81-2

Circulating Tumour Cells in Clinical Practice: Can Lessons Learned From Locally Advanced/Metastatic Breast and Prostate Cancers Guide Their use for other Cancer Types? Lights and Shadows

Michela Buglione, Luca Triggiani, Nadia Pasinetti, Loredana Costa, Sara Pedretti, Marta Maddalo, Paolo Borghetti, Camillo Almici, Salvatore Grisanti, Stefano Maria Magrini

Michela Buglione, Luca Triggiani, Nadia Pasinetti, Loredana Costa, Sara Pedretti, Marta Maddalo, Paolo Borghetti, Stefano Maria Magrini, Radiation Oncology Department, University and Spedali Civili, Brescia; P.le Spedali Civili 1, Brescia, Italy
Camillo Almici, Immunohematology Department, Spedali Civili, Brescia; P.le Spedali Civili 1 – Brescia, Italy
Salvatore Grisanti, Medical Oncology Department, University and Spedali Civili, Brescia; P.le Spedali Civili 1, Brescia, Italy.

Correspondence to: Michela Buglione, Radiation Oncology Department, University and Spedali Civili, Brescia, Piazzale Spedali Civili, 1, Italy.
Email: michela.buglione@unibs.it
Telephone: +39-030-3995271
Fax: +39-030-399601
Received: July 11, 2015
Revised: September 25, 2015
Accepted: September 30, 2015
Published online: December 10, 2015

ABSTRACT

Circulating tumour cells are cells probably involved in different phases of the metastatic process. They can be identified using standardized and repeatable methods and are characterized by EpCAM; Citokeratin 8, 18, 19 and DAPI positivity and CD45 negativity. CTC’s identification in patients with metastatic breast and prostate cancer has been demonstrated to have a prognostic and predictive role. It is not still clear if these cells can be used also in patients with non metastatic breast and prostate cancer, or in patients with different diseases (eg. metastatic and/or locally advanced head and neck cancer) or if they can used to to biologically characterize the disease during its natural history. This review, leaving from the report of the knowledge already consolidated, is devoted to identify the new trends in the research and in the clinical application of CTC, both in locally advanced/localized prostate and breast cancer, and in head and neck cancer.

© 2015 ACT. All rights reserved.

Key words: Circulating tumour cells; Breast cancer; Prostate cancer; Head and neck cancer; Review

Buglione M, Triggiani L, Pasinetti N, Costa L, Pedretti S, Maddalo M, Borghetti P, Almici C, Grisanti S, Magrini SM. Circulating Tumour Cells in Clinical Practice: Can Lessons Learned From Locally Advanced/Metastatic Breast and Prostate Cancers Guide Their use for other Cancer Types? Lights and Shadows. Journal of Tumor 2015; 3(3): 322-342 Available from: URL: http://www.ghrnet.org/index.php/JT/article/view/1506

The cells probably involved in tumour progression

The process of metastatic diffusion of cancer cells is not completely understood. There are a lot of hypotheses and many different, incompletely defined cells are probably involved.

Cancer stem cells (CSC) are characterized by properties conferring them the ability to induce cancer, to survive to the treatment and to produce the regrowth of the tumor both at the primary site and in distant ones[1,2]

Disseminated tumour cells (DTC) have been identified in the bone marrow and may be a marker of minimal residual disease (a concept defined for hematologic neoplasms and not yet directly demonstrated for other solid tumors)[3]

Circulating tumour cells (CTC) probably represent an intermediate step between these two cell types. They can be defined as cells shed from the primary tumour and circulating in the bloodstream. They could be responsible, together with other host factors, of the metastatic process. CTCs are very rarely picked up in the blood; they can be identified using different methods[4, 5].

Only one of these is considered as a standardized and repeatable method. This method identifies cells characterized by EpCAM, Citokeratin 8, 18 and 19, DAPI positivity and CD45 negativity as CTC[6]. The problem of this method is that it could miss cells that are transforming themselves (from epithelial to mesenchymal). The epithelial to mesenchymal transformation (EMT) has been recognised as one of the steps involved in the metastatic process[7-9].

Moreover, the group at MSKCC suggested the role of CTC in the self-seeding process by using an animal model (2009). This hypothesis could explain the occurrence of late local relapses[10,11]. According to this model, a particular type of “aggressive CTC” could be the cause of both local relapse and metastatic disease. This is not really conclusively proven and different authors cast some doubts about this conclusion.

The identification and the recognition of these different types of cells (CSC, DTC and CTC) could give to the physician the possibility to monitor and characterize disease before and during the treatment, as a kind of “liquid biopsy”[12-15].

CTC’s identification has already been demonstrated to have a prognostic and predictive value in metastatic breast, prostate[16,17].

However, in the daily clinical activity we are very far from this. There are still a lot of open questions to be solved before CTC determination can be applied in clinical practice.

Among them:

- can CTCs be used to predict survival also in non metastatic, locally advanced tumours (in particular breast, prostate cancer)?

- can CTCs be used in other tumours (e.g. head and neck cancer)?

- can CTCs be used to biologically characterize the disease during its clinical evolution?

Breast cancer

Breast cancer is the more frequent tumour affecting females. Nowadays, most of these tumours are diagnosed in an early stage, but a lot of them can progress to metastatic disease. Many different therapeutic options (chemotherapy, hormonal and target therapy) are available to treat and to prolong survival of these patients.

The already available prognostic and predictive factors are patient-related (menopausal status, age) and disease related (hormonal status, disease stage, biological factors as Her2 expression). In metastatic patients the absence/presence of visceral disease has a relevant prognostic value. Tumour markers are often used to monitor the evolution of metastatic disease and its response to the treatment; unfortunately, these factors cannot be used to accurately predict progression free and overall survival during treatment[18]. In fact, they cannot anticipate the duration of response to therapy and their changes often do not mirror the clinical evolution of disease.

The discovery of DTC and CTC led clinical investigators to hypothesize they could solve the problem of “anticipating response” to treatment in patients with metastatic breast cancer. Similarly, the identification of DTC or CTC could be used to select patients with localized or locally advanced breast cancer requiring a more aggressive treatment.

Clinical relevance of CTC in metastatic breast cancer

CTC have already been identified as a relevant prognostic factor in metastatic breast cancer. The first evidence of their role was published by Cristofanilli in 2005[19]. In patients submitted to chemotherapy for metastatic breast cancer, he demonstrated the prognostic value of the presence (cut-off value, 5 CTC/7.5 ml) and of the variation in number (increase or reduction) of CTC’s during chemotherapy. In 2006[16] he concluded that the changes of number of CTC’s during the treatment can predict survival and response better than and before than conventional staging by imaging. CTC’s prognostic value in metastatic breast cancer was lastly confirmed in a recent retrospective analysis collecting the data of 1944 patients[20]; 991 of them had ≥ 5 CTC/7.5 ml before chemotherapy and worse progression free and overall survival; similarly, an increase of CTC’s during weeks 3-5 and 6-8 after the beginning of chemotherapy was related to worse progression and overall survival. CTC count during the treatment has a better prognostic value than other tumour markers (CA 15.3 and CEA). Many prospective/retrospective/observational studies confirmed these data. The more recent reports are listed in Table1.

An additional role of CTC identification in metastatic cancer can be the characterization of the differences between primary disease and metastatic disease in terms of Her2 and ER expression; this could lead to understand if the metastatic process changed the “type” of tumour and consequently to select different treatment options according to these findings[15].

The data about the difference in HER2 over-expression between primary tumour and the CTC’s detected in metastatic patients point to the same conclusion (<15% in primary tumour vs 15-90% in CTC’s); the different percentages of CTC’s positivity are probably related to the methods used to identify and characterize the CTC itself[24-28]. Two clinical prospective trials are addressing the possible benefit of using trastuzumab and lapatinib to treat patients with HER2 negative metastatic breast cancer (MBC) but harbouring Her2 positive CTC[29].

The data about ER expression in CTC’s are scarce; CTC’s ER positivity range between 18 to 57 %, a clearly lower value than in the primary tumour (70%)[30-31]; again, differences between studies can be related to the different identification and determination methods. The predictive role of ER-CTC status is, anyway, not clear, since clinical studies are still lacking.

The possible role of CTC’s in locally advanced/localized breast cancer

The clinical significance of CTC’s in locally advanced, localized breast cancer is far from being clear.

Their use has been investigated in different retrospective and prospective trials based on the hypotesis that CTC’s could be an early signal of residual disease and that they could initiate the metastatic process, consequently changing the natural history of the disease.

In fact, it would be very useful if early CTC’s detection could help in defining the prognosis of patients with early, node negative breast cancer patients, identifying those deserving a more aggressive treatment.

Some trials demonstrated that CTC detection is related to a worse disease free, metastases free, cancer specific and overall survival (Table 2).

Adjuvant chemotherapy and CTC

In his German trial (SUCCESS trials) Rack demonstrated in 2010 and more recently[32-33] the relevance of CTC detection (cut-off ≥1) before and after adjuvant chemotherapy, to predict DFS and OS, also as an independent factor.

Neoadjuvant chemotherapy and CTC

Contrasting results are evident regarding the use of CTC’s before and after neo-adjuvant chemotherapy. In patients submitted to neo-adjuvant chemotherapy CTC’s evolution during treatment is independent from the primary tumour response and it is not associated to any of its clinical and pathological features; some studies relate CTC’s reduction with a peculiar pattern of relapse[34].

An unexpected finding is that in some studies CTC’s status after neo-adjuvant chemotherapy does not seem to have any impact on prognosis[35]; in other reports, it relates with a worse DFS if the CTC’s number is ≥1 and with a worse OS if their number is ≥5[34].

A still unsolved problem is how the information from clinical studies can really help the physician to improve survival also in patients with CTC positive determination. The future should again rely on the biological characterization of these cells in order to guide a tailored treatment.

Can CTC be used to guide treatment in non-metastatic breast cancer?

The problem of understanding if CTC’s can be used to change the treatment of patients with localized breast cancer is not a little one. The results from the only study already published seem to show that an early change of treatment in patients with an increasing CTC number does not translate in a better OS: they maintain a prognosis worse than that of patients without CTC or with a reduced number of CTC after 21 days of treatment[21].

Different prospective clinical trials are now on-going to give an answer to this strategic question.

Table 3 shows only the trials registered at the clinicaltrials.gov website.

Prostate cancer

Prostate cancer is most common tumor in males. It is often discovered occasionally and as a very localized disease (low-risk disease). For these patients the therapeutic options can vary from “watchful waiting” to radiotherapy alone to surgery alone depending also by patient preference.

The patients defined at high risk (locally advanced tumour, high Gleason score and high PSA level)[40] are usually treated with conservative treatment: radiotherapy (with or without pelvic lymph-nodal irradiation[41] depending on the risk of nodal involvement and clinical practice of the different institutions) and hormonal treatment, better if more than two years[42].

The usual medical treatment of high risk or early metastatic disease is hormonal therapy (LHRH analogue or total androgenic block).

A substantial proportion of high risk patients progresses to metastatic disease and can become resistant to hormonal treatment.

There are no clear methods, apart from the already mentioned prognostic factors, to predict the clinical evolution of disease and to identify patients to be consequently treated with the more aggressive treatment options.

The rationale suggesting clinical investigators to search disseminated or, better, circulating tumour cells in the bone marrow or in the blood, respectively, also in prostate cancer is the same already discussed for breast cancer: to identify a simple, more effective method to predict survival and to guide the treatment.

The evidence about CTC prognostic value, however, is weaker for prostate than for breast cancer.

CTC in metastatic hormone sensible (HSPC) or castrate resistant (CRPC) prostate cancer

It has been demonstrated that CTC are detectable in patients with metastatic PC; the number and the evolution of CTC number during treatment is significantly related to survival and response to therapy.

The results of the recently published SWOG-S0421 trial confirmed the prognostic importance, in terms of OS, of CTC enumeration and change in number between baseline and day 21 in a phase III trial of patients treated with docetaxel with or without atrasentan[43]. Median CTC number at day 0 was 5 cell; a CTC number lower and higher than 5 was associated with PSA levels, bone pain, liver disease, haemoglobin level, alkaline phosphatase and PSA and RECIST response. OS was equal to 26 months for patients with < 5 CTC and 13 months in those with >5 CTC; a raising count between days 0 and 21 was associated with shorter OS. Less recent phase II studies showed the same results in terms of number of CTC before the treatment[44-45] and change of the number during the treatment[46]. These results induced FDA to authorize the use of CTC detection for the evaluation of metastatic CRPC using a standardized method.

The subsequent step will be to verify the utility of CTC count as surrogate of survival in CRPC. The requisite to consider a parameter as a response-indicator biomarker are very restrictive and has to be confirmed and re-confirmed in prospective randomized trials[47].

In HSPC only a few little studies suggest the utility of CTC. In 2011 Goodmann[48] demonstrated that CTC number is directly related to ALP and LDH levels and not related to PSA and testosterone levels (33 patients); at multivariate analysis only CTC at baseline had independent predictive value in terms of duration and magnitude of response to hormonal therapy. In 2012 Folkersma[49] compared 30 pts with localized PC, 30 patients with metastatic PC and 30 healthy volunteers. A significant correlation between all the marker of disease extension but Gleason score was demonstrated (PSA, T, N, M class). A prognostic correlation in terms of OS and PFS was evident if CTC count was >4; these data were confirmed at multivariate analysis.

It is still not clear if CTC enumeration can be used as an early marker of the evolution of disease and to early predict the loss of hormonal sensibility. More data are necessary to confirm this data and to allow a practice of change of treatment based on CTC count.

Localized prostate cancer

CTC appear to be less frequently detectable in localized than in metastatic PC. The studies in this patient setting are scarce; recently[50] it has been demonstrated that only 5/36 patients with newly diagnosed high risk PC (age >70, median PSA 14.1, median GS 9) had CTC in their blood. The presence of CTC did not correlate with other already confirmed prognostic factors (PSA, GS, stage T). Same results were evident comparing the percentage of CTC positivity between control subjects (20) and patients (20) with locally advanced PC (median PSA 21ng/ml, T>cT3a) with a p=0.66[51].

The low frequency of CTC positivity in localized prostate cancer forced clinical investigators to search different methods to identify CTC: CTC-chip platform[52], RT-PCR for prostate specific genes as a surrogate for counting CTC and as a mean to predict PC recurrence after prostatectomy[53].

Lowes in 2012[54] investigated the role of CTC in patients treated with radical radiotherapy (RT) for locally advanced PC and did not found a reduction of CTC number after RT; however, PSA responders could have a more pronounced reduction of CTC number than non responders (even if this observation does not reach statistical significance).

Again in this setting of patients the correlation between CTC and DTC in the bone marrow has been looked for, but the correlation was difficult to demonstrate. DTC itself were associated with worse prognosis in terms of probability of metastatic disease (multivariate analysis)[55-58].

Nowadays it is still not clear if the simple enumeration of CTC with standardized methods could be used in clinical practice for localized prostate cancer, either as a prognostic or a predictive factor.

Molecular characterization of CTC

The future studies on CTC in prostate cancer will probably address the biological characterization of these cells, in order to directly target the treatment to specific and different types of prostate cancer. A lot of different methods can be used to characterize the genomic and proteomic pattern of CTC including immunohistochemistry, immunofluorescence (presence or loss of phosphatase and tensin homolog PTEN; assess the number of copy of the AR gene), gene copy number analysis (detection of DNA mutations and altered copy number), genome sequencing analysis (hotspot mutation of phosphoinositide 3-kinase PI3K/AKT genes in the circulating genomic material) and epigenetic studies[59].

The interest of these different methods to track biological targets is the opportunity to monitor the disease during its natural history, to identify the evolution of the tumor in response to different treatment, to try to develop specific targeted treatments.

Clinical trials addressing future developments in CTC use

The lack of definitive answers as far as the clinical impact of CTC measurement is concerned is also testified by the ongoing trials reported in Table 4.

The main goals of these trials are: the choice of the better method to identify CTC; the clinical advantages of the use of newer identification methods; the impact of molecular characterization of CTC on survival.

How to apply the lessons learned from breast and prostate cancers to other cancer types: the case of head and neck tumours (HNC)

The interest for a possible relationship between CTC count and outcome in head and neck cancer has been recently growing because of the difficulties in predicting survival with the already known prognostic factors.

Different combinations of surgery, radiotherapy and chemotherapy are the mainstay of the treatment of head and neck cancer. The choice of the actual treatment derives from the tumor site (naso-, meso-, hypo-pharynx, larynx) and, within each site, from T, N and M categories and from their interplay, defining disease stage. Surgery is often the first approach, but radiotherapy is very often chosen to also in locally advanced cases, aiming at the cure of the disease with less functional damage[60].

Overall and progression free survival times after primary treatment can be predicted using mainly the same parameters (T, N, M).

Only recently some biological factors have been added to the diagnostic armamentarium as prognostic and predictive factors. The most frequently used biological predictors are the EGFR status (since EGFR is the target for some effective drugs, like cetuximab, and even if EGFR positivity is overwhelmingly frequent in HNC)[61, 62]; and the HPV positivity, mainly for meso-pharyngeal cancer, identifying patients with a better outcome even after less aggressive treatments[63]. The evidence on the predictive efficacy of these markers is growing, but they cannot be considered a standard mean to drive treatment choice.

Locally advanced disease

The interest about CTC determination in head and neck cancer is grown only in the last two-three years.

The research steps were not dissimilar from the case of breast and prostate cancer: 1. Try to identify CTC in the blood; 2. Try to correlate their presence and number to the other already known prognostic and predictive factor; and finally 3. Try to understand if they could more effectively predict response to treatment and prognosis.

Differently from the case of breast and prostate cancers, early studies were focused on locally advanced disease.

The results were not uniform.

Hristozova did not found a statistically significant correlation between CTC identification, T stage and tumour volume; an association with N2b status was however reported[64]; Bozec did not find any correlation with the already known prognostic factors (T, N, M)[65]; Buglione found an higher frequency of CTC detection in patients with stage IV disease, as compared with earlier stages, but without statistically significant differences[66].

The percentage of CTC positivity before primary treatment ranges between 15% and 43% and it is strongly dependent from the search method, being lower with standardized methods (Table 5)

Some authors demonstrated differences between CTC identification in different sites and also different clinical implications for CTC detection according to the tumor site: for example, a favourable prognostic significance in oro-pharyngeal cancer and a negative prognostic impact for the other disease sites have been reported[67] Our group detected CTC more frequently in oro- and hypo-pharyngeal cancer and SNUC cases[66].

Recently Gröbe[68] in a selected series of patients treated with surgery for oral cancer demonstrated a correlation between T, N, M category, CTC detection rate (in blood samples) and DTC positivity (in bone marrow). CTC and DTC were present in 12.5% and 20% of the patients, respectively. It is very interesting to note that CTC and DTC were identified together only in two patients. He demonstrated, both at univariate and multivariate analysis a statistically significant correlation between CTC/DTC detection and recurrence free survival.

As far as survival is concerned, again, the results reported are not uniform. Buglione[66] reported significantly better complete/partial response rates in patients without CTC or with a decreasing number of CTC during the treatment; no statistical evaluation of DFS or OS was done because the follow up period was too short. A decrease in the CTC number or their absence throughout the treatment was related with non-progressive disease, after both complete or incomplete remission, and with the number of patients alive and NED (no evidence of disease) (p = 0.009). Tinhofer[67] demonstrated that CTC presence was not predictive of OS or DFS in the entire series (144 patients); in oro-pharyngeal carcinomas (OPC, 63 patients) CTC detection was associated with improved DFS (CTC+ vs CTC-: 100% vs. 79%; p=0.059); the reverse was true for carcinomas of other sites (n=81; CTC+ vs CTC-: 29% vs 75%; p=0.001). In multivariate analysis, CTC remained an independent prognostic marker for DFS (p=0.002) and OS (p=0.016) in non-OPC.

Only one study tried to relate CTC presence to its biological characterization and to treatment response[69]. The number of CTC-positive samples significantly increased after radiotherapy. This radiation-induced increase in CTC numbers was less pronounced when radiotherapy was combined with cetuximab compared to the combination with cisplatin/5-fluorouracil. The Cetuximab regimen was also effective in reducing phosphorylate EGFR (pEGFR) expression in CTCs.

Metastatic disease

More than 50% of the cases with stage III-IV head and neck cancer recur with metastatic disease. One third of all treated patients will respond to systemic treatment with complete or – more often- partial remission, even if cure is almost never obtained. It is difficult to predict both which patients will recur with metastatic disease and which patients will respond to the treatment. Only one (not validated) prognostic score has been published to try to predict progression free and overall survival in metastatic disease. This score considers factors like performance status, weight loss, primary site, response to radiotherapy and tumour cell differentiation[70].

As for breast and prostate cancers, it is possible to use CTC to predict the outcome of patients treated with chemotherapy.

Our group explored also the use of CTC in metastatic patients. Grisanti[71] confirmed the presence of CTC before chemotherapy (26%) and during treatment. He demonstrated that at univariate analysis ≥2 CTCs are related with a prognosis poorer than 0-1 CTC; the presence of ≥1 CTC was related to worse PFS and OS. At multivariate analysis, disease control after systemic therapy was obtained in 8% of CTC-positive patients as opposed to 45% of the CTC-negative ones (p 0.03).

An important research effort will be needed do better define the possible clinical use of CTC in this clinical setting; the data collected, even if interesting, are in fact far from being sound enough to find a clear clinical application.

Also for HNC, the final goal resides in the biological characterization of these cells to try to guide the treatment directly according to specific targets.

Probably because the rarity of this kind of tumours the on-going prospective trials devoted to understand the role of CTC are unfortunately very rare (Table 6).

Conclusions

A lot of scientific and economic energies have been devoted to understand the role of CTC to enhance the knowledge of the natural history of some cancer types.

Metastatic breast cancer was the first disease for which FDA in USA approved the use of CTCs as a prognostic/predictive factor. Metastatic prostate cancer was the second. Despite this important acknowledgement, until now, the use of this new biologic instrument did not impact significantly on the choice of the treatment.

CTC determination in metastatic breast and prostate cancer can be effective when used as prognostic and predictive factor. We know that the presence of CTC before the beginning of the treatment correlates with worse progression free and overall survival, but neither the knowledge of its change during the treatment neither the early change of treatment guided by CTC determination seem to modify the outcome.

Results are even scarcer in locally advanced and early breast and prostate cancer. For early disease, questions far outweigh answers. Correlations between CTC determination and the other already used prognostic factors remain unclear and it is impossible to use CTC determination to guide treatment choice.

The reason of these difficulties probably depends from the methods of determination (probably not all the cells are correctly identified) and from the lack of biologic characterization. In locally advanced and early stage disease ineffective detection methods probably are not able to identify the few, possibly present, CTC.

In addition, disease biological features change during its natural history; the treatment should be probably targeted on these new targets rather than on those of disease at presentation.

The ongoing trials in breast and prostate cancer are trying to answer these open questions in order to confirm or discard, also from an health economics point of view, the utility of CTC determination and characterization.

The problem is obviously much more complex for head and neck cancer.

Difficulties with HNC are amplified by the heterogeneity of the disease(s) in terms of staging and prognosis (epi-pharyngeal tumor are very different from meso-pharyngeal or hypo-pharyngeal or laryngeal tumors and all they are different from oral cavity tumors) and treatment (different combinations of surgery, radiotherapy and chemotherapy or biological treatment).

Results on correlations between CTC determination and stage, site of primary are therefore very different in the different trials. The percentage of CTC positivity varies very much between the different trials and according to the the different methods of determination.

The results of the only prospective trial on metastatic disease confirm the prognostic role of CTC before the beginning of chemotherapy and the significance of CTC reduction during the treatment, in terms of disease control.

CTC determination has been applied also to other neoplasms, not covered by this review. Among them at least lung cancer should be quoted[74-76].

However, in an era of reduced resources for health systems it is easily understandable that very costly predictive and prognostic tests should be fully validated before being adopted in the routine clinical practice.

The study of CTC has important biological implications; it could advance further our knowledge of the neoplastic phenotype and of its variations after therapy. However, clinical applications are in their infancy and should be considered very cautiously. This does not mean that research in this field should be abandoned; rather, more sound biological bases should probably be built before attempting extensive clinical testing.

Acknowledgements

Prof Piero Nicolai, Prof Roberto Maroldi, Prof Alfredo Berruti, Dott.ssa Mirella Marini, Dott.ssa Monica Mangoni

CONFLICT OF INTERESTS

There are no conflicts of interest with regard to the present study.

REFERENCES

1.Krause M, Yaromina A, Eicheler W, Koch U, Baumann M Cancer stem cells: targets and potential biomarkers for radiotherapy. Clin Cancer Res 2011; 17(23):7224-9.

2.Baumann M, Krause M.CD44: a cancer stem cell-related biomarker with predictive potential for radiotherapy. Clin Cancer Res 2010; 16(21):5091-3.

3.Wiedswang G, Borgen E, Karesen R, Kvalheim G, Nesland JM, Qvist H,Schlichting E, Sauer T, Janbu J, Harbitz T, Naume B: Detection of isolated tumor cells in bone marrow is an independent prognostic factor in breast cancer. J Clin Oncol 2003; 21:3469-3478.

4.Harouaka R, Kang Z, Zheng Si-Yang, Cao L. Circulating tumor cells: Advances in isolation and analysis, and challenges for clinical applications Pharmacology & Therapeutics 2014; 141: 209–221 PMID: 24134902

5.Bitting RL, Boominathan R, Rao C, Kemeny G, Foulk B, Garcia-Blanco MA, Connelly M, Armstrong AJ Development of a method to isolate circulating tumor cells using mesenchymal-based capture Methods 2013; 64: 129–136.

6.Allard WJ, Matera J, Miller MC, Repollet M, Connelly MC, Rao C, Tibbe AG, Uhr JW, Terstappen LW. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin Cancer Res.2004; 10(20): 6897-6904

7.Chaffer CL, Weinberg RA A perspective on cancer cell metastasis. Science 2011; 331(6024):1559-64.

8.Foroni C, Broggini M, Generali D, Damia G Epithelial–mesenchymal transition and breast cancer: Role, molecular mechanisms and clinical impact. Cancer Treatment Reviews 2012; 38: 689-697

9.Yang J, Weinberg RA Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell 2008, 14:818–829.

10.Kim MY, Oskarsson T, Acharyya S, Nguyen DX, Zhang XH, Norton L, Massagué J Tumor self-seeding by circulating cancer cells. Cell 2009 24: 139(7):1315-26.

11.Aguirre-Ghiso JA On the theory of tumor self-seeding: implications for metastasis progression in humans Breast Cancer Research 2010, 12(2):304.

12.Riethdorf S, Wikman H, Pantel K Review: Biological relevance of disseminated tumor cells in cancer patients Int. J. Cancer 2008 123: 1991–2006

13.Alix-Panabie` C and Pantel K Circulating Tumor Cells: Liquid Biopsy of Cancer Clinical Chemistry 2013 59(1): 110–118

14.Liberko M, Kolostova K, Bobek V Essentials of circulating tumor cells for clinical research and practice, Critical Reviews in Oncology/Hematology 2013 88: 338–356

15.Friedlander TW, Premasekharan G, Paris PL Looking back, to the future of circulating tumor cells. Pharmacol Ther. 2014 142(3): 271-80.

16.Budd GT, Cristofanilli M, Ellis MJ, Stopeck A, Borden E, Miller MC, Matera J, Repollet M, Doyle GV, Terstappen LW, Hayes DF Circulating Tumor Cells versus Imaging--Predicting Overall Survival in Metastatic Breast Cancer Clin Cancer Res 2006; 12: 6403-6409.

17.de Bono JS, Scher HI, Montgomery RB, Parker C, Miller MC, Tissing H, Doyle GV, Terstappen LW, Pienta KJ, Raghavan D Circulating Tumor Cells Predict Survival Benefit from Cancer Treatment in Metastatic Castration-Resistant Prostate Clin Cancer Res 2008 14: 6302-6309.

18.Novaković S. Tumor markers in clinical oncology Radiol Oncol 2004; 38(2): 73-83

19.Cristofanilli M, Hayes DF, Budd GT, Ellis MJ, Stopeck A, Reuben JM, Doyle GV, Matera J, Allard WJ, Miller MC, Fritsche HA,. Hortobagyi GN, Terstappen LW Circulating Tumor Cells: A Novel Prognostic Factor for Newly Diagnosed Metastatic Breast Cancer J Clin Oncol. 2005 23(7): 1420-30.

20.Bidard FC, Peeters DJ, Fehm T, Nolé F, Gisbert-Criado R, Mavroudis D, Grisanti S, Generali D, Garcia-Saenz JA, Stebbing J, Caldas C, Gazzaniga P, Manso L, Zamarchi R, Fernandez de Lascoiti A, De Mattos-Arruda L, Ignatiadis M, Lebofsky R, van Laere SJ, Meier-Stiegen F, Sandri MT, Vidal-Martinez J, Politaki E, Consoli F, Bottini A, Diaz-Rubio E, Krell J et al. Clinical validity of circulating tumour cells in patients with metastatic breast cancer: a pooled analysis of individual patient data Lancet Oncol 2014 15: 406–14.

21.Jeffrey B. Smerage, William E. Barlow, Gabriel N. Hortobagyi, Eric P. Winer, Brian Leyland-Jones, Gordan Srkalovic, Sheela Tejwani, Anne F. Schott, Mark A. O’Rourke, Danika L. Lew, Gerald V. Doyle, Julie R. Gralow, Robert B. Livingston, and Daniel F. Hayes Circulating Tumor Cells and Response to Chemotherapy in Metastatic Breast Cancer: SWOG S0500 J Clin Oncol 2014.

22.Martín M, Custodio S, de Las Casas ML, García-Sáenz JÁ, de la Torre JC, Bellón-Cano JM, López-Tarruella S, Vidaurreta-Lazaro M, de la Orden V, Jerez Y, Márquez-Rodas I, Casado A, Sastre J, Díaz-Rubio E Circulating Tumor Cells Following First Chemotherapy Cycle: An Early and Strong Predictor of Outcome in Patients With Metastatic Breast Cancer The Oncologist 2013 18:917–923.

23.Wallwiener M, Riethdorf S, Hartkopf AD, Modugno C, Nees J, Madhavan D, Sprick MR, Schott S, Domschke C, Baccelli I, Schönfisch B, Burwinkel B, Marmé F, Heil J, Sohn C, Pantel K, Trumpp A, Schneeweiss A Serial enumeration of circulating tumor cells predicts treatment response and prognosis in metastatic breast cancer: a prospective study in 393 patients BMC Cancer 2014 14:512.

24.Krawczyk N, Banys M, Neubauer H, Solomayer EF, Gall C, Hahn M, Becker S, Bachmann R, Wallwiener D, Fehm T HER2 status on persistent disseminated tumor cells after adjuvant therapy may differ from initial HER2 status on primary tumor. Anticancer Res 2009 29:4019–24.

25.Pestrin M, Bessi S, Galardi F, Truglia M, Biggeri A, Biagioni C, Cappadona S, Biganzoli L, Giannini A, Di Leo A Correlation of HER2 status between primary tumors and corresponding circulating tumor cells in advanced breast cancer patients. Breast Cancer Res Treat 2009 118:523–30.

26.Meng S, Tripathy D, Shete S, Ashfaq R, Haley B, Perkins S, Beitsch P, Khan A, Euhus D, Osborne C, Frenkel E, Hoover S, Leitch M, Clifford E, Vitetta E, Morrison L, Herlyn D, Terstappen LW, Fleming T, Fehm T, Tucker T, Lane N, Wang J, Uhr J HER-2 gene amplification can be acquired as breast cancer progresses. Proc Natl Acad Sci U S A 2004 101:9393–8.

27.Pectasides D, Gaglia A, Arapantoni-Dadioti P, Bobota A, Valavanis C, Kostopoulou V, Mylonakis N, Karabelis A, Pectasides M, Economopoulos T. HER-2/neu status of primary breast cancer and corresponding metastatic sites in patients with advanced breast cancer treated with trastuzumab-based therapy. Anticancer Res 2006 26:647–53.

28.Hayes DF, Walker TM, Singh B, Vitetta ES, Uhr JW, Gross S, Rao C, Doyle GV, Terstappen LW Monitoring expression of HER-2 on circulating epithelial cells in patients with advanced breast cancer. Int J Oncol 2002 21:1111–7.

29.Bidard FC, Fehm T, Ignatiadis M, Smerage JB, Alix-Panabières C, Janni W, Messina C, Paoletti C, Müller V, Hayes DF, Piccart M, Pierga JY. Clinical application of circulating tumor cells in breast cancer: overview of the current interventional trials. Cancer Metastasis Rev 2013 32(1–2) 179–88.

30.Aktas B, Müller V, Tewes M, Zeitz J, Kasimir-Bauer S, Loehberg CR, Rack B, Schneeweiss A, Fehm T Comparison of estrogen and progesterone receptor status of circulating tumor cells and the primary tumor in metastatic breast cancer patients Gynecol Oncol 2011 122(2) 356–60.

31.Ditsch N, Mayer B, Rolle M, Untch M, Schildberg FW, Funke I Estrogen receptor expression profile of disseminated epithelial tumor cells in bone marrow of breast cancer patients. Recent Results Cancer Res 2003 162:141–7.

32.Rack B Prognostic relevance of circulating tumor cells in the peripheral blood of primary breast cancer patients 33rd Annual San Antonio Breast Cancer Symposium 2010.

33.Rack B, Schindlbeck C, Jückstock J, Andergassen U, Hepp P, Zwingers T, Friedl TW, Lorenz R, Tesch H, Fasching PA, Fehm T, Schneeweiss A, Lichtenegger W, Beckmann MW, Friese K, Pantel K, Janni W; SUCCESS Study Group. Circulating tumor cells predict survival in early average to high risk breast cancer patients. J Natl Cancer Inst 2014 15 106(5).

34.Pierga JY, Bidard FC, Mathiot C, Brain E, Delaloge S, Giachetti S, de Cremoux P, Salmon R, Vincent-Salomon A, Marty M Circulating tumor cell detection predicts early metastatic relapse after neoadjuvant chemotherapy in large operable and locally advanced breast cancer in a phase II randomized trial Clin Cancer Res 2008 14(21) 7004–10.

35.Pierga JY, Petit T, Delozier T, Ferrero JM, Campone M, Gligorov J, Lerebours F, Roché H, Bachelot T, Charafe-Jauffret E, Pavlyuk M, Kraemer S, Bidard FC, Viens P Neoadjuvant bevacizumab, trastuzumab, and chemotherapy for primary inflammatory HER2-positive breast cancer (BEVERLY-2): an open-label, single-arm phase 2 study Lancet Oncol 2012 13(4) 375–84.

36.36 Rack B, Schindlbeck C, Jückstock J, Andergassen U, Hepp P, Zwingers T, Friedl TW, Lorenz R, Tesch H, Fasching PA, Fehm T, Schneeweiss A, Lichtenegger W, Beckmann MW, Friese K, Pantel K, Janni W; SUCCESS Study Group. Circulating tumor cells predict survival in early average to high risk breast cancer patients. J Natl Cancer Inst. 2014 May 15;106(5).

37.Bidard FC, Mathiot C, Delaloge E, Brain E, Giacchetti S, de Cremoux P., Marty M. Single circulating tumor cell detection and overall survival in nonmetastatic breast cancer Annals of Oncology 2010 21:729-733.

38.Molloy TJ, Bosma AJ, Baumbusch LO, Synnestvedt M, Borgen E, Russnes HG, Schlichting E, van’t Veer LJ, Naume B. The prognostic significance of tumour cell detection in the peripheral blood versus the bone marrow in 733 early-stage breast cancer patients Breast Cancer Res 2011 13(3).

39.Franken B, de Groot MR, Mastboom WJB, Vermes I, van der Palen J, Tibbe AGJ, Terstappen LW. circulating tumor cells, disease recurrent and survival in newly diagnosed breast cancer Breast Cancer Res 2012, 14 (5): R133.

40.D’Amico AV, Whittington R, Malkowicz SB, Schultz D, Blank K, Broderick GA, Tomaszewski JE, Renshaw AA, Kaplan I, Beard CJ, Wein A Biochemical outcome after radical prostatectomy, external beam radiation therapy, or interstitial radiation therapy for clinically localized prostate cancer. JAMA 1998 280:969-974.

41.Pegurri L. Buglione M, Girelli G, Guarnieri A, Meattini I, Ricardi U, Mangoni M, Gabriele P, Bellavita R, Krengli M, Bonetta A, Cagna E, Bunkheila F, Borghesi S, Signor M, Di Marco A, Bertoni F, Stefanacci M, Gatta R, De Bari B, Magrini SM Changes in patterns of practice for prostate cancer radiotherapy in Italy 1995-2003. A survey of the Prostate Cancer Study Group of the Italian Radiation Oncology Society. Tumori 2014 100(1):31-7.

42.D’Amico AV, Denham JW, Bolla M, Collette L, Lamb DS, Tai KH, Steigler A, Chen MH. Short- vs long-term androgen suppression plus external beam radiation therapy and survival in men of advanced age with node-negative high-risk adenocarcinoma of the prostate. Cancer 2007 109(10):2004-10.

43.Goldkorn A, Ely B, Quinn DI, Tangen CM, Fink LM, Xu T, Twardowski P, Van Veldhuizen PJ, Agarwal N, Carducci MA, Monk JP 3rd, Datar RH, Garzotto M, Mack PC, Lara P Jr, Higano CS, Hussain M, Thompson IM Jr, Cote RJ, Vogelzang NJ. Circulating tumor cell counts are prognostic of overall survival in SWOG S0421: a phase III trial of docetaxel with or without atrasentan for metastatic castration-resistant prostate cancer. J Clin Oncol 2014 32(11):1136-42.

44.De Bono JS, Scher HI, Montgomery RB, Parker C, Miller MC, Tissing H, Doyle GV, Terstappen LW, Pienta KJ, Raghavan D Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin. Cancer Res. 2008, 14, 6302–6309.

45.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.

46.Quinn DI, Tangen C.M, Hussain M, Lara PN Jr, Goldkorn A, Moinpour CM, Garzotto MG, Mack PC, Carducci MA, Monk JP Docetaxel and atrasentan versus docetaxel and placebo for men with advanced castration-resistant prostate cancer (swog s0421): A randomised phase 3 trial. Lancet Oncol. 2013, 14, 893–900.

47.Doyen J, Alix-Panabières C, Hofman P, Parks SK, Chamorey E, Naman H, Hannoun-Lévi J-M Circulating tumor cells in prostate cancer: A potential surrogate marker of survival Critical Reviews in Oncology/Hematology 2012 81:241–256.

48.Goodman OB Jr, Symanowski JT, Loudyi A, Fink LM, Ward DC, Vogelzang NJ. Circulating tumor cells as a predictive biomarker in patients with hormone-sensitive prostate cancer. Clin Genitourin Cancer 2011 9(1):31-8.

49.Folkersma R, Manso San J, Romo G, Sierra MJ, Gómez O. Prognostic significance of circulating tumor cell count in patients with metastatic hormone-sensitive prostate cancer. Urology 2012 80(6):1328-32.

50.Loh J, Jovanovic L, Lehman M, Capp A, Pryor D, Harris M, Nelson C, Martin J. Circulating tumor cell detection in high‑risk non‑metastatic prostate cancer. J Cancer Res Clin Oncol 2014 Jul 16.

51.Thalgott M, Rack B, Maurer T, Souvatzoglou M, Eiber M, Kress V, Heck MM, Andergassen U, Nawroth R, Gschwend JE Detection of circulating tumor cells in different stages of prostate cancer. J. Cancer Res. Clin. Oncol 2013 139: 755–763.

52.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: 25.

53.Yates DR, Roupret M, Drouin SJ, Comperat E, Ricci S, Lacave R, Sebe P, Cancel-Tassin G, Bitker MO, Cussenot O Quantitative RT-PCR analysis of PSA and prostate-specific membrane antigen mRNA to detect circulating tumor cells improves recurrence-free survival nomogram prediction after radical prostatectomy. Prostate 2012, 72, 1382–1388.

54.Lowes LE, Lock M, Rodrigues G, D’Souza D, Bauman G, Ahmad B, Venkatesan V, Allan AL, Sexton T. Circulating tumour cells in prostate cancer patients receiving salvage radiotherapy Clin Transl Oncol 2012 14(2):150-6.

55.Riethdorf S, Wikman H, Pantel K. Review: Biological relevance of disseminated tumor cells in cancer patients. Int J Cancer 2008 123(9):1991-2006.

56.Berg A, Berner A, Lilleby W, Bruland OS, Fossa SD, Nesland JM, Kvalheim G Impact of disseminated tumor cells in bone marrow at diagnosis in patients with non metastatic prostate cancer treated by definitive radiotherapy. Int. J. Cancer Suppl 2007 120, 1603–1609.

57.Lilleby W, Stensvold A, Mills I.G, Nesland JM Disseminated tumor cells and their prognostic significance in nonmetastatic prostate cancer patients. Int. J. Cancer Suppl 2013 133, 149–155.

58.Kollermann J, Weikert S, Schostak M, Kempkensteffen C, Kleinschmidt K, Rau T, Pantel K Prognostic significance of disseminated tumor cells in the bone marrow of prostate cancer patients treated with neoadjuvant hormone treatment. J. Clin. Oncol 2008 26: 4928–4933.

59.Lorente D, Mateo J, de Bono JS Molecular Characterization and Clinical Utility of Circulating Tumor Cells in the Treatment of Prostate Cancer asco.org/edbook | 2014.

60.Corvò R Evidence-based radiation oncology in head and neck squamous cell carcinoma. Radiother Oncol 2007 85(1):156-70.

61.Ang KK, Zhang Q, Rosenthal DI, Nguyen-Tan PF, Sherman EJ, Weber RS, Galvin JM, Bonner JA, Harris J, El-Naggar AK, Gillison ML, Jordan RC, Konski AA, Thorstad WL, Trotti A, Beitler JJ, Garden AS, Spanos WJ, Yom SS, Axelrod RS Randomized Phase III Trial of Concurrent Accelerated Radiation Plus Cisplatin With or Without Cetuximab for Stage III to IV Head and Neck Carcinoma: RTOG 0522 J Clin Oncol. 2014 Aug 25. pii: JCO.2013.53.5633.

62.Bonner JA, Harari PM, Giralt J, Azarnia N, Shin DM, Cohen RB, Jones CU, Sur R, Raben D, Jassem J, Ove R, Kies MS, Baselga J, Youssoufian H, Amellal N, Rowinsky EK, Ang KK Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck. N Engl J Med 2006 354(6): 567-78.

63.Barry B, Ortholan C Human papilloma virus in head and neck cancer. Cancer Radiother. 2014.

64.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) Annals of Oncology 2011 22: 1878–1885.

65.Bozec A, Ilie M, Dassonville O, Long E, Poissonnet G, Santini J, Chamorey E, Ettaiche M, Chauvière D, Peyrade F, Hebert C, Benezery K, Sudaka A, Haudebourg J, Selva E, Hofman P Significance of circulating tumor cell detection using the CellSearch system in patients with locally advanced head and neck squamous cell carcinoma Eur Arch Otorhinolaryngol 2013 270:2745–2749.

66.Buglione M, Grisanti S, Almici C, Mangoni M, Polli C, Consoli F, Verardi R, Costa L, Paiar F, Pasinetti N, Bolzoni A, Marini M, Simoncini E, Nicolai P, Biti G, Magrini SM.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. European Journal of Cancer 2012 48, 3019– 3026.

67.Tinhofer I, Konschak R, Stromberger C, Raguse JD, Dreyer JH, Jöhrens K, Keilholz U, Budach V Detection of circulating tumor cells for prediction of recurrence after adjuvant chemoradiation in locally advanced squamous cell carcinoma of the head and neck Ann Oncol 2014

68.Grobe A, Blessmann M, Hanken H, Friedrich RE, Schon G, Wikner J, Effenberger KE, Kluwe L, Heiland M, Pantel K, Riethdorf S. 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.

69.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 Radiation Oncol Biol Phys 2012 83(5): e685ee690.

70.Argiris A, Li Y, Forastiere A. Prognostic factors and long-term survivorship in patients with recurrent or metastatic carcinoma of the head and neck. Cancer 2004 101(10): 2222-9.

71.Grisanti S, Almici C, Consoli F, Buglione M, Verardi R, Bolzoni-Villaret A, Bianchetti A, Ciccarese C, Mangoni M, Ferrari L, Biti G, Marini M, Ferrari VD, Nicolai P, Magrini SM, Berruti A. Circulating tumor cells in patients with recurrent or metastatic head and neck carcinoma: prognostic and predictive significance PLoS One.2014 9(8): e103918.

72.Nichols AC, Lowes LE, Szeto CCT, Basmaji J, Dhaliwal S, Chapeskie C, Todorovic B, Read N, Venkatesan V, Hammond A, Palma DA, Winquist E, Ernst S, Fung K, Franklin JH, Yoo J, Koropatnick J, Mymryk JS, Barrett JW, Allan AL Detection of circulating tumor cells in advanced head and neck cancer using the cellsearch system Head Neck 34: 1440–1444, 2012.

73.He S, Li P He S Long T Zhang N Fang J Yu Z Detection of circulating tumour cells with the CellSearch system in patients with advanced-stage head and neck cancer: preliminary results. J Laryngol Otol 2013 127: 788.

74.Krebs MG, Sloane R, Priest L, Lancashire L, Hou JM, Greystoke A, Ward TH, Ferraldeschi R, Hughes A, Clack G, Ranson M, Dive C, Blackhall FH. Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J Clin Oncol 2011 29(12): 1556-63.

75.Martin OA, Anderson RL, Russell PA, Cox RA, Ivashkevich A, Swierczak A, Doherty JP, Jacobs DH, Smith J, Siva S, Daly PE, Ball DL, Martin RF, MacManus MP Mobilization of viable tumor cells into the circulation during radiation therapy. Int J Radiat Oncol Biol Phys 2014 88(2): 395-403.

76.Hofman V, Ilie MI, Long E, Selva E, Bonnetaud C, Molina T, Vénissac N, Mouroux J, Vielh P Hofman P Detection of circulating tumor cells as a prognostic factor in patients undergoing radical surgery for non-small-cell lung carcinoma: comparison of the efficacy of the CellSearch Assay™ and the isolation by size of epithelial tumor cell method. Int J Cancer 2011 129(7):1651-60.

Refbacks

  • There are currently no refbacks.