Circulating and disseminated tumor cells as a possible prognostic factor for oncological diseases

封面

如何引用文章

全文:

详细

Background. Distant metastasis remains the main cause of death for patients, despite some successes in the diagnosis and treatment of solid tumors. This demonstrates the significance to study the methods for the timely detection of tumor micrometastases and to find the role of tumor cells preserved in peripheral blood or disseminated tumor cells in bone marrow after the end of the treatment.

Aim. To generalize the data concerning the significance and methodology of the determining circulating tumor cells in peripheral blood and disseminated tumor cells in bone marrow in case of different types of cancer.

Materials and methods. Foreign and domestic articles concerning this topic, published in eLibrary, PubMed, Medline, Scopus databases have been analyzed.

Results. The literary review deals with the modern methods of determination, we have analyzed the importance of identification of circulating and disseminated tumor cells as the clinical predictors for specific nosological types of cancer.

Conclusion. The enumeration of circulating and disseminated tumor cells could be a new generation of screening and evaluation of the efficacy of cancer therapy. However, it is necessary and up to date to develop the methodology of their quantitative and qualitative identification and to find the methods for evaluating functional activity.

作者简介

Andrei Kaprin

National Medical Research Radiological Centre

Email: vesper04@mail.ru
ORCID iD: 0000-0001-8784-8415

D. Sci. (Med.), Prof., Acad. RAS

俄罗斯联邦, Obninsk

Svetlana Zatsarenko

Tsyb Medical Radiological Research Centre – branch of the National Medical Research Radiological Centre

编辑信件的主要联系方式.
Email: vesper04@mail.ru
ORCID iD: 0000-0002-6335-1401
SPIN 代码: 7894-9405

Res. Officer

俄罗斯联邦, Obninsk

Sergei Ivanov

Tsyb Medical Radiological Research Centre – branch of the National Medical Research Radiological Centre

Email: vesper04@mail.ru
ORCID iD: 0000-0001-7689-6032

D. Sci. (Med.), Prof. RAS

俄罗斯联邦, Obninsk

Lyudmila Grivtsova

Tsyb Medical Radiological Research Centre – branch of the National Medical Research Radiological Centre

Email: grivtsova@mail.ru
ORCID iD: 0000-0001-9103-9688
SPIN 代码: 4423-6844

D. Sci. (Biol.)

俄罗斯联邦, Obninsk

参考

  1. Тюляндин С.А., Жуков Н.В. Правда о российской онкологии: проблемы и возможные решения. Общероссийская общественная организация «Российское общество клинической онкологии». М., 2018 [Tyulyandin SA, Zhukov NV. The truth about Russian oncology: problems and possible solutions. All-Russian public organization “Russian Society of Clinical Oncology”. Мoscow, 2018 (in Russian)].
  2. Шляхтунов Е.А. Минимальная остаточная болезнь при солидных опухолях – современное состояние проблемы. Новости хирургии. 2014;22(6):735-42 [Shlyakhtunov EA. Minimal residual disease in solid tumors is the current state of the problem. Novosti khirurgii. 2014;22(6):735-42 (in Russian)].
  3. Тупицын Н.Н. Циркулирующие и диссеминированные раковые клетки при раке молочной железы и раке яичников. Онкогинекология. 2013;1:12-8 [Tupitsyn NN. Circulating and disseminated cancer cells in breast and ovarian cancer. Onkoginekologiia. 2013;1:12-8 (in Russian)].
  4. Pantel K, Alix-Panabiеres C, Riethdorf S. Cancer micrometastases. Nat Rev Clin Oncol. 2009;6(6):339-51. doi: 10.1038/nrclinonc.2009.44
  5. Alix-Panabiеres C, Schwarzenbach H, Pantel K. Circulating Tumor cells and circulating tumor DNA. Ann Rev Med. 2012;63:199-215. doi: 10.1146/annurev-med-062310-094219
  6. Аликс Панабьерес Е., Пантел К. Циркулирующие опухолевые клетки: жидкостная биопсия рака. Клиническая лабораторная диагностика. 2014;59(4):60-4 [Alix-Panabieres C, Pantel K. The circulating tumor cells: liquid biopsy of cancer. Klinicheskaia laboratornaia diagnostika. 2014;59(4):60-4 (in Russian)].
  7. Кит О.И., Новикова И.А., Никипелова Е.А., и др. Циркулирующие опухолевые клетки как высокоспецифичный прогностический фактор. Обзор литературы. Международный журнал прикладных и фундаментальных исследований. 2014;12(2):252-7 [Kit OI, Novikova IA, Nikipelova EA, et al. Circulating tumor cells as a highly specific prognostic factor. Literature review. Mezhdunarodnyi zhurnal prikladnykh i fundamental’nykh issledovanii. 2014;12(2):252-7 (in Russian)].
  8. Ненахова Ю.Н., Лядов В.К., Поддубная И.В. Циркулирующие опухолевые клетки: биология, методы выделения, клиническое значение при раке молочной железы. Современная Онкология. 2016;18(3):76-82 [Nenakhova YuN, Lyadov VK, Poddubnaya IV. Circulating tumor cells: biology, isolation methods, clinical significance in breast cancer. Journal of Modern Oncology. 2016;18(3):76-82 (in Russian)].
  9. Бжадуг О.Б., Гривцова Л.Ю., Тупицын Н.Н., и др. Циркулирующие опухолевые клетки в крови больных местнораспространенным и диссеминированным раком молочной железы. Вестник РОНЦ им. Н.Н. Блохина РАМН. 2007;18(2):19-22 [Bzhadug OB, Grivtsova LYu, Tupitsyn NN, et al. Circulating tumor cells in the blood of patients with locally advanced and disseminated breast cancer. Journal of N.N. Blokhin Russian Cancer Research Center RAMS. 2007;18(2):19-22 (in Russian)].
  10. Cristofanilli M, Budd G, Ellis MJ, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;351:781-91. doi: 10.1056/NEJMoa040766
  11. Zhang L, Riethdorf S, Wu G, et al. Meta-analysis of the prognostic value of circulating tumor cells in breast cancer. Clin Cancer Res. 2012;18(20):5701-10. doi: 10.1158/1078-0432.CCR-12-1587
  12. Riethdorf S, Muller V, Zhang L, 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(9):2634-45. doi: 10.1158/1078-0432.CCR-09-2042
  13. Fehm T, Müller V, Aktas B, et al. HER2 status of circulating tumor cells in patients with metastatic breast cancer: a prospective, multicenter trial. Breast Cancer Res Treat. 2010;124:403-12. doi: 10.1007/s10549-010-1163-x
  14. Cristofanilli M, Pierga JY, Reuben J, et al. The clinical use of circulating tumor cells (CTCs) enumeration for staging of metastatic breast cancer (MBC): International expert consensus paper. Crit Rev Oncol Hematol. 2019;134:39-45. doi: 10.1016/j.critrevonc.2018.12.004
  15. Кайгородова Е.В., Тарабановская Н.А., Суркова П.В., и др. Наличие в крови различных популяций циркулирующих опухолевых клеток у больных раком молочной железы до лечения: связь с пятилетней безметастатической выживаемостью. Сибирский онкологический журнал. 2020;19(6):57-65 [Kaigorodova EV, Tarabanovskaya NA, Surkova PV, et al. The presence of various populations of circulating tumor cells in the blood of breast cancer patients before treatment: association with five-year metastasis-free survival. Siberian Journal of Oncology. 2020;19(6):57-65 (in Russian)]. doi: 10.21294/1814-4861-2020-19-6-57-65
  16. Пономарев В.Е. Значение поражения лимфатических узлов и наличия циркулирующих опухолевых клеток при первично-операбельном раке молочной железы: дис. … канд. мед. наук. М., 2020. Режим доступа: https://www.ronc.ru/. Ссылка активна на 15.08.2020 [Ponomarev VE. Znachenie porazheniia limfaticheskikh uzlov i nalichiia tsirkuliruiushchikh opukholevykh kletok pri pervichno-operabel’nom rake molochnoi zhelezy: dis. … kand. med. nauk. Мoscow, 2020. Available at: https://www.ronc.ru/. Accessed: 15.08.2020 (in Russian)].
  17. Danila DC, Heller G, Gignac GA, et al. Circulating tumor cell number and prognosis in progressive castration-resistant prostate cancer. Clin Cancer Res. 2007;13(23):7053-58. doi: 10.1158/1078-0432.CCR-07-1506
  18. Resel FL, San JML, Galante RI, et al. Prognostic significance of circulating tumor cell count in patients with metastatic hormone-sensitive prostate cancer. Urology. 2012;80(6):1328-32. doi: 10.1016/j.urology.2012.09.001
  19. Cieślikowski WA, Budna-Tukan J, Świerczewska M, et al. Circulating Tumor Cells as a Marker of Disseminated Disease in Patients with Newly Diagnosed High-Risk Prostate Cancer. Cancers (Basel). 2020;12(1):160. doi: 10.3390/cancers12010160
  20. Cohen SJ, Punt CJ, Iannotti N, 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(19):3213-21. doi: 10.1200/JCO.2007.15.8923
  21. Tol J, Koopman M, Miller MC, et al. Circulating tumour cells early predict progression-free and overall survival in advanced colorectal cancer patients treated with chemotherapy and targeted agents. Ann Oncol. 2010;21(15):1006-12. doi: 10.1093/annonc/mdp463
  22. Tan Y, Wu H. The significant prognostic value of circulating tumor cells in colorectal cancer: A systematic review and meta-analysis. Curr Probl Cancer. 2018;42(1):95-106. doi: 10.1016/j.currproblcancer.2017.11.002
  23. Krebs MG, Sloane R, Pries L, et al. Evaluation and prognostic significance of circulating tumor cells in patients with non-small-cell lung cancer. J Clin Oncol. 2011;29(12):1556-63. doi: 10.1200/JCO.2010.28.7045
  24. Poveda A, Kaye SB, McCormack R, et al. Circulating tumor cells predict progression free survival and overall survival in patients with relapsed/recurrent advanced ovarian cancer. Gynecol Oncol. 2011;122(3):567-72. doi: 10.1016/j.ygyno.2011.05.028
  25. Khoja L, Lorigan P, Zhou C, et al. Biomarker utility of circulating tumor cells in metastatic cutaneous melanoma. J Invest Dermatol. 2013;133(6):1582-90. doi: 10.1038/jid.2012.468
  26. Gorges K, Wiltfang L, Gorges TM, et al. Intra-Patient Heterogeneity of Circulating Tumor Cells and Circulating Tumor DNA in Blood of Melanoma Patients. Cancers (Basel). 2019;11(11):1685. doi: 10.3390/cancers11111685
  27. Guibert N, Delaunay M, Lusque A, et al. PD-L1 expression in circulating tumor cells of advanced non-small cell lung cancer patients treated with nivolumab. Lung Cancer. 2018;120:108-12. doi: 10.1016/j.lungcan.2018.04.001
  28. Bergmann S, Coym A, Ott L, et al. Evaluation of PD-L1 expression on circulating tumor cells (CTCs) in patients with advanced urothelial carcinoma (UC). Oncoimmunology. 2020;9(1):1738-98. doi: 10.1080/2162402X.2020.1738798
  29. Sänger N, Effenberger KE, Riethdorf S, et al. Disseminated tumor cells in the bone marrow of patients with ductal carcinoma in situ. Int J Cancer. 2011;129(10):2522-6. doi: 10.1002/ijc.25895
  30. Hüsemann Y, Geigl JB, Schubert F, et al. Systemic spread is an early step in breast cancer. Cancer Cell. 2008;13(1):58-68. doi: 10.1016/j.ccr.2007.12.003
  31. Гривцова Л.Ю., Тупицын Н.Н. Иммунологическая оценка гемодилюции костного мозга при лабораторных исследованиях (на основании теста М. Локен). Медицинский алфавит. 2015;4(18):67-70 [Grivtsova LYu, Tupitsyn NN. Immunological assessment of bone marrow hemodilution in laboratory studies (based on the M. Loken test). Medical Alphabet. 2015;4(18):67-70 (in Russian)].
  32. Козловская С.П., Антоненкова Н.Н., Жаврид Э.А. Современное состояние проблемы лечения пациенток с микрометастазами в костном мозге. Опухоли женской репродуктивной системы. 2016;12(1):12-8 [Kozlovskaya SP, Antonenkova NN, Zhavrid EA. The current state of the problem of treating patients with bone marrow micrometastases. Tumors of Female Reproductive System. 2016;12(1):12-8 (in Russian)].
  33. Давыдов М.И., Тупицын Н.Н., Григорьева Т.А., и др. Метод проточной цитометрии в оценке минимального поражения костного мозга у больных раком. Иммунология гемопоэза. 2014;2(1):8-17 [Davydov MI, Tupitsyn NN, Grigor’eva TA, et al. Flow cytometry method for assessing minimal bone marrow damage in cancer patients. Hematopoiesis Immunology. 2014;2(1):8-17 (in Russian)].
  34. Родионов В.В., Богомолова О.А., Кометова В.В. Метод проточной цитометрии в диагностике диссеминированных опухолевых клеток в костном мозге у больных раком молочной железы. Ульяновский медико-биологический журнал. 2013;1:77-82 [Rodionov VV, Bogomolova OA, Kometova VV. Flow cytometry method in the diagnosis of disseminated tumor cells in the bone marrow in patients with breast cancer. Ulyanovsk Medico-Biological Journal. 2013;1:77-82 (in Russian)].
  35. Braun S, Vogl FD, Naume B, et al. A pooled analysis of bone marrow micrometastasis in breast cancer. N Engl J Med. 2005;353(8):793-802. doi: 10.1056/NEJMoa050434
  36. Gebauer G, Fehm T, Merkle E, et al. Epithelial cells in bone marrow of breast cancer patients at time of primary surgery. Clinical outcome during long-term follow-up. J Clin Oncol. 2001;19(16):3669-74. doi: 10.1200/JCO.2001.19.16.3669
  37. Janni W, Rack B, Kasprowicz N, et al. DTCs in breast cancer: clinical research and practice. Recent Results Cancer Res. 2012;195:173-8. doi: 10.1007/978-3-642-28160-0_15
  38. NCCN Guidelines. Version 1.2021. Breast Cancer NCCN Evidence BlocksTM. 2021 Jan 15:MS3.
  39. Wang G, Wang S, Li Y, et al. Clinical study of disseminated tumor cells in bone marrow of patients with gastric cancer. Hepatogastroenterology. 2013;60(122):273-6. doi: 10.5754/hge12599
  40. Джуманазаров Т.М., Чулкова С.В., Тупицын Н.Н., и др. Детекция диссеминированных опухолевых клеток и их взаимосвязь с популяцией костномозговых лимфоцитов у больных немелкоклеточным раком легкого. Современная Онкология. 2020;22(3):94-9 [Djumanazarov TM, Chulkova SV, Tupitsyn NN, et al. Detection of disseminated tumor cells and their relationship with a population of bone marrow lymphocytes in patients with non-small cell lung cancer. Journal of Modern Oncology. 2020;22(3):94-9 (in Russian)]. doi: 10.26442/18151434.2020.3.200137

版权所有 © Consilium Medicum, 2021

Creative Commons License
此作品已接受知识共享署名-非商业性使用 4.0国际许可协议的许可。
 


##common.cookie##