Preview

Известия Национальной академии наук Беларуси. Серия биологических наук

Расширенный поиск

Циркулирующие опухолевые клетки и циркулирующие раковые стволовые клетки и их детекция методом проточной цитофлуориметрии

https://doi.org/10.29235/1029-8940-2021-66-3-370-384

Аннотация

В обзоре приведено описание циркулирующих раковых стволовых клеток (цРСК) и циркулирующих опухолевых клеток (ЦОК) в крови человека и методов их определения. цРСК являются одними из главных инициаторов рецидива онкологических заболеваний, что делает их основной мишенью для разработки новых методов лечения. ЦОК являются относительно новыми биомаркерами для ранней диагностики метастазирования, мониторинг содержания которых дает ценную информацию на всех этапах ведения онкопациентов, включая раннюю диагностику заболевания, оценку риска рецидива болезни, определение эффективности химиотерапии с возможностью последующей ее коррекции.

Об авторах

Т. А. Позняк
Институт биофизики и клеточной инженерии НАН Беларуси
Беларусь

Позняк Татьяна Анатольевна - кандидат биологических наук, старший научный сотрудник.

ул. Академическая, 27, 220072, Минск.



А. Е. Гончаров
Институт биофизики и клеточной инженерии НАН Беларуси
Беларусь

Гончаров Андрей Евгеньевич - кандидат медицинских наук, доцент, директор.

ул. Академическая, 27, 220072, Минск.



В. М. Абашкин
Институт биофизики и клеточной инженерии НАН Беларуси
Беларусь

Абашкин Виктор Михайлович - младший научный сотрудник.

ул. Академическая, 27, 220072, Минск.



А. И. Становая
Институт биофизики и клеточной инженерии НАН Беларуси
Беларусь

Становая Алеся Игоревна - младший научный сотрудник.

ул. Академическая, 27, 220072, Минск.



А. В. Прохоров
Белорусский государственный медицинский университет
Беларусь

Прохоров Александр Викторович - доктор медицинских наук, профессор, заведующий кафедрой.

пр. Дзержинского, 83, 220116, Минск.



Д. Г. Щербин
Институт биофизики и клеточной инженерии НАН Беларуси
Беларусь

Щербин Дмитрий Григорьевич - доктор биологических наук, доцент, заведующий лабораторией.

ул. Академическая, 27, 220072, Минск.



Список литературы

1. Рак (онкологические заболевания) [Electronic resource] // Zdrav. Expert. - Mode of access: http://zdrav.expert/index.php/Ctat'ya:Rak_(onkologicheckiye_zabolevaniya). - Date of access: 14.03.2021.

2. Randomized trial of irinotecan and cetuximab with or without vemurafenib in BRAF-mutant metastatic colorectal cancer (SWOG 1406) / S. Kopetz [et al.] // J. Clin. Oncol. - 2017. - Vol. 35, N 4. - P. 285-294. https://doi.org/10.1200/jco.20.01994

3. Cancer stem cells, a fuzzy evolving concept: a cell population or a cell property? / А. Antoniou [et al.] // Cell Cycle. -2013. - Vol. 12, N 24. - P. 3743-3748. https://doi.org/10.4161/cc.27305

4. Understanding the cancer stem cell / S. Bomken [et al.] // Br. J. Cancer. - 2010. - Vol. 103, N 4. - P. 439-445. https://doi.org/10.1038/sj.bjc.6605821

5. Bonnet, D. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell / D. Bonnet, J. E. Dick // Nat. Med. - 1997. - Vol. 3, N 7. - P. 730-737. https://doi.org/10.1038/nm0797-730

6. Eun, K. Cancer stem cell heterogeneity: origin and new perspectives on CSC targeting / K. Eun, S. W. Ham, H. Kim // BMB Rep. - 2017. - Vol. 50, N 3. - P. 117-125. https://doi.org/10.5483/bmbrep.2017.50.3.222

7. Lang, F. Stem Cell Hierarchy and Clonal Evolution in Acute Lymphoblastic Leukemia / F. Lang, B. Wojcik, M. A. Rieger // Stem Cells Int. - 2015. - Vol. 2015. - P. 1-13. https://doi.org/10.1155/2015/137164

8. Visvader, J. E. Cancer stem cells: current status and evolving complexities / J. E. Visvader, G. J. Lindeman // Cell Stem Cell. - 2012. - Vol. 10, N 6. - P. 717-728. https://doi.org/10.1016/j.stem.2012.05.007

9. Mani S. A. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell, 2008, vol. 133, pp. 704-715. htpps://doi.org/10.1016/j.cell.2008.03.027

10. Hollier, B. G. The epithelial-to-mesenchymal transition and cancer stem cells: a coalition against cancer therapies / B. G. Hollier, K. Evans, S. A. Mani // J. Mammary Gland Biol. Neoplasia. - 2009. - Vol. 14, N 1. - P. 29-43. https://doi.org/10.1007/s10911-009-9110-3

11. Молекулярные маркеры раковых стволовых клеток, верифицированные in vivo / Я. С. Ким [и др.] // Биомед. химия. - 2016. - Т. 62, № 3. - C. 228-238.

12. Therapeutic strategies targeting cancer stem cells and their microenvironment / H. R. Sun [et al.] // Front. Oncol. -2019. - Vol. 9, N 9. - P. 1104. https://doi.org/10.3389/fonc.2019.01104

13. Fillmore, C. M. Human breast cancer cell lines contain stem-like cells that selfrenew, give rise to phenotypically diverse progeny and survive chemotherapy / C. M. Fillmore, C. Kuperwasser // Breast Cancer Res. - 2008. - Vol. 10, N 2. -Art. R25. https://doi.org/10.1186/bcr1982

14. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome / C. Ginestier [et al.] // Cell Stem Cell. - 2007. - Vol. 1, N 5. - P. 555-567. https://doi.org/10.1016/j.stem.2007.08.014

15. Begicevic, R. R. ABC transporters in cancer stem cells: beyond chemoresistance / R. R Begicevic, M. Falasca // Int. J. Mol. Sci. - 2017. - Vol. 18. - P. 2362. https://doi.org/10.3390/ijms18112362

16. Hanahan D. Hallmarks of cancer: the next generation / D. Hanahan, R. A. Weinberg // Cell. - 2011. - Vol. 144, N 5. -P. 646-674. https://doi.org/10.1016/j.cell.2011.02.013

17. Książkiewicz, M. Epithelial-mesenchymal transition: a hallmark in metastasis formation linking circulating tumor cells and cancer stem cells / M. Książkiewicz, A. Markiewicz, A. J. Zaczek // Pathobiology. - 2012. - Vol. 79, N 4. -P. 195-208. https://doi.org/10.1159/000337106

18. Epithelial-to-mesenchymal transitions and circulating tumor cells / A. Bonnomet [et al.] // J. Mammary Gland Biol. Neoplasia. - 2010. - Vol. 15, N 2. - P. 261-273. https://doi.org/10.1007/s10911-010-9174-0

19. Kalluri, R. EMT: when epithelial cells decide to become mesenchymal-like cells / R. Kalluri // J. Clin. Invest. - 2009. -Vol. 119, N 6. - P. 1417-1419. https://doi.org/10.1172/jci39675

20. Kalluri, R. The basics of epithelial-mesenchymal transition / R. Kalluri, R. A. Weinberg // J. Clin. Invest. - 2009. -Vol. 119, N 6. - P. 1420-1428. https://doi.org/10.1172/jci39104

21. Chaffer, C. L. Mesenchymal to epithelial transition in development and disease / C. L. Chaffer, E. W. Thompson, E. D. Williams // Cells Tissues Organs. - 2007. - Vol. 185, N 1-3. - P. 7-19. https://doi.org/10.1159/000101298

22. Chao, Y. L. Breast carcinoma cells re-express E-cadherin during mesenchymal to epithelial reverting transition / Y. L. Chao, C. R. Shepard, A. Wells // Mol. Cancer. - 2010. - Vol. 9, N 1. - Art. 179. https://doi.org/10.1186/1476-4598-9-179

23. Klymkowsky, M. W. Epithelialmesenchymal transition: a cancer researcher's conceptual friend and foe / M. W. Klymkowsky, P. Savagner // Am. J. Pathol. - 2009. - Vol. 174, N 5. - P. 1588-1593. https://doi.org/10.2353/ajpath.2009.080545

24. Tsuji, T. Epithelial-mesenchymal transition and cell cooperativity in metastasis / T. Tsuji, S. Ibaragi, G. F. Hu // Cancer Res. - 2009. - Vol. 69, N 18. - P. 7135-7139. https://doi.org/10.1158/0008-5472.can-09-1618

25. Hall, C. Circulating tumor cells in breast cancer patients / C. Hall, L. Valad, A. Lucci // Crit. Rev. Oncog. - 2016. -Vol. 21, N 1-2. - P. 125-139. https://doi.org/10.1615/critrevoncog.2016016120

26. Циркулирующие опухолевые клетки (ЦОК) при раке молочной железы: прогностическая значимость и методы выделения / Д. А. Зубцов [и др.] // Тр. МФТИ. - 2012. - Т. 4, № 3. - С. 18-26.

27. Circulating tumour cells as prognostic markers in progressive, castration-resistant prostate cancer: a reanalysis of IMMC38 trial data / H. I. Scher // Lancet Oncol. - 2009. - Vol. 10, N 3. - P. 233-239. https://doi.org/10.1016/s1470-2045(08)70340-1

28. Bidard. F. C. Circulating tumor cells in breast cancer / F. C. Bidard, C. Proudhon, J. Y. Pierga // Mol. Oncol. - 2016. -Vol. 10, N 3. - P 418-430. https://doi.org/10.1016Zj.molonc.2016.01.001

29. Multistep nature of metastatic inefficiency: dormancy of solitary cells after successful extravasation and limited survival of early micrometastases / K. J. Luzzi [et al.] // Am. J. Pathol. - 1998. - Vol. 153, N 3. - P. 865-873. https://doi.org/10.1016/s0002-9440(10)65628-3

30. Increased expression of apoptosis inhibitor protein XIAP contributes to anoikis resistance of circulating human prostate cancer metastasis precursor cells / O. Berezovskaya [et al.] // Cancer Res. - 2005. - Vol. 65, N 6. - P. 2378-2386. https://doi.org/10.1158/0008-5472.can-04-2649

31. Gray, J. W. / Evidence emerges for early metastasis and parallel evolution of primary and metastatic tumors / J. W. Gray // Cancer Cell. - 2003. - Vol. 4, N 1. - P. 4-6. https://doi.org/10.1016/s1535-6108(03)00167-3

32. Spread of human cancer cells occurs with probabilities indicative of a nongenetic mechanism / J. S. Michaelson [et al.] // Br. J. Cancer. - 2005. - Vol. 93, N 11. - P. 1244-1249. https://doi.org/10.1038/sj.bjc.6602848

33. Methods for isolating circulating epithelial cells and criteria for their classification as carcinoma cells / T. Fehm [et al.] // Cytotherapy. - 2005. - Vol. 7, N 2. - P. 171-185. https://doi.org/10.1080/14653240510027082

34. Man, Y. Currently Used markers for CTC isolation - advantages, limitations and impact on cancer prognosis / Y. Man, Q. Wang, W. Kemmner // J. Clin. Exp. Pathol. - 2011. - Vol. 1, N 1. - Art. 102. https://doi.org/10.4172/2161-0681.1000102

35. Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: a validation study of the CellSearch system / S. Riethdorf [et al.] // Clin. Cancer Res. - 2007. - Vol. 13, N 3. - P. 920-928. https://doi.org/10.1158/1078-0432.ccr-06-1695

36. Circulating tumor cells in solid tumor in metastatic and localized stages / L. M. Maestro [et al.] // Anticancer Res. -2009. - Vol. 29, N 11. - P. 4839-4843.

37. Circulating tumour cells early predict progression-free and overall survival in advanced colorectal cancer patients treated with chemotherapy and targeted agents / J. Tol [et al.] // Ann. Oncol. - 2010. - Vol. 21, N 5. - P. 1006-1012. https://doi.org/10.1093/annonc/mdp463

38. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance / K. R. Fischer [et al.] // Nature. - 2015. - Vol. 527, N 7579. - P. 472-476. htpps://doi.org/10.1038/nature15748

39. The initial detection and partial characterization of circulating tumor cells in neuroendocrine prostate cancer / H. Beltran [et al.] // Clin. Cancer Res. - 2016. - Vol. 22, N 6. - P. 1510-1519. https://doi.org/10.1158/1078-0432.ccr-15-0137

40. Lianidou, E. S. Circulating tumor cells as emerging tumor biomarkers in breast cancer / E. S. Lianidou, A. Markou // Clin. Chem. Lab. Med. - 2011. - Vol. 49, N 10. - P. 1579-1590. https://doi.org/10.1515/cclm.2011.628

41. Eslami, S. Z. Circulating tumor cells: moving forward into clinical applications / S. Z. Eslami, L. E. Cortes-Hernandez, C. Alix-Panabieres // Precis Cancer Med. March. - 2020. - Vol. 3, N 4. - P 781-791. https://pubmed.ncbi.nlm.nih.gov/31377345/

42. Antiepithelial cell adhesion molecule antibodies and the detection of circulating normallike breast tumor cells / A. M. Sieuwerts [et al.] // J. Natl. Cancer Inst. - 2009. - Vol. 101, N 1. - P. 61-66. https://doi.org/10.1093/jnci/djn419

43. Molecular biomarker analyses using circulating tumor cells / E. A. Punnoose [et al.] // PLoS ONE. - 2010. - Vol. 5, N 9. - P. e12517. https://doi.org/10.1371/journal.pone.0012517

44. Detection of EpCAM negative and cytokeratin-negative circulating tumor cells in peripheral blood / S. D. Mikolajczyk [et al.] // J. Oncol. - 2011. - Vol. 2011. - P. 252361. https://doi.org/10.1155/2011/252361

45. EGFR-based immunoisolation as a recovery target for low-EpCAM CTC subpopulation / A. Vila [et al.] // PLoS ONE. -2016. - Vol. 11, N 10. - P. e0163705. https://doi.org/10.1371/journal.pone.0163705

46. Long term survival following the detection of circulating tumour cells in head and neck squamous cell carcinoma / S. C. Winter [et al.] // BMC Cancer. - 2009. - Vol. 9, N 1. - Art. 424. https://doi.org/10.1186/1471-2407-9-424

47. Cell-free RNA content in peripheral blood as potential biomarkers for detecting circulating tumor cells in non-small cell lung carcinoma / X. M. Yu [et al.] // Int. J. Mol. Sci. - 2016. - Vol. 17, N 11. - P. 1845. https://doi.org/10.3390/ijms17111845

48. Correlation of HER2 status between primary tumors and corresponding circulating tumor cells in advanced breast cancer patients / M. Pestrin [et al.] // Breast Cancer Res. Treat. - 2009. - Vol. 118, N 3. - P. 523-530. https://doi.org/10.1007/s10549-009-0461-7

49. Detection of HER2-positive circulating tumor cells using the LiquidBiopsy System in breast cancer / W. Chen [et al.] // Clin. Breast Cancer. - 2019. - Vol. 19, N 1. - P. 239-246. https://doi.org/10.1016/j.clbc.2018.10.009

50. Prognostic significance of MUC-1 in circulating tumor cells in patients with metastatic pancreatic adenocarcinoma / E. Dotan [et al.] // Pancreas. - 2016. - Vol. 45, N 8. - P. 1131-1135. https://doi.org/10.1097/mpa.0000000000000619

51. MUC1-positive circulating tumor cells and MUC1 protein predict chemotherapeutic efficacy in the treatment of metastatic breast cancer / J.-P. Cheng [et al.] // J. Cancer. - 2011. - Vol. 30, N 1. - P. 54-61. https://doi.org/10.5732/cjc.010.10239

52. Influence of surgical manipulation and surgical modality on the molecular detection of circulating tumor cells from colorectal cancer / Soo Yeun Park [et al.] // J. Korean Surg. Soc. - 2012. - Vol. 82, N 6. - P. 356-364. https://doi.org/10.4174/jkss.2012.82.6.356

53. Circulating tumour cells: molecular properties and anti-cancer treatment monitoring / A. Baigenzhin [et al.] // J. Clin. Med. Kazakhstan. - 2013. - Vol. 4, N 30. - P. 9-13.

54. Paterlini-Brechot, P. // Circulating tumor cells (CTC) detection: clinical impact and future directions / P. Paterlini-Brechot, N. L. Benali // Cancer Lett. - 2007. - Vol. 253, N 2. - P. 180-204. https://doi.org/10.1016/j.canlet.2006.12.014

55. Circulating tumor cell count is a prognostic factor in metastatic colorectal cancer patients receiving first-line chemotherapy plus bevacizumab: a Spanish Cooperative Group for the Treatment of Digestive Tumors study / J. Sastre [et al.] // Oncologist. - 2012. - Vol. 17, N 7. - P. 947-955. https://doi.org/10.1634/theoncologist.2012-0048

56. Paoletti, C. Circulating tumor cells / C. Paoletti, D. F. Hayes // Novel Biomarkers in the Continuum of Breast Cancer / ed. V. Stearns. - Cham, 2016. - Vol. 882. - P. 235-258. https://doi.org/10.1007/978-3-319-22909-6_10

57. Gwatkin, R. B. L. Practical flow cytometry, by H. M. Shapiro, Wiley-Liss, New York, 3rd ed., 1994, 542 p. / R. B. L. Gwatkin // Mol. Rep. Dev. - 1995. - Vol. 41, N 4. - P. 540. https://doi.org/10.1002/mrd.1080410419

58. Ten-color 15-antibody flow cytometry panel for immunophenotyping of lymphocyte population / A. Rajab [et al.] // Int. J. Lab. Hematol. - 2017. - Vol. 39, suppl. 1. - P. 76-85. https://doi.org/10.1111/ijlh.12678

59. Postoperative metastasis prediction based on portal vein circulating tumor cells detected by flow cytometry in periampullary or pancreatic cancer / L. Tao [et al.] // Cancer Manag. Res. - 2019. - Vol. 11. - P. 7405-7425. https://doi.org/10.2147/cmar.s210332

60. Isolation and characterization of living circulating tumor cells in patients by immunomagnetic negative enrichment coupled with flow cytometry / Y. Lu [et al.] // Cancer. - 2015. - Vol. 121, N 17. - P. 3036-3045 https://doi.org/10.1002/cncr.2944

61. Xu, J. How do we use multicolor flow cytometry to detect minimal residual disease in acute myeloid leukemia? / J. Xu, J. L. Jorgensen, S. A. Wang // Clin. Lab. Med. - 2017. - Vol. 37, N 4. - P. 787-802. https://doi.org/10.1016/j.cll.2017.07.004

62. Способ определения циркулирующих опухолевых клеток : пат. BY 23371 / А. Е. Гончаров, О. B. Тимохина. -Опубл. 30.04.2021.


Рецензия

Просмотров: 837


Creative Commons License
Контент доступен под лицензией Creative Commons Attribution 4.0 License.


ISSN 1029-8940 (Print)
ISSN 2524-230X (Online)