Network


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

Hotspot


Dive into the research topics where Annelies Jorritsma-Smit is active.

Publication


Featured researches published by Annelies Jorritsma-Smit.


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.


The Lancet Gastroenterology & Hepatology | 2016

Molecular fluorescence-guided surgery of peritoneal carcinomatosis of colorectal origin: a single-centre feasibility study

Niels J. Harlaar; Marjory Koller; Steven J. de Jongh; Barbara L. van Leeuwen; Patrick H. J. Hemmer; S. Kruijff; Robert J. van Ginkel; Lukas B. Been; Johannes S. de Jong; Gursah Kats-Ugurlu; Matthijs D. Linssen; Annelies Jorritsma-Smit; Marleen van Oosten; Wouter B. Nagengast; Vasilis Ntziachristos; Gooitzen M. van Dam

BACKGROUND Optimum cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy (HIPEC) is essential for the curative treatment of peritoneal carcinomatosis of colorectal origin. At present, surgeons depend on visual inspection and palpation for tumour detection. Improved detection of tumour tissue using molecular fluorescence-guided surgery could not only help attain a complete cytoreduction of metastatic lesions, but might also prevent overtreatment by avoiding resection of benign lesions. METHODS For this non-randomised, single-centre feasibility study, we enrolled patients with colorectal peritoneal metastases scheduled for cytoreductive surgery and HIPEC. 2 days before surgery, 4·5 mg of the near-infrared fluorescent tracer bevacizumab-IRDye800CW was administered intravenously. The primary objectives were to determine the safety and feasibility of molecular fluorescence-guided surgery using bevacizumab-IRDye800CW. Molecular fluorescence-guided surgery was deemed safe if no allergic or anaphylactic reactions were recorded and no serious adverse events were attributed to bevacizumab-IRDye800CW. The technique was deemed feasible if bevacizumab-IRDye800CW enabled detection of fluorescence signals intraoperatively. Secondary objectives were correlation of fluorescence with histopathology by back-table imaging of the fresh surgical specimen and semi-quantitative ex-vivo analyses of formalin-fixed paraffin embedded (FFPE) tissue on all peritoneal lesions. Additionally, VEGF-α staining and fluorescence microscopy was done. This study is registered with the Netherlands Trial Registry, number NTR4632. FINDINGS Between July 3, 2014, and March 2, 2015, seven patients were enrolled in the study. One patient developed an abdominal sepsis 5 days postoperatively and another died from an asystole 4 days postoperatively, most probably due to a cardiovascular thromboembolic event. However, both serious adverse events were attributed to the surgical cytoreductive surgery and HIPEC procedure. No serious adverse events related to bevacizumab-IRDye800CW occurred in any of the patients. Intraoperatively, fluorescence was seen in all patients. In two patients, additional tumour tissue was detected by molecular fluorescence-guided surgery that was initially missed by the surgeons. During back-table imaging of fresh surgical specimens, a total of 80 areas were imaged, marked, and analysed. All of the 29 non-fluorescent areas were found to contain only benign tissue, whereas tumour tissue was detected in 27 of 51 fluorescent areas (53%). Ex-vivo semi-quantification of 79 FFPE peritoneal lesions showed a tumour-to-normal ratio of 6·92 (SD 2·47). INTERPRETATION Molecular fluorescence-guided surgery using the near-infrared fluorescent tracer bevacizumab-IRDye800CW is safe and feasible. This technique might be of added value for the treatment of patients with colorectal peritoneal metastases through improved patient selection and optimisation of cytoreductive surgery. A subsequent multicentre phase 2 trial is needed to make a definitive assessment of the diagnostic accuracy and the effect on clinical decision making of molecular fluorescence-guided surgery. FUNDING FP-7 Framework Programme BetaCure and SurgVision BV.


Cancer Research | 2017

Threshold Analysis and Biodistribution of Fluorescently Labeled Bevacizumab in Human Breast Cancer.

Maximilian Koch; Johannes S. de Jong; Jürgen Glatz; Panagiotis Symvoulidis; Laetitia E. Lamberts; Arthur Adams; Mariëtte E.G. Kranendonk; Anton G.T. Terwisscha van Scheltinga; Michaela Aichler; 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; P. Beatriz Garcia-Allende; Paul J. van Diest; Elisabeth G.E. de Vries; Axel Walch; Gooitzen M. van Dam; Vasilis Ntziachristos

In vivo tumor labeling with fluorescent agents may assist endoscopic and surgical guidance for cancer therapy as well as create opportunities to directly observe cancer biology in patients. However, malignant and nonmalignant tissues are usually distinguished on fluorescence images by applying empirically determined fluorescence intensity thresholds. Here, we report the development of fSTREAM, a set of analytic methods designed to streamline the analysis of surgically excised breast tissues by collecting and statistically processing hybrid multiscale fluorescence, color, and histology readouts toward precision fluorescence imaging. fSTREAM addresses core questions of how to relate fluorescence intensity to tumor tissue and how to quantitatively assign a normalized threshold that sufficiently differentiates tumor tissue from healthy tissue. Using fSTREAM we assessed human breast tumors stained in vivo with fluorescent bevacizumab at microdose levels. Showing that detection of such levels is achievable, we validated fSTREAM for high-resolution mapping of the spatial pattern of labeled antibody and its relation to the underlying cancer pathophysiology and tumor border on a per patient basis. We demonstrated a 98% sensitivity and 79% specificity when using labeled bevacizumab to outline the tumor mass. Overall, our results illustrate a quantitative approach to relate fluorescence signals to malignant tissues and improve the theranostic application of fluorescence molecular imaging. Cancer Res; 77(3); 623-31. ©2016 AACR.


European Journal of Pharmaceutics and Biopharmaceutics | 2016

Development, preclinical safety, formulation, and stability of clinical grade bevacizumab-800CW, a new near infrared fluorescent imaging agent for first in human use

Eva J. ter Weele; Anton G.T. Terwisscha van Scheltinga; Matthijs D. Linssen; Wouter B. Nagengast; Ingo Lindner; Annelies Jorritsma-Smit; Elisabeth G.E. de Vries; Jos G. W. Kosterink; Marjolijn N. Lub-de Hooge

There is a dire need for better visualization of cancer and analysis of specific targets in vivo. Molecular imaging with fluorescence is gaining more and more attention, as it allows detection of these targets and has advantages over radioactivity, such as no radiation dose, and lower costs. A key challenge in optical imaging however, is translation of the newly developed tracers from pre-clinical phase to clinical application. We describe the development and safety testing of clinical grade bevacizumab-800CW, an antibody-based targeted agent for non-invasive imaging of vascular endothelial growth factor A (VEGF-A). Development included implementing the manufacturing process and analytical methods according to current Good Manufacturing Practice (cGMP), formulation studies, extended characterization and stability testing. For safety pharmacology an extended single dose toxicity study in mice was performed. Bevacizumab-800CW was formulated in isotonic phosphate buffered sodium chloride solution at pH 7. The production was robust and showed a reproducible labeling efficiency, and no impurities. The binding affinity to VEGF-A remained intact. The optimized product meets all release specifications, is stable up to at least 3months and its characteristics did not significantly differ from the unlabeled bevacizumab. Toxicity testing in mice showed no remarkable findings. In conclusion, sterile bevacizumab-800CW (6mg=6ml) can be produced in stock according to current Good Manufacturing Practice. It is ready for first-in-human use.


Clinical Cancer Research | 2017

Zr-89-Lumretuzumab PET Imaging before and during HER3 Antibody Lumretuzumab Treatment in Patients with Solid Tumors

Frederike Bensch; Laetitia E. Lamberts; Michaël M. Smeenk; Annelies Jorritsma-Smit; Marjolijn N. Lub-de Hooge; Anton G.T. Terwisscha van Scheltinga; Johan R. de Jong; Jourik A. Gietema; Carolien P. Schröder; Marlene Thomas; Wolfgang Jacob; Keelara Abiraj; Celine Adessi; Georgina Meneses-Lorente; Ian James; Martin Weisser; Adrienne H. Brouwers; Elisabeth G.E. de Vries

Purpose: We evaluated biodistribution and tumor targeting of 89Zr-lumretuzumab before and during treatment with lumretuzumab, a human epidermal growth factor receptor 3 (HER3)–targeting monoclonal antibody. Experimental Design: Twenty patients with histologically confirmed HER3-expressing tumors received 89Zr-lumretuzumab and underwent positron emission tomography (PET). In part A, 89Zr-lumretuzumab was given with additional, escalating doses of unlabeled lumretuzumab, and scans were performed 2, 4, and 7 days after injection to determine optimal imaging conditions. In part B, patients were scanned following tracer injection before (baseline) and after a pharmacodynamic (PD)-active lumretuzumab dose for saturation analysis. HER3 expression was determined immunohistochemically in skin biopsies. Tracer uptake was calculated as standardized uptake value (SUV). Results: Optimal PET conditions were found to be 4 and 7 days after administration of 89Zr-lumretuzumab with 100-mg unlabeled lumretuzumab. At baseline using 100-mg unlabeled lumretuzumab, the tumor SUVmax was 3.4 (±1.9) at 4 days after injection. SUVmean values for normal blood, liver, lung, and brain tissues were 4.9, 6.4, 0.9 and 0.2, respectively. Saturation analysis (n = 7) showed that 4 days after lumretuzumab administration, tumor uptake decreased by 11.9% (±8.2), 10.0% (±16.5), and 24.6% (±20.9) at PD-active doses of 400, 800, and 1,600 mg, respectively, when compared with baseline. Membranous HER3 was completely downregulated in paired skin biopsies already at and above 400-mg lumretuzumab. Conclusions: PET imaging showed biodistribution and tumor-specific 89Zr-lumretuzumab uptake. Although, PD-active lumretuzumab doses decreased 89Zr-lumretuzumab uptake, there was no clear evidence of tumor saturation by PET imaging as the tumor SUV did not plateau with increasing doses. Clin Cancer Res; 23(20); 6128–37. ©2017 AACR.


Theranostics | 2018

Potential Red-Flag Identification of Colorectal Adenomas with Wide-Field Fluorescence Molecular Endoscopy

Elmire Hartmans; Jolien J. Tjalma; Matthijs D. Linssen; Pilar Beatriz Garcia Allende; Marjory Koller; Annelies Jorritsma-Smit; Mariana e Silva de Oliveira Nery; Sjoerd G. Elias; Arend Karrenbeld; Elisabeth G.E. de Vries; Jan H. Kleibeuker; Gooitzen M. van Dam; Dominic J. Robinson; Vasilis Ntziachristos; Wouter B. Nagengast

Adenoma miss rates in colonoscopy are unacceptably high, especially for sessile serrated adenomas / polyps (SSA/Ps) and in high-risk populations, such as patients with Lynch syndrome. Detection rates may be improved by fluorescence molecular endoscopy (FME), which allows morphological visualization of lesions with high-definition white-light imaging as well as fluorescence-guided identification of lesions with a specific molecular marker. In a clinical proof-of-principal study, we investigated FME for colorectal adenoma detection, using a fluorescently labelled antibody (bevacizumab-800CW) against vascular endothelial growth factor A (VEGFA), which is highly upregulated in colorectal adenomas. Methods: Patients with familial adenomatous polyposis (n = 17), received an intravenous injection with 4.5, 10 or 25 mg of bevacizumab-800CW. Three days later, they received NIR-FME. Results: VEGFA-targeted NIR-FME detected colorectal adenomas at all doses. Best results were achieved in the highest (25 mg) cohort, which even detected small adenomas (<3 mm). Spectroscopy analyses of freshly excised specimen demonstrated the highest adenoma-to-normal ratio of 1.84 for the 25 mg cohort, with a calculated median tracer concentration in adenomas of 6.43 nmol/mL. Ex vivo signal analyses demonstrated NIR fluorescence within the dysplastic areas of the adenomas. Conclusion: These results suggest that NIR-FME is clinically feasible as a real-time, red-flag technique for detection of colorectal adenomas.


Nature Communications | 2018

Implementation and benchmarking of a novel analytical framework to clinically evaluate tumor-specific fluorescent tracers

Marjory Koller; Si-Qi Qiu; Matthijs D. Linssen; Liesbeth Jansen; Wendy Kelder; Jakob de Vries; Inge Kruithof; Guo-Jun Zhang; Dominic J. Robinson; Wouter B. Nagengast; Annelies Jorritsma-Smit; Bert van der Vegt; Gooitzen M. van Dam

During the last decade, the emerging field of molecular fluorescence imaging has led to the development of tumor-specific fluorescent tracers and an increase in early-phase clinical trials without having consensus on a standard methodology for evaluating an optical tracer. By combining multiple complementary state-of-the-art clinical optical imaging techniques, we propose a novel analytical framework for the clinical translation and evaluation of tumor-targeted fluorescent tracers for molecular fluorescence imaging which can be used for a range of tumor types and with different optical tracers. Here we report the implementation of this analytical framework and demonstrate the tumor-specific targeting of escalating doses of the near-infrared fluorescent tracer bevacizumab-800CW on a macroscopic and microscopic level. We subsequently demonstrate an 88% increase in the intraoperative detection rate of tumor-involved margins in primary breast cancer patients, indicating the clinical feasibility and support of future studies to evaluate the definitive clinical impact of fluorescence-guided surgery.Fluorescent tracers are being tested in clinical trials to improve detection of tumor margins, but procedures are not standardised. Here, the authors develop an analytical framework that is compatible with the workflow in the operating theatre, and show that it leads to an 88% increase in intraoperative detection of tumor margins in patients with breast cancer.


Clinical and Preclinical Optical Diagnostics | 2017

Fluorescently labeled bevacizumab in human breast cancer: defining the classification threshold

Maximilian Koch; Johannes S. de Jong; Jürgen Glatz; Panagiotis Symvoulidis; Laetitia E. Lamberts; Arthur Adams; Mariëtte E.G. Kranendonk; Anton G.T. Terwisscha van Scheltinga; Michaela Aichler; 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; P. Beatriz Garcia-Allende; Paul J. van Diest; Elisabeth G.E. de Vries; Axel Walch; Gooitzen M. van Dam; Vasilis Ntziachristos

In-vivo fluorescently labelled drug (bevacizumab) breast cancer specimen where obtained from patients. We propose a new structured method to determine the optimal classification threshold in targeted fluorescence intra-operative imaging.


British Journal of Clinical Pharmacology | 2017

Ocular myasthenic syndrome, adverse reaction to omalizumab?: A case report

Willemien S. Kalteren; Marco W.J. Schreurs; Annelies Jorritsma-Smit; Daan Touw; Jan Willem de Jong; Petra G. M. A. Zweers; Fransje Reesink

University of Groningen, University Medical Center Groningen, The Netherlands, Department of Immunology, Erasmus MC, University Medical Center Rotterdam, The Netherlands, Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, The Netherlands, Department of Lung Diseases, Ommelander Hospital Groningen, The Netherlands, Netherlands Pharmacovigilance Centre Lareb, The Netherlands, and Department of Neurology, Ommelander Hospital Groningen, The Netherlands


Gut | 2017

Near-infrared fluorescence molecular endoscopy detects dysplastic oesophageal lesions using topical and systemic tracer of vascular endothelial growth factor A

Wouter B. Nagengast; Elmire Hartmans; Pilar Beatriz Garcia-Allende; Frans Peters; Matthijs D. Linssen; Maximilian Koch; Marjory Koller; Jolien J. Tjalma; Arend Karrenbeld; Annelies Jorritsma-Smit; Jan H. Kleibeuker; Gooitzen M. van Dam; Vasilis Ntziachristos

Collaboration


Dive into the Annelies Jorritsma-Smit's collaboration.

Top Co-Authors

Avatar

Wouter B. Nagengast

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Matthijs D. Linssen

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Gooitzen M. van Dam

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Marjory Koller

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Elisabeth G.E. de Vries

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marjolijn N. Lub-de Hooge

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

Liesbeth Jansen

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge