- 1Department of Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
- 2Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
Introduction: Cancer origin patterns significantly influence cancer phenotypes and hallmark characteristics.
Method: In the present review, four distinct carcinogenesis trajectories that contribute to malignant transformation: mutator phenotype, chromosomal instability, dysmetabolism, and stemness, are explored.
Results and Discussion: In the mutator phenotype trajectory, deficiencies in DNA repair or synthesis systems lead to hypermutation and accumulation of oncogenic alterations. The chromosomal instability trajectory involves aneuploidy-induced copy number alterations in oncogenes and tumor suppressor genes. Dysmetabolic carcinogenesis is driven by the accumulation of oncometabolites due to alterations in metabolic genes. The stemness trajectory refers to the malignant transformation of cells possessing stem-like properties under oncogenic stimuli. Each trajectory independently promotes carcinogenesis and endows cancer cells with distinct characteristics. Notably, the primary oncogenic drivers in each trajectory can self-reinforce and form spontaneous-reinforcing loops that amplify oncogenic signals. Although crosstalk exists among trajectories, evidence suggests they are mutually exclusive during cancer origin. Therefore, targeting specific carcinogenesis trajectories and disrupting the self-reinforcing oncogenic loops may represent novel therapeutic strategies. Understanding carcinogenesis trajectories provides a framework for future cancer research and treatment approaches.
1 Introduction
Cancer is a group of diseases characterized by the uncontrolled proliferation of transformed cells, which are subject to evolution by natural selection (Brown et al., 2023). Challenges in cancer management are largely attributed to the inherent complexities of cancer biology (Hanahan and Weinberg, 2011; Hanahan, 2022), as evidenced by its extensive hallmarks and intricate crosstalk. Although the hallmarks of cancer are universal features of malignant cells (Hanahan, 2022), their significance varies across different cancer types and context. For example, certain cancers exhibit hypermutations, whereas others may present somatic copy number variants. The variable significance of cancer hallmarks can be attributed to tumor heterogeneity, plasticity, and origin variations (Jassim et al., 2023). The pattern of cancer origin is particularly crucial in determining cancer phenotypes. In this article, we aim to explore and delineate several trajectories through which carcinogenesis occurs, providing a comprehensive overview of the mechanisms underlying cancer initiation. We refer to these pathways as “carcinogenesis trajectories.” This article solely focuses on the process of cancer origin, rather than cancer progression.
Typically, there are four primary carcinogenic trajectories: mutator phenotype, chromosomal instability (CIN), dysmetabolism, and stemness. The mutator phenotype trajectory posits that deficiencies in DNA repair or synthesis systems exert a mutator effect. Specifically, initially occurring mismatch repair deficiency can impair the function of other normal mismatch repair genes, forming a “mutator-mutates-other-mutators” loop. This loop progressively damages DNA repair systems and drives carcinogenesis by accelerating mutation rates and accumulating oncogenic mutations. The chromosomal instability (CIN) trajectory suggests that CIN induces aneuploidy, which in turn reduces chromosomal stability and exacerbates aneuploidy—forming a “CIN-aneuploidy-CIN” loop. This loop leads to progressive copy number alterations (CNAs) in genes; once CNAs in cancer-related genes reach the threshold for malignant transformation, cancer originates. The dysmetabolic trajectory proposes that the accumulation of oncometabolites (a consequence of alterations in metabolism-related genes) amplifies dysmetabolism, establishing a “dysmetabolism-oncometabolite-dysmetabolism” loop. These oncometabolites then promote oncogenic signaling and induce cancer origin. The stemness trajectory indicates that cells with stemness properties undergo malignant transformation in response to oncogenic stimuli, generating cancer cells that retain stemness characteristics. In the carcinogenesis process, oncogenic drivers can self-maintain and amplify, triggering an oncogenic cascade that accumulates oncogenic signals until cellular transformation occurs. Notably, the longer the carcinogenic loop persists, the more oncogenic signals are activated—and consequently, the higher the probability of malignant transformation. Therefore, targeting these carcinogenic trajectories and disrupting self-reinforcing oncogenic loops may present a potential therapeutic strategy.
2 Mutator phenotype carcinogenesis trajectory
2.1 Mutator phenotype theory
Accumulation of oncogenic mutations is a predominant mechanism underlying carcinogenesis. Various genotoxic agents, such as ionizing radiation, ultraviolet light from sunlight, and chemicals, can result in DNA damage, accumulation of mutations, and malignant transformation (Ciccia and Elledge, 2010). Under normal conditions, DNA damage repair systems maintain a low mutation rate in normal cells, which is typically insufficient to accumulate the numerous oncogenic mutations required for malignant transformation (Fox et al., 2013; Loeb, 2016). Therefore, the existence of “mutation accelerators” has been proposed to account for the carcinogenesis process (Loeb, 2010; Kentsis and Frank, 2020). In this context, the mutator phenotype theory has been postulated, suggesting the presence of a specific phenotype that contributes to the acceleration of the mutation rate and malignant transformation. In general, the mutator phenotype is characterized by deficiencies in mismatch repair genes (Bak et al., 2014; Roberts and Gordenin, 2014) and polymerase proofreading mechanisms (Albertson et al., 2009), which can lead to hypermutation (Shlien et al., 2015) and increase susceptibility to various cancers (Jeggo et al., 2016).
2.2 Mismatch repair deficiency and microsatellite instability
Mismatch repair genes, including MLH1, MSH2, MSH6, and PMS2, correct spontaneous mutations in repetitive DNA sequences (Pećina-Šlaus et al., 2020; Vilar and Gruber, 2010). The predominant consequence of a deficient mismatch repair system is microsatellite instability (MSI), which is characterized by length alterations in simple repeated microsatellite sequences and the accumulation of frameshift mutations in repeated sequences of target genes. Various mechanisms can compromise mismatch repair function and lead to MSI. First, MSI can be caused by direct germline or somatic mutations in mismatch repair genes. Second, epigenetic mechanisms can cause MSI. For example, the 3′end deletion of the epithelial cell adhesion molecule gene, which is located upstream of MSH2, can lead to the methylation of the MSH2 promoter and subsequently cause MSI (Ligtenberg et al., 2009). Epigenetic silencing of the MLH1 promoter via CpG island hypermethylation can also lead to MSI (Herman et al., 1998; Leung et al., 1999). Third, in some scenarios, the MSI phenotype can emerge without genetic or epigenetic mismatch repair gene alterations. For instance, ARID1A facilitates the recruitment of MSH2 to chromatin, enabling effective mismatch repair during DNA replication. Inactivation of ARID1A impairs the mismatch repair effect of MSH2 and can lead to an MSI phenotype (Shen et al., 2018). Moreover, trimethylation of histone H3 at Lys36 (H3K36 me3) is an epigenetic histone mark required for the mismatch repair process (Li et al., 2013). Loss of the H3K36 trimethyl-transferase SETD2 impairs the mismatch repair process and induces an MSI phenotype (Li et al., 2013). Fourth, overexpression of miR-155 or miR-21 can downregulate the expression of mismatch repair proteins, thereby inducing an MSI phenotype (Valeri et al., 2010a; Valeri et al., 2010b).
MSI occurs in various tumors (Quaas et al., 2021; Hause et al., 2016), serving as a hallmark of cancer. However, MSI also drives carcinogenesis, primarily by promoting frameshift mutations (Yamamoto and Imai, 2015; Imai and Yamamoto, 2008) that affect various target genes. These include DNA repair genes, such as MSH3 and MSH6. Mutations in these genes can further increase mutation rates, exacerbate MSI, and predispose to carcinogenesis (Malkhosyan et al., 1996). Cancer-related genes are also direct targets of frameshift mutations by MSI; the activation of oncogenes and inactivation of tumor suppressor genes confer growth and survival advantages that promote carcinogenesis (Gylfe et al., 2013; Tuupanen et al., 2014; An et al., 2015). For example, the pro-apoptotic gene BAX can be inactivated by MSI-induced frameshift mutations, contributing to oncogenesis (Woerner et al., 2005). In addition, MSI can target epigenetic modifier genes, such as histone deacetylase 2 (HDAC2) (Ropero et al., 2006) and histone methyltransferase genes (Choi et al., 2014). Notably, altered histone modifications resulting from these mutations are closely related to carcinogenesis (Calcagno et al., 2013).
2.3 Polymerase proofreading deficiency
DNA polymerases are enzymes involved in DNA synthesis. Heterozygous mutations in DNA polymerases can abrogate their ability to excise mis-incorporated non-complementary nucleotides, leading to a high frequency of single-nucleotide variant mutation rate and the development of a hypermutated phenotype (Liu et al., 2018; Goldsby et al., 2002). Mutations in DNA polymerases can increase susceptibility to various cancers (Rayner et al., 2016; Magrin et al., 2021). The DNA polymerase epsilon (POLE) gene encodes the catalytic subunit of DNA polymerase epsilon, whereas the polymerase delta 1 (POLD1) gene encodes the catalytic subunit of DNA polymerase delta (Mur et al., 2020). Notably, loss-of-function mutations in POLE and POLD1 can decrease the fidelity of DNA replication, leading to a hypermutation phenotype and ultimately promoting carcinogenesis (Venkatesan et al., 2007).
2.4 Hallmarks of mutator phenotype carcinogenesis trajectory
The mutator phenotype carcinogenesis trajectory promotes carcinogenesis through the rapid accumulation of oncogenic genetic or epigenetic alterations via MSI-induced frameshift mutations. Several hallmark characteristics define this carcinogenesis trajectory. First, this trajectory is characterized by a hypermutation phenotype, accompanied by low levels of somatic copy number alterations and chromosomal instability (Camps et al., 2006). Second, it represents an efficient pathway to cancer development. Specifically, while carcinogenesis via the chromosomal instability trajectory typically requires more than 10 years, the mismatch repair deficiency trajectory can lead to cancer within just a few years (De’ Angelis et al., 2018). Third, this trajectory exhibits a high tumor mutational burden and increased production of neoantigens, correlating with enhanced response to immunotherapy (Magrin et al., 2021; Rochefort et al., 2021).
Notably, MSI is a continuous, rather than discrete phenotype (Hause et al., 2016). Deficiency in mismatch repair genes can lead to MSI, whereas frameshift mutations in MSH3 and MSH6 can further aggravate mismatch repair deficiency and increase the level of MSI (Malkhosyan et al., 1996), creating a “mutator mutates other mutators” phenotype. This self-reinforcing “mutator–MSI–mutator” loop can progressively alter gene expression through frameshift mutations until sufficient cancer-related genes are affected, leading to cancer initiation (Figure 1).
Figure 1. Mismatch repair deficiency leads to microsatellite instability (MSI). Frameshift mutations induced by MSI impair addition mismatch repair genes, forming a self-reinforcing “mutator mutate other mutator” loop, progressively promoting hypermutations. Once a sufficient number of cancer driver genes are affected, cancer emerges.
3 Chromosomal instability carcinogenesis trajectory
3.1 Chromosomal instability and aneuploidy
Chromosomal instability (CIN) is a type of genomic instability characterized by alterations in chromosome copy number or chromosome structure. DNA damage, particularly in regions adjacent to telomeres, may lead to chromosome fusion and trigger a breakage–fusion–bridge cycle. During this process, dicentric chromosomes form chromosome bridges, which break during subsequent cell divisions, leading to CIN (Murnane, 2012; Chakravarti et al., 2021). Defects in any cellular process involved in DNA replication, chromosomal segregation, and DNA repair (such as homologous recombination deficiency) may potentially lead to CIN. CIN is commonly induced by factors such as ionizing radiation, tobacco smoke constituents (Saletta et al., 2007), viral infections (Cosper et al., 2023; Shumilov et al., 2017), and aging.
Aneuploidy, a characteristic consequence of CIN, refers to copy number alteration affecting either the entire chromosome arm (excluding the short arms of acrocentric chromosomes) or whole chromosomes (Ben-David and Amon, 2020). Aneuploidy represents a manifestation of an imbalanced karyotype (Schukken and Foijer, 2018) and typically arises from errors in chromosomal segregation in euploid cells. Various mechanisms contribute to aneuploidy, including the activation of specific oncogenes (Woo and Poon, 2004), oxidative or mechanical stress (Li and Zhu, 2022), centrosome overduplication (Vitre and Cleveland, 2012), DNA replication stress (Burrell et al., 2013), incorrect kinetochore–microtubule attachments (Li and Zhu, 2022), hypomethylation (Gaudet et al., 2003), and alterations in centromeric DNA and its binding factors (Worrall et al., 2018; Dumont et al., 2020).
3.2 Effects of aneuploidy on carcinogenesis
Aneuploidy simultaneously affects a large number of genes and can lead to dramatic alterations in gene dosage, gene expression profiles (Sdeor et al., 2024), cell biology and immune activity (Kuang and Li, 2022). It plays diverse roles in determining cell fate, and its most prominent effect is the modulation of cellular fitness. Generally, aneuploidy induces imbalanced protein production and increased proteotoxic stress, which can reduce cell fitness, disrupt normal cellular biological activity, and lead to cell death (Zhu et al., 2018; Ben-David and Amon, 2020). Notably, aneuploidy can inhibit carcinogenesis through its detrimental and lethal effects on affected cells (Hirpara et al., 2018; Williams et al., 2008). For example, aneuploidy-induced p53 activation can inhibit proliferation and induce apoptosis (Santaguida and Amon, 2015). Sometimes, aneuploidy exhibits a neutral effect because the resulting copy number alterations do not significantly change gene expression profile owing to compensatory or buffering mechanisms, or because the altered expression is neither advantageous nor detrimental for cell survival (Sdeor et al., 2024). Rarely, specific copy number alterations may confer adaptive phenotypes, promote proliferation, and increase cell fitness (Pavelka et al., 2010; Bollen et al., 2021; Ben-David et al., 2014). This survival advantage allows cells to accumulate additional copy number alterations.
Similar to CIN, aneuploidy is not only a hallmark of cancer (Taylor et al., 2018) but also a driver of cancer. However, aneuploidy alone is insufficient to initiate cancer (Kuang and Li, 2022; Sdeor et al., 2024; Vasudevan et al., 2021). The first stage of carcinogenesis involving aneuploidy requires fitness selection, where aneuploid cells that fail to successfully adapt to genomic alterations are eliminated (Sdeor et al., 2024). Notably, low levels of aneuploidy can suppress carcinogenesis (Williams et al., 2008; Stingele et al., 2012), whereas a high degree of aneuploidy promotes it (Taylor et al., 2018; Buccitelli et al., 2017; Davoli et al., 2017). This phenomenon may be explained by the fact that only aneuploid cells with sufficient fitness survive and accumulate extensive aneuploidy.
The second stage of aneuploidy-mediated carcinogenesis involves copy number alterations in cancer-related genes. In aneuploid cells, chromosomal missegregation can lead to copy number gains in oncogenes and loss of heterozygosity in tumor suppressor genes. As alterations in as few as two to eight driver genes can result in cancer (Hart et al., 2015; Alonso-Curbelo et al., 2021), once copy number alterations affect a sufficient number of cancer-related genes and reach a transformation threshold, cancer initiation occurs. For example, in glioblastoma tumor samples, a gain of chromosome 7 and loss of chromosome 10 have been observed (Brennan et al., 2013; Bredel et al., 2009); chromosome 7 carries oncogenes, typically EGFR and HGFR, while chromosome 10 harbors the tumor suppressor gene PTEN (Körber et al., 2019).
3.3 Hallmarks of chromosomal instability carcinogenesis trajectory
The chromosomal instability carcinogenesis trajectory refers to tumorigenicity driven by aneuploidy-induced fitness selection and copy number alterations in cancer-related genes (Sack et al., 2018; Davoli et al., 2013). This trajectory is characterized by high somatic copy number alterations, low mutation counts, and the involvement of p53. Functional p53 can be activated by aneuploidy (Santaguida and Amon, 2015) and induce apoptosis in aneuploid cells (Soto et al., 2017; Thompson and Compton, 2010). Given that p53 can reduce the fitness of these cells, loss-of-function mutations in this gene are favored during the CIN carcinogenesis trajectory. This hypothesis is supported by the fact that colorectal cancer cells exhibiting CIN are more likely to acquire p53 mutations at the same time (Guinney et al., 2015).
As mentioned above, CIN can cause aneuploidy, which in turn can lead to massive cellular consequences, including further increases in CIN (Chunduri and Storchová, 2019). The degree of CIN depends on the identity and number of aneuploid chromosomes (Zhu et al., 2012). For example, aberrant expression of mitotic transcription regulators induced by aneuploidy can further increase the degree of CIN in breast cancer (Pfister et al., 2018). Aneuploidy exerts complex effects on genomic stability. In certain contexts, it may confer a genome-stabilizing function (Duesberg et al., 1998; Li et al., 2009; Bökenkamp et al., 2025), whereas in other scenarios, aneuploidy itself is sufficient to induce structural instability of chromosomes (Fabarius et al., 2003; Potapova et al., 2013; Lamm et al., 2016). Aneuploidy disrupts the balance of enzyme complexes and alters the dosage of gene products encoded on aneuploid chromosomes. Specifically, dysregulation of genes responsible for DNA synthesis, maintenance, and nucleotide pool homeostasis can trigger subsequent DNA breakage, thereby eliciting chromosomal deletions, amplifications, and rearrangements (Fabarius et al., 2003; Potapova et al., 2013; Lamm et al., 2016). Notably, aneuploidy induces CIN, which in turn amplifies the degree of aneuploidy, creating a spontaneous, self-reinforcing “CIN–aneuploidy–CIN” loop that continuously drives the carcinogenesis process (Figure 2) (Potapova et al., 2013; Fabarius et al., 2003).
Figure 2. Chromosomal instability (CIN) leads to the emergence of aneuploid cells. Aneuploidy, in turn, can drive genomic instability and aggravate CIN. This establishes a self-reinforcing “CIN–aneuploidy–CIN” loop, characterized by progressively accumulating copy number alterations. Once a sufficient number of cancer-related genes are affected, cancer emerges.
4 Dysmetabolic carcinogenesis trajectory
4.1 Dysregulated metabolisms and carcinogenesis
Cell metabolism can directly control cell survival, proliferation, and differentiation, which are intricately correlated with carcinogenesis (Brunner and Finley, 2023). Dysregulated metabolism is both a hallmark of cancer and an important inducer of its development (Pavlova et al., 2022). For example, fatty acid metabolism, oxidative phosphorylation, and the citric acid cycle are enhanced in the early stages of precancerous lung squamous cell carcinoma lesions (Nie et al., 2021; Mascaux et al., 2019). These data indicate that metabolic remodeling begins early in the precancerous stages and that mutations in metabolic enzymes may act as initial drivers of carcinogenesis. Moreover, mutations in metabolic regulatory genes can lead to aberrant metabolism and predispose individuals to various tumors (Tomlinson et al., 2002; Clark et al., 2014). Environmental factors, such as hypoxia, local nutrient availability, microbiota, chronic inflammation, and diet, can also promote tumor initiation, at least partly through metabolic mechanisms (Brunner and Finley, 2023). Furthermore, cancer transformation can occur without any mutations, driven only by disruptions in cellular bioenergetics (Losman et al., 2013).
4.2 Oncometabolites in carcinogenesis
The mechanisms by which metabolic dysregulation leads to carcinogenesis are typically explained by the “oncometabolite theory.” This theory posits that the accumulation of oncometabolites, induced by mutations in mitochondrial DNA or nuclear genes encoding key metabolic enzymes, triggers oncogenic signaling cascades (Sciacovelli and Frezza, 2016). Oncometabolites are small-molecule components that accumulate due to disruptions in the tricarboxylic acid cycle (Krebs cycle). Their accumulation can drive carcinogenesis by promoting oncogenic signaling in the absence of other genetic alterations (Liu and Yang, 2021; Yong et al., 2020; Losman et al., 2013). Some studies describe oncometabolites as “drivers” of carcinogenesis, whereas other studies define oncometabolites as “products of cancer cells” (Yang et al., 2013). In this review, we discuss oncometabolites from the perspective of their role as “drivers” of carcinogenesis.
Typical oncometabolites include fumarate (Yang et al., 2012), succinate, 2-hydroxyglutarate (Du and Hu, 2021), lysophosphatidic acid (Karalis and Poulogiannis, 2024), and lactate (San-Millán et al., 2019; Raychaudhuri et al., 2019; Pandkar et al., 2023). Oncometabolite accumulation can be induced by various factors, including mutations in genes encoding key metabolic enzymes, hypoxia (Intlekofer et al., 2015; Wise et al., 2011), hyperglycemic conditions (Manuel et al., 2020; Blatnik et al., 2008), or mitochondrial dysfunction (Baysal et al., 2000). For example, fumarate hydratase (FH) is a nuclear-encoded Krebs cycle enzyme complex that mediates the reversible reaction between fumarate and malate (Pollard et al., 2005). Mutations in the gene encoding FH result in the accumulation of large amounts of cytoplasmic oncometabolite fumarate (Tomlinson et al., 2002). Hyperglycemia can also induce fumarate accumulation (Manuel et al., 2020) by impairing mitochondrial function (Nishikawa et al., 2000; Thomas et al., 2012). Fumarate accumulation can also be induced by high glucose levels in combination with metformin treatment (Pollard et al., 2005). Moreover, succinate dehydrogenase (SDH) is a Krebs cycle enzyme complex (mitochondrial enzyme) that converts succinate to fumarate to fuel the Krebs cycle. Loss-of-function mutations in SDH-encoding genes disrupt the assembly of respiratory complex II and compromise succinate oxidation, leading to the accumulation of the oncometabolite succinate (Pollard et al., 2005). Another example is isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2, respectively), which are homodimeric enzymes that convert isocitrate to α-ketoglutarate in an NADP + -dependent manner during the Krebs cycle (Xu et al., 2011). Overexpression of genes encoding IDH increases enzyme activity and leads to the accumulation of oncometabolites, including D2-hydroxyglutarate (D2-HG) (Xu et al., 2011; Ward et al., 2010; Dang et al., 2009). The accumulation of 2-HG can also be driven by hypoxia (Wise et al., 2011; Oldham et al., 2015), phosphoglycerate dehydrogenase (Fan et al., 2015), or the MYC pathway (Terunuma et al., 2014).
Oncometabolites can promote oncogenic signaling through multiple mechanisms: (1) Oncometabolites can directly promote oncogenic signaling by regulating the expression of hypoxia inducible factor (HIF) genes (Laukka et al., 2016), which in turn promote oncogenic signals and carcinogenesis (Adam et al., 2011). For example, fumarate can competitively inhibit HIF prolyl hydroxylase, preventing the proteasomal degradation of HIF (Yogev et al., 2010). Moreover, HF can induce pseudohypoxia and promote HIFs (Shanmugasundaram et al., 2014; Sudarshan et al., 2009). Succinate accumulation can lead to aberrant stabilization of HIFs, even under normoxic conditions (Selak et al., 2005). Accumulation of fumarate and 2-HG can activate the mTOR axis, which plays a pro-oncogenic role (Esparza-Moltó and Cuezva, 2018; Sourbier et al., 2014). Moreover, R-2HG accumulation can induce hypersuccinylation in the mitochondria and promote resistance to apoptosis via the BCL-2 pathway (Li et al., 2015). (2) Oncometabolites can promote a CpG island methylation phenotype (CIMP) (Whitehall et al., 2014); promoter hypermethylation associated with CIMP can inactivate tumor suppressor genes, thereby promoting carcinogenesis (Otani et al., 2013). For example, IDH1 mutations can induce CIMP through the production of oncometabolites 2-HG (Whitehall et al., 2014). Mutations in SDH can lead to CIMP by inhibiting 2-OG-dependent histone and DNA demethylases (Letouzé et al., 2013). In addition, succinate accumulation can decrease histone methylation by inhibiting histone lysine demethylases, leading to promoter hypermethylation (Xiao et al., 2012). (3) Oncometabolites can induce excessive production of reactive oxygen species (ROS) (Ishii et al., 2005), whose accumulation can cause irreversible DNA damage and protein oxidation, thereby promoting oncogenic signaling (Ziech et al., 2011; Moreno-Sánchez et al., 2013).
The metabolic-related genes mutations can independently promote carcinogenesis. For example, FH translocates from the cytosol to the nucleus upon DNA damage (Yogev et al., 2010). Mutations in FH increase cellular sensitivity to DNA damage induced by ionizing radiation (Johnson et al., 2018). Similarly, IDH mutations disrupt chromosomal topology and induce oncogene expression (Flavahan et al., 2016). Notably, these carcinogenesis mechanisms occur independently of the effects of oncometabolites.
4.3 Hallmarks of the dysmetabolic carcinogenesis trajectory
The dysmetabolic carcinogenesis trajectory refers to the aberrant accumulation of oncometabolites resulting from genetic or epigenetic alterations in metabolism-related genes, which promote oncogenic signaling and carcinogenesis. Oncometabolites arising from dysregulated metabolism can further disrupt metabolic processes and promote oncometabolite production (Li et al., 2015), forming a spontaneous, self-reinforcing “dysmetabolism–oncometabolites–dysmetabolism” loop that drives carcinogenesis (Figure 3).
Figure 3. Mutations in metabolism-related genes or mitochondrial dysfunction lead to accumulation of oncometabolites. This accumulation further dysregulates metabolism, forming a self-reinforcing loop that promotes oncogenic signaling and carcinogenesis.
5 Stemness carcinogenesis trajectory
The cancer stem cell theory suggests that cancer originates from normal tissue stem cells or stem-like cells. Notably, cells with stem and progenitor characteristics in normal tissues are ideal candidates for transformation owing to their long lifespan, which enables the accumulation of adequate oncogenic mutations (Smalley and Ashworth, 2003), inherent self-renewal and proliferation abilities (Dontu et al., 2003), multidirectional differentiation ability, and access to stemness-maintaining signals from their niches. Malignant transformation has been observed in various stem cells, including neural (Wang et al., 2009), mammary basal (Molyneux et al., 2010), hepatic (Chiba et al., 2010), thyroid (Nagayama et al., 2016), prostate (Losada-García et al., 2023), and hematopoietic stem cells (Shlush et al., 2014). In this section, we briefly review the emergence of stem-like cells and focus on stem cell transformation.
5.1 Cells with stemness identity
The stem cell population represents not only distinct cell types but also a plastic phenotype (Thankamony et al., 2020; Raz and Yamashita, 2025). Non-stem cells can acquire stem cell-like properties under certain conditions. First, stem cells depend on their microenvironments, gaining or losing their stemness identity as they enter or exit the stem cell niche (Gehart and Clevers, 2019). For example, the transcription factor Achaete-Scute Homolog 2 can lead to the dedifferentiation of crypt cells, enabling them to acquire stem cell status in the absence of Leucine-Rich Repeat-Containing G-Protein Coupled Receptor 5 (LGR5+) stem cells (Murata et al., 2020). Second, the stemness of normal tissue stem cells is maintained by developmental signaling pathways. Aberrant activation of developmental signals through genetic or epigenetic alterations can induce non-stem cells to adopt a stem-like state. For instance, the activation of NF-κΒ can stimulate WNT signaling, leading to intestinal villus cell dedifferentiation and acquisition of stem cell properties (Schwitalla et al., 2013). Third, genetic epigenetic alterations affecting differentiation-related processes also contribute to stemness acquisition. For example, the Sterile Alpha Motif Domain (SMAD4) is a transcription factor that promotes differentiation. The loss of SMAD4, combined with the activation of the WNT pathway, impairs normal differentiation and drives intestinal epithelial cells toward a stem cell-like phenotype (Perekatt et al., 2018). Therefore, in the absence of pre-existing stem cells, differentiated cells can acquire stemness identity and become potential targets for malignant transformation (Bajaj et al., 2020).
5.2 Stem cell transformation
The initial level of stemness, together with secondary oncogenic drivers, determines the likelihood and outcome of cellular transformation. Stemness level influences the transformation potential; generally, undifferentiated stem cells are more responsive to oncogenic mutations than their differentiated counterparts (Lytle et al., 2018). Moreover, oncogenic drivers can transform undifferentiated cells but not differentiated cells. For instance, the activation of Sox2 and Stat3 can promote the malignant transformation of undifferentiated esophageal basal cells but not differentiated suprabasal cells (Liu et al., 2013). In response to Ras and p53 alterations, hair follicle stem cells can transform into squamous carcinoma cells, whereas their progeny, the transit-amplifying cells, cannot form benign tumors under the same genetic conditions (White et al., 2011; Lapouge et al., 2011). Melanoblasts, the precursors of melanocytes, are responsive to oncogenic mutations and undergo malignant transformation more readily than differentiated melanocytes (Baggiolini et al., 2021). Similarly, APC deletion leads to transformation in intestinal stem cells but not in short-lived transit-amplifying cells (Barker et al., 2009). Moreover, the level of stemness contributes to the transformed cancer types. For example, BCR–ABL translocation in hematopoietic stem cells leads to chronic myelogenous leukemia, whereas in progenitor cells, it leads to acute lymphoid leukemia (Li et al., 1999; Neering et al., 2007). Loss-of-function mutations in the tumor suppressor genes P53, NF1, and PTEN of neural stem cells, neural progenitors, and oligodendrocyte progenitors induce distinct subtypes of glioblastoma (Alcantara Llaguno et al., 2015). Secondary oncogenic drivers also contribute to malignant transformation and influence the resulting cancer type. For example, activation of hedgehog signaling in either neuronal stem or granule precursor cells can give rise to medulloblastomas with similar molecular phenotypes (Schüller et al., 2008; Yang et al., 2008). Finally, whether a specific signaling acts as a stemness maintainer or a secondary oncogenic driver depends on the cellular context. For example, although Sonic hedgehog signaling generally plays a role in maintaining stemness, its activation in neuronal progenitors leads to medulloblastoma formation (Yang et al., 2008), acting as a secondary oncogenic driver in carcinogenesis.
5.3 Hallmarks of stemness carcinogenesis trajectory
The stemness carcinogenesis trajectory refers to the malignant transformation of stem-like cells under secondary oncogenic stimuli. This carcinogenesis trajectory is characterized by a stemness phenotype and high degree of aggressiveness. For example, medulloblastoma originating from stem cells progresses more rapidly than that arising from progenitor cells (Yang et al., 2008). This heightened malignancy may be attributed to the emergence of cancer stem cells (Bajaj et al., 2020; Smith et al., 2015; Zhang et al., 2008), which are associated with disease progression, metastasis, recurrence, and resistance to treatment (Chu et al., 2024). The stemness trajectory is closely associated with, and may be considered as a shortcut of, the chromosomal instability trajectory (Figure 4).
Figure 4. Normal cells can acquire chromosomal instability (CIN) through radiation, chemical carcinogens, or viruses. Most cells exhibiting CIN ultimately die, as CIN is generally disadvantageous to survival. However, a small subset of cells may acquire survival and proliferative advantages through the generation of aneuploidy; this explains the latent period in the carcinogenesis process. Cells that gain such survival and proliferative advantages may undergo malignant transformation upon acquiring even minimal additional oncogenic signals. Stem cells, which inherently possess a long lifespan and intrinsic survival advantages, may undergo malignant transformation under oncogenic stimuli.
6 Crosstalk among carcinogenesis trajectories
Intracellular signaling pathways exhibit extensive crosstalk, allowing a single oncogenic signal to initiate a cascade that activates a series of downstream oncogenic signals. While cancer may initially arise from a specific carcinogenesis trajectory, it can later involve mechanisms from other trajectories as the disease progresses (Figure 5). Trajectory shifts can occur during carcinogenesis, influenced by fluctuations in the underlying driving forces.
Figure 5. Carcinogenesis trajectories have extensive crosstalk and promote each other via distinctive mechanisms.
Recently, FH was demonstrated to function as a DNA repair factor in the non-homologous end-joining (NHEJ) process. Mutations in FH can impair the NHEJ process, potentially leading to CIN, thereby establishing a link between metabolic dysregulation and CIN. Furthermore, the accumulation of the oncometabolite fumarate can induce epithelial-mesenchymal transition, which is associated with the activation of stemness signaling (Sciacovelli et al., 2016). Oncometabolites can also activate the HIF-1α/Notch pathway, which is involved in cellular dedifferentiation and stemness maintenance (Xiang and Semenza, 2019; Mu et al., 2021).
Theoretically, in the chromosomal instability carcinogenesis trajectory, aneuploidy can disrupt mismatch repair systems through loss of heterozygosity, leading to MSI. Aneuploidy may also affect metabolism-related genes, leading to oncometabolite accumulation. For example, aneuploidy involving chromosome 21 can alter the dosage of genes encoding the DNA methyltransferase DNMT3L, causing hypermethylation or hypomethylation at CpG sites (Muskens et al., 2021; Lu et al., 2016). Aberrant methylation of CpG islands in gene promoters can inactivate tumor suppressor genes, thereby promoting carcinogenesis (Otani et al., 2013). CIN can also promote stemness by activating pro-tumorigenic antiviral signaling (Baba et al., 2024). Aneuploidy, such as the loss of chromosome arm 8p, can alter lipid metabolism and lead to dysmetabolism (Cai et al., 2016).
Frameshift mutations induced by MSI can affect genes responsible for chromosomal integrity (Koh et al., 2005). Both hRAD50 and hMRE11 are involved in the NHEJ repair process, and MSI-induced frameshift mutations in these genes contribute to CIN (Koh et al., 2005). Moreover, MSI-related frameshift mutations can involve non-coding regions, resulting in aberrant production of microRNAs (miRNAs). These dysregulated miRNA may further promote copy number amplifications or deletions.
In the stemness carcinogenesis trajectory, cells with stem-like properties are also susceptible CIN, leading to an aneuploidy phenotype (Gao et al., 2016).
7 The carcinogenesis trajectories and cancer origin theory
Various models have been proposed to explain the origin of cancer (Jassim et al., 2023). Theories of cancer origin typically include the somatic mutation (Bailey et al., 2018), stem cell (Tu et al., 2021), mutator phenotype (Loeb, 2001), atavistic model (Lineweaver et al., 2021; Bussey and Davies, 2021), and bioenergetic theory (Gyamfi et al., 2022; Brunner and Finley, 2023; Martínez-Reyes and Chandel, 2021; Pavlova et al., 2022). Each theory successfully aligns with certain scenarios but fails to explain oncogenesis in others. For instance, cancer stem cell theory proposes that cancer-initiating cells are derived from normal stem or progenitor cells (Lytle et al., 2018). However, cancer stem cells (CSCs) have yet to be identified in many types of cancers (Ishizawa et al., 2010). Furthermore, the prevailing view today is that cancer has a genetic component. Therefore, the accumulation of genetic mutations and epigenetic alterations, which regulate cell replication, cell division, cell metabolism, and cell growth, drive oncogenesis (Hanahan, 2022). However, there are reports that many carcinogens do not damage DNA (Riva et al., 2020), and simple oncogenic mutations are not sufficient for initiating tumorigenesis without additional regulatory factors, such as the microenvironment (Yuan et al., 2024). Emerging evidence indicates the regulatory roles of dysregulated metabolism in carcinogenesis (Gyamfi et al., 2022; Kong et al., 2024). The dysregulation of mitochondria or oncometabolites has been proposed to drive oncogenesis. However, in some cancers, only genetic abnormalities are found, without abnormal mitochondrial function (Kiebish and Seyfried, 2005; Seyfried, 2015). The atavistic theory suggests that cancer onset and progression represent a type of reversion from a multicellular to a unicellular phenotype (Lineweaver et al., 2021; Bussey and Davies, 2021). During the evolution of multicellular organisms, genetic programs inherited from the unicellular stage—such as those regulating “unlimited proliferation” and “autonomous migration”—are silenced.
In this context, the proposal of an integrated and rational theory on the origin of cancer is crucial. Here, we propose that carcinogenesis trajectories suggest that cancer may have multiple origins. Each trajectory can independently contribute to carcinogenesis; additionally, they may crosstalk and collectively promote the activation and accumulation of oncogenic signals. For instance, the mutator phenotype trajectory is aligned with somatic mutation, both of which focus on “mutations”. The dysmetabolic trajectory align with the bioenergetic theory, both of which emphasize the oncogenic effects of metabolic alterations. The stemness trajectory is aligned with cancer stem cells theory of cancer origin. Another interesting finding is that unicellular organisms lack the complex DNA repair mechanisms and cell cycle checkpoints found in multicellular organisms. They have a higher probability of chromosomal replication errors, fragment recombination, or loss, resulting in an inherently high rate of chromosomal variation. This is not a “defect” but a necessary adaptation for survival, as it enables them to rapidly generate genetic diversity to adapt to adverse environments. Based on this view, the CIN trajectory is partially aligned with atavistic theory.
8 Spontaneous-reinforcing oncogenic loop
The activation of oncogenes and the inactivation of tumor suppressor genes do not always lead to cancer, as pro-tumorigenic signals may be neutralized or buffered by redundant cellular signaling pathways. Therefore, the oncogenesis process depends on the cellular context. Based on the carcinogenesis trajectories discussed above, a notable finding emerges-for malignant transformation to happen, the initial oncogenic driver itself needs to self-reinforce, thereby promoting the progression of oncogenic events. For example, the degree of aneuploidy can spontaneously worsen owing to genomic instability induced by the aneuploidy itself (Burrell et al., 2013; Lamm et al., 2016; Ly et al., 2019; Garribba and Santaguida, 2022; Nicholson and Cimini, 2013). Oncometabolites generated through dysmetabolism can disrupt mitochondrial function, exacerbating metabolic dysfunction (Li et al., 2015). Moreover, the mutator phenotype resulting from mismatch repair deficiency can impair the function of other normal mismatch repair genes (Yamamoto and Imai, 2015), progressively accelerating the mutation rate. These findings suggest that the primary drivers of carcinogenesis should be specific oncogenic events which are capable of self-reinforcing, maintaining, and amplifying their effects. The formation of a spontaneous, self-reinforcing oncogenic loop may be necessary for the initiation of cancer.
9 Therapeutic significance of carcinogenesis trajectory theory
The origin of a tumor underlies a distinct tumorigenic pathway that directly affects its phenotype. A typical example is cancers originating from mismatch repair deficiency, which exhibit an MSI phenotype regardless of whether their origin is hereditary or sporadic (Imai and Yamamoto, 2008). Cancers of distinct origins generally respond differently to different treatments; therefore, targeting the carcinogenesis trajectory and halting the spontaneous-reinforcing oncogenic loop may be a potential treatment strategy. Various drugs have been designed and implemented in clinical practice to treat cancers by targeting specific hallmarks. One such hallmark is the production of oncometabolites, which occupy a central role in the “dysmetabolism–oncometabolites–dysmetabolism” spontaneous-reinforcing oncogenic loop. Therapeutic strategies have been designed to target the production of these oncometabolites. For example, the accumulation of the oncometabolite 2HD, which is caused by IDH mutations, can lead to oncogenesis (Gunn et al., 2023). A small-molecule inhibitor of mutant IDH1, which reduces the production of 2-HD, has been used to treat IDH-mutated acute myeloid leukemia (Mellinghoff et al., 2020) and cholangiocarcinoma (Abou-Alfa et al., 2020). The second hallmark involves the survival advantage of cells with stemness characteristics, which is essential to cancers originating from the stemness carcinogenesis trajectory. Various strategies have been designed to target cancer stemness. For example, all-trans retinoic acid has been designed to target stemness-related genes, such as ALDH, SOX2, and KLF4, to target cancer stem cells in gastric cancer (Nguyen et al., 2016). Tranylcypromine analogs have been used to induce cancer stem cell differentiation in the treatment of acute leukemia (Harris et al., 2012). A third targeted hallmark is aneuploidy, which can induce genomic instability and further exacerbate aneuploidy by itself. Therapeutic strategies targeting aneuploid cells have been designed for cancer treatment (Bhatia et al., 2024; Sdeor et al., 2024; Rendo et al., 2025). Another hallmark is DNA methylation, which can be caused by dysmetabolism. DNA methyltransferase inhibitors can reverse DNA hypermethylation, and hypomethylation at CpG regions enables the re-expression of silenced tumor suppressor genes in cancer cells (Lee et al., 2024). We hypothesize that the anti-tumor effects of these drugs may be attributed, at least in part, to their disruption of spontaneous-reinforcing oncogenic loops.
10 Conclusions and future perspectives
The origin of cancer could be fundamentally attributed to the aberrant activation of oncogenic signals. These pro-tumorigenic signals engaged in an extremely complex crosstalk, unlocking the plasticity of cancer cells and complicating cancer treatment. Although oncogenic signaling pathways often interact to varying degrees, there is still a predominant pathway by which cancer originates and acquires cancer-specific phenotypes and hallmarks; this pathway is defined as “the carcinogenesis trajectory”. In this article, we reviewed several typical carcinogenesis trajectories, including genomic instability trajectories (CIN and MSI), dysmetabolism-driven carcinogenesis trajectory, and stemness-related carcinogenesis trajectory, and discussed the crosstalk between them. The carcinogenesis trajectory is a dynamic process as well as a cellular state in which a specific phenotype can become self-sustaining and self-amplifying. Notably, a spontaneous-reinforcing carcinogenesis loop can be observed in each trajectory. This “carcinogenesis loop” possesses a self-reinforcing ability that progressively maintains and amplifies oncogenic signals, ultimately driving the emergence of cancer. Therefore, targeting the carcinogenesis trajectory and halting the carcinogenesis loop could serve as a potential strategy for cancer prevention and treatment.
Data availability statement
The original contributions presented in the study are included in the article/supplementary material, further inquiries can be directed to the corresponding author.
Author contributions
RW: Funding acquisition, Software, Writing – original draft. ZY: Conceptualization, Funding acquisition, Supervision, Writing – original draft, Writing – review and editing.
Funding
The authors declare that financial support was received for the research and/or publication of this article. This study was supported by the 345 Talent Project of Shengjing Hospital of China Medical University (Grant Number M1437 to ZY; Grant Number M1418 to RW), High-Quality Development Projects of China Medical University supported by Department of Science and Technology of Liaoning Province (Grant Number 2023JH2/20200151 to ZY) and Natural Science Foundation General Program of Department of Science and Technology of Liaoning Province (Grant Number 2025-MS-216 to RW).
Acknowledgements
We would like to thank Editage (www.editage.com) for the English language editing.
Conflict of interest
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.
Generative AI statement
The authors declare that no Generative AI was used in the creation of this manuscript.
Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
Abou-Alfa, G. K., Macarulla, T., Javle, M. M., Kelley, R. K., Lubner, S. J., Adeva, J., et al. (2020). Ivosidenib in IDH1-mutant, chemotherapy-refractory cholangiocarcinoma (ClarIDHy): a multicentre, randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 21, 796–807. doi:10.1016/S1470-2045(20)30157-1
Adam, J., Hatipoglu, E., O’Flaherty, L., Ternette, N., Sahgal, N., Lockstone, H., et al. (2011). Renal cyst formation in Fh1-deficient mice is independent of the Hif/Phd pathway: roles for fumarate in KEAP1 succination and Nrf2 signaling. Cancer Cell 20, 524–537. doi:10.1016/j.ccr.2011.09.006
Albertson, T. M., Ogawa, M., Bugni, J. M., Hays, L. E., Chen, Y., Wang, Y., et al. (2009). DNA polymerase epsilon and delta proofreading suppress discrete mutator and cancer phenotypes in mice. Proc. Natl. Acad. Sci. U. S. A. 106, 17101–17104. doi:10.1073/pnas.0907147106
Alcantara Llaguno, S. R., Wang, Z., Sun, D., Chen, J., Xu, J., Kim, E., et al. (2015). Adult lineage-restricted CNS progenitors specify distinct glioblastoma subtypes. Cancer Cell 28, 429–440. doi:10.1016/j.ccell.2015.09.007
Alonso-Curbelo, D., Ho, Y.-J., Burdziak, C., Maag, J. L. V., Morris, J. P., Chandwani, R., et al. (2021). A gene-environment-induced epigenetic program initiates tumorigenesis. Nature 590, 642–648. doi:10.1038/s41586-020-03147-x
An, C. H., Je, E. M., Yoo, N. J., and Lee, S. H. (2015). Frameshift mutations of cadherin genes DCHS2, CDH10 and CDH24 genes in gastric and colorectal cancers with high microsatellite instability. Pathol. Oncol. Res. 21, 181–185. doi:10.1007/s12253-014-9804-8
Baba, S. A., Zakeri, A., and Desgrosellier, J. S. (2024). Chromosomal instability as an architect of the cancer stemness landscape. Front. Cell Dev. Biol. 12, 1450614. doi:10.3389/fcell.2024.1450614
Baggiolini, A., Callahan, S. J., Montal, E., Weiss, J. M., Trieu, T., Tagore, M. M., et al. (2021). Developmental chromatin programs determine oncogenic competence in melanoma. Science 373, eabc1048. doi:10.1126/science.abc1048
Bailey, M. H., Tokheim, C., Porta-Pardo, E., Sengupta, S., Bertrand, D., Weerasinghe, A., et al. (2018). Comprehensive characterization of cancer driver genes and mutations. Cell 173, 371–385.e18. doi:10.1016/j.cell.2018.02.060
Bajaj, J., Diaz, E., and Reya, T. (2020). Stem cells in cancer initiation and progression. J. Cell Biol. 219, e201911053. doi:10.1083/jcb.201911053
Bak, S. T., Sakellariou, D., and Pena-Diaz, J. (2014). The dual nature of mismatch repair as antimutator and mutator: for better or for worse. Front. Genet. 5, 287. doi:10.3389/fgene.2014.00287
Barker, N., Ridgway, R. A., van Es, J. H., van de Wetering, M., Begthel, H., van den Born, M., et al. (2009). Crypt stem cells as the cells-of-origin of intestinal cancer. Nature 457, 608–611. doi:10.1038/nature07602
Baysal, B. E., Ferrell, R. E., Willett-Brozick, J. E., Lawrence, E. C., Myssiorek, D., Bosch, A., et al. (2000). Mutations in SDHD, a mitochondrial complex II gene, in hereditary paraganglioma. Science 287, 848–851. doi:10.1126/science.287.5454.848
Ben-David, U., and Amon, A. (2020). Context is everything: aneuploidy in cancer. Nat. Rev. Genet. 21, 44–62. doi:10.1038/s41576-019-0171-x
Ben-David, U., Arad, G., Weissbein, U., Mandefro, B., Maimon, A., Golan-Lev, T., et al. (2014). Aneuploidy induces profound changes in gene expression, proliferation and tumorigenicity of human pluripotent stem cells. Nat. Commun. 5, 4825. doi:10.1038/ncomms5825
Bhatia, S., Khanna, K. K., and Duijf, P. H. G. (2024). Targeting chromosomal instability and aneuploidy in cancer. Trends Pharmacol. Sci. 45, 210–224. doi:10.1016/j.tips.2024.01.009
Blatnik, M., Thorpe, S. R., and Baynes, J. W. (2008). Succination of proteins by fumarate: mechanism of inactivation of glyceraldehyde-3-phosphate dehydrogenase in diabetes. Ann. N. Y. Acad. Sci. 1126, 272–275. doi:10.1196/annals.1433.047
Bökenkamp, J.-E., Keuper, K., Redel, S., Barthel, K., Johnson, L., Becker, A., et al. (2025). Proteogenomic analysis reveals adaptive strategies for alleviating the consequences of aneuploidy in cancer. EMBO J. 44, 1829–1865. doi:10.1038/s44318-025-00372-w
Bollen, Y., Stelloo, E., van Leenen, P., van den Bos, M., Ponsioen, B., Lu, B., et al. (2021). Reconstructing single-cell karyotype alterations in colorectal cancer identifies punctuated and gradual diversification patterns. Nat. Genet. 53, 1187–1195. doi:10.1038/s41588-021-00891-2
Bredel, M., Scholtens, D. M., Harsh, G. R., Bredel, C., Chandler, J. P., Renfrow, J. J., et al. (2009). A network model of a cooperative genetic landscape in brain tumors. JAMA 302, 261–275. doi:10.1001/jama.2009.997
Brennan, C. W., Verhaak, R. G. W., McKenna, A., Campos, B., Noushmehr, H., Salama, S. R., et al. (2013). The somatic genomic landscape of glioblastoma. Cell 155, 462–477. doi:10.1016/j.cell.2013.09.034
Brown, J. S., Amend, S. R., Austin, R. H., Gatenby, R. A., Hammarlund, E. U., and Pienta, K. J. (2023). Updating the definition of cancer. Mol. Cancer Res. 21, 1142–1147. doi:10.1158/1541-7786.MCR-23-0411
Brunner, J. S., and Finley, L. W. S. (2023). Metabolic determinants of tumour initiation. Nat. Rev. Endocrinol. 19, 134–150. doi:10.1038/s41574-022-00773-5
Buccitelli, C., Salgueiro, L., Rowald, K., Sotillo, R., Mardin, B. R., and Korbel, J. O. (2017). Pan-cancer analysis distinguishes transcriptional changes of aneuploidy from proliferation. Genome Res. 27, 501–511. doi:10.1101/gr.212225.116
Burrell, R. A., McClelland, S. E., Endesfelder, D., Groth, P., Weller, M.-C., Shaikh, N., et al. (2013). Replication stress links structural and numerical cancer chromosomal instability. Nature 494, 492–496. doi:10.1038/nature11935
Bussey, K. J., and Davies, P. C. W. (2021). Reverting to single-cell biology: the predictions of the atavism theory of cancer. Prog. Biophys. Mol. Biol. 165, 49–55. doi:10.1016/j.pbiomolbio.2021.08.002
Cai, Y., Crowther, J., Pastor, T., Abbasi Asbagh, L., Baietti, M. F., De Troyer, M., et al. (2016). Loss of chromosome 8p governs tumor progression and drug response by altering lipid metabolism. Cancer Cell 29, 751–766. doi:10.1016/j.ccell.2016.04.003
Calcagno, D. Q., Gigek, C. O., Chen, E. S., Burbano, R. R., Smith, M., and de, A. C. (2013). DNA and histone methylation in gastric carcinogenesis. World J. Gastroenterol. 19, 1182–1192. doi:10.3748/wjg.v19.i8.1182
Camps, J., Armengol, G., del Rey, J., Lozano, J. J., Vauhkonen, H., Prat, E., et al. (2006). Genome-wide differences between microsatellite stable and unstable colorectal tumors. Carcinogenesis 27, 419–428. doi:10.1093/carcin/bgi244
Chakravarti, D., LaBella, K. A., and DePinho, R. A. (2021). Telomeres: history, health, and hallmarks of aging. Cell 184, 306–322. doi:10.1016/j.cell.2020.12.028
Chiba, T., Seki, A., Aoki, R., Ichikawa, H., Negishi, M., Miyagi, S., et al. (2010). Bmi1 promotes hepatic stem cell expansion and tumorigenicity in both Ink4a/Arf-dependent and -independent manners in mice. Hepatology 52, 1111–1123. doi:10.1002/hep.23793
Choi, Y. J., Oh, H. R., Choi, M. R., Gwak, M., An, C. H., Chung, Y. J., et al. (2014). Frameshift mutation of a histone methylation-related gene SETD1B and its regional heterogeneity in gastric and colorectal cancers with high microsatellite instability. Hum. Pathol. 45, 1674–1681. doi:10.1016/j.humpath.2014.04.013
Chu, X., Tian, W., Ning, J., Xiao, G., Zhou, Y., Wang, Z., et al. (2024). Cancer stem cells: advances in knowledge and implications for cancer therapy. Signal Transduct. Target Ther. 9, 170. doi:10.1038/s41392-024-01851-y
Chunduri, N. K., and Storchová, Z. (2019). The diverse consequences of aneuploidy. Nat. Cell Biol. 21, 54–62. doi:10.1038/s41556-018-0243-8
Ciccia, A., and Elledge, S. J. (2010). The DNA damage response: making it safe to play with knives. Mol. Cell 40, 179–204. doi:10.1016/j.molcel.2010.09.019
Clark, G. R., Sciacovelli, M., Gaude, E., Walsh, D. M., Kirby, G., Simpson, M. A., et al. (2014). Germline FH mutations presenting with pheochromocytoma. J. Clin. Endocrinol. Metab. 99, E2046–E2050. doi:10.1210/jc.2014-1659
Cosper, P. F., Hrycyniak, L. C. F., Paracha, M., Lee, D. L., Wan, J., Jones, K., et al. (2023). HPV16 E6 induces chromosomal instability due to polar chromosomes caused by E6AP-dependent degradation of the mitotic kinesin CENP-E. Proc. Natl. Acad. Sci. U. S. A. 120, e2216700120. doi:10.1073/pnas.2216700120
Dang, L., White, D. W., Gross, S., Bennett, B. D., Bittinger, M. A., Driggers, E. M., et al. (2009). Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 462, 739–744. doi:10.1038/nature08617
Davoli, T., Xu, A. W., Mengwasser, K. E., Sack, L. M., Yoon, J. C., Park, P. J., et al. (2013). Cumulative haploinsufficiency and triplosensitivity drive aneuploidy patterns and shape the cancer genome. Cell 155, 948–962. doi:10.1016/j.cell.2013.10.011
Davoli, T., Uno, H., Wooten, E. C., and Elledge, S. J. (2017). Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 355, eaaf8399. doi:10.1126/science.aaf8399
De’ Angelis, G. L., Bottarelli, L., Azzoni, C., De’ Angelis, N., Leandro, G., Di Mario, F., et al. (2018). Microsatellite instability in colorectal cancer. Acta Biomed. 89, 97–101. doi:10.23750/abm.v89i9-S.7960
Dontu, G., Al-Hajj, M., Abdallah, W. M., Clarke, M. F., and Wicha, M. S. (2003). Stem cells in normal breast development and breast cancer. Cell Prolif. 36 (Suppl. 1), 59–72. doi:10.1046/j.1365-2184.36.s.1.6.x
Du, X., and Hu, H. (2021). The roles of 2-Hydroxyglutarate. Front. Cell Dev. Biol. 9, 651317. doi:10.3389/fcell.2021.651317
Duesberg, P., Rausch, C., Rasnick, D., and Hehlmann, R. (1998). Genetic instability of cancer cells is proportional to their degree of aneuploidy. Proc. Natl. Acad. Sci. U. S. A. 95, 13692–13697. doi:10.1073/pnas.95.23.13692
Dumont, M., Gamba, R., Gestraud, P., Klaasen, S., Worrall, J. T., De Vries, S. G., et al. (2020). Human chromosome-specific aneuploidy is influenced by DNA-dependent centromeric features. EMBO J. 39, e102924. doi:10.15252/embj.2019102924
Esparza-Moltó, P. B., and Cuezva, J. M. (2018). The role of mitochondrial H+-ATP synthase in cancer. Front. Oncol. 8, 53. doi:10.3389/fonc.2018.00053
Fabarius, A., Hehlmann, R., and Duesberg, P. H. (2003). Instability of chromosome structure in cancer cells increases exponentially with degrees of aneuploidy. Cancer Genet. Cytogenet 143, 59–72. doi:10.1016/s0165-4608(03)00003-7
Fan, J., Teng, X., Liu, L., Mattaini, K. R., Looper, R. E., Vander Heiden, M. G., et al. (2015). Human phosphoglycerate dehydrogenase produces the oncometabolite D-2-hydroxyglutarate. ACS Chem. Biol. 10, 510–516. doi:10.1021/cb500683c
Flavahan, W. A., Drier, Y., Liau, B. B., Gillespie, S. M., Venteicher, A. S., Stemmer-Rachamimov, A. O., et al. (2016). Insulator dysfunction and oncogene activation in IDH mutant gliomas. Nature 529, 110–114. doi:10.1038/nature16490
Fox, E. J., Prindle, M. J., and Loeb, L. A. (2013). Do mutator mutations fuel tumorigenesis? Cancer Metastasis Rev. 32, 353–361. doi:10.1007/s10555-013-9426-8
Gao, R., Davis, A., McDonald, T. O., Sei, E., Shi, X., Wang, Y., et al. (2016). Punctuated copy number evolution and clonal stasis in triple-negative breast cancer. Nat. Genet. 48, 1119–1130. doi:10.1038/ng.3641
Garribba, L., and Santaguida, S. (2022). The dynamic instability of the aneuploid genome. Front. Cell Dev. Biol. 10, 838928. doi:10.3389/fcell.2022.838928
Gaudet, F., Hodgson, J. G., Eden, A., Jackson-Grusby, L., Dausman, J., Gray, J. W., et al. (2003). Induction of tumors in mice by genomic hypomethylation. Science 300, 489–492. doi:10.1126/science.1083558
Gehart, H., and Clevers, H. (2019). Tales from the crypt: new insights into intestinal stem cells. Nat. Rev. Gastroenterol. Hepatol. 16, 19–34. doi:10.1038/s41575-018-0081-y
Goldsby, R. E., Hays, L. E., Chen, X., Olmsted, E. A., Slayton, W. B., Spangrude, G. J., et al. (2002). High incidence of epithelial cancers in mice deficient for DNA polymerase Delta proofreading. Proc. Natl. Acad. Sci. U. S. A. 99, 15560–15565. doi:10.1073/pnas.232340999
Guinney, J., Dienstmann, R., Wang, X., de Reyniès, A., Schlicker, A., Soneson, C., et al. (2015). The consensus molecular subtypes of colorectal cancer. Nat. Med. 21, 1350–1356. doi:10.1038/nm.3967
Gunn, K., Myllykoski, M., Cao, J. Z., Ahmed, M., Huang, B., Rouaisnel, B., et al. (2023). (R)-2-Hydroxyglutarate inhibits KDM5 histone lysine demethylases to drive transformation in IDH-mutant cancers. Cancer Discov. 13, 1478–1497. doi:10.1158/2159-8290.CD-22-0825
Gyamfi, J., Kim, J., and Choi, J. (2022). Cancer as a metabolic disorder. Int. J. Mol. Sci. 23, 1155. doi:10.3390/ijms23031155
Gylfe, A. E., Kondelin, J., Turunen, M., Ristolainen, H., Katainen, R., Pitkänen, E., et al. (2013). Identification of candidate oncogenes in human colorectal cancers with microsatellite instability. Gastroenterology 145, 540–543.e22. doi:10.1053/j.gastro.2013.05.015
Hanahan, D. (2022). Hallmarks of cancer: new dimensions. Cancer Discov. 12, 31–46. doi:10.1158/2159-8290.CD-21-1059
Hanahan, D., and Weinberg, R. A. (2011). Hallmarks of cancer: the next generation. Cell 144, 646–674. doi:10.1016/j.cell.2011.02.013
Harris, W. J., Huang, X., Lynch, J. T., Spencer, G. J., Hitchin, J. R., Li, Y., et al. (2012). The histone demethylase KDM1A sustains the oncogenic potential of MLL-AF9 leukemia stem cells. Cancer Cell 21, 473–487. doi:10.1016/j.ccr.2012.03.014
Hart, J. R., Zhang, Y., Liao, L., Ueno, L., Du, L., Jonkers, M., et al. (2015). The butterfly effect in cancer: a single base mutation can remodel the cell. Proc. Natl. Acad. Sci. U. S. A. 112, 1131–1136. doi:10.1073/pnas.1424012112
Hause, R. J., Pritchard, C. C., Shendure, J., and Salipante, S. J. (2016). Classification and characterization of microsatellite instability across 18 cancer types. Nat. Med. 22, 1342–1350. doi:10.1038/nm.4191
Herman, J. G., Umar, A., Polyak, K., Graff, J. R., Ahuja, N., Issa, J. P., et al. (1998). Incidence and functional consequences of hMLH1 promoter hypermethylation in colorectal carcinoma. Proc. Natl. Acad. Sci. U. S. A. 95, 6870–6875. doi:10.1073/pnas.95.12.6870
Hirpara, A., Bloomfield, M., and Duesberg, P. (2018). Speciation theory of carcinogenesis explains karyotypic individuality and long latencies of cancers. Genes (Basel) 9, 402. doi:10.3390/genes9080402
Imai, K., and Yamamoto, H. (2008). Carcinogenesis and microsatellite instability: the interrelationship between genetics and epigenetics. Carcinogenesis 29, 673–680. doi:10.1093/carcin/bgm228
Intlekofer, A. M., Dematteo, R. G., Venneti, S., Finley, L. W. S., Lu, C., Judkins, A. R., et al. (2015). Hypoxia induces production of L-2-Hydroxyglutarate. Cell Metab. 22, 304–311. doi:10.1016/j.cmet.2015.06.023
Ishii, T., Yasuda, K., Akatsuka, A., Hino, O., Hartman, P. S., and Ishii, N. (2005). A mutation in the SDHC gene of complex II increases oxidative stress, resulting in apoptosis and tumorigenesis. Cancer Res. 65, 203–209. doi:10.1158/0008-5472.203.65.1
Ishizawa, K., Rasheed, Z. A., Karisch, R., Wang, Q., Kowalski, J., Susky, E., et al. (2010). Tumor-initiating cells are rare in many human tumors. Cell Stem Cell 7, 279–282. doi:10.1016/j.stem.2010.08.009
Jassim, A., Rahrmann, E. P., Simons, B. D., and Gilbertson, R. J. (2023). Cancers make their own luck: theories of cancer origins. Nat. Rev. Cancer 23, 710–724. doi:10.1038/s41568-023-00602-5
Jeggo, P. A., Pearl, L. H., and Carr, A. M. (2016). DNA repair, genome stability and cancer: a historical perspective. Nat. Rev. Cancer 16, 35–42. doi:10.1038/nrc.2015.4
Johnson, T. I., Costa, A. S. H., Ferguson, A. N., and Frezza, C. (2018). Fumarate hydratase loss promotes mitotic entry in the presence of DNA damage after ionising radiation. Cell Death Dis. 9, 913. doi:10.1038/s41419-018-0912-3
Karalis, T., and Poulogiannis, G. (2024). The emerging role of LPA as an oncometabolite. Cells 13, 629. doi:10.3390/cells13070629
Kentsis, A., and Frank, S. A. (2020). Developmental mutators and early onset cancer. Front. Pediatr. 8, 189. doi:10.3389/fped.2020.00189
Kiebish, M. A., and Seyfried, T. N. (2005). Absence of pathogenic mitochondrial DNA mutations in mouse brain tumors. BMC Cancer 5, 102. doi:10.1186/1471-2407-5-102
Koh, K. H., Kang, H. J., Li, L. S., Kim, N.-G., You, K. T., Yang, E., et al. (2005). Impaired nonhomologous end-joining in mismatch repair-deficient Colon carcinomas. Lab. Invest. 85, 1130–1138. doi:10.1038/labinvest.3700315
Kong, L. R., Gupta, K., Wu, A. J., Perera, D., Ivanyi-Nagy, R., Ahmed, S. M., et al. (2024). A glycolytic metabolite bypasses “two-hit” tumor suppression by BRCA2. Cell 187, 2269–2287.e16. doi:10.1016/j.cell.2024.03.006
Körber, V., Yang, J., Barah, P., Wu, Y., Stichel, D., Gu, Z., et al. (2019). Evolutionary trajectories of IDHWT glioblastomas reveal a common path of early tumorigenesis instigated years ahead of initial diagnosis. Cancer Cell 35, 692–704.e12. doi:10.1016/j.ccell.2019.02.007
Kuang, X., and Li, J. (2022). Chromosome instability and aneuploidy as context-dependent activators or inhibitors of antitumor immunity. Front. Immunol. 13, 895961. doi:10.3389/fimmu.2022.895961
Lamm, N., Ben-David, U., Golan-Lev, T., Storchová, Z., Benvenisty, N., and Kerem, B. (2016). Genomic instability in human pluripotent stem cells arises from replicative stress and chromosome condensation defects. Cell Stem Cell 18, 253–261. doi:10.1016/j.stem.2015.11.003
Lapouge, G., Youssef, K. K., Vokaer, B., Achouri, Y., Michaux, C., Sotiropoulou, P. A., et al. (2011). Identifying the cellular origin of squamous skin tumors. Proc. Natl. Acad. Sci. U. S. A. 108, 7431–7436. doi:10.1073/pnas.1012720108
Laukka, T., Mariani, C. J., Ihantola, T., Cao, J. Z., Hokkanen, J., Kaelin, W. G., et al. (2016). Fumarate and succinate regulate expression of hypoxia-inducible genes via TET enzymes. J. Biol. Chem. 291, 4256–4265. doi:10.1074/jbc.M115.688762
Lee, A. V., Nestler, K. A., and Chiappinelli, K. B. (2024). Therapeutic targeting of DNA methylation alterations in cancer. Pharmacol. Ther. 258, 108640. doi:10.1016/j.pharmthera.2024.108640
Letouzé, E., Martinelli, C., Loriot, C., Burnichon, N., Abermil, N., Ottolenghi, C., et al. (2013). SDH mutations establish a hypermethylator phenotype in paraganglioma. Cancer Cell 23, 739–752. doi:10.1016/j.ccr.2013.04.018
Leung, S. Y., Yuen, S. T., Chung, L. P., Chu, K. M., Chan, A. S., and Ho, J. C. (1999). hMLH1 promoter methylation and lack of hMLH1 expression in sporadic gastric carcinomas with high-frequency microsatellite instability. Cancer Res. 59, 159–164. Available online at: https://aacrjournals.org/cancerres/article/59/1/159/505045/hMLH1-Promoter-Methylation-and-Lack-of-hMLH1.
Li, R., and Zhu, J. (2022). Effects of aneuploidy on cell behaviour and function. Nat. Rev. Mol. Cell Biol. 23, 250–265. doi:10.1038/s41580-021-00436-9
Li, S., Ilaria, R. L., Million, R. P., Daley, G. Q., and Van Etten, R. A. (1999). The P190, P210, and P230 forms of the BCR/ABL oncogene induce a similar chronic myeloid leukemia-like syndrome in mice but have different lymphoid leukemogenic activity. J. Exp. Med. 189, 1399–1412. doi:10.1084/jem.189.9.1399
Li, L., McCormack, A. A., Nicholson, J. M., Fabarius, A., Hehlmann, R., Sachs, R. K., et al. (2009). Cancer-causing karyotypes: chromosomal equilibria between destabilizing aneuploidy and stabilizing selection for oncogenic function. Cancer Genet. Cytogenet 188, 1–25. doi:10.1016/j.cancergencyto.2008.08.016
Li, F., Mao, G., Tong, D., Huang, J., Gu, L., Yang, W., et al. (2013). The histone mark H3K36me3 regulates human DNA mismatch repair through its interaction with MutSα. Cell 153, 590–600. doi:10.1016/j.cell.2013.03.025
Li, F., He, X., Ye, D., Lin, Y., Yu, H., Yao, C., et al. (2015). NADP(+)-IDH mutations promote hypersuccinylation that impairs mitochondria respiration and induces apoptosis resistance. Mol. Cell 60, 661–675. doi:10.1016/j.molcel.2015.10.017
Ligtenberg, M. J. L., Kuiper, R. P., Chan, T. L., Goossens, M., Hebeda, K. M., Voorendt, M., et al. (2009). Heritable somatic methylation and inactivation of MSH2 in families with Lynch syndrome due to deletion of the 3’ exons of TACSTD1. Nat. Genet. 41, 112–117. doi:10.1038/ng.283
Lineweaver, C. H., Bussey, K. J., Blackburn, A. C., and Davies, P. C. W. (2021). Cancer progression as a sequence of atavistic reversions. Bioessays 43, e2000305. doi:10.1002/bies.202000305
Liu, Y., and Yang, C. (2021). Oncometabolites in cancer: current understanding and challenges. Cancer Res. 81, 2820–2823. doi:10.1158/0008-5472.CAN-20-3730
Liu, K., Jiang, M., Lu, Y., Chen, H., Sun, J., Wu, S., et al. (2013). Sox2 cooperates with inflammation-mediated Stat3 activation in the malignant transformation of foregut basal progenitor cells. Cell Stem Cell 12, 304–315. doi:10.1016/j.stem.2013.01.007
Liu, Y., Sethi, N. S., Hinoue, T., Schneider, B. G., Cherniack, A. D., Sanchez-Vega, F., et al. (2018). Comparative molecular analysis of gastrointestinal adenocarcinomas. Cancer Cell 33, 721–735.e8. doi:10.1016/j.ccell.2018.03.010
Loeb, L. A. (2001). A mutator phenotype in cancer. Cancer Res. 61, 3230–3239. Available online at: https://aacrjournals.org/cancerres/article/61/8/3230/508633/A-Mutator-Phenotype-in-Cancer1.
Loeb, L. A. (2010). Mutator phenotype in cancer: origin and consequences. Semin. Cancer Biol. 20, 279–280. doi:10.1016/j.semcancer.2010.10.006
Loeb, L. A. (2016). Human cancers express a mutator phenotype: hypothesis, origin, and consequences. Cancer Res. 76, 2057–2059. doi:10.1158/0008-5472.CAN-16-0794
Losada-García, A., Salido-Guadarrama, I., Cortes-Ramirez, S. A., Cruz-Burgos, M., Morales-Pacheco, M., Vazquez-Santillan, K., et al. (2023). SFRP1 induces a stem cell phenotype in prostate cancer cells. Front. Cell Dev. Biol. 11, 1096923. doi:10.3389/fcell.2023.1096923
Losman, J.-A., Looper, R. E., Koivunen, P., Lee, S., Schneider, R. K., McMahon, C., et al. (2013). (R)-2-hydroxyglutarate is sufficient to promote leukemogenesis and its effects are reversible. Science 339, 1621–1625. doi:10.1126/science.1231677
Lu, J., Mccarter, M., Lian, G., Esposito, G., Capoccia, E., Delli-Bovi, L. C., et al. (2016). Global hypermethylation in fetal cortex of down syndrome due to DNMT3L overexpression. Hum. Mol. Genet. 25, 1714–1727. doi:10.1093/hmg/ddw043
Ly, P., Brunner, S. F., Shoshani, O., Kim, D. H., Lan, W., Pyntikova, T., et al. (2019). Chromosome segregation errors generate a diverse spectrum of simple and complex genomic rearrangements. Nat. Genet. 51, 705–715. doi:10.1038/s41588-019-0360-8
Lytle, N. K., Barber, A. G., and Reya, T. (2018). Stem cell fate in cancer growth, progression and therapy resistance. Nat. Rev. Cancer 18, 669–680. doi:10.1038/s41568-018-0056-x
Magrin, L., Fanale, D., Brando, C., Fiorino, A., Corsini, L. R., Sciacchitano, R., et al. (2021). POLE, POLD1, and NTHL1: the last but not the least hereditary cancer-predisposing genes. Oncogene 40, 5893–5901. doi:10.1038/s41388-021-01984-2
Malkhosyan, S., Rampino, N., Yamamoto, H., and Perucho, M. (1996). Frameshift mutator mutations. Nature 382, 499–500. doi:10.1038/382499a0
Manuel, A. M., Walla, M. D., Dorn, M. T., Tanis, R. M., Piroli, G. G., and Frizzell, N. (2020). Fumarate and oxidative stress synergize to promote stability of C/EBP homologous protein in the adipocyte. Free Radic. Biol. Med. 148, 70–82. doi:10.1016/j.freeradbiomed.2019.12.037
Martínez-Reyes, I., and Chandel, N. S. (2021). Cancer metabolism: looking forward. Nat. Rev. Cancer 21, 669–680. doi:10.1038/s41568-021-00378-6
Mascaux, C., Angelova, M., Vasaturo, A., Beane, J., Hijazi, K., Anthoine, G., et al. (2019). Immune evasion before tumour invasion in early lung squamous carcinogenesis. Nature 571, 570–575. doi:10.1038/s41586-019-1330-0
Mellinghoff, I. K., Ellingson, B. M., Touat, M., Maher, E., De La Fuente, M. I., Holdhoff, M., et al. (2020). Ivosidenib in isocitrate dehydrogenase 1-Mutated advanced glioma. J. Clin. Oncol. 38, 3398–3406. doi:10.1200/JCO.19.03327
Molyneux, G., Geyer, F. C., Magnay, F.-A., McCarthy, A., Kendrick, H., Natrajan, R., et al. (2010). BRCA1 basal-like breast cancers originate from luminal epithelial progenitors and not from basal stem cells. Cell Stem Cell 7, 403–417. doi:10.1016/j.stem.2010.07.010
Moreno-Sánchez, R., Hernández-Esquivel, L., Rivero-Segura, N. A., Marín-Hernández, A., Neuzil, J., Ralph, S. J., et al. (2013). Reactive oxygen species are generated by the respiratory complex II--evidence for lack of contribution of the reverse electron flow in complex I. FEBS J. 280, 927–938. doi:10.1111/febs.12086
Mu, R., Zou, Y.-K., Tu, K., Wang, D.-B., Tang, D., Yu, Z., et al. (2021). Hypoxia promotes pancreatic cancer cell dedifferentiation to stem-like cell phenotypes with high tumorigenic potential by the HIF-1α/Notch signaling pathway. Pancreas 50, 756–765. doi:10.1097/MPA.0000000000001828
Mur, P., García-Mulero, S., Del Valle, J., Magraner-Pardo, L., Vidal, A., Pineda, M., et al. (2020). Role of POLE and POLD1 in familial cancer. Genet. Med. 22, 2089–2100. doi:10.1038/s41436-020-0922-2
Murata, K., Jadhav, U., Madha, S., van Es, J., Dean, J., Cavazza, A., et al. (2020). Ascl2-Dependent cell dedifferentiation drives regeneration of ablated intestinal stem cells. Cell Stem Cell 26, 377–390.e6. doi:10.1016/j.stem.2019.12.011
Murnane, J. P. (2012). Telomere dysfunction and chromosome instability. Mutat. Res. 730, 28–36. doi:10.1016/j.mrfmmm.2011.04.008
Muskens, I. S., Li, S., Jackson, T., Elliot, N., Hansen, H. M., Myint, S. S., et al. (2021). The genome-wide impact of trisomy 21 on DNA methylation and its implications for hematopoiesis. Nat. Commun. 12, 821. doi:10.1038/s41467-021-21064-z
Nagayama, Y., Shimamura, M., and Mitsutake, N. (2016). Cancer stem cells in the thyroid. Front. Endocrinol. (Lausanne) 7, 20. doi:10.3389/fendo.2016.00020
Neering, S. J., Bushnell, T., Sozer, S., Ashton, J., Rossi, R. M., Wang, P.-Y., et al. (2007). Leukemia stem cells in a genetically defined murine model of blast-crisis CML. Blood 110, 2578–2585. doi:10.1182/blood-2007-02-073031
Nguyen, P. H., Giraud, J., Staedel, C., Chambonnier, L., Dubus, P., Chevret, E., et al. (2016). All-trans retinoic acid targets gastric cancer stem cells and inhibits patient-derived gastric carcinoma tumor growth. Oncogene 35, 5619–5628. doi:10.1038/onc.2016.87
Nicholson, J. M., and Cimini, D. (2013). Cancer karyotypes: survival of the fittest. Front. Oncol. 3, 148. doi:10.3389/fonc.2013.00148
Nie, M., Yao, K., Zhu, X., Chen, N., Xiao, N., Wang, Y., et al. (2021). Evolutionary metabolic landscape from preneoplasia to invasive lung adenocarcinoma. Nat. Commun. 12, 6479. doi:10.1038/s41467-021-26685-y
Nishikawa, T., Edelstein, D., Du, X. L., Yamagishi, S., Matsumura, T., Kaneda, Y., et al. (2000). Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature 404, 787–790. doi:10.1038/35008121
Oldham, W. M., Clish, C. B., Yang, Y., and Loscalzo, J. (2015). Hypoxia-mediated increases in L-2-hydroxyglutarate coordinate the metabolic response to reductive stress. Cell Metab. 22, 291–303. doi:10.1016/j.cmet.2015.06.021
Otani, K., Li, X., Arakawa, T., Chan, F. K. L., and Yu, J. (2013). Epigenetic-mediated tumor suppressor genes as diagnostic or prognostic biomarkers in gastric cancer. Expert Rev. Mol. Diagn 13, 445–455. doi:10.1586/erm.13.32
Pandkar, M. R., Sinha, S., Samaiya, A., and Shukla, S. (2023). Oncometabolite lactate enhances breast cancer progression by orchestrating histone lactylation-dependent c-Myc expression. Transl. Oncol. 37, 101758. doi:10.1016/j.tranon.2023.101758
Pavelka, N., Rancati, G., Zhu, J., Bradford, W. D., Saraf, A., Florens, L., et al. (2010). Aneuploidy confers quantitative proteome changes and phenotypic variation in budding yeast. Nature 468, 321–325. doi:10.1038/nature09529
Pavlova, N. N., Zhu, J., and Thompson, C. B. (2022). The hallmarks of cancer metabolism: still emerging. Cell Metab. 34, 355–377. doi:10.1016/j.cmet.2022.01.007
Pećina-Šlaus, N., Kafka, A., Salamon, I., and Bukovac, A. (2020). Mismatch repair pathway, genome stability and cancer. Front. Mol. Biosci. 7, 122. doi:10.3389/fmolb.2020.00122
Perekatt, A. O., Shah, P. P., Cheung, S., Jariwala, N., Wu, A., Gandhi, V., et al. (2018). SMAD4 suppresses WNT-driven dedifferentiation and oncogenesis in the differentiated gut epithelium. Cancer Res. 78, 4878–4890. doi:10.1158/0008-5472.CAN-18-0043
Pfister, K., Pipka, J. L., Chiang, C., Liu, Y., Clark, R. A., Keller, R., et al. (2018). Identification of drivers of aneuploidy in breast tumors. Cell Rep. 23, 2758–2769. doi:10.1016/j.celrep.2018.04.102
Pollard, P. J., Brière, J. J., Alam, N. A., Barwell, J., Barclay, E., Wortham, N. C., et al. (2005). Accumulation of krebs cycle intermediates and over-expression of HIF1alpha in tumours which result from germline FH and SDH mutations. Hum. Mol. Genet. 14, 2231–2239. doi:10.1093/hmg/ddi227
Potapova, T. A., Zhu, J., and Li, R. (2013). Aneuploidy and chromosomal instability: a vicious cycle driving cellular evolution and cancer genome chaos. Cancer Metastasis Rev. 32, 377–389. doi:10.1007/s10555-013-9436-6
Quaas, A., Rehkaemper, J., Rueschoff, J., Pamuk, A., Zander, T., Hillmer, A., et al. (2021). Occurrence of high microsatellite-Instability/Mismatch repair deficiency in nearly 2,000 human adenocarcinomas of the gastrointestinal tract, pancreas, and bile ducts: a study from a large German comprehensive cancer center. Front. Oncol. 11, 569475. doi:10.3389/fonc.2021.569475
Raychaudhuri, D., Bhattacharya, R., Sinha, B. P., Liu, C. S. C., Ghosh, A. R., Rahaman, O., et al. (2019). Lactate induces pro-tumor reprogramming in intratumoral plasmacytoid dendritic cells. Front. Immunol. 10, 1878. doi:10.3389/fimmu.2019.01878
Rayner, E., van Gool, I. C., Palles, C., Kearsey, S. E., Bosse, T., Tomlinson, I., et al. (2016). A panoply of errors: polymerase proofreading domain mutations in cancer. Nat. Rev. Cancer 16, 71–81. doi:10.1038/nrc.2015.12
Raz, A. A., and Yamashita, Y. M. (2025). Rewinding the clock: mechanisms of dedifferentiation. Curr. Opin. Genet. Dev. 93, 102353. doi:10.1016/j.gde.2025.102353
Rendo, V., Schubert, M., Khuu, N., Suarez Peredo Rodriguez, M. F., Whyte, D., Ling, X., et al. (2025). A compendium of amplification-related gain of sensitivity genes in human cancer. Nat. Commun. 16, 1077. doi:10.1038/s41467-025-56301-2
Riva, L., Pandiri, A. R., Li, Y. R., Droop, A., Hewinson, J., Quail, M. A., et al. (2020). The mutational signature profile of known and suspected human carcinogens in mice. Nat. Genet. 52, 1189–1197. doi:10.1038/s41588-020-0692-4
Roberts, S. A., and Gordenin, D. A. (2014). Hypermutation in human cancer genomes: footprints and mechanisms. Nat. Rev. Cancer 14, 786–800. doi:10.1038/nrc3816
Rochefort, P., Desseigne, F., Bonadona, V., Dussart, S., Coutzac, C., Sarabi, M., et al. (2021). Immune checkpoint inhibitor sensitivity of DNA repair deficient tumors. Immunotherapy 13, 1205–1213. doi:10.2217/imt-2021-0024
Ropero, S., Fraga, M. F., Ballestar, E., Hamelin, R., Yamamoto, H., Boix-Chornet, M., et al. (2006). A truncating mutation of HDAC2 in human cancers confers resistance to histone deacetylase inhibition. Nat. Genet. 38, 566–569. doi:10.1038/ng1773
Sack, L. M., Davoli, T., Li, M. Z., Li, Y., Xu, Q., Naxerova, K., et al. (2018). Profound tissue specificity in proliferation control underlies cancer drivers and aneuploidy patterns. Cell 173, 499–514.e23. doi:10.1016/j.cell.2018.02.037
Saletta, F., Matullo, G., Manuguerra, M., Arena, S., Bardelli, A., and Vineis, P. (2007). Exposure to the tobacco smoke constituent 4-aminobiphenyl induces chromosomal instability in human cancer cells. Cancer Res. 67, 7088–7094. doi:10.1158/0008-5472.CAN-06-4420
San-Millán, I., Julian, C. G., Matarazzo, C., Martinez, J., and Brooks, G. A. (2019). Is lactate an oncometabolite? Evidence supporting a role for lactate in the regulation of transcriptional activity of cancer-related genes in MCF7 breast cancer cells. Front. Oncol. 9, 1536. doi:10.3389/fonc.2019.01536
Santaguida, S., and Amon, A. (2015). Short- and long-term effects of chromosome mis-segregation and aneuploidy. Nat. Rev. Mol. Cell Biol. 16, 473–485. doi:10.1038/nrm4025
Schukken, K. M., and Foijer, F. (2018). CIN and aneuploidy: different concepts, different consequences. Bioessays 40, 1700147. doi:10.1002/bies.201700147
Schüller, U., Heine, V. M., Mao, J., Kho, A. T., Dillon, A. K., Han, Y.-G., et al. (2008). Acquisition of granule neuron precursor identity is a critical determinant of progenitor cell competence to form Shh-induced medulloblastoma. Cancer Cell 14, 123–134. doi:10.1016/j.ccr.2008.07.005
Schwitalla, S., Fingerle, A. A., Cammareri, P., Nebelsiek, T., Göktuna, S. I., Ziegler, P. K., et al. (2013). Intestinal tumorigenesis initiated by dedifferentiation and acquisition of stem-cell-like properties. Cell 152, 25–38. doi:10.1016/j.cell.2012.12.012
Sciacovelli, M., and Frezza, C. (2016). Oncometabolites: unconventional triggers of oncogenic signalling cascades. Free Radic. Biol. Med. 100, 175–181. doi:10.1016/j.freeradbiomed.2016.04.025
Sciacovelli, M., Gonçalves, E., Johnson, T. I., Zecchini, V. R., da Costa, A. S. H., Gaude, E., et al. (2016). Fumarate is an epigenetic modifier that elicits epithelial-to-mesenchymal transition. Nature 537, 544–547. doi:10.1038/nature19353
Sdeor, E., Okada, H., Saad, R., Ben-Yishay, T., and Ben-David, U. (2024). Aneuploidy as a driver of human cancer. Nat. Genet. 56, 2014–2026. doi:10.1038/s41588-024-01916-2
Selak, M. A., Armour, S. M., MacKenzie, E. D., Boulahbel, H., Watson, D. G., Mansfield, K. D., et al. (2005). Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-alpha prolyl hydroxylase. Cancer Cell 7, 77–85. doi:10.1016/j.ccr.2004.11.022
Seyfried, T. N. (2015). Cancer as a mitochondrial metabolic disease. Front. Cell Dev. Biol. 3, 43. doi:10.3389/fcell.2015.00043
Shanmugasundaram, K., Nayak, B., Shim, E.-H., Livi, C. B., Block, K., and Sudarshan, S. (2014). The oncometabolite fumarate promotes pseudohypoxia through noncanonical activation of NF-κB signaling. J. Biol. Chem. 289, 24691–24699. doi:10.1074/jbc.M114.568162
Shen, J., Ju, Z., Zhao, W., Wang, L., Peng, Y., Ge, Z., et al. (2018). ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade. Nat. Med. 24, 556–562. doi:10.1038/s41591-018-0012-z
Shlien, A., Campbell, B. B., de Borja, R., Alexandrov, L. B., Merico, D., Wedge, D., et al. (2015). Combined hereditary and somatic mutations of replication error repair genes result in rapid onset of ultra-hypermutated cancers. Nat. Genet. 47, 257–262. doi:10.1038/ng.3202
Shlush, L. I., Zandi, S., Mitchell, A., Chen, W. C., Brandwein, J. M., Gupta, V., et al. (2014). Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature 506, 328–333. doi:10.1038/nature13038
Shumilov, A., Tsai, M.-H., Schlosser, Y. T., Kratz, A.-S., Bernhardt, K., Fink, S., et al. (2017). Epstein-barr virus particles induce centrosome amplification and chromosomal instability. Nat. Commun. 8, 14257. doi:10.1038/ncomms14257
Smalley, M., and Ashworth, A. (2003). Stem cells and breast cancer: a field in transit. Nat. Rev. Cancer 3, 832–844. doi:10.1038/nrc1212
Smith, B. A., Sokolov, A., Uzunangelov, V., Baertsch, R., Newton, Y., Graim, K., et al. (2015). A basal stem cell signature identifies aggressive prostate cancer phenotypes. Proc. Natl. Acad. Sci. U. S. A. 112, E6544–E6552. doi:10.1073/pnas.1518007112
Soto, M., Raaijmakers, J. A., Bakker, B., Spierings, D. C. J., Lansdorp, P. M., Foijer, F., et al. (2017). p53 prohibits propagation of chromosome segregation errors that produce structural aneuploidies. Cell Rep. 19, 2423–2431. doi:10.1016/j.celrep.2017.05.055
Sourbier, C., Ricketts, C. J., Matsumoto, S., Crooks, D. R., Liao, P.-J., Mannes, P. Z., et al. (2014). Targeting ABL1-mediated oxidative stress adaptation in fumarate hydratase-deficient cancer. Cancer Cell 26, 840–850. doi:10.1016/j.ccell.2014.10.005
Stingele, S., Stoehr, G., Peplowska, K., Cox, J., Mann, M., and Storchova, Z. (2012). Global analysis of genome, transcriptome and proteome reveals the response to aneuploidy in human cells. Mol. Syst. Biol. 8, 608. doi:10.1038/msb.2012.40
Sudarshan, S., Sourbier, C., Kong, H.-S., Block, K., Valera Romero, V. A., Yang, Y., et al. (2009). Fumarate hydratase deficiency in renal cancer induces glycolytic addiction and hypoxia-inducible transcription factor 1alpha stabilization by glucose-dependent generation of reactive oxygen species. Mol. Cell Biol. 29, 4080–4090. doi:10.1128/MCB.00483-09
Taylor, A. M., Shih, J., Ha, G., Gao, G. F., Zhang, X., Berger, A. C., et al. (2018). Genomic and functional approaches to understanding cancer aneuploidy. Cancer Cell 33, 676–689.e3. doi:10.1016/j.ccell.2018.03.007
Terunuma, A., Putluri, N., Mishra, P., Mathé, E. A., Dorsey, T. H., Yi, M., et al. (2014). MYC-Driven accumulation of 2-hydroxyglutarate is associated with breast cancer prognosis. J. Clin. Invest. 124, 398–412. doi:10.1172/JCI71180
Thankamony, A. P., Saxena, K., Murali, R., Jolly, M. K., and Nair, R. (2020). Cancer stem cell plasticity - a deadly deal. Front. Mol. Biosci. 7, 79. doi:10.3389/fmolb.2020.00079
Thomas, S. A., Storey, K. B., Baynes, J. W., and Frizzell, N. (2012). Tissue distribution of S-(2-succino)cysteine (2SC), a biomarker of mitochondrial stress in obesity and diabetes. Obes. (Silver Spring) 20, 263–269. doi:10.1038/oby.2011.340
Thompson, S. L., and Compton, D. A. (2010). Proliferation of aneuploid human cells is limited by a p53-dependent mechanism. J. Cell Biol. 188, 369–381. doi:10.1083/jcb.200905057
Tomlinson, I. P. M., Alam, N. A., Rowan, A. J., Barclay, E., Jaeger, E. E. M., Kelsell, D., et al. (2002). Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat. Genet. 30, 406–410. doi:10.1038/ng849
Tu, S.-M., Zhang, M., Wood, C. G., and Pisters, L. L. (2021). Stem cell theory of cancer: origin of tumor heterogeneity and plasticity. Cancers (Basel) 13, 4006. doi:10.3390/cancers13164006
Tuupanen, S., Hänninen, U. A., Kondelin, J., von Nandelstadh, P., Cajuso, T., Gylfe, A. E., et al. (2014). Identification of 33 candidate oncogenes by screening for base-specific mutations. Br. J. Cancer 111, 1657–1662. doi:10.1038/bjc.2014.429
Valeri, N., Gasparini, P., Braconi, C., Paone, A., Lovat, F., Fabbri, M., et al. (2010a). MicroRNA-21 induces resistance to 5-fluorouracil by down-regulating human DNA MutS homolog 2 (hMSH2). Proc. Natl. Acad. Sci. U. S. A. 107, 21098–21103. doi:10.1073/pnas.1015541107
Valeri, N., Gasparini, P., Fabbri, M., Braconi, C., Veronese, A., Lovat, F., et al. (2010b). Modulation of mismatch repair and genomic stability by miR-155. Proc. Natl. Acad. Sci. U. S. A. 107, 6982–6987. doi:10.1073/pnas.1002472107
Vasudevan, A., Schukken, K. M., Sausville, E. L., Girish, V., Adebambo, O. A., and Sheltzer, J. M. (2021). Aneuploidy as a promoter and suppressor of malignant growth. Nat. Rev. Cancer 21, 89–103. doi:10.1038/s41568-020-00321-1
Venkatesan, R. N., Treuting, P. M., Fuller, E. D., Goldsby, R. E., Norwood, T. H., Gooley, T. A., et al. (2007). Mutation at the polymerase active site of mouse DNA polymerase Delta increases genomic instability and accelerates tumorigenesis. Mol. Cell Biol. 27, 7669–7682. doi:10.1128/MCB.00002-07
Vilar, E., and Gruber, S. B. (2010). Microsatellite instability in colorectal cancer-the stable evidence. Nat. Rev. Clin. Oncol. 7, 153–162. doi:10.1038/nrclinonc.2009.237
Vitre, B. D., and Cleveland, D. W. (2012). Centrosomes, chromosome instability (CIN) and aneuploidy. Curr. Opin. Cell Biol. 24, 809–815. doi:10.1016/j.ceb.2012.10.006
Wang, Y., Yang, J., Zheng, H., Tomasek, G. J., Zhang, P., McKeever, P. E., et al. (2009). Expression of mutant p53 proteins implicates a lineage relationship between neural stem cells and malignant astrocytic glioma in a murine model. Cancer Cell 15, 514–526. doi:10.1016/j.ccr.2009.04.001
Ward, P. S., Patel, J., Wise, D. R., Abdel-Wahab, O., Bennett, B. D., Coller, H. A., et al. (2010). The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 17, 225–234. doi:10.1016/j.ccr.2010.01.020
White, A. C., Tran, K., Khuu, J., Dang, C., Cui, Y., Binder, S. W., et al. (2011). Defining the origins of Ras/p53-mediated squamous cell carcinoma. Proc. Natl. Acad. Sci. U. S. A. 108, 7425–7430. doi:10.1073/pnas.1012670108
Whitehall, V. L. J., Dumenil, T. D., McKeone, D. M., Bond, C. E., Bettington, M. L., Buttenshaw, R. L., et al. (2014). Isocitrate dehydrogenase 1 R132C mutation occurs exclusively in microsatellite stable colorectal cancers with the CpG island methylator phenotype. Epigenetics 9, 1454–1460. doi:10.4161/15592294.2014.971624
Williams, B. R., Prabhu, V. R., Hunter, K. E., Glazier, C. M., Whittaker, C. A., Housman, D. E., et al. (2008). Aneuploidy affects proliferation and spontaneous immortalization in Mammalian cells. Science 322, 703–709. doi:10.1126/science.1160058
Wise, D. R., Ward, P. S., Shay, J. E. S., Cross, J. R., Gruber, J. J., Sachdeva, U. M., et al. (2011). Hypoxia promotes isocitrate dehydrogenase-dependent carboxylation of α-ketoglutarate to citrate to support cell growth and viability. Proc. Natl. Acad. Sci. U. S. A. 108, 19611–19616. doi:10.1073/pnas.1117773108
Woerner, S. M., Kloor, M., Mueller, A., Rueschoff, J., Friedrichs, N., Buettner, R., et al. (2005). Microsatellite instability of selective target genes in HNPCC-associated colon adenomas. Oncogene 24, 2525–2535. doi:10.1038/sj.onc.1208456
Woo, R. A., and Poon, R. Y. C. (2004). Activated oncogenes promote and cooperate with chromosomal instability for neoplastic transformation. Genes Dev. 18, 1317–1330. doi:10.1101/gad.1165204
Worrall, J. T., Tamura, N., Mazzagatti, A., Shaikh, N., van Lingen, T., Bakker, B., et al. (2018). Non-random mis-segregation of human chromosomes. Cell Rep. 23, 3366–3380. doi:10.1016/j.celrep.2018.05.047
Xiang, L., and Semenza, G. L. (2019). Hypoxia-inducible factors promote breast cancer stem cell specification and maintenance in response to hypoxia or cytotoxic chemotherapy. Adv. Cancer Res. 141, 175–212. doi:10.1016/bs.acr.2018.11.001
Xiao, M., Yang, H., Xu, W., Ma, S., Lin, H., Zhu, H., et al. (2012). Inhibition of α-KG-dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors. Genes Dev. 26, 1326–1338. doi:10.1101/gad.191056.112
Xu, W., Yang, H., Liu, Y., Yang, Y., Wang, P., Kim, S.-H., et al. (2011). Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell 19, 17–30. doi:10.1016/j.ccr.2010.12.014
Yamamoto, H., and Imai, K. (2015). Microsatellite instability: an update. Arch. Toxicol. 89, 899–921. doi:10.1007/s00204-015-1474-0
Yang, Z. J., Ellis, T., Markant, S. L., Read, T. A., Kessler, J. D., Bourboulas, M., et al. (2008). Medulloblastoma can be initiated by deletion of patched in lineage-restricted progenitors or stem cells. Cancer Cell 14, 135–145. doi:10.1016/j.ccr.2008.07.003
Yang, M., Soga, T., Pollard, P. J., and Adam, J. (2012). The emerging role of fumarate as an oncometabolite. Front. Oncol. 2, 85. doi:10.3389/fonc.2012.00085
Yang, M., Soga, T., and Pollard, P. J. (2013). Oncometabolites: linking altered metabolism with cancer. J. Clin. Invest. 123, 3652–3658. doi:10.1172/JCI67228
Yogev, O., Yogev, O., Singer, E., Shaulian, E., Goldberg, M., Fox, T. D., et al. (2010). Fumarase: a mitochondrial metabolic enzyme and a cytosolic/nuclear component of the DNA damage response. PLoS Biol. 8, e1000328. doi:10.1371/journal.pbio.1000328
Yong, C., Stewart, G. D., and Frezza, C. (2020). Oncometabolites in renal cancer. Nat. Rev. Nephrol. 16, 156–172. doi:10.1038/s41581-019-0210-z
Yuan, S., Almagro, J., and Fuchs, E. (2024). Beyond genetics: driving cancer with the tumour microenvironment behind the wheel. Nat. Rev. Cancer 24, 274–286. doi:10.1038/s41568-023-00660-9
Zhang, F., Chen, X. P., Zhang, W., Dong, H. H., Xiang, S., Zhang, W. G., et al. (2008). Combined hepatocellular cholangiocarcinoma originating from hepatic progenitor cells: immunohistochemical and double-fluorescence immunostaining evidence. Histopathology 52, 224–232. doi:10.1111/j.1365-2559.2007.02929.x
Zhu, J., Pavelka, N., Bradford, W. D., Rancati, G., and Li, R. (2012). Karyotypic determinants of chromosome instability in aneuploid budding yeast. PLoS Genet. 8, e1002719. doi:10.1371/journal.pgen.1002719
Zhu, J., Tsai, H. J., Gordon, M. R., and Li, R. (2018). Cellular stress associated with aneuploidy. Dev. Cell 44, 420–431. doi:10.1016/j.devcel.2018.02.002
Keywords: cancer origin, tumorigenesis, oncogenesis, carcinogenesis, malignant transformation, cancer initiation, tumorigenic pathway
Citation: Wang R and Yan Z (2025) Carcinogenesis trajectories. Front. Cell Dev. Biol. 13:1671115. doi: 10.3389/fcell.2025.1671115
Received: 22 July 2025; Accepted: 21 November 2025;
Published: 09 December 2025.
Edited by:
Olga V. Anatskaya, Russian Academy of Sciences (RAS), RussiaReviewed by:
Ella L. Kim, Johannes Gutenberg University Mainz, GermanyAndrzej Kasperski, University of Zielona Góra, Poland
Copyright © 2025 Wang and Yan. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Zhaopeng Yan, enB5YW5AY211LmVkdS5jbg==
†ORCID: Rui Wang, orcid.org/0000-0003-1728-1868; Zhaopeng Yan, orcid.org/0000-0003-1247-2067