Cancer Res Treat.  2023 Jul;55(3):720-736. 10.4143/crt.2023.468.

Cellular Dormancy in Cancer: Mechanisms and Potential Targeting Strategies

Affiliations
  • 1College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea

Abstract

Cancer is a leading cause of disease-related mortality worldwide. Drug resistance is one of the primary reasons for the failure of anticancer therapy. There are a number of underlying mechanisms for anticancer drug resistance including genetic/epigenetic modifications, microenvironmental factors, and tumor heterogeneity. In the present scenario, researchers have focused on these novel mechanisms and strategies to tackle them. Recently, researchers have recognized the ability of cancer to become dormant because of anticancer drug resistance, tumor relapse, and progression. Currently, cancer dormancy is classified into “tumor mass dormancy” and “cellular dormancy.” Tumor mass dormancy represents the equilibrium between cell proliferation and cell death under the control of blood supply and immune responses. Cellular dormancy denotes the state in which cells undergo quiescence and is characterized by autophagy, stress-tolerance signaling, microenvironmental cues, and epigenetic modifications. Cancer dormancy has been regarded as the stem of primary or distal recurrent tumor formation and poor clinical outcomes in cancer patients. Despite the insufficiency of reliable models of cellular dormancy, the mechanisms underlying the regulation of cellular dormancy have been clarified in numerous studies. A better understanding of the biology of cancer dormancy is critical for the development of effective anticancer therapeutic strategies. In this review, we summarize the characteristics and regulatory mechanisms of cellular dormancy, introduce several potential strategies for targeting cellular dormancy, and discuss future perspectives.

Keyword

Neoplasms; Dormancy; Recurrence; Cancer progression; Anticancer drug resistance; Cellular dormancy

Figure

  • Fig. 1 Mechanisms underlying maintenance and awakening of dormant cancer cells. Cancer dormancy is classified as tumor mass dormancy and cellular dormancy. Tumor mass dormancy is the equilibrium between cell proliferation and cell death, which is regulated by blood supply and the immune system. Cellular dormancy is the status of reversible growth arrest and characterized by cell cycle arrest at the G0/G1 phase of the cell cycle, induction of CDK inhibitors, reduction of proliferation markers (such as Ki67 and PCNA), p38 MAPK activation, compacted chromatin structure, and reduction of cellular metabolism. Several mechanisms, such as autophagy, stress-tolerance signaling, microenvironmental cues, and epigenetic modifications, are involved in the maintenance of cellular dormancy. Dormant cancer cells escape from the dormant status via changing autocrine soluble factors autonomously and/or interacting with surrounding stromal cells in the microenvironments. CDK, cyclin-dependent kinase; MAPK, mitogen-activated protein kinase; PCNA, proliferating cell nuclear antigen.

  • Fig. 2 Intracellular modulation involved in cellular dormancy. Regulation of cell cycle machinery, such as the Rb-E2F and DREAM complexes, ECM-mediated signal transduction, p38 MAPK activation, growth factors (TGF-β family growth factors and IGFs), and ER stress-induced UPR and acquisition of senescence, autophagy, EMT, and cancer stem cell–associated phenotypes are known to be associated with cellular dormancy. DREAM, dimerization partner, Rb-like, E2F and multi-vulval class B; ECM, extracellular matrix; EMT, epithelial-mesenchymal transition; ER, endoplasmic reticulum; IGF, insulin-like growth factor; MAPK, mitogen-activated protein kinase; Rb, retinoblastoma; TGF-β, transforming growth factor-β; UPR, unfolded protein response.

  • Fig. 3 A proposed mechanism underlying the regulation of dormant cancer cells. Regulation of the UPR machinery for ER homeostasis is essential for the survival of dormant cancer cells in the presence of sustained ER stress, such as chemotherapy. RGS2 plays an important role in the survival of dormant cancer cells by maintaining protein homeostasis against chemotherapy or hostile microenvironments. ATF, activating transcription factor; ER, endoplasmic reticulum; ERAD, ER-associated protein degradation; IRE1, inositol requiring enzyme 1; PERK, protein kinase R-like endoplasmic reticulum kinase; RGS2, regulator of G protein signaling 2; Ub, ubiquitin; UPR, unfolded protein response; XBP1, X-box binding protein 1.

  • Fig. 4 Tumor relapse by awakening slow-cycling/dormant cancer cells through interaction with stromal cells in the tumor microenvironment. Increased recruitment of fibroblasts and vascular endothelial cells under proinflammatory and proliferation-promoting conditions mediated by various cytokines and growth factors stimulate the proliferation of slow-cycling/dormant cancer cells, leading to relapsed tumor formation and cancer progression. ACC, active-cycling cancer cells; CAF, cancer-associated fibroblast; CDK, cyclin-dependent kinase; COX-2, cyclooxygenase 2; EGFR, epidermal growth factor receptor; IL, interleukin; PCNA, proliferating cell nuclear antigen; PGE2, prostaglandin E2; SCC, slow-cycling cancer cells; TGF-β2, transforming growth factor-β2.


Reference

References

1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021; 71:209–49.
2. Wang X, Zhang H, Chen X. Drug resistance and combating drug resistance in cancer. Cancer Drug Resist. 2019; 2:141–60.
3. Ruth JR, Pant DK, Pan TC, Seidel HE, Baksh SC, Keister BA, et al. Cellular dormancy in minimal residual disease following targeted therapy. Breast Cancer Res. 2021; 23:63.
4. Tachtsidis A, McInnes LM, Jacobsen N, Thompson EW, Saunders CM. Minimal residual disease in breast cancer: an overview of circulating and disseminated tumour cells. Clin Exp Metastasis. 2016; 33:521–50.
Article
5. Kang Y, Pantel K. Tumor cell dissemination: emerging biological insights from animal models and cancer patients. Cancer Cell. 2013; 23:573–81.
6. Badia-Ramentol J, Linares J, Gomez-Llonin A, Calon A. Minimal residual disease, metastasis and immunity. Biomolecules. 2021; 11:130.
Article
7. Mohme M, Riethdorf S, Pantel K. Circulating and disseminated tumour cells: mechanisms of immune surveillance and escape. Nat Rev Clin Oncol. 2017; 14:155–67.
8. Yeh AC, Ramaswamy S. Mechanisms of cancer cell dormancy: another hallmark of cancer? Cancer Res. 2015; 75:5014–22.
9. Endo H, Inoue M. Dormancy in cancer. Cancer Sci. 2019; 110:474–80.
10. Goss PE, Chambers AF. Does tumour dormancy offer a therapeutic target? Nat Rev Cancer. 2010; 10:871–7.
Article
11. Damen MP, van Rheenen J, Scheele C. Targeting dormant tumor cells to prevent cancer recurrence. FEBS J. 2021; 288:6286–303.
Article
12. Recasens A, Munoz L. Targeting cancer cell dormancy. Trends Pharmacol Sci. 2019; 40:128–41.
13. Sosa MS, Bragado P, Aguirre-Ghiso JA. Mechanisms of disseminated cancer cell dormancy: an awakening field. Nat Rev Cancer. 2014; 14:611–22.
Article
14. Phan TG, Croucher PI. The dormant cancer cell life cycle. Nat Rev Cancer. 2020; 20:398–411.
Article
15. Aguirre-Ghiso JA. Models, mechanisms and clinical evidence for cancer dormancy. Nat Rev Cancer. 2007; 7:834–46.
Article
16. Indraccolo S, Stievano L, Minuzzo S, Tosello V, Esposito G, Piovan E, et al. Interruption of tumor dormancy by a transient angiogenic burst within the tumor microenvironment. Proc Natl Acad Sci U S A. 2006; 103:4216–21.
17. Swann JB, Smyth MJ. Immune surveillance of tumors. J Clin Invest. 2007; 117:1137–46.
Article
18. Aqbi HF, Wallace M, Sappal S, Payne KK, Manjili MH. IFN-gamma orchestrates tumor elimination, tumor dormancy, tumor escape, and progression. J Leukoc Biol. 2018; 103:1219–23.
19. Correia AL, Guimaraes JC, Auf der Maur P, De Silva D, Trefny MP, Okamoto R, et al. Hepatic stellate cells suppress NK cell-sustained breast cancer dormancy. Nature. 2021; 594:566–71.
Article
20. Liu Y, Liang X, Yin X, Lv J, Tang K, Ma J, et al. Blockade of IDO-kynurenine-AhR metabolic circuitry abrogates IFN-gamma-induced immunologic dormancy of tumor-repopulating cells. Nat Commun. 2017; 8:15207.
21. Muller-Hermelink N, Braumuller H, Pichler B, Wieder T, Mailhammer R, Schaak K, et al. TNFR1 signaling and IFN-gamma signaling determine whether T cells induce tumor dormancy or promote multistage carcinogenesis. Cancer Cell. 2008; 13:507–18.
22. Braumuller H, Wieder T, Brenner E, Assmann S, Hahn M, Alkhaled M, et al. T-helper-1-cell cytokines drive cancer into senescence. Nature. 2013; 494:361–5.
23. Basu S, Dong Y, Kumar R, Jeter C, Tang DG. Slow-cycling (dormant) cancer cells in therapy resistance, cancer relapse and metastasis. Semin Cancer Biol. 2022; 78:90–103.
24. Santos-de-Frutos K, Djouder N. When dormancy fuels tumour relapse. Commun Biol. 2021; 4:747.
Article
25. Townson JL, Chambers AF. Dormancy of solitary metastatic cells. Cell Cycle. 2006; 5:1744–50.
Article
26. Spiliotaki M, Mavroudis D, Kapranou K, Markomanolaki H, Kallergi G, Koinis F, et al. Evaluation of proliferation and apoptosis markers in circulating tumor cells of women with early breast cancer who are candidates for tumor dormancy. Breast Cancer Res. 2014; 16:485.
Article
27. Gilje B, Nordgard O, Tjensvoll K, Janssen EA, Soiland H, Smaaland R, et al. Mitotic activity and bone marrow micrometastases have independent prognostic value in node positive breast cancer patients. Breast Cancer Res Treat. 2011; 128:137–46.
Article
28. Kato TA, Haskins JS. Mitotic index analysis. Methods Mol Biol. 2023; 2519:17–26.
Article
29. Oki T, Nishimura K, Kitaura J, Togami K, Maehara A, Izawa K, et al. A novel cell-cycle-indicator, mVenus-p27K-, identifies quiescent cells and visualizes G0–G1 transition. Sci Rep. 2014; 4:4012.
Article
30. Moore N, Houghton J, Lyle S. Slow-cycling therapy-resistant cancer cells. Stem Cells Dev. 2012; 21:1822–30.
Article
31. Yumoto K, Berry JE, Taichman RS, Shiozawa Y. A novel method for monitoring tumor proliferation in vivo using fluorescent dye DiD. Cytometry A. 2014; 85:548–55.
Article
32. Chauvistre H, Shannan B, Daignault-Mill SM, Ju RJ, Picard D, Egetemaier S, et al. Persister state-directed transitioning and vulnerability in melanoma. Nat Commun. 2022; 13:3055.
33. Tario JD Jr, Humphrey K, Bantly AD, Muirhead KA, Moore JS, Wallace PK. Optimized staining and proliferation modeling methods for cell division monitoring using cell tracking dyes. J Vis Exp. 2012; e4287.
34. Ebinger S, Ozdemir EZ, Ziegenhain C, Tiedt S, Castro Alves C, Grunert M, et al. Characterization of rare, dormant, and therapy-resistant cells in acute lymphoblastic leukemia. Cancer Cell. 2016; 30:849–62.
35. Preciado JA, Aksan A. Method to isolate dormant cancer cells from heterogeneous populations. Methods Mol Biol. 2022; 2394:19–29.
Article
36. Pradhan S, Sperduto JL, Farino CJ, Slater JH. Engineered in vitro models of tumor dormancy and reactivation. J Biol Eng. 2018; 12:37.
Article
37. Gu Y, Bui T, Muller WJ. Exploiting mouse models to recapitulate clinical tumor dormancy and recurrence in breast cancer. Endocrinology. 2022; 163:bqac055.
38. Sosa MS, Avivar-Valderas A, Bragado P, Wen HC, Aguirre-Ghiso JA. ERK1/2 and p38alpha/beta signaling in tumor cell quiescence: opportunities to control dormant residual disease. Clin Cancer Res. 2011; 17:5850–7.
39. Aguirre-Ghiso JA, Estrada Y, Liu D, Ossowski L. ERK(MAPK) activity as a determinant of tumor growth and dormancy; regulation by p38(SAPK). Cancer Res. 2003; 63:1684–95.
Article
40. Barney LE, Hall CL, Schwartz AD, Parks AN, Sparages C, Galarza S, et al. Tumor cell-organized fibronectin maintenance of a dormant breast cancer population. Sci Adv. 2020; 6:eaaz4157.
Article
41. Rehman SK, Haynes J, Collignon E, Brown KR, Wang Y, Nixon AM, et al. Colorectal cancer cells enter a diapause-like DTP state to survive chemotherapy. Cell. 2021; 184:226–42.
42. Cho J, Min HY, Lee HJ, Hyun SY, Sim JY, Noh M, et al. RGS2-mediated translational control mediates cancer cell dormancy and tumor relapse. J Clin Invest. 2021; 131:e136779.
Article
43. Pranzini E, Raugei G, Taddei ML. Metabolic features of tumor dormancy: possible therapeutic strategies. Cancers (Basel). 2022; 14:547.
Article
44. Valcourt JR, Lemons JM, Haley EM, Kojima M, Demuren OO, Coller HA. Staying alive: metabolic adaptations to quiescence. Cell Cycle. 2012; 11:1680–96.
45. Ranganathan AC, Adam AP, Zhang L, Aguirre-Ghiso JA. Tumor cell dormancy induced by p38SAPK and ER-stress signaling: an adaptive advantage for metastatic cells? Cancer Biol Ther. 2006; 5:729–35.
Article
46. Evertts AG, Manning AL, Wang X, Dyson NJ, Garcia BA, Coller HA. H4K20 methylation regulates quiescence and chromatin compaction. Mol Biol Cell. 2013; 24:3025–37.
Article
47. Milanovic M, Fan DN, Belenki D, Dabritz JH, Zhao Z, Yu Y, et al. Senescence-associated reprogramming promotes cancer stemness. Nature. 2018; 553:96–100.
Article
48. Truskowski K, Amend SR, Pienta KJ. Dormant cancer cells: programmed quiescence, senescence, or both? Cancer Meta-stasis Rev. 2023; 42:37–47.
49. Schmitt CA, Wang B, Demaria M. Senescence and cancer: role and therapeutic opportunities. Nat Rev Clin Oncol. 2022; 19:619–36.
50. Iwasa H, Han J, Ishikawa F. Mitogen-activated protein kinase p38 defines the common senescence-signalling pathway. Genes Cells. 2003; 8:131–44.
Article
51. Prunier C, Alay A, van Dijk M, Ammerlaan KL, van Gelderen S, Marvin DL, et al. Breast cancer dormancy is associated with a 4NG1 state and not senescence. NPJ Breast Cancer. 2021; 7:140.
52. Triana-Martinez F, Loza MI, Dominguez E. Beyond tumor suppression: senescence in cancer stemness and tumor dormancy. Cells. 2020; 9:346.
Article
53. Saleh T, Tyutyunyk-Massey L, Gewirtz DA. Tumor cell escape from therapy-induced senescence as a model of disease recurrence after dormancy. Cancer Res. 2019; 79:1044–6.
Article
54. Plaks V, Kong N, Werb Z. The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell Stem Cell. 2015; 16:225–38.
Article
55. Desai A, Yan Y, Gerson SL. Concise reviews: cancer stem cell targeted therapies: toward clinical success. Stem Cells Transl Med. 2019; 8:75–81.
Article
56. Pattabiraman DR, Weinberg RA. Tackling the cancer stem cells: what challenges do they pose? Nat Rev Drug Discov. 2014; 13:497–512.
Article
57. Medema JP. Cancer stem cells: the challenges ahead. Nat Cell Biol. 2013; 15:338–44.
58. Lambert AW, Weinberg RA. Linking EMT programmes to normal and neoplastic epithelial stem cells. Nat Rev Cancer. 2021; 21:325–38.
Article
59. Weidenfeld K, Barkan D. EMT and stemness in tumor dormancy and outgrowth: are they intertwined processes? Front Oncol. 2018; 8:381.
60. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008; 133:704–15.
61. Pradella D, Naro C, Sette C, Ghigna C. EMT and stemness: flexible processes tuned by alternative splicing in development and cancer progression. Mol Cancer. 2017; 16:8.
Article
62. Wang SS, Jiang J, Liang XH, Tang YL. Links between cancer stem cells and epithelial-mesenchymal transition. Onco Targets Ther. 2015; 8:2973–80.
63. Ocana OH, Corcoles R, Fabra A, Moreno-Bueno G, Acloque H, Vega S, et al. Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell. 2012; 22:709–24.
Article
64. Jia Q, Yang F, Huang W, Zhang Y, Bao B, Li K, et al. Low Levels of Sox2 are required for melanoma tumor-repopulating cell dormancy. Theranostics. 2019; 9:424–35.
Article
65. Ohta Y, Fujii M, Takahashi S, Takano A, Nanki K, Matano M, et al. Cell-matrix interface regulates dormancy in human colon cancer stem cells. Nature. 2022; 608:784–94.
Article
66. Zhou N, Wu X, Yang B, Yang X, Zhang D, Qing G. Stem cell characteristics of dormant cells and cisplatin-induced effects on the stemness of epithelial ovarian cancer cells. Mol Med Rep. 2014; 10:2495–504.
Article
67. Fischer M, Schade AE, Branigan TB, Muller GA, DeCaprio JA. Coordinating gene expression during the cell cycle. Trends Biochem Sci. 2022; 47:1009–22.
Article
68. Giacinti C, Giordano A. RB and cell cycle progression. Oncogene. 2006; 25:5220–7.
Article
69. Kwon JS, Everetts NJ, Wang X, Wang W, Della Croce K, Xing J, et al. Controlling depth of cellular quiescence by an Rb-E2F network switch. Cell Rep. 2017; 20:3223–35.
Article
70. Sadasivam S, DeCaprio JA. The DREAM complex: master coordinator of cell cycle-dependent gene expression. Nat Rev Cancer. 2013; 13:585–95.
Article
71. Litovchick L, Florens LA, Swanson SK, Washburn MP, DeCaprio JA. DYRK1A protein kinase promotes quiescence and senescence through DREAM complex assembly. Genes Dev. 2011; 25:801–13.
Article
72. MacDonald J, Ramos-Valdes Y, Perampalam P, Litovchick L, DiMattia GE, Dick FA. A systematic analysis of negative growth control implicates the DREAM complex in cancer cell dormancy. Mol Cancer Res. 2017; 15:371–81.
Article
73. Guiley KZ, Liban TJ, Felthousen JG, Ramanan P, Litovchick L, Rubin SM. Structural mechanisms of DREAM complex assembly and regulation. Genes Dev. 2015; 29:961–74.
Article
74. Schade AE, Oser MG, Nicholson HE, DeCaprio JA. Cyclin D-CDK4 relieves cooperative repression of proliferation and cell cycle gene expression by DREAM and RB. Oncogene. 2019; 38:4962–76.
75. Boichuk S, Parry JA, Makielski KR, Litovchick L, Baron JL, Zewe JP, et al. The DREAM complex mediates GIST cell quiescence and is a novel therapeutic target to enhance imatinib-induced apoptosis. Cancer Res. 2013; 73:5120–9.
Article
76. Wang P, Karakose E, Argmann C, Wang H, Balev M, Brody RI, et al. Disrupting the DREAM complex enables proliferation of adult human pancreatic beta cells. J Clin Invest. 2022; 132:e157086.
77. Kim MJ, Cervantes C, Jung YS, Zhang X, Zhang J, Lee SH, et al. PAF remodels the DREAM complex to bypass cell quiescence and promote lung tumorigenesis. Mol Cell. 2021; 81:1698–714.
Article
78. Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. J Cell Sci. 2010; 123:4195–200.
Article
79. Yue B. Biology of the extracellular matrix: an overview. J Glaucoma. 2014; 23(8 Suppl 1):S20–3.
80. Hynes RO, Naba A. Overview of the matrisome: an inventory of extracellular matrix constituents and functions. Cold Spring Harb Perspect Biol. 2012; 4:a004903.
81. Naba A, Clauser KR, Ding H, Whittaker CA, Carr SA, Hynes RO. The extracellular matrix: Tools and insights for the “omics” era. Matrix Biol. 2016; 49:10–24.
Article
82. Winkler J, Abisoye-Ogunniyan A, Metcalf KJ, Werb Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat Commun. 2020; 11:5120.
Article
83. Wei Y, Lukashev M, Simon DI, Bodary SC, Rosenberg S, Doyle MV, et al. Regulation of integrin function by the urokinase receptor. Science. 1996; 273:1551–5.
Article
84. Aguirre Ghiso JA, Kovalski K, Ossowski L. Tumor dormancy induced by downregulation of urokinase receptor in human carcinoma involves integrin and MAPK signaling. J Cell Biol. 1999; 147:89–104.
Article
85. Aguirre-Ghiso JA, Liu D, Mignatti A, Kovalski K, Ossow-ski L. Urokinase receptor and fibronectin regulate the ERK(MAPK) to p38(MAPK) activity ratios that determine carcinoma cell proliferation or dormancy in vivo. Mol Biol Cell. 2001; 12:863–79.
86. Barkan D, Kleinman H, Simmons JL, Asmussen H, Kamaraju AK, Hoenorhoff MJ, et al. Inhibition of metastatic outgrowth from single dormant tumor cells by targeting the cytoskeleton. Cancer Res. 2008; 68:6241–50.
Article
87. Barkan D, El Touny LH, Michalowski AM, Smith JA, Chu I, Davis AS, et al. Metastatic growth from dormant cells induced by a col-I-enriched fibrotic environment. Cancer Res. 2010; 70:5706–16.
Article
88. Cho J, Lee HJ, Hwang SJ, Min HY, Kang HN, Park AY, et al. The interplay between slow-cycling, chemoresistant cancer cells and fibroblasts creates a proinflammatory niche for tumor progression. Cancer Res. 2020; 80:2257–72.
Article
89. Keeratichamroen S, Lirdprapamongkol K, Svasti J. Mechanism of ECM-induced dormancy and chemoresistance in A549 human lung carcinoma cells. Oncol Rep. 2018; 39:1765–74.
Article
90. Di Martino JS, Nobre AR, Mondal C, Taha I, Farias EF, Fertig EJ, et al. A tumor-derived type III collagen-rich ECM niche regulates tumor cell dormancy. Nat Cancer. 2022; 3:90–107.
Article
91. Almog N, Ma L, Raychowdhury R, Schwager C, Erber R, Short S, et al. Transcriptional switch of dormant tumors to fast-growing angiogenic phenotype. Cancer Res. 2009; 69:836–44.
Article
92. Kazerounian S, Yee KO, Lawler J. Thrombospondins in cancer. Cell Mol Life Sci. 2008; 65:700–12.
93. Rouanne M, Adam J, Goubar A, Robin A, Ohana C, Louvet E, et al. Osteopontin and thrombospondin-1 play opposite roles in promoting tumor aggressiveness of primary resected non-small cell lung cancer. BMC Cancer. 2016; 16:483.
Article
94. Boyerinas B, Zafrir M, Yesilkanal AE, Price TT, Hyjek EM, Sipkins DA. Adhesion to osteopontin in the bone marrow niche regulates lymphoblastic leukemia cell dormancy. Blood. 2013; 121:4821–31.
Article
95. Parker AL, Cox TR. The role of the ECM in lung cancer dormancy and outgrowth. Front Oncol. 2020; 10:1766.
Article
96. Rodrigues LR, Teixeira JA, Schmitt FL, Paulsson M, Lindmark-Mansson H. The role of osteopontin in tumor progression and metastasis in breast cancer. Cancer Epidemiol Biomarkers Prev. 2007; 16:1087–97.
97. Jiang Y, Zhang H, Wang J, Liu Y, Luo T, Hua H. Targeting extracellular matrix stiffness and mechanotransducers to improve cancer therapy. J Hematol Oncol. 2022; 15:34.
Article
98. Doue M, Okwieka A, Berquand A, Gorisse L, Maurice P, Velard F, et al. Carbamylation of elastic fibers is a molecular substratum of aortic stiffness. Sci Rep. 2021; 11:17827.
99. Schrader J, Gordon-Walker TT, Aucott RL, van Deemter M, Quaas A, Walsh S, et al. Matrix stiffness modulates proliferation, chemotherapeutic response, and dormancy in hepatocellular carcinoma cells. Hepatology. 2011; 53:1192–205.
Article
100. Kondapaneni RV, Rao SS. Matrix stiffness and cluster size collectively regulate dormancy versus proliferation in brain metastatic breast cancer cell clusters. Biomater Sci. 2020; 8:6637–46.
101. Anlas AA, Nelson CM. Soft microenvironments induce chemoresistance by increasing autophagy downstream of integrin-linked kinase. Cancer Res. 2020; 80:4103–13.
102. Liu Y, Lv J, Liang X, Yin X, Zhang L, Chen D, et al. Fibrin stiffness mediates dormancy of tumor-repopulating cells via a Cdc42-driven Tet2 epigenetic program. Cancer Res. 2018; 78:3926–37.
Article
103. Sistigu A, Musella M, Galassi C, Vitale I, De Maria R. Tuning cancer fate: tumor microenvironment’s role in cancer stem cell quiescence and reawakening. Front Immunol. 2020; 11:2166.
Article
104. Butturini E, Carcereri de Prati A, Boriero D, Mariotto S. Tumor dormancy and interplay with hypoxic tumor microenvironment. Int J Mol Sci. 2019; 20:4305.
Article
105. Ju S, Wang F, Wang Y, Ju S. CSN8 is a key regulator in hypoxia-induced epithelial-mesenchymal transition and dormancy of colorectal cancer cells. Mol Cancer. 2020; 19:168.
106. Ameri K, Jahangiri A, Rajah AM, Tormos KV, Nagarajan R, Pekmezci M, et al. HIGD1A regulates oxygen consumption, ROS production, and AMPK activity during glucose deprivation to modulate cell survival and tumor growth. Cell Rep. 2015; 10:891–9.
Article
107. Bildik G, Liang X, Sutton MN, Bast RC Jr, Lu Z. DIRAS3: an imprinted tumor suppressor gene that regulates RAS and PI3K-driven cancer growth, motility, autophagy, and tumor dormancy. Mol Cancer Ther. 2022; 21:25–37.
Article
108. Sutton MN, Huang GY, Zhou J, Mao W, Langley R, Lu Z, et al. Amino acid deprivation-induced autophagy requires upregulation of DIRAS3 through reduction of E2F1 and E2F4 transcriptional repression. Cancers (Basel). 2019; 11:603.
Article
109. Wang L, Shang Z, Zhou Y, Hu X, Chen Y, Fan Y, et al. Autophagy mediates glucose starvation-induced glioblastoma cell quiescence and chemoresistance through coordinating cell metabolism, cell cycle, and survival. Cell Death Dis. 2018; 9:213.
Article
110. Zhang M, Peng R, Wang H, Yang Z, Zhang H, Zhang Y, et al. Nanog mediated by FAO/ACLY signaling induces cellular dormancy in colorectal cancer cells. Cell Death Dis. 2022; 13:159.
Article
111. Dong X, Xue H, Mo F, Lin YY, Lin D, Wong NKY, et al. Modeling androgen deprivation therapy-induced prostate cancer dormancy and its clinical implications. Mol Cancer Res. 2022; 20:782–93.
Article
112. Kurppa KJ, Liu Y, To C, Zhang T, Fan M, Vajdi A, et al. Treatment-induced tumor dormancy through YAP-mediated transcriptional reprogramming of the apoptotic pathway. Cancer Cell. 2020; 37:104–22.
Article
113. Min M, Spencer SL. Spontaneously slow-cycling subpopulations of human cells originate from activation of stress-response pathways. PLoS Biol. 2019; 17:e3000178.
Article
114. Wagner EF, Nebreda AR. Signal integration by JNK and p38 MAPK pathways in cancer development. Nat Rev Cancer. 2009; 9:537–49.
Article
115. Adam AP, George A, Schewe D, Bragado P, Iglesias BV, Ranganathan AC, et al. Computational identification of a p38SAPK-regulated transcription factor network required for tumor cell quiescence. Cancer Res. 2009; 69:5664–72.
Article
116. Sosa MS, Parikh F, Maia AG, Estrada Y, Bosch A, Bragado P, et al. NR2F1 controls tumour cell dormancy via SOX9- and RARbeta-driven quiescence programmes. Nat Commun. 2015; 6:6170.
117. Cai B, Chang SH, Becker EB, Bonni A, Xia Z. p38 MAP kinase mediates apoptosis through phosphorylation of BimEL at Ser-65. J Biol Chem. 2006; 281:25215–22.
Article
118. Zhuang S, Demirs JT, Kochevar IE. p38 mitogen-activated protein kinase mediates bid cleavage, mitochondrial dysfunction, and caspase-3 activation during apoptosis induced by singlet oxygen but not by hydrogen peroxide. J Biol Chem. 2000; 275:25939–48.
Article
119. Sui X, Kong N, Ye L, Han W, Zhou J, Zhang Q, et al. p38 and JNK MAPK pathways control the balance of apoptosis and autophagy in response to chemotherapeutic agents. Cancer Lett. 2014; 344:174–9.
120. Kroeger H, Grimsey N, Paxman R, Chiang WC, Plate L, Jones Y, et al. The unfolded protein response regulator ATF6 promotes mesodermal differentiation. Sci Signal. 2018; 11:eaan5785.
Article
121. Fu Y, Li J, Lee AS. GRP78/BiP inhibits endoplasmic reticulum BIK and protects human breast cancer cells against estrogen starvation-induced apoptosis. Cancer Res. 2007; 67:3734–40.
Article
122. Schewe DM, Aguirre-Ghiso JA. ATF6alpha-Rheb-mTOR signaling promotes survival of dormant tumor cells in vivo. Proc Natl Acad Sci U S A. 2008; 105:10519–24.
123. Gawrzak S, Rinaldi L, Gregorio S, Arenas EJ, Salvador F, Urosevic J, et al. MSK1 regulates luminal cell differentiation and metastatic dormancy in ER(+) breast cancer. Nat Cell Biol. 2018; 20:211–21.
124. Aman Y, Schmauck-Medina T, Hansen M, Morimoto RI, Simon AK, Bjedov I, et al. Autophagy in healthy aging and disease. Nat Aging. 2021; 1:634–50.
Article
125. Akkoc Y, Peker N, Akcay A, Gozuacik D. Autophagy and cancer dormancy. Front Oncol. 2021; 11:627023.
Article
126. Vera-Ramirez L. Cell-intrinsic survival signals. The role of autophagy in metastatic dissemination and tumor cell dormancy. Semin Cancer Biol. 2020; 60:28–40.
Article
127. Chen Y, Gibson SB. Three dimensions of autophagy in regulating tumor growth: cell survival/death, cell proliferation, and tumor dormancy. Biochim Biophys Acta Mol Basis Dis. 2021; 1867:166265.
Article
128. Gupta A, Roy S, Lazar AJ, Wang WL, McAuliffe JC, Reynoso D, et al. Autophagy inhibition and antimalarials promote cell death in gastrointestinal stromal tumor (GIST). Proc Natl Acad Sci U S A. 2010; 107:14333–8.
Article
129. Vera-Ramirez L, Vodnala SK, Nini R, Hunter KW, Green JE. Autophagy promotes the survival of dormant breast cancer cells and metastatic tumour recurrence. Nat Commun. 2018; 9:1944.
Article
130. Lu Z, Luo RZ, Lu Y, Zhang X, Yu Q, Khare S, et al. The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. J Clin Invest. 2008; 118:3917–29.
131. Lu Z, Baquero MT, Yang H, Yang M, Reger AS, Kim C, et al. DIRAS3 regulates the autophagosome initiation complex in dormant ovarian cancer cells. Autophagy. 2014; 10:1071–92.
Article
132. Weiss A, Attisano L. The TGFbeta superfamily signaling pathway. Wiley Interdiscip Rev Dev Biol. 2013; 2:47–63.
Article
133. Bragado P, Estrada Y, Parikh F, Krause S, Capobianco C, Farina HG, et al. TGF-beta2 dictates disseminated tumour cell fate in target organs through TGF-beta-RIII and p38alpha/beta signalling. Nat Cell Biol. 2013; 15:1351–61.
134. Yumoto K, Eber MR, Wang J, Cackowski FC, Decker AM, Lee E, et al. Axl is required for TGF-beta2-induced dormancy of prostate cancer cells in the bone marrow. Sci Rep. 2016; 6:36520.
135. Kobayashi A, Okuda H, Xing F, Pandey PR, Watabe M, Hirota S, et al. Bone morphogenetic protein 7 in dormancy and metastasis of prostate cancer stem-like cells in bone. J Exp Med. 2011; 208:2641–55.
136. Pollak MN, Schernhammer ES, Hankinson SE. Insulin-like growth factors and neoplasia. Nat Rev Cancer. 2004; 4:505–18.
Article
137. Rajbhandari N, Lin WC, Wehde BL, Triplett AA, Wagner KU. Autocrine IGF1 signaling mediates pancreatic tumor cell dormancy in the absence of oncogenic drivers. Cell Rep. 2017; 18:2243–55.
138. Shimizu T, Sugihara E, Yamaguchi-Iwai S, Tamaki S, Koyama Y, Kamel W, et al. IGF2 preserves osteosarcoma cell survival by creating an autophagic state of dormancy that protects cells against chemotherapeutic stress. Cancer Res. 2014; 74:6531–41.
Article
139. Worster DT, Schmelzle T, Solimini NL, Lightcap ES, Millard B, Mills GB, et al. Akt and ERK control the proliferative response of mammary epithelial cells to the growth factors IGF-1 and EGF through the cell cycle inhibitor p57Kip2. Sci Signal. 2012; 5:ra19.
140. Mercer TR, Mattick JS. Structure and function of long noncoding RNAs in epigenetic regulation. Nat Struct Mol Biol. 2013; 20:300–7.
Article
141. Ruksha TG. MicroRNAs’ control of cancer cell dormancy. Cell Div. 2019; 14:11.
Article
142. Nishikawa S, Dewi DL, Ishii H, Konno M, Haraguchi N, Kano Y, et al. Transcriptomic study of dormant gastrointestinal cancer stem cells. Int J Oncol. 2012; 41:979–84.
143. Almog N, Briggs C, Beheshti A, Ma L, Wilkie KP, Rietman E, et al. Transcriptional changes induced by the tumor dormancy-associated microRNA-190. Transcription. 2013; 4:177–91.
Article
144. Vera-Ramirez L, Hunter KW. Tumor cell dormancy as an adaptive cell stress response mechanism. F1000Res. 2017; 6:2134.
Article
145. Schonthal AH. Endoplasmic reticulum stress: its role in disease and novel prospects for therapy. Scientifica (Cairo). 2012; 2012:857516.
146. Wang M, Kaufman RJ. The impact of the endoplasmic reticulum protein-folding environment on cancer development. Nat Rev Cancer. 2014; 14:581–97.
Article
147. Hetz C. The unfolded protein response: controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Cell Biol. 2012; 13:89–102.
148. Lee K, Tirasophon W, Shen X, Michalak M, Prywes R, Okada T, et al. IRE1-mediated unconventional mRNA splicing and S2P-mediated ATF6 cleavage merge to regulate XBP1 in signaling the unfolded protein response. Genes Dev. 2002; 16:452–66.
Article
149. Maurel M, Chevet E, Tavernier J, Gerlo S. Getting RIDD of RNA: IRE1 in cell fate regulation. Trends Biochem Sci. 2014; 39:245–54.
Article
150. Han J, Back SH, Hur J, Lin YH, Gildersleeve R, Shan J, et al. ER-stress-induced transcriptional regulation increases protein synthesis leading to cell death. Nat Cell Biol. 2013; 15:481–90.
151. Bartkowiak K, Kwiatkowski M, Buck F, Gorges TM, Nilse L, Assmann V, et al. Disseminated tumor cells persist in the bone marrow of breast cancer patients through sustained activation of the unfolded protein response. Cancer Res. 2015; 75:5367–77.
Article
152. Pommier A, Anaparthy N, Memos N, Kelley ZL, Gouronnec A, Yan R, et al. Unresolved endoplasmic reticulum stress engenders immune-resistant, latent pancreatic cancer metastases. Science. 2018; 360:eaao4908.
Article
153. Ranganathan AC, Zhang L, Adam AP, Aguirre-Ghiso JA. Functional coupling of p38-induced up-regulation of BiP and activation of RNA-dependent protein kinase-like endoplasmic reticulum kinase to drug resistance of dormant carcinoma cells. Cancer Res. 2006; 66:1702–11.
Article
154. Jaud M, Philippe C, Di Bella D, Tang W, Pyronnet S, Laurell H, et al. Translational regulations in response to endoplasmic reticulum stress in cancers. Cells. 2020; 9:540.
Article
155. Kehrl JH, Sinnarajah S. RGS2: a multifunctional regulator of G-protein signaling. Int J Biochem Cell Biol. 2002; 34:432–8.
Article
156. Nguyen CH, Ming H, Zhao P, Hugendubler L, Gros R, Kimball SR, et al. Translational control by RGS2. J Cell Biol. 2009; 186:755–65.
Article
157. Zmijewski JW, Song L, Harkins L, Cobbs CS, Jope RS. Oxidative stress and heat shock stimulate RGS2 expression in 1321N1 astrocytoma cells. Arch Biochem Biophys. 2001; 392:192–6.
158. Ota A, Sawai M, Sakurai H. Stress-induced transcription of regulator of G protein signaling 2 (RGS2) by heat shock transcription factor HSF1. Biochimie. 2013; 95:1432–6.
Article
159. Nguyen CH, Zhao P, Sobiesiak AJ, Chidiac P. RGS2 is a component of the cellular stress response. Biochem Biophys Res Commun. 2012; 426:129–34.
Article
160. Park SY, Nam JS. The force awakens: metastatic dormant cancer cells. Exp Mol Med. 2020; 52:569–81.
Article
161. Adamski V, Hattermann K, Kubelt C, Cohrs G, Lucius R, Synowitz M, et al. Entry and exit of chemotherapeutically-promoted cellular dormancy in glioblastoma cells is differentially affected by the chemokines CXCL12, CXCL16, and CX3CL1. Oncogene. 2020; 39:4421–35.
162. Hirano R, Okamoto K, Shinke M, Sato M, Watanabe S, Watanabe H, et al. Tissue-resident macrophages are major tumor-associated macrophage resources, contributing to early TNBC development, recurrence, and metastases. Commun Biol. 2023; 6:144.
Article
163. Liu H, Ling CC, Yeung WH, Pang L, Liu J, Zhou J, et al. Monocytic MDSC mobilization promotes tumor recurrence after liver transplantation via CXCL10/TLR4/MMP14 signaling. Cell Death Dis. 2021; 12:489.
Article
164. Albrengues J, Shields MA, Ng D, Park CG, Ambrico A, Poindexter ME, et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science. 2018; 361:eaao4227.
Article
165. Hast J, Schiffer IB, Neugebauer B, Teichman E, Schreiber W, Brieger J, et al. Angiogenesis and fibroblast proliferation precede formation of recurrent tumors after radiation therapy in nude mice. Anticancer Res. 2002; 22:677–88.
166. Cho J, Min HY, Pei H, Wei X, Sim JY, Park SH, et al. The ATF6-EGF pathway mediates the awakening of slow-cycling chemoresistant cells and tumor recurrence by stimulating tumor angiogenesis. Cancers (Basel). 2020; 12:1772.
Article
167. Huang X, Wang L, Guo H, Zhang W, Shao Z. Single-cell transcriptomics reveals the regulative roles of cancer associated fibroblasts in tumor immune microenvironment of recurrent osteosarcoma. Theranostics. 2022; 12:5877–87.
168. Bushnell GG, Deshmukh AP, den Hollander P, Luo M, Soundararajan R, Jia D, et al. Breast cancer dormancy: need for clinically relevant models to address current gaps in knowledge. NPJ Breast Cancer. 2021; 7:66.
Article
169. Heidrich I, Deitert B, Werner S, Pantel K. Liquid biopsy for monitoring of tumor dormancy and early detection of disease recurrence in solid tumors. Cancer Metastasis Rev. 2023; 42:161–82.
Article
170. Morin C, Moyret-Lalle C, Mertani HC, Diaz JJ, Marcel V. Heterogeneity and dynamic of EMT through the plasticity of ribosome and mRNA translation. Biochim Biophys Acta Rev Cancer. 2022; 1877:188718.
Article
171. Brown MS, Abdollahi B, Wilkins OM, Lu H, Chakraborty P, Ognjenovic NB, et al. Phenotypic heterogeneity driven by plasticity of the intermediate EMT state governs disease progression and metastasis in breast cancer. Sci Adv. 2022; 8:eabj8002.
Article
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr