Circulating tumor cell

Circulating tumor cells (CTCs) are cells that have shed into the vasculature from a primary tumor and circulate in the bloodstream. CTCs thus constitute seeds for subsequent growth of additional tumors (metastasis) in vital distant organs, triggering a mechanism that is responsible for the vast majority of cancer-related deaths.[1]

CTCs were observed for the first time in 1869 in the blood of a man with metastatic cancer by Thomas Ashworth, who postulated that “cells identical with those of the cancer itself being seen in the blood may tend to throw some light upon the mode of origin of multiple tumours existing in the same person”. A thorough comparison of the morphology of the circulating cells to tumor cells from different lesions led Ashworth to conclude that “One thing is certain, that if they [CTC] came from an existing cancer structure, they must have passed through the greater part of the circulatory system to have arrived at the internal saphena vein of the sound leg”.[2]

The importance of CTC's in modern cancer research began in the mid 1990's with the demonstration [J. Uhr, UT-Dallas, L. Terstappen and P. Liberti, Immunicon, Philadelphia] that CTC's exist early on in the course of the disease. Those results were made possible by exquisitely sensitive magnetic separation technology employing Ferrofluids (colloidal magnetic nanoparticles) and high gradient magnetic separators invented by Liberti at Immunicon and motivated by theoretical calculations by Liberti and Terstappen that indicated very small tumors shedding cells at less than 1.0% per day should result in detectable cells in blood. A variety of other technologies have been applied to CTC enumeration and identification since that time.

Modern cancer research has demonstrated that CTCs derive from clones in the primary tumor, validating Ashworth's remarks.[3] The significant efforts put into understanding the CTCs biological properties have demonstrated the critical role circulating tumor cells play in the metastatic spread of carcinoma.[4] Furthermore, highly sensitive, single-cell analysis demonstrated a high level of heterogeneity seen at the single cell level for both protein expression and protein localization and the CTCs reflected both the primary biopsy and the changes seen in the metastatic sites.

Tissue biopsies are poor diagnostic procedures: they are invasive, cannot be used repeatedly, and are ineffective in understanding metastatic risk, disease progression, and treatment effectiveness.[5] CTCs thus could be considered a “liquid biopsy” which reveals metastasis in action, providing live information about the patient’s disease status.[6] Analysis of blood samples found a propensity for increased CTC detection as the disease progressed in individual patients.[7] Blood tests are easy and safe to perform and multiple samples can be taken over time. By contrast, analysis of solid tumors necessitates invasive procedures that might limit patient compliance. The ability to monitor the disease progression over time could facilitate appropriate modification to a patient's therapy, potentially improving their prognosis and quality of life. The important aspect of the ability to prognose the future progression of the disease is elimination (at least temporarily) of the need for a surgery when the repeated CTC counts are low and not increasing; the obvious benefits of avoiding the surgery include avoiding the risk related to the innate tumor-genicity of cancer surgeries. To this end, technologies with the requisite sensitivity and reproducibility to detect CTCs in patients with metastatic disease have recently been developed.[8][9][10][11][12][13][14][15]

Types of CTCs

1. Traditional CTCs are confirmed cancer cells with an intact, viable nucleus; express cytokeratins, which demonstrate epithelial origin; have an absence of CD45, indicating the cell is not of hematopoietic origin; and are often larger cells with irregularity shape or subcellular morphology.[16]

2. Cytokeratin negative (CK-) CTCs are cancer stem cells or cells undergoing epithelial-mesenchymal transition (EMT). CK-CTCs may be the most resistant and most prone to metastasis; express neither cytokeratins nor CD45; have morphology similar to a cancer cell; and importantly have gene or protein expression or genomics associated with cancer.[17]

3. Apoptotic CTCs are traditional CTCs that are undergoing apoptosis (cell death): Epic Sciences technology identifies nuclear fragmentation or cytoplasmic blebbing associated with apoptosis. Measuring the ratio of traditional CTC to apoptotic CTCs – from baseline to therapy – provides clues to a therapy’s efficacy in targeting and killing cancer cells.[17]

4. Small CTCs are cytokeratin positive and CD45 negative, but with sizes and shapes similar to white blood cells. Importantly, small CTCs have cancer-specific biomarkers that identify them as CTCs. Small CTCs have been implicated in progressive disease and differentiation into small cell carcinomas, which often require a different therapeutic course.

5. CTC Clusters are two or more individual CTCs bound together. The CTC cluster may contain traditional, small or CK- CTCs. These clusters have cancer-specific biomarkers that identify them as CTCs. These clusters are associated with increased metastatic risk and poor prognosis.[18]

Frequency of CTCs

Figure 1: cell number of various blood cells in whole blood versus CTC

The detection of CTCs may have important prognostic and therapeutic implications but because their numbers can be very small, these cells are not easily detected.[19] It is estimated that among the cells that have detached from the primary tumor, only 0.01% can form metastases.[20]

Circulating tumor cells are found in frequencies on the order of 1-10 CTC per mL of whole blood in patients with metastatic disease.[21] For comparison, a mL of blood contains a few million white blood cells and a billion red blood cells, see figure 1. This low frequency, associated to difficulty of identifying cancerous cells, means that a key component of understanding CTCs biological properties require technologies and approaches capable of isolating 1 CTC per mL of blood, either by enrichment, or better yet with enrichment-free assays that identify all CTC subtypes in sufficiently high definition to satisfy diagnostic pathology image-quantity requirements in patients with a variety of cancer types.[17] To date CTCs have been detected in several epithelial cancers (breast, prostate, lung, and colon)[22][23][24][25] and clinical evidences indicate that patients with metastatic lesions are more likely to have CTCs isolated.

CTCs are usually (in 2011) captured from the vasculature by using specific antibodies able to recognize specific tumoral marker (usually EpCAM);[26] however this approach is biased by the need for a sufficient expression of the selected protein on the cell surface, event necessary for the enrichment step. Moreover, since EpCAM and other proteins (e.g. cytokeratins) are not expressed in some tumors and can be down regulated during the epithelial to mesenchymal transition (EMT), new enrichment strategies are required.[27]

First evidence indicates that CTC markers applied in human medicine are conserved in other species. Five of the more common markers including CK19 are also useful to detect CTC in the blood of dogs with malignant mammary tumors.[28][29]

Newer approaches are able to identify more cells out 7.5 ml of blood, like isoflux or maintrac.[30][31]

Clinical Utility

Figure 2: Kaplan Meier Analysis of overall survival before starting a new line of therapy for patients with metastatic breast, colorectal and prostate cancer. Patients were divided into those with Favorable and Unfavorable CTC (Unfavorable: >5 CTC/7.5mL for breast and prostate, >3 CTC/7.5mL for colon) [32]

To date, a variety of research methods have been developed to isolate and enumerate CTC.[33] The only U.S. Food and Drug Administration (FDA) cleared methodology for enumeration of CTC in whole blood is the CellSearch system.[34] Extensive clinical testing done using this method shows that presence of CTC is a strong prognostic factor for overall survival in patients with metastatic breast, colorectal or prostate cancer, see figure 2 [35][36][37][38][39][40][41]

Detection Methods

CellSearch Method

This method is based on the use of iron nano-particles coated with a polymer layer carrying biotin analogues and conjugated with antibodies anti EpCAM for capturing CTCs, and on the use of an analyzer to take images of isolated cells upon their staining with specific fluorescent antibody conjugates. Blood is sampled in an EDTA tube with an added preservative. Upon arrival in the lab, 7.5mL of blood is centrifuged and placed in a preparation system. This system first enriches the tumor cells immunomagnetically by means of ferrofluid nano-particles and a magnet. Subsequently recovered cells are permeabilized and stained with a nuclear stain, a fluorescent antibody conjugate against CD45 (leukocyte marker), and cytokeratin 8, 18 and 19 (CKs). The sample is then scanned on an analyzer which takes images of the nuclear, cytokeratin, and CD45 stains.[42] To be considered a CTC a cell must contain a nucleus, be positive for cytoplasmic expression of cytokeratin as well as negative for the expression of CD45 marker, and have a diameter larger than 5 µm. If the total number of tumor cells found to meet the criteria cited above is 5 or more, a blood sample is positive. In studies done on prostate, breast and colon cancer patients, median survival of metastatic patients with positive samples is about half the median survival of metastatic patients with negative samples. This system is characterized by a recovery capacity of 93% and a detection limit of one CTC per 7.5 mL of whole blood. Despite its sensitivity and reproducibility, the CellSearch Method requires specific equipment to perform the analysis.

Epic Sciences Method

This method involves technology to separate nucleated cells from red blood cells, which lack a nucleus. All nucleated cells, including normal white blood cells and CTCs, are exposed to fluorescent-tagged antibodies specific for cancer biomarkers. In addition, Epic’s imaging system captures pictures of all the cells on the slide (approximately 3 million), records the precise coordinates of each cell, and analyzes each cell for 90 different parameters, including the fluorescence intensity of the four fluorescent markers and 86 different morphological parameters. Epic can also use FISH and other staining techniques to look for abnormalities such as duplications, deletions, and rearrangements. The imaging and analysis technology also allows for the coordinates of every cell on a slide to be known so that a single cell can be retrieved from the slide for analysis using next-generation sequencing. A hematopathology-trained algorithm incorporates numerous morphology measurements as well as expression from cytokeratin and CD45. The algorithm then proposes candidate CTCs that a trained reader confirms. Cells of interest are analyzed for relevant phenotypic and genotypic markers, with regional white blood cells included as negative controls.[43] Epic’s molecular assays measure protein expression and also interrogate genomic abnormalities in CTCs for more than 20 different cancer types.

maintrac

Maintrac is a diagnostic platform applying microscopic methods to identify rare cells in body fluids and their molecular characteristics.

Concerning circulating tumor cells, maintrac is using an approach based on microscopic identification of circulating tumor cells. To prevent damage and loss of the cells during the process, maintrac uses just two steps towards the identification. In contrast to many other methods, maintrac does not purify the cells or enriches them, but identifies them within the context of the other blood compounds. To obtain vital cells and to reduce stress of those cells, blood cells are prepared by only one centrifugation step and erythrocyte lysis. Like CellSearch maintrac uses an EpCAM antibody. It is, however, not used for enrichment but rather as a fluorescent marker to identify those cells. Together with the nuclear staining with propidium iodide the maintrac method can distinguish between dead and living cells. Only vital, propidium excluding EpCAM positive cells are counted as potential tumor cells. Only living cells can grow into tumors, therefore dying EpCAM positive cells can do no harm. The suspension is analysed by fluorescence microscopy, which automatically counts the events. Simultaneous event galleries are recorded to verify whether the software found a true living cell and to differentiate between skin epithelial cells for example. Close validation of the method showed that additional antibodies of cytokeratins or CD45 did not have any advantage.[31][44]

Unlike other methods maintrac does not use the single cell count as a prognostic marker, rather maintrac utilizes the dynamics of the cell count. Rising tumor cell numbers are an important factor that tumor activity is ongoing. Decreasing cell counts are a sign for a successful therapy.

Therefore maintrac can be used in following situations:

[47]

Circulating tumor cells as early diagnosis for cancer recurrence:[48][49]

Studies showed that under certain circumstances also EpCAM positive cells can be found in the blood.[50] Inflammation diseases like Morbus Crohn or Colitits Ulcerosa also show increased levels of EpCAM positive cells. Patients with severe skin burns can also carry EpCAM positive cells in the blood, which may falsify results. So use of EpCAM positive cells as a tool for early diagnosis is not recommended.

Other Methods

CTCs are pivotal to understanding the biology of metastasis and promise potential as a biomarker to noninvasively evaluate tumor progression and response to treatment. However, isolation and characterization of CTCs represent a major technological challenge, since CTCs make up a minute number of the total cells in circulating blood, 1–10 CTCs per mL of whole blood compared to a few million white blood cells and a billion red blood cells.[51] Therefore the major challenge for CTC researchers is the prevailing difficulty of CTC purification that allows the molecular characterization of CTCs. Several methods have been developed to isolate CTCs in the peripheral blood and essentially fall into two categories: biological methods and physical methods.

Another approach is the in vivo capture of CTCs of GILUPI GmbH.[57][58] An antibody coated metal wire is inserted into a peripheral vein and stays there for a defined period (30 min). During this time, CTCs from the blood can bind to the antibodies (currently anti-EpCAM). After the incubation time, the wire is removed, washed and the native CTCs, isolated from the blood of the patient, can be further analysed. Molekular genetics as well as immunoflourescent staining and several other methods are possible.[59][60] Advantage of this method is the higher blood volume that can be analysed for CTCs (approx. 750 ml in 30 min compared to 7.5 ml of a drawn blood sample).

Characterization of CTC

Any useful method for isolation of CTCs must allow (i) their identification and enumeration and (ii) their characterization through immunocytochemistry, fluorescence in situ hybridization (FISH) DNA and RNA assays, and all other relevant molecular techniques using DNA and RNA. When circulating tumor cells are captured from blood using filtration devices (such as ScreenCell isolation device), further morphological and molecular characterization is required to reveal important predictive information and report changes in CTC biology, for example during tumor relapse. ViewRNA assay for CTCs characterization is the only in situ hybridization technology that allows multiplex, single-molecule RNA detection of any RNA target. The exceptional sensitivity and specificity is achieved by using proprietary probe design, simultaneous branched DNA (bDNA) signal amplification and background suppression.

CTCs characterization using ViewRNA for multiplex in situ RNA analysis

The captured CTCs on the filter membrane of a ScreenCell isolation device, are transferred to a 24-well cell culture plate for enumeration/characterization using ViewRNA ISH Cell Assay. A target-specific probe set containing 20 oligonucleotide pairs hybridizes to the target RNA. An oligo pair hybridization event is essential for support of the signal amplification structure, which is assembled by a series of sequential hybridization steps. Each fully assembled amplification structure is contained within 40−50 bp of target RNA with the capacity for 400-fold signal amplification. Therefore, a typical target-specific probe set (containing 20 oligo pairs) can generate 8,000-fold signal amplification at the location of the target RNA.

Further Characterisation of CTC

Some drugs are particularly effective against cancers which fit certain requirements. For example Herceptin is very effective in patients who are Her2 positive, but much less effective in patients who are Her2 negative. Once the primary tumor is removed, biopsy of the current state of the cancer through traditional tissue typing is not possible anymore.[61] Often tissue sections of the primary tumor, removed years prior, are used to do the typing. Further characterisation of CTC may help determining the current tumor phenotype. FISH assays has been performed on CTC to as well as determination of IGF-1R, Her2, Bcl-2, [ERG (gene)|ERG], PTEN, AR status using immunofluorescence.[62][63][64][65][66][67]

Morphological Definition

Morphological appearance is judged by human operators and is therefore subject to large inter operator variation.[68] Several CTC enumeration methods exist which use morphological appearance to identify CTC, which may also apply different morphological criteria. A recent study in prostate cancer showed that many different morphological definitions of circulating tumor cells have similar prognostic value, even though the absolute number of cells found in patients and normal donors varied by more than a decade between different morphological definitions.[69]

See also

References

  1. Gupta, GP; Massagué, J (Nov 17, 2006). "Cancer metastasis: building a framework.". Cell 127 (4): 679–95. doi:10.1016/j.cell.2006.11.001. PMID 17110329.
  2. Ashworth, T. R (1869). "A case of cancer in which cells similar to those in the tumours were seen in the blood after death". Australian Medical Journal 14: 146–7.
  3. Fehm, T; Sagalowsky, A; Clifford, E; Beitsch, P; Saboorian, H; Euhus, D; Meng, S; Morrison, L; Tucker, T; Lane, N; Ghadimi, BM; Heselmeyer-Haddad, K; Ried, T; Rao, C; Uhr, J (Jul 2002). "Cytogenetic evidence that circulating epithelial cells in patients with carcinoma are malignant". Clinical cancer research : an official journal of the American Association for Cancer Research 8 (7): 2073–84. PMID 12114406.
  4. Fidler IJ (2003). "Timeline: The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis revisited". Nature Reviews Cancer 3 (6): 453–8. doi:10.1038/nrc1098. PMID 12778135.
  5. Marrinucci, D; Bethel, K; Luttgen, M; Nieva, J; Kuhn, P; Kuhn, P (Sep 2009). "Circulating tumor cells from well-differentiated lung adenocarcinoma retain cytomorphologic features of primary tumor type". Archives of Pathology & Laboratory Medicine 133 (9): 1468–71. doi:10.1043/1543-2165-133.9.1468 (inactive 2015-04-17). PMID 19722757.
  6. Attard, G; Swennenhuis, JF; Olmos, D; Reid, AH; Vickers, E; A'Hern, R; Levink, R; Coumans, F; Moreira, J; Riisnaes, R; Oommen, NB; Hawche, G; Jameson, C; Thompson, E; Sipkema, R; Carden, CP; Parker, C; Dearnaley, D; Kaye, SB; Cooper, CS; Molina, A; Cox, ME; Terstappen, LW; de Bono, JS (Apr 1, 2009). "Characterization of ERG, AR and PTEN gene status in circulating tumor cells from patients with castration-resistant prostate cancer". Cancer Research 69 (7): 2912–8. doi:10.1158/0008-5472.CAN-08-3667. PMID 19339269.
  7. Cohen, SJ; Punt, CJ; Iannotti, N; Saidman, BH; Sabbath, KD; Gabrail, NY; Picus, J; Morse, M; Mitchell, E; Miller, MC; Doyle, GV; Tissing, H; Terstappen, LW; Meropol, NJ (Jul 1, 2008). "Relationship of circulating tumor cells to tumor response, progression-free survival, and overall survival in patients with metastatic colorectal cancer". Journal of clinical oncology : official journal of the American Society of Clinical Oncology 26 (19): 3213–21. doi:10.1200/JCO.2007.15.8923. PMID 18591556.
  8. Yu M., Haber D. A.; et al. (2012). "RNA sequencing of pancreatic circulating tumour cells implicates WNT signalling in metastasis". Nature 487 (7408): 510–3. Bibcode:2012Natur.487..510Y. doi:10.1038/nature11217. PMC 3408856. PMID 22763454.
  9. Sleijfer S, Gratama JW, Sieuwerts AM; et al. (2007). "Circulating tumour cell detection on its way to routine diagnostic implementation?". Eur J Cancer 43 (18): 2645–50. doi:10.1016/j.ejca.2007.09.016. PMID 17977713.
  10. Hayes DF, Smerage J.; Smerage (2008). "Is There a Role for Circulating Tumor Cells in the Management of Breast Cancer?". Clin Cancer Res 14 (12): 3646–50. doi:10.1158/1078-0432.CCR-07-4481. PMID 18559576.
  11. Pantel K, Alix-Panabières C, Riethdorf; Alix-Panabières; Riethdorf (2009). "Cancer micrometastases". Nature Reviews Clinical Oncology 6 (6): 339–51. doi:10.1038/nrclinonc.2009.44. PMID 19399023.
  12. Pantel K, Riethdorf S.; Riethdorf (2009). "Pathology: are circulating tumor cells predictive of overall survival?". Nature Reviews Clinical Oncology 6 (4): 190–1. doi:10.1038/nrclinonc.2009.23. PMID 19333222.
  13. Panteleakou Z, Lembessis P, Sourla A; et al. (2009). "Detection of circulating tumor cells in prostate cancer patients: methodological pitfalls and clinical relevance". Mol Med 15 (3–4): 101–14. doi:10.2119/molmed.2008.00116. PMC 2600498. PMID 19081770.
  14. Esmaeilsabzali H, Beischlag TV, Cox ME, Parameswaran AM, Park EJ.; Beischlag; Cox; Parameswaran; Park (2013). "Detection and isolation of circulating tumor cells: principles and methods". Biotechnol Adv. 31 (7): 1063–84. doi:10.1016/j.biotechadv.2013.08.016. PMID 23999357.
  15. 1 2 Nieva, J; Wendel, M; Luttgen, MS; Marrinucci, D; Bazhenova, L; Kolatkar, A; Santala, R; Whittenberger, B; Burke, J; Torrey, M; Bethel, K; Kuhn, P (Feb 2012). "High-definition imaging of circulating tumor cells and associated cellular events in non-small cell lung cancer patients: a longitudinal analysis". Physical Biology 9 (1): 016004. Bibcode:2012PhBio...9a6004N. doi:10.1088/1478-3975/9/1/016004. PMC 3388002. PMID 22306961.
  16. Racila, E; Euhus, D; Weiss, AJ; Rao, C; McConnell, J; Terstappen, LW; Uhr, JW (Apr 1998). "Detection and characterization of carcinoma cells in the blood". Proceedings of the National Academy of Sciences 95 (8): 4589–4594. Bibcode:1998PNAS...95.4589R. doi:10.1073/pnas.95.8.4589. PMC 22534. PMID 9539782.
  17. 1 2 3 Marrinucci, Dena; Bethel, Kelly; Kolatkar, Anand; Luttgen, Madelyn; Malchiodi, Michael; Baehring, Franziska; Voigt, Katharina; Lazar, Daniel; Nieva, Jorge; Bazhenova, Lyudmilda; Ko, Andrew; Korn, W. Michael; Schram, Ethan; Coward, Michael; Yang, Xing; Metzner, Thomas; Lamy, Rachelle; Honnatti, Meghana; Yoshioka, Craig; Kunken, Joshua; Petrova, Yelena; Sok, Devin; Nelson, David; Kuhn, Peter (Feb 2012). "Fluid Biopsy in Patients with Metastatic Prostate, Pancreatic and Breast Cancers". Physical Biology 9 (1): 016003. Bibcode:2012PhBio...9a6003M. doi:10.1088/1478-3975/9/1/016003. PMC 3387996. PMID 22306768.
  18. Aceto, Nicola; Bardia, Aditya; Miyamoto, David; Donaldson, Maria; Wittner, Ben; Spencer, Joel; Yu, Min; Pely, Adam; Engstrom, Amanda; Zhu, Huili; Brannigan, Brian; Kapur, Ravi; Stott, Shannon; Shioda, Toshi; Ramaswamy, Sridhar; Ting, David; Lin, Charles; Toner, Mehmet; Haberemail, Daniel; Maheswaranemail, Shyamala (28 Aug 2014). "Circulating Tumor Cell Clusters Are Oligoclonal Precursors of Breast Cancer Metastasis". Cell 158 (5): 1110–22. doi:10.1016/j.cell.2014.07.013. PMID 25171411. |first21= missing |last21= in Authors list (help)
  19. Ghossein RA, Bhattacharya S, Rosai J; Bhattacharya; Rosai (1999). "Molecular detection of micrometastases and circulating tumor cells in solid tumors". Clin. Cancer Res. 5 (8): 1950–60. PMID 10473071.
  20. Zhe, X; Cher M.L.; Bonfil R.D. (2011). "Circulating tumor cells: finding the needle in the haystack". Am J Cancer Res 1 (6): 740–751. PMC 3195935. PMID 22016824.
  21. Miller, MC; Doyle, GV; Terstappen, LW (2010). "Significance of Circulating Tumor Cells Detected by the CellSearch System in Patients with Metastatic Breast Colorectal and Prostate Cancer.". Journal of oncology 2010: 617421. doi:10.1155/2010/617421. PMC 2793426. PMID 20016752.
  22. Swaby, RF; Cristofanilli, M (Apr 21, 2011). "Circulating tumor cells in breast cancer: a tool whose time has come of age.". BMC medicine 9: 43. doi:10.1186/1741-7015-9-43. PMC 3107794. PMID 21510857.
  23. Danila, DC; Fleisher, M; Scher, HI (Jun 15, 2011). "Circulating tumor cells as biomarkers in prostate cancer.". Clinical cancer research : an official journal of the American Association for Cancer Research 17 (12): 3903–12. doi:10.1158/1078-0432.CCR-10-2650. PMID 21680546.
  24. Tanaka, F; Yoneda, K; Kondo, N; Hashimoto, M; Takuwa, T; Matsumoto, S; Okumura, Y; Rahman, S; Tsubota, N; Tsujimura, T; Kuribayashi, K; Fukuoka, K; Nakano, T; Hasegawa, S (Nov 15, 2009). "Circulating tumor cell as a diagnostic marker in primary lung cancer". Clinical cancer research : an official journal of the American Association for Cancer Research 15 (22): 6980–6. doi:10.1158/1078-0432.CCR-09-1095. PMID 19887487.
  25. Negin, BP; Cohen, SJ (Jun 2010). "Circulating tumor cells in colorectal cancer: past, present, and future challenges". Current treatment options in oncology 11 (1–2): 1–13. doi:10.1007/s11864-010-0115-3. PMID 20143276.
  26. Man, Yicun; Wang, Qing; Kemmner, Wolfgang (1 January 2011). "Currently Used Markers for CTC Isolation - Advantages, Limitations and Impact on Cancer Prognosis". Journal of Clinical & Experimental Pathology 01 (1). doi:10.4172/2161-0681.1000102.
  27. Mikolajczyk, SD; Millar, LS; Tsinberg, P; Coutts, SM; Zomorrodi, M; Pham, T; Bischoff, FZ; Pircher, TJ (2011). "Detection of EpCAM-Negative and Cytokeratin-Negative Circulating Tumor Cells in Peripheral Blood.". Journal of oncology 2011: 252361. doi:10.1155/2011/252361. PMC 3090615. PMID 21577258.
  28. da Costa A, Oliveira JT, Gärtner F, Kohn B, Gruber AD, Klopfleisch R.; Oliveira; Gärtner; Kohn; Gruber; Klopfleisch (2010). "Potential markers for detection of circulating canine mammary tumor cells in the peripheral blood". Veterinary Journal 190 (1): 165–8. doi:10.1016/j.tvjl.2010.09.027. PMID 21051248.
  29. da Costa, A (2013). "Multiple RT-PCR markers for the detection of circulating tumour cells of metastatic canine mammary tumours.". Veterinary Journal 196 (1): 34–39. doi:10.1016/j.tvjl.2012.08.021. PMID 23036177.
  30. Harb, W., Fan, A., Tran, T., Danila, D.C., Keys, D., Schwartz, M., and Ionescu-Zanetti, C. (2013). "Mutational Analysis of Circulating Tumor Cells Using a Novel Microfluidic Collection Device and qPCR Assay". Transl. Oncol 6 (6): 528–538. doi:10.1593/tlo.13367. PMC 3799195. PMID 24151533.
  31. 1 2 3 Pachmann K., Camara O., Kavallaris A., Krauspe S., Malarski N., Gajda M., Kroll T., Jorke C., Hammer U., Altendorf-Hofmann A.; et al. (2008). "Monitoring the Response of Circulating Epithelial Tumor Cells to Adjuvant Chemotherapy in Breast Cancer Allows Detection of Patients at Risk of Early Relapse". J. Clin. Oncol 26: 1208–1215. doi:10.1200/JCO.2007.13.6523.
  32. MC Miller, GV Doyle, LWMM Terstappen; Doyle; Terstappen (2010). "Significance of Circulating Tumor Cells detected by the CellSearch System in Patients with Metastatic Breast Colorectal and Prostate Cancer". Journal of Oncology 2010: 1–8. doi:10.1155/2010/617421. PMC 2793426. PMID 20016752.
  33. Paterlini-Brechot P, Benali NL.; Benali (2007). "Circulating tumor cells (CTC) detection: Clinical impact and future directions". Cancer Lett. 253 (2): 180–204. doi:10.1016/j.canlet.2006.12.014. PMID 17314005.
  34. "Veridex CellSearch Website". March 2010. Retrieved 2010-03-14.
  35. "Veridex LLC. CellSearch circulating tumor cell kit premarket notification—expanded indications for use—metastatic prostate cancer" (PDF). March 2010. Retrieved 2010-03-14.
  36. Cristofanilli M, Budd GT, Ellis MJ; et al. (2004). "Circulating Tumor Cells, Disease Progression and Survival in Metastatic Breast Cancer". NEJM 351 (8): 781–91. doi:10.1056/NEJMoa040766. PMID 15317891.
  37. Budd G, Cristofanilli M, Ellis M; et al. (2006). "Circulating Tumor Cells versus Imaging - Predicting Overall Survival in Metastatic Breast Cancer". Clin Can Res 12: 6404–09.
  38. Cohen SJ, Punt CJ, Iannotti N; et al. (2008). "The Relationship of Circulating Tumor Cells to Tumor Response, Progression-Free Survival, and Overall Survival in Patients with Metastatic Colorectal Cancer". JCO 26 (19): 3213–21. doi:10.1200/JCO.2007.15.8923. PMID 18591556.
  39. JS DeBono, HI Scher, RB Montgomery; et al. (2008). "Circulating Tumor Cells (CTC) predict survival benefit from treatment in metastatic castration resistant prostate cancer (CRPC)". Clin Can Res 14 (19): 6302–9. doi:10.1158/1078-0432.CCR-08-0872.
  40. Allard W J, Matera J, Miller MC; et al. (2004). "Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with non-malignant diseases". Clin Can Res 10 (20): 6897–6904. doi:10.1158/1078-0432.CCR-04-0378. PMID 15501967.
  41. Riethdorf; Fritsche, H; Müller, V; Rau, T; Schindlbeck, C; Rack, B; Janni, W; Coith, C; et al. (2007). "Detection of Circulating Tumor Cells in Peripheral Blood of Patients with Metastatic Breast Cancer: A Validation Study of the CellSearch System". Clin Cancer Res 13 (3): 920–8. doi:10.1158/1078-0432.CCR-06-1695. PMID 17289886.
  42. "An Introduction to the CellSearch™" (PDF).
  43. Bethel, Kelly; Luttgen2, Madelyn; Damani, Samir; Kolatkar2, Anand; Lamy, Rachelle; Sabouri-Ghomi, Mohsen; Topol, Sarah; Topol2, Eric; Kuhn, Peter (9 Jan 2014). "Fluid phase biopsy for detection and characterization of circulating endothelial cells in myocardial infarction". Physical Biology 11 (1): 016002. Bibcode:2014PhBio..11a6002B. doi:10.1088/1478-3975/11/1/016002. PMC 4143170. PMID 24406475.
  44. Pachmann K., Camara O., Kavallaris A., Schneider U., Schünemann S., Höffken K. (2005). "Quantification of the response of circulating epithelial cells to neodadjuvant treatment for breast cancer: a new tool for therapy monitoring". Breast Cancer Res. 7: R975–979. doi:10.1186/bcr1328.
  45. Camara O., Rengsberger M., Egbe A., Koch A., Gajda M., Hammer U., Jorke C., Rabenstein C., Untch M., Pachmann K. (2007). "The relevance of circulating epithelial tumor cells (CETC) for therapy monitoring during neoadjuvant (primary systemic) chemotherapy in breast cancer". Ann. Oncol. 18: 1484–1492. doi:10.1093/annonc/mdm206.
  46. Pachmann K., Camara O., Kohlhase A., Rabenstein C., Kroll T., Runnebaum I.B., Hoeffken K. (2010). "Assessing the efficacy of targeted therapy using circulating epithelial tumor cells (CETC): the example of SERM therapy monitoring as a unique tool to individualize therapy". J. Cancer Res. Clin. Oncol 137: 821–828. doi:10.1007/s00432-010-0942-4.
  47. Pachmann K., Camara O., Kroll T., Gajda M., Gellner A.K., Wotschadlo J., Runnebaum I.B. (2011). "Efficacy control of therapy using circulating epithelial tumor cells (CETC) as "Liquid Biopsy": trastuzumab in HER2/neu-positive breast carcinoma". J. Cancer Res. Clin. Oncol 137: 1317–1327. doi:10.1007/s00432-011-1000-6.
  48. Hekimian K., Meisezahl S., Trompelt K., Rabenstein C., Pachmann K. (2012). "Epithelial Cell Dissemination and Readhesion: Analysis of Factors Contributing to Metastasis Formation in Breast Cancer". ISRN Oncol 2012: 1–8. doi:10.5402/2012/601810. PMC 3317055. PMID 22530147.
  49. Rolle A., Günzel R., Pachmann U., Willen B., Höffken K., Pachmann K. (2005). "Increase in number of circulating disseminated epithelial cells after surgery for non-small cell lung cancer monitored by MAINTRAC is a predictor for relapse: A preliminary report". World J. Surg. Oncol 3: 18. doi:10.1186/1477-7819-3-18. PMC 1087511. PMID 15801980.
  50. Camara Oumar, Kavallaris Andreas, Nöschel Helmut, Rengsberger Matthias, Jörke Cornelia, Pachmann Katharina (2006). "Seeding of Epithelial Cells into Circulation During Surgery for Breast Cancer: The Fate of Malignant and Benign Mobilized Cells". World Journal of Surgical Oncology 4: 67. doi:10.1186/1477-7819-4-67.
  51. Yu M.; et al. (2011). "Circulating tumor cells: approaches to isolation and characterization". The journal of Cell Biology 192 (3): 373–382. doi:10.1083/jcb.201010021. PMC 3101098. PMID 21300848.
  52. Nagrath, Sunitha; Sequist, Lecia V.; Maheswaran, Shyamala; Bell, Daphne W.; Irimia, Daniel; Ulkus, Lindsey; Smith, Matthew R.; Kwak, Eunice L.; Digumarthy, Subba; Muzikansky, Alona; Ryan, Paula; Balis, Ulysses J.; Tompkins, Ronald G.; Haber, Daniel A.; Toner, Mehmet (December 2007). "Isolation of rare circulating tumour cells in cancer patients by microchip technology". Nature 450 (7173): 1235–1239. Bibcode:2007Natur.450.1235N. doi:10.1038/nature06385. PMC 3090667. PMID 18097410.
  53. Hoshino, Kazunori; Huang, Yu-Yen; Lane, Nancy; Huebschman, Michael; Uhr, Jonathan W.; Frenkel, Eugene P.; Zhang, Xiaojing (Oct 2011). "Microchip-based immunomagnetic detection of circulating tumor cells". Lab on a Chip 11 (20): 3449–3457. doi:10.1039/c1lc20270g. PMID 21863182.
  54. Peng, Chen; Yu-yen, Huang; Hoshino, Kazunori; Xiaojing, Zhang (2014). "Multiscale immunomagnetic enrichment of circulating tumor cells: from tubes to microchips". Lab on a Chip 14: 446–458. doi:10.1039/C3LC51107C.
  55. Wang, Huiqiang; Chen, Nanhai G.; Minev, Boris R.; Zimmermann, Martina; Aguilar, Richard J.; Zhang, Qian; Sturm, Julia B.; Fend, Falko; Yu, Yong A.; Cappello, Joseph; Lauer, Ulrich M.; Szalay, Aladar A. (September 2013). "Optical Detection and Virotherapy of Live Metastatic Tumor Cells in Body Fluids with Vaccinia Strains". PLoS ONE 8 (9): e71105. Bibcode:2013PLoSO...871105W. doi:10.1371/journal.pone.0071105. PMID 24019862.
  56. Desitter I.; et al. (2011). "A New Device for Rapid Isolation by Size and Characterization of Rare Circulating Tumor Cells". Anticancer research 31 (2): 427–442.
  57. http://www.gilupi.de
  58. Saucedo-Zeni, Nadia, et al. "A novel method for the in vivo isolation of circulating tumor cells from peripheral blood of cancer patients using a functionalized and structured medical wire." International journal of oncology 41.4 (2012): 1241-1250. http://www.ncbi.nlm.nih.gov/pubmed/22825490
  59. Luecke, Klaus, et al. "The GILUPI CellCollector as an in vivo tool for circulating tumor cell enumeration and molecular characterization in lung cancer patients." ASCO Annual Meeting Proceedings. Vol. 33. No. 15_suppl. 2015. http://hwmaint.meeting.ascopubs.org/cgi/content/abstract/33/15_suppl/e22035
  60. Scheumann, N., et al. "50PENUMERATION AND MOLECULAR CHARACTERIZATION OF CIRCULATING TUMOR CELLS IN LUNG CANCER PATIENTS USING THE GILUPI CELLCOLLECTOR™, AN EFFECTIVE IN VIVO DEVICE FOR CAPTURING CTCS." Annals of Oncology 26.suppl 1 (2015): i14-i14. http://annonc.oxfordjournals.org/content/26/suppl_1/i14.1.short
  61. Meng S, Tripathy D, Shete S; et al. (2004). "HER-2 Gene Amplification can be acquired as breast cancer progresses". PNAS 101 (25): 9393–8. Bibcode:2004PNAS..101.9393M. doi:10.1073/pnas.0402993101. PMC 438987. PMID 15194824.
  62. Hayes DF, Walker TM, Singh B; et al. (2002). "Monitoring Expression of HER-2 on Circulating Epithelial Cells in Patients with advanced Breast Cancer". Int J of Oncology 21: 1111–8.
  63. O'Hara SM, Moreno JG, Zweitzig DR; et al. (2004). "Multigene Reverse Transcription-PCR Profiling of Circulating Tumor Cells in Hormone-Refractory Prostate Cancer". Clin Chem 50 (5): 826–835. doi:10.1373/clinchem.2003.028563. PMID 14988224.
  64. de Bono JS, Attard G, Adjei A; et al. (2007). "Potential Applications for Circulating Tumor Cells expressing the Insulin Growth Factor-I Receptor". Clin Can Res 13 (12): 3611–6. doi:10.1158/1078-0432.CCR-07-0268.
  65. Attard G, Swennenhuis JF, Olmos D; et al. (2009). "Characterization of ERG, AR and PTEN status in Circulating Tumor Cells from Patients with Castration-Resistant Prostate Cancer". Cancer Research 69 (7): 2912–8. doi:10.1158/0008-5472.CAN-08-3667. PMID 19339269.
  66. Swennenhuis JF, Tibbe AGJ, Levink R; et al. (2009). "Characterization of Circulating Tumor Cells by Fluorescence In-Situ Hybridization". Cytometry Part A 75A (6): 520–7. doi:10.1002/cyto.a.20718.
  67. Karp DD, Pollak MN, Cohen RB; et al. (2009). "Pharmacokinetics and Pharmacodynamics of the IGF-IR Inhibitor Figitumumab (CP-751,871) in Combination with Paclitaxel and Carboplatin". Journal of Thoracic Oncology 4 (11): 1397–1403. doi:10.1097/JTO.0b013e3181ba2f1d. PMC 2941876. PMID 19745765.
  68. AGJ Tibbe, MC Miller, LWMM Terstappen (2007). "Statistical Considerations for Enumeration of Circulating Tumor Cells". Cytometry Part A 71A: 132–142. doi:10.1002/cyto.a.20369.
  69. F. A. W. Coumans, C. J. M. Doggen, G. Attard; et al. (2010). "All circulating EpCAM1CK1CD452 objects predict overall survival in castration-resistant prostate cancer". Annals of Oncology 21 (9): 1851–7. doi:10.1093/annonc/mdq030. PMID 20147742.
This article is issued from Wikipedia - version of the Wednesday, May 04, 2016. The text is available under the Creative Commons Attribution/Share Alike but additional terms may apply for the media files.