Skip to main content

An immunotherapeutic approach to decipher the role of long non-coding RNAs in cancer progression, resistance and epigenetic regulation of immune cells

Abstract

Immunotherapeutic treatments are gaining attention due to their effective anti-tumor response. Particularly, the revolution of immune checkpoint inhibitors (ICIs) produces promising outcomes for various cancer types. However, the usage of immunotherapy is limited due to its low response rate, suggesting that tumor cells escape the immune surveillance. Rapid advances in transcriptomic profiling have led to recognize immune-related long non-coding RNAs (LncRNAs), as regulators of immune cell-specific gene expression that mediates immune stimulatory as well as suppression of immune response, indicating LncRNAs as targets to improve the efficacy of immunotherapy against tumours. Moreover, the immune-related LncRNAs acting as epigenetic modifiers are also under deep investigation. Thus, herein, is a summarised knowledge of LncRNAs and their regulation in the adaptive and innate immune system, considering their importance in autophagy and predicting putative immunotherapeutic responses.

Background

Cancer immunotherapy (CI) is a rapidly advancing treatment regimen supporting the primary mode of treatments like surgery, chemotherapy, radiation, and targeted therapy [1]. The conventional cancer treatment generally failed to produce beneficial responses in patients with late-stage disease treatment compared to early non-metastatic cancer patients. Besides, the traditional treatment modalities are often associated with side effects like disfigurement, cytotoxicity on normal cells, and hair loss [2]. However, CI has fewer side effects, and it aims to engage the immune system directly to recognize and eradicate tumor cells. The main attributes of CI are the specificity, breadth of response, and memory against the tumor antigens that can provide better clinical outcomes, and thus can increase the quality of life, particularly in metastatic patients [3].

Immunotherapy has a multi-faceted approach like the use of monoclonal antibodies, checkpoint inhibitors, cancer vaccine, and cell-based therapy [4]. These modalities can be either passive or active immune therapeutics. In passive immune therapeutics, the ICIs are variant monoclonal antibodies that block checkpoint receptors facilitating the T cell activation to clear the malignant cells. Recently employed ICIs are anti-PDL1s /anti-CTLA-4s that are passive immunotherapy agents [5]. While, active immune therapeutics specifically aim to generate durable, long-lasting memory responses, by utilizing immune cells like dendritic cells, which are the potent antigen-presenting cells and immunostimulatory cytokines [6]. However, there are several ongoing challenges for immunotherapy that make this passive and active immune therapeutics, highly challenging. One among them is that many persons fail to respond to the CI due to high mutation burdens [7]. This indicates that CI must be personalized through careful understanding and identification of the rate-limiting step working on a specific patient. These can be addressed by providing combinational strategies to overcome the cancer burden through utilizing high throughput technologies to characterize patients’ specific landscape like identifying a neoantigen and immune molecular signature pertain to the individuals, which may solve this to an extent [8]. Besides, several factors influence an effective CI like key evasion tactics including abrogated expression of cancer antigens and checkpoint receptor ligands which majorly prevent the entry of tumor-infiltrating lymphocytes (TILs) in tumor mass, decreased antigen presentation, b2-microglobulin alterations, severe exhaustion of T cells, and increased activation and recruitment of immunosuppressive cells or induction of suppressive cytokines such as IL-10 and TGF-β [9]. This concerns the specificity of CI in eliciting an immune response, overcoming the mechanisms that cancer cells employ to evade immune surveillance, and ensures that the activated immune cells have access to the malignant tissues. Hence, there is a persistent need in identifying more specific biomarkers, better predictive tools, and assays to identify patients who will respond to these immunotherapies.

To understand the depth of immune resistance, studies were focused on the tumor microenvironment, but they are unable to provide the detailed mechanisms of immune evasion to date. Intriguingly, recent studies showed that this issue can be resolved by considering the crucial involvement of non-coding RNAs in tumor cell-intrinsic factors that mediate tumor cell escape from immune surveillance [10, 11]. Through the evolution of high throughput sequence technology, rapid screening of non-coding RNAs was made possible in a limited time as never before [12, 13]. The research revealed that only a small portion of approximately 20,000 genes are protein-coding, which accounts for <2% of the human genome encodes, whereas 90% of the genome is transcribed into non-coding RNAs [14]. Of these non-coding RNAs, microRNAs (miRNAs) are widely studied and differentially expressed with several human pathologies, including cancer. Furthermore, miRNAs are the regulators of innate and adaptive immunity and their maintenance, helping in the generation of immune progenitors and their differentiation, modulating the functions of mature immune cells [15, 16].

Unlike miRNAs, long non-coding RNA is 200 nucleotides long, transcribed by RNA polymerase II, and regulated by splicing (via processing at the 5′ and 3′ ends), and exported to the cytoplasm [17]. LncRNAs respond to several cellular functions ranging from modification of chromatins leading to RNA stability through translational control. Biochemically, LncRNAs exert their function via RNA-RNA, RNA-DNA, or RNA-protein interactions [18, 19]. Moreover, LncRNAs are deregulated in several ailment conditions and could categorize cellular abnormalities leading to cancer. It may even account for delivering a prognostic value or therapeutic options for cancer patients [20, 21]. For example, MALAT1 is over-expressed in several cancer types, and knockdown of MALAT1 reduces both the proliferation and metastasis of tumor cells in several mouse models [22]. Besides, MALAT1 also acts as an epigenetic modifier by regulating the PRC2 complex [23]. Emerging studies also revealed the role of LncRNAs in differentiation, proliferation, and immune cells’ activation [24]. Thus, this review provides a critical analysis of LncRNAs in the regulation of cancer resistance by direct and indirect modification of T cells, B cells, Treg cells, natural killer cells (NKs), macrophages and myeloid-derived suppressor cells (MDSCs), and autophagosomes functionality, projecting it as a tool to treat various cancers, as a personalized treatment regimen.

LncRNAs as modulators of the adaptive immune system

Adaptive immune responses are referred to the generation of antigen-specific lymphocytes by host organisms against the subsequent exposure of similar antigens or if reinfected with the same type of pathogen. The adaptive immune system consists of T cells and B cells [25].

Inhibition of LncRNAs potentiates immunotherapeutic strategies of cytotoxic T lymphocytes (CTLs) by curtailing the functionality of T regulatory cells (Tregs)

The T lymphocytes stimulate the activation of the immune cell to fight against cancerous cells [26]. Moreover, T cells signal other immune cells to participate in the immune responses. There are several T cell sub-sets, which cause the death of cancer cells and prevent their progression. One among them is the CTLs which possess the surface expression of CD8+. These CTLs are involved in the destruction of targeted cells through apoptosis by utilizing the granules containing digestive enzymes or cytotoxic cytokines (Tumour Necrosis Factor-TNF and Interferon γ - IFN γ) [27]. Cancer cells are known to resists the effect of CTLs by reorganizing the cellular function through the expression of coding RNAs and LncRNAs leading to an altered cellular response to the native immune system. Recent studies also showed the role of LncRNAs in resisting the action of CTLs in cancers [28, 29].

Nuclear-enriched autosomal transcript 1 (NEAT1) is a nuclear paraspeckle localized LncRNA reported in different types of cancer [30, 31]. In vivo experiments showed that the inhibition of NEAT1 decrease CD8+T cell apoptosis and gain active cytolytic function via the miR-155/Tim-3 pathway leading to enhanced immune activity (Fig. 1). Moreover, Tim-3 upregulated in exhausted T cells in tumors as well as in chronic infections. In turn, the increased expression of this Tim-3 leads to the death of CD8+ T cells. Hence Tim-3 has emerged as a potential immunotherapeutic target which is currently explored clinically [32]. However, Ji et al. (2018) [33] reported the role of Lnc-Tim-3 in hepatocellular carcinoma in the regulation of Tim-3 protein expressions. They stated that an increase of Lnc-Tim-3 specifically binds with Tim-3 protein and blocks its interaction with Bat-3 thus suppressing downstream Lck/NFAT1/AP-1 signaling, leading to nuclear localization of Bat-3, and enhancing p300-dependent p53 and RelA transcriptional activation of anti-apoptosis genes including MDM2 and Bcl2 leads to survival and proliferation of exhausted T cells (Fig. 1). This study indicates Lnc-TIM-3 can be used as to target to deploy the T cell exhaustion and proliferation.

Fig. 1
figure 1

LncRNA mediated molecular pathways in T Cells. In CTLs and Th cells, due to TCR signalling, there may be a Ca2+ influx, which can activate calmodulin. This promotes deacetylation of NILKA promoter, causing nuclear translocation of p300 inhibiting NF-kB pathway and activating AICD. In CD8+ cells, Lnc-NEAT1 regulates miR-155p mediated Tim-3 activation. While binding of Lnc-Tim-3 to Tim-3 protein inhibit Lck/NFAT1/AP-1 pathway and permit the localisation of Bat-3 towards p300 mediated activation of RelA and p53. In Th1 cells, EZH2 binds to LSD1, causing nuclear localization of EZH2, which inactivates transcription of MAF-4 gene by recruiting Linc-MAF-4 and H3K27 trimethylation of the promotor. In CD4+/CD8+ cells, Lnc-NeST and Lnc-HOTTIP binds to WDR5 protein of SET/MLL/WRD5 complex. The NeST can even recruit STAT to WDR5 through methylation. STAT4, thus gets localised to nucleus causing activation of IFN gamma pathways

Furthermore, Huang et al. (2018) [34] reported an increased expression of nuclear factor κB interacting long non coding RNA (LncRNA NKILA) in cytotoxic T lymphocytes leading to the poor prognosis in breast and lung cancer. But knockdown of NKILA significantly decreased the tumor growth by increasing the CTLs mediated activation induced cell death (AICD), by the histone-deacetylase (HDAC)-mediated P300 nuclear localization, post calcium influx and calmodulin activation (Fig. 1). Besides, long intergenic non-coding RNA (LincRNA) Tmevgpg1/ Nettoie Salmonella pas Theiler’s (NeST) identified in Thelier’s virus infection [35] revealed co-expression with interferon γ (IFNγ). It can stimulate epigenetic remodelling through its interaction with WDR5 mainly, depends on the transcription factors T-bet and STAT4. Hence this NeST is classified as an enhancer RNA [36, 37], was recognized for its proximity-based cis-transactivation of IFNγ. Moreover, the LincRNA-MAF-4 is also involved in the development of Thl response, mediated through the epigenetic silencing of the transcriptional factor MAF-4, by the recruitment of the chromatin modifiers like polycomb Group proteins (PRC1 and PRC2) which facilitates the deposition of H3K27me3 (transcriptional repression) by utilising the catalytic component partner mainly Enhance of Zeste Homolog 2 (EZH2). In addition, Lysine specific demethylase 1 (LSD1) also facilitates H3K4me3 (transcriptional activation) and H3K27me3 at their gene regulatory regions. Thus LincRNA-MAF-4 interacts with the transcriptional repressors LSD1 and EZH2 to facilitate the H3K27me3 trimethylation mark at the promoter of MAF to silence its expression (Fig. 1) [38, 39].

Cancer cells mediated immune suppression by several mechanisms involves an accumulation of immune-suppressive tumor-infiltrating lymphocytes in the tumor microenvironment [40]. One among them is Tregs which are the most potent and well-studied suppressive phenotypes found in the tumor microenvironment. Increasing evidence shows that Treg cells may also play an important role in immune evasion mechanisms employed by cancer. In addition, recent evidence shows that tumor-derived LncRNA plays a pivotal role in Treg maintenance and differentiation [41]. For instance, Jiang et al. (2017) [42] pointed out the novel role of Lnc-epidermal growth factor receptor (EGFR) in differentiating Tregs, causing suppression of CTLs in hepatocellular carcinoma (Fig. 2). The study identified an upregulation of Lnc-EGFR in Tregs and was well-correlated with the expression of EGFR/Foxp3 and tumor size, but negatively correlated with IFN-γ expression. Even though many studies evidenced the FOXp3 Tregs are involved in the progression of the tumor, a recent study [43] showed the two different populations of Tregs called FOXp3 low and FOXp3 high have a different role in promoting tumor. In line with this Zemmour et al. (2017) [44] discovered the role of flicr (Foxp3 long intergenic noncoding RNA) in regulating the expression of FOXp3 in Treg. The increased expression of IL-2 followed by the T cell receptor (TCR) signaling are involved in intensifying the suppressive functions of Foxp3 in Tregs by activating conserved noncoding sequence-2 (CNS-2) enhancer by inhibiting flicr. Hence, there was an urge to study the role of LncRNA in controlling Treg functionality further to potentiate the immunotherapeutic strategies, including miR regulation, downstream of LncRNA regulatory mechanisms.

Fig. 2
figure 2

Molecular targets of LncRNAs responsible for T regulatory cells differentiation and inhibition of proliferation. In Treg cells, Lnc-EGFR stimulates FOXp3 causing inhibition of IFN. While Lnc-SNHG1 and Lnc-SOX5/SOX3 stimulates IDO production. Lnc-SNHG20, Lnc-MALAT1, Lnc-CECR7 and LINC00473 accelerates PD-1 and PD-L1. While Lnc-NKX2-1-AS1 and Lnc-Rp11-51IM6.8 inhibit PD-1 and PD-L1

To understand the role of LncRNA and miR interplay in IDO production, Pei et al. (2018) [45] demonstrated the interaction of LncRNA-SNHG1 (Small nucleolar RNA host gene 1) with miR-448 in the regulation of the IDO expression in TILs. The study shows miR-448 negatively regulates the IDO expression, especially in CD4+ TILs not in CD4+ cells in circulating peripheral blood. Lnc-SNHG1 sponges miR-448 in turn, leading to upregulation of IDO expression in hepatocellular carcinomic cells [46] suggests LncRNA-SNHG1 might be used as potential therapeutic target in enhancing the immunotherapy by inhibiting the expression of LncRNA. Moreover, the increased expression of Lnc-SOX5 and SOX3, in the tongue and colon cancer was found to be inhibiting the T cell activity by upregulating IDO1 [47]. Previous studies also showed the IDO1 mediated immunosuppression facilitating the progression of tumor growth viz., upregulation of Treg and myeloid-derived suppressor cells (MDSC) regulatory cells (Fig. 2).

Besides, LncRNA is also involved in the regulation of CTLA4 and PD-L1 mediated Treg propagation. A recent report [48] showed the role of LncRNA RP11-571M6.8 driver LncRNA in promoting human cancers. The increased expression of LncRNA RP11-571M6.8 had lowered the expression of immune checkpoint proteins such as PD-1, PD-L1, and CTLA4. In addition, its expression is also well correlated with the exhausted Tregs, particularly in glioblastoma multiforme (GBM) datasets. Moreover, Zhou et al. (2019) [49] showed that LINC00473 LncRNA involved in the sponging of miR195-5p in pancreatic cancer leads to the upregulation of PD-L1 expression. In-line inhibition of LINC00473 causes enhancement of CTLs causing suppression of tumor progression. Moreover, Kathuria et al. (2018) [50] also reported that LncRNAs NKX2-1-AS1 and NXX2-1 in regulating the PD-L1 expression in lung cancer. Where NXX2-1 protein is involved in the upregulation of PD-L1 but the NKX2-1-AS1 LncRNA disturbs the upregulation of PD-L1 by reducing the CD274 gene transcript which codes for PD-L1 (Fig. 2). Thus suggest the screening the pattern of potential LncRNA which curtail the PD-L1 expression between cancer patients can predict the responders and non-responder in immune checkpoint therapy.

LncRNA small nucleolar RNA host gene 20 (SNHG20) also plays a crucial role in increasing the expression of PD-L1, p-ATM, and p-JAK1/2 in esophageal squamous cell carcinoma. The modulation of this signaling pathway promotes proliferation and metastasis of cancer cells [51]. LncRNA CECR7 (cat eye syndrome chromosome region, candidate 7) regulates the CTLA4 expression associated with the overall survival of hepatocellular carcinoma [52]. However, in diabetic pancreatic cancer, the expression of CECR7 was downregulated suggesting its expression pattern varies between different cancers (Fig. 2) [53].

However, MALAT1 was the most widely studied LncRNA which was initially identified in lung cancer [54]. Later it was also reported in driving H3K27me3 mediated repression of tumor suppressor genes through its interaction with the members of PRC2 complex mediating tumor progression and metastasis [55]. In addition, Zhao et al (2016) [56] revealed its role in regulating the innate immune response in NF-kB dependent manner. Moreover, a recent study by Wang et al. (2019) [57] indicated its involvement in sponging miR-195 causing upregulation of PD-L1, leading to immune escape and apoptosis of CTLs (Fig. 2). Hence, cancer specific expression of MALAT1 can be explored for targeted therapy as well as biomarkers in cancer.

The role of Lnc RNA in B cell development and malignancy

The B cells play an important role as an initiator of humoral immune responses. In addition, B cells are also involved in the initiation of T cell immune responses, which are essential for regulating immune homeostasis [58]. Petri et al. (2015) [59] also showed the importance of LncRNA in the regulation of B cells in every stage of its differentiation. LncRNAs such as long intergenic ncRNAs (LincRNA) CTC 436K13.6 and SMAD1-AS1, MYB-AS1, and LEF1-AS1 which are antisense transcript encoded by their opposite sense strands possess a crucial function in the early B cell formations. Among the gene expressed, CRNDE (colorectal neoplasia differentially expressed) which found expressed in preB1, B2, and centroblasts (rapidly dividing B cells that achieve further diversification of the Ig repertoire through the somatic hypermutation of the V regions of Ig genes) and low in centrocytes (generate new antibody variants that are selected according to their affinity to the cognate antigen, ensuring increased affinity between the Ig and the antigen). Besides, the expression of this CRNDE function as the metabolic regulator in the early stage of B cell development is believed to favour the metabolic switch to aerobic glycolysis in tumor cells. Another LncRNA BIC (B-cell integration cluster) gives rise to miR-155-5p, and it plays a central role in hematopoiesis, inflammation, and immune response. The increased expression of BIC and miR-155 has been found in many mature B- cell malignancies [34]. Impaired production of soluble Fas receptor (sFAS) was associated with poor prognosis in non-Hodgkin’s lymphomas [60]. In line the expression of sFAS was tightly regulated by LncRNA FAS-AS1, where it binds with RNA binding motif protein5 (RBM5), preventing the alternative splicing of FAS pre-mRNA required for sFAS production. But the FAS-AS1 transcript was hyper methylated by over-expressed EZH2- enzymatic component of PRC2 complex leads to repression of FAS-AS1, in turn causes aberrant expression of sFAS in B-cell lymphoma. Parallelly, Zhou et al. (2016) [61] showed the downregulation of this LncRNA causes increased tumor size and lymph node metastasis in breast cancer patients. These data necessitate the use of EZH2 inhibitors in combination with the chemotherapeutic targets helps in augmenting the immune cell mediated cell killing. A recent bioinformatics analysis [62] in NSCLC revealed that LncRNA-GVINP1 was associated with regulation of the B cell receptor signaling pathway associated with the overall survival period of NSCLC patients. Kelin et al. (2010) [63] reported that LncRNA DLEU2 along with miR-15a/16-1, controls the proliferation of B cells, by the dysregulation of this cluster leading to uncontrolled proliferation in CLL (Fig. 3).

Fig. 3
figure 3

LncRNA mediated molecular pathways in B cell maturation. In bone marrow, the development of pro/pre/immature B cells are mediated by Lnc-CRNDE, SMAD1-AS1, MYB-AS1 and LEF1-AS1. lncRNA BIC plays a major role in hemotopoises peripheral compartments Naïve B cells and FAS activations are mediated by LncRNAs BIC and FAS-AS1. The BCR signalling is mediated by Lnc-GVINP1. Linc-BM742401, Oct-4 mediated MIAT and MALAT1 cause CLL proliferation, Tumoral B cell formation and multiple myeloma, respectively

MIAT which was initially discovered in myocardial infarction [64] suggested constituting a regulatory loop with Oct4 in tumoral mature B-cell [65]. Further both MIAT and Oct4 were demonstrated to be essential for the tumour cell survival supporting the proliferation of malignant mature B cells [66]. Hence high expression of MIAT is associated to pathogenesis in cancer patients, can considered for potential biomarker as well as therapeutic target. LincRNA BM742401 was frequently methylated in chronic lymphocytic leukemia (CLL) [67] was previously reported that lower expression was correlated with poor survival in gastric cancer [68]. Particularly, the promoter region of BM742401 which was unmethylated in peripheral B-cells collected from healthy donors but methylated in 50% CLL samples at diagnosis. The methylation of this BM742401 in CLL samples was significantly associated with higher lymphocyte counts and correlated with the advanced stage. Further MALAT1 expression also significantly upregulated in multiple myeloma (MM) patients compared to treated or healthy donors. In addition, increased expression of MALAT1 was seen in relapsed MM patients [69]. These studies indicate the important role of non-coding RNA in curtailing the function of adaptive soldier’s which leads to tumour escape from immune surveillance causing tumour progression (Fig. 3).

LncRNA act as regulators of innate immune warriors

The innate immune responses are not specific to a particular pathogen. They mainly depend on the pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) that are conserved features of pathogens [70]. The major effector cells of the innate immune system which are involved indirectly in targeting the cancer cells are natural killer cells (NKs), dendritic cells (DCs), macrophages, and myeloid-derived suppressor cells (MDSCs). In this section, we concentrated on the regulation of LncRNAs in NK, macrophages, and MDSCs.

Reinforcing cancer resistance by curtailing the functionality of innate cytotoxic NK cells

Natural killer cells are important members of the innate immune system by providing a natural defence against infections involved in mediating antitumor immune responses. The growing evidence suggests the role of LncRNAs in NK cells’ development and tumor escape. A recent study [71] reported that LncRNA GAS5 is critical for IFNγ secretion, which got downregulated in NK cells of liver cancer patients. In addition, the study indicated that knocking down this GAS5 may lead to a decrease in the percentage of CD107a+ NKs, causing decreased apoptosis of HepG2 and Huh7 cells. However, the overexpression of LncRNA GAS5 decreased miR-544 expression, by increasing runt related transcription factor 3 (RUNX3) expression, favouring increased activated NK cell-mediated cytotoxicity suggesting a potential therapeutic target for cancer (Fig. 4).

Fig. 4
figure 4

Major LncRNAs mediated molecular events occurring in the cancer immune surveillance. RPPH1 and TUC339 present in the cancer cell-secreted exosomes facilitate the expansion of M2 macrophages and MM2P, UCA1 also causes the M2 phenotype induction in cancer. On the other hand, lncRNA such as Lnc-COX2, CCDC26, and RP11-419k12.1 mediates the M1 macrophage induction. MDSC can be derived from both infiltrating neutrophils or monocytes, here AK036396 induces MDSC from infiltrating neutrophils through stabilizing the ficolin B expression. Further, Lnc-CHOP and RNCR3 both induce the H3K4 trimethylation in the promoter region of arginine, iNOS, and COX2 facilitates M-MDSC generation. NK cells infiltration consider as a potent anti-tumor response where LncGAS5 and ARSP91 facilitates the increased NK cell activity, while RAB30-AS1, ARAP-AS1, PRMT5-AS1, and ZFAS1 causes aberrant cell growth particularly in NKTCL carcinoma

Further, whole transcriptome analysis of natural killer/T cell lymphoma (NKTCL), normal NK and NK lymphoma cell lines revealed 166 LncRNA which had more than 1.5-fold overexpression. The LncRNAs such as RAB30-AS1, ARAP-AS1, and PRMT5-AS1 are suggested to have a biological function on cell growth [72]. In addition, Askarian-Amiri et al. (2011) [73] discovered the novel LncRNA ZNFX1 antisense RNA 1 (ZFAS1) transcribed from the antisense gene ZNFX1, which hosts for three SNORD (small nucleolar RNAs c/D box) with extreme stability for more than 16 hours. Up-regulated in cancers such as glioma, colorectal, gastric, hepatocellular, ovarian cancers and down-regulated in breast cancer. Furthermore, ZFAS1-correlated genes are associated with the upregulation of many pathways, including NF-kB signaling, b-catenin independent Wnt signaling, and p53-dependent apoptosis and the cell cycle pathways (Fig. 4) [74].

The other study revealed the role of LncRNA small nucleolar RNA host gene 12 (SNHG12) acting as a direct transcriptional target of c-Myc. Thus c-Myc-mediated increased expression of LncRNA SNHG12 facilitates proliferation and resistance to cisplatin in NKTCL [75]. This proposes SNHG12 may serve as diagnostic and druggable therapeutic target with promising clinical potential in cancer. Further, Ma et al. (2019) [76] demonstrated that circular RNA of AR-suppressed PABPC1 91 bp (ARSP91) was involved in the enhancement of cytotoxicity activity in natural killer cells against hepatocellular carcinoma. Subsequent mechanistic research revealed that upregulating UL16 binding protein 1 (ULBP1) expression in hepatocellular carcinoma cells led to the enhanced innate immune surveillance, by strengthening cytotoxic NK cells (Fig. 4).

Decisive role of LncRNA in polarising the tissue-associated macrophages

Monocytes from the bloodstream are migrated to the tissues to differentiate as macrophages. Moreover, recent reports also evidenced that most tissue-resident macrophages originate during embryonic development [77]. They play a critical mechanism in eliminating the apoptotic bodies, harmful foreign substances, and cancer cells. Further, the macrophages can polarize to M1 that are activated macrophages involved in triggering an anti-tumor response. The M2 causes immune suppression led to the development of tumor-associated macrophages (TAMs), which play a crucial role in the tumor microenvironment as significant molecules of tumor propagation [78]. Many studies revealed the importance of LncRNA in deciding the polarization pattern of TAM.

In evidence, the LncRNA Cox2 highly expressed in M1 macrophages is induced in response to LPS and TLR1 and 2 (toll-like receptor) through MyD88 and NF-κB-dependent manner. Moreover, the silencing of this LncRNA-Cox-2 using siRNA causes a reduction in IL12 and TNF α and increased expression of M2 features with higher IL10 and arginase-1. Besides, the proliferation, migration, invasion, angiogenesis, and epithelial-mesenchymal transition (EMT) of hepatocellular carcinoma cells were increased when co-cultured with LncRNA-Cox-2 knockdown M2 macrophages, which needs to be validated in other cancers to consider as potential therapeutic target [79]. LncRNA-CASC2c expression abrogated the M2 phenotype and decreased the GBM tumor growth by inhibiting the M2 polarization [80]. Furthermore, a recent report [81] proved that the expression of LncRNA RPPH1 is associated with advanced TNM (Tumour Node metastasis) stage and owes for poor prognosis in colorectal cancer (CRC). The cancer cells release exosomes containing LncRNA RPPH1 transported into macrophages causes induced polarization of M2 phenotype. Collectively these studies indicate the necessity of targeting LncRNA, in turn, improving the immune therapeutics (Fig. 4).

Li et al. (2018) [82] showed the HCC exosome-derived Lnc-RNATUC339 is involved in the polarization of macrophages toward the M2 phenotype. In line Cao et al. (2019) [83] reported the induction of IL4 and IL13 in cell line and animal models showed upregulation of 25 LncRNA identified using microarray analysis of RAW264.7 and bone marrow-derived macrophages. Out of 25 LncRNA, the novel LncRNA MM2P is specifically expressed in M2 polarized macrophages. Further, knockdown of LncRNA MM2P blocked the M2 polarization, particularly, it weakened the angiogenesis promoting factor of M2 by inhibiting the phosphorylation of STAT6. Indicates lncRNA MM2P might involve in activation and maintenance of STAT6 causes sustained M2 polarisation. Chen et al. (2015) [84], reported that an activated LncRNA UCA1 promoted M2 macrophage infiltration, leading to tumor progression in breast cancer. In addition, the differential expression status of LncRNAs decides the fate of infiltrating CD14+ monocytes polarization. Gabrusiewicz et al. (2016) [85] identified the varied expression pattern of three LncRNA 14orf139, SNORA25, and CCDC26 in glioblastoma dictated the polarization of infiltrating CD14+TAM. Furthermore, the study highlighted that the increased expression of two LncRNAs (CCDC26 and RP11-419k12.1) favours the M1 TAM whereas the LncRNA RP11-26E5.1 was favouring M2 TAM (Fig. 4).

Chemokines play a vital role in the differentiation of macrophages in the tumor microenvironment. In specific the increased expression of CCL2 is crucial for tumor metastasis. In evidence, a recent report showed that LncRNA LNMAT1 (lymph node metastasis-associated transcript 1) recruits hnRNPL to CCL2 promotor, leads to H3K4me3 methylation causes transcriptional activation of CCL2 responsive genes. Upregulated expression of CCL2 led to the recruitment of macrophages which favours accelerated lymphatic metastasis in bladder cancer through VEGF-C secretion [86]. In line HOTAIR1 was also associated with CCL2 mediated recruitment of TAMs and MDSC [87]. LncRNA CCAT1 and NIFK-AS1 controls, the expression of miR (148a and 146a) favours the generation of M1 TAM, which was downregulated in various cancer types, causing M2 polarization and tumor cell invasion [88, 89]. Kong et al. 2018 [90] discovered a novel LncRNA-ZFPM-AS1 (ZFPM2 antisense RNA 1) was associated with gastric carcinogenesis. In addition, their results indicated the direct binding of this LncRNA to macrophage migration inhibitory factor (MIF) prevent the degradation of MIF acts as a potent destabilizer of p53. Further, knockdown of MIF caused reduced expression of ZFPM-AS1, resulting in a p53 increase in gastric cancer. Thus, the studies indicates LncRNA can function as both- oncogenes where it involved in the promotion of M2 by inhibiting the M1 polarization and tumour suppressor which facilitates the M1 generation while inhibiting the M2-related gene expression.

Recruitment of myeloid derived suppressor cells towards tumour microenvironment through LncRNA expression

Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of immature granulocytes/monocytes that accumulate in cancer, and other disease conditions like autoimmunity [91, 92]. They release arginase-1 (Arg1), NO synthase 2 (NOS2), NADPH oxidase 2 (NOX2), cyclooxygenase-2 (Cox2), and various toxic and regulatory substances like H2O2 and reactive oxygen species (ROS) [93]. Causing damage in nucleic acids, proteins, and lipids, they attenuate many immune effectors particularly curtailing the T cell activities. MDSC is classified into two major subtypes based on their cell surface phenotype and morphology: polymorphonuclear MDSC (PMN-MDSC) and monocytic MDSC (M-MDSC) [94]. MDSC is also involved in the generation of induced T regulatory cells (iTregs) [95] in tumor microenvironments causing poor survival and a low response to immune checkpoint inhibitors (ICIs). Likely, a recent study by Ma et al. (2019) [96] showed there was an increase in the MDSC counts in cancer compared to premalignant or healthy donors contributing to poor survival.

The growing evidence indicates the role of LncRNAs in recruiting these immune suppressive cells to the tumor milieu. Shang et al. (2019) [97] discovered that LncRNA Olfr29-ps1 in mouse models were 43% like the OR1F2P gene in humans. The expression of this LncRNAs is upregulated in the presence of Granulocyte-macrophage colony-stimulating factor (GM-CSF) and IL6, compared to GM-CSF, through the STAT3 and JAK3 pathways. The induced expression of this LncRNA enhances the differentiation of immune cells to M-MDSC rather than PMN-MDSC. Moreover, the N6-methyladenosine (m6A) is specific to cell type regulating the function of eukaryotic messenger RNAs (mRNA) and LncRNAs. In this context, m6A modification was increased upon proinflammatory cytokine induction. Further, this modulation favours the Olfr29-ps1 in sponging the miR-214-3p acting as a competing endogenous RNA (ceRNA) mediating in the differentiation of MDSC. Another study [98] showed the role of Lnc-CHOP mediating the differentiation of MDSC to M-MDSC through its interaction with C/EBP homologous protein (CHOP) and the C/EBPβ isoform LIP. Consequently, the interaction also promoted the epigenetic modification in H3K4me3 on the promoter region of Arg1, NOS2, NOX2, and Cox2 favours potent immunosuppressive function. Moreover, the expression of CHOP is also regulated by another LncRNA called retinal non-coding RNA3 (RNCR3) by sponging miR-185-5p [99] which affects the expansion of MDSC (Fig. 4). These reveal the complex role of LncRNA where two different non-coding RNA regulating the CHOP expression. Hence the context-dependent role of non-coding RNA activation needs to broaden might potentiates the targeted therapy in cancer.

Tian et al. 2018 [100] confirmed the role of LncRNA RUNX1 overlapping RNA (RUNXOR) in regulating the MDSC in lung cancer. The increased expression of this RUNXOR causes a decrease in the expression of runt-related transcription factor1 RUNX1 which acts as a tumour suppressor by promoting the differentiation of MDSCs in lung cancer. The expression of LncRNA was also differentially regulated in lung adenocarcinoma and squamous cell lung carcinoma indicates the screening of this LncRNA might help in identifying lung cancer subtypes. In addition, miR-9 negatively regulates the RUNX1 expression in lewis lung carcinoma. Collectively, these studies showed the negative regulation of RUNXOR and miR-9 by competing the RUNX1. Hypoxia is one of the hallmarks of the tumor microenvironment where Hypoxia-inducible factors (HIFs) play an important role in the regulation of cellular responses to hypoxia including regulation of immune cell response. Zhang et al. (2019) [101] showed the hypoxia-mediated increase in the LncRNA-plasmacytoma variant translocation 1 (Pvt1) in G-MDSC particularly by increasing the expression of c-Myc in tumour bearing mice. Moreover, the report inferred both subtypes of MDSC (M-MDSC and G-MDSC) isolated from the tumor tissue showed an upregulated expression of PVT1 with increased Arg1 and ROS expression. A recent report showed the increased expression of LncRNA AK036396 favours the immunosuppression of PMN-MDSC [102] through stabilizing the ficolin B protein that acts as a surrogate for PMN-MDSC development (Fig. 4).

Directives of LncRNA in autophagy

Autophagy is a conserved catabolic process involved in cellular homeostasis and is required to maintain normal cellular physiology under stressful conditions [103]. It has been reported to modulate both innate and adaptive immune responses, through regulation of homeostasis, survival, activation, proliferation, and differentiation of the immune system. Moreover, it also affects the cytokine release and transport of antibodies [104, 105]. Autophagy favours immunotherapy by safeguarding the regulation of immunostimulatory signals by antigens delivery towards the immune cells, lymphocyte development, thymus selection, and immune homeostasis [106].

Several data revealed many aspects of autophagy are regulated by LncRNAs [107,108,109]. Specifically, one of the mechanisms by which LncRNAs regulate cellular functions is by acting as ceRNA. The MALAT1 LncRNA is a potent autophagy inducer that protects brain microvascular endothelial cells (BMEC) against oxygen-glucose deprivation/re-oxygenation (OGD/R) by sponging miR-26. In turn, it can cause upregulation of miR-26b targeting ULK2 leading to the survival of BMEC by promoting autophagy under OGD/R condition [110]. Likewise, Li et al. (2018) [111] identified the role of MALAT1 in tumorigenesis and tumor cell survival of glioma through autophagy. The study reported MALAT1 damage the suppressive effect of miR-101 on glioma cell autophagy by acting as ceRNA through upregulation of miR-101 targeting STMN1, RAB5A, and ATG4D [112].

HOTAIRM1 plays a crucial role in myeloid cell differentiation through degradation of oncoprotein PML-RARA by regulating the autophagy pathway. The study showed a decrease in the HOTAIRM1 expression in APL (acute promyelocytic leukemia) leading to the inhibition of all-trans-retinoic acid (ATRA) mediated degradation of PML-RARA causes repression of promyelocytic to granulocytic cellular differentiation. The HOTAIRM1 can regulate the autophagy pathway by acting as ceRNA by sponging the miRNAs, miR-20a/106b, and miR-125b, which targets crucial genes in autophagy through ULK1, E2F1, and DRAM2 which facilitates for the degradation of PML [113]. This reveals over-expression of HOTAIRM1 might be a potential therapeutic target for APL. The NLR family proteins NLRP3 acts as a sensor of the pathogen, environmental and host-derived factors. This NLRP3 the inflammasome is a multimeric protein complex requires for the release of proinflammatory cytokines IL-1β and IL-18 in mature form and also initiates the inflammatory form of cell death. Intriguingly, lincRNA-Cox2 could act as both an activator and suppressor of innate immune responses, playing an important role in NLRP3 inflammasome formation through binding with the NF-κB p65 subunit regulation, thus promoting its nuclear translocation and transcription. Besides, knockdown of lincRNA-Cox2 repressed inflammasome activation and prohibited the LincRNA-Cox2 triggered caspase-1 activation, causing decreased IL-1β secretion and weakened TIR-domain-containing adapter-inducing interferon-β (TRIF) cleavage, by prompting the TRIF-mediated autophagy [109] (Fig. 5).

Fig. 5
figure 5

LncRNA involved in various processes of autophagy – induction, initiation, phagophore nucleation, autolysosome fusion. MALAT1 is the well-studied LncRNA involved in all the steps of autophagy. The other LncRNA GAS5 indicated its role in increased NK cell cytotoxicity also involved in the initiation and phagophore nucleation

Thus, based on the above discussion, the use of LncRNAs for cancer immunotherapy in a personalized manner can be taken as a branch of cancer therapy soon. Moreover, a recent study [114] showed the association of LncRNAs in immunotherapy responses by predictions using computational algorithms. Based on the immune-related LncRNA signature and CTL infiltration, the patients were categorized into 4 different classes. Among the four classes, the immune active class showed increased oxidative stress. Conversely, the study highlighted the patients with low LncRNA score had prolonged survival with increased immune activity compared to high LncRNA score. Furthermore, the upregulated and downregulated set of LncRNA and miRNA also act as a predictor of monitoring the chimeric antigen receptor T cells (CAR-T) therapy efficacy. Moreover, the number of LncRNAs enriched in NK cell-mediated cytotoxicity was upregulated upon CD19 CAR-T cell infusion. Particularly LncRNA RN7SL1 which was reported [114] to be associated with immune processes displayed a high degree of co-expression with histone genes including HIST1H4B and HIST1H2BL. Further, a set of 22 differentially expressed miRNA had a similar pattern in remissive patients. Especially, the expression of miR-27a-3p, a tumor suppressor in B-ALL cell lines, was upregulated after CAR-T therapy, potentially regulates a set of crosstalk genes (e.g., CEBPE and CHD4), and participates in the immune response pathways [114].

Future prospective

The collective studies revealed the multitasking role of single LncRNA in many cancers. For instance, MALAT1 was proved to be involved in epigenetic modifications as well as autophagy induction through sponging miRNA. HOTAIR not only promotes HLAG expression [115] to enhance the antigen presentation but also down-regulates CD4+/CD8+ T-cell subsets, and also involve in autophagosome elongation and closure suggesting the effects of LncRNAs, which needs further studies to understand its mechanisms clearly. Lnc-TIM-3 and LncSNHG1 which plays a major role in the sustained expansion of exhaustive T cells might be used as the potential therapeutic target can be combined with the cell-mediated therapies like dendritic cell vaccine, adoptive T cell therapy to enhance the treatment efficacy. GAS5 which is down-regulated in many cancers likes CRC, NSCLC, breast, and pancreatic cancer indicates the over-expression of GAS5 aids in regression of the tumour. It was evidenced that LncRNA prostate cancer antigen 3 (PCA3) is overexpressed in 95% of prostate cancers and can be used as molecular markers [116] in prostate cancers, which was approved by the US food and drug administration in 2012 [117]. Furthermore, MALAT1 and CRADE can be used as a biomarker where it was detected in the serum of NSCLC and CRC in exosomal form [118]. Macrophages constitute a major population in tumour microenvironment where the increased polarization towards the M2 supports cancer proliferation. For instance, the LncRNA RPPH1 and UCA1 increase facilitates the metastasis in CRC by increasing M2 conversion needs further work in other cancer to use as a potential therapeutic target might sustain the activation of M1 macrophages. Moreover, ZFAS1 LncRNA down-regulated in breast cancer and upregulated in many cancer types indicates the context-specific regulation which needs to be studied in the future.

Figure 6 gives an overall account of major LncRNAs which might act as a potential target in cancer immunotherapy. Moreover, Table 1 shows an account of major LncRNAs working on development of lymphoma, melanoma, and myeloma of human cancer types (courtesy to http://www.cuilab.cn/lncrnadisease) [167]. Hence, in view of the above, the LncRNA gained great potential as prognostic/diagnostic markers and therapeutic targets providing great insight into the prospects of LncRNAs in cancer immunotherapy.

Fig. 6
figure 6

Potential LncRNAs can aid as targets for cancer immunotherapy. The figure shows relevant LncRNAs can be used as a potential target in enhancing the immunotherapeutic strategies either by upregulation of its expression (eg. GAS5 and FAS-AS1) can increase anti-tumor response or silencing of LncRNA expression (eg., MM2P and Tim-3) might mediates an increase in anti-tumor response by inhibiting the Treg, M2, MDSC expansion

Table 1 Account of LncRNAs responsible for blood cells related cancer and malignancies

Conclusion

The functionality of LncRNA is complex and competes with both miRNA and mRNA to regulate the expression of the protein. Recent studies revealed that LncRNAs act as epigenetic modifiers involved in promoting innate immune memory responses. Future explorations of immune-related LncRNAs in predicting immune responses are needed, as potential candidatures for specific immune cell-mediated tumor evasion and cellular apoptosis.

Availability of data and materials

Not applicable.

Abbreviations

AICD:

Activation induced cell death

ALL:

Acute lymphoblastic leukemia

AML:

Acute myeloid leukemia

APL:

Acute promyelocytic leukemia

Arg1:

Arginase-1

ARSP91:

AR-suppressed PABPC1 91 bp

ATRA:

All-trans-retinoic acid

BCL:

B-cell lymphoma

BIC:

B-cell integration cluster

BM:

Burkitt lymphoma

BMEC:

Brain microvascular endothelial cells

CAR-T:

Chimeric antigen receptor T cells

CECR7:

Cat eye syndrome chromosome region, candidate 7

ceRNA:

Competing endogenous RNA

CHOP:

C/EBP homologous protein

CI:

Cancer immunotherapy

CLL:

Chronic lymphocytic leukemia

CML:

Chronic myeloid leukemia

CNS-2:

Conserved noncoding sequence-2

Cox2:

Cyclooxygenase-2

CRNDE:

Colorectal neoplasia differentially expressed

CRC:

Colorectal cancer

CTLs:

Cytotoxic T lymphocytes

DAMPs:

Damage-associated molecular patterns

DCs:

Dendritic cells

DLBCL:

Diffuse large B-cell lymphoma

EGFR:

Epidermal growth factor receptor

EMT:

Epithelial-mesenchymal transition

EZH2:

Enhance of Zeste Homolog 2

Flicr:

Foxp3 long intergenic noncoding RNA

GBM:

Glioblastoma multiforme

GM-CSF:

Granulocyte-macrophage colony-stimulating factor

G-MDSC:

Granulocytes-MDSC

HDAC:

Histone-deacetylase

HIFs:

Hypoxia-inducible factors

ICIs:

Immune checkpoint inhibitors

IFN γ:

Interferon γ

iTregs:

induced T regulatory cells

LincRNA:

Long intergenic non-coding RNA

LncRNA NKILA:

Nuclear factor κB interacting long non coding RNA

LncRNAs:

Long non-coding RNAs

LNMAT1:

Lymph node metastasis-associated transcript 1

LSD1:

Lysine specific demethylase 1

M-MDSC:

Monocytic MDSC

MDS:

Myelodysplastic syndromes

MDSC:

Myeloid-derived suppressor cells

MIF:

Macrophage migration inhibitory factor

MM:

Multiple myeloma

mRNA:

messenger RNA

nNEAT1:

Nuclear-enriched autosomal transcript 1

NeST:

Nettoie Salmonella pas Theiler’s

NKs:

Natural killer cells

NKTCL:

Natural killer/T cell lymphoma

NOS2:

NO synthase 2

NOX2:

NADPH oxidase 2

OGD/R:

Oxygen-glucose deprivation/re-oxygenation

p-ATM:

PD-L1, ataxia telangiectasia mutated kinase

PAMPs:

Pathogen-associated molecular patterns

PMN-MDSC:

Polymorphonuclear MDSC

PRC:

Polycomb group protein

Pvt1:

Plasmacytoma variant translocation 1

RBM5:

RNA binding motif protein5

RNCR3:

Retinal non-coding RNA3

ROS:

Reactive oxygen species

RUNX:

Runt related transcription factor

RUNXOR:

RUNX1 overlapping RNA

sFAS:

soluble FAS receptor

SNHG1:

Small nucleolar RNA host gene 1

SNHG20:

Small nucleolar RNA host gene 20

SNORD:

Small nucleolar RNAs C/D box

TAMs:

Tumor-associated macrophages

TCR:

T cell receptor

TILs:

Tumor-infiltrating lymphocytes

TLR:

Toll-like receptor

TNM:

Tumour node metastasis

TNF:

Tumour necrosis factor

TRAIL:

TNF-related apoptosis-inducing ligand

Tregs:

T regulatory cells

TRIF:

TIR-domain-containing adapter-inducing interferon-β

ULBP1:

UL16 binding protein 1

ZFAS1:

ZNFX1 antisense RNA 1

ZFPM-AS1:

ZFPM2 antisense RNA 1

References

  1. Emens LA, Ascierto PA, Darcy PK, Demaria S, Eggermont AMM, et al. Cancer immunotherapy: Opportunities and challenges in the rapidly evolving clinical landscape. Eur J Cancer. 2017;81:116–29. https://doi.org/10.1016/j.ejca.2017.01.035.

    Article  CAS  PubMed  Google Scholar 

  2. Nurgali K, Jagoe RT, Abalo R. Editorial: Adverse effects of cancer chemotherapy: Anything new to improve tolerance and reduce sequelae? Front Pharmacol. 2018;9:245. https://doi.org/10.3389/fphar.2018.00245.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kaufman HL, Atkins MB, Subedi P, Wu J, Chambers J, Joseph Mattingly T, et al. The promise of Immuno-oncology: implications for defining the value of cancer treatment. J Immunother Cancer. 2019;7(1):129. https://doi.org/10.1186/s40425-019-0594-0.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Papaioannou NE, Beniata OV, Vitsos P, Tsitsilonis O, Samara P. Harnessing the immune system to improve cancer therapy. Ann Transl Med. 2016;4(14):261. https://doi.org/10.21037/atm.2016.04.01.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Galluzzi L, Vacchelli E, Bravo-San Pedro JM, Buqué A, Senovilla L, Baracco EE, et al. Classification of current anticancer immunotherapies. Oncotarget. 2014;5(24):12472–508. https://doi.org/10.18632/oncotarget.2998.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Rossi M, Young JW. Human dendritic cells: potent antigen-presenting cells at the crossroads of innate and adaptive immunity. J Immunol. 2005;175(3):1373–81. https://doi.org/10.4049/jimmunol.175.3.1373.

    Article  CAS  PubMed  Google Scholar 

  7. Zugazagoitia J, Guedes C, Ponce S, Ferrer I, Molina-Pinelo S, Paz-Ares L. Current challenges in cancer treatment. Clin Ther. 2016;38(7):1551–66. https://doi.org/10.1016/j.clinthera.2016.03.026.

    Article  PubMed  Google Scholar 

  8. Kakimi K, Karasaki T, Matsushita H, Sugie T. Advances in personalized cancer immunotherapy. Breast Cancer. 2017;24(1):16–24. https://doi.org/10.1007/s12282-016-0688-1.

    Article  PubMed  Google Scholar 

  9. Sambi M, Bagheri L, Szewczuk MR. Current challenges in cancer immunotherapy: Multimodal approaches to improve efficacy and patient response rates. J Oncol. 2019;2019:4508794. https://doi.org/10.1155/2019/4508794.

  10. Murciano-Goroff YR, Warner AB, Wolchok JD. The future of cancer immunotherapy: microenvironment-targeting combinations. Cell Res. 2020;30(6):507–19. https://doi.org/10.1038/s41422-020-0337-2.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Mpekris F, Voutouri C, Baish JW, Duda DG, Munn LL, Stylianopoulos T, et al. Combining microenvironment normalization strategies to improve cancer immunotherapy. Proc Natl Acad Sci U S A. 2020;117(7):3728–37. https://doi.org/10.1073/pnas.1919764117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kung JT, Colognori D, Lee JT. Long noncoding RNAs: past, present, and future. Genetics. 2013;193(3):651–69. https://doi.org/10.1534/genetics.112.146704.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Denaro N, Merlano MC, Lo NC. Long noncoding RNAs as regulators of cancer immunity. Mol Oncol. 2019;13(1):61–73. https://doi.org/10.1002/1878-0261.12413.

    Article  CAS  PubMed  Google Scholar 

  14. Stein LD. Human genome: end of the beginning. Nature. 2004;431(7011):915–6. https://doi.org/10.1038/431915a.

    Article  CAS  PubMed  Google Scholar 

  15. Sonkoly E, Ståhle M, Pivarcsi A. MicroRNAs and immunity: novel players in the regulation of normal immune function and inflammation. Semin Cancer Biol. 2008;18(2):131–40. https://doi.org/10.1016/j.semcancer.2008.01.005.

    Article  CAS  PubMed  Google Scholar 

  16. Lee HM, Nguyen DT, Lu LF. Progress and challenge of microRNA research in immunity. Front Genet. 2014;5:178. https://doi.org/10.3389/fgene.2014.00178.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Statello L, Guo CJ, Chen LL, Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 2021;22(2):96–118. https://doi.org/10.1038/s41580-020-00315-9.

    Article  CAS  PubMed  Google Scholar 

  18. Connerty P, Lock RB, de Bock CE. Long Non-coding RNAs: Major regulators of cell stress in cancer. Front Oncol. 2020;10:285. https://doi.org/10.3389/fonc.2020.00285.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Tuck AC, Tollervey D. A transcriptome-wide atlas of RNP composition reveals diverse classes of mRNAs and lncRNAs. Cell. 2013;154(5):996–1009. https://doi.org/10.1016/j.cell.2013.07.047.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Cipolla GA, de Oliveira JC, Salviano-Silva A, Lobo-Alves SC, Lemos DS, Oliveira LC, et al. Long non-coding RNAs in multifactorial diseases: Another layer of complexity. noncoding RNA. 2018;4(2):13. https://doi.org/10.3390/ncrna4020013.

    Article  CAS  PubMed Central  Google Scholar 

  21. Wang KC, Chang HY. Molecular Mechanisms of Long Noncoding RNAs. Mol Cell. 2011;43(6):904–14 https://doi.org/10.1016/j.molcel.2011.08.018.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gutschner T, Hämmerle M, Eissmann M, Hsu J, Kim Y, Hung G, et al. The noncoding RNA MALAT1 is a critical regulator of the metastasis phenotype of lung cancer cells. Cancer Res. 2013;73(3):1180–9. https://doi.org/10.1158/0008-5472.CAN-12-2850.

    Article  CAS  PubMed  Google Scholar 

  23. Qu D, Sun WW, Li L, Ma L, Sun L, Jin X, et al. Long noncoding RNA MALAT1 releases epigenetic silencing of HIV-1 replication by displacing the polycomb repressive complex 2 from binding to the LTR promoter. Nucleic Acids Res. 2019;47(6):3013–27. https://doi.org/10.1093/nar/gkz117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Satpathy AT, Chang HY. Long noncoding RNA in hematopoiesis and immunity. Immunity. 2015;42(5):792–804. https://doi.org/10.1016/j.immuni.2015.05.004.

    Article  CAS  PubMed  Google Scholar 

  25. Bonilla FA, Oettgen HC. Adaptive immunity. J Allergy Clin Immunol. 2010;125(2):S33–40. https://doi.org/10.1016/j.jaci.2009.09.017.

    Article  PubMed  Google Scholar 

  26. Katz SG, Rabinovich PM. T Cell Reprogramming Against Cancer. Methods Mol Biol. 2020;2097:3–44. https://doi.org/10.1007/978-1-0716-0203-4_1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Martínez-Lostao L, Anel A, Pardo J. How do cytotoxic lymphocytes kill cancer cells? Clin Cancer Res. 2015;21(22):5047–56. https://doi.org/10.1158/1078-0432.CCR-15-0685.

    Article  CAS  PubMed  Google Scholar 

  28. Luo Y, Yang J, Yu J, Liu X, Yu C, Hu J, et al. Long non-coding RNAs: Emerging roles in the immunosuppressive tumor microenvironment. Front Oncol. 2020;10:48. https://doi.org/10.3389/fonc.2020.00048.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Zhang L, Xu X, Su X. Noncoding RNAs in cancer immunity: functions, regulatory mechanisms, and clinical application. Mol Cancer. 2020;19(1):48. https://doi.org/10.1186/s12943-020-01154-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Pan LJ, Zhong TF, Tang RX, Li P, Dang YW, Huang SN, et al. Upregulation and clinicopathological significance of long non-coding NEAT1 RNA in NSCLC tissues. Asian Pac J Cancer Prev. 2015;16(7):2851–5. https://doi.org/10.7314/apjcp.2015.16.7.2851.

    Article  PubMed  Google Scholar 

  31. Peng W, Wang Z, Fan H. LncRNA NEAT1 impacts cell proliferation and apoptosis of colorectal cancer via regulation of Akt signaling. Pathol Oncol Res. 2017;23(3):651–6. https://doi.org/10.1007/s12253-016-0172-4.

    Article  CAS  PubMed  Google Scholar 

  32. Yan K, Fu Y, Zhu N, Wang Z, Hong JL, Li Y, et al. Repression of lncRNA NEAT1 enhances the antitumor activity of CD8+T cells against hepatocellular carcinoma via regulating miR-155/Tim-3. Int J Biochem Cell Biol. 2019;110:1–8. https://doi.org/10.1016/j.biocel.2019.01.019.

    Article  CAS  PubMed  Google Scholar 

  33. Ji J, Yin Y, Ju H, Xu X, Liu W, Fu Q, et al. Long non-coding RNA Lnc-Tim3 exacerbates CD8 T cell exhaustion via binding to Tim-3 and inducing nuclear translocation of Bat3 in HCC. Cell Death Dis. 2018;9(5):478. https://doi.org/10.1038/s41419-018-0528-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Huang D, Chen J, Yang L, Ouyang Q, Li J, Lao L, et al. NKILA lncRNA promotes tumor immune evasion by sensitizing T cells to activation-induced cell death. Nat Immunol. 2018;19(10):1112–25. https://doi.org/10.1038/s41590-018-0207-y.

    Article  CAS  PubMed  Google Scholar 

  35. Vigneau S, Rohrlich PS, Brahic M, Bureau JF. Tmevpg1, a candidate gene for the control of Theiler's virus persistence, could be implicated in the regulation of gamma interferon. J Virol. 2003;77(10):5632–8. https://doi.org/10.1128/jvi.77.10.5632-5638.2003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Collier SP, Henderson MA, Tossberg JT, Aune TM. Regulation of the Th1 genomic locus from Ifng through Tmevpg1 by T-bet. J Immunol. 2014;193(8):3959–65. https://doi.org/10.4049/jimmunol.1401099.

    Article  CAS  PubMed  Google Scholar 

  37. Gomez JA, Wapinski OL, Yang YW, Bureau JF, Gopinath S, Monack DM, et al. The NeST long ncRNA controls microbial susceptibility and epigenetic activation of the interferon-γ locus. Cell. 2013;152(4):743–54. https://doi.org/10.1016/j.cell.2013.01.015.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ahmad I, Valverde A, Ahmad F, Naqvi AR. Long noncoding RNA in myeloid and lymphoid cell differentiation, polarization and function. Cells. 2020;9(2):269. https://doi.org/10.3390/cells9020269.

    Article  CAS  PubMed Central  Google Scholar 

  39. Ranzani V, Rossetti G, Panzeri I, Arrigoni A, Bonnal RJ, Curti S, et al. The long intergenic noncoding RNA landscape of human lymphocytes highlights the regulation of T cell differentiation by linc-MAF-4. Nat Immunol. 2015;16(3):318–25. https://doi.org/10.1038/ni.3093.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Badalamenti G, Fanale D, Incorvaia L, Barraco N, Listì A, Maragliano R, et al. Role of tumor-infiltrating lymphocytes in patients with solid tumors: Can a drop dig a stone? Cell Immunol. 2019;343:103753. https://doi.org/10.1016/j.cellimm.2018.01.013.

    Article  CAS  PubMed  Google Scholar 

  41. Vinay DS, Ryan EP, Pawelec G, Talib WH, Stagg J, Elkord E, et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin Cancer Biol. 2015;35:S185–98. https://doi.org/10.1016/j.semcancer.2015.03.004.

    Article  CAS  PubMed  Google Scholar 

  42. Jiang R, Tang J, Chen Y, Deng L, Ji J, Xie Y, et al. The long noncoding RNA lnc-EGFR stimulates T-regulatory cells differentiation thus promoting hepatocellular carcinoma immune evasion. Nat Commun. 2017;8:15129. https://doi.org/10.1038/ncomms15129.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Saito T, Nishikawa H, Wada H, Nagano Y, Sugiyama D, Atarashi K, et al. Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med. 2016;22(6):679–84. https://doi.org/10.1038/nm.4086.

    Article  CAS  PubMed  Google Scholar 

  44. Zemmour D, Pratama A, Loughhead SM, Mathis D, Benoist C. Flicr, a long noncoding RNA, modulates Foxp3 expression and autoimmunity. Proc Natl Acad Sci U S A. 2017;114(17):E3472–80. https://doi.org/10.1073/pnas.1700946114.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pei X, Wang X, Li H. LncRNA SNHG1 regulates the differentiation of Treg cells and affects the immune escape of breast cancer via regulating miR-448/IDO. Int J Biol Macromol. 2018;118(Pt A):24–30. https://doi.org/10.1016/j.ijbiomac.2018.06.033.

    Article  CAS  PubMed  Google Scholar 

  46. Zhang M, Wang W, Li T, Yu X, Zhu Y, Ding F, et al. Long noncoding RNA SNHG1 predicts a poor prognosis and promotes hepatocellular carcinoma tumorigenesis. Biomed Pharmacother. 2016;80:73–9. https://doi.org/10.1016/j.biopha.2016.02.036.

    Article  CAS  PubMed  Google Scholar 

  47. Wang L, Ye S, Wang J, Gu Z, Zhang Y, Zhang C, et al. HuR stabilizes lnc-Sox5 mRNA to promote tongue carcinogenesis. Biochemistry (Mosc). 2017;82(4):438–45. https://doi.org/10.1134/S0006297917040046.

    Article  CAS  Google Scholar 

  48. Zhang Y, Liao G, Bai J, Zhang X, Xu L, Deng C, et al. Identifying cancer driver lncRNAs bridged by functional effectors through integrating multi-omics data in human cancers. Mol Ther Nucleic Acids. 2019;17:362–73. https://doi.org/10.1016/j.omtn.2019.05.030.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zhou WY, Zhang MM, Liu C, Kang Y, Wang JO, Yang XH. Long noncoding RNA LINC00473 drives the progression of pancreatic cancer via upregulating programmed death-ligand 1 by sponging microRNA-195-5p. J Cell Physiol. 2019;234(12):23176–89. https://doi.org/10.1002/jcp.28884.

    Article  CAS  PubMed  Google Scholar 

  50. Kathuria H, Millien G, McNally L, Gower AC, Tagne JB, Cao Y, et al. NKX2-1-AS1 negatively regulates CD274/PD-L1, cell-cell interaction genes, and limits human lung carcinoma cell migration. Sci Rep. 2018;8(1):14418. https://doi.org/10.1038/s41598-018-32793-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang C, Jiang F, Su C, Xie P, Xu L. Upregulation of long noncoding RNA SNHG20 promotes cell growth and metastasis in esophageal squamous cell carcinoma via modulating ATM-JAK-PD-L1 pathway. J Cell Biochem. 2019. https://doi.org/10.1002/jcb.28444.

  52. Zhang J, Fan D, Jian Z, Chen GG, Lai PB. Cancer specific long noncoding RNAs show differential expression patterns and competing endogenous RNA potential in hepatocellular carcinoma. PLoS One. 2015;10(10):e0141042. https://doi.org/10.1371/journal.pone.0141042.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Yao K, Wang Q, Jia J, Zhao H. A competing endogenous RNA network identifies novel mRNA, miRNA and lncRNA markers for the prognosis of diabetic pancreatic cancer. Tumour Biol. 2017;39(6):1010428317707882. https://doi.org/10.1177/1010428317707882.

    Article  CAS  PubMed  Google Scholar 

  54. Liu X, Huang G, Zhang J, Zhang L, Liang Z. Prognostic and clinicopathological significance of long noncoding RNA MALAT-1 expression in patients with non-small cell lung cancer: A meta-analysis. PLoS One. 2020;15(10):e0240321. https://doi.org/10.1371/journal.pone.0240321.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Hanly DJ, Esteller M, Berdasco M. Interplay between long non-coding RNAs and epigenetic machinery: emerging targets in cancer? Philos Trans R Soc Lond Ser B Biol Sci. 2018;373(1748):20170074. https://doi.org/10.1098/rstb.2017.0074.

    Article  CAS  Google Scholar 

  56. Zhao G, Su Z, Song D, Mao Y, Mao X. The long noncoding RNA MALAT1 regulates the lipopolysaccharide-induced inflammatory response through its interaction with NF-κB. FEBS Lett. 2016;590(17):2884–95. https://doi.org/10.1002/1873-3468.12315.

    Article  CAS  PubMed  Google Scholar 

  57. Wang QM, Lian GY, Song Y, Huang YF, Gong Y. LncRNA MALAT1 promotes tumorigenesis and immune escape of diffuse large B cell lymphoma by sponging miR-195. Life Sci. 2019;231:116335. https://doi.org/10.1016/j.lfs.2019.03.040.

    Article  CAS  PubMed  Google Scholar 

  58. Ron Y, De Baetselier P, Gordon J, Feldman M, Segal S. Defective induction of antigen-reactive proliferating T cells in B cell-deprived mice. Eur J Immunol. 1981;11(12):964–8. https://doi.org/10.1002/eji.1830111203.

    Article  CAS  PubMed  Google Scholar 

  59. Petri A, Dybkær K, Bøgsted M, Thrue CA, Hagedorn PH, Schmitz A, et al. Long noncoding RNA expression during human B-cell development. PLoS One. 2015;10(9):e0138236. https://doi.org/10.1371/journal.pone.0138236.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Sehgal L, Mathur R, Braun FK, Wise JF, Berkova Z, Neelapu S, et al. FAS-antisense 1 lncRNA and production of soluble versus membrane Fas in B-cell lymphoma. Leukemia. 2014;28(12):2376–87. https://doi.org/10.1038/leu.2014.126.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Zhou SW, Su BB, Zhou Y. Expression of FAS-AS1 in breast cancer and its effect on the host gene sFas. Zhonghua Yi Xue Za Zhi. 2016;96(12):949–53. https://doi.org/10.3760/cma.j.issn.0376-2491.2016.12.009.

    Article  CAS  PubMed  Google Scholar 

  62. Zhou W, Liu T, Saren G, Liao L, Fang W, Zhao H. Comprehensive analysis of differentially expressed long non-coding RNAs in non-small cell lung cancer. Oncol Lett. 2019;18(2):1145–56. https://doi.org/10.3892/ol.2019.10414.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Klein U, Lia M, Crespo M, Siegel R, Shen Q, Mo T, et al. The DLEU2/miR-15a/16-1 cluster controls B cell proliferation and its deletion leads to chronic lymphocytic leukemia. Cancer Cell. 2010;17(1):28–40. https://doi.org/10.1016/j.ccr.2009.11.019.

    Article  CAS  PubMed  Google Scholar 

  64. Ishii N, Ozaki K, Sato H, Mizuno H. Susumu Saito, Takahashi A, et al. Identification of a novel non-coding RNA, MIAT, that confers risk of myocardial infarction. J Hum Genet. 2006;51(12):1087–99. https://doi.org/10.1007/s10038-006-0070-9.

    Article  CAS  PubMed  Google Scholar 

  65. Sheik Mohamed J, Gaughwin PM, Lim B, Robson P, Lipovich L. Conserved long noncoding RNAs transcriptionally regulated by Oct4 and Nanog modulate pluripotency in mouse embryonic stem cells. RNA. 2010;16(2):324–37. https://doi.org/10.1261/rna.1441510.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Sattari A, Siddiqui H, Moshiri F, Ngankeu A, Nakamura T, Kipps TJ, et al. Upregulation of long noncoding RNA MIAT in aggressive form of chronic lymphocytic leukemias. Oncotarget. 2016;7(34):54174–82. https://doi.org/10.18632/oncotarget.11099.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Wang LQ, Wong KY, Li ZH, Chim CS. Epigenetic silencing of tumor suppressor long non-coding RNA BM742401 in chronic lymphocytic leukemia. Oncotarget. 2016;7(50):82400–10. https://doi.org/10.18632/oncotarget.12252.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Park SM, Park SJ, Kim HJ, Kwon OH, Kang TW, Sohn HA, et al. A known expressed sequence tag, BM742401, is a potent lincRNA inhibiting cancer metastasis. Exp Mol Med. 2013;45(7):e31. https://doi.org/10.1038/emm.2013.59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Cho SF, Chang YC, Chang CS, Lin SF, Liu YC, Hsiao HH, et al. MALAT1 long non-coding RNA is overexpressed in multiple myeloma and may serve as a marker to predict disease progression. BMC Cancer. 2014;14:809. https://doi.org/10.1186/1471-2407-14-809.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Roh JS, Sohn DH. Damage-associated molecular patterns in inflammatory diseases. Immune Netw. 2018;18(4):e27. https://doi.org/10.4110/in.2018.18.e27.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Fang P, Xiang L, Chen W, Li S, Huang S, Li J, et al. LncRNA GAS5 enhanced the killing effect of NK cell on liver cancer through regulating miR-544/RUNX3. Innate Immun. 2019;25(2):99–109. https://doi.org/10.1177/1753425919827632.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Baytak E, Gong Q, Akman B, Yuan H, Chan WC, Küçük C. Whole transcriptome analysis reveals dysregulated oncogenic lncRNAs in natural killer/T-cell lymphoma and establishes MIR155HG as a target of PRDM1. Tumour Biol. 2017;39(5):1010428317701648. https://doi.org/10.1177/1010428317701648.

    Article  CAS  PubMed  Google Scholar 

  73. Askarian-Amiri ME, Crawford J, French JD, Smart CE, Smith MA, Clark MB, et al. SNORD-host RNA Zfas1 is a regulator of mammary development and a potential marker for breast cancer. RNA. 2011;17(5):878–91. https://doi.org/10.1261/rna.2528811.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Mei M, Zhang M. Non-coding RNAs in natural killer/T-cell lymphoma. Front Oncol. 2019;9:515. https://doi.org/10.3389/fonc.2019.00515.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Zhu L, Zhang X, Fu X, Li Z, Sun Z, Wu J, et al. c-Myc mediated upregulation of long noncoding RNA SNHG12 regulates proliferation and drug sensitivity in natural killer/T-cell lymphoma. J Cell Biochem. 2019;120(8):12628–37. https://doi.org/10.1002/jcb.28529.

    Article  CAS  PubMed  Google Scholar 

  76. Ma Y, Zhang C, Zhang B, Yu H, Yu Q. circRNA of AR-suppressed PABPC1 91 bp enhances the cytotoxicity of natural killer cells against hepatocellular carcinoma via upregulating UL16 binding protein 1. Oncol Lett. 2019;17(1):388–97. https://doi.org/10.3892/ol.2018.9606.

    Article  CAS  PubMed  Google Scholar 

  77. Epelman S, Lavine KJ, Randolph GJ. Origin and functions of tissue macrophages. Immunity. 2014;41(1):21–35. https://doi.org/10.1016/j.immuni.2014.06.013.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Italiani P, Boraschi D. From monocytes to M1/M2 macrophages: Phenotypical vs. functional differentiation. Front Immunol. 2014;5:514. https://doi.org/10.3389/fimmu.2014.00514.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Ye Y, Xu Y, Lai Y, He W, Li Y, Wang R, et al. Long non-coding RNA cox-2 prevents immune evasion and metastasis of hepatocellular carcinoma by altering M1/M2 macrophage polarization. J Cell Biochem. 2018;119(3):2951–63. https://doi.org/10.1002/jcb.26509.

    Article  CAS  PubMed  Google Scholar 

  80. Zhang Y, Feng J, Fu H, Liu C, Yu Z, Sun Y, et al. Coagulation factor X regulated by CASC2c recruited macrophages and induced M2 polarization in glioblastoma multiforme. Front Immunol. 2018;9:1557. https://doi.org/10.3389/fimmu.2018.01557.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Liang ZX, Liu HS, Wang FW, Xiong L, Zhou C, Hu T, et al. LncRNA RPPH1 promotes colorectal cancer metastasis by interacting with TUBB3 and by promoting exosomes-mediated macrophage M2 polarization. Cell Death Dis. 2019;10(11):829. https://doi.org/10.1038/s41419-019-2077-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Li X, Lei Y, Wu M, Li N. Regulation of macrophage activation and polarization by HCC-derived exosomal lncRNA TUC339. Int J Mol Sci. 2018;19(10):2958. https://doi.org/10.3390/ijms19102958.

    Article  CAS  PubMed Central  Google Scholar 

  83. Cao J, Dong R, Jiang L, Gong Y, Yuan M, You J, et al. LncRNA-MM2P identified as a modulator of macrophage M2 polarization. Cancer Immunol Res. 2019;7(2):292–305. https://doi.org/10.1158/2326-6066.CIR-18-0145.

    Article  CAS  PubMed  Google Scholar 

  84. Chen S, Shao C, Xu M, Ji J, Xie Y, Lei Y, et al. Macrophage infiltration promotes invasiveness of breast cancer cells via activating long non-coding RNA UCA1. Int J Clin Exp Pathol. 2015;8(8):9052–61.

    PubMed  PubMed Central  Google Scholar 

  85. Gabrusiewicz K, Rodriguez B, Wei J, Hashimoto Y, Healy LM, Maiti SN, et al. Glioblastoma-infiltrated innate immune cells resemble M0 macrophage phenotype. JCI Insight. 2016;1(2):e85841. https://doi.org/10.1172/jci.insight.85841.

    Article  PubMed Central  Google Scholar 

  86. Chen C, He W, Huang J, Wang B, Li H, Cai Q, et al. LNMAT1 promotes lymphatic metastasis of bladder cancer via CCL2 dependent macrophage recruitment. Nat Commun. 2018;9(1):3826. https://doi.org/10.1038/s41467-018-06152-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Leija Montoya G, González Ramírez J, Sandoval Basilio J, Serafín Higuera I, Isiordia Espinoza M, González González R, et al. Long non-coding RNAs: Regulators of the activity of myeloid-derived suppressor cells. Front Immunol. 2019;10:1734. https://doi.org/10.3389/fimmu.2019.01734.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Liu J, Ding D, Jiang Z, Du T, Liu J, Kong Z. Long non-coding RNA CCAT1/miR-148a/PKCζ prevents cell migration of prostate cancer by altering macrophage polarization. Prostate. 2019;79(1):105–12. https://doi.org/10.1002/pros.23716.

    Article  CAS  PubMed  Google Scholar 

  89. Zhou YX, Zhao W, Mao LW, Wang YL, Xia LQ, Cao M, et al. Long non-coding RNA NIFK-AS1 inhibits M2 polarization of macrophages in endometrial cancer through targeting miR-146a. Int J Biochem Cell Biol. 2018;104:25–33. https://doi.org/10.1016/j.biocel.2018.08.017.

    Article  CAS  PubMed  Google Scholar 

  90. Kong F, Deng X, Kong X, Du Y, Li L, Zhu H, et al. ZFPM2-AS1, a novel lncRNA, attenuates the p53 pathway and promotes gastric carcinogenesis by stabilizing MIF. Oncogene. 2018;37(45):5982–96. https://doi.org/10.1038/s41388-018-0387-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Rajabinejad M, Salari F, Gorgin Karaji A, Rezaiemanesh A. The role of myeloid-derived suppressor cells in the pathogenesis of rheumatoid arthritis; anti- or pro-inflammatory cells? Artif Cells Nanomed Biotechnol. 2019;47(1):4149–58. https://doi.org/10.1080/21691401.2019.1687504.

    Article  CAS  PubMed  Google Scholar 

  92. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9(3):162–74. https://doi.org/10.1038/nri2506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Law AMK, Valdes-Mora F, Gallego-Ortega D. Myeloid-derived suppressor cells as a therapeutic target for cancer. Cells. 2020;9(3):561. https://doi.org/10.3390/cells9030561.

    Article  CAS  PubMed Central  Google Scholar 

  94. Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021:1–14. https://doi.org/10.1038/s41577-020-00490-y.

  95. Hatziioannou A, Alissafi T, Verginis P. Myeloid-derived suppressor cells and T regulatory cells in tumors: unraveling the dark side of the force. J Leukoc Biol. 2017;102(2):407–21. https://doi.org/10.1189/jlb.5VMR1116-493R.

    Article  CAS  PubMed  Google Scholar 

  96. Ma P, Beatty PL, McKolanis J, Brand R, Schoen RE, Finn OJ. Circulating myeloid derived suppressor cells (MDSC) that Accumulate in Premalignancy Share Phenotypic and Functional Characteristics With MDSC in Cancer. Front Immunol. 2019;10:1401. https://doi.org/10.3389/fimmu.2019.01401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Shang W, Gao Y, Tang Z, Zhang Y, Yang R. The pseudogene Olfr29-ps1 promotes the suppressive function and differentiation of monocytic MDSCs. Cancer Immunol Res. 2019;7(5):813–27. https://doi.org/10.1158/2326-6066.CIR-18-0443.

    Article  CAS  PubMed  Google Scholar 

  98. Gao Y, Wang T, Li Y, Zhang Y, Yang R. Lnc-chop promotes immunosuppressive function of myeloid-derived suppressor cells in tumor and inflammatory environments. J Immunol. 2018;200(8):2603–14. https://doi.org/10.4049/jimmunol.1701721.

    Article  CAS  PubMed  Google Scholar 

  99. Shang W, Tang Z, Gao Y, Qi H, Su X, Zhang Y, et al. LncRNA RNCR3 promotes Chop expression by sponging miR-185-5p during MDSC differentiation. Oncotarget. 2017;8(67):111754–69. https://doi.org/10.18632/oncotarget.22906.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Tian X, Ma J, Wang T, Tian J, Zheng Y, Peng R, et al. Long non-coding RNA RUNXOR accelerates MDSC-mediated immunosuppression in lung cancer. BMC Cancer. 2018;18(1):660. https://doi.org/10.1186/s12885-018-4564-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Zheng Y, Tian X, Wang T, Xia X, Cao F, Tian J, et al. Long noncoding RNA Pvt1 regulates the immunosuppression activity of granulocytic myeloid-derived suppressor cells in tumor-bearing mice. Mol Cancer. 2019;18(1):61. https://doi.org/10.1186/s12943-019-0978-2.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Tian X, Zheng Y, Yin K, Ma J, Tian J, Zhang Y, et al. LncRNA AK036396 inhibits maturation and accelerates immunosuppression of polymorphonuclear myeloid-derived suppressor cells by enhancing the stability of Ficolin B. Cancer Immunol Res. 2020;8(4):565–77. https://doi.org/10.1158/2326-6066.CIR-19-0595.

    Article  CAS  PubMed  Google Scholar 

  103. Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell. 2011;147(4):728–41. https://doi.org/10.1016/j.cell.2011.10.026.

    Article  CAS  PubMed  Google Scholar 

  104. Kirkegaard K, Taylor MP, Jackson WT. Cellular autophagy: surrender, avoidance and subversion by microorganisms. Nat Rev Microbiol. 2004;2(4):301–14. https://doi.org/10.1038/nrmicro865.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Nardacci R, Ciccosanti F, Marsella C, Ippolito G, Piacentini M, Fimia GM. Role of autophagy in HIV infection and pathogenesis. J Intern Med. 2017;281(5):422–32. https://doi.org/10.1111/joim.12596.

    Article  CAS  PubMed  Google Scholar 

  106. Jiang GM, Tan Y, Wang H, Peng L, Chen HT, Meng XJ, et al. The relationship between autophagy and the immune system and its applications for tumor immunotherapy. Mol Cancer. 2019;18(1):17. https://doi.org/10.1186/s12943-019-0944-z.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Yao H, Han B, Zhang Y, Shen L, Huang R. Non-coding RNAs and Autophagy. Adv Exp Med Biol. 2019;1206:199–220. https://doi.org/10.1007/978-981-15-0602-4_10.

    Article  CAS  PubMed  Google Scholar 

  108. Yang L, Wang H, Shen Q, Feng L, Jin H. Long non-coding RNAs involved in autophagy regulation. Cell Death Dis. 2017;8(10):e3073. https://doi.org/10.1038/cddis.2017.464.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Xue Z, Zhang Z, Liu H, Li W, Guo X, Zhang Z, et al. lincRNA-Cox2 regulates NLRP3 inflammasome and autophagy mediated neuroinflammation. Cell Death Differ. 2019;26(1):130–45. https://doi.org/10.1038/s41418-018-0105-8.

    Article  CAS  PubMed  Google Scholar 

  110. Li Z, Li J, Tang N. Long noncoding RNA Malat1 is a potent autophagy inducer protecting brain microvascular endothelial cells against oxygen-glucose deprivation/reoxygenation-induced injury by sponging miR-26b and upregulating ULK2 expression. Neuroscience. 2017;354:1–10. https://doi.org/10.1016/j.neuroscience.2017.04.017.

    Article  CAS  PubMed  Google Scholar 

  111. Li ZX, Zhu QN, Zhang HB, Hu Y, Wang G, Zhu YS. MALAT1: a potential biomarker in cancer. Cancer Manag Res. 2018;10:6757–68. https://doi.org/10.2147/CMAR.S169406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Fu Z, Luo W, Wang J, Peng T, Sun G, Shi J, et al. Malat1 activates autophagy and promotes cell proliferation by sponging miR-101 and upregulating STMN1, RAB5A and ATG4D expression in glioma. Biochem Biophys Res Commun. 2017;492(3):480–6. https://doi.org/10.1016/j.bbrc.2017.08.070.

    Article  CAS  PubMed  Google Scholar 

  113. Chen ZH, Wang WT, Huang W, Fang K, Sun YM, Liu SR, et al. The lncRNA HOTAIRM1 regulates the degradation of PML-RARA oncoprotein and myeloid cell differentiation by enhancing the autophagy pathway. Cell Death Differ. 2017;24(2):212–24. https://doi.org/10.1038/cdd.2016.111.

    Article  CAS  PubMed  Google Scholar 

  114. Yu Y, Zhang W, Li A, Chen Y, Ou Q, He Z, et al. Association of long noncoding RNA biomarkers with clinical immune subtype and prediction of immunotherapy response in patients with cancer. JAMA Netw Open. 2020;3(4):e202149. https://doi.org/10.1001/jamanetworkopen.2020.2149.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Song B, Guan Z, Liu F, Sun D, Wang K, Qu H. Long non-coding RNA HOTAIR promotes HLA-G expression via inhibiting miR-152 in gastric cancer cells. Biochem Biophys Res Commun. 2015;464(3):807–13. https://doi.org/10.1016/j.bbrc.2015.07.040.

    Article  CAS  PubMed  Google Scholar 

  116. Osses DF, Roobol MJ, Schoots IG. Prediction medicine: Biomarkers, risk calculators and magnetic resonance imaging as risk stratification tools in prostate cancer diagnosis. Int J Mol Sci. 2019;20(7):1637. https://doi.org/10.3390/ijms20071637.

    Article  CAS  PubMed Central  Google Scholar 

  117. Sartori DA, Chan DW. Biomarkers in prostate cancer: what’s new? Curr Opin Oncol. 2014;26(3):259–64. https://doi.org/10.1097/CCO.0000000000000065.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Zhang R, Xia Y, Wang Z, Zheng J, Chen Y, Li X, et al. Serum long non coding RNA MALAT-1 protected by exosomes is up-regulated and promotes cell proliferation and migration in non-small cell lung cancer. Biochem Biophys Res Commun. 2017;490(2):406–14. https://doi.org/10.1016/j.bbrc.2017.06.055 Epub 2017 Jun 13. PMID: 28623135.

    Article  CAS  PubMed  Google Scholar 

  119. Subhash S, Andersson PO, Kosalai ST, Kanduri C, Kanduri M. Global DNA methylation profiling reveals new insights into epigenetically deregulated protein coding and long noncoding RNAs in CLL. Clin Epigenetics. 2016;8:106. https://doi.org/10.1186/s13148-016-0274-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Guo G, Kang Q, Zhu X, Chen Q, Wang X, Chen Y, et al. A long noncoding RNA critically regulates Bcr-Abl-mediated cellular transformation by acting as a competitive endogenous RNA. Oncogene. 2015;34(14):1768–79. https://doi.org/10.1038/onc.2014.131.

    Article  CAS  PubMed  Google Scholar 

  121. Chen L, Wang W, Cao L, Li Z, Wang X. Long non-coding RNA CCAT1 acts as a competing endogenous RNA to regulate cell growth and differentiation in acute myeloid leukemia. Mol Cell. 2016;39(4):330–6. https://doi.org/10.14348/molcells.2016.2308.

    Article  CAS  Google Scholar 

  122. Hirano T, Yoshikawa R, Harada H, Harada Y, Ishida A, Yamazaki T. Long noncoding RNA, CCDC26, controls myeloid leukemia cell growth through regulation of KIT expression. Mol Cancer. 2015;14:90. https://doi.org/10.1186/s12943-015-0364-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Wang XQ, Crutchley JL, Dostie J. Shaping the genome with non-coding RNAs. Curr Genomics. 2011;12(5):307–21. https://doi.org/10.2174/138920211796429772.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Lerner M, Harada M, Lovén J, Castro J, Davis Z, Oscier D, et al. DLEU2, frequently deleted in malignancy, functions as a critical host gene of the cell cycle inhibitory microRNAs miR-15a and miR-16-1. Exp Cell Res. 2009;315(17):2941–52. https://doi.org/10.1016/j.yexcr.2009.07.001.

    Article  CAS  PubMed  Google Scholar 

  125. Kasar S, Underbayev C, Yuan Y, Hanlon M, Aly S, Khan H, et al. Therapeutic implications of activation of the host gene (Dleu2) promoter for miR-15a/16-1 in chronic lymphocytic leukemia. Oncogene. 2014;33(25):3307–15. https://doi.org/10.1038/onc.2013.291.

    Article  CAS  PubMed  Google Scholar 

  126. Morenos L, Chatterton Z, Ng JL, Halemba MS, Parkinson-Bates M, Mechinaud F, et al. Hypermethylation and down-regulation of DLEU2 in paediatric acute myeloid leukaemia independent of embedded tumour suppressor miR-15a/16-1. Mol Cancer. 2014;13:123. https://doi.org/10.1186/1476-4598-13-123.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Mertens D, Philippen A, Ruppel M, Allegra D, Bhattacharya N, Tschuch C, et al. Chronic lymphocytic leukemia and 13q14: miRs and more. Leuk Lymphoma. 2009;50(3):502–5. https://doi.org/10.1080/10428190902763509.

    Article  CAS  PubMed  Google Scholar 

  128. Barwick BG, Gupta VA, Vertino PM, Boise LH. Cell of origin and genetic alterations in the pathogenesis of multiple myeloma. Front Immunol. 2019;10:1121. https://doi.org/10.3389/fimmu.2019.01121.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Dinger ME, Amaral PP, Mercer TR, Pang KC, Bruce SJ, Gardiner BB, et al. Long noncoding RNAs in mouse embryonic stem cell pluripotency and differentiation. Genome Res. 2008;18(9):1433–45. https://doi.org/10.1101/gr.078378.108.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Mourtada-Maarabouni M, Pickard MR, Hedge VL, Farzaneh F, Williams GT. GAS5, a non-protein-coding RNA, controls apoptosis and is downregulated in breast cancer. Oncogene. 2009;28(2):195–208. https://doi.org/10.1038/onc.2008.373.

    Article  CAS  PubMed  Google Scholar 

  131. Nakamura Y, Takahashi N, Kakegawa E, Yoshida K, Ito Y, Kayano H, et al. The GAS5 (growth arrest-specific transcript 5) gene fuses to BCL6 as a result of t(1;3)(q25;q27) in a patient with B-cell lymphoma. Cancer Genet Cytogenet. 2008;182(2):144–9. https://doi.org/10.1016/j.cancergencyto.2008.01.013 PMID: 18406879.

    Article  CAS  PubMed  Google Scholar 

  132. Mourtada-Maarabouni M, Williams GT. Role of GAS5 noncoding RNA in mediating the effects of rapamycin and its analogues on mantle cell lymphoma cells. Clin Lymphoma Myeloma Leuk. 2014;14(6):468–73. https://doi.org/10.1016/j.clml.2014.02.011 Epub 2014 Mar 3. PMID: 24703244.

    Article  PubMed  Google Scholar 

  133. Zhang YY, Huang SH, Zhou HR, Chen CJ, Tian LH, Shen JZ. Role of HOTAIR in the diagnosis and prognosis of acute leukemia. Oncol Rep. 2016;36(6):3113–22. https://doi.org/10.3892/or.2016.5147.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Oh EJ, Kim SH, Yang WI, Ko YH, Yoon SO. Long non-coding RNA HOTAIR expression in diffuse large B-cell lymphoma: In relation to polycomb repressive complex pathway proteins and H3K27 trimethylation. J Pathol Transl Med. 2016;50(5):369–76. https://doi.org/10.4132/jptm.2016.06.06.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Yan Y, Han J, Li Z, Yang H, Sui Y, Wang M. Elevated RNA expression of long non‑coding HOTAIR promotes cell proliferation and predicts a poor prognosis in patients with diffuse large B cell lymphoma. Mol Med Rep. 2016;13(6):5125–31. https://doi.org/10.3892/mmr.2016.5190.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Wang XQ, Dostie J. Reciprocal regulation of chromatin state and architecture by HOTAIRM1 contributes to temporal collinear HOXA gene activation. Nucleic Acids Res. 2017;45(3):1091–104. https://doi.org/10.1093/nar/gkw966.

    Article  CAS  PubMed  Google Scholar 

  137. Zhao H, Zhang X, Frazão JB, Condino-Neto A, Newburger PE. HOX antisense lincRNA HOXA-AS2 is an apoptosis repressor in all trans retinoic acid treated NB4 promyelocytic leukemia cells. J Cell Biochem. 2013;114(10):2375–83. https://doi.org/10.1002/jcb.24586.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Peng W, Wu J, Feng J. Long noncoding RNA HULC predicts poor clinical outcome and represents pro-oncogenic activity in diffuse large B-cell lymphoma. Biomed Pharmacother. 2016;79:188–93. https://doi.org/10.1016/j.biopha.2016.02.032.

    Article  CAS  PubMed  Google Scholar 

  139. Mangiavacchi A, Sorci M, Masciarelli S, Larivera S, Legnini I, Iosue I, et al. The miR-223 host non-coding transcript linc-223 induces IRF4 expression in acute myeloid leukemia by acting as a competing endogenous RNA. Oncotarget. 2016;7(37):60155–68. https://doi.org/10.18632/oncotarget.11165.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Ghazavi F, De Moerloose B, Van Loocke W, Wallaert A, Helsmoortel HH, Ferster A, et al. Unique long non-coding RNA expression signature in ETV6/RUNX1-driven B-cell precursor acute lymphoblastic leukemia. Oncotarget. 2016;7(45):73769–80. https://doi.org/10.18632/oncotarget.12063.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Conde L, Riby J, Zhang J, Bracci PM, Skibola CF. Copy number variation analysis on a non-Hodgkin lymphoma case-control study identifies an 11q25 duplication associated with diffuse large B-cell lymphoma. PLoS One. 2014;9(8):e105382. https://doi.org/10.1371/journal.pone.0105382.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Trimarchi T, Bilal E, Ntziachristos P, Fabbri G, Dalla-Favera R, Tsirigos A, et al. Genome-wide mapping and characterization of Notch-regulated long noncoding RNAs in acute leukemia. Cell. 2014;158(3):593–606. https://doi.org/10.1016/j.cell.2014.05.049.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Peng W, Feng J. Long noncoding RNA LUNAR1 associates with cell proliferation and predicts a poor prognosis in diffuse large B-cell lymphoma. Biomed Pharmacother. 2016;77:65–71. https://doi.org/10.1016/j.biopha.2015.12.001.

    Article  CAS  PubMed  Google Scholar 

  144. Benetatos L, Hatzimichael E, Dasoula A, Dranitsaris G, Tsiara S, Syrrou M, et al. CpG methylation analysis of the MEG3 and SNRPN imprinted genes in acute myeloid leukemia and myelodysplastic syndromes. Leuk Res. 2010;34(2):148–53. https://doi.org/10.1016/j.leukres.2009.06.019.

    Article  CAS  PubMed  Google Scholar 

  145. Zhang X, Zhou Y, Mehta KR, Danila DC, Scolavino S, Johnson SR, et al. A pituitary-derived MEG3 isoform functions as a growth suppressor in tumor cells. J Clin Endocrinol Metab. 2003;88(11):5119–26. https://doi.org/10.1210/jc.2003-030222.

    Article  CAS  PubMed  Google Scholar 

  146. Zhuang W, Ge X, Yang S, Huang M, Zhuang W, Chen P, et al. Upregulation of lncRNA MEG3 promotes osteogenic differentiation of mesenchymal stem cells from multiple myeloma patients by targeting BMP4 transcription. Stem Cells. 2015;33(6):1985–97. https://doi.org/10.1002/stem.1989.

    Article  CAS  PubMed  Google Scholar 

  147. Doose G, Haake A, Bernhart SH, López C, Duggimpudi S, Wojciech F, et al. MINCR is a MYC-induced lncRNA able to modulate MYC’s transcriptional network in Burkitt lymphoma cells. Proc Natl Acad Sci U S A. 2015;112(38):E5261–70. https://doi.org/10.1073/pnas.1505753112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Emmrich S, Streltsov A, Schmidt F, Thangapandi VR, Reinhardt D, Klusmann JH. LincRNAs MONC and MIR100HG act as oncogenes in acute megakaryoblastic leukemia. Mol Cancer. 2014;13:171. https://doi.org/10.1186/1476-4598-13-171.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Wang Y, Wu P, Lin R, Rong L, Xue Y, Fang Y. LncRNA NALT interaction with NOTCH1 promoted cell proliferation in pediatric T cell acute lymphoblastic leukemia. Sci Rep. 2015;5:13749. https://doi.org/10.1038/srep13749.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Gao C, Zhang J, Wang Q, Ren C. Overexpression of lncRNA NEAT1 mitigates multidrug resistance by inhibiting ABCG2 in leukemia. Oncol Lett. 2016;12(2):1051–7. https://doi.org/10.3892/ol.2016.4738.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Zeng C, Yu X, Lai J, Yang L, Chen S, Li Y. Overexpression of the long non-coding RNA PVT1 is correlated with leukemic cell proliferation in acute promyelocytic leukemia. J Hematol Oncol. 2015;8:126. https://doi.org/10.1186/s13045-015-0223-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Nagoshi H, Taki T, Hanamura I, Nitta M, Otsuki T, Nishida K, et al. Frequent PVT1 rearrangement and novel chimeric genes PVT1-NBEA and PVT1-WWOX occur in multiple myeloma with 8q24 abnormality. Cancer Res. 2012;72(19):4954–62. https://doi.org/10.1158/0008-5472.CAN-12-0213.

    Article  CAS  PubMed  Google Scholar 

  153. Carramusa L, Contino F, Ferro A, Minafra L, Perconti G, Giallongo A, et al. The PVT-1 oncogene is a Myc protein target that is overexpressed in transformed cells. J Cell Physiol. 2007;213(2):511–8. https://doi.org/10.1002/jcp.21133.

    Article  CAS  PubMed  Google Scholar 

  154. Enciso-Mora V, Broderick P, Ma Y, Jarrett RF, Hjalgrim H, Hemminki K, et al. A genome-wide association study of Hodgkin's lymphoma identifies new susceptibility loci at 2p16.1 (REL), 8q24.21 and 10p14 (GATA3). Nat Genet. 2010;42(12):1126–30. https://doi.org/10.1038/ng.696.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Graham M, Adams JM. Chromosome 8 breakpoint far 3’ of the c-myc oncogene in a Burkitt’s lymphoma 2;8 variant translocation is equivalent to the murine pvt-1 locus. EMBO J. 1986;5(11):2845–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Shtivelman E, Henglein B, Groitl P, Lipp M, Bishop JM. Identification of a human transcription unit affected by the variant chromosomal translocations 2;8 and 8;22 of Burkitt lymphoma. Proc Natl Acad Sci U S A. 1989;86(9):3257–60. https://doi.org/10.1073/pnas.86.9.3257.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Tsutsumi Y, Chinen Y, Sakamoto N, Nagoshi H, Nishida K, Kobayashi S, et al. Deletion or methylation of CDKN2A/2B and PVT1 rearrangement occur frequently in highly aggressive B-cell lymphomas harboring 8q24 abnormality. Leuk Lymphoma. 2013;54(12):2760–4. https://doi.org/10.3109/10428194.2013.790543.

    Article  CAS  PubMed  Google Scholar 

  158. Tanaka R, Satoh H, Moriyama M, Satoh K, Morishita Y, Yoshida S, et al. Intronic U50 small-nucleolar-RNA (snoRNA) host gene of no protein-coding potential is mapped at the chromosome breakpoint t(3;6)(q27;q15) of human B-cell lymphoma. Genes Cells. 2000;5(4):277–87. https://doi.org/10.1046/j.1365-2443.2000.00325.x.

    Article  CAS  PubMed  Google Scholar 

  159. Zhang L, Xu HG, Lu C. A novel long non-coding RNA T-ALL-R-LncR1 knockdown and Par-4 cooperate to induce cellular apoptosis in T-cell acute lymphoblastic leukemia cells. Leuk Lymphoma. 2014;55(6):1373–82. https://doi.org/10.3109/10428194.2013.829574.

    Article  CAS  PubMed  Google Scholar 

  160. Saitou M, Sugimoto J, Hatakeyama T, Russo G, Isobe M. Identification of the TCL6 genes within the breakpoint cluster region on chromosome 14q32 in T-cell leukemia. Oncogene. 2000;19(23):2796–802. https://doi.org/10.1038/sj.onc.1203604.

    Article  CAS  PubMed  Google Scholar 

  161. Wong KY, Li Z, Zhang X, Leung GK, Chan GC, Chim CS. Epigenetic silencing of a long non-coding RNA KIAA0495 in multiple myeloma. Mol Cancer. 2015;14:175. https://doi.org/10.1186/s12943-015-0444-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Coccaro N, Zagaria A, Tota G, Anelli L, Orsini P, Casieri P, et al. Overexpression of the LSAMP and TUSC7 genes in acute myeloid leukemia following microdeletion/duplication of chromosome 3. Cancer Genet. 2015;208(10):517–22. https://doi.org/10.1016/j.cancergen.2015.07.006.

    Article  CAS  PubMed  Google Scholar 

  163. Hughes JM, Legnini I, Salvatori B, Masciarelli S, Marchioni M, Fazi F, et al. C/EBPα-p30 protein induces expression of the oncogenic long non-coding RNA UCA1 in acute myeloid leukemia. Oncotarget. 2015;6(21):18534–44. https://doi.org/10.18632/oncotarget.4069.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Plass C, Yu F, Yu L, Strout MP, El-Rifai W, Elonen E, et al. Restriction landmark genome scanning for aberrant methylation in primary refractory and relapsed acute myeloid leukemia; involvement of the WIT-1 gene. Oncogene. 1999;18(20):3159–65. https://doi.org/10.1038/sj.onc.1202651.

    Article  CAS  PubMed  Google Scholar 

  165. Dallosso AR, Hancock AL, Malik S, Salpekar A, King-Underwood L, Pritchard-Jones K, et al. Alternately spliced WT1 antisense transcripts interact with WT1 sense RNA and show epigenetic and splicing defects in cancer. RNA. 2007;13(12):2287–99. https://doi.org/10.1261/rna.562907.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Zeng MN, Ma WL. Zheng WL, Bioinformatics analysis of microRNA comprehensive regulatory network in B- cell acute lymphoblastic leukemia. 2016;37(7):585–90. https://doi.org/10.3760/cma.j.issn.0253-2727.2016.07.009.

  167. The LncRNA and Disease Database. http://www.cuilab.cn/lncrnadisease.

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by grants from the National Natural Science Foundation of China (81872772, 81960546, 81700169, U1812403, 21867009, and U1812403-4-4), The Science and Technology Department of Guizhou Province (QKHJC[2018]1409, QKHZC [2019]2762, QKHPTRC[2020]5008 and QKHZC[2020]4Y203), The 100 Leading Talents of Guizhou Province (fund for Y.M.L) P2018-KF11, QZYY-2019-022, The Base of International Scientific and Technological Cooperation of Guizhou Province ([2017]5802), Guizhou Provincial Talent Base for Druggability from Natural Medicine ([2018]3), and the Fund of High-Level Innovation Talents of Guizhou (2015-4029) (fund for X.C.S).

Author information

Authors and Affiliations

Authors

Contributions

KMV, HD, and BG wrote the original draft, prepared tables, figures and corrected the manuscript; WL, JS, CW, and AH provided critical suggestions; YBD, XS, YL, and BG revised the paper. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Yaacov Ben-David, Xiangchun Shen, Yanmei Li or Babu Gajendran.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Varier, K.M., Dhandapani, H., Liu, W. et al. An immunotherapeutic approach to decipher the role of long non-coding RNAs in cancer progression, resistance and epigenetic regulation of immune cells. J Exp Clin Cancer Res 40, 242 (2021). https://doi.org/10.1186/s13046-021-01997-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13046-021-01997-5

Keywords