Next Article in Journal
Novel Bacillus-Infecting Phage Bquatquinnuvirus eskimopiis (Strains B450T and B450C), Founder of a New Genus, and the Properties of Its Endolysin
Previous Article in Journal
Inflammation as a Prognostic Marker in Cardiovascular Kidney Metabolic Syndrome: A Systematic Review
Previous Article in Special Issue
Fueling the Seed: Growth Factors and Cytokines Driving Cancer Stem Cells in Gynecological Malignancies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Decoding Breast Cancer: Emerging Molecular Biomarkers and Novel Therapeutic Targets for Precision Medicine

by
Dámaris P. Intriago-Baldeón
1,†,
Eduarda Sofía Pérez-Coral
1,†,
Martina Isabella Armas Samaniego
2,
Vanessa I. Romero
2,3,
Juan Carlos Pozo Palacios
4,* and
Gabriele Davide Bigoni-Ordóñez
5,6,*
1
Grupo de Investigación en Biomedicina Experimental y Aplicada, Facultad de Ciencias de la Salud, Universidad Internacional SEK (UISEK), Quito 170120, Ecuador
2
Escuela de Medicina, Colegio de Ciencias de la Salud, Universidad San Francisco de Quito, Quito 170901, Ecuador
3
Instituto de Microbiología, Colegio de Ciencias Biológicas y Ambientales, Universidad San Francisco de Quito, Quito 170901, Ecuador
4
Facultad de Ciencias Médicas Eugenio Espejo, Campus Cuenca, Universidad UTE, Cuenca 010109, Ecuador
5
Carrera de Laboratorio Clínico, Facultad de Ciencias Médicas, Universidad de Cuenca, Cuenca 010107, Ecuador
6
Grupo de Investigación en Salud Sexual y Reproductiva, Facultad de Ciencias Médicas, Universidad de Cuenca, Cuenca 010107, Ecuador
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2026, 27(1), 138; https://doi.org/10.3390/ijms27010138
Submission received: 5 November 2025 / Revised: 13 December 2025 / Accepted: 17 December 2025 / Published: 22 December 2025

Abstract

Breast cancer is the most frequent gynecological malignancy and the main cause of cancer death in the female population worldwide. One of the most significant challenges in its clinical management is the molecular heterogeneity of malignant breast tumors, which is reflected in the current molecular classification of these entities. In each of these tumor molecular subtypes, distinct genetic alterations are involved, and several intracellular signaling pathways contribute to defining their biological identity and clinical response. This literature review summarized the main classic and emerging biomarkers in breast cancer, along with the therapies associated with them. There are several classic biomarkers associated with this disease, such as estrogen and progesterone receptors, the HER2 receptor, and the Ki-67 cell proliferation marker. Given the limitations of these biomarkers, new biomarkers have been identified, including the TP53 tumor suppressor gene, the EGFR, different types of RNAs, plus epigenetic and immunological biomarkers. The integration of classic and emerging biomarkers along with new therapeutic targets in the clinical practice has promoted a thorough understanding of the high molecular complexity of breast cancer and the development of precision medicine strategies which increase the chances of therapeutic success.

1. Introduction

Breast cancer is one of the most frequent and challenging malignant neoplasms worldwide. It is defined as a heterogeneous group of diseases characterized by the uncontrolled growth and division of breast cells with molecular alterations [1]. This excessive cell proliferation results from the acquisition and integration of several hallmarks of cancer, such as self-sufficiency in growth signals, inhibition of apoptosis, sustained angiogenesis, and evasion of the anticancer immune response, which together promote the formation of malignant breast tumors with metastatic potential [2,3]. Despite increasing advances in early detection and personalized treatments, the incidence and mortality of breast cancer continue to rise in the female population at the global scale, due to its high molecular complexity and heterogeneity [4,5]. In this context, the identification of molecular biomarkers is highly relevant, since these molecules allow the characterization of molecular subtypes of breast cancer, contribute to the clinical management of the disease, predict tumor response to medical treatments, and help to establish more accurate prognoses [6,7]. Therefore, the search for new biomarkers has been promoted, which, in conjunction with classic biomarkers, provide detailed information on the biological characteristics of malignant breast tumors and favor the development and administration of personalized therapies that are adjusted to the molecular profiles of each patient’s tumors [8,9]. Given the need to gather updated scientific evidence on biomarkers for this malignant neoplasm, this literature review summarized the main classic and emerging biomarkers in breast cancer, along with the therapies associated with them.

2. Epidemiology and Risk Factors in Breast Cancer

Breast cancer is the most common malignant neoplasm and one of the leading causes of cancer death among women worldwide. According to recent World Health Organization (WHO) estimates, in 2022, more than 2.3 million new cases were diagnosed, and approximately 670,000 deaths from this cause were recorded [10]. The global burden of the disease continues to rise, with an estimated annual increase of 1–5% in incidence in about half of the countries evaluated [11]. Although mortality rates have declined steadily in countries with a very high Human Development Index (HDI), attributed to improved screening and therapies, critical gaps persist in regions with a low HDI, contributing to higher mortality rates [10,11,12,13]. Moreover, there is a high incidence of breast cancer in Australia and New Zealand, North America, and Northern Europe, while the highest mortality rates are found in Melanesia, Western Africa, and Micronesia/Polynesia [12,13]. Furthermore, it has been projected that, by 2050, there will be more than 3 million new breast cancer cases each year, along with a 68% increase in mortality, if global prevention strategies are not strengthened [11].
There are multiple risk factors associated with the development of breast cancer, which can be classified as modifiable and non-modifiable [14]. Among the non-modifiable risk factors that predispose individuals to developing this disease are being female, which is associated with greater stimulation by hormones such as estrogen and androgens [15], being over the age of 50, as this is related to the accumulation of cellular alterations that may increase the likelihood of carcinogenesis [16,17], having a family history of breast cancer in first-degree relatives [18,19], the presence of mutations in genes such as BRCA1 and BRCA2 [20,21], belonging to specific ethnic groups such as being a non-Hispanic Caucasian or African American woman [22,23,24], having an early onset of menstruation and a late onset of menopause [25], having high breast tissue density [26,27], having a personal history of breast cancer [28], having a previous diagnosis of other benign conditions affecting the mammary glands [29], and having previously undergone radiation therapy [30]. In this context, it is worth mentioning that early pregnancy and breastfeeding are considered protective factors against breast cancer; however, pregnancy at age 35 is associated with an increased risk of developing this disease [31,32,33]. On the other hand, the modifiable risk factors that increase the probability of developing breast cancer are the use of hormone replacement therapies [34,35], the administration of drugs such as diethylstilbestrol during pregnancy [36,37], lack of physical activity, as exercise reduces the risk of developing this disease [38,39,40], overweight and obesity in postmenopausal women [41], high consumption of alcoholic beverages and tobacco [42,43], vitamin deficiencies such as vitamin D [44], exposure to artificial light at night [45,46], high consumption of processed foods in the daily diet [47], and exposure to chemicals that could have carcinogenic effects [48,49].

3. Genetic Basis for Breast Cancer

Multifactorial diseases like breast cancer result from the interaction of genetic and non-genetic factors. Identifying these components allows for a better understanding of malignant breast tumor biology and clinical heterogeneity. The most relevant factors are described below:

3.1. Non-Genetic Factors

Recently documented cases in multiple patient cohorts reveal an increased risk of developing breast cancer due to diverse non-genetic factors. These factors include environmental factors such as alcohol consumption [50] and hormone replacement therapy [51], hormonal factors such as prolonged estrogen exposure in cases of early menarche or late menopause [52], reproductive factors such as number of children and breastfeeding [53], and metabolic factors, such as sedentary lifestyle and postmenopausal obesity [54]. It has recently been shown that these hormonal, reproductive, metabolic, and environmental factors can induce epigenetic changes [55], such as hypermethylation of tumor suppressor gene promoters such as BRCA1 [56], CDH1, RASSF1A, and PTEN, or histone modifications (EZH2, HDAC1), which alter gene expression and modulate the effect of genetic variants related to breast cancer risk [57,58]. This interaction among environment, epigenome, and genotype could explain the differences in breast cancer incidence and clinical presentation observed across populations [59].

3.2. Genetic Factors

The development of next-generation sequencing (NGS) has promoted the identification of multiple genes related to breast cancer predisposition. The genetic factors can be classified into germline and somatic variants [60]. Germline variants are inherited and are associated with a medium to a high risk of cancer; it is estimated that 5 to 10% of all breast cancers are caused by germline variants. Updated clinical guidelines recommend, at a minimum, evaluating multigene panels that contain BRCA1, BRCA2, PALB2, TP53, PTEN, CDH1, STK11, ATM, and CHEK2 to assess the risk of germline variants [61]. Somatic variants are not inherited but significantly influence prognosis and treatment response, depending on the molecular subtype, and are responsible for the majority of cancers [62]. In general, these genetic factors can be classified into high-penetrance variants, intermediate-penetrance variants, and low-penetrance polymorphisms:
  • High-Penetrance Variants: These genetic variants can occur in the BRCA1 and BRCA2 genes, with cumulative lifetime risks of 55–70% and 45–69%, respectively [63]. However, the risk varies depending on the variant type and the population studied. Other high-penetrance genes are TP53 (Li-Fraumeni syndrome) [64], PTEN (Cowden syndrome), and CDH1 (hereditary lobular carcinoma). These genes have been recognized as responsible for hereditary cancer predisposition syndromes, in addition to their association with breast cancer [65].
  • Intermediate Penetrance Variants: These genetic variants have been identified in genes like PALB2, CHEK2, and ATM, and are known to confer a moderate to high risk of developing breast cancer, although a lower risk than that associated with BRCA1 and BRCA2 [66]. The estimated cumulative risk, situated between 20 and 50% for PALB2 [67] and 20–40% for CHEK2 [68], relies on factors like family history and demographic group. Regarding ATM, certain variations have also been linked to a moderate risk [69].
  • Low-Penetrance Polymorphisms: These genetic variants can be analyzed using polygenic risk scores (PRS), which quantify cumulative risk based on hundreds of single nucleotide polymorphisms (SNPs) and adjust the individual risk of breast cancer [70,71].

3.3. Alterations in Intracellular Signaling Pathways Linked to Breast Cancer

Germline and somatic mutations, along with epigenetic alterations, can disrupt intracellular signaling pathways that control and regulate cell proliferation and differentiation, ultimately shaping the tumor phenotype [72]. These pathways include:
  • PI3K/AKT/mTOR signaling pathway: Mutations in the PIK3CA gene described in hormone receptor-positive (HR+) luminal breast carcinomas plus loss of PTEN or hypermethylation of its promoter induce AKT phosphorylation and mTORC1/2 activation [73]. This signaling pathway stimulates cell proliferation and resistance to genotoxic stress. For this reason, this pathway is used as a therapeutic target for PI3K and mTOR inhibitors [74].
  • MAPK signaling pathway (RAS/RAF/MEK/ERK): Amplification of receptor tyrosine kinases (RTKs), such as epidermal growth factor receptor-2 (HER2/ERBB2) and fibroblast growth factor receptor-1 (FGFR1), mutations in RAS/BRAF, or cross-activation with the PI3K/AKT signaling pathway induces cell proliferation, improved cell migration and suppression of apoptosis (associated with aggressive phenotypes of breast cancer such as triple-negative (TNBC) and HER2-positive) [75]. Simultaneous activation of the MAPK and PI3K/AKT signaling pathways induces adaptive therapeutic resistance [76].
  • Wnt/β-catenin signaling pathway: Wnt overactivation due to SFRP1 hypermethylation or CTNNB1 mutations leads to nuclear accumulation of β-catenin, which in turn regulates the expression of genes associated with cell plasticity and metastasis. This molecular mechanism has been identified in basal and metaplastic tumors [77]. The interaction of this pathway with the PI3K/AKT and MAPK signaling pathways contributes to tumor heterogeneity and treatment resistance [78].

4. Molecular Basis for Breast Cancer

4.1. Establishment of Molecular Subtypes of Breast Cancer

From a biological point of view, malignant breast tumors are clinically heterogeneous entities, and their molecular classification has enabled the identification of subtypes with distinct prognostic and therapeutic profiles [79]. Malignant breast tumors have been classified into several intrinsic molecular subtypes based on the analysis of their mRNA expression profiles. In 2000, the study conducted by Perou et al. established four intrinsic molecular subtypes of breast cancer based on gene expression data obtained from complementary DNA (cDNA) microarrays that analyzed 8102 genes from 65 human breast cancer specimens derived from 42 different patients: Luminal, HER2-enriched, Basal-like, and Normal Breast-like [80]. Subsequently, the Luminal subtype was divided into two categories: Luminal A and Luminal B subtypes [81,82]. In addition, it has been established that the Normal Breast-like subtype represents biological samples that were contaminated with healthy breast tissue or healthy germline DNA; therefore, this subtype was excluded from the current molecular classification for the disease [2,14,83].
Moreover, the results of the Cancer Genome Atlas (TCGA) project, which analyzed tumor and germline DNA samples from 825 patients at the genomic, transcriptomic, and proteomic levels to establish clusters of tumors with molecular similarities, confirmed the existence of four intrinsic subtypes of breast cancer based solely on mRNA expression profiles: Luminal A, Luminal B, HER2-enriched, and Basal-like [84]. Furthermore, a fifth intrinsic molecular subtype called Claudin-low was added to this classification, which was first described in 2007 in a study by Herschkowitz et al. that analyzed the gene expression profiles of 13 breast tumors obtained from murine models of the disease, using cDNA microarrays, and then compared these data with those derived from malignant human breast tumors [85]. Each of these five intrinsic molecular subtypes of breast cancer exhibit unique biological characteristics that directly influence its clinical behavior and prognosis, which are described below (Figure 1).

4.2. Characteristics of Breast Cancer Molecular Subtypes

4.2.1. Luminal A

The histological grade of malignant breast tumors belonging to the luminal A subtype is low. These tumors are characterized by the expression of estrogen receptors (ER) and/or progesterone receptors (PR), as well as low-molecular-weight cytokeratins. In addition, these tumors do not express HER2/ERBB2, exhibit low Ki-67 expression, and have a low risk of recurrence. Luminal A malignant breast tumors are the most prevalent, accounting for approximately 50% of all diagnosed breast cancer cases. These tumors are associated with slow clinical progression, a good prognosis, and an excellent response to hormone therapies [86,87,88]. These tumors express a robust luminal genetic signature, including ESR1, GATA3, XBP1, and FOXA1 genes [84], and show low expression of genes related to cell proliferation [89].

4.2.2. Luminal B

Malignant breast tumors of the luminal B subtype are histologically high grade and tend to have a worse prognosis than luminal A tumors; approximately 20% of all diagnosed cases of breast cancer correspond to this molecular subtype [88,90]. These tumors can be classified into two subgroups: HER2-positive luminal B and HER2-negative luminal B. HER2-positive luminal B tumors express ERs, often overexpress HER2, and may exhibit varying Ki-67 and PR receptor expression. On the other hand, HER2-negative luminal B tumors express ERs and do not express HER2 receptors; in addition, these tumors have at least one of the following characteristics: high expression of the Ki-67 cell proliferation marker, low or no expression of the PR receptor, and a high risk of recurrence [86,87]. Luminal B malignant breast tumors show high expression of genes related to cell proliferation, as well as low-molecular-weight cytokeratins [84,88]. The response of these tumors to hormone therapies and chemotherapy is variable; when these tumors are more biologically aggressive and less sensitive to hormone therapies, combined systemic treatment is usually administered [87,88].

4.2.3. HER2-Enriched

HER2-enriched malignant breast tumors have a high histological grade, are associated with a high proliferative index and an increased risk of metastasis, and have a worse prognosis compared to luminal A and luminal B malignant breast tumors. These tumors show intermediate expression of the luminal genetic signature and do not express the ER and PR hormone receptors. Instead, they are characterized by amplification of the HER2/ERBB2 gene, located on chromosome 17q12, which leads to overexpression of the HER2 receptor [84,86,88,91]. Approximately, 15% of all diagnosed breast cancer cases correspond to this molecular subtype; these tumors have a high expression of the Ki-67 cell proliferation marker and mutations in the TP53 gene [88]. It is worth mentioning that therapies targeting the HER2 receptor, such as Trastuzumab and Pertuzumab, which can be administered along with other cancer treatments, such as chemotherapy, have significantly improved disease-free survival (DFS) in patients with this molecular subtype of the disease [92].

4.2.4. Basal-like/Triple-Negative (TNBC)

Malignant basal-like breast tumors represent approximately 15% of all diagnosed cases of breast cancer, have high proliferation rates, and exhibit high expression of basal cytokeratins and epidermal growth factor receptor (EGFR), along with low expression of the luminal A genetic signature. These tumors are characterized by high chromosomal instability and germline mutations in the BRCA1 gene [84]. Approximately, 80% of these malignant tumors correspond to the TNBC subtype [2]. TNBC is a group of heterogeneous malignant breast tumors that do not express ER and PR hormone receptors and the HER2 receptor. In addition, these malignant tumors exhibit high Ki-67 expression and TP53 mutations [6,86,88,93]. TNBC tumors are more common in young women, especially the ones of African descent and/or with BRCA1 gene mutations [6,94,95]. This molecular subtype of breast cancer is associated with an aggressive clinical progression, limited specific treatment options, and a poorer overall prognosis [96].

4.2.5. Claudin Low

Malignant breast tumors that belong to the Claudin-low subtype are generally triple-negative, have a poor prognosis, exhibit low or no expression of luminal differentiation markers, and have a high expression of markers related to the epithelial–mesenchymal transition (EMT), genes associated with the immune response, and cancer stem cell-like features. This molecular subtype of breast cancer has a response to standard preoperative chemotherapy that is intermediate between the one seen in luminal tumors and the one observed in basal-like tumors [97].

5. Classic Biomarkers for Breast Cancer

Molecular biomarkers complement traditional clinicopathological characteristics (such as tumor size, histological grade, and lymph node involvement) in guiding personalized therapeutic strategies [98]. They allow the disease to be detected and categorized at early stages, predict therapeutic response, estimate prognosis, and assess the risk of recurrence [99,100]. The so-called classic biomarkers of breast cancer (ER, PR and androgen (AR) hormone receptors, the HER2 receptor, and the Ki-67 cell proliferation marker) were the first ones to be studied and standardized and remain pillars of the clinical management of this disease, despite the emergence of new molecular candidates.

5.1. Hormone Receptors

Breast tissue development and differentiation are primarily regulated by estrogen and progesterone, which are hormones that act through the nuclear receptors ER and PR [101]. Both ER and PR are predominantly expressed in luminal epithelial cells, and their immunohistochemical identification is essential for diagnosis, prognosis prediction, and therapeutic selection in breast cancer.

5.1.1. ER

The ER comprises two subtypes: ERα encoded by the ESR1 gene, located at chromosome 6q25.1-q25.2, and ERβ encoded by the ESR2 gene, located at chromosome 14q23.2-q23.3. ERα is expressed in 70–75% of malignant luminal breast tumors [101,102,103]. ERβ modulates and counteracts ERα-mediated cell hyperproliferation [6,104]. ER has six functional domains: the N-terminal AF-1-independent A/B domain, the DNA-binding domain (DBD), the hinge region (D), the ligand-binding domain (LBD), the C-terminal domain, and the AF-2 transcriptional activation domain [101]. These domains regulate the transcription of genes, such as CCND1, FOXM1, IGF-1, MYC, BCL2, and PGR, which are involved in cell proliferation and survival [104]. In patients treated with antiestrogens, mutations in the LBD of ESR1 are associated with therapeutic resistance [105].

5.1.2. PR

The PR exists in two main isoforms, PR-A (94 kDa) and PR-B (114 kDa), both encoded by the PGR gene located at chromosome 11q22.1 but transcribed by alternative promoters [106]. PR-A, which is predominantly nuclear, can inhibit the transcriptional activity of PR-B, which is the primary mediator of progesterone-induced signaling [107]. The alteration in the PR-A: PR-B ratio occurs at the early stages of mammary tumorigenesis, in both tumor and adjacent cells. PR consists of the DBD, the LDB, the amino-terminal and the transcriptional activation domains (AF-1, AF-2, and additional AF-3). PR modulates ERα-mediated estrogen signaling by regulating the expression of genes, such as RANKL, WNT4, and CCND1, which activate signaling pathways, such as PI3K/AKT and MAPK. PR loss is linked to a higher probability of recurrence and development of endocrine resistance, mainly to tamoxifen and exemestane [108].

5.1.3. AR

The AR, encoded on chromosome Xq12, is a ligand-dependent transcription factor that regulates genes involved in proliferation, differentiation, and metabolic balance [109]. Recent evidence has strengthened its role as an emerging biomarker in breast cancer, particularly in TNBC, where AR expression contributes to a distinct molecular phenotype known as the luminal androgen receptor (LAR) subtype [110]. This subgroup shows AR-driven transcriptional activity and unique therapeutic vulnerabilities. Contemporary reviews and translational studies highlight that AR-positive TNBC may exhibit differential prognosis and can respond to androgen-signaling inhibition, supporting ongoing interest in AR-targeted approaches [111]. Together, these findings position AR as an increasingly relevant biomarker with growing clinical and molecular implications in TNBC.

5.2. HER2/ERBB2

The HER2/ERBB2 is a tyrosine kinase receptor encoded by the ERBB2 gene, located at chromosome 17q12, which is composed of three domains: the extracellular domain (ECD) with four subdomains (I–IV), the transmembrane domain (TMD), and the intracellular region [112]. Receptor activation through homo- or heterodimerization induces autophosphorylation of tyrosine residues, which triggers signaling pathways, such as MAPK and PI3K/AKT/mTOR [112,113]. The HER2 receptor is amplified or overexpressed in 15–30% breast carcinomas, especially in those that are more aggressive and have a poor prognosis [113]. Intratumoral heterogeneity in HER2 receptor expression may reduce the efficacy of targeted therapies [114]. Point mutations that affect the structure of the ECD or the intracellular tyrosine kinase region, independent of amplification or overexpression, are associated with therapeutic resistance [115,116,117,118].

5.3. Ki-67 Cell Proliferation Marker

Ki-67 is a nuclear antigen of approximately 359 kDa encoded by the MKI67 gene, which is actively expressed during the G1, S, G2, and M phases of the cell cycle, but is absent in quiescent cells (G0) [7,119]. It is overexpressed in approximately 16.67% of breast carcinomas, particularly in enlarged hyperplastic lobular units [120]. In oncology, it is used as an index of cell proliferation, and it is associated with a poor prognosis; expression above 60% is associated with a higher risk of recurrence and lower survival rates [121]. It is also used as a predictive biomarker of response to endocrine therapies. For example, an increase in its expression after treatment is associated with a poorer response and lower recurrence-free survival, while high baseline expression does not necessarily predict poor response [121]. Despite its clinical utility, there are limitations in the consensus on the cutoff point to distinguish between low- and high-risk tumors, in interobserver variability, and in the standardization of quantification methods, traditionally performed by microscopy [122,123,124].

6. Emerging Molecular Biomarkers for Breast Cancer

A wide range of molecular alterations have been identified as potential biomarkers for breast cancer, including genetic mutations, aberrant receptor signaling, dysregulated expression of non-coding RNAs, epigenetic modifications, and immune-related markers. These biomarkers reflect tumor heterogeneity and have important implications for diagnosis, prognosis, and therapeutic decision-making (Figure 2).

6.1. TP53 Tumor Suppressor Gene

TP53 is a tumor suppressor gene located on chromosome 17p13.1 that encodes the p53 transcription factor, which is composed of a transactivation domain (TAD), a DBD, a tetramerization domain, a proline-rich domain, and a regulatory domain [125]. TP53 is the most frequently mutated gene in human cancer, with both loss-of-function and gain-of-function mutations that can alter cell cycle regulation, apoptosis, senescence, DNA repair, and accumulate genetic alterations [126,127]. Therefore, malignant tumors harboring TP53 mutations are associated with rapid progression, resistance to conventional therapies, and poor prognosis.
Approximately 85% of women carrying TP53 mutations eventually develop breast cancer, either due to germline (5–8%) or somatic (37%) mutations; these mutations predominate in triple-negative, HER2-enriched, and basal-like breast cancers, and they are usually located in exons 5–8 [126]. In addition, certain TP53 mutations could induce immunogenicity in breast cancer through the regulation of several p53-mediated signaling pathways, which could be associated with a better prognosis in TP53-mutated malignant breast tumors; this result implies that TP53 mutation status could be considered as a potential biomarker to classify patients who might be responsive to immunotherapies [128].

6.2. EGFR/HER1/ERBB1

The EGFR is a tyrosine kinase receptor consisting of an ECD, a TMD, and an intracellular region with tyrosine kinase activity [129]. Its activation occurs after the binding of ligands that induce its dimerization, autophosphorylation, and subsequent activation of the MAPK, PI3K/AKT, and JAK-STAT signaling pathways [76,129]. EGFR signaling can be amplified by the increased expression of ligands, such as TGF-α, Heparin-Binding EGF-Like Growth Factor (HBEGF), and amphiregulin (AREG), which can be produced endogenously in response to lifestyle or environmental factors [130]. It is overexpressed or aberrantly activated in 14–45% of breast carcinomas, especially in TNBC, and it is associated with increased proliferation, resistance to apoptosis, metastasis, EMT, and poor clinical prognosis [131,132,133]. The therapeutic potential of EGFR-targeted monoclonal antibodies and tyrosine kinase inhibitors is limited, with a tendency toward recurrence and the development of intrinsic or acquired resistance [134]. This is mainly attributed to tumor heterogeneity, gain-of-function mutations, chromosomal rearrangements, and aberrant or compensatory activation of signaling pathways [130]. In this context, pharmacological inhibition of EGFR may disrupt its crosstalk with other receptor tyrosine kinases, such as IGF-1R, which promote cell proliferation. Consequently, breast cancer cells may activate compensatory signaling pathways, thereby reducing the efficacy of anti-EGFR therapies [135].

6.3. Different Types of RNAs

Non-coding RNAs add an additional regulatory dimension to breast cancer biology. Through their influence on transcription, RNA processing, and protein interaction networks, these molecules help shape tumor behavior and contribute to clinically relevant phenotypes. Among the most extensively studied groups are microRNAs, long non-coding RNAs, and circular RNAs. Each category participates in distinct molecular mechanisms that affect tumor growth, progression, and therapeutic response.

6.3.1. MicroRNAs

MicroRNAs (miRNAs) are short non-coding RNA molecules—typically 15 to 25 nucleotides long—that fine-tune gene expression by pairing with complementary sequences in target mRNAs, promoting their degradation or blocking translation. Through this post-transcriptional regulation, miRNAs participate in biological processes such as epigenetic remodeling, control of protein turnover, and gene silencing. Disruption of miRNA expression or activity can alter key signaling pathways and contribute to cancer initiation and progression [136,137,138]. Functionally, cancer-related miRNAs fall broadly into two subgroups: oncogenic miRNAs (oncomiRs), which become overexpressed and enhance tumor-promoting traits, and tumor-suppressive miRNAs, which restrain cell growth, modulate immune responses, or promote apoptosis, thereby counteracting malignant transformation [139,140].
Clinically, miRNAs have attracted considerable attention because they can be reliably detected in both tissues and body fluids, offering opportunities for minimally invasive diagnostic assays. Numerous studies have reported that women with breast cancer display distinct miRNA expression patterns in blood and tissue samples, which differ markedly from healthy or pre-malignant states [141,142,143,144,145]. These signatures often correlate with tumor subtype: miRNA expression tends to be higher in hormone-receptor-positive tumors and lower in basal-like cancers. Additionally, certain miRNAs show expression patterns associated with BRCA1, BRCA2, and TP53, both before and after chemotherapy [146].
Several individual miRNAs illustrate these roles; for example, miR-155, whose expression is normally restrained by BRCA1/2, promotes proliferation and migration by suppressing SOCS1 and enhancing MMP-16 levels [146,147]. miR-21 is frequently elevated across breast cancer cohorts, particularly in triple-negative cases, where it contributes to tumor aggressiveness in part by downregulating PTEN [146,148]. The miR-200 family (miR-200a/b/c, miR-141, miR-429) regulates epithelial–mesenchymal transitions by inhibiting transcriptional repressors, such as ZEB1/2, although under certain conditions these same molecules can facilitate metastatic spread [146,149,150,151,152,153,154,155].
Furthermore, studies have been conducted to advance miRNA-based therapeutics into the clinical setting, by identifying strategies that could enhance the pharmacological effects of these molecules; for example, a study led by Abdelaal et al. (2023) [156] synthesized and tested a fully modified version of miR-34a (FM-miR-34a) which was conjugated to a synthetically simplistic ligand. This molecule is more stable than its partially modified version, and it exhibited strong silencing of its gene targets; moreover, it inhibited proliferation, migration and invasion of the MB-231 breast cancer human cell line in vitro. In addition, it induced a substantial reduction in tumor growth in mice because of its conjugation to folate (FM-FolamiR-34a); these results suggest that miR-34a could become an anticancer agent with clinical potential.

6.3.2. lncRNAs

Long non-coding RNAs (lncRNAs) are transcripts longer than 200 nucleotides that, despite lacking protein-coding potential, exert regulatory effects at multiple cellular levels [157]. Found in both the nucleus and cytoplasm, lncRNAs modulate gene expression by interacting with DNA, transcription factors, mRNAs, miRNAs, or protein complexes, influencing processes such as chromatin organization, transcriptional activation or repression, and RNA stability [158,159].
Because many lncRNAs exhibit tissue-specific expression patterns and participate in epigenetic regulation of oncogenes and tumor-suppressor genes, they are being explored as candidates for diagnostic and prognostic use. In breast cancer, numerous lncRNAs show characteristic dysregulation. HOX transcript antisense intergenic RNA (HOTAIR) is a well-known example: its overexpression enhances proliferation, promotes EMT, and facilitates metastasis [160,161]. Elevated HOTAIR levels correlate with lymph-node involvement and poorer overall survival (OS), especially in TNBC [162,163,164]. H19, one of the earliest lncRNAs linked to breast cancer, regulates the H19/IGF2 imprinting axis and is frequently upregulated across multiple tumor types [165]. NEAT1 contributes to chemoresistance by sequestering pro-apoptotic miRNAs in TNBC [166]. Likewise, CUPID1 and CUPID2 influence stress-response pathways and hormone-receptor signaling, and their dysregulation has been associated with susceptibility to ER- and PR-positive breast cancers [167].

6.3.3. circRNAs

Circular RNAs (circRNAs) represent a specialized subgroup of non-coding RNAs characterized by a covalently closed loop generated through a back-splicing mechanism that links the 3′ and 5′ ends of exons and/or introns. This circular architecture provides strong resistance to exonuclease degradation, contributing to their stability and making them attractive candidates as biomarkers [168]. Their presence in biological fluids—including plasma and saliva—also supports the development of non-invasive diagnostic approaches [169]. The best-described mechanism of circRNA function is their ability to act as miRNA sponges, sequestering specific miRNAs and thereby modulating entire regulatory networks involved in cancer pathogenesis [170]. However, circRNAs may also interact with RNA-binding proteins, participate in transcriptional regulation, and, in some cases, encode small peptides with biological activity.
A variety of circRNAs have been implicated in breast cancer progression. circHMCU enhances proliferative and metastatic behavior by binding to members of the let-7 miRNA family, which leads to increased expression of MYC, HMGA2, and CCND1 [171]. circ-Dnmt1, frequently upregulated in malignant breast cells, promotes autophagy and supports tumor survival; its inhibition reduces cell proliferation and tumor growth in vivo [172]. Conversely, circRNAs, such as circCCDC85A, hsa_circ_0072309, and circVRK1, tend to be downregulated in breast cancer, and their re-expression suppresses proliferation, invasion, and migration [173,174,175]. Additional molecules, including hsa_circ_0006743 and hsa_circ_0002496, are enriched in early-stage breast cancers [176]. Some circRNAs also influence therapeutic outcomes: circRNA_0025202 enhances tamoxifen sensitivity in ER-positive malignant breast tumors [177], while hsa_circ_0000199, which is elevated in TNBC, modulates response to chemotherapy and improves treatment efficacy when suppressed [178].

6.4. Epigenetic Biomarkers

In cancer, epigenetic mechanisms such as DNA methylation and histone post-translational modifications are abnormally dysregulated, leading to changes in gene expression without altering the DNA sequence [179,180]. These mechanisms regulate the activation or silencing of oncogenes and tumor suppressor genes and may be involved in the processes of cell proliferation, apoptosis, and differentiation [181]. Recently, there has been an increase in reports of recurrent mutations in genes encoding epigenetic modulators associated with EMT, pluripotency, and drug resistance [182,183]. Therefore, it has been proposed that epigenetic patterns could serve as biomarkers for diagnosis and prediction of prognosis and therapeutic response, as well as potential targets for the development of new pharmacological strategies.

6.4.1. DNA Methylation in Promoter Regions

DNA methylation is a heritable and reversible epigenetic process mediated by DNA methyltransferases, encoded by the DNMT1, DNMT3A, and DNMT3B genes, which catalyze the addition of a methyl group to the carbon-5 of cytosines [184,185]. This event occurs mainly in regions rich in dinucleotides (CpG), such as the promoters and regulators of oncogenes and tumor suppressor genes. On the other hand, demethylation occurs via Ten-Eleven Translocation (TET) dioxygenases, which oxidize 5-methylcytosine (5mC) to generate 5-hydroxymethylcytosine (5hmC) [186,187,188]. In general, cancer cells exhibit global DNA hypomethylation and promoter hypermethylation. Hypomethylation of promoters of oncogenes, such as CRY2 and KPNA2, and drug resistance-associated genes, such as MDR1, favors their activation and increases genomic instability, contributing to uncontrolled cell proliferation and metastasis [183]. Hypermethylation in promoters of tumor suppressor genes, such as BRCA1, APC, CDH1, CCND2, CTNNB1, FOXA1, SOX10, p16, and RASSF1A, causes their transcriptional silencing [183,189,190,191]. In breast cancer patients, hypo- and hypermethylation events have been observed during the early stages of the disease. At the same time, changes in methylation patterns have been reported during the transition from healthy mammary tissue to ductal carcinoma in situ (DCIS), with minimal epigenetic modifications between DCIS and the invasive form [179,189].

6.4.2. Histone Modifications

Histone modifications consist of post-translational alterations of the N-terminal tails of histone proteins, including methylation, acetylation, phosphorylation, ubiquitination, and sumoylation. These processes catalyzed by enzymes, such as histone acetyltransferases (HATs), histone deacetylases (HDACs), and histone methyltransferases (HMTs), which modify the structure of chromatin and, therefore, the accessibility of DNA to the transcriptional machinery [192]. In breast cancer, alterations in histone acetylation and methylation contribute to the dysregulation of gene expression and the development of aggressive malignant tumor phenotypes.
It has been reported that in the more aggressive subtypes of breast cancer, such as basal-like and TNBC, global levels of H3 lysine 27 trimethylation (H3k27me) are reduced compared to less aggressive subtypes, like luminal A and HER2-enriched, suggesting that higher H3K27me levels could be linked to a better prognosis [191,193,194]. A similar trend has been observed for H4R3me2, which is found at moderate to low levels in poor-prognosis subtypes [195]. Likewise, an increase in histone acetylation has been observed at lysine residues, such as H3K9ac (associated with HER2-enriched tumors, poor prognosis, and reduced OV), H3K18ac (associated with hormone receptor-positive malignant breast tumors), and H4K12ac (found in adjacent normal breast tissues to luminal and triple-negative malignant breast tumors) [196,197,198].

6.5. Immunological Biomarkers

In recent years, the emergence of new cancer treatments, such as immunotherapies that stimulate the antitumor immune response, has promoted the generation of scientific literature that analyzes the immunogenicity of breast cancer and the execution of clinical studies to identify new biomarkers that could reflect this immunogenicity and predict the response of malignant breast tumors to immunotherapy [199,200,201,202,203]. Malignant breast tumors that belong to the HER2-enriched and TNBC molecular subtypes are often highly immunogenic, while malignant breast tumors that express ER and PR hormone receptors tend to have a medium to low level of immunogenicity. Highly immunogenic malignant breast tumors that are sensitive to immunotherapy are characterized by a high expression of tumor-infiltrating lymphocytes (TILs) and programmed death-ligand 1 (PD-L1) [204,205]. In this regard, TILs and PD-L1 have emerged as potential biomarkers of immunotherapy response in breast cancer, as described below.

6.5.1. PD-L1

PD-L1 is a 33 kDa type 1 transmembrane protein ligand encoded by the PD-L1 gene (also known as CD274 and B7-H1) and expressed in different types of activated immune cells, such as T lymphocytes, B lymphocytes, macrophages, and dendritic cells. PD-L1 can also be expressed on the surface of malignant tumor cells [202]. The PD-L1 ligand binds to the PD-1 receptor, encoded by the PDCD1 gene (also known as CD279), which is present on T lymphocytes, B lymphocytes, natural killer (NK) cells, macrophages, and some subtypes of activated dendritic cells; as a result, PD-L1 participates in the PD-1/PD-L1 signaling pathway, which plays an important immunoregulatory role by suppressing the activation of immune cells in normal physiological contexts and in diseases, such as cancer [201,202,203,204,205]. In cancer, the interaction between the PD-L1 ligand expressed on malignant tumor cells and the PD-1 receptor located on cytotoxic T lymphocytes (CD8+) suppresses the antitumor activity of these immune cells and promotes cancer immune evasion. Therefore, PD-L1 expression contributes to tumor growth and progression primarily by inhibiting antitumor immune responses and promoting an immunosuppressive tumor microenvironment [201,202,203,204,205].
Based on the knowledge of the biological effects induced by the PD-1/PD-L1 interaction, immunotherapy drugs have been designed to block ligand–receptor binding and improve the ability of the CD8+ T lymphocytes to identify and eliminate malignant tumor cells. In this context, examples of immune checkpoint inhibitors include the anti-PD-1 monoclonal antibodies, such as Pembrolizumab and Nivolumab, and the anti-PD-L1 monoclonal antibodies, such as Atezolizumab and Durvalumab [206]. In clinical practice, the presence or absence of PD-L1 in malignant tumors is assessed by immunohistochemistry (IHC), which is the gold standard for determining which patients are eligible for immunotherapy based on immune checkpoint inhibitors [206]. For example, in TNBC, analysis of PD-L1 expression in immune and malignant breast tumor cells by IHC is a crucial step in identifying patients who may respond favorably to immunotherapy [207].

6.5.2. TILs

TILs are a group of immune cells composed of lymphocytes, plasma cells, monocytes, and NK-T cells that may be scattered throughout the stroma of a malignant tumor (stromal TILs) or present within the malignant tumor (intratumoral TILs); the presence of TILs within a malignant tumor is directly related to a patient’s antitumor immune response. Based on their characteristics, TILs can be classified into cellular subgroups, such as CD8+ T lymphocytes, tissue-resident memory T lymphocytes, CD4+ helper T lymphocytes, CD4+ regulatory T lymphocytes, CD4+ follicular helper T lymphocytes, and tumor-infiltrating B lymphocytes. However, the functions of each TIL subpopulation and their clinical significance remain unknown [208,209,210].
At the pathological level, the evaluation of TILs in malignant tumor tissue should focus only on stromal TILs; this is assessed using hematoxylin and eosin staining and by analyzing the ratio between the intratumoral stromal area infiltrated by lymphocytes and plasma cells and the total intratumoral stromal area. Considering the percentage of stromal TILs present in malignant tumor tissue, three outcomes can be established: low percentage of TILs (when there are 0 to 10% stromal TILs), intermediate percentage of TILs (11 to 40%), and high percentage of TILs (greater than 40%) [210]. In this context, there is growing scientific evidence supporting the predictive and prognostic role of TILs in breast cancer. For example, in TNBC, the most immunogenic molecular subtype of breast cancer, a high percentage of TILs is associated with a better response to chemotherapy, a favorable prognosis, and prolonged survival. Finally, several studies suggest that a high percentage of TILs predicts response to neoadjuvant immunotherapy, either alone or in combination with chemotherapy, in early-stage TNBC, as well as response to immune checkpoint inhibitor-based immunotherapy in advanced or metastatic TNBC [204,208,209,210,211,212,213].

7. Advancements in Liquid Biopsy for the Clinical Management of Breast Cancer

In recent decades, liquid biopsy has emerged as an innovative minimally invasive technique that detects several types of molecular biomarkers, tumor cells, and metabolites in a wide variety of biological samples, such as blood and other body fluids, for the diagnosis, monitoring, and treatment of multiple cancer types, including breast cancer [214]. Conventional diagnostic techniques, such as tissue biopsy, have limited capacity to provide a comprehensive view of the dynamics of malignant breast tumor evolution; therefore, liquid biopsy offers the opportunity to conduct repeated sampling and analysis of biological samples to evaluate biomarker expression in malignant breast tumors at different disease stages, monitor tumor heterogeneity, and understand their complex biological characteristics [215,216,217,218]. Examples of liquid biopsy biomarkers are circulating tumor ctDNA, circulating tumor cells (CTCs), non-coding RNAs (ncRNAs), tumor-educated platelets (TEPs), exosomes, proteins, and metabolites; the analyses of these molecular biomarkers contribute to breast cancer’s early detection and screening, selection of the most suitable treatment modalities for patients, prognosis prediction, and monitoring of residual disease [219].
In the context of liquid biopsy for early diagnosis and clinical monitoring of breast cancer, ctDNA constitutes DNA molecules that are released by malignant breast cancer cells into the circulatory system, because of apoptosis or necrosis. It has been shown that there is a high concentration of ctDNA in plasma samples from breast cancer patients when compared to healthy counterparts; this highlights the importance of analyzing ctDNA levels for the early diagnosis of breast cancer, since its accuracy is the highest among other tumor molecular biomarkers [219,220,221]. Also, tumor-specific mutations can be identified in ctDNA samples; this shows the potential of ctDNA as a biomarker for clinical use, which can provide useful information regarding the genetic landscape of malignant breast tumors. In this case, NGS and droplet digital PCR (ddPCR) are used to evaluate the genetic alterations that drive breast cancer progression and response to treatments [215,222]. On the other hand, CTCs are breast cancer cells that detach from the primary malignant breast tumors because of the induction of the EMT process; this scenario allows them to enter the bloodstream and could reach other tissues in the body and establish metastatic colonies [223,224,225]. Since CTCs play a crucial role in metastatic dissemination, their detection in the clinical setting can provide useful insights regarding tumor progression and response to therapies [225]; clinical studies have shown that the presence of CTCs in early breast cancer patients is associated with worse OS and DFS rates [226,227,228].

8. Emerging Therapeutic Targets and Their Clinical Application in Breast Cancer

8.1. Therapies Targeting Altered Signaling Pathways in Breast Cancer

The PI3K/AKT/mTOR signaling cascade remains one of the most intensively explored therapeutic axes in breast cancer. Several inhibitors directed at this pathway have reached clinical implementation or are undergoing advanced evaluation. Among them, alpelisib is already approved for HR+/HER2- tumors harboring PIK3CA mutations, illustrating the relevance of molecular profiling in treatment selection [229]. Agents that block AKT, such as capivasertib, have also shown meaningful clinical benefit when paired with fulvestrant in Phase III studies [230].
Drugs targeting mTOR—specifically everolimus and temsirolimus—act predominantly on mTORC1, suppressing protein synthesis and slowing tumor cell expansion. Current research is prioritizing multi-drug regimens that incorporate endocrine therapy, chemotherapy, MAPK pathway inhibitors, or immunotherapy in an effort to mitigate resistance and improve therapeutic durability [231,232].
Resistance to pathway inhibition is frequently driven by genetic and metabolic adaptations, including PIK3CA mutations, compensatory shifts between PI3K isoforms, and broader metabolic rewiring involving glycolysis, lipid biosynthesis, or autophagy [233]. To counter these changes, several next-generation strategies are under development. These include dual PI3K/mTOR inhibitors, compounds that modulate autophagy, inhibitors of glutaminase or fatty acid synthase, and targeted degraders generated through PROTAC technology [231,234,235].
In estrogen receptor–positive tumors, Selective Estrogen Receptor Degraders (SERDs) and proteolysis-targeting chimeras (PROTACs) offer new options to disrupt or eliminate proteins that sustain tumor growth [236]. Other experimental agents, such as BIBR1591, which interfere with telomerase activity and triggers apoptosis through transcriptional alterations, exemplify how targeting specific molecular vulnerabilities may broaden therapeutic possibilities [237]. Additionally, acrolein-based delivery systems and Pro-FTY, a selective blocker of sphingosine-1-phosphate (S1P) signaling, have shown activity in multidrug-resistant models while avoiding lymphocytopenia, suggesting potential value in combination regimens [238].
As these therapeutic options evolve, progress in biomarker-guided patient selection, the incorporation of immunomodulatory strategies, and the use of AI-enhanced drug discovery and imaging technologies are expected to speed the development of more personalized oncology tools [239].

8.2. Immunotherapy and Checkpoint Inhibitors

Immunotherapy continues to introduce new avenues to strengthen antitumor responses in breast cancer. Expression of PD-L1 remains a key contributor to immune suppression within the tumor microenvironment, often limiting the efficacy of PD-1/PD-L1 blockade. However, studies combining these inhibitors with localized radiotherapy have reported improved outcomes in TNBC without notable increases in toxicity [232,240]. The identification of additional checkpoint systems, such as BTLA/HVEM, further expands opportunities to enhance immune activation and diversify therapeutic strategies [241].
This growing understanding of immune regulation reinforces the potential of combination of regimens and next-generation immunotherapies to overcome resistance mechanisms and achieve more sustained clinical responses. Parallel advances in targeted therapy complement these findings. For instance, trastuzumab deruxtecan has demonstrated superior activity compared with trastuzumab emtansine in HER2-positive metastatic disease, although careful monitoring for interstitial lung disease continues to be crucial [242]. Additional molecules under investigation include FTY720, which may reduce chemotherapy-related neuropathic symptoms while enhancing anticancer effects, and optimized clofarabine derivatives with improved potency and safety in TNBC models [243,244]. These developments collectively support a shift toward increasingly individualized therapeutic approaches.

8.3. Nucleic Acid–Based Therapies

Advances in RNA and DNA-based technologies have opened new perspectives for breast cancer treatment. miRNAs are of particular interest due to their ability to regulate immune checkpoint expression. Certain miRNAs—such as miR-4477a—have demonstrated both immunomodulatory and antitumor effects in breast cancer cell models [243,245]. circRNAs, exemplified by circ-ARHGER28, are also gaining attention for their diagnostic potential and their influence on signaling pathways such as PI3K/AKT/mTOR [246]. The telomerase inhibitor BIBR1591, previously mentioned, further illustrates how targeting nucleic-acid associated processes can induce apoptotic responses [237]. Moreover, CRISPR-based functional genomic screening is transforming the discovery pipeline by identifying genes involved in tumor progression and therapeutic resistance, offering a foundation for future targeted interventions—even though these tools have not yet been adapted into clinical therapies for breast cancer [247]. Furthermore, gene therapy remains a powerful frontier, aiming to correct defective genes or boost immune responses. Together, these approaches illustrate the expanding landscape of personalized and mechanism-driven therapies in breast cancer [248].

8.4. Nanotechnology Applied to Targeted Drug Delivery

Nanotechnology has opened new possibilities for improving targeted therapy in breast cancer. Among the most promising tools are graphene-based quantum dots, which possess favorable optical characteristics, good biocompatibility, and the capacity to enhance the precision and efficiency of drug delivery [238,249]. Recent nanosystems illustrate this potential: for example, nitrogen-doped graphene quantum dots loaded with organotin (IV) compounds have demonstrated improved solubility and a more selective delivery profile, while also offering theranostic capabilities by supporting both imaging and treatment within the same platform [238,250].
Other nanomaterials have also shown synergistic benefits. Combinations of silver graphene quantum dots with radiotherapy or agents such as 17-AAG have produced stronger anticancer responses than individual treatments, particularly through increased induction of apoptosis [251]. Additional strategies continue to emerge. One example includes hybrid nanocapsules composed of Fe3O4, chitosan, and hyaluronic acid, which have proven effective for guiding drugs or genetic material toward CD44-expressing tumor cells with high specificity [252].
Together, these advances illustrate how nanoscale delivery systems can enhance selectivity, reduce toxicity, and support the development of more precise therapeutic approaches for breast cancer.

8.5. Limitations in Emerging Therapeutics for Breast Cancer

Despite the progress achieved through targeted therapies, immunotherapies, and advanced molecular technologies, several challenges remain inherent to emerging therapeutic strategies. Resistance continues to be one of the most persistent obstacles, driven by the capacity of tumor cells to undergo genetic and metabolic adaptation. Furthermore, the reliance of many treatments on specific biomarkers, receptor profiles, or pathway alterations restricts their applicability to relatively narrow patient subgroups, limiting widespread clinical benefit. The heterogeneity and biological complexity characteristics of breast cancer further complicate efforts to develop interventions capable of producing consistent responses across diverse disease subtypes. Even so, advances in molecular profiling, drug design, and integrative technologies are gradually shaping a path toward more flexible and durable treatment frameworks.
Despite the growing number of innovative therapeutic approaches—ranging from pathway-targeted agents and immunotherapies to nucleic acid–based strategies and nanotechnology—current options for breast cancer still face substantial challenges. Tumor evolution, metabolic adaptability, and both inter- and intratumoral heterogeneity continue to undermine the durability of treatment responses. Moreover, many emerging therapies rely on narrowly defined biomarkers or molecular alterations, limiting their applicability to specific patient subsets and reducing the potential for broader clinical benefit. To contextualize these advances within their real-world constraints, Table 1 provides a consolidated overview of the most relevant investigational therapeutic targets, including trial phase, mechanism of action, reported adverse events, and key limitations identified to date. This synthesis highlights not only the progress made but also the critical gaps that must be addressed as these therapies move from preclinical promises toward clinical translation.

8.6. Clinical Application of Classic and Emerging Biomarkers in Breast Cancer

8.6.1. Utility in Early Diagnosis and Risk Stratification

Across clinical guidelines, there is consensus that ER, PR, and HER2 testing are central for early breast cancer (EBC) risk assessment, with IHC and confirmatory in situ hybridization (ISH) used to classify tumors as luminal, HER2-positive, or TNBC [293,294,295,296,297,298,299]. ASCO (American Society of Clinical Oncology) highlights reporting of low ER positivity (1–10%) mandates HER2 testing and recommends multigene assays, such as Oncotype DX®, in ER+/HER2-, node-negative tumors to guide chemotherapy [294,295,296].
ESMO (European Society for Medical Oncology) also requires ER/PR/HER2 plus Ki-67, emphasizes preoperative endocrine response as an additional stratifier, and advises germline BRCA1/2 testing in candidates for Poly (ADP-ribose) polymerases (PARP) inhibitors [293,298]. The Pan-Asian adapted ESMO guidelines align with these principles, emphasizing the same pathology panel, multigene signatures for HR+/HER2- EBC when chemotherapy benefit is uncertain, and BRCA1/2 testing for both hereditary and therapeutic purposes [299]. The 2024 Spanish consensus (SEOM–SEAP) agrees on ER/PR/HER2/Ki-67 testing but extends scope with standardized HER2 scoring (0 vs. 1+) to enable T-DXd use in HER2-low disease, and formally validates several genomic assays, giving the strongest evidence to MammaPrint® [297].

8.6.2. Role in Therapeutic Selection and Response Monitoring

The consensus across guidelines is that biomarkers guide systemic therapy decisions in both EBC and advanced breast cancer (ABC), while the role of liquid biopsy for monitoring remains investigational (Table 2). ASCO supports testing for PIK3CA to identify candidates for alpelisib, gBRCA1/2 for PARP inhibitors, and PD-L1 in TNBC for immune checkpoint inhibitor therapy; it also considers Microsatellite Instability–High (MSI-H)/Deficient Mismatch Repair (dMMR), Tumor Mutational Burden (TMB)-high, and Neurotrophic Tyrosine Receptor Kinase (NTRK) fusions actionable when matching therapies are available, but does not recommend circulating tumor DNA (ctDNA) or circulating tumor cells (CTCs) for routine monitoring [300]. ESMO advises retesting ER/PR/HER2 at metastatic diagnosis, incorporates BRCA1/2, PIK3CA, and PD-L1 as standard in ABC, and limits broader genomic profiling to situations where results would directly influence management or enable trial access, applying the ESMO Scale for Clinical Actionability of Molecular Targets, which ranks biomarkers from Tier I (standard of care) to Tier X (non-actionable) [293,298].
The Pan-Asian consensus applies these principles to regional practice, recommending ER/PR/HER2 and gBRCA1/2 in EBC, applying multigene assays for HR+/HER2- disease when chemotherapy benefit is uncertain, and clarifying that PD-L1 should not be used in EBC; they also advise against unnecessary imaging or biomarkers unless clinically justified [299]. The Spanish consensus aligns with ASCO and ESMO on PIK3CA, BRCA1/2, and PD-L1, but distinctively includes ESR1 mutations, often detected in ctDNA, as a biomarker to guide endocrine sequencing and the use of elacestrant after Cyclin-Dependent Kinase 4/6 inhibitor (CDK4/6i) in HR+/HER2- ABC, while similarly restricting liquid biopsy to applications in detecting therapeutic resistance rather than surveillance [297].

9. Current Limitations in the Clinical Assessment of Classic and Emerging Biomarkers for Breast Cancer

The clinical assessment of classic and emerging molecular biomarkers is crucial for improving the diagnosis, treatment, and monitoring of breast cancer. Current research regarding biomarkers for this disease is focused on identifying novel molecules which are characterized by their high sensitivity and specificity, plus reproducibility, to be implemented in real clinical settings. In addition, ideal biomarkers should be easily quantifiable, user-friendly, cost-effective, and measurable to ensure clinically interpretable results from readily accessible biological fluids or specimens [301]. Nevertheless, crucial limitations related to their validation, integration, standardization, and implementation in clinical practice persist in both classic biomarkers, such as Ki-67, and in emerging ones, such as PD-L1 [302].
Although Ki-67 was discovered during the 1980s and has been extensively studied since this date, its sole adoption and utility in decision-making for clinical practice remains debatable. Generally, the percentage of Ki-67 is determined by calculating the labeling index (LI) from a tissue hotspot evaluated by immunohistochemistry (IHC); however, the lack of standardization and inconsistent reproducibility related to this classic biomarker have promoted the application of complementary genomics tests, such as Oncotype Dx, MammaPrint, Endo-Predict, and Prosigna [303]. Even though the integration of Ki-67 with other diagnostic tools provides a more comprehensive biological analysis of malignant breast tumors, the implementation of additional tests increases the cost of the diagnostic process, affecting accessibility, particularly in developing countries where economic resources are limited.
Another limitation associated with the classic biomarker Ki-67 is its analytical and methodological validation. Currently, for Ki-67, there are no established criteria for the appropriate collection, fixation, and processing of breast cancer specimens during the preanalytical phase. It is worth mentioning that the clinical assessment of Ki-67 requires stricter control on the preanalytical variables, such as the type of fixative, time to fixation, and duration of fixation, as the prolonged, delayed, or insufficient fixation results in a considerable reduction in the LI [304]. Regarding analytical considerations, the main challenge is the intra- and inter-observer variability between pathologists, which is largely attributed to factors, such as intratumoral heterogeneity, difficulties in identifying a tissue hotspot region, and the lack of a well-defined measurement approach—either a score system or a counting method. Moreover, the absence of universally accepted Ki-67 cut-off values further complicates interpretation, particularly with intermediate LI. According to Mikami et al. (2013) [305], while the visual counting method shows high reproducibility, the selection of the tissue hotspot remains a critical factor, and variability becomes especially problematic in malignant breast tumors with intermediate Ki-67 levels (5–25%).
To address these limitations, the multidisciplinary group International Ki-67 in Breast Cancer Working Group (IKWG) was established in 2011, bringing together experts in pathology, medical oncology, public health, biostatistics, and biomedical research. This group aims to standardize the clinical assessment of Ki-67, through the development of evidence-based guidelines, recommendations, cut-off proposals, and training courses for pathologists. However, there is no universal regulatory framework, so the adoption of the guidelines provided by IKWG depends on each laboratory/hospital/pathologist. In parallel, the need for improved reproducibility has prompted the development of automated and semi-automated approaches for Ki-67 clinical assessment based on IKWG’s guidelines. For example, Fernezlian et al. (2023) proposed the concept of a nuclear gradient (NG) of Ki-67 categorized into NG1, NG2, and NG3/4, and analyzed through a semi-automated microscopic image approach based on staining intensity, nuclear distribution, and cell-cycle–related patterns [306]. Similarly, AI approaches have demonstrated high concordance with manual evaluation, often with improved reproducibility, reduced inter- and intra-observer variability, and greater workflow efficiency [307,308,309,310]. Importantly, the performance of AI models depends strongly on the type of algorithm employed (Convolutional Neural Networks, CNNs, or Scale-Invariant Feature Transform, SIFT), the quality of the tissue biopsy images, and the robustness of the training data [309,311]. However, both semi-automated and AI-driven strategies are still being investigated.
On the other hand, there are several limitations associated with the emerging biomarker PD-L1. For instance, currently, there is an absence of a unique standardized protocol for the analysis of PD-L1 expression in malignant breast tumor tissue [206]. Moreover, there are several molecular assays available for PD-L1 detection that include distinct antibody clones for PD-L1 evaluation; also, there are different clinically validated scoring methods for this biomarker. In this context, these tests are not directly interchangeable [206,312]. In addition, during the clinical assessment of PD-L1, interobserver variability can occur, so it is recommended that pathologists attend training sessions for PD-L1 evaluation to improve interobserver reproducibility [206]. Furthermore, it has been shown that there is a discordance in the PD-L1 expression between primary malignant breast tumors and metastatic breast tumors; this scenario highlights the importance of appropriate tissue sampling from metastatic biopsies and the urgent need for standardization of laboratory protocols for PD-L1 evaluation [313]. Nowadays, these scenarios negatively impact the ability of pathologists to correctly identify breast cancer patients who are best suited for immunotherapy based on anti-PD-L1 therapeutic approaches.

10. Current Challenges in the Implementation of Liquid Biopsy at the Clinical Setting for Breast Cancer Management

Despite the advantages associated with liquid biopsy, several challenges hinder its implementation in clinical scenarios to aid in the diagnosis and treatment of breast cancer patients [219]. One of the challenges is the low concentrations of molecular biomarkers, such as ctDNA, in biological samples, like blood specimens, which negatively affects the sensitivity and specificity of liquid biopsy assays; thus, it is necessary to improve the efficiency and performance of laboratory protocols for biomarker extraction and molecular detection technologies [314,315,316,317,318]. Another challenge is the lack of standardized protocols for liquid biopsy tests, which should include a consensus on aspects related to sample collection, its processing before molecular analyses, and the reporting of results; these guidelines would facilitate the implementation of liquid biopsy in the clinical setting [319,320,321,322,323,324]. In addition, a third challenge is the absence of well-designed long-term clinical studies, which would provide key insights regarding the feasibility of the application of liquid biopsy for the clinical monitoring of breast cancer cases [325].

11. Conclusions and Future Perspectives

Breast cancer remains a global health priority not only because of its high incidence and mortality, but also because it represents a biologically diverse set of diseases driven by layered genetic, epigenetic, and microenvironmental programs. Across this review, the evidence supports a central message: meaningful clinical progress increasingly depends on linking tumor biology to actionable biomarkers, rather than relying on clinicopathological features alone. Classic markers such as ER, PR, HER2, and Ki-67 still anchor clinical routine decision-making, enabling subtype definition, therapeutic selection, and prognostic stratification. However, their limitations—particularly variability in assessment and inconsistent cut-offs—highlight the need for better standardization, harmonized reporting frameworks, and robust quality control to ensure that biomarker information translates reliably into patient benefit.
At the same time, emerging biomarkers are reshaping how breast cancer is understood and managed. Alterations in TP53 and EGFR, non-coding RNA networks (miRNAs, lncRNAs, circRNAs), epigenetic signatures, and immune-contexture indicators, such as PD-L1 and TILs, provide a richer description of tumor behavior, treatment sensitivity, and resistance trajectories. These candidates also reflect key biological vulnerabilities that are being exploited therapeutically through pathway inhibition (e.g., PI3K/AKT/mTOR), antibody–drug conjugates, immune checkpoint blockade, and next-generation strategies, including targeted protein degradation, nucleic-acid–based approaches, and nanotechnology-enabled delivery. Yet, the same biology that creates therapeutic opportunities also fuels failure: inter- and intratumoral heterogeneity, clonal evolution, and metabolic adaptation frequently limit the durability and generalizability of therapy responses, especially when interventions are restricted to narrowly defined molecular subgroups.
On the other hand, liquid biopsy further illustrates both promise and constraint. ctDNA, CTCs, exosomes, and circulating RNA signatures offer an appealing route to capture tumor dynamics in real time, potentially improving early detection, monitoring of minimal residual disease, and identification of therapy resistance mechanisms. Nevertheless, low analyte abundance, preanalytical variability, and the lack of universally accepted analytical and clinical validation pathways remain major barriers to routine implementation. In this setting, well-designed longitudinal studies, standardized workflows from sampling to reporting, and clinically meaningful endpoints will be essential to move liquid biopsy from an investigational tool toward a dependable component of health care.
Looking forward, the most impactful advances are likely to emerge from integrative, biomarker-guided frameworks that combine multi-omics profiling with rigorous clinical validation and equitable implementation strategies. This includes harmonizing biomarker assessment across laboratories, improving reproducibility through digital pathology and AI-assisted scoring where appropriate, and prioritizing biomarkers that are not only biologically informative but also feasible in real-world health systems. Ultimately, closing the gap between discovery and clinical utility will require coordinated efforts that connect mechanistic insight, trial design, and implementation science—so that precision oncology in breast cancer becomes both more effective and more accessible.

Author Contributions

D.P.I.-B., E.S.P.-C., J.C.P.P., M.I.A.S. and V.I.R., wrote the manuscript and participated in scientific discussions. G.D.B.-O. provided the concept design and scientific direction, designed the figures for the review, led scientific discussions, and contributed to the editing and drafting of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This review was funded by Universidad de Cuenca and Universidad UTE.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We thank the Vicerrectorado de Investigación de la Universidad de Cuenca for their support in the publication of this article, and we thank the Universidad UTE for the funds allocated to the payment of the APC for open access publication. All figures were created with Mind the Graph.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ABCAdvanced breast cancer
ADCAntibody–drug conjugate
AEAdverse effects
AR Androgen receptor
AREGAmphiregulin
ASCOAmerican Society of Clinical Oncology
CD8+Cytotoxic T lymphocytes
CDK4/6iCyclin-Dependent Kinase 4/6 inhibitor
cDNAComplementary DNA
circRNAsCircular RNAs
CTCsCirculating tumor cells
ctDNACirculating tumor DNA
DHinge region
DBDDNA-binding domain
DCISDuctal carcinoma in situ
DFSDisease-free survival
ddPCRDroplet digital PCR
dMMRDeficient Mismatch Repair
EBCEarly breast cancer
ECDExtracellular domain
EGFR/HER1/ERBB1Epidermal growth factor receptor
EMTEpithelial–mesenchymal transition
EREstrogen receptors
ESCATESMO Scale for Clinical Actionability of molecular Targets
ESMOEuropean Society for Medical Oncology
ESR1Estrogen Receptor 1
FM-miR-34aFully modified version of miR-34a
gBRCA1/2Germline BRCA1/2
H3k27meH3 lysine 27 trimethylation
HATsHistone acetyltransferases
HBEGFHeparin-Binding EGF-Like Growth Factor
HDACsHistone deacetylases
HDIHuman Development Index
HDRHomologous Recombination Deficiency
HER2Growth factor receptor-2
HER2/ERBB2Human epidermal growth factor receptor-2
HMTsHistone methyltransferases
HOTAIRHOX transcript antisense intergenic RNA
HR+Hormone receptor-positive
IHCImmunohistochemistry
IKWGInternational Ki67 in Breast Cancer Working Group
ISHIn Situ Hybridization
LARLuminal androgen receptor
LDBLigand-binding domain
LILabeling index
lncRNAsLong non-coding RNAs
MBCMetastatic Breast Cancer
miRNAsMicroRNAs
MSI-HMicrosatellite Instability–High
ncRNAsnon-coding RNAs
NGSNext-generation sequencing
NKNatural killer cells
NTRKNeurotrophic Tyrosine Receptor Kinase
oncomiRsOncogenic miRNAs
OSOverall survival
PARPPoly (ADP-ribose) polymerases
PD-L1Programmed death-ligand 1
PIK3CAPhosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Alpha
PRProgesterone receptors
PROTACsProteolysis-targeting chimeras
PRSPolygenic risk score
RTKsReceptor tyrosine kinases
S1PSphingosine-1-phosphate
SERDsSelective Estrogen Receptor Degraders
SNPsSingle nucleotide polymorphisms
TADTransactivation domain
TCGACancer Genome Atlas
T-DXdTrastuzumab deruxtecan
TEPsTumor-educated platelets
TILsTumor-infiltrating lymphocytes
TMBTumor Mutational Burden
TMDTransmembrane domain
TNBCTriple-negative breast cancer
TROP2Trophoblast Cell-Surface Antigen 2
VEGFVascular endothelial growth factor
WHOWorld Health Organization

References

  1. Turashvili, G.; Brogi, E. Tumor Heterogeneity in Breast Cancer. Front. Med. 2017, 4, 227. [Google Scholar] [CrossRef]
  2. Nolan, E.; Lindeman, G.J.; Visvader, J.E. Deciphering Breast Cancer: From Biology to the Clinic. Cell 2023, 186, 1708–1728. [Google Scholar] [CrossRef]
  3. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef]
  4. Siegel, R.L.; Kratzer, T.B.; Giaquinto, A.N.; Sung, H.; Jemal, A. Cancer Statistics, 2025. CA Cancer J. Clin. 2025, 75, 10–45. [Google Scholar] [CrossRef]
  5. Xiong, X.; Zheng, L.W.; Ding, Y.; Chen, Y.F.; Cai, Y.W.; Wang, L.P.; Huang, L.; Liu, C.C.; Shao, Z.M.; Yu, K. Da Breast Cancer: Pathogenesis and Treatments. Signal Transduct. Target. Ther. 2025, 10, 49. [Google Scholar] [CrossRef]
  6. Lopez-Gonzalez, L.; Sanchez Cendra, A.; Sanchez Cendra, C.; Roberts Cervantes, E.D.; Espinosa, J.C.; Pekarek, T.; Fraile-Martinez, O.; García-Montero, C.; Rodriguez-Slocker, A.M.; Jiménez-Álvarez, L.; et al. Exploring Biomarkers in Breast Cancer: Hallmarks of Diagnosis, Treatment, and Follow-Up in Clinical Practice. Medicina 2024, 60, 168. [Google Scholar] [CrossRef] [PubMed]
  7. Beňačka, R.; Szabóová, D.; Guľašová, Z.; Hertelyová, Z.; Radoňák, J. Classic and New Markers in Diagnostics and Classification of Breast Cancer. Cancers 2022, 14, 5444. [Google Scholar] [CrossRef]
  8. Rassy, E.; Mosele, M.F.; Di Meglio, A.; Pistilli, B.; Andre, F. Precision Oncology in Patients with Breast Cancer: Towards a ‘Screen and Characterize’ Approach. ESMO Open 2024, 9, 103716. [Google Scholar] [CrossRef] [PubMed]
  9. Papalexis, P.; Georgakopoulou, V.E.; Drossos, P.V.; Thymara, E.; Nonni, A.; Lazaris, A.C.; Zografos, G.; Spandidos, D.; Kavantzas, N.; Thomopoulou, G.E. Precision Medicine in Breast Cancer (Review). Mol. Clin. Oncol. 2024, 21, 78. [Google Scholar] [CrossRef] [PubMed]
  10. World Health Organization (WHO). Breast Cancer. Available online: https://www.who.int/news-room/fact-sheets/detail/breast-cancer (accessed on 29 October 2025).
  11. Kim, J.; Harper, A.; McCormack, V.; Sung, H.; Houssami, N.; Morgan, E.; Mutebi, M.; Garvey, G.; Soerjomataram, I.; Fidler-Benaoudia, M.M. Global Patterns and Trends in Breast Cancer Incidence and Mortality across 185 Countries. Nat. Med. 2025, 31, 1154–1162. [Google Scholar] [CrossRef]
  12. Ferlay, J.; Ervik, M.; Lam, F.; Laversanne, M.; Colombet, M.; Mery, L.; Piñeros, M.; Znaor, A.; Soerjomataram, I.; Bray, F. Global Cancer Observatory: Cancer Today (Version 1.1). Available online: https://gco.iarc.who.int/today (accessed on 29 October 2025).
  13. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global Cancer Statistics 2022: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  14. Łukasiewicz, S.; Czeczelewski, M.; Forma, A.; Baj, J.; Sitarz, R.; Stanisławek, A. Breast Cancer-Epidemiology, Risk Factors, Classification, Prognostic Markers, and Current Treatment Strategies-An Updated Review. Cancers 2021, 13, 4287. [Google Scholar] [CrossRef]
  15. Endogenous Hormones and Breast Cancer Collaborative Group; Key, T.J.; Appleby, P.N.; Reeves, G.K.; Travis, R.C.; Alberg, A.J.; Barricarte, A.; Berrino, F.; Krogh, V.; Sieri, S.; et al. Sex Hormones and Risk of Breast Cancer in Premenopausal Women: A Collaborative Reanalysis of Individual Participant Data from Seven Prospective Studies. Lancet Oncol. 2013, 14, 1009–1019. [Google Scholar] [CrossRef]
  16. Benz, C.C. Impact of Aging on the Biology of Breast Cancer. Crit. Rev. Oncol. Hematol. 2008, 66, 65–74. [Google Scholar] [CrossRef]
  17. McGuire, A.; Brown, J.; Malone, C.; McLaughlin, R.; Kerin, M. Effects of Age on the Detection and Management of Breast Cancer. Cancers 2015, 7, 908–929. [Google Scholar] [CrossRef]
  18. Brewer, H.R.; Jones, M.E.; Schoemaker, M.J.; Ashworth, A.; Swerdlow, A.J. Family History and Risk of Breast Cancer: An Analysis Accounting for Family Structure. Breast Cancer Res. Treat. 2017, 165, 193–200. [Google Scholar] [CrossRef]
  19. Liu, L.; Hao, X.; Song, Z.; Zhi, X.; Zhang, S.; Zhang, J. Correlation between Family History and Characteristics of Breast Cancer. Sci. Rep. 2021, 11, 6360. [Google Scholar] [CrossRef]
  20. Shiovitz, S.; Korde, L.A. Genetics of Breast Cancer: A Topic in Evolution. Ann. Oncol. 2015, 26, 1291–1299. [Google Scholar] [CrossRef]
  21. Baretta, Z.; Mocellin, S.; Goldin, E.; Olopade, O.I.; Huo, D. Effect of BRCA Germline Mutations on Breast Cancer Prognosis. Medicine 2016, 95, e4975. [Google Scholar] [CrossRef]
  22. Hill, D.A.; Prossnitz, E.R.; Royce, M.; Nibbe, A. Temporal Trends in Breast Cancer Survival by Race and Ethnicity: A Population-Based Cohort Study. PLoS ONE 2019, 14, e0224064. [Google Scholar] [CrossRef]
  23. Yedjou, C.G.; Sims, J.N.; Miele, L.; Noubissi, F.; Lowe, L.; Fonseca, D.D.; Alo, R.A.; Payton, M.; Tchounwou, P.B. Health and Racial Disparity in Breast Cancer. Adv. Exp. Med. Biol. 2019, 1152, 31–49. [Google Scholar] [CrossRef] [PubMed]
  24. American Cancer Society. Breast Cancer Facts & Figures 2024–2025; American Cancer Society: Atlanta, GA, USA, 2024. [Google Scholar]
  25. Collaborative Group on Hormonal Factors in Breast Cancer Menarche, Menopause, and Breast Cancer Risk: Individual Participant Meta-Analysis, Including 118 964 Women with Breast Cancer from 117 Epidemiological Studies. Lancet Oncol. 2012, 13, 1141–1151. [CrossRef]
  26. Bodewes, F.T.H.; van Asselt, A.A.; Dorrius, M.D.; Greuter, M.J.W.; de Bock, G.H. Mammographic Breast Density and the Risk of Breast Cancer: A Systematic Review and Meta-Analysis. Breast 2022, 66, 62–68. [Google Scholar] [CrossRef]
  27. Mokhtary, A.; Karakatsanis, A.; Valachis, A. Mammographic Density Changes over Time and Breast Cancer Risk: A Systematic Review and Meta-Analysis. Cancers 2021, 13, 4805. [Google Scholar] [CrossRef]
  28. Schacht, D.V.; Yamaguchi, K.; Lai, J.; Kulkarni, K.; Sennett, C.A.; Abe, H. Importance of a Personal History of Breast Cancer as a Risk Factor for the Development of Subsequent Breast Cancer: Results From Screening Breast MRI. Am. J. Roentgenol. 2014, 202, 289–292. [Google Scholar] [CrossRef] [PubMed]
  29. Dyrstad, S.W.; Yan, Y.; Fowler, A.M.; Colditz, G.A. Breast Cancer Risk Associated with Benign Breast Disease: Systematic Review and Meta-Analysis. Breast Cancer Res. Treat. 2015, 149, 569–575. [Google Scholar] [CrossRef]
  30. Hou, N.; Wang, Z.; Ling, Y.; Hou, G.; Zhang, B.; Zhang, X.; Shi, M.; Chu, Z.; Wang, Y.; Hu, J.; et al. Radiotherapy and Increased Risk of Second Primary Cancers in Breast Cancer Survivors: An Epidemiological and Large Cohort Study. Breast 2024, 78, 103824. [Google Scholar] [CrossRef]
  31. Subramani, R.; Lakshmanaswamy, R. Pregnancy and Breast Cancer. Prog. Mol. Biol. Transl. Sci. 2017, 151, 81–111. [Google Scholar]
  32. Stordal, B. Breastfeeding Reduces the Risk of Breast Cancer: A Call for Action in High-income Countries with Low Rates of Breastfeeding. Cancer Med. 2023, 12, 4616–4625. [Google Scholar] [CrossRef]
  33. Migliavacca Zucchetti, B.; Peccatori, F.A.; Codacci-Pisanelli, G. Pregnancy and Lactation: Risk or Protective Factors for Breast Cancer? In Diseases of the Breast During Pregnancy and Lactation; Advances in Experimental Medicine and Biology; Alipour, S., Omranipour, R., Eds.; Springer: Cham, Switzerland, 2020; Volume 1252, pp. 195–197. [Google Scholar]
  34. Narod, S.A. Hormone Replacement Therapy and the Risk of Breast Cancer. Nat. Rev. Clin. Oncol. 2011, 8, 669–676. [Google Scholar] [CrossRef] [PubMed]
  35. Vinogradova, Y.; Coupland, C.; Hippisley-Cox, J. Use of Hormone Replacement Therapy and Risk of Breast Cancer: Nested Case-Control Studies Using the QResearch and CPRD Databases. BMJ 2020, 371, m3873. [Google Scholar] [CrossRef]
  36. Hoover, R.N.; Hyer, M.; Pfeiffer, R.M.; Adam, E.; Bond, B.; Cheville, A.L.; Colton, T.; Hartge, P.; Hatch, E.E.; Herbst, A.L.; et al. Adverse Health Outcomes in Women Exposed In Utero to Diethylstilbestrol. N. Engl. J. Med. 2011, 365, 1304–1314. [Google Scholar] [CrossRef]
  37. Hilakivi-Clarke, L. Maternal Exposure to Diethylstilbestrol during Pregnancy and Increased Breast Cancer Risk in Daughters. Breast Cancer Res. 2014, 16, 208. [Google Scholar] [CrossRef]
  38. Chen, X.; Wang, Q.; Zhang, Y.; Xie, Q.; Tan, X. Physical Activity and Risk of Breast Cancer: A Meta-Analysis of 38 Cohort Studies in 45 Study Reports. Value Health 2019, 22, 104–128. [Google Scholar] [CrossRef]
  39. Dixon-Suen, S.C.; Lewis, S.J.; Martin, R.M.; English, D.R.; Boyle, T.; Giles, G.G.; Michailidou, K.; Bolla, M.K.; Wang, Q.; Dennis, J.; et al. Physical Activity, Sedentary Time and Breast Cancer Risk: A Mendelian Randomisation Study. Br. J. Sports Med. 2022, 56, 1157–1170. [Google Scholar] [CrossRef] [PubMed]
  40. Boraka, Ö.; Klintman, M.; Rosendahl, A.H. Physical Activity and Long-Term Risk of Breast Cancer, Associations with Time in Life and Body Composition in the Prospective Malmö Diet and Cancer Study. Cancers 2022, 14, 1960. [Google Scholar] [CrossRef]
  41. Dehesh, T.; Fadaghi, S.; Seyedi, M.; Abolhadi, E.; Ilaghi, M.; Shams, P.; Ajam, F.; Mosleh-Shirazi, M.A.; Dehesh, P. The Relation between Obesity and Breast Cancer Risk in Women by Considering Menstruation Status and Geographical Variations: A Systematic Review and Meta-Analysis. BMC Women’s Health 2023, 23, 392. [Google Scholar] [CrossRef] [PubMed]
  42. Starek-Świechowicz, B.; Budziszewska, B.; Starek, A. Alcohol and Breast Cancer. Pharmacol. Rep. 2023, 75, 69–84. [Google Scholar] [CrossRef]
  43. He, Y.; Si, Y.; Li, X.; Hong, J.; Yu, C.; He, N. The Relationship between Tobacco and Breast Cancer Incidence: A Systematic Review and Meta-Analysis of Observational Studies. Front. Oncol. 2022, 12, 961970. [Google Scholar] [CrossRef] [PubMed]
  44. Hossain, S.; Beydoun, M.A.; Beydoun, H.A.; Chen, X.; Zonderman, A.B.; Wood, R.J. Vitamin D and Breast Cancer: A Systematic Review and Meta-Analysis of Observational Studies. Clin. Nutr. ESPEN 2019, 30, 170–184. [Google Scholar] [CrossRef]
  45. Luo, Z.; Liu, Z.; Chen, H.; Liu, Y.; Tang, N.; Li, H. Light at Night Exposure and Risk of Breast Cancer: A Meta-Analysis of Observational Studies. Front. Public Health 2023, 11, 1276290. [Google Scholar] [CrossRef]
  46. Ma, S.; Alsabawi, Y.; El-Serag, H.B.; Thrift, A.P. Exposure to Light at Night and Risk of Cancer: A Systematic Review, Meta-Analysis, and Data Synthesis. Cancers 2024, 16, 2653. [Google Scholar] [CrossRef]
  47. Fiolet, T.; Srour, B.; Sellem, L.; Kesse-Guyot, E.; Allès, B.; Méjean, C.; Deschasaux, M.; Fassier, P.; Latino-Martel, P.; Beslay, M.; et al. Consumption of Ultra-Processed Foods and Cancer Risk: Results from NutriNet-Santé Prospective Cohort. BMJ 2018, 360, k322. [Google Scholar] [CrossRef] [PubMed]
  48. Ugalde-Resano, R.; Gamboa-Loira, B.; Mérida-Ortega, Á.; Rincón-Rubio, A.; Flores-Collado, G.; Piña-Pozas, M.; López-Carrillo, L. Biological Concentrations of DDT Metabolites and Breast Cancer Risk: An Updated Systematic Review and Meta-Analysis. Rev. Environ. Health 2024, 40, 225–236. [Google Scholar] [CrossRef]
  49. Wang, R.; Wang, P.; Zhou, Y.; Wang, Y.; Xu, C.; Wang, Z.; Wang, W. Association between Long-Term Ambient Air Pollution Exposure and the Incidence of Breast Cancer: A Meta-Analysis Based on Updated Evidence. Ecotoxicol. Environ. Saf. 2025, 289, 117472. [Google Scholar] [CrossRef]
  50. McDonald, J.A.; Goyal, A.; Terry, M.B. Alcohol Intake and Breast Cancer Risk: Weighing the Overall Evidence. Curr. Breast Cancer Rep. 2013, 5, 208–221. [Google Scholar] [CrossRef]
  51. Mesnage, R.; Phedonos, A.; Arno, M.; Balu, S.; Corton, J.C.; Antoniou, M.N. Editor’s Highlight: Transcriptome Profiling Reveals Bisphenol A Alternatives Activate Estrogen Receptor Alpha in Human Breast Cancer Cells. Toxicol. Sci. 2017, 158, 431–443. [Google Scholar] [CrossRef] [PubMed]
  52. Stordal, B.; Harvie, M.; Antoniou, M.N.; Bellingham, M.; Chan, D.S.M.; Darbre, P.; Karlsson, O.; Kortenkamp, A.; Magee, P.; Mandriota, S.; et al. Breast Cancer Risk and Prevention in 2024: An Overview from the Breast Cancer UK-Breast Cancer Prevention Conference. Cancer Med. 2024, 13, e70255. [Google Scholar] [CrossRef]
  53. Collaborative Group on Hormonal Factors in Breast Cancer. Breast Cancer and Breastfeeding: Collaborative Reanalysis of Individual Data from 47 Epidemiological Studies in 30 Countries, Including 50 302 Women with Breast Cancer and 96 973 Women without the Disease. Lancet 2002, 360, 187–195. [Google Scholar] [CrossRef] [PubMed]
  54. Teras, L.R.; Patel, A.V.; Wang, M.; Yaun, S.-S.; Anderson, K.; Brathwaite, R.; Caan, B.J.; Chen, Y.; Connor, A.E.; Eliassen, A.H.; et al. Sustained Weight Loss and Risk of Breast Cancer in Women 50 Years and Older: A Pooled Analysis of Prospective Data. J. Natl. Cancer Inst. 2020, 112, 929–937. [Google Scholar] [CrossRef]
  55. Soldado-Gordillo, A.; Álvarez-Mercado, A.I. Epigenetics, Microbiota, and Breast Cancer: A Systematic Review. Life 2024, 14, 705. [Google Scholar] [CrossRef]
  56. Yang, H.; Fang, Y.; Wang, H.; Lu, T.; Chen, Q.; Liu, H. Progress in Epigenetic Research of Breast Cancer: A Bibliometric Analysis since the 2000s. Front. Oncol. 2025, 15, 1619346. [Google Scholar] [CrossRef]
  57. Chen, Y.; Zhu, H.; Luo, Y.; Tong, S.; Liu, Y. EZH2: The Roles in Targeted Therapy and Mechanisms of Resistance in Breast Cancer. Biomed. Pharmacother. 2024, 175, 116624. [Google Scholar] [CrossRef]
  58. Cortellesi, E.; Savini, I.; Veneziano, M.; Gambacurta, A.; Catani, M.V.; Gasperi, V. Decoding the Epigenome of Breast Cancer. Int. J. Mol. Sci. 2025, 26, 2605. [Google Scholar] [CrossRef]
  59. Saha, S.; Mahapatra, S.; Khanra, S.; Mishra, B.; Swain, B.; Malhotra, D.; Saha, S.; Panda, V.K.; Kumari, K.; Jena, S.; et al. Decoding Breast Cancer Treatment Resistance through Genetic, Epigenetic, and Immune-Regulatory Mechanisms: From Molecular Insights to Translational Perspectives. Cancer Drug Resist. 2025, 8, 36. [Google Scholar] [CrossRef]
  60. Srinivasan, P.; Bandlamudi, C.; Jonsson, P.; Kemel, Y.; Chavan, S.S.; Richards, A.L.; Penson, A.V.; Bielski, C.M.; Fong, C.; Syed, A.; et al. The Context-Specific Role of Germline Pathogenicity in Tumorigenesis. Nat. Genet. 2021, 53, 1577–1585. [Google Scholar] [CrossRef] [PubMed]
  61. Cardoso, F.; Paluch-Shimon, S.; Schumacher-Wulf, E.; Matos, L.; Gelmon, K.; Aapro, M.S.; Bajpai, J.; Barrios, C.H.; Bergh, J.; Bergsten-Nordström, E.; et al. 6th and 7th International Consensus Guidelines for the Management of Advanced Breast Cancer (ABC Guidelines 6 and 7). Breast 2024, 76, 103756. [Google Scholar] [CrossRef] [PubMed]
  62. Park, K.H.; Choi, J.Y.; Lim, A.-R.; Kim, J.W.; Choi, Y.J.; Lee, S.; Sung, J.S.; Chung, H.-J.; Jang, B.; Yoon, D.; et al. Genomic Landscape and Clinical Utility in Korean Advanced Pan-Cancer Patients from Prospective Clinical Sequencing: K-MASTER Program. Cancer Discov. 2022, 12, 938–948. [Google Scholar] [CrossRef] [PubMed]
  63. Petrucelli, N.; Daly, M.B.; Pal, T. BRCA1- and BRCA2-Associated Hereditary Breast and Ovarian Cancer; Adam, M.P., Feldman, J., Mirzaa, G.M., Pagon, R.A., Wallace, S.E., Eds.; GeneReviews, University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  64. Schneider, K.; Zelley, K.; Nichols, K.E.; Schwartz Levine, A.; Garber, J. Li-Fraumeni Syndrome; Adam, M.P., Fedman, J., MIrzaa, G.M., Pagon, R.A., Wallace, S.E., Eds.; GeneReviews, University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  65. Subaşıoğlu, A.; Güç, Z.G.; Gür, E.Ö.; Tekindal, M.A.; Atahan, M.K. Genetic, Surgical and Oncological Approach to Breast Cancer, with BRCA1, BRCA2, CDH1, PALB2, PTEN and TP53 Variants. Eur. J. Breast Health 2023, 19, 55–69. [Google Scholar] [CrossRef]
  66. Hung, C.-C.; Moi, S.-H.; Huang, H.-I.; Hsiao, T.-H.; Huang, C.-C. Polygenic Risk Score-Based Prediction of Breast Cancer Risk in Taiwanese Women with Dense Breast Using a Retrospective Cohort Study. Sci. Rep. 2024, 14, 6324. [Google Scholar] [CrossRef]
  67. Ruberu, T.L.M.; Braun, D.; Parmigiani, G.; Biswas, S. Meta-Analysis of Breast Cancer Risk for Individuals with PALB2 Pathogenic Variants. Genet. Epidemiol. 2024, 48, 448–454. [Google Scholar] [CrossRef]
  68. Hanson, H.; Pal, T.; Tischkowitz, M.; Stewart, D. CHEK2-Related Cancer Predisposition; Adam, M.P., Feldman, J., Mirzaa, G.M., Pagon, R.A., Wallace, S.E., Eds.; GeneReviews, University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  69. Seca, M.; Narod, S.A. Breast Cancer and ATM Mutations: Treatment Implications. Hered. Cancer Clin. Pract. 2024, 22, 26. [Google Scholar] [CrossRef]
  70. Roberts, E.; Howell, S.; Evans, D.G. Polygenic Risk Scores and Breast Cancer Risk Prediction. Breast 2023, 67, 71–77. [Google Scholar] [CrossRef]
  71. Mbuya-Bienge, C.; Pashayan, N.; Kazemali, C.D.; Lapointe, J.; Simard, J.; Nabi, H. A Systematic Review and Critical Assessment of Breast Cancer Risk Prediction Tools Incorporating a Polygenic Risk Score for the General Population. Cancers 2023, 15, 5380. [Google Scholar] [CrossRef] [PubMed]
  72. Wu, J.; Mamidi, T.K.K.; Zhang, L.; Hicks, C. Deconvolution of the Genomic and Epigenomic Interaction Landscape of Triple-Negative Breast Cancer. Cancers 2019, 11, 1692. [Google Scholar] [CrossRef]
  73. Hayama, S.; Nakamura, R.; Ishige, T.; Sangai, T.; Sakakibara, M.; Fujimoto, H.; Ishigami, E.; Masuda, T.; Nakagawa, A.; Teranaka, R.; et al. The Impact of PIK3CA Mutations and PTEN Expression on the Effect of Neoadjuvant Therapy for Postmenopausal Luminal Breast Cancer Patients. BMC Cancer 2023, 23, 384. [Google Scholar] [CrossRef] [PubMed]
  74. Tada, H.; Miyashita, M.; Harada-Shoji, N.; Ebata, A.; Sato, M.; Motonari, T.; Yanagaki, M.; Kon, T.; Sakamoto, A.; Ishida, T. Clinicopathogenomic Analysis of PI3K/AKT/PTEN-Altered Luminal Metastatic Breast Cancer in Japan. Breast Cancer 2025, 32, 208–216. [Google Scholar] [CrossRef] [PubMed]
  75. Gautam, S.; Maurya, R.; Vikal, A.; Patel, P.; Thakur, S.; Singh, A.; Gupta, G.D.; Kurmi, B. Das Understanding Drug Resistance in Breast Cancer: Mechanisms and Emerging Therapeutic Strategies. Med. Drug Discov. 2025, 26, 100210. [Google Scholar] [CrossRef]
  76. Bahar, M.E.; Kim, H.J.; Kim, D.R. Targeting the RAS/RAF/MAPK Pathway for Cancer Therapy: From Mechanism to Clinical Studies. Signal Transduct. Target. Ther. 2023, 8, 455. [Google Scholar] [CrossRef]
  77. Song, P.; Gao, Z.; Bao, Y.; Chen, L.; Huang, Y.; Liu, Y.; Dong, Q.; Wei, X. Wnt/β-Catenin Signaling Pathway in Carcinogenesis and Cancer Therapy. J. Hematol. Oncol. 2024, 17, 46. [Google Scholar] [CrossRef]
  78. Van Schie, E.H.; Van Amerongen, R. Aberrant WNT/CTNNB1 Signaling as a Therapeutic Target in Human Breast Cancer: Weighing the Evidence. Front. Cell Dev. Biol. 2020, 8, 25. [Google Scholar] [CrossRef]
  79. Zhang, X. Molecular Classification of Breast Cancer: Relevance and Challenges. Arch. Pathol. Lab. Med. 2023, 147, 46–51. [Google Scholar] [CrossRef]
  80. Perou, C.M.; Sørlie, T.; Eisen, M.B.; Van De Rijn, M.; Jeffrey, S.S.; Ress, C.A.; Pollack, J.R.; Ross, D.T.; Johnsen, H.; Akslen, L.A.; et al. Molecular Portraits of Human Breast Tumours. Nature 2000, 406, 747–752. [Google Scholar] [CrossRef]
  81. Sørlie, T.; Perou, C.M.; Tibshirani, R.; Aas, T.; Geisler, S.; Johnsen, H.; Hastie, T.; Eisen, M.B.; Van De Rijn, M.; Jeffrey, S.S.; et al. Gene Expression Patterns of Breast Carcinomas Distinguish Tumor Subclasses with Clinical Implications. Proc. Natl. Acad. Sci. USA 2001, 98, 10869. [Google Scholar] [CrossRef] [PubMed]
  82. Prat, A.; Perou, C.M. Deconstructing the Molecular Portraits of Breast Cancer. Mol. Oncol. 2011, 5, 5–23. [Google Scholar] [CrossRef]
  83. Provenzano, E.; Ulaner, G.A.; Chin, S.F. Molecular Classification of Breast Cancer. PET Clin. 2018, 13, 325–338. [Google Scholar] [CrossRef]
  84. Cancer Genome Atlas Network. Comprehensive Molecular Portraits of Human Breast Tumors. Nature 2012, 490, 61. [Google Scholar] [CrossRef]
  85. Herschkowitz, J.I.; Simin, K.; Weigman, V.J.; Mikaelian, I.; Usary, J.; Hu, Z.; Rasmussen, K.E.; Jones, L.P.; Assefnia, S.; Chandrasekharan, S.; et al. Identification of Conserved Gene Expression Features between Murine Mammary Carcinoma Models and Human Breast Tumors. Genome Biol. 2007, 8, R76. [Google Scholar] [CrossRef] [PubMed]
  86. Goldhirsch, A.; Winer, E.P.; Coates, A.S.; Gelber, R.D.; Piccart-Gebhart, M.; Thürlimann, B.; Senn, H.J.; Albain, K.S.; André, F.; Bergh, J.; et al. Personalizing the Treatment of Women with Early Breast Cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann. Oncol. 2013, 24, 2206. [Google Scholar] [CrossRef]
  87. Roy, M.; Fowler, A.M.; Ulaner, G.A.; Mahajan, A. Molecular Classification of Breast Cancer. PET Clin. 2023, 18, 441–458. [Google Scholar] [CrossRef] [PubMed]
  88. Makki, J. Diversity of Breast Carcinoma: Histological Subtypes and Clinical Relevance. Clin. Med. Insights Pathol. 2015, 8, 23. [Google Scholar] [CrossRef] [PubMed]
  89. Eroles, P.; Bosch, A.; Alejandro Pérez-Fidalgo, J.; Lluch, A. Molecular Biology in Breast Cancer: Intrinsic Subtypes and Signaling Pathways. Cancer Treat. Rev. 2012, 38, 698–707. [Google Scholar] [CrossRef]
  90. Ades, F.; Zardavas, D.; Bozovic-Spasojevic, I.; Pugliano, L.; Fumagalli, D.; De Azambuja, E.; Viale, G.; Sotiriou, C.; Piccart, M. Luminal B Breast Cancer: Molecular Characterization, Clinical Management, and Future Perspectives. J. Clin. Oncol. 2014, 32, 2794–2803. [Google Scholar] [CrossRef] [PubMed]
  91. Rediti, M.; Venet, D.; Joaquin Garcia, A.; Maetens, M.; Vincent, D.; Majjaj, S.; El-Abed, S.; Di Cosimo, S.; Ueno, T.; Izquierdo, M.; et al. Identification of HER2-Positive Breast Cancer Molecular Subtypes with Potential Clinical Implications in the ALTTO Clinical Trial. Nat. Commun. 2024, 15, 10402. [Google Scholar] [CrossRef]
  92. Carvalho, E.; Canberk, S.; Schmitt, F.; Vale, N. Molecular Subtypes and Mechanisms of Breast Cancer: Precision Medicine Approaches for Targeted Therapies. Cancers 2025, 17, 1102. [Google Scholar] [CrossRef]
  93. Collignon, J.; Lousberg, L.; Schroeder, H.; Jerusalem, G. Triple-Negative Breast Cancer: Treatment Challenges and Solutions. Breast Cancer Targets Ther. 2016, 8, 93. [Google Scholar] [CrossRef]
  94. Yin, L.; Duan, J.J.; Bian, X.W.; Yu, S.C. Triple-Negative Breast Cancer Molecular Subtyping and Treatment Progress. Breast Cancer Res. 2020, 22, 61. [Google Scholar] [CrossRef]
  95. Plasilova, M.L.; Hayse, B.; Killelea, B.K.; Horowitz, N.R.; Chagpar, A.B.; Lannin, D.R. Features of Triple-Negative Breast Cancer: Analysis of 38,813 Cases from the National Cancer Database. Medicine 2016, 95, e4614. [Google Scholar] [CrossRef]
  96. Jiang, Y.Z.; Ma, D.; Suo, C.; Shi, J.; Xue, M.; Hu, X.; Xiao, Y.; Yu, K.D.; Liu, Y.R.; Yu, Y.; et al. Genomic and Transcriptomic Landscape of Triple-Negative Breast Cancers: Subtypes and Treatment Strategies. Cancer Cell 2019, 35, 428–440.e5. [Google Scholar] [CrossRef]
  97. Prat, A.; Parker, J.S.; Karginova, O.; Fan, C.; Livasy, C.; Herschkowitz, J.I.; He, X.; Perou, C.M. Phenotypic and Molecular Characterization of the Claudin-Low Intrinsic Subtype of Breast Cancer. Breast Cancer Res. 2010, 12, R68. [Google Scholar] [CrossRef]
  98. Neves Rebello Alves, L.; Dummer Meira, D.; Poppe Merigueti, L.; Correia Casotti, M.; do Prado Ventorim, D.; Ferreira Figueiredo Almeida, J.; Pereira de Sousa, V.; Cindra Sant’Ana, M.; Gonçalves Coutinho da Cruz, R.; Santos Louro, L.; et al. Biomarkers in Breast Cancer: An Old Story with a New End. Genes 2023, 14, 1364. [Google Scholar] [CrossRef] [PubMed]
  99. Barzaman, K.; Karami, J.; Zarei, Z.; Hosseinzadeh, A.; Kazemi, M.H.; Moradi-Kalbolandi, S.; Safari, E.; Farahmand, L. Breast Cancer: Biology, Biomarkers, and Treatments. Int. Immunopharmacol. 2020, 84, 106535. [Google Scholar] [CrossRef]
  100. Moar, K.; Pant, A.; Saini, V.; Pandey, M.; Maurya, P.K. Potential Diagnostic and Prognostic Biomarkers for Breast Cancer: A Compiled Review. Pathol. Res. Pract. 2023, 251, 154893. [Google Scholar] [CrossRef]
  101. Clusan, L.; Ferrière, F.; Flouriot, G.; Pakdel, F. A Basic Review on Estrogen Receptor Signaling Pathways in Breast Cancer. Int. J. Mol. Sci. 2023, 24, 6834. [Google Scholar] [CrossRef] [PubMed]
  102. Porras, L.; Ismail, H.; Mader, S. Positive Regulation of Estrogen Receptor Alpha in Breast Tumorigenesis. Cells 2021, 10, 2966. [Google Scholar] [CrossRef]
  103. Maqsood, Q.; Khan, M.U.; Fatima, T.; Khalid, S.; Malik, Z.I. Recent Insights Into Breast Cancer: Molecular Pathways, Epigenetic Regulation, and Emerging Targeted Therapies. Breast Cancer 2025, 19, 11782234251355663. [Google Scholar] [CrossRef]
  104. Mal, R.; Magner, A.; David, J.; Datta, J.; Vallabhaneni, M.; Kassem, M.; Manouchehri, J.; Willingham, N.; Stover, D.; Vandeusen, J.; et al. Estrogen Receptor Beta (ERβ): A Ligand Activated Tumor Suppressor. Front. Oncol. 2020, 10, 587386. [Google Scholar] [CrossRef] [PubMed]
  105. Arao, Y.; Korach, K.S. The Physiological Role of Estrogen Receptor Functional Domains. Essays Biochem. 2021, 65, 867–875. [Google Scholar] [CrossRef]
  106. Li, Z.; Wei, H.; Li, S.; Wu, P.; Mao, X. The Role of Progesterone Receptors in Breast Cancer. Drug Des. Devel. Ther. 2022, 16, 305–314. [Google Scholar] [CrossRef]
  107. Vang, A.; Salem, K.; Fowler, A.M. Progesterone Receptor Gene Polymorphisms and Breast Cancer Risk. Endocrinology 2023, 164, bqad020. [Google Scholar] [CrossRef]
  108. Azeez, J.M.; Susmi, T.R.; Remadevi, V.; Ravindran, V.; Sasikumar, S.A.; Ayswarya, R.n.S. Sreeja Sreeharshan New Insights into the Functions of Progesterone Receptor (PR) Isoforms and Progesterone Signaling. Am. J. Cancer Res. 2021, 11, 5214–5232. [Google Scholar]
  109. Heinlein, C.A.; Chang, C. Androgen Receptor (AR) Coregulators: An Overview. Endocr. Rev. 2002, 23, 175–200. [Google Scholar] [CrossRef]
  110. Kolyvas, E.A.; Caldas, C.; Kelly, K.; Ahmad, S.S. Androgen Receptor Function and Targeted Therapeutics across Breast Cancer Subtypes. Breast Cancer Res. 2022, 24, 79. [Google Scholar] [CrossRef] [PubMed]
  111. Hackbart, H.; Cui, X.; Lee, J.S. Androgen Receptor in Breast Cancer and Its Clinical Implication. Transl. Breast Cancer Res. 2023, 4, 30. [Google Scholar] [CrossRef]
  112. Zhu, K.; Yang, X.; Tai, H.; Zhong, X.; Luo, T.; Zheng, H. HER2-Targeted Therapies in Cancer: A Systematic Review. Biomark. Res. 2024, 12, 16. [Google Scholar] [CrossRef]
  113. Dieci, M.V.; Miglietta, F.; Griguolo, G.; Guarneri, V. Biomarkers for HER2-Positive Metastatic Breast Cancer: Beyond Hormone Receptors. Cancer Treat. Rev. 2020, 88, 102064. [Google Scholar] [CrossRef]
  114. Marchiò, C.; Annaratone, L.; Marques, A.; Casorzo, L.; Berrino, E.; Sapino, A. Evolving Concepts in HER2 Evaluation in Breast Cancer: Heterogeneity, HER2-Low Carcinomas and Beyond. Semin. Cancer Biol. 2021, 72, 123–135. [Google Scholar] [CrossRef]
  115. Samanta, A.; Karim, A.; Ahamed, A.; Hassan, M.K.; Alam, S.S.M.; Ali, S.; Hoque, M. Damaging Non-Synonymous Mutations in the Extracellular Domain of HER2 Potentially Alter the Efficacy of Herceptin-Mediated Breast Cancer Therapy. Egypt. J. Med. Hum. Genet. 2025, 26, 149. [Google Scholar] [CrossRef]
  116. Zhang, Y.; Fan, W.; Su, F.; Zhang, X.; Du, Y.; Li, W.; Gao, Y.; Hu, W.; Zhao, J. Discussion on the Mechanism of HER2 Resistance in Esophagogastric Junction and Gastric Cancer in the Era of Immunotherapy. Hum. Vaccines Immunother. 2025, 21, 2459458. [Google Scholar] [CrossRef] [PubMed]
  117. Ishiyama, N.; O’Connor, M.; Salomatov, A.; Romashko, D.; Thakur, S.; Mentes, A.; Hopkins, J.F.; Frampton, G.M.; Albacker, L.A.; Kohlmann, A.; et al. Computational and Functional Analyses of HER2 Mutations Reveal Allosteric Activation Mechanisms and Altered Pharmacologic Effects. Cancer Res. 2023, 83, 1531–1542. [Google Scholar] [CrossRef] [PubMed]
  118. Gaibar, M.; Beltrán, L.; Romero-Lorca, A.; Fernández-Santander, A.; Novillo, A. Somatic Mutations in HER2 and Implications for Current Treatment Paradigms in HER2-Positive Breast Cancer. J. Oncol. 2020, 2020, 6375956. [Google Scholar] [CrossRef] [PubMed]
  119. Zhang, A.; Wang, X.; Fan, C.; Mao, X. The Role of Ki67 in Evaluating Neoadjuvant Endocrine Therapy of Hormone Receptor-Positive Breast Cancer. Front. Endocrinol. 2021, 12, 687244. [Google Scholar] [CrossRef]
  120. Kreipe, H.; Harbeck, N.; Christgen, M. Clinical Validity and Clinical Utility of Ki67 in Early Breast Cancer. Ther. Adv. Med. Oncol. 2022, 14, 17588359221122725. [Google Scholar] [CrossRef]
  121. Smith, I.; Robertson, J.; Kilburn, L.; Wilcox, M.; Evans, A.; Holcombe, C.; Horgan, K.; Kirwan, C.; Mallon, E.; Sibbering, M.; et al. Long-Term Outcome and Prognostic Value of Ki67 after Perioperative Endocrine Therapy in Postmenopausal Women with Hormone-Sensitive Early Breast Cancer (POETIC): An Open-Label, Multicentre, Parallel-Group, Randomised, Phase 3 Trial. Lancet Oncol. 2020, 21, 1443–1454, Erratum in Lancet Oncol. 2020, 21, e553. https://doi.org/10.1016/S1470-2045(20)30677-X. [Google Scholar] [CrossRef] [PubMed]
  122. Li, W.; Lu, N.; Chen, C.; Lu, X. Identifying the Optimal Cutoff Point of Ki-67 in Breast Cancer: A Single-Center Experience. J. Int. Med. Res. 2023, 51, 03000605231195468. [Google Scholar] [CrossRef]
  123. Nielsen, T.O.; Leung, S.C.Y.; Rimm, D.L.; Dodson, A.; Acs, B.; Badve, S.; Denkert, C.; Ellis, M.J.; Fineberg, S.; Flowers, M.; et al. Assessment of Ki67 in Breast Cancer: Updated Recommendations From the International Ki67 in Breast Cancer Working Group. J. Natl. Cancer Inst. 2021, 113, 808–819. [Google Scholar] [CrossRef]
  124. Escala-Cornejo, R.; Olivares-Hernández, A.; García Muñoz, M.; Figuero-Pérez, L.; Vallejo, J.M.; Miramontes-González, J.P.; Sancho de Salas, M.; Gómez Muñoz, M.A.; Tamayo, R.S.; García, G.M.; et al. Identifying the Best Ki-67 Cut-Off for Determining Luminal Breast Cancer Subtypes Using Immunohistochemical Analysis and PAM50 Genomic Classification. EMJ Oncol. 2020, 8, 47–48. [Google Scholar] [CrossRef]
  125. Hernández Borrero, L.J.; El-Deiry, W.S. Tumor Suppressor P53: Biology, Signaling Pathways, and Therapeutic Targeting. Biochim. Biophys. Acta (BBA)-Rev. Cancer 2021, 1876, 188556. [Google Scholar] [CrossRef]
  126. Hwang, S.H.; Baek, S.H.; Lee, M.J.; Kook, Y.; Bae, S.J.; Ahn, S.G.; Jeong, J. Clinical Relevance of TP53 Mutation and Its Characteristics in Breast Cancer with Long-Term Follow-Up Date. Cancers 2024, 16, 3899. [Google Scholar] [CrossRef]
  127. Paulino, P.J.I.V.; Che Omar, M.T. Identification of High-Risk Signatures and Therapeutic Targets through Molecular Characterization and Immune Profiling of TP53-Mutant Breast Cancer. J. Genet. Eng. Biotechnol. 2025, 23, 100574. [Google Scholar] [CrossRef]
  128. Liu, Z.; Jiang, Z.; Gao, Y.; Wang, L.; Chen, C.; Wang, X. TP53 Mutations Promote Immunogenic Activity in Breast Cancer. J. Oncol. 2019, 2019, 5952836. [Google Scholar] [CrossRef]
  129. Tito, C.; Masciarelli, S.; Colotti, G.; Fazi, F. EGF Receptor in Organ Development, Tissue Homeostasis and Regeneration. J. Biomed. Sci. 2025, 32, 24. [Google Scholar] [CrossRef]
  130. Shyamsunder, S.; Lu, Z.; Takiar, V.; Waltz, S.E. Challenges and Resistance Mechanisms to EGFR Targeted Therapies in Head and Neck Cancers and Breast Cancer: Insights into RTK Dependent and Independent Mechanisms. Oncotarget 2025, 16, 508–530. [Google Scholar] [CrossRef]
  131. Li, X.; Zhao, L.; Chen, C.; Nie, J.; Jiao, B. Can EGFR Be a Therapeutic Target in Breast Cancer? Biochim. Biophys. Acta (BBA)-Rev. Cancer 2022, 1877, 188789. [Google Scholar] [CrossRef]
  132. Oshi, M.; Gandhi, S.; Tokumaru, Y.; Yan, L.; Yamada, A.; Matsuyama, R.; Ishikawa, T.; Endo, I.; Takabe, K. Conflicting Roles of EGFR Expression by Subtypes in Breast Cancer. Am. J. Cancer Res. 2021, 11, 5094–5110. [Google Scholar]
  133. Silva Rocha, F.; da Silva Maués, J.H.; Brito Lins Pereira, C.M.; Moreira-Nunes, C.A.; Rodriguez Burbano, R.M. Analysis of Increased EGFR and IGF-1R Signaling and Its Correlation with Socio-Epidemiological Features and Biological Profile in Breast Cancer Patients: A Study in Northern Brazil. Breast Cancer Targets Ther. 2021, 13, 325–339. [Google Scholar] [CrossRef] [PubMed]
  134. Uribe, M.L.; Marrocco, I.; Yarden, Y. EGFR in Cancer: Signaling Mechanisms, Drugs, and Acquired Resistance. Cancers 2021, 13, 2748. [Google Scholar] [CrossRef] [PubMed]
  135. Soni, U.K.; Jenny, L.; Hegde, R.S. IGF-1R Targeting in Cancer—Does Sub-Cellular Localization Matter? J. Exp. Clin. Cancer Res. 2023, 42, 273. [Google Scholar] [CrossRef]
  136. Dai, X.; Kaushik, A.C.; Zhang, J. The Emerging Role of Major Regulatory RNAs in Cancer Control. Front. Oncol. 2019, 9, 920. [Google Scholar] [CrossRef] [PubMed]
  137. Peng, Y.; Croce, C.M. The Role of MicroRNAs in Human Cancer. Signal Transduct. Target. Ther. 2016, 1, 15004. [Google Scholar] [CrossRef]
  138. Alles, J.; Fehlmann, T.; Fischer, U.; Backes, C.; Galata, V.; Minet, M.; Hart, M.; Abu-Halima, M.; Grässer, F.A.; Lenhof, H.P.; et al. An Estimate of the Total Number of True Human MiRNAs. Nucleic Acids Res. 2019, 47, 3353–3364. [Google Scholar] [CrossRef]
  139. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef]
  140. Vaschetto, L.M. MiRNA Activation Is an Endogenous Gene Expression Pathway. RNA Biol. 2018, 15, 826–828. [Google Scholar] [CrossRef]
  141. Babaei, G.; Raei, N.; Toofani Milani, A.; Gholizadeh-Ghaleh Aziz, S.; Pourjabbar, N.; Geravand, F. The Emerging Role of MiR-200 Family in Metastasis: Focus on EMT, CSCs, Angiogenesis, and Anoikis. Mol. Biol. Rep. 2021, 48, 6935–6947. [Google Scholar] [CrossRef]
  142. Gulati, R.; Mitra, T.; Rajiv, R.; Rajan, E.J.E.; Pierret, C.; Enninga, E.A.L.; Janardhanan, R. Exosomal MicroRNAs in Breast Cancer: Towards Theranostic Applications. Front. Mol. Biosci. 2024, 11, 1330144. [Google Scholar] [CrossRef] [PubMed]
  143. Huang, G.L.; Sun, J.; Lu, Y.; Liu, Y.; Cao, H.; Zhang, H.; Calin, G.A. MiR-200 Family and Cancer: From a Meta-Analysis View. Mol. Asp. Med. 2019, 70, 57–71. [Google Scholar] [CrossRef]
  144. Huang, S.K.; Luo, Q.; Peng, H.; Li, J.; Zhao, M.; Wang, J.; Gu, Y.Y.; Li, Y.; Yuan, P.; Zhao, G.H.; et al. A Panel of Serum Noncoding RNAs for the Diagnosis and Monitoring of Response to Therapy in Patients with Breast Cancer. Med. Sci. Monit. 2018, 24, 2476. [Google Scholar] [CrossRef] [PubMed]
  145. Ludwig, N.; Leidinger, P.; Becker, K.; Backes, C.; Fehlmann, T.; Pallasch, C.; Rheinheimer, S.; Meder, B.; Stähler, C.; Meese, E.; et al. Distribution of MiRNA Expression across Human Tissues. Nucleic Acids Res. 2016, 44, 3865–3877. [Google Scholar] [CrossRef] [PubMed]
  146. Jelski, W.; Okrasinska, S.; Mroczko, B. MicroRNAs as Biomarkers of Breast Cancer. Int. J. Mol. Sci. 2025, 26, 4395. [Google Scholar] [CrossRef]
  147. Sochor, M.; Basova, P.; Pesta, M.; Dusilkova, N.; Bartos, J.; Burda, P.; Pospisil, V.; Stopka, T. Oncogenic MicroRNAs: MiR-155, MiR-19a, MiR-181b, and MiR-24 Enable Monitoring of Early Breast Cancer in Serum. BMC Cancer 2014, 14, 448. [Google Scholar] [CrossRef]
  148. Chen, J.; Wang, X. MicroRNA-21 in Breast Cancer: Diagnostic and Prognostic Potential. Clin. Transl. Oncol. Off. Publ. Fed. Span. Oncol. Soc. 2014, 16, 225–233. [Google Scholar] [CrossRef] [PubMed]
  149. Madhavan, D.; Zucknick, M.; Wallwiener, M.; Cuk, K.; Modugno, C.; Scharpff, M.; Schott, S.; Heil, J.; Turchinovich, A.; Yang, R.; et al. Circulating MiRNAs as Surrogate Markers for Circulating Tumor Cells and Prognostic Markers in Metastatic Breast Cancer. Clin. Cancer Res. 2012, 18, 5972–5982. [Google Scholar] [CrossRef]
  150. Guo, H.; Zhang, N.; Huang, T.; Shen, N. MicroRNA-200c in Cancer Generation, Invasion, and Metastasis. Int. J. Mol. Sci. 2025, 26, 710. [Google Scholar] [CrossRef]
  151. Chen, H.; Li, Z.; Zhang, L.; Zhang, L.; Zhang, Y.; Wang, Y.; Xu, M.; Zhong, Q. MicroRNA-200c Inhibits the Metastasis of Triple-Negative Breast Cancer by Targeting ZEB2, an Epithelial-Mesenchymal Transition Regulator. Ann. Clin. Lab. Sci. 2020, 50, 519–527. [Google Scholar]
  152. Taha, M.; Mitwally, N.; Soliman, A.S.; Yousef, E. Potential Diagnostic and Prognostic Utility of MiR-141, MiR-181b1, and MiR-23b in Breast Cancer. Int. J. Mol. Sci. 2020, 21, 8589. [Google Scholar] [CrossRef]
  153. Li, Y.; Meng, X.; Luo, Y.; Luo, S.; Li, J.; Zeng, J.; Huang, X.; Wang, J. The Oncogenic MiR-429 Promotes Triple-Negative Breast Cancer Progression by Degrading DLC1. Aging 2023, 15, 9809–9821. [Google Scholar] [CrossRef]
  154. Ye, Z.B.; Ma, G.; Zhao, Y.H.; Xiao, Y.; Zhan, Y.; Jing, C.; Gao, K.; Liu, Z.H.; Yu, S.J. MiR-429 Inhibits Migration and Invasion of Breast Cancer Cells in Vitro. Int. J. Oncol. 2015, 46, 531–538. [Google Scholar] [CrossRef]
  155. Li, P.; Xu, T.; Zhou, X.; Liao, L.; Pang, G.; Luo, W.; Han, L.; Zhang, J.; Luo, X.; Xie, X.; et al. Downregulation of MiRNA-141 in Breast Cancer Cells Is Associated with Cell Migration and Invasion: Involvement of ANP32E Targeting. Cancer Med. 2017, 6, 662. [Google Scholar] [CrossRef]
  156. Abdelaal, A.M.; Sohal, I.S.; Iyer, S.; Sudarshan, K.; Kothandaraman, H.; Lanman, N.A.; Low, P.S.; Kasinski, A.L. A First-in-Class Fully Modified Version of MiR-34a with Outstanding Stability, Activity, and Anti-Tumor Efficacy. Oncogene 2023, 42, 2985–2999. [Google Scholar] [CrossRef] [PubMed]
  157. Chen, Y.; Li, Z.; Chen, X.; Zhang, S. Long Non-Coding RNAs: From Disease Code to Drug Role. Acta Pharm. Sin. B 2021, 11, 340–354. [Google Scholar] [CrossRef] [PubMed]
  158. Iyer, M.K.; Niknafs, Y.S.; Malik, R.; Singhal, U.; Sahu, A.; Hosono, Y.; Barrette, T.R.; Prensner, J.R.; Evans, J.R.; Zhao, S.; et al. The Landscape of Long Noncoding RNAs in the Human Transcriptome. Nat. Genet. 2015, 47, 199–208. [Google Scholar] [CrossRef]
  159. Quinn, J.J.; Chang, H.Y. Unique Features of Long Non-Coding RNA Biogenesis and Function. Nat. Rev. Genet. 2016, 17, 47–62. [Google Scholar] [CrossRef]
  160. Wu, D.; Zhu, J.; Fu, Y.; Li, C.; Wu, B. LncRNA HOTAIR Promotes Breast Cancer Progression through Regulating the MiR-129-5p/FZD7 Axis. Cancer Biomark. 2021, 30, 203–212. [Google Scholar] [CrossRef] [PubMed]
  161. Raju, G.S.R.; Pavitra, E.; Bandaru, S.S.; Varaprasad, G.L.; Nagaraju, G.P.; Malla, R.R.; Huh, Y.S.; Han, Y.-K. HOTAIR: A Potential Metastatic, Drug-Resistant and Prognostic Regulator of Breast Cancer. Mol. Cancer 2023, 22, 65. [Google Scholar] [CrossRef] [PubMed]
  162. Gupta, R.A.; Shah, N.; Wang, K.C.; Kim, J.; Horlings, H.M.; Wong, D.J.; Tsai, M.-C.; Hung, T.; Argani, P.; Rinn, J.L.; et al. Long Non-Coding RNA HOTAIR Reprograms Chromatin State to Promote Cancer Metastasis. Nature 2010, 464, 1071–1076. [Google Scholar] [CrossRef]
  163. Deng, S.; Wang, J.; Zhang, L.; Li, J.; Jin, Y. LncRNA HOTAIR Promotes Cancer Stem-Like Cells Properties by Sponging MiR-34a to Activate the JAK2/STAT3 Pathway in Pancreatic Ductal Adenocarcinoma. Onco Targets Ther. 2021, 14, 1883–1893. [Google Scholar] [CrossRef]
  164. Collina, F.; Aquino, G.; Brogna, M.; Cipolletta, S.; Buonfanti, G.; De Laurentiis, M.; Di Bonito, M.; Cantile, M.; Botti, G. LncRNA HOTAIR Up-Regulation Is Strongly Related with Lymph Nodes Metastasis and LAR Subtype of Triple Negative Breast Cancer. J. Cancer 2019, 10, 2018–2024. [Google Scholar] [CrossRef] [PubMed]
  165. Matouk, I.J.; DeGroot, N.; Mezan, S.; Ayesh, S.; Abu-lail, R.; Hochberg, A.; Galun, E. The H19 Non-Coding RNA Is Essential for Human Tumor Growth. PLoS ONE 2007, 2, e845. [Google Scholar] [CrossRef]
  166. Shin, V.Y.; Chen, J.; Cheuk, I.W.-Y.; Siu, M.-T.; Ho, C.-W.; Wang, X.; Jin, H.; Kwong, A. Long Non-Coding RNA NEAT1 Confers Oncogenic Role in Triple-Negative Breast Cancer through Modulating Chemoresistance and Cancer Stemness. Cell Death Dis. 2019, 10, 270. [Google Scholar] [CrossRef]
  167. Betts, J.A.; Moradi Marjaneh, M.; Al-Ejeh, F.; Lim, Y.C.; Shi, W.; Sivakumaran, H.; Tropée, R.; Patch, A.-M.; Clark, M.B.; Bartonicek, N.; et al. Long Noncoding RNAs CUPID1 and CUPID2 Mediate Breast Cancer Risk at 11q13 by Modulating the Response to DNA Damage. Am. J. Hum. Genet. 2017, 101, 255–266. [Google Scholar] [CrossRef]
  168. Kristensen, L.S.; Andersen, M.S.; Stagsted, L.V.W.; Ebbesen, K.K.; Hansen, T.B.; Kjems, J. The Biogenesis, Biology and Characterization of Circular RNAs. Nat. Rev. Genet. 2019, 20, 675–691. [Google Scholar] [CrossRef] [PubMed]
  169. Zhao, W.; Dong, M.; Pan, J.; Wang, Y.; Zhou, J.; Ma, J.; Liu, S. Circular RNAs: A Novel Target among Non-coding RNAs with Potential Roles in Malignant Tumors (Review). Mol. Med. Rep. 2019, 20, 3463–3474. [Google Scholar] [CrossRef] [PubMed]
  170. Zhang, M.; Bai, X.; Zeng, X.; Liu, J.; Liu, F.; Zhang, Z. CircRNA-MiRNA-MRNA in Breast Cancer. Clin. Chim. Acta 2021, 523, 120–130. [Google Scholar] [CrossRef]
  171. Song, X.; Liang, Y.; Sang, Y.; Li, Y.; Zhang, H.; Chen, B.; Du, L.; Liu, Y.; Wang, L.; Zhao, W.; et al. CircHMCU Promotes Proliferation and Metastasis of Breast Cancer by Sponging the Let-7 Family. Mol. Ther. Nucleic Acids 2020, 20, 518–533, Erratum in Mol. Ther. Nucleic Acids. 2021, 26, 1240, https://doi.org/10.1016/j.omtn.2021.11.001. [Google Scholar] [CrossRef]
  172. Du, W.W.; Yang, W.; Li, X.; Awan, F.M.; Yang, Z.; Fang, L.; Lyu, J.; Li, F.; Peng, C.; Krylov, S.N.; et al. A Circular RNA Circ-DNMT1 Enhances Breast Cancer Progression by Activating Autophagy. Oncogene 2018, 37, 5829–5842. [Google Scholar] [CrossRef] [PubMed]
  173. Meng, L.; Chang, S.; Sang, Y.; Ding, P.; Wang, L.; Nan, X.; Xu, R.; Liu, F.; Gu, L.; Zheng, Y.; et al. Circular RNA CircCCDC85A Inhibits Breast Cancer Progression via Acting as a MiR-550a-5p Sponge to Enhance MOB1A Expression. Breast Cancer Res. 2022, 24, 1. [Google Scholar] [CrossRef]
  174. Li, Y.; Li, H. Circular RNA VRK1 Correlates with Favourable Prognosis, Inhibits Cell Proliferation but Promotes Apoptosis in Breast Cancer. J. Clin. Lab. Anal. 2020, 34, e22980. [Google Scholar] [CrossRef]
  175. Yan, L.; Zheng, M.; Wang, H. Circular RNA Hsa_circ_0072309 Inhibits Proliferation and Invasion of Breast Cancer Cells via Targeting MiR-492. Cancer Manag. Res. 2019, 11, 1033–1041. [Google Scholar] [CrossRef]
  176. Rao, A.K.D.M.; Arvinden, V.R.; Ramasamy, D.; Patel, K.; Meenakumari, B.; Ramanathan, P.; Sundersingh, S.; Sridevi, V.; Rajkumar, T.; Herceg, Z.; et al. Identification of Novel Dysregulated Circular RNAs in Early-stage Breast Cancer. J. Cell. Mol. Med. 2021, 25, 3912–3921. [Google Scholar] [CrossRef]
  177. 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. 2019, 27, 1638–1652, Erratum in Mol. Ther. 2021, 29, 3525–3527. https://doi.org/10.1016/j.ymthe.2021.11.002. [Google Scholar] [CrossRef]
  178. Li, H.; Xu, W.; Xia, Z.; Liu, W.; Pan, G.; Ding, J.; Li, J.; Wang, J.; Xie, X.; Jiang, D. Hsa_circ_0000199 Facilitates Chemo-Tolerance of Triple-Negative Breast Cancer by Interfering with MiR-206/613-Led PI3K/Akt/MTOR Signaling. Aging 2021, 13, 4522–4551. [Google Scholar] [CrossRef]
  179. Sarvari, P.; Sarvari, P.; Ramírez-Díaz, I.; Mahjoubi, F.; Rubio, K. Advances of Epigenetic Biomarkers and Epigenome Editing for Early Diagnosis in Breast Cancer. Int. J. Mol. Sci. 2022, 23, 9521. [Google Scholar] [CrossRef]
  180. Brown, L.J.; Achinger-Kawecka, J.; Portman, N.; Clark, S.; Stirzaker, C.; Lim, E. Epigenetic Therapies and Biomarkers in Breast Cancer. Cancers 2022, 14, 474. [Google Scholar] [CrossRef]
  181. Biswas, S.; Rao, C.M. Epigenetics in Cancer: Fundamentals and Beyond. Pharmacol. Ther. 2017, 173, 118–134. [Google Scholar] [CrossRef] [PubMed]
  182. Zhuang, J.; Huo, Q.; Yang, F.; Xie, N. Perspectives on the Role of Histone Modification in Breast Cancer Progression and the Advanced Technological Tools to Study Epigenetic Determinants of Metastasis. Front. Genet. 2020, 11, 603552. [Google Scholar] [CrossRef] [PubMed]
  183. Sarkar, S.; Venkatesh, D.; Kandasamy, T.; Ghosh, S.S. Epigenetic Modulations in Breast Cancer: An Emerging Paradigm in Therapeutic Implications. Front. Biosci. 2024, 29, 287. [Google Scholar] [CrossRef]
  184. Okano, M.; Bell, D.W.; Haber, D.A.; Li, E. DNA Methyltransferases Dnmt3a and Dnmt3b Are Essential for De Novo Methylation and Mammalian Development. Cell 1999, 99, 247–257. [Google Scholar] [CrossRef]
  185. Ma, L.; Li, C.; Yin, H.; Huang, J.; Yu, S.; Zhao, J.; Tang, Y.; Yu, M.; Lin, J.; Ding, L.; et al. The Mechanism of DNA Methylation and MiRNA in Breast Cancer. Int. J. Mol. Sci. 2023, 24, 9360. [Google Scholar] [CrossRef]
  186. Feng, L.; Lou, J. DNA Methylation Analysis. Methods Mol. Biol. 2019, 1894, 181–227. [Google Scholar]
  187. Pastor, W.A.; Aravind, L.; Rao, A. TETonic Shift: Biological Roles of TET Proteins in DNA Demethylation and Transcription. Nat. Rev. Mol. Cell Biol. 2013, 14, 341–356. [Google Scholar] [CrossRef] [PubMed]
  188. Lakshminarasimhan, R.; Liang, G. The Role of DNA Methylation in Cancer. In Advances in Experimental Medicine and Biology; Springer: Cham, Switzerland, 2016; pp. 151–172. [Google Scholar]
  189. Vietri, M.; D’elia, G.; Benincasa, G.; Ferraro, G.; Caliendo, G.; Nicoletti, G.; Napoli, C. DNA Methylation and Breast Cancer: A Way Forward (Review). Int. J. Oncol. 2021, 59, 98. [Google Scholar] [CrossRef]
  190. Prajzendanc, K.; Domagała, P.; Hybiak, J.; Ryś, J.; Huzarski, T.; Szwiec, M.; Tomiczek-Szwiec, J.; Redelbach, W.; Sejda, A.; Gronwald, J.; et al. BRCA1 Promoter Methylation in Peripheral Blood Is Associated with the Risk of Triple-negative Breast Cancer. Int. J. Cancer 2020, 146, 1293–1298. [Google Scholar] [CrossRef] [PubMed]
  191. Kang, Z.; Wang, J.; Liu, J.; Du, L.; Liu, X. Epigenetic Modifications in Breast Cancer: From Immune Escape Mechanisms to Therapeutic Target Discovery. Front. Immunol. 2025, 16, 1584087, Erratum in Front. Immunol. 2025, 16, 1643911. https://doi.org/10.3389/fimmu.2025.1643911 . [Google Scholar] [CrossRef]
  192. Grosselin, K.; Durand, A.; Marsolier, J.; Poitou, A.; Marangoni, E.; Nemati, F.; Dahmani, A.; Lameiras, S.; Reyal, F.; Frenoy, O.; et al. High-Throughput Single-Cell ChIP-Seq Identifies Heterogeneity of Chromatin States in Breast Cancer. Nat. Genet. 2019, 51, 1060–1066. [Google Scholar] [CrossRef]
  193. Holm, K.; Grabau, D.; Lövgren, K.; Aradottir, S.; Gruvberger-Saal, S.; Howlin, J.; Saal, L.H.; Ethier, S.P.; Bendahl, P.-O.; Stål, O.; et al. Global H3K27 Trimethylation and EZH2 Abundance in Breast Tumor Subtypes. Mol. Oncol. 2012, 6, 494–506. [Google Scholar] [CrossRef]
  194. Healey, M.A.; Hu, R.; Beck, A.H.; Collins, L.C.; Schnitt, S.J.; Tamimi, R.M.; Hazra, A. Association of H3K9me3 and H3K27me3 Repressive Histone Marks with Breast Cancer Subtypes in the Nurses’ Health Study. Breast Cancer Res. Treat. 2014, 147, 639–651. [Google Scholar] [CrossRef] [PubMed]
  195. Elsheikh, S.E.; Green, A.R.; Rakha, E.A.; Powe, D.G.; Ahmed, R.A.; Collins, H.M.; Soria, D.; Garibaldi, J.M.; Paish, C.E.; Ammar, A.A.; et al. Global Histone Modifications in Breast Cancer Correlate with Tumor Phenotypes, Prognostic Factors, and Patient Outcome. Cancer Res. 2009, 69, 3802–3809. [Google Scholar] [CrossRef]
  196. Berger, L.; Kolben, T.; Meister, S.; Kolben, T.M.; Schmoeckel, E.; Mayr, D.; Mahner, S.; Jeschke, U.; Ditsch, N.; Beyer, S. Expression of H3K4me3 and H3K9ac in Breast Cancer. J. Cancer Res. Clin. Oncol. 2020, 146, 2017–2027. [Google Scholar] [CrossRef]
  197. Idrissou, M.; Boisnier, T.; Sanches, A.; Khoufaf, F.Z.H.; Penault-Llorca, F.; Bignon, Y.-J.; Bernard-Gallon, D. TIP60/P400/H4K12ac Plays a Role as a Heterochromatin Back-up Skeleton in Breast Cancer. Cancer Genom.-Proteom. 2020, 17, 687–694. [Google Scholar] [CrossRef]
  198. Hałasa, M.; Wawruszak, A.; Przybyszewska, A.; Jaruga, A.; Guz, M.; Kałafut, J.; Stepulak, A.; Cybulski, M. H3K18Ac as a Marker of Cancer Progression and Potential Target of Anti-Cancer Therapy. Cells 2019, 8, 485. [Google Scholar] [CrossRef] [PubMed]
  199. Denkert, C.; von Minckwitz, G.; Darb-Esfahani, S.; Lederer, B.; Heppner, B.I.; Weber, K.E.; Budczies, J.; Huober, J.; Klauschen, F.; Furlanetto, J.; et al. Tumour-Infiltrating Lymphocytes and Prognosis in Different Subtypes of Breast Cancer: A Pooled Analysis of 3771 Patients Treated with Neoadjuvant Therapy. Lancet Oncol. 2018, 19, 40–50. [Google Scholar] [CrossRef]
  200. Gao, Z.H.; Li, C.X.; Liu, M.; Jiang, J.Y. Predictive and Prognostic Role of Tumour-Infiltrating Lymphocytes in Breast Cancer Patients with Different Molecular Subtypes: A Meta-Analysis. BMC Cancer 2020, 20, 1150. [Google Scholar] [CrossRef]
  201. Simon, S.; Labarriere, N. PD-1 Expression on Tumor-Specific T Cells: Friend or Foe for Immunotherapy? Oncoimmunology 2017, 7, e1364828. [Google Scholar] [CrossRef]
  202. Vranic, S.; Cyprian, F.S.; Gatalica, Z.; Palazzo, J. PD-L1 Status in Breast Cancer: Current View and Perspectives. Semin. Cancer Biol. 2021, 72, 146–154. [Google Scholar] [CrossRef] [PubMed]
  203. Debien, V.; De Caluwé, A.; Wang, X.; Piccart-Gebhart, M.; Tuohy, V.K.; Romano, E.; Buisseret, L. Immunotherapy in Breast Cancer: An Overview of Current Strategies and Perspectives. NPJ Breast Cancer 2023, 9, 7. [Google Scholar] [CrossRef]
  204. He, Y.; Jiang, Z.; Chen, C.; Wang, X. Classification of Triple-Negative Breast Cancers Based on Immunogenomic Profiling. J. Exp. Clin. Cancer Res. 2018, 37, 327. [Google Scholar] [CrossRef] [PubMed]
  205. Nagarajan, D.; McArdle, S.E.B. Immune Landscape of Breast Cancers. Biomedicines 2018, 6, 20. [Google Scholar] [CrossRef]
  206. Erber, R.; Hartmann, A. Understanding PD-L1 Testing in Breast Cancer: A Practical Approach. Breast Care 2020, 15, 481–490. [Google Scholar] [CrossRef] [PubMed]
  207. Marletta, S.; Fusco, N.; Munari, E.; Luchini, C.; Cimadamore, A.; Brunelli, M.; Querzoli, G.; Martini, M.; Vigliar, E.; Colombari, R.; et al. Atlas of PD-L1 for Pathologists: Indications, Scores, Diagnostic Platforms and Reporting Systems. J. Pers. Med. 2022, 12, 1073. [Google Scholar] [CrossRef]
  208. El Bairi, K.; Haynes, H.R.; Blackley, E.; Fineberg, S.; Shear, J.; Turner, S.; de Freitas, J.R.; Sur, D.; Amendola, L.C.; Gharib, M.; et al. The Tale of TILs in Breast Cancer: A Report from The International Immuno-Oncology Biomarker Working Group. NPJ Breast Cancer 2021, 7, 150. [Google Scholar] [CrossRef]
  209. Lam, B.M.; Verrill, C. Clinical Significance of Tumour-Infiltrating B Lymphocytes (TIL-Bs) in Breast Cancer: A Systematic Literature Review. Cancers 2023, 15, 1164. [Google Scholar] [CrossRef] [PubMed]
  210. Salgado, R.; Denkert, C.; Demaria, S.; Sirtaine, N.; Klauschen, F.; Pruneri, G.; Wienert, S.; Van den Eynden, G.; Baehner, F.L.; Penault-Llorca, F.; et al. The Evaluation of Tumor-Infiltrating Lymphocytes (TILs) in Breast Cancer: Recommendations by an International TILs Working Group 2014. Ann. Oncol. 2015, 26, 259–271. [Google Scholar] [CrossRef] [PubMed]
  211. Stanton, S.E.; Adams, S.; Disis, M.L. Variation in the Incidence and Magnitude of Tumor-Infiltrating Lymphocytes in Breast Cancer Subtypes: A Systematic Review. JAMA Oncol. 2016, 2, 1354–1360. [Google Scholar] [CrossRef]
  212. Bianchini, G.; De Angelis, C.; Licata, L.; Gianni, L. Treatment Landscape of Triple-Negative Breast Cancer—Expanded Options, Evolving Needs. Nat. Rev. Clin. Oncol. 2022, 19, 91–113. [Google Scholar] [CrossRef]
  213. Nanda, R.; Chow, L.Q.M.; Dees, E.C.; Berger, R.; Gupta, S.; Geva, R.; Pusztai, L.; Pathiraja, K.; Aktan, G.; Cheng, J.D.; et al. Pembrolizumab in Patients With Advanced Triple-Negative Breast Cancer: Phase Ib KEYNOTE-012 Study. J. Clin. Oncol. 2016, 34, 2460–2467. [Google Scholar] [CrossRef]
  214. Bortolini Silveira, A.; Bidard, F.C.; Tanguy, M.L.; Girard, E.; Trédan, O.; Dubot, C.; Jacot, W.; Goncalves, A.; Debled, M.; Levy, C.; et al. Multimodal Liquid Biopsy for Early Monitoring and Outcome Prediction of Chemotherapy in Metastatic Breast Cancer. NPJ Breast Cancer 2021, 7, 115. [Google Scholar] [CrossRef]
  215. Pantel, K.; Alix-Panabières, C. Liquid Biopsy and Minimal Residual Disease—Latest Advances and Implications for Cure. Nat. Rev. Clin. Oncol. 2019, 16, 409–424. [Google Scholar] [CrossRef]
  216. Serrano, M.J.; Garrido-Navas, M.C.; Mochon, J.J.D.; Cristofanilli, M.; Gil-Bazo, I.; Pauwels, P.; Malapelle, U.; Russo, A.; Lorente, J.A.; Ruiz-Rodriguez, A.J.; et al. Precision Prevention and Cancer Interception: The New Challenges of Liquid Biopsy. Cancer Discov. 2020, 10, 1635–1644. [Google Scholar] [CrossRef] [PubMed]
  217. Kilgour, E.; Rothwell, D.G.; Brady, G.; Dive, C. Liquid Biopsy-Based Biomarkers of Treatment Response and Resistance. Cancer Cell 2020, 37, 485–495. [Google Scholar] [CrossRef]
  218. Sala, M.; Ros, M.; Saltel, F. A Complex and Evolutive Character: Two Face Aspects of ECM in Tumor Progression. Front. Oncol. 2020, 10, 1620. [Google Scholar] [CrossRef]
  219. Qiu, P.; Yu, X.; Zheng, F.; Gu, X.; Huang, Q.Q.; Qin, K.; Hu, Y.; Liu, B.; Xu, T.; Zhang, T.; et al. Advancements in Liquid Biopsy for Breast Cancer: Molecular Biomarkers and Clinical Applications. Cancer Treat. Rev. 2025, 139, 102979. [Google Scholar] [CrossRef]
  220. Lin, Z.; Neiswender, J.; Fang, B.; Ma, X.; Zhang, J.; Hu, X. Value of Circulating Cell-Free DNA Analysis as a Diagnostic Tool for Breast Cancer: A Meta-Analysis. Oncotarget 2017, 8, 26625–26636. [Google Scholar] [CrossRef] [PubMed]
  221. Huang, Z.H.; Li, L.H.; Hua, D. Quantitative Analysis of Plasma Circulating DNA at Diagnosis and during Follow-up of Breast Cancer Patients. Cancer Lett. 2006, 243, 64–70. [Google Scholar] [CrossRef]
  222. Hench, I.B.; Hench, J.; Tolnay, M. Liquid Biopsy in Clinical Management of Breast, Lung, and Colorectal Cancer. Front. Med. 2018, 5, 9. [Google Scholar] [CrossRef]
  223. Li, Y.; Wu, S.; Bai, F. Molecular Characterization of Circulating Tumor Cells—From Bench to Bedside. Semin. Cell Dev. Biol. 2018, 75, 88–97. [Google Scholar] [CrossRef] [PubMed]
  224. Kowalik, A.; Kowalewska, M.; Góźdź, S. Current Approaches for Avoiding the Limitations of Circulating Tumor Cells Detection Methods—Implications for Diagnosis and Treatment of Patients with Solid Tumors. Transl. Res. 2017, 185, 58–84.e15. [Google Scholar] [CrossRef] [PubMed]
  225. Papadaki, M.A.; Stoupis, G.; Theodoropoulos, P.A.; Mavroudis, D.; Georgoulias, V.; Agelaki, S. Circulating Tumor Cells with Stemness and Epithelial-to-Mesenchymal Transition Features Are Chemoresistant and Predictive of Poor Outcome in Metastatic Breast Cancer. Mol. Cancer Ther. 2019, 18, 437–447. [Google Scholar] [CrossRef] [PubMed]
  226. Zhang, L.; Riethdorf, S.; Wu, G.; Wang, T.; Yang, K.; Peng, G.; Liu, J.; Pantel, K. Meta-Analysis of the Prognostic Value of Circulating Tumor Cells in Breast Cancer. Clin. Cancer Res. 2012, 18, 5701–5710. [Google Scholar] [CrossRef]
  227. Janni, W.J.; Rack, B.; Terstappen, L.W.M.M.; Pierga, J.Y.; Taran, F.A.; Fehm, T.; Hall, C.; De Groot, M.R.; Bidard, F.C.; Friedl, T.W.P.; et al. Pooled Analysis of the Prognostic Relevance of Circulating Tumor Cells in Primary Breast Cancer. Clin. Cancer Res. 2016, 22, 2583–2593. [Google Scholar] [CrossRef]
  228. Budd, G.T.; Cristofanilli, M.; Ellis, M.J.; Stopeck, A.; Borden, E.; Miller, M.C.; Matera, J.; Repollet, M.; Doyle, G.V.; Terstappen, L.W.M.M.; et al. Circulating Tumor Cells versus Imaging—Predicting Overall Survival in Metastatic Breast Cancer. Clin. Cancer Res. 2006, 12, 6403–6409. [Google Scholar] [CrossRef]
  229. Miricescu, D.; Totan, A.; Stanescu-Spinu, I.-I.; Badoiu, S.C.; Stefani, C.; Greabu, M. PI3K/AKT/MTOR Signaling Pathway in Breast Cancer: From Molecular Landscape to Clinical Aspects. Int. J. Mol. Sci. 2020, 22, 173. [Google Scholar] [CrossRef]
  230. Braglia, L.; Zavatti, M.; Vinceti, M.; Martelli, A.M.; Marmiroli, S. Deregulated PTEN/PI3K/AKT/MTOR Signaling in Prostate Cancer: Still a Potential Druggable Target? Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2020, 1867, 118731. [Google Scholar] [CrossRef] [PubMed]
  231. Garg, P.; Ramisetty, S.; Nair, M.; Kulkarni, P.; Horne, D.; Salgia, R.; Singhal, S.S. Strategic Advancements in Targeting the PI3K/AKT/MTOR Pathway for Breast Cancer Therapy. Biochem. Pharmacol. 2025, 236, 116850. [Google Scholar] [CrossRef] [PubMed]
  232. Singh, S.; Barik, D.; Lawrie, K.; Mohapatra, I.; Prasad, S.; Naqvi, A.R.; Singh, A.; Singh, G. Unveiling Novel Avenues in MTOR-Targeted Therapeutics: Advancements in Glioblastoma Treatment. Int. J. Mol. Sci. 2023, 24, 14960. [Google Scholar] [CrossRef]
  233. Browne, I.M.; Okines, A.F.C. Resistance to Targeted Inhibitors of the PI3K/AKT/MTOR Pathway in Advanced Oestrogen-Receptor-Positive Breast Cancer. Cancers 2024, 16, 2259. [Google Scholar] [CrossRef] [PubMed]
  234. Truong, T.H.; Roman Ortiz, N.I.; Ufondu, C.A.; Lee, S.-J.; Ostrander, J.H. Emerging Mechanisms of Therapy Resistance in Metastatic ER+ Breast Cancer. Endocrinology 2025, 166, bqaf127. [Google Scholar] [CrossRef]
  235. Li, X.; Pu, W.; Zheng, Q.; Ai, M.; Chen, S.; Peng, Y. Proteolysis-Targeting Chimeras (PROTACs) in Cancer Therapy. Mol. Cancer 2022, 21, 99. [Google Scholar] [CrossRef]
  236. Djafari, J.; Fernández-Lodeiro, J.; Santos, H.M.; Lorenzo, J.; Rodriguez-Calado, S.; Bértolo, E.; Capelo-Martínez, J.L.; Lodeiro, C. Study and Preparation of Multifunctional Poly(L-Lysine)@Hyaluronic Acid Nanopolyplexes for the Effective Delivery of Tumor Suppressive MiR-34a into Triple-Negative Breast Cancer Cells. Materials 2020, 13, 5309. [Google Scholar] [CrossRef]
  237. Alsarraf, Z.; Nori, A.; Oraibi, A.; Al-Hussaniy, H.; Jabbar, A. BIBR1591 induces apoptosis in breast cancer cell line and increases expression of DAPK1, AND NR4A3. Cancer 2024, 9, 156–160. [Google Scholar]
  238. Nagahashi, M.; Komatsu, M.; Urano, S.; Kuroiwa, M.; Takahashi, Y.; Morimoto, K.; Pradipta, A.R.; Tanaka, K.; Miyoshi, Y. An Acrolein-Based Drug Delivery System Enables Tumor-Specific Sphingosine-1-Phosphate Targeting in Breast Cancer without Lymphocytopenia. Cancer Res. Commun. 2025, 5, 981–993. [Google Scholar] [CrossRef]
  239. Liu, B.; Zhou, H.; Tan, L.; Siu, K.T.H.; Guan, X.-Y. Exploring Treatment Options in Cancer: Tumor Treatment Strategies. Signal Transduct. Target. Ther. 2024, 9, 175. [Google Scholar] [CrossRef]
  240. Chavez-MacGregor, M.; Miao, J.; Pusztai, L.; Goetz, M.P.; Rastogi, P.; Ganz, P.A.; Mamounas, E.P.; Paik, S.; Bandos, H.; Razaq, W.; et al. Phase III Randomized, Placebo-Controlled Trial of Endocrine Therapy ± 1 Year of Everolimus in Patients With High-Risk, Hormone Receptor–Positive, Early-Stage Breast Cancer. J. Clin. Oncol. 2024, 42, 3012–3021. [Google Scholar] [CrossRef] [PubMed]
  241. Mohamed, A.H.; Obeid, R.A.; Fadhil, A.A.; Amir, A.A.; Adhab, Z.H.; Jabouri, E.A.; Ahmad, I.; Alshahrani, M.Y. BTLA and HVEM: Emerging Players in the Tumor Microenvironment and Cancer Progression. Cytokine 2023, 172, 156412. [Google Scholar] [CrossRef] [PubMed]
  242. Cortés, J.; Kim, S.-B.; Chung, W.-P.; Im, S.-A.; Park, Y.H.; Hegg, R.; Kim, M.H.; Tseng, L.-M.; Petry, V.; Chung, C.-F.; et al. Trastuzumab Deruxtecan versus Trastuzumab Emtansine for Breast Cancer. N. Engl. J. Med. 2022, 386, 1143–1154. [Google Scholar] [CrossRef]
  243. Zabeti Touchaei, A.; Vahidi, S. MicroRNAs as Regulators of Immune Checkpoints in Cancer Immunotherapy: Targeting PD-1/PD-L1 and CTLA-4 Pathways. Cancer Cell Int. 2024, 24, 102. [Google Scholar] [CrossRef]
  244. Singh, S.K.; Spiegel, S. Sphingosine-1-Phosphate Signaling: A Novel Target for Simultaneous Adjuvant Treatment of Triple Negative Breast Cancer and Chemotherapy-Induced Neuropathic Pain. Adv. Biol. Regul. 2020, 75, 100670. [Google Scholar] [CrossRef]
  245. Tülüce, Y.; Köstekci, S.; Karakuş, F.; Keleş, A.Y.; Tunçyürekli, M. Investigation the Immunotherapeutic Potential of MiR-4477a Targeting PD-1/PD-L1 in Breast Cancer Cell Line Using a CD8+ Co-Culture Model. Mol. Biol. Rep. 2025, 52, 326. [Google Scholar] [CrossRef] [PubMed]
  246. Tao, X.; Na, L.; Hu, E.-X.; Wang, J.; Wu, L.-G.; Zhang, X.; Wang, L.-B. Clinical Diagnostic Value of Circ-ARHGER28 for Breast Cancer and Its Effect on MCF 7 Cell Proliferation and Apoptosis. Anticancer Res. 2024, 44, 2877–2886. [Google Scholar] [CrossRef]
  247. Samuels, M.; Besta, S.; Betrán, A.L.; Nia, R.S.; Xie, X.; Gu, X.; Shu, Q.; Giamas, G. CRISPR Screening Approaches in Breast Cancer Research. Cancer Metastasis Rev. 2025, 44, 59. [Google Scholar] [CrossRef]
  248. Ritika; Rani, S.; Malviya, R.; Rajput, S.; Belagodu Sridhar, S.; Kaushik, D. Understanding the Prospective of Gene Therapy for the Treatment of Breast Cancer. Rev. Senol. Patol. Mamar. 2025, 38, 100683. [Google Scholar] [CrossRef]
  249. Unidirwade, D.S.; Lade, S.N.; Umekar, M.J.; Burle, S.S.; Rangari, S.W. Innovative Theranostic Potential of Graphene Quantum Dot Nanocomposites in Breast Cancer. Med. Oncol. 2025, 42, 404. [Google Scholar] [CrossRef]
  250. Gómez, I.J.; Ovejero-Paredes, K.; Méndez-Arriaga, J.M.; Pizúrová, N.; Filice, M.; Zajíčková, L.; Prashar, S.; Gómez-Ruiz, S. Organotin(IV)-Decorated Graphene Quantum Dots as Dual Platform for Molecular Imaging and Treatment of Triple Negative Breast Cancer. Chem.-A Eur. J. 2023, 29, e202301845. [Google Scholar] [CrossRef]
  251. Esgandari, K.; Mohammadian, M.; Zohdiaghdam, R.; Rastin, S.J.; Alidadi, S.; Behrouzkia, Z. Combined Treatment with Silver Graphene Quantum Dot, Radiation, and 17-AAG Induces Anticancer Effects in Breast Cancer Cells. J. Cell. Physiol. 2021, 236, 2817–2828. [Google Scholar] [CrossRef] [PubMed]
  252. Mohajeri, S.; Yaghoubi, H.; Bourang, S.; Noruzpour, M. Multifunctional Magnetic Nanocapsules for Dual Delivery of SiRNA and Chemotherapy to MCF-7 Cells (Breast Cancer Cells). Naunyn-Schmiedeberg′s Arch. Pharmacol. 2025, 398, 17957–17979. [Google Scholar] [CrossRef] [PubMed]
  253. Rugo, H.S.; Lacouture, M.E.; Goncalves, M.D.; Masharani, U.; Aapro, M.S.; O’Shaughnessy, J.A. A Multidisciplinary Approach to Optimizing Care of Patients Treated with Alpelisib. Breast 2022, 61, 156–167. [Google Scholar] [CrossRef] [PubMed]
  254. Darapu, H.; McKnight, M. Capivasertib-Induced Refractory Hyperglycemia in a Nondiabetic Patient with Metastatic Breast Cancer. AACE Endocrinol. Diabetes 2025, in press. [Google Scholar] [CrossRef]
  255. Kolinsky, M.P.; Rescigno, P.; Bianchini, D.; Zafeiriou, Z.; Mehra, N.; Mateo, J.; Michalarea, V.; Riisnaes, R.; Crespo, M.; Figueiredo, I.; et al. A Phase I Dose-Escalation Study of Enzalutamide in Combination with the AKT Inhibitor AZD5363 (Capivasertib) in Patients with Metastatic Castration-Resistant Prostate Cancer. Ann. Oncol. 2020, 31, 619–625. [Google Scholar] [CrossRef]
  256. Kushwaha, S.; Xu, X. Target of Rapamycin (TOR)–Based Therapy for Cardiomyopathy: Evidence From Zebrafish and Human Studies. Trends Cardiovasc. Med. 2012, 22, 39–43. [Google Scholar] [CrossRef]
  257. Unnikrishnan, A.; Deepa, S.S.; Herd, H.R.; Richardson, A. Extension of Life Span in Laboratory Mice. In Conn’s Handbook of Models for Human Aging; Elsevier: Amsterdam, The Netherlands, 2018; pp. 245–270. [Google Scholar]
  258. Mao, B.; Zhang, Q.; Ma, L.; Zhao, D.-S.; Zhao, P.; Yan, P. Overview of Research into MTOR Inhibitors. Molecules 2022, 27, 5295. [Google Scholar] [CrossRef]
  259. Wullschleger, S.; Loewith, R.; Hall, M.N. TOR Signaling in Growth and Metabolism. Cell 2006, 124, 471–484. [Google Scholar] [CrossRef]
  260. Naing, A.; Aghajanian, C.; Raymond, E.; Olmos, D.; Schwartz, G.; Oelmann, E.; Grinsted, L.; Burke, W.; Taylor, R.; Kaye, S.; et al. Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of AZD8055 in Advanced Solid Tumours and Lymphoma. Br. J. Cancer 2012, 107, 1093–1099. [Google Scholar] [CrossRef]
  261. del Campo, J.M.; Birrer, M.; Davis, C.; Fujiwara, K.; Gollerkeri, A.; Gore, M.; Houk, B.; Lau, S.; Poveda, A.; González-Martín, A.; et al. A Randomized Phase II Non-Comparative Study of PF-04691502 and Gedatolisib (PF-05212384) in Patients with Recurrent Endometrial Cancer. Gynecol. Oncol. 2016, 142, 62–69. [Google Scholar] [CrossRef]
  262. Wylaź, M.; Kaczmarska, A.; Pajor, D.; Hryniewicki, M.; Gil, D.; Dulińska-Litewka, J. Exploring the Role of PI3K/AKT/MTOR Inhibitors in Hormone-Related Cancers: A Focus on Breast and Prostate Cancer. Biomed. Pharmacother. 2023, 168, 115676. [Google Scholar] [CrossRef] [PubMed]
  263. Liu, C.; Xing, W.; Yu, H.; Zhang, W.; Si, T. ABCB1 and ABCG2 Restricts the Efficacy of Gedatolisib (PF-05212384), a PI3K Inhibitor in Colorectal Cancer Cells. Cancer Cell Int. 2021, 21, 108. [Google Scholar] [CrossRef]
  264. Low, L.E.; Kong, C.K.; Yap, W.-H.; Siva, S.P.; Gan, S.H.; Siew, W.S.; Ming, L.C.; Lai-Foenander, A.S.; Chang, S.K.; Lee, W.-L.; et al. Hydroxychloroquine: Key Therapeutic Advances and Emerging Nanotechnological Landscape for Cancer Mitigation. Chem. Biol. Interact. 2023, 386, 110750. [Google Scholar] [CrossRef] [PubMed]
  265. Rahim, R.; Strobl, J.S. Hydroxychloroquine, Chloroquine, and All-Trans Retinoic Acid Regulate Growth, Survival, and Histone Acetylation in Breast Cancer Cells. Anticancer Drugs 2009, 20, 736–745. [Google Scholar] [CrossRef]
  266. Falchook, G.; Infante, J.; Arkenau, H.-T.; Patel, M.R.; Dean, E.; Borazanci, E.; Brenner, A.; Cook, N.; Lopez, J.; Pant, S.; et al. First-in-Human Study of the Safety, Pharmacokinetics, and Pharmacodynamics of First-in-Class Fatty Acid Synthase Inhibitor TVB-2640 Alone and with a Taxane in Advanced Tumors. EClinicalMedicine 2021, 34, 100797. [Google Scholar] [CrossRef] [PubMed]
  267. Kelly, W.; Diaz Duque, A.E.; Michalek, J.; Konkel, B.; Caflisch, L.; Chen, Y.; Pathuri, S.C.; Madhusudanannair-Kunnuparampil, V.; Floyd, J.; Brenner, A. Phase II Investigation of TVB-2640 (Denifanstat) with Bevacizumab in Patients with First Relapse High-Grade Astrocytoma. Clin. Cancer Res. 2023, 29, 2419–2425. [Google Scholar] [CrossRef]
  268. Loomba, R.; Bedossa, P.; Grimmer, K.; Kemble, G.; Bruno Martins, E.; McCulloch, W.; O’Farrell, M.; Tsai, W.-W.; Cobiella, J.; Lawitz, E.; et al. Denifanstat for the Treatment of Metabolic Dysfunction-Associated Steatohepatitis: A Multicentre, Double-Blind, Randomised, Placebo-Controlled, Phase 2b Trial. Lancet Gastroenterol. Hepatol. 2024, 9, 1090–1100. [Google Scholar] [CrossRef]
  269. Serhan, H.A.; Bao, L.; Cheng, X.; Qin, Z.; Liu, C.-J.; Heth, J.A.; Udager, A.M.; Soellner, M.B.; Merajver, S.D.; Morikawa, A.; et al. Targeting Fatty Acid Synthase in Preclinical Models of TNBC Brain Metastases Synergizes with SN-38 and Impairs Invasion. NPJ Breast Cancer 2024, 10, 43. [Google Scholar] [CrossRef] [PubMed]
  270. Campone, M.; De Laurentiis, M.; Jhaveri, K.; Hu, X.; Ladoire, S.; Patsouris, A.; Zamagni, C.; Cui, J.; Cazzaniga, M.; Cil, T.; et al. Vepdegestrant, a PROTAC Estrogen Receptor Degrader, in Advanced Breast Cancer. N. Engl. J. Med. 2025, 393, 556–568. [Google Scholar] [CrossRef] [PubMed]
  271. Gough, S.M.; Flanagan, J.J.; Teh, J.; Andreoli, M.; Rousseau, E.; Pannone, M.; Bookbinder, M.; Willard, R.; Davenport, K.; Bortolon, E.; et al. Oral Estrogen Receptor PROTAC Vepdegestrant (ARV-471) Is Highly Efficacious as Monotherapy and in Combination with CDK4/6 or PI3K/MTOR Pathway Inhibitors in Preclinical ER+ Breast Cancer Models. Clin. Cancer Res. 2024, 30, 3549–3563. [Google Scholar] [CrossRef]
  272. Robertson, J.F.R.; Shao, Z.; Noguchi, S.; Bondarenko, I.; Panasci, L.; Singh, S.; Subramaniam, S.; Ellis, M.J. Fulvestrant Versus Anastrozole in Endocrine Therapy–Naïve Women With Hormone Receptor–Positive Advanced Breast Cancer: Final Overall Survival in the Phase III FALCON Trial. J. Clin. Oncol. 2025, 43, 1539–1545, Erratum in J. Clin. Oncol. 2025, 43, 1045 https://doi.org/10.1200/JCO-25-00116.. [Google Scholar] [CrossRef] [PubMed]
  273. Carlson, R.W. The History and Mechanism of Action of Fulvestrant. Clin. Breast Cancer 2005, 6, S5–S8. [Google Scholar] [CrossRef]
  274. Jhaveri, K.L.; Neven, P.; Casalnuovo, M.L.; Kim, S.-B.; Tokunaga, E.; Aftimos, P.; Saura, C.; O’Shaughnessy, J.; Harbeck, N.; Carey, L.A.; et al. Imlunestrant with or without Abemaciclib in Advanced Breast Cancer. N. Engl. J. Med. 2025, 392, 1189–1202. [Google Scholar] [CrossRef]
  275. Al-Karmalawy, A.A.; Mousa, M.H.A.; Sharaky, M.; Mourad, M.A.E.; El-Dessouki, A.M.; Hamouda, A.O.; Alnajjar, R.; Ayed, A.A.; Shaldam, M.A.; Tawfik, H.O. Lead Optimization of BIBR1591 To Improve Its Telomerase Inhibitory Activity: Design and Synthesis of Novel Four Chemical Series with In Silico, In Vitro, and In Vivo Preclinical Assessments. J. Med. Chem. 2024, 67, 492–512. [Google Scholar] [CrossRef]
  276. Aboushanab, A.R.; El-Moslemany, R.M.; El-Kamel, A.H.; Mehanna, R.A.; Bakr, B.A.; Ashour, A.A. Targeted Fisetin-Encapsulated β-Cyclodextrin Nanosponges for Breast Cancer. Pharmaceutics 2023, 15, 1480. [Google Scholar] [CrossRef]
  277. Hirata, N.; Yamada, S.; Yanagida, S.; Ono, A.; Kanda, Y. FTY720 Inhibits Expansion of Breast Cancer Stem Cells via PP2A Activation. Int. J. Mol. Sci. 2021, 22, 7259. [Google Scholar] [CrossRef] [PubMed]
  278. Chung, W.-P.; Huang, W.-L.; Liao, W.-A.; Hung, C.-H.; Chiang, C.-W.; Cheung, C.H.A.; Su, W.-C. FTY720 in Resistant Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer. Sci. Rep. 2022, 12, 241. [Google Scholar] [CrossRef]
  279. Takasaki, T.; Hagihara, K.; Satoh, R.; Sugiura, R. More than Just an Immunosuppressant: The Emerging Role of FTY720 as a Novel Inducer of ROS and Apoptosis. Oxid. Med. Cell. Longev. 2018, 2018, 4397159. [Google Scholar] [CrossRef]
  280. Canadian Agency for Drugs and Technologies in Health Pembrolizumab (Keytruda). CADTH Reimbursement Review: Therapeutic Area: Triple-Negative Breast Cancer; Canadian Agency for Drugs and Technologies in Health: Ottawa, ON, Canada, 2023. [Google Scholar]
  281. He, Y.; Ramesh, A.; Gusev, Y.; Bhuvaneshwar, K.; Giaccone, G. Molecular Predictors of Response to Pembrolizumab in Thymic Carcinoma. Cell Rep. Med. 2021, 2, 100392. [Google Scholar] [CrossRef] [PubMed]
  282. Aleem, A.; Shah, H. Atezolizumab. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  283. Hamidi, H.; Senbabaoglu, Y.; Beig, N.; Roels, J.; Manuel, C.; Guan, X.; Koeppen, H.; Assaf, Z.J.; Nabet, B.Y.; Waddell, A.; et al. Molecular Heterogeneity in Urothelial Carcinoma and Determinants of Clinical Benefit to PD-L1 Blockade. Cancer Cell 2024, 42, 2098–2112.e4. [Google Scholar] [CrossRef]
  284. Sledge, G.; Xiu, J.; Mahtani, R.L.; Sandoval Leon, A.C.; Oberley, M.J.; Radovich, M.; Spetzler, D. Abstract PS13-09: Mechanisms of Resistance to Trastuzumab Deruxtecan in Breast Cancer Elucidated by Multi-Omic Molecular Profiling. Clin. Cancer Res. 2025, 31, PS13-09-PS13-09. [Google Scholar] [CrossRef]
  285. Tolaney, S.M.; Jiang, Z.; Zhang, Q.; Barroso-Sousa, R.; Park, Y.H.; Rimawi, M.F.; Saura, C.; Schneeweiss, A.; Toi, M.; Chae, Y.S.; et al. Trastuzumab Deruxtecan plus Pertuzumab for HER2-Positive Metastatic Breast Cancer. N. Engl. J. Med. 2025. [Google Scholar] [CrossRef]
  286. Han, T.-Y.; Hou, L.-S.; Li, J.-X.; Huan, M.-L.; Zhou, S.-Y.; Zhang, B.-L. Bone Targeted MiRNA Delivery System for MiR-34a with Enhanced Anti-Tumor Efficacy to Bone-Associated Metastatic Breast Cancer. Int. J. Pharm. 2023, 635, 122755. [Google Scholar] [CrossRef] [PubMed]
  287. Lei, R.; Long, Y.; Li, Q.; Xie, Q.; Ling, X.; Xie, M.; Zhou, H.; Zhang, B. Circular RNA Circ_ARHGEF28 Inhibits MST1/2 Dimerization to Suppress Hippo Pathway to Induce Cisplatin Resistance in Ovarian Cancer. Cancer Cell Int. 2024, 24, 256. [Google Scholar] [CrossRef]
  288. Wang, Z.; Li, Y.; Xiao, Y.; Lin, H.; Yang, P.; Humphries, B.; Gao, T.; Yang, C. Integrin A9 Depletion Promotes B-catenin Degradation to Suppress Triple-negative Breast Cancer Tumor Growth and Metastasis. Int. J. Cancer 2019, 145, 2767–2780. [Google Scholar] [CrossRef]
  289. Misra, G.; Qaisar, S.; Singh, P. CRISPR-Based Therapeutic Targeting of Signaling Pathways in Breast Cancer. Biochim. et Biophys. Acta (BBA)-Mol. Basis Dis. 2024, 1870, 166872. [Google Scholar] [CrossRef]
  290. Shen, L.; Liu, Y.; Tso, P.; Wang, D.Q.-H.; Davidson, W.S.; Woods, S.C.; Liu, M. Silencing Steroid Receptor Coactivator-1 in the Nucleus of the Solitary Tract Reduces Estrogenic Effects on Feeding and Apolipoprotein A-IV Expression. J. Biol. Chem. 2018, 293, 2091–2101. [Google Scholar] [CrossRef] [PubMed]
  291. Ward, E.; Varešlija, D.; Charmsaz, S.; Fagan, A.; Browne, A.L.; Cosgrove, N.; Cocchiglia, S.; Purcell, S.P.; Hudson, L.; Das, S.; et al. Epigenome-Wide SRC-1–Mediated Gene Silencing Represses Cellular Differentiation in Advanced Breast Cancer. Clin. Cancer Res. 2018, 24, 3692–3703. [Google Scholar] [CrossRef]
  292. Ganji Arjenaki, R.; Samieepour, G.; Sadat Ebrahimi, S.E.; Pirali Hamedani, M.; Saffari, M.; Seyedhamzeh, M.; Kamali, A.N.; Najdian, A.; Shafiee Ardestani, M. Development of Novel Radiolabeled Antibody-Conjugated Graphene Quantum Dots for Targeted in Vivo Breast Cancer Imaging and Biodistribution Studies. Arab. J. Chem. 2024, 17, 105518. [Google Scholar] [CrossRef]
  293. Gennari, A.; André, F.; Barrios, C.H.; Cortés, J.; de Azambuja, E.; DeMichele, A.; Dent, R.; Fenlon, D.; Gligorov, J.; Hurvitz, S.A.; et al. ESMO Clinical Practice Guideline for the Diagnosis, Staging and Treatment of Patients with Metastatic Breast Cancer. Ann. Oncol. 2021, 32, 1475–1495. [Google Scholar] [CrossRef]
  294. Allison, K.H.; Hammond, M.E.H.; Dowsett, M.; McKernin, S.E.; Carey, L.A.; Fitzgibbons, P.L.; Hayes, D.F.; Lakhani, S.R.; Chavez-MacGregor, M.; Perlmutter, J.; et al. Estrogen and Progesterone Receptor Testing in Breast Cancer: ASCO/CAP Guideline Update. J. Clin. Oncol. 2020, 38, 1346–1366. [Google Scholar] [CrossRef]
  295. Henry, N.L.; Somerfield, M.R.; Dayao, Z.; Elias, A.; Kalinsky, K.; McShane, L.M.; Moy, B.; Park, B.H.; Shanahan, K.M.; Sharma, P.; et al. Biomarkers for Systemic Therapy in Metastatic Breast Cancer: ASCO Guideline Update. J. Clin. Oncol. 2022, 40, 3205–3221. [Google Scholar] [CrossRef]
  296. Wolff, A.C.; Somerfield, M.R.; Dowsett, M.; Hammond, M.E.H.; Hayes, D.F.; McShane, L.M.; Saphner, T.J.; Spears, P.A.; Allison, K.H. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: ASCO–College of American Pathologists Guideline Update. J. Clin. Oncol. 2023, 41, 3867–3872. [Google Scholar] [CrossRef] [PubMed]
  297. Colomer, R.; González-Farré, B.; Ballesteros, A.I.; Peg, V.; Bermejo, B.; Pérez-Mies, B.; de la Cruz, S.; Rojo, F.; Pernas, S.; Palacios, J. Biomarkers in Breast Cancer 2024: An Updated Consensus Statement by the Spanish Society of Medical Oncology and the Spanish Society of Pathology. Clin. Transl. Oncol. 2024, 26, 2935–2951. [Google Scholar] [CrossRef] [PubMed]
  298. Loibl, S.; André, F.; Bachelot, T.; Barrios, C.H.; Bergh, J.; Burstein, H.J.; Cardoso, M.J.; Carey, L.A.; Dawood, S.; Del Mastro, L.; et al. Early Breast Cancer: ESMO Clinical Practice Guideline for Diagnosis, Treatment and Follow-Up. Ann. Oncol. 2024, 35, 159–182. [Google Scholar] [CrossRef] [PubMed]
  299. Park, K.H.; Loibl, S.; Sohn, J.; Park, Y.H.; Jiang, Z.; Tadjoedin, H.; Nag, S.; Saji, S.; Yusof, M.M.; Villegas, E.M.B.; et al. Pan-Asian Adapted ESMO Clinical Practice Guidelines for the Diagnosis, Treatment and Follow-up of Patients with Early Breast Cancer. ESMO Open 2024, 9, 102974. [Google Scholar] [CrossRef]
  300. Al Sukhun, S.; Temin, S.; Barrios, C.H.; Antone, N.Z.; Guerra, Y.C.; Chavez-MacGregor, M.; Chopra, R.; Danso, M.A.; Gomez, H.L.; Homian, N.M.; et al. Systemic Treatment of Patients With Metastatic Breast Cancer: ASCO Resource–Stratified Guideline. JCO Glob. Oncol. 2024, 10, e2300285. [Google Scholar] [CrossRef]
  301. Kohaar, I.; Hodges, N.A.; Srivastava, S. Biomarkers in Cancer Screening. Hematol. Oncol. Clin. N. Am. 2024, 38, 869–888. [Google Scholar] [CrossRef]
  302. Alismail, H. Review: Merging from Traditional to Potential Novel Breast Cancer Biomarkers. J. King Saud Univ. Sci. 2024, 36, 103551. [Google Scholar] [CrossRef]
  303. Mooghal, M.; Anjum, S.; Khan, W.; Tariq, H.; Babar, A.; Vohra, L.M. Artificial Intelligence-Powered Optimization of KI-67 Assessment in Breast Cancer: Enhancing Precision and Workflow Efficiency. a Literature Review. J. Pak. Med. Assoc. 2024, 74, S109–S116. [Google Scholar] [CrossRef]
  304. Arima, N.; Nishimura, R.; Osako, T.; Nishiyama, Y.; Fujisue, M.; Okumura, Y.; Nakano, M.; Tashima, R.; Toyozumi, Y. The Importance of Tissue Handling of Surgically Removed Breast Cancer for an Accurate Assessment of the Ki-67 Index. J. Clin. Pathol. 2016, 69, 255–259. [Google Scholar] [CrossRef] [PubMed]
  305. Mikami, Y.; Ueno, T.; Yoshimura, K.; Tsuda, H.; Kurosumi, M.; Masuda, S.; Horii, R.; Toi, M.; Sasano, H. Interobserver Concordance of Ki67 Labeling Index in Breast Cancer: Japan Breast Cancer Research Group Ki67 Ring Study. Cancer Sci. 2013, 104, 1539–1543. [Google Scholar] [CrossRef]
  306. Fernezlian, S.M.; Baldavira, C.M.; de Souza, M.L.F.; Farhat, C.; de Vilhena, A.F.; Pereira, J.C.N.; de Campos, J.R.M.; Takagaki, T.; Balancin, M.L.; Ab’Saber, A.M.; et al. A Semi-Automated Microscopic Image Analysis Method for Scoring Ki-67 Nuclear Immunostaining. Braz. J. Med. Biol. Res. 2023, 56, e12922. [Google Scholar] [CrossRef] [PubMed]
  307. Cai, L.; Yan, K.; Bu, H.; Yue, M.; Dong, P.; Wang, X.; Li, L.; Tian, K.; Shen, H.; Zhang, J.; et al. Improving Ki67 Assessment Concordance by the Use of an Artificial Intelligence-empowered Microscope: A Multi-institutional Ring Study. Histopathology 2021, 79, 544–555. [Google Scholar] [CrossRef]
  308. Deng, Y.; Li, F.L.; Qin, H.Y.; Zhou, Y.Y.; Zhou, Q.Q.; Mei, J.; Li, L.; Liu, H.H.; Wang, Y.Z.; Bu, H.; et al. Sichuan Da Xue Xue Bao [Application Test of the AI-Automatic Diagnostic System for Ki-67 in Breast Cancer]. Journal of Sichuan University. Med. Sci. Ed. 2021, 52, 693–697. [Google Scholar] [CrossRef]
  309. Xie, N.; Zhou, H.; Yu, L.; Huang, S.; Tian, C.; Li, K.; Jiang, Y.; Hu, Z.-Y.; Ouyang, Q. Artificial Intelligence Scale-Invariant Feature Transform Algorithm-Based System to Improve the Calculation Accuracy of Ki-67 Index in Invasive Breast Cancer: A Multicenter Retrospective Study. Ann. Transl. Med. 2022, 10, 1067. [Google Scholar] [CrossRef] [PubMed]
  310. Li, L.; Han, D.; Yu, Y.; Li, J.; Liu, Y. Artificial Intelligence-Assisted Interpretation of Ki-67 Expression and Repeatability in Breast Cancer. Diagn. Pathol. 2022, 17, 20. [Google Scholar] [CrossRef]
  311. Chen, X.; Chen, D.G.; Zhao, Z.; Balko, J.M.; Chen, J. Artificial Image Objects for Classification of Breast Cancer Biomarkers with Transcriptome Sequencing Data and Convolutional Neural Network Algorithms. Breast Cancer Res. 2021, 23, 96. [Google Scholar] [CrossRef] [PubMed]
  312. Sigurjonsdottir, G.; De Marchi, T.; Ehinger, A.; Hartman, J.; Bosch, A.; Staaf, J.; Killander, F.; Niméus, E. Comparison of SP142 and 22C3 PD-L1 Assays in a Population-Based Cohort of Triple-Negative Breast Cancer Patients in the Context of Their Clinically Established Scoring Algorithms. Breast Cancer Res. 2023, 25, 123. [Google Scholar] [CrossRef] [PubMed]
  313. Boman, C.; Zerdes, I.; Mårtensson, K.; Bergh, J.; Foukakis, T.; Valachis, A.; Matikas, A. Discordance of PD-L1 Status between Primary and Metastatic Breast Cancer: A Systematic Review and Meta-Analysis. Cancer Treat. Rev. 2021, 99, 102257. [Google Scholar] [CrossRef]
  314. Xie, W.; Suryaprakash, S.; Wu, C.; Rodriguez, A.; Fraterman, S. Trends in the Use of Liquid Biopsy in Oncology. Nat. Rev. Drug Discov. 2023, 22, 612–613. [Google Scholar] [CrossRef]
  315. Siravegna, G.; Marsoni, S.; Siena, S.; Bardelli, A. Integrating Liquid Biopsies into the Management of Cancer. Nat. Rev. Clin. Oncol. 2017, 14, 531–548. [Google Scholar] [CrossRef] [PubMed]
  316. Moser, T.; Heitzer, E. Surpassing Sensitivity Limits in Liquid Biopsy. Science 2024, 383, 260–261. [Google Scholar] [CrossRef]
  317. Martin-Alonso, C.; Tabrizi, S.; Xiong, K.; Blewett, T.; Sridhar, S.; Crnjac, A.; Patel, S.; An, Z.; Bekdemir, A.; Shea, D.; et al. Priming Agents Transiently Reduce the Clearance of Cell-Free DNA to Improve Liquid Biopsies. Science 2024, 383, eadf2341. [Google Scholar] [CrossRef]
  318. Oxnard, G.R.; Paweletz, C.P.; Kuang, Y.; Mach, S.L.; O’Connell, A.; Messineo, M.M.; Luke, J.J.; Butaney, M.; Kirschmeier, P.; Jackman, D.M.; et al. Noninvasive Detection of Response and Resistance in EGFR-Mutant Lung Cancer Using Quantitative next-Generation Genotyping of Cell-Free Plasma DNA. Clin. Cancer Res. 2014, 20, 1698–1705. [Google Scholar] [CrossRef] [PubMed]
  319. Ding, Y.; Li, W.; Wang, K.; Xu, C.; Hao, M.; Ding, L. Perspectives of the Application of Liquid Biopsy in Colorectal Cancer. BioMed Res. Int. 2020, 2020, 6843180. [Google Scholar] [CrossRef] [PubMed]
  320. Batool, S.M.; Yekula, A.; Khanna, P.; Hsia, T.; Gamblin, A.S.; Ekanayake, E.; Escobedo, A.K.; You, D.G.; Castro, C.M.; Im, H.; et al. The Liquid Biopsy Consortium: Challenges and Opportunities for Early Cancer Detection and Monitoring. Cell Rep. Med. 2023, 4, 101198. [Google Scholar] [CrossRef] [PubMed]
  321. Im, Y.R.; Tsui, D.W.Y.; Diaz, L.A.; Wan, J.C.M. Next-Generation Liquid Biopsies: Embracing Data Science in Oncology. Trends Cancer 2021, 7, 283–292. [Google Scholar] [CrossRef]
  322. Darrigues, L.; Pierga, J.Y.; Bernard-Tessier, A.; Bièche, I.; Silveira, A.B.; Michel, M.; Loirat, D.; Cottu, P.; Cabel, L.; Dubot, C.; et al. Circulating Tumor DNA as a Dynamic Biomarker of Response to Palbociclib and Fulvestrant in Metastatic Breast Cancer Patients. Breast Cancer Res. 2021, 23, 31. [Google Scholar] [CrossRef] [PubMed]
  323. Wang, K.; Wang, X.; Pan, Q.; Zhao, B. Liquid Biopsy Techniques and Pancreatic Cancer: Diagnosis, Monitoring, and Evaluation. Mol. Cancer 2023, 22, 167. [Google Scholar] [CrossRef] [PubMed]
  324. Zhao, Y.; Tang, J.; Jiang, K.; Liu, S.Y.; Aicher, A.; Heeschen, C. Liquid Biopsy in Pancreatic Cancer—Current Perspective and Future Outlook. Biochim. Biophys. Acta Rev. Cancer 2023, 1878, 188868. [Google Scholar] [CrossRef] [PubMed]
  325. Zhou, H.; Zhu, L.; Song, J.; Wang, G.; Li, P.; Li, W.; Luo, P.; Sun, X.; Wu, J.; Liu, Y.; et al. Liquid Biopsy at the Frontier of Detection, Prognosis and Progression Monitoring in Colorectal Cancer. Mol. Cancer 2022, 21, 86. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the five main intrinsic subtypes of breast cancer. Each intrinsic subtype shows distinct molecular signatures and clinical outcomes, ranging from hormone receptor-positive Luminal types to highly aggressive Basal-like and Claudin-low tumors.
Figure 1. Schematic representation of the five main intrinsic subtypes of breast cancer. Each intrinsic subtype shows distinct molecular signatures and clinical outcomes, ranging from hormone receptor-positive Luminal types to highly aggressive Basal-like and Claudin-low tumors.
Ijms 27 00138 g001
Figure 2. Summary of the main emerging molecular biomarkers in breast cancer. Emerging biomarkers include non-coding RNAs, epigenetic, genomic, and immune markers that collectively contribute to tumor progression, therapeutic response, and precision oncology.
Figure 2. Summary of the main emerging molecular biomarkers in breast cancer. Emerging biomarkers include non-coding RNAs, epigenetic, genomic, and immune markers that collectively contribute to tumor progression, therapeutic response, and precision oncology.
Ijms 27 00138 g002
Table 1. Summary of Novel Therapeutic Targets Being Investigated in Breast Cancer Trials.
Table 1. Summary of Novel Therapeutic Targets Being Investigated in Breast Cancer Trials.
TherapyDrug NameClinical Trial Stage/ConditionActionAdverse Effects (AE)/LimitationsReference
8.1 Therapies Targeting Altered Signaling Pathways in breast cancer
PI3K/AKT/mTOR pathway inhibitorsAlpelisib (BYL719)
+ fulvestrant
FDA-approved for HR+/HER2-treatment with PIK3CA variants
(NCT02437318)
Inhibits cellular proliferation
by targeting both wild-type PI3K-alpha and PI3K-alpha containing canonical variants
AE: Hyperglycemia, diarrhea, nausea, decreased appetite, rash, or maculopapular rash.[229,231,253]
Limitations: It is less potent against either PI3K-sigma or -gamma.
AKT inhibitorsCapivasertib (AZD5363)Phase III + fulvestrant for hormone receptor- positive and HER2 negative advanced breast cancer (ABC) with PIK3CA/AKT1/PTEN alterations
(NCT04305496)
Potent inhibitor of the AKT protein (Protein Kinase B or PKB) across all three isoforms (AKT1, 2,3), blocking PI3K. AKT signaling pathway that drives cancer cell growth, survival, and metabolismAE: Hyperglycemia, severe hyperglycemia with mixed diabetic ketoacidosis (DKA) and hyperosmolar hyperglycemic state (HHS).[230,254,255]
Limitation: It is CYP3A4-dependent metabolism which causes drug interactions and variability. Thus, the development of resistance through alternative signaling pathways, and the need for specific biomarkers.
mTOR inhibitorsRapamycinPhase II for HER-2 receptor positive metastatic breast cancer-Rapamycin + Trastuzumab (NCT00411788)Targets FKPB12/mTORC1.
Inhibits lymphocyte activation and induces cell cycle arrest.
Rapamycin to FKBP12 binding inhibits the activity of mTORC1 leading to a decrease in protein synthesis, increased autophagy, and inhibition of cell growth.
AE Rapamycin: Bone pain, diarrhea, headache, blurred vision, and chest pain.
AE Everolimus: Loss of appetite, fatigue, diarrhea, swelling, and nausea.
AE Temirolimus: Rash, itching, chest pain, hives, and flushing.
[232,256,257]
Everolimus (RAD001)
Temsirolimus (CCI-779)
Phase III for patients with breast cancer (NCT01674140)
Phase III for rhabdomyosarcoma patients (NCT00703625)
Targets mTORC1.
Reduces vascular endothelial growth factor (VEGF) expression and inhibits glycolysis.
Inhibits mTOR activity and regulates cell division
Limitation: Suppression of mTOR triggers compensatory signaling circuits acting on upstream nodes, which in turn enhance tumor cell viability, division, and metastatic potential.[232,240,258,259]
AZD8055Phase I for solid tumors and lymphoma.
(Withdrawn)
Phase I for breast cancer and lung cancer-AZD8055 + paclitaxel
(NCT02193633)
Target both mTORC1 and mTORC2 complexes and induce cell deathAE: Increased alanine aminotransferase (22%), increased aspartate aminotransferase (22%) and fatigue (16%).[258,260]
Limitation: Variants in mTOR with clinical relevance increase its catalytic function and diminish the effectiveness of inhibitors.
Dual PI3K/mTOR inhibitorsGedatolisibPhase III + fulvestrant and CDK4/6 inhibitors in individuals with HR+/HER2− locally advanced or metastatic breast cancer.
(NCT03400254)
It inhibits PI3K and mTOR. Inhibits tumor growth and increase survival time.AE: Nausea (53%), mucosal inflammation (50%), decreased appetite (40%), diarrhea (38%), fatigue (35%), and dysgeusia and vomiting (each 30%).[231,261,262,263]
Limitation: Resistance driven by ABC transporters (ABCB1, ABCG2), difficulty preventing metastasis from dormant cells, and side effects such as stomatitis remain concerns, even though hyperglycemia and diarrhea occur less frequently than with other PI3K inhibitors.
Autophagy inhibitorsHydroxychloroquineFDA approved for treating malaria, rheumatoid arthritis, and lupus.
Currently studying as an anticancer drug in combination with other therapies (like Palbociclib).
(NCT04841148)
Increase protein acetylation. Inhibition of malignant cell growth, viability.AE: Fatigue, nausea, diarrhea, constipation, and appetite loss.[234,264,265]
Limitation: Its use is constrained by poor bioavailability and broad, non-targeted distribution. In addition, HCQ’s therapeutic impact is reduced because it does not efficiently cross tumor cell membranes within the acidic tumor microenvironment
Glutaminase or fatty acid synthase inhibitorsFASN inhibitor (TVB-2640)Phase I for treatment for combinatorial treatment for breast cancer and KRAS-positive lung cancer.
(NCT02223247)
It suppresses AKT phosphorylation, promotes apoptosis in tumor cells, increases the sensitivity of chemotherapy-resistant tumors to treatment, and reduces tumor growth in mouse xenograft models.AE: Dry eyes, fatigue, dry skin, mucositis, and nauseas.[234,266]
Limitation: reliance on combination therapies due to limited monotherapy responses, the need to manage adverse effects such as palmar–plantar erythrodysesthesia, and strict patient eligibility criteria that exclude individuals with significant gastrointestinal or cardiac conditions.
Denifanstat (TVB-2640)Phase II for relapsing high-grade astrocytoma
(NCT03032484)
Phase III for patients with metabolic dysfunction-associated steatohepatitis and F2/F3 fibrosis
(NCT06594523)
Acts synergistically with the topoisomerase inhibitor SN-38 in TNBC brain metastasis cell lines, increases FAS expression, reduces the expression of cell-cycle–related genes, and decreases the motility of these TNBC bone marrow cells.AE: Dry skin, dry eye and sometimes alopecia.[234,267,268,269]
Limitations: More studies are required for effectiveness directly on breast cancer patients. Fat-soluble aids absorption but increases off-target effects. Clinical trials have limitations such as small sample size and short treatment durations.
PROTACsVepdegestrant
(ARV-471)
Phase III for advanced metastatic breast cancer.
(NCT05654623)
Block or degrade tumor-driving proteins and are associated with significantly longer progression-free survival compared with fulvestrant.AE: Fatigue, liver enzyme elevations, nausea and hot flashes.[263,270,271]
Limitation: Limited oral absorption due to their size and polarity, intricate linker optimization, risks of degrading unintended proteins, delivery and cell-permeability barriers, potential overload of the proteasome, immunogenicity concerns, and difficulties in assessing pharmacokinetics and pharmacodynamics.
SERDsFulvestrant (Faslodex)FDA-approved for advanced ER-positive breast cancer
(NCT01602380)
Binds ER in cancer cells, blocking estrogen signaling, and accelerating receptor degradation, thereby reducing tumor growth in hormone receptor–positive breast cancer.AE: Bone pain, diarrhea, fatigue, hot flashes, nausea, and headache.[272,273]
Limitation: Poor water solubility, necessitating monthly intramuscular injections. Mechanism involves ER degradation, leading to potential resistance via receptor upregulation.
Imlunesrtant (Inluriyo)Phase III for ER+/HER2-, and ABC
(NCT04975308)
Binds to ER, particularly ERα, causing a conformational change that marks the receptor for degradation via the proteasome system, effectively eliminating it from the cell.AE: Fatigue, decreased calcium and neutrophils, musculoskeletal pain, and increased liver enzymes (AST/ALT).[234,274]
Limitation: Resistance to ESR1 variants and inconsistency in brain penetration.
Telomerase inhibitorBIBR1591In silico, In vitro, In vivo pre-clinical assessmentsInduces apoptosis in altered gene expression, having anticancer effect as its expression of CDH13, DAPK1, and NR4A3 genes.AE: Low hemoglobin/calcium/neutrophils/platelets, fatigue, musculoskeletal pain, diarrhea, nausea, constipation, elevated liver enzymes (AST/ALT), and increased cholesterol/triglycerides.[237,275]
Limitation: Longer treatment times (lag phase) to see effects due to telomere shortening, challenges with potency, selectivity, and drug-like properties, and potential for off-target effects as it impacts cancer cell proliferation and apoptosis
S1P signaling inhibitorPro-FTY
Fisetin
Phase II for breast cancer survivors–PROFFi (NCT06113016)Efficient against multi-drug-resistant breast cancer.
Targets S1P signaling in cancer cells, potentially affecting immune cells (lymphocytes) and tumor growth.
AE: Diarrhea, nausea, constipation, and possible liver enzyme changes.[275,276]
Limitation: Poor bioavailability (rapid metabolism, low solubility), requiring advanced delivery systems (nanoparticles) for better tumor targeting, and understanding its complex interaction with the Nrf2/HO-1 pathway which can promote cancer survival (careful doses consideration).
FTY720
(Fingolimod)
Soon to start early phase clinical trials for HER-2 positive breast cancer resistant to trastuzumab
Phase III for relapsing-remitting multiple scleorosis (NCT00662649)
Induces apoptosis through ROS, suppressing survival pathways such as Akt/mTOR, and targeting cancer stem cells by lowering Oct4/Sox2/Nanog. It also limits metastasis by reducing migration and invasion (including MMP activity), disrupting cytoskeletal structures, and can act synergistically with drugs like tamoxifen or EGFR inhibitors, partly via PP2A activation and modulation of the autotaxin–LPA axis.AE: Headache, nausea, fatigue, dizziness, cardiovascular problems, fungal infections, and enzyme elevation. [244,277,278,279]
Limitation: It requires in vivo phosphorylation by SphK1/2 to activate it (FTY720-P).
It also has off-target effects by affecting S1P1,3,4,5 receptors and CB1.
Has more side effects for its variable cellular response.
8.2 Immunotherapy and checkpoint inhibitors
PD-1/PD-L1 inhibitorsPembrolizumab (Keytruda)
Targeting PD-1
Phase III for triple-negative breast cancer.
(NCT02819518)
Expression of PD-L1 in ER/PR negative breast tumors (NCT03197389)
Blocks the PD-1 receptor on T cells, preventing cancer cells from using the PD-L1/PD-L2 pathway to evade the immune system, thereby allowing the body to attack the tumors.AE: Diarrhea, nausea, fatigue, pain, rash, itching, cough, and fever.[280,281]
Limitation: It is ineffective in tumors with low PD-L1 expression and can develop resistance even in those with high PD-L1 levels. It may also overstimulate the immune system as an off-target effect.
Atezolizumab (Tecentriq)
Targeting PD-L1
Phase IIIb for advanced or metastatic PD-L1-positive triple-negative breast cancer
(NCT04148911)
Blocks the PD-L1 protein on cancer and immune cells, preventing it from binding to PD-1 on T cells. This keeps the immune response from being switched off, allowing the immune system to attack the tumor.AE: Fatigue, cough, decreased appetite, difficulty in breathing, anemia, constipation, fever, and diarrhea.[282,283]
Limitation: It is susceptible to resistance through alternative immune checkpoints (such as CTLA-4), tumor heterogeneity in PD-L1 expression, poor T-cell infiltration, or the development of an immunosuppressive tumor microenvironment (e.g., expansion of myeloid-derived suppressor cells).
ImmunotherapyTrastuzumab deruxtecan
(T-DXd)
Phase III for HER2-positive metastatic breast cancer, and first-line metastatic breast cancer
(NCT04784715)
It is an antibody–drug conjugate (ADC) that links trastuzumab (which targets the HER2 protein) to a potent chemotherapy payload (deruxtecan). This design allows it to kill HER2-positive cancer cells and, through a “bystander effect,” also nearby tumor cells, improving treatment outcomes in HER2-positive and HER2-low breast cancer and other solid tumors.AE: Fatigue, constipation, diarrhea, alopecia, neutropenia, increased liver enzymes, and anemia.
[242,284,285]
Limitation: It has challenges in predicting the response it would generate. It is not immune to resistance mechanisms of cancer cells like decreased HER2 expression, altered ADC internalization, and issues with payload release/action (like CTSL activity).
8.3 Nucleic Acid-Based Therapies
miRNA therapymiR-4477aPre-clinical assessments in co-culture in vitro breast cancer cell line.They bind to three separate regions of PD-L1 mRNA with high affinity (94%, 88%, and 80%), allowing them to effectively target and suppress key regulatory pathways essential for cancer cell function.AE: No clinical trial performed yet, so there are any reported.[243,245,286]
Limitation: It has challenges in delivery efficiency, understanding complex roles in cell cycles, and instability. Likewise, there are no clinical trials performed, so the is limited information.
Circular RNAs therapycirc-ARHGER28Pre-clinical assessments on MCF-7 cells for breast cancer.It functions as a tumor suppressor in breast cancer by limiting cell proliferation and inducing apoptosis, mainly through inhibition of the PI3K/AKT/mTOR pathway, ultimately slowing tumor growth and highlighting its therapeutic potential.
It promotes cisplatin resistance in ovarian cancer.
AE: No clinical trial performed yet, so there are any reported.[246,287]
Limitation: It has been evidence of resistance to treatment in pre-clinical trials by genome evolution of cancer cells. There is still limited access to information.
CRISPR-based technologiesDisrupting ITGA9 (Integrin Alpha 9)Pre-clinical assessments on mouse models.Inhibits or reduces the ITGA9 function, often using antagonists, shRNA, or siRNA, to block its role in cell adhesion, migration, and inflammation.AE: No clinical trial performed yet, so there are any reported.[247,248,288,289]
Limitation: It involves complex pathway compensation (like other integrins, α5β1), context-dependent effects (strong in inflammation/angiogenesis), and varied mechanisms for achieving depletion (gene loss vs. epigenetic silencing). There are complex interactions that need understanding and limited studies.
Silencing SRC-1Pre-clinical assessments in ex vivo metastatic tumors of endocrine-treated human breast cancer.Suppressing SRC-1 reduces metastatic spread in breast cancer, even though it does not significantly affect primary tumor growth.AE: No clinical trial performed yet, so there are any reported.[247,290,291]
Limitation: SRC-1 is suppress but it is a key coactivator for hormone receptors, nuclear organization, and multiple signaling pathways. Thus, disrupting SRC-1 can interfere with these essential functions, causing altered cellular responses.
8.4 Nanotechnology Applied to Targeted Drug Delivery
Graphene quantum dotsGraphene quantum dots +
Pembrolizumab
Pre-clinical assessments in in vivo human breast cancer imaging.Provide effective and targeted delivery with enhanced solubility, reduced toxicity, and theragnostic potential by enabling simultaneous treatment and imaging.
High tumor activity and specific targeting using a radiolabeled probe.
AE: No clinical trial performed yet, so any reported.[249,292]
Limitation: It has complex synthesis, and unknown long-term biodistribution.
There is limited access to information and studies.
Table 2. Comparison of biomarker recommendations in breast cancer across major clinical guidelines.
Table 2. Comparison of biomarker recommendations in breast cancer across major clinical guidelines.
Routine Clinical Use (Standard of Care)
ESMO [293,298] ASCO [300] Pan-Asian [299] Spanish [297]
ER, PR, HER2 (IHC ± ISH) for all invasive cancers; re-test in MBCER, PR (IHC) for all invasive cancersER, PR (IHC) for all invasive cancersER, PR, HER2, Ki-67 (IHC ± ISH) in all EBC
Ki-67 as part of initial risk assessmentHER2 (IHC/ISH) for all invasive cancersHER2 (IHC ± ISH) for all invasive cancersHER2-low reporting (0 vs. 1+) to enable T-DXd
Multigene assays (HR+/HER2- EBC, uncertain chemo benefit)Oncotype DX (HR+/HER2-, node-negative)Validated multigene assays (HR+/HER2- EBC)Oncotype DX®, MammaPrint®, Prosigna®, EndoPredict® in ER+/HER2- EBC
PD-L1 testing in metastatic TNBCPD-L1 testing in TNBCPD-L1 testing in metastatic TNBC
PIK3CA testing in HR+/HER2- MBCPIK3CA mutations → alpelisibPIK3CA testing in HR+/HER2- ABC
gBRCA1/2 in EBC (adjuvant olaparib) and in HER2- MBC (PARP inhibitors)gBRCA1/2 mutations → PARP inhibitorsgBRCA1/2 testing for adjuvant olaparibgBRCA1/2 in high-risk HER2- EBC and advanced disease
MSI-H/dMMR, TMB-high for immunotherapySame
NTRK fusions for TRK inhibitorsSame
ESR1 mutations (ctDNA) to guide endocrine therapy (HR+/HER2- ABC).
Research/Limited or optional use
ESMO [293,298] ASCO [300] Pan-Asian [299] Spanish [297]
ESR1, somatic BRCA, HER2-low (optional)ESR1 (emerging)
MSI-H/dMMR, TMB-high, NTRK (only if matched drugs available)SameSameSame
TILs (prognostic; no treatment cut-offs)SameSameSame
TROP2 (investigational for ADCs)SameNot required for sacituzumab govitecan
PALB2 (possible PARP use, not established)Same
ctDNA/CTCs (prognostic, not for therapy decisions)Same
HRD beyond BRCA, AKT-pathway, broad NGS (investigational)Same
Not recommended
ESMO [293,298] ASCO [300] Pan-Asian [299] Spanish [297]
PD-L1 in EBCRoutine HRD testing outside BRCAPD-L1 in EBC (not predictive in neoadjuvant TNBC)Routine MSI-H/dMMR testing (rare in breast cancer)
Broad genomic profiling or ctDNA when not actionableRoutine TROP2 testingRoutine lab tumor markers/extensive imaging for allRoutine broad NGS for off-label therapy
Routine ctDNA/CTCs for monitoringPD-L1 for EBC (benefit in TNBC regardless of PD-L1)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Intriago-Baldeón, D.P.; Pérez-Coral, E.S.; Armas Samaniego, M.I.; Romero, V.I.; Pozo Palacios, J.C.; Bigoni-Ordóñez, G.D. Decoding Breast Cancer: Emerging Molecular Biomarkers and Novel Therapeutic Targets for Precision Medicine. Int. J. Mol. Sci. 2026, 27, 138. https://doi.org/10.3390/ijms27010138

AMA Style

Intriago-Baldeón DP, Pérez-Coral ES, Armas Samaniego MI, Romero VI, Pozo Palacios JC, Bigoni-Ordóñez GD. Decoding Breast Cancer: Emerging Molecular Biomarkers and Novel Therapeutic Targets for Precision Medicine. International Journal of Molecular Sciences. 2026; 27(1):138. https://doi.org/10.3390/ijms27010138

Chicago/Turabian Style

Intriago-Baldeón, Dámaris P., Eduarda Sofía Pérez-Coral, Martina Isabella Armas Samaniego, Vanessa I. Romero, Juan Carlos Pozo Palacios, and Gabriele Davide Bigoni-Ordóñez. 2026. "Decoding Breast Cancer: Emerging Molecular Biomarkers and Novel Therapeutic Targets for Precision Medicine" International Journal of Molecular Sciences 27, no. 1: 138. https://doi.org/10.3390/ijms27010138

APA Style

Intriago-Baldeón, D. P., Pérez-Coral, E. S., Armas Samaniego, M. I., Romero, V. I., Pozo Palacios, J. C., & Bigoni-Ordóñez, G. D. (2026). Decoding Breast Cancer: Emerging Molecular Biomarkers and Novel Therapeutic Targets for Precision Medicine. International Journal of Molecular Sciences, 27(1), 138. https://doi.org/10.3390/ijms27010138

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop