Network


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

Hotspot


Dive into the research topics where Martin C. Boonstra is active.

Publication


Featured researches published by Martin C. Boonstra.


Circulation Research | 2014

Inhibition of 14q32 MicroRNAs miR-329, miR-487b, miR-494, and miR-495 Increases Neovascularization and Blood Flow Recovery After Ischemia

Sabine M.J. Welten; A.J.N.M. Bastiaansen; Rob C. M. de Jong; Margreet R. de Vries; Erna Peters; Martin C. Boonstra; Søren Paludan Sheikh; Nicola La Monica; Ekambar R. Kandimalla; Paul H.A. Quax; A. Yaël Nossent

Rationale: Effective neovascularization is crucial for recovery after cardiovascular events. Objective: Because microRNAs regulate expression of up to several hundred target genes, we set out to identify microRNAs that target genes in all pathways of the multifactorial neovascularization process. Using www.targetscan.org, we performed a reverse target prediction analysis on a set of 197 genes involved in neovascularization. We found enrichment of binding sites for 27 microRNAs in a single microRNA gene cluster. Microarray analyses showed upregulation of 14q32 microRNAs during neovascularization in mice after single femoral artery ligation. Methods and Results: Gene silencing oligonucleotides (GSOs) were used to inhibit 4 14q32 microRNAs, miR-329, miR-487b, miR-494, and miR-495, 1 day before double femoral artery ligation. Blood flow recovery was followed by laser Doppler perfusion imaging. All 4 GSOs clearly improved blood flow recovery after ischemia. Mice treated with GSO-495 or GSO-329 showed increased perfusion already after 3 days (30% perfusion versus 15% in control), and those treated with GSO-329 showed a full recovery of perfusion after 7 days (versus 60% in control). Increased collateral artery diameters (arteriogenesis) were observed in adductor muscles of GSO-treated mice, as well as increased capillary densities (angiogenesis) in the ischemic soleus muscle. In vitro, treatment with GSOs led to increased sprout formation and increased arterial endothelial cell proliferation, as well as to increased arterial myofibroblast proliferation. Conclusions: The 14q32 microRNA gene cluster is highly involved in neovascularization. Inhibition of 14q32 microRNAs miR-329, miR-487b, miR-494, and miR-495 provides a promising tool for future therapeutic neovascularization.


Ejso | 2014

Real-Time Intraoperative Detection of Breast Cancer using Near-infrared Fluorescence Imaging and Methylene Blue

Quirijn R.J.G. Tummers; F.P.R. Verbeek; Boudewijn E. Schaafsma; Martin C. Boonstra; J.R. van der Vorst; G.J. Liefers; C.J.H. van de Velde; John V. Frangioni; Alexander L. Vahrmeijer

BACKGROUND Despite recent developments in preoperative breast cancer imaging, intraoperative localization of tumor tissue can be challenging, resulting in tumor-positive resection margins during breast conserving surgery. Based on certain physicochemical similarities between Technetium((99m)Tc)-sestamibi (MIBI), an SPECT radiodiagnostic with a sensitivity of 83-90% to detect breast cancer preoperatively, and the near-infrared (NIR) fluorophore Methylene Blue (MB), we hypothesized that MB might detect breast cancer intraoperatively using NIR fluorescence imaging. METHODS Twenty-four patients with breast cancer, planned for surgical resection, were included. Patients were divided in 2 administration groups, which differed with respect to the timing of MB administration. N = 12 patients per group were administered 1.0 mg/kg MB intravenously either immediately or 3 h before surgery. The mini-FLARE imaging system was used to identify the NIR fluorescent signal during surgery and on post-resected specimens transferred to the pathology department. Results were confirmed by NIR fluorescence microscopy. RESULTS 20/24 (83%) of breast tumors (carcinoma in N = 21 and ductal carcinoma in situ in N = 3) were identified in the resected specimen using NIR fluorescence imaging. Patients with non-detectable tumors were significantly older. No significant relation to receptor status or tumor grade was seen. Overall tumor-to-background ratio (TBR) was 2.4 ± 0.8. There was no significant difference between TBR and background signal between administration groups. In 2/4 patients with positive resection margins, breast cancer tissue identified in the wound bed during surgery would have changed surgical management. Histology confirmed the concordance of fluorescence signal and tumor tissue. CONCLUSIONS This feasibility study demonstrated an overall breast cancer identification rate using MB of 83%, with real-time intraoperative guidance having the potential to alter patient management.


International Journal of Cancer | 2014

Intraoperative fluorescence delineation of head and neck cancer with a fluorescent Anti-epidermal growth factor receptor nanobody

P. B. A. A. van Driel; J.R. van der Vorst; F.P.R. Verbeek; Sabrina Oliveira; Thomas J. A. Snoeks; Stijn Keereweer; B. Chan; Martin C. Boonstra; John V. Frangioni; P.M.P. van Bergen en Henegouwen; Alexander L. Vahrmeijer; Clemens W.G.M. Löwik

Intraoperative near‐infrared (NIR) fluorescence imaging is a technology with high potential to provide the surgeon with real‐time visualization of tumors during surgery. Our study explores the feasibility for clinical translation of an epidermal growth factor receptor (EGFR)‐targeting nanobody for intraoperative imaging and resection of orthotopic tongue tumors and cervical lymph node metastases. The anti‐EGFR nanobody 7D12 and the negative control nanobody R2 were conjugated to the NIR fluorophore IRDye800CW (7D12‐800CW and R2‐800CW). Orthotopic tongue tumors were induced in nude mice using the OSC‐19‐luc2‐cGFP cell line. Tumor‐bearing mice were injected with 25 µg 7D12‐800CW, R2–800CW or 11 µg 800CW. Subsequently, other mice were injected with 50 or 75 µg of 7D12‐800CW. The FLARE imaging system and the IVIS spectrum were used to identify, delineate and resect the primary tumor and cervical lymph node metastases. All tumors could be clearly identified using 7D12‐800CW. A significantly higher tumor‐to‐background ratio (TBR) was observed in mice injected with 7D12–800CW compared to mice injected with R2‐800CW and 800CW. The highest average TBR (2.00 ± 0.34 and 2.72 ± 0.17 for FLARE and IVIS spectrum, respectively) was observed 24 hr after administration of the EGFR‐specific nanobody. After injection of 75 µg 7D12‐800CW cervical lymph node metastases could be clearly detected. Orthotopic tongue tumors and cervical lymph node metastases in a mouse model were clearly identified intraoperatively using a recently developed fluorescent EGFR‐targeting nanobody. Translation of this approach to the clinic would potentially improve the rate of radical surgical resections.


Current Pharmaceutical Design | 2011

Clinical Applications of the Urokinase Receptor (uPAR) for Cancer Patients

Martin C. Boonstra; Hein W. Verspaget; S. Ganesh; F.J.G.M. Kubben; Alexander L. Vahrmeijer; Cornelis J. H. van de Velde; Peter J. K. Kuppen; Paul H.A. Quax; Cornelis F. M. Sier

Since decades the urokinase plasminogen activator (uPA) system has been associated with the invasion of malignant cells. The receptor of urokinase (uPAR) is one of the key players in this proteolytic cascade, because it focuses uPAs proteolytic activity to the cell surface and in addition functions as a signaling receptor. uPAR is highly expressed in virtually all human cancers, suggesting possible clinical applications as diagnostic marker, predictive tool of survival or clinical response, and as a target for therapy and imaging. This review summarizes the possibilities of uPAR in clinical applications for cancer patients.


Journal of Controlled Release | 2016

EGFR targeted nanobody–photosensitizer conjugates for photodynamic therapy in a pre-clinical model of head and neck cancer

Pieter B. A. A. Van Driel; Martin C. Boonstra; Maxime D. Slooter; Raimond Heukers; Marieke A. Stammes; Thomas J. A. Snoeks; Henriëtte S. de Bruijn; Paul J. van Diest; Alexander L. Vahrmeijer; Paul M.P. van Bergen en Henegouwen; Cornelis J. H. van de Velde; Clemens W.G.M. Löwik; Dominic J. Robinson; Sabrina Oliveira

Photodynamic therapy (PDT) induces cell death through local light activation of a photosensitizer (PS) and has been used to treat head and neck cancers. Yet, common PS lack tumor specificity, which leads to collateral damage to normal tissues. Targeted delivery of PS via antibodies has pre-clinically improved tumor selectivity. However, antibodies have long half-lives and relatively poor tissue penetration, which could limit therapeutic efficacy and lead to long photosensitivity. Here, in this feasibility study, we evaluate at the pre-clinical level a recently introduced format of targeted PDT, which employs nanobodies as targeting agents and a water-soluble PS (IRDye700DX) that is traceable through optical imaging. In vitro, the PS solely binds to cells and induces phototoxicity on cells overexpressing the epidermal growth factor receptor (EGFR), when conjugated to the EGFR targeted nanobodies. To investigate whether this new format of targeted PDT is capable of inducing selective tumor cell death in vivo, PDT was applied on an orthotopic mouse tumor model with illumination at 1 h post-injection of the nanobody–PS conjugates, as selected from quantitative fluorescence spectroscopy measurements. In parallel, and as a reference, PDT was applied with an antibody–PS conjugate, with illumination performed 24 h post-injection. Importantly, EGFR targeted nanobody–PS conjugates led to extensive tumor necrosis (approx. 90%) and almost no toxicity in healthy tissues, as observed through histology 24 h after PDT. Overall, results show that these EGFR targeted nanobody–PS conjugates are selective and able to induce tumor cell death in vivo. Additional studies are now needed to assess the full potential of this approach to improving PDT.


BMC Cancer | 2014

Expression of uPAR in tumor-associated stromal cells is associated with colorectal cancer patient prognosis: a TMA study

Martin C. Boonstra; F.P.R. Verbeek; Andrew P. Mazar; Hendrica A.J.M. Prevoo; Peter J. K. Kuppen; Cornelis J. H. van de Velde; Alexander L. Vahrmeijer; Cornelis F. M. Sier

BackgroundThe receptor for urokinase-type plasminogen activator (uPAR) is associated with cancer development and progression. Within the tumor microenvironment uPAR is expressed by malignant cells as well as tumor-associated stromal cells. However, the contribution of uPAR expression in these stromal cells to malignancy and patient survival in colorectal cancer is still unclear. This study compares the association of uPAR expression in both colorectal tumor-associated stromal cells and neoplastic cells with clinico-pathological characteristics and patient survival using tissue micro arrays (TMA).MethodsImmunohistochemical staining of uPAR expression was performed on tumor tissue from 262 colorectal cancer patients. Kaplan-Meier, log rank, and uni- and multivariate Cox’s regression analyses were used to calculate associations between uPAR expression and patient survival.ResultsIn the colorectal tumor-associated stromal microenvironment, uPAR is expressed in macrophages, (neoangiogenic) endothelial cells and myofibroblasts. uPAR expression in tumor-associated stromal cells and neoplastic cells (and both combined) were negatively associated with overall survival (OS) and Disease Free Survival (DFS). Uni- and multivariate Cox’s regression analysis for combined uPAR expression in tumor-associated stromal and neoplastic cells showed significant and independent negative associations with OS and DFS. Only uPAR expression in tumor-associated stromal cells showed independent significance in the uni- and multivariate analysis for DFS.ConclusionThis study demonstrates a significant independent negative association between colorectal cancer patient survival and uPAR expression in especially tumor-associated stromal cells.


International Journal of Cancer | 2015

Preclinical evaluation of a novel CEA-targeting near-infrared fluorescent tracer delineating colorectal and pancreatic tumors

Martin C. Boonstra; Berend Tolner; Boudewijn E. Schaafsma; Leonora S.F. Boogerd; Hendrica A.J.M. Prevoo; Guarav Bhavsar; Peter J. K. Kuppen; Cornelis F. M. Sier; Bert A. Bonsing; John V. Frangioni; Cornelis J. H. van de Velde; Kerry A. Chester; Alexander L. Vahrmeijer

Surgery is the cornerstone of oncologic therapy with curative intent. However, identification of tumor cells in the resection margins is difficult, resulting in nonradical resections, increased cancer recurrence and subsequent decreased patient survival. Novel imaging techniques that aid in demarcating tumor margins during surgery are needed. Overexpression of carcinoembryonic antigen (CEA) is found in the majority of gastrointestinal carcinomas, including colorectal and pancreas. We developed ssSM3E/800CW, a novel CEA‐targeted near‐infrared fluorescent (NIRF) tracer, based on a disulfide‐stabilized single‐chain antibody fragment (ssScFv), to visualize colorectal and pancreatic tumors in a clinically translatable setting. The applicability of the tracer was tested for cell and tissue binding characteristics and dosing using immunohistochemistry, flow cytometry, cell‐based plate assays and orthotopic colorectal (HT‐29, well differentiated) and pancreatic (BXPC‐3, poorly differentiated) xenogeneic human–mouse models. NIRF signals were visualized using the clinically compatible FLARE™ imaging system. Calculated clinically relevant doses of ssSM3E/800CW selectively accumulated in colorectal and pancreatic tumors/cells, with highest tumor‐to‐background ratios of 5.1 ± 0.6 at 72 hr postinjection, which proved suitable for intraoperative detection and delineation of tumor boarders and small (residual) tumor nodules in mice, between 8 and 96 hr postinjection. Ex vivo fluorescence imaging and pathologic examination confirmed tumor specificity and the distribution of the tracer. Our results indicate that ssSM3E/800CW shows promise as a diagnostic tool to recognize colorectal and pancreatic cancers for fluorescent‐guided surgery applications. If successfully translated clinically, this tracer could help improve the completeness of surgery and thus survival.


Molecular Imaging and Biology | 2015

Characterization and evaluation of the artemis camera for fluorescence-guided cancer surgery.

P. B. A. A. van Driel; M. van de Giessen; Martin C. Boonstra; Thomas J. A. Snoeks; Stijn Keereweer; Sabrina Oliveira; C.J.H. van de Velde; Boudewijn P. F. Lelieveldt; Alexander L. Vahrmeijer; Clemens W.G.M. Löwik; Jouke Dijkstra

PurposeNear-infrared (NIR) fluorescence imaging can provide the surgeon with real-time visualization of, e.g., tumor margins and lymph nodes. We describe and evaluate the Artemis, a novel, handheld NIR fluorescence camera.ProceduresWe evaluated minimal detectable cell numbers (FaDu-luc2, 7D12-IRDye 800CW), preclinical intraoperative detection of sentinel lymph nodes (SLN) using indocyanine green (ICG), and of orthotopic tongue tumors using 7D12-800CW. Results were compared with the Pearl imager. Clinically, three patients with liver metastases were imaged using ICG.ResultsMinimum detectable cell counts for Artemis and Pearl were 2 × 105 and 4 × 104 cells, respectively. In vivo, seven SLNs were detected in four mice with both cameras. Orthotopic OSC-19-luc2-cGFP tongue tumors were clearly identifiable, and a minimum FaDu-luc2 tumor size of 1 mm3 could be identified. Six human malignant lesions were identified during three liver surgery procedures.ConclusionsBased on this study, the Artemis system has demonstrated its utility in fluorescence-guided cancer surgery.


World Journal of Gastroenterology | 2016

Near-infrared fluorescence sentinel lymph node detection in gastric cancer: A pilot study.

Quirijn R.J.G. Tummers; Leonora S.F. Boogerd; Wobbe O. de Steur; F.P.R. Verbeek; Martin C. Boonstra; Henricus J.M. Handgraaf; John V. Frangioni; Cornelis J. H. van de Velde; Henk H. Hartgrink; Alexander L. Vahrmeijer

AIM To investigate feasibility and accuracy of near-infrared fluorescence imaging using indocyanine green: nanocolloid for sentinel lymph node (SLN) detection in gastric cancer. METHODS A prospective, single-institution, phase I feasibility trial was conducted. Patients suffering from gastric cancer and planned for gastrectomy were included. During surgery, a subserosal injection of 1.6 mL ICG:Nanocoll was administered around the tumor. NIR fluorescence imaging of the abdominal cavity was performed using the Mini-FLARE™ NIR fluorescence imaging system. Lymphatic pathways and SLNs were visualized. Of every detected SLN, the corresponding lymph node station, signal-to-background ratio and histopathological diagnosis was determined. Patients underwent standard-of-care gastrectomy. Detected SLNs outside the standard dissection planes were also resected and evaluated. RESULTS Twenty-six patients were enrolled. Four patients were excluded because distant metastases were found during surgery or due to technical failure of the injection. In 21 of the remaining 22 patients, at least 1 SLN was detected by NIR Fluorescence imaging (mean 3.1 SLNs; range 1-6). In 8 of the 21 patients, tumor-positive LNs were found. Overall accuracy of the technique was 90% (70%-99%; 95%CI), which decreased by higher pT-stage (100%, 100%, 100%, 90%, 0% for respectively Tx, T1, T2, T3, T4 tumors). All NIR-negative SLNs were completely effaced by tumor. Mean fluorescence signal-to-background ratio of SLNs was 4.4 (range 1.4-19.8). In 8 of the 21 patients, SLNs outside the standard resection plane were identified, that contained malignant cells in 2 patients. CONCLUSION This study shows successful use of ICG:Nanocoll as lymphatic tracer for SLN detection in gastric cancer. Moreover, tumor-containing LNs outside the standard dissection planes were identified.


Oncotarget | 2016

Concordance of folate receptor-α expression between biopsy, primary tumor and metastasis in breast cancer and lung cancer patients

Leonora S.F. Boogerd; Martin C. Boonstra; Ann Jean Beck; Ayoub Charehbili; Charlotte E.S. Hoogstins; Hendrica A.J.M. Prevoo; Sunil Singhal; Philip S. Low; Cornelis J. H. van de Velde; Alexander L. Vahrmeijer

Folate receptor alpha (FRα) is known to be upregulated in a variety of cancers, including non-small cell lung cancer (NSCLC) and breast cancer. To ensure reliable implementation of diagnostic- and therapeutic agents, concordance of FRα expression between biopsy, primary tumor and metastases is important. Using immunohistochemistry (Mab 26B3.F2) these concordances were investigated in 60 NSCLC and 40 breast cancer patients. False positivity of FRα expression on breast and lung cancer biopsies was limited to less than 5%. In NSCLC, FRα expression was shown in 21/34 adenocarcinomas and 4/26 squamous cell carcinomas (SCC). Concordance of FRα expression between biopsy and primary tumor was achieved in respectively 83% and 91% of adenocarcinomas and SCCs. Approximately 80% of all local and distant metastases of NSCLC patients showed concordant FRα expression as their corresponding primary tumor. In breast cancer, FRα positivity was shown in 12/40 biopsies, 20/40 lumpectomies and 6/20 LN metastases, with concordance of 68% between biopsy and primary tumor and 60% between primary tumor and LN metastases. In conclusion, this study shows high concordance rates of FRα expression between biopsies and metastases compared to primary NSCLC and breast cancers, underscoring the applicability of FRα-targeted agents in these patients.

Collaboration


Dive into the Martin C. Boonstra's collaboration.

Top Co-Authors

Avatar

Alexander L. Vahrmeijer

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cornelis F. M. Sier

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hendrica A.J.M. Prevoo

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peter J. K. Kuppen

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Quirijn R.J.G. Tummers

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

John V. Frangioni

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

C.J.H. van de Velde

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Leonora S.F. Boogerd

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Bert A. Bonsing

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge