Abstract
In today's rapidly advancing field of medical research, non-coding RNA (ncRNA) and nanomedicine have emerged as promising areas of study for therapeutic and diagnostic approaches. ncRNAs, previously considered "junk DNA" and hence insignificant, are now being documented for their remarkably extraordinary regulatory roles in gene expression and various cellular processes. These molecules acquire various forms, comprising microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and small interfering RNAs (siRNAs), each with its distinct functions. The enormous benefits of ncRNA therapies include ease of sequence design and creation, functional flexibility, charge and protection, and the opportunity for patient-specific management. Nanomedicine, on the other hand, combines nanotechnology and medicine through developing innovative solutions for disease treatment and diagnosis. This article provides an overview of the technical aspects and potential of commercializing the design and targeting of ncRNAs using nanocarriers and nano-delivery systems for miRNA delivery. Furthermore, the impact of nanomedicine on the healthcare industry, as well as its therapeutic and diagnostic applications, has been investigated. Overall, this study will provide insight into novel systems for the treatment and diagnosis of ncRNA.
Keywords: Nanomedicine, Non-coding RNA, Diagnostic approach, Therapeutic approach, Commercialization opportunities
Introduction
Non-coding RNA
Non-coding RNA (ncRNA) refers to a class of RNA molecules that do not code for proteins.1 Previously dismissed as "junk DNA," ncRNAs have proven to be crucial players in gene regulation, cellular development, and disease progression. These molecules come into various forms, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and small interfering RNAs (siRNAs), each with its distinct functions (Fig. 1). Long non-protein-coding RNAs > 200 nucleotides in length, some of which play crucial roles in a variety of biological processes such as promoter-specific gene regulation, epigenetic control of chromatin, X-chromosome inactivation, mRNA stability, and imprinting. Small ncRNAs are symbolized by a wide range of identified and recently discovered RNA species, with many being related to 5′ or 3′ regions of protein-coding genes. This class includes well-documented siRNAs, miRNAs, piRNAs, and others.2,3
Fig. 1.
The different classification of ncRNA: Function and their regulatory role
Nanomedicine: Developing healthcare at the nanoscale
Nanomedicine is the application of nanotechnology in the field of medicine. It involves the design, development, and use of nanoscale materials and devices for various healthcare purposes. Nanomedicine also holds great promise for improving patient outcomes and revolutionizing healthcare. To realize the impact of nanomedicine, it is desirable to understand the nanoscale. The nanoscale refers to dimensions ranging from 1 to 100 nm, where one nanometer is equivalent to one billionth of a meter.5 At this scale, materials exhibit unique physical, chemical, and biological properties that vary from their bulk complements. These properties consist of increased surface area, enhanced reactivity, and improved cellular interactions. Scientists can create innovative solutions for medical challenges by manipulating, using, and engineering materials at the nanoscale.6
Nanomedicine has the potential to revolutionize healthcare by enabling precise drug delivery, imaging, and diagnostics at the molecular level.
Quantitative real-time PCR (qRT-PCR), digital droplet PCR (ddPCR), RNA, and sequencing (RNA seq) are common ways to investigate ncRNA potential biomarkers.7 By applying different materials and devices at the nanoscale, nanomedicine can offer innovative solutions for targeted treatments with reduced side effects and better-quality therapeutic outcomes.8 For this purpose, nano-sensors/biosensors (Fig. 2), and nanoparticles (NPs) as nanocarriers are some of the main fields that play significant roles in the nanomedicine scope.9 Multi-functionalized NPs and nano-based sensors have been developed by targeted action via binding specified ligands to target the tissues for the diagnosis and treatment of cancer.10
Fig. 2.
Enhancement of lncRNA detection by using functionalized NPs.
Regulatory roles of microRNAs and nanocarriers
miRNA, a type of small non-coding RNA, regulates gene expression by binding to target messenger RNAs (mRNAs) and either inhibiting their translation or promoting their degradation. They play key roles in cellular processes such as development, differentiation, and apoptosis. 2 Furthermore, dysregulation of specific miRNAs has been linked to various diseases, including cancer, cardiovascular disorders, and neurodegenerative conditions.3
It is believed that among ncRNA, miRNA can be effectively applied for cancer treatment, as well as for many other purposes. For this purpose, miRNA-nanocarriers are engineered to deliver miRNA molecules to specific cellular targets with unparalleled precision. In cancer treatment, for example, these nanocarriers can deliver miRNAs that prevent tumor growth while sparing healthy tissues.
There are five major nano-delivery systems groups for miRNA delivery. (1) miRNAs can be chemically conjugated to nucleic acid/protein NPs such as antibodies, aptamers, and pRNA to support delivery. (2) Inorganic NPs are a novel delivery system with a small size of about 1-70 nm. (3) Cell-derived membrane nanocarriers can also be utilized up to 200 nm in size. (4) Lipid-based delivery systems are popular due to their high gene transfection efficiency. (5) Polymers are another efficient delivery strategy with their large size compared to other systems (up to 500 nm) (Fig. 3). It is believed that by tailoring treatment regimens based on an individual's miRNA profile, healthcare providers can improve therapy outcomes, minimize adverse effects, and enhance overall patient care.
Fig. 3.
Schematic illustration of nanocarriers for miRNA delivery.
Diagnostic and therapeutic applications of ncRNA
Although no pure ncRNA therapeutics are yet fully FDA-approved, RNA-targeting therapies are approved; none are classic "non-coding" RNAs, such as miRNAs or lncRNAs, used directly as drugs. Approved agents are primarily synthetic antisense oligonucleotides (ASOs) and siRNAs designed to target specific mRNAs. Multiple siRNA drugs are now FDA-approved, representing the most mature class of therapeutic ncRNAs. Several ASO drugs are approved, targeting non-coding regions or mechanisms involving ncRNAs.11
Hence, the unique characteristics and regulatory capabilities of ncRNA make them valuable targets for diagnostic and therapeutic interventions, particularly in cancer (Table 1).
Table 1. Summary of evaluating ncRNA as biomarkers for cancer .
| ncRNA | Associated cancer | Application | Ref. |
| H19 (lncRNAs) | Gastric cancer | Diagnostic, prognostic | 12,13 |
| let-7 | Lung cancer | Diagnostic, prognostic | 14,15 |
| circHIPK3 | Liver & colorectal cancer | Diagnostic, prognostic, therapeutic target | 16,17 |
| HOTAIR (lncRNA) | Breast & colorectal cancers | Diagnostic, prognostic, diagnose metastasis | 18-23 |
| MALAT1 (lncRNA) | Lung & breast cancers | Diagnostic, prognostic, diagnose metastasis | 24,25 |
| lncRNA GAS5 | Breast & prostate cancer | Diagnostic, prognostic, therapeutic target | 26,27 |
| lncRNA PCA3 | Prostate cancer | Diagnostic, prognostic | 28,29 |
| circPVT1 | Gastric & colorectal cancer | Diagnostic, prognostic | 18,30-32 |
| lncRNA SChLAP1 | Prostate cancer | Diagnostic, prognostic, diagnose metastasis | 33,34 |
| piRNAs | Various cancers | Diagnostic, prognostic | 35,36 |
| miR-21 | Various cancers | Diagnostic, prognostic, therapeutic target | 37,38 |
| miR-155 | Breast Cancer & esophageal squamous cell carcinoma | Diagnostic, prognostic | 39-42 |
| miR-34a | Prostate, lung & breast cancers | Diagnostic, prognostic, therapeutic target | 43-46 |
| miR-125b | Breast & ovarian cancer | Diagnostic, prognostic, therapeutic target | 47-52 |
| miR-15b miR-21 |
Colorectal cancer | Diagnostic | NCT06738225 |
| miR-20a | Gastric cancer | Diagnostic | NCT05901376 |
| miR-21 | Gastric cancer | Diagnostic | NCT05901376 |
| miR-106b | Gastric cancer | Diagnostic | NCT05901376 |
| miR-199a | Gastric cancer | Diagnostic | NCT05901376 |
| miR-22 | Gastric cancer | Diagnostic | NCT05901376 |
| Sha-miR-71a | Bilharzial BlC | Diagnostic, prognostic | NCT05697224 |
Researchers have explored the use of ncRNAs as biomarkers for disease detection and prognosis.53 ncRNA-therapies can also provide enormous benefits, including ease of sequence design and creation, functional flexibility, charge and protection, and the opportunity for patient-specific management.54 In addition, ncRNAs can facilitate the conversion of proteins into the cellular cytoplasm without requiring nuclear entry, and are not expected to interact with the host genome, therefore verifying the safety of these treatments.55
By analyzing the expression profiles of specific ncRNAs, healthcare professionals can gain insights into disease states and tailor treatment strategies accordingly. Additionally, the therapeutic potential of ncRNA lies in its ability to regulate gene expression.
Small non-coding RNAs regulate gene expression post-transcriptionally, typically by binding to target mRNAs with partial complementarity, leading to mRNA cleavage or inhibition of protein synthesis. The outcome depends on the degree of complementarity between the siRNA/miRNA and its target. Perfect complementarity induces endonucleolytic cleavage of the mRNA. In contrast, imperfect pairing is more common in mammals, results in translational repression. This occurs either through disruption of the translational machinery (leading to truncated proteins) or by sequestering mRNAs into cytoplasmic P-bodies. Within P-bodies, mRNAs may undergo degradation by exonucleases or deadenylation by poly(A)-specific nucleases.56
The ability to regulate miRNA expression in vivo holds promise as a foundation for developing novel therapies. Several strategies have already been established to modulate miRNA levels. To increase miRNA activity, researchers can employ: (i) miRNA mimics, (ii) small synthetic double-stranded molecules that are processed into functional miRNAs, (iii) miRNA expression vectors to induce cellular miRNA production, or (iv) direct delivery of mature miRNAs. Conversely, to suppress miRNA activity, antagomirs and miRNA sponges synthetic sequences complementary to target miRNAs can be used to block their interaction with endogenous mRNA. Given that miRNAs play key roles in cancer-related processes such as cell proliferation, apoptosis, differentiation, invasion, metastasis, and tumorigenesis, these regulatory approaches may offer significant therapeutic potential.57
Like miRNAs, siRNAs act as post-transcriptional regulators and have been investigated for their therapeutic potential in various diseases, including cancer, hepatitis, and metabolic and genetic disorders. In recent years, siRNAs have garnered significant attention due to their potential clinical applications. Several miRNA- and siRNA-based therapies are currently under evaluation in clinical trials, and three Food and Drug Administration (FDA) approved RNA interference (RNAi) drugs based on siRNA are available for targeting primary hyperoxaluria type 1, acute hepatic porphyria, and transthyretin-mediated amyloidosis.56 Hence, diagnostic applications of small non-coding RNAs, such as miRNAs, which are stable in biofluids (blood, saliva, urine) and serve as non-invasive biomarkers for various cancers, including glioblastoma (e.g., miR-21) and breast cancer (e.g., miR-155). siRNAs are also used in liquid biopsies for detecting oncogenic mutations. piRNAs, though less studied, show promise in early-stage cancer detection (e.g., piR-823 in colorectal cancer). Therapeutic applications are a significant feature of ncRNA, where miRNA mimics (e.g., miR-34a) and antagomirs (anti-miRs) are utilized in clinical trials for cancer and cardiovascular diseases, respectively. Furthermore, siRNA-based drugs (e.g., Patisiran for amyloidosis) were used to leverage RNAi to silence disease-causing genes.58
Understanding tRNA modifications as medium ncRNAs is highly significant because even minor disruptions in this balance, such as the absence of a single tRNA, can lead to tissue degeneration or death. Recent studies have shown that tRNA expression can be post-transcriptionally regulated by microRNAs (miRNAs). Additionally, dysregulation in tRNA expression, misacylation by aminoacyl-tRNA synthetases, and tRNA hypomodification can all impact gene expression, potentially contributing to diseases such as cancer, neurodegenerative disorders, and metabolic conditions. Given that aberrant tRNA levels can modulate gene expression, deciphering the mechanisms controlling tRNA expression and the consequences of its dysregulation is crucial. Unraveling these processes could pave the way for novel therapeutic strategies, enabling targeted and personalized treatments for various diseases. 59
Small RNAs derived from tRNAs have garnered significant interest as potential biomarkers. Several studies have identified circulating tRNA-derived fragments (tRFs) as diagnostic tools for various diseases, including epilepsy, clear cell renal cell carcinoma, and gastric cancer (GC). In line with these findings, Wang et al observed significantly reduced plasma levels of specific tRFs—tRF-GluCTC-003, tRF-GlyCCC-007, tRF-GlyCCC-008, tRF-LeuCAA-003, tRF-SerTGA-001, and tRF-SerTGA-002—in patients with early breast cancer (EBC).60
The authors proposed that these 5′-derived tRFs could serve as potential biomarkers for the in situ diagnosis of EBC, though further validation with a larger sample size is required. In this line, Zhang et al. reported that tRF-3019a is overexpressed in GC and directly targets the tumor suppressor gene FBXO47 (F-box protein 47). Their findings revealed that tRF-3019a promotes GC malignancy by suppressing FBXO47, highlighting its critical role in GC progression. These results suggest that tRF-3019a may function as an oncogenic factor, positioning it as a promising diagnostic biomarker or therapeutic target for GC.61 Beyond GC-associated tRFs, Green et al also observed a significant downregulation of tRF-3003a in osteoarthritic cartilage, implicating its potential role in osteoarthritis (OA).56,62
Dysregulation of rRNA biogenesis kinetics, for example, in breast cancer (BC), is linked to elevated levels of intermediate rRNA species that are less efficient in mRNA translation. Recent studies suggest a silencing mechanism that inhibits pre-rRNA expression when rRNA processing is defective. Notably, ribosomes exhibit structural and functional heterogeneity, and their varying affinities for different mRNAs represent an emerging mechanism of translational control in gene expression. However, rRNA synthesis can be disrupted at multiple stages, including alterations in rDNA copy number, impaired rDNA transcription, and errors in rRNA processing and modification, ultimately leading to defective ribosome assembly. Such dysregulation may promote aberrant protein aggregation, thereby disrupting proteostasis.56
In this context, diagnostic applications of medium ncRNAs, such as tRFs, have been identified in the dysregulation of neurodegenerative diseases and cancers, serving as novel biomarkers. Also, snoRNAs (e.g., SNORD78 in lung cancer) correlate with tumor progression. 63 On the other hand, therapeutic applications of modified tRNAs are explored for suppressing nonsense mutations in genetic disorders. snoRNA-targeting therapies are also being tested for ribosomopathies and cancers.64
Linear lncRNAs are currently being explored in clinical trials as noninvasive biomarkers, detectable in circulating blood or urine. Their expression levels can indicate disease severity or reveal specific patterns in certain types of cancer. For example, Htoo et al demonstrated that elevated PCA3 lncRNA levels in urine correlate with prostate cancer progression. Similarly, Kumarswamy et al identified LIPCAR lncRNA in plasma as a potential prognostic biomarker for cardiovascular mortality. Additionally, Lorenzen et al showed that circulating TAPSAKI lncRNA levels could predict mortality in patients with acute kidney injury.56 To disrupt lncRNA activity, several strategies can be employed: (i) Transcriptional modulation: Altering the promoter activity of the lncRNA-coding region to suppress transcription. (ii) RNAi and antisense targeting: Using siRNAs, shRNAs, or modified antisense oligonucleotides (e.g., gapmers) to silence lncRNAs, which can lead to epigenetic derepression and subsequent activation of sense genes. (iii) Aptamer-based disruption: Employing aptamers to bind specific lncRNA structural domains, interfering with their interactions with binding partners. (iv) Ribozyme-mediated degradation: Utilizing ribozymes to cleave and degrade target lncRNAs. (v) Small-molecule/peptide inhibitors: Designing synthetic molecules or peptides to block lncRNA interactions with regulatory factors. To enhance or restore lncRNA expression to normal levels, strategies leveraging clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) technology can be employed. Specifically, a catalytically inactive Cas9 (dCas9) fused to the transcriptional activator domain VP64 can be used to activate a target promoter. However, plasmid or viral vector-based approaches may yield ambiguous results, as some lncRNAs influence gene expression within their native genomic contexts. While lncRNAs hold significant promise as therapeutic agents, several challenges hinder their full understanding and application. These include the lack of humanized models or organoid cultures, the involvement of lncRNAs in diverse molecular mechanisms, and their multifunctional roles.65 However, Diagnostic applications of lncRNAs are determined by HOTAIR (in breast cancer) and MALAT1 (in lung cancer) as prognostic markers.66 Linc-p21 is also associated with chemoresistance in multiple types of cancer. As a therapeutic target, ASO targeting lncRNAs (e.g., targeting NEAT1) is being evaluated in glioblastoma. Moreover, CRISPR-based lncRNA editing is being investigated for epigenetic modulation.67
Techniques such as RNAi utilize a variety of interfering RNAs to silence disease-causing or mimic silenced/haploinsufficient genes selectively to restore regular gene expression.2 In addition, the development of delivery systems, including nanocarriers, has further facilitated the efficient delivery of these RNA molecules to target cells, thereby opening new avenues for precise and personalized medicine.68
Overall, ncRNA-based systems have been explored in several syndromes, and many have progressed to clinical trials. However, to make an RNA product suitable for biomedical applications, specific conditions must be met, and RNA purity, stability, and bioactivity must be verified. So, in the following, a viewpoint on the key challenges and advanced approaches for the broad diagnostic and therapeutic applications (Fig. 4) of ncRNA is introduced.
Fig. 4.
ncRNA is used as a diagnostic and therapeutic indicator of cancer. (A) ncRNA-based treatments might target the ncRNA by exploiting RNAi therapeutic molecules and/or using tiny molecular suppressors of their protein associates. These helpful paths could be proper for oral or intravenous administration. Additionally, targeted therapies such as gene editing, gene silencing, and gene expression via nucleic acid nanoassembly have enhanced the chances of RNA therapy.(B) Tumor cells and various body fluids are used for diagnosis. ncRNA isolation and detection. The identification of ncRNAs associated with cancer has been facilitated by the advancement of several high-performance expression analysis technologies. Nucleic acid nanoassemblies for ncRNA detection and imaging may be employed for cancer identification and prognosis, and can serve as therapeutic biomarkers.
Diagnostic Advancements through Nanomedicine
One of the main areas where nanomedicine is making significant advances is in diagnosis. For instance, nanosensors are being developed to detect diseases at an early stage.9 In addition, nano-based sensors/biosensors can be designed to detect specific biomarkers or abnormal cellular activities with remarkable accuracy, enabling the early detection of conditions such as cancer, diabetes, infectious diseases, and neurological disorders.69
It has been verified that ncRNAs exhibit remarkable stability in whole blood, which can be utilized as novel biomarkers for specific syndromes, including cancers (Fig. 5).70 Among ncRNAs, miRNAs have been studied in most studies, and they are involved in many biological processes.71-74 For instance, most liver cancer-associated miRNAs have been investigated by Shi et al. in a clinical system.75 Based on their investigation, four miRNAs were up-regulated and five miRNAs were down-regulated in liver cancer tissues.
Fig. 5.
RNA-based therapeutic in cancer. ncRNAs via modification can be effective in the prevention of proliferation, metastasis, angiogenesis, drug resistance, DNA damage, and invasion.
The integration of nanomedicine with imaging modalities has revolutionized medical imaging, offering high-resolution images for precise diagnosis.76 For this case, NPs are being used as contrast agents in medical imaging techniques such as MRI and CT scans, allowing for more accurate and detailed visualization of tissues and organs.77
Another thrilling application of nanomedicine in diagnostic systems is the improvement of liquid biopsy tests.78 These tests can be applied to nanoscale technologies to detect and discover circulating tumor cells or fragments of tumor DNA in the circulation.79,80
Nanomedicine has revolutionized liquid biopsy procedures by improving the sensitivity and specificity of ncRNA detection. Liquid biopsies, which analyze biomarkers in bodily fluids such as blood, rely on nanotechnology to isolate microRNAs and lncRNAs from complex biological matrices. Liquid biopsies offer a non-invasive alternative to traditional tissue biopsies and can provide valuable information about the presence, progression, and treatment response of cancer. NPs, such as gold nanoparticles and magnetic beads, functionalized with antibodies or oligonucleotides, enable the selective capture of ncRNAs even at low concentrations. For example, a study by Wang et al demonstrated that silica-coated magnetic nanoparticles efficiently extracted exosomal microRNAs from plasma, enhancing detection limits by 100-fold compared to conventional methods.81 Moreover, gold nanoparticles and quantum dots enhance extraction efficiency by selectively binding to ncRNAs, enabling their isolation even at minimal concentrations.82 This approach minimizes sample loss and improves diagnostic accuracy, particularly in early-stage cancers where ncRNA levels are typically low.
Furthermore, advanced nanoplatforms for ncRNA enrichment are also introduced. Nanotechnology-based platforms, such as exosome isolation kits employing antibody-coated nanoparticles, have been instrumental in enriching tumor-derived exosomes containing ncRNAs. Exosomes, which carry ncRNAs, are crucial for cancer diagnostics but are challenging to isolate due to their small size. Silicon-based nanowires and polymer nanoparticles have demonstrated high affinity for exosomal ncRNAs, facilitating their purification from complex biofluids.82 A notable example is the use of lipid-based nanoparticles to extract exosomal lncRNAs in cancer patients, enabling early diagnosis with high accuracy. These nanoplatforms not only enhance yield but also preserve RNA integrity, ensuring reliable downstream analysis.83
The integration of nanomedicine in liquid biopsies holds immense potential for personalized medicine, particularly in oncology. For example, a study by Dogra et al84 demonstrated that nanoparticle-based enrichment of miR-155 in the blood of lung cancer patients correlated with treatment response, highlighting its prognostic value. Despite these advances, challenges such as standardization and biocompatibility remain. Future research should focus on optimizing nanoparticle designs for clinical scalability while minimizing off-target effects. As nanomedicine continues to evolve, its role in liquid biopsy-based ncRNA diagnostics is expected to expand, paving the way for earlier and more accurate disease detection.
To date, electrochemical, optical, and electromechanical systems (including mass, surface stress, and resonance) based on various biological responses have been developed using DNA-based biosensors for the recognition of cancer-associated biomarkers. Among several DNA-based biosensors, electrochemical ones offer an outstanding capacity for biomarker detection due to their striking benefits, including ease, rapidity, cost-effectiveness, and the opportunity for miniaturization.85,86 Most electrochemical assays were established to identify overexpressed oncogenic miRNAs by enhancing the monitoring of the signal from cancer cells and comparing it to that from healthy cells. Cancer suppressor miRNAs, which are underexpressed in cancer cells, are typically not targeted because their levels are frequently below the detection limits of the assays. Therefore, recently, more sensitive and specific systems have been developed for miRNA determination.87-90 In this case, a sensitive and specific technique for the electrocatalytic detection of target miRNA (miR-107) by gold-loaded nanoporous superparamagnetic magnetic nanocubes (Au-NPFe2O3NC) has been developed. The proposed system was employed to determine miR-107 levels in cancer cell lines with remarkable reproducibility and high specificity. RT-qPCR was applied as a standard method. The obtained system displayed a high translational potential for monitoring miRNAs in biological fluid samples.91
Therapeutic Innovations with NPs
Nanomedicine is updating the field of therapeutics by supporting targeted drug delivery and personalized medicine.92,93 NPs, such as liposomes and polymeric NPs as efficient nanocarriers, can be engineered and commonly applied to encapsulate drugs and deliver them directly to the site of action.94,95 This targeted approach lessens side effects and enriches the efficacy of the treatment. NP-based therapies can be functionalized with ligands that specifically bind to receptors on diseased cells, further enhancing drug delivery and reducing off-target effects.50,96 Additionally, nano-based therapies have shown promising results in battling multidrug-resistant pathogens and overcoming biological barriers.
Nanomedicine has shown great promise for cancer treatment.97,98 NP-based therapies, such as gold NPs and carbon nanotubes, can selectively target cancer cells while maintaining healthy tissues.99,100 These NPs can be loaded with chemotherapy drugs or therapeutic agents and delivered directly to the tumor site, maximizing the treatment's effectiveness.101 Additionally, nanorobots are being developed to navigate through the bloodstream and deliver drugs with precision, minimizing systemic toxicity.17,102
Some types of NPs have been discovered for ncRNA delivery, including liposomes, polymeric NPs, dendrimers, and inorganic NPs. NPs employed in the delivery of therapeutic ncRNAs for FDA-approved and clinical-stage candidates exhibit diverse structural characteristics tailored to enhance stability, targeting, and cellular uptake.
Liposomes, composed of lipid bilayers, are one of the most extensively investigated NPs for ncRNA delivery.103,104 They can be applied as encapsulated agents for both hydrophilic and hydrophobic ncRNAs, providing protection and controlled release. In this line, LNPs, such as those used in Patisiran (ONPATTRO®), feature ionizable lipids, phospholipids, and cholesterol to deliver siRNA, leveraging their biocompatibility and endosomal escape capabilities. PEG-lipids were used to encapsulate siRNA, enabling endosomal escape and hepatic delivery.105 Also, LNPs modified with targeting ligands (e.g., GalNAc for hepatocyte-specific delivery) are being verified for miRNA therapeutics in cancer and metabolic diseases.106
Polymeric NPs, on the other hand, are composed of biocompatible polymers, such as poly(lactic-co-glycolic acid) (PLGA) and polyethylene glycol (PEG). These NPs can be easily modified to improve stability, targeting, and release kinetics. Polymeric NPs, including PLGA and polyethyleneimine (PEI), offer controlled release and cationic surfaces for nucleic acid complexation. In clinical trials, novel formulations like cyclodextrin-based polymers (e.g., CALAA-01) and gold NPs functionalized with oligonucleotides demonstrate improved biocompatibility and tumor targeting.107
Dendrimers, with their highly branched structure, offer a high payload capacity and efficient cellular uptake. Inorganic NPs, such as gold NPs and quantum dots, offer unique optical and magnetic properties that can be exploited for imaging and therapeutic purposes.108-110 Additionally, exosome-based systems leverage natural vesicular structures for enhanced biodistribution. Key modifications, such as PEGylation and ligand conjugation (e.g., GalNAc for hepatocyte targeting), further refine pharmacokinetics and tissue specificity. These advancements highlight the critical role of nanoparticle design in overcoming biological barriers for effective ncRNA therapeutics.105
Furthermore, advancements in extracellular vesicle (EV)-based NPs and peptide-derived carriers offer promising alternatives with reduced immunogenicity. Despite challenges such as scalability and off-target effects, the integration of smart NPs responsive to pH, enzymes, or redox conditions holds promise for precision therapy. Collectively, these innovations underscore the pivotal role of nanotechnology in realizing the clinical potential of ncRNA therapeutics. Besides the potential of NP-based ncRNA therapeutics, several challenges and boundaries should be addressed carefully.111,112 One of the main worries is the stability and degradation of ncRNAs within the NPs.113 Nucleases and other enzymes introduced into the biological milieu can degrade ncRNAs, leading to reduced therapeutic efficacy.104,114 Chemical modifications and encapsulation strategies within protective matrices have been applied to overcome this issue.115 Another challenge is achieving targeted delivery of ncRNAs to specific cells or tissues.116 While surface modification of NPs with targeting ligands can enhance specificity, further optimization is still needed to ensure efficient and selective delivery. In addition, the immunogenicity and toxicity of NPs should be carefully evaluated to diminish adverse effects.
To improve the effectiveness of NP-based ncRNA therapeutics, several approaches have been discovered.113,116,117 NPs along with ligands or antibodies, can boost cellular uptake and targeting by surface modification.118-120 Therapeutic agents such as small molecules or proteins integrated with ncRNAs can enrich the effectiveness.121,122 Mixture therapy attitudes, where several ncRNAs or therapeutic compounds are conveyed simultaneously, have also exposed beneficial consequences. These policies aim to overcome the challenges of diseases by targeting multiple pathways or molecular targets simultaneously.
Recent innovations in NP-based ncRNA therapeutics have confirmed their potential in numerous syndromes.123-125 For instance, NP-based delivery of tumor suppressor ncRNAs has revealed promising potential in animal studies.126,127 Correspondingly, in Alzheimer's and Parkinson's, NP-mediated delivery of neuroprotective ncRNAs has displayed neurorestorative impacts.115 Some clinical evaluations are presently ongoing to assess the safety and efficiency of NP-based ncRNA therapeutics in humans, emphasizing their promising potential in the healthcare system.
Looking ahead, NP-based ncRNA therapeutics play a significant role in personalized medicine and disease-specific targeting.128-130 With advancements in nanomedicine and our understanding of ncRNA biology, it is possible to design NPs that can selectively deliver ncRNAs to specific cell types or disease sites.131,132 This opens up new avenues for precision medicine, where therapies can be tailored to individual patients based on their genetic profile and disease characteristics. Furthermore, NP-based ncRNA therapeutics can also be employed in gene editing and gene therapy applications, offering potential cures for genetic disorders.
Overall, therapeutic advances with NPs for ncRNA have developed the field of molecular medicine. NPs provide unique advantages in terms of stability, protection, and targeted delivery of ncRNA therapeutics. A comparative analysis of nano-delivery systems for ncRNAs reveals distinct advantages and limitations based on their design and composition. LNPs, especially liposomes, are widely used due to their high biocompatibility and efficient encapsulation of ncRNAs like siRNA and miRNA; however, they may suffer from instability and rapid clearance in vivo.70 Polymeric NPs, such as PLGA or chitosan, offer controlled release and protection against enzymatic degradation, but can exhibit cytotoxicity and low transfection efficiency.133 Inorganic NPs, such as gold or silica-based systems, provide tunable surfaces for functionalization and enhanced cellular uptake; however, their potential long-term toxicity and poor biodegradability remain concerns.134 Each system thus presents trade-offs between delivery efficiency, safety, and therapeutic applicability.
Emerging nano-delivery platforms, such as exosomes and hybrid systems, aim to overcome these limitations by leveraging natural biocompatibility and targeting capabilities. Exosomes, as endogenous vesicles, minimize immune responses and enhance the tissue-specific delivery of ncRNAs, but their large-scale production and heterogeneity pose challenges.135 Hybrid systems combining lipids, polymers, or inorganic materials
attempt to synergize the benefits of multiple approaches, improving stability and targeting precision.136 However, the complexity of fabrication and potential batch-to-batch variability may hinder clinical translation. The choice of delivery system ultimately depends on the specific ncRNA (e.g., siRNA, lncRNA, or circRNA), desired pharmacokinetics, and the target tissue, necessitating further optimization for personalized therapeutic applications.
Despite the challenges and limitations in therapeutic strategies, ongoing research in nanomedicine is paving the way for the development of safe and effective NP-based ncRNA therapeutics. With further optimization and clinical validation, these innovative approaches have the potential to transform the treatment landscape for various diseases, bringing us closer to the realization of personalized and precision medicine. In this context, the efficiency of treatment achieved through the transfer of ncRNA using nano-delivery systems for cancer treatment is represented in Table 2.
Table 2. Summary of cancer treatment with the delivery of ncRNA therapeutics through nano-delivery systems .
| ncRNA | Delivery system | Cancer type | Therapeutic impact | Ref. |
|---|---|---|---|---|
| MT1DP | Folate-modified liposome NPs | NSCLC | Raised erastin-induced ferroptosis by augmentation of Malondialdehyde and ROS levels, enhancement of intracellular Ferrous iron concentration, and reduction of glutathione levels | 137 |
| MDC1 | Thermosensitive magnetic cationic liposomes | Cervical cancer | Magnetic cationic liposomes cause Overwhelmed with definite adverse responses and improved the inhibition of cell growth related to cervical cancer |
138 |
| LINC01257 | Lipid NPs | AML | LNPs lessen cell count after 48 h of treatment, damage Kasumi-1 cell proliferation without disturbing healthy PBMCs | 109 |
| NRCP | DOPC nanoliposomes | Ovarian cancer | Considerably diminished tumor growth NRCP playing as a middle-associated partner between STAT1 and RNA polymerase II, leading to amplified expression of downstream target genes | 139 |
| Malat1 | Liposomal spherical nucleic acid constructs | Lung adenocarcinoma | Boosted the tumor suppressor, interferon-induced protein with IFIT2 | 140 |
| LCDR | NT-NPs | Lung adenocarcinoma | siLCDR/AUTP multiplexes precisely target the nucleus to suppress the effective gene, declining cancer growth of patient-derived xenografts of lung adenocarcinoma | 141 |
| CCAT1 | CSNPs | Colorectal cancer | Expressively limited HT-29 tumor growth, with suitable biosafety and biocompatibility in the animal model | 142 |
| lncAFAP1-AS1 | PDSA polymer NPs | Triple-negative breast cancer | Silencing lncAFAP1-AS1 expression and scavenging the elevated GSH, leading to synergistic reversal of radioresistance. Enhanced the radiosensitivity and improved the radiotherapy effect | 13 |
| DANCR | RGD-PEG-ECO NPs | Triple-negative breast cancer | Meaningfully limited the survival, invasion, migration, and proliferation of the TNBC cell lines | 38 |
| ANRIL | DTBP-3NP-siANRIL NPs | Hepatocellular carcinoma | Signaling the expression of miR-203a and its following genes and augmented the ratios of NK cells and T cells | 15 |
| MALAT1 | s-PGEA-FA NPs | Esophageal squamous cell carcinoma | Effectively inhibiting esophageal squamous cell carcinoma development | 143 |
| MEG3 | PuPGEA NPs | Hepatocellular carcinoma | Effectively inhibiting tumor growth and inducing tumor necrosis | 21 |
| MEG3 | CNC@CB8 @PGEA NPs | Hepatocellular carcinoma | Effectively inhibiting HCC tumor growth | 117 |
| MEG3 | PAMAM-PEG-EpDT3 NPs | CRPC | Noteworthy anti-CRPC outcome, both in the animal model and in vitro study | 22 |
| MALAT1 | ASO-Au-TAT NPs | Lung cancer | Reducing MALAT1 expression level, decreasing migration capability in vitro and reducing metastatic tumor nodule formation in an animal study | 23 |
| OUM1 | ICG-MOF-RGD NPs | UM | Conquers UM proliferation and metastasis and enhances cisplatin sensitivity in UM cells | 25 |
| MALAT1 | Single wall carbon nanotube (SWCNT)-antiMALAT1 | MM | Inducing DNA damage and cell apoptosis in vivo | 36 |
| LINC00589 | PMSNs | GC | Conquer the metastatic ability of GC cells in an animal model and in vitro study | 45 |
| miR-122 | Multivalent rubber-like RNA NPs | Liver cancer | Silencing of drug exporters and oncogenic proteins, as well as inhibition of tumor growth | 46 |
| miR-218 | LA-PAMAM | Liver cancer | Diminished tumor progression and amended liver histological features | 41 |
| miR-451 | calcium carbonate NPs | Bladder cancer | Suppression of multidrug resistance and augmented growth of intracellular Adr with anticancer properties | 42 |
| miR-199a-3p | Omentum-derived exosomes | Ovarian cancer | Inhibition of cell proliferation and invasion | 27 |
| miR-let-7c-5p | SiO2-polyethyleneimine NPs | Cervical cancer | Suppression of cell proliferation and migration | 144 |
| miR-200c | CXCR4-targeted polymeric poly-Lglutamic acid-coated NPs | Colon cancer | Enhanced immune responses against tumors | 29 |
| miR-139-5p | R9 modified with125 I-labeled RGD and Ce6 | Cancer in general | Boosted the radiotherapy sensitivity with low toxicity | 31 |
| let-7i | Nano-graphene oxide platform | Cancer in general | Retreated intracellular drug and improved photothermal therapy with chemical agents | 32 |
| miR-532-3p | PLGA-PEG-VB12 NPs | Gastric cancer | Mitochondrial impairment, amplified apoptosis, and limitation of cell proliferation | 51 |
| miR-181a | GDY-CeO2 nanozymes | Esophageal cancer | Improvement of tumor hypoxia and radiation-induced DNA damage, and inhibition of tumor growth | 52 |
| miR-15a and miR-16–1 | Cationic PEGylated niosomes | Prostate cancer | Augmented apoptosis of tumor cells | 34 |
| miR-320 | a combination of TAT-coated SLNs with peptides containing the NGR motif | Head and neck cancer | Declined Oxa-associated toxicities and high antitumor value | 145 |
| miR-181a | ZIF-8 nano-complexes | Rectal cancer | Improved radiosensitivity, limited proliferation, lessened migration, and boosted apoptosis | 146 |
| miR-30a-5p | MMNs | Ocular melanoma | Enriched pro-inflammatory anticancer immunity against skin cancer | 147 |
| MRX34 (miR-34a mimic) | Liposome | Solid tumor | Phase I terminated | NCT02862145 |
| Atu027 | Liposome | Solid tumors | Phase I completed | NCT00938574 |
| siG12D LODER | PLGA matrix | LAPC | Phase I completed Phase II |
NCT01188785 NCT01676259 |
| TKM-080301 | LNP | NET and ACC HCC Liver cancer |
Phase I/II completed Phase I/II completed Phase I completed |
NCT01262235 NCT02191878 NCT01437007 |
| EphA2 siRNA | DOPC neutral liposome | Advanced or recurrent solid tumor | Phase I | NCT01591356 |
| NU-0129 | Gold nanoparticle | Glioblastoma | Phase I completed | NCT03020017 |
| ALN-VSP02 | Co-delivery of two siRNAs with LNP | Advanced solid tumor with liver involvement |
Phase I completed | NCT00882180 |
| CALAA-01 | Cyclodextrin nanoparticles targeting transferrin receptor |
Solid tumor | Phase I terminated | NCT00689065 |
| DCR-MYC | LNP | Solid tumor, multiple myeloma, or lymphoma HCC |
Phase I terminated Phase I/II terminated |
NCT02110563 NCT02314052 |
NSCLC, non-small-cell lung cancer; AML,acute myeloid leukemia; CRPC, Castration-resistant prostate cancer; UM,Uveal melanoma; MM, Multiple myeloma; GC,gastric cancer.
Commercialization opportunities, overcoming obstacles, and conclusion
The convergence of ncRNA and nanomedicine presents substantial commercialization opportunities in the healthcare industry. Although traditional detection methods for ncRNAs, such as qRT-PCR, northern blotting, and microarray, are widely applied, they have some limitations that discourage their use, including laborious techniques, long processing times, sample size requirements, varying sensitivities of the kits and instruments, and false-positive results. In terms of technical aspects, analytical companies, utilizing sensor and biosensor devices, can develop non-invasive tests that use nano-sensors/biosensors to identify ncRNAs as biomarkers, thereby providing accurate and timely disease detection. Pharmaceutical companies, alternatively, can capitalize on the targeted drug delivery systems offered by nanomedicine, improving drug efficacy and controlled release. This can be achieved by constructing more stable, longer-lasting, and less toxic antisense or mimic oligonucleotides to downregulate or upregulate a specific ncRNA, respectively, for therapeutic purposes.
Advances in RNAi technologies, such as siRNA and miRNA-based therapies, have led to FDA-approved treatments, including Patisiran, for hereditary transthyretin amyloidosis. Nanocarriers, such as LNPs and polymeric nanoparticles, enhance delivery efficiency, reducing off-target effects and improving bioavailability.
Companies like Alnylam and Moderna are leveraging these innovations, with increasing investments in RNA-nanomedicine hybrids for the treatment of cancer, cardiovascular diseases, and rare genetic disorders. Additionally, diagnostics utilizing exosomal ncRNAs as biomarkers for early cancer detection present lucrative opportunities for biotechnology firms. However, scalability, manufacturing consistency, and regulatory hurdles remain key challenges to widespread adoption.148
While the prospects are promising, several challenges must be addressed for effective commercialization to occur. Safety concerns surrounding the use of nanomaterials, regulatory frameworks, and manufacturing scalability are among the key hurdles. Stability issues, immune system clearance, and inefficient tissue targeting limit therapeutic efficacy. Recent innovations, such as exosome-based carriers, offer improved biocompatibility and natural tropism for specific cells, enhancing delivery precision.
Clinical trials, such as those investigating exosome-delivered miR-34a for solid tumors (NCT03608631), highlight both promise and pitfalls, e.g., variable patient responses and manufacturing complexities. Furthermore, regulatory agencies demand rigorous safety assessments, necessitating standardized protocols for nanoparticle characterization. Collaborative efforts between academia, industry, and regulators are essential to address these challenges.
Nanorobotics holds immense potential in the field of ncRNA therapeutics and diagnostics, yet several challenges must be addressed for clinical translation. One major obstacle is the precise delivery of ncRNA molecules to target cells without degradation or off-target effects. Nanorobots equipped with molecular recognition systems can enhance specificity by binding to overexpressed biomarkers on diseased cells, thereby improving the accuracy of therapeutic interventions. Additionally, advancements in biocompatible materials and propulsion mechanisms, such as magnetic or enzymatic propulsion, are overcoming biological barriers, including immune clearance and vascular dynamics.149 Nanorobotics integrated with ncRNA-based therapies offers unprecedented control over gene regulation and disease detection. For instance, nanorobots carrying CRISPR-Cas9 and guide RNA can perform precise gene editing, while those loaded with fluorescent reporters enable real-time imaging of tumor-associated ncRNAs. Furthermore, adaptive nanorobots can respond to microenvironmental cues (e.g., pH, enzymes) to release payloads selectively, minimizing systemic toxicity. Such innovations bridge the gap between ncRNA biology and clinical applications, paving the way for personalized medicine.150
Despite progress, scalability and long-term safety remain hurdles in nanorobotic-ncRNA systems. Manufacturing nanorobots with uniform properties at scale requires sophisticated techniques, such as DNA origami or 3D nanoprinting. Immunogenicity and unintended biodistribution also pose risks, necessitating rigorous preclinical testing. Computational modeling and AI-driven design are being employed to predict the behavior of nanorobots in vivo, thereby optimizing their efficacy and safety profiles. Addressing these challenges will be critical for regulatory approval and clinical adoption.150
The convergence of nanorobotics and ncRNA nanomedicine is revolutionizing therapeutic and diagnostic paradigms. By overcoming delivery barriers, enhancing precision, and improving biocompatibility, nanorobots are unlocking new avenues for treating cancers, genetic disorders, and infectious diseases. Future research should focus on scalable fabrication, smart responsiveness, and rigorous clinical trials to translate these technologies from bench to bedside. As the field advances, interdisciplinary collaboration will be key to harnessing the full potential of nanorobotics in ncRNA medicine.
Exosomes have also emerged as promising carriers for ncRNAs due to their biocompatibility, low immunogenicity, and ability to cross biological barriers. These nanoscale vesicles facilitate intercellular communication by transferring functional ncRNAs to target cells, modulating gene expression, and cellular functions. Their endogenous origin minimizes toxicity and enhances stability, making them superior to synthetic nanoparticles for therapeutic delivery. Additionally, exosomes can be engineered to enhance targeting efficiency, allowing for the precise delivery of ncRNA-based therapeutics in diseases such as cancer and neurodegenerative disorders. Despite their potential, exosome-based ncRNA delivery faces challenges, including low yield during isolation, heterogeneity, and inefficient loading of therapeutic ncRNAs. Advances in nanotechnology have addressed these issues by optimizing isolation techniques (e.g., ultracentrifugation, size-exclusion chromatography) and developing novel loading strategies, such as electroporation and sonication. Surface modification with ligands (e.g., peptides, antibodies) enhances tissue-specific targeting, while genetic engineering of parent cells allows for customized exosome production.
Furthermore, integrating exosomes with synthetic nanoparticles (hybrid systems) improves payload capacity and pharmacokinetics, overcoming limitations in clinical scalability. Hence, Exosome-based carriers represent a transformative platform for ncRNA delivery, bridging the gap between nanomedicine and clinical applications. While challenges remain in standardization and large-scale production, ongoing advancements in bioengineering and nanotechnology are paving the way for scalable, targeted therapies. Additionally, translating scientific discoveries into marketable products requires substantial investments in developing basic research, conducting clinical trials, and protecting intellectual property.
Nanomedicine and ncRNA represent cutting-edge fields with tremendous potential in the field of healthcare. The complex regulatory roles of ncRNA and the precision of nanotechnology hold promising solutions for the development of innovative diagnostic and therapeutic approaches. By leveraging the power of these technologies, we can advance disease detection, enhance treatment outcomes, and pave the way for a more personalized and efficient healthcare system.
In conclusion, the potential synergy derived from combining ncRNA and nanomedicine offers a pathway to address unmet medical needs. By driving scientific advancements, fostering collaborations, and embracing commercialization opportunities, we can unlock the full potential of these cutting-edge technologies and shape the future of healthcare systems. Further advancements in this interdisciplinary field will contribute to the progression of precision medicine and patient satisfaction.
Review Highlights
What is the current knowledge?
ncRNAs, previously considered 'junk DNA' and insignificant, are now being documented for their remarkably extraordinary regulatory roles in gene expression and various cellular processes.
Technical aspects and potential of commercializing the design and targeting of ncRNAