Review Open Access
Copyright ©2014 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Clin Oncol. May 10, 2014; 5(2): 114-124
Published online May 10, 2014. doi: 10.5306/wjco.v5.i2.114
Squamous cell carcinoma of the oral cavity and circulating tumour cells
Johannes Wikner, Alexander Gröbe, Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
Klaus Pantel, Sabine Riethdorf, Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
Author contributions: All of the authors substantially contributed to the conception and design of this paper, drafting the article and revising it critically for important intellectual content and the approval of the final version; Wikner J performed the initial literature search, which was updated by the other authors.
Supported by Hamburger Stiftung zur Förderung der Krebsbekämpfung; No. 188 to Gröbe A and Riethdorf S; ERC Advanced Investigator Grant “DISSECT” (Pantel K), No. 269081
Correspondence to: Sabine Riethdorf, PhD, Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany. s.riethdorf@uke.de
Telephone: +49-40-74151891 Fax: +49-40-741056546
Received: December 17, 2013
Revised: February 25, 2014
Accepted: March 13, 2014
Published online: May 10, 2014

Abstract

Due to a lack of substantial improvement in the outcome of patients suffering from oral squamous cell carcinoma (OSCC) during the past decades, current staging methods need to be revised. This disease is associated with poor survival rates despite considerable advances in diagnosis and treatment. The early detection of metastases is an important indicator of survival, prognosis and relapse. Therefore, a better understanding of the mechanisms underlying metastasis is crucial. Exploring alternative measures apart from common procedures is needed to identify new prognostic markers. Similar to previous findings predominantly for other solid tumours, recently published studies demonstrate that circulating tumour cells (CTCs) and disseminated tumour cells (DTCs) might serve as prognostic markers and could supplement routine staging in OSCC. Thus, the detection of CTCs/DTCs is a promising tool to determine the individual need for therapeutic intervention. Encouraging results and new approaches point to the future use of targeted therapies for OSCC, an exceedingly heterogeneous subgroup of head and neck cancer. This review focuses on summarising technologies currently used to detect CTCs/DTCs. The translational relevance for OSCC is highlighted. The inherent challenges in detecting CTCs/DTCs will be emphasised.

Key Words: Circulating tumour cells, Disseminated tumour cells, Oral squamous cell carcinoma, Head and neck squamous cell carcinoma, Bone marrow, Peripheral blood, Micrometastasis, Minimal residual disease, Epithelial-mesenchymal transition

Core tip: Oral squamous cell carcinoma (OSCC), among head and neck cancer, is related to poor survival rates despite considerable advances in diagnosis and treatment. Therefore, detecting tumour cell dissemination early and understanding the underlying mechanisms are crucial for predicting prognosis, relapse and survival. According to previous findings, circulating tumour cells (CTCs) and disseminated tumour cells (DTCs) might serve as prognostic markers to supplement routine staging and support determining individual therapeutic interventions. This review focuses on summarising the current knowledge about the detection of CTCs/DTCs with special emphasis on patients suffering from OSCC. The translational relevance of CTCs/DTCs and challenges for clinical application are highlighted.



INTRODUCTION

Because of its biological heterogeneity and complex behaviour, head and neck cancer must be considered a conglomeration of several tumour types. The vast majority of patients are diagnosed with squamous cell carcinoma (SCC). With an estimated yearly incidence worldwide of half a million persons, nearly 50% of patients die due to tumour-related complications[1,2]. The disease ranks age dependently among the leading types of solid cancer and is responsible for more than 65000 annual deaths in Europe[3,4]. Early stage carcinomas are only observed in one third of the patients [Union for International Cancer Control stage (UICC) I-II] while two thirds present with advanced disease (UICC-Stage III-IV). More than 50% of patients suffering from head neck squamous cell carcinoma (HNSCC) sustain local relapses while only up to 25% develop distant metastases. However, the prognosis of these patients is still poor[5-7].

Although the most important prognostic indicator for relapse is lymph node metastasis in the neck, the incidence of distant metastasis has increased dramatically[8,9]. Despite considerable advances in diagnosis[10] and therapeutic options[11], the 5-year survival rate has remained stable at approximately 50% during the past decades among all tumour stages[12,13]. This finding might also be due to an altered pattern of the clinical disease itself. While fewer patients currently present with a locoregional relapse, more patients develop a distant disease and recurrences due to occult cervical metastasis. These micrometastases are assumed to contribute to increased mortality and morbidity[14,15]. Thus, improved methods in staging oral squamous cell carcinoma (OSCC) are needed to detect early metastatic spread and any residual tumour, hence improving decisions about individual therapeutic interventions. The establishment of a resilient model of genetic instability, such as a loss of heterozygosity at distinct chromosomes and the consecutive progression from premalignant lesions to invasive tumours in head and neck cancer, led to a better understanding of the disease[2,16-19]. Due to the paradigm shift of considering solid cancers a systemic disease, the value and promise of circulating tumour cells (CTCs) in peripheral blood (PB) and disseminated tumour cells (DTCs) in bone marrow (BM) for cancer patients regarding diagnosis, prognosis, and response to therapy came into focus. While sampling BM from the easily accessible iliac crest is well established in diagnosing leukemic diseases, the full acceptance of this practice as an additional diagnostic tool for patients with solid tumours is still lacking. For carcinomas, including SCC, BM should operate as a so-called “homing organ” for CTCs becoming DTCs in the nomenclature. The clinical relevance of CTC/DTC detection in carcinoma patients has been part of extensive research, and encouraging results exist for an association between CTC detection and the prognosis in patients with other solid tumours, such as metastatic breast, prostate, and gastrointestinal cancers[20-28]. For HNSCC, a putative prognostic value was identified as well, revealing a positive correlation between CTCs/DTCs to relapse and metastasis[29-32]. In this context, it is important to note that the clinical behaviour and outcome of OSCC differ from those of the oropharynx and other HNSCC. Although there is no sharp distinction between OSCC and HNSCC in general, not all aspects and findings are transferrable. Even the main therapeutic options vary between the OSCC and other HNSCC. For OSCC, surgical removal is the primary treatment, whereas there is an ongoing debate about the preferred treatment options, such as primary radiation, (induction-) chemotherapy and supplemental targeted therapy, for laryngeal or nasopharyngeal SCC[33,34]. However, recently published data suggest that DTCs and CTCs in patients with OSCC are independent prognostic markers of disease-free survival[35]. Identifying clinical behaviour and understanding the inherent patterns of OSCC dissemination might result in meaningful contributions to the treatment of those patients. The application of new technologies that make the detection of CTCs/DTCs feasible might allow for more accurate staging and could help select patients and provide targeted therapies. To identify patients in need of systemic therapy in addition to local resection and irradiation, it is important to identify patients suffering from non-localised but “circulating” disease to expand the indication for systemic therapy. With new prognostic markers in prospect, the therapeutic options could substantially improve[36-38].

DETECTION AND CHARACTERISATION OF CTCS

Several techniques have been established to detect the rare CTCs/DTCs. To put the rarity of these cells in perspective, by applying currently available methods, it is estimated that there might be only one tumour cell among millions of other cells, even in metastatic disease. To increase the efficiency of detecting these cells, an initial enrichment step is needed. Basic enrichment approaches rely on either physical properties, such as size, density or dielectrophoretic mobility that might be characteristic at least for subgroups of CTCs/DTCs, or on the expression of specific surface molecules captured by magnetic bead-coated antibodies. Subsequently, one has to decide between cytometric/immunological approaches on one hand and molecular [mostly real-time reverse transcription-polymerase chain reaction (RT-PCR)-based] techniques on the other hand to detect the CTCs/DTCs among the simultaneously enriched non-tumoural cells[37].

A widely used method is gradient centrifugation using Ficoll-Hypaque, which utilises the density of different cell types. However, during enrichment, a substantial loss of cell-material is described[39]. The advantages of using immunocytochemical detection of enriched CTCs/DTCs include the possibility to characterise these cells by size and shape as well as by the nucleus-plasma relation. A set of monoclonal antibodies against various epithelial proteins is available, including various keratins of the cytoskeleton, surface adhesion molecules or growth factor receptors. The pan-keratin-antibody A45-B/B3 is still widely used for detecting DTCs via an alkaline phosphatase anti-alkaline phosphatase technique with New Fuchsin as a substrate[35,40]. Automated screening for potential CTCs/DTCs and visualisation can be performed with the ACIS™-system (Chromavision, San Juan Capistrano, CA, United States)[41,42].

Thus far, one of the most advanced methods for capturing and enumerating CTCs from PB is represented by the CellSearch™-system (Veridex, Raritan, NJ, United States), which provides automated enrichment and immunostaining of CTCs. It is the first food and drug administration-cleared device in CTC detection for solid cancers and has been approved for metastasised prostate, breast and colon cancers[23,43-46]. Gröbe et al[35] succeeded in detecting CTCs in a small subset of OSCC patients as well. The underlying principle uses immunomagnetic bead separation of EpCAM-positive tumour cells followed by immunofluorescent staining with anti-keratin antibodies and the exclusion of leukocytes using an anti-CD45-antibody. The CellSearch system benefits from selecting for epithelial features, such as the epithelial cell adhesion molecule (EpCAM) exposed at the surface of the vast majority of epithelial but not on normal blood cells.

A semi-automated process via fluorescence microscopy scans visualises the results. Subsequently, nucleated cells, with a diameter of at least 4 μm and characterised by keratin-positivity and CD45-negativity, are accepted and designated as epithelial cells as surrogates for tumour cells[20,44]. Other promising EpCAM-based tools were presented recently, including the microfluid-based “CTC”-chips consisting of arrays of anti-EpCAM antibody-coated microposts able to capture EpCAM-positive cells under controlled laminar flow of whole blood[47,48]. However, these techniques still lack tumour cell specificity and an application for head and neck tumours. Moreover, widespread application must be proven in large clinical trials.

Furthermore, other techniques were used for the detection of CTCs in patients with HNSCC. The Surfaced enhanced Raman Spectroscopy with epidermal growth factor receptor (EGFR) as a targeting ligand has been proven for the detection of putative CTCs in 19 patients by Wang et al[49]. The potential use of EGFR expression and activation in finding CTCs during the course of combined chemo- or bioradiotherapy regimens, accentuating the aspect of monitoring the therapeutic response in HNSCC, was described by Tinhofer et al[50] using flow cytometry[49-51]. Alternative approaches should be mentioned, however, these approaches still lack proof for their utilisation in OSCC. For example, the detection of CTCs from large blood volumes after leukapheresis is promising and paves the way for the analysis of higher numbers of CTCs either by fluorescence-activated cell sorting, immunocytochemistry or molecular approaches[52,53]. Moreover, real time monitoring of CTCs is desirable, such as during the course of therapeutic interventions. In this context, CTC detection in vivo is provided by the GILUPI cell collector device using a wire coated with anti-EpCAM antibodies ready to accumulate CTCs after insertion into a vein for 30 min. The detection of CTCs and the exclusion of leukocytes can be confirmed by immunostaining with anti-keratin and anti-CD45-antibodies, respectively[54]. To prove reliability and applicability for different tumour types and clinical relevance, several clinical trials are ongoing[37].

The detection of viable CTCs/DTCs after secretion of specific proteins during a 48 h short term culture is possible using epithelial immunospots (EPISPOT)[55]. For the EPISPOT approach, leukocytes must be depleted by negative selection of haematopoietic cells, such as using the common leukocyte antigen CD45.

To conclude, of the various introduced techniques, consideration should be given to molecular technologies. These technologies involve either DNA or complementary DNA (mRNA)-based PCR-amplification. These methods rely on gene expression patterns or the detection of known gene mutations, amplifications, other genomic aberrations or methylation patterns in tumour cells with the restriction that currently no universally applicable marker exists due to strong inter- and intra-tumoural heterogeneity. Nevertheless, using epithelium-specific targets, such as keratin 19 encoding transcripts, RT-PCR approaches are promising[56,57].

Wicha et al[58] have recently suggested, for breast and other carcinomas, that not all CTCs in these patients can be found with the approaches that are currently available and that not all detected CTCs are aggressive and have the potential to initiate metastases or recurrences, and these suggestions might also be true for OSCC. Therefore, strong efforts must be made to detect CTCs that have lost their epithelium-specific features and instead favour mesenchymal characteristics as the cells may be undergoing epithelial-mesenchymal transition (EMT). Investigations applying ultrasensitive imaging procedures, gene expression analyses and next generation sequencing approaches are ongoing and have already yielded interesting data[38,59-61].

A DISCUSSION OF THE RESULTS FROM STUDIES ON HEAD AND NECK CANCER

In comparison to other cancer types, the clinical impact of CTCs in OSCC and other HNSCC patients (Table 1) is still unclear. Therefore, investigating a correlation between these cells and clinico-pathological features is crucial. The research on the outcome of patients suffering from OSCC is impaired by comorbidities, such as chronic obstructive pulmonary disease, malnutrition, loss of function due to surgery and therapy, which can influence decisive parameters. Moreover, a sharp distinction between OSCC and HNSCC is hindered by similar but different causative pathogens, such as the contribution of human papillomaviruses (HPV) in the carcinogenesis of a subset of HNSCC, which is found less often in OSCC[62]. The incidence of the distinctly different disease of HPV-related head and neck cancers from other head and neck carcinomas is increasing[63]. This finding will influence further studies regarding etiology and outcome and therefore must be included when evaluating these studies, especially those of patients undergoing different therapy-regimens.

Table 1 Detection of circulating and disseminated tumour cells in oral squamous cell carcinoma and head neck squamous cell cancer.
SourceEntitynAssay systemCTC-positive (%)DTC-positive (%)Clinical relevanceNumber of analysed OSCCRef. yr of release
PBOSCC20RT-PCR: CK19 mRNA20.0Detection of CK19-mRNA when tumour is incisedOSCC only[85], 2000
PBHNSCC77RT-PCR: CK19 mRNA2.1No significance for CTCsVarious sites n = 13 OSCC n = 23 oropharyngeal[79], 2001
PB/ CVB BMHNSCC40ICC: Keratins CK1-8, 10, 14-16 and 19 RT-PCR: E48 mRNA10.0 (PCR) 20.0 (ICC)20.0 (PCR) 32.0 (ICC)E48 mRNA and DMFS/DFS preoperative in BM (P = 0.001/P = 0.002) and CVB (P < 0.001/P = 0.013) E48-mRNA and DMFS/DFS intraoperative in CVB (P = 0.026/0.001) ICC and DMFS preoperative in BM (P = 0.0006) and CVB (P < 0.001) ICC and DFS preoperative in BM (P = 0.036) and CVB (P = 0.042)OSCC only[81], 2003
BMHNSCC162RT-PCR: E48 mRNA40.0DTC on DFS (P = 0.1460) DMFS (P = 0.2912) N > or = 2 DMFS (P = 0.0210)Various sites n = 13 OSCC n = 23 oropharyngeal[80], 2004
BMHNSCC176ICC: CK1930.7OS and N-involvement (P = 0.0001), OS and DTC (0.0066)Various sites n = 37 OSCC n = 43 oropharyngeal[29], 2004
PBHNSCC21Immunomagnetic separation: EpCAM33.3Occurrence of CTCs more frequent in stage III-IV than in stageI-II; connection with recurrence presumed; NSVarious sites n = 7 OSCC/ oropharyngeal[83], 2007
PBOSCC25RT-PCR: CK19 mRNA16Detection of CK19-mRNA when tumour is incisedOSCC only[108], 2008
PBHNSCC48Immunomagnetic separation: ICC: CK3-6H571DFS and NO CTC (P = 0.01) clinical outcome and CTCs (P = 0.04)Various sites n = 25 OSCC n = 10 oropharyngeal[30], 2010
PBHNSCC42Flow cytometry: EpCAM, CD45 RT-PCR: EGFR mRNA43CTC and N in flow cytometry (P = 0.014), CTC and N in RT-PCRVarious sites n = 9 OSCC n = 17 oropharyngeal[31], 2011
PBHNSCC15CellSearch™40CTC and M (P = 0.04)Various sites n = 4 OSCC n =7 oropharyngeal[84], 2012
PBHNSCC31Flow cytometry: EpCAM; EGFR, CD4529 pEGFR in 55% of CTC+-casesIncreasing number of CTC-positive cases at end of treatmentVarious sites n =5 OSCC n =18 oropharyngeal[50], 2012
PBUnder RCT HNSCC33Flow cytometry: EpCAM CD45 keratins 7, 8 EGFR33 pEGFR in 36.4% of CTC+-casesNone given for comparing two methodsVarious sites n = 0 OSCC (not specified) n = 13 oropharyngeal[51], 2012
PBHNSCC73CellSearch™15.1Decreasing CTC counts and clinical response (P = 0.017)Various sites n = 3 OSCC n = 39 oropharyngeal[82], 2012
BMHNSCC105ICC: keratins 8, 18 and 1914.5No significance for DTCs (76/105 tested)Various sites n = 72 OSCC n = 8 oropharyngeal[68], 2012
PB BMOSCC110CellSearch™ ICC: keratins 8, 18 and 1912.5 (Cell search)20.0 (ICC)CTCs and T (P = 0.04), N and DTCs (P = 0.02), M and CTCs (P = 0.004), M and DTCs (P = 0.005), DTCs and RFS (P < 0.001)OSCC only[35], 2014

With reference to the concept of field cancerisation (FC) introduced by Slaughter et al[64] in the 1950’s, related problems in OSCC should also be considered because locoregional failure can lead to distant metastasis in HNSCC[65]. FC is thought to be undetectable by routine procedures thereby potentially causing a second primary tumour[66]. Furthermore, the entire mucous membrane of the respiratory and upper digestive tract is at risk for neoplasia[67]. Graveland et al[68] investigated the potential association of minimal residual cancer in deep surgical margins and DTCs in BM aspirates with clinico-histopathological parameters and outcomes and concluded that the presence of vasoinvasive and infiltrative growth in HNSCC is a significant risk factor for locoregional recurrence and disease-free survival. Furthermore, they concluded that, at present, there seemed to be no role for the molecular analysis of deep surgical margins and BM aspirates in predicting outcomes with the methods used for 105 HNSCC patients enrolled in this study. To improve histopathological staging in previously tumour cell-negative margins, the overexpression of p53 frequently found in primary tumours was revealed to be indicative of an increased risk of local recurrence[69,70]. Clinically relevant tumour cell spread is rarely found beyond the level of histopathological detection. Thus, a molecular assessment of minimal residual disease is needed to fill the gap between histopathological findings and outcomes in HNSCC, including OSCC[71]. Several serum tumour markers have been evaluated with regard to clinical value in HNSCC and OSCC, but most of them demonstrated poor sensitivity[72,73]. This article focuses on the potential haematogenous spread of tumour cells while OSCC most often spreads in lymphatic vessels[74-76], potentially decreasing the relevance regarding OSCC/HNSCC. However, there are increasing signs that CTCs are clinically relevant in the pathogenesis and recurrence of head and neck tumours. The total number of suspicious cells most likely depends on the method applied. OSCC is characterised by specific expression patterns for different keratins, capable of being detected in CTCs/DTCs[77], leading to technical limitations that must be considered. The detection methods have been optimised for CTCs or DTCs derived from adenocarcinomas such as breast, prostate, colorectal or hepatocellular carcinomas, as reported by Schulze et al[78] in the latter case. Therefore, an adaption to the unique biology of OSCC may be required.

Nevertheless, one of the first reports of prognostic value in a homogenous group of OSCC has been recently published by Gröbe et al[35], which describes a strong and independent prognostic impact of CTCs and DTCs for disease-free survival, which outperformed current prognosticators. This finding might provide evidence that patients who are positive for tumour cells in the BM or PB may suffer from a more aggressive disease. Thus, CTCs/DTCs detected in OSCC patients might serve as prognostic markers, predicting relapse at various disease stages, supplementing the current routine staging procedures. In this study, 110 patients were enrolled. Anti-keratin immunostaining after Ficoll density gradient centrifugation to detect DTCs and applying the CellSearch™-System to analyse PB represent techniques with the current highest degree of international standardisation[42,44]. Nineteen patients (21%) exhibited DTCs in the BM, while 11 patients (13.6%) were positive for CTCs in the PB. According to other literature data, CTCs/DTCs are traceable in the PB of HNSCC patients[29,31]. Wollenberg et al[29], for example, detected DTCs from BM aspirates, depending on tumour stage.

Gröbe et al[35] found no significant correlation between the detection of CTCs and the presence of DTCs (P = 0.68), suggesting that both analyses might provide supplementary results. Distant haematogenous metastases were less frequently observed compared to locoregional relapses. Consistent with other studies examining more heterogeneous groups of HNSCC patients[29,79,80], the subsequent development of recurrent disease was observed more frequently when patients were positive for CTCs/DTCs[35,81]. However, there are several incongruous and moderately significant results demonstrating that the higher the disease stage, the higher the yield of detected CTCs. Although statistically not significant, a trend regarding this conclusion was shown by Buglione et al[82] last year, comparing T1 to higher tumour stages in HNSCC, but in the OSCC subgroup (13/152 patients), no CTCs were found. Previously, Guney et al[83] found similar results for HNSCC but in a smaller cohort of patients. A correlation to tumour stage was found, though not significant for DTCs, and the total number of CTCs was higher in T3-4 tumours compared to T1-2 tumours[29]. These findings suggest that the size of the primary tumour is not always prognostically relevant. This hypothesis is strengthened by significantly higher incidences of CTC positivity in patients with locally advanced disease when considering the N-stage, holding true even in a multivariate analysis[31]. Moreover, CTC detection was significantly associated with the presence of lung nodules[84]. The tumour mass limits local resection by potentially decreasing safety margins because of neighbouring structures, such as the sinus at the skull base, which reaffirms the problem of an assumed CF. Nevertheless, a longer disease-free survival of patients without detectable CTCs compared to CTC-positive patients was stated by Jatana et al[32]. Unfortunately, most of the studies cited were performed on small patient cohorts for other groups of HNSCC[83,84]. In a distinctly larger group of 176 HNSCC patients, the presence of DTCs in BM showed prognostic relevance for overall survival. In half of the DTC-positive patients, the disease recurred, whereas in the total cohort, recurrences were only observed in 27% of the patients[29].

The importance of CTC detection in the context of therapy is still controversially discussed. For OSCC patients, there are studies investigating the dissemination of tumour cells after surgery and tumour cell spread due to surgical procedures[32,85], but among these studies, only a few have investigated the impact of metastases and survival regarding this issue. These facts are crucial in OSCC as surgery is the current gold standard and primary treatment option.

Before, during and following radiotherapy the detection of keratin-19-positive cells coincided with local failure, distant metastasis and moreover, the diagnosis of anaemia[79]. Regarding the therapeutic response in HNSCC with decreased levels of previously detected CTCs, a positive effect on outcome was presumed by Buglione et al[82] because partial or complete response could be stated along with these findings.

A fact that might impair the analysis of detected CTCs/DTCs is the subjective evaluation by even experienced independent readers of common immunocytochemical methods, such as the use of the anti-keratin antibody A45-B/B3. However, false-positive results are seldom found[86]. The criteria to evaluate the morphology of potential CTCs and immunostaining reactions are crucial in defining comparable results[39,42]. This knowledge is even important to evaluate results obtained for blood samples analysed with the semi-automated, highly standardised CellSearch system. To overcome these problems, different international ring studies have begun to reduce interobserver variability of the results[44,87]. The successful CellSearch system has proven its usefulness in OSCC/HNSCC, although not in all advanced stage patients. However, even in metastasised, advanced disease, CTCs were not detectable in all cases[35]. Nichols et al[84] detected almost 100% of spiked HNSCC cell line cells (FaDu) using the CellSearch system, however only 6 out of 15 HNSCC patients tested CTC-positive using this approach. The results are in accordance with those of other studies but suggest that, in patients, either the incidence or number of potentially detectable CTCs is very low or that not all present CTCs are detectable with the currently applied approaches. Furthermore, the system is ruling out non-EpCAM and/or non-keratin expressing CTCs. This escape from detection has been previously demonstrated for breast cancer where, in aggressive tumours, EpCAM expression might be down-regulated, and this finding has also been assumed for aggressive forms of solid cancers other than OSCC[88].

