<<

Ruksha  Div (2019) 14:11 https://doi.org/10.1186/s13008-019-0054-8

REVIEW Open Access ’ control of cancer cell dormancy Tatiana G. Ruksha*

Abstract ‘Dormancy’, in the context of carcinogenesis, is a biological phenomenon of decreased cancer cell proliferation and metabolism. In view of their ability to remain quiescent, cancer cells are able to avoid cell death induced by chemo- therapeutic agents, and thereby give rise to tumor relapse at a later stage. Being a dynamic event, the dormant state is controlled by several epigenetic mechanisms, including the action of microRNAs. The present review highlights microRNAs that have been shown to be dysregulated in dormant cancer cells among diferent tumor types. Micro- accomplish their control of cancer cell quiescence by targeting regulators and signaling pathways involved in maintenance, including the AKT/phosphoinositide 3- (PI3K) pathway. MicroRNAs, as components of intercellular vesicles, enable interactions to occur between cancer cells and cells of the microenviron- ment, resulting in the cancer cells either acquiring the quiescent state or, oppositely, stimulating them to proliferate. Taken together, the evidence obtained to date has collectively confrmed the involvement of microRNAsin cancer cell dormancy. Modulation of the various processes may enable optimization of the treatment of metastatic tumors. Keywords: Quiescence, Dormancy, Cancer, microRNA,

Background nuclear receptor expression, which regulates the gene In terms of carcinogenesis, dormancy may be identi- expression of quiescent cells [4]. Te hydroxymethylation fed as a biological state of tumors associated with a low enzyme, Tet2, was revealed to stimulate expression of proliferation rate and low metabolic activity. It is under- the cell-cycle inhibitors,-dependent kinase inhibi- stood that a limited number of cancer cells reside in a tor 1A (CDKN1A) andCDKN1B, resulting in dormancy quiescent state for an unspecifed period until unknown of the melanoma cells [5]. Less is understood regarding stimuli induce their division, in addition to other biologi- the abilities of microRNAs to infuence cancer quies- cal activities that are specifc to cancer cells. Epigenetic cent cells. More importantly, it is not yet clear whether mechanisms are considered to fulfl the role of such stim- the dormant state of tumor cells should be shifted or not uli. Among the various epigenetic mechanisms, the func- for therapeutic purposes. For that reason, the goal of the tioning of microRNAs results in translational repression present review is to summarize the research data that or decay of target mRNAs. Each microRNA may have have been published concerning the role of microRNAs up to several hundred mRNA targets, and several genes in cancer dormancy, and their impact as modulators of may be regulated by one unique microRNA. Te biogen- tumor relapse and progression. esis, functioning and regulation of microRNAs have been thoroughly analyzed in numerous reviews [1–3]; there- Main text fore, the present review will not focus in any great detail Tumor dormancy defnition and methods used on these aspects. Recently, it was shown that epigenetic for quiescent cells identifcation therapies may afect dormant cells. Te DNA-methylat- Te frst data concerning the role of microRNAs in can- ing agent, 5-aza-2′-deoxycytidine (5-aza-C), in combi- cer was published in 2002, when the research group of nation with retinoic acid, was shown to restore orphan C. Croce reported on microRNA-15 (miR-15) and miR- 16 downregulation in chronic lymphocytic leukemia cells [6]. Later it was discovered that mic are implicated *Correspondence: [email protected] Department of Pathological Physiology, Krasnoyarsk State Medical in fundamental carcinogenesis-associated processes, University, P. Zeleznyaka str., 1, Krasnoyarsk 660022, Russia including regulation via microRNAs targeting genes

© The Author(s) 2019. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/ publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated. Ruksha Cell Div (2019) 14:11 Page 2 of 7

associated with the cell cycle, , DNA repair, cells, formed microscopically sized tumors at the site oncosupression, and cancer cell migration, invasion and of injection, which remained stable for up to 125 and evasion of the immune response. 88 days for the osteosarcoma and glioblastoma tumor Tumor cell quiescence may be defned as a reversible models, respectively. miR-190-overexpressed dormant state of growth arrest in which cells enter the ­G0-phase microtumors were shown to contain proliferative cells, of the cell cycle [7]. Dormant tumors characterized by a and miR-190 gene target and signaling pathway analyses balance of apoptosis and cell proliferation results in the revealed that miR-190-associated dormancy was asso- absence of progression and clinical symptoms. Cancer ciated with antigenic pattern expression redistribution stem cells, which are determined as a cancer cell subpop- on cancer cells, followed by immune escape. To identify ulation responsible for tumor initiation and progression, microRNAs involved in regulating the shift of breast can- can be dormancy competent, although not only cancer cer cells from the dormant to the metastatic state, Gian- stem cells can become quiescent. Acquisition quiescence cotti et al. [14] constructed cDNA libraries originating by cancer cells is a fexible process that can be induced by from metastatic breast cancer cells. Subsequently, cDNA stress factors, including chemotherapy [8]. libraries were transduced into breast cancer 4TO7 cells Core aspects of any study on cancer cell dormancy will that resided in the lungs, but did not give rise to metas- include the choice of approaches that are used to char- tasis. Murine microRNAs shown previously to give rise acterize the dormant, quiescent state of the cancer cells, to breast cancer metastasis in the lung were divided into and to diferentiate that pool of cells within the total pop- sets of up to 97 microRNAs transfected into 4TO7 cells, ulation of cells in the tumor. Even though the phenom- followed by injection into mice. Te tumors that formed enon of tumor dormancy was frst described in 1934 [9], in mice were examined further for their microRNA a lack of unbiased methods for objectively studying this expression levels, and miR-340, miR-346, and a combina- topic has restricted the quantity and the quality of the tion of miR-138 and miR-223 were identifed in each ani- research results that have been obtained. mal where breast cancer lung metastases had developed, Cell cycle phase distribution monitoring via several indicating that the latter could be implicated in the trans- standard procedures, including fow cytometric analysis formation of breast cancer cells from the quiescent to to determine cells residing in the quiescent(G0) and gap the metastatic state. A similar study performed by Mori 1 ­(G1) phases, may be used to investigate the population et al. [15] on gastrointestinal cancer stem cells separated of dormant cells. Furthermore, cells that are resistant to them into CD13­ −/CD90+ proliferating cancer stem cells various chemotherapeutic agents, and those which bear and ­CD13+/CD90− dormant cancer stem cells. Te lat- cancer stem cell markers, are currently being investi- ter were characterized according to their downregulated gated. To confrm the proliferative state of cells, cell expression of miR-101 compared with ­CD13−/CD90+ proliferation-specifc markers, such as the Ki-67 antigen, cells. Bioinformatic analysis revealed that miR-101 was have been used, as well as the incorporation of dyes [10]. implicated in the cytokine/chemokine regulatory path- As cellular quiescence is regulated by and cyclin- way (specifcally, interleukin-8 and CXCL5), which again dependent [11], this phenomenon may be inves- demonstrated the involvement of the immune system tigated by estimating their and mRNA levels. in the mechanism of maintenance of dormancy of can- Dormant tumor formation in vivo may be studied via the cer stem cells. Te CD13­ +/CD90−dormant cancer stem formation of small tumors that are stable in size, which cells were further characterized by their association with develop within a lag period of time following injection of regulation of the tumor protein (TP53) signaling cancer cells into a model animal. Patient-derived xeno- pathway, which is involved with chemoresistance devel- graft tissues implanted into immunocompromised mice opment upon treatment with genotoxic agents. is one method that may be utilized for this process [12]. Dormant cancer cells residing in osteosarcoma, glio- blastoma, breast cancer and liposarcoma in vivo mod- MicroRNA profling of quiescent cells els were established by Abdollahi et al. [18]. All of these Measuring the expression of microRNAs is a standard tumor types were characterized by high proliferation approach for identifying the exact molecules that are and apoptotic rates, as well as impaired angiogenesis. crucial in the regulation of a process that is being inves- MicroRNA profling revealed similar microRNA expres- tigated. MicroRNA profling is used to characterize the sion patterns in all of these tumor types, where 16 out of unique expression pattern of quiescent cells. Almog et al. 19 microRNAs (84%) were upregulated compared with [13] performed an array scanning analysis in dormant non-dormant tumors of the same origin. Restoration tumor models of osteosarcoma and glioblastoma, where of the levels of the most upregulated microRNAs, miR- miR-190 was identifed to be one of the most upregu- 580, miR-588 and miR-190, in fast-growing glioblastoma lated microRNAs. miR-190, overexpressed in cancer cells resulted in tumor growth delay. Gene targets of the Ruksha Cell Div (2019) 14:11 Page 3 of 7

aforementioned microRNAs included tissue inhibitor of this pathway, bone morphogenetic protein and activin metalloproteinases 3, hypoxia-induced factor-1α, basic membrane bound inhibitor (BAMBI) [19]. fbroblast growth factors and the K-Ras tumor , Cancer cell quiescence is also maintained by heat- which are well known carcinogenesis-related genes. shock , of which heat-shock protein90 (Hsp90) has been shown to regulate protein function associ- ated with cancer cell ‘stemness’ and quiescence. In that MicroRNAs and their target genes are implicated regard, Hsp90-specifc inhibitors were revealed to induce in proliferation‑quiescent state maintenance a decrease in the expression of the cancer stem cell mark- To further elucidate the role of microRNAs in can- ers, CD44, and aldehyde dehydrogenase (ALDH). In cer cells’ shift from dormancy to the proliferative state, addition, microRNA expression profles were subjected Watson et al. [17] restored expression of the miR- to alterations when microRNAs involved in the regula- 200a/200b/429 cluster in the murine mammary tumor tion of signaling pathways such as those connected with cell line. Te miR-200a/200b/429 cluster, which is mitogen-activated protein kinase (MAPK), β-catenin, involved in regulating the proliferation and migration and cell cycle/proliferation were revealed to be dysregu- of stem cells by targeting mesenchymal transcription lated [20]. factors, belongs to a microRNA family that is highly Several studies have shown that miR-221 and miR-222 conserved among vertebrates. Reconstruction of the act as dormancy regulators. Acute lymphoblastic leu- expression of the aforementioned microRNAs in the kemia cells co-cultured with bone marrow stromal cells murine mammary tumor cell line resulted in a decrease and primary human osteoblasts demonstrated decreased in cell proliferation, as well as conversion of the cells’ expression levels of miR-221 and miR-222, microRNAs morphology into the epithelial type. Furthermore, it led that were described previously as leukemia cell prolifera- to a diminution of the levels of the transcription fac- tion regulators targeting the cell cycle inhibitor, CDKN1B tors Snail, Twist1, Twist2, and Zeb1, and induced tumor (or p27). Previously, it was shown that p27 is involved transition to the state of dormancy, as determined by the in the quiescence of acute lymphoblastic leukemia cells formation of small size tumors in vivo. miR-200c has pre- [21]. Targeted downregulation of miR-221 and miR-222, viously been shown to be involved in maintenance of the followed by p27 overexpression, resulted in cell cycle dormant state of osteosarcoma tumors. In the study by arrest and an increase in chemotherapy resistance in the Tiram et al. [18], the osteosarcoma cell line was gener- presence of bone marrow niche components, including ated by using non-tumorigenic osteosarcoma cells, which stromal cells and osteoblasts, as well as independently gave rise to small tumors that did not exhibit any signs of of these cells. Modulation of miR-221 and miR-222 was angiogenesis and were stable in terms of tumor volume also revealed to trigger quiescence of leukemia cells for a time period lasting up to 140 days in mice. miR- [22]. Similar results were obtained in glioblastoma cells, 200c was one of the microRNAs that were diferently where the levels of miR-221 and miR-222 were shown expressed in dormant-tumor-forming cells compared to be increased in cells as they were entering into the with fast-growing ones, and its overexpression was asso- cell cycle. Again, p27, and also p57, were validated as ciated with a prolonged survival rate in vivo. Terefore, direct gene targets of these aforementioned microRNAs. these aforementioned data demonstrates miR-200 family Transfection with antimiR-222 inhibitor resulted in an involvement in tumor dormancy regulation. increased number of cells residing in ­G0 phase. By con- Several experiments have been aimed at elucidating trast, miR-221 and miR-222 were shown to stimulate the mechanisms underlying the transition of cancer cells cells to enter the synthesis (S) phase, followed by apop- between the proliferative and dormant states. ΔNp63α, totic death. Anti-miR-222/223 transfection in mesen- which is an isoform of p63 protein, was shown to be chymal stem cells transformed them to dormant cancer implicated in mammary stem cell quiescence. In MCF7 cells via exosomes and gap junctions, which resulted in luminal estrogen-receptor-positive breast cancer cells, a moderation of the proliferation rate of cancer cells and ΔNp63α stimulated reversible growth arrest, indicating prolonged survival rates in vivo [23]. Terefore, low lev- its role in dormancy state development. MiR-205 was els of miR-221 and miR-222 in quiescent cancer cells are shown to be the most upregulated microRNA in MCF7 considered to act as a mechanism that protects against cells overexpressing ΔNp63α. Te application of anmiR- an undesired shift from the quiescent to the proliferative 20-specifc mimic led to a decrease in the cell prolif- state, prior to the onset of apoptosis [11]. eration rate, and caused dormancy-associated protein Turning our attention to bone marrow breast can- dysregulation, including components of the bone mor- cer cells, these are characterized by the CD44­ +/CD24− phogenetic protein signaling pathway target genes, inhib- expression pattern. MiR-23b originating from bone itor of DNA binding 1 (ID1) and ID3,andan inhibitor of marrow increased the number of ­CD44− cells, thereby Ruksha Cell Div (2019) 14:11 Page 4 of 7

showing its potential to induce breast cancer dormancy, demonstrating bimodal efects on populations of regula- as ­CD44− cells were revealed to have a decreased pro- tory T cells (Tregs) and T helper 17 (T17) cells, and on liferation rate, reduced invasive capabilities, as well as interleukin-17 levels [27]. Mesenchymal stem cells and being less sensitive to the chemotherapeutic agent, doc- breast cancer cells communicate via gap junctions and etaxel [24]. MiR-424 and miR-503 were able to afect exosomes, both of which are characterized by a specifc the cell cycle, leading to an increase in the number of ­G1 microRNA profle. MiR-222/223, as mentioned above, cells in colon cancer and osteosarcoma cells by inhibit- are involved in cancer cell drug resistance [28], and these ing Cdc25A protein, which has an important role in cell microRNAs were revealed to be overexpressed in mesen- cycle-dependent kinase activation [25]. Taking together, chymal stem cell-derived exosomes, which indicates their these reports indicate the impact of microRNAs on cell ability to act as signal transducers for a dynamic state of cycle-related genes, resulting in entry to the cell cycle or cancer cells upon microenvironmental stimuli. its arrest. MiR-126 is overexpressed in normal stem cells and progenitor hematopoietic cells, and is associated with MicroRNAs support interactions between proliferating/ an increased number of dormant leukemia stem cells. quiescent cells and microenvironment As endothelial cells were shown to express the highest Te tumor microenvironment itself is considered to rates of miR-126 among the components of the tumor be actively involved in carcinogenesis. Its components microenvironment, communication between these communicate via microRNAs with cancer cells, result- cells and leukemia stem cells was inferred. Co-culture ing in their switch from a dormant to a proliferative state of endothelial cells and leukemia stem cells confrmed (Fig. 1). Wells et al. [26] demonstrated that the micro- the occurrence of miR-126 trafcking from endothe- RNA profle of breast cancer cell-derived exosomes dif- lial cells to leukemia stem cells via extracellular vesicles. fered considerably from that of hepatic niche-derived Modulation of the expression level of miR-126 in leu- exosomes. MicroRNAs derived from the latter were kemia cells resulted in an increase in their proliferation able to modulate cell proliferation of breast cancer cells, and apoptosis rates, as well as diminished colony forma- triggering several signaling pathways, and restoring tion. MiR-126 downregulation in endothelial cells and the epithelial-type morphology of cells via induction of chronic myelogenous leukemia stem cells induced antitu- the re-expression of the epithelial marker, E-cadherin mor efects of tyrosine kinase inhibitors, a phenomenon [26]. Bone marrow is an important site for quiescent that was determined via their increased survival rates in breast cancer cells. Mesenchymal stem cells commu- miR126a-knockout mice [29]. As has been demonstrated nicate to distinct breast cancer cells subsets diferently, in hematopoietic stem cells, miR-126 may regulate the expression of several components of the phospho- inositide 3-kinase (PI3K)/AKT signaling pathway [30], which was defned as an essential component of dormant estrogen-dependent breast cancer cells [31].

Impact of microRNA level alterations on cell quiescence Tere is no clear evidence to suggest if the shift between the quiescent and proliferative states can be considered a therapeutic goal. Terefore, gain-and-loss of function studies may help to provide further understanding. Pan- creatic ductal adenocarcinoma stem cells resistant to chemotherapeutic agents were revealed to comprise the population of quiescent cells that are characterized by resistance to gemcitabine, predominantly existing in the ­G0 and ­G1, but not the S, cell cycle phases, increased lev- els of CDKN1A and CDKN1C, and decreased levels of . MicroRNA profling revealed the miR-17-92 cluster to be downregulated in gemcitabine-resistant cells compared with vehicle-treated pancreatic ductal adeno- carcinoma stem cells, whereas the levels of the miR-17-92 cluster were upregulated in pancreatic cancer bulk tumor Fig. 1 Summary diagram of microRNAs implicated in tumor cell quiescence tissues. MiR-17-92 levels were decreased in diferenti- ated pancreatic cancer cells, leading to an enhancement Ruksha Cell Div (2019) 14:11 Page 5 of 7 [ 25 ] [ 18 ] [ 17 ] [ 13 ] [ 29 ] [ 15 ] References [ 14 ] [ 21 , 14 ] [ 22 ] [ 19 ] [ 14 ] [ 16 ] [ 16 ] [ 25 ] -

1 -to-G 0 cells 1 G cells, cell cycle arrest via targeting M-phase cell cycle via targeting cells, arrest 1 ­ G of induced factor 1α, basic fbroblast growth factor, factor, growth induced factor 1α, basic fbroblast K-Ras/metastasis binding protein 3/cell proliferation binding protein tion, apoptosis, and colony formation and colony tion, apoptosis, tency 1B, p27/drug resistance 1B, inhibitor 1B (or p27)/cell cycle sensitiza - 1B (or inhibitor regulation, agents cytotoxic tion to target genes (ID1 andID3), and the inhibitor of this genes (ID1 andID3), and the inhibitor target BAMBI/induction of quiescence in cells pathway, phase transition enhancement induced factor-1α, basic fbroblast growth factor, factor, growth basic fbroblast induced factor-1α, K-Ras/induction and dormancy of antiangiogenic EphA5, Angiomotin pathways promoting induced factor-1α, basic fbroblast growth factor, factor, growth basic fbroblast induced factor-1α, K-Ras/induction and dormancy of antiangiogenic EphA5, Angiomotin pathways promoting inducer phosphotase , cyclin in the number E, Cdc25A/Increase D, Cyclin Tissue inhibitor of metalloproteinases 3, hypoxia- of metalloproteinases inhibitor Tissue Snail, Twist1, Twist2, Twist2, Zeb1/cell Twist1, proliferation Snail, Nuclear factor I/B, human T cell leukemia virus type I human Nuclear factor I/B, - PI3K/AKT components/prolifera pathway signaling , ALK1, and transcription factor TBX3/G p21, ALK1, and transcription factor JARID1A, JMJD1B, TP53INP1, EZH2/stem cell pluripoTP53INP1, EZH2/stem JARID1A, JMJD1B, Target/key biological events regulated events biological Target/key Increase in the number of ­ Drug resistance enhancement to carboplatin enhancement to Drug resistance Cell cycle cyclin-dependentCell inhibitor kinase inhibitor The cell cycle inhibitor, cyclin-dependent kinase cell cycleThe inhibitor, Bone morphogenetic protein signaling pathway pathway signaling Bone morphogenetic protein Tissue inhibitor of metalloproteinases 3, hypoxia- of metalloproteinases inhibitor Tissue Tissue inhibitor of metalloproteinases 3, hypoxia- of metalloproteinases inhibitor Tissue detection of anti- H3 antibodies detection of anti-histone tent measurement, Kaplan–Meier measurement, tent analysis and Ki-67 immunohistochemistry dormancy for models in vivo cells identifcation by immunocytochemistrycells identifcation by nostaining deoxyuridine staining immunostaining deoxyuridine-Ki-67 double immunostaining cellular DNA content measurement, Ki-67 measurement, immu - cellular DNA content nostaining cellular DNA content measurement, Ki-67 measurement, immu - cellular DNA content nostaining Flow cytometryFlow cell cycle analysis for Kaplan–Meier survival analysis for Cell proliferation measured by immunofuorescent immunofuorescent by measured proliferation Cell In vitro proliferation assay based on cellular DNA con - assay In proliferation vitro Fluorescence activated cell sortingFluorescence Cell cycle cytometry,Cell fow Ki-67 analysis by negative Fluorescence activated cell sortingFluorescence Method used for quiescent cell identifcation cell Method used for quiescent Flow cytometryFlow cell cycle analysis for Flow cytometryFlow cell cycle Ki-67 analysis, for immu - Flow cytometryFlow cell cycle 5-bromo-2 ′ - analysis, for Cell proliferation evaluation by fow cytometry, fow evaluation p27 by proliferation Cell ‐ 2 ′ Flow cell cycle 5 ‐ bromo analysis, for Tumor volume analysis, proliferation assay based on assay proliferation analysis, volume Tumor Tumor volume analysis, proliferation assay based on assay proliferation analysis, volume Tumor

− /CD90 + CD13 quiescent cancer stem cells quiescent cancer stem 0 cells enous leukemia cells stem subpopulation cells quiescent cells liposarcoma/quiescent cells liposarcoma/quiescent liposarcoma/quiescent cells liposarcoma/quiescent Colon cancer, osteosarcoma/proliferating cancer osteosarcoma/proliferating cancer, Colon Breast cancer/G Breast Osteosarcoma/dormant-tumor-forming cells Osteosarcoma/dormant-tumor-forming Breast cancer/low proliferative cells proliferative cancer/low Breast Osteosarcoma, glioblastoma/dormant cells Osteosarcoma, - myelog leukemia/chronic myelogenous Chronic Pancreatic cancer/quiescent cancer stem cells cancer/quiescent cancer stem Pancreatic Cancer type involved/defnition used type involved/defnition Cancer studied cells to describe Colon cancer, osteosarcoma/proliferating cancer osteosarcoma/proliferating cancer, Colon Breast cancer/breast cancer dormant cells stem cancer/breast Breast Acute lymphoblastic leukemia, breast cancer/ lymphoblastic leukemia, breast Acute Acute lymphoblastic leukemia/quiescent cells Acute cancer/liver cancer stem cell ­ cancer stem cancer/liver Liver Osteosarcoma, glioblastoma, breast cancer, cancer, breast glioblastoma, Osteosarcoma, Osteosarcoma, glioblastoma, breast cancer, cancer, breast glioblastoma, Osteosarcoma, Characteristics of cancer cell quiescent state-associated microRNAs quiescent cell of cancer Characteristics cluster miR-503 miR-205 miR-200c miR-200a/200b/429 miR-190 miR-126 miR-17-92 1 Table MicroRNA and activin kinase 1/3, BAMBI bound inhibitor bone morphogenetic Δ-isomerase membrane protein 1, PI3K phosphoinositide 3-kinase, ID1/3 isopentenyl-diphosphate AKL1 Activin receptor-like miR-424 miR-223 miR-222 miR-221 miR-101 miR-588 miR-580 Ruksha Cell Div (2019) 14:11 Page 6 of 7

in the level of the stem cell marker, CD133, as well as an Consent for publication Not applicable. increase in the number of cells in ­G0/. Further- more, anmiR-17-92 inhibitor stimulated tumor growth Competing interests in vivo. Subsequent gene target analysis revealed that The author declares there is no competing interest. the miR-17-92 cluster activated the cell-cycle regulator Received: 1 November 2018 Accepted: 11 September 2019 p21, activin receptor-like kinase 1 (ALK1), and the tran- scription factor, TBX3, which is implicated in cell cycle and cancer progression control.TBX3 itself infuenced the transforming growth factor-β1 signal pathway [12]. References In summary, the identifcation of microRNAs, which can 1. Creugny A, Fender A, Pfefer S. Regulation of primary microRNA process- ing. FEBS Lett. 2018;592(12):1980–96. be key regulators of cancer quiescence, could be useful to 2. Kian R, Moradi S, Ghorbian S. Role of components of microRNA machin- prevent minimal residual disease [8]. ery in carcinogenesis. Exp Oncol. 2018;40(1):2–9. 3. Liu Y, Lv J, Liang X, Yin X, Zhang L, Chen D, Jin X, Fiskesund R, Tang K, Ma J, Zhang H, Dong W, Mo S, Zhang T, Cheng F, Zhou Y, Xie J, Wang N, Huang Conclusions B. Fibrin stifness mediates dormancy of tumor-repopulating cells via a Cdc42-driven tet2 epigenetic program. Cancer Res. 2018;78(14):3926–37. To conclude, several mechanisms underpinning the 4. Sosa MS, Parikh F, Maia AG, Estrada Y, Bosch A, Bragado P, Ekpin E, George involvement of microRNAs in regulation of the quiescent A, Zheng Y, Lam HM, Morrissey C, Chung CY, Farias EF, Bernstein E, Aguirre-Ghiso JA. NR2F1 controls tumour cell dormancy via SOX9- and state of cancer cells, and their shift to a proliferative state, RARβ-driven quiescence programmes. Nat Commun. 2015;30(6):6170. have been elucidated (Table 1). MicroRNAs take part in 5. Treiber T, Treiber N, Meister G. Regulation of microRNA biogenesis and its the quiescent-proliferative state of cancer cells and its crosstalk with other cellular pathways. Nat Rev Mol Cell Biol. 2018. https​:// doi.org/10.1038/s4158​0-018-0070-6. dynamic balance. Proper microRNA profling for various 6. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, et al. Frequent cancer types is required to determine if specifc micro- deletions and down-regulation of micro-RNA genes miR-15 and miR-16 RNAs of quiescent cancer cells exist. Further functional at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 2002;99(24):15524–9. studies may help to elucidate specifc roles of these epige- 7. Ghajar CM. Metastasis prevention by targeting the dormant niche. Nat netic regulators in tumor dormancy. Terapeutic strate- Rev Cancer. 2015;15:238–47. gies targeting microRNAs may be hypothesized, but their 8. Talukdar S, Bhoopathi P, Emdad L, Das S, Sarkar D, Fisher PB. Dormancy and cancer stem cells: an enigma for cancer therapeutic targeting. Adv usefulness and applicability require validation by further Cancer Res. 2019;141:43–84. research, including in vivo studies, before any frm con- 9. Willis RA. The Spread of tumours in the human body. London: J&A. clusions may be reached. A broadening of the experimen- Churchill; 1934. 10. Romar GA, Kupper TS, Divito SJ. Research techniques made simple: tech- tal systems investigated to include other cancer types, niques to assess cell proliferation. J Invest Dermatol. 2016;136:e1–7. both in vitro and in vivo, is required, as the majority of 11. Medina R, Zaidi SK, Liu CG, Stein JL, van Wijnen AJ, Croce CM, Stein GS. the studies performed to date have been conducted MicroRNAs 221 and 222 bypass quiescence and compromise cell survival. Cancer Res. 2008;68(8):2773–801. on breast cancer and leukemia models. An improved 12. Ciof M, Trabulo SM, Sanchez-Ripoll Y, Miranda-Lorenzo I, Lonardo E, understanding of the diversity of cancer cells, and their Dorado J, Reis Vieira C, Ramirez JC, Hidalgo M, Aicher A, Hahn S, Sainz associated sensitivities to chemotherapeutic agents, will B Jr, Heeschen C. The miR-17-92 cluster counteracts quiescence and chemoresistance in a distinct subpopulation of pancreatic cancer stem optimize personalized therapy approaches, which are cells. Gut. 2015;64(12):1936–48. necessary if cancer survival rates are to be improved. 13. Almog N, Briggs C, Beheshti A, Ma L, Wilkie KP, Rietman E, Hlatky L. Transcriptional changes induced by the tumor dormancy-associated microRNA-190. Transcription. 2013;4(4):177–91. Abbreviations 14. Gao H, Chakraborty G, Lee-Lim AP, Mavrakis KJ, Wendel HG, Giancotti FG. ALDH: aldehyde dehydrogenase; MAPK: mitogen-activated protein kinase; Th: Forward genetic screens in mice uncover mediators and suppressors of T helper cell; Treg: regulatory T cell. metastatic reactivation. Proc Natl Acad Sci USA. 2014;111(46):16532–7. 15. Nishikawa S, Dewi DL, Ishii H, Konno M, Haraguchi N, Kano Y, Fukusumi Acknowledgements T, Ohta K, Noguchi Y, Ozaki M, Sakai D, Satoh T, Doki Y, Mori M. Transcrip- Not applicable. tome study of dormant gastrointestinal cancer stem cells. Int J Oncol. 2012;41(3):979–84. Authors’ contributions 16. Almog N, Ma L, Schwager C, Brinkmann BG, Beheshti A, Vajkoczy P, Folk- TR analyzed and interpreted the data, made fgure and table. She wrote the man J, Hlatky L, Abdollahi A. Consensus micro RNAs governing the switch fnal manuscript. The author read and approved the fnal manuscript. of dormant tumors to the fast-growing angiogenic phenotype. PLoS ONE. 2012;7(8):e44001. Funding 17. Watson KL, Jones RA, Bruce A, Moorehead RA. The miR-200b/200a/429 This study was supported by a Grant from the Russian Science Foundation cluster prevents metastasis and induces dormancy in a murine claudin- (project no. 19-15-00110). low mammary tumor cell line. Exp Cell Res. 2018;369(1):17–26. 18. Tiram G, Segal E, Krivitsky A, Shreberk-Hassidim R, Ferber S, Ofek P, Uda- Availability of data and materials gawa T, Edry L, Shomron N, Roniger M, Kerem B, Shaked Y, Aviel-Ronen Not applicable. S, Barshack I, Calderón M, Haag R, Satchi-Fainaro R. Identifcation of dor- mancy-associated microRNAs for the design of osteosarcoma-targeted Ethics approval and consent to participate dendritic polyglycerol nanopolyplexes. ACS Nano. 2016;10(2):2028–45. Not applicable. Ruksha Cell Div (2019) 14:11 Page 7 of 7

19. Amin R, Morita-Fujimura Y, Tawarayama H, Semba K, Chiba N, Fuku- 27. Patel SA, Dave MA, Bliss SA, Giec-Ujda AB, Bryan M, Pliner LF, Rameshwar P. moto M, Ikawa S. ΔNp63α induces quiescence and downregulates the ­Treg/Th17 polarization by distinct subsets of breast cancer cells is dictated BRCA1 pathway in estrogen receptor-positive luminal breast cancer by the interaction with mesenchymal stem cells. J Cancer Stem Cell Res. cell line MCF7 but not in other breast cancer cell lines. Mol Oncol. 2014;2:e1003. 2016;10(4):575–93. 28. Lim PK, Bliss SA, Patel SA, Taborga M, Dave MA, Gregory LA, Greco SJ, 20. Subramanian C, Kovatch KJ, Sim MW, Wang G, Prince ME, Carey TE, Davis Bryan M, Patel PS, Rameshwar P. Gap junction-mediated import of micro- R, Blagg BSJ, Cohen MS. Novel C-terminal heat shock protein 90 inhibitors RNA from bone marrow stromal cells can elicit cell cycle quiescence in (KU711 and Ku757) are efective in targeting head and neck squamous breast cancer cells. Cancer Res. 2011;71(5):1550–60. cell carcinoma cancer stem cells. Neoplasia. 2017;19(12):1003–11. 29. Zhang B, Nguyen LXT, Li L, Zhao D, Kumar B, Wu H, Lin A, Pellicano F, 21. Furuichi Y, Goi K, Inukai T, Sato H, Nemoto A, Takahashi K, Akahane K, Hopcroft L, Su YL, Copland M, Holyoake TL, Kuo CJ, Bhatia R, Snyder DS, Hirose K, Honna H, Kuroda I, Zhang X, Kagami K, Hayashi Y, Harigaya Ali H, Stein AS, Brewer C, Wang H, McDonald T, Swiderski P, Troadec E, K, Nakazawa S, Sugita K. Fms-like tyrosine kinase 3 ligand stimulation Chen CC, Dorrance A, Pullarkat V, Yuan YC, Perrotti D, Carlesso N, Forman induces MLL-rearranged leukemia cells into quiescence resistant to SJ, Kortylewski M, Kuo YH, Marcucci G. Bone marrow niche trafcking antileukemic agents. Cancer Res. 2007;67(20):9852–61. of miR-126 controls the self-renewal of leukemia stem cells in chronic 22. Moses BS, Evans R, Slone WL, Piktel D, Martinez I, Craig MD, Gibson LF. myelogenous leukemia. Nat Med. 2018;24(4):450–62. Bone marrow microenvironment niche regulates miR-221/222 in acute 30. Lechman ER, Gentner B, van Galen P, Giustacchini A, Saini M, Boccalatte lymphoblastic leukemia. Mol Cancer Res. 2016;14(10):909–19. FE, Hiramatsu H, Restuccia U, Bachi A, Voisin V, Bader GD, Dick JE, Naldini 23. Bliss SA, Sinha G, Sandiford OA, Williams LM, Engelberth DJ, Guiro K, L. Attenuation of miR-126 activity expands HSC in vivo without exhaus- Isenalumhe LL, Greco SJ, Ayer S, Bryan M, Kumar R, Ponzio NM, Ramesh- tion. Cell Stem Cell. 2012;11(6):799–811. war P. Mesenchymal stem cell-derived exosomes stimulate cycling 31. Barrios J, Wieder R. Dual FGF-2 and intergrin alpha5beta1 signaling quiescence and early breast cancer dormancy in bone marrow. Cancer mediate GRAF-induced RhoA inactivation in a model of breast cancer Res. 2016;76(19):5832–44. dormancy. Cancer Microenviron. 2009;2(1):33–47. 24. Ono M, Kosaka N, Tominaga N, Yoshioka Y, Takeshita F, Takahashi RU, Yoshida M, Tsuda H, Tamura K, Ochiya T. Exosomes from bone marrow mesenchymal stem cells contain a microRNA that promotes dormancy in Publisher’s Note metastatic breast cancer cells. Sci Signal. 2014;7(332):63. Springer Nature remains neutral with regard to jurisdictional claims in pub- 25. Sarkar S, Dey BK, Dutta A. MiR-322/424 and -503 are induced during mus- lished maps and institutional afliations. cle diferentiation and promote cell cycle quiescence and diferentiation by down-regulation of Cdc25A. Mol Biol Cell. 2010;21(13):2138–49. 26. Dioufa N, Clark AM, Ma B, Beckwitt CH, Wells A. Bi-directional exosome- driven intercommunication between the hepatic niche and cancer cells. Mol Cancer. 2017;16(1):172.

Ready to submit your research ? Choose BMC and benefit from:

• fast, convenient online submission • thorough peer review by experienced researchers in your field • rapid publication on acceptance • support for research data, including large and complex data types • gold Open Access which fosters wider collaboration and increased citations • maximum visibility for your research: over 100M website views per year

At BMC, research is always in progress.

Learn more biomedcentral.com/submissions