Fact-checked by Grok 2 weeks ago

Oncomir

An oncomir, also spelled oncomiR, is a (miRNA)—a short, approximately 22 nucleotides in length—that acts as an to promote cancer development by post-transcriptionally repressing the expression of tumor suppressor genes. These miRNAs are typically overexpressed in cancer cells, where they regulate critical cellular processes such as , , , , , and immune evasion, thereby contributing to tumor initiation and progression. The concept of oncomiRs emerged from early studies on miRNAs, with the first miRNA (lin-4) discovered in in 1993, but their link to human cancer was established in 2002 when deletions of miR-15 and miR-16 were found at chromosome 13q14 in (CLL) patients. Subsequent research revealed that oncomiRs can be deregulated through genetic alterations (e.g., amplification or translocation), epigenetic changes, or transcriptional dysregulation, leading to their aberrant expression across diverse malignancies including , , and colorectal cancers. Mechanistically, oncomiRs bind to the 3' untranslated regions of target mRNAs via the (RISC), inhibiting translation or inducing mRNA degradation to silence genes like PTEN and PDCD4. Notable examples include the miR-17-92 cluster, which promotes lymphomagenesis and by modulating E2F1 and pathways; miR-21, an anti-apoptotic oncomir overexpressed in and colorectal cancers that targets tropomyosin 1 and PTEN; and miR-155, which enhances proliferation and immune suppression in various hematological and solid tumors. OncomiRs can also be transferred between cells via exosomes, influencing the to foster , such as by promoting regulatory T-cell activity or inhibiting natural killer cells. Clinically, circulating exosomal oncomiRs like miR-21-5p serve as promising non-invasive biomarkers for cancer and , while therapeutic strategies involving antagomiRs (miRNA inhibitors) have shown potential in preclinical models to suppress tumor growth and reverse chemoresistance.

Overview

Definition and Classification

Oncomirs, also known as oncogenic microRNAs (miRNAs), are a subset of small non-coding RNAs, approximately 22 in length, that promote oncogenesis by dysregulating at the post-transcriptional level. Typically, oncomirs exert their effects through overexpression in cancer cells, leading to the suppression of tumor suppressor genes or the enhancement of oncogenic signaling pathways, thereby facilitating tumor initiation and progression. As a class of miRNAs, oncomirs are distinguished from tumor-suppressor miRNAs, which are generally underexpressed in cancers and inhibit oncogenes; in contrast, oncomirs function analogously to oncogenes by driving malignant phenotypes when upregulated. Understanding their dysregulation requires knowledge of miRNA biogenesis: miRNAs are transcribed as primary transcripts (pri-miRNAs) by , then processed in the nucleus by the Drosha-DGCR8 complex into precursor miRNAs (pre-miRNAs), which are exported to the and further cleaved by into mature miRNAs that incorporate into the (RISC) to target mRNAs. This canonical pathway underscores how alterations in processing or expression can amplify oncomir activity in cancer. Oncomirs contribute to core by modulating key biological processes, such as sustaining proliferative signaling through inhibition of regulators and evading via targeting pro-apoptotic factors. For instance, their regulatory actions can enhance cell survival and unchecked growth without triggering mechanisms. miRNAs in general in by binding to target mRNAs, leading to translational repression or degradation. Numerous miRNAs have been implicated as potential oncomirs across various cancer types, highlighting their widespread role in oncogenesis.

Historical Development

The discovery of microRNAs (miRNAs) as key regulators of gene expression began in the early 1990s with studies in the nematode Caenorhabditis elegans. In 1993, Victor Ambros and colleagues identified lin-4 as the first miRNA, a small non-coding RNA that negatively regulates the protein LIN-14 through antisense complementarity, thereby controlling developmental timing. This finding established miRNAs as post-transcriptional regulators, though it was initially viewed as an anomaly. Building on this, in 2000, Gary Ruvkun's group discovered let-7, another small RNA that similarly modulates heterochronic genes like lin-41 and is highly conserved across species, including humans, highlighting the broad evolutionary role of miRNAs in gene regulation. David Bartel's subsequent work in the early 2000s further elucidated miRNA biogenesis and target prediction mechanisms, providing a foundational framework for understanding their regulatory potential. The link between miRNAs and cancer emerged in the mid-2000s, marking the birth of oncomir research. In 2005, Lin He and colleagues identified the miR-17-92 cluster as the first oncogenic miRNA, demonstrating its amplification in B-cell lymphomas and its ability to promote lymphomagenesis by inhibiting in cooperation with ; this study coined the term "oncomir" to describe miRNAs with tumor-promoting functions. Shortly thereafter, profiling studies revealed miR-21 as overexpressed in tissues compared to normal adjacent samples, where it targets tumor suppressor genes like PDCD4 to enhance invasion and proliferation. Similarly, between 2006 and 2008, miR-155 was established as an oncomir in leukemias, with sustained expression in hematopoietic stem cells driving myeloproliferative disorders and pre-B-cell lymphomas through suppression of genes like SHIP1. From 2010 onward, high-throughput sequencing transformed oncomir research by enabling genome-wide miRNA profiling in large cohorts. (TCGA) initiative, starting with in 2012 and expanding to pan-cancer analyses, integrated miRNA data with genomic alterations, uncovering context-specific oncomir roles such as tissue-dependent deregulation patterns across 33 tumor types. By the mid-2020s, these datasets had facilitated the identification of over 10,000 in miRNA genes and refined understandings of oncomir contributions to tumor heterogeneity, solidifying their status as critical cancer drivers.

Biological Mechanisms

General Regulatory Mechanisms

OncomiRs, or oncogenic microRNAs, are upregulated in cancer through multiple mechanisms that enhance their expression and oncogenic potential. Genomic of chromosomal regions containing oncomiR genes leads to increased copy numbers and thus elevated expression levels. Transcriptional activation occurs when oncogenes such as bind to promoter regions of oncomiR clusters, driving their overexpression in proliferating cancer cells. Epigenetic modifications, including hypomethylation of oncomiR promoters mediated by enzymes like demethylases, further contribute to this upregulation by making transcriptional machinery more accessible. Additionally, alterations in the (TME), such as , induce factors like HIF-1α to transcriptionally activate specific oncomiRs, adapting their expression to stressful conditions that favor tumor survival. Once upregulated, oncomiRs exert their effects primarily by binding to the 3' untranslated regions (3' UTRs) of target messenger RNAs (mRNAs), leading to translational repression or mRNA destabilization and degradation via the (RISC). This disrupts broad cellular pathways, such as the suppression of tumor suppressors like PTEN, which normally inhibits the PI3K/AKT signaling cascade; reduced PTEN activity results in unchecked PI3K/AKT activation, promoting , survival, and evasion of . Such interactions allow oncomiRs to coordinately deregulate multiple downstream effectors, amplifying oncogenic signaling networks. The function of oncomiRs is highly context-dependent, varying across different cancer types due to differences in cellular milieu, genetic background, and TME cues. In epithelial cancers, oncomiRs can drive epithelial-mesenchymal transition () by repressing E-cadherin and activating mesenchymal markers, enhancing cellular motility and invasiveness essential for . This context-specific regulation means the same oncomiR may promote tumor progression in one cancer type while having minimal effects in another, influenced by reciprocal feedback loops with transcription factors like ZEB or that fine-tune EMT dynamics. Recent insights from 2023 to 2025 highlight the role of other non-coding RNAs, such as long non-coding RNAs (), in modulating oncomiR expression and activity through sponging or stabilizing interactions within the TME. Furthermore, extracellular vesicles (), including exosomes, facilitate oncomiR dissemination by packaging and transferring them between cancer cells and stromal components, thereby propagating oncogenic signals to distant sites and contributing to pre-metastatic niche formation.

Oncomir Addiction

Oncomir addiction refers to the phenomenon in which cancer cells develop a critical dependence on the sustained activity of specific oncogenic microRNAs (oncomiRs) to maintain their , even in the presence of numerous genetic alterations. This concept parallels oncogene addiction, where tumors rely on a single driver despite genomic complexity, making oncomiRs pivotal nodes in oncogenic networks. Inhibition of these oncomiRs can lead to rapid tumor regression or , highlighting their role beyond initiation in sustaining cancer progression. Mechanistically, oncomiRs perpetuate addiction by stabilizing oncogenic signaling pathways through regulatory feedback loops and by suppressing tumor suppressor genes, thereby bypassing normal cellular safeguards. For instance, oncomiRs such as miR-21 can downregulate protein 4 (PDCD4), preventing and allowing unchecked , while clusters like miR-17-92 form with transcription factors such as and to amplify proliferative signals. This creates a where cancer cells, having rewired their regulatory landscape around the oncomiR, cannot survive its disruption, as alternative pathways fail to compensate for the loss of these finely tuned networks. Evidence for oncomir stems from preclinical models demonstrating tumor collapse upon oncomir suppression. In a 2010 mouse model of miR-21-driven pre-B-cell , conditional overexpression of miR-21 induced tumors, but its doxycycline-inducible shutdown via antisense triggered rapid regression in established tumors, confirming dependency. Similar results were observed with miR-155 inhibition in models, where antagomiRs delayed tumor growth by restoring suppressor functions. More recent preclinical data from 2025 using / to knock out miR-21 in lung adenocarcinoma cell lines (A549) showed reduced proliferation (by ~31% at 72 hours), increased via upregulated PTEN and PDCD4, and enhanced chemosensitivity to drugs like , underscoring sustained reliance in solid tumors. Therapeutically, exploiting oncomir addiction enables selective cancer cell killing while sparing normal cells, as non-addicted tissues do not depend on these miRNAs. Antisense and locked nucleic acids (LNAs) targeting addicted oncomiRs have shown promise in models, with miR-21 inhibition in preclinical studies enhancing drug efficacy and suggesting potential for combination therapies. This approach leverages the "" created by addiction, positioning oncomir inhibitors as precision tools in .

Key Examples

miR-21 and miR-155

miR-21, a well-characterized oncomir, is frequently overexpressed in numerous solid tumors, where it acts as a key regulator of oncogenic processes. By targeting tumor suppressor genes such as PTEN, PDCD4, and TPM1, miR-21 promotes , invasion, and resistance to in cancer cells. In , miR-21 enhances tumor progression through multiple signaling pathways, including those involving and PTEN suppression, contributing to poor prognosis as highlighted in a 2024 review. Similarly, in and colorectal cancers, elevated miR-21 levels correlate with increased and reduced patient survival by inhibiting and epithelial-mesenchymal transition () regulators. miR-155, another prominent oncomir, is upregulated in both hematologic malignancies and solid tumors, often transcribed from the gene as its host transcript. It targets genes like and SHIP1, thereby enhancing cell survival, proliferation, and inflammatory responses that facilitate immune evasion in the . In lymphomas, miR-155 overexpression drives lymphomagenesis by promoting B-cell transformation and resistance to , while in , it supports tumor progression through pathways involving and signaling. This upregulation contributes to chronic , a hallmark of cancer, by modulating immune function and suppressing antitumor immunity. Both miR-21 and miR-155 contribute to , a critical step in cancer , but miR-21 predominantly drives this process in solid tumors like and colorectal cancers through direct suppression of molecules, whereas miR-155 exerts stronger effects in immune-related contexts, such as lymphomas, by altering inflammatory signaling. Experimental studies have demonstrated that disrupting miR-21 expression reduces tumor and , leading to decreased tumorigenesis. Likewise, miR-155 in tumor cells impairs glucose metabolism, resulting in slower tumor growth.

miR-17-92 Cluster and Others

The miR-17-92 cluster, recognized as the first identified oncomir, was discovered in 2005 through studies showing its amplification and overexpression in human B-cell lymphomas, where it functions as a polycistronic unit encoding multiple mature microRNAs. This cluster comprises six principal members: miR-17, miR-18a, miR-19a, miR-19b, miR-20a, and miR-92a, transcribed from a single primary transcript located at 13q31.33. Its expression is primarily regulated by the MYC, which binds to the promoter region to drive polycistronic transcription, thereby amplifying oncogenic signaling in responsive cells. Key targets include E2F1 (repressed by miR-17 and miR-20a to modulate progression and ) and PTEN (targeted by miR-19 to inhibit tumor suppression and promote survival). The cluster plays a pivotal role in B-cell lymphomas, where its enforced expression cooperates with MYC to accelerate lymphomagenesis, and extends to solid tumors such as , colon, and cancers, enhancing and inhibiting . Paralogous clusters, miR-106b-25 and miR-106a-363, share and functional overlap with miR-17-92, often exhibiting coordinated dysregulation in cancers to reinforce oncogenic networks. The miR-17-92 cluster demonstrates cooperative dynamics among its members, with miR-19 driving proliferation by suppressing PTEN and regulators, while miR-92a promotes by targeting anti-angiogenic factors like thrombospondin-1, collectively amplifying tumor growth and vascularization. Genomic amplification of the cluster occurs frequently across malignancies, including lung cancers, and variably in other solid tumors, contributing to its oncogenic potency. In certain contexts, such as MYC-driven lymphomas, cancer cells exhibit addiction to the cluster, where its inhibition disrupts feedback loops with and , leading to tumor regression. Beyond the miR-17-92 cluster, other notable oncomirs illustrate the diversity of microRNA-driven oncogenesis. For instance, miR-569, amplified at 3q26.2 in epithelial cancers including , acts as an oncomir by directly targeting TP53INP1, a tumor suppressor that induces cell cycle arrest and , thereby enhancing tumor aggressiveness, , and chemoresistance. miR-181a-5p exhibits a dual role across malignancies, functioning as a tumor suppressor in many but oncogenic in specific contexts such as , where its upregulation promotes invasion and survival by targeting regulators like FBXO11. Similarly, miR-184 displays context-dependent oncogenic activity in head and neck squamous cell carcinomas, where it is upregulated to drive and inhibit through upregulation of c-Myc, contrasting its tumor-suppressive effects in other head and neck subtypes like . Recent investigations, including 2024 studies, have highlighted the miR-17-92 cluster's role in modulating the (TME), where its components influence immune infiltration and metabolic reprogramming to foster an immunosuppressive niche supportive of tumor progression.

Clinical and Research Applications

Diagnostic and Biomarker Roles

Oncomirs circulating in biofluids such as , , and exosomes enable non-invasive liquid biopsies for cancer detection, , and treatment monitoring, offering a dynamic view of tumor activity. Encapsulated within exosomes, these miRNAs exhibit superior stability compared to protein markers, as their protection shields them from degradation in circulation, while their high abundance—up to $10^9 particles per mL—facilitates sensitive detection even at early disease stages. Unlike proteins, which often derive from apoptotic s and lack tumor-specific signatures, exosomal oncomirs reflect live secretions, providing mechanistic insights into oncogenesis. For instance, urinary miRNA ensembles, as blood byproducts, have emerged as promising for early detection in urological and other cancers, with profiles distinguishing malignant from benign conditions. In , circulating multi-miRNA panels in plasma demonstrate robust diagnostic performance for non-small cell lung cancer (NSCLC) subtypes, achieving 83% sensitivity for and 92% for in a 2024 validation cohort of over 4,000 patients, with area under the curve () values up to 0.98. Panels incorporating miR-21, a key oncomir upregulated in NSCLC, enhance early identification when combined with , as shown in studies of peripheral from patients with pulmonary nodules. Similarly, a 2025 meta-analysis of 29 studies on reported pooled sensitivity of 85% and specificity of 84% ( 0.90) for plasma-based multi-miRNA panels, outperforming single-miRNA assays by integrating diverse regulatory pathways like PI3K/AKT. Tissue-based oncomir profiling from tumor biopsies supports subtype classification and prognostic stratification. In lymphomas, miR-155 signatures in biopsies distinguish activated B-cell-like from B-cell-like and predict outcomes, with high expression linked to shorter overall survival and in multiple cohorts. Elevated miR-21 in tumor tissues across cancers, including and , correlates with aggressive disease; a 2025 meta-analysis of studies (pooled HR 2.37 for overall survival, 95% 1.42–3.98; HR 1.97 for disease-free survival, 95% 1.39–2.80) underscores its role in forecasting poor prognosis, particularly in triple-negative subtypes (HR 5.69). Multi-miR panels in tissues further refine predictions by capturing heterogeneous expression patterns, surpassing individual oncomirs in accuracy for survival forecasting. Despite these advances, oncomir biomarkers encounter hurdles in clinical adoption, including standardization of detection via qPCR and NGS, where variability in sample collection, , and —lacking consensus internal controls—compromises reproducibility. Specificity remains challenging due to miRNA causing and elevated levels in inflammatory or non-cancerous states, necessitating refined protocols to distinguish tumor-derived signals.

Therapeutic Strategies Including Anti-oncomirs

Therapeutic strategies targeting oncomirs primarily involve inhibiting their oncogenic activity to restore tumor suppressor pathways and disrupt cancer progression. Anti-oncomirs, such as synthetic antisense including antagomirs and locked nucleic acids (LNAs), are designed to bind and sequester oncomirs, preventing their with target mRNAs. These inhibitors have shown promise in preclinical models by reducing tumor growth and enhancing , particularly for overexpressed oncomirs like miR-21, miR-155, and the miR-17-92 cluster. For miR-21, a key oncomir in (GBM), LNA-based inhibitors and antagomirs delivered via lipid nanoparticles have demonstrated reduced tumor proliferation and increased sensitivity to in preclinical studies. Similarly, CRISPR-SaCas9-mediated editing of the miR-21 locus using a single (AAV) vector in GBM mouse models led to up to 180-fold reduction in miR-21 expression, upregulation of suppressors like PTEN and PDCD4, and a 31% increase in median survival (from 21.5 to 28.5 days). In hematological malignancies, inhibition of miR-155 with the LNA-modified MRG-106 (cobomarsen) in preclinical (CTCL) models induced antileukemic effects, including transcriptome changes consistent with target de-repression and reduced . For the miR-17-92 cluster, preclinical inhibition using targeted inhibitors like MIR17PTi slowed tumor progression in by modulating pathways such as PTEN and HIF-1α. Beyond direct inhibition, miRNA mimics of tumor suppressor miRNAs can indirectly counteract oncomir effects by restoring regulatory balance, such as miR-34a mimics that enhance antitumor drug efficacy in various cancers. CRISPR-based editing of oncomir loci offers precise genomic disruption, as seen in GBM models targeting miR-21 to exploit oncomir addiction vulnerabilities. Combination therapies integrating anti-oncomirs with address resistance; for instance, anti-miR-21 with in GBM liposomes overcomes multidrug resistance by downregulating ABC transporters like ABCC1. Similarly, miR-155 inhibitors combined with standard agents in AML models sensitize cells by modulating and pathways. Effective delivery remains a critical challenge, with nanoparticle systems (e.g., , lipid-based) and viral vectors (e.g., AAV, lentiviral) enabling tumor-specific targeting while protecting anti-oncomirs from degradation. Lipid nanoparticles improve stability and biodistribution but face issues like low encapsulation efficiency (due to miRNA hydrophilicity) and rapid clearance by the . Viral vectors offer high but risk immunogenicity and . Off-target effects, immune activation, and poor endosomal escape further complicate translation, though exosome-based delivery shows potential for crossing barriers like the blood-brain barrier in GBM. Clinical progress includes phase I/II trials for miR-155 inhibitors like cobomarsen in (NCT02580552), demonstrating safety and partial responses, with preclinical extensions to . For miR-21, ongoing preclinical work in GBM has shown promise, while miR-17-92 targeting remains largely preclinical, with safety data from related miR-17 inhibitors in non-cancer trials (e.g., NCT04536688). These advances highlight anti-oncomirs' potential, though optimizing and minimizing are essential for broader efficacy.

Resources and Further Study

Databases and Tools

Several major databases serve as foundational resources for oncomir research, providing curated data on miRNA sequences, annotations, and cancer-specific interactions. miRBase remains the primary repository for sequences and annotations, cataloging over 48,000 mature miRNAs from 271 organisms in its latest release (v22.1 as referenced in 2025 studies). Updated annotations in 2024 incorporated improved structural predictions and expression data integration, facilitating oncomir identification across species. OncomiRDB, launched in 2014, specializes in experimentally verified oncogenic and tumor-suppressive s, compiling direct functional evidence such as regulation of cancer-related phenotypes from literature sources. It enables targeted queries for oncomir-target interactions, with over 1,000 entries linking miRNAs to validated pathways. miRTarBase, updated in 2025, collects over 3.8 million experimentally validated miRNA-target interactions from more than 13,000 articles, including cancer-specific data on and therapeutic implications; it supports oncomir research through searchable MTIs and integration with tools for network analysis. (TCGA) miRNA data portals, accessible via the Genomic Data Commons, offer comprehensive cancer-specific expression profiles from thousands of tumor samples across 33 cancer types, including raw sequencing data and normalized datasets for oncomir dysregulation analysis. Computational tools complement these databases by predicting and analyzing oncomir functions. TargetScan employs a seed-matching to forecast conserved miRNA target sites in mRNAs, prioritizing 6-8mer sites in 3' UTRs for high-confidence predictions; it is widely used for oncomir studies due to its integration of evolutionary conservation scores. DIANA-microT (now microT-CDS) utilizes to score miRNA-mRNA interactions based on and site , supporting batch predictions for multiple oncomirs and exporting results in tabular formats. CircInteractome focuses on miRNA binding sites within circular RNAs, aiding analysis of circulating miRNA interactions in extracellular vesicles; it incorporates TargetScan predictions to map potential regulatory networks in cancer biofluids. Practical usage of these resources involves straightforward querying and . For instance, researchers can query miR-21 targets in TargetScan to retrieve predicted genes like PTEN, then cross-reference expression datasets from TCGA portals to validate dysregulation in specific cancers. Downloading miRBase files allows alignment with OncomiRDB for functional of novel oncomirs. Recent 2025 updates have integrated AI-driven pattern recognition into pipelines combining NGS data from TCGA with tools like microT-CDS, enabling automated discovery of oncomir signatures through models for target prioritization. Despite their utility, these databases and tools face limitations requiring careful curation. Context-dependency of miRNA targeting—such as tissue-specific expression or post-transcriptional modifications—often leads to false positives in predictions, necessitating experimental validation beyond computational outputs. Additionally, while most resources like miRBase and TargetScan are open-access, proprietary platforms (e.g., certain TCGA-derived analytics suites) restrict full data access, hindering in global research efforts.

Emerging Research Directions

Recent research has increasingly focused on the role of oncomirs in the (TME) and , particularly their contributions to immune evasion and stromal interactions. Oncomirs such as miR-21 and miR-155 are upregulated in the TME under hypoxic and acidic conditions, promoting () and the recruitment of suppressive immune cells like () and regulatory T cells (Tregs). Exosomal transfer of miR-21 from cancer cells or to stromal cells downregulates tumor suppressors like PTEN and PDCD4, enhancing activity, degradation, invasion, and via pathways such as YAP1/HIF-1α. These mechanisms facilitate immune evasion by suppressing antitumor responses and preparing premetastatic niches, as evidenced in head and neck (HNSCC) models where exosomal miR-21 correlates with poor . The context-dependent functions of oncomirs highlight their dual roles across cancer types, complicating therapeutic targeting but offering nuanced insights into tumor biology. For instance, miR-184 acts as an oncomir in HNSCC and osteosarcoma (with controversial evidence in glioma) by promoting proliferation and inhibiting apoptosis through targeting genes like SOX7, INPPL1, and BCL2L1. Conversely, it functions as a tumor suppressor in lung, breast, colorectal, gastric, prostate, endometrial, ovarian, renal cell, hepatocellular, and retinoblastoma carcinomas, where downregulation enhances migration, invasion, and EMT via pathways such as Wnt/β-catenin and AKT/mTORC1. Advancements in liquid biopsy have leveraged these context-specific profiles, with circulating miRNAs enabling non-invasive detection; a 2023 study using droplet digital PCR on plasma from 268 genitourinary cancer patients identified panels like hsa-miR-155-5p/hsa-miR-375-3p for renal cell carcinoma (80.54% specificity) and hsa-miR-126-3p/hsa-miR-375-3p for bladder cancer (94.87% specificity), supporting early diagnosis and risk stratification. Gaps in oncomir research include limited exploration of (ncRNA) crosstalk and AI-driven discovery, alongside underrepresentation of recent studies in genitourinary cancers. NcRNA crosstalk, such as interactions between oncomirs and long non-coding RNAs (lncRNAs) or RNA-binding proteins, modulates tumor progression by altering networks, yet comprehensive models remain sparse. and have accelerated miRNA identification, with 2024 reviews highlighting for predicting oncogenic miRNAs across cancers like gastric and head and neck, achieving high accuracy in survival models via explainable . Studies from 2023-2025 on genitourinary malignancies, such as miR-155-5p promoting proliferation in via JADE-1 targeting and miR-371a-3p as a for testicular tumors, underscore diagnostic potential but lack integration into broader therapeutic frameworks. Looking ahead, oncomir profiling promises to advance by tailoring therapies to individual miRNA signatures, while integration with could enhance efficacy. Circulating miRNA panels may guide patient stratification for inhibitors, with miR-21 levels predicting responses in cancer chemoimmunotherapy. Combining miRNA modulation with vaccines or checkpoint blockade, as in miRNA-enhanced therapies, holds potential to overcome resistance and boost antitumor immunity.

References

  1. [1]
    OncomiRs: the discovery and progress of microRNAs in cancers
    Recent evidences indicate that miRNAs can function as tumor suppressors and oncogenes, and the miRNAs with a role in cancer are designated as oncogenic miRNAs ( ...Missing: definition | Show results with:definition
  2. [2]
    OncomiRs as noncoding RNAs having functions in cancer - Frontiers
    The overexpression of oncomiRs in cancer cells promote cancer through the regulation of the expression of multiple tumor suppressor gene targets with cancer- ...
  3. [3]
    The regulatory mechanisms of oncomiRs in cancer - ScienceDirect
    The current review summarises the different mechanisms controlling the upregulation of oncomiR expression in cancer cells and the tumour microenvironment (TME).
  4. [4]
    microRNAs as oncogenes and tumor suppressors - ScienceDirect.com
    Feb 1, 2007 · These oncogene miRNAs, called “oncomirs”, usually promote tumor development by negatively inhibiting tumor suppressor genes and/or genes that ...Review · Mirna Biogenesis · Microrna Microarray
  5. [5]
    OncomiR or Tumor Suppressor? The Duplicity of MicroRNAs in ...
    Generally, oncogenic miRNAs (oncomiRs) are overexpressed in cancers while tumor suppressive miRNAs are underexpressed. When these oncomiRs or tumor suppressor ...Missing: definition | Show results with:definition
  6. [6]
    The Drosha-DGCR8 complex in primary microRNA processing
    The nuclear RNase III Drosha cleaves primary miRNAs (pri-miRNAs) to release hairpin-shaped pre-miRNAs that are subsequently cut by the cytoplasmic RNase III ...
  7. [7]
    The Duality of OncomiR Addiction in the Maintenance and ...
    OncomiRs perpetuate cancer through regulation of biological pathways that include apoptosis, proliferation, differentiation, cell senescence, and angiogenesis.
  8. [8]
    CMC: Cancer miRNA Census – a list of cancer-related miRNA genes
    Jan 23, 2024 · We created a list of 165 cancer-related miRNA genes called the Cancer miRNA Census (CMC). The list is based on a score, built on various types of functional ...
  9. [9]
    The C. elegans heterochronic gene lin-4 encodes small RNAs with ...
    The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14 ... Cell. 1993 Dec 3;75(5):843-54. doi: 10.1016/0092-8674(93) ...
  10. [10]
    Pan-cancer analysis of somatic mutations in miRNA genes
    In this study, we identified 7110 mutations in miRNA genes across 33 cancer types based on data available in the TCGA repository. Most of the mutations were ...
  11. [11]
    A Comprehensive Review of Cancer MicroRNA Therapeutic ...
    MiRNA binding through a partial complementary sequence (seed sequence) within the target mRNA 3'UTR primarily inhibits translation or promotes its degradation.2. Altered Microrna... · 2.4. Mirna Replacement... · 3.5. Nanoparticles
  12. [12]
  13. [13]
    Regulation of cancer metastasis by microRNAs - PMC
    Many studies have shown that microRNAs could directly target genes playing a central role in epithelia-mesenchymal-transition (EMT), a cellular transformation ...
  14. [14]
    Extracellular RNA in oncogenesis, metastasis and drug resistance
    Aug 6, 2024 · We review the roles of specific miRNAs that have been detected in EVPs and that play a role in oncogenesis and metastasis. KEYWORDS:.
  15. [15]
  16. [16]
  17. [17]
    The miR-17/92 cluster: a comprehensive update on its genomics ...
    Nov 11, 2013 · The miR-17/92 cluster is among the best-studied microRNA clusters. Interest in the cluster and its members has been increasing steadily.
  18. [18]
    Genetic dissection of the miR-17∼92 cluster of microRNAs in Myc ...
    The miR-17∼92 cluster is frequently amplified or overexpressed in human cancers and has emerged as the prototypical oncogenic polycistron microRNA (miRNA).
  19. [19]
    A Polycistronic MicroRNA Cluster, miR-17-92, Is Overexpressed in ...
    Nov 1, 2005 · The miR-17-92 cluster, which comprises seven miRNAs and resides in intron 3 of the C13orf25 gene at 13q31.3, is markedly overexpressed in lung cancers.
  20. [20]
    Consequences of the feedback loops involving miR-17-92, E2F, and ...
    The cluster is composed of 7 mature miRs, namely, miR-17–5p, miR-17–3p, miR-18a, miR-19a, miR-20a, miR-19b, and miR-92–1 (Fig. 2A). The nucleotide sequences and ...Microrna Regulation Of A... · Results And Discussion · Mir-17-92 As An Oncogene And...
  21. [21]
    Copy Number Gain of hsa-miR-569 at 3q26.2 Leads to Loss of ...
    ... Copy Number Gain of hsa-miR-569 at 3q26.2 Leads to Loss of TP53INP1 and Aggressiveness of Epithelial Cancers. Pradeep Chaluvally-Raghavan. Pradeep Chaluvally ...
  22. [22]
    Role of miR‑181a‑5p in cancer (Review) - Spandidos Publications
    Aug 3, 2023 · Numerous studies have demonstrated that miR‑181a‑5p exerts regulatory influence on cancer development and progression, acting as an oncomiR or ...
  23. [23]
    microRNA-184 in the landscape of human malignancies: a review to ...
    Nov 24, 2023 · It is documented that miR-184 shows widely deregulation in numerous cancers with controversial functions either as an oncomiR in some cancers ...
  24. [24]
    Unraveling the Multifaceted Role of the miR-17-92 Cluster in ...
    Feb 28, 2024 · The miR-17-92 cluster is upregulated in colon cancer and contributes to tumor development by promoting angiogenesis through the suppression of ...
  25. [25]
    Exosomes as a new frontier of cancer liquid biopsy
    Feb 18, 2022 · Compared with CTCs and ctDNA, exosomes have shown greater advantages in liquid biopsy. First, the presence of large amounts of exosomes (~ 109 ...
  26. [26]
    Early Cancer Detection via Multi-microRNA Profiling of Urinary ...
    Oct 18, 2024 · Urinary miRNA ensembles, which are blood byproducts, have the potential to serve as a cancer warning sign, the same as the blood miRNA ensembles ...
  27. [27]
    Circulating miRNA panels as a novel non-invasive diagnostic ...
    Aug 30, 2024 · Circulating miRNA signatures demonstrate diagnostic and prognostic value for NSCLC and may guide treatment decisions in clinical practice.
  28. [28]
    Clinical value of peripheral blood miR-21 and miR-486 combined ...
    Sep 20, 2024 · This study aimed to investigate the clinical significance of combining peripheral blood miR-21 and miR-486 with CT for the early cancer diagnosis in pulmonary ...<|separator|>
  29. [29]
    Diagnostic Potential of Cross-Specimen microRNA Panels as ...
    Aug 23, 2025 · This systematic review and meta-analysis evaluated the diagnostic accuracy of multi-miRNA panels and explored their mechanistic relevance to ...
  30. [30]
    The Role of microRNA-155 as a Biomarker in Diffuse Large B-Cell ...
    Nov 21, 2024 · The study found that increased expression of miR-155 was consistently linked to lower survival rates (OS, PFS, event-free survival) in patients ...
  31. [31]
    The Prognostic Power of miR-21 in Breast Cancer - NIH
    Oct 6, 2025 · This systematic review and meta-analysis aimed to clarify the prognostic significance of miR-21 in BC and explore its molecular mechanisms ...
  32. [32]
    MicroRNAs: circulating biomarkers for the early detection of ... - Nature
    Jun 5, 2024 · Early studies faced challenges in establishing robust biomarker panels and overcoming the heterogeneity of cancer types.
  33. [33]
    MicroRNA in cancer therapy: breakthroughs and challenges in early ...
    Apr 21, 2025 · This review examines the dual role of miRNAs in solid and hematological malignancies, starting from their dysregulation in various cancer types.
  34. [34]
    microRNAs (miRNAs) in Glioblastoma Multiforme (GBM) - MDPI
    According to the latest research, two basic strategies for miRNA-based therapy have been proposed, the first of which is to restore the downward-regulated ...Micrornas (mirnas) In... · 4. Tumor Suppressor Mirnas · 6. The Use Of Mirnas In Gbm...
  35. [35]
    CRISPR targeting of mmu-miR-21a through a single adeno ...
    Jan 8, 2025 · In this study, we demonstrated that disruption of genomic mmu-miR-21a with a single AAV vector influenced glioma development resulting in beneficial anti-tumor ...<|separator|>
  36. [36]
    Preclinical Results Supporting Therapeutic Development of Mrg-106 ...
    Dec 3, 2015 · Inhibition of miR-155-5p resulted in transcriptome changes consistent with miR-155-5p target gene modulation, reduction in cell ...
  37. [37]
    Targeting oncomiRNAs and mimicking tumor suppressor miRNAs
    One of the solid results emerging from different independent studies is that miRNA replacement therapy can enhance the antitumor effects of the antitumor drugs.
  38. [38]
    MicroRNA-Based Combinatorial Cancer Therapy: Effects of ... - NIH
    Combination therapies have been proposed to overcome therapeutic resistance via the combined inhibition of different mechanisms. For example, the combination of ...
  39. [39]
    MicroRNA delivery through nanoparticles - PMC - PubMed Central
    The main challenges in miRNA delivery with nanocarriers are related to their low encapsulation efficiency [60], and the need for cell targeting, as shown ...
  40. [40]
    Advances in MicroRNA Therapeutics: From Preclinical to Clinical ...
    In this review, we present recent advances in the development of miRNA therapeutics currently in preclinical or clinical stages for treating both rare genetic ...Advances In Microrna... · Mirna Therapeutics... · Mirna-Based Therapeutics In...<|control11|><|separator|>
  41. [41]
    MicroRNAs as Emerging Therapeutic Targets Modulating the Tumor ...
    For instance, exosomal miR-21 can downregulate PDCD4, TPM1, and PTEN in neighboring cells, promoting tumor invasion and immune evasion [17]. This bidirectional ...
  42. [42]
    microRNA-184 in the landscape of human malignancies: a review to ...
    Nov 24, 2023 · It is documented that miR-184 shows widely deregulation in numerous cancers with controversial functions either as an oncomiR in some cancers or ...<|control11|><|separator|>
  43. [43]
    Crosstalk between RNA-binding proteins and non-coding RNAs in ...
    This article has provided a comprehensive review of the roles played by RNA-binding proteins and their associated non-coding RNAs in tumor biology.
  44. [44]
    Advancing miRNA cancer research through artificial intelligence
    Dec 17, 2024 · This review comprehensively examines current developments in the application of machine learning techniques in miRNA research across diverse cancers.Missing: oncomirs 2023-2025
  45. [45]
    MicroRNAs in Genitourinary Malignancies: An Exciting Frontier of ...
    In this review, we describe how novel miRNAs can be used in the treatment of prostate, urothelial, kidney, testicular, and other genitourinary cancers, both in ...
  46. [46]
    Circulating miRNAs as potentially predictive biomarkers for ... - Nature
    Aug 30, 2025 · Circulating miRNAs as potentially predictive biomarkers for chemoimmunotherapy in advanced biliary tract cancer: a post-hoc analysis of the ...
  47. [47]
    A new era of cancer immunotherapy: vaccines and miRNAs - PMC
    Apr 1, 2025 · This review examines the important role of vaccines in cancer therapy, focusing on recent advancements such as dendritic cell vaccines, mRNA vaccines, and ...