Change in number and size of circulating tumor cells with high telomerase activity during treatment of patients with gastric cancer

  • Authors:
    • Hiroaki Ito
    • Noriko Yamaguchi
    • Manabu Onimaru
    • Satoshi Kimura
    • Tohru Ohmori
    • Fumihiro Ishikawa
    • Jun Sato
    • Shun Ito
    • Haruhiro Inoue
  • View Affiliations

  • Published online on: October 11, 2016     https://doi.org/10.3892/ol.2016.5239
  • Pages: 4720-4726
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Detection of circulating tumor cells (CTCs) in peripheral blood is useful for estimating the prognosis of patients with cancer. We previously reported the detection of CTCs by OBP‑401, a telomerase‑specific, replication‑selective, oncolytic adenoviral agent carrying the green fluorescent protein (GFP) gene. We demonstrated that the number of large (L)‑GFP+ cells (≥7.735 µm in diameter) in peripheral blood samples correlated significantly with the prognosis of treatment-naïve gastric cancer patients, whereas the number of small (S)‑GFP+ cells (<7.735 µm in diameter) did not. In the present study, we studied the change in the number of GFP+ cells during treatment, and analyzed the association between the number of GFP+ cells in blood samples and the outcome of patients. Peripheral blood samples were obtained from 37 gastric patients prior and subsequent to surgery (three samples per time point). Upon infection of blood cells with OBP‑401, GFP+ cells of different sizes were counted and measured. The association between the number of GFP+ cells and surgical outcome was determined by statistical analysis. The median follow‑up period after surgery was 39 months. Although the difference was not significant, patients with ≥6 L‑GFP+ cells in preoperative blood samples had a lower relapse‑free survival rate than patients with 0‑5 L‑GFP+ cells. There was no significant correlation between the number of L‑GFP+ cells in postoperative blood samples and the prognosis of patients receiving adjuvant therapy. Although the difference was not significant, the number of S‑GFP+ cells in samples from patients who had received postoperative chemotherapy was higher than in those who had not. The number of L‑GFP+ cells was not significantly correlated with the relapse‑free survival rate in gastric cancer patients who underwent surgery. The number of S‑GFP+ cells was relatively high in samples from patients who had received postoperative chemotherapy.

Introduction

The presence of circulating tumor cells (CTCs) in peripheral blood indicates a systemic disease stage (1), and detection of CTCs in peripheral blood is useful for estimating prognosis and monitoring disease progression in breast (2), prostate (3), skin (4) and colon (5) malignancies. The commonly used techniques for enrichment and detection of CTCs are density gradient separation (6,7), direct enrichment by filtration (8), immunomagnetic separation (9), flow cytometry (10), reverse transcription-polymerase chain reaction (RT-PCR) (11,12) and microchip technology (13). The CELLSEARCH® System (Veridex LLC, Raritan, NJ, USA) (14) is a widely used automated method of immunomagnetic cell enrichment (1517) that detects CTCs by detection of epithelial markers such as epithelial cell adhesion molecule (EpCAM) and cytokeratin. The expression of these markers, however, decreases during the epithelial-mesenchymal transition (EMT) (18,19); thus, CTCs undergoing EMT may escape detection.

Increased telomerase activity is a common characteristic of malignant tumors, and telomerase activity strongly correlates with carcinogenesis and disease progression (2022). As a novel means of detecting cells with high telomerase activity in peripheral blood samples of cancer patients, a telomerase-specific, replication-competent adenovirus variant termed OBP-401 (TelomeScan®; Oncolys BioPharma, Inc., Tokyo, Japan) was used (22). In this variant, the human telomerase reverse transcriptase (TERT) gene promoter drives the expression of the viral genes adenovirus early region 1A (E1A) and E1B, while the cytomegalovirus promoter drives the expression of the green fluorescent protein (GFP) gene, which serves as a marker of viral replication (22).

In our previous study (23), we used OBP-401 to detect CTCs in patients with gastric cancer. Gastric cancer is the fifth most common cancer worldwide and the third most frequent cause of cancer-associated mortality, affecting ~723,000 people in 2012 (24). In that study, blood samples from 65 treatment-naïve gastric cancer patients were collected prior to surgery (23). CTCs are larger than normal blood cells (25,26), and it was observed that the GFP+ cells in these samples with a diameter ≥7.735 µm (termed L-GTP+ cells) expressed EpCAM and other CTC markers (23). Additional results indicated that the number of L-GFP+ cells was significantly correlated with venous invasion (23) and overall survival after a median follow-up period of 3 years (27).

The number of CTCs following initial treatment correlates significantly with treatment efficacy and prognosis in patients with breast (2), prostate (3) and colorectal cancer (5). However, whether this is the case for patients with gastric cancer, is controversial. In the present study, the number of CTCs (defined as L-GFP+ cells) were determined prior and subsequent to treatment.

Materials and methods

Patients and healthy volunteers

The present report is an interim analysis of part of our prospective study of CTCs as GFP+ cells in patients with treatment-naïve gastric adenocarcinoma who underwent surgery at the Digestive Disease Center of Showa University Northern Yokohama Hospital (Tsuzuki-ku, Japan) between April 2010 and May 2011. Peripheral blood samples (three samples per time point) were obtained from 37 patients before surgery and at 4 and 24 weeks after surgery. The inclusion criteria were as follows: i) Histologically confirmed adenocarcinoma of the stomach by endoscopic biopsy; ii) clinical solitary tumor; iii) no prior endoscopic resection, chemotherapy or radiotherapy; iv) age, 20–80 years; v) Eastern Cooperative Oncology Group performance status (28) of 0 or 1; vi) sufficient organ function; and vii) written informed consent. The exclusion criteria were as follows: i) Synchronous or metachronous malignancy; ii) pregnancy or breast-feeding; iii) active or chronic viral hepatitis; iv) active bacterial or fungal infection; v) diabetes mellitus; vi) systemic administration of corticosteroids; and vii) unstable hypertension. The pathological stage of the disease was determined according to the 7th edition of the American Joint Committee on Cancer/International Union Against Cancer tumor-node-metastasis classification system (29). The depth of the tumor invasion in 4 patients without gastrectomy and the regional lymph node status of 7 patients without sufficient lymphadenectomy were surgically diagnosed.

All patients were followed up at our hospital every 3 months after surgery. The patients underwent endoscopy and computed tomography at least once per year depending on their disease stage and course. Postoperative therapy was allowed if required.

Peripheral blood samples were collected from healthy volunteers to establish the cut-off size of GFP+ cells. All volunteers were employees of Sysmex Corporation (Kobe, Japan), and were interviewed individually prior to sample collection. The volunteer group consisted of 7 men (mean age, 31.4 years; range, 24–39 years) and 3 women (mean age, 33.7 years; range, 26–48 years). All volunteers underwent medical check-ups upon employment and annually thereafter. Check-ups included medical interviews, auscultation, chest radiography, and blood and urine analyses. None of the volunteers was receiving medical treatment, pregnant or breast-feeding, or had donated blood within the month previous to the initiation of the study.

The present study was approved by the Institutional Review Board of Showa University Northern Yokohama Hospital (Tsuzuki-ku, Japan; approval number 0903-03). The study protocol was explained to patients and volunteers prior to obtention of written informed consent. Our study was registered with the University Hospital Medical Information Network in Japan (Tokyo, Japan; registration number 000004026).

Viruses

OBP-401, a telomerase-specific, replication-selective adenovirus variant in which the TERT promoter drives the expression of the viral EIA and EIB genes, and into which the GFP gene is integrated (22), was used. The sensitivity and specificity of the OBP-401 assay have been reported by Kim et al (30). Briefly, the assay was evaluated five times using MDA-MB-468 breast carcinoma cells as positive controls. The numbers of GFP+ cells in samples containing 1 MDA-MB-468 cell and 7.5 ml blood were 1, 1, 1, 2 and 3, respectively, while the numbers of GFP+ cells in samples containing 20 MDA-MB-468 cells and 7.5 ml blood were 15, 17, 19, 22 and 24, respectively. Viral samples were stored at −80°C.

Sample preparation and immunostaining

Details of sample preparation and assay are described in our previous report (23). A 7.5-ml peripheral vein blood sample was obtained from each patient before surgery and at 4 and 24 weeks after surgery. Blood was collected in tubes containing citric acid, phosphoric acid and dextrose, and stored at 4°C. Samples were assayed within 48 h of collection. Samples were centrifuged for 5 min at 540 × g, and the plasma phase was removed. Cell pellets were washed four times with PBS (Sigma-Aldrich; Merck Millipore, Darmstadt, Germany) and twice with RPMI medium (Sigma-Aldrich; Merck Millipore). Upon suspension in RPMI medium, cells were infected with 4×108 plaque-forming units of OBP-401 for 24 h at 37°C. Dead cells were stained with the red fluorescent reactive dye L23102 (Thermo Fisher Scientific, Inc., Waltham, MA, USA). Upon inactivation of OBP-401, cells were fixed with 2% paraformaldehyde for 20 min at room temperature and treated with a surface-active agent (Emalgen 2025 G; Kao Chemicals, Tokyo, Japan) for 10 min at 40°C to degrade red blood cells. Cells were incubated with phycoerythrin-labeled anti-human cluster of differentiation (CD) 45 antibody (catalogue number 368510; BioLegend, San Diego, CA, USA; 1:5 dilution) for 30 min at 25°C, which was diluted into PBS containing 2% fetal bovine serum (FBS; Sigma-Aldrich; Merck Millipore). Cells were washed with PBS containing 2% FBS and mounted on glass slides for microscopic analysis (two slides per sample).

Determination of GFP fluorescence intensity threshold

The threshold for GFP fluorescence intensity was determined as previously reported (23). Briefly, blood samples (7.5 ml) from healthy volunteers were mixed with ~30,000 A549 (lung carcinoma), HepG2 (hepatocellular carcinoma), HEC-1 (endometrial carcinoma), KATO-III (gastric carcinoma), SBC-3 (small cell lung carcinoma), LNCaP (prostate adenocarcinoma), MDA-MB-468 (breast carcinoma) or OVCAR-3 (ovarian carcinoma) cells. The A549, HepG2, HEC-1, KATO-III, SBC-3 and MDA-MB-468 cell lines were obtained from the Health Science Research Resources Bank (Osaka, Japan), while the LNCap and OVCAR-3 cell lines were obtained from the RIKEN Cell Bank (Tokyo, Japan). Cell lines were cultured according to the vendor's specifications. The samples were assayed using OBP-401, and GFP+ cells were visualized by fluorescence microscopy and counted. The GFP signal intensity threshold was determined to be 2.85×107 molecules of equivalent fluorochrome on the basis of the minimal GFP intensity level observed in the blood samples mixed with the cells. There was no significant difference in cell size prior and subsequent to OBP-401 infection.

Determination of cell size threshold

In our previous study (23), the various sizes of GFP+ cells in each sample made it difficult to determine which GFP+ cells best represented CTCs. To establish a constant value, the optimum threshold derived from a receiver operating characteristic curve analysis based on cell size was used to define GFP+ CTCs; this threshold was 7.735 µm. Thus, two groups of GFP+ cells were analyzed: Small (S)-GFP+ cells, measuring <7.735 µM in diameter, and large (L)-GFP+ cells, measuring ≥7.735 µm in diameter (27).

Cell counting and analysis. In a blind analysis, GFP+ cells on slides were counted on a computer-controlled fluorescence microscope (IX71; Olympus Corporation, Tokyo, Japan). Cells with fluorescent emission counts of ≥2.85×107 molecules of equivalent fluorochrome were counted as GFP+ cells. The criteria for a GFP+ cell did not include the presence of epithelial markers such as EpCAM or cytokeratin, since tumor cells undergoing EMT have been reported to lose epithelial markers (31). CD45+ cells were excluded from the analysis.

Statistical analysis

All statistical analyses were performed using JMP Pro 11.1.1 software (SAS Institute, Cary, NC, USA). The Kaplan-Meier analysis and the Wilcoxon test were used to calculate relapse-free survival rates and for non-parametric comparisons of the numbers of GFP+ cells. The relapse-free survival rate of the patients who underwent non-curative surgery declined to 0 months. P≤0.05 was considered to indicate a statistically significant difference.

Results

Participant characteristics

The clinicopathological characteristics of 37 patients (26 men and 11 women; mean age, 60.7 years; range, 33–76 years) are summarized in Table I. The median follow-up period of the surviving patients was 39 months. A total of 33 patients underwent pathological curative surgery, and 5 of these patients experienced disease recurrence. In total, 6 patients succumbed to disease; 16 patients underwent distal gastrectomy; 18 patients underwent total gastrectomy; and 3 underwent exploratory laparotomy. Of the 37 patients, 17 received chemotherapy post-surgery; 13 patients received oral chemotherapy (S-1); and 4 received oral chemotherapy combined with infusion (S-1/cisplatin and S-1/docetaxel).

Table I.

Patient characteristics and pathological findings.

Table I.

Patient characteristics and pathological findings.

VariableNumber of patients
Gender
  Male26
  Female11
Age, years (mean; range)60.7; 33–76
Gastrectomy
  Distal16
  Total18
  None  3
Curability
  R033
  R1  0
  R2  4
TNM stage
  I22
  II  5
  III  6
  IV  4
Depth of tumor invasion
  T120
  T2  5
  T3  5
  T4  7
Lymph node metastasis
  N022
  N1  4
  N2  3
  N3  8
Distant metastasis
  M033
  M1  4
Main histological typea
  Differentiated13
  Undifferentiated24
Lymphatic invasion
  L018
  L116
  LX  3
Venous invasion
  V019
  V1–215
  VX  3
Postoperative chemotherapy
  Yes (oral)13
  Yes (oral and infusion)  4
  No20
Outcome
  Relapse free28
  Relapse  5
  Non-curative surgery  4

a Well differentiated, moderately differentiated and papillary adenocarcinoma were categorized as differentiated. Signet-ring cell carcinoma, poorly differentiated adenocarcinoma and mucinous adenocarcinoma were categorized as undifferentiated. TNM, tumor-node-metastasis.

Association between the number of L-GFP+ cells and patient outcome

The association between the number of L-GFP+ cells and the relapse-free survival rate is shown in Fig. 1. Of the 37 patients enrolled in our study, 33 and 30 were relapse free at 4 and 24 weeks, respectively, after surgery. There were no significant differences in the relapse-free survival rates of patients with ≥6 L-GFP+ cells and patients with <6 L-GFP+ cells, either before (P=0.072; Fig. 1A) or at 4 (P=0.109; Fig. 1B) or 24 (P=0.342; Fig. 1C) weeks after surgery. At 4 weeks after surgery, the relapse-free survival rate was, however, relatively higher in patients with ≥6 GFP+ cells than in patients with <6 GFP+ cells.

Association between patient outcome and changes in the number of GFP+ cells

The numbers of L-GFP+ cells in peripheral blood samples from patients without relapse following curative surgery (R-), patients with relapse following curative surgery (R+) and patients who underwent non-curative surgery (Non-C) were compared at three time points. The mean numbers of L-GFP+ cells at 0, 4 and 24 weeks after surgery were as follows: R-, 5.6, 8.3 and 6.1, respectively; R+, 6.0, 4.0 and 7.2, respectively; and Non-C, 6.8, 0.8 and 4.0, respectively (Fig. 2A). There was a significant difference between the number of L-GFP+ cells in samples from the R- patients and the Non-C patients at 4 weeks after surgery (P=0.034).

Figure 2.

Association of GFP+ cell number with patient outcome. (A and B) The black dots indicate the number of GFP+ cells in 7.5-ml blood samples from chemotherapy-negative patients. The white dots indicate the number of GFP+ cells in samples from patients receiving chemotherapy. The bottom and top of the box represent the lower and upper quartiles, respectively, while the band across the box represents the median. The lower and upper bars at the ends of the whiskers indicate the lowest datum within 1.5 interquartile ranges of the lower quartile and the highest datum within 1.5 interquartile ranges of the upper quartile, respectively. The green bars indicate the mean value. (A) Numbers of L-GFP+ cells (≥7.735 µm in diameter). The mean numbers of L-GFP+ cells before surgery, 4 weeks post-surgery and 24 weeks post-surgery were as follows: R-, 5.6, 8.3 and 6.1; R+, 6.0, 4.0 and 7.2; and Non-C, 6.8, 0.8 and 4.0, respectively. There was a significant difference in the number of L-GFP+ cells in samples from R- patients and Non-C patients (P=0.034). (B) Numbers of S-GFP+ cells (<7.735 µm in diameter). The mean numbers of S-GFP+ cells before surgery, 4 weeks post-surgery and 24 weeks post-surgery were as follows: R-, 8.7, 11.3 and 11.6; R+, 11.8, 10.4 and 13.0; and Non-C, 10.3, 10.8 and 20.0, respectively. Although there were no significant differences, the number of S-GFP+ cells in samples from Non-C patients 24 weeks after surgery was relatively higher than the numbers of S-GFP+ cells in the samples from R- (P=0.932) and R+ patients (P=0.713). R-, patients without relapse; R+, patients with relapse; Non-C, patients who underwent non-curative surgery; GFP, green fluorescent protein; L, large; S, small.

The mean numbers of S-GFP+ cells at 0, 4 and 24 weeks after surgery were as follows: R-, 8.7, 11.3 and 11.6, respectively; R+, 11.8, 10.4 and 13.0, respectively; and Non-C, 10.3, 10.8 and 20.0, respectively (Fig. 2B). There were no significant differences between the three groups either prior or subsequent to surgery. The number of S-GFP+ cells in samples from Non-C patients at 24 weeks after surgery was, however, relatively high.

The associations between the number of GFP+ cells, postoperative chemotherapy and outcome are represented in Fig. 3. Of the 37 patients, 6 and 17 were receiving postoperative chemotherapy 4 and 24 weeks after surgery. The number of L-GFP+ cells was similar prior and subsequent to surgery, regardless of chemotherapy (Fig. 3A). Although there was no significant difference, the number of S-GFP+ cells was relatively high in the samples from the patients who were receiving postoperative chemotherapy 24 weeks after surgery (Fig. 3B).

Discussion

In the present study, the association between the number of CTCs (L-GFP+ cells) and treatment outcome was assessed in patients with gastric cancer, which was the third leading cause of cancer-associated mortality worldwide in 2012 (24). The usefulness of CTCs for diagnosis and estimation of prognosis has been reported for breast (15,31), prostate (32), lung (33), esophagus (11,12), colon (34) and gastric cancer (23,27). The results of the present study indicate that the number of CTCs during treatment is unrelated to prognosis in gastric cancer.

A major finding of our study is that there is no significant correlation between patient prognosis and the number of L-GFP+ cells in samples from gastric cancer patients following surgery. This result is consistent with that of our previous study, which used PCR to detect CTCs in patients with esophageal cancer (11). Significant associations between the number of CTCs during treatment and prognosis have been reported for patients with breast (2), prostate (3) and colorectal (5) cancer. The difference in the results of these studies and the present study may reflect the type of cancer examined. The CTC number in our study may have been affected by the postoperative chemotherapy regimens, which were not the same for all patients. In addition, the median follow-up period of 3 years in our study may be too short to accurately evaluate the association between CTCs and prognosis. It is contrary to our expectation that the number of L-GFP+ cells in samples from the Non-C patients was significantly lower than R- patients 4 weeks after surgery. Since the number of Non-C patients in the present study is small, this significant difference should be confirmed by a large-scale prospective study.

The second major finding of our study is that the number of S-GFP+ cells increases following chemotherapy, which suggests that chemotherapy reduces the size of CTCs. The present authors suggest that the ratio of drug-resistant CTCs (e.g., cancer stem-like cells) to drug-sensitive CTCs increases following chemotherapy. Therefore, drug-resistant CTCs may be smaller than drug-sensitive CTCs. It is also possible that OBP-401 infection increases telomerase activity in non-cancer cells, which are smaller than CTCs.

A limitation of our study is that the metastatic potential of the GFP+ cells was not determined. Additional studies in a larger population of patients with different cancer types are required to clarify the clinical applicability of CTC detection. Future studies should analyze the functions of viable CTCs upon cell sorting and identify CTCs with metastatic potential using additional tools such as DNA ploidy analysis (35,36). Gene expression profiling of CTCs and primary tumors will also provide important insights into the mechanisms of cancer metastasis.

In summary, the results of the present study indicate that CTCs may be useful as predictors of disease progression in treatment-naïve gastric cancer patients; however, CTCs do not constitute a prognostic factor in patients during treatment.

There was no significant association between the change in the number of CTCs, treatment or prognosis in gastric cancer patients who underwent curative surgery. However, the present study used a short follow-up period, and only a small number of participants were included. In addition, whether all GFP+ cells have true metastatic potential was unclear. Therefore, further studies are warranted to confirm the findings of the present study.

Acknowledgements

The present study was supported in part by a Grant-in-Aid for Challenging Exploratory Research (grant number 23659308) from Japan Society for the Promotion of Science (Tokyo, Japan). The authors are grateful to all the patients and volunteers who donated blood for the study. The authors would like to thank Professor Toshiyoshi Fujiwara (Okayama University Graduate School of Medicine, Okayama, Japan) for helpful comments and suggestions; Mr. Yasuo Urata (Oncolys BioPharma, Tokyo, Japan) for supplying OBP-401; Dr Yukio Tsujino, Dr Toshiyuki Ozawa and Dr Akinori Masago (Sysmex Corporation, Kobe, Japan) for their valuable support; and the clinical staff of Showa University Northern Yokohama Hospital (Yokohama, Japan).

References

1 

Liotta LA, Kleinerman J and Saidel GM: Quantitative relationships of intravascular tumor cells, tumor vessels, and pulmonary metastases following tumor implantation. Cancer Res. 34:997–1004. 1974.PubMed/NCBI

2 

Camara O, Rengsberger M, Egbe A, Koch A, Gajda M, Hammer U, Jörke C, Rabenstein C, Untch M and Pachmann K: The relevance of circulating epithelial tumor cells (CETC) for therapy monitoring during neoadjuvant (primary systemic) chemotherapy in breast cancer. Ann Oncol. 18:1484–1492. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Scher HI, Jia X, de Bono JS, Fleisher M, Pienta KJ, Raghavan D and Heller G: Circulating tumour cells as prognostic markers in progressive, castration-resistant prostate cancer: A reanalysis of IMMC38 trial data. Lancet Oncol. 10:233–239. 2009. View Article : Google Scholar : PubMed/NCBI

4 

A L Reid, Millward M, Pearce R, Lee M, Frank MH, Ireland A, Monshizadeh L, Rai T, Heenan P, Medic S, et al: Markers of circulating tumour cells in the peripheral blood of patients with melanoma correlate with disease recurrence and progression. Br J Dermatol. 168:85–92. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Lu CY, Tsai HL, Uen YH, Hu HM, Chen CW, Cheng TL, Lin SR and Wang JY: Circulating tumor cells as a surrogate marker for determining clinical outcome to mFOLFOX chemotherapy in patients with stage III colon cancer. Br J Cancer. 108:791–797. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Hanahan D and Weinberg RA: The hallmarks of cancer. Cell. 100:57–70. 2000. View Article : Google Scholar : PubMed/NCBI

7 

Arai Y, Kanamaru H, Yoshimura K, Okubo K, Kamoto T and Yoshida O: Incidence of lymph node metastasis and its impact on long-term prognosis in clinically localized prostate cancer. Int J Urol. 5:459–465. 1998. View Article : Google Scholar : PubMed/NCBI

8 

Vona G, Sabile A, Louha M, Sitruk V, Romana S, Schütze K, Capron F, Franco D, Pazzagli M, Vekemans M, et al: Isolation by size of epithelial tumor cells: A new method for the immunomorphological and molecular characterization of circulating tumor cells. Am J Pathol. 156:57–63. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Talasaz AH, Powell AA, Huber DE, Berbee JG, Roh KH, Yu W, Xiao W, Davis MM, Pease RF, Mindrinos MN, et al: Isolating highly enriched populations of circulating epithelial cells and other rare cells from blood using a magnetic sweeper device. Proc Natl Acad Sci USA. 106:3970–3975. 2009. View Article : Google Scholar : PubMed/NCBI

10 

He W, Wang H, Hartmann LC, Cheng JX and Low PS: In vivo quantitation of rare circulating tumor cells by multiphoton intravital flow cytometry. Proc Natl Acad Sci USA. 104:11760–11765. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Ito H, Kanda T, Nishimaki T, Sato H, Nakagawa S and Hatakeyama K: Detection and quantification of circulating tumor cells in patients with esophageal cancer by real-time polymerase chain reaction. J Exp Clin Cancer Res. 23:455–464. 2004.PubMed/NCBI

12 

Honma H, Kanda T, Ito H, Wakai T, Nakagawa S, Ohashi M, Koyama Y, Valera VA, Akazawa K and Hatakeyama K: Squamous cell carcinoma-antigen messenger RNA level in peripheral blood predicts recurrence after resection in patients with esophageal squamous cell carcinoma. Surgery. 139:678–685. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Nagrath S, Sequist LV, Maheswaran S, Bell DW, Irimia D, Ulkus L, Smith MR, Kwak EL, Digumarthy S, Muzikansky A, et al: Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature. 450:1235–1239. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Cohen SJ, Punt CJ, Iannotti N, Saidman BH, Sabbath KD, Gabrail NY, Picus J, Morse M, Mitchell E, Miller MC, et al: Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer. J Clin Oncol. 26:3213–3221. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Riethdorf S, Fritsche H, Müller V, Rau T, Schindlbeck C, Rack B, Janni W, Coith C, Beck K, Jänicke F, et al: Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: A validation study of the cell search system. Clin Cancer Res. 13:920–928. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Davis JW, Nakanishi H, Kumar VS, Bhadkamkar VA, McCormack R, Fritsche HA, Handy B, Gornet T and Babaian RJ: Circulating tumor cells in peripheral blood samples from patients with increased serum prostate specific antigen: Initial results in early prostate cancer. J Urol. 179:2187–2191; discussion 2191. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Hou JM, Greystoke A, Lancashire L, Cummings J, Ward T, Board R, Amir E, Hughes S, Krebs M, Hughes A, et al: Evaluation of circulating tumor cells and serological cell death biomarkers in small cell lung cancer patients undergoing chemotherapy. Am J Pathol. 175:808–816. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Książkiewicz M, Markiewicz A and Zaczek AJ: Epithelial-mesenchymal transition: A hallmark in metastasis formation linking circulating tumor cells and cancer stem cells. Pathobiology. 79:195–208. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Gorges TM, Tinhofer I, Drosch M, Röse L, Zollner TM, Krahn T and von Ahsen O: Circulating tumour cells escape from EpCAM-based detection due to epithelial-to-mesenchymal transition. BMC Cancer. 12:1782012. View Article : Google Scholar : PubMed/NCBI

20 

Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho PL, Coviello GM, Wright WE, Weinrich SL and Shay JW: Specific association of human telomerase activity with immortal cells and cancer. Science. 266:2011–2015. 1994. View Article : Google Scholar : PubMed/NCBI

21 

Blackburn EH: Telomere states and cell fates. Nature. 408:53–56. 2000. View Article : Google Scholar : PubMed/NCBI

22 

Fujiwara T, Kagawa S, Kishimoto H, Endo Y, Hioki M, Ikeda Y, Sakai R, Urata Y, Tanaka N and Fujiwara T: Enhanced antitumor efficacy of telomerase-selective oncolytic adenoviral agent OBP-401 with docetaxel: Preclinical evaluation of chemovirotherapy. Int J Cancer. 119:432–440. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Ito H, Inoue H, Sando N, Kimura S, Gohda K, Sato J, Murakami K, Ito S, Odaka N, Satodate H and Kudo SE: Prognostic impact of detecting viable circulating tumour cells in gastric cancer patients using a telomerase-specific viral agent: A prospective study. BMC Cancer. 12:3462012. View Article : Google Scholar : PubMed/NCBI

24 

World Health Organization, . International Agency for Research on Cancer. GLOBOCAN 2012. Estimated Cancer Incidence, Mortality and Prevalence World Wide in 2012. http://globocan.iarc.fr/Pages/fact_sheets_cancer.aspxAccessed June 9, 2014.

25 

Lin HK, Zheng S, Williams AJ, Balic M, Groshen S, Scher HI, Fleisher M, Stadler W, Datar RH, Tai YC and Cote RJ: Portable filter-based microdevice for detection and characterization of circulating tumor cells. Clin Cancer Res. 16:5011–5018. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Zheng S, Lin HK, Lu B, Williams A, Datar R, Cote RJ and Tai YC: 3D microfilter device for viable circulating tumor cell (CTC) enrichment from blood. Biomed Microdevices. 13:203–213. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Ito H, Inoue H, Kimura S, Ohmori T, Ishikawa F, Gohda K and Sato J: Prognostic impact of the number of viable circulating cells with high telomerase activity in gastric cancer patients: A prospective study. Int J Oncol. 45:227–234. 2014.PubMed/NCBI

28 

Oken MM, Creech RH, Tormey DC, Horton J, Davis TE, McFadden ET and Carbone PP: Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol. 5:649–655. 1982. View Article : Google Scholar : PubMed/NCBI

29 

Sobin LH, Gospodarowicz MK and Wittekind C: TNM Classification of Malignant Tumours. 7th. Blackwell Publishing Ltd.; Oxford: 2010

30 

Kim SJ, Masago A, Tamaki Y, Akazawa K, Tsukamoto F, Sato J, Ozawa T, Tsujino Y and Noguchi S: A novel approach using telomerase-specific replication-selective adenovirus for detection of circulating tumor cells in breast cancer patients. Breast Cancer Res Treat. 128:765–773. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Cristofanilli M, Budd GT, Ellis MJ, Stopeck A, Matera J, Miller MC, Reuben JM, Doyle GV, Allard WJ, Terstappen LW and Hayes DF: Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 351:781–791. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Moreno JG, Miller MC, Gross S, Allard WJ, Gomella LG and Terstappen LW: Circulating tumor cells predict survival in patients with metastatic prostate cancer. Urology. 65:713–718. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Krebs MG, Sloane R, Priest L, Lancashire L, Hou JM, Greystoke A, Ward TH, Ferraldeschi R, Hughes A, Clack G, et al: Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J Clin Oncol. 29:1556–1563. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Katsumata K, Sumi T, Mori Y, Hisada M, Tsuchida A and Aoki T: Detection and evaluation of epithelial cells in the blood of colon cancer patients using RT-PCR. Int J Clin Oncol. 11:385–389. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Bonsing BA, Beerman H, Kuipers-Dijkshoorn N, Fleuren GJ and Cornelisse CJ: High levels of DNA index heterogeneity in advanced breast carcinomas. Evidence for DNA ploidy differences between lymphatic and hematogenous metastases. Cancer. 71:382–391. 1993. View Article : Google Scholar : PubMed/NCBI

36 

Klijanienko J, el-Naggar AK, de Braud F, Rodriguez-Peralto JL, Rodriguez R, Itzhaki M, Russo A, Janot F, Luboinski B and Cvitkovic E: Tumor vascularization, mitotic index, histopathologic grade, and DNA ploidy in the assessment of 114 head and neck squamous cell carcinomas. Cancer. 75:1649–1656. 1995. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2016
Volume 12 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ito H, Yamaguchi N, Onimaru M, Kimura S, Ohmori T, Ishikawa F, Sato J, Ito S and Inoue H: Change in number and size of circulating tumor cells with high telomerase activity during treatment of patients with gastric cancer. Oncol Lett 12: 4720-4726, 2016
APA
Ito, H., Yamaguchi, N., Onimaru, M., Kimura, S., Ohmori, T., Ishikawa, F. ... Inoue, H. (2016). Change in number and size of circulating tumor cells with high telomerase activity during treatment of patients with gastric cancer. Oncology Letters, 12, 4720-4726. https://doi.org/10.3892/ol.2016.5239
MLA
Ito, H., Yamaguchi, N., Onimaru, M., Kimura, S., Ohmori, T., Ishikawa, F., Sato, J., Ito, S., Inoue, H."Change in number and size of circulating tumor cells with high telomerase activity during treatment of patients with gastric cancer". Oncology Letters 12.6 (2016): 4720-4726.
Chicago
Ito, H., Yamaguchi, N., Onimaru, M., Kimura, S., Ohmori, T., Ishikawa, F., Sato, J., Ito, S., Inoue, H."Change in number and size of circulating tumor cells with high telomerase activity during treatment of patients with gastric cancer". Oncology Letters 12, no. 6 (2016): 4720-4726. https://doi.org/10.3892/ol.2016.5239