Network


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

Hotspot


Dive into the research topics where Maarten Neerincx is active.

Publication


Featured researches published by Maarten Neerincx.


Endoscopy | 2010

Colonic work-up after incomplete colonoscopy: significant new findings during follow-up

Maarten Neerincx; J. S. Terhaar sive Droste; C. J. J. Mulder; Mirre Räkers; Joep F. W. M. Bartelsman; Ruud J. Loffeld; Hans Tuynman; R. M. Brohet; R. W. M. Van Der Hulst

BACKGROUND AND STUDY AIMS Cecal intubation is not achieved in 2 - 23 % of colonoscopies. The efforts made by physicians to visualize the remaining colon and the number of missed significant lesions are unknown. This study evaluates 1) the reasons for incomplete colonoscopy, 2) the rates of complete colonic evaluation after incomplete colonoscopy, and 3) the number of (pre-) malignant lesions missed by incomplete colonoscopy. PATIENTS AND METHODS In this population-based cohort study index colonoscopies were performed between September and December 2005. Prospectively collected data from consecutive patients with an incomplete colonoscopy were analyzed. For up to 18 months after the index colonoscopy, any further examinations performed in these patients were identified retrospectively. These secondary examinations included: repeat colonoscopy, computed tomography (CT) colonography, barium enema, abdominal CT scan, and surgery involving the colorectum. RESULTS Of 5278 colonoscopies, 511 were incomplete (9.7 %). The most frequent causes of incomplete colonoscopy were looping of the scope (20.4 %), patient discomfort (15.3 %), and obstructing tumor (13.9 %). Secondary examination was performed in 278 patients (54.4 %) after incomplete colonoscopy. Patients undergoing surveillance after colorectal cancer (CRC) (78.9 %) and those with anemia (73.1 %) most frequently received a secondary examination. Incomplete colonoscopies due to stenosis (78.9 %), severe inflammation (77.8 %) or an obstructing tumor (74.6 %) were most frequently followed by a secondary examination. In all of the follow-up examinations, CRC was diagnosed in 18 patients (3.5 %) and advanced adenoma in four patients (0.8 %). CONCLUSIONS In 4.3 % of the patients, advanced neoplasia was missed by incomplete colonoscopy. Our data therefore suggest that additional imaging is obligatory to visualize the remaining colon adequately.


Cancer Prevention Research | 2012

DNA methylation of phosphatase and actin regulator 3 detects colorectal cancer in stool and complements FIT

Linda J.W. Bosch; Frank A. Oort; Maarten Neerincx; Carolina Khalid-de Bakker; Jochim S. Terhaar sive Droste; Veerle Melotte; Daisy Jonkers; Ad Masclee; Sandra Mongera; Madeleine Grooteclaes; Joost Louwagie; Wim Van Criekinge; Veerle M.H. Coupé; Chris J. Mulder; Manon van Engeland; Beatriz Carvalho; Gerrit A. Meijer

Using a bioinformatics-based strategy, we set out to identify hypermethylated genes that could serve as biomarkers for early detection of colorectal cancer (CRC) in stool. In addition, the complementary value to a Fecal Immunochemical Test (FIT) was evaluated. Candidate genes were selected by applying cluster alignment and computational analysis of promoter regions to microarray-expression data of colorectal adenomas and carcinomas. DNA methylation was measured by quantitative methylation-specific PCR on 34 normal colon mucosa, 71 advanced adenoma, and 64 CRC tissues. The performance as biomarker was tested in whole stool samples from in total 193 subjects, including 19 with advanced adenoma and 66 with CRC. For a large proportion of these series, methylation data for GATA4 and OSMR were available for comparison. The complementary value to FIT was measured in stool subsamples from 92 subjects including 44 with advanced adenoma or CRC. Phosphatase and Actin Regulator 3 (PHACTR3) was identified as a novel hypermethylated gene showing more than 70-fold increased DNA methylation levels in advanced neoplasia compared with normal colon mucosa. In a stool training set, PHACTR3 methylation showed a sensitivity of 55% (95% CI: 33–75) for CRC and a specificity of 95% (95% CI: 87–98). In a stool validation set, sensitivity reached 66% (95% CI: 50–79) for CRC and 32% (95% CI: 14–57) for advanced adenomas at a specificity of 100% (95% CI: 86–100). Adding PHACTR3 methylation to FIT increased sensitivity for CRC up to 15%. PHACTR3 is a new hypermethylated gene in CRC with a good performance in stool DNA testing and has complementary value to FIT. Cancer Prev Res; 5(3); 464–72. ©2011 AACR.


Cancer Epidemiology, Biomarkers & Prevention | 2011

Higher Fecal Immunochemical Test Cutoff Levels: Lower Positivity Rates but Still Acceptable Detection Rates for Early-Stage Colorectal Cancers

J. S. Terhaar sive Droste; Frank A. Oort; R. W. M. Van Der Hulst; H. A. van Heukelem; Ruud J. Loffeld; S. T. van Turenhout; I. Ben Larbi; Shannon L. Kanis; Maarten Neerincx; Mirre Räkers; Veerle M.H. Coupé; Anneke A. Bouman; G. A. Meijer; C. J. J. Mulder

Background: Adjusting the threshold for positivity of quantitative fecal immunochemical tests (FIT) allows for controlling the number of follow-up colonoscopies in a screening program. However, it is unknown to what extent higher cutoff levels affect detection rates of screen-relevant neoplasia. This study aimed to assess the effect of higher cutoff levels of a quantitative FIT on test positivity rate and detection rate of early-stage colorectal cancers (CRC). Methods: Subjects above 40 years old scheduled for colonoscopy in 5 hospitals were asked to sample a single FIT (OC sensor) before colonoscopy. Screen-relevant neoplasia were defined as advanced adenoma or early-stage cancer (stage I and II). Positivity rate, sensitivity, and specificity were evaluated at increasing cutoff levels of 50 to 200 ng/mL. Results: In 2,145 individuals who underwent total colonoscopy, 79 patients were diagnosed with CRC, 38 of which were with early-stage disease. Advanced adenomas were found in 236 patients. When varying cutoff levels from ≥50 to ≥200 ng/mL, positivity rates ranged from 16.5% to 10.2%. With increasing cutoff levels, sensitivity for early-stage CRCs and for screen-relevant neoplasia ranged from 84.2% to 78.9% and 47.1% to 37.2%, respectively. Conclusions: Higher FIT cutoff levels substantially decrease test positivity rates with only limited effects on detection rates of early-stage CRCs. However, spectrum bias resulting in higher estimates of sensitivity than would be expected in a screening population may be present. Impact: Higher cutoff levels can reduce strain on colonoscopy capacity with only a modest decrease in sensitivity for curable cancers. Cancer Epidemiol Biomarkers Prev; 20(2); 272–80. ©2010 AACR.


Oncogenesis | 2015

MiR expression profiles of paired primary colorectal cancer and metastases by next-generation sequencing

Maarten Neerincx; Daud Sie; M.A. van de Wiel; N.C.T. van Grieken; Jan Dirk Burggraaf; Henk L. Dekker; Paul P. Eijk; Bauke Ylstra; Cornelis Verhoef; G. A. Meijer; Tineke E. Buffart; Henk M.W. Verheul

MicroRNAs (miRs) have been recognized as promising biomarkers. It is unknown to what extent tumor-derived miRs are differentially expressed between primary colorectal cancers (pCRCs) and metastatic lesions, and to what extent the expression profiles of tumor tissue differ from the surrounding normal tissue. Next-generation sequencing (NGS) of 220 fresh-frozen samples, including paired primary and metastatic tumor tissue and non-tumorous tissue from 38 patients, revealed expression of 2245 known unique mature miRs and 515 novel candidate miRs. Unsupervised clustering of miR expression profiles of pCRC tissue with paired metastases did not separate the two entities, whereas unsupervised clustering of miR expression profiles of pCRC with normal colorectal mucosa demonstrated complete separation of the tumor samples from their paired normal mucosa. Two hundred and twenty-two miRs differentiated both pCRC and metastases from normal tissue samples (false discovery rate (FDR) <0.05). The highest expressed tumor-specific miRs were miR-21 and miR-92a, both previously described to be involved in CRC with potential as circulating biomarker for early detection. Only eight miRs, 0.5% of the analysed miR transcriptome, were differentially expressed between pCRC and the corresponding metastases (FDR <0.1), consisting of five known miRs (miR-320b, miR-320d, miR-3117, miR-1246 and miR-663b) and three novel candidate miRs (chr 1-2552-5p, chr 8-20656-5p and chr 10-25333-3p). These results indicate that previously unrecognized candidate miRs expressed in advanced CRC were identified using NGS. In addition, miR expression profiles of pCRC and metastatic lesions are highly comparable and may be of similar predictive value for prognosis or response to treatment in patients with advanced CRC.


BMC Bioinformatics | 2014

ShrinkBayes: a versatile R-package for analysis of count-based sequencing data in complex study designs

Mark A. van de Wiel; Maarten Neerincx; Tineke E. Buffart; Daoud Sie; Henk M.W. Verheul

BackgroundComplex designs are common in (observational) clinical studies. Sequencing data for such studies are produced more and more often, implying challenges for the analysis, such as excess of zeros, presence of random effects and multi-parameter inference. Moreover, when sample sizes are small, inference is likely to be too liberal when, in a Bayesian setting, applying a non-appropriate prior or to lack power when not carefully borrowing information across features.ResultsWe show on microRNA sequencing data from a clinical cancer study how our software ShrinkBayes tackles the aforementioned challenges. In addition, we illustrate its comparatively good performance on multi-parameter inference for groups using a data-based simulation. Finally, in the small sample size setting, we demonstrate its high power and improved FDR estimation by use of Gaussian mixture priors that include a point mass.ConclusionShrinkBayes is a versatile software package for the analysis of count-based sequencing data, which is particularly useful for studies with small sample sizes or complex designs.


Gut | 2013

The future of colorectal cancer: implications of screening

Maarten Neerincx; Tineke E. Buffart; Chris J. Mulder; Gerrit A. Meijer; Henk M.W. Verheul

In 2003 the countries of the EU agreed to start screening for colorectal cancer (CRC). Currently, several EU countries have implemented a screening programme. As the result of the detection of prevalent cancers with a biannual faecal immunochemical test (FIT) accompanied by an aging population and an increased risk of developing CRC, the incidence of CRC will rise with 35% by 2020 in the Netherlands.1 ,2 Implementation of a screening programme will lead to a more favourable prognosis at diagnosis through the detection of CRC at an early, asymptomatic stage. Of all cancers detected with a single guaiac faecal occult blood test (GFOBT) or FIT, 64–71% will be at stage I or II, compared with 45–60% in matched non-invited symptomatic populations, with FIT showing superiority over GFOBT.3 ,4 The introduction of CRC screening will detect on average 1600 additional stage I and II CRCs per year in the first few years after its introduction in the Netherlands.4 ,5 Interval cancers will still be diagnosed in between screening rounds because not all cancers will be detected when asymptomatic.6 The stage distribution of these interval cancers will mimic those diagnosed in a symptomatic non-screened population.3 ,7 When taking these interval cancers into account, computational models reveal an increase in the proportion of CRCs diagnosed at stages I and II from 53% to 80% without and with annual FIT screening, respectively. Accordingly, a decrease in the proportion of CRCs diagnosed at stages III or IV from 47% to 20% with screening has been predicted. Identical shifts in stage distribution have been observed for all investigated screening strategies.8 Although there might be a selection bias towards an unfavourable stage distribution in the anticipated 40% of the invited population not attending CRC screening, preliminary studies …


Journal of Clinical Oncology | 2018

Loss of Chromosome 18q11.2-q12.1 Is Predictive for Survival in Patients With Metastatic Colorectal Cancer Treated With Bevacizumab

Erik van Dijk; Hedde D. Biesma; Martijn Cordes; Dominiek Smeets; Maarten Neerincx; Sudipto Das; Paul P. Eijk; Verena Murphy; Anna Barat; Orna Bacon; Jochen H. M. Prehn; Johannes Betge; Timo Gaiser; Bozena Fender; Gerrit A. Meijer; Deborah A. McNamara; Rut Klinger; Miriam Koopman; Matthias Ebert; Elaine Kay; Bryan T. Hennessey; Henk M.W. Verheul; William M. Gallagher; Darran P. O’Connor; Cornelis J. A. Punt; Fotios Loupakis; Diether Lambrechts; Annette T. Byrne; Nicole C.T. van Grieken; Bauke Ylstra

Purpose Patients with metastatic colorectal cancer (mCRC) have limited benefit from the addition of bevacizumab to standard chemotherapy. However, a subset probably benefits substantially, highlighting an unmet clinical need for a biomarker of response to bevacizumab. Previously, we demonstrated that losses of chromosomes 5q34, 17q12, and 18q11.2-q12.1 had a significant correlation with progression-free survival (PFS) in patients with mCRC treated with bevacizumab in the CAIRO2 clinical trial but not in patients who did not receive bevacizumab in the CAIRO trial. This study was designed to validate these findings. Materials and Methods Primary mCRC samples were analyzed from two cohorts of patients who received bevacizumab as first-line treatment; 96 samples from the European multicenter study Angiopredict (APD) and 81 samples from the Italian multicenter study, MOMA. A third cohort of 90 samples from patients with mCRC who did not receive bevacizumab was analyzed. Copy number aberrations of tumor biopsy specimens were measured by shallow whole-genome sequencing and were correlated with PFS, overall survival (OS), and response. Results Loss of chromosome 18q11.2-q12.1 was associated with prolonged PFS most significantly in both the cohorts that received bevacizumab (APD: hazard ratio, 0.54; P = .01; PFS difference, 65 days; MOMA: hazard ratio, 0.55; P = .019; PFS difference, 49 days). A similar association was found for OS and overall response rate in these two cohorts, which became significant when combined with the CAIRO2 cohort. Median PFS in the cohort of patients with mCRC who did not receive bevacizumab and in the CAIRO cohort was similar to that of the APD, MOMA, and CAIRO2 patients without an 18q11.2-q12.1 loss. Conclusion We conclude that the loss of chromosome 18q11.2-q12.1 is consistently predictive for prolonged PFS in patients receiving bevacizumab. The predictive value of this loss is substantiated by a significant gain in OS and overall response rate.


Digestion | 2016

Outcome of Colorectal Cancer Patients Treated with Combination Bevacizumab Therapy: A Pooled Retrospective Analysis of Three European Cohorts from the Angiopredict Initiative

Johannes Betge; Ana Barat; Verena Murphy; Thomas Hielscher; Nicole C.T. van Grieken; Sebastian Belle; T Zhan; Nicolai Härtel; Melanie Kripp; Orna Bacon; Martijn Cordes; Elaine Kay; Henk M.W. Verheul; Maarten Neerincx; Bryan T. Hennessy; Ralf Hofheinz; Timo Gaiser; Bauke Ylstra; Jochen H. M. Prehn; Diether Lambrechts; Annette T. Byrne; Matthias P.A. Ebert; Nadine Schulte

Background/Aims: This study is aimed at analyzing the survival rates and prognostic factors of stage IV colorectal cancer patients from 3 European cohorts undergoing combination chemotherapy with bevacizumab. Methods: Progression free-survival (PFS) and overall survival (OS) were analyzed in 172 patients using the Kaplan-Meier method and uni- and multivariable Cox proportional hazards regression models. Results: The median PFS was 9.7 and the median OS 27.4 months. Patients treated at centers in Germany (n = 97), Ireland (n = 32), and The Netherlands (n = 43) showed a median PFS of 9.9, 9.2, and 9.7 months, OS of 34.0, 20.5, and 25.1 months, respectively. Patients >65 years had a significantly shorter PFS (9.5 vs. 9.8 months) but not OS (27.4 vs. 27.5 months) than younger patients. High tumor grade (G3/4) was associated with a shorter PFS, T4 classification with both shorter PFS and OS. Fluoropyrimidine (FP) chemotherapy backbones (doublets and single) had comparable outcomes, while patients not receiving FP backbones had a shorter PFS. In multivariable analysis, age and non-FP backbone were associated with inferior PFS, T4 classification and therapy line >2nd were significantly associated with poor PFS and OS. Conclusion: The observed survival rates confirm previous studies and demonstrate reproducible benefits of combination bevacizumab regimens. Classification T4, non-FP chemotherapy backbone, and age >65 were associated with inferior outcome.


PLOS ONE | 2018

Combination of a six microRNA expression profile with four clinicopathological factors for response prediction of systemic treatment in patients with advanced colorectal cancer

Maarten Neerincx; Dennis Poel; Daoud Sie; Nicole C.T. van Grieken; Ram C. Shankaraiah; Floor S.W. Van Der Wolf-De Lijster; Jan Hein T.M. van Waesberghe; Jan Dirk Burggraaf; Paul P. Eijk; Cornelis Verhoef; Bauke Ylstra; Gerrit A. Meijer; Mark A. van de Wiel; Tineke E. Buffart; Henk M.W. Verheul

Background First line chemotherapy is effective in 75 to 80% of patients with metastatic colorectal cancer (mCRC). We studied whether microRNA (miR) expression profiles can predict treatment outcome for first line fluoropyrimidine containing systemic therapy in patients with mCRC. Methods MiR expression levels were determined by next generation sequencing from snap frozen tumor samples of 88 patients with mCRC. Predictive miRs were selected with penalized logistic regression and posterior forward selection. The prediction co-efficients of the miRs were re-estimated and validated by real-time quantitative PCR in an independent cohort of 81 patients with mCRC. Results Expression levels of miR-17-5p, miR-20a-5p, miR-30a-5p, miR-92a-3p, miR-92b-3p and miR-98-5p in combination with age, tumor differentiation, adjuvant therapy and type of systemic treatment, were predictive for clinical benefit in the training cohort with an AUC of 0.78. In the validation cohort the addition of the six miR signature to the four clinicopathological factors demonstrated a significant increased AUC for predicting treatment response versus those with stable disease (SD) from 0.79 to 0.90. The increase for predicting treatment response versus progressive disease (PD) and for patients with SD versus those with PD was not significant. in the validation cohort. MiR-17-5p, miR-20a-5p and miR-92a-3p were significantly upregulated in patients with treatment response in both the training and validation cohorts. Conclusion A six miR expression signature was identified that predicted treatment response to fluoropyrimidine containing first line systemic treatment in patients with mCRC when combined with four clinicopathological factors. Independent validation demonstrated added predictive value of this miR-signature for predicting treatment response versus SD. However, added predicted value for separating patients with PD could not be validated. The clinical relevance of the identified miRs for predicting treatment response has to be further explored.


European Journal of Cancer | 2016

Chromosome 18q11.2 loss as a predictive marker for response to bevacizumab in metastatic colorectal cancer

Bauke Ylstra; Martijn Cordes; Hedde D. Biesma; Henk M.W. Verheul; Maarten Neerincx; Miriam Koopman; C.J.A. Punt; G. A. Meijer; Verena Murphy; Anna Barat; Johannes Betge; Matthias P. Ebert; Timo Gaiser; Bozena Fender; Rut Klinger; Sudipto Das; Dominiek Smeets; Diether Lambrechts; Annette T. Byrne; N.C.T. van Grieken

28 EORTC – NCI – AACR Symposium on Molecular Targets and Cancer Therapeutics 29 November 2016 - 02 December 2016

Collaboration


Dive into the Maarten Neerincx's collaboration.

Top Co-Authors

Avatar

Gerrit A. Meijer

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Henk M.W. Verheul

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Bauke Ylstra

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chris J. Mulder

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Henk L. Dekker

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Tineke E. Buffart

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

G. A. Meijer

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Martijn Cordes

VU University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge