Open Access

Bladder Cancer-related microRNAs With In Vivo Efficacy in Preclinical Models

ULRICH H. WEIDLE 1
  &  
FABIAN BIRZELE 2

1Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany

2Roche Pharma Research and Early Development, Pharmaceutical Sciences,Roche Innovation Center Basel, Basel, Switzerland

Cancer Diagnosis & Prognosis Sep-Oct; 1(4): 245-263 DOI: 10.21873/cdp.10033
Received 23 April 2021 | Revised 10 December 2024 | Accepted 15 June 2021
Corresponding author
Ulrich H. Weidle, Roche Innovation Center Munich, Roche Diagnostics GmbH, Nonnenwald 2, D-82372 Penzberg, Germany. E-mail weidle49@t-online.de and Fabian Birzele, Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland. E-mail fabian.birzele@roche.com
pdf image icon

Abstract

Progressive and metastatic bladder cancer remain difficult to treat. In this review, we critique seven up-regulated and 25 down-regulated microRNAs in order to identify new therapeutic entities and corresponding targets. These microRNAs were selected with respect to their efficacy in bladder cancer-related preclinical in vivo models. MicroRNAs and related targets interfering with chemoresistance, cell-cycle, signaling, apoptosis, autophagy, transcription factor modulation, epigenetic modification and metabolism are described. In addition, we highlight microRNAs targeting transmembrane receptors and secreted factors. We discuss druggability issues for the identified targets.
Keywords: Apoptosis, migration, invasion and proliferation, bladder cancer-related targets, cell cycle, epigenetic modifiers, orthotopic and xenograft models, tumor growth and metastasis, review

In 2020 in the US, 81,400 new cases of bladder cancer (BCa) were diagnosed, with a death toll of 18,000 (1). In the European Union 40,000 cases of BCa are diagnosed annually (2). The vast majority (90%) of cases are urothelial carcinomas which originate in the innermost tissue layer of the bladder and progress to muscle-invasive BCa, finally metastasizing to lymph nodes, bone, lung, liver and peritoneum (3). In this review, we focus on urothelial carcinoma of the bladder as BCa. Non-muscle-invasive BCa has a favorable prognosis and is treated with transurethral resection, intravesical chemotherapy and immunotherapy with Bacillus Calmette-Guerin (3). Only 60% of patients with muscle-invasive BCa are alive 5 years after treatment with neoadjuvant chemotherapy and surgery (4). In patients with metastatic disease, the medium survival time is 15 months (5). In addition to chemotherapy, several new agents have been approved for treatment of BCa such as erdafitinib for fibroblast growth factor receptor 3 (FGFR3) mutations and FGFR-related fusion proteins), enfortumab vedotin (monoclonal antibody to nectin 4 conjugated to monomethyl auristatin E), checkpoint inhibitory monoclonal antibodies such as pembrolizumab and nivolumab directed against programmed cell death protein 1, atezolizumab, durvalumab and atezolizumab directed against programmed death-ligand 1). These agents provide clinical benefit in progressive and metastatic scenarios of BCa but development of resistance is a commonly observed phenomenon (6,7). Therefore, the identification of new targets and development of new treatment regimens is a very important issue (8-10). In this review, we focus on microRNAs (miRs) with documented activity in preclinical in vivo models as a tool for the identification of new targets and as possible new entities for treatment of BCa.

miRs and their Role in Oncology

miRs are synthesized in the nucleus as precursors, processed and transported to the cytoplasm (11,12). Finally, one strand of a 22-nucleotide duplex is maintained (guide strand) while the other strand (passenger strand) is degraded (11,12). Binding of the guide strand to the 3’-untranslated region (3’-UTR) of the corresponding mRNA leads to degradation or translational repression of the target mRNA (11,12). miRs can interfere with expression of several target genes and can therefore modulate several pathways and potentially can rewire oncogenic networks (13). miRs can act as tumor suppressors or as oncogenes in a context-dependent manner (14-16). We have recently summarized the role of miRs with respect to the growth and metastasis of breast (17) and prostate cancer (18), non-small cell lung carcinoma (19), and hepatocellular (20) and pancreatic carcinoma (21). The role of miRs in BCa has been reviewed in (22,23). Here, we focus on BCa-related miRs with activity in preclinical in vivo models as tools for target identification and potential new entities for treatment of BCa.

miRs Up-regulated in BCa

miRs affecting apoptosis, autophagy and signaling

miR-21 targets protein phosphatase 2 regulatory subunit Bα (PPP2R2A). Locked nucleic acid miR-21 blocked anchorage-dependent growth and invasive capacity of RT-112 and 5637 BCa cells in vitro (24) and RT-112 xenografts after tail-vein or intraperitoneal injection (24). PPP2R2A was identified as a target of miR-21 (Figure 1) (24). Suppression of PPP2R2A activates the extracellular signal-regulated kinase (ERK) signaling pathway (24). PPP2R2A functions as a tumor suppressor in BCa and is a determinant of substrate specificity of the heterotrimeric serine/threonine protein phosphatase 2A (25,26). In non-small cell lung cancer, miR-136 was shown to promote ERK1/2 activation by targeting PPP2R2A (27).

miR-24-3p targets death-effector domain-containing protein (DEED). miR-24-3p (Figure 1) promoted proliferation migration and invasion, inhibited apoptosis and participated in autophagy in T24 and HBC BCa cells (28). miR-24 promoted growth of T24 BCa cells subcutaneously injected into nude mice (28). DEED was identified as a target of miR-24-3p (28). In BCa, miR-24-3p is overexpressed and expression of DEED is low (28). DEED functions as a tumor suppressor and reverses epithelial mesenchymal transition (EMT) by activating autophagy (29,30). DEED is the final target of a series of events by which cluster of differentiation 95-induced signals are transferred to nucleoli to shut-off cellular biosynthetic activities (31). DEED binds to DNA and reconstitutes mono-nucleosomes and inhibits transcription in a reconstituted in vitro system (31).

miR-135a targets glycogen synthase kinase-3β (GSK-3β). Mao et al. found that miR-135a (Figure 1) was increased in BCa tissue in comparison to adjacent normal tissue. miR-135a increased cell proliferation and migration and reduced apoptosis of T24 BCa cells. They showed in nude mice that miR-135a overexpression stimulated growth of T24 xenografts, whilst its inhibition suppressed xenograft growth. Glycogen synthase kinase-3β (GSK-3β) was identified as a target of miR-135a. miR-135a activated WNT/β-catenin signaling and EMT by inhibition of GSK-3β signaling (32). Other studies have also shown that GSK-3β inhibits WNT signaling, attenuates proliferation and stimulates apoptosis of BCa cells (33,34). β-Catenin activation is related to BCa progression (35). GSK-3β has been identified as a prognostic marker and potential target in BCa (35).

miR-495 targets phosphatase and tensin homolog deleted on chromosome 10 (PTEN). miR-495 (Figure 1) is overexpressed in BCa and corresponding cell lines. High miR-495 expression was correlated with larger tumor size, advanced TNM stage and lymph node metastasis. miR-495 was shown to promote BCa proliferation and invasion by targeting PTEN (36). In nude mice, miR-495 accelerated the growth of subcutaneous BCa xenografts in association with down-regulation of PTEN (36). PTEN metabolizes phosphatidylinositol 3,4,5 triphosphate, the ligand product of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K), thereby opposing activation of the PI3K/AKT serine/threonine kinase 1 (AKT)/mechanistic target of rapamycin (mTOR) signaling network (37-39). In BCa, down-regulation of PTEN has been correlated with poor prognosis, chemoresistance and progression (40). Targeting PI3K/AKT/mTOR is an important pathway for therapeutic intervention in BCa (41).

miR-516A targets PH domain leucine rich containing protein phosphatase 2 (PHLPP2). miR-516A (Figure 1) was shown to be up-regulated in BCa tissues and cell lines. miR-516A mediated proliferation and anchorage-independent growth of UMUC3 and J82 BCa cell lines in vitro and promoted tumor growth in nude mice of UMUC3 and J82 transfected with miR-516A. PHLPP2 has been identified as a target of miR-516A. The miR-516A–PHLPP2 cascade promoted E3 ubiquitin ligase-mediated Beclin1 (BECN1) degradation and thus attenuating autophagy and promoting BCa growth (42). PHLPP2 and its closely related paralog PHLPP1 are members of the protein phosphatase 2C family of Mg2+- and Mn2+-dependent phosphatases (43). They can inactivate signaling of AKT and protein kinase C by dephosphorylation of C-terminal hydrophobic phosphorylation motifs (43,44). BECN1 is a control protein that assembles cofactors for triggering autophagy induction (45,46). Autophagy has a multifaceted role in cancer and its function is context-dependent (45,46).

miRs Affecting Several Targets

miR-193a-3p targets splicing factor serine/arginine rich (SRSF2), plasminogen activator urokinase (PLAU), hypermethylated in cancer 2 (HIC2), lysyl oxidase-like 4 (LOXL4) and transcription factor homeobox C9 (HOXC9)

miR-193a-3p (Figure 2) regulates the activation of chemoresistance-associated signaling pathways. miR-193a-3p was found to have a positive effect on growth and chemoresistance of BCa xenografts after intra-tumoral injection in nude mice. Three direct targets have been identified as mediating these effects: Splicing factor serine/arginine rich (SRSF2), urokinase plasminogen activator (uPA) and hypermethylated in cancer 2 (HIC2) (47). SRSF2 is a regulator of splicing and nuclear-cytoplasmic transportation of mature RNA (48). Interaction of uPA with its receptor protects against anoikis by increasing the level of BCL2 apoptosis regulator- like 1-xL (49), which is in contradiction to the results described above which implicate down-regulation of uPA by miR-193a-3p in protection against apoptosis by a not yet resolved mechanism (47). HIC2 is hypermethylated in cancer and acts as a transcriptional repressor (50). Inhibition of these three targets results in modulation of five signaling pathways: DNA damage, NOTCH, nuclear factor ĸB (NFĸB), MYC and oxidative stress (50).

Another group has identified lysyl oxidase-like 4 (LOXL4) as a direct target of miR-193a, contributing to growth and chemoresistance of BCa cells. miR-193-3p levels were found to be higher in chemoresistant (H-bc and UM-UC-3) compared to chemosensitive BCa cell lines. Intra-tumoral injection of miR-193a-3p promoted growth of 5637 and H-bc BCa xenografts. miR-193a-3p activates the miR-193a/LOXL4/oxidative stress pathway with involvement of transcription factor nuclear facor erythroid 2-related factor (NRF2) (51). LOXL4 is an extracellular copper-dependent amine oxidase which plays a key role in matrix stability and integrity by catalyzing cross-links between collagens and elastin, leading to the preduction of insoluble fibers and is often deregulated in cancer (52). In BCa, LOXL4 has been shown to inhibit tumor growth (53).

Furthermore, it has been shown that miR-193-3p promotes chemoresistance by targeting transcription factor homeobox C9 (HOXC9) (54). HOXC9 participates in the miR-193a/HOXC9/DNA-damage response/oxidative stress pathway axis (54) and is involved in phenotype switching in breast cancer (55).

mir-145 targets forkhead box protein 1 (FOXO1) – stage-dependent function. miR-145 (Figure 2) was shown to be up-regulated in BCa tissues and T24T metastatic BCa cell line. Ectopic expression of miR-145 inhibited anchorage-independent growth of T24 BCa cells, however in T24T BCa cells, promotion of anchorage-independent growth by miR-145 was observed. Up-regulation of FOXO1 and its effector p27 was observed in T24 cells, whilst in T24T cells, these were down-regulated. miR-145 promoted xenograft formation in nude mice injected with T24T transfectants. From a mechanistic point of view, miR-145 was shown to bind to the 3’-UTR of FOXO1 in both T24 and T24T cell lines (56). miR-145 enhanced FOXO1 transcription by inhibiting phosphorylation of signal transducer and activator of transcription 3 (STAT3) at Tyr 705 in T24 cells but not in T24 cells, indicating that these differential effects are mediated by FOXO1. miR-145 was found to inhibit STAT3 phosphorylation at Tyr705 through targeting Janus kinase 2 (JAK2) mRNA coding sequence. JAK2 is responsible for phosphorylation of STAT3. Knock-down of STAT3 impaired miR-145 promotion of xenograft formation by T24T transfectants (56). FOXO1 induces cyclin-dependent kinase (CDK) inhibitor p27 and suppresses cyclin D1 and D2 expression (57). STAT3 is a transcription factor which plays a key role in growth and apoptosis of tumor cells (58,59). However, according to the data retrieved from the The Cancer Genome Atlas (TCGA) (Figure 3), miR-145 is down-regulated at the RNA steady-state level in BCa.

Down-regulated miRs

miRs affecting cell-cycle-related targets

miR-34b targets cyclin D2 (CCND2) and P2Y purinoreceptor 1 (P2RY1). miR-34b-3p (Figure 4) was shown to repress multi-drug chemoresistance of BCa cells 5637 and EJ. In a nude mouse xenograft model, miR-34b-3p inhibited tumor growth and paclitaxel resistance. CCND2 and P2RY1 have been identified as direct targets of miR-34b-3p (60). CCND2 forms complexes with CDK4/6, which mediate G1/S transition (61). P2RY1 is a G-protein-coupled receptor with ADP as an antagonist, couples to phospholipase C and triggers Ca2+ release from intracellular reservoirs; it is also involved in platelet aggregation (62,63). The details of how P2RY1 contributes to the phenomena as described above has not yet been resolved.

miR-124 targets CDK4. miR-124 (Figure 4) was found to be down-regulated in BCa tissue samples. CDK4 has been identified as a target of miR-124. miR-124 inhibited growth of HT1197 BCa cells by inducing cell-cycle arrest. miR-124 retarded growth of HT1197 BCa xenografts (64). Cell-cycle deregulation is a key driver of disease progression in BCa and it has been shown independently that inhibition of CDK4/6 controls proliferation of BCa cells (65). CDK4/6 inhibitors are being tested in more than 80 cancer-related clinical trials, including a trial in BCa (9,66).

miR-124-3p targets aurora kinase A (AURKA). miR-124-3p (Figure 4) was found to be down-regulated in BCa tissues and cell lines: miR-124-3p inhibited proliferation and invasion and induced apoptosis of BCa cells in vitro and inhibited growth of BCa cell xenografts in vivo. AURKA has been identified as a target for miR-124-3p (67). In BCa, AURKA was shown to promote cell-cycle progression, anti-apoptopic signaling, EMT and stem cell-like properties of cancer cells (68). Independent research has indicated that AURKA is a diagnostic biomarker for BCa detection, contributes to its aggressive behavior and predicts poor prognosis (69-71). Orally administrable AURKA inhibitor alisertib is being evaluated in several clinical studies in patients with cancer and is in phase III studies in patients with refractory peripheral T-cell lymphoma (72,73). In patients with advanced BCa, phase II studies are ongoing (74).

miiR-146a-3p targets pituitary tumor transforming gene 1 (PTTG1). miR-146a-3p (Figure 4) suppressed migration, invasion and cell-cycle progression and induced senescence in EJ and T24 BCa cells. miR-146a-3p inhibited growth and metastasis to the lungs of EJ BCa cells after tail vein injection into nude mice. PTTG1 was identified as a direct target of miR-146a-3p. PTTG1 expression was negatively correlated with that of miR-146a-3p in BCa tissues and cell lines (75). PTTG1 overexpression was shown to inhibit p21, an inducer of G1-phase arrest (76). p21 has been shown as a target of PTTG1 in pituitary tumor cells (76) In addition, PTTG1 is involved in cell transformation, aneuploidy and survival (77,78).

miR-1180-5p targets CDK4/6, cyclin D1 (CCND1) and cyclin D2 (CCND2). miR-1180-5p (Figure 4) attenuated proliferation, inhibited colony formation and induced cell-cycle arrest of T24 and EJ BCa cells. miR-1180-5p suppressed tumorigenicty of EJ cells after subcutaneous transplantation into nude mice. miR-1180-5p induced p21, which down-regulates cell-cycle regulators such as CDK4/6, CCND1 and CCNA2 (80). The activation of the p21 gene by miR-1180-5p is an example of a direct gene activation by a miR, other examples have been reported (79,80,81). p21 competitively binds to cyclins and reduces the formation of cyclin–CDK complexes (82). CDK4 and CDK6 are among the targets of p21 (83). CDK4/6 inhibitors palbociclib and ribociclib have been approved for treatment of hormone receptor-positive human epidermal growth factor receptor (EGFR) 2-negative metastatic breast cancer (84). Preclinical validation studies show that CDK4/6 inhibitors might be considered as clinical candidates for treatment of BCa (66,85).

miR-3619-5p targets CDK2 and β-catenin. Expression of miR-3619-5p (Figure 4) and p21 was found to be reduced in BCa tissues and cell lines, and associated with cancer progression. Low expression of p21 or decrease in both p21 and miR-3619-5p were associated with poor survival in patients with BCa (86). miR-3519-5p inhibited proliferation, migration, invasion, EMT and survival of 5637 and T24 BCa cells. Growth of T24 tumors in nude mice was delayed by overexpression of miR-3619-5p. miR-3619-5p was shown to up-regulate p21 by interaction with its promoter and targets binding sites in the 3’-UTRs of CDK2 and β-catenin (86). Low p21 expression predicted tumor recurrence and poor prognosis in BCa (87). β-Catenin-mediated WNT signaling was found to activated in BCa (88). CDK2 is under validation as a target for treatment of BCa (89).

miRs Targeting Transmembrane Proteins and Secreted Factors

miR-31 and miR-328-3p target integrin α5 (ITNΑ5). miR-31 (Figure 5) was down-regulated in BCa tissues and its expression correlated with individual progression. In T24 and 5637 BCa cells, miR-31 suppressed proliferation, invasion and migration and increased sensitivity to mitomycin C in vitro. ITNΑ5 has been identified as a target of miR-31 and inhibited AKT and ERK pathways in BCa cells. In nude mice, T24 BCa cells overexpressing miR-31 exhibited reduced tumor growth and increased sensitivity to mitomycin C (90). It has been shown that ITNΑ5 facilitates BCa invasion through enhanced contraction forces (91).

miR-328-3p (Figure 5) was down-regulated in BCa tissues and inhibited tumorigenesis by ITNΑ5 and inactivation of the PI3K pathway in BCa. Expression of miR-328-3p in T24 BCa cells inhibited proliferation in vitro and tumor growth in nude mice (92). Furthermore, a positive correlation between ITNΑ5 expression and malignant phenotype has been found in BCa (93).

miR-122 targets vascular endothelial growth factor (VEGFC). miR-122 (Figure 5) was down-regulated in BCa. miR-122 was shown to inhibit proliferation, migration, invasion and colony formation, and sensitized HT1376 BCa to cisplatin treatment. In HT1376 xenografts, miR-122 inhibited tumor growth and angiogenesis. VEGFC has been identified as a direct target of miR-122 (94). VEGFC participates in tumor angiogenesis and lymphangiogenesis. It is expressed in endothelial and tumor cells and mediates VEGFR2 and VEGFR3 signaling (95,96). In BCa, VEGFC expression is correlated with lymph node density and microvessel density (97).

miR-202 targets EGFR. A strong correlation between down-regulation of miR-202 (Figure 5) in BCa tissue and clinical characteristics such as T-classification, N-classification, recurrence and mortality was noted. In vitro, miR-202 inhibited cell proliferation, migration and invasion by T24, BIU87 and EJ BCa cells. Overexpression of miR-202 in these cells inhibited their growth as xenografts in nude mice after subcutaneous implantation. EGFR has been identified as a direct target of miR-202. Knock-down of EGFR inhibited cell proliferation, invasion and migration in T24, BIU87 and EJ Ba cell lines. (98). EGFR expression is deregulated in BCa and is correlated with disease progression (99,100). EGFR is involved in the pathogenesis and progression of several types of cancer (101). EGFR is a possible target for treatment of BCa (102).

miR-210-3p targets FGFR like-1 (FGFRL1). miR-210-3p (Figure 5) was found to be down-regulated in BCa. miR-210-3p suppressed growth of BCa cells T24 and SW780 in vitro and in vivo after implantation into the flanks of nude mice. FGFRL1 was identified as a direct target of miR-210-3p. The effect of miR-210-3p on BCa growth and migration was blocked by inhibition of FGFLR1 (103). The FGFR family consists of five transmembrane receptors which are deregulated in many types of cancer with downstream effectors such as phospholipase C, RAS–mitogen-activated protein kinase and PI3K-AKT (104). Erdafitinib was approved for treatment of metastatic or locally advanced BCa with activating mutations in FGFR3 or a fusion protein containing FGFR2 or -3 (105). Several clinical studies with compounds inhibiting FGFRs are ongoing in patients with metastatic or advanced BCa (106,107). FGFRL1, the fifth FGFR, lacks a protein tyrosine domain but contains a short intracellular tail with histidine-rich motifs (108). Aberrant expression of FGFRL1 has been noted in ovarian tumors (109). In BCa, its function has to be validated in more detail.

miRs Affecting Signaling

miR-100 targets mTOR). miR-100 (Figure 6) expression was found to reduced in human BCa tissues. miR-100 suppressed proliferation and migration of EJ and 5637 BCa cells in vitro. mTOR was identified as a direct target of miR-100. It reduced growth of subcutaneously or intravesically implanted EJ cells in nude mice (110). mTOR is a downstream effector of the PI3K/AKT signaling pathway and its inhibition results in induction of apoptosis and inhibition of the cell cycle, angiogenesis and protein translation (111-113). The PI3K/AKT/mTOR axis exhibits frequent molecular alterations in BCa but clinical trials with mTOR inhibitors have not met their endpoints (114). Reduction of miR-100 expression in BCa samples in comparison to non-transformed bladder tissues was confirmed by data from the TCGA (Figure 3).

miR-608 targets flotilin 1 (FLOT1). miR-608 (Figure 6) was found to be down-regulated in BCa. In vitro, miR-608 induced G1 phase arrest and inhibition of colony formation by T24 and UMUC3 BCa cells. UMUC3 cells transfected with miR-608 gave rise to slower growing tumors in nude mice (115). FOXO3a suppressed the expression of CCND1 and other related cell regulators by inducing tumor suppressors p21 and p27 (115-117). Up-regulation of miR-608 led to inhibition of AKT/FOXO3a signaling in BCa cells. FLOT1 has been identified as a direct target of miR-608 (115). FLOT1 is a scaffolding protein of lipid raft microdomains and a highly conserved lipid raft marker (118,119). It is involved in signal transduction, cell adhesion, cytoskeleton remodeling, endocytosis and acts as a signaling mediator by anchoring various receptors of signaling pathways to the cell membrane (118,119). The mechanistic underpinnings of inhibition of AKT/FOXO3 signaling and FLOT1 remain to be worked out. Independent research has shown overexpression of FLOT1 is involved in proliferation and recurrence of BCa (120). The tractability of FLOT1 for drug discovery is a critical issue.

miR-4324 targets RAC GTPase activating protein-1 (RACGAP1). miR-4324 (Figure 6) was shown to be down-regulated in BCa tissues. In T24 and UMUC3 BCa cells, miR-4324 inhibited growth, invasion and migration, induced G0/G1 phase cell-cycle arrest and mediated a reduction of colony-forming ability. In nude mice, UMUC3 cells transfected with miR-4324 exhibited reduced tumor growth after subcutaneous implantation and reduced lung metastasis after injection into the tail vein. RACGAP1 was identified as the target of miR-4324. RACGAP1 was shown to mediates doxorubicin resistance in BCa cell line UMUC3. miR-4324 is an estrogen receptor 1 target gene which is down-regulated by promoter methylation (121). RACGAP1 increases expression of STAT3 and mediates its translocation into the nucleus and acts as a nuclear chaperone for STAT3 (121-123). RACGAP1 was also found to regulate the activation of GTPases, RAS-related C3 botulinum toxin substrate 1 (RAC1) and cell division control protein 42 homolog (CDC42) trigger cytokinesis and cytoskeletal reorganization, regulating chemotaxis, cell polarity, migration and metastasis (124). RACGAP1 mediated recurrence and metastasis and was also found to regulate the polycomb repressive complex 1, which promotes cytoskeletal and transcriptional pathways enhancing cell motility (125). Targeting the miR-4234–RACGAP1–STAT3-estrogen receptor 1 feedback loop is a potential target for drug discovery in BCa. Tractability of RACGAP1 for drug discovery is a pending issue.

miRs Affecting Apoptosis

miR-138-5p targets survivin. miR-138-5p (Figure 6) was shown to inhibit proliferation of T24 BCa cells in vitro and growth of T24 transfectants in nude mice after subcutaneous implantation. Survivin has been identified as a direct target of miR-138-5p (126). Survivin is a member of the inhibitor of apoptosis (IAP) family of anti-apoptotic proteins containing a single baculovirus IAP repeat domain (126-128). Survivin protects cancer cells from apoptosis and autophagy, localizes to the mitotic spindle and interacts with tubulin during mitosis, activates AKT and PI3K signaling, induces angiogenesis and stemness and is a direct target of the WNT signaling pathway (127,128). Research has shown that survivin can induce proliferation of BCa cells (129). Expression of survivin is correlated with tumor grade and worse prognosis in patients with BCa (130,131). Survivin is an important target for anticancer drugs, however, clinical studies did not reach the projected endpoints. Therfore, new optimized compounds need to be developed (132).

miR-200c targets X-linked inhibitor of apoptosis (XIAP). miR-200c (Figure 6) expressed in T24 BCa cells attenuated lung metastasis after tail vein injection into nude mice and improved survival. XIAP was identified as a target of miR-200c. Expression of XIAP in BCa tissues was correlated with metastasis (133). XIAP contains a baculovirus IAP repeat, a ubiquitin-binding domain and a carboxy-terminal RING finger which catalyzes the ubiquitinylation of caspases 3, 7 and 9 (134). XIAP can be inhibited by small molecules targeting the BIR-domain or mimetics of the natural XIAP antagonist, referred to as second mitochondria-derived activator of caspase mimetics (135,136). XIAP promoted BCa cell survival and invasion in vitro and lung metastasis in nude mice (137,138). XIAP has been identified as a prognostic marker of early recurrence of non-muscular invasive BCa (139). ASTX 660, a non-peptidometic small-molecule antagonist of XIAP and cellular inhibitor of apoptosis 1/2 (cIAP1/2) is being clinically evaluated in patients with advanced cancer and lymphoma (140).

miRs Interfering With Autophagy

miR-154 targets autophagy-related 7 (ATG7). miR-154 (Figure 6) has been shown to be down-regulated in BCa and to suppress proliferation, migration and invasion of T24 and UMUC3 BCa cells in vitro. In nude mice, miR-154 inhibited proliferation and tumorigenesis of T24 BCa cells. ATG7 was identified as a direct target of miR-154 (141). ATG7 is essential for autophagy and autophagosome formation (142). Autophagy can prolong survival of cancer cells but can also activate tumor-suppressor genes in a context-dependent way (143). In BCa cells, inhibition of autophagy causes apoptosis and attenuation of invasion and migration via EMT inhibition (144,145). ATG7 is overexpressed in BCa and its inhibition might be a promising strategy for treatment of BCa (146).

miR Targeting Transcription Factors

miR-15 targets B-lymphoma Moloney murine insertion region 1 (BMI1). miR-15 (Figure 6) was found to be down-regulated in BCa tissues and its expression level inversely correlated with survival. miR-15 inhibited proliferation, migration and invasion of T24, Blu87 and HT1376 BCa cells. miR-15 inhibited EMT and PI3K/AKT signaling. In nude mice, growth of T24 cells transfected with miR-15 was inhibited after subcutaneous implantation. BMI1 was identified as a target of miR-15 (147). BMI1 is a member of the polycomb group of transcription factors which can act as repressors of transcription (148). BMI1 acts as an oncogene, inhibits PTEN, represses p16 and p19 (INKA/ARF genes) and is involved in self-renewal of cancer stem cells (148). Independently, miR-15 was shown to inhibit proliferation, migration and invasion of BCa cells (149,150). BMI1 was indicated as a prognostic marker in patients with BCa. Patients with higher BMI1 expression were found to have shorter survival (151).

miR-370 targets transcription factor SOX12 (SOX12). miR-370 (Figure 6) was reduced in BCa and inhibited proliferation, migration and invasion of 5637 BCa cells. In nude mice, tumor growth of 5637 cells transfected with miR-370 was inhibited after their subcutaneous implantation. Transcription factor SOX12 was identified as a direct target of miR-370 (152). SOX12 contains a DNA-binding high-mobility group domain (153). SOX12 has been poorly validated in BCa (154). However, in colorectal cancer, gastric cancer and hepatocellular carcinoma, SOX12 has been shown to promote proliferation and metastasis (154-156).

miRs Involved in Epigenetic Modification

miR-124 targets ubiquitin-like containing PH and ring domains, 1 (UHRF1). miR-124 (Figure 7) inhibited proliferation, colony formation, migration, invasion and vascular mimicry of T24 and J82 BCa cells. Intratumoral injection of miR-124 into T24 xenografts induced suppression of tumor growth in nude mice. UHRF1, a RING-finger like type R3 ubiquitin ligase, was identified as a direct target of miR-124 (157). UHRF-1 recruits DNA methyltransferase 1 to hemi-methylated DNA, coordinates methylation and histone modifications, mediates silencing of tumor-suppressor genes and is involved in DNA repair (158). Epigenetic repression of regulator of G-protein signaling by UHRF1 has been shown to promote BCa progression (159). Expression levels of UHRF1 was shown to be negatively correlated with clinical outcomes of patients with BCa (160).

miR-411 targets myeloid/lymphoid or mixed lineage translocated to 1 (MLLT1). miR-411 (Figure 7) has been shown to be down-regulated in BCa tissues and cell lines. In vitro, miR-141 suppressed proliferation and induced G2/M cell-cycle arrest in T24 and UMUC3 BCa cell lines. G2/M cell-cycle arrest is mediated by up-regulation of p21. miR-411 also attenuated growth of T24 and UMUC3 xenografts in nude mice after subcutaneous implantation (161). MLLT1 has been identified as a target of miR-411. MLLT1 is a histone acetylation reader and plays a role in chromatin remodeling, expression of oncogenes such as MYC and homeobox genes via histone acetylation and is a critical component of the super elongation complex (162). The mechanism by which miR-411 induces p21 still has to be resolved. MLLT1 plays a role as an oncogene in leukemia development (162). Small molecules disrupting the YEATS reader pocket of MLLT1 are candidates for treatment of acute myeloid leukemia (162). In BCa, further validation of MLLT1 is necessary with respect to its role as a therapeutic target.

miRs Interfering With Metabolism-related Targets

miR-1-3p targets glutaminase (GLS). miR-1-3p (Figure 7) expression was reduced in BCa and corresponding cell lines. In BCa cell lines T24 and UMUC3, miR-1-3p inhibited proliferation, invasion and migration in vitro. In nude mice, T24 cells transfected with miR-124 exhibited reduced tumor growth after subcutaneous implantation. GLS was identified as a direct target of miR-1-3p (163). Increased glutamine metabolism (glutaminolysis) is a hallmark of cancer and a key metabolic change in cancer cells (164,165). Small-molecule inhibitors of GLS are currently under preclinical validation (166). Data derived from the TCGA confirm the down-regulation of miR-1 in BCa tissue in comparison to non-transformed bladder tissues (Figure 3).

miR-145 targets MYC/polypyrimidine tract binding protein 1 (PTBP1)/ muscle pyruvate kinase isoenzymes 1/2 (PKM) axis. miR-145 (Figure 7) was shown to be down-regulated in clinical BCa and corresponding cell lines. In BCa cell lines T24 and 253JB-V, miR-145 inhibited growth and induced apoptosis in vitro. Intra-tumoral injection of miR-145 into 253JB-V xenografts exerted an antitumoral effect in nude mice (167). Inhibition of miR-145 induced differential splicing of PKM isoforms PKM1 and PKM2 in favor of PKM2, which induces the Warburg effect, resulting in increased glycolysis regardless of oxygen availability (167,168). The observed shift in isoforms was shown to be induced by down-regulation of MYC and up-regulation of PTBP1 (167). PTBP1 is part of the heterogenous nuclear ribonucleoprotein family, which are regulators of splicing (169). MYC has been identified as a direct target of miR-145 (170). miR-145 was shown to impair the MYC–PTBP1–PKM axis and also inhibit mitogen-activated protein kinase/ERK and PI3K/AKT signaling (167). Down-regulation of miR-145 in BCa tissues in comparison to corresponding normal bladder tissues is confirmed by data from TCGA (Figure 3).

miR-153 targets indoleamine 2,3 dioxygenase1 (IDO1). miR-153 (Figure 7) is down-regulated in BCa tissues and cell lines. In T24 and UMUC3 BCa cells, miR-153 induced apoptosis, reduced colony formation, and inhibited invasion, migration and EMT in vitro. miR-153 inhibited tube formation in human umbilical vein endothelial cells and angiogenesis in the chorioallantoic membrane assay. In nude mice, miR-153 inhibited tumor growth of T24 xenografts transduced with miR-153 after subcutaneous implantation. IDO1 was identified as a direct target of miR-153 (171). IDO1 is a rate-limiting enzyme in tryptophan metabolism (172). IDO1 also induces interleukin 6 (IL6) which interacts with IL6 receptor and induces STAT3 and VEGF, which promotes angiogenesis (171). IDO1 expression predicted poorer survival in patients with early-stage BCa (173). IDO1 also induces immuno-suppression. Several IDO1 inhibitors are under clinical evaluation in patients with cancer (174). Recently, IDO1 inhibitor epacadostat has failed in patients with melanoma (175). Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma did not improve progression-free survival or overall survival. The exploration of the clinical value of IDO1 inhibitors in combination with other agents is ongoing.

miR-612 targets malic enzyme 1 (ME1). miR-612 (Figure 7) was down-regulated in BCa tissues and cell lines. In T24 BCa cell line, miR-612 reduced tumor growth, colony formation, migration, invasion and EMT in vitro and in xenografts in nude mice after subcutaneous implantation. ME1 has been identified as a direct target of miR-612 (176). ME1 links the glycolytic and citric cycles and is important for NADP production, glutamine metabolism and lipogenesis (177). ME1 is a mediator of EMT by inhibition of expression of E-cadherin and induction of membrane metalloproteinase 9, N-cadherin and vimentin (177). ME1 was shown to promote EMT and metastasis in hepatocellular carcinoma and is a marker for poor prognosis (178).

miRs Affecting Several Targets

miR-582-5p and miR-3p target protein geranylgeranyl transferase type 1 subunit β (PGGT1B), leucine-rich repeat kinase 2 (LRRK2), DIX domain-containing 1 (DIXDC1) and RAS-related GTP-binding protein 27A (RAB27A). miR-582-5p and miR-3p (Figure 7) were down-regulated in BCa tissues and corresponding cell lines. miR-582-5p and miR-3p interfered with proliferative and invasive abilities of UMUC3, J82 and TCCSUP BCa cells. In an orthotopic BCa xenograft model, tumor growth and lung metastasis of UMUC3 BCa cells were inhibited by transurethrally delivered miR-582-5p or miR-3p complexes. PGGT1B, LRRK2, DIXDC1 and RAB27A were identified as targets of miR-582-5p and miR-3p (179). PGGT1B geranylates ras homolog family members, disrupts F-actin organization and promotes motility, invasion and metastasis (180,181). LRRK2 directly phosphorylates AKT1, resulting in cell survival and prevention of apoptosis (182). DIXDC1 up-regulates CCND1, down-regulates p21 and inhibits G1/S arrest (183). RAB27A functions as a promoter of proliferation, invasion and metastasis (184). RAB27A was shown to promote BCa cell proliferation and chemoresistance through regulation of NFĸB signaling (185).

miR-502 targets CCND1, DNA methyltransferase 3β (DNMT3B) and nucleolar protein 14 (NOP14). miR-502 (Figure 7) has been shown to be frequently down-regulated in BCa. miR-502 inhibited proliferation of T24 and UMUC3 BCa cell lines in vitro and growth of UMUC3 cells transfected with miR-502 in nude mice after subcutaneous implantation (186). miR-502 directly targeted CCND1 (187), NOP14 (188,189) and DNMT3B) (190,191). CCND1 forms a complex with CDK4 and CDK6, which are responsible for G1/S transition (187).

Conclusion

We have identified seven miRs up-regulated and 25 down-regulated in BCa-related tissues with efficacy in preclincal BCa-related in vivo models. Up-regulated miRs can be inhibited by single-stranded RNAs, with 12-25 nucleotides complementary to the corresponding mRNA (192) or with miR sponges which contain multiple miR-binding sites which compete with the specific mRNA for binding of the corresponding miR (193). Inhibition of miRs can also be achieved with small molecules which interfere with their transcription or their secondary structure, but specificity issues of the identified compounds are a major issue (194,195). Candidates drug targets are miR-21, miR-24-3p, miR-135a, miR-495, miR-516A and miR-193a-3p (Figures 1 and 2). Inhibition of miR-193a might be of relevance in overcoming chemoresistance to several drugs (Figure 2). Expression of miR-145 is context-dependent and therefore not a priority target. In the case of PPP2R2A, DEED, PTEN, PHLPP2 and FOXO1 (Figures 1 and 2) druggabilty due to the absence of protein pockets or unresolved interactions with other proteins is a critical issue with the exception of GSK-3β.

Down-regulated miRs can be substituted by reconstitution therapy, their corresponding targets are candidates for inhibition by small molecules or antibody-based entities. Functional reconstitution of miRs can be achieved with miR mimetics, double-stranded RNAs designed to mimic endogenous mature miRs or to express the corresponding miR with plasmid- or virus-based expression vectors in corresponding recipient cells (195,196). Analysis of down-regulated miRs provides several promising miRs for reconstitution therapy and targets for inhibitory compounds. All of the down-regulated miRs need to be validated in more detail with respect to their propensity for functional reconstitution and resulting physiological consequences. miR-34b, miR-124, miR-146-3p, miR-1180-5p and miR-3619-5p have targets such as CDK2/4/6 and AURKA which are druggable and which have a high priority in development of BCa agents (Figure 4) (9). miR-31, miR-328, miR-122, miR-202 and miR-210-3p have targets such as INTΑ5, VEGFC, EGFR and FGFRL1 (Figure 5), which are candidates for further validation studies. In the signaling category, miR-100 points to mTOR as a target (Figure 5). Validation of mTOR inhibitors is actively being pursued in BCa-related settings (9). miR-608 and miR-4324 have FLOT1 and RACGAP1 as targets but druggability is a critical issue (Figure 6). miR-138-5p and miR-200c target anti-apoptotic proteins such as survivin and XIAP, corresponding inhibitors for which are already under validation in clinical studies (Figure 6). miR-154 identifies autophagy protein ATG7 and transcription factors, and miR-15 and miR-370 have BMI and SOX12 (miR-370) as possible targets for therapeutic intervention in patients with BCa, but druggability is a critical issue (Figure 6). miR-124 and miR-411 identify epigenetic modifiers such as UHRF1 and MLLT1 as new targets for small-molecule epigenetic modifiers but here tractability issues also have been resolved (Figure 7). miR1-3p, miR-145a, miR-153 and miR-612 have metabolism-related targets such as GLS, PKM1 and -2, IDO1 and ME1 as possible new targets for intervention with small molecules. Inhibitors of these targets are under preclinical validation or are under clinical investigation as outlined in the previous sections.

Interestingly, miR-145, targeting FOXO1 or PTBP1/PKM, can be up- or down-regulated in BCa-related cell lines and tissues (Figures 2 and 7). miR-124c targets AURKA, CDK4 and UHRF1 (Figures 4 and 7) with in vivo efficacy against each of these targets. miR-502 and miR-582-3p and miR-5p modulate several targets. The contributions of each target to in vivo efficacy have to be resolved in further detail (Figure 7).

Various issues have to addressed in the preclinical validation process, case by case. These issues are not discussed in further detail in this review. Critical issues are: The optimization of pharmacokinetic and pharmacodynamic properties due to removal by the reticulo-endothelial system, renal excretion, entry into tumor cells and efficiency of intracellular endosomal escape. Further critical issues are hybridization-dependent and -independent side-effects, immunogenicity, hematological and hepato-toxicity, and cytokine-release syndrome (195-202). Recently the field of miR-based therapeutics has witnessed several setbacks, mainly due to toxicity issues (203). The upcoming years will show whether proof-of-concept clinical studies with miR-based therapeutics are feasible.

Conflicts of Interest

FB is and UHW was an employee of Roche.

Authors’ Contributions

FB and UHW jointly conceived and prepared the manuscript.

References

1 Siegel RL Miller KD & Jemal A Cancer statistics, 2020. CA Cancer J Clin. 70(1) 7 - 30 2020. PMID: 31912902. DOI: 10.3322/caac.21590
2 Burger M Catto JW Dalbagni G Grossman HB Herr H Karakiewicz P Kassouf W Kiemeney LA La Vecchia C Shariat S & Lotan Y Epidemiology and risk factors of urothelial bladder cancer. Eur Urol. 63(2) 234 - 241 2013. PMID: 22877502. DOI: 10.1016/j.eururo.2012.07.033
3 Shinagare AB Ramaiya NH Jagannathan JP Fennessy FM Taplin ME & Van den Abbeele AD Metastatic pattern of bladder cancer: correlation with the characteristics of the primary tumor. AJR Am J Roentgenol. 196(1) 117 - 122 2011. PMID: 21178055. DOI: 10.2214/AJR.10.5036
4 Grossman HB Natale RB Tangen CM Speights VO Vogelzang NJ Trump DL deVere White RW Sarosdy MF Wood DP Jr Raghavan D & Crawford ED Neoadjuvant chemotherapy plus cystectomy compared with cystectomy alone for locally advanced bladder cancer. N Engl J Med. 349(9) 859 - 866 2003. PMID: 12944571. DOI: 10.1056/NEJMoa022148
5 von der Maase H Sengelov L Roberts JT Ricci S Dogliotti L Oliver T Moore MJ Zimmermann A & Arning M Long-term survival results of a randomized trial comparing gemcitabine plus cisplatin, with methotrexate, vinblastine, doxorubicin, plus cisplatin in patients with bladder cancer. J Clin Oncol. 23(21) 4602 - 4608 2005. PMID: 16034041. DOI: 10.1200/JCO.2005.07.757
6 Patel VG Oh WK & Galsky MD Treatment of muscle-invasive and advanced bladder cancer in 2020. CA Cancer J Clin. 70(5) 404 - 423 2020. PMID: 32767764. DOI: 10.3322/caac.21631
7 DeGeorge KC Holt HR & Hodges SC Bladder cancer: Diagnosis and treatment. Am Fam Physician. 96(8) 507 - 514 2017. PMID: 29094888.
Pubmed |
8 Knowles MA & Hurst CD Molecular biology of bladder cancer: new insights into pathogenesis and clinical diversity. Nat Rev Cancer. 15(1) 25 - 41 2015. PMID: 25533674. DOI: 10.1038/nrc3817
9 Carneiro BA Meeks JJ Kuzel TM Scaranti M Abdulkadir SA & Giles FJ Emerging therapeutic targets in bladder cancer. Cancer Treat Rev. 41(2) 170 - 178 2015. PMID: 25498841. DOI: 10.1016/j.ctrv.2014.11.003
10 Felsenstein KM & Theodorescu D Precision medicine for urothelial bladder cancer: update on tumour genomics and immunotherapy. Nat Rev Urol. 15(2) 92 - 111 2018. PMID: 29133939. DOI: 10.1038/nrurol.2017.179
11 Hata A & Kashima R Dysregulation of microRNA biogenesis machinery in cancer. Crit Rev Biochem Mol Biol. 51(3) 121 - 134 2016. PMID: 26628006. DOI: 10.3109/10409238.2015.1117054
12 Gulyaeva LF & Kushlinskiy NE Regulatory mechanisms of microRNA expression. J Transl Med. 14(1) 143 2016. PMID: 27197967. DOI: 10.1186/s12967-016-0893-x
13 Peter ME Targeting of mRNAs by multiple miRNAs: the next step. Oncogene. 29(15) 2161 - 2164 2010. PMID: 20190803. DOI: 10.1038/onc.2010.59
14 Fasoulakis Z Daskalakis G Diakosavvas M Papapanagiotou I Theodora M Bourazan A Alatzidou D Pagkalos A & Kontomanolis E MicroRNAs determining carcinogenesis by regulating oncogenes and tumor suppressor genes during cell cycle. MicroRNA. 9(2) 82 - 92 2021. DOI: 10.2174/2211536608666190919161849
15 Gailhouste L & Ochiya T Cancer-related microRNAs and their role as tumor suppressors and oncogenes in hepatocellular carcinoma. Histol Histopathol. 28(4) 437 - 451 2013. PMID: 23224781. DOI: 10.14670/HH-28.437
16 Zhang B Pan X Cobb GP & Anderson TA microRNAs as oncogenes and tumor suppressors. Dev Biol. 302(1) 1 - 12 2007. PMID: 16989803. DOI: 10.1016/j.ydbio.2006.08.028
17 Weidle UH Dickopf S Hintermair C Kollmorgen G Birzele F & Brinkmann U The role of micro RNAs in breast cancer metastasis: Preclinical validation and potential therapeutic targets. Cancer Genomics Proteomics. 15(1) 17 - 39 2018. PMID: 29275360. DOI: 10.21873/cgp.20062
18 Weidle UH Epp A Birzele F & Brinkmann U the functional role of prostate cancer metastasis-related micro-RNAs. Cancer Genomics Proteomics. 16(1) 1 - 19 2019. PMID: 30587496. DOI: 10.21873/cgp.20108
19 Weidle UH Birzele F & Nopora A MicroRNAs as potential targets for therapeutic intervention with metastasis of non-small cell lung cancer. Cancer Genomics Proteomics. 16(2) 99 - 119 2019. PMID: 30850362. DOI: 10.21873/cgp.20116
20 Weidle UH Schmid D Birzele F & Brinkmann U MicroRNAs involved in metastasis of hepatocellular carcinoma: Target candidates, functionality and efficacy in animal models and prognostic relevance. Cancer Genomics Proteomics. 17(1) 1 - 21 2020. PMID: 31882547. DOI: 10.21873/cgp.20163
21 Weidle UH Birzele F & Nopora A Pancreatic ductal adenocarcinoma: MicroRNAs affecting tumor growth and metastasis in preclinical in vivo models. Cancer Genomics Proteomics. 16(6) 451 - 464 2019. PMID: 31659100. DOI: 10.21873/cgp.20149
22 Xie Y Ma X Chen L Li H Gu L Gao Y Zhang Y Li X Fan Y Chen J & Zhang X MicroRNAs with prognostic significance in bladder cancer: a systematic review and meta-analysis. Sci Rep. 7(1) 5619 2017. PMID: 28717125. DOI: 10.1038/s41598-017-05801-3
23 Li Q Wang H Peng H Huang Q Huyan T Huang Q Yang H & Shi J MicroRNAs: Key players in bladder cancer. Mol Diagn Ther. 23(5) 579 - 601 2019. PMID: 31325035. DOI: 10.1007/s40291-019-00410-4
24 Koutsioumpa M Chen HW O’Brien N Koinis F Mahurkar-Joshi S Vorvis C Soroosh A Luo T Issakhanian S Pantuck AJ Georgoulias V Iliopoulos D Slamon DJ & Drakaki A MKAD-21 suppresses the oncogenic activity of the miR-21/PPP2R2A/ERK molecular network in bladder cancer. Mol Cancer Ther. 17(7) 1430 - 1440 2018. PMID: 29703843. DOI: 10.1158/1535-7163.MCT-17-1049
25 Shi Y Serine/threonine phosphatases: mechanism through structure. Cell. 139(3) 468 - 484 2009. PMID: 19879837. DOI: 10.1016/j.cell.2009.10.006
26 Fowle H Zhao Z & Graña X PP2A holoenzymes, substrate specificity driving cellular functions and deregulation in cancer. Adv Cancer Res. 144 55 - 93 2019. PMID: 31349904. DOI: 10.1016/bs.acr.2019.03.009
27 Shen S Yue H Li Y Qin J Li K Liu Y & Wang J Upregulation of miR-136 in human non-small cell lung cancer cells promotes Erk1/2 activation by targeting PPP2R2A. Tumour Biol. 35(1) 631 - 640 2014. PMID: 23959478. DOI: 10.1007/s13277-013-1087-2
28 Yu G Jia Z & Dou Z miR-24-3p regulates bladder cancer cell proliferation, migration, invasion and autophagy by targeting DEDD. Oncol Rep. 37(2) 1123 - 1131 2017. PMID: 28000900. DOI: 10.3892/or.2016.5326
29 Lv Q Hua F & Hu ZW DEDD, a novel tumor repressor, reverses epithelial-mesenchymal transition by activating selective autophagy. Autophagy. 8(11) 1675 - 1676 2012. PMID: 22874565. DOI: 10.4161/auto.21438
30 Hua F Xue JF Lü XX & Hu ZW [DEDD decreases Smad3 activity, promotes tumor cell apoptosis and inhibits proliferation]. Yao Xue Xue Bao. 48(5) 680 - 685 2013. PMID: 23888690.
Pubmed |
31 Stegh AH Schickling O Ehret A Scaffidi C Peterhänsel C Hofmann TG Grummt I Krammer PH & Peter ME DEDD, a novel death effector domain-containing protein, targeted to the nucleolus. EMBO J. 17(20) 5974 - 5986 1998. PMID: 9774341. DOI: 10.1093/emboj/17.20.5974
32 Mao XW Xiao JQ Li ZY Zheng YC & Zhang N Effects of microRNA-135a on the epithelial-mesenchymal transition, migration and invasion of bladder cancer cells by targeting GSK3β through the Wnt/β-catenin signaling pathway. Exp Mol Med. 50(1) e429 2018. PMID: 29350680. DOI: 10.1038/emm.2017.239
33 Li Y Zheng Y Izumi K Ishiguro H Ye B Li F & Miyamoto H Androgen activates β-catenin signaling in bladder cancer cells. Endocr Relat Cancer. 20(3) 293 - 304 2013. PMID: 23447569. DOI: 10.1530/ERC-12-0328
34 Yang X Wang X Nie F Liu T Yu X Wang H Li Q Peng R Mao Z Zhou Q & Li G miR-135 family members mediate podocyte injury through the activation of Wnt/β-catenin signaling. Int J Mol Med. 36(3) 669 - 677 2015. PMID: 26134897. DOI: 10.3892/ijmm.2015.2259
35 Naito S Bilim V Yuuki K Ugolkov A Motoyama T Nagaoka A Kato T & Tomita Y Glycogen synthase kinase-3beta: a prognostic marker and a potential therapeutic target in human bladder cancer. Clin Cancer Res. 16(21) 5124 - 5132 2010. PMID: 20889919. DOI: 10.1158/1078-0432.CCR-10-0275
36 Tan M Mu X Liu Z Tao L Wang J Ge J & Qiu J microRNA-495 promotes bladder cancer cell growth and invasion by targeting phosphatase and tensin homolog. Biochem Biophys Res Commun. 483(2) 867 - 873 2017. PMID: 28069380. DOI: 10.1016/j.bbrc.2017.01.019
37 Álvarez-Garcia V Tawil Y Wise HM & Leslie NR Mechanisms of PTEN loss in cancer: It’s all about diversity. Semin Cancer Biol. 59 66 - 79 2019. PMID: 30738865. DOI: 10.1016/j.semcancer.2019.02.001
38 Worby CA & Dixon JE PTEN. Annu Rev Biochem. 83 641 - 669 2014. PMID: 24905788. DOI: 10.1146/annurev-biochem-082411-113907
39 Lee YR Chen M & Pandolfi PP The functions and regulation of the PTEN tumour suppressor: new modes and prospects. Nat Rev Mol Cell Biol. 19(9) 547 - 562 2018. PMID: 29858604. DOI: 10.1038/s41580-018-0015-0
40 Ashrafizadeh M Zarrabi A Samarghandian S & Najafi M PTEN: What we know of the function and regulation of this onco-suppressor factor in bladder cancer. Eur J Pharmacol. 881 173226 2020. PMID: 32485246. DOI: 10.1016/j.ejphar.2020.173226
41 Houédé N & Pourquier P Targeting the genetic alterations of the PI3K-AKT-mTOR pathway: its potential use in the treatment of bladder cancers. Pharmacol Ther. 145 1 - 18 2015. PMID: 24929024. DOI: 10.1016/j.pharmthera.2014.06.004
42 Jin H Ma J Xu J Li H Chang Y Zang N Tian Z Wang X Zhao N Liu L Chen C Xie Q Lu Y Fang Z Huang X Huang C & Huang H Oncogenic role of MIR516A in human bladder cancer was mediated by its attenuating PHLPP2 expression and BECN1-dependent autophagy. Autophagy. 17(4) 840 - 854 2021. PMID: 32116109. DOI: 10.1080/15548627.2020.1733262
43 Brognard J Sierecki E Gao T & Newton A PHLPP and a second isoform, PHLPP2, differentially attenuate the amplitude of Akt signaling by regulating distinct Akt isoforms. Molecular Cell. 25(6) 917 - 931 2019. DOI: 10.1016/j.molcel.2007.02.017
44 Gao T Brognard J & Newton AC The phosphatase PHLPP controls the cellular levels of protein kinase C. J Biol Chem. 283(10) 6300 - 6311 2008. PMID: 18162466. DOI: 10.1074/jbc.M707319200
45 Levy JMM Towers CG & Thorburn A Targeting autophagy in cancer. Nat Rev Cancer. 17(9) 528 - 542 2017. PMID: 28751651. DOI: 10.1038/nrc.2017.53
46 Onorati AV Dyczynski M Ojha R & Amaravadi RK Targeting autophagy in cancer. Cancer. 124(16) 3307 - 3318 2018. PMID: 29671878. DOI: 10.1002/cncr.31335
47 Lv L Deng H Li Y Zhang C Liu X Liu Q Zhang D Wang L Pu Y Zhang H He Y Wang Y Yu Y Yu T & Zhu J The DNA methylation-regulated miR-193a-3p dictates the multi-chemoresistance of bladder cancer via repression of SRSF2/PLAU/HIC2 expression. Cell Death Dis. 5 e1402 2014. PMID: 25188512. DOI: 10.1038/cddis.2014.367
48 Cazalla D Zhu J Manche L Huber E Krainer AR & Cáceres JF Nuclear export and retention signals in the RS domain of SR proteins. Mol Cell Biol. 22(19) 6871 - 6882 2002. PMID: 12215544. DOI: 10.1128/MCB.22.19.6871-6882.2002
49 Alfano D Iaccarino I & Stoppelli MP Urokinase signaling through its receptor protects against anoikis by increasing BCL-xL expression levels. J Biol Chem. 281(26) 17758 - 17767 2006. PMID: 16632475. DOI: 10.1074/jbc.M601812200
50 Deltour S Pinte S Guérardel C & Leprince D Characterization of HRG22, a human homologue of the putative tumor suppressor gene HIC1. Biochem Biophys Res Commun. 287(2) 427 - 434 2001. PMID: 11554746. DOI: 10.1006/bbrc.2001.5624
51 Deng H Lv L Li Y Zhang C Meng F Pu Y Xiao J Qian L Zhao W Liu Q Zhang D Wang Y Zhang H He Y & Zhu J miR-193a-3p regulates the multi-drug resistance of bladder cancer by targeting the LOXL4 gene and the oxidative stress pathway. Mol Cancer. 13 234 2014. PMID: 25311867. DOI: 10.1186/1476-4598-13-234
52 Ye M Song Y Pan S Chu M Wang ZW & Zhu X Evolving roles of lysyl oxidase family in tumorigenesis and cancer therapy. Pharmacol Ther. 215 107633 2020. PMID: 32693113. DOI: 10.1016/j.pharmthera.2020.107633
53 Li T Wu C Gao L Qin F Wei Q & Yuan J Lysyl oxidase family members in urological tumorigenesis and fibrosis. Oncotarget. 9(28) 20156 - 20164 2018. PMID: 29732010. DOI: 10.18632/oncotarget.24948
54 Lv L Li Y Deng H Zhang C Pu Y Qian L Xiao J Zhao W Liu Q Zhang D Wang Y Zhang H He Y & Zhu J MiR-193a-3p promotes the multi-chemoresistance of bladder cancer by targeting the HOXC9 gene. Cancer Lett. 357(1) 105 - 113 2015. PMID: 25444900. DOI: 10.1016/j.canlet.2014.11.002
55 Hur H Lee JY Yang S Kim JM Park AE & Kim MH HOXC9 induces phenotypic switching between proliferation and invasion in breast cancer cells. J Cancer. 7(7) 768 - 773 2016. PMID: 27162534. DOI: 10.7150/jca.13894
56 Jiang G Huang C Li J Huang H Jin H Zhu J Wu XR & Huang C Role of STAT3 and FOXO1 in the divergent therapeutic responses of non-metastatic and metastatic bladder cancer cells to miR-145. Mol Cancer Ther. 16(5) 924 - 935 2017. PMID: 28223425. DOI: 10.1158/1535-7163.MCT-16-0631
57 Monsalve M & Olmos Y The complex biology of FOXO. Curr Drug Targets. 12(9) 1322 - 1350 2011. PMID: 21443460. DOI: 10.2174/138945011796150307
58 Johnson DE O’Keefe RA & Grandis JR Targeting the IL-6/JAK/STAT3 signalling axis in cancer. Nat Rev Clin Oncol. 15(4) 234 - 248 2018. PMID: 29405201. DOI: 10.1038/nrclinonc.2018.8
59 Furtek SL Backos DS Matheson CJ & Reigan P Strategies and approaches of targeting STAT3 for cancer treatment. ACS Chem Biol. 11(2) 308 - 318 2016. PMID: 26730496. DOI: 10.1021/acschembio.5b00945
60 Tan Y Zhang T Zhou L Liu S & Liang C MiR-34b-3p represses the multidrug-chemoresistance of bladder cancer cells by regulating the CCND2 and P2RY1 genes. Med Sci Monit. 25 1323 - 1335 2019. PMID: 30778022. DOI: 10.12659/MSM.913746
61 Hu W Liu Q Pan J & Sui Z MiR-373-3p enhances the chemosensitivity of gemcitabine through cell cycle pathway by targeting CCND2 in pancreatic carcinoma cells. Biomed Pharmacother. 105 887 - 898 2018. PMID: 30021382. DOI: 10.1016/j.biopha.2018.05.091
62 Yanachkov IB Chang H Yanachkova MI Dix EJ Berny-Lang MA Gremmel T Michelson AD Wright GE & Frelinger AL 3rd New highly active antiplatelet agents with dual specificity for platelet P2Y1 and P2Y12 adenosine diphosphate receptors. Eur J Med Chem. 107 204 - 218 2016. PMID: 26588064. DOI: 10.1016/j.ejmech.2015.10.055
63 Gremmel T Yanachkov IB Yanachkova MI Wright GE Wider J Undyala VV Michelson AD Frelinger AL 3rd & Przyklenk K Synergistic inhibition of both P2Y1 and P2Y12 adenosine diphosphate receptors as novel approach to rapidly attenuate platelet-mediated thrombosis. Arterioscler Thromb Vasc Biol. 36(3) 501 - 509 2016. PMID: 26743169. DOI: 10.1161/ATVBAHA.115.306885
64 Zhang T Wang J Zhai X Li H Li C & Chang J MiR-124 retards bladder cancer growth by directly targeting CDK4. Acta Biochim Biophys Sin (Shanghai). 46(12) 1072 - 1079 2014. PMID: 25348738. DOI: 10.1093/abbs/gmu105
65 Sathe A Koshy N Schmid SC Thalgott M Schwarzenböck SM Krause BJ Holm PS Gschwend JE Retz M & Nawroth R CDK4/6 inhibition controls proliferation of bladder cancer and transcription of RB1. J Urol. 195(3) 771 - 779 2016. PMID: 26318986. DOI: 10.1016/j.juro.2015.08.082
66 Carneiro BA Meeks JJ Kuzel TM Scaranti M Abdulkadir SA & Giles FJ Emerging therapeutic targets in bladder cancer. Cancer Treat Rev. 41(2) 170 - 178 2015. PMID: 25498841. DOI: 10.1016/j.ctrv.2014.11.003
67 Yuan Q Sun T Ye F Kong W & Jin H MicroRNA-124-3p affects proliferation, migration and apoptosis of bladder cancer cells through targeting AURKA. Cancer Biomark. 19(1) 93 - 101 2017. PMID: 28269755. DOI: 10.3233/CBM-160427
68 Yan M Wang C He B Yang M Tong M Long Z Liu B Peng F Xu L Zhang Y Liang D Lei H Subrata S Kelley KW Lam EW Jin B & Liu Q Aurora-A kinase: A potent oncogene and target for cancer therapy. Med Res Rev. 36(6) 1036 - 1079 2016. PMID: 27406026. DOI: 10.1002/med.21399
69 Guo M Lu S Huang H Wang Y Yang MQ Yang Y Fan Z Jiang B & Deng Y Increased AURKA promotes cell proliferation and predicts poor prognosis in bladder cancer. BMC Syst Biol. 12(Suppl 7) 118 2018. PMID: 30547784. DOI: 10.1186/s12918-018-0634-2
70 Mobley A Zhang S Bondaruk J Wang Y Majewski T Caraway NP Huang L Shoshan E Velazquez-Torres G Nitti G Lee S Lee JG Fuentes-Mattei E Willis D Zhang L Guo CC Yao H Baggerly K Lotan Y Lerner SP Dinney C McConkey D Bar-Eli M & Czerniak B Aurora kinase A is a biomarker for bladder cancer detection and contributes to its aggressive behavior. Sci Rep. 7 40714 2017. PMID: 28102366. DOI: 10.1038/srep40714
71 de Martino M Shariat SF Hofbauer SL Lucca I Taus C Wiener HG Haitel A Susani M & Klatte T Aurora A Kinase as a diagnostic urinary marker for urothelial bladder cancer. World J Urol. 33(1) 105 - 110 2015. PMID: 24562316. DOI: 10.1007/s00345-014-1267-8
72 Tayyar Y Jubair L Fallaha S & McMillan NAJ Critical risk-benefit assessment of the novel anti-cancer aurora a kinase inhibitor alisertib (MLN8237): A comprehensive review of the clinical data. Crit Rev Oncol Hematol. 119 59 - 65 2017. PMID: 29065986. DOI: 10.1016/j.critrevonc.2017.09.006
73 O’Connor OA Özcan M Jacobsen ED Roncero JM Trotman J Demeter J Masszi T Pereira J Ramchandren R Beaven A Caballero D Horwitz SM Lennard A Turgut M Hamerschlak N d’Amore FA Foss F Kim WS Leonard JP Zinzani PL Chiattone CS Hsi ED Trümper L Liu H Sheldon-Waniga E Ullmann CD Venkatakrishnan K Leonard EJ Shustov AR & Lumiere Study Investigators Randomized phase III study of alisertib or investigator’s choice (selected single agent) in patients with relapsed or refractory peripheral T-cell lymphoma. J Clin Oncol. 37(8) 613 - 623 2019. PMID: 30707661. DOI: 10.1200/JCO.18.00899
74 Necchi A Lo Vullo S Mariani L Raggi D Giannatempo P Calareso G Togliardi E Crippa F Di Genova N Perrone F Colecchia M Paolini B Pelosi G Nicolai N Procopio G Salvioni R & De Braud FG An open-label, single-arm, phase 2 study of the Aurora kinase A inhibitor alisertib in patients with advanced urothelial cancer. Invest New Drugs. 34(2) 236 - 242 2016. PMID: 26873642. DOI: 10.1007/s10637-016-0328-9
75 Xiang W Wu X Huang C Wang M Zhao X Luo G Li Y Jiang G Xiao X & Zeng F PTTG1 regulated by miR-146a-3p promotes bladder cancer migration, invasion, metastasis and growth. Oncotarget. 8(1) 664 - 678 2017. PMID: 27893422. DOI: 10.18632/oncotarget.13507
76 Yang K Zheng XY Qin J Wang YB Bai Y Mao QQ Wan Q Wu ZM & Xie LP Up-regulation of p21WAF1/Cip1 by saRNA induces G1-phase arrest and apoptosis in T24 human bladder cancer cells. Cancer Lett. 265(2) 206 - 214 2008. PMID: 18358601. DOI: 10.1016/j.canlet.2008.02.014
77 Chesnokova V Zonis S Kovacs K Ben-Shlomo A Wawrowsky K Bannykh S & Melmed S p21(Cip1) restrains pituitary tumor growth. Proc Natl Acad Sci U S A. 105(45) 17498 - 17503 2008. PMID: 18981426. DOI: 10.1073/pnas.0804810105
78 Vlotides G Eigler T & Melmed S Pituitary tumor-transforming gene: physiology and implications for tumorigenesis. Endocr Rev. 28(2) 165 - 186 2007. PMID: 17325339. DOI: 10.1210/er.2006-0042
79 Ge Q Wang C Chen Z Li F Hu J & Ye Z The suppressive effects of miR-1180-5p on the proliferation and tumorigenicity of bladder cancer cells. Histol Histopathol. 32(1) 77 - 86 2017. PMID: 27112784. DOI: 10.14670/HH-11-772
80 Place RF Li LC Pookot D Noonan EJ & Dahiya R MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc Natl Acad Sci U S A. 105(5) 1608 - 1613 2008. PMID: 18227514. DOI: 10.1073/pnas.0707594105
81 Huang V Place RF Portnoy V Wang J Qi Z Jia Z Yu A Shuman M Yu J & Li LC Upregulation of Cyclin B1 by miRNA and its implications in cancer. Nucleic Acids Res. 40(4) 1695 - 1707 2012. PMID: 22053081. DOI: 10.1093/nar/gkr934
82 Wang C Chen Z Ge Q Hu J Li F Hu J Xu H Ye Z & Li LC Up-regulation of p21(WAF1/CIP1) by miRNAs and its implications in bladder cancer cells. FEBS Lett. 588(24) 4654 - 4664 2014. PMID: 25447520. DOI: 10.1016/j.febslet.2014.10.037
83 Parry D Mahony D Wills K & Lees E Cyclin D-CDK subunit arrangement is dependent on the availability of competing INK4 and p21 class inhibitors. Mol Cell Biol. 19(3) 1775 - 1783 1999. PMID: 10022865. DOI: 10.1128/MCB.19.3.1775
84 Choo JR & Lee SC CDK4-6 inhibitors in breast cancer: current status and future development. Expert Opin Drug Metab Toxicol. 14(11) 1123 - 1138 2018. PMID: 30360668. DOI: 10.1080/17425255.2018.1541347
85 Rubio C Martínez-Fernández M Segovia C Lodewijk I Suarez-Cabrera C Segrelles C López-Calderón F Munera-Maravilla E Santos M Bernardini A García-Escudero R Lorz C Gómez-Rodriguez MJ de Velasco G Otero I Villacampa F Guerrero-Ramos F Ruiz S de la Rosa F Domínguez-Rodríguez S Real FX Malats N Castellano D Dueñas M & Paramio JM CDK4/6 inhibitor as a novel therapeutic approach for advanced bladder cancer independently of RB1 status. Clin Cancer Res. 25(1) 390 - 402 2019. PMID: 30242024. DOI: 10.1158/1078-0432.CCR-18-0685
86 Zhang Q Miao S Han X Li C Zhang M Cui K Xiong T Chen Z Wang C & Xu H MicroRNA-3619-5p suppresses bladder carcinoma progression by directly targeting β-catenin and CDK2 and activating p21. Cell Death Dis. 9(10) 960 2018. PMID: 30237499. DOI: 10.1038/s41419-018-0986-y
87 Tang K Wang C Chen Z Xu H & Ye Z Clinicopathologic and prognostic significance of p21 (Cip1/Waf1) expression in bladder cancer. Int J Clin Exp Pathol. 8(5) 4999 - 5007 2015. PMID: 26191193.
Pubmed |
88 Garg M & Maurya N WNT/β-catenin signaling in urothelial carcinoma of bladder. World J Nephrol. 8(5) 83 - 94 2019. PMID: 31624709. DOI: 10.5527/wjn.v8.i5.83
89 Jung JH You S Oh JW Yoon J Yeon A Shahid M Cho E Sairam V Park TD Kim KP & Kim J Integrated proteomic and phosphoproteomic analyses of cisplatin-sensitive and resistant bladder cancer cells reveal CDK2 network as a key therapeutic target. Cancer Lett. 437 1 - 12 2018. PMID: 30145203. DOI: 10.1016/j.canlet.2018.08.014
90 Xu T Qin L Zhu Z Wang X Liu Y Fan Y Zhong S Wang X Zhang X Xia L Zhang X Xu C & Shen Z MicroRNA-31 functions as a tumor suppressor and increases sensitivity to mitomycin-C in urothelial bladder cancer by targeting integrin α5. Oncotarget. 7(19) 27445 - 27457 2016. PMID: 27050274. DOI: 10.18632/oncotarget.8479
91 Mierke CT Frey B Fellner M Herrmann M & Fabry B Integrin α5β1 facilitates cancer cell invasion through enhanced contractile forces. J Cell Sci. 124(Pt 3) 369 - 383 2011. PMID: 21224397. DOI: 10.1242/jcs.071985
92 Yan T & Ye XX MicroRNA-328-3p inhibits the tumorigenesis of bladder cancer through targeting ITGA5 and inactivating PI3K/AKT pathway. Eur Rev Med Pharmacol Sci. 23(12) 5139 - 5148 2019. PMID: 31298367. DOI: 10.26355/eurrev_201906_18178
93 Saito T Kimura M Kawasaki T Sato S & Tomita Y Correlation between integrin alpha 5 expression and the malignant phenotype of transitional cell carcinoma. Br J Cancer. 73(3) 327 - 331 1996. PMID: 8562338. DOI: 10.1038/bjc.1996.57
94 Wang Y Xing QF Liu XQ Guo ZJ Li CY & Sun G MiR-122 targets VEGFC in bladder cancer to inhibit tumor growth and angiogenesis. Am J Transl Res. 8(7) 3056 - 3066 2016. PMID: 27508026.
Pubmed |
95 Chen JC Chang YW Hong CC Yu YH & Su JL The role of the VEGF-C/VEGFRs axis in tumor progression and therapy. Int J Mol Sci. 14(1) 88 - 107 2012. PMID: 23344023. DOI: 10.3390/ijms14010088
96 Karaman S & Detmar M Mechanisms of lymphatic metastasis. J Clin Invest. 124(3) 922 - 928 2014. PMID: 24590277. DOI: 10.1172/JCI71606
97 Miyata Y Kanda S Ohba K Nomata K Hayashida Y Eguchi J Hayashi T & Kanetake H Lymphangiogenesis and angiogenesis in bladder cancer: prognostic implications and regulation by vascular endothelial growth factors-A, -C, and -D. Clin Cancer Res. 12(3 Pt 1) 800 - 806 2006. PMID: 16467091. DOI: 10.1158/1078-0432.CCR-05-1284
98 Zhang L Xu J Yang G Li H & Guo X miR-202 inhibits cell proliferation, migration, and invasion by targeting epidermal growth factor receptor in human bladder cancer. Oncol Res. 26(6) 949 - 957 2018. PMID: 29298735. DOI: 10.3727/096504018X15149787144385
99 Arfaoui AT Mejri S Belhaj R Karkni W Chebil M & Rammeh S Prognostic value of immunohistochemical expression profile of epidermal growth factor receptor in urothelial bladder cancer. J Immunoassay Immunochem. 37(4) 359 - 367 2016. PMID: 26919632. DOI: 10.1080/15321819.2016.1146757
100 Di Maida F Mari A Scalici Gesolfo C Cangemi A Allegro R Sforza S Cocci A Tellini R Masieri L Russo A Carini M Minervini A & Serretta V Epidermal growth factor receptor (EGFR) cell expression during adjuvant treatment after transurethral resection for non-muscle-invasive bladder cancer: A new potential tool to identify patients at higher risk of disease progression. Clin Genitourin Cancer. 17(4) e751 - e758 2019. PMID: 31126772. DOI: 10.1016/j.clgc.2019.04.008
101 Arteaga CL & Engelman JA ERBB receptors: from oncogene discovery to basic science to mechanism-based cancer therapeutics. Cancer Cell. 25(3) 282 - 303 2014. PMID: 24651011. DOI: 10.1016/j.ccr.2014.02.025
102 Sjödahl G Jackson CL Bartlett JM Siemens DR & Berman DM Molecular profiling in muscle-invasive bladder cancer: more than the sum of its parts. J Pathol. 247(5) 563 - 573 2019. PMID: 30604486. DOI: 10.1002/path.5230
103 Yang X Shi L Yi C Yang Y Chang L & Song D MiR-210-3p inhibits the tumor growth and metastasis of bladder cancer via targeting fibroblast growth factor receptor-like 1. Am J Cancer Res. 7(8) 1738 - 1753 2017. PMID: 28861329.
Pubmed |
104 Katoh M & Nakagama H FGF receptors: cancer biology and therapeutics. Med Res Rev. 34(2) 280 - 300 2014. PMID: 23696246. DOI: 10.1002/med.21288
105 Roskoski R Jr The role of fibroblast growth factor receptor (FGFR) protein-tyrosine kinase inhibitors in the treatment of cancers including those of the urinary bladder. Pharmacol Res. 151 104567 2020. PMID: 31770593. DOI: 10.1016/j.phrs.2019.104567
106 De Keukeleire S De Maeseneer D Jacobs C & Rottey S Targeting FGFR in bladder cancer: ready for clinical practice. Acta Clin Belg. 75(1) 49 - 56 2020. PMID: 31671027. DOI: 10.1080/17843286.2019.1685738
107 Casadei C Dizman N Schepisi G Cursano MC Basso U Santini D Pal SK & De Giorgi U Targeted therapies for advanced bladder cancer: new strategies with FGFR inhibitors. Ther Adv Med Oncol. 11 1758835919890285 2019. PMID: 31803255. DOI: 10.1177/1758835919890285
108 Trueb B Biology of FGFRL1, the fifth fibroblast growth factor receptor. Cellular and Molecular Life Sciences. 68(6) 951 - 964 2019. DOI: 10.1007/s00018-010-0576-3
109 Schild C & Trueb B Aberrant expression of FGFRL1, a novel FGF receptor, in ovarian tumors. Int J Mol Med. 16(6) 1169 - 1173 2005. PMID: 16273302.
Pubmed |
110 Xu C Zeng Q Xu W Jiao L Chen Y Zhang Z Wu C Jin T Pan A Wei R Yang B & Sun Y miRNA-100 inhibits human bladder urothelial carcinogenesis by directly targeting mTOR. Mol Cancer Ther. 12(2) 207 - 219 2013. PMID: 23270926. DOI: 10.1158/1535-7163.MCT-12-0273
111 Guertin DA & Sabatini DM Defining the role of mTOR in cancer. Cancer Cell. 12(1) 9 - 22 2007. PMID: 17613433. DOI: 10.1016/j.ccr.2007.05.008
112 Castedo M Ferri KF & Kroemer G Mammalian target of rapamycin (mTOR): pro- and anti-apoptotic. Cell Death Differ. 9(2) 99 - 100 2002. PMID: 11840159. DOI: 10.1038/sj.cdd.4400978
113 Pinto-Leite R Arantes-Rodrigues R Sousa N Oliveira PA & Santos L mTOR inhibitors in urinary bladder cancer. Tumour Biol. 37(9) 11541 - 11551 2016. PMID: 27235118. DOI: 10.1007/s13277-016-5083-1
114 Sathe A & Nawroth R Targeting the PI3K/AKT/mTOR pathway in bladder cancer. Methods Mol Biol. 1655 335 - 350 2018. PMID: 28889395. DOI: 10.1007/978-1-4939-7234-0_23
115 Liang Z Wang X Xu X Xie B Ji A Meng S Li S Zhu Y Wu J Hu Z Lin Y Zheng X Xie L & Liu B MicroRNA-608 inhibits proliferation of bladder cancer via AKT/FOXO3a signaling pathway. Mol Cancer. 16(1) 96 2017. PMID: 28549468. DOI: 10.1186/s12943-017-0664-1
116 Schmidt M Fernandez de Mattos S van der Horst A Klompmaker R Kops GJ Lam EW Burgering BM & Medema RH Cell cycle inhibition by FoxO forkhead transcription factors involves downregulation of cyclin D. Mol Cell Biol. 22(22) 7842 - 7852 2002. PMID: 12391153. DOI: 10.1128/MCB.22.22.7842-7852.2002
117 Huang H & Tindall DJ Dynamic FoxO transcription factors. J Cell Sci. 120(Pt 15) 2479 - 2487 2007. PMID: 17646672. DOI: 10.1242/jcs.001222
118 Langhorst MF Reuter A & Stuermer CA Scaffolding microdomains and beyond: the function of reggie/flotillin proteins. Cell Mol Life Sci. 62(19-20) 2228 - 2240 2005. PMID: 16091845. DOI: 10.1007/s00018-005-5166-4
119 Bickel PE Scherer PE Schnitzer JE Oh P Lisanti MP & Lodish HF Flotillin and epidermal surface antigen define a new family of caveolae-associated integral membrane proteins. J Biol Chem. 272(21) 13793 - 13802 1997. PMID: 9153235. DOI: 10.1074/jbc.272.21.13793
120 Guan Y Song H Zhang G & Ai X Overexpression of flotillin-1 is involved in proliferation and recurrence of bladder transitional cell carcinoma. Oncol Rep. 32(2) 748 - 754 2014. PMID: 24890092. DOI: 10.3892/or.2014.3221
121 Ge Q Lu M Ju L Qian K Wang G Wu CL Liu X Xiao Y & Wang X miR-4324-RACGAP1-STAT3-ESR1 feedback loop inhibits proliferation and metastasis of bladder cancer. Int J Cancer. 144(12) 3043 - 3055 2019. PMID: 30511377. DOI: 10.1002/ijc.32036
122 Hirose K Kawashima T Iwamoto I Nosaka T & Kitamura T MgcRacGAP is involved in cytokinesis through associating with mitotic spindle and midbody. J Biol Chem. 276(8) 5821 - 5828 2001. PMID: 11085985. DOI: 10.1074/jbc.M007252200
123 Kawashima T Bao YC Minoshima Y Nomura Y Hatori T Hori T Fukagawa T Fukada T Takahashi N Nosaka T Inoue M Sato T Kukimoto-Niino M Shirouzu M Yokoyama S & Kitamura T A Rac GTPase-activating protein, MgcRacGAP, is a nuclear localizing signal-containing nuclear chaperone in the activation of STAT transcription factors. Mol Cell Biol. 29(7) 1796 - 1813 2009. PMID: 19158271. DOI: 10.1128/MCB.01423-08
124 Vega FM & Ridley AJ Rho GTPases in cancer cell biology. FEBS Lett. 582(14) 2093 - 2101 2008. PMID: 18460342. DOI: 10.1016/j.febslet.2008.04.039
125 Ke HL Ke RH Li ST Li B Lu HT & Wang XQ Expression of RACGAP1 in high grade meningiomas: a potential role in cancer progression. J Neurooncol. 113(2) 327 - 332 2013. PMID: 23525949. DOI: 10.1007/s11060-013-1121-7
126 Yang R Liu M Liang H Guo S Guo X Yuan M Lian H Yan X Zhang S Chen X Fang F Guo H & Zhang C miR-138-5p contributes to cell proliferation and invasion by targeting Survivin in bladder cancer cells. Mol Cancer. 15(1) 82 2016. PMID: 27978829. DOI: 10.1186/s12943-016-0569-4
127 Altieri DC Targeting survivin in cancer. Cancer Lett. 332(2) 225 - 228 2013. PMID: 22410464. DOI: 10.1016/j.canlet.2012.03.005
128 Wheatley SP & Altieri DC Survivin at a glance. J Cell Sci. 132(7) jcs223826 2019. PMID: 30948431. DOI: 10.1242/jcs.223826
129 Zhang J Wang S Han F Li J Yu L Zhou P Chen Z Xue S Dai C & Li Q MicroRNA-542-3p suppresses cellular proliferation of bladder cancer cells through post-transcriptionally regulating survivin. Gene. 579(2) 146 - 152 2018. DOI: 10.1016/j.gene.2015.12.048
130 Jeon C Kim M Kwak C Kim HH & Ku JH Prognostic role of survivin in bladder cancer: a systematic review and meta-analysis. PLoS One. 8(10) e76719 2013. PMID: 24204662. DOI: 10.1371/journal.pone.0076719
131 Apollo A Ortenzi V Scatena C Zavaglia K Aretini P Lessi F Franceschi S Tomei S Sepich CA Viacava P Mazzanti CM & Naccarato AG Molecular characterization of low grade and high grade bladder cancer. PLoS One. 14(1) e0210635 2019. PMID: 30650148. DOI: 10.1371/journal.pone.0210635
132 Martínez-García D Manero-Rupérez N Quesada R Korrodi-Gregório L & Soto-Cerrato V Therapeutic strategies involving survivin inhibition in cancer. Med Res Rev. 39(3) 887 - 909 2019. PMID: 30421440. DOI: 10.1002/med.21547
133 Jin H Xue L Mo L Zhang D Guo X Xu J Li J Peng M Zhao X Zhong M Xu D Wu XR Huang H & Huang C Downregulation of miR-200c stabilizes XIAP mRNA and contributes to invasion and lung metastasis of bladder cancer. Cell Adh Migr. 13(1) 236 - 248 2019. PMID: 31240993. DOI: 10.1080/19336918.2019.1633851
134 Deveraux QL & Reed JC IAP family proteins--suppressors of apoptosis. Genes Dev. 13(3) 239 - 252 1999. PMID: 9990849. DOI: 10.1101/gad.13.3.239
135 Cossu F Milani M Mastrangelo E & Lecis D Targeting the BIR domains of inhibitor of apoptosis (IAP) proteins in cancer treatment. Comput Struct Biotechnol J. 17 142 - 150 2019. PMID: 30766663. DOI: 10.1016/j.csbj.2019.01.009
136 Fulda S Smac mimetics to therapeutically target IAP proteins in cancer. Int Rev Cell Mol Biol. 330 157 - 169 2017. PMID: 28215531. DOI: 10.1016/bs.ircmb.2016.09.004
137 Fu X Pang X Qi H Chen S Li Y & Tan W XIAP inhibitor Embelin inhibits bladder cancer survival and invasion in vitro. Clin Transl Oncol. 18(3) 277 - 282 2016. PMID: 26209051. DOI: 10.1007/s12094-015-1363-2
138 Yu Y Jin H Xu J Gu J Li X Xie Q Huang H Li J Tian Z Jiang G Chen C He F Wu XR & Huang C XIAP overexpression promotes bladder cancer invasion in vitro and lung metastasis in vivo via enhancing nucleolin-mediated Rho-GDIβ mRNA stability. Int J Cancer. 142(10) 2040 - 2055 2018. PMID: 29250796. DOI: 10.1002/ijc.31223
139 Li M Song T Yin ZF & Na YQ XIAP as a prognostic marker of early recurrence of nonmuscular invasive bladder cancer. Chin Med J (Engl). 120(6) 469 - 473 2007. PMID: 17439739.
Pubmed |
140 Mita MM LoRusso PM Papadopoulos KP Gordon MS Mita AC Ferraldeschi R Keer H Oganesian A Su XY Jueliger S & Tolcher AW A phase I study of ASTX660, an antagonist of inhibitors of apoptosis proteins, in adults with advanced cancers or lymphoma. Clin Cancer Res. 26(12) 2819 - 2826 2020. PMID: 31900279. DOI: 10.1158/1078-0432.CCR-19-1430
141 Zhang J Mao S Wang L Zhang W Zhang Z Guo Y Wu Y Yi F & Yao X MicroRNA 154 functions as a tumor suppressor in bladder cancer by directly targeting ATG7. Oncol Rep. 41(2) 819 - 828 2019. PMID: 30483807. DOI: 10.3892/or.2018.6879
142 Kimmelman AC & White E Autophagy and tumor metabolism. Cell Metab. 25(5) 1037 - 1043 2017. PMID: 28467923. DOI: 10.1016/j.cmet.2017.04.004
143 Chen N & Karantza V Autophagy as a therapeutic target in cancer. Cancer Biol Ther. 11(2) 157 - 168 2011. PMID: 21228626. DOI: 10.4161/cbt.11.2.14622
144 Lin YC Lin JF Wen SI Yang SC Tsai TF Chen HE Chou KY & Hwang TI Inhibition of high basal level of autophagy induces apoptosis in human bladder cancer cells. J Urol. 195(4 Pt 1) 1126 - 1135 2016. PMID: 26519656. DOI: 10.1016/j.juro.2015.10.128
145 Tong H Yin H Hossain MA Wang Y Wu F Dong X Gao S Zhan K & He W Starvation-induced autophagy promotes the invasion and migration of human bladder cancer cells via TGF-β1/Smad3-mediated epithelial-mesenchymal transition activation. J Cell Biochem. 120(4) 5118 - 5127 2019. PMID: 30320898. DOI: 10.1002/jcb.27788
146 Li F Guo H Yang Y Feng M Liu B Ren X & Zhou H Autophagy modulation in bladder cancer development and treatment (Review). Oncol Rep. 42(5) 1647 - 1655 2019. PMID: 31436298. DOI: 10.3892/or.2019.7286
147 Zhang L Wang CZ Ma M & Shao GF MiR-15 suppressed the progression of bladder cancer by targeting BMI1 oncogene via PI3K/AKT signaling pathway. Eur Rev Med Pharmacol Sci. 23(20) 8813 - 8822 2019. PMID: 31696468. DOI: 10.26355/eurrev_201910_19276
148 Wang MC Li CL Cui J Jiao M Wu T Jing LI & Nan KJ BMI-1, a promising therapeutic target for human cancer. Oncol Lett. 10(2) 583 - 588 2015. PMID: 26622537. DOI: 10.3892/ol.2015.3361
149 Liu L Qiu M Tan G Liang Z Qin Y Chen L Chen H & Liu J miR-200c inhibits invasion, migration and proliferation of bladder cancer cells through down-regulation of BMI-1 and E2F3. J Transl Med. 12 305 2014. PMID: 25367080. DOI: 10.1186/s12967-014-0305-z
150 Cheng Y Yang X Deng X Zhang X Li P Tao J & Lu Q MicroRNA-218 inhibits bladder cancer cell proliferation, migration, and invasion by targeting BMI-1. Tumour Biol. 36(10) 8015 - 8023 2015. PMID: 25967457. DOI: 10.1007/s13277-015-3532-x
151 Qin ZK Yang JA Ye YL Zhang X Xu LH Zhou FJ Han H Liu ZW Song LB & Zeng MS Expression of Bmi-1 is a prognostic marker in bladder cancer. BMC Cancer. 9 61 2009. PMID: 19228380. DOI: 10.1186/1471-2407-9-61
152 Wang Y Ma DL Yu CH Sha KF Zhao MJ & Liu TJ MicroRNA-370 suppresses SOX12 transcription and acts as a tumor suppressor in bladder cancer. Eur Rev Med Pharmacol Sci. 24(5) 2303 - 2312 2020. PMID: 32196581. DOI: 10.26355/eurrev_202003_20496
153 Dy P Penzo-Méndez A Wang H Pedraza CE Macklin WB & Lefebvre V The three SoxC proteins--Sox4, Sox11 and Sox12--exhibit overlapping expression patterns and molecular properties. Nucleic Acids Res. 36(9) 3101 - 3117 2008. PMID: 18403418. DOI: 10.1093/nar/gkn162
154 Du F Chen J Liu H Cai Y Cao T Han W Yi X Qian M Tian D Nie Y Wu K Fan D & Xia L SOX12 promotes colorectal cancer cell proliferation and metastasis by regulating asparagine synthesis. Cell Death Dis. 10(3) 239 2019. PMID: 30858360. DOI: 10.1038/s41419-019-1481-9
155 Du F Feng W Chen S Wu S Cao T Yuan T Tian D Nie Y Wu K Fan D & Xia L Sex determining region Y-box 12 (SOX12) promotes gastric cancer metastasis by upregulating MMP7 and IGF1. Cancer Lett. 452 103 - 118 2019. PMID: 30922917. DOI: 10.1016/j.canlet.2019.03.035
156 Yuan P Meng L & Wang N SOX12 upregulation is associated with metastasis of hepatocellular carcinoma and increases CDK4 and IGF2BP1 expression. Eur Rev Med Pharmacol Sci. 21(17) 3821 - 3826 2017. PMID: 28975985.
Pubmed |
157 Wang X Wu Q Xu B Wang P Fan W Cai Y Gu X & Meng F MiR-124 exerts tumor suppressive functions on the cell proliferation, motility and angiogenesis of bladder cancer by fine-tuning UHRF1. FEBS J. 282(22) 4376 - 4388 2015. PMID: 26310391. DOI: 10.1111/febs.13502
158 Sidhu H & Capalash N UHRF1: The key regulator of epigenetics and molecular target for cancer therapeutics. Tumour Biol. 39(2) 1010428317692205 2017. PMID: 28218043. DOI: 10.1177/1010428317692205
159 Ying L Lin J Qiu F Cao M Chen H Liu Z & Huang Y Epigenetic repression of regulator of G-protein signaling 2 by ubiquitin-like with PHD and ring-finger domain 1 promotes bladder cancer progression. FEBS J. 282(1) 174 - 182 2015. PMID: 25323766. DOI: 10.1111/febs.13116
160 Saidi S Popov Z Janevska V & Panov S Overexpression of UHRF1 gene correlates with the major clinicopathological parameters in urinary bladder cancer. Int Braz J Urol. 43(2) 224 - 229 2017. PMID: 28128913. DOI: 10.1590/S1677-5538.IBJU.2016.0126
161 Jin H Sun W Zhang Y Yan H Liufu H Wang S Chen C Gu J Hua X Zhou L Jiang G Rao D Xie Q Huang H & Huang C MicroRNA-411 downregulation enhances tumor growth by upregulating MLLT11 expression in human bladder cancer. Mol Ther Nucleic Acids. 11 312 - 322 2018. PMID: 29858066. DOI: 10.1016/j.omtn.2018.03.003
162 Zhou J Ng Y & Chng WJ ENL: structure, function, and roles in hematopoiesis and acute myeloid leukemia. Cell Mol Life Sci. 75(21) 3931 - 3941 2018. PMID: 30066088. DOI: 10.1007/s00018-018-2895-8
163 Zhang J Wang L Mao S Liu M Zhang W Zhang Z Guo Y Huang B Yan Y Huang Y & Yao X miR-1-3p contributes to cell proliferation and invasion by targeting glutaminase in bladder cancer cells. Cell Physiol Biochem. 51(2) 513 - 527 2018. PMID: 30458442. DOI: 10.1159/000495273
164 Jang M Kim SS & Lee J Cancer cell metabolism: implications for therapeutic targets. Exp Mol Med. 45 e45 2013. PMID: 24091747. DOI: 10.1038/emm.2013.85
165 Masisi BK El Ansari R Alfarsi L Rakha EA Green AR & Craze ML The role of glutaminase in cancer. Histopathology. 76(4) 498 - 508 2020. PMID: 31596504. DOI: 10.1111/his.14014
166 Song M Kim SH Im CY & Hwang HJ Recent development of small molecule glutaminase inhibitors. Curr Top Med Chem. 18(6) 432 - 443 2018. PMID: 29793408. DOI: 10.2174/1568026618666180525100830
167 Takai T Yoshikawa Y Inamoto T Minami K Taniguchi K Sugito N Kuranaga Y Shinohara H Kumazaki M Tsujino T Takahara K Ito Y Akao Y & Azuma H A novel combination RNAi toward Warburg effect by replacement with miR-145 and silencing of PTBP1 induces apoptotic cell death in bladder cancer cells. Int J Mol Sci. 18(1) 179 2017. PMID: 28106737. DOI: 10.3390/ijms18010179
168 Vander Heiden MG Cantley LC & Thompson CB Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 324(5930) 1029 - 1033 2009. PMID: 19460998. DOI: 10.1126/science.1160809
169 Clower CV Chatterjee D Wang Z Cantley LC Vander Heiden MG & Krainer AR The alternative splicing repressors hnRNP A1/A2 and PTB influence pyruvate kinase isoform expression and cell metabolism. Proc Natl Acad Sci U S A. 107(5) 1894 - 1899 2010. PMID: 20133837. DOI: 10.1073/pnas.0914845107
170 Derouet MF Dakpo E Wu L Zehong G Conner J Keshavjee S de Perrot M Waddell T Elimova E Yeung J & Darling GE miR-145 expression enhances integrin expression in SK-GT-4 cell line by down-regulating c-Myc expression. Oncotarget. 9(20) 15198 - 15207 2018. PMID: 29632636. DOI: 10.18632/oncotarget.24613
171 Zhang W Mao S Shi D Zhang J Zhang Z Guo Y Wu Y Wang R Wang L Huang Y & Yao X MicroRNA-153 decreases tryptophan catabolism and inhibits angiogenesis in bladder cancer by targeting indoleamine 2,3-dioxygenase 1. Front Oncol. 9 619 2019. PMID: 31355138. DOI: 10.3389/fonc.2019.00619
172 Liu M Wang X Wang L Ma X Gong Z Zhang S & Li Y Targeting the IDO1 pathway in cancer: from bench to bedside. J Hematol Oncol. 11(1) 100 2018. PMID: 30068361. DOI: 10.1186/s13045-018-0644-y
173 Tsai YS Jou YC Tsai HT Cheong IS & Tzai TS Indoleamine-2,3-dioxygenase-1 expression predicts poorer survival and up-regulates ZEB2 expression in human early stage bladder cancer. Urol Oncol. 37(11) 810.e17 - 810.e27 2019. PMID: 31253481. DOI: 10.1016/j.urolonc.2019.05.005
174 Le Naour J Galluzzi L Zitvogel L Kroemer G & Vacchelli E Trial watch: IDO inhibitors in cancer therapy. Oncoimmunology. 9(1) 1777625 2020. PMID: 32934882. DOI: 10.1080/2162402X.2020.1777625
175 Long GV Dummer R Hamid O Gajewski TF Caglevic C Dalle S Arance A Carlino MS Grob JJ Kim TM Demidov L Robert C Larkin J Anderson JR Maleski J Jones M Diede SJ & Mitchell TC Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. Lancet Oncol. 20(8) 1083 - 1097 2019. PMID: 31221619. DOI: 10.1016/S1470-2045(19)30274-8
176 Liu M Chen Y Huang B Mao S Cai K Wang L & Yao X Tumor-suppressing effects of microRNA-612 in bladder cancer cells by targeting malic enzyme 1 expression. Int J Oncol. 52(6) 1923 - 1933 2018. PMID: 29620192. DOI: 10.3892/ijo.2018.4342
177 Murai S Ando A Ebara S Hirayama M Satomi Y & Hara T Inhibition of malic enzyme 1 disrupts cellular metabolism and leads to vulnerability in cancer cells in glucose-restricted conditions. Oncogenesis. 6(5) e329 2017. PMID: 28481367. DOI: 10.1038/oncsis.2017.34
178 Wen D Liu D Tang J Dong L Liu Y Tao Z Wan J Gao D Wang L Sun H Fan J & Wu W Malic enzyme 1 induces epithelial-mesenchymal transition and indicates poor prognosis in hepatocellular carcinoma. Tumour Biol. 36(8) 6211 - 6221 2015. PMID: 25753478. DOI: 10.1007/s13277-015-3306-5
179 Uchino K Takeshita F Takahashi RU Kosaka N Fujiwara K Naruoka H Sonoke S Yano J Sasaki H Nozawa S Yoshiike M Kitajima K Chikaraishi T & Ochiya T Therapeutic effects of microRNA-582-5p and -3p on the inhibition of bladder cancer progression. Mol Ther. 21(3) 610 - 619 2013. PMID: 23295946. DOI: 10.1038/mt.2012.269
180 Kusama T Mukai M Tatsuta M Matsumoto Y Nakamura H & Inoue M Selective inhibition of cancer cell invasion by a geranylgeranyltransferase-I inhibitor. Clin Exp Metastasis. 20(6) 561 - 567 2003. PMID: 14598891. DOI: 10.1023/a:1025898316728
181 Virtanen SS Sandholm J Yegutkin G Kalervo Väänänen H & Härkönen PL Inhibition of GGTase-I and FTase disrupts cytoskeletal organization of human PC-3 prostate cancer cells. Cell Biol Int. 34(8) 815 - 826 2010. PMID: 20446922. DOI: 10.1042/CBI20090288
182 Ohta E Kawakami F Kubo M & Obata F LRRK2 directly phosphorylates Akt1 as a possible physiological substrate: impairment of the kinase activity by Parkinson’s disease-associated mutations. FEBS Lett. 585(14) 2165 - 2170 2011. PMID: 21658387. DOI: 10.1016/j.febslet.2011.05.044
183 Wang L Cao XX Chen Q Zhu TF Zhu HG & Zheng L DIXDC1 targets p21 and cyclin D1 via PI3K pathway activation to promote colon cancer cell proliferation. Cancer Sci. 100(10) 1801 - 1808 2009. PMID: 19572978. DOI: 10.1111/j.1349-7006.2009.01246.x
184 Wang JS Wang FB Zhang QG Shen ZZ & Shao ZM Enhanced expression of Rab27A gene by breast cancer cells promoting invasiveness and the metastasis potential by secretion of insulin-like growth factor-II. Mol Cancer Res. 6(3) 372 - 382 2008. PMID: 18337447. DOI: 10.1158/1541-7786.MCR-07-0162
185 Liu J Gong X Zhu X Xue D Liu Y & Wang P Rab27A overexpression promotes bladder cancer proliferation and chemoresistance through regulation of NF-ĸB signaling. Oncotarget. 8(43) 75272 - 75283 2017. PMID: 29088864. DOI: 10.18632/oncotarget.20775
186 Ying Y Li J Xie H Yan H Jin K He L Ma X Wu J Xu X Fang J Wang X Zheng X Liu B & Xie L CCND1, NOP14 and DNMT3B are involved in miR-502-5p-mediated inhibition of cell migration and proliferation in bladder cancer. Cell Prolif. 53(2) e12751 2020. PMID: 31971654. DOI: 10.1111/cpr.12751
187 Fu M Wang C Li Z Sakamaki T & Pestell RG Minireview: Cyclin D1: normal and abnormal functions. Endocrinology. 145(12) 5439 - 5447 2004. PMID: 15331580. DOI: 10.1210/en.2004-0959
188 Liu PC & Thiele DJ Novel stress-responsive genes EMG1 and NOP14 encode conserved, interacting proteins required for 40S ribosome biogenesis. Mol Biol Cell. 12(11) 3644 - 3657 2001. PMID: 11694595. DOI: 10.1091/mbc.12.11.3644
189 Du Y Liu Z You L Hou P Ren X Jiao T Zhao W Li Z Shu H Liu C & Zhao Y Pancreatic cancer progression relies upon mutant p53-induced oncogenic signaling mediated by NOP14. Cancer Res. 77(10) 2661 - 2673 2017. PMID: 28280038. DOI: 10.1158/0008-5472.CAN-16-2339
190 Rhee I Bachman KE Park BH Jair KW Yen RW Schuebel KE Cui H Feinberg AP Lengauer C Kinzler KW Baylin SB & Vogelstein B DNMT1 and DNMT3b cooperate to silence genes in human cancer cells. Nature. 416(6880) 552 - 556 2002. PMID: 11932749. DOI: 10.1038/416552a
191 Zo RB & Long Z MiR-124-3p suppresses bladder cancer by targeting DNA methyltransferase 3B. J Cell Physiol. 234(1) 464 - 474 2018. PMID: 29893409. DOI: 10.1002/jcp.26591
192 Ling H Fabbri M & Calin GA MicroRNAs and other non-coding RNAs as targets for anticancer drug development. Nat Rev Drug Discov. 12(11) 847 - 865 2013. PMID: 24172333. DOI: 10.1038/nrd4140
193 Broderick JA & Zamore PD MicroRNA therapeutics. Gene Ther. 18(12) 1104 - 1110 2011. PMID: 21525952. DOI: 10.1038/gt.2011.50
194 Gambari R Brognara E Spandidos DA & Fabbri E Targeting oncomiRNAs and mimicking tumor suppressor miRNAs: Νew trends in the development of miRNA therapeutic strategies in oncology (Review). Int J Oncol. 49(1) 5 - 32 2016. PMID: 27175518. DOI: 10.3892/ijo.2016.3503
195 MacLeod AR & Crooke ST RNA therapeutics in oncology: Advances, challenges, and future directions. J Clin Pharmacol. 57(Suppl) 10 S43 - S59 2017. PMID: 28921648. DOI: 10.1002/jcph.957
196 Li Z & Rana TM Therapeutic targeting of microRNAs: current status and future challenges. Nat Rev Drug Discov. 13(8) 622 - 638 2014. PMID: 25011539. DOI: 10.1038/nrd4359
197 Garofalo M Leva GD & Croce CM MicroRNAs as anti-cancer therapy. Curr Pharm Des. 20(33) 5328 - 5335 2014. PMID: 24479801. DOI: 10.2174/1381612820666140128211346
198 Malek A Merkel O Fink L Czubayko F Kissel T & Aigner A In vivo pharmacokinetics, tissue distribution and underlying mechanisms of various PEI(-PEG)/siRNA complexes. Toxicol Appl Pharmacol. 236(1) 97 - 108 2009. PMID: 19371615. DOI: 10.1016/j.taap.2009.01.014
199 Wang AZ Langer R & Farokhzad OC Nanoparticle delivery of cancer drugs. Annu Rev Med. 63 185 - 198 2012. PMID: 21888516. DOI: 10.1146/annurev-med-040210-162544
200 Rozema DB Lewis DL Wakefield DH Wong SC Klein JJ Roesch PL Bertin SL Reppen TW Chu Q Blokhin AV Hagstrom JE & Wolff JA Dynamic PolyConjugates for targeted in vivo delivery of siRNA to hepatocytes. Proc Natl Acad Sci USA. 104(32) 12982 - 12987 2007. PMID: 17652171. DOI: 10.1073/pnas.0703778104
201 Kanasty R Dorkin JR Vegas A & Anderson D Delivery materials for siRNA therapeutics. Nat Mater. 12(11) 967 - 977 2013. PMID: 24150415. DOI: 10.1038/nmat3765
202 Nair JK Willoughby JL Chan A Charisse K Alam MR Wang Q Hoekstra M Kandasamy P Kel’in AV Milstein S Taneja N O’Shea J Shaikh S Zhang L van der Sluis RJ Jung ME Akinc A Hutabarat R Kuchimanchi S Fitzgerald K Zimmermann T van Berkel TJ Maier MA Rajeev KG & Manoharan M Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J Am Chem Soc. 136(49) 16958 - 16961 2014. PMID: 25434769. DOI: 10.1021/ja505986a
203 Jones D Setbacks shadow microRNA therapies in the clinic. Nat Biotechnol. 36(10) 909 - 910 2018. PMID: 30307922. DOI: 10.1038/nbt1018-909
cdp > Vol 1 - 4 > Pages 245 - 263