A genetic explanation of Slaughter's concept of field cancerization: evidence and clinical implications.

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.

[1]  R H Hruban,et al.  Molecular assessment of histopathological staging in squamous-cell carcinoma of the head and neck. , 1995, The New England journal of medicine.

[2]  R. Stern,et al.  p53 mutation in nonmelanoma skin cancers occurring in psoralen ultraviolet a-treated patients: evidence for heterogeneity and field cancerization. , 2002, The Journal of investigative dermatology.

[3]  S. Gartler,et al.  DISTRIBUTION OF GLUCOSE-6-PHOSPHATE DEHYDROGENASE ELECTROPHORETIC VARIANTS IN DIFFERENT TISSUES OF HETEROZYGOTES. , 1965, American journal of human genetics.

[4]  R. Iggo,et al.  Field cancerisation and polyclonal p53 mutation in the upper aero- digestive tract , 1997, Oncogene.

[5]  G. Snow,et al.  Mutated p53 as a molecular marker for the diagnosis of head and neck cancer , 2002, The Journal of pathology.

[6]  E. Gabrielson,et al.  Multiple head and neck tumors: evidence for a common clonal origin. , 1996, Cancer research.

[7]  W. Hittelman Genetic Instability in Epithelial Tissues at Risk for Cancer , 2001, Annals of the New York Academy of Sciences.

[8]  B. Vogelstein,et al.  A genetic model for colorectal tumorigenesis , 1990, Cell.

[9]  A. Leunig,et al.  A comparative study of normal inspection, autofluorescence and 5‐ALA‐induced PPIX fluorescence for oral cancer diagnosis , 2002, International journal of cancer.

[10]  D. Slaughter,et al.  “Field cancerization” in oral stratified squamous epithelium. Clinical implications of multicentric origin , 1953, Cancer.

[11]  B. Reid,et al.  p53-mutant clones and field effects in Barrett's esophagus. , 1999, Cancer research.

[12]  N. Wright,et al.  Field cancerization, clonality, and epithelial stem cells: the spread of mutated clones in epithelial sheets , 1999, The Journal of pathology.

[13]  M. K Rollert,et al.  Oral white lesions with special reference to precancerous and tobacco-related lesions: Conclusions of an international symposium held in Uppsala, Sweden, May 18–21 1994 , 1996 .

[14]  P. V. van Diest,et al.  Comparative molecular and histological grading of epithelial dysplasia of the oral cavity and the oropharynx , 2003, The Journal of pathology.

[15]  C. R. Leemans,et al.  Discordance of genetic alterations between primary head and neck tumors and corresponding metastases associated with mutational status of the TP53 gene , 2002, Genes, chromosomes & cancer.

[16]  G. Snow,et al.  Mutagen sensitivity as a biomarker for second primary tumors after head and neck squamous cell carcinoma. , 2000, Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology.

[17]  A. Harłozińska,et al.  Field effect of human colon carcinoma on normal mucosa: relevance of carcinoembryonic antigen expression. , 1996, Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine.

[18]  S. Lippman,et al.  Frequent microsatellite alterations at chromosomes 9p21 and 3p14 in oral premalignant lesions and their value in cancer risk assessment , 1996, Nature Medicine.

[19]  S. Gollin Chromosomal alterations in squamous cell carcinomas of the head and neck: Window to the biology of disease , 2001, Head & neck.

[20]  C. R. Leemans,et al.  Multiple head and neck tumors frequently originate from a single preneoplastic lesion. , 2002, The American journal of pathology.

[21]  K. Schepman,et al.  Malignant transformation of oral leukoplakia: a follow-up study of a hospital-based population of 166 patients with oral leukoplakia from The Netherlands. , 1998, Oral oncology.

[22]  C. R. Leemans,et al.  Persistence of genetically altered fields in head and neck cancer patients: biological and clinical implications. , 2001, Clinical cancer research : an official journal of the American Association for Cancer Research.

[23]  I. Jacobs,et al.  Molecular evidence of a common clonal origin and subsequent divergent clonal evolution in vulval intraepithelial neoplasia, vulval squamous cell carcinoma and lymph node metastases , 2002, International journal of cancer.

[24]  J. Langdon,et al.  Detection of minimal residual cancer to investigate why oral tumors recur despite seemingly adequate treatment. , 2000, Clinical cancer research : an official journal of the American Association for Cancer Research.

[25]  Chen-Yang Shen,et al.  Monoclonality and surface lesion‐specific microsatellite alterations in premalignant and malignant neoplasia of uterine cervix: a local field effect of genomic instability and clonal evolution , 1999, Genes, chromosomes & cancer.

[26]  A Semjonow,et al.  Cytogenetic analysis of multifocal bladder cancer supports a monoclonal origin and intraepithelial spread of tumor cells. , 2001, Cancer research.

[27]  W. Hong,et al.  Dysregulated cyclin D1 expression early in head and neck tumorigenesis: in vivo evidence for an association with subsequent gene amplification , 1998, Oncogene.

[28]  D. Sidransky,et al.  Allelic losses in OraTest-directed biopsies of patients with prior upper aerodigestive tract malignancy. , 2001, Clinical cancer research : an official journal of the American Association for Cancer Research.

[29]  T. Axéll,et al.  Oral white lesions with special reference to precancerous and tobacco- related lesions: conclusions of an international symposium held in Uppsala, Sweden, May 18-21 1994. International Collaborative Group on Oral White Lesions. , 1996, Journal of oral pathology & medicine : official publication of the International Association of Oral Pathologists and the American Academy of Oral Pathology.

[30]  G. Snow,et al.  A panel of biomarkers of carcinogenesis of the upper aerodigestive tract as potential intermediate endpoints in chemoprevention trials , 1993, Cancer.

[31]  Y. Miller,et al.  Widely dispersed p53 mutation in respiratory epithelium. A novel mechanism for field carcinogenesis. , 1997, The Journal of clinical investigation.

[32]  N D Le,et al.  Use of allelic loss to predict malignant risk for low-grade oral epithelial dysplasia. , 2000, Clinical cancer research : an official journal of the American Association for Cancer Research.

[33]  J. Califano,et al.  Second esophageal tumors in patients with head and neck squamous cell carcinoma: an assessment of clonal relationships. , 1999, Clinical cancer research : an official journal of the American Association for Cancer Research.

[34]  R. Tarone,et al.  Frequent clones of p53-mutated keratinocytes in normal human skin. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[35]  O. Yoshida,et al.  Clonal and chronological genetic analysis of multifocal cancers of the bladder and upper urinary tract. , 1998, Cancer research.

[36]  P. Nichols,et al.  Mosaicism in human epithelium: macroscopic monoclonal patches cover the urothelium. , 1995, The Journal of urology.

[37]  Fiona M. Watt,et al.  Separation of human epidermal stem cells from transit amplifying cells on the basis of differences in integrin function and expression , 1993, Cell.

[38]  J. Minna,et al.  Multiple clonal abnormalities in the bronchial epithelium of patients with lung cancer. , 1999, Journal of the National Cancer Institute.

[39]  K. Hemminki,et al.  Loss of heterozygosity in tumour-adjacent normal tissue of breast and bladder cancer. , 2001, European journal of cancer.

[40]  G. Snow,et al.  Second primary tumors and field cancerization in oral and oropharyngeal cancer: Molecular techniques provide new insights and definitions , 2002 .

[41]  P. Nowell The clonal evolution of tumor cell populations. , 1976, Science.

[42]  L. Cupples,et al.  Genetically abnormal clones in histologically normal breast tissue. , 1998, The American journal of pathology.

[43]  B. Nickoloff,et al.  Estimation of size of clonal unit for keratinocytes in normal human skin. , 2002, Archives of pathology & laboratory medicine.