Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Ann Oncol ; 31(11): 1536-1544, 2020 11.
Article in English | MEDLINE | ID: mdl-32861806

ABSTRACT

BACKGROUND: In AURA3 (NCT02151981), osimertinib, a third-generation epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor (TKI), significantly prolonged progression-free survival and improved response in patients with EGFR T790M advanced non-small-cell lung cancer (NSCLC) and progression on prior EGFR-TKI treatment. We report the final AURA3 overall survival (OS) analysis. PATIENTS AND METHODS: Adult patients were randomized 2 : 1 to osimertinib (80 mg orally, once daily) or pemetrexed plus carboplatin/cisplatin (platinum-pemetrexed) intravenously, every 3 weeks (≤6 cycles). Patients could crossover to osimertinib on progression confirmed by blinded independent central review. OS and safety were secondary end points. RESULTS: A total of 279 patients were randomly assigned to receive osimertinib and 140 to platinum-pemetrexed (136 received treatment). At data cut-off (DCO; 15 March 2019), 188 patients (67%) receiving osimertinib versus 93 (66%) receiving platinum-pemetrexed had died. The hazard ratio (HR) for OS was 0.87 [95% confidence interval (CI) 0.67-1.12; P = 0.277]; the median OS was 26.8 months (95% CI 23.5-31.5) versus 22.5 months (95% CI 20.2-28.8) for osimertinib and platinum-pemetrexed, respectively. The estimated 24- and 36-month survival was 55% versus 43% and 37% versus 30%, respectively. After crossover adjustment, there was an HR of 0.54 (95% CI 0.18-1.6). Time to first subsequent therapy or death showed a clinically meaningful advantage toward osimertinib (HR 0.21, 95% CI 0.16-0.28; P < 0.001). At DCO, 99/136 (73%) patients in the platinum-pemetrexed arm had crossed over to osimertinib, 66/99 (67%) of whom had died. The most common adverse events possibly related to study treatment were diarrhea (32%; grade ≥3, 1%) and rash (grouped term; 32%; grade ≥3, <1%) in the osimertinib arm, versus nausea (47%; grade ≥3, 3%) in the platinum-pemetrexed arm. CONCLUSIONS: In patients with T790M advanced NSCLC, no statistically significant benefit in OS was observed for osimertinib versus platinum-pemetrexed, which possibly reflects the high crossover rate of patients from platinum-pemetrexed to osimertinib. CLINICAL TRIALS NUMBER: ClinicalTrials.gov NCT02151981; https://clinicaltrials.gov/ct2/show/NCT02151981.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Acrylamides , Adult , Aniline Compounds/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Pemetrexed/therapeutic use , Platinum/therapeutic use , Protein Kinase Inhibitors/adverse effects , Survival Analysis
2.
Ann Oncol ; 25(2): 409-15, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24368400

ABSTRACT

BACKGROUND: Preclinical data suggest combining a mammalian target of rapamycin inhibitor with erlotinib could provide synergistic antitumor effects in advanced non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS: In this multicenter, open-label, phase II study, patients with advanced NSCLC that progressed after one to two previous chemotherapy regimens were randomized 1:1 to erlotinib 150 mg/day±everolimus 5 mg/day. Primary end point was the disease control rate (DCR) at 3 months; secondary end points included progression-free survival (PFS) and safety. RESULTS: One hundred thirty-three patients received everolimus-erlotinib (n=66) or erlotinib alone (n=67). The DCR at 3 months was 39.4% and 28.4%, respectively. The probability for the difference in disease control at 3 months to be ≥15% was estimated to be 29.8%, which was below the prespecified probability threshold of ≥40%. Median PFS was 2.9 and 2.0 months, respectively. Grade 3/4 adverse events occurred in 72.7% and 32.3% of patients, respectively. Grade 3/4 stomatitis was observed in 31.8% of combination therapy recipients. CONCLUSIONS: Everolimus 5 mg/day plus erlotinib 150 mg/day was not considered sufficiently efficacious per the predefined study criteria. The combination does not warrant further investigation based on increased toxicity and the lack of substantial improvement in disease stabilization.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Non-Small-Cell Lung/mortality , Diarrhea/chemically induced , Disease-Free Survival , Erlotinib Hydrochloride , Everolimus , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Male , Middle Aged , Quinazolines/administration & dosage , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Treatment Outcome
3.
Clin Cancer Res ; 7(10): 3127-34, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11595705

ABSTRACT

PURPOSE: To better understand the role of G(1)-S transition regulator abnormalities in the pathogenesis of advanced premalignant lesions of the upper aerodigestive tract and the biological effects of chemoprevention, we studied biopsies obtained sequentially from participants in a prospective trial using 13-cis retinoic acid, IFN-alpha, and alpha-tocopherol for 12 months. EXPERIMENTAL DESIGN: Cyclin D1 and p16 expression were analyzed by immunohistochemistry, loss of heterozygosity by polymerase chain reacting amplification, and then electrophoretic separation of the products, methylation of the p16 promoter by methylation-specific polymerase chain reacting, and cyclin D1 gene amplification by fluorescence in situ hybridization. RESULTS: Baseline dysregulation of cyclin D1 expression was found in 50% (14 of 28) and was reversed in 6 of 14 cases, whereas p16 expression was lost in 46% (13 of 28) and regained in 2 of 13 cases. Loss of heterozygosity at 9p21 occurred in 68% and p16(INK4a) promoter methylation occurred in 75% of cases, with increasing frequency from mild to severe dysplasia. Cyclin D1 gene amplification was identified in two cases. Cyclin D1 protein dysregulation at last follow-up alone and in combination with p16 loss was associated with histological progression and cancer development (P < 0.01). CONCLUSIONS: Additional study of these alterations in a larger sample and exploration of the upstream signaling partners of these cell cycle regulators in vivo is warranted to identify cancer risk profiles that would be meaningful targets for chemopreventive intervention.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclins/genetics , Head and Neck Neoplasms/pathology , Precancerous Conditions/pathology , Adult , Aged , Cell Cycle Proteins/physiology , Chromosomes, Human, Pair 9/genetics , Cyclin D , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclins/metabolism , DNA/genetics , DNA/metabolism , DNA Methylation , Female , Gene Expression Regulation , Head and Neck Neoplasms/prevention & control , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Loss of Heterozygosity , Male , Microsatellite Repeats , Middle Aged , Polymerase Chain Reaction/methods , Precancerous Conditions/genetics , Precancerous Conditions/metabolism
4.
Bull Cancer ; 88(4): 351-61, 2001 Apr.
Article in French | MEDLINE | ID: mdl-11371369

ABSTRACT

Lung as well as head and neck cancer represent an important public health problem worldwide, with lung cancer being the leading cause of cancer death in western countries. Although early stage disease is often curable with surgery or radiotherapy, the majority of patients present with advanced disease in which despite advances in combined modality therapy the outcomes have not dramatically improved. Furthermore, patients cured of their initial early stage lung or head and neck carcinoma are at high risk for development of second primary tumors, which pose the main threat to their survival. An alternative approach in reducing the incidence and thus mortality of these cancers is chemoprevention, the use of agents to reverse, halt or delay carcinogenesis. The carcinogenesis process in lung and head and neck cancer results from a dysregulation of cellular proliferation, differentiation and cell death resulting from field-wide exposure of the upper and lower airway track to tobacco smoking. This review article presents main data regarding the actual understanding of lung and head and neck carcinogenesis, as well as results of major chemoprevention trials in this field.


Subject(s)
Biomarkers, Tumor/genetics , Genetic Markers/genetics , Lung Neoplasms/diagnosis , Otorhinolaryngologic Neoplasms/diagnosis , Anticarcinogenic Agents/therapeutic use , Chromosome Aberrations/genetics , Clinical Trials as Topic , Genetic Predisposition to Disease/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/prevention & control , Otorhinolaryngologic Neoplasms/genetics , Otorhinolaryngologic Neoplasms/prevention & control , Risk Factors
5.
Int J Cancer ; 88(6): 852-5, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11093804

ABSTRACT

Head-and-neck squamous-cell cancer (HNSCC) is an important public-health problem, accounting for approximately 40,300 new cancer cases and 12,000 cancer deaths annually in the United States (Greenlee et al., [2000]). Patients with early-stage disease are often cured with surgery or radiotherapy but are at high risk for second primary tumor (SPT) development (Lippman and Hong, [1989]), and the majority of patients present with advanced disease, for which the outcomes have not markedly improved despite advances in combined-modality therapy (Vokes et al., [1993]). HNSCC arises from transformation of the genetic material of normal cells, followed by successive genetic alterations in a multistep fashion, leading to clonal evolution of progeny cells with a proliferative advantage (Vogelstein and Kinzler, [1993]), induced by tobacco carcinogens (Slaughter et al., [1953]). Chemoprevention aims at reversal of this process through re-regulation of growth and differentiation and possibly elimination of genetically and phenotypically aberrant clones. Chemoprevention studies in upper aerodigestive tract (UADT) cancers are based on these fundamental premises and the identification of molecular genetic and biologic cellular changes. These alterations represent biomarkers of the carcinogenesis process and ultimately, if validated, could serve as intermediate end points for these studies.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/genetics , Retinoids/therapeutic use , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/prevention & control , Clinical Trials as Topic , DNA Methylation , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Gene Silencing , Genes, p16/genetics , Genes, p53/genetics , Genetic Markers , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/prevention & control , Humans , Loss of Heterozygosity , Receptors, Retinoic Acid/metabolism
6.
Curr Opin Oncol ; 12(3): 240-5, 2000 May.
Article in English | MEDLINE | ID: mdl-10841196

ABSTRACT

Head and neck cancer is an important public health problem worldwide, accounting for approximately 40,400 new cancer cases and 12,300 cancer deaths annually in the US. Although early-stage disease is often curable with surgery or radiotherapy, the majority of patients present with advanced disease in which despite advances in combined modality therapy the outcomes have not dramatically improved. Furthermore, patients cured of their initial early-stage head and neck squamous cell carcinoma are at high risk for development of second primary tumors, which pose the main threat to survival. An alternative approach in reducing the incidence and thus mortality associated with these cancers is chemoprevention, the use of agents to reverse, halt, or delay carcinogenesis. The carcinogenesis process in head and neck cancer results from a dysregulation of cellular proliferation, differentiation, and cell death resulting from field-wide exposure of the upper aerodigestive tract to tobacco smoking. Newly acquired knowledge in the field of tumor biology and of the genetic changes underlying carcinogenesis through the use of new molecular technology represents the basis on which chemoprevention efforts should be based.


Subject(s)
Head and Neck Neoplasms/prevention & control , Chemoprevention , Clinical Trials as Topic , Genetic Predisposition to Disease , Head and Neck Neoplasms/genetics , Humans , Retinoids/therapeutic use
7.
Arch Otolaryngol Head Neck Surg ; 125(10): 1083-9, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10522499

ABSTRACT

OBJECTIVES: To evaluate the efficacy and secondarily the toxic effects of biochemopreventive therapy (high-dose isotretinoin [13-cis-retinoic acid], alpha-tocopherol, and interferon alfa) in the reversal of advanced premalignant lesions of the upper aerodigestive tract and to correlate the therapeutic events with modulation of biomarkers. DESIGN: Prospective, nonrandomized chemoprevention trial. SETTING: Tertiary cancer care referral center and ambulatory care. PARTICIPANTS: Thirty-six patients with advanced premalignant lesions of the upper aerodigestive tract, without cancer during the 2 years before the intervention, with evaluable lesions, and without retinoid therapy for 3 months before the trial. INTERVENTION: Administration of oral isotretinoin (100 mg/m2 per day), oral alpha-tocopherol (1200 IU/d), and subcutaneous interferon alfa (3 megaunits per square meter twice weekly) for 12 months, with serial biopsies and clinical examination at 0, 6, 12, and 18 months from study start. MAIN OUTCOME MEASURES: Clinical and histologic responses to the intervention. RESULTS: Of the 36 patients, evaluation was possible in 30 for response at 6 months and in 21 at 12 months. At 6 months, there were 10 pathologic complete responses and 7 partial responses; at 12 months, 7 complete and 3 partial responses. A striking difference in response was observed in favor of laryngeal lesions (9/19 [47%] complete response rate at 6 months and 7/14 [50%] at 12 months vs 1/11 [9%] and 0/7 [0%], respectively, for oral lesions). Toxic effects were acceptable and did not exceed grade 3. CONCLUSION: Biochemoprevention is a promising biologic approach for laryngeal dysplasia and needs to be investigated further.


Subject(s)
Antineoplastic Agents/therapeutic use , Interferon-alpha/therapeutic use , Isotretinoin/therapeutic use , Laryngeal Neoplasms/prevention & control , Mouth Neoplasms/prevention & control , Precancerous Conditions/drug therapy , Vitamin E/therapeutic use , Adult , Aged , Antineoplastic Agents/administration & dosage , Chemoprevention , Female , Humans , Interferon-alpha/administration & dosage , Isotretinoin/administration & dosage , Male , Middle Aged , Prospective Studies , Treatment Outcome , Vitamin E/administration & dosage
9.
Oncogene ; 17(18): 2313-22, 1998 Nov 05.
Article in English | MEDLINE | ID: mdl-9811462

ABSTRACT

Cyclin D1 proto-oncogene is a key regulator of the mammalian cell-cycle acting at the restriction point in late G1. Amplification of the cyclin D1 locus, located on chromosome 11q13, as well as cyclin D1 protein overexpression have been reported in several human malignancies. The purpose of this study was to evaluate cyclin D1 gene copy status and protein expression during the multistep process of head and neck tumorigenesis, using a combination of fluorescence in situ hybridization and immunohistochemistry techniques. From 29 selected patients presenting with head and neck squamous carcinoma and whose tumor cytospins had been previously screened for presence (16 cases) or absence (13 cases) of amplification at the 11q13 band, we analysed 46 paraffin-embedded tissue specimens that demonstrated, besides the primary tumor, the presence of contiguous adjacent normal tissue and/or premalignant lesions. Of the 16 amplified cases, nine demonstrated a continuous progression from premalignant to invasive carcinoma and seven (77.7%) of these cases showed cyclin D1 gene amplification in premalignant lesions prior to development of invasive carcinoma. Increased cyclin D1 protein expression was observed in all 16 amplified tumors and five of the 13 (38.4%) non-amplified tumors. Interestingly, dysregulated cyclin D1 expression was also found in the premalignant lesions adjacent to all 16 amplified tumors, and it appeared to precede cyclin D1 gene amplification. In contrast no dysregulated expression was detected in the premalignant lesions of the non-amplified tumors. In conclusion, these findings provide strong evidence for early dysregulation of cyclin D1 expression during the tumorigenesis process and suggest that dysregulated increased expression precedes and possibly enables gene amplification.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cyclin D1/metabolism , Fibroblast Growth Factors/metabolism , Gene Amplification , Head and Neck Neoplasms/metabolism , Neoplasm Proteins/metabolism , Precancerous Conditions/metabolism , Proto-Oncogene Proteins/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cyclin D1/genetics , Fibroblast Growth Factor 3 , Fibroblast Growth Factors/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , In Situ Hybridization, Fluorescence , Neoplasm Proteins/genetics , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics
10.
Clin Cancer Res ; 4(3): 539-44, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9533520

ABSTRACT

The 18q chromosomal region is frequently lost in head and neck squamous cell carcinomas (HNSCCs). Several candidate tumor suppressor genes have been mapped to this chromosomal region, including DCC, DPC4, and MADR2. The latter two genes are members of the Smad family, key downstream mediators in the transforming growth factor beta signaling pathway, and their alterations could confer resistance to transforming growth factor beta and contribute to tumorigenesis. Nevertheless, genetic alterations of DCC and DPC4 in HNSCC have not been frequently reported. To further investigate the extent and significance of the loss of the 18q chromosomal region in HNSCC, we performed detailed mapping at this region in a set of 50 primary HNSCCs using 19 highly polymorphic microsatellite markers. We detected loss of heterozygosity in 84% of the tumors tested and were able to identify three minimal deleted regions encompassing markers D18S467-D18S474 at 18q12 (4 cM), D18S1099-D18S487 at 18q21.1 (3 cM), and D18S69-41 at 18q21.1-q21.2 (2 cM). Of these minimal deleted regions, only one harbors a known candidate tumor suppressor gene, DCC, which maps telomeric to D18S46. In addition, the role of the MADR2 gene in HNSCCs was investigated by examining nine HNSCC cell lines for alterations of the gene by reverse transcription-PCR and direct sequencing analysis. No mutations or polymorphisms were detected, making this gene an unlikely target of the frequent loss at 18q in HNSCC. Our data indicate high frequency of loss of heterozygosity at 18q in HNSCC and the presence of at least two as yet unidentified tumor suppressor genes in this chromosomal region. Additional efforts to identify these putative tumor suppressor genes are warranted.


Subject(s)
Carcinoma, Squamous Cell/genetics , Chromosome Deletion , Chromosomes, Human, Pair 18 , Genes, Tumor Suppressor , Head and Neck Neoplasms/genetics , Tumor Suppressor Proteins , Carcinoma, Squamous Cell/pathology , Cell Adhesion Molecules/genetics , Chromosome Mapping , DCC Receptor , DNA-Binding Proteins/genetics , Genetic Markers , Head and Neck Neoplasms/pathology , Humans , Multigene Family , Polymerase Chain Reaction , Receptors, Cell Surface , Smad2 Protein , Smad4 Protein , Trans-Activators/genetics , Tumor Cells, Cultured
11.
Cancer J Sci Am ; 3(2): 92-9, 1997.
Article in English | MEDLINE | ID: mdl-9099459

ABSTRACT

PURPOSE: To determine the efficacy of the combination of cisplatin, fluorouracil, and high-dose l-leucovorin (PFL) as organ-preserving induction therapy followed by radiotherapy in untreated patients with advanced squamous cell carcinoma of the head and neck. PATIENTS AND METHODS: This was a phase II study of PFL in 47 patients with resectable stage III (n = 20) and IV (n = 27) M0 squamous cell carcinoma of the head and neck, including larynx (n = 20), hypopharynx (n = 14), and oropharynx (n = 13). The PFL regimen consisted of cisplatin 25 mg/m2 on days 1 through 5, fluorouracil 800 mg/m2 CI on days 2 through 6, and l-leucovorin 250 mg/m2 on days 1 through 6, all by continuous intravenous infusion every 21 to 28 days for three courses. The primary study endpoint was initial response to and local disease control rate with PFL as induction chemotherapy, with an aim to confirm the previously reported complete response rate of 60% to 70%. RESULTS: Of 47 patients enrolled, 46 were evaluable for response to PFL, 14 (30%) achieved a complete response, and 25 (54%) achieved a partial response, for an overall response rate of 84%. Of 39 patients evaluable for response after radiation therapy, 27 (69%) achieved a complete response and 11 (28%) a partial response. Local disease control was achieved in 37 of 46 (80%). Grade 3 or 4 toxic effects occurred frequently, with neutropenia in 27 (59%) of 46 evaluable patients, thrombocytopenia in 30%, mucositis in 41%, diarrhea in 13%, and nausea/ vomiting in 13%, but there were no treatment-related deaths. With a median follow-up of 35 months there have been nine recurrences (four local/regional and five distant) and 17 deaths (12 in patients with disease progression and five not directly related to the primary tumor). Second primary tumors have developed in six patients. At 3 years 62% of the patients remain alive with no disease progression, and the 3-year survival estimate with preserved organ function is 66%. CONCLUSION: PFL induction chemotherapy produced only a modest complete response rate, possibly due to suboptimal dose intensity, and was associated with substantial, although not life-threatening, toxicity. Newer regimens and treatment modalities are still needed in the management of advanced squamous cell carcinoma of the head and neck.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cisplatin/adverse effects , Cisplatin/therapeutic use , Female , Fluorouracil/adverse effects , Fluorouracil/therapeutic use , Head and Neck Neoplasms/mortality , Head and Neck Neoplasms/pathology , Humans , Leucovorin/adverse effects , Leucovorin/therapeutic use , Male , Middle Aged , Mouth Mucosa/radiation effects , Neoplasm Staging , Radiation Injuries/etiology , Stomatitis/etiology , Survival Rate
13.
Proc Soc Exp Biol Med ; 216(2): 283-90, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9349698

ABSTRACT

Over the last few years, we have witnessed tremendous progress in both basic and clinical research on retinoids. Preclinical studies have indicated the potential of retinoids in cancer prevention and therapy, but the actual successful application of retinoids in clinical chemoprevention trials has been the recent and exciting development in the field of retinoid research. Our understanding of the role of retinoids in normal developmental processes and the differentiation of normal and malignant cells, and the fundamental discovery of the nuclear retinoid receptors that act as transcription modulating factors regulating specific gene expression have been major advances in the field of basic retinoid research. Chemoprevention is the newest research approach in our efforts to control upper-aerodigestive tract cancers, which have one of the lowest cure rates among epithelial malignancies, and in which the occurrence of second primary tumors further burdens the dismal prognosis of patients. The efficacy of retinoids in the reversal of oral premalignant lesions and the prevention of second primary tumors has generated tremendous enthusiasm among retinoid researchers, particularly those in the field of chemoprevention. Current explorations of combinations of retinoids with biologic response modifiers such as alpha-interferon, as well as new receptor-selective retinoids, hold promise for the future.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Chemoprevention , Head and Neck Neoplasms/prevention & control , Retinoids/therapeutic use , Clinical Trials as Topic , Head and Neck Neoplasms/etiology , Head and Neck Neoplasms/therapy , Humans , Leukoplakia, Oral/drug therapy , Neoplasms, Second Primary/prevention & control , Retinoids/chemistry , Retinoids/metabolism , Risk Factors
15.
Cancer Metastasis Rev ; 15(1): 53-76, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8842479

ABSTRACT

One way to explain the development of head and neck cancer is through the theories of field cancerization, i.e., the exposure of an entire field of tissue to repeated carcinogenic insult, and multistep process, i.e., development of multiple cancers in a predisposed filed through a series of recognizable stages. Recent molecular genetic studies of histologically normal and premalignant epithelia of high-risk subjects and studies of malignant tumors in aerodigestive tract epithelia have identified a continuum of accumulated specific genetic alterations that possibly occur during the clonal evolution of tumors, namely, during the multistep process. Second primary or multiple primary tumors arise in the same fields as independent clones, with similar but unique molecular genetic and/or cellular alterations. Consequently, the assessment of these genetic and phenotypic alterations has been integrated into clinical chemoprevention trials in an effort to identify biomarkers that are also risk predictors and intermediate end points. This review covers candidate biomarkers of the processes of field cancerization and multistep tumor development in aerodigestive tract epithelia, including general and specific genetic markers, proliferation markers, and squamous differentiation markers.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/etiology , Head and Neck Neoplasms/genetics , Carcinoma, Squamous Cell/prevention & control , Genetic Markers , Head and Neck Neoplasms/prevention & control , Humans , Molecular Biology
SELECTION OF CITATIONS
SEARCH DETAIL
...