26.10.2012 Views

Abstract Book 2010 - CIMT Annual Meeting

Abstract Book 2010 - CIMT Annual Meeting

Abstract Book 2010 - CIMT Annual Meeting

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>Abstract</strong>book <strong>2010</strong><br />

8 th <strong>Annual</strong> <strong>Meeting</strong>


Dear colleagues, members and friends,<br />

On behalf of the organizing committee, we would like to welcome you to<br />

the 8th annual <strong>CIMT</strong> meeting in Mainz from May 26-28, <strong>2010</strong>. The newly<br />

formed Scientific Program Committee has done an outstanding job this<br />

year putting together a comprehensive program which covers the whole<br />

range of innovative developments in immunotherapy.<br />

The scope of this year's meeting stretches from tumor immunosupression,<br />

regulatory issues, the potential of adjuvants and new adoptive cellular<br />

therapies to therapeutic vaccination. Because the gap between "bench and<br />

bedside" is still wide, we put focus on the question how translation can<br />

be successful. We will have the opportunity to share three days of plenary<br />

sessions, workshops and guided poster sessions. The expanded short<br />

talk sessions and the guided poster sessions are again main tracks in the<br />

program. An industry exhibition and satellite workshops accompany the<br />

conference.<br />

Your participation makes this meeting exciting. Welcome to Mainz!<br />

Ch. Huber (Mainz)<br />

P. Johnson (Southampton)<br />

U. Kalinke (Hannover)<br />

C. Melief (Leiden)<br />

H.G. Rammensee (Tuebingen)<br />

G. Schuler (Erlangen)<br />

P. Walden (Berlin)<br />

3


The Association<br />

The association for Cancer Immunotherapy (<strong>CIMT</strong>) is a non profit organisation<br />

and was founded in the fall of 2002 as an information and<br />

education platform for immunological cancer therapy. Physicians and<br />

researchers from different fields of clinical and theoretical medicine<br />

from the universities of Berlin, Erlangen, Mainz and Tuebingen were the<br />

founders of the association.<br />

<strong>CIMT</strong> is located at the university medical center of the Johannes Gutenberg<br />

University Mainz.<br />

The association is financed by donations, congress fees and sponsoring.<br />

Our Goals<br />

The purpose of <strong>CIMT</strong> is to enhance the development of cancer immunotherapies<br />

by:<br />

* Establishing a communication platform in order to facilitate the com-<br />

munication between scientists of all faculties with interest in cancer<br />

immunotherapy. We improve knowledge exchange between industry<br />

based scientists, academic scientists, regulatory authorities and phy-<br />

sicians alike. Our scientific meetings, educational symposia and pu-<br />

blications serve this purpose.<br />

* Education of health-professionals with annual meetings, advanced<br />

education seminars and publication of textbooks.<br />

* Cooperation with partners in related consortia, regulatory agencies,<br />

journals, academic institutions and companies.<br />

* Initiation of working parties that actively accelerate the development<br />

in the field.<br />

4<br />

Who We Are


Managing Committee:<br />

Christoph Huber, chair<br />

Mainz, Germany<br />

Peter Johnson<br />

Southampton, UK<br />

Ulrich Kalinke<br />

Hannover, Germany<br />

Cornelis Melief<br />

Leiden, Netherlands<br />

Executive Director - Science:<br />

Sebastian Kreiter<br />

Mainz, Germany<br />

Scientific Secretary:<br />

Mustafa Diken<br />

Mainz, Germany<br />

Who We Are<br />

Hans Georg Rammensee<br />

Tuebingen, Germany<br />

Gerold Schuler<br />

Erlangen, Germany<br />

Peter Walden<br />

Berlin, Germany<br />

Executive Director - Translational Medicine:<br />

<strong>CIMT</strong> - Education:<br />

Cedrik Britten<br />

Mainz, Germany<br />

Torsten Seppmann<br />

Mainz, Germany<br />

5


<strong>CIMT</strong> - Office:<br />

6<br />

Kristiane Preuss<br />

Isny, Germany<br />

Scientific Advisory Board:<br />

Philipp Beckhove<br />

Heidelberg, Germany<br />

Peter Brossart<br />

Bonn, Germany<br />

Ulrich Keilholz<br />

Berlin, Germany<br />

Christian Ottensmeier<br />

Southampton, UK<br />

Giorgio Parmiani<br />

Milan, Italy<br />

Ugur Sahin<br />

Mainz, Germany<br />

Who We Are<br />

<strong>CIMT</strong> - Communications:<br />

Christine Castle<br />

Mainz, Germany<br />

Michael Schmitt<br />

Kiel, Germany<br />

Joachim Schultze<br />

Bonn, Germany<br />

Harpreet Singh<br />

Tuebingen, Germany<br />

Freda Stevenson<br />

Southampton, UK<br />

Thomas Tüting<br />

Bonn, Germany


Visit our website<br />

www.cimt.eu<br />

find us on facebook<br />

Websites<br />

www.facebook.com/cimtmainz<br />

follow us on twitter<br />

www.twitter.com/C_IMT<br />

7


Program Committee and Speakers<br />

<strong>2010</strong> Program Committee Members:<br />

8<br />

Giorgio Parmiani<br />

Unit of immune-biotherapy of melanoma<br />

and solid tumors<br />

Fondazione Centro san Raffaele del Monte<br />

Tabor<br />

Milan, Italy<br />

Graham Pawelec<br />

Zentrum für Medizinische Forschung<br />

Sektion Transplantationsimmunologie/Immunhämatologie,<br />

Medizinische Klinik II<br />

Tuebingen, Germany<br />

Christian Ottensmeier<br />

Cancer Sciences Division<br />

Southampton University Hospitals<br />

Southampton, United Kingdom<br />

Axel Hoos<br />

Co-Chairman, CVC Executive Committee;<br />

Program leader in Immunology/Oncology<br />

at Bristol-Myers Squibb<br />

Bristol Myers Squibb Inc.<br />

Wallingford, USA<br />

Hansjoerg Schild<br />

Institute for Immunology<br />

JGU Universitätsmedizin Mainz<br />

Mainz, Germany<br />

Harpreet Singh<br />

Immatics<br />

Tuebingen, Germany<br />

Cecile Gouttefangeas<br />

Department of Immunology<br />

University of Tuebingen<br />

Tuebingen, Germany<br />

Dolores Schendel<br />

Institut für Molekulare Immunologie (IMI)<br />

Helmholtz Zentrum München<br />

München, Germany<br />

Freda Stevenson<br />

Molecular Immunology Group, Cancer<br />

Sciences Division<br />

School of Medicine, University of<br />

Southampton<br />

Southampton, United Kingdom<br />

Wolfgang Herr<br />

Innere Medizin<br />

JGU Universitätsmedizin Mainz<br />

Mainz, Germany<br />

Antoine Tesniere<br />

Institut Gustave Roussy<br />

INSERM Unit U848, „Apoptosis, Cancer &<br />

Immunity“<br />

Paris, France<br />

Sebastian Kreiter<br />

Institute for Translational Oncology (TrOn)<br />

Johannes Gutenberg-Universität Mainz<br />

Mainz, Germany<br />

Cedrik Britten<br />

III. Medizinische Klinik und Poliklinik<br />

Johannes Gutenberg-Universität Mainz<br />

Mainz, Germany<br />

Sjoerd H. van der Burg<br />

Department for Clinical Oncology<br />

Leiden University Medical Center<br />

Leiden, Netherlands


<strong>2010</strong> Speakers:<br />

G. J. Adema<br />

Radboud University Nijmegen Medical<br />

Centre<br />

Nijmegen Centre for Molecular Life Sciences<br />

(NCMLS)<br />

Nijmegen, Nethterlands<br />

Prof. Dr. Adema holds the chair in Molecular Immunology at<br />

the NCMLS Department of Tumor Immunology. His research<br />

mainly focuses on the molecular analysis of dendritic cells and<br />

regulatory T cells, and their function in the immune system in<br />

health and disease. The knowledge gathered in the fundamental<br />

immunological studies is translated into pre-clinical and clinical<br />

vaccination studies of cancer patients with autologous,<br />

antigen-loaded dendritic cell vaccines.<br />

James Allison<br />

Ludwig Center for Cancer Immunotherapy<br />

New York, USA<br />

James Allison has a longstanding interest the in the mechanisms<br />

of T cell activation and its regulation, as well as developing<br />

novel strategies for immunotherapy of cancer. He has made many<br />

significant contributions to our understanding T cell costimulation<br />

and inhibition, and conceived the notion of immunological<br />

checkpoint blockade for the therapy of cancer. He is currently<br />

the Chairman of the Immunology Program, Director of the Ludwig<br />

Center for Cancer Immunotherapy, David H. Koch Chair in<br />

Immunologic Studies, Attending Immunologist at the Memorial<br />

Sloan Kettering Cancer Center. He is also an Investigator of the<br />

Howard Hughes Medical Institute, a Member of the National<br />

academy of Sciences, and of the Institute of Medicine.<br />

Victor Appay<br />

INSERM<br />

Paris, France<br />

Dr. Appay leads the HIV pathogenesis and Immunosenescence<br />

group at the Institut National de la Santé et Recherche Médicale<br />

(INSERM). His main research interest is exploring the factors<br />

that govern the development and maintenance of effective<br />

CD8+T cell responses in HIV infection using basic immunology.<br />

The work of his team also focuses on the development with<br />

progressive HIV disease of premature immunosenescence, or<br />

immune ageing.<br />

John Barrett<br />

National Heart, Lung and Blood Institute<br />

National Institutes of Health<br />

Bethesda, USA<br />

John Barrett is Chief of the National Heart, Lung and Blood<br />

Institute‘s Stem Cell Allotransplantation Section of the Hemato-<br />

logy Branch, National Heart, Lung and Blood Institute, National<br />

Institutes of Health, in Bethesda, USA. His research is directed<br />

to defining the antigens involved in graft-versus-host disease,<br />

graft-versus-leukemia and graft versus tumor immunity and<br />

designing improved ways to reconstitute the immune system<br />

after stem cell transplantation using technologies to selectively<br />

eliminate harmful alloreactive T cells from the transplant while<br />

conserving graft-versus-leukemia reactivity.<br />

Philipp Beckhove<br />

Deutsches Krebsforschungszentrum<br />

Heidelberg, Germany<br />

PD. Dr. Philipp Beckhove leads an independent research group<br />

at the German Cancer Research Center. The objective of Dr.<br />

Beckhove’s research group „Translational Immunology“ is to<br />

gain new insights into the immune defence system of cancerous<br />

cells and to evolve the results from basic research through to<br />

clinical treatments. The pursuit of which is based on close interdisciplinary<br />

collaboration with the oncological departments of<br />

the university hospital. New therapeutic concepts developed by<br />

the research group are being “translated” into clinical application<br />

under the roof of the National Center for Tumor Diseases<br />

in Heidelberg (NCT).<br />

J. S. Bromberg<br />

Mount Sinai School of Medicine<br />

New York, USA<br />

Prof. Dr. Bromberg has great experience in starting, building,<br />

and reorganizing critical components of multiorgan transplant<br />

programs. In addition to his clinical transplantation practice,<br />

Prof. Bromberg is pursuing basic and clinical research involving<br />

immunology and transplantation. His current projects are<br />

focused on determination of key mechanisms in T cell homing<br />

and tolerance induction, investigation of signals important for<br />

induction and inhibition of Foxp3 expression in regulatory T<br />

cells (Treg) as well as interaction of Tregs with other cells in<br />

different disease models of autoimmunity.<br />

Federica Cavallo<br />

University of Torino<br />

Torino, Italy<br />

Federica Cavallo, PhD, is Associate Professor of Immunology<br />

(General Pathology) of the University of Torino. She is among<br />

the founders of the new Molecular Biotechnology Center (MBC)<br />

of the University of Torino, where her laboratory is now located.<br />

She has a successful experience in basic and translational cancer<br />

immunotherapy, and has a considerable expertise in transgenic<br />

mouse models of cancer and DNA vaccination. At present her<br />

lab is combining immune regulators of various kind and vaccines<br />

to prevent and cure autochthonous cancers in pre-clinical<br />

models. A major effort is devoted to grab new knowledge on<br />

tumor and microenvironment transcriptome from microarray<br />

9


<strong>2010</strong> Speakers:<br />

analysis and next generation sequencing data. These technologies<br />

are currently exploited to tease apart new mechanisms<br />

of carcinogenesis inhibition and to spot both miRNA and new<br />

oncoantigens to be exploited in a new generation of antitumor<br />

vaccines.<br />

10<br />

Helen X. Chen<br />

Investigational Drug Branch, Cancer<br />

Therapy Evaluation Program<br />

National Cancer Institute<br />

Bethesda, USA<br />

Dr. Helen Chen, M.D. is currently Associate Branch Chief of the<br />

Investigational Drug Branch of National Cancer Institute (NCI)’s<br />

Cancer Therapy Evaluation Program (CTEP). She is responsible<br />

for oversight of NCI-sponsored clinical trials involving antiangiogenesis<br />

therapies, EGFR/HER2 and IGF-1R targeting agents,<br />

and monoclonal antibody therapeutics. She is also involved in<br />

the development of NCI’s program for combination of molecular<br />

targeted investigational agents. She has authored more than 30<br />

publications and book chapters on these areas, and has been on<br />

scientific committees for AACR and AACR-EORTC-NCI <strong>Annual</strong><br />

<strong>Meeting</strong>s, the ASCO-NCI-EORTC <strong>Annual</strong> <strong>Meeting</strong> on Molecular<br />

Markers, and the International Symposium for Antiangiogenesis<br />

Therapies. Dr. Chen a diplomate of the American Board of<br />

Internal Medicine, and also serves as an attending physician in<br />

the NCI’s Phase I clinic.<br />

Mark M. Davis<br />

Stanford University School of Medicine<br />

Stanford, USA<br />

Professor Dr. Davis, Director of the Stanford Institute for Immunology,<br />

Transplantation and Infection (ITI), is a professor of<br />

microbiology and immunology and a Howard Hughes Medical<br />

Institute investigator. He is well known for identification in<br />

the 1980s of the elusive T-Cell receptor genes, which allow T<br />

lymphocytes to fight disease causing microbes, and he and his<br />

group have made many subsequent discoveries about this type<br />

of molecule and how it functions.<br />

Jolanda M. de Vries<br />

Nijmegen Centre for Molecular Life<br />

Sciences)<br />

Nijmegen, Netherlands<br />

Dr. Jolanda de Vries is Assistant Professor at the Department<br />

of Tumor Immunology at the Nijmegen Centre for Molecular<br />

Life Sciences. She was one of the pioneers to translate dendritic<br />

cell biology into potential clinical applications. The first clinical<br />

phase I/II studies in which patients were vaccinated with DCs<br />

loaded with tumor-specific peptides were initiated in 1997. She<br />

also developed a novel immuno-monitoring assay that is highly<br />

predictive for extended survival after vaccination with DCs (J<br />

Clin Oncology 2005). Her primary scientific interest continues<br />

along the line of DC-immunotherapy and in particular the<br />

migration and imaging of DC. For example, in-vivo imaging of<br />

ex-vivo labeled cells using MRI (Nature Biotechnology 2005).<br />

New opportunities for other cell-types (e.g. subsets of DCs) are<br />

now being developed. She recently completed the first plasmacytoid<br />

DC vaccination trial.<br />

M. V. Dhodapkar<br />

Yale University School of Medicine<br />

New Haven, USA<br />

Prof. Dr. Dhodapkar is the chief of Section of Hematology at Yale<br />

School of Medicine and the director of Department of Hematologic<br />

Malignancies at Yale Cancer Center. He is interested in<br />

studying the biology of the human immune system and how<br />

it interacts with growing tumors in patients. His main reseach<br />

goal is understanding and harnessing the properties of the<br />

immune system to detect, prevent and treat cancer, with a particular<br />

focus on multiple myeloma.<br />

Mary L. (Nora) Disis<br />

University of Washington<br />

School of Medicine<br />

Washington, USA<br />

Mary L. (Nora) Disis, M.D. is the Associate Dean for Translational<br />

Health Sciences in the University of Washington<br />

(UW) School of Medicine, Professor of Medicine and Adjunct<br />

Professor of Pathology and Obstetrics and Gynecology at UW<br />

and a Member of the Fred Hutchinson Cancer Research Center<br />

(FHCRC). Dr. Disis is an expert in breast and ovarian cancer<br />

immunology and translational research. Her research interest is<br />

in the discovery of new molecular targets in breast and ovarian<br />

cancer for the development of vaccine and cellular therapy for<br />

the treatment of those malignancies.<br />

Mark E. Dudley<br />

National Cancer Institute<br />

Bethesda, USA<br />

Dr. Mark E. Dudley is the head of the Cell Production Facility at<br />

the Surgery Branch, National Cancer Institute. The Cell Production<br />

Facility translates basic research discoveries into cell and<br />

gene therapies for patients with cancer. Current projects include<br />

closed system bioreactors, retroviral and lentiviral gene therapy<br />

vectors, and artificial antigen presenting cells. These systems<br />

are being developed in the context of ongoing clinical trials to<br />

treat patients with advanced melanoma and other cancers.


<strong>2010</strong> Speakers:<br />

D. I. Gabrilovich<br />

University of South Florida<br />

Tampa, USA<br />

Prof. Dr. Gabrilovich holds a chair in tumor immunology at the<br />

Department of Oncologic Sciences-University of South Florida.<br />

His research mainly focuses understanding the biology and<br />

mechanism of tumor-associated immunosuppression and exploring<br />

approaches to eliminate immunosuppression. His team is<br />

also working on development of novel effective cancer vaccines<br />

including genetically modified dendritic cells and the combination<br />

of apoptosis-inducing therapy and immunotherapy.<br />

John B.A.G. Haanen<br />

Netherlands Cancer Institute<br />

Antoni van Leeuwenhoek Hospital<br />

Leiden, Netherlands<br />

Prof. Dr. John B.A.G. Haanen, MD PhD is head of the Division<br />

of Medical Oncology at the Netherlands Cancer Institute/Antoni<br />

van Leeuwenhoek Hospital. His main research interest is immunotherapy<br />

of cancer and he was recently appointed professor of<br />

translational immunotherapy of cancer at the Leiden University.<br />

The main topics of his research are on the development of<br />

DNA-based vaccine strategies for the treatment of cancer and<br />

adoptive cellular therapy. In close collaboration with prof. Ton<br />

Schumacher he has developed a program for T cell receptor gene<br />

therapy.<br />

Mirjam Heemskerk<br />

Leiden University Medical Center<br />

Leiden, Netherlands<br />

Mirjam H. M. Heemskerk is currently Associate Professor and<br />

Head of the Laboratory of Experimental Hematology at the<br />

LUMC Department of Hematology which she joined to pioneer<br />

the retroviral introduction of antigen specific T cell receptors as<br />

a means to engineer leukemia specific immunity. Her research<br />

interests are characterisation of alloimmune responses and<br />

occurrence of delayed immune reconstitution after HLA mismatched<br />

stem cell transplantation, high throughput identification<br />

of hematopoiesis-restricted minor histocompatibility antigens<br />

by reverse immunology approaches and TCR gene therapy<br />

of hematological malignancies. A clinical trial is being set up to<br />

treat patients with hematological malignancies after allogeneic<br />

stem cell transplantation with TCR engineered T cells.<br />

Thomas Hünig<br />

Institute for Virology and Immunobiology<br />

University of Würzburg<br />

Würzburg, Germany<br />

Prof. Dr. Hünig holds a chair in Immunobiology at the Institute<br />

for Virology and Immunobiology. He was co-founder of the<br />

company TeGenero that developed the CD28-superagonist antibodyTGN1412.<br />

His main focus of research is regulation of T cell<br />

activation by CD28 and CTLA4.<br />

Ulrich Kalinke<br />

Twincore<br />

Hannover, Germany<br />

Prof. Dr. Kalinke is the Executive Director of TWINCORE<br />

Research Center in Hannover and former Head of the Department<br />

of Immunology at Paul-Ehrlich-Institut (Langen). His<br />

main research area is experimental infection focusing on the<br />

influence of interferons on adaptive immunity and how different<br />

pathogens prevent the release of interferons.<br />

Samir N. Khleif<br />

National Cancer Institute<br />

Bethesda, USA<br />

Prof. Dr. Khleif is head of the Cancer Vaccine Section, Vaccine<br />

Branch at the NCI and he also serves as a Special Assistant to the<br />

Commissioner of the Food and Drug Administration, leading<br />

the Critical Path Initiative for oncology. His reseach focuses on<br />

integrating translational basic laboratory research and clinical<br />

trials to understand the interaction between tumor cells and the<br />

immune system and to develop cancer vaccines.<br />

Andreas Mackensen<br />

University Erlangen<br />

Erlangen, Germany<br />

Professor Dr. Andreas Mackensen holds the chair position for<br />

Hematology/Oncology at the University Hospital of Erlangen.<br />

His main research area is the development of immunotherapeutic<br />

strategies for the treatment of malignant diseases.<br />

Raj K. Puri<br />

Division of Cellular and Gene Therapies<br />

U.S. FDA´s Center for Biologics Evaluation<br />

and Research<br />

Silver Spring, USA<br />

Raj K. Puri, M.D., Ph.D. is the Director of the Division of Cellular<br />

and Gene Therapies (DCGT) in the U.S. FDA’s Center for<br />

Biologics Evaluation and Research. He is also a Chief of Tumor<br />

Vaccines and Biotechnology Branch (TVBB). Dr. Puri oversees<br />

investigational new drug (INDs), investigational device exemptions<br />

(IDEs), and biological license applications (BLAs) for<br />

tumor vaccines, immunotherapy, cellular and gene therapy, tissue<br />

engineering, and xenotransplantation products and deve-<br />

11


<strong>2010</strong> Speakers:<br />

lopment of policies and guidance documents in these cutting<br />

edge areas of medical research.<br />

Dr Rezvani is a Clinical Senior Lecturer and Consultant Haematologist<br />

at Imperial College in London. She is the clinical lead in<br />

allogeneic stem cell transplantation and clinical director of the<br />

Jacie accredited GMP cellular facility.<br />

As a research fellow at the NIH from 2001-2008, her laboratory<br />

studies in immune responses to leukemia led to the initiation<br />

of 3 FDA approved clinical trials of vaccination in patients<br />

with myeloid malignancies. She received a highly competitive<br />

award from the Higher Education Funding Council for England<br />

(HEFCE) and started her position as a clinical senior lecturer<br />

at Imperial College in January 2008 where she has an active<br />

research laboratory program in transplantation and tumor<br />

immunology.<br />

12<br />

Katy Rezvani<br />

Imperial College London<br />

London, England<br />

Antoni Ribas<br />

University of California<br />

Los Angeles, USA<br />

Antoni Ribas, MD, is Associate Professor of Medicine and Surgery<br />

at University of California, Los Angeles. He practices oncology<br />

focused on new drug development for melanoma, and has<br />

an NIH-funded research laboratory focused also in melanoma.<br />

He also serves as Director of the Cell and Gene Therapy Core<br />

Facility, Assistant Director of the Tumor Immunology Program,<br />

Assistant Director of the Human Gene Medicine Program, General<br />

Clinical Research Center Advisory Board Member, and trains<br />

faculty at the Institute for Stem Cell Biology and Medicine.<br />

Dr. Ribas is a permanent committee member of the NCI grant<br />

review panels. Dr. Ribas‘ clinical and laboratory research involves<br />

the development of novel strategies for the treatment of cancer,<br />

including anti-CTLA4 antibodies, cancer vaccines, BRAF<br />

inhibitor and Mek inhibitor targeted therapy, nanoparticle delivery<br />

of siRNA, and gene therapy strategies.<br />

Martina Schuessler-Lenz<br />

Paul-Ehrlich Institut<br />

Langen, Germany<br />

Dr. Schuessler-Lenz is a Senior Clinical Assessor at the Division<br />

Medical Biotechnology of the Paul-Ehrlich Institut, and the<br />

alternate German member of the Committee for Advanced Therapies<br />

(CAT) of the European Medicines Agency. Her main area<br />

of expertise is the clinical development of Advanced Therapy<br />

Medicinal Products and of cancer immunotherapies.<br />

Protul A. Shrikant<br />

Roswell Park Cancer Insitute<br />

New York, USA<br />

Dr. Protul A. Shrikant completed a National Multiple Sclerosis<br />

Society Fellowship at the University of Minnesota, Center for<br />

Immunology (1999) and joined Roswell Park Cancer Institute in<br />

2000 as an Assistant Professor in the Department of Immunology<br />

(2000), where he currently is an Associate Professor with<br />

tenure and serves on the advisory council for the Centre for<br />

Immunotherapy. He also holds an adjunct appointment with the<br />

Departments of Microbiology and Immunology at State University<br />

of New York at Buffalo. His research is focussed on characterizing<br />

the molecular pathways by which instructions program<br />

CD8 T cell responses and exploit the insights for developing<br />

novel strategies for tumor immunity.<br />

Masato Tanaka<br />

RIKEN Research Center for Allergy<br />

and Immunology<br />

Yokohama, Japan<br />

Masato Tanaka, M.D. and Ph.D., graduated from Tokyo Medical<br />

and Dental University in 1988. I worked for Osaka Bioscience<br />

Institute and Osaka University Medical School, and engaged<br />

in research of the molecular mechanisms of apoptosis in laboratory<br />

of Prof. Shigekazu Nagata. I worked for Tularik Inc. in<br />

San Francisco, USA to investigate the molecular mechanisms<br />

of inflammatory reaction in 1997-1999. Since 2002, I became<br />

the team leader of Laboratory for Innate Cellular Immunity,<br />

RIKEN Research Center for Allergy and Immunology (RCAI),<br />

Yokohama Japan. In recent years, I focused on the physiological<br />

and pathological roles of apoptotic cell clearance by phagocytes,<br />

and strategic approach for immune regulation by dead<br />

cell clearance.<br />

David L. Urdal<br />

Dendreon Corporation<br />

Seattle, USA<br />

Dr. Urdal has been Chief Scientific Officer and a Director of<br />

Dendreon Corporation since he joined the company in July of<br />

1995. Dendreon (Nasdaq: DNDN) has been dedicated to targeting<br />

cancer and transforming lives through the discovery and<br />

development of novel products like Provenge®, an active immunotherapy<br />

for prostate cancer that has shown promise in Phase<br />

III clinical trials.<br />

From 1982 to 1995, he held various positions with Immunex<br />

Corporation, including President of Immunex Manufacturing<br />

Corporation, Vice President and Director of Development, and<br />

Head of the Departments of Biochemistry and Membrane Biochemistry.<br />

At Immunex he participated in the discovery, development<br />

and commercialization of hematopoietic growth factors,<br />

cytokines and cytokine receptor drugs, like Leukine® and<br />

Enbrel®. Dr. Urdal received an M.S. in Public Health and a Ph.D.<br />

in Biochemical Oncology from the University of Washington. He<br />

is inventor on 16 patents and author on 79 publications.


<strong>2010</strong> Speakers:<br />

Weiping Zou<br />

Department of Surgery and Comprehensive<br />

Cancer Center<br />

University of Michigan School of<br />

Medicine<br />

Ann Arbor, USA<br />

Weiping Zou is a Professor of Surgery, Immunology and Biology<br />

and the director for Translational Research at the University of<br />

Michigan. His research interests are in tumor immunopathology<br />

and immunotherapy, with an emphasis on the cross-talk among<br />

immune cell subsets, stromal cells, tumor cells and tumor stem<br />

cells in the tumor microenvironment, and its impact on tumor<br />

immunity, tolerance and therapy.<br />

13


Scientific Program <strong>CIMT</strong> <strong>2010</strong> Day 1 (May 26 th )<br />

09:00-10:00 Welcome coffee<br />

14<br />

Welcome addresses<br />

10:00-10:05 C. Huber (<strong>CIMT</strong> Chairman, Mainz)<br />

10:05-10:10 G. Krausch (University President, Mainz)<br />

10:10-10:15 R. Urban (CSO, University Medicine Mainz)<br />

Session 1: Cellular therapies<br />

Chairpersons: M. Theobald (Mainz), S. Mielke (Würzburg)<br />

10:20-10:50 J. Barrett (Bethesda)<br />

Selective depletion of alloreactive T cells: promises and challenges<br />

10:50-11:20 M. Heemskerk (Leiden)<br />

Risks and benefits of TCR gene therapy<br />

11:20-11:50 A. Mackensen (Erlangen)<br />

11:50-13:30 Lunch break<br />

Industry satellite symposium I:<br />

Cellular therapy in solid tumors - New perspectives<br />

(organized by Miltenyi Biotec)<br />

12:15-12.40 J. de Vries (Radboud University Nijmegen Medical Center, Netherlands)<br />

Human plasmacytoid dendritic cells in cancer and cancer therapy;<br />

12:40-13.50 Philipp Beckhove (German Cancer Research Center Heidelberg, Germany)<br />

Regulation of T cell responses in cancer<br />

13.05-13.30 Mark E. Dudley (National Cancer Institute, NIH, Bethesda, MD, USA)<br />

Adoptive T cell therapy for melanoma<br />

11:50-13:00 <strong>CIMT</strong> members meeting<br />

Session 2: Tumor vaccination<br />

Chairperson: C. Ottensmeier (Southampton), C. Melief (Leiden)<br />

13:30-14:00 J.B. Haanen (Amsterdam)<br />

Therapeutic DNA vaccination by tattooing: from preclinical development to a first<br />

clinical trial<br />

14:00-14:30 K. Rezvani (London)<br />

The role of immunotherapy in the eradication of minimal residual disease in CML<br />

14:30-15:00 F. Cavallo (Torino)<br />

DNA Vaccination in Prevention and Cure of Autoch thonus Mammary Carcinoma in<br />

Her2-tg Mice – Implications for Clinical Testing<br />

15:00-15:30 D. Urdal (Dendreon, Seattle)<br />

Sipuleucel-T: Active cellular immunotherapy for castrate resistant prostate cancer<br />

15:30-15:50 Coffee break<br />

Short talk session I: Immune monitoring<br />

Chairpersons: C. Britten (Mainz), J. de Vries (Nijmegen)<br />

15:50-16:05 T. de Gruijl (Amsterdam) (# 021)<br />

Immune and clinical response monitoring in patients with metastatic hormone-refrac<br />

tory prostate cancer receiving combined Prostate GVAX and anti-CTLA4 immunotherapy


16:05-16:20 K. Laske (Tuebingen) (# 028)<br />

T-cell immunomonitoring in patients with renal cell carcinoma after multi-peptide<br />

vaccination<br />

16:20-16:35 M. Guidoboni (Meldola) (# 034)<br />

Changes of immune cell infiltrates induced by dendritic cell vaccination in melanoma<br />

patients’ tumor tissues: an immunohistochemical study<br />

16:35-16:50 S. Voland (Erlangen) (# 035)<br />

Characterisation and functional analysis of T-cell responses in melanoma patients<br />

vaccinated with peptide-loaded dentritic cells<br />

16:50-17:05 E. Aarntzen (Bemmel) (# 037)<br />

Skin-derived functional specific Tcells predict clinical outcome upon DC vaccination in<br />

stage III and IV melanoma patients<br />

Short talk session II: Therapeutic tumor vaccination<br />

Chairpersons: S. Grabbe (Mainz), P. thor Straten (Copenhagen)<br />

15:50-16:05 C Maccalli (Milan) (# 078)<br />

Definition of the immunological properties of cancer stem cells isolated from human<br />

glioblastoma<br />

16:05-16:20 I. Poschke (Stockholm) (# 079)<br />

Immature immunosuppressive CD14+HLA-DR-/low cells in melanoma patients are<br />

Stat3hi and over-express CD80, CD83 and DC-Sign<br />

16:20-16:35 E. Lion (Borgerhout) (# 081)<br />

Poly(I:C)-electroporated myeloid leukemic cell lines become highly susceptible to NK<br />

cell-mediated killing and phagocytosis by immature DC<br />

16:35-16:50 A. Bonertz (Heidelberg) (# 082)<br />

Antigen-specific Treg in colorectal carcinoma control T cell responses against a limited<br />

tumor antigen repertoire<br />

16:50-17:05 C. Lutz-Nicoladoni (Innsbruck) (# 087)<br />

Reinforcement of Cancer Immunotherapy by Adoptive Transfer of cblb-deficient<br />

Cytotoxic T Lymphocytes combined with a Dentritic Cell Vaccine<br />

Short talk session III: New targets & new leads<br />

T. Wölfel (Mainz), S. Kreiter (Mainz)<br />

15:50-16:05 R. Ashfield (Abington) (# 062)<br />

ImmTACs: bi-functional reagents for redirected tumour cell killing<br />

16:05-16:20 N. Cesares (Pamplona) (# 065)<br />

Suppression of Treg activity by a FOXP3 inhibitor peptide<br />

16:20-16:35 M. H. Andersen (Herlev) (# 069)<br />

Cellular Immune Responses Against Indoleamine 2,3-dioxygenase<br />

16:35-16:50 W. van Esch (Amsterdam) (# 072)<br />

T cell epitope discovery through HLA class I peptide ligand exchange and combinatori<br />

al coding<br />

16:50-17:05 N. Schaft (Erlangen) (# 074)<br />

Transfer of mRNA encoding chimeric antigen receptors specific for MCSP into CD4+<br />

and CD8+ T cells<br />

Key Note Lecture<br />

Chairperson: C. Britten (Mainz)<br />

17:10-17:50 Mark M. Davis (Stanford)<br />

The coming golden age of human immunology<br />

19:30-23:00 Speakers dinner<br />

15


Scientific Program <strong>CIMT</strong> <strong>2010</strong> Day 2 (May 27 th )<br />

16<br />

Session 3: Combination Partners for Vaccination<br />

Chairpersons: A. Hoos (New York), G. Schuler (Erlangen)<br />

08:40-09:10 J. Allison (New York)<br />

Immune checkpoint blockade in cancer therapy: New insights and opportunities<br />

09:10-09:40 A. Ribas (Los Angeles)<br />

Immunosensitization: Targeted therapies to improve tumor immunotherapy<br />

09:40-10:20 H. Chen (Bethesda)<br />

CTEP/NCI’s model to facilitate clinical trials for combination between investigational<br />

agents<br />

10:20-10:50 Coffee break<br />

Short talk session IV: CIP-Session<br />

Chairpersons: C. Ottensmeier (Southampton), M. Welters (Leiden)<br />

10:45-11:00 C. Britten (Mainz)<br />

Overview on CIP activities in 2009<br />

11:00-11:20 C. Gouttefangeas (Tuebingen)<br />

Update on the CIP HLA-multimer panels<br />

11:20-11:40 C. Ottensmeier (Southampton)<br />

Plans for the new ELISPOT panels (IVS T cell in UK and freezing medium in Mainz<br />

11:40-12:00 S. Singh (Leiden)<br />

A stable standard sample for harmonisation and validation of T cell assays<br />

12:00-12:20 S. Janetzki (New York)<br />

Influence of harmonization on assay performance – the CIC experience<br />

Short talk session V: Cellular therapy<br />

Chairpersons: M. Kalos (Philadelphia), R. G. Meyer (Mainz)<br />

10:50-11:05 N. Auphan-Anezin (Marseille) (# 045)<br />

Optimizing effector and memory properties of CD8 T cells for adoptive tumor<br />

immunotherapy<br />

11:05-11:20 D. Schendel (München) (# 048)<br />

High avidity T cell clones specific for tumor-associated antigens and how to find them<br />

11:20-11:35 D. Stolle (Mainz) (# 053)<br />

Generation of HCMV-/TAA-bispecific human T cells by either the genetic equipment of<br />

HCMV+ T cells with tumor-reactive TCRs or the combined retroviral transduction of<br />

bulk human T cells with HCMV-/ TAA-specific TCRs<br />

11:35-11:50 A. Bloetz (Mainz) (# 054)<br />

Allo-reactivity to HLA class II mismatch alleles in CD4+ T-cell subsets<br />

11:50-12:05 H. Echchannaoui (Mainz) (# 058)<br />

Evaluation of the safety of p53 TCR gene transfer in a humanized mouse model<br />

Short talk session VI: Tumor biology / interaction with<br />

the immune system<br />

Chairpersons: V. Umansky (Heidelberg), D. Schendel (Munich)<br />

10:50-11:05 M. Ramacher (Heidelberg) (# 091)<br />

Immunosuppression in transgenic mouse melanoma model induced by myeloid de<br />

rived suppressor cells


11:05-11:20 M. Koslowski (Mainz) (# 093)<br />

Tumor-associated genomic DNA hypomethylation effects positive autoregulation of HIF-1α<br />

11:20-11:35 E. Quaglino (Turin) (# 094)<br />

miRNAs expression profiles during ErbB2 driven mammary carcinogenesis<br />

11:35-11:50 C. Zimmer (Essen) (# 102)<br />

Impact of Endoplasmic reticulum aminopeptidases (ERAP) on T cell epitope genera<br />

tion in melanoma cells<br />

11:50-12:05 L. Hansmann (Regensburg) (# 104)<br />

Isolation of intact genomic DNA from FOXP3-stained and FACS-sorted regulatory T<br />

cells for epigenetic analysis<br />

12:05-14:00 Lunch break<br />

Industry satellite symposium II<br />

12.30-14:00 (organized by Beckman Coulter)<br />

Dr. Andreas Boehmler / Dr. Michael Kapinsky<br />

Multilaser and Multicolour Flowcytometry -Upcoming challenges!<br />

New concepts in experimental setup and analysis<br />

Session 4: Regulation – how translation can be successful<br />

(organized by the <strong>CIMT</strong> RRP)<br />

Chairperson: T. Hinz (PEI), H. Singh (Tuebingen)<br />

14:00-14:15 U. Kalinke (Hannover)<br />

Regulatory challenges in translational research<br />

14:15-14:35 R. K. Puri (FDA-CBER, Rockville)<br />

Regulatory considerations for biologics for cancer therapy<br />

14:35-14:55 T. Hünig (Würzburg)<br />

Functional impairment of circulating T-cells questions validity of conventional PBMC<br />

testing: the case of the CD28 superagonist TGN1412<br />

14:55-15:15 M. Schuessler-Lenz (PEI, Langen)<br />

The role of the European Committee for Advanced Therapies in the Evaluation of Cancer<br />

Immunotherapies<br />

15:30-16:10 Panel Discussion<br />

Poster Session I<br />

Guided poster session (+ coffee & cookies)<br />

16:15-18.00 Therapeutic vaccination I, Chair: V. Lennerz (Mainz)<br />

Immune monitoring I, Chair: C. Bain (Lyon)<br />

Cellular therapy I, Chair: H. Voß (Mainz)<br />

New targets & new leads I, Chair: K. Thielemans (Brussels)<br />

Tumor biology & interaction with the immune system I, Chair: S. Pascolo (Zurich)<br />

Enhancing immunity & adjuvants, Chair: M. Radsak (Mainz)<br />

Social event<br />

19:30-23:30 Casual dinner in the “Rheingold” lounge<br />

17


Scientific Program <strong>CIMT</strong> <strong>2010</strong> Day 3 (May 28 th )<br />

18<br />

Poster Session II<br />

Guided poster session (+ pretzels)<br />

08:30-09:50 Therapeutic vaccination II, Chair: M. Schmitt (Rostock)<br />

Immune monitoring II, Chair: S. Groß (Erlangen)<br />

Cellular therapy II, Chair: R. Stripecke (Hannover)<br />

New targets & new leads II, Chair: V. Umansky (Heidelberg)<br />

Tumor biology & interaction with the immune system II, Chair: U. Hartwig (Mainz)<br />

09:50-10:20 Coffee break<br />

Session 5: Chemo-Immunotherapy<br />

Chairperson: A. Tesnière (Paris), H.-G. Rammensee (Tuebingen)<br />

10:20-10:50 S. Khleif (Bethesda)<br />

Combination of immunotherapy and molecular targeted therapy: manipulating the<br />

inhibitory arm of the immune system<br />

10:50-11:20 M. Tanaka (Yokohama)<br />

Role of Sinus macrophages in the induction of anti-tumor immunity<br />

11:20-11:50 D. Gabrilovitch (Tampa)<br />

Mechanism of synergistic effect of immunotherapy and chemotherapy<br />

11:50-12:20 M. Dhodapkar (Yale)<br />

Immunity to pluripotency genes in spontaneous and therapy induced immunity<br />

12:20-13:50 Lunch break<br />

Industry satellite symposium III<br />

(organized by Becton Dickinson)<br />

12:30 13:10 Dr. Andreas Weigert (Medizinische Fakultät der Goethe-Universität Frankfurt/<br />

Main, Institut für Biochemie 1/Pathobiochemie)<br />

Characterization of tumor infiltrating immune cell populations by multicolor flow<br />

cytometry<br />

13:10-13:50 Dr. Marieke Essers (DKFZ Heidelberg, HI-STEM-Heidelberg Institute for<br />

Stem Cell Technologies and Experimental Medicine)<br />

Activation of dormant hematopoetic stem cells<br />

Session 6: Immune monitoring<br />

(organized by the <strong>CIMT</strong> Immunoguiding Program)<br />

Chairperson: Cécile Gouttefangeas (Tuebingen), S. H. van der Burg (Leiden)<br />

13:50-14:20 M.-L. (Nora) Disis (Washington)<br />

Immunomodulation of breast cancer<br />

14:20-14:50 V. Appay (Paris)<br />

Assessing qualitative attributes of HIV specific CD8+T cells: from phenotype to clonotype<br />

14:50-15:20 W. Zou (Ann Arbor)<br />

T cells subsets in the tumor microenvironment<br />

15:20-15:45 Coffee break


Session 7: Poised, Suppressed, Tolerized–Mechanisms of T-cell Immunity<br />

Chairpersons: H.J. Schild (Mainz), W. Herr (Mainz)<br />

15:45-16:15 J. S. Bromberg (New York)<br />

Induction and migration of regulatory T cells<br />

16:15-16:45 G. Adema (Nijmegen)<br />

Regulatory T cells in immunotherapy of cancer<br />

16:45-17:15 P. A. Shrikant (Buffalo)<br />

MenTORed CD8+ T cells for tumor immunity<br />

Closing words: Christoph Huber & Hansjörg Schild<br />

19


Sponsors, Partners & Industry exhibitors<br />

We gratefully acknowledge the recurring support that reaches us every<br />

year from our sponsors, partners and industry exhibitors.<br />

Sponsors<br />

Merck<br />

Roche<br />

Novartis<br />

Partners<br />

Deutsche Krebshilfe<br />

FZI Mainz<br />

20<br />

Wyeth<br />

Beckman Coulter<br />

Ganymed Pharmaceuticals<br />

Cancer Immunotherapy Consortium<br />

Becton Dickinson<br />

Miltenyi Biotec<br />

Immatics<br />

Cancer Immunology Immunotherapy (CII)


Industry exhibitors<br />

R&D Systems<br />

CTL Europe<br />

Millipore<br />

Bachem<br />

JPT Peptide<br />

Peprotech<br />

Thymed<br />

Caliper Life Sciences<br />

Bio Rad<br />

CellGenix<br />

Meda Pharma<br />

Invitrogen Deutschland<br />

Becton Dickinson<br />

Miltenyi Biotec<br />

Invatis<br />

Beckman Coulter<br />

Bio Sys GmbH<br />

Qiagen GmbH<br />

Charles River<br />

eBioscience Company<br />

Sanquin Reagents<br />

Immudex<br />

21


Poster Presentations<br />

22<br />

General Information<br />

<strong>Abstract</strong>s that have been selected for poster presentation have been divided into seven sessions:<br />

Therapeutic vaccination<br />

Immune monitoring<br />

Cellular therapy<br />

New targets & new leads<br />

Tumor biology & interaction with the immune system<br />

Enhancing immunity & adjuvants<br />

Posters will be discussed in a guided poster session. Presenting authors should be prepared<br />

to present and discuss their posters (about 5-10 minutes time).


Guided Poster Sessions<br />

Session Date & time Chair Posters<br />

Therapeutic vaccination I<br />

Immune monitoring I<br />

Cellular therapy I<br />

New targets & new leads I<br />

Tumor biology & interaction<br />

with the immune system I<br />

Enhancing immunity &<br />

adjuvants<br />

Therapeutic vaccination II<br />

Immune monitoring II<br />

Cellular therapy II<br />

New targets & new leads II<br />

Tumor biology & interaction<br />

with the immune system II<br />

27.05.<strong>2010</strong>, 16:15 – 18:00 hrs<br />

27.05.<strong>2010</strong>, 16:15 – 18:00 hrs<br />

27.05.<strong>2010</strong>, 16:15 – 18:00 hrs<br />

27.05.<strong>2010</strong>, 16:15 – 18:00 hrs<br />

27.05.<strong>2010</strong>, 16:15 – 18:00 hrs<br />

27.05.<strong>2010</strong>, 16:15 – 18:00 hrs<br />

28.05.<strong>2010</strong>, 8:30 – 9:50 hrs<br />

28.05.<strong>2010</strong>, 8:30 – 9:50 hrs<br />

28.05.<strong>2010</strong>, 8:30 – 9:50 hrs<br />

28.05.<strong>2010</strong>, 8:30 – 9:50 hrs<br />

28.05.<strong>2010</strong>, 8:30 – 9:50 hrs<br />

V. Lennerz (Mainz)<br />

C. Bain (Lyon)<br />

H. Voß (Mainz)<br />

K. Thielemans (Brussels)<br />

S. Pascolo (Zurich)<br />

M. Radsak (Mainz)<br />

M. Schmitt (Rostock)<br />

S. Groß (Erlangen)<br />

R. Stripecke (Hannover)<br />

V. Umansky (Heidelberg)<br />

U. Hartwig (Mainz)<br />

001-012<br />

021-031<br />

L124<br />

040-050<br />

061-069<br />

078-092<br />

107-118<br />

L126<br />

013-020,<br />

L119-L122<br />

032-039<br />

051-060<br />

070-077,<br />

L123, L125<br />

093-106<br />

23


<strong>Abstract</strong> Nr. Title Poster session Oral short talk<br />

001 Bontkes<br />

002 Kotsiou<br />

003 Altvater<br />

004 Haenssle<br />

005 Vyth-Dreese<br />

006 Bernard<br />

007 Navarrete<br />

008 Schroeder<br />

009 Kuhn<br />

010 Mikyskova<br />

24<br />

Dendritic cells loaded with amplified mRNA isolated<br />

from leukemic stem cell like cells induce specific<br />

T-cells<br />

Antigen specific monoclonal antibodies directed<br />

against the HA-1 and HA-2 minor histocompatibility<br />

antigens<br />

Presentation of tumor-associated epitopes by activated<br />

human γδ T cells induces expansion of specific<br />

CD8+ T cells from naïve precursors<br />

Dendritic cells loaded with HSP70-expressing heatkilled<br />

melanoma cells efficiently activate autologous<br />

T cells<br />

MART-1 specific T cells after DNA tattoo vaccination<br />

of melanoma patients<br />

Investigating the impact of autophagy modulation<br />

on dendritic cell cancer vaccines<br />

Upfront Immunization With Autologous Recombinant<br />

Idiotype Fab Fragment Without Prior Cytoreduction<br />

in Indolent B-Cell Lymphoma<br />

In vitro characterization of an irradiated genetransfer<br />

medicinal product consisting of a prostate<br />

cancer derived cell line constitutively secreting<br />

interleukin-2 and interferon-gamma<br />

Phosphorothioate cap analogs increase stability and<br />

translational efficiency of RNA vaccines in immature<br />

dendritic cells and induce superior immune<br />

responses in vivo<br />

Interleukin 12 genetically-modified vaccines augment<br />

the effect of chemotherapy of human papilloma<br />

virus 16-associated tumours with gemcitabine<br />

Therapeutic vaccination I<br />

Therapeutic vaccination I<br />

Therapeutic vaccination I<br />

Therapeutic vaccination I<br />

Therapeutic vaccination I<br />

Therapeutic vaccination I<br />

Therapeutic vaccination I<br />

Therapeutic vaccination I<br />

Therapeutic vaccination I<br />

Therapeutic vaccination I


<strong>Abstract</strong> Nr. Title<br />

011 Kuhs<br />

012 Gueckel<br />

013 Babiarova<br />

014 Abbas<br />

015 Vogt<br />

016 van Nuffel<br />

017 Schaft<br />

018 Haenssle<br />

019 Buhl<br />

020 Nielsen<br />

L119 Dutoit<br />

L120 Hilf<br />

Clinical Development of lentiviral vector-induced<br />

“SMART-DC” for melanoma immunotherapy<br />

targeted modification of HLA-A*02-restricted mucin-<br />

1-derived peptides results in induction of cytotoxic<br />

T cells cross-reactive with naturally presented<br />

peptides<br />

Development of different types of vaccine against<br />

WT1 tumor antigen<br />

Vaccine development through antigen targeting to<br />

mannose receptor<br />

Inhibition of tumor growth after subcutaneous<br />

injection of dendritic cells engineered to express<br />

angiogenesis-related flk-1 antigen in a murine HCC<br />

model<br />

T cell specificities after vaccination of melanoma<br />

patients with mRNA loaded DC<br />

Optimization of the T-cell stimulation capacity of a<br />

dendritic-cell-based melanoma vaccine<br />

Dendritic cells loaded with HSP70-expressing heatkilled<br />

melanoma cells efficiently activate autologous<br />

T cells<br />

Cryopreservation of high-concentrated peripheral<br />

blood mononuclear cells by controlled-rate freezer<br />

results in higher cell yields for dendritic cell-based<br />

immunotherapy<br />

The CDX-1307 vaccine: regulatory and clinical steps<br />

in developing a regimen<br />

Functional immune reactivity to antigens of the ovel<br />

IMA950 vaccine peptide set in glioma patients<br />

IMA950 - a novel multi-peptide cancer vaccine for<br />

treatment of glioblastoma<br />

Poster session<br />

Therapeutic vaccination I<br />

Therapeutic vaccination I<br />

Therapeutic vaccination II<br />

Therapeutic vaccination II<br />

Therapeutic vaccination II<br />

Therapeutic vaccination II<br />

Therapeutic vaccination II<br />

Therapeutic vaccination II<br />

Therapeutic vaccination II<br />

Therapeutic vaccination II<br />

Therapeutic vaccination II<br />

Therapeutic vaccination II<br />

Oral short talk<br />

25


<strong>Abstract</strong> Nr. Title Poster session Oral short talk<br />

L121 Pawlowski<br />

L122 van de Ven<br />

021 Santegoets<br />

022 Pfirschke<br />

023 Veron<br />

024 Zupke<br />

025 Savelyeva<br />

026 Tanriverdi-<br />

Akhisaroglu<br />

027 Low<br />

028 Laske<br />

029 Brix<br />

030 Brix<br />

26<br />

Synergy of the immunomodulators GM-CSF and<br />

Imiquimod for peptide-based therapeutic vaccines<br />

Exposure of CD34+ precursors to cytostatic<br />

anthraquinone-derivatives induces rapid dendritic<br />

cell differentiation<br />

Immune and clinical response monitoring in patients<br />

with metastatic hormone-refractory prostate<br />

cancer receiving combined Prostate GVAX and anti-<br />

CTLA4 immunotherapy<br />

The repertoire of melanoma antigens recognized by<br />

T cell responses in malignant melanoma patients<br />

Selection, validation and evaluation of a 30-Plex<br />

Luminex kit for immunomonitoring of a phase I<br />

clinical trial<br />

Nanoparticles for efficient labeling of dendritic cells<br />

to be used in cellular immunotherapy<br />

Vaccination and immune memory: modelling the<br />

bystander activation of CD4+ memory T cells in<br />

mice<br />

Spontaneous tumour antigen specific T cell responses<br />

occur frequently in patients with Carcinoma<br />

of Unknown Primary and are predominantly<br />

directed against EGFR, telomerase, MAGE-3 and P53<br />

Culturing PBMC in the presence of antigen results<br />

in a measurable expansion of memory T cells<br />

T-cell immunomonitoring in patients with renal cell<br />

carcinoma after multi-peptide vaccination<br />

Sensitive and reliable monitoring of cancer specific<br />

T cell responses<br />

MHC Dextramers: improved detection of antigenspecific<br />

T-cells in fluid samples and in situ<br />

Therapeutic vaccination II<br />

Therapeutic vaccination II<br />

Immune monitoring I<br />

Immune monitoring I<br />

Immune monitoring I<br />

Immune monitoring I<br />

Immune monitoring I<br />

Immune monitoring I<br />

Immune monitoring I<br />

Immune monitoring I<br />

Immune monitoring I<br />

Immune monitoring I<br />

26.05.<strong>2010</strong>,<br />

15:50 – 16:05 hrs<br />

26.05.<strong>2010</strong>,<br />

16:05 – 16:20 hrs


<strong>Abstract</strong> Nr. Title<br />

L124 Zhang<br />

031 Gross<br />

032 Zelba<br />

033 Singh<br />

034 Guidoboni<br />

035 Voland<br />

036 Wagner<br />

037 Aarntzen<br />

038 Moodie<br />

039 Attig<br />

040 Kotsiou<br />

Goal of ELISPOT proficiency accomplished: ELI-<br />

SPOT assays provide reproducible results among<br />

different laboratories for T-cell immune monitoring<br />

— even in hands of ELISPOT novices<br />

Monitoring of vaccine-specific regulatory and helper<br />

CD4+ and cytolytic CD8+ T cells after vaccination<br />

with autologous antigen-loaded dendritic cells<br />

NY-ESO-1-specific T cells in long-term melanoma<br />

survivors<br />

A stable standard sample for harmonisation and<br />

validation of T cell assay Satwinder Kaur Singh,<br />

Changes of immune cell infiltrates induced by dendritic<br />

cell vaccination in melanoma patients‘ tumor<br />

tissues: an immunohistochemical study<br />

Characterisation and functional analysis of T-cell<br />

responses in melanoma patients vaccinated with<br />

peptide-loaded dentritic cells<br />

GPI-anchor negative T cells with impaired effector<br />

function persist after Alemtuzumab mediated T-cell<br />

depletion<br />

skin-derived functional specific Tcells predict clinical<br />

outcome upon DC vaccination in stage III and<br />

IV melanoma patients<br />

Response definition criteria for ELISPOT assays<br />

revisited<br />

Recommendations for HLA-peptide multimer<br />

staining experiments - Results from the second<br />

HLA-peptide multimer proficiency phase organized<br />

by the Cancer Immunotherapy Consortium<br />

Application of MHC class I single chain trimers for<br />

leukaemia immunotherapy<br />

Poster session<br />

Immune monitoring I<br />

Immune monitoring II<br />

Immune monitoring II<br />

Immune monitoring II<br />

Immune monitoring II<br />

Immune monitoring II<br />

Immune monitoring II<br />

Immune monitoring II<br />

Immune monitoring II<br />

Immune monitoring II<br />

Cellular therapy I<br />

Oral short talk<br />

26.05.<strong>2010</strong>,<br />

16:05 – 16:20 hrs<br />

26.05.<strong>2010</strong>,<br />

16:05 – 16:20 hrs<br />

26.05.<strong>2010</strong>,<br />

16:05 – 16:20 hrs<br />

27


<strong>Abstract</strong> Nr. Title Poster session Oral short talk<br />

041 Wittmann<br />

042 Weigand<br />

043 Liang<br />

044 Kalos<br />

045 Grange<br />

046 Singh<br />

047 Blaudszun<br />

048 Schendel<br />

049 Albrecht<br />

050 Klobuch<br />

28<br />

Comparing genetically engineered T cells for chimeric<br />

TCR versus CD16 + Trastuzumab for adoptive<br />

immunotherapy against HER2 positive breast<br />

carcinomas<br />

Generation of CD4+ T cells with specificity for<br />

FMNL1<br />

Transfer of human T-cell receptor (TCR) containing<br />

murine chimeric constant betagamma-chain sequences<br />

reduces the risk of mixed heterodimers and<br />

enhanced the proliferation of transduced T cells<br />

Clearance of established leukemia in a mouse xenograft<br />

model by a single injection of primary T cells<br />

gene-modified to express chimeric antigen receptors<br />

that target CD19<br />

Optimizing effector and memory properties of CD8<br />

T cells for adoptive tumor immunotherapy<br />

Search for FLT3- ITD-reactive CD8+ T cells in the<br />

peripheral blood of healthy donors<br />

Adoptive transfer of ex vivo activated and antineoplastic<br />

drug loaded T lymphocytes retargeted to<br />

EpCAM expressing tumours<br />

High avidity T cell clones specific for tumor-associated<br />

antigens and how to find them<br />

IL-21 treated naive CD45RA+ T cells represent a reliable<br />

source for generating AML-reactive cytotoxic<br />

T lymphocytes with high proliferative potential and<br />

early differentiation phenotype<br />

T cell receptor RNA transfer into non-reactive<br />

human T lymphocytes turns them into potent CMVspecific<br />

effector cells<br />

Cellular therapy I<br />

Cellular therapy I<br />

Cellular therapy I<br />

Cellular therapy I<br />

Cellular therapy I<br />

Cellular therapy I<br />

Cellular therapy I<br />

Cellular therapy I<br />

Cellular therapy I<br />

Cellular therapy I<br />

27.05.<strong>2010</strong><br />

10:50 – 11:05 hrs<br />

27.05.<strong>2010</strong><br />

11:05 – 11:20 hrs


<strong>Abstract</strong> Nr. Title<br />

051 Distler<br />

052 Salguero<br />

053 Stolle<br />

054 Bloetz<br />

055 Stumpf<br />

056 Hoyer<br />

057 Foerster<br />

058 Echchannaoui<br />

059 Bourquin<br />

060 Schmitt<br />

Alloreactivity to HLA class I mismatch alleles mainly<br />

resides in CCR7+ naive and central memory CD8<br />

T cells<br />

Dramatic early expansion of CMV-reactive human<br />

T cells in vivo is supported by lentiviral vector-induced<br />

engineered dendritic cells<br />

Generation of HCMV-/TAA-bispecific human T cells<br />

by either the genetic equipment of HCMV+ T cells<br />

with tumor-reactive TCRs or the combined retroviral<br />

transduction of bulk human T cells with HCMV-/<br />

TAA-specific TCRs<br />

Allo-reactivity to HLA class II mismatch alleles in<br />

CD4+ T-cell subsets<br />

Indirect presentation of HLA class II restricted minor<br />

histocompatibility antigens is a characteristic<br />

of HLA-DM resistant antigens<br />

Examination of T-helper cell / DC cross-talk by the<br />

transfection of T cells with RNA coding for TCRs<br />

Generation of multivirus-specific CD4+ and CD8+<br />

T cells for adoptive immunotherapy<br />

Evaluation of the safety of p53 TCR gene transfer in<br />

a humanized mouse model<br />

Efficient eradication of subcutaneous but not of<br />

autochthonous gastric tumors by adoptive T cell<br />

transfer in a SV40 T antigen mouse model<br />

WT1 and RHAMM specific CD8+ T cells can be<br />

isolated and purified by streptamers for adoptive<br />

transfer<br />

Poster session<br />

Cellular therapy II<br />

Cellular therapy II<br />

Cellular therapy II<br />

Cellular therapy II<br />

Cellular therapy II<br />

Cellular therapy II<br />

Cellular therapy II<br />

Cellular therapy II<br />

Cellular therapy II<br />

Cellular therapy II<br />

Oral short talk<br />

27.05.<strong>2010</strong><br />

11:20 – 11:35 hrs<br />

27.05.<strong>2010</strong><br />

11:35 – 11:50 hrs<br />

27.05.<strong>2010</strong><br />

11:50 – 12:05 hrs<br />

29


<strong>Abstract</strong> Nr. Title Poster session Oral short talk<br />

061 Buchwald<br />

062 Ashfield<br />

063 Woelfel<br />

064 Hornig<br />

065 Casares<br />

066 Dettmar<br />

067 Hirschhaeuser<br />

068 Guthmann<br />

069 Sørensen<br />

070 Andersen<br />

30<br />

The ubiquitin-conjugase-8 targets active fms-like<br />

tyrosine kinase-3 for proteasomal degradation<br />

ImmTACs: bi-functional reagents for redirected<br />

tumour cell killing<br />

Identification of acute myeloid leukemia-associated<br />

antigens recognized by allogeneic CD8+ T lymphocytes<br />

Combinatorial approach of a bispecific antibody<br />

and costimulatory antibody fusion proteins for<br />

targeted cancer immunotherapy<br />

Suppression of Treg activity by a FOXP3 inhibitor<br />

peptide<br />

Identification of clinically useful combinations of<br />

trifunctional antibodies with chemotherapy using<br />

different preclinical models<br />

Efficacy of catumaxomab in tumor spheroid killing<br />

is mediated by its trifunctional mode of action<br />

Cellular and humoral immune response to N-glycolyl-GM3<br />

elicited by racotumomab, an anti-idiotypic<br />

vaccine<br />

Cellular Immune Responses Against Indoleamine<br />

2,3-dioxygenase<br />

Identification of highly potent regulatory CD8+<br />

T-cells specific for Heme Oxygenase-1<br />

New targets & new leads I<br />

New targets & new leads I<br />

New targets & new leads I<br />

New targets & new leads I<br />

New targets & new leads I<br />

New targets & new leads I<br />

New targets & new leads I<br />

New targets & new leads I<br />

New targets & new leads I<br />

New targets & new leads I<br />

26.05.<strong>2010</strong><br />

15:50 – 16:05 hrs<br />

26.50.<strong>2010</strong><br />

16:05 – 16:20 hrs<br />

26.50.<strong>2010</strong><br />

16:20 – 16:35 hrs


<strong>Abstract</strong> Nr. Title<br />

071 Kleemann<br />

072 van Esch<br />

073 Suso<br />

074 Krug<br />

075 Lubojanski<br />

076 Staege<br />

077 Feger<br />

L123 Kyzirakos<br />

L125 Huijbers<br />

078 Maccalli<br />

079 Poschke<br />

080 Flores-<br />

Guzman<br />

Identification of T-cell defined varicella-zoster virus<br />

proteins<br />

T cell epitope discovery through HLA class I peptide<br />

ligand exchange and combinatorial coding<br />

Identification of novel CD4+ and CD8+ T cell<br />

epitopes from telomerase (hTERT)<br />

Transfer of mRNA encoding chimeric antigen receptors<br />

specific for MCSP into CD4+ and CD8+ T cells<br />

Identification of T cell-defined melanoma antigens<br />

Tumor antigens in Hodgkin Lymphoma<br />

Analysis of HPV L1 specific T-cell immunity<br />

Identification of novel EBV-specific MHC class-II<br />

epitopes<br />

Vaccination against the extra domain-B<br />

of fibronectin as a novel tumor therapy<br />

Definition of the immunological properties of cancer<br />

stem cells isolated from human glioblastoma<br />

Immature immunosuppressive CD14+HLA-DR-/low<br />

cells in melanoma patients are Stat3hi and overexpress<br />

CD80, CD83 and DC-Sign<br />

Potential cancer stem cells in ret transgenic mouse<br />

model of spontaneous melanoma<br />

Poster session<br />

New targets & new leads II<br />

New targets & new leads II<br />

New targets & new leads II<br />

New targets & new leads II<br />

New targets & new leads II<br />

New targets & new leads II<br />

New targets & new leads II<br />

New targets & new leads II<br />

New targets & new leads II<br />

Tumor biology & interaction<br />

with the immune system I<br />

Tumor biology & interaction<br />

with the immune system I<br />

Tumor biology & interaction<br />

with the immune system I<br />

Oral short talk<br />

26.50.<strong>2010</strong><br />

16:35 – 16:50 hrs<br />

26.50.<strong>2010</strong><br />

16:50 – 17:05 hrs<br />

26.05.<strong>2010</strong><br />

15:50 – 16:05 hrs<br />

26.05.<strong>2010</strong><br />

16:05 – 16:20 hrs<br />

31


<strong>Abstract</strong> Nr. Title Poster session Oral short talk<br />

081 Lion<br />

082 Bonertz<br />

083 Ge<br />

084 Chen<br />

085 Dubrot<br />

086 Chachibaia-<br />

Saginashvili<br />

087 Lutz-Nicoladoni<br />

088 Schmid<br />

089 Sha<br />

090 Strothmeyer<br />

32<br />

Poly(I:C)-electroporated myeloid leukemic cell lines<br />

become highly susceptible to NK cell-mediated killing<br />

and phagocytosis by immature DC<br />

Antigen-specific Treg in colorectal carcinoma<br />

control T cell responses against a limited tumor<br />

antigen repertoire<br />

tumor-induced emigration of antigen-specific treg<br />

enables the bone marrow to foster spontaneous<br />

anti-tumor t-cell responses in breast cancer patients<br />

Disturbed NK cell function in CML patients at<br />

diagnosis does not recover under treatment with<br />

imatinib mesylate<br />

Treatment with anti-CD137 mAbs causes intense<br />

accumulations of liver T cells without selective antitumor<br />

immunotherapeutic effects in this organ<br />

Identification of clinically useful combinations of<br />

trifunctional antibodies with chemotherapy using<br />

different preclinical models<br />

Reinforcement of Cancer Immunotherapy by Adoptive<br />

Transfer of cblb-deficient Cytotoxic T Lymphocytes<br />

combined with a Dentritic Cell Vaccine<br />

Development of a novel transgenic mouse model for<br />

melanoma<br />

Influence of activation of human immune effector<br />

cells on the penetration into tumor spheroids<br />

Comparative Analysis of Predicted HLA Binding<br />

of Immunoglobulin Idiotype Sequences Indicates<br />

T Cell-Mediated Immunosurveillance in Follicular<br />

Lymphoma<br />

Tumor biology & interaction<br />

with the immune system I<br />

Tumor biology & interaction<br />

with the immune system I<br />

Tumor biology & interaction<br />

with the immune system I<br />

Tumor biology & interaction<br />

with the immune system I<br />

Tumor biology & interaction<br />

with the immune system I<br />

Tumor biology & interaction<br />

with the immune system I<br />

Tumor biology & interaction<br />

with the immune system I<br />

Tumor biology & interaction<br />

with the immune system I<br />

Tumor biology & interaction<br />

with the immune system I<br />

Tumor biology & interaction<br />

with the immune system I<br />

26.50.<strong>2010</strong><br />

16:20 – 16:35 hrs<br />

26.05.<strong>2010</strong><br />

16:35 – 16:50 hrs<br />

26.05.<strong>2010</strong><br />

16:50 – 17:05 hrs


<strong>Abstract</strong> Nr. Title<br />

091 Ramacher<br />

092 Tomsitz<br />

093 Koslowski<br />

094 Quaglino<br />

095 Lanzardo<br />

096 Halama<br />

097 Fraszczak<br />

098 Trad<br />

099 Hemmerling<br />

100 Koop<br />

Immunosuppression in transgenic mouse melanoma<br />

model induced by myeloid derived suppressor<br />

cells<br />

Establishment of a pre-clinical NOD/LtSz-scid<br />

IL2Rgammac null (NSG) mouse model to evaluate<br />

immunotherapeutic strategies for acute myeloid<br />

leukemia (AML)<br />

Tumor-associated genomic DNA hypomethylation<br />

effects positive autoregulation of HIF-1α<br />

A miRNAs expression profiles during ErbB2 driven<br />

mammary carcinogenesis<br />

Attenuation of PI3K/Akt-mediated tumorigenic<br />

signals through PTEN activation by DNA vaccineinduced<br />

anti-ErbB2 antibodies<br />

Natural Killer Cells are decreased in Human Colorectal<br />

Cancer Tissue and Liver Metastases despite<br />

High Levels of NK-Recruiting Chemokines<br />

Killer dendritic cells inhibit Treg differentiation<br />

Tumor infiltrating CD11c+ dendritic cells suppress<br />

T cell activation<br />

Human Langerhans cells reconstitute in skin xenografts<br />

Downregulation of cancer/testis antigen 45 (CT45)<br />

in human HT1080 cells analysed by proteomic<br />

screening<br />

Poster session<br />

Tumor biology & interaction<br />

with the immune system I<br />

Tumor biology & interaction<br />

with the immune system I<br />

Tumor biology & interaction<br />

with the immune system II<br />

Tumor biology & interaction<br />

with the immune system II<br />

Tumor biology & interaction<br />

with the immune system II<br />

Tumor biology & interaction<br />

with the immune system II<br />

Tumor biology & interaction<br />

with the immune system II<br />

Tumor biology & interaction<br />

with the immune system II<br />

Tumor biology & interaction<br />

with the immune system II<br />

Tumor biology & interaction<br />

with the immune system II<br />

Oral short talk<br />

27.05.<strong>2010</strong><br />

10:50 – 11:05 hrs<br />

27.05.<strong>2010</strong><br />

11:05 – 11:20 hrs<br />

27.05.<strong>2010</strong><br />

11:20 – 11:35 hrs<br />

33


<strong>Abstract</strong> Nr. Title Poster session Oral short talk<br />

101 Karakhanova<br />

102 Zimmer<br />

103 Heidemann<br />

Krogh<br />

104 Hansmann<br />

105 Lupp<br />

106 Castle<br />

107 Sevko<br />

108 Diaz-Valdés<br />

109 Bauer<br />

110 Femel<br />

34<br />

IFN-α up-regulates B7-H1 expression on dendritic<br />

cells and pancreatic tumor cells<br />

Impact of endoplasmic reticulum aminopeptidases<br />

(ERAP) on T cell epitope generation in melanoma<br />

cells<br />

the phospho-regulated calcium-binding cancer-testis<br />

antigen cabyr, interacts with glycolysis-regulating<br />

enzymes in lung cancer cells<br />

Isolation of intact genomic DNA from FOXP3stained<br />

and FACS-sorted regulatory T cells for<br />

epigenetic analysis<br />

Mechanisms of Treg-mediated suppression of graftversus-host<br />

disease<br />

DNA copy number, including telomeres and mitochondria,<br />

assayed using next-generation sequencing<br />

Paclitaxel in ultra-low doses reduces immunosuppression<br />

in ret transgenic tumor bearing mice<br />

Transforming growth beta 1 increases monocyte<br />

chemotactin protein-1 and interleukin-10 expression<br />

in A375 human melanoma cells enhancing<br />

monocyte migration and impairing dendritic cell<br />

function<br />

The TLR9 ligand CpG reduces the suppressive<br />

function of myeloid-derived suppressor cells via<br />

interferon alpha<br />

Identification of a potent, non-toxic adjuvant for use<br />

in therapeutic vaccines<br />

Tumor biology & interaction<br />

with the immune system II<br />

Tumor biology & interaction<br />

with the immune system II<br />

Tumor biology & interaction<br />

with the immune system II<br />

Tumor biology & interaction<br />

with the immune system II<br />

Tumor biology & interaction<br />

with the immune system II<br />

Tumor biology & interaction<br />

with the immune system II<br />

Enhancing immunity &<br />

adjuvants<br />

Enhancing immunity &<br />

adjuvants<br />

Enhancing immunity &<br />

adjuvants<br />

Enhancing immunity &<br />

adjuvants<br />

27.05.<strong>2010</strong><br />

11:35 – 11:50 hrs<br />

27.05.<strong>2010</strong><br />

11:50 – 12:05 hrs


<strong>Abstract</strong> Nr. Title<br />

111 Diekmann<br />

112 Reinis<br />

113 Kermer<br />

114 Lehmann<br />

115 Lladser<br />

116 Hotz<br />

117 Gonzalez-<br />

Carmona<br />

118 Trojandt<br />

L126 Klier<br />

mTOR inhibition by Rapamycin after intranodal<br />

RNA immunization improves the CD8+ memory<br />

T-cell response qualitatively and quantitatively<br />

DNA methyltransferase inhibitors as modulators of<br />

anti-tumour immunity: implications for their use as<br />

adjuvants for immunotherapy<br />

Antibody fusion proteins for cancer immunotherapy<br />

mimicking IL-15 trans presentation at the tumor site<br />

Modified vaccinia virus Ankara (MVA) - a safe vector<br />

virus and an adjuvant system for immunotherapy<br />

DAI (ZBP1/DLM-1) as a novel genetic adjuvant for<br />

cancer DNA vaccines that promotes CTL responses,<br />

overcomes tolerance and confers long-term tumor<br />

protection.<br />

A rational protocol of repeated TLR7 stimulation circumvents<br />

induction of TLR tolerance and translates<br />

into efficient anti-tumor therapy<br />

Combining subcutaneous inoculation of AFPexpressing<br />

DC with intraperitoneal injection of IL-12expressing<br />

DC increases survival in an established<br />

orthotopic HCC model<br />

Effects of the anti-cancer agent topotecan on human<br />

monocyte-derived dendritic cells<br />

Combined bacterial antibody therapy for colorectal<br />

carcinoma<br />

Poster session<br />

Enhancing immunity &<br />

adjuvants<br />

Enhancing immunity &<br />

adjuvants<br />

Enhancing immunity &<br />

adjuvants<br />

Enhancing immunity &<br />

adjuvants<br />

Enhancing immunity &<br />

adjuvants<br />

Enhancing immunity &<br />

adjuvants<br />

Enhancing immunity &<br />

adjuvants<br />

Enhancing immunity &<br />

adjuvants<br />

Enhancing immunity &<br />

adjuvants<br />

Oral short talk<br />

35


36<br />

Therapeutic vaccination


001 Bontkes | Therapeutic vaccination<br />

Dendritic cells loaded with amplified mRNA isolated from<br />

leukemic stem cell like cells induce specific T-cells<br />

Hetty J. Bontkes, Jurjen M. Ruben, Willemijn A. van den Ancker, Theresia M. Westers, Gert<br />

J. Ossenkoppele, and Arjan A. van de Loosdrecht<br />

Department of Hematology , Cancer Center Amsterdam, V-ICI, VU University Medical Center,<br />

Amsterdam, The Netherlands<br />

It is thought that recurrent acute myeloid leukemia<br />

(AML) originates from chemotherapy resistant quiescent<br />

leukemic stem- or leukemia initiating cells<br />

(LSC). The application of immunotherapeutic approaches<br />

to eradicate LSC remaining after first line<br />

chemotherapy may contribute to improved disease<br />

outcome. Vaccination strategies have used dendritic<br />

cells (DC) ex vivo pulsed with tumor-derived<br />

whole lysates as modalities to present a broad range<br />

of tumor antigens to T cells to stimulate effective<br />

anti-tumor T-cell immunity in vivo. It is likely that<br />

certain proteins expressed by LSC have a distinct<br />

antigenicity as compared to more mature AML<br />

blasts and thus provide targets for specific T-cells.<br />

Even without identification of specific antigens,<br />

LSC can be a useful source of tumor antigens in DC<br />

vaccination-based immunotherapy. CD34+CD38-<br />

LSC can be identified using malignant stem cell<br />

associated cell surface markers including C-type<br />

lectin-like molecule-1 (CLL-1) and lineage markers<br />

such as CD7 and CD56. However, the low frequency<br />

of these cells precludes the use of LSC derived apoptotic<br />

cells or lysates for DC loading. Alternatively,<br />

mRNA isolated from LSC can be amplified and<br />

subsequently transfected into DC. We have made<br />

use of the HLA-A2 positive CD38- AML derived cell<br />

line MUTZ-3 which contains a subpopulation of<br />

CD34+CLL1+ cells which resembles the phenotype<br />

of a putative LSC. CLL1+CD34+ stem cell like cells<br />

as well as CLL1+CD34- CLL1-CD34+ and CLL1-<br />

CD34-cells were isolated by FACS sorting and total<br />

RNA was isolated. mRNA was converted to cDNA<br />

and amplified by PCR using the SMART system.<br />

Subsequently, mRNA was in vitro transcribed<br />

from the amplified cDNA. Amplification of CLL1<br />

and survivin transcripts was confirmed by RT-PCR.<br />

Mature monocyte derived DC (MoDC) were generated<br />

from HLA-A2 positive healthy donor blood and<br />

transfected with amplified CLL1+CD34+ derived<br />

mRNA and used to stimulate autologous CD8β+<br />

T-cells. After three weekly re-stimulations with<br />

CLL1+CD34+ mRNA transfected DC, specificity of<br />

the T-cells was analyzed either by IFNγ production<br />

upon stimulation with autologous immature MoDC<br />

transfected with GFP mRNA, mRNA amplified from<br />

total MUTZ-3 cells or sorted subpopulations, or by<br />

cytotoxicity towards MUTZ-3 subpopulations. The<br />

generated T-cells specifically produced IFNγ in response<br />

to MUTZ-3(subpopulation) mRNA loaded<br />

iDC but not in response to GFP mRNA transfected<br />

DC. Furthermore, the generated CTL killed the<br />

CLL1+CD34+ and CLL1+CD34- populations more<br />

efficiently compared to the CLL1-CD34+ populations.<br />

We show that MoDC transfected with RNA amplified<br />

from one MUTZ-3 sub-population resembling<br />

the phenotype of LCS cells are capable of inducing<br />

T-cells which recognize both cells transfected with<br />

mRNA from the LSC resembling MUTZ-3 subset as<br />

well as the more mature subsets. The efficacy and<br />

feasibility of this approach is currently tested in an<br />

autologous AML setting in vitro.<br />

37


002 Kotsiou | Therapeutic vaccination<br />

Antigen specific monoclonal antibodies directed against the<br />

HA-1 and HA-2 minor histocompatibility antigens<br />

Eleni Kotsiou 1,2 , Jonathan Silk 3 , Vincenzo Cerundolo 3 , Julian Dyson 2 , and Keith G. Gould 1<br />

1<br />

Department of Immunology, Wright-Fleming Institute, St Mary‘s Campus, Imperial College,<br />

London W2 1PG, UK<br />

2<br />

Immunobiology Section, Commonwealth Building, Hammersmith Hospital, Imperial College,<br />

London W12 0NN, UK<br />

3<br />

Tumour Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford,<br />

Oxford OX3 9DS, UK<br />

38<br />

Monoclonal antibodies with the specificity of a T<br />

cell receptor (TCR) (also known as TCR mimic antibodies)<br />

can be used for the targeting of tumor or<br />

viral epitopes and are potentially very useful in the<br />

clinical setting.<br />

The HA-1 and HA-2 minor histocompatibility (H)<br />

antigens are restricted by human leukocyte antigen<br />

(HLA) A2 and expressed only on normal and malignant<br />

haematopoietic cells. We have produced<br />

soluble (lacking cytoplasmic and transmembrane<br />

regions) HA-1 and HA-2 HLA-A2 single chain<br />

trimers (SCTs) where the peptide, β2-microglobulin<br />

and major histocompatibility complex (MHC) heavy<br />

chain are covalently linked together via glycine/<br />

serine rich linkers. We then used the soluble SCT<br />

proteins to immunize HHD (HLA-A2 transgenic)<br />

mice for the production of antigen specific monoclonal<br />

antibodies by taking advantage of the ‘humanized’<br />

immune system of the transgenic mice<br />

which will recognize the HLA-A2 molecule as ‘self’<br />

but the peptide epitope as foreign.<br />

The screening of the serum of the immunized mice<br />

(after three rounds of immunization) revealed the<br />

presence of antibodies preferentially reacting with<br />

the HA-1 and HA-2 epitopes. The screening of hybridoma<br />

clones produced after fusion of spleen cells<br />

from innunized mice with myeloma cells showed<br />

that, rather than recognising the peptide, the majority<br />

of the monoclonal antibodies were directed<br />

against other immunogenic epitopes present in the<br />

sequence of the HLA-A2 SCTs.<br />

The development of new SCT fusion proteins which<br />

can potentially improve the immunization strategy<br />

will be described.


003 Altvater | Therapeutic vaccination<br />

Presentation of tumor-associated epitopes by activated human<br />

γδ T cells induces expansion of specific CD8+ T cells from<br />

naïve precursors<br />

Bianca Altvater 1 , Silke Landmeier 1 , Sibylle Pscherer 1 , Anna Hansmeier 1 , Barbara Savoldo 2 ,<br />

H. Juergens 1 , Claudia Rossig 1<br />

1 University Children´s Hospital Münster, Department of Pediatric Hematology and Oncology,<br />

Albert-Schweitzer-Str. 33, 48149 Münster<br />

2 Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, U.S.A<br />

Efficient antigen presentation is an important<br />

prerequisite for the induction of therapeutic T-cell<br />

immunity against viral and tumor antigens. In an<br />

Epstein Barr virus (EBV) model, we previously demonstrated<br />

that bisphosphonate-activated γδ T cells<br />

are potent stimulators of functional EBV-specific T<br />

cell responses in both normal donors and Hodgkin<br />

lymphoma patients. Here, we extended these observations<br />

by investigating the capacity of activated γδ<br />

T cells to induce primary T-cell responses against<br />

the tumor-associated, poorly immunogeneic antigens<br />

PRAME and STEAP-1 from the naive T-cell<br />

repertoire.<br />

Peripheral blood-derived γδ T cells were expanded<br />

from three individual HLA-A2 positive healthy<br />

donors by stimulation with the aminobisphosphonate<br />

zoledronic acid (1 µg/ml) in the presence of<br />

rhIL-2 (100 U/ml) and rhIL-15 (10 ng/ml). To investigate<br />

the ability of activated γδ T cells to stimulate<br />

expansion of PRAME- and STEAP-1 specific<br />

cytotoxic T cells (CTL) in vitro, purified autologous<br />

CD8+ T cells were cocultured with either dendritic<br />

cells (DCs) or activated γδ T cells pulsed with<br />

pools of HLA-A2 restricted peptides derived from<br />

either STEAP-1 or PRAME, followed by two stimulations<br />

with peptide-pulsed K-562 cells, genemodified<br />

to express human HLA-A2, CD80, CD40L,<br />

and OX40L (K-562-APCs). ELISPOT analysis demonstrated<br />

efficient and equivalent induction of<br />

specific CD8+ T-cell responses by both DCs and<br />

γδ T-APCs. Both types of CTLs had a predominant<br />

effector memory phenotype (CCR7-CD45R0+).<br />

Even though antigen-experienced precursor CTLs<br />

against the tumor-associated antigens are absent in<br />

the majority of healthy donors, these experiments<br />

do not formally prove that the expanded T cells are<br />

a consequence of T-cell priming. To demonstrate<br />

the origin of CTLs from antigen-naïve T cells, we<br />

performed additional coculture experiments using<br />

CD45RA+ T cells, purified from the CD8+ T-cell<br />

population by magnetic cell selection, as responder<br />

cells. Again, ELISPOT analysis after primary stimulation<br />

with PRAME peptide-pulsed autologous<br />

DCs or γδ T-APCs demonstrated specific and potent<br />

CTL reactivity against both pooled and individual<br />

PRAME peptides. Importantly, γδ T-APC induced<br />

CTLs were not inferior to those generated using the<br />

DC gold standard.<br />

Thus, γδ T cells represent a promising source of<br />

highly efficient professional APC for antigen-specific<br />

immunotherapy of cancer. In current experiments,<br />

we are establishing the capacity of γδ T<br />

cells endogenously expressing STEAP-1 or PRAME<br />

full-length antigen in inducing functional primary<br />

CTL responses.<br />

39


004 Haenssle | Therapeutic vaccination<br />

Dendritic cells loaded with HSP70-expressing heat-killed melanoma<br />

cells efficiently activate autologous T cells<br />

Holger A. Haenssle 1 , Susanne Knudsen 1 , Anke Schardt 2 , Timo Buhl 1 , Lars Boeckmann 1 ,<br />

Michael P. Schön 1<br />

1 Department of Dermatology and Venereology, Georg August University, Göttingen, Germany<br />

2 Max Planck Institute for Experimental Medicine, Göttingen, Germany<br />

40<br />

There is considerable interest to develop strategies<br />

that enhance cross-presentation pathways of dendritic<br />

cells (DCs) to elicit a strong cytotoxic T cell<br />

activation for cancer vaccination protocols.<br />

In order to best exploit the enhanced cross-presentation<br />

of antigens bound to heat shock protein 70<br />

(HSP70), we analyzed melanoma cell preparations<br />

for their HSP70 expression. For generation of heatnecrotic<br />

cell material melanoma cells were kept at<br />

42°C for 90 min and then heated to 56°C for a final<br />

30 min. Apoptotic Annexin V + /Propidium iodide–<br />

melanoma cells were generated by irradiation with<br />

1.0 J/cm2 UV-B. Western blotting revealed strong<br />

upregulation of HSP70 after heat-killing in contrast<br />

to UV-B irradiation. The uptake of fluorescently<br />

labeled heat-killed necrotic vs. apoptotic melanoma<br />

cells by DCs at various levels of maturation was assessed.<br />

Immature DCs (iDCs) internalized HSP70expressing<br />

necrotic material to a much higher extent<br />

(61% ± 7%) than apoptotic material from UV-Birradiated<br />

cells (48% ± 5%). Maturation-inducing<br />

cytokines did not affect the uptake when added<br />

simultaneously with the tumor cell preparations.<br />

Loading DCs with heat-necrotic or apoptotic melanoma<br />

cells slightly reduced CD83 expression while<br />

leaving CD208 (DC-LAMP) expression unchanged.<br />

As determined by IFN-γ-detecting ELISPOT assays,<br />

iDCs loaded with HSP70-expressing heat-killed<br />

melanoma cells activated autologous T cells most<br />

effectively when used without any further maturation,<br />

whereas DCs loaded with apoptotic material<br />

required maturation.<br />

In conclusion, HSP70-expressing melanoma cells<br />

could be generated by heat-killing. Loading immature<br />

DCs with heat-killed melanoma cells resulted<br />

in a superior priming of autologous T cells<br />

in vitro.


005 Vyth-Dreese | Therapeutic vaccination<br />

MART-1 specific T cells after DNA tattoo vaccination of<br />

melanoma patients<br />

Florry Vyth-Dreese 1 , Martin van der Maas 1 , Willeke van de Kasteele 1 , Trees Dellemijn 1 ,<br />

Sandra Adriaansz 2 , Henk Mallo 2 , Bastiaan Nuijen 3 , Christian Ottensmeier 4 , Lindsey Low 4 ,<br />

Christian Blank 1,2 , Ton Schumacher 1 and John Haanen 1,2<br />

1 Division of Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital,<br />

1066 CX Amsterdam, The Netherlands<br />

2 Medical Oncology Department, The Netherlands Cancer Institute-Antoni van Leeuwenhoek<br />

Hospital, 1066 CX Amsterdam, The Netherlands<br />

3 Pharmacy, Slotervaart Hospital, 1066 EC Amsterdam, The Netherlands<br />

4 Cancer Sciences Division, University of Southampton, Southampton SO16 6YD, United Kingdom<br />

Recently, we developed a novel, rapid and potent<br />

intradermal DNA vaccination method, called DNA<br />

tattooing. Previous studies in experimental systems<br />

showed a strong increase in vaccine-specific T cells<br />

compared to intramuscular vaccination, parallelled<br />

by robust anti-tumor T cell responses and rejection<br />

of established subcutaneous tumors. These<br />

results prompted us to initiate a phase I clinical<br />

study with an inhouse manufactured GMP-grade<br />

DNA vaccine, pDermatt. Here we report data from<br />

the immunomonitoring of our first phase I clinical<br />

trial of DNA tattoo vaccination of stage IV melanoma<br />

patients using this DNA vaccine encoding a<br />

tetanus toxin fragment c-modified MART-1 epitope<br />

fusion protein.<br />

Sofar, six patients were treated with DNA tattoo<br />

vaccination on day 0, 3 and 6 as a prime and on day<br />

28, 31 and 34 as a boost vaccination. Per cohort of<br />

3 patients the vaccine dose was doubled from 0.5<br />

mg to 1 mg DNA per tattoo. Flow cytometry and<br />

EliSpot assays were performed to examine the presence<br />

and specificity of T cells in peripheral blood<br />

samples, taken before therapy and at week 2, 4, 6<br />

and 8. Biopsies were taken from the vaccination site<br />

at week 2 and 6 and examined for the presence of<br />

immune cell subsets and specific T cells by immunohistochemistry<br />

and flow cytometry.<br />

To enable flow cytometric analysis for specific T<br />

cells, biopsies were taken from the vaccination<br />

site and cultured in vitro for 2 weeks in high dose<br />

IL-2 (6000 IU/ml). In 5/6 patients, compared to 0/6<br />

normal skin biopsies obtained from breast cancer<br />

operations, this resulted in outgrowth of MART-<br />

1-specific CD8 T lymphocytes, with 3/5 patient<br />

samples showing 30-50% MART-1 specific CD8 T<br />

cells. Highest numbers were obtained from week<br />

6 biopsies with a preferential increase in MART-1<br />

modified specific T cells.<br />

Virtually no therapy induced increases were observed<br />

in percentages of MART-1 specific peripheral<br />

blood CD8 T cells. EliSpot analysis revealed therapy<br />

induced IFNγ responses to MART-1 modified and<br />

wild type peptides in 2/3 patients tested in cohort<br />

1. These responses followed similar, although<br />

delayed, kinetics compared to tetanus fragment c<br />

responses which served as internal controls.<br />

Examination of immune cells at patient vaccination<br />

sites showed similar numbers of CD8 T cells, compared<br />

to normal skin, located in the subepidermal<br />

or dermal, but not epidermal areas. An increase<br />

was found for activated DC expressing CD80. From<br />

these data it is concluded that DNA tattoo vaccination<br />

induced MART-1 specific CD8 T cell migration<br />

to the vaccination site.<br />

41


006 Bernard | Therapeutic vaccination<br />

Investigating the impact of autophagy modulation on dendritic<br />

cell cancer vaccines<br />

Dannie Bernard 1 , Julian J. Lum 2 , Yonghong Wan 1 and Jonathan L. Bramson 1<br />

1 Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, L8N 3Z5, Canada<br />

2 Deeley Research Center, BC Cancer Agency, Vancouver Island, British Columbia, V8R 6V5, Canada<br />

42<br />

Autophagy has been linked to extended survival<br />

when cells are faced with cellular stress. Ex-vivo<br />

derived dendritic cells (DCs) undergo substantial<br />

stress upon antigen loading and in-vivo delivery<br />

and the importance of autophagy in protecting<br />

DCs from these stresses is poorly understood.<br />

We have employed 2 strategies to investigate the<br />

impact of autophagy on DC vaccines. In the first<br />

case, we employed rapamycin as an inducer of autophagy<br />

in an effort to precondition the DCs prior<br />

to infection with recombinant adenovirus (Ad)<br />

and recombinant vesicular stomatitis virus (VSV).<br />

Kinetic analyses showed that rapamycin was effective<br />

at inhibiting its target mTOR within 5 hours of<br />

treatment and the inhibition was maintained for at<br />

least 24 hours post-transduction. Preconditioning<br />

of DCs with rapamycin did not affect infectivity,<br />

as measured by GFP expression, nor did it affect<br />

maturation, defined by MHC II, CD40, CD86, IL-12<br />

and TNF-a expression. However, we noted a 10-fold<br />

reduction in type I interferon secretion following<br />

VSV transduction, but not Ad transduction. We<br />

are currently conducting in-vivo experiments with<br />

rapamycin-preconditioned DC vaccines and results<br />

will be discussed at the meeting. In the second<br />

case, we have investigated the role of basal autophagy<br />

in the context of DC vaccination. We have<br />

crossed ATG5fl/fl mice with CD11c-Cre mice to generate<br />

conditional knockout mice in which DCs are<br />

autophagy-deficient. Results obtained thus far from<br />

in-vitro phenotyping experiments of virally trans-<br />

duced DCs indicate that autophagy deficiency does<br />

not significantly impact maturation of the cells.<br />

The importance of autophagy following delivery of<br />

DC vaccines still remains to be determined. This<br />

work was supported by grants from CIHR, OCRN<br />

and TFF.


007 Navarrete | Therapeutic vaccination<br />

Upfront Immunization With Autologous Recombinant Idiotype<br />

Fab Fragment Without Prior Cytoreduction in Indolent B-Cell<br />

Lymphoma<br />

Marcelo Navarrete 1 , Kristina Heining-Mikesch 1 , Frank Schüler 2 , Cristina Bertinetti-Lapatki 1 ,<br />

Gabriele Ihorst 3 , Andrea Keppler-Hafkemeyer 1 , Gottfried Dölken 2 , Hendrik Veelken 1<br />

1 Department of Hematology/Oncology, University Medical Center Freiburg, Freiburg, Germany<br />

2 Department of Hematology and Oncology, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany<br />

3 Department of Medical Biometry and Statistics, University Medical Center Freiburg, Freiburg, Germany<br />

The monoclonal immunoglobulin ("Idiotype", Id)<br />

of B-cell lymphomas constitutes a tumor-specific<br />

antigen. Id immunization of follicular lymphoma<br />

(FL) patients as remission maintenance after conventional<br />

chemotherapy induces anti-Id antibodies<br />

that are associated with improved PFS. To ease<br />

the challenge of manufacturing Id vaccines on a<br />

patient-individual basis, we have developed the<br />

expression of Id protein as Fab fragment (FabId)<br />

in E. coli based on validated Ig clonality criteria<br />

(Bertinetti et al., EJH 2006). In a phase I trial, we<br />

have demonstrated the feasibility and tolerability<br />

of intradermal injection of FabId with the adjuvant<br />

MF59 in conjunction with subcutaneous GM-CSF<br />

(Bertinetti et al., Cancer Res 2006).<br />

Since the effects of upfront Id vaccination in lymphoma<br />

patients without prior chemotherapy are<br />

unknown, we conducted a phase II trial designed<br />

to demonstrate the immunological efficacy of FabId<br />

vaccination as upfront intervention for asymptomatic<br />

lymphoma patients and for remission consolidation.<br />

Given the strong but indirect evidence for<br />

Id-directed, T cell-mediated immuno-surveillance<br />

in FL (see poster by Strothmeyer et al.), we investigated<br />

vaccination-induced T cell responses. 21<br />

patients with untreated indolent B-cell lymphoma<br />

and 20 patients in remission after cytoreductive<br />

therapy received 6 intradermal injections of FabId<br />

with subcutaneous sargramostim. Circulating<br />

antigen-reactive T cells were measured by IFNγ<br />

ELISpot that was validated by blinded interlabo-<br />

ratory testing (Britten et al., CII 2008). Humoral<br />

immune responses were assessed by ELISA.<br />

FabId vaccination induced immune responses in<br />

76% of untreated and in 80% of remission patients.<br />

Poisson regression analyses and epitope mapping<br />

demonstrated specificity of T cell responses for<br />

individual Id features. In untreated patients, cellular<br />

immunity was associated with superior PFS<br />

(p=0.002) and with durable remissions in 6 of 15<br />

FL (p=0.04). In remission maintenance, anti-Id antibodies<br />

correlated with PFS (p=0.02). Prior rituximab<br />

and low B-cell counts were associated with<br />

failure to develop anti-Id antibodies. FabId immunization<br />

effectively induces immune responses in<br />

both pretreated and untreated lymphoma patients.<br />

Similar to other Id vaccines, humoral immunity is<br />

associated with control of residual lymphoma. In<br />

contrast, cellular anti-Id immunity may be effective<br />

against untreated lymphoma. Sustained remissions<br />

in patients with vaccination-induced cellular immunity<br />

suggest clinical benefit and warrant randomized<br />

trials comparing this vaccine with expectant<br />

management as upfront strategy for asymptomatic<br />

follicular lymphoma.<br />

43


008 Schroeder | Therapeutic vaccination<br />

In vitro characterization of an irradiated gene-transfer medicinal<br />

product consisting of a prostate cancer derived cell line<br />

constitutively secreting interleukin-2 and interferon-gamma<br />

Petra Schroeder 1 , Carsten Lindemann 1 , Uwe Irmer 1 , Henning Weigt 2 , Bernd Eisele 2 , Klaus Kuehlcke 1<br />

1 EUFETS GmbH, Vollmersbachstraße 66, 55743 Idar-Oberstein, Germany<br />

2 Vakzine Projekt Management GmbH, Mellendorfer Straße 9, 30625 Hannover, Germany<br />

44<br />

In a first in man Phase I/II study, a prostate cancer<br />

cell line constitutively secreting the pro-inflammatory<br />

cytokines interferon-gamma (IFN-γ) and interleukin-2<br />

(IL-2) has shown anti-tumor response<br />

(median PSA doubling time prolonged from 63 days<br />

to 114 days, p = 0.0035; Brill et al., J Gene Med<br />

2007 and Hum Gene Ther 2009). Further drug development<br />

requires thorough characterization of<br />

the tumour vaccine. We designed a non-clinical<br />

program regarding aspects of safety, stability and<br />

the proof of principle of the tumour vaccine adopting<br />

an in vitro approach based on human cells<br />

to rectify the limitations of homologous animal<br />

models. A clinical scale manufacturing process<br />

including centralized irradiation was developed<br />

in parallel. Considering safety and stability of the<br />

tumour vaccine we tested the viability, colony formation,<br />

phenotype (surface marker expression of<br />

the prostate specific antigens PSA, PSMA, EpCAM),<br />

the release and bioactivity of IL-2 and IFN-γ, vector<br />

integrity and the release of replication competent<br />

retrovirus besides the generic tests stipulated for<br />

such products. The absence of tumourigenicity,<br />

e.g. outgrowth of potentially remaining replication<br />

competent cells, of the finally irradiated cells was<br />

controlled by the assessment of proliferating cells<br />

in long term cultures and proliferation assays. In<br />

order to show the induction of an immunological<br />

reaction which is considered as a prerequisite to<br />

induce an anti-tumour response we evaluated the<br />

allogenic response towards the irradiated cells,<br />

analysed the deposition of complement and the<br />

uptake of cellular particles by phagocytes. The stability<br />

and safety as well as the functionality and biologic<br />

activity of the cellular product could be reliably<br />

demonstrated. Results of phenotyping, colony<br />

formation, bioassays and the proof of principle will<br />

be presented.


009 Kuhn | Therapeutic vaccination<br />

Phosphorothioate cap analogs increase stability and translational<br />

efficiency of RNA vaccines in immature dendritic cells and<br />

induce superior immune responses in vivo<br />

Andreas N. Kuhn 1 , Mustafa Diken 1 , Sebastian Kreiter 1 , Abderraouf Selmi 1 , Joanna Kowalska 2 ,<br />

Jacek Jemielity 2 , Edward Darzynkiewicz 2 , Christoph Huber 1 , Özlem Türeci 1 , and Ugur Sahin 1<br />

1 Department of Internal Medicine III, Division of Translational and Experimental Oncology,<br />

University Medicine Mainz, Germany<br />

2 Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Poland<br />

Vaccination with in vitro transcribed RNA coding<br />

for tumor antigens is considered a promising approach<br />

for cancer immunotherapy and has already<br />

entered human clinical testing.<br />

One of the basic objectives for development of RNA<br />

as a vaccine is the optimization of immunobioavailability<br />

of the encoded antigen in vivo. In previous<br />

work, we have developed plasmid templates<br />

for in vitro transcription of RNA-encoded antigens<br />

with modified 3‘ structures (3‘ UTR and poly(A)tail)<br />

stabilizing the RNA and optimizing its translational<br />

performance in dendritic cells (DCs), the<br />

major antigen-presenting cells (1). Moreover, we<br />

achieved significant leverage of MHC class I and II<br />

presentation of the antigen in DCs by introducing<br />

routing signals (2). By combining these measures<br />

we were able to profoundly improve properties of<br />

RNA-encoded vaccines.<br />

Yet another important structural element of in vitro<br />

transcribed RNA is the 5‘ cap structure. Capping of<br />

in vitro synthesized RNA is achieved by transcribing<br />

the DNA template in the presence of the cap<br />

dinucleotide m7GpppG as structural homolog of the<br />

endogenous cap structure. Recently, several synthetic<br />

cap analogs have been described with modifications<br />

that influence stability and translational<br />

characteristics of the respective RNA species.<br />

Up to now, such modified cap analogs have been<br />

primarily employed to study RNA metabolism.<br />

Their impact, however, on bioavailability of the<br />

encoded protein in antigen presenting cells such<br />

as DCs and eventually on induction of potent T-cell<br />

responses in vivo has never been investigated. Therefore<br />

we have preclinically evaluated novel cap<br />

analogs for immunopharmacological improvement<br />

of vaccines based on antigen-encoding RNA. We<br />

demonstrate that RNAs capped with the D1 diastereoisomer<br />

of m27,2‘-OGppSpG (beta-S-ARCA) have<br />

increased stability and translational efficiency in<br />

immature but not mature DCs. Accordingly, in vivo<br />

delivery of the antigen as beta-S-ARCA(D1)-capped<br />

RNA species led to increased protein expression<br />

and enhanced priming and expansion of naïve antigen-specific<br />

T-cells in mice. We expect that our<br />

findings pave the way for inauguration of modified<br />

cap analogs into RNA vaccine development and<br />

will likely contribute to a better clinical outcome.<br />

(1) Holtkamp et al. (2006) Blood 108: 4009-17<br />

(2) Kreiter et al. (2008) J Immunol 180: 309-18<br />

45


010 Mikyskova | Therapeutic vaccination<br />

Interleukin 12 genetically-modified vaccines augment the effect<br />

of chemotherapy of human papilloma virus 16-associated<br />

tumours with gemcitabine<br />

Romana Mikyšková, Jana Šímová, Marie Indrová, Jan Bubeník, Jana Bieblová, Milan Reiniš<br />

Department of Tumour Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic,<br />

Prague, Czech Republic<br />

46<br />

Experiments were designed to examine whether<br />

local administration of genetically-modified interleukin<br />

12 (IL-12)-producing vaccine can enhance<br />

the chemotherapy of human papilloma virus (HPV)<br />

16-associated tumour with gemcitabine, a cytostatic<br />

agent that is known to reduce myeloid derived<br />

suppressor cells. Major histocompatibility class I<br />

(MHC I) positive non-metastasizing murine carcinoma<br />

TC-1 model was used to examine the effect<br />

of local IL-12 gene therapy in the minimal residual<br />

tumour disease induced by intraperitoneal cytoreductive<br />

therapy with gemcitabine. Mice with<br />

established 0.2-0.3 cm in diameter TC-1 tumours<br />

were treated with gemcitabine and, subsequently,<br />

with irradiated, IL-12-producing tumour cells.<br />

It was found that local administration of IL-12producing<br />

vaccine enhanced the effect of cytoreductive<br />

therapy with gemcitabine in TC-1 (MHC I<br />

positive) tumours. To investigate the antitumour<br />

and antimetastatic effect of IL-12 plus gemcitabine<br />

combination in another clinically relevant setting,<br />

surgical minimal residual tumour disease of MHC<br />

I-deficient MK16 tumours was used. Mice with established<br />

1 cm in diameter MK16 tumours were<br />

operated and subsequently treated with gemcitabine<br />

and/or IL-12-producing vaccine. Combination of<br />

the IL-12 genetically-modified tumour vaccine with<br />

gemcitabine treatment after surgery significantly<br />

reduced the percentage and size of MK16 tumour<br />

recurrences as well as the percentage and number<br />

of lung metastases, as compared to the group of<br />

operated-only mice and groups treated with gemcitabine<br />

or IL-12-producing vaccine alone. The therapeutic<br />

effectiveness exerted by the gemcitabine plus<br />

IL-12 combination in the MK16 tumour model was<br />

demonstrated by the detection of interferon gamma<br />

(IFNγ) production by spleen cells using ELISPOT<br />

and ELISA test. The highest production of IFNγ was<br />

found in the group treated with gemcitabine plus<br />

IL-12-producing cells. The percentage of myeloid<br />

derived suppressor cells (CD11b+/Gr-1+ cells)<br />

was analyzed three days after administration of<br />

the IL-12-producing vaccine. The lowest percentage<br />

of CD11b+/Gr-1+ was found in the group treated<br />

with the combination of gemcitabine plus IL-12producing<br />

cells. Taken together, we conclude that<br />

IL-12 augments the effectiveness of chemotherapy<br />

of HPV16-associated tumours with gemcitabine<br />

and increases the anti-tumour immune response.<br />

This work was supported by grants No. 301/09/1024 and No.<br />

301/07/1410 from the Grant Agency of the Czech Republic and by<br />

grant of the Clinigene project EU-FP6-NOE No. 018933.


011 Kuhs | Therapeutic vaccination<br />

Clinical Development of lentiviral vector-induced „SMART-DC“<br />

for melanoma immunotherapy<br />

Sandra Kuhs 1 , Ralf Gutzmer 2 , Bala Sai Sundarasetty 1 , Sylvia Borchers 1 , Gustavo Salguero 1 ,<br />

Arnold Ganser 1 , Rainer Blasczyk 3 , Henk Garritsen 4 , Thomas Woelfel 5 , Farzin Farzaneh 6 and<br />

Renata Stripecke 1<br />

1 Dept. of Hematology, Hemostasis, Oncology and Stem Cell Transplantation<br />

2 Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Poland<br />

3 Dept. of Transfusion Medicine, Hannover Medical School<br />

4 Institute for Clinical Transfusion Medicine, Städtisches Klinikum Braunschweig gGmbH<br />

5 Hematology and Oncology, University of Mainz, Germany<br />

6 Dept. of Hematological Medicine, Kings College, London, UK<br />

Clinical ex vivo production of dendritic cells (DC)<br />

is costly, time-consuming and difficult for largescale<br />

clinical trials. In addition, ex vivo grown DCs<br />

are not highly viable once re-infused in the body<br />

and antigen-presentation and bio-distribution are<br />

sub-optimal. We developed a novel technology for<br />

production of DC consisting of lentiviral vector<br />

(LV) transduction of growth factors and full-length<br />

antigens into monocytes that results into induction<br />

of “SMART-DCs” (Self-differentiated Myeloidderived<br />

Antigen-presenting-cells Reactive against<br />

Tumors). This concept has been extensively tested<br />

and validated in pre-clinical melanoma mouse<br />

models for safety and for induction of protective<br />

immunity. Here, self-inactivating lentiviral vectors<br />

containing interspacing 2A elements and a nonencoding<br />

Wpre were produced in a bicistronic (coexpressing<br />

simultaneously human GM-CSF and<br />

IL-4) or tricistronic design (expressing in addition<br />

the melanoma-associated antigens MART-1 or<br />

TRP-2). Co-expression of all the transgenes was validated<br />

in 293T cells transduced with these vectors.<br />

Overnight transduction of fresh or cryopreserved<br />

CD14+ monocytes resulted in persistent autocrine<br />

production of GM-CSF (average 7 ng/ml) and IL-4<br />

(average 0.8 ng/ml), inducing the self-differentiation<br />

of long-lived (up to 3 weeks) human SMART-<br />

DCs, which fully recapitulated the immunophenotype<br />

of conventional DCs. For future GMP process<br />

development, we evaluated monocyte transduction<br />

in a closed GMP-grade bag system. We were able<br />

to demonstrate that SMART-DCs were produced<br />

more effectively in bags than in culture flasks. Monocytes<br />

co-transduced with the bicistronic vector<br />

plus a vector expressing full length MART-1 were<br />

recognized by T cell clones specific for MART-1<br />

in an HLA-restricted manner, assessed by IFN-γ-<br />

ELISPOT-Assay. PBMCs that were primed/boosted<br />

in vitro with autologous SMART-DCs expressing<br />

MART-1 and assayed by IFN-γ-ELISPOT-Assay<br />

(using peptide-pulsed T2 cells as target cells) demonstrated<br />

the induction of MART-1-specific T cell<br />

responses. Pre-clinical validation of SMART-DCs<br />

engineered with tricistronic vectors for MART-1<br />

or TRP2 expression, for the assessment of their<br />

potency to stimulate and expand autologous T cells<br />

obtained from melanoma patients is underway. A<br />

pre-GMP lentiviral production for generation of<br />

GMP compliant vector will follow, for development<br />

of a phase I immunotherapy clinical trial for melanoma<br />

patients.<br />

47


012 Gueckel | Therapeutic vaccination<br />

Targeted modification of HLA-A*02-restricted mucin-1-derived<br />

peptides results in induction of cytotoxic T cells cross-reactive<br />

with naturally presented peptides<br />

Brigitte Gückel 1 , Simone Kayser 1 , Helen Hörzer 1 , Janet Peper 1 , Despina Rudolf 2 , Dorothee<br />

Wernet 3 , Stefan Stevanovic 2 , Dagmar Sigurdardottir 2<br />

1 Department of Obstetrics and Gynecology, University of Tuebingen, 72076 Tuebingen, Germany<br />

2 Department of Immunology, Institute for Cell Biology, University of Tuebingen, 72076 Tuebingen, Germany<br />

3 Institute of Clinical and Experimental Transfusion Medicine, University Hospital Tuebingen, 72076 Tuebingen,<br />

Germany<br />

48<br />

The characterization of T-cell epitopes derived<br />

from tumor associated antigens fostered peptidebased<br />

cancer vaccines. Well characterized mucin<br />

(MUC)-1 derived HLA-A*02-restricted epitopes<br />

have been used for vaccinations in renal cell<br />

cancer, breast and ovarian cancer; however, the clinical<br />

efficacy had not yet met expectations. Altered<br />

peptide ligands (APLs) that might have enhanced<br />

HLA-binding and thus, improved immunogenicity<br />

are under investigation.<br />

In this study, we tested the CD8+ T-cell responses<br />

against two MUC-1 epitopes. The (TαL) substitution<br />

at the anchor position 2 in peptide M1.1 (MUC-1950-<br />

958) induced a 10-fold increase in peptide-MHC<br />

binding, as shown by T2-binding assay. Replacement<br />

of (LαK) at position 1 in the M1.2 peptide<br />

(MUC-112-20) increased its solubility in aqueous<br />

solution while not affecting its MHC-binding.<br />

To compare the CD8+ T-cell response against<br />

MUC-1 peptides and their APLs, peripheral blood<br />

lymphocytes of healthy donors were stimulated<br />

with peptide-pulsed antigen-presenting cells for<br />

several rounds. The frequency of peptide-reactive<br />

CD8+ T cells was determined by intracellular IFNγ-staining.<br />

While T cells specific for the natural<br />

peptides and the M1.2-APL were rarely detected,<br />

many donors produced significant frequencies of<br />

CD8+ T cells reactive with the M1.1-APL. M1.2-<br />

APL-specific CD8+ T cells were simultaneously<br />

recognized by HLA-A*02 tetramers containing the<br />

natural and the modified peptide, which suggests<br />

that the modification in the anchor peptide did not<br />

significantly affect T-cell recognition. Similarly,<br />

M1.1-APL-specific T cells efficiently lysed T2 cells<br />

loaded with either the natural or the modified<br />

peptide. Interestingly, a similar cross-reactivity was<br />

seen with rare M1.2-specific T cells. More importantly,<br />

M1.1-APL-specific T cells lysed the MUC-1<br />

highly expressing tumour cell line MCF-7 which<br />

presents the natural M1.1 epitope.<br />

In summary, CD8+ T cells induced against both<br />

MUC-1-APLs are able to cross-react with naturally<br />

presented parental epitopes and, in the case of<br />

M1.1-APL, the ease of induction of peptide-specific<br />

T cells makes this peptide a good candidate for vaccination<br />

strategies.


013 Babiarova | Therapeutic vaccination<br />

Development of different types of vaccine against WT1<br />

tumor antigen<br />

Katarina Babiarova, Luda Kutinova, Eva Brabcova, Jitka Krystofova, Petr Hainz and Sarka<br />

Nemeckova<br />

Department of Experimental Virology, Institute of Hematology and Blood Transfusion, Prague 2, U Nemocnice 1,<br />

CZ-128 20, Czech Republic<br />

The Wilms’ tumor gene 1 (WT1) encodes a tran-<br />

scription factor, initially identified as a suppressor<br />

in the etiology of Wilms’ tumor. It is expressed at<br />

high levels in a variety of human cancer including<br />

leukemia and various types of solid tumors. WT1<br />

protein has been reported to serve as target antigen<br />

for tumor-specific immune responses.<br />

The aims of this study were to prepare experimental<br />

vaccines of different type against WT1 tumor<br />

antigen and to evaluate their potency in immunotherapy<br />

of tumors in mouse model. We prepared<br />

DNA vaccine pBSC/WT1-GUS based on the fusion<br />

gene β-glucoronidase from E.coli (GUS) which<br />

was shown previously to have superior anti-tumor<br />

effect. We also constructed the viral vaccine P13-<br />

WT1-GUS based on the recombinant vaccinia virus<br />

(rVACV) strain P13 with the expression of fusion<br />

protein WT1-GUS under the control of either earlylate<br />

(H5) or synthetic early-late (E/L) VACV promoters.<br />

Every vaccine contains a fragment of murine<br />

WT1 gene coding for a peptide with the length of<br />

155aa (94.-249.aa) containing several CTL epitopes.<br />

We also used peptide vaccines derived from WT1<br />

with motifs predicted to bind to Db murine MHC<br />

I. The efficacy of these vaccines was tested in the<br />

immunization experiments in mice. The ELISPOT-<br />

IFNγ assay and measurement of the intracellular<br />

IFNγ was used to detect T cell immune response<br />

specific for WT1 peptides. Highest response was<br />

elicited by the RMFPNAPYL peptide. The immunization<br />

of mice either with pBSC/WT1-GUS or P13-<br />

WT1-GUS in combination with WT1 peptide vaccine<br />

P(126-135) as a booster revealed a low WT1 peptide<br />

specific T cell immune response in about 50% of<br />

immunized mice. Similar results were found after<br />

the immunization of mice using combination of all<br />

three types of vaccines. Anti-tumor effect of immunization<br />

with the different combination of our<br />

three vaccines were determined in mouse model of<br />

WT-1 positive tumors induced by the TRAMP-C2<br />

cells. We observed that priming with rVACV based<br />

vaccine combined with booster with RMFPNAPYL<br />

peptide in incomplete Freund adjuvant resulted in<br />

enhancement of the growth of TRAMP-C2 cells in<br />

mice. However immunization with one vaccine<br />

only (P13-WT1-GUS) was able to decelerate the<br />

growth of TRAMP-C2 tumors.<br />

This work was supported by grants NS 10660-3/2009 from IGA<br />

MZ ?R and 78608 from GAUK.<br />

49


014 Abbas | Therapeutic vaccination<br />

Vaccine development through antigen targeting to mannose<br />

receptor<br />

Zaigham Abbas 1 , Richard Pleass 2 , Giuseppe Mantovani 3 , Lindy Durrant 1 and Luisa Martinez-Pomares 1<br />

1 School of Molecular Medical Sciences, University of Nottingham<br />

2 School of Biology, University of Nottingham<br />

3 School of Pharmacy, University of Nottingham<br />

50<br />

Dendritic cells (DC) are unique antigen presenting<br />

cells which play a major role on the antigen presentation<br />

and initiation of the immune response<br />

by regulating B- and T- cell activation. Antigen<br />

targeting to DC receptors is an effective, safe and<br />

specific method for the vaccine development. MR<br />

is an endocytic receptor expressed by subpopulations<br />

of DC. Antigen targeting through MR leads<br />

to enhanced antigen uptake and presentation to T<br />

cells which makes it a favourite receptor for the development<br />

of vaccines against diseases that require<br />

T-cell immunity such cancer and viral infections.<br />

We have used two approaches to target antigen to<br />

MR; (i) MR-specific chimeric antibodies carrying<br />

several model antigens and (ii) antigens conjugated<br />

to novel glycopolymers. The binding efficiency of<br />

the chimeric antibodies has been assessed by using<br />

ELISA and BIACORE and the glycopolymers have<br />

been tested for their interaction with MR. These<br />

glycopolymers will be coupled to a mutated form<br />

of the model antigen ovalbumin (OVA) lacking Nglycosylation<br />

sites as native OVA is an excellent MR<br />

ligand. These novel reagents are currently being<br />

tested for their ability to induce T-cell activation<br />

in vitro using co-cultures of antigen presenting<br />

cells and T cells and in-vivo. Our results will<br />

greatly benefit the development of antigen delivery<br />

methods suitable for robust T cell activation.


015 Vogt | Therapeutic vaccination<br />

Inhibition of tumor growth after subcutaneous injection of dendritic<br />

cells engineered to express angiogenesis-related flk-1 antigen<br />

in a murine HCC model<br />

Annabelle Vogt 1 , Georges Decker 1 , Esther Raskopf 1 , Volker Schmitz 1 , Tilman Sauerbruch 1 ,<br />

Wolfgang H. Caselmann 2 and Maria A. González-Carmona 1<br />

1 Department of Internal Medicine I, University of Bonn, Bonn, Germany<br />

2 Bavarian State Ministry of the Environment and Public Health, Munich, Germany<br />

Background: Dendritic cells (DC) are professio-<br />

nal antigen presenting cells able to prime T-cells<br />

against tumor associated antigens (TAA) such<br />

as AFP. However, tumor cells downregulate the<br />

expression of TAA inducing immune tolerance.<br />

VEGFR-2 (flk-1) is a receptor expressed by proliferative<br />

endothelial cells, which are mainly involved<br />

in the tumorangiogenesis. Its ligand VEGF has been<br />

shown to stimulate endothelial cell mitogenesis and<br />

cell migration. The aim of this study was to induce<br />

a more effective immune response by targeting the<br />

angiogenesis-associated antigen, flk-1 in vivo. Therefore,<br />

DC were engineered to express soluble flk-1<br />

using an adenoviral vector (Ad) encoding for flk-1.<br />

Methods: Ad-flk1 encoding for murine flk-1 was<br />

constructed using the AdEasy system. As control,<br />

Ad-LacZ was used. DC were obtained from the bone<br />

marrow of C3H-mice and adenovirally transduced<br />

on day 6. To study the effect of the vaccination with<br />

flk-1-expressing DC in the angiogenesis, C3H-mice<br />

were immunized by subcutaneous (s.c.) injection<br />

of 106 flk- or LacZ-expressing DC. One week later,<br />

500µl of Matrigel enriched with VEGF was injected<br />

s.c. in the abdomen. Density of newly formed vascular<br />

vessels was evaluated by CD31-staining. To<br />

evaluate antitumoral effects, 106 Hepa129-cells<br />

were injected s.c. into the right flank of C3H-mice<br />

to induce a tumor. Mice were treated twice with 106<br />

flk1- or AdLacZ-transduced DC.<br />

Results: A significant inhibition regarding the formation<br />

of vascular vessels could be assessed one<br />

week after the vaccination with flk-1 expressing<br />

DC (3,5+/-1,6 vascular vessels in the group of mice<br />

treated with flk-1 DC vs. 31,75+/-8 in the group<br />

treated with LacZ-DC, p


016 van Nuffel | Therapeutic vaccination<br />

T cell specificities after vaccination of melanoma patients with<br />

mRNA loaded DC<br />

An MT Van Nuffel 1 , Daphné Benteyn 1 , Sofie Wilgenhof 1,2 , Jurgen Corthals 1 , Carlo Heirman 1 , Bart Neyns 2 ,<br />

Kris Thielemans 1 and Aude Bonehill 1<br />

1 Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium<br />

2 Department of Medical Oncology, Universitair Ziekenhuis Brussel, Brussels, Belgium<br />

52<br />

The incidence of melanoma doubles every 10 years.<br />

The use of full-length tumor associated antigens<br />

(TAAs) to load dendritic cells (DCs) is proposed<br />

to improve DC based immunotherapy because<br />

this should allow vaccination irrespective of the<br />

patient’s HLA-type and stimulation of a broad spectrum<br />

of TAA-recognizing T cells.<br />

Our objective was to investigate the in vivo stimulatory<br />

potency of DCs loaded by electroporation with<br />

defined, full-length TAA-encoding mRNA. Defined<br />

mRNA was chosen as antigen carrier because it can<br />

be modified to enhance the antigen presentation by<br />

DCs to both CD8+ and CD4+ T cells. The exact T<br />

cell specificities of vaccine stimulated T cells were<br />

analyzed.<br />

Histologically confirmed AJCC stage IV melanoma<br />

patients included in a phase I clinical study<br />

donated a skin biopsy one week after the last of<br />

four biweekly DC-vaccines. Vaccination occurred<br />

with an equal mix of TriMix-DC (i.e. DCs matured<br />

using constitutive active TLR4, CD40L and CD70<br />

mRNA) co-electroporated with mRNA encoding<br />

gp100, tyrosinase, Mage-C2 or Mage-A3 fused to<br />

the HLA-class II targeting sequence of DC-Lamp.<br />

Skin biopsies were taken 72h after induction of the<br />

delayed type hypersensitivity (DTH) response by<br />

intradermal injection of a small amount of vaccine<br />

DCs. DTH-infiltrating lymphocytes (DIL) were cultured<br />

during 2,5 weeks in the presence of 100 IU/<br />

ml IL-2. The DIL were re-stimulated overnight by<br />

autologous EBV-B cells electroporated with TAA<br />

mRNA. Activated T cells were detected by both<br />

CD137 upregulation in flow cytometry and cytokine<br />

secretion (IFN-γ and TNF-α) using a cytokine<br />

bead assay. Once T cells specific for a vaccine<br />

antigen were found, the recognized epitope was determined<br />

using autologous EBV-B cells loaded with<br />

overlapping peptides covering the TAAs. The HLA<br />

restriction was assessed by allogeneic EBV-B cells<br />

expressing relevant HLA-types and vaccine TAA.<br />

This way, we found CD8+ and CD4+ T cells<br />

specific for the previously unidentified epitopes<br />

RTCQCSGNF and YPPLHEWVLREG, derived<br />

from the tumor antigens tyrosinase and Mage-A3,<br />

respectively. These epitopes were presented respectively<br />

in the less common HLA-B57 and the<br />

common HLA-DQB1*05. In vivo stimulation of the<br />

CD8+ T cells by the TriMix-DCs was confirmed by<br />

the analysis of blood samples obtained pre- and<br />

postvaccination.<br />

In conclusion, vaccination of melanoma patients<br />

with mRNA loaded DCs leads to in vivo stimulation<br />

of CD8+ and CD4+ T cells recognizing previously<br />

unknown epitopes presented in both common and<br />

less common HLA-types. This finding underscores<br />

the broadness of the induced T cell response and<br />

thereby the strength of using mRNA loaded DCs.<br />

On the other hand, it shows that DC-therapy is no<br />

longer solely suitable for a selected population with<br />

a certain HLA-type but can be beneficial for all patients.


017 Schaft | Therapeutic vaccination<br />

Optimization of the T-cell stimulation capacity of a dendriticcell-based<br />

melanoma vaccine<br />

Niels Schaft 1 ,*, Jan Dörrie 1 ,*, Tanja Schunder 1 , Christian Wohn 1 , Verena Wellner 1 , Stefanie<br />

Baumann 1 and Gerold Schuler 1<br />

1 Department of Dermatology, University Hospital Erlangen, Erlangen, Germany<br />

* Contributed equally<br />

Tumor-antigen-loaded dendritic cells (DC) are pro-<br />

mising tools as therapeutic vaccine for the treat-<br />

ment of malignant melanoma. Although immune<br />

responses, such as induction of tumor-antigen-specific<br />

T cells, were observed in many studies with<br />

DC vaccines, these did not correlate with clinical<br />

responses. Electroporation of monocyte-derived<br />

DC with RNA, to load them with antigen, or to manipulate<br />

their function, has become a widely used<br />

method, also in clinical applications. Our aim in<br />

this study was to optimize the T-cell stimulation capacity<br />

of DC. Therefore, we electroporated an optimized<br />

CD40L-encoding RNA (optCD40L) alone, or<br />

a combination of (non-optimized) CD40L-, CD70-,<br />

and constitutively active TLR4 (caTLR4)-encoding<br />

RNAs (TriMix, as described by K. Thielemans et<br />

al.) into mature or immature DC, respectively. In<br />

addition, these DC were electroporated with a RNA<br />

encoding a tumor antigen (Ag). We chose MelanA<br />

as a model-Ag, for its well-characterized and highly<br />

immunogenic HLA-A2-presented peptide EAAGI-<br />

GILTV. The optCD40L transfection in mature DC<br />

resulted in enhanced expression of the maturation<br />

markers CD25, CD40, CCR7, CD70, and OX40L,<br />

and in secretion of the pro-inflammatory cytokines<br />

IL-12p70, IL-6, IL-8, and TNF-alpha. Simultaneously,<br />

the DC retained their capacity to migrate in a<br />

CCR7-dependent way. The DC‘s capacity to expand<br />

autologous T cells was analyzed by in vitro stimulation<br />

and subsequent HLA-A2-MelanA tetramerstaining<br />

in combination with phenotyping (CCR7/<br />

CD45RA) for T-cell function. The capability of the<br />

DC to induce expansion of T cells in a second and<br />

third round of stimulation was improved by the<br />

transfection of optCD40L RNA. The TriMix transfection<br />

in immature DC resulted in maturation of<br />

these DC, as shown by up-regulation of maturation<br />

markers, and a secretion of IL-12p70. However, the<br />

T-cell expansion capacity was less efficient compared<br />

to cytokine-cocktail-matured, MelanA-RNAelectroporated<br />

DC.<br />

We believe that these studies point the way to improved<br />

DC that will induce better and longer lasting<br />

immune responses in the vaccination against<br />

cancer.<br />

53


018 Haenssle | Therapeutic vaccination<br />

Dendritic cells loaded with HSP70-expressing heat-killed melanoma<br />

cells efficiently activate autologous T cells<br />

Holger A. Haenssle 1 , Susanne Knudsen 1 , Anke Schardt 2 , Timo Buhl 1 , Lars Boeckmann 1 ,<br />

Michael P. Schön 1<br />

1 Department of Dermatology and Venereology, Georg August University, Göttingen, Germany<br />

2 Max Planck Institute for Experimental Medicine, Göttingen, Germany<br />

54<br />

There is considerable interest to develop strategies<br />

that enhance cross-presentation pathways of dendritic<br />

cells (DCs) to elicit a strong cytotoxic T cell<br />

activation for cancer vaccination protocols.<br />

In order to best exploit the enhanced cross-presentation<br />

of antigens bound to heat shock protein<br />

70 (HSP70), we analyzed melanoma cell preparations<br />

for their HSP70 expression. For generation of<br />

heat-necrotic cell material melanoma cells were<br />

kept at 42°C for 90 min and then heated to 56°C<br />

for a final 30 min. Apoptotic Annexin V /Propidium<br />

iodide- melanoma cells were generated by<br />

irradiation with 1.0 J/cm2 UV-B. Western blotting<br />

revealed strong upregulation of HSP70 after heatkilling<br />

in contrast to UV-B irradiation. The uptake<br />

of fluorescently labeled heat-killed necrotic vs.<br />

apoptotic melanoma cells by DCs at various levels<br />

of maturation was assessed. Immature DCs (iDCs)<br />

internalized HSP70-expressing necrotic material to<br />

a much higher extent (61% ± 7%) than apoptotic<br />

material from UV-B-irradiated cells (48% ± 5%).<br />

Maturation-inducing cytokines did not affect the<br />

uptake when added simultaneously with the tumor<br />

cell preparations. Loading DCs with heat-necrotic<br />

or apoptotic melanoma cells slightly reduced CD83<br />

expression while leaving CD208 (DC-LAMP) expression<br />

unchanged. As determined by IFN- -detecting<br />

ELISPOT assays, iDCs loaded with HSP70-expressing<br />

heat-killed melanoma cells activated autologous<br />

T cells most effectively when used without<br />

any further maturation, whereas DCs loaded with<br />

apoptotic material required maturation. In conclusion,<br />

HSP70-expressing melanoma cells could be<br />

generated by heat-killing. Loading immature DCs<br />

with heat-killed melanoma cells resulted in a superior<br />

priming of autologous T cells in vitro.


019 Buhl | Therapeutic vaccination<br />

Cryopreservation of high-concentrated peripheral blood mononuclear<br />

cells by controlled-rate freezer results in higher cell<br />

yields for dendritic cell-based immunotherapy<br />

Timo Buhl 1 , Anke Schardt 1 , Tobias Legler 2 , Michael P. Schön 1 , Holger A. Hänßle 1<br />

1 Department of Dermatology and<br />

2 Department of Transfusion Medicine of Georg August University, Göttingen, Germany<br />

Cryopreservation of immature or mature dendritic<br />

cells (DC) has been described as a suitable method<br />

to achieve large numbers of DC for immunotherapeutic<br />

trials against cancer. Recently it was shown<br />

that cryopreservation of peripheral blood mononuclear<br />

cells (PBMC) with subsequent differentiation<br />

into DC is superior compared to cryopreservation<br />

of immature or mature DC in terms of resulting DC<br />

quantity and immuno-stimulating capacity. The<br />

aim of our study was to establish an optimized<br />

protocol for the cryopreservation of highly-concentrated<br />

PBMC for DC-based immunotherapy. Cryopreserved<br />

cell preparations were analyzed regarding<br />

recovery, viability, phenotype, and functional<br />

properties. Results were compared to fresh DC<br />

generated instantly from the same donor. In contrast<br />

to PBMC frozen by standard isopropyl alcohol<br />

freezing containers, PBMC cryopreservation in an<br />

automated controlled-rate freezer (CRF) resulted<br />

in significantly higher cell yields of immature and<br />

mature DC after thawing and subsequent differentiation<br />

to DC. The CRF is connected to liquid nitrogen,<br />

allowing the freezing program to exactly<br />

control and adjust the temperature of the freezing<br />

chamber. Therefore, the crystallization heat at the<br />

freezing point of the sample can be compensated<br />

by a sharp temperature decrease in the freezing<br />

chamber. Immature DC yields and total protein<br />

content after using CRF were comparable to freshly<br />

generated DC and exceeded results of standard isopropyl<br />

alcohol freezing by approximately 50%. DC<br />

generated after both freezing protocols of PBMC revealed<br />

no relevant phenotypic or functional differences.<br />

Our analyses included phenotypic markers,<br />

allogenic T cell stimulatory assays, viability tests<br />

and microarray cytokine profiles. The latter showed<br />

that few cytokines were differently secreted during<br />

DC maturation depending on the method of cryopreservation.<br />

The majority of cytokine levels was<br />

not altered in correlation with the method of cryopreservation.<br />

In summary, automated controlled<br />

rate freezing of PBMC represents a much improved<br />

freezing method capable of increasing DC yields for<br />

cancer immunotherapy.<br />

55


020 Nielsen | Therapeutic vaccination<br />

The CDX-1307 vaccine: regulatory and clinical steps in<br />

developing a regimen<br />

Michaela Nielsen, Larry Thomas, Laura Vitale, Li-Zhen He, Venky Ramakrishna, Jennifer<br />

Green, Thomas Davis<br />

Celldex Therapeutics, Inc., Needham, Massachusetts 02494, USA<br />

56<br />

CDX-1307 is a dendritic cell (DC)-targeting cancer<br />

vaccine, consisting of a monoclonal antibody<br />

against the mannose receptor, CD206, fused to the<br />

β-chain of human chorionic gonadotropin (hCGβ).<br />

hCGβ is frequently expressed by common epithelial<br />

cancers, may facilitate cancer progression,<br />

and correlates with poor prognosis. The antibody<br />

portion of the vaccine targets it to CD206 on DCs,<br />

where the molecule is internalized and processed<br />

into peptides that are presented on the DC surface.<br />

The CDX-1307 vaccine regimen was developed in<br />

a step-wise approach. Without a suitable animal<br />

model available, the initial Phase I study was a<br />

dose-escalation with intradermal administration of<br />

CDX-1307 in patients with incurable cancer. Safety<br />

of the vaccine was established by demonstrating<br />

tolerability in cohorts of 6 patients at 0.3, 1.0, and<br />

2.5 mg respectively, given every two weeks for a<br />

total of 4 doses.<br />

Pre-clinical data and clinical experience demonstrated<br />

that inclusion of adjuvants leads to optimal<br />

immune responses. The cytokine GM-CSF, which<br />

mobilizes and activates antigen-presenting cells<br />

(APCs) and up-regulates CD206 on APCs, was thus<br />

proposed to be added to the study regimen. After a<br />

toxicology study with CDX-1307 + GM-CSF in hMRtransgenic<br />

mice showed no noteworthy adverse reactions,<br />

combination treatment with CDX-1307 and<br />

GM-CSF was approved and initiated.<br />

Further studies indicated that immune responses<br />

are significantly enhanced by toll-like receptor<br />

(TLR) agonists. Two such agents were proposed<br />

to be added to the CDX-1307 + GM-CSF regimen<br />

in additional cohorts: the TLR7/8 antagonist Resiquimod<br />

(3M) or the TLR3 antagonist Hiltonol®<br />

(Poly-ICLC). Both cohorts were approved based on<br />

previous human experience with the antagonists,<br />

which were proposed to be used in a concentration<br />

below their established maximum tolerated doses<br />

(MTD).<br />

Our pre-clinical studies showed that combining<br />

TLR agonists enhances the immune response and<br />

anti-tumor activity of cancer vaccines. Therefore,<br />

a regimen that combines GM-CSF, Resiquimod and<br />

Hiltonol® (Poly-ICLC) was proposed. This regimen<br />

was approved after review of toxicology data from<br />

a rabbit local reactogenicity study with various<br />

combinations of the three adjuvants. One particular<br />

challenge of the combination regimen is the<br />

local administration of four different agents, one<br />

of which is topically administered (Resiquimod),<br />

while the others are injected. Most rabbits that received<br />

the three adjuvant combination developed<br />

minor skin reactogenicity, with only one animal<br />

developing some slowly resolving subcutaneous<br />

edema.<br />

The clinical data demonstrated that the combination<br />

of the TLR agonists increased the frequency and<br />

severity of local reactions, but they were limited to<br />

grade 1 and 2 and resolved. In fact no dose limiting<br />

toxicities were reported, while robust anti-hCG-β<br />

humoral and T cell responses were reported. Evidence<br />

of clinical impact was observed in a subset of<br />

patients, and was noted to be associated with generation<br />

of anti-hCG-β responses. These data provide<br />

support for further evaluation of the CDX-1307<br />

vaccine regimen in patients with hCG-β-expressing<br />

tumors. A Phase II study in patients with newly diagnosed,<br />

resectable hCGβ-positive bladder cancer<br />

involving the full combination regimen has been<br />

accepted by the FDA and is pending initiation.


L119 Dutoit | Therapeutic vaccination<br />

Functional immune reactivity to antigens of the novel<br />

IMA950 vaccine peptide set in glioma patients<br />

Valérie Dutoit 1 , Norbert Hilf 2 , Steffen Walter 2 , Toni Weinschenk 2 , Philippe Beckhove 3 , Christel<br />

Herold-Mende 4 , Paul Walker 1 , Harpreet Singh 2 and Pierre-Yves Dietrich 1<br />

1 Laboratory of tumor immunology, Oncology department, Geneva University Hospital, Geneva, Switzerland<br />

2 immatics biotechnologies GmbH, Tuebingen, Germany<br />

3 Translational Immunology Unit, The German Cancer Research Center, Heidelberg, Germany<br />

4 Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg, Germany<br />

We have recently identified a new set of HLA-A2restricted<br />

glioma antigens isolated by direct elution<br />

from tumor cells for use in glioma immunotherapy.<br />

These antigens were selected from a large panel<br />

of eluted peptides according to overexpression in<br />

glioma compared to normal brain and other tissues,<br />

and relevance in tumorigenesis. In vitro peptide<br />

immunogenicity was investigated in healthy individuals,<br />

guiding the final vaccine peptide composition.<br />

Here, in order to validate the use of the novel peptide<br />

set for multipeptide vaccination, T cell reactivity<br />

and absence of tolerance was investigated in patients<br />

with malignant glioma. In addition, peptidespecific<br />

T cell function was assessed in vitro. All<br />

patients responded to several glioma peptides, suggesting<br />

that this multipeptide vaccine may benefit<br />

the majority of HLA-A2+ patients. Moreover,<br />

glioma peptide-specific CD8+ T cell clones generated<br />

from the blood of glioma patients were able to<br />

efficiently kill antigen-expressing tumor cells, but<br />

not K562 cells. Finally, CD8+ T cells specific for a<br />

brevican epitope were detected at the tumor site in<br />

one patient, showing that, for this antigen, spontaneously<br />

elicited specific T cells are retained at the<br />

tumor site. Altogether, our results validate the use<br />

of the ten novel glioma antigens in multipeptide<br />

vaccination and adoptive cell therapies for patients<br />

with glioma.<br />

57


L120 Hilf | Therapeutic vaccination<br />

IMA950 - a novel multi-peptide cancer vaccine for treatment of<br />

glioblastoma<br />

Norbert Hilf 1 , Oliver Schoor 1 , Valérie Dutoit 2 , Toni Weinschenk 1 , Steffen Walter 1 , Peter Lewandrowski<br />

1 , Sylvia Flohr 1 , Claudia Trautwein 1 , Cécile Gouttefangeas 3 , Stefan Stevanovic 3 ,<br />

Hans-Georg Rammensee 3 , Philipp Beckhove 4 , Christel Herold-Mende 5 , Pierre-Yves Dietrich 2 ,<br />

Harpreet Singh 1<br />

1<br />

immatics biotechnologies GmbH, Tuebingen, Germany<br />

2<br />

Laboratory of Tumor Immunology, Oncology, Geneva University Hospital, Geneva, Switzerland immatics bio<br />

technologies GmbH, Tuebingen, Germany<br />

3<br />

Department of Immunology, Institute for Cell Biology, University of Tuebingen, Germany<br />

4 Translational Immunology Group, German Cancer Research Center, Heidelberg, Germany<br />

5 Division of Neurosurgical Research, Department of Neurosurgery, University of Heidelberg, Germany<br />

58<br />

Despite promising results of various therapeutic<br />

approaches currently under investigation, including<br />

active immunotherapy, glioblastoma (GBM)<br />

remains one of the most fatal tumor types. IMA950<br />

is a novel peptide vaccine aiming at the induction<br />

of a broad T-cell response against a variety of<br />

tumor-relevant antigens expressed by GBM cells.<br />

IMA950 consists of 11 synthetic tumor-associated<br />

peptides (TUMAPs). Nine of the TUMAPs bind to<br />

the human leukocyte antigen (HLA) class I allele<br />

A*02 expressed by ~ 45% of the Caucasian population<br />

and two TUMAPs bind to various HLA class<br />

II (DR) alleles expressed by the majority of patients.<br />

Therefore, cytotoxic T-lymphocyte (CTL) as well as<br />

T helper responses are expected to arise from vaccination<br />

with IMA950.<br />

IMA950 was rationally designed employing the<br />

XPRESIDENT approach: More than 3,000 different<br />

HLA-A*02 peptides were identified from more than<br />

30 primary GBM specimens by mass spectrometry.<br />

TUMAPs were ranked according to a variety of<br />

criteria, including degree and frequency of mRNA<br />

over-expression of the source antigens in GBM<br />

compared with normal tissues, known relevance of<br />

the source antigens in oncogenic processes, in vitro<br />

immunogenicity in healthy donors, and feasibility<br />

of pharmaceutical development.<br />

Analysis of the „over-presentation“ on GBM samples<br />

directly at the peptide level confirmed the relevance<br />

of the finally selected TUMAPs using a novel<br />

approach allowing label-free HLA-peptide quanti-<br />

tation directly by mass spectrometry.<br />

In conclusion, the strong GBM association of IMA950<br />

TUMAPs indicated by overpresentation together<br />

with pre-clinical immunogenicity results suggest<br />

that this multipeptide vaccine may be a benefit to<br />

the majority of HLA-A*02-positive patients. Clinical<br />

development is planned to start within <strong>2010</strong> with<br />

several phase I trials investigating the safety and<br />

immunogenicity of the vaccine in different patient<br />

populations and using various immunomodulatory<br />

regimens in order to explore the most promising<br />

strategy for further development.


L121 Pawlowski | Therapeutic vaccination<br />

Synergy of the immunomodulators GM-CSF and Imiquimod for<br />

peptide-based therapeutic vaccines<br />

Nina Pawlowski, Norbert Hilf, Sylvia Flohr, Toni Weinschenk, Oliver Schoor, Harpreet Singh<br />

immatics biotechnologies GmbH, Tuebingen, Germany<br />

All FDA- or EMA-approved drugs currently used<br />

as immunomodulators with therapeutic peptidebased<br />

cancer vaccines are of only moderate efficacy<br />

in terms of direct activation of antigen-presenting<br />

cells which is a key prerequisite for efficient priming<br />

of cytotoxic T cells (CTLs). Because the reliable<br />

induction of tumor-directed immune responses is<br />

crucial for the efficacy of therapeutic cancer vaccines,<br />

novel immunomodulator regimens have to be<br />

introduced into clinical testing to achieve a better<br />

clinical outcome. Therefore, we analyzed safety<br />

and efficacy of several approved immunomodulatory<br />

substances and combinations thereof in a dedicated<br />

pre-clinical screening program.<br />

The combination of subcutaneously (s.c.) applied<br />

granulocyte-macrophage colonystimulating factor<br />

(GM-CSF) with topical application of the TLR7<br />

ligand imiquimod resulted in synergistic enhancement<br />

of peptide-vaccine induced CTL responses<br />

without any signs of toxicity or cumulated adverse<br />

effects.<br />

GM-CSF injected s.c. combined with imiquimod<br />

cream applied topically to the skin of C57BL/6<br />

mice at the vaccination site significantly increased<br />

the number of vaccinespecific CTLs induced by<br />

s.c. vaccinations with model MHC class I-binding<br />

peptides compared with either substance alone.<br />

The induced immune response was comparable to<br />

the effect of CpG deoxyoligonucleotides or poly-IC,<br />

two potent immunomodulators currently not FDA-/<br />

EMA-approved and therefore not widely available<br />

for clinical trials in humans. However, the timing<br />

of imiquimod application in relation to the vaccination<br />

schedule is crucial, as the synergistic effect<br />

of imiquimod and GM-CSF was only observed, if<br />

imiquimod was applied at the day of vaccination<br />

and/or 24 h later, but not if imiquimod was applied<br />

the day before peptide vaccination.<br />

In total, 45 mice were immunized with the combination<br />

of GM-CSF and imiquimod without any<br />

signs of local or systemic toxicities. Further results<br />

from in vitro activation assays with human PBMCs<br />

and isolated antigen-presenting cells and the distinct<br />

molecular mechanisms of immunostimulation<br />

employed by both substances provide further<br />

support for the anticipated safety of this regimen<br />

in humans.<br />

GM-CSF and imiquimod is therefore considered as<br />

a promising and safe alternative immunomodulator<br />

regimen for upcoming immunotherapeutic approaches<br />

and is currently explored by the authors<br />

in a clinical trial in colorectal cancer patients.<br />

59


L122 van de Ven | Therapeutic vaccination<br />

Exposure of CD34+ precursors to cytostatic anthraquinone-derivatives<br />

induces rapid dendritic cell differentiation<br />

Rieneke van de Ven 1,2 , Anneke Reurs 1,3 , Pepijn Wijnands 1,3 , Sandra van Wetering 3 , Ada<br />

Kruisbeek 3 , Erik Hooijberg 1 , George Scheffer 1 , Rik Scheper 1,3 and Tanja de Gruijl 2<br />

1<br />

Department of Pathology, VU University medical center, Amsterdam, The Netherlands<br />

2<br />

Medical Oncology Laboratory, VU University medical center, Amsterdam, The Netherlands<br />

3 DC Prime B.V., Amsterdam, The Netherlands<br />

60<br />

Appropriate activation of Dendritic Cells (DC) is<br />

essential for successful active vaccination and induction<br />

of cell-mediated immunity. The scarcity of<br />

precursor cells, as well as long culture methods,<br />

has hampered wide-scale application of DC vaccines<br />

derived from CD34+ precursors, despite their<br />

suggested superior efficacy over the more commonly<br />

applied monocyte-derived DC (MoDC). Here, employing<br />

the CD34+/CD14+ Acute Myeloid Leukemia-derived<br />

human DC progenitor cell line MUTZ3,<br />

we show that cytostatic anthraquinone-derivatives<br />

(i.e. the anthracenedione mitoxantrone and the<br />

related anthracyclin doxorubicin) induce rapid differentiation<br />

of CD34+ DC precursors into functional<br />

antigen presenting cells (APC) in a three-day<br />

protocol. The drugs were found to act specifically<br />

on CD34+, and not on CD14+ DC precursors. Importantly,<br />

these observations were confirmed for<br />

primary CD34+ and CD14+ DC precursors from<br />

peripheral blood. Mitoxantrone-generated DC were<br />

fully differentiated within three days and after an<br />

additional twenty-four hours of maturation, were<br />

as capable as control 9-day differentiated and<br />

matured DC to migrate towards the lymph nodehoming<br />

chemokines CCL19 and CCL21, to induce<br />

primary allogeneic T cell proliferation, and to prime<br />

functional MART1-specific CD8+ T lymphocytes.<br />

Anthraquinone-derivatives like mitoxantrone may<br />

thus be employed either as a differentiation-inducing<br />

agent for rapid in vitro generation of DC, or<br />

might even be exploited to mature DC precursors<br />

in vivo in support of DC-based therapies.


Immune monitoring<br />

61


021 Santegoets | Immune monitoring<br />

Immune and clinical response monitoring in patients with metastatic<br />

hormone-refractory prostate cancer receiving combined<br />

Prostate GVAX and anti-CTLA4 immunotherapy<br />

Saskia J.A.M. Santegoets 1 , A.J.M. van den Eertwegh 1 , A.G.M. Stam 2 , R.J. Scheper 2 , S.M.<br />

Lougheed 1 , P.E.T. Scholten 2 , B.M.E. von Blomberg 2 , H. Gall 1 , K. Jooss 3 , N. Sacks 3 , T. Harding<br />

3 , K. Hege 3 , I. Lowy 4 , W.R. Gerritsen 1 , and T.D. de Gruijl 1<br />

1 Department of Medical Oncology, VU University medical center, Amsterdam, The Netherlands<br />

2 Department of Pathology, VU University medical center, Amsterdam, The Netherlands<br />

3 Cell Genesys Inc., South San Francisco, CA, USA<br />

4 Medarex In.c, Bloomsbury, NJ, USA<br />

62<br />

The effects of a GM-CSF-secreting allogeneic pro-<br />

state cancer vaccine (Prostate GVAX) and the anti-<br />

CTLA4 antibody Ipilimumab were investigated in<br />

a Phase I dose escalation/expansion trial of patients<br />

with metastatic, hormone-refractory prostate<br />

cancer (mHRPC). Patients received a 500 million<br />

cell GVAX priming dose on day 1 followed by 12 biweekly<br />

intradermal administrations of 300 million<br />

cells, while Ipilimumab was administered every 4<br />

wks from day 1 for a total of 6 times. Initially, 12 patients<br />

were enrolled in cohorts of 3 and each cohort<br />

was assigned an escalating dose of Ipilimumab at<br />

0.3, 1, 3 or 5 mg/kg. 16 additional patients were<br />

enrolled in the expansion cohort of 3 mg/kg Ipilimumab.<br />

PSA declines of more than 50% (Partial<br />

Response, PR) were observed in 5 of 22 patients<br />

in the 3-5 mg/kg Ipilimumab dose cohorts, and<br />

were associated with Autoimmune Breakthrough<br />

Events (ABE), including Grade 2 or 3 hypophysitis<br />

and Grade 3 alveolitis. PSA stabilizations (Stable<br />

Disease, SD) were observed in 1/3 patients in lower<br />

(0.3 and 1 mg/kg), and in 7 of 22 in the higher (3-5<br />

mg/kg) dose levels. Moreover, regressing bone and<br />

lymph node metastases were observed in 2/5 PR<br />

patients.<br />

Immune response monitoring was performed to<br />

identify changes that might predict or correlate<br />

with clinical efficacy. Significant increases in frequencies<br />

of activated and effector CD4+ and CD8+<br />

T cells were observed by HLA-DR and ICOS expression<br />

upon administration of high (3 and 5 mg/kg)<br />

but not of low (0.3 and 1 mg/kg) Ipilimumab doses;<br />

a relation to clinical behaviour was only observed<br />

for HLA-DR with earlier and more pronounced increases<br />

in patients with PR or SD. As an indication<br />

of tumor-specific responsiveness HLA-Tetramer<br />

(Tm) and seroreactivity to NY-ESO and PSMA were<br />

tested. For NY-ESO, therapy-induced increased seroreactivity<br />

was observed in 6/28 patients, which in<br />

2 patients was confirmed to coincide with increased<br />

Tm reactivity. PSMA seroconversions were observed<br />

in a total of 12/28 patients (and, of note, in<br />

4/5 PR), but no Tm positivity at any time was found<br />

in a total of 15 patients tested. PSMA seroconversion<br />

in the higher dose levels was associated with<br />

increased overall survival (P=0.062). In addition,<br />

significantly increased ex vivo levels of IL-4 in both<br />

CD4+ and CD8+ T cells were observed in patients<br />

with PR or SD (P


022 Pfirschke | Immune monitoring<br />

The repertoire of melanoma antigens recognized by T cell responses<br />

in malignant melanoma patients<br />

Christina Pfirschke 1 , Christoffer Gebhardt 2 , Alexander Enk 2 , Philipp Beckhove 1<br />

1 Translational Immunology Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany<br />

2 Department of Dermatology, University of Heidelberg, Heidelberg, Germany<br />

Malignant melanoma, the leading cause of skin cancer<br />

related death, is worldwide constantly increasing.<br />

During the recent years the development of novel, improved<br />

immunotherapeutic strategies have therefore<br />

become highly important. Pre-existing memory CD4<br />

and CD8 T cell responses are present in patients with<br />

many tumor entities and provide a repertoire of functional<br />

and potentially productive immune cells. Boosting<br />

patients tailored of such T cells with selected<br />

long peptides resembles a new strategy in melanoma<br />

immunotherapy.<br />

We here evaluated the presence and specificity of<br />

the repertoire of preexisting tumor-antigen specific<br />

T cells in 38 melanoma patients analyzed 1-4 month<br />

after primary tumor rejection. We have determined by<br />

an ex vivo short-term IFNγ ELISPOT assay the reactivity<br />

of spontaneously induced T cells in the blood of<br />

malignant melanoma patients against a broad variety<br />

of melanoma tumor-associated antigens (TAAs). To<br />

this end, long peptides (50aa) that cover several MHC<br />

class I- and II-restricted epitopes were used as test antigens<br />

while human IgG was investigated as negative<br />

control antigen. As antigen presenting cells, we used<br />

dendritic cells generated in vitro from monocytes<br />

and pulsed with different synthetic 50aa peptides<br />

derived from immunodominant regions of described<br />

melanoma TAAs like MelanA/MART-1, tyrosinase,<br />

gp100/pmel17, NY-Eso-1, p53, MDM2, NA17-A, TRP2,<br />

MAGEA1, MAGE-C2, GAGE-1, MIF and RAB38/NY-<br />

Mel-1. In order to analyze the role of regulatory T cells<br />

(Tregs) in suppressing TAA-specific T cell responses,<br />

Tregs were depleted from a part of the purified CD3+<br />

T cell fractions using αCD25-magnetic beads.<br />

More than 80% of the analyzed T cell samples of melanoma<br />

patients reacted against at least one tested<br />

peptide. T cells of HLA-A2 positive and HLA-A2 negative<br />

patients recognized the polypeptides to a similar<br />

extent. Additionally, more than 70% of the responding<br />

T cell samples reacted in a polyvalent manner<br />

to 4 or more tested peptides at the same time. Some<br />

peptides, such as MelanA/MART-1 (56%), tyrosinase<br />

(55%) and NA17-A (50%), as well as MAGE-A1<br />

(44%), MDM2 (42%), GAGE-1 (41%), RAB38/NY-Mel<br />

(40%), TRP2 (35%) and p53 (35%) were preferentially<br />

recognized whereas other melanoma TAAs, such<br />

as MIF (30%), MAGE-C2 (29%), gp100/pmel17 (24%)<br />

and NY-Eso-1 (18%) elicited lower spontaneous T cell<br />

responses. Interestingly, the depletion of Tregs from<br />

the investigated T cell fractions did not result in an<br />

increase of T cell response compared with the non<br />

Treg depleted T cell samples.<br />

Summarized, our data demonstrate that in a majority<br />

of malignant melanoma patients in the peripheral<br />

blood a polyvalent repertoire of tumor-reactive<br />

memory T cells exists. In contrast to other kinds of<br />

tumors investigated, including colon cancer, for malignant<br />

melanoma we did not detect a major role of<br />

Tregs for control of tumor reactive memory T cells.<br />

Moreover, some analyzed antigens appear to be dominant<br />

targets of spontaneous effector T cell responses<br />

providing a potential new criterion for selection of<br />

vaccine antigens for melanoma immunotherapy.<br />

63


023 Veron | Immune monitoring<br />

Selection, validation and evaluation of a 30-Plex Luminex kit<br />

for immunomonitoring of a phase I clinical trial<br />

Laure Veron 1 , Delphine Olivier 1 , Bérangère Marie-Bastien 2 , Benoît Grellier 2 , Edwige Bonfils 2 ,<br />

Bastien Calmels 2 , Philippe Ancian 2 , Jean Yves Bonnefoy 2 , Geneviève Inchauspé 1 , Christine Bain 1<br />

1<br />

Transgene SA, Centre of Infectiology, LYON, FRANCE<br />

2<br />

Transgene SA, Parc d‘innovation, ILLKIRCH GRAFFENSTADEN, FRANCE<br />

64<br />

Multiparametric technologies, such as the Luminex<br />

Bead Array technology, deserve to be evaluated<br />

for their potency to monitor immune responses<br />

induced by immunotherapeutic products.<br />

In a preliminary step, the Linco human 30-Plex kit<br />

was selected among three commercial kits based on<br />

technical (sample volume, time consumption and<br />

internal quality controls) and performance criteria<br />

(sensitivity) for further validation. The validation<br />

study was designed to evaluate the performance of<br />

the Linco 30-Plex kit for the measurement of analyte<br />

concentrations in antigen-stimulated PBMC culture<br />

supernatants. We also evaluated the impact of the<br />

in vitro stimulation step on the precision of this<br />

read-out for the determination of antigen-specific<br />

immune responses. Our data revealed that 1/ Accuracy<br />

and precision of the Linco 30-Plex assay were<br />

within acceptable limits for immunoassays, except<br />

for MCP-1 (Monocyte Chemotactic Protein-1) ; 2/<br />

Depending on the analyte, a matrix effect of the<br />

complete medium was observed, especially at the<br />

1/10 dilution ; 3/ Linearity of dilutions was observed,<br />

especially for analytes that displayed a good<br />

recovery at low concentrations and 4/ The in vitro<br />

stimulation step was the main source of variability<br />

and should be taken into account for the analysis<br />

of samples from clinical trials.<br />

This Linco 30-plex kit was then evaluated within<br />

the framework of a phase I clinical trial in 15 HCV<br />

(Hepatitis C virus)-chronically infected patients<br />

who received escalating doses of TG4040, a MVA<br />

(Modified vaccine Ankara) -based therapeutic<br />

vaccine encoding 3 HCV non structural proteins.<br />

Different cytokine profiles were observed in the 15<br />

patients, especially a contrasting profile between<br />

the best TG4040 responder, showing a decrease<br />

of more than 0.5 log of the viral load (VL), and a<br />

TG4040 non responder patient (ie who displayed<br />

less than 0.5 log VL decrease), with respectively a<br />

broad panel of Th1/pro-inflammatory biased cytokines<br />

for the former and a limited Th2 biased cytokines<br />

for the latter.<br />

Overall, Luminex technology can be used to follow<br />

immune responses after therapeutic vaccination.<br />

Different parameters however need to be carefully<br />

taken into account for a correct interpretation of<br />

the data, such as matrix effect, and particularly the<br />

variabilitity of the in vitro stimulation step


024 Zupke | Immune monitoring<br />

Nanoparticles for efficient labeling of dendritic cells to be used<br />

in cellular immunotherapy<br />

Oliver Zupke 1 , Eva Distler 1 , Daniela Baumann 2 , Dennis Strand 3 , Ralf Georg Meyer 1 , Katharina<br />

Landfester 2 , Wolfgang Herr 1 , Volker Mailänder 1 , 2 *<br />

1 Dept. of Medicine III, Hematology & Oncology, University Medical Center, Mainz, Germany<br />

2 Max Planck Institute (MPI) for Polymer Research, Mainz, Germany<br />

3 Dept. of Medicine I, University Medical Center, Mainz, Germany<br />

The goal of this project is to provide a platform for<br />

the evaluation of cellular immune therapies by monitoring<br />

the fate of administered cells in vivo over<br />

time. Therefore, the effective labeling of immune<br />

cells is essential. Here, immunostimulatory cells<br />

like dendritic cells (DCs) and immune effector cells<br />

like CD8 and CD4 T lymphocytes are of high interest.<br />

In the first step, a broad panel of polymeric<br />

nanoparticles (NPs) with different surface functionalities<br />

(synthesized at the MPI for Polymer Research,<br />

Mainz) was screened for their suitability as<br />

labeling agents for human monocyte-derived DCs.<br />

These NPs contain a fluorescent dye (PMI, perylenmonoimide),<br />

allowing the quantitation of cellular<br />

uptake by flow cytometry. We observed that aminofunctionalized<br />

NPs are most efficient for DC labeling.<br />

In subsequent experiments, the concentration<br />

as well as the incubation time of NPs was optimized,<br />

regarding a maximal cellular uptake and on<br />

the same time minimal toxic effects, as determined<br />

by 7-AAD stainings. To confirm intracellular localization<br />

and to exclude mere attachment of NPs at the<br />

cell surface, NP-loaded immature and mature DCs<br />

were also analyzed by confocal laser scanning microscopy.<br />

Kinetic studies demonstrated that labeled<br />

mature DCs still contained high amounts of NPs<br />

for a culture period of up to 8 days, what will be of<br />

importance for later in vivo trafficking studies. In<br />

further experiments, the influence of NP loading<br />

on phenotype and cellular function of DCs was<br />

investigated. Flow cytometry analysis of different<br />

maturation, homing, and costimulatory molecules<br />

demonstrated that NP labeling by this protocol<br />

had no significant influence on DC phenotype. A<br />

possibly negative influence of NP loading on the<br />

immunostimulatory functions of DCs could be excluded<br />

on the basis of allo-MLR (mixed lymphocyte<br />

reaction) and cytokine ELISPOT assays. CD4 and<br />

CD8 T cells stimulated in MLRs with NP-labeled,<br />

allogeneic DCs demonstrated comparably strong<br />

proliferation in 3H-Thymidin assays compared to<br />

stimulation with unlabeled DCs. In addition, IFN-γ<br />

spot production of alloreactive T cells (stimulated<br />

with HLA-mismatched DCs) as well as virus-specific<br />

memory T cells (stimulated with CMV peptideloaded<br />

DCs) was not impaired by NP labeling of the<br />

antigen-presenting cells. Furthermore the ability<br />

for processing viral protein antigens (Influenza A)<br />

and their presentation via MHC II molecules to CD4<br />

T cells was not affected by NPs.<br />

In summary, we developed a protocol using amino-functionalized,<br />

fluorochrome-containing polymeric<br />

nanoparticles that allows the efficient labeling<br />

of human dendritic cells, without significant<br />

negative impact on cellular phenotype or function<br />

of DCs as antigen-presenting cells. Currently, the<br />

protocol is adapted to human leukemia-, virus-, and<br />

alloreactive T lymphocytes. Furthermore, experiments<br />

with superparamagnetic iron oxide NPs are<br />

ongoing, with respect to intended in vivo trafficking<br />

studies of transferred immune cells in humanized<br />

mice by magnetic resonance imaging.<br />

65


025 Savelyeva | Immune monitoring<br />

Vaccination and immune memory: modelling the bystander<br />

activation of CD4+ memory T cells in mice<br />

Natalia Savelyeva, Amy Suchacki, Stephen Thirdborough, Freda K. Stevenson & Gianfranco Di Genova<br />

Cancer Sciences Division, Southampton University Hospitals, Tremona RD, Southampton, UK<br />

66<br />

CD4+ T-helper (Th) cells have a key role in pro-<br />

moting both B and T cell-mediated immunity.<br />

Knowledge of the mechanisms involved in the<br />

maintenance of vaccine-induced or natural CD4<br />

T cell immunity is crucial for the development of<br />

successful vaccination strategies against infectious<br />

diseases and cancer. We previously demonstrated<br />

that in healthy volunteers vaccination with tetanus<br />

toxoid (TT) induced bystander activation of Th<br />

memory cells of unrelated specificities; however<br />

antibody responses by B cells remained vaccinespecific<br />

(Di Genova G et al. Blood 2006). We hypothesized<br />

that this bystander activation could<br />

represent a mechanism which contributes to their<br />

long-term maintenance. We have now modelled<br />

this phenomenon in mice, confirming the results<br />

observed in humans and paving the way to a better<br />

understanding of the immunological mechanisms<br />

responsible for it. Briefly, in a first model two populations<br />

of CD4+ memory T cells specific for two<br />

distinct antigens were generated simultaneously in<br />

C57BL/6 mice. When mice were re-challenged with<br />

one of the two antigens, this caused not only the<br />

expected recall immune response but also expansion<br />

in the number of cytokine-producing cells specific<br />

for the unrelated antigen. In a second model,<br />

CD4+ TCR transgenic T cells (OT-II), either naïve or<br />

previously activated in vitro with cognate antigen,<br />

were CFSE-labelled and transferred into wild type<br />

recipient mice which were immune to TT. Recipient<br />

mice were then challenged with TT antigen and<br />

susceptibility of OT-II cells to bystander activation<br />

and proliferation was tested. Antigen-activated but<br />

not naïve cells were responsive and underwent bystander<br />

proliferation. This was proportional to the<br />

strength of the TT-specific memory T-cell response<br />

and appeared to be mediated by the common<br />

gamma chain cytokines IL-2 and IL-7. These data<br />

support the principle that CD4+ T cells at various<br />

stages of differentiation, from activated to memory<br />

cells, can respond to cytokines generated during<br />

parallel immune responses, such as those induced<br />

in human subjects by vaccines or natural exposure<br />

to environmental antigens. This might represent a<br />

mechanism which could contribute to their antigenindependent<br />

maintenance. The findings have high<br />

relevance for the measurement of T-cell responses<br />

in patients who are being vaccinated.


026 Tanriverdi-Akhisaroglu | Immune monitoring<br />

Spontaneous tumour antigen specific T cell responses occur<br />

frequently in patients with Carcinoma of Unknown<br />

Primary and are predominantly directed against EGFR,<br />

telomerase, MAGE-3 and P53<br />

Serpil Tanriverdi-Akhisaroglu 1 , Harald Löffler 2 , Yingzi Ge 1 , Kim Pietsch 1 , Andreas Bonertz 1 ,<br />

Alwin Krämer 2 , Philipp Beckhove 1<br />

1 Division of Translational Immunology<br />

2 Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center, Heidelberg<br />

Introduction: Carcinoma of unknown primary<br />

origin (CUP) represents a so far largely uncharacterized<br />

tumour entity. Since treatment options<br />

and conventional therapy regimens only provide<br />

very limited success (median overall survival of<br />

3-11 months), alternative treatment options need<br />

to be assessed. Tumour immunotherapy has recently<br />

proven clinical efficiency and might provide an<br />

option for the treatment of CUP.<br />

Purpose: As a first basis for the development of<br />

tumour immunotherapy for CUP, we here evaluated,<br />

to what extent CUP patients develop spontaneous<br />

tumour antigen specific T cell responses. In<br />

addition, we characterized the antigen specificities<br />

of tumour-reactive T cells of CUP patients in comparison<br />

to patients with colorectal and pancreatic<br />

carcinoma, as well as breast cancer.<br />

Patients and Methods: We analyzed T cell responses<br />

from peripheral blood of 18 CUP patients. The presence<br />

of tumour antigen specific T cells was assessed<br />

by short term ex vivo IFN-γ ELISPOT assay.<br />

As test antigens, we used a broad panel of long<br />

synthetic peptides derived from immunogenic sequences<br />

of the following defined tumour antigens:<br />

MUC-11-100, MUC-1(1137-157)5, EGFR(479-528),<br />

p53(118-167), Her-2/neu(351-384), CEA(569-618),<br />

Mage3(271-314-157), NYESO1, survivin(93-142),<br />

telomerase(958-1007), heparanase(1-50, 163-212).<br />

As negative control antigen, we used human IgG.<br />

Test or control antigens were loaded on autologous<br />

DCs at a concentration of 200µg/ml and pulsed DCs<br />

were incubated on coated IFN-γ ELISPOT plates for<br />

40 hours. Spots were measured automatically by an<br />

ELISPOT reader and T cell reactivity was assumed<br />

if spot numbers in triplicate test wells were significantly<br />

increased compared to triplicate control<br />

wells. Test results were compared to results of peripheral<br />

blood-derived T cell samples from patients<br />

with pancreatic- , colorectal- or breast cancer or<br />

from healthy volunteers.<br />

Results: Interestingly, we observed in all (100%)<br />

patients pre-existing, tumour antigenspecific T<br />

cells with an average number of 3.4 antigens simultaneously<br />

recognized. This compares favourably to<br />

spontaneous T cell responses detectable in patients<br />

with pancreatic cancer (85%), breast cancer (61%),<br />

colorectal cancer (60%) and strongly exceeded<br />

the minor rates of TA-specific T cell responses in<br />

healthy individuals. CUP patients developed predominant<br />

T cell responses against EGFR (50%),<br />

telomerase (47%), Mage3 (40%) and p53 (39%),<br />

whereas responses against the same peptides were<br />

less frequent in CRC patients (12%,0%,20% and<br />

21%, respectively), pancreatic carcinoma patients<br />

(23%,20%,23% and 26%) and also in breast cancer<br />

patients (47 %,21%, 29% and 41%). Particularly, T<br />

cell responses against telomerase were significantly<br />

increased in CUP patients as compared to the<br />

other defined tumour entities (p=0.03).<br />

Conclusion: CUP patients develop polyvalent T cell<br />

responses against a distinct repertoire of tumour<br />

antigens, particularly against EGFR, telomerase,<br />

MAGE-3 and P53, suggesting that these antigens<br />

may be candidates for antigen specific immunotherapy<br />

against CUP.<br />

67


027 Low | Immune monitoring<br />

Culturing PBMC in the presence of antigen results in a measurable<br />

expansion of memory T cells<br />

Lindsey Low, Ann Mander, Kathy Tier, Christian Ottensmeier<br />

Cancer Research UK Clinical Centre, Cancer Sciences Division, University of Southampton<br />

68<br />

Following an in vivo encounter with an antigen,<br />

there is a primary expansion of effector CD8 T cells,<br />

followed by a contraction in which 90 – 95% of<br />

effector cells die. A small population of memory<br />

cells remain which can persist long term, and will<br />

expand to produce effector cells if the antigen is encountered<br />

again. Culturing of PBMC in the presence<br />

of antigen is believed to stimulate this expansion,<br />

producing antigen-specific effector cells which can<br />

be detected using standard immunological assays.<br />

The protocol was developed using cryporeserved<br />

PBMC from HLA A2 positive heathy donors, cultured<br />

in the presence of IL-2 with the viral peptide<br />

antigens, cytomegalovirus (CMV), CMV pp65 493-<br />

499, (NLVPMVAVT); influenza A, Matrix 1 58-66,<br />

(GILCFVFTL); Epstein Barr virus (EBV), BMLFI<br />

259-217 (GLCTLVAML), and measles, non-structural<br />

C protein 84-92 (KLWESPQEI). Variables optimized<br />

included cell concentration, peptide concentration,<br />

well size, time spent in culture, feeding<br />

regimen, medium and use of serum. The resulting<br />

protocol was shown to give an optimum yield of antigen-specific<br />

effector cells at the end of the culture<br />

process. Cultured cells were used in immunological<br />

assays to determine phenotype and fuctionality.<br />

FACS analysis of pre- and post-culture cells has been<br />

used to show a measureable post-culture increase<br />

in the population of effector cells. Use of cultured<br />

cells in standard IFNγ ELISpots shows a noticeable<br />

amplification of antigen-specific response rates<br />

when compared to non-cultured cells for both the<br />

test viral antigens and for low frequency tumour<br />

associated antigens in clinical trial usage.<br />

This indirect measurement of the memory cell population<br />

provides a sensitive and reliable method to<br />

monitor the long term effectiveness of immunotherapeutic<br />

vaccination.


028 Laske | Immune monitoring<br />

T-cell immunomonitoring in patients with renal cell carcinoma<br />

after multi-peptide vaccination<br />

Karoline Laske 1 , Annemarie Dröge 1 , Susan Feyerabend 2 , Jörg Hennenlotter 2 , Jens Bedke 2 , Patricia<br />

Hrstic 1 , Stefan Stevanovic 1 , Arnulf Stenzl 2 , Cécile Gouttefangeas 1 , Hans-Georg Rammensee 1<br />

1 Institute for Cell Biology, Department of Immunology, Eberhard Karls University Tuebingen,<br />

Auf der Morgenstelle 15, 72076 Tuebingen, Germany<br />

2 Department of Urology, Eberhard Karls University Tuebingen, Hoppe-Seyler-Str. 3, 72076 Tuebingen,<br />

Germany<br />

In this clinical trial, patients with advanced or<br />

metastatic renal cell carcinoma after complete resection<br />

receive a multi-peptide cocktail containing<br />

HLA-class I and II binding peptides. The vaccine is<br />

applied either intradermally with GM-CSF as adjuvant<br />

or subcutaneously in Montanide ISA52 at<br />

18 vaccination time points over 12 months or until<br />

progression. Computer tomography imaging is performed<br />

to evaluate progression-free survival every<br />

3 months. HLA-A*02 positive or negative patients<br />

receive an individual peptide cocktail containing<br />

between 3 and 12 HLA-class I binding peptides.<br />

These peptides are known epitopes or ligands<br />

derived from selected tumor-associated antigens.<br />

Furthermore, the peptide cocktail for HLA-A*02 positive<br />

patients contains one peptide (from the influenza<br />

virus) as recall control and one peptide (from<br />

HBV) as priming control for the vaccination.<br />

Additionally, 3 to 4 HLA-class II binding peptides<br />

are included for CD4+ T-cell stimulation. Until<br />

now, 17/40 patients have been recruited and T-cell<br />

monitoring was performed for 9 patients. Blood<br />

was collected at each vaccination time point, serum<br />

and peripheral blood mononuclear cells (PBMCs)<br />

were isolated and frozen. For T-cell monitoring,<br />

PBMCs from different time points were thawed and<br />

stimulated with the individual peptide cocktail for<br />

12 days. Detection of specific T-cells was performed<br />

by IFN-gamma Elispot, tetramer staining (for<br />

HLA-A*02 positive patients) and intracellular cytokine<br />

staining. So far, all 7 monitored HLA-A*02<br />

positive patients developed a strong CD4+ T-cell<br />

response against 2 of the 4 HLAclass II binding<br />

epitopes (derived from carbonic anhydrase 9 and<br />

cyclin-D1 protein) which were detected by Elispot<br />

and further characterized by intracellular cytokine<br />

staining. We could detect a CD8+ T-cell response<br />

in 4 of 7 HLA-A*02 positive patients. The most frequently<br />

recognized peptides are from the proteins<br />

cyclin-D1 and guanylate cyclase 1.<br />

69


029 Brix | Immune monitoring<br />

Culturing PBMC in the presence of antigen results in a measurable<br />

expansion of memory T cells<br />

Liselotte Brix, Tina Jakobsen, and Henrik Pedersen<br />

Immudex, Copenhagen, Denmark<br />

70<br />

Sensitive and reliable monitoring of cellular immune<br />

responses is becoming increasingly important in<br />

cancer vaccine and immunotherapeutic development.<br />

Flow analysis using conventional fluorescent<br />

MHC multimers like Tetramers and Pentamers has<br />

made a great impact in this field enabling visualization,<br />

enumeration and phenotypic characterization<br />

of antigen-specific T-cells. However, shortcomings<br />

such as difficulties in detecting low-affinity interactions,<br />

e.g. tumor-specific T-cell responses, low<br />

reproducibility and reagent stability are still prominent<br />

problems.<br />

MHC Dextramer reagents are a new generation of<br />

fluorescent MHC multimers that solve some of those<br />

problems. Thus, we have used MHC Dextramers to<br />

detect low-affinity T cells (i.e. T cells carrying TCR<br />

with low affinity for the MHC-peptide complex).<br />

In contrast to Tetramers, the Dextramers allowed<br />

enumeration of T cells carrying TCRs with a Kd of<br />

> 250 µM. To our knowledge this is the first time<br />

that antigen-specific T cells with such low-affinity<br />

TCR have been detected with a fluorescent MHC<br />

multimer reagent.<br />

When staining T cells of higher affinity-TCR’s (5<br />

µM – 100 µM), Dextramers had much higher resolution<br />

than the corresponding Tetramers. Dextramers<br />

thus provide a more reliable means for identification<br />

of true positive antigen-specific T cells.<br />

The increased sensitivity and resolution of the<br />

Dextramers probably reflects the higher number of<br />

MHC-peptide complexes and fluorochromes, since<br />

the high number of MHC-peptide complexes increases<br />

their avidity for the specific T-cells, and the<br />

higher number of fluorochromes enhances staining<br />

intensity. As a result, Dextramers have higher sensitivity,<br />

higher staining intensity and higher signalto-noise<br />

ratio.<br />

The dextran backbone of the MHC Dextramer<br />

stabilizes the attached proteins. When analyzing<br />

the stability of the MHC Dextramers, a given<br />

blood sample could be stained reproducibly well<br />

using the same batch of a MHC Dextramer for<br />

more than 4 month. Likewise, different batches<br />

of MHC Dextramers provided similar frequencies<br />

of antigen-specific T cells with comparable staining<br />

intensities when staining the same sample.<br />

We also investigated the impact of freezing and<br />

thawing MHC Dextramers. Surprisingly, the MHC<br />

Dextramer reagent could be subjected to repeated<br />

freeze-thaw cycles without affecting the staining<br />

intensity, resolution or the number of positive cells<br />

detected.<br />

In summary, MHC Dextramers are sensitive, highly<br />

stable reagents that are suitable to minimize assayto-assay<br />

variation, and that are particularly useful<br />

reagents in the immune monitoring of cancer immunotherapy.


030 Brix | Immune monitoring<br />

MHC Dextramers: improved detection of antigen-specific T-cells<br />

in fluid samples and in situ<br />

Liselotte Brix, Tina Jakobsen, and Henrik Pedersen<br />

Immudex, Copenhagen, Denmark<br />

Sensitive and reliable monitoring of cellular immune<br />

responses is becoming increasingly important in<br />

vaccine and immunotherapeutic development.<br />

Flow analysis using fluorescent MHC multimers<br />

has made a great impact in this field enabling visualization,<br />

enumeration and phenotypic characterization<br />

of antigen specific T-cells. Here we discuss<br />

how MHC Dextramers, a new generation of MHC<br />

multimer reagents, can be used for advanced detection<br />

of antigen specific T-cells in comprehensive<br />

applications.<br />

MHC Dextramer reagents carry a higher number<br />

of MHC-molecules and more fluorochromes than<br />

conventional MHC multimers. This increases their<br />

avidity for the specific T-cell and enhances their<br />

staining intensity, thereby increasing resolution and<br />

the signal-to-noise ratio. Furthermore the dextran<br />

backbone of the MHC Dextramer stabilizes the attached<br />

MHC-molecules and fluorochrome proteins.<br />

This allows specialized applications such as in situ<br />

staining (e.g. detection of antigen-specific T-cells<br />

in frozen tissue sections), detection of very rare<br />

events, combined staining with high numbers of<br />

MHC Dextramer reagents in same tube and MHC<br />

Dextramer staining combined with extracellular<br />

and intracellular staining for e.g. surface proteins<br />

and cytokines.<br />

Such specialized applications are useful in improving<br />

immune monitoring of cancer immunotherapeutics.<br />

71


L124 Zhang | Immune monitoring<br />

Goal of ELISPOT proficiency accomplished: ELISPOT assays provide<br />

reproducible results among different laboratories for T-cell<br />

immune monitoring — even in hands of ELISPOT novices<br />

W. Zhang 1 , R. Caspell 1 , A. Y. Karulin 1 , M. Ahmad 2 , N. Haicheur 3 , A. Abdelsalam 4 , K.<br />

Johannesen 5 , V. Vignard 6 , P. Dudzik 7 , K. Georgakopoulou 8 , A. Mihaylova 9 , K. Silina 10 , N<br />

Aptsiauri 11 , V. Adams 12 , P. V. Lehmann 1,13 , and S. McArdle 2<br />

1 Cellular Technology Ltd., Shaker Hts. Ohio, USA<br />

2 Nottingham Trent University, Nottingham, UK<br />

3 Hôpital Européen Georges Pompidou, Paris, France<br />

4 University of Pittsburgh, Pittsburgh, PA, USA<br />

5<br />

Norwegian Radium Institute, Oslo,Norway<br />

6<br />

INSERM, Nantes, France<br />

7<br />

Jagiellonian University Medical College, Krakow, Poland<br />

8 Immunology Center, St. Savas Cancer Hospital, Athens, Greece<br />

9<br />

UMBAL “Alexandrovska”, Sofia, Bulgaria<br />

10<br />

Biomedical Research Study Centre, Riga, Latvia<br />

11<br />

Hospital Universitario Virgen De la Nieves, Granada, Spain<br />

12<br />

Onyvax Ltd, London, UK<br />

13<br />

Department of Pathology, Case Western Reserve University, Cleveland OH, USA<br />

72<br />

T cell monitoring remains challenging in tumor<br />

vaccine trials due to the necessity of testing live<br />

cells in functional assays, and the low frequencies<br />

of tumor antigen-specific T cells. Recent multi-center<br />

initiatives that aimed at harmonizing T<br />

cell assays have drawn attention to alarming- 30-,<br />

20- and 150- fold inter-laboratory variations in test<br />

results for ELISPOT, ICS and tetramers, respectively<br />

(Immunity, 2009, 31: 527-528). The authors concluded:<br />

“The high degree in variability makes the<br />

comparison between any two labs become a game<br />

of chance”. Puzzled and alarmed by this message,<br />

we undertook a similar effort for ELISPOT together<br />

with a European consortium, NEUCAPs. For<br />

our study, we required that all participants from<br />

the eleven reporting labs follow the same detailed<br />

protocol using one uniform platform, and that the<br />

study participants had never previously conducted<br />

an ELISPOT assay (the results of their first attempt<br />

were recorded for the study). While three of the<br />

labs failed with the basic logistics of the trial, eight<br />

detected the peptide-specific CD8+ T-cells in frequencies<br />

approximating the values established by<br />

the Reference Laboratory. These results show that<br />

ELISPOT can produce comparable, reliable data<br />

(even from untrained personnel) if a standardized<br />

platform for the assay and data analysis is followed.<br />

Since ELISPOT assays have been qualified<br />

and validated for regulated studies, they are ideal<br />

candidates for robust and reproducible monitoring<br />

of Tcell immunity in tumor vaccine trials.


031 Gross | Immune monitoring<br />

Monitoring of vaccine-specific regulatory and helper CD4+<br />

and cytolytic CD8+ T cells after vaccination with autologous<br />

antigen-loaded dendritic cells<br />

Stefanie Gross, Annett Hamann, Gerold Schuler, Eckhart Kämpgen, Beatrice Schuler-Thurner<br />

Department of Dermatology, University of Erlangen, Germany<br />

74<br />

Cancer vaccines have been shown to induce tumorspecific<br />

CD8+ cytolytic T cells (CTL) in the blood<br />

of cancer patients. However, clinical outcome often<br />

remains poor. This ‚‘cancer vaccine paradox‘‘ can<br />

nowadays be readily explained by tumor-escape<br />

and immune-regulatory mechanisms. It has been<br />

suspected, that cancer vaccines of different types<br />

might not only induce the desired CTL response<br />

but simultaneously also expand antigen-specific<br />

regulatory T cells (Treg), thus counteracting productive<br />

immune responses and preventing clinical<br />

efficacy of cancer vaccines. Of note is that it has<br />

been claimed that mature dendritic cells (DC) are<br />

not only expanding antigen-specific helper T cells<br />

but also the disadvantageous Tregs. The induction<br />

of antigen-specific Tregs by DC and other vaccines<br />

has, however, not yet been studied systematically<br />

by advanced immunomonitoring techniques.<br />

Frequencies and nature of vaccine-induced CD8+<br />

cytolytic and CD4+ helper or regulatory T cells in<br />

vaccinated cancer patients were analyzed ex vivo<br />

by 9 color flow cytometry. MHC Class I and MHC-<br />

Class II multimer-staining for different tyrosinase,<br />

MelanA, gp100, NY-ESO1 or MAGE3 epitopes was<br />

combined with intracellular staining for different<br />

signature-transcription factors such as FoxP3<br />

(Treg), GATA-3 (Th2) and RoR t (Th17) for CD4+<br />

T cells or function-associated molecules like perforine<br />

and granzyme B for CD8+ T cells.<br />

In contrast to common belief our data so far do<br />

not support the notion that tumor vaccination with<br />

autologous mature dendritic cells bears the risk<br />

to induce immuno-suppressive regulatory T cells.<br />

Remarkably, the same vaccine induced different<br />

immune responses in patients (Th1, Th2, granzyme<br />

B), pinpointing the likely benefit of patientselection<br />

and vaccination at early disease stages.


032 Zelba | Immune monitoring<br />

NY-ESO-1-specific T cells in long-term melanoma survivors<br />

Henning Zelba 1 , Benjamin Weide 2 , Claus Garbe 3 , Graham Pawelec 1 , Evelyna Derhovanessian 1<br />

1 Department of Internal Medicine II, Section for Transplantation Immunology and Immunohaematology,<br />

University Hospital, Tuebingen, Germany<br />

2 Department of Dermatology, Division of Dermatooncology, University Hospital, Tuebingen, Germany<br />

Immunotherapy has now become a potentially<br />

effective cancer treatment modality for certain<br />

patients. Theoretically any protein expressed abnormally<br />

by the tumour can serve as a target for<br />

antibodies or T cells, and there is an ever-increasing<br />

list of possible antigen targets. However, it<br />

is not clear whether targeting particular antigens<br />

can result in a better clinical outcome than others.<br />

Moreover, non-immunotherapeutic treatments may<br />

also result in the generation of immune responses<br />

against certain of these target antigens, which may<br />

be associated with improved patient survival.<br />

To test this, we are analysing exceptionally longterm<br />

melanoma survivors (LTS) in comparison to<br />

patients with bad prognosis who progressed during<br />

treatment and died within the usual time period.<br />

Patients included those receiving chemotherapy ( e.<br />

g. Dacarbazin, Temodal,…) and Immunotherapy (<br />

e. g. IL-2 injection, ...).<br />

We analysed CD4+ and CD8+ T-cell responses<br />

against the four common melanomaassociated antigens<br />

Melan-A, MAGE-A3, Survivin and NY-ESO-<br />

1. Antigen-specific T-cell responses were detected<br />

using peptide mixtures spanning the whole sequence<br />

of the molecule after one round of in vitro<br />

sensitization for 12 days. Intracellular production<br />

of six different cytokines (IFN-γ, TNF, IL-2, IL-4,<br />

IL-10 and IL-17) was measured simultaneously in<br />

both CD4+ and CD8+ T-cells by 14 colour multiparameter<br />

flow cytometry, allowing analysis of<br />

phenotype and function (TH1, TH2, TH17) at the<br />

single-cell level.<br />

In this way, we have identified NY-SO-1-specific T<br />

cells in 7 of 10 LTS but in only 1 of 6 short-term survivors.<br />

The prevalence of T-cells specific for Melan-<br />

A, MAGE-A3 or Survivin did not differ between LTS<br />

and other patients. Similar results were obtained<br />

in an independent vaccination trial targeting the<br />

above antigens. NY-ESO-1-specific immune responses<br />

were found in 3 of 5 patients with a good<br />

clinical outcome after vaccination, but in only 1 of<br />

7 patients with a poor clinical outcome. MAGE-A3specific<br />

responses were induced in the majority of<br />

patients after vaccination, but there was no difference<br />

between LTS and poor responders.<br />

In all LTS patients, as well as in the patients with<br />

good clinical outcome in the vaccination trial,<br />

CD4+ and CD8+ NY-ESO-1-specific T cells produced<br />

high levels of IFN-γ and TNF. In contrast,<br />

NY-ESO-1-specific T-cells in the two patients with<br />

bad prognosis did not produce any or only small<br />

amounts of these cytokines. Taken together, our<br />

data suggest that NY-ESO-1 is likely to be a promising<br />

target antigen for melanoma patients and<br />

might be a good marker to predict the clinical<br />

outcome of individual patients.<br />

75


033 Singh | Immune monitoring<br />

A stable standard sample for harmonisation and validation<br />

of T cell assay<br />

Satwinder Kaur Singh 1 , Bart Tummers 1 , Ton Schumacher 2 , Kees Franken 3 , Marij Welters 3 ,<br />

Cedrik M. Britten 4 , Sjoerd H. van der Burg 1<br />

1 Deparment of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands<br />

2 Department of Immunology, Dutch Cancer Institute, Amsterdam, the Netherlands<br />

3 Immunohematology and Blood transfusion, Leiden University Medical Center, Leiden, the Netherlands<br />

4 Third Medical Department, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany<br />

76<br />

Immune monitoring of clinical trials is a challenging<br />

task as different methods and protocols are applied<br />

to quantify vaccine-induced T cell responses. Heterogeneity<br />

in protocols and lack of standards make it<br />

difficult to compare results obtained from different<br />

clinical trials. Harmonisation of quantitative assays<br />

critically depends on samples containing defined<br />

numbers of antigen-specific T cells, however, the<br />

availability of such samples is limiting at best.<br />

We have developed a standard sample using the<br />

HLA-A2-restricted NY-ESO-1-specific T cell receptor<br />

(TCR)-transduced T cells spiked into autologous<br />

PBMC population. The NY-ESO-1- specific TCR<br />

was retrovirally transduced into purified CD8+<br />

T cells and NY-ESO-1-specific T cells were sorted<br />

using HLA-peptide tetramer to obtain >99% TCRtransduced<br />

pure CD8+ T-cell population. These<br />

purified cells were clonally expanded and then<br />

spiked either at a concentration of 0.25% or 0.5%<br />

into autologous PBMCs. Multiple batches were extensively<br />

tested for stability up to 6 months in time.<br />

The percentage of detected tetramer-positive T cells<br />

in subsequently tested frozen vials varied little to<br />

the calculated number of spiked cells (CV


034 Guidoboni | Immune monitoring<br />

Changes of immune cell infiltrates induced by dendritic cell<br />

vaccination in melanoma patients‘ tumor tissues:<br />

an immunohistochemical study<br />

Massimo Guidoboni 1 , Laura Ridolfi 1 , Annamaria Granato 1 , Massimiliano Petrini 1 , Laura<br />

Fiammenghi 1 , Valentina Ancarani 1 , Elena Pancisi 1 , Linda Valmorri 1 , Angela Riccobon 1 ,<br />

Roberta Gafà 2 , Giovanni Lanza 2 , Dino Amadori 3 , Ruggero Ridolfi 1<br />

1 Immunotherapy and Somatic Cell Therapy Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei<br />

Tumori, Meldola (FC), Italy<br />

2 Department of Pathology, University of Ferrara, Italy<br />

3 Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Meldola (FC), Italy<br />

We have been treated more than 40 patients in a<br />

phase I/II study of therapeutic vaccination using<br />

DC pulsed with autologous tumor lysate or homogenate<br />

in advanced melanoma. Objective responses<br />

are observed in a limited number of patients, clinical<br />

responses rates as evaluated by classical responses<br />

criteria are still disappointing, and we are not able<br />

to identify patients who will benefit from DC vaccination.<br />

Spontaneous and vaccine-induced antitumor<br />

immune responses have been extensively characterized<br />

in peripheral circulating compartment,<br />

and in some cases this approach predicted clinical<br />

response to immunotherapy; however, we do not<br />

know whether this translate into efficient immune<br />

cell recruiting in all tumor sites. To shed light on<br />

changes induced by DC vaccine on the composition<br />

of immune effectors in tumor sites, we evaluated<br />

tumor infiltrating lymphocytes (TILs) in 12<br />

tumor biopsies taken from 4 patients before and at<br />

different times after DC vaccination for metastatic<br />

melanoma. Two patients showed complete clinical<br />

responses (CR) after at least 6 courses of vaccine.<br />

In one of these patients DC vaccination induced<br />

huge amount of CD8+ activated cytotoxic lymphocytes<br />

(24,89 CD8+ TILs/100 melanoma cells)<br />

in one tumor site compared to prevaccine biopsy;<br />

however, negligible levels of CD8+ TILs together<br />

with considerable amount of CD4+ cells were found<br />

in one other relapsed site, suggesting that immune<br />

escape had occurred. The other patient showing<br />

CR underwent relapse and progressive surgical de-<br />

bulking of metachronous localizations led to surgical<br />

CR still lasting. Tumor biopsies taken after<br />

vaccination showed lower amount of CD8+ TILs<br />

than those collected before (2,13 and 1,38 vs 8,44<br />

CD8+ TILs/100 tumor cells), showing that complete<br />

surgical removal of „immune escaped“ lesions<br />

may lead to persistent CR in patients with good<br />

clinical status. One patient undergone progression<br />

after Ipilimumab failure showed considerable<br />

levels of CD8+ TILs. After 9 courses of DC vaccine<br />

with stable disease (SD), she relapsed and progressing<br />

lesion was removed, with decreased amount of<br />

tumor infiltrating CD8+ lymphocytes in this tumor<br />

site. The last patient has been vaccinated for 4 years<br />

with still lasting SD, and further tumor biopsies<br />

were taken to prepare additional doses of vaccine.<br />

Treatment induced lower increase of CD8+ TILs<br />

in postvaccine biopsies than those observed in the<br />

other patients (3,28 and 1,93 vs 1,12 CD8+ TILs/100<br />

tumor cells), suggesting that lower immune pressure<br />

might allow to reach an equilibrium between<br />

immune system and tumor which slow occurrence<br />

of immune escape. Although performed on a<br />

limited number of cases, this study firstly explored<br />

the possible role for „tissue immune monitoring“ in<br />

the clinical setting and strongly indicate its use to<br />

direct clinical decision in multimodal therapeutic<br />

approach.<br />

77


035 Voland | Immune monitoring<br />

Characterisation and functional analysis of T-cell responses in<br />

melanoma patients vaccinated with peptide-loaded<br />

dentritic cells<br />

Steve Voland 1 , Stefanie Gross 1 , Beatrice Schuler-Thurner 1 , Pierre Coulie 2 , and Gerold Schuler 1<br />

1 Dept. Dermatology, University Hospital Erlangen, Erlangen, Germany<br />

2 de Duve Institute and Université catholique de Louvain, Brussels<br />

78<br />

T cell responses in melanoma patients vaccinated<br />

with autologous monocyte-derived dendritic cells<br />

(DC) loaded with peptides from different tumorassociated<br />

antigens (TAA) were characterized for<br />

their functional capacity.<br />

To assign a certain functional capacity (cytolytic<br />

activity, cytokine production and degranulation<br />

upon restimulation) to specific T cell clones we<br />

chose a limiting-dilution based approach. Frozen<br />

aliquots of PBMC from 5 melanoma patients who<br />

had received four vaccinations were thawed,<br />

loaded with peptide and seeded in a 96well plate<br />

followed by a 14-day culture. The cells were restimulated<br />

with autologous peptide-loaded PBMC<br />

at day 7. By splitting the cells two times during<br />

the 14-day culture four plates with identical clonal<br />

composition were obtained. Comparative analyses<br />

of each corresponding well were performed with<br />

the following assays:<br />

1. percentage of peptide-specific T cells, determined<br />

by MHC tetramer binding<br />

2. intracellular cytokine production (interferon-γ,<br />

interleukin 2, TNF-α) and degranulation (by CD107a<br />

mobilization) after antigenic stimulation<br />

3. cytolytic activity determined by a standard 51Crrelease<br />

assay<br />

In all 5 patients vaccine-specific CD8+ T cells were<br />

detected after in vitro presensitation with peptide.<br />

Detected responses differed in magnitudes and<br />

overall functional capacity. In most cases a positive<br />

correlation between lytic activity and antigenspecificity<br />

(MHC tetramer positivity) was found.<br />

Furthermore the lytic activity correlated positively<br />

with certain cytokine profiles with a pronounced<br />

IFN-γ and TNF-α proportion and to a lower extend<br />

also with IL-2 and CD107a.<br />

From our data can be concluded that vaccination<br />

with autologous monocyte-derived DCs loaded<br />

with TAA-derived peptides is capapble to induce<br />

antigen-specific CD8+ T cells, with the potential<br />

to produce different immune-stimulatory cytokines<br />

and which show cell mediated cytotoxitcity.<br />

Further investigations of the induced T cells will be<br />

conducted to determine the breadth of the induced<br />

immune response by analysis of the different T cell<br />

clones and their affinities.


036 Wagner | Immune monitoring<br />

GPI-anchor negative T cells with impaired effector function persist<br />

after Alemtuzumab mediated T-cell depletion<br />

Eva Maria Wagner 1 , Aline Naomi Lay 1 , Caroline Goetze 1 , Diana Wolff 1 , Julia Hemmerling 1 ,<br />

Matthias Theobald 1 , Wolfgang Herr 1 , Ralf Georg Meyer 1<br />

1 Department of Hematology/Oncology at the University Medical Center of the Johannes Gutenberg-University<br />

Mainz, Germany<br />

The monoclonal anti-CD52 antibody Alemtuzumab<br />

is frequently used for T-cell depletion (TCD) in the<br />

context of allogeneic hematopoietic stem cell transplantation<br />

(HSCT). We have previously demonstrated<br />

that substantial proportions of reconstituting T<br />

cells remained CD52 negative for years, especially<br />

in patients who did not receive donor lymphocyte<br />

infusions. CD52 is a glycosylphosphatidylinositol<br />

(GPI)-anchored molecule of unknown function. By<br />

flow-cytometry, we demonstrated that the lack of<br />

CD52-expression was associated with decreased<br />

expression of further GPI-anchored molecules like<br />

CD55 and CD59. We therefore analyzed CD52 negative<br />

T cells of 20 patients after HSCT with a fluorescent<br />

aerolysin staining (FLAER) that directly labels<br />

the GPI-anchor and found that the loss of CD52 and<br />

further GPI-anchored proteins in T cells was due to<br />

the loss of the GPI-anchors themselves. In contrast,<br />

CD52 positive T cells of the same patients showed<br />

no reduced FLAIR-signal and normal expression<br />

of CD55 and CD59. Patients who underwent Alemtuzumab-mediated<br />

TCD prior to allogeneic HSCT<br />

suffer from long-lasting immunologic dysfunction<br />

even when peripheral T cell counts have reconstituted<br />

to normal range. Hence, we compared GPI-anchor<br />

positive and negative T cells for their response<br />

to viral and allogeneic stimuli. When FACS-sorted<br />

CD52 negative and positive T cell populations were<br />

expanded in vitro (IL2, OKT3, feeder-cells), we observed<br />

that GPI-anchor expression remained absent<br />

in the CD52 negative T-cell cultures. In contrast, the<br />

CD52 positive T-cell cultures showed constant expression<br />

of GPI-anchors for more than 16 weeks. In<br />

addition, the CD52 expression had no influence on<br />

the growth-kinetics following antigen-independent<br />

stimulation with IL2 and OKT3. However, CD52 positive<br />

T cells showed enhanced proliferation after specific<br />

stimulation with CMV-pp65 peptides. In IFN-γ<br />

ELISPOT assays using CMV-peptide (pp65 and IE-1)<br />

loaded autologous dendritic cells as stimulators,<br />

specific spot production was only detected among<br />

the GPI-anchor positive T cells. To investigate on<br />

T-cell function ex vivo, we performed IFN-γ Secretion-assays<br />

with T cells directly isolated from patients.<br />

CD52 negative T cells again showed a reduced<br />

IFN-γ-secretion after stimulation with either autologous<br />

DCs loaded with the CMV-peptides or with<br />

allogeneic DCs. Nonetheless, T cells with CMV-pp65<br />

specific T cell receptors were present in the CD52<br />

positive as well as the CD52 negative subpopulations,<br />

as proven by tetramer-staining. Despite the<br />

functional differences, the percentages of naïve and<br />

memory T cells did not differ between both populations.<br />

In summary, we demonstrated that after Alemtuzumab-based<br />

TCD, substantial proportions of<br />

reconstituting donor T cells lost the expression of<br />

GPI-anchors. GPI-anchor negative T cells showed<br />

reduced proliferative capacity as well as reduced<br />

IFN-γ secretion in response to CMV or allogeneic<br />

stimulation. Our data suggest that a loss of GPI-anchored<br />

molecules might contribute to an impaired<br />

function of reconstituting donor T cells after Alemtuzumab-mediated<br />

TCD. We will further investigate<br />

on the impact of different GPI-anchored molecules<br />

on effector-functions of different T-cell subsets.<br />

79


037 Aarntzen | Immune monitoring<br />

Skin-derived functional specific Tcells predict clinical outcome<br />

upon DC vaccination in stage III and IV melanoma patients<br />

Erik Aarntzen 1,2 , Annemiek de Boer 1 , Nicole Meeusen-Scharenborg 1 , Danita Schuurhuis 1 ,<br />

Gerty Schreibelt 1 , Joost Lesterhuis 1,2 , Hans Jacobs 1 , Michelle van Rossum 3 , Carl Figdor 1 ,<br />

Gosse Adema 1 , Kees Punt 2 , Jolanda de Vries 1<br />

1<br />

Centro de investigación médica aplicada (CIMA)<br />

2<br />

Bristol Myers-Squibb Pharmaceutical research institute, Princeton, NJ<br />

3 Sidney Kimmel Cancer Center. Johns Hopkins Medical School. Baltimore, BA<br />

80<br />

Purpose: To improve immunotherapy against<br />

cancer, tumor-specific immunomonitoring is essen-<br />

tial. In this study, we analyze the predictive value of<br />

T cell cultures from Delayed Type Hypersensitivity<br />

(DTH) skin sites as monitoring tool after dendritic<br />

cell (DC) vaccination in melanoma patients.<br />

Patients and methods: In our ongoing trials, HLA-<br />

A2.1 positive stage III and IV melanoma patients<br />

are vaccinated with mature DC loaded with the<br />

tumor associated antigens gp100 and tyrosinase.<br />

After vaccination, tumor-specific response is routinely<br />

evaluated in blood and cultures from DTH<br />

infiltrating lymphocytes (DIL).<br />

Results: Almost all stage III and IV patients show<br />

both proliferative and humoral responses to the<br />

control antigen KLH. Tumor-specific CD8+ Tcells<br />

are rarely detected in peripheral blood (6 out of 73<br />

patients tested). In the vast majority of patients,<br />

sufficient cell numbers are obtained from DIL cultures<br />

for tetramerstaining and functional analysis.<br />

We show a strong predictive value of the presence<br />

of functional specific CD8+ Tcells in DIL cultures<br />

for favorable clinical outcome in both stage III<br />

(n=75) and IV (n=85) melanoma patients.<br />

Conclusion: Our findings show that the presence<br />

of functional tumor-specific Tcells from DTH skin<br />

biopsies highly predict clinical outcome.


038 Moodie | Immune monitoring<br />

Response definition criteria for ELISPOT assays revisited<br />

Zoe Moodie 1 , Leah Price 2 , Cécile Gouttefangeas 3 , Ann Mander 4 , Syliva Janetzki 5 , Marij J.P.<br />

Welters 6 , Christian Ottensmeier 4 , Sjoerd H. van der Burg 7 and Cedrik M. Britten 8<br />

1 Statistical Center for HIV/AIDS Research & Prevention (SCHARP) Fred Hutchinson Cancer Research<br />

2 Department of Biostatistics, New York University, New York, NY USA, Center, Seattle, WA USA<br />

3 Department of Immunology, University of Tuebingen, Tuebingen, Germany<br />

4 Experimental Cencer Medicine Centre and Cancer Sciences Division, Southampton University Hospitals, Southampton, UK<br />

5 ZellNet Consulting, Inc., Fort Lee, NJ, USA<br />

6 Department of Immunhematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands<br />

7 Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands<br />

8 8 III. Medical Department, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany<br />

No consensus exists on to how to determine if an<br />

immune response occurred based on raw data from<br />

an ELISPOT assay. The goal of this study was to<br />

enable investigators to understand currently available<br />

methods for response determination and to rationally<br />

select and apply a scientifically sound method appropriate<br />

to their specific laboratory setting. Five representative<br />

approaches were applied to data sets from<br />

the <strong>CIMT</strong> Immunoguiding Program and the response<br />

detection and false positive rates of each approach<br />

were compared. Simulation studies were also performed<br />

to compare empirical and statistical approaches.<br />

We conclude that both empirical and statistical approaches<br />

may serve as appropriate response definition<br />

criteria. However, we recommend the use of a nonparametric<br />

statistical test. To enable use of the recommended<br />

statistical tests, a web-based user interface<br />

was created: http://www.scharp.org/zoe/runDFR/.<br />

This interface allows the user to upload data from an<br />

ELISPOT assay and obtain the response determination<br />

based on the recommended statistical tests.<br />

Furthermore, we recommend the use of six medium<br />

control wells or four wells each for both medium<br />

control and experimental conditions be performed in<br />

order to increase the sensitivity of the assay. The study<br />

provides an estimate for replicate variation and limit<br />

of detection that can be expected from an average<br />

lab. The authors recommend that replicates with high<br />

variation and below the estimated limit of detection<br />

should be filtered out or interpreted with caution.<br />

81


039 Attig | Immune monitoring<br />

Recommendations for HLA-peptide multimer staining experiments<br />

- Results from the second HLA-peptide multimer proficiency<br />

phase organized by the Cancer Immunotherapy Consortium<br />

Sebastian Attig 1 *, Leah Price 2 *, Sylvia Janetzki 3 , Michael Kalos 4 , Michael Pride 5 , Lisa Mc<br />

Neil 5 , Tim Clay 6 , Jianda Yuan 7 , Kunle Odunsi 8 , Axel Hoos 9 , Pedro Romero 10 , Cedrik M. Britten<br />

1,11 for the CRI-CIC Assay Working Group<br />

1<br />

Division of Translational and Experimental Oncology, Department of Internal Medicine III, University Medical<br />

Center of the Johannes-Gutenberg University, Mainz, Germany<br />

2<br />

Department of Biostatistics, New York University, New York, NY USA<br />

3<br />

ZellNet Consulting, Inc., Fort Lee, NJ USA<br />

4<br />

Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Abramson<br />

Family Cancer Research Institute, Philadelphia, PA USA<br />

5<br />

ZellNet Consulting, Inc., Fort Lee, NJ, USA<br />

6<br />

Surgery and Immunology, Duke University Medical Center, Durham, NC, USA<br />

7<br />

Ludwig Center for Cancer Immunotherapy, Immunology Program, Sloan-Kettering Institute, New York,<br />

NY USA<br />

8<br />

Departments of Gynecologic Oncology and Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA<br />

9<br />

Bristol-Myers Squibb, Wallingford, CT USA<br />

10<br />

Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Lausanne Branch, University<br />

Hospital (CHUV), Lausanne, Switzerland<br />

11<br />

Clinical Development, BioNTech AG, Mainz, Germany<br />

82<br />

The Cancer research Institute‘s Cancer Immuno-<br />

therapy Consortium (CRI-CIC) has conducted a<br />

HLA-peptide multimer (MULTIMER) proficiency<br />

panel focusing on the impact of DUMP channel use.<br />

The study showed that the introduction of a dump<br />

channel did not impact on the detection rate of participating<br />

labs but did reduce the non-specific MUL-<br />

TIMER binding within the group. Both the use of<br />

DUMP channel and dead cell marker may decrease<br />

the amount of false positive events in CD8-positive<br />

population thus leading to a lower limit of detection<br />

of the test and provide a more accurate measure of<br />

the true MULTIMER specific binding. In 88% of<br />

all the CMV-MULTIMER replicates a response was<br />

detected, demonstrating that low frequency CMV<br />

responses were successfully detected by most of<br />

the labs. Variation of duplicate staining was below<br />

30% in 70% of participating labs indicating a very<br />

good reproducibility of results even for low frequency<br />

T cell responses. Introduction of a dump<br />

channel also decreased the variability in reported<br />

frequencies of pp65-specific CD8 T cells across the<br />

group. However, the response detection rates for<br />

the Melan-A responses were much lower which was<br />

due to the particular staining characteristics of the<br />

Melan-A-specific MULTIMER. Results from this<br />

proficiency panel phase clearly confirm the value<br />

of previously published harmonization guidelines,<br />

and have led to additional recommendations on<br />

assay performance and how to best present data<br />

from MULTIMER experiments.


Cellular therapy<br />

83


040 Kotsiou | Cellular therapy<br />

Application of MHC class I single chain trimers for leukaemia<br />

immunotherapy<br />

Eleni Kotsiou 1,2 , Julian Dyson 2 , Keith G Gould 1<br />

1<br />

Department of Immunology, Wright-Fleming Institute, St Mary’s campus, Imperial College,<br />

London W2 1PG, UK<br />

2<br />

Immunobiology Section, Commonwealth Building, Hammersmith Hospital, Imperial College,<br />

London W12 ONN, UK<br />

84<br />

Major Histocompatibility Complex (MHC) class I<br />

single chain trimers (SCTs) consist of antigenic<br />

peptide, β2-microglobulin and MHC heavy chain<br />

all joined together in a single polypeptide molecule<br />

via flexible linkers. SCTs are extremely valuable<br />

tools for the stimulation of CD8+ T cell responses<br />

because of their enhanced stability compared to<br />

conventional MHC molecules and can therefore be<br />

used for the expansion of peptide specific CD8+ T<br />

cells either in vitro or in vivo.<br />

We have expressed the human minor histocompatibility<br />

(H) antigens, HA-1 and HA-2 (expressed only<br />

on haematopoietic cells and restricted by human<br />

leukocyte antigen (HLA) A2) and the mouse HY<br />

minor transplantation antigens originating from<br />

the Smcy and Uty genes (restricted by H2-Db) in the<br />

SCT format. For the HLA-A2 SCTs, different forms<br />

were constructed by modifying the CD8 binding<br />

region (CD8 enhanced and non-binding variants)<br />

whereas in both mouse and human SCTs we introduced<br />

cysteine residues in the heavy chain and<br />

first linker that have been shown to form a disulfide<br />

bond, anchoring the peptide more efficiently<br />

(disulfide-trap (dt) variants).<br />

All human and mouse SCTs had good cell surface<br />

expression as determined by specific antibody<br />

staining of TAP2- deficient CHO cell transfectants.<br />

Soluble forms of HA-1 and HA-2 SCTs were obtained<br />

by retroviral transduction of human embryonic<br />

kidney (HEK) 293 T cells, and subsequent purification<br />

of the soluble protein from culture superna-<br />

tant. As the HA-1 and HA-2 minor H antigens are<br />

only expressed on normal and malignant haematopoietic<br />

cells, they are good candidates for therapeutic<br />

targeting. For this reason soluble SCTs were<br />

used for the in vitro expansion of allo-restricted<br />

CD8+ T cells specific for HA-1 and HA-2 minor H<br />

antigens from HLA-A2 negative individuals. Furthermore<br />

the minor male specific transplantation<br />

antigens DbUty and DbSmcy were expressed as<br />

SCTs and dtSCTs and will be introduced into bone<br />

marrow dendritic cells by retroviral transduction.<br />

Their potential to expand antigen specific CD8+<br />

T cells in vitro and in vivo as a vaccination model<br />

will be assessed. Further results will be presented<br />

illustrating the applications and functions of MHC<br />

class I SCTs.


041 Wittmann | Cellular therapy<br />

Comparing genetically engineered T cells for chimeric TCR<br />

versus CD16 + Trastuzumab for adoptive immunotherapy<br />

against HER2 positive breast carcinomas<br />

Sandrine Valsesia-Wittmann 1 , Béatrice Clémenceau 2 , Anne-Catherine Jallas 1 , Anne-Claire<br />

Doffin 3 , Jenny Valladeau 3 , Raphaël Rousseauy, Christophe Caux 1,3 and Henri Vié 2<br />

1 Centre Léon Bérard -plateforme d’Innovation en Immunomonitoring et Immunothérapie-<br />

28 rue Laennec-69008 LYON, France<br />

2 INSERM CRCNA U892 - Institut de Recherche Thérapeutique de l‘Université de Nantes-<br />

44007 NANTES, France<br />

3 CLB-INSERM U590, Cytokines et cancers, Lyon, France<br />

4 Roche, Bale, Switzerland<br />

The HER2 receptor is overexpressed in 25% of<br />

breast cancers and is associated with poor prognosis.<br />

Trastuzumab, a monoclonal antibody targeting<br />

HER2 has been demonstrated to improve survival<br />

of HER2 overexpressing metastatic breast cancer.<br />

However, majority of patients who initially respond<br />

to trastuzumab develop resistance within one year<br />

of treatment initiation, and in the adjuvant setting<br />

15% of patients still relapse despite trastuzumabbased<br />

therapy.<br />

The goal of this project is to develop and compare<br />

efficiency of two adoptive immunotherapy approaches<br />

by genetically engineered T cells to overcome<br />

this resistance. For the first approach, we developed<br />

T lymphocytes armed with an anti-HER2<br />

chimaeric TCR (scFvanti-Her2(FRP5)-CD28TM-<br />

CD3zeta) to directly kill HER2 positive cells. For<br />

the second approach, we developed T lymphocytes<br />

armed with a high affinity IgG FcR (FcgRIIIa, CD16)<br />

linked to its transduction chain FceRIg (CD16/g).<br />

In this latter case, the HER2 antigen is pre-targeted<br />

by trastuzumab to induce a “two step killing”<br />

through Antibodies Dependant Cellular Cytotoxicity<br />

(ADCC).<br />

Results obtained in vitro demonstrated the high<br />

direct killing efficiency of anti-HER2 chimaeric<br />

TCR CTL against HER2 amplified breast carcinoma<br />

cells BT474 or SKBR3 (>75%). However, abnormal<br />

reactivity of these CTL against undetectable HER2<br />

expression was observed (15 to 30% of lysis). This<br />

constitutive non specific activation of chimaeric<br />

TCR might represent an important limitation point<br />

for clinical use.<br />

On the other hand, “two step killing” with CD16/g-<br />

CTL demonstrated a low but very specific cytotoxicity<br />

against HER2 positive cells through ADCC<br />

only in the presence of Trastumumab (20% to<br />

40% of lysis at 30:1 ET). The moderate efficiency<br />

of specific lysis is linked to low levels of CD16/g<br />

expression and uncorrelated to level of HER2 targeted<br />

antigen expression. In order to improve our<br />

strategy and before transfert in animal model, we<br />

are developing new constructs with: (i) modification<br />

of the transduction domain of chimaeric TCR<br />

to block constitutive activation and limit non specific<br />

lysis and (ii) increase the efficiency of CD16/g<br />

expression at the surface of effectors to enhance<br />

ADCC mechanism.<br />

Furthermore, we are comparing in vivo efficacy<br />

of both approaches (“one step” TCR vs “two<br />

step”ADCC) to induce regression of HER2 + or -<br />

breast carcinoma xenograft in immunodeficient<br />

mice model NOD-SCID b2-/-.<br />

Our last results will be presented. Because the<br />

alteration of ADCC mechanisms during trastuzumab<br />

treatment is one rational explanation for<br />

the acquired resistance, improving effectors might<br />

represent a safe adjuvant treatment to prevent resistance.<br />

85


042 Weigand | Cellular therapy<br />

Generation of CD4+ T cells with specificity for FMNL1<br />

Luise Weigand 1 , Xiaoling Liang 1 , Ingrid Schuster 2 , Elfriede Eppinger 1 , Yanyan Han 1 ,<br />

Matthias Schiemann 3 , Elisabeth Kremmer 2 , Andreas Moosmann 4 , Josef Mautner 5 , Christian<br />

Peschel 1 , Angela Krackhardt 1<br />

1<br />

Department of Hematology/Oncology, Klinikum Rechts der Isar, Technische Universität München,<br />

Munich, Germany<br />

2<br />

Division Helmholtz Zentrum München – German Research Center for Environmental Health,<br />

Institute of Molecular Immunology, Munich, Germany<br />

3<br />

Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München,<br />

Munich, Germany<br />

4<br />

Helmholtz Zentrum München – German Research Center for Environmental Health, Clinical Cooperation<br />

Group Molecular Oncology and Ludwig-Maximilians-Universität München, Klinik für Hals-Nasen- und<br />

Ohrenkrankheiten, Munich, Germany<br />

5<br />

Helmholtz Zentrum München – German Research Center for Environmental Health, Institute for Clinical<br />

and Molecular Biology, Munich, Germany<br />

86<br />

Adoptive T cell therapies in the context of allogeneic<br />

stem cell transplantation have curative potential,<br />

especially in hematological malignancies. Patients<br />

with leukemic relapse after stem cell transplantation<br />

are treated with donor lymphocyte infusions<br />

which may induce a beneficial Graft-versus-leukemia<br />

effect but also detrimental Graft-versus-host<br />

disease. This approach may be improved by transferring<br />

T cells with selected specificities. We have<br />

previously generated allorestricted CD8+ T cells<br />

with specificity for the SEREX-derived tumor-associated<br />

antigen formin-like protein 1 (FMNL1). As<br />

it has been repeatedly shown, that CD4+ T cells<br />

play a critical role in tumor eradication and maintenance<br />

of immune memory, the aim of this project<br />

was to generate CD4+ T cells directed towards a<br />

peptide derived from FMNL1.<br />

We used two different methods to isolate FMNL1specific<br />

CD4+ T cells: Dendritic cells were pulsed<br />

with either one out of nine peptide pools from an<br />

FMNL1 peptide-library or recombinant protein to<br />

prime autologous PBMC or CD45RO- T cells. Using<br />

dendritic cells pulsed with overlapping peptides<br />

derived from the FMNL1 peptide library as stimu-<br />

lator cells, several T cell clones (e. g. Ga3.7) highly<br />

specific for the same FMNL1 peptide could be<br />

isolated. However, these clones did not recognize<br />

natural targets. In contrast, using protein-pulsed<br />

dendritic cells as stimulator cells, we were able to<br />

generate CD4+ T cell clones reactive against cells<br />

naturally expressing FMNL1. Intensive testing of<br />

one specific clone (Aa2.2) showed that recognition<br />

of target cells could be enhanced by transduction<br />

with FMNL1 and blocked by an HLA-DR specific<br />

antibody. Moreover preliminary data show, that<br />

Aa2.2 recognizes HLA-DR matched chronic lymphocytic<br />

leukemia cells. Both, the peptide-specific<br />

clone Ga3.7 and Aa2.2 show a T helper cell type 2<br />

(TH2) cytokine profile with antigen-specific secretion<br />

of IL-5, IL-10 and IL-13. TCR genes of relevant<br />

clones have been isolated and are, actually, transduced<br />

in indicator cell lines in order to confirm the<br />

MHC restriction element and to define the recognized<br />

epitope.


043 Liang | Cellular therapy<br />

Transfer of human T-cell receptor (TCR) containing murine<br />

chimeric constant betagamma-chain sequences reduces the risk<br />

of mixed heterodimers and enhanced the proliferation of<br />

transduced T cells<br />

Xiaoling Liang 1 , Yanyan Han 1 , Ingrid G. Schuster 2 , Luise U. Weigand 1 , Elfriede Eppinger 1 ,<br />

Dirk Busch 3 , Wolfgang Uckert 4 , Christian Peschel 1 , Angela Krackhardt 1<br />

1 Technical University Munich, Klinikum Rechts der Isar, Medizinische Klinik III,<br />

Innere Medizin mit Schwerpunkt Hämatologie und Onkologie, Munich, Germany<br />

2 Helmholtz Zentrum München-German Research Center for Environmental Health,<br />

Molecular Immunology, Munich, Germany<br />

3 Technical University Munich, Medical Microbiology, Immunology and Hygiene, Munich, Germany<br />

4 Max Delbrück Centrum, Berlin, Germany<br />

Adoptive transfer of tumor-associated antigen (TAA)<br />

or virus specific T-cell receptor (TCR)-transduced<br />

T cells may represent an attractive and promising<br />

approach to specifically treat malignant diseases<br />

and has been previously successfully applied in the<br />

clinic. However, there are different concerns which<br />

need to be addressed to further improve this therapeutic<br />

approach: First, the formation βγof heterodimers<br />

between endogenous and transduced TCR<br />

chains may abrogate specific TCR function and<br />

harbours a particular risk for unknown specificities.<br />

Second, longterm survival of TCR-transduced<br />

T cells has been demonstrated to be critical for the<br />

effectivity of this approach and needs to be further<br />

improved.<br />

We have previously identified several HLA-A2-allorestricted<br />

T-cell receptors with specificity for the<br />

breast cancer associated antigen HER2/neu and<br />

the hematopoietic lineage specific protein Forminlike<br />

protein 1 (FMNL1) which is overexpressed in<br />

diverse malignant cell lines and native leukemia<br />

cells. The FMNL1-specific TCR SK22 represents a<br />

weak TCR with low interchain affinity which was<br />

significantly improved by current optimization stra-<br />

tegies including murinization of constant chains<br />

and codon-optimization. We additionally modified<br />

this TCR as well as two other TCR with alternative<br />

specificities as HER2/neu and GP100 by introducing<br />

murinized constant chain betagamma (βγ)<br />

chimera, which results in reduced pairing of heterodimers<br />

in PBMC. More significantly, proliferation<br />

of T cells was clearly enhanced in effector cells and<br />

T cell clones transduced with αβγ compared to αβ<br />

TCR. Preliminary results suggest that also different<br />

cytokine patterns might be expressed. In contrast<br />

to a previous publication, we observed a significant<br />

polarization of CARMA-1 to the immunogical<br />

synapse in different TCR containing the chimeric<br />

chain combination and no decrease in Fas-Ligand<br />

expression in transduced T cells suggesting that<br />

altered CARMA-1 signaling might not be the only<br />

explanation for enhanced proliferation of T cells<br />

transduced or transgenic for the βγ-TCR.<br />

In conclusion, our data show that the transfer of<br />

TCR chain genes containing optimized murinized<br />

chimeric βγ-constant chains may have advantages<br />

and might be an alternative or synergistic choice<br />

for TCR optimization for adoptive T cell transfer.<br />

87


044 Kalos | Cellular therapy<br />

Clearance of established leukemia in a mouse xenograft model<br />

by a single injection of primary T cells gene-modified to express<br />

chimeric antigen receptors that target CD19<br />

Michael Kalos 1 , David M. Barrett 2 , Carmine Carpenito 1 , Stephan A. Grupp 2 and Carl June 1<br />

1 Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA<br />

2 Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA<br />

88<br />

We are evaluating the potential to use T-cells re-<br />

directed by genetic engineering to target human<br />

tumors. Chimeric antigen receptors (CAR), developed<br />

by combining antigen binding domains from<br />

antibodies or T cell receptors (TcR) to T cell signalling<br />

domains offer a potentially powerful platform<br />

to mediate tumor re-targeting.<br />

To re-target T cells against B cell malignancies we<br />

are focusing on the CD19 molecule, a membrane<br />

glycoprotein found on human B lymphocytes and<br />

expressed by the vast majority of B cell tumors. We<br />

have generated a CAR that consists of an extracellular<br />

scFv domain derived from an antibody that<br />

targets CD19 linked to the intracellular signalling<br />

domain of the TcR zeta chain. To criticaly evaluate<br />

the effects of including signaling domains from T cell<br />

costimulatory on CAR functionality and anti-CD19targeting<br />

CAR we introduced signalling domains<br />

from CD28 or CD137 (41BB) domains into the anti-<br />

CD19 CAR, transduced such CAR into primary<br />

human CD8+ and CD4+ T cells, and evaluated<br />

the ability of the transduced T cells to eradicate<br />

established aggressive CD19+ leukemia cell line<br />

Nalm-6 in the highly immunodeficient NOG mouse<br />

model. We evaluated primary lymphocytes gene<br />

modified to express anti-CD19-targeting CAR that<br />

contain TcRζ —alone, CD28-TcRζ, CD137-TcRζ—,<br />

and CD28-CD137-TcRζ — signaling domains.<br />

Mice treated with a single dose of T cells engineered<br />

to express each of the anti-CD19 CAR demonstrated<br />

statistically significant improved survival over con-<br />

trols animals. The CD19-CD28-CD137-ζ — CAR mediated<br />

the longest median overall survival and the<br />

most mice surviving until the end of the study at<br />

Day 180. In vivo bioluminescent imaging shows the<br />

kinetics of leukemia clearance to be rapid (within<br />

72 hours), and proves a sensitive tool for tracking<br />

disease relapse.<br />

These data demonstrate the potential of CAR directed<br />

to the CD19 molecule to effectively redirect T<br />

cells to eradicate established bone marrow disease<br />

with a single injection of CAR modified cells. Future<br />

efforts will focus on enhancing in vivo persistence<br />

and surveillance capabilities of these CAR T-cells,<br />

as well as investigating the ability of CAR T cells to<br />

clear repeated challenges of leukemia.


045 Grange | Cellular therapy<br />

Optimizing effector and memory properties of CD8 T cells<br />

for adoptive tumor immunotherapy<br />

Magali Grange 1 - 3 , Michel Buferne 1 - 3 , Lee Leserman 1 - 3 , Anne-Marie Schmitt-Verhulst 1 - 3 and<br />

Nathalie Auphan-Anezin 1 - 3<br />

Centre d'Immunologie de Marseille-Luminy, Parc Scientifique de Luminy, Case 906,<br />

13288 Marseille cedex 9, France.<br />

1<br />

INSERM U631, Parc Scientifique de Luminy, Case 906, 13288 Marseille, France<br />

2<br />

CNRS UMR6102, Parc Scientifique de Luminy, Case 906, 13288 Marseille, France<br />

3<br />

Université Aix-Marseille, Parc Scientifique de Luminy, Case 906, 13288 Marseille, France.<br />

Relative to normal tissues, tumor cells ectopically<br />

express or overexpress tumor-assoiated antigens<br />

(TAA). Therefore, targeted destruction of malignancies<br />

by enhancing CD8 T lymphocyte (TL) responses<br />

to TAA is an attractive therapeutic modality<br />

that is currently being evaluated. This protocol<br />

typically relies on the ex vivo expansion of TAAspecific<br />

CD8 effector TL in medium containing IL-2<br />

to support their survival and proliferation. While<br />

this allows the generation of large numbers of TAAspecific<br />

TL, it appears that these cells are terminally<br />

differentiated effector TL that rapidly become<br />

senescent. Therefore, alternatives to the use of IL-2<br />

need to be developed. Several groups including<br />

ours have shown that the IL-2R/STAT5 signaling<br />

reinforces the gene expression program initiated<br />

by TCR engagement in effector TL, including CD4<br />

Th2, CD4 Th1 and CD8 TL. For the latter subset<br />

we have shown that a constitutively active STAT5a<br />

transcription factor (STAT5CA) mimicked IL-2R signaling<br />

to enhance CD8 TL effector functions in<br />

vitro. Expression of the same STAT5CA mutant has<br />

been shown by others to promote the differentiation<br />

and the self renewal of hematopoietic stem<br />

cells, a cell population that shares a transcriptional<br />

signature with self renewing memory TL.<br />

We now report on the potential of STAT5CA expressing<br />

TAA-specific CD8 TL to gain both potent antitumor<br />

CD8 effector activity and long-term maintenance<br />

in vivo through the regulation of genes<br />

responsible for self-renewal.<br />

We show that expression of STAT5CA in CD8 TL<br />

favors their differentiation into long-lived effector<br />

cells which manifest the phenotypic and migratory<br />

properties (tissue homing) characteristic of effector<br />

TL and the high potential for Ag recall responses<br />

and absence of senescent features reminiscent of<br />

central memory TL. Nevertheless, the transduced<br />

CD8 TL return to quiescence and remain under<br />

control of TCR-derived signals for their survival<br />

and the activation of effector functions. Importantly,<br />

the characteristics conferred on TL by expression<br />

of STAT5CA were obtained with both monoclonal<br />

(TCR transgenic) and polyclonal CD8 TL. Therefore,<br />

active STAT5, by counteracting the induction of<br />

senescence, has distinct effects from IL-2 mediated<br />

signals that promote the differentiation of terminally<br />

differentiated effector TL. Finally, in adoptive<br />

therapy, STAT5CA-expressing CD8 effector TL demonstrated<br />

increased efficiency to cure mice with<br />

established tumors.<br />

Therefore, the novelty of our approach resides in the<br />

modification of a “master switch” gene as a useful<br />

strategy to optimize differentiation of CD8 TL into<br />

functional, long-lived and replication competent<br />

effector TL, through the concerted regulation of<br />

molecules involved in co-stimulation, memory fate,<br />

tissue infiltration, function and potential for secondary<br />

response.<br />

89


046 Singh | Cellular therapy<br />

Search for FLT3- ITD-reactive CD8+ T cells in the peripheral<br />

blood of healthy donors<br />

Vijay Singh 1 , Claudine Graf 1 , Volker Lennerz 1 , Catherine Wölfel 1 , Thomas Wölfel 1<br />

1<br />

III. Medizinische Klinik (Hematology & Oncology), University Medical Center of the<br />

Johannes Gutenberg-University, Mainz, Germany<br />

90<br />

FLT3-ITDs (fibroblast-macrophage stimulating<br />

factor receptor (FMS)-like tyrosine kinase receptor<br />

3 –internal tandem duplications) support leukemic<br />

transformation by constitutive phosphorylation<br />

resulting in uncontrolled activation and their presence<br />

is associated with worse prognosis. We have<br />

previously demonstrated that FLT3-ITD-derived neoantigens<br />

can be recognized by autologous AMLreactive<br />

CD8+ T cells (1, unpublished). We have<br />

chosen three immunogenic FLT3-ITDs identified in<br />

patients VE, IN and QQ, known to be restricted by<br />

HLA-B*27:05, -A*32:01 and -A*11:01, respectively.<br />

We set out to generate stable CD8+ T-cell populations<br />

and clones against these FLT3-ITDs from<br />

allogeneic healthy donors carrying at least one relevant<br />

FLT3-ITD-antigen presenting HLA I allele.<br />

In brief, blood-derived CD8+ T cells were stimulated<br />

under quasi-limiting dilution conditions with<br />

autologous irradiated CD8- PBMCs electroporated<br />

with in vitro-transcribed RNA encoding FLT3-ITD<br />

(2). On day 12, responder T cells were tested in<br />

an IFN-γ ELISPOT assay for recognition of the appropriate<br />

FLT3-ITD. ITD-reactive microcultures<br />

were restimulated twice with autologous irradiated<br />

CD8-PBMCs or adherent cells (monocytes)<br />

electroporated with FLT3-ITD mRNA. On day 26,<br />

to assess their HLA restriction, responder T cells<br />

were tested against COS-7 cells co-transfected with<br />

all donor HLA I alleles (predicted on the basis of<br />

2-digit typing) and the respective FLT3-ITD in an<br />

IFN-γELISPOT assay. Until now, we have analyzed<br />

16 healthy donors for FLT3-ITD response on d12.<br />

We observed ITD reactivity in seven donors. The<br />

frequencies of ITD-reactive T cells were estimated<br />

to be in the range of 4.25 x 10-7 to 2.5 x 10-6. In<br />

3 donors ITD-reactive T cells could be propagated<br />

over two further stimulation periods applying monocytes<br />

as presenting cells. In one donor we found<br />

that anti-FLT3-ITD (VE) T cells were restricted by<br />

HLA-Cw*07:02 whereas in the remaining donors<br />

the restricting HLA alleles were not identified. In<br />

further experiments we will try to derive stable<br />

T cell clones against FLT3-ITDs from allogeneic<br />

sources. These T cells or their receptors could be<br />

used in adoptive transfer therapies.<br />

(1) Graf C et al. Blood 109:2985-8, 2007;<br />

(2) Teufel R et al. Cell Mol Life Sci 62:1755-62, 2005


047 Blaudszun | Cellular therapy<br />

Adoptive transfer of ex vivo activated and antineoplastic drug<br />

loaded T lymphocytes retargeted to EpCAM expressing tumours<br />

André-René Blaudszun 1 , Anja Philippi 1 , Alice Borth 1 , Gerhard Moldenhauer 2 , Philipp Beckhove<br />

2 , Hyeck-Hee Lee 1 and Ute Steinfeld 1<br />

1 Cellular Immunotherapy Group, KIST Europe Forschungsgesellschaft mbH, Saarbrücken, Germany<br />

2 Translational Immunology Unit, German Cancer Research Center, Heidelberg, Germany<br />

T lymphocytes are often the first choice for adop-<br />

tive cell therapy (ACT) against cancers, because<br />

of their intrinsic cytolytic capacity. Unfortunately,<br />

clinical trials have shown that the effectiveness of<br />

ACT is limited. Among various therapeutic options,<br />

chemotherapy remains the mainstay of treatment<br />

for neoplasms. However, therapies based on antineoplastic<br />

drugs are often accompanied by severe<br />

adverse effects, because they are also harmful to<br />

healthy constantly dividing cells and tissues.<br />

Here, we describe a novel cancer therapy approach<br />

that combines the specificity of retargeted T cells<br />

with the high efficacy of chemotherapies. To enhance<br />

the natural cytotoxic function of human lymphocytes,<br />

ex vivo activated and expanded T cells were<br />

loaded with a liposomal preparation of the secondgeneration<br />

anthracycline idarubicin (LipoIda). Although<br />

T lymphocyte viability was impaired upon<br />

LipoIda treatment, mice experiments were carried<br />

out to test the effectiveness of the living drug carriers<br />

in vivo. When SKOV-3 xenograft mice were<br />

treated with either unloaded or idarubicin-loaded<br />

allogenic T cells in combination with the bispecific<br />

antibody EpCAM×CD3, tumour growth was more<br />

efficiently inhibited by the drug-containing cells.<br />

To analyse the observed effect in more detail, we<br />

compared the action of idarubicin-loaded T cells<br />

with the effect of a co-administration of unloaded<br />

T cells and free drug. Xenograft mice treated with<br />

drug-containing T lymphocytes showed a stronger<br />

inhibition of tumour growth than the control<br />

animals suggesting that the combined T cell-drug<br />

effect of LipoIda-treated T cells on malignant cell<br />

proliferation is rather synergistic than additive.<br />

Thus, redirection of antineoplastic drug-loaded<br />

T lymphocytes represents a promising course of<br />

action to treat human cancers. Since the administered<br />

idarubicin amount lies below conventional<br />

therapeutic doses, strong adverse effects typically<br />

caused by chemotherapy are unlikely to occur in<br />

our approach.<br />

91


048 Schendel | Cellular therapy<br />

High avidity T cell clones specific for tumor-associated<br />

antigens and how to find them<br />

Dolores J. Schendel 1,2 , Susanne Wilde, Daniel Sommermeyer 3 , Matthias Leisegang 3 , Stefani<br />

Spranger 1 , Slavoljub Milosevic 1 , Bernhard Frankenberger 1 and Wolfgang Uckert 3,4<br />

1<br />

Institute of Molecular Immunology and<br />

2<br />

Clinical Cooperation Group “Immune Monitoring”, Helmholtz Zentrum München, German Research Center for<br />

Environmental Health, Munich, Germany<br />

3<br />

Max-Delbrück-Center for Molecular Medicine, Berlin, Germany<br />

4 Humboldt University Berlin, Institute of Biology, Berlin, Germany<br />

92<br />

Since many tumor-associated antigens (TAA) re-<br />

present over-expressed self-proteins, T cells with<br />

high avidities have been eliminated during negative<br />

selection in the thymus to prevent autoimmunity.<br />

Therefore, we compared self-restricted and allo-restricted<br />

T cell lines and clones with the aim to identify<br />

properties that discriminate high avidity from<br />

low avidity T cells. Autologous CD8+ T cells were<br />

primed using either dendritic cells (DC) of HLA-A2donors<br />

pulsed with in vitro transcribed (ivt) RNA<br />

encoding a selected TAA plus allogeneic HLA-A2<br />

ivt-RNA or DC of HLA-A2+ donors loaded with<br />

TAA encoding ivt-RNA alone. Multimer+CD8+<br />

cells were sorted, cloned by limiting dilution and<br />

uncloned cells were expanded as bulk lines using<br />

antigen-independent stimulation.<br />

Since high-intensity multimer staining was shown<br />

previously to indicate strong TCR-ligand interactions,<br />

mean fluorescence intensities (MFI) of multimer<br />

binding was used as a first estimate of structural<br />

TCR-MHC/peptide binding affinity. A second estimate<br />

was made based on loss of multimer binding<br />

over time (i.e. multimer off-rate), since a slower<br />

off-rate suggests that TCR-ligand interactions are<br />

more stable and of higher structural affinity. Differences<br />

were seen directly in primed cultures and<br />

after sorting of bulk lines from various priming cultures.<br />

Multimer+CD8+ cells of HLA-A2+ donors<br />

had lower MFI of multimer binding compared to<br />

HLA-A2- donors and percentages of double-positive<br />

cells were also lower. A more rapid loss of<br />

multimer binding was measured over time. These<br />

characteristics of multimer binding indicated that<br />

self-restricted T cells were of lower TCR affinity<br />

compared to allo-restricted T cells. At the clonal<br />

level, we observed more disparities regarding the<br />

multimer analyses since there was a trend but no<br />

significant correlation between multimer analysis<br />

and functional avidity, as measured by responses<br />

to titrated amounts of TAA specific peptide pulsed<br />

onto T2 cells. These results revealed that multimer<br />

MFI and off-rates alone do not conclusively predict<br />

functional avidity of T cell clones. These discrepancies<br />

were retained when TCR from selected<br />

clones were expressed as transgenic proteins in<br />

activated lymphocytes.


049 Albrecht | Cellular therapy<br />

IL-21 treated naive CD45RA+ T cells represent a reliable source<br />

for generating AML-reactive cytotoxic T lymphocytes with high<br />

proliferative potential and early differentiation phenotype<br />

Jana Albrecht 1 , Michaela Frey 1 , Daniel Teschner 1 , Alexander Carbol 2 , Matthias Theobald 1 ,<br />

Wolfgang Herr 1 , Eva Distler 1<br />

1 Dept. of Medicine III, Hematology & Oncology, University Medical Center, Mainz, Germany<br />

2 Center for Blood Transfusion, University Medical Center, Mainz, Germany<br />

Specific allogeneic T cell therapy inducing selective<br />

and durable graft-versus-leukemia effects requires<br />

in vitro strategies to generate highly proliferating<br />

leukemia-reactive T cells with an early differentiation<br />

phenotype and strong effector functions.<br />

Here, we introduce an approach for the reliable<br />

generation of such leukemia-reactive cytotoxic T<br />

lymphocytes (CTLs) from naive CD45RA+ CD8+<br />

T cells of healthy donors. The protocol includes<br />

the stimulation of immunomagnetically isolated<br />

naive T cells with primary acute myeloid leukemia<br />

(AML) blasts of patient origin in HLA class I-matched<br />

mini-mixed lymphocyte/ leukemia cultures<br />

(mini-MLLCs), according to our recently published<br />

protocol (Distler et al., Exp. Hematol. 36:451-463,<br />

2008). In 8 different donor/AML-patient pairs we<br />

succesfully isolated AML-reactive CTLs from naive<br />

CD45RA+ precursors. If IL-21 was added during<br />

the primary stimulation phase in addition to the<br />

previously used cytokines IL-7, IL-12, and IL-15, the<br />

procedure was even more efficient. AML-reactive T<br />

cell cultures could be easily expanded by weekly<br />

antigen-specific stimulation to cell numbers exceeding<br />

108 within a few weeks. Most CTLs expressed<br />

one or up to three TCR Vβ chains, suggesting<br />

mono- or oligoclonality. For up to 10 weeks<br />

of in vitro culture the majority of CTL clones and<br />

lines retained strong HLA class-I restricted effector<br />

functions (cytolysis and IFN-γ secretion). On the<br />

basis of reactivity pattern with patient cells, CTLs<br />

could be divided into two groups: Those that exclu-<br />

sively reacted with primary AML blasts, suggesting<br />

leukemia-assosiated antigens as targets, and those<br />

that recognized both primary AML blasts and patient-derived<br />

lymphoblastoid cell lines (LCLs), indicating<br />

minor histocompatiliby antigens as targets.<br />

Donor-derived LCLs or the NK cell target K562 were<br />

not lysed. By the use of monoclonal anti-HLA class<br />

I-blocking antibodies and cross reactivity analyses<br />

against partially HLA class I-matched leukemia<br />

samples, we could identify HLA-A01, -A02, -B13,<br />

-B15, -B57, -Cw03, -Cw06 and -Cw07 as restriction<br />

elements. Most importantly, leukemia-reactive<br />

CTLs retained expression of the early T cell differentiation<br />

markers CD27, CD28 (costimulation),<br />

CD62L, CXCR4 (homing) and CD127 (IL7Rα) for<br />

several weeks during in vitro expansion. For some<br />

of these markers, the effect was even enhanced by<br />

IL-21. This expression pattern may be favorable for<br />

CTL survival and homing in vivo after adoptive<br />

transfer.<br />

In summary, AML-reactive CTLs generated with<br />

IL-21 from the naive CD8+ CD45RA+ subset of<br />

healthy donors are promising candidates for adoptive<br />

immunotherapy because of their expression of<br />

early phenotype markers along with a high proliferative<br />

potential and strong effector functions.<br />

93


050 Klobuch | Cellular therapy<br />

T cell receptor RNA transfer into non-reactive human<br />

T lymphocytes turns them into potent CMV-specific effector<br />

cells<br />

Sebastian Klobuch 1 , Katrin Besold 2 , Bodo Plachter 2 , Mirjam H. M. Heemskerk 3 , Niels<br />

Schaft 4 , Ralf-Holger Voss 1 , Matthias Theobald 1 , Wolfgang Herr 1 , Simone Thomas 1<br />

1 Department of Hematology & Oncology, University Medical Center, Mainz, Germany<br />

2<br />

Institute of Virology, University Medical Center, Mainz, Germany<br />

3<br />

Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands<br />

4 Department of Dermatology, University Hospital of Erlangen, Erlangen, Germany<br />

94<br />

Cytomeglovirus (CMV)-associated disease is a<br />

life-threatening complication in patients after allogeneic<br />

hematopoietic stem cell transplantation<br />

(HSCT). Although antiviral drug therapy is successfully<br />

used to reduce the risk of CMV disease,<br />

long-term virus control requires the reestablishment<br />

of protective antiviral T cell immunity in<br />

the host. The latter is challenging, particularly if<br />

the donor is CMV seronegative or possesses only<br />

a weak CMV-specific memory T cell response and<br />

thus, no CMV-reactive T cells are being transferred<br />

from donor to recipient during HSCT. Grafting<br />

nonreactive T cells of CMV seronegative donors by<br />

virusantigen specific T cell receptors (TCR) may be<br />

an efficient means to transfer CMV specific T cell<br />

function into allogeneic HSCT recipients. In this<br />

study, we have reprogrammed T cells of CMV-seronegative<br />

donors with human TCR recognizing the<br />

immunodominant HLA-A*0201- binding epitope<br />

495-503 derived from the CMV pp65 protein. To<br />

overcome the limitations of retroviral TCR gene<br />

transduction that hamper clinical translation, we<br />

used in vitro transcribed RNA encoding CMV-specific<br />

TCR for electroporation of non-reactive human<br />

T cells. After RNA transfection of anti-CD3 stimulated<br />

peripheral blood mononuclear cells (PBMCs),<br />

TCR expression levels were sufficient to trigger<br />

IFN-g secretion and cytolytic activity against pp65<br />

peptide-pulsed target cells and moreover against<br />

human fibroblast upon CMV infection. Due to the<br />

instability of the introduced RNA molecules, TCR<br />

expression and effector function was measureable<br />

for a time period of at least 3 days. We also observed<br />

that TCR RNA transfection of CD4+ T cells<br />

turned them into potent CMVpp65/HLA-A*0201specific<br />

T helper cells. This was demonstrated by<br />

co-incubating them with immature dendritic cells<br />

(DC), which resulted in maturation of DC only in<br />

the presence of the CMV pp65 epitope.<br />

Next, we transfected pure naïve and memory CD8+<br />

T cell subsets isolated from peripheral blood of<br />

CMV-seronegative donors. Although 90% of naïve<br />

CD8+ T cells were CMVpp65/HLA-A*0201 tetramer<br />

positive after electroporation, they mediated only<br />

marginal lysis toward CMV-infected fibroblasts.<br />

In contrast, memory CD8+ T cells showed strong<br />

TCR expression and cytotoxicity upon antigen recognition<br />

up to one week. In summary, our data demonstrate<br />

that non-reactive human T cells can be<br />

easily redirected with CMVpp65 TCR RNA, thereby<br />

gaining CMV-specific T cell effector function for a<br />

considerable time period.<br />

We therefore believe that CMVpp65 TCR RNA has<br />

the potential to be further developed as a therapeutic<br />

‘off-the-shelf’ reagent for CMV-seropositive<br />

patients who undergo allogeneic HSCT from CMVseronegative<br />

donors.


051 Distler | Cellular therapy<br />

Alloreactivity to HLA class I mismatch alleles mainly resides in<br />

CCR7+ naive and central memory CD8 T cells<br />

Eva Distler 1 , Saliha Asdufan 1 , Michaela Frey 1 , Udo F. Hartwig 1 , Matthias Theobald 1 , Wolfgang<br />

Herr 1<br />

1 Dept. of Medicine III, Hematology & Oncology, University Medical Center, Mainz, Germany<br />

Reducing the incidence of severe graft-versus-host<br />

disease (GVHD) remains the major challenge in<br />

patients who receive stem cell transplants from<br />

HLA-mismatch donors. Selective depletion of alloreactive<br />

donor T cells can prevent GVHD, but is<br />

still a complex technical procedure that requires<br />

the in vitro culture of T cells for several days. Here,<br />

we studied CD8 T cell subsets of 5 healthy donors,<br />

sorted by flow cytometry according to expression<br />

of the differentiation markers CD45RA, CD45RO,<br />

CCR7 and CD62L for the ability to respond to allo-<br />

HLA class I alleles in vitro. To detect pure alloreactive<br />

T cell responses and minimize the interference<br />

by other specificities, we used HLA-deficient K562<br />

cells transfected with single HLA-A, -B or -C alleles<br />

for weekly stimulations. We observed the strongest<br />

anti-HLA mismatch proliferation in sorted CCR7pos<br />

and CD62Lpos fractions, comprising naive (TN)<br />

and central memory CD8 T cells (TCM). In contrast,<br />

the CCR7neg and CD62Lneg counterpart fractions<br />

containing effector memory (TEM) and terminal<br />

effector CD8 T cells showed much lower allo-HLA<br />

induced in vitro expansion. The CD45RApos subset<br />

enriched for TN cells and devoid of TCM and TEM<br />

cells demonstrated less strong allo-proliferation<br />

compared to corresponding CD45RAneg cells. Low<br />

allo-proliferation was observed for CD45ROpos and<br />

CD45ROneg populations. Interestingly, the growth<br />

pattern of CD8 T cell subsets was consistent, regardless<br />

of HLA-A, -B, or -C mismatch alleles were<br />

used for stimulation. Cultures derived from all CD8<br />

T cell subsets were analyzed for allo-HLA class I reactivity<br />

by IFN-γ ELISPOT assay starting on day 11.<br />

The strongest anti-HLA mismatch reactivity was<br />

detected in the CCR7pos subset. Lower level of alloreactivity<br />

was observed in CD62Lpos, CD45ROneg,<br />

and CD45RApos fractions. All counterpart fractions<br />

showed even less strong anti-HLA responses.<br />

Again, these findings were consistent for all HLA<br />

alleles tested. Our results thus demonstrate that distinct<br />

CD8 T cell subsets vary substantially in the<br />

ability to respond to HLA class I mismatch alleles.<br />

Although all subsets enriched for TN and TCM<br />

produced substantial alloreactivity, the CCR7pos<br />

fraction exceeded others in terms of proliferation<br />

and effector function. Thus, in vitro removal of<br />

CCR7pos CD8 T cells may be a straightforward culture-independent<br />

allodepletion procedure. Further<br />

experiments comparing anti-HLA mismatch and<br />

self-HLA-restricted anti-leukemia reactivity of T<br />

cell subsets are ongoing.<br />

95


052 Salguero | Cellular therapy<br />

Dramatic early expansion of CMV-reactive human T cells in<br />

vivo is supported by lentiviral vector-induced engineered dendritic<br />

cells<br />

Gustavo Salguero 1 , Bala Sai Sundarasetty 1 , Dirk Wedekind 2 , Sylvia Borchers 1 , Gregor<br />

Warnecke 3 , Ann-Kathrin Knöfel 3 , Rainer Blasczyk 4 , Britta Eis-Vesper 4 , Eva Mischak-Weissinger<br />

1 , Arnold Ganser 1 , Renata Stripecke 1<br />

1<br />

Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation,<br />

Hannover Medical School, Hannover, Germany<br />

2<br />

Central Animal Laboratory, Hannover Medical School, Hannover, Germany<br />

3 Department of Experimental Transplantation, Hannover Medical School, Hannover, Germany<br />

4 Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany<br />

96<br />

Dendritic cell (DC) based immunotherapy is a<br />

potent strategy to induce antigen-specific responses<br />

against cancer and infections. Strategies<br />

to enhance the viability, biodistribution and immunostimulatoy<br />

capacity of ex vivo generated DC in<br />

vivo, may also be critical to determine the success<br />

of adoptive T cell therapies. We have previously<br />

shown that genetically reprogramming of human<br />

monocytes using bicistronic lentiviral vectors (LV)<br />

expressing GM-CSF and IL-4 induces self-differentiated<br />

DCs (SMART-DC) with improved engraftment,<br />

viability and biodistribution in vivo (Sundarasetty<br />

et al., <strong>CIMT</strong> 2009). Here, we assessed the in vivo<br />

potential of SMART-DCs to support engraftment<br />

and expansion of human T cells reactive against<br />

cytomegalovirus (CMV). Human SMART-DCs were<br />

generated from CD14+ monocytes obtained from<br />

CMV-seropositive donors and co-transduced with<br />

a LV expressing the full-length CMV protein pp65<br />

(SMART-DC/pp65) for analyses of antigen-specific<br />

responses. SMART-DC/pp65 showed a stable DC<br />

immunophenotype in vitro (CD209+, HLA-DR+,<br />

CD86+) and high viability in vitro (>3 weeks) and<br />

in vivo (>6 weeks). NOD.Rag1-/-.IL2rγ-/- immunodeficient<br />

mice were preconditioned with SMART-<br />

DC or with SMART-DC/pp65 injected s.c. and 7<br />

days later infused i.v. with autologous T cell expressing<br />

the firefly luciferase (fLUC). The engraftment,<br />

expansion and biodistribution of T cells were<br />

evaluated by serial in vivo bioluminescence image<br />

analysis, flow cytometry and immunohistochemi-<br />

stry. Control arms with “conventional” DCs alone<br />

or pulsed with pp65 overlapping peptides were included.<br />

SMART-DC-preconditioned mice showed<br />

significantly enhanced engraftment of fLUC-T cells<br />

in spleen (day 14, 6-fold p


053 Stolle | Cellular therapy<br />

Generation of HCMV-/TAA-bispecific human T cells by<br />

either the genetic equipment of HCMV+ T cells with<br />

tumor-reactive TCRs or the combined retroviral transduction<br />

of bulk human T cells with HCMV-/TAA-specific TCRs<br />

Diana Stolle 1 , Beate Hauptrock 1 , Hakim Echchannaoui 1 , Edite Antunes 1 , Simone Thomas 1 ,<br />

Sebastian Klobuch 1 , Wolfgang Herr 1 , Matthias Theobald 1 , Ralf-Holger Voss 1<br />

1 Department of Hematology & Oncology, University Medical Center, Mainz, Germany<br />

Immune suppression after allogeneic stem cell<br />

transplantation causes reactivation of human cytomegalovirus<br />

(HCMV) and comes along with increased<br />

mortality in HCMV positive patients. The<br />

aim of cellular immunotherapy is the eradication<br />

of tumor cells and the decrease of the HCMV viral<br />

load by adoptive transfer of HCMV-/tumor associated<br />

antigen (TAA)-bispecific T lymphocytes.<br />

Therefore, we performed two different strategies:<br />

First, we generated HCMV-reactive (HCMV+) T<br />

cells from seropositive human blood donors using<br />

pp65(NLVPMVATV(495-503)) peptide-specific stimulation.<br />

In this case one or two restimulations<br />

led to the efficient accumulation of up to 80 %<br />

HCMV-specific T cells. To obtain a pure HCMV T<br />

cell culture we sorted the cells with CMV pp65specific<br />

streptamers by flow cytometry. Cytotoxicity<br />

was assessed in a 51-Chromium release assay<br />

using either pp65- loaded K562-A2 or HCMV-infected<br />

HLA-A*0201+ human fibroblasts as target<br />

cells. For distinct HCMV+ donors, minimal pp65<br />

peptide concentrations were in the range of as low<br />

as 0.03 nM to 100 nM triggering specific lysis at<br />

CD8+ HCMV+ to target ratios of 5:1 up to 20:1.<br />

They were indicative of high TCR affinities for the<br />

cognate antigen reflected by EC50-values ranging<br />

from 0.1 nM to 1.8 nM in peptide titration. Most importantly,<br />

HCMV T cells were efficiently capable of<br />

lysing CMV-infected human fibroblasts. After their<br />

antigen-specific expansion, HCMV-reactive T cells<br />

were equipped with a p53- or gp100- tumor anti-<br />

genspecific TCR by either RNA transfection or retroviral<br />

transduction. RNA electroporation yielded<br />

the generation of p53- as well as gp100- bispecific<br />

HCMV+ T cells: Both subsets proved efficient<br />

bifunctionality in CTL-assay and IFNg-Elisa after<br />

endogenous (pp65)- or exogenous (p53/gp100)<br />

peptide-specific stimulation. Preliminary data indicated<br />

that in retroviral transduction either TCR,<br />

the endogenous TCR CMV or the introduced TCR,<br />

are prone to be down-regulated dependent on the<br />

particular HCMV/TAA TCR combination.<br />

Our second approach was the simultaneous retroviral<br />

transduction of bulk human T cells with a<br />

HCMV- as well as p53 tumor antigen- specific TCR.<br />

Both retroviral TCR constructs contained drug selection<br />

cassettes which allowed for a normalization<br />

of TCR expression. In tetramer analysis, we<br />

were able to detect both, single TCR+ T cells and a<br />

substantial fraction of HCMV/TAA double TCR+ T<br />

cells. Endogenous TCRs were still present. Bifunctionality<br />

was corroborated in CTL-assays and in intracellular<br />

IFNg secretion stainings for specifically<br />

restimulated T cells.<br />

In summary, our data demonstrate two opportunities<br />

to generate virus (HCMV) and tumor antigen<br />

bispecific human T cells. Most importantly, the<br />

expression level of the HCMV- as well as tumor<br />

antigen- specific TCR is sufficient for specific<br />

antigen recognition.<br />

97


054 Bloetz | Cellular therapy<br />

Allo-reactivity to HLA class II mismatch alleles in CD4+<br />

T-cell subsets<br />

Andrea Blötz 1 , Eva Distler 1 , Elke Schnürer 1 , Simone Thomas 1 , Ugur Sahin 1 , Matthias Theobald<br />

1 , Wolfgang Herr 1<br />

1 Dept. of Medicine III – Hematology & Oncology, University Medical Center, Mainz, Germany<br />

98<br />

In allogeneic hematopoietic stem cell transplan-<br />

tation (allo-HSCT), allo-reactive T lymphocytes<br />

of donor origin recognize antigens expressed on<br />

patient-derived leukemia cells, thereby mediating<br />

the beneficial graft-versus-leukemia effect. If alloantigens<br />

are expressed on non-hematopoietic recipient<br />

tissues, donor T cells also induce graft-versushost<br />

disease (GvHD). Since HLA mismatch alleles<br />

represent major targets of allo-reactive T lymphocytes,<br />

patient and donor are usually matched for<br />

the class I molecules HLA-A, -B, -C, and for the<br />

class II molecules HLA-DRB1 and -DQB1, in order<br />

do reduce the risk of GvHD. The HLA-DPB1 locus,<br />

however, is still ignored in donor selection. Interestingly,<br />

clinical studies have demonstrated that<br />

disparities at HLA-DQB1 and distinct HLA-DPB1<br />

alleles do not adversely affect the outcome of allo-<br />

HSCT. Because HLA class II is predominantly expressed<br />

on hematopoietic cells, allo-reactive CD4+<br />

donor T cells recognizing HLA-DQB1 or permissive<br />

HLA-DPB1 mismatch alleles may primarily target<br />

patient leukemic and hematopoietic cells, while<br />

sparing non-hematopoietic recipient tissues. In this<br />

ongoing study we analyze the allo-reactive potential<br />

of CD4+ T-lymphocyte subsets by stimulating<br />

them in vitro against single HLA class II mismatch<br />

alleles. For that purpose, CD4+ peripheral blood T<br />

lymphocytes of healthy donors are sorted by flow<br />

cytometry according to the expression or absence<br />

of the differentiation markers CD45RA, CD45RO,<br />

CD62L and CCR7. As standard antigen-presenting<br />

cells for class II-mismatch stimulations, we use the<br />

HLA-deficient cell line K562 upon electroporation<br />

with in vitro transcribed RNA coding for the alpha<br />

and beta chains of single HLA class II molecules<br />

(HLA-DQ: DQA1-010201/DQB1-060201; HLA-DR:<br />

DRA1-0101/DRB1-0701). This procedure results in<br />

transient HLA expression for up to one week with<br />

strongest staining intensity (>80% positive cells)<br />

24h after electroporation. In addition, transfectants<br />

do not lose HLA class II expression upon freezing,<br />

thawing and irradiation cycles. In a first series of<br />

experiments, sorted CD4 T-cell subsets of 3 healthy<br />

donors were stimulated weekly with mismatched<br />

K562/HLA-DR or -DQ transfectants, respectively,<br />

and were tested for allo-reactivity 5 days after the<br />

first (d12) and second (d19) restimulation in IFN-γ<br />

ELISPOT assays. The strongest allo-HLA II reactivity<br />

was detected in the CD45RApos and CD62Lpos<br />

as well as in the CD45ROneg CD4+ subsets, in contrast<br />

to the corresponding counterpart fractions,<br />

confirming that allo-reactivity can be found mainly<br />

in naive and central memory T cells. Alloreactivity<br />

of the CCR7pos and CCR7neg cell fractions varied<br />

considerably. In all cases, anti-HLA class II reactivity<br />

of CD4 populations could be blocked by monoclonal<br />

antibodies to the respective HLA class II<br />

molecule used for stimulation. Proliferation did not<br />

show clear differences between individual CD4+<br />

T-cell subsets. In summary, we show herein that<br />

the transient expression of HLA class II molecules<br />

in K562 cells by RNA electroporation is a rapid and<br />

efficient approach to detect and stimulate allo-HLA<br />

class II reactive CD4+ T-cell responses. The strongest<br />

class II mismatch reactivity was found in the<br />

naive and central memory CD4+ T-cell subsets.<br />

This approach based on K562 cells and HLA class<br />

II RNA as „off-the-shelf” reagents allows to rapidly<br />

generate CD4+ T cells with reactivity to single allo-<br />

HLA-DQ/DP alleles, which may be of potential use<br />

in adoptive immunotherapy of leukemias.


055 Stumpf | Cellular therapy<br />

Indirect presentation of HLA class II restricted minor histocompatibility<br />

antigens is a characteristic of HLA-DM resistant<br />

antigens<br />

Anita Stumpf 1 , Edith van der Meijden 1 , Roel Willemze 1 , Fred Falkenburg 1 , Marieke Griffioen 1<br />

1 Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands<br />

HLA-matched allogeneic stem cell transplantation<br />

is an effective treatment for haematological malignancies.<br />

The anti-tumor effect is mediated by donor<br />

T-cells recognizing polymorphic peptides in HLA<br />

molecules which are differentially expressed on<br />

patient and donor cells due to amino acid differences<br />

encoded by single nucleotide polymorphisms,<br />

the so-called minor histocompatibility antigens<br />

(MiHAs). Donor T-cells recognizing MiHAs<br />

with broad expression on hematopoietic as well as<br />

non-hematopoietic cells may not only induce beneficial<br />

antitumor immunity, but also detrimental<br />

graft versus host disease (GvHD). We recently identified<br />

5 HLA class II restricted MiHAs which were<br />

not directly recognized on nonhematopoietic cells<br />

even after HLA class II upregulation by cytokine<br />

treatment. However, it was recently shown in mice<br />

(Wang, ASH 2009, abstract no. 689) that indirect<br />

presentation of host antigens by donor antigen presenting<br />

cells (APC) contributed to CD4+ T cell mediated<br />

GvHD. Here, we analyzed whether this is a<br />

general phenomenon by analyzing processing and<br />

intercellular transfer of six HLA class II restricted<br />

antigens.<br />

As a model to investigate recognition by CD4+ T<br />

cells, we exogenously pulsed the nonhematopoietic<br />

HeLa cell line transduced with the respective HLA<br />

class II alleles with bacteria expressing six different<br />

recombinant proteins. Antigen processing was<br />

analyzed after retroviral transfer of invariant chain<br />

(Ii) with or without HLA-DM. Intercellular transfer<br />

of antigens was analyzed by co-incubation of HLA<br />

II+/MiHA- cells with HLA II-/MiHA+ cells or their<br />

culture supernatants.<br />

Recognition of all HLA class II restricted antigens<br />

was increased upon transduction of Ii. Additional<br />

transfer of HLA-DM, however, had divergent effects.<br />

Following HLA-DM transfer, 3 antigens were similarly<br />

recognized (DM-resistant), whereas recognition<br />

of the other 3 antigens was completely abolished<br />

(DM-sensitive). We observed intercellular transfer<br />

of DM-resistant, but not DM-sensitive, antigens to<br />

MiHA- /HLA II+ cells after co-incubation with<br />

MiHA+/HLA II- cells. Also in the absence of direct<br />

cellcell contact the antigen could be transferred by<br />

cell-derived particles as was shown by incubation<br />

with supernatant from MiHA+ cells. This transfer<br />

was not mediated by free peptides as 30kDa filtration<br />

of the supernatant abolished recognition.<br />

In summary, we show that some antigens can be<br />

intercellularly transferred leading to T cell recognition<br />

of MiHA-/HLA II+ cells. However, this phenomenon<br />

was restricted to a group of antigens that<br />

showed HLA-DM-resistant processing and presentation.<br />

These data suggest that late after allogeneic<br />

stem cell transplantation, when patient-derived<br />

APCs are replaced by the donor, only HLA-DMresistant<br />

antigens may be indirectly presented by<br />

donor APC, and therefore not every MiHA mismatch<br />

may evoke late onset GvHD.<br />

99


056 Hoyer | Cellular therapy<br />

Examination of T-helper cell / DC cross-talk by the transfection<br />

of T cells with RNA coding for TCRs<br />

Stefanie Hoyer 1 , Sabrina Schievelbein 1 , Gerold Schuler 1 , Jan Dörrie 1 ,*, and Niels Schaft 1 ,*<br />

1 Department of Dermatology, University Hospital Erlangen, Erlangen, Germany<br />

* Contributed equally<br />

100<br />

The goal of immunotherapy of cancer is to initiate<br />

an adaptive immune response against the tumor.<br />

But up to now, cancer immunotherapy has mainly<br />

focused on the generation of tumor-specific CD8+<br />

T cells, even though CD4+ T-cell help is also required<br />

for a long-lasting cytotoxic T lymphocyte<br />

response. However, the exact mechanism by<br />

which CD4+ T cells license dendritic cells (DC)<br />

and thereby modulate the priming and expansion<br />

of CTLs is not yet fully understood. For this<br />

purpose, we transferred melanoma-antigen-specific<br />

TCRs by RNA-electroporation into CD4+ T cells<br />

and co-cultured them with peptide-loaded DC to<br />

elucidate T-cell / DC cross-talk. We either introduced<br />

MAGE-A3/HLA-DP4-specific or gp100/HLA-<br />

A2-specific TCR-RNA into CD4+ T cells, which<br />

mainly resulted in antigen-specific Th1 cytokine<br />

secretion after co-cultivation with peptide-loaded<br />

DC. Besides, phenotypic changes were assessed in<br />

a time-dependent manner. We detected a clear antigen-specific<br />

up-regulation of maturation markers<br />

like CD25, CD40, CD80, CD86, and CD70 on both immature<br />

and mature DC after 20 hours of co-cultivation<br />

with T cells. In correlation, CD25 was antigenspecifically<br />

up-regulated as an activation marker<br />

on CD4+ T cells. The early activation marker CD69<br />

was antigen-specifically up-regulated already after<br />

2 hours, while CD27- and CCR7-expression on the<br />

T cells remained almost constant during the stimulation.<br />

Moreover, T-cell activation was completely<br />

cell-cell-contact dependent, while the maturation<br />

of the DC was in part mediated by soluble factors,<br />

as revealed by transwell experiments. These data<br />

indicate that DC / CD4+ T-cell cross-talk is a bidirectional<br />

process. Moreover, we currently examine<br />

the influence of pre-activation of CD4+ T cells on<br />

the antigen-specific cross-talk, and whether there<br />

is an antigen-specific cross-talk between DC and<br />

CD8+ T cells. Since TCR-transfected CD4+ T cells<br />

can induce DC maturation, they may be used to<br />

provide T-cell help to induce more efficient CD8+<br />

T-cell responses for the immunotherapy of cancer.


057 Foerster | Cellular therapy<br />

Generation of multivirus-specific CD4+ and CD8+ T cells<br />

for adoptive immunotherapy<br />

Anna Foerster, Verena Lasmanowicz, Olaf Brauns, Sven Kramer, Petra Jekow, Wolfgang Rönspeck,<br />

Jürgen Schmitz, Mario Assenmacher, Anne Richter<br />

Department of Research & Development, Miltenyi Biotec GmbH, Bergisch Gladbach, Germany<br />

Adoptive transfer of T lymphocyte populations<br />

with specificities for several antigens is an attractive<br />

strategy for the treatment of multiple infections<br />

in immunocompromised patients or of cancer. We<br />

have established a protocol for the generation of<br />

multiantigen-specific CD4+ and CD8+ T cells<br />

using newly developed pools of peptides for in vitro<br />

T cell stimulation and a subsequent magnetic enrichment<br />

of functional T cells according to IFN-γ<br />

secretion.Peptide pools consist of synthetic peptides<br />

of mainly 15 amino acid (aa) length with 11<br />

aa overlap covering the complete antigenic protein.<br />

We have analyzed the efficiency of peptide pools<br />

compared to recombinant proteins and immunodominant<br />

HLA-A2 and –B7-restricted peptides for<br />

short-term in vitro restimulation and induction of<br />

IFN-γ in cytomegalovirus (CMV) pp65 and IE-1 specific<br />

CD4+ and CD8+ T cells derived from CMVinfected<br />

healthy blood donors.<br />

The results show that the peptide pools as well as<br />

the recombinant proteins are useful for efficient<br />

activation of CD4+ T helper cells. In contrast, efficient<br />

stimulation of CD8+ T cells is achieved only<br />

using either the overlapping 15-mer peptide pools<br />

or the immunodominant peptide epitopes of 8-10<br />

amino acids. The latter are well defined only for<br />

a limited panel of antigens and are restricted to<br />

certain HLA alleles.<br />

To test the usability of peptide pools to generate<br />

multivirus-specific T cells for adoptive immunotherapy<br />

we stimulated PBMC from leukapheresis of<br />

healthy donors with a combination of four peptide<br />

pools selected from CMV pp65 and IE-1, adenovirus<br />

(AdV) hexon, and Epstein-Barr-Virus (EBV) EBNA-1<br />

and BZLF-1 for four hours. The Large Scale IFN-γ<br />

Secretion Assay Enrichment Kit was used to magnetically<br />

enrich IFN-γ secreting T cells to a purity of<br />

>90%. Antigen specificity and functionality of the<br />

enriched T cells were controlled after expansion.<br />

Cells expanded between 4 and 745 fold within 9-14<br />

days. Expanded cells contained high frequencies<br />

of pp65495-503/A2 tetramer+ and pp65417-426/B7<br />

tetramer+ CD8+ T cells. Additionally after restimulation<br />

of the expanded cells with the mixture<br />

of peptide pools and intracellular IFN-γ staining,<br />

21-53% of CD4+ T cells and 53-87% of CD8+ T<br />

cells produced IFN-γ. Moreover comparing the stimulation<br />

of PBMC with reactivation of the T cell<br />

lines with each peptide pool separately showed that<br />

the specificity for each antigen sustained during<br />

the enrichment and expansion phase.<br />

The concomitant addition of four peptide pools increases<br />

the competition of peptides to be loaded on<br />

MHC molecules, which might decrease the efficient<br />

activation of each antigen-specific T cell. Therefore<br />

we included in our study the separation of PBMC<br />

in four samples, separate loading of the samples<br />

with one peptide pool for two hours, and recombination<br />

of PBMC for T cell stimulation for four<br />

hours. We found comparable results in the number<br />

of enriched cells, expansion rate of T cells, and the<br />

antigen-specificity of the T cell lines for concomitant<br />

and separate antigen loading.<br />

In summary, we have established a protocol for<br />

rapid in vitro generation of multivirus-specific<br />

CD4+ and CD8+ T cells using a combination of<br />

peptide pools from several antigens for restimulation<br />

and subsequent magnetic selection of IFN-γ<br />

secreting T cells.<br />

101


058 Echchannaoui | Cellular therapy<br />

Evaluation of the safety of p53 TCR gene transfer in a<br />

humanized mouse model<br />

Hakim Echchannaoui 1 , Edite Antunes 1 , Carina Lotz 2 , Simone Thomas 1 , Ralf-Holger Voss 1<br />

and Matthias Theobald 1<br />

1 Department of Hematology & Oncology, University Medical Center, Mainz, Germany<br />

2 Actelion Pharmaceuticals Ltd, Allschwil, Switzerland<br />

102<br />

Adoptive cell therapy with T cells retrovirally trans-<br />

duced with tumor-associated antigen (TAA) specific<br />

TCRs is a promising approach for immunotherapy<br />

in patients with haematological malignancies. The<br />

TAA p53 is over-expressed in approximately 50% of<br />

human tumors. We have reported that HLA-A*0201<br />

(A2.1) transgenic mice can be used to circumvent<br />

self-tolerance to universal human TAA and to generate<br />

efficient tumor-reactive CTL. We used A2.1<br />

transgenic mice, in which the mouse CD8 molecule<br />

cannot efficiently interact with A2.1 to generate a<br />

high-affinity, CD8-independent p53(264-272) specific<br />

TCR. Retroviral expression of CD8-independent<br />

p53-specific TCR into T cells, allowed CD8+ T lymphocytes<br />

to acquire a broad tumor-specific CTL activity<br />

but also redirected CD4+ T cells into potent<br />

tumor-reactive, p53-specific T helper cells. However<br />

a particular safety concern with TCR gene transfer,<br />

is the formation of mixed TCR heterodimers<br />

between the introduced TCR α and β chains with<br />

the endogenous TCR chains, resulting in the potential<br />

generation of autoreactive T cells. To reduce<br />

the formation of TCR mixed dimers an additional<br />

inter-chain disulfide bond between the TCR α and β<br />

chain constant domains was introduced. We further<br />

improved the expression level of p53 TCR transgene<br />

using codon-optimization of the TCR sequence.<br />

Mouse T cells transduced with cysteine-modified<br />

(Cys) and codon-optimized (Opt) p53(264-272)A2.1<br />

TCR showed higher expression levels of the introduced<br />

TCR as compared to p53(264-272)-specific<br />

WT TCR. Importantly, p53(264-272)A2.1-Opt.Cys<br />

TCR transduced CTL recognized and killed a wide<br />

variety of malignant A2.1 tumor cells with altered<br />

p53 expression, but not p53-deficient A2.1 cells<br />

more efficiently as compared to CTL transduced<br />

with the WT p53(264-272)-specific TCR.<br />

However, when p53(264-272)A2.1-Opt.Cys TCR<br />

transduced mouse T cells are adoptively transferred<br />

into p53-deficient partially humanized (A2Kb)<br />

mice, under conditioning-induced lymphopenia,<br />

expansion of infused T cells following high dose<br />

IL-2 administration is associated with the development<br />

of lethal autoimmunity similarly to mice<br />

receiving p53(264-272)-specific WT TCR transduced<br />

T cells due to the formation of self-reactive<br />

TCRs. The so called off-target toxicity observed in<br />

our preclinical mouse model highlights the need<br />

for further improvement of TCR gene therapy to<br />

prevent TCR mispairing-induced autoimmunity. To<br />

address this concern, a novel single chain p53TCR<br />

construct was engineered and is currently under<br />

investigation.


059 Bourquin | Cellular therapy<br />

Efficient eradication of subcutaneous but not of autochthonous<br />

gastric tumors by adoptive T cell transfer in a SV40 T antigen<br />

mouse model<br />

Carole Bourquin 1 , Philip von der Borch 1 , Christine Zoglmeier 1 , David Anz 1 , Nadja Sandholzer<br />

1 , Cornelia Wurzenberger 1 , Robert Kammerer 2,3 , Wolfgang Zimmermann 2 , Stefan Endres 1<br />

1 Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology,<br />

University Hospital of Munich, 80336 Munich, Germany<br />

2 Tumor Immunology Laboratory, LIFE-Center, Grosshadern, Ludwig-Maximilian University,<br />

Marchioninistr. 23, 81377 Munich, Germany<br />

3 Institute of Immunology, Friedrich-Loeffler-Institut, Paul-Ehrlich-Straße 28, 72076 Tuebingen, Germany<br />

In stomach cancer, there is a need for new thera-<br />

peutic strategies, in particular for the treatment of<br />

unresectable tumors and micrometastases. We investigated<br />

the efficacy of immunotherapy in an autochthonous<br />

model of gastric cancer, the CEA424-<br />

SV40 T antigen (TAg) transgenic mice. Treatment<br />

efficacy against both the autochthonous tumors<br />

and subcutaneous tumors induced by the derived<br />

cell line mGC3 were assessed. In wild-type mice,<br />

a dendritic cell vaccine loaded with irradiated<br />

tumor cells combined with CpG oligonucleotides<br />

induced efficient cytotoxic T cell and memory responses<br />

against mGC3 subcutaneous tumors. In<br />

contrast, neither subcutaneous nor autochthonous<br />

tumors responded to vaccination in CEA424-SV40<br />

TAg mice, indicating tolerance to the SV40 TAg.<br />

To examine whether tumors in these mice were<br />

principally accessible to immunotherapy, splenocytes<br />

from immune wild-type mice were adoptively<br />

transferred into CEA424-SV40 Tag transgenic mice.<br />

Treated mice showed complete regression of the<br />

subcutaneous tumors associated with intratumoral<br />

infiltrates of CD8 and CD4 T cells. In contrast,<br />

the autochthonous gastric tumors in the same mice<br />

were poorly infiltrated and did not regress.<br />

Thus, even in the presence of an active antitumoral<br />

T cell response, autochthonous gastric tumors<br />

do not respond to immunotherapy. This is the first<br />

comparison of the efficacy of adoptive T cell transfer<br />

between transplanted subcutaneous tumors<br />

and autochthonous tumors in the same animals.<br />

Our results suggest that in gastric cancer patients,<br />

even a strong antitumor T cell response will not<br />

efficiently penetrate the tumor in the absence of additional<br />

therapeutic strategies targeting the tumor<br />

microenvironment.<br />

103


060 Schmitt | Cellular therapy<br />

WT1 and RHAMM specific CD8+ T cells can be isolated<br />

and purified by streptamers for adoptive transfer<br />

Anita Schmitt, Xu Xun, Xinchao Wang, Inken Hilgendorf, Kersten Borchert, Mathias<br />

Freund, Michael Schmitt<br />

Department of Internal Medicine III, University of Rostock, Rostock, Germany<br />

104<br />

Background: A positive selection of leukemia as-<br />

sociated antigen (LAA)-specific T cells would be<br />

highly desirable to amplify the GVL effect and to<br />

decrease the risk of GVHD. Wilms Tumor gene 1<br />

(WT1) and the receptor for hyaluronic acid mediated<br />

motility (RHAMM) are LAAs recognized by<br />

CD8+ T lymphocytes. Streptamers constitute a<br />

novel multimer technology to select specific CD8+<br />

T cells, available at good manufacturing production<br />

(GMP) level. MATERIAL AND METHODS: Both tetramers<br />

and streptamers were used to detect the<br />

frequency of HLA-A2 restricted CD8+ T cells in<br />

the naïve peripheral blood (PB) from both healthy<br />

donors (HDs) and AML patients. RHAMM- and<br />

WT1-specific CD8+ T cells were further characterized<br />

for the expression of CD27, CD28, CD45RA<br />

and CCR7. In the next step, LAA-specific cells were<br />

positive selected by MACS columns after labeling<br />

with streptamers and thereafter immunophenotyped.<br />

Moreover, mixed lymphocyte peptide cultures<br />

(MLPCs) were performed to enrich WT1 specific<br />

T cells derived from the PB of HDs. RHAMM<br />

and WT1 specific cells were subjected to carboxylfluorescein<br />

succimidylester (CFSE) based proliferation<br />

and cytotoxicity assays.<br />

Results: 21 of 40 HDs showed naïve LAA specific T<br />

cell frequencies of 0.5 to 1.6% of all CD8+ T cells.<br />

In AML patients in complete remission significantly<br />

higher frequencies of LAA-specific T cells than<br />

in patients at diagnosis could be detected, up to<br />

6.0% of all CD8+ T cells. These cells revealed to be<br />

CD8+CD27-CD28+CD45RA+CCR7- effector T cells<br />

in flow cytometry. After positive selection by MACS<br />

columns a purity of 20-87% could be achieved for<br />

LAA-specific T cells. After a maximum of three<br />

rounds of MLPC, only a frequency of 2-5% could<br />

be achieved, thus demonstrating the power of the<br />

streptamer technology. Both tetramer- and streptamer-based<br />

selections yielded similar amounts of<br />

LAA-specific T cells.<br />

After positive selection, these T cells preserved an<br />

effector T cell phenotype and showed active proliferation<br />

in CFSE staining. Streptamer-selected T cells<br />

showed a trend towards higher CD28 expression<br />

thus indicating a more activated T cell status.<br />

Conclusion: In summary, the streptamer technology<br />

allows to select highly pure fractions of RHAMMand<br />

WT1-specific effector T cells with proliferative<br />

and cytotoxic properties. In analogy to DLIs specific<br />

for viral antigens such as CMVpp65, production<br />

of leukemia specific DLIs is feasible on a GMP level.<br />

Further leukemia antigens are currently evaluated<br />

by our group.


New targets & new leads<br />

105


061 Buchwald | New targets & new leads<br />

The ubiquitin-conjugase-8 targets active fms-like tyrosine<br />

kinase-3 for proteasomal degradation<br />

Marc Buchwald 1 , Thorsten Heinzel 1 , Frank D. Böhmer 2 , Oliver H. Krämer 1,3<br />

1 Friedrich-Schiller-University Jena, Center for Molecular Biomedicine Institute of Biochemistry and Biophysics<br />

2<br />

Friedrich-Schiller-University Jena, Center for Molecular Biomedicine Institute of Molecular Cell Biology,<br />

Hans-Knöll-Str. 2, D-07745 Jena, Germany<br />

3<br />

Friedrich-Schiller-University Jena, Center for Molecular Biomedicine, PD Dr. O. H. Krämer,<br />

Tel/Fax: 03641-949-362/949-352, e-mail: Oliver.Kraemer@uni-jena.de<br />

106<br />

The class III receptor tyrosine kinase FMS-like tyro-<br />

sine kinase 3 (FLT3) regulates normal hematopoiesis<br />

and immunological functions. Nonetheless, constitutively<br />

active mutant FLT3 (FLT3-ITD) causally<br />

contributes to transformation and is associated with<br />

poor prognosis of acute myeloid leukemia (AML)<br />

patients. Histone deacetylase inhibitors (HDACi)<br />

can counteract deregulated gene expression profiles<br />

and decrease oncoprotein stability, which renders<br />

them candidate drugs for AML treatment. However,<br />

these drugs have pleiotropic effects and it is often<br />

unclear how they correct oncogenic transcriptomes<br />

and proteomes. We report here that treatment of<br />

AML cells with the HDACi LBH589 induces the ubiquitin-conjugating<br />

enzyme UBCH8 and degradation<br />

of FLT3-ITD. Gain- and loss-of-function approaches<br />

demonstrate that UBCH8 and the ubiquitin-ligase<br />

SIAH1 physically interact with and target FLT3-ITD<br />

for proteasomal degradation. These ubiquitinylating<br />

enzymes though have a significantly lesser effect<br />

on wildtype FLT3. Furthermore, physiological and<br />

pharmacological stimulation of FLT3 phosphorylation,<br />

inhibition of FLT3-ITD auto-phosphorylation,<br />

and analyzing kinase-inactive FLT3-ITD revealed<br />

that tyrosine phosphorylation determines their proteasomal<br />

degradation. These results provide novel<br />

insights into anti-leukemic activities of HDACi and<br />

position UBCH8, which has been implicated primarily<br />

in processes in the nucleus, as a previously<br />

unrecognized important modulator of FLT3-ITD<br />

stability and leukemic cell survival.


062 Ashfield | New targets & new leads<br />

ImmTACs: bi-functional reagents for redirected tumour<br />

cell killing<br />

Rebecca Ashfield 1 , Linda Hibbert 1 , Nathaniel Liddy 1 , Giovanna Bossi 1 , Katherine Adams 1 ,<br />

Anna Lissina 2 , Tara Mahon 1 , Namir Hassan 1 , Jessie Gavarret 1 Frayne Bianchi 1 , Nicholas<br />

Pumphrey 1 , Kristin Ladell 2 , Emma Gostick 2 , Andrew Sewell 2 , Nikolai Lissin 1 , Peter Molloy 1 ,<br />

Yi Li 1 , Brian Cameron 1 , Malkit Sami 1 Emma Baston 1 , Penio Todorov 1 , Samantha Paston 1 ,<br />

Rebecca Dennis 1 , Andy Johnson 1 , David Price 2 , Annelise Vuidepot 1 , Daniel Williams 1 , Bent<br />

Jakobsen 1<br />

1 Immunocore Ltd, 57C Milton Park, Abingdon, Oxfordshire, OX14 4RX UK<br />

2 Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, Wales, UK<br />

The human immune system can theoretically<br />

identify malignant cells by inspecting cell surface<br />

Class I HLA -peptide complexes for the presence of<br />

disease-associated epitopes. Indeed, many cancer<br />

patients generate CD8 cyto-toxic T cell responses<br />

to tumour-associated antigens; the majority of patients,<br />

however, fail to clear tumours since T cell<br />

avidity for self-antigens tends to be weak, and<br />

cancer cells employ escape mechanisms for avoiding<br />

destruction by T cells. To overcome these<br />

issues, we have engineered novel, bi-functional<br />

protein therapeutics termed ImmTACs (Immune<br />

Mobilising mTCR Against Cancer) which re-direct<br />

the immune system to target and destroy tumour<br />

cells with a high degree of potency and specificity.<br />

An ImmTAC comprises a high affinity ‘monoclonal’<br />

T cell Receptor (mTCR) targeting a cancer-associated<br />

HLA-peptide complex, fused to an anti-CD3<br />

scFv domain which activates an anti-tumour T cell<br />

response.<br />

We demonstrate that ImmTACs against a number<br />

of different cancer-associated antigens can target<br />

and kill tumour cells expressing as few as 5-20 epitopes<br />

per cell with pico-Molar potency. ImmTACs<br />

preferentially activate effector memory CD8 T cells,<br />

resulting in secretion of multiple cytokines and<br />

tumour cell killing; a single activated T cell can kill<br />

multiple antigen positive tumour cells. Furthermore,<br />

we demonstrate that the reagents are able to<br />

inhibit tumour growth in mouse xenograft models.<br />

In vitro ImmTAC potency translates to a dose of<br />

less than 1mg in humans, representing a significant<br />

advance over existing targeted anti-cancer therapies<br />

including monoclonal antibodies.<br />

107


063 Domning | New targets & new leads<br />

Identification of acute myeloid leukemia-associated antigens<br />

recognized by allogeneic CD8+ T lymphocytes<br />

Sabine Domning 1 , Sylvia Köhler 1 , Eva Distler 1 , Christine Pohl 2 , Volker Lennerz 1 , Wolfgang<br />

Herr 1 , Thomas Wölfel 1 , Catherine Wölfel 1<br />

1 III. Medizinische Klinik, University Medical Center of the Johannes Gutenberg University, Mainz/Germany<br />

2 Institut für Pathologie, University Medical Center of the Johannes Gutenberg University, Mainz/Germany<br />

108<br />

CTL clones 1C6 and 4D7 against acute myeloid<br />

leukemia cells of patient MZ201 (AML FAB M5)<br />

were recently generated in allogeneic mixed lymphocyte<br />

leukemic cell cultures (allo- MLLC) using<br />

CD8+CD62Lhigh+ cells isolated from unrelated<br />

HLA class I-matched donor 332 (Distler, E., et al.,<br />

2008). Both CTL clones were used for T cell-directed<br />

expression screening of a cDNA library constructed<br />

from MZ201-AML cells as previously described<br />

(Wölfel, C., et al., 2008). Library screening with<br />

4D7 led to the identification of a novel minor histocompatibility<br />

antigen, PLAUR-317P, encoded by a<br />

known polymorphic allele of the plasminogen activator<br />

urokinase receptor gene (PLAUR, syn.: uPAR,<br />

CD87; SNP rs4760) and restricted by HLAB* 56:01.<br />

Prolin (P) on position 317 generated an aggretope. A<br />

10mer carrying the polymorphic residue on its 2nd<br />

position (PLAUR-317P316-325: SPTITLLMTA) was<br />

recognized at lowest concentrations (half maximal<br />

lysis at 5nM) whereas the homologous PLAUR-<br />

317L316- 325 was not recognized. The frequency of<br />

PLAUR-317P was determined via LightCycler PCR.<br />

Seventy-four of 252 individuals (29,4%) carried one<br />

PLAUR-317P allele, which exceeds the frequency<br />

indicated for SNP rs4760 in public domain databases.<br />

Approximately 2% of individuals were homozygous<br />

for PLAUR-317P. PLAUR is known to be overexpressed<br />

in AML, especially in subtype FAB M5,<br />

in multiple myeloma and in various solid cancers.<br />

As the target of 1C6 lymphocytes we identified chemokine<br />

(C-X-C motif) ligand 3 (CXCL3). Its immu-<br />

nogenic peptide (RLLRVALLL, amino acids 14-22)<br />

was recognized in association with HLA-A*02:01<br />

and was derived from the signal sequence of CXCL3.<br />

The same peptide is also encoded by the related<br />

genes CXCL1 and CXCL2, that both induce recognition<br />

by 1C6 upon transfection. Due to its hydrophobicity<br />

a rather high concentration of the synthetic<br />

peptide was required in reconstitution assays (halfmaximal<br />

lysis induced at 0,3 µM). 1C6 recognized<br />

HLA-A*02:01+ leukemic cells of myeloid origin<br />

(AML and CML) (Distler, E., et al., 2008). Monocytes<br />

and endothelial cells, but not epithelial cells,<br />

isolated from HLA-A*02:01+ donors were weakly<br />

recognized, but their recognition could be strongly<br />

enhanced by pretreatment with proinflammatory<br />

molecules (IL1-ß, TNF-alpha and LPS).<br />

Using cDNA expression cloning with allogeneic<br />

leukemia-reactive T cells generated from a healthy<br />

donor we identified two antigens of distinct categories<br />

(mHag, structurally unaltered) that are<br />

overexpressed in myeloid leukemias and, among<br />

non-malignant cells, preferentially expressed in hematopoetic<br />

cells. Both antigens are candidates for<br />

GvL effects and GvHD.<br />

References: - Distler, E. et al. Acute myeloid leukemia (AML)reactive<br />

cytotoxic T lymphocyte clones rapidly expanded from<br />

CD8(+) CD62L(high)+ T cells of healthy donors prevent AML<br />

engraftment in NOD/SCID IL2Rgamma(null) mice. Exp. Hematol.<br />

36:451-463, 2008 - Wölfel, C. et al. Dissection and molecular analysis<br />

of alloreactive CD8+ T cell responses in allogeneic haematopoietic<br />

stem cell transplantation. Cancer Immunol. Immunother.<br />

57:849- 857, 2008


064 Hornig | New targets & new leads<br />

Combinatorial approach of a bispecific antibody and costimulatory<br />

antibody fusion proteins for targeted cancer<br />

immunotherapy<br />

Nora Hornig, Philipp Diebolder, Yvonne Eichinger, Roland E. Kontermann & Dafne Müller<br />

Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany<br />

Recombinant bispecific antibodies have shown to<br />

be able to retarget cytotoxic T cells to tumor cells<br />

in a MHC-independent manner, triggering effector<br />

cell activation and consecutive tumor cell killing.<br />

Considering that costimulation is an essential requirement<br />

not only to initiate T cell activation, but<br />

also for the regulation of a proper T cell response,<br />

we propose a combinatorial approach by a recombinant<br />

bispecific antibody and targeted costimulatory<br />

ligands in order to generate a highly efficient<br />

antitumoral immune response. In our model<br />

system we used a bispecific antibody recognizing<br />

the fibroblast activation protein (FAP) on tumor<br />

cells and CD3 on T cells. For targeted costimulation<br />

we generated antibody fusion proteins composed<br />

of a tumor binding antibody moiety and the extracellular<br />

domain of the costimulatory ligands B7.2<br />

and 4-1BBL, respectively. In view of the concomitant<br />

binding requirement of these constructs to the<br />

tumor cell, different antibody moieties, targeting<br />

the antigens FAP and endoglin on the tumor cell,<br />

were used. Costimulatory effects were assayed by<br />

incubating the fusion proteins with the bispecific<br />

antibody on tumor cells in presence of PBMCs and<br />

monitoring IL-2 and IFN-γ release. We showed that<br />

T cell activation induced by the bispecific antibody<br />

was significantly enhanced by targeted costimulation<br />

either with the B7.2 or the 4-1BBL fusion<br />

protein in a concentration-dependent and ligandspecific<br />

manner.<br />

Moreover, further signal enhancement was achie-<br />

ved by the combined application of both costimulatory<br />

fusion proteins, showing a synergistic effect.<br />

Thus, application of recombinant bispecific antibodies<br />

with combinations of tumor directed costimulatory<br />

fusion proteins of B7.2 and 4-1BBL might<br />

be a promising approach for efficient retargeting<br />

and activation of cytotoxic T lymphocytes in cancer<br />

immunotherapy.<br />

109


065 Casares | New targets & new leads<br />

Suppression of Treg activity by a FOXP3 inhibitor peptide<br />

Noelia Casares, Laura Arribillaga, Francesc Rudilla, Diana Llópiz, José Ignacio Riezu-Boj,<br />

Pablo Sarobe, Francisco Borrás-Cuesta, Jesús Prieto and Juan José Lasarte<br />

Gene Therapy and Hepatology Area, University of Navarra, Center for Applied MedicalResearch (CIMA),<br />

Pamplona, Spain<br />

110<br />

Down-regulation of regulatory T (Treg) cell func-<br />

tion might be beneficial to enhance the immuno-<br />

genicity of viral and tumor vaccines or to induce<br />

breakdown of immunotolerance. Although the mechanism<br />

of suppression used by Treg cells remains<br />

controversial, it is already accepted that FOXP3 is<br />

the master gene in this subpopulation of cells.<br />

In this study, using a phage display random peptide<br />

library, we have identified a peptide, called P60,<br />

able to bind FOXP3 when measured by surface<br />

plasmon resonance. In vitro studies demonstrate<br />

that P60 inhibits murine and human Treg activity.<br />

In mice, it was found that P60 was able to restore<br />

the proliferation of murine spleen cells in response<br />

to anti-CD3 stimulation, which was inhibited by<br />

the presence of Treg. In human, P60 restored proliferation<br />

and IFN-g production of human peripheral<br />

mononuclear cells in a mixed leukocyte reaction<br />

inhibited by the addition of Tregs. Related to the<br />

mechanism of action, when we used a FOXP3-GFP<br />

transfection system in 293 cells, we found that P60<br />

is able to inhibit nuclear translocation of FOXP3.<br />

Moreover, P60 reduced the capacity of FOXP3 to<br />

inhibit NFAT and NF-kB transcriptional activity in<br />

Jurkat and 293 cells respectively.<br />

In vivo, selective inhibition of FOXP3 with P60 in<br />

newborn mice induced a lymphoproliferative autoimmune<br />

syndrome resembling the reported pathology<br />

in scurfy mice lacking functional FOXP3.<br />

However, P60 administration didn’t cause toxic<br />

effects in adult mice and, when given to BALB/c<br />

mice immunized with the cytotoxic T-cell epitope<br />

AH1 from CT26 tumor cells, it induced protection<br />

against tumor implantation. Similarly, P60 improved<br />

the antiviral efficacy of a recombinant adenovirus<br />

expressing NS3 protein from hepatitis C virus.<br />

These findings demonstrate that functional inhibition<br />

of Treg by the FOXP3-inhibitory peptide P60<br />

constitutes a strategy to enhance antitumor and<br />

antiviral immunotherapies.


066 Dettmar | New targets & new leads<br />

Identification of clinically useful combinations of trifunctional<br />

antibodies with chemotherapy using different preclinical<br />

models<br />

Kirsten Dettmar 1 , Petra Schroeder 2 , Carsten Lindemann 2 , Andrea Eberhardt 1 , Franziska<br />

Hirschhaeuser 3 , Diane Seimetz 1 , Judith Atz 1<br />

1 Fresenius Biotech GmbH, Munich, Germany<br />

2 EUFETS GmbH, Idar-Oberstein, Germany<br />

3 Institute of Physiology and Pathophysiology, University Medical Center of the Johannes Gutenberg-University<br />

Mainz, Germany<br />

The trifunctional bispecific antibodies catumaxo-<br />

mab (anti-EpCAM x anti-CD3) and ertumaxomab<br />

(anti-HER-2/neu x anti-CD3) exert their mechanism<br />

of action by simultaneous recruitment and activation<br />

of two different types of immune effector cells<br />

(T-cells and accessory cells) at the tumor site. Thus<br />

tumor cells and immune effector cells are brought<br />

into close proximity leading to physiological co-stimulation<br />

of T-cells, improved tumor cell elimination<br />

by different immunologic killing mechanisms<br />

and phagocytosis, as well as processing and presentation<br />

of tumor material by bound and activated<br />

accessory cells.<br />

Despite the development of different antibody therapies,<br />

chemotherapy (CTX) still represents the mainstay<br />

of cancer therapy. Therefore many antibodies<br />

are either used in combination with the established<br />

chemotherapeutic therapies or in short intervals afterwards.<br />

However, the commonly observed hematological<br />

toxicities of chemotherapeutics are regularly associated<br />

with some degree of immunosuppression.<br />

On the other hand, some of these chemotherapeutic<br />

agents are reported to support or even enhance the<br />

anti-tumor effect of immunotherapeutic drugs in<br />

certain doses.<br />

A largely intact immune system is the key for trifunctional<br />

antibodies to exhibit their full mode of action.<br />

Since chemotherapy might impact the number and<br />

/ or function of immune effector cells, we evaluated<br />

in different preclinical in vitro models whether the<br />

efficacy of trifunctional bispecific antibodies would<br />

be influenced when combined with chemotherapeutic<br />

drugs from different drug classes.<br />

The preclinical studies performed included in vitro<br />

cytotoxicity assays with established tumor cell lines<br />

evaluating potential synergy by using the method<br />

of Chou and Talalay. Furthermore, results from 3D<br />

tumor spheroids and from an autologous human<br />

ex vivo setting are presented. In addition, immune<br />

cells from cancer patients were obtained at different<br />

time points before, during and after chemotherapy<br />

and were assessed for their in vitro cytotoxicity mediated<br />

by trifunctional antibodies.<br />

So far the results from the different in vitro assay<br />

systems used indicate synergy for the combination<br />

of the trifunctional antibodies with 5-Fluorouracil<br />

(5-FU), cisplatin and epirubicin. Furthermore, no<br />

negative influence of these chemotherapeutic drugs<br />

was observed on the trifunctional mode of action<br />

in vitro. Catumaxomab and ertumaxomab were<br />

able to mediate an effective killing of tumor cells<br />

with immune cells from chemotherapy patients<br />

shortly after chemotherapy. The respective patients<br />

immune cells can be activated by these bispecific<br />

antibodies one week after chemotherapy with pyrimidine<br />

antagonists (5-FU) and alkylating agents<br />

(cisplatin) resulting in significant tumor cell killing<br />

in vitro.<br />

These results provide a basis for the possible combination<br />

of these drugs with trifunctional antibodies<br />

in the clinical setting.<br />

111


067 Hirschhaeuser | New targets & new leads<br />

Efficacy of catumaxomab in tumor spheroid killing is mediated<br />

by its trifunctional mode of action<br />

Franziska Hirschhaeuser 1 , Kirsten Dettmar 2 , Judith Atz 2 and Wolfgang Mueller-Klieser 1<br />

1 Institute of Physiology and Pathophysiology, University Medical Center of the Johannes Gutenberg-University<br />

Mainz, Germany<br />

2<br />

Fresenius Biotech GmbH, Munich, Germany<br />

112<br />

Catumaxomab is an intact trifunctional bispecific<br />

antibody targeting human EpCAM (epithelial cell<br />

adhesion molecule) and CD3 with further binding to<br />

activating Fcγ receptor type I, IIa and III. We chose<br />

multicellular tumor spheroids (MCTS) of human<br />

EpCAM-positive FaDu tumor cells in co-culture with<br />

human peripheral blood mononuclear cells as an adequate<br />

three-dimensional in vitro model for pharmacological<br />

testing of catumaxomab.<br />

Besides catumaxomab, we used the F(ab`)2 fragments<br />

of catumaxomab and the parental antibodies 26/II/6<br />

(anti-human CD3) and HO-3 (anti-human EpCAM), as<br />

well as BiLu (anti-human EpCAM x anti-mouse CD3)<br />

which is not able to bind to human T cells. We assessed<br />

the spheroid volume growth, cytokine release (IL 2, IL<br />

6, IFN γ, TNF α), and immunohistological stainings for<br />

different immune cell types and for proliferation.<br />

After 6 days, spheroids cultured with the EpCAMtargeting<br />

antibody HO-3 showed almost the same<br />

volume as control spheroids without antibody. Combination<br />

of 2.5 ng/ml HO-3 with 2.5 ng/ml 26/II/6<br />

resulted in spheroid volumes which were reduced by<br />

61%; and approaches with 5.0 ng/ml 26/II/6 showed a<br />

70% volume reduction. Catumaxomab induced a 91%<br />

decrease in spheroid volume. Comparing the effects of<br />

26/II/6 and HO-3 to catumaxomab, demonstrates less<br />

efficacy of the parental antibodies either used alone or<br />

in combination. The importance of the CD3 binding<br />

site was emphasized by further control experiments<br />

with BiLu. Similar to HO-3, BiLu showed no tumor<br />

spheroid killing. The essential function of the intact<br />

Fc region was demonstrated in control experiments<br />

using the F(ab’)2 fragment of catumaxomab: The<br />

bispecific antibody fragment lacking this functional<br />

binding site resulted in a dose-dependent volume reduction<br />

of FaDu spheroids ranging from 54-81% for<br />

concentrations of 1.0, 2.5 and 10.0 ng/ml, while catumaxomab<br />

caused a decrease of 90-100% within the<br />

same concentration range. The highest concentrations<br />

of secreted IL 2 and IFN γ were observed in the setting<br />

with tumor spheroids, PBMCs and catumaxomab.<br />

Cultures of PBMC and 5.0 ng/ml 26/II/6 yielded the<br />

highest levels for IL 6 and TNF α. In contrast, incubation<br />

with BiLu did not induce cytokine secretion,<br />

and the incubation with F(ab’)2 resulted in increased<br />

cytokine levels only for 10.0 ng/ml, but several-fold<br />

less than induced by catumaxomab. The immunological<br />

stainings showed that the infiltrating lymphocytes<br />

are mainly CD8+ T cells in spheroids treated with<br />

catumaxomab and its F(ab’)2 fragment, whereas the<br />

total amount of CD45+ cells in catumaxomab treated<br />

spheroids was higher.<br />

In summary, the results show, that all binding partners<br />

of the postulated tricell complex have to be<br />

present to exert catumaxomab’s full mode of action.<br />

These distinct effects of catumaxomab are based on<br />

the unique composition of the trifunctional bispecific<br />

antibody.<br />

Acknowledgements<br />

The technical assistance of S. Dahms-Prätorius and R. Santos is<br />

greatly acknowledged.<br />

Supported by Fresenius Biotech GmbH and TRION Pharma GmbH<br />

(Munich, Germany).


068 Guthmann | New targets & new leads<br />

Cellular and humoral immune response to N-glycolyl-GM3<br />

elicited by racotumomab, an anti-idiotypic vaccine<br />

Marcelo D Guthmann 1 , Cecilia Venier 1 , Estrella Levy 1 , Mónica A Castro 2 , Gabriela Cinat 2 ,<br />

Roberto Gómez 3 , Ana María Vázquez 4 , Leonardo Fainboim 1<br />

1 Hospital de Clínicas José de San Martín, University of Buenos Aires, Argentina<br />

2 Instituto de Oncología A. Roffo, University of Buenos Aires, Argentina<br />

3 Laboratorio Elea, Buenos Aires, Argentina<br />

4 Center of Molecular Immunology, Havana, Cuba<br />

Gangliosides are a family of sialylated glycolipids<br />

which are normal components of the cell membrane.<br />

They have been found to be important actors in<br />

multiple aspects of cellular interaction with its environment<br />

and with transmembrane signaling. As<br />

such, they are involved in cancer progression and<br />

have become the focus of several immunotherapeutic<br />

approaches. Not all gangliosides are equally<br />

immunogenic. N-acetyl-GM3, the main gangliosides<br />

on the cell surface and the most abundant<br />

ganglioside in normal serum, is one of the most<br />

immunologically tolerated member of the family.<br />

In contrast, N-glycolyl-GM3, is not expressed in<br />

human normal tissues due to a species-specific<br />

genetic mutation that abrogates the biosynthesis of<br />

N-glycolylneuraminic acid (Neu5Gc). Neu5Gc has<br />

been reported, however, in human tumors, and its<br />

presence might be derived from dietary sources<br />

or a yet unknown alternate synthesis pathway. Nglycolyl-GM3<br />

is expressed in melanoma, breast and<br />

lung cancer cells, and is highly immunogenic. As a<br />

result, it has been considered as a target of choice<br />

for immunotherapy.<br />

We investigated with an extended vaccination protocol<br />

the immunogenicity and toxicity profile of racotumomab,<br />

an anti-idiotypic vaccine mimicking<br />

N-glycolyl-GM3. The year-long vaccination scheme<br />

consisted of six bi-weekly intradermal injections<br />

followed by 10 monthly boosters. Nineteen patients<br />

with high risk (stage III) or metastatic breast<br />

cancer were vaccinated with different dose levels<br />

of racotumomab (0.5, 1 and 2 mg). The humoral<br />

and cellular responses to racotumomab and to the<br />

targeted ganglioside were assessed at baseline and<br />

throughout the treatment. Anti-idiotype antibodies<br />

and anti-N-glycolyl-GM3 IgM and IgG antibodies<br />

were determined by ELISA. Serum antibody reactivity<br />

was also tested against P3X63 Ag8 653 murine<br />

myeloma cells and B16 murine melanoma cells.<br />

Frequency of N-glycolyl-GM3—reactive cells was<br />

calculated by IFNγ ELISPOT. For that purpose, cryopreserved<br />

PBMC and autologous DC were incubated<br />

with N-glycolyl-GM3—containing liposomes.<br />

Equivalent amounts of unloaded liposomes were<br />

added to control wells. After a 24-h culture, cells in<br />

each well were resuspended and transferred to triplicate<br />

wells in an IFNγ-precoated ELISPOT plate.<br />

Subsequent steps for detection of IFNγ-secreting<br />

cells followed standard ELISPOT procedures. Patients<br />

with no response at baseline and with a posttreatment<br />

two-fold increase in the number of spots<br />

in N-glycolyl-GM3 stimulated wells (versus unstimulated<br />

wells) were considered as responsive.<br />

All patients showed a strong antibody response to<br />

N-glycolyl GM3. In addition, ganglioside-specific<br />

IFNγ responses were recorded in 5 of 13 evaluable<br />

patients.<br />

113


069 Sørensen | New targets & new leads<br />

Cellular Immune Responses Against Indoleamine<br />

2,3-dioxygenase<br />

Rikke Bæk Sørensen, Inge Marie Svane, Per thor Straten and Mads Hald Andersen<br />

Center for Cancer Immune Therapy (CCIT), Department of Hematology, 54P4, University Hospital Herlev,<br />

Herlev Ringvej 75, DK-2730 Herlev, Denmark<br />

114<br />

Indoleamine 2,3-dioxygenase (IDO) is an immu-<br />

noregulatory enzyme that are implicated in sup-<br />

pressing T-cell immunity in normal and patholo-<br />

gical settings. Expression of IDO has been shown<br />

to induce T-cell anergy and/or the generation of<br />

adaptive regulatory T cells. In cancer patients IDO<br />

is expressed within the tumor itself as well as in<br />

antigen-presenting cells in tumor-draining lymph<br />

nodes, where it promotes the establishment of peripheral<br />

immune tolerance to tumor antigens. In the<br />

present study, we tested the notion whether IDO<br />

itself may be subject to immune responses. The<br />

study unveiled spontaneous cytotoxic T-cell reactivity<br />

against IDO in peripheral blood as well as<br />

in the tumor microenvironment of different cancer<br />

patients. We demonstrate that these IDO reactive T<br />

cells are indeed peptide specific, cytotoxic effector<br />

cells. Hence, IDO reactive T cells are able to recognize<br />

and kill tumor cells including directly isolated<br />

AML blasts as well as IDO-expressing dendritic<br />

cells, i.e. one of the major immune suppressive cell<br />

populations. Consequently, IDO may serve as an<br />

important and widely applicable target for anticancer<br />

immunotherapeutic strategies. Furthermore,<br />

as emerging evidence suggests that IDO constitutes<br />

a significant counter-regulatory mechanism<br />

induced by pro-inflammatory signals, IDO-based<br />

immunotherapy holds the promise to boost anticancer<br />

immunotherapy in general.<br />

Furthermore, we show that spontaneous cytotoxic<br />

T-cell reactivity against IDO exists in healthy<br />

individuals. Such IDO-specific T cells are able to<br />

boost immunity against CMV antigens by eliminating<br />

IDO+ suppressive cells. IDO-specific T-cells<br />

are induced in healthy individuals by non-specific<br />

inflammatory stimuli such as IFN-γ and IL-2. IDO<br />

specific T cells may play a vital role for the mounting<br />

or keeping of an effective adaptive immune<br />

response.


070 Andersen | New targets & new leads<br />

Identification of highly potent regulatory CD8+ T-cells<br />

specific for Heme Oxygenase-1<br />

Mads Hald Andersen 1 , Rikke Bæk Sørensen 1 , Marie Brimnes 1 , Inge Marie Svane 1 , Jürgen C<br />

Becker 2 , Per thor Straten 1<br />

1 Center for Cancer Immune Therapy (CCIT), Department of Hematology, Herlev University Hospital,<br />

Dk-2730 Herlev, Denmark<br />

2 Department of Dermatology, University of Würzburg, D-97080 Würzburg, Germany<br />

Regulatory elements are essential for the home-<br />

ostasis of the immune system. While regulatory<br />

CD4+T cells have received much attention over the<br />

past years, less is known about regulatory CD8+ T<br />

cells; moreover, potential antigens recognized by<br />

these cells remain elusive. In the present study we<br />

describe Heme Oxygenase-1 (HO-1) specific, HLA<br />

restricted, CD8 positive T-cells, which are able to<br />

suppress cells immune responses with an hitherto<br />

unseen efficacy. HO-1 has been defined as a “protective<br />

gene” with anti-inflammatory functions and<br />

is expressed at high levelt in malignant cells. Remarkably,<br />

HLA-restricted, HO-1 specific CD8 T-cells<br />

were readily detected directly ex vivo in peripheral<br />

blood lymphocytes obtained from cancer patients<br />

and in situ among tumor infiltrating T-cells. These<br />

HO-1 specific T cells were not only able to suppress<br />

IFN-γ release of antigen specific T cells, but also<br />

inhibited both the activity of CD4+ and CD8+ T<br />

cells in proliferation and cytotoxic assays. Notably,<br />

the inhibitory effect of HO-1 specific T cells was<br />

far more pronounced compared to conventional<br />

CD4+CD25+CD127- regulatory T-cells. The inhibitory<br />

activity of HO-1 specific T cells seems at least<br />

partly to be mediated by soluble factors. The identification<br />

of a new subset of highly potent, antigen<br />

specific CD8+ regulatory T-cells open new avenues<br />

for therapeutic interventions both in autoimmune<br />

diseases and cancer.<br />

115


071 Kleemann | New targets & new leads<br />

Identification of T-cell defined varicella-zoster virus proteins<br />

Patrick Kleemann 1 , Stefan Tenzer 2 , Eva Distler 1 , Eva Wagner 1 Ralf G. Meyer 1 , Sebastian<br />

Klobuch 1 , Simone Thomas 1 , Steffi Aue 3 , Bodo Plachter 3 , Wolfgang Herr 1<br />

1 Dept. of Medicine III, Hematology & Oncology, University Medical Center, Mainz, Germany<br />

2 Institute for Immunology, University Medical Center, Mainz, Germany<br />

3 Institute for Virology, University Medical Center, Mainz, Germany<br />

116<br />

Reactivated infection with varicella-zoster virus<br />

(VZV) causes herpes zoster. In immunodeficient<br />

cancer patients (e.g. after hematopoietic stem cell<br />

transplantation) dissemination of the virus can lead<br />

to life-threatening disease. Although an efficient<br />

live attenuated VZV vaccine for zoster prophylaxis<br />

exists, it is not approved in immunocompromised<br />

patients due to safety reasons. Knowledge of immunogeneic<br />

VZV proteins would allow designing a noninfectious<br />

nonhazardous subunit vaccine suitable<br />

for patients with immunodeficiencies. The objective<br />

of this study is to identify T-cell defined virus<br />

proteins of a VZV-infected Vero cell extract that we<br />

have recently described as a reliable antigen format<br />

for IFN-g ELISPOT assays (Distler et al. Biol. Blood<br />

Marrow Transplant. 2008, 14:1417-24). We first<br />

separated the VZV-infected/-uninfected Vero cell<br />

extracts by ultracentrifugation and reverse-phase<br />

HPLC. The collected fractions were screened for<br />

reactivity with peripheral blood mononuclear cells<br />

(PBMC) of VZV-seropositive healthy individuals<br />

by IFN-g ELISPOT assay. Using this strategy, we<br />

successfully identified bioactive HPLC fractions<br />

that contained immunogeneic VZV material. The<br />

immune reactivity was mediated by CD4+ memory<br />

T cells of VZV-seropositive healthy individuals as<br />

demonstrated by experiments with T-cell subpopulations.<br />

We next analyzed the bioactive HPLC<br />

fractions with MALDI and ESI mass spectrometry<br />

techniques and identified three (glycoprotein<br />

E; glycoprotein B and immediate early protein 62)<br />

different VZV derived proteins. In subsequent experiments<br />

we expressed these VZV-encoded proteins<br />

in dendritic cells (DCs) by in vitro transcribed<br />

RNA and screened transfected DCs with autologous<br />

T cells. Based on these experiments we established<br />

a protocol to perform these studies with PBMC of<br />

healthy donors and patients after hematopoietic<br />

stem cell transplantation. The work will hopefully<br />

lead to protein candidates of VZV to be included<br />

in a subunit vaccine, which can be safely used for<br />

zoster prophylaxis in immunocompromised cancer<br />

patients.


072 van Esch | New targets & new leads<br />

T cell epitope discovery through HLA class I peptide ligand exchange<br />

and combinatorial coding<br />

Wim van Esch 1 , Juk Yee Mok 1 , Annemieke Molenaar 1 , Arnold Bakker 2 , Mireille Toebes 2 ,<br />

Sine Reker Hadrup 2 , Chengyi Shu 2 , Ernst Soethout 3 , Josine van Beek 3 , Huib Ovaa 4 and Ton<br />

Schumacher 2<br />

1 Dept. R&D, Sanquin Reagents, Amsterdam, The Netherlands<br />

2 Div. Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands<br />

3 Netherlands Vaccine Institute, Bilthoven, The Netherlands<br />

4 Div. Cellular Biochemistry, The Netherlands Cancer Institute, Amsterdam, The Netherlands<br />

Human major histocompatibility complex class I<br />

(HLA-I) consists of beta2-microglobulin, a polymorphic<br />

heavy chain and a peptide ligand. Peptides<br />

in context with HLA-I on the cell surface are<br />

recognized by appropriate cytotoxic T cells. Our<br />

aim is to develop a platform for high-throughput<br />

identification of HLA-I ligands and T cell detection<br />

using flow cytometry in order to detect new<br />

disease-specific epitopes/T cells. To this end, conditional<br />

ligands that can be cleaved in the HLAbound<br />

state upon UV irradiation were designed<br />

for different HLA alleles. Cleavage of HLA-I-bound<br />

conditional ligand in the presence of another experimental<br />

ligand results in peptide exchange.<br />

Only those peptides that fit in the peptide binding<br />

groove will be able to maintain the integrity of the<br />

empty HLA complex, and the efficiency of HLA-I<br />

stabilization by experimental ligands can be determined<br />

by ELISA. Subsequently, HLA-I-binding<br />

peptides identified in this manner are tested for<br />

their immunological relevance. To this purpose, a<br />

combinatorial coding technology has been developed<br />

that allows the parallel detection of a multitude<br />

of different T cell populations in a single sample.<br />

Detection of antigen-specific T cells from peripheral<br />

blood by combinatorial coding is as efficient<br />

as detection with conventionally fluorescently-labeled<br />

HLA multimers and allows comprehensive<br />

screens to be performed. Using these technologies,<br />

several screens for HLA ligands have been performed,<br />

either manually or using robotics, using<br />

peptides potentially associated with infectious or<br />

non-infectious diseases. Large-scale screenings of<br />

their immunological relevance by analysis of T cell<br />

responses using combinatorial coding are ongoing.<br />

Together these techniques form a highly efficient<br />

platform for T cell epitope discovery in a broad<br />

range of human diseases and for the monitoring of<br />

disease- and therapy-induced T cell immunity.<br />

117


073 Suso | New targets & new leads<br />

Identification of novel CD4+ and CD8+ T cell epitopes from<br />

telomerase (hTERT)<br />

Else Marit Inderberg Suso 1 , Anne-Marie Rasmussen 1 , Sissel Trachsel 1 , Steinar Aamdal 2 ,<br />

Svein Dueland 2 , Gunnar Kvalheim 3 , Gustav Gaudernack 1<br />

1 Section for Immunology, Oslo University Hospital-Radiumhospitalet, Norway<br />

2 Dept for Clinical Research, Oslo University Hospital-Radiumhospitalet, Norway<br />

3 Section for Cell Therapy, Oslo University Hospital-Radiumhospitalet, Norway<br />

118<br />

We have a number of patients who have been vacci-<br />

nated with the telomerase peptide vaccine GV1001<br />

or with dendritic cells (DCs) loaded with hTERT<br />

mRNA.<br />

Among these patients, several have had an extraordinary<br />

clinical course, involving disease stabilization<br />

over many years, partial and even complete<br />

remission of the disease. These patients offer a<br />

unique opportunity to study in detail the immune<br />

response against the vaccine.<br />

We have identified several novel hTERT epitopes by<br />

studying responses against a panel of 24 overlapping<br />

15-mer hTERT peptides as well as one 30-mer<br />

peptide. The results indicate that a profound epitope<br />

spreading within the hTERT antigen takes place in<br />

some patients following vaccination with a single<br />

T helper epitope (GV1001), and that many of these<br />

epitopes are recognized also following vaccination<br />

with hTERT transfected DCs. CD4+ T-cell clones<br />

specific for some of these peptides have been generated.<br />

In addition, CD8+ T cells specific for novel<br />

hTERT epitopes have been identified by pentamer<br />

staining.<br />

The recognition of these epitopes in a large number<br />

of patients further indicates a high degree of immunogenicity<br />

and HLA promiscuity. This also suggests<br />

that these peptides may be clinically relevant<br />

epitopes. Their identification is important for the<br />

development of the next generation of immunotherapy.<br />

We are now developing a second generation<br />

of hTERT peptide vaccines as well as T-cell receptor<br />

transfer based on “clinically validated” T cell receptors<br />

cloned from long term survivors.


074 Krug | New targets & new leads<br />

Transfer of mRNA encoding chimeric antigen receptors specific<br />

for MCSP into CD4+ and CD8+ T cells<br />

Christian Krug 1 , Katrin Birkholz 1 , Hinrich Abken 2 , Michael Schwenkert 3 , Georg Fey 3 , Gerold<br />

Schuler 1 , Jan Dörrie 1 ,*, and Niels Schaft 1 ,*<br />

1 Department of Dermatology, University Hospital Erlangen, Erlangen, Germany<br />

2 Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany<br />

3 Chair of Genetics, University of Erlangen-Nuremberg, Erlangen, Germany<br />

* contributed equally<br />

Adoptive transfer of bulk T cells with a tumoran-<br />

tigen-specific T-cell receptor (TCR) is an innova-<br />

tive and promising approach to treat malignancies.<br />

Chimeric antigen receptors (CAR), which consist<br />

of a scFv directed against a cancer surface antigen<br />

and the signaling domain of CD3-zeta and usually<br />

also of CD28 molecules, are an attractive alternative<br />

for normal TCR since MHC-restricted antigen<br />

presentation is not required. To avoid persistent<br />

auto-aggression, a reported life threatening risk of<br />

engineered T cells with constitutive CAR expression,<br />

we explored mRNA electroporation for transient<br />

receptor expression in human T cells.<br />

CAR, consisting of different antigen-binding and signaling<br />

domains, specific for melanoma-associated<br />

chondroitin sulfate proteoglycan (MCSP), which is<br />

expressed on melanomas, basal cell carcinomas,<br />

and some forms of childhood leukemias, were efficiently<br />

transfected into CD4+ and CD8+ T cells.<br />

Expression kinetics of the CAR were studied, and at<br />

day 9 after electroporation CAR-expression had disappeared.<br />

Upon specific stimulation with MCSP+<br />

tumor cells, transfected CD4+ and CD8+ T cells<br />

secreted the cytokines IL-2, TNF-alpha, and IFNgamma.<br />

Moreover, the reprogrammed T cells were<br />

capable of killing target cells in an antigen-specific<br />

manner. The comparison of different CAR showed<br />

that using a binding domain with a higher affinity<br />

and stability improved the recognition of tumor<br />

cells. Furthermore, the incorporation of a CD28<br />

signaling domain sometimes improved the CAR<br />

surface expression, and always improved the lytic<br />

capacity and cytokine secretion of the T cells.<br />

In aggregate, this study shows for the first time a<br />

direct comparison of CAR with different scFv specific<br />

for the same antigen. Furthermore, RNA electroporation<br />

provides us with a technique to rapidly<br />

compare and choose CAR best suited for the immunotherapy<br />

of cancer.<br />

119


075 Lubojanski | New targets & new leads<br />

Identification of T cell-defined melanoma antigens<br />

S. Lubojanski 1 , M. Fatho 1 , A. Paschen 2 , D. Eberts 1 , C. Wölfel 1 , D. Schadendorf 2 , V. Lennerz<br />

1 , T. Wölfel 1<br />

1 III. Medizinische Klinik, University Medical Center Mainz<br />

2 Department of Dermatology, University Hospital Essen<br />

120<br />

CD8+ T lymphocytes can identify and eliminate<br />

cancer cells by recognizing peptides derived from<br />

tumor associated antigens (TAA) via HLA class I<br />

molecules. Although cancer cells are less immunogenic<br />

than pathogens, current knowledge predicts<br />

a high degree of individuality in tumor/T cell interaction.<br />

Even though some TAA and their epitopes<br />

have been identified, increasing knowledge of their<br />

identity should indicate the way to an effective strategy<br />

for specific immunotherapy [1].<br />

The aim of this work is to identify and characterize<br />

T cell-defined antigens in the melanoma model<br />

MA-MEL-86. This model was derived from a female<br />

patient who had been first vaccinated with peptides<br />

and later on with tumor-lysate pulsed DCs.<br />

Three permanent melanoma cells lines had been<br />

established at different time points from resected<br />

lymph node metastases (MA-MEL-86A, -86B and<br />

-86C). These lines differ in their expression of<br />

the HLA I alleles. While MA-MEL 86A expresses<br />

HLA-A*01:01/-A*24:02, HLA-B*08:01/-B*15:01 and<br />

HLA-C*03:03/ -C*07:01; MA-MEL-86C had lost one<br />

haplotype (HLA-A*24:02, -B*15:01, -C*03:03), and<br />

due to a mutation in the ß2-microglobulin gene,<br />

MA-MEL-86B does not express HLA class I alleles<br />

at all. The enrichment of the melamoma-reactive T<br />

cells in autologous mixed lymphocyte-tumor cell<br />

cultures (MLTCs) was performed by weekly stimulation<br />

of peripheral blood lymphocytes (PBL)<br />

or PBL-derived CD8+ T cells against MA-MEL-86A<br />

and MA-MEL-86C, respectively. MLTC responders<br />

and T cell clones derived thereof were applied to expression<br />

screening of an antigen panel comprising<br />

39 known melanoma-associated antigens in association<br />

with the patient´s HLA class I alleles. They<br />

were also applied to expression screening of cDNA<br />

libraries constructed from cell lines MA-MEL-86A<br />

and -86C. For a detailed molecular analysis of the<br />

anti-tumor T cell repertoire. So far, three distinct<br />

antigens were identified in this model. MLTC responders<br />

stimulated with MA-MEL-86A were found<br />

to target a mutated neoantigen in association with<br />

HLA-B*15:01 (HERPUD1mut/B15). In addition,<br />

tumor-reactive T cells stimulated with MA-MEL-<br />

86C recognized known peptides from tyrosinase<br />

and gp100 both restricted by HLA-A*01:01. T cells<br />

against MA-MEL-86 cells predominantly recognize<br />

antigens not contained in the above mentioned<br />

panel. They are currently applied to further screening<br />

experiments.<br />

[1] Lennerz, V. et al. The response of autologous T cells to a<br />

human melanoma is dominated by mutated neoantigens. Proc<br />

Natl Acad Sci U S A. 102(44):16013-8, 2005


076 Staege | New targets & new leads<br />

Tumor antigens in Hodgkin lymphoma<br />

Martin S. Staege 1 , Carolin Winkler 1 , Daniela Max 1 , Wolfgang Altermann 2 , Gerald Schlaf 2 ,<br />

Ursula Banning 1 , Dieter Körholz 1<br />

1 Department of Pediatrics, Childrens Cancer Research Center, Martin-Luther-University Halle-Wittenberg,<br />

06120 Halle, Germany<br />

2 University Medical Center, HLA-Laboratory, Martin-Luther-University Halle-Wittenberg,<br />

06120 Halle, Germany<br />

Hodgkin’s lymphoma (HL) is a lymphoproliferative<br />

disorder of unknown etiology. In most cases, molecular<br />

markers have suggested a B cell origin of the<br />

tumor cells. However, HL cells exhibit a characteristic<br />

gene expression profile which discriminates<br />

these cells from all other hematopoietic cells. Despite<br />

improvements in the therapy of patients with HL, a<br />

significant number of patients cannot be cured with<br />

current therapy strategies. In addition, toxicity of<br />

the currently used therapy is associated with high<br />

risk for secondary malignancies, cardiac complications<br />

or treatment-induced infertility. Cytotoxic T<br />

lymphocytes (CTL) can kill HL cells and have been<br />

used clinically for treatment of Epstein-Barr viruspositive<br />

HL. For patients with EBV-negative HL this<br />

strategy cannot be employed and alternative target<br />

structures have to be defined. In order to establish<br />

a system for the stimulation of HL-reactive T cells,<br />

we used dendritic cells (DC) as antigen presenting<br />

cells for autologous T cells. Because no specific antigens<br />

for EBV-negative HL are available, we transfected<br />

DC with RNA from established HL cell lines.<br />

After stimulation of peripheral blood mononuclear<br />

cells (PBMC) with RNA-transfected DC we assessed<br />

the reactivity of primed PBMC by interferon gamma<br />

ELISPOT. When we transfected DC (HLA-A*02,24;<br />

B*15; Cw*03,07; DRB1*04,11; DQB1*03) with RNA<br />

from HL cell line HDLM-2 (HLA-A*01,02; B*08,44;<br />

Cw*05,07; DRB1*13,15; DQB1*06) and used these<br />

DC for priming of the PBMC from the same DC-donor,<br />

we observed that the stimulated cells reacted<br />

with HDLM-2 cells but not with irrelevant tumor<br />

cells (TE671 sarcoma cells) or autologous PBMC. Interestingly,<br />

these HDLM-2-primed cells also reacted<br />

with HL cell line KM-H2 (HLA-A*11,24; B*52,15;<br />

Cw*04,12; DRB1*04,11; DQB1*03) but not with HL<br />

cell lines L-1236 (HLA-A*02; B*51; Cw*02; DRB1*14;<br />

DQB1*05) or L-540 (HLA-A*03,11; B*51; Cw*02,15;<br />

DRB1*04,11; DQB1*03). Because HDLM-2 cells and<br />

KM-H2 cells have completely different HLA haplotypes<br />

and KM-H2 cells share 6/10 HLA molecules<br />

with the used PBMC, these observations suggest<br />

the presence of HL-specific antigens expressed in<br />

both HL cell lines. These antigens are presented in<br />

the context of HLA class-I molecules of both cell<br />

lines leading to the T cell response. By analysis of<br />

Gene Expression Omnibus (GEO) microarray data<br />

sets from HL cell lines and primary HL samples<br />

in comparison with normal tissues und normal<br />

B cells we identified HL-associated transcripts.<br />

Among these transcripts we found known cancer<br />

testis (CT) antigens (e.g. PRAME (preferentially<br />

expressed antigen in melanoma) or CT45) and<br />

new antigens with CT-like expression pattern, e.g.<br />

ZBTB32 (zinc finger and BTB domain containing<br />

32). These antigens might be targets for future HLspecific<br />

immune therapy or for monitoring of HLdirected<br />

immune responses.<br />

121


077 Feger | New targets & new leads<br />

Analysis of HPV L1 specific T-cell immunity<br />

Thomas Feger 1 , Andreas Kaufmann 2 , Hans-Georg Rammensee 1 , Stefan Stevanovic 1<br />

1 Eberhard Karls University, Immunology, Tuebingen<br />

2 Charite Campus Benjamin Franklin, Gynecology, Berlin<br />

122<br />

There are over 200 different types of the human<br />

papillomavirus (HPV) described. Especially types<br />

16 and 18, but also types 31 and 45, are associated<br />

with cervical cancer. Types 16 and 18 alone are<br />

thought to be responsible for about 70% of all cervical<br />

cancer. Preventive vaccines based on L1 protein<br />

virus-like particles were licensed in 2007. Although<br />

the antibody response against L1 has been well documented,<br />

not much is known about the T-cell response<br />

to the viral L1 protein. Identification of class<br />

I and class II specific T-cell epitopes is essential for<br />

our understanding of HPV immunity, however.<br />

We used the epitope prediction algorithm SYFPEIT-<br />

HI to identify HPV 6, 11, 16 and 18 epitope candidates<br />

from the viral L1 protein. Class I and class II<br />

epitope candidates were synthesized and screened<br />

for their capability of memory T-cell activation with<br />

IFN-γ ELISPOT and intracellular cytokine staining<br />

using in vitro stimulated PBMCs of healthy donors.<br />

The identified specificities were used to select and<br />

clone epitope reactive T cells.<br />

Several novel class II epitopes from different regions<br />

of the L1 protein of the HPV strains 6, 11, 16 and<br />

18 were identified. T-cell responses against these<br />

epitopes are detectable in healthy donors. Analysis<br />

of the established T-cell clones showed cross<br />

reactivity against T-cell epitopes from strain 6, 11<br />

and 31, and against epitopes from strain 18 and 45,<br />

respectively.<br />

The knowledge of these L1 epitopes enables us to<br />

follow and compare T-cell responses elicited by<br />

vaccination or normal HPV infection. Furthermore,<br />

it explains the protection against additional HPV<br />

strains that are not included in but elicited by the<br />

preventive HPV vaccines.


L123 Kyzirakos | New targets & new leads<br />

Identification of novel EBV-specific MHC class-II epitopes<br />

Christina Kyzirakos 1 , Hans-Georg Rammensee 1 , Stefan Stevanovic 1<br />

1 Universität Tuebingen, Auf der Morgenstelle, Tuebingen, Germany<br />

Epstein-Barr Virus (EBV) persists in the vast majo-<br />

rity of adult individuals (>95%) as a lifelong latent<br />

infection of the host’s B-lymphocyte pool. Although<br />

chronic infections remain asymptomatic in most<br />

cases, immunocompromised patients can suffer<br />

from severe and lifethreatening EBV-associated diseases,<br />

such as post-transplant lymphoproliferative<br />

disorders (PTLD). Thus, immunotherapeutic strategies<br />

using adoptively transferred EBV-specific T<br />

cells are promising. One option is the generation<br />

and expansion of CD4+ and CD8+ T lymphocytes<br />

by using EBV-specific synthetic peptides for the stimulation<br />

of pre-existing memory T cells. Aim of<br />

our study was to identify a set of MHC class-II peptides<br />

which could be recognized by specific CD4+<br />

T helper cells of basically every individual.<br />

15 amino acid long candidate epitopes of nine immunodominant<br />

antigens, three of the latent cycle<br />

(EBNA3A, LMP1 and LMP2) and six of the lytic<br />

cycle (BMLF1, BMRF1, BRLF1, BZLF1, BNRF1 and<br />

BLLF1) of the virus were predicted using the SYF-<br />

PEITHI epitope prediction programme (www.syfpeithi.de).<br />

For each antigen promiscuitive peptides<br />

with high SYFPEITHI scores were tested for immunogenicity<br />

using an IFN-γ-ELISPOT. PBMCs of<br />

at least 15 healthy, randomly chosen blood donors<br />

were cultured for 12 days in the presence of each<br />

candidate peptide. Functional and phenotypic analysis<br />

of T cells of several donors was performed by<br />

multicolor flow cytometry.<br />

Although 76 out of 107 tested peptides could be<br />

identified as T-cell epitopes, only five of them were<br />

defined as immunodominant, as more than 50% of<br />

tested blood donors showed peptide-specific T cell<br />

responses. So far, 19 of the tested peptides could be<br />

identified as MHC class-II epitopes.<br />

In conclusion, we could identify several new EBVspecific<br />

MHC class-II epitopes which can be used<br />

for promising immunotherapeutic approaches for<br />

the treatment of EBV-associated diseases like PTLD,<br />

as well as for diagnostic purposes.<br />

123


L125 Huijbers | New targets & new leads<br />

Vaccination against the extra domain-B of fibronectin as a<br />

novel tumor therapy<br />

Elisabeth J. M. Huijbers 1 , Maria Ringvall 1 , Julia Femel 1 , Sebastian Kalamajski 1 , Agneta<br />

Lukinius 2 , Magnus Åbrink 1 , Lars Hellman 3 and Anna-Karin Olsson 1<br />

1 Department of Medical Biochemistry and Microbiology, Uppsala University, BMC, SE-751 23 Uppsala, Sweden<br />

2 Department of Medical Cell Biology, Uppsala University, BMC, SE-751 23 Uppsala, Sweden<br />

3 Department Cell and Molecular Biology, Uppsala University, BMC, SE-751 24 Uppsala, Sweden<br />

124<br />

During physiological angiogenesis (e.g. wound<br />

healing and proliferation of the endometrium),<br />

embryonic development and tumor angiogenesis<br />

certain extra domains of the extracellular matrix<br />

molecules fibronectin and tenascin-C are inserted<br />

into the parent molecule by alternative splicing<br />

leading to the formation of different isoforms.<br />

Splice variants of both fibronectin and tenascin-C<br />

are often co-expressed specifically around neovasculature<br />

and tumor vasculature but have low or no<br />

expression in normal adult tissue, rendering them<br />

specific targets for anti-tumor therapy.<br />

In our approach we have targeted the extra domain-<br />

B (ED-B), which is a 91 amino acid domain alternatively<br />

spliced into fibronectin and highly conserved<br />

between species. It is identical in mouse, human,<br />

rabbit, dog, monkey and several other species. To<br />

evoke an immune response against the self-antigen<br />

ED-B we used a recombinant fusion protein consisting<br />

of a bacterial thioredoxin (TRX) part fused<br />

with the extra domain B (ED-B), termed TRX-EDB.<br />

This fusion protein has been injected together<br />

with Freund’s adjuvant, a strong immunostimulator,<br />

into eight-weeks old female wild type C57bl6<br />

mice. Mice were boostered twice in a period of five<br />

weeks before they were inoculated subcutaneously<br />

with T241 fibrosarcoma cells, a tumor type known<br />

to express ED-B. After a tumor growth period of<br />

three weeks animals were sacrificed and blood and<br />

tumors were removed. Nineteen out of 20 vaccinated<br />

mice responded with production of anti-ED-B<br />

antibodies and showed a 70% reduction in tumor<br />

size compared to control animals injected with<br />

Freund’s adjuvant and vehicle, lacking anti-ED-<br />

B antibodies. Staining of murine grade III glioma<br />

tissue, to see whether the serum from TRX-EDB<br />

mice could detect native ED-B, showed an extensive<br />

vascular staining pattern compared to normal brain<br />

tissue, which is devoid of ED-B. This shows that<br />

the anti-ED-B antibodies are able to detect native<br />

tissue ED-B. Quantification of tumor necrotic area<br />

revealed a tendency towards a greater necrotic area<br />

in TRX-EDB injected compared to control animals.<br />

Further investigation of the tumors with electron<br />

microscopy revealed morphological changes of the<br />

tumor vasculature of animals with anti-ED-B antibodies,<br />

which was consistent with an immune<br />

response towards the tumor vasculature expressing<br />

ED-B. Moreover in tumors from animals with<br />

anti-ED-B antibodies an increased number of infiltrating<br />

neutrophils was observed, compared to<br />

controls, confirming an immune attack of the vasculature.<br />

Furthermore we detected an increased<br />

amount of extravasated fibrinogen, indicative of<br />

vascular leakage, in tumors of animals with anti-<br />

ED-B antibodies compared to controls.<br />

Therefore ED-B and possibly other tumor vascular<br />

antigens such as the extra domain A of fibronectin<br />

or the extra domain C of tenascin-C alone or in<br />

combination are interesting candidate targets for<br />

treatment of solid tumors. Since the choice of adjuvant<br />

is of major importance for treatment success<br />

in humans, we addressed this issue in a second<br />

study and found a non-toxic biodegradable adjuvant<br />

as potent as Freund’s in inducing an immune<br />

response against a self-antigen (see poster by Julia<br />

Femel et al.)


125


126<br />

Tumor biology & interaction with the immune system


078 Maccalli | Tumor biology & interaction with the immune system<br />

Definition of the immunological properties of cancer stem cells<br />

isolated from human glioblastoma<br />

Cristina Maccalli 1 , Tiziano Di Tomaso 1 , Ena Wang 2 , Stefania Mazzoleni 3 , Gloria Sovena 1 ,<br />

Soldano Ferrone 4 , Rossella Galli 3 , Francesco M. Marincola 2 , Giorgio Parmiani 1<br />

1 Unit of Immuno-Biotherapy of Solid Tumors, San Raffaele Foundation Scientific Institute, Milan, Italy<br />

2 Department of Tranfusion Medicine, National Institutes of Health, Bethesda, MD, USA<br />

3 Neural Stem Cell Unit, San Raffaele Foundation Scientific Institute, Milan, Italy<br />

4 Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, USA<br />

The main objectives of our study is to assess<br />

whether cancer stem cells (CSCs) isolated from glioblastoma<br />

multiforme (GBM) patients can be exploited<br />

as source of antigens to elicit T cell-mediated<br />

immune responses and their validation to design<br />

immunotherapeutic protocols for GBM. We have<br />

characterized, by immunofluorescence and cytofluorimetric<br />

or confocal microscopy analysis, the<br />

immune profile of 14 CSC and of 7 FBS tumor line<br />

pairs isolated from GBM samples. Moreover, PBMCs<br />

isolated from GBM patients were in vitro stimulated<br />

with autologous CSCs and the specific reactivity of<br />

T lymphocytes against GBM CSCs was evaluated by<br />

IFN-g, Granzyme B release (ELISPOT) or CD107a<br />

mobilization. We have assessed that both GBM CSC<br />

and their FBS-cultured non-CSC pairs showed a<br />

low immunogenic profile (MHC, NKG2DL and APM<br />

molecules). Up-regulation of most of these molecules<br />

was induced by IFNs or 5-Aza CdR, though<br />

more efficiently in FBS than in CSCs. Notably, T<br />

cell responses, both CD4+- and CD8+-mediated,<br />

against GBM could be raised in one GBM patient by<br />

stimulating in vitro PBMCs with autologous CSCs<br />

pre-treated with IFN-β. However, TH2-mediated<br />

responses with more efficient recognition of FBS<br />

tumor cell than CSCs, could be found in 3 additional<br />

GBM patients. One key finding of this study<br />

is that CSCs but not FBS cells inhibit allogeneic T<br />

cell proliferation. This immune-inhibitory activity<br />

mediated by GBM CSCs was not dependent on the<br />

secretion of TGFβ1 or 2, that were preferentially<br />

down-modulated in CSC lines, nor on IL-10 and<br />

IL1-3 that were undetectable in the supernatant of<br />

these cell lines. Expression of the indoleamine 2,3dioxygenase<br />

(IDO), a molecule implicated in the generation<br />

of immune tolerance, was detected in both<br />

CSCs and FBS tumor cell lines with high increase<br />

following IFN-g treatment of the cells. Furthermore,<br />

we found that both CSCs and FBS tumor cells<br />

expressed immune response inhibitory molecules,<br />

such as CTLA-4, PD-1 and PDL-1. Furthermore, a<br />

differential gene signature that was confirmed at<br />

the protein levels for some immunological-related<br />

molecules was also found between CSC and FBS<br />

lines. Moreover, this latter analysis provided information<br />

of some candidate molecules associated<br />

with CSCs that we are functionally investigating.<br />

Altogether, these results indicate a lower immunogenicity<br />

and higher suppressive activity of GBM<br />

CSC compared to autologous FBS lines and the need<br />

to identify immunomodulatory agents that can efficiently<br />

restore the expression of immunogenic molecules<br />

on CSCs.<br />

127


079 Poschke | Tumor biology & interaction with the immune system<br />

Immature immunosuppressive CD14+HLA-DR-/low cells in<br />

melanoma patients are Stat3hi and over-express CD80,<br />

CD83 and DC-Sign<br />

Isabel Poschke 2 , Dimitrios Mougiakakos 2 , Johan Hansson, Giuseppe V. Masucci 3 ,<br />

Rolf Kiessling 3<br />

1 Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden<br />

2 I.P. and D.M. contributed equally to this study<br />

3 G.V.M. and R.K. contributed equally to this study as principal investigators<br />

128<br />

Myeloid derived suppressor cells (MDSC) have<br />

emerged as key immune-modulators in various<br />

tumor models and human malignancies, but their<br />

characteristics in humans remain to be unequivocally<br />

defined.<br />

In this study, we have examined CD14+HLA-DR-/<br />

low MDSC in advanced 34 malignant melanoma<br />

(MM) patients. Their frequency in peripheral<br />

blood is significantly increased and associated with<br />

disease activity. Contrary to the common notion<br />

that MDSC are a heterogeneous population of exclusively<br />

immature cells, we find co-expression<br />

of markers associated with mature phenotype.<br />

We demonstrate for the first time over-expression<br />

of CD80, CD83 and DC-SIGN in human MDSC.<br />

Further, increased levels of Stat3, an important regulator<br />

in MDSC development and function, were<br />

noted in patient-derived CD14+HLA-DR-/low cells.<br />

Stat3 was altered towards an active, phosphorylated<br />

state in the HLA-DR- population of CD14+ cells<br />

and was more reactive to activating stimuli in patients.<br />

The described MM-MDSCs utilize arginase<br />

in conjunction with other undefined mechanisms<br />

to suppress CD4+- and CD8+-T-cells. Several observations<br />

suggest a redox imbalance in MDSC and<br />

indicate an important role of Stat3-dependent oxidative<br />

stress in MDSC-mediated T-cell suppression.<br />

These results emphasize the diversity of MDSC in<br />

human cancer and provide potential targets for therapeutic<br />

interventions.


080 Flores-Guzman | Tumor biology & interaction with the immune system<br />

Potential cancer stem cells in ret transgenic mouse model of<br />

spontaneous melanoma<br />

Fernando Flores-Guzman, Dirk Schadendorf and Viktor Umansky<br />

German Cancer Reserarch Center (DKFZ), Heidelberg, Germany<br />

The cancer stem cell (CSC) hypothesis suggests that<br />

neoplastic clones are maintained exclusively by a<br />

rare fraction of tumor cells with stem cells properties.<br />

CSC could represent disseminated dormant tumor<br />

cells without clinical signs of progression. We used a<br />

transgenic mouse spontaneous melanoma model, in<br />

which after a short latency, around 25% of all transgenic<br />

mice developed skin tumors with metastases<br />

in distant organs like liver and lungs. We found that<br />

CD133+ melanoma cells represent a small subset in<br />

primary skin tumors and lymph node metastases<br />

(< 1.5%). The amount of CD133+ melanoma cells<br />

both in primary skin tumors and lymph node metastases<br />

correlated with the primary tumor weight<br />

and the stage of tumor progression. We detected<br />

TRP2+CD133+ single melanoma cells in the bone<br />

marrow of tumor bearing mice suggesting thereby<br />

that these cells could be potential melanoma stem<br />

cells. We also found CD20, CD24, CD44 and CD166<br />

expression in primary tumors, which correlated<br />

with the tumor progression. Another potential melanoma<br />

stem cell marker nestin was express in most<br />

melanoma cells, whereas the multidrug resistance<br />

ABCB5 marker was expressed on less than 3% cells<br />

from primary skin tumors.<br />

In conclusion, our data demonstrate an existence<br />

of the subpopulation of CD133+ melanoma cells in<br />

primary skin tumors, metastatic lymph nodes and<br />

in the bone marrow of tumor bearing transgenic<br />

mice and suggest that these subsets could be potential<br />

CSC.<br />

129


081 Lion | Tumor biology & interaction with the immune system<br />

Poly(I:C)-electroporated myeloid leukemic cell lines become<br />

highly susceptible to NK cell-mediated killing and phagocytosis<br />

by immature DC<br />

Eva Lion 1 , Sébastien Anguille 1 , 2 , Evelien Smits 1 , Zwi Berneman 1 , 2 , Viggo Van Tendeloo 1<br />

1 Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium<br />

2 Laboratory of Experimental Hematology, Antwerp University Hospital, Antwerp, Belgium<br />

130<br />

Natural killer (NK) cells and dendritic cells (DC) are<br />

proven to exert important functions in anti-tumor<br />

defence. Targeting both immune cells for immunebased<br />

therapy is currently advised. We recently<br />

showed that electroporation (EP) of acute myeloid<br />

leukemic (AML) cell lines with the synthetic double-stranded<br />

RNA TLR3-ligand poly(I:C) enhances<br />

their capacity to act on DC and NK cells. Aside from<br />

the fact that tumor cells themselves become apoptotic<br />

and start to secrete marked amounts of type<br />

I interferon (IFN), they are able to mature DC and<br />

promote NK cell IFN-γ. In this study, we explored the<br />

effect of poly(I:C)-EP AML cell lines on the killing<br />

capacity of NK cells and the phagocytic potential<br />

of DC. Additionally, we assessed these functionalities<br />

in three-party cocultures to see for functional<br />

cross-talk. We hypothesized that poly(I:C)-induced<br />

apoptosis and the presence of IFN-α would increase<br />

the NK cell cytotoxicity. In turn, this could lead to<br />

an increased tumor cell up-take by DC.<br />

All experiments were performed with fresh autologous<br />

monocyte-derived immature DC and purified<br />

NK cells of healthy donors. The K562 and U-937<br />

leukemic cell lines were used as targets. Flow cytometric<br />

detection of cytotoxicity and phagocytosis<br />

was acquired simultaneously. Cytotoxicity was determined<br />

based on the viability (annexin V- propidium<br />

iodide-) of PKH67-labeled AML cells. Phagocytosis<br />

of PKH67+ tumor cells by violet-labeled DC<br />

was expressed as the % PKH67+violet+ cells of all<br />

violet+ DC, selected for single cells.<br />

Our data demonstrate that poly(I:C)-EP of AML<br />

cell lines results in an increased susceptibility to<br />

NK cell-mediated killing (p=0.0023 for K562 and<br />

p


082 Bonertz | Tumor biology & interaction with the immune system<br />

Antigen-specific Treg in colorectal carcinoma control T cell<br />

responses against a limited tumor antigen repertoire<br />

Andreas Bonertz 1 , Jürgen Weitz 2 , Kim Pietsch 1 , Christoph Schlude 1 , Simone Jünger 1 , Yingzi<br />

Ge 1 , Kashayarsha Khazaie 3 , Moritz Koch 2 , Philipp Beckhove 1<br />

1 Translational Immunology Unit, The German Cancer Research Center, Heidelberg, Germany<br />

2 Department of Visceral Surgery, University Hospital of Heidelberg, Heidelberg, Germany<br />

3 Division of Gastroenterology, Northwestern University Feinberg School of Medicine,<br />

Robert Lurie Comprehensive Cancer Center, Chicago, Illinois, USA<br />

Previous studies suggest that regulatory T cells<br />

(Treg) may suppress a tumor-specific immune<br />

response against established tumors in patients.<br />

Here, we examined how a depletion of Treg influences<br />

the immune reactivity of colorectal carcinoma<br />

(CRC) patients against tumor associated antigens<br />

(TAA) and screened for the specificity of Treg<br />

in the same patients.<br />

We analyzed the T cell (TC) repertoires from the<br />

peripheral blood (PB) of 170 CRC patients for the<br />

presence and frequencies of spontaneously induced<br />

effector/memory TC against synthetic polypeptides<br />

spanning immunogenic regions from 9 different<br />

TAAs (CEA, EGFR, Heparanase, Her2/neu, Mage-3,<br />

Muc-1, p53, Survivin, Telomerase) before and after<br />

depletion of Treg by short-term IFNγ enzyme-linked<br />

immunospot analysis. In comparison, we assayed<br />

PB from 32 healthy donors for the presence of<br />

TAA-specific TC. Patients and healthy donors were<br />

divided into HLA-A2 positive and negative cohorts.<br />

Furthermore, we evaluated for the first time the<br />

specificities of Treg in the same patients for the respective<br />

TAAs. For this, dendritic cells were pulsed<br />

with TAA-peptides or IgG control antigen, coincubated<br />

with autologous Treg and analyzed for the<br />

Treg capacity to suppress in an antigen-dependent<br />

manner the proliferation of polyclonally activated<br />

conventional TC.<br />

In the majority of patients (60%), TAA specific TC<br />

were detected. The TAA recognition pattern was<br />

highly diverse within the CRC patients and was<br />

independent of the patients HLAtype. Healthy individuals<br />

contained significantly less TAA-reactive<br />

TC. After Treg depletion, the proportion of TAAreactive<br />

patients increased to 86%. The strongest<br />

increases of recognition were found for Muc-1 (14%<br />

vs. 39%), EGFR (13% vs. 38%), and CEA (18% vs.<br />

39%), which was also associated with an increase<br />

of the frequencies of TC specific for these antigens.<br />

Treg reacted specifically for individual TAAs in the<br />

PB of 40% of CRC patients.<br />

Treg-induced suppression of proliferation was most<br />

prevalently observed after Treg activation with<br />

Muc-1, HER2/neu, and CEA, whereas Treg specific<br />

for p53 were not observed. Interestingly, specificities<br />

of memory TC and Treg markedly differed<br />

in the majority of patients, leaving the possibility<br />

of using selected sets of TAA for tumor vaccinations<br />

that induce optimal effector TC responses but<br />

minimal Treg activity.<br />

131


083 Ge | Tumor biology & interaction with the immune system<br />

tumor-induced emigration of antigen-specific treg enables<br />

the bone marrow to foster spontaneous anti-tumor t-cell<br />

responses in breast cancer patients<br />

Yingzi Ge 1 , Christoph Domschke 2 , Helge Philipp Frebel 3 , Christopher Schnappauf 4 , Michael<br />

Hillier 1 , Florian Schuetz 2 and Philipp Beckhove 1<br />

1 Translational Immunology Unit, German Cancer Research Center, Heidelberg, Germany;<br />

2 Department of Obstetrics and Gynecology, Heidelberg University Women’s Hospital<br />

3 Institute of Microbiology, ETH Zurich, Zurich, Switzerland<br />

4 Heidelberg University Children’s Hospital<br />

132<br />

Adequate frequency and activity of tumor-specific<br />

T cells are crucial for successful cancer immunotherapy.<br />

Bone marrow from patients with breast<br />

cancer has been shown to serve as a reservoir of<br />

tumor-responding memory/effector T cells (Tmem/<br />

eff). We detected that a depletion of regulatory T<br />

cells (Treg) unmasked a comparable population of<br />

anti-tumor T cells in the blood to that in the bone<br />

marrow, as determined with defined tumor-associated<br />

antigens (TAA) as well as tumor-cell lysates<br />

in IFN-γ ELISpot assay. The higher reactivity of<br />

TAA-specific Tmem/eff in the bone marrow is due<br />

to firstly, a remarkably lower frequency of resident<br />

Treg, as shown by FACS analysis, and secondly,<br />

a significantly reduced suppressive capacity of<br />

Treg upon TAA activation in vitro. Interestingly,<br />

we also found that T-cell tumor-responsiveness in<br />

the bone marrow inversely correlated to the frequency<br />

of intratumoral Treg, suggesting a mobilization<br />

of activated Treg into the blood. In support<br />

of this, we could identify a panel of tissue-homing<br />

receptors which are up-regulated on activated Treg<br />

derived from the bone marrow in comparison to<br />

those from the blood. The frequency of Treg expressing<br />

those breast tumor-relevant chemokine receptors<br />

was associated with the latency of Tmem/<br />

eff anti-tumor reactivity in the circulation. Collectively,<br />

we propose that tumor-specific Treg in the<br />

bone marrow acquire migratory and suppressive<br />

capacity upon activation and sequentially home to<br />

tumor tissue. This recruitment in turn endows the<br />

bone marrow a unique environment devoid of TAAspecific<br />

Treg and capable of fostering anti-tumor<br />

Tmem/eff.


084 Chen | Tumor biology & interaction with the immune system<br />

Disturbed NK cell function in CML patients at diagnosis does<br />

not recover under treatment with imatinib mesylate<br />

Christiane I-U. Chen 1 , Steffen Koschmieder 2 , Linda Kamp 2 , Holden T Maecker 3 , Wolfgang<br />

Berdel 2 , Heribert Jürgens, 1 Peter P Lee 4 , Claudia Rössig 1<br />

1 Department of Pediatric Hematology and Oncolocy, University Hospital Muenster, Germany<br />

2 Department of Medicine, Division of Hematology and Oncology, University Hospital Muenster, Germany<br />

3 Human Immune Monitoring Center, Stanford University School of Medicine, USA<br />

4 Department of Medicine, Division of Hematology, Stanford University, USA<br />

Even though the tyrosine kinase inhibitor imati-<br />

nib mesylate (imatinib) has become the first-line<br />

therapy for patients with chronic myeloid leukemia<br />

(CML), allogeneic hematopoietic stem cell transplantation<br />

(HSCT) remains the only curative treatment.<br />

The important contribution of the immune<br />

system to curing CML is evidenced by the potent<br />

therapeutic effects of donor lymphocyte infusions.<br />

In addition to cytotoxic T cells, natural killer (NK)<br />

cells may be involved in immunologic control of the<br />

disease. Here, we used a transgenic tet-off inducible<br />

CML mouse model as well as peripheral blood<br />

samples from CML patients to explore the role of<br />

NK cells in CML.<br />

Splenic lymphocyte subpopulations from CML<br />

mice were analyzed by flow cytometry. Under continuous<br />

tetracycline treatment (tet-on), these mice<br />

have a bcr-abl-negative phenotype, whereas withdrawal<br />

of tetracycline induces bcr-abl transgene<br />

expression, resulting in initiation of leukemia and<br />

manifestation of a CML-like phenotype (tet-off, bcrabl+).<br />

Bcr-abl+ mice had significantly decreased<br />

relative numbers of NK cells compared to control<br />

mice (5.5% vs 13.8%, p = 0.045). While ex vivo expansion<br />

of purified NK cell populations from tet-on<br />

and tet-off mice demonstrated similar proliferative<br />

responses to stimulation with high-dose interleukin-2<br />

(IL-2), the cytolytic capacity of NK cells expanded<br />

from bcr-abl+ mice against K-562 cells was<br />

significantly reduced (10.7% vs. 27.3% at an E:T<br />

ratio of 1:1, p < 0.05).<br />

To address the functionality of NK cells in human<br />

CML, we quantified NK cells in peripheral blood<br />

samples from 13 patients at diagnosis and at several<br />

time points during imatinib-induced remission<br />

(range 10-59 months). Compared to age-matched<br />

healthy controls, NK cells were decreased at diagnosis<br />

(4,4% vs 13%, p < 0.05) and did not recover<br />

to normal levels under imatinib treatment (6.2% vs<br />

10%, p < 0.05). Functional experiments revealed<br />

reduced expansion of NK cells in response to coincubation<br />

with K-562/4-1BBL/mbIL-15 stimulator<br />

cells in CML patients versus healthy controls<br />

(26-fold expansion vs 48-fold expansion within 10<br />

days, p < 0.05). Moreover, the cytolytic activity of<br />

human CML NK cells was reduced both at diagnosis<br />

(8.3% lysis of K-562 target cells at an E:T ratio<br />

of 1:1) and under treatment with imatinib (11%)<br />

compared to control (18.9%, p < 0.05).<br />

We conclude that patients with CML have both<br />

quantitative and qualitative defects within their<br />

NK cell compartment, which can be reproduced<br />

by transgenic bcr-abl expression in mice. NK cells<br />

do not recover to normal levels and normal function<br />

under imatinib treatment, even after obtaining<br />

complete remission. Further work will aim at<br />

identifying the underlying mechanisms of NK cell<br />

deficiency in CML, and the development of strategies<br />

to exploit NK cells for immunotherapy of the<br />

disease.<br />

133


085 Dubrot | Tumor biology & interaction with the immune system<br />

Treatment with anti-CD137 mAbs causes intense<br />

accumulations of liver T cells without selective antitumor<br />

mmunotherapeutic effects in this organ<br />

Juan Dubrot 1 , , Francisca Milheiro 1 , Carlos Alfaro 1 , Asis Palazón 1 , Ivan Martinez-Forero 1 ,<br />

Aizea Morales-Kastresana 1 , María C. Ochoa 1 , Sandra Hervás Stubbs 1 , Maria Jure-Kunkel 2 ,<br />

Lieping Chen 3 and Ignacio Melero 1<br />

1 Centro de investigación médica aplicada (CIMA)<br />

2 Bristol Myers-Squibb Pharmaceutical research institute, Princeton, NJ<br />

3 Sidney Kimmel Cancer Center. Johns Hopkins Medical School. Baltimore, BA<br />

134<br />

Background/Aims: Cancer therapy with agonist<br />

anti-CD137 mAbs has been shown to induce im-<br />

mune-mediated tumor rejections in mice and equi-<br />

valent agents of this kind are currently being tested<br />

in cancer patients. Previous reports indicated that<br />

CD137 stimulation induces polyclonal infiltrates of<br />

T lymphocytes in the liver. This study characterizes<br />

the liver infiltrates, the target-dependency of the<br />

phenomena and addresses the question of whether<br />

tumors nested in the liver are a more favourable<br />

target for CD137-based immunotherapy.<br />

Methods: Liver infiltrates were studied with conventional<br />

histology and multiple colour flow cytometry<br />

of total liver leukocytes. CD137-/- mice, mice<br />

with a single rearrangement of the TCR (OT-1 mice)<br />

and Rag-/- mice were used to clarify molecular requirements.<br />

Mice implanted with MC38 colon carcinomas<br />

either subcutaneously or inside the liver<br />

were used for comparative studies under treatment<br />

with agonist anti-CD137 mAbs.<br />

Results: CD137 treatment caused mononuclear<br />

inflammation in portal spaces of the liver, which<br />

gave rise to moderate increases in transaminases<br />

without signs of cholestasis. Marked increases in<br />

the numbers of CD8+ T cells were observed, including<br />

CD8+ T lymphocytes co-expressing CD11c.<br />

Infiltrates were absent in CD137-/- mice and mitigated<br />

in mice harbouring a single transgenic TCR on<br />

their CD8 T cells. Despite of the tumor-independent<br />

accumulation of T cells in the liver, immunotherapeutic<br />

effects were not more prominent against<br />

tumors located in this organ.<br />

Conclusions: Target-dependent effects of CD137<br />

stimulation lead to liver infiltration with T cells,<br />

but lymphocyte enrichment in this organ does not<br />

privilege this site for immunotherapeutic effects<br />

against transplanted tumors.


086 Chachibaia-Saginashvili | Tumor biology & interaction with the immune system<br />

Role of T & B-cells in malignant transition from chronic<br />

autoimmune inflammation during Sjogren ’s syndrome and<br />

treatment approach<br />

Tamar Chachibaia 1 , Ekaterina Aptsiauri 2<br />

1 Georgian Sjogren Association, Medical School AIETI, Department of Immunology, Tbilisi, Georgia<br />

2 Bristol Myers-Squibb Pharmaceutical research institute, Princeton, NJ<br />

3 Rare Diseases Alliance Georgia, Tbilisi<br />

Sjogren’s syndrome (SS) is a common systemic<br />

autoimmune disease, which was first described in<br />

1933 by Henrik Sjögren. It occurs as an isolated disorder<br />

(primary form), also known as the Sicca syndrome,<br />

or more often in association with another<br />

autoimmune disease (secondary form). Women are<br />

affected in 90% of cases, mostly in their peri/post<br />

menopause period of life.<br />

SS is characterized by dysfunction and destruction<br />

of the lachrymal and salivary glands associated<br />

with lymphocytic infiltration and immunological<br />

hyperreactivity. The decrease in tears and saliva<br />

(sicca syndrome) is the result of histo-pathologically<br />

evidenced lymphocytic infiltration and fibrosis<br />

of this exocrine glands. The infiltrate contains<br />

predominantly activated CD4+ helper T cells and<br />

some B cells, including plasma cells that secrete<br />

antibody locally.<br />

As might be expected from the presence of autoantibodies,<br />

a variety of functional abnormalities<br />

has been detected in the T cells, B cells, and macrophages.<br />

The development of parotid lymphoma<br />

in patients with SS are thought to represent endpoints<br />

of multi-step process of local antigen-driven<br />

chronic inflammation that is characterized by<br />

organ-specific B- or T-cell proliferation, polyclonality,<br />

oligoclonality and, eventually, monoclonality.<br />

Of interest are studies that indicate the presence of<br />

a monoclonal B-cell population within the salivary<br />

glands that gives rise to monoclonal immunoglobulins<br />

or light chains in the serum.<br />

About 5% of patients with Sjögren's syndrome<br />

will develop some form of lymphoid malignancy.<br />

(Tzioufas AG, Voulgarelis M (2007). Patients with<br />

Sjögren's syndrome have higher rate an approximately<br />

40-fold higher risk of developing lymphoid<br />

malignancies compared to both patients with<br />

other autoimmune diseases and healthy people.<br />

(Tzioufas AG & Voulgarelis M, 2007). Patients with<br />

severe cases are much more likely to develop lymphomas<br />

than patients with mild or moderate cases.<br />

(Smedby et al, 2006). The most common lymphomas<br />

are salivary extranodal marginal zone B cell<br />

lymphomas or non-Hodgkin lymphoma (Voulgarelis<br />

M. & Skopouli FN 2007) and diffuse large B-cell<br />

lymphoma (Smedby et al, 2006).<br />

The gain in knowledge regarding the cellular mechanisms<br />

of T and B lymphocyte activity in the<br />

pathogenesis of Sjögren’s syndrome (SS) and the<br />

current availability of various biological agents<br />

(anti-TNF-α, IFN-α, anti-CD20, and anti-CD22) have<br />

resulted in new strategies for therapeutic intervention.<br />

Currently, B cell-directed therapies seem to<br />

be more promising than T cell-related therapies.<br />

However, large, randomized, placebo-controlled<br />

clinical trials are needed to confirm the promising<br />

results of these early studies.<br />

135


087 Lutz-Nicoladoni | Tumor biology & interaction with the immune system<br />

Reinforcement of Cancer Immunotherapy by Adoptive<br />

Transfer of cblb-deficient Cytotoxic T Lymphocytes<br />

combined with a Dentritic Cell Vaccine<br />

Christina Lutz-Nicoladoni 1,2 , Patrizia Stoitzner 3 , Magdalena Pircher 1,2 , Stephanie Wallner<br />

1,2 , Thomas Gruber 4 , Anna Maria Wolf 1,2 , Günther Gastl 1,2 , Josef Penninger 5 , Gottfried<br />

Baier 4 , Dominik Wolf 1,2<br />

1 Tyrolean Cancer Research Institute, Tumorimmunology, Innsbruck, Austria<br />

2 Medical University Innsbruck, Internal Medicine V, Hematology and Oncology, Innsbruck, Austria<br />

3 Medical University Innsbruck, Department of Dermatology and Venerology, Austria<br />

4 Medical University Innsbruck, Department for Medical Genetics, Molecular and Clinical Pharmacology, Austria<br />

5 Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria<br />

136<br />

During the last decades rapid progress in immuno-<br />

logy facilitated the development of tumor therapy<br />

based on cell transfer. Autologous tumorinfiltrating<br />

lymphocytes (TILs) are transferred after ex<br />

vivo clonal expansion or ex vivo transduction with<br />

tumor antigen specific T cell receptors (TCR) and<br />

subsequent expansion into patients. Adoptive T<br />

cell transfers (ATC) are often combined with lymphodepleting<br />

therapies. However, the success of<br />

cancer immunotherapy is limited by potent endogenous<br />

immune-evasion mechanisms, which are<br />

centrally mediated by transforming growth factor<br />

beta TGF-β. The RING E3 ubiquitin ligase casitas B<br />

cell lymphoma (Cbl-b) is a negative key regulator<br />

of T cell activation. Cblb-deficient T lymphocytes<br />

are uncoupled from the requirement of CD28 costimulation,<br />

are anergy-resistent and resistant to inhibition<br />

by TGF-β. As a consequence, cblb-deficient<br />

animals reject tumors via cytotoxic CD8+ T cells<br />

(CTLs), which makes Cbl-b an ideal target in adoptive<br />

T cell transfer therapy.<br />

Here we provide evidence that cblb-/- CTLs are<br />

hyper-responsive to TCR/CD28-stimulation in vitro<br />

and protected from the negative effects induced by<br />

TGF-β. Nevertheless and unexpectedly, adoptive<br />

transfer of polyclonal, non TCR-transgenic cblb-deficient<br />

CTLs into tumor bearing immunocompetent<br />

wildtype (wt) mice is not sufficient to reject B16ova<br />

or EG7 tumors in vivo. Thus, cblb-deficient transferred<br />

CD8+ T cells were in vivo re-activated by a<br />

dendritic cell (DC) vaccine (i.e. SIINFEKL-pulsed<br />

DC). In strict contrast to ATC monotherapy, this approach<br />

now delayed tumor outgrowth and significantly<br />

increased survival rates, which is paralleled<br />

by an increased CTL infiltration rate to the tumor<br />

site as well as enrichment of tumor antigen-specific<br />

and interferon-gamma (IFN-g)-secreting CTLs in<br />

the draining lymphnodes. Moreover, in vivo cytolytic<br />

activity was increased in tumor bearing mice<br />

treated with DCs and cblb-deficient CTLs compared<br />

to those treated with DCs plus wt CTLs.<br />

In summary, we provide experimental evidence that<br />

genetic inactivation of Cbl-b in polyclonal, non-TCR<br />

transgenic adoptively transferred CTLs serves as a<br />

novel “adjuvant approach”, suitable to augment the<br />

effectiveness of established anti-cancer immunotherapy<br />

in immuno-competent recipients.


088 Schmid | Tumor biology & interaction with the immune system<br />

Development of a novel transgenic mouse model for melanoma<br />

Beate Schmid 1 , Danielle Arnold-Schild 1 , Mustafa Diken 2 , Christine Tertilt 1,3 , Markus Radsak<br />

4 , Hansjörg Schild 1<br />

1 Johannes Gutenberg University Medical Center, Dept. of Immunology,<br />

Langenbeckstr. 1, 55131 Mainz, Germany<br />

2 Johannes Gutenberg University Medical Center, III. Dept. Medicine, Hematology/Oncology,<br />

Obere Zahlbacher Str. 63, 55131 Mainz, Germany<br />

3 Johannes Gutenberg University Medical Center, Dept. of Pediatrics,<br />

Langenbeckstr. 1, 55131 Mainz, Germany<br />

4 Johannes Gutenberg University Medical Center, III. Dept. Medicine, Hematology/Oncology,<br />

Langenbeckstr. 1, 55131 Mainz, Germany<br />

Novel cancer therapeutics need to be evaluated<br />

in animal models before application in humans.<br />

Models using transplanted tumor cell lines may lead<br />

to tumor growth in mice, but their characteristics<br />

often substantially differ from autochthonously developing<br />

tumors in vivo. This may be important for<br />

the development of tumor tolerance and the establishment<br />

of anti-tumor immunity. Therefore, new<br />

animal models are needed where the endogenous<br />

development of tumors can be controlled and manipulated.<br />

To create an autochthonous tumor model<br />

we generated BAC transgenic mice with inducible<br />

expression of the melanoma oncogenes BrafV600E,<br />

Cdk4R24C and Mitf under the control of the melanocyte<br />

specific tyrosinase promoter. Furthermore<br />

we introduced the gene for firefly luciferase for monitoring<br />

of oncogene expression by bioluminescent<br />

imaging. Transcription of these genes is controlled<br />

by the tamoxifen inducible recombinase CreERT2<br />

also expressed specifically in melanocytes. In vitro<br />

studies of C22 cells transfected with the vector<br />

pcDNA3.1 containing the oncogene-luciferase expression<br />

construct indeed showed an increase in<br />

Braf and Cdk4 expression by western blot as well<br />

as luciferase activity indicating the functionality<br />

of the construct. The pronucleus injection of the<br />

final BAC construct resulted in three founders confirmed<br />

by PCR and southern blot analysis. Analysis<br />

of these mouse lines in vivo by bioluminescent<br />

imaging showed some base line luciferase activity<br />

in one mouse line indicating leakiness of the trans-<br />

genic construct. However, tamoxifen treatment<br />

greatly enhanced the luciferase activity indicating<br />

that our construct is operating as intended. Further<br />

analyses to characterize the tissue specific transgene<br />

expression and kinetics of tumor development<br />

are ongoing. After completion of these basic studies<br />

this novel mouse model will be a valuable tool for<br />

the preclinical assessment of novel cancer therapeutics.<br />

137


089 Sha | Tumor biology & interaction with the immune system<br />

Influence of activation of human immune effector cells<br />

on the penetration into tumor spheroids<br />

Weixiao Sha 1 , Franziska Hirschhaeuser 1 , Stefan Walenta 1 , Kirsten Dettmar 2 and Wolfgang<br />

Mueller-Klieser 1<br />

1 Institute of Physiology and Pathophysiology, University Medical Center of the Johannes Gutenberg-University<br />

Mainz, Germany<br />

2 Fresenius Biotech GmbH, Munich, Germany<br />

138<br />

The trifunctional bispecific antibody catumaxo-<br />

mab (anti-EpCAM x anti-CD3) was designed for<br />

anti-tumor immunotherapy by targeting the tumor-associated<br />

antigen human EpCAM (epithelial<br />

cell adhesion molecule) on tumor cells and human<br />

CD3, expressed on T lymphocytes. In addition, catumaxomab<br />

recruits simultaneously Fcγ receptorpositive<br />

accessory cells, to promote the formation<br />

of a so-called “tri-cell-complex” which finally leads<br />

to the destruction of tumor cells by immune cells.<br />

MCTS (multicellular tumor spheroids) of the FaDu<br />

cell line (over-expressing EpCAM) were co-cultured<br />

with PBMCs (peripheral blood mononuclear cells)<br />

and incubated with the test agents in microtiter<br />

plates. With the objective to compare the influence<br />

of the different binding sites of catumaxomab we<br />

additionally used the F(ab`)2 fragments of catumaxomab<br />

lacking a Fc-region and BiLu (anti-human<br />

EpCAM x anti-mouse CD3) which is unable to bind<br />

to human T cells. Furthermore, LPS (Lipopolysaccharid)<br />

and con A (Concanavalin A) were applied<br />

to the test system. To asses the immune cell infiltration,<br />

rapidly frozen spheroids were cryosectioned<br />

and stained for immunohistology. Antibodies<br />

against CD45, CD14, CD2, CD4 and CD8 were used<br />

to discriminate different subtypes of infiltrating<br />

leukocytes in MCTS. Cytokine release (IFN γ, TNF<br />

α, IL 2, IL 6) gave further information on the type of<br />

immune response. Besides immunological parameters,<br />

the metabolic effects of the treatment with the<br />

different agents were analyzed by measurement of<br />

the extracellular lactate production in co-culture<br />

supernatants.<br />

Catumaxomab-mediated stimulation of PBMCs led<br />

to a massive penetration of spheroids by CD2+ T<br />

cells after 3 days and the volumes of the MCTS<br />

were significantly decreased. CD8+ T lymphocytes<br />

were identified as the dominating subtype of tumor<br />

infiltrating T cells. Higher levels of IL-2 through catumaxomab<br />

application were detected after 1 day,<br />

while no IL-2 was detectable after 3 days. Incubation<br />

with the F(ab`)2 fragment of catumaxomab resulted<br />

in similar results. Catumaxomab stimulation<br />

of co-cultures also resulted in increased concentrations<br />

of extracellular lactate. On the other side, IFN<br />

γ and TNF α secretion as well as lactate concentration<br />

was elevated in PBMC cultures stimulated<br />

with catumaxomab in the absence of tumor cells,<br />

whereas incubation with F(ab`)2 showed no detectable<br />

effect. These catumaxomab-mediated effects<br />

were enhanced in the presence of PBMC and tumor<br />

cells. Furthermore, BiLu did not reveal any effects<br />

in co-cultures of tumor cells with human PBMC.<br />

In summary, it was demonstrated that the catumaxomab-mediated<br />

stimulation of PBMCs in MCTS<br />

co-culture plays a pivotal role for tumor infiltration<br />

by cytotoxic T lymphocytes and secretion of antitumoral<br />

cytokines, thus underscoring the importance<br />

of the trifunctional mode of action for the<br />

initiation of an effective anti-tumoral response.<br />

Supported by Fresenius Biotech GmbH and TRION Pharma GmbH<br />

(Munich, Germany).


090 Strothmeyer | Tumor biology & interaction with the immune system<br />

Comparative Analysis of Predicted HLA Binding of<br />

Immunoglobulin Idiotype Sequences Indicates T Cell-Mediated<br />

Immunosurveillance in Follicular Lymphoma<br />

Anna-Maria Strothmeyer, Katja Zirlik, Marcus Dühren-von Minden, Marcelo Navarrete,<br />

Kristina Heining-Mikesch, Hendrik Veelken<br />

Department of Hematology/Oncology, University Medical Center Freiburg, 79106 Freiburg, Germany<br />

Active immunization against the individual B cell<br />

receptor ("Idiotype", Id) of B cell lymphomas can<br />

induce Id-specific immune responses (see accompanying<br />

poster by Navarrete et al.). Epitope mapping<br />

experiments with Id peptides show that these in vivo-induced<br />

T cell responses are directed preferentially<br />

against individual Id epitopes located in CDR,<br />

whereas FR and C region epitopes are ignored. We<br />

therefore speculated that Id-specific T cell immunity<br />

might occur naturally in lymphoma patients,<br />

and conducted a reverse immunology study of the<br />

Id in 39 follicular lymphoma (FL) patients. Clonal<br />

Id transcripts were identified from tumor biopsies<br />

by A-PCR. HLA-A and B alleles were determined<br />

by serotyping and high-resolution PCR genotyping.<br />

Potentially HLA-presentable nonameric peptides<br />

from all Ids and their corresponding germ-line (GL)<br />

VH and VL genes were identified for all analyzable<br />

HLA alleles of this cohort by reverse immunology<br />

(bimas.cit.nih.gov). Identified peptides were<br />

ranked according to their predicted score, and the<br />

sum of the scores for the 20 highest ranking peptides<br />

was calculated for each HLA allele. For every<br />

patient's HLA type, the sum scores of all Ids on<br />

his/her HLA alleles were added to yield a theoretical<br />

measure of every Id's immunogenicity. This<br />

"autologous" sum score was compared to the mean<br />

sum scores of all other 38 idiotypes in a matchedpair<br />

analysis. Separate analyses were performed for<br />

CDR peptides (containing at least 2 AA in any CDR)<br />

vs. non-CDR-peptides (as defined by imgt.cines.fr)<br />

and Id vs. corresponding GL sequences.<br />

The sum scores of Id VH sequences were lower for<br />

the patient's own Id than for the mean of the allogeneic<br />

Ids (p=0.0017). Every CDR (CDR1: p


091 Ramacher | Tumor biology & interaction with the immune system<br />

Immunosuppression in transgenic mouse melanoma<br />

model induced by myeloid derived suppressor cells<br />

Marcel Ramacher 1 , Michal Baniyash 2 and Viktor Umansky 1<br />

1 German Cancer Research Center (DKFZ), Heidelberg Germany<br />

2 Lautenberg Center for General and Tumor Immunology, Jerusalem, Israel<br />

140<br />

Melanoma is known for its poor response to current<br />

immunotherapies due to immunosuppressive cells<br />

in the tumor microenvironment like myeloid derived<br />

suppressor cells (MDSC). We performed the studies<br />

on the ret transgenic mouse model of malignant<br />

melanoma, which resembles human melanoma as<br />

regards to histopathology and clinical development.<br />

After a short latency, 25% of mice develop melanoma<br />

metastasizing to lymph nodes, lungs and liver.<br />

We found that MDSC from tumor bearing mice were<br />

characterized by increased numbers and higher<br />

nitric oxide (NO) production and arginase-1 (ARG1)<br />

expression than those from tumor free mice or nontransgenic<br />

littermates. This phenomenon could be<br />

due to exosomes released by melanoma cells in<br />

vivo. In fact, upon treatment of non-transgenic littermates<br />

with melanoma derived exosomes, MDSC<br />

amounts were significantly elevated.<br />

To investigate the effect of NO and reactive oxygen<br />

species produced by MDSC on CD4+CD25- conventional<br />

(helper) T cells and CD4+CD25+ regulatory<br />

T cells (Treg), these subsets were separated from<br />

spleens and treated with NO donor DETA-NONOate<br />

or hydrogen peroxide in vitro. Both factors induced<br />

much higher apoptosis in conventional T cells than<br />

in Tregs suggesting thereby the Treg resistance<br />

to the immunosuppressive microenvironment. To<br />

reduce the MDSC immunosuppressive effect and<br />

to improve antitumor immune responses, tumor<br />

bearing mice were treated orally with an inhibitor<br />

of phosphodiesterase (PDE)-5 sildenafil (Viagra).<br />

Lower NO production and ARG1 expression in<br />

MDSC was correlated with the inhibition of melanoma<br />

progression. We suggest that an effective<br />

immunotherapy should include the inhibition of<br />

MDSC immunosuppressive functions.


092 Tomsitz | Tumor biology & interaction with the immune system<br />

Establishment of a pre-clinical NOD/LtSz-scid IL2Rgammac null<br />

(NSG) mouse model to evaluate immunotherapeutic strategies<br />

for acute myeloid leukemia (AML)<br />

Dirk Tomsitz 1 , Ariane Brunk 1 , Marion Nonn 1 , Alexander Hohberger 1 , Shamsul A. Khan 1 ,<br />

Eva Distler 1 , Matthias Theobald 1 , Wolfgang Herr 1 , Udo F. Hartwig 1<br />

1 Dept. of Medicine III - Hematology and Oncology, Johannes-Gutenberg-University Medical Center,<br />

Mainz, Germany<br />

Introduction: Acute myeloid leukemia (AML) is a<br />

cancer of the myeloid line of blood cells, characte-<br />

rized by the clonal expansion of immature mye-<br />

loblasts potentially initiating from rare leukemic<br />

stem cells. Xenotransplantation of human AML into<br />

immunodeficient mice is essential for establishing<br />

a preclinical model to i) assess the potency of modified<br />

donor lymphocyte grafts to induce immunotherapeutic<br />

graft-versus-leukemia (GVL)-reactivity<br />

and ii) to study the properties of leukemic stem<br />

cell biology. Here we report the successful engraftment<br />

of 8 out of 14 patient-derived, primary AML<br />

samples in NOD/LtSz-scid IL2Rgammacnull (NSG)<br />

mice within 2-12 weeks, with a mean of 0,2-40%<br />

human leukemic cells in the bone marrow (BM). In<br />

first studies, we also show the successful eradication<br />

of engrafted AML-blasts using AML-reactive,<br />

HLA-mismatched T lymphocytes.<br />

Methods: Mice were used unconditioned or irradiated<br />

with 150 cGy 4-16h prior to intravenous (i.v.)<br />

transfer of 5x104-1x107 primary AML cells. Engraftment<br />

kinetics and dose-dependencies were analyzed<br />

by means of clinicopathological criteria and<br />

flow cytometry. AML-reactive T cells were generated<br />

in a HLA-mismatched setting over 3 weeks from<br />

PBMCs isolated from buffy coats. One week after<br />

transplantation of 5x105 AML cells from patient<br />

MZ667, AML-bearing mice were i.v. injected with<br />

5x106 CD8+ leukemia-reactive T lymphocytes.<br />

Results: Stable AML-engraftment in NSG recipients<br />

was detected upon transplantation of 8 out of 14<br />

primary AML samples by determing the percentage<br />

of CD33+/CD45+ cells in blood, spleen and BM.<br />

Expression of Flt3 mutations appeared to promote<br />

engraftment of AML-samples. In addition to leukemic<br />

blasts, we frequently observed co-engraftment<br />

of human T cells present in the AML-graft, possib-<br />

ly facilitated due to irradiation of recipients. In 4<br />

cases, stable AML engraftment was inhibited by<br />

T cell outgrowth and induction of xenoreactivity.<br />

However, in general irradiation augmented the<br />

engraftment efficacy of AML. Thus, we attempted<br />

to circumvent T cell growth by a) depleting T cells<br />

from AML-samples using MACS-technology® or b)<br />

treating animals with immunosuppressiva such as<br />

Cyclosporin A, Mycophenolatemofetil and Tacrolimus.<br />

Immunomagnetic T cell depletion resulted in<br />

stable but decelerated AML engraftment without<br />

T cell outgrowth, whereas preliminary results indicated<br />

that treatment of transplanted mice with<br />

immunsuppressiva abrogate engraftment of both<br />

AML and T cells. Finally, we started to explore our<br />

model for investigating GVL-responses of AMLreactive<br />

human T cell lines generated by repetitive<br />

stimulation of healthy donor lymphocytes with<br />

AML blasts. In first studies using HLA-mismatched<br />

AML MZ667-reactive CD8+ T cells we could demonstrate<br />

that following adoptive transfer of T<br />

lymphocytes into AML MZ667 bearing mice, leukemic<br />

blasts could be completely eradicated, whereas<br />

in controls stable engraftment was detectable in the<br />

BM.<br />

Conclusions: These results suggest that NSG mice<br />

represent a valuable tool for the establishment of a<br />

preclinical AML model to evaluate the immunotherapeutic<br />

GVL-reactivity of ex vivo modified T cellgrafts<br />

as well as to analyze properties of leukemia<br />

stem cell biology.<br />

141


093 Koslowski | Tumor biology & interaction with the immune system<br />

Tumor-associated genomic DNA hypomethylation<br />

effects positive autoregulation of HIF-1α<br />

Michael Koslowski 1 , Ulrich Luxemburger 1 , Özlem Türeci 2 , Ugur Sahin 1<br />

1 TRON gGmbH, Langenbeckstr., 55131 Mainz<br />

2 Ganymed Pharmaceuticals AG, Freiligrathstr.12, 55131 Mainz<br />

142<br />

Hypoxia-inducible factor 1α (HIF-1α) is frequently<br />

overexpressed in human cancers and controls the<br />

expression of several genes that have been implicated<br />

in tumor growth and progression. Activity of<br />

HIF-1α in cancer cells is regulated at the transcriptional,<br />

translational and posttranslational level by<br />

multiple inter- and co-acting molecular pathways.<br />

We show for the first time that tumor-associated<br />

genomic DNA hypomethylation facilitates positive<br />

autoregulation of HIF-1α, resulting in amplification<br />

of hypoxia-induced transactivation of HIF-1α target<br />

genes. Applying bisulfite sequencing, methylationspecific<br />

PCR, transcription factor binding assays,<br />

and chromatin immunoprecipitation we found that<br />

the HIF-1α promoter harbors a hypoxia-response<br />

element that is normally repressed by methylation<br />

of a CpG dinucleotide located in the core element.<br />

However, in colon cancer cell lines and in primary<br />

colon cancer samples we found frequent demethylation<br />

of this element, enabling binding and positive<br />

autoregulation of HIF-1α. Our results provide novel<br />

and highly unexpected insights into the complexity<br />

of HIF-1α regulation in cancer cells and implicate<br />

that tumor-associated genomic DNA hypomethylation<br />

is a positive factor for HIF-1α mediated effects<br />

on malignant cell growth.


094 Quaglino | Tumor biology & interaction with the immune system<br />

A miRNAs expression profiles during ErbB2 driven mammary<br />

carcinogenesis<br />

Elena Quaglino 1 , Maddalena Arigoni 1 , Elisabetta Ercole 1 , Federica Riccardo 1 , Guido Forni 1 ,<br />

Raffaele Calogero 2 and Federica Cavallo 1<br />

1 Molecular Biotechnology Center (MBC), University of Torino, Italy<br />

2 Bioinformatics and Genomics Unit, Department of Clinical and Biological Science, University of Torino, Italy<br />

MicroRNAs (miRNAs) are noncoding RNAs that<br />

regulate global gene expression. They often act<br />

synergistically to repress target genes, and their<br />

deregulation can contribute to the initiation and<br />

progression of a variety of cancers. In view of the<br />

roles played by miRNAs in cancer progression, we<br />

performed a miRNAs microarray analysis aimed at<br />

identifying the modulation of miRNAs expression<br />

profiles during the progression of authoctonous<br />

mammary carcinomas arising in mice transgenic<br />

for the activated transforming rat ErbB2 oncogene<br />

(BALB-neuT mice). BALB-neuT mice constitute<br />

a suitable cancer-prone model, since inexorably<br />

the females develop an invasive and metastatic<br />

mammary cancer in all of their ten mammary<br />

glands with a step wide pattern and a systemic metastatic<br />

spread similar to that observed in human<br />

mammary cancer. Using Applied Biosystems<br />

Megaplex low density miRNA expression arrays<br />

miRNAs expression signature of diffused atypical<br />

hyperplasia (6 week-old BALB-neuT versus 6 weekold<br />

BALB-c females mammary glands) and invasive<br />

carcinoma (19 week-old BALB-neuT versus 19<br />

week-old BALB-c females mammary glands) were<br />

identified. PCA shows a wide difference in miRNAs<br />

expression between diffused atypical hyperplasia<br />

and invasive carcinoma. Moreover, rank product<br />

analysis allowed the detection of 6 and 4 miRNAs<br />

whose expression is correlated to invasive carcinoma<br />

and to diffused atypical hyperplasia respectively.<br />

Among those that were found to be upregu-<br />

lated in invasive carcinoma miR-135a and miR-135b<br />

were found to be expressed also in several lines<br />

derived from BALB-neuT mammary tumors, suggesting<br />

their role in mammary cancer progression.<br />

By contrast, the expression of miR-741 resulted<br />

undetectable in all the mouse cell lines derived<br />

from BALB-neuT mammary tumors, even if, its expression<br />

was upregulated in mammary BALBneuT<br />

tumors, suggesting its role in the tumor microenvironment<br />

phenotype. Modulation (overexpression<br />

or down-modulation) of miR-135b in breast cancer<br />

cells is under investigation in order to evaluate its<br />

role in the in vivo tumor growth, in vitro matrigel<br />

invasion as well as in vivo lung metastasis formation.<br />

143


095 Lanzardo | Tumor biology & interaction with the immune system<br />

Attenuation of PI3K/Akt-mediated tumorigenic signals through<br />

PTEN activation by DNA vaccine-induced anti-ErbB2<br />

antibodies<br />

Stefania Lanzardo 1 , Alessandra Porzia 2 , Arianna Citti 2 , Federica Cavallo 1 , Guido Forni 1 ,<br />

Angela Santoni 2 , Ricciarda Galandrini 2 , Rossella Paolini 2<br />

1 Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of Turin, Italy<br />

2 Department of Experimental Medicine, Institute Pasteur-Fondazione Cenci Bolognetti,<br />

Sapienza University, Rome, Italy<br />

144<br />

Vaccination of BALB/c mice with a plasmid enco-<br />

ding for the extracellular (EC) and transmembrane<br />

(TM) domains of rat ErbB2 (EC-TMneu) protects<br />

from a lethal challenge of ErbB2+ tumor cells<br />

by eliciting both a cell mediated and a humoral<br />

immune response. ECTMneu vaccination also<br />

leads to the inhibition of spontaneous mammary<br />

tumors in rat ErbB2 transgenic mice mainly by eliciting<br />

anti-rat ErbB2 Abs capable of inhibiting in<br />

vitro the expansion of ErbB2+ cells. However, the<br />

molecular mechanisms underlying Ab-induced retardation/rejection<br />

of tumor growth have not been<br />

elucidated.<br />

By studying BALB/c mice deficient in immune components<br />

we show that the protective immunity to<br />

rat ErbB2+ tumors rests on the antibody response<br />

elicited by the electroporation of a DNA vaccine<br />

encoding the extracellular and transmembrane<br />

domains of rat ErbB2. In vivo the adoptive transfer<br />

of vaccine-elicited anti-rat ErbB2 antibodies<br />

protected against a challenge of rat ErbB2+ carcinoma<br />

cells (TUBO cells). In vitro such antibodies<br />

inhibited TUBO cell growth by impairing cell cycle<br />

progression and inducing apoptosis. To correlate<br />

intrinsic mechanisms of antibody action with their<br />

tumor-inhibitory potential, first we showed that<br />

TUBO cells constitutively express phosphorylated<br />

transgenic ErbB2/autochthonous ErbB3 heterodimers,<br />

and exhibit a basal level of Akt phosphorylation<br />

suggesting a constitutive activation of the<br />

PI3K/Akt pathway. The treatment with anti-ErbB2<br />

antibodies caused a drastic reduction of the basal<br />

level of Akt phosphorylation in the absence of an<br />

impairment of PI3K enzymatic activity. Notably,<br />

the same antibody treatment induced an increase<br />

of PTEN phosphatase activity that correlated with a<br />

reduced PTEN tyrosine phosphorylation. In conclusion,<br />

vaccine-induced anti-ErbB2 antibodies directly<br />

affect the transformed phenotype of rat ErbB2+<br />

tumors by impairing ErbB2-mediated PI3K/Akt signalling.


096 Halama | Tumor biology & interaction with the immune system<br />

Natural Killer Cells are decreased in Human Colorectal Cancer<br />

Tissue and Liver Metastases despite High Levels of<br />

NK-Recruiting Chemokines<br />

Niels Halama 1,2 Monika Braun 3 , Christoph Kahlert 4 , Anna Spille 1 , Christian Quack 3 , Nuh<br />

Rahbari 4 , Moritz Koch 4 , Jürgen Weitz 4 , Inka Zoernig 1 , Peter Schirmacher 5 , Karsten Brand 2 ,<br />

Niels Grabe 2 and Christine S. Falk 3<br />

1 Medical Oncology, National Center for Tumor Diseases, University of Heidelberg, Heidelberg, Germany<br />

2 Hamamatsu Tissue Imaging and Analysis (TIGA) Center, Institute for Medical Biometry and Informatics,<br />

University of Heidelberg, Heidelberg, Germany<br />

3 Immune Monitoring Unit, NCT, DKFZ and Institute for Immunology<br />

Im Neuenheimer Feld 305, 69120 Heidelberg<br />

4 Department of Surgery, University of Heidelberg, Heidelberg, Germany<br />

5 Institute of Pathology, University of Heidelberg, Heidelberg, Germany<br />

Background & aims Tumor infiltrating T lympho-<br />

cytes in colorectal cancer (CRC) have prognostic<br />

impact, but the role of NK cells in CRC tissue<br />

remains unknown. The contribution of intratumoral<br />

cytokines and chemokines in shaping the composition<br />

of the inflammatory lymphocytic infiltrate<br />

is also unclear.<br />

Methods: In this study, localization and densities<br />

of NK and T cells within primary CRC, CRC liver<br />

metastases, and normal tissues were analyzed on<br />

whole tissue sections. In parallel, the most important<br />

cytokines and chemokines were quantified in<br />

paired serum, primary CRC and adjacent mucosa<br />

samples and in CRC liver metastases and correlated<br />

with NK and T cell infiltration, respectively.<br />

Results: The different compartments displayed<br />

characteristic differences like significantly higher<br />

chemokine concentrations in CRC tissue. Most<br />

importantly, despite high local chemokine levels,<br />

NK cells were generally scarce within CRC tumor<br />

tissues, independent of HLA class I expression.<br />

Adjacent normal mucosa contained normal levels of<br />

NK cells. In contrast, corresponding T cell numbers<br />

varied substantially and were positively correlated<br />

with higher chemokine levels.<br />

Conclusions: Our findings indicate a distinct regulation<br />

of NK cells versus T cells in the CRC tumor<br />

microenvironment. NK cell migration into CRC<br />

tumor tissue is obviously impaired early during<br />

tumor development by mechanisms that do not<br />

affect T cell infiltration.<br />

145


097 Fraszczak | Tumor biology & interaction with the immune system<br />

Killer dendritic cells inhibit Treg differentiation<br />

Jennifer Fraszczak 1 , Malika Trad 1 , Nona Janikashvili 1,2 , Daniela Lakomy 1 , Maxime Samson<br />

1 , Sylvain Audia 1 , Julien Vinit 1 , Nicolas Larmonier 2 , Bernard Bonnotte 1<br />

1 CR Inserm 866, Faculty of medicine, 7 boulevard Jeanne d’Arc, 21000 Dijon, France<br />

2 Department of Paediatrics, Steele Children's Research Centre, University of Arizona, Tucson, AZ 85724, USA<br />

146<br />

Known for decades as the principal messengers of<br />

the immune system, dendritic cells (DC) play a critical<br />

role in the initiation and regulation of immune<br />

responses against tumor. We have recently reported<br />

on the non-conventional direct tumor killing activity<br />

of DC. We have demonstrated that mouse DC generated<br />

from bone marrow precursors are endowed<br />

with the capability to kill multiple types of cancer<br />

cells and have identified peroxynitrites as the main<br />

effector molecules underlying this cytotoxic activity.<br />

Importantly these killer DC (KDC) are capable<br />

of presenting tumor antigens from the cancer cells<br />

they had killed to specific T cells thereby inducing<br />

an efficient antitumor immune response. We here<br />

evaluate whether KDC may modulate regulatory T<br />

cells (Treg), critical contributors of cancer-induced<br />

immune tolerance and major obstacles for tumor<br />

immunotherapy. Our results indicate that KDC significantly<br />

impair the polarization of naive T cells<br />

into FoxP3-expressing immunosuppressive Treg<br />

but foster their differentiation into T bet-expressing<br />

Th-1 cells. The cellular and molecular bases<br />

responsible for this negative modulation of Treg by<br />

KDC is currently under investigation.


098 Trad | Tumor biology & interaction with the immune system<br />

Tumor infiltrating CD11c+ dendritic cells suppress T cell<br />

activation<br />

Malika Trad 1 , Jennifer Fraszczak 1 , Daniela Lakomy 1 , Maxime Samson 1 , Sylvain Audia 1 ,<br />

Julien Vinit 1 , Emmanuel Katsanis 2 , Nicolas Larmonier 2 * and Bernard Bonnotte 1<br />

1 CR INSERM 866, Université de Bourgogne, Dijon, France<br />

2 Department of Pediatrics, Steele Children’s Research Center, University of Arizona, Tucson, USA<br />

* Equal contribution<br />

Many tumors, including breast cancers are infil-<br />

trated by dendritic cells (DC), known primarily for<br />

their capability to induce and sustain anti-tumoral<br />

immune responses. However, a significant deficit<br />

in the function of DC has been reported in cancer<br />

patients and in various animal tumor models,<br />

which may account for tumor escape from the<br />

immune system. Using the murine breast cancer<br />

model 4T1, we have investigated the phenotypical<br />

and functional characteristics of tumor-infiltrating<br />

DC (TIDC). Our results indicate that TIDC, isolated<br />

from 4T1 tumor-bearing Balb/c mice based on the<br />

expression of the marker CD11c, exhibit a mixed<br />

phenotype associating the expression of mature DC<br />

markers (co-stimulatory molecules) and the Gr-1<br />

molecule (a marker of immunosuppressive myeloid<br />

cells). Functionally, TIDC were unable to induce<br />

allogeneic T cell proliferation and produced low<br />

amount of the pro-inflammatory cytokine IL-12,<br />

required for the T cell activation. Conversely, TIDC<br />

secreted the immunosuppressive cytokine IL-10<br />

and were capable of suppressing the proliferation<br />

of T lymphocytes induced with anti-CD3 and anti-<br />

CD28. The mechanism(s) underlying T cell suppression<br />

by TIDC did not depend on nitric oxide,<br />

but may involve the enzyme indoleamine 2,3-dioxygenase<br />

(IDO) which was significantly upregulated<br />

in these cells.<br />

147


099 Hemmerling | Tumor biology & interaction with the immune system<br />

Human Langerhans cells reconstitute in skin xenografts<br />

Julia Hemmerling 1 , Joanna Wegner-Kops 1 , Diana Wolff 1 , Maria Sommer 1 , Eva M. Wagner 1 ,<br />

Esther von Stebut 2 , Udo F. Hartwig 1 , Rudolf E. Schopf 2 , Matthias Theobald 1 , Wolfgang<br />

Herr 1 , Ralf G. Meyer 1<br />

1 Department of Hematology/Oncology and<br />

2 Department of Dermatology, at the University Medical Center of the Johannes Gutenberg-University<br />

Mainz, Germany<br />

148<br />

Dendritic cells (DC) of the skin, e.g. epidermal Lan-<br />

gerhans cells (LC), are potent regulators of T cells.<br />

They therefore are interesting targets for autologous<br />

T-cell stimulation in the context of vaccinestrategies<br />

as well as for the manipulation of allo-reactive<br />

T-cells in graft-versus-host disease (GVHD).<br />

In an attempt to study the biology of human LC in<br />

vivo, we transplanted human skin to immunodeficient<br />

NOD/SCID/γcnull (NSG) mice and studied the<br />

impact of xeno-transplantation on the distribution<br />

of LC in comparison to that of CD11c positive dermal<br />

DC. We analyzed skin xenografts at different time<br />

points after transplantation and found that during<br />

wound healing, LC were absent in grafts prepared<br />

4 to 6 weeks after transplantation. However, in<br />

many animals analyzed beyond week 6, LC were<br />

again present with almost normal distribution.<br />

These findings were re-confirmed by staining with<br />

antibodies against human CD1a, CD207/Langerin,<br />

S100 and HLA-DR. CD11c positive human dermal<br />

cells were also transiently reduced in numbers, but<br />

never vanished from the dermis after xenografting.<br />

In subsequent experiments, we analyzed the skin<br />

grafts by sequential biopsies and again demonstrated<br />

that more than 50 % of the skin grafts were<br />

devoid of LC in week 6. Three to 5 weeks later,<br />

LC were detected again in these transplants with a<br />

slightly reduced density compared to normal skin.<br />

In congenic mouse models of hematopoietic stem<br />

cell transplantation, murine LC have been shown<br />

to re-populate after local skin injury without the<br />

need of blood-derived precursors. Up to now, there<br />

are only few data supporting this hypothesis for<br />

human LC. By demonstrating the re-establishment<br />

of LC in the xenografts in the absence of any human<br />

hematopoiesis, our data confirm that human LC are<br />

able to re-populate the skin after local depletion.<br />

We further analyzed the proliferative activity of<br />

LC in the xenografts by double staining of CD207/<br />

Langerin-positive cells with Ki67 and found a significantly<br />

increased proliferative activity of LC<br />

compared to healthy skin (31% vs. 5%). Proliferation<br />

is a unique feature of LC among DC. Our data<br />

suggest that this contributes to LC-reconstitution.<br />

In summary, we introduce a human skin xenograftmodel<br />

that allows studying the biology of LC and<br />

dermal DC. Our findings help to unravel the phenomenon<br />

of local LC-reconstitution in human skin.<br />

This model might also help to study the role of skin<br />

DC for inflammatory skin disease and skin GVHD<br />

as well as for vaccine-strategies in vivo.


100 Koop | Tumor biology & interaction with the immune system<br />

Downregulation of cancer/testis antigen 45 (CT45) in<br />

human HT1080 cells analysed by proteomic screening<br />

Anja Koop 1 , Hendrik Schmidt 1 , Melanie Nebendahl 1 , Christoph Gelhaus 2 , Matthias Leippe 2 ,<br />

Ottmar Janssen 1 , Hans-Jürgen Heidebrecht 3<br />

1 Institute of Immunology, UK-SH, Campus Kiel<br />

2 Department of Zoophysiology, University of Kiel<br />

3 Institute of Pathology, UK S-H, Campus Kiel<br />

The cancer/testis antigen (CT) family is defined by<br />

its specific expression pattern. In most cases, CT<br />

antigens are exclusively expressed in germline cells<br />

and in some tumors. However, as for many other<br />

CT antigens, little is known about the function of<br />

CT45. Since various CT antigens induce antigen<br />

specific cellular or humoral immune responses,<br />

they are regarded as vaccine targets for anti-cancer<br />

immunotherapy.<br />

To get an idea about the function of CT45, we<br />

looked for differentially expressed proteins in the<br />

CT45 positive human fibrosarcoma cellline HT1080<br />

after downregulation of CT45. Therefore five different<br />

cellpreparations of scrRNA and siRNA treated<br />

HT1080 were prepared 48h after treatment. Only<br />

cellpreparations with significantly downregulated<br />

CT45 expression were used for these experiments.<br />

In order to determine regulated proteins, lysates<br />

were analysed by 2D-DIGE and differentially expressed<br />

proteins were identified by peptide mass<br />

finger printing. Equal amounts of each sample were<br />

labelled with Cy3 or Cy5. After isoelectric focusing<br />

proteins were separated on 12,5% polyacrylamid<br />

gels (20x24cm). The in gel fluorescence corresponding<br />

to the protein concentration was detected by<br />

an fluorescence scanner. Differentially expressed<br />

proteins were picked, proteolytically cleaved by<br />

trypsin and then subjected to MALDI analysis. One<br />

of the differentially expressed proteins is Annexin1<br />

(ANXA1). In all five cellular lysates of 48h siRNA<br />

treated HT1080 cells it was significantly downre-<br />

gulated. ANAX1 regulates Phospholipase A2 activity<br />

and is considered to play an important role in<br />

the tumorgenesis of breast cancer. Further experiments<br />

will resolve the significance of the ANAX1<br />

downregulation.<br />

The work is sponsored by the Medical Faculty of the CAU Kiel<br />

and the Wilhelm-Sander-Stiftung.<br />

149


101 Karakhanova | Tumor biology & interaction with the immune system<br />

IFN-α up-regulates B7-H1 expression on dendritic cells<br />

and pancreatic tumor cells<br />

Svetlana Karakhanova 1 , Ralf Jesenofsky 2 , Susanne Serba 1 , Alexandr V. Bazhin 1 , Jan Schmidt 1<br />

1 Department of Surgery, University of Heidelberg, Germany<br />

2 II. University Hospital, Mannheim, Germany<br />

150<br />

Pancreatic carcinoma is one of the most aggressive<br />

human malignant tumors and most lethal cancers<br />

worldwide. Despite recent advances in surgery, radiation<br />

and chemotherapy, outcome of pancreatic<br />

cancer could be improved only partially. Therefore<br />

novel approaches are strongly needed. Immunotherapy<br />

represents such an option. Clinical data<br />

show promising results for approaches combining<br />

chemotherapeutics with activating cytokines, like<br />

IFN-α. However, in parallel with immune stimulating<br />

effects of this treatment, the presence and induction<br />

of immunosuppressive mechanisms should<br />

be considered in the course of such a pancreatic<br />

cancer therapy. Regulatory molecules of the B7-H<br />

family, especially B7-H1 (PD-L1, CD274), play an important<br />

role in the regulation of immune responses.<br />

In human pancreatic carcinoma tissue B7-H1 is upregulated<br />

and the patients, demonstrating increased<br />

B7-H1 expression have a poor prognosis. B7-H1<br />

expression in pancreatic tumors contributes to the<br />

tumor immune evasion and tumor progression and,<br />

as recently demonstrated, blocking of B7-H1 improves<br />

anti-tumor effects in a mouse pancreatic cancer<br />

model.<br />

In our study we aim to investigate the expression<br />

and function of B7-H1 molecules in the context of<br />

IFN-α therapy of pancreatic cancer.<br />

Using Flow cytometry, cytological methods as well<br />

as RT-PCR approaches, we showed that B7-H1 expression<br />

is up-regulated on human dendritic cells<br />

(DC) and some human pancreatic cancer cell lines<br />

upon treatment with IFN-α. Interestingly, in some<br />

cell lines we observed additive effect of 5-FU (chemotherapeutic<br />

agents used for pancreatic carcinoma<br />

treatment) and IFN-α on the increase of B7-H1<br />

expression. To confirm the inhibitory potential of<br />

IFN-α -induced B7-H1 molecule, we performed cocultures<br />

of T-Cells and IFN-α-treated DC with and<br />

without blocking of B7-H1 with specific antibodies<br />

and measured proliferation and cytokine production<br />

from T-cells. In ongoing study we are using an<br />

orthotopic mouse model of pancreatic carcinoma to<br />

demonstrate that additional blocking of B7-H1 molecule<br />

during 5-FU and IFN-α combinatory therapy<br />

may additionally improve the outcome of this treatment.<br />

Our data demonstrate that, despite the stimulation<br />

of anti-tumor effects, IFN-α up regulates the expression<br />

of immunosuppressive B7-H1 molecules,<br />

which could limit the effect of immunotherapy<br />

with IFN-α. Thus, it could be of advantage to reduce<br />

undesirable site effects of the IFN-α-induced B7-H1<br />

expression on the tumor and immune cells.


102 Zimmer | Tumor biology & interaction with the immune system<br />

Impact of endoplasmic reticulum aminopeptidases (ERAP)<br />

on T cell epitope generation in melanoma cells<br />

Christin Zimmer 1 , Silke Lubojanski 2 , Tanja Dannenberg 1 , Xhao Fang 1 , Ulrike Seifert 3 , Dirk<br />

Schadendorf 1 , Annette Paschen 1<br />

1 Department of Dermatology, University Hospital Essen, Essen, Germany<br />

2 Department of Medicine III, University Medical Centre, Mainz, Germany<br />

3 Institute for Biochemistry, University Medicine Charite, Berlin, Germany<br />

Tumor cells presenting antigen epitopes by HLA<br />

class I molecules can be specifically recognized and<br />

effectively killed by autologous cytotoxic CD8+ T<br />

lymphocytes (CTLs). Different proteolytic/peptidolytic<br />

components are involved in the intracellular<br />

generation of antigen epitopes. The cytosolic, multi-catalytic<br />

proteasome cleaves the tumor antigen<br />

into peptide fragments of different length, thereby<br />

defining the C-terminus for the majority of peptides<br />

presented by HLA class I molecules. While some<br />

of the proteasome products are of the correct size<br />

for direct binding to HLA class I molecules others<br />

are N-terminally elongated peptide precursors that<br />

require further trimming by aminopeptidases. In<br />

the endoplasmic reticulum of human cells two<br />

aminopeptidases, ERAP1 and ERAP2, have been<br />

identified. Their function in epitope generation by<br />

precursor trimming has been clearly demonstrated.<br />

Thus, the aim of this study is to determine the influence<br />

of ERAPs on the presentation of specific<br />

tumor antigen epitopes by melanoma cells. At first,<br />

the expression level of ERAP1 and ERAP2 in several<br />

cell lines established from different metastases of<br />

melanoma patients was determined by quantitative<br />

real time PCR and Western Blot analysis. All cell<br />

lines tested expressed ERAP1 and ERAP2 at mRNA<br />

and protein level, albeit at different quantities. To<br />

gain further insight into the role of EARP1 in the<br />

generation of specific CTL epitopes, its expression<br />

was down-regulated either by transient transfection<br />

of melanoma cells with siRNA or stable<br />

transfection with a shRNA expression plasmid. In<br />

either case, knockdown of ERAP1 expression was<br />

verified by RT-PCR and/or Western Blot analysis.<br />

Interestingly, ERAP1 down-regulation increased<br />

the recognition of melanoma cells UKRV-Mel-15<br />

by CTLs specific for an epitope (Melan-A26/27-35)<br />

derived from the melanoma differentiation antigen<br />

Melan-A/MART-1. Conversely, overexpression of<br />

ERAP1 decreased the presentation of the Melan-A/<br />

MART-126/27-35 epitope, pointing to a destructive<br />

role of ERAP1 in the processing of this epitope. In<br />

contrast, down-regulation of ERAP1 in Ma-Mel-86a<br />

cells did not affect the stimulation of autologous<br />

CD8+ T cells directed against a mutated neoantigen<br />

(please see abstract by Lubojanski et al.).<br />

Our results point to a more epitope-specific role of<br />

ERAP1 in antigen presentation: ERAP1 activity can<br />

either contribute to or interfere with the generation<br />

of specific epitopes while others might even be generated<br />

independently.<br />

151


103 Heidemann Krogh | Tumor biology & interaction with the immune system<br />

The phospho-regulated calcium-binding cancer-testis antigen<br />

cabyr, interacts with glycolysis-regulating enzymes in lung cancer<br />

cells<br />

Anette Heidemann Krogh, Allan Stensballe 2 , Anne Elbæk, Lotte Hatting Pugholm, Evo Kristina<br />

Søndergaard, Rikke Bæk, Malene Jørgensen, Kim Varming, and Søren Naaby-Hansen<br />

1 Department of Clinical Immunology, Aalborg sygehus, Århus University Hospitals, Aalborg, Denmark<br />

2 Department of Biotechnology, Aalborg University, Aalborg, Denmark<br />

152<br />

Background: CABYR was originally identified as a<br />

human sperm flagellum protein associated with the<br />

fibrous sheath structure and involved in capacitation-induced<br />

hyperactivated motility. Six variants of<br />

CABYR containing two coding regions (CR-A and<br />

CR-B) were cloned from human testis cDNA libraries,<br />

including five variants with alternative splice<br />

deletions. CABYR possesses several motifs and<br />

domains for self-assembly and protein interactions.<br />

Although CABYR transcripts corresponding to the<br />

entire CR-A are testis-specific, small amounts of<br />

minor splice variants are present in motile cilia of<br />

normal human bronchus and fallopian tubes. More<br />

interesting, CABYR expression has recently been<br />

demonstrated in a variety of solid tumours, including<br />

brain, lung, and head & neck squamous cell<br />

cancers.<br />

In testis and sperm CABYR seems to play a role in<br />

the regulation of calcium sequestration and episodic<br />

release, and to form a scaffold with other testisspecific<br />

AKAPs that serve to integrate PKA, Rho and<br />

calcium signalling. CABYR is finally thought to act as<br />

a Ca2+-shuttle in a high order fibrous sheath protein<br />

complex that mediate glycolysis-driven energy<br />

generation in the distal flagellum. While CABYR’<br />

roles in spermatogenesis and gamete function are<br />

emerging, its function in cancer cells remains to be<br />

demonstrated. Here we report recent data from an<br />

ongoing study of CABYR in lung cancer.<br />

Results and Discussion<br />

To define CABYR’ interactions in the lung adeno-<br />

carcinoma cell line A549, immunoprecipitation (IP)<br />

with monoclonal antibodies against CABYR was<br />

combined with western blot (WB) and mass spectrometry<br />

(MS) analysis. Butyrate-induced protein<br />

1, a protein tyrosine phosphatase and the regulatory<br />

subunit of serine/threonine-protein phosphatase<br />

2A, were co-precipitated with CABYR. These<br />

putative interactions are in accordance with our<br />

previous findings of serine- and threonine-phosphorylation<br />

in testicular CABYR and that CABYR’<br />

calcium-binding capacity is regulated by tyrosine<br />

phosphorylation.<br />

More notable, several enzymes involved in the regulation<br />

of glycolysis, including phosphofructokinase,<br />

pyruvate kinase, fructose-biphosphate aldolase,<br />

malate dehydrogenase, and the testis-specific<br />

subunit of lactate dehydrogenase, as well as hypoxia-inducible<br />

factor prolyl hydroxylase 3 and ADP/<br />

ATP translocase 1, also co-purified with CABYR<br />

from A549 cells. The interaction between CABYR<br />

and lactate dehydrogenase was confirmed by bidirectional<br />

IP and WB.<br />

Taken together, these results suggest that CABYR<br />

participates in the regulation of anaerobic glycolysis<br />

and energy generation in lung cancer, and that<br />

CABYR expression is an indicator of aggressive<br />

tumour growth. The regulation of the CABYR interactome,<br />

and cancer cells ability to survive and proliferate<br />

under hypoxic conditions following RNAimediated<br />

downregulation of CABYR, are currently<br />

being investigated.


104 Hansmann | Tumor biology & interaction with the immune system<br />

Isolation of intact genomic DNA from FOXP3-stained and<br />

FACS-sorted regulatory T cells for epigenetic analysis<br />

Leo Hansmann, Christian Schmidl, Tina J. Boeld, Reinhard Andreesen, Petra Hoffmann,<br />

Michael Rehli, Matthias Edinger<br />

Department of Hematology & Oncology, University Hospital Regensburg, 93042 Regensburg, Germany<br />

The CD4+ T cell compartment contains cells of<br />

different lineages and differentiation states that<br />

include Th1, Th2, Th17 and natural regulatory T<br />

(Treg) cells. Due to the lack of exclusive surface<br />

markers for these T cell subpopulations, they can<br />

be differentiated from each other only by their cytokine<br />

secretion profile, their functional characteristics<br />

or, most reliably, their expression lineage defining<br />

transcription factors, including T-bet, GATA-3,<br />

RORC or FOXP3 for Th1, Th2, Th17 and Treg cells,<br />

respectively. We recently showed that stable expression<br />

of the Foxp3 gene in human Treg cells<br />

is regulated in parts by DNA methylation (Baron<br />

et al. Eur J Immunol 2007, Hoffmann et al. Eur J<br />

Immunol 2009, Schmidl et al. Genome Res 2009).<br />

Yet, due to the lack of methods for the isolation<br />

of intact genomic DNA from fixed and intranuclearly-stained<br />

cells we thus far had to rely on surrogate<br />

markers for the identification and isolation<br />

of human Tregs for epigenetic analyses. We now<br />

present a modified phenol-based DNA isolation protocol<br />

permitting the reliable purification of intact<br />

high-molecular genomic DNA for further downstream<br />

applications, such as MALDI-TOF MS based<br />

DNA methylation analysis, that includes bisulphite<br />

conversion and PCR amplification of the extracted<br />

DNA. This method enables us to investigate human<br />

Treg cell epigenetics with the same accuracy as in<br />

murine systems where transgenic reporter mice are<br />

usually used for the unequivocal identification and<br />

isolation of Foxp3+ Treg cells. We present the first<br />

methylation analyses of the FOXP3 locus in human<br />

conventional T- and Treg cells sorted for intranuclear<br />

FOXP3 expression.<br />

153


105 Lupp | Tumor biology & interaction with the immune system<br />

Mechanisms of Treg-mediated suppression<br />

of graft-versus-host disease<br />

Corinna Lupp 1 , Tobias Bopp 1 , Edgar Schmitt 1 , Hansjörg Schild 1 and Markus P. Radsak 1,2<br />

1 Institute for Immunology, Universitätsmedizin of the Johannes Gutenberg-University, Mainz, Germany<br />

2 III. Dept. of Medicine, University Hospital, Johannes Gutenberg-University, Mainz, Germany<br />

154<br />

The therapeutic efficacy of allogenic bone marrow<br />

transplantation for many hematological malignancies<br />

relies on the graft-versus leukemia effect by eliminating<br />

residual leukemic cells. The recognition of<br />

host alloantigens by donor T cells is most likely the<br />

basis of graft-versus-tumor effects, but is oftentimes<br />

also the cause of graft-versus-host disease (GvHD),<br />

a key contributor to the high morbidity and mortality<br />

rates in the context of bone marrow transplantation.<br />

Regulatory FoxP3+ CD4+ T cells (Tregs) are<br />

known to suppress the activation of conventional T<br />

cells (e.g. CD8+ or CD4+ T cells) and represent a<br />

therapeutic paradigm for the control of GvHD.<br />

In our present study we analyzed potential mechanisms<br />

to prevent or attenuate acute graft-versushost<br />

reactions in an allogenic MHC mismatched<br />

transplantation model.<br />

To study the suppressive effects of Tregs in detail we<br />

performed allogenic mixed lymphocyte reactions<br />

(MLR) in vitro and allogenic bone marrow/T cell<br />

transplantations in mice. In transplantation experiments<br />

we used BALB/c mice as recipients which<br />

became irradiated lethally and got a bone marrow<br />

and T cell transplant from C57BL/6 donor mice.<br />

Some mice additionally got Tregs from Interleukin<br />

10 deficient (IL-10 -/-) or the wildtype C57BL/6<br />

donors. The control group which did not get Tregs<br />

developed severe graft-versus-host disease symptoms<br />

after transplantation.<br />

Additionally transplanted Tregs from IL-10 -/- as<br />

well as from wildtype donors could attenuate cli-<br />

nical GvHD symptoms. And also in vitro the Tregmediated<br />

suppression of the alloreactive effector T<br />

cells’ proliferation occurred independent of IL-10.<br />

The analysis of the phenotype of dendritic cells<br />

(DCs) that had been cultured with Treg cells discovered<br />

a reduced expression of the costimulatory<br />

molecules CD 80 and CD 86. We could show that<br />

Tregs communicate with allogenic DCs via direct<br />

cell-to-cell contact, demonstrated by the transfer of<br />

the dye calcein from calcein-labeled Tregs to DCs.<br />

This calcein transfer could partially be prevented<br />

through the preincubation of the Tregs with the gap<br />

junction blocking peptide GAP 27. Only a slight dye<br />

transfer to the conventional T cells was detectable in<br />

the MLR indicating that the Tregs preferentially interact<br />

with the allogenic antigen presenting cells.<br />

In conclusion, our results demonstrate that Tregs<br />

interact with allogenic antigen presenting cells via<br />

gap junction intercellular communication. There<br />

is only a little cell-contact dependent interaction<br />

between the Tregs and the effector T cells and the<br />

cytokine IL-10 seems not to be indispensable for the<br />

inhibition of alloreactivity.<br />

These data provide the basis for future concepts to<br />

manipulate allogenic T cell responses to prevent<br />

GvHD.


106 Castle | Tumor biology & interaction with the immune system<br />

DNA copy number, including telomeres and mitochondria,<br />

assayed using next-generation sequencing<br />

John C. Castle* 1 , Christopher D. Armour 2 , Matthew Biery 2 , David Haynor 3 , Heather Bouzek<br />

3 , Tao Xie 4 , Ronghua Chen 5 , Kira Misura 6 , Stuart Jackson 5 , Jason M. Johnson 5 , Carol A.<br />

Rohl 5 , Christopher K. Raymond 2<br />

1 Institute for Translational Oncology and Immunology (TrOn), Mainz, Germany<br />

2 NuGEN Technologies, Inc., Seattle, Washington, USA<br />

3 University of Washington, Seattle, Washington, USA<br />

4 Pfizer, Inc., San Diego, California, USA<br />

5 Merck Research Laboratories, Boston, Massachusetts, USA<br />

6 Amgen, Inc., Seattle, Washington, USA<br />

DNA copy number aberrations are found in tumors<br />

and are potentially carcinogenic. We sought to<br />

develop an improved lab-automatable, cost-efficient,<br />

accurate platform using next-generation sequencing<br />

to profile nuclear, mitochondrial, and<br />

telomeric DNA copy number. This platform draws<br />

on the unbiased nature of next-generation sequencing<br />

and incorporates techniques developed for<br />

RNA expression profiling. To demonstrate this platform,<br />

we assayed tumor-derived cell line UMC-11<br />

using 5 million 33 nt reads and found tremendous<br />

copy number variation, including regions of single<br />

and homogeneous deletions and amplifications<br />

to 29 copies; 5 times more mitochondria and 4<br />

times less telomeric sequence than a pool of nondiseased,<br />

blood-derived DNA; and that UMC-11<br />

was derived from a male individual. The described<br />

assay outputs absolute copy number, outputs an<br />

error estimate (p-value), and is more accurate than<br />

array-based platforms at high copy number. The<br />

platform enables profiling of mitochondrial levels<br />

and telomeric length. The assay is lab-automatable<br />

and has a genomic resolution and cost that are<br />

tunable based on the number of sequence reads.<br />

155


156<br />

Enhancing immunity & adjuvants


107 Sevko | Enhancing immunity & adjuvants<br />

Paclitaxel in ultra-low doses reduces immunosuppression<br />

in ret transgenic tumor bearing mice<br />

Alexandra Sevko 1 , Michael Shurin 2 , Viktor Umansky 1<br />

1 German Cancer Research Center, Heidelberg, Germany<br />

2 Departments of Pathology and Immunology, University of Pittsburgh Medical Center, Pittsburgh,<br />

Pennsylvania, USA<br />

It has been recently shown that chemotherapeutic<br />

drugs such as paclitaxel, 5-fluoruracil or doxorubicin<br />

applied in ultra-low, noncytotoxic doses stimulate<br />

dendritic cell activity, and induce antitumor<br />

immune responses in mouse transplantable tumor<br />

models.<br />

We studied effects of ultra low-dose paclitaxel on<br />

tumor progression in ret transgenic mouse model<br />

of spontaneous melanoma, which closely resembles<br />

human melanoma regarding histopathology and<br />

clinical development.<br />

We showed that the survival of melanoma bearing<br />

mice upon the treatment was significantly increased<br />

as compared to non-treated group that correlated<br />

with a significant decrease in the tumor weight<br />

in mice from paclitaxel-treated group. To evaluate<br />

the mechanism of this antitumor effect, we focused<br />

on immunosuppressive cells like myeloid derived<br />

suppressor cells (MDSC) and regulatory T cells<br />

(Treg). Paclitaxel was found to decrease the number<br />

of MDSC infiltrating primary tumors and metastatic<br />

lymph nodes. Importantly, the amount of MDSC producing<br />

immunosuppressive agent nitric oxide was<br />

also decreased in these melanoma lesions and in the<br />

bone marrow. In addition, paclitaxel induced a reduction<br />

of Treg infiltrating primary tumors without<br />

affecting the total number of CD4+ T cells.<br />

We suggest that ultra low-dose paclitaxel therapy<br />

can neutralize tumor-induced immune suppression<br />

leading to the delayed tumor progression and prolonged<br />

survival of tumor bearing mice.<br />

157


108 Diaz-Valdés | Enhancing immunity & adjuvants<br />

Transforming growth beta 1 increases monocyte chemotactin<br />

protein-1 and interleukin-10 expression in A375 human<br />

melanoma cells enhancing monocyte migration and impairing<br />

dendritic cell function<br />

Nancy Díaz-Valdés 1 , María Basagoiti 1 , Javier Dotor 2 , Iñaki Monreal 1 , Jose Ignacio Riezu-<br />

Boj 1 , Francisco Borrás-Cuesta 1 , Pablo Sarobe 1 and Esperanza Feijoó 2<br />

1 Universidad de Navarra, Centro de Investigación Médica Aplicada, Area de Hepatología y Terapia Génica,<br />

Pamplona, Spain<br />

2 DIGNA Biotech, Madrid, Spain<br />

158<br />

Melanoma progression is associated to the expres-<br />

sion of different growth factors, cytokines and<br />

chemokines, which collaborate on processes such<br />

as cell proliferation, angiogenesis, metastasis and<br />

immunosuppression. Transforming growth beta 1<br />

(TGFβ1) is a pleiotropic cytokine involved not only<br />

in physiological processes but also in cancer development.<br />

Here we analyzed in melanoma cells<br />

the effect that TGFβ1 has on monocyte chemotactin<br />

protein-1 (MCP-1) and interleukin-10 (IL-10) expression,<br />

two known factors responsible for melanoma<br />

progression. TGFβ1 treatment of A375 human<br />

melanoma cells increased the expression of MCP-1<br />

and IL-10 and this effect was mediated by the crosstalk<br />

between Smad and the AKT and BRAF-MAPK<br />

signaling pathways. Conditioned medium from<br />

TGFβ1-treated A375 cells (TGFβ-MCM) enhanced<br />

MCP-1-dependent migration of monocytes, which<br />

expressed high levels of TGFβ, basic fibroblast<br />

growth factor and vascular endothelial growth<br />

factor mRNA. Concerning IL-10, it had an autocrine<br />

effect on A375 cells, enhancing their proliferation.<br />

Moreover, TGFβ-MCM induced a functional<br />

impairment of dendritic cells, characterized by the<br />

lower production of proinflammatory factors tumor<br />

necrosis factor-alfa and interleukin-12 and upregulation<br />

of immunoregulatory molecules programed<br />

death ligand-1, herpesvirus entry mediator and<br />

cyclooxygenase-2, mediated by IL-10. Additionally,<br />

TGFβ-MCM increased the expression of the inhibitory<br />

molecules indolamine 2, 3-dioxigenasa, and<br />

immunoglobulin-like transcript receptor in DC and<br />

significantly decreased the interferon gamma IFNγ<br />

production by T-cells in mixed leukocyte reaction<br />

assays. Finally, the effect of the 15-mer TGF-β1 inhibitor<br />

peptide P144 (Disitertide) was analysed in<br />

vitro and in vivo. In vitro, P144 inhibited TGF-β1dependent<br />

Smad2 phosphorylation and expression<br />

of MCP-1 and IL-10 in A375 cells. In vivo, treatment<br />

with P144 of A375-tumor-bearing nude mice significantly<br />

reduced tumor volume. These results show<br />

new effects of TGFβ1 on melanoma cells and their<br />

consequences on tumor progression and immunosuppression,<br />

strongly reinforcing the role of this<br />

cytokine as a molecular target in melanoma.


109 Bauer | Enhancing immunity & adjuvants<br />

The TLR9 ligand CpG reduces the suppressive function of<br />

myeloid-derived suppressor cells via interferon alpha<br />

Helen Bauer 1 , Christine Zoglmeier 1 , Daniel Nörenberg 1 , Georg Wedekind 1 , Philipp Bittner 1 ,<br />

Stefan Endres 1 , Carole Bourquin 1<br />

1 Division of Clinical Pharmacology, Ludwig-Maximilian University Munich, Germany<br />

Tumor cells employ a wide range of mechanisms<br />

to evade recognition in tumor-bearing hosts. The<br />

induction and expansion of myeloid-derived suppressor<br />

cells (MDSC), a heterogenous population<br />

of immature, bone-marrow-derived myeloid cells,<br />

is one of these mechanisms. In mice, MDSCs are<br />

commonly defined as Gr1+CD11b+ and display<br />

a remarkable ability to suppress T cell responses<br />

in vitro via different mechanisms such as ROS-induction,<br />

arginase and iNOS activity. In our studies,<br />

we investigated the effect of a stimulation of the<br />

innate immune system with the TLR9 ligand CpG<br />

on the function and phenotype of MDSCs in tumorbearing<br />

mice.<br />

To investigate the effect of TLR9 activation on<br />

MDSCs, we treated C26 tumor-bearing mice with<br />

CpG and analysed the expression of a range of<br />

surface markers by FACS analysis. We show that<br />

a number of markers associated with MDSC maturation<br />

and activation is upregulated by CpG treatment.<br />

Investigation of the suppressive function of<br />

MDSCs revealed reduced suppressivity of MDSCs<br />

from CpG-treated mice. Similar results were obtained<br />

in an orthotopic model of gastric cancer. In<br />

vitro, CpG treatment of isolated MDSCs demonstrated<br />

that the reduction in suppressivity by CpG is<br />

not the result of a direct activation of MDSCs, but<br />

is mediated by cytokines released from other cell<br />

types. Both in in vivo and in vitro experiments we<br />

show that interferon alpha released by plasmacytoid<br />

dendritic cells following CpG treatment plays<br />

a major role in inducing MDSC maturation and reduction<br />

of suppressivity.<br />

In summary, our results show that MDSCs are<br />

strongly affected by TLR9 activation, resulting in<br />

drastic changes in MDSC phenotype and function.<br />

In addition, we provide evidence that interferon<br />

alpha induced by TLR9 activation is a major factor<br />

in promoting MDSC maturation and reduction of<br />

MDSC suppressivity in CpG-treated tumor-bearing<br />

mice.<br />

159


110 Femel | Enhancing immunity & adjuvants<br />

Identification of a potent, non-toxic adjuvant<br />

for use in therapeutic vaccines<br />

Julia Femel 1 , Elisabeth JM Huijbers 1 , Maria Ringvall 1 , Lars Hellman 2 , Anna-Karin Olsson 1<br />

1 Department of Medical Biochemistry and Microbiology, Uppsala University, BMC, SE-75123 Uppsala, Sweden<br />

2 Department of Cell and Molecular Biology, Uppsala University, BMC, SE-75124 Uppsala, Sweden<br />

160<br />

Monoclonal antibody-based therapies have made an<br />

important contribution to current treatment strategies<br />

for cancer and autoimmune disease. However,<br />

their cost-intensive production could possibly lead<br />

to limited availability of the therapy. Therefore obtaining<br />

antibodies produced by the body through<br />

therapeutic vaccination is an interesting alternative,<br />

due to the low amount of required recombinant<br />

protein. To circumvent the tolerance of the<br />

immune system against self-antigens, an efficient<br />

vaccination technology as well as a potent adjuvant<br />

is required. The lack of potent, but at the same time<br />

non-toxic and biodegradable adjuvants that can be<br />

used in the clinic, has been a major limiting factor<br />

for the development of therapeutic vaccines.<br />

We have identified the biodegradable squalenebased<br />

Montanide ISA720 combined with phosphorothioate<br />

stabilized CpG oligo 1826 (Montanide/<br />

CpG 1826) as an adjuvant able to break tolerance<br />

against a self-molecule. A therapeutic vaccine<br />

against tumor angiogenesis has been developed in<br />

our group. This vaccine successfully breaks selftolerance<br />

against the extra-domain B (ED-B) of fibronectin<br />

using Freund’s adjuvant and the recombinant<br />

protein Trx-EDB (see poster by Elisabeth<br />

Huijbers et al.). To compare the immunostimulatory<br />

properties of Freund’s adjuvant and Montanide/CpG<br />

1826, we have vaccinated two groups of<br />

C57BL/6 mice with Trx-EDB in combination with<br />

either Montanide/CpG 1826 or Freund’s adjuvant.<br />

Blood samples were collected regularly and analy-<br />

zed for anti-ED-B antibodies.<br />

Comparing the ability of Montanide/CpG 1826 and<br />

Freund’s adjuvant to stimulate an antibody response<br />

against the extra-domain B of fibronectin revealed<br />

that Montanide/CpG 1826 induced higher anti-ED-<br />

B antibody titers, as well as less variation between<br />

individual animals, than Freund’s. Furthermore<br />

antibodies in animals treated with Montanide/<br />

CpG 1826 were still detectable after eleven months,<br />

while antibodies in mice treated with Freund’s adjuvant<br />

had returned to base levels at this time point.<br />

Analysis of the IgG isotypes showed that IgG1 and<br />

IgG2b were the main isotypes in both treatment<br />

groups. However, preliminary data indicate that<br />

Montanide/CpG 1826 might have the capability to<br />

induce antibodies with higher affinity to ED-B than<br />

antibodies induced with Freund’s adjuvant.<br />

With Montanide/CpG 1826 we have identified a<br />

non-toxic alternative to Freund's adjuvant, which<br />

is at least as potent with respect to inducing an<br />

immune response against a self-antigen. This will<br />

facilitate introduction of therapeutic vaccines to the<br />

clinic.


111 Diekmann | Enhancing immunity & adjuvants<br />

mTOR inhibition by Rapamycin after intranodal<br />

RNA immunization improves the CD8+ memory<br />

T-cell response qualitatively and quantitatively<br />

Jan Diekmann 1 , Sebastian Attig 1 , Sebastian Kreiter 1 , Raouf Selmi 1 , Mustafa Diken 1 , Özlem<br />

Türeci 1 and Ugur Sahin 1,2<br />

1 Universitätsmedizin, Johannes Gutenberg-Universität Mainz, Germany<br />

2 Institute for Translational Oncoclogy (TrOn), Johannes Gutenberg-Universität Mainz, Germany<br />

Memory T cell formation is one of the most im-<br />

portant factors predicting the success of a cancer<br />

vaccine. By inducing a long lasting antigen-specific<br />

T cell memory, residual cancer cells and micro-metastasises<br />

can be targeted and thereby a possible<br />

relapse of the tumour can be prevented. Recent<br />

reports have suggested the key metabolic kinase<br />

mammalian target of rapamycin (mTOR) as an intrinsic<br />

regulator of memory formation in the CD8+<br />

T cell compartment. The inhibitor of mTOR, Rapamycin,<br />

is used in the clinic to suppress immune rejection<br />

in the setting of organ transplants. Paradoxically<br />

it has been shown that Rapamycin treatment<br />

of mice during and after the course of an infection<br />

enhanced the formation and differentiation of the<br />

memory CD8+ T cell pool and also accelerated the<br />

transition from effector to memory cells.<br />

In order to investigate the effect of Rapamycin on<br />

the CD8+ T cell reponse in the setting of intranodal<br />

RNA immunization, we treated C57BL/6J mice<br />

with Rapamycin (Rapamune) for 3 weeks (d10 –<br />

d31) after a course of three RNA immunizations<br />

(d0, d3, d6) with IVT-RNA (20µg) coding for the<br />

OVA-derived SIINFEKL epitope (SIINFEKL-RNA).<br />

Immunized mice treated with the carrier of the drug<br />

served as a control group. During the treatment, the<br />

contraction of the SIINFEKL-specific CD8+ T cells<br />

was almost identical in both groups. However, the<br />

phenotype of the SIINFEKL-specific cells was markedly<br />

different: the Rapamycin-group developed<br />

significantly more CD127+/KLRG1- cells, potential<br />

precursors of long-lived central memory cells. Additionally,<br />

these cells showed a higher proliferative<br />

capacity after a rechallenge with the antigen (SIIN-<br />

FEKL peptide) at day +35. In this study, we show,<br />

for the first time, the beneficiary effect of Rapamycin<br />

on memory T cell differentiation in the setting<br />

of anti-cancer vaccination.<br />

These findings indicate that mTOR inhibitors could<br />

be used to improve the establishment of a longlasting<br />

and potent memory CD8+ T cell population<br />

after intranodal RNA-immunization, which<br />

could augment the outcome after tumour relapse/<br />

therapy.<br />

161


112 Reinis | Enhancing immunity & adjuvants<br />

DNA methyltransferase inhibitors as modulators of anti-tumour<br />

immunity: implications for their use as adjuvants for<br />

immunotherapy<br />

Milan Reiniš, Jana Šímová, Marie Indrová, Romana Mikyšková, Veronika Polláková, Ivan<br />

Štěpánek, Jana Bieblová and Jan Bubeník<br />

Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic<br />

162<br />

Epigenetic changes, such as aberrant DNA methyla-<br />

tion, play an important role in cancer cell deve-<br />

lopment. Notably, they can also be crucial for the<br />

development of tumour cell variants that escape<br />

the immune surveillance. Tumour cells frequently<br />

downregulate the expression of genes involved in<br />

antigen presentation and interactions with immune<br />

cells (e.g. MHC and costimulatory molecules). Expression<br />

of these genes can be silenced by DNA methylation<br />

in tumour cells and thus influence tumour<br />

surveillance. DNA methylation can be blocked by<br />

DNA methyltransferase inhibitors (DNMTi), promising<br />

anti-tumour agents. Recently, we have demonstrated<br />

on various murine MHC class I-deficient<br />

tumour cell lines that DNMTi (5-azacytidine<br />

and 2’-deoxy-5-azacytidine), used either alone or<br />

in combination with histone deacetylase inhibitors,<br />

upregulate expression of antigen-presenting<br />

machinery genes as well as cell surface expression<br />

of MHC class I and other costimulatory/inhibitory<br />

and adhesive molecules. Our further analysis<br />

revealed that DNMTi can have similar effects on<br />

the expression of selected immunoactive genes in<br />

tumour cells as IFNγ.<br />

Further, we have investigated the impact of in vivo<br />

administration of DNMTi on the growth of experimental<br />

MHC class I-deficient tumours, using a<br />

murine model for HPV-16-associated tumours. We<br />

have demonstrated their adjuvant/additive effects<br />

in anti-tumour immunotherapy either with unmethylated<br />

CpG oligodeoxynucleotides or with IL-<br />

12-producing cellular vaccines. Similarly to the in<br />

vitro treatment, administration of DNMTi upregulated<br />

MHC class I and other immune molecules<br />

on tumour cells, inducing their sensitivity to the<br />

CD8+-mediated immune response.<br />

Since in vivo administration of DNMTi can also<br />

affect differentiation and function of immune cells,<br />

we have tested changes in effector and regulatory<br />

cell populations in tumour-bearing animals upon<br />

the treatment. Our data indicate that, despite putative<br />

immunosuppressive effects (dendritic cell maturation<br />

impairment, block of cytokine production<br />

or induction of T regulatory cells), treatment with<br />

DNMTi combined with immunotherapy is an attractive<br />

anti-tumour therapeutic setting.<br />

This work was supported by grants Nos. 301/07/1410, 301/10/2174<br />

and 301/09/1024, Grant Agency of the Czech Republic, and in<br />

part by the Clinigene Network of Excellence for the Advancement<br />

of Gene Transfer and Therapy, EU-FP6 Project No. 018933.


113 Kermer | Enhancing immunity & adjuvants<br />

Antibody fusion proteins for cancer immunotherapy mimicking<br />

IL-15 trans presentation at the tumor site<br />

Vanessa Kermer, Volker Baum, Roland E. Kontermann & Dafne Müller<br />

Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany<br />

Cytokines driving the immune response are po-<br />

werful tools for cancer immunotherapy, but their<br />

application is generally limited by severe systemic<br />

toxicity. Targeted approaches by means of antibody-cytokine<br />

fusion proteins might enable to focus<br />

the cytokine activity to the tumor site, thereby<br />

reducing unwanted side effects. Here we investigated<br />

the possibility to improve the efficiency of<br />

IL-15 presentation in a targeted approach by the<br />

incorporation of an IL-15R chain fragment, mimicking<br />

physiological trans presentation. Therefor,<br />

antibody cytokine fusion proteins were generated<br />

composed of an antibody moiety targeting the<br />

tumor stromal fibroblast activation protein (FAP),<br />

an extended IL-15Rαsushi domain (RD) and IL-15.<br />

These fusion proteins exhibited antibody-mediated<br />

specific binding and cytokine activity. Comparing<br />

the cytokine effect of antibody fusion proteins<br />

with and without the RD component, we observed<br />

differences for the soluble and membrane-bound<br />

form in stimulating proliferation of cells expressing<br />

the intermediate (IL-15Rβγ) and high affinity (IL-<br />

15Rαβγ) IL-15 receptor. For the fusion proteins in<br />

solution, the presence of the receptor domain (RD)<br />

reduced the stimulatory activity on CTLL-2 (IL-<br />

15Rαβγ) cells and increased the stimulatory effect<br />

on Mo7e (IL-15Rβγ) cells and PBMCs. In the targeted,<br />

membrane-bound form of the fusion protein,<br />

the presence of RD did not interfere with the proliferation<br />

of CTLL-2 (IL-15Rβγ) cells and clearly enhanced<br />

the proliferation of Mo7e (IL-15Rβγ) cells.<br />

Thus, targeted presentation of IL-15 in combination<br />

with the RD in form of an antibody fusion protein<br />

appears as a promising approach to further improve<br />

the antitumor efficiency of IL-15.<br />

163


114 Lehmann | Enhancing immunity & adjuvants<br />

Modified vaccinia virus Ankara (MVA) - a safe vector virus<br />

and an adjuvant system for immunotherapy<br />

Michael H. Lehmann 1 , Ulrich Kalinke 2 , Gerd Sutter 1<br />

1 Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-Universität München,<br />

80539 Munich, Germany<br />

2 TWINCORE, Centre of Experimental and Clinical Infection Research, 30625 Hannover, Germany<br />

164<br />

Modified vaccinia virus Ankara (MVA) is a highly<br />

attenuated strain of vaccinia virus (VACV) being<br />

generated through serial passage in chicken<br />

embryo fibroblast cells [1]. Until 1988, MVA was<br />

used for safer vaccination against smallpox and has<br />

been administered to more than 100 000 humans<br />

without documentation of the severe adverse reactions<br />

associated with the use of conventional VACV<br />

vaccines. Analysis of the MVA genome revealed that<br />

during the attenuation process the virus had lost a<br />

substantial amount of genetic information including<br />

many viral genes regulating virus-host interaction.<br />

In consequence, MVA is unable to propagate<br />

in human and many other mammalian cells but<br />

can efficiently express viral and recombinant genes<br />

[2]. This phenotype strongly supported its use as<br />

viral vector and, by today, a variety of recombinant<br />

MVA vaccines are undergoing clinical testing in<br />

immune prophylaxis and immune therapy against<br />

infectious diseases and cancer.<br />

Here, we investigated the immune stimulatory properties<br />

of MVA. Surprisingly, MVA but not other<br />

VACV strains induced expression of type I IFNs [3].<br />

We detected IFN-beta both in human monocytic<br />

cells and in the lungs of mice intranasally infected<br />

with MVA. Additionally, infection of human cells<br />

or mice with MVA but not with other VACV strains<br />

efficiently induced the expression of several chemokines<br />

(CCL2, CCL3, CCL4, CXCL1, CXCL10),<br />

which led to the fast attraction of leukocytes including<br />

monocytes, neutrophils, CD4+ and CD8+<br />

lymphocytes to the site of infection [4]. Since active<br />

immigration of immune cells to the site of immunization<br />

is a prerequisite for induction of an effective<br />

immune response, MVA vaccines naturally comprise<br />

this hallmark of a potent vaccine adjuvant.<br />

In summary, the properties making MVA an ideal<br />

vector for immunotherapeutic protocols are (i) its<br />

clinical safety as replication deficient virus, (ii) its<br />

ability to easily transfer and express foreign genetic<br />

material in target cells of choice and, (iii) its capacity<br />

to activate type I interferons, chemokines and<br />

thereby a rapid immigration of leukocytes.<br />

References<br />

[1] A. Mayr, E. Munz, Veränderungen von Vaccinevirus durch<br />

Dauerpassagen auf Hühnerembryofibroblasten-Kulturen,<br />

Zentralbl. Bakteriol. Orig. A 195 (1964) 24-35.<br />

[2] G. Sutter, B. Moss, Nonreplicating vaccinia vector efficient<br />

ly expresses recombinant genes, Proc. Natl. Acad. Sci. USA<br />

89 (1992) 10847-10851.<br />

[3] Z. Waibler, M. Anzaghe, H. Ludwig, S. Akira, S. Weiss, G.<br />

Sutter, U. Kalinke, Modified vaccinia virus Ankara induces<br />

Toll-like receptor-independent type I interferon responses, J.<br />

Virol. 81 (2007) 12102-12110.<br />

[4] M.H. Lehmann, W. Kastenmuller, J.D. Kandemir, F. Brandt,<br />

Y. Suezer, G. Sutter, Modified vaccinia virus ankara triggers<br />

chemotaxis of monocytes and early respiratory immigration<br />

of leukocytes by induction of CCL2 expression, J. Virol. 83<br />

2009) 2540-2552.


115 Lladser | Enhancing immunity & adjuvants<br />

DAI (ZBP1/DLM-1) as a novel genetic adjuvant for cancer DNA<br />

vaccines that promotes CTL responses, overcomes tolerance and<br />

confers long-term tumor protection.<br />

Alvaro Lladser 1 , Dimitrios Mougiakakos 1 , Helena Tufvesson 1 , Andrew F.G. Quest 2 , Karl<br />

Ljungberg 1 *, Rolf Kiessling 1 *<br />

1 Immune and Gene Therapy Laboratory, Cancer Center Karolinska, Department of Oncology and Pathology,<br />

Karolinska Institutet. Karolinska Hospital R8:01, SE-17176 Stockholm, Sweden<br />

2 Laboratory of Cellular Communication, FONDAP Center for Molecular Studies of the Cell, Program of Cellular<br />

and Molecular Biology, Facultad de Medicina, Universidad de Chile, Santiago, Chile<br />

* KL and RK contributed equally to this work<br />

DNA vaccination is an attractive approach to induce<br />

antigen-specific cytotoxic T cell (CTL) responses<br />

capable of providing long-lasting protective immunity<br />

against cancer. Although effective in animal<br />

models, DNA vaccines against cancer have shown<br />

limited efficacy in clinical trials. Therefore, efficient<br />

delivery systems and powerful adjuvants are<br />

needed for DNA vaccines to overcome tumor-associated<br />

T cell tolerance. Indeed, the early activation<br />

of innate immune receptors and downstream signaling<br />

pathways are essential to promote effective<br />

adaptive immunity. Consequently, DNA vaccines<br />

benefit from adjuvants that boost innate immunity<br />

signaling. We have used the recently described cytosolic<br />

DNA sensor Z-DNA binding protein 1 (ZBP1<br />

also known as DLM-1), termed DNA-dependent activator<br />

of interferon regulatory factors (DAI) as a<br />

genetic adjuvant for DNA vaccines. In vivo electroporation<br />

(EP) of mice with a DAI-encoding plasmid<br />

(pDAI) promoted transcription of genes encoding<br />

type I interferons (IFNs), proinflammatory cytokines,<br />

chemokines and co-stimulatory molecules.<br />

Co-immunization of pDAI and antigen-encoding<br />

plasmids enhanced both in vivo antigen-specific<br />

proliferation and induction of CTLs. Moreover, codelivered<br />

pDAI overcame CTL-tolerance to a tumorassociated<br />

antigen (TAA) and conferred long-term<br />

anti-tumor protection. The DAI-adjuvanted CTL induction<br />

required NF-κB activation and intact type I<br />

IFN signaling, but not interferon regulatory factor<br />

(IRF) 3. This study demonstrates the potential of<br />

using intracellular innate sensors as genetic adju-<br />

vants to improve DNA vaccine potency.<br />

Acknowledgements: Research described here has<br />

been supported by grants to RK from the Swedish<br />

Cancer Society, the Swedish Medical Research<br />

Council, the Cancer Society of Stockholm, the European<br />

Union (Grant “EUCAAD” and “DC-THERA”),<br />

the Karolinska Institutet, “ALFProject” grants<br />

from the Stockholm City Council. AFGQ has received<br />

support from ICGEB (International Center of<br />

Genetic Engineering and Biotechnology, Trieste,<br />

Italy) grant CRP/CH102-01, Wellcome Trust award<br />

WT06491I/Z/01/Z and FONDAP grant 15010006.<br />

AL has been supported by a Fellowship for Postgraduate<br />

Studies “Presidente de la República” from<br />

CONICYT, Chile. DM was supported by a grant of<br />

the German Research Association (DFG). KL has<br />

been supported by a postdoctoral fellowship from<br />

the Swedish Society for Medical Research.<br />

165


116 Hotz | Enhancing immunity & adjuvants<br />

A rational protocol of repeated TLR7 stimulation<br />

circumvents induction of TLR tolerance and translates<br />

into efficient anti-tumor therapy<br />

Christian Hotz 1 , Andreas Völkl 1 , Daniel Noerenberg 1 , Nadja Sandholzer 1 , Cornelia Wurzenberger<br />

1 , David Anz 1 , Stefan Endres 1 and Carole Bourquin 1<br />

1 Center for Integrated Protein Science Munich, Division of Clinical Pharmacology,<br />

Department of Internal Medicine, Ludwig-Maximilian University of Munich, Munich, Germany.<br />

166<br />

Topical application of small molecule toll-like re-<br />

ceptor 7 (TLR7) agonists is often highly effective<br />

for the treatment of skin tumors, whereas their systemic<br />

application has been largely unsuccessful<br />

for the immunotherapy of cancer. One reason may<br />

be that repeated application of certain TLR ligands<br />

can induce a state of immune unresponsiveness,<br />

termed TLR tolerance. We show here that a single<br />

injection of the TLR7 agonist R848 in mice induces<br />

a short period of increased responsiveness followed<br />

by a state of hyporesponsiveness lasting several<br />

days. We demonstrate for the first time that TLR7<br />

tolerance occurs in plasmacytoid and myeloid dendritic<br />

cells, that play a critical role in the initiation<br />

and amplification of antitumor immune responses,<br />

and results in inhibited secretion of the key cytokines<br />

IFN-κ, IL-12p70 and IL-6 both in vitro and in<br />

vivo. We further show that TLR7 tolerance in plasmacytoid<br />

dendritic cells is accompanied by downregulation<br />

of the adaptor protein interleukin-1 receptor-associated<br />

kinase (IRAK-1). Based on these<br />

findings, we have designed a novel strategy for the<br />

treatment of murine CT26 tumors using cycles of<br />

repeated R848 injections separated by 5-day treatment-free<br />

intervals. We show that this protocol<br />

circumvents TLR7 tolerance and translates into<br />

efficient cancer immunotherapy whereas single injections<br />

given within shorter intervals do not.


117 Gonzalez-Carmona | Enhancing immunity & adjuvants<br />

Combining subcutaneous inoculation of AFP-expressing DC<br />

with intraperitoneal injection of IL-12-expressing DC increases<br />

survival in an established orthotopic HCC model<br />

Maria A. González-Carmona 1 , Annabelle Vogt 1 , Georges Decker 1 , Isabelle Bahadori 1 , Esther<br />

Raskopf 1 , Volker Schmitz 1 , Tilman Sauerbruch 1 and Wolfgang H. Caselmann 2<br />

1 Department of Internal Medicine I, University of Bonn, Bonn, Germany<br />

2 Bavarian State Ministry of the Environment and Public Health, Munich, Germany<br />

Background: Dendritic cells (DC) are a heterogene-<br />

ous population of professional antigen-presenting<br />

cells capable of priming CD4 and CD8 T-cell responses<br />

against tumor-associated antigens (TAA)<br />

such as alpha-fetoprotein (AFP). The presence of<br />

an immunosuppressive tumor-environment going<br />

along with the suppression of DC is one of the main<br />

reasons for a lack of immune responses after immunotherapy.<br />

IL-12 as a major Th1 cytokine is essential<br />

for the induction of cellular immune responses. In<br />

this study, the antitumoral effect of a vaccination<br />

with s.c. inoculated AFP-expressing DC combined<br />

with the intraperitoneal injection of IL-12-expressing<br />

DC in order to achieve an inflammatory tumor<br />

environment was analyzed in an orthotopic HCC<br />

model.<br />

Methods: Three adenoviral vectors were used: AdmIL12,<br />

Ad-mAFP and Ad-LacZ as a control. DC<br />

were obtained from the bone marrow of C3H-mice,<br />

cultured with GM-CSF and IL-4 and adenoviral<br />

transduced on day 6. 1x105 AFP-positive Hepa129cells<br />

were injected into the liver after laparotomy<br />

to induce orthotopic tumors. In a first experiment,<br />

tumor-bearing mice were treated twice with 1x106<br />

mIL-12-expressing or LacZ-expressing DC intraperitoneally.<br />

In a second experiment, mice were<br />

treated with the combination of 1x106 s.c. inoculated<br />

AFP-expressing DC followed by i.p. injection<br />

of 1x106 mIL-12-expressing DC. Survival time and<br />

occurrence of malignant ascites were closely monitored<br />

and documented.<br />

Results: In the first experiment 100% of mice in the<br />

control group (LacZ-expressing DC) developed mali-<br />

gnant ascites as a sign of tumor progression whereas<br />

only 30% of the mice treated with IL-12-expressing<br />

DC treated mice had developed. Furthermore, the<br />

mean survival after tumor induction was significantly<br />

prolonged in the mice group treated with<br />

Ad-mIL12-expressing DC (p


118 Trojandt | Enhancing immunity & adjuvants<br />

Effects of the anti-cancer agent topotecan on human<br />

monocyte-derived dendritic cells<br />

Stefanie Trojandt, Stephan Grabbe, Angelika B. Reske-Kunz and Matthias Bros<br />

Clinical Research Unit Allergology, Department of Dermatology, Medical Center of the Johannes Gutenberg-University,<br />

55101 Mainz, Germany<br />

168<br />

The camptothecin analogue topotecan has shown<br />

a remarkable activity against a range of cancers<br />

and tumors. Topotecan interacts with the enzyme<br />

topoisomerase I which is involved in DNA replication<br />

and repair. It forms a stable complex with this<br />

enzyme, thereby provoking the selective inhibition<br />

of topoisomerase I. This results in irreversible DNA<br />

double-strands breaks, giving rise to apoptosis and<br />

cell death.<br />

Due to the fact that dendritic cells (DCs) play an<br />

important role in anti-tumor immune responses,<br />

we raised the issue of whether topotecan may have<br />

an impact on DC activation.<br />

In this study we investigated the effect of topotecan<br />

on the molecular characteristics of DCs regarding<br />

the expression of DC-relevant molecules and their<br />

T cell stimulatory capacity. Human monocytes<br />

were differentiated to immature DCs and were<br />

stimulated with a maturation cocktail (TNFalpha,<br />

IL-1beta, Prostaglandin2). During stimulation with<br />

the cocktail the cells were treated with different<br />

concentrations of topotecan. At the concentrations<br />

used topotecan had no effect on the viability of the<br />

DCs. Contact of the DCs with this agent resulted in<br />

an impaired stimulatory capacity for allogenic T<br />

cells. Nevertheless the treated DC populations expressed<br />

high levels of HLA-DR and CD86. However,<br />

the expression of the costimulatory molecule CD80<br />

and the maturation marker CD83 was slightly impaired.<br />

In summary, we showed a modification in the func-<br />

tional activity of DCs after their treatment with the<br />

anti-tumor agent topotecan.


L126 Klier | Enhancing immunity & adjuvants<br />

Combined bacterial antibody therapy for colorectal carcinoma<br />

U. Klier 1 , C. Maletzki 1,2 , E. Klar 1 , Bernd Kreikemeyer 3 , M. Linnebacher 1<br />

1 Section of Molecular Oncology and Immunotherapy, Department of General Surgery,<br />

University of Rostock, Germany<br />

2 Division of Gastroenterology, Department of Internal Medicine,<br />

University of Rostock, Germany<br />

3 Department of Medical Micobiology and Hospital Hygiene, Institute of Medical Microbiology,<br />

Virology and Hygiene, University of Rostock, Germany<br />

Background & Aims: Here, we analyzed the po-<br />

tential of inactivated Staphylococcus aureus loaded<br />

with therapeutic monoclonal antibodies (mAb), for<br />

treatment of colorectal carcinomas in vitro and in<br />

vivo. The aim was to induce an inflammatory reaction<br />

first against bacteria followed by the activation<br />

of tumor specific immune responses.<br />

Material & Methods: Colorectal carcinoma cell<br />

lines, established in our laboratory, were treated<br />

with S. aureus, antibodies (Panitumumab (anti-<br />

EGFR), or Trastuzumab (anti- Her2neu)) or a combination<br />

of both for 24 and 48 hours respectively.<br />

Cell proliferation and viability were assessed by<br />

BrdU-incorporation assay and Calcein AM staining.<br />

Then, we tested the ability of these experimental<br />

agents to stimulate T cells and determined<br />

the changing in the pattern of Toll like receptors.<br />

Additionally, antitumoral effects were examined in<br />

co-culture experiments using peripheral blood mononuclear<br />

cells (PBMCs) from healthy donors. As<br />

in vivo model, Balb/c mice with established CT26<br />

tumors (stably expressing Her2/neu) received local<br />

injections of the therapeutics or saline as a control<br />

(total of 10 injections given twice a week, n=6).<br />

Tumor-infiltrating leukocytes were analyzed by<br />

immunohistochemistry.<br />

Results: Proliferation and survival of tumor cells<br />

was slightly affected by the therapeutical agents.<br />

However, antitumoral effects were boosted after<br />

co-culture with PBMCs in the presence of bacteria<br />

and mAbs. Flow cytometric analysis of PBMCs re-<br />

vealed an increase of CD4+/25+ and CD8+/25+<br />

cells (24 h), indicative for activation of T cells by<br />

S. aureus. In vivo, S. aureus treatment induced a<br />

delay of tumor growth. These effects were accompanied<br />

by increased numbers of tumor-infiltrating<br />

CD11b+ granulocytes/macrophages, CD4+ as well<br />

as CD8+ T cells. Similar results were obtained after<br />

combination therapy.<br />

Conclusion: Our results demonstrate that S. aureus<br />

is capable to elicit direct antitumoral and immunostimulatory<br />

effects without the need of a targeted<br />

therapy with mAbs. Current investigations focus<br />

on testing specific antitumoral T cell responses<br />

subsequent to stimulation with S. aureus.<br />

169


Imprint<br />

Postal Address<br />

<strong>CIMT</strong> - Association for Cancer Immunotherapy<br />

Center for Translational Oncology and Immunology<br />

Gutenberg University<br />

Mainz Langenbeckstr. 1, Building 708<br />

55131 Mainz Germany<br />

Telephone: +49-06131-17-5905<br />

Telefax: +49-06131-17-3490<br />

Website: www.cimt.eu<br />

E-Mail: office@cimt.eu<br />

Concept and Design<br />

Das LABOR - Büro für Gestaltung<br />

Rheinallee 88 / Geb. 25<br />

55120 Mainz, Germany<br />

Telephone: 06131 - 30 46 76 2<br />

Website: www.das-labor.net<br />

© Immunologische Krebs-Therapie e.V. (Association for Cancer Immunotherapy). All rights reserved.<br />

170<br />

Register of associations<br />

Amtsgericht Mainz: VR 3783<br />

Print<br />

Buchbinderei Schulz<br />

Am Heckerpfad 12<br />

55128 Mainz<br />

Telephone: 06131 - 3 41 47<br />

Website: www.buchbinderei-schulz.de


171

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!