Hence, serious consideration should be given to EMT, which is associated with a loss of epithelial features, enhancing the migratory capability due to altered plasticity[88,89]. Stem cell-like cells seem to be most notably affected, as dynamic changes in these cells were identified by Mani et al[90] and Yu et al[91]. Thus, increasingly false-negative results might correlate to EMT[92]. The detection of EpCAM-negative cells might be possible by using other surface markers, such as N-cadherin, EGFR or cancer stem cell markers, such as CD44[89,93,94]. In colorectal carcinomas, plastin3 was identified as a potential target for cells undergoing EMT because its expression is not altered during this process[95]. Superiority over EpCAM-based methods must be proven, and expression of plastin3 in cancers other than colorectal cancer must be analysed. EGFR, frequently overexpressed in HNSCC, is a meaningful target for CTC enrichment[50,96]. Due to the biological heterogeneity of CTCs/DTCs, the most aggressive subsets initiating metastasis must still be categorised[97,98]. In OSCC, Nagata et al[99] performed molecular analyses, identifying integrins as potential biomarkers for the risk of locoregional and haematogenous dissemination. The role of the expression rates of integrin alpha-3 for locoregional and ITGB4 for haematogenous dissemination and, therefore, cancer cell motility and anchorage-independent survival, which are vital for OSCC recurrence and metastasis, has previously been shown. Furthermore, the importance of integrin beta1 in HNSCC and stimulated vascular endothelial growth factor secretion by alphaB-crystallin in HNSCC was specified[100,101].

In addition to these considerations, there might be a spread of tumour-derived molecules on a subcellular level, such as micro-RNAs. Their role in the formation of metastases derived from OSCC is currently unclear[102,103].

According to current hypotheses, most DTCs remain in a dormant state and probably never initiate a relapse or metastasis[56,104], whereas others return to a proliferating state by still unknown molecular alterations or changes in environmental conditions[105,106]. Interestingly, also a re-circulation from the dormant site back to the primary location of the cancer has been reported in an mouse breast cancer model[107], but because of the high rate of locoregional failure, the concept does not seem apparent for OSCC in the reviewers opinion due to the concept of FC[67]. Therefore, more obvious challenges must be faced when considering of this phenomenon. The spread of tumour cells during surgery remains unclear but has been proven for fine needle aspiration or incision biopsy in OSCC[85,108]. A total of 71% of 48 patients harboured CTCs detected by Jatana et al[30] in HNSCC. Here, samples were taken at the time of surgery. Circulating epithelial cells also can be found in non-malignant diseases, such as inflammatory diseases of the colon, with the current assays[109].

Nevertheless, the appealing potential of detecting CTCs/DTCs as prognostic markers in OSCC from non-invasively accessible blood samples must be emphasised. When occult tumour cells are ubiquitously spread, PB may be a way to detect these cells, facilitating a “liquid biopsy”[37]. Out of a set of several markers, a decision must be made, and the occurrence of CTCs is a very rare event. Researchers are faced with the problem that often processed samples cannot be used a second time for other methods, and the samples display only a cross section of the entire circulating blood. A reliable, morphological identification, for example, with fixation of the cells and nuclear staining might prevent subsequent molecular characterisation. These analyses provide just a snapshot of tumour cell dissemination in contrast to BM, which may represent a reservoir where DTCs can be collected over a longer period of time[104]. Many CTCs are assumed to undergo apoptosis quickly[110], as the lifespan of CTCs in the circulation is estimated to be only between one and two hours[111]. Alternative approaches, such as the “CTC-Chip”, punctuate an improvement in CTC yield and purity but only give a possible outlook due to the small number of examined cohorts and have not been tested for OSCC yet[47,112].

To identify potential targets for individualised therapies and to use repeated CTC assessments in individual patients for treatment surveillance is of the utmost importance[37]. A potential marker in breast cancer is the human epidermal growth factor receptor 2 (HER2), the expression of which can also be monitored on CTCs, thus giving patients with HER2-negative primary tumours but HER2-positive CTCs a chance to be treated by a HER2-targeted therapy[113,114]. Currently, there is no biomarker available for OSCC yet. BM might be an interesting target organ for therapeutic interventions by considering DTCs as therapeutic targets. The data described here point to the potential future utility of drugs targeting DTCs in the BM of OSCC patients. The present high expenses will decrease when evidence is found that these methods are suitable for OSCC and can be included in standard protocols for tumour staging.

CONCLUSION

A set of promising techniques for the detection of CTCs/DTCs has been developed in the past decade, proving their usefulness in solid cancers with predominantly haematogenous dissemination. Less invasive diagnostic tools might be appropriate to refine the current staging methods. The non-invasiveness of collecting samples from PB and obtaining BM specimens might provide additional treatment options in patients suffering from OSCC. For OSCC, the current data show that CTCs/DTCs might have a clinical impact on the prognosis and survival, even outperforming current prognosticators. Circulating tumor cells and DTCs must be considered independent markers in OSCC as a subgroup of head and neck tumours, but the ongoing debate remains too controversial to be considered evidence. The findings presented in this review are not convincing for oral cancer in all aspects yet. Nevertheless, the potential is striking, guiding therapy towards individual-centred medicine with the possibility of early intervention in this severe disease. Despite encouraging results, further investigation of the clinical relevance of CTCs and DTCs is needed as validation in large multicentre trials with synchronised measuring methods and defined outcomes and time points is lacking. In particular, the risk of an early locoregional relapse is difficult to assess because of different problems in the field of HNSCC and OSCC. It is important to continue investigations in homogenous cohorts to decrease the still unaltered morbidity in patients suffering from OSCC. Due to being one of the most active areas of translational cancer research, the technologies presented herein offer a number of compelling advantages. The enumeration of CTCs and DTCs as biomarkers to predict tumour relapse may become feasible in the near future, but researchers in this extraordinarily dynamic field are still facing technical and clinical hurdles. In addition to further improvements, there are challenges beyond the mere detection of CTCs/DTCs. In particular, the detection of carcinoma cells undergoing EMT, a down-regulation of epithelial features when entering vessels, the transient circulation of cells, the mechanisms behind leaving the “homing organs” and the states of dormancy need to be investigated. Whether EpCAM- or keratin-positive cells detected by the described techniques are involved in initiating metastasis is also controversially discussed. To address this question further, molecular analysis of CTCs/DTCs is needed.

A profound understanding of OSCC as a systemic disease supported by molecular investigation of CTCs/DTCs gives new perspectives on the disease and provides the chance to identify potential targets for individualised therapies. Repeated CTC assessments might offer treatment surveillance in individual patients prospectively and might predict local and systemic relapse with a higher sensitivity at various disease stages compared to routine staging procedures in OSCC.

Footnotes

P- Reviewers: Chui YL, Msaouel P, Munoz M, Radoauane Y S- Editor: Zhai HH L- Editor: A E- Editor: Liu SQ

References
1.  Parkin DM, Bray F, Ferlay J, Pisani P. Estimating the world cancer burden: Globocan 2000. Int J Cancer. 2001;94:153-156.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Haddad RI, Shin DM. Recent advances in head and neck cancer. N Engl J Med. 2008;359:1143-1154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 699]  [Cited by in F6Publishing: 680]  [Article Influence: 42.5]  [Reference Citation Analysis (0)]
3.  Barnes L World health organisation classification of tumours, pathology and genetics, head and neck tumours. Lyon: IARC Press 2005; .  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Kademani D, Bell RB, Bagheri S, Holmgren E, Dierks E, Potter B, Homer L. Prognostic factors in intraoral squamous cell carcinoma: the influence of histologic grade. J Oral Maxillofac Surg. 2005;63:1599-1605.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Zbären P, Lehmann W. Frequency and sites of distant metastases in head and neck squamous cell carcinoma. An analysis of 101 cases at autopsy. Arch Otolaryngol Head Neck Surg. 1987;113:762-764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 113]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
6.  Slootweg PJ, Bolle CW, Koole R, Hordijk GJ. Cause of death in squamous cell carcinoma of the head and neck. An autopsy study on 31 patients. J Craniomaxillofac Surg. 1992;20:225-227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
7.  Wheeler SE, Shi H, Lin F, Dasari S, Bednash J, Thorne S, Watkins S, Joshi R, Thomas SM. Enhancement of head and neck squamous cell carcinoma proliferation, invasion, and metastasis by tumor-associated fibroblasts in preclinical models. Head Neck. 2014;36:385-392.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Shah JP. Patterns of cervical lymph node metastasis from squamous carcinomas of the upper aerodigestive tract. Am J Surg. 1990;160:405-409.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 601]  [Cited by in F6Publishing: 502]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
9.  Forastiere A, Koch W, Trotti A, Sidransky D. Head and neck cancer. N Engl J Med. 2001;345:1890-1900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 974]  [Cited by in F6Publishing: 1009]  [Article Influence: 43.9]  [Reference Citation Analysis (0)]
10.  Yongkui L, Jian L, Wanghan L. 18FDG-PET/CT for the detection of regional nodal metastasis in patients with primary head and neck cancer before treatment: a meta-analysis. Surg Oncol. 2013;22:e11-e16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 32]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
11.  Teymoortash A, Werner JA. Current advances in diagnosis and surgical treatment of lymph node metastasis in head and neck cancer. GMS Curr Top Otorhinolaryngol Head Neck Surg. 2012;11:Doc04.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Cohen EE, Lingen MW, Vokes EE. The expanding role of systemic therapy in head and neck cancer. J Clin Oncol. 2004;22:1743-1752.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Cooper JS, Pajak TF, Forastiere AA, Jacobs J, Campbell BH, Saxman SB, Kish JA, Kim HE, Cmelak AJ, Rotman M. Postoperative concurrent radiotherapy and chemotherapy for high-risk squamous-cell carcinoma of the head and neck. N Engl J Med. 2004;350:1937-1944.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Partridge M. Head and neck cancer and precancer: can we use molecular genetics to make better predictions? Ann R Coll Surg Engl. 1999;81:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Psychogios G, Mantsopoulos K, Bohr C, Koch M, Zenk J, Iro H. Incidence of occult cervical metastasis in head and neck carcinomas: development over time. J Surg Oncol. 2013;107:384-387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 32]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
16.  Jin YT, Myers J, Tsai ST, Goepfert H, Batsakis JG, el-Naggar AK. Genetic alterations in oral squamous cell carcinoma of young adults. Oral Oncol. 1999;35:251-256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 40]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
17.  Jin C, Jin Y, Wennerberg J, Akervall J, Dictor M, Mertens F. Karyotypic heterogeneity and clonal evolution in squamous cell carcinomas of the head and neck. Cancer Genet Cytogenet. 2002;132:85-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 55]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
18.  Zhang Z, Shi Q, Wang LE, Sturgis EM, Spitz MR, El-Naggar AK, Hong WK, Wei Q. No Association between hOGG1 Ser326Cys polymorphism and risk of squamous cell carcinoma of the head and neck. Cancer Epidemiol Biomarkers Prev. 2004;13:1081-1083.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Li G, Sturgis EM, Wang LE, Chamberlain RM, Amos CI, Spitz MR, El-Naggar AK, Hong WK, Wei Q. Association of a p73 exon 2 G4C14-to-A4T14 polymorphism with risk of squamous cell carcinoma of the head and neck. Carcinogenesis. 2004;25:1911-1916.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 72]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
20.  Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, Reuben JM, Doyle GV, Allard WJ, Terstappen LW. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;351:781-791.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3360]  [Cited by in F6Publishing: 3257]  [Article Influence: 162.9]  [Reference Citation Analysis (0)]
21.  Cohen SJ, Punt CJ, Iannotti N, Saidman BH, Sabbath KD, Gabrail NY, Picus J, Morse M, Mitchell E, Miller MC. Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26:3213-3221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1354]  [Cited by in F6Publishing: 1353]  [Article Influence: 84.6]  [Reference Citation Analysis (0)]
22.  de Bono JS, Attard G, Adjei A, Pollak MN, Fong PC, Haluska P, Roberts L, Melvin C, Repollet M, Chianese D. Potential applications for circulating tumor cells expressing the insulin-like growth factor-I receptor. Clin Cancer Res. 2007;13:3611-3616.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Shaffer DR, Leversha MA, Danila DC, Lin O, Gonzalez-Espinoza R, Gu B, Anand A, Smith K, Maslak P, Doyle GV. Circulating tumor cell analysis in patients with progressive castration-resistant prostate cancer. Clin Cancer Res. 2007;13:2023-2029.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Danila DC, Heller G, Gignac GA, Gonzalez-Espinoza R, Anand A, Tanaka E, Lilja H, Schwartz L, Larson S, Fleisher M. Circulating tumor cell number and prognosis in progressive castration-resistant prostate cancer. Clin Cancer Res. 2007;13:7053-7058.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Effenberger KE, Schroeder C, Eulenburg C, Reeh M, Tachezy M, Riethdorf S, Vashist YK, Izbicki JR, Pantel K, Bockhorn M. Disseminated tumor cells in pancreatic cancer-an independent prognosticator of disease progression and survival. Int J Cancer. 2012;131:E475-E483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
26.  Fehm T, Banys M, Rack B, Janni W, Marth C, Blassl C, Hartkopf A, Trope C, Kimmig R, Krawczyk N. Pooled analysis of the prognostic relevance of disseminated tumor cells in the bone marrow of patients with ovarian cancer. Int J Gynecol Cancer. 2013;23:839-845.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
27.  Wallwiener M, Hartkopf AD, Baccelli I, Riethdorf S, Schott S, Pantel K, Marmé F, Sohn C, Trumpp A, Rack B. The prognostic impact of circulating tumor cells in subtypes of metastatic breast cancer. Breast Cancer Res Treat. 2013;137:503-510.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 68]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
28.  Wang G, Wang S, Li Y, Yu Y, Song Z, Zhao Q, Tian Z. Clinical study of disseminated tumor cells in bone marrow of patients with gastric cancer. Hepatogastroenterology. 2013;60:273-276.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Wollenberg B, Walz A, Kolbow K, Pauli C, Chaubal S, Andratschke M. Clinical relevance of circulating tumour cells in the bone marrow of patients with SCCHN. Onkologie. 2004;27:358-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 19]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
30.  Jatana KR, Balasubramanian P, Lang JC, Yang L, Jatana CA, White E, Agrawal A, Ozer E, Schuller DE, Teknos TN. Significance of circulating tumor cells in patients with squamous cell carcinoma of the head and neck: initial results. Arch Otolaryngol Head Neck Surg. 2010;136:1274-1279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 91]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
31.  Hristozova T, Konschak R, Stromberger C, Fusi A, Liu Z, Weichert W, Stenzinger A, Budach V, Keilholz U, Tinhofer I. The presence of circulating tumor cells (CTCs) correlates with lymph node metastasis in nonresectable squamous cell carcinoma of the head and neck region (SCCHN). Ann Oncol. 2011;22:1878-1885.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Jatana KR, Lang JC, Chalmers JJ. Identification of circulating tumor cells: a prognostic marker in squamous cell carcinoma of the head and neck? Future Oncol. 2011;7:481-484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
33.  Lim AM, Corry J, Collins M, Peters L, Hicks RJ, D’Costa I, Coleman A, Chua M, Solomon B, Rischin D. A phase II study of induction carboplatin and gemcitabine followed by chemoradiotherapy for the treatment of locally advanced nasopharyngeal carcinoma. Oral Oncol. 2013;49:468-474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
34.  Ferlito A, Thompson LD, Cardesa A, Gnepp DR, Devaney KO, Rodrigo JP, Hunt JL, Rinaldo A, Takes RP. The importance of histological types for treatment and prognosis in laryngeal cancer. Eur Arch Otorhinolaryngol. 2013;270:401-403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 7]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
35.  Gröbe A, Blessmann M, Hanken H, Friedrich RE, Schön G, Wikner J, Effenberger KE, Kluwe L, Heiland M, Pantel K. Prognostic relevance of circulating tumor cells in blood and disseminated tumor cells in bone marrow of patients with squamous cell carcinoma of the oral cavity. Clin Cancer Res. 2014;20:425-433.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Pantel K, Alix-Panabières C. Detection methods of circulating tumor cells. J Thorac Dis. 2012;4:446-447.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Alix-Panabières C, Pantel K. Circulating tumor cells: liquid biopsy of cancer. Clin Chem. 2013;59:110-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 748]  [Cited by in F6Publishing: 795]  [Article Influence: 72.3]  [Reference Citation Analysis (2)]
38.  Pantel K, Alix-Panabières C. Real-time liquid biopsy in cancer patients: fact or fiction? Cancer Res. 2013;73:6384-6388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 290]  [Cited by in F6Publishing: 316]  [Article Influence: 28.7]  [Reference Citation Analysis (0)]
39.  Borgen E, Naume B, Nesland JM, Kvalheim G, Beiske K, Fodstad O, Diel I, Solomayer EF, Theocharous P, Coombes RC. Standardization of the immunocytochemical detection of cancer cells in BM and blood: I. establishment of objective criteria for the evaluation of immunostained cells. Cytotherapy. 1999;1:377-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 168]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
40.  Solá M, Margelí M, Castellá E, Cirauqui B, Mariscal A, Rull M, Julian JF, Luna M, Vallejo V, Fraile M. Detection of disseminated tumor cells in locally advanced breast cancer patients before primary systemic therapy. Breast. 2013;22:908-913.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
41.  Braun S, Vogl FD, Naume B, Janni W, Osborne MP, Coombes RC, Schlimok G, Diel IJ, Gerber B, Gebauer G. A pooled analysis of bone marrow micrometastasis in breast cancer. N Engl J Med. 2005;353:793-802.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1017]  [Cited by in F6Publishing: 943]  [Article Influence: 49.6]  [Reference Citation Analysis (0)]
42.  Fehm T, Braun S, Muller V, Janni W, Gebauer G, Marth C, Schindlbeck C, Wallwiener D, Borgen E, Naume B. A concept for the standardized detection of disseminated tumor cells in bone marrow from patients with primary breast cancer and its clinical implementation. Cancer. 2006;107:885-892.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 176]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
43.  Hayes DF, Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Miller MC, Matera J, Allard WJ, Doyle GV, Terstappen LW. Circulating tumor cells at each follow-up time point during therapy of metastatic breast cancer patients predict progression-free and overall survival. Clin Cancer Res. 2006;12:4218-4224.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Riethdorf S, Fritsche H, Müller V, Rau T, Schindlbeck C, Rack B, Janni W, Coith C, Beck K, Jänicke F. Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: a validation study of the CellSearch system. Clin Cancer Res. 2007;13:920-928.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Sastre J, Maestro ML, Puente J, Veganzones S, Alfonso R, Rafael S, García-Saenz JA, Vidaurreta M, Martín M, Arroyo M. Circulating tumor cells in colorectal cancer: correlation with clinical and pathological variables. Ann Oncol. 2008;19:935-938.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Gorges TM, Pantel K. Circulating tumor cells as therapy-related biomarkers in cancer patients. Cancer Immunol Immunother. 2013;62:931-939.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 79]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
47.  Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D, Ulkus L, Smith MR, Kwak EL, Digumarthy S, Muzikansky A. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature. 2007;450:1235-1239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2735]  [Cited by in F6Publishing: 2490]  [Article Influence: 155.6]  [Reference Citation Analysis (0)]
48.  Stott SL, Lee RJ, Nagrath S, Yu M, Miyamoto DT, Ulkus L, Inserra EJ, Ulman M, Springer S, Nakamura Z. Isolation and characterization of circulating tumor cells from patients with localized and metastatic prostate cancer. Sci Transl Med. 2010;2:25ra23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 379]  [Cited by in F6Publishing: 404]  [Article Influence: 28.9]  [Reference Citation Analysis (0)]
49.  Wang X, Qian X, Beitler JJ, Chen ZG, Khuri FR, Lewis MM, Shin HJ, Nie S, Shin DM. Detection of circulating tumor cells in human peripheral blood using surface-enhanced Raman scattering nanoparticles. Cancer Res. 2011;71:1526-1532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 275]  [Cited by in F6Publishing: 229]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
50.  Tinhofer I, Hristozova T, Stromberger C, Keilhoiz U, Budach V. Monitoring of circulating tumor cells and their expression of EGFR/phospho-EGFR during combined radiotherapy regimens in locally advanced squamous cell carcinoma of the head and neck. Int J Radiat Oncol Biol Phys. 2012;83:e685-e690.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 54]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
51.  Hristozova T, Konschak R, Budach V, Tinhofer I. A simple multicolor flow cytometry protocol for detection and molecular characterization of circulating tumor cells in epithelial cancers. Cytometry A. 2012;81:489-495.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 43]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
52.  Eifler RL, Lind J, Falkenhagen D, Weber V, Fischer MB, Zeillinger R. Enrichment of circulating tumor cells from a large blood volume using leukapheresis and elutriation: proof of concept. Cytometry B Clin Cytom. 2011;80:100-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 58]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
53.  Fischer JC, Niederacher D, Topp SA, Honisch E, Schumacher S, Schmitz N, Zacarias Föhrding L, Vay C, Hoffmann I, Kasprowicz NS. Diagnostic leukapheresis enables reliable detection of circulating tumor cells of nonmetastatic cancer patients. Proc Natl Acad Sci USA. 2013;110:16580-16585.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 158]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
54.  Saucedo-Zeni N, Mewes S, Niestroj R, Gasiorowski L, Murawa D, Nowaczyk P, Tomasi T, Weber E, Dworacki G, Morgenthaler NG. A novel method for the in vivo isolation of circulating tumor cells from peripheral blood of cancer patients using a functionalized and structured medical wire. Int J Oncol. 2012;41:1241-1250.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Alix-Panabières C, Vendrell JP, Pellé O, Rebillard X, Riethdorf S, Müller V, Fabbro M, Pantel K. Detection and characterization of putative metastatic precursor cells in cancer patients. Clin Chem. 2007;53:537-539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 136]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
56.  Pantel K, Brakenhoff RH, Brandt B. Detection, clinical relevance and specific biological properties of disseminating tumour cells. Nat Rev Cancer. 2008;8:329-340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 883]  [Cited by in F6Publishing: 927]  [Article Influence: 57.9]  [Reference Citation Analysis (0)]
57.  Sieuwerts AM, Kraan J, Bolt-de Vries J, van der Spoel P, Mostert B, Martens JW, Gratama JW, Sleijfer S, Foekens JA. Molecular characterization of circulating tumor cells in large quantities of contaminating leukocytes by a multiplex real-time PCR. Breast Cancer Res Treat. 2009;118:455-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 154]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
58.  Wicha MS, Hayes DF. Circulating tumor cells: not all detected cells are bad and not all bad cells are detected. J Clin Oncol. 2011;29:1508-1511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 139]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
59.  Mostert B, Kraan J, Sieuwerts AM, van der Spoel P, Bolt-de Vries J, Prager-van der Smissen WJ, Smid M, Timmermans AM, Martens JW, Gratama JW. CD49f-based selection of circulating tumor cells (CTCs) improves detection across breast cancer subtypes. Cancer Lett. 2012;319:49-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
60.  Heitzer E, Auer M, Gasch C, Pichler M, Ulz P, Hoffmann EM, Lax S, Waldispuehl-Geigl J, Mauermann O, Lackner C. Complex tumor genomes inferred from single circulating tumor cells by array-CGH and next-generation sequencing. Cancer Res. 2013;73:2965-2975.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 406]  [Cited by in F6Publishing: 433]  [Article Influence: 39.4]  [Reference Citation Analysis (0)]
61.  Gasch C, Bauernhofer T, Pichler M, Langer-Freitag S, Reeh M, Seifert AM, Mauermann O, Izbicki JR, Pantel K, Riethdorf S. Heterogeneity of epidermal growth factor receptor status and mutations of KRAS/PIK3CA in circulating tumor cells of patients with colorectal cancer. Clin Chem. 2013;59:252-260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 182]  [Cited by in F6Publishing: 183]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
62.  Gröbe A, Hanken H, Kluwe L, Schöllchen M, Tribius S, Pohlenz P, Clauditz T, Grob T, Simon R, Sauter G. Immunohistochemical analysis of p16 expression, HPV infection and its prognostic utility in oral squamous cell carcinoma. J Oral Pathol Med. 2013;42:676-681.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Turksma AW, Braakhuis BJ, Bloemena E, Meijer CJ, Leemans CR, Hooijberg E. Immunotherapy for head and neck cancer patients: shifting the balance. Immunotherapy. 2013;5:49-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
64.  Slaughter DP, Southwick HW, Smejkal W. Field cancerization in oral stratified squamous epithelium; clinical implications of multicentric origin. Cancer. 1953;6:963-968.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 13]  [Reference Citation Analysis (0)]
65.  Dragovic AF, Caudell JJ, Spencer SA, Carroll WR, Nabell LA, Bonner JA. Locoregional failure and the risk of distant metastasis after modern radiotherapy for head and neck cancer. Head Neck. 2013;35:381-387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
66.  Braakhuis BJ, Tabor MP, Kummer JA, Leemans CR, Brakenhoff RH. A genetic explanation of Slaughter’s concept of field cancerization: evidence and clinical implications. Cancer Res. 2003;63:1727-1730.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Angadi PV, Savitha JK, Rao SS, Sivaranjini Y. Oral field cancerization: current evidence and future perspectives. Oral Maxillofac Surg. 2012;16:171-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 68]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
68.  Graveland AP, Braakhuis BJ, Eerenstein SE, de Bree R, Bloemena E, de Maaker M, van den Brekel MW, Dijk F, Mesker WE, Tanke HJ. Molecular diagnosis of minimal residual disease in head and neck cancer patients. Cell Oncol (Dordr). 2012;35:367-375.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
69.  Brennan JA, Mao L, Hruban RH, Boyle JO, Eby YJ, Koch WM, Goodman SN, Sidransky D. Molecular assessment of histopathological staging in squamous-cell carcinoma of the head and neck. N Engl J Med. 1995;332:429-435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 526]  [Cited by in F6Publishing: 489]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
70.  van Houten VM, Tabor MP, van den Brekel MW, Denkers F, Wishaupt RG, Kummer JA, Snow GB, Brakenhoff RH. Molecular assays for the diagnosis of minimal residual head-and-neck cancer: methods, reliability, pitfalls, and solutions. Clin Cancer Res. 2000;6:3803-3816.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Gath HJ, Brakenhoff RH. Minimal residual disease in head and neck cancer. Cancer Metastasis Rev. 1999;18:109-126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 43]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
72.  Seoane Lestón J, Diz Dios P. Diagnostic clinical aids in oral cancer. Oral Oncol. 2010;46:418-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 59]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
73.  Blessmann M, Kaifi JT, Schurr PG, Cihan A, Kalinin V, Trump F, Atac A, Heiland M, Pohlenz P, Blake F. Short tandem repeat polymorphism in exon 4 of esophageal cancer related gene 2 predicts relapse of oral squamous cell carcinoma. Oral Oncol. 2008;44:143-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
74.  Stephan K H, Sandro J S. SPECT/CT for Lymphatic Mapping of Sentinel Nodes in Early Squamous Cell Carcinoma of the Oral Cavity and Oropharynx. Int J Mol Imaging. 2011;2011:106068.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 17]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
75.  Michikawa C, Uzawa N, Kayamori K, Sonoda I, Ohyama Y, Okada N, Yamaguchi A, Amagasa T. Clinical significance of lymphatic and blood vessel invasion in oral tongue squamous cell carcinomas. Oral Oncol. 2012;48:320-324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 49]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
76.  Watanabe S, Kato M, Kotani I, Ryoke K, Hayashi K. Lymphatic Vessel Density and Vascular Endothelial Growth Factor Expression in Squamous Cell Carcinomas of Lip and Oral Cavity: A Clinicopathological Analysis with Immunohistochemistry Using Antibodies to D2-40, VEGF-C and VEGF-D. Yonago Acta Med. 2013;56:29-37.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Moll R, Divo M, Langbein L. The human keratins: biology and pathology. Histochem Cell Biol. 2008;129:705-733.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 896]  [Cited by in F6Publishing: 934]  [Article Influence: 58.4]  [Reference Citation Analysis (0)]
78.  Schulze K, Gasch C, Staufer K, Nashan B, Lohse AW, Pantel K, Riethdorf S, Wege H. Presence of EpCAM-positive circulating tumor cells as biomarker for systemic disease strongly correlates to survival in patients with hepatocellular carcinoma. Int J Cancer. 2013;133:2165-2171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 177]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
79.  Pajonk F, Schlessmann S, Guttenberger R, Henke M. Epithelial cells in the peripheral blood of patients with cancer of the head and neck: incidence, detection and possible clinical significance. Radiother Oncol. 2001;59:213-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
80.  Colnot DR, Nieuwenhuis EJ, Kuik DJ, Leemans CR, Dijkstra J, Snow GB, van Dongen GA, Brakenhoff RH. Clinical significance of micrometastatic cells detected by E48 (Ly-6D) reverse transcription-polymerase chain reaction in bone marrow of head and neck cancer patients. Clin Cancer Res. 2004;10:7827-7833.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Partridge M, Brakenhoff R, Phillips E, Ali K, Francis R, Hooper R, Lavery K, Brown A, Langdon J. Detection of rare disseminated tumor cells identifies head and neck cancer patients at risk of treatment failure. Clin Cancer Res. 2003;9:5287-5294.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Buglione M, Grisanti S, Almici C, Mangoni M, Polli C, Consoli F, Verardi R, Costa L, Paiar F, Pasinetti N. Circulating tumour cells in locally advanced head and neck cancer: preliminary report about their possible role in predicting response to non-surgical treatment and survival. Eur J Cancer. 2012;48:3019-3026.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 67]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
83.  Guney K, Yoldas B, Ozbilim G, Derin AT, Sarihan S, Balkan E. Detection of micrometastatic tumor cells in head and neck squamous cell carcinoma. A possible predictor of recurrences? Saudi Med J. 2007;28:216-220.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Nichols AC, Lowes LE, Szeto CC, Basmaji J, Dhaliwal S, Chapeskie C, Todorovic B, Read N, Venkatesan V, Hammond A. Detection of circulating tumor cells in advanced head and neck cancer using the CellSearch system. Head Neck. 2012;34:1440-1444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 91]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
85.  Kusukawa J, Suefuji Y, Ryu F, Noguchi R, Iwamoto O, Kameyama T. Dissemination of cancer cells into circulation occurs by incisional biopsy of oral squamous cell carcinoma. J Oral Pathol Med. 2000;29:303-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 84]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
86.  Pantel K, Izbicki J, Passlick B, Angstwurm M, Haussinger K, Thetter O, Riethmuller G. Frequency and prognostic significance of isolated tumour cells in bone marrow of patients with non-small-cell lung cancer without overt metastases. Lancet. 1996;347:649-653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 276]  [Cited by in F6Publishing: 290]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
87.  Kraan J, Sleijfer S, Strijbos MH, Ignatiadis M, Peeters D, Pierga JY, Farace F, Riethdorf S, Fehm T, Zorzino L. External quality assurance of circulating tumor cell enumeration using the CellSearch(®) system: a feasibility study. Cytometry B Clin Cytom. 2011;80:112-118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 90]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
88.  Sieuwerts AM, Kraan J, Bolt J, van der Spoel P, Elstrodt F, Schutte M, Martens JW, Gratama JW, Sleijfer S, Foekens JA. Anti-epithelial cell adhesion molecule antibodies and the detection of circulating normal-like breast tumor cells. J Natl Cancer Inst. 2009;101:61-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 349]  [Cited by in F6Publishing: 342]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
89.  Balasubramanian P, Lang JC, Jatana KR, Miller B, Ozer E, Old M, Schuller DE, Agrawal A, Teknos TN, Summers TA. Multiparameter analysis, including EMT markers, on negatively enriched blood samples from patients with squamous cell carcinoma of the head and neck. PLoS One. 2012;7:e42048.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 93]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
90.  Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133:704-715.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6211]  [Cited by in F6Publishing: 6535]  [Article Influence: 408.4]  [Reference Citation Analysis (0)]
91.  Yu M, Bardia A, Wittner BS, Stott SL, Smas ME, Ting DT, Isakoff SJ, Ciciliano JC, Wells MN, Shah AM. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science. 2013;339:580-584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1745]  [Cited by in F6Publishing: 1824]  [Article Influence: 165.8]  [Reference Citation Analysis (0)]
92.  Gorges TM, Tinhofer I, Drosch M, Röse L, Zollner TM, Krahn T, von Ahsen O. Circulating tumour cells escape from EpCAM-based detection due to epithelial-to-mesenchymal transition. BMC Cancer. 2012;12:178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 393]  [Cited by in F6Publishing: 405]  [Article Influence: 33.8]  [Reference Citation Analysis (0)]
93.  Mikolajczyk SD, Millar LS, Tsinberg P, Coutts SM, Zomorrodi M, Pham T, Bischoff FZ, Pircher TJ. Detection of EpCAM-Negative and Cytokeratin-Negative Circulating Tumor Cells in Peripheral Blood. J Oncol. 2011;2011:252361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 181]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
94.  Yao X, Labelle M, Lamb CR, Dugan JM, Williamson CA, Spencer DR, Christ KR, Keating RO, Lee WD, Paradis GA. Determination of 35 cell surface antigen levels in malignant pleural effusions identifies CD24 as a marker of disseminated tumor cells. Int J Cancer. 2013;133:2925-2933.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
95.  Yokobori T, Iinuma H, Shimamura T, Imoto S, Sugimachi K, Ishii H, Iwatsuki M, Ota D, Ohkuma M, Iwaya T. Plastin3 is a novel marker for circulating tumor cells undergoing the epithelial-mesenchymal transition and is associated with colorectal cancer prognosis. Cancer Res. 2013;73:2059-2069.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in F6Publishing: 182]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
96.  Gröbe A, Eichhorn W, Fraederich M, Kluwe L, Vashist Y, Wikner J, Smeets R, Simon R, Sauter G, Heiland M. Immunohistochemical and FISH analysis of EGFR and its prognostic value in patients with oral squamous cell carcinoma. J Oral Pathol Med. 2014;43:205-210.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Stoecklein NH, Hosch SB, Bezler M, Stern F, Hartmann CH, Vay C, Siegmund A, Scheunemann P, Schurr P, Knoefel WT. Direct genetic analysis of single disseminated cancer cells for prediction of outcome and therapy selection in esophageal cancer. Cancer Cell. 2008;13:441-453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 178]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
98.  Wikman H, Vessella R, Pantel K. Cancer micrometastasis and tumour dormancy. APMIS. 2008;116:754-770.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 120]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
99.  Nagata M, Noman AA, Suzuki K, Kurita H, Ohnishi M, Ohyama T, Kitamura N, Kobayashi T, Uematsu K, Takahashi K. ITGA3 and ITGB4 expression biomarkers estimate the risks of locoregional and hematogenous dissemination of oral squamous cell carcinoma. BMC Cancer. 2013;13:410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 37]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
100.  Wang D, Müller S, Amin AR, Huang D, Su L, Hu Z, Rahman MA, Nannapaneni S, Koenig L, Chen Z. The pivotal role of integrin β1 in metastasis of head and neck squamous cell carcinoma. Clin Cancer Res. 2012;18:4589-4599.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  van de Schootbrugge C, Bussink J, Span PN, Sweep FC, Grénman R, Stegeman H, Pruijn GJ, Kaanders JH, Boelens WC. αB-crystallin stimulates VEGF secretion and tumor cell migration and correlates with enhanced distant metastasis in head and neck squamous cell carcinoma. BMC Cancer. 2013;13:128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
102.  Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11:426-437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1954]  [Cited by in F6Publishing: 1989]  [Article Influence: 153.0]  [Reference Citation Analysis (0)]
103.  Diaz LA, Williams RT, Wu J, Kinde I, Hecht JR, Berlin J, Allen B, Bozic I, Reiter JG, Nowak MA. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature. 2012;486:537-540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1259]  [Cited by in F6Publishing: 1292]  [Article Influence: 107.7]  [Reference Citation Analysis (0)]
104.  Pantel K, Alix-Panabières C, Riethdorf S. Cancer micrometastases. Nat Rev Clin Oncol. 2009;6:339-351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 488]  [Cited by in F6Publishing: 528]  [Article Influence: 35.2]  [Reference Citation Analysis (0)]
105.  Uhr JW, Pantel K. Controversies in clinical cancer dormancy. Proc Natl Acad Sci USA. 2011;108:12396-12400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 181]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
106.  Joosse SA, Pantel K. Biologic challenges in the detection of circulating tumor cells. Cancer Res. 2013;73:8-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 152]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
107.  Kim MY, Oskarsson T, Acharyya S, Nguyen DX, Zhang XH, Norton L, Massagué J. Tumor self-seeding by circulating cancer cells. Cell. 2009;139:1315-1326.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 926]  [Cited by in F6Publishing: 984]  [Article Influence: 70.3]  [Reference Citation Analysis (0)]
108.  Dyavanagoudar S, Kale A, Bhat K, Hallikerimath S. Reverse transcriptase polymerase chain reaction study to evaluate dissemination of cancer cells into circulation after incision biopsy in oral squamous cell carcinoma. Indian J Dent Res. 2008;19:315-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 21]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
109.  Pantel K, Denève E, Nocca D, Coffy A, Vendrell JP, Maudelonde T, Riethdorf S, Alix-Panabières C. Circulating epithelial cells in patients with benign colon diseases. Clin Chem. 2012;58:936-940.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 201]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
110.  Méhes G, Witt A, Kubista E, Ambros PF. Circulating breast cancer cells are frequently apoptotic. Am J Pathol. 2001;159:17-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 190]  [Cited by in F6Publishing: 200]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
111.  Meng S, Tripathy D, Frenkel EP, Shete S, Naftalis EZ, Huth JF, Beitsch PD, Leitch M, Hoover S, Euhus D. Circulating tumor cells in patients with breast cancer dormancy. Clin Cancer Res. 2004;10:8152-8162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 666]  [Cited by in F6Publishing: 681]  [Article Influence: 35.8]  [Reference Citation Analysis (0)]
112.  Stott SL, Hsu CH, Tsukrov DI, Yu M, Miyamoto DT, Waltman BA, Rothenberg SM, Shah AM, Smas ME, Korir GK. Isolation of circulating tumor cells using a microvortex-generating herringbone-chip. Proc Natl Acad Sci USA. 2010;107:18392-18397.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Riethdorf S, Müller V, Zhang L, Rau T, Loibl S, Komor M, Roller M, Huober J, Fehm T, Schrader I. Detection and HER2 expression of circulating tumor cells: prospective monitoring in breast cancer patients treated in the neoadjuvant GeparQuattro trial. Clin Cancer Res. 2010;16:2634-2645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 383]  [Cited by in F6Publishing: 369]  [Article Influence: 26.4]  [Reference Citation Analysis (0)]
114.  Pierga JY, Bidard FC, Cropet C, Tresca P, Dalenc F, Romieu G, Campone M, Mahier Aït-Oukhatar C, Le Rhun E, Gonçalves A. Circulating tumor cells and brain metastasis outcome in patients with HER2-positive breast cancer: the LANDSCAPE trial. Ann Oncol. 2013;24:2999-3004.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]