Open Access

Decoding the role of cancer stem cells in digestive tract tumors: Mechanisms and therapeutic implications (Review)

  • Authors:
    • Xiansheng Cao
    • Xuejing Geng
    • Chunlei Zhang
    • Lei Li
  • View Affiliations

  • Published online on: June 25, 2025     https://doi.org/10.3892/ijo.2025.5767
  • Article Number: 61
  • Copyright: © Cao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer stem cells (CSCs) are a distinct subpopulation of cells within tumors, characterized by their ability to self‑renew, differentiate and promote tumorigenesis. CSCs have critical roles in the initiation, progression and therapeutic resistance of digestive tract tumors, including in esophageal, gastric, colorectal and pancreatic cancer. The present review comprehensively explores the biology of CSCs, their interactions with the tumor microenvironment and their clinical relevance in predicting patient prognosis and guiding treatment strategies. The emerging therapeutic approaches that target CSCs, including pathway inhibitors, monoclonal antibodies and combination therapies, are also discussed, highlighting the potential of these strategies to improve patient outcomes in digestive tract cancer types. Additionally, future research directions and challenges in developing effective CSC‑targeted therapies are addressed, emphasizing the need for innovative strategies to overcome treatment resistance and increase therapeutic efficacy.

Introduction

Cancer stem cells (CSCs) are a specialized subset of tumor cells with properties akin to those of normal stem cells, such as self-renewal (the ability to generate identical copies of themselves) and differentiation (the capacity to form diverse cell types). Unlike normal stem cells, which maintain tissue balance, CSCs drive uncontrolled tumor growth and are linked to treatment resistance and relapse (1). These cells are hypothesized to be responsible for tumor initiation, progression and resistance to conventional therapies (2). Over the past decade, extensive research has focused on understanding the biology of CSCs and their role in cancer progression. CSCs are believed to maintain tumor growth through their unique self-renewal capacity and resistance to apoptosis, making them critical targets for therapeutic intervention (1). Studies have identified specific surface markers and signaling pathways that regulate CSC behavior, such as the Wnt, Notch and Hedgehog pathways (1,3). These findings provide insights into the mechanisms that underlie CSC-driven tumorigenesis and highlight potential therapeutic strategies to target these cells.

Digestive tract tumors, including esophageal, gastric, colorectal and pancreatic cancer, are among the most common and aggressive malignancies worldwide (4). According to the latest global cancer statistics, gastric cancer is the fifth most common cancer and the third leading cause of cancer-related death, whereas colorectal cancer is the third most common cancer globally (4). Esophageal cancer and pancreatic cancer are also associated with high mortality rates (4). These tumors often exhibit high rates of recurrence and metastasis, which contribute to poor patient outcomes (5). Previous studies have implicated CSCs in the pathogenesis and progression of digestive tract tumors. For example, CSCs have been shown to drive tumor initiation, promote metastasis and contribute to therapeutic resistance in gastric and colorectal cancer (6,7). The identification of CSC-specific markers and pathways has provided new avenues for developing targeted therapies aimed at improving patient survival and quality of life.

Research on CSCs in digestive tract tumors has advanced significantly in recent years. Studies have identified specific CSC markers, such as CD44, CD133 and leucine rich repeat containing G protein-coupled receptor 5 (LGR5), which are associated with tumor aggressiveness and poor prognosis (8,9). Furthermore, high expression of high mobility group box 2 (HMGB2), a non-histone chromatin-binding protein, has been linked to poor survival outcomes in patients with gastric cancer and colorectal cancer. A meta-analysis of multiple studies revealed that elevated HMGB2 is associated with a shorter overall survival (OS) time in patients with digestive cancer, highlighting its potential as a prognostic biomarker and therapeutic target (10). Additionally, research has explored the role of CSCs in promoting epithelial-mesenchymal transition (EMT) and metastasis in hepatocellular carcinoma and pancreatic ductal adenocarcinoma. These findings underscore the importance of CSCs in driving tumor progression and therapeutic resistance in digestive tract cancer (11,12).

The present review aims to comprehensively explore the biology of CSCs in digestive tract tumors, focusing on their role in tumorigenesis, progression and therapeutic resistance. The current understanding of CSC-specific markers and signaling pathways are critically evaluated, highlighting their clinical relevance in predicting patient outcomes and guiding treatment strategies. Furthermore, the emerging therapeutic approaches that target CSCs, including small molecule inhibitors, monoclonal antibodies and combination therapies are discussed. By integrating findings from preclinical studies and clinical trials, the present review seeks to provide insights into the potential of CSC-targeted therapies to improve patient outcomes in digestive cancer. Future research directions and challenges in developing effective CSC-targeted therapies will also be addressed, emphasizing the need for innovative strategies to overcome treatment resistance and increase therapeutic efficacy.

Biology of CSCs

The biology of CSCs is complex and multifaceted, involving intricate interactions between genetic and epigenetic factors, signaling pathways and the tumor microenvironment (TME) (13). Understanding these mechanisms is crucial for developing effective therapeutic strategies targeting CSCs and improving patient outcomes in digestive cancer.

Definition and characteristics

CSCs are a distinct subpopulation of cancer cells characterized by their ability to self-renew, differentiate into various cell types and drive tumorigenesis. CSCs are hypothesized to be responsible for tumor initiation, progression and resistance to conventional therapies (1). Normal stem cells in tissues (such as the gut lining or bone marrow) are strictly controlled to replace damaged cells and maintain organ function. By contrast, CSCs acquire genetic and epigenetic abnormalities that disrupt this regulation, allowing them to proliferate uncontrollably and spread to other organs (metastasis) (2). Studies have shown that CSCs can repopulate tumors even after therapy, making them a critical target for therapeutic intervention (14,15). For example, in gastric cancer, CSCs have been implicated in therapeutic resistance and tumor relapse, highlighting their role in maintaining tumor growth and heterogeneity (16).

Identification and markers

The identification and characterization of CSCs rely on specific surface markers and functional assays. Surface markers are proteins on cell membranes that can act as 'barcodes' to identify specific cell types. In CSCs, markers such as CD44, CD133 and aldehyde dehydrogenase 1 (ALDH1; an enzyme involved in metabolism) are often upregulated and are correlated with aggressive tumor behavior. For example, CD44+ CSCs in colorectal cancer show enhanced ability to initiate tumors and resist drugs (17,18). CD44, a transmembrane glycoprotein, is a defining CSCs marker in multiple cancer types, including breast, colorectal and head and neck malignancies. CD44+ CSCs exhibit enhanced tumor-initiating capacity and resistance to conventional therapies (19). CD133 (prominin-1) is another widely studied marker, implicated in CSC tumorigenicity in glioblastoma, liver and pancreatic cancer. CD133+ cells exhibit increased sphere-forming ability and heightened tumorigenicity in xenograft models (20). ALDH1 is an intracellular enzyme frequently used to identify CSCs in breast, lung and prostate cancer. High ALDH1 activity is linked to enhanced self-renewal, chemoresistance and tumor initiation (20). However, the variability in marker expression across different cancer types underscores the need for context-dependent analysis of CSC markers.

While CD44 and CD133 remain widely used CSC markers, their expression varies significantly across digestive tumor subtypes. For instance, CD44+ cells dominate in colorectal cancer, whereas CD133+ populations are more prevalent in pancreatic CSCs (21). Notably, Tian et al (22) demonstrated heterogeneous CD44/CD133 expression in esophageal squamous cell carcinoma (ESCC), with only 40% of tumors showing co-expression, questioning their universal applicability. Single-cell sequencing further revealed that ALDH1+ gastric CSCs (GCSCs) comprise distinct subclones with divergent Wnt and Notch dependencies (23), underscoring the need for context-specific marker validation.

Mechanisms of CSC maintenance

The maintenance of CSCs is regulated by complex molecular pathways and influenced by both genetic and epigenetic factors. Key signaling pathways include Wnt/β-catenin (24), Notch (25) and Hedgehog (26), which are critical for CSC self-renewal and differentiation. For instance, aberrant activation of the Wnt/β-catenin pathway leads to β-catenin accumulation and nuclear translocation, activating genes associated with proliferation, invasion and survival (27). This dysregulation is observed in colorectal and breast cancer, where nuclear β-catenin expression is correlated with increased tumor aggressiveness and recurrence (28,29). Epigenetic factors, such as DNA methylation and histone modification, also serve significant roles in maintaining CSCs properties (30). Studies have shown that epigenetic modifications can silence tumor suppressor genes and activate oncogenes, contributing to CSC maintenance and tumorigenesis (31,32). Additionally, genetic mutations and chromosomal instability can drive the acquisition of stem cell-like properties in cancer cells, further promoting tumor progression and therapeutic resistance (33).

Interaction between CSCs and the TME

The interaction between CSCs and the TME is a dynamic and complex process that significantly impacts tumor progression, therapeutic resistance and patient outcomes (2). Understanding these interactions is crucial for developing novel therapeutic strategies targeting both CSCs and their microenvironment to improve treatment efficacy in digestive cancer.

TME components

The TME is the 'ecosystem' surrounding cancer cells, including both living cells (such as immune cells and fibroblasts) and non-living elements [such as proteins and nutrients in the extracellular matrix (ECM)]. This environment supports tumor growth by providing nutrients, promoting blood vessel formation and shielding cancer cells from the immune system (34). Cellular components include cancer-associated fibroblasts (CAFs), immune cells [such as macrophages, T cells and natural killer (NK) cells] and endothelial cells. Non-cellular components consist of the ECM, growth factors, cytokines and metabolites (35). These components collectively influence tumor progression, metastasis and therapeutic resistance (Fig. 1). For example, CAFs promote tumor growth by secreting growth factors and remodeling the ECM, while immune cells can either suppress or enhance tumor growth depending on their polarization state (36). The ECM provides structural support and facilitates cell migration and invasion, contributing to metastasis (37).

Among the TME components, cytokines play a pivotal role in shaping the behavior of CSCs. For instance, interleukin-6 (IL-6) and IL-8 have been shown to activate critical signaling pathways such as STAT3 in CSCs, thereby promoting their self-renewal and survival (38). Matrix metalloproteinases (MMPs), which are enzymes that degrade the ECM, also contribute to CSC maintenance by facilitating tumor invasion and creating niches conducive to CSC proliferation (39). The intricate interplay between these TME components and CSCs highlights the importance of considering the microenvironment in therapeutic strategies.

CSC and TME interactions

The TME plays a critical role in supporting the survival and proliferation of CSCs. Hypoxia, a common feature of the TME, enhances the properties of CSCs by activating hypoxia-inducible factors (HIFs), which promote self-renewal and resistance to apoptosis (25) (Fig. 1). Studies have shown that hypoxia increases the expression of CSCs markers such as CD44 and ALDH1, driving tumor progression and therapeutic resistance (25,40). Additionally, angiogenesis within the TME provides nutrients and oxygen to CSCs, further promoting their survival and proliferation. For instance, HMGB2, a non-histone chromatin-binding protein, has been implicated in promoting angiogenesis and enhancing the properties of CSCs in digestive tract tumors. High HMGB2 expression is correlated with shorter OS and disease-free survival (DFS), highlighting its role in supporting CSC behavior (10).

Recent studies have also highlighted the role of the ECM in CSC maintenance (1,41). The ECM, composed of collagen, fibronectin and laminin, interacts with CSCs through integrin receptors. This interaction activates downstream signaling pathways such as focal adhesion kinase (FAK) and PI3K/AKT, which are essential for CSC survival and chemoresistance (1). For example, fibronectin has been found to enhance the stemness of colorectal CSCs by activating the FAK/PI3K/AKT pathway, underscoring the role of the ECM in maintaining CSC properties within the TME (1).

Immune evasion mechanisms

CSCs employ various strategies to evade anti-tumor immunity, often interacting with immunosuppressive cells such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) (Fig. 1). Tregs suppress anti-tumor immune responses by inhibiting cytotoxic T cells, while MDSCs promote immune tolerance through the production of immunosuppressive cytokines (42,43). CSCs can secrete factors that attract and activate these immunosuppressive cells, creating a favorable microenvironment for tumor growth (44). Furthermore, CSCs express immune checkpoint molecules such as programmed death-ligand 1 (PD-L1), which inhibit T cell activity and enhance immune evasion (45). For example, HMGB2 has been shown to promote immune escape in non-small cell lung cancer by upregulating PD-L1 expression, highlighting its role in immune evasion mechanisms (46).

Moreover, CSCs can modulate the TME to reduce the infiltration of cytotoxic T cells and NK cells. For instance, in colorectal cancer, the fatty acid desaturase 1 (FADS1)/dihydroxydodecanoic acid (DDA) axis is activated under hypoxic conditions in CSCs, impairing NK cell cytotoxicity (47). This mechanism not only facilitates immune evasion but also enhances the metastatic potential of CSCs. Additionally, CSCs can exploit metabolic pathways to alter the TME. Lactate, a byproduct of glycolysis, can drive metastasis of normoxic colorectal CSCs (CCSCs) via peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α)-dependent oxidative phosphorylation (48). This metabolic adaptation not only supports the survival of CSCs but also creates a hostile environment for immune cells.

Metabolic adaptation of CSCs

CSCs exhibit notable metabolic flexibility, enabling them to survive and thrive in the harsh TME. CSCs can switch between glycolysis and oxidative phosphorylation depending on the availability of nutrients and oxygen. For example, in hypoxic conditions, CSCs upregulate glycolysis-related enzymes and transporters to maintain energy production (25). This metabolic reprogramming is often accompanied by the activation of specific signaling pathways, such as the HIF-1α pathway, which further enhances the stemness and survival of CSCs.

Furthermore, CSCs can utilize alternative metabolic substrates to adapt to nutrient limitations. In colorectal cancer, vitamin D has been shown to promote ferroptosis in CCSCs by downregulating SLC7A11, a cystine transporter critical for redox balance (49). This suggests that CSCs can alter their metabolic pathways to resist oxidative stress and maintain cellular homeostasis. Additionally, CSCs can exploit the metabolic products of surrounding cells. For instance, in gastric cancer, prostaglandin D2 (PGD2)/prostaglandin D2 receptor 2 (PTGDR2) signaling inhibits autophagy via autophagy-related protein 4B (ATG4B) ubiquitination, reducing CSC stemness (50). This highlights the intricate interplay between metabolic adaptation and signaling pathways in CSCs.

CSCs in digestive tract tumors

CSCs play a pivotal role in the pathogenesis and progression of digestive tract tumors, driving tumorigenesis, metastasis and therapeutic resistance. Understanding the biology of CSCs and their molecular mechanisms is crucial for developing effective therapeutic strategies targeting these cells, which could significantly improve patient outcomes in digestive system cancer.

Esophageal cancer

Esophageal CSCs drive tumor initiation, progression and therapeutic resistance through diverse molecular mechanisms (Table I). This subsection synthesizes key findings on CSC-related pathways in esophageal cancer, critically evaluates their implications and highlights unresolved controversies.

Table I

Studies on the mechanism of action of cancer stem cells in esophageal cancer.

Table I

Studies on the mechanism of action of cancer stem cells in esophageal cancer.

First author, yearTargetsStudy typesMechanism of actionPhenotype in tumor(Refs.)
Chen et al, 2012DDR pathwayIn vitro and xenograftInhibits Chk1/2 activation and p53 signaling, attenuates DNA damage response and promotes CSC survivalChemoresistance and tumor recurrence(51)
Song et al, 2014YAP1/SOX9In vitro and animal modelsHippo/YAP1 pathway activates SOX9, enhancing CSC self-renewal and tumorigenicityTumor initiation and increased invasiveness(53)
Xu et al, 2016STAT3/miR-181bIn vitroSTAT3 synergizes with the miR-181b/CYLD axis to promote CSC proliferationAccelerated cell proliferation and chemoresistance(54)
Liu et al, 2019Hsp27-AKT-HK2In vitroHsp27 activates HK2-dependent glycolysis via AKT, maintaining CSC metabolism and stemnessMetabolic reprogramming and tumor growth(55)
Kai et al, 2022MYH9Preclinical studiesMYH9 activates the PI3K/AKT/mTOR axis, driving CSC oncogenicityTumorigenesis and increased invasiveness(56)
Wang et al, 2017CXCL12/CXCR4Animal modelsAutocrine CXCL12/CXCR4 signaling enhances MMP secretion, promoting ECM degradation and metastasisMetastasis dissemination(57)
Yue et al, 2015TGF-β1/SmadIn vitroTGF-β1 promotes CSC migration via Smad-dependent EMT activationEMT activation and increased invasiveness(58)
Wei et al, 2024QSOX1/PD-L1Animal modelsQSOX1 upregulates PD-L1, inhibiting CD8+ T cell infiltration and mediating immune evasionImmune evasion and dormant CSC survival(59)
Guo et al, 2022miR-637/WASHClinical sample analysismiR-637 loss activates the WASH/IL-8 pathway, enhancing CSC stemness and metastasisAccelerated metastasis and poor prognosis(60)
Xun et al, 2024RGS1/CXCR4In vitromiR-191-3p targets RGS1, activating CXCR4/PI3K/AKT signaling and maintaining CSC stemnessEnhanced stemness(61)
Yu et al, 2020DNA methylationGenome-wide analysisHypermethylation of tumor suppressor genes (such as CDH1) promotes CSC epigenetic reprogrammingIncreased tumor heterogeneity(62)
Gupta et al, 2021CD44/CD133Clinical sample analysisHeterogeneous expression of CD44/CD133 reflects the diversity of esophageal cancer subtypesControversy over marker specificity(63)
Xu et al, 2020STAT3/HSP90In vitroSTAT3 silencing enhances the cytotoxic effect of HSP90 inhibitor SNX-2112 on CSCsIncreased chemosensitivity(64)
Liu et al, 2021Hsp27/GPX4In vitroHsp27-GPX4 axis mediates ferroptosis resistance in CSCsFerroptosis resistance and enhanced survival(65)

[i] CSCs, cancer stem cells; ESCC, esophageal squamous cell carcinoma; DDR, DNA damage response; Chk1/2, checkpoint kinase 1/2; YAP1, Yes-associated protein 1; SOX9, SRY-box transcription factor 9; STAT3, signal transducer and activator of transcription 3; CYLD, cylindromatosis; Hsp27, heat shock protein 27; AKT, protein kinase B; HK2, hexokinase 2; MYH9, myosin heavy chain 9; CXCL12, C-X-C motif chemokine ligand 12; CXCR4, C-X-C motif chemokine receptor 4; MMP, matrix metalloproteinase; TGF-β1, transforming growth factor β1; QSOX1, quiescin sulfhydryl oxidase 1; PD-L1, programed death-ligand 1; WASH, Wiskott-Aldrich syndrome protein and SCAR homolog; IL-8, interleukin-8; RGS1, regulator of G-protein signaling 1; CDH1, cadherin 1; lncFERO, Long non-coding RNA FERO; GPX4, glutathione peroxidase 4.

Quiescence and attenuated DNA damage response (DDR) promote CSC survival

CSCs in ESCC exhibit a quiescent state, enabling evasion of conventional therapies targeting rapidly dividing cells. Chen et al (51) demonstrated that ESCC CSCs downregulate DDR pathways, reducing apoptosis under genotoxic stress. This quiescence is mediated by suppressed checkpoint kinase 1/2 activation and impaired p53 signaling, allowing CSCs to accumulate mutations and survive chemotherapy. However, this study relied on in vitro sphere-forming assays and xenograft models, which may not fully recapitulate the TME in human patients. Subsequent studies, such as that by Zhao et al (52), corroborated these findings but emphasized heterogeneity in DDR pathways across CSC subpopulations, suggesting context-dependent survival mechanisms.

Activation of pro-survival signaling pathways

Multiple signaling cascades converge to sustain CSC self-renewal and chemoresistance. The Hippo/Yes-associated protein 1 (YAP1) pathway is a critical regulator in esophageal cancer. Song et al (53) revealed that YAP1 upregulates SRY-box transcription factor 9, enhancing CSC properties such as tumorigenicity and spheroid formation. Similarly, Xu et al (54) identified a STAT3/miRNA (miR)-181b/cylindromatosis axis that promotes CSC proliferation by modulating IL-6/STAT3 signaling. While these studies highlight pathway-specific roles, conflicting evidence exists regarding cross-talk between pathways. For instance, Liu et al (55) linked the HSP27/AKT/hexokinase 2 (HK2) axis to CSC metabolic reprogramming, showing that AKT activation sustains stemness via HK2-dependent glycolysis. This contrasts with the study by Kai et al (56), in which myosin heavy chain 9 was implicated in activating PI3K/AKT/mTOR to drive CSC oncogenesis, suggesting overlapping yet distinct metabolic dependencies.

Cytokine autocrine loops and metastatic dissemination

Autocrine signaling mechanisms facilitate CSC invasion and metastasis in esophageal cancer. Wang et al (57) demonstrated that C-X-C motif chemokine ligand 12/C-X-C motif chemokine receptor 4 (CXCR4) axis activation in CSCs enhances MMP secretion, promoting ECM degradation and metastatic spread. Conversely, Yue et al (58) linked TGF-β1 to CSC migration via Smad-dependent EMT activation. Despite mechanistic clarity, these studies predominantly utilized monolayer cell cultures, neglecting the contribution of stromal cells in the TME. Recent work by Wei et al (59) addressed this gap, showing that quiescin sulfhydryl oxidase 1 (QSOX1) in CSCs upregulates PD-L1 to exclude CD8+ T cells, illustrating how cytokine networks synergize with immune evasion.

Epigenetic and post-transcriptional regulation

Dysregulation of non-coding RNAs and epigenetic modifiers underpins CSC plasticity in esophageal cancer. Guo et al (60) reported that miR-637 loss activates the Wiskott-Aldrich syndrome protein and SCAR homolog/IL-8 pathway, augmenting CSC stemness and metastasis. Similarly, Xun et al (61) identified miR-191-3p as a suppressor of regulator of G-protein signaling 1, which inhibits CXCR4/PI3K/AKT signaling. These findings align with genome-wide methylation analyses by Yu et al (62), which revealed hypermethylation of tumor suppressor genes (such as Cadherin 1) in CSCs. However, inconsistencies arise in biomarker specificity; for example, Gupta et al (63) found variable CD44/CD133 expression across ESCC subtypes, questioning the universality of these markers.

Therapeutic resistance and immune evasion

CSCs employ multifaceted strategies to resist treatment in esophageal cancer. Xu et al (64) showed that STAT3 silencing sensitizes CSCs to the HSP90 inhibitor, SNX-2112, while Liu et al (65) linked ferroptosis resistance to HSP27-glutathione peroxidase 4 (GPX4) upregulation. Notably, Wei et al (59) uncovered a novel immune evasion mechanism where QSOX1 elevates PD-L1, enabling CSCs to bypass cytotoxic T cell surveillance. These studies underscore the need for combinatorial therapies targeting both CSCs and immune cells, although clinical validation remains limited.

Gastric cancer

GCSCs are central to tumor initiation, metastasis and therapeutic resistance. The functional regulation of GCSCs involves intricate signaling networks, epigenetic reprogramming and interactions with the TME (Table II). In this subsection, key mechanisms governing GCSC biology are dissected, representative studies are critically evaluated and therapeutic implications are discussed.

Table II

Studies on the mechanism of action of tumor stem cells in gastric cancer.

Table II

Studies on the mechanism of action of tumor stem cells in gastric cancer.

First author, yearTargetsStudy typesMechanism of actionPhenotype in tumor(Refs.)
Xu et al, 2019BMX-ARHGAPIn vitroBMX-ARHGAP fusion protein activates the JAK/STAT3 pathway, maintaining GCSC survival Chemoresistance(67)
Wu et al, 2013 miR-19b/20a/92aIn vitromiR cluster targets PTEN, activating AKT/mTOR signaling and maintaining GCSC self-renewalEnhanced stemness(68)
Han et al, 2015ATOH1/Notch1In vitroATOH1 inhibits Notch1, inducing GCSC differentiationDifferentiation promotion and tumor suppression(69)
Shen et al, 2021ONECUT2/SOX2In vitroONECUT2 upregulates SOX2 via miR-15a-5p, enhancing GCSC stemnessEnhanced stemness(70)
Li et al, 2018 Notch/autophagyIn vitroNotch signaling induces autophagy, protecting GCSCs from 5-FU cytotoxicity Chemoresistance(71)
Xin et al, 2020RAB37 methylationIn vitroMethionine promotes RAB37 methylation, inhibiting autophagy and enhancing GCSC survivalSurvival under nutrient stress(72)
Togano et al, 2021Autophagy inhibitorsIn vitroAutophagy inhibition weakens GCSC viability in hypoxic nichesReduced survival(73)
Zhang et al, 2024PGD2/PTGDR2Animal modelsPGD2/PTGDR2 signaling inhibits autophagy via ATG4B ubiquitination, reducing GCSC stemnessReduced stemness(50)
Yang et al, 2017HIF-1α/SnailIn vitroHIF-1α induces EMT in GCSCs via Snail, promoting metastasisEMT activation and enhanced metastasis(74)
Sun et al, 2020HER2Patient-derived xenograftsHER2 promotes GCSC self-renewal and invasion; HER2 inhibition reduces tumorigenicityAttenuated invasiveness(75)
Seeneevassen et al, 2020LIF/HippoIn vitroLIF activates Hippo kinases, suppressing GCSC tumorigenicityTumor suppression(76)
Wang et al, 2017ABC transportersIn vitroALDH1+ GCSCs upregulate ABC transporters to resist docetaxel/cisplatin/5-FU treatment Chemoresistance(78)
Zhang et al, 2021lncFEROAnimal models Chemotoxicity-induced exosomal lncFERO suppresses ferroptosis, maintaining GCSC stemnessStemness maintenance(79)
Mao et al, 2024SCD1/mTORIn vitroSCD1 inhibits ferroptosis via the SQLE/cholesterol/mTOR axis, enhancing GCSC stemnessFerroptosis resistance and enhanced stemness(80)
Ni et al, 2024TGF-β/SmadAnimal modelsCelastrus orbiculatus extract inhibits the TGF-β/Smad pathway, weakening GCSC tumorigenicityTumor growth inhibition(81)

[i] GCSCs, gastric cancer stem cells; miR, microRNA; GSK3β, glycogen synthase kinase 3 β; BMX-ARHGAP, bone marrow X kinase-Rho GTPase activating protein; JAK/STAT3, Janus kinase/signal transducer and activator of transcription 3; PTEN, phosphatase and tensin homolog; ATOH1, atonal transcription factor 1; ONECUT2, one cut homeobox 2; SOX2, SRY-box transcription factor 2; 5-FU, 5-Fluorouracil; HIF-1α, hypoxia-inducible factor 1 alpha; LIF, leukemia inhibitory factor; PGD2/PTGDR2, prostaglandin D2/prostaglandin D2 receptor 2; ATG4B, autophagy-related protein 4B; SCD1, Stearoyl-CoA desaturase 1; SQLE, Squalene epoxidase; mTOR, mechanistic target of rapamycin.

Wnt/β-catenin signaling in GCSC proliferation and chemoresistance

The Wnt/β-catenin pathway is a cornerstone of GCSC self-renewal. Mao et al (66) demonstrated that β-catenin activation enhances GCSC proliferation, while salinomycin inhibits this pathway, reducing tumorigenicity in xenograft models. Similarly, Wu et al (34) identified miR-483-5p as a promoter of Wnt/β-catenin signaling, driving GCSC invasion and self-renewal via direct suppression of GSK3β. However, discrepancies exist in downstream effectors. Xu et al (67) reported that the bone marrow X kinase-Rho GTPase activating protein fusion proteins activate Janus kinase/STAT3, bypassing canonical Wnt signaling to sustain GCSC survival. These findings suggest pathway plasticity, where GCSCs utilize both canonical and non-canonical Wnt signaling to adapt to therapeutic pressures.

Epigenetic and post-transcriptional regulation of stemness

Epigenetic modifiers and miRNAs tightly regulate GCSC plasticity. Wu et al (68) revealed that the miR-19b/20a/92a cluster maintains GCSC self-renewal by targeting PTEN and activating AKT/mTOR signaling. By contrast, Han et al (69) showed that atonal transcription factor 1 (ATOH1) induces GCSC differentiation by suppressing Notch1, highlighting a tumor suppressive role of transcription factors. The detailed mechanism involves the direct interaction of ATOH1 with the Notch1 signaling pathway. Notch1 is a well-known promoter of stemness and tumorigenicity in various cancer types, including gastric cancer. By inhibiting Notch1, ATOH1 effectively reduces the self-renewal and proliferative capacity of cancer stem cells, thus exerting a tumor suppressive effect. Notably, Shen et al (70) identified one cut homeobox 2 as a miR-15a-5p target that amplifies GCSC stemness through SOX2 upregulation. Such studies underscore the dual role of epigenetic regulators in either sustaining or suppressing GCSC phenotypes, depending on cellular context.

Autophagy and metabolic adaptations

Autophagy enables GCSCs to survive stress and resist therapy. Li et al (71) linked Notch-induced autophagy to chemoresistance, where inhibition of autophagy sensitizes GCSCs to 5-fluorouracil (5-FU). Xin et al (72) further demonstrated that methionine represses autophagy by methylating RAB37, enhancing GCSC survival under nutrient deprivation. Recent work by Togano et al (73) corroborated these findings, showing that autophagy inhibitors impair GCSC viability in hypoxic niches. Conversely, Zhang et al (50) reported that PGD2/PTGDR2 signaling suppresses GCSC stemness by promoting ATG4B ubiquitination and inhibiting autophagic flux. These contrasting roles of autophagy emphasize the need for context-specific therapeutic targeting.

Pro-survival signaling and the TME

GCSCs exploit microenvironmental cues to evade immune surveillance and foster metastasis. Yang et al (74) revealed that HIF-1α induces EMT in GCSCs via Snail upregulation, facilitating dissemination under hypoxia. Sun et al (75) implicated HER2 in GCSC invasion, where HER2 inhibition reduces tumorigenicity in patient-derived xenografts. Additionally, Seeneevassen et al (76) identified leukemia inhibitory factor as a Hippo pathway activator that suppresses GCSC tumorigenicity, suggesting a dual role for cytokines in niche regulation. Recent studies also highlight the influence of the microbiome. PGD2/PTGDR2 signaling, downregulated in gastric cancer tissues, restricts GCSC self-renewal by inhibiting STAT3 phosphorylation (50,77), aligning with findings from GCSC models.

Therapeutic resistance and emerging targets

GCSCs employ diverse mechanisms to resist conventional therapies. Wang et al (78) showed that docetaxel/cisplatin/5-FU regimens fail to eradicate ALDH1+ GCSCs due to upregulated ABC transporters. Zhang et al (79) uncovered exosomal long non-coding RNA FERO as a ferroptosis suppressor, linking chemotoxicity to GCSC stemness preservation. Novel strategies targeting metabolic vulnerabilities, such as inhibition of stearoyl-CoA desaturase 1 to induce ferroptosis via cholesterol-mTOR axis disruption (80), show promise. Similarly, Ni et al (81) demonstrated that Celastrus orbiculatus extract suppresses GCSCs through TGF-β/Smad pathway inhibition.

Colorectal cancer

CCSCs are pivotal drivers of tumor initiation, progression, chemoresistance and metastasis. The unique biological properties and regulatory mechanisms of CCSCs have been extensively studied, revealing complex interactions between intrinsic signaling pathways, epigenetic modifications and the TME (Table III). In this subsection, the key mechanisms underlying CCSC functionality, supported by representative studies, are summarized and their implications for therapeutic targeting are discussed.

Table III

Studies on the mechanism of action of tumor stem cells in colorectal cancer.

Table III

Studies on the mechanism of action of tumor stem cells in colorectal cancer.

First author, yearTargetsStudy typesMechanism of actionPhenotype in tumor(Refs.)
Chen et al, 2017miR-199a/bIn vitromiR-199a/b activates Wnt/β-catenin signaling, upregulating ABCG2-mediated cisplatin efflux Chemoresistance(82)
Li et al, 2017KDM3/H3K9me2In vitroKDM3 demethylates H3K9me2, activating Wnt/β-catenin signaling and promoting stemnessTumor initiation and enhanced stemness(83)
Hua et al, 2019 TRIB3/β-cateninAnimal modelsTRIB3 stabilizes β-catenin/TCF4 complexes, amplifying CCSC stemness and EMTEnhanced metastasis(84)
Yu et al, 2017SATB2In vitroSATB2 acts as a Wnt-driven transcriptional coactivator, inducing CCSC transformationTumor initiation(85)
Zhu et al, 2021SOX2/Beclin1In vitroSOX2 mediates autophagy-dependent chemoresistance via β-catenin/Beclin1 signaling Chemoresistance(86)
Izumi et al, 2017FBXW7/c-MycPreclinical studiesFBXW7 downregulation stabilizes c-Myc, enhancing CCSC survival Chemoresistance(87)
Mukohyama et al, 2019miR-221/QKIIn vitromiR-221 targets QKI, releasing its inhibition on CCSC proliferationAccelerated(89)
Liu et al, 2022Lactate/PGC-1αAnimal modelsLactate drives metastasis of normoxic CCSCs via PGC-1α-dependent oxidative phosphorylationEnhanced liver metastasis(48)
Guo et al, 2023Vitamin D/SLC7A11In vitroVitamin D inhibits SLC7A11, inducing ferroptosis in CCSCsIncreased ferroptosis sensitivity(49)
Geng et al, 2024FADS1/DDAIn vitroUnder hypoxia, CCSCs employ the FADS1/DDA axis to impair NK cell cytotoxicityImmune evasion(47)
Liu et al, 2020CAFs ExosomesIn vitroCAFs secrete exosomes rich in TGF-β1, enhancing CCSC radioresistance via SMAD signaling Radioresistance(90)
Montalbán-Hernández et al, 2022Monocyte FusionAnimal modelsCCSCs fuse with monocytes to form hybrid cells that promote metastasis and immune evasionAccelerated metastasis(91)
Cavallucci et al, 2022Fusobacterium nucleatumIn vitroF. nucleatum directly activates proinflammatory and stemness pathways in CCSCsRemodeling of the inflammatory microenvironment(92)
Zou et al, 2022CD44/EMTIn vitroCD44 knockdown inhibits SNAIL/TWIST1, reducing CCSC invasivenessAttenuated invasiveness(94)
Vishnubalaji et al, 2018MAPK/FAKIn vitroALDH1+ CCSCs activate the MAPK/FAK pathways to resist oxidative stress and immune surveillanceResistance to oxidative stress and immune evasion(95)

[i] CCSCs, colorectal cancer stem cells; ABCG2, ATP-binding cassette subfamily G member 2; KDM3, lysine-specific demethylase 3; H3K9me2, Histone H3 lysine 9 dimethylation; TRIB3, Tribbles pseudokinase 3; TCF4, T-cell factor 4; SATB2, Special AT-rich sequence-binding protein 2; SOX2, SRY-box transcription factor 2; FBXW7, F-box and WD repeat domain-containing 7; PGC-1α, Peroxisome proliferator-activated receptor γ coactivator 1-α; SLC7A11, solute carrier family 7 member 11; FADS1, fatty acid desaturase 1; DDA, dihydroxydodecanoic acid; CAFs, cancer-associated fibroblasts; TGF-β1, Transforming growth factor β1; ALDH1, Aldehyde Dehydrogenase 1; NK, natural killer; EMT, epithelial-mesenchymal transition; TWIST1, Twist family bHLH transcription factor 1; FAK, Focal adhesion kinase.

Wnt/β-catenin signaling as a core regulatory axis

The Wnt/β-catenin pathway is a central regulator of CCSC self-renewal and chemoresistance. Chen et al (82) demonstrated that miR-199a/b upregulation in ALDH1+ CCSCs activates Wnt/β-catenin signaling, enhancing ATP-binding cassette subfamily G member 2 (ABCG2)-mediated drug efflux and cisplatin resistance. Similarly, Li et al (83) identified that lysine-specific demethylase 3 epigenetically activates Wnt/β-catenin by removing repressive H3K9 methylation marks, thereby promoting stemness and tumorigenicity. These findings align with Hua et al (84), who showed that Tribbles pseudokinase 3 stabilizes β-catenin/T-cell factor 4 complexes, amplifying stemness and EMT in CCSCs. However, discrepancies arise in the role of downstream effectors. While Yu et al (85) implicated Special AT-rich sequence-binding protein 2 as a Wnt-driven transcriptional coactivator, Zhu et al (86) highlighted SOX2-mediated β-catenin/Beclin1 crosstalk in chemoresistance, suggesting pathway plasticity across CCSC subpopulations.

Chemoresistance via ubiquitination and epigenetic modulation

CCSCs evade chemotherapy through dynamic post-translational modifications. Izumi et al (87) revealed that F-box and WD repeat domain-containing 7 (FBXW7), an E3 ubiquitin ligase, is downregulated in CCSCs, leading to c-Myc stabilization and enhanced survival under 5-FU treatment. Conversely, Honma et al (88) reported that FBXW7 upregulation degrades pro-survival proteins, sensitizing CCSCs to oxaliplatin. This paradox may reflect context-dependent roles of FBXW7 in different chemotherapeutic regimens. Epigenetically, Mukohyama et al (89) identified miR-221 as a driver of chemoresistance by targeting Quaking homolog, KH domain RNA-binding protein (QKI), a tumor suppressor that restrains CCSC proliferation. These studies underscore the need for personalized strategies targeting ubiquitination or miRNA networks.

Metabolic reprogramming and hypoxic adaptation

CCSCs exhibit metabolic flexibility to survive hostile microenvironments. Liu et al (48) demonstrated that lactate, a byproduct of glycolysis, induces PGC-1α-dependent oxidative phosphorylation in normoxic CCSCs, facilitating liver metastasis. Conversely, Guo et al (49) found that vitamin D triggers ferroptosis in CCSCs by suppressing SLC7A11, a cystine transporter critical for redox balance. These contrasting mechanisms highlight the dual role of metabolism in CCSC survival and vulnerability. Additionally, hypoxia-driven immune evasion was explored by Geng et al (47), in which it was observed that CCSCs upregulated FADS1/DDA to impair NK cell cytotoxicity, a process exacerbated by hypoxic conditions.

Interaction with the TME

The niche surrounding CCSCs notably influences their behavior. Liu et al (90) showed that CAFs secrete exosomes enriched with TGF-β1, which enhance CCSC radioresistance via Smad-dependent stemness pathways. Montalbán-Hernández et al (91) further revealed that CCSCs fuse with monocytes to form hybrid cells capable of immune evasion and metastatic dissemination. These findings align with those of Cavallucci et al (92), who identified Fusobacterium nucleatum as a pathobiont that directly activates pro-inflammatory and stemness pathways in CCSCs, emphasizing the role of the microbiome in CCSC regulation.

EMT and immune evasion

EMT is a hallmark of CCSC plasticity. Tamura et al (93) linked E-cadherin loss to Nanog upregulation, which drives CCSC proliferation. Similarly, Zou et al (94) demonstrated that CD44 knockdown suppresses EMT and invasiveness by inhibiting Snail and Twist family bHLH transcription factor 1. Immune evasion mechanisms were highlighted by Vishnubalaji et al (95), where ALDH1+ CCSCs were shown to activate MAPK and FAK pathways to resist oxidative stress and immune surveillance. These studies collectively suggest that targeting EMT regulators or immune checkpoints could disrupt CCSC dissemination.

Clinical relevance of CSCs in digestive tract tumors

CSCs significantly influence clinical outcomes in digestive tract tumors by driving therapeutic resistance, recurrence and metastasis. This section evaluates their prognostic value, mechanisms of drug resistance and emerging therapeutic strategies in clinical trials, integrating findings from key studies to highlight translational implications.

Prognostic significance

CSC-associated markers serve as robust predictors of patient survival and treatment response. In esophageal cancer, CD44 and CD133 expression is associated with resistance to neoadjuvant chemotherapy and poor survival. Specifically, Agawa et al (96) demonstrated that CD44+/CD133+ CSCs in ESCC predict poor pathological response to cisplatin/5-FU regimens, with a 3-year survival rate of 28 vs. 72% for marker-negative patients. Similarly, Claudin 4 (CLDN4)-high ESCC cells exhibit stem-like properties and resistance to concurrent chemoradiotherapy, as shown by Lin et al (97), linking CLDN4 to reduced OS [hazard ratio (HR), 2.1; P=0.003]. Meta-analyses by Trevellin et al (98) further confirmed that CSC markers (such as ALDH1 and CD44) are consistently associated with shorter DFS across esophageal and gastric cancer.

In gastric cancer, ALDH1+ CSCs are linked to chemoresistance and recurrence. Nishikawa et al (99) reported that ALDH1+ cells survive 5-FU treatment due to upregulated ABCG2, with ALDH1 positivity correlating with a 2.3-fold increased risk of relapse. Gong et al (100) identified epithelial cell transforming 2 (ECT2) as a novel prognostic marker, where high ECT2 expression promotes stemness via Wnt/β-catenin signaling and predicts poor differentiation (HR, 1.9; P=0.01). Circulating GCSCs detected in peripheral blood are also correlated with advanced tumor stage and metastasis (P<0.001) (101).

For colorectal cancer, Catalano et al (102) revealed that thyroid hormone receptor activation reduces CSC viability by suppressing the Wnt and bone morphogenetic protein 4 pathways, improving survival in patients with low Nanog expression (HR, 0.6; P=0.04). Conversely, Prieur et al (103) demonstrated that anti-progastrin antibodies targeting Wnt-driven CSCs enhance chemosensitivity in KRAS-mutated colorectal cancer, with a 40% reduction in tumor sphere formation.

Drug resistance

CSCs evade therapy through intrinsic and extrinsic mechanisms. In esophageal cancer, ABT-263 (a BCL-2 inhibitor) synergizes with chemotherapy by depleting CSCs via apoptosis induction, achieving a 60% reduction in tumor volume in xenografts (104). Nanog, a stemness marker, mediates resistance to cisplatin; iron chelators targeting Nanog reduce chemoresistance by 50% in vitro (105).

GCSCs employ metabolic adaptations and epigenetic reprogramming. Xu et al (106) showed that 5-FU enriches ALDH1+ CSCs through reactive oxygen species-mediated autophagy, while methionine restriction sensitizes CSCs by inhibiting RAB37 methylation (107). HNRNPA2B1, an RNA-binding protein, stabilizes c-Myc mRNA in CSCs, and its knockdown restores oxaliplatin sensitivity (P<0.01) (108).

In colorectal cancer, dual PI3K/mTOR inhibitors induce the differentiation of CD133+ CSCs, reducing tumorigenicity by 70% (109). CD44+/CD133+ CSCs are paradoxically sensitive to trifluridine (110), suggesting metabolic vulnerabilities. However, sphingosine kinase 1/HIF-1 axis inhibition (111) and aurora kinase A/YAP1) targeting (112) overcome microenvironment-driven resistance, suppressing metastasis by 45-60%.

Therapeutic targeting of CSCs

The development of therapies targeting CSCs in digestive tract tumors has gained momentum, with strategies focusing on disrupting CSC-specific pathways, enhancing drug delivery and overcoming therapeutic resistance (Table IV). This section critically evaluates current approaches, integrating findings from preclinical and clinical studies, and discusses challenges and future directions.

Table IV

Studies on the relevance of tumor stem cells in the treatment of digestive tract tumors.

Table IV

Studies on the relevance of tumor stem cells in the treatment of digestive tract tumors.

First author, yearIntervening measuresTumorStudy typesTargetsSignaling pathwaysClinical value(Refs.)
Akrami et al, 2018IbuprofenGastric cancerIn vitro and animal modelsWnt/β-cateninInhibits Wnt/β-catenin signaling, reducing proliferation and tumorigenicityPotential for chemoprevention and adjuvant therapy(113)
Chen et al, 2018Phenethyl isothiocyanateColorectal cancerIn vitro and animal modelsWnt/β-cateninDownregulates Wnt/β-catenin, impairing sphere formation and chemoresistanceEnhances chemotherapeutic efficacy(114)
Chen et al, 2017(-)-EGCGColorectal cancerIn vitro and animal modelsWnt/β-cateninSuppresses Wnt/β-catenin pathwayPotential for chemoprevention(115)
Qi et al, 2022TET1/FOXO4Gastric cancerIn vitro and animal modelsWnt/β-cateninInhibits metastasis and self-renewalPotential for metastasis prevention(116)
Cao et al, 2021ApatinibGastric cancerIn vitro and animal modelsSonic HedgehogReduces stemness markers (such as CD44 and ALDH1) and tumor growthPromising in advanced gastric cancer treatment(119)
Yang et al, 2020miR-135aEsophageal cancerIn vitroSmoothenedTargets Hedgehog signaling pathway, inhibiting invasion and migrationPromising for targeted therapy(120)
Giraud et al, 2020VerteporfinGastric cancerIn vitro and animal modelsYAP1/TAZ-TEADDisrupts transcriptional activity, reducing spheroid formation and metastasisPotential for metastasis prevention(121)
Jang et al, 2020Tankyrase inhibitorsColorectal cancerIn vitro and animal modelsAXIN, c-KITDownregulates c-KIT via AXIN, impairing viabilityOvercomes resistance to MEK inhibitors(122)
Hu et al, 2024Src signaling inhibitorsGastric cancerIn vitroSTAT3, AKTBlocks stemness gene expression and proliferationEnhances efficacy of targeted therapies(123)
Song et al, 2021Pan-BCL-2 inhibitorGastroesophageal carcinomaIn vitro and animal modelsBCL-2 family proteinsInduces apoptosis in CD44+ CSCsSynergizes with chemotherapy(125)
Park et al, 2020Caffeic acidColorectal cancerIn vitroPI3K/AktBlocks PI3K/Akt signaling axisEnhances chemotherapeutic response(126)
Wen et al, 2015EvodiamineGastric cancerIn vitroWnt pathwayInhibits self-renewalPotential for adjuvant therapy(117)
Sun et al, 2024CinobufaciniGastric cancerIn vitro and animal models AKT/GSK-3β/β-cateninEnhances therapeutic response of 5-FUOvercomes chemoresistance(118)
Yao et al, 2014Hyaluronic acid functionalized carbon nanotubes loaded with salinomycinGastric cancerIn vitro-Selectively kills GCSCsEnhances targeted delivery(127)
Andrade et al, 2021Polymeric micelles targeting CD44v6 receptorColorectal cancerIn vitro and animal modelsCD44v6Increases niclosamide efficacy and reduces CTCsEnhances targeted therapy(128)
Quarni et al, 2019Mithramycin AColorectal cancerIn vitro-Inhibits colorectal cancer growthPotential for monotherapy or combination therapy(129)
AlShamaileh et al, 2017Aptamer-mediated survivin RNAiColorectal cancerIn vitroSurvivinEnables 5-FU to eliminate CCSCsEnhances chemotherapeutic efficacy(130)

[i] GCSCs, gastric cancer stem cells; CCSCs, colorectal cancer stem cells; EGCG, (-)-epigallocatechin-3-gallate; PI3K/Akt, phosphatidylinositol 3-kinase/Akt; YAP1/TAZ-TEAD, Yes-associated protein 1/transducer of transcription-TEA domain family transcription factor; AHCC/ETAS, active hexose correlated compound/epigallocatechin gallate; ALDH1, aldehyde dehydrogenase 1; CTCs, circulating tumor cells; 5-FU, 5-fluorouracil.

Targeting CSC-specific pathways
Wnt/β-catenin pathway

The Wnt/β-catenin pathway is normally active during embryonic development to guide cell fate decisions. Multiple Wnt/β-catenin pathway inhibitors have shown efficacy in gastric and colorectal cancer. For instance, ibuprofen suppresses GCSCs by inhibiting Wnt/β-catenin signaling, reducing proliferation and tumorigenicity in vitro and in vivo (113) (Fig. 2). Similarly, phenethyl isothiocyanate and epigallocatechin-3-gallate downregulate Wnt/β-catenin in CCSCs, impairing sphere formation and chemoresistance (114,115). However, variability exists in downstream effects; for example, TET1/FOXO4-mediated Wnt inhibition suppresses metastasis but fails to eliminate quiescent GCSCs (116), highlighting the need for combinatorial approaches. Furthermore, evodiamine and cinobufagin inhibit GCSC self-renewal via the Wnt/β-catenin and AKT/GSK-3β pathways, respectively (117,118). Notably, cinobufagin enhances 5-FU sensitivity by suppressing β-catenin nuclear translocation (118), illustrating the promise of phytochemicals in overcoming chemoresistance.

Hedgehog pathway

Apatinib, a tyrosine kinase inhibitor, targets Sonic Hedgehog in GCSCs, reducing stemness markers (such as CD44 and ALDH1) and tumor growth (119) (Fig. 2). Conversely, miR-135a inhibits Hedgehog signaling in esophageal CSCs by targeting Smoothened, but its clinical application is limited by delivery challenges (120). These studies underscore the context-dependent role of this pathway, necessitating biomarker-guided therapies.

YAP1/transducer of transcription-TEA domain family transcription factor (TAZ-TEAD) axis

Verteporfin, a YAP1/TAZ-TEAD inhibitor, suppresses GCSC tumorigenicity by disrupting transcriptional activity, leading to reduced spheroid formation and metastasis (121) (Fig. 2). This aligns with findings in colorectal cancer, where tankyrase inhibitors downregulate YAP1-associated c-KIT, impairing CCSC viability (122). However, YAP1 crosstalk with other pathways (such as PI3K/AKT) may necessitate dual targeting to prevent resistance.

Src and BCL signaling

Src kinase inhibition blocks GCSC proliferation and EMT by suppressing STAT3 and AKT phosphorylation (123) (Fig. 2). Similarly, in CRC, MEK inhibitors combined with CSC-targeting agents overcome resistance by depleting ALDH1+ CCSCs (124). These findings emphasize the potential of kinase inhibitors in disrupting CSC survival networks. The pan-BCL-2 inhibitor, navitoclax (ABT-263), synergizes with chemotherapy in gastroesophageal carcinoma, inducing apoptosis in CD44+ CSCs and reducing tumor recurrence (125). However, heterogeneity in BCL-2 family expression across CSC subpopulations may limit efficacy, as observed in colorectal cancer models (126).

Combination therapies

Combining CSC-targeted agents with conventional therapies improves outcomes by addressing bulk tumors and residual CSCs (Fig. 2). For example, salinomycin-loaded carbon nanotubes selectively kill GCSCs while sparing normal cells, enhancing the efficacy of 5-FU (127). In colorectal cancer, polymeric micelles targeting CD44v6 deliver niclosamide, synergizing with oxaliplatin to reduce CCSC-driven metastasis (128). Similarly, mithramycin A represses ABCG2 in CRCSCs, sensitizing them to chemotherapy (129). In addition, aptamer-mediated survivin knockdown in CCSCs enhances 5-FU-induced apoptosis and reduces immune evasion (130). Furthermore, inhibition of cholesterol synthesis in GCSCs attenuates NK cell evasion, suggesting a role for metabolic-immune crosstalk in combinatorial regimens (131).

Challenges and future directions

CSCs serve pivotal roles in the initiation, progression and therapeutic resistance of digestive tract tumors. The heterogeneity and plasticity of these cells pose significant challenges for effective treatment (132). Emerging therapeutic strategies targeting CSC-specific pathways, immune modulation and combination therapies offer promising avenues to overcome resistance and enhance patient outcomes (133). Future research should focus on integrating biomarker-driven approaches and innovative technologies to advance precision medicine in the field of digestive oncology.

Despite promising preclinical data, the clinical translation of CSC-targeted therapies remains challenging. For example, vismodegib, a Hedgehog inhibitor, failed to improve survival in gastric cancer trials due to compensatory YAP1 activation in residual CSCs (134,135). Similarly, Wnt inhibitors such as PRI-724 have shown limited efficacy against colorectal cancer, as CAF-derived IL-6 reactivates β-catenin via STAT3 (136). These failures highlight the need to target CSC-stroma crosstalk. Emerging strategies such as microbiota modulation (including F. nucleatum eradication) and metabolic-immune combinations (including vitamin D + anti-PD-1) may overcome these hurdles by simultaneously disrupting CSC niche support and immune evasion (137,138).

Identifying and targeting CSCs is technically challenging due to their heterogeneity and plasticity. Universal markers such as CD44 and CD133 show variable expression across tumor subtypes, necessitating context-specific validation (139). Furthermore, CSC subpopulations exhibit divergent responses to therapies. For instance, delta-like protein 1 inhibition suppresses Wnt-driven CCSCs but spares LGR5+ subsets (140), underscoring the need for multitargeted strategies. Single-cell sequencing and spatial transcriptomics may elucidate clonal dynamics, guiding personalized therapies. Furthermore, the TME shields CSCs via cytokine loops and stromal support. Blocking proprotein convertases disrupts GCSC-TME crosstalk, reducing invasiveness (141). Similarly, ruthenium-xanthoxylin complexes target HSP90 in CRCSCs, thereby overcoming stroma-mediated resistance (142). Advances in genomic and proteomic technologies have enabled tailored interventions against CSC heterogeneity in digestive tract tumors.

However, preclinical models have inherent limitations. Although animal models provide mechanistic insights, they often fail to recapitulate human tumor complexity and microenvironmental interactions, leading to clinical discrepancies (143). Additionally, tumor heterogeneity and CSC plasticity complicate therapy development, as CSCs can adapt to microenvironmental changes and therapeutic pressures. The TME, which is crucial for CSC support, is difficult to target without affecting normal tissue homeostasis (144). Furthermore, a lack of robust biomarkers for patient stratification and treatment response assessment hinders the development of effective CSC-targeted therapies.

Nanomaterials and CRISPR-based approaches offer precision in future treatment. For example, SPION-driven atranorin induces ferroptosis in GCSCs by modulating Xc-/GPX4 (6,145). Additionally, low-dose vitamin C promotes CCSC differentiation via β-catenin membrane retention, a strategy that is compatible with immune checkpoint inhibitors (146). Although preclinical data are promising, clinical trials remain limited. The active hexose correlated compound/epigallocatechin gallate combination reduces LGR5+ CCSCs in early-phase studies (147); however, its scalability and toxicity require further evaluation. Future research should prioritize biomarkers (such as ALDH1 and CD44) to stratify patients and optimize trial designs.

In addition, the application of artificial intelligence (AI) in predicting the vulnerabilities of CSCs has recently been demonstrated, providing new ideas and methods for the development of targeted therapies against CSCs (148). For example, deep learning models have been employed to analyze large-scale CSC genomic and transcriptomic data, aiming to identify potential therapeutic targets and drug candidates. These models can predict the drug sensitivity and resistance of CSCs, thereby offering a theoretical basis for drug repurposing (149). For example, studies have utilized deep learning algorithms to analyze the gene expression profiles of CSCs from various digestive tract tumors and to predict their responses to different drugs (150,151). The results have shown that some conventional drugs may have potential inhibitory effects on CSCs. Further experimental verification is ongoing to explore the feasibility of these drugs in clinical applications. This integration of AI with CSC research not only increases the efficiency of target discovery but also optimizes therapeutic strategies, offering new avenues for the individualized treatment of digestive tract tumors.

Finally, patient-derived organoids (PDOs) have garnered significant attention as promising tools for personalized therapy screening. PDOs are three-dimensional cellular models cultured from patient tumor tissues (152). PDOs retain the histological structure and genetic characteristics of the original tumor to some extent, making them powerful tools for evaluating drug efficacy and guiding clinical treatment. A previous study has shown that PDOs can effectively mimic the TME and CSC niche of digestive tract tumors (153). By screening various drugs using PDOs, researchers can identify therapeutic regimens that are effective against CSCs and the bulk tumor cells, providing a basis for personalized treatment. For example, researchers successfully established PDO models from patients with gastric or pancreatic ductal adenocarcinoma, and drug screening experiments revealed that certain drugs could specifically targeted CSCs in the PDOs, inhibiting tumor growth and metastasis (154,155). These findings demonstrate the potential of PDOs to help predict therapeutic responses and guide clinical treatment. The application of PDOs in personalized therapy screening offers a new approach for improving the treatment outcomes of digestive tract tumors.

In conclusion, although significant progress has been made in understanding the biology of CSCs and their role in digestive tract tumors, several challenges remain in translating this knowledge into clinical practice. Future research should focus on addressing the limitations of current CSC markers, reconciling discrepancies between preclinical and clinical findings and developing strategies to overcome CSC plasticity and microenvironment-driven resistance. By integrating innovative technologies and biomarker-driven approaches, the field of CSC-targeted therapies can be advanced and precision medicine in the field of digestive oncology can be improved.

Conclusion

CSCs play a pivotal role in the initiation, progression and therapeutic resistance of digestive tract tumors. The heterogeneity and plasticity of CSCs pose significant challenges to effective treatment. Emerging therapeutic strategies targeting CSC-specific pathways, immune modulation and combination therapies offer promising avenues to overcome resistance and enhance patient outcomes. Future research should focus on integrating biomarker-driven approaches and innovative technologies to advance precision medicine in digestive oncology.

Availability of data and materials

Not applicable.

Authors' contributions

XC and XG equally contributed to the study by conducting in-depth literature reviews on CSCs in digestive tract tumors from PubMed, analyzing data related to their biological characteristics and functions in esophageal and gastric cancer as well as drafting relevant sections. CZ focused on CSCs in colorectal cancer, collecting and analyzing data from PubMed and writing the corresponding content. LL supervised the entire project, critically evaluated the data and drafts based on the PubMed literature and refined the manuscript to meet the requirements of the journal. Data authentication is not applicable. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

No funding was received.

References

1 

Haddadin L and Sun X: Stem cells in cancer: From mechanisms to therapeutic strategies. Cells. 14:5382025. View Article : Google Scholar : PubMed/NCBI

2 

El-Tanani M, Rabbani SA, Satyam SM, Rangraze IR, Wali AF, El-Tanani Y and Aljabali AAA: Deciphering the role of cancer stem cells: Drivers of tumor evolution, therapeutic resistance, and precision medicine strategies. Cancers (Basel). 17:3822025. View Article : Google Scholar : PubMed/NCBI

3 

Takebe N, Miele L, Harris PJ, Jeong W, Bando H, Kahn M, Yang SX and Ivy SP: Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: Clinical update. Nat Rev Clin Oncol. 12:445–464. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Gonzalez RS, Raza A, Propst R, Adeyi O, Bateman J, Sopha SC, Shaw J and Auerbach A: Recent advances in digestive tract tumors: Updates from the 5th edition of the world health organization 'blue book'. Arch Pathol Lab Med. 145:607–626. 2021. View Article : Google Scholar

6 

Li K, Dan Z and Nie YQ: Gastric cancer stem cells in gastric carcinogenesis, progression, prevention and treatment. World J Gastroenterol. 20:5420–5426. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Ayob AZ and Ramasamy TS: Cancer stem cells as key drivers of tumour progression. J Biomed Sci. 25:202018. View Article : Google Scholar : PubMed/NCBI

8 

Jahangiri L: Cancer stem cell markers and properties across gastrointestinal cancers. Curr. Tissue Microenviron. Rep. 4:77–89. 2023. View Article : Google Scholar

9 

Sarabia-Sánchez MA, Tinajero-Rodríguez JM, Ortiz-Sánchez E and Alvarado-Ortiz E: Cancer stem cell markers: Symphonic masters of chemoresistance and immune evasion. Life Sci. 355:1230152024. View Article : Google Scholar : PubMed/NCBI

10 

Zhu Y, He S, Wang Z, Xi H, Lu W and Lin X: Predictive and clinicopathological importance of HMGB2 in various carcinomas: A meta and bioinformatic approach. Sci Rep. 15:110032025. View Article : Google Scholar : PubMed/NCBI

11 

Yang Y, Meng WJ and Wang ZQ: Cancer stem cells and the tumor microenvironment in gastric cancer. Front Oncol. 11:8039742022. View Article : Google Scholar : PubMed/NCBI

12 

Zhang H, Steed A, Co M and Chen X: Cancer stem cells, epithelial-mesenchymal transition, ATP and their roles in drug resistance in cancer. Cancer Drug Resist. 4:684–709. 2021.PubMed/NCBI

13 

Sinha S, Hembram KC and Chatterjee S: Targeting signaling pathways in cancer stem cells: A potential approach for developing novel anti-cancer therapeutics. Int Rev Cell Mol Biol. 385:157–209. 2024. View Article : Google Scholar : PubMed/NCBI

14 

Becerril-Rico J, Alvarado-Ortiz E, Toledo-Guzmán ME, Pelayo R and Ortiz-Sánchez E: The cross talk between gastric cancer stem cells and the immune microenvironment: a tumor-promoting factor. Stem Cell Res Ther. 12:4982021. View Article : Google Scholar : PubMed/NCBI

15 

Kapoor-Narula U and Lenka N: Cancer stem cells and tumor heterogeneity: Deciphering the role in tumor progression and metastasis. Cytokine. 157:1559682022. View Article : Google Scholar : PubMed/NCBI

16 

Otaegi-Ugartemendia M, Matheu A and Carrasco-Garcia E: Impact of cancer stem cells on therapy resistance in gastric cancer. Cancers (Basel). 14:14572022. View Article : Google Scholar : PubMed/NCBI

17 

Wang C, Xie J, Guo J, Manning HC, Gore JC and Guo N: Evaluation of CD44 and CD133 as cancer stem cell markers for colorectal cancer. Oncol Rep. 28:1301–1308. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Dhumal SN, Choudhari SK, Patankar S, Ghule SS, Jadhav YB and Masne S: Cancer stem cell markers, CD44 and ALDH1, for assessment of cancer risk in OPMDs and lymph node metastasis in oral squamous cell carcinoma. Head Neck Pathol. 16:453–465. 2022. View Article : Google Scholar :

19 

Hassn Mesrati M, Syafruddin SE, Mohtar MA and Syahir A: CD44: A multifunctional mediator of cancer progression. Biomolecules. 11:18502021. View Article : Google Scholar : PubMed/NCBI

20 

Gopalan V Islam F and Lam AK: Surface markers for the identification of cancer stem cells. Methods Mol Biol. 1692:17–29. 2018. View Article : Google Scholar

21 

Makohon-Moore A and Iacobuzio-Donahue CA: Pancreatic cancer biology and genetics from an evolutionary perspective. Nat Rev Cancer. 16:553–565. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Tian S, Ma R, Liu Y, Chen F, Huang X, Yang Q, Nian W and Fan Z: Clinicopathological significance of cancer stem cell marker CD44/SOX2 in esophageal squamous cell carcinoma (ESCC) patients and construction of a nomogram to predict overall survival. Transl Cancer Res. 13:2971–2984. 2024. View Article : Google Scholar : PubMed/NCBI

23 

Huang G, Yuan C, Zhang C, Yang F, Tan Y, Chen D, Li H and Qian K: Single-cell sequencing reveals the immune microenvironment associated with gastric cancer. Genes Dis. 12:1012182025. View Article : Google Scholar

24 

Xue C, Chu Q, Shi Q, Zeng Y, Lu J and Li L: Wnt signaling pathways in biology and disease: Mechanisms and therapeutic advances. Signal Transduct Target Ther. 10:1062025. View Article : Google Scholar : PubMed/NCBI

25 

Shang T, Jia Z, Li J, Cao H, Xu H, Cong L, Ma D, Wang X and Liu J: Unraveling the triad of hypoxia, cancer cell stemness, and drug resistance. J Hematol Oncol. 18:322025. View Article : Google Scholar : PubMed/NCBI

26 

Wang S, Wang Y, Xun X, Zhang C, Xiang X, Cheng Q, Hu S, Li Z and Zhu J: Hedgehog signaling promotes sorafenib resistance in hepatocellular carcinoma patient-derived organoids. J Exp Clin Cancer Res. 39:222020. View Article : Google Scholar : PubMed/NCBI

27 

Tufail M, Jiang CH and Li N: Wnt signaling in cancer: From biomarkers to targeted therapies and clinical translation. Mol Cancer. 24:1072025. View Article : Google Scholar : PubMed/NCBI

28 

Lanauze CB, Sehgal P, Hayer K, Torres-Diz M, Pippin JA, Grant SFA and Thomas-Tikhonenko A: Colorectal Cancer-Associated Smad4 R361 Hotspot Mutations Boost Wnt/β-Catenin Signaling through Enhanced Smad4-LEF1 Binding. Mol Cancer Res. 19:823–833. 2021. View Article : Google Scholar : PubMed/NCBI

29 

López-Knowles E, Zardawi SJ, McNeil CM, Millar EK, Crea P, Musgrove EA, Sutherland RL and O'Toole SA: Cytoplasmic localization of beta-catenin is a marker of poor outcome in breast cancer patients. Cancer Epidemiol Biomarkers Prev. 19:301–309. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Aine M, Nacer DF, Arbajian E, Veerla S, Karlsson A, Häkkinen J, Johansson HJ, Rosengren F, Vallon-Christersson J, Borg A and Staaf J: The DNA methylation landscape of primary triple-negative breast cancer. Nat Commun. 16:30412025. View Article : Google Scholar : PubMed/NCBI

31 

Zhang Z and Zhang Y: Transcriptional regulation of cancer stem cell: regulatory factors elucidation and cancer treatment strategies. J Exp Clin Cancer Res. 43:992024. View Article : Google Scholar : PubMed/NCBI

32 

Darwiche N: Epigenetic mechanisms and the hallmarks of cancer: An intimate affair. Am J Cancer Res. 10:1954–1978. 2020.PubMed/NCBI

33 

Liu B, Peng Z, Zhang H, Zhang N, Liu Z, Xia Z, Huang S, Luo P and Cheng Q: Regulation of cellular senescence in tumor progression and therapeutic targeting: mechanisms and pathways. Mol Cancer. 24:1062025. View Article : Google Scholar : PubMed/NCBI

34 

Quail DF and Joyce JA: Microenvironmental regulation of tumor progression and metastasis. Nat Med. 19:1423–1437. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Jia H, Chen X, Zhang L and Chen M: Cancer associated fibroblasts in cancer development and therapy. J Hematol Oncol. 18:362025. View Article : Google Scholar : PubMed/NCBI

36 

Shi Z, Hu C, Li Q and Sun C: Cancer-associated fibroblasts as the 'Architect' of the lung cancer immune microenvironment: Multidimensional roles and synergistic regulation with radiotherapy. Int J Mol Sci. 26:32342025. View Article : Google Scholar

37 

Yu S, Wang S, Wang X and Xu X: The axis of tumor-associated macrophages, extracellular matrix proteins, and cancer-associated fibroblasts in oncogenesis. Cancer Cell Int. 24:3352024. View Article : Google Scholar : PubMed/NCBI

38 

Huang B, Lang X and Li X: The role of IL-6/JAK2/STAT3 signaling pathway in cancers. Front Oncol. 12:10231772022. View Article : Google Scholar :

39 

Li YR, Fang Y, Lyu Z, Zhu Y and Yang L: Exploring the dynamic interplay between cancer stem cells and the tumor microenvironment: Implications for novel therapeutic strategies. J Transl Med. 21:6862023. View Article : Google Scholar : PubMed/NCBI

40 

Rabinovich I, Sebastião APM, Lima RS, Urban CA, Junior ES, Anselmi KF, Elifio-Esposito S, De Noronha L and Moreno-Amaral AN: Cancer stem cell markers ALDH1 and CD44+/CD24− phenotype and their prognosis impact in invasive ductal carcinoma. Eur J Histochem. 62:29432018.

41 

Wang D, Li Y, Ge H, Ghadban T, Reeh M and Güngör C: The extracellular matrix: A key accomplice of cancer stem cell migration, metastasis formation, and drug resistance in PDAC. Cancers (Basel). 14:39982022. View Article : Google Scholar : PubMed/NCBI

42 

Tie Y, Tang F, Wei YQ and Wei XW: Immunosuppressive cells in cancer: Mechanisms and potential therapeutic targets. J Hematol Oncol. 15:612022. View Article : Google Scholar : PubMed/NCBI

43 

Yin B, Cai Y, Chen L, Li Z and Li X: Immunosuppressive MDSC and Treg signatures predict prognosis and therapeutic response in glioma. Int Immunopharmacol. 141:1129222024. View Article : Google Scholar : PubMed/NCBI

44 

Lin Y, Song Y, Zhang Y, Li X, Kan L and Han S: New insights on anti-tumor immunity of CD8+ T cells: Cancer stem cells, tumor immune microenvironment and immunotherapy. J Transl Med. 23:3412025. View Article : Google Scholar :

45 

Galassi C, Musella M, Manduca N, Maccafeo E and Sistigu A: The immune privilege of cancer stem cells: A key to understanding tumor immune escape and therapy failure. Cells. 10:23612021. View Article : Google Scholar : PubMed/NCBI

46 

Luo H, Hu B, Gu XR, Chen J, Fan XQ, Zhang W, Wang RT, He XD, Guo W, Dai N, et al: The miR-23a/27a/24 − 2 cluster drives immune evasion and resistance to PD-1/PD-L1 blockade in non-small cell lung cancer. Mol Cancer. 23:2852024. View Article : Google Scholar

47 

Geng S, Zhu L, Wang Y, Liu Q, Yu C, Shi S and Yu S: Co-Colorectal cancer stem cells employ the FADS1/DDA axis to evade NK cell-mediated immunosuppression after co-cultured with NK cells under hypoxia. Int Immunopharmacol. 143(Pt 3): 1135352024. View Article : Google Scholar : PubMed/NCBI

48 

Liu S, Zhao H, Hu Y, Yan C, Mi Y, Li X, Tao D and Qin J: Lactate promotes metastasis of normoxic colorectal cancer stem cells through PGC-1α-mediated oxidative phosphorylation. Cell Death Dis. 13:6512022. View Article : Google Scholar

49 

Guo S, Zhao W, Zhang W, Li S, Teng G and Liu L: Vitamin D promotes ferroptosis in colorectal cancer stem cells via SLC7A11 downregulation. Oxid Med Cell Longev. 2023:47721342023. View Article : Google Scholar : PubMed/NCBI

50 

Zhang Q, Tian H, Ge K and Wang F, Gao P, Chen A, Wang L, Zhao Y, Lian C and Wang F: PGD2/PTGDR2 signaling pathway affects the self-renewal capacity of gastric cancer stem cells by regulating ATG4B ubiquitination. Front Oncol. 14:14960502024. View Article : Google Scholar

51 

Chen Y, Li D, Wang D, Liu X, Yin N, Song Y, Lu SH, Ju Z and Zhan Q: Quiescence and attenuated DNA damage response promote survival of esophageal cancer stem cells. J Cell Biochem. 113:3643–3652. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Zhao Y, Bao Q, Schwarz B, Zhao L, Mysliwietz J, Ellwart J, Renner A, Hirner H, Niess H, Camaj P, et al: Stem cell-like side populations in esophageal cancer: A source of chemotherapy resistance and metastases. Stem Cells Dev. 23:180–192. 2014. View Article : Google Scholar

53 

Song S, Ajani JA, Honjo S, Maru DM, Chen Q, Scott AW, Heallen TR, Xiao L, Hofstetter WL, Weston B, et al: Hippo coactivator YAP1 upregulates SOX9 and endows esophageal cancer cells with stem-like properties. Cancer Res. 74:4170–4182. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Xu DD, Zhou PJ, Wang Y, Zhang L, Fu WY, Ruan BB, Xu HP, Hu CZ, Tian L, Qin JH, et al: Reciprocal activation between STAT3 and miR-181b regulates the proliferation of esophageal cancer stem-like cells via the CYLD pathway. Mol Cancer. 15:402016. View Article : Google Scholar : PubMed/NCBI

55 

Liu CC, Chou KT, Hsu JW, Lin JH, Hsu TW, Yen DH, Hung SC and Hsu HS: High metabolic rate and stem cell characteristics of esophageal cancer stem-like cells depend on the Hsp27-AKT-HK2 pathway. Int J Cancer. 145:2144–2156. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Kai JD, Cheng LH, Li BF, Kang K, Xiong F, Fu JC and Wang S: MYH9 is a novel cancer stem cell marker and prognostic indicator in esophageal cancer that promotes oncogenesis through the PI3K/AKT/mTOR axis. Cell Biol Int. 46:2085–2094. 2022. View Article : Google Scholar : PubMed/NCBI

57 

Wang X, Cao Y, Zhang S, Chen Z, Fan L, Shen X, Zhou S and Chen D: Stem cell autocrine CXCL12/CXCR4 stimulates invasion and metastasis of esophageal cancer. Oncotarget. 8:36149–36160. 2017. View Article : Google Scholar : PubMed/NCBI

58 

Yue D, Zhang Z, Li J, Chen X, Ping Y, Liu S, Shi X, Li L, Wang L, Huang L, et al: Transforming growth factor-beta1 promotes the migration and invasion of sphere-forming stem-like cell subpopulations in esophageal cancer. Exp Cell Res. 336:141–149. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Wei JR, Zhang B, Zhang Y, Chen WM, Zhang XP, Zeng TT, Li Y, Zhu YH, Guan XY and Li L: QSOX1 facilitates dormant esophageal cancer stem cells to evade immune elimination via PD-L1 upregulation and CD8 T cell exclusion. Proc Natl Acad Sci USA. 121:e24075061212024. View Article : Google Scholar : PubMed/NCBI

60 

Guo M, Lian J, Liu Y, Dong B, He Q, Zhao Q, Zhang H, Qi Y, Zhang Y and Huang L: Loss of miR-637 promotes cancer cell stemness via WASH/IL-8 pathway and serves as a novel prognostic marker in esophageal squamous cell carcinoma. Biomark Res. 10:772022. View Article : Google Scholar : PubMed/NCBI

61 

Xun J, Ma Y, Wang B, Jiang X, Liu B, Gao R, Zhai Q, Cheng R, Wu X, Wu Y and Zhang Q: RGS1 targeted by miR-191-3p inhibited the stemness properties of esophageal cancer cells by suppressing CXCR4/PI3K/AKT signaling. Acta Histochem. 126:1521902024. View Article : Google Scholar : PubMed/NCBI

62 

Yu X, Teng Y, Jiang X, Yuan H and Jiang W: Genome-Wide DNA methylation pattern of cancer stem cells in esophageal cancer. Technol Cancer Res Treat. 19:15330338209837932020. View Article : Google Scholar : PubMed/NCBI

63 

Gupta P, Rizvi SZ, Lal N, Gupta V, Srivastav AN and Musa O: Expression of CD44 and CD133 stem cell markers in squamous cell carcinoma of esophagus. Indian J Pathol Microbiol. 64:472–478. 2021. View Article : Google Scholar : PubMed/NCBI

64 

Xu DD, Chen SH, Zhou PJ, Wang Y, Zhao ZD, Wang X, Huang HQ, Xue X, Liu QY, Wang YF and Zhang R: Suppression of Esophageal Cancer Stem-like Cells by SNX-2112 Is Enhanced by STAT3 Silencing. Front Pharmacol. 11:5323952020. View Article : Google Scholar :

65 

Liu CC, Li HH, Lin JH, Chiang MC, Hsu TW, Li AF, Yen DH, Hsu HS and Hung SC: Esophageal Cancer Stem-like Cells Resist Ferroptosis-Induced Cell Death by Active Hsp27-GPX4 Pathway. Biomolecules. 12:482021. View Article : Google Scholar

66 

Mao J, Fan S, Ma W, Fan P, Wang B, Zhang J, Wang H, Tang B, Zhang Q, Yu X, et al: Roles of Wnt/β-catenin signaling in the gastric cancer stem cells proliferation and salinomycin treatment. Cell Death Dis. 5:e10392014. View Article : Google Scholar

67 

Xu XF, Gao F, Wang JJ, Long C, Chen X, Tao L, Yang L, Ding L and Ji Y: BMX-ARHGAP fusion protein maintains the tumorigenicity of gastric cancer stem cells by activating the JAK/STAT3 signaling pathway. Cancer Cell Int. 19:1332019. View Article : Google Scholar : PubMed/NCBI

68 

Wu Q, Yang Z, Wang F, Hu S, Yang L, Shi Y and Fan D: MiR-19b/20a/92a regulates the self-renewal and proliferation of gastric cancer stem cells. J Cell Sci. 126(Pt 18): 4220–4229. 2013.PubMed/NCBI

69 

Han ME, Baek SJ, Kim SY, Kang CD and Oh SO: ATOH1 can regulate the tumorigenicity of gastric cancer cells by inducing the differentiation of cancer stem cells. PLoS One. 10:e01260852015. View Article : Google Scholar : PubMed/NCBI

70 

Shen C, Wang J, Xu Z, Zhang L, Gu W and Zhou X: ONECUT2 which is targeted by hsa-miR-15a-5p enhances stemness maintenance of gastric cancer stem cells. Exp Biol Med (Maywood). 246:2645–2659. 2021. View Article : Google Scholar : PubMed/NCBI

71 

Li LQ, Pan D, Zhang SWY, Xie D, Zheng XL and Chen H: Autophagy regulates chemoresistance of gastric cancer stem cells via the Notch signaling pathway. Eur Rev Med Pharmacol Sci. 22:3402–3407. 2018.PubMed/NCBI

72 

Xin L, Li SH, Liu C, Zeng F, Cao JQ, Zhou LQ, Zhou Q and Yuan YW: Methionine represses the autophagy of gastric cancer stem cells via promoting the methylation and phosphorylation of RAB37. Cell Cycle. 19:2644–2652. 2020. View Article : Google Scholar : PubMed/NCBI

73 

Togano S, Yashiro M, Masuda G, Sugimoto A, Miki Y, Yamamoto Y, Sera T, Kushiyama S, Nishimura S, Kuroda K, et al: Gastric cancer stem cells survive in stress environments via their autophagy system. Sci Rep. 11:206642021. View Article : Google Scholar : PubMed/NCBI

74 

Yang SW, Zhang ZG, Hao YX, Zhao YL, Qian F, Shi Y, Li PA, Liu CY and Yu PW: HIF-1α induces the epithelial-mesenchymal transition in gastric cancer stem cells through the Snail pathway. Oncotarget. 8:9535–9545. 2017. View Article : Google Scholar : PubMed/NCBI

75 

Sun LF, Yang K, Wang YG, Liu YX, Hou PX, Lu ZH, Chen XL, Zhang WH, Zhou ZG, Mo XM and Hu JK: The Role of HER2 in self-renewal, invasion, and tumorigenicity of gastric cancer stem cells. Front Oncol. 10:16082020. View Article : Google Scholar : PubMed/NCBI

76 

Seeneevassen L, Giraud J, Molina-Castro S, Sifré E, Tiffon C, Beauvoit C, Staedel C, Mégraud F, Lehours P, Martin OCB, et al: Leukaemia inhibitory factor (LIF) inhibits cancer stem cells tumorigenic properties through hippo kinases activation in gastric cancer. Cancers (Basel). 12:20112020. View Article : Google Scholar : PubMed/NCBI

77 

Zhang Q and Wang F, Huang Y, Gao P, Wang N, Tian H, Chen A, Li Y and Wang F: PGD2/PTGDR2 signal affects the viability, invasion, apoptosis, and stemness of gastric cancer stem cells and prevents the progression of gastric cancer. Comb Chem High Throughput Screen. 27:933–946. 2024. View Article : Google Scholar

78 

Wang X, Zhang F, Yang J, Huang X, Chao X, Ayidu A and Abudureyimu A: The chemotherapeutic effect of docetaxel, cisplatin and fluorouracil regimen on gastric cancer stem cells. J Nanosci Nanotechnol. 17:983–999. 2017. View Article : Google Scholar : PubMed/NCBI

79 

Zhang H, Wang M, He Y, Deng T, Liu R, Wang W, Zhu K, Bai M, Ning T, Yang H, et al: Chemotoxicity-induced exosomal lncFERO regulates ferroptosis and stemness in gastric cancer stem cells. Cell Death Dis. 12:11162021. View Article : Google Scholar : PubMed/NCBI

80 

Mao X, Wang L, Chen Z, Huang H, Chen J, Su J, Li Z, Shen G, Ren Y, Li Z, et al: SCD1 promotes the stemness of gastric cancer stem cells by inhibiting ferroptosis through the SQLE/cholesterol/mTOR signalling pathway. Int J Biol Macromol. 275(Pt 2): 1336982024. View Article : Google Scholar : PubMed/NCBI

81 

Ni T, Chu Z, Tao L, Zhao Y, Lv M, Zhu M, Luo Y, Sunagawa M, Wang H and Liu Y: Celastrus orbiculatus extract suppresses gastric cancer stem cells through the TGF-β/Smad signaling pathway. J Nat Med. 78:100–113. 2024. View Article : Google Scholar

82 

Chen B, Zhang D, Kuai J, Cheng M, Fang X and Li G: Upregulation of miR-199a/b contributes to cisplatin resistance via Wnt/β-catenin-ABCG2 signaling pathway in ALDHA1(+) colorectal cancer stem cells. Tumour Biol. 39:10104283177151552017. View Article : Google Scholar

83 

Li J, Yu B, Deng P, Cheng Y, Yu Y, Kevork K, Ramadoss S, Ding X, Li X and Wang CY: KDM3 epigenetically controls tumorigenic potentials of human colorectal cancer stem cells through Wnt/β-catenin signalling. Nat Commun. 8:151462017. View Article : Google Scholar

84 

Hua F, Shang S, Yang YM, Zhang HZ, Xu TL, Yu JJ, Zhou DD, Cui B, Li K, Lv XX, et al: TRIB3 interacts With β-Catenin and TCF4 to increase stem cell features of colorectal cancer stem cells and tumorigenesis. Gastroenterology. 156:708–721.e15. 2019. View Article : Google Scholar

85 

Yu W, Ma Y, Shankar S and Srivastava RK: SATB2/β-catenin/TCF-LEF pathway induces cellular transformation by generating cancer stem cells in colorectal cancer. Sci Rep. 7:109392017. View Article : Google Scholar

86 

Zhu Y, Huang S, Chen S, Chen J, Wang Z, Wang Y and Zheng H: SOX2 promotes chemoresistance, cancer stem cells properties, and epithelial-mesenchymal transition by β-catenin and Beclin1/autophagy signaling in colorectal cancer. Cell Death Dis. 12:4492021. View Article : Google Scholar

87 

Izumi D, Ishimoto T, Miyake K, Eto T, Arima K, Kiyozumi Y, Uchihara T, Kurashige J, Iwatsuki M, Baba Y, et al: Colorectal cancer stem cells acquire chemoresistance through the upregulation of F-Box/WD repeat-containing protein 7 and the consequent degradation of c-Myc. Stem Cells. 35:2027–2036. 2017. View Article : Google Scholar : PubMed/NCBI

88 

Honma S, Hisamori S, Nishiuchi A, Itatani Y, Obama K, Shimono Y and Sakai Y: F-Box/WD repeat domain-containing 7 induces chemotherapy resistance in colorectal cancer stem cells. Cancers (Basel). 11:6352019. View Article : Google Scholar : PubMed/NCBI

89 

Mukohyama J, Isobe T, Hu Q, Hayashi T, Watanabe T, Maeda M, Yanagi H, Qian X, Yamashita K, Minami H, et al: miR-221 Targets QKI to Enhance the tumorigenic capacity of human colorectal cancer stem cells. Cancer Res. 79:5151–5158. 2019. View Article : Google Scholar : PubMed/NCBI

90 

Liu L, Zhang Z, Zhou L, Hu L, Yin C, Qing D, Huang S, Cai X and Chen Y: Cancer associated fibroblasts-derived exosomes contribute to radioresistance through promoting colorectal cancer stem cells phenotype. Exp Cell Res. 391:1119562020. View Article : Google Scholar : PubMed/NCBI

91 

Montalbán-Hernández K, Cantero-Cid R, Casalvilla-Dueñas JC, Avendaño-Ortiz J, Marín E, Lozano-Rodríguez R, Ter rón-A rcos V, Vica r io-Bravo M, Ma rcano C, Saavedra-Ambrosy J, et al: Colorectal cancer stem cells fuse with monocytes to form tumour hybrid cells with the ability to migrate and evade the immune system. Cancers (Basel). 14:34452022. View Article : Google Scholar : PubMed/NCBI

92 

Cavallucci V, Palucci I, Fidaleo M, Mercuri A, Masi L, Emoli V, Bianchetti G, Fiori ME, Bachrach G, Scaldaferri F, et al: Proinflammatory and cancer-promoting pathobiont fusobacterium nucleatum directly targets colorectal cancer stem cells. Biomolecules. 12:12562022. View Article : Google Scholar : PubMed/NCBI

93 

Tamura S, Isobe T, Ariyama H, Nakano M, Kikushige Y, Takaishi S, Kusaba H, Takenaka K, Ueki T, Nakamura M, et al: E-cadherin regulates proliferation of colorectal cancer stem cells through NANOG. Oncol Rep. 40:693–703. 2018.PubMed/NCBI

94 

Zou W, Zhang Y, Bai G, Zhuang J, Wei L, Wang Z, Sun M and Wang J: siRNA-induced CD44 knockdown suppresses the proliferation and invasion of colorectal cancer stem cells through inhibiting epithelial-mesenchymal transition. J Cell Mol Med. 26:1969–1978. 2022. View Article : Google Scholar : PubMed/NCBI

95 

Vishnubalaji R, Manikandan M, Fahad M, Hamam R, Alfayez M, Kassem M, Aldahmash A and Alajez NM: Molecular profiling of ALDH1(+) colorectal cancer stem cells reveals preferential activation of MAPK, FAK, and oxidative stress pro-survival signalling pathways. Oncotarget. 9:13551–13564. 2018. View Article : Google Scholar : PubMed/NCBI

96 

Agawa K, Yamashita K, Nakagawa A, Yamada K, Watanabe A, Mukohyama J, Saito M, Fujita M, Takiguchi G, Urakawa N, et al: Simple cancer stem cell markers predict neoadjuvant chemotherapy resistance of esophageal squamous cell carcinoma. Anticancer Res. 41:4117–4126. 2021. View Article : Google Scholar : PubMed/NCBI

97 

Lin CH, Li HY, Liu YP, Kuo PF, Wang WC, Lin FC, Chang WL, Sheu BS, Wang YC, Hung WC, et al: High-CLDN4 ESCC cells harbor stem-like properties and indicate for poor concurrent chemoradiation therapy response in esophageal squamous cell carcinoma. Ther Adv Med Oncol. 11:17588359198753242019. View Article : Google Scholar : PubMed/NCBI

98 

Trevellin E, Pirozzolo G, Fassan M and Vettor R: Prognostic value of stem cell markers in esophageal and esophagogastric junction cancer: A meta-analysis. J Cancer. 11:4240–4249. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Nishikawa S, Konno M, Hamabe A, Hasegawa S, Kano Y, Ohta K, Fukusumi T, Sakai D, Kudo T, Haraguchi N, et al: Aldehyde dehydrogenase high gastric cancer stem cells are resistant to chemotherapy. Int J Oncol. 42:1437–1442. 2013. View Article : Google Scholar : PubMed/NCBI

100 

Gong DY, Chen X, Yang TL, Wang Y, Guo Y, Zeng JH and Chen SZ: Upregulation of ECT2 is associated with transcriptional program of cancer stem cells and predicts poor clinical outcome in gastric cancer. Oncol Lett. 20:542020.PubMed/NCBI

101 

Becerril-Rico J, Grandvallet-Contreras J, Ruíz-León MP, Dorantes-Cano S, Ramírez-Vidal L, Tinajero-Rodríguez JM and Ortiz-Sánchez E: Circulating gastric cancer stem cells as blood screening and prognosis factor in gastric cancer. Stem Cells Int. 2024:99991552024. View Article : Google Scholar : PubMed/NCBI

102 

Catalano V, Dentice M, Ambrosio R, Luongo C, Carollo R, Benfante A, Todaro M, Stassi G and Salvatore D: Activated thyroid hormone promotes differentiation and chemotherapeutic sensitization of colorectal cancer stem cells by regulating Wnt and BMP4 signaling. Cancer Res. 76:1237–1244. 2016. View Article : Google Scholar

103 

Prieur A, Cappellini M, Habif G, Lefranc MP, Mazard T, Morency E, Pascussi JM, Flacelière M, Cahuzac N, Vire B, et al: Targeting the Wnt pathway and cancer stem cells with anti-progastrin humanized antibodies as a potential treatment for K-RAS-mutated colorectal cancer. Clin Cancer Res. 23:5267–5280. 2017. View Article : Google Scholar : PubMed/NCBI

104 

Chen Q, Song S, Wei S, Liu B, Honjo S, Scott A, Jin J, Ma L, Zhu H, Skinner HD, et al: ABT-263 induces apoptosis and synergizes with chemotherapy by targeting stemness pathways in esophageal cancer. Oncotarget. 6:25883–25896. 2015. View Article : Google Scholar : PubMed/NCBI

105 

Narusaka T, Ohara T, Noma K, Nishiwaki N, Katsura Y, Kato T, Sato H, Tomono Y, Kikuchi S, Tazawa H, et al: Nanog is a promising chemoresistant stemness marker and therapeutic target by iron chelators for esophageal cancer. Int J Cancer. 149:347–357. 2021. View Article : Google Scholar : PubMed/NCBI

106 

Xu ZY, Tang JN, Xie HX, Du YA, Huang L, Yu PF and Cheng XD: 5-Fluorouracil chemotherapy of gastric cancer generates residual cells with properties of cancer stem cells. Int J Biol Sci. 11:284–294. 2015. View Article : Google Scholar : PubMed/NCBI

107 

Liu C, Wang JL, Wu DZ, Yuan YW and Xin L: Methionine restriction enhances the chemotherapeutic sensitivity of colorectal cancer stem cells by miR-320d/c-Myc axis. Mol Cell Biochem. 477:2001–2013. 2022. View Article : Google Scholar : PubMed/NCBI

108 

Yu M, Fei B and Chu S: Targeting HNRNPA2B1 to overcome chemotherapy resistance in gastric cancer stem cells: Mechanisms and therapeutic potential. J Biol Chem. 301:1082342025. View Article : Google Scholar : PubMed/NCBI

109 

Kim MJ, Koo JE, Han GY, Kim B, Lee YS, Ahn C and Kim CW: Dual-Blocking of PI3K and mTOR improves chemotherapeutic effects on SW620 human colorectal cancer stem cells by inducing differentiation. J Korean Med Sci. 31:360–370. 2016. View Article : Google Scholar : PubMed/NCBI

110 

Tsunekuni K, Konno M, Haraguchi N, Koseki J, Asai A, Matsuoka K, Kobunai T, Takechi T, Doki Y, Mori M and Ishii H: CD44/CD133-positive colorectal cancer stem cells are sensitive to trifluridine exposure. Sci Rep. 9:148612019. View Article : Google Scholar : PubMed/NCBI

111 

Khoei SG, Sadeghi H and Dermani FK: Targeting the SPHK1/HIF1 PATHWAY TO INHIBIT colorectal cancer stem cells niche. J Gastrointest Cancer. 51:716–717. 2020. View Article : Google Scholar

112 

Rio-Vilariño A, Cenigaonandia-Campillo A, García-Bautista A, Mateos-Gómez PA, Schlaepfer MI, Del Puerto-Nevado L, Aguilera O, García-García L, Galeano C, de Miguel I, et al: Inhibition of the AURKA/YAP1 axis is a promising therapeutic option for overcoming cetuximab resistance in colorectal cancer stem cells. Br J Cancer. 130:1402–1413. 2024. View Article : Google Scholar : PubMed/NCBI

113 

Akrami H, Moradi B, Borzabadi Farahani D and Mehdizadeh K: Ibuprofen reduces cell proliferation through inhibiting Wnt/β catenin signaling pathway in gastric cancer stem cells. Cell Biol Int. 42:949–958. 2018. View Article : Google Scholar : PubMed/NCBI

114 

Chen Y, Li Y, Wang XQ, Meng Y, Zhang Q, Zhu JY, Chen JQ, Cao WS, Wang XQ, Xie CF, et al: Phenethyl isothiocyanate inhibits colorectal cancer stem cells by suppressing Wnt/β-catenin pathway. Phytother Res. 32:2447–2455. 2018. View Article : Google Scholar : PubMed/NCBI

115 

Chen Y, Wang XQ, Zhang Q, Zhu JY, Li Y, Xie CF, Li XT, Wu JS, Geng SS, Zhong CY and Han HY: (-)-epigallocatechin-3-gallate inhibits colorectal cancer stem cells by suppressing Wnt/β-catenin pathway. Nutrients. 9:5722017. View Article : Google Scholar

116 

Qi J, Cui D, Wu QN, Zhao Q, Chen ZH, Li L, Birchmeier W, Yu Y and Tao R: Targeting Wnt/β-catenin signaling by TET1/FOXO4 inhibits metastatic spreading and self-renewal of cancer stem cells in gastric cancer. Cancers (Basel). 14:32322022. View Article : Google Scholar

117 

Wen Z, Feng S, Wei L, Wang Z, Hong D and Wang Q: Evodiamine, a novel inhibitor of the Wnt pathway, inhibits the self-renewal of gastric cancer stem cells. Int J Mol Med. 36:1657–1663. 2015. View Article : Google Scholar : PubMed/NCBI

118 

Sun J, Zhang S and Wang M, Cheng H, Wang Y, He S, Zuo Q, Wang N, Li Q and Wang M: Cinobufacini enhances the therapeutic response of 5-Fluorouracil against gastric cancer by targeting cancer stem cells via AKT/GSK-3β/β-catenin signaling axis. Transl Oncol. 47:1020542024. View Article : Google Scholar

119 

Cao W, Li Y, Sun H, Yang C, Zhu J, Xie C, Li X, Wu J, Geng S, Wang L, et al: Apatinib suppresses gastric cancer stem cells properties by inhibiting the sonic hedgehog pathway. Front Cell Dev Biol. 9:6798062021. View Article : Google Scholar : PubMed/NCBI

120 

Yang C, Zheng X, Ye K, Sun Y, Lu Y, Fan Q and Ge H: miR-135a inhibits the invasion and migration of esophageal cancer stem cells through the hedgehog signaling pathway by targeting Smo. Mol Ther Nucleic Acids. 19:841–852. 2020. View Article : Google Scholar : PubMed/NCBI

121 

Giraud J, Molina-Castro S, Seeneevassen L, Sifré E, Izotte J, Tiffon C, Staedel C, Boeuf H, Fernandez S, Barthelemy P, et al: Verteporfin targeting YAP1/TAZ-TEAD transcriptional activity inhibits the tumorigenic properties of gastric cancer stem cells. Int J Cancer. 146:2255–2267. 2020. View Article : Google Scholar

122 

Jang MK, Mashima T and Seimiya H: Tankyrase inhibitors target colorectal cancer stem cells via AXIN-dependent downregulation of c-KIT tyrosine kinase. Mol Cancer Ther. 19:765–776. 2020. View Article : Google Scholar : PubMed/NCBI

123 

Hu CT, Lin CF, Shih HM, You RI, Wu WS and Chen TC: Blockade of Src signaling prevented stemness gene expression and proliferation of patient-derived gastric cancer stem cells. Tzu Chi Med J. 37:65–71. 2024. View Article : Google Scholar

124 

Lamichhane A, Shahi Thakuri P, Singh S, Rafsanjani Nejad P, Heiss J, Luker GD and Tavana H: Therapeutic targeting of cancer stem cells prevents resistance of colorectal cancer cells to MEK inhibition. ACS Pharmacol Transl Sci. 5:724–734. 2022. View Article : Google Scholar : PubMed/NCBI

125 

Song S, Chen Q, Li Y, Lei G, Scott A, Huo L, Li CY, Estrella JS, Correa A, Pizzi MP, et al: Targeting cancer stem cells with a pan-BCL-2 inhibitor in preclinical and clinical settings in patients with gastroesophageal carcinoma. Gut. 70:2238–2248. 2021. View Article : Google Scholar : PubMed/NCBI

126 

Park SR, Kim SR, Hong IS and Lee HY: A novel therapeutic approach for colorectal cancer stem cells: Blocking the PI3K/Akt signaling axis with caffeic acid. Front Cell Dev Biol. 8:5859872020. View Article : Google Scholar

127 

Yao HJ, Zhang YG, Sun L and Liu Y: The effect of hyaluronic acid functionalized carbon nanotubes loaded with salinomycin on gastric cancer stem cells. Biomaterials. 35:9208–9223. 2014. View Article : Google Scholar : PubMed/NCBI

128 

Andrade F, Rafael D, Vilar-Hernández M, Montero S, Martínez-Trucharte F, Seras-Franzoso J, Díaz-Riascos ZV, Boullosa A, García-Aranda N, Cámara-Sánchez P, et al: Polymeric micelles targeted against CD44v6 receptor increase niclosamide efficacy against colorectal cancer stem cells and reduce circulating tumor cells in vivo. J Control Release. 331:198–212. 2021. View Article : Google Scholar : PubMed/NCBI

129 

Quarni W, Dutta R, Green R, Katiri S, Patel B, Mohapatra SS and Mohapatra S: Mithramycin a inhibits colorectal cancer growth by targeting cancer stem cells. Sci Rep. 9:152022019. View Article : Google Scholar : PubMed/NCBI

130 

AlShamaileh H, Wang T, Xiang D, Yin W, Tran PH, Barrero RA, Zhang PZ, Li Y, Kong L, Liu K, et al: Aptamer-mediated survivin RNAi enables 5-fluorouracil to eliminate colorectal cancer stem cells. Sci Rep. 7:58982017. View Article : Google Scholar : PubMed/NCBI

131 

Zhu L and Wang H: Cholesterol-regulated cellular stiffness may enhance evasion of NK cell-mediated cytotoxicity in gastric cancer stem cells. FEBS Open Bio. 14:855–866. 2024. View Article : Google Scholar : PubMed/NCBI

132 

Chen W, Dong J, Haiech J, Kilhoffer MC and Zeniou M: Cancer stem cell quiescence and plasticity as major challenges in cancer therapy. Stem Cells Int. 2016:17409362016. View Article : Google Scholar : PubMed/NCBI

133 

Huang T, Song X, Xu D, Tiek D, Goenka A, Wu B, Sastry N, Hu B and Cheng SY: Stem cell programs in cancer initiation, progression, and therapy resistance. Theranostics. 10:8721–8743. 2020. View Article : Google Scholar : PubMed/NCBI

134 

Kim R, Ji JH, Kim JH, Hong JY, Lim HY, Kang WK, Lee J and Kim ST: Safety and anti-tumor effects of vismodegib in patients with refractory advanced gastric cancer: A single-arm, phase-II trial. J Cancer. 13:1097–1102. 2022. View Article : Google Scholar : PubMed/NCBI

135 

Rao X, Zhang C, Luo H, Zhang J, Zhuang Z, Liang Z and Wu X: Targeting gastric cancer stem cells to enhance treatment response. Cells. 11:28282022. View Article : Google Scholar : PubMed/NCBI

136 

Zhong B, Cheng B, Huang X, Xiao Q, Niu Z, Chen YF, Yu Q, Wang W and Wu XJ: Colorectal cancer-associated fibroblasts promote metastasis by up-regulating LRG1 through stromal IL-6/STAT3 signaling. Cell Death Dis. 13:162021. View Article : Google Scholar : PubMed/NCBI

137 

Luo J, Chen H, Ma F, Xiao C, Sun B, Liu Y, Tang H, Yang Y, Liu W and Luo Z: Vitamin D metabolism pathway polymorphisms are associated with efficacy and safety in patients under anti-PD-1 inhibitor therapy. Front Immunol. 13:9374762022. View Article : Google Scholar : PubMed/NCBI

138 

Krishnamurthy N and Kurzrock R: Targeting the Wnt/beta-catenin pathway in cancer: Update on effectors and inhibitors. Cancer Treat Rev. 62:50–60. 2018. View Article : Google Scholar

139 

Mohan A, Raj Rajan R, Mohan G, Kollenchery Puthenveettil P and Maliekal TT: Markers and reporters to reveal the hierarchy in heterogeneous cancer stem cells. Front Cell Dev Biol. 9:6688512021. View Article : Google Scholar : PubMed/NCBI

140 

Li Y, Lu Y, Wu M, Wang H, Gong Y and Gu Y: Neogambogic acid suppresses characteristics and growth of colorectal cancer stem cells by inhibition of DLK1 and Wnt/β-catenin pathway. Eur J Pharmacol. 929:1751122022. View Article : Google Scholar

141 

Zaafour A, Seeneevassen L, Nguyen TL, Genevois C, Nicolas N, Sifré E, Giese A, Porcheron C, Descarpentrie J, Dubus P, et al: Inhibition of proprotein convertases activity results in repressed stemness and invasiveness of cancer stem cells in gastric cancer. Gastric Cancer. 27:292–307. 2024. View Article : Google Scholar : PubMed/NCBI

142 

Santos LS, Silva VR, de Castro MVL, Dias RB, Valverde LF, Rocha CAG, Soares MBP, Quadros CA, Dos Santos ER, Oliveira RMM, et al: New ruthenium-xanthoxylin complex eliminates colorectal cancer stem cells by targeting the heat shock protein 90 chaperone. Cell Death Dis. 14:8322023. View Article : Google Scholar : PubMed/NCBI

143 

Soufizadeh P, Mansouri V and Ahmadbeigi N: A review of animal models utilized in preclinical studies of approved gene therapy products: trends and insights. Lab Anim Res. 40:172024. View Article : Google Scholar : PubMed/NCBI

144 

Johnson JI, Decker S, Zaharevitz D, Rubinstein LV, Venditti JM, Schepartz S, Kalyandrug S, Christian M, Arbuck S, Hollingshead M and Sausville EA: Relationships between drug activity in NCI preclinical in vitro and in vivo models and early clinical trials. Br J Cancer. 84:1424–1431. 2001. View Article : Google Scholar : PubMed/NCBI

145 

Ni Z, Nie X, Zhang H, Wang L, Geng Z, Du X, Qian H, Liu W and Liu T: Atranorin driven by nano materials SPION lead to ferroptosis of gastric cancer stem cells by weakening the mRNA 5-hydroxymethylcytidine modification of the Xc-/GPX4 axis and its expression. Int J Med Sci. 19:1680–1694. 2022. View Article : Google Scholar : PubMed/NCBI

146 

Shanavas S, Sen U, Banerjee R, Shenoy PS and Bose B: Effective targeting of colorectal cancer stem cells by inducing differentiation mediated by low-dose vitamin C via β-catenin retention in the cell membrane. J Cell Biochem. 126:e306862025. View Article : Google Scholar

147 

Paganelli F, Chiarini F, Palmieri A, Martinelli M, Sena P, Bertacchini J, Roncucci L, Cappellini A, Martelli AM, Bonucci M, et al: The Combination of AHCC and ETAS decreases migration of colorectal cancer cells, and reduces the expression of LGR5 and Notch1 genes in cancer stem cells: A novel potential approach for integrative medicine. Pharmaceuticals (Basel). 14:13252021. View Article : Google Scholar : PubMed/NCBI

148 

Mao Y, Shangguan D, Huang Q, Xiao L, Cao D, Zhou H and Wang YK: Emerging artificial intelligence-driven precision therapies in tumor drug resistance: Recent advances, opportunities, and challenges. Mol Cancer. 24:1232025. View Article : Google Scholar : PubMed/NCBI

149 

Zhang Z, Wang ZX, Chen YX, Wu HX, Yin L, Zhao Q, Luo HY, Zeng ZL, Qiu MZ and Xu RH: Integrated analysis of single-cell and bulk RNA sequencing data reveals a pan-cancer stemness signature predicting immunotherapy response. Genome Med. 14:452022. View Article : Google Scholar : PubMed/NCBI

150 

Xia X, Zhu C, Zhong F and Liu L: TransCDR: A deep learning model for enhancing the generalizability of drug activity prediction through transfer learning and multimodal data fusion. BMC Biol. 22:2272024. View Article : Google Scholar : PubMed/NCBI

151 

Zhao X, Singhal A, Park S, Kong J, Bachelder R and Ideker T: Cancer mutations converge on a collection of protein assemblies to predict resistance to replication stress. Cancer Discov. 14:508–523. 2024. View Article : Google Scholar : PubMed/NCBI

152 

Varinelli L, Illescas O, Lorenc EJ, Battistessa D, Di Bella M, Zanutto S and Gariboldi M: Organoids technology in cancer research: from basic applications to advanced ex vivo models. Front Cell Dev Biol. 13:15693372025. View Article : Google Scholar : PubMed/NCBI

153 

Beshiri ML, Tice CM, Tran C, Nguyen HM, Sowalsky AG, Agarwal S, Jansson KH, Yang Q, McGowen KM, Yin J, et al: A PDX/Organoid biobank of advanced prostate cancers captures genomic and phenotypic heterogeneity for disease modeling and therapeutic screening. Clin Cancer Res. 24:4332–4345. 2018. View Article : Google Scholar : PubMed/NCBI

154 

Zhu Z, Shen J, Ho PC, Hu Y, Ma Z and Wang L: Transforming cancer treatment: Integrating patient-derived organoids and CRISPR screening for precision medicine. Front Pharmacol. 16:15631982025. View Article : Google Scholar : PubMed/NCBI

155 

Li P, Huang M, Li M, Li G, Ma Y, Zhao Y, Wang X, Zhang Y and Shi C: Combining molecular characteristics and therapeutic analysis of PDOs predict clinical responses and guide PDAC personalized treatment. J Exp Clin Cancer Res. 44:722025. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2025
Volume 67 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cao X, Geng X, Zhang C and Li L: Decoding the role of cancer stem cells in digestive tract tumors: Mechanisms and therapeutic implications (Review). Int J Oncol 67: 61, 2025.
APA
Cao, X., Geng, X., Zhang, C., & Li, L. (2025). Decoding the role of cancer stem cells in digestive tract tumors: Mechanisms and therapeutic implications (Review). International Journal of Oncology, 67, 61. https://doi.org/10.3892/ijo.2025.5767
MLA
Cao, X., Geng, X., Zhang, C., Li, L."Decoding the role of cancer stem cells in digestive tract tumors: Mechanisms and therapeutic implications (Review)". International Journal of Oncology 67.1 (2025): 61.
Chicago
Cao, X., Geng, X., Zhang, C., Li, L."Decoding the role of cancer stem cells in digestive tract tumors: Mechanisms and therapeutic implications (Review)". International Journal of Oncology 67, no. 1 (2025): 61. https://doi.org/10.3892/ijo.2025.5767