Network


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

Hotspot


Dive into the research topics where Rafke Schoffelen is active.

Publication


Featured researches published by Rafke Schoffelen.


Molecular Cancer Therapeutics | 2010

Pretargeted Immuno–Positron Emission Tomography Imaging of Carcinoembryonic Antigen–Expressing Tumors with a Bispecific Antibody and a 68Ga- and 18F-Labeled Hapten Peptide in Mice with Human Tumor Xenografts

Rafke Schoffelen; Robert M. Sharkey; David M. Goldenberg; Gerben M. Franssen; William J. McBride; Edmund A. Rossi; Chien-Hsing Chang; Peter Laverman; Jonathan A. Disselhorst; Annemarie Eek; Winette T. A. van der Graaf; Wim J.G. Oyen; Otto C. Boerman

18F-Fluorodeoxyglucose (18F-FDG) is the most common molecular imaging agent in oncology, with a high sensitivity and specificity for detecting several cancers. Antibodies could enhance specificity; therefore, procedures were developed for radiolabeling a small (∼1451 Da) hapten peptide with 68Ga or 18F to compare their specificity with 18F-FDG for detecting tumors using a pretargeting procedure. Mice were implanted with carcinoembryonic antigen (CEA; CEACAM5)–expressing LS174T human colonic tumors and a CEA-negative tumor, or an inflammation was induced in thigh muscle. A bispecific monoclonal anti-CEA × anti-hapten antibody was given to mice, and 16 hours later, 5 MBq of 68Ga- or 18F-labeled hapten peptides were administered intravenously. Within 1 hour, tissues showed high and specific targeting of 68Ga-IMP-288, with 10.7 ± 3.6% ID/g uptake in the tumor and very low uptake in normal tissues (e.g., tumor-to-blood ratio of 69.9 ± 32.3), in a CEA-negative tumor (0.35 ± 0.35% ID/g), and inflamed muscle (0.72 ± 0.20% ID/g). 18F-FDG localized efficiently in the tumor (7.42 ± 0.20% ID/g) but also in the inflamed muscle (4.07 ± 1.13% ID/g) and in several normal tissues; thus, pretargeted 68Ga-IMP-288 provided better specificity and sensitivity. Positron emission tomography (PET)/computed tomography images reinforced the improved specificity of the pretargeting method. 18F-labeled IMP-449 distributed similarly in the tumor and normal tissues as the 68Ga-labeled IMP-288, indicating that either radiolabeled hapten peptide could be used. Thus, pretargeted immuno-PET does exceptionally well with short-lived radionuclides and is a highly sensitive procedure that is more specific than 18F-FDG-PET. Mol Cancer Ther; 9(4); 1019–27. ©2010 AACR.


British Journal of Cancer | 2013

Development of an imaging-guided CEA-pretargeted radionuclide treatment of advanced colorectal cancer: first clinical results

Rafke Schoffelen; Otto C. Boerman; David M. Goldenberg; Robert M. Sharkey; C.M.L. van Herpen; Gerben M. Franssen; William J. McBride; Chien-Hsing Chang; Edmund A. Rossi; W.T.A. van der Graaf; Wim J.G. Oyen

Background:Radiolabelled antibody targeting of cancer is limited by slow blood clearance. Pretargeting with a non-radiolabelled bispecific monoclonal antibody (bsMAb) followed by a rapidly clearing radiolabelled hapten peptide improves tumour localisation. The primary goals of this first pretargeting study in patients with the anti-CEACAM5 × anti-hapten (HSG) bsMAb, TF2, and the radiolabelled hapten-peptide, IMP288, were to assess optimal pretargeting conditions and safety in patients with metastatic colorectal cancer (CRC).Methods:Different dose schedules were studied in four cohorts of five patients: (1) shortening the interval between the bsMAb and peptide administration (5 days vs 1 day), (2) escalating the TF2 dose (from 75 to 150 mg), and (3) reducing the peptide dose (from 100 to 25 μg). After confirmation of tumour targeting by 111In-IMP288, patients were treated with a bsMAb/177Lu-IMP288 cycle.Results:Rapid and selective tumour targeting of the radiolabelled peptide was visualised within 1 h, with high tumour-to-tissue ratios (>20 at 24 h). Improved tumour targeting was achieved with a 1-day interval between the administration of the bsMAb and the peptide and with the 25-μg peptide dose. High 177Lu-IMP288 doses (2.5–7.4 GBq) were well tolerated, with some manageable TF2 infusion reactions, and transient grades 3–4 thrombocytopaenia in 10% of the patients who received 177Lu-IMP288.Conclusion:This phase I study demonstrates for the first time that pretargeting with bsMAb TF2 and radiolabelled IMP288 in patients with CEA-expressing CRC is feasible and safe. With this pretargeting method, tumours are specifically and rapidly targeted.


EJNMMI research | 2012

Pretargeted immuno-PET of CEA-expressing intraperitoneal human colonic tumor xenografts: a new sensitive detection method

Rafke Schoffelen; Winette T. A. van der Graaf; Robert M. Sharkey; Gerben M. Franssen; William J. McBride; Chien-Hsing Chang; Peter Laverman; David M. Goldenberg; Wim J.G. Oyen; Otto C. Boerman

BackgroundIn this study, pretargeted immuno-positron-emission tomography [PET] with a bispecific monoclonal anti-carcinoembryonic antigen [CEA] (CEACAM5) × anti-hapten antibody (bispecific monoclonal antibody [bsmAb]) and a small (1.5 kD) peptide labeled with 68Ga was compared to fludeoxyglucose [18F-FDG]-PET for detecting intraperitoneal [i.p.] CEA-expressing human colonic tumor xenografts in nude mice.MethodsTwo groups of female BALB/c nude mice were inoculated with LS174T human colonic tumor cells i.p. One group received 5 MBq 18F-FDG, and the other received intravenous injections of the bsmAb, followed 16 h later with 5 MBq of 68Ga-labeled peptide. One hour after the radiolabeled peptide or FDG was given, micro-PET/computed tomography images were acquired. Thereafter, the uptake of the 68Ga or 18F in dissected tissue was determined.ResultsWithin 1 h, high uptake of the 68Ga-labeled peptide in the tumor lesions (23.4 ± 7.2% ID/g) and low background activity levels were observed (e.g., tumor-to-intestine ratio, 58 ± 22). This resulted in a clear visualization of all intra-abdominal tumor lesions ≥ 10 μL and even some tumors as small as 5 μL (2 mm diameter). 18F-FDG efficiently localized in the tumors (8.7 ± 3.1% ID/g) but also showed physiological uptake in various normal tissues (e.g., tumor-to-intestine ratio, 3.9 ± 1.1).ConclusionsPretargeted immuno-PET with bsmAb and a 68Ga-labeled peptide could be a very sensitive imaging method for imaging colonic cancer, disclosing occult lesions.


The Journal of Nuclear Medicine | 2012

Quantitative Immuno-SPECT Monitoring of Pretargeted Radioimmunotherapy with a Bispecific Antibody in an Intraperitoneal Nude Mouse Model of Human Colon Cancer

Rafke Schoffelen; W.T.A. van der Graaf; Robert M. Sharkey; Gerben M. Franssen; William J. McBride; Chien-Hsing Chang; D. L. Bos; David M. Goldenberg; Wim J.G. Oyen; Otto C. Boerman

The prospects for using pretargeted immuno-SPECT to monitor the response to pretargeted radioimmunotherapy were examined. In this study, a bispecific anticarcinoembryonic antigen (CEACAM5; CD66e) × antihapten monoclonal antibody, TF2, was used in combination with a small (1.5 kD) peptide, IMP288, labeled with 111In and 177Lu. Methods: First, tumor uptake of 111In-IMP288 and 177Lu-IMP288, as determined by immuno-SPECT, was validated by ex vivo counting. Two groups of female BALB/c nude mice had LS174T tumors implanted in the peritoneal cavity. They received intravenous injections of TF2, followed by 10 MBq of 111In-IMP288 or 90 MBq of 177Lu-IMP288. A control group of non–tumor-bearing mice received TF2 and 111In-IMP288. One hour after the radiolabeled IMP288 was given, small-animal SPECT/CT images were acquired, and subsequently animals were dissected. Furthermore, a survival study was performed in 3 groups of 10 mice with intraperitoneal tumors: mice received TF2 and 177Lu-IMP288 (60 MBq), nonpretargeted 177Lu-IMP288 (60 MBq), or phosphate-buffered saline. Immuno-SPECT scans were acquired directly after therapy and at 14 and 45 d after therapy. Tumor growth was analyzed in the successive scans in each animal. Results: 111In- and 177Lu-labeled IMP288 had similar in vivo distribution. The activity measured in the pretargeted immuno-SPECT images correlated well with the uptake measured in the dissected tumors (Pearson r = 0.99, P < 0.05). In the therapy study, the SPECT images showed rapid and selective tumor targeting with high tumor-to-background contrast (30 ± 12) as early as 1 h after injection. The successive images of the treated mice showed delayed tumor growth in the pretargeted radioimmunotherapy group, corresponding with their prolonged survival. Conclusion: Pretargeted immuno-SPECT with TF2 and 111In- or 177Lu-IMP288 can be used to predict and confirm tumor targeting and monitor the therapeutic effect of pretargeted radioimmunotherapy.


Cancer Research | 2012

Abstract 1755: Pretargeted radioimmunotherapy of an anti-CEA bispecific antibody and 177Lu-labeled peptide: a phase I study in patients with advanced colorectal cancer

Rafke Schoffelen; Otto C. Boerman; David M. Goldenberg; Robert M. Sharkey; Carla M.L. Herpen; Chien-Hsing Chang; Gerben M. Franssen; William J. McBride; Edmund A. Rossi; Winette T. A. van der Graaf; Wim J.G. Oyen

Table of Contents * Pretargeted radioimmunotherapy of an anti-CEA bispecific antibody and 177Lu-labeled peptide: a phase I study in patients with advanced colorectal cancer * Abstract Objectives: Pretargeted radioimmunotherapy (PRIT) with bispecific monoclonal antibodies (bsMAb) in combination with a radiolabeled peptide reduces the radiation dose of conventional RIT to normal tissues by rapid tumor targeting and fast blood clearance of the radiolabeled molecule. In this phase I trial, PRIT was investigated in 20 patients with advanced colorectal cancer (CRC), using a humanized recombinant bsMAb, TF2, targeting carcinoembryonic antigen (CEACAM5) and the hapten-peptide, IMP288, which was radiolabeled with177Lu. The safety, pharmacokinetics, and tumor targeting of these agents were studied, by optimizing the dose schedule of the pretargeting system. Methods: We included four cohorts with five patients per cohort. We studied the interval between the bsMAb and peptide administration and its consequences for the blood clearance of the radiolabeled peptide (cohort 1: 5 days interval vs next cohorts: 1 day). We escalated TF2 dose to saturate tumor antigens (cohort 3: 150 mg TF2, vs other cohorts: 75 mg). Furthermore, we investigated the effect of labelling the same radioactivity doses to a lower peptide dose, which ideally would increase the fractional targeting to the tumors (cohort 4: 25 µg IMP288 vs other cohorts: 100 µg) A pre-therapy cycle with 111In-labeled IMP288 was used to predict the pharmacokinetics and tumor targeting of 177Lu-IMP288, and to assure safety of high 177Lu activity doses (3.7 GBq in cohort 1 and ≤ 7.4 GBq in cohorts 2-4). Toxicity was determined by NCI-CTC v3.0. Whole-body planar and SPECT images were acquired. Results: Toxicity was generally very mild, with only two patients showing grade 3-4 thrombocytopenia, and one with grade 3 dyspnea. Dose escalation did not increase toxicity. Rapid localization of the radiolabeled peptide in the tumor lesions was clearly visualized with scintigraphy within 1 h of injection, with excellent tumor-to-normal tissue ratios (>20 at 24 h after injection in all cohorts). Low absorbed radiation doses to the red bone marrow ( Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1755. doi:1538-7445.AM2012-1755


Cancer Research | 2011

Abstract 5282: Pretargeted immunoPET for imaging human colonic cancer in a mouse model

Rafke Schoffelen; Gerben M. Franssen; William J. McBride; Jonathan A. Disselhorst; Robert M. Sharkey; Peter Laverman; Winette T. A. van der Graaf; Wim J.G. Oyen; David M. Goldenberg; Otto C. Boerman

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL Objective: Molecular imaging with pretargeting bispecific antibodies allows rapid and specific targeting of tumors for PET imaging. In this study, pretargeted immunoPET with a bispecific monoclonal anti-carcinoembryonic antigen (CEA; CEACAM5) × anti-hapten antibody (bsMAb) and a small (1.5 kD) peptide labeled with a short-lived positron-emitter 68Ga was compared to FDG for detection of intraperitoneal CEA-expressing human colonic tumor xenografts in nude mice. Methods: In BALB/c nude mice LS174T human colonic tumor cells were injected intraperitoneally. One group (n=5) received 5 MBq FDG i.v., and another group (n=5) received the bsMAb i.v. and 5 MBq of 68Ga-labeled peptide i.v. 16 h later. One hour after the injection of the peptide or FDG, PET/CT images were acquired. One day later the same procedure was repeated vice versa, and after microPET imaging the uptake of the 68Ga or 18F in dissected tissue was determined. Results: Within one hour, specific tumor targeting of 68Ga-labeled peptide (23.4 ± 7.2% ID/g) combined with the low background activity levels resulted in the clear visualization of the small intra-abdominal tumor lesions (1.5-5 mm). High tumor/background radioactivity concentration ratios were obtained (e.g. tumor/intestines 57.5 ± 22.4). FDG localized efficiently in the tumors (8.7 ± 3.1 % ID/g), but with physiological uptake in various normal tissues (ratio tumor/intestines 4.0 ± 0.9). Conclusion: Pretargeted immunoPET performs exceptionally well with short-lived radionuclides, and is a highly sensitive procedure for dectection of small tumor deposits that is also more specific than FDG-PET. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 5282. doi:10.1158/1538-7445.AM2011-5282


