Network


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

Hotspot


Dive into the research topics where Ruud H. Brakenhoff is active.

Publication


Featured researches published by Ruud H. Brakenhoff.


Nature Reviews Cancer | 2011

The molecular biology of head and neck cancer

C. René Leemans; Boudewijn J. M. Braakhuis; Ruud H. Brakenhoff

Head and neck squamous cell carcinomas (HNSCCs) are caused by tobacco and alcohol consumption and by infection with high-risk types of human papillomavirus (HPV). Tumours often develop within preneoplastic fields of genetically altered cells. The persistence of these fields after treatment presents a major challenge, because it might lead to local recurrences and second primary tumours that are responsible for a large proportion of deaths. Aberrant signalling pathways have been identified in HNSCCs and inhibition of epidermal growth factor receptor (EGFR) has proved a successful therapeutic strategy. In this Review, we discuss the recent literature on tumour heterogeneity, field cancerization, molecular pathogenesis and the underlying causative cancer genes that can be exploited for novel and personalized treatments of patients with HNSCC.


Nature Reviews Cancer | 2004

Dissecting the metastatic cascade

Klaus Pantel; Ruud H. Brakenhoff

Despite recent progress in gene-expression profiling studies, the biology underlying the various patterns of metastasis that are observed in different tumour types remains unclear. The detection and characterization of disseminated tumour cells in patients with cancer has provided important new information about the cascade of metastatic events. This information has important implications for cancer prognosis and for therapy.


Nature Reviews Cancer | 2008

Detection, clinical relevance and specific biological properties of disseminating tumour cells

Klaus Pantel; Ruud H. Brakenhoff; Burkhard Brandt

Most cancer deaths are caused by haematogenous metastatic spread and subsequent growth of tumour cells at distant organs. Disseminating tumour cells present in the peripheral blood and bone marrow can now be detected and characterized at the single-cell level. These cells are highly relevant to the study of the biology of early metastatic spread and provide a diagnostic source in patients with overt metastases. Here we review the evidence that disseminating tumour cells have a variety of uses for understanding tumour biology and improving cancer treatment.


International Journal of Cancer | 2007

A novel algorithm for reliable detection of human papillomavirus in paraffin embedded head and neck cancer specimen.

Serge J. Smeets; Albertus T. Hesselink; Ernst-Jan M. Speel; Annick Haesevoets; Peter J.F. Snijders; Michael Pawlita; Chris J. L. M. Meijer; Boudewijn J. M. Braakhuis; C. Ren É Leemans; Ruud H. Brakenhoff

Human papillomavirus type 16 (HPV16) plays a role in the development of a subgroup of head and neck squamous cell carcinomas (HNSCC). However, uncertainty exists about the true impact of HPV in this tumor type as conflicting reports have been published with prevalence rates from 0 to 100%. We aimed to find a detection algorithm of a biologically and thus clinically meaningful infection, applicable for high‐throughput screening of frozen and formalin‐fixed paraffin embedded (FFPE) specimens. By considering detection of HPV E6 oncogene expression in frozen biopsies as gold standard for a meaningful HPV infection, the value of several assays was evaluated on FFPE tumor specimens and sera of 48 HNSCC patients. The following assays were evaluated on FFPE tissue samples: HPV DNA general primer (GP)5+/6+ PCR, viral load analysis, HPV16 DNA FISH detection, HPV16 E6 mRNA RT‐PCR, p16 immunostaining, and on corresponding serum samples detection of antibodies against the HPV16 proteins L1, E6 and E7. Comparing single assays on FFPE tissue samples detection of E6 expression by RT‐PCR was superior, but application remains at present limited to HPV16 detection. Most suitable algorithm with 100% sensitivity and specificity appeared p16 immunostaining followed by GP5+/6+ PCR on the p16‐positive cases. We show that clinically meaningful viral HPV infections can be more reliably measured in FFPE HNSCC samples in a standard and high throughput manner, paving the way for prognostic and experimental vaccination studies, regarding not only HNSCC, but possibly also cancer types with HPV involvement in subgroups such as penile and anal cancer.


International Journal of Cancer | 2001

Biological evidence that human papillomaviruses are etiologically involved in a subgroup of head and neck squamous cell carcinomas

Viola M. M. van Houten; Peter J.F. Snijders; Michiel W. M. van den Brekel; J. Alain Kummer; Chris J. L. M. Meijer; Bart van Leeuwen; Fedor Denkers; Ludi E. Smeele; Gordon B. Snow; Ruud H. Brakenhoff

High‐risk human papillomaviruses (HPVs) have been proposed to be associated with a subset of head and neck cancers (HNSCCs). However, clear biological evidence linking HPV‐mediated oncogenesis to the development of HNSCC is hardly available. An important biological mechanism underlying HPV‐mediated carcinogenesis is the inactivation of p53 by the HPV E6 oncoprotein. In the present study we investigated this biological relationship between HPV and HNSCC. In total 84 HNSCC tumors were analyzed for the presence of high‐risk HPV nucleic acids by DNA polymerase chain reaction‐enzyme immunoassay (PCR‐EIA) and E6 reverse transcriptase (RT)‐PCR as well as for the presence of mutations in the p53 gene. We found 20/84 HPV16 DNA‐positive cases with one or more DNA assays, 10 of which were consistently positive with all assays. Only 9/20 cases showed E6 mRNA expression, indicative for viral activity. Only these nine E6 mRNA‐positive cases all lacked a p53 mutation, whereas both the other HPV DNA‐positive and HPV‐DNA negative tumors showed p53 mutations in 36% and 63% of the cases, respectively. Moreover, only in lymph node metastases of HPV E6 mRNA‐positive tumors both viral DNA and E6 mRNA were present. Our study provides strong biological evidence for a plausible etiological role of high‐risk HPV in a subgroup of HNSCC. Analysis of E6 mRNA expression by RT‐PCR or alternatively, semiquantitative analyses of the viral load, seem more reliable assays to assess HPV involvement in HNSCC than the very sensitive DNA PCR analyses used routinely.


Head and Neck-journal for The Sciences and Specialties of The Head and Neck | 2002

Second primary tumors and field cancerization in oral and oropharyngeal cancer: Molecular techniques provide new insights and definitions†

Boudewijn J. M. Braakhuis; Maarten P. Tabor; C. René Leemans; Isaäc van der Waal; Gordon B. Snow; Ruud H. Brakenhoff

Second primary tumors (SPTs) are a significant problem in treating oral and oropharyngeal squamous cell carcinoma and have a negative impact on survival. In most studies the definition of SPT is based on the criteria of Warren and Gates, published in 1932. These criteria, however, are ill‐defined and lead to confusion. Recent molecular studies have shown that a tumor can be surrounded by a mucosal field consisting of genetically altered cells. Furthermore, evidence has been provided that SPTs (defined by classical criteria) can share some or even all genetic markers with the index tumor, indicating that both tumors have arisen from a common cell clone. We propose that these secondary neoplastic lesions should not be considered SPTs, implying that the present concept of SPT needs revision. This review describes a novel classification of the secondary tumors that develop after treatment of a carcinoma in the oral cavity or oropharynx. On the basis of the molecular analysis of the tumors and the genetically altered mucosal field in between, we propose definitions for a “true SPT,” a local recurrence, a “SFT” (second field tumor derived from the same genetically altered mucosal field as the primary tumor), and a metastasis. Considering the etiologic differences of these lesions, we believe that an accurate molecular definition is essential to make headway with the clinical management of oral and oropharyngeal cancer.


Oncogene | 2006

Genome-wide DNA copy number alterations in head and neck squamous cell carcinomas with or without oncogene-expressing human papillomavirus

Serge J. Smeets; Boudewijn J. M. Braakhuis; S Abbas; P. J. F. Snijders; Bauke Ylstra; M.A. van de Wiel; G. A. Meijer; C R Leemans; Ruud H. Brakenhoff

Oncogene-expressing human papillomavirus type 16 (HPV16) is found in a subset of head and neck squamous cell carcinomas (HNSCC). HPV16 drives carcinogenesis by inactivating p53 and pRb with the viral oncoproteins E6 and E7, paralleled by a low level of mutations in TP53 and allelic loss at 3p, 9p, and 17p, genetic changes frequently found in HNSCCs of nonviral etiology. We hypothesize that two pathways to HNSCC exist: one determined by HPV16 and the other by environmental carcinogens. To define the critical genetic events in these two pathways, we now present a detailed genome analysis of HNSCC with and without HPV16 involvement by employing high-resolution microarray comparative genomic hybridization. Four regions showed alterations in HPV-negative tumors that were absent in HPV-positive tumors: losses at 3p11.2–26.3, 5q11.2–35.2, and 9p21.1–24, and gains/amplifications at 11q12.1–13.4. Also, HPV16-negative tumors demonstrated loss at 18q12.1–23, in contrast to gain in HPV16-positive tumors. Seven regions were altered at high frequency (>33%) in both groups: gains at 3q22.2-qter, 5p15.2-pter, 8p11.2-qter, 9q22–34.1, and 20p–20q, and losses at 11q14.1-qter and 13q11–33. These data show that HNSCC arising by environmental carcinogens are characterized by genetic alterations that differ from those observed in HPV16-induced HNSCC, and most likely occur early in carcinogenesis. A number of genetic changes are shared in both tumor groups and can be considered crucial in the later stages of HNSCC progression.


Nucleic Acids Research | 2006

BAC to the future! or oligonucleotides : a perspective for micro array comparative genomic hybridization (array CGH)

Bauke Ylstra; Paul van den IJssel; Beatriz Carvalho; Ruud H. Brakenhoff; Gerrit A. Meijer

The array CGH technique (Array Comparative Genome Hybridization) has been developed to detect chromosomal copy number changes on a genome-wide and/or high-resolution scale. It is used in human genetics and oncology, with great promise for clinical application. Until recently primarily PCR amplified bacterial artificial chromosomes (BACs) or cDNAs have been spotted as elements on the array. The large-scale DNA isolations or PCR amplifications of the large-insert clones necessary for manufacturing the arrays are elaborate and time-consuming. Lack of a high-resolution highly sensitive (commercial) alternative has undoubtedly hindered the implementation of array CGH in research and diagnostics. Recently, synthetic oligonucleotides as arrayed elements have been introduced as an alternative substrate for array CGH, both by academic institutions as well as by commercial providers. Oligonucleotide libraries or ready-made arrays can be bought off-the-shelf saving considerable time and efforts. For RNA expression profiling, we have seen a gradual transition from in-house printed cDNA-based expression arrays to oligonucleotide arrays and we expect a similar transition for array CGH. This review compares the different platforms and will attempt to shine a light on the ‘BAC to the future’ of the array CGH technique.


American Journal of Pathology | 2002

Multiple Head and Neck Tumors Frequently Originate from a Single Preneoplastic Lesion

Maarten P. Tabor; Ruud H. Brakenhoff; Jacqueline E. van der Wal; G. B. Snow; C. René Leemans; Boudewijn J. M. Braakhuis

The development of second primary tumors has a negative impact on the prognosis of head and neck squamous cell carcinoma. Previously, we detected genetically altered and tumor-related mucosal lesions in the resection margins in 25% of unselected head and neck squamous cell carcinoma patients (Tabor MP, Brakenhoff RH, van Houten VMM, Kummer JA, Snel MHJ, Snijders PJF, Snow GB, Leemans CR, Braakhuis BJM: Persistence of genetically altered fields in head and neck cancer patients: biological and clinical implications. Clin Cancer Res 2001, 7: 1523-1532). The aim of this study was to determine whether first and second primary tumors are clonally related and originate from a single genetically altered field. From 10 patients we analyzed the first tumor of the oral cavity or oropharynx, the >3-cm remote second primary tumor, and the mucosa from the tumor-free margins from both resection specimens. We compared TP53 mutations and loss of heterozygosity profiles using 19 microsatellite markers at chromosomes 3p, 9p, 13q, and 17p. In all patients, genetically altered mucosal lesions were detected in at least one resection margin from both first and second primary tumor. Evidence for a common clonal origin of the first tumor, second primary tumor, and the intervening mucosa was found for at least 6 of 10 patients. Our results indicate that a proportion of multiple primary tumors have developed within a single preneoplastic field. Based on different etiology and clinical consequences, we propose that independent second primary tumors should be distinguished from second field tumors, that arise from the same genetically altered field the first tumor has developed from.


International Journal of Cancer | 2000

Molecular cloning and immunogenicity of renal cell carcinoma-associated antigen G250

Karin Grabmaier; Joost L. M. Vissers; Mirjam de Weijert; Jeannette C. Oosterwijk-Wakka; Adrie van Bokhoven; Ruud H. Brakenhoff; Elfriede Noessner; P.F.A. Mulders; Gerard Merkx; Carl G. Figdor; Gosse J. Adema; Egbert Oosterwijk

The molecular cloning of the cDNA and gene encoding the renal cell carcinoma (RCC)‐associated protein G250 is described. This protein is one of the best markers for clear cell RCC: all clear‐cell RCC express this protein, whereas no expression can be detected in normal kidney and most other normal tissue. Antibody studies have indicated that this molecule might serve as a therapeutic target. In view of the induction/up‐regulation of G250 antigen in RCC, its restricted tissue expression and its possible role in therapy, we set out to molecularly define the G250 antigen, which we identified as a transmembrane protein identical to the tumor‐associated antigen MN/CAIX. We determined, by FISH analysis, that the G250/MN/CAIX gene is located on chromosome 9p12‐13. In view of the relative immunogenicity of RCC, we investigated whether the G250 antigen can be recognized by TIL derived from RCC patients. The initial characterization of 18 different TIL cultures suggests that anti‐G250 reactivity is rare. Int. J. Cancer 85:865–870, 2000.

Collaboration


Dive into the Ruud H. Brakenhoff's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

C. René Leemans

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Elisabeth Bloemena

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter J.F. Snijders

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Arjen Brink

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Serge J. Smeets

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

Dirk J. Kuik

VU University Medical Center

View shared research outputs
Top Co-Authors

Avatar

C.R. Leemans

VU University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge