Abstract
Objectives
Development of targeted therapies for pancreatic cancer could be enhanced by a reliable method for noninvasive tumor cell assessment. In this pilot study we isolated and phenotypically characterized circulating tumor cells (CTC) from patients with metastatic pancreatic cancer and explored their relationship to clinical outcome.
Methods
Peripheral blood from 50 patients was collected at treatment initiation and first disease evaluation for CTC enumeration and phenotyping by CellSearch® system. Expression of human mucin 1 (MUC-1) was performed.
Results
48 and 37 patients had evaluable samples at baseline and first disease evaluation, respectively. The cohort was 62% male, with a median age of 63 years. At least 1 CTC/7.5mL was detected in 23 patients (48%) pre-treatment and 11 patients (30%) at first disease evaluation. No difference was seen in overall survival between patients with ≥ 1 CTC vs no CTC at baseline (p=0.14). Patients with MUC-1 expressing CTC (n=10) had shorter median overall survival compared to those with MUC-1 negative CTC (n=13) (2.7 vs. 9.6m; p=0.044).
Conclusions
CTC enumeration and phenotypic characterization from metastatic pancreatic cancer patients is feasible. No correlation was found between CTC isolation and survival. However, the presence of MUC-1 expressing CTC demonstrated a trend toward inferior survival.
Keywords: Pancreatic Cancer, Circulating Tumor Cells, MUC-1, Mucin-1
Introduction
Pancreatic cancer is the fourth leading cause of cancer-related death in the United States, with approximately 40,000 new cases and approximately the same number of deaths reported yearly1. Systemic chemotherapy is the mainstay of treatment for locally advanced and metastatic pancreatic cancer (mPC)2. Despite the most aggressive chemotherapy regimens available today, median survival remains less than 1 year 3,4. Thus, there is an urgent need for advances in the management of these patients through better systemic therapy. The development of effective therapies in pancreatic cancer is impeded by the relative inaccessibility of tumor for repeat biopsy to document therapeutic effect or evaluate tumor evolution through various lines of therapy. The capture of circulating tumor cells (CTCs) from patients is one potential strategy for in vivo pharmacodynamic assessment of pancreatic cancer cells.
CTCs studied in multiple cancers including breast, prostate, and colorectal cancers correlate with underlying tumor biology including genetic alterations, biomarker expression, and prognosis5–8. Our group previously demonstrated the prognostic significance of CTC enumeration in the peripheral blood of patients with metastatic colon cancer9. The presence of CTC at treatment initiation and during therapy conferred a poorer prognosis regardless of the treatment type10,11. The utility of CTC enumeration in pancreatic cancer patients has not been clearly defined.
MUC-1 is a large trans-membrane glycoprotein highly expressed in a subset of pancreatic adenocarcinomas compared to non-cancerous pancreatic tissue12. Increased expression of MUC-1 has been associated with increased metastatic properties of pancreatic cancer cells, possibly by induction of epithelial to mesenchymal transition13. Increased MUC-1 expression in a tumor correlates with higher stage and poorer outcome14. In addition, the presence of circulating anti-MUC-1 IgG antibodies is associated with improved survival among pancreatic cancer patients irrespective of stage15. Agents targeting MUC-1 are now being evaluated in clinical trials in pancreatic cancer16–19.
We conducted a pilot study to prospectively evaluate CTC yield using the CellSearch® system and to assess their relationship to clinical outcome in patients with metastatic pancreatic cancer (mPC). We further assessed expression of MUC-1 on captured CTCs and its relationship to clinical outcome.
Patients and Methods
Study Design
Patients 18 years or older with metastatic adenocarcinoma of the pancreas were enrolled to this study. Enrollment occurred shortly after diagnosis of mPC and prior to the initiation of front line systemic therapy. Patient demographics, tumor characteristics, and treatment regimens were recorded. Patients provided written informed consent according to institutional and federal requirements, following the approval of the protocol by the institutional review board of Fox Chase Cancer Center.
Fifty milliliters (mL) of peripheral blood were obtained from patients at study entry and at first disease evaluation (6–10 weeks after treatment initiation). Levels of the tumor marker CA-19-9 were recorded at the same timepoints. Overall survival (OS) was defined as the time between date of study enrollment and death.
Specimen Analysis
CTC evaluation was performed using the CellSearch® assay (Janssen Diagnostics LLC) as previously described20. Peripheral blood samples for CTC were collected in two 10 mL CellSave Perservative tubes™, and processed using the CellSearch™ Epithelial Cell kits. All automated enrichments were performed on the CellTracks™ AutoPrep System. Data was collected and analyzed on the CellTracks™ Analyzer II. Immunomagnetic enrichment of CTC using the Cell Tracks AutoPrep System has been described in detail by Kagan et al. 21. The assay was performed on a 7.5mL of blood sample from each patient at each timepoint. Cells were fluorescently labeled with anti-CD45APC, anti-pan cytokeratin-PE (CK8, 18, 19), DAPI, and anti- MUC-1-FITC (provided by Janssen LLC Huntington Valley). Images for analyses were sorted by computer-assisted software selecting events based on the parameters of CD45 negative, cytokeratin positive and DAPI positive. Selected CTCs were assessed for MUC-1 expression.
Statistical Methods
A sample size of 50 was selected as a pilot cohort for evaluation of CTCs in metastatic pancreatic cancer patients. The number of CTCs at different time points was summarized. Based on the distribution of CTC category, we dichotomized them into two categories (0 vs. ≥ 1). Differences in demographic and treatment characteristics by CTC count were assessed using Chi-square, Fisher’s exact, or Wilcoxon rank sum tests as appropriate. OS was visually examined using Kaplan-Meier curves and median survival estimated based on CTC counts and MUC-1 expression. Wilcoxon tests were used to test any difference in survival. The relation between CA19-9 and CTC number was examined. Spearman rank correlation was computed and tested, since both distributions of CA19-9 and CTC counts were skewed.. Analyses were performed using SAS statistical software ver. 9.3 (Cary, NC). All tests were considered significant using p-value <0.05.
Results
Patient Characteristics
Fifty patients were enrolled in this study between March 2007 and March of 2011. Patient characteristics for the entire cohort are shown in Table 1. Patient and treatment characteristics were balanced between the group of patients with CTC ≥ 1 versus the group with CTC=0. Median age was 61 years (range: 39–86 years), with the majority being Caucasian (84%) males (62%). The majority of patients received gemcitabine-based therapy (96%). Thirteen patients (26%) had received chemotherapy for localized disease prior to study enrollment, of which 12 received radiation and 2 underwent surgical resection.
Table 1.
Patient Demographics and Treatment Characteristics
All patients N=50* n (%) |
Baseline CTC
|
p-value | ||
---|---|---|---|---|
CTC=0 N=25 n (%) |
CTC ≥1 N=23 n (%) |
|||
Sex | ||||
Female | 19 (38.0%) | 12 (48.0%) | 7 (30.4%) | 0.21 |
Male | 31 (62.0%) | 13 (52.0%) | 16 (69.6%) | |
Median Age at diagnosis | 61.5 (38–86) | 61 (49–84) | 62 (38–86) | 0.80 |
years (range) | ||||
Race | ||||
Caucasian | 42 (84.0%) | 21 (84.0%) | 20 (87.0%) | 0.77 |
African American | 7 (14.0%) | 4 (16.0%) | 3 (13.0%) | |
American Indian | 1 (2.0%) | |||
CA 19-9 (U/ML) | ||||
Median CA 19-9 at baseline | 1810 | 511 | 2090 | 0.93 |
<37 | 9 (18.0%) | 3 (12.0%) | 6 (26.1%) | 0.080 |
37–1000 | 13 (26.0%) | 10 (40.0%) | 3 (13.0%) | |
>1000 | 28 (56.0%) | 12 (48.0%) | 14 (60.9%) | |
Metastatic sites | ||||
Liver | 33 (66.0%) | 17 (68.0%) | 16 (69.6%) | 0.91 |
Lung | 8 (16.0%) | 3 (12.0%) | 4 (17.4%) | 0.70 |
Peritoneum | 18 (36.0%) | 8 (32.0%) | 7 (30.0%) | 0.82 |
Lymph nodes | 6 (12.0%) | 3 (12.0%) | 2 (8.7%) | 1.00 |
Bone | 2 (4.0%) | 1 (4.0%) | 1 (4.0%) | 1.00 |
Therapy** | ||||
Gemcitabine | 46 (96.0%) | 23 (92.0%) | 23 (100%) | 0.17 |
FOLFIRINOX | 3 (6.0%) | 2 (8.0%) | 1 (4.3%) | 0.60 |
Gemcitabine/nab-paclitaxel | 1 (2.0%) | 0 (0%) | 1 (4.3%) | 0.29 |
Any Imexon, inj. *** | 21 (42.0%) | 7 (28.0%) | 12 (52.2%) | 0.087 |
50 patients were enrolled in the study. Only 48 patients had samples available for CTC evaluation pre-treatment.
Some patients in this cohort received treatment with multiple agents.
21 untreated mPC patients were enrolled in a Phase Ib study of imexon injection plus gemcitabine38.
Circulating Tumor Cell Yield
Forty-eight patients provided samples that were evaluable for CTC enumeration at baseline, and 37 provided evaluable samples at first disease evaluation. Figure 1 demonstrates CTC yield at baseline and first disease evaluation. At baseline, 23 of 48 patients (48%) had ≥ 1 CTC/7.5mL peripheral blood. Thirty percent (11 of 37 patients) had ≥ 1 CTC/7.5mL peripheral blood at first disease evaluation. Isolation of ≥2 CTC/7.5mL was rare, and was noted in only 6 patients (13%) pre-treatment and 3 patients (8%) at first disease evaluation.
Figure 1. CTC yield among mPC patients pre-treatment and at first disease evaluation.
Peripheral blood CTC yield among 48 patients with available samples for enumeration pre-treatment and 37 patients with available samples for enumeration at first disease evaluation.
Among the 37 patients who submitted samples at the two time points, we evaluated the change in CTC count between the two samples. Of the patients who presented with 0 CTC, 84% (19/22) remained in this category at first disease evaluation. For patients with ≥ 1 CTC at presentation, approximately half had no CTCs detected at first disease evaluation (47%, 7/15 patients). No association was seen between treatment response (partial response, stable disease, or progressive disease) and CTC level at baseline (p=0.66), or at first disease evaluation (p=0.90) (Table 2). No correlation was found between CTC enumeration and absolute CA19-9 levels pre-treatment or at first disease evaluation (Spearman correlation coefficients 0.04 and -0.214, respectively; p>0.20 data not shown).
Table 2.
Association Between Overall Response and CTC Level Pre-treatment (A) and First Disease Evaluation (B)
Pre-treatment | 1st disease evaluation | |||||||
---|---|---|---|---|---|---|---|---|
Response | CTC = 0 (n = 25) | CTC 1 (n = 23) | CTC = 0** (n = 26) | CTC ≥ 1*** (n = 11) | ||||
N | % | N | % | N | % | N | % | |
PR | 5 | 20.0 | 3 | 13.0 | 6 | 23.1 | 2 | 18.2 |
SD | 11 | 44.0 | 9 | 39.1 | 13 | 50.0 | 5 | 45.4 |
PD | 6 | 24.0 | 5 | 21.7 | 7 | 26.9 | 4 | 36.4 |
Not evaluable | 3 | 12.0 | 6 | 26.1 | - | - | - | - |
P Value | P = 0.66 | P = 0.90 |
PR-partial response, SD- stable disease, PD-progressive disease
Patients that were not evaluable and those that did not have CTC enumeration at first disease evaluation were excluded.
Relation of circulating tumor cell yield to overall survival
With a median follow up of 7.5 months (range – 1–24months), the median OS of our cohort was 7.5 months (95% CI: 5.3–9.9 months). Patients who did not have CTC detected at baseline had longer median survival compared to those with ≥ 1 CTC, but this difference was not statistically significant (8.8 months; 95% CI: 6.8–10.9 vs. 6.3 months; 95%CI: 2.6–9.7; p=0.14) (Figure 2A). At first disease evaluation, 37 patients provided follow-up samples for CTC enumeration. A similar non-significant difference was noted in the median OS of patients without CTC and patients with ≥ 1 CTC/7.5mL at first disease evaluation [9.6 months (95% CI: 7.1–10.9 months) vs. 8.8 months (95%CI: 2.9–13.8) respectively; p=0.25] (Figure 2B).
Figure 2. Overall survival by CTC level pre-treatment.
OS based on CTC count at baseline (A) and first disease evaluation (B). (A) Kaplan-Meier curve estimates of overall survival based on CTC enumeration pre-treatment (0 vs. ≥ 1); N=48; median OS 8.8 months vs. 6.3months, respectively; p=0.139. (B) Kaplan-Meier curve estimates of OS based on CTC enumeration at first disease evaluation (0 vs. ≥1); N=37; Median OS 9.6 months vs 8.8 months; respectively; p=0.25.
Relation of MUC-1 expression in circulating tumor cells to overall survival
MUC-1 expression was found in 10 samples (43%) of the 23 patients with ≥ 1 CTC at baseline. Patients with MUC-1 positive CTC had shorter median OS [2.7 months (95%CI: 0.1–7.6)] compared to patients with MUC-1 negative CTC [9.6 months (95%CI: 3.9–12.8); p=0.044] or no isolated CTC [8.8 months (95%CI: 6.0–10.9) p=0.014] (Figure 3).
Figure 3. Overall survival by MUC-1 expression at baseline.
OS based on MUC-1 expression. Kaplan-Meier curve estimates of OS based on CTC enumeration and MUC-1 expression pre-treatment (CTC=0 (n=25) – mOS=8.8 months; MUC-1 positive CTC (n=10)- mOS=2.7 months; MUC-1 negative CTC (n=13) – mOS=9.6 months; p=0.014). Median follow-up 7.5 m (range 1–24 months).
Discussion
The range of CTC detection among pancreatic cancer patients is between 30–100% across different studies using various methods22–26. The initial studies evaluating CTC detection using the CellSearch® system demonstrated that up to 40% of pancreatic cancer patients with various disease stages had detectable CTC 20,27. Available data regarding the correlation between CTC number and clinical outcomes is conflicting. Soeth et al. demonstrated a statistically significant correlation between preoperative CTC detection and decreased OS among early stage pancreatic cancer patients23. Similarly Kurihara et al. reported worse survival among advanced pancreatic cancer patients with ≥ 1 CTC27. In a recent meta-analysis of 9 cohort studies with a total of 623 pancreatic cancer patients of various disease stages, CTC positivity was associated with poorer progression-free survival (HR=1.89, 95 %CI: 1.25–4.0, p<0.001), and worse OS (HR=1.23, 95%CI: 0.88–2.08, p<0.001)28. Other groups failed to show any correlation between clinical outcomes and CTC enumeration in the locally advanced or metastatic setting29, 30.
In this pilot study we prospectively evaluated the ability to characterize CTC enriched by the CellSearch® system from the peripheral blood of mPC patients and assess a correlation with clinical outcomes. Although data from the original cohort of patients evaluated by this assay found 6 of 16 patients with pancreatic cancer with ≥ 3.5 CTC/7.5ml of peripheral blood, our results demonstrate a lower rate of CTC enumeration 20. For the current study, we utilized a threshold of ≥ 1 CTC to declare a sample positive. In our cohort, 48% had ≥ 1 CTC/7.5ml peripheral blood and very few patients had ≥ 2 CTC/7.5ml. This yield is less than that noted in other epithelial malignancies such as metastatic breast, colon and prostate with 30–65 percent of patient having ≥ 5 CTC/7.5 ml peripheral blood6,8,10. Differences in biology between pancreatic cancer and other epithelial malignancies likely play an important role in CTC recovery. Specifically, the lower rate of hematogenous spread of metastatic disease in pancreatic cancer as compared to other cancers31,32. The lower percentage of mPC patients with detectable CTC mirrors data from other groups studying CTC in pancreatic cancer10,25,26,33.
The threshold of ≥ 1 CTC has been used in previous studies evaluating CTC in pancreatic cancer patients. Bidard et al analyzed a cohort of 79 patients with locally advanced pancreatic cancer for the presence of ≥ 1 CTC pre-treatment using the CellSearch® system and detected CTC in 11% of patients overall (5% at presentation and 9% after 2 months of therapy). CTC positivity was associated with poor differentiated and shorter OS33. Similarly, in our study, patients with ≥ 1 CTC demonstrated a trend towards worse OS compared to patients with no CTC isolated. This trend is most clearly seen in the first 3 months of follow up (Figure 3), yet it did not translate into a survival benefit. The small sample size may have limited our ability to detect a survival benefit in this pilot study. Furthermore, the results may change if tested on a cohort of mPC patients treated with novel therapeutic regimens.
The isolation of CTC represents a potential platform for drug development and molecular analysis of cancers, and provides a non-invasive method to study the tumor in-vivo34. Our study evaluated the prognostic value of MUC-1 expression on isolated CTC, which was found in 43% of patients. We demonstrated a statistically significant difference in OS between patients with MUC-1 positive versus MUC-1 negative CTC. This finding is supported by other biologic observations, as MUC-1 is thought to enhance metastatic properties of pancreatic cancer cells and promote mesenchymal transition. MUC-1 expression was shown to correlate with worst outcomes13,14. We could not perform an analysis of MUC-1 expression on corresponding tumor tissue due to lack of available specimens, as most patients had limited material from a diagnostic fine needle aspiration. As there are several investigational agents targeting MUC-1 in clinical trials, the ability to test the expression of this protein on CTC as a “liquid biopsy” could be utilized in the future to select treatments for patients or measure treatment response16,19,35.
Our study has several limitations. As a pilot study, it is by its nature limited from drawing firm conclusions due to the small sample size and the heterogeneity of delivered chemotherapy. Given the trend of an association between CTC and OS, a larger cohort of patients would be required to reach a definitive conclusion. The low CTC yield in our study raises potential questions regarding the sensitivity of the assay. Utilizing a lower threshold of ≥ 1 CTC increases the rate of “positivity” and the risk of error. Given the potential utility of CTC evaluation in this disease, in which abundant tissue availability is often limited, additional studies are needed to develop more sensitive platforms and novel techniques for CTC isolation in pancreatic cancer36,37.
In summary, this pilot study demonstrates the feasibility of capturing and characterizing CTC with the CellSearch® system in patients with mPC. The sample size was too small to make definitive conclusions regarding the association of CTC number and survival. However, our data suggests that patients with MUC-1 positive CTC may have shorter survival than those with MUC-1 negative CTC. Further studies to test the prognostic impact of CTCs and their potential as a pharmcodynamic marker for MUC-1 targeted therapies in pancreatic cancer are warranted.
Acknowledgments
Supported by ACS MRSG-06-003-01-CCE and by Cancer Center Support Grant 3 P30 CA006927-47S4
Footnotes
Prior presentation: Presented in part at the Annual Meeting of the American Society of Clinical Oncology, June 4–10, 2010, Chicago, Illinois
Conflict of Interest: No conflict of interest to report.
References
- 1.Siegel R, Ma J, Zou Z, et al. Cancer statistics, 2014. CA Cancer J Clin. 2014;64:9–29. doi: 10.3322/caac.21208. [DOI] [PubMed] [Google Scholar]
- 2.Hidalgo M. Pancreatic cancer. N Engl J Med. 2010;362:1605–1617. doi: 10.1056/NEJMra0901557. [DOI] [PubMed] [Google Scholar]
- 3.Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–1703. doi: 10.1056/NEJMoa1304369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–1825. doi: 10.1056/NEJMoa1011923. [DOI] [PubMed] [Google Scholar]
- 5.Alix-Panabieres C, Pantel K. Challenges in circulating tumour cell research. Nat Rev Cancer. 2014;14:623–631. doi: 10.1038/nrc3820. [DOI] [PubMed] [Google Scholar]
- 6.Cristofanilli M, Budd GT, Ellis MJ, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;351:781–791. doi: 10.1056/NEJMoa040766. [DOI] [PubMed] [Google Scholar]
- 7.Hayes DF, Cristofanilli M, Budd GT, et al. 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. doi: 10.1158/1078-0432.CCR-05-2821. [DOI] [PubMed] [Google Scholar]
- 8.Moreno JG, Miller MC, Gross S, et al. Circulating tumor cells predict survival in patients with metastatic prostate cancer. Urology. 2005;65:713–718. doi: 10.1016/j.urology.2004.11.006. [DOI] [PubMed] [Google Scholar]
- 9.Cohen SJ, Alpaugh RK, Gross S, et al. Isolation and characterization of circulating tumor cells in patients with metastatic colorectal cancer. Clin Colorectal Cancer. 2006;6:125–132. doi: 10.3816/CCC.2006.n.029. [DOI] [PubMed] [Google Scholar]
- 10.Cohen SJ, Punt CJ, Iannotti N, 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. 2008;26:3213–3221. doi: 10.1200/JCO.2007.15.8923. [DOI] [PubMed] [Google Scholar]
- 11.Cohen SJ, Punt CJ, Iannotti N, et al. Prognostic significance of circulating tumor cells in patients with metastatic colorectal cancer. Ann Oncol. 2009;20:1223–1229. doi: 10.1093/annonc/mdn786. [DOI] [PubMed] [Google Scholar]
- 12.Masaki Y, Oka M, Ogura Y, et al. Sialylated MUC1 mucin expression in normal pancreas, benign pancreatic lesions, and pancreatic ductal adenocarcinoma. Hepato-gastroenterology. 1999;46:2240–2245. [PubMed] [Google Scholar]
- 13.Roy LD, Sahraei M, Subramani DB, et al. MUC1 enhances invasiveness of pancreatic cancer cells by inducing epithelial to mesenchymal transition. Oncogene. 2011;30:1449–1459. doi: 10.1038/onc.2010.526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hinoda Y, Ikematsu Y, Horinochi M, et al. Increased expression of MUC1 in advanced pancreatic cancer. J Gastroenterol. 2003;38:1162–1166. doi: 10.1007/s00535-003-1224-6. [DOI] [PubMed] [Google Scholar]
- 15.Hamanaka Y, Suehiro Y, Fukui M, et al. Circulating anti-MUC1 IgG antibodies as a favorable prognostic factor for pancreatic cancer. Int J Cancer. 2003;103:97–100. doi: 10.1002/ijc.10801. [DOI] [PubMed] [Google Scholar]
- 16.Gold DV, Karanjawala Z, Modrak DE, et al. PAM4-reactive MUC1 is a biomarker for early pancreatic adenocarcinoma. Clin Cancer Res. 2007;13:7380–7387. doi: 10.1158/1078-0432.CCR-07-1488. [DOI] [PubMed] [Google Scholar]
- 17.Rong Y, Jin D, Wu W, et al. Induction of protective and therapeutic anti-pancreatic cancer immunity using a reconstructed MUC1 DNA vaccine. BMC Cancer. 2009;9:191. doi: 10.1186/1471-2407-9-191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Rong Y, Qin X, Jin D, et al. A phase I pilot trial of MUC1-peptide-pulsed dendritic cells in the treatment of advanced pancreatic cancer. Clin Exp Med. 2012;12:173–180. doi: 10.1007/s10238-011-0159-0. [DOI] [PubMed] [Google Scholar]
- 19.Gold DV, Modrak DE, Ying Z, et al. New MUC1 serum immunoassay differentiates pancreatic cancer from pancreatitis. J Clin Oncol. 2006;24:252–258. doi: 10.1200/JCO.2005.02.8282. [DOI] [PubMed] [Google Scholar]
- 20.Allard WJ, Matera J, Miller MC, et al. 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;10:6897–6904. doi: 10.1158/1078-0432.CCR-04-0378. [DOI] [PubMed] [Google Scholar]
- 21.Kagan M, Howard D, Bendele T, et al. A sample preparation and analysis system for identification of circulating tumor cells. J Clinical Ligand Assay. 2002;25:104–110. [Google Scholar]
- 22.Ko A, Scott J, Tempero M, et al. Detection and significance of circulating tumor cells in patients with metastatic pancreatic cancer receiving systmic therapy. J Clin Oncol. 2007;25:4596. abstract. [Google Scholar]
- 23.Soeth E, Grigoleit U, Moellmann B, et al. Detection of tumor cell dissemination in pancreatic ductal carcinoma patients by CK 20 RT-PCR indicates poor survival. J Cancer Res Clin Oncol. 2005;131:669–676. doi: 10.1007/s00432-005-0008-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Nagrath S, Sequist LV, Maheswaran S, et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature. 2007;450:1235–1239. doi: 10.1038/nature06385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ren C, Han C, Zhang J, et al. Detection of apoptotic circulating tumor cells in advanced pancreatic cancer following 5-fluorouracil chemotherapy. Cancer Biol Ther. 2011;12:700–706. doi: 10.4161/cbt.12.8.15960. [DOI] [PubMed] [Google Scholar]
- 26.Marrinucci D, Bethel K, Kolatkar A, et al. Fluid biopsy in patients with metastatic prostate, pancreatic and breast cancers. Phys Biol. 2012;9:016003. doi: 10.1088/1478-3975/9/1/016003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kurihara T, Itoi T, Sofuni A, et al. Detection of circulating tumor cells in patients with pancreatic cancer: a preliminary result. J Hepatobiliary Pancreat Surg. 2008;15:189–195. doi: 10.1007/s00534-007-1250-5. [DOI] [PubMed] [Google Scholar]
- 28.Han L, Chen W, Zhao Q. Prognostic value of circulating tumor cells in patients with pancreatic cancer: a meta-analysis. Tumour Biol. 2014;35:2473–2480. doi: 10.1007/s13277-013-1327-5. [DOI] [PubMed] [Google Scholar]
- 29.Khoja L, Backen A, Sloane R, et al. A pilot study to explore circulating tumour cells in pancreatic cancer as a novel biomarker. Br J Cancer. 2012;106:508–516. doi: 10.1038/bjc.2011.545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.de Albuquerque A, Kubisch I, Breier G, et al. Multimarker gene analysis of circulating tumor cells in pancreatic cancer patients: a feasibility study. Oncology. 2012;82:3–10. doi: 10.1159/000335479. [DOI] [PubMed] [Google Scholar]
- 31.Braun S, Pantel K, Muller P, et al. Cytokeratin-positive cells in the bone marrow and survival of patients with stage I, II, or III breast cancer. N Engl J Med. 2000;342:525–533. doi: 10.1056/NEJM200002243420801. [DOI] [PubMed] [Google Scholar]
- 32.Weckermann D, Muller P, Wawroschek F, et al. Micrometastases of bone marrow in localized prostate cancer: correlation with established risk factors. J Clin Oncol. 1999;17:3438–3443. doi: 10.1200/JCO.1999.17.11.3438. [DOI] [PubMed] [Google Scholar]
- 33.Bidard FC, Huguet F, Louvet C, et al. Circulating tumor cells in locally advanced pancreatic adenocarcinoma: the ancillary CirCe 07 study to the LAP 07 trial. Ann Oncol. 2013;24:2057–2061. doi: 10.1093/annonc/mdt176. [DOI] [PubMed] [Google Scholar]
- 34.Yu KH, Ricigliano M, Hidalgo M, et al. Pharmacogenomic modeling of circulating tumor and invasive cells for prediction of chemotherapy response and resistance in pancreatic cancer. Clin Cancer Res. 2014;20:5281–5289. doi: 10.1158/1078-0432.CCR-14-0531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ramanathan RK, Lee KM, McKolanis J, et al. Phase I study of a MUC1 vaccine composed of different doses of MUC1 peptide with SB-AS2 adjuvant in resected and locally advanced pancreatic cancer. Cancer Immunol Immunother. 2005;54:254–264. doi: 10.1007/s00262-004-0581-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sheng W, Ogunwobi OO, Chen T, et al. Capture, release and culture of circulating tumor cells from pancreatic cancer patients using an enhanced mixing chip. Lab Chip. 2014;14:89–98. doi: 10.1039/c3lc51017d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kulemann B, Pitman MB, Liss AS, et al. Circulating tumor cells found in patients with localized and advanced pancreatic cancer. Pancreas. 2015;44:547–550. doi: 10.1097/MPA.0000000000000324. [DOI] [PubMed] [Google Scholar]
- 38.Cohen SJ, Zalupski MM, Modiano MR, et al. A phase I study of imexon plus gemcitabine as first-line therapy for advanced pancreatic cancer. Cancer Chemother Pharmacol. 2010;66:287–294. doi: 10.1007/s00280-009-1162-y. [DOI] [PMC free article] [PubMed] [Google Scholar]