The Metastatic Cascade as the Basis for Liquid Biopsy Development

Zahra Eslami-S, Luis Enrique Cortés-Hernández, Catherine Alix-Panabières, Zahra Eslami-S, Luis Enrique Cortés-Hernández, Catherine Alix-Panabières

Abstract

The metastatic cascade describes the process whereby aggressive cancer cells leave the primary tumor, travel through the bloodstream, and eventually reach distant organs to develop one or several metastases. During the last decade, innovative technologies have exploited the recent biological knowledge to identify new circulating biomarkers for the screening and early detection of cancer, real-time monitoring of treatment response, assessment of tumor relapse risk (prognosis), identification of new therapeutic targets and resistance mechanisms, patient stratification, and therapeutic decision-making. These techniques are broadly described using the term of Liquid Biopsy. This field is in constant progression and is based on the detection of circulating tumor cells, circulating free nucleic acids (e.g., circulating tumor DNA), circulating tumor-derived extracellular vesicles, and tumor-educated platelets. The aim of this review is to describe the biological principles underlying the liquid biopsy concept and to discuss how functional studies can expand the clinical applications of these circulating biomarkers.

Keywords: circulating tumor cells; circulating-tumor DNA; extracellular vesicles; liquid biopsy; metastatic cascade; tumor educated platelets.

Copyright © 2020 Eslami-S, Cortés-Hernández and Alix-Panabières.

Figures

Figure 1
Figure 1
(A) Advantages of liquid biopsy vs. tissue biopsy during the metastatic cascade. (A) Compared with tissue biopsy (limitation for serial sampling and difficulty to access certain organs, such as brain and lung), liquid biopsy allows the real-time monitoring of cancer progression during the liquid phase of the metastatic cascade through the detection of different circulating analytes. These circulating analytes have specific biological functions and provide complementary information that can be continuously evaluated during cancer progression. (B) Comparison of tissue and liquid biopsies. Comparison of the relevant medical features of tissue biopsy and liquid biopsy.
Figure 2
Figure 2
Liquid biopsy and the metastatic cascade. During the metastatic cascade, different analytes are involved: (A) Primary tumor: tumor growth at the primary site. (B) Cancer cell proliferation: release of DNA from necrotic/apoptotic cells when due to the continuous cancer cell proliferation, passive diffusion of oxygen and nutrients is no longer enough to cover the cell requirement. (C) Angiogenesis: EVs are involved in neo-angiogenesis. (D) Local invasion, detachment, and intravasation: Cancer cells acquire significant changes to survive the physical interactions and mechanical forces in blood and tissues. Tumor cells also undergo epithelial-to-mesenchymal transition (EMT) to move within the connective tissue and then secrete exosomes that contribute to induce endothelial permeability for intravasation through endothelial cells. (E) Embolization and survival: CTCs can survive and adapt in the blood by forming CTC clusters/micro-emboli that display higher metastatic potential by increasing cancer cell survival and reducing apoptosis. Clustering with neutrophils can also promote cell-cycle progression and survival in CTCs, further promoting metastatic development. CTCs also gain physical and immune protection by interacting with platelets. (F) Arrest and extravasation in the target organ: CTCs interact with endothelial cells, mainly in capillaries where they become trapped. CTCs and platelets might release EVs that can modify the cytoskeleton of endothelial cells and increase the blood vessel permeability, thus allowing CTC extravasation. (G) Micro-metastases: CTCs need a favorable niche. Before CTC arrival, this niche is called pre-metastatic niche, and exosomes play a major role in its preparation. (H) Metastases: CTCs that arrive in distant organs, such as liver or lung, will form a metastatic lesion, thus concluding the metastatic cascade.
Figure 3
Figure 3
Advantages of liquid biopsy to monitor tumor progression. Liquid biopsy has broad potential applications for cancer diagnosis and treatment, including screening for early diagnosis, study of tumor heterogeneity, and clonal evolution, and detection of minimal residual disease. During cancer progression, liquid biopsy might help clinicians to stratify patients for treatment personalization and to monitor the treatment response and the development of resistance. It can also be used for the tumor molecular characterization, and its minimally invasive nature allows repeat sampling to monitor changes over time without the need for a tissue biopsy.

References

    1. Paget S. The distribution of secondary growths in cancer of the breast. Lancet. (1889) 133:571–3. 10.1016/S0140-6736(00)49915-0
    1. Fidler IJ. Metastasis: quantitative analysis of distribution and fate of tumor emboli labeled with 125 I-5-iodo-2'-deoxyuridine. J Natl Cancer Inst. (1970) 45:773–82.
    1. Chambers AF, Naumov GN, Vantyghem SA, Tuck AB. Molecular biology of breast cancer metastasis. Clinical implications of experimental studies on metastatic inefficiency. Breast Cancer Res. (2000) 2:400–7. 10.1186/bcr86
    1. Fidler IJ. The pathogenesis of cancer metastasis: the “seed and soil” hypothesis revisited. Nat Rev Cancer. (2003) 3:453–8. 10.1038/nrc1098
    1. Poste G, Fidler IJ. The pathogenesis of cancer metastasis. Nature. (1980) 283:139–46. 10.1038/283139a0
    1. Lambert AW, Pattabiraman DR, Weinberg RA. Emerging biological principles of metastasis. Cell. (2017) 168:670–91. 10.1016/j.cell.2016.11.037
    1. Pantel K, Alix-Panabières C. Liquid biopsy and minimal residual disease — latest advances and implications for cure. Nat Rev Clin Oncol. (2019) 16:409–24. 10.1038/s41571-019-0187-3
    1. Eslami-S Z, Cortes-Hernandez LE, Cayrefourcq L, Alix-Panabieres C. The different facets of liquid biopsy: a Kaleidoscopic view. Cold Spring Harb Perspect Med. (2019) 10:a037333. 10.1101/cshperspect.a037333
    1. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LAJ, Kinzler KW. Cancer genome landscapes. Science. (2013) 339:1546–58. 10.1126/science.1235122
    1. Campbell P, Getz G, Korbel J, Joshua M, Jennings J, Stein L, et al. Pan-cancer analysis of whole genomes. Nature. (2020) 578:82–93. 10.1038/s41586-020-1969-6
    1. Al Tameemi W, Dale TP, Al-Jumaily RMK, Forsyth NR. Hypoxia-modified cancer cell metabolism. Front cell Dev Biol. (2019) 7:4. 10.3389/fcell.2019.00004
    1. Merker JD, Oxnard GR, Compton C, Diehn M, Hurley P, Lazar AJ, et al. Circulating tumor DNA analysis in patients with cancer: American society of clinical oncology and college of American pathologists joint review. J Clin Oncol. (2018) 36:1631–41. 10.1200/JCO.2017.76.8671
    1. Lo YMD, Chan KCA, Sun H, Chen EZ, Jiang P, Lun FMF, et al. . Maternal plasma DNA sequencing reveals the genome-wide genetic and mutational profile of the fetus. Sci Transl Med. (2010) 2:61ra91 LP. 10.1126/scitranslmed.3001720
    1. Thierry AR, Mouliere F, Gongora C, Ollier J, Robert B, Ychou M, et al. . Origin and quantification of circulating DNA in mice with human colorectal cancer xenografts. Nucleic Acids Res. (2010) 38:6159–75. 10.1093/nar/gkq421
    1. Kustanovich A, Schwartz R, Peretz T, Grinshpun A. Life and death of circulating cell-free DNA. Cancer Biol Ther. (2019) 20:1057–67. 10.1080/15384047.2019.1598759
    1. Lampignano R, Neumann MHD, Weber S, Kloten V, Herdean A, Voss T, et al. . Multicenter evaluation of circulating cell-free DNA extraction and downstream analyses for the development of standardized (pre)analytical work flows. Clin Chem. (2019) 66:clinchem.2019.306837. 10.1373/clinchem.2019.306837
    1. Jeppesen DK, Fenix AM, Franklin JL, Higginbotham JN, Zhang Q, Zimmerman LJ, et al. . Reassessment of exosome composition. Cell. (2019) 177:428–45.e18. 10.1016/j.cell.2019.02.029
    1. Sun K, Jiang P, Chan KCA, Wong J, Cheng YKY, Liang RHS, et al. . Plasma DNA tissue mapping by genome-wide methylation sequencing for noninvasive prenatal, cancer, and transplantation assessments. Proc Natl Acad Sci USA. (2015) 112:E5503–12. 10.1073/pnas.1508736112
    1. Razavi P, Li BT, Brown DN, Jung B, Hubbell E, Shen R, et al. . High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. Nat Med. (2019) 25:1928–37. 10.1038/s41591-019-0652-7
    1. Martincorena I, Fowler JC, Wabik A, Lawson ARJ, Abascal F, Hall MWJ, et al. . Somatic mutant clones colonize the human esophagus with age. Science. (2018) 362:911–7. 10.1126/science.aau3879
    1. Mattox AK, Bettegowda C, Zhou S, Papadopoulos N, Kinzler KW, Vogelstein B. Applications of liquid biopsies for cancer. Sci Transl Med. (2019) 11:eaay1984. 10.1126/scitranslmed.aay1984
    1. Chabon JJ, Hamilton EG, Kurtz DM, Esfahani MS, Moding EJ, Stehr H, et al. . Integrating genomic features for non-invasive early lung cancer detection. Nature. (2020) 580:245–51. 10.1038/s41586-020-2140-0
    1. Gavelli G, Giampalma E. Sensitivity and specificity of chest X-ray screening for lung cancer: review article. Cancer. (2000) 89(Suppl. 11):2453–6. 10.1002/1097-01422000120189:112453::;2-d
    1. Leal A, van Grieken NCT, Palsgrove DN, Phallen J, Medina JE, Hruban C, et al. . White blood cell and cell-free DNA analyses for detection of residual disease in gastric cancer. Nat Commun. (2020) 11:525. 10.1038/s41467-020-14310-3
    1. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. (2020) 367:eaau6977. 10.1126/science.aau6977
    1. Thakur BK, Zhang H, Becker A, Matei I, Huang Y, Costa-Silva B, et al. . Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res. (2014) 24:766–9. 10.1038/cr.2014.44
    1. Fernando MR, Jiang C, Krzyzanowski GD, Ryan WL. New evidence that a large proportion of human blood plasma cell-free DNA is localized in exosomes. PLoS ONE. (2017) 12:e0183915. 10.1371/journal.pone.0183915
    1. Vagner T, Spinelli C, Minciacchi VR, Balaj L, Zandian M, Conley A, et al. . Large extracellular vesicles carry most of the tumour DNA circulating in prostate cancer patient plasma. J Extracell Vesicles. (2018) 7:1505403. 10.1080/20013078.2018.1505403
    1. Thery C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, et al. . Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International society for extracellular vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. (2018) 7:1535750. 10.1080/20013078.2018.1461450
    1. Cohen JD, Li L, Wang Y, Thoburn C, Afsari B, Danilova L, et al. . Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. (2018) 359:926–30. 10.1126/science.aar3247
    1. Xu J, Zhang X, Pelayo R, Monestier M, Ammollo CT, Semeraro F, et al. . Extracellular histones are major mediators of death in sepsis. Nat Med. (2009) 15:1318–21. 10.1038/nm.2053
    1. Nishimoto S, Fukuda D, Higashikuni Y, Tanaka K, Hirata Y, Murata C, et al. . Obesity-induced DNA released from adipocytes stimulates chronic adipose tissue inflammation and insulin resistance. Sci Adv. (2016) 2:e1501332. 10.1126/sciadv.1501332
    1. Hsu YL, Hung JY, Chang WA, Lin YS, Pan YC, Tsai PH, et al. . Hypoxic lung cancer-secreted exosomal miR-23a increased angiogenesis and vascular permeability by targeting prolyl hydroxylase and tight junction protein ZO-1. Oncogene. (2017) 36:4929–42. 10.1038/onc.2017.105
    1. Schillaci O, Fontana S, Monteleone F, Taverna S, Di Bella MA, Di Vizio D, et al. . Exosomes from metastatic cancer cells transfer amoeboid phenotype to non-metastatic cells and increase endothelial permeability: their emerging role in tumor heterogeneity. Sci Rep. (2017) 7:4711. 10.1038/s41598-017-05002-y
    1. Amit M, Takahashi H, Dragomir MP, Lindemann A, Gleber-Netto FO, Pickering CR, et al. . Loss of p53 drives neuron reprogramming in head and neck cancer. Nature. (2020) 578:449–54. 10.1038/s41586-020-1996-3
    1. Maru Y. Premetastasis. Cold Spring Harb Perspect Med. (2019) a036897. [Epub ahead of print]. 10.1101/cshperspect.a036897.
    1. Holdenrieder S, Pagliaro L, Morgenstern D, Dayyani F. Clinically meaningful use of blood tumor markers in oncology. Biomed Res Int. (2016) 2016:9795269. 10.1155/2016/9795269
    1. Jiang T, Shi T, Zhang H, Hu J, Song Y, Wei J, et al. . Tumor neoantigens: from basic research to clinical applications. J Hematol Oncol. (2019) 12:93. 10.1186/s13045-019-0787-5
    1. Hofman P, Heeke S, Alix-Panabières C, Pantel K. Liquid biopsy in the era of immuno-oncology: is it ready for prime-time use for cancer patients? Ann Oncol. (2019) 30:1448–59. 10.1093/annonc/mdz196
    1. Gros A, Parkhurst MR, Tran E, Pasetto A, Robbins PF, Ilyas S, et al. . Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat Med. (2016) 22:433–8. 10.1038/nm.4051
    1. Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat Rev Mol Cell Biol. (2019) 20:69–84. 10.1038/s41580-018-0080-4
    1. Wirtz D, Konstantopoulos K, Searson PC. The physics of cancer: the role of physical interactions and mechanical forces in metastasis. Nat Rev Cancer. (2011) 11:512–22. 10.1038/nrc3080
    1. Gilmore AP. Anoikis. Cell Death Differ. (2005) 12:1473–7. 10.1038/sj.cdd.4401723
    1. Hu Z, Ding J, Ma Z, Sun R, Seoane JA, Scott Shaffer J, et al. . Quantitative evidence for early metastatic seeding in colorectal cancer. Nat Genet. (2019) 51:1113–22. 10.1038/s41588-019-0423-x
    1. Bidard F-C, Peeters DJ, Fehm T, Nolé F, Gisbert-Criado R, Mavroudis D, et al. . Clinical validity of circulating tumour cells in patients with metastatic breast cancer: a pooled analysis of individual patient data. Lancet Oncol. (2014) 15:406–14. 10.1016/S1470-2045(14)70069-5
    1. Mazel M, Jacot W, Pantel K, Bartkowiak K, Topart D, Cayrefourcq L, et al. . Frequent expression of PD-L1 on circulating breast cancer cells. Mol Oncol. (2015) 9:1773–82. 10.1016/j.molonc.2015.05.009
    1. Riethdorf S, Müller V, Zhang L, Rau T, Loibl S, Komor M, et al. . Detection and HER2 expression of circulating tumor cells: prospective monitoring in breast cancer patients treated in the neoadjuvant GeparQuattro trial. Clin Cancer Res. (2010) 16:2634–45. 10.1158/1078-0432.CCR-09-2042
    1. Scher HI, Lu D, Schreiber NA, Louw J, Graf RP, Vargas HA, et al. . Association of AR-V7 on circulating tumor cells as a treatment-specific biomarker with outcomes and survival in castration-resistant prostate Cancer. JAMA Oncol. (2016) 2:1441–9. 10.1001/jamaoncol.2016.1828
    1. Eslami-S Z, Cortés-Hernández LE, Alix-Panabières C. Circulating tumor cells: moving forward into clinical applications. Precis Cancer Med March. (2020) 3:4 10.21037/pcm.2019.11.07
    1. Cortés-Hernández LE, Eslami-SZ, Pantel K, Alix-Panabières C. Molecular and functional characterization of circulating tumor cells: from discovery to clinical application. Clin Chem. (2019) 66:303586. 10.1373/clinchem.2019.303586
    1. Denève E, Riethdorf S, Ramos J, Nocca D, Coffy A, Daurès JP, et al. . Capture of viable circulating tumor cells in the liver of colorectal cancer patients. Clin Chem. (2013) 59:1384–92. 10.1373/clinchem.2013.202846
    1. Aceto N, Bardia A, Miyamoto DT, Donaldson MC, Wittner BS, Spencer JA, et al. . Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell. (2014) 158:1110–22. 10.1016/j.cell.2014.07.013
    1. Gkountela S, Castro-Giner F, Szczerba BM, Vetter M, Landin J, Scherrer R, et al. . Circulating tumor cell clustering shapes DNA methylation to enable metastasis seeding. Cell. (2019) 176:98–112.e14. 10.1016/j.cell.2018.11.046
    1. Szczerba BM, Castro-Giner F, Vetter M, Krol I, Gkountela S, Landin J, et al. . Neutrophils escort circulating tumour cells to enable cell cycle progression. Nature. (2019) 566:553–7.10.1038/s41586-019-0915-y
    1. Hong SM, Jung D, Kiemen A, Gaida MM, Yoshizawa T, Braxton AM, et al. Three-dimensional visualization of cleared human pancreas cancer reveals that sustained epithelial-to-mesenchymal transition is not required for venous invasion. Mod Pathol. (2019) 33:639–47. 10.1038/s41379-019-0409-3
    1. Padmanaban V, Krol I, Suhail Y, Szczerba BM, Aceto N, Bader JS, et al. . E-cadherin is required for metastasis in multiple models of breast cancer. Nature. (2019) 573:439–44. 10.1038/s41586-019-1526-3
    1. Cabel L, Proudhon C, Gortais H, Loirat D, Coussy F, Pierga JY, et al. . Circulating tumor cells: clinical validity and utility. Int J Clin Oncol. (2017) 22:421–30. 10.1007/s10147-017-1105-2
    1. Tissot T, Massol F, Ujvari B, Alix-Panabieres C, Loeuille N, Thomas F. Metastasis and the evolution of dispersal. Proc Biol Sci. (2019) 286:20192186. 10.1098/rspb.2019.2186
    1. Jiang X, Wong KHK, Khankhel AH, Zeinali M, Reategui E, Phillips MJ, et al. . Microfluidic isolation of platelet-covered circulating tumor cells. Lab Chip. (2017) 17:3498–503. 10.1039/C7LC00654C
    1. Gasic GJ, Gasic TB, Galanti N, Johnson T, Murphy S. Platelet-tumor-cell interactions in mice. The role of platelets in the spread of malignant disease. Int J cancer. (1973) 11:704–18. 10.1002/ijc.2910110322
    1. Best MG, Sol N, Kooi I, Tannous J, Westerman BA, Rustenburg F, et al. . RNA-Seq of tumor-educated platelets enables blood-based pan-cancer, multiclass, and molecular pathway cancer diagnostics. Cancer Cell. (2015) 28:666–76. 10.1016/j.ccell.2015.09.018
    1. Kuznetsov HS, Marsh T, Markens BA, Castano Z, Greene-Colozzi A, Hay SA, et al. . Identification of luminal breast cancers that establish a tumor-supportive macroenvironment defined by proangiogenic platelets and bone marrow-derived cells. Cancer Discov. (2012) 2:1150–65. 10.1158/-12-0216
    1. Heeke S, Mograbi B, Alix-Panabières C, Hofman P. Never travel alone: the crosstalk of circulating tumor cells and the blood microenvironment. Cells. (2019) 8:714. 10.3390/cells8070714
    1. Menter DG, Tucker SC, Kopetz S, Sood AK, Crissman JD, Honn K V. Platelets and cancer: a casual or causal relationship: revisited. Cancer Metastasis Rev. (2014) 33:231–69. 10.1007/s10555-014-9498-0
    1. McAllister SS, Weinberg RA. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat Cell Biol. (2014) 16:717–27. 10.1038/ncb3015
    1. Hou JM, Krebs MG, Lancashire L, Sloane R, Backen A, Swain RK, et al. . Clinical significance and molecular characteristics of circulating tumor cells and circulating tumor microemboli in patients with small-cell lung cancer. J Clin Oncol. (2012) 30:525–32. 10.1200/JCO.2010.33.3716
    1. Giuliano M, Shaikh A, Lo HC, Arpino G, De Placido S, Zhang XH, et al. . Perspective on circulating tumor cell clusters: why it takes a village to metastasize. Cancer Res. (2018) 78:845–52. 10.1158/0008-5472.CAN-17-2748
    1. Gay LJ, Felding-Habermann B. Contribution of platelets to tumour metastasis. Nat Rev Cancer. (2011) 11:123–34. 10.1038/nrc3004
    1. Wang Y, Sun Y, Li D, Zhang L, Wang K, Zuo Y, et al. . Platelet P2Y12 is involved in murine pulmonary metastasis. PLoS ONE. (2013) 8:e80780. 10.1371/journal.pone.0080780
    1. Orellana R, Kato S, Erices R, Bravo ML, Gonzalez P, Oliva B, et al. . Platelets enhance tissue factor protein and metastasis initiating cell markers, and act as chemoattractants increasing the migration of ovarian cancer cells. BMC Cancer. (2015) 15:290. 10.1186/s12885-015-1304-z
    1. Labelle M, Begum S, Hynes RO. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell. (2011) 20:576–90. 10.1016/j.ccr.2011.09.009
    1. Xiong G, Chen J, Zhang G, Wang S, Kawasaki K, Zhu J, et al. . Hsp47 promotes cancer metastasis by enhancing collagen-dependent cancer cell-platelet interaction. Proc Natl Acad Sci USA. (2020) 117:3748–58. 10.1073/pnas.1911951117
    1. Zhu J, Xiong G, Fu H, Evers BM, Zhou BP, Xu R. Chaperone Hsp47 drives malignant growth and invasion by modulating an ECM gene network. Cancer Res. (2015) 75:1580–91. 10.1158/0008-5472.CAN-14-1027
    1. Osmani N, Follain G, García León MJ, Lefebvre O, Busnelli I, Larnicol A, et al. . Metastatic tumor cells exploit their adhesion repertoire to counteract shear forces during intravascular arrest. Cell Rep. (2019) 28:2491–500.e5. 10.1016/j.celrep.2019.07.102
    1. Follain G, Osmani N, Azevedo AS, Allio G, Mercier L, Karreman MA, et al. . Hemodynamic forces tune the arrest, adhesion, and extravasation of circulating tumor cells. Dev Cell. (2018) 45:33–52.e12. 10.1016/j.devcel.2018.02.015
    1. Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. (2012) 18:883–91. 10.1038/nm.2753
    1. Ward Y, Lake R, Faraji F, Sperger J, Martin P, Gilliard C, et al. . Platelets promote metastasis via binding tumor CD97 leading to bidirectional signaling that coordinates transendothelial migration. Cell Rep. (2018) 23:808–22. 10.1016/j.celrep.2018.03.092
    1. Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, et al. . Tumour exosome integrins determine organotropic metastasis. Nature. (2015) 527:329–35. 10.1038/nature15756
    1. Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, et al. . Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. (2015) 17:816–26. 10.1038/ncb3169
    1. Rodrigues G, Hoshino A, Kenific CM, Matei IR, Steiner L, Freitas D, et al. . Tumour exosomal CEMIP protein promotes cancer cell colonization in brain metastasis. Nat Cell Biol. (2019) 21:1403–12. 10.1038/s41556-019-0404-4
    1. Agnoletto C, Corrà F, Minotti L, Baldassari F, Crudele F, Cook JW, et al. . Heterogeneity in circulating tumor cells: the relevance of the stem-cell subset. Cancers. (2019) 11:483. 10.3390/cancers11040483
    1. Zhang L, Ridgway LD, Wetzel MD, Ngo J, Yin W, Kumar D, et al. . The identification and characterization of breast cancer CTCs competent for brain metastasis. Sci Transl Med. (2013) 5:180ra48. 10.1126/scitranslmed.3005109

Source: PubMed

3
Předplatit