Clinical Cancer Research | 2010

Abstract A32: Pretargeted immunoPET of CEA-expressing in intraperitoneal human colonic tumor xenografts in nude mice

Rafke Schoffelen; Robert M. Sharkey; Gerben M. Franssen; David M. Goldenberg; William J. McBride; Edmund A. Rossi; Chien-Hsing Chang; Peter Laverman; Winette T.A. Graaf; Wim J.G. Oyen; Otto C. Boerman

Objective: Radiolabeled antibodies were being developed commercially for the detection of several cancer types. However, imaging with radiolabeled antibodies require a relatively long interval between injection and imaging acquisition for adequate contrast to develop. The pretargeting method is a 2-step method: a unlabeled bsMAb with affinity for the tumor and a small radiolabeled molecule is injected, followed by the a radiolabeled compound. This aims to optimize the imaging system by specific tumor accumulation and fast blood clearance of the radiopharmacon, which allows imaging within one hour. Short-lived positron emitting radionuclides, e.g. 68Ga (half life: 68 min) match the pharmacokinetics of the small molecule. In this study, pretargeted immunoPET with a bispecific monoclonal anti-carcinoembryonic antigen (CEA) x anti-histamine-succinyl-glycine (HSG) antibody, TF2, and a radiolabeled peptide, IMP288, was compared to 18F-FDG in detecting CEA-expressing tumors in nude mice. Methods: Groups of five BALB/c nude mice were inoculated with CEA-expressing human colonic tumor cells intraperitoneally. One group received 5 MBq 18F-FDG intravenously (i.v.). Another group received TF2 and 5 MBq 68Ga-IMP288 i.v. 16 h later. After one hour PET/CT images were acquired. One day later this was repeated vice versa. Uptake of 68Ga-IMP288 or 18F-FDG in the tumors and other dissected tissues was determined. Results: High, specific tumor uptake of 68Ga-IMP288 (23.4 ± 7.2% ID/g) was observed, with very low accretion in the normal tissues (intestines: 0.50 ± 0.19 % ID/g; liver: 1.83 ± 0.45 % ID/g). This resulted in high tumor-to-background ratios (e.g. tumor-to-intestines 57.5 ± 22.4) and in the clear visualization of the small intra-abdominal tumor lesions (1.5–5 mm), as soon as one hour after the injection of 68Ga-IMP288. 18F-FDG localized efficiently in the tumors (8.7 ± 3.1 % ID/g), but with physiological uptake in various normal tissues (intestines: 2.15 ± 0.61 % ID/g; liver: 2.78 ± 0.52 % ID/g). The low tumor-to-background ratio (e.g. tumor-to-intestines 4.0 ± 0.9) complicated the detection of the tumors. Conclusions: This study indicated that pretargeted immunoPET with TF2 and 68Ga-IMP288 is a specific and sensitive method for detecting colon cancer. Citation Information: Clin Cancer Res 2010;16(7 Suppl):A32


Journal of Clinical Oncology | 2011

Androgen Receptor–Positive Salivary Duct Carcinoma: A Disease Entity With Promising New Treatment Options

Heidy C.J. Jaspers; Berit M. Verbist; Rafke Schoffelen; Vera Mattijssen; Piet J. Slootweg; Winette T. A. van der Graaf; Carla M.L. van Herpen


European Journal of Nuclear Medicine and Molecular Imaging | 2014

Predictive patient-specific dosimetry and individualized dosing of pretargeted radioimmunotherapy in patients with advanced colorectal cancer

Rafke Schoffelen; Wietske Woliner-van der Weg; Eric P. Visser; David M. Goldenberg; Robert M. Sharkey; William J. McBride; Chien-Hsing Chang; Edmund A. Rossi; Winette T. A. van der Graaf; Wim J.G. Oyen; Otto C. Boerman


The Journal of Nuclear Medicine | 2011

Phase I clinical study of the feasibility of pretargeted radioimmunotherapy (PT-RAIT) in patients with colorectal cancer (CRC): First results

Rafke Schoffelen; Otto C. Boerman; Winette T. A. van der Graaf; Carla M.L. van Herpen; Robert M. Sharkey; William H. McBride; Chien-Hsing Chang; Edmund A. Rossi; David M. Goldenberg; Wim J.G. Oyen

Collaboration


Dive into the Rafke Schoffelen's collaboration.

Top Co-Authors

Avatar

David M. Goldenberg

Roswell Park Cancer Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Otto C. Boerman

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Winette T. A. van der Graaf

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Wim J.G. Oyen

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Gerben M. Franssen

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Chien-Hsing Chang

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

William J. McBride

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

Edmund A. Rossi

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peter Laverman

Radboud University Nijmegen

View shared research outputs
Researchain Logo
Decentralizing Knowledge