Circulating Cells with Macrophage-like Characteristics in Cancer: The Importance of Circulating Neoplastic-Immune Hybrid Cells in Cancer
Abstract
:Simple Summary
Abstract
1. Cancer Biomarkers: In Search of the Holy Grail
2. Circulating Tumor Cells
3. Tumor-Associated Macrophages, Phagocytosis, and Cancer-Associated Macrophage-Like Cells (CAMLs)
3.1. Tumor-Associated Macrophages
3.2. Cancer-Associated Macrophage-Like Cells
4. Tumor Cell Fusion, Tumor-Immune Hybrids, and Circulating Hybrid Cells
4.1. Intratumoral Tumor–Immune Hybrids
4.2. Tumor–Immune Hybrids in Circulation
4.3. CHCs as a Biomarker in Human Malignancy
5. Discussion
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Lieberman, D.A.; Weiss, D.G.; Bond, J.H.; Ahnen, D.J.; Garewal, H.; Chejfec, G. Use of colonoscopy to screen asymptomatic adults for colorectal cancer. Veterans Affairs Cooperative Study Group 380. N. Engl. J. Med. 2000, 343, 162–168. [Google Scholar] [CrossRef] [PubMed]
- Qumseya, B.; Sultan, S.; Bain, P.; Jamil, L.; Jacobson, B.; Anandasabapathy, S.; Agrawal, D.; Buxbaum, J.L.; Fishman, D.S.; Gurudu, S.R.; et al. ASGE guideline on screening and surveillance of Barrett’s esophagus. Gastrointest. Endosc. 2019, 90, 335–359. [Google Scholar] [CrossRef] [Green Version]
- Zhang, M.; Qin, X.; Xu, W.; Wang, Y.; Song, Y.; Garg, S.; Luan, Y. Engineering of a dual-modal phototherapeutic nanoplatform for single NIR laser-triggered tumor therapy. J. Colloid Interface Sci. 2021, 594, 493–501. [Google Scholar] [CrossRef] [PubMed]
- O’Connor, J.P.; Aboagye, E.O.; Adams, J.E.; Aerts, H.J.; Barrington, S.F.; Beer, A.J.; Boellaard, R.; Bohndiek, S.E.; Brady, M.; Brown, G.; et al. Imaging biomarker roadmap for cancer studies. Nat. Rev. Clin. Oncol. 2017, 14, 169–186. [Google Scholar] [CrossRef] [PubMed]
- Rosenkrantz, A.B.; Duszak, R., Jr.; Babb, J.S.; Glover, M.; Kang, S.K. Discrepancy Rates and Clinical Impact of Imaging Secondary Interpretations: A Systematic Review and Meta-Analysis. J. Am. Coll. Radiol. 2018, 15, 1222–1231. [Google Scholar] [CrossRef]
- Schlemmer, H.P.; Bittencourt, L.K.; D’Anastasi, M.; Domingues, R.; Khong, P.L.; Lockhat, Z.; Muellner, A.; Reiser, M.F.; Schilsky, R.L.; Hricak, H. Global Challenges for Cancer Imaging. J. Glob. Oncol. 2018, 4, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Ariga, R.; Bloom, K.; Reddy, V.B.; Kluskens, L.; Francescatti, D.; Dowlat, K.; Siziopikou, P.; Gattuso, P. Fine-needle aspiration of clinically suspicious palpable breast masses with histopathologic correlation. Am. J. Surg. 2002, 184, 410–413. [Google Scholar] [CrossRef]
- Strimbu, K.; Tavel, J.A. What are biomarkers? Curr. Opin. HIV AIDS 2010, 5, 463–466. [Google Scholar] [CrossRef]
- Cooperman, A.M.; Iskandar, M.E.; Wayne, M.G.; Steele, J.G. Prevention and Early Detection of Pancreatic Cancer. Surg. Clin. N. Am. 2018, 98, 1–12. [Google Scholar] [CrossRef]
- Locker, G.Y.; Hamilton, S.; Harris, J.; Jessup, J.M.; Kemeny, N.; Macdonald, J.S.; Somerfield, M.R.; Hayes, D.F.; Bast, R.C., Jr. ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2006, 24, 5313–5327. [Google Scholar] [CrossRef] [PubMed]
- Brabletz, T. EMT and MET in metastasis: Where are the cancer stem cells? Cancer Cell 2012, 22, 699–701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dalerba, P.; Cho, R.W.; Clarke, M.F. Cancer stem cells: Models and concepts. Annu. Rev. Med. 2007, 58, 267–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leon, S.A.; Shapiro, B.; Sklaroff, D.M.; Yaros, M.J. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 1977, 37, 646–650. [Google Scholar]
- Stroun, M.; Anker, P.; Maurice, P.; Lyautey, J.; Lederrey, C.; Beljanski, M. Neoplastic characteristics of the DNA found in the plasma of cancer patients. Oncology 1989, 46, 318–322. [Google Scholar] [CrossRef]
- de Kok, J.B.; van Solinge, W.W.; Ruers, T.J.; Roelofs, R.W.; van Muijen, G.N.; Willems, J.L.; Swinkels, D.W. Detection of tumour DNA in serum of colorectal cancer patients. Scand. J. Clin. Lab. Investig. 1997, 57, 601–604. [Google Scholar] [CrossRef]
- 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. Off. J. Am. Soc. Clin. Oncol. 2008, 26, 3213–3221. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Krebs, M.G.; Sloane, R.; Priest, L.; Lancashire, L.; Hou, J.M.; Greystoke, A.; Ward, T.H.; Ferraldeschi, R.; Hughes, A.; Clack, G.; et al. Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2011, 29, 1556–1563. [Google Scholar] [CrossRef]
- Torphy, R.J.; Tignanelli, C.J.; Kamande, J.W.; Moffitt, R.A.; Herrera Loeza, S.G.; Soper, S.A.; Yeh, J.J. Circulating tumor cells as a biomarker of response to treatment in patient-derived xenograft mouse models of pancreatic adenocarcinoma. PLoS ONE 2014, 9, e89474. [Google Scholar] [CrossRef]
- Court, C.M.; Ankeny, J.S.; Sho, S.; Winograd, P.; Hou, S.; Song, M.; Wainberg, Z.A.; Girgis, M.D.; Graeber, T.G.; Agopian, V.G.; et al. Circulating Tumor Cells Predict Occult Metastatic Disease and Prognosis in Pancreatic Cancer. Ann. Surg. Oncol. 2018, 25, 1000–1008. [Google Scholar] [CrossRef] [PubMed]
- Miller, M.C.; Doyle, G.V.; Terstappen, L.W. Significance of Circulating Tumor Cells Detected by the CellSearch System in Patients with Metastatic Breast Colorectal and Prostate Cancer. J. Oncol. 2010, 2010, 617421. [Google Scholar] [CrossRef] [PubMed]
- van Dalum, G.; Stam, G.J.; Scholten, L.F.; Mastboom, W.J.; Vermes, I.; Tibbe, A.G.; De Groot, M.R.; Terstappen, L.W. Importance of circulating tumor cells in newly diagnosed colorectal cancer. Int. J. Oncol. 2015, 46, 1361–1368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [Green Version]
- Boral, D.; Vishnoi, M.; Liu, H.N.; Yin, W.; Sprouse, M.L.; Scamardo, A.; Hong, D.S.; Tan, T.Z.; Thiery, J.P.; Chang, J.C.; et al. Molecular characterization of breast cancer CTCs associated with brain metastasis. Nat. Commun. 2017, 8, 196. [Google Scholar] [CrossRef] [Green Version]
- Giuliano, M.; Giordano, A.; Jackson, S.; De Giorgi, U.; Mego, M.; Cohen, E.N.; Gao, H.; Anfossi, S.; Handy, B.C.; Ueno, N.T.; et al. Circulating tumor cells as early predictors of metastatic spread in breast cancer patients with limited metastatic dissemination. Breast Cancer Res. BCR 2014, 16, 440. [Google Scholar] [CrossRef] [Green Version]
- Lucci, A.; Hall, C.S.; Lodhi, A.K.; Bhattacharyya, A.; Anderson, A.E.; Xiao, L.; Bedrosian, I.; Kuerer, H.M.; Krishnamurthy, S. Circulating tumour cells in non-metastatic breast cancer: A prospective study. Lancet Oncol. 2012, 13, 688–695. [Google Scholar] [CrossRef]
- Bidard, F.C.; Michiels, S.; Riethdorf, S.; Mueller, V.; Esserman, L.J.; Lucci, A.; Naume, B.; Horiguchi, J.; Gisbert-Criado, R.; Sleijfer, S.; et al. Circulating Tumor Cells in Breast Cancer Patients Treated by Neoadjuvant Chemotherapy: A Meta-analysis. J. Natl. Cancer Inst. 2018, 110, 560–567. [Google Scholar] [CrossRef]
- Gast, C.E.; Silk, A.D.; Zarour, L.; Riegler, L.; Burkhart, J.G.; Gustafson, K.T.; Parappilly, M.S.; Roh-Johnson, M.; Goodman, J.R.; Olson, B.; et al. Cell fusion potentiates tumor heterogeneity and reveals circulating hybrid cells that correlate with stage and survival. Sci. Adv. 2018, 4, eaat7828. [Google Scholar] [CrossRef] [Green Version]
- Ashworth, T. A case of cancer in which cells similar to those in the tumors were seen in the blood after death. Austrailian Med. J. 1869, 1869, 146–147. [Google Scholar]
- Adams, D.L.; Martin, S.S.; Alpaugh, R.K.; Charpentier, M.; Tsai, S.; Bergan, R.C.; Ogden, I.M.; Catalona, W.; Chumsri, S.; Tang, C.M.; et al. Circulating giant macrophages as a potential biomarker of solid tumors. Proc. Natl. Acad. Sci. USA 2014, 111, 3514–3519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sutton, T.L.; Walker, B.S.; Wong, M.H. Circulating Hybrid Cells Join the Fray of Circulating Cellular Biomarkers. Cell Mol. Gastroenterol. Hepatol. 2019, 8, 595–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dietz, M.S.; Sutton, T.L.; Walker, B.S.; Gast, C.E.; Zarour, L.; Sengupta, S.K.; Swain, J.R.; Eng, J.; Parappilly, M.; Limbach, K.; et al. Relevance of circulating hybrid cells as a non-invasive biomarker for myriad solid tumors. Sci. Rep. 2021, 11, 13630. [Google Scholar] [CrossRef]
- Scher, H.I.; Jia, X.; de Bono, J.S.; Fleisher, M.; Pienta, K.J.; Raghavan, D.; Heller, G. Circulating tumour cells as prognostic markers in progressive, castration-resistant prostate cancer: A reanalysis of IMMC38 trial data. Lancet Oncol. 2009, 10, 233–239. [Google Scholar] [CrossRef] [Green Version]
- Rosenberg, R.; Gertler, R.; Friederichs, J.; Fuehrer, K.; Dahm, M.; Phelps, R.; Thorban, S.; Nekarda, H.; Siewert, J.R. Comparison of two density gradient centrifugation systems for the enrichment of disseminated tumor cells in blood. Cytometry 2002, 49, 150–158. [Google Scholar] [CrossRef] [PubMed]
- Campton, D.E.; Ramirez, A.B.; Nordberg, J.J.; Drovetto, N.; Clein, A.C.; Varshavskaya, P.; Friemel, B.H.; Quarre, S.; Breman, A.; Dorschner, M.; et al. High-recovery visual identification and single-cell retrieval of circulating tumor cells for genomic analysis using a dual-technology platform integrated with automated immunofluorescence staining. BMC Cancer 2015, 15, 360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Elvington, E.S.; Salmanzadeh, A.; Stremler, M.A.; Davalos, R.V. Label-free isolation and enrichment of cells through contactless dielectrophoresis. J. Vis. Exp. JoVE 2013, 79, e50634. [Google Scholar] [CrossRef] [Green Version]
- Fernandez, S.V.; Bingham, C.; Fittipaldi, P.; Austin, L.; Palazzo, J.; Palmer, G.; Alpaugh, K.; Cristofanilli, M. TP53 mutations detected in circulating tumor cells present in the blood of metastatic triple negative breast cancer patients. Breast Cancer Res. BCR 2014, 16, 445. [Google Scholar] [CrossRef] [Green Version]
- Gupta, V.; Jafferji, I.; Garza, M.; Melnikova, V.O.; Hasegawa, D.K.; Pethig, R.; Davis, D.W. ApoStream(), a new dielectrophoretic device for antibody independent isolation and recovery of viable cancer cells from blood. Biomicrofluidics 2012, 6, 24133. [Google Scholar] [CrossRef] [Green Version]
- Freidin, M.B.; Tay, A.; Freydina, D.V.; Chudasama, D.; Nicholson, A.G.; Rice, A.; Anikin, V.; Lim, E. An assessment of diagnostic performance of a filter-based antibody-independent peripheral blood circulating tumour cell capture paired with cytomorphologic criteria for the diagnosis of cancer. Lung Cancer 2014, 85, 182–185. [Google Scholar] [CrossRef]
- Farace, F.; Massard, C.; Vimond, N.; Drusch, F.; Jacques, N.; Billiot, F.; Laplanche, A.; Chauchereau, A.; Lacroix, L.; Planchard, D.; et al. A direct comparison of CellSearch and ISET for circulating tumour-cell detection in patients with metastatic carcinomas. Br. J. Cancer 2011, 105, 847–853. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adams, D.L.; Stefansson, S.; Haudenschild, C.; Martin, S.S.; Charpentier, M.; Chumsri, S.; Cristofanilli, M.; Tang, C.M.; Alpaugh, R.K. Cytometric characterization of circulating tumor cells captured by microfiltration and their correlation to the CellSearch((R)) CTC test. Cytom. Part A J. Int. Soc. Anal. Cytol. 2015, 87, 137–144. [Google Scholar] [CrossRef] [Green Version]
- Raimondi, C.; Nicolazzo, C.; Gradilone, A.; Giannini, G.; De Falco, E.; Chimenti, I.; Varriale, E.; Hauch, S.; Plappert, L.; Cortesi, E.; et al. Circulating tumor cells: Exploring intratumor heterogeneity of colorectal cancer. Cancer Biol. Ther. 2014, 15, 496–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Talasaz, A.H.; Powell, A.A.; Huber, D.E.; Berbee, J.G.; Roh, K.H.; Yu, W.; Xiao, W.; Davis, M.M.; Pease, R.F.; Mindrinos, M.N.; et al. Isolating highly enriched populations of circulating epithelial cells and other rare cells from blood using a magnetic sweeper device. Proc. Natl. Acad. Sci. USA 2009, 106, 3970–3975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Winer-Jones, J.P.; Vahidi, B.; Arquilevich, N.; Fang, C.; Ferguson, S.; Harkins, D.; Hill, C.; Klem, E.; Pagano, P.C.; Peasley, C.; et al. Circulating tumor cells: Clinically relevant molecular access based on a novel CTC flow cell. PLoS ONE 2014, 9, e86717. [Google Scholar] [CrossRef]
- Nagrath, S.; Sequist, L.V.; Maheswaran, S.; Bell, D.W.; Irimia, D.; Ulkus, L.; Smith, M.R.; Kwak, E.L.; Digumarthy, S.; Muzikansky, A.; et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature 2007, 450, 1235–1239. [Google Scholar] [CrossRef] [Green Version]
- Brychta, N.; Drosch, M.; Driemel, C.; Fischer, J.C.; Neves, R.P.; Esposito, I.; Knoefel, W.; Mohlendick, B.; Hille, C.; Stresemann, A.; et al. Isolation of circulating tumor cells from pancreatic cancer by automated filtration. Oncotarget 2017, 8, 86143–86156. [Google Scholar] [CrossRef] [Green Version]
- Chen, F.; Wang, S.; Fang, Y.; Zheng, L.; Zhi, X.; Cheng, B.; Chen, Y.; Zhang, C.; Shi, D.; Song, H.; et al. Feasibility of a novel one-stop ISET device to capture CTCs and its clinical application. Oncotarget 2017, 8, 3029–3041. [Google Scholar] [CrossRef] [Green Version]
- Millner, L.M.; Linder, M.W.; Valdes, R., Jr. Circulating tumor cells: A review of present methods and the need to identify heterogeneous phenotypes. Ann. Clin. Lab. Sci. 2013, 43, 295–304. [Google Scholar]
- Zhang, Y.; Wang, F.; Ning, N.; Chen, Q.; Yang, Z.; Guo, Y.; Xu, D.; Zhang, D.; Zhan, T.; Cui, W. Patterns of circulating tumor cells identified by CEP8, CK and CD45 in pancreatic cancer. Int. J. Cancer 2015, 136, 1228–1233. [Google Scholar] [CrossRef]
- Poruk, K.E.; Valero, V.; Saunders, T.; Blackford, A.L.; Griffin, J.F.; Poling, J.; Hruban, R.H.; Anders, R.A.; Herman, J.; Zheng, L.; et al. Circulating Tumor Cell Phenotype Predicts Recurrence and Survival in Pancreatic Adenocarcinoma. Ann. Surg. 2016, 264, 1073–1081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Poruk, K.E.; Blackford, A.L.; Weiss, M.J.; Cameron, J.L.; He, J.; Goggins, M.; Rasheed, Z.A.; Wolfgang, C.L.; Wood, L.D. Circulating Tumor Cells Expressing Markers of Tumor-Initiating Cells Predict Poor Survival and Cancer Recurrence in Patients with Pancreatic Ductal Adenocarcinoma. Clin. Cancer Res. An. Off. J. Am. Assoc. Cancer Res. 2017, 23, 2681–2690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gemenetzis, G.; Groot, V.P.; Yu, J.; Ding, D.; Teinor, J.A.; Javed, A.A.; Wood, L.D.; Burkhart, R.A.; Cameron, J.L.; Makary, M.A.; et al. Circulating Tumor Cells Dynamics in Pancreatic Adenocarcinoma Correlate with Disease Status: Results of the Prospective CLUSTER Study. Ann. Surg. 2018, 268, 408–420. [Google Scholar] [CrossRef] [PubMed]
- Andree, K.C.; van Dalum, G.; Terstappen, L.W. Challenges in circulating tumor cell detection by the CellSearch system. Mol. Oncol. 2016, 10, 395–407. [Google Scholar] [CrossRef] [Green Version]
- 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. Off. J. Am. Soc. Clin. Oncol. 2014, 32, 3483–3489. [Google Scholar] [CrossRef]
- Cabel, L.; Berger, F.; Cottu, P.; Loirat, D.; Rampanou, A.; Brain, E.; Cyrille, S.; Bourgeois, H.; Kiavue, N.; Deluche, E.; et al. Clinical utility of circulating tumour cell-based monitoring of late-line chemotherapy for metastatic breast cancer: The randomised CirCe01 trial. Br. J. Cancer 2021, 124, 1207–1213. [Google Scholar] [CrossRef]
- Bidard, F.C.; Jacot, W.; Kiavue, N.; Dureau, S.; Kadi, A.; Brain, E.; Bachelot, T.; Bourgeois, H.; Gonçalves, A.; Ladoire, S.; et al. Efficacy of Circulating Tumor Cell Count-Driven vs. Clinician-Driven First-line Therapy Choice in Hormone Receptor-Positive, ERBB2-Negative Metastatic Breast Cancer: The STIC CTC Randomized Clinical Trial. JAMA Oncol. 2021, 7, 34–41. [Google Scholar] [CrossRef]
- Xu, Y.; Qin, T.; Li, J.; Wang, X.; Gao, C.; Xu, C.; Hao, J.; Liu, J.; Gao, S.; Ren, H. Detection of Circulating Tumor Cells Using Negative Enrichment Immunofluorescence and an In Situ Hybridization System in Pancreatic Cancer. Int. J. Mol. Sci. 2017, 18, 622. [Google Scholar] [CrossRef] [Green Version]
- Cai, J.; Huang, L.; Huang, J.; Kang, L.; Lin, H.; Huang, P.; Zhu, P.; Wang, J.; Dong, J.; Wang, L.; et al. Associations between the cyclooxygenase-2 expression in circulating tumor cells and the clinicopathological features of patients with colorectal cancer. J. Cell. Biochem. 2019, 120, 4935–4941. [Google Scholar] [CrossRef]
- Messaritakis, I.; Sfakianaki, M.; Papadaki, C.; Koulouridi, A.; Vardakis, N.; Koinis, F.; Hatzidaki, D.; Georgoulia, N.; Kladi, A.; Kotsakis, A.; et al. Prognostic significance of CEACAM5mRNA-positive circulating tumor cells in patients with metastatic colorectal cancer. Cancer Chemother. Pharmacol. 2018, 82, 767–775. [Google Scholar] [CrossRef]
- Wang, W.; Wan, L.; Wu, S.; Yang, J.; Zhou, Y.; Liu, F.; Wu, Z.; Cheng, Y. Mesenchymal marker and LGR5 expression levels in circulating tumor cells correlate with colorectal cancer prognosis. Cell. Oncol. 2018, 41, 495–504. [Google Scholar] [CrossRef] [PubMed]
- Zhao, X.H.; Wang, Z.R.; Chen, C.L.; Di, L.; Bi, Z.F.; Li, Z.H.; Liu, Y.M. Molecular detection of epithelial-mesenchymal transition markers in circulating tumor cells from pancreatic cancer patients: Potential role in clinical practice. World J. Gastroenterol. 2019, 25, 138–150. [Google Scholar] [CrossRef] [PubMed]
- Zhao, R.; Cai, Z.; Li, S.; Cheng, Y.; Gao, H.; Liu, F.; Wu, S.; Liu, S.; Dong, Y.; Zheng, L.; et al. Expression and clinical relevance of epithelial and mesenchymal markers in circulating tumor cells from colorectal cancer. Oncotarget 2017, 8, 9293–9302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Franklin, R.A.; Liao, W.; Sarkar, A.; Kim, M.V.; Bivona, M.R.; Liu, K.; Pamer, E.G.; Li, M.O. The cellular and molecular origin of tumor-associated macrophages. Science 2014, 344, 921–925. [Google Scholar] [CrossRef] [Green Version]
- Henze, A.T.; Mazzone, M. The impact of hypoxia on tumor-associated macrophages. J. Clin. Investig. 2016, 126, 3672–3679. [Google Scholar] [CrossRef]
- Morita, Y.; Zhang, R.; Leslie, M.; Adhikari, S.; Hasan, N.; Chervoneva, I.; Rui, H.; Tanaka, T. Pathologic evaluation of tumor-associated macrophage density and vessel inflammation in invasive breast carcinomas. Oncol. Lett. 2017, 14, 2111–2118. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.; Bae, J.S. Tumor-Associated Macrophages and Neutrophils in Tumor Microenvironment. Mediat. Inflamm. 2016, 2016, 6058147. [Google Scholar] [CrossRef] [Green Version]
- Malyshev, I.; Malyshev, Y. Current Concept and Update of the Macrophage Plasticity Concept: Intracellular Mechanisms of Reprogramming and M3 Macrophage “Switch” Phenotype. BioMed Res. Int. 2015, 2015, 341308. [Google Scholar] [CrossRef] [Green Version]
- Hirayama, D.; Iida, T.; Nakase, H. The Phagocytic Function of Macrophage-Enforcing Innate Immunity and Tissue Homeostasis. Int. J. Mol. Sci. 2017, 19, 92. [Google Scholar] [CrossRef] [Green Version]
- Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
- Chen, D.; Zhang, X.; Li, Z.; Zhu, B. Metabolic regulatory crosstalk between tumor microenvironment and tumor-associated macrophages. Theranostics 2021, 11, 1016–1030. [Google Scholar] [CrossRef] [PubMed]
- Cheng, N.; Bai, X.; Shu, Y.; Ahmad, O.; Shen, P. Targeting tumor-associated macrophages as an antitumor strategy. Biochem. Pharm. 2021, 183, 114354. [Google Scholar] [CrossRef] [PubMed]
- Mosser, D.M.; Edwards, J.P. Exploring the full spectrum of macrophage activation. Nat. Rev. Immunol. 2008, 8, 958–969. [Google Scholar] [CrossRef]
- Chanmee, T.; Ontong, P.; Konno, K.; Itano, N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers 2014, 6, 1670–1690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lavin, Y.; Mortha, A.; Rahman, A.; Merad, M. Regulation of macrophage development and function in peripheral tissues. Nat. Rev. Immunol. 2015, 15, 731–744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martinez, F.O.; Gordon, S. The M1 and M2 paradigm of macrophage activation: Time for reassessment. F1000Prime Rep. 2014, 6, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef] [Green Version]
- Zhou, J.; Tang, Z.; Gao, S.; Li, C.; Feng, Y.; Zhou, X. Tumor-Associated Macrophages: Recent Insights and Therapies. Front. Oncol. 2020, 10, 188. [Google Scholar] [CrossRef]
- Italiani, P.; Boraschi, D. From Monocytes to M1/M2 Macrophages: Phenotypical vs. Functional Differentiation. Front. Immunol. 2014, 5, 514. [Google Scholar] [CrossRef] [Green Version]
- Unanue, E.R. Antigen-presenting function of the macrophage. Annu. Rev. Immunol. 1984, 2, 395–428. [Google Scholar] [CrossRef]
- Vignery, A. Osteoclasts and giant cells: Macrophage-macrophage fusion mechanism. Int. J. Exp. Pathol. 2000, 81, 291–304. [Google Scholar] [CrossRef] [PubMed]
- Tang, C.M.; Zhu, P.; Li, S.; Makarova, O.V.; Amstutz, P.T.; Adams, D.L. Blood-based biopsies-clinical utility beyond circulating tumor cells. Cytom. Part A J. Int. Soc. Anal. Cytol. 2018, 93, 1246–1250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mu, Z.; Benali-Furet, N.; Uzan, G.; Znaty, A.; Ye, Z.; Paolillo, C.; Wang, C.; Austin, L.; Rossi, G.; Fortina, P.; et al. Detection and Characterization of Circulating Tumor Associated Cells in Metastatic Breast Cancer. Int. J. Mol. Sci. 2016, 17, 1665. [Google Scholar] [CrossRef] [PubMed]
- Adams, D.L.; Adams, D.K.; Alpaugh, R.K.; Cristofanilli, M.; Martin, S.S.; Chumsri, S.; Tang, C.M.; Marks, J.R. Circulating Cancer-Associated Macrophage-Like Cells Differentiate Malignant Breast Cancer and Benign Breast Conditions. Cancer Epidemiol. Biomark. Prev. 2016, 25, 1037–1042. [Google Scholar] [CrossRef] [Green Version]
- Mu, Z.; Wang, C.; Ye, Z.; Rossi, G.; Sun, C.; Li, L.; Zhu, Z.; Yang, H.; Cristofanilli, M. Prognostic values of cancer associated macrophage-like cells (CAML) enumeration in metastatic breast cancer. Breast Cancer Res. Treat. 2017, 165, 733–741. [Google Scholar] [CrossRef] [PubMed]
- Gironda, D.J.; Adams, D.L.; He, J.; Xu, T.; Gao, H.; Qiao, Y.; Komaki, R.; Reuben, J.M.; Liao, Z.; Blum-Murphy, M.; et al. Cancer associated macrophage-like cells and prognosis of esophageal cancer after chemoradiation therapy. J. Transl. Med. 2020, 18, 413. [Google Scholar] [CrossRef]
- Augustyn, A.; Adams, D.L.; He, J.; Qiao, Y.; Verma, V.; Liao, Z.; Tang, C.M.; Heymach, J.V.; Tsao, A.S.; Lin, S.H. Giant Circulating Cancer-Associated Macrophage-Like Cells Are Associated With Disease Recurrence and Survival in Non-Small-Cell Lung Cancer Treated With Chemoradiation and Atezolizumab. Clin. Lung Cancer 2021, 22, e451–e465. [Google Scholar] [CrossRef]
- Saman, S.; Stagno, M.J.; Warmann, S.W.; Malek, N.P.; Plentz, R.R.; Schmid, E. Biomarkers Apo10 and TKTL1: Epitope-detection in monocytes (EDIM) as a new diagnostic approach for cholangiocellular, pancreatic and colorectal carcinoma. Cancer Biomark. 2020, 27, 129–137. [Google Scholar] [CrossRef] [Green Version]
- Grimm, M.; Kraut, W.; Hoefert, S.; Krimmel, M.; Biegner, T.; Teriete, P.; Cetindis, M.; Polligkeit, J.; Kluba, S.; Munz, A.; et al. Evaluation of a biomarker based blood test for monitoring surgical resection of oral squamous cell carcinomas. Clin. Oral. Investig. 2016, 20, 329–338. [Google Scholar] [CrossRef]
- Aichel, O. Über Zellverschmelzung mit Qualitativ Abnormer Chromosomenverteilung als Ursache der Geschwulstbildung [About Cell Fusion with Qualitatively Abnormal Chromosome Distribution as Cause for Tumor Formation]; Wilhelm Engelmann: Leipzig, Germany, 1911. [Google Scholar]
- Singec, I.; Snyder, E.Y. Inflammation as a matchmaker: Revisiting cell fusion. Nat. Cell Biol. 2008, 10, 503–505. [Google Scholar] [CrossRef]
- Johansson, C.B.; Youssef, S.; Koleckar, K.; Holbrook, C.; Doyonnas, R.; Corbel, S.Y.; Steinman, L.; Rossi, F.M.; Blau, H.M. Extensive fusion of haematopoietic cells with Purkinje neurons in response to chronic inflammation. Nat. Cell Biol. 2008, 10, 575–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nygren, J.M.; Liuba, K.; Breitbach, M.; Stott, S.; Thoren, L.; Roell, W.; Geisen, C.; Sasse, P.; Kirik, D.; Bjorklund, A.; et al. Myeloid and lymphoid contribution to non-haematopoietic lineages through irradiation-induced heterotypic cell fusion. Nat. Cell Biol. 2008, 10, 584–592. [Google Scholar] [CrossRef] [PubMed]
- Rizvi, A.Z.; Swain, J.R.; Davies, P.S.; Bailey, A.S.; Decker, A.D.; Willenbring, H.; Grompe, M.; Fleming, W.H.; Wong, M.H. Bone marrow-derived cells fuse with normal and transformed intestinal stem cells. Proc. Natl. Acad. Sci. USA 2006, 103, 6321–6325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Powell, A.E.; Anderson, E.C.; Davies, P.S.; Silk, A.D.; Pelz, C.; Impey, S.; Wong, M.H. Fusion between Intestinal epithelial cells and macrophages in a cancer context results in nuclear reprogramming. Cancer Res. 2011, 71, 1497–1505. [Google Scholar] [CrossRef] [Green Version]
- Davies, P.S.; Powell, A.E.; Swain, J.R.; Wong, M.H. Inflammation and proliferation act together to mediate intestinal cell fusion. PLoS ONE 2009, 4, e6530. [Google Scholar] [CrossRef]
- Silk, A.D.; Gast, C.E.; Davies, P.S.; Fakhari, F.D.; Vanderbeek, G.E.; Mori, M.; Wong, M.H. Fusion between hematopoietic and epithelial cells in adult human intestine. PLoS ONE 2013, 8, e55572. [Google Scholar] [CrossRef] [Green Version]
- Duelli, D.; Lazebnik, Y. Cell fusion: A hidden enemy? Cancer Cell 2003, 3, 445–448. [Google Scholar] [CrossRef] [Green Version]
- Pawelek, J.M. Tumour-cell fusion as a source of myeloid traits in cancer. Lancet Oncol. 2005, 6, 988–993. [Google Scholar] [CrossRef]
- Pawelek, J.M. Tumour cell hybridization and metastasis revisited. Melanoma Res. 2000, 10, 507–514. [Google Scholar] [CrossRef]
- Pawelek, J.M.; Chakraborty, A.K. Fusion of tumour cells with bone marrow-derived cells: A unifying explanation for metastasis. Nat. Rev. Cancer 2008, 8, 377–386. [Google Scholar] [CrossRef]
- Pawelek, J.M.; Chakraborty, A.K. The cancer cell–Leukocyte fusion theory of metastasis. Adv. Cancer Res. 2008, 101, 397–444. [Google Scholar] [CrossRef] [PubMed]
- Ramakrishnan, M.; Mathur, S.R.; Mukhopadhyay, A. Fusion-derived epithelial cancer cells express hematopoietic markers and contribute to stem cell and migratory phenotype in ovarian carcinoma. Cancer Res. 2013, 73, 5360–5370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walker, B.S.; Sutton, T.L.; Zarour, L.; Hunter, J.G.; Wood, S.G.; Tsikitis, V.L.; Herzig, D.O.; Lopez, C.D.; Chen, E.Y.; Mayo, S.C.; et al. Circulating Hybrid Cells: A Novel Liquid Biomarker of Treatment Response in Gastrointestinal Cancers. Ann. Surg. Oncol. 2021, 28, 8567–8578. [Google Scholar] [CrossRef] [PubMed]
- Henn, T.E.; Anderson, A.N.; Hollett, Y.R.; Sutton, T.L.; Walker, B.S.; Swain, J.R.; Sauer, D.A.; Clayburgh, D.R.; Wong, M.H. Circulating hybrid cells predict presence of occult nodal metastases in oral cavity carcinoma. Head Neck 2021, 43, 2193–2201. [Google Scholar] [CrossRef] [PubMed]
- Clawson, G.A.; Matters, G.L.; Xin, P.; Imamura-Kawasawa, Y.; Du, Z.; Thiboutot, D.M.; Helm, K.F.; Neves, R.I.; Abraham, T. Macrophage-tumor cell fusions from peripheral blood of melanoma patients. PLoS ONE 2015, 10, e0134320. [Google Scholar] [CrossRef] [Green Version]
- Clawson, G.A.; Matters, G.L.; Xin, P.; McGovern, C.; Wafula, E.; dePamphilis, C.; Meckley, M.; Wong, J.; Stewart, L.; D’Jamoos, C.; et al. “Stealth dissemination” of macrophage-tumor cell fusions cultured from blood of patients with pancreatic ductal adenocarcinoma. PLoS ONE 2017, 12, e0184451. [Google Scholar] [CrossRef] [Green Version]
- Cheng, K.S.; Pan, R.; Pan, H.; Li, B.; Meena, S.S.; Xing, H.; Ng, Y.J.; Qin, K.; Liao, X.; Kosgei, B.K.; et al. ALICE: A hybrid AI paradigm with enhanced connectivity and cybersecurity for a serendipitous encounter with circulating hybrid cells. Theranostics 2020, 10, 11026–11048. [Google Scholar] [CrossRef]
- Lazova, R.; Laberge, G.S.; Duvall, E.; Spoelstra, N.; Klump, V.; Sznol, M.; Cooper, D.; Spritz, R.A.; Chang, J.T.; Pawelek, J.M. A Melanoma Brain Metastasis with a Donor-Patient Hybrid Genome following Bone Marrow Transplantation: First Evidence for Fusion in Human Cancer. PLoS ONE 2013, 8, e66731. [Google Scholar] [CrossRef]
- LaBerge, G.S.; Duvall, E.; Grasmick, Z.; Haedicke, K.; Pawelek, J. A Melanoma Lymph Node Metastasis with a Donor-Patient Hybrid Genome following Bone Marrow Transplantation: A Second Case of Leucocyte-Tumor Cell Hybridization in Cancer Metastasis. PLoS ONE 2017, 12, e0168581. [Google Scholar] [CrossRef]
- Manjunath, Y.; Mitchem, J.B.; Suvilesh, K.N.; Avella, D.M.; Kimchi, E.T.; Staveley-O’Carroll, K.F.; Deroche, C.B.; Pantel, K.; Li, G.; Kaifi, J.T. Circulating Giant Tumor-Macrophage Fusion Cells Are Independent Prognosticators in Patients With NSCLC. J. Thorac. Oncol. 2020, 15, 1460–1471. [Google Scholar] [CrossRef]
- Lizier, M.; Anselmo, A.; Mantero, S.; Ficara, F.; Paulis, M.; Vezzoni, P.; Lucchini, F.; Pacchiana, G. Fusion between cancer cells and macrophages occurs in a murine model of spontaneous neu+ breast cancer without increasing its metastatic potential. Oncotarget 2016, 7, 60793–60806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rappa, G.; Mercapide, J.; Lorico, A. Spontaneous formation of tumorigenic hybrids between breast cancer and multipotent stromal cells is a source of tumor heterogeneity. Am. J. Pathol. 2012, 180, 2504–2515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ding, J.; Jin, W.; Chen, C.; Shao, Z.; Wu, J. Tumor associated macrophage× cancer cell hybrids may acquire cancer stem cell properties in breast cancer. PLoS ONE 2012, 7, e41942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cao, M.F.; Chen, L.; Dang, W.Q.; Zhang, X.C.; Zhang, X.; Shi, Y.; Yao, X.H.; Li, Q.; Zhu, J.; Lin, Y.; et al. Hybrids by tumor-associated macrophages × glioblastoma cells entail nuclear reprogramming and glioblastoma invasion. Cancer Lett. 2019, 442, 445–452. [Google Scholar] [CrossRef]
- Joly, E.; Hudrisier, D. What is trogocytosis and what is its purpose? Nat. Immunol. 2003, 4, 815. [Google Scholar] [CrossRef]
- Xue, J.; Zhu, Y.; Sun, Z.; Ji, R.; Zhang, X.; Xu, W.; Yuan, X.; Zhang, B.; Yan, Y.; Yin, L.; et al. Tumorigenic hybrids between mesenchymal stem cells and gastric cancer cells enhanced cancer proliferation, migration and stemness. BMC Cancer 2015, 15, 793. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.N.; Huang, Y.H.; Zhao, L. Fusion of macrophages promotes breast cancer cell proliferation, migration and invasion through activating epithelial-mesenchymal transition and Wnt/β-catenin signaling pathway. Arch. Biochem. Biophys. 2019, 676, 108137. [Google Scholar] [CrossRef]
- Fan, H.; Lu, S. Fusion of human bone hemopoietic stem cell with esophageal carcinoma cells didn’t generate esophageal cancer stem cell. Neoplasma 2014, 61, 540–545. [Google Scholar] [CrossRef]
- Weichert, W.; Knösel, T.; Bellach, J.; Dietel, M.; Kristiansen, G. ALCAM/CD166 is overexpressed in colorectal carcinoma and correlates with shortened patient survival. J. Clin. Pathol. 2004, 57, 1160–1164. [Google Scholar] [CrossRef]
- Jayasinghe, C.; Simiantonaki, N.; Michel-Schmidt, R.; Kirkpatrick, C.J. Endothelial VEGFR-3 expression in colorectal carcinomas is associated with hematogenous metastasis. Oncol. Rep. 2009, 22, 1093–1100. [Google Scholar] [CrossRef] [Green Version]
- Planagumà, J.; Díaz-Fuertes, M.; Gil-Moreno, A.; Abal, M.; Monge, M.; García, A.; Baró, T.; Thomson, T.M.; Xercavins, J.; Alameda, F.; et al. A differential gene expression profile reveals overexpression of RUNX1/AML1 in invasive endometrioid carcinoma. Cancer Res. 2004, 64, 8846–8853. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lartigue, L.; Merle, C.; Lagarde, P.; Delespaul, L.; Lesluyes, T.; Le Guellec, S.; Pérot, G.; Leroy, L.; Coindre, J.M.; Chibon, F. Genome remodeling upon mesenchymal tumor cell fusion contributes to tumor progression and metastatic spread. Oncogene 2020, 39, 4198–4211. [Google Scholar] [CrossRef]
- Ogle, B.M.; Cascalho, M.; Platt, J.L. Biological implications of cell fusion. Nat. Rev. Mol. Cell Biol. 2005, 6, 567–575. [Google Scholar] [CrossRef]
- Yamanaka, S.; Blau, H.M. Nuclear reprogramming to a pluripotent state by three approaches. Nature 2010, 465, 704–712. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cowan, C.A.; Atienza, J.; Melton, D.A.; Eggan, K. Nuclear reprogramming of somatic cells after fusion with human embryonic stem cells. Science 2005, 309, 1369–1373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Blau, H.M.; Blakely, B.T. Plasticity of cell fate: Insights from heterokaryons. Semin. Cell Dev. Biol. 1999, 10, 267–272. [Google Scholar] [CrossRef]
- Blau, H.M.; Chiu, C.P.; Webster, C. Cytoplasmic activation of human nuclear genes in stable heterocaryons. Cell 1983, 32, 1171–1180. [Google Scholar] [CrossRef]
- Pomerantz, J.; Blau, H.M. Nuclear reprogramming: A key to stem cell function in regenerative medicine. Nat. Cell Biol. 2004, 6, 810–816. [Google Scholar] [CrossRef]
- Itokowa, T.; Zhu, M.L.; Troiano, N.; Bian, J.; Kawano, T.; Insogna, K. Osteoclasts lacking Rac2 have defective chemotaxis and resorptive activity. Calcif. Tissue Int. 2011, 88, 75–86. [Google Scholar] [CrossRef] [Green Version]
- Islam, M.Q.; Meirelles Lda, S.; Nardi, N.B.; Magnusson, P.; Islam, K. Polyethylene glycol-mediated fusion between primary mouse mesenchymal stem cells and mouse fibroblasts generates hybrid cells with increased proliferation and altered differentiation. Stem Cells Dev. 2006, 15, 905–919. [Google Scholar] [CrossRef]
- Goldenberg, D.M.; Zagzag, D.; Heselmeyer-Haddad, K.M.; Berroa Garcia, L.Y.; Ried, T.; Loo, M.; Chang, C.H.; Gold, D.V. Horizontal transmission and retention of malignancy, as well as functional human genes, after spontaneous fusion of human glioblastoma and hamster host cells in vivo. Int. J. Cancer 2012, 131, 49–58. [Google Scholar] [CrossRef] [PubMed]
- Goldenberg, D.M.; Gold, D.V.; Loo, M.; Liu, D.; Chang, C.H.; Jaffe, E.S. Horizontal transmission of malignancy: In-vivo fusion of human lymphomas with hamster stroma produces tumors retaining human genes and lymphoid pathology. PLoS ONE 2013, 8, e55324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yilmaz, Y.; Lazova, R.; Qumsiyeh, M.; Cooper, D.; Pawelek, J. Donor Y chromosome in renal carcinoma cells of a female BMT recipient: Visualization of putative BMT-tumor hybrids by FISH. Bone Marrow Transplant. 2005, 35, 1021–1024. [Google Scholar] [CrossRef] [Green Version]
- Xu, M.H.; Gao, X.; Luo, D.; Zhou, X.D.; Xiong, W.; Liu, G.X. EMT and acquisition of stem cell-like properties are involved in spontaneous formation of tumorigenic hybrids between lung cancer and bone marrow-derived mesenchymal stem cells. PLoS ONE 2014, 9, e87893. [Google Scholar] [CrossRef] [PubMed]
- Luo, F.; Liu, T.; Wang, J.; Li, J.; Ma, P.; Ding, H.; Feng, G.; Lin, D.; Xu, Y.; Yang, K. Bone marrow mesenchymal stem cells participate in prostate carcinogenesis and promote growth of prostate cancer by cell fusion in vivo. Oncotarget 2016, 7, 30924–30934. [Google Scholar] [CrossRef] [Green Version]
- Laberge, G.S.; Duvall, E.; Haedicke, K.; Pawelek, J. Leukocyte(-)Cancer Cell Fusion-Genesis of a Deadly Journey. Cells 2019, 8, 170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Toyoshima, K.; Hayashi, A.; Kashiwagi, M.; Hayashi, N.; Iwatsuki, M.; Ishimoto, T.; Baba, Y.; Baba, H.; Ohta, Y. Analysis of circulating tumor cells derived from advanced gastric cancer. Int. J. Cancer 2015, 137, 991–998. [Google Scholar] [CrossRef] [PubMed]
- Lustberg, M.B.; Balasubramanian, P.; Miller, B.; Garcia-Villa, A.; Deighan, C.; Wu, Y.; Carothers, S.; Berger, M.; Ramaswamy, B.; Macrae, E.R.; et al. Heterogeneous atypical cell populations are present in blood of metastatic breast cancer patients. Breast Cancer Res. BCR 2014, 16, R23. [Google Scholar] [CrossRef] [Green Version]
- de Wit, S.; Zeune, L.L.; Hiltermann, T.J.N.; Groen, H.J.M.; Dalum, G.V.; Terstappen, L. Classification of Cells in CTC-Enriched Samples by Advanced Image Analysis. Cancers 2018, 10, 377. [Google Scholar] [CrossRef] [Green Version]
- Nel, I.; Jehn, U.; Gauler, T.; Hoffmann, A.C. Individual profiling of circulating tumor cell composition in patients with non-small cell lung cancer receiving platinum based treatment. Transl. Lung Cancer Res. 2014, 3, 100–106. [Google Scholar] [CrossRef]
- Allan, A.L.; Vantyghem, S.A.; Tuck, A.B.; Chambers, A.F.; Chin-Yee, I.H.; Keeney, M. Detection and quantification of circulating tumor cells in mouse models of human breast cancer using immunomagnetic enrichment and multiparameter flow cytometry. Cytom. Part A J. Int. Soc. Anal. Cytol. 2005, 65, 4–14. [Google Scholar] [CrossRef] [PubMed]
- Liu, Q.; Liao, Q.; Zhao, Y. Myeloid-derived suppressor cells (MDSC) facilitate distant metastasis of malignancies by shielding circulating tumor cells (CTC) from immune surveillance. Med. Hypotheses 2016, 87, 34–39. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Meng, Q.H.; Noh, H.; Somaiah, N.; Torres, K.E.; Xia, X.; Batth, I.S.; Joseph, C.P.; Liu, M.; Wang, R.; et al. Cell-surface vimentin-positive macrophage-like circulating tumor cells as a novel biomarker of metastatic gastrointestinal stromal tumors. Oncoimmunology 2018, 7, e1420450. [Google Scholar] [CrossRef]
- Reduzzi, C.; Vismara, M.; Gerratana, L.; Silvestri, M.; De Braud, F.; Raspagliesi, F.; Verzoni, E.; Di Cosimo, S.; Locati, L.D.; Cristofanilli, M.; et al. The curious phenomenon of dual-positive circulating cells: Longtime overlooked tumor cells. Semin. Cancer Biol. 2020, 60, 344–350. [Google Scholar] [CrossRef]
- Sajay, B.N.; Chang, C.P.; Ahmad, H.; Khuntontong, P.; Wong, C.C.; Wang, Z.; Puiu, P.D.; Soo, R.; Rahman, A.R. Microfluidic platform for negative enrichment of circulating tumor cells. Biomed. Microdevices 2014, 16, 537–548. [Google Scholar] [CrossRef] [PubMed]
- Lustberg, M.; Jatana, K.R.; Zborowski, M.; Chalmers, J.J. Emerging technologies for CTC detection based on depletion of normal cells. Minimal Residual Dis. Circ. Tumor Cells Breast Cancer 2012, 195, 97–110. [Google Scholar] [CrossRef] [Green Version]
- Takao, M.; Takeda, K. Enumeration, characterization, and collection of intact circulating tumor cells by cross contamination-free flow cytometry. Cytom. Part A J. Int. Soc. Anal. Cytol. 2011, 79, 107–117. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Riethdorf, S.; Fritsche, H.; Muller, V.; Rau, T.; Schindlbeck, C.; Rack, B.; Janni, W.; Coith, C.; Beck, K.; Janicke, F.; et al. Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: A validation study of the CellSearch system. Clin. Cancer Res. An. Off. J. Am. Assoc. Cancer Res. 2007, 13, 920–928. [Google Scholar] [CrossRef] [Green Version]
- Aguirre, L.A.; Montalbán-Hernández, K.; Avendaño-Ortiz, J.; Marín, E.; Lozano, R.; Toledano, V.; Sánchez-Maroto, L.; Terrón, V.; Valentín, J.; Pulido, E.; et al. Tumor stem cells fuse with monocytes to form highly invasive tumor-hybrid cells. Oncoimmunology 2020, 9, 1773204. [Google Scholar] [CrossRef]
- Yan, Y.Y.; Guo, Q.R.; Wang, F.H.; Adhikari, R.; Zhu, Z.Y.; Zhang, H.Y.; Zhou, W.M.; Yu, H.; Li, J.Q.; Zhang, J.Y. Cell-Free DNA: Hope and Potential Application in Cancer. Front. Cell Dev. Biol. 2021, 9, 639233. [Google Scholar] [CrossRef] [PubMed]
- Tie, J.; Wang, Y.; Cohen, J.; Li, L.; Hong, W.; Christie, M.; Wong, H.L.; Kosmider, S.; Wong, R.; Thomson, B.; et al. Circulating tumor DNA dynamics and recurrence risk in patients undergoing curative intent resection of colorectal cancer liver metastases: A prospective cohort study. PLoS Med. 2021, 18, e1003620. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Sutton, T.L.; Patel, R.K.; Anderson, A.N.; Bowden, S.G.; Whalen, R.; Giske, N.R.; Wong, M.H. Circulating Cells with Macrophage-like Characteristics in Cancer: The Importance of Circulating Neoplastic-Immune Hybrid Cells in Cancer. Cancers 2022, 14, 3871. https://doi.org/10.3390/cancers14163871
Sutton TL, Patel RK, Anderson AN, Bowden SG, Whalen R, Giske NR, Wong MH. Circulating Cells with Macrophage-like Characteristics in Cancer: The Importance of Circulating Neoplastic-Immune Hybrid Cells in Cancer. Cancers. 2022; 14(16):3871. https://doi.org/10.3390/cancers14163871
Chicago/Turabian StyleSutton, Thomas L., Ranish K. Patel, Ashley N. Anderson, Stephen G. Bowden, Riley Whalen, Nicole R. Giske, and Melissa H. Wong. 2022. "Circulating Cells with Macrophage-like Characteristics in Cancer: The Importance of Circulating Neoplastic-Immune Hybrid Cells in Cancer" Cancers 14, no. 16: 3871. https://doi.org/10.3390/cancers14163871
APA StyleSutton, T. L., Patel, R. K., Anderson, A. N., Bowden, S. G., Whalen, R., Giske, N. R., & Wong, M. H. (2022). Circulating Cells with Macrophage-like Characteristics in Cancer: The Importance of Circulating Neoplastic-Immune Hybrid Cells in Cancer. Cancers, 14(16), 3871. https://doi.org/10.3390/cancers14163871