Network


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

Hotspot


Dive into the research topics where Remond J.A. Fijneman is active.

Publication


Featured researches published by Remond J.A. Fijneman.


Journal of Proteomics | 2010

Proteomics of colorectal cancer: Overview of discovery studies and identification of commonly identified cancer-associated proteins and candidate CRC serum markers

Connie R. Jimenez; Jaco C. Knol; Gerrit A. Meijer; Remond J.A. Fijneman

Colorectal cancer (CRC) is a common cause of cancer-related mortality in the developed world. Improved methods for early detection and disease management are urgently needed. Many efforts in the past 5 years have been devoted to protein biomarker discovery for early detection of CRC. Here, we discuss identity-based studies employing tandem mass spectrometry that analyzed clinical material as well as model systems. Through meta-analysis we provide a list of CRC-associated tissue proteins discovered in multiple studies, with the greater majority being 2D gel-based discoveries coupled to MS/MS. So far only a limited number of CRC-associated proteins have been validated in serum for non-invasive testing for CRC. This list includes several intracellular and nuclear proteins that a priori would not have been considered candidate biomarkers based on their predicted subcellular localization. Finally, we highlight promising new directions that combine targeted analyses of subcellular proteomes, like the cell surface, secretome, exosome, and nuclear matrix, with nanoLC-MS/MS-based proteomics. We anticipate that in the near future, these novel mass spectrometry-based in-depth approaches will uncover many novel, specific CRC marker candidates in clinical tissues and that their targeted validation with multi-reaction monitoring MS will speed up development of non-invasive tests in feces and serum/plasma.


Clinical Colorectal Cancer | 2011

Molecular Tests for Colorectal Cancer Screening

Linda J.W. Bosch; Beatriz Carvalho; Remond J.A. Fijneman; Connie R. Jimenez; Herbert M. Pinedo; Manon van Engeland; Gerrit A. Meijer

Detecting and removing high-risk adenomas and early colorectal cancer (CRC) can reduce mortality of this disease. The noninvasive fecal occult blood test (FOBT; guaiac-based or immunochemical) is widely used in screening programs and although effective, it leaves room for improvement in terms of test accuracy. Molecular tests are expected to be more sensitive, specific and informative than current detection tests, and are promising future tools for CRC screening. This review provides an overview of the performances of DNA, RNA, and protein markers for CRC detection in stool and blood. Most emphasis currently is on DNA and protein markers. Among DNA markers there is trend to move away from mutation markers in favor of methylation markers. The recent boost in proteomics research leads to many new candidate protein markers. Usually in small series, some markers show better performance than the present FOBT. Evaluation in large well-controlled randomized trials is the next step needed to take molecular markers for CRC screening to the next level and warrant implementation in a screening setting.


European Journal of Cancer | 2011

Loss of lamin A/C expression in stage II and III colon cancer is associated with disease recurrence

E.J.Th. Belt; Remond J.A. Fijneman; E.G. van den Berg; Herman Bril; P.M. Delis-van Diemen; Marianne Tijssen; H.F. van Essen; E.S.M. de Lange-de Klerk; Jeroen A.M. Beliën; Hein B.A.C. Stockmann; S. Meijer; Gerrit A. Meijer

AIM OF THE STUDY Loss of the nuclear lamina protein lamin A/C (LMNA) has been observed in several human malignancies. The present study aimed to investigate associations between LMNA expression and clinical outcome in colon cancer patients. PATIENTS AND METHODS Clinicopathological data and formalin-fixed paraffin embedded tissues were collected from 370 stage II and III colon cancer patients. Tissue microarrays were constructed, stained for lamin A/C and evaluated microscopically. Microsatellite instability status was determined for 318 tumours. RESULTS Low levels of LMNA expression were observed in 17.8% of colon tumours, with disease recurrence occurring in 45.5% of stage II and III colon cancer patients with LMNA-low expressing tumours compared to 29.6% of patients with LMNA-high expressing tumours (p=0.01). For stage II patients, disease recurrence was observed for 35.7% of LMNA-low compared to 20.3% of LMNA-high expressing tumours (p=0.03). Microsatellite stable (MSS) tumours exhibited more frequently low LMNA expression than microsatellite instable (MSI) tumours (21% versus 9.8%; p=0.05). Interestingly, disease recurrence among LMNA-low and LMNA-high expressing MSS tumours varied significantly for stage III patients who had not received adjuvant chemotherapy (100% versus 37.8%; p<0.01) while no such difference was observed for patients who received adjuvant chemotherapy (46.7% versus 46.0%; p=0.96). CONCLUSION These data indicate that low expression of LMNA is associated with an increased disease recurrence in stage II and III colon cancer patients, and suggest that these patients in particular may benefit from adjuvant chemotherapy.


Science Translational Medicine | 2017

Direct detection of early-stage cancers using circulating tumor DNA

Jillian Phallen; Mark Sausen; Vilmos Adleff; Alessandro Leal; Carolyn Hruban; James White; Valsamo Anagnostou; Jacob Fiksel; Stephen Cristiano; Eniko Papp; Savannah Speir; Thomas Reinert; Mai-Britt Worm Ørntoft; Brian Woodward; Derek Murphy; Sonya Parpart-Li; David Riley; Monica Nesselbush; Naomi Sengamalay; Andrew Georgiadis; Qing Kay Li; Mogens Rørbæk Madsen; Frank Viborg Mortensen; Joost Huiskens; Cornelis J. A. Punt; Nicole C.T. van Grieken; Remond J.A. Fijneman; G. A. Meijer; Hatim Husain; Robert B. Scharpf

Noninvasive liquid biopsy analysis of circulating tumor DNA permits direct detection of early-stage cancers. Finding smaller needles in haystacks The detection and analysis of cell-free DNA in patients’ blood are becoming increasingly accepted in oncology. However, this approach has generally been applied for the monitoring of patients with existing tumors. It has not been useful for early diagnosis of cancer because of insufficient sensitivity to detect really small tumors that only shed minute quantities of DNA into the blood, as well as difficulties with identifying cancer-associated genetic changes without knowing what mutations are present in the primary tumor. A method developed by Phallen et al., called targeted error correction sequencing, addresses both of these limitations and demonstrates the feasibility of detecting circulating cell-free DNA from many early tumors, suggesting its potential use for cancer screening. Early detection and intervention are likely to be the most effective means for reducing morbidity and mortality of human cancer. However, development of methods for noninvasive detection of early-stage tumors has remained a challenge. We have developed an approach called targeted error correction sequencing (TEC-Seq) that allows ultrasensitive direct evaluation of sequence changes in circulating cell-free DNA using massively parallel sequencing. We have used this approach to examine 58 cancer-related genes encompassing 81 kb. Analysis of plasma from 44 healthy individuals identified genomic changes related to clonal hematopoiesis in 16% of asymptomatic individuals but no alterations in driver genes related to solid cancers. Evaluation of 200 patients with colorectal, breast, lung, or ovarian cancer detected somatic mutations in the plasma of 71, 59, 59, and 68%, respectively, of patients with stage I or II disease. Analyses of mutations in the circulation revealed high concordance with alterations in the tumors of these patients. In patients with resectable colorectal cancers, higher amounts of preoperative circulating tumor DNA were associated with disease recurrence and decreased overall survival. These analyses provide a broadly applicable approach for noninvasive detection of early-stage tumors that may be useful for screening and management of patients with cancer.


Clinical Biochemistry | 2013

Proteomics in colorectal cancer translational research: biomarker discovery for clinical applications.

Meike de Wit; Remond J.A. Fijneman; Henk M.W. Verheul; Gerrit A. Meijer; Connie R. Jimenez

Colorectal cancer (CRC) is a major cause of cancer-related death in the western world. Screening to detect the disease in an early stage is the most effective approach to tackle this problem. In addition, better diagnostic tools for assessment of prognosis and prediction of response to drug therapy will allow for personalized therapies and better outcomes. Protein biomarkers that reflect tumor biology have the potential to address a wide range of clinical needs. These include diagnostic (screening) biomarkers for early detection, prognostic biomarkers for estimation of disease outcome, predictive biomarkers for adjuvant treatment stratification, and surveillance biomarkers for disease monitoring and treatment response. An important source for the discovery of potential biomarkers comes from mass spectrometry based proteomics research of the biology of CRC development. Here, we review recent colon cancer proteomics studies directed at identification of biomarker proteins. These include studies that use preclinical models (i.e. cell lines or murine tissues) as well as clinical materials (e.g. tissue and stool samples). We separately highlight some studies that focused on identification of cancer stem cell (CSC) related proteins in tumor spheroids, an in vitro model system for investigating CRC treatment response. Recent proteomics studies have generated many new candidate protein biomarkers. However, the lack of follow-up studies that lead to biomarker verification and/or validation remains a limiting factor in the translation of these candidate biomarkers into clinical applications. This is partly due to technological limitations which are bound to diminish with new technologies, including selected reaction monitoring mass spectrometry (SRM-MS). Antibodies are still required, though, both to perform high-throughput validation as well as to develop cost-effective tests for routine use in a clinical setting.


Gut | 2012

TPX2 and AURKA promote 20q amplicon-driven colorectal adenoma to carcinoma progression

Anke H. Sillars-Hardebol; Beatriz Carvalho; Marianne Tijssen; Jeroen A.M. Beliën; Meike de Wit; Pien M. Delis-van Diemen; Fredrik Pontén; Mark A. van de Wiel; Remond J.A. Fijneman; Gerrit A. Meijer

Background and objective Progression of a colorectal adenoma to invasive cancer occurs in a minority of adenomas and is the most crucial step in colorectal cancer pathogenesis. In the majority of cases, this is associated with gain of a substantial part of chromosome 20q, indicating that multiple genes on the 20q amplicon may drive carcinogenesis. The aim of this study was to identify genes located on the 20q amplicon that promote progression of colorectal adenoma to carcinoma. Design Functional assays were performed for 32 candidate driver genes for which a positive correlation between 20q DNA copy number and mRNA expression had been demonstrated. Effects of gene knockdown on cell viability, anchorage-independent growth, and invasion were analysed in colorectal cancer cell lines with 20q gain. Colorectal tumour protein expression was examined by immunohistochemical staining of tissue microarrays. Results TPX2, AURKA, CSE1L, DIDO1, HM13, TCFL5, SLC17A9, RBM39 and PRPF6 affected cell viability and/or anchorage-independent growth. Chromosome 20q DNA copy number status correlated significantly with TPX2 and AURKA protein levels in a series of colorectal adenomas and carcinomas. Moreover, downmodulation of TPX2 and AURKA was shown to inhibit invasion. Conclusion These data identify TPX2 (20q11) and AURKA (20q13.2) as two genes located on distinct regions of chromosome 20q that promote 20q amplicon-driven progression of colorectal adenoma to carcinoma. Therefore the selection advantage imposed by 20q gain in tumour progression is achieved by gain-of-function of multiple cancer-related genes—knowledge that can be translated into novel tests for early diagnosis of progressive adenomas.


Molecular & Cellular Proteomics | 2010

Subnuclear Proteomics in Colorectal Cancer IDENTIFICATION OF PROTEINS ENRICHED IN THE NUCLEAR MATRIX FRACTION AND REGULATION IN ADENOMA TO CARCINOMA PROGRESSION

Jakob Albrethsen; Jaco C. Knol; Sander R. Piersma; Thang V. Pham; Meike de Wit; Sandra Mongera; Beatriz Carvalho; Henk M.W. Verheul; Remond J.A. Fijneman; Gerrit A. Meijer; Connie R. Jimenez

Abnormalities in nuclear phenotype and chromosome structure are key features of cancer cells. Investigation of the protein determinants of nuclear subfractions in cancer may yield molecular insights into aberrant chromosome function and chromatin organization and in addition may yield biomarkers for early cancer detection. Here we evaluate a proteomics work flow for profiling protein constituents in subnuclear domains in colorectal cancer tissues and apply this work flow to a comparative analysis of the nuclear matrix fraction in colorectal adenoma and carcinoma tissue samples. First, we established the reproducibility of the entire work flow. In a reproducibility analysis of three nuclear matrix fractions independently isolated from the same colon tumor homogenate, 889 of 1,047 proteins (85%) were reproducibly identified at high confidence (minimally two peptides per protein at 99% confidence interval at the protein level) with an average coefficient of variance for the number of normalized spectral counts per protein of 30%. This indicates a good reproducibility of the entire work flow from biochemical isolation to nano-LC-MS/MS analysis. Second, using spectral counting combined with statistics, we identified proteins that are significantly enriched in the nuclear matrix fraction relative to two earlier fractions (the chromatin-binding and intermediate filament fractions) isolated from six colorectal tissue samples. The total data set contained 2,059 non-redundant proteins. Gene ontology mining and protein network analysis of nuclear matrix-enriched proteins revealed enrichment for proteins implicated in “RNA processing” and “mRNA metabolic process.” Finally, an explorative comparison of the nuclear matrix proteome in colorectal adenoma and carcinoma tissues revealed many proteins previously implicated in oncogenesis as well as new candidates. A subset of these differentially expressed proteins also exhibited a corresponding change at the mRNA level. Together, the results show that subnuclear proteomics of tumor tissue is feasible and a promising avenue for exploring oncogenesis.


Tumor Biology | 2010

Identification of key genes for carcinogenic pathways associated with colorectal adenoma-to-carcinoma progression

Anke H. Sillars-Hardebol; Beatriz Carvalho; Meike de Wit; Cindy Postma; Pien M. Delis-van Diemen; Sandra Mongera; Bauke Ylstra; Mark A. van de Wiel; Gerrit A. Meijer; Remond J.A. Fijneman

Colorectal adenomas form a biologically and clinically distinct intermediate stage in development of colorectal cancer (CRC) from normal colon epithelium. Only 5% of adenomas progress into adenocarcinomas, indicating that malignant transformation requires other biological alterations than those involved in adenoma formation. The present study aimed to explore which cancer-related biological processes are affected during colorectal adenoma-to-carcinoma progression and to identify key genes within these pathways that can serve as tumor markers for malignant transformation. The activity of 12 cancer-related biological processes was compared between 37 colorectal adenomas and 31 adenocarcinomas, using the pathway analysis tool Gene Set Enrichment Analysis. Expression of six gene sets was significantly increased in CRCs compared to adenomas, representing chromosomal instability, proliferation, differentiation, invasion, stroma activation, and angiogenesis. In addition, 18 key genes were identified for these processes based on their significantly increased expression levels. For AURKA and PDGFRB, increased mRNA expression levels were verified at the protein level by immunohistochemical analysis of a series of adenomas and CRCs. This study revealed cancer-related biological processes whose activities are increased during malignant transformation and identified key genes which may be used as tumor markers to improve molecular characterization of colorectal tumors.


Journal of Proteomics | 2014

Colorectal cancer candidate biomarkers identified by tissue secretome proteome profiling.

Meike de Wit; Huub Kant; Sander R. Piersma; Thang V. Pham; Sandra Mongera; Maaike van Berkel; Epie Boven; Fredrik Pontén; Gerrit A. Meijer; Connie R. Jimenez; Remond J.A. Fijneman

UNLABELLED Colorectal cancer (CRC) is a major health problem. Biomarkers associated with molecular changes in cancer cells can aid early detection, diagnosis, prognosis, therapy selection, and disease monitoring. Tumor tissue secretomes are a rich source of candidate biomarkers. To identify CRC protein biomarkers, secretomes of four pairs of human CRC tissue and patient-matched normal colon tissue samples, and secretomes of five CRC cell lines were analyzed by GeLC-MS/MS. Subsequent data analysis was based on label-free spectral counting, Ingenuity Pathway Analysis, Secretome/SignalP, STRING and Cytoscape, resulting in 2703 protein identifications in the tissue secretomes, of which 409 proteins were significantly more present in CRC samples than in controls. Biomarker selection of 76 candidates was based on consistent and abundant over-representation in cancer- compared to control-secretomes, and presumed neoplastic origin. Overlap analysis with previously obtained datasets revealed 21 biomarkers suited for early detection of CRC. Immunohistochemistry confirmed overexpression in CRC of one candidate marker (MCM5). In conclusion, a human reference dataset of 76 candidate biomarkers was identified for which we illustrate that combination with existing pre-clinical datasets allows pre-selection of biomarkers for blood- or stool-based assays to support clinical management of CRC. Further dedicated validation studies are required to demonstrate their clinical applicability. BIOLOGICAL SIGNIFICANCE Tissue secretome proteomes are a rich source of candidate biomarkers. Several secretome proteome datasets have been obtained from pre-clinical in vitro and in vivo colorectal cancer (CRC) model systems, yielding promising CRC biomarkers obtained under well-defined experimentally controlled conditions. However, which of these biomarker proteins are actually secreted by human CRC samples was not known. To our knowledge, this is the first study that directly compares secretome proteomes from clinically relevant human CRC tissues to patient-matched normal colon tissues. We identified 76 human CRC protein biomarkers that may facilitate blood-based or stool-based assay development to support clinical management of CRC. Overlap analysis with datasets from well-defined pre-clinical studies helps to determine what clinical application suits these human CRC biomarkers best, i.e. early detection, diagnosis, prognosis, therapy selection, and/or disease monitoring of CRC. This is demonstrated for a CRC mouse model dataset, revealing 21 human CRC biomarkers suited for early detection of CRC.


Virchows Archiv | 2008

Presence of HIF-1 and related genes in normal mucosa, adenomas and carcinomas of the colorectum

Astrid E. Greijer; Pien M. Delis-van Diemen; Remond J.A. Fijneman; Rachel H. Giles; Emile E. Voest; Victor W.M. van Hinsbergh; Gerrit A. Meijer

Expression of the transcription factor hypoxia-inducible factor 1 (HIF-1), which plays a key role in cellular adaptation to hypoxia, was investigated in normal colorectal mucosa (ten), adenomas (61), and carcinomas (23). Tissue samples were analyzed for HIF-1α, its upstream regulators, von Hippel–Lindau factor, AKT, and mammalian target of rapamycin (mTOR) and its downstream targets glucose transporter 1 (GLUT1), carbonic anhydrase IX, stromal-cell-derived factor 1 (SDF-1) by immunohistochemistry. In normal colorectal mucosa, HIF-1α was observed in almost all nuclei of surface epithelial cells, probably secondary to a gradient of oxygenation, as indicated by pimonidazole staining. The same staining pattern was present in 87% of adenomas. In carcinomas, HIF-1α was present predominantly around areas of necrosis (78%). Active AKT and mTOR, were present in all adenomas, carcinomas, and in normal colorectal mucosa. GLUT1 and SDF-1 were present in the normal surface epithelium of all adenoma cases, whereas in the carcinoma GLUT1 was located around necrotic regions and SDF-1 was present in all epithelial cells. In conclusion, HIF-1α appears to be physiologically expressed in the upper part of the colorectal mucosa. The present observations support that upregulation of HIF-1α and its downstream targets GLUT1 and SDF-1 in colorectal adenomas and carcinomas may be due to hypoxia, in close interaction with an active phosphatidylinositol 3-kinases–AKT–mTOR pathway.

Collaboration


Dive into the Remond J.A. Fijneman's collaboration.

Top Co-Authors

Avatar

Gerrit A. Meijer

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Beatriz Carvalho

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Connie R. Jimenez

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Meike de Wit

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sander R. Piersma

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Veerle M.H. Coupé

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

G. A. Meijer

Netherlands Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Jeroen A.M. Beliën

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Thang V. Pham

VU University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge