Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 10(11): 3607-13, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15173066

ABSTRACT

PURPOSE: Surgeons treating patients with head and neck squamous cell carcinoma (HNSCC) rely heavily on histology to decide whether the resection margins are tumor free and subsequent adjuvant treatments can be omitted. However, despite the presence of tumor-free margins, 10-30% of HNSCC patients still develop a locally recurrent tumor. Evidence is available that recurrent cancer develops from either (a). outgrowth of a relatively small number of tumor cells that have not been detected by the pathologist or (b). a precursor lesion in which additional genetic alterations have led again to invasive cancer. EXPERIMENTAL DESIGN: In a retrospective study on 13 HNSCC cases, we analyzed the primary tumor, its surrounding histologically tumor-free resection margins, and local recurrences for loss of heterozygosity (22 microsatellite markers on 6 chromosomes) and TP53 mutations to determine the origin of the recurrent cancer. RESULTS: A precursor lesion was absent in 5 of 13 (39%) cases, and the genetic similarity of the primary and recurrent cancer was high, providing evidence that residual cancer cells were the origin of recurrence. For the remaining eight cases (61%) a genetically related precursor lesion (field) was detected, and for five of these cases, evidence was found that both the primary and recurrent carcinoma originated from this field. The remaining three cases were less conclusive. CONCLUSIONS: This study explains the pathobiology of locally recurrent HNSCC in patients with histologically tumor-free resection margins and indicates that the development of novel therapies to decrease the local recurrence rates in HNSCC should not only be focused on eradicating residual cancer cells but also on the precursor lesions that are left behind.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Female , Genes, p53 , Humans , Loss of Heterozygosity , Male , Microsatellite Repeats , Middle Aged , Mutation , Neoplasm Recurrence, Local , Neoplasm, Residual , Recurrence , Retrospective Studies , Treatment Outcome
2.
Cancer Res ; 63(8): 1727-30, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12702551

ABSTRACT

The concept of "field cancerization" was first introduced by Slaughter et al. [D. P, Slaughter et al., Cancer (Phila.), 6: 963-968, 1953] in 1953 when studying the presence of histologically abnormal tissue surrounding oral squamous cell carcinoma. It was proposed to explain the development of multiple primary tumors and locally recurrent cancer. Organ systems in which field cancerization has been described since then are: head and neck (oral cavity, oropharynx, and larynx), lung, vulva, esophagus, cervix, breast, skin, colon, and bladder. Recent molecular findings support the carcinogenesis model in which the development of a field with genetically altered cells plays a central role. In the initial phase, a stem cell acquires genetic alterations and forms a "patch," a clonal unit of altered daughter cells. These patches can be recognized on the basis of mutations in TP53, and have been reported for head and neck, lung, skin, and breast cancer. The conversion of a patch into an expanding field is the next logical and critical step in epithelial carcinogenesis. Additional genetic alterations are required for this step, and by virtue of its growth advantage, a proliferating field gradually displaces the normal mucosa. In the mucosa of the head and neck, as well as the esophagus, such fields have been detected with dimensions of >7 cm in diameter, whereas they are usually not detected by routine diagnostic techniques. Ultimately, clonal divergence leads to the development of one or more tumors within a contiguous field of preneoplastic cells. An important clinical implication is that fields often remain after surgery of the primary tumor and may lead to new cancers, designated presently by clinicians as "a second primary tumor" or "local recurrence," depending on the exact site and time interval. In conclusion, the development of an expanding preneoplastic field appears to be a critical step in epithelial carcinogenesis with important clinical consequences. Diagnosis and treatment of epithelial cancers should not only be focused on the tumor but also on the field from which it developed.


Subject(s)
Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/genetics , Carcinoma, Squamous Cell/pathology , Genetic Predisposition to Disease , Head and Neck Neoplasms/pathology , Humans , Risk Factors
3.
J Pathol ; 199(3): 354-60, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12579537

ABSTRACT

Histological grading of epithelial dysplasia in the oral cavity and oropharynx is used to predict the risk for cancer and to determine the treatment strategy. This grading, however, is subjective and not well reproducible. Recent publications have shown that molecular markers are promising in cancer risk assessment. The aim of the present study was to compare classical histological and molecular grading and to relate these to the proliferation rate by quantitative assessment of Ki-67 staining. Forty-three samples were analysed from the margins of patients who had undergone resection of their squamous cell carcinoma in the oral cavity/oropharynx. Three experienced pathologists performed the histological grading. With the consensus score, 12 samples were classified as normal and 31 as dysplastic (21 mild, six moderate, and four severe). Loss of heterozygosity (LOH) was assessed in the same samples with 15 microsatellite markers at chromosomes 3p, 9p, 17p, 8p, 13q, and 18q, and was present in 28 of the 43 samples. Twenty-four of the 28 cases (86%) with LOH were classified as dysplastic and four as normal. All ten samples with moderate and severe dysplasia and 14 of 21 samples with mild dysplasia contained LOH. In four of 12 biopsies classified as normal, LOH was found. A very striking and significant difference of the Ki-67 index was observed between LOH-positive and LOH-negative cases, 36.6 +/- 11.1% versus 19.4 +/- 2.8% positive cells, respectively. In mild dysplasia, 13 of 14 lesions containing LOH had a higher Ki-67 index than all seven lesions without LOH. Thus, in the oral cavity/oropharynx, LOH is more frequently found in the histologically higher-grade lesions (moderate dysplasia or worse) and in the lower grade lesions when a high proliferation rate is present. Assessment of proliferation with Ki-67 is a better surrogate for LOH than histological grading.


Subject(s)
Mouth Neoplasms/genetics , Oropharyngeal Neoplasms/genetics , Precancerous Conditions/genetics , Carcinoma, Squamous Cell , Cell Division , Humans , Ki-67 Antigen/metabolism , Loss of Heterozygosity , Microsatellite Repeats , Mouth Neoplasms/pathology , Neoplasm Staging , Observer Variation , Oropharyngeal Neoplasms/pathology , Precancerous Conditions/pathology , Prognosis , Sensitivity and Specificity
4.
J Pathol ; 198(4): 476-86, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12434417

ABSTRACT

In total, 10-30% of patients with head and neck squamous cell carcinoma (HNSCC) develop local recurrences despite seemingly adequate tumour resection. This may result from minimal residual cancer (MRC): small numbers of tumour cells left behind in the surgical margins, undetectable by routine histopathology. In recent studies, p53 mutations have been considered as selective and sensitive DNA markers of cancer cells. There are two potential problems in using mutated-p53 DNA as a marker. Firstly, p53 mutations occur early in progression and might therefore detect unresected precursor lesions besides tumour cells. Secondly, DNA is a very stable biomolecule that might lead to false-positive results. These two potential problems have been evaluated in this study. Fifty patients with a radical tumour resection were included, of whom 30 showed a p53 mutation in the primary tumour. Histopathologically tumour-free surgical margins were quantitatively analysed for mutated p53 by molecular diagnosis (plaque assay) and subsequent (immuno)histopathology. p53 mutated DNA was detected in the surgical margins of 19/30 patients. Immunohistochemistry confirmed the presence of small tumour foci in 2/19 mutated p53-positive cases. In 7/19 cases, the tumour-specific p53 mutation was found in unresected dysplastic mucosal precursor lesions. Moreover, in a number of cases small p53-immunostained patches were detected, but the mutations found were never tumour-related. By screening contralateral exfoliated cells and plaque assays on RNA it was shown that detection of mutated-p53 DNA is prone to false-positive results. In conclusion, using p53 mutations as a marker, both MRC and unresected mutated p53-positive mucosal precursor lesions are detected within surgical margins. Molecular assessment of surgical margins using p53 mutations enables the selection of HNSCC patients at high risk for tumour recurrence, but tumour RNA seems at present to be a more specific biomolecule for analysis than tumour DNA.


Subject(s)
Carcinoma, Squamous Cell/diagnosis , Genes, p53 , Genetic Markers , Head and Neck Neoplasms/diagnosis , Mutation , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/surgery , DNA Mutational Analysis , DNA, Neoplasm/genetics , False Positive Reactions , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/surgery , Humans , Immunoenzyme Techniques , Neoplasm Recurrence, Local/genetics , Neoplasm, Residual , Precancerous Conditions/genetics , RNA, Neoplasm/genetics , Tumor Suppressor Protein p53/metabolism
5.
Am J Pathol ; 161(3): 1051-60, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12213734

ABSTRACT

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.


Subject(s)
Microsatellite Repeats , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mutation , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/pathology , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/pathology , Precancerous Conditions/genetics , Female , Genes, p16 , Genes, p53 , Humans , Male , Precancerous Conditions/pathology
6.
Oncology ; 63(1): 56-63, 2002.
Article in English | MEDLINE | ID: mdl-12187072

ABSTRACT

BACKGROUND AND OBJECTIVE: Retinoids show promise in the treatment of various (pre)malignancies, including head and neck squamous cell carcinoma (HNSCC). It has been shown that metabolic pathways of retinoids are important in their anticancer effect and that these pathways may change during HNSCC carcinogenesis. We have previously reported that HNSCC cells have a 17-fold greater turnover rate of retinoic acid (RA) than normal oral keratinocytes from noncancer controls, and that the formation of polar metabolites such as 4-oxo-RA and 4-hydroxy-RA is only seen in HNSCC cell lines. We aimed to establish whether this altered retinoid metabolism is an intrinsic characteristic of HNSCC patients. METHODS: The normal mucosa of cancer and noncancer patients was the source of keratinocyte cultures. The cells were exposed to RA for various time periods, and the levels of various retinoids were measured in the culture medium and cell pellets with reverse-phase liquid chromatography. RESULTS: Cells from cancer patients were morphologically normal and showed no genetic aberrations (i.e. loss of heterozygosity). The RA turnover rate in normal oral keratinocytes of cancer patients was 15 times higher (p = 0.003) than that in normal oral keratinocytes of noncancer controls, with average turnover rates of 218.6 and 14.8 pmol/mg protein/h, respectively. Specific profiles of RA metabolites were similar. CONCLUSION: The observed higher RA metabolism in noncancer cells of HNSCC patients suggests that individuals with a relatively high RA turnover have an increased risk of developing HNSCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Keratinocytes/metabolism , Tretinoin/metabolism , Adult , Aged , Cells, Cultured , Female , Humans , Loss of Heterozygosity , Male , Middle Aged , Mouth/pathology
7.
Head Neck ; 24(2): 198-206, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11891950

ABSTRACT

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.


Subject(s)
Carcinoma/classification , Carcinoma/genetics , Molecular Diagnostic Techniques , Mouth Neoplasms/classification , Mouth Neoplasms/genetics , Neoplasms, Second Primary/classification , Neoplasms, Second Primary/genetics , Oropharyngeal Neoplasms/classification , Oropharyngeal Neoplasms/genetics , Carcinoma/diagnosis , Humans , Mouth Neoplasms/diagnosis , Neoplasms, Second Primary/diagnosis , Oropharyngeal Neoplasms/diagnosis
8.
Genes Chromosomes Cancer ; 33(2): 168-77, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11793443

ABSTRACT

Ample molecular data are available on the progression from normal mucosa to invasive head and neck squamous cell carcinoma (HNSCC), but information on further genetic progression to metastatic disease is scarce. To obtain insight into the metastatic process, we compared 23 primary HNSCCs with 25 corresponding lymph node metastases (LNMs) and 10 corresponding distant metastases (DMs) with respect to TP53 mutations and patterns of loss of heterozygosity (LOH) based on 26 microsatellite markers on six chromosome arms (3p, 9p, 17p, 13q, 8p, and 18q). In 18 of the 23 patients, a TP53 mutation was detected in the primary tumor, and in all cases the same TP53 mutation was present in the corresponding LNM or DM. In nine of 20 patients with LNMs and three of seven patients with DMs, the LOH pattern of metastasis differed from that of the corresponding primary tumor by at least one marker. Microsatellite markers located on chromosome arms 13q, 8p, and 18q were most frequently discordant, providing evidence that alterations at these chromosomes occur late in HNSCC carcinogenesis. Moreover, evidence was found that DMs had developed directly from the primary tumor and not from LNMs. Remarkably, we observed that the mutational status of the TP53 gene is associated significantly with the degree of genetic differences between primary HNSCCs and corresponding metastases. All patients with TP53 wild-type primary tumors showed significantly more discordant LOH patterns in the corresponding LNMs and DMs than patients with TP53-mutated tumors. The percentages were 100% versus 27% (LNMs) and 100% versus 0% (DMs), respectively (P = 0.008 and P = 0.029; two-sided Fisher exact test). This finding suggests that TP53-mutated tumors need fewer additional genetic alterations to develop metastases compared with TP53 wild-type primary tumors.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/secondary , Chromosome Aberrations , Genes, p53/genetics , Head and Neck Neoplasms/genetics , Aged , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Carcinoma, Squamous Cell/pathology , DNA Mutational Analysis , Female , Head and Neck Neoplasms/pathology , Heart Neoplasms/genetics , Heart Neoplasms/pathology , Heart Neoplasms/secondary , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/secondary , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Loss of Heterozygosity/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lymph Nodes/pathology , Male , Microsatellite Repeats/genetics , Middle Aged , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/secondary , Splenic Neoplasms/genetics , Splenic Neoplasms/pathology , Splenic Neoplasms/secondary
SELECTION OF CITATIONS
SEARCH DETAIL
...