Open Access

Role of RUNX2 in breast cancer development and drug resistance (Review)

  • Authors:
    • Wentao Si
    • Chen Kan
    • Leisheng Zhang
    • Feifei Li
  • View Affiliations

  • Published online on: March 15, 2023     https://doi.org/10.3892/ol.2023.13762
  • Article Number: 176
  • Copyright: © Si et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Breast cancer is the most common malignancy and ranks second among the causes of tumor‑associated death in females. The recurrence and drug resistance of breast cancer are intractable due to the presence of breast cancer stem cells (BCSCs), which are adequate to initiate tumor formation and refractory to conventional remedies. Runt‑related transcription factor 2 (RUNX2), a pivotal transcription factor in mammary gland and bone development, has also been related to metastatic cancer and BCSCs. State‑of‑the‑art research has indicated the retention of RUNX2 expression in a more invasive subtype of breast cancer, and in particular, triple‑negative breast cancer development and drug resistance are associated with estrogen receptor signaling pathways. The present review mainly focused on the latest updates on RUNX2 in BCSCs and their roles in breast cancer progression and drug resistance, providing insight that may aid the development of RUNX2‑based diagnostics and treatments for breast cancer in clinical practice.

Introduction

Breast cancer, a phenomenon of uncontrolled proliferation of breast epithelial cells caused by carcinogenic factors, is the most common malignancy and ranks second among the causes of tumor-associated death in females; it has become a public health issue and endangers women's health worldwide (1). According to the data released by the International Agency for Research on Cancer of the World Health Organization, breast cancer surpassed lung cancer in 2020 and became the world's most significant cancer type with the most newly-diagnosed cases and deaths among females (2). Women with early breast cancer or locoregional relapse are usually able to be cured by multidisciplinary remedies, such as surgery, radiotherapy, chemotherapy and pharmacotherapy. However, for those with metastatic breast cancer, the clinical treatment outcomes are still far from satisfactory and only palliative care (e.g., focusing on the prolongation of survival and alleviation of symptoms) may be possible, largely due to the metastatic heterogeneity and the elusive pathogenesis of breast cancer. For instance, as reviewed by Li et al (3), triple-negative breast cancer (TNBC) may have worse cause-specific survival and overall survival than the non-TNBC counterpart in all stages and substages, regardless of influencing factors from univariate and multivariate analyses (e.g., tumor grade, age, ethnicity, surgery and radiation treatments).

Runt-related transcription factor 2 (RUNX2) belongs to the RUNX family (including RUNX1-3) and has been recognized as a key modulator and master transcription factor for osteogenesis, as well as prostate and skeletal development (47). To date, RUNX2 has been involved in diverse physiological processes, including osteogenic differentiation of mesenchymal stem/stromal cells, chondrocyte hypertrophy, immunomodulation, vascular invasion and endothelial cell migration via modulating a variety of signaling cascades (e.g., MAPK and NK-κB pathway macrophage reprogramming) (811). Meanwhile, the dysregulation and alteration of RUNX2 expression or activity may result in arteriosclerosis, skeletal dysplasia (e.g., cleidocranial dysplasia) and tumorigenesis (4,1214). For instance, RUNX2 is involved in the progression of various tumor types, such as osteosarcoma, renal cell carcinoma, gastric cancer and breast cancer (1520). For instance, a recent study by our group reported the facilitating effect of RUNX2 during aggressiveness and chemoresistance of TNBC cells via activating MMP1, which was significantly associated with poor prognosis (21). Of note, other studies have also indicated the involvement of RUNX2 in breast cancer stem cells (BCSCs) and breast cancer progression (22,23). For instance, Zhang et al (23,24) found that RUNX2 was required for the activity of CD44+/CD24−/low BCSCs during breast cancer development, while miR-205/RUNX2 axis was further identified with s negative regulatory effect upon the activity of CD44+/CD24 BCSCs. Taken together, these studies indicated the involvement of RUNX2 in BCSCs and its roles in breast cancer diagnosis and drug resistance, revealing its promising prospective clinical application and utility as an antitumor drug target in the future.

The present review article mainly focused on the roles of RUNX2 and BCSCs in the progression and management of breast cancer. Furthermore, current advances in therapeutic approaches for breast cancer, ranging from RUNX2-based remedies to drug resistance, were summarized. Collectively, the systematic and detailed research on RUNX2 has constituted a prospective area of diagnosis and treatment strategy innovation for breast cancer.

Breast cancer and classification

Breast cancers are divided into different molecular subtypes, including luminal A, luminal B, human epidermal growth factor receptor 2 (HER2)-positive and basal-like breast cancer. Among them, luminal A is a class of breast cancer with an immunohistochemical index showing estrogen receptor (ER) positivity, progesterone receptor (PR) positivity and HER2 negativity. Of the aforementioned subtypes, luminal A breast cancer is the most common type of breast cancer, which may be targeted by hormonal therapy with a favorable prognosis (25,26).

Distinct from luminal A breast cancer, the HER2+ luminal B subtype features poor differentiation and a worse prognosis (27). HER2-positive breast cancer accounts for 12–20% of all invasive breast cancers, which exhibits multifaceted deteriorative characteristics, including a higher degree of malignancy, faster disease progression, greater likelihood of relapse and metastasis, and poor prognosis (28). HER2-targeted therapies have been widely used for the treatment of HER2-positive breast cancer, such as monoclonal antibodies, kinase inhibitors and antibody-drug conjugates (ADCs). Basal-like breast cancer is positive for basal cytokeratin and epidermal growth factor receptors and/or c-Kit expression, and it accounts for ~15% of breast cancers. It usually features rapid development of local and distant metastases, and thus, it has relatively high mortality rates (29). Gene expression profiling typically classifies triple-negative breast cancer (TNBC) as a major subtype of basal-like breast cancer without the expression of ER, PR or HER2 (30), which is clinically characterized by a strong aggressive nature, high metastatic potential, proneness to relapse and poor prognosis (31). Although poly(ADP ribose) polymerase inhibitors (e.g., programmed cell death-1 monoclonal antibody, trophoblast cell surface antigen 2 ADC) have been used for a subset of TNBCs, the prognosis of patients with TNBC is still far from satisfactory (Table I).

Table I.

Molecular typing of breast cancer.

Table I.

Molecular typing of breast cancer.

SubtypeERPRHER2
Luminal A++-
Luminal B (HER2-negative)+--
HER2-positive (ER-positive)+Any+
HER2-positive (ER-negative)--+
Basal-like (Triple negative)---

[i] The information presented is derived from Ref. (132). ER, estrogen receptor; PR, progesterone receptor; HER2, human epidermal growth factor receptor 2.

Status of clinical treatment of breast cancer

Breast cancer is a heterogeneous disease with highly aggressive and complex biological features, and the clinical treatment and prognosis of different patients vary greatly (32). As mentioned above, patients with early breast cancer may be effectively cured and the focus of treatment is to avoid overtreatment and undertreatment (33). However, metastatic breast cancer cannot be radically cured by current clinical remedies and its management mainly focuses on prolonging survival time and maintaining quality of life instead (34).

Currently, the major clinical remedies of breast cancer are endocrine therapy, molecular targeted therapy and chemotherapy (26). For instance, patients with stage I and II breast cancers are recommended to undergo breast-conserving surgery and radiation treatment, whereas those with node-positive breast cancer are systemically treated with chemotherapy, endocrine therapy and trastuzumab. As to patients with stage III breast cancer, chemotherapy is applied to facilitate breast-conserving surgery (35).

However, as the molecular phenotypes vary across breast cancers, breast cancer is not sensitive to either conventional endocrine therapy or molecular targeted therapies (36). Therefore, chemotherapy remains the primary strategy for breast cancer management. However, the efficacy of conventional postoperative adjuvant chemotherapy is unreliable, and residual metastatic lesions eventually lead to tumor recurrence, which largely attributes to secondary tumor recurrence and metastasis caused by acquired chemotherapy resistance (37,38). Thus, there is an urgent need to explore the drug resistance mechanisms and to identify novel therapeutic targets to finally overcome the resistance of breast cancer to chemotherapy, which is of great clinical significance for clinical practice in the future. Of note, state-of-the-art updates have indicated the role of the transcription factor RUNX2 in BCSCs and drug resistance in breast cancer, which will provide overwhelming new references for the development of the next generation of therapeutic targets and innovative drugs for breast cancer management.

The RUNX family

RUNX is a highly conserved transcription factor family that has an important role in the regulation of gene expression involved in embryonic development and cell differentiation. Recent studies have demonstrated that members of the RUNX transcription factor family are involved in the differentiation of a variety of hematopoietic cells. RUNX2 is required for osteogenesis, whereas RUNX1 and RUNX3 control blood cell development at different stages of cell lineage specification.

RUNX1 is also known as acute myeloid leukemia gene 1. Mutations in this gene are common in patients with lymphoblastic and myeloid leukemia (39). It has been suggested that RUNX1 deficiency is associated with familial platelet disorders. Familial platelet disorders predispose to myeloid leukemia and are associated with thrombocytopenia, as well as marked reductions in B-lymphoid, T-lymphoid and myeloid lineages (40).

RUNX2 has a key role in the development of bone and cartilage tissue, while abnormal expression of RUNX2 is associated with a variety of cancer types, and in particular, the process of tumor bone metastasis (41). To date, RUNX2 has been proven to be involved in melanoma, thyroid cancer and hepatocellular carcinoma (4244). For instance, Guan et al (44) found that circular (circ) RNA_102272 promoted cisplatin resistance of hepatocellular carcinoma via inhibiting the targeting effect of microRNA (miR)-326 upon RUNX2. Of note, Matthijssens et al (45) reported that RUNX2 was able to significantly induce glycolysis and oxidative phosphorylation, and resulted in increased mitochondrial activity, which collectively suggested the promoting effect of RUNX2 in accelerating tumor cell metabolism, and in particular, in benefiting the invasion and migration of leukemic cells via mediating the interaction between glycolysis and mitochondrial respiration. RUNX2 also functions in the development of gastric cancer, including tumor invasion and metastasis by simultaneously facilitating the proliferation of gastric cancer cells and increasing the self-renewal potential (19). Ji et al (46) found that metastasis associated lung adenocarcinoma transcript 1 was able to modulate the transcription and translation of RUNX2 to further increase the metastasis of recurrent colorectal cancer via binding to miR-15 family members, inhibiting LDL receptor related protein 6 expression and enhancing β-catenin signaling, or binding to splicing factor proline and glutamine rich (SFPQ) and dissociating the SFPQ/polypyrimidine tract binding protein 2 dimer. Collectively, RUNX2 holds the potential to function as an important indicator for the detection of multiple cancers in the early stage, which also serves as a promising therapeutic target for clinical practice.

RUNX3 has a significant role in the occurrence and development of a variety of human cancers. Zhang et al (47) indicated that RUNX3 inhibits colorectal cancer proliferation and metastasis. Liu et al (48) found that RUNX3 inhibits glutamine metabolism in gastric cancer. A study identified a NO•/RUNX3/kynurenine metabolic axis, which enhances disease aggressiveness in pancreatic cancer (49).

To summarize, the RUNX family not only plays a role in normal physiological functions, but abnormal expression of any member of the family may cause a variety of different diseases.

Physiological and pathological roles of RUNX2

RUNX2, together with RUNX1 and RUNX3, is an essential transcription factor of the RUNX family (50). To date, studies have indicated the role of RUNX2 in numerous physiological and pathological processes, such as osteoblast differentiation, chondrocyte maturation (51), skeletal and mammary gland development (22), osteoarthritis, osteosarcoma, prostatic carcinoma, gastric cancer and even breast cancer (19,5255).

Roles of RUNX2 in physiological processes

RUNX2 has important roles in various types of cells, such as chondrocytes, osteoblasts and mesenchymal stem/stromal cells (MSCs) (56). Meanwhile, RUNX2 has been reported essential for breast development (57), which also exhibits good interaction with twist family BHLH transcription factor (TWIST)1 in regulating cranial neural crest-derived cell fate and thus guides craniofacial muscle development (58).

For decades, RUNX2 has been recognized as a key transcription factor for bone formation and osteoblast differentiation, as identified by cell sorting, lineage tracing and single-cell transcriptome analysis. For instance, Shu et al (59) developed a dual-recombinase fate-mapping system for the capture of the spatio-temporal skeletal progenitor transition during postnatal bone formation. During intramembranous ossification, RUNX2 promotes the differentiation of MSCs into anterior osteoblasts and immature osteoblasts (12). Furthermore, Liu and Lee (60) reviewed the regulatory network of key transcription factors governing bone formation, such as Msh homeobox 2, TWIST and promyelocytic leukemia zinc-finger protein. During endochondral ossification, RUNX2 maintains the survival of terminal hypertrophic cartilage and promotes its trans-differentiation into osteoblasts (61). Despite the fast-growing understanding of the transcriptional mechanisms of osteogenesis, the detailed mechanisms of RUNX2-related skeletal development and precise orchestration of osteogenesis remain largely elusive.

RUNX2 has also been reported to have a crucial function during breast development via directly regulating the expression pattern of a number of genes related to mammary gland development (62). Studies have revealed the expression of RUNX2 in mammary tissues, including basal cells and luminal cells, which thereby participates in breast development. For instance, Inman and Shore (63) and Sato et al (64) verified that osteopontin may act as a target of RUNX2 and exert its function in breast differentiation in breast epithelial cells during pregnancy and lactation.

RUNX2 is associated with breast cancer

Besides its critical role in physiological development, RUNX2 also functions as an oncogene in numerous types of cancer. RUNX2 has been associated with a wide variety of processes, including tumor progression and heterogeneity, via mediating the responses of cells to signaling pathways hyperactive in tumors (65), such as the connective tissue growth factor-RUNX2-RANKL axis (66), miR-130a-5p-RUNX2-serine/threonine kinase 32A network (67), the bone morphogenetic protein (BMP)/TGF-β-RUNX2 loop (65) and the Zic family member 2-RUNX2-nucleolar and coiled-body phosphoprotein 1 signaling axis (68). To date, RUNX2 has emerged as a key mediator in the metastasis of cancers with preinvasive and promigratory behaviors in osteosarcoma, breast cancer, thyroid cancer, prostatic carcinoma and melanoma cells (69). Furthermore, numerous investigations of the molecular mechanisms of RUNX2 have revealed the mode of action in tumor metastasis and growth via activating the expression of bone matrix and adhesion proteins, matrix metalloproteinase (MMP) and angiogenic factors in cancer cells (70,71).

As mentioned above, breast cancer is composed of distinct subtypes with multiple stages during tumor progression. Of note, RUNX2 has been reported to have an important role in bone metastasis, which is the final stage of breast cancer development (72). For instance, phosphorylation of RUNX2 mediated by tyrosine kinase ABL is adequate to promote breast cancer invasion (73), while the interaction with core binding factor subunit β protein is sufficient to guide breast cancer cell invasion (53).

During tumor metastasis, cancer cells function via autophagy to recover nutrients to maintain their own survival and RUNX2 promotes autophagy by increasing acetylation of α-tubulin sub-units of microtubules, and thus promotes the metastasis of breast cancer cells (74). It has been indicated that the integrin subunit α5 (ITGA5)β3 expressed on the surface of breast cancer cells was able to anchor cells to osteoblasts by binding the tripeptide Arg-Gly-Asp motif of bone matrix proteins (75). During the aforementioned process, RUNX2 facilitated breast cancer cell recruitment and colonization in bone in an ITGA5-dependent manner (76). SET domain containing 7, histone lysine methyltransferase (SET7)/9 in breast cancer is able to activate target gene expression through histone methylation or directly act as a target via non-histone methylation. It has also been indicated that SET7/9 is able to promote multiple malignant biological behaviors in the development of breast cancer by activating RUNX2 (77). TGF-β is indispensable in normal physiological function (78) and had a complex ‘double-edged sword’ role in the occurrence and development of tumors (79). Furthermore, RUNX2 may promote bone metastasis in breast cancer cells through the activation of the TGF-β signaling pathway (80). In addition, accumulating evidence has indicated that multiple miRNAs (e.g., miR-30, miR-135, miR-203, miR-205, miR-505-3P and miR-590-3P) are adequate to inhibit the development and metastasis of breast cancer via targeting RUNX2 (23,71,8184). Taken together, abnormal expression of RUNX2 may be associated with the occurrence and progression of breast cancer, and the underlying molecular mechanisms, including target genes and the concomitant signaling pathways, remain to be fully elucidated.

BCSCs

CSCs and epithelial-mesenchymal transition (EMT) are the major elements contributing to the metastasis and recurrence of cancers (85,86). In general, CSCs are unique subpopulations of solid tumors (e.g., breast cancer, lung cancer and stomach cancer) with stem cell-related characteristics, such as self-renewal, differentiation and tumorigenic potential, which have re-emerged as a hot topic of increased interest in the field (87,88). In breast cancer, BCSCs promote angiogenesis in mammary tissues by dedifferentiating to endothelial cells and secreting proangiogenic or angiogenic factors, which collectively facilitates metastasis and therapy resistance of breast cancer (89). Of note, studies have demonstrated that a series of signaling pathways are involved in orchestrating the phenotypes of CSCs, including the Hippo, Wnt, Notch and Hedgehog pathways (90). Meanwhile, drug efflux transporters and multi-drug resistance genes expressed in BCSCs also confer resistance against conventional chemotherapeutic drugs (90).

Breast cancer and BCSCs

Despite the inspiring advancements in radiation and chemotherapies, breast cancer is still an intractable disease owing to tumor relapse and drug resistance caused by BCSCs (90). BCSCs participate in the occurrence and development of breast cancer and usually result in cancer relapse with enhanced aggressiveness (91). Although only a small proportion of breast cancer cells may lead to tumors xenografts, these tumor-initiating cells are able to reconstruct tumors with a similar heterogeneity to that of the primary tumor, which indicates the distinct stem cell-like plasticity of BCSCs (92). To date, a large number of studies have indicated the pivotal role of the different pathways involved in the regulation of BCSCs, such as Wnt, Hedgehog and Notch signaling. For instance, Katoh (93) summarized Wnt signaling cascades cross-talk with the fibroblast growth factor, Notch, Hedgehog and TGF-β/BMP signaling cascades and regulate the expression of functional CSC markers. Furthermore, Ibrahim et al (94) verified syndecan-1 as a novel biomarker for inflammatory TNBC and modulating the BCSC phenotype via orchestrating the IL-6/STAT3-Notch and EGFR signal cascades.

According to the currently recognized stem cell markers for breast cancer, BCSCs may be divided into the CD44+/CD24 and aldehyde dehydrogenase (ALDH)+ subtypes (95). The most rapidly proliferating ALDH+ BCSCs have an epithelial cell morphology, whereas the CD44+/CD24 mesenchymal subsets display declined proliferation but high invasion and metastasis (96). In general, based on the expression levels of CD44, breast cancers of different molecular subtypes show variations in the proportion of CD44+/CD24 BCSCs (97). For instance, the proportion of CD44+/CD24 BCSCs in basal-like breast cancer is significantly higher than that in luminal A and luminal B breast cancers (98), which may be accountable for the worse prognosis of basal-like breast cancer as compared with that of the luminal A and luminal B subtypes (Table II). Collectively, the existence of BCSCs has tremendously increased the metastasis, angiogenesis and therapy resistance of breast cancer, as well as the secondary tumor formation in patients. Therefore, devising therapeutic interventions with multidisciplinary strategies to target BCSCs would be of great help in boosting patients' survival rates and in increasing the sensitivity to anti-tumor drugs for breast cancer.

Table II.

BCSC markers.

Table II.

BCSC markers.

First author, yearMarkerBCSCsLocationFunction(Refs.)
Dzobo, 2021CD44+Cell membraneCell-cell interactions, cell adhesion and migration(133)
Fillmore, 2007CD24-Cell membraneCell differentiation(134)
Tomita, 2016ALDH1+CytoplasmOxidize xenobiotic and intracellular aldehydes(135)

[i] BCSC, breast cancer stem cell; ALDH1, aldehyde dehydrogenase 1.

RUNX2 and BCSCs

State-of-the-art renewal has indicated the involvement of RUNX2 in various cancer types. The pro-cancer role of RUNX2 in breast cancer is known to be related to BCSCs (7). Furthermore, several studies have indicated a potential link between RUNX2 and CD44. For instance, in colorectal cancer, RUNX2 interacts with brahma-related gene 1 to target CD44 for promoting invasion and migration of colorectal cancer cells (99). In prostate cancer cells, RUNX2 forms a complex with intracellular domain of CD44 as a co-transcriptional factor, activates the expression of metastasis-related genes, and contributes to migration and tumor formation (54).

As a surface marker of BCSCs, CD44 rather than CD24 is positively correlated with RUNX2 expression, which suggests the association of RUNX2 with CD44+/CD24 BCSCs. For instance, Zhang et al (24) found that RUNX2 promoted the malignant biological behavior of breast cancer cells by regulating the proportion of BCSCs. As a common event in breast cancer progression, PR Ser294 phosphorylation is required to maintain BCSCs fate via cooperation with the growth factor-initiated signaling pathway and key phosphor-PR targets (e.g., solute carrier family 37 member 2 and RUNX2). With the aid of the microsphere formation test, RUNX2 has been proven to act as an important driver of BCSC formation in vitro (100). In detail, RUNX2 has a critical influence in both EMT and BCSCs, and the ectopic expression of RUNX2 may subsequently induce the occurrence of EMT via the regulation of key pathways (e.g., TGF-β, Wnt) (101). Meanwhile, RUNX2 also has a critical role in CD44+/CD24 MCF10AT1 cells with BCSC characteristics (102).

Taken together, RUNX2 has a critical role in promoting the development and metastasis of breast cancer via regulating the proportion of BCSCs, which provides new insight for the further dissection of the pathogenesis and therapeutic remedies for breast cancer.

Drug resistance in breast cancer

Attributed to the rapid development of technologies, the diagnosis and treatment of breast cancer have markedly improved; however, drug resistance in breast cancer remains an issue, as the mechanisms comprise disorders in the orchestration of hormones, apoptosis and efflux pump activation, signaling pathways and oncogenes (103).

Relationship between CSCs and drug resistance

CSCs are self-renewal cells with high tumorigenic potential. CSCs are able to adapt to changes in the surrounding environment and are more resistant to radiotherapy and chemotherapy than other cells in the tumor. Several studies have indicated that the dual effects of intrinsic and extrinsic factors contributes to CSC-mediated therapy resistance.

CSCs have a very slow cell cycle and possess an anti-apoptotic machinery, DNA repair systems and persistent stemness, which lead to drug resistance during cancer treatment (104). Besides, CSC drug resistance may also be caused by external factors, such as the tumor microenvironment (TME). When the TME is always in a state of nutrition, metabolism and oxygen deficiency, it promotes the adaptation of CSCs to the TME (105). CSC drug resistance is complex. CSCs promote tumor progression, treatment resistance and disease recurrence, which may achieved by their sustained proliferation, invasion into normal tissue, promotion of angiogenesis, evasion of the immune system and resistance to conventional anticancer therapies (106,107).

BCSCs and drug resistance in breast cancer

Cancer metastasis and drug resistance currently remain major challenges in cancer therapy (106). Increasing evidence suggests that CSCs favor cancer metastasis and drug resistance, leading to relapse of cancer and death of patients (108). On the one hand, BCSCs have been considered the result of overactivation of mutant normal breast stem cells during self-renewal. On the other hand, BCSCs are resultant of the dedifferentiation of cancer cells caused by somatic mutations or microenvironmental components during treatment (109).

The proportion of BCSCs was found to be significantly increased in chemoresistant and radioresistant breast cancer cell lines and human tissues (110). Furthermore, overactivation of the anti-apoptotic PI3K signaling pathway and the antioxidant nuclear factor E2-related factor 2 signaling pathway collectively contribute to cellular resistance and resistance of BCSCs to radiation-induced ROS attack and apoptosis compared with non-BCSCs (111,112). Further mechanistic studies revealed that BCSCs with a high level of free radical scavenger expression were more tolerant to the hypoxic environment and thus conferred a radiation-resistant phenotype by reducing the accumulation of intracellular ROS (113). In addition, certain cell surface pumps in BCSCs (e.g., ATP binding cassette subfamily G member 2) are able to impair the intracellular accumulation of anticancer drugs (114). Collectively, the variations in BCSCs are responsible for increased drug resistance of breast cancer.

RUNX2 and drug resistance of breast cancers

Overexpression of RUNX2 commonly leads to reduced sensitivity of cancer cells (e.g., osteosarcoma) to chemotherapy, which may thus be used as a reliable marker for the evaluation of chemotherapy resistance (115). Conversely, loss of RUNX2 expression is adequate to increase the sensitivity of osteosarcoma cells to the chemotherapeutic agent doxorubicin (116). First, Sugimoto et al (117) found that silencing RUNX2 was able to enhance the sensitivity of AsPC-1 cells in P53-deficient human pancreatic cancer to gemcitabine (GEM) by stimulating Tap63 (isoform of p63 gene)-mediated cell death. Furthermore, Ozaki et al (118) verified that increased Tap73-dependent cell death mediated by RUNX2 depletion enhanced the sensitivity of P53-mutant pancreatic cancer MiaPaCa-2 cells to GEM. In addition, RUNX2 was found to weaken the proapoptotic activity and enhance the sensitivity to GEM drugs of P53-mutant Panc-1 pancreatic cancer cells via suppressing Tap63 expression (118). These findings demonstrate that RUNX2 is associated with drug sensitivity to GEM in pancreatic cancer cells lacking functional P53.

In breast cancer, Tamoxifen (TAM) resistance is responsible for a large proportion of breast cancer-associated mortalities. Jeselsohn et al (119) found that RUNX2 was able to interact with ERα to directly induce SOX9 transcription and reduce the sensitivity of breast cancer cells to TAM. Of note, Geter et al (120) identified RUNX2 as the only RUNX family member that was transcriptionally or translationally upregulated in both TAM-resistant LCC9 cells and patient-derived xenograft derived from tamoxifen-resistant cell lines, suggesting that RUNX2 has a core role in the drug resistance of breast cancer. The study's results revealed that RUNX2 promoted the metastasis of breast cancer, and knockdown of RUNX2 was able to significantly restore the sensitivity of breast cancer to TAM (120). Furthermore, Othman et al (121) found that silencing of RUNX2 increased the sensitivity of breast cancer cells to microtubule-targeting agents. Wang et al (122) demonstrated that serum miR-4530 may sensitize breast cancer to taxane- and anthracycline-based neoadjuvant chemotherapy by suppressing RUNX2. A recent study revealed that RUNX2 directly targeted MMP1 and facilitates aggressiveness and chemoresistance of TNBC cells (21).

However, the specific mechanisms of RUNX2-associated drug resistance in breast cancer still require confirmation and further elucidation. Systematic and detailed dissection of the underlying mechanisms will contribute to conquering drug resistance and provide new references for the treatment of breast cancer (Fig. 1).

Discussion

RUNX2 has been recognized as a master transcription factor for osteogenesis and bone metastases of invasive breast cancer (72). In highly metastatic breast cancer, RUNX2 phosphorylation has associations with the invasive capacity of MDA-MB-231 cells by modulating the activity of tyrosine kinase ABL, which is involved in the activation of the classical BMP-SMAD signaling cascades (17). It was further indicated that RUNX2 facilitates the aggressiveness and chemoresistance of TNBC and the concomitant tumorigenesis and cancer progression via activating MMP1, which thus provides a basis for developing RUNX2-MMP1 axis-based novel candidates for breast cancer diagnostics (21).

Longitudinal studies have highlighted the potential utility of RUNX2 as a pivotal contributor to bone-specific metastasis during breast cancer. For instance, Li et al (76) verified the facilitating effect of RUNX2 and the targeted ITGA5 during the adhesion and attraction of breast cancer cells, as well as osteotropism and bone colonization. In contrast to the aforementioned studies indicating the critical role of RUNX2 in bone-specific metastasis or progression of breast cancer, the present review mainly focused on the latest updates on the interaction between RUNX2 and BCSCs and their biofunctions, which contribute to the aggressiveness and drug resistance of breast cancer. Furthermore, the systematic and detailed investigation of the RUNX2-BCSCs axis in breast cancer and knowledge regarding its utility in diagnosis and treatment are still far from satisfactory and further preclinical and clinical investigations are required in the future.

Of note, current progress has also highlighted the potential application of developing computational models based on the data fusion paradigm for identification of the disease-related non-coding RNAs (ncRNAs) as a future direction in disease assessment (123,124). For instance, Huang et al (123) summarized the advances in miRNAs and algorithm design for complex diseases by computational models, which may help overcome the obstacles to disease prediction. Simultaneously, Chen et al (125) and Wang et al (126) put forward the limitations and future directions of the disease prediction utility of long ncRNAs (lncRNAs) or circular RNAs (circRNAs) for complex diseases by combing experimental technology with computational prediction algorithms. Similarly, a series of ncRNAs have been identified to be linked to the disease progression of breast cancer or TNBC, such as the miR-20a-5p/high mobility group AT-hook 2 axis, lncRNA-CDC6/miRNA-215 and circRNA_002502/miR-182-5p/forkhead box O3a axis (127130). In addition, Yin et al (131) performed a weighted gene co-expression network analysis and competing endogenous RNA network analysis based on the Cancer Genome Atlas database and identified the prospective association of 3,301 key modules and 453 genes with breast cancer prognosis. Therewith, it would be of great interest and importance to perform further studies to predict the association of RUNX2 and BCSCs with breast cancer development and drug resistance based on powerful computational model construction, which may also benefit the development of the framework of oncology diagnostics and therapeutics from a different perspective in the future. The present review mainly summarized the research progress on RUNX2 and BCSC in terms of breast cancer progression and diagnosis, may benefit the identification of key mechanisms of breast cancer and supply references for the concomitant anticancer treatment and drug development in the future.

As a malignant tumor type with high morbidity and mortality rates, breast cancer persistently endangers the health and lives of females. While chemotherapy remains the major remedy for breast cancer, drug resistance severely limits the efficacy and usually results in tumor metastasis and recurrence.

In recent years, RUNX2 in breast cancer has attracted considerable attention, as increasing evidence indicated its pivotal role in tumor development and metastasis, as well as its association with BCSCs-related drug resistance. Based on the current literature, it may be presumed that RUNX2 enhances drug resistance in breast cancer by regulating the proportion and biofunction of BCSCs, which will vastly enhance the current understanding of the pathogenesis and provide reliable targets for clinical treatment and drug development in the future.

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (grant nos. 82173377 and 81302319 to FL), Major Research Project of the Education Department of Anhui Province (grant no. KJ2018ZD018), the National Natural Science Foundation of China (grant no. 82260031), the Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences (grant no. 2019PT320005) and the 2021 Central-Guided Local Science and Technology Development Fund (grant no. ZYYDDFFZZJ-1).

Availability of data and materials

Not applicable.

Authors' contributions

WS and FL designed the study, performed data analysis and interpretation, wrote the manuscript and gave final approval of the manuscript. WS and CK performed the literature search and wrote the manuscript. FL and LZ reviewed the manuscript. All authors contributed to the review article, and have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Akram M, Iqbal M, Daniyal M and Khan AU: Awareness and current knowledge of breast cancer. Biol Res. 50:332017. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2021. CA Cancer J Clin. 71:7–33. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Li X, Yang J, Peng L, Sahin AA, Huo L, Ward KC, O'Regan R, Torres MA and Meisel JL: Triple-negative breast cancer has worse overall survival and cause-specific survival than non-triple-negative breast cancer. Breast Cancer Res Treat. 161:279–287. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Gomathi K, Akshaya N, Srinaath N, Moorthi A and Selvamurugan N: Regulation of Runx2 by post-translational modifications in osteoblast differentiation. Life Sci. 245:1173892020. View Article : Google Scholar : PubMed/NCBI

5 

Liu DD, Zhang CY, Liu Y, Li J, Wang YX and Zheng SG: RUNX2 regulates osteoblast differentiation via the BMP4 signaling pathway. J Dent Res. 101:1227–1237. 2022. View Article : Google Scholar : PubMed/NCBI

6 

Komori T: Runx2, a multifunctional transcription factor in skeletal development. J Cell Biochem. 87:1–8. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Li Y, Ge C and Franceschi RT: Role of Runx2 in prostate development and stem cell function. Prostate. 81:231–241. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Westendorf JJ: Transcriptional co-repressors of Runx2. J Cell Biochem. 98:54–64. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Zhang L, Wei Y, Chi Y, Liu D, Yang S, Han Z and Li Z: Two-step generation of mesenchymal stem/stromal cells from human pluripotent stem cells with reinforced efficacy upon osteoarthritis rabbits by HA hydrogel. Cell Biosci. 11:62021. View Article : Google Scholar : PubMed/NCBI

10 

Zhang L, Wang H, Liu C, Wu Q, Su P, Wu D, Guo J, Zhou W, Xu Y, Shi L and Zhou J: MSX2 initiates and accelerates mesenchymal stem/stromal cell specification of hPSCs by regulating TWIST1 and PRAME. Stem Cell Reports. 11:497–513. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Lu J and Zhang H, Pan J, Hu Z, Liu L, Liu Y, Yu X, Bai X, Cai D and Zhang H: Fargesin ameliorates osteoarthritis via macrophage reprogramming by downregulating MAPK and NF-κB pathways. Arthritis Res Ther. 23:1422021. View Article : Google Scholar : PubMed/NCBI

12 

Kim HJ, Kim WJ and Ryoo HM: Post-translational regulations of transcriptional activity of RUNX2. Mol Cells. 43:160–167. 2020.PubMed/NCBI

13 

Zhang Y and Duan X: A novel 90-kbp deletion of RUNX2 associated with cleidocranial dysplasia. Genes (Basel). 13:11282022. View Article : Google Scholar : PubMed/NCBI

14 

Ukkat J, Hoang-Vu C, Trojanowicz B and Rebelo A: Osteocalcin, osteopontin and RUNX2 expression in patients' leucocytes with arteriosclerosis. Diseases. 9:192021. View Article : Google Scholar : PubMed/NCBI

15 

Zhang X, Ren Z, Liu B and Wei S: RUNX2 mediates renal cell carcinoma invasion through calpain2. Biol Pharm Bull. 45:1653–1659. 2022. View Article : Google Scholar : PubMed/NCBI

16 

Wysokinski D, Blasiak J and Pawlowska E: Role of RUNX2 in breast carcinogenesis. Int J Mol Sci. 16:20969–20993. 2015. View Article : Google Scholar : PubMed/NCBI

17 

He F, Matsumoto Y, Asano Y, Yamamura Y, Katsuyama T, La Rose J, Tomonobu N, Komalasari NLGY, Sakaguchi M, Rottapel R and Wada J: RUNX2 phosphorylation by tyrosine kinase ABL promotes breast cancer invasion. Front Oncol. 11:6652732021. View Article : Google Scholar : PubMed/NCBI

18 

Song X, Liu J, Liu B, Piao C, Kong C and Li Z: RUNX2 interacts with SCD1 and activates Wnt/β-catenin signaling pathway to promote the progression of clear cell renal cell carcinoma. Cancer Med. Oct 6–2022.(Epub ahead of print).

19 

Guo Z, Zhou K, Wang Q, Huang Y, Ji J, Peng Y, Zhang X, Zheng T, Zhang Z, Chong D and Yang Z: The transcription factor RUNX2 fuels YAP1 signaling and gastric cancer tumorigenesis. Cancer Sci. 112:3533–3544. 2021. View Article : Google Scholar : PubMed/NCBI

20 

Li N, Luo D, Hu X, Luo W, Lei G, Wang Q, Zhu T, Gu J, Lu Y and Zheng Q: RUNX2 and osteosarcoma. Anticancer Agents Med Chem. 15:881–887. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Si W, Xu X, Wan L, Lv F, Wei W, Xu X, Li W, Huang D, Zhang L and Li F: RUNX2 facilitates aggressiveness and chemoresistance of triple negative breast cancer cells via activating MMP1. Front Oncol. 12:9960802022. View Article : Google Scholar : PubMed/NCBI

22 

Ferrari N, McDonald L, Morris JS, Cameron ER and Blyth K: RUNX2 in mammary gland development and breast cancer. J Cell Physiol. 228:1137–1142. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Zhang L, Liu L, Xu X, He X, Wang G, Fan C, Zheng Q and Li F: miR-205/RunX2 axis negatively regulates CD44+/CD24 breast cancer stem cell activity. Am J Cancer Res. 10:1871–1887. 2020.PubMed/NCBI

24 

Zhang P, Liu L, Zhang L, He X, Xu X, Lu Y and Li F: Runx2 is required for activity of CD44+/CD24−/low breast cancer stem cell in breast cancer development. Am J Transl Res. 12:2305–2318. 2020.PubMed/NCBI

25 

Kudela E, Samec M, Koklesova L, Liskova A, Kubatka P, Kozubik E, Rokos T, Pribulova T, Gabonova E, Smolar M and Biringer K: miRNA expression profiles in luminal A breast cancer-implications in biology, prognosis, and prediction of response to hormonal treatment. Int J Mol Sci. 21:76912020. View Article : Google Scholar : PubMed/NCBI

26 

Harbeck N and Gnant M: Breast cancer. Lancet. 389:1134–1150. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Ades F, Zardavas D, Bozovic-Spasojevic I, Pugliano L, Fumagalli D, de Azambuja E, Viale G, Sotiriou C and Piccart M: Luminal B breast cancer: Molecular characterization, clinical management, and future perspectives. J Clin Oncol. 32:2794–2803. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Loibl S and Gianni L: HER2-positive breast cancer. Lancet. 389:2415–2429. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Alexandrou S, George SM, Ormandy CJ, Lim E, Oakes SR and Caldon CE: The proliferative and apoptotic landscape of basal-like breast cancer. Int J Mol Sci. 20:6672019. View Article : Google Scholar : PubMed/NCBI

30 

Perou CM, Sørlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, Pollack JR, Ross DT, Johnsen H, Akslen LA, et al: Molecular portraits of human breast tumours. Nature. 406:747–752. 2000. View Article : Google Scholar : PubMed/NCBI

31 

Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH, Allred DC, Bartlett JM, Bilous M, Fitzgibbons P, et al: Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American society of clinical oncology/college of American pathologists clinical practice guideline update. Arch Pathol Lab Med. 138:241–256. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Dagogo-Jack I and Shaw AT: Tumour heterogeneity and resistance to cancer therapies. Nat Rev Clin Oncol. 15:81–94. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Cardoso F, Kyriakides S, Ohno S, Penault-Llorca F, Poortmans P, Rubio IT, Zackrisson S and Senkus E; ESMO Guidelines Committee. Electronic address, : simpleclinicalguidelines@esmo.org: Early breast cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up†. Ann Oncol. 30:1194–1220. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Tosello G, Torloni MR, Mota BS, Neeman T and Riera R: Breast surgery for metastatic breast cancer. Cochrane Database Syst Rev. 3:CD0112762018.PubMed/NCBI

35 

Maughan KL, Lutterbie MA and Ham PS: Treatment of breast cancer. Am Fam Physician. 81:1339–1346. 2010.PubMed/NCBI

36 

Pondé NF, Zardavas D and Piccart M: Progress in adjuvant systemic therapy for breast cancer. Nat Rev Clin Oncol. 16:27–44. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Waks AG and Winer EP: Breast cancer treatment: A review. JAMA. 321:288–300. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Abderrahman B and Jordan VC: Telling details of breast-cancer recurrence. Nature. 553:1552018. View Article : Google Scholar

39 

Miyoshi H, Shimizu K, Kozu T, Maseki N, Kaneko Y and Ohki M: t(8;21) breakpoints on chromosome 21 in acute myeloid leukemia are clustered within a limited region of a single gene, AML1. Proc Natl Acad Sci USA. 88:10431–10434. 1991. View Article : Google Scholar : PubMed/NCBI

40 

Schlegelberger B and Heller PG: RUNX1 deficiency (familial platelet disorder with predisposition to myeloid leukemia, FPDMM). Semin Hematol. 54:75–80. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Ito Y, Bae SC and Chuang LS: The RUNX family: developmental regulators in cancer. Nat Rev Cancer. 15:81–95. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Cecconi D, Brandi J, Manfredi M, Serena M, Dalle Carbonare L, Deiana M, Cheri S, Parolini F, Gandini A, Marchetto G, et al: Runx2 stimulates neoangiogenesis through the Runt domain in melanoma. Sci Rep. 9:80522019. View Article : Google Scholar : PubMed/NCBI

43 

Vitale E, Sauta E, Gugnoni M, Torricelli F, Manicardi V and Ciarrocchi A: A multimodal integrative approach to model transcriptional addiction of thyroid cancer on RUNX2. Cancer Commun (Lond). 42:892–896. 2022. View Article : Google Scholar : PubMed/NCBI

44 

Guan Y, Zhang Y, Hao L and Nie Z: CircRNA_102272 promotes cisplatin-resistance in hepatocellular carcinoma by decreasing MiR-326 targeting of RUNX2. Cancer Manag Res. 12:12527–12534. 2020. View Article : Google Scholar : PubMed/NCBI

45 

Matthijssens F, Sharma ND, Nysus M, Nickl CK, Kang H, Perez DR, Lintermans B, Van Loocke W, Roels J, Peirs S, et al: RUNX2 regulates leukemic cell metabolism and chemotaxis in high-risk T cell acute lymphoblastic leukemia. J Clin Invest. 131:e1415662021. View Article : Google Scholar : PubMed/NCBI

46 

Ji Q, Cai G, Liu X, Zhang Y, Wang Y, Zhou L, Sui H and Li Q: MALAT1 regulates the transcriptional and translational levels of proto-oncogene RUNX2 in colorectal cancer metastasis. Cell Death Dis. 10:3782019. View Article : Google Scholar : PubMed/NCBI

47 

Zhang F, Su T and Xiao M: RUNX3-regulated circRNA METTL3 inhibits colorectal cancer proliferation and metastasis via miR-107/PER3 axis. Cell Death Dis. 13:5502022. View Article : Google Scholar : PubMed/NCBI

48 

Liu H, Xue Q, Cai H, Jiang X, Cao G, Chen T, Chen Y and Wang D: RUNX3-mediated circDYRK1A inhibits glutamine metabolism in gastric cancer by up-regulating microRNA-889-3p-dependent FBXO4. J Transl Med. 20:1202022. View Article : Google Scholar : PubMed/NCBI

49 

Wang L, Tang W, Yang S, He P, Wang J, Gaedcke J, Ströbel P, Azizian A, Ried T, Gaida MM, et al: NO/RUNX3/kynurenine metabolic signaling enhances disease aggressiveness in pancreatic cancer. Int J Cancer. 146:3160–3169. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Mevel R, Draper JE, Lie-A-Ling M, Kouskoff V and Lacaud G: RUNX transcription factors: Orchestrators of development. Development. 146:dev1482962019. View Article : Google Scholar : PubMed/NCBI

51 

Komori T: Regulation of proliferation, differentiation and functions of osteoblasts by Runx2. Int J Mol Sci. 20:16942019. View Article : Google Scholar : PubMed/NCBI

52 

Nagata K, Hojo H, Chang SH, Okada H, Yano F, Chijimatsu R, Omata Y, Mori D, Makii Y, Kawata M, et al: Runx2 and Runx3 differentially regulate articular chondrocytes during surgically induced osteoarthritis development. Nat Commun. 13:61872022. View Article : Google Scholar : PubMed/NCBI

53 

Villanueva F, Araya H, Briceño P, Varela N, Stevenson A, Jerez S, Tempio F, Chnaiderman J, Perez C, Villarroel M, et al: The cancer-related transcription factor RUNX2 modulates expression and secretion of the matricellular protein osteopontin in osteosarcoma cells to promote adhesion to endothelial pulmonary cells and lung metastasis. J Cell Physiol. 234:13659–13679. 2019. View Article : Google Scholar : PubMed/NCBI

54 

Senbanjo LT, AlJohani H, Majumdar S and Chellaiah MA: Characterization of CD44 intracellular domain interaction with RUNX2 in PC3 human prostate cancer cells. Cell Commun Signal. 17:802019. View Article : Google Scholar : PubMed/NCBI

55 

Yin X, Teng X, Ma T, Yang T, Zhang J, Huo M, Liu W, Yang Y, Yuan B, Yu H, et al: RUNX2 recruits the NuRD(MTA1)/CRL4B complex to promote breast cancer progression and bone metastasis. Cell Death Differ. 29:2203–2217. 2022. View Article : Google Scholar : PubMed/NCBI

56 

Chan WCW, Tan Z, To MKT and Chan D: Regulation and role of transcription factors in osteogenesis. Int J Mol Sci. 22:54452021. View Article : Google Scholar : PubMed/NCBI

57 

Coffman JA: Runx transcription factors and the developmental balance between cell proliferation and differentiation. Cell Biol Int. 27:315–324. 2003. View Article : Google Scholar : PubMed/NCBI

58 

Han X, Feng J, Guo T, Loh YE, Yuan Y, Ho TV, Cho CK, Li J, Jing J, Janeckova E, et al: Runx2-Twist1 interaction coordinates cranial neural crest guidance of soft palate myogenesis. Elife. 10:e623872021. View Article : Google Scholar : PubMed/NCBI

59 

Shu HS, Liu YL, Tang XT, Zhang XS, Zhou B, Zou W and Zhou BO: Tracing the skeletal progenitor transition during postnatal bone formation. Cell Stem Cell. 28:2122–2136.e3. 2021. View Article : Google Scholar : PubMed/NCBI

60 

Liu TM and Lee EH: Transcriptional regulatory cascades in Runx2-dependent bone development. Tissue Eng Part B Rev. 19:254–263. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Qin X, Jiang Q, Nagano K, Moriishi T, Miyazaki T, Komori H, Ito K, Mark KV, Sakane C, Kaneko H and Komori T: Runx2 is essential for the transdifferentiation of chondrocytes into osteoblasts. PLoS Genet. 16:e10091692020. View Article : Google Scholar : PubMed/NCBI

62 

Owens TW, Rogers RL, Best S, Ledger A, Mooney AM, Ferguson A, Shore P, Swarbrick A, Ormandy CJ, Simpson PT, et al: Runx2 is a novel regulator of mammary epithelial cell fate in development and breast cancer. Cancer Res. 74:5277–5286. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Inman CK and Shore P: The osteoblast transcription factor Runx2 is expressed in mammary epithelial cells and mediates osteopontin expression. J Biol Chem. 278:48684–48689. 2003. View Article : Google Scholar : PubMed/NCBI

64 

Sato M, Morii E, Komori T, Kawahata H, Sugimoto M, Terai K, Shimizu H, Yasui T, Ogihara H, Yasui N, et al: Transcriptional regulation of osteopontin gene in vivo by PEBP2alphaA/CBFA1 and ETS1 in the skeletal tissues. Oncogene. 17:1517–1525. 1998. View Article : Google Scholar : PubMed/NCBI

65 

Pratap J, Lian JB, Javed A, Barnes GL, van Wijnen AJ, Stein JL and Stein GS: Regulatory roles of Runx2 in metastatic tumor and cancer cell interactions with bone. Cancer Metastasis Rev. 25:589–600. 2006. View Article : Google Scholar : PubMed/NCBI

66 

Kim B, Kim H, Jung S, Moon A, Noh DY, Lee ZH, Kim HJ and Kim HH: A CTGF-RUNX2-RANKL axis in breast and prostate cancer cells promotes tumor progression in bone. J Bone Miner Res. 35:155–166. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Ma F, Xie Y, Lei Y, Kuang Z and Liu X: The microRNA-130a-5p/RUNX2/STK32A network modulates tumor invasive and metastatic potential in non-small cell lung cancer. BMC Cancer. 20:5802020. View Article : Google Scholar : PubMed/NCBI

68 

Wu CY, Li L, Chen SL, Yang X, Zhang CZ and Cao Y: A Zic2/Runx2/NOLC1 signaling axis mediates tumor growth and metastasis in clear cell renal cell carcinoma. Cell Death Dis. 12:3192021. View Article : Google Scholar : PubMed/NCBI

69 

Cohen-Solal KA, Boregowda RK and Lasfar A: RUNX2 and the PI3K/AKT axis reciprocal activation as a driving force for tumor progression. Mol Cancer. 14:1372015. View Article : Google Scholar : PubMed/NCBI

70 

Tandon M, Gokul K, Ali SA, Chen Z, Lian J, Stein GS and Pratap J: Runx2 mediates epigenetic silencing of the bone morphogenetic protein-3B (BMP-3B/GDF10) in lung cancer cells. Mol Cancer. 11:272012. View Article : Google Scholar : PubMed/NCBI

71 

Pranavkrishna S, Sanjeev G, Akshaya RL, Rohini M and Selvamurugan N: A computational approach on studying the regulation of TGF-β1-stimulated Runx2 expression by MicroRNAs in human breast cancer cells. Comput Biol Med. 137:1048232021. View Article : Google Scholar : PubMed/NCBI

72 

Vishal M, Swetha R, Thejaswini G, Arumugam B and Selvamurugan N: Role of Runx2 in breast cancer-mediated bone metastasis. Int J Biol Macromol. 99:608–614. 2017. View Article : Google Scholar : PubMed/NCBI

73 

Fang Y, Xue Z, Zhao L, Yang X, Yang Y, Zhou X, Feng S and Chen K: Calycosin stimulates the osteogenic differentiation of rat calvarial osteoblasts by activating the IGF1R/PI3K/Akt signaling pathway. Cell Biol Int. 43:323–332. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Tandon M, Othman AH, Ashok V, Stein GS and Pratap J: The role of Runx2 in facilitating autophagy in metastatic breast cancer cells. J Cell Physiol. 233:559–571. 2018. View Article : Google Scholar : PubMed/NCBI

75 

Schneider JG, Amend SR and Weilbaecher KN: Integrins and bone metastasis: Integrating tumor cell and stromal cell interactions. Bone. 48:54–65. 2011. View Article : Google Scholar : PubMed/NCBI

76 

Li XQ, Lu JT, Tan CC, Wang QS and Feng YM: RUNX2 promotes breast cancer bone metastasis by increasing integrin α5-mediated colonization. Cancer Lett. 380:78–86. 2016. View Article : Google Scholar : PubMed/NCBI

77 

Si W, Zhou J, Zhao Y, Zheng J and Cui L: SET7/9 promotes multiple malignant processes in breast cancer development via RUNX2 activation and is negatively regulated by TRIM21. Cell Death Dis. 11:1512020. View Article : Google Scholar : PubMed/NCBI

78 

Morikawa M, Derynck R and Miyazono K: TGF-β and the TGF-β family: Context-dependent roles in cell and tissue physiology. Cold Spring Harb Perspect Biol. 8:a0218732016. View Article : Google Scholar : PubMed/NCBI

79 

Larson C, Oronsky B, Carter CA, Oronsky A, Knox SJ, Sher D and Reid TR: TGF-beta: A master immune regulator. Expert Opin Ther Targets. 24:427–438. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Li XQ, Du X, Li DM, Kong PZ, Sun Y, Liu PF, Wang QS and Feng YM: ITGBL1 is a Runx2 transcriptional target and promotes breast cancer bone metastasis by activating the TGFβ signaling pathway. Cancer Res. 75:3302–3313. 2015. View Article : Google Scholar : PubMed/NCBI

81 

Taipaleenmäki H, Browne G, Akech J, Zustin J, van Wijnen AJ, Stein JL, Hesse E, Stein GS and Lian JB: Targeting of Runx2 by miR-135 and miR-203 impairs progression of breast cancer and metastatic bone disease. Cancer Res. 75:1433–1444. 2015. View Article : Google Scholar : PubMed/NCBI

82 

Croset M, Pantano F, Kan CWS, Bonnelye E, Descotes F, Alix-Panabières C, Lecellier CH, Bachelier R, Allioli N, Hong SS, et al: miRNA-30 family members inhibit breast cancer invasion, osteomimicry, and bone destruction by directly targeting multiple bone metastasis-associated genes. Cancer Res. 78:5259–5273. 2018. View Article : Google Scholar : PubMed/NCBI

83 

Zhao P, Guan H, Dai Z, Ma Y, Zhao Y and Liu D: Long noncoding RNA DLX6-AS1 promotes breast cancer progression via miR-505-3p/RUNX2 axis. Eur J Pharmacol. 865:1727782019. View Article : Google Scholar : PubMed/NCBI

84 

Rohini M, Gokulnath M, Miranda PJ and Selvamurugan N: miR-590-3p inhibits proliferation and promotes apoptosis by targeting activating transcription factor 3 in human breast cancer cells. Biochimie. 154:10–18. 2018. View Article : Google Scholar : PubMed/NCBI

85 

Batlle E and Clevers H: Cancer stem cells revisited. Nat Med. 23:1124–1134. 2017. View Article : Google Scholar : PubMed/NCBI

86 

Babaei G, Aziz SGG and Jaghi NZZ: EMT, cancer stem cells and autophagy; the three main axes of metastasis. Biomed Pharmacother. 133:1109092021. View Article : Google Scholar : PubMed/NCBI

87 

Peitzsch C, Tyutyunnykova A, Pantel K and Dubrovska A: Cancer stem cells: The root of tumor recurrence and metastases. Semin Cancer Biol. 44:10–24. 2017. View Article : Google Scholar : PubMed/NCBI

88 

Ishiguro T, Ohata H, Sato A, Yamawaki K, Enomoto T and Okamoto K: Tumor-derived spheroids: Relevance to cancer stem cells and clinical applications. Cancer Sci. 108:283–289. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Baccelli I, Schneeweiss A, Riethdorf S, Stenzinger A, Schillert A, Vogel V, Klein C, Saini M, Bäuerle T, Wallwiener M, et al: Identification of a population of blood circulating tumor cells from breast cancer patients that initiates metastasis in a xenograft assay. Nat Biotechnol. 31:539–544. 2013. View Article : Google Scholar : PubMed/NCBI

90 

Butti R, Gunasekaran VP, Kumar TVS, Banerjee P and Kundu GC: Breast cancer stem cells: Biology and therapeutic implications. Int J Biochem Cell Biol. 107:38–52. 2019. View Article : Google Scholar : PubMed/NCBI

91 

Dittmer J: Breast cancer stem cells: Features, key drivers and treatment options. Semin Cancer Biol. 53:59–74. 2018. View Article : Google Scholar : PubMed/NCBI

92 

Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ and Clarke MF: Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA. 100:3983–3988. 2003. View Article : Google Scholar : PubMed/NCBI

93 

Katoh M: Canonical and non-canonical WNT signaling in cancer stem cells and their niches: Cellular heterogeneity, omics reprogramming, targeted therapy and tumor plasticity (review). Int J Oncol. 51:1357–1369. 2017. View Article : Google Scholar : PubMed/NCBI

94 

Ibrahim SA, Gadalla R, El-Ghonaimy EA, Samir O, Mohamed HT, Hassan H, Greve B, El-Shinawi M, Mohamed MM and Götte M: Syndecan-1 is a novel molecular marker for triple negative inflammatory breast cancer and modulates the cancer stem cell phenotype via the IL-6/STAT3, Notch and EGFR signaling pathways. Mol Cancer. 16:572017. View Article : Google Scholar : PubMed/NCBI

95 

Colacino JA, Azizi E, Brooks MD, Harouaka R, Fouladdel S, McDermott SP, Lee M, Hill D, Madden J, Boerner J, et al: Heterogeneity of human breast stem and progenitor cells as revealed by transcriptional profiling. Stem Cell Reports. 10:1596–1609. 2018. View Article : Google Scholar : PubMed/NCBI

96 

Liu S, Cong Y, Wang D, Sun Y, Deng L, Liu Y, Martin-Trevino R, Shang L, McDermott SP, Landis MD, et al: Breast cancer stem cells transition between epithelial and mesenchymal states reflective of their normal counterparts. Stem Cell Reports. 2:78–91. 2013. View Article : Google Scholar : PubMed/NCBI

97 

Zöller M: CD44: Can a cancer-initiating cell profit from an abundantly expressed molecule? Nat Rev Cancer. 11:254–267. 2011. View Article : Google Scholar : PubMed/NCBI

98 

Vikram R, Chou WC, Hung SC and Shen CY: Tumorigenic and metastatic role of CD44−/low/CD24−/low cells in luminal breast cancer. Cancers (Basel). 12:12392020. View Article : Google Scholar : PubMed/NCBI

99 

Yan X, Han D, Chen Z, Han C, Dong W, Han L, Zou L, Zhang J, Liu Y and Chai J: RUNX2 interacts with BRG1 to target CD44 for promoting invasion and migration of colorectal cancer cells. Cancer Cell Int. 20:5052020. View Article : Google Scholar : PubMed/NCBI

100 

Knutson TP, Truong TH, Ma S, Brady NJ, Sullivan ME, Raj G, Schwertfeger KL and Lange CA: Posttranslationally modified progesterone receptors direct ligand-specific expression of breast cancer stem cell-associated gene programs. J Hematol Oncol. 10:892017. View Article : Google Scholar : PubMed/NCBI

101 

Valenti MT, Serafini P, Innamorati G, Gili A, Cheri S, Bassi C and Dalle Carbonare L: Runx2 expression: A mesenchymal stem marker for cancer. Oncol Lett. 12:4167–4172. 2016. View Article : Google Scholar : PubMed/NCBI

102 

Fritz AJ, Hong D, Boyd J, Kost J, Finstaad KH, Fitzgerald MP, Hanna S, Abuarqoub AH, Malik M, Bushweller J, et al: RUNX1 and RUNX2 transcription factors function in opposing roles to regulate breast cancer stem cells. J Cell Physiol. 235:7261–7272. 2020. View Article : Google Scholar : PubMed/NCBI

103 

Muley H, Fadó R, Rodriguez-Rodriguez R and Casals N: Drug uptake-based chemoresistance in breast cancer treatment. Biochem Pharmacol. 177:1139592020. View Article : Google Scholar : PubMed/NCBI

104 

Dingwall S, Lee JB, Guezguez B, Fiebig A, McNicol J, Boreham D, Collins TJ and Bhatia M: Neoplastic human embryonic stem cells as a model of radiation resistance of human cancer stem cells. Oncotarget. 6:22258–22269. 2015. View Article : Google Scholar : PubMed/NCBI

105 

Najafi M, Mortezaee K and Majidpoor J: Cancer stem cell (CSC) resistance drivers. Life Sci. 234:1167812019. View Article : Google Scholar : PubMed/NCBI

106 

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

107 

Garcia-Mayea Y, Mir C, Masson F, Paciucci R and LLeonart ME: Insights into new mechanisms and models of cancer stem cell multidrug resistance. Semin Cancer Biol. 60:166–180. 2020. View Article : Google Scholar : PubMed/NCBI

108 

Smith AG and Macleod KF: Autophagy, cancer stem cells and drug resistance. J Pathol. 247:708–718. 2019. View Article : Google Scholar : PubMed/NCBI

109 

Pece S, Tosoni D, Confalonieri S, Mazzarol G, Vecchi M, Ronzoni S, Bernard L, Viale G, Pelicci PG and Di Fiore PP: Biological and molecular heterogeneity of breast cancers correlates with their cancer stem cell content. Cell. 140:62–73. 2010. View Article : Google Scholar : PubMed/NCBI

110 

Phillips TM, McBride WH and Pajonk F: The response of CD24(−/low)/CD44+ breast cancer-initiating cells to radiation. J Natl Cancer Inst. 98:1777–1785. 2006. View Article : Google Scholar : PubMed/NCBI

111 

Ryoo IG, Choi BH and Kwak MK: Activation of NRF2 by p62 and proteasome reduction in sphere-forming breast carcinoma cells. Oncotarget. 6:8167–8184. 2015. View Article : Google Scholar : PubMed/NCBI

112 

Bai J, Chen WB, Zhang XY, Kang XN, Jin LJ, Zhang H and Wang ZY: HIF-2α regulates CD44 to promote cancer stem cell activation in triple-negative breast cancer via PI3K/AKT/mTOR signaling. World J Stem Cells. 12:87–99. 2020. View Article : Google Scholar : PubMed/NCBI

113 

Bai X, Ni J, Beretov J, Graham P and Li Y: Cancer stem cell in breast cancer therapeutic resistance. Cancer Treat Rev. 69:152–163. 2018. View Article : Google Scholar : PubMed/NCBI

114 

Leccia F, Del Vecchio L, Mariotti E, Di Noto R, Morel AP, Puisieux A, Salvatore F and Ansieau S: ABCG2, a novel antigen to sort luminal progenitors of BRCA1- breast cancer cells. Mol Cancer. 13:2132014. View Article : Google Scholar : PubMed/NCBI

115 

Sadikovic B, Thorner P, Chilton-Macneill S, Martin JW, Cervigne NK, Squire J and Zielenska M: Expression analysis of genes associated with human osteosarcoma tumors shows correlation of RUNX2 overexpression with poor response to chemotherapy. BMC Cancer. 10:2022010. View Article : Google Scholar : PubMed/NCBI

116 

Roos A, Satterfield L, Zhao S, Fuja D, Shuck R, Hicks MJ, Donehower LA and Yustein JT: Loss of Runx2 sensitises osteosarcoma to chemotherapy-induced apoptosis. Br J Cancer. 113:1289–1297. 2015. View Article : Google Scholar : PubMed/NCBI

117 

Sugimoto H, Nakamura M, Yoda H, Hiraoka K, Shinohara K, Sang M, Fujiwara K, Shimozato O, Nagase H and Ozaki T: Silencing of RUNX2 enhances gemcitabine sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells through the stimulation of TAp63-mediated cell death. Cell Death Discov. 1:150102015. View Article : Google Scholar : PubMed/NCBI

118 

Ozaki T, Nakamura M, Ogata T, Sang M, Yoda H, Hiraoka K, Sang M and Shimozato O: Depletion of pro-oncogenic RUNX2 enhances gemcitabine (GEM) sensitivity of p53-mutated pancreatic cancer Panc-1 cells through the induction of pro-apoptotic TAp63. Oncotarget. 7:71937–71950. 2016. View Article : Google Scholar : PubMed/NCBI

119 

Jeselsohn R, Cornwell M, Pun M, Buchwalter G, Nguyen M, Bango C, Huang Y, Kuang Y, Paweletz C, Fu X, et al: Embryonic transcription factor SOX9 drives breast cancer endocrine resistance. Proc Natl Acad Sci USA. 114:E4482–E4491. 2017. View Article : Google Scholar : PubMed/NCBI

120 

Geter PA, Ernlund AW, Bakogianni S, Alard A, Arju R, Giashuddin S, Gadi A, Bromberg J and Schneider RJ: Hyperactive mTOR and MNK1 phosphorylation of eIF4E confer tamoxifen resistance and estrogen independence through selective mRNA translation reprogramming. Genes Dev. 31:2235–2249. 2017. View Article : Google Scholar : PubMed/NCBI

121 

Othman A, Winogradzki M, Patel S, Holmes W, Blank A and Pratap J: The role of Runx2 in microtubule acetylation in bone metastatic breast cancer cells. Cancers (Basel). 14:34362022. View Article : Google Scholar : PubMed/NCBI

122 

Wang XX, Ye FG, Zhang J, Li JJ, Chen QX, Lin PY and Song CG: Serum miR-4530 sensitizes breast cancer to neoadjuvant chemotherapy by suppressing RUNX2. Cancer Manag Res. 10:4393–4400. 2018. View Article : Google Scholar : PubMed/NCBI

123 

Huang L, Zhang L and Chen X: Updated review of advances in microRNAs and complex diseases: Taxonomy, trends and challenges of computational models. Brief Bioinform. 23:bbac3582022. View Article : Google Scholar : PubMed/NCBI

124 

Chen X, Xie D, Zhao Q and You ZH: MicroRNAs and complex diseases: From experimental results to computational models. Brief Bioinform. 20:515–539. 2019. View Article : Google Scholar : PubMed/NCBI

125 

Chen X, Yan CC, Zhang X and You ZH: Long non-coding RNAs and complex diseases: From experimental results to computational models. Brief Bioinform. 18:558–576. 2017.PubMed/NCBI

126 

Wang CC, Han CD, Zhao Q and Chen X: Circular RNAs and complex diseases: From experimental results to computational models. Brief Bioinform. 22:bbab2862021. View Article : Google Scholar : PubMed/NCBI

127 

Zhao W, Geng D, Li S, Chen Z and Sun M: LncRNA HOTAIR influences cell growth, migration, invasion, and apoptosis via the miR-20a-5p/HMGA2 axis in breast cancer. Cancer Med. 7:842–855. 2018. View Article : Google Scholar : PubMed/NCBI

128 

Xu J, Wu KJ, Jia QJ and Ding XF: Roles of miRNA and lncRNA in triple-negative breast cancer. J Zhejiang Univ Sci B. 21:673–689. 2020. View Article : Google Scholar : PubMed/NCBI

129 

Kong X, Duan Y, Sang Y, Li Y, Zhang H, Liang Y, Liu Y, Zhang N and Yang Q: LncRNA-CDC6 promotes breast cancer progression and function as ceRNA to target CDC6 by sponging microRNA-215. J Cell Physiol. 234:9105–9117. 2019. View Article : Google Scholar : PubMed/NCBI

130 

Sang Y, Chen B, Song X, Li Y, Liang Y, Han D, Zhang N, Zhang H, Liu Y, Chen T, et al: circRNA_0025202 regulates tamoxifen sensitivity and tumor progression via regulating the miR-182-5p/FOXO3a axis in breast cancer. Mol Ther. 27:1638–1652. 2019. View Article : Google Scholar : PubMed/NCBI

131 

Yin X, Wang P, Yang T, Li G, Teng X, Huang W and Yu H: Identification of key modules and genes associated with breast cancer prognosis using WGCNA and ceRNA network analysis. Aging (Albany NY). 13:2519–2538. 2020. View Article : Google Scholar : PubMed/NCBI

132 

Joshi H and Press MF: Molecular oncology of breast cancer. The Breast. Elsevier; Amsterdam, The Netherlands: pp. pp282–307.e5. 2018, Available online:. https://www.sciencedirect.com/science/article/pii/B978032335955900022230–May. 2021 View Article : Google Scholar

133 

Dzobo K and Sinkala M: Cancer stem cell marker CD44 plays multiple key roles in human cancers: immune suppression/evasion, drug resistance, epithelial-mesenchymal transition, and metastasis. OMICS. 25:313–332. 2021. View Article : Google Scholar : PubMed/NCBI

134 

Fillmore C and Kuperwasser C: Human breast cancer stem cell markers CD44 and CD24: Enriching for cells with functional properties in mice or in man? Breast Cancer Res. 9:3032007. View Article : Google Scholar : PubMed/NCBI

135 

Tomita H, Tanaka K, Tanaka T and Hara A: Aldehyde dehydrogenase 1A1 in stem cells and cancer. Oncotarget. 7:11018–11032. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2023
Volume 25 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Si W, Kan C, Zhang L and Li F: Role of RUNX2 in breast cancer development and drug resistance (Review). Oncol Lett 25: 176, 2023
APA
Si, W., Kan, C., Zhang, L., & Li, F. (2023). Role of RUNX2 in breast cancer development and drug resistance (Review). Oncology Letters, 25, 176. https://doi.org/10.3892/ol.2023.13762
MLA
Si, W., Kan, C., Zhang, L., Li, F."Role of RUNX2 in breast cancer development and drug resistance (Review)". Oncology Letters 25.5 (2023): 176.
Chicago
Si, W., Kan, C., Zhang, L., Li, F."Role of RUNX2 in breast cancer development and drug resistance (Review)". Oncology Letters 25, no. 5 (2023): 176. https://doi.org/10.3892/ol.2023.13762