Abstract
It is well known that long non-coding RNAs (lncRNAs) play an important role in the regulation of tumor genesis and development. They can modulate gene expression of transcriptional regulation, epigenetic regulation of chromatin modification, and post-transcriptional regulation, thus influencing the biological behavior of tumors, such as cell proliferation, apoptosis, cell cycle, invasion, and migration. Tumor angiogenesis not only provides nutrients and helps excrete metabolites, but it also opens a pathway for tumor metastasis. Anti-angiogenic therapy has become one of the effective treatment methods for tumor. But its drug resistance leads to the limitation of clinical application. Recent studies have shown that lncRNAs are closely related to tumor angiogenesis and anti-angiogenic therapy resistance, which provides a new direction for tumor research. lncRNAs are expected to be new targets for tumor therapy. For the first time to our knowledge, this paper reviews advancement of lncRNAs in tumor angiogenesis and anti-angiogenic therapy resistance and further discusses their potential clinical application.
Keywords: MT: non-coding RNAs, long non-coding RNAs, angiogenesis, anti-angiogenic therapy, drug resistance, cancer stem cells, cancer
Graphical abstract
lncRNAs play an important role in tumor genesis and development. This paper systematically reviews the role of lncRNAs in tumor angiogenesis and antiangiogenic therapy resistance. It not only reveals the potential of lncRNAs as tumor biomarkers and therapeutic targets, but it also provides a new idea for the treatment of tumor.
Introduction
Currently, cancer is a major public health problem worldwide and results in heavy social and economic burden.1 The latest Global Cancer Date Report indicates that an estimated 19.3 million new cancer cases and nearly 10 million cancer deaths occurred worldwide in 2020.2 The occurrence and development of tumors is an intricate process affected by multiple factors and manifested in multiple stages, among which angiogenesis plays an insignificant role.3 Angiogenesis is a commonality of almost all solid tumors, concerned with tumor grade, malignancy, and poor clinical outcomes.4,5 Anti-angiogenic therapy has come into being. The combination of anti-angiogenic therapy and other treatment methods has been proven effective.6, 7, 8 Whereas, many studies have shown that anti-angiogenic therapy, like other anti-tumor drugs, inevitably develops drug resistance.9 It has been discovered that about three-quarters of the human genome can be transcribed, but nearly 98% of the human genome does not encode proteins, and only 2% does.10 Non-coding RNAs (ncRNAs) without protein-coding function are considered to be non-functional evolutionary junk.11 As gene sequence technology has developed rapidly, the lncRNA, which is a type of ncRNA with a length of more than 200 nucleotides, has been extensively studied over the past decade. Examples of lncRNAs regulating tumor and therapeutic resistance are nothing new.12, 13, 14, 15, 16 Our research group found LINC01234 was significantly upregulated in gastric cancer (GC) and non-small cell lung cancer (NSCLC), associated with poor prognosis. LINC01234 upregulated CBFB via sponge miR-204-5p to boost the occurrence and development of GC.17 A positive feedback loop, c-Myc–LINC01234–HNRNPA2B1–miR-106b-5p–CRY2–c-Myc, was crucial to the progression of NSCLC.18 It also promoted NSCLC metastasis by activating VAV3 as a miR-27b-3p/miR-340-5p ceRNA or directly inhibiting BTG2.19 In addition, lncRNA CASC9 promoted NSCLC resistance to gefitinib by recruiting EZH2 to inhibit DUSP1.20 With the deep-going research, the function of lncRNAs in angiogenesis and anti-angiogenic therapy resistance has been revealed in cancer.21,22 Here, we first summarize the roles of lncRNAs in tumor angiogenesis and anti-angiogenic therapy resistance.
lncRNAs
The lncRNA is mainly transcribed from the antisense chains and spacers of protein-coding genes. Compared with protein-coding RNAs, lncRNAs are expressed at a lower level but are still characterized by tissue and cell specificity, high interspecific conservation, and high coefficient of variation.23 lncRNAs are involved in biological processes through adjustment of gene expression, including transcriptional regulation, epigenetic regulation of chromatin modification, and post-transcriptional regulation.24, 25, 26, 27, 28, 29 For instance, lncRNA H19 stimulated osteogenic differentiation of bone marrow mesenchymal stems cells by means of targeting the miR-149/SDF-1 axis.30 Importantly, lncRNAs are also involved in many pathological processes to influence many diseases,25 especially tumors.31 According to their effects, they can be divided into oncogenes and suppressor genes. lncRNA VCAN-AS1 promotes the progression of GC by interacting with eIF4A3 to downregulate p53.32 On the contrary, lncRNA GAS5 inhibits the metastasis of GC via positively modulating p53.33 Firstly, lncRNAs are expressed in almost all types of cancers. Secondly, lncRNAs are expressed variously in the same tumor tissue or cell. Using gene chip technology, He et al. found 2,669 upregulated and 3,506 downregulated lncRNAs in GC.34 And last, the expression levels of the same lncRNA are different in disparate tumors. H19 is upregulated in GC,35 breast cancer,36 colorectal cancer (CRC),37 cholangiocarcinoma,38 and NSCLC.39 But it is downregulated in papillary thyroid carcinoma.40
Angiogenesis and anti-angiogenic therapy
In 1971, Folkman first proposed the hypothesis that tumor growth and metastasis depended on angiogenesis.41 Angiogenesis is a highly dynamic, sophisticated process. New blood vessels are generated from existing germinated or ungerminated vascular beds, including degradation of vascular basement membranes; activation, proliferation, and migration of vascular endothelial cells; and reconstruction of new blood vessels and vascular networks.
Tumor micro environment (TME) refers to the local internal environment composed of locally infiltrated immune cells, mesenchymal cells, extracellular matrix, and active mediators, together with tumor cells and tumor stem cells, namely the site of angiogenesis.42,43 Due to the rapid growth of tumor cells, the blood supply cannot meet the growth needs of tumor cells, leading to hypoxia and acidosis in the TME. Hypoxia induces cells to produce proangiogenic factors, which turn on the switch of tumor angiogenesis.44 The continuous secretion of proangiogenic factors in the TME leads to abnormal vascular structure with a circuitous leakage state, and ultimately it results in poor blood perfusion of the tumor. This result further aggravates hypoxia and acidosis in the TME. Abnormal tumor blood vessels cause the immunosuppressive state of TME.45 A large number of immunosuppressive cells and factors are released and collected, including myeloid-derived suppressor cells (MDSCs), dendritic cells (DCs), M2 phenotype of tumor associated macrophages (TAMs), T regulatory cells (Tregs), IL-10, TGF-β, and so on. The increased expression of PD-L1 in tumor cells inhibited the infiltration of cytotoxic T lymphocyte (CTL) to tumor cells. The cytotoxic effect of NK cells is reduced or even inactivated. The differentiation and maturation of DCs are inhibited, and the antigen presenting ability is decreased. The immunosuppressive state of TME eventually gives rise to immune escape of tumor cells. In addition, hypoxia can induce dedifferentiation of tumor cells and enhance the stem cell phenotype of tumor cells.46 Anti-angiogenic therapy can inhibit angiogenesis and normalize abnormal tumor blood vessels. Vascular normalization can reprogram the immunosuppressive state into an immune activated state, and the activation of immune cells can in turn promote vascular normalization, thus forming a good positive feedback loop (Figure 1).47,48 It is the theoretical basis for anti-angiogenic therapy in combination with immune checkpoint inhibitors in the treatment of malignancies.49
Figure 1.
Relationship between tumor angiogenesis and immune microenvironment before and after anti-angiogenic therapy
In February 2004, the US FDA authorized bevacizumab in combination with 5-FU for the first-line treatment of metastatic colorectal cancer.50 Up to now, angiogenesis theory and inhibitors have gained widespread acceptance. At present, anti-angiogenic drugs are divided into three categories: (1) macromolecular monoclonal antibodies (bevacizumab, ramucirumab), (2) small molecule multi-target anti-angiogenic drugs (sorafenib, sunitinib, and so on), and (3) endostatin (endostar). Anti-angiogenic therapy is more effective in combination with other anti-tumor therapies, especially immunotherapy. Compared with bevacizumab + chemotherapy, atezolizumab + bevacizumab + chemotherapy significantly improved progression-free survival (PFS) and overall survival (OS) of patients with metastatic non-squamous NSCLC, no matter what PD-L1 and EGFR or ALK gene altered status.51 Choueiri et al. first reported that avelumab combined with axitinib improved PFS in patients with advanced clear cell renal cancer (RC).52 However, the resistance of anti-angiogenic therapy critically imposes restrictions on its clinical application.53,54
Correlation between lncRNAs and angiogenesis in cancer
lncRNAs can regulate angiogenesis in cardiovascular disease,55,56 diabetes,57,58 and even tumors. It is common knowledge that the vascular endothelial growth factors (VEGF), Notch, and angiogenin (Ang) signaling pathways are the vital signaling pathways involved in angiogenesis.59 lncRNAs are closely connected with angiogenesis and may regulate tumor angiogenesis (Figure 2, Table 1).
Figure 2.
The partial mechanisms of lncRNAs involved in tumor angiogenesis and resistance to anti-angiogenic therapy
Table 1.
Correlation between lncRNAs and angiogenesis in tumors
Classification | LncRNA | Cancer | Expression | Mechanism | Biological functions | Ref |
---|---|---|---|---|---|---|
CeRNA | LINC00173.v1 | lung squamous cell carcinoma | up | ΔNp63α/LINC00173.v1/miR-511-5p/VEGFA axis | promoting tumorigenesis and angiogenesis | 60 |
MYLK-AS1 | hepatocellular carcinoma | up | miR-424-5p/E2F7/VEGFR-2 axis | promoting proliferation, invasion, migration, and angiogenesis | 61 | |
MALAT1 | up | miR-140/VEGFA axis | promoting angiogenesis and changing polarization of macrophage | 62 | ||
SNHG17 | colorectal carcinoma | up | miR-23a-3p/CXCL12 axis | promoting viability proliferation, migration, and angiogenesis | 63 | |
SNHG22 | gastric carcinoma | up | miR-361-3p/HMGA1/Wnt/β-catenin axis | promoting proliferation, angiogenesis, and transition from G1 phase to S phase; inhibition of apoptosis | 64 | |
TUG1 | osteosarcoma | up | miR-143-5p/HIF-1α | promoting invasion and angiogenesis | 65 | |
Signaling pathway | SRRM2-AS | nasopharyngeal carcinoma | up | MYLK/cGMP/PKG axis | promoting proliferation, colony formation, angiogenesis; regulating cell cycle; inhibition apoptosis | 66 |
LINC01314 | gastric carcinoma | down | KLK4/Wnt/β-catenin axis | inhibiting migration, invasion, and angiogenesis | 67 | |
Angiogenic factors and receptors | EPIC1 | non-small cell lung cancer | up | Ang2/Tie2 axis | promoting angiogenesis | 68 |
Recruitment of RNA polymerase | RAB11B-AS1 | breast cancer | up | HIF-2/Rab11B-AS1/RNA Pol II/VEGFA and ANGPTL4 | promoting migration, invasion, angiogenesis, and distant metastasis | 69 |
Exosome | TUG1 | cervical cancer | up | exosome transfer | promoting angiogenesis | 70 |
Gene transcription | LINC00261 | prostate cancer | down | DKK3/GATA6/VEGF and CD31 | inhibiting proliferation, migration, invasion, tumorigenicity, and angiogenesis | 71 |
SLC26A4-AS1 | glioma | down | NFKB1/NPTX1 | Inhibiting cell viability, proliferation, migration, invasion and angiogenesis | 72 | |
LINC00320 | down | NFKB1/AQP9 | inhibit proliferation and angiogenesis | 73 | ||
Tumor associated macrophage | MALAT1 | thyroid cancer | up | TAMs FGF2 protein | promoting proliferation, migration, invasion, angiogenesis | 74 |
Cancer stem cells | H19 | hepatocellular carcinoma | up | exosome transfer | promoting tube formation and cell-cell adhesion | 75 |
Head and neck tumors
Nasopharyngeal carcinoma (NPC) with obvious regional aggregation, most frequently occurring in Southeast Asian countries, is a malignant tumor usually occurring in the mucosa and epithelium of the nasopharynx.76 Chen et al. reported a novel lncRNA SRRM2-AS that is regarded as a potential marker for angiogenesis in NPC. Overexpression of SRRM2-AS was confirmed in NPC tissues. Moreover, further investigations revealed that silencing SRRM2-AS inhibited angiogenesis in NPC by activating MYLK-mediated cGMP/PKG signaling pathway.66 It was reported that lncRNA MALAT1 might be a new biomarker for thyroid cancer. Fibroblast growth factors 2 (FGF2) is a VEGF-independent angiogenic factor.77 MALAT1 and FGF2 were dramatically overexpressed in thyroid cancer tissues and cells (FTC133). It was noteworthy that both of them also presented high expression in TAMs. The culture medium (CM) from TAMs can effectively promote the proliferation, migration, and invasion of FTC133 cells. The CM from M2 macrophages and TAMs effectively increased the angiogenesis of human umbilical vein endothelial cells (HUVECs). After downregulation of MALAT1 in TAMs, the CM could restrain proliferation, migration, and invasion of FTC133 cells and reduce angiogenesis. The same results were observed when FGF2 in TAMs was downregulated. Mechanism studies have found that MALAT1 promoted angiogenesis and enhanced cell proliferation, migration, and invasion by mediating FGF2 protein secretion of TAMs in thyroid cancer (Figure 2A).74
Respiratory tumors
Lung cancer remains one of the leading causes of death from malignancies worldwide, despite significant reductions in the death rate from lung cancer after intensive research and standardized treatment.2 The result of bioinformatics analysis implied LINC00173.v1 was mainly expressed in lung cancer tissues and significantly upregulated in lung squamous cell carcinoma. Overexpression of LINC00173.v1 was a symbol of poorer OS and PFS. This result was confirmed in situ hybridization. Silencing LINC00173.v1 inhibited the proliferation and migration of vascular endothelial cells in vivo and in vitro. The mechanism experiment proved that LINC00173.v1 increased the expression of VEGFA by sponging miR-511-5p to boost the proliferation and migration of vascular endothelial cells in lung squamous cell carcinoma. In a xenograft model of the mice, miR-511-5p/VEGFA axis may be the mechanism of LINCC00173.v1 inducing tumorigenesis. Moreover, inhibiting the expression of LINC00173.v1 has therapeutic effect in SQC. ΔNp63α is one of the p53/p63/p73 family transcription factors, which is essential to the occurrence,78 development, and even drug resistance of a variety of tumors.79 Further studies demonstrated that ΔNp63α raised the transcription of LINC00173.v1. These events suggested that LINC00173.v1 may be an underlying anti-angiogenic therapeutic target of lung squamous cell carcinoma (Figure 2B).60 Hou et al. found that lncRNA EPIC1 is upregulated in NSCLC cells and tissues. Mechanistically, EPIC1 enhanced tumor angiogenesis by activating the Ang2/Tie2 axis in NSCLC. Angiogenesis assay in chick chorioallantoic membranes (CAM) showed that overexpression of EPIC1 promoted CAM angiogenesis, while silencing EPIC1 inhibited CAM angiogenesis. A xenograft nude mice model confirmed that EPIC1 overexpression significantly increased tumor angiogenesis, the number of CD31 marked channels, and Ang2 levels. What is more, samples from patients with NSCLC also confirmed the results. EPIC1 might be a biomarker for angiogenesis in NSCLC.68
Digestive system tumors
The morbidity and mortality of various digestive system tumors are among the top 10 malignancies in the world.2 LINC01314(CTXND1) was low in GC by analyzed GC Chip Data GSE19826. In vivo and in vitro, upregulation of LINC01314 or downregulation of KLK4 reduced the proliferation, migration, invasion, and angiogenesis of GC cells and decreased micro-vessel density. Bioinformatic analysis showed that there was a specific binding site between KLK4 and LINC01314. Dual-luciferase reporter gene assay confirmed the result. The Wnt/β-catenin signaling pathway participated in the adjustment of various pathophysiological procedure.80, 81, 82 Abnormal Wnt/β-catenin signaling pathway can induce many diseases.83, 84, 85 In western blot analysis, the expression levels of KLK4, Wnt-1, β-catenin, Cyclin D1, and N-cadherin were decreased after the upregulation of LINC01314. Animal experiments showed that upregulation of LINC01314 or downregulation of KLK4 can inhibit tumor growth and reduce the micro-vessel density. The preceding results revealed that overexpressed LINC01314 can inhibit the Wnt/β-catenin signaling pathway by downregulating KLK4 to suppress the angiogenesis in GC.67 lncRNA SNHG22 can upregulate HMGA1 through sponge miR-361-3p to activate Wnt/β-catenin pathway, thereby promoting GC progression (Figure 2C).64 1,081 lncRNAs, 127 microRNAs, and 1,983 differentially expressed mRNAs were screened from RNA-seq data of hepatocellular carcinoma (HCC) patients in the TCGA database. More importantly, lncRNA MYLK-AS1/miR-424-5p/E2F7 axis was the only ceRNA regulatory axis negatively correlated with OS. MiR-424-5p has the dual effects of oncogene and tumor suppressor gene. In LSCC,86 esophageal cancer,87 CRC,88 and thyroid cancer,89 miR-424-5p mainly intensifies the proliferation, invasion, migration, and drug resistance of tumor cells. However, it has the opposite effect on liver,90 breast,91 and nasopharyngeal cancers.92 Mechanistically, MYLK-AS1 accelerated angiogenesis by regulating the miR-424-5p/E2F7 axis to activate the VEGFR-2 signaling pathway in HCC. This result was further confirmed in xenograft mice tumors.61 Hou et al. covered that miR-140/VEGFA axis may be the potential mechanism for regulating HCC angiogenesis by MALAT1.62 In colorectal adenocarcinoma (CRA), SNHG17 boosted CRA angiogenesis through regulating the miR-23a-3p/CXCL12 axis to induce proliferation and migration of CRA cells.63
Genitourinary system tumors
According to the latest data, breast cancer overtakes lung cancer as the most common malignancy.2 Niu et al. established breast cancer cell lines of different oxygen concentrations and found that hypoxia could induce upregulation of lncRNA RAB11B-AS1. HIF, a highly conserved transcription factor, can regulate the expression of multifarious genes responsible for specific physiological responses.93 ChIP-qPCR analysis showed that HIF-2 induced the overexpression of RAB11B-AS1 in hypoxia. Gain-of-function experiments demonstrated that upregulation of RAB11B-AS1 enhanced invasion and migration of breast cancer cells. The results were opposite in loss-of-function experiment. Animal studies showed that RAB11B-AS1 promoted angiogenesis and metastasis. Mechanistically, RAB11B-AS1 recruited RNA Pol II to enhance hypoxia-induced angiogenic factors, VEGFA, and ANGPTL4, thus motivating angiogenesis in breast cancer (Figure 2D).69 Lei et al. reported that lncRNA TUG1 was transferred by exosomes to the HUVECs to promote angiogenesis in cervical cancer (CC). TUG1 was meaningly upregulated in HeLa and CaSki cell lines and their exosomes. HUVECs treated with HeLa-Exo and CaSki-Exo significantly enhanced TUG1 levels and angiogenesis. Silencing TUG1 attenuated the angiogenic potential for HeLa-Exo and CaSki-Exo in HUVECs. As a consequence, TUG1 might be a therapeutic target gene for the CC (Figure 2E).70 In addition, Li et al. obtained 667 prostate cancer-related genes through analysis dataset GSE45016 in the GEO database. LINC00261 had the highest expression differential multiple and was underexpressed in both prostate cancer tissues and cells. Overexpression of LINC00261 inhibited proliferation, migration, invasion, and tube formation of prostate cancer cells. GATA6, a member of the GATA family, is essential for the development of early human organs. Its expression disorders are linked to many human diseases, including cancer.94 RIP experiment and ChIP assay revealed that LINC00261 could bind to GATA6, and GATA6 could be combined with DKK3 promoter. The expression of VEGF and CD31 decreased after LINC00261 or DKK3 overexpression. Tumorigenesis in nude mice and immunohistochemical staining supported the results. Altogether, LINC00261 promoted the expression of DKK3 by recruiting GATA6 to refrain proliferation, migration, invasion, and angiogenesis of prostate cancer.71
Other tumors
Glioma is the most common primary intracranial tumor with extensive angiogenesis. Neovascularization affords ideal conditions for tumor cells to infiltrate and migrate.95 It has been reported that lncRNAs are closely correlated with glioma angiogenesis.96 Analysis of database GSE104291 indicated that there were 91 genes with high expression and 268 genes with low expression.73 lncRNA SLC26A4-AS1 was meaningly downregulated in glioma tissues and cells. Upregulation of SLC26A4-AS1 in U251 and SHG44 cells inhibited the proliferation, migration, and angiogenesis of U251 and SHG44 cells. RIP and ChIP assays made clear that SLC26A4-AS1 upregulated NPTX1 by recruiting NFKB1 into the NPTX1 promoter. Silencing NPTX1 or NFKB1 partially reversed the invasive and proangiogenic properties of glioma cells induced by overexpression of SLC26A4-AS1. Animal experiment proved that SLC26A4-AS1 inhibited glioma tumor growth and angiogenesis by upregulation of NPTX1. Taken together, SLC26A4-AS1 might be a potential therapeutic target for glioma.72 However, LINC00320 inhibited AQP9 expression by recruiting NFKB1 to the promoter region of AQP9, thus acting as a tumor suppressor gene in glioma (Figure 2F).73 Osteosarcoma is a common and highly malignant bone tumor, mostly occurring in adolescents. lncRNA TUG1 was overexpressed in osteosarcoma tissues and is associated with poor prognosis of patients. TGF-β secreted by CAFs motivated the expression of TUG1 in osteosarcoma cells. Bioinformatics analysis showed that miR-143-5p was a potential target binding miRNA of TUG1, and HIF-1α was the target gene of miR-143-5p. Further experiments verified that TUG1 positively regulated HIF-1α through acting ceRNA of miR-143-5p, thereby promoting the invasion and angiogenesis of osteosarcoma cells. The xenografted tumor experiments in nude mice showed that silencing TUG1 refrained tumor growth, peritoneal diffusion, and metastasis in vivo. In short, TUG1 might be a potential therapeutic target and prognostic indicator in osteosarcoma.65
Cancer stem cells (CSCs)
CSCs can secrete plenty of VEGF to promote tumor angiogenesis. In the meantime, CSCs depend on vascular niches. CSCs and angiogenesis can form a positive feedback loop to boost the occurrence and development of tumors.97 CD90 + HCC cells are described as cancer stem-cell-like that exhibit aggressive and metastatic phenotypes. CD90 + cells isolated from Huh7 cell line were found to have mesenchymal phenotype and release exosomes in large quantities in HCC. Exosomes produced by CD90 + Huh7 cells promoted angiogenesis and cell-cell adhesion of HUVECs. Mechanism studies have shown that CD90+ Huh7 cells can overexpress H19 and transfer H19 into HUVECs by secreting exosomes, thus promoting angiogenesis and adhesion of endothelial monolayer in HUVECs.75 Due to the difficulty in successfully isolating and identifying CSCs, studies on CSCs are still in infancy. There are few studies on lncRNAs, angiogenesis, and CSCs. Scientific and technological progress will solve the problem of isolation and identification of CSCs in the future. The studies of lncRNAs, angiogenesis, and CSCs will also embrace a qualitative leap by then.
Correlation between lncRNAs and anti-angiogenic therapy resistance in cancer
Anti-angiogenic therapy, as an important means of tumor therapy, has achieved good efficacy in clinical application. However, the shortcoming of drug resistance has also been highlighted in the long-term clinical application. Many studies have attested that lncRNAs are closely related to anti-angiogenic therapy resistance (Table 2).
Table 2.
Correlation between lncRNAs and anti-angiogenic therapy resistance in tumors
Classification | Drugs | Cancer | LncRNA | Role | Mechanism | Ref |
---|---|---|---|---|---|---|
CeRNA | sorafenib | hepatocellular carcinoma | MALAT1 | up | regulating miR-140-5p/Aurora-A axis | 98 |
TTN-AS1 | up | regulating miR-16-5p/cyclin E1/PTEN/AKT axis | 99 | |||
sunitinib | renal cancer | HOTAIR | up | regulating miR-17-5p/Beclin1 axis | 100 | |
Gene transcription | sunitinib | renal cancer | SNHG12 | up | regulating CDCA3 by stabilizing transcription factor SP1 | 101 |
CCAT1 | up | regulating c-Myc | 102 | |||
Signal pathway | anlotinib | non-small cell lung cancer | NEAT1 | up | regulating Wnt/β-catenin signaling pathway | 103 |
Sorafenib
Sorafenib is a multi-kinase inhibitor targeted VEGFR and platelet-derived growth factor receptor (PDGFR) and others.104 As a first-line treatment for advanced HCC, it has effectively ameliorated the prognosis of patients with HCC.105 However, patients gradually develop resistance, leading to poor treatment.106 Fan et al. established HCC cell lines resistant to sorafenib (HepG2-R and SMMC-R). Microarray analysis results displayed that there were 293 upregulated lncRNAs and 207 downregulated lncRNAs in sorafenib-resistant HCC cells, among which MALAT1 was significantly overexpressed. The knockdown of MALAT1 in HepG2-R and SMMC-R cells resulted in the decrease of IC50 value, inhibition of cell proliferation and migration, G2–M cell-cycle arrest, and increase of apoptosis. Aurora-A not only promotes the genesis and progression of tumors, but it also mediates the resistance of chemotherapy and radiotherapy and participates in immunotherapy.107,108 In clinical trials, its selective inhibitors or siRNA have been demonstrated to have anti-tumor effects.109,110 Mechanically, MALAT1 upregulated the expression of Aurora-A by sponging miR-140-5p to intensify sorafenib resistance of HCC cells. Clinical patient tissue tests further confirmed these results. Patients with high MALAT1 expression had poor prognosis. More importantly, animal studies indicated that silencing MALAT1 promoted the anti-tumor efficacy of sorafenib in vivo (Figure 2G).98 In HCC, lncRNA TTN-AS1 was a potential therapeutic target of HCC. It is common knowledge that cyclin E1 is the tumorigenic factor and biomarker of numerous malignant tumors, such as, breast cancer,111 osteosarcoma,112 etc. Research has demonstrated that lncRNA TTN-AS1 can competitively inhibit miR-16-5p to upregulate cyclin E1 and regulate the PTEN/AKT signaling pathway to enhance sorafenib resistance of HCC. In a tumor-forming model in nude mice, knockdown of TTN-AS1 reduced sorafenib resistance in vivo.99
Sunitinib
Sunitinib, a multitarget RTK inhibitor with strong anti-angiogenesis, has been approved for imatinib-resistant stromal tumor, renal, and pancreatic neuroendocrine tumors. Sunitinib has been proved to extend PFS and OS of patients with RC, but its efficacy has been overshadowed by drug resistance after the sensitive phase.113 Li et al. recognized a novel lncRNA, HOTAIR, which was an enhancing factor of sunitinib resistance in RC. HOTAIR promoted sunitinib resistance in RC by negatively regulating miR-17-5p to promote Beclin1-mediated autophagy. Knockdown of HOTAIR partially reversed sunitinib resistance of RC cells in vivo. Thus, miR-17-5p/Beclin1 axis might be the potential mechanism of HOTAIR promoting sunitinib resistance of RC.100 In renal cell carcinoma (RCC), lncRNA CCAT1 granted resistance to sunitinib in a c-Myc-dependent manner, providing a new target for improving sunitinib therapy (Figure 2H).102 In addition, Liu et al. first proposed lncRNA SNHG12 could upregulate the expression of CDCA3 by stabilizing transcription factor SP1 to facilitate tumor progressions and resistance to sunitinib in RCC. RNA sequence and bioinformatics analysis demonstrated that SNHG12 was highly expressed in RCC tissues and cells resistant to sunitinib, associating with poor prognoses. Downregulation of SNHG12 meaningly reduced the proliferation, migration, and invasion ability of tumor cells, and partially reversed the resistance to sunitinib. CDCA3, a potential marker for poor prognoses, often acts as an oncogene in digestive system tumors and NSCLC.114, 115, 116 GSEA analysis and rescue assays confirmed that SNHG12 accelerated the progression of RCC by positively regulating CDCA3. RIP and ChIP experiments confirmed that SNHG12 activated CDCA3 transcription by binding and stabilizing SP1. Animal experiments made clear that knockdown SNHG12 inhibited tumor progression and reversed sunitinib resistance. Thus, SNHG12 may be a potential therapeutic target for the reversal of sunitinib resistance in RCC (Figure 2I).101
Anlotinib
Anlotinib is a multi-target inhibitor that targets c-kit, PDGFR, fibroblast growth factor receptor, and VEGFR. In 2018, it was approved for patients with NSCLC who progressed after treatment of at least two drugs in China117 It has been shown to prolong PFS and OS in patients with advanced refractory NSCLC.118 Previous studies reported that lncRNA NEAT1 played an oncogene role in NSCLC. It promoted the progression of NSCLC by regulating the miR-376b-3p/SULF1 and miR-204/NUAK1 axis, and it even mediated the paclitaxel resistance of NSCLC by activating the AKT/mTOR signaling pathway.119, 120, 121 Gu et al. reported that NEAT1 was involved in regulating anlotinib sensitivity in NSCLC. In vivo, the knockdown of NEAT1 in A549 and NCI-H1975 cells enhanced the sensitivity of the cells to anlotinib. Compared with anlotinib alone group, tumor volume and weight were significantly reduced in the NEAT1 knockdown combined anlotinib group in vivo. Further studies showed that NEAT1 knockdown promoted the cytotoxicity of anlotinib by inhibiting the Wnt/β-catenin signaling pathway. Downregulation of NEAT1 can enhance the anti-tumor effect of antitinib in mouse xenograft tumor model (Figure 2J).103
Conclusions and perspectives
Cancer poses a serious threat to the global economy and human health. The functions of ncRNAs have been discovered through high-throughput sequence technology. We mainly reviewed lncRNAs that can play ceRNAs of miRNAs, target directly proangiogenic factors and their receptors, or act on some signaling pathways to regulate tumor angiogenesis. The same lncRNA regulates angiogenesis by different mechanisms in different tumors. In addition, we also reviewed most of the lncRNAs regulate the mechanism of anti-angiogenic therapy resistance through sponging miRNA. Ye et al. discovered anisomycin inhibited angiogenesis in ovarian cancer by weakening the molecular sponge effect of lncRNA MEG3/miR-421/PDGFRA axis.122 Tretinoin can target and inhibit the TR4-mediated expression of lncRNA TASR/AXL axis, thereby reducing sunitinib resistance in RCC.123 These results further confirmed the feasibility of lncRNAs as potential targets for tumor therapy. Wang et al. reported ASRPS secreted by LINC00908 had anti-tumor angiogenesis effects in triple-negative breast cancer. Mechanically, ASRPS downregulated STAT3 phosphorylation by directly binding to STAT3 to decrease VEGF.124 Tang et al. prepared an artificial lncRNA (alncRNA) that could target multiple miRNAs to regulate the PTEN/AKT axis. Ad5-alncRNA enhanced the inhibitory effect of sorafenib on HCC cells.125 These investigations suggested that lncRNAs might even have potential as a therapeutic agent for cancer. In conclusion, lncRNAs, which have great potential for both tumor angiogenesis and anti-angiogenic therapy resistance, may become potential targets or even new medicines for tumor in the future. Antisense oligonucleotide technology and overexpression vector construction technology make it possible to treat diseases by regulating the expression of lncRNAs. Compared with traditional chemotherapy, lncRNA targeted therapy has the following advantages: (1) highly selectivity, (2) high specificity, (3) high affinity, (4) high degree of individualization, and (5) fewer side effects. Liquid biopsy has been gradually used to detect tumor biomarkers.126 Exosomes can prevent lncRNAs from degradation by nuclease to increase their stability. Researchers have demonstrated that circulating and exosomal lncRNAs are biomarkers for diagnosis and prognosis of malignant tumors.127, 128, 129, 130, 131Therefore, lncRNAs have great potential for clinical application of tumor diagnosis and treatment. At present, lncRNAs have not been fully identified and mechanisms have not been thoroughly studied. Large amounts of basic experiments and clinical trials will be needed to further explore and verify their clinical application value.
Acknowledgments
This work was supported by grants from the National Natural Science Foundation of China (no. 82072591), the Key Research and Development Plan (Social Development) of the Science and Technology Department of Jiangsu Province (no. BE2019760), and the “123” advantageous disciplines and core technologies of the Second Affiliated Hospital of Nanjing Medical University.
Author contributions
J.L., Q.Z., D.Y., and F.X. wrote and drafted the manuscript and figures. Z.W. designed the manuscript. Z.W. and J.L. revised the manuscript. All authors contributed to the article and approved the submitted version.
Declaration of interests
The authors declare no competing interests.
References
- 1.Siegel R.L., Miller K.D., Fuchs H.E. Cancer statistics, 2021. CA Cancer J. Clin. 2021;71:7–33. doi: 10.3322/caac.21654. [DOI] [PubMed] [Google Scholar]
- 2.Sung H., Ferlay J., Siegel R.L., Laversanne M., Soerjomataram I., Jemal A. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021;71:209–249. doi: 10.3322/caac.21660. [DOI] [PubMed] [Google Scholar]
- 3.Lugano R., Ramachandran M., Dimberg A. Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell Mol. Life Sci. 2020;77:1745–1770. doi: 10.1007/s00018-019-03351-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Weidner N., Semple J.P., Welch W.R., Folkman J. Tumor angiogenesis and metastasis--correlation in invasive breast carcinoma. N. Engl. J. Med. 1991;324:1–8. doi: 10.1056/NEJM199101033240101. [DOI] [PubMed] [Google Scholar]
- 5.Fox S.B. Tumour angiogenesis and prognosis. Histopathology. 1997;30:294–301. doi: 10.1046/j.1365-2559.1997.d01-606.x. [DOI] [PubMed] [Google Scholar]
- 6.Chekerov R., Hilpert F., Mahner S., El-Balat A., Harter P., De Gregorio N., Fridrich C., Markmann S., Potenberg J., Lorenz R., et al. Sorafenib plus topotecan versus placebo plus topotecan for platinum-resistant ovarian cancer (TRIAS): a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 2018;19:1247–1258. doi: 10.1016/S1470-2045(18)30372-3. [DOI] [PubMed] [Google Scholar]
- 7.Rani V., Prabhu A. Combining angiogenesis inhibitors with radiation: advances and challenges in cancer treatment. Curr. Pharm. Des. 2021;27:919–931. doi: 10.2174/1381612826666201002145454. [DOI] [PubMed] [Google Scholar]
- 8.Manegold C., Dingemans A.C., Gray J.E., Nakagawa K., Nicolson M., Peters S., et al. The potential of combined immunotherapy and antiangiogenesis for the synergistic treatment of advanced NSCLC. J. Thorac. Oncol. 2017;12:194–207. doi: 10.1016/j.jtho.2016.10.003. [DOI] [PubMed] [Google Scholar]
- 9.Bottsford-Miller J.N., Coleman R.L., Sood A.K. Resistance and escape from antiangiogenesis therapy: clinical implications and future strategies. J. Clin. Oncol. 2012;30:4026–4034. doi: 10.1200/JCO.2012.41.9242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Djebali S., Davis C.A., Merkel A., Dobin A., Lassmann T., Mortazavi A., Tanzer A., Lagarde J., Lin W., Schlesinger F., et al. Landscape of transcription in human cells. Nature. 2012;489:101–108. doi: 10.1038/nature11233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Mattick J.S., Makunin I.V. Non-coding RNA. Hum. Mol. Genet. 15 Spec. No. 2006;1:R17–R29. doi: 10.1093/hmg/ddl046. [DOI] [PubMed] [Google Scholar]
- 12.Yang X., Meng L., Zhong Y., Hu F., Wang L., Wang M. The long intergenic noncoding RNA GAS5 reduces cisplatin-resistance in non-small cell lung cancer through the miR-217/LHPP axis. Aging (Albany NY) 2021;13:2864–2884. doi: 10.18632/aging.202352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Jiang X., Guan J., Xu Y., Ren H., Jiang J., Wudu M., Wang Q., Su H., Zhang Y., Zhang B., et al. Silencing of CASC8 inhibits non-small cell lung cancer cells function and promotes sensitivity to osimertinib via FOXM1. J. Cancer. 2021;12:387–396. doi: 10.7150/jca.47863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wang H., Lu B., Ren S., Wu F., Wang X., Yan C., Wang Z. Long noncoding RNA LINC01116 contributes to gefitinib resistance in non-small cell lung cancer through regulating IFI44. Mol. Ther. Nucleic Acids. 2020;19:218–227. doi: 10.1016/j.omtn.2019.10.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Huang D., Chen J., Yang L., Ouyang Q., Li J., Lao L., Zhao J., Liu J., Lu Y., Xing Y., et al. NKILA lncRNA promotes tumor immune evasion by sensitizing T cells to activation-induced cell death. Nat. Immunol. 2018;19:1112–1125. doi: 10.1038/s41590-018-0207-y. [DOI] [PubMed] [Google Scholar]
- 16.Yu Z., Wang G., Zhang C., Liu Y., Chen W., Wang H., Liu H. LncRNA SBF2-AS1 affects the radiosensitivity of non-small cell lung cancer via modulating microRNA-302a/MBNL3 axis. Cell Cycle. 2020;19:300–316. doi: 10.1080/15384101.2019.1708016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Chen X., Chen Z., Yu S., Nie F., Yan S., Ma P., Chen Q., Wei C., Fu H., Xu T., et al. Long noncoding RNA LINC01234 functions as a competing endogenous RNA to regulate CBFB expression by sponging miR-204-5p in gastric cancer. Clin. Cancer Res. 2018;24:2002–2014. doi: 10.1158/1078-0432.CCR-17-2376. [DOI] [PubMed] [Google Scholar]
- 18.Chen Z., Chen X., Lei T., Gu Y., Gu J., Huang J., Lu B., Yuan L., Sun M., Wang Z. Integrative analysis of NSCLC identifies LINC01234 as an oncogenic lncRNA that interacts with HNRNPA2B1 and regulates miR-106b biogenesis. Mol. Ther. 2020;28:1479–1493. doi: 10.1016/j.ymthe.2020.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chen Z., Chen X., Lu B., Gu Y., Chen Q., Lei T., Nie F., Gu J., Huang J., Wei C., et al. Up-regulated LINC01234 promotes non-small-cell lung cancer cell metastasis by activating VAV3 and repressing BTG2 expression. J. Hematol. Oncol. 2020;13:7. doi: 10.1186/s13045-019-0842-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Chen Z., Chen Q., Cheng Z., Gu J., Feng W., Lei T., Huang J., Pu J., Chen X., Wang Z. Long non-coding RNA CASC9 promotes gefitinib resistance in NSCLC by epigenetic repression of DUSP1. Cell Death Dis. 2020;11:858. doi: 10.1038/s41419-020-03047-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Teppan J., Barth D.A., Prinz F., Jonas K., Pichler M., Klec C. Involvement of long non-coding RNAs (lncRNAs) in tumor angiogenesis. Noncoding RNA. 2020;6:42. doi: 10.3390/ncrna6040042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Qu L., Ding J., Chen C., Wu Z.J., Liu B., Gao Y., Chen W., Liu F., Sun W., Li X.F., et al. Exosome-transmitted lncARSR promotes sunitinib resistance in renal cancer by acting as a competing endogenous RNA. Cancer Cell. 2016;29:653–668. doi: 10.1016/j.ccell.2016.03.004. [DOI] [PubMed] [Google Scholar]
- 23.Guo C.J., Ma X.K., Xing Y.H., Zheng C.C., Xu Y.F., Shan L., Zhang J., Wang S., Wang Y., Carmichael G.G., et al. Distinct processing of lncRNAs contributes to non-conserved functions in stem cells. Cell. 2020;181:621–636.e622. doi: 10.1016/j.cell.2020.03.006. [DOI] [PubMed] [Google Scholar]
- 24.Anastasiadou E., Jacob L.S., Slack F.J. Non-coding RNA networks in cancer. Nat. Rev. Cancer. 2018;18:5–18. doi: 10.1038/nrc.2017.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Beermann J., Piccoli M.T., Viereck J., Thum T. Non-coding RNAs in development and disease: background, mechanisms, and therapeutic approaches. Physiol. Rev. 2016;96:1297–1325. doi: 10.1152/physrev.00041.2015. [DOI] [PubMed] [Google Scholar]
- 26.Slack F.J., Chinnaiyan A.M. The role of non-coding RNAs in oncology. Cell. 2019;179:1033–1055. doi: 10.1016/j.cell.2019.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zhang X., Wang W., Zhu W., Dong J., Cheng Y., Yin Z., Shen F. Mechanisms and functions of long non-coding RNAs at multiple regulatory levels. Int. J. Mol. Sci. 2019;20:5573. doi: 10.3390/ijms20225573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kopp F., Mendell J.T. Functional classification and experimental dissection of long noncoding RNAs. Cell. 2018;172:393–407. doi: 10.1016/j.cell.2018.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Statello L., Guo C.J., Chen L.L., Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nat. Rev. Mol. Cell Biol. 2021;22:96–118. doi: 10.1038/s41580-020-00315-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Li G., Yun X., Ye K., Zhao H., An J., Zhang X., Han X., Li Y., Wang S. Long non-coding RNA-H19 stimulates osteogenic differentiation of bone marrow mesenchymal stem cells via the microRNA-149/SDF-1 axis. J. Cell Mol Med. 2020;24:4944–4955. doi: 10.1111/jcmm.15040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Sanchez Calle A., Kawamura Y., Yamamoto Y., Takeshita F., Ochiya T. Emerging roles of long non-coding RNA in cancer. Cancer Sci. 2018;109:2093–2100. doi: 10.1111/cas.13642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Feng L., Li J., Li F., Li H., Bei S., Zhang X., Yang Z. Long noncoding RNA VCAN-AS1 contributes to the progression of gastric cancer via regulating p53 expression. J. Cell Physiol. 2020;235:4388–4398. doi: 10.1002/jcp.29315. [DOI] [PubMed] [Google Scholar]
- 33.Liu Y., Yin L., Chen C., Zhang X., Wang S. Long non-coding RNA GAS5 inhibits migration and invasion in gastric cancer via interacting with p53 protein. Dig. Liver Dis. 2020;52:331–338. doi: 10.1016/j.dld.2019.08.012. [DOI] [PubMed] [Google Scholar]
- 34.He Z., Duan Z., Chen L., Li B., Zhou Y. Long non-coding RNA Loc490 inhibits gastric cancer cell proliferation and metastasis by upregulating RNA-binding protein Quaking. Aging (Albany NY) 2020;12:17681–17693. doi: 10.18632/aging.103876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Li J., Wang X., Wang Y., Yang Q. H19 promotes the gastric carcinogenesis by sponging miR-29a-3p: evidence from lncRNA-miRNA-mRNA network analysis. Epigenomics. 2020;12:989–1002. doi: 10.2217/epi-2020-0114. [DOI] [PubMed] [Google Scholar]
- 36.Li Y., Ma H.Y., Hu X.W., Qu Y.Y., Wen X., Zhang Y., Xu Q.Y. LncRNA H19 promotes triple-negative breast cancer cells invasion and metastasis through the p53/TNFAIP8 pathway. Cancer Cell Int. 2020;20:200. doi: 10.1186/s12935-020-01261-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zhang Y., Huang W., Yuan Y., Li J., Wu J., Yu J., He Y., Wei Z., Zhang C. Long non-coding RNA H19 promotes colorectal cancer metastasis via binding to hnRNPA2B1. J. Exp. Clin. Cancer Res. 2020;39:141. doi: 10.1186/s13046-020-01619-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Yu A., Zhao L., Kang Q., Li J., Chen K., Fu H. Transcription factor HIF1alpha promotes proliferation, migration, and invasion of cholangiocarcinoma via long noncoding RNA H19/microRNA-612/Bcl-2 axis. Transl. Res. 2020;224:26–39. doi: 10.1016/j.trsl.2020.05.010. [DOI] [PubMed] [Google Scholar]
- 39.Zheng Z.H., Wu D.M., Fan S.H., Zhang Z.F., Chen G.Q., Lu J. Upregulation of miR-675-5p induced by lncRNA H19 was associated with tumor progression and development by targeting tumor suppressor p53 in non-small cell lung cancer. J. Cell Biochem. 2019;120:18724–18735. doi: 10.1002/jcb.29182. [DOI] [PubMed] [Google Scholar]
- 40.Lan X., Sun W., Dong W., Wang Z., Zhang T., He L., Zhang H. Downregulation of long noncoding RNA H19 contributes to the proliferation and migration of papillary thyroid carcinoma. Gene. 2018;646:98–105. doi: 10.1016/j.gene.2017.12.051. [DOI] [PubMed] [Google Scholar]
- 41.Folkman J. Tumor angiogenesis: therapeutic implications. N. Engl. J. Med. 1971;285:1182–1186. doi: 10.1056/NEJM197111182852108. [DOI] [PubMed] [Google Scholar]
- 42.Hinshaw D.C., Shevde L.A. The tumor microenvironment innately modulates cancer progression. Cancer Res. 2019;79:4557–4566. doi: 10.1158/0008-5472.CAN-18-3962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Vitale I., Manic G., Coussens L.M., Kroemer G., Galluzzi L. Macrophages and metabolism in the tumor microenvironment. Cell Metab. 2019;30:36–50. doi: 10.1016/j.cmet.2019.06.001. [DOI] [PubMed] [Google Scholar]
- 44.Shweiki D., Itin A., Soffer D., Keshet E. Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis. Nature. 1992;359:843–845. doi: 10.1038/359843a0. [DOI] [PubMed] [Google Scholar]
- 45.Rahma O.E., Hodi F.S. The intersection between tumor angiogenesis and immune suppression. Clin. Cancer Res. 2019;25:5449–5457. doi: 10.1158/1078-0432.CCR-18-1543. [DOI] [PubMed] [Google Scholar]
- 46.Kubota S., Tanaka M., Endo H., Ito Y., Onuma K., Ueda Y., Kamiura S., Yoshino K., Kimura T., Kondo J., et al. Dedifferentiation of neuroendocrine carcinoma of the uterine cervix in hypoxia. Biochem. Biophys. Res. Commun. 2020;524:398–404. doi: 10.1016/j.bbrc.2020.01.024. [DOI] [PubMed] [Google Scholar]
- 47.Yi M., Jiao D., Qin S., Chu Q., Wu K., Li A. Synergistic effect of immune checkpoint blockade and anti-angiogenesis in cancer treatment. Mol. Cancer. 2019;18:60. doi: 10.1186/s12943-019-0974-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Huang Y., Kim B.Y.S., Chan C.K., Hahn S.M., Weissman I.L., Jiang W. Improving immune-vascular crosstalk for cancer immunotherapy. Nat. Rev. Immunol. 2018;18:195–203. doi: 10.1038/nri.2017.145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Fukumura D., Kloepper J., Amoozgar Z., Duda D.G., Jain R.K. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat. Rev. Clin. Oncol. 2018;15:325–340. doi: 10.1038/nrclinonc.2018.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Muhsin M., Graham J., Kirkpatrick P. Bevacizumab. Nat. Rev. Drug Discov. 2004;3:995–996. doi: 10.1038/nrd1601. [DOI] [PubMed] [Google Scholar]
- 51.Socinski M.A., Jotte R.M., Cappuzzo F., Orlandi F., Stroyakovskiy D., Nogami N., Rodriguez-Abreu D., Moro-Sibilot D., Thomas C.A., Barlesi F., et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N. Engl. J. Med. 2018;378:2288–2301. doi: 10.1056/NEJMoa1716948. [DOI] [PubMed] [Google Scholar]
- 52.Choueiri T.K., Motzer R.J., Rini B.I., Haanen J., Campbell M.T., Venugopal B., Kollmannsberger C., Gravis-Mescam G., Uemura M., Lee J.L., et al. Updated efficacy results from the JAVELIN Renal 101 trial: first-line avelumab plus axitinib versus sunitinib in patients with advanced renal cell carcinoma. Ann. Oncol. 2020;31:1030–1039. doi: 10.1016/j.annonc.2020.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Iwamoto H., Abe M., Yang Y., Cui D., Seki T., Nakamura M., Hosaka K., Lim S., Wu J., He X., et al. Cancer lipid metabolism confers antiangiogenic drug resistance. Cell Metab. 2018;28:104–117.e105. doi: 10.1016/j.cmet.2018.05.005. [DOI] [PubMed] [Google Scholar]
- 54.Kuczynski E.A., Reynolds A.R. Vessel co-option and resistance to anti-angiogenic therapy. Angiogenesis. 2020;23:55–74. doi: 10.1007/s10456-019-09698-6. [DOI] [PubMed] [Google Scholar]
- 55.Si X., Zheng H., Wei G., Li M., Li W., Wang H., Guo H., Sun J., Li C., Zhong S., et al. circRNA Hipk3 induces cardiac regeneration after myocardial infarction in mice by binding to Notch1 and miR-133a. Mol. Ther. Nucleic Acids. 2020;21:636–655. doi: 10.1016/j.omtn.2020.06.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Li Y., Yan C., Fan J., Hou Z., Han Y. MiR-221-3p targets Hif-1alpha to inhibit angiogenesis in heart failure. Lab. Invest. 2021;101:104–115. doi: 10.1038/s41374-020-0450-3. [DOI] [PubMed] [Google Scholar]
- 57.Zou J., Liu K.C., Wang W.P., Xu Y. Circular RNA COL1A2 promotes angiogenesis via regulating miR-29b/VEGF axis in diabetic retinopathy. Life Sci. 2020;256:117888. doi: 10.1016/j.lfs.2020.117888. [DOI] [PubMed] [Google Scholar]
- 58.Cheng N., Li X., Zhao C., Ren S., Chen X., Cai W., Zhao M., Zhang Y., Li J., Wang Q., et al. Microarray expression profile of long non-coding RNAs in EGFR-TKIs resistance of human non-small cell lung cancer. Oncol. Rep. 2015;33:833–839. doi: 10.3892/or.2014.3643. [DOI] [PubMed] [Google Scholar]
- 59.Caporarello N., Lupo G., Olivieri M., Cristaldi M., Cambria M.T., Salmeri M., Anfuso C.D. Classical VEGF, Notch and Ang signalling in cancer angiogenesis, alternative approaches and future directions (Review) Mol. Med. Rep. 2017;16:4393–4402. doi: 10.3892/mmr.2017.7179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Chen J., Liu A., Wang Z., Wang B., Chai X., Lu W., et al. LINC00173.v1 promotes angiogenesis and progression of lung squamous cell carcinoma by sponging miR-511-5p to regulate VEGFA expression. Mol Cancer. 2020;19:98. doi: 10.1186/s12943-020-01217-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Teng F., Zhang J.X., Chang Q.M., Wu X.B., Tang W.G., Wang J.F., et al. Correction to: LncRNA MYLK-AS1 facilitates tumor progression and angiogenesis by targeting miR-424-5p/E2F7 axis and activating VEGFR-2 signaling pathway in hepatocellular carcinoma. J Exp Clin Cancer Res. 2020;39 doi: 10.1186/s13046-020-01780-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hou Z.H., Xu X.W., Fu X.Y., Zhou L.D., Liu S.P., Tan D.M. Long non-coding RNA MALAT1 promotes angiogenesis and immunosuppressive properties of HCC cells by sponging miR-140. Am J Physiol Cell Physiol. 2020;318:C649–C663. doi: 10.1152/ajpcell.00510.2018. [DOI] [PubMed] [Google Scholar]
- 63.Liu Y., Li Q., Tang D., Li M., Zhao P., Yang W., et al. SNHG17 promotes the proliferation and migration of colorectal adenocarcinoma cells by modulating CXCL12-mediated angiogenesis. Cancer Cell International. 2020;20 doi: 10.1186/s12935-020-01621-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Cui X., Zhang H., Chen T., Yu W., Shen K. Long Noncoding RNA SNHG22 Induces Cell Migration, Invasion, and Angiogenesis of Gastric Cancer Cells via microRNA-361-3p/HMGA1/Wnt/β-Catenin Axis. Cancer Management and Research. 2020;12:12867–12883. doi: 10.2147/CMAR.S281578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Yu X., Hu L., Li S., Shen J., Wang D., Xu R., et al. Long non-coding RNA Taurine upregulated gene 1 promotes osteosarcoma cell metastasis by mediating HIF-1alpha via miR-143-5p. Cell Death Dis. 2019;10 doi: 10.1038/s41419-019-1509-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Chen S., Lv L., Zhan Z., Wang X., You Z., Luo X., et al. Silencing of long noncoding RNA SRRM2-AS exerts suppressive effects on angiogenesis in nasopharyngeal carcinoma via activating MYLK-mediated cGMP-PKG signaling pathway. J Cell Physiol. 2020;235:7757–7768. doi: 10.1002/jcp.29382. [DOI] [PubMed] [Google Scholar]
- 67.Tang L., Wen J.B., Wen P., Li X., Gong M., Li Q. Long non-coding RNA LINC01314 represses cell migration, invasion, and angiogenesis in gastric cancer via the Wnt/beta-catenin signaling pathway by down-regulating KLK4. Cancer Cell Int. 2019;19:94. doi: 10.1186/s12935-019-0799-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hou Y., Jia H., Cao Y., Zhang S., Zhang X., Wei P., et al. LncRNA EPIC1 promotes tumor angiogenesis via activating the Ang2/Tie2 axis in non-small cell lung cancer. Life Sci. 2021;267:118933. doi: 10.1016/j.lfs.2020.118933. [DOI] [PubMed] [Google Scholar]
- 69.Niu Y., Bao L., Chen Y., Wang C., Luo M., Zhang B., et al. HIF2-Induced Long Noncoding RNA RAB11B-AS1 Promotes Hypoxia-Mediated Angiogenesis and Breast Cancer Metastasis. Cancer Res. 2020;80:964–975. doi: 10.1158/0008-5472.CAN-19-1532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Lei L., Mou Q. Exosomal taurine up-regulated 1 promotes angiogenesis and endothelial cell proliferation in cervical cancer. Cancer Biol Ther. 2020;21:717–725. doi: 10.1080/15384047.2020.1764318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Li Y., Li H., Wei X. Long noncoding RNA LINC00261 suppresses prostate cancer tumorigenesis through upregulation of GATA6-mediated DKK3. Cancer Cell Int. 2020;20 doi: 10.1186/s12935-020-01484-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Li H., Yan R., Chen W., Ding X., Liu J., Chen G., et al. Long non coding RNA SLC26A4-AS1 exerts antiangiogenic effects in human glioma by upregulating NPTX1 via NFKB1 transcriptional factor. FEBS J. 2021;288:212–228. doi: 10.1111/febs.15325. [DOI] [PubMed] [Google Scholar]
- 73.Chang L., Bian Z., Xiong X., Liu J., Wang D., Zhou F., et al. Long Non-coding RNA LINC00320 Inhibits Tumorigenicity of Glioma Cells and Angiogenesis Through Downregulation of NFKB1-Mediated AQP9. Front Cell Neurosci. 2020;14 doi: 10.3389/fncel.2020.542552. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 74.Huang J.K., Ma L., Song W.H., Lu B.Y., Huang Y.B., Dong H.M., et al. LncRNA-MALAT1 Promotes Angiogenesis of Thyroid Cancer by Modulating Tumor-Associated Macrophage FGF2 Protein Secretion. J Cell Biochem. 2017;118:4821–4830. doi: 10.1002/jcb.26153. [DOI] [PubMed] [Google Scholar]
- 75.Conigliaro A., Costa V., Lo Dico A., Saieva L., Buccheri S., Dieli F., et al. CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA. Mol Cancer. 2015;14 doi: 10.1186/s12943-015-0426-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Guan S., Wei J., Huang L., Wu L. Chemotherapy and chemo-resistance in nasopharyngeal carcinoma. Eur J Med Chem. 2020;207:112758. doi: 10.1016/j.ejmech.2020.112758. [DOI] [PubMed] [Google Scholar]
- 77.Eguchi R., Wakabayashi I. HDGF enhances VEGFdependent angiogenesis and FGF2 is a VEGFindependent angiogenic factor in nonsmall cell lung cancer. Oncol Rep. 2020;44:14–28. doi: 10.3892/or.2020.7580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Niu M., He Y., Xu J., Ding L., He T., Yi Y., et al. Noncanonical TGF-beta signaling leads to FBXO3-mediated degradation of DeltaNp63alpha promoting breast cancer metastasis and poor clinical prognosis. PLoS Biol. 2021;19:e3001113. doi: 10.1371/journal.pbio.3001113. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 79.Hao T., Gan Y.H. ΔNp63α promotes the expression and nuclear translocation of PTEN, leading to cisplatin resistance in oral cancer cells. Am J Transl Res. 2020;12:6187–6203. [PMC free article] [PubMed] [Google Scholar]
- 80.Zhou J.Y., Huang D.G., Zhu M., Gao C.Q., Yan H.C., Li X.G., et al. Wnt/beta-catenin-mediated heat exposure inhibits intestinal epithelial cell proliferation and stem cell expansion through endoplasmic reticulum stress. J Cell Physiol. 2020;235:5613–5627. doi: 10.1002/jcp.29492. [DOI] [PubMed] [Google Scholar]
- 81.Wei H., Beeson G.C., Ye Z., Zhang J., Yao H., Damon B., et al. Activation of Wnt/beta-catenin signalling and HIF1alpha stabilisation alters pluripotency and differentiation/proliferation properties of human-induced pluripotent stem cells. Biol Cell. 2021;113:133–145. doi: 10.1111/boc.202000055. [DOI] [PubMed] [Google Scholar]
- 82.Lorzadeh S., Kohan L., Ghavami S., Azarpira N. Autophagy and the Wnt signaling pathway: A focus on Wnt/beta-catenin signaling. Biochim Biophys Acta Mol Cell Res. 2021;1868:118926. doi: 10.1016/j.bbamcr.2020.118926. [DOI] [PubMed] [Google Scholar]
- 83.Serafino A., Giovannini D., Rossi S., Cozzolino M. Targeting the Wnt/beta-catenin pathway in neurodegenerative diseases: recent approaches and current challenges. Expert Opin Drug Discov. 2020;15:803–822. doi: 10.1080/17460441.2020.1746266. [DOI] [PubMed] [Google Scholar]
- 84.Schunk S.J., Floege J., Fliser D., Speer T. WNT-beta-catenin signalling - a versatile player in kidney injury and repair. Nat Rev Nephrol. 2021;17:172–184. doi: 10.1038/s41581-020-00343-w. [DOI] [PubMed] [Google Scholar]
- 85.Li K., Zhang J., Tian Y., He Y., Xu X., Pan W., et al. The Wnt/beta-catenin/VASP positive feedback loop drives cell proliferation and migration in breast cancer. Oncogene. 2020;39:2258–2274. doi: 10.1038/s41388-019-1145-3. [DOI] [PubMed] [Google Scholar]
- 86.Li Y., Liu J., Hu W., Zhang Y., Sang J., Li H., et al. miR-424-5p Promotes Proliferation, Migration and Invasion of Laryngeal Squamous Cell Carcinoma. Onco Targets Ther. 2019;12:10441–10453. doi: 10.2147/OTT.S224325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Cui Y., Yang J., Bai Y., Zhang Y., Yao Y., Zheng T., et al. miR-424-5p regulates cell proliferation and migration of esophageal squamous cell carcinoma by targeting SIRT4. J Cancer. 2020;11:6337–6347. doi: 10.7150/jca.50587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Dai W., Zhou J., Wang H., Zhang M., Yang X., Song W. miR-424-5p promotes the proliferation and metastasis of colorectal cancer by directly targeting SCN4B. Pathol Res Pract. 2020;216 doi: 10.1016/j.prp.2019.152731. [DOI] [PubMed] [Google Scholar]
- 89.Liu X., Fu Y., Zhang G., Zhang D., Liang N., Li F., et al. miR-424-5p Promotes Anoikis Resistance and Lung Metastasis by Inactivating Hippo Signaling in Thyroid Cancer. Mol Ther Oncolytics. 2019;15:248–260. doi: 10.1016/j.omto.2019.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Wu J., Yang B., Zhang Y., Feng X., He B., Xie H., et al. miR-424-5p represses the metastasis and invasion of intrahepatic cholangiocarcinoma by targeting ARK5. Int J Biol Sci. 2019;15:1591–1599. doi: 10.7150/ijbs.34113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Dastmalchi N., Hosseinpourfeizi M.A., Khojasteh S.M.B., Baradaran B., Safaralizadeh R. Tumor suppressive activity of miR-424-5p in breast cancer cells through targeting PD-L1 and modulating PTEN/PI3K/AKT/mTOR signaling pathway. Life Sciences. 2020;259 doi: 10.1016/j.lfs.2020.118239. [DOI] [PubMed] [Google Scholar]
- 92.Zhao C., Zhao F., Chen H., Liu Y., Su J. MicroRNA-424-5p inhibits the proliferation, migration, and invasion of nasopharyngeal carcinoma cells by decreasing AKT3 expression. Braz J Med Biol Res. 2020;53 doi: 10.1590/1414-431X20209029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Madu C.O., Wang S., Madu C.O., Lu M. Angiogenesis in Breast Cancer Progression, Diagnosis, and Treatment. J. Cancer. 2020;11:4474–4494. doi: 10.7150/jca.44313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Sun Z., Yan B. Multiple roles and regulatory mechanisms of the transcription factor GATA6 in human cancers. Clin Genet. 2020;97:64–72. doi: 10.1111/cge.13630. [DOI] [PubMed] [Google Scholar]
- 95.Tu J., Fang Y., Han D., Tan X., Jiang H., Gong X., et al. Activation of nuclear factor-kappaB in the angiogenesis of glioma: Insights into the associated molecular mechanisms and targeted therapies. Cell Prolif. 2021;54 doi: 10.1111/cpr.12929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Xu H., Zhao G., Zhang Y., Jiang H., Wang W., Zhao D., et al. Long non-coding RNA PAXIP1-AS1 facilitates cell invasion and angiogenesis of glioma by recruiting transcription factor ETS1 to upregulate KIF14 expression. J Exp Clin Cancer Res. 2019;38 doi: 10.1186/s13046-019-1474-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Eyler C.E., Rich J.N. Survival of the fittest: cancer stem cells in therapeutic resistance and angiogenesis. J Clin Oncol. 2008;26:2839–2845. doi: 10.1200/JCO.2007.15.1829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Fan L., Huang X., Chen J., Zhang K., Gu Y.H., Sun J., et al. Long Noncoding RNA MALAT1 Contributes to Sorafenib Resistance by Targeting miR-140-5p/Aurora-A Signaling in Hepatocellular Carcinoma. Mol Cancer Ther. 2020;19:1197–1209. doi: 10.1158/1535-7163.MCT-19-0203. [DOI] [PubMed] [Google Scholar]
- 99.Zhou Y., Huang Y., Dai T., Hua Z., Xu J., Lin Y., et al. LncRNA TTN-AS1 intensifies sorafenib resistance in hepatocellular carcinoma by sponging miR-16-5p and upregulation of cyclin E1. Biomed Pharmacother. 2021;133 doi: 10.1016/j.biopha.2020.111030. [DOI] [PubMed] [Google Scholar]
- 100.Li D., Li C., Chen Y., Teng L., Cao Y., Wang W., et al. LncRNA HOTAIR induces sunitinib resistance in renal cancer by acting as a competing endogenous RNA to regulate autophagy of renal cells. Cancer Cell Int. 2020;20 doi: 10.1186/s12935-020-01419-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Liu Y., Cheng G., Huang Z., Bao L., Liu J., Wang C., et al. Long noncoding RNA SNHG12 promotes tumour progression and sunitinib resistance by upregulating CDCA3 in renal cell carcinoma. Cell Death Dis. 2020;11 doi: 10.1038/s41419-020-2713-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Shan L., Liu W., Zhan Y. Long Non-coding RNA CCAT1 Acts as an Oncogene and Promotes Sunitinib Resistance in Renal Cell Carcinoma. Front Oncol. 2020;10 doi: 10.3389/fonc.2020.516552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Gu G., Hu C., Hui K., Chen T., Zhang H., Jiang X. NEAT 1 knockdown enhances the sensitivity of human non-small-cell lung cancer cells to anlotinib. Aging (Albany NY). 2021;13:13941–13953. doi: 10.18632/aging.203004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Llovet J.M., Ricci S., Mazzaferro V., Hilgard P., Gane E., Blanc J.F., et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–390. doi: 10.1056/NEJMoa0708857. [DOI] [PubMed] [Google Scholar]
- 105.Cheng A.L., Kang Y.K., Chen Z., Tsao C.J., Qin S., Kim J.S., et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. The Lancet Oncology. 2009;10:25–34. doi: 10.1016/S1470-2045(08)70285-7. [DOI] [PubMed] [Google Scholar]
- 106.Tang W., Chen Z., Zhang W., Cheng Y., Zhang B., Wu F., et al. The mechanisms of sorafenib resistance in hepatocellular carcinoma: theoretical basis and therapeutic aspects. Signal Transduct Target Ther. 2020;5 doi: 10.1038/s41392-020-0187-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Lin X., Xiang X., Hao L., Wang T., Lai Y., Abudoureyimu M., et al. The role of Aurora-A in human cancers and future therapeutics. Am J Cancer Res. 2020;10:2705–2729. [PMC free article] [PubMed] [Google Scholar]
- 108.Jalalirad M., Haddad T.C., Salisbury J.L., Radisky D., Zhang M., Schroeder M., et al. Aurora-A kinase oncogenic signaling mediates TGF-β-induced triple-negative breast cancer plasticity and chemoresistance. Oncogene. 2021;40:2509–2523. doi: 10.1038/s41388-021-01711-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Lai C.H., Chen R.Y., Hsieh H.P., Tsai S.J., Chang K.C., Yen C.J., et al. A selective Aurora-A 5’-UTR siRNA inhibits tumor growth and metastasis. Cancer Lett. 2020;472:97–107. doi: 10.1016/j.canlet.2019.12.031. [DOI] [PubMed] [Google Scholar]
- 110.Miura A., Sootome H., Fujita N., Suzuki T., Fukushima H., Mizuarai S., et al. TAS-119, a novel selective Aurora A and TRK inhibitor, exhibits antitumor efficacy in preclinical models with deregulated activation of the Myc, beta-Catenin, and TRK pathways. Invest New Drugs. 2021;39:724–735. doi: 10.1007/s10637-020-01019-9. [DOI] [PubMed] [Google Scholar]
- 111.Milioli H.H., Alexandrou S., Lim E., Caldon C.E. Cyclin E1 and cyclin E2 in ER+ breast cancer: prospects as biomarkers and therapeutic targets. Endocr Relat Cancer. 2020;27:R93–R112. doi: 10.1530/ERC-19-0501. [DOI] [PubMed] [Google Scholar]
- 112.Wei R., Thanindratarn P., Dean D.C., Hornicek F.J., Guo W., Duan Z. Cyclin E1 is a prognostic biomarker and potential therapeutic target in osteosarcoma. J Orthop Res. 2020;38:1952–1964. doi: 10.1002/jor.24659. [DOI] [PubMed] [Google Scholar]
- 113.Nassif E., Thibault C., Vano Y., Fournier L., Mauge L., Verkarre V., et al. Sunitinib in kidney cancer: 10 years of experience and development. Expert Rev Anticancer Ther. 2017;17:129–142. doi: 10.1080/14737140.2017.1272415. [DOI] [PubMed] [Google Scholar]
- 114.Zhang Y., Yin W., Cao W., Chen P., Bian L., Ni Q. CDCA3 is a potential prognostic marker that promotes cell proliferation in gastric cancer. Oncol Rep. 2019;41:2471–2481. doi: 10.3892/or.2019.7008. [DOI] [PubMed] [Google Scholar]
- 115.Zhang W., Lu Y., Li X., Zhang J., Zheng L., Zhang W., et al. CDCA3 promotes cell proliferation by activating the NF-kappaB/cyclin D1 signaling pathway in colorectal cancer. Biochem Biophys Res Commun. 2018;500:196–203. doi: 10.1016/j.bbrc.2018.04.034. [DOI] [PubMed] [Google Scholar]
- 116.Adams M.N., Burgess J.T., He Y., Gately K., Snell C., Zhang S.D., et al. Expression of CDCA3 Is a Prognostic Biomarker and Potential Therapeutic Target in Non–Small Cell Lung Cancer. Journal of Thoracic Oncology. 2017;12:1071–1084. doi: 10.1016/j.jtho.2017.04.018. [DOI] [PubMed] [Google Scholar]
- 117.Zhou M., Chen X., Zhang H., Xia L., Tong X., Zou L., et al. China National Medical Products Administration approval summary: anlotinib for the treatment of advanced non-small cell lung cancer after two lines of chemotherapy. Cancer Commun (Lond). 2019;39 doi: 10.1186/s40880-019-0383-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Han B., Li K., Wang Q., Zhang L., Shi J., Wang Z., et al. Effect of Anlotinib as a Third-Line or Further Treatment on Overall Survival of Patients With Advanced Non-Small Cell Lung Cancer: The ALTER 0303 Phase 3 Randomized Clinical Trial. JAMA Oncol. 2018;4:1569–1575. doi: 10.1001/jamaoncol.2018.3039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Chen L.M., Niu Y.D., Xiao M., Li X.J., Lin H. LncRNA NEAT1 regulated cell proliferation, invasion, migration and apoptosis by targeting has-miR-376b-3p/SULF1 axis in non-small cell lung cancer. Eur Rev Med Pharmacol Sci. 2020;24:4810–4821. doi: 10.26355/eurrev_202005_21170. [DOI] [PubMed] [Google Scholar]
- 120.Zhao M.M., Ge L.Y., Yang L.F., Zheng H.X., Chen G., Wu L.Z., et al. LncRNA NEAT1/miR-204/NUAK1 Axis is a Potential Therapeutic Target for Non-Small Cell Lung Cancer. Cancer Manag Res. 2020;12:13357–13368. doi: 10.2147/CMAR.S277524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Li B., Gu W., Zhu X. NEAT1 mediates paclitaxel-resistance of non-small cell of lung cancer through activation of Akt/mTOR signalling pathway. Journal of Drug Targeting. 2019;27:1061–1067. doi: 10.1080/1061186X.2019.1585437. [DOI] [PubMed] [Google Scholar]
- 122.Ye W., Ni Z., Yicheng S., Pan H., Huang Y., Xiong Y., Liu T. Anisomycin inhibits angiogenesis in ovarian cancer by attenuating the molecular sponge effect of the lncRNAMeg3/miR421/PDGFRA axis. Int. J. Oncol. 2019;55:1296–1312. doi: 10.3892/ijo.2019.4887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Shi H., Sun Y., He M., Yang X., Hamada M., Fukunaga T., Zhang X., Chang C. Targeting the TR4 nuclear receptor-mediated lncTASR/AXL signaling with tretinoin increases the sunitinib sensitivity to better suppress the RCC progression. Oncogene. 2020;39:530–545. doi: 10.1038/s41388-019-0962-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Wang Y., Wu S., Zhu X., Zhang L., Deng J., Li F., Guo B., Zhang S., Wu R., Zhang Z., et al. LncRNA-encoded polypeptide ASRPS inhibits triple-negative breast cancer angiogenesis. J. Exp. Med. 2020;217:20190950. doi: 10.1084/jem.20190950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Tang S., Tan G., Jiang X., Han P., Zhai B., Dong X., Qiao H., Jiang H., Sun X. An artificial lncRNA targeting multiple miRNAs overcomes sorafenib resistance in hepatocellular carcinoma cells. Oncotarget. 2016;7:73257–73269. doi: 10.18632/oncotarget.12304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Eslami S.Z., Cortes-Hernandez L.E., Cayrefourcq L., Alix-Panabieres C. The different facets of liquid biopsy: a kaleidoscopic view. Cold Spring Harb Perspect. Med. 2020;10:a037333. doi: 10.1101/cshperspect.a037333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Tong Y.S., Wang X.W., Zhou X.L., Liu Z.H., Yang T.X., Shi W.H., Xie H.W., Lv J., Wu Q.Q., Cao X.F. Identification of the long non-coding RNA POU3F3 in plasma as a novel biomarker for diagnosis of esophageal squamous cell carcinoma. Mol. Cancer. 2015;14:3. doi: 10.1186/1476-4598-14-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Weber D.G., Casjens S., Brik A., Raiko I., Lehnert M., Taeger D., Gleichenhagen J., Kollmeier J., Bauer T.T., Brüning T., et al. Circulating long non-coding RNA GAS5 (growth arrest-specific transcript 5) as a complement marker for the detection of malignant mesothelioma using liquid biopsies. Biomarker Res. 2020;8:15. doi: 10.1186/s40364-020-00194-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Guo X., Lv X., Ru Y., Zhou F., Wang N., Xi H., Zhang K., Li J., Chang R., Xie T., et al. Circulating exosomal gastric cancer-associated long noncoding RNA1 as a biomarker for early detection and monitoring progression of gastric cancer: a multiphase study. JAMA Surg. 2020;155:572–579. doi: 10.1001/jamasurg.2020.1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Baassiri A., Nassar F., Mukherji D., Shamseddine A., Nasr R., Temraz S. Exosomal non coding RNA in LIQUID biopsies as a promising biomarker for colorectal cancer. Int. J. Mol. Sci. 2020;21:1398. doi: 10.3390/ijms21041398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Lee Y.R., Kim G., Tak W.Y., Jang S.Y., Kweon Y.O., Park J.G., Lee H.W., Han Y.S., Chun J.M., Park S.Y., et al. Circulating exosomal noncoding RNAs as prognostic biomarkers in human hepatocellular carcinoma. Int. J. Cancer. 2019;144:1444–1452. doi: 10.1002/ijc.31931. [DOI] [PubMed] [Google Scholar]