Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Sigrid Stroobants is active.

Publication


Featured researches published by Sigrid Stroobants.


Journal of Clinical Oncology | 2007

Revised Response Criteria for Malignant Lymphoma

Bruce D. Cheson; Beate Pfistner; Malik E. Juweid; Randy D. Gascoyne; Lena Specht; Sandra J. Horning; Bertrand Coiffier; Richard I. Fisher; Anton Hagenbeek; Emanuele Zucca; Steven T. Rosen; Sigrid Stroobants; T. Andrew Lister; Richard T. Hoppe; Martin Dreyling; Kensei Tobinai; Julie M. Vose; Joseph M. Connors; Massimo Federico; Volker Diehl

PURPOSE Standardized response criteria are needed to interpret and compare clinical trials and for approval of new therapeutic agents by regulatory agencies. METHODS The International Working Group response criteria (Cheson et al, J Clin Oncol 17:1244, 1999) were widely adopted, but required reassessment because of identified limitations and the increased use of [18F]fluorodeoxyglucose-positron emission tomography (PET), immunohistochemistry (IHC), and flow cytometry. The International Harmonization Project was convened to provide updated recommendations. RESULTS New guidelines are presented incorporating PET, IHC, and flow cytometry for definitions of response in non-Hodgkins and Hodgkins lymphoma. Standardized definitions of end points are provided. CONCLUSION We hope that these guidelines will be adopted widely by study groups, pharmaceutical and biotechnology companies, and regulatory agencies to facilitate the development of new and more effective therapies to improve the outcome of patients with lymphoma.


The Lancet | 2001

Safety and efficacy of imatinib (STI571) in metastatic gastrointestinal stromal tumours: a phase I study

Allan Van Oosterom; Ian Judson; Jaap Verweij; Sigrid Stroobants; Eugenio Donato di Paola; Sasa Dimitrijevic; Marc Martens; Andrew Webb; Raf Sciot; Martine Van Glabbeke; Sandra Leta Silberman; Ole Steen Nielsen

BACKGROUND Gastrointestinal stromal tumours (GISTs) are rare tumours of the gastrointestinal tract characterised by cell-surface expression of the tyrosine kinase KIT (CD117). No effective systemic treatment is available. Imatinib (STI571) inhibits a similar tyrosine kinase, BCR-ABL, leading to responses in chronic myeloid leukaemia, and has also been shown to inhibit KIT. We did a phase I study to identify the dose-limiting toxic effects of imatinib in patients with advanced soft tissue sarcomas including GISTs. METHODS 40 patients (of whom 36 had GISTs) received imatinib at doses of 400 mg once daily, 300 mg twice daily, 400 mg twice daily, or 500 mg twice daily. Toxic effects and haematological, biochemical, and radiological measurements were assessed during 8 weeks of follow-up. 18Fluorodeoxy-glucose positron-emission tomography (PET) was used for response assessment in one centre. FINDINGS Five patients on 500 mg imatinib twice daily had dose-limiting toxic effects (severe nausea, vomiting, oedema, or rash). Inhibition of tumour growth was seen in all but four patients with GISTs, resulting in 19 confirmed partial responses and six as yet unconfirmed partial responses or more than 20% regressions. 24 of 27 clinically symptomatic patients showed improvement, and 29 of 36 were still on treatment after more than 9 months. PET scan responses predicted subsequent computed tomography responses. INTERPRETATION Imatinib at a dose of 400 mg twice daily is well tolerated during the first 8 weeks, side-effects diminish with continuing treatment, and it has significant activity in patients with advanced GISTs. Our results provide evidence of a role for KIT in GISTs, and show the potential for the development of anticancer drugs based on specific molecular abnormalities present in cancers.


Journal of Clinical Oncology | 2007

Use of Positron Emission Tomography for Response Assessment of Lymphoma: Consensus of the Imaging Subcommittee of International Harmonization Project in Lymphoma

Malik E. Juweid; Sigrid Stroobants; Otto S. Hoekstra; Felix M. Mottaghy; Markus Dietlein; Ali Guermazi; Gregory A. Wiseman; Lale Kostakoglu; Klemens Scheidhauer; Andreas K. Buck; Ralph Naumann; Karoline Spaepen; Rodney J. Hicks; Wolfgang A. Weber; Sven N. Reske; Markus Schwaiger; Lawrence H. Schwartz; Josée M. Zijlstra; Barry A. Siegel; Bruce D. Cheson

PURPOSE To develop guidelines for performing and interpreting positron emission tomography (PET) imaging for treatment assessment in patients with lymphoma both in clinical practice and in clinical trials. METHODS An International Harmonization Project (IHP) was convened to discuss standardization of clinical trial parameters in lymphoma. An imaging subcommittee developed consensus recommendations based on published PET literature and the collective expertise of its members in the use of PET in lymphoma. Only recommendations subsequently endorsed by all IHP subcommittees were adopted. RECOMMENDATIONS PET after completion of therapy should be performed at least 3 weeks, and preferably at 6 to 8 weeks, after chemotherapy or chemoimmunotherapy, and 8 to 12 weeks after radiation or chemoradiotherapy. Visual assessment alone is adequate for interpreting PET findings as positive or negative when assessing response after completion of therapy. Mediastinal blood pool activity is recommended as the reference background activity to define PET positivity for a residual mass > or = 2 cm in greatest transverse diameter, regardless of its location. A smaller residual mass or a normal sized lymph node (ie, < or = 1 x 1 cm in diameter) should be considered positive if its activity is above that of the surrounding background. Specific criteria for defining PET positivity in the liver, spleen, lung, and bone marrow are also proposed. Use of attenuation-corrected PET is strongly encouraged. Use of PET for treatment monitoring during a course of therapy should only be done in a clinical trial or as part of a prospective registry.


European Journal of Nuclear Medicine and Molecular Imaging | 2010

FDG PET and PET/CT: EANM procedure guidelines for tumour PET imaging: version 1.0

Ronald Boellaard; Michael O'Doherty; Wolfgang A. Weber; Felix M. Mottaghy; Markus N. Lonsdale; Sigrid Stroobants; Wim J.G. Oyen; Joerg Kotzerke; Otto S. Hoekstra; Jan Pruim; Paul Marsden; Klaus Tatsch; Corneline J. Hoekstra; Eric P. Visser; Bertjan Arends; Fred J. Verzijlbergen; Josée M. Zijlstra; Emile F.I. Comans; Adriaan A. Lammertsma; Anne M. J. Paans; Antoon T. M. Willemsen; Thomas Beyer; Andreas Bockisch; Cornelia Schaefer-Prokop; Dominique Delbeke; Richard P. Baum; Arturo Chiti; Bernd J. Krause

The aim of this guideline is to provide a minimum standard for the acquisition and interpretation of PET and PET/CT scans with [18F]-fluorodeoxyglucose (FDG). This guideline will therefore address general information about [18F]-fluorodeoxyglucose (FDG) positron emission tomography-computed tomography (PET/CT) and is provided to help the physician and physicist to assist to carrying out, interpret, and document quantitative FDG PET/CT examinations, but will concentrate on the optimisation of diagnostic quality and quantitative information.


European Journal of Cancer | 2003

18FDG-Positron emission tomography for the early prediction of response in advanced soft tissue sarcoma treated with imatinib mesylate (Glivec).

Sigrid Stroobants; J. Goeminne; M. Seegers; Sasa Dimitrijevic; P. Dupont; J. Nuyts; M. Martens; B. van den Borne; P. Cole; Raphael Sciot; Herlinde Dumez; Sandra Leta Silberman; Luc Mortelmans; A.T. van Oosterom

Imatinib mesylate (Glivec, formerly STI571) is the first effective systemic treatment for gastrointestinal stromal tumours (GISTs). Major changes in tumour volume, however, tend to occur late after the start of treatment. The aim of this study was to evaluate if [18F]-fluorodeoxyglucose-positron emission tomography (FDG-PET) can be used for the early evaluation of response to imatinib mesylate treatment in soft-tissue sarcomas (STS). 21 patients (17 GIST, 4 other STS) underwent FDG-PET imaging prior to and 8 days after the start of treatment. PET response (European Organization for Research and Treatment (EORTC) guidelines) was observed in 13 GISTs (11 Complete Responders, 2 partial responders. Subsequent computerised tomography (CT) response Response Evaluation Criteria in Solid Tumours (RECIST) was observed in 10 of these patients after a median follow up of 8 weeks. Stable or progressive disease was observed on PET in 8 patients and none of them achieved a response on CT. PET response was also associated with a longer progression-free survival (PFS) (92% versus 12% at 1 year, P=0.00107). We conclude that FDG-PET is an early and sensitive method to evaluate an early response to imatinib treatment.


Journal of Clinical Oncology | 2001

Prognostic Value of Positron Emission Tomography (PET) With Fluorine-18 Fluorodeoxyglucose ([18F]FDG) After First-Line Chemotherapy in Non-Hodgkin’s Lymphoma: Is [18F]FDG-PET a Valid Alternative to Conventional Diagnostic Methods?

Karoline Spaepen; Sigrid Stroobants; Patrick Dupont; Steven Van Steenweghen; José Thomas; Peter Vandenberghe; Lucien Vanuytsel; Guy Bormans; Jan Balzarini; Christine De Wolf-Peeters; Luc Mortelmans; Gregor Verhoef

PURPOSE A complete remission (CR) after first-line therapy is associated with longer progression-free survival (PFS). However, defining CR is not always easy because of the presence of residual masses. Metabolic imaging with fluorine-18 fluorodeoxyglucose ([18F]FDG) positron emission tomography (PET) offers the ability to differentiate between viable and fibrotic inactive tissue. In this study, we evaluated the value of PET in detecting residual disease and, hence, predicting relapse after first-line treatment in patients with non-Hodgkins lymphoma (NHL). PATIENTS AND METHODS Ninety-three patients with histologically proven NHL, who underwent a whole-body [18F]FDG-PET study after completion of first-line chemotherapy and who had follow-up of at least 1 year, were included. Persistence or absence of residual disease on PET was related to PFS using Kaplan-Meier survival analysis. RESULTS Sixty-seven patients showed a normal PET scan after first-line chemotherapy; 56 of 67 remained in CR, with a median follow-up of 653 days. Nine of these patients with a residual mass considered as unconfirmed CR received additional radiotherapy. Only 11 of 67 patients relapsed (median PFS, 404 days). Persistent abnormal [18F]FDG uptake was seen in 26 patients, and all of them relapsed (median PFS, 73 days). Because standard restaging also suggested residual disease, 12 patients received immediate secondary treatment. In 14 of 26 patients, only PET predicted persistent disease. From these patients, relapse was proven either by biopsy (n = 8) or by progressive disease on computed tomography or magnetic resonance imaging (n = 6). CONCLUSION Persistent abnormal [18F]FDG uptake after first-line chemotherapy in NHL is highly predictive for residual or recurrent disease. In relapsing patients, PFS was significantly shorter after a positive scan than after a negative scan.


European Journal of Nuclear Medicine and Molecular Imaging | 2015

FDG PET/CT: EANM procedure guidelines for tumour imaging: version 2.0

Ronald Boellaard; Roberto Delgado-Bolton; Wim J.G. Oyen; Francesco Giammarile; Klaus Tatsch; Wolfgang Eschner; Fred J. Verzijlbergen; Sally Barrington; Lucy Pike; Wolfgang A. Weber; Sigrid Stroobants; Dominique Delbeke; Kevin J. Donohoe; Scott Holbrook; Michael M. Graham; Giorgio Testanera; Otto S. Hoekstra; Josée M. Zijlstra; Eric P. Visser; Corneline J. Hoekstra; Jan Pruim; Antoon T. M. Willemsen; Bertjan Arends; Joerg Kotzerke; Andreas Bockisch; Thomas Beyer; Arturo Chiti; Bernd J. Krause

The purpose of these guidelines is to assist physicians in recommending, performing, interpreting and reporting the results of FDG PET/CT for oncological imaging of adult patients. PET is a quantitative imaging technique and therefore requires a common quality control (QC)/quality assurance (QA) procedure to maintain the accuracy and precision of quantitation. Repeatability and reproducibility are two essential requirements for any quantitative measurement and/or imaging biomarker. Repeatability relates to the uncertainty in obtaining the same result in the same patient when he or she is examined more than once on the same system. However, imaging biomarkers should also have adequate reproducibility, i.e. the ability to yield the same result in the same patient when that patient is examined on different systems and at different imaging sites. Adequate repeatability and reproducibility are essential for the clinical management of patients and the use of FDG PET/CT within multicentre trials. A common standardised imaging procedure will help promote the appropriate use of FDG PET/CT imaging and increase the value of publications and, therefore, their contribution to evidence-based medicine. Moreover, consistency in numerical values between platforms and institutes that acquire the data will potentially enhance the role of semiquantitative and quantitative image interpretation. Precision and accuracy are additionally important as FDG PET/CT is used to evaluate tumour response as well as for diagnosis, prognosis and staging. Therefore both the previous and these new guidelines specifically aim to achieve standardised uptake value harmonisation in multicentre settings.


Journal of Clinical Oncology | 1999

Prognostic Importance of the Standardized Uptake Value on 18F-Fluoro-2-Deoxy-Glucose–Positron Emission Tomography Scan in Non–Small-Cell Lung Cancer: An Analysis of 125 Cases

Johan Vansteenkiste; Sigrid Stroobants; Patrick Dupont; Paul De Leyn; Erik Verbeken; Georges Deneffe; Luc Mortelmans; Maurits Demedts

PURPOSE The amount of radio-labeled (18)F-fluoro-2-deoxy-glucose (FDG) uptake, a measurement of the increased glucose metabolism of non-small-cell lung cancer (NSCLC) cells, has recently been correlated with proliferation capacity. The Standardized Uptake Value (SUV), a semi-quantitative measurement of FDG uptake on positron emission tomography (PET) scan, could thus be of prognostic significance. PATIENTS AND METHODS We analyzed the follow-up of 125 potentially operable NSCLC patients, previously included in three of our prospective PET protocols. Performance status, maximal tumor diameter, tumor-cell type, SUV, and final staging were analyzed for their possible association with survival. RESULTS Sixty-five patients had stage I or II NSCLC, 37 had stage IIIA, and 23 had stage IIIB. Treatment was complete resection in 91 cases. In a univariate analysis, performance status (P =.002), stage (P =.001), tumor diameter (P =.06), tumor-cell type (P =.03), and SUV greater than 7 (P =.001) were correlated with survival. For SUV, group dichotomy with a cut-off SUV of 7 had the best discriminative value for prognosis, both in the total and surgical cohort. A multivariate Cox analysis identified performance status (P =.02), stage (P =.01), and SUV (P =.007) as important for the prognosis. In the surgical group, patients with a resected tumor less than 3 cm had an expected 2-year survival of 86%, if the SUV was below 7, and 60%, if above 7. Nearly all resected tumors larger than 3 cm had SUVs greater than 7 and an expected 2-year survival of 43%. CONCLUSION We conclude that the FDG uptake in primary NSCLC on PET has an important prognostic value and could be complementary to other well-known factors in the decision on adjuvant treatment protocols.


Radiotherapy and Oncology | 2000

The impact of 18F-fluoro-2-deoxy-d-glucose positron emission tomography (FDG-PET) lymph node staging on the radiation treatment volumes in patients with non-small cell lung cancer

Luc Vanuytsel; Johan Vansteenkiste; Sigrid Stroobants; Paul De Leyn; Walter De Wever; Eric Verbeken; Giovanna Gatti; Dominique Huyskens; Gerald Kutcher

Abstract Purpose : 18 F-fluoro-2-deoxy-d-glucose positron emission tomography (FDG-PET) combined with computer tomography (PET-CT) is superior to CT alone in mediastinal lymph node (LN) staging in non-small cell lung cancer (NSCLC). We studied the potential impact of this non-invasive LN staging procedure on the radiation treatment plan of patients with NSCLC. Patients and methods : The imaging and surgical pathology data from 105 patients included in two previously published prospective LN staging protocols form the basis for the present analysis. For 73 of these patients, with positive LNs on CT and/or on PET, a theoretical study was performed in which for each patient the gross tumour volume (GTV) was defined based on CT and on PET-CT data. For each GTV, the completeness of tumour coverage was assessed, using the available surgical pathology data as gold standard. A more detailed analysis was done for the first ten consecutive patients in whom the PET-CT-GTV was smaller than the CT-GTV. Theoretical radiation treatment plans were constructed based on both CT-GTV and PET-CT-GTV. Dose-volume histograms for the planning target volume (PTV), for the total lung volume and the lung volume receiving more than 20 Gy ( V lung(20) ), were calculated. Results : Data from 988 assessed LN stations were available. In the subgroup of 73 patients with CT or PET positive LNs, tumour coverage improved from 75% when the CT-GTV was used to 89% with the PET-CT-GTV ( P =0.005). In 45 patients (62%) the information obtained from PET would have led to a change of the treatment volumes. For the ten patients in the dosimetry study, the use of PET-CT to define the GTV, resulted in an average reduction of the PTV by 29±18% (±1 SD) ( P =0.002) and of the V lung(20) of 27±18% (±1 SD) ( P =0.001). Conclusion : In patients with NSCLC considered for curative radiation treatment, assessment of locoregional LN tumour extension by PET will improve tumour coverage, and in selected patients, will reduce the volume of normal tissues irradiated, and thus toxicity. This subgroup of patients could then become candidates for treatment intensification.


IEEE Transactions on Medical Imaging | 1999

Simultaneous maximum a posteriori reconstruction of attenuation and activity distributions from emission sinograms

Johan Nuyts; Patrick Dupont; Sigrid Stroobants; Roel Benninck; Luc Mortelmans; Paul Suetens

In order to perform attenuation correction in emission tomography an attenuation map is required. The authors propose a new method to compute this map directly from the emission sinogram, eliminating the transmission scan from the acquisition protocol. The problem is formulated as an optimization task where the objective function is a combination of the likelihood and an a priori probability. The latter uses a Gibbs prior distribution to encourage local smoothness and a multimodal distribution for the attenuation coefficients. Since the attenuation process is different in positron emission tomography (PET) and single photon emission tomography (SPECT), a separate algorithm for each case is derived. The method has been tested on mathematical phantoms and on a few clinical studies. For PET, good agreement was found between the images obtained with transmission measurements and those produced by the new algorithm in an abdominal study. For SPECT, promising simulation results have been obtained for nonhomogeneous attenuation due to the presence of the lungs.

Collaboration


Dive into the Sigrid Stroobants's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luc Mortelmans

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patrick Dupont

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Johan Vansteenkiste

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Johan Nuyts

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar

Otto S. Hoekstra

VU University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge