Gauging the Impact of Cancer Treatment Modalities on Circulating Tumor Cells (CTCs)
Abstract
1. Introduction
2. CTCs and Metastatic Efficiency
3. Mechanisms for Tumor Cells to Enter the Vasculature
4. Current Anti-Tumor Therapies may Inadvertently Increase CTCs
4.1. Surgical Procedures
4.2. Radiotherapy
4.3. Chemotherapy
5. Potential Therapies to Reduce CTCs
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Steeg, P.S. Targeting metastasis. Nat. Rev. Cancer 2016, 16, 201–218. [Google Scholar] [CrossRef]
- Momma, T.; Hamblin, M.R.; Wu, H.C.; Hasan, T. Photodynamic therapy of orthotopic prostate cancer with benzoporphyrin derivative: Local control and distant metastasis. Cancer Res. 1998, 58, 5425–5431. [Google Scholar] [PubMed]
- Goldfarb, Y.; Ben-Eliyahu, S. Surgery as a risk factor for breast cancer recurrence and metastasis: Mediating mechanisms and clinical prophylactic approaches. Breast Dis. 2006, 26, 99–114. [Google Scholar] [CrossRef] [PubMed]
- Shi, R.Y.; Yao, Q.Y.; Wu, L.M.; Xu, J.R. Breast Lesions: Diagnosis Using Diffusion Weighted Imaging at 1.5T and 3.0T-Systematic Review and Meta-Analysis. Clin. Breast Cancer 2018, 18, e305–e320. [Google Scholar] [CrossRef] [PubMed]
- Ramirez-Galvan, Y.A.; Cardona-Huerta, S.; Ibarra-Fombona, E.; Elizondo-Riojas, G. Apparent diffusion coefficient (ADC) value to evaluate BI-RADS 4 breast lesions: Correlation with pathological findings. Clin. Imaging 2015, 39, 51–55. [Google Scholar] [CrossRef]
- Folkman, J. Angiogenesis. Annu. Rev. Med. 2006, 57, 1–18. [Google Scholar] [CrossRef]
- Butler, T.P.; Gullino, P.M. Quantitation of cell shedding into efferent blood of mammary adenocarcinoma. Cancer Res. 1975, 35, 512–516. [Google Scholar]
- Chambers, A.F.; Groom, A.C.; MacDonald, I.C. Dissemination and growth of cancer cells in metastatic sites. Nat. Rev. Cancer 2002, 2, 563–572. [Google Scholar] [CrossRef]
- Fidler, I.J. Metastasis: Quantitative analysis of distribution and fate of tumor emboli labeled with 125 I-5-iodo-2’-deoxyuridine. J. Natl. Cancer Inst. 1970, 45, 773–782. [Google Scholar]
- Luzzi, K.J.; MacDonald, I.C.; Schmidt, E.E.; Kerkvliet, N.; Morris, V.L.; Chambers, A.F.; Groom, A.C. Multistep nature of metastatic inefficiency: Dormancy of solitary cells after successful extravasation and limited survival of early micrometastases. Am. J. Pathol. 1998, 153, 865–873. [Google Scholar] [CrossRef]
- Coumans, F.A.; Siesling, S.; Terstappen, L.W. Detection of cancer before distant metastasis. BMC Cancer 2013, 13, 283. [Google Scholar] [CrossRef] [PubMed]
- Van’t Veer, L.J.; Dai, H.; van de Vijver, M.J.; He, Y.D.; Hart, A.A.; Mao, M.; Peterse, H.L.; van der Kooy, K.; Marton, M.J.; Witteveen, A.T.; et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 2002, 415, 530–536. [Google Scholar] [CrossRef] [PubMed]
- Van de Vijver, M.J.; He, Y.D.; van’t Veer, L.J.; Dai, H.; Hart, A.A.; Voskuil, D.W.; Schreiber, G.J.; Peterse, J.L.; Roberts, C.; Marton, M.J.; et al. A gene-expression signature as a predictor of survival in breast cancer. N. Engl. J. Med. 2002, 347, 1999–2009. [Google Scholar] [CrossRef]
- Weigelt, B.; Glas, A.M.; Wessels, L.F.; Witteveen, A.T.; Peterse, J.L.; van’t Veer, L.J. Gene expression profiles of primary breast tumors maintained in distant metastases. Proc. Natl. Acad. Sci. USA 2003, 100, 15901–15905. [Google Scholar] [CrossRef] [PubMed]
- Bernards, R.; Weinberg, R.A. A progression puzzle. Nature 2002, 418, 823. [Google Scholar] [CrossRef] [PubMed]
- Husemann, Y.; Geigl, J.B.; Schubert, F.; Musiani, P.; Meyer, M.; Burghart, E.; Forni, G.; Eils, R.; Fehm, T.; Riethmuller, G.; et al. Systemic spread is an early step in breast cancer. Cancer Cell 2008, 13, 58–68. [Google Scholar] [CrossRef]
- Larson, C.J.; Moreno, J.G.; Pienta, K.J.; Gross, S.; Repollet, M.; O’Hara, S.M.; Russell, T.; Terstappen, L.W. Apoptosis of circulating tumor cells in prostate cancer patients. Cytom. Part A 2004, 62, 46–53. [Google Scholar] [CrossRef]
- Mehes, G.; Witt, A.; Kubista, E.; Ambros, P.F. Circulating breast cancer cells are frequently apoptotic. Am. J. Pathol. 2001, 159, 17–20. [Google Scholar] [CrossRef]
- Fukumura, D.; Jain, R.K. Tumor microvasculature and microenvironment: Targets for anti-angiogenesis and normalization. Microvasc. Res. 2007, 74, 72–84. [Google Scholar] [CrossRef]
- Paoletti, C.; Li, Y.; Muniz, M.C.; Kidwell, K.M.; Aung, K.; Thomas, D.G.; Brown, M.E.; Abramson, V.G.; Irvin, W.J., Jr.; Lin, N.U.; et al. Significance of Circulating Tumor Cells in Metastatic Triple-Negative Breast Cancer Patients within a Randomized, Phase II Trial: TBCRC 019. Clin. Cancer Res. 2015, 21, 2771–2779. [Google Scholar] [CrossRef]
- Jansson, S.; Bendahl, P.O.; Larsson, A.M.; Aaltonen, K.E.; Ryden, L. Prognostic impact of circulating tumor cell apoptosis and clusters in serial blood samples from patients with metastatic breast cancer in a prospective observational cohort. BMC Cancer 2016, 16, 433. [Google Scholar] [CrossRef] [PubMed]
- Hou, J.M.; Krebs, M.; Ward, T.; Morris, K.; Sloane, R.; Blackhall, F.; Dive, C. Circulating tumor cells, enumeration and beyond. Cancers 2010, 2, 1236–1250. [Google Scholar] [CrossRef] [PubMed]
- Wong, A.D.; Searson, P.C. Mitosis-Mediated Intravasation in a Tissue-Engineered Tumor-Microvessel Platform. Cancer Res. 2017, 77, 6453–6461. [Google Scholar] [CrossRef] [PubMed]
- Nicolson, G.L. Metastatic tumor cell attachment and invasion assay utilizing vascular endothelial cell monolayers. J. Histochem. Cytochem. 1982, 30, 214–220. [Google Scholar] [CrossRef]
- Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Investig. 2009, 119, 1420–1428. [Google Scholar] [CrossRef]
- Nieto, M.A.; Huang, R.Y.; Jackson, R.A.; Thiery, J.P. Emt: 2016. Cell 2016, 166, 21–45. [Google Scholar] [CrossRef]
- Thiery, J.P. Epithelial-mesenchymal transitions in tumour progression. Nat. Rev. Cancer 2002, 2, 442–454. [Google Scholar] [CrossRef]
- Lambert, A.W.; Pattabiraman, D.R.; Weinberg, R.A. Emerging Biological Principles of Metastasis. Cell 2017, 168, 670–691. [Google Scholar] [CrossRef]
- Rhim, A.D.; Mirek, E.T.; Aiello, N.M.; Maitra, A.; Bailey, J.M.; McAllister, F.; Reichert, M.; Beatty, G.L.; Rustgi, A.K.; Vonderheide, R.H.; et al. EMT and dissemination precede pancreatic tumor formation. Cell 2012, 148, 349–361. [Google Scholar] [CrossRef]
- Mani, S.A.; Guo, W.; Liao, M.J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef]
- Morel, A.P.; Lievre, M.; Thomas, C.; Hinkal, G.; Ansieau, S.; Puisieux, A. Generation of breast cancer stem cells through epithelial-mesenchymal transition. PLoS ONE 2008, 3, e2888. [Google Scholar] [CrossRef] [PubMed]
- Fan, F.; Samuel, S.; Evans, K.W.; Lu, J.; Xia, L.; Zhou, Y.; Sceusi, E.; Tozzi, F.; Ye, X.C.; Mani, S.A.; et al. Overexpression of snail induces epithelial-mesenchymal transition and a cancer stem cell-like phenotype in human colorectal cancer cells. Cancer Med. 2012, 1, 5–16. [Google Scholar] [CrossRef]
- Pang, R.; Law, W.L.; Chu, A.C.; Poon, J.T.; Lam, C.S.; Chow, A.K.; Ng, L.; Cheung, L.W.; Lan, X.R.; Lan, H.Y.; et al. A subpopulation of CD26+ cancer stem cells with metastatic capacity in human colorectal cancer. Cell Stem Cell 2010, 6, 603–615. [Google Scholar] [CrossRef] [PubMed]
- Long, H.; Xiang, T.; Qi, W.; Huang, J.; Chen, J.; He, L.; Liang, Z.; Guo, B.; Li, Y.; Xie, R.; et al. CD133+ ovarian cancer stem-like cells promote non-stem cancer cell metastasis via CCL5 induced epithelial-mesenchymal transition. Oncotarget 2015, 6, 5846–5859. [Google Scholar] [CrossRef] [PubMed]
- Rasheed, Z.A.; Yang, J.; Wang, Q.; Kowalski, J.; Freed, I.; Murter, C.; Hong, S.M.; Koorstra, J.B.; Rajeshkumar, N.V.; He, X.; et al. Prognostic significance of tumorigenic cells with mesenchymal features in pancreatic adenocarcinoma. J. Natl. Cancer Inst. 2010, 102, 340–351. [Google Scholar] [CrossRef] [PubMed]
- Kong, D.; Banerjee, S.; Ahmad, A.; Li, Y.; Wang, Z.; Sethi, S.; Sarkar, F.H. Epithelial to mesenchymal transition is mechanistically linked with stem cell signatures in prostate cancer cells. PLoS ONE 2010, 5, e12445. [Google Scholar] [CrossRef]
- Zhou, D.; Kannappan, V.; Chen, X.; Li, J.; Leng, X.; Zhang, J.; Xuan, S. RBP2 induces stem-like cancer cells by promoting EMT and is a prognostic marker for renal cell carcinoma. Exp. Mol. Med. 2016, 48, e238. [Google Scholar] [CrossRef]
- Bednarz-Knoll, N.; Alix-Panabieres, C.; Pantel, K. Plasticity of disseminating cancer cells in patients with epithelial malignancies. Cancer Metastasis Rev. 2012, 31, 673–687. [Google Scholar] [CrossRef]
- Grosse-Wilde, A.; Fouquier d’Herouel, A.; McIntosh, E.; Ertaylan, G.; Skupin, A.; Kuestner, R.E.; del Sol, A.; Walters, K.A.; Huang, S. Stemness of the Hybrid Epithelial/Mesenchymal State in Breast Cancer and Its Association with Poor Survival. PLoS ONE 2015, 10, e0126522. [Google Scholar] [CrossRef]
- Li, W.; Kang, Y. Probing the Fifty Shades of EMT in Metastasis. Trends Cancer 2016, 2, 65–67. [Google Scholar] [CrossRef]
- Bonnomet, A.; Syne, L.; Brysse, A.; Feyereisen, E.; Thompson, E.W.; Noel, A.; Foidart, J.M.; Birembaut, P.; Polette, M.; Gilles, C. A dynamic in vivo model of epithelial-to-mesenchymal transitions in circulating tumor cells and metastases of breast cancer. Oncogene 2012, 31, 3741–3753. [Google Scholar] [CrossRef] [PubMed]
- Trimboli, A.J.; Fukino, K.; de Bruin, A.; Wei, G.; Shen, L.; Tanner, S.M.; Creasap, N.; Rosol, T.J.; Robinson, M.L.; Eng, C.; et al. Direct evidence for epithelial-mesenchymal transitions in breast cancer. Cancer Res. 2008, 68, 937–945. [Google Scholar] [CrossRef] [PubMed]
- Celia-Terrassa, T.; Meca-Cortes, O.; Mateo, F.; Martinez de Paz, A.; Rubio, N.; Arnal-Estape, A.; Ell, B.J.; Bermudo, R.; Diaz, A.; Guerra-Rebollo, M.; et al. Epithelial-mesenchymal transition can suppress major attributes of human epithelial tumor-initiating cells. J. Clin. Investig. 2012, 122, 1849–1868. [Google Scholar] [CrossRef] [PubMed]
- Lundgren, K.; Nordenskjold, B.; Landberg, G. Hypoxia, Snail and incomplete epithelial-mesenchymal transition in breast cancer. Br. J. Cancer 2009, 101, 1769–1781. [Google Scholar] [CrossRef]
- Hong, T.; Watanabe, K.; Ta, C.H.; Villarreal-Ponce, A.; Nie, Q.; Dai, X. An Ovol2-Zeb1 Mutual Inhibitory Circuit Governs Bidirectional and Multi-Step Transition between Epithelial and Mesenchymal States. PLoS Comput. Biol. 2015, 11, e1004569. [Google Scholar] [CrossRef]
- Sampson, V.B.; David, J.M.; Puig, I.; Patil, P.U.; de Herreros, A.G.; Thomas, G.V.; Rajasekaran, A.K. Wilms’ tumor protein induces an epithelial-mesenchymal hybrid differentiation state in clear cell renal cell carcinoma. PLoS ONE 2014, 9, e102041. [Google Scholar] [CrossRef]
- Schliekelman, M.J.; Taguchi, A.; Zhu, J.; Dai, X.; Rodriguez, J.; Celiktas, M.; Zhang, Q.; Chin, A.; Wong, C.H.; Wang, H.; et al. Molecular portraits of epithelial, mesenchymal, and hybrid States in lung adenocarcinoma and their relevance to survival. Cancer Res. 2015, 75, 1789–1800. [Google Scholar] [CrossRef]
- Bailey, P.C.; Martin, S.S. Insights on CTC Biology and Clinical Impact Emerging from Advances in Capture Technology. Cells 2019, 8, 553. [Google Scholar] [CrossRef]
- Ferreira, M.M.; Ramani, V.C.; Jeffrey, S.S. Circulating tumor cell technologies. Mol. Oncol. 2016, 10, 374–394. [Google Scholar] [CrossRef]
- Boya, M.; Chu, C.H.; Liu, R.; Ozkaya-Ahmadov, T.; Sarioglu, A.F. Circulating Tumor Cell Enrichment Technologies. Recent Results Cancer Res. 2020, 215, 25–55. [Google Scholar] [CrossRef]
- Banko, P.; Lee, S.Y.; Nagygyorgy, V.; Zrinyi, M.; Chae, C.H.; Cho, D.H.; Telekes, A. Technologies for circulating tumor cell separation from whole blood. J. Hematol. Oncol. 2019, 12, 48. [Google Scholar] [CrossRef]
- Shen, Z.; Wu, A.; Chen, X. Current detection technologies for circulating tumor cells. Chem. Soc. Rev. 2017, 46, 2038–2056. [Google Scholar] [CrossRef] [PubMed]
- Rawal, S.; Yang, Y.P.; Cote, R.; Agarwal, A. Identification and Quantitation of Circulating Tumor Cells. Annu. Rev. Anal. Chem. (Palo Alto Calif.) 2017, 10, 321–343. [Google Scholar] [CrossRef] [PubMed]
- Khetani, S.; Mohammadi, M.; Nezhad, A.S. Filter-based isolation, enrichment, and characterization of circulating tumor cells. Biotechnol. Bioeng. 2018, 115, 2504–2529. [Google Scholar] [CrossRef] [PubMed]
- Cheung, K.J.; Gabrielson, E.; Werb, Z.; Ewald, A.J. Collective invasion in breast cancer requires a conserved basal epithelial program. Cell 2013, 155, 1639–1651. [Google Scholar] [CrossRef] [PubMed]
- Cheung, K.J.; Padmanaban, V.; Silvestri, V.; Schipper, K.; Cohen, J.D.; Fairchild, A.N.; Gorin, M.A.; Verdone, J.E.; Pienta, K.J.; Bader, J.S.; et al. Polyclonal breast cancer metastases arise from collective dissemination of keratin 14-expressing tumor cell clusters. Proc. Natl. Acad. Sci. USA 2016, 113, E854–E863. [Google Scholar] [CrossRef] [PubMed]
- Wyckoff, J.B.; Wang, Y.; Lin, E.Y.; Li, J.F.; Goswami, S.; Stanley, E.R.; Segall, J.E.; Pollard, J.W.; Condeelis, J. Direct visualization of macrophage-assisted tumor cell intravasation in mammary tumors. Cancer Res. 2007, 67, 2649–2656. [Google Scholar] [CrossRef]
- Yamaguchi, H.; Wyckoff, J.; Condeelis, J. Cell migration in tumors. Curr. Opin. Cell Biol. 2005, 17, 559–564. [Google Scholar] [CrossRef]
- Condeelis, J.; Pollard, J.W. Macrophages: Obligate partners for tumor cell migration, invasion, and metastasis. Cell 2006, 124, 263–266. [Google Scholar] [CrossRef]
- Stegner, D.; Dutting, S.; Nieswandt, B. Mechanistic explanation for platelet contribution to cancer metastasis. Thromb. Res. 2014, 133 (Suppl. 2), S149–S157. [Google Scholar] [CrossRef]
- Fidler, I.J. The relationship of embolic homogeneity, number, size and viability to the incidence of experimental metastasis. Eur. J. Cancer 1973, 9, 223–227. [Google Scholar] [CrossRef]
- Liotta, L.A.; Saidel, M.G.; Kleinerman, J. The significance of hematogenous tumor cell clumps in the metastatic process. Cancer Res. 1976, 36, 889–894. [Google Scholar] [PubMed]
- Molnar, B.; Ladanyi, A.; Tanko, L.; Sreter, L.; Tulassay, Z. Circulating tumor cell clusters in the peripheral blood of colorectal cancer patients. Clin. Cancer Res. 2001, 7, 4080–4085. [Google Scholar] [PubMed]
- Cho, E.H.; Wendel, M.; Luttgen, M.; Yoshioka, C.; Marrinucci, D.; Lazar, D.; Schram, E.; Nieva, J.; Bazhenova, L.; Morgan, A.; et al. Characterization of circulating tumor cell aggregates identified in patients with epithelial tumors. Phys. Biol. 2012, 9, 016001. [Google Scholar] [CrossRef] [PubMed]
- Stott, S.L.; Hsu, C.H.; Tsukrov, D.I.; Yu, M.; Miyamoto, D.T.; Waltman, B.A.; Rothenberg, S.M.; Shah, A.M.; Smas, M.E.; Korir, G.K.; et al. Isolation of circulating tumor cells using a microvortex-generating herringbone-chip. Proc. Natl. Acad. Sci. USA 2010, 107, 18392–18397. [Google Scholar] [CrossRef] [PubMed]
- Yu, M.; Bardia, A.; Wittner, B.S.; Stott, S.L.; Smas, M.E.; Ting, D.T.; Isakoff, S.J.; Ciciliano, J.C.; Wells, M.N.; Shah, A.M.; et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 2013, 339, 580–584. [Google Scholar] [CrossRef]
- Aceto, N.; Bardia, A.; Miyamoto, D.T.; Donaldson, M.C.; Wittner, B.S.; Spencer, J.A.; Yu, M.; Pely, A.; Engstrom, A.; Zhu, H.; et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell 2014, 158, 1110–1122. [Google Scholar] [CrossRef]
- Updyke, T.V.; Nicolson, G.L. Malignant melanoma cell lines selected in vitro for increased homotypic adhesion properties have increased experimental metastatic potential. Clin. Exp. Metastasis 1986, 4, 273–284. [Google Scholar] [CrossRef]
- Sleeman, J.P.; Nazarenko, I.; Thiele, W. Do all roads lead to Rome? Routes to metastasis development. Int. J. Cancer 2011, 128, 2511–2526. [Google Scholar] [CrossRef]
- Roh, J.; Muelleman, T.; Tawfik, O.; Thomas, S.M. Perineural growth in head and neck squamous cell carcinoma: A review. Oral Oncol. 2015, 51, 16–23. [Google Scholar] [CrossRef]
- Marchesi, F.; Piemonti, L.; Mantovani, A.; Allavena, P. Molecular mechanisms of perineural invasion, a forgotten pathway of dissemination and metastasis. Cytokine Growth Factor Rev. 2010, 21, 77–82. [Google Scholar] [CrossRef] [PubMed]
- Lugassy, C.; Kleinman, H.K.; Engbring, J.A.; Welch, D.R.; Harms, J.F.; Rufner, R.; Ghanem, G.; Patierno, S.R.; Barnhill, R.L. Pericyte-like location of GFP-tagged melanoma cells: Ex vivo and in vivo studies of extravascular migratory metastasis. Am. J. Pathol. 2004, 164, 1191–1198. [Google Scholar] [CrossRef]
- Martin, O.A.; Anderson, R.L.; Narayan, K.; MacManus, M.P. Does the mobilization of circulating tumour cells during cancer therapy cause metastasis? Nat. Rev. Clin. Oncol. 2017, 14, 32–44. [Google Scholar] [CrossRef] [PubMed]
- Cohen, S.J.; Punt, C.J.; Iannotti, N.; Saidman, B.H.; Sabbath, K.D.; Gabrail, N.Y.; Picus, J.; Morse, M.; Mitchell, E.; Miller, M.C.; 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. [Google Scholar] [CrossRef] [PubMed]
- Cristofanilli, M.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Matera, J.; Miller, M.C.; Reuben, J.M.; Doyle, G.V.; Allard, W.J.; Terstappen, L.W.; et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N. Engl. J. Med. 2004, 351, 781–791. [Google Scholar] [CrossRef]
- de Bono, J.S.; Scher, H.I.; Montgomery, R.B.; Parker, C.; Miller, M.C.; Tissing, H.; Doyle, G.V.; Terstappen, L.W.; Pienta, K.J.; Raghavan, D. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin. Cancer Res. 2008, 14, 6302–6309. [Google Scholar] [CrossRef]
- Mocellin, S.; Del Fiore, P.; Guarnieri, L.; Scalerta, R.; Foletto, M.; Chiarion, V.; Pilati, P.; Nitti, D.; Lise, M.; Rossi, C.R. Molecular detection of circulating tumor cells is an independent prognostic factor in patients with high-risk cutaneous melanoma. Int. J. Cancer 2004, 111, 741–745. [Google Scholar] [CrossRef]
- Moreno, J.G.; O’Hara, S.M.; Gross, S.; Doyle, G.; Fritsche, H.; Gomella, L.G.; Terstappen, L.W. Changes in circulating carcinoma cells in patients with metastatic prostate cancer correlate with disease status. Urology 2001, 58, 386–392. [Google Scholar] [CrossRef]
- van Dalum, G.; van der Stam, G.J.; Tibbe, A.G.; Franken, B.; Mastboom, W.J.; Vermes, I.; de Groot, M.R.; Terstappen, L.W. Circulating tumor cells before and during follow-up after breast cancer surgery. Int. J. Oncol. 2015, 46, 407–413. [Google Scholar] [CrossRef]
- Rahbari, N.N.; Aigner, M.; Thorlund, K.; Mollberg, N.; Motschall, E.; Jensen, K.; Diener, M.K.; Buchler, M.W.; Koch, M.; Weitz, J. Meta-analysis shows that detection of circulating tumor cells indicates poor prognosis in patients with colorectal cancer. Gastroenterology 2010, 138, 1714–1726. [Google Scholar] [CrossRef]
- Steinert, G.; Scholch, S.; Koch, M.; Weitz, J. Biology and significance of circulating and disseminated tumour cells in colorectal cancer. Langenbeck’s Arch. Surg. 2012, 397, 535–542. [Google Scholar] [CrossRef]
- Peach, G.; Kim, C.; Zacharakis, E.; Purkayastha, S.; Ziprin, P. Prognostic significance of circulating tumour cells following surgical resection of colorectal cancers: A systematic review. Br. J. Cancer 2010, 102, 1327–1334. [Google Scholar] [CrossRef] [PubMed]
- Kapeleris, J.; Kulasinghe, A.; Warkiani, M.E.; Vela, I.; Kenny, L.; O’Byrne, K.; Punyadeera, C. The Prognostic Role of Circulating Tumor Cells (CTCs) in Lung Cancer. Front. Oncol. 2018, 8, 311. [Google Scholar] [CrossRef] [PubMed]
- Kulasinghe, A.; Kapeleris, J.; Kimberley, R.; Mattarollo, S.R.; Thompson, E.W.; Thiery, J.P.; Kenny, L.; O’Byrne, K.; Punyadeera, C. The prognostic significance of circulating tumor cells in head and neck and non-small-cell lung cancer. Cancer Med. 2018, 7, 5910–5919. [Google Scholar] [CrossRef] [PubMed]
- National Breast Cancer Foundation, Inc. Breast Anatomy. Available online: www.nationalbreastcancer.org (accessed on 1 September 2019).
- Kaplan, R.N.; Riba, R.D.; Zacharoulis, S.; Bramley, A.H.; Vincent, L.; Costa, C.; MacDonald, D.D.; Jin, D.K.; Shido, K.; Kerns, S.A.; et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 2005, 438, 820–827. [Google Scholar] [CrossRef] [PubMed]
- Hansen, E.; Wolff, N.; Knuechel, R.; Ruschoff, J.; Hofstaedter, F.; Taeger, K. Tumor cells in blood shed from the surgical field. Arch. Surg. 1995, 130, 387–393. [Google Scholar] [CrossRef]
- Hashimoto, M.; Tanaka, F.; Yoneda, K.; Takuwa, T.; Matsumoto, S.; Okumura, Y.; Kondo, N.; Tsubota, N.; Tsujimura, T.; Tabata, C.; et al. Significant increase in circulating tumour cells in pulmonary venous blood during surgical manipulation in patients with primary lung cancer. Interact. Cardiovasc. Thorac. Surg. 2014, 18, 775–783. [Google Scholar] [CrossRef]
- Papavasiliou, P.; Fisher, T.; Kuhn, J.; Nemunaitis, J.; Lamont, J. Circulating tumor cells in patients undergoing surgery for hepatic metastases from colorectal cancer. Bayl. Univ. Med. Cent. Proc. 2010, 23, 11–14. [Google Scholar] [CrossRef]
- Matsutani, N.; Sawabata, N.; Yamaguchi, M.; Woo, T.; Kudo, Y.; Kawase, A.; Shiono, S.; Iinuma, H.; Morita, S.; Kawamura, M. Does lung cancer surgery cause circulating tumor cells?—A multicenter, prospective study. J. Thorac. Dis. 2017, 9, 2419–2426. [Google Scholar] [CrossRef]
- Yu, J.J.; Xiao, W.; Dong, S.L.; Liang, H.F.; Zhang, Z.W.; Zhang, B.X.; Huang, Z.Y.; Chen, Y.F.; Zhang, W.G.; Luo, H.P.; et al. Effect of surgical liver resection on circulating tumor cells in patients with hepatocellular carcinoma. BMC Cancer 2018, 18, 835. [Google Scholar] [CrossRef]
- Haga, N.; Onagi, A.; Koguchi, T.; Hoshi, S.; Ogawa, S.; Akaihata, H.; Hata, J.; Hiraki, H.; Honda, R.; Tanji, R.; et al. Perioperative Detection of Circulating Tumor Cells in Radical or Partial Nephrectomy for Renal Cell Carcinoma. Ann. Surg. Oncol. 2019. [Google Scholar] [CrossRef] [PubMed]
- Wei, X.Q.; Ma, Y.; Chen, Y.; Liu, X.; Zhao, M.; Zhou, L.W. Laparoscopic surgery for early cervical squamous cell carcinoma and its effect on the micrometastasis of cancer cells. Medicine (Baltimore) 2018, 97, e11921. [Google Scholar] [CrossRef] [PubMed]
- Wind, J.; Tuynman, J.B.; Tibbe, A.G.; Swennenhuis, J.F.; Richel, D.J.; van Berge Henegouwen, M.I.; Bemelman, W.A. Circulating tumour cells during laparoscopic and open surgery for primary colonic cancer in portal and peripheral blood. Eur. J. Surg. Oncol. 2009, 35, 942–950. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Yan, W.; Yang, X.; Chen, L.; Fan, L.; Liu, H.; Liu, K.; Zhang, Y.; Jiang, J. Less micrometastatic risk related to circulating tumor cells after endoscopic breast cancer surgery compared to open surgery. BMC Cancer 2019, 19, 1070. [Google Scholar] [CrossRef]
- Juratli, M.A.; Sarimollaoglu, M.; Siegel, E.R.; Nedosekin, D.A.; Galanzha, E.I.; Suen, J.Y.; Zharov, V.P. Real-time monitoring of circulating tumor cell release during tumor manipulation using in vivo photoacoustic and fluorescent flow cytometry. Head Neck 2014, 36, 1207–1215. [Google Scholar] [CrossRef]
- Juratli, M.A.; Siegel, E.R.; Nedosekin, D.A.; Sarimollaoglu, M.; Jamshidi-Parsian, A.; Cai, C.; Menyaev, Y.A.; Suen, J.Y.; Galanzha, E.I.; Zharov, V.P. In Vivo Long-Term Monitoring of Circulating Tumor Cells Fluctuation during Medical Interventions. PLoS ONE 2015, 10, e0137613. [Google Scholar] [CrossRef]
- Dyavanagoudar, S.; Kale, A.; Bhat, K.; Hallikerimath, S. Reverse transcriptase polymerase chain reaction study to evaluate dissemination of cancer cells into circulation after incision biopsy in oral squamous cell carcinoma. Indian J. Dent. Res. 2008, 19, 315–319. [Google Scholar] [CrossRef]
- Kusukawa, J.; Suefuji, Y.; Ryu, F.; Noguchi, R.; Iwamoto, O.; Kameyama, T. Dissemination of cancer cells into circulation occurs by incisional biopsy of oral squamous cell carcinoma. J. Oral Pathol. Med. 2000, 29, 303–307. [Google Scholar] [CrossRef]
- Robertson, E.G.; Baxter, G. Tumour seeding following percutaneous needle biopsy: The real story! Clin. Radiol. 2011, 66, 1007–1014. [Google Scholar] [CrossRef]
- Mathenge, E.G.; Dean, C.A.; Clements, D.; Vaghar-Kashani, A.; Photopoulos, S.; Coyle, K.M.; Giacomantonio, M.; Malueth, B.; Nunokawa, A.; Jordan, J.; et al. Core needle biopsy of breast cancer tumors increases distant metastases in a mouse model. Neoplasia 2014, 16, 950–960. [Google Scholar] [CrossRef]
- Joosse, S.A.; Beyer, B.; Gasch, C.; Nastaly, P.; Kuske, A.; Isbarn, H.; Horst, L.J.; Hille, C.; Gorges, T.M.; Cayrefourcq, L.; et al. Tumor-Associated Release of Prostatic Cells into the Blood after Transrectal Ultrasound-Guided Biopsy in Patients with Histologically Confirmed Prostate Cancer. Clin. Chem. 2019. [Google Scholar] [CrossRef] [PubMed]
- Von Essen, C.F. Radiation enhancement of metastasis: A review. Clin. Exp. Metastasis 1991, 9, 77–104. [Google Scholar] [CrossRef] [PubMed]
- Kaplan, H.S.; Murphy, E.D. The effect of local roentgen irradiation on the biological behavior of a transplantable mouse carcinoma; increased frequency of pulmonary metastasis. J. Natl. Cancer Inst. 1949, 9, 407–413. [Google Scholar] [PubMed]
- Sheldon, P.W.; Fowler, J.F. The effect of low-dose pre-operative X-irradiation of implanted mouse mammary carcinomas on local recurrence and metastasis. Br. J. Cancer 1976, 34, 401–407. [Google Scholar] [CrossRef] [PubMed]
- Camphausen, K.; Moses, M.A.; Beecken, W.D.; Khan, M.K.; Folkman, J.; O’Reilly, M.S. Radiation therapy to a primary tumor accelerates metastatic growth in mice. Cancer Res. 2001, 61, 2207–2211. [Google Scholar] [PubMed]
- Koonce, N.A.; Juratli, M.A.; Cai, C.; Sarimollaoglu, M.; Menyaev, Y.A.; Dent, J.; Quick, C.M.; Dings, R.P.M.; Nedosekin, D.; Zharov, V.; et al. Real-time monitoring of circulating tumor cell (CTC) release after nanodrug or tumor radiotherapy using in vivo flow cytometry. Biochem. Biophys. Res. Commun. 2017, 492, 507–512. [Google Scholar] [CrossRef]
- Gomez-Casal, R.; Bhattacharya, C.; Ganesh, N.; Bailey, L.; Basse, P.; Gibson, M.; Epperly, M.; Levina, V. Non-small cell lung cancer cells survived ionizing radiation treatment display cancer stem cell and epithelial-mesenchymal transition phenotypes. Mol. Cancer 2013, 12, 94. [Google Scholar] [CrossRef]
- Rofstad, E.K.; Mathiesen, B.; Galappathi, K. Increased metastatic dissemination in human melanoma xenografts after subcurative radiation treatment: Radiation-induced increase in fraction of hypoxic cells and hypoxia-induced up-regulation of urokinase-type plasminogen activator receptor. Cancer Res. 2004, 64, 13–18. [Google Scholar] [CrossRef]
- Bernier, J.; Hall, E.J.; Giaccia, A. Radiation oncology: A century of achievements. Nat. Rev. Cancer 2004, 4, 737–747. [Google Scholar] [CrossRef]
- Eriksson, D.; Stigbrand, T. Radiation-induced cell death mechanisms. Tumour Biol. 2010, 31, 363–372. [Google Scholar] [CrossRef]
- Dewey, W.C.; Ling, C.C.; Meyn, R.E. Radiation-induced apoptosis: Relevance to radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 1995, 33, 781–796. [Google Scholar] [CrossRef]
- Palumbo, S.; Comincini, S. Autophagy and ionizing radiation in tumors: The “survive or not survive” dilemma. J. Cell. Physiol. 2013, 228, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Brunner, T.B.; Kunz-Schughart, L.A.; Grosse-Gehling, P.; Baumann, M. Cancer stem cells as a predictive factor in radiotherapy. Semin. Radiat. Oncol. 2012, 22, 151–174. [Google Scholar] [CrossRef] [PubMed]
- Butof, R.; Dubrovska, A.; Baumann, M. Clinical perspectives of cancer stem cell research in radiation oncology. Radiother. Oncol. 2013, 108, 388–396. [Google Scholar] [CrossRef]
- Vilalta, M.; Rafat, M.; Giaccia, A.J.; Graves, E.E. Recruitment of circulating breast cancer cells is stimulated by radiotherapy. Cell Rep. 2014, 8, 402–409. [Google Scholar] [CrossRef]
- Early Breast Cancer Trialists’ Collaborative Group; Darby, S.; McGale, P.; Correa, C.; Taylor, C.; Arriagada, R.; Clarke, M.; Cutter, D.; Davies, C.; Ewertz, M.; et al. Effect of radiotherapy after breast-conserving surgery on 10-year recurrence and 15-year breast cancer death: Meta-analysis of individual patient data for 10,801 women in 17 randomised trials. Lancet 2011, 378, 1707–1716. [Google Scholar] [CrossRef]
- Martin, O.A.; Anderson, R.L.; Russell, P.A.; Cox, R.A.; Ivashkevich, A.; Swierczak, A.; Doherty, J.P.; Jacobs, D.H.; Smith, J.; Siva, S.; et al. Mobilization of viable tumor cells into the circulation during radiation therapy. Int. J. Radiat. Oncol. Biol. Phys. 2014, 88, 395–403. [Google Scholar] [CrossRef]
- Dorsey, J.F.; Kao, G.D.; MacArthur, K.M.; Ju, M.; Steinmetz, D.; Wileyto, E.P.; Simone, C.B., 2nd; Hahn, S.M. Tracking viable circulating tumor cells (CTCs) in the peripheral blood of non-small cell lung cancer (NSCLC) patients undergoing definitive radiation therapy: Pilot study results. Cancer 2015, 121, 139–149. [Google Scholar] [CrossRef]
- Goodman, C.R.; Seagle, B.L.; Friedl, T.W.P.; Rack, B.; Lato, K.; Fink, V.; Cristofanilli, M.; Donnelly, E.D.; Janni, W.; Shahabi, S.; et al. Association of Circulating Tumor Cell Status with Benefit of Radiotherapy and Survival in Early-Stage Breast Cancer. JAMA Oncol. 2018, 4, e180163. [Google Scholar] [CrossRef]
- Dong, Q.; Huang, J.; Zhou, Y.; Li, L.; Bao, G.; Feng, J.; Sha, H. Hematogenous dissemination of lung cancer cells during surgery: Quantitative detection by flow cytometry and prognostic significance. Lung Cancer 2002, 37, 293–301. [Google Scholar] [CrossRef]
- Paez-Ribes, M.; Allen, E.; Hudock, J.; Takeda, T.; Okuyama, H.; Vinals, F.; Inoue, M.; Bergers, G.; Hanahan, D.; Casanovas, O. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 2009, 15, 220–231. [Google Scholar] [CrossRef] [PubMed]
- Ebos, J.M.; Lee, C.R.; Cruz-Munoz, W.; Bjarnason, G.A.; Christensen, J.G.; Kerbel, R.S. Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell 2009, 15, 232–239. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Gong, J.; Zhang, Q.; Lu, Z.; Gao, J.; Li, Y.; Cao, Y.; Shen, L. Dynamic monitoring of circulating tumour cells to evaluate therapeutic efficacy in advanced gastric cancer. Br. J. Cancer 2016, 114, 138–145. [Google Scholar] [CrossRef] [PubMed]
- Yan, W.T.; Cui, X.; Chen, Q.; Li, Y.F.; Cui, Y.H.; Wang, Y.; Jiang, J. Circulating tumor cell status monitors the treatment responses in breast cancer patients: A meta-analysis. Sci. Rep. 2017, 7, 43464. [Google Scholar] [CrossRef]
- Smerage, J.B.; Barlow, W.E.; Hortobagyi, G.N.; Winer, E.P.; Leyland-Jones, B.; Srkalovic, G.; Tejwani, S.; Schott, A.F.; O’Rourke, M.A.; Lew, D.L.; et al. Circulating tumor cells and response to chemotherapy in metastatic breast cancer: SWOG S0500. J. Clin. Oncol. 2014, 32, 3483–3489. [Google Scholar] [CrossRef]
- Ebos, J.M.; Kerbel, R.S. Antiangiogenic therapy: Impact on invasion, disease progression, and metastasis. Nat. Rev. Clin. Oncol. 2011, 8, 210–221. [Google Scholar] [CrossRef]
- Obenauf, A.C.; Zou, Y.; Ji, A.L.; Vanharanta, S.; Shu, W.; Shi, H.; Kong, X.; Bosenberg, M.C.; Wiesner, T.; Rosen, N.; et al. Therapy-induced tumour secretomes promote resistance and tumour progression. Nature 2015, 520, 368–372. [Google Scholar] [CrossRef]
- Lin, T.H.; Lee, S.O.; Niu, Y.; Xu, D.; Liang, L.; Li, L.; Yeh, S.D.; Fujimoto, N.; Yeh, S.; Chang, C. Differential androgen deprivation therapies with anti-androgens casodex/bicalutamide or MDV3100/Enzalutamide versus anti-androgen receptor ASC-J9(R) Lead to promotion versus suppression of prostate cancer metastasis. J. Biol. Chem. 2013, 288, 19359–19369. [Google Scholar] [CrossRef]
- Volk-Draper, L.; Hall, K.; Griggs, C.; Rajput, S.; Kohio, P.; DeNardo, D.; Ran, S. Paclitaxel therapy promotes breast cancer metastasis in a TLR4-dependent manner. Cancer Res. 2014, 74, 5421–5434. [Google Scholar] [CrossRef]
- Pool, S.E.; Bison, S.; Koelewijn, S.J.; van der Graaf, L.M.; Melis, M.; Krenning, E.P.; de Jong, M. mTOR inhibitor RAD001 promotes metastasis in a rat model of pancreatic neuroendocrine cancer. Cancer Res. 2013, 73, 12–18. [Google Scholar] [CrossRef]
- Itescu, S.; Burke, E.; Lietz, K.; John, R.; Mancini, D.; Michler, R.; Rose, E.; Oz, M.; Edwards, N. Intravenous pulse administration of cyclophosphamide is an effective and safe treatment for sensitized cardiac allograft recipients. Circulation 2002, 105, 1214–1219. [Google Scholar] [CrossRef][Green Version]
- Schiavoni, G.; Mattei, F.; Di Pucchio, T.; Santini, S.M.; Bracci, L.; Belardelli, F.; Proietti, E. Cyclophosphamide induces type I interferon and augments the number of CD44(hi) T lymphocytes in mice: Implications for strategies of chemoimmunotherapy of cancer. Blood 2000, 95, 2024–2030. [Google Scholar] [CrossRef]
- Man, S.; Zhang, Y.; Gao, W.; Yan, L.; Ma, C. Cyclophosphamide promotes pulmonary metastasis on mouse lung adenocarcinoma. Clin. Exp. Metastasis 2008, 25, 855–864. [Google Scholar] [CrossRef] [PubMed]
- Yamauchi, K.; Yang, M.; Hayashi, K.; Jiang, P.; Yamamoto, N.; Tsuchiya, H.; Tomita, K.; Moossa, A.R.; Bouvet, M.; Hoffman, R.M. Induction of cancer metastasis by cyclophosphamide pretreatment of host mice: An opposite effect of chemotherapy. Cancer Res. 2008, 68, 516–520. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y.J.; Muldoon, L.L.; Dickey, D.T.; Lewin, S.J.; Varallyay, C.G.; Neuwelt, E.A. Cyclophosphamide enhances human tumor growth in nude rat xenografted tumor models. Neoplasia 2009, 11, 187–195. [Google Scholar] [CrossRef] [PubMed]
- Daenen, L.G.; Roodhart, J.M.; van Amersfoort, M.; Dehnad, M.; Roessingh, W.; Ulfman, L.H.; Derksen, P.W.; Voest, E.E. Chemotherapy enhances metastasis formation via VEGFR-1-expressing endothelial cells. Cancer Res. 2011, 71, 6976–6985. [Google Scholar] [CrossRef] [PubMed]
- Kaigorodova, E.V.; Savelieva, O.E.; Tashireva, L.A.; Tarabanovskaya, N.A.; Simolina, E.I.; Denisov, E.V.; Slonimskaya, E.M.; Choynzonov, E.L.; Perelmuter, V.M. Heterogeneity of Circulating Tumor Cells in Neoadjuvant Chemotherapy of Breast Cancer. Molecules 2018, 23, 727. [Google Scholar] [CrossRef]
- Papadaki, M.A.; Stoupis, G.; Theodoropoulos, P.A.; Mavroudis, D.; Georgoulias, V.; Agelaki, S. Circulating Tumor Cells with Stemness and Epithelial-to-Mesenchymal Transition Features Are Chemoresistant and Predictive of Poor Outcome in Metastatic Breast Cancer. Mol. Cancer Ther. 2019, 18, 437–447. [Google Scholar] [CrossRef]
- Balzer, E.M.; Whipple, R.A.; Cho, E.H.; Matrone, M.A.; Martin, S.S. Antimitotic chemotherapeutics promote adhesive responses in detached and circulating tumor cells. Breast Cancer Res. Treat. 2010, 121, 65–78. [Google Scholar] [CrossRef]
- Charpentier, M.S.; Whipple, R.A.; Vitolo, M.I.; Boggs, A.E.; Slovic, J.; Thompson, K.N.; Bhandary, L.; Martin, S.S. Curcumin targets breast cancer stem-like cells with microtentacles that persist in mammospheres and promote reattachment. Cancer Res. 2014, 74, 1250–1260. [Google Scholar] [CrossRef]
- Matrone, M.A.; Whipple, R.A.; Balzer, E.M.; Martin, S.S. Microtentacles tip the balance of cytoskeletal forces in circulating tumor cells. Cancer Res. 2010, 70, 7737–7741. [Google Scholar] [CrossRef] [PubMed]
- Whipple, R.A.; Matrone, M.A.; Cho, E.H.; Balzer, E.M.; Vitolo, M.I.; Yoon, J.R.; Ioffe, O.B.; Tuttle, K.C.; Yang, J.; Martin, S.S. Epithelial-to-mesenchymal transition promotes tubulin detyrosination and microtentacles that enhance endothelial engagement. Cancer Res. 2010, 70, 8127–8137. [Google Scholar] [CrossRef] [PubMed]
- Robinson, B.D.; Jones, J.G. Tumor microenvironment of metastasis (TMEM): A novel tissue-based assay for metastatic risk in breast cancer. Future Oncol. 2009, 5, 919–921. [Google Scholar] [CrossRef] [PubMed]
- Robinson, B.D.; Sica, G.L.; Liu, Y.F.; Rohan, T.E.; Gertler, F.B.; Condeelis, J.S.; Jones, J.G. Tumor microenvironment of metastasis in human breast carcinoma: A potential prognostic marker linked to hematogenous dissemination. Clin. Cancer Res. 2009, 15, 2433–2441. [Google Scholar] [CrossRef]
- Rohan, T.E.; Xue, X.; Lin, H.M.; D’Alfonso, T.M.; Ginter, P.S.; Oktay, M.H.; Robinson, B.D.; Ginsberg, M.; Gertler, F.B.; Glass, A.G.; et al. Tumor microenvironment of metastasis and risk of distant metastasis of breast cancer. J. Natl. Cancer Inst. 2014, 106. [Google Scholar] [CrossRef]
- Oktay, M.H.; Gertler, F.B.; Liu, Y.F.; Rohan, T.E.; Condeelis, J.S.; Jones, J.G. Correlated immunohistochemical and cytological assays for the prediction of hematogenous dissemination of breast cancer. J. Histochem. Cytochem. 2012, 60, 168–173. [Google Scholar] [CrossRef]
- Rastogi, P.; Anderson, S.J.; Bear, H.D.; Geyer, C.E.; Kahlenberg, M.S.; Robidoux, A.; Margolese, R.G.; Hoehn, J.L.; Vogel, V.G.; Dakhil, S.R.; et al. Preoperative chemotherapy: Updates of National Surgical Adjuvant Breast and Bowel Project Protocols B-18 and B-27. J. Clin. Oncol. 2008, 26, 778–785. [Google Scholar] [CrossRef]
- Gianni, L.; Dafni, U.; Gelber, R.D.; Azambuja, E.; Muehlbauer, S.; Goldhirsch, A.; Untch, M.; Smith, I.; Baselga, J.; Jackisch, C.; et al. Treatment with trastuzumab for 1 year after adjuvant chemotherapy in patients with HER2-positive early breast cancer: A 4-year follow-up of a randomised controlled trial. Lancet Oncol. 2011, 12, 236–244. [Google Scholar] [CrossRef]
- Harney, A.S.; Arwert, E.N.; Entenberg, D.; Wang, Y.; Guo, P.; Qian, B.Z.; Oktay, M.H.; Pollard, J.W.; Jones, J.G.; Condeelis, J.S. Real-Time Imaging Reveals Local, Transient Vascular Permeability, and Tumor Cell Intravasation Stimulated by TIE2hi Macrophage-Derived VEGFA. Cancer Discov. 2015, 5, 932–943. [Google Scholar] [CrossRef]
- Karagiannis, G.S.; Goswami, S.; Jones, J.G.; Oktay, M.H.; Condeelis, J.S. Signatures of breast cancer metastasis at a glance. J. Cell Sci. 2016, 129, 1751–1758. [Google Scholar] [CrossRef]
- Karagiannis, G.S.; Pastoriza, J.M.; Wang, Y.; Harney, A.S.; Entenberg, D.; Pignatelli, J.; Sharma, V.P.; Xue, E.A.; Cheng, E.; D’Alfonso, T.M.; et al. Neoadjuvant chemotherapy induces breast cancer metastasis through a TMEM-mediated mechanism. Sci. Transl. Med. 2017, 9. [Google Scholar] [CrossRef] [PubMed]
- Chang, Y.S.; Jalgaonkar, S.P.; Middleton, J.D.; Hai, T. Stress-inducible gene Atf3 in the noncancer host cells contributes to chemotherapy-exacerbated breast cancer metastasis. Proc. Natl. Acad. Sci. USA 2017, 114, E7159–E7168. [Google Scholar] [CrossRef] [PubMed]
- Goss, P.E.; Chambers, A.F. Does tumour dormancy offer a therapeutic target? Nat. Rev. Cancer 2010, 10, 871–877. [Google Scholar] [CrossRef] [PubMed]
- Chakrabarti, K.R.; Hessler, L.; Bhandary, L.; Martin, S.S. Molecular Pathways: New Signaling Considerations When Targeting Cytoskeletal Balance to Reduce Tumor Growth. Clin. Cancer Res. 2015, 21, 5209–5214. [Google Scholar] [CrossRef] [PubMed]
- Balzer, E.M.; Whipple, R.A.; Thompson, K.; Boggs, A.E.; Slovic, J.; Cho, E.H.; Matrone, M.A.; Yoneda, T.; Mueller, S.C.; Martin, S.S. c-Src differentially regulates the functions of microtentacles and invadopodia. Oncogene 2010, 29, 6402–6408. [Google Scholar] [CrossRef] [PubMed]
- Boggs, A.E.; Vitolo, M.I.; Whipple, R.A.; Charpentier, M.S.; Goloubeva, O.G.; Ioffe, O.B.; Tuttle, K.C.; Slovic, J.; Lu, Y.; Mills, G.B.; et al. alpha-Tubulin acetylation elevated in metastatic and basal-like breast cancer cells promotes microtentacle formation, adhesion, and invasive migration. Cancer Res. 2015, 75, 203–215. [Google Scholar] [CrossRef]
- Vitolo, M.I.; Boggs, A.E.; Whipple, R.A.; Yoon, J.R.; Thompson, K.; Matrone, M.A.; Cho, E.H.; Balzer, E.M.; Martin, S.S. Loss of PTEN induces microtentacles through PI3K-independent activation of cofilin. Oncogene 2013, 32, 2200–2210. [Google Scholar] [CrossRef]
- Whipple, R.A.; Balzer, E.M.; Cho, E.H.; Matrone, M.A.; Yoon, J.R.; Martin, S.S. Vimentin filaments support extension of tubulin-based microtentacles in detached breast tumor cells. Cancer Res. 2008, 68, 5678–5688. [Google Scholar] [CrossRef]
- Whipple, R.A.; Cheung, A.M.; Martin, S.S. Detyrosinated microtubule protrusions in suspended mammary epithelial cells promote reattachment. Exp. Cell Res. 2007, 313, 1326–1336. [Google Scholar] [CrossRef]
- Yoon, J.R.; Whipple, R.A.; Balzer, E.M.; Cho, E.H.; Matrone, M.A.; Peckham, M.; Martin, S.S. Local anesthetics inhibit kinesin motility and microtentacle protrusions in human epithelial and breast tumor cells. Breast Cancer Res. Treat. 2011, 129, 691–701. [Google Scholar] [CrossRef]
- Chiang, S.P.; Cabrera, R.M.; Segall, J.E. Tumor cell intravasation. Am. J. Physiol. Cell Physiol. 2016, 311, C1–C14. [Google Scholar] [CrossRef] [PubMed]
- Gupta, G.P.; Massague, J. Platelets and metastasis revisited: A novel fatty link. J. Clin. Investig. 2004, 114, 1691–1693. [Google Scholar] [CrossRef] [PubMed]
- Labelle, M.; Begum, S.; Hynes, R.O. Platelets guide the formation of early metastatic niches. Proc. Natl. Acad. Sci. USA 2014, 111, E3053–E3061. [Google Scholar] [CrossRef] [PubMed]
- Uppal, A.; Wightman, S.C.; Ganai, S.; Weichselbaum, R.R.; An, G. Investigation of the essential role of platelet-tumor cell interactions in metastasis progression using an agent-based model. Theor. Biol. Med. Model. 2014, 11, 17. [Google Scholar] [CrossRef]
- Gkountela, S.; Castro-Giner, F.; Szczerba, B.M.; Vetter, M.; Landin, J.; Scherrer, R.; Krol, I.; Scheidmann, M.C.; Beisel, C.; Stirnimann, C.U.; et al. Circulating Tumor Cell Clustering Shapes DNA Methylation to Enable Metastasis Seeding. Cell 2019, 176, 98–112.e114. [Google Scholar] [CrossRef]
- Zijlstra, A.; Von Lersner, A.; Yu, D.; Borrello, L.; Oudin, M.; Kang, Y.; Sahai, E.; Fingleton, B.; Stein, U.; Cox, T.R.; et al. The importance of developing therapies targeting the biological spectrum of metastatic disease. Clin. Exp. Metastasis 2019, 36, 305–309. [Google Scholar] [CrossRef]
- Anderson, R.L.; Balasas, T.; Callaghan, J.; Coombes, R.C.; Evans, J.; Hall, J.A.; Kinrade, S.; Jones, D.; Jones, P.S.; Jones, R.; et al. A framework for the development of effective anti-metastatic agents. Nat. Rev. Clin. Oncol. 2019, 16, 185–204. [Google Scholar] [CrossRef]
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mathias, T.J.; Chang, K.T.; Martin, S.S.; Vitolo, M.I. Gauging the Impact of Cancer Treatment Modalities on Circulating Tumor Cells (CTCs). Cancers 2020, 12, 743. https://doi.org/10.3390/cancers12030743
Mathias TJ, Chang KT, Martin SS, Vitolo MI. Gauging the Impact of Cancer Treatment Modalities on Circulating Tumor Cells (CTCs). Cancers. 2020; 12(3):743. https://doi.org/10.3390/cancers12030743
Chicago/Turabian StyleMathias, Trevor J., Katarina T. Chang, Stuart S. Martin, and Michele I. Vitolo. 2020. "Gauging the Impact of Cancer Treatment Modalities on Circulating Tumor Cells (CTCs)" Cancers 12, no. 3: 743. https://doi.org/10.3390/cancers12030743
APA StyleMathias, T. J., Chang, K. T., Martin, S. S., & Vitolo, M. I. (2020). Gauging the Impact of Cancer Treatment Modalities on Circulating Tumor Cells (CTCs). Cancers, 12(3), 743. https://doi.org/10.3390/cancers12030743