Review Open Access Logo

Cancer cell dormancy: An update to 2025

Bui Dinh Khan 1, 2, *
Nguyen Thi Yen Nhi 1, 2
Tran Ngo The Nhan 1, 2
Pham Duy Khuong 2
  1. VNUHCM-US Stem Cell Institute, University of Science, Viet Nam National University Ho Chi Minh City, Viet Nam
  2. Vietnam National University Ho Chi Minh City, Viet Nam
Correspondence to: Bui Dinh Khan, VNUHCM-US Stem Cell Institute, University of Science, Viet Nam National University Ho Chi Minh City, Viet Nam; Vietnam National University Ho Chi Minh City, Viet Nam. Email: [email protected].
Volume & Issue: Vol. 12 No. 7 (2025) | Page No.: 7559-7575 | DOI: 10.15419/ttm97s19
Published: 2025-07-31

Online metrics


Statistics from the website

  • HTML Views: 125
  • PDF Views: 26
  • XML Views: 1

Statistics from Dimensions

This article is published with open access by BioMedPress. This article is distributed under the terms of the Creative Commons Attribution License (CC-BY 4.0) which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited. 

Abstract

About 70 years ago, scientists first observed groups of cancer cells in a “temporary mitotic arrest,” a dormant state that complicates treatment and increases the risk of recurrence. Recent updates have provided novel insights into the mechanisms driving cancer cell dormancy, especially in relation to how dormant cells evolve and develop resistance to treatments over time. This phenomenon is particularly concerning in breast cancer, where dormant cells can 'wake up' after extended periods, contributing to cancer relapse. Dormancy, akin to hibernation in animals, occurs when cancer cells enter a resting phase (G0/G1 phase) in response to stressors like nutrient deprivation or hypoxia. Key signaling pathways have been identified that regulate the balance between proliferation and dormancy, with some pathways playing critical roles in maintaining dormancy for years. Notably, cancer dormancy has been linked to enhanced stemness and increased resistance to therapies, making drug resistance a significant challenge. Despite promising advancements, existing strategies to target dormant cancer cells have not yet achieved complete eradication, leaving surviving cells that can trigger relapse. A particularly important future direction is the development of combination therapies, which hold potential for preventing recurrence and improving patient outcomes by targeting multiple mechanisms that govern dormancy and reactivation.

Introduction

According to statistics from the Global Cancer Observatory (GLOBOCAN) in 2022, the global incidence of cancer is on an upward trend. Nearly 20 million new cancer cases were reported, and the number of deaths reached approximately 10 million1. Numerous studies have been carried out to identify the underlying causes responsible for the severity and lethality of cancer.

The observation of tumor recurrence in the human body dates back not only to modern times but also to the early centuries A.D. In De medicina, a medical treatise compiled by the ancient Roman encyclopedist Aulus Cornelius Celsus, it was noted that numerous patients experienced the redevelopment of malignant tumors following a period of remission after surgical excision2. In 1934, Rupert Allan Willis first hypothesized that cancer cells stayed silent in tissues where they were localized, as he observed delayed metastases in patients who showed no local recurrence following the removal of their primary tumor3. In 1954, Geoffrey Hadfield proposed that cancer cells, intentionally stalling their activities, entered a phase known as “temporary mitotic arrest”4. Gimbrone (1972) demonstrated that tumor dormancy can be maintained by preventing neovascularization, highlighting angiogenesis as a critical switch for tumor progression5. This dormant state renders cancer cells difficult to be targeted and eradicated. Recently, using single-cell RNA-sequencing technology, Wang (2022) confirmed that nearly 400 genes exhibit altered expression in dormant cancer cells residing within bone niches6. In addition, a multiomics analysis by Laguillaume (2024) provided insights into the genetic and non-genetic mechanisms governing cancer cell dormancy, highlighting both the complexity and cancer type-specificity of this phenomenon. The identification of both shared and distinct molecular signatures across two models lays a foundation for developing therapeutic strategies aimed at eliminating dormant cells to prevent recurrence and metastasis7. Over time, dormancy allows cancer cells to evolve into populations resistant to current therapies. These findings have established the foundation for oncologists’ understanding of dormant cancer cells as a potential threat to patients when these cells become reactivated (Figure 1).

The concept of cancer dormancy first emerged from German pathologist Rudolf Virchow’s clinical observations in the late nineteenth century, when he noted that some malignant cells survived post-treatment and later gave rise to recurrent tumors with similar histological traits8. In the 1930s and 1950s, early concepts describing non-proliferative or slowly cycling cancer cell populations were proposed3, 4. Gimbrone (1972) and Folkman . (1995) summarized that tumor dormancy depends on suppressed angiogenesis, suggesting that reactivation and progression require neovascularization, thereby identifying angiogenesis as a key therapeutic target5, 9. Since the 2000s, in-depth studies have uncovered molecular and cellular mechanisms underlying the dormant state of cancer cells, emphasizing the pivotal roles of microenvironment signals and the immune system, and suggesting potential biomarkers for precision medicine in diagnosis and relapse prevention10, 11, 12, 13.

In this review, we primarily focus on the signaling pathways that lead cancer cells into a dormant state. Also, we discuss how dormant cancer cells are reawakened and examine the similarities between dormant cancer cells and cancer stem cells. Lastly, this review highlights novel strategies targeting dormant cells, which have been explored in , , and clinical settings. However, further research is needed to gain a deeper understanding of the mechanisms involved in cancer dormancy, to develop more precise treatment, and to prevent tumor relapse in the future.

Figure 1

A brief history of studies in cancer dormancy.

Current Understanding of the Mechanisms That Maintain Dormancy in Cancer

After observing dormant populations of cancer cells in breast cancer patients, Retsky . (2005) reported that cancer recurrence arises from the reactivation of these dormant cells14. Mechanistically, this unique dormant state mirrors the hibernation mechanism seen in certain animals exposed to harsh conditions in temperate and cold regions15. Cancer cell dormancy can be understood as an intermediary between the G0 and G1 phases of the cell cycle—a “resting” state in which they restrict metabolism and proliferation12, 16. To enter this silent state, stress factors such as oxygen deprivation, limited nutrients, or chemical stress prompt changes in intracellular metabolism, allowing cancer cells to endure adverse conditions without being destroyed12.

Most researchers maintain that activation of the extracellular signal-regulating kinases (ERKs) plays a decisive role in determining whether cells proliferate or enter a dormant state. Additionally, p38, a member of the mitogen-activated protein kinase (MAPK) family, is vital in regulating both normal cell survival and cancer cell behavior17. When excessive cell proliferation is triggered by extracellular ERK signaling, the p38 kinase becomes activated to counterbalance ERK activity, driving the cell into an intermediate state within the G0/G1 phase. The upregulation of ERK and p38 MAPKs is considered the first "golden" signal that influences both cell proliferation and dormancy18. While ERK phosphorylation promotes proliferation, p38 phosphorylation opposes it by inducing cellular dormancy. Therefore, a lower ERK/p38 expression ratio is an indicator of the dormant state in cancer cells. Furthermore, proteins encoded by the F-box and WD repeat domain containing 7 (FBXW7) gene regulate mitotic activity by targeting key proteins such as cyclin E and c-Myc for degradation19, 20, thereby suppressing proliferation and maintaining quiescence21. These findings underscore the importance of cell cycle-regulating signaling pathways in controlling dormancy.

Tumor growth factor beta (TGF-β) stimulates the expression of cell cycle-related and dormancy-related genes in cancer cells such as p15, p21, and p2722. Studies by Bragado . (2015) and Sosa (2013) demonstrated that TGF-β2 and all-trans retinoic acid (atRA), derived from the bone microenvironment, can cooperate with intrinsic tumor signals to promote cellular dormancy. These dormant cells are characterized by growth arrest, enhanced survival, and strong expression of pluripotency-associated genes23, 24. Kobayashi (2011) showed that bone morphogenetic protein 7 (BMP-7), secreted by bone stromal cells, induces dormancy in prostate cancer cells via the p38 pathway and upregulation of the metastasis suppressor gene NDRG125. Buijs . (2007) highlighted the capacity of BMP-7 to selectively inhibit bone metastasis26. More recently, Nobre. (2021) reaffirmed the dormancy-regulating roles of TGF-β and BMP-7 signaling pathways originating from NG2/Nestin mesenchymal stem cells (MSCs)27. Thus, in the bone microenvironment, atRA, TGF-β2, and BMP-7 are supplied in excess, fostering long-term stability and dormancy of cancer cells for years or even decades. Additionally, abscisic acid in bone marrow and serum induces G0 cell cycle arrest in prostate cancer cells via PPARγ signaling28. Prostate cancer cells have also been found to adhere to osteoblasts within the hematopoietic stem cell niche, resulting in TBK1 upregulation, which suppresses mTOR signaling and promotes cellular dormancy and drug resistance29. Furthermore, leukemia inhibitory factor from bone marrow stromal cells inhibits the growth of breast cancer cells and drives dormancy in bone through its ligand and STAT3 signaling30. These findings reveal a complex network of signaling interactions between bone marrow cells and dormant cancer cells.

Uniquely among mechanisms supporting cancer dormancy, breast cancer cells cannibalize MSCs, potentially acquiring a dormant phenotype via TWIST1 and MAPK upregulation31. A hybrid breast cancer cell line, MDA-MSC-hyb5, remained dormant in mouse tissue and, once activated by unknown factors, developed 1.8-fold faster than its parental MDA-MB-231 cells32. Bartosh . (2016) hypothesized that key factors of the senescence-associated secretory phenotype become upregulated after MSC cannibalism, inducing a dormant phenotype by spreading growth arrest signals31. Adipose mesenchymal stem cells may promote cancer dormancy and increase chemoresistance via exosomal microRNAs, which are transferred to breast cancer cells in co-culture33. However, interaction with dormant breast cancer cells can trigger tenascin production by adipose-derived stem cells, subsequently heightening cancer invasiveness34. MSCs also contribute to reduced proliferation, enhanced adhesion, and decreased migration of cancer cells via their extracellular vesicles35. These findings clarify the mechanisms that precipitate tumor relapse and metastasis.

PI3K/AKT is a critical biological pathway in cancer cells, given its direct involvement in cell proliferation, survival, and metabolism36. Inhibiting PI3K/AKT is an effective approach to prevent the aggressive growth of various cancer cells37, 38, 39, 40, 41. Endo (2014) observed that colorectal cancer cells entered dormancy when AKT was blocked under hypoxia and limited growth factor availability42. Experiments on different cancer cell types show that impairing the PI3K/AKT signaling pathway upregulates p21 and p2743, 44, 45, 46, two central inhibitors that can modify the cell cycle and halt proliferation.

Epigenetic modifications have recently gained prominence in uncovering the mechanisms of cancer cell dormancy. Several studies highlight their crucial roles across cancer types. For instance, Sun (2022) reported that in head and neck squamous cell carcinoma (HNSCC), TGF-β and p38 signals stimulate macroH2A1 or macroH2A2 overexpression in an autocrine loop, thereby inducing reversible dormancy and preventing metastasis formation47. Likewise, Ferrer-Diaz (2024) suggested that H3K4 methylation is vital for breast cancer stem cell survival, with methyltransferases KMT2B and KMT2D maintaining a dormant and drug-resistant phenotype48. Additionally, ovo-like (OVOL) transcription factors appear to foster dormancy traits in aggressive breast cancer cells, including growth arrest, morphological changes, and reduced migration. OVOL1 and OVOL2 upregulate E-cadherin while downregulating mesenchymal markers—an expression pattern associated with poor prognosis in ERlow breast cancer patients. OVOLs also enhance chromosome 1 open reading frame 116 (C1orf116)—a potential autophagy receptor—to preserve the epithelial phenotype and modulate tumor behavior. The OVOL-C1orf116 axis alters metabolism by lowering glutathione-related metabolites and raising amino acids, indicating a capacity for redox regulation and amino acid recycling during dormancy49. In ER+ mammary carcinoma (ER+ MC) cells, hypomethylation upregulates Trefoil factor 3 (TFF3), sustaining dormancy-related traits and therapy resistance via a BCL2-dependent mechanism50. In parallel, Llinas-Bertran (2024) revealed that extended dormancy in ER+ MC cells induced by tamoxifen or aromatase inhibitors was not linked to genetic mutations but rather to epigenetic alterations, specifically increased heterochromatin marks (H3K9me2, H3K27me3, and H4K20me3)51. Laguillaumie . (2024) discovered that dormant melanoma cells keep genetic stability, whereas their leukemia counterparts accumulate mutations during dormancy, implying ongoing genetic evolution. This study also identified distinct CNVs, epigenetic marks, and changes in gene and protein expression—particularly in metabolic and dormancy-associated genes such as Vars2, Eno1, Nudt5, and Capg7. These observations not only pinpoint potential therapeutic targets but also demonstrate clinical relevance through patient-derived data.

Just as some animals hibernate to withstand unfavorable conditions, cancer cells seem to adopt dormancy as a survival strategy. Based on the evidence discussed, it is clear that signaling pathways regulating dormancy in various cancers are highly diverse. When conditions improve or undergo certain changes, these dormant cells reactivate and regain malignancy—they are widely considered a primary cause of cancer recurrence after treatment.

The “awakening” of dormant cancer cells

Several studies have investigated how the reactivation of cancer cells from a dormant state greatly contributes to tumor recurrence, leading to serious complications such as malignant transformation, metastasis, or even death52. Some studies have shown the possibility of carcinogenesis and awakening from dormancy in cancer cells under chronic inflammatory conditions among patients with pre-existing diseases53. Attenuated interferon-gamma (IFNγ) has been shown to facilitate the escape of cancer cells from dormancy and promote relapse54, 55, 56, 57, as IFNγ serves as a key mediator of dormancy induction in cancer sustained by natural killer cells, potentially through the IDO-Kyn-AhR signaling pathway58, 59. Although immune cells, such as CD8+ T cells, can recognize mutant antigens expressed on cancer cells and keep them in a dormant state60, chronic inflammation can reactivate dormant cancer cells via the EMT process and seed them onto new metastatic sites61. Lipopolysaccharide (LPS) maintains inflammation via neutrophil recruitment and causes dormant cancer cells in mice to reenter their cell cycle. In the course of inflammation, NETs are formed and stimulate the resurgence of dormant cancer cells. , analysis of the mechanism showed that NE and MMP-9, two NET-associated proteases, sequentially cleave laminin. Similar to the former demonstration, this loss of laminin causes dormant cancer cell proliferation via the initiation of integrin α3β1 signaling pathways62.

Multiple signaling pathways and metabolic alterations have been implicated in the reactivation of dormant cancer cells, contributing to tumor recurrence. In HER2-downregulated dormant breast cancer cells, enhanced fatty acid oxidation generates ATP but also leads to reactive oxygen species (ROS) accumulation, subsequently activating NRF2. Stabilized NRF2 restores redox homeostasis and supports nucleotide biosynthesis through thioredoxin reductase and the pentose phosphate pathway, thereby facilitating tumor resurgence63. In mouse models of lung cancer, PMN-MDSCs promote dormancy escape via S100A8/A9 signaling. , stress hormones such as epinephrine, norepinephrine, cortisol, and serotonin trigger PMN activation under exogenous S100A8/A9 treatment, which in turn initiates myeloperoxidase and several oxidized or hydrolyzed phospholipids. These lipids can upregulate FGFR signaling and drive proliferation of previously dormant cancer cells. Clinically, elevated S100A8/A9 levels in NSCLC patients correlates with an earlier relapse following tumor resection64.

Additional pathways implicated in dormancy escape include NOTCH4-mediated activation of breast cancer stem cells, Hedgehog (Hh)-driven EMT induction, and ATF6-EGF signaling in NSCLC, which promotes angiogenesis and reactivates slow-cycling tumor cells65, 66, 67. Hypoxia-induced LOXL2 expression in dormant MCF-7 cells also facilitates EMT and a phenotypic transition68. Complementing these mechanisms, recent findings demonstrate that exosome-mediated delivery of NR2F1 and its lncRNA regulator NR2F1-AS1 drives the switch from dormancy to proliferation in enzalutamide-resistant prostate cancer. NR2F1-AS1, via interaction with SRSF1, stabilizes NR2F1 expression, leading to hormonal receptor maintenance and sustained proliferative signaling, while HnRNPA2B1 facilitates their packaging into exosomes69. These coordinated metabolic, immunologic, and transcriptional reprogramming events highlight a complex regulatory network underlying cancer dormancy and reactivation.

In general, dormant cancer cells remain quiescent, not growing or expanding. Meanwhile, angiogenesis helps to provide oxygen and nutrients to promote tumor growth. Thus, angiogenesis is considered critical for breaking dormancy in tumors. ARHI is highly expressed on normal ovarian and breast epithelial cells but significantly downregulated or absent in 60-70% of breast and ovarian cancer cases70. ARHI often acts as a tumor suppressor since its re-expression upregulates cell cycle proteins such as p53, p21, and p27, which eventually results in cell cycle arrest in breast cancer cells71. In the highly-expressed-ARHI xenograft model, TIMP3 and CDH1, two anti-angiogenic factors, are upregulated during dormancy since their DNA methylation level decreased. In contrast, increasing methylation leads to the downregulation of these two factors and drives tumor recurrent growth. It is consistent with immunohistochemical staining of PCNA and CD31 results, where they were both reduced in dormant xenografts and escalated in those with progressive growth72. Remodeling of the endosteal niche mediated by osteoclasts can also lead to the reactivation of dormant myeloma cells73, further emphasizing the significance of cell-cell and cell-niche interactions in this process. These insights open new avenues for the development of targeted therapies and the enhancement of treatment efficacy.

Epigenetic regulation plays a critical role in both maintaining cancer cell dormancy and enabling their eventual reactivation. For instance, the histone methyltransferase G9a, which mediates H3K9 methylation, has emerged as a key factor in this process. Under hypoxic conditions, G9a cooperates with hypoxia-inducible factors to repress differentiation genes and activate survival and angiogenic programs, promoting dormancy and tumor growth74, 75. Post treatment, G9a continues to silence pro-inflammatory genes, enabling immune evasion and long-term cellular quiescence76. It also promotes immunosuppression through the Notch pathway and represses adhesion molecules like EpCAM, supporting reactivation and metastasis upon dormancy exit77, 78. Besides, regulators such as EZH1/279, LSD180, HDACs81, and DNMTs82, 83 often interact and form complex networks that control gene expression programs associated with cancer dormancy and reactivation84. Targeting these factors may offer promising therapeutic avenues to prevent cancer relapse in clinical trials.

Awakening cancer cells from dormancy represents a promising area of research for the development of advanced cancer therapies. Based on the above content, we summarized the factors influencing the dormancy and reactivation of cancer cells in Table 1. However, a deeper understanding of this mechanism is essential, and further research is required to translate these findings into clinical practice.

Table 1

Summary of factors that influence the 'sleep' and 'wakefulness' of dormant cancer cells

Factors

Dormancy maintainance

Dormancy exit

Signaling

Upregulation of TGF- 2, BMP-7, p38 MAPK

Downregulation of PI3K/AKT

NOTCH4, ERK, FGFR, integrin α3 β1

Microenvironment

Hypoxia, nutrient depletion, bone marrow niches stability

Chronic inflammation, angiogenesis, uncontrolable microenvironment

Immunity

IFN γ mediator, CD8+ T-cell

Downregulated IFN γ

Upregulated NETs

Reactivation through MDSCs

Metabolism

Cell growth arresst

Low energy usage

ROS, NRF2, PPP promote recurrence

Extracellular signals

Upregulation of TBK, ARHI

Downregulation of mTOR

Upregulation of mTOR or methylation of CDH1/TIMP3

Downregulation of ARHI

Epigenetic regulators

Histone modifications (e.g. gene methylation, macroH2A1/2 overexpression)

Gene regulation (e.g. OVOL upregulates C1orf116, BCL2-mediated TFF3 hypomethylation)

Enhanced epigenetic-related enzyme activites (e.g. G9a, HDACs, DNMTs)

Reprogramming of epigenetic pathways (e.g. EpCAM reduction)

Dormancy in cancer and cancer stem cells

According to previous studies, cancer cells in a dormant state have been proven to present some properties that resemble cancer stem cells (CSCs), such as resistance to drugs, the ability to evade the immune system, and the capacity to metastasize85, 86. Thus, there may be a complex relationship between cancer stemness and dormancy. Some CSCs remain in a dormant state under nutrient-depleted conditions. BEX2 is significantly expressed in CD274 cholangiocarcinoma cells and maintains these CSCs in dormancy by interacting with the TUFM mitochondrial protein, which eventually leads to mitochondrial dysfunction87. BEX2 is also required for the dormancy of hepatocellular carcinoma88. Colorectal stem-like cancer cells cultured in serum-deprived conditions become dormant because fatty acid oxidation mediates Nanog expression, and Nanog binds to the promoter of p21 and p27 to enhance these cell cycle blockers89. As Nanog plays a vital role in regulating stem cell properties, this relationship between CSC maintenance and dormancy needs to be carefully considered.

Hypoxia plays a pivotal role in both inducing cancer dormancy and maintaining CSCs90, 91, 92, 93, 94, 95, 96. It not only drives cellular quiescence but also reshapes metabolic and signaling pathways critical for tumor progression, immune evasion, and therapeutic resistance97, 98. Chromosome 4 open reading frame 47 (C4orf47), a direct target of HIF-1α, upregulates several cell cycle repressors and downregulates cell cycle promoters to induce G0/G1 arrest and cellular dormancy99. C4orf47 also enhances CD44 expression and sustains CSC-like phenotypes in pancreatic and gallbladder cancers under hypoxia93, 99. Additionally, in salivary adenoid cystic carcinoma, HIF-1α has been recently found to increase DEC2 by lowering miR-922, which possibly influences dormancy via lipid metabolism100. Expanding on these insights, recent studies show that hypoxia-induced metabolic shifts, such as L-2-hydroxyglutarate (L-2HG) accumulation via lactate dehydrogenase activity, preserve stemness and immune evasion in pancreatic cancer101. In triple-negative breast cancer, oxidative ATM activation under hypoxia maintains CSC traits and enhances therapy resistance102. Glioma stem-like cells also upregulate hypoxia-inducible gene 2 (HIG-2) under low oxygen, leading to lipid accumulation and Wnt/β-catenin activation via FZD10, which increases insulin-like growth factor binding protein 2 (IGFBP2) secretion and suppresses radioimmunogenicity103. In renal cell carcinoma, a multi-omics model confirmed that hypoxia modulates stemness and immune infiltration104. Similarly, in head and neck squamous cell carcinoma, hypoxia-driven circulating tumor cells exhibit altruistic behavior and therapy resistance independent of genetic mutations105. Collectively, these findings underscore the multifaceted role of hypoxia in orchestrating dormancy and stemness through metabolic reprogramming, epigenetic regulation, and microenvironmental adaptation, marking it as a central target in overcoming cancer recurrence and resistance. Notably, neoadjuvant bevacizumab improved oxygenation in glioblastoma, reduced CSC characteristics, and enhanced treatment response106.

Autophagy activates when cells are in a state of stress to help cancer stem cells survive under these conditions107. When cancer stem cells enter a quiescent state, they do not proliferate, and autophagy is often highly activated. Dormant MCF-7 breast cancer cells express stem cell markers such as CD44 and ALDH1. MCF-7 breast cancer cells undergo autophagy activation to maintain a dormant status, as autophagy inhibition leads to the reversal of the dormant phenotype108. Alyssa . (2019) demonstrated that a high autophagic level reduces Pfkfb3 expression via p62/SQSTM1 and guides breast cancer cells to cellular dormancy109. Because autophagy plays a vital role in controlling the survival of dormant cancer cells, inhibition of autophagic flux results in apoptosis in breast cancer cells, suggesting a therapeutic approach for breast cancer recurrence110. Likewise, autophagy is indispensable for the quiescence of ovarian cancer stem-like cells111. Downregulation of autophagy similarly forces glioblastoma cells to exit dormancy, but this condition also enhances the expression of stemness markers such as Oct-4 and CD133112. The influence of the autophagy process on the expression of stemness markers varies among different cancer types, as autophagy may act as a “double-edged sword” in cancer, with its role in promoting or preventing tumorigenesis still unclear113. However, autophagy strongly affects the promotion of cellular dormancy in general. Harsh conditions within the tumor microenvironment can cause CSCs or dormant cancer cells to accumulate damage, which potentially leads to cell death. Autophagy functions as a protective mechanism that helps manage intracellular stress and stabilize genome integrity by recycling damaged organelles or misfolded proteins114. Autophagy not only keeps CSCs or dormant cancer cells in an inactive state but also enables them to initiate tumor formation when environmental conditions become favorable, allowing them to exit dormancy and re-enter the cell cycle110, 115, 116, 117.

Drug resistance in cancer stem cells and dormant cancer cells

Another aspect that links cancer stem cells with dormant cancer cells is their shared capacity for resistance to drugs and therapies. Drug resistance in cancer stem cells is largely mediated by ATP-binding cassette (ABC) transporter proteins118. These transmembrane transporters are not exclusive to cancer cells, they are also found in various normal cells throughout the body, where they regulate the transport of substances across the cell membrane, contributing to endocrine and exocrine balance. In cancer cells, mutations often occur in the nucleotide-binding domain of ABC transporters, which not only alter drug transport but also impair intracellular energy transfer. As a result, therapeutic agents are unable to exert cytotoxic effects, contributing to drug resistance through ABC transporters119. In addition, metastatic cancer cells infiltrate the lymphatic and vascular systems, disseminating to distant organs before entering a dormant state. During dormancy, they may secrete growth factors and cytokines in preparation for future activation and metastasis formation120. This behavior is considered a key step in the development of metastatic tumors and represents a major cause of cancer-related mortality.

As mentioned above, the ratio between activity of p38 and ERK1/2 is an important indicator in identifying the level of proliferation and dormancy in cancer cells. In particular, p38 acts as a central controller of many pathways related to drug resistance in different types of tumors. SOX9, a downstream signal of p38, has been implicated in drug resistance in breast, esophageal, and cholangiocarcinoma stem cells121, 122, 123. SOX2 and SOX9 are strongly expressed in breast and lung cancer stem cells through DKK1 inhibiting the Wnt signaling pathway, which maintains dormancy in cancer cells124. Although suppression of the Wnt pathway often enhances the therapeutic effect of cancer drugs125, 126, 127, 128, 129, a study in lung cancer cells has also shown that chemotherapy resistance persists even when this pathway is inhibited130. In HEp3 cells, activation of two stress response regulators, protein kinase-like ER kinase (PERK) and BiP, depends on p38 signal. Upregulation of BiP inhibits Bax, an important signal in drug-induced apoptosis, and renders HEp3 cells resistant to several chemotherapeutic drugs131.

In some cases, dormant cancer cells also adjust cellular metabolism to acquire a drug-resistant phenotype. Colorectal dormant cancer cells collaborate with cancer-associated fibroblasts to promote glutamine metabolism and reduce toxicity of 5-fluorouracil (5-FU)132. Glucose starvation could induce massive death of glioblastoma cells under temozolomide (TMZ) or carboplatin treatment, but a small subset of cells escapes and becomes persistent to these chemotherapeutic drugs, entering a quiescent state by enhancing autophagy133. Prostate cancer cells, which are adapted to grow in medium containing the androgen receptor antagonist 2-hydroxy-flutamide, undergo epigenetic modifications. These prostate cancer cells are quiescent and less sensitive to docetaxel, while upregulating pluripotent transcription factors such as Nanog and Oct4. Observations on prostate cancer biopsies confirm that phosphocholine metabolism and methylation are restricted while deacetylation is upregulated. These lead to the reprogramming of prostate cancer cells into cancer stem-like cells134.

These pieces of evidence prove that signaling pathways connecting drug resistance and dormancy in cancer cells are highly complex since dormant cancer cells may use any mechanism to conquer chemotherapeutic agents. However, drug resistance in dormant cancer cells is not always permanent and can be influenced by specific regulatory factors. Kinoshita . (2012) demonstrated that reduced expression of nuclear pore protein NUP62 induces cisplatin resistance in dormant ovarian cancer cells, which can be reversed by restoring NUP62 expression135. Ebinger (2016) further demonstrated that dormant leukemia cells cultured exhibited reduced drug resistance compared to their previously dormant counterparts . This finding highlights the potential efficacy of therapeutic strategies that specifically target dormant leukemia cells after their mobilization from the bone marrow niches, which are believed to play a critical role in maintaining cancer cell dormancy136. Therefore, more studies are needed to unveil this association. Dormant cancer cells and cancer stem cells share features like quiescence and drug resistance, yet differ in origin, regulatory pathways, and roles in tumor progression (Table 2).

Table 2

Comparisons between cancer stem cells and dormant cancer cells

Aspects

Cancer stem cells

Dormant cancer cells

Proliferation

Quiescent or slow-cycling, can re-enter cell cycle

Quiescent (G0/G1 phase), can be reactivated

Drug desistance

ABC transporters, autophagy, metabolic reprogramming

p38 signaling, autophagy, niche protection, metabolic shift

Immune evasion

Low immunogenicity, immune checkpoint regulation

Secrete factors to evade immunity; reside in protected niches

Hypoxia response

Maintains stemness; upregulates CD44, Nanog, SOX2, via HIF-1α

Hypoxia triggers dormancy via HIF-1α, C4orf47, p21/p27 induction

Autophagy role

Maintains quiescence and stress tolerance

Maintains dormancy and survival under stress

Angiogenesis

Produces angiogenic factors to support tumor growth

Relies on angiogenesis for reactivation and recurrence

Metastasis initiation

Capable of initiating and maintaining metastases

Disseminate early and remain dormant before forming metastases

Stemness markers

CD44, ALDH1, Nanog, SOX2/9, Oct4

May express CSC markers during dormancy (e.g., CD44+, ALDH1+)

Metabolic features

Uses fatty acid oxidation, glucose regulation under stress

Adapts metabolism for survival (e.g., glutamine use, epigenetic shift)

Therapeutic implications

Targeting stemness pathways or ABC transporters

Targeting dormancy regulators, autophagy, niche exit strategies

Targeting dormancy in cancer therapies

Currently, treatment options targeting dormancy in cancer focus on three main strategies: (1) maintaining the dormant state of cancer cells, (2) reactivating dormant cells followed by chemotherapy to eliminate them, or (3) completely eradicating these cells while they remain dormant137. Although option (1), which involves peacefully coexisting with dormant cancer cells, carries a high risk due to the potential for uncontrolled reactivation and unforeseen clinical outcomes, options (2) and (3) provide more proactive alternatives. They aim to directly target and eradicate dormant cell populations, thereby significantly minimizing the risk of cancer recurrence in patients.

For type (1), dormant cells must be strictly kept in a “peaceful hibernation” state. Once awakened, they may become more malignant, aggressively increase in population, and metastasize137. Therefore, maintaining dormancy-inducing signals while inhibiting proliferative cues remains a top priority. Tamoxifen, an estrogen receptor antagonist, has been proven to prevent the growth of dormant breast cancer cells and significantly extend patient survival times138. The glutaminase inhibitor CB-839 hinders the reawakening of dormant tumor cells, as recurrent breast cancer cells are sensitive to glutaminase inhibition under the metabolic control of NRF263. In mice, NET-mediated laminin remodeling activates dormant lung cancer cells, but chimeric mouse immunoglobulin G2a antibodies can counteract this process62. Regucalcin sustains dormancy in prostate cancer cells by stimulating p38, downregulating ERK, and reducing angiogenesis through increased miR-23c expression139. Gene therapy strategies targeting uPAR and Src successfully induce a dormancy phenotype in neuroblastoma140 and breast cancer cells141. Targeting the β2-adrenergic receptor in the PMN with ICI-118,551 abrogates norepinephrine-induced S100A8/A9 secretion, preventing dormant tumor cell reactivation64. Palbociclib, a CDK4/6 inhibitor that prevents cells from transitioning from the G0/G1 phase to the S phase, significantly increases survival rates in breast cancer patients. The FDA approved its clinical use for various cancer types in 2016142. Although this strategy is theoretically feasible, it has also been validated , , and clinical trials. Recently, certain miRNAs have been identified as key targets for sustaining “peaceful hibernation,” as their inhibition may foster dormancy induction33. Furthermore, epigenetic modification strategies have shown promise. For instance, Clements (2021) demonstrated that histone deacetylase inhibitors reduce proliferation rates in various cancer cell lines, elevate the expression of dormancy-related genes, and delay tumor recurrence143. However, this approach still entails potential risks, as not all cancer cells become truly dormant; some merely exhibit reduced proliferation, which may ultimately trigger tumorigenesis137.

For strategies (2) and (3), several agents are used to disrupt dormancy, making cells more vulnerable to chemotherapy and facilitating their removal. Both and data indicate that granulocyte-colony stimulating factor (G-CSF) can reactivate dormant leukemia stem cells, allowing their re-entry into the normal cell cycle. Subsequent therapy with cytarabine or imatinib then effectively eliminates these reactivated cells144. Nimustine and bortezomib have each been shown to decrease dormant breast cancer and myeloma cell populations, respectively, an effect not seen with therapies targeting only proliferating cells73, 145. Combining a Src inhibitor (AZD0530) with a MEK1/2 inhibitor (AZD6244) prevents dormant breast cancer cells from responding to external stimuli, ultimately inducing apoptosis146. Xie . (2022) reported that erlotinib and CB-839 effectively kill dormant and drug-resistant colorectal cancer cells132. Moreover, certain genetic modifications in glioblastoma cells reactivate dormant cells and heighten temozolomide-induced apoptosis112. Dwyer (2024) demonstrated that inhibiting autophagy in breast cancer cells, either through pharmacological agents or genetic manipulation, not only postpones tumor recurrence but also diminishes dormant cell survival, as shown in both and studies117. Hu . (2023) demonstrated that STING pathway activation suppresses the reactivation of dormant metastatic cells in lung adenocarcinoma. Additionally, STING agonist treatment eliminates these cells in a T and NK cell-dependent manner, offering a promising strategy to prevent metastatic relapse147. Growing research is evaluating more compounds for their efficacy in targeting dormant cancer cells, utilizing both studies and clinical trials (Table 3).

Table 3

Strategies developed over the past decade (2015-2025) to treat dormant cancer cells

Methods or Compounds

Cancer type

Outcomes

Referrence

Dormancy maintainance

Indirect or direct eradication of dormant cells

Granulocyte-colony stimulating factor (GCS-F) plus cytarabine or imatinib

Leukemia

v

144

Valproic acid

Breast cancer

v

30

Palbociclib

Various cancer

v

142

IFNg plus IDO/AhR inhibitor (Interferon gamma + Indoleamine 2,3-dioxygenase / Aryl hydrocarbon receptor inhibitors)

Melanoma

v

59

Nimustine, bortezomib

Myeloma

v

145

Immunoglobulin G2a antibodies

Lung cancer

v

62

Itraconazole

Colorectal cancer

v

148

Interferon beta (IFN-beta)

Glioma

Breast cancer

v

149, 150

FBXW7 ablation

Breast cancer

v

21

ICI-118,551 hydrochloride (ICI-118,551)

Lung cancer

v

64

Exosomal miRNAs

Various cancer

v

33

Glutaminase inhibitor (CB-839)

Breast cancer

v

v

63, 132

Yes-associated protein (YAP) activation

Lung cancer

v

151

Genetics modification

Glioblastoma

v

112

Regucalcin

Prostate cancer

v

139

Targeting enhancer of zeste homolog 1/2 (EZH1/2)

Leukemia

v

152, 153

Histone deacetylase inhibitors (HDAC inhibitors)

Breast cancer

v

143

5-azacytidine plus all-trans retinoic acid (atRA) reprogramming

HNSCC

v

154

uPAR and Scr downregulation via gene therapy

Neuroblastoma

Breast cancer

v

140, 141

MacroH2A2 restoration

HNSCC

v

47

Erlotinib

Breast cancer

v

132

Gedatosilib

Breast cancer

v

155

STING activation

Lung cancer

v

147

T-cell immunotherapies

Breast cancer

v

156

Trefoil factor 3 (TFF3) depletion plus CDK4/6 inhibitor

Breast cancer

v

50

To date, current treatments have not been able to successfully eliminate all dormant cancer cells. Even a minimal population of cells that survive these interventions may later acquire malignant potential, leading to disease relapse and a worsened prognosis for patients. Optimal cancer treatment requires either sustaining dormancy or eliminating these dormant cells, but both strategies remain challenging. Continual therapy to maintain dormancy is often impractical, while reactivating cells to kill them can significantly worsen outcomes if therapies fail157. To address these limitations, a comprehensive understanding of the biological mechanisms governing dormancy and reactivation is essential for developing safer and more effective treatment strategies.

Future directions in cancer dormancy

Understanding and targeting cancer cell dormancy represents a critical frontier in oncology research, particularly in addressing tumor relapse and metastasis that can occur months or even years after initial treatment. Current efforts must focus on elucidating the molecular mechanisms that govern dormancy, including intracellular signaling pathways158, 159, epigenetic regulation160, metabolic reprogramming161, and interactions with the tumor microenvironment159. The development of physiologically relevant and models162, 163, along with the identification of robust dormancy-specific biomarkers, is essential for tracking dormant cells and distinguishing them from therapy-induced quiescent populations164. Future directions include developing targeted therapies to either eradicate dormant cells or maintain them in a non-proliferative state indefinitely137, 165, integrating immunological approaches to modulate dormancy surveillance86, and leveraging liquid biopsy and multi-omics technologies for early detection of reactivation events13. In the near future, the establishment of large-scale clinical databases and interdisciplinary collaborations will be pivotal in translating dormancy research into precision medicine strategies166.

Conclusion

The significance of cancer cell dormancy studies has grown markedly because they provide critical insights into many cases of late relapse, metastasis, and therapy resistance. Although dormant cancer cells remain in a non-dividing state, they still possess reproductive capabilities, making them difficult to eliminate with standard therapies, as many are concealed in protective microenvironments167. To build upon new findings, future studies should integrate mechanistic insights (. signaling, epigenetics, metabolic control) alongside advanced techniques such as liquid biopsy and multi-omics to continuously detect and monitor dormancy-related changes in real time. Furthermore, developing reliable dormancy-specific biomarkers and enhanced preclinical models will improve our ability to clearly distinguish dormant cells from therapy-induced quiescent cells. Ultimately, revolutionizing cancer management post-treatment and preventing disease recurrence could be achieved by devising innovative strategies to eradicate or durably contain dormant cells, marking a breakthrough in precision oncology.

Abbreviations

AhR: aryl hydrocarbon receptor, ALDH1: aldehyde dehydrogenase 1, ARHI: aplasia rag homolog member 1, atRA: all-trans retinoic acid, BCL2: B-cell lymphoma 2, BMP-7: bone morphogenetic protein 7, C4orf47: chromosome 4 open reading frame 47, C1orf116: chromosome 1 open reading frame 116, CNV: copy number variation, DNMT: DNA methyltransferase, EMT: epithelial-to-mesenchymal transition, ERK: extracellular signal-regulated kinase, ER+: estrogen receptor positive, EZH1/2: enhancer of zeste homolog 1/2, FBXW7: F-box and WD repeat domain containing 7, FGFR: fibroblast growth factor receptor, HDAC: histone deacetylase, HNSCC: head and neck squamous cell carcinoma, IDO: indolamine 2,3-dioxygenaseIFN-β: interferon beta, Kyn: kynurenine, LOXL2: lysyl oxidase like 2, LPS: lipopolysaccharide, LSD1: lysine-specific demethylase 1, miRNAs: micro ribonucleotide acids, MMP-9: matrix metalloproteinase-9, NETs: neutrophil extracellular traps, NE: neutrophil elastase, NK cell: natural killer cell, NRF2: nuclear factor-like 2, NSCLC: non-small-cell lung cancer, Pfkfb3: 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3, PMN: polymorphonuclear neutrophils, PMN-MDSC: polymorphonuclear myeloid-derived suppressor cell, PPARγ: peroxisome proliferator-activated receptor gamma, STAT3: signal transducer and activator of transcription 3, TBK1: TANK binding kinase 1, TFF3: Trefoil factor 3

Acknowledgments

None.

Author’s contributions

Bui Dinh Khan took primary responsibility for the layout and content of this manuscript. Nguyen Thi Yen Nhi, Pham Duy Khuong, and Tran Ngo The Nhan contributed equally to this work. We would also like to thank Phung Van Khai (School of Biomedical Sciences, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Clayton, Victoria, Australia) for providing feedback on the content, correcting spelling and grammatical errors, and improving wording of this manuscript. All figures were illustrated using free tools and templates from Canva. All authors read and approved the final version of the manuscript.

Funding

This paper is funded by University of Science, Vietnam National University Ho Chi Minh City (VNUHCM) under grant number T2024-82.

Availability of data and materials

Not applicable.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Declaration of generative AI and AI-assisted technologies in the writing process

The authors declare that they have not used generative AI (a type of artificial intelligence technology that can produce various types of content including text, imagery, audio and synthetic data. Examples include ChatGPT, NovelAI, Jasper AI, Rytr AI, DALL-E, ) and AI-assisted technologies in the writing process before submission.

Competing interests

The authors declare that they have no competing interests.

References

  1. F. Bray, M. Laversanne, H. Sung, J. Ferlay, R.L. Siegel, I. Soerjomataram. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a Cancer Journal for Clinicians 2024; 74(3): 229-63.
  2. A.C. Celsus, W. Spencer. De medicina. London: W. 1935, Heinemann ltd. 1935; :
  3. R.A. Willis. The spread of tumours in the human body. J. & A. Churchill 1934; :
  4. G. Hadfield. The dormant cancer cell. BMJ (Clinical Research Ed.) 1954; 2(4888): 607-10.
  5. M.A. Gimbrone, S.B. Leapman, R.S. Cotran, J. Folkman. Tumor dormancy in vivo by prevention of neovascularization. The Journal of Experimental Medicine 1972; 136(2): 261-76.
  6. X. Wang, J. Yu, J. Yan, K. Peng, H. Zhou. Single-cell sequencing reveals MYC targeting gene MAD2L1 is associated with prostate cancer bone metastasis tumor dormancy. BMC Urology 2022; 22(1): 37.
  7. M.O. Laguillaumie, S. Titah, A. Guillemette, B. Neve, F. Leprêtre, P. Ségard. Deciphering genetic and nongenetic factors underlying tumour dormancy: insights from multiomics analysis of two syngeneic MRD models of melanoma and leukemia. Biological Research 2024; 57(1): 59.
  8. H. David. Rudolf Virchow and modern aspects of tumor pathology. Pathology, Research and Practice 1988; 183(3): 356-64.
  9. J. Folkman. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nature Medicine 1995; 1(1): 27-30.
  10. J.A. Aguirre-Ghiso. Models, mechanisms and clinical evidence for cancer dormancy. Nature Reviews. Cancer 2007; 7(11): 834-46.
  11. A.C. Yeh, S. Ramaswamy. Mechanisms of cancer cell dormancy\textemdashanother hallmark of cancer?. Cancer Research 2015; 75(23): 5014-22.
  12. H. Endo, M. Inoue. Dormancy in cancer. Cancer Science 2019; 110(2): 474-80.
  13. M.S. Ganesh, R. Revanth, C.M. Bharathi. Advanced Biomarkers and Precision Medicine: Innovative Strategies to Prevent Cancer Recurrence. Journal of Cancer Research Updates 2025; 14: 1-11.
  14. M. Retsky, R. Demicheli, W.J. Hrushesky. Does surgery induce angiogenesis in breast cancer? Indirect evidence from relapse pattern and mammography paradox. International Journal of Surgery 2005; 3(3): 179-87.
  15. J. Dittmer. The Mechanism Enabling Hibernation in Mammals. In: Iwaya-Inoue, M., Sakurai, M., Uemura, M. (eds) Survival Strategies in Extreme Cold and Desiccation. Advances in Experimental Medicine and Biology 2017; 1081:
  16. Y. Horii, T. Shiina, Y. Shimizu. ERKMAPK activity as a determinant of tumor growth and dormancy; regulation by p38SAPK. Cancer research 2018; 63(7): 1684-95.
  17. J.A. Aguirre-Ghiso. ERKMAPK activity as a determinant of tumor growth and dormancy; regulation by p38SAPK. Cancer Research 2003; 63(7): 1684-95.
  18. Y. Fan, X. Ren, Y. Wang, E. Xu, S. Wang, R. Ge. Metformin inhibits the proliferation of canine mammary gland tumor cells through the AMPK/AKT/mTOR signaling pathway in vitro. Oncology Letters 2021; 22(6): 1-7.
  19. I. Herr, K.M. Debatin. Cellular stress response and apoptosis in cancer therapy. Blood 2001; 98(9): 2603-14.
  20. M.H. Manjili. Tumor dormancy and relapse: from a natural byproduct of evolution to a disease state. Cancer Research 2017; 77(10): 2564-9.
  21. H. Shimizu, S. Takeishi, H. Nakatsumi, K.I. Nakayama. Prevention of cancer dormancy by Fbxw7 ablation eradicates disseminated tumor cells. JCI Insight 2019; 4(4): e125138.
  22. C. Prunier, D. Baker, P. ten Dijke, L. Ritsma. TGF-β family signaling pathways in cellular dormancy. Trends in Cancer 2019; 5(1): 66-78.
  23. M.S. Sosa, F. Parikh, A.G. Maia, Y. Estrada, A. Bosch, P. Bragado. NR2F1 controls tumour cell dormancy via SOX9-and RARβ-driven quiescence programmes. Nature Communications 2015; 6(1): 6170.
  24. P. Bragado, Y. Estrada, F. Parikh, S. Krause, C. Capobianco, H.G. Farina. TGF-β2 dictates disseminated tumour cell fate in target organs through TGF-β-RIII and p38α/β signalling. Nature Cell Biology 2013; 15(11): 1351-61.
  25. A. Kobayashi, H. Okuda, F. Xing, P.R. Pandey, M. Watabe, S. Hirota. Bone morphogenetic protein 7 in dormancy and metastasis of prostate cancer stem-like cells in bone. The Journal of Experimental Medicine 2011; 208(13): 2641-55.
  26. J.T. Buijs, C.A. Rentsch, G. van der Horst, P.G. van Overveld, A. Wetterwald, R. Schwaninger. BMP7, a putative regulator of epithelial homeostasis in the human prostate, is a potent inhibitor of prostate cancer bone metastasis in vivo. The American Journal of Pathology 2007; 171(3): 1047-57.
  27. A.R. Nobre, E. Risson, D.K. Singh, J.S. Di Martino, J.F. Cheung, J. Wang. Bone marrow NG2+/Nestin+ mesenchymal stem cells drive DTC dormancy via TGF-β2. Nature Cancer 2021; 2(3): 327-39.
  28. Y. Jung, F.C. Cackowski, K. Yumoto, A.M. Decker, Y. Wang, M. Hotchkin. Abscisic acid regulates dormancy of prostate cancer disseminated tumor cells in the bone marrow. Neoplasia : An International Journal for Oncology Research 2021; 23(1): 102-11.
  29. J.K. Kim, Y. Jung, J. Wang, J. Joseph, A. Mishra, E.E. Hill. TBK1 regulates prostate cancer dormancy through mTOR inhibition. Neoplasia : An International Journal for Oncology Research 2013; 15(9): 1064-74.
  30. R.W. Johnson, E.C. Finger, M.M. Olcina, M. Vilalta, T. Aguilera, Y. Miao. Induction of LIFR confers a dormancy phenotype in breast cancer cells disseminated to the bone marrow. Nature Cell Biology 2016; 18(10): 1078-89.
  31. T.J. Bartosh, M. Ullah, S. Zeitouni, J. Beaver, D.J. Prockop. Cancer cells enter dormancy after cannibalizing mesenchymal stem/stromal cells (MSCs). Proceedings of the National Academy of Sciences of the United States of America 2016; 113(42): 6447-56.
  32. C. Melzer, J. von der Ohe, T. Luo, R. Hass. Spontaneous fusion of MSC with breast cancer cells can generate tumor dormancy. International Journal of Molecular Sciences 2021; 22(11): 5930.
  33. N. Mohd Ali, S.K. Yeap, W.Y. Ho, L. Boo, H. Ky, D.A. Satharasinghe. Adipose MSCs suppress MCF7 and MDA-MB-231 breast cancer metastasis and EMT pathways leading to dormancy via exosomal-miRNAs following co-culture interaction. Pharmaceuticals (Basel, Switzerland) 2020; 14(1): 8.
  34. M. Nyström, A.T. Lauvrud, S. Pérez-Díaz, P.J. Kingham, R. Wiberg. Interaction of adipose-derived stem cells with active and dormant breast cancer cells. Journal of Plastic, Reconstructive {&}amp; Aesthetic Surgery : JPRAS 2023; 83: 69-76.
  35. J. Casson, O.G. Davies, C.A. Smith, M.J. Dalby, C.C. Berry. Mesenchymal stem cell-derived extracellular vesicles may promote breast cancer cell dormancy. Journal of Tissue Engineering 2018; 9:
  36. N. Jiang, Q. Dai, X. Su, J. Fu, X. Feng, J. Peng. Role of PI3K/AKT pathway in cancer: the framework of malignant behavior. Molecular Biology Reports 2020; 47(6): 4587-629.
  37. B. Ye, L.L. Jiang, H.T. Xu, D.W. Zhou, Z.S. Li. Exression and PI3K/AKT pathway in gastric cancer and its blockade suppresses tumor growth and metastasis. International Journal of Immunopathology and Pharmacology 2012; 25(3): 627-36.
  38. S. Zhu, W. Jiao, Y. Xu, L. Hou, H. Li, J. Shao. Palmitic acid inhibits prostate cancer cell proliferation and metastasis by suppressing the PI3K/Akt pathway. Life Sciences 2021; 286:
  39. C. Li, X. Yang, C. Chen, S. Cai, J. Hu. Isorhamnetin suppresses colon cancer cell growth through the PI3K/TOR pathway. Molecular Medicine Reports 2014; 9(3): 935-40.
  40. N. Gulati. The antiproliferative effect of Quercetin in cancer cells is mediated via inhibition of the PI3K-Akt/PKB pathway. Anticancer Research 2006; 26: 1177-81.
  41. S. Kang, S.M. Dong, B.R. Kim, M.S. Park, B. Trink, H.J. Byun. Thioridazine induces apoptosis by targeting the PI3K/Akt/mTOR pathway in cervical and endometrial cancer cells. Apoptosis : An International Journal on Programmed Cell Death 2012; 17(9): 989-97.
  42. H. Endo, H. Okuyama, M. Ohue, M. Inoue. Dormancy of cancer cells with suppression of AKT activity contributes to survival in chronic hypoxia. PLoS One 2014; 9(6): e98858.
  43. Q. Guo, Y. Xiong, Y. Song, K. Hua, S. Gao. ARHGAP17 suppresses tumor progression and up-regulates P21 and P27 expression via inhibiting PI3K/AKT signaling pathway in cervical cancer. Gene 2019; 692: 9-16.
  44. R. Kannaiyan, K.A. Manu, L. Chen, F. Li, P. Rajendran, A. Subramaniam. Celastrol inhibits tumor cell proliferation and promotes apoptosis through the activation of c-Jun N-terminal kinase and suppression of PI3 K/Akt signaling pathways. Apoptosis : An International Journal on Programmed Cell Death 2011; 16(10): 1028-41.
  45. X. Yang, Y. Feng, Y. Liu, X. Ye, X. Ji, L. Sun. Fuzheng Jiedu Xiaoji formulation inhibits hepatocellular carcinoma progression in patients by targeting the AKT/CyclinD1/p21/p27 pathway. Phytomedicine : International Journal of Phytotherapy and Phytopharmacology 2021; 87: 153575.
  46. J.H. Yoon, J.W. Shin, T.H. Pham, Y.J. Choi, H.W. Ryu, S.R. Oh. Methyl lucidone induces apoptosis and G2/M phase arrest via the PI3K/Akt/NF-κB pathway in ovarian cancer cells. Pharmaceutical Biology 2020; 58(1): 51-9.
  47. D. Sun, D.K. Singh, S. Carcamo, D. Filipescu, B. Khalil, X. Huang. MacroH2A impedes metastatic growth by enforcing a discrete dormancy program in disseminated cancer cells. Science Advances 2022; 8(48): eabo0876.
  48. A.I. Ferrer-Diaz, G. Sinha, A. Petryna, R. Gonzalez-Bermejo, Y. Kenfack, O. Adetayo. Revealing role of epigenetic modifiers and DNA oxidation in cell-autonomous regulation of Cancer stem cells. Cell Communication and Signaling : CCS 2024; 22(1): 119.
  49. D. Drago-Garcia, S. Giri, R. Chatterjee, A. Simoni-Nieves, M. Abedrabbo, A. Genna. Re-epithelialization of cancer cells increases autophagy and DNA damage: implications for breast cancer dormancy and relapse. Science Signaling 2025; 18(883): eado3473.
  50. S. Chen, X. Zhang, B. Basappa, T. Zhu, V. Pandey, P.E. Lobie. TFF3 facilitates dormancy of anti-estrogen treated ER+ mammary carcinoma. Communications Medicine 2025; 5(1): 45.
  51. D. Rosano, E. Sofyali, H. Dhiman, C. Ghirardi, D. Ivanoiu, T. Heide. Long-term multimodal recording reveals epigenetic adaptation routes in dormant breast cancer cells. Cancer Discovery 2024; 14(5): 866-89.
  52. S. Hadad, D.G. Hardie, V. Appleyard, A.M. Thompson. Effects of metformin on breast cancer cell proliferation, the AMPK pathway and the cell cycle. Clinical {&}amp; Translational Oncology : Official Publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico 2014; 16(8): 746-52.
  53. A.P. Adam, A. George, D. Schewe, P. Bragado, B.V. Iglesias, A.C. Ranganathan. Computational identification of a p38SAPK-regulated transcription factor network required for tumor cell quiescence. Cancer Research 2009; 69(14): 5664-72.
  54. K. Hallermalm, K. Seki, A. De Geer, B. Motyka, R.C. Bleackley, M.J. Jager. Modulation of the tumor cell phenotype by IFN-γ results in resistance of uveal melanoma cells to granule-mediated lysis by cytotoxic lymphocytes. The Journal of Immunology : Official Journal of the American Association of Immunologists 2008; 180(6): 3766-74.
  55. P. Namjoshi, L. Showalter, B.J. Czerniecki, G.K. Koski. T-helper 1-type cytokines induce apoptosis and loss of HER-family oncodriver expression in murine and human breast cancer cells. Oncotarget 2019; 10(57): 6006-20.
  56. K.K. Payne, R.C. Keim, L. Graham, M.O. Idowu, W. Wan, X.Y. Wang. Tumor-reactive immune cells protect against metastatic tumor and induce immunoediting of indolent but not quiescent tumor cells. Journal of Leukocyte Biology 2016; 100(3): 625-35.
  57. M. Kmieciak, K.K. Payne, X.Y. Wang, M.H. Manjili. IFN-γ Rα is a key determinant of CD8+ T cell-mediated tumor elimination or tumor escape and relapse in FVB mouse. PLoS One 2013; 8(12): e82544.
  58. A.L. Correia, J.C. Guimaraes, P. Auf der Maur, D. De Silva, M.P. Trefny, R. Okamoto. Hepatic stellate cells suppress NK cell-sustained breast cancer dormancy. Nature 2021; 594(7864): 566-71.
  59. Y. Liu, X. Liang, X. Yin, J. Lv, K. Tang, J. Ma. Blockade of IDO-kynurenine-AhR metabolic circuitry abrogates IFN-γ-induced immunologic dormancy of tumor-repopulating cells. Nature Communications 2017; 8(1): 15207.
  60. J. Eyles, A.L. Puaux, X. Wang, B. Toh, C. Prakash, M. Hong. Tumor cells disseminate early, but immunosurveillance limits metastatic outgrowth, in a mouse model of melanoma. The Journal of Clinical Investigation 2010; 120(6): 2030-9.
  61. A.D. Rhim, E.T. Mirek, N.M. Aiello, A. Maitra, J.M. Bailey, F. McAllister. EMT and dissemination precede pancreatic tumor formation. Cell 2012; 148(1-2): 349-61.
  62. J. Albrengues, M.A. Shields, D. Ng, C.G. Park, A. Ambrico, M.E. Poindexter. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 2018; 361(6409): eaao4227.
  63. D.B. Fox, N.M. Garcia, B.J. McKinney, R. Lupo, L.C. Noteware, R. Newcomb. NRF2 activation promotes the recurrence of dormant tumour cells through regulation of redox and nucleotide metabolism. Nature Metabolism 2020; 2(4): 318-34.
  64. M. Perego, V.A. Tyurin, Y.Y. Tyurina, J. Yellets, T. Nacarelli, C. Lin. Reactivation of dormant tumor cells by modified lipids derived from stress-activated neutrophils. Science Translational Medicine 2020; 12(572): eabb5817.
  65. M.S. D'arcy. Cell death: a review of the major forms of apoptosis, necrosis and autophagy. Cell Biology International 2019; 43(6): 582-92.
  66. S.A. Patel, S.H. Ramkissoon, M. Bryan, L.F. Pliner, G. Dontu, P.S. Patel. Delineation of breast cancer cell hierarchy identifies the subset responsible for dormancy. Scientific Reports 2012; 2(1): 906.
  67. J. Cho, H.Y. Min, H. Pei, X. Wei, J.Y. Sim, S.H. Park. The ATF6-EGF pathway mediates the awakening of slow-cycling chemoresistant cells and tumor recurrence by stimulating tumor angiogenesis. Cancers (Basel) 2020; 12(7): 1772.
  68. K. Weidenfeld, S. Schif-Zuck, H. Abu-Tayeh, K. Kang, O. Kessler, M. Weissmann. Dormant tumor cells expressing LOXL2 acquire a stem-like phenotype mediating their transition to proliferative growth. Oncotarget 2016; 7(44): 71362-77.
  69. W. Chen, Y. Mao, Y.Y. Zhan, W. Li, J. Wu, X. Mao. Exosome-delivered NR2F1-AS1 and NR2F1 drive phenotypic transition from dormancy to proliferation in treatment-resistant prostate cancer via stabilizing hormonal receptors. Journal of Nanobiotechnology 2024; 22(1): 761.
  70. Y. Yu, R. Luo, Z. Lu, W. Wei Feng, D. Badgwell, J.P. Issa. Biochemistry and biology of ARHI (DIRAS3), an imprinted tumor suppressor gene whose expression is lost in ovarian and breast cancers. Methods in Enzymology 2006; 407: 455-68.
  71. X. Zuo, Y. Qin, X. Zhang, Q. Ning, S. Shao, M. Luo. Breast cancer cells are arrested at different phases of the cell cycle following the re-expression of ARHI. Oncology Reports 2014; 31(5): 2358-64.
  72. T. Lyu, N. Jia, J. Wang, X. Yan, Y. Yu, Z. Lu. Expression and epigenetic regulation of angiogenesis-related factors during dormancy and recurrent growth of ovarian carcinoma. Epigenetics 2013; 8(12): 1330-46.
  73. M.A. Lawson, M.M. McDonald, N. Kovacic, W. Hua Khoo, R.L. Terry, J. Down. Osteoclasts control reactivation of dormant myeloma cells by remodelling the endosteal niche. Nature Communications 2015; 6(1): 8983.
  74. J. Ueda, J.C. Ho, K.L. Lee, S. Kitajima, H. Yang, W. Sun. The hypoxia-inducible epigenetic regulators Jmjd1a and G9a provide a mechanistic link between angiogenesis and tumor growth. Molecular and Cellular Biology 2014; 34(19): 3702-20.
  75. F. Casciello, F. Al-Ejeh, G. Kelly, D.J. Brennan, S.F. Ngiow, A. Young. G9a drives hypoxia-mediated gene repression for breast cancer cell survival and tumorigenesis. Proceedings of the National Academy of Sciences of the United States of America 2017; 114(27): 7077-82.
  76. N.W. Mabe, N.M. Garcia, S.E. Wolery, R. Newcomb, R.C. Meingasner, B.A. Vilona. G9a promotes breast cancer recurrence through repression of a pro-inflammatory program. Cell Reports 2020; 33(5): 108341.
  77. Y. Cao, B. Liu, L. Cai, Y. Li, Y. Huang, Y. Zhou. G9a promotes immune suppression by targeting the Fbxw7/Notch pathway in glioma stem cells. CNS Neuroscience & Therapeutics 2023; 29(9): 2508-21.
  78. M.W. Chen, K.T. Hua, H.J. Kao, C.C. Chi, L.H. Wei, G. Johansson. H3K9 histone methyltransferase G9a promotes lung cancer invasion and metastasis by silencing the cell adhesion molecule Ep-CAM. Cancer Research 2010; 70(20): 7830-40.
  79. S.H. Lee, Y. Li, H. Kim, S. Eum, K. Park, C.H. Lee. The role of EZH1 and EZH2 in development and cancer. BMB Reports 2022; 55(12): 595-601.
  80. D. Kim, K.I. Kim, S.H. Baek. Roles of lysine-specific demethylase 1 (LSD1) in homeostasis and diseases. Journal of Biomedical Science 2021; 28(1): 41.
  81. Y. Li, E. Seto. HDACs and HDAC inhibitors in cancer development and therapy. Cold Spring Harbor Perspectives in Medicine 2016; 6(10):
  82. D.J. Kim. The Role of the DNA Methyltransferase Family and the Therapeutic Potential of DNMT Inhibitors in Tumor Treatment. Current Oncology (Toronto, Ont.) 2025; 32(2): 88.
  83. E. Hirata, K. Ishibashi, S. Kohsaka, K. Shinjo, S. Kojima, Y. Kondo. The Brain Microenvironment Induces DNMT1 Suppression and Indolence of Metastatic Cancer Cells. iScience 2020; 23(9): 101480.
  84. M. Tufail, C.H. Jiang, N. Li. Tumor dormancy and relapse: understanding the molecular mechanisms of cancer recurrence. Military Medical Research 2025; 12(1): 7.
  85. F. Francescangeli, M.L. De Angelis, R. Rossi, A. Cuccu, A. Giuliani, R. De Maria. Dormancy, stemness, and therapy resistance: interconnected players in cancer evolution. Cancer Metastasis Reviews 2023; 42(1): 197-215.
  86. X. Jiang, L. Liang, G. Chen, C. Liu. Modulation of immune components on stem cell and dormancy in cancer. Cells 2021; 10(11): 2826.
  87. K. Tamai, M. Nakamura-Shima, R. Shibuya-Takahashi, S.I. Kanno, A. Yasui, M. Mochizuki. BEX2 suppresses mitochondrial activity and is required for dormant cancer stem cell maintenance in intrahepatic cholangiocarcinoma. Scientific Reports 2020; 10(1): 21592.
  88. D. Fukushi, R. Shibuya-Takahashi, M. Mochizuki, H. Fujimori, T. Kogure, T. Sugai. BEX2 is required for maintaining dormant cancer stem cell in hepatocellular carcinoma. Cancer Science 2021; 112(11): 4580-92.
  89. M. Zhang, R. Peng, H. Wang, Z. Yang, H. Zhang, Y. Zhang. Nanog mediated by FAO/ACLY signaling induces cellular dormancy in colorectal cancer cells. Cell Death & Disease 2022; 13(2): 159.
  90. S. Ju, F. Wang, Y. Wang, S. Ju. CSN8 is a key regulator in hypoxia-induced epithelial\textendashmesenchymal transition and dormancy of colorectal cancer cells. Molecular Cancer 2020; 19(1): 1-7.
  91. K. Hoppe-Seyler, F. Bossler, C. Lohrey, J. Bulkescher, F. Rösl, L. Jansen. Induction of dormancy in hypoxic human papillomavirus-positive cancer cells. Proceedings of the National Academy of Sciences of the United States of America 2017; 114(6): 990-8.
  92. A. Carcereri de Prati, E. Butturini, A. Rigo, E. Oppici, M. Rossin, D. Boriero. Metastatic breast cancer cells enter into dormant state and express cancer stem cells phenotype under chronic hypoxia. Journal of Cellular Biochemistry 2017; 118(10): 3237-48.
  93. L. Na, S. Masuda, S. Nagao, S. Morisaki, N. Iwamoto, K. Sakanashi. C4orf47 Contributes to the Induction of Stem-like Properties in Gallbladder Cancer Under Hypoxia. Anticancer Research 2023; 43(5): 1925-32.
  94. M. Mimeault, S.K. Batra. Hypoxia-inducing factors as master regulators of stemness properties and altered metabolism of cancer-and metastasis-initiating cells. Journal of Cellular and Molecular Medicine 2013; 17(1): 30-54.
  95. D. Zhang, L. Yang, X. Liu, J. Gao, T. Liu, Q. Yan. Hypoxia modulates stem cell properties and induces EMT through N-glycosylation of EpCAM in breast cancer cells. Journal of Cellular Physiology 2020; 235(4): 3626-33.
  96. P. Wang, S. Gong, B. Liao, J. Pan, J. Wang, D. Zou. HIF1α/HIF2α induces glioma cell dedifferentiation into cancer stem cells through Sox2 under hypoxic conditions. Journal of Cancer 2022; 13(1): 1-14.
  97. S. Cheng, M. Guo, Z. Liu, Y. Fu, H. Wu, C. Wang. Morphine Promotes the Angiogenesis of Postoperative Recurrent Tumors and Metastasis of Dormant Breast Cancer Cells. Pharmacology 2019; 104(5-6): 276-86.
  98. B.D. Khan, H.N. Chau, N.T. Nhi. Roles of hypoxia in tumor progression and novel strategies for cancer treatment. Biomedical Research and Therapy 2022; 9(10): 5361-74.
  99. S. Nagao, H. Onishi, M. Kawamoto, S. Masuda, L. Na, S. Morisaki. C4orf47 contributes to the dormancy of pancreatic cancer under hypoxic conditions. Journal of Cancer 2023; 14(2): 306-17.
  100. L. Dai, H. Xian, H. Wang, M. Li, M. Zhang, X. Liang. Hypoxia induced cell dormancy of salivary adenoid cystic carcinoma through miR-922/DEC2 axis. Translational Oncology 2024; 40: 101868.
  101. V.K. Gupta, N.S. Sharma, B. Durden, V.T. Garrido, K. Kesh, D. Edwards. Hypoxia-driven oncometabolite L-2HG maintains stemness-differentiation balance and facilitates immune evasion in pancreatic cancer. Cancer Research 2021; 81(15): 4001-13.
  102. X. Zhou, X. Liu, X. Wan, M. Xu, R. Wang, D. Yang. Oxidized ATM governs stemness of breast cancer stem cell through regulating ubiquitylation and acetylation switch. Biochemical and Biophysical Research Communications 2024; 691: 149243.
  103. Y. Yang, T. Sun, X. Xue, H. Tan, Y. Li, W. Yang. HIG-2 promotes glioma stemness and radioresistance mediated by IGFBP2-rich microparticles in hypoxia. Apoptosis : An International Journal on Programmed Cell Death 2025; 30(1): 297-319.
  104. B. Xiong, W. Liu, Y. Liu, T. Chen, A. Lin, J. Song. A Multi-Omics Prognostic Model Capturing Tumor Stemness and the Immune Microenvironment in Clear Cell Renal Cell Carcinoma. Biomedicines 2024; 12(10): 2171.
  105. L. Pathak, B. Pal, J. Talukdar, P.J. Saikia, S. Sandhya, W. Tasabehji. Hypoxia-driven mobilization of altruistic cancer stem cells in platinum-treated head and neck cancer. Frontiers in Immunology 2024; 15: 1336882.
  106. J. Takei, N. Fukasawa, T. Tanaka, Y. Yamamoto, R. Tamura, H. Sasaki. Impact of neoadjuvant bevacizumab on neuroradiographic response and histological findings related to tumor stemness and the hypoxic tumor microenvironment in glioblastoma: paired comparison between newly diagnosed and recurrent glioblastomas. Frontiers in Oncology 2022; 12: 898614.
  107. S.Y. Park, J.S. Nam. The force awakens: metastatic dormant cancer cells. Experimental & Molecular Medicine 2020; 52(4): 569-81.
  108. M. Chaterjee, K.L. van Golen. Breast cancer stem cells survive periods of farnesyl-transferase inhibitor-induced dormancy by undergoing autophagy. Bone marrow research 2011; 2011(11): 362938.
  109. A. La Belle Flynn, B.C. Calhoun, A. Sharma, J.C. Chang, A. Almasan, W.P. Schiemann. Autophagy inhibition elicits emergence from metastatic dormancy by inducing and stabilizing Pfkfb3 expression. Nature Communications 2019; 10(1): 3668.
  110. L. Vera-Ramirez, S.K. Vodnala, R. Nini, K.W. Hunter, J.E. Green. Autophagy promotes the survival of dormant breast cancer cells and metastatic tumour recurrence. Nature Communications 2018; 9(1): 1944.
  111. Q. Wang. Autophagy is indispensable for the self-renewal and quiescence of ovarian cancer spheroid cells with stem cell-like properties. Oxidative medicine and cellular longevity 2018; 2018(1): 7010472.
  112. A. Brunel, S. Hombourger, E. Barthout, S. Battu, D. Kögel, P. Antonietti. Autophagy inhibition reinforces stemness together with exit from dormancy of polydisperse glioblastoma stem cells. Aging 2021; 13(14): 18106-30.
  113. B. Levine. Autophagy and cancer. Nature 2007; 446(7137): 745-7.
  114. W. Bu, X. Hao, T. Yang, J. Wang, Q. Liu, X. Zhang. Autophagy contributes to the maintenance of genomic integrity by reducing oxidative stress. Oxidative Medicine and Cellular Longevity 2020; 2020(1): 2015920.
  115. M. Shimonosono, K. Tanaka, S. Flashner, S. Takada, N. Matsuura, Y. Tomita. Alcohol metabolism enriches squamous cell carcinoma cancer stem cells that survive oxidative stress via autophagy. Biomolecules 2021; 11(10): 1479.
  116. H.F. Aqbi, L. Tyutyunyk-Massey, R.C. Keim, S.E. Butler, T. Thekkudan, S. Joshi. Autophagy-deficient breast cancer shows early tumor recurrence and escape from dormancy. Oncotarget 2018; 9(31): 22113-22.
  117. S. Dwyer, J. Ruth, H.E. Seidel, A.A. Raz, L.A. Chodosh. Autophagy is required for mammary tumor recurrence by promoting dormant tumor cell survival following therapy. Breast Cancer Research : BCR 2024; 26(1): 143.
  118. C.J. Bailey. Metformin: historical overview. Diabetologia 2017; 60(9): 1566-76.
  119. H. Akrami, F. Mahmoodi, S. Havasi, A. Sharifi. PlGF knockdown inhibited tumor survival and migration in gastric cancer cell via PI3K/Akt and p38MAPK pathways. Cell Biochemistry and Function 2016; 34(3): 173-80.
  120. E. Koustas, E.M. Trifylli, P. Sarantis, N.I. Kontolatis, C. Damaskos, N. Garmpis. The Implication of Autophagy in Gastric Cancer Progression. Life (Basel, Switzerland) 2021; 11(12): 1304.
  121. G. Domenici, I. Aurrekoetxea-Rodríguez, B.M. Simões, M. Rábano, S.Y. Lee, J.S. Millán. A Sox2/Sox9 signalling axis maintains human breast luminal progenitor and breast cancer stem cells. Oncogene 2019; 38(17): 3151-69.
  122. L. Wang, Z. Zhang, X. Yu, X. Huang, Z. Liu, Y. Chai. Unbalanced YAP\SOX9 circuit drives stemness and malignant progression in esophageal squamous cell carcinoma. Oncogene 2019; 38(12): 2042-55.
  123. X. Yuan, J. Li, C. Coulouarn, T. Lin, L. Sulpice, D. Bergeat. SOX9 expression decreases survival of patients with intrahepatic cholangiocarcinoma by conferring chemoresistance. British Journal of Cancer 2018; 119(11): 1358-66.
  124. S. Malladi, D.G. Macalinao, X. Jin, L. He, H. Basnet, Y. Zou. Metastatic latency and immune evasion through autocrine inhibition of WNT. Cell 2016; 165(1): 45-60.
  125. L. He, H. Zhu, S. Zhou, T. Wu, H. Wu, H. Yang. Wnt pathway is involved in 5-FU drug resistance of colorectal cancer cells. Experimental & Molecular Medicine 2018; 50(8): 1-12.
  126. M. Kobune, H. Chiba, J. Kato, K. Kato, K. Nakamura, Y. Kawano. Wnt3/RhoA/ROCK signaling pathway is involved in adhesion-mediated drug resistance of multiple myeloma in an autocrine mechanism. Molecular Cancer Therapeutics 2007; 6(6): 1774-84.
  127. X. Wu, F. Luo, J. Li, X. Zhong, K. Liu. Tankyrase 1 inhibitior XAV939 increases chemosensitivity in colon cancer cell lines via inhibition of the Wnt signaling pathway. International Journal of Oncology 2016; 48(4): 1333-40.
  128. L. Liu, H. Zhu, Y. Liao, W. Wu, L. Liu, L. Liu. Inhibition of Wnt/β-catenin pathway reverses multi-drug resistance and EMT in Oct4+/Nanog+ NSCLC cells. Biomedicine & Pharmacotherapy 2020; 127: 110225.
  129. J. Fu. Wnt/β-catenin inhibition reverses multidrug resistance in pediatric acute lymphoblastic leukemia. Oncology Reports 2019; 41(2): 1387-94.
  130. F. Guo, Z. Cao, H. Guo, S. Li. The action mechanism of lncRNA-HOTAIR on the drug resistance of non-small cell lung cancer by regulating Wnt signaling pathway. Experimental and Therapeutic Medicine 2018; 15(6): 4885-9.
  131. A.C. Ranganathan, L. Zhang, A.P. Adam, J.A. Aguirre-Ghiso. 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 Research 2006; 66(3): 1702-11.
  132. L. Xie, R. Huang, H. Huang, X. Liu, J. Yu. Transcriptomics and metabolomics identify drug resistance of dormant cell in colorectal cancer. Frontiers in Pharmacology 2022; 13: 879751.
  133. L. Wang, Z. Shang, Y. Zhou, X. Hu, Y. Chen, Y. Fan. Autophagy mediates glucose starvation-induced glioblastoma cell quiescence and chemoresistance through coordinating cell metabolism, cell cycle, and survival. Cell Death {&}amp; Disease 2018; 9(2): 213.
  134. J.M. Mora-Rodríguez, B.G. Sánchez, A. Sebastián-Martín, A. Díaz-Yuste, M. Sánchez-Chapado, A.M. Palacín. Resistance to 2-Hydroxy-Flutamide in Prostate Cancer Cells Is Associated with the Downregulation of Phosphatidylcholine Biosynthesis and Epigenetic Modifications. International Journal of Molecular Sciences 2023; 24(21): 15626.
  135. Y. Kinoshita, T. Kalir, J. Rahaman, P. Dottino, D.S. Kohtz. Alterations in nuclear pore architecture allow cancer cell entry into or exit from drug-resistant dormancy. The American Journal of Pathology 2012; 180(1): 375-89.
  136. S. Ebinger, E.Z. Özdemir, C. Ziegenhain, S. Tiedt, C. Castro Alves, M. Grunert. Characterization of rare, dormant, and therapy-resistant cells in acute lymphoblastic leukemia. Cancer Cell 2016; 30(6): 849-62.
  137. A. Recasens, L. Munoz. Targeting cancer cell dormancy. Trends in Pharmacological Sciences 2019; 40(2): 128-41.
  138. C. Davies, H. Pan, J. Godwin, R. Gray, R. Arriagada, V. Raina. Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial. Lancet 2013; 381(9869): 805-16.
  139. S. Sharma, X. Pei, F. Xing, S.Y. Wu, K. Wu, A. Tyagi. Regucalcin promotes dormancy of prostate cancer. Oncogene 2021; 40(5): 1012-26.
  140. A.A. Shmakova, P.S. Klimovich, K.D. Rysenkova, V.S. Popov, A.S. Gorbunova, A.A. Karpukhina. Urokinase Receptor uPAR Downregulation in Neuroblastoma Leads to Dormancy, Chemoresistance and Metastasis. Cancers (Basel) 2022; 14(4): 994.
  141. C.T. Ho. Snail induces dormancy in disseminated luminal type A breast cancer through Src inhibition. American Journal of Cancer Research 2022; 12(8): 3932.
  142. B. O'leary, R.S. Finn, N.C. Turner. Treating cancer with selective CDK4/6 inhibitors. Nature Reviews. Clinical Oncology 2016; 13(7): 417-30.
  143. M.E. Clements, L. Holtslander, C. Edwards, V. Todd, S.D. Dooyema, K. Bullock. HDAC inhibitors induce LIFR expression and promote a dormancy phenotype in breast cancer. Oncogene 2021; 40(34): 5314-26.
  144. M.A. Essers, A. Trumpp. Targeting leukemic stem cells by breaking their dormancy. Molecular Oncology 2010; 4(5): 443-50.
  145. M. Pajic, S. Blatter, C. Guyader, M. Gonggrijp, A. Kersbergen, A. Küçükosmanoğlu. Selected alkylating agents can overcome drug tolerance of G0-like tumor cells and eradicate BRCA1-deficient mammary tumors in mice. Clinical Cancer Research 2017; 23(22): 7020-33.
  146. L.H. El Touny, A. Vieira, A. Mendoza, C. Khanna, M.J. Hoenerhoff, J.E. Green. Combined SFK/MEK inhibition prevents metastatic outgrowth of dormant tumor cells. The Journal of Clinical Investigation 2014; 124(1): 156-68.
  147. J. Hu, F.J. Sánchez-Rivera, Z. Wang, G.N. Johnson, Y. Ho, K. Ganesh. STING inhibits the reactivation of dormant metastasis in lung adenocarcinoma. Nature 2023; 616(7958): 806-13.
  148. Q. Lan, S. Peyvandi, N. Duffey, Y.T. Huang, D. Barras, W. Held. Type I interferon/IRF7 axis instigates chemotherapy-induced immunological dormancy in breast cancer. Oncogene 2019; 38(15): 2814-29.
  149. X.X. Han, S. Jin, L.M. Yu, M. Wang, X.Y. Hu, D.Y. Hu. Interferon-beta inhibits human glioma stem cell growth by modulating immune response and cell cycle related signaling pathways. Cell Regeneration (London, England) 2022; 11(1): 23.
  150. K.J. Kurppa. Treatment-induced tumor dormancy through YAP-mediated transcriptional reprogramming of the apoptotic pathway. Cancer cell 2020; 37(1): 104--122. e12.
  151. K.H. Chang, M. Basyal, Z. Alaniz, D. Honma, S. Tsutsumi, C.E. Dos Santos. Targeting EZH1/2 to Restore Chemosensitivity in Dormant Myeloid Leukemia Stem Cells. Blood 2021; 138: 2943.
  152. H. Akiyama, Y. Nishida, K.H. Chang, A.D. Bedoy, M. Muftuoglu, W. Ma. Dual targeting of EZH2 and EZH1 drives exit of leukemia stem cells from quiescence and potentiates chemotherapy in acute myeloid leukemia. Blood Cancer Journal 2025; 15(1): 76.
  153. D.K. Singh. Epigenetic reprogramming of DCCs into dormancy suppresses metastasis via restored TGFβ–SMAD4 signaling. bioRxiv 2021; 2021: 454684.
  154. R.E. Shor, J. Dai, S.Y. Lee, L. Pisarsky, I. Matei, S. Lucotti. The PI3K/mTOR inhibitor Gedatolisib eliminates dormant breast cancer cells in organotypic culture, but fails to prevent metastasis in preclinical settings. Molecular Oncology 2022; 16(1): 130-47.
  155. E.T. Goddard. Immune evasion of dormant disseminated tumor cells is due to their scarcity and can be overcome by T cell immunotherapies. Cancer Cell 2024; 42(1): 119-134. e12.
  156. S.J. Buczacki, S. Popova, E. Biggs, C. Koukorava, J. Buzzelli, L. Vermeulen. Itraconazole targets cell cycle heterogeneity in colorectal cancer. The Journal of Experimental Medicine 2018; 215(7): 1891-912.
  157. W. Liu, A.H. Kovacs, J. Hou. Cancer Cells in Sleep Mode: Wake Them to Eliminate or Keep Them Asleep Forever?. Cells 2024; 13(23): 2022.
  158. R. Liu, Y. Zhao, S. Su, A. Kwabil, P.C. Njoku, H. Yu. Unveiling cancer dormancy: intrinsic mechanisms and extrinsic forces. Cancer Letters 2024; 591:
  159. W.A. Weston, A.R. Barr. A cell cycle centric view of tumour dormancy. British Journal of Cancer 2023; 129(10): 1535-45.
  160. X. Su, Y. Li, Y. Ren, M. Cao, G. Yang, J. Luo. A new strategy for overcoming drug resistance in liver cancer: epigenetic regulation. Biomedicine & Pharmacotherapy 2024; 176: 116902.
  161. K.K. Payne. Cellular stress responses and metabolic reprogramming in cancer progression and dormancy. Seminars in cancer biology 2022; 78: 45-48.
  162. A. Mahmoud, K. Ganesh. Mouse models of metastasis and dormancy. Cold Spring Harbor Perspectives in Medicine 2024; 14(8): a041386.
  163. G.G. Bushnell, A.P. Deshmukh, P. den Hollander, M. Luo, R. Soundararajan, D. Jia. Breast cancer dormancy: need for clinically relevant models to address current gaps in knowledge. NPJ Breast Cancer 2021; 7(1): 66.
  164. E. Risson, A.R. Nobre, V. Maguer-Satta, J.A. Aguirre-Ghiso. The current paradigm and challenges ahead for the dormancy of disseminated tumor cells. Nature Cancer 2020; 1(7): 672-80.
  165. E. Boydell, M. Borgeaud, P. Tsantoulis. Dormant Tumor Cells: Current Opportunities and Challenges in Clinical Practice. Onco 2025; 5(1): 3.
  166. J. Ingham, J.L. Ruan, M.A. Coelho. Breaking barriers: we need a multidisciplinary approach to tackle cancer drug resistance. BJC Reports 2025; 3(1): 11.
  167. J. Agudo, J.A. Aguirre-Ghiso, M. Bhatia, L.A. Chodosh, A.L. Correia, C.A. Klein. Targeting cancer cell dormancy. Nature Reviews. Cancer 2024; 24(2): 97-104.

Comments