Network


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

Hotspot


Dive into the research topics where Anton G.T. Terwisscha van Scheltinga is active.

Publication


Featured researches published by Anton G.T. Terwisscha van Scheltinga.


The Journal of Nuclear Medicine | 2011

Intraoperative Near-Infrared Fluorescence Tumor Imaging with Vascular Endothelial Growth Factor and Human Epidermal Growth Factor Receptor 2 Targeting Antibodies

Anton G.T. Terwisscha van Scheltinga; Gooitzen M. van Dam; Wouter B. Nagengast; Vasilis Ntziachristos; Harry Hollema; Jennifer Lynn Herek; Carolien P. Schröder; Jos G. W. Kosterink; Marjolijn N. Lub-de Hoog; Elisabeth G.E. de Vries

Fluorescence imaging is currently attracting much interest as a method for intraoperative tumor detection, but most current tracers lack tumor specificity. Therefore, this technique can be further improved by tumor-specific detection. With tumor-targeted antibodies bound to a radioactive label, tumor-specific SPECT or PET is feasible in the clinical setting. The aim of the present study was to apply antibody-based tumor detection to intraoperative optical imaging, using preclinical in vivo mouse models. Methods: Anti–vascular endothelial growth factor (VEGF) antibody bevacizumab and anti–human epidermal growth factor receptor (HER) 2 antibody trastuzumab were labeled with the near-infrared (NIR) fluorescence dye IRDye 800CW. Tumor uptake of the fluorescent tracers and their 89Zr-labeled radioactive counterparts for PET was determined in human xenograft–bearing athymic mice during 1 wk after tracer injection, followed by ex vivo biodistribution and pathologic examination. Intraoperative imaging of fluorescent VEGF- or HER2-positive tumor lesions was performed in subcutaneous tumors and in intraperitoneal dissemination tumor models. Results: Tumor-to-background ratios, with fluorescent imaging, were 1.93 ± 0.40 for bevacizumab and 2.92 ± 0.29 for trastuzumab on day 6 after tracer injection. Real-time intraoperative imaging detected tumor lesions at even the submillimeter level in intraperitoneal dissemination tumor models. These results were supported by standard histology, immunohistochemistry, and fluorescence microscopy analyses. Conclusion: NIR fluorescence–labeled antibodies targeting VEGF or HER2 can be used for highly specific and sensitive detection of tumor lesions in vivo. These preclinical findings encourage future clinical studies with NIR fluorescence–labeled tumor-specific antibodies for intraoperative-guided surgery in cancer patients.


Journal of Clinical Oncology | 2015

Antibody Positron Emission Tomography Imaging in Anticancer Drug Development

Laetitia E. Lamberts; Simon Williams; Anton G.T. Terwisscha van Scheltinga; Marjolijn N. Lub-de Hooge; Carolien P. Schröder; Jourik A. Gietema; Adrienne H. Brouwers; Elisabeth G.E. de Vries

More than 50 monoclonal antibodies (mAbs), including several antibody-drug conjugates, are in advanced clinical development, forming an important part of the many molecularly targeted anticancer therapeutics currently in development. Drug development is a relatively slow and expensive process, limiting the number of drugs that can be brought into late-stage trials. Development decisions could benefit from quantitative biomarkers, enabling visualization of the tissue distribution of (potentially modified) therapeutic mAbs to confirm effective whole-body target expression, engagement, and modulation and to evaluate heterogeneity across lesions and patients. Such biomarkers may be realized with positron emission tomography imaging of radioactively labeled antibodies, a process called immunoPET. This approach could potentially increase the power and value of early trials by improving patient selection, optimizing dose and schedule, and rationalizing observed drug responses. In this review, we summarize the available literature and the status of clinical trials regarding the potential of immunoPET during early anticancer drug development.


Clinical Cancer Research | 2012

Measurement of tumor VEGF-A levels with 89Zr-bevacizumab PET as an early biomarker for the antiangiogenic effect of everolimus treatment in an ovarian cancer xenograft model

Arne R.M. van der Bilt; Anton G.T. Terwisscha van Scheltinga; Hetty Timmer-Bosscha; Carolien P. Schröder; Linda Pot; Jos G. W. Kosterink; Ate G.J. van der Zee; Marjolijn N. Lub-de Hooge; Steven de Jong; Elisabeth G.E. de Vries; Anna K.L. Reyners

Purpose: The mTOR pathway is frequently activated in ovarian cancers. mTOR inhibitors, such as everolimus, can reduce VEGF-A production by cancer cells. We investigated whether early everolimus treatment effects could be monitored by positron emission tomography (PET) with 89Zr-bevacizumab. Experimental Design: The effect of everolimus on VEGF-A secretion was determined in a panel of human ovarian cancer cell lines and in A2780luc+ ovarian cancer cells xenografted subcutaneously in BALB/c mice. Mice received daily 10 mg/kg everolimus intraperitoneally (i.p.) for 14 days. PET scans with the tracer 89Zr-labeled bevacizumab were conducted before and after treatment. Ex vivo 89Zr-bevacizumab biodistribution and correlative tissue analyses were conducted. Tumor VEGF-A levels were measured with ELISA and mean vascular density (MVD) was determined with immunohistochemistry. Results: Everolimus treatment reduced VEGF-A levels in the supernatant of all cell lines. Everolimus lowered 89Zr-bevacizumab tumor uptake by 21.7% ± 4.0% [mean standardized uptake value (SUVmean) 2.3 ± 0.2 vs. 2.9 ± 0.2, P < 0.01]. Ex vivo biodistribution also showed lower tracer uptake in the tumors of treated as compared with control animals (7.8 ± 0.8%ID/g vs. 14.0 ± 1.7%ID/g, P < 0.01), whereas no differences were observed for other tissues. This coincided with lower VEGF-A protein levels in tumor lysates in treated versus untreated tumors (P = 0.04) and reduced MVD (P < 0.01). Conclusion: Tumor VEGF-A levels are decreased by everolimus. 89Zr-bevacizumab PET could be used to monitor tumor VEGF-A levels as an early biomarker of the antiangiogenic effect of mTOR inhibitor therapy. Clin Cancer Res; 18(22); 6306–14. ©2012 AACR.


Pharmacology & Therapeutics | 2014

HER3, serious partner in crime Therapeutic approaches and potential biomarkers for effect of HER3-targeting

Arjan Kol; Anton G.T. Terwisscha van Scheltinga; Hetty Timmer-Bosscha; Laetitia E. Lamberts; Frederike Bensch; Elisabeth G.E. de Vries; Carolina P. Schröder

The human epidermal growth factor receptor (HER) family members are targeted by a growing numbers of small molecules and monoclonal antibodies. Resistance against the epidermal growth factor receptor (EGFR) and HER2-targeting agents is a clinically relevant problem forcing research on optimizing targeting of the HER family. In view of its overexpression in tumors, and compensatory role in HER signaling, HER3 has gained much interest as a potential additional target within the HER family. It is the only member of the HER family lacking intrinsic tyrosine kinase activity and therefore its role in cancer has long been underestimated. Drugs that block HER3 or interfere with HER3 dimer signaling, including fully human anti-HER3 antibodies, bispecific antibodies and tyrosine kinase inhibitors (TKIs), are currently becoming available. Several compounds have already entered clinical trial. In the meantime potential biomarkers are tested such as tumor analysis of HER3 expression, functional assays for downstream effector molecules and molecular imaging techniques. This review describes the biology and relevance of HER3 in cancer, agents targeting HER3 and potential biomarkers for effect of HER3-targeting.


Clinical Cancer Research | 2017

Tumor-specific uptake of fluorescent bevacizumab-IRDye800CW microdosing in patients with primary breast cancer: a phase I feasibility study

Laetitia E. Lamberts; Maximillian Koch; Johannes S. de Jong; Arthur Adams; Jürgen Glatz; Mariëtte E.G. Kranendonk; Anton G.T. Terwisscha van Scheltinga; Liesbeth Jansen; Jakob de Vries; Marjolijn N. Lub-de Hooge; Carolien P. Schröder; Annelies Jorritsma-Smit; Matthijs D. Linssen; Esther de Boer; Bert van der Vegt; Wouter B. Nagengast; Sjoerd G. Elias; Sabrina Oliveira; Arjen J. Witkamp; Willem P. Th. M. Mali; Elsken van der Wall; Paul J. van Diest; Elisabeth G.E. de Vries; Vasilis Ntziachristos; Gooitzen M. van Dam

Purpose: To provide proof of principle of safety, breast tumor–specific uptake, and positive tumor margin assessment of the systemically administered near-infrared fluorescent tracer bevacizumab–IRDye800CW targeting VEGF-A in patients with breast cancer. Experimental Design: Twenty patients with primary invasive breast cancer eligible for primary surgery received 4.5 mg bevacizumab–IRDye800CW as intravenous bolus injection. Safety aspects were assessed as well as tracer uptake and tumor delineation during surgery and ex vivo in surgical specimens using an optical imaging system. Ex vivo multiplexed histopathology analyses were performed for evaluation of biodistribution of tracer uptake and coregistration of tumor tissue and healthy tissue. Results: None of the patients experienced adverse events. Tracer levels in primary tumor tissue were higher compared with those in the tumor margin (P < 0.05) and healthy tissue (P < 0.0001). VEGF-A tumor levels also correlated with tracer levels (r = 0.63, P < 0.0002). All but one tumor showed specific tracer uptake. Two of 20 surgically excised lumps contained microscopic positive margins detected ex vivo by fluorescent macro- and microscopy and confirmed at the cellular level. Conclusions: Our study shows that systemic administration of the bevacizumab–IRDye800CW tracer is safe for breast cancer guidance and confirms tumor and tumor margin uptake as evaluated by a systematic validation methodology. The findings are a step toward a phase II dose-finding study aimed at in vivo margin assessment and point to a novel drug assessment tool that provides a detailed picture of drug distribution in the tumor tissue. Clin Cancer Res; 23(11); 2730–41. ©2016 AACR.


mAbs | 2014

ImmunoPET and biodistribution with human epidermal growth factor receptor 3 targeting antibody 89Zr-RG7116

Anton G.T. Terwisscha van Scheltinga; Marjolijn N. Lub-de Hooge; Keelara Abiraj; Carolien P. Schröder; Linda Pot; Birgit Bossenmaier; Marlene Thomas; Gabriele Hölzlwimmer; Thomas Friess; Jos G. W. Kosterink; Elisabeth G.E. de Vries

The humanized monoclonal antibody with high affinity for the human epidermal growth factor receptor (HER) 3, RG7116, is a glycoengineered, IgG1 class antibody. By labeling RG7116 with zirconium-89 (89Zr) we aimed to visualize in vivo HER3 expression and study the biodistribution of this antibody in human tumor-bearing mice. Biodistribution of 89Zr-RG7116 was studied in subcutaneously xenografted FaDu tumor cells (HER3-positive). Dose-dependency of 89Zr-RG7116 organ distribution and specific tumor uptake was assessed by administering doses ranging from 0.05 to 10 mg/kg RG7116 to SCID/Beige mice. Biodistribution was analyzed at 24 and 144 h after injection. MicroPET imaging was performed at 1, 3, and 6 days after injection of 1.0 mg/kg 89Zr-RG7116 in the FaDu, H441, QG-56 and Calu-1 xenografts with varying HER3 expression. The excised tumors were analyzed for HER3 expression. Biodistribution analyses showed a dose- and time-dependent 89Zr-RG7116 tumor uptake in FaDu tumors. The highest tumor uptake of 89Zr-RG7116 was observed in the 0.05 mg/kg dose group with 27.5%ID/g at 144 h after tracer injection. MicroPET imaging revealed specific tumor uptake of 89Zr-RG7116 in FaDu and H441 models with an increase in tumor uptake over time. Biodistribution data was consistent with the microPET findings in FaDu, H441, QG56 and Calu-1 xenografts, which correlated with HER3 expression levels. In conclusion, 89Zr-RG7116 specifically accumulates in HER3 expressing tumors. PET imaging with this tracer provides real-time non-invasive information about RG7116 distribution, tumor targeting and tumor HER3 expression levels.


The Journal of Nuclear Medicine | 2014

In Vivo Visualization of MET Tumor Expression and Anticalin Biodistribution with the MET-Specific Anticalin 89Zr-PRS-110 PET Tracer

Anton G.T. Terwisscha van Scheltinga; Marjolijn N. Lub-de Hooge; Marlon Hinner; Remy B. Verheijen; Andrea Allersdorfer; Martin Hülsmeyer; Wouter B. Nagengast; Carolien P. Schröder; Jos G. W. Kosterink; Elisabeth G.E. de Vries; Laurent Audoly; Shane Olwill

Anticalins are a novel class of biopharmaceuticals, displaying highly desirable attributes as imaging agents. The anticalin PRS-110 was rationally engineered to target the oncogene MET with high affinity and specificity. The aim of this study was to visualize MET expression and analyze biodistribution of 89Zr-labeled PRS-110 in human tumor–bearing mice. Methods: 89Zr-PRS-110 was generated. For biodistribution studies (96 h after injection of tracer) 10 μg of 89Zr-PRS-110 (with 0–490 μg of unlabeled PRS-110) were injected into BALB/c mice bearing high MET-expressing H441 non–small cell lung cancer xenografts. Further characterization with PET imaging was performed at 6, 24, 48, and 96 h after injection of 50 μg of 89Zr-PRS-110 into mice bearing H441, primary glioblastoma U87-MG (intermediate MET), or ovarian cancer A2780 (low MET) xenografts. Drug distribution was also analyzed ex vivo using fluorescently labeled PRS-110. Results: Biodistribution analyses showed a dose-dependent tumor uptake of 89Zr-PRS-110, with the highest fractional tumor uptake at 10 μg of 89Zr-PRS-110, with no unlabeled PRS-110. Small-animal PET imaging supported by biodistribution data revealed specific tumor uptake of 89Zr-PRS-110 in the MET-expressing H441 and U87-MG tumors whereas the MET-negative A2780 tumor model showed a lower uptake similar to a non-MET binder anticalin control. Tumor uptake increased up to 24 h after tracer injection and remained high, whereas uptake in other organs decreased over time. Ex vivo fluorescence revealed intracellular presence of PRS-110. Conclusion: 89Zr-PRS-110 specifically accumulates in MET-expressing tumors in a receptor density–dependent manner. PET imaging provides real-time noninvasive information about PRS-110 distribution and tumor accumulation in preclinical models.


Pharmacology & Therapeutics | 2014

Associate editor: B. TeicherHER3, serious partner in crime: Therapeutic approaches and potential biomarkers for effect of HER3-targeting

Arjan Kol; Anton G.T. Terwisscha van Scheltinga; Hetty Timmer-Bosscha; Laetitia E. Lamberts; Frederike Bensch; Elisabeth G.E. de Vries; Carolina P. Schröder

The human epidermal growth factor receptor (HER) family members are targeted by a growing numbers of small molecules and monoclonal antibodies. Resistance against the epidermal growth factor receptor (EGFR) and HER2-targeting agents is a clinically relevant problem forcing research on optimizing targeting of the HER family. In view of its overexpression in tumors, and compensatory role in HER signaling, HER3 has gained much interest as a potential additional target within the HER family. It is the only member of the HER family lacking intrinsic tyrosine kinase activity and therefore its role in cancer has long been underestimated. Drugs that block HER3 or interfere with HER3 dimer signaling, including fully human anti-HER3 antibodies, bispecific antibodies and tyrosine kinase inhibitors (TKIs), are currently becoming available. Several compounds have already entered clinical trial. In the meantime potential biomarkers are tested such as tumor analysis of HER3 expression, functional assays for downstream effector molecules and molecular imaging techniques. This review describes the biology and relevance of HER3 in cancer, agents targeting HER3 and potential biomarkers for effect of HER3-targeting.


Molecular Imaging | 2013

Feasibility of Vascular Endothelial Growth Factor Imaging in Human Atherosclerotic Plaque Using Zr-89-Bevacizumab Positron Emission Tomography

Reza Golestani; Clark J. Zeebregts; Anton G.T. Terwisscha van Scheltinga; Marjolijn N. Lub-de Hooge; Gooitzen M. van Dam; Andor W. J. M. Glaudemans; Rudi Dierckx; René A. Tio; Albert J. H. Suurmeijer; Hendrikus Boersma; Wouter B. Nagengast; Riemer H. J. A. Slart

Intraplaque angiogenesis is associated with the occurrence of atherosclerotic plaque rupture. Cardiovascular molecular imaging can be used for the detection of rupture-prone plaques. Imaging with radiolabeled bevacizumab, a monoclonal anti-vascular endothelial growth factor (VEGF)-A, can depict VEGF levels corresponding to the angiogenic status in tumors. We determined the feasibility of 89Zr-bevacizumab imaging for the detection of VEGF in carotid endarterectomy (CEA) specimens. Five CEA specimens were coincubated with 89Zr-bevacizumab and aspecific 111In-labeled IgG to determine the specificity of bevacizumab accumulation. In 11 CEA specimens, 89Zr-bevacizumab micro-positron emission tomography (PET) was performed following 2 hours of incubation. Specimens were cut in 4 mm wide segments and were stained for VEGF and CD68. In each segment, the mean percent incubation dose per gram of tissue (%Inc/g) and tissue to background ratio were determined. A 10-fold higher accumulation of 89Zr-bevacizumab compared to 111In-IgG uptake was demonstrated by gamma counting. The mean %Inc/ghot spot was 2.2 ± 0.9 with a hot spot to background ratio of 3.6 ± 0.8. There was a significant correlation between the segmental tissue to background uptake ratio and the VEGF score (ρ = .74, p < .001). It is feasible to detect VEGF tissue concentration within CEA specimens using 89Zr-bevacizumab PET. 89Zr-bevacizumab accumulation in plaques is specific and correlates with immunohistochemistry scores.


The Journal of Nuclear Medicine | 2016

Molecular Fluorescence Endoscopy Targeting Vascular Endothelial Growth Factor A for Improved Colorectal Polyp Detection

Jolien J. Tjalma; P. Beatriz Garcia-Allende; Elmire Hartmans; Anton G.T. Terwisscha van Scheltinga; Wytske Boersma-van Ek; Juergen Glatz; Maximilian Koch; Yasmijn van Herwaarden; Tanya M. Bisseling; Iris D. Nagtegaal; Hetty Timmer-Bosscha; Jan J. Koornstra; A Karrenbeld; Jan H. Kleibeuker; Gooitzen M. van Dam; Vasilis Ntziachristos; Wouter B. Nagengast

Small and flat adenomas are known to carry a high miss-rate during standard white-light endoscopy. Increased detection rate may be achieved by molecular fluorescence endoscopy with targeted near-infrared (NIR) fluorescent tracers. The aim of this study was to validate vascular endothelial growth factor A (VEGF-A) and epidermal growth factor receptor (EGFR)–targeted fluorescent tracers during ex vivo colonoscopy with an NIR endoscopy platform. Methods: VEGF-A and EGFR expression was determined by immunohistochemistry on a large subset of human colorectal tissue samples—48 sessile serrated adenomas/polyps, 70 sporadic high-grade dysplastic adenomas, and 19 hyperplastic polyps—and tissue derived from patients with Lynch syndrome—78 low-grade dysplastic adenomas, 57 high-grade dysplastic adenomas, and 31 colon cancer samples. To perform an ex vivo colonoscopy procedure, 14 mice with small intraperitoneal EGFR-positive HCT116luc tumors received intravenous bevacizumab-800CW (anti-VEGF-A), cetuximab-800CW (anti-EGFR), control tracer IgG-800CW, or sodium chloride. Three days later, 8 resected HCT116luc tumors (2–5 mm) were stitched into 1 freshly resected human colon specimen and followed by an ex vivo molecular fluorescence colonoscopy procedure. Results: Immunohistochemistry showed high VEGF-A expression in 79%–96% and high EGFR expression in 51%–69% of the colorectal lesions. Both targets were significantly overexpressed in the colorectal lesions, compared with the adjacent normal colon crypts. During ex vivo molecular fluorescence endoscopy, all tumors could clearly be delineated for both bevacizumab-800CW and cetuximab-800CW tracers. Specific tumor uptake was confirmed with fluorescent microscopy showing, respectively, stromal and cell membrane fluorescence. Conclusion: VEGF-A is a promising target for molecular fluorescence endoscopy because it showed a high protein expression, especially in sessile serrated adenomas/polyps and Lynch syndrome. We demonstrated the feasibility to visualize small tumors in real time during colonoscopy using a NIR fluorescence endoscopy platform, providing the endoscopist a wide-field red-flag technique for adenoma detection. Clinical studies are currently being performed in order to provide in-human evaluation of our approach.

Collaboration


Dive into the Anton G.T. Terwisscha van Scheltinga's collaboration.

Top Co-Authors

Avatar

Elisabeth G.E. de Vries

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Marjolijn N. Lub-de Hooge

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Carolien P. Schröder

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Wouter B. Nagengast

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Jos G. W. Kosterink

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Arjan Kol

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Laetitia E. Lamberts

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Steven de Jong

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Martin Pool

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Gooitzen M. van Dam

University Medical Center Groningen

View shared research outputs
Researchain Logo
Decentralizing Knowledge