Next Article in Journal
Development of an Inflammation-Triggered In Vitro “Leaky Gut” Model Using Caco-2/HT29-MTX-E12 Combined with Macrophage-like THP-1 Cells or Primary Human-Derived Macrophages
Next Article in Special Issue
A Facile Synthesis and Molecular Characterization of Certain New Anti-Proliferative Indole-Based Chemical Entities
Previous Article in Journal
Transcriptomic Complexity of Culm Growth and Development in Different Types of Moso Bamboo
Previous Article in Special Issue
RanBP1: A Potential Therapeutic Target for Cancer Stem Cells in Lung Cancer and Glioma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Clinical Utility of lncRNAs and Their Application as Molecular Biomarkers in Breast Cancer

by
Cristian Arriaga-Canon
1,*,†,
Laura Contreras-Espinosa
1,2,†,
Sergio Aguilar-Villanueva
3,
Enrique Bargalló-Rocha
3,
José Antonio García-Gordillo
4,
Paula Cabrera-Galeana
4,
Clementina Castro-Hernández
1,
Francisco Jiménez-Trejo
5 and
L. A. Herrera
1,6,*
1
Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C.P. 14080, Mexico
2
Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, Mexico City C.P. 04510, Mexico
3
Departamento de Tumores Mamarios, Instituto Nacional de Cancerología, Tlalpan, Mexico City C.P. 14080, Mexico
4
Departamento de Oncología Médica de Mama, Instituto Nacional de Cancerología, Tlalpan, Mexico City C.P. 14080, Mexico
5
Instituto Nacional de Pediatría, Coyoacán, Mexico City C.P. 04530, Mexico
6
Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey C.P. 64710, Mexico
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(8), 7426; https://doi.org/10.3390/ijms24087426
Submission received: 23 February 2023 / Revised: 4 April 2023 / Accepted: 10 April 2023 / Published: 18 April 2023
(This article belongs to the Special Issue Novel Biological Molecules for Cancer Treatments 2.0)

Abstract

:
Given their tumor-specific and stage-specific gene expression, long non-coding RNAs (lncRNAs) have demonstrated to be potential molecular biomarkers for diagnosis, prognosis, and treatment response. Particularly, the lncRNAs DSCAM-AS1 and GATA3-AS1 serve as examples of this because of their high subtype-specific expression profile in luminal B-like breast cancer. This makes them candidates to use as molecular biomarkers in clinical practice. However, lncRNA studies in breast cancer are limited in sample size and are restricted to the determination of their biological function, which represents an obstacle for its inclusion as molecular biomarkers of clinical utility. Nevertheless, due to their expression specificity among diseases, such as cancer, and their stability in body fluids, lncRNAs are promising molecular biomarkers that could improve the reliability, sensitivity, and specificity of molecular techniques used in clinical diagnosis. The development of lncRNA-based diagnostics and lncRNA-based therapeutics will be useful in routine medical practice to improve patient clinical management and quality of life.

1. Introduction

Long non-coding RNAs (lncRNAs) are known to be tissue [1], cancer specific [2], and disease specific in breast cancer [3]. In particular, their expression has also been reported to be distinct under several conditions, for example, in cancer progression or in disease resistant to systemic treatments [4]. Currently, several examples of lncRNAs have been described as potential clinical biomarkers for predicting response to therapy or for prognosis in breast cancer, such as HOTAIR, H19, and DSCAM-AS1. HOTAIR, a −2 kb lncRNA located in chromosome 12, regulates gene expression by recruiting PRC2 and LSD1 complexes to their target gene regions, modulating the enrichment of H3K27 and H3K4 methylation [5], and it has also been described that HOTAIR overexpression increases cell proliferation by an enhanced estrogen receptor signaling pathway [6]. H19 is also a −2 kb lncRNA, located in chromosome 11, and evidence suggests that it modulates gene expression by the miRNA sponge mechanism [7]. These two lncRNAs, HOTAIR and H19, have been demonstrated to be overexpressed in breast cancer; this has been related with predictive value to assess resistance to neoadjuvant chemotherapy in breast cancer [8,9]. HOTAIR has been particularly associated with metastasis in breast cancer [10]. Additionally, DSCAM-AS1 is another lncRNA of −1 kb of length, located in chromosome 21, whose molecular mechanism is still unknown, but several studies suggest that it has a miRNA sponge function, particularly in carcinogenesis [11], and its silencing induces a reduction in proliferation in breast cancer cell lines [12]; its overexpression has been associated with resistance to tamoxifen therapy [13]. Additionally, GATA3-AS1 was first identified as a lncRNA, differentially expressed in breast cancer patients by Zhang, et al. [14]. This lncRNA of ~2 kb of length is in chromosome 10, and it has also been associated with Th2 lymphocytes differentiation [15] by regulating its adjacent gene GATA3, a transcriptional factor involved in immune response [16] and breast cancer [17]. The mechanisms by which GATA3-AS1 regulates the function of GATA3 are not fully understood, but Gibbons et al. described a molecular mechanism in human T lymphocytes in which GATA3-AS1 forms an R-Loop in GATA3 locus; this structure allows for the recruitment of a chromatin remodeler, such as MLL, leading to the activation of GATA3 transcription [16]. Because of its expression in breast cancer, GATA3-AS1 has also been proposed as a potential biomarker of response to neoadjuvant chemotherapy in patients with locally advanced breast cancer of Luminal B subtype [17]; it is also related to the immune response since it regulates the differentiation of Th2 lymphocytes, as mentioned before [16]. Furthermore, Zhang et al. reported that its expression is related to the immune response of mammary tumors by stabilizing protein levels of PD-L1 in triple negative breast cancer [18]. Taken together, these results suggest that GATA3-AS1 could be used in clinical practice as a biomarker that would form a part of targeted therapy strategies for PD-L1 positive tumors in triple negative breast cancer, for which pertuzumab, atezolizumab, and nab-paclitaxel are implemented as treatments [19]. Thus, lncRNAs are associated with several clinical conditions, such as resistance to therapy or metastasis development in breast cancer. This gives the possibility to propose them as novel molecular biomarkers for clinical practice. However, as is the case for many lncRNAs, the research to determine their clinical application is limited by several factors, including study design and the number of samples used to determine the clinical utility of lncRNAs.

2. Sample Size Calculation for Biomarker Discovery in Clinical Research

Currently, it is necessary to direct research efforts towards the use of lncRNAs in targeted therapies with sample sizes that enable adequate sensitivity and specificity [20]. For example, the studies performed by Contreras-Espinosa et al. and other research works in translational medicine, in which they identified the lncRNAs GATA3-AS1 [17], LINC02544 [21], and H19 [9], are related to neoadjuvant chemotherapy response in luminal breast cancer phenotypes, while lncRNA AC009283.1 was associated with tumorigenic cell signaling pathways in HER-2 enriched breast cancer phenotypes [22]; together, these results demonstrated the association of lncRNAs with different clinical characteristics in breast cancer. However, a common characteristic of these studies is the small sample size. For the identification of a molecule as a novel biomarker, like a lncRNA, the study population should be as large as the study needs to obtain from it a reliable result about lncRNAs to be candidates for biomarker development, which means that the study must be planned with adequate statistical criteria, considering the lowest possible p-value (p-value < 0.05), which would allow obtaining lncRNAs as biomarker candidates. To identify the greatest potential for predictive biomarkers with clinical applicability, several requirements have been described previously by Pepe et al. who designed the prospective-specimen-collection, retrospective-blinded-evaluation (ProBE) for selecting samples [23], including specific criteria, such as the definition of biomarker performance and the proportion of useful markers that the study should identify and the tolerable number of useless markers. This represents the guidelines that are now applied routinely in validation research, but not in discovery research, and they could help improve the quality, as well as usefulness, in lncRNA biomarker discovery.
To achieve this goal, the sample size must be properly determined depending on the phase of the study and the type of study to be carried out [24]. For the discovery phase in preclinical studies for biomarker discovery, based on RNA expression, it is important to consider implementing the Next Generation Sequencing approach, due to its ability to analyze the entire transcriptome through the RNA-Seq technique [25]. Particularly, in the discovery phase for preclinical studies based on RNA-Seq data analysis, the sample size can be small (n > 3) because the statistical power of the study relies on the type of transcript of interest (protein coding genes or ncRNAs) and in the reduction of the dispersion generated by the biological heterogeneity [26,27]. The principal parameters that can be used to improve statistical power are the design of the library (a paired-end library enhances more statistical power compared to single-end libraries), the design of the study (a tissue level expression analysis decreases dispersion compared to population study), and the depth of the sequencing (a study with a depth greater than 25 million reads increases the statistical power) [26]. However, it is desirable to have at least three biological replicates for each biological condition to be analyzed, for example, for the discovery of predictive biomarkers, three samples per each responder patient and three samples per each non-responder patient should be considered in the experimental design of a case-control study (n = 6) [28]. Additionally, it is necessary to determine the data distribution for subsequent statistical analysis, which includes the use of normal distribution tests, such as Kolmogorov-Smirnov and Shapiro-Wilk tests [29]. If data is normally distributed, then parametric tests, such as t-test, ANOVA test, linear regression, and Pearson correlation test, are implemented to determine the clinical association of the candidate lncRNA [30]. If data follows a non-normal distribution, then non-parametric tests, such as Fisher’s exact test, Mann-Whitney U test, and Spearman correlation test, are implemented to determine these associations [31], which should be validated in a larger and independent cohort of patients [32].
For the validation phase, it is assumed that the results of the discovery phase will be replicated, and to achieve this, we must consider fitting the sample size of the replication phase based on the conditional power calculation, which is the probability that the replication study leads to a statistically significant conclusion, as is described by Micheloud and Held [28]. Once the candidates for use as predictive biomarkers are selected, the results should be validated in larger cohorts in the Replication Phase, considering only the expression levels of candidate RNAs and their predictive value. This must be done by determining the data distribution, calculating sensitivity, and specificity for each candidate or for the whole set, which leads to the development of a molecular signature, which assumes normal distribution [33,34]. The aim of this method is to determine the true positives plus false negatives value based on the pre-established distribution value (Z = 1.96), sensitivity, specificity, and the maximum confidence interval of 95% (W = 0.1). For biomarkers, in general, the minimum acceptable values for sensitivity and specificity are 0.8 (80%) and 0.6 (60%), respectively [35]; thus, it is desirable to set these values for further calculations and determination of the optimal sample size (N). Finally, each sample size (sensitivity and specificity) is added to obtain the total sample size required for the study [33], which is statistically optimal for obtaining reliable information to determine the utility of the candidate biomarker in clinical practice. Here, we present a compilation of the scientific literature dealing with lncRNAs, and their clinical utility in breast cancer is presented, for which we performed an exhaustive search conducted by Pubmed, Google Scholar, and Connected Papers using keywords relating to lncRNAs and their clinical application in breast cancer, which includes “biomarkers”, “non-coding RNAs”, “lncRNAs”, “breast cancer”, “prognosis”, “prediction”, “RNA-Seq”, “transcriptome”, “sample size”, and “clinical trials”.

Sample Size Determination for lncRNA Studies in Breast Cancer Research

One example of optimal sample size is the study of Niknafs et al., in which they included 946 patients for the discovery phase and 758 patients for the validation phase. They described the usefulness of the lncRNA DSCAM-AS1 as a biomarker of resistance to tamoxifen in endocrine therapy because of its overexpression in patients with estrogen receptor positive breast cancer [13], as it is also the case for GATA3-AS1 [17], for which the accurate sample size for the replication phase should be 1295 patients [33]. Despite this, the use of a convenient sample size in studies for lncRNA biomarker discovery is also relevant for the identification of predictive biomarkers because several studies have identified predictive biomarkers in neoadjuvant chemotherapy, such as H19 [9], MALAT1 [36], and LINC02544 [21], which, despite the reduced sample size, showed a potential utility in clinical application. Additionally, public databases in which the results of the expression analyses are stored by massive parallel sequencing of coding genes, such as GEPIA [37], as well as non-coding genes, such as TANRIC [38], both corresponding to the expression profiles of patients found in the Cancer Genome Atlas (TCGA) cohorts, validates the results obtained in studies such as those mentioned above, in which it was not possible to obtain more samples. In summary, the importance of properly implementing the design of a clinical study ensures the reliability of the obtained data in the results, and therefore, it could be validated in a replication phase to determine the clinical value of a biomarker based on lncRNA.

3. The Current Use of lncRNAs as Clinical Biomarkers in Clinical Practice

Although their usefulness in clinical practice is poorly understood, the use of lncRNAs as predictive biomarkers in response to therapy has advantages compared to protein-based and mRNA-based biomarkers [39] since they present tissue and stage specific expression [40]; this gives them greater sensitivity and specificity [41], particularly in tumors with hormone sensitivity, such as the prostatic adenocarcinoma, in which some lncRNAs with clinical utility, such as SChLAP1 [42], lncRNA-p21 [43], and PCA3 [44], have been identified. As their association with prostate cancer has already been established, this allows for their use in clinical practice. For example, SChLAP1 is a lncRNA whose length is 854 nt, is transcribed from chromosome 2, and is differentially expressed in bladder normal tissue and prostate cancer tissue. It was first identified in paraffin-embedded tissue biopsies by in situ hybridization (ISH). The biological function of SChLAP1 is related to the regulation of the SWI/SNF chromatin-modifying complex; this lncRNA antagonizes the genome-wide localization of this protein complex, which is related to the promotion of invasiveness and metastasis in LNCaP and 22Rv1, as well as in Du145 cancer cell lines [45]. Moreover, SChLAP1 expression has also been associated with metastasis (odds ratio [OR] 2.45, 95% CI 1.70–3.53; p-value < 0·0001) and cancer progression (hazard ratio = 1.99, p-value = 0.032) in prostate cancer patients [46]. Additionally, lincRNA-p21, which is a lncRNA, has been shown to be differentially expressed in prostate cancer [47]; its biological function is principally the regulation of apoptosis, cell proliferation [48], and cell cycle by its interaction with MDM2 and STAT3 [47]. lincRNA-p21 has also been related to disease progression in prostate cancer in preclinical studies, as its overexpression in castration-resistant patients who were treated with enzalutamide is associated with less overall survival (p-value = 0.04), which indicates that lincRNA-p21 could also be a useful predictive biomarker for enzalutamide treatment [47]. Finally, the Prostate Cancer Antigen 3 (PCA3), a lncRNA of 3 Kb in length transcribed in chromosome 9, is present in prostate cancer with high tissue-expression specificity, described first by Bussemakers et al. in 1999 [49]. Currently, PCA3 is also an auxiliary biomarker in prostate cancer; its use was approved by the Food and Drug Administration (FDA) in 2012 [44] due to its clinical utility by reducing the number of unnecessary biopsies in patients. Additionally, PCA3 has been reported to be related to the survival of prostate tumor cells by regulating the androgen receptor signaling pathway, as well as regulating the epithelial-mesenchymal transition (EMT) by modulating some targets, such as E-cadherin and TWIST [50,51]. Furthermore, it has been used in gene signature PROGENSA to determine which patients with a previous negative biopsy [52] need a second biopsy [53]. As described above, the use of molecular biomarkers based on lncRNA expression for prostate cancer has demonstrated the utility of this RNA biotype in clinical practice. Likewise, this could be extended to breast cancer clinical application since both carcinomas are characterized as hormone-sensitive [54], and there is experimental evidence of lncRNA expression related with clinical outcome, such as lincRNA-ROR, in which PCA3 regulates EMT by modulating E-cadherin functions [55]. Thus, it is necessary to implement more research to have similar results in biomarker discovery for breast cancer.
Moreover, in prostate cancer research, it has been established that, although the lncRNA expression itself has clinical utility, the identification and detection of different biotypes, such as mRNAs, and genetic fusions also has utility in clinical practice [56]. Indeed, there are reports in scientific literature that demonstrate that the combination of lncRNA, mRNA, and genetic fusions in molecular signatures has improved the sensitivity or specificity of assays based priorly only in the expression of one gene [57]. One example is PROGENSA, which is based on PCA3 expression and is associated with a sensitivity of 66–72% and a specificity of 58–76% [58,59], while Mi Prostate Score, an urinary test based on the detection of PSA (mRNA), PCA3 (lncRNA), and TMPRSS2-ERG (genetic fusion) [60], has an associated sensitivity value of 95%. This demonstrates that the combinatorial use of mRNAs, lncRNAs, and genetic fusions can improve the results of laboratory tests for prostate cancer, and this could be extended to breast cancer research.

3.1. Challenges and Perspectives for lncRNA Clinical Application as Predictive Biomarkers for Breast Cancer Management

For breast cancer, there are few studies that support the use of lncRNAs or the combination with other biotypes as molecular predictive or prognostic biomarkers in clinical practice, and none of them have been approved for commercial distribution in prostate cancer, as in the case of PROGENSA, although there is already evidence in scientific literature about their potential as biomarkers in decision-making for the management of breast cancer patients [61,62,63]. The best example to describe the potential clinical utility of a lncRNA in patients with breast cancer is the study performed by Berger et al. in which the existence of lncRNA-coding gene regulation networks, such as NEAT1, TERC, and TUG1, together with other mRNAs, such as ESR1, AR, and SOX2, make it possible to classify patients with gynecological cancers and breast cancer into 6 clusters, which are related directly to their phenotypes and mainly to the immune response, as well as to the expression of hormone receptors in patients particularly associated with the estrogen receptor signaling pathway. This biomarker can be used for diagnostic, predictive, and prognostic purposes in breast cancer patients [64]. Furthermore, this has also been demonstrated by Niknafs et al., who described the use of DSCAM-AS1 expression as part of the characteristics of luminal tumors that are positive to hormone receptor expression [13]. It has also been described by Contreras-Espinosa et al. for GATA3-AS1 [17] and for the LINC01087, which expression profile is also related with luminal phenotypes in breast cancer [65]. This suggests that GATA3-AS1 expression may be a relevant molecular characteristic that defines luminal tumors [66]. However, there are additional emerging lncRNAs that have been described as potential biomarkers in cancer, such as HOTAIR, DSCAM-AS1, and GATA3-AS1 in breast cancer [8,13,17], MALAT1 in lung cancer [67], H19 in colorectal cancer [68], HULC in liver cancer [69], UCA1 in bladder cancer [70], and DLEU1 in endometrial cancer [71]. Among other lincRNAs [72], the applicability of lncRNAs in the molecular diagnostic area and their use in laboratory tests for clinical diagnosis in the near future largely depends on the expansion of knowledge about their association with different clinical variables, such as response to treatment and overall survival, as well as their inclusion in clinical trials in order to determine and validate the benefits of their use in clinical routine, as it has been made for coding genes before [73]. Thus, there are still many studies to be carried out in order to include lncRNAs more frequently in laboratory tests for the patient’s workup and treatment, not only in oncology, but also for other pathologies, like cardiovascular diseases [74] and diabetes [75], which are also leading causes of morbidity worldwide [76]. Taken together, these results suggest that lncRNAs may be relevant biomolecules that could allow oncologists to differentiate patients who do not respond to therapy, regardless of the molecular heterogeneity of breast tumors, which represents an important challenge in oncology practice [77].

3.2. The Use of lncRNAs as Molecular Biomarkers in the RNA-Based Therapeutics Era

A molecular feature advantage that distinguishes lncRNAs is their stability in biological samples, such as blood, urine, or saliva (median half-life ~3.5 h) [78]. This is due to their transport in exosomes, microvesicles, apoptotic bodies, high density lipoprotein capsules, or into circulating tumor cells [79], in contrast with mRNAs, which are characterized by their instability in body fluids (median half-life < 2 h) [80]. This allows the detection of lncRNAs by non-invasive techniques through the use of liquid biopsies, such as urine and saliva, and less-invasive methods, such as serum and plasma [40], as has been reported for lncRNAs HOTAIR [8] and H19 [9] in breast cancer, as well as for MALAT1, which has been shown to be a serological marker in breast cancer [36] and a diagnostic biomarker for oral squamous cell carcinoma that can be detected by saliva testing [81]. The detection of these lncRNA is performed by a quantitative polymerase chain reaction (qPCR) in RNA extracted from serum or saliva obtained from patients. Likewise, it is possible to detect lncRNAs with the use of other techniques with higher sensitivity, such as ISH-RNA, which has been used for the rapid detection of markers, such as HER2 in breast cancer, [82] with greater sensitivity and specificity (99% and 98%, respectively) when compared to HER2 immunohistochemistry (IHC) assay detection (95% and 98%, respectively) [83]. The ISH-RNA assay has also allowed the detection of lncRNA SNHG3 as a potential diagnostic biomarker, distinguishing between normal breast tissue and cancerous breast tissues [84]. Furthermore, there are novel molecular approaches, such as spatial transcriptomics, which allow for the identification of a signature based on 798 transcripts, including the lncRNA LINC00657, that could be implemented in machine learning methods to distinguish invasive breast cancer [62]. In summary, the implementation of molecular biology techniques for lncRNA-based biomarkers detection in clinical practice could improve the reliability of the results of laboratory tests and the accuracy of oncological diagnosis.
As discussed above, the implementation of PCA3, DSCAM-AS1, and GATA3-AS1 as other lncRNA molecular biomarkers represents a novel approach for the clinical management of the oncological patient (Figure 1) since their expression is tissue-specific, disease-specific, and is associated to a particular stability in body fluids [2], contributing to the development of precision medicine; this is because lncRNA-based biomarkers offer simple and reliable tests [85]. Altogether, this represents the lncRNA-based diagnostics [86,87], a new concept in medicine which integrates the potential use of lncRNAs as molecular biomarkers, with application in clinical practice, that will improve patient management in three main aspects. The first is the use of non-invasive techniques for laboratory tests (e.g., liquid biopsies); this has proven to be useful in clinical routines as the urine analysis, which is currently in practice with the use of PCA3 [88]. The implementation of these molecular assays, with fluids like urine and saliva, have the main objective of benefitting patient management because these methods allow the oncologists to perform the diagnostic and follow up of patients in less invasive manners, with the accuracy improved, due to the capability of these non-invasive methods to avoid some bias, like tumor cell heterogeneity [89]. Hence, the detection of lncRNAs by the implementation of non-invasive methods, such as urine and saliva analysis, is a promising improvement in clinical routine. Second, the use of time and cost-efficient detection techniques, such as qPCR, which take ~2 h to get results [90] in contrast to IHC, which takes approximately 2 days or more [91], will directly impact the optimization of the oncologist decision making, for example, in the decision for treatment selection for breast cancer patients. Third, the improvement in result accuracy for laboratory tests. Because of the high specific expression profile of lncRNAs, as well as their sensibility and specificity, differential diagnosis and early diagnosis are easier and can also be combined with pathological imaging processing that involves the use of X-ray imaging, magnetic resonance imaging, nuclear medicine imaging, and ultrasound imaging, which are techniques with routinary use in clinical practice [92]. Thus, the combinatorial use of molecular and image biomarkers could lead to the improvement in diagnosis, prediction, and prognosis values [93], which have been demonstrated by the implementation of machine learning algorithms for the integration of molecular imaging and clinical data [94,95,96]. However, to achieve the implementation of combinatorial biomarkers in breast cancer, the development of appropriate research protocols is necessary to demonstrate and validate their usefulness in clinical practice.

3.3. The Current Challenges for lncRNA Research and for Their Implementation as Molecular Biomarkers in Routine Clinical Practice

Finally, it should be noted that the principal objectives in the investigation of the use of lncRNAs as current molecular biomarkers in breast cancer have mostly been aimed at determining the biological function of lncRNA [100,101] or their ability to describe mammary tumors molecularly, as is the case of lncRNA EPIC1 described by Wang et al., which has been identified as an oncogene in breast cancer that promotes cell cycle and has been associated with poor overall survival (hazard ratio ~2, p-value = 0.005) [99]. However, it has not yet been possible to apply this knowledge in biomarker development for routine used in clinical practice, as it occurs with PCA3 in prostate cancer [44]. This happens because of three main reasons: (1) the sample size used for the discovery and validation of these potential biomarkers, (2) the lack of clinical trials focused on exploring the association of lncRNAs with clinical variables, and (3) the lack of clarity and accurate use of clinical definitions that involve biomarkers and clinical assessments, which make the development of new clinical tools based on novel molecular markers, such as lncRNAs, and their inclusion in clinical practice difficult [102]. However, their advantages over other biomolecules, such as proteins and mRNAs, have been demonstrated [40], as is the case of GATA3-AS1, which predicts neoadjuvant chemotherapy resistance in luminal B-like breast cancer patients with a sensitivity of 92% and specificity of 75% (p-value = 0.0001) [17] compared to Ki-67, a clinical biomarker for neoadjuvant chemotherapy response prediction in breast cancer (sensitivity: 95.7%, specificity: 54.3%, p-value  =  0.002) [103]. Another example is H19 [68,104] and DSCAM-AS1 [13,105]; not only has their biological function been described, but so has their applicability in clinical practice by establishing associations with clinical variables, such as estrogen receptor expression, which has potential application for diagnosis (sensitivity, 100.0%; specificity, 97.0%; p-value < 0.001), as well as predictive and prognostic features [105]. Furthermore, there are also studies for lncRNAs that are used as genetic signatures [4,14,106]. Wang et al. reported the use of a gene signature based on lncRNA expression that included NEAT1, and its predictive value for neoadjuvant chemotherapy response was described (sensitivity, 69.9%; specificity, 77.8%; p-value < 0.0001) [106]. However, the sample size in these studies is small and this has not allowed for the scale up of the applications of lncRNAs to the dimensions of a clinical trial, as is the case of H19 [9], HOTAIR [8], MALAT1 [36], and GATA3-AS1 [17], which have predictive value for neoadjuvant chemotherapy response. Furthermore, in general, there are few studies in the clinical setting that investigate the potential use of lncRNAs as diagnostic, predictive, or prognostic biomarkers in clinical practice for cancer (Supplementary Table S1) since there are only a few studies in preclinical stages and only three clinical trials that include lncRNAs as biomarkers developed specifically for breast cancer research (Table 1) and are not specifically dedicated to a particular lncRNA or do not yet have a publication related to lncRNA application [107,108,109]. This is despite the evidence for their roles as regulators of cell proliferation and survival in mammary tumors (Table 1), which represents a disadvantage in the development of lncRNA-based biomarkers in breast cancer, particularly in locally advanced stages, and makes their inclusion to molecular signatures harder, even if their utility could improve the sensibility and specificity of molecular biomarkers based on coding genes. This has been demonstrated for H19, which has a sensitivity of 56% in the identification of metastatic breast cancer [110] compared with CA-15-3, a tumor marker with a sensitivity of 42% in the same prognostic application [111]. This is evidence that lncRNA inclusion enhances gene signatures for clinical trials.
Practice, as is shown by Liu et al., with the comparison of 5-gene signature integrated by 4 mRNAs (OR7C1, TBX2, RSPH4A, and C2orf61) and the lncRNA AC10538, with 21-gene signature Oncotype Dx, in predicting overall survival in early-stage breast cancer patients showed that a 5-gene signature performed better in prognosis value than 21-gene in a TCGA cohort (areas under curves 0.807 and 0.572, respectively) [121]. This demonstrates that the integration of lncRNA to an mRNA signature could improve the prognosis value, as was also demonstrated for the combinatorial use of lncRNA MALAT1 and Oncotype Dx, for predicting the diagnosis of early-stage breast cancer [122].
Similar results have been reported for Acute Lymphoblastic Leukemia, in which a 7-gene signature, including one lncRNA, LINC00652, and six mRNAs, INSL3, NIPAL2, REN, RIMS2, RPRM, and SNAP91, had an area under the curve of 0.9 for predicting 5-year overall survival [123]. Moreover, Zhou and collaborators described a 12 lncRNA gene expression panel that is capable of predicting the risk of recurrence in breast cancer patients [124]. Another example is the study performed by Shen and collaborators, in which they demonstrated that a 11-lncRNA gene signature, that has an associated prognostic value (HR = 1.328 in univariate Cox regression analysis, with p-value < 0.001 and HR = 1.266 in multivariate Cox regression analysis, with p-value < 0.001), is also related with immune cell infiltration in breast tumors, which could have a clinical application for breast cancer immunotherapy [125]. Additional studies of the identification of molecular signature biomarkers based on lncRNA expression profiles are listed in Supplementary Table S2. This evidence suggests the utility of lncRNAs, in clinical practice, as molecular biomarkers of treatment response prediction and prognosis in cancer and their potential inclusion in already used molecular signatures, like Oncotype Dx [126]. The usefulness of lncRNAs in breast cancer, particularly for locally advanced stages, can therefore be more than just identifying biological properties and treatment response to systemic therapy [39], as is the case of HOTAIR, which is related to lymph node metastasis [10] and HUMT (p-value < 0.001) [127].
As well as the above examples, there are numerous examples of other lncRNAs that have shown potential as biomarkers for predicting the response to treatment in breast cancer in studies conducted in mouse animal models at the preclinical level (Table 2). However, as discussed above, efforts need to be directed to move this research to phase I clinical studies, not only for response prediction biomarkers, but also to biomarkers with diagnostic and prognostic applications in order to determine the benefits that they can bring to the management of oncology patients, including in other areas, such as surgery.
Additionally, in surgical practice, they could help in predicting lymph node burden and, therefore, avoid extensive axillary surgery in patients eligible for genomic tests [151], as it has been done recently with epigenetic signatures [152]. They could also work as biomarkers to predict objective response rates (ORR) to specific therapies in the neoadjuvant setting so tumor downstaging could be more precisely inferred [153]. In metastatic disease, these molecules can also help us to identify progression free survival in specific settings [154], such as oligometastatic disease in luminal-like tumors [155]. In the neoadjuvant setting, differential expression of lncRNAs could help to identify the probability of achieving low residual cancer burden (RCB) [156], and perhaps even its expression in residual disease might identify luminal-like patients that could benefit from adjuvant therapies if their absence/presence is associated with recurrence or death, since pathologic complete response is not an adequate biological surrogate of survival in this molecular subtype [157]. In addition, lncRNAs could have an impact on the development of new therapies based on RNA or RNA-based therapeutics [158] for breast cancer patients. Particularly, lncRNA could be incorporated for immunotherapy purposes [159], for example, as enhancers of a cancer vaccine’s response, as is the case of LINK-A, a lncRNA whose inhibition by locked nucleic acids synergistically suppresses tumor progression when it is combined with immune checkpoint blockers (or ICB therapy) [160], which have the same effect that the PVX-410 cancer vaccine [161] has. This in turn suggests that LINK-A could be a biomarker for PVX-410 vaccine response or a sensitizer for this vaccine [162]. Another example of the potential use of lncRNAs in cancer therapy is their use as targeted therapies for the treatment of metastatic cancer with the use of systems of release, such as the MALAT-1 antisense oligonucleotide, stabilized in antisense oligonucleotide-loaded nanostructure, co-functionalized in Au nanoparticles, which reduced metastatic tumors in vivo [163]. Altogether, these results show the potential applicability of lncRNA in different RNA-based therapies, including immunotherapy, which is one of the most promising perspectives for lncRNA therapeutics in cancer.

4. Conclusions

The trend of lncRNA research in breast cancer is based on the proposal of a new type of precision medicine that relies on the use of ncRNAs, since it is still in the dark matter of human genome [164] and since its application in the clinical area has not been explored deeply. This is due to the persistence of the exclusively and prioritized persistence of coding genes as biomarkers [165,166,167], although they continue to be of clinical utility when merging with gene signature tests, such as Oncotype Dx, Mammaprint, and Prosigna/PAM50 [168]. It has been shown that they do not represent the complete molecular description of mammary tumors [169], for which it is necessary to integrate the knowledge that research areas, such as genomics, transcriptomics, proteomics, epigenomics, metabolomics, and interactomics, have provided [170,171]. In the future, this will allow for the development of precision medicine for the benefit of cancer patients [97].
Thus, the new era of molecular diagnosis should consider the issues discussed above and, at least, allow for the reflection of three perspectives in its development. First, the integration of lncRNAs in clinical trials for the study and analysis of their potential applicability in clinical diagnosis. Second, the use of lncRNAs as diagnostic molecular biomarkers using non-invasive tests, as is the case for PCA3 detection in urine and for other promising lncRNAs, such as HOTAIR in serological tests and MALAT-1 in salivary tests, which represent an advance in the proper management of oncology patients, as we discussed before. Third, the integration of lncRNAs in commercial gene signatures and laboratory tests with diagnostic purposes, as was previously discussed, to improve the accuracy and reliability of diagnostic results, which will be reflected in the enhancement of oncology management strategies and in the amelioration of cancer patients’ quality of life. Finally, lncRNAs are part of the RNA world that have potential use as molecular biomarkers, which could be used in the near future as part of routine testing in breast cancer management.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms24087426/s1. References [172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192] are cited in the Supplementary Materials.

Author Contributions

Project development: C.A.-C. and L.A.H.; Clinical information: J.A.G.-G., P.C.-G., F.J.-T., E.B.-R. and S.A.-V.; Basic research information: L.C.-E., C.C.-H. and C.A.-C.; Figures: L.C.-E., C.A.-C. and L.A.H.; Manuscript writing: C.A.-C., L.C.-E., J.A.G.-G., S.A.-V., F.J.-T. and L.A.H.; Manuscript editing: C.A.-C. and L.A.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Consejo Nacional de Ciencia y Tecnología (CONACYT) grant A3-S-46689. L.C.-E. is a Ph.D. student in the Programa de Posgrado en Ciencias Biológicas, UNAM, and received a fellowship from CONACYT with Currículum Vitae Único (CVU)-1003211.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank the National Cancer Institute of Mexico (INCan), the National Institute of Genomic Medicine (INMEGEN), and the Programa de Posgrado en Ciencias Biológicas, UNAM for their support. This manuscript is part of the productivity of L.C.-E.’s studies aimed at obtaining a Ph.D. in Science in the field of Biomedicine. Figures were created with BioRender.com (accessed on 30 January 2023).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Ribonucleic acids (RNA); long non-coding RNAs (lncRNAs); prospective-specimen-collection, retrospective-blinded-evaluation (ProBE); RNA Sequencing (RNA-Seq); non-coding RNAs (ncRNAs); the Cancer Genome Atlas (TCGA); in situ hybridization (ISH); Prostate Cancer Antigen 3 (PCA3); Food and Drug Administration (FDA); messenger RNA (mRNA); quantitative polymerase chain reaction (qPCR); in situ hybridization of RNA (ISH-RNA); immunohistochemistry (IHC); identifier (ID); polymerase chain reaction (PCR); objective response rates (ORR); residual cancer burden (RCB).

References

  1. 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] [PubMed]
  2. Yan, X.; Hu, Z.; Feng, Y.; Hu, X.; Yuan, J.; Zhao, S.D.; Zhang, Y.; Yang, L.; Shan, W.; He, Q.; et al. Comprehensive Genomic Characterization of Long Non-Coding RNAs across Human Cancers. Cancer Cell 2015, 28, 529–540. [Google Scholar] [CrossRef] [PubMed]
  3. Bjørklund, S.S.; Aure, M.R.; Häkkinen, J.; Vallon-Christersson, J.; Kumar, S.; Evensen, K.B.; Fleischer, T.; Tost, J.; Sahlberg, K.K.; Mathelier, A.; et al. Subtype and Cell Type Specific Expression of LncRNAs Provide Insight into Breast Cancer. Commun. Biol. 2022, 5, 834. [Google Scholar] [CrossRef]
  4. Ferrando, L.; Cirmena, G.; Garuti, A.; Scabini, S.; Grillo, F.; Mastracci, L.; Isnaldi, E.; Marrone, C.; Gonella, R.; Murialdo, R.; et al. Development of a Long Non-Coding RNA Signature for Prediction of Response to Neoadjuvant Chemoradiotherapy in Locally Advanced Rectal Adenocarcinoma. PLoS ONE 2020, 15, e0226595. [Google Scholar] [CrossRef]
  5. Zhu, C.; Wang, X.; Wang, Y.; Wang, K. Functions and Underlying Mechanisms of LncRNA HOTAIR in Cancer Chemotherapy Resistance. Cell Death Discov. 2022, 8, 383. [Google Scholar] [CrossRef] [PubMed]
  6. Xue, X.; Yang, Y.A.; Zhang, A.; Fong, K.-W.; Kim, J.; Song, B.; Li, S.; Zhao, J.C.; Yu, J. LncRNA HOTAIR Enhances ER Signaling and Confers Tamoxifen Resistance in Breast Cancer. Oncogene 2016, 35, 2746–2755. [Google Scholar] [CrossRef]
  7. Alipoor, B.; Parvar, S.N.; Sabati, Z.; Ghaedi, H.; Ghasemi, H. An Updated Review of the H19 LncRNA in Human Cancer: Molecular Mechanism and Diagnostic and Therapeutic Importance. Mol. Biol. Rep. 2020, 47, 6357–6374. [Google Scholar] [CrossRef]
  8. Lu, R.; Zhang, J.; Zhang, W.; Huang, Y.; Wang, N.; Zhang, Q.; Qu, S. Circulating HOTAIR Expression Predicts the Clinical Response to Neoadjuvant Chemotherapy in Patients with Breast Cancer. Cancer Biomark. 2018, 22, 249–256. [Google Scholar] [CrossRef]
  9. Özgür, E.; Ferhatoğlu, F.; Şen, F.; Saip, P.; Gezer, U. Circulating LncRNA H19 May Be a Useful Marker of Response to Neoadjuvant Chemotherapy in Breast Cancer. Cancer Biomark. 2020, 27, 11–17. [Google Scholar] [CrossRef]
  10. 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]
  11. Ghafouri-Fard, S.; Khoshbakht, T.; Taheri, M.; Ebrahimzadeh, K. A Review on the Carcinogenic Roles of DSCAM-AS1. Front. Cell Dev. Biol. 2021, 9, 758513. [Google Scholar] [CrossRef] [PubMed]
  12. Elhasnaoui, J.; Miano, V.; Ferrero, G.; Doria, E.; Leon, A.E.; Fabricio, A.S.C.; Annaratone, L.; Castellano, I.; Sapino, A.; De Bortoli, M. DSCAM-AS1-Driven Proliferation of Breast Cancer Cells Involves Regulation of Alternative Exon Splicing and 3′-End Usage. Cancers 2020, 12, 1453. [Google Scholar] [CrossRef] [PubMed]
  13. Niknafs, Y.S.; Han, S.; Ma, T.; Speers, C.; Zhang, C.; Wilder-Romans, K.; Iyer, M.K.; Pitchiaya, S.; Malik, R.; Hosono, Y.; et al. The LncRNA Landscape of Breast Cancer Reveals a Role for DSCAM-AS1 in Breast Cancer Progression. Nat. Commun. 2016, 7, 12791. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, Y.; Wagner, E.K.; Guo, X.; May, I.; Cai, Q.; Zheng, W.; He, C.; Long, J. Long Intergenic Non-Coding RNA Expression Signature in Human Breast Cancer. Sci. Rep. 2016, 6, 37821. [Google Scholar] [CrossRef] [PubMed]
  15. Gibbons, H.R.; Spurlock, C.F.; Aune, T.M. A Long-Noncoding RNA, Gata3-AS1, Is a Positive Transcriptional Regulator of Transcription Factor Gata3 in TH2 Cells. J. Immunol. 2017, 198, 150.11. [Google Scholar] [CrossRef]
  16. Gibbons, H.R.; Shaginurova, G.; Kim, L.C.; Chapman, N.; Spurlock, C.F.I.; Aune, T.M. Divergent LncRNA GATA3-AS1 Regulates GATA3 Transcription in T-Helper 2 Cells. Front. Immunol. 2018, 9, 2512. [Google Scholar] [CrossRef]
  17. Contreras-Espinosa, L.; Alcaraz, N.; De La Rosa-Velázquez, I.A.; Díaz-Chávez, J.; Cabrera-Galeana, P.; Vega, R.R.; Reynoso-Noveron, N.; Maldonado-Martínez, H.A.; González-Barrios, R.; Montiel-Manríquez, R.; et al. Transcriptome Analysis Identifies GATA3-AS1 as a Long Noncoding RNA Associated with Resistance to Neoadjuvant Chemotherapy in Locally Advanced Breast Cancer Patients. J. Mol. Diagn. 2021, 23, 1306–1323. [Google Scholar] [CrossRef]
  18. Zhang, M.; Wang, N.; Song, P.; Fu, Y.; Ren, Y.; Li, Z.; Wang, J. LncRNA GATA3-AS1 Facilitates Tumour Progression and Immune Escape in Triple-Negative Breast Cancer through Destabilization of GATA3 but Stabilization of PD-L1. Cell Prolif. 2020, 53, e12855. [Google Scholar] [CrossRef]
  19. Lyons, T.G. Targeted Therapies for Triple-Negative Breast Cancer. Curr. Treat. Options Oncol. 2019, 20, 82. [Google Scholar] [CrossRef]
  20. Pepe, M.S.; Li, C.I.; Feng, Z. Improving the Quality of Biomarker Discovery Research: The Right Samples and Enough of Them. Cancer Epidemiol. Biomark. Prev. 2015, 24, 944–950. [Google Scholar] [CrossRef] [PubMed]
  21. Guo, Z.-H.; Yao, L.-T.; Guo, A.-Y. Clinical and Biological Impact of LINC02544 Expression in Breast Cancer after Neoadjuvant Chemotherapy. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 10573–10585. [Google Scholar] [CrossRef] [PubMed]
  22. Cedro-Tanda, A.; Ríos-Romero, M.; Romero-Córdoba, S.; Cisneros-Villanueva, M.; Rebollar-Vega, R.G.; Alfaro-Ruiz, L.A.; Jiménez-Morales, S.; Domínguez-Reyes, C.; Villegas-Carlos, F.; Tenorio-Torres, A.; et al. A LncRNA Landscape in Breast Cancer Reveals a Potential Role for AC009283.1 in Proliferation and Apoptosis in HER2-Enriched Subtype. Sci. Rep. 2020, 10, 13146. [Google Scholar] [CrossRef] [PubMed]
  23. Pepe, M.S.; Feng, Z.; Janes, H.; Bossuyt, P.M.; Potter, J.D. Pivotal Evaluation of the Accuracy of a Biomarker Used for Classification or Prediction: Standards for Study Design. J. Natl. Cancer. Inst. 2008, 100, 1432–1438. [Google Scholar] [CrossRef] [PubMed]
  24. Gadbury, G.L.; Page, G.P.; Edwards, J.; Kayo, T.; Prolla, T.A.; Weindruch, R.; Permana, P.A.; Mountz, J.D.; Allison, D.B. Power and Sample Size Estimation in High Dimensional Biology. Stat. Methods Med. Res. 2004, 13, 325–338. [Google Scholar] [CrossRef]
  25. Auer, P.L.; Doerge, R.W. Statistical Design and Analysis of RNA Sequencing Data. Genetics 2010, 185, 405–416. [Google Scholar] [CrossRef]
  26. Corchete, L.A.; Rojas, E.A.; Alonso-López, D.; De Las Rivas, J.; Gutiérrez, N.C.; Burguillo, F.J. Systematic Comparison and Assessment of RNA-Seq Procedures for Gene Expression Quantitative Analysis. Sci. Rep. 2020, 10, 19737. [Google Scholar] [CrossRef]
  27. Stark, R.; Grzelak, M.; Hadfield, J. RNA Sequencing: The Teenage Years. Nat. Rev. Genet. 2019, 20, 631–656. [Google Scholar] [CrossRef]
  28. Micheloud, C.; Held, L. Power Calculations for Replication Studies. Stat. Sci. 2022, 37, 369–379. [Google Scholar] [CrossRef]
  29. Mishra, P.; Pandey, C.M.; Singh, U.; Gupta, A.; Sahu, C.; Keshri, A. Descriptive Statistics and Normality Tests for Statistical Data. Ann. Card. Anaesth. 2019, 22, 67–72. [Google Scholar] [CrossRef]
  30. Savani, B.N.; Barrett, A.J. CHAPTER 49-How to Build and Use a Stem Cell Transplant Database. In Hematopoietic Stem Cell Transplantation in Clinical Practice; Treleaven, J., Barrett, A.J., Eds.; Churchill Livingstone: Edinburgh, UK, 2009; pp. 505–512. ISBN 978-0-443-10147-2. [Google Scholar]
  31. Rosner, B.A. Fundamentals of Biostatistics; Thomson-Brooks/Cole: Boston, MA, USA, 2006; ISBN 978-0-534-41820-5. [Google Scholar]
  32. Fang, Z.; Cui, X. Design and Validation Issues in RNA-Seq Experiments. Brief. Bioinform. 2011, 12, 280–287. [Google Scholar] [CrossRef]
  33. Negida, A.; Fahim, N.K.; Negida, Y. Sample Size Calculation Guide-Part 4: How to Calculate the Sample Size for a Diagnostic Test Accuracy Study Based on Sensitivity, Specificity, and the Area Under the ROC Curve. Adv. J. Emerg. Med. 2019, 3, e33. [Google Scholar] [CrossRef]
  34. Hajian-Tilaki, K.O.; Hanley, J.A.; Joseph, L.; Collet, J.-P. A Comparison of Parametric and Nonparametric Approaches to ROC Analysis of Quantitative Diagnostic Tests. Med. Decis. Mak. 1997, 17, 94–102. [Google Scholar] [CrossRef] [PubMed]
  35. Ray, P.; Manach, Y.L.; Riou, B.; Houle, T.T.; Warner, D.S. Statistical Evaluation of a Biomarker. Anesthesiology 2010, 112, 1023–1040. [Google Scholar] [CrossRef]
  36. Zidan, H.E.; Karam, R.A.; El-Seifi, O.S.; Abd Elrahman, T.M. Circulating Long Non-Coding RNA MALAT1 Expression as Molecular Biomarker in Egyptian Patients with Breast Cancer. Cancer Genet. 2018, 220, 32–37. [Google Scholar] [CrossRef]
  37. Tang, Z.; Li, C.; Kang, B.; Gao, G.; Li, C.; Zhang, Z. GEPIA: A Web Server for Cancer and Normal Gene Expression Profiling and Interactive Analyses. Nucleic Acids Res. 2017, 45, W98–W102. [Google Scholar] [CrossRef]
  38. Li, J.; Han, L.; Roebuck, P.; Diao, L.; Liu, L.; Yuan, Y.; Weinstein, J.N.; Liang, H. TANRIC: An Interactive Open Platform to Explore the Function of LncRNAs in Cancer. Cancer Res. 2015, 75, 3728–3737. [Google Scholar] [CrossRef] [PubMed]
  39. Jin, H.; Du, W.; Huang, W.; Yan, J.; Tang, Q.; Chen, Y.; Zou, Z. LncRNA and Breast Cancer: Progress from Identifying Mechanisms to Challenges and Opportunities of Clinical Treatment. Mol. Ther. Nucleic Acids 2021, 25, 613–637. [Google Scholar] [CrossRef] [PubMed]
  40. Xi, X.; Li, T.; Huang, Y.; Sun, J.; Zhu, Y.; Yang, Y.; Lu, Z.J. RNA Biomarkers: Frontier of Precision Medicine for Cancer. Non-Coding RNA 2017, 3, 9. [Google Scholar] [CrossRef] [PubMed]
  41. Jiang, S.; Cheng, S.-J.; Ren, L.-C.; Wang, Q.; Kang, Y.-J.; Ding, Y.; Hou, M.; Yang, X.-X.; Lin, Y.; Liang, N.; et al. An Expanded Landscape of Human Long Noncoding RNA. Nucleic Acids Res. 2019, 47, 7842–7856. [Google Scholar] [CrossRef]
  42. Mehra, R.; Udager, A.M.; Ahearn, T.U.; Cao, X.; Feng, F.Y.; Loda, M.; Petimar, J.S.; Kantoff, P.; Mucci, L.A.; Chinnaiyan, A.M. Overexpression of the Long Non-Coding RNA SChLAP1 Independently Predicts Lethal Prostate Cancer. Eur. Urol. 2016, 70, 549–552. [Google Scholar] [CrossRef]
  43. Işın, M.; Uysaler, E.; Özgür, E.; Köseoğlu, H.; Şanlı, Ö.; Yücel, Ö.B.; Gezer, U.; Dalay, N. Exosomal LncRNA-P21 Levels May Help to Distinguish Prostate Cancer from Benign Disease. Front. Genet. 2015, 6, 168. [Google Scholar] [CrossRef] [PubMed]
  44. Lemos, A.E.G.; Matos, A.D.R.; Ferreira, L.B.; Gimba, E.R.P. The Long Non-Coding RNA PCA3: An Update of Its Functions and Clinical Applications as a Biomarker in Prostate Cancer. Oncotarget 2019, 10, 6589–6603. [Google Scholar] [CrossRef] [PubMed]
  45. Prensner, J.R.; Iyer, M.K.; Sahu, A.; Asangani, I.A.; Cao, Q.; Patel, L.; Vergara, I.A.; Davicioni, E.; Erho, N.; Ghadessi, M.; et al. The Long Noncoding RNA SChLAP1 Promotes Aggressive Prostate Cancer and Antagonizes the SWI/SNF Complex. Nat. Genet. 2013, 45, 1392–1398. [Google Scholar] [CrossRef]
  46. Mehra, R.; Shi, Y.; Udager, A.M.; Prensner, J.R.; Sahu, A.; Iyer, M.K.; Siddiqui, J.; Cao, X.; Wei, J.; Jiang, H.; et al. A Novel RNA in Situ Hybridization Assay for the Long Noncoding RNA SChLAP1 Predicts Poor Clinical Outcome after Radical Prostatectomy in Clinically Localized Prostate Cancer. Neoplasia 2014, 16, 1121–1127. [Google Scholar] [CrossRef] [PubMed]
  47. Luo, J.; Wang, K.; Yeh, S.; Sun, Y.; Liang, L.; Xiao, Y.; Xu, W.; Niu, Y.; Cheng, L.; Maity, S.N.; et al. LncRNA-P21 Alters the Antiandrogen Enzalutamide-Induced Prostate Cancer Neuroendocrine Differentiation via Modulating the EZH2/STAT3 Signaling. Nat. Commun. 2019, 10, 2571. [Google Scholar] [CrossRef] [PubMed]
  48. Amirinejad, R.; Rezaei, M.; Shirvani-Farsani, Z. An Update on Long Intergenic Noncoding RNA P21: A Regulatory Molecule with Various Significant Functions in Cancer. Cell Biosci. 2020, 10, 82. [Google Scholar] [CrossRef]
  49. Bussemakers, M.J.; van Bokhoven, A.; Verhaegh, G.W.; Smit, F.P.; Karthaus, H.F.; Schalken, J.A.; Debruyne, F.M.; Ru, N.; Isaacs, W.B. DD3: A New Prostate-Specific Gene, Highly Overexpressed in Prostate Cancer. Cancer Res. 1999, 59, 5975–5979. [Google Scholar]
  50. Xue, W.-J.; Ying, X.-L.; Jiang, J.-H.; Xu, Y.-H. Prostate Cancer Antigen 3 as a Biomarker in the Urine for Prostate Cancer Diagnosis: A Meta-Analysis. J. Cancer Res. Ther. 2014, 10 (Suppl. S3), C218–C221. [Google Scholar] [CrossRef]
  51. Jiang, G.; Su, Z.; Liang, X.; Huang, Y.; Lan, Z.; Jiang, X. Long Non-Coding RNAs in Prostate Tumorigenesis and Therapy (Review). Mol. Clin. Oncol. 2020, 13, 76. [Google Scholar] [CrossRef]
  52. Reid, J.; Gen-Probe, Incorporated. Clinical Evaluation of the PROGENSA(R) PCA3 Assay in Men with a Previous Negative Biopsy Result; Clinical Trial ID: NCT01024959. 2012. Available online: https://beta.clinicaltrials.gov/study/NCT01024959?distance=50&term=NCT01024959&rank=1 (accessed on 22 February 2023).
  53. Durand, X.; Moutereau, S.; Xylinas, E.; de la Taille, A. ProgensaTM PCA3 Test for Prostate Cancer. Expert Rev. Mol. Diagn. 2011, 11, 137–144. [Google Scholar] [CrossRef]
  54. Chaput, G.; Sumar, N. Endocrine Therapies for Breast and Prostate Cancers. Can. Fam. Physician 2022, 68, 271–276. [Google Scholar] [CrossRef] [PubMed]
  55. Hou, P.; Zhao, Y.; Li, Z.; Yao, R.; Ma, M.; Gao, Y.; Zhao, L.; Zhang, Y.; Huang, B.; Lu, J. LincRNA-ROR Induces Epithelial-to-Mesenchymal Transition and Contributes to Breast Cancer Tumorigenesis and Metastasis. Cell Death Dis. 2014, 5, e1287. [Google Scholar] [CrossRef] [PubMed]
  56. Udager, A.M.; Tomlins, S.A. Molecular Biomarkers in the Clinical Management of Prostate Cancer. Cold Spring Harb. Perspect. Med. 2018, 8, a030601. [Google Scholar] [CrossRef]
  57. Alarcón-Zendejas, A.P.; Scavuzzo, A.; Jiménez-Ríos, M.A.; Álvarez-Gómez, R.M.; Montiel-Manríquez, R.; Castro-Hernández, C.; Jiménez-Dávila, M.A.; Pérez-Montiel, D.; González-Barrios, R.; Jiménez-Trejo, F.; et al. The Promising Role of New Molecular Biomarkers in Prostate Cancer: From Coding and Non-Coding Genes to Artificial Intelligence Approaches. Prostate Cancer Prostatic Dis. 2022, 25, 431–443. [Google Scholar] [CrossRef] [PubMed]
  58. Ramos, C.G.; Valdevenito, R.; Vergara, I.; Anabalon, P.; Sanchez, C.; Fulla, J. PCA3 Sensitivity and Specificity for Prostate Cancer Detection in Patients with Abnormal PSA and/or Suspicious Digital Rectal Examination. First Latin American Experience. Urol. Oncol. 2013, 31, 1522–1526. [Google Scholar] [CrossRef]
  59. van Gils, M.P.M.Q.; Hessels, D.; van Hooij, O.; Jannink, S.A.; Peelen, W.P.; Hanssen, S.L.J.; Witjes, J.A.; Cornel, E.B.; Karthaus, H.F.M.; Smits, G.A.H.J.; et al. The Time-Resolved Fluorescence-Based PCA3 Test on Urinary Sediments after Digital Rectal Examination; a Dutch Multicenter Validation of the Diagnostic Performance. Clin. Cancer Res. 2007, 13, 939–943. [Google Scholar] [CrossRef]
  60. Tosoian, J.J.; Trock, B.J.; Morgan, T.M.; Salami, S.S.; Tomlins, S.A.; Spratt, D.E.; Siddiqui, J.; Kunju, L.P.; Botbyl, R.; Chopra, Z.; et al. Use of the MyProstateScore Test to Rule Out Clinically Significant Cancer: Validation of a Straightforward Clinical Testing Approach. J. Urol. 2021, 205, 732–739. [Google Scholar] [CrossRef]
  61. Guo, W.; Wang, Q.; Zhan, Y.; Chen, X.; Yu, Q.; Zhang, J.; Wang, Y.; Xu, X.; Zhu, L. Transcriptome Sequencing Uncovers a Three–Long Noncoding RNA Signature in Predicting Breast Cancer Survival. Sci. Rep. 2016, 6, 27931. [Google Scholar] [CrossRef]
  62. Yoosuf, N.; Navarro, J.F.; Salmén, F.; Ståhl, P.L.; Daub, C.O. Identification and Transfer of Spatial Transcriptomics Signatures for Cancer Diagnosis. Breast Cancer Res. 2020, 22, 6. [Google Scholar] [CrossRef]
  63. Fan, C.-N.; Ma, L.; Liu, N. Systematic Analysis of LncRNA-MiRNA-MRNA Competing Endogenous RNA Network Identifies Four-LncRNA Signature as a Prognostic Biomarker for Breast Cancer. J. Transl. Med. 2018, 16, 264. [Google Scholar] [CrossRef]
  64. Berger, A.C.; Korkut, A.; Kanchi, R.S.; Hegde, A.M.; Lenoir, W.; Liu, W.; Liu, Y.; Fan, H.; Shen, H.; Ravikumar, V.; et al. A Comprehensive Pan-Cancer Molecular Study of Gynecologic and Breast Cancers. Cancer Cell 2018, 33, 690–705.e9. [Google Scholar] [CrossRef]
  65. De Palma, F.D.E.; Del Monaco, V.; Pol, J.G.; Kremer, M.; D’Argenio, V.; Stoll, G.; Montanaro, D.; Uszczyńska-Ratajczak, B.; Klein, C.C.; Vlasova, A.; et al. The Abundance of the Long Intergenic Non-Coding RNA 01087 Differentiates between Luminal and Triple-Negative Breast Cancers and Predicts Patient Outcome. Pharmacol. Res. 2020, 161, 105249. [Google Scholar] [CrossRef] [PubMed]
  66. Jézéquel, P.; Gouraud, W.; Ben Azzouz, F.; Guérin-Charbonnel, C.; Juin, P.P.; Lasla, H.; Campone, M. Bc-GenExMiner 4.5: New Mining Module Computes Breast Cancer Differential Gene Expression Analyses. Database 2021, 2021, baab007. [Google Scholar] [CrossRef]
  67. Pan, J.; Bian, Y.; Cao, Z.; Lei, L.; Pan, J.; Huang, J.; Cai, X.; Lan, X.; Zheng, H. Long Noncoding RNA MALAT1 as a Candidate Serological Biomarker for the Diagnosis of Non-small Cell Lung Cancer: A Meta-analysis. Thorac. Cancer 2020, 11, 329–335. [Google Scholar] [CrossRef] [PubMed]
  68. Han, D.; Gao, X.; Wang, M.; Qiao, Y.; Xu, Y.; Yang, J.; Dong, N.; He, J.; Sun, Q.; Lv, G.; et al. Long Noncoding RNA H19 Indicates a Poor Prognosis of Colorectal Cancer and Promotes Tumor Growth by Recruiting and Binding to EIF4A3. Oncotarget 2016, 7, 22159–22173. [Google Scholar] [CrossRef] [PubMed]
  69. Xie, H.; Ma, H.; Zhou, D. Plasma HULC as a Promising Novel Biomarker for the Detection of Hepatocellular Carcinoma. Biomed. Res. Int. 2013, 2013, 136106. [Google Scholar] [CrossRef]
  70. Fan, Y.; Shen, B.; Tan, M.; Mu, X.; Qin, Y.; Zhang, F.; Liu, Y. Long Non-Coding RNA UCA1 Increases Chemoresistance of Bladder Cancer Cells by Regulating Wnt Signaling. FEBS J. 2014, 281, 1750–1758. [Google Scholar] [CrossRef]
  71. Shan, L.; Zhao, T.; Wang, Y. Upregulation of Serum LncRNA DLEU1 Predicts Progression of Premalignant Endometrial Lesion and Unfavorable Clinical Outcome of Endometrial Cancer. Technol. Cancer Res. Treat 2020, 19, 1533033820965589. [Google Scholar] [CrossRef]
  72. Bhan, A.; Soleimani, M.; Mandal, S.S. Long Noncoding RNA and Cancer: A New Paradigm. Cancer Res. 2017, 77, 3965–3981. [Google Scholar] [CrossRef]
  73. Selleck, M.J.; Senthil, M.; Wall, N.R. Making Meaningful Clinical Use of Biomarkers. Biomark. Insights 2017, 12, 1177271917715236. [Google Scholar] [CrossRef]
  74. Matkovich, S.J.; Edwards, J.R.; Grossenheider, T.C.; Strong, C.D.G.; Dorn, G.W. Epigenetic Coordination of Embryonic Heart Transcription by Dynamically Regulated Long Noncoding RNAs. Proc. Natl. Acad. Sci. USA 2014, 111, 12264–12269. [Google Scholar] [CrossRef]
  75. Sathishkumar, C.; Prabu, P.; Mohan, V.; Balasubramanyam, M. Linking a Role of LncRNAs (Long Non-Coding RNAs) with Insulin Resistance, Accelerated Senescence, and Inflammation in Patients with Type 2 Diabetes. Hum. Genom. 2018, 12, 41. [Google Scholar] [CrossRef] [PubMed]
  76. World Health Organization. World Health Statistics 2021: Monitoring Health for the SDGs, Sustainable Development Goals; World Health Organization: Geneva, Switzerland, 2021. [Google Scholar]
  77. Harbeck, N.; Penault-Llorca, F.; Cortes, J.; Gnant, M.; Houssami, N.; Poortmans, P.; Ruddy, K.; Tsang, J.; Cardoso, F. Breast Cancer. Nat. Rev. Dis. Prim. 2019, 5, 66. [Google Scholar] [CrossRef] [PubMed]
  78. Galamb, O.; Barták, B.K.; Kalmár, A.; Nagy, Z.B.; Szigeti, K.A.; Tulassay, Z.; Igaz, P.; Molnár, B. Diagnostic and Prognostic Potential of Tissue and Circulating Long Non-Coding RNAs in Colorectal Tumors. World J. Gastroenterol. 2019, 25, 5026–5048. [Google Scholar] [CrossRef] [PubMed]
  79. Shi, T.; Gao, G.; Cao, Y. Long Noncoding RNAs as Novel Biomarkers Have a Promising Future in Cancer Diagnostics. Dis. Markers 2016, 2016, e9085195. [Google Scholar] [CrossRef] [PubMed]
  80. Yang, E.; van Nimwegen, E.; Zavolan, M.; Rajewsky, N.; Schroeder, M.; Magnasco, M.; Darnell, J.E. Decay Rates of Human MRNAs: Correlation with Functional Characteristics and Sequence Attributes. Genome Res. 2003, 13, 1863–1872. [Google Scholar] [CrossRef]
  81. Tang, H.; Wu, Z.; Zhang, J.; Su, B. Salivary LncRNA as a Potential Marker for Oral Squamous Cell Carcinoma Diagnosis. Mol. Med. Rep. 2013, 7, 761–766. [Google Scholar] [CrossRef] [PubMed]
  82. Alba, J.; Gutierrez, J.; Coupe, V.M.; Fernández, B.; Vázquez-Boquete, Á.; Alba, J.; Forteza, J.; García-Caballero, T. HER2 Status Determination Using RNA-ISH--a Rapid and Simple Technique Showing High Correlation with FISH and IHC in 141 Cases of Breast Cancer. Histol. Histopathol. 2012, 27, 1021–1027. [Google Scholar] [CrossRef] [PubMed]
  83. Arnould, L.; Roger, P.; MacGrogan, G.; Chenard, M.-P.; Balaton, A.; Beauclair, S.; Penault-Llorca, F. Accuracy of HER2 Status Determination on Breast Core-Needle Biopsies (Immunohistochemistry, FISH, CISH and SISH vs. FISH). Mod. Pathol. 2012, 25, 675–682. [Google Scholar] [CrossRef]
  84. Jiang, H.; Li, X.; Wang, W.; Dong, H. Long Non-Coding RNA SNHG3 Promotes Breast Cancer Cell Proliferation and Metastasis by Binding to MicroRNA-154-3p and Activating the Notch Signaling Pathway. BMC Cancer 2020, 20, 838. [Google Scholar] [CrossRef]
  85. Ginsburg, G.S.; Phillips, K.A. Precision Medicine: From Science to Value. Health Aff. (Millwood) 2018, 37, 694–701. [Google Scholar] [CrossRef] [PubMed]
  86. Hu, Q.; Ma, H.; Chen, H.; Zhang, Z.; Xue, Q. LncRNA in Tumorigenesis of Non-Small-Cell Lung Cancer: From Bench to Bedside. Cell Death Discov. 2022, 8, 359. [Google Scholar] [CrossRef] [PubMed]
  87. Dey Ghosh, R.; Guha Majumder, S. Circulating Long Non-Coding RNAs Could Be the Potential Prognostic Biomarker for Liquid Biopsy for the Clinical Management of Oral Squamous Cell Carcinoma. Cancers 2022, 14, 5590. [Google Scholar] [CrossRef]
  88. Hessels, D.; Klein Gunnewiek, J.M.T.; van Oort, I.; Karthaus, H.F.M.; van Leenders, G.J.L.; van Balken, B.; Kiemeney, L.A.; Witjes, J.A.; Schalken, J.A. DD3(PCA3)-Based Molecular Urine Analysis for the Diagnosis of Prostate Cancer. Eur. Urol. 2003, 44, 8–15; discussion 15–16. [Google Scholar] [CrossRef]
  89. Li, J.; Guan, X.; Fan, Z.; Ching, L.-M.; Li, Y.; Wang, X.; Cao, W.-M.; Liu, D.-X. Non-Invasive Biomarkers for Early Detection of Breast Cancer. Cancers 2020, 12, 2767. [Google Scholar] [CrossRef]
  90. Rogers-Broadway, K.-R.; Karteris, E. Amplification Efficiency and Thermal Stability of QPCR Instrumentation: Current Landscape and Future Perspectives. Exp. Ther. Med. 2015, 10, 1261–1264. [Google Scholar] [CrossRef] [PubMed]
  91. Reasons for Delays in Getting Your Biopsy and Cytology Test Results. Available online: https://www.cancer.org/treatment/understanding-your-diagnosis/tests/testing-biopsy-and-cytology-specimens-for-cancer/how-long-does-testing-take.html (accessed on 10 December 2021).
  92. Zhang, Y.; XIA, K.; LI, C.; WEI, B.; Zhang, B. Review of Breast Cancer Pathologigcal Image Processing. Biomed. Res. Int. 2021, 2021, 1994764. [Google Scholar] [CrossRef] [PubMed]
  93. Gamble, P.; Jaroensri, R.; Wang, H.; Tan, F.; Moran, M.; Brown, T.; Flament-Auvigne, I.; Rakha, E.A.; Toss, M.; Dabbs, D.J.; et al. Determining Breast Cancer Biomarker Status and Associated Morphological Features Using Deep Learning. Commun. Med. 2021, 1, 14. [Google Scholar] [CrossRef] [PubMed]
  94. Lobato-Delgado, B.; Priego-Torres, B.; Sanchez-Morillo, D. Combining Molecular, Imaging, and Clinical Data Analysis for Predicting Cancer Prognosis. Cancers 2022, 14, 3215. [Google Scholar] [CrossRef]
  95. Li, Y.; Wu, F.-X.; Ngom, A. A Review on Machine Learning Principles for Multi-View Biological Data Integration. Brief. Bioinform. 2016, 19, 325–340. [Google Scholar] [CrossRef]
  96. Ektefaie, Y.; Yuan, W.; Dillon, D.A.; Lin, N.U.; Golden, J.A.; Kohane, I.S.; Yu, K.-H. Integrative Multiomics-Histopathology Analysis for Breast Cancer Classification. Npj Breast Cancer 2021, 7, 147. [Google Scholar] [CrossRef]
  97. Ou, F.-S.; Michiels, S.; Shyr, Y.; Adjei, A.A.; Oberg, A.L. Biomarker Discovery and Validation: Statistical Considerations. J. Thorac. Oncol. 2021, 16, 537–545. [Google Scholar] [CrossRef]
  98. Ren, S.; Peng, Z.; Mao, J.-H.; Yu, Y.; Yin, C.; Gao, X.; Cui, Z.; Zhang, J.; Yi, K.; Xu, W.; et al. RNA-Seq Analysis of Prostate Cancer in the Chinese Population Identifies Recurrent Gene Fusions, Cancer-Associated Long Noncoding RNAs and Aberrant Alternative Splicings. Cell Res. 2012, 22, 806–821. [Google Scholar] [CrossRef] [PubMed]
  99. Wang, Z.; Yang, B.; Zhang, M.; Guo, W.; Wu, Z.; Wang, Y.; Jia, L.; Li, S.; Cancer Genome Atlas Research Network; Xie, W.; et al. lncRNA Epigenetic Landscape Analysis Identifies EPIC1 as an Oncogenic lncRNA that Interacts with MYC and Promotes Cell-Cycle Progression in Cancer. Cancer Cell 2018, 33, 706–720.e9. [Google Scholar] [CrossRef] [PubMed]
  100. 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] [PubMed]
  101. Huang, X.-J.; Xia, Y.; He, G.-F.; Zheng, L.-L.; Cai, Y.-P.; Yin, Y.; Wu, Q. MALAT1 Promotes Angiogenesis of Breast Cancer. Oncol. Rep. 2018, 40, 2683–2689. [Google Scholar] [CrossRef] [PubMed]
  102. FDA-NIH Biomarker Working Group. BEST (Biomarkers, EndpointS, and Other Tools) Resource; National Institutes of Health:: Bethesda, MD, USA; Food and Drug Administration: Silver Spring, MD, USA, 2006. [Google Scholar]
  103. Ács, B.; Zámbó, V.; Vízkeleti, L.; Szász, A.M.; Madaras, L.; Szentmártoni, G.; Tőkés, T.; Molnár, B.Á.; Molnár, I.A.; Vári-Kakas, S.; et al. Ki-67 as a Controversial Predictive and Prognostic Marker in Breast Cancer Patients Treated with Neoadjuvant Chemotherapy. Diagn. Pathol. 2017, 12, 20. [Google Scholar] [CrossRef] [PubMed]
  104. Gao, H.; Hao, G.; Sun, Y.; Li, L.; Wang, Y. Long Noncoding RNA H19 Mediated the Chemosensitivity of Breast Cancer Cells via Wnt Pathway and EMT Process. Onco. Targets Ther. 2018, 11, 8001–8012. [Google Scholar] [CrossRef] [PubMed]
  105. Moradi, M.-T.; Fallahi, H.; Rahimi, Z. The Clinical Significance of Circulating DSCAM-AS1 in Patients with ER-Positive Breast Cancer and Construction of Its Competitive Endogenous RNA Network. Mol. Biol. Rep. 2020, 47, 7685–7697. [Google Scholar] [CrossRef]
  106. Wang, G.; Chen, X.; Liang, Y.; Wang, W.; Shen, K. A Long Noncoding RNA Signature That Predicts Pathological Complete Remission Rate Sensitively in Neoadjuvant Treatment of Breast Cancer. Transl. Oncol. 2017, 10, 988–997. [Google Scholar] [CrossRef]
  107. TA(E)C-GP Versus A(E)C-T for the High Risk TNBC Patients and Validation of the MRNA-LncRNA Signature-No Study Results Posted-ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/results/NCT02641847 (accessed on 16 September 2021).
  108. Vogin, G.; Behm-Ansmant, I. Institut de Cancérologie de Lorraine Study of RNA and Heat Shock Protein (HSP) Derived Biomarkers in Radiation-Induced Fibrosis in Patients Treated for Breast Cancer.; Clinical Trial ID: NCT03000764. 2018. Available online: https://beta.clinicaltrials.gov/study/NCT03000764?distance=50&term=NCT03000764&rank=1 (accessed on 22 February 2023).
  109. Lu, J. Weekly Paclitaxel and Cisplatin to Treat Hormone Receptor Positive and Triple Negative Breast Cancer Patients (SHPD002); Clinical Trial ID: NCT02221999. 2019. Available online: https://beta.clinicaltrials.gov/study/NCT02221999?distance=50&term=NCT02221999&rank=1 (accessed on 22 February 2023).
  110. Zhang, K.; Luo, Z.; Zhang, Y.; Zhang, L.; Wu, L.; Liu, L.; Yang, J.; Song, X.; Liu, J. Circulating LncRNA H19 in Plasma as a Novel Biomarker for Breast Cancer. Cancer Biomark. 2016, 17, 187–194. [Google Scholar] [CrossRef] [PubMed]
  111. Lumachi, F.; Brandes, A.A.; Ermani, M.; Bruno, G.; Boccagni, P. Sensitivity of Serum Tumor Markers CEA and CA 15-3 in Breast Cancer Recurrences and Correlation with Different Prognostic Factors. Anticancer Res. 2000, 20, 4751–4755. [Google Scholar]
  112. Ma, T.; Liang, Y.; Li, Y.; Song, X.; Zhang, N.; Li, X.; Chen, B.; Zhao, W.; Wang, L.; Yang, Q. LncRNA LINP1 Confers Tamoxifen Resistance and Negatively Regulated by ER Signaling in Breast Cancer. Cell. Signal. 2020, 68, 109536. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, Y.; He, Q.; Hu, Z.; Feng, Y.; Fan, L.; Tang, Z.; Yuan, J.; Shan, W.; Li, C.; Hu, X.; et al. Long Noncoding RNA LINP1 Regulates Repair of DNA Double-Strand Breaks in Triple-Negative Breast Cancer. Nat. Struct. Mol. Biol. 2016, 23, 522–530. [Google Scholar] [CrossRef] [PubMed]
  114. Liu, Y.; Zhang, P.; Wu, Q.; Fang, H.; Wang, Y.; Xiao, Y.; Cong, M.; Wang, T.; He, Y.; Ma, C.; et al. Long Non-Coding RNA NR2F1-AS1 Induces Breast Cancer Lung Metastatic Dormancy by Regulating NR2F1 and ΔNp63. Nat. Commun. 2021, 12, 5232. [Google Scholar] [CrossRef]
  115. Shi, Q.; Li, Y.; Li, S.; Jin, L.; Lai, H.; Wu, Y.; Cai, Z.; Zhu, M.; Li, Q.; Li, Y.; et al. LncRNA DILA1 Inhibits Cyclin D1 Degradation and Contributes to Tamoxifen Resistance in Breast Cancer. Nat. Commun. 2020, 11, 5513. [Google Scholar] [CrossRef]
  116. Lin, A.; Hu, Q.; Li, C.; Xing, Z.; Ma, G.; Wang, C.; Li, J.; Ye, Y.; Yao, J.; Liang, K.; et al. The LINK-A LncRNA Interacts with PI(3,4,5)P3 to Hyperactivate AKT and Confer Resistance to AKT Inhibitors. Nat. Cell Biol. 2017, 19, 238–251. [Google Scholar] [CrossRef]
  117. Lin, A.; Li, C.; Xing, Z.; Hu, Q.; Liang, K.; Han, L.; Wang, C.; Hawke, D.H.; Wang, S.; Zhang, Y.; et al. The LINK-A LncRNA Activates Normoxic HIF1α Signalling in Triple-Negative Breast Cancer. Nat. Cell Biol. 2016, 18, 213–224. [Google Scholar] [CrossRef]
  118. Liu, J.; Lao, L.; Chen, J.; Li, J.; Zeng, W.; Zhu, X.; Li, J.; Chen, X.; Yang, L.; Xing, Y.; et al. The IRENA LncRNA Converts Chemotherapy-Polarized Tumor-Suppressing Macrophages to Tumor-Promoting Phenotypes in Breast Cancer. Nat. Cancer 2021, 2, 457–473. [Google Scholar] [CrossRef]
  119. Lin, X.; Dinglin, X.; Cao, S.; Zheng, S.; Wu, C.; Chen, W.; Li, Q.; Hu, Q.; Zheng, F.; Wu, Z.; et al. Enhancer-Driven LncRNA BDNF-AS Induces Endocrine Resistance and Malignant Progression of Breast Cancer through the RNH1/TRIM21/MTOR Cascade. Cell Rep. 2020, 31, 107753. [Google Scholar] [CrossRef]
  120. Chang, K.-C.; Diermeier, S.D.; Yu, A.T.; Brine, L.D.; Russo, S.; Bhatia, S.; Alsudani, H.; Kostroff, K.; Bhuiya, T.; Brogi, E.; et al. MaTAR25 LncRNA Regulates the Tensin1 Gene to Impact Breast Cancer Progression. Nat. Commun. 2020, 11, 6438. [Google Scholar] [CrossRef] [PubMed]
  121. Liu, Q.; Wang, Z.; Kong, X.; Wang, X.; Qi, Y.; Gao, R.; Fang, Y.; Wang, J. A Novel Prognostic Signature of MRNA-LncRNA in Breast Cancer. DNA Cell Biol. 2020, 39, 671–682. [Google Scholar] [CrossRef] [PubMed]
  122. Huang, Z.; Qin, Q.; Xia, L.; Lian, B.; Tan, Q.; Yu, Y.; Mo, Q. Significance of Oncotype DX 21-Gene Test and Expression of Long Non-Coding RNA MALAT1 in Early and Estrogen Receptor-Positive Breast Cancer Patients. CMAR 2021, 13, 587–593. [Google Scholar] [CrossRef] [PubMed]
  123. Qi, H.; Chi, L.; Wang, X.; Jin, X.; Wang, W.; Lan, J. Identification of a Seven-LncRNA-MRNA Signature for Recurrence and Prognostic Prediction in Relapsed Acute Lymphoblastic Leukemia Based on WGCNA and LASSO Analyses. Anal. Cell. Pathol. 2021, 2021, e6692022. [Google Scholar] [CrossRef]
  124. Zhou, M.; Zhong, L.; Xu, W.; Sun, Y.; Zhang, Z.; Zhao, H.; Yang, L.; Sun, J. Discovery of Potential Prognostic Long Non-Coding RNA Biomarkers for Predicting the Risk of Tumor Recurrence of Breast Cancer Patients. Sci. Rep. 2016, 6, 31038. [Google Scholar] [CrossRef]
  125. Shen, Y.; Peng, X.; Shen, C. Identification and Validation of Immune-Related LncRNA Prognostic Signature for Breast Cancer. Genomics 2020, 112, 2640–2646. [Google Scholar] [CrossRef]
  126. McVeigh, T.P.; Kerin, M.J. Clinical Use of the Oncotype DX Genomic Test to Guide Treatment Decisions for Patients with Invasive Breast Cancer. Breast Cancer (Dove Med. Press) 2017, 9, 393–400. [Google Scholar] [CrossRef]
  127. Zheng, S.; Yang, L.; Zou, Y.; Liang, J.; Liu, P.; Gao, G.; Yang, A.; Tang, H.; Xie, X. Long Non-Coding RNA HUMT Hypomethylation Promotes Lymphangiogenesis and Metastasis via Activating FOXK1 Transcription in Triple-Negative Breast Cancer. J. Hematol. Oncol. 2020, 13, 17. [Google Scholar] [CrossRef]
  128. Chang, L.; Hu, Z.; Zhou, Z.; Zhang, H. Linc00518 Contributes to Multidrug Resistance Through Regulating the MiR-199a/MRP1 Axis in Breast Cancer. Cell Physiol. Biochem. 2018, 48, 16–28. [Google Scholar] [CrossRef]
  129. Gao, X.; Qin, T.; Mao, J.; Zhang, J.; Fan, S.; Lu, Y.; Sun, Z.; Zhang, Q.; Song, B.; Li, L. PTENP1/MiR-20a/PTEN Axis Contributes to Breast Cancer Progression by Regulating PTEN via PI3K/AKT Pathway. J. Exp. Clin. Cancer Res. 2019, 38, 256. [Google Scholar] [CrossRef]
  130. Zheng, R.; Jia, J.; Guan, L.; Yuan, H.; Liu, K.; Liu, C.; Ye, W.; Liao, Y.; Lin, S.; Huang, O. Long Noncoding RNA Lnc-LOC645166 Promotes Adriamycin Resistance via NF-ΚB/GATA3 Axis in Breast Cancer. Aging 2020, 12, 8893–8912. [Google Scholar] [CrossRef] [PubMed]
  131. Liu, C.; Jiang, F.; Zhang, X.; Xu, X. Long Non-Coding RNA UCA1 Modulates Paclitaxel Resistance in Breast Cancer via MiR-613/CDK12 Axis. CMAR 2020, 12, 2777–2788. [Google Scholar] [CrossRef] [PubMed]
  132. Zhao, C.; Ling, X.; Xia, Y.; Yan, B.; Guan, Q. LncRNA UCA1 Promotes SOX12 Expression in Breast Cancer by Regulating M6A Modification of MiR-375 by METTL14 through DNA Methylation. Cancer Gene Ther. 2022, 29, 1043–1055. [Google Scholar] [CrossRef] [PubMed]
  133. Zhang, M.; Wang, Y.; Jiang, L.; Song, X.; Zheng, A.; Gao, H.; Wei, M.; Zhao, L. LncRNA CBR3-AS1 Regulates of Breast Cancer Drug Sensitivity as a Competing Endogenous RNA through the JNK1/MEK4-Mediated MAPK Signal Pathway. J. Exp. Clin. Cancer Res. 2021, 40, 41. [Google Scholar] [CrossRef] [PubMed]
  134. Li, S.; Mi, X.; Sun, M.; Zhang, J.; Hao, M.; Cao, Q.; Diao, K.; Wang, J.; Li, H.; Liu, B. LncRNA EGFR-AS1 Regulates Breast Cancer Cells Function and Sensitivity to Docetaxel Via the MiR-149-5p/ELP5 Axis. Res. Sq. 2021. [Google Scholar] [CrossRef]
  135. Yao, N.; Fu, Y.; Chen, L.; Liu, Z.; He, J.; Zhu, Y.; Xia, T.; Wang, S. Long Non-Coding RNA NONHSAT101069 Promotes Epirubicin Resistance, Migration, and Invasion of Breast Cancer Cells through NONHSAT101069/MiR-129-5p/Twist1 Axis. Oncogene 2019, 38, 7216–7233. [Google Scholar] [CrossRef]
  136. Wang, J.; Xie, S.; Yang, J.; Xiong, H.; Jia, Y.; Zhou, Y.; Chen, Y.; Ying, X.; Chen, C.; Ye, C.; et al. The Long Noncoding RNA H19 Promotes Tamoxifen Resistance in Breast Cancer via Autophagy. J. Hematol. Oncol. 2019, 12, 81. [Google Scholar] [CrossRef]
  137. Han, J.; Han, B.; Wu, X.; Hao, J.; Dong, X.; Shen, Q.; Pang, H. Knockdown of LncRNA H19 Restores Chemo-Sensitivity in Paclitaxel-Resistant Triple-Negative Breast Cancer through Triggering Apoptosis and Regulating Akt Signaling Pathway. Toxicol. Appl. Pharmacol. 2018, 359, 55–61. [Google Scholar] [CrossRef]
  138. Zhang, H.; Zhang, J.; Dong, L.; Ma, R. LncRNA ATXN8OS Enhances Tamoxifen Resistance in Breast Cancer. Open Med. (Wars) 2020, 16, 68–80. [Google Scholar] [CrossRef]
  139. Yu, Q.; Li, Y.; Peng, S.; Li, J.; Qin, X. Exosomal-Mediated Transfer of OIP5-AS1 Enhanced Cell Chemoresistance to Trastuzumab in Breast Cancer via up-Regulating HMGB3 by Sponging MiR-381-3p. Open Med. 2021, 16, 512–525. [Google Scholar] [CrossRef]
  140. Han, M.; Qian, X.; Cao, H.; Wang, F.; Li, X.; Han, N.; Yang, X.; Yang, Y.; Dou, D.; Hu, J.; et al. LncRNA ZNF649-AS1 Induces Trastuzumab Resistance by Promoting ATG5 Expression and Autophagy. Mol. Ther. 2020, 28, 2488–2502. [Google Scholar] [CrossRef] [PubMed]
  141. Zhang, H.; Zhang, X.-Y.; Kang, X.-N.; Jin, L.-J.; Wang, Z.-Y. LncRNA-SNHG7 Enhances Chemotherapy Resistance and Cell Viability of Breast Cancer Cells by Regulating MiR-186. CMAR 2020, 12, 10163–10172. [Google Scholar] [CrossRef] [PubMed]
  142. Wu, H.; Gu, J.; Zhou, D.; Cheng, W.; Wang, Y.; Wang, Q.; Wang, X. LINC00160 Mediated Paclitaxel-And Doxorubicin-Resistance in Breast Cancer Cells by Regulating TFF3 via Transcription Factor C/EBPβ. J. Cell. Mol. Med. 2020, 24, 8589–8602. [Google Scholar] [CrossRef] [PubMed]
  143. Zheng, P.; Dong, L.; Zhang, B.; Dai, J.; Zhang, Y.; Wang, Y.; Qin, S. Long Noncoding RNA CASC2 Promotes Paclitaxel Resistance in Breast Cancer through Regulation of MiR-18a-5p/CDK19. Histochem. Cell Biol. 2019, 152, 281–291. [Google Scholar] [CrossRef]
  144. Du, C.; Wang, Y.; Zhang, Y.; Zhang, J.; Zhang, L.; Li, J. LncRNA DLX6-AS1 Contributes to Epithelial–Mesenchymal Transition and Cisplatin Resistance in Triple-Negative Breast Cancer via Modulating Mir-199b-5p/Paxillin Axis. Cell Transplant. 2020, 29, 0963689720929983. [Google Scholar] [CrossRef]
  145. Tang, L.; Wei, D.; Xu, X.; Mao, X.; Mo, D.; Yan, L.; Xu, W.; Yan, F. Long Non-Coding RNA MIR200CHG Promotes Breast Cancer Proliferation, Invasion, and Drug Resistance by Interacting with and Stabilizing YB-1. Npj Breast Cancer 2021, 7, 94. [Google Scholar] [CrossRef]
  146. Liang, Y.; Song, X.; Li, Y.; Sang, Y.; Zhang, N.; Zhang, H.; Liu, Y.; Duan, Y.; Chen, B.; Guo, R.; et al. A Novel Long Non-Coding RNA-PRLB Acts as a Tumor Promoter through Regulating MiR-4766-5p/SIRT1 Axis in Breast Cancer. Cell Death Dis. 2018, 9, 563. [Google Scholar] [CrossRef]
  147. Cai, Y.; He, J.; Zhang, D. Long Noncoding RNA CCAT2 Promotes Breast Tumor Growth by Regulating the Wnt Signaling Pathway. Onco. Targets Ther. 2015, 8, 2657–2664. [Google Scholar] [CrossRef]
  148. Zhou, D.; Gu, J.; Wang, Y.; Luo, B.; Feng, M.; Wang, X. Long Noncoding RNA CCAT2 Reduces Chemosensitivity to 5-Fluorouracil in Breast Cancer Cells by Activating the MTOR Axis. J. Cell. Mol. Med. 2022, 26, 1392–1401. [Google Scholar] [CrossRef]
  149. Luo, L.; Zhang, J.; Tang, H.; Zhai, D.; Huang, D.; Ling, L.; Wang, X.; Liu, T.; Zhang, Q.; Zhang, Z.; et al. LncRNA SNORD3A Specifically Sensitizes Breast Cancer Cells to 5-FU by Sponging MiR-185-5p to Enhance UMPS Expression. Cell Death Dis. 2020, 11, 329. [Google Scholar] [CrossRef]
  150. Yao, X.; Tu, Y.; Xu, Y.; Guo, Y.; Yao, F.; Zhang, X. Endoplasmic Reticulum Stress Confers 5-Fluorouracil Resistance in Breast Cancer Cell via the GRP78/OCT4/LncRNA MIAT/AKT Pathway. Am. J. Cancer Res. 2020, 10, 838–855. [Google Scholar] [PubMed]
  151. Ye, T.; Ye, Z. LncRNA NORAD as a Novel Predictor of Lymph Node Metastasis and Prognosis in Solid Tumors: A Systematic Review and Meta-Analysis. Technol. Cancer Res. Treat 2020, 19, 1533033820963586. [Google Scholar] [CrossRef] [PubMed]
  152. Ensenyat-Mendez, M.; Rünger, D.; Orozco, J.I.J.; Le, J.; Baker, J.L.; Weidhaas, J.; Marzese, D.M.; DiNome, M.L. Epigenetic Signatures Predict Pathologic Nodal Stage in Breast Cancer Patients with Estrogen Receptor-Positive, Clinically Node-Positive Disease. Ann. Surg. Oncol. 2022, 29, 4716–4724. [Google Scholar] [CrossRef]
  153. Yu, Y.; Zhang, W.; Li, A.; Chen, Y.; Ou, Q.; He, Z.; Zhang, Y.; Liu, R.; Yao, H.; Song, E. Association of Long Noncoding RNA Biomarkers with Clinical Immune Subtype and Prediction of Immunotherapy Response in Patients With Cancer. JAMA Netw. Open 2020, 3, e202149. [Google Scholar] [CrossRef] [PubMed]
  154. Stelcer, E.; Konkol, M.; Głȩboka, A.; Suchorska, W.M. Liquid Biopsy in Oligometastatic Prostate Cancer—A Biologist’s Point of View. Front. Oncol. 2019, 9, 775. [Google Scholar] [CrossRef]
  155. Cock, L.D.; Heylen, J.; Wildiers, A.; Punie, K.; Smeets, A.; Weltens, C.; Neven, P.; Billen, J.; Laenen, A.; Wildiers, H. Detection of Secondary Metastatic Breast Cancer by Measurement of Plasma CA 15.3. ESMO Open 2021, 6, 100203. [Google Scholar] [CrossRef] [PubMed]
  156. Laas, E.; Labrosse, J.; Hamy, A.-S.; Benchimol, G.; de Croze, D.; Feron, J.-G.; Coussy, F.; Balezeau, T.; Guerin, J.; Lae, M.; et al. Determination of Breast Cancer Prognosis after Neoadjuvant Chemotherapy: Comparison of Residual Cancer Burden (RCB) and Neo-Bioscore. Br. J. Cancer 2021, 124, 1421–1427. [Google Scholar] [CrossRef]
  157. Müller, C.; Schmidt, G.; Juhasz-Böss, I.; Jung, L.; Huwer, S.; Solomayer, E.-F.; Juhasz-Böss, S. Influences on Pathologic Complete Response in Breast Cancer Patients after Neoadjuvant Chemotherapy. Arch. Gynecol. Obstet. 2021, 304, 1065–1071. [Google Scholar] [CrossRef]
  158. Winkle, M.; El-Daly, S.M.; Fabbri, M.; Calin, G.A. Noncoding RNA Therapeutics—Challenges and Potential Solutions. Nat. Rev. Drug Discov. 2021, 20, 629–651. [Google Scholar] [CrossRef]
  159. Di Martino, M.T.; Riillo, C.; Scionti, F.; Grillone, K.; Polerà, N.; Caracciolo, D.; Arbitrio, M.; Tagliaferri, P.; Tassone, P. MiRNAs and LncRNAs as Novel Therapeutic Targets to Improve Cancer Immunotherapy. Cancers 2021, 13, 1587. [Google Scholar] [CrossRef]
  160. Hu, Q.; Ye, Y.; Chan, L.-C.; Li, Y.; Liang, K.; Lin, A.; Egranov, S.D.; Zhang, Y.; Xia, W.; Gong, J.; et al. Oncogenic LncRNA Downregulates Cancer Cell Antigen Presentation and Intrinsic Tumor Suppression. Nat. Immunol. 2019, 20, 835–851. [Google Scholar] [CrossRef] [PubMed]
  161. Nooka, A.K.; Wang, M.L.; Yee, A.J.; Kaufman, J.L.; Bae, J.; Peterkin, D.; Richardson, P.G.; Raje, N.S. Assessment of Safety and Immunogenicity of PVX-410 Vaccine with or Without Lenalidomide in Patients with Smoldering Multiple Myeloma: A Nonrandomized Clinical Trial. JAMA Oncol. 2018, 4, e183267. [Google Scholar] [CrossRef] [PubMed]
  162. Luo, Y.; Yang, J.; Yu, J.; Liu, X.; Yu, C.; Hu, J.; Shi, H.; Ma, X. Long Non-Coding RNAs: Emerging Roles in the Immunosuppressive Tumor Microenvironment. Front. Oncol. 2020, 10, 48. [Google Scholar] [CrossRef] [PubMed]
  163. Gong, N.; Teng, X.; Li, J.; Liang, X.-J. Antisense Oligonucleotide-Conjugated Nanostructure-Targeting LncRNA MALAT1 Inhibits Cancer Metastasis. ACS Appl. Mater. Interfaces 2019, 11, 37–42. [Google Scholar] [CrossRef]
  164. Evans, J.R.; Feng, F.Y.; Chinnaiyan, A.M. The Bright Side of Dark Matter: LncRNAs in Cancer. J. Clin. Investig. 2016, 126, 2775–2782. [Google Scholar] [CrossRef] [PubMed]
  165. Duffy, M.J.; Harbeck, N.; Nap, M.; Molina, R.; Nicolini, A.; Senkus, E.; Cardoso, F. Clinical Use of Biomarkers in Breast Cancer: Updated Guidelines from the European Group on Tumor Markers (EGTM). Eur. J. Cancer 2017, 75, 284–298. [Google Scholar] [CrossRef]
  166. Korde, L.A.; Somerfield, M.R.; Carey, L.A.; Crews, J.R.; Denduluri, N.; Hwang, E.S.; Khan, S.A.; Loibl, S.; Morris, E.A.; Perez, A.; et al. Neoadjuvant Chemotherapy, Endocrine Therapy, and Targeted Therapy for Breast Cancer: ASCO Guideline. J. Clin. Oncol. 2021, 39, 1485–1505. [Google Scholar] [CrossRef]
  167. Zhang, K.; Luo, Z.; Zhang, Y.; Song, X.; Zhang, L.; Wu, L.; Liu, J. Long Non-Coding RNAs as Novel Biomarkers for Breast Cancer Invasion and Metastasis (Review). Oncol. Lett. 2017, 14, 1895–1904. [Google Scholar] [CrossRef]
  168. Xin, L.; Liu, Y.-H.; Martin, T.A.; Jiang, W.G. The Era of Multigene Panels Comes? The Clinical Utility of Oncotype DX and MammaPrint. World J. Oncol. 2017, 8, 34–40. [Google Scholar] [CrossRef]
  169. Manjang, K.; Tripathi, S.; Yli-Harja, O.; Dehmer, M.; Glazko, G.; Emmert-Streib, F. Prognostic Gene Expression Signatures of Breast Cancer Are Lacking a Sensible Biological Meaning. Sci. Rep. 2021, 11, 156. [Google Scholar] [CrossRef]
  170. Lehmann, B.D.; Colaprico, A.; Silva, T.C.; Chen, J.; An, H.; Ban, Y.; Huang, H.; Wang, L.; James, J.L.; Balko, J.M.; et al. Multi-Omics Analysis Identifies Therapeutic Vulnerabilities in Triple-Negative Breast Cancer Subtypes. Nat. Commun. 2021, 12, 6276. [Google Scholar] [CrossRef] [PubMed]
  171. Nguyen, Q.-H.; Nguyen, H.; Nguyen, T.; Le, D.-H. Multi-Omics Analysis Detects Novel Prognostic Subgroups of Breast Cancer. Front. Genet. 2020, 11, 1265. [Google Scholar] [CrossRef] [PubMed]
  172. Zhu, L.; Cui, K.; Weng, L.; Yu, P.; Du, Y.; Zhang, T.; Liu, H.; Li, B.; Ma, W. A Panel of 8-LncRNA Predicts Prognosis of Breast Cancer Patients and Migration of Breast Cancer Cells. PLoS ONE 2021, 16, e0249174. [Google Scholar] [CrossRef]
  173. Fan, C.-N.; Ma, L.; Liu, N. Comprehensive Analysis of Novel Three-Long Noncoding RNA Signatures as a Diagnostic and Prognostic Biomarkers of Human Triple-Negative Breast Cancer. J. Cell. Biochem. 2019, 120, 3185–3196. [Google Scholar] [CrossRef] [PubMed]
  174. Sun, J.; Chen, X.; Wang, Z.; Guo, M.; Shi, H.; Wang, X.; Cheng, L.; Zhou, M. A Potential Prognostic Long Non-Coding RNA Signature to Predict Metastasis-Free Survival of Breast Cancer Patients. Sci. Rep. 2015, 5, 16553. [Google Scholar] [CrossRef] [PubMed]
  175. Ma, J.; Liu, S.; Chen, J.; Liu, Q. Metabolism-Related Long Non-Coding RNAs (LncRNAs) as Potential Biomarkers for Predicting Risk of Recurrence in Breast Cancer Patients. Bioengineered 2021, 12, 3726–3736. [Google Scholar] [CrossRef]
  176. Lai, J.; Chen, B.; Zhang, G.; Li, X.; Mok, H.; Liao, N. Molecular Characterization of Breast Cancer: A Potential Novel Immune-Related LncRNAs Signature. J. Transl. Med. 2020, 18, 416. [Google Scholar] [CrossRef]
  177. Wang, K.; Li, J.; Xiong, Y.-F.; Zeng, Z.; Zhang, X.; Li, H.-Y. A Potential Prognostic Long Noncoding RNA Signature to Predict Recurrence among ER-Positive Breast Cancer Patients Treated with Tamoxifen. Sci. Rep. 2018, 8, 3179. [Google Scholar] [CrossRef]
  178. Sun, M.; Liu, X.; Xia, L.; Chen, Y.; Kuang, L.; Gu, X.; Li, T. A Nine-LncRNA Signature Predicts Distant Relapse-Free Survival of HER2-Negative Breast Cancer Patients Receiving Taxane and Anthracycline-Based Neoadjuvant Chemotherapy. Biochem. Pharmacol. 2021, 189, 114285. [Google Scholar] [CrossRef]
  179. Li, Z.; Zheng, J.; Feng, Y.; Li, Y.; Liang, Y.; Liu, Y.; Wang, X.; Yang, Q. Integrated Analysis Identifies a Novel LncRNA Prognostic Signature Associated with Aerobic Glycolysis and Hub Pathways in Breast Cancer. Cancer Med. 2021, 10, 7877–7892. [Google Scholar] [CrossRef]
  180. Li, J.; Wang, W.; Xia, P.; Wan, L.; Zhang, L.; Yu, L.; Wang, L.; Chen, X.; Xiao, Y.; Xu, C. Identification of a Five-LncRNA Signature for Predicting the Risk of Tumor Recurrence in Breast Cancer Patients. Int. J. Cancer 2018, 143, 2150–2160. [Google Scholar] [CrossRef] [PubMed]
  181. Shen, S.; Chen, X.; Hu, X.; Huo, J.; Luo, L.; Zhou, X. Predicting the Immune Landscape of Invasive Breast Carcinoma Based on the Novel Signature of Immune-related LncRNA. Cancer Med. 2021, 10, 6561–6575. [Google Scholar] [CrossRef] [PubMed]
  182. Li, Y.; Wang, S.; Li, C.; Li, Y.; Wang, S.; Li, C. Four-LncRNA Immune Prognostic Signature for Triple-Negative Breast Cancer. MBE 2021, 18, 3939–3956. [Google Scholar] [CrossRef]
  183. Zhang, X.; Zhang, X.; Li, G.; Hao, Y.; Liu, L.; Zhang, L.; Chen, Y.; Wu, J.; Wang, X.; Yang, S.; et al. A Novel Necroptosis-Associated LncRNA Signature Can Impact the Immune Status and Predict the Outcome of Breast Cancer. J. Immunol. Res. 2022, 2022, 3143511. [Google Scholar] [CrossRef] [PubMed]
  184. Zhang, Y.; Li, Z.; Chen, M.; Chen, H.; Zhong, Q.; Liang, L.; Li, B. Identification of a New Eight-Long Noncoding RNA Molecular Signature for Breast Cancer Survival Prediction. DNA Cell Biol. 2019, 38, 1529–1539. [Google Scholar] [CrossRef] [PubMed]
  185. Meng, J.; Li, P.; Zhang, Q.; Yang, Z.; Fu, S. A Four-Long Non-Coding RNA Signature in Predicting Breast Cancer Survival. J. Exp. Clin. Cancer Res. 2014, 33, 84. [Google Scholar] [CrossRef]
  186. Liu, D. Identification of a Prognostic LncRNA Signature for ER-Positive, ER-Negative and Triple-Negative Breast Cancers. Breast Cancer Res. Treat. 2020, 183, 95–105. [Google Scholar] [CrossRef]
  187. Ma, W.; Zhao, F.; Yu, X.; Guan, S.; Suo, H.; Tao, Z.; Qiu, Y.; Wu, Y.; Cao, Y.; Jin, F. Immune-Related LncRNAs as Predictors of Survival in Breast Cancer: A Prognostic Signature. J. Transl. Med. 2020, 18, 442. [Google Scholar] [CrossRef]
  188. Wu, Q.; Li, Q.; Zhu, W.; Zhang, X.; Li, H. Identification of Autophagy-related Long Non-coding RNA Prognostic Signature for Breast Cancer. J. Cell Mol. Med. 2021, 25, 4088–4098. [Google Scholar] [CrossRef]
  189. Shen, S.; Yang, D.; Yang, Y.; Chen, Y.; Xiong, J.; Hu, X. A Novel Prognostic Ferroptosis-Related LncRNA Signature Associated with Immune Landscape in Invasive Breast Cancer. Dis. Markers 2022, 2022, 9168556. [Google Scholar] [CrossRef]
  190. Liu, Z.; Mi, M.; Li, X.; Zheng, X.; Wu, G.; Zhang, L. A LncRNA Prognostic Signature Associated with Immune Infiltration and Tumour Mutation Burden in Breast Cancer. J. Cell Mol. Med. 2020, 24, 12444–12456. [Google Scholar] [CrossRef] [PubMed]
  191. Li, X.; Li, Y.; Yu, X.; Jin, F. Identification and Validation of Stemness-Related LncRNA Prognostic Signature for Breast Cancer. J. Transl. Med. 2020, 18, 331. [Google Scholar] [CrossRef] [PubMed]
  192. Chen, F.; Yang, J.; Fang, M.; Wu, Y.; Su, D.; Sheng, Y. Necroptosis-related LncRNA to Establish Novel Prognostic Signature and Predict the Immunotherapy Response in Breast Cancer. J. Clin. Lab. Anal. 2022, 36, e24302. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Workflow for lncRNA validation as biomarkers with clinical utility and application. The process of implementing a lncRNA-based biomarker consists of 5 principal steps: discovery, validation, verification, clinical application, and clinical utility [97]. In the discovery step, the objective is to select a lncRNA (or a set) differentially expressed in the condition of interest, like treatment response, and implement it in selected patients. In the validation step, the sample size is increased to determine lncRNA robustness to define the clinical condition of interest and follow the sample size calculation recommended in [20]. In the verification step, lncRNA expression is determined by a clinical laboratory technique, such as qPCR, to verify its viability to be detected in the clinical routine. In the clinical application step, the functionality of the lncRNA as a biomarker for diagnosis, prediction, or prognosis is determined by assessing its sensibility and specificity [20]. Finally, in the clinical utility step, the accuracy of the lncRNA as biomarker is tested in a larger sample size and could be included in clinical trials. (A) PCA3 is an example of a lncRNA that has been validated for clinical application in prostate cancer diagnosis because it represents an FDA-approved lncRNA for clinical purposes. It was discovered from a sample size of 11 patients in the discovery phase [49] and 507 male patients were included in the validation phase in a clinical trial [53]. Additionally, PCA3 is associated with a sensitivity ranging from 54% to 82% and a specificity range of 56.3% to 89%, which justifies its use in clinical practice [50]. Although PCA3 was identified by Northern blot technique [49], it has been validated in other studies by high throughput sequencing technologies, which is the principal tool for current biomarker discovery [98]. (B) EPIC1 is a lncRNA that was identified from the analysis of 6475 tumor samples in the discovery phase and 534 samples in the validation phase [99]. However, it has not yet been verified as a biomarker for clinical utility or for clinical application in the prognosis of breast cancer. (C) GATA3-AS1 is a lncRNA proposed as a potential clinical biomarker in predicting treatment response in breast cancer because it has been demonstrated by Zhang et al. that it is overexpressed in breast neoplasia in a differential expression analysis for 85 paired tumor-normal samples and 830 tumor samples in the discovery phase. For the validation step, 50 paired tumor-normal samples and 23 healthy samples were included [14]. Recently, Contreras-Espinosa et al. also identified this lncRNA by a machine learning approach in a sample size of 11 patients for the discovery step and 68 patients for the validation step, which demonstrated its utility as a predictive biomarker [17]. However, it has not been validated for clinical application yet, as other lncRNA which have been proposed as molecular biomarkers for treatment response prediction in breast cancer, like HOTAIR [8] and MALAT1 [36], have not been included in clinical trials for the analysis of their applicability in diagnosis. lncRNA: long non-coding RNA; ISH: in situ hybridization; PCR: Polymerase Chain Reaction; qPCR: quantitative PCR. Created in BioRender.com.
Figure 1. Workflow for lncRNA validation as biomarkers with clinical utility and application. The process of implementing a lncRNA-based biomarker consists of 5 principal steps: discovery, validation, verification, clinical application, and clinical utility [97]. In the discovery step, the objective is to select a lncRNA (or a set) differentially expressed in the condition of interest, like treatment response, and implement it in selected patients. In the validation step, the sample size is increased to determine lncRNA robustness to define the clinical condition of interest and follow the sample size calculation recommended in [20]. In the verification step, lncRNA expression is determined by a clinical laboratory technique, such as qPCR, to verify its viability to be detected in the clinical routine. In the clinical application step, the functionality of the lncRNA as a biomarker for diagnosis, prediction, or prognosis is determined by assessing its sensibility and specificity [20]. Finally, in the clinical utility step, the accuracy of the lncRNA as biomarker is tested in a larger sample size and could be included in clinical trials. (A) PCA3 is an example of a lncRNA that has been validated for clinical application in prostate cancer diagnosis because it represents an FDA-approved lncRNA for clinical purposes. It was discovered from a sample size of 11 patients in the discovery phase [49] and 507 male patients were included in the validation phase in a clinical trial [53]. Additionally, PCA3 is associated with a sensitivity ranging from 54% to 82% and a specificity range of 56.3% to 89%, which justifies its use in clinical practice [50]. Although PCA3 was identified by Northern blot technique [49], it has been validated in other studies by high throughput sequencing technologies, which is the principal tool for current biomarker discovery [98]. (B) EPIC1 is a lncRNA that was identified from the analysis of 6475 tumor samples in the discovery phase and 534 samples in the validation phase [99]. However, it has not yet been verified as a biomarker for clinical utility or for clinical application in the prognosis of breast cancer. (C) GATA3-AS1 is a lncRNA proposed as a potential clinical biomarker in predicting treatment response in breast cancer because it has been demonstrated by Zhang et al. that it is overexpressed in breast neoplasia in a differential expression analysis for 85 paired tumor-normal samples and 830 tumor samples in the discovery phase. For the validation step, 50 paired tumor-normal samples and 23 healthy samples were included [14]. Recently, Contreras-Espinosa et al. also identified this lncRNA by a machine learning approach in a sample size of 11 patients for the discovery step and 68 patients for the validation step, which demonstrated its utility as a predictive biomarker [17]. However, it has not been validated for clinical application yet, as other lncRNA which have been proposed as molecular biomarkers for treatment response prediction in breast cancer, like HOTAIR [8] and MALAT1 [36], have not been included in clinical trials for the analysis of their applicability in diagnosis. lncRNA: long non-coding RNA; ISH: in situ hybridization; PCR: Polymerase Chain Reaction; qPCR: quantitative PCR. Created in BioRender.com.
Ijms 24 07426 g001
Table 1. List of clinical trials and preclinical studies in breast cancer research that also included lncRNAs.
Table 1. List of clinical trials and preclinical studies in breast cancer research that also included lncRNAs.
Clinical Trials for lncRNA *
Clinical Trial IDTitleDiseaseNumber of ParticipantsPhase
NCT02641847TA(E)C-GP Versus A(E)C-T for the High Risk TNBC Patients and Validation of the mRNA-lncRNA Signature.Triple Negative Breast Cancer, Breast Cancer.503Phase 2
Phase 3
NCT03000764RNA and Heat Shock Protein Biomarkers in Radiation-induced Fibrosis in Breast Cancer (SPLICI-Rad).Breast Carcinoma, Fibrosis.20Not Applicable
NCT02221999Weekly Paclitaxel and Cisplatin to Treat Hormone Receptor Positive and Triple Negative Breast Cancer Patients (SHPD002).Tubular Breast Cancer, Mucinous Breast Cancer, Invasive Ductal Breast Cancer, Inflammatory Breast Cancer.250Phase 2
Phase 3
LncRNA in Preclinical studies
LncRNABiological Function +DiseaseApplicationReference
LINP1Proliferation and migrationTriple Negative Breast CancerPredictive biomarker for radiotherapy response[112,113]
NR2F1-AS1 (NAS1)EMT and invasionLung Metastasis Breast CancerPrognostic biomarker for metastasis-free survival and relapse-free survival[114]
DILA1Cell cycle progression and proliferationER-Positive Breast CancerPredictive biomarker for tamoxifen response.
Prognostic biomarker for relapse-free survival
[115]
LINK-AProliferation and survivalTriple Negative Breast CancerBiomarker for stratification of Triple Negative Breast Cancer Patients[116,117]
IRENAProliferationInvasive Breast CarcinomaPrognostic biomarker. Disease-free survival[118]
BDNF-ASProliferationTriple Negative Breast CancerPredictive biomarker for tamoxifen response[119]
LINC01271Proliferation and migrationBreast CancerMetastasis-related biomarker[120]
* Clinical trials were consulted in https://clinicaltrials.gov/ (accessed on 13 January 2023). Search terms: [LncRNA] AND [Breast Cancer]. + Studies performed in breast cancer cell lines.
Table 2. Studies in mouse models for lncRNA biomarker discovery in breast cancer.
Table 2. Studies in mouse models for lncRNA biomarker discovery in breast cancer.
LncRNADrugExpression StatusResponseReference
linc00518DoxorubicinUpregulatedPromotes resistance[128]
PTENP1DoxorubicinDownregulatedPromotes resistance[129]
LOC645166DoxorubicinUpregulatedPromotes resistance[130]
UCA1Doxorubicin
Tamoxifen
Paclitaxel
UpregulatedPromotes resistance[131,132]
CBR3-AS1DoxorubicinUpregulatedPromotes resistance[133]
EGFR-AS1DocetaxelUpregulatedPromotes resistance[134]
NONHSAG048143.2DocetaxelUpregulatedPromotes resistance[135]
DILA1TamoxifenUpregulatedPromotes resistance[115]
BDNF-ASTamoxifenUpregulatedPromotes resistance[119]
LINP1TamoxifenUpregulatedPromotes resistance[112]
H19Tamoxifen
Paclitaxel
UpregulatedPromotes resistance[136,137]
ATXN8OSTamoxifenUpregulatedPromotes resistance[138]
OIP5-AS1TrastuzumabUpregulatedPromotes resistance[139]
ZNF649-AS1TrastuzumabUpregulatedPromotes resistance[140]
SNHG7TrastuzumabUpregulatedPromotes resistance[141]
LINC00160PaclitaxelUpregulatedPromotes resistance[142]
CASC2PaclitaxelUpregulatedPromotes resistance[143]
DLX6-AS1Cisplatin, CarboplatinUpregulatedPromotes resistance[144]
MIR200CHGCisplatin, CarboplatinUpregulatedPromotes resistance[145]
PRLB5-FluorouracilUpregulatedPromotes resistance[146]
CCAT25-FluorouracilUpregulatedPromotes resistance[147,148]
SNORD3A5-FluorouracilUpregulatedPromotes resistance[149]
MIAT5-FluorouracilUpregulatedPromotes resistance[150]
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

Arriaga-Canon, C.; Contreras-Espinosa, L.; Aguilar-Villanueva, S.; Bargalló-Rocha, E.; García-Gordillo, J.A.; Cabrera-Galeana, P.; Castro-Hernández, C.; Jiménez-Trejo, F.; Herrera, L.A. The Clinical Utility of lncRNAs and Their Application as Molecular Biomarkers in Breast Cancer. Int. J. Mol. Sci. 2023, 24, 7426. https://doi.org/10.3390/ijms24087426

AMA Style

Arriaga-Canon C, Contreras-Espinosa L, Aguilar-Villanueva S, Bargalló-Rocha E, García-Gordillo JA, Cabrera-Galeana P, Castro-Hernández C, Jiménez-Trejo F, Herrera LA. The Clinical Utility of lncRNAs and Their Application as Molecular Biomarkers in Breast Cancer. International Journal of Molecular Sciences. 2023; 24(8):7426. https://doi.org/10.3390/ijms24087426

Chicago/Turabian Style

Arriaga-Canon, Cristian, Laura Contreras-Espinosa, Sergio Aguilar-Villanueva, Enrique Bargalló-Rocha, José Antonio García-Gordillo, Paula Cabrera-Galeana, Clementina Castro-Hernández, Francisco Jiménez-Trejo, and L. A. Herrera. 2023. "The Clinical Utility of lncRNAs and Their Application as Molecular Biomarkers in Breast Cancer" International Journal of Molecular Sciences 24, no. 8: 7426. https://doi.org/10.3390/ijms24087426

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