Next Article in Journal
Histone Deacetylase Inhibitors Downregulate Calcium Pyrophosphate Crystal Formation in Human Articular Chondrocytes
Previous Article in Journal
Exudative versus Nonexudative Age-Related Macular Degeneration: Physiopathology and Treatment Options
Previous Article in Special Issue
Long Non-Coding RNA ANRIL as a Potential Biomarker of Chemosensitivity and Clinical Outcomes in Osteosarcoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

MicroRNA Signatures in the Upper Urinary Tract Urothelial Carcinoma Scenario: Ready for the Game Changer?

1
Biorek S.r.l., San Raffaele Scientific Institute, 20132 Milan, Italy
2
Department of Medical Oncology Livestrong Cancer Institutes, Dell Medical School, University of Texas at Austin, Austin, TX 78723, USA
3
Urological Research Institute, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
4
Faculty of Medicine and Surgery,, Università Vita-Salute San Raffaele, 20132 Milan, Italy
5
Nephrology, Dialysis, and Transplantation, Università degli Studi di Cagliari, G. Brotzu Hospital, 09134 Cagliari, Italy
6
Department of Medical Oncology and Medical Education, Dell Medical School, Livestrong Cancer Institutes, University of Texas at Austin, Austin, TX 78723, USA
7
Unit of Urology, San Raffaele Scientific Institute, 20132 Milan, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(5), 2602; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23052602
Submission received: 16 December 2021 / Revised: 20 February 2022 / Accepted: 24 February 2022 / Published: 26 February 2022

Abstract

:
Upper urinary tract urothelial carcinoma (UTUC) represents a minor subgroup of malignancies arising in the urothelium of the renal pelvis or ureter. The estimated annual incidence is around 2 cases per 100,000 people, with a mean age at diagnosis of 73 years. UTUC is more frequently diagnosed in an invasive or metastatic stage. However, even though the incidence of UTUC is not high, UTUC tends to be aggressive and rapidly progressing with a poor prognosis in some patients. A significant challenge in UTUC is ensuring accurate and timely diagnosis, which is complicated by the non-specific nature of symptoms seen at the onset of disease. Moreover, there is a lack of biomarkers capable of identifying the early presence of the malignancy and guide-tailored medical treatment. However, the growing understanding of the molecular biology underlying UTUC has led to the discovery of promising new biomarkers. Among these biomarkers, there is a class of small non-coding RNA biomarkers known as microRNAs (miRNAs) that are particularly promising. In this review, we will analyze the main characteristics of UTUC and focus on microRNAs as possible novel tools that could enter clinical practice in order to optimize the current diagnostic and prognostic algorithm.

1. Introduction

Upper urinary tract urothelial carcinoma (UTUC) is a minor subgroup of malignancies arising in the urothelium of the renal pelvis or ureter (5–10%) [1]. The estimated annual incidence is around two cases per 100,000 people [2], with a mean age at diagnosis of 73 years, and a male predominance of 2–3:1 is reported [2,3].
UTUC can be multifocal and/or metachronous, with an underlying oligoclonality or, more often, monoclonality [4]. In 17% of UTUC cases, a concurrent bladder cancer is present [5], while a recurrence in the bladder or in the contralateral upper tract is observed in 4.6–47% and in 2–6% of cases, respectively [6,7,8,9].

2. Risk Factors and Genetics of UTUCs

UTUCs are usually sporadic, though familial/hereditary UTUCs may be observed. Indeed, up to 20% of familial/hereditary UTUC cases seem to be related to Lynch’s syndrome (LS), which is a hereditary non-polyposis colorectal carcinoma (HNPCC)-related tumor [10] that is characterized by a female predominance, younger age at onset [11] and a greater susceptibility to developing a contralateral UTUC [12]. The European Association of Urology (EAU) UTUC guidelines (GL) provide a weak recommendation on evaluation based on the Amsterdam criteria, in patients suspected of having LS, to identify individuals and families at risk for HNPCC-related tumors [13].
Beyond hereditary and genetic risk-associated factors, environmental factors such as smoking and, in certain geographical areas, aristolochic acid are strongly linked to sporadic UTUCs. This supports a causative role for carcinogenic agents [13] especially in individuals who are more susceptible to UTUC. Both tobacco product use and exposure to aristolochic acid should be assessed as risk factors according to the 2021 EAU guidelines for upper tract urothelial cancer (weak recommendation) [14]. Indeed, aristolochic acid is a recently investigated, potential carcinogenic agent, known to cause Balkan endemic nephropathy/Chinese herb nephropathy, and to induce a p53 gene mutation, thus resulting in a higher regional UTUC risk [15]. For instance, the incidence in Taiwan is so high that one-fourth of all UTUCs arise from the upper urothelial tract [16], whereas tobacco use, on the other hand, is a known risk factor for UTUC with a relative risk (RR) of 2.5–7 [17]. An association with worse oncological outcome after a radical nephroureterectomy has been observed, with an increased risk of UC recurrence and mortality [18]. Moreover, UTUC are associated with Black foot vasculitis and CYC treatment [17].

3. Diagnosis and Staging

UTUCs are more frequently diagnosed in an invasive or metastatic stage, comprising approximately 60% and 7% of total cases, respectively [15]. As a consequence, the diagnosis of UTUC is often incidental or discovered upon further evaluation prompted by clinical manifestations due to local or systemic disease [19]. The most common presenting symptoms include, in order of their prevalence, visible or non-visible hematuria (73–80%) [20,21], flank pain (30%), or a palpable mass, the latter of which is infrequently observed (6%) [21]. Patients with UTUC who develop systemic symptoms require an appropriate evaluation to assess the presence, site and number of metastases, all of which are associated with a worse prognosis [19].
A variety of diagnostic modalities are employed to facilitate both the detection and staging of UTUC and multifocal UC, as well as more complete risk stratification and prognostic evaluation in the pre-operative setting. This, in turn, supports a multidisciplinary team-decision making process focused on making the most appropriate decisions on optimal treatment strategies [15].
In patients suspected of having an UTUC, whether arising from an unexpected incidental lesion detected during an imaging procedure or following an initial evaluation for hematuria and/or flank pain, the first-choice modality for diagnosis and staging is computed tomography urography (CT-U), according to the EAU UTUC guidelines [14]. Of available imaging techniques, CT-U has the highest diagnostic accuracy with a greater than 90% sensitivity and specificity (92–97% and 93–95%, respectively) [22,23], a 79% positive predictive value and a high negative predictive value (99%) [22]. Furthermore, imaging findings, such as more severe hydronephrosis or CT-texture analysis, seem to predict non-organ-confined disease, although the use of CT-U in risk stratification needs further study to be validated for clinical practice [24].
An alternative to CT-U is magnetic resonance urography [25], though its use is only weakly recommended and it must be used with caution in patients with category G4 or G5 chronic kidney disease (CKD) [14]. Nonetheless, relative safety in this context has been observed; however, there is the risk of systemic nephrogenic fibrosis when receiving one dose of group II gadolinium-based contrast administration below 0.07% in CKD G4–G5 patients, below 0.5% in CKD G5 non-dialysis subjects, and below 0.2% for dialysis-dependent patients [26].
An integral part of the uro-oncologic work-up is the use of urethrocystoscopy to identify or exclude concomitant bladder cancer, since UCs are often multifocal. The aim is to achieve precise disease staging and early diagnosis of multifocal UC in order to tailor treatment to individual patients [5,15].
Another useful diagnostic tool is cytology, although it is only modestly predictive for high grade or muscle-invasive UC in patients who have already undergone a radical surgery. Therefore, it should only be performed selectively for the affected upper tract [27] and it should be integrated with other endoscopic and imaging data.
Indeed, positive or suspicious cytology findings require in-depth evaluation and close follow-up even when endoscopy is negative [28]. As a matter of fact, abnormal cytologic findings may suggest a high-grade UTUC even when a bladder lesion is not identified by cystoscopy, and is considered a significative pre-operative variable in UTUC risk-stratification. Moreover, an abnormal cytology examination may be observed in bladder or prostatic-urethra in situ carcinomas undetected by endoscopic examination (e.g., hexaminolevulinate-guided fluorescence cystoscopy) [28,29] or other additional diagnostic techniques (e.g., fluorescence in situ hybridization) [28].
Diagnostic flexible ureterorenoscopy (URS) allows direct endoscopic visualization of visible lesions in the upper urinary tract through white light endoscopy [15,30]. Moreover, growing evidence on the use of techniques such as narrow-band imaging or photodynamic diagnostic flexible URS support a possible role in the detection of flat lesions that would otherwise be overlooked [31,32]. The other advantage of URS is its ability to perform not only cytologic but also biopsy sampling of any identified suspicious lesions, although tumor under-grading may occur and it is unreliable for pre-RNU staging purposes, with a 63% overall accuracy (when used as the sole staging technique) [33], since lamina propria is often not included in URS biopsies (in up to 32% of cases). Furthermore, the accuracy of histopathological examination of URS bioptic sampling can be limited by inadequate tissue-sampling artefacts. On the other hand, URS biopsy grade appears to correlate with pathological stage and is considered a remarkable pre-operative variable in risk stratification [30]. In summation, URS is not an upfront diagnostic-staging technique and should be used in the setting of insufficient diagnostic and risk stratification data collected by UCT, cystoscopy and cytology [30,34].
CT of the chest, abdomen and pelvis is performed as a means to detect distant metastases, allowing thorough staging of UTUC, and consequently directly guiding treatment planning in case of metastatic or non-metastatic disease [14]. Encouraging, but preliminary data on FDG-PET/CT staging are emerging, with 85% sensitivity in detecting metastases, nonetheless insufficient to support its indication in clinical practice guidelines at the moment [35].
UTUC can be stratified according to stage as non-invasive papillary tumors, carcinoma in situ and invasive carcinomas, [36] (Herrera Puerto et al., 1989) the latter being the most common scenario observed at diagnosis and staging should be expressed according to the 8th edition of tumor, node, metastasis (TNM) classification [14].
In summary, distinguishing a non-muscle invasive from a muscle-invasive UTUC can be challenging in the pre-operatory setting, since URS is not a reliable staging tool. For this reason, a number of pre-operative prognostic factors, predictive tools such as pre-radical nephroureterectomy models and prognostic nomograms have been evaluated to risk stratify UTUC, with the aim of differentiating between organ-confined and non-organ confined/muscle-invasive UTUC and select those who can benefit the most from a non-radical surgical approach [34].

4. Classification, Prognosis and Risk-Stratification

The histological classification and grading of UTUCs are based on the WHO classification, since most of the published data still use the 1973 WHO classification [37]. UTUC can also present with a wide spectrum of histologic variants, introducing further prognostic variability as they are well-recognized postoperative negative prognostic factors [38].
UTUCs are often diagnosed as invasive cancer and prognosis is strongly influenced by the TNM stage. In a series, 5-year overall survival was 73%; for pT0, pTa and pTis it was 94%, while for invasive stages it dropped to 75%, 54% and 12% for pT2, pT3 and pT4, respectively [39].
The latest EAU UTUC guidelines suggest, as a weak recommendation because of the lack of level 1 supporting evidence, risk stratifying UTUC using preoperative factors for therapeutic guidance, identifying low- and high-risk cancers, the former being more likely to benefit from kidney treatment, the latter from a radical surgical approach. Moreover, chronological age alone should not preclude RNU with curative intent [14].
A wide array of prognostic factors have been investigated [40]. They are usually divided as pre-operative and post-operative, the former being of relevance in risk-stratification and in surgery planning, and for this reason are often incorporated in predictive tools used to identify non-organ-confined high-risk UTUC [14].
Before surgery, the presence of several significant prognostic factors can be taken into account by the clinician, such as a high-grade URS biopsy finding, positive or high-grade cytology findings older age [30,41,42], smoking [18,43], ureteral location and multifocality [43,44,45], locally advanced disease with hydronephrosis [46,47], a surgical delay over 12 weeks [48,49], systemic symptoms suggestive for advanced-metastatic disease [14] obesity and higher body mass index [50] high pre-treatment-derived neutrophil–lymphocyte ratio [51,52].
The most important intra- and post-operative prognostic factors reported in the literature are primarily tumor stage and gradewhile other relevant factors include lymph node (LN) involvement and extra-nodal extension [53], lymph vascular invasion, positive surgical margins [54,55], variant histology, extensive tumor necrosis [56,57,58], sessile growth pattern [59,60] concomitant CIS in organ-confined UTUC, history of bladder CIS [59,61] and distal ureteral management [14].
As in bladder cancer, a wide spectrum of molecular markers (related to cell adhesion, microsatellite instability, cell differentiation, angiogenesis, cell proliferation, epithelial-mesenchymal transition, mitosis, apoptosis, vascular invasion, programmed death-ligand 1 expression, c-MET), including miRNA, have been studied. Nevertheless, there is still insufficient evidence of their prognostic impact to support their use in clinical practice, since at present their use is not clinically validated [14,40].

5. Treatment Management

Treatment decision-making is guided by prognostic evaluation using tumor stage. As a matter of fact, non-metastatic and metastatic UTUCs require different management with different treatment goals: potentially curative and palliative, respectively [34,54,56,62].
A non-metastatic UTUC can be risk stratified as a low-risk carcinoma (unifocal disease, tumor size < 2 cm, low-grade cytology, low-grade URS biopsy, a non-invasive aspect on CT urography) or as a high-risk carcinoma (multifocal disease, tumor size ≥ 2 cm, hydronephrosis, high-grade cytology, high-grade URS biopsy, variant histology, prior radical cystectomy for high-grade bladder cancer) [34,63,64]
Low-risk carcinomas are treated with kidney-sparing surgery as survival rates are similar to those after RNU [65]. Depending on the tumor characteristics, the clinician may choose endoscopic ablation with flexible ureteroscopy [66,67], segmental resection or a percutaneous approach [65,68].
Low-level evidence supports the use of upper urinary tract instillation of Bacillus Calmette–Guerin (BCG) or mitomycin C [69,70], performed either with an anterograde, retrograde or combined approach [71,72,73], since both treatment modalities result in similar overall survival, recurrence and progression rates [74].
The standard of care for high-risk carcinomas is RNU, with an open [39], laparoscopic [75,76] or robotic approach [77,78]. On the contrary, in patients with a solitary kidney and/or impaired renal function, kidney-sparing management should be considered on a case-by-case basis [79]. Along with the radical resection of the tumor, the homolateral kidney and ureter, the patient is offered a bladder cuff resection, which consists in a vesical ureteral orifice resection, to further reduce the risk of bladder UC recurrence [80,81,82].
Open RNU, laparoscopic and robotic approaches all have similar oncological outcomes in patients with organ-confined UTUCs, while for non-organ-confined cancers (cT3/4, cN+, cM+ in the TNM classification) an open RNU approach is the preferred choice [83,84,85,86,87].
In the setting of high risk UTUC already in the muscle-invasive stage, also performing a lymph node dissection (LND) is associated with lower recurrence rates, improved cancer-specific survival [88], and improved survival regardless of the “N” stage [89,90,91]. Two to ten days after RNU, the intravesical instillation of a single dose of BCG or Mitomycin C is associated with reduced bladder UC recurrence rate [92,93]. EAU GLs for UTUC also suggest, with a weak strength recommendation, intravesical chemotherapeutic instillation in the setting of kidney-sparing surgery [14].
The role of neoadjuvant chemotherapy (NeoCT) has been evaluated in patients with advanced UTUC; despite the lack of randomized controlled trials, contemporary literature is rapidly growing and has also been recently enriched with prospective evidence that led it to be considered as a favorable option. NeoCT usually includes a platinum compound and has been associated with lower pathological downstaging: >60% of high-grade patients showed a ≤1 ypT1 stage after neoadjuvant therapy in a phase II trial. Furthermore, a recent metanalysis observed a 38% pooled pathologic tumor downstaging rate. Moreover, NeoCT increased complete response rates (especially in high-grade UTUC, in up to 14% of cases), and lower disease recurrence and mortality rates versus standard radical surgery [34,64].
Adjuvant chemotherapy is an option for patients with locally advanced UTUC, though controversies remain based on the conflicting results reported in the current literature [94,95,96,97] and the greater susceptibility to chemotherapy-induced kidney injury in patients with an iatrogenic solitary kidney [98,99]. Current evidence supports the use of a platinum-based regimen over non-platinum regimens. UTUC-specific randomized clinical trials (RCTs) are scarce and generally support the use of an early combination of a platinum compound with another antineoplastic agent (e.g., Cisplatin + Gemcitabine) [100]. EAU GLs strongly recommend postoperative systemic chemotherapy with platinum-based regimens in patients with muscle-invasive UTUC [14].
Metastatic UTUC treatment is primarily based on systemic chemotherapy as a palliative treatment [34,62,101]. Even in the presence of metastases, there is room for some surgical options. As a matter of fact, RNU might be offered to symptomatic patients with resectable, locally advanced disease, as it may improve quality of life and outcomes [102,103,104], gaining a “weak recommendation” in the EAU UTUC guidelines if a resection is still technically feasible [14].
Metastasectomy is an option, although the absence of evidence supporting its efficacy imposes a case-by-case evaluation with the patient [14]. Current UTUC-specific evidence on systemic chemotherapy is growing, though bladder-cancer-related literature still plays a big role in supporting chemotherapy practices in UTUC patients. Chemotherapy should be offered to all patients with metastatic disease since cisplatin-based regimens can improve median survival, while single-agent and carboplatin-based combinations are less effective than cisplatin-based combination chemotherapy in terms of complete response and survival [62,64,101,105].
Platinum-based combination chemotherapy is the treatment of choice in the first-line management of cisplatin-eligible patients [62,64,101]. Cisplatin can be combined with Gemcitabine (GC) or Methotrexate + Vinblastine + Adriamycin (MVAC and high-dose MVAC) or Paclitaxel + Gemcitabine (PCG) and the use of these regimens is strongly recommended by the EAU GLs [14].
Carboplatin might be a first-choice agent in cisplatin-ineligible patients, coupled with another antineoplastic agent in a carboplatin-based regimen [106]. In the first line setting, two immune checkpoint inhibitors (ICIs) have been approved for UTUC management. Currently, pembrolizumab [107] and atezolizumab [108,109] have been approved by the FDA and EMA and are clinically available, though the EAU GLs weakly support their use in this setting due to the scarce, but growing, literature [14].
If a patient treated in the first line setting displays progression during or after administration of chemotherapy or ICIs, several second-line agents can be used, though the EAU GLs strongly recommend only pembrolizumab, atezolizumab or nivolumab, while they state that vinflunine monotherapy should be offered as a third or subsequent treatment line or as second-line treatment if immune checkpoint inhibitors or combination chemotherapy are not feasible [14].
At the moment, different ICIs have been approved by the FDA and EMA for metastatic UTUC-patients in the second-line setting, as they share similar efficacy and safety, though UTUC-specific literature is scarce. Pembrolizumab and Atezolizumab can also be used in this context [110,111,112,113], whereas Avelumab, Durvalumab and Nivolumab have only been approved as second-line agents [114,115,116,117].
Given the wide array of treatment-related toxicities, as with other antineoplastic agents, UTUC-patients treated with ICIs require multidisciplinary care [118].
Management of UTUCs remains difficult due to the limitations of the current predictive and prognostic tools. Therefore, specific biomarkers must be identified to predict outcomes and tailor personalized treatment and surveillance strategies in order to increase survival and decrease morbidity in UTUC affected patients.

6. MicroRNAs as Biomarkers in UTUC: Proof in Principle

The growing understanding of the molecular biology underlying UTUC has led to the discovery of promising new biomarkers. Among the most promising is a class of small non-coding RNA biomarkers known as microRNAs (miRNAs).
miRNAs are relatively small (approximately 18–24 nucleotides), single-stranded endogenous RNA molecules that negatively regulate gene expression at the post-transcriptional level by binding to the 3′-untranslated region of target messenger RNA (mRNA) [119,120]. They can repress translation or lead to degradation of mRNA targets based on imperfect or perfect complementarity between the miRNA and the mRNA sequence, respectively [119]. According to the latest release of the miRBase database (v.22), there are now more than 2600 unique mature human miRNAs [121]. A single miRNA can regulate up to 200 mRNA transcripts [122], and thousands of human genes are conserved targets of miRNAs [123]. Given this vast majority of mRNA targets regulated by miRNAs, miRNAs potentially influence almost all genetic pathways [122]. Indeed, miRNAs play a crucial role in diverse biological processes including development, cell growth, differentiation, apoptosis and proliferation [124]. The dysregulation of these processes is a hallmark of cancer [125]. As might be expected, an altered expression of miRNAs has been associated with the pathogenesis of cancer, including initiation and progression of cancer, as well as with many other pathological conditions [126].
It has been estimated that more than half of the miRNA genes are located in cancer-associated genomic regions or in fragile sites [127]. Based on the role of their mRNA targets, deregulated miRNAs can act as oncogenes (oncomiRs) or tumor-suppressor genes (tumor suppressive miRs) [128]. OncomiRs are mostly overexpressed and tumor suppressive miRs are under-expressed in cancer [126].
Aberrant expression levels of miRNA transcripts, either downregulated or upregulated, in comparison with those in the corresponding normal tissues have been observed in a broad variety of human malignant cancers [129,130,131,132,133,134,135,136,137,138]. miRNA expression profiles can be unique for different cancer types and may also characterize tumor histology. In addition, miRNA expression can change during tumor progression, reflecting the clinicopathological features of the tumor such as grade, stage, aggressiveness, vascular invasion and proliferation index [128,139]. Thus, miRNAs could be ideal candidates as diagnostic and prognostic biomarkers. miRNAs could also be involved in chemoresistance, as has been shown in many studies [128,140,141,142,143,144,145,146,147,148], and consequently could also be used as predictive biomarkers as well as constituting possible therapeutic targets.
miRNAs are not only present in tissue cells but can also be released into the extracellular space and then transported into bodily fluids, such as peripheral blood and urine. These cell-free or circulating miRNAs are key regulators of cellular crosstalk, modulating gene expression in recipient cells under normal and pathological conditions, such as cancer. Indeed, as part of a cell-to-cell communication mechanism in cancer, circulating miRNAs have been associated with cell proliferation and migration, metastasis, epithelial–mesenchymal transition (EMT), angiogenesis and evasion of immune response [149,150]. miRNAs can be released by cells through both vesicle trafficking and protein carrier mechanisms: (i) packaged into extracellular vesicles (EVs), such as exosomes, microvescicles, or apoptotic bodies; (ii) associated with high-density lipoprotein (HDL); (iii) or associated with RNA-binding proteins such as argonaute proteins (Ago1 or Ago2), nucleophosmin (NPM1), or ribosomal proteins [151,152,153,154,155].
miRNAs are thus also protected from degradation by RNase and are very stable in biofluids. Recipient cells can then uptake EVs carrying miRNAs via different mechanisms, including direct membrane fusion, endocytosis, micropinocytosis, phagocytosis and receptor binding. HDL-binding miRNAs can also be delivered to recipient cells via unknown mechanisms. On the other hand, there is no evidence of AGO2–miRNA complexes or NPM1-bound miRNAs being delivered into recipient cells [151,152,153,154,155].
Levels of circulating miRNAs in the plasma and serum of healthy individuals were found to be constant [156,157], and miRNA signatures in blood are similar in men and women and independent of the patient’s age [158,159]. On the other hand, specific expression profiles of circulating miRNAs have been shown in a variety of cancers, including bladder [160], renal [161], gastric [162], pancreatic [163], brain [164] and lung cancers [165,166] and could be used to distinguish cancer patients from healthy individuals, as well as for the prognosis of the disease.
Another characteristic that further supports miRNAs’ potential as reliable biomarkers is their high stability in biological samples as well as in stored samples, including fixed tissue, blood and other bodily fluids. Indeed, fixed tissue can provide a tremendous source of material for the discovery of cancer-related biomarkers, and it has been shown that differing formalin fixation times do not significantly influence miRNA profiles in formalin-fixed, paraffin-embedded (FFPE) specimens, and that miRNAs are well preserved even over prolonged FFPE block storage [167,168]. In the same way, circulating miRNAs are stable after being submitted to severe conditions such as boiling, extreme pH, long storage, and several freeze–thaw cycles, that would normally degrade most RNAs [156,169]. They are also protected from degradation by RNase, as explained above.
Consequently, miRNAs could be reliable biomarkers for clinical application due to (i) their altered levels during pathological processes or diseases such as cancer; (ii) their high stability in biological samples; and (iii) the highly sensitive, accurate, and reproducible measurement methods through which they can be quantified (such as quantitative polymerase chain reaction (qPCR)). Regarding circulating miRNAs, another great advantage consists of their ease of access through non-invasive or minimally invasive isolation methods.

7. Methods for Collecting and Measuring MicroRNAs from UTUC Patients

The rapid progression of technologies such as next-generation sequencing (NGS), microarrays and qPCR and the development of powerful bioinformatic tools has made genome-wide miRNA analysis and the consequent discovery of candidate miRNAs as diagnostic, prognostic and predictive cancer biomarkers or therapeutic targets more feasible [170].
However, miRNA profiling, especially in bodily fluids, could still be very challenging both because of the intrinsic characteristics of miRNAs (i.e., small size, GC content, high degree of sequence similarity within miRNA families, low abundance in circulation) and because of pre-analytical and analytical variables [170].
Pre-analytical variables, including sample collection, processing methods and sample quality can greatly impact the results. Regarding plasma and serum samples, the type of blood collection tube used, the time interval between blood draw and plasma/serum preparation and the different methods used for the separation of plasma and serum from whole blood are important pre-analytical factors to be considered [171,172,173]. Indeed, heparin in blood-collection tubes could inhibit PCR reaction, and procedures such as centrifugation or filtration could lead to different amounts of blood cell, cell fragment and platelet contamination in serum and plasma samples [171,172,173,174,175,176]. In addition, hemolysis must be evaluated in serum and plasma samples as it may alter circulating miRNA levels by up to 50-fold [177], even with an erythrocyte contamination as low as 0.008% [178]. It must also be taken into consideration that serum has a different miRNA profile compared to plasma due to the coagulation process [179]. Regarding urine samples, one of the main underlying reasons for variability among samples is urine concentration, which could differ significantly among individuals, at different times of the day (e.g., urine collected in the early morning is generally more concentrated than urine collected later in the day) and depending on physiological or pathological conditions. In addition, when analyzing miRNAs in cell-free urine supernatant, care should be taken at all steps of sample handling and processing to minimize cell lysis, as is the case for plasma and serum samples, because RNA levels from residual cells are several orders of magnitude higher than cell-free RNA levels.
Therefore, considering that miRNAs are present at very low levels in bodily fluids, small variations in these pre-analytical variables may significantly alter circulating miRNA levels.
In order to obtain reliable results, it is also very important to use efficient methods of RNA extraction and cDNA synthesis to verify the RNA extraction and reverse transcription efficacy, e.g., using spike-in methods, and to adopt appropriate normalization strategies for differential expression analysis when miRNA expression data are obtained [170,180].
Among the various strategies for miRNA detection, RT-qPCR is the one with the highest sensitivity and accuracy and it is considered the gold standard technique for nucleic acid quantification, offering relatively simple and cost-efficient processing of samples and data [181]. RT-qPCR has increased throughput with the introduction of microfluidic card and array platforms, which allow for large-scale miRNA detection at the same time yet still with high specificity, sensitivity and high dynamic range. However, with such technology it is possible to detect only annotated miRNAs and only medium throughput can be reached [170]. Microarray is another technique for miRNA profiling based on nucleic acid hybridization between target molecules and their corresponding complementary probes. The methodology is high throughput, being able to analyze up to thousands of miRNAs in one assay, but only among those that are already known. It is also less expensive than amplification-based arrays, but it has lower dynamic range, sensitivity and specificity than RT-qPCR and NGS [170]. NGS is another promising quantitative technology for miRNA profiling that has the great advantage of allowing both the detection of known miRNAs and the discovery of novel miRNAs [170].
However, this technology is expensive and necessitates special equipment and expert bioinformaticians, so it cannot be considered user- and/or lab-friendly [170]. The use of different technologies and platforms may be one of the main reasons for the inconsistent results reported by many published studies regarding miRNAs as biomarkers of cancer, highlighting the importance of using standardized methods.
Besides the potential technical biases discussed above, an additional aspect to be considered regarding biomarker discovery is the intrinsic interindividual variability (e.g., genetic and epidemiologic variability) and the influence of disease-independent factors on circulating miRNAs levels (e.g., drug administration [182], smoking habits [183], diet [100,184] and physical activity [185]. Therefore, it is crucial during experimental design to assess the number of samples or participants that has to be included in the study to reach statistical power.
Generally, numerous steps of validation are required before biomarkers can be considered for clinical application. After the first screening and exploratory phase aimed at identifying candidate miRNAs as potential biomarkers in cancer, systematic evaluation of miRNA candidates must be performed to evaluate their ability to discriminate different conditions with high sensitivity and specificity in a larger and independent confirmatory study cohort. Ultimately, with standardized measurements and even larger cohorts of patients, the accuracy of the previous results must be revalidated to confirm miRNA candidates as biomarkers in the general population.
Finally, for the reason reported above regarding interindividual variability and environmental factors and considering that cancer is a complex and multifactorial disease [186], it is not surprising that, if used alone, miRNA could lack high sensitivity or specificity. On the other hand, a set of two or more miRNAs, a “miRNA signature”, either alone or in combination with clinical–pathological features, could show high discriminatory power [161,187,188,189,190,191,192] and have better potential for clinical use as biomarkers.
In this review, we summarize the most relevant findings of published research regarding miRNAs as candidate biomarkers of UTUC.

8. Promising MicroRNA Biomarkers in UTUC

Although the etiology of UTUC is similar to that of bladder cancer (BC), since they both are urothelial carcinomas, their natural history is significantly different. BC is mostly (75–80%) diagnosed as a superficial tumor, while UTUC is often invasive at diagnosis and thus the prognosis is poorer [193]. Given the similarities of UTUC and BC, it is likely that they share common molecular features in the development of the tumor, related to the miRNA expression, which is likely to be similarly altered. To test this hypothesis, the expression of eleven miRNAs (miR-10a, miR-21, miR-96, miR-135, miR-141, miR-182, miR-200b, miR-205, miR-429, miR-520b, miR-1244), which were previously shown to be increased in BC, was studied in normal ureter and cancerous tissue samples of patients undergoing nephroureterectomy (47 UTUC tissue samples and 36 corresponding samples of histologically normal ureters) [194]. Most miRNAs selected resulted significantly upregulated in tumor tissues; miR-205 was overexpressed in poorly differentiated UTUC. The same analysis was then performed on circulating serum miRNAs from 44 UTUC patients and 34 controls with non-malignant urological diseases. MiR-141 confirmed an increased levels in UTUC with an AUC of 0.726 (95% confidence interval 0.609–0.843) as diagnostic marker. Finally, serum miRNA levels were associated with clinical–pathological parameters, showing that miR-10a and miR-135 were reduced in muscle-invasive UTUC (pTa/pT1 vs. pT2-4). No correlations between age, sex, metastasis or grading and circulating miRNA levels were identified [194]. Notably, circulating miR-141 was found to be elevated in multiple tumor types including prostate, colon and cervical cancer [195], and associated with the drug resistance in cancer chemotherapy [196], suggesting that it can be a useful biomarker in multiple cancer settings.
Since UTUC originates from the urothelial lining of the renal pelvis and calyces, a possible anatomic overlap with renal cell carcinoma (RCC) can occur. A correct diagnosis is critical to define proper surgery and post-surgical treatments. For this purpose, miRNA expression has been investigated to distinguish the most prevalent RCC subtypes and UTUC from the normal kidney [197]. The analysis of tissue sections derived from 24 ccRCC, 5 papRCC, 3 chRCC, 5 UTUC patients and 40 normal men showed that 434 miRNAs were significantly deregulated in cancerous versus normal tissues. In particular, miRNAs specific to each tumor entity were identified: miR-3648, miR-3656 and miR-3687 discriminated UTUC from the normal tissue with a median AUC > 0.94 [197].
UTUC can be caused also by toxic molecules found in certain industries or the environment, of which aristolochic acid (AA) is the most well-known and understood. It is related to nitrophenanthrene carboxylic acid and is a carcinogenic, mutagenic and nephrotoxic compound, which can be isolated from members of the plant family of Aristolochiaceae, whose consumption results in chronic kidney disease (CKD) and UTUC [198]. To investigate whether there is a difference in miRNA expression between AA-induced UTUC and common UTUC, samples from 20 patients/group were collected. Patients who experienced AA-induced UTUC previously received a renal transplant and were treated with a standard immunosuppressive regimen [198,199]. After discovery and validation processes, the expression of miR-488 and miR-181c was found to be significantly different between the two groups. Moreover, the expression of miR-488 was higher in stage I and II than stage III and IV tumors, while miR-181c was increased in tumors > 3 cm, even if these correlations were not validated in normal UTUC patients [199].
Hypoxia represents one of the most relevant environmental elements able to promote the metastatic process. As a matter of fact, hypoxia acts as protagonist in several different biological assets such as metabolism, angiogenesis, innate immunity and induction of stemness through the hypoxia-inducible factors (HIFs) which are the major components of hypoxia signaling pathways [200].
Human solid tumors are normally less oxygenated in comparison to the normal parenchyma from which they derived. The most prominent causes of cancer hypoxia are related to a deficiency or to an inappropriate vascularization, which can also be promoted by chronic anemia, a frequent condition that affects lots of oncology patients in clinical practice [201].
Unfortunately, due to this pathological characteristic, cancer cells tend to be more resistant to treatment, especially to radiotherapy and chemotherapy, resulting in an increased tumor metastases incidence. For these issues, the hypoxia phenomenon symbolizes a hallmark of several solid cancers, being strongly linked to malignant progression and poor clinical outcomes [202]. A recent study investigated the role of miR-210 as a possible oncogenic factor in the development and establishment of UTUC [203]. miR-210 plays a crucial role in the regulation of hypoxia-induced pathogenesis [204,205]. In fact, different lines of evidence highlighted that this biological mechanism is mediated by HIF-1α through the hypoxia-responsive factor situated on the proximal promoter of the miR-210-coding DNA. Therefore, miR-210 is usually considered as a robust HIF target [206].
Moreover, several works showed that miR-210 downregulation significantly reduced clonogenicity, migration and invasion, promoted cell apoptosis, augmented the percentage of cells in the G1 phase and diminished the percentage of cells in the S phase in vitro [203,207]. In this paper, the authors compared the expression of miR-210 between tumoral tissues affected by UTUC and paired non-cancerous urothelium [207]. The total cohort was composed of eighty-three patients with UTUC who received nephroureterectomy in a tertiary university hospital from 2013 to 2015 and the clinicopathologic data were collected by reviewing medical records. The article underlined the significant upregulation of miR-210 in each tumoral specimen with respect to the healthy adjacent noncancerous urothelium. Moreover, miR-210 was found over-expressed especially in high-stage and high-grade UTUC in comparison to low grade. MiR-210 levels could distinguish the neoplasm from the healthy parenchyma with an AUC of 0.904 (95% CI = 0.843–0.865, p < 0.001) and a sensitivity and specificity of 80% and 90%, respectively [207].
This finding represents a crucial point because, if appropriately validated on a larger cohort, miR-210 levels could be fundamental for the pathologists to identify precancerous lesions before histological characteristics are established. In addition, miR-210 and its targets may be possible intriguing candidates for the UTUC personalized therapy [203,207].
Another interesting perspective related to the altered expression of a miR in the UTUC scenario is proposed by Hsu WC [208]. In this work, the authors investigated the role of miR-145-5p in the regulation of ADP ribosylation factors 6 (ARF6) in the tumorigenesis of UTUC. MiR-145-5 p is a well-studied tumor suppressor microRNA, which is downregulated in many cancers [209] and it is related to poor prognosis in prostate, gastric, cervical, glioblastoma and non-small cell lung carcinoma (NSCLC) [210,211,212,213,214]. ADP-ribosylation factor 6 (ARF6), expressed widely in mammalian cells, is a small protein that is involved in several biological mechanisms, such as membrane trafficking and actin cytoskeletal rearrangement [215]. Different studies showed that the activation and high expression of ARF 6 protein are significantly associated with the invasion and metastasis of several types of cancers, such as breast cancer, pancreatic cancer, lung cancer, etc. [216].
In the UTUC panorama, the mechanism by which ARF6 modulates the migration and aggression of the cancer cells in the urothelium district remains unknown. To elucidate this aspect, the authors analyzed 114 formalin-fixed UTUC tissues and 40 paired noncancerous urothelium samples obtained from nephroureterectomy [208]. In the molecular analysis, ARF6 expression was observed to be higher in UTUC tissues than paired adjacent normal tissues whereas a reverse correlation between ARF6 and miR-145-5p was found in the neoplastic tissues [208]. In vitro experiments showed that miR-145-5 p overexpression blocked UTUC cells migration and invasion by negatively regulating ARF6 expression [208]. Further, the authors revealed that miR-145-5-p controlled tumor cell migration and invasion by suppressing the expression of Matrix Metallopeptidase 2 (MMP2), N-cadherin, Focal adhesion kinase and Matrix Metallopeptidase 7 (MMP7) [208]. However, all the above effects were reversed by ARF6. These findings suggest that miR-145-5p may suppress UTUC cell motility and invasion by targeting ARF6/MMP7 through the epithelial–mesenchymal transition [208].
In conclusion, this work underlined that the downregulation of miR-145-5p could be a biomarker of disease-free survival or cancer-specific survival in the UTUC asset; in parallel, the study hypothesized that miR-145 therapies and other ARF6-targeting inhibitors could be novel promising drugs for UTUC patients [208].
Previous works investigated the function of miR-30a-5p in the oncological asset as a tumor suppressor which is often downregulated in several cancerous tissues [217].
Mir-30a-5p normally reduces cell proliferation, migration and invasion processes [218]. Furthermore, this oncosuppressor miR represents a biomarker of focal and segmental glomerulosclerosis and drives the epithelial-to-mesenchymal transition (EMT) in colorectal, pulmonary and gastric cancers [219,220]. The EMT process plays a fundamental role in the aforementioned biological mechanisms related to cancer aggressiveness and recent articles highlighted that mir-30a-5p blocks the invasion of breast cancer cells thanks to the suppression of EMT progression [221].
A recent paper revealed a possible crosslink between UTUC and miR-30a-5p [222]. Three selected UTUC samples were analyzed with NGS and microarray techniques to define the most prominent miRNA signatures and their possible mRNA targets in UTUC tissues. Among all miRs signatures, miR-30a-5p was significantly downregulated in UTUC tumors compared to adjacent normal tissues. The authors further validated the different expression of the selected miR using qPCR in other 22 human samples collected from cancerous tissues of UTUC and matched adjacent normal urothelium in patients who underwent nephroureterectomy [222]. The functional role of miR-30a-5p in UTUC development was further validated using a UTUC cell line (BFTC-909) [222]. The study clearly demonstrated that miR-30a-5p expression was significantly reduced in UTUC samples in comparison to their healthy counterparts, suggesting its involvement in the proliferation and metastasis of UTUC cells [222]. Moreover, the pathway enrichment analysis also underlined the involvement of MAPK and PI3K/Akt pathways in the tumorigenesis of UTUC. Finally, the study offered the first in vitro proof that miR-30a-5p over-expression significantly augmented claudin-5 expression in UTUC BFTC-909 cells, giving the idea that miR-30a-5p was probably able to counteract EMT through interrupting the functioning of mediators in the tight junction pathway [222]. In fact, claudins are the major proteic components of tight junction structure and are responsible for the maintenance of intercellular adhesion, determining epithelial cell polarity [223]. A loss of claudins expression is strictly connected with aggressive neoplasms and high recurrence in several cancers [224].
Results of global analysis of miRNA expression patterns from patients to assess stage and prognosis of urothelial carcinoma have yielded interesting candidates worthy of further analysis. Using a miRNA array, Izquierdo et al. screened 754 miRNAs from 150 patient samples [225]. Of the total pool of miRNAs, twenty-six were found to be differentially expressed in patients with progressing or non-progressing disease. These included miRNA-31 and miRNA-149 which, upon analysis of tissue samples of 18 patients, exhibited the greatest fold change. Both miRNA-31 and miRNA-149 were, in general, expressed more highly in the non-progression group compared to the progression group. Subsequent multivariate regression analysis revealed that both of these miRNAs were independent prognostic factors of tumor progression and pathological tumor stage, whereas miRNA-149 was also an independent prognostic factor of cancer specific survival [225]. These miRNAs are aberrantly expressed in several other cancer types (i.e., clear-cell RCC, squamous cell carcinoma of the tongue, prostate cancer, glioblastoma and astrocytoma) and appear to be involved in regulation of numerous immune genes, cell structure genes and cell signaling genes [225].
More recently, a study of serum samples from 33 patients with UTUC was conducted to identify biomarkers of tumor progression and cancer survival [226]. From a pool of 800 miRNAs that were evaluated, 38 were discovered to be differentially expressed in those patients who progressed compared to those whose UTUC did not progress. Independent validation of these 38 miRNAs in 21 UTUC patients (8 progressing and 13 non-progressing) refined the number of differentially expressed miRNAs to 18, of which miRNA-151b was a significant prognostic factor for tumor progression and cancer-specific survival [226]. They posit that the observed downregulation of miRNA-151b may impact downstream gene targets such as CCNE1 that is involved in T-cell signaling, DNA damage and cell cycle regulation, which has been shown to be altered in other cancer types such as breast, pancreas, lung and prostate cancers [226]. However, this is only one of 53 gene targets of miRNA-151b they identified in a network analysis of protein–protein interactions using two different software platforms. Thus, more work is clearly needed to map out and validate the underlying molecular implications of miRNA-151b downregulation in progressing UTUC.
While the precise causes and etiology of UTUC are not known, there are many associated risk factors that have been implicated which may contribute to disease onset [227]. These include geographic location, exposure to occupational and environmental toxins/chemicals, radiation therapy, bladder cancer, inflammation, tobacco smoke, as well as several other risk factors [227,228,229].
Along these lines, a recent study on the geographically restricted Balkan Endemic Neuropathy (BEN), which is strongly associated with UTUC, found a number of miRNAs that are differentially expressed in UTUC tissues of patients with UTUC that arose in BEN patients and in patients with non-BEN UTUC in comparison to normal kidney tissues [230]. Using miRNA expression data analyzed by two different statistical analysis software programs they identified miRNAs that result differentially expressed that were common to both programs for further analysis. In the BEN-UTUC patient group (n = 7) they found 10 differentially expressed miRNAs whereas in the non-BEN-UTUC patient group (n = 5) they found 15 differentially expressed miRNAs in comparison to normal kidney tissues. Of these, miRNA-205-5p was found to be upregulated in both BEN-UTUC and non-BEN-UTUC samples suggesting a role for this miRNA in the pathogenesis of UTUC [230]. Pathway analysis of miRNA-205-5p revealed potential gene targets, including extracellular matrix genes among others [230].
Tao et al. performed a miRNAs profiling of serum samples from 12 UTUC patients and 12 controls (cancer free patients of similar age); then, through RT-qPCR they validated 13 miRNAs as differentially expressed in serum samples of 46 UTUC patients compared to 30 controls [231]. Of these, statistical analysis revealed that miR-664a-3p, miR-431-5p, miR-423-5p, miR-191-5p, miR-33b-3p, miR-26a-5p, miR-22-3p, miR-16-5p, let-7b-5p and let-7c all had the potential to serve as diagnostic biomarkers of UTUC [231]. Notably, the expression levels of let-7b, let-7c and miR-22 have been shown to be altered in other types of cancers such as breast cancer, nasopharyngeal cancer, esophageal squamous cell carcinoma (ESCC) and non-small cell lung cancer (NSCLC) [231].
Finally, using tissue samples from 157 patients that had undergone radical nephroureterectomy, Browne et al. sought to determine if miRNA expression patterns could be used to stratify tumors by grade (high grade versus low grade) and whether the tumor invaded muscle [232]. They discovered that high-grade UTUC differentially expressed miR-29b-2-5p, miR-18a-5p, miR-223-3p and miR-199a-5p compared to low-grade UTUC [232]. Importantly, none of these were identified in the aforementioned studies. They also observed miRNAs differentially expressed in muscle-invasive compared with non-muscle-invasive samples: miR-10b-5p, miR-26a-5p-5p, miR-31-5p and miR-146b-5p [232]. Of these, miRNA-31 was previously reported [225]. The implications of this study are profound and, if confirmed, offer a level of fidelity to detection that could redefine staging and grading as well as open the door to development of novel targeted therapeutics.
All findings reported in this section are summarized in Table 1.
The miRNAs identified in UTUC have many distinct molecular actions that need to be explored more carefully to better understand how they can be used clinically in diagnosis or to tailor therapeutics for management (Figure 1). A comprehensive summary of the main cited studies on the diagnostic and prognostic significance of miRNA in UTUC is listed in Table 1.

9. Conclusions

The biological characteristics of circulating as well as tissue miRNAs make them promising candidates as biomarkers of UTUC that could have diagnostic, prognostic and predictive utility. These circulating and tissue miRNAs may also represent potential therapeutic targets that could lead to development of novel therapeutic approaches.

Author Contributions

Writing—original draft preparation, A.C. (Alessandra Cinque), A.C. (Anna Capasso), R.V., M.F., M.W.L., R.M., and F.T.; writing—review and editing, A.C. (Alessandra Cinque), A.C. (Anna Capasso), R.V., M.F., M.W.L., R.M., and F.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Biorek s.r.l. Capasso is financially supported by CPRIT grant RR160093 (S.G.E “CPRIT SCHOLAR IN CANCER RESEARCH) and the DoD grant W81XWH-20-1-0366 (awarded to Anna Capasso). The funding bodies had no role in the design of the study and collection, analysis and interpretation of data or in writing the manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

Cinque is founder and president of Biorek s.r.l.; Trevisani is the founder and CEO of Biorek s.r.l.; The other co-authors declare no conflicts of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Raman, J.D.; Messer, J.; Sielatycki, J.A.; Hollenbeak, C.S. Incidence and survival of patients with carcinoma of the ureter and renal pelvis in the USA, 1973-2005. BJU Int. 2011, 107, 1059–1064. [Google Scholar] [CrossRef]
  3. Joung, J.Y.; Lim, J.; Oh, C.M.; Jung, K.W.; Cho, H.; Kim, S.H.; Seo, H.K.; Park, W.S.; Chung, J.; Lee, K.H.; et al. Current Trends in the Incidence and Survival Rate of Urological Cancers in Korea. Cancer Res. Treat. 2017, 49, 607–615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Hafner, C.; Knuechel, R.; Zanardo, L.; Dietmaier, W.; Blaszyk, H.; Cheville, J.; Hofstaedter, F.; Hartmann, A. Evidence for oligoclonality and tumor spread by intraluminal seeding in multifocal urothelial carcinomas of the upper and lower urinary tract. Oncogene 2001, 20, 4910–4915. [Google Scholar] [CrossRef] [Green Version]
  5. Cosentino, M.; Palou, J.; Gaya, J.M.; Breda, A.; Rodriguez-Faba, O.; Villavicencio-Mavrich, H. Upper urinary tract urothelial cell carcinoma: Location as a predictive factor for concomitant bladder carcinoma. World J. Urol. 2013, 31, 141–145. [Google Scholar] [CrossRef]
  6. Xylinas, E.; Rink, M.; Margulis, V.; Karakiewicz, P.; Novara, G.; Shariat, S.F.; Upper Tract Urothelial Carcinoma, C. Multifocal carcinoma in situ of the upper tract is associated with high risk of bladder cancer recurrence. Eur. Urol. 2012, 61, 1069–1070. [Google Scholar] [CrossRef]
  7. Kates, M.; Badalato, G.M.; Gupta, M.; McKiernan, J.M. Secondary bladder cancer after upper tract urothelial carcinoma in the US population. BJU Int. 2012, 110, 1325–1329. [Google Scholar] [CrossRef]
  8. Li, W.M.; Shen, J.T.; Li, C.C.; Ke, H.L.; Wei, Y.C.; Wu, W.J.; Chou, Y.H.; Huang, C.H. Oncologic outcomes following three different approaches to the distal ureter and bladder cuff in nephroureterectomy for primary upper urinary tract urothelial carcinoma. Eur. Urol. 2010, 57, 963–969. [Google Scholar] [CrossRef] [PubMed]
  9. Petros, F.G. Epidemiology, clinical presentation, and evaluation of upper-tract urothelial carcinoma. Transl. Androl. Urol. 2020, 9, 1794–1798. [Google Scholar] [CrossRef]
  10. Audenet, F.; Colin, P.; Yates, D.R.; Ouzzane, A.; Pignot, G.; Long, J.A.; Soulie, M.; Phe, V.; Bensadoun, H.; Guy, L.; et al. A proportion of hereditary upper urinary tract urothelial carcinomas are misclassified as sporadic according to a multi-institutional database analysis: Proposal of patient-specific risk identification tool. BJU Int. 2012, 110, E583–E589. [Google Scholar] [CrossRef] [PubMed]
  11. Therkildsen, C.; Eriksson, P.; Hoglund, M.; Jonsson, M.; Sjodahl, G.; Nilbert, M.; Liedberg, F. Molecular subtype classification of urothelial carcinoma in Lynch syndrome. Mol. Oncol. 2018, 12, 1286–1295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Mork, M.; Hubosky, S.G.; Roupret, M.; Margulis, V.; Raman, J.; Lotan, Y.; O’Brien, T.; You, N.; Shariat, S.F.; Matin, S.F. Lynch Syndrome: A Primer for Urologists and Panel Recommendations. J. Urol. 2015, 194, 21–29. [Google Scholar] [CrossRef]
  13. American Association of Neurological Surgeons (AANS); American Society of Neuroradiology (ASNR); Cardiovascular and Interventional Radiology Society of Europe (CIRSE); Canadian Interventional Radiology Association (CIRA); Congress of Neurological Surgeons (CNS); European Society of Minimally Invasive Neurological Therapy (ESMINT); European Society of Neuroradiology (ESNR); European Stroke Organization (ESO); Society for Cardiovascular Angiography and Interventions (SCAI); Society of Interventional Radiology (SIR); et al. Multisociety Consensus Quality Improvement Revised Consensus Statement for Endovascular Therapy of Acute Ischemic Stroke. Int. J. Stroke 2018, 13, 612–632. [Google Scholar] [CrossRef]
  14. Roupret, M.; Babjuk, M.; Burger, M.; Capoun, O.; Cohen, D.; Comperat, E.M.; Cowan, N.C.; Dominguez-Escrig, J.L.; Gontero, P.; Hugh Mostafid, A.; et al. European Association of Urology Guidelines on Upper Urinary Tract Urothelial Carcinoma: 2020 Update. Eur. Urol. 2021, 79, 62–79. [Google Scholar] [CrossRef]
  15. Soria, F.; Shariat, S.F.; Lerner, S.P.; Fritsche, H.M.; Rink, M.; Kassouf, W.; Spiess, P.E.; Lotan, Y.; Ye, D.; Fernandez, M.I.; et al. Epidemiology, diagnosis, preoperative evaluation and prognostic assessment of upper-tract urothelial carcinoma (UTUC). World J. Urol. 2017, 35, 379–387. [Google Scholar] [CrossRef]
  16. Chen, C.H.; Dickman, K.G.; Moriya, M.; Zavadil, J.; Sidorenko, V.S.; Edwards, K.L.; Gnatenko, D.V.; Wu, L.; Turesky, R.J.; Wu, X.R.; et al. Aristolochic acid-associated urothelial cancer in Taiwan. Proc. Natl. Acad. Sci. USA 2012, 109, 8241–8246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Colin, P.; Koenig, P.; Ouzzane, A.; Berthon, N.; Villers, A.; Biserte, J.; Roupret, M. Environmental factors involved in carcinogenesis of urothelial cell carcinomas of the upper urinary tract. BJU Int. 2009, 104, 1436–1440. [Google Scholar] [CrossRef]
  18. Rink, M.; Xylinas, E.; Margulis, V.; Cha, E.K.; Ehdaie, B.; Raman, J.D.; Chun, F.K.; Matsumoto, K.; Lotan, Y.; Furberg, H.; et al. Impact of smoking on oncologic outcomes of upper tract urothelial carcinoma after radical nephroureterectomy. Eur. Urol. 2013, 63, 1082–1090. [Google Scholar] [CrossRef] [Green Version]
  19. Raman, J.D.; Shariat, S.F.; Karakiewicz, P.I.; Lotan, Y.; Sagalowsky, A.I.; Roscigno, M.; Montorsi, F.; Bolenz, C.; Weizer, A.Z.; Wheat, J.C.; et al. Does preoperative symptom classification impact prognosis in patients with clinically localized upper-tract urothelial carcinoma managed by radical nephroureterectomy? Urol. Oncol. 2011, 29, 716–723. [Google Scholar] [CrossRef] [PubMed]
  20. Cowan, N.C. CT urography for hematuria. Nat. Rev. Urol. 2012, 9, 218–226. [Google Scholar] [CrossRef]
  21. Inman, B.A.; Tran, V.T.; Fradet, Y.; Lacombe, L. Carcinoma of the upper urinary tract: Predictors of survival and competing causes of mortality. Cancer 2009, 115, 2853–2862. [Google Scholar] [CrossRef]
  22. Cowan, N.C.; Turney, B.W.; Taylor, N.J.; McCarthy, C.L.; Crew, J.P. Multidetector computed tomography urography for diagnosing upper urinary tract urothelial tumour. BJU Int. 2007, 99, 1363–1370. [Google Scholar] [CrossRef]
  23. Janisch, F.; Shariat, S.F.; Baltzer, P.; Fajkovic, H.; Kimura, S.; Iwata, T.; Korn, P.; Yang, L.; Glybochko, P.V.; Rink, M.; et al. Diagnostic performance of multidetector computed tomographic (MDCTU) in upper tract urothelial carcinoma (UTUC): A systematic review and meta-analysis. World J. Urol. 2020, 38, 1165–1175. [Google Scholar] [CrossRef] [PubMed]
  24. Honda, Y.; Nakamura, Y.; Teishima, J.; Goto, K.; Higaki, T.; Narita, K.; Akagi, M.; Terada, H.; Kaichi, Y.; Fujii, S.; et al. Clinical staging of upper urinary tract urothelial carcinoma for T staging: Review and pictorial essay. Int. J. Urol. 2019, 26, 1024–1032. [Google Scholar] [CrossRef]
  25. Takahashi, N.; Glockner, J.F.; Hartman, R.P.; King, B.F.; Leibovich, B.C.; Stanley, D.W.; Fitz-Gibbon, P.D.; Kawashima, A. Gadolinium enhanced magnetic resonance urography for upper urinary tract malignancy. J. Urol. 2010, 183, 1330–1365. [Google Scholar] [CrossRef] [PubMed]
  26. Weinreb, J.C.; Rodby, R.A.; Yee, J.; Wang, C.L.; Fine, D.; McDonald, R.J.; Perazella, M.A.; Dillman, J.R.; Davenport, M.S. Use of Intravenous Gadolinium-based Contrast Media in Patients with Kidney Disease: Consensus Statements from the American College of Radiology and the National Kidney Foundation. Radiology 2021, 298, 28–35. [Google Scholar] [CrossRef]
  27. Messer, J.; Shariat, S.F.; Brien, J.C.; Herman, M.P.; Ng, C.K.; Scherr, D.S.; Scoll, B.; Uzzo, R.G.; Wille, M.; Eggener, S.E.; et al. Urinary cytology has a poor performance for predicting invasive or high-grade upper-tract urothelial carcinoma. BJU Int. 2011, 108, 701–705. [Google Scholar] [CrossRef] [PubMed]
  28. Barkan, G.A.; Wojcik, E.M.; Nayar, R.; Savic-Prince, S.; Quek, M.L.; Kurtycz, D.F.; Rosenthal, D.L. The Paris System for Reporting Urinary Cytology: The Quest to Develop a Standardized Terminology. Adv. Anat. Pathol. 2016, 23, 193–201. [Google Scholar] [CrossRef]
  29. Witjes, J.A.; Redorta, J.P.; Jacqmin, D.; Sofras, F.; Malmstrom, P.U.; Riedl, C.; Jocham, D.; Conti, G.; Montorsi, F.; Arentsen, H.C.; et al. Hexaminolevulinate-guided fluorescence cystoscopy in the diagnosis and follow-up of patients with non-muscle-invasive bladder cancer: Review of the evidence and recommendations. Eur. Urol. 2010, 57, 607–614. [Google Scholar] [CrossRef]
  30. Baard, J.; de Bruin, D.M.; Zondervan, P.J.; Kamphuis, G.; de la Rosette, J.; Laguna, M.P. Diagnostic dilemmas in patients with upper tract urothelial carcinoma. Nat. Rev. Urol. 2017, 14, 181–191. [Google Scholar] [CrossRef]
  31. Kata, S.G.; Aboumarzouk, O.M.; Zreik, A.; Somani, B.; Ahmad, S.; Nabi, G.; Buist, R.; Goodman, C.; Chlosta, P.; Golabek, T.; et al. Photodynamic diagnostic ureterorenoscopy: A valuable tool in the detection of upper urinary tract tumour. Photodiagnosis Photodyn. Ther. 2016, 13, 255–260. [Google Scholar] [CrossRef]
  32. Bus, M.T.; de Bruin, D.M.; Faber, D.J.; Kamphuis, G.M.; Zondervan, P.J.; Laguna Pes, M.P.; de Reijke, T.M.; Traxer, O.; van Leeuwen, T.G.; de la Rosette, J.J. Optical diagnostics for upper urinary tract urothelial cancer: Technology, thresholds, and clinical applications. J. Endourol. 2015, 29, 113–123. [Google Scholar] [CrossRef] [PubMed]
  33. Guarnizo, E.; Pavlovich, C.P.; Seiba, M.; Carlson, D.L.; Vaughan, E.D., Jr.; Sosa, R.E. Ureteroscopic biopsy of upper tract urothelial carcinoma: Improved diagnostic accuracy and histopathological considerations using a multi-biopsy approach. J. Urol. 2000, 163, 52–55. [Google Scholar] [CrossRef]
  34. EAU. EAU Guidelines; European Association of Urology: Arnhem, The Netherlands, 2021. [Google Scholar]
  35. Tanaka, H.; Yoshida, S.; Komai, Y.; Sakai, Y.; Urakami, S.; Yuasa, T.; Yamamoto, S.; Masuda, H.; Koizumi, M.; Kohno, A.; et al. Clinical Value of 18F-Fluorodeoxyglucose Positron Emission Tomography/Computed Tomography in Upper Tract Urothelial Carcinoma: Impact on Detection of Metastases and Patient Management. Urol. Int. 2016, 96, 65–72. [Google Scholar] [CrossRef] [PubMed]
  36. Herrera Puerto, J.; Soler Fernandez, J.; Dominguez Bravo, C.; Laguna Alvarez, G.; Caballero Gomez, M.; Ruiz de la Hermosa, J.; Asuar Aydillo, S. [Sarcomatoid carcinoma of the kidney: Presentation of a case]. Actas Urol. Esp. 1989, 13, 467–469. [Google Scholar]
  37. Soukup, V.; Capoun, O.; Cohen, D.; Hernandez, V.; Babjuk, M.; Burger, M.; Comperat, E.; Gontero, P.; Lam, T.; MacLennan, S.; et al. Prognostic Performance and Reproducibility of the 1973 and 2004/2016 World Health Organization Grading Classification Systems in Non-muscle-invasive Bladder Cancer: A European Association of Urology Non-muscle Invasive Bladder Cancer Guidelines Panel Systematic Review. Eur. Urol. 2017, 72, 801–813. [Google Scholar] [CrossRef] [Green Version]
  38. Amin, M.B. Histological variants of urothelial carcinoma: Diagnostic, therapeutic and prognostic implications. Mod. Pathol. 2009, 22 (Suppl. 2), S96–S118. [Google Scholar] [CrossRef]
  39. Margulis, V.; Shariat, S.F.; Matin, S.F.; Kamat, A.M.; Zigeuner, R.; Kikuchi, E.; Lotan, Y.; Weizer, A.; Raman, J.D.; Wood, C.G.; et al. Outcomes of radical nephroureterectomy: A series from the Upper Tract Urothelial Carcinoma Collaboration. Cancer 2009, 115, 1224–1233. [Google Scholar] [CrossRef]
  40. Lughezzani, G.; Burger, M.; Margulis, V.; Matin, S.F.; Novara, G.; Roupret, M.; Shariat, S.F.; Wood, C.G.; Zigeuner, R. Prognostic factors in upper urinary tract urothelial carcinomas: A comprehensive review of the current literature. Eur. Urol. 2012, 62, 100–114. [Google Scholar] [CrossRef] [PubMed]
  41. Kim, H.S.; Jeong, C.W.; Kwak, C.; Kim, H.H.; Ku, J.H. Association between demographic factors and prognosis in urothelial carcinoma of the upper urinary tract: A systematic review and meta-analysis. Oncotarget 2017, 8, 7464–7476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Shariat, S.F.; Godoy, G.; Lotan, Y.; Droller, M.; Karakiewicz, P.I.; Raman, J.D.; Isbarn, H.; Weizer, A.; Remzi, M.; Roscigno, M.; et al. Advanced patient age is associated with inferior cancer-specific survival after radical nephroureterectomy. BJU Int. 2010, 105, 1672–1677. [Google Scholar] [CrossRef]
  43. Simsir, A.; Sarsik, B.; Cureklibatir, I.; Sen, S.; Gunaydin, G.; Cal, C. Prognostic factors for upper urinary tract urothelial carcinomas: Stage, grade, and smoking status. Int. Urol. Nephrol. 2011, 43, 1039–1045. [Google Scholar] [CrossRef]
  44. Ouzzane, A.; Colin, P.; Xylinas, E.; Pignot, G.; Ariane, M.M.; Saint, F.; Hoarau, N.; Adam, E.; Azemar, M.D.; Bensadoun, H.; et al. Ureteral and multifocal tumours have worse prognosis than renal pelvic tumours in urothelial carcinoma of the upper urinary tract treated by nephroureterectomy. Eur. Urol. 2011, 60, 1258–1265. [Google Scholar] [CrossRef]
  45. Chromecki, T.F.; Cha, E.K.; Fajkovic, H.; Margulis, V.; Novara, G.; Scherr, D.S.; Lotan, Y.; Raman, J.D.; Kassouf, W.; Bensalah, K.; et al. The impact of tumor multifocality on outcomes in patients treated with radical nephroureterectomy. Eur. Urol. 2012, 61, 245–253. [Google Scholar] [CrossRef] [PubMed]
  46. Hurel, S.; Roupret, M.; Seisen, T.; Comperat, E.; Phe, V.; Droupy, S.; Audenet, F.; Pignot, G.; Cathelineau, X.; Guy, L.; et al. Influence of preoperative factors on the oncologic outcome for upper urinary tract urothelial carcinoma after radical nephroureterectomy. World J. Urol. 2015, 33, 335–341. [Google Scholar] [CrossRef] [PubMed]
  47. Ito, Y.; Kikuchi, E.; Tanaka, N.; Miyajima, A.; Mikami, S.; Jinzaki, M.; Oya, M. Preoperative hydronephrosis grade independently predicts worse pathological outcomes in patients undergoing nephroureterectomy for upper tract urothelial carcinoma. J. Urol. 2011, 185, 1621–1626. [Google Scholar] [CrossRef] [PubMed]
  48. Xia, L.; Taylor, B.L.; Pulido, J.E.; Guzzo, T.J. Impact of surgical waiting time on survival in patients with upper tract urothelial carcinoma: A national cancer database study. Urol. Oncol. 2018, 36, 10.e15–10.e22. [Google Scholar] [CrossRef] [PubMed]
  49. Sundi, D.; Svatek, R.S.; Margulis, V.; Wood, C.G.; Matin, S.F.; Dinney, C.P.; Kamat, A.M. Upper tract urothelial carcinoma: Impact of time to surgery. Urol. Oncol. 2012, 30, 266–272. [Google Scholar] [CrossRef] [Green Version]
  50. Ehdaie, B.; Chromecki, T.F.; Lee, R.K.; Lotan, Y.; Margulis, V.; Karakiewicz, P.I.; Novara, G.; Raman, J.D.; Ng, C.; Lowrance, W.T.; et al. Obesity adversely impacts disease specific outcomes in patients with upper tract urothelial carcinoma. J. Urol. 2011, 186, 66–72. [Google Scholar] [CrossRef] [Green Version]
  51. Dalpiaz, O.; Pichler, M.; Mannweiler, S.; Martin Hernandez, J.M.; Stojakovic, T.; Pummer, K.; Zigeuner, R.; Hutterer, G.C. Validation of the pretreatment derived neutrophil-lymphocyte ratio as a prognostic factor in a European cohort of patients with upper tract urothelial carcinoma. Br. J. Cancer 2014, 110, 2531–2536. [Google Scholar] [CrossRef] [Green Version]
  52. Vartolomei, M.D.; Kimura, S.; Ferro, M.; Vartolomei, L.; Foerster, B.; Abufaraj, M.; Shariat, S.F. Is neutrophil-to-lymphocytes ratio a clinical relevant preoperative biomarker in upper tract urothelial carcinoma? A meta-analysis of 4385 patients. World J. Urol. 2018, 36, 1019–1029. [Google Scholar] [CrossRef]
  53. Fajkovic, H.; Cha, E.K.; Jeldres, C.; Donner, G.; Chromecki, T.F.; Margulis, V.; Novara, G.; Lotan, Y.; Raman, J.D.; Kassouf, W.; et al. Prognostic value of extranodal extension and other lymph node parameters in patients with upper tract urothelial carcinoma. J. Urol. 2012, 187, 845–851. [Google Scholar] [CrossRef]
  54. Petrelli, F.; Yasser Hussein, M.I.; Vavassori, I.; Barni, S. Prognostic Factors of Overall Survival in Upper Urinary Tract Carcinoma: A Systematic Review and Meta-analysis. Urology 2017, 100, 9–15. [Google Scholar] [CrossRef]
  55. Colin, P.; Ouzzane, A.; Yates, D.R.; Audenet, F.; Pignot, G.; Arvin-Berod, A.; Merigot de Treigny, O.; Laurent, G.; Valeri, A.; Irani, J.; et al. Influence of positive surgical margin status after radical nephroureterectomy on upper urinary tract urothelial carcinoma survival. Ann. Surg. Oncol. 2012, 19, 3613–3620. [Google Scholar] [CrossRef] [PubMed]
  56. Zigeuner, R.; Shariat, S.F.; Margulis, V.; Karakiewicz, P.I.; Roscigno, M.; Weizer, A.; Kikuchi, E.; Remzi, M.; Raman, J.D.; Bolenz, C.; et al. Tumour necrosis is an indicator of aggressive biology in patients with urothelial carcinoma of the upper urinary tract. Eur. Urol. 2010, 57, 575–581. [Google Scholar] [CrossRef] [PubMed]
  57. Seitz, C.; Gupta, A.; Shariat, S.F.; Matsumoto, K.; Kassouf, W.; Walton, T.J.; Fritsche, H.M.; Otto, W.; Tritschler, S.; Bastian, P.J.; et al. Association of tumor necrosis with pathological features and clinical outcome in 754 patients undergoing radical nephroureterectomy for upper tract urothelial carcinoma: An international validation study. J. Urol. 2010, 184, 1895–1900. [Google Scholar] [CrossRef] [PubMed]
  58. Deuker, M.; Stolzenbach, L.F.; Colla Ruvolo, C.; Nocera, L.; Tian, Z.; Roos, F.C.; Becker, A.; Kluth, L.A.; Tilki, D.; Shariat, S.F.; et al. Upper Urinary Tract Tumors: Variant Histology Versus Urothelial Carcinoma. Clin. Genitourin. Cancer 2021, 19, 117–124. [Google Scholar] [CrossRef]
  59. Schieda, N.; Alrashed, A.; Flood, T.A.; Samji, K.; Shabana, W.; McInnes, M.D. Comparison of Quantitative MRI and CT Washout Analysis for Differentiation of Adrenal Pheochromocytoma From Adrenal Adenoma. AJR Am. J. Roentgenol. 2016, 206, 1141–1148. [Google Scholar] [CrossRef]
  60. Remzi, M.; Haitel, A.; Margulis, V.; Karakiewicz, P.; Montorsi, F.; Kikuchi, E.; Zigeuner, R.; Weizer, A.; Bolenz, C.; Bensalah, K.; et al. Tumour architecture is an independent predictor of outcomes after nephroureterectomy: A multi-institutional analysis of 1363 patients. BJU Int. 2009, 103, 307–311. [Google Scholar] [CrossRef]
  61. Wheat, J.C.; Weizer, A.Z.; Wolf, J.S., Jr.; Lotan, Y.; Remzi, M.; Margulis, V.; Wood, C.G.; Montorsi, F.; Roscigno, M.; Kikuchi, E.; et al. Concomitant carcinoma in situ is a feature of aggressive disease in patients with organ confined urothelial carcinoma following radical nephroureterectomy. Urol. Oncol. 2012, 30, 252–258. [Google Scholar] [CrossRef]
  62. Pham, M.N.; Apolo, A.B.; De Santis, M.; Galsky, M.D.; Leibovich, B.C.; Pisters, L.L.; Siefker-Radtke, A.O.; Sonpavde, G.; Steinberg, G.D.; Sternberg, C.N.; et al. Upper tract urothelial carcinoma topical issue 2016: Treatment of metastatic cancer. World J. Urol. 2017, 35, 367–378. [Google Scholar] [CrossRef]
  63. Azizi, M.; Cheriyan, S.K.; Peyton, C.C.; Foerster, B.; Shariat, S.F.; Spiess, P.E. Optimal Management of Upper Tract Urothelial Carcinoma: An Unmet Need. Curr. Treat. Options Oncol. 2019, 20, 40. [Google Scholar] [CrossRef]
  64. Leow, J.J.; Chong, Y.L.; Chang, S.L.; Valderrama, B.P.; Powles, T.; Bellmunt, J. Neoadjuvant and Adjuvant Chemotherapy for Upper Tract Urothelial Carcinoma: A 2020 Systematic Review and Meta-analysis, and Future Perspectives on Systemic Therapy. Eur. Urol. 2021, 79, 635–654. [Google Scholar] [CrossRef]
  65. Seisen, T.; Peyronnet, B.; Dominguez-Escrig, J.L.; Bruins, H.M.; Yuan, C.Y.; Babjuk, M.; Bohle, A.; Burger, M.; Comperat, E.M.; Cowan, N.C.; et al. Oncologic Outcomes of Kidney-sparing Surgery Versus Radical Nephroureterectomy for Upper Tract Urothelial Carcinoma: A Systematic Review by the EAU Non-muscle Invasive Bladder Cancer Guidelines Panel. Eur. Urol. 2016, 70, 1052–1068. [Google Scholar] [CrossRef] [PubMed]
  66. Cutress, M.L.; Stewart, G.D.; Wells-Cole, S.; Phipps, S.; Thomas, B.G.; Tolley, D.A. Long-term endoscopic management of upper tract urothelial carcinoma: 20-year single-centre experience. BJU Int. 2012, 110, 1608–1617. [Google Scholar] [CrossRef] [PubMed]
  67. Cutress, M.L.; Stewart, G.D.; Zakikhani, P.; Phipps, S.; Thomas, B.G.; Tolley, D.A. Ureteroscopic and percutaneous management of upper tract urothelial carcinoma (UTUC): Systematic review. BJU Int. 2012, 110, 614–628. [Google Scholar] [CrossRef]
  68. Roupret, M.; Traxer, O.; Tligui, M.; Conort, P.; Chartier-Kastler, E.; Richard, F.; Cussenot, O. Upper urinary tract transitional cell carcinoma: Recurrence rate after percutaneous endoscopic resection. Eur. Urol. 2007, 51, 709–713. [Google Scholar] [CrossRef]
  69. Redrow, G.P.; Guo, C.C.; Brausi, M.A.; Coleman, J.A.; Fernandez, M.I.; Kassouf, W.; Keeley, F.X., Jr.; Margulis, V.; Raman, J.D.; Roupret, M.; et al. Upper Urinary Tract Carcinoma In Situ: Current Knowledge, Future Direction. J. Urol. 2017, 197, 287–295. [Google Scholar] [CrossRef] [PubMed]
  70. Giannarini, G.; Kessler, T.M.; Birkhauser, F.D.; Thalmann, G.N.; Studer, U.E. Antegrade perfusion with bacillus Calmette-Guerin in patients with non-muscle-invasive urothelial carcinoma of the upper urinary tract: Who may benefit? Eur. Urol. 2011, 60, 955–960. [Google Scholar] [CrossRef]
  71. Irie, A.; Iwamura, M.; Kadowaki, K.; Ohkawa, A.; Uchida, T.; Baba, S. Intravesical instillation of bacille Calmette-Guerin for carcinoma in situ of the urothelium involving the upper urinary tract using vesicoureteral reflux created by a double-pigtail catheter. Urology 2002, 59, 53–57. [Google Scholar] [CrossRef]
  72. Horiguchi, H.; Yoneyama, T.; Hatakeyama, S.; Tokui, N.; Sato, T.; Fujita, N.; Yamamoto, H.; Tobisawa, Y.; Yoneyama, T.; Hashimoto, Y.; et al. Impact of bacillus Calmette-Guerin therapy of upper urinary tract carcinoma in situ: Comparison of oncological outcomes with radical nephroureterectomy. Med. Oncol. 2018, 35, 41. [Google Scholar] [CrossRef]
  73. Yossepowitch, O.; Lifshitz, D.A.; Dekel, Y.; Ehrlich, Y.; Gur, U.; Margel, D.; Livne, P.M.; Baniel, J. Assessment of vesicoureteral reflux in patients with self-retaining ureteral stents: Implications for upper urinary tract instillation. J. Urol. 2005, 173, 890–893. [Google Scholar] [CrossRef]
  74. Foerster, B.; D’Andrea, D.; Abufaraj, M.; Broenimann, S.; Karakiewicz, P.I.; Roupret, M.; Gontero, P.; Lerner, S.P.; Shariat, S.F.; Soria, F. Endocavitary treatment for upper tract urothelial carcinoma: A meta-analysis of the current literature. Urol. Oncol. 2019, 37, 430–436. [Google Scholar] [CrossRef]
  75. Roupret, M.; Smyth, G.; Irani, J.; Guy, L.; Davin, J.L.; Saint, F.; Pfister, C.; Wallerand, H.; Rozet, F. Oncological risk of laparoscopic surgery in urothelial carcinomas. World J. Urol. 2009, 27, 81–88. [Google Scholar] [CrossRef] [PubMed]
  76. Ong, A.M.; Bhayani, S.B.; Pavlovich, C.P. Trocar site recurrence after laparoscopic nephroureterectomy. J. Urol. 2003, 170, 1301. [Google Scholar] [CrossRef] [PubMed]
  77. Clements, M.B.; Krupski, T.L.; Culp, S.H. Robotic-Assisted Surgery for Upper Tract Urothelial Carcinoma: A Comparative Survival Analysis. Ann. Surg. Oncol. 2018, 25, 2550–2562. [Google Scholar] [CrossRef]
  78. Aboumohamed, A.A.; Krane, L.S.; Hemal, A.K. Oncologic Outcomes Following Robot-Assisted Laparoscopic Nephroureterectomy with Bladder Cuff Excision for Upper Tract Urothelial Carcinoma. J. Urol. 2015, 194, 1561–1566. [Google Scholar] [CrossRef] [PubMed]
  79. Su, X.; Fang, D.; Zhang, L.; Xiong, G.; Hao, H.; Zhao, G.; Yang, K.; Xing, Y.; Li, X.; Zhou, L. Treatment strategies for upper tract urothelial carcinoma (UTUC) of a solitary kidney: A single-institutional analysis of 61 cases. Int. Urol. Nephrol. 2016, 48, 1601–1608. [Google Scholar] [CrossRef] [PubMed]
  80. Seisen, T.; Granger, B.; Colin, P.; Leon, P.; Utard, G.; Renard-Penna, R.; Comperat, E.; Mozer, P.; Cussenot, O.; Shariat, S.F.; et al. A Systematic Review and Meta-analysis of Clinicopathologic Factors Linked to Intravesical Recurrence After Radical Nephroureterectomy to Treat Upper Tract Urothelial Carcinoma. Eur. Urol. 2015, 67, 1122–1133. [Google Scholar] [CrossRef]
  81. Xylinas, E.; Rink, M.; Cha, E.K.; Clozel, T.; Lee, R.K.; Fajkovic, H.; Comploj, E.; Novara, G.; Margulis, V.; Raman, J.D.; et al. Impact of distal ureter management on oncologic outcomes following radical nephroureterectomy for upper tract urothelial carcinoma. Eur. Urol. 2014, 65, 210–217. [Google Scholar] [CrossRef]
  82. Phe, V.; Cussenot, O.; Bitker, M.O.; Roupret, M. Does the surgical technique for management of the distal ureter influence the outcome after nephroureterectomy? BJU Int. 2011, 108, 130–138. [Google Scholar] [CrossRef]
  83. Favaretto, R.L.; Shariat, S.F.; Chade, D.C.; Godoy, G.; Kaag, M.; Cronin, A.M.; Bochner, B.H.; Coleman, J.; Dalbagni, G. Comparison between laparoscopic and open radical nephroureterectomy in a contemporary group of patients: Are recurrence and disease-specific survival associated with surgical technique? Eur. Urol. 2010, 58, 645–651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Walton, T.J.; Novara, G.; Matsumoto, K.; Kassouf, W.; Fritsche, H.M.; Artibani, W.; Bastian, P.J.; Martinez-Salamanca, J.I.; Seitz, C.; Thomas, S.A.; et al. Oncological outcomes after laparoscopic and open radical nephroureterectomy: Results from an international cohort. BJU Int. 2011, 108, 406–412. [Google Scholar] [CrossRef] [PubMed]
  85. Ni, S.; Tao, W.; Chen, Q.; Liu, L.; Jiang, H.; Hu, H.; Han, R.; Wang, C. Laparoscopic versus open nephroureterectomy for the treatment of upper urinary tract urothelial carcinoma: A systematic review and cumulative analysis of comparative studies. Eur. Urol. 2012, 61, 1142–1153. [Google Scholar] [CrossRef] [PubMed]
  86. Rodriguez, J.F.; Packiam, V.T.; Boysen, W.R.; Johnson, S.C.; Smith, Z.L.; Smith, N.D.; Shalhav, A.L.; Steinberg, G.D. Utilization and Outcomes of Nephroureterectomy for Upper Tract Urothelial Carcinoma by Surgical Approach. J. Endourol. 2017, 31, 661–665. [Google Scholar] [CrossRef]
  87. Ariane, M.M.; Colin, P.; Ouzzane, A.; Pignot, G.; Audouin, M.; Cornu, J.N.; Albouy, B.; Guillotreau, J.; Neuzillet, Y.; Crouzet, S.; et al. Assessment of oncologic control obtained after open versus laparoscopic nephroureterectomy for upper urinary tract urothelial carcinomas (UUT-UCs): Results from a large French multicenter collaborative study. Ann. Surg. Oncol. 2012, 19, 301–308. [Google Scholar] [CrossRef]
  88. Dominguez-Escrig, J.L.; Peyronnet, B.; Seisen, T.; Bruins, H.M.; Yuan, C.Y.; Babjuk, M.; Bohle, A.; Burger, M.; Comperat, E.M.; Gontero, P.; et al. Potential Benefit of Lymph Node Dissection During Radical Nephroureterectomy for Upper Tract Urothelial Carcinoma: A Systematic Review by the European Association of Urology Guidelines Panel on Non-muscle-invasive Bladder Cancer. Eur. Urol. Focus 2019, 5, 224–241. [Google Scholar] [CrossRef]
  89. Dong, F.; Xu, T.; Wang, X.; Shen, Y.; Zhang, X.; Chen, S.; Zhong, S.; Zhang, M.; Ding, Q. Lymph node dissection could bring survival benefits to patients diagnosed with clinically node-negative upper urinary tract urothelial cancer: A population-based, propensity score-matched study. Int. J. Clin Oncol. 2019, 24, 296–305. [Google Scholar] [CrossRef]
  90. Lenis, A.T.; Donin, N.M.; Faiena, I.; Salmasi, A.; Johnson, D.C.; Drakaki, A.; Gollapudi, K.; Blumberg, J.; Belldegrun, A.; Pantuck, A.; et al. Role of surgical approach on lymph node dissection yield and survival in patients with upper tract urothelial carcinoma. Urol. Oncol. 2018, 36, 9.e1–9.e9. [Google Scholar] [CrossRef]
  91. Kondo, T.; Hashimoto, Y.; Kobayashi, H.; Iizuka, J.; Nakazawa, H.; Ito, F.; Tanabe, K. Template-based lymphadenectomy in urothelial carcinoma of the upper urinary tract: Impact on patient survival. Int. J. Urol. 2010, 17, 848–854. [Google Scholar] [CrossRef]
  92. O’Brien, T.; Ray, E.; Singh, R.; Coker, B.; Beard, R.; British Association of Urological Surgeons Section of, O. Prevention of bladder tumours after nephroureterectomy for primary upper urinary tract urothelial carcinoma: A prospective, multicentre, randomised clinical trial of a single postoperative intravesical dose of mitomycin C (the ODMIT-C Trial). Eur. Urol. 2011, 60, 703–710. [Google Scholar] [CrossRef]
  93. Ito, A.; Shintaku, I.; Satoh, M.; Ioritani, N.; Aizawa, M.; Tochigi, T.; Kawamura, S.; Aoki, H.; Numata, I.; Takeda, A.; et al. Prospective randomized phase II trial of a single early intravesical instillation of pirarubicin (THP) in the prevention of bladder recurrence after nephroureterectomy for upper urinary tract urothelial carcinoma: The THP Monotherapy Study Group Trial. J. Clin. Oncol. 2013, 31, 1422–1427. [Google Scholar] [CrossRef]
  94. Goldberg, H.; Klaassen, Z.; Chandrasekar, T.; Sayyid, R.; Kulkarni, G.S.; Hamilton, R.J.; Fleshner, N.E. Does perioperative chemotherapy improve survival in upper tract urothelial carcinoma? A population based analysis. Oncotarget 2018, 9, 18797–18810. [Google Scholar] [CrossRef] [Green Version]
  95. Necchi, A.; Lo Vullo, S.; Mariani, L.; Moschini, M.; Hendricksen, K.; Rink, M.; Sosnowski, R.; Dobruch, J.; Raman, J.D.; Wood, C.G.; et al. Adjuvant chemotherapy after radical nephroureterectomy does not improve survival in patients with upper tract urothelial carcinoma: A joint study by the European Association of Urology-Young Academic Urologists and the Upper Tract Urothelial Carcinoma Collaboration. BJU Int. 2018, 121, 252–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Fujita, K.; Taneishi, K.; Inamoto, T.; Ishizuya, Y.; Takada, S.; Tsujihata, M.; Tanigawa, G.; Minato, N.; Nakazawa, S.; Takada, T.; et al. Adjuvant chemotherapy improves survival of patients with high-risk upper urinary tract urothelial carcinoma: A propensity score-matched analysis. BMC Urol. 2017, 17, 110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Seisen, T.; Krasnow, R.E.; Bellmunt, J.; Roupret, M.; Leow, J.J.; Lipsitz, S.R.; Vetterlein, M.W.; Preston, M.A.; Hanna, N.; Kibel, A.S.; et al. Effectiveness of Adjuvant Chemotherapy After Radical Nephroureterectomy for Locally Advanced and/or Positive Regional Lymph Node Upper Tract Urothelial Carcinoma. J. Clin. Oncol. 2017, 35, 852–860. [Google Scholar] [CrossRef]
  98. Kaag, M.G.; O’Malley, R.L.; O’Malley, P.; Godoy, G.; Chen, M.; Smaldone, M.C.; Hrebinko, R.L.; Raman, J.D.; Bochner, B.; Dalbagni, G.; et al. Changes in renal function following nephroureterectomy may affect the use of perioperative chemotherapy. Eur. Urol. 2010, 58, 581–587. [Google Scholar] [CrossRef] [Green Version]
  99. Lane, B.R.; Smith, A.K.; Larson, B.T.; Gong, M.C.; Campbell, S.C.; Raghavan, D.; Dreicer, R.; Hansel, D.E.; Stephenson, A.J. Chronic kidney disease after nephroureterectomy for upper tract urothelial carcinoma and implications for the administration of perioperative chemotherapy. Cancer 2010, 116, 2967–2973. [Google Scholar] [CrossRef] [PubMed]
  100. Witwer, K.W. XenomiRs and miRNA homeostasis in health and disease: Evidence that diet and dietary miRNAs directly and indirectly influence circulating miRNA profiles. RNA Biol. 2012, 9, 1147–1154. [Google Scholar] [CrossRef]
  101. Darr, C.; Hadaschik, B.A.; Tschirdewahn, S. [Systemic treatment of metastatic tumors of the upper urinary tract]. Urologe A 2019, 58, 30–33. [Google Scholar] [CrossRef]
  102. Seisen, T.; Jindal, T.; Karabon, P.; Sood, A.; Bellmunt, J.; Roupret, M.; Leow, J.J.; Vetterlein, M.W.; Sun, M.; Alanee, S.; et al. Efficacy of Systemic Chemotherapy Plus Radical Nephroureterectomy for Metastatic Upper Tract Urothelial Carcinoma. Eur. Urol. 2017, 71, 714–718. [Google Scholar] [CrossRef] [PubMed]
  103. Moschini, M.; Xylinas, E.; Zamboni, S.; Mattei, A.; Niegisch, G.; Yu, E.Y.; Bamias, A.; Agarwal, N.; Sridhar, S.S.; Sternberg, C.N.; et al. Efficacy of Surgery in the Primary Tumor Site for Metastatic Urothelial Cancer: Analysis of an International, Multicenter, Multidisciplinary Database. Eur. Urol. Oncol. 2020, 3, 94–101. [Google Scholar] [CrossRef] [PubMed]
  104. Nazzani, S.; Preisser, F.; Mazzone, E.; Marchioni, M.; Bandini, M.; Tian, Z.; Mistretta, F.A.; Shariat, S.F.; Soulieres, D.; Saad, F.; et al. Survival Effect of Nephroureterectomy in Metastatic Upper Urinary Tract Urothelial Carcinoma. Clin. Genitourin. Cancer 2019, 17, e602–e611. [Google Scholar] [CrossRef] [PubMed]
  105. Moschini, M.; Shariat, S.F.; Roupret, M.; De Santis, M.; Bellmunt, J.; Sternberg, C.N.; Tombal, B.; Collette, L. Impact of Primary Tumor Location on Survival from the European Organization for the Research and Treatment of Cancer Advanced Urothelial Cancer Studies. J. Urol. 2018, 199, 1149–1157. [Google Scholar] [CrossRef] [PubMed]
  106. Galsky, M.D.; Chen, G.J.; Oh, W.K.; Bellmunt, J.; Roth, B.J.; Petrioli, R.; Dogliotti, L.; Dreicer, R.; Sonpavde, G. Comparative effectiveness of cisplatin-based and carboplatin-based chemotherapy for treatment of advanced urothelial carcinoma. Ann. Oncol. 2012, 23, 406–410. [Google Scholar] [CrossRef]
  107. Balar, A.V.; Castellano, D.; O’Donnell, P.H.; Grivas, P.; Vuky, J.; Powles, T.; Plimack, E.R.; Hahn, N.M.; de Wit, R.; Pang, L.; et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): A multicentre, single-arm, phase 2 study. Lancet Oncol. 2017, 18, 1483–1492. [Google Scholar] [CrossRef]
  108. Galsky, M.D.; Arija, J.A.A.; Bamias, A.; Davis, I.D.; De Santis, M.; Kikuchi, E.; Garcia-Del-Muro, X.; De Giorgi, U.; Mencinger, M.; Izumi, K.; et al. Atezolizumab with or without chemotherapy in metastatic urothelial cancer (IMvigor130): A multicentre, randomised, placebo-controlled phase 3 trial. Lancet 2020, 395, 1547–1557. [Google Scholar] [CrossRef]
  109. Balar, A.V.; Galsky, M.D.; Rosenberg, J.E.; Powles, T.; Petrylak, D.P.; Bellmunt, J.; Loriot, Y.; Necchi, A.; Hoffman-Censits, J.; Perez-Gracia, J.L.; et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: A single-arm, multicentre, phase 2 trial. Lancet 2017, 389, 67–76. [Google Scholar] [CrossRef] [Green Version]
  110. Rosenberg, J.E.; Hoffman-Censits, J.; Powles, T.; van der Heijden, M.S.; Balar, A.V.; Necchi, A.; Dawson, N.; O’Donnell, P.H.; Balmanoukian, A.; Loriot, Y.; et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: A single-arm, multicentre, phase 2 trial. Lancet 2016, 387, 1909–1920. [Google Scholar] [CrossRef] [Green Version]
  111. Powles, T.; Duran, I.; van der Heijden, M.S.; Loriot, Y.; Vogelzang, N.J.; De Giorgi, U.; Oudard, S.; Retz, M.M.; Castellano, D.; Bamias, A.; et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): A multicentre, open-label, phase 3 randomised controlled trial. Lancet 2018, 391, 748–757. [Google Scholar] [CrossRef]
  112. Sternberg, C.N.; Loriot, Y.; James, N.; Choy, E.; Castellano, D.; Lopez-Rios, F.; Banna, G.L.; De Giorgi, U.; Masini, C.; Bamias, A.; et al. Primary Results from SAUL, a Multinational Single-arm Safety Study of Atezolizumab Therapy for Locally Advanced or Metastatic Urothelial or Nonurothelial Carcinoma of the Urinary Tract. Eur. Urol. 2019, 76, 73–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Bamias, A.; Merseburger, A.S.; Loriot, Y.; James, N.; Choy, E.; Castellano, D.; Lopez-Rios, F.; Calabro, F.; Kramer, M.; de Velasco, G.; et al. SAUL, a single-arm study of atezolizumab for chemotherapy-pretreated locally advanced or metastatic carcinoma of the urinary tract: Outcomes by key baseline factors, PD-L1 expression and prior platinum therapy. ESMO Open 2021, 6, 100152. [Google Scholar] [CrossRef]
  114. Patel, M.R.; Ellerton, J.; Infante, J.R.; Agrawal, M.; Gordon, M.; Aljumaily, R.; Britten, C.D.; Dirix, L.; Lee, K.W.; Taylor, M.; et al. Avelumab in metastatic urothelial carcinoma after platinum failure (JAVELIN Solid Tumor): Pooled results from two expansion cohorts of an open-label, phase 1 trial. Lancet Oncol. 2018, 19, 51–64. [Google Scholar] [CrossRef]
  115. Powles, T.; O’Donnell, P.H.; Massard, C.; Arkenau, H.T.; Friedlander, T.W.; Hoimes, C.J.; Lee, J.L.; Ong, M.; Sridhar, S.S.; Vogelzang, N.J.; et al. Efficacy and Safety of Durvalumab in Locally Advanced or Metastatic Urothelial Carcinoma: Updated Results From a Phase 1/2 Open-label Study. JAMA Oncol. 2017, 3, e172411. [Google Scholar] [CrossRef] [PubMed]
  116. Sharma, P.; Callahan, M.K.; Bono, P.; Kim, J.; Spiliopoulou, P.; Calvo, E.; Pillai, R.N.; Ott, P.A.; de Braud, F.; Morse, M.; et al. Nivolumab monotherapy in recurrent metastatic urothelial carcinoma (CheckMate 032): A multicentre, open-label, two-stage, multi-arm, phase 1/2 trial. Lancet Oncol. 2016, 17, 1590–1598. [Google Scholar] [CrossRef] [Green Version]
  117. Sharma, P.; Retz, M.; Siefker-Radtke, A.; Baron, A.; Necchi, A.; Bedke, J.; Plimack, E.R.; Vaena, D.; Grimm, M.O.; Bracarda, S.; et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): A multicentre, single-arm, phase 2 trial. Lancet Oncol. 2017, 18, 312–322. [Google Scholar] [CrossRef]
  118. Grimm, M.O.; Bex, A.; De Santis, M.; Ljungberg, B.; Catto, J.W.F.; Roupret, M.; Hussain, S.A.; Bellmunt, J.; Powles, T.; Wirth, M.; et al. Safe Use of Immune Checkpoint Inhibitors in the Multidisciplinary Management of Urological Cancer: The European Association of Urology Position in 2019. Eur. Urol. 2019, 76, 368–380. [Google Scholar] [CrossRef] [Green Version]
  119. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [Green Version]
  120. Guo, H.; Ingolia, N.T.; Weissman, J.S.; Bartel, D.P. Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature 2010, 466, 835–840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Kozomara, A.; Birgaoanu, M.; Griffiths-Jones, S. miRBase: From microRNA sequences to function. Nucleic Acids Res. 2019, 47, D155–D162. [Google Scholar] [CrossRef]
  122. Esquela-Kerscher, A.; Slack, F.J. Oncomirs—microRNAs with a role in cancer. Nat. Rev. Cancer 2006, 6, 259–269. [Google Scholar] [CrossRef] [PubMed]
  123. Lewis, B.P.; Burge, C.B.; Bartel, D.P. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005, 120, 15–20. [Google Scholar] [CrossRef] [Green Version]
  124. Krol, J.; Loedige, I.; Filipowicz, W. The widespread regulation of microRNA biogenesis, function and decay. Nat. Rev. Genet. 2010, 11, 597–610. [Google Scholar] [CrossRef]
  125. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  126. Rupaimoole, R.; Slack, F.J. MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat. Rev. Drug Discov. 2017, 16, 203–222. [Google Scholar] [CrossRef] [PubMed]
  127. Calin, G.A.; Sevignani, C.; Dumitru, C.D.; Hyslop, T.; Noch, E.; Yendamuri, S.; Shimizu, M.; Rattan, S.; Bullrich, F.; Negrini, M.; et al. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc. Natl. Acad. Sci. USA 2004, 101, 2999–3004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Koturbash, I.; Zemp, F.J.; Pogribny, I.; Kovalchuk, O. Small molecules with big effects: The role of the microRNAome in cancer and carcinogenesis. Mutat. Res. 2011, 722, 94–105. [Google Scholar] [CrossRef] [PubMed]
  129. Onyshchenko, K.V.; Voitsitskyi, T.V.; Grygorenko, V.M.; Saidakova, N.O.; Pereta, L.V.; Onyschuk, A.P.; Skrypkina, I.Y. Expression of micro-RNA hsa-miR-30c-5p and hsa-miR-138-1 in renal cell carcinoma. Exp Oncol. 2020, 42, 115–119. [Google Scholar] [CrossRef] [PubMed]
  130. Persson, H.; Kvist, A.; Rego, N.; Staaf, J.; Vallon-Christersson, J.; Luts, L.; Loman, N.; Jonsson, G.; Naya, H.; Hoglund, M.; et al. Identification of new microRNAs in paired normal and tumor breast tissue suggests a dual role for the ERBB2/Her2 gene. Cancer Res. 2011, 71, 78–86. [Google Scholar] [CrossRef] [Green Version]
  131. Zhang, J.X.; Song, W.; Chen, Z.H.; Wei, J.H.; Liao, Y.J.; Lei, J.; Hu, M.; Chen, G.Z.; Liao, B.; Lu, J.; et al. Prognostic and predictive value of a microRNA signature in stage II colon cancer: A microRNA expression analysis. Lancet Oncol. 2013, 14, 1295–1306. [Google Scholar] [CrossRef]
  132. Xu, B.; Liu, J.; Xiang, X.; Liu, S.; Zhong, P.; Xie, F.; Mou, T.; Lai, L. Expression of miRNA-143 in Pancreatic Cancer and Its Clinical Significance. Cancer Biother. Radiopharm. 2018, 33, 373–379. [Google Scholar] [CrossRef]
  133. Chen, Y.; Min, L.; Ren, C.; Xu, X.; Yang, J.; Sun, X.; Wang, T.; Wang, F.; Sun, C.; Zhang, X. miRNA-148a serves as a prognostic factor and suppresses migration and invasion through Wnt1 in non-small cell lung cancer. PLoS ONE 2017, 12, e0171751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Tian, F.; Li, R.; Chen, Z.; Shen, Y.; Lu, J.; Xie, X.; Ge, Q. Differentially Expressed miRNAs in Tumor, Adjacent, and Normal Tissues of Lung Adenocarcinoma. BioMed Res. Int. 2016, 2016, 1428271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Mahdavinezhad, A.; Mousavibahar, S.H.; Poorolajal, J.; Yadegarazari, R.; Jafari, M.; Shabab, N.; Saidijam, M. Association between tissue miR-141, miR-200c and miR-30b and bladder cancer: A matched case-control study. Urol. J. 2015, 12, 2010–2013. [Google Scholar]
  136. Fendler, A.; Jung, M.; Stephan, C.; Honey, R.J.; Stewart, R.J.; Pace, K.T.; Erbersdobler, A.; Samaan, S.; Jung, K.; Yousef, G.M. miRNAs can predict prostate cancer biochemical relapse and are involved in tumor progression. Int. J. Oncol. 2011, 39, 1183–1192. [Google Scholar] [CrossRef]
  137. Gao, B.; Ning, S.; Li, J.; Liu, H.; Wei, W.; Wu, F.; Tang, Y.; Feng, Y.; Li, K.; Zhang, L. Integrated analysis of differentially expressed mRNAs and miRNAs between hepatocellular carcinoma and their matched adjacent normal liver tissues. Oncol. Rep. 2015, 34, 325–333. [Google Scholar] [CrossRef] [Green Version]
  138. Ramdas, L.; Giri, U.; Ashorn, C.L.; Coombes, K.R.; El-Naggar, A.; Ang, K.K.; Story, M.D. miRNA expression profiles in head and neck squamous cell carcinoma and adjacent normal tissue. Head Neck 2009, 31, 642–654. [Google Scholar] [CrossRef]
  139. Calin, G.A.; Croce, C.M. MicroRNA signatures in human cancers. Nat. Rev. Cancer 2006, 6, 857–866. [Google Scholar] [CrossRef] [PubMed]
  140. Hummel, R.; Hussey, D.J.; Haier, J. MicroRNAs: Predictors and modifiers of chemo- and radiotherapy in different tumour types. Eur. J. Cancer 2010, 46, 298–311. [Google Scholar] [CrossRef]
  141. Lei, L.; Huang, Y.; Gong, W. miR-205 promotes the growth, metastasis and chemoresistance of NSCLC cells by targeting PTEN. Oncol. Rep. 2013, 30, 2897–2902. [Google Scholar] [CrossRef] [Green Version]
  142. Xu, M.; Jin, H.; Xu, C.X.; Bi, W.Z.; Wang, Y. MiR-34c inhibits osteosarcoma metastasis and chemoresistance. Med. Oncol. 2014, 31, 972. [Google Scholar] [CrossRef]
  143. Meng, F.; Henson, R.; Lang, M.; Wehbe, H.; Maheshwari, S.; Mendell, J.T.; Jiang, J.; Schmittgen, T.D.; Patel, T. Involvement of human micro-RNA in growth and response to chemotherapy in human cholangiocarcinoma cell lines. Gastroenterology 2006, 130, 2113–2129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Giovannetti, E.; Funel, N.; Peters, G.J.; Del Chiaro, M.; Erozenci, L.A.; Vasile, E.; Leon, L.G.; Pollina, L.E.; Groen, A.; Falcone, A.; et al. MicroRNA-21 in pancreatic cancer: Correlation with clinical outcome and pharmacologic aspects underlying its role in the modulation of gemcitabine activity. Cancer Res. 2010, 70, 4528–4538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Zhou, M.; Liu, Z.; Zhao, Y.; Ding, Y.; Liu, H.; Xi, Y.; Xiong, W.; Li, G.; Lu, J.; Fodstad, O.; et al. MicroRNA-125b confers the resistance of breast cancer cells to paclitaxel through suppression of pro-apoptotic Bcl-2 antagonist killer 1 (Bak1) expression. J. Biol. Chem. 2010, 285, 21496–21507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Nakajima, G.; Hayashi, K.; Xi, Y.; Kudo, K.; Uchida, K.; Takasaki, K.; Yamamoto, M.; Ju, J. Non-coding MicroRNAs hsa-let-7g and hsa-miR-181b are Associated with Chemoresponse to S-1 in Colon Cancer. Cancer Genomics Proteomics 2006, 3, 317–324. [Google Scholar] [PubMed]
  147. Weiss, G.J.; Bemis, L.T.; Nakajima, E.; Sugita, M.; Birks, D.K.; Robinson, W.A.; Varella-Garcia, M.; Bunn, P.A., Jr.; Haney, J.; Helfrich, B.A.; et al. EGFR regulation by microRNA in lung cancer: Correlation with clinical response and survival to gefitinib and EGFR expression in cell lines. Ann. Oncol. 2008, 19, 1053–1059. [Google Scholar] [CrossRef]
  148. Cai, Z.; Zhang, F.; Chen, W.; Zhang, J.; Li, H. miRNAs: A Promising Target in the Chemoresistance of Bladder Cancer. Onco Targets Ther. 2019, 12, 11805–11816. [Google Scholar] [CrossRef] [Green Version]
  149. Ortiz-Quintero, B. Extracellular MicroRNAs as Intercellular Mediators and Noninvasive Biomarkers of Cancer. Cancers 2020, 12, 3455. [Google Scholar] [CrossRef]
  150. Becker, A.; Thakur, B.K.; Weiss, J.M.; Kim, H.S.; Peinado, H.; Lyden, D. Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis. Cancer Cell 2016, 30, 836–848. [Google Scholar] [CrossRef] [Green Version]
  151. Valadi, H.; Ekstrom, K.; Bossios, A.; Sjostrand, M.; Lee, J.J.; Lotvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef] [Green Version]
  152. Vickers, K.C.; Palmisano, B.T.; Shoucri, B.M.; Shamburek, R.D.; Remaley, A.T. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat. Cell Biol. 2011, 13, 423–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Arroyo, J.D.; Chevillet, J.R.; Kroh, E.M.; Ruf, I.K.; Pritchard, C.C.; Gibson, D.F.; Mitchell, P.S.; Bennett, C.F.; Pogosova-Agadjanyan, E.L.; Stirewalt, D.L.; et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc. Natl. Acad. Sci. USA 2011, 108, 5003–5008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Wang, K.; Zhang, S.; Weber, J.; Baxter, D.; Galas, D.J. Export of microRNAs and microRNA-protective protein by mammalian cells. Nucleic Acids Res. 2010, 38, 7248–7259. [Google Scholar] [CrossRef] [Green Version]
  155. Turchinovich, A.; Weiz, L.; Langheinz, A.; Burwinkel, B. Characterization of extracellular circulating microRNA. Nucleic Acids Res. 2011, 39, 7223–7233. [Google Scholar] [CrossRef] [PubMed]
  156. Chen, X.; Ba, Y.; Ma, L.; Cai, X.; Yin, Y.; Wang, K.; Guo, J.; Zhang, Y.; Chen, J.; Guo, X.; et al. Characterization of microRNAs in serum: A novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res 2008, 18, 997–1006. [Google Scholar] [CrossRef] [Green Version]
  157. Gilad, S.; Meiri, E.; Yogev, Y.; Benjamin, S.; Lebanony, D.; Yerushalmi, N.; Benjamin, H.; Kushnir, M.; Cholakh, H.; Melamed, N.; et al. Serum microRNAs are promising novel biomarkers. PLoS ONE 2008, 3, e3148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Mooney, C.; Raoof, R.; El-Naggar, H.; Sanz-Rodriguez, A.; Jimenez-Mateos, E.M.; Henshall, D.C. High Throughput qPCR Expression Profiling of Circulating MicroRNAs Reveals Minimal Sex- and Sample Timing-Related Variation in Plasma of Healthy Volunteers. PLoS ONE 2015, 10, e0145316. [Google Scholar] [CrossRef] [Green Version]
  159. Hunter, M.P.; Ismail, N.; Zhang, X.; Aguda, B.D.; Lee, E.J.; Yu, L.; Xiao, T.; Schafer, J.; Lee, M.L.; Schmittgen, T.D.; et al. Detection of microRNA expression in human peripheral blood microvesicles. PLoS ONE 2008, 3, e3694. [Google Scholar] [CrossRef] [Green Version]
  160. Piao, X.M.; Jeong, P.; Kim, Y.H.; Byun, Y.J.; Xu, Y.; Kang, H.W.; Ha, Y.S.; Kim, W.T.; Lee, J.Y.; Woo, S.H.; et al. Urinary cell-free microRNA biomarker could discriminate bladder cancer from benign hematuria. Int. J. Cancer 2019, 144, 380–388. [Google Scholar] [CrossRef]
  161. Cinque, A.; Vago, R.; Trevisani, F. Circulating RNA in Kidney Cancer: What We Know and What We Still Suppose. Genes 2021, 12, 835. [Google Scholar] [CrossRef]
  162. Cui, L.; Zhang, X.; Ye, G.; Zheng, T.; Song, H.; Deng, H.; Xiao, B.; Xia, T.; Yu, X.; Le, Y.; et al. Gastric juice MicroRNAs as potential biomarkers for the screening of gastric cancer. Cancer 2013, 119, 1618–1626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Wang, J.; Raimondo, M.; Guha, S.; Chen, J.; Diao, L.; Dong, X.; Wallace, M.B.; Killary, A.M.; Frazier, M.L.; Woodward, T.A.; et al. Circulating microRNAs in Pancreatic Juice as Candidate Biomarkers of Pancreatic Cancer. J. Cancer 2014, 5, 696–705. [Google Scholar] [CrossRef] [Green Version]
  164. Kopkova, A.; Sana, J.; Machackova, T.; Vecera, M.; Radova, L.; Trachtova, K.; Vybihal, V.; Smrcka, M.; Kazda, T.; Slaby, O.; et al. Cerebrospinal Fluid MicroRNA Signatures as Diagnostic Biomarkers in Brain Tumors. Cancers 2019, 11, 1546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Kim, J.E.; Eom, J.S.; Kim, W.Y.; Jo, E.J.; Mok, J.; Lee, K.; Kim, K.U.; Park, H.K.; Lee, M.K.; Kim, M.H. Diagnostic value of microRNAs derived from exosomes in bronchoalveolar lavage fluid of early-stage lung adenocarcinoma: A pilot study. Thorac. Cancer 2018, 9, 911–915. [Google Scholar] [CrossRef]
  166. Watabe, S.; Kikuchi, Y.; Morita, S.; Komura, D.; Numakura, S.; Kumagai-Togashi, A.; Watanabe, M.; Matsutani, N.; Kawamura, M.; Yasuda, M.; et al. Clinicopathological significance of microRNA-21 in extracellular vesicles of pleural lavage fluid of lung adenocarcinoma and its functions inducing the mesothelial to mesenchymal transition. Cancer Med. 2020, 9, 2879–2890. [Google Scholar] [CrossRef] [PubMed]
  167. Xi, Y.; Nakajima, G.; Gavin, E.; Morris, C.G.; Kudo, K.; Hayashi, K.; Ju, J. Systematic analysis of microRNA expression of RNA extracted from fresh frozen and formalin-fixed paraffin-embedded samples. RNA 2007, 13, 1668–1674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Leite, K.R.; Canavez, J.M.; Reis, S.T.; Tomiyama, A.H.; Piantino, C.B.; Sanudo, A.; Camara-Lopes, L.H.; Srougi, M. miRNA analysis of prostate cancer by quantitative real time PCR: Comparison between formalin-fixed paraffin embedded and fresh-frozen tissue. Urol. Oncol. 2011, 29, 533–537. [Google Scholar] [CrossRef]
  169. Mall, C.; Rocke, D.M.; Durbin-Johnson, B.; Weiss, R.H. Stability of miRNA in human urine supports its biomarker potential. Biomark. Med. 2013, 7, 623–631. [Google Scholar] [CrossRef] [Green Version]
  170. Tiberio, P.; Callari, M.; Angeloni, V.; Daidone, M.G.; Appierto, V. Challenges in using circulating miRNAs as cancer biomarkers. BioMed Res. Int. 2015, 2015, 731479. [Google Scholar] [CrossRef]
  171. Kroh, E.M.; Parkin, R.K.; Mitchell, P.S.; Tewari, M. Analysis of circulating microRNA biomarkers in plasma and serum using quantitative reverse transcription-PCR (qRT-PCR). Methods 2010, 50, 298–301. [Google Scholar] [CrossRef] [Green Version]
  172. Cheng, H.H.; Yi, H.S.; Kim, Y.; Kroh, E.M.; Chien, J.W.; Eaton, K.D.; Goodman, M.T.; Tait, J.F.; Tewari, M.; Pritchard, C.C. Plasma processing conditions substantially influence circulating microRNA biomarker levels. PLoS ONE 2013, 8, e64795. [Google Scholar] [CrossRef] [Green Version]
  173. Moret, I.; Sanchez-Izquierdo, D.; Iborra, M.; Tortosa, L.; Navarro-Puche, A.; Nos, P.; Cervera, J.; Beltran, B. Assessing an improved protocol for plasma microRNA extraction. PLoS ONE 2013, 8, e82753. [Google Scholar] [CrossRef]
  174. Willems, M.; Moshage, H.; Nevens, F.; Fevery, J.; Yap, S.H. Plasma collected from heparinized blood is not suitable for HCV-RNA detection by conventional RT-PCR assay. J. Virol. Methods 1993, 42, 127–130. [Google Scholar] [CrossRef]
  175. Garcia, M.E.; Blanco, J.L.; Caballero, J.; Gargallo-Viola, D. Anticoagulants interfere with PCR used to diagnose invasive aspergillosis. J. Clin. Microbiol. 2002, 40, 1567–1568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Kim, D.J.; Linnstaedt, S.; Palma, J.; Park, J.C.; Ntrivalas, E.; Kwak-Kim, J.Y.; Gilman-Sachs, A.; Beaman, K.; Hastings, M.L.; Martin, J.N.; et al. Plasma components affect accuracy of circulating cancer-related microRNA quantitation. J. Mol. Diagn. 2012, 14, 71–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Pritchard, C.C.; Kroh, E.; Wood, B.; Arroyo, J.D.; Dougherty, K.J.; Miyaji, M.M.; Tait, J.F.; Tewari, M. Blood cell origin of circulating microRNAs: A cautionary note for cancer biomarker studies. Cancer Prev. Res. 2012, 5, 492–497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Kirschner, M.B.; Kao, S.C.; Edelman, J.J.; Armstrong, N.J.; Vallely, M.P.; van Zandwijk, N.; Reid, G. Haemolysis during sample preparation alters microRNA content of plasma. PLoS ONE 2011, 6, e24145. [Google Scholar] [CrossRef] [PubMed]
  179. Wang, K.; Yuan, Y.; Cho, J.H.; McClarty, S.; Baxter, D.; Galas, D.J. Comparing the MicroRNA spectrum between serum and plasma. PLoS ONE 2012, 7, e41561. [Google Scholar] [CrossRef]
  180. McDonald, J.S.; Milosevic, D.; Reddi, H.V.; Grebe, S.K.; Algeciras-Schimnich, A. Analysis of circulating microRNA: Preanalytical and analytical challenges. Clin. Chem. 2011, 57, 833–840. [Google Scholar] [CrossRef] [Green Version]
  181. Schmittgen, T.D.; Lee, E.J.; Jiang, J.; Sarkar, A.; Yang, L.; Elton, T.S.; Chen, C. Real-time PCR quantification of precursor and mature microRNA. Methods 2008, 44, 31–38. [Google Scholar] [CrossRef] [Green Version]
  182. de Boer, H.C.; van Solingen, C.; Prins, J.; Duijs, J.M.; Huisman, M.V.; Rabelink, T.J.; van Zonneveld, A.J. Aspirin treatment hampers the use of plasma microRNA-126 as a biomarker for the progression of vascular disease. Eur. Heart J. 2013, 34, 3451–3457. [Google Scholar] [CrossRef]
  183. Badrnya, S.; Baumgartner, R.; Assinger, A. Smoking alters circulating plasma microvesicle pattern and microRNA signatures. Thromb. Haemost. 2014, 112, 128–136. [Google Scholar] [CrossRef] [Green Version]
  184. Ross, S.A.; Davis, C.D. MicroRNA, nutrition, and cancer prevention. Adv. Nutr. 2011, 2, 472–485. [Google Scholar] [CrossRef]
  185. Aoi, W.; Sakuma, K. Does regulation of skeletal muscle function involve circulating microRNAs? Front. Physiol. 2014, 5, 39. [Google Scholar] [CrossRef]
  186. Fouad, Y.A.; Aanei, C. Revisiting the hallmarks of cancer. Am. J. Cancer Res. 2017, 7, 1016–1036. [Google Scholar]
  187. Ying, L.; Du, L.; Zou, R.; Shi, L.; Zhang, N.; Jin, J.; Xu, C.; Zhang, F.; Zhu, C.; Wu, J.; et al. Development of a serum miRNA panel for detection of early stage non-small cell lung cancer. Proc. Natl. Acad. Sci. USA 2020, 117, 25036–25042. [Google Scholar] [CrossRef] [PubMed]
  188. Yokoi, A.; Matsuzaki, J.; Yamamoto, Y.; Yoneoka, Y.; Takahashi, K.; Shimizu, H.; Uehara, T.; Ishikawa, M.; Ikeda, S.I.; Sonoda, T.; et al. Integrated extracellular microRNA profiling for ovarian cancer screening. Nat. Commun. 2018, 9, 4319. [Google Scholar] [CrossRef]
  189. Urabe, F.; Matsuzaki, J.; Yamamoto, Y.; Kimura, T.; Hara, T.; Ichikawa, M.; Takizawa, S.; Aoki, Y.; Niida, S.; Sakamoto, H.; et al. Large-scale Circulating microRNA Profiling for the Liquid Biopsy of Prostate Cancer. Clin. Cancer Res. 2019, 25, 3016–3025. [Google Scholar] [CrossRef] [Green Version]
  190. Zhang, J.; Song, Y.; Zhang, C.; Zhi, X.; Fu, H.; Ma, Y.; Chen, Y.; Pan, F.; Wang, K.; Ni, J.; et al. Circulating MiR-16-5p and MiR-19b-3p as Two Novel Potential Biomarkers to Indicate Progression of Gastric Cancer. Theranostics 2015, 5, 733–745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Zhou, C.; Chen, Z.; Dong, J.; Li, J.; Shi, X.; Sun, N.; Luo, M.; Zhou, F.; Tan, F.; He, J. Combination of serum miRNAs with Cyfra21-1 for the diagnosis of non-small cell lung cancer. Cancer Lett. 2015, 367, 138–146. [Google Scholar] [CrossRef] [PubMed]
  192. Min, L.; Zhu, S.; Chen, L.; Liu, X.; Wei, R.; Zhao, L.; Yang, Y.; Zhang, Z.; Kong, G.; Li, P.; et al. Evaluation of circulating small extracellular vesicles derived miRNAs as biomarkers of early colon cancer: A comparison with plasma total miRNAs. J. Extracell. Vesicles 2019, 8, 1643670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Verhoest, G.; Shariat, S.F.; Chromecki, T.F.; Raman, J.D.; Margulis, V.; Novara, G.; Seitz, C.; Remzi, M.; Roupret, M.; Scherr, D.S.; et al. Predictive factors of recurrence and survival of upper tract urothelial carcinomas. World J. Urol. 2011, 29, 495–501. [Google Scholar] [CrossRef] [Green Version]
  194. Kriebel, S.; Schmidt, D.; Holdenrieder, S.; Goltz, D.; Kristiansen, G.; Moritz, R.; Fisang, C.; Muller, S.C.; Ellinger, J. Analysis of tissue and serum microRNA expression in patients with upper urinary tract urothelial cancer. PLoS ONE 2015, 10, e0117284. [Google Scholar] [CrossRef] [PubMed]
  195. Cavallari, I.; Ciccarese, F.; Sharova, E.; Urso, L.; Raimondi, V.; Silic-Benussi, M.; D’Agostino, D.M.; Ciminale, V. The miR-200 Family of microRNAs: Fine Tuners of Epithelial-Mesenchymal Transition and Circulating Cancer Biomarkers. Cancers 2021, 13, 5874. [Google Scholar] [CrossRef] [PubMed]
  196. Gao, Y.; Feng, B.; Han, S.; Zhang, K.; Chen, J.; Li, C.; Wang, R.; Chen, L. The Roles of MicroRNA-141 in Human Cancers: From Diagnosis to Treatment. Cell Physiol. Biochem. 2016, 38, 427–448. [Google Scholar] [CrossRef] [PubMed]
  197. Zaravinos, A.; Lambrou, G.I.; Mourmouras, N.; Katafygiotis, P.; Papagregoriou, G.; Giannikou, K.; Delakas, D.; Deltas, C. New miRNA profiles accurately distinguish renal cell carcinomas and upper tract urothelial carcinomas from the normal kidney. PLoS ONE 2014, 9, e91646. [Google Scholar] [CrossRef]
  198. Cosyns, J.P.; Jadoul, M.; Squifflet, J.P.; Van Cangh, P.J.; van Ypersele de Strihou, C. Urothelial malignancy in nephropathy due to Chinese herbs. Lancet 1994, 344, 188. [Google Scholar] [CrossRef]
  199. Tao, L.; Zeng, Y.; Wang, J.; Liu, Z.; Shen, B.; Ge, J.; Liu, Y.; Guo, Y.; Qiu, J. Differential microRNA expression in aristolochic acid-induced upper urothelial tract cancers ex vivo. Mol. Med. Rep. 2015, 12, 6533–6546. [Google Scholar] [CrossRef] [Green Version]
  200. Eales, K.L.; Hollinshead, K.E.; Tennant, D.A. Hypoxia and metabolic adaptation of cancer cells. Oncogenesis 2016, 5, e190. [Google Scholar] [CrossRef] [Green Version]
  201. Muz, B.; de la Puente, P.; Azab, F.; Azab, A.K. The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy. Hypoxia (Auckl) 2015, 3, 83–92. [Google Scholar] [CrossRef] [Green Version]
  202. Al Tameemi, W.; Dale, T.P.; Al-Jumaily, R.M.K.; Forsyth, N.R. Hypoxia-Modified Cancer Cell Metabolism. Front. Cell Dev. Biol. 2019, 7, 4. [Google Scholar] [CrossRef] [Green Version]
  203. Ke, H.L.; Li, W.M.; Lin, H.H.; Hsu, W.C.; Hsu, Y.L.; Chang, L.L.; Huang, C.N.; Li, C.C.; Chang, H.P.; Yeh, H.C.; et al. Hypoxia-regulated MicroRNA-210 Overexpression is Associated with Tumor Development and Progression in Upper Tract Urothelial Carcinoma. Int. J. Med. Sci. 2017, 14, 578–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Huang, X.; Ding, L.; Bennewith, K.L.; Tong, R.T.; Welford, S.M.; Ang, K.K.; Story, M.; Le, Q.T.; Giaccia, A.J. Hypoxia-inducible mir-210 regulates normoxic gene expression involved in tumor initiation. Mol. Cell 2009, 35, 856–867. [Google Scholar] [CrossRef] [Green Version]
  205. el-Maraghy, S.S. Aflatoxins and fungal flora in lentil (Lens esculenta L.). Mycopathologia 1988, 102, 31–35. [Google Scholar] [CrossRef]
  206. Ullmann, P.; Qureshi-Baig, K.; Rodriguez, F.; Ginolhac, A.; Nonnenmacher, Y.; Ternes, D.; Weiler, J.; Gabler, K.; Bahlawane, C.; Hiller, K.; et al. Hypoxia-responsive miR-210 promotes self-renewal capacity of colon tumor-initiating cells by repressing ISCU and by inducing lactate production. Oncotarget 2016, 7, 65454–65470. [Google Scholar] [CrossRef] [Green Version]
  207. Liu, L.L.; Li, D.; He, Y.L.; Zhou, Y.Z.; Gong, S.H.; Wu, L.Y.; Zhao, Y.Q.; Huang, X.; Zhao, T.; Xu, L.; et al. miR-210 protects renal cell against hypoxia-induced apoptosis by targeting HIF-1 alpha. Mol. Med. 2017, 23, 258–271. [Google Scholar] [CrossRef] [Green Version]
  208. Hsu, W.C.; Li, W.M.; Lee, Y.C.; Huang, A.M.; Chang, L.L.; Lin, H.H.; Wu, W.J.; Li, C.C.; Liang, P.I.; Ke, H.L. MicroRNA-145 suppresses cell migration and invasion in upper tract urothelial carcinoma by targeting ARF6. FASEB J. 2020, 34, 5975–5992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Li, H.; Zhao, S.; Chen, X.; Feng, G.; Chen, Z.; Fan, S. MiR-145 modulates the radiosensitivity of non-small cell lung cancer cells by suppression of TMOD3. Carcinogenesis, 1093; -9. [Google Scholar] [CrossRef]
  210. Zhang, X.; Wu, J. Prognostic role of microRNA-145 in prostate cancer: A systems review and meta-analysis. Prostate Int. 2015, 3, 71–74. [Google Scholar] [CrossRef] [Green Version]
  211. Xia, X.; Yang, B.; Zhai, X.; Liu, X.; Shen, K.; Wu, Z.; Cai, J. Prognostic role of microRNA-21 in colorectal cancer: A meta-analysis. PLoS ONE 2013, 8, e80426. [Google Scholar] [CrossRef]
  212. Tang, W.; Zhang, X.; Tan, W.; Gao, J.; Pan, L.; Ye, X.; Chen, L.; Zheng, W. miR-145-5p Suppresses Breast Cancer Progression by Inhibiting SOX2. J. Surg. Res. 2019, 236, 278–287. [Google Scholar] [CrossRef] [PubMed]
  213. Zhang, Y.; Ta, W.W.; Sun, P.F.; Meng, Y.F.; Zhao, C.Z. Diagnostic and prognostic significance of serum miR-145-5p expression in glioblastoma. Int. J. Clin Exp Pathol 2019, 12, 2536–2543. [Google Scholar]
  214. Aftab, M.; Poojary, S.S.; Seshan, V.; Kumar, S.; Agarwal, P.; Tandon, S.; Zutshi, V.; Das, B.C. Urine miRNA signature as a potential non-invasive diagnostic and prognostic biomarker in cervical cancer. Sci. Rep. 2021, 11, 10323. [Google Scholar] [CrossRef]
  215. Zaoui, K.; Smith, H.W.; Park, M.; Duhamel, S. ARF6 controls RHOB targeting to endosomes regulating cancer cell invasion. Mol Cell Oncol. 2020, 7, 1766932. [Google Scholar] [CrossRef]
  216. Sabe, H. Requirement for Arf6 in cell adhesion, migration, and cancer cell invasion. J. Biochem. 2003, 134, 485–489. [Google Scholar] [CrossRef] [PubMed]
  217. Baraniskin, A.; Birkenkamp-Demtroder, K.; Maghnouj, A.; Zollner, H.; Munding, J.; Klein-Scory, S.; Reinacher-Schick, A.; Schwarte-Waldhoff, I.; Schmiegel, W.; Hahn, S.A. MiR-30a-5p suppresses tumor growth in colon carcinoma by targeting DTL. Carcinogenesis 2012, 33, 732–739. [Google Scholar] [CrossRef] [PubMed]
  218. Wei, W.; Yang, Y.; Cai, J.; Cui, K.; Li, R.X.; Wang, H.; Shang, X.; Wei, D. MiR-30a-5p Suppresses Tumor Metastasis of Human Colorectal Cancer by Targeting ITGB3. Cell Physiol. Biochem. 2016, 39, 1165–1176. [Google Scholar] [CrossRef]
  219. Zhang, W.; Zhang, C.; Chen, H.; Li, L.; Tu, Y.; Liu, C.; Shi, S.; Zen, K.; Liu, Z. Evaluation of microRNAs miR-196a, miR-30a-5P, and miR-490 as biomarkers of disease activity among patients with FSGS. Clin. J. Am. Soc. Nephrol. 2014, 9, 1545–1552. [Google Scholar] [CrossRef] [Green Version]
  220. Wang, L.L.; Zhang, X.H.; Zhang, X.; Chu, J.K. MiR-30a increases cisplatin sensitivity of gastric cancer cells through suppressing epithelial-to-mesenchymal transition (EMT). Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 1733–1739. [Google Scholar] [PubMed]
  221. Chang, C.W.; Yu, J.C.; Hsieh, Y.H.; Yao, C.C.; Chao, J.I.; Chen, P.M.; Hsieh, H.Y.; Hsiung, C.N.; Chu, H.W.; Shen, C.Y.; et al. MicroRNA-30a increases tight junction protein expression to suppress the epithelial-mesenchymal transition and metastasis by targeting Slug in breast cancer. Oncotarget 2016, 7, 16462–16478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Chung, Y.H.; Li, S.C.; Kao, Y.H.; Luo, H.L.; Cheng, Y.T.; Lin, P.R.; Tai, M.H.; Chiang, P.H. MiR-30a-5p Inhibits Epithelial-to-Mesenchymal Transition and Upregulates Expression of Tight Junction Protein Claudin-5 in Human Upper Tract Urothelial Carcinoma Cells. Int. J. Mol. Sci. 2017, 18, 1826. [Google Scholar] [CrossRef] [Green Version]
  223. Ikenouchi, J.; Matsuda, M.; Furuse, M.; Tsukita, S. Regulation of tight junctions during the epithelium-mesenchyme transition: Direct repression of the gene expression of claudins/occludin by Snail. J. Cell Sci. 2003, 116, 1959–1967. [Google Scholar] [CrossRef] [Green Version]
  224. Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2014, 15, 178–196. [Google Scholar] [CrossRef] [Green Version]
  225. Izquierdo, L.; Ingelmo-Torres, M.; Mallofre, C.; Lozano, J.J.; Verhasselt-Crinquette, M.; Leroy, X.; Colin, P.; Comperat, E.; Roupret, M.; Alcaraz, A.; et al. Prognostic value of microRNA expression pattern in upper tract urothelial carcinoma. BJU Int. 2014, 113, 813–821. [Google Scholar] [CrossRef] [Green Version]
  226. Montalbo, R.; Izquierdo, L.; Ingelmo-Torres, M.; Lozano, J.J.; Capitan, D.; Alcaraz, A.; Mengual, L. Prognostic value of circulating microRNAs in upper tract urinary carcinoma. Oncotarget 2018, 9, 16691–16700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Soualhi, A.; Rammant, E.; George, G.; Russell, B.; Enting, D.; Nair, R.; Van Hemelrijck, M.; Bosco, C. The incidence and prevalence of upper tract urothelial carcinoma: A systematic review. BMC Urol 2021, 21, 110. [Google Scholar] [CrossRef]
  228. Di Giovanni, P.; Di Martino, G.; Scampoli, P.; Cedrone, F.; Meo, F.; Lucisano, G.; Romano, F.; Staniscia, T. Arsenic Exposure and Risk of Urothelial Cancer: Systematic Review and Meta-Analysis. Int. J. Environ. Res. Public Health 2020, 17, 3105. [Google Scholar] [CrossRef] [PubMed]
  229. Blaszyk, H.; Wang, L.; Dietmaier, W.; Hofstadter, F.; Burgart, L.J.; Cheville, J.C.; Hartmann, A. Upper tract urothelial carcinoma: A clinicopathologic study including microsatellite instability analysis. Mod. Pathol. 2002, 15, 790–797. [Google Scholar] [CrossRef]
  230. Popovska-Jankovic, K.; Noveski, P.; Jankovic-Velickovic, L.; Stojnev, S.; Cukuranovic, R.; Stefanovic, V.; Toncheva, D.; Staneva, R.; Polenakovic, M.; Plaseska-Karanfilska, D. MicroRNA Profiling in Patients with Upper Tract Urothelial Carcinoma Associated with Balkan Endemic Nephropathy. Biomed Res. Int. 2016, 2016, 7450461. [Google Scholar] [CrossRef] [Green Version]
  231. Tao, J.; Yang, X.; Li, P.; Wei, J.; Deng, X.; Cheng, Y.; Qin, C.; Ju, X.; Meng, X.; Li, J.; et al. Identification of circulating microRNA signatures for upper tract urothelial carcinoma detection. Mol. Med. Rep. 2015, 12, 6752–6760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Browne, B.M.; Stensland, K.D.; Patel, C.K.; Sullivan, T.; Burks, E.J.; Canes, D.; Raman, J.D.; Warrick, J.; Reiger-Christ, K.M. MicroRNA Expression Profiles in Upper Tract Urothelial Carcinoma Differentiate Tumor Grade, Stage, and Survival: Implications for Clinical Decision-Making. Urology 2019, 123, 93–100. [Google Scholar] [CrossRef]
Figure 1. Diversity of miRNA Targets in UTUC. Examples of miRNAs identified in studies on UTUC biomarker screening. In most cases, the candidate miRNAs alter numerous genes and have pleotropic actions in normal and cancerous cells. The downstream actions presented represent some of the most common actions of these miRNAs.
Figure 1. Diversity of miRNA Targets in UTUC. Examples of miRNAs identified in studies on UTUC biomarker screening. In most cases, the candidate miRNAs alter numerous genes and have pleotropic actions in normal and cancerous cells. The downstream actions presented represent some of the most common actions of these miRNAs.
Ijms 23 02602 g001
Table 1. miRNAs as diagnostic or prognostic biomarkers of UTUC.
Table 1. miRNAs as diagnostic or prognostic biomarkers of UTUC.
ReferenceBiomarker or Panel of Biomarkers and LevelsSample TypeCollection Processing (Sample Processing, Storage Condition)Study Design:
Retrospective–Prospective;
Monocenter-Multicenter
Discovery
Cohort/Validation Cohort, Histotypes
Participant
Characteristics, TNM Stage and Fuhrman Grade
Evaluation Method (RNA Extraction, Retrotrascription, and Detection Technology) and Normalization StrategyDiagnostic/Prognostic Value and Statistical Analysis/Results
[194]Serum miRNAs.
↑ miR-141 (UTUC patients vs. controls);
↓ miR-10a, ↓ miR135 in muscle-invasive UTUC (pTa/pT1 vs. pT2–4).
Tissue miRNAs.
↑ miR-21, ↑ miR-96, ↑ miR-135, ↑ miR-141,
↑ miR-182, ↑ miR-205, ↑ miR-429, miR-520b, ↑ miR-10a, ↑ miR-200b (in UTUC tissue vs. normal tissue);
↑ miR-205 in undifferentiated UTUC (G3 vs. G2/G1).
Blood;
Tissue samples
Pre-operative blood samples centrifuged at 2500× g × 10 min and stored in cryotubules at −80 °C until use;
Formalin-fixed,
paraffin-embedded tissue.
Prospective;
multicenter (serum cohort), monocenter (tissue cohort)
DC: Serum cohort: 44 UTUC, 34 controls.
DC: Tissue cohort: 47 UTUC, 36 controls.
Serum cohort.
UTUC: Age: 67.9 y; Gender: 17 female; TNM: pTa/pT1 25 pts, pT2-pT4 19 pts; Grading: G1 6 pts, G2 28 pts, G3 10 pts.
Control: Age: 63.5 y; Gender: 11 female;
Tissue cohort.
UTUC: Age: 68.9 y; Gender: 21 female; TNM: pTa/pT1 16 pts, pT2-pT4 31 pts; Grading: G1 9 pts, G2 20 pts, G3 18 pts.
Control: Age: 69.1 y; Gender: 14 female;
Total RNA isolated from serum samples with mirVana PARIS Kit (Life Technologies) Total RNA extracted from tissues with the RecoverAll Total Nucleic Acid Isolation Kit (Life Technologies).
Retrotrascription with miScript II RT Kit (Qiagen), qPCR using miScript SYBR Green PCR Kit (Qiagen). Tissue-miRNAs normalized against RNU1-4, SNORD43, and SNORD48, serum-miRNAs normalized against RNU1-4 and SNORD43
Diagnostic. Serum miR-141: AUC = 0.726 (70.5% sensitivity and 73.5% specificity)
Prognostic. Serum miR-10a (p = 0.003) and miR-135 (p = 0.040)
[197]↑miR-3648, ↑ miR-3656, ↑ miR-3687 (UTUC vs. normal kidney tissue)Tissue samplesDC: Formalin-fixed, paraffin-embedded tissue samples
VC: freshly frozen tissue samples
Retrospective,
monocenter
DC: 24 ccRCC, 5 papRCC, 3 chRCC, 5 UTUC, 40 normal tissue samples
VC: 10 ccrCC, 3 papRCC, 4 chRCC, 3 UTUC, 20 normal kidney samples
DC: ccRCC: Age: 70 y; Gender: 4 female; Stage: 13 T1, 6 T2, 5 T3, 0 T4, 2 metastatic samples; Grade: 0 G1, 12 G2, 9 G4, 3 G4.
papRCC: Age: 67 y; Gender: 1 female; Stage: 4 T1, 1 T2, 0 T3, 0 T4; Grade: 0 G1, 2 G2, 3 G3, 0 G4.
chRCC: Age: 68 y; Gender: 3 female; Stage: 0 T1, 3 T2, 0 T3, 0 T4, 0 metastatic; Grade: 0 G1, 1 G2, 2 G3, 0 G4.
UTUC: Age: 76 y; Gender: 1 female; Stage: 3 T1, 1 T2, 1 T3, 0 T4, 0 metastatic; Grade: 0 G1, 1 G2, 3 G3, 0 G4.
Total RNA extracted from FFPE tissue samples with High Pure FFPE RNA Micro Kit (Roche Applied Science); Total RNA extracted from frozen tissue samples with mirVANA miRNA Isolation Kit (Ambion).
MicroRNA microarray analysis with miRCURY LNA microRNA Array, 6th gen (Exiqon).
Retrotranscription using miRCURY LNA Universal RT cDNA synthesis kit (Exiqon). qPCR using SYBR Green master mix (Exiqon, Vedbaek, Denmark) and LNA microRNA-specific primers on a ViiA 7 Real-Time PCR System (Applied Biosystems) and SNORA66, U6snRNA, RNU1A1 as endogenous reference genes for normalization.
Diagnostic. miR-3648, miR-3656, miR-3687 discriminated UTUC from normal kidney tissue with a median AUC > 0.94, p < 0.001
[199]↑ miR-488, ↓ miR-181c (AAN-UTUC vs. non-AAN-UTUC);
↑ miR-488 (stage I-II vs. stage III-IV AAN-UTUC);
↑ miR-181 (>3 cm vs. <3 cm AAN-UUC)
Tissue samplesFormalin-fixed, paraffin-embedded tissue samplesRetrospective,
Monocenter
20 cases of AAN-UTUC and 20 controls (non AAN-UTUC)AAN-group: Age: 63.9 ± 1.64 y; Gender: 11 female; Stage: 9 stage I–II, 11 stage III–IV; Tumor differentiation: 4 well, 7 moderate, 9 poor.
Non AAN-group: Age: 65.6 ± 1.66 y; Gender: 7 female; Stage: 7 stage I–II, 13 stage III–IV; Tumor differentiation: 9 well, 5 moderate, 6 poor.
RNA isolation using mirVanaTM miRNA isolation kit (Life Technologies); miRNA microarray analysis using Affymetrix GeneChip miRNA arrays (Affymetrix); reverse transcription using a stem-loop RT primer; qPCR reaction with TaqMan PCR Master Mix-UNG using an ABI 7900 HT real-time PCR system (Applied Biosystems).Diagnostic.
miR-488 and miR-181c: AAN-UTUC vs. non-AAN-UTUC (p < 0.05);
Prognostic.
miR-488: stage I–II vs. stage III–IV AAN-UUC (p = 0.038); miR-181c: >3 cm vs. <3 cm AAN-UUC (p = 0.049).
[203]↑ miR-210 (UTUC tissue vs. normal tissue); high-stage and high-grade tumors vs. low-stage and low-grade tumors)Tissue samplesFreshly frozen tissue samplesRetrospective,
monocenter
A total of 83 surgically removed UTUC cases provided 83 UTUC tissue samples and 50 paired normal urothelium tissue samplesAll cohort: Age: 42 pts <70 y and 41 pts ≥70 y pts; Gender: 48 females; Stage: 50 organ-confined (T1–T2), 33 locally advanced (T3–T4); Grade: 11 Low, 72 HighTotal RNA extracted with Quick-RNA™ MiniPrep Kit (Zymo research, Reverse-transcription using miR-210-specific stem-loop primer; qPCR using TaqMan miRNA assay (Applied Biosystems) on 7500HT Fast Real Time PCR System (Applied Biosystems). RNU6B as endogenous reference gene for normalization.Diagnostic.
UTUC from non-cancerous
Urothelium with an AUC of 0.904 (95% CI = 0.843–0.865, p < 0.001), with 80% sensitivity and 90% specificity;
Prognostic:
high stage vs. low stage, p = 0.02; high grade vs. low grade, p = 0.049
[208]↓ miR-145-5p
(UTUC vs. normal urothelium tissues)
Tissue samplesFormalin-fixed, paraffin-embedded tissue samplesRetrospective, monocenter114 UTUC samples and 40 paired non tumoral urothelium samples.
Subsequent focal analysis of miR-145-5p was restricted to 61 pairs of UTUC samples and normal tumor-adjacent tissue samples
DC. Age: 41 pts <65y, 73 pts ≥65y; Gender: 65 females; Stage: 75 T1–T2, 39 T3–T4; Grade: 24 low, 90 high.Total RNA extracted with TRIzol reagent (Invitrogen); reverse transcription with TaqMan MicroRNA Reverse Transcription Kit (Applied Biosystem). qPCR using TaqMan MicroRNA Assays on StepOnePlus Real-Time PCR System (Applied Biosystems). U6 as endogenous reference gene for normalization.Diagnosis. UTUC vs. normal urothelium tissues (p < 0.001).
[222]↓ miR-30a-5p
(UTUC tissues vs. normal tissues)
Tissue samplesFresh tissues were immediately immersed in RNA later (Qiagen) after nephroureterectomy, stored at 4 °C overnight, and then stored at −80 °CRetrospective, monocenter22 UTUC pts (renal pelvis) who underwent nephroureterectomy without neoadjuvant therapy provided 22 UTUC tissue samples and 14 Adjacent non-tumoral tissue collected from non-cancer areas (by gross appearance) of renal pelvis mucosaDC. Age: 4 < 60y, 18 ≥ 60 y; Gender: 13 female; Stage: 12 T1-T2, 10 T3-T4, 15 N0, 7 N1, 9 M0, 13 M1; Grade: 5 Low, 17 High.Total RNA extracted with TRIzol Reagent (Invitrogen); reverse transcription with TaqMan MicroRNA reverse transcription kit (ABI); qPCR with TaqMan microRNA assays using Brilliant III QPCR Master Mixes with low ROX on AriaMx Real-Time PCR system (Agilent). U6 as endogenous reference gene for normalization.miR-30a-5p is significantly downregulated in UTUC samples (p < 0.001).
[225]↑ miRNA-31,↑ miRNA-149 (non-progressing UTUC vs. progressing UTUCTissue samplesFormalin-fixed paraffin-embedded tissue samplesRetrospective, multicenterA total of 150 patients with RNU-treated UTUCTotal cohort: Age: 70 y; Gender: 34 female; Stage: 26 pTa, 42 pT1, 28 pT2, 38 pT3, 16 pT4; Grade: 14 G1, 48 G2, 88 G3.
DC: 18 randomly selected UTUC cases (9 non-progressing UTUC and 9 progressing UTUC)
VC: remaining 132 UTUC pts
Total RNA isolation using the RecoverAll Total Nucleic Acid Isolation Kit (Ambion). Screening Phase: Reverse transcription with TaqMan miRNA reverse transcription kit with Megaplex RT primers for Human Pool A and B (Applied Biosystems). Pre-Amplification with Megaplex PreAmp primers (Applied Biosystems); qPCR with TaqMan® Human MicroRNA Array A + B Cards Set v2.0 on ABI7900HT Real-Time PCR system (Applied Biosystems) and global mean as normalization method.
Validation phases:
RT-qPCR with miRCURY Locked Nucleic Acid kit (Exiqon) on ABI7900HT Real-Time PCR system (Applied Biosystems). hsa-miR-218 as endogenous reference gene for normalization.
Prognosis. miR-31 and miR-149 were independent prognostic factors of tumor progression (HR 0.88, p < 0.001 and HR 0.78, p < 0.001, respectively); miR-149 expression was an independent prognostic factor of cancer-specific survival (HR 0.82; p = 0.018).
[226]↓ miR-151b
(Progressing vs. non-progressing
UTUC pts)
Serum samplesPre-operative blood samples collected in a BD vacutainer sterile tube coated with silicone and micronized silica particles, left to clot for a minimum of one hour and, within four hours, centrifuged for 15 min at 3500 rpm at 4 °C; serum transferred to a cryotube and stored at −80 °C.Prospective, monocenterA total of 33 pts with UTUC were analyzed in two phases,
DC. 5 progressing and 7 non-progressing.
VC: 8 progressing and 13 non-progressing UTUC pts.
Total cohort. Age: 70 y; Gender: 10 female; Stage: 7 pTa, 5 pT1, 7 pT2, 12 pT3, 2 pT4; Grade: 6 Low, 27 High.
DC: progressing pts: Stage and Grade: 1 pT2, 3 pT3 and 1 pT4, all high grade;
non-progressing: Stage and Grade: 2 pTa, 1 pT2 and 4 pT3, all high-grade.
VC: progressing pts: Stage and Grade: 1 pTa low grade, 1 pT1 low-grade, 2 pT2 high grade, 3 pT3 high grade and 1 pT4 high grade; non-progressing: Stage and Grade: 4 pTa low grade, 4 pT1 high grade, 3 pT2 high grade and 2 pT3 high grade).
Total RNA extracted with mirVana PARIS Kit (Thermosfisher Scientific), miRNAs expression profiling using nCounter Human v3 miRNA expression Assay Kit (NanoString Technologies). hsa-miR-16-5p, hsa-miR-484, hsa-miR-126-3p, hsa-miR-191-5p, hsa-miR-93-5p and hsa-miR-24-3p as endogenous reference genes for normalization.Prognosis. miR-151b prognostic factors for tumor progression (HR = 0.33, p < 0.001) and cancer specific survival (HR = 0.25, p < 0.001).
[230]hsa-miR-205-5p, ↑ hsa-miR-4322, ↑ hsa-miR-99b-3p, ↑ hsa-miR-3620-3p, ↑ hsa-miR-373-5p, ↑ hsa-miR-3656, ↑ hsa-miR-1290, ↓ hsa-miR-30a-5p, ↓ hsa-miR-127-3p and ↓ hsa-miR-154-5p in BEN-UTUC vs. normal kidney tissue;
hsa-miR-205-5p, ↑ hsa-miR-205-3p, ↑ has-miR-224-5p, ↑ hsa-miR-224-3p, ↑ hsa-miR-197-3p, ↑ hsa-miR-182-5p, ↑ hsa-miR-183-5p, ↑ hsa-miR-96-5p, ↑ hsa-miR-203a-3p, ↑ hsa-miR-149-5p, ↑ hsa-miR-141-3p, ↑ hsa-miR-200c-3p, ↑ hsa-miR-1260a, ↑ hsa-miR-210-3p, ↓ hsa-miR-663b
in non-BEN-UTUC vs. normal kidney tissue
Tissue samplesFormalin-fixed paraffine embedded tissue samplesRetrospective, monocenter7 BEN-UTUC, 5 non-BEN-UTUC and 8 normal kidney tissues.DC. BEN-UTUC: Age: 66 y; Gender: 2 female; Stage: 4 Low, 2 high, 1 unknown; Grade: 4 low, 2 high, 1 unknown;
Non-BEN-UTUC: Age: 62.8 y; Gender: 3 female; Stage: 3 low, 2 high; Grade: 1 low, 4 high.
Normal kidney tissue samples: unspecified clinical–pathological features.
Total RNA extraction with FFPE DNA/RNA Kit (Qiagen).
miRNAs expression profiling: “miRNA Microarray System with miRNA Complete Labeling and Hyb Kit,” version 2.4 with Agilent Sure Print G3 Human v16 miRNA Array Kit (Agilent Technologies);
RT-qPCR Validation: reverse transcription with TaqMan_ MicroRNA Reverse Transcription Kit (Life Technologies); qPCR with TaqMan Universal PCR Master Mix and TaqMAN MicroRNA Assays (Life Technologies).
Diagnostic.
BEN-UTUC vs. normal kidney tissue, p < 0.05;
non-BEN-UTUC vs. normal kidney tissue, p < 0.05.
[231]↑ miR-664a-3p
↑ miR-431-5p ↑ miR-423-5p ↑ miR-191-5p
↑ miR-33b-3p ↑ miR-26a-5p ↑ miR-22-3p ↑ miR-16-5p
↑ let-7b-5p
↑ let-7c
(UTUC patients vs. controls)
Serum samplesBlood drawn within 1 day after admission, the coagulated blood samples were collected in tubes containing a separating gel and clot activator, then centrifugated at 1500× g for 15 min at 4 °C. The supernatant was centrifugated at 1500× g for 15 min and stored at −80 °C.Retrospective, monocenter58 UTUC patients and 42 cancer-free controls with non-neoplastic hematuria (12 UTUC pts and 12 controls in the training set, 46 UTUC pts and 30 controls in the validation set)DC. UTUC: Age: 28 <68 y, 30 ≥68 y; Gender: 17 female; Stage: 21 I stage, 14 II stage, 20 III stage, 3 stage IV; Grade: 1 papillary urothelial neoplasia of low malignant potential, 17 low, 40 high.
Normal: Age: 22 <60 y, 20 ≥68 y; Gender: 12 female.
RNA extracted with miRNeasy Serum/Plasma kit (Qiagen); miRNAs expression profiling: deep sequencing platform Illumina HiSeq™ 2000; RT-qPCR Validation: All-in-One miRNA RT-qPCR Detection kit (GeneCapoiea-FulenGen). qPCR with SYBR green on Applied Biosystems Step One Plus System (Applied Biosystems). RNU6-2 as endogenous reference gene for normalization.Diagnosis.
UTUC patients vs. controls, AUC > 0.8
[232]↓ hsa-miR-10a-5p, ↓ hsa-miR-10b-5p, ↓ hsa-miR-26a-5p, ↓ hsa-miR-29b-2-5p, ↓ hsa-miR-31-5p, ↑ hsa-miR-146b-5p, ↑ hsa-miR-223-3p (G3–G4 vs. G1–G2 and ≥ pT2 vs. pTa/pT1);
↑ hsa-miR-18a-5p and ↓ hsa-miR-199a-5p (G3–G4 vs. G1–G2); hsa-miR-30c-5p (≥pT2 vs. pTa/pT1).
Tissue samplesFormalin fixed paraffin embedded tissue samplesRetrospective, multicenter157 patients with UTUC (35 constituted the screening cohort while 123 constituted the validation cohort)DC: Age: 73.2 y; Gender: 49% female; Stage: 16 pTa, 3 pT1, 4 pT2, 10 pT3, 3 pT4; Grade: 42.9% low, 57.1% high.
VC: 70.9 y; Gender: 33%; Stage: 45 pTa, 31 pT1, 15 pT2, 28 pT3, 4 pT4; Grade: 37% low, 63% high.
Total RNA extracted with RecoverAll® Total Nucleic Acid Isolation Kit (Ambion). RT-qPCR using miRCURY LNA™ reagents (Exiqon).
Screening analysis with Human miRNome panels I and II version 4, analyzing 752 miRNA (Exiqon), using global mean as normalization method.
RT-qPCR Validation using miRNA LNA primer sets.
Prognostic. G3–G4 vs. G1–G2: miR-29b-2-5p, miR-18a-5p, miR-223-3p and miR-199a-5p, with AUC = 0.86 (83% sensitivity, 85% specificity); ≥pT2 vs. pTa/pT1: miR-10b-5p, miR-26a-5p-5p, miR-31-5p and miR-146b-5p, with AUC = 0.9 (64% sensitivity, 96% specificity).
UTUC—upper tract urothelial cancer; miRNA—microRNA; DC—discovery cohort; VC—validation cohort; pts—patients; RT-qPCR—Reverse Transcription quantitative PCR; RNU1-4—U1 Small Nuclear 4 RNA; SNORD43—Small Nucleolar RNA, C/D Box 43; SNORD48—Small Nucleolar RNA, C/D Box 48; AUC—area under the curve; ccRCC—clear cell Renal Cell Carcinoma; papRCC—papillary Renal Cell Carcinoma; FFPE—formalin-fixed paraffin-embedded; SNORA66—Small Nucleolar RNA, H/ACA Box 66; U6snRNA—U6 spliceosomal RNA; RNU1A1—U1 Small Nuclear 1 RNA; AAN-UTUC—Aristolochic Acid-induced upper tract urothelial cancer; RNU6B—U6B small nucleolar RNA; BEN-UTUC—Upper Tract Urothelial Cancer patients living in endemic regions for Balkan endemic Nephropathy; U6 Small Nuclear 2 RNA).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cinque, A.; Capasso, A.; Vago, R.; Floris, M.; Lee, M.W.; Minnei, R.; Trevisani, F. MicroRNA Signatures in the Upper Urinary Tract Urothelial Carcinoma Scenario: Ready for the Game Changer? Int. J. Mol. Sci. 2022, 23, 2602. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23052602

AMA Style

Cinque A, Capasso A, Vago R, Floris M, Lee MW, Minnei R, Trevisani F. MicroRNA Signatures in the Upper Urinary Tract Urothelial Carcinoma Scenario: Ready for the Game Changer? International Journal of Molecular Sciences. 2022; 23(5):2602. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23052602

Chicago/Turabian Style

Cinque, Alessandra, Anna Capasso, Riccardo Vago, Matteo Floris, Michael W. Lee, Roberto Minnei, and Francesco Trevisani. 2022. "MicroRNA Signatures in the Upper Urinary Tract Urothelial Carcinoma Scenario: Ready for the Game Changer?" International Journal of Molecular Sciences 23, no. 5: 2602. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23052602

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop