• Open Access
    Review

    Molecular interaction of metastasis suppressor genes and tumor microenvironment in breast cancer

    Sathammai Sathappa Supuramanian
    Sid Dsa
    Sitaram Harihar *

    Explor Target Antitumor Ther. 2023;4:912–932 DOI: https://doi.org/10.37349/etat.2023.00173

    Received: February 27, 2023 Accepted: August 03, 2023 Published: October 11, 2023

    Academic Editor: Sufi Thomas, University of Kansas Medical Center, USA

    This article belongs to the special issue Therapeutic Targeting of the Tumor Microenvironment

    Abstract

    Breast cancer (BC) is a leading cause of cancer-related deaths in women worldwide where the process of metastasis is a major contributor to the mortality associated with this disease. Metastasis suppressor genes are a group of genes that play a crucial role in preventing or inhibiting the spread of cancer cells. They suppress the metastasis process by inhibiting colonization and by inducing dormancy. These genes function by regulating various cellular processes in the tumor microenvironment (TME), such as cell adhesion, invasion, migration, and angiogenesis. Dysregulation of metastasis suppressor genes can lead to the acquisition of an invasive and metastatic phenotype and lead to poor prognostic outcomes. The components of the TME generally play a necessary in the metastasis progression of tumor cells. This review has identified and elaborated on the role of a few metastatic suppressors associated with the TME that have been shown to inhibit metastasis in BC by different mechanisms, such as blocking certain cell signaling molecules involved in cancer cell migration, invasion, enhancing immune surveillance of cancer cells, and promoting the formation of a protective extracellular matrix (ECM). Understanding the interaction of metastatic suppressor genes and the components of TME has important implications for the development of novel therapeutic strategies to target the metastatic cascade. Targeting these genes or their downstream signaling pathways offers a promising approach to inhibiting the spread of cancer cells and improves patient outcomes.

    Keywords

    Tumor microenvironment, metastasis suppressor genes, breast cancer, breast cancer metastasis suppressor 1

    Introduction

    Breast cancer (BC) is a diverse disease that is the most prevalent cancer in the world, particularly among women [1]. BC causes more women to lose disability-adjusted life years (DALYs) than other cancers. The World Health Organization (WHO) estimates that 2.3 million women receive a BC diagnosis each year. BC starts in the lining cells (epithelium) of the ducts (85%) or lobules (15%) of the glandular tissue of the breast [2]. Estrogen exposure can alter the genetic alterations and DNA damage that leads to BC. Classification of BC depends on the presence or absence of estrogen receptor alpha (ERα), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Invasive or infiltrating pre-cancer, which is less common and more severe, spreads into the surrounding breast tissue after starting in a milk duct. The in situ pre-cancer, which begins in a milk duct but does not extend to the rest of the breast tissue, is distinct from the invasive one. One in eight women (12.4%) may get invasive BC in their lifetime. Frequent screening for BC, primarily via mammography, can significantly decrease the chance of death from the disease [3]. Some of the risk factors, both changeable and immutable, include age, family history, mutations, alcohol consumption, smoking, and more [4]. Furthermore, a significant percentage of those who are diagnosed with BC disclose having a first-degree relative who has the disease [5]. BC susceptibility gene 1 (BRCA1) and BRCA2 are two significant genes with high penetrance found on chromosomes 17 and 13, respectively. They primarily raise the possibility of developing BC [6]. An assortment of breast tumors that are ER-negative, PR-negative, and HER2-negative collectively fall under the term “triple-negative BC (TNBC)”. TNBC accounts for over 80% of BCs with BRCA1 germline mutations, while BRCA1 or BRCA2 germline mutations account for 11–16% of all TNBCs. TNBC frequently has a worse prognosis and is physiologically more aggressive [7, 8]. Some of the treatment strategies include surgery, radiotherapy, chemotherapy, and hormone therapy [4].

    The biological mechanism known as the metastatic cascade allows for the spread of cancerous cells from their original site to a secondary site and the emergence of new cancer cells [9]. In contrast to other organs like the liver and brain, BC has been observed to spread more frequently to the bone and lungs [10]. It is well-recognized that circulating tumor cells are crucial to the metastasis of carcinomas. Because a favorable microenvironment is vital for the growth and progression of malignant tumors, it is necessary for the proliferation of metastatic tumor cells [11]. The possibility exists that tumor cells may manufacture substances to prepare the “soil” prior to metastasis in order to establish a “pre-metastatic niche” that would support future metastatic locales [10, 12]. The evidence is mounting in favor of the hypothesis that strategies emphasizing the interactions between the tumor and TME may pave the way for a new generation of therapies. Numerous pre-clinical or clinical studies have demonstrated that targeting tumor microenvironment (TME) with directed therapy reduces tumor growth, metastasis, and chemoresistance [13, 14]. Interaction between the tumor cell and its microenvironment is necessary to establish metastasis; therefore, cutting off this interaction should lessen the likelihood of metastasis.

    A protein known as a metastasis suppressor slows or stops metastases (secondary tumors) from growing and spreading throughout the body of a cancerous organism [15]. Metastasis suppressors frequently appear to selectively regulate how cells react to environmental signals by modifying signaling cascades that regulate downstream gene expression [16, 17]. Nevertheless, metastasis suppressors exist in both cells and the extracellular milieu; they each have a unique method of action and control a different stage of the metastatic cascade [16]. This review’s primary focus is metastasis suppressor genes related to BC and their function in regulating the TME. Understanding the interaction between the components of the TME and metastasis suppressor genes in order to inhibit the metastasis of BC may pave the way for potential therapeutic opportunities.

    TME in BC

    A tumor is a heterogeneous assemblage of invading and resident host cells, secreted chemicals, and extracellular matrix (ECM) rather than just a collection of cancer cells [18]. Tumor growth and progression are influenced by two distinct pathways, namely genetic and epigenetic alterations in the tumor cells and the reorganization of the TME elements through reciprocal and dynamic interaction [19]. Through intricate signaling networks, tumor cells, the driving force being the TME, regulate cellular and no-cellular functions to take advantage of non-malignant cells for their interests [19, 20].

    The stages of the metastatic cascade depend on interactions between the microenvironment and cancer cells. Immune cells and their mediators promote metastatic formation in this regional milieu and distant organs. Blood vessels, fibroblasts, ECM, signaling molecules, immune cells, non-neoplastic cells, tumor cells, and more make up the TME (Figure 1), which promotes the growth and spread of cancer [19, 21]. Using a variety of markers, the cellular segmentation of tumors and their associated TMEs allowed the detection of tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), T and B lymphocytes, natural killer (NK) cells, and cancer stem cells (CSCs) [22]. The microenvironments in which tumors grow, invade, and metastasize are intricate and dynamic. In order to target them and use them as a potential therapeutic target, it is crucial to understand the involvement of the TME in BC.

    Cellular components of the TME and surrounding BC cells. This figure was created with BioRender

    In the microenvironment of solid tumors, fibroblasts are one of the most prevalent cell types. Breast, pancreatic, colon, and prostate carcinomas are among those where fibroblasts are most prevalent [23]. The most prevalent cell type in the stroma of BC, CAFs, produce an abundance of chemokines, growth hormones, and ECM proteins that may encourage metastasis and dissemination [24]. Compared to their regular counterparts, the growth of tumors in xenograft models was greatly enhanced by fibroblasts from primary human invasive breast carcinomas [25]. The interaction of integrin three and CAF-derived interleukin 32 (IL32) at the tumor cell membrane, which enables the cross-talk between CAFs and breast tumor cells, promotes BC invasion [26]. GREMLIN 1, an antagonist of bone morphogenetic protein (BMP) signaling significantly impacts the connection between BC cells and CAFs, which encourages cancer cell invasion [27]. Another study claimed that BC’s epithelial-mesenchymal transition (EMT), invasion, and metastasis are caused by the exosome-derived microRNA-18b (miR-18b) from CAFs, which targets transcription elongation factor A like 7 (TCEAL7) and activates the nuclear factor-kappa B (NF-κB) pathway [28]. Also, through transfer from nearby CAF cells, patient-derived CAF exosomal miR-500a-5p can give BC cells an aggressive phenotype [29]. Given that CAF is frequently seen in brain metastases and that they encourage BC cell invasion, colony formation, and transmigration in vitro, they may also substantially impact the development of brain metastases in BC patients [30]. Furthermore, it has been demonstrated that signal transducer and activator of transcription 3 (STAT3) promotes the production of angiopoetin-like 4 (ANGPTL4), matrix metalloproteinase 13 (MMP13), and stanniocalcin-1 (STC1) by fibroblasts associated with the disease, which in turn promotes the growth of BC [31].

    The triple-negative and HER2-positive BC subtypes have shown prognostic and predictive utility in immune cells and tumor-infiltrating (TI) lymphocytes (TILs) [32, 33]. T cells comprising cluster of differentiation 4+ (CD4+) helper cells, Forkhead box P3+ (FOXP3+), regulatory T (Treg), and effector cells such as NK cells and CD8+ T cells comprise most of the lymphocytes that infiltrate tumors [34]. TI Tregs interact with ECM elements, tumor and stromal cells, and other cells in the TME to generate an immune-suppressive phenotype. Due to Treg infiltration into the TME, TNBC is incredibly susceptible to medication resistance and early recurrence (TME) [35]. According to theory, the tumor creates an immunosuppressive microenvironment by secreting IL10 and transforming growth factor-β1 (TGF-β1) and increasing the activities of effector cells by releasing prostaglandin E2 and TGF-β1 signaling [36]. In the stroma of strongly perfused samples, close to but not inside blood vessels, most BCs are thought to have more CD4+ naive T cells than neighboring normal tissue. Their quantity is also related to TI Tregs, their clonotypic T cell receptor (TCR) sequences are most comparable to those of TI Tregs, and the presence of naive CD4+ T cells in tumors without CD8+ T cells is a blatantly unfavorable prognostic indicator.

    Furthermore, it is known that TAM-derived C-C motif ligand 18 (CCL18) attracts the bulk of naive peripheral blood (PB) CD4+ T cells that differentiate into Tregs in human BC [37]. Though it is unknown how exosomes impact the environment around tumors, the relationship between exosomes and the immune system is vital in initiating and developing malignancies [38]. IL20 receptor subunit alpha (IL20RA) signaling, which explicitly enhances BC stemness and creates an immune milieu favorable to tumor growth, may be involved in forming numerous tumor formations [39]. Due to the tumor cells’ exceptional flexibility in response to extra-tumoral and various metabolic stimuli, the TME is essential [40].

    TAMs, a significant cell population in BC, engage in a particular function that promotes tissue remodeling, angiogenesis, and suppression of adaptive immunity. Additionally, they produce a variety of compounds that aid in the development of tumors, including vascular endothelial growth factor (VEGF), cytokines, and enzymes that encourage angiogenesis, invasion, and metastasis [41]. According to recent research, epigenetically controlled genes mediate the communication between cancer and non-cancer host cells in the TME.

    Metastasis suppressor genes

    The phrase “metastasis suppressor genes” refers to a class of genes that inhibits the ability of cancer cells to spread by concentrating on a critical stage of the invasion process without affecting tumorigenicity. To be precise, metastasis suppressors mainly influence downstream events other than basic tumor formation, whereas tumor suppressors inhibit metastasis by inhibiting primary tumor growth. The first metastasis suppressor, NME/Nm23 nucleoside diphosphate kinase (NDPK) 1 (NME1), was identified in 1988 by comparing the differentially expressed genes in high- and low-metastatic murine melanoma cell lines. Nm23 and Nm23-H1 are other names for NME1 [42]. More than 30 metastasis suppressors have been discovered based on the functional criteria. Both inside the cells and in the extracellular environment, metastasis suppressors have a variety of methods of action, and each one controls a different stage of the metastatic cascade [16, 43]. Not all the metastasis suppressor mechanisms are studied. However, some research suggests that it modulates the TME in various prospects, a potential therapeutic target for treating tumors. This review mainly focuses on some of the metastasis suppressor genes that suppress metastasis either inhibiting migration or colonization. This also includes the primary research of the genes in BC in particular in vitro studies. The activity of the metastasis suppressor gene in different BC cell lines and its effect on the TME is listed in Table 1.

    Metastasis suppressor genes and modulation of TME in BC cell lines

    Metastasis

    suppressor

    genes

    Cell linesEffect on the TMEReferences
    BRMS1

    MDA-231 and

    MDA-435

    Reduced localization of β1 integrin to adhesion-associated cellular protrusions[44]
    KISS1

    MDA-MB-31Br and

    CN34Brm2Ctgl

    Overexpression of epithelial marker E-cadherin[45]
    NM23MDA-MB-435Suppresses MT1-MMP activity[46]
    GSNMCF and MDA-MB-231TGF-β1 signaling leads to epigenetic alterations in gelsolin expression[47]
    CADM1MCF7EMT-induced E-cadherin and CADM1 modifications result in enhanced vulnerability to NK cell cytotoxicity[48]
    KAI1MDA-MB-231Induces apoptosis by the production of intracellular ROS and downregulates EGFR signaling[49, 50]
    NDRG1MCF7 and MDA-MB-435Upregulates E-cadherin and β-catenin by inhibiting SMAD/PSMAD32 and downregulation of TWIST, SNAIL, and SLUG[51, 52]
    RhoGDI2MDA-MB-231D4-GDI knockdown inhibits cell growth and invasion through activating Rac-dependent p38 and JNK signaling[53]
    MAPK14MDA-MB-231, Hs587T, and SUM159Cell migration, induction of EMT, p38 activation, and genomic regulation of NR4A1 expression by cis-acting-catenin/TCF/LEF complexes are all TGF-induced responses[54]
    GAS1MDADecreases endothelial migration leading to decreased vascularization[55]
    Display full size

    BRMS1: BC metastasis suppressor 1; GSN: gelsolin; CADM1: cell adhesion molecule 1; KAI1: kangai1-anticancer; NDRG1: N-myc downstream-regulated gene 1; RhoGDI2: Rho guanine diphosphate-dissociation inhibitor 2; MAPK14: mitogen-activated protein kinase 14; GAS1: growth arrest specific 1; MT1-MMP: membrane type 1 MMP; ROS: reactive oxygen species; EGFR: epidermal growth factor receptor; PSMAD32: SMAD family member; TWIST: Twist family BHLH transcription factor; SNAIL: Snail family transcriptional repressor; D4-GDI: D4-guanine diphosphate-dissociation inhibitor; JNK: c-Jun N-terminal kinase; NR4A1: nuclear receptor subfamily 4 group A member 1; TCF: T cell factor; LEF: lymphoid enhancer factor

    BRMS1

    Human BC and melanoma cell lines’ ability to metastasize is inhibited but not their tumorigenicity by the metastasis suppressor gene known as BRMS1, which is located at chromosome 11q13. This gene produces a protein that belongs to the mammalian switch independent 3 (mSin3a) family of histone deacetylase complexes (HDACs) and is primarily present in the nucleus. Hence it is convincible that the BRMS1 acts as a metastasis suppressor through epigenetic regulation [56]. BRMS1 suppresses metastatic regulation of BC subtypes, including TNBC, ER/PR+, ER, PR+, HER2, and HER2+ [5761]. Since tumor promotion and prevention are delicately balanced by the immune system’s involvement, cellular interactions with the immune system may be impacted by BRMS1 [62, 63]. In the systemic circulation of immunocompromised mice, BRMS1 expression has been shown to drastically diminish the survival of BC cells [64, 65]. The expression of BRMS1 was assessed in cell lines such as MDA-MB-231, MDA-MB-435, MCF-10A, and HCC-1937.

    BRMS1 was shown to be hypermethylated in MDA-MB-231, moderately methylated in HCC-1937 and MDA-MB-435, but unmethylated in MCF-10A when the DNA methylation status of BC cell lines, BC tissues, and associated non-malignant breast tissues was evaluated using major sperm protein (MSP) [66]. BRMS1 induces anoikis by upregulating the expression of pro-apoptotic genes that are associated with cells that are unable to adhere [65]. Cell adherence to the matrix is a crucial phase in determining whether the metastatic cascade is successful or unsuccessful at each stage. The remodeling of the cytoskeleton brought on by cell/matrix interaction is inhibited by BRMS1 expression. In BRMS1-expressing cells, adhesion-associated reductions in myocardin-related transcription factor (MRTF) and serum response factor (SRF) levels occur upon contact with the ECM; both proteins are intricately linked to the dynamic switching of globular (destabilized) to fibrillar (stabilized) actin filaments [67]. The oncogene Cullin3 encourages BC cells’ metastasis and EMT by degrading BRMS1 [68]. By reducing TWIST1 and SNAIL levels, BRMS1 significantly decreased TGF-β1-induced BC cells’ EMT and invasion [69]. JARID1C, a histone demethylase that stimulates the growth of cancer cells and controls transcription and chromatin remodeling, is overexpressed in BC, especially in cases where the disease has spread to distant organs. Increased migration and invasion result from JARID1C modification, suppressing BRMS1 expression by demethylating histone H3 lysine 4 (H3K4) and lowering H3K4me3 at the BRMS1 gene promoter [70]. BRMS1 expression may serve as both a potential biomarker for selecting patients for clinical trials as well as a clinical prognostic indicator, capable of predicting relapse or response to targeted therapy. Recent research reveals that inhibiting the tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) pathway makes cells more susceptible to cell death, suggesting that BRMS1+ BC patients may benefit from TRAIL-targeted therapy [71].

    KISS1

    KISS1 was first identified as a metastasis suppressor, but more recently, it has been found to play various roles in social behavior, reproduction, metabolism, and fertility [72]. It was given the name KISS1 in remembrance of the area where it was discovered—the home of Hershey’s chocolate “Kisses” [73]. In 1996, when human chromosome 6 was introduced to a cancer cell, the lab of Dr. Danny Welch in Hershey, Pennsylvania, isolated a complementary DNA (cDNA) from the cell, preventing metastasis [74]. It can inhibit melanoma and BC metastasis. The KISS1-derived peptides were given the kisspeptins (KPs)—KP54, 14, 13, and 10—following the number of amino acids in the peptide chain. Due to its anti-metastatic properties, one of them, KP54, was initially known as metastin [75]. When blocking KISS1 with a specific short peptide antagonist (p234) impacts TGF-mediated cell invasion and MMP9 synthesis, it is proven that KISS1 plays a vital role in mediating the pro-invasive effects of TGF as a downstream target of the canonical TGF/SMAD2 signaling pathway [76]. MMP9 and other proteases encourage tumor invasion by destroying the ECM. The tumor-suppressive role is thought to prevent tumor invasion by suppressing MMP9 production and activity and MAPK [77]. Maintaining the epithelial state and reducing BC cell invasiveness are two effects of KISS1 signaling through PKD1, which also performs tasks related to its function as a metastasis suppressor. In TNBC cells, melatonin boosted the expression of KISS1, which was mediated through GATA binding protein 3 (GATA3). Melatonin’s inhibition of KISS1 remained as a result [78]. IL30 increased the proliferation, motility, and inflammatory environment in TNBC cells, aiding KISS1-dependent metastasis. Treatment with IL30 in vivo enhanced the expansion of intra-tumoral CD11b+/Gr1+ myeloid cell infiltrates, vascular dissemination, and cancer cell proliferation in TNBC [79]. It was demonstrated that the KISS1 receptor (KISS1R) regulated the expression of the drug efflux transporter BC resistance protein (BCRP), a crucial regulator of the multi-drug resistance phenotype in BC significantly elevated in TNBC, demonstrating that KISS1R-induced drug resistance was dependent on the activity of adenosine triphosphate-binding cassette (ABC) transporters [80]. KISS1 stops the migration of BC cells by inhibiting the NF-κB pathway and RhoA activation that TNF-α causes. Although KP10 did not impact cancer cell proliferation, KISS1 overexpression and KP10 stimulation reduced TNF-α-induced NF-κB activity and suppressed TNF-α-induced cell migration, respectively, and lowered TNF-α-induced cell attachment to fibronectin in BC cells [81]. According to studies on the function of KISS1 in MDA-MB-231 and MDA-MB-157 cells, the suppression of MMP9 and MMP2 activity is linked to the reduction of metastasis caused by KP [82]. NF-κB p50 and MMP9 expression were discovered to be adversely linked with the expression of the KISS1-1 protein [83]. According to some studies, the stromal cell-derived factor-1 (SDF-1)/C-X-C chemokine receptor type 4 (CXCR4) system may be crucial for BC invasion and EMT. KP-10 therapy inhibits CXCR4 expression to decrease invasion, and EMT brought on by SDF-1 [84]. A recent study found that TGF-β1, but not SMAD, inhibits human trophoblast cell invasion by increasing KP synthesis through the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway [85].

    Non-metastatic 23

    The non-metastatic (Nm) protein 23 H1 is the molecular name for the common enzyme (NDPK A, Nm23-H1). Steeg et al. [42, 86] initially discovered this metastasis suppressor protein. The human Nnm23 genes Nm23-H1 and Nm23-H2 have been located on chromosome 17q21. The phosphorylation of Nm23/NME histidine 118, which is implicated in the activities of the enzymes NDPK and histidine protein kinase (HPK), has received considerable attention. The processes through which Nm23 inhibits metastasis and motility still need to be better understood [87]. Loss of heterozygosity, spontaneous mutations, and polymorphisms in the Nm23 gene are rarely found in malignancies; as a result, Nm23 protein levels are likely to impact the tumor cells’ ability to metastasize [88]. Previously, in the human MDA-MB-435 (BC) and K-1735 TK (melanoma) cell lines, the overexpression of Nm23 was linked to a reduction in tumor-spreading potential [89]. EMT, triggered by stress caused by tumor hypoxia and growth factor depletion, leads to invasion and metastasis and is linked to CSCs characteristics. In Panc-1/MDA-MB-231 cells, serum starvation and hypoxia reduced the expression of the prototypical Nm23-H1 [90]. In past investigations, the less aggressive MCF-7 cells, which express much of Nm23-H1, and the invasive MDA-MB-231 cells, which express little to no Nm23-H1, demonstrated a relationship between messenger RNA (mRNA) levels and protein expression. This implies that transcriptional processes might control the expression of Nm23-H1. Several transcription factors, including CCCTC-binding factor (CTCF) and early growth response factor 1 (EGR1), can influence the transcription of NME1. This might either result in BC cells expressing more Nm23-H1, which would promote a phenotype that is less likely to spread, or it could result in Nm23-H1 production being reduced [91]. Treatment with an NDPK A and B inhibitor or a purinoreceptor antagonist attenuates the effects of MDA-MB-231 extra vesicles on the mice’s pulmonary vascular leakage and experimental lung metastases [92]. An additional experiment demonstrates that NME1 (Nm23-H1) was found to be the primary component of these exosomes when it was transfected into exosomes formed from two cancer cell lines (MDA-MB-231T and MDA-MB-435). These exosomes altered the endocytic pathways of receiving tumor cells via NME1, decreasing their motility and migration in vitro more than exosomes from control transfectants [93]. Invasive breast carcinomatous lesions exhibit a highly anti-correlated expression of NME1 and cortical MT1-MMP, according to RNA sequencing (RNAseq) data [46].

    GSN

    GSN is the first tumor suppressor gene in human basal carcinoma to be impacted by histone acetylation [94]. The actin-binding protein GSN plays a crucial role in controlling the formation and disassembly of actin filaments. Additionally, it was revealed that specific tumor cells had lower cellular levels of GSN and that overexpressing GSN by gene transfer reduces tumorigenicity. GSN’s carboxyl terminus is crucial for suppressing metastasis, reducing chemotaxis, and delaying cell spreading [95]. In MCF-7 cells, down-regulating GSN resulted in a concentration-dependent increase in migratory activity and vice versa [96]. GSN’s epigenetic alteration by TGF-β1 may affect the EMT process in BC cells [47]. Results from gel shift and supershift assays, Southwestern blotting studies, and gel shift and supershift assays suggest that the 27-bp GSN cis-element is preferentially bindable by activating transcription factor 1 (ATF1) in cancer cells. This shows that ATF1 is involved in silencing the GSN promoter, as opposed to its transactivating effect on other types of promoters [97]. The study suggests that GSN is downregulated through epigenetic processes.

    Furthermore, the more significant role in this repression is played by hypoacetylation of histones as opposed to hypermethylation of regulatory cytosine-guanine (CpG) sites [94]. Rodent and human BCs share a common dysregulation of cyclin D1 and GSN, and it appears that, at least in part, a changed transcription rate is responsible for both genes’ malfunction [98]. On the other hand, several studies assert that the invasive motile characteristics of cells and cell aggregation are considerably diminished in various cancer cell types, including MDA-MB-231 and PC-3 cells when CapG or GSN is downregulated. These findings suggest that GSN and CapG may operate as tumor activators [99]. Thus, further research needs to be done to understand the role of GSN clearly.

    CADM1

    CADM1 or Tslc1, Necl2, Ra175, IgSF4a, and SynCAM, a transmembrane protein member of the immunoglobulin superfamily is predominantly implicated in cell-cell interactions [100]. TSLC1 is highlighted as a tumor suppressor in lung adenocarcinoma [101], but recent studies have highlighted the metastatic suppressive properties of CADM1 in breast carcinomas [102].

    From a macro-perspective, higher clinical presentations of CADM1 have been correlated with tumor size and staging [103, 104]. Studies have co-related poor patient survival rates due to hypermethylation of the promoter region of the CADM1 gene leading to transcriptional repression in metastatic BC. At the same time, very little research has gone into understanding the molecular aspects of its mechanisms [105108]. Research studies have implicated CADM1’s protective role in several stages of the metastatic cascade in BC [109] and ovarian cancer [110].

    A study conducted by Faraji et al. [102] explored the anti-metastatic and anti-proliferative roles of CADM1 in association with components involved in immunosurveillance in the TME. Expression of CADM1 in metastatic murine mammary cell lines suppressed metastasis without affecting primary tumorigenesis. At the same time, the scratch wound assay demonstrated a significant reduction in the relative motility of CADM1+ BC cells compared to the control specimen [102]. Furthermore, when CD8+ T cells were reduced in immune-competent mice, the metastasis-suppressing impact of CADM1 was partially phenocopied in mice lacking T cell-mediated immunity [102].

    Cancer development and progression are impacted by cell cytotoxicity and tumor rejection caused by CD8+ T cell-mediated immunity and NK cells [111]. Class-I restricted T cell adhesion molecule (Crtam) has been shown to interact with CADM1 and regulate immunoediting processes by generating interferon, which may stimulate pro-inflammatory signals to induce cytotoxicity. It is a crucial biomarker of activated CD8+ T cells and NK cells [112, 113]. The hypothesis was tested by Faraji et al. [102] and team and a significant increase in interferon-γ levels from the lymph nodes of mice comprising cadm1 expressing neoplasms was observed. Reports also show NK-mediated cytotoxicity depends on the balance of cellular E-cadherin (a noted marker for EMT) and CADM1 owing to homophilic interactions mediated by membrane-associated guanylate kinases and CADM1’s C-terminal intracellular domain [114, 115].

    CADM11 has much potential to be targeted for therapeutic applications in invasive late-stage cancers owing to the co-relation between non-coding RNA expression and CADM1 concentration. Several reports indicate the down-regulation of CADM1 due to the expression of long non-coding (lncRNA) and miR. A study conducted by Zhang et al. [103] conclusively demonstrated the effects of miR-155-3p associated with the downregulation of CADM1, leading to tumor progression and metastasis, and apoptosis. Hence, CADM1 looks to be a promising and potential target to treat multiple late-stage metastatic cancers.

    CD82

    KAI1/CD82 was first identified in T-cell activation research and is a type III transmembrane glycoprotein member of the tetraspanin protein family. It is also known as R2, C33, IA4, or 4F9 [116, 117]. Studies identified the KAI1 gene on human chromosome 11, which comprises ten exons and nine introns [118]. Tetraspanins are proteins on the cell membrane consisting of 4 putative domains, N-terminal and C-terminal, and small and large extracellular domains. Overall functional activity of the KAI1 protein is influenced by its domains where glycosylation of its N-terminal is crucial for adhesion molecules and cell surface receptors. At the same time, polar molecules spanning the cellular membrane are associated with the maintenance of protein conformation and cell migration, invasion, and metastasis [119121].

    The metastatic cascade involves various steps such as intravasation, cell migration, extravasation, and colonization [122]. KAI1 protein is known to down-regulate intercellular adhesion processes and prevent the colonization of metastatic cancer cells [123]. The alteration of cell adhesion factors as such in the TME regulates the migratory behavior and metastatic potential of cancer cells. Overexpression of KAI1/CD82 induces apoptosis via the production of intracellular ROS, which is thought to be the downstream product of the release of intracellular anti-oxidant glutathione [124].

    In vitro, studies that established CD82’s anti-metastatic function specifically in mammary tumor cells and found that knocking down CD82 in highly metastatic MDA-MB-231 BC cells promoted functional inactivation of MAPK pathway and EGFR signaling, which increased cell migration and invasiveness, were found to be correlated with the metastatic suppressive effects of KAI1/CD82 [49, 50]. Studies by He et al. [125] and Odintsova et al. [126] reveal that CD82 slows the downregulation of EGFR signaling in the tetraspanin web which describes the lateral interactions between tetraspanins and their partners in the cell membrane and is crucial for developing tumor cells. Multiple reports indicate KAI1/CD82 interacts with tetraspanin protein molecules such as: (A) CD9 and CD81 [127, 128]; (B) β1 and β2 integrins that lead to the formation of p130CAS–Crk complex, thus regulating inhibition of cell migration [129133]; (C) major histocompatibility complex (MHC) class I and II molecules for immunomodulation in the TME [133, 134]; (D) heparin-binding epidermal growth factor [135] desensitizes epidermal growth factor signaling via endocytosis of EGFR to suppress metastasis [126]; (E) intracellular signaling molecules such as protein kinase C recruit the integrin-tetraspanin protein complex to regulate cell motility and migration [136] and are also connected to the urokinase-type plasminogen activator receptor (uPAR)’s intracellular dispersion into focal adhesions, which is mediated by the α5β1 integrins [137].

    NDRG1

    NDRG1 is a member of the 4 NDRG gene family determined to suppress oncogenesis and tumor progression in different cancers such as breast, prostate, brain, colon, pancreas, and rectum [138, 139]. Contradictory evidence also suggests the oncogenic effects of NDRG1 in kidney and liver cancers [138].

    The mature protein is said to be primarily located in the cytoplasm but is also dependent on cell type; for instance, NDRG1 is predominantly found in the plasma membrane of lactating mammary epithelial cells, the inner membrane of the mitochondria in cells lining the proximal renal tubule and nucleus of prostatic epithelial cells [138, 139]. NDRG1 has been to said disrupt several signaling pathways as a pivotal response to regulate cell proliferation, cell-cell adhesion, and motility under stress conditions like hypoxia and reoxygenation and also have varying effects on the TME [138, 140142].

    Several chemical and biological signals regulate NDRG1 concentration in the cell [140]. Epigenetic modifications to the NDRG1 gene can lead to reduced expression. In contrast, several factors can increase expressions, such as heavy metal ions, iron depletion, hypoxia-inducible factor 1, and DNA-damaging agents [138, 139, 143]. It is also said to decrease the expression of p53 under hypoxic conditions, thus preventing apoptosis [144]. Various reports have linked the expression of NDRG1 with suppressing tumor growth and development [51, 52, 145]. It mediates the upregulation of several epithelial markers like E-cadherin and β-catenin and downregulates mesenchymal phenotypes occurring in EMT via inhibiting SMAD/PSMAD32 expression and downregulating SNAIL, SLUG, and TWIST expression to suppress cell migration and metastasis [51, 52]. NDRG1 expression in cancer cells is also associated with inhibition in β-catenin phosphorylation at Ser33/37 and Thr41 residues which protect the cell from β-catenin, a widely known pro-oncogenic transcription factor in the Wnt signaling pathway and thus suppresses cancer metastasis [52, 146]. This statement is backed up by a study that showed suppressed metastasis in mouse mammary cancer models [147]. Reports also indicated decreased expression of ErbB receptors such as EGFR, HER2, and HER3 to NDRG1 expression preventing the formation of HER2/HER3 dimers and disrupting major oncogenic pathways primarily associated with mammary tumors [145, 148]. Stabilizing the RhoGDI-cell division control (CDC42) complex, which produces cell protuberances at the cell’s leading edge in colorectal cancer cell lines, is another mechanism through which NDRG1 is connected to increased invasiveness [142]. Hence, metastatic suppressive activities at the molecular level are observed with NDRG1 expression and can be further explored.

    However, overwhelming contradictory evidence links the expression of NDRG1 with oncogenic effects in aggressive BCs [149152]. A study correlated increased expression of NDRG1 with advanced tumor progression, thus highlighting it as an important prognostic biomarker in invasive BC [152]. Hence, NDRG1 needs to be further explored and studied to develop into a novel therapeutic agent against cancer. A summary of molecular interaction between these metastatic suppressors and chemical components of the TME has been given in Figure 2.

    Molecular interactions between metastatic suppressors and chemical components of the TME. This figure was created with BioRender

    Conclusions

    Due to a greater understanding of the molecular process behind the initiation and progression of cancer, targeted cancer therapies have achieved tremendous advancements in recent decades [153]. Many biological functions in the stage of metastasis are controlled by metastasis suppressor genes [154]. An increased chance of abnormal cell proliferation, differentiation, cell death, and cancer development exists when metastasis suppressor genes are not functioning properly. The downregulation of metastasis suppressor genes has been discovered in various cancers like breast, pancreatic, ovarian, bladder, colorectal, neck, uterine, and lung cancer. One of the reasons for this downregulation can be the modulation in the TME such as lymphocytes, CAF, TAM, ECM, exosomes, CTCs, etc. New delivery techniques and more sophisticated gene expression systems are being studied as potential cancer treatments and cures. Lack of tumor selectivity is one of the problems with current cancer treatments. Therefore, one of the main goals of cancer gene therapy is to specifically target tumor cell gene expression [155]. The classification of the BC subtypes, specifically HR+/HER2+, HER2+, and HR+/HER2, determines the majority of treatment plans. Local treatments for Nm BCs include surgery (which may need axillary lymph node removal) and radiotherapy [156]. Different directed gene therapy strategies have been created employing tumor-specific promoters to treat BC [157]. After screening a library of recombinant secreted microenvironmental proteins, fibroblast growth factor 2 (FGF2) was discovered to be an essential regulator of anti-estrogen resistance, mechanistic target of rapamycin complex 1 (mTORC1) inhibition, and phosphatidylinositol 3-kinase inhibition in ER+ BC [158]. When used in conjunction with oncolytic virotherapy for the treatment of breast and other metastatic cancers, KISS1 transgenic expression has the potential to be an efficient tool for CRAd-mediated cytotoxicity in BC cells [159]. Additionally, it is true that customized care and precision medicine, which are directed at specific patients and patient subgroups, are gaining popularity in oncology, where the focus is on delaying the onset of the disease and the treatment plan is devised to maximize effectiveness while minimizing side effects [160]. lncRNA-based therapy and miR therapeutics are the other treatments for BC based on epigenetic drugs by which metastasis suppressor genes are epigenetically inactivated [156]. Further studies about the TME and the metastasis suppressor genes need to be done to recognize their exact role in tumors. The TME contributes to the growth and invasion of cancer cells and their components help the cells for migration. When suppressor genes enter the scene to inhibit the metastatic process of tumor cells, what role do the components of the TME play? Will the interaction of these components and genes make any remarkable changes in the metastatic cascade? If so, then how can this be used as a potential target for therapies? To answer these questions, a detailed review has been done to study the relationship between the TME and the metastasis suppressor genes in BC. Some of the metastasis suppressor genes are downregulated which leads to metastasis in several tumors. Some studies suggest that modulation in the TME is associated with cancer progression and proliferation. Hence, understanding the role of each metastasis suppressor gene in the TME is important in order to of them as a potential therapeutic target.

    Abbreviations

    ATF-1:

    activating transcription factor-1

    BC:

    breast cancer

    BRMS1:

    breast cancer metastasis suppressor 1

    CADM1:

    cell adhesion molecule 1

    CAFs:

    cancer-associated fibroblasts

    CD4+:

    cluster of differentiation 4+

    CXCR4:

    C-X-C chemokine receptor type 4

    D4-GDI:

    D4-guanine diphosphate-dissociation inhibitor

    ECM:

    extracellular matrix

    EGFR:

    epidermal growth factor receptor

    EMT:

    epithelial-mesenchymal transition

    ERα:

    estrogen receptor alpha

    GSN:

    gelsolin

    HER2:

    human epidermal growth factor 2

    IL32:

    interleukin 32

    KAI1:

    kangai1-anticancer

    KISS1R:

    KISS1 receptor

    KPs:

    kisspeptins

    MAPK:

    mitogen-activated protein kinase

    miR-18b:

    microRNA-18b

    MMP13:

    matrix metalloproteinase 13

    MT1-MMP:

    membrane type 1 matrix metalloprotease

    NDPK:

    nucleoside diphosphate kinase

    NDRG1:

    N-myc downstream-regulated gene 1

    NF-κB:

    nuclear factor-kappa B

    NK:

    natural killer

    Nm:

    non-metastatic

    NME1:

    NME/Nm23 nucleoside diphosphate kinase 1

    PR:

    progesterone receptor

    SNAIL:

    Snail family transcriptional repressor

    TAMs:

    tumor-associated macrophages

    TGF-β1:

    transforming growth factor-β1

    TME:

    tumor microenvironment

    TNBC:

    triple-negative breast cancer

    TNF:

    tumor necrosis factor

    Treg:

    regulatory T

    TWIST:

    Twist family BHLH transcription factor

    Declarations

    Author contributions

    SSS: Conceptualization, Methodology, Investigation, Writing—original draft, Writing—review & editing. SD: Methodology, Investigation, Software, Writing—original draft, Visualization. SH: Conceptualization, Visualization, Writing—review & editing, Project administration, Supervision.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2023.

    References

    Weigelt B, Geyer FC, Reis-Filho JS. Histological types of breast cancer: how special are they? Mol Oncol. 2010;4:192208. [DOI] [PubMed] [PMC]
    Sun YS, Zhao Z, Yang ZN, Xu F, Lu HJ, Zhu ZY, et al. Risk factors and preventions of breast cancer. Int J Biol Sci. 2017;13:138797. [DOI] [PubMed] [PMC]
    Niell BL, Freer PE, Weinfurtner RJ, Arleo EK, Drukteinis JS. Screening for breast cancer. Radiol Clin North Am. 2017;55:114562. [PubMed]
    Łukasiewicz S, Czeczelewski M, Forma A, Baj J, Sitarz R, Stanisławek A. Breast cancer—epidemiology, risk factors, classification, prognostic markers, and current treatment strategies—an updated review. Cancers (Basel). 2021;13:4287. [DOI] [PubMed] [PMC]
    Collaborative Group on Hormonal Factors in Breast Cancer. Familial breast cancer: collaborative reanalysis of individual data from 52 epidemiological studies including 58,209 women with breast cancer and 101,986 women without the disease. Lancet. 2001;358:138999. [DOI] [PubMed]
    Shiovitz S, Korde LA. Genetics of breast cancer: a topic in evolution. Ann Oncol. 2015;26:12919. [DOI] [PubMed] [PMC]
    Plasilova ML, Hayse B, Killelea BK, Horowitz NR, Chagpar AB, Lannin DR. Features of triple-negative breast cancer: analysis of 38,813 cases from the national cancer database. Medicine (Baltimore). 2016;95:e4614. [DOI] [PubMed] [PMC]
    Newman LA, Reis-Filho JS, Morrow M, Carey LA, King TA. The 2014 Society of Surgical Oncology Susan G. Komen for the Cure Symposium: triple-negative breast cancer. Ann Surg Oncol. 2015;22:87482. [DOI] [PubMed]
    Mego M, Mani SA, Cristofanilli M. Molecular mechanisms of metastasis in breast cancer—clinical applications. Nat Rev Clin Oncol. 2010;7:693701. [DOI] [PubMed]
    Minn AJ, Kang Y, Serganova I, Gupta GP, Giri DD, Doubrovin M, et al. Distinct organ-specific metastatic potential of individual breast cancer cells and primary tumors. J Clin Invest. 2005;115:4455. [DOI] [PubMed] [PMC]
    Weigelt B, Peterse JL, van ’t Veer LJ. Breast cancer metastasis: markers and models. Nat Rev Cancer. 2005;5:591602. [DOI] [PubMed]
    Scully OJ, Bay BH, Yip G, Yu Y. Breast cancer metastasis. Cancer Genomics Proteomics. 2012;9:31120. [PubMed]
    Mehraj U, Dar AH, Wani NA, Mir MA. Tumor microenvironment promotes breast cancer chemoresistance. Cancer Chemother Pharmacol. 2021;87:14758. [DOI] [PubMed]
    Bahrami A, Hassanian SM, Khazaei M, Hasanzadeh M, Shahidsales S, Maftouh M, et al. The therapeutic potential of targeting tumor microenvironment in breast cancer: rational strategies and recent progress. J Cell Biochem. 2018;119:11122. [DOI] [PubMed]
    Yan J, Yang Q, Huang Q. Metastasis suppressor genes. Histol Histopathol. 2013;28:28592. [DOI] [PubMed] [PMC]
    Cook LM, Hurst DR, Welch DR. Metastasis suppressors and the tumor microenvironment. Semin Cancer Biol. 2011;21:11322. [DOI] [PubMed] [PMC]
    Vaidya KS, Welch DR. Metastasis suppressors and their roles in breast carcinoma. J Mammary Gland Biol Neoplasia. 2007;12:17590. [DOI] [PubMed] [PMC]
    Anderson NM, Simon MC. The tumor microenvironment. Curr Biol. 2020;30:R9215. [DOI] [PubMed] [PMC]
    Baghban R, Roshangar L, Jahanban-Esfahlan R, Seidi K, Ebrahimi-Kalan A, Jaymand M, et al. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal. 2020;18:59. [DOI] [PubMed] [PMC]
    Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21:30922. [DOI] [PubMed]
    Whiteside TL. The tumor microenvironment and its role in promoting tumor growth. Oncogene. 2008;27:590412. [DOI] [PubMed] [PMC]
    Hui L, Chen Y. Tumor microenvironment: sanctuary of the devil. Cancer Lett. 2015;368:713. [DOI] [PubMed]
    Allen M, Louise Jones J. Jekyll and Hyde: the role of the microenvironment on the progression of cancer. J Pathol. 2011;223:16276. [DOI] [PubMed]
    Folgueira MAAK, Maistro S, Katayama MLH, Roela RA, Mundim FGL, Nanogaki S, et al. Markers of breast cancer stromal fibroblasts in the primary tumour site associated with lymph node metastasis: a systematic review including our case series. Biosci Rep. 2013;33:e00085. [DOI] [PubMed] [PMC]
    Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121:33548. [DOI] [PubMed]
    Wen S, Hou Y, Fu L, Xi L, Yang D, Zhao M, et al. Cancer-associated fibroblast (CAF)-derived IL32 promotes breast cancer cell invasion and metastasis via integrin β3-p38 MAPK signalling. Cancer Lett. 2019;442:32032. [DOI] [PubMed]
    Ren J, Smid M, Iaria J, Salvatori DCF, van Dam H, Zhu HJ, et al. Cancer-associated fibroblast-derived Gremlin 1 promotes breast cancer progression. Breast Cancer Res. 2019;21:109. [DOI] [PubMed] [PMC]
    Yan Z, Sheng Z, Zheng Y, Feng R, Xiao Q, Shi L, et al. Cancer-associated fibroblast-derived exosomal miR-18b promotes breast cancer invasion and metastasis by regulating TCEAL7. Cell Death Dis. 2021;12:1120. [DOI] [PubMed] [PMC]
    Chen B, Sang Y, Song X, Zhang D, Wang L, Zhao W, et al. Exosomal miR-500a-5p derived from cancer-associated fibroblasts promotes breast cancer cell proliferation and metastasis through targeting USP28. Theranostics. 2021;11:393247. [DOI] [PubMed] [PMC]
    Choi YP, Lee JH, Gao MQ, Kim BG, Kang S, Kim SH, et al. Cancer-associated fibroblast promote transmigration through endothelial brain cells in three-dimensional in vitro models. Int J Cancer. 2014;135:202433. [DOI] [PubMed]
    Avalle L, Raggi L, Monteleone E, Savino A, Viavattene D, Statello L, et al. STAT3 induces breast cancer growth via ANGPTL4, MMP13 and STC1 secretion by cancer associated fibroblasts. Oncogene. 2022;41:145667. [DOI] [PubMed]
    Denkert C, von Minckwitz G, Brase JC, Sinn BV, Gade S, Kronenwett R, et al. Tumor-infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2-positive and triple-negative primary breast cancers. J Clin Oncol. 2015;33:98391. [DOI] [PubMed]
    Shohdy KS, Almeldin DS, Ghaly R, Kassem L, Pagani O. Prognostic impact of cytotoxic CD4 T cells in tumor immune microenvironment of patients with breast cancer. J Immunother Precis Oncol. 2021;5:79. [DOI] [PubMed] [PMC]
    Soysal SD, Tzankov A, Muenst SE. Role of the tumor microenvironment in breast cancer. Pathobiology. 2015;82:14252. [DOI] [PubMed]
    Malla RR, Vasudevaraju P, Vempati RK, Rakshmitha M, Merchant N, Nagaraju GP. Regulatory T cells: their role in triple-negative breast cancer progression and metastasis. Cancer. 2022;128:117183. [DOI] [PubMed]
    Jiang X. Harnessing the immune system for the treatment of breast cancer. J Zhejiang Univ Sci B. 2014;15:115. [DOI] [PubMed] [PMC]
    Su S, Liao J, Liu J, Huang D, He C, Chen F, et al. Blocking the recruitment of naive CD4+ T cells reverses immunosuppression in breast cancer. Cell Res. 2017;27:46182. [DOI] [PubMed] [PMC]
    Qiu P, Guo Q, Yao Q, Chen J, Lin J. Characterization of exosome-related gene risk model to evaluate the tumor immune microenvironment and predict prognosis in triple-negative breast cancer. Front Immunol. 2021;12:736030. [DOI] [PubMed] [PMC]
    Gao W, Wen H, Liang L, Dong X, Du R, Zhou W, et al. IL20RA signaling enhances stemness and promotes the formation of an immunosuppressive microenvironment in breast cancer. Theranostics. 2021;11:256480. [DOI] [PubMed] [PMC]
    Tang Y, Tian W, Xie J, Zou Y, Wang Z, Li N, et al. Prognosis and dissection of immunosuppressive microenvironment in breast cancer based on fatty acid metabolism-related signature. Front Immunol. 2022;13:843515. [DOI] [PubMed] [PMC]
    Solinas G, Germano G, Mantovani A, Allavena P. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. J Leukoc Biol. 2009;86:106573. [DOI] [PubMed]
    Hartsough MT, Steeg PS. Nm23-H1: genetic alterations and expression patterns in tumor metastasis. Am J Hum Genet. 1998;63:610. [DOI] [PubMed] [PMC]
    Yoshida BA, Sokoloff MM, Welch DR, Rinker-Schaeffer CW. Metastasis-suppressor genes: a review and perspective on an emerging field. J Natl Cancer Inst. 2000;92:171730. [DOI] [PubMed]
    Khotskaya YB, Beck BH, Hurst DR, Han Z, Xia W, Hung MC, et al. Expression of metastasis suppressor BRMS1 in breast cancer cells results in a marked delay in cellular adhesion to matrix. Mol Carcinog. 2014;53:101126. [DOI] [PubMed] [PMC]
    Ulasov I, Borovjagin A, Fares J, Yakushov S, Malin D, Timashev P, et al. MicroRNA 345 (miR345) regulates KISS1-E-cadherin functional interaction in breast cancer brain metastases. Cancer Lett. 2020;481:2431. [DOI] [PubMed]
    Lodillinsky C, Fuhrmann L, Irondelle M, Pylypenko O, Li XY, Bonsang-Kitzis H, et al. Metastasis-suppressor NME1 controls the invasive switch of breast cancer by regulating MT1-MMP surface clearance. Oncogene. 2021;40:401932. [DOI] [PubMed] [PMC]
    Chen ZY, Wang PW, Shieh DB, Chiu KY, Liou YM. Involvement of gelsolin in TGF-beta 1 induced epithelial to mesenchymal transition in breast cancer cells. J Biomed Sci. 2015;22:90. [DOI] [PubMed] [PMC]
    Chockley PJ, Chen J, Chen G, Beer DG, Standiford TJ, Keshamouni VG. Epithelial-mesenchymal transition leads to NK cell-mediated metastasis-specific immunosurveillance in lung cancer. J Clin Invest. 2018;128:138496. [DOI] [PubMed] [PMC]
    Malik FA, Sanders AJ, Kayani MA, Jiang WG. Effect of expressional alteration of KAI1 on breast cancer cell growth, adhesion, migration and invasion. Cancer Genomics Proteomics. 2009;6:20513. [PubMed]
    Odintsova E, Voortman J, Gilbert E, Berditchevski F. Tetraspanin CD82 regulates compartmentalisation and ligand-induced dimerization of EGFR. J Cell Sci. 2003;116:455766. [DOI] [PubMed]
    Lane DJR, Mills TM, Shafie NH, Merlot AM, Saleh Moussa R, Kalinowski DS, et al. Expanding horizons in iron chelation and the treatment of cancer: role of iron in the regulation of ER stress and the epithelial-mesenchymal transition. Biochim Biophys Acta. 2014;1845:16681. [DOI] [PubMed]
    Chen Z, Zhang D, Yue F, Zheng M, Kovacevic Z, Richardson DR. The iron chelators Dp44mT and DFO inhibit TGF-β-induced epithelial-mesenchymal transition via up-regulation of N-Myc downstream-regulated gene 1 (NDRG1). J Biol Chem. 2012;287:1701628. [DOI] [PubMed] [PMC]
    Zhang Y, Rivera Rosado LA, Moon SY, Zhang B. Silencing of D4-GDI inhibits growth and invasive behavior in MDA-MB-231 cells by activation of Rac-dependent p38 and JNK signaling. J Biol Chem. 2009;284:1295665. [DOI] [PubMed] [PMC]
    Hedrick E, Safe S. Transforming growth factor β/NR4A1-inducible breast cancer cell migration and epithelial-to-mesenchymal transition is p38α (mitogen-activated protein kinase 14) dependent. Mol Cell Biol. 2017;37:e0030617. [DOI] [PubMed] [PMC]
    Jiménez A, López-Ornelas A, Estudillo E, González-Mariscal L, González RO, Segovia J. A soluble form of GAS1 inhibits tumor growth and angiogenesis in a triple negative breast cancer model. Exp Cell Res. 2014;327:30717. [DOI] [PubMed]
    Meehan WJ, Samant RS, Hopper JE, Carrozza MJ, Shevde LA, Workman JL, et al. Breast cancer metastasis suppressor 1 (BRMS1) forms complexes with retinoblastoma-binding protein 1 (RBP1) and the mSin3 histone deacetylase complex and represses transcription. J Biol Chem. 2004;279:15629. [DOI] [PubMed]
    Zhang Z, Yamashita H, Toyama T, Yamamoto Y, Kawasoe T, Iwase H. Reduced expression of the breast cancer metastasis suppressor 1 mRNA is correlated with poor progress in breast cancer. Clin Cancer Res. 2006;12:64104. [DOI] [PubMed]
    Hicks DG, Yoder BJ, Short S, Tarr S, Prescott N, Crowe JP, et al. Loss of breast cancer metastasis suppressor 1 protein expression predicts reduced disease-free survival in subsets of breast cancer patients. Clin Cancer Res. 2006;12:67028. [DOI] [PubMed] [PMC]
    Frolova N, Edmonds MD, Bodenstine TM, Seitz R, Johnson MR, Feng R, et al. A shift from nuclear to cytoplasmic breast cancer metastasis suppressor 1 expression is associated with highly proliferative estrogen receptor-negative breast cancers. Tumour Biol. 2009;30:14859. [DOI] [PubMed] [PMC]
    Lin L, Cai M, Dai Y, Zheng Z, Jiang F, Shi L, et al. Breast cancer metastasis suppressor gene, breast cancer metastasis suppressor 1, may be associated with clinicopathological features of breast cancer. J Cancer Res Ther. 2018;14:S36874. [DOI] [PubMed]
    Qiu R, Shi H, Wang S, Leng S, Liu R, Zheng Y, et al. BRMS1 coordinates with LSD1 and suppresses breast cancer cell metastasis. Am J Cancer Res. 2018;8:203045. [PubMed] [PMC]
    Janssen LME, Ramsay EE, Logsdon CD, Overwijk WW. The immune system in cancer metastasis: friend or foe? J Immunother Cancer. 2017;5:79. [DOI] [PubMed] [PMC]
    Zimmermann RC, Welch DR. BRMS1: a multifunctional signaling molecule in metastasis. Cancer Metastasis Rev. 2020;39:75568. [DOI] [PubMed] [PMC]
    Hedley BD, Vaidya KS, Phadke P, MacKenzie L, Dales DW, Postenka CO, et al. BRMS1 suppresses breast cancer metastasis in multiple experimental models of metastasis by reducing solitary cell survival and inhibiting growth initiation. Clin Exp Metastasis. 2008;25:72740. [DOI] [PubMed]
    Phadke PA, Vaidya KS, Nash KT, Hurst DR, Welch DR. BRMS1 suppresses breast cancer experimental metastasis to multiple organs by inhibiting several steps of the metastatic process. Am J Pathol. 2008;172:80917. [DOI] [PubMed] [PMC]
    Kong B, Lv ZD, Wang Y, Jin LY, Ding L, Yang ZC. Down-regulation of BRMS1 by DNA hypermethylation and its association with metastatic progression in triple-negative breast cancer. Int J Clin Exp Pathol. 2015;8:1107683. [PubMed] [PMC]
    Rey M, Irondelle M, Waharte F, Lizarraga F, Chavrier P. HDAC6 is required for invadopodia activity and invasion by breast tumor cells. Eur J Cell Biol. 2011;90:12835. [DOI] [PubMed]
    Huo X, Li S, Shi T, Suo A, Ruan Z, Guo H, et al. Cullin3 promotes breast cancer cells metastasis and epithelial-mesenchymal transition by targeting BRMS1 for degradation. Oncotarget. 2015;6:4195975. [DOI] [PubMed] [PMC]
    Cho KH, Yu SL, Cho DY, Park CG, Lee HY. Breast cancer metastasis suppressor 1 (BRMS1) attenuates TGF-β1-induced breast cancer cell aggressiveness through downregulating HIF-1α expression. BMC Cancer. 2015;15:829. [DOI] [PubMed] [PMC]
    Wang Q, Wei J, Su P, Gao P. Histone demethylase JARID1C promotes breast cancer metastasis cells via down regulating BRMS1 expression. Biochem Biophys Res Commun. 2015;464:65966. [DOI] [PubMed]
    Lombardi G, Di Cristofano C, Capodanno A, Iorio MC, Aretini P, Isola P, et al. High level of messenger RNA for BRMS1 in primary breast carcinomas is associated with poor prognosis. Int J Cancer. 2007;120:116978. [DOI] [PubMed]
    Harihar S, Ray S, Narayanan S, Santhoshkumar A, Ly T, Welch DR. Role of the tumor microenvironment in regulating the anti-metastatic effect of KISS1. Clin Exp Metastasis. 2020;37:20923. [DOI] [PubMed] [PMC]
    Beck BH, Welch DR. The KISS1 metastasis suppressor: a good night kiss for disseminated cancer cells. Eur J Cancer. 2010;46:12839. [DOI] [PubMed] [PMC]
    Lee JH, Miele ME, Hicks DJ, Phillips KK, Trent JM, Weissman BE, et al. KiSS-1, a novel human malignant melanoma metastasis-suppressor gene. J Natl Cancer Inst. 1996;88:17317. [DOI] [PubMed]
    Kirby HR, Maguire JJ, Colledge WH, Davenport AP. International Union of Basic and Clinical Pharmacology. LXXVII. Kisspeptin receptor nomenclature, distribution, and function. Pharmacol Rev. 2010;62:56578. [DOI] [PubMed] [PMC]
    Ulasov IV, Borovjagin AV, Timashev P, Cristofanili M, Welch DR. KISS1 in breast cancer progression and autophagy. Cancer Metastasis Rev. 2019;38:493506. [DOI] [PubMed] [PMC]
    Guzman S, Brackstone M, Wondisford F, Babwah AV, Bhattacharya M. KISS1/KISS1R and breast cancer: metastasis promoter. Semin Reprod Med. 2019;37:197206. [DOI] [PubMed]
    Kim TH, Cho SG. Melatonin-induced KiSS1 expression inhibits triple-negative breast cancer cell invasiveness. Oncol Lett. 2017;14:25116. [DOI] [PubMed] [PMC]
    Airoldi I, Cocco C, Sorrentino C, Angelucci D, Di Meo S, Manzoli L, et al. Interleukin-30 promotes breast cancer growth and progression. Cancer Res. 2016;76:621829. [DOI] [PubMed]
    Blake A, Dragan M, Tirona RG, Hardy DB, Brackstone M, Tuck AB, et al. G protein-coupled KISS1 receptor is overexpressed in triple negative breast cancer and promotes drug resistance. Sci Rep. 2017;7:46525. [DOI] [PubMed] [PMC]
    Cho SG, Li D, Stafford LJ, Luo J, Rodriguez-Villanueva M, Wang Y, et al. KiSS1 suppresses TNFα-induced breast cancer cell invasion via an inhibition of RhoA-mediated NF-κB activation. J Cell Biochem. 2009;107:113949. [DOI] [PubMed] [PMC]
    Song GQ, Zhao Y. Kisspeptin-10 inhibits the migration of breast cancer cells by regulating epithelial-mesenchymal transition. Oncol Rep. 2015;33:66974. [DOI] [PubMed]
    Yi X, Li CY, Zhang SH, Wang XH, Li ZQ, Yang F. Relationship and clinical significance of KiSS-1, nuclear factor kappa B (NF-kappaB), p50, and matrix metalloproteinase 9 expression in breast cancer. Zhonghua Bing Li Xue Za Zhi. 2008;37:23842. Chinese. [PubMed]
    Gründker C, Bauerschmitz G, Knapp J, Schmidt E, Olbrich T, Emons G. Inhibition of SDF-1/CXCR4-induced epithelial-mesenchymal transition by kisspeptin-10. Breast Cancer Res Treat. 2015;152:4150. [DOI] [PubMed]
    Fang L, Yan Y, Gao Y, Wu Z, Wang Z, Yang S, et al. TGF-β1 inhibits human trophoblast cell invasion by upregulating kisspeptin expression through ERK1/2 but not SMAD signaling pathway. Reprod Biol Endocrinol. 2022;20:22. [DOI] [PubMed] [PMC]
    Steeg PS, De la Rosa A, Flatow U, MacDonald NJ, Benedict M, Leone A. Nm23 and breast cancer metastasis. Breast Cancer Res Treat. 1993;25:17587. [DOI] [PubMed]
    Kim B, Lee KJ. Activation of Nm23-H1 to suppress breast cancer metastasis via redox regulation. Exp Mol Med. 2021;53:34657. [DOI] [PubMed] [PMC]
    Khan I, Steeg PS. The relationship of NM23 (NME) metastasis suppressor histidine phosphorylation to its nucleoside diphosphate kinase, histidine protein kinase and motility suppression activities. Oncotarget. 2017;9:10185202. [DOI] [PubMed] [PMC]
    Boissan M, Lacombe ML. NM23, an example of a metastasis suppressor gene. Bull Cancer. 2012;99:43140. French. [DOI] [PubMed]
    Rasool RU, Nayak D, Chakraborty S, Jamwal VL, Mahajan V, Katoch A, et al. Differential regulation of NM23-H1 under hypoxic and serum starvation conditions in metastatic cancer cells and its implication in EMT. Eur J Cell Biol. 2017;96:16471. [DOI] [PubMed]
    Wong KM, Song J, Wong YH. CTCF and EGR1 suppress breast cancer cell migration through transcriptional control of Nm23-H1. Sci Rep. 2021;11:491. [DOI] [PubMed] [PMC]
    Duan S, Nordmeier S, Byrnes AE, Buxton ILO. Extracellular vsicle-mediated purinergic signaling contributes to host microenvironment plasticity and metastasis in triple negative breast cancer. Int J Mol Sci. 2021;22:597. [DOI] [PubMed] [PMC]
    Khan I, Gril B, Hoshino A, Yang HH, Lee MP, Difilippantonio S, et al. Metastasis suppressor NME1 in exosomes or liposomes conveys motility and migration inhibition in breast cancer model systems. Clin Exp Metastasis. 2022;39:81531. [DOI] [PubMed]
    Mielnicki LM, Ying AM, Head KL, Asch HL, Asch BB. Epigenetic regulation of gelsolin expression in human breast cancer cells. Exp Cell Res. 1999;249:16176. [DOI] [PubMed]
    Fujita H, Okada F, Hamada J, Hosokawa M, Moriuchi T, Koya RC, et al. Gelsolin functions as a metastasis suppressor in B16-BL6 mouse melanoma cells and requirement of the carboxyl-terminus for its effect. Int J Cancer. 2001;93:77380. [DOI] [PubMed]
    Stock AM, Klee F, Edlund K, Grinberg M, Hammad S, Marchan R, et al. Gelsolin is associated with longer metastasis-free survival and reduced cell migration in estrogen receptor-positive breast cancer. Anticancer Res. 2015;35:527785. [PubMed]
    Dong Y, Asch HL, Ying A, Asch BB. Molecular mechanism of transcriptional repression of gelsolin in human breast cancer cells. Exp Cell Res. 2002;276:32836. [DOI] [PubMed]
    Dong Y, Asch HL, Medina D, Ip C, Ip M, Guzman R, et al. Concurrent deregulation of gelsolin and cyclin D1 in the majority of human and rodent breast cancers. Int J Cancer. 1999;81:9308. [DOI] [PubMed]
    Van den Abbeele A, De Corte V, Van Impe K, Bruyneel E, Boucherie C, Bracke M, et al. Downregulation of gelsolin family proteins counteracts cancer cell invasion in vitro. Cancer Lett. 2007;255:5770. [DOI] [PubMed]
    Kuramochi M, Fukuhara H, Nobukuni T, Kanbe T, Maruyama T, Ghosh HP, et al. TSLC1 is a tumor-suppressor gene in human non-small-cell lung cancer. Nat Genet. 2001;27:42730. [DOI] [PubMed]
    Watabe K, Ito A, Koma YI, Kitamura Y. IGSF4: a new intercellular adhesion molecule that is called by three names, TSLC1, SgIGSF and SynCAM, by virtue of its diverse function. Histol Histopathol. 2003;18:13219. [DOI] [PubMed]
    Faraji F, Pang Y, Walker RC, Nieves Borges R, Yang L, Hunter KW. Cadm1 is a metastasis susceptibility gene that suppresses metastasis by modifying tumor interaction with the cell-mediated immunity. PLoS Genet. 2012;8:e1002926. [DOI] [PubMed] [PMC]
    Zhang G, Zhong L, Luo H, Wang S. MicroRNA-155-3p promotes breast cancer progression through down-regulating CADM1. Onco Targets Ther. 2019;12:79938002. [DOI] [PubMed] [PMC]
    Wikman H, Westphal L, Schmid F, Pollari S, Kropidlowski J, Sielaff-Frimpong B, et al. Loss of CADM1 expression is associated with poor prognosis and brain metastasis in breast cancer patients. Oncotarget. 2014;5:307687. [DOI] [PubMed] [PMC]
    Heller G, Geradts J, Ziegler B, Newsham I, Filipits M, Markis-Ritzinger EM, et al. Downregulation of TSLC1 and DAL-1 expression occurs frequently in breast cancer. Breast Cancer Res Treat. 2007;103:28391. [DOI] [PubMed]
    Allinen M, Peri L, Kujala S, Lahti-Domenici J, Outila K, Karppinen SM, et al. Analysis of 11q21-24 loss of heterozygosity candidate target genes in breast cancer: indications of TSLC1 promoter hypermethylation. Genes Chromosomes Cancer. 2002;34:3849. [DOI] [PubMed]
    Overmeer RM, Henken FE, Snijders PJ, Claassen-Kramer D, Berkhof J, Helmerhorst TJ, et al. Association between dense CADM1 promoter methylation and reduced protein expression in high-grade CIN and cervical SCC. J Pathol. 2008;215:38897. [DOI] [PubMed]
    Vermeulen MA, van Deurzen CHM, Doebar SC, de Leng WWJ, Martens JWM, van Diest PJ, et al. Promoter hypermethylation in ductal carcinoma in situ of the male breast. Endocr Relat Cancer. 2019;26:57584. [DOI] [PubMed]
    Saito M, Goto A, Abe N, Saito K, Maeda D, Ohtake T, et al. Decreased expression of CADM1 and CADM4 are associated with advanced stage breast cancer. Oncol Lett. 2018;15:24016. [DOI] [PubMed] [PMC]
    Si X, Xu F, Xu F, Wei M, Ge Y, Chenge S. CADM1 inhibits ovarian cancer cell proliferation and migration by potentially regulating the PI3K/Akt/mTOR pathway. Biomed Pharmacother. 2020;123:109717. [DOI] [PubMed]
    Galibert L, Diemer GS, Liu Z, Johnson RS, Smith JL, Walzer T, et al. Nectin-like protein 2 defines a subset of T-cell zone dendritic cells and is a ligand for class-I-restricted T-cell-associated molecule. J Biol Chem. 2005;280:2195564. [DOI] [PubMed]
    Boles KS, Barchet W, Diacovo T, Cella M, Colonna M. The tumor suppressor TSLC1/NECL-2 triggers NK-cell and CD8+ T-cell responses through the cell-surface receptor CRTAM. Blood. 2005;106:77986. [DOI] [PubMed]
    Giangreco A, Hoste E, Takai Y, Rosewell I, Watt FM. Epidermal Cadm1 expression promotes autoimmune alopecia via enhanced T cell adhesion and cytotoxicity. J Immunol. 2012;188:151422. [DOI] [PubMed]
    Murakami S, Sakurai-Yageta M, Maruyama T, Murakami Y. Trans-homophilic interaction of CADM1 activates PI3K by forming a complex with MAGuK-family proteins MPP3 and Dlg. PLoS One. 2014;9:e110062. [DOI] [PubMed] [PMC]
    Masuda M, Yageta M, Fukuhara H, Kuramochi M, Maruyama T, Nomoto A, et al. The tumor suppressor protein TSLC1 is involved in cell-cell adhesion. J Biol Chem. 2002;277:310149. [DOI] [PubMed]
    Gaugitsch HW, Hofer E, Huber NE, Schnabl E, Baumruker T. A new superfamily of lymphoid and melanoma cell proteins with extensive homology to Schistosoma mansoni antigen Sm23. Eur J Immunol. 1991;21:37783. [DOI] [PubMed]
    van ’t Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002;415:5306. [DOI] [PubMed]
    Dong JT, Isaacs WB, Barrett JC, Isaacs JT. Genomic organization of the human KAI1 metastasis-suppressor gene. Genomics. 1997;41:2532. [DOI] [PubMed]
    Zhao Y, Sato Y, Isaji T, Fukuda T, Matsumoto A, Miyoshi E, et al. Branched N-glycans regulate the biological functions of integrins and cadherins. FEBS J. 2008;275:193948. [DOI] [PubMed]
    Takahashi M, Kuroki Y, Ohtsubo K, Taniguchi N. Core fucose and bisecting GlcNAc, the direct modifiers of the N-glycan core: their functions and target proteins. Carbohydr Res. 2009;344:138790. [DOI] [PubMed]
    Bari R, Zhang YH, Zhang F, Wang NX, Stipp CS, Zheng JJ, et al. Transmembrane interactions are needed for KAI1/CD82-mediated suppression of cancer invasion and metastasis. Am J Pathol. 2009;174:64760. [DOI] [PubMed] [PMC]
    Welch DR, Hurst DR. Defining the hallmarks of metastasis. Cancer Res. 2019;79:301127. [DOI] [PubMed] [PMC]
    Tsai YC, Weissman AM. Dissecting the diverse functions of the metastasis suppressor CD82/KAI1. FEBS Lett. 2011;585:316673. [DOI] [PubMed] [PMC]
    Schoenfeld N, Bauer MKA, Grimm S. The metastasis suppressor gene C33/CD82/KAI1 induces apoptosis through reactive oxygen intermediates. FASEB J. 2004;18:15860. [DOI] [PubMed]
    He B, Liu L, Cook GA, Grgurevich S, Jennings LK, Zhang XA. Tetraspanin CD82 attenuates cellular morphogenesis through down-regulating integrin α6-mediated cell adhesion. J Biol Chem. 2005;280:334654. [DOI] [PubMed]
    Odintsova E, Sugiura T, Berditchevski F. Attenuation of EGF receptor signaling by a metastasis suppressor, the tetraspanin CD82/KAI-1. Curr Biol. 2000;10:100912. [DOI] [PubMed]
    Claas C, Stipp CS, Hemler ME. Evaluation of prototype transmembrane 4 superfamily protein complexes and their relation to lipid rafts. J Biol Chem. 2001;276:797484. [DOI] [PubMed]
    Hemler ME. Tetraspanin proteins mediate cellular penetration, invasion, and fusion events and define a novel type of membrane microdomain. Annu Rev Cell Dev Biol. 2003;19:397422. [DOI] [PubMed]
    Ono M, Handa K, Withers DA, Hakomori S. Motility inhibition and apoptosis are induced by metastasis-suppressing gene product CD82 and its analogue CD9, with concurrent glycosylation. Cancer Res. 1999;59:23359. [PubMed]
    Lee JH, Seo YW, Park SR, Kim YJ, Kim KK. Expression of a splice variant of KAI1, a tumor metastasis suppressor gene, influences tumor invasion and progression. Cancer Res. 2003;63:724755. [PubMed]
    Iwata S, Kobayashi H, Miyake-Nishijima R, Sasaki T, Souta-Kuribara A, Nori M, et al. Distinctive signaling pathways through CD82 and β1 integrins in human T cells. Eur J Immunol. 2002;32:132837. [DOI] [PubMed]
    Mannion BA, Berditchevski F, Kraeft SK, Chen LB, Hemler ME. Transmembrane-4 superfamily proteins CD81 (TAPA-1), CD82, CD63, and CD53 specifically associated with integrin alpha 4 beta 1 (CD49d/CD29). J Immunol. 1996;157:203947. [PubMed]
    Shibagaki N, Hanada Ki, Yamashita H, Shimada S, Hamada H. Overexpression of CD82 on human T cells enhances LFA-1 / ICAM-1-mediated cell-cell adhesion: functional association between CD82 and LFA-1 in T cell activation. Eur J Immunol. 1999;29:408191. [DOI] [PubMed]
    Lagaudrière-Gesbert C, Lebel-Binay S, Wiertz E, Ploegh HL, Fradelizi D, Conjeaud H. The tetraspanin protein CD82 associates with both free HLA class I heavy chain and heterodimeric beta 2-microglobulin complexes. J Immunol. 1997;158:27907. [PubMed]
    Nakamura K, Mitamura T, Takahashi T, Kobayashi T, Mekada E. Importance of the major extracellular domain of CD9 and the epidermal growth factor (EGF)-like domain of heparin-binding EGF-like growth factor for up-regulation of binding and activity. J Biol Chem. 2000;275:1828490. [DOI] [PubMed]
    Zhang XA, Bontrager AL, Hemler ME. Transmembrane-4 superfamily proteins associate with activated protein kinase C (PKC) and link PKC to specific β1 integrins. J Biol Chem. 2001;276:2500513. [DOI] [PubMed]
    Bass R, Werner F, Odintsova E, Sugiura T, Berditchevski F, Ellis V. Regulation of urokinase receptor proteolytic function by the tetraspanin CD82. J Biol Chem. 2005;280:148118. [DOI] [PubMed]
    Fang BA, Kovačević Ž, Park KC, Kalinowski DS, Jansson PJ, Lane DJ, et al. Molecular functions of the iron-regulated metastasis suppressor, NDRG1, and its potential as a molecular target for cancer therapy. Biochim Biophys Acta. 2014;1845:119. [DOI] [PubMed]
    Bae DH, Jansson PJ, Huang ML, Kovacevic Z, Kalinowski D, Lee CS, et al. The role of NDRG1 in the pathology and potential treatment of human cancers. J Clin Pathol. 2013;66:9117. [DOI] [PubMed]
    Sun J, Zhang D, Bae DH, Sahni S, Jansson P, Zheng Y, et al. Metastasis suppressor, NDRG1, mediates its activity through signaling pathways and molecular motors. Carcinogenesis. 2013;34:194354. [DOI] [PubMed]
    Lachat P, Shaw P, Gebhard S, van Belzen N, Chaubert P, Bosman FT. Expression of NDRG1, a differentiation-related gene, in human tissues. Histochem Cell Biol. 2002;118:399408. [DOI] [PubMed]
    Aikemu B, Shao Y, Yang G, Ma J, Zhang S, Yang X, et al. NDRG1 regulates Filopodia-induced colorectal cancer invasiveness via modulating CDC42 activity. Int J Biol Sci. 2021;17:171630. [DOI] [PubMed] [PMC]
    Patel JH, Loboda AP, Showe MK, Showe LC, McMahon SB. Analysis of genomic targets reveals complex functions of MYC. Nat Rev Cancer. 2004;4:5628. [DOI] [PubMed]
    Taketomi Y, Sunaga K, Tanaka S, Nakamura M, Arata S, Okuda T, et al. Impaired mast cell maturation and degranulation and attenuated allergic responses in Ndrg1-deficient mice. J Immunol. 2007;178:704253. [DOI] [PubMed]
    Kovacevic Z, Chikhani S, Lui GYL, Sivagurunathan S, Richardson DR. The iron-regulated metastasis suppressor NDRG1 targets NEDD4L, PTEN, and SMAD4 and inhibits the PI3K and Ras signaling pathways. Antioxid Redox Signal. 2013;18:87487. [DOI] [PubMed]
    Jin R, Liu W, Menezes S, Yue F, Zheng M, Kovacevic Z, et al. The metastasis suppressor NDRG1 modulates the phosphorylation and nuclear translocation of β-catenin through mechanisms involving FRAT1 and PAK4. J Cell Sci. 2014;127:311630. [DOI] [PubMed]
    Liu W, Xing F, Iiizumi-Gairani M, Okuda H, Watabe M, Pai SK, et al. N-myc downstream regulated gene 1 modulates Wnt-β-catenin signalling and pleiotropically suppresses metastasis. EMBO Mol Med. 2012;4:93108. [DOI] [PubMed] [PMC]
    Wang Z. ErbB Receptors and Cancer. Methods Mol Biol. 2017;1652:335. [DOI] [PubMed]
    Sevinsky CJ, Khan F, Kokabee L, Darehshouri A, Maddipati KR, Conklin DS. NDRG1 regulates neutral lipid metabolism in breast cancer cells. Breast Cancer Res. 2018;20:55. [DOI] [PubMed] [PMC]
    Li EY, Huang WY, Chang YC, Tsai MH, Chuang EY, Kuok QY, et al. Aryl hydrocarbon receptor activates NDRG1 transcription under hypoxia in breast cancer cells. Sci Rep. 2016;6:20808. [DOI] [PubMed] [PMC]
    Nagai MA, Gerhard R, Fregnani JH, Nonogaki S, Rierger RB, Netto MM, et al. Prognostic value of NDRG1 and SPARC protein expression in breast cancer patients. Breast Cancer Res Treat. 2011;12:114. [DOI] [PubMed]
    Mao XY, Fan CF, Wei J, Liu C, Zheng HC, Yao F, et al. Increased N-myc downstream-regulated gene 1 expression is associated with breast atypia-to-carcinoma progression. Tumour Biol. 2011;32:12716. [DOI] [PubMed]
    Liu Y, Hu X, Han C, Wang L, Zhang X, He X, et al. Targeting tumor suppressor genes for cancer therapy. Bioessays. 2015;37:127786. [DOI] [PubMed] [PMC]
    Kuo KK, Hsiao PJ, Chang WT, Chuang SC, Yang YH, Wuputra K, et al. Therapeutic strategies targeting tumor suppressor genes in pancreatic cancer. Cancers (Basel). 2021;13:3920. [DOI] [PubMed] [PMC]
    Chen C, Yue D, Lei L, Wang H, Lu J, Zhou Y, et al. Promoter-operating targeted expression of gene therapy in cancer: current stage and prospect. Mol Ther Nucleic Acids. 2018;11:50814. [DOI] [PubMed] [PMC]
    Viera M, Yip GWC, Shen HM, Baeg GH, Bay BH. Targeting CD82/KAI1 for precision therapeutics in surmounting metastatic potential in breast cancer. Cancers (Basel). 2021;13:4486. [DOI] [PubMed] [PMC]
    Montaño-Samaniego M, Bravo-Estupiñan DM, Méndez-Guerrero O, Alarcón-Hernández E, Ibáñez-Hernández M. Strategies for targeting gene therapy in cancer cells with tumor-specific promoters. Front Oncol. 2020;10:605380. [DOI] [PubMed] [PMC]
    Shee K, Yang W, Hinds JW, Hampsch RA, Varn FS, Traphagen NA, et al. Therapeutically targeting tumor microenvironment-mediated drug resistance in estrogen receptor-positive breast cancer. J Exp Med. 2018;215:895910. [DOI] [PubMed] [PMC]
    Platonov ME, Borovjagin AV, Kaverina N, Xiao T, Kadagidze Z, Lesniak M, et al. KISS1 tumor suppressor restricts angiogenesis of breast cancer brain metastases and sensitizes them to oncolytic virotherapy in vitro. Cancer Lett. 2018;417:7588. [DOI] [PubMed]
    Alimirzaie S, Bagherzadeh M, Akbari MR. Liquid biopsy in breast cancer: a comprehensive review. Clin Genet. 2019;95:64360. [DOI] [PubMed]