Abstract
Cancer stem cells (CSCs) also known as tumor stem cells (TSCs), are pivotal in cancer development and progression. They can be identified through specific markers and surface proteins (e.g., CD44, CD133) that differ from those on non-CSC tumor cells. As well high CSC levels often correlate with poor prognosis, aggressive disease, and resistance to conventional therapies. CSCs are more resistant to standard treatments like chemotherapy and radiation, leading to relapse and metastasis. Therapies targeting CSC-specific pathways and markers are developing to improve treatment efficacy and prevent recurrence. Understanding their role and mechanisms is essential for developing more effective and targeted cancer treatments. While significant progress has been made in developing therapies to target tumor stem cells, much work remains. Tracking CSC markers can help monitor residual disease and predict the likelihood of recurrence, allowing for more tailored treatment approaches. Research into CSC biomarkers continues to evolve, with ongoing studies aiming to refine their use for better diagnosis, prognosis, and treatment of various cancers. Overall, using CSCs as biomarkers offers a promising avenue for more personalized and effective cancer treatment strategies.
Introduction
Normal stem cells, including embryonic and adult types, are crucial for growth, development, and tissue repair, while tumor stem cells self-renew and divide into various cancer cell types [1]. Cancer stem cells (CSCs) also known as Tumor stem cells (TSCs), are a subpopulation of cells within a tumor that possess unique properties compared to the rest of the tumor cells, first found in the context of acute myeloid leukemia in 1994 [2,3], later on also confirmed in cancers of brain, breast, lung, liver, pancreas, colon, and prostate cancer [4]. CSCs exhibit self-renewal, differentiation, and tumor initiation and maintenance, generating new cells to sustain tumors, forming various types of cancer cells, and presumably initiating and maintaining tumors [5]. CSCs, unlike other cancer cells, can produce tumors upon transplantation, exhibiting resistance and metastasis, potentially contributing to cancer spread through tissue invasion and bloodstream entry [6]. CSCs often exhibit resistance to conventional therapies such as chemotherapy and radiation, which can lead to relapse and metastasis [7,8]. CSCs' regulation is less understood than normal stem cells, potentially affecting signaling pathways and tumor microenvironment interactions. Understanding the differences between normal stem cells and CSCs is crucial for developing targeted cancer therapies [9].
Contribution of TSCs to Cancer Development
Tumor growth and maintenance
CSCs are thought to be responsible for initiating tumors. They can form new tumors even from a small number of cells, which makes them crucial in the initial stages of cancer development [10]. CSCs can self-renew and are essential for initiating and sustaining tumor growth. Even at low cell numbers, TSCs can initiate tumors when transplanted into immunocompromised mice [11]. CSCs generate diverse cancer cell populations, maintain tumor growth, and interact with their surrounding microenvironment to maintain the tumor's diverse population, including immune cells to protect themselves from immune clearance and to provide signals that support their survival and proliferation [12].
Invasion and spread
CSCs use immune checkpoint molecules and extracellular vesicles to evade immune surveillance, enabling them to spread and establish secondary tumors, despite their crucial role in maintaining self-tolerance [13,14]. These pathways help to maintain self-tolerance, preventing the immune system from attacking normal cells indiscriminately [15]. However, some cancers exploit these checkpoints to protect themselves from immune attack. Here are some key immune checkpoint molecules including CTLA-4 (Cytotoxic T Lymphocyte Antigen-4) [16], PD-1 (Programmed Cell Death Protein 1) and PD-L1 (Programmed Death Ligand 1) [17], TIM-3 (T Cell Immunoglobulin and Mucin Domain-Containing Protein 3) [18] and CD27, CD28, CD40, and CD137, etc [19,20]. Certainly, Immune checkpoint inhibitors, which are used in cancer treatment, can have both common and rarer side effects. Natural killer (NK) cells can also recognize and kill CSCs, but CSCs often develop mechanisms to escape this targeting [21]. There are reports that exosomes from CSCs interact with immune cells, affecting signaling pathways and immune responses. CSC-derived exosomes can suppress T-cell function and induce immunosuppressive macrophages as well exosome-based drug delivery systems show promise in targeting CSCs with improved precision and reduced side effects [22].
Genetic and epigenetic alterations
CSCs' genetic and epigenetic alterations drive cancer development, modulating cell growth, survival, and differentiation, impacting tumor response and contributing to therapeutic resistance [23]. Epigenetic mechanisms impact CSC behavior in various ways as DNA methylation involves hypermethylation of tumor suppressor genes that can silence their expression, for example, genes like p16INK4a (a tumor suppressor gene) are frequently hypermethylated in CSCs [24]. Global DNA hypomethylation can activate oncogenes or repetitive elements that promote genomic instability and CSC properties. Furthermore, histones undergo modifications (acetylation, methylation, phosphorylation) that affect chromatin structure. These modifications regulate gene accessibility, impacting CSC properties [25]. CSCs often carry mutations in genes that drive cancer, such as tumor protein p53(TP53), Kirsten rat sarcoma viral oncogene homolog (KRAS), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), and breast cancer susceptibility genes (BRCA1/2) [26]. Overexpression or activation of transcription factors like Oct4, Sox2, Nanog, and KLF4 are commonly observed in CSCs. These factors influence stem-like characteristics and pluripotency of CSCs, which can be enhanced by mutations, contributing to tumor growth and therapy resistance [27,28]. Moreover, dysregulation of miRNAs can influence CSC self-renewal and differentiation [29]. Key pathways (Wnt/β-catenin, Notch, Hedgehog, TGF-β/BMP) are altered in CSCs through epigenetic changes. These alterations maintain stemness, differentiation, and drug resistance in CSCs [3].
Biomarkers
Cancer stem cells (CSCs) serve diverse roles in cancer progression, providing biomarkers for prognosis and therapy response, although their clinical application is still evolving and limited in routine practice [30]. Here are some of the clinically relevant cancer stem cell biomarkers that have been explored or approved for use in cancer diagnostics or treatment planning:
CD44 and its variants
CD44 is a transmembrane glycoprotein, highly expressed in CSCs and involved in cell adhesion, migration, and interaction with the extracellular matrix. It influences various signaling pathways, including the Wnt, Notch, and Hedgehog pathways, essential for maintaining CSC properties [31]. CD44 has multiple isoforms, with CD44s being the most common and smallest, and CD44v being overexpressed in tumor cells [32]. CD44 is a widely used marker for cancer stem cells in various malignancies, including breast, pancreas, prostate, colorectal, ovarian, lung, liver, head/neck, blood, bladder, gastric, brain, bone, and cervical. Also, it is found in stem cells in various regions [33,34]. The CD44 expression is elevated in metastatic lymph nodes of breast cancer compared to original tumors, unlike CD44 knockdown yields contrary results [35]. CD44 overexpression increases cell invasion and migration, leading to worse prognosis and more aggressive tumor behavior. The many isoforms of CD44, a crucial component of colorectal cancer, affect the disease's onset, progression, metastasis, and therapy resistance [36]. CD44, a potential therapeutic target in leukemia treatment, has the potential to eradicate CSCs and prevent relapse [37]. CD44-targeting strategies are being explored in clinical trials to inhibit tumor growth and metastasis, aiding in the identification of CSCs, although not directly used as a treatment target. Although, a few medications that target CD44 have been authorized for use in clinical trials [32]. Hyaluronic acid (HA) is used for CD44 targeted delivery to eradicate cancer cells and CSCs, while silibinin (SIL) and cabazitaxel (CBX) are co-loaded onto cationic liposomes for prostate cancer stem cell treatment [38,39].
CD45
It is also known as protein tyrosine phosphatase receptor type C (PTPRC). CD45 is used to identify and isolate cancer stem cells in various types of cancer, including leukemia [40, 41] and solid tumors [42]. High CD45 expression levels have been associated with better prognosis in certain cancers, such as bladder cancer [43]. Glatting et al., [44] reported that when tagged with 90Y, the anti-CD45 monoclonal antibody YAML568 can deliver radiation to hematopoietic regions selectively. Targeting CD45 and its associated pathways can potentially enhance the effectiveness of cancer treatments, especially immunotherapies [45]. The bispecific Abs (target CD45 and Y-DOTA) effectively delivered radiation to leukemia cells, reducing tumor growth and improving survival rates in both murine and human leukemia models [46].
CD87
CD87, also known as the Urokinase-type plasminogen activator receptor (uPAR), is indeed used as a cancer stem cell (CSC) marker in lung cancer. It helps distinguish lung cancer stem cells from other types of cancer stem cells, such as those found in blood cancers [47]. Targeting CD87 could potentially disrupt processes such as cell adhesion, migration, and interaction with the extracellular matrix (ECM), making it a promising therapeutic target for lung cancer treatment [48]. uPAR-targeted therapeutic strategies have shown potential in animal models, but no uPAR-targeted agents have been developed or evaluated in cancer clinical trials. Although phase I clinical trials employing 64Cu-DOTA-AE105 are already in progress, humanized ATN-658 is still awaiting translation to diagnose aggressive malignancies and evaluate cancer aggressiveness [49].
CD90
CD90, also known as Thy-1, is a glycoprotein, an important biomarker for cancer stem cells in multiple cancer types, including the brain [50], liver [51], colorectal [52], and breast cancers [53]. CD90 is not exclusively expressed by cancer stem cells. It is also expressed in mesenchymal stem cells and liver stem cells [54]. CD90 expression, particularly in triple-negative breast cancer (TNBC), is an aggressive subtype associated with poor prognosis and limited treatment options [53]. According to previous studies, CD90-positive cells express more CD133 than CD90-negative tumor cells, and signaling analyses showed that CD90 induction of CD133 requires β3 integrin and adenosine monophosphate (AMP)-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) [55,56]. During in vitro studies, the survival and carcinogenic potential of CD90+ HCC cells are found to be controlled by CD44 [57].
CD133 (Prominin-1)
CD133 is another stem cell marker that has been used to identify cancer stem cells in several cancers, including glioblastoma, colon cancer, pancreatic cancer, and breast cancer [58]. CD133+ cells are enriched in the CSC population and are associated with increased tumorigenicity and resistance to chemotherapy. Research is ongoing to assess the therapeutic targeting of CD133, particularly in combination with other CSC markers [59]. Currently authorized treatments do not specifically target CD133, it is utilized in experimental treatments and clinical studies to detect the existence of cancer stem cells [60,61]. Several approaches, such as Sorafenib, a multi-kinase inhibitor, are being explored, which diminishes CD133 expression and suppresses tumor growth in vivo. Additionally, Nifuroxazide, a STAT3 inhibitor, diminishes CD133 expression and suppresses tumor growth in vivo [62]. Trifluridine (FTD)/tipiracil (TPI) is an oral combination medication that may be efficacious against colorectal cancer cells with high levels of CD44 and CD133 expression [63].
Leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5)
Leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5) is a promising biomarker for cancer stem cells (CSCs) [64]. Aberrant overexpression of Lgr5 is common in some human cancers, leading to the potentiation of canonical Wnt/β-catenin signaling [65]. Lgr5 is used to identify and isolate CSCs in various types of cancers, including colorectal, gastric, and esophageal cancers [66].
Epithelial cell adhesion molecule (EpCAM)
EpCAM is a cell surface glycoprotein overexpressed in many epithelial cancers, including breast, colorectal, and lung cancers [67]. It is involved in the regulation of cell cycle progression and can act as a transcription factor by activating genes like c-myc, cyclin A, and cyclin. EpCAM has oncogenic potential and can contribute to cancer progression by interacting with various signaling pathways, such as Wnt/β-catenin, TGF-β/SMAD, and PI3K/AKT/mTOR [68]. It is used in liquid biopsy techniques, such as the detection of circulating tumor cells (CTCs), which are thought to originate from cancer stem cells. Targeting EpCAM is being explored as a strategy to eliminate cancer stem cells in solid tumors [69]. Moreover, bispecific antibodies and CAR T-cell therapy have shown promise in eliminating CSCs and improving treatment outcomes. While not yet FDA-approved as a direct CSC target, EpCAM is being explored as part of clinical trials targeting CTCs in cancers [70].
Aldehyde dehydrogenase 1 (ALDH1)
In breast cancer, high populations of ALDH1 are associated with triple-negative breast cancer (TNBC), a subtype that is particularly aggressive and difficult to treat [71] ALDH1 is an enzyme that detoxifies aldehydes and is a key marker for cancer stem cells in various cancers, including breast, liver, and lung cancers [72]. Elevated ALDH1 expression is linked to poor prognosis, chemoresistance, and aggressive tumor behavior. Clinical trials are investigating ALDH1 as a target for therapy, as well as its use as a biomarker to assess the stem cell population in tumors. ALDH1A3 with the other two markers CD24-/CD44 + and CD24 + /CD44 + was found to be associated with poor prognosis, limited response to chemotherapy, higher chances of metastasis, and aggressive tumor formation in breast cancers [73]. However, direct FDA approval for therapeutic targeting of ALDH1 in CSCs is not yet available.
Human epidermal growth factor receptor 2 (HER2)
HER2 is an important biomarker and therapeutic target in breast cancer, particularly in breast cancer stem cells (BCSCs) [74]. It was found to be overexpressed in approximately 15-20% of breast cancers (oncologypro.esmo.org). While not strictly a cancer stem cell marker, HER2 overexpression is associated with aggressive breast cancers, particularly HER2-positive breast cancer. HER2 signaling has bidirectional communication with stemness-related pathways, influencing both the maintenance of BCSCs and the response to targeted therapies. It interacts with stemness-related pathways, such as Notch and Wnt/β-catenin, contributing to the self-renewal and differentiation properties of BCSCs [74]. A splice variant of full-length HER2 mRNA, d16HER2, has been identified as one of the most oncogenic isoforms, significantly implicated in tumorigenesis and epithelial-mesenchymal transition (EMT) [75]. HER2-targeted therapies, such as trastuzumab (Herceptin), Pertuzumab (Perjeta), Ado-Trastuzumab Emtansine (T-DM1, Kadcyla), Lapatinib (Tykerb), Neratinib (Nerlynx), Tucatinib (Tukysa) are used to treat HER2-positive breast cancer [76]. HER2-positive cancer stem cells are often more resistant to conventional therapies, making HER2-targeted therapy an important approach. These treatments aim at eradicating both differentiated cancer cells and cancer stem cells that express HER2 [77].
Cytokeratins (e.g., CK5, CK14, CK19)
Cytokeratins are proteins found in the cytoskeleton of epithelial cells and are used as biomarkers for certain types of cancer stem cells [78]. In cancers like breast cancer, certain cytokeratins (e.g., CK5 and CK14) are expressed in basal-like breast cancer subtypes, which are often triple-negative (lacking estrogen receptor, progesterone receptor, and HER2), often harbor cancer stem cells [79,80]. There is a strong association between CK5/14-positive tumors and BRCA1 mutations. These tumors are often found in patients with hereditary breast cancer [79]. CK19, an upregulated gene in ovarian cancer tissue, promotes cell invasion, proliferation, and migration, activating the Wnt/β-catenin signaling pathway through β-catenin, TCF7, LEF1, c-MYC, and cyclin D1 [81]. A novel CSC marker, CK19 is linked to TGFb/Smad signaling and EMT in hepatocellular carcinoma (HCC) [82]. CK expression patterns help identify stem-like cells and predict the aggressiveness of tumors. Cytokeratin-based markers are used for diagnostic purposes, particularly in pathology to distinguish different subtypes of cancer. However, direct therapeutic targeting of these markers is still under investigation [83].
Trophoblast antigen 2 (Trop2)
Trop2 is a transmembrane protein involved in cell proliferation and survival and is expressed in many cancer stem cells [84]. Trop2 is overexpressed in various cancers, including breast cancer, and is associated with tumor progression and stem-like properties. The FDA approved sacituzumab govitecan in 2020 for treating metastatic triple-negative breast cancer, and in 2021 for hormone receptor-positive, HER2-negative breast cancer. This therapy is particularly beneficial for treating cancers that express high levels of Trop2, which are often resistant to standard treatments [85]. This FDA-approved antibody-drug conjugate targets Trop2 for treating metastatic triple-negative breast cancer (TNBC) [86].
Ki-67 (Proliferation marker)
Ki-67 is expressed in all active phases of the cell cycle (G1, S, G2, and M) but is absent in resting (G0) cells. It is widely used to assess the proliferation rate of tumor cells [87]. However, its role in CSCs extends beyond mere proliferation, as it is also involved in maintaining the stem cell phenotype. Genetic disruption of Ki-67 in human epithelial breast and colon cancer cells deplete the cancer stem cell niche [88]. It is widely used in clinical settings to predict outcomes in various cancers, including breast [89], prostate [90], and colon cancers [91]. Ki-67 is routinely used in clinical diagnostics to guide treatment decisions, especially in breast cancer, where it helps to classify tumors as high or low grade based on proliferation [92]. Future studies should focus on standardizing Ki67 measurement and exploring its combination with other biomarkers to improve its predictive value.
P-glycoprotein (MDR1)
P-glycoprotein, a membrane-bound transporter, is involved in multidrug resistance by pumping out drugs from cells. It is often overexpressed in cancer stem cells, contributing to their resistance to chemotherapy [93]. P-glycoprotein in CSCs can be used to predict drug resistance and guide treatment strategies [94]. In clinical practice, P-glycoprotein inhibitors (e.g., verapamil) are sometimes used in combination with chemotherapy to overcome resistance, though the direct targeting of P-glycoprotein in CSCs is not yet FDA-approved [95]. Here’s an overview of how CSCs are utilized as biomarkers in different cancers, discussed in Table 1.
Hence, CD133, CD44, and EpCAM have been recognized as prospective cancer stem cell markers in various solid and non-solid tumors.
|
Types of Cancer |
Markers |
References |
|
Breast Cancer |
CD29+, CD49f+, CD90+, CD 133, CD44+/CD24-, ALDH1, HER2, and EpCAM. |
[30, 96-101] |
|
Colorectal Cancer |
CD133, CD44, CD24, CD29, CD166, EpCAM, CXCR4 ALDH1, Oct4, Sox2, Nanog and Lgr5 |
[102-105] |
|
Lung Cancer |
CD44+, CD133, CD166+, CD90+ CD133, CD87+, ALDH1, and Sox2 |
[106-111] |
|
Ovarian Cancer |
CD133, CD44, CD24, CD117 or ALDH1, PDL-1, CD105, CD106, SOX2, EpCAM, Nestin and SSEA1 |
[112-121] |
|
Glioblastoma |
CD49f+ CD90+ CD44+, CD36+ CD133, Nestin, and Sox2, EGFR+ A2B5+, L1CAM+, CD133+ |
[122-125] |
|
Prostate Cancer |
EpCAM+, CD117+, α2β1+, ALDH+, CD44+, EZH2+, CXCR4+, E-cadherin+ CD133+, CD44, ALDH1, and AR variants |
[126-131] |
|
Pancreatic Cancer |
CD24, CD44, CD133, ESA, ALDH1, Nanog, EpCAM, CXCR4, DCLK1, c-MET, ABCG2 and Lgr5 |
[132-135] |
|
Hematologic Malignancies (e.g.,Leukemias, Lymphomas) |
CD45, CD38, CD19, CD22, CD34, CD123, Lgr5, Oct4, Sox2, Nanog, c-kit, ABCG2, ALDH, CLL-1 and her lineage-specific markers. |
[136,137 ] |
|
Cervical Cancer |
CD44, CD133 (Promin), CD111 (c-Kit), CD24, CD117, OCT4, ALDH1, LGR5 |
[138-143] |
|
Liver Cancer |
CD133, CD90, CD44, CD24, CD13, EpCAM, and cytokeratin 19 (CK19), |
[144,145] |
Therapeutic Strategies
Cancer treatment researchers are exploring strategies to target specific markers or pathways associated with CSCs to achieve long-term remission and prevent relapse [146]. CSCs are often more resistant to conventional treatments such as chemotherapy and radiation. This resistance is due to various factors, including enhanced DNA repair, drug efflux pumps (ATP-binding cassette (ABC), and some enzymes such as aldehyde dehydrogenase (ALDH)) that can expel therapeutic agents from the cells. Also, the state of quiescence results in the maintenance of cells in a latent or quiescent condition, hence reducing their vulnerability to therapeutic interventions that specifically target actively dividing cells. Additionally, in relapse and recurrence, where conventional therapies may not effectively eliminate CSCs, these cells can survive and lead to relapse or recurrence of the tumor after initial treatment [147].
CSCs express specific surface markers that differentiate them from other tumor cells. Targeting these markers can help selectively eradicate TSCs. Monoclonal Antibodies targeting specific CSC markers like CD44, CD133, ALDH1, or ESA (epithelial-specific antigen) are being developed. For example, catumaxomab, an antibody targeting EpCAM and receptor of T cells-CD3, has been investigated for CSC targeting [148,149]. Chimeric antigen receptor T-cell (CAR-T) therapy can be engineered to recognize and kill CSCs by targeting surface antigens such as CD133 and CD44 [150]. Whereas, targeting signaling pathways (Wnt/β-catenin pathway, Notch pathway, Hedgehog pathway, PI3K/Akt/mTOR pathway) can inhibit CSC function and reduce tumor recurrence [3]. Agents like decitabine and azacitidine, which inhibit DNA methyltransferases, can reverse the silencing of tumor suppressor genes in CSCs. Drugs like vorinostat, panobinostat, and romidepsin can modulate histone acetylation and disrupt CSC self-renewal and differentiation [151]. EZH2, a key Polycomb repressive complex 2 (PRC2) member, is often overexpressed in CSCs [152]. Inhibitors like tazemetostat are being explored to target EZH2 and prevent CSC maintenance [153]. Bromodomain and extra-terminal domain (BET) inhibitors like JQ1 target proteins that regulate gene expression via histone acetylation, showing potential in eliminating CSCs by modulating transcriptional networks critical for stemness [154].
CSCs play a critical role in mediating resistance to anti-cancer therapies through a variety of mechanisms (Figure 1). Furthermore, pharmacological repurposing and the integration of micronutrients are capable of targeting both malignancies and cancer stem cells. However, ongoing research has led to the development of various approaches aimed at targeting and eradicating these cells [155,156]. CSCs often rely on aerobic glycolysis for energy production (the Warburg effect). Targeting enzymes like hexokinase 2 or pyruvate kinase M2 can inhibit glucose metabolism and reduce CSC viability [157]. While inhibiting fatty acid oxidation or metabolism, it can also target CSCs, as they depend on this pathway for maintaining stemness and growth [158]. Targeting mitochondrial functions, such as metformin or IACS-010759, can disrupt the metabolic networks that sustain CSCs [159]. Agents like retinoids, vitamin D analogs, and histone deacetylase inhibitors (HDACi) can promote the differentiation of CSCs, decreasing their ability to self-renew and initiate tumors [7,160]. Bone morphogenetic protein (BMP) signaling can promote differentiation and decrease stemness in CSCs. Activating this pathway by BMP agonists is being studied as a potential therapy [161]. Drugs targeting immune checkpoints like PD-1/PD-L1 and CTLA-4 can enhance anti-tumor immunity and potentially eliminate CSCs [162,163]. Targeting CSC-specific antigens through cancer vaccines could provide a strategy to selectively activate immune responses against TSCs [164]. Engineered viruses can be designed to specifically target and eliminate cancer stem cells while preserving normal cells. For example, oncolytic adenoviruses have been designed to target CSCs specifically [165]. Nanocarriers can deliver gene therapies that target key genes involved in CSC self-renewal and survival [166].
Figure 1. Various mechanisms responsible for mediating resistance to anti-cancer therapies by CSCs.
Combining traditional chemotherapy with CSC-targeting agents (e.g., Wnt inhibitors, HDAC inhibitors) can enhance the effectiveness of treatment and prevent tumor recurrence [167]. Combining immune checkpoint inhibitors with epigenetic modulators can potentially enhance the immune response to CSCs by modifying their immune profile [168]. Blocking the formation of blood vessels (angiogenesis) in the tumor microenvironment can starve CSCs of nutrients and oxygen, making them more vulnerable to treatment. Drugs like bevacizumab (anti-VEGF) are being explored [169]. Intervening with the stromal cells in the tumor microenvironment, such as fibroblasts and mesenchymal stem cells, can disrupt the supportive niche and reduce CSC survival [170]. Gene editing technologies such as CRISPR/Cas9 can directly inactivate genes critical for cancer stem cell self-renewal, survival, or metastasis. Focusing on oncogenes or tumor suppressors that govern stemness may diminish CSC viability and avert recurrence [171]. Also, matrix metalloproteinase inhibitors target enzymes involved in tissue remodeling and could impact the supportive microenvironment for CSCs [172]. CSCs' heterogeneity and varying markers make developing one-size-fits-all treatments challenging. Careful targeting is necessary to avoid damaging normal stem cells or causing significant side effects. The study emphasizes the importance of understanding and overcoming the resistance mechanisms of CSCs to various therapies, as they are often more resistant to conventional treatments [173].
Concluding Remarks
Researchers are working on better ways to identify and isolate CSCs to understand their biology and develop targeted therapies. Additionally, finding reliable biomarkers for CSCs is crucial for early detection and monitoring treatment response. CSC markers can be used for early detection of cancer, as they may be present in circulating tumor cells or biopsy samples before conventional markers become evident. The presence and proportion of CSCs often correlate with a more aggressive disease course and poorer prognosis. While several cancer stem cell biomarkers are being studied and utilized in clinical trials, few have received formal FDA approval for direct therapeutic targeting. However, CD44, ALDH1, HER2, and Trop2 represent the most clinically significant CSC markers currently used to guide treatment decisions or included in FDA-approved therapies (e.g., trastuzumab for HER2+ cancers, sacituzumab govitecan for Trop2-expressing cancers). The clinical utility of CSC markers is rapidly evolving as research continues to uncover new potential targets and therapies aimed at eradicating cancer stem cells and improving patient outcomes. Developing drugs that specifically target CSCs, or their pathways can potentially improve treatment outcomes and reduce relapse rates. Overall, understanding the role of tumor stem cells is essential for developing more effective cancer treatments and improving patient outcomes.
Abbreviations
ALDH: Aldehyde Dehydrogenase; AR: Androgen Receptor; ABC: ATP-Binding Cassette; BRCA1/2: Breast Cancer Susceptibility genes; BET: Bromodomain and Extra-Terminal Domain; BMP: Bone Morphogenetic Protein; SIL: Silibinin; CBX: Cabazitaxel; CSCS: Cancer Stem Cells; CAR: Chimeric Antigen Receptor; CAR-T: Chimeric Antigen Receptor T-cell; CD: Cluster Domain; CTLA-4: Cytotoxic T Lymphocyte Antigen-4; CK: Cytokeratin; DCs: Dendritic Cells; ESA: Epithelial-Specific Antigen; EVS: Extracellular Vesicles; HER2: Human Epidermal Growth Factor Receptor 2; HDACI: Histone Deacetylase Inhibitors; KRAS: Kirsten Rat Sarcoma Viral Oncogene Homolog; NK-Cells: Natural Killer Cell; OVS: Oncolytic Viruses; PD-1: Programmed Cell Death Protein 1; PD-L1: Programmed Death Ligand 1; PIK3CA: Phosphatidylinositol-4, 5-bisphosphate 3-kinase Catalytic Subunit Alpha; PTPRC: Protein Tyrosine Phosphatase Receptor Type C; TIM-3: T Cell Immunoglobulin and Mucin Domain-Containing Protein 3; TP53: Tumor Protein p53; TSCS: Tumor Stem Cells; Sox2: SRY-Box Transcription Factor 2
Declarations
Competing interests
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
No authors have any potential conflicts of interest to disclose.
Funding disclosure statement
Not applicable as this work was not supported by the Funding Agency or under any grant.
Ethics approval statements
Not applicable.
Author contributions
Conceptualization: Linz Buoy George; Study design: Dhara Jani; Original draft preparation and analysis: Urja Joshi; Formal analysis and investigation: Hyacinth Highland; Review and editing: All authors.
Data, material, and/or code availability
Not applicable.
References
2. Dick JE. Acute myeloid leukemia stem cells. Ann NY Acad Sci. 2005 Jun;1044(1):1-5.
3. Zeng Z, Fu M, Hu Y, Wei Y, Wei X, Luo M. Regulation and signaling pathways in cancer stem cells: implications for targeted therapy for cancer. Mol Cancer. 2023 Oct 18;22(1):172.
4. Wu B, Shi X, Jiang M, Liu H. Cross-talk between cancer stem cells and immune cells: potential therapeutic targets in the tumor immune microenvironment. Mol Cancer. 2023 Feb 21;22(1):38.
5. Garg S, Yadav R, Garg K, Khan H, Kumar D. Cancer stem cells and maintenance of tumor heterogeneity/microenvironment. In: Pathak S, Banerjee A, eds. Cancer Stem Cells and Signaling Pathways. New York: Academic Press; 2024. pp. 517-29.
6. Borlongan MC, Saha D, Wang H. Tumor microenvironment: A niche for cancer stem cell immunotherapy. Stem Cell Rev Rep. 2024 Jan;20(1):3-24.
7. Guo Q, Zhou Y, Xie T, Yuan Y, Li H, Shi W, et al. Tumor microenvironment of cancer stem cells: Perspectives on cancer stem cell targeting. Genes & Diseases. 2024 May 1;11(3):101043.
8. Kulus M, Farzaneh M, Bryja A, Zehtabi M, Azizidoost S, Abouali Gale Dari M, et al. Phenotypic Transitions the Processes Involved in Regulation of Growth and Proangiogenic Properties of Stem Cells, Cancer Stem Cells and Circulating Tumor Cells. Stem Cell Rev Rep. 2024 Feb 19:1-3.
9. Stouras I, Vasileiou M, Kanatas PF, Tziona E, Tsianava C, Theocharis S. Metabolic profiles of cancer stem cells and normal stem cells and their therapeutic significance. Cells. 2023 Nov 22;12(23):2686.
10. Nairuz T, Mahmud Z, Manik RK, Kabir Y. Cancer stem cells: an insight into the development of metastatic tumors and therapy resistance. Stem Cell Rev Rep. 2023 Aug;19(6):1577-95.
11. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med.1997 Jul 1;3(7):730-7.
12. Plaks V, Kong N, Werb Z. The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells?. Cell Stem Cell. 2015 Mar 5;16(3):225-38.
13. Hinshaw DC, Shevde LA. The tumor microenvironment innately modulates cancer progression. Cancer Res. 2019 Sep 15;79(18):4557-66.
14. Sistigu A, Musella M, Galassi C, Vitale I, De Maria R. Tuning cancer fate: tumor microenvironment's role in cancer stem cell quiescence and reawakening. Front Immunol. 2020 Oct 21;11:2166.
15. Miko E, Meggyes M, Doba K, Barakonyi A, Szereday L. Immune checkpoint molecules in reproductive immunology. Front Immunol. 2019 Apr 18;10:846.
16. Zheng F, Dang J, Zhang H, Xu F, Ba D, Zhang B, et al. Cancer stem cell vaccination with PD-L1 and CTLA-4 blockades enhances the eradication of melanoma stem cells in a mouse tumor model. J Immunother. 2018 Oct 1;41(8):361-8.
17. Ai L, Chen J, Yan H, He Q, Luo P, Xu Z, et al. Research status and outlook of PD-1/PD-L1 inhibitors for cancer therapy. Drug Des Devel Ther. 2020 Sep 8:3625-49.
18. Gao L, Yu S, Zhang X. Hypothesis: Tim-3/galectin-9, a new pathway for leukemia stem cells survival by promoting expansion of myeloid-derived suppressor cells and differentiating into tumor-associated macrophages. Cell Biochem Biophys. 2014 Sep;70:273-7.
19. Lou Y, Diao L, Cuentas ER, Denning WL, Chen L, Fan YH, et al. Epithelial–mesenchymal transition is associated with a distinct tumor microenvironment including elevation of inflammatory signals and multiple immune checkpoints in lung adenocarcinoma. Clin Cancer Res. 2016 Jul 15;22(14):3630-42.
20. Marhelava K, Pilch Z, Bajor M, Graczyk-Jarzynka A, Zagozdzon R. Targeting negative and positive immune checkpoints with monoclonal antibodies in therapy of cancer. Cancers. 2019 Nov 8;11(11):1756.
21. Guo F, Zhang Y, Bai L, Cui J. Natural killer cell therapy targeting cancer stem cells: old wine in a new bottle. Cancer Lett. 2023 Jul 26:216328.
22. Keyvani-Ghamsari S, Khorsandi K, Rasul A, Zaman MK. Current understanding of epigenetics mechanism as a novel target in reducing cancer stem cells resistance. Clin Epigenetics. 2021 Dec;13(1):120.
23. Toh TB, Lim JJ, Chow EK. Epigenetics in cancer stem cells. Mol. Cancer. 2017 Dec;16:1-20.
24. VD P, V V. Targeting epigenetic alterations in cancer stem cells. Front Mol Med. 2022 Sep 20;2:1011882.
25. Quagliano A, Gopalakrishnapillai A, Barwe SP. Understanding the mechanisms by which epigenetic modifiers avert therapy resistance in cancer. Front Oncol. 2020 Jun 24;10:992.
26. Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013 May 30;368(22):2059-74.
27. Li W, Tian E, Chen ZX, Sun G, Ye P, Yang S, et al. Identification of Oct4-activating compounds that enhance reprogramming efficiency. Proc Natl Acad Sci USA. 2012 Dec 18;109(51):20853-8.
28. Zhang Q, Han Z, Zhu Y, Chen J, Li W. The role and specific mechanism of OCT4 in cancer stem cells: a review. Int J Stem Cells. 2020 Nov 30;13(3):312-25.
29. Ortiz-Sánchez E. Overview: Epigenetic Regulation in Cancer Stem Cells by Methylation. Austin J Cancer Clin Res. 2014;1(1):1007.
30. Conde I, Ribeiro AS, Paredes J. Breast cancer stem cell membrane biomarkers: therapy targeting and clinical implications. Cells. 2022 Mar 9;11(6):934.
31. Mu W, Xu Y, Gu P, Wang W, Li J, Ge Y, et al. Exosomal CD44 cooperates with integrin α6β4 to support organotropic metastasis via regulating tumor cell motility and target host cell activation. Engineering. 2021 Oct 1;7(10):1413-23.
32. Li M, Sun J, Zhang W, Zhao Y, Zhang S, Zhang S. Drug delivery systems based on CD44-targeted glycosaminoglycans for cancer therapy. Carbohyd Polym. 2021 Jan 1;251:117103.
33. Basakran NS. CD44 as a potential diagnostic tumor marker. Saudi Med J. 2015;36(3):273.
34. Morath I, Hartmann TN, Orian-Rousseau V. CD44: More than a mere stem cell marker. Int J Biochem Cell Biol. 2016 Dec 1;81:166-73.
35. Vadhan A, Hou MF, Vijayaraghavan P, Wu YC, Hu SC, Wang YM, et al. CD44 promotes breast cancer metastasis through AKT-mediated downregulation of nuclear FOXA2. Biomedicines. 2022 Oct 5;10(10):2488.
36. Ziranu P, Pretta A, Aimola V, Cau F, Mariani S, D’Agata AP, et al. CD44: A New Prognostic Marker in Colorectal Cancer?. Cancers. 2024 Apr 19;16(8):1569.
37. Xu H, Niu M, Yuan X, Wu K, Liu A. CD44 as a tumor biomarker and therapeutic target. Exp Hematol Oncol. 2020 Dec;9:1-4.
38. Mahira S, Kommineni N, Husain GM, Khan W. Cabazitaxel and silibinin co-encapsulated cationic liposomes for CD44 targeted delivery: a new insight into nanomedicine based combinational chemotherapy for prostate cancer. Biomed Pharmacother. 2019 Feb 1;110:803-17.
39. Kesharwani P, Chadar R, Sheikh A, Rizg WY, Safhi AY. CD44-targeted nanocarrier for cancer therapy. Front Pharmacol. 2022 Mar 31;12:800481.
40. Nakamura A, Tsurusawa M, Kato A, Taga T, Hatae Y, Miyake M, et al. Prognostic Impact of CD45 Antigen Expression in High-Risk, Childhood B-Cell Precursor Acute Lymphoblastic Leukemia: Children's Cancer and Leukemia Study Group (CCLSG). Leuk. Lymphoma. 2001 Jan 1;42(3):393-8.
41. Harfmann M, Schröder T, Głów D, Jung M, Uhde A, Kröger N, et al. CD45-Directed CAR-T Cells with CD45 Knockout Efficiently Kill Myeloid Leukemia and Lymphoma Cells In Vitro Even after Extended Culture. Cancers. 2024 Jan 12;16(2):334.
42. Saha A, Palchaudhuri R, Lanieri L, Hyzy S, Riddle MJ, Panthera J, et al. Alloengraftment without significant toxicity or GVHD in CD45 antibody-drug conjugate–conditioned Fanconi anemia mice. Blood. 2024 May 23;143(21):2201-16.
43. Rheinländer A, Schraven B, Bommhardt U. CD45 in human physiology and clinical medicine. Immunol.Lett. 2018 Apr 1;196:22-32.
44. Glatting G, Müller M, Koop B, Hohl K, Friesen C, Neumaier B, et al. Anti-CD45 monoclonal antibody YAML568: A promising radioimmunoconjugate for targeted therapy of acute leukemia. J Nucl Med. 2006 Aug 1;47(8):1335-41.
45. Park SY, Kim JY, Jang GB, Choi JH, Kim JH, Lee CJ, et al. Aberrant activation of the CD45-Wnt signaling axis promotes stemness and therapy resistance in colorectal cancer cells. Theranostics. 2021;11(18):8755.
46. Orozco JJ, Kenoyer AL, Lin Y, O'Steen S, Guel R, Nartea ME, et al. Therapy of myeloid leukemia using novel bispecific fusion proteins targeting CD45 and 90Y-DOTA. Mol Cancer Ther. 2020 Dec 1;19(12):2575-84.
47. Skurikhin EG, Ermakova N, Zhukova M, Pershina O, Pan E, Pakhomova A, et al. Analysis of circulating tumor and cancer stem cells provides new opportunities in diagnosis and treatment of small cell lung cancer. Int J Mol Sci. 2022 Sep 17;23(18):10853.
48. Ren Y, Bao X, Feng M, Xing B, Lian W, Yao Y, et al. CD87-targeted BiTE and CAR-T cells potently inhibit invasive nonfunctional pituitary adenomas. Sci China Life Sci. 2024 Oct;67(10):2169-85.
49. Zhai BT, Tian H, Sun J, Zou JB, Zhang XF, Cheng JX, et al. Urokinase-type plasminogen activator receptor (uPAR) as a therapeutic target in cancer. J Transl Med. 2022 Mar 18;20(1):135.
50. He J, Liu Y, Zhu T, Zhu J, DiMeco F, Vescovi AL, et al. CD90 is identified as a candidate marker for cancer stem cells in primary high-grade gliomas using tissue microarrays. Mol Cell Proteomics. 2012 Jun 1;11(6):M111.010744.
51. Niu Q, Ye S, Zhao L, Qian Y, Liu F. The role of liver cancer stem cells in hepatocellular carcinoma metastasis. Cancer Biol. Ther. 2024 Dec 31;25(1):2321768.
52. Aboushousha T, Osama M, Mohamed M, Khaled Y, Elmeligy HA, Ossama Y. Significance of CD70, VEGF, and CD90 Immunohistochemical Expression in Colorectal Cancer. Asian Pac J Cancer Prev. 2024 Oct 1;25(10):3691-9.
53. Sriwidyani NP, Dewi IG, Yuniawaty NG. High CD90 Expression is A Predictor of Axillary Nodal Metastasis in Breast Carcinoma. Indones. J Cancer. 2024 Jun 27;18(2):157-62.
54. Shaikh MV, Kala M, Nivsarkar M. CD90 a potential cancer stem cell marker and a therapeutic target. Cancer Biomark. 2016 Jan 1;16(3):301-7.
55. Jia (a) Q, Zhang X, Deng T, Gao J. Positive correlation of Oct4 and ABCG2 to chemotherapeutic resistance in CD90+ CD133+ liver cancer stem cells. Cell Reprogramming. 2013 Apr 1;15(2):143-50.
56. Jia (b) SQ, Ren JJ, Dong PD, Meng XK. Probing the hepatic progenitor cell in human hepatocellular carcinoma. Gastroenterol Res Pract. 2013;2013(1):145253.
57. El-Houseini ME, Ismail A, Abdelaal AA, El-Habashy AH, Abdallah ZF, Mohamed MZ, et al. Role of TGF-β1 and C-Kit mutations in the development of hepatocellular carcinoma in hepatitis C virus-infected patients: in vitro study. Biochem (Mosc). 2019 Aug;84:941-53.
58. Li Z. CD133: a stem cell biomarker and beyond. Exp Hematol Oncol. 2013 Dec;2:1-8.
59. Barzegar Behrooz A, Syahir A, Ahmad S. CD133: beyond a cancer stem cell biomarker. J Drug Target. 2019 Mar 16;27(3):257-69.
60. Asadzadeh Z, Mansoori B, Mohammadi A, Kazemi T, Mokhtarzadeh A, Shanehbandi D, et al. The combination effect of Prominin1 (CD133) suppression and Oxaliplatin treatment in colorectal cancer therapy. Biomed Pharmacother. 2021 May 1;137:111364.
61. Pospieszna J, Dams-Kozlowska H, Udomsak W, Murias M, Kucinska M. Unmasking the deceptive nature of cancer stem cells: The role of cd133 in revealing their secrets. Int J Mol Sci. 2023 Jun 30;24(13):10910.
62. Ghoshal S, Fuchs BC, Tanabe KK. STAT3 is a key transcriptional regulator of cancer stem cell marker CD133 in HCC. Hepatobiliary Surg Nutr. 2016 Jun;5(3):201.
63. Tsunekuni K, Konno M, Haraguchi N, Koseki J, Asai A, Matsuoka K, et al. CD44/CD133-positive colorectal cancer stem cells are sensitive to trifluridine exposure. Sci Rep. 2019 Oct 16;9(1):14861.
64. Xu L, Lin W, Wen L, Li G. Lgr5 in cancer biology: functional identification of Lgr5 in cancer progression and potential opportunities for novel therapy. Stem Cell Res Ther. 2019 Jul 29;10(1):219.
65. Walcher L, Kistenmacher AK, Suo H, Kitte R, Dluczek S, Strauß A, et al. Cancer stem cells—origins and biomarkers: perspectives for targeted personalized therapies. Front Immunol. 2020 Aug 7;11:1280.
66. Morgan RG, Mortensson E, Williams AC. Targeting LGR5 in Colorectal Cancer: therapeutic gold or too plastic?. Br J Cancer. 2018 May 29;118(11):1410-8.
67. Liu Y, Wang Y, Sun S, Chen Z, Xiang S, Ding Z, et al. Understanding the versatile roles and applications of EpCAM in cancers: from bench to bedside. Exp Hematol Oncol. 2022 Nov 11;11(1):97.
68. Jiang X, Wang S, Liang Q, Liu Y, Liu L. Unraveling the multifaceted role of EpCAM in colorectal cancer: an integrated review of its function and interplay with non-coding RNAs. Med Oncol. 2023 Dec 27;41(1):35.
69. MacLean MR, Walker OL, Arun RP, Fernando W, Marcato P. Informed by Cancer Stem Cells of Solid Tumors: Advances in Treatments Targeting Tumor-Promoting Factors and Pathways. Int J Mol Sci. 2024 Apr 7;25(7):4102.
70. Luo T, Fang W, Lu Z, Zheng R, Yin W, Wang S, et al. Epithelial Cell Adhesion Molecule (EpCAM)-Targeted CAR-T Cells (IMC001) in Patients with Advanced Gastric Cancer: A Phase I Dose-Escalation Trial. Research Square. 2024. DOI:10.21203/rs.3.rs-4381902/v1.
71. Chen B, Liu J. Advances in ovarian tumor stem cells and therapy. Cell Biochem Biophys. 2024 Jul 3:1-22.
72. Lavudi K, Nuguri SM, Pandey P, Kokkanti RR, Wang QE. ALDH and cancer stem cells: Pathways, challenges, and future directions in targeted therapy. Life Sci. 2024 Aug 31:123033.
73. Ansari N, Gaurav K, Anand A, Singh KR, Agarwal P, Agarwal A, et al. Expression of cancer stem cell markers (CD24, CD44 & ALDH1A3 isoform) in Breast Cancer in Indian population considering clinicopathological characteristics and response to neoadjuvant chemotherapy–a prospective analysis from a university hospital. Indian J Surg Oncol. 2024 Dec;15(4):874-83.
74. Pupa SM, Ligorio F, Cancila V, Franceschini A, Tripodo C, Vernieri C, et al. HER2 signaling and breast cancer stem cells: The bridge behind HER2-positive breast cancer aggressiveness and therapy refractoriness. Cancers. 2021 Sep 24;13(19):4778.
75. Marra A, Chandarlapaty S, Modi S. Management of patients with advanced-stage HER2-positive breast cancer: current evidence and future perspectives. Nat Rev Clin Oncol. 2024 Mar;21(3):185-202.
76. Zhu K, Yang X, Tai H, Zhong X, Luo T, Zheng H. HER2-targeted therapies in cancer: a systematic review. Biomark Res. 2024 Feb 2;12(1):16.
77. Yoon J, Oh DY. HER2-targeted therapies beyond breast cancer—An update. Nat Rev Clin Oncol. 2024 Sep;21(9):675-700.
78. Saha SK, Kim K, Yang GM, Choi HY, Cho SG. Cytokeratin 19 (KRT19) has a role in the reprogramming of cancer stem cell-like cells to less aggressive and more drug-sensitive cells. Int J Mol Sci. 2018 May 9;19(5):1423.
79. Laakso M, Loman N, Borg Å, Isola J. Cytokeratin 5/14-positive breast cancer: true basal phenotype confined to BRCA1 tumors. Mod Pathol. 2005 Oct 1;18(10):1321-8.
80. McGinn O, Riley D, Finlay-Schultz J, Paul KV, Kabos P, Sartorius CA. Cytokeratins 5 and 17 maintain an aggressive epithelial state in basal-like breast cancer. Mol Cancer Res. 2022 Sep 2;20(9):1443-55.
81. Lu Q, Qu H, Lou T, Liu C, Zhang Z. CK19 promotes ovarian cancer development by impacting on Wnt/β-catenin pathway. OncoTargets Ther. 2020 Mar 24:2421-31.
82. Kawai T, Yasuchika K, Ishii T, Katayama H, Yoshitoshi EY, Ogiso S, et al. Keratin 19, a cancer stem cell marker in human hepatocellular carcinoma. Clin Cancer Res. 2015 Jul 1;21(13):3081-91.
83. Yang MH, Imrali A, Heeschen C. Circulating cancer stem cells: the importance to select. Chin J Cancer Res. 2015 Oct;27(5):437.
84. Goldenberg DM, Stein R, Sharkey RM. The emergence of trophoblast cell-surface antigen 2 (TROP-2) as a novel cancer target. Oncotarget. 2018 Jun 6;9(48):28989.
85. Zhou DD, Zhai XT, Zhang LW, Xie ZH, Wang Y, Zhen YS, et al. A new TROP2-targeting antibody-drug conjugate shows potent antitumor efficacy in breast and lung cancers. NPJ Precis Oncol. 2024 Apr 23;8(1):94.
86. Sakach E, Sacks R, Kalinsky K. Trop-2 as a Therapeutic Target in Breast Cancer. Cancers. 2022;14:5936.
87. Li LT, Jiang G, Chen Q, Zheng JN. Ki67 is a promising molecular target in the diagnosis of cancer. Mol Med Rep. 2015 Mar 1;11(3):1566-72.
88. Cidado J, Wong HY, Rosen DM, Cimino-Mathews A, Garay JP, Fessler AG, et al. Ki-67 is required for maintenance of cancer stem cells but not cell proliferation. Oncotarget. 2016 Feb 2;7(5):6281.
89. Tsang JY, Huang YH, Luo MH, Ni YB, Chan SK, Lui PC, et al. Cancer stem cell markers are associated with adverse biomarker profiles and molecular subtypes of breast cancer. Breast cancer Res Treat. 2012 Nov;136:407-17.
90. Matsika A, Srinivasan B, Day C, Mader SA, Kiernan DM, Broomfield A, et al. Cancer stem cell markers in prostate cancer: an immunohistochemical study of ALDH1, SOX2 and EZH2. Pathology. 2015 Dec 1;47(7):622-8.
91. Margaritescu C, Pirici D, Cherciu I, Barbalan A, Cârtân T, Saftoiu A. CD133/CD166/Ki-67 triple immunofluorescence assessment for putative cancer stem cells in colon carcinoma. J Gastrointest Liver Dis. 2014 Jun 1;23(2):161-70.
92. Davey MG, Hynes SO, Kerin MJ, Miller N, Lowery AJ. Ki-67 as a prognostic biomarker in invasive breast cancer. Cancers. 2021;13:4455.
93. Heming CP, Muriithi W, Macharia LW, Niemeyer Filho P, Moura-Neto V, Aran V. P-glycoprotein and cancer: what do we currently know?. Heliyon. 2022 Oct 1;8(10):e11171.
94. Tian Y, Lei Y, Wang Y, Lai J, Wang J, Xia F. Mechanism of multidrug resistance to chemotherapy mediated by P‑glycoprotein. Int J Oncol. 2023 Nov 1;63(5):1-9.
95. Nanayakkara AK, Follit CA, Chen G, Williams NS, Vogel PD, Wise JG. Targeted inhibitors of P-glycoprotein increase chemotherapeutic-induced mortality of multidrug resistant tumor cells. Sci Rep. 2018 Jan 17;8(1):967.
96. Shackleton M, Vaillant F, Simpson KJ, Stingl J, Smyth GK, Asselin-Labat ML, et al. Generation of a functional mammary gland from a single stem cell. Nature. 2006 Jan 5;439(7072):84-8.
97. Fillmore CM, Kuperwasser C. Human breast cancer cell lines contain stem-like cells that self-renew, give rise to phenotypically diverse progeny and survive chemotherapy. Breast Cancer Res. 2008 Apr;10:1-3.
98. Pece S, Tosoni D, Confalonieri S, Mazzarol G, Vecchi M, Ronzoni S, et al. Biological and molecular heterogeneity of breast cancers correlates with their cancer stem cell content. Cell. 2010 Jan 8;140(1):62-73.
99. Lu H, Clauser KR, Tam WL, Fröse J, Ye X, Eaton EN, et al. A breast cancer stem cell niche supported by juxtacrine signalling from monocytes and macrophages. Nat Cell Biol. 2014 Nov;16(11):1105-17.
100. Ricardo S, Vieira AF, Gerhard R, Leitão D, Pinto R, Cameselle-Teijeiro JF, et al. Breast cancer stem cell markers CD44, CD24 and ALDH1: expression distribution within intrinsic molecular subtype. J Clin Pathol. 2011 Nov 1;64(11):937-46.
101. Zhang X, Powell K, Li L. Breast cancer stem cells: biomarkers, identification and isolation methods, regulating mechanisms, cellular origin, and beyond. Cancers. 2020 Dec 14;12(12):3765.
102. Zhou Y, Xia L, Wang H, Oyang L, Su M, Liu Q, et al. Cancer stem cells in progression of colorectal cancer. Oncotarget. 2017 Dec 22;9(70):33403.
103. Zhao H, Han R, Wang Z, Xian J, Bai X. Colorectal cancer stem cells and targeted agents. Pharmaceutics. 2023 Dec 12;15(12):2763.
104. Omran MM, Fouda MS, Mekkawy SA, Tabll AA, Abdelaziz AG, Omran AM, et al. Molecular Biomarkers and Signaling Pathways of Cancer Stem Cells in Colorectal Cancer. Technol Cancer Res Treat. 2024 May;23:15330338241254061.
105. Soleimani A, Saeedi N, Al-Asady AM, Nazari E, Hanaie R, Khazaei M, et al. Colorectal Cancer Stem Cell Biomarkers: Biological Traits and Prognostic Insights. Curr Pharm Des. 2024 May 1;30(18):1386-97.
106. Gutova M, Najbauer J, Gevorgyan A, Metz MZ, Weng Y, Shih CC, et al. Identification of uPAR-positive chemoresistant cells in small cell lung cancer. PloS One. 2007 Feb 28;2(2):e243.
107. Jiang F, Qiu Q, Khanna A, Todd NW, Deepak J, Xing L, et al. Aldehyde dehydrogenase 1 is a tumor stem cell-associated marker in lung cancer. Mol. Cancer Res. 2009 Mar 17;7(3):330-8.
108. Yan X, Luo HU, Zhou X, Zhu B, Wang Y, Bian X. Identification of CD90 as a marker for lung cancer stem cells in A549 and H446 cell lines. Oncology Rep. 2013 Dec 1;30(6):2733-40.
109. Romeo HE, Arcos ML. Clinical relevance of stem cells in lung cancer. World J. Stem Cells. 2023 Jun 6;15(6):576.
110. Zheng Y, Wang L, Yin L, Yao Z, Tong R, Xue J, et al. Lung cancer stem cell markers as therapeutic targets: an update on signaling pathways and therapies. Front Oncol. 2022 May 26;12:873994.
111. Tachezy M, Zander H, Wolters-Eisfeld G, Müller J, Wicklein D, Gebauer F, et al. Activated leukocyte cell adhesion molecule (CD166): an “inert” cancer stem cell marker for non-small cell lung cancer?. Stem Cells. 2014 Jun 1;32(6):1429-36.
112. Taskiran C, Erdem O, Onan A, Arisoy O, Acar A, Vural C, et al. The prognostic value of endoglin (CD105) expression in ovarian carcinoma. Int J Gynecol Cancer. 2006 Sep-Oct;16(5):1789-93.
113. Wang YC, Yo YT, Lee HY, Liao YP, Chao TK, Su PH, et al. ALDH1-bright epithelial ovarian cancer cells are associated with CD44 expression, drug resistance, and poor clinical outcome. Am J Pathol. 2012 Mar 1;180(3):1159-69.
114. Huang J, Zhang J, Li H, Lu Z, Shan W, Mercado-Uribe I, et al. VCAM1 expression correlated with tumorigenesis and poor prognosis in high grade serous ovarian cancer. Am J Transl Res. 2013;5(3):336.
115. Onisim A, Iancu M, Vlad C, Kubelac P, Fetica B, Fulop A, et al. Expression of Nestin and CD133 in serous ovarian carcinoma. J Buon. 2016 Sep 1;21(5):1168-75.
116. Parte SC, Batra SK, Kakar SS. Characterization of stem cell and cancer stem cell populations in ovary and ovarian tumors. J Ovarian Res. 2018 Dec;11:1-6.
117. Klemba A, Purzycka-Olewiecka JK, Wcisło G, Czarnecka AM, Lewicki S, Lesyng B, et al. Surface markers of cancer stem-like cells of ovarian cancer and their clinical relevance. Contemp Oncol /Współczesna Onkologia. 2018 Mar 5;2018(1):48-55.
118. Kim DY, Kim H, Ko EJ, Koh SB, Kim H, Lee JY, et al. Correlation analysis of cancer stem cell marker CD133 and human endogenous retrovirus (HERV)-K env in SKOV3 ovarian cancer cells. Genes Genomics. 2024 Apr;46(4):511-8.
119. Suster NK, Virant-Klun I. Presence and role of stem cells in ovarian cancer. World J Stem Cells. 2019 Jul 7;11(7):383.
120. Masadah R, Ikram D, Riadi R, Tangdiung Y, Nelwan BJ, Ghaznawie M, et al. CD133, CD47, and PD-L1 Expression in Ovarian High-grade Serous Carcinoma and Its Association with Metastatic Disease: A Cross-sectional Study. Asian Pac J Cancer Prev.: APJCP. 2024;25(1):249.
121. Davidson B, Holth A, Dong HP. Expression of the cancer stem cell marker SSEA1 is associated with poor survival in metastatic high-grade serous carcinoma. Virchows Archiv. 2020 Nov;477(5):677-85.
122. Mazzoleni S, Politi LS, Pala M, Cominelli M, Franzin A, Sergi Sergi L, et al. Epidermal growth factor receptor expression identifies functionally and molecularly distinct tumor-initiating cells in human glioblastoma multiforme and is required for gliomagenesis. Cancer Res. 2010 Oct 1;70(19):7500-13.
123. Hale JS, Otvos B, Sinyuk M, Alvarado AG, Hitomi M, Stoltz K, et al. Cancer stem cell-specific scavenger receptor CD36 drives glioblastoma progression. Stem Cells. 2014 Jul 1;32(7):1746-58.
124. Sasmita AO, Wong YP, Ling AP. Biomarkers and therapeutic advances in glioblastoma multiforme. Asia‐Pac J Clin Oncol. 2018 Feb;14(1):40-51.
125. Hassn Mesrati M, Behrooz AB, Y. Abuhamad A, Syahir A. Understanding glioblastoma biomarkers: knocking a mountain with a hammer. Cells. 2020 May 16;9(5):1236.
126. Richardson GD, Robson CN, Lang SH, Neal DE, Maitland NJ, Collins AT. CD133, a novel marker for human prostatic epithelial stem cells. J Cell Sci. 2004 Jul 15;117(16):3539-45.
127. Darash-Yahana M, Pikarsky E, Abramovitch R, Zeira E, Pal B, Karplus R, et al. Role of high expression levels of CXCR4 in tumor growth, vascularization, and metastasis. FASEB J. 2004 Aug;18(11):1240-2.
128. Ugolkov AV, Eisengart LJ, Luan C, Yang XJ. Expression analysis of putative stem cell markers in human benign and malignant prostate. Prostate. 2011 Jan 1;71(1):18-25.
129. Deng Z, Wu Y, Ma W, Zhang S, Zhang YQ. Adoptive T-cell therapy of prostate cancer targeting the cancer stem cell antigen EpCAM. BMC Immunol. 2015 Dec;16:1-9.
130. Kerr BA, Miocinovic R, Smith AK, West XZ, Watts KE, Alzayed AW, et al. CD117+ cells in the circulation are predictive of advanced prostate cancer. Oncotarget. 2015;6(3):1889.
131. Li K, Wang Q, Tang X, Akakuru OU, Li R, Wang Y, et al. Advances in prostate cancer biomarkers and probes. Cyborg and Bionic Systems. 2024 Jun 27;5:0129.
132. Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, Wicha M, Clarke MF, Simeone DM. Identification of pancreatic cancer stem cells. Cancer Res. 2007 Feb 1;67(3):1030-7.
133. Yousefi-Ahmadipour A, Pirsadeghi A, Allahverdi A, Asadi F. Pancreatic cancer and stem cell. In: Pathak S, Banerjee A, eds. Cancer Stem Cells and Signaling Pathways. New York: Academic Press; 2024 Jan 1. pp. 363-79.
134. Rasheed ZA, Yang J, Wang Q, Kowalski J, Freed I, Murter C, et al. Prognostic significance of tumorigenic cells with mesenchymal features in pancreatic adenocarcinoma. J Natl Cancer Inst. 2010 Mar 3;102(5):340-51.
135. Matsuoka I, Kasai T, Onaga C, Ozaki A, Motomura H, Maemura Y, et al. Co‑expression of SLC20A1 and ALDH1A3 is associated with poor prognosis, and SLC20A1 is required for the survival of ALDH1‑positive pancreatic cancer stem cells. Oncology Lett. 2024 Jul 5;28(3):426.
136. Darwish NH, Sudha T, Godugu K, Elbaz O, Abdelghaffar HA, Hassan EE, et al. Acute myeloid leukemia stem cell markers in prognosis and targeted therapy: potential impact of BMI-1, TIM-3 and CLL-1. Oncotarget. 2016 Sep 9;7(36):57811.
137. Bełdzińska-Gądek K, Zarzycka E, Pastuszak K, Borman K, Lewandowski K, Zaucha JM, et al. Immune escape of B-cell lymphoblastic leukemic cells through a lineage switch to acute myeloid leukemia. Leuk Lymphoma. 2024 Jul 28;65(9):1292-302.
138. Yang Y, Wang Y, Yin C, Li X. Clinical significance of the stem cell gene Oct-4 in cervical cancer. Tumor Biol. 2014 Jun;35:5339-45.
139. Tulake W, Yuemaier R, Sheng L, Ru M, Lidifu D, Abudula A. Upregulation of stem cell markers ALDH1A1 and OCT4 as potential biomarkers for the early detection of cervical carcinoma. Oncol Lett. 2018 Nov 1;16(5):5525-34.
140. Organista‑Nava J, Gómez‑Gómez Y, Garibay‑Cerdenares OL, Leyva‑Vázquez MA, Illades‑Aguiar B. Cervical cancer stem cell‑associated genes: Prognostic implications in cervical cancer. Oncol Lett. 2019 Jul 1;18(1):7-14.
141. Fahmi MN, Hertapanndika IN, Kusuma F. The prognostic value of cancer stem cell markers in cervical cancer: a systematic review and meta-analysis. Asian Pac J Cancer Prev: APJCP. 2021 Dec;22(12):4057.
142. Almeida TA, Dos Santos OP, Saddi VA, Pereira JX, da Costa Machado H, Santos Carneiro MA, et al. Association of CD133, ALDH1, CD117 and OCT4 expression with prognosis of patients with cervical cancer. Virchows Arch. 2024 Jul 9:1-1.
143. Mendoza-Almanza G, Ortiz-Sanchez E, Rocha-Zavaleta L, Rivas-Santiago C, Esparza-Ibarra E, Olmos J. Cervical cancer stem cells and other leading factors associated with cervical cancer development. Oncol Lett. 2019 Oct;18(4):3423-32.
144. Yamashita T, Ji J, Budhu A, Forgues M, Yang W, Wang HY, et al. EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features. Gastroenterology. 2009 Mar 1;136(3):1012-24.
145. Wilson GS, Hu Z, Duan W, Tian A, Wang XM, McLeod D, et al. Efficacy of using cancer stem cell markers in isolating and characterizing liver cancer stem cells. Stem Cells Devel. 2013 Oct 1;22(19):2655-64.
146. Lathia J, Liu H, Matei D. The clinical impact of cancer stem cells. The Oncologist. 2020 Feb 1;25(2):123-31.
147. Izadpanah A, Mohammadkhani N, Masoudnia M, Ghasemzad M, Saeedian A, Mehdizadeh H, et al. Update on immune‐based therapy strategies targeting cancer stem cells. Cancer Med. 2023 Sep;12(18):18960-80.
148. Mau-Sørensen M, Dittrich C, Dienstmann R, Lassen U, Büchler W, Martinius H, et al. A phase I trial of intravenous catumaxomab: a bispecific monoclonal antibody targeting EpCAM and the T cell coreceptor CD3. Cancer Chemother Pharmacol. 2015 May;75:1065-73.
149. Knödler M, Körfer J, Kunzmann V, Trojan J, Daum S, Schenk M, et al. Randomised phase II trial to investigate catumaxomab (anti-EpCAM× anti-CD3) for treatment of peritoneal carcinomatosis in patients with gastric cancer. Br J Cancer. 2018 Aug 1;119(3):296-302.
150. Masoumi J, Jafarzadeh A, Abdolalizadeh J, Khan H, Philippe J, Mirzaei H, et al. Cancer stem cell-targeted chimeric antigen receptor (CAR)-T cell therapy: Challenges and prospects. Acta Pharm Sin B. 2021 Jul 1;11(7):1721-39.
151. Davalos V, Esteller M. Cancer epigenetics in clinical practice. CA: Cancer J Clin. 2023 Jul;73(4):376-424.
152. Parreno V, Martinez AM, Cavalli G. Mechanisms of Polycomb group protein function in cancer. Cell Res. 2022 Mar;32(3):231-53.
153. Gounder M, Schöffski P, Jones RL, Agulnik M, Cote GM, Villalobos VM, et al. Tazemetostat in advanced epithelioid sarcoma with loss of INI1/SMARCB1: an international, open-label, phase 2 basket study. Lancet Oncol. 2020 Nov 1;21(11):1423-32.
154. Czerwinska P, Mackiewicz AA. Bromodomain (BrD) Family Members as Regulators of Cancer Stemness—A Comprehensive Review. Int J Mol Sci. 2023 Jan 4;24(2):995.
155. Satheesh NJ, Samuel SM, Büsselberg D. Combination therapy with vitamin C could eradicate cancer stem cells. Biomolecules. 2020 Jan 3;10(1):79.
156. Ajmeera D, Ajumeera R. Drug repurposing: A novel strategy to target cancer stem cells and therapeutic resistance. Genes Dis. 2024 Jan 1;11(1):148-75.
157. Deshmukh A, Deshpande K, Arfuso F, Newsholme P, Dharmarajan A. Cancer stem cell metabolism: a potential target for cancer therapy. Mol Cancer. 2016 Dec;15:1-0.
158. Singh MK, Han S, Kim S, Kang I. Targeting Lipid Metabolism in Cancer Stem Cells for Anticancer Treatment. Int J Mol Sci. 2024 Oct 17;25(20):11185.
159. Saini N, Yang X. Metformin as an anti-cancer agent: actions and mechanisms targeting cancer stem cells. Acta Biochim Biophys Sin. 2018 Feb 1;50(2):133-43.
160. Maucort C, Di Giorgio A, Azoulay S, Duca M. Differentiation of Cancer Stem Cells by Using Synthetic Small Molecules: Toward New Therapeutic Strategies against Therapy Resistance. ChemMedChem. 2021 Jan 8;16(1):14-29.
161. Todd GM, Gao Z, Hyvönen M, Brazil DP, Ten Dijke P. Secreted BMP antagonists and their role in cancer and bone metastases. Bone. 2020 Aug 1;137:115455
162. Quezada SA, Peggs KS. Exploiting CTLA-4, PD-1 and PD-L1 to reactivate the host immune response against cancer. Br J Cancer. 2013 Apr;108(8):1560-5.
163. Su X, Li J, Xu X, Ye Y, Wang C, Pang G, et al. Strategies to enhance the therapeutic efficacy of anti-PD-1 antibody, anti-PD-L1 antibody and anti-CTLA-4 antibody in cancer therapy. J Transl Med. 2024 Aug 9;22(1):751.
164. Barati M, Akhondi M, Mousavi NS, Haghparast N, Ghodsi A, Baharvand H, et al. Pluripotent stem cells: cancer study, therapy, and vaccination. Stem Cell Rev Rep. 2021 Dec;17(6):1975-92.
165. Faghihkhorasani A, Dalvand A, Derafsh E, Tavakoli F, Younis NK, Yasamineh Set al. The role of oncolytic virotherapy and viral oncogenes in the cancer stem cells: a review of virus in cancer stem cells. Cancer Cell Int. 2023 Oct 25;23(1):250.
166. Abballe L, Spinello Z, Antonacci C, Coppola L, Miele E, Catanzaro G, Miele E. Nanoparticles for drug and gene delivery in pediatric brain tumors’ cancer stem cells: Current knowledge and future perspectives. Pharmaceutics. 2023 Feb;15(2):505.
167. Kharkar PS. Cancer stem cell (CSC) inhibitors in oncology—a promise for a better therapeutic outcome: state of the art and future perspectives. J Med Chem. 2020 Dec 16;63(24):15279-307
168. Prasanna T, Wu F, Yip D, Rao S. Immune modulation via epigenetic targeting to overcome immune checkpoint inhibitor resistance. Immunotherapy. 2019 Aug;11(15):1263-6.
169. Dzobo K, Senthebane DA, Ganz C, Thomford NE, Wonkam A, Dandara C. Advances in therapeutic targeting of cancer stem cells within the tumor microenvironment: an updated review. Cells. 2020 Aug 13;9(8):1896.
170. De Francesco EM, Sotgia F, Lisanti MP. Cancer stem cells (CSCs): metabolic strategies for their identification and eradication. Biochem J. 2018 May 15;475(9):1611-34.
171. Rabaan AA, AlSaihati H, Bukhamsin R, Bakhrebah MA, Nassar MS, Alsaleh AA, et al. Application of CRISPR/Cas9 technology in cancer treatment: a future direction. Curr Oncol. 2023 Feb 6;30(2):1954-76.
172. Lv S, Liu Y, Xie C, Xue C, Du S, Yao J. Emerging role of interactions between tumor angiogenesis and cancer stem cells. J Controlled Release. 2023 Aug 1;360:468-81.
173. Spokeviciute B, Kholia S, Brizzi MF. Chimeric antigen receptor (CAR) T-cell therapy: harnessing extracellular vesicles for enhanced efficacy. Pharmacol Res. 2024 Aug 13:107352.

