Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Co Ne n t e n t s x t Annual Rep ort 2006 Department of Medical Oncology VU University Medical Center Amsterdam, The Netherlands V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Ne x t Annual Rep o rt 2006 Department of Medical Oncology VU University Medical Center Amsterdam, The Netherlands Editor: Dr. H.J. Broxterman Department of Medical Oncology VU University Medical Center De Boelelaan 1117 1081 HV Amsterdam The Netherlands www.vu-medicaloncology.org V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr e v i ou s Contents C ONTENT S Ne x t DEPARTMENT STAFF 4 INTRODUCTION 8 SYMPOSIA, LECTURES AND COURSES 9 PHASE I/II CLINICAL RESEARCH 12 PSYCHO-ONCOLOGY AND PALLIATIVE CARE SECTION 14 ONCOLOGY WARD 17 OUTPATIENT CLINIC AND DAY CARE UNIT 19 ACTIVITIES OF THE RESEARCH DIVISIONS Division of Angiogenesis Division of Gene Therapy Division of Immunotherapy Division of Pharmacology Oncoproteomics Laboratory 20 20 24 28 34 39 PhD THESES 43 SCIENTIFIC PUBLICATIONS 44 C OLO F ON Copyright © 2007, Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands. All rights reserved. No part of this publication may be reproduced or transmitted in any form or by means, electronic or mechanical, including photocopying, recording, or any information storage and retrieval system, without the written permission of the Department of Medical Oncology, VU University Medical Center. Production: Lobbezoo Medical Communications, Hilversum, the Netherlands. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Ne n t e n t s Department staff D C l i n i ca l S t aff Prof. G. Giaccone, MD, PhD, head of department Prof. E. Boven, MD, PhD Dr. J. Buter, MD, PhD Dr. A.J.M. van den Eertwegh, MD, PhD Dr. W.R. Gerritsen, MD, PhD Dr. C.J. van Groeningen, MD, PhD Dr. K. Hoekman, MD, PhD Dr. B. Kuenen, MD, PhD Prof. H.M. Pinedo, MD, PhD e p ar t m e n t C l i n i ca l S t aff T ra i n i n g s t aff in Drs. A. Beeker, MD (until May 1) Drs. H.P. van den Berg, MD Drs. A. Goosens, MD (until March 1) Drs. S. van den Heiligenberg, MD Drs. A. Keijzer, MD Dr. J.J. van der Vliet, MD, PhD Psycho-Oncology and P a l l i at i v e C ar e S t aff M.S.A. Boddaert, MD, palliative care physician Dr. M.H.M. van der Linden, PhD, clinical psychologist S u p p o r t i v e S t aff Members of clinical, laboratory and nursing staff during a brain-storm session. Standing from left to right: dr. C. Jiménez, dr. A.J.M. van den Eertwegh, dr. F.A.E. Kruyt, dr.B. Kuenen, dr. H.J. Broxterman, prof. J. Lankelma, dr.V.W. van Beusechem, dr. K. Hoekman, dr. J. Buter, prof. G.J. Peters, prof. V.W.M. van Hinsbergh; Sitting from left to right: H. Peltenbrug, dr. T.D. de Gruijl; In front from left to right: M.S.A. Boddaert, prof. E. Boven, dr. M.H.M. van der Linden, dr. W.R.Gerritsen.. x t L. Groenewegen, IT consultant M.R. Regtuit, financial manager Drs. A. Carper, MD, personal assistant to prof. H.M. Pinedo O n c o l o g y R e s e arc h L a b o rat o r i e s Prof. G.J. Peters, PhD, head of laboratories Dr. F.A.E. Kruyt, PhD, molecular biologist, deputy head of laboratory Division of Angiogenesis Prof. V.W.M. van Hinsbergh, head of division Prof. E. Boven, MD, PhD, laboratory head Dr. K. Hoekman, MD, PhD, project leader Prof. J. Lankelma, PhD, chemist, project leader Dr. H.J. Broxterman, PhD, biochemist, project leader M.N.A. Bijman, MSc, medical biologist (until September 1) Dr. W.P.H. de Boer, PhD, theoretical physicist (guest) Dr. R. Fernández Luque, PhD, physicist (guest) Dr. R. Fijneman, PhD, molecular biologist Dr. A. Greijer, PhD, biologist (until February 1) Dr. M.L. Janmaat, PhD, biologist (until November 1) H. van Cruijsen, MD A.A.M. van der Veldt, MD Dr. B.C. Kuenen, MD, PhD Dr. R.R. de Haas, PhD, senior research technician Ing. M.P.A. van Berkel, technician Ing. H. Dekker, technician Ing. P.M. Delis-van Diemen, technician Ing. Y. Yuana, Msc, technician (until August 1) L. Vroling, MSc, medical biologist V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Division of Gene Therapy Dr. W.R. Gerritsen, MD, PhD, head of division Dr. V.W. van Beusechem, PhD, medical biologist, laboratory head Prof. C. Dirven, MD, neurosurgeon Ing. A. Huizenga, technician Dr. F.H.E. Schagen, PhD, biochemist (until August 1) Dr. M.L.M. Lamfers, PhD, medical biologist (until August 1) S. Idema, MSc, MD Ing. I.H. van der Meulen-Muileman, technician Ing. S. Moeniralm, technician Division Immunotherapy (in c o o p e r at i o n w i t h t h e D e pa r t m e n t o f P at h o l o g y ) Dr. A.J.M. van den Eertwegh, MD, PhD, oncologist/immunologist, head of division Prof. R.J. Scheper, PhD, tumor immunologist, laboratory head Dr. T.D. de Gruijl, PhD, tumor immunologist, group leader Dr. H.J. Bontkes, PhD, tumor immmunologist Dr. B. Hangalapura, PhD biologist (as of May 1) Dr. D. Oosterhoff, PhD medical biologist S.J.A.M. Santegoets, MSc, medical biologist J. Lindenberg, Msc, medical biologist (as of 10 August 1) J. Molling, MSc, medical biologist M. Moreno, Msc, medical biologist of Ne n t e n t s R. van de Ven, MSc, medical biologist H. van Cruijsen, MD B.J.R. Sluijter, MD S.M. Lougheed, MSc, medical biologist, technician Ing. N. Ravenhorst, technician Ing. A.W. Reurs, technician Ing. A. Stam, technician P.G.J.T.B. Wijnands, MSc, medical biologist, technician D i v i s i o n o f P h a r m ac o l o g y Prof. G. Giaccone, MD, PhD, head of division Prof. G.J. Peters, PhD, biochemist, head of laboratory Dr. F.A.E. Kruyt, PhD, molecular biologist, deputy head of laboratory Dr. J.A. Rodriguez, PhD, molecular biologist, project leader I.V. Bijnsdorp, MSc, med. biologist A. Checinszka, MSc, biochemist (until February 1) C. Lemos, MSc, biochemist J. Sigmond, MSc, medical biologist (until April 1) J. Voortman, MSc, MD A. Watts, Msc, biologist (as of June 1) Dr. R.J. Honeywell, PhD, senior research technician Ing. K. Smid, senior research technician Ing. S. Span, senior research technician A. Adema, MSc, technician Ing. K. Floor, technician x t Ing. M. Gallegos-Ruiz, Msc Ing. E. Hoebe, technician Ing. G.A.M. Kathmann, technician Ing. A.C. Laan, technician Ing. P. Noordhuis, MSc (as of October 1) Ing. E. Torun, technician (until July 1) O n c o p r o t e o m i c s L a b o r at o r y Dr. C. Jiménez, PhD, chemist, laboratory head Dr. J.C. Knol, PhD, chemist (as of April 1) Dr. S. Piersma, PhD, chemist (as of May 1) Z. El Filali, MSc, technician (as of May 1) Dr. T. Pham, PhD, bioinformatics (as of October 1) P s y c h o - o n c o l o g y a n d P a l l i at i v e Care Section Dr. W.R. Gerritsen, MD, PhD, chairman M.S.A. Boddaert, MD E.J. Douwes Jr., lay expert F. Eskens, medical social worker, Department of Medical Social Work Dr. K. Hoekman, MD, PhD Dr. M.H.M. van der Linden, PhD, clinical psychologist J. van Ooijen, nurse oncology day care unit V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr e v i ou s Co O n c o l o g y W ar d N u rs i n g S t aff R e s e arc h N u rs e s H. Peltenburg, head oncology ward & outpatient clinic C. Stouthart, deputy head nurse T. Blankwater A. Bos H. Bos J. Brandenburgh A. Dekkers M. Douwes A. Frantzen A. Hellebrekers-Bos Eijssen L. Jansma B. Jorritsma S. Kooiman J. van de Lans J. Oosterling E. Pauw H. Refat T. Rensink R. Schopenhauer J. Schouten J. van Staveren A. Stapert L. Tuinman J. ter Veen B. Verdegaal M. IJmker-Westeneng H.E. Gall E. Doeleman I. van der Horst R. Ruijter C. Tillier W ar d A ss i s t a n t s D. van Giffen C. Hogers C. de Reus M. Verhoef T. van Os A d m i n i s t rat i v e S t aff O n c o l o g y W ar d I. Charan B. Guman M. van den Haak J. Commandeur M.L.C. Latupeirissa M. Secici D at a M a n a g e rs K. Groot V. Hartog F. Terpoorten Ne n t e n t s x t N u rs i n g S t aff D ay C ar e U n i t N. Boere K. van der Jagt T. Graas I. Kamping-Steenkist B. Krolis M. Koeree L. Lenten-Cetinel J. van Loo J. van Ooijen A. Paulus M. Piet-Krijnen C. Vree-Bergh M. Nunez Sandez, departmental assistant A d m i n i s t rat i v e S t aff O u t pat i e n t C l i n i c J. Stolker, head secretary S. van der Feen J. van der Heide A. Kuipers I. de Ruyter L. van Straten J. Versteeg V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s n t e n t s Ne x t V o l u n t e e rs A. Bisterbosch J. Lantink C. Manshanden H. de Winter S e cr e t ar i a l S t aff M.H. Plieger, head secretary C. Clarinda-Overmeer S. van Geloven M. Klaassen P. Voogd Secretarial and support staff. From left to right: S. van Geloven, P. Voogd, M. Regtuit, M. Klaassen, C. Clarinda-Overmeer. Front: M.H. Plieger. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr e v i ou s Co In t r o d u c t i o n I The year 2006 has seen an increase in the number of clinical studies started by the department as well as in the number of patients included in clinical studies. A strong program focussing on phase I-II studies and on translational studies has been firmly established. In general, a shift towards targeted therapies and molecular research related to the mechanisms of action of those therapies has been seen. In fact, within the phase I unit emphasis has been on the development of agents targeting the Epidermal Growth Factor (EGF) receptor, angiogenesis inhibitors, as well as novel immunotherapies. A major development in 2006 was moving the laboratory facilities of the department into the new building of the Cancer Center Amsterdam (CCA), located adjacent to the outpatient clinic of the hospital. CCA opened its doors officially on June 19, 2006. Also, the new core facility for proteomics , established in collaboration with other departments interested in using this novel technology platform, got its place in the new CCA building. The new CCA laboratory facility aims at bringing together under one roof all research laboratories of the major departments of the VU University Medical Center conducting cancer research (i.e., medical oncology, pathology, n t e n t s Ne x t n t r o d u c t i o n haematology, head and neck surgery, dermatology and pediatric oncology). New cancer research laboratory of the Cancer Center Amsterdam up a student exchange program with both the University of Bologna, Italy, and the University of Turin, Italy, with students from both universities spending a number of months in our laboratory to gain research experience. A selection from the diversity of activities of the Department of Medical Oncology has been highlighted here. These and other activities are described in more detail in this Annual Report. We are confident that ongoing activities, new developments and initiatives in our research, organization (phase I/II unit) and education will lead to further improvements in cancer patient care in the years to come. In the year 2006, our international collaboration with foreign institutions has continued, in particular with Johns Hopkins Cancer Center in Baltimore (HIF-1 research), Institut GustaveRoussy in Paris (gene therapy of malignant gliomas), University of Alabama in Birmingham, Alabama (novel viral vectors for gene therapy), and Harvard University (angiogenesis). Our collaboration with Johns Hopkins is fostered by an international grant from Aegon. Prof. Van Hinsbergh coordinates this line of research for the department. The department has also set V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Ne n t e n t s x t Symposia, lectures and courses S y m p o s i a TAT 2 0 0 6, M arc h 1 6 - 1 8 , Amste rdam , The Netherlands In March 2006, the 4th International Symposium on Targeted Anticancer Therapies (TAT 2006) was held at the VU University in Amsterdam. This meeting was organised by the NDDO Research Foundation (congress director: Dr. M.W. Lobbezoo; www.nddo.org ) together with the European Society for Medical Oncology (www. esmo.org) and the Department of Medical Oncology. The department head, prof. G. Giaccone, served as Symposium President. The TAT series builds on a tradition of symposia organised at the VU on new anticancer drug development. TAT 2006 demonstrated that this annual meeting series has come of age. It provides an excellent forum for the presentation and discussion of virtually all aspects of targeted anticancer therapies in (pre)clinical development. Compared to the meeting in 2005, some important adjustments in the focus and the format of the meeting have been implemented successfully in 2006. These included the focussing on early-phase clinical development, , l e c t u r e s a n d resulting in more original presentations on phase I and II studies, and stricter abstract review, resulting in a higher rejection rate and better overall quality of the scientific presentations. Overall, well over 400 people representing 42 different countries attended TAT 2006, a clear increase in attendance over TAT 2005. About 70% of delegates were from European countries, 20% from the USA and Canada, and 10% from the rest of the world. Compared to TAT 2005, the relative size of the delegations from Asia and the rest of the world has increased at the expense of European delegations, illustrating the further globalization of the TAT series. Delegates’ evaluations of various aspects of TAT 2006 ranged between 3.2 and 4.2 on a scale from 1 to 5. TAT 2006 witnessed the start of a special task force on ‘Methodology for the Development of Innovative Cancer Therapies’ (MDICT). This forum of experts from leading academic cancer research institutes, supplement with observers from, industry and regulatory agencies, attempts to develop practical guidance on the optimal development of innovative anticancer agents. Dr. Elizabeth Eisenhauer presented an extensive summary of MDICT’s first session on important issues in the design of phase I studies of targeted c o u rs e s agents in the main symposium. A separate report on the first MDICT meeting has been offered for publication in a scientific journal. One of the highlights was the NDDO Award Lecture on ‘Adventures in targeted cancer therapy: From tumour hypoxia to the cancer genome’, presented by professor Paul Workman from the Institute of Cancer Research in Sutton, UK. Professor Workman and his co-workers have an impressive track record in drug design and discovery as well as in early-phase clinical and translational research. Professor Paul Workman (left) receives the NDDO Award 2006 from Dr. Coenraad van Kalken, Director NDDO Research Foundation (right) during the TAT 2006 Opening Ceremony. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Much attention has recently been drawn by drugs showing promising clinical activity in advanced renal cell cancer. In his Keynote Lecture, professor Martin Gore reviewed the most advanced drugs in clinical development, sorafenib (Nexavar®) and sunitinib (Sutent®), together with several promising drug candidates with less mature data. Sorafenib and sunitinib have recently obtained regulatory approval for this indication in the USA, the European Union, and several other countries. Among the many agents in active preclinical and clinical development and discussed during TAT 2006, AMN107 stood out by its remarkable clinical activity in a phase I study in imatinibresistant CML. AMN107 is a novel inhibitor of Bcr-Abl, the target of imatinib (Glivec®) in chronic myeloid leukaemia. x t O p e n d ay f o r pat i e n t s : “ K a n k e r a n d e rs b e l i c h t ” M ay 3 , 2 0 0 6 On May 3, 2006, we celebrated the fifth anniversary of the VU University Medical Center, which is a combination of the former VU Hospital and the VU School of Medicine. For our patients we organized a memorable program “Kanker anders belicht”, (“a different light on cancer”), loaded with live music and interesting information for cancer patients. Mary-Lou van Steenis elegantly presented the program. Among others, she introduced the choir “Zingen voor je leven” (“Singing for life”) directed by Harry de Beer . The choir makes use of different songs to express their emotions related to (previous) illness, such as “This little light of mine”, “What a wonderful world” and “With a little help from my friends”. The next meetings, TAT 2007 and TAT 2008, have been scheduled for March 8-10, 2007 and March 20-22, 2008, respectively. The choir “Zingen voor je leven” directed by Harry de Beer 10 Ne n t e n t s Harry de Beer Jacobine Donker being interviewed by Mary-Lou van Steenis Mr. Frits J. Duparc, MSc, director of the Mauritshuis Museum, The Hague, showed old paintings expressing loyalty and care. True friendship, fidelity in marriage, and care for the sick have often been subjects of imaginations on canvas. Jacobine Donker directed a short movie in which she followed two patients in their daily life. Both have to cope with limitations caused by their treatment, but can live a happy life. A short piano intermezzo was given by Mark van der Feen. Prof. Paul van der Valk demonstrated the possibilities the modern pathologist has to discover tumor characteristics at the molecular level. Laboratory research aimed at improving cancer treatment was illustrated by a short movie directed by Andrew Fallon. He followed a patient who participated in a clinical immunotherapy trial and showed the laboratory activities required in preparation of his treatment. Prof. Giuseppe Giaccone demonstrated the possibilities we V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s develop in the clinic to further improve therapies directed against specific targets in tumors. Participants were very enthusiastic about the market place in the Amstelzaal Foyer, which could be visited at lunch time. Many organizations were present, either with focus on a particular tumor type, on handling symptoms of disease or on special clothing. Attentive public in the Amstelzaal A few written reactions from patients, apart from the many words of thanks: - ‘we had a wonderful day and learned a lot’ -‘a very impressive and emotional program, which enabled us to manage disease in a better way’ -‘fantastic atmosphere and an excellent day; why is the next one planned only in 5 years?’ 11 n t e n t s Ne x t T e ac h i n g The department contributed to teaching, both to the new curriculum for medical students and to the Master in Oncology program. The Master in Oncology program was established in 2002 and was recognized by the Minister of Education as one of two top-masters of the VU University. In 2006, the master program performed a self evaluation in order to renew its recognition. Prof. Peters is chair of the examination committee and one of the organizers of the course on Innovative Therapies, in which students are taught on both standard and novel therapies. This also includes an intensive study of the literature to formulate, present and defend a research project. In 2006, twenty-five students passed their master. Some of the new masters in oncology continued their career by doing a Ph.D. thesis in the Department of Medical Oncology. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Ne n t e n t s x t Phase 1/11 clinical research P O r g a n i z at i o n h as e and fac i l i t i e s As in 2004 and 2005, the phase I/II research unit has been used very intensively in 2006. The unit consists of a three-bed patient room on the oncology ward with beds equipped with 2 monitoring devices, offering the possibility to monitor heart rate, blood pressure, oxygen saturation and body temperature of patients participating in trials. With the devices, also EKGs and analyses of heart rhythm (72 hour full disclosure) can be performed. In 2005, the laboratory for processing and storage of blood samples moved to a larger room near the phase I/II unit. It is equipped with centrifuges, flow chamber, and -20°C and -80°C freezers with a 24 hour temperature alarm system. The aim of the research unit is to improve care for patients participating in trials and professional competence in adhering to study protocols. Due to the increasing complexity of drug development in recent years, a dedicated team is required. The research unit is currently managed by five research nurses: Rita Ruijter, Helen Gall, Inge van der Horst, Corinne Tillier and Ellen Doeleman. The unit is supervised by Dr. B. Kuenen, medical oncologist, who also chairs all the phase I/II 12 1 / 1 1 c l i n i ca l r e s e arc h meetings at the department. The phase I/II team covers the full period from protocol development to completion of the trials. Their tasks consist of starting up studies, co-ordination of all trial related procedures, extra checks of in- and exclusion criteria, final enrolment of patients, pharmacokinetic sampling and work-up, planning visits and assessments according to the protocol, follow-up of patients, including scoring of toxicity, all logistics, sample shipment, etc. Staff of the phase I/II research unit (from left to right): Ellen Doeleman, Inge van der Horst, Rita Ruijter, Corinne Tillier, Bart Kuenen, Helen Gall C l i n i ca l studies Pharmacokinetic/pharmacodynamic evaluations were performed in all phase I studies and some of the phase II/III studies. A major focus of our department is, besides participating in phase II and III trials, performing phase I trials. The various phase I, II and III studies can be subdivided into three main categories: those investigating classical chemotherapy compounds, those investigating targeted compounds and those investigating combinations of targeted compounds or combinations of a targeted compound and chemotherapy. The targeted compounds under study in our department are mainly directed at two targets: the epidermal growth factor receptor (EGFR) pathway, which is involved in tumor growth in several tumor types, and the vascular endothelial growth factor (VEGF) pathway, which is the main player in the formation of new blood vessels (angiogenesis). Examples of trials with EGFR inhibitors are • a phase I dose escalation study of the fully humanised monoclonal antibody IMC-11F8; • a phase I/II study of erlotinib in combination with radiotherapy in rectal cancer; • a feasibility phase I/II study of erlotinib added to radiotherapy plus gemcitabine in locally advanced irresectable pancreatic cancer; • a metabolism study of erlotinib in smokers versus non-smokers; • a nationwide Dutch trial randomising patients with colorectal cancer to capecitabine, oxaliplatin and bevacizumab with or without cetuximab (CAIRO2) V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s n t e n t s Ne x t Angiogenesis inhibitors under study in our department include sunitinib, SU014813, AZD2171, BMS582664, VEGF-trap, and bevacizumab. Other targets investigated in the clinic are the apoptotic machinery (anti-survivin and AMG951), which is also extensively studied in our preclinical department, and m-TOR (RAD001). Another main focus of our clinical research program is immunotherapy with a special interest in vaccinations combined with an anti-CTLA4 monoclonal antibody (MDX010) or chemotherapy. A number of trials with these combinations are open for accrual in prostate cancer and melanoma. The department aims at performing translational research and therefore many side-studies related to the clinical trial protocols are being performed. Examples are an extensive analysis of the EGFR expression patterns, mutational status and downstream effects in tumors of patients treated with EGFR-inhibitors, analysis of maturation of dendritic cells during treatment with angiogenesis inhibitors, the mechanism of hypertension in patients treated with angiogenesis inhibitors, detection of circulating endothelial cells and precursors in patients treated with angiogenesis inhibitors, extensive immunological monitoring of patients in the vaccination/anti-CTLA4 trials and performing proteomics of blood samples. 13 V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr P Co e v i ou s x t Psycho-oncology and palliative care section - s y c h o O r g a n i z at i o n o n c o l o g y The section of Psycho-oncology and Palliative Care continued its efforts to improve the quality of care within the department*. In 2005, the section started a scientific research network in collaboration with the Department of Medical Psychology, the Department of Otolaryngology, Head and Neck Surgery, the Department of Social Medicine and the Ingeborg Douwes Center for Psycho-Oncology to investigate quality of life and palliative care. The focus in 2006 was on developing new scientific projects. This led to grant applications for several projects concerning an individualised quality of life intervention for hospitalised patients with advanced cancer, the selection and validation of instruments for measuring outcome of care in oncology patients without curative treatment options and screening of ‘high risk’ patients with the Emotional Distress Thermometer (J. Holland, 2005). We plan starting these projects in 2007. In 2006, psychological and palliative care has been extended to patients of several departments in the VU University Medical Center. Besides providing patient care, both dr.M.H.M. van der Linden and drs. M.S.A Boddaert, continued educating health practitioners in psychosocial 14 Ne n t e n t s a n d pa l l i a t i v e and palliative care to enhance professional expertise in the field of care for cancer patients. In addition, both the medical faculty and the hospital education centre extended psychooncology and palliative care education in their curriculum for medical students and oncology nurses. In collaboration with the regional Comprehensive Cancer Center Amsterdam (IKA) and Leiden University Medical Center, master classes on psycho-oncology for medical students and psychology students were organized. P a l l i at i v e car e s e c t i o n double rooms on the ward were transformed to single rooms and fitted with rooming-in capacity**. Official opening of these rooms took place in October 2005 and 115 patients were admitted to these rooms for symptom control throughout 2006. car e A comprehensive business plan to provide multidisciplinary palliative care to the Department of Medical Oncology gained hospital-wide support and was implemented in 2004 and 2005. The Board of Directors of the VU Univeristy Medical Center acknowledged palliative care as an important area of expertise to be developed within the center and therefore, the implementation of our plans took place in close collaboration with all departments involved in setting up the VUmc Expertise Centre for Palliative Care. As a preliminary result, four fully operational palliative care beds were opened on the oncology ward in 2005. For this purpose, four View of palliative care room on the oncology ward As a result of increased hospital-wide awareness of palliative care, referrals to the department’s consultant in palliative medicine increased steadily to 125 new consultations throughout 2006. Close collaboration within the hospital’s Palliative & Supportive Care Team between prof. dr. W.W.A. Zuurmond, MD, anaesthesiologist, drs. P. Doornaert, MD, radiotherapist, drs. M.S.A. Boddaert, MD, dr. M.H.M. van der Linden and B.Tuit, specialist nurse, has led V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s to the availibility of 24-hour palliative and supportive care for patients admitted to our hospital. In addition, as a courtesy to the regional Comprehensive Cancer Center Amsterdam (IKA) the hospital’s Palliative and Supportive Care Team is available for regional consultation by phone outside office hours and during weekends. Through these interdepartmental collaborations the Department of Medical Oncology is now an active participant in the VUmc Expertise Centre for Palliative Care that was launched in October 2006. Psycho-oncology car e Oncology hostesses The department’s oncology hostesses work as volunteers. In 2006 again they did a great job in accompanying and caring for 637 out of the 782 new patients of our outpatient clinic. car e The referral system between oncologists and the department’s clinical psychologist and medical social worker has been working swiftly and effectively. The number of referrals is stable: more than one third of all new patients (329 out of 782 new patients in 2006) consulted the psychosocial team (medical social worker, clinical psychologist and consultant in palliative medicine). In conformity with the ‘National Program of KWF Kankerbestrijding 2005-2010’, the psycho-oncology team started introducing a validated screening instrument to improve the referral system by which high-risk patients will be identified. Dr. M.H.M. van der Linden started a collaboration with dr. I. Verdonck-de Leeuw 15 (psychologist of the Department of Otolaryngology, Head and Neck Surgery) to investigate distress in head and neck cancer patients and their spouses. In 2006, a study was completed in which group therapy was offered to these patients and their spouses in collaboration with the Ingeborg Douwes Center. I n t e g ra l Ne n t e n t s S p e c i a l n u r s e c o n s u ltat i o n In the outpatient clinic, specialist nurses continued to provide follow-up care by phone for patients starting a new line of therapy. The frequency of weekly phone-in hours for patients and their families was increased to a daily frequency, such that they may contact a specialist nurse about specific problems or situations at hand. T r a n s m u r a l P a l l i at i v e C a r e G u i d e lines Supported by a grant from AGIS healthcare insurance company, eight transmural Palliative Care Guidelines for a variety of symptoms (bowel obstruction, pain, dyspnoea, confusion, nausea x t and vomiting, ascites) were compiled by one of the department’s senior nurses and the specialist nurse on the Palliative and Supportive Care Team. The aim of these guidelines is to inform healthcare professionals coming to the homes of our patients on the care that was initiated during admission. By doing so, we aim at improving the continuity of care and possibly prevent readmission. F a m i ly S u p p o r t P r o j e c t The Family Support Project was initiated in 2001 by prof.dr. H.M. Pinedo. It aims at improving communication between cancer patients, their family members and their doctors. Following tremendous fundraising activities, the first “Time-Out” family room on the oncology ward was opened on March 24, 2005***. Here, family members and patients have the opportunity to take a break. It is a quiet and comfortable place in close vicinity to their loved ones, where an experienced oncology nurse is available for support and for relieving concerns. So far, the Family Support Project has been a great succes. Between March 2005 and December 2006, 300 families used thr ‘Time-Out’ familiy room to celebrate birthdays and marriages, to do homework and share joyous and sad moments together in private. In 2006, the “TimeOutkamers VU Medisch Centrum” Foundation initiated new efforts to V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s finance family rooms in the outpatient clinic, the intensive care unit and on the surgical ward to expand support for the family of cancer patients throughout the center. The official opening of the second and third family room is planned to take place in 2007. n t e n t s Ne x t lso a note of thanks is due to the Ingeborg A Douwes Stichting for organizing a benefit dinner and theatre show at “Theater Carre” in June 2006, to financially support psychosocial care of patients with cancer. ** A special note of thanks is due to dr. K. Hoekman and the community of Urk, the Roparun Foundation, several individual sponsors, the Ingeborg Douwes Stichting, the Telegraaf Puzzelactie and individuals who joined a benefit dinner at the Mauritshuis Museum in April 2005 for their generous financial support. View of the “Time-Out” family room on the oncology ward S pecial notes of thanks * A special note of thanks is due to prof. H.M. Pinedo and dr. W.R. Gerritsen of the VUmc Cancer Center Amsterdam. Their continued efforts in obtaining financial support for palliative medicine and psychosocial care have enabled drs. M.S.A. Boddaert and dr. M.H.M. van der Linden to continue their work within the department as well as hospital-wide. 16 *** A special note of thanks is due to the ‘TimeOutkamers VU Medisch Centrum’ Foundation for recruiting sponsors for the Family Support Project. Through its generous donation to the VUmc Cancer Center Amsterdam the Department of Medical Oncology will be able to realise two additonal family rooms. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s x t O n c o l o g y Wa r d O A number of innovations aimed at increasing the quality of patient care as well as improving the quality control process on the oncology ward were started in 2005 and were continued enthusiastically this year. Among the activities are the assignment of a nurse specialist providing specialist support to family only. This means that one nurse works 20 hours per week as the coordinating family room nurse, while at the same time the ward’s team of nurses work on a rotation of two days each in the family room. An important development was the opening of four palliative care rooms on the ward in late 2005, which are now fully operative. Patients are admitted to these rooms for a maximum of 11 days per admission. During this period, they receive multidisciplinary treatment for multiple symptomatic palliative problems. After the patient’s symptoms have been regulated, the patient is discharged to the home situation with specialist home care, or to a palliative unit of either a nursing or residential home or one of the hospices in the Netherlands. The annual training for nurses in the fall of 2006 covered the following subjects: treatment path for colon cancer and ongoing studies, depression and coping with cancer, prevention and 17 Ne n t e n t s n c o l o g y W ar d treatment of stomatitis during chemotherapy, cachexia and nutrition with cancer and information about cetuximab. The new informative movie: “Chemotherapy, what is it?” for patients who will undergo cancer treatment, was shown. Both the movie and the training were well received. This special movie is the final project of Lisette Saveur and Marleen Westeneng who were trained as oncology nurses. It took a while before the plan could be carried out, to get consent, financing and the question whether to ask professionals or patient-actors, but once everything was cleared, the director, Bob Aardewerk, started with a lot of commitment, effort and enthusiasm. In addition to the Dutch version several foreign language versions will be prepared (Turkish, Moroccon-Arabic) and the final result will be available by May 1, 2007 for use in other institutions as well. In November 2006, the day care unit received a cheque of €30.000 from “Stichting Roparun”, which enabled the purchase of two Paxman equipments for cooling the scalp of patients. This is a highly valuable extension of our possibility to improve treatment and in particular the quality of life of certain patients, who will now suffer less hair loss caused by chemotherapy treatment. As of early 2007, this treatment will be offered to patients. S af e t y management Two projects, which are in fact the forerunners of an integrated safety management system, are ongoing. The first project concerns the safety of managing and administering chemotherapy. Three SOPs (Standard Operating Procedures) were developed at the end of this project. In doing so, stepwise checkpoints were introduced and everyone’s responsibility in the process from arranging of cystostatics, to administration to the correct patient at the correct time and in the correct dosage has been laid down in writing. The second project concerns the ‘Safe Reporting of Incidents” (SRI; blamefree reporting) project. The objective of this project is to achieve a decentralised safety management system.This is a project embedded in the national action plan from the Ministry of Health, Welfare and Sport in which 24 leading hospitals have been designated to improve care and efficiency in health care. Safety is an important part of this project. The SRI project on the ward is multidisciplinary in nature. Nurses and doctors can report (near) errors and (near) accidents in a very accessible and easy fashion. ‘Incidents’ is a V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s n t e n t s Ne x t neutral, non-accusing term. Within this system, the barriers in medical and nursing healthcare procedures are tested and analysed with regard to their functionality. Every incident reported is analysed by a multidisciplinary working group, which meets weekly. Reporting trends are addressed by means of improvement trajectories, which will either improve the barriers or place them earlier in the process such that incidents either do not reach the patient at all or do not affect the patient in a significant way. 18 V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s n t e n t s Ne x t O Out-patient clinic and day care unit u t p a t i e n t c l i n i c a n d d a y car e u n i t In 2006, 803 new patients were referred to our outpatient clinic. Thirty-six percent of our patients are from Amsterdam, 20% from the province of Noord-Holland outside of Amsterdam, and the remainder from other provinces, indicating that a considerable number of patients were willing to travel (sometimes for more than an hour) to get access to our care. The distribution of tumor types among the patients attending our outpatient clinic and day care unit is depicted in Figure 1. About one-third of the patients had a tumor of the digestive tract and another one-third of the urogenital tract. Ovarian/cervix 7% Rest 19% Lung 6% ENT 10% GU tract 18% Breast 13% GI tract 27% Since most of our patients had never visited the VU University Medical Center before being referred, all new patients visiting our outpatient clinic are escorted by a hostess, who will contact them before travelling to the hospital. They also explain our procedures to them. After consultation with a medical oncologist, the hostess arranges all further appointments for the patient. The hostess also alerts the patient to the possibility of counselling by the psycho-oncology team whenever necessary. Almost all patients were seen by our staff within one week of registration. Apart from patients seeking a second opinion, most patients preferred to be treated at the VU University Medical Center. When possible, patients often consented to participating in one of our clinical trials. The flexible attitude of all involved in the day care unit, dealing with complicated treatments, is very important for the high-quality care we offer our patients. The continuous excellent collaboration with the pharmacy department, blood transfusion service and clinical laboratories has been instrumental in achieving this. Figure 1. Distribution of tumor types among patients attending the outpatient clinic and day care unit. 19 V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Ne n t e n t s x t A Activities of the research divisions c t i v i t i e s o f t h e r e s e arc h d i v i s i o n s Division of Angiogenesis The Division of Angiogenesis aims at improving the understanding of tumor-induced angiogenesis and establishing the clinical value of anti-angiogenic therapy for cancer patients. The growth of tumors and their metastases depends on tumor-induced neo-vascularization (angiogenesis). The process of angiogenesis is initiated and maintained by tumor-derived growth factors, which are generated due to genetic abnormalities of tumor cells and by local hypoxia. Hypoxia stabilizes the HIF-1α protein, which is a transcription factor for many genes involved in angiogenesis. Among tumor-induced pro-angiogenic factors, vascular endothelial growth factor (VEGF) has been recognized as one of the most important stimulatory factors of angiogenesis. Consequently, VEGF and its receptors (mainly VEGFR2 and VEGFR1) are primary targets for anticancer therapy. Evidence that inhibition of the VEGFVEGFR pathway is of clinical benefit in cancer patients is growing. It has been shown that the monoclonal antibody bevacizumab (Avastin®), which is directed against VEGF, has activity as a single agent in renal cell cancer. Moreover, the 20 combination of bevacizumab and chemotherapy was associated with an increased number of tumor responses, prolonged time to progression and prolonged overall survival in colorectal cancer and non-small cell lung cancer. Members of the Division of Angiogenesis of Medical Oncology together with the members of the Department of Physiology after their weekly common Thursday morning meeting In addition, the combination of bevacizumab and an anti-EGFR agent appeared to be synergistic in patients with advanced lung cancer. Finally, several tyrosine kinase (TK) inhibitors of VEGFR-2 have shown impressive activity in renal cell cancer (i.e. sunitinib). VEGF is a dominant factor in the biology of this tumor type. These encouraging data have led to a sharp increase in the number of clinical trials evaluating antiVEGF and anti-VEGFR agents with or without other biological agents, chemotherapy and/or radiotherapy. Clinical trials with second- and third-generation VEGFR and VEGF inhibitors are being performed. These clinical trials are accompanied by side studies to learn more about the effects of anti-angiogenic agents on their target, the tumor vasculature’s endothelium as well as on host tissue (e.g. the normal vessel endothelium). These side-studies include investigations of circulating endothelial progenitor cells, immunological parameters, endothelium-specific marker proteins and complete protein profiles (proteomics). In addition, effects of antiangiogenic therapy on the normal physiology of vessels (regulation of the blood pressure) are being studied and new imaging techniques are being used to monitor anti-tumor effects. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s The specific research projects of the division are: • Clinical trials of anti-angiogenic agents •Circulating endothelial cells (CECs) and endothelial progenitor cells (CEPs) • Role of HIF-1 in tumor-induced angiogenesis • Angiogenesis in ovarian cancer •Integrative tumor cell biology for improvement of cancer treatment Clinical trials of a n t i - a n g i o g e n i c ag e n t s In 2006, all data collected in the phase I clinical trial of SU014813, carried out by prof. dr. E. Boven in collaboration with the Hamburg University Hospital, Germany, were evaluated for toxicity and efficacy. SU014813 is an oral TK inhibitor of the VEGF receptors 1 and 2, PDGFRβ, c-Kit and Flt-3. A total of 77 patients have been included in the trial, 38 at a schedule of 4 weeks on treatment and one week off, and 39 at a schedule of continuous daily dosing. Most side effects were, in general, well tolerable. Few grade 3-4 toxicities occurred and the ones that did occur were mainly obeserved with the continuous dose schedule: diarrhea and fatigue were most prominent. Twelve patients responded to SU014813, while 32 patients experienced stabilization of disease. Thirteen patients were on treatment for a period > 12 months. Presently, an international phase II trial is ongoing with SU014813 being administered at 100 mg per day 21 Ne n t e n t s continuously in patients with advanced breast cancer, who were previously treated with a taxane and an anthracycline. Antitumor activity has been noticed in several patients, including patients treated by our department. An extended access program of SU011248 (sunitinib; Sutent®) in advanced renal cell cancer was coordinated by prof. dr. E. Boven and dr. A.J.M. van den Eertwegh and was carried out by A.A.M. van der Veldt, MSc,) in collaboration with the group of dr. J. Haanen, Netherlands Cancer Institute, Amsterdam. A total of 83 patients were treated, including elderly patients age and patients with poor performance status. Several patients, especially those with clear cell carcinoma histology, responded to the drug for an extended period of time. Of interest, some patients with their primary tumor still in situ, known to be resistant to cytokine therapy, showed massive tumor necrosis upon treatment with sunitinib. Several side-studies were done to obtain more insight in the mechanism of action of sunitinib, in particular a possible role of circulating endothelial cells and circulating dendritic cells as well as the mechanism of hypertension. The data are currently being analysed. Presently, a phase I clinical trial of the combination of sunitinib and irinotecan in patients with advanced solid malignancies is x t ongoing to determine the doses that can be safely combined (prof. E. Boven in collaboration with Institut Gustave Roussy, Paris, France). H. van Cruijsen, MD (under the supervision of dr. K. Hoekman and dr. T. de Gruijl) has performed a phase I/II study with escalating doses of AZD2171, an oral TK inhibitor of VEGFR-2, PGDFR-β and c-kit, in combination with 250 or 500 mg of an oral TK inhibitor of EGFR (gefitinib). This study is conducted in cooperation with the Department of Medical Oncology, University Medical Center Utrecht and the Department of Medical Oncology, Radboud University Nijmegen Medical Center. Side-studies in this trial included dynamic CT imaging of tumors in a subset of patients and measuring effects of the therapeutic agents on circulating dendritic cell subpopulations (for details of the results, see Division of Immunotherapy). V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s C i r c u l at i n g e n d o t h e l i a l c e l l s ( CEC s ) a n d e n d o t h e l i a l p r o g e n i t o r c e l l s ( CEP s ) Testing novel anti-angiogenic drug combinations in the clinic is hampered by the lack of suitable methods for target validation and efficacy. Strong evidence from preclinical studies suggests that the number of circulating endothelial cells originating from the (tumor) vasculature (CECs) may serve as an early indicator of anti-angiogenic and/or antivascular effects. In addition, in preclinical models, circulating endothelial progenitor cells (CEPs), which originate from the bone marrow, have been shown to contribute to neo-vascularisation. Therefore the quantification of pre-treatment numbers of CEPs and CECs and changes in these number during therapy might be markers of efficacy of anti-angiogenic therapy. Currently, there is no validated protocol to measure these cell populations longitudinally in cancer patients. Supported by a grant from the EU-FP6 program “Angiotargeting” (dr. H.J. Broxterman, dr. K.Hoekman, prof. dr. V.W.M. van Hinsbergh), we have designed a flow cytometric method for the quantification of populations of CECs and CEPs in peripheral blood of cancer patients in clinical trials. Since almost all anti-angiogenic agents and combinations presently under study in the clinic target VEGF or its receptor VEGFR-2, 22 Ne n t e n t s we have in particular focussed on measuring cell populations expressing VEGFR-2. A study protocol has been set up to establish the reliability and reproducibility of a 4-colour flow cytometry procedure to measure the frequencies of subpopulations of cells with endothelial and progenitor markers, including CD34, AC133, CD146, CD31, and CD105, in combination with VEGFR-2 as well as CD45 (to exclude hematopoetic cells). L. Vroling, MSc, found, when comparing 20 healthy volunteers and 14 patients with advanced cancer, that the population of CD45negCD34brightCD133neg CD146posVEGFR-2pos cells was increased in the cancer patients. This novel “small EC-like” cell population is currently being investigated further in vitro and in clinical trials in patients with non-small cell lung cancer (NSCLC) treated with bevacizumab and the EGFR blocker erlotinib and patients with renal cell cancer treated with sunitinib for its potential as a biomarker of anti-angiogenic, antitumor effects. Another population of circulating cells, previously identified in a number of studies as CECs, has been shown to exist mostly of large platelets. Other studies in colorectal and non-small cell lung cancer patients are being planned. Emerging evidence suggests that human blood contains bone marrow (BM)-derived endothelial progenitor cells (EPCs) that contribute to x t postnatal neovascularization. Clinical trials demonstrated that administration of BM-cells may enhance neovascularization. Most studies, however, used crude BM cell populations. Identifying the role of different cell populations is important for developing improved cellular therapies. L. Vroling, MSc, and M. Weij have set up a culture technique for so-called “lateoutgrowth endothelial progenitor cells”, from human cord blood. These cells are characterised by a typical cobblestone-like appearance, and have a marker profile consistent with endothelial cell lineage (CD45neg CD14neg CD31bright CD146pos CD144pos). These cells also express EGFR and will therefore be used to investigate the effects of combined EGFR and VEGFR inhibition (e.g. bevazicumab with ertolitinib) on EPCs as models for tumor endothelial cells. Role of HIF - 1 in tumor-induced angiogenesis H ypoxia I nducible F actor -1 (HIF-1) in breast and colon tumors HIF-1 is an important transcription factor that stimulates tumor growth and metastasis via several pathways, including the induction of angiogenesis-regulating growth factors VEGF and SDF-1. Activation of HIF-1 depends on the presence of its α-subunit, which in the presence of oxygen is rapidly degraded after V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s proline hydroxylation and subsequent interaction with von Hippel Lindau (VHL) protein in the proteasome. Hypoxia increases HIF-1α levels by inhibiting prolyl-hydroxylase-mediated hydroxylation and thereby preventing HIF-1α degradation. In line with such a mechanism we found that silencing prolyl-hydroxylase-2 in endothelial cells by siRNA resulted in sustained activation of HIF-1α. The expression of HIF-1α-induced genes was also investigated in biopsies of colon carcinomas, adenomas and non-malignant colon biopsy specimens. This was done in collaboration with prof. G.A. Meijer (Department of Pathology, VU University Medical Center), supported by an AEGON fellowship. HIF-1α was clearly demonstrated in colon tissue and its correlation with hypoxia, GLUT-1, CAIX and other proteins was further investigated. Special emphasis was given to tumor vascularization and the occurrence of genetic instability in the patients’ biopsies. A mouse colon tumor model was set up to investigate the role of angiogenesis, hypoxiaassociated factors (including HIF-1α) and monocytes in the progression of colon tumor (dr. R. Fijneman). E xperimental studies on hypoxia and HIF-1α In collaboration with dr. M.A. Engelse 23 Ne n t e n t s (Department of Physiology, VU University Medical Center), in vitro parameters of angiogenesis were studied for prolonged periods under various oxygen concentrations. mRNA expression profiles of human macro- and microvascular endothelial cells kept at hypoxic, normoxic and ambient air atmosphere for several months were analyzed. A selection of the differentially expressed genes is currently investigated with respect to their possible role in angiogenesis. A n g i o g e n e s i s i n o va r i a n c a n c e r In a project carried out by M. Bijman, MSc, the anti-angiogenic properties of microtubuletargeting agents have been studied. Earlier, it was shown that docetaxel, epothilone B and vinblastine at non-toxic concentrations efficiently inhibited functional properties of endothelial cells, while cisplatin and doxorubicin were not effective. It now appeared that docetaxel also interferes with actin dynamics of human ovarian cancer cells, but does not significantly inhibit microtubule dynamics. Again, cisplatin and doxorubicin were not effective. Combining docetaxel with cetuximab (an inhibitor of EGFR) and trastuzumab (an inhibitor of HER2) or pertuzumab (an inhibitor of dimerization of HER2 with EGFR or HER2 with HER3) resulted in increased antitumor effects in ovarian cancer cells. x t In a project carried out by dr. M. Janmaat, the focus was on the role of platelet-derived growth factor (PDGF) signalling in ovarian cancer. PDGF signalling is not only recognized as an important process in angiogenesis, as it is a growth factor for the development of the supporting cells in the vasculature, the pericytes, but also as a factor involved in the tumor-associated fibroblast (stroma) response. Our data were consistent with such a paracrine role, while autocrine PDGF signalling in ovarian cancer cells appeared of no importance for tumor proliferation and migration. I n t e g r at i v e tumor cell biology for i m p r o v e m e n t o f c a n c e r t r e at m e n t Starting from the moment of anticancer drug administration to a patient, many processes are involved in generating the overall tumor response to the drug, such as pharmacokinetics, drug metabolism, drug transport to the target and interactions of the drug with the cellular biochemical machinery, including cellular defense (such as damage repair). Advanced knowledge on the integration of the dynamic responses of all these processes will lead to better treatment and better individual tailoring of anti-cancer treatments. Parts of these processes have been mimicked and modelled in vitro using breast cancer cells in tissue culture and in silico (prof. dr. J. Lankelma) in a way that simulates treatment of V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s breast cancer with doxorubicin. Another project aims at the design of methods for monitoring tumor responses dynamically in patients, because sensitive and selective biomarkers for this purpose are currently lacking. To search for new biomarkers in urine, we are developing a micro-HPLC system with post-column derivatization, rendering peptides fluorescent, followed by laser-induced fluorescence detection. The method has been applied in five patients with colorectal cancer, treated with bevacizumab and chemotherapy. We have collected urine samples two days before and three weeks after the start of treatment and we have observed dynamically changing chromatographic peaks during this time period. More information on the identity of these peaks will be obtained by tandem mass spectrometry. Division of G e n e T h e ra p y Research by the Division of Gene Therapy focuses on the use of recombinant viruses as platforms for new anti-cancer therapies. Currently, the main research aims are to selectively target cancer cells and to enhance cancer cell killing by selectively replicating viruses. The final goal is to translate new gene-based treatment modalities into clinical applications in cancer patients. In addition, the Gene Therapy Laboratory houses the newly established CCA/V-ICI core facility for 24 Ne n t e n t s functional oncogenomics research by RNA interference library screening (RIFOL). anti-tumor activity was obtained from bone scan improvement, measurable tumor regression and improvement in bone pain. Interestingly, clinical responses were associated with endocrine-related immune breakthrough events that were treatable with standard hormone replacement therapy. Members of the Division of Gene Therapy. Gene therapy clinical studies Currently, one clinical study is ongoing as a combined effort of the Divisions of Gene Therapy and Immunotherapy and the Departments of Urology of the VU University Medical Center and the Antonie van Leeuwenhoekhuis/Netherlands Cancer Institute, Amsterdam. This phase I study, sponsored by Cell Genesys and Medarex, involves multiple vaccinations with a fixed dose of lethally irradiated GM-CSF-expressing prostate cancer cells and dose escalation of a human antibody against CTLA-4 of patients witt metastatic, hormone-refractory prostate cancer. This year, enrolment of the dose escalation cohorts was completed. Dr. W.R. Gerritsen presented preliminary results at the 2006 ASCO Annual Meeting. At higher dose levels, partial PSA responses were observed. Additional evidence for x t In our preclinical studies, we found that the CRAd Ad5-∆24RGD can be successfully applied in the treatment of glioblastoma multiforme. Researchers from the MD Anderson Cancer Center confirmed the potential of this virus in an intracranial tumor model. We combined our efforts with the MD Anderson group and with prof. D.T. Curiel (University of Alabama, Birmingham, AL). Jointly, we applied for a grant from the Rapid Access to Intervention Development (RAID) program of the U.S. National Cancer Institute. In 2004, we received final confirmation of approval of the grant. In 2005, a clinical-grade virus batch was prepared under the RAID program. This year, toxicology studies were done and the final report on these studies is expected in Q1 of 2007. The clinical study that will be done in Amsterdam will involve convection-enhanced delivery of Ad5-∆24RGD into the tumor bed after surgical resection of recurrent glioma. In the phase I dose escalation part of the study, we will investigate possible toxicity of this procedure. Next, we will perform a modified phase II study to investigate the possible V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s anti-tumor efficacy of the treatment. We expect to start the clinical study in the second half of 2007. G e n e t h e r a p y p r e c l i n i c a l r e s e a rc h The main areas of preclinical research of the Division of Gene Therapy are: •Tumor-specific targeting of recombinant viruses •Augmentation of virus-mediated cancer cell killing •Preclinical evaluation of gene-based (combination) treatments T umor - specific targeting of recombinant viruses We are exploring the utility of adenoviral vectors (AdV), conditionally replicative adenoviruses (CRAds), and recombinant coronaviruses as anti-cancer agents. To make these viruses most effective and selective for cancer cells, we target their infection specifically to surface molecules that are highly expressed on tumor cells. Previously, we successfully redirected the binding of AdV to a variety of tumor cell types by using adapter molecules that on one side bind to the AdV capsid and on the other side bind to a cell surface protein of choice. We also extrapolated this two-component targeting approach to CRAds. Recently, we showed that these targeted CRAds could be made very specific for cells expressing the target molecule by introducing 25 Ne n t e n t s two mutations in virus capsid genes. Currently, we are using the adapter targeting strategy together with the Department of Otolaryngology (dr. R.H. Brakenhoff and H.J.T. van Zeeburg; supported by the Dutch Cancer Society) to develop targeted CRAds for early intervention of pre-malignant oral and oropharyngeal lesions. Potential target molecules that are highly expressed on squamous cells were already identified and their utility to mediate adenovirus infection via antibody binding was confirmed. In addition, together with the Virology Division of Utrecht University, we constructed targeted recombinant murine and feline coronaviruses for selective lysis of human cancer cells (dr. H. Verheije and dr. T. Würdinger; supported by the Dutch Cancer Society). Several findings provided interesting leads for further development of recombinant coronaviruses as anti-cancer agents. Coronaviruses could be targeted to cell surface molecules by incorporating genes encoding the appropriate adapter molecules into their genome. These viruses replicated in human cells expressing the target receptor and killed these cells via syncytium formation. Another interesting observation was that acute myeloid leukemia cells could be infected via their Fc receptor using an antibody directed against the coronavirus spike protein. Most of these studies are described in the thesis of dr. Würdinger, “The development of tumor-selective coronaviruses as anti-cancer agents”, which he successfully x t defended at Utrecht University on February 1, 2006, and for which he received the Greiner Award of the Dutch Society of Gene Therapy. Despite the success of targeting via bispecific adapter molecules, it would be preferred to develop targeted AdV and CRAds as singlecomponent genetic medicines. This requires the physical incorporation of specific tumortargeting ligands into the viral capsid. Based on the close resemblance of the adenovirus fiber protein and the reovirus attachment protein σ1, dr. F.H.E. Schagen designed a fiber/σ1 fusion protein lacking all known native binding sites. A specific binding ligand (His) for an artificial receptor molecule (HisR) was incorporated (Figure 2). Cells expressing the receptor were transduced up to 50-fold more efficient than parental cells lacking the receptor. Importantly, the targeted AdV exhibited 1,000 to 10,000-fold reduced infection of liver cells and did not bind to human red blood cells. Moreover, following systemic administration to mice, the virus showed prolonged persistence in the circulation and reduced transduction of all organs tested. Together with the Division of Immunotherapy (dr. T.D. de Gruijl and dr. D. Oosterhoff) we are currently exploiting this targeting platform to develop AdV targeted towards dendritic cells. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s A ugmentation of adenovirus - mediated cancer cell killing Figure 2. Targeting AdV by genetic modification of the AdV capsid. (A) Parental 293 cells and derivative 293-HisR cells, expressing an artificial receptor for His-tagged molecules, were transduced with an AdV with native tropism (AdGL) or with a similar AdV carrying His-tagged fiber/σ1 fusion proteins (AdGLHisTar). Transduction was analyzed by fluorescence microscopy for GFP reporter gene expression. AdGLHisTar efficiently transduced 293-HisR, but not 293 cells. (B) 293-HisR cells were transduced as in A, after pre-incubating the AdV with monoclonal antibodies directed against the fiber knob domain or the His-tag as indicated. Whereas the anti-fiber knob antibody neutralized transduction by native AdGL, the anti-His antibody neutralized transduction by targeted AdGLHisTar. This confirmed that targeted transduction was mediated via the His targeting-ligand. 26 Ne n t e n t s CRAds are promising new agents with potential utility to treat cancer. Adenoviruses kill host cells by exploiting multiple cell death mechanisms of which a p53 tumor suppressor protein-dependent pathway appears to be important for rapid lysis. As the p53 pathway is dysfunctional in most cancers this limits the efficacy of CRAd-based therapies. We therefore set out to develop more effective oncolytic agents by restoring functional p53 expression during CRAd replication in cancer cells (Dr. V.W. van Beusechem; supported by the Royal Netherlands Academy of Arts and Sciences). In previous years, we showed that a CRAd expressing wild type p53 killed most tested human cancer cell lines, primary tumor specimens, and tumor xenografts in vivo more effectively. The anti-cancer effects of CRAdp53 were augmented by chemotherapy and radiotherapy. We also made two tailored CRAds for more effective treatment of cancers with high expression of the p53 inhibitors MDM2 and HPV E6 protein, respectively. For this, we used p53 variants that do not bind to these inhibitors. Recently, we found that CRAd-p53 is also more effective in combination with the specific MDM2 antagonist Nutlin-3 (H.C.A. Graat; Department of Orthopedic Surgery). Together, these observations showed that oncolytic adenovirus therapy is most effective if p53 is x t expressed and p53 inhibitors are eliminated. The biotechnology start-up company ORCA Therapeutics obtained an exclusive license on our CRAd-p53 technology. Currently, we are pursuing the further development of p53expressing CRAds for early clinical evaluation together with ORCA Therapeutics. An entirely new way to augment the anti-cancer potency of CRAds could be by suppressing virus inhibitory molecules in cancer cells. To this end, we are exploring the utility of RNA interference (RNAi). RNAi is a very specific mechanism to silence gene expression by sequence-specific mRNA degradation. We already constructed CRAds expressing short hairpin RNAs that can trigger RNAi. These CRAds caused efficient and specific silencing of a target gene in cancer cells. Currently, we are focusing our attention to silencing inhibitors of p53. Our final goal is to identify virus inhibitory molecules in cancer cells by a functional genomics approach. This year, we obtained a research grant from the Technology Foundation STW (dr. V.W. van Beusechem and dr. W.R. Gerritsen) for this project. This research will benefit from the initiative to establish the RNA Interference Functional Oncogenomics Laboratory (see below). V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s P reclinical evaluation of gene - based ( combination ) treatments To translate new virus-based treatment modalities into clinical applications in cancer patients, we investigated the efficacy of adenoviruses in preclinical models of osteosarcoma and glioblastoma multiforme. Dr. M.A. Witlox (Department of Orthopedic Surgery) completed her studies on the utility of adenovirus gene transfer vectors and oncolytic adenoviruses for the treatment of osteosarcoma (OS) by defending her thesis entitled “Gene therapy of osteosarcoma. Preclinical studies with adenoviral vectors and adenoviral oncolysis”on December 15, 2006. An important finding of these studies was that the primary receptor for adenovirus, CAR, is hardly expressed by most osteosarcomas. Therefore, effective gene therapy or virotherapy of OS requires targeted virus entry via alternative cell receptors. In particular, integrin molecules were found very useful for this purpose. Because metastatic disease, in particular to the lungs, is the major threat to patients with OS, H.C.A. Graat (Department of Orthopedic Surgery) focused his studies on treating OS metastases. In collaboration with prof. E.S. Kleinerman (Division of Pediatrics, M.D. Anderson Cancer Center, Houston, TX, USA) he set up a mouse model of human OS lung metastasis. Intravenous delivery of the integrintargeted CRAd Ad5-∆24RGD to mice with 27 Ne n t e n t s established OS lung metastases decreased the number of lung metastases and total lung weight, strongly suggesting that systemic treatment of OS with CRAds is feasible. Previously, we found that the CRAd Ad5∆24RGD kills glioblastoma cells effectively in vitro and in vivo. Furthermore, the efficacy of this treatment was enhanced by combining it with irradiation. This year, dr. M.L.M. Lamfers (Department of Neurosurgery) assessed the effects of these treatments in an intracranial glioma animal model. Surprisingly, and in contrast to findings in cell culture and in subcutaneous glioma tumors, combination treatment was not significantly more effective than treatment with virus alone. Apparently, glioma tumors respond differently to treatments in their natural brain environment than when placed in an ectopic environment. Another interesting observation made by dr. Lamfers in collaboration with the group of prof. E.A. Chiocca (Molecular Neuro-oncology Laboratories, Massachusetts General Hospital, Charlestown, MA, USA) was that the immune system appears to reject oncolytic adenovirusinfected glioma cells in the brain. She monitored virus replication in an intracranial mouse model of malignant glioma, using a tumor-selective adenovirus expressing luciferase and in vivo bioluminescence imaging. Luciferase expression x t was lost rapidly, whereas a replication defective control vector tested in the same model demonstrated stable luciferase expression. In immunodeficient NOD/SCID mice, luciferase expression by the oncolytic adenovirus was lost less rapidly, suggesting a role for the immune system. This was corroborated by the observation of infiltrated inflammatory cells in injected tumors. Treatment of mice with the immunomodulator cyclophosphamide, which decreased tumor infiltration by immune cells, prolonged transgene expression. Thus, intracranial anti-tumor efficacy of oncolytic adenovirus may be hampered by an immune response against the replicating virus, which can at least partially be circumvented by pretreatment with immunomodulating agents. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s RNA I n t e r f e r e n c e F u n c t i o n a l O n c o g e n o m i c s L a b o r at o r y On June 19, 2006, during the opening event of the new research building of the VUmc Cancer Center Amsterdam (CCA), Dr. V.W. van Beusechem received a research award from the Foundation SBGO to introduce a new platform technology, i.e. RNA interference library screening. The biological process of RNA interference (RNAi) provides a wealth of opportunities for functional oncogenomics research. Using RNAi, the genetic cause of a particular function can be assessed by individually suppressing gene products in cancer cells and studying changes in the behavior of these cells. Pinpointing gene products that are functionally associated with aberrant processes in cancer cells contributes to a better understanding of cancer cell biology as well as to identifying targets for therapy. To provide researchers at CCA/V-ICI with facilities to conduct RNAi library screens, a core 28 Ne n t e n t s facility was established in the Gene Therapy laboratories of the Department of Medical Oncology. This RNA Interference Functional Oncogenomics Laboratory (RIFOL) provides researchers access to the Dharmacon siARRAY Human Genome Library. This library includes siRNA reagents that target over 21,000 unique genes of the human genome. RIFOL maintains equipment to support mammalian cell-based medium-throughput screening efforts. The RIFOL user group, with members from several CCA/V-ICI departments, provides an internal forum for researchers to share protocols and exchange screening data. Furthermore, RIFOL joined the Genome-Wide RNAi Global Initiative, an alliance of Dharmacon, Inc. and leading international research centers pioneering the use of whole-genome RNAi screening. Interactions with this community of researchers exploiting this exiting new technology should be very helpful to advance the productivity of our research. Division of I m m u n o t h e ra p y The Division of Immunotherapy is a close collaboration between the Departments of Medical Oncology and Pathology. Under the supervision of dr. A.J.M. van den Eertwegh and prof. dr. R.J. Scheper, the division aims at translating preclinical studies into new x t immunological approaches for the treatment of cancer. In 2006 the division experienced considerable growth, both in terms of number of people and diversity of research subjects. Members of the Division of Immunotherapy paying tribute to prof. H.M. Pinedo. In 2006, dendritic cell (DC)-targeted tumor vaccination remained a major focus of research, in which we have a long-standing collaboration with the division of Gene Therapy (dr. W.R. Gerritsen and dr. V.W. van Beusechem) and the Division of Human Gene Therapy of the University of Alabama at Birmingham (UAB; dr. D.T. Curiel). Further topics of research were the suppressive effects of tumors on DC differentiation and maturation, potentiation of DC functions in vivo, chemo-immunotherapeutic approaches, the role of ABC-transporters in DC physiology, the development of DC lines for V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s semi-allogeneic vaccination, and identification of novel tumor-associated antigens for vaccine development. The division was also involved in monitoring of DC and tumor-specific T cell function in several clinical immunotherapeutic studies. Lines of research within the division of immunotherapy: • Tumor cell vaccination • Immunopotentiation • Dendritic cell-based therapies • NKT cell-based therapies Tumor c e l l va c c i n at i o n A utologous tumor cell vaccination in renal cell carcinoma In 2004 we started a phase II study in metastatic renal cell carcinoma, vaccinating patients with an autologous whole tumor cell vaccine in combination with Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF) and PF3512676 (formerly known as CPG 7909). GMCSF recruits and activates myeloid dendritic cells (MDC), while bacterially derived un-methylated CpG sequences like PF-3512676 activate plasmacytoid DC (PDC) and further boost both natural killer (NK) and cytotoxic T lymphocyte (CTL) responses. This study was coordinated by dr. A.J.M. van den Eertwegh. Vaccine 29 Ne n t e n t s preparation was performed according to current Good Manufacturing Practice (cGMP) in our vaccination facility headed by dr. E. Hooijberg. The first three induction vaccinations were given weekly with all subsequent booster vaccinations given every three months. After completing tumor cell vaccinations, patients were treated with subcutaneous injections of PF-3512676 once every two weeks and IFN-α subcutaneous three times per week with the aim of further enhancing innate immunity and the effector phase of the adaptive immune response. Primary endpoints of this study were response rate and feasibility, and secondary endpoints were toxicity, efficacy of immunization and survival. This year we included the 12th patient in the study, a patient with metastatic renal cell carcinoma. All patients tolerated the vaccines very well (no ulceration) and after 3 vaccinations a significant DTH response against the autologous tumor cells was observed in all patients, indicating that a specific tumor response was induced. In addition, we observed a partial remission in 25% of patients. Clinical follow-up is ongoing. A llogeneic tumor cell vaccination and anti -CTLA-4 in prostate cancer A phase I/II clinical trial started last year (led by dr. W.R. Gerritsen and dr. A.J.M. van den Eertwegh), studies the anti-tumor activity of a prostate specific immunotherapy based on two x t Granulocyte/Macrophage-Colony Stimulating Factor (GM-CSF)-transduced allogeneic cell lines (Prostate GVAX®) combined with the immunomodulatory anti-CTLA4 antibody ipilimumab in men with metastatic, hormonerefractory prostate cancer. The anticipated synergy of these novel agents may lead to augmented T cell-mediated anti-tumor immunity via improved dendritic cell function, and blockade of inhibitory feedback loops in tumorspecific T cells. Preliminary results showed PSA responses in 5/6 patients at the two highest ipilimumab dose levels, as well as regressing of bone and lymph node metastases, and improvement of bone pain in three of these five PSA responders. S. Lougheed and A. Stam receive a shipment of cell lines from Cell Genesys Inc. for the GVAX and ipilimumab immunomonitoring studies. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Under supervision of dr. T.D. Gruijl and S.J.A.M. Santegoets, MSc, we are monitoring dendritic and T-cell function in these patients to identify changes that correlate with clinical efficacy. The first results convincingly showed generalized T cell activation and tumor-specific serological responses following therapy, in conjunction with profound decreases in PSA serum levels. To increase the chances of tumor-specific T cell detection, both blood and injection sites are monitored for T cell reactivity. These analyses are expected to yield valuable data concerning immune effects achieved by the GVAX/ipilimumab combination. The first promising clinical data from the phase I study were presented by dr. W.R. Gerritsen at the 2006 annual meeting of ASCO in Atlanta. The accrual of 16 additional patients for the phase II trial is currently ongoing. I m m u n o p o t e n t i at i o n L ocoregional treatment with GM-CSF and PF 3512676 (CPG7909) of melanoma patients Early melanoma development is accompanied by impaired immune effector functions in the initial tumor-draining lymph node, the so-called sentinel lymph node (SLN). Most notably, a reduced frequency and activation state of DC may well interfere with the activation of anti30 Ne n t e n t s tumor effector T cells. In collaboration with the Department of Oncological Surgery (prof. dr. P.A.M. van Leeuwen and dr. S. Meijer), we have performed two phase II studies of clinically stage I/II melanoma patients who received either GMCSF or the CpG–B oligodinucleotide (ODN) PF3512676 intradermally around the excision site of the primary melanoma. Both treatments resulted in increased frequencies of tumor antigenspecific CD8+ T cells in the corresponding SLN. The observed rise in melanoma-reactive CD8+T cells upon GM-CSF administration was associated with an increased frequency of (most likely skin-derived) CD1a+CD83+ myeloid DC in the SLN. In contrast, rises in frequency of melanoma-reactive CD8+ T cells, observed upon CpG-B ODN administration in both SLN and blood, were associated with the activation of SLN-resident plasmacytoid DC rather than of CD1a+ myeloid DC. In addition, CpG-B administration was associated with significantly lower Treg frequencies as well as the presence of a newly identified CD11chiCD123+CD83+ mature myeloid SLN-DC subset, further characterized by the expression of TRAIL, CCR7, and high levels of co-stimulatory molecules. These studies thus provide insight into the different DC subsets that are present in melanoma-draining lymph nodes and how these may be targeted in order to activate tumor-reactive CD8+ T cells. The latter appear to have been recruited to the x t SLN in early stages of melanoma development but have remained functionally “dormant”. Clinical strategies aiming at immune modulation of the SLN may help to (re)activate these tumorreactive T cells and thus afford local as well as systemic control of metastatic outgrowth. On September 29, 2006, dr. R.J.C.L.M. Vuylsteke successfully defended his PhD thesis on the GMCSF studies described above. By the end of 2007, B.G. Molenkamp is expected to defend a thesis on the PF-3512676 studies. Meanwhile, B.J.R. Sluijter, MSc, has just concluded the accrual of patients in our third phase II trial, comparing the effects of combined local administration of PF-3512676 and GM-CSF with the effects of PF3512676 alone. N eoadjuvant chemotherapy and GM-CSF in breast cancer Putative beneficial effects of GM-CSF administration were also studied in conjunction with neoadjuvant chemotherapy in patients with locally advanced breast cancer (LABC) in an international multicenter phase III trial (Spinoza Trial, initiated by prof.dr. H.M. Pinedo). We hypothesized that improved survival might result from chemotherapy-induced tumor antigen release in combination with long-term preservation of the tumor-draining lymph nodes and GM-CSF-induced DC mobilization and activation. GM-CSF may correct the V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s hampered development and functioning of DC in these patients while chemotherapy-induced tumor cell death may decrease the levels of tumor-derived immunosuppressive cytokines. These circumstances should facilitate optimal activation of tumor-specific T cells with the capacity to eradicate both primary tumor cells and micrometastases. Phenotypic and functional DC and T cell studies were performed in peripheral blood and TDLN biopsies over the course of treatment by dr. T.D. de Gruijl, S.M. Lougheed, MSc, and A.M. Stam. We found significant decreases in myeloid DC frequencies in the peripheral blood of LABC patients at the start of treatment, as compared to sex- and age-matched healthy controls. During treatment these frequencies increased in LABC patients, finally even exceeding levels observed in healthy controls. Myeloid DC frequencies were significantly increased in TDLN from GM-CSFtreated patients in comparison to G-CSF-treated patients. Interestingly, CEA, Her-2/neu, and/or survivin-specific CTL responses were detected in TDLN in two out of four tested patients, but not in the blood of the same patients. This is an indication of local specific CTL priming. The long-term clinical follow-up analysis is ongoing. 31 VEGFR inhibition : DC differentiation Defective systemic DC differentiation, maturation and function is one of the mechanisms underlying impaired anti-tumor immunity in cancer patients. Tumor-derived, vascular endothelial growth factor (VEGF) has been implicated as a major DC-suppressive factor. In a collaborative effort with the Division of Angiogenesis (dr. K. Hoekman and prof. dr. E. Boven), H. van Cruijsen, MD, is performing clinical and preclinical studies to investigate the potential beneficial effect of VEGF receptor (VEGFR) tyrosine kinase (TK) inhibitors on defective DC differentiation in cancer patients. Peripheral blood DC (PBDC) precursor and subset frequencies were measured in patients with advanced renal cell cancer before and after treatment with the anti-angiogenic, VEGFR-TK inhibitors AZD2171 or sunitinib. It was found that a population of immature myeloid cells (ImC) and a population of myeloid suppressor cells (MSC) was significantly increased, while more mature myeloid DC precursors (pMDC) and specifically the myeloid DC subsets-1 (BDCA-1+/CD1c+) and -2 (BDCA-3+) were significantly reduced in the blood of cancer patients as compared to healthy subjects. Remarkably, plasmacytoid DC (BDCA2+) were also significantly lower. After treatment with AZD2171, pMDC frequencies did not increase, while sunitinib treatment did normalize effects on Ne n t e n t s x t pMDC values in the blood of patients with advanced renal cell cancer. ABC transporters and DC functioning Looking for new ways to optimize DC-based cancer immunotherapy, we are investigating possible roles of ATP-binding cassette (ABC) transporters in physiological DC processes such as differentiation and migration. These ABC transporters are involved in multidrug resistance (MDR) in patients undergoing chemotherapy. Knowledge of the possible role of these transporters in immune effector functions may aid in the rational design of future chemoimmunotherapeutic approaches. R. van de Ven, MSc, works on a NKB-funded project. From April to August 2006, she visited the laboratory of prof. dr. D.T. Curiel to construct adenoviral vectors encoding siRNAs for various ABC transporters. These studies have so far uncovered roles for MRP1 in DC differentiation, for MRP4 in DC migration and for BCRP in Langerhans cell (LC) differentiation. Modulation of the expression of these ABC transporters to enhance DC development and function (e.g. by cytostatic drugs) may be a novel way to support DC-based tumor immunotherapy approaches. R. van de Ven received a Young Investigator’s Award for this work at the 2006 Annual Meeting of the American Association for Cancer Research (AACR), held in Washington DC. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Dendritic In Co e v i ou s cell-based therapies situ targeting and modulation of dendritic cells for immunotherapy : reversal of suppression Vaccination with DC generated in vitro holds great promise, but is hampered by suboptimal migration of DC to the draining lymph nodes. Direct in vivo targeting of vectors carrying the genetic code of tumor antigens to DC may thus present an attractive alternative. Unfortunately, in cancer patients, hampered DC differentiation and activation has been reported. High systemic levels of tumor-derived suppressive cytokines like IL-10, IL-6 and VEGF have been shown to interfere with proper DC function, resulting in tolerance rather than immune activation. In a Zon/MW-funded VIDI project awarded to dr. T.D. de Gruijl, dr. D. Oosterhoff and J.J. Lindenberg, MSc, are investigating ways to target DC in situ, while bypassing or reversing tumorinduced immune suppression. A human skin explant model is used to study effects of suppressive factors on DC migration and antigen presentation. A shift from mature CD83+ DC to immature CD14+BDCA3+ macrophage-like cells was observed within seven days after migration from IL-10 injected skin. These macrophage-like cells displayed poor T cell-stimulatory ability and lacked 32 Ne n t e n t s expression of CCR7, thus precluding their migration to T cell areas in lymph nodes. Of note, DC migrating from breast tumor-overlying skin samples underwent a similar accelerated DC-to-macrophage switch. Recent studies have demonstrated that hyperactivation of the Jak2/STAT3 pathway by the suppressive cytokines IL-10 or IL-6 might be responsible for hampered DC differentiation in cancer. In order to prevent (or revert) abnormal differentiation into suppressive DC in cancer patients, we have constructed a panel of replication deficient adenoviral (Ad) vectors expressing both GFP and short hairpin RNAs directed against different key proteins of the Jak/STAT pathway, such as STAT3. These adenoviral vectors are used to transduce suppressed DCs; effects on DC phenotype and function are being studied. In order to identify useful targeting motifs on DC in human skin explants and tumordraining lymph node samples, we will use a screening system based on Ad vectors in which a Staphylococcus aureus protein A domain is incorporated, that can bind the Fc domain of immunoglobulins. This system allows the utilisation of antibodies directed against DC surface markers to test their efficacy as targeting molecules for high-efficiency transduction and simultaneous activation of the targeted DC. In collaboration with Crucell (dr. M. Havenga), we x t found Ad35 to efficiently transduce maturing DC in the dermal tissue environment through binding to CD46. In a collaborative study with UAB (dr. D.T. Curiel), R. van de Ven and J.J. Lindenberg, MSc, are similarly investigating DC targeting potential of Ad5/3 vectors that bind CD80 and CD86. In a related NKB-funded project, dr. B. Hangalapura and P.G.J.T.B. Wijnands, MSc, are investigating the utility of CD40-targeted Ad vectors for in vivo DC-based tumor vaccination. The interaction of CD40, a cell surface marker expressed by DC, and its ligand is critical for the generation of protective cell-mediated tumor immunity. We used CFm40L, a bispecific adaptor molecule with the ectodomain of CAR on one end linked to the extracellular domain of mouse CD40 ligand on the other end by a trimerization motif (provided by dr. A. Pereboev, UAB). This adaptor molecule bridges the fiber knob domain of Ad to CD40 on both mouse (m) and human (hu) DC. Re-targeting of Ad to CD40 using CFm40L enhances adenoviral infection and also induces phenotypic and functional maturation of DC. Melanoma antigen recognized by T cell-1 (MART1) and tyrosinase related protein2 (TRP2) are recognized by melanoma specific CTLs. Using CFm40L-targeted Ad encoding huMART1 to infect human DC in vitro, superior priming ability of fully functional CTL was V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s established. We are currently using CFm40Ltargeted Ad encoding mTRP2 to target DC in vivo and examine the effects in terms of T cell activation and tumor protection in the B16 melanoma model. D evelopment of the human MUTZ-3 dendritic cell line model for semi - allogeneic tumor vaccination In a separate line of research the human acute myeloid leukemia (AML) cell line MUTZ-3 and newly generated AML lines are developed to obtain DC in vitro. These DC will be used as allogeneic, standardized off-the-shelf alternatives to autologous DC for DC-based tumor vaccination. Together with the VUmc spin-off DCPrime (prof. dr. A.M. Kruisbeek and prof. dr. R.J. Scheper), we are currently planning phase I clinical trials, in which patients with AML, glioblastoma or prostate cancer will be vaccinated with MUTZ-3 derived DC (either unloaded, fused to glioma cell lines or pulsed with prostate tumor RNA), as an HLA-A2/A3-matched allogeneic vaccine. In preclinical proof of principle studies, we have recently shown that DC can be generated from the CD34+ MUTZ-3 cells. CD14+ MUTZ-3 precursors can either differentiate to Langerhans cells (LC; displaying typical Birbeck granules; Figure 3), or to interstitial DC. To assess the utility of these MUTZ-3-derived DC subsets 33 Ne n t e n t s for immunotherapeutic applications, we tested their potential to migrate to paracortical lymph node areas, their ability to prime tumor-specific CTL, and their cytokine release profile. From these studies, we concluded that the MUTZ-3 cell line may provide a continuous supply of DC and LC precursors for tumor immunotherapy approaches. Before the end of 2007, S.J.A.M. Santegoets is expected to finish her PhD thesis based on these studies. Figure 3. Birbeck granules provide evidence that MUTZ-3 cells can differentiate into bona fide Langerhans cells. NKT cell-based therapies Key features of invariant natural killer (iNKT) cells are an invariant T cell receptor gene usage, CD1d restriction, high levels of cytokine production, particularly interleukin-4 (IL-4) and interferon-γ (IFN-γ), and cytotoxic potential. x t Animal studies showed that iNKT cells, depending on their predominant cytokine profile, are important regulatory cells in autoimmunity (Th2, IL-4) and tumor immunity (Th1, IFN-γ). iNKT cells recognize hydrophobic ligands like α-galactosyl-ceramide (α-GalCer), in conjunction with the antigen presenting molecule CD1d. Under supervision of dr. B.M.E. von Blomberg, head of the Laboratory for Medical Immunology, the size of the circulating iNKT cell population in various types of cancer was compared with matched healthy volunteers. This population was found to be decreased in cancer patients. Whereas the number of T cells or NK cells had no prognostic value, patients with a strongly diminished iNKT cell fraction prior to therapy had significantly decreased disease-specific survival. We are currently developing strategies to increase the frequency of iNKT1 cells in cancer patients, which will hopefully enhance their antitumor immunity. One such approach involves treatment with the ligand α-GalCer, systemically or locally by intradermal administration. Adoptive transfer of iNKT cells expanded in vitro is an alternative approach, the validity of which was tested in a murine tumor model. In a NKBfunded study, dr. H.J. Bontkes and J.W. Molling, MSc, developed a culture method for the expansion of spleen-derived murine polyclonal iNKT cell lines. These iNKT cells could elicit anti-tumor responses in vivo; tumor outgrowth V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s was inhibited after intradermal co-injection with B16.F10 melanoma tumor cells and the formation of lung metastases was inhibited after intravenous co-injection with B16.F10. These data show that cultured murine iNKT cells retain their function and that adoptive transfer of iNKT cells may indeed enhance anti-tumor responses. J.W. Molling is currently finishing his PhD thesis based on these studies. Dr. H.J. Bontkes and M. Moreno, MSc, are currently investigating the influence of iNKT1 cells on tumor-reactive NK and CTL activation and expansion to define the mechanism by which metastasis formation was inhibited. F u r t h e r c o l l a b o r at i v e p r o j e c t s •Development of tissue engineered, autologous human skin for studying sensitization and immuno-vaccination approaches: in collaboration with the Department of Dermatology (dr. S. Gibbs) human, fully defined, immuno-competent in vitro skin models (containing Langerhans cells) are developed by K. Ouwehand, MSc, and employed to study immunogenicity of vaccine formulations and allergens. •Optimisation of strategies for DC vaccination in AML: in collaboration with the Department of Hematology (dr. A. van de Loosdrecht and prof. dr. G. Ossenkoppele) novel approaches to DC-based AML vaccination are explored. 34 •CTL activation markers as prognostic factors in melanoma: in a collaboration with the Department of Pathology (dr. J.J. Oudejans) intratumoral or intranodal CTL activation and apoptosis (resistance)-related markers are studied by I. van Houdt, MSc, for their possible significance in melanoma progression. Division Ne n t e n t s of x t her PhD studies in the group of prof. Peters funded by an exchange grant. P h ar m ac o l o g y In the Division of Pharmacology research is mainly focused on the mechanism of action of, and resistance to, cytotoxic and novel targeted anticancer agents in relation to pharmacogenomic, pharmacodynamic and pharmacokinetic parameters. This research was coordinated in the laboratory by prof. dr. G.J. Peters and in the clinic by prof.dr. G. Giaccone and dr. C. van Groeningen. Dr. F.A.E. Kruyt supervised the research on cell death mechanisms, dr. J. Rodriguez on targeted agents in the laboratory and prof.dr. Giaccone in the clinic. Following current clinical practice in medical oncology, research on several targeted agents is becoming more and more integrated in all research lines. In this year, the division hosted a number of foreign scientists working in the various research groups, in particular Elisa Giovannetti, PhD, from Italy, Cecilia Ceresa, MSc, from Italy, Yerigeri Mayur, PhD, from India, and Christina Tekle, MSc, from Norway, while Clara Lemos, MSc, from Portugal performs Members of the group of prof. Peters Clinical p h a r m ac o k i n e t i c s a n d p h a r m ac o d y na m i c s The flavonoid monoHER has been investigated for its protective effects against doxorubicininduced cardiotoxicity. Scheduling of monoHER was investigated in an animal model by A. Bruynzeel, MD, while in a clinical study the potential protective effects of monoHer in humans were investigated by taking biopsies after repeated administration of doxorubicin with monoHER. Differences in metabolism of the flavonoid between humans and rats due to conjugation and sulfatation have been investigated. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Application of liquid chromatography (LC) coupled to mass-spectrometry (LC-MS-MS) continued to increase. A sensitive LC-MS-MS detection method for gemcitabine, developed by dr. R. Honeywell, was used to measure the low plasma concentration of gemcitabine (50150 nM) after administration as a hepatic artery infusion, and (phosphorylated) gemcitabine in brain tumors. These data demonstrated for the first time that gemcitabine passes the bloodbrain/tumor barrier. The same method was applied in the quantification of deoxycytidine (CdR) in various clinical schedules. These studies demonstrated that gemcitabine treatment resulted in an increase of CdR. Other applications of LC-MS-MS include the measurement of folates in patients enrolled in a randomized phase II study, comparing gemcitabine-cisplatin with or without folate supplementation. This study was initiated in 2005 and patients are still being entered. The LC-MS-MS method also enabled measurement of the accumulation of deoxyuridine after administration of the thymidylate synthase (TS) inhibitor pemetrexed (Alimta®). These data are being related to the clinical effects of treatment. A small adaptation of this assay allowed the analysis of DNA methylation patterns. Methylation of deoxycytidine in the promoter region of individual genes will effectively turn off gene function. The degree of methylation was 35 investigated in several cell lines and in paired cells of colon tumors and normal mucosa. T r a n s l at i o n a l Ne n t e n t s r e s e a rc h , p h a r m ac o g e n o m i c s Pharmacogenomic features of various populations are considered increasingly important as a tool for selecting the most optimal treatment for patients, aiming at reducing toxicity and/or increasing antitumor activity of an anticancer agent. The study in which optimal treatment was based on the tumoral expression of thymidylate synthase (TS) and dihydropyrimidine dehydrogenase (DPD), the TS/DPD study, is now being followed by a retrospective study of historic samples using micro-array analysis of the whole genome, and array-CGH analysis of the DNA, in collaboration with dr. B. Ylstra and prof. G.A. Meijer of the Department of Pathology. In a small cohort of 15 patients, non-supervised clustering of the array-CGH data revealed a significant difference between responding and non-responding patients. Further pharmacogenomics research focuses on enzymes that play an important role in folate homeostasis. Since it is known that a 677-TT homozygosity of methylene tetrahydrofolate reductase (MTHFR) is associated with lower enzyme activity, this polymorphism will increase x t the concentration of the substrate of TS and may affect the efficacy of drugs targeting TS, such as 5-fluorouracil and pemetrexed. Therefore, polymorphism of MTHFR together with that of other folate-related enzymes has been determined in normal and tumor tissues of patients being treated with antifolates. In colon cancer patients, an association between MTHFR polymorphism and response was observed. A ntimetabolites , DNA repair , cell cycle antagonists and multidrug resistance proteins Antimetabolites interfere with cellular functioning at many levels. Not only do they compete with normal metabolites for incorporation into RNA or DNA, affecting cellular proliferation, they also directly affect other processes such as cell cycle regulation and angiogenesis. Thus, the antimetabolites, methotrexate and 5-fluorouracil (5-FU) appear to have additional mechanisms of action, which involve interference with signalling. Vice versa, alterations in cellular signalling processes can profoundly affect the antitumor activity of these agents. For instance, the activity and expression of thymidine phosphorylase (TP), also known as platelet-derived endothelial growth factor (PD-ECGF), is controlled by cytokines such as IL-8. TP plays a major role in cellular thymidine homeostasis; one of the products of this V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s reaction, deoxyribose-1-phosphate (dRib-1-P), is thought to play a major role in angiogenesis. TP also catalyses the last and rate-limiting step of the activation of capecitabine (Xeloda®) to 5-FU, namely that of 5’deoxy-5-fluorouridine (5’DFUR) to 5-FU. Inhibition of TP may prevent tumorspecific angiogenesis. Irene Bijnsdorp, MSc, therefore investigated the downstream effects of TP inhibition both in tumor cells and human umbilical vein edothelial cells (HUVECs). The first data indicate differential effects in these cell types, opening up possibilities for selective treatment with combinations of TP inhibitor (TPI) such as the formulation TAS-102, which consists of trifluorothymidine (TFT) and TPI. Overexpression of drug transporters may confer resistance to a number of unrelated drugs. Previously it was demonstrated that folates are substrates of MRPs, but also that the expression of MRPs is affected by folate supplementation or depletion. Clara Lemos, MSc, demonstrated that folate homeostasis modulates breast cancer resistance protein (BCRP or ABCG1) and multidrug resistance protein 1 (MRP1) expression differently in cell lines from various origins. The change in BCRP expression was accompanied by loss of resistance to mitoxantrone in MCF7/MR LF cells and induction of mitoxantrone resistance in Caco-2 LF cells. Interestingly, it was found that BCRP localisation was either 36 cytoplasmic (Caco-2 cells) or in the plasma membrane (MCF-7/MR cells). Only in the latter cells, folate deprivation induced loss of BCRP expression. A visiting scientist from India, Y. Mayur, PhD, investigated whether N10substituted fluoro-acridone derivatives might be a substrate for one of the efflux pumps Pglycoprotein, MRP1-5 or BCRP. I nteraction Ne n t e n t s of cytotoxic drugs with targeted agents In order to increase selectivity of cancer treatment, convential cytotoxic treatment is increasingly being combined with newer drugs interfering with cellular signal transduction pathways. Cellular proliferation and functioning is controlled by growth factors which activate membrane proteins by phosphorylation, followed by a cascade of phosphorylation reactions inside the cell. Erlotinib (Tarceva®), a potent small molecule inhibitor of the tyrosine kinase domain of epidermal growth factor receptor (EGFR) was combined with both the platinum analog satraplatin (C. Ceresa , MSc) and the antifolate pemetrexed (E. Giovannetti, MD). Pemetrexed was also combined with the protein kinase C-β (PKC-β) inhibitor enzastaurin (C. Tekle, PhD). The active metabolite of satraplatin, JM118, and erlotinib appeared to be synergistic in non- small cell lung cancer (NSCLC), colon x t and ovarian cancer cell lines by interfering with p-ERK and p-AKT signalling downstream of EGFR, while p38 signalling was not modulated. The interaction between erlotinib and pemetrexed was investigated in six NSCLC cell lines, characterized by a large variation in thymidylate synthase (TS) expression, the target of pemetrexed, and by the presence of mutations of EGFR, K-ras, and PTEN, all determinants of erlotinib sensitivity. Synergistic cytotoxicity was detected in all cell lines, mostly with the pemetrexed (24h) → erlotinib (72h) sequence, associated with a significant induction of apoptosis. Pemetrexed increased EGFR phosphorylation and reduced AKT phosphorylation, while erlotinib significantly reduced TS activity, which was additionally reduced by the combination. In two of the cell lines, the interaction of pemetrexed with enzastaurin was investigated, showing enhanced pemetrexed- induced cell death by enzaustarin. Enzastaurin reduced the phosphorylation of targets in cell signalling pathways such as AKT and GSK3b as well as TS expression, thus potentially enhancing pemetrexed activity. T argeting apoptosis and kinase pathways for cancer therapy Research by the group of dr. F.A.E. Kruyt and prof. G. Giaccone ranges from fundamental to clinical research on the mechanisms that V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s determine life-and-death decisions in tumor cells. The decisive step in the action of standard anticancer drugs and novel targeted agents is their ability to induce molecular pathways in tumor cells that lead to cell death by activation of apoptosis. For this reason, the gain in fundamental knowledge of molecular mechanisms underlying cell death regulation is key for the rational design of novel treatment strategies and the development of targeted medicines. In addition, molecular pathways that regulate apoptosis are often defective in tumor cells and are likely to contribute to drug resistance and poor patient survival. Hence, apoptotic regulators may have diagnostic and prognostic value. Parallel to the preclinical work, clinical studies with novel targeted agents are conducted. Members of the group of prof. Giaccone and dr. Kruyt 37 C onventional Ne n t e n t s chemotherapy and apoptosis We have discovered that in chemotherapyresistant non-small cell lung cancer (NSCLC) cells, the apoptotic pathway headed by caspase-9 is blocked. Caspases are a group of proteases that execute the apoptotic program. A. Checinska, MSc, supported by a grant of the Dutch Cancer Society (NKB), is exploring the mechanism underlying caspase-9 inhibition. She will defend her thesis in March 2007. In this context, the involvement of members of the inhibitor-ofapoptosis protein family (IAPs) that are able to bind to, and inhibit caspases, is evaluated. In her studies, a potential small molecule drug that is able to mimic the activity of Smac (a kind gift from dr. X. Wang, Dallas) is used. Smac is a cellular protein that can inactivate IAPs. Also mass spectrometry-based proteomic strategies are employed to identify caspase-9 binding proteins in NSCLC cells, studies that are conducted in collaboration with dr. C. Jiménez, VU University, Amsterdam). L. Bröker, MSc, has finished her studies on the mechanisms underlying apoptosis activation by microtubule-interacting agents. She will defend her thesis in 2007. One of the main findings in her studies was that these potent anticancer drugs induce apoptosis via caspase-independent pathways. In particular, the lysosomal protease, cathepsin B, was found to mediate cell killing by x t the microtubule-stabilizing agents, paclitaxel and epothilone B. T argeted medicines Based on our mechanistic studies, we hypothesize that agents that can restore caspase9 activation or that bypass the blockade will have therapeutic benefit in NSCLC. Results by J. Voortman, MD, MSc, supported by a NWOAGIKO grant and A. Checinska showed that the proteasome inhibitor, bortezomib, (PS-341, Velcade®) is able to restore caspase-9 activation in NSCLC cells. Apart from the preclinical work, J. Voortman also coordinated a clinical study in which the efficacy of this agent, in combination with other anticancer agents, in the treatment of solid tumors is examined. As a means to bypassing caspase-9, current research is focusing on tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL) that activates caspase8-dependent apoptosis. Two PhD students supported by a Dutch Cancer Society grant (in collaboration with dr. S. de Jong and prof. dr. E.G.E. de Vries, University Medical Centre Groningen) are currently working on the efficacy and mechanism of action of combinations of TRAIL and bortezomib in NSCLC models (A. Watts, MSc, in Amsterdam and J. Stegehuis in Groningen). V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Dr. F.A.E. Kruyt obtained a grant from the Top Institute Pharma (TIPharma), a national governmental initiative to stimulate the interaction between industry and academic centres. TIPharma aims at achieving leadership in research and education in areas critical for the international competitiveness of the pharmaceutical industry in the Netherlands. Together with the academic groups of dr. S. de Jong and prof. dr. E.G.E. de Vries, and prof. dr. W. Quax (University Medical Centre Groningen), prof. dr. J. Borst (Netherlands Cancer Institute, Amsterdam), prof. dr. J.P. Medema (Academic Medical Center, Amsterdam), a collaboration has been established with Organon (Oss) and Pepscan (Lelystad) to study TNF ligands in cancer. At the VU University Medical Center, we will in particular study newly developed agonistic TRAIL receptor-directed monoclonal antibodies for anticancer activity, and determine the involvement of kinase pathways in modulating the response. Signaling induced by the epidermal growth factor receptor (EGFR) is known to affect survival of NSCLC cells. Agents targeting EGFR have provided novel promising therapeutic strategies. M. Janmaat, MSc, studied the mechanism and efficacy of EGFR inhibitors, including the monoclonal antibody C225 (cetuximab) and the small molecule EGFR-tyrosine kinase inhibitors 38 Ne n t e n t s x t gefitinib (Iressa®) and erlotinib (Tarceva®). He found that their efficacy was related to the activation status of the MEK/Erk (MAPK) and PI3K/Akt kinase pathways. In October 2006, M. Janmaat successfully defended his thesis entitled “Targeting Erb receptors as anticancer therapy: factors of sensitivity and resistance”. The recent discovery that mutations in the ATP binding site of EGFR confer sensitivity to small-molecule EGFR inhibitors in advanced NSCLC patients has led us to the sequencing of tumor samples of patients enrolled in phase II studies of EGFR inhibitors in esophageal cancer and NSCLC. A grant from the Dutch Cancer Society (NKB) has been obtained for this work. M. Gallegos-Ruiz, MSc, is spearheading this work. In addition to setting up the techniques to detect mutations in EGFR and K-Ras, we are performing an extensive analysis of a large series of NSCLC samples. Using tissue microarrays, the expression of several proteins in the EGFR pathway and related signaling networks are being examined. Furthermore, expression microarrays and array comparative genomic hybridization (aCGH), in collaboration with dr. B. Ylstra, are being carried out to gain a better understanding of the genetic changes that occur during NSCLC development. Dr. Maarten Janmaat successfully defended his thesis; Laudatio by prof. Giaccone R egulation of inhibitors of (IAP s ) Research by dr. J. Rodriguez, supported by the Walter Bruckerhoff Stiftung, focuses on the functional implications of regulated nuclearcytoplasm shuttling of proteins that regulate key cellular processes, such as apoptosis and cell cycle progression. Current research focuses on survivin, a member of the IAP family with a dual role in apoptosis and mitosis. Survivin shuttles between the nucleus and the cytoplasm. Several splice variants of survivin, which undergo differential transport and localize to different cellular compartments have been identified. The role of each variant in tumor development and apoptosis proteins V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s progression is being investigated. Using RNA interference and mutagenesis, we are exploring several aspects of survivin biology and its relation with other proteins, such as the kinase aurora B, both during mitosis and interphase. On the other hand, using novel imaging techniques, such as bimolecular fluorescence complementation (BiFC), the dimerization ability of survivin in vivo is being explored as a potential target for anticancer therapies. In this regard, we have recently identified a functional role for survivin dimerization: regulation of its nuclear export. These studies are carried out in collaboration with dr. R. Medema and dr. S. Lens (University Medical Center Utrecht) and with the group of dr. M. Fornerod (Netherlands Cancer Institute, Amsterdam). B. Vischioni, MSc, obtained her PhD degree on November 14, 2006 after defending her thesis entitled “Expression and subcellular localization of inhibitor of apoptosis (IAP) proteins in human tissues: mechanisms and clinical implications”. She studied the regulation of the nucleo-cytoplasmic localization of the survivin relatives cIAP1, cIAP2 and XIAP. She has found that cIAP1 and cIAP2, but not XIAP are nucleocytoplasmic shuttling proteins. She mapped several CRM1-dependent nuclear export signals (NES) in these IAPs. Extending these mechanistic studies, B. Vischioni has also 39 Ne n t e n t s investigated the distribution of IAPs in normal tissues and tumor samples. She has also explored the expression of the kinase aurora B, a partner of the IAP survivin in NSCLC tumor samples and the effect of novel aurora B inhibitors on the growth of NSCLC cells in vitro. These studies are essential to predict side effects in patients undergoing treatment with IAP inhibitors, drugs that are presently entering clinical trials. P r o g n o s t i c m a r k e r s i n NSCLC The prognostic value of expression of the apoptotic regulator TUCAN has been studied by A. Checinska. Although expression did not correlate with treatment outcome in NSCLC patients, cytoplasmic localization of TUCAN predicted shorter survival. Dr. F. Barlesi, a guest from the Assistance Publique Hopiteaux de Marseille, France, studied global histone modifications as prognostic factor for resected NSCLC patients. His data indicate that methylation and acetylation of histone H3 are valuable prognostic markers for a subset of early-stage NSCLC patients. Another visitor in our department, dr. P Zucali from Italy, studied the relationship between activation of the c-Met receptor pathway and response to EGFR inhibitors in NSCLC. Using tissue microarrays and in vitro experiments, he found that activation of c-Met may be marker of intrinsic resistance to gefitinib and that x t combined inhibition of the c-Met and EGFR pathway may be a suitable therapeutic approach for some NSCLC patients. These studies were performed in collaboration with prof. G. Meijer, prof. P. van der Valk and dr. J. Oudejans from the Department of Pathology. O n c o P r o t e o m i cs L a b o rat o r y CCA O ONCO VUmc PL Proteomics Laboratory The OncoProteomics Laboratory (OPL) has been founded in April 2006 together with the establishment of the cancer research building of the VUmc Cancer Center Amsterdam (CCA). OPL was created to provide a state-of-the-art proteomics infrastructure and knowledge center for CCA/V-ICI researchers. Proteomics is a relatively new field. It creates a link between genomic information and biological function through global studies of protein expression, protein modification and proteinprotein interactions. Until now, proteomics has been a technology-driven science. Emphasis in the coming years will be on applying proteomics to the understanding of biological function in V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s healthy organisms and in disease (Figure 4). to high-throughput MALDI-TOF/TOF mass spectrometry, and 2) label-free (nano)-LC-LTQFT mass spectrometry of sub-fractionated biofluids and tumor samples for in-depth profiling. Code Readout DN A ~22.000 R NA ~100.000 R e s e a rc h x t s t r at e g y A. D evelopment of robust automated mass spectrometry - based methods for biomarker discovery , with a focus on subproteomes P R OT E IN > 500.000 W ork ing m a c hine ry Figure 4. Approximation of the number of genes, messenger RNA molecules and proteins in humans, illustrating the complexity of the proteome M i s s i o n o f OPL The mission of OPL is to develop and implement innovative proteomics technologies and data analysis methods to improve diagnosis and treatment of cancer. To this end, the major focus is on developing and implementing robust strategies for biomarker discovery in tumor tissue, if available, and in biofluids such as blood-serum/plasma that can be collected noninvasively as well as on proximal fluid such as CSF and nipple aspirate fluid. In addition, cancer cell conditioned media and tumor secretomes hold great promise for discovery of candidate biomarkers. Samples are profiled using two complementary platforms for the discovery of diagnostic, predictive and drug response patterns and biomarkers: 1) an automated magnetic bead peptide capture method coupled 40 Ne n t e n t s O r g a n i s at i o n o f OPL OPL is a facilitating center where most projects are shaped in close interaction with the collaborators. Moreover, a substantial part of the activities is core research to develop and implement proteomics methods of general interest for cancer researchers and clinicians. Head of the unit is a scientist/coordinator who is leading a team of scientists (a mass spectrometrist, a computer scientist and a biochemical technician as well as some technicians on collaborative temporary projects). OPL is housed by the Department of Medical Oncology. The head of OPL is assisted by a ‘Program Advisory Committee’ with representatives from the major V-ICI departments (Medical Oncology, Hematology, Pathology, ENT, Molecular, Cellular Biology and Immunology, and Epidemiology and Biostatistics) who meet every two months with the head of OPL to discuss and assist in prioritizing projects. The lack of reliable, robust and easily assessable biomarkers greatly hampers cancer management. In recent years, proteomics has raised the hope of identifying novel biomarkers for cancer diagnosis and detection. This hope is based on the ability of proteomic technologies, such as mass spectrometry, to identify hundreds of proteins in complex biofluids. OPL focuses on two complementary approaches for the discovery of diagnostic and predictive peptide patterns and identification of candidate biomarkers in body fluids and tumors (Figure 5). The first goal was to implement robust, scalable methods for patient sample preparation that can be performed in parallel and in an automated manner. Pre-analytical variables of serum sample handling and analytical aspects of the high through-put (HTP) profiling workflow have been investigated (Jiménez et al., work submitted and in preparation). Therefore, the automated method can now be applied to cancer profiling studies. In several phase I studies of the Department of Medical Oncology, serum for proteomics analysis has been collected. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close OPL focuses on two complementary approaches for the discovery of diagnostic and predictive peptide patterns and identification of candidate biomarkers in body fluids and tumors (Figure 5). The first goal was to implement robust, scalable methods for patient sample preparation that can be performed in P parallel r e v and i ou s automated manner. Contents in an HIGH-THROUGHPUT POTENTIAL DIAGNOSTIC TOOL IN-DEPTH HIGH RESOLUTION & DYNAMIC RANGE, IDENTIFIED BIOMARKERS Patients M AS S SP E CT RO M ET R Y Peptide mass profile Multi-dimensional Proteomic profile HTP In-depth Pattern recognition M AL D I-T OF- M S 100 Diseas e % intensity 165 100 5 182 6 199 7 216 8 165 5 182 6 199 7 216 Disease protein Signature Retention time (min) 233 9 control Mass (m/z) 8 233 9 Nano- LC -L TQF TM S Novel biomarkers DIAGNOSIS PROGNOSIS DRUG RESPONSE Diseas e Contro l Mass (m/z) Figure 5. Complementary mass spectrometry-based proteomics approaches for discovery of cancer signatures and Figure 5. Complementary mass spectrometry-based approaches formethod discovery of cancer signatures biomarker at OPL. LEFT: High-throughput MALDI-TOFproteomics mass spectrometry-based coupled to spectral data and biomarker at OPL. LEFT: High-throughput MALDI-TOF mass spectrometry-based method coupled to mining of blood-serum peptides and proteins for the discovery of predictive, diagnostic and drug-response signatures. spectral data mining of blood-serum peptides and proteins for the discovery of predictive, diagnostic and drugTissue response homogenates can also Tissue be subjected to this type analysis. RIGHT: sub-fractionation method coupled signatures. homogenates canofalso be subjected toProteome this type of analysis. RIGHT: Proteome subto (nano)Liquid-Chromatography-LTQ-FT mass spectrometry for in-depth profiling of peptides and proteins and discovery/identification of predictive and early detection biomarkers in patient samples. Subproteomes of interest are 37 the nucleus and secreted proteins. In blood, the low various subcellular tissue fractions including the plasma membrane, molecular weight proteome as well as the glycoproteome are of particular interest. 41 Ne x t Robust methods for the capture of subproteomes from tissue and blood for in-depth profiling by nanoLCFTMS and discovery of identified candidate biomarkers are currently being developed and explored in several (collaborative) projects (among others with dr. F. Kruyt , dr. H.J. Broxterman, dr. K. Hoekman and with dr. R.Fijneman, prof. dr. G. Meijer and dr. B. Carvalho of the Department of Pathology). B. D ata analysis We are developing robust data analysis methods for label-free quantitation of peptide ion abundance, because this approach represents a promising (and economic) avenue for quantitation at high sensitivity. The additional dimension of separation allows for profiling of a much larger number of peptides in complex mixtures. We are implementing open-source tools as well as home-made algorithms (Horizon Breakthrough project granted to Drs. Marchiori and Jiménez) for the analysis of LC-MS datasets. The sensitivity of LC-MS profiling is higher, but the throughput of this method is considerably less than the MALDI-TOF-MS approach (~ 10 samples per day), so we will use high quality, well-characterized samples for this approach. Statistically significant differential peptides will be followed up in targeted tandem MS experiments to determine the sequence and thus peptide/protein identities of candidate markers. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s n t e n t s Ne x t Because this method yields identified proteins, it can be more easily coupled to future antibodybased screening. C. R esearch proteomics Proteomics applied to model systems (in cell culture or animal studies) is well established and can solve a range of questions related to protein expression levels, composition of protein complexes, proteolytic processing, post-translational modifications as well as (subcellular) localization and secretion into the medium. Therefore these types of projects have been started in 2006 immediately after the required instruments were up and running. 42 V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Ne n t e n t s x t PhD theses 2006 PhD Würdinger T. – The development of tumorselective Coronaviruses as anti-cancer agents. Universiteit Utrecht. Promotor: prof.dr. P.J.M. Rottier. Co-promotores: dr. W.R. Gerritsen, dr. V.W. van Beusechem, dr. M.H.Verheije. PrintPartners Ipskamp B.V., Enschede, 2006: 186 pp. Date: 01-02-2006. Vuylsteke R.J.C.L.M. – Clinical and immunological aspects of the sentinel lymph node procedure in melanoma. Vrije Universiteit Amsterdam. Promotores: prof.dr. P.A.M. van Leeuwen, prof.dr. S.Meijer, prof.dr.R.J. Scheper. Co-promotor: dr. T.D. de Gruijl. Febodruk BV, Enschede, 2006; ISBN 90 8659 0144 4: 159 pp. Date: 29-09-2006. theses 2006 Vischioni B. – Expression and subcellular localization of inhibitor of apoptosis (IAP) proteins in human tissues: mechanisms and clinical implications. Vrije Universiteit Amsterdam. Promotor: prof. dr. G. Giaccone. Co-promotor: dr. JA. Rodriguez. PrintPartners Ipskamp B.V., Enschede, 2006: 192 pp. Date: 14-11-2006. Witlox M.A. – Gene therapy of osteosarcoma. Preclinical studies with adeoviral vectors and adenoviral oncolysis. Vrije Universiteit Amsterdam. Promotor: prof. dr. P.I.J.M. Wuisman. Co-promotores: dr. V.W. van Beusechem, dr. W.R. Gerritsen. Haveka B.V., 2006. 161 pp. Date: 15-12-2006. Dr. Barbara Vischioni with her paranymphs; Laudatio by prof. Giaccone Janmaat M.L. – Targeting ErbB receptors as anticancer therapy: Factors of sensitivity and resistance. Vrije Universiteit Amsterdam. Promotor: prof. dr. G. Giaccone. Co-promotor: dr. JA. Rodriguez. PrintPartners Ipskamp B.V., Enschede, 2006: 155 pp. Date: 11-10-2006. 43 V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Ne n t e n t s x t Scientific publicatons Scientific Abou El Hassan MA, Braam SR, Kruyt FA. A real-time RT-PCR assay for the quantitative determination of adenoviral gene expression in tumor cells. J Virol Methods 2006; 133: 53-61. Abou El Hassan MA, Braam SR, Kruyt FA. Paclitaxel and vincristine potentiate adenoviral oncolysis that is associated with cell cycle and apoptosis modulation, whereas they differentially affect the viral life cycle in non-small-cell lung cancer cells. Cancer Gene Ther 2006; 13:1105-1114. Achille M, Gallegos-Ruiz M, Giaccone G, Soria JC. Response to erlotinib in first-line treatment of non-small-cell lung cancer in a white male smoker with squamous-cell histology. Clin Lung Cancer 2006; 8: 214-216. Ackland SP, Clarke SJ, Beale P, Peters GJ. Thymidylate synthase inhibitors. Update Cancer Ther. 2006; 1: 403-427. Adema AD, Hubeek I, Zuurbier L, Floor K, Albertioni F, Kaspers GJL, Peters GJ. Cellular resistance against troxacitabine in human cell lines and pediatric patient acute myeloid leukemia blast cells. Nucleosides Nucleotides Nucleic Acids 2006; 25: 981-986. 44 p u b l i ca t o n s Bergman AM, Peters GJ. Gemcitabine. mechanism of action and resistance. From: Cancer drug discovery and development: deoxynucleoside analogs in cancer therapy. Edited by: G.J. Peters. Humana Press Inc., Totowa, NJ, USA. 2006; pp. 225-251. Biesma B, Manegold C, Smit HJ, Willems L, Legrand C, Passioukov A, van Meerbeeck JP, Giaccone G; for the EORTC Lung Cancer Group. Docetaxel and cisplatin as induction chemotherapy in patients with pathologicallyproven stage IIIA N2 non-small cell lung cancer: a phase II study of the European organization for research and treatment of cancer (EORTC 08984). Eur J Cancer 2006; 42: 1399-1406. Bijman MNA, Van Nieuw Amerongen GP, Laurens N, Van Hinsbergh VWM, Boven E. Microtubule-targeting agents inhibit angiogenesis at subtoxic concentrations, a process associated with inhibition of Rac1 and Cdc42 activity and changes in the endothelial cytoskeleton. Clin Cancer Res 2006; 5:2348-2357. Boddaert MS, Gerritsen WR, Pinedo HM. On our way to targeted therapy for cachexia in cancer? Curr Opin Oncol 2006; 18: 335-340. Bontkes HJ, Ruizendaal JJ, Kramer D, Santegoets SJ, Scheper RJ, de Gruijl TD, Meijer CJ, Hooijberg E. Constitutively active STAT5b induces cytokine-independent growth of the acute myeloid leukemia-derived MUTZ-3 cell line and accelerates its differentiation into mature dendritic cells. J Immunother 2006; 29: 188-200. Bottomley A, Gaafar R, Manegold C, Burgers S, Coens C, Legrand C, Vincent M, Giaccone G, van Meerbeeck J; EORTC Lung-Cancer Group; Natl Cancer Inst, Canada. Short-term treatment-related symptoms and quality of life: results from an international randomized phase III study of cisplatin with or without raltitrexed in patients with malignant pleural mesothelioma: an EORTC Lung-Cancer Group and National Cancer Institute, Canada, Intergroup Study. J Clin Oncol 2006; 24: 1435-1442 (Erratum in: J Clin Oncol 2006; 24: 2601). V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Bröker LE, de Vos FY, van Groeningen CJ, Kuenen BC, Gall HE, Woo MH, Voi M, Gietema JA, de Vries EG, Giaccone G. Phase I trial with BMS-275183, a novel oral taxane with promising antitumor activity. Clin Cancer Res 2006; 12: 1760-1767. Broxterman HJ. Circulerende endotheelcellen bij de kankerpatiënt. Angiogenese Journaal 2006; 1, no. 2: 1-5. Checinska A, Giaccone G, Hoogeland B, Ferreira CG, Rodriguez JA, Kruyt FA. TUCAN /CARDINAL/CARD8 and apoptosis resistance in non-small cell lung cancer cells. BMC Cancer 2006; 6: 166. Checinska A, Oudejans JJ, Span SW, Rodriguez JA, Kruyt FA, Giaccone G. The expression of TUCAN, an inhibitor of apoptosis protein, in patients with advanced non-small cell lung cancer treated with chemotherapy. Anticancer Res 2006; 26: 3819-3824. De Bree R, van der Valk P, Kuik DJ, van Diest PJ, Doornaert P, Buter J, Eerenstein SE, Langendijk JA, van der Waal I, Leemans CR. Prognostic factors in adult soft tissue sarcomas of the head and neck: a single-centre experience. Oral Oncol 2006; 42: 703-709. 45 Ne n t e n t s De Bruin M, Temmink OH, Hoekman K, Pinedo HM, Peters GJ. Role of platelet derived endothelial cell growth factor / thymidine phosphorylase in health and disease. Cancer Ther 2006; 4: 99-124. De Gruijl TD, Ophorst OJ, Goudsmit J, Verhaagh S, Lougheed SM, Radosevic K, Havenga MJ, Scheper RJ. Intradermal delivery of adenoviral type-35 vectors leads to high efficiency transduction of mature, CD8+ T cell-stimulating skin-emigrated dendritic cells. J Immunol 2006; 177: 2208-2215. De Gruijl TD, Sombroek CC, Lougheed SM, Oosterhoff D, Buter J, van den Eertwegh AJ, Scheper RJ, Pinedo HM. A postmigrational switch among skin-derived dendritic cells to a macrophage-like phenotype is predetermined by the intracutaneous cytokine balance. J Immunol 2006; 176: 7232-7242. Efficace F, Bottomley A, Smit EF, Lianes P, Legrand C, Debruyne C, Schramel F, Smit HJ, Gaafar R, Biesma B, Manegold C, Coens C, Giaccone G, van Meerbeeck J; the EORTC Lung Cancer Group and Quality of Lif Unit. Is a patient’s self-reported health-related quality of life a prognostic factor for survival in non-smallcell lung cancer patients? A multivariate analysis of prognostic factors of EORTC study 08975. Ann Oncol 2006; 17: 1698-1704. x t Fijneman, RJ. Paving the road towards noninvasive molecular imaging. Cell Oncol 2006; 28: 125-126. Fontijn D, Duyndam MC, van Berkel MP, Yuana Y, Shapiro LH, Pinedo HM, Broxterman HJ, Boven E. CD13/aminopeptidase N overexpression by basic fibroblast growth factor enhanced invasiveness of 1F6 human melanoma cells. Br J Cancer 2006; 94: 1627-1636. Giaccone G, Gallegos-Ruiz M. Le Chevalier T, Thatcher N, Smit EF, Rodriguez JA, Janne P, Oulid-Aissa D, Soria J-C. Erlotinib for frontline treatment of advanced non-small cell lung cancer: a phase II study. Clin Cancer Res 2006; 12: 6049-6055. Giaccone G, Wilmink H, Paul MA, van der Valk P. Systemic treatment of malignant thymoma: a decade experience at a single institution. Am J Clin Oncol 2006; 29: 336-344. Graat HCA, Witlox MA, Schagen FHE, Kaspers GJL, Helder MN, Bras J, Schaap GR, Gerritsen WR, Wuisman PIJM, Van Beusechem VW. Different susceptibility of osteosarcoma cell lines and primary cells to treatment with oncolytic adenovirus and doxorubicin or cisplatin. Br J Cancer 2006; 94: 1837-1844. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Haanen JB, Baars A, Gomez R, Weder P, Smits M, de Gruijl TD, von Blomberg BM, Bloemena E, Scheper RJ, van Ham SM, Pinedo HM, van den Eertwegh AJ. Melanoma-specific tumorinfiltrating lymphocytes but not circulating melanoma-specific T cells may predict survival in resected advanced-stage melanoma patients. Cancer Immunol Immunother 2006; 55: 451-458. Haveman J, Sigmond J, Van Bree C, Franken NA, Koedooder C, Peters GJ. Time course of enhanced activity of deoxycytidine kinase and thymidine kinase 1 and 2 in cultured human squamous lung carcinoma cells, SW-1573, induced by gamma-irradiation. Oncol Rep 2006; 16: 901905. Honeywell R, van Groeningen CJ, Laan AC, Strocchi E, Ruiter R, Giaccone G, Peters GJ. Analysis of deoxycytidine accumulation in gemcitabine treated patients. Nucleosides Nucleotides Nucleic Acids 2006; 25: 1225-1232. Hooijberg JH, De Vries NA, Kaspers GJL, Pieters R, Jansen G, Peters GJ. Multidrug resistance proteins and folate supplementation: therapeutic implications for antifolates and other classes of drugs in cancer treatment. Cancer Chemother Pharmacol 2006; 58: 1-12. 46 Ne n t e n t s Hornberg JJ, Bruggeman FJ, Westerhoff HV, Lankelma J. Cancer: a Systems Biology disease. Biosystems 2006; 83: 81-90. Hornberg JJ, Dekker H, Peters PH, Langerak P, Lankelma J, van Zoelen EJ. Epidermal growth factor receptor-induced activator protein 1 activity controls density-dependent growth inhibition in normal rat kidney fibroblasts. Mol Biotechnol 2006; 34: 101-108. Hubeek I, Kaspers GJL, Ossenkoppele GJ, Peters GJ. Cytosine arabinoside. Metabolism, mechanisms of resistance and clinical pharmacology. From: Cancer drug discovery and development: deoxy-nucleoside analogs in cancer therapy. Edited by: G.J. Peters. Humana Press Inc., Totowa, NJ, USA. 2006; pp. 119-151. Hubeek I, Peters GJ, Broekhuizen AJF, Zwaan ChM,Kaaijk P, Van Wering ER, Gibson BES, Creutzig U, Janka-Schaub GE, Den Boer ML, Pieters R, Kaspers GJL. In vitro sensitivity and cross-resistance to deoxynucleoside analogs in childhood acute leukemia. Haematologica 2006; 91: 17-23. x t Janmaat ML, Gallegos-Ruiz M, Rodriguez JA, Meijer GA, Vervenne WL, Richel DJ, Van Groeningen C, Giaccone G. Predictive factors for outcome in a phase II study of gefitinib in second-line treatment of advanced esophageal cancer patients. J Clin Oncol 2006; 24: 1612-1619. Janmaat ML, Rodriguez JA, Gallegos-Ruiz M, Kruyt FAE, Giaccone G. Enhanced cytotoxicity induced by gefitinib and specific inhibitors of the Ras or phosphatidyl inositol-3-kinase pathways in non-small cell lung cancer cells. Int J Cancer 2006; 118: 209-214. Janmaat ML, Rodriguez JA, Giaccone G. Correspondence in reply to: Personeni N., Epidermal growth factor receptor gene copy number in esophageal cancer and outcome prediction to gefitinib: does intratumoral heterogeneity matter? J Clin Oncol 2006; 24: 5466-5467. Jellema AP, Slotman BJ, Muller MJ, Leemans CR, Smeele LE, Hoekman K, Aaronson NK, Langendijk JA. Radiotherapy alone, versus radiotherapy with amifostine 3 times weekly, versus radiotherapy with amifostine 5 times weekly: A prospective randomized study in squamous cell head and neck cancer. Cancer 2006; 107: 544-553. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Jiménez CR. Proteomics in de Oncologie. In: Kanker, tijdschrift van de Nederlandse Vereniging voor Oncologie; april 2006. Kramer GW, Legrand CL, van Schil P, Uitterhove L, Smit EF, Schramel F, Biesma B, Tjan-Heijnen V, van Zandwijk N, Splinter T, Giaccone G, van Meerbeeck JP; on behalf of EORTC-Lung Cancer Group. Quality assurance of thoracic radiotherapy in EORTC 08941: a randomised trial of surgery versus thoracic radiotherapy in patients with stage IIIA non-small-cell lung cancer (NSCLC) after response to induction chemotherapy. Eur J Cancer 2006; 42: 1391-1398. Kroep JR, Peters, GJ, Nagourney RA. Clinical activity of gemcitabine as a single agent and in combination. From: Cancer drug discovery and development: deoxynucleoside analogs in cancer therapy. Edited by: G.J. Peters. Humana Press Inc., Totowa, NJ, USA. 2006; pp. 253-287. Kroep JR, Smit EF, Giaccone G, van den Born K, Beijnen JH, van Groeningen CJ, van der Vijgh WJ, Postmus PE, Pinedo HM, Peters GJ. Pharmacology of the paclitaxel-cisplatin, gemcitabine-cisplatin, and paclitaxel-gemcitabine combinations in patients with advanced nonsmall cell lung cancer. Cancer Chemother Pharmacol 2006; 58: 509-516. 47 Ne n t e n t s Lamfers MLM, Fulci G, Gianni D, Tang Y, Kurozumi K, Kaur B, Moeniralm S, Saeki Y, Carette JE, Weissleder R, Vandertop WP, Van Beusechem VW, Dirven CMF, Chiocca EA. Cyclophosphamide increases transgene expression mediated by an oncolytic adenovirus in gliomabearing mice monitored by bioluminescence imaging. Mol Ther 2006; 14: 779-788. Lammertsma AA, Hoekstra CJ, Giaccone G, Hoekstra OS. How should we analyse FDG PET studies for monitoring tumour response? Eur J Nucl Med Mol Imaging 2006; 33 (Suppl 13): 16-21. Lens SMA, Rodriguez JA, Vader G, Span SW, Giaccone G, Medema RH. Uncoupling the central spindle-associated function of the Chromosome Passenger Complex from its role at centromeres. Mol Biol Cell 2006; 17: 1897-1909. Mauritz R, Peters GJ. Pharmacogenetics of colon cancer and potential implications for 5-fluorouracil-based chemotherapy. Current Pharmacogenomics 2006; 4: 57-67. Molenkamp BG, van Leeuwen PA, van den Eertwegh AJ, Sluijter BJ, Scheper RJ, Meijer S, de Gruijl TD. Immunomodulation of the melanoma sentinel lymph node: a novel adjuvant therapeutic option. Immunobiology 2006; 211: 651-661. x t Padrón JM, Peters GJ. Cytotoxicity of sphingoid marine compound analogs in mono- and multilayered solid tumor cell cultures. Invest New Drugs 2006; 24: 195-202. Pauwels B, Korst AEC, Pattyn GGO, Lambrechts HAJ, Kamphuis JAE, De Pooter CMJ, Peters GJ, Lardon F, Vermorken JB. The relation between deoxycytidine kinase activity and the radiosensitizing effect of gemcitabine in eight different human tumor cell lines. BMC Cancer 2006; 6: 142 (doi: 10.1186/ 1471-2407-6-142). Peltenburg H, Schopenhauer R, van der Linden MHM. The Family Support project: a unique concept of family care within the hospital setting. Psychooncology 2006; 15: S1-S478. Peters GJ. Preface. From: Cancer drug discovery and development: deoxynucleoside analogs in cancer therapy. Edited by: G.J. Peters. Humana Press Inc., Totowa, NJ, USA. 2006; pp. V-VII. Peters GJ, Carrey EA, Sebésta I. Purine and pyrimidine metabolism, a firm base for a transformed society. Nucleosides Nucleotides Nucleic Acids 2006; 25: 971-974. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Peters GJ, van Moorsel CJ, Lakerveld B, Smid K, Noordhuis P, Comijn EC, Weaver D, Willey JC, Voorn D, van der Vijgh WJ, Pinedo HM. Effects of gemcitabine on cis-platinum-DNA adduct formation and repair in a panel of gemcitabine and cisplatin-sensitive or -resistant human ovarian cancer cell lines. Int J Oncol 2006; 28: 237-244. Santegoets SJ, Schreurs MW, Masterson AJ, Liu YP, Goletz S, Baumeister H, Kueter EW, Lougheed SM, van den Eertwegh AJ, Scheper RJ, Hooijberg E, de Gruijl TD. In vitro priming of tumor-specific cytotoxic T lymphocytes using allogeneic dendritic cells derived from the human MUTZ-3 cell line. Cancer Immunol Immunother 2006; 55: 1480-1490. Rodriguez JA, Giaccone G. Keynote comment: Are large-scale cancer-genomics projects ready to use? Lancet Oncol 2006; 7: 190-191. Savonije JH, van Groeningen CJ, Wormhoudt LW, Giaccone G. Early Intervention with epoetin alfa during platinum-based chemotherapy: an analysis of quality-of-life results of a multicenter, randomized, controlled trial compared with population normative data. Oncologist 2006; 11: 197-205. Rodriguez JA, Lens SMA, Span SW, Vader G, Medema RH, Kruyt FAE, Giaccone G. Subcellular localization and nucleocytoplasmic transport of the chromosomal passenger proteins before nuclear envelope breakdown. Oncogene 2006; 25: 4867-4879. Santegoets SJ, Masterson AJ, van der Sluis PC, Lougheed SM, Fluitsma DM, van den Eertwegh AJ, Pinedo HM, Scheper RJ, de Gruijl TD. A CD34(+) human cell line model of myeloid dendritic cell differentiation: evidence for a CD14(+)CD11b(+) Langerhans cell precursor. J Leukoc Biol 2006; 80: 1337-1344. 48 Ne n t e n t s Savonije JH, van Groeningen CJ, Wormhoudt LW, Giaccone G. Early intervention with epoetin alfa during platinum-based chemotherapy: an analysis of the results of a multicenter, randomized, controlled trial based on initial hemoglobin level. Oncologist 2006; 11: 206-216. Savonije JH, van Groeningen CJ, van den Broek WJ, Rentinck ME, Corstens FH, Gundy C, Giaccone G, Marx JJ. Iron absorption during epoetin alfa therapy for chemotherapy-associated anaemia. Cancer Invest 2006; 24: 562-566. x t Schagen FHE, Wensveen FM, Carette JE, Dermody TS, Gerritsen WR, Van Beusechem VW. Genetic targeting of adenovirus vectors using a reovirus σ1-based attachment protein. Mol Ther 2006; 13: 997-1005. Sigmond J, Haveman J, Castro Kreder N, Loves WJ, Van Bree C, Franken NA, Peters GJ. Enhanced activity of deoxycytidine kinase after pulsed low dose rate and single dose gammairradiation. Nucleosides Nucleotides Nucleic Acids 2006; 25: 1177-1180. Smid K, Bergman AM, Eijk PP, Veerman G, Ruiz van Haperen VWT, Van den IJssel P, Ylstra B, Peters GJ. Micro-array analysis of resistance for gemcitabine results in increased expression of ribonucleotide reductase subunits. Nucleosides Nucleotides Nucleic Acids, 2006; 25: 1001-1007. Smorenburg CH, Peters GJ, van Groeningen CJ, Noordhuis P, Smid K, van Riel AM, Dercksen W, Pinedo HM, Giaccone G. Phase II study of tailored chemotherapy for advanced colorectal cancer with either 5-fluouracil and leucovorin or oxaliplatin and irinotecan based on the expression of thymidylate synthase and dihydropyrimidine dehydrogenase. Ann Oncol 2006; 17: 35-42. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s Temmink OH, De Bruin M, Laan AC, Turksma AW, Cricca S, Masterson AJ, Noordhuis P, Peters GJ. The role of thymidine phosphorylase and uridine phosphorylase in (fluoro) pyrimidine metabolism in peripheral blood mononuclear cells. Int J Biochem Cell Biol 2006; 38: 1759-1765. Temmink OH, Hoogeland MFM, Fukushima M, Peters GJ.Low folate conditions may enhance the interaction of trifluorothymidine with antifolates in colon cancer cells. Cancer Chemother Pharmacol 2006; 57: 171-179. Tijink BM, Buter J, de Bree R, Giaccone G, Lang MS, Staab A, Leemans CR, van Dongen GA. A phase I dose escalation study with anti-CD44v6 bivatuzumab mertansine in patients with incurable squamous cell carcinoma of the head and neck or esophagus. Clin Cancer Res 2006; 12(20 Pt 1): 6064-6072. Van der Linden MHM, Ruijter S, Verdonck-de Leeuw IM, Kleverlaan N. Children of parents with cancer, an exploratory study of the Dutch website ‘Cancer Phantoms’. Psychooncology 2006; 15: S46-47. Van der Vliet HJ, van Vonderen MG, Molling JW, Bontkes HJ, Reijm M, Reiss P, van Agtmael MA, Danner SA, van den Eertwegh AJ, von Blomberg BM, Scheper RJ. Cutting edge: Rapid recovery 49 Ne n t e n t s of NKT cells upon institution of highly active antiretroviral therapy for HIV-1 infection. J Immunol 2006; 177: 5775-5778. Van de Ven R, de Jong MC, Reurs AW, Schoonderwoerd AJ, Jansen G, Hooijberg JH, Scheffer GL, de Gruijl TD, Scheper RJ. Dendritic cells require multidrug resistance protein 1 (ABCC1) transporter activity for differentiation. J Immunol 2006; 176: 5191-5198. Van Heijst JW, Niessen HW, Musters RJ, van Hinsbergh VW, Hoekman K, Schalkwijk CG. Argpyrimidine-modified Heat shock protein 27 in human non-small cell lung cancer: a possible mechanism for evasion of apoptosis. Cancer Lett 2006; 241: 309-319. Van Hinsbergh VWM, Engelse MA, Quax PHA. Pericellular proteases in angiogenesis and vasculogenesis. Arterioscler Thromb Vasc Biol 2006; 26: 716-728. Verdonck-de Leeuw IM, Borggreven PA, Eerenstein SEJ, van der Linden MHM, Rinkel RNPM, de Bree R, Leemans CR. Psychosociale gevolgen en functionele gevolgen van hoofdhalskanker. Nederlands Tijdschrift voor Oncologie 2006; 3: 185-191. x t Verdonck-de Leeuw IM, Eerenstein SEJ, van der Linden MHM, Kuik DJ, de Bree R, Leemans CR. Distress in spouses and patients with head and neck cancer. Psychooncology 2006; 15: S1-S478. Vergeer MR, Doornaert P, Leemans CR, Buter J, Slotman BJ, Langendijk JA. Control of nodal metastases in squamous cell head and neck cancer treated by radiation therapy or chemoradiation. Radiother Oncol 2006; 79: 3944. Verheije MH, Würdinger T, Van Beusechem VW, De Haan CAM, Gerritsen WR, Rottier PJM. Redirecting coronavirus to a non-native receptor through a virus-encoded targeting adapter. J Virol 2006; 80: 1250-1260. Vischioni B, Oudejans JJ, Vos W, Rodriguez JA, Giaccone G. Frequent overexpression of aurora B kinase, a novel drug target, in non-small cell lung carcinoma patients. Mol Cancer Ther 2006; 5: 2905-2913. Vischioni B, van der Valk P, Span SW, Kruyt FAE, Rodriguez JA, Giaccone G. Expression and localization of inhibitor of apoptosis proteins in normal human tissues. Human Pathol 2006; 37: 78-86. V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close Pr Co e v i ou s n t e n t s Voortman J, Giaccone G. Severe reversible cardiac failure after bortezomib treatment combined with chemotherapy in a non-small cell lung cancer patient: a case report. BMC Cancer 2006; 6: 129. Vuylsteke RJ, Molenkamp BG, van Leeuwen PA, Meijer S, Wijnands PG, Haanen JB, Scheper RJ, de Gruijl TD. Tumor-specific CD8+ T cell reactivity in the sentinel lymph node of GM-CSF-treated stage I melanoma patients is associated with high myeloid dendritic cell content. Clin Cancer Res 2006; 12: 2826-2833. Würdinger T, Verheije MH., Van der Aa LM, Bosch BJ, De Haan CAM, Van Beusechem VW, Gerritsen WR, Rottier PJM. Antibodymediated targeting of coronaviral vectors to the Fc receptor on acute myeloid leukemia cells. Leukemia 2006; 20: 2182-2184. Zucali PA, Giaccone G. Biology and management of malignant pleural mesothelioma. Eur J Cancer 2006; 42: 2706-2714. 50 V U m c M e d i c a l O n c o l o g y – A n n u a l R e p or t 2 0 0 6 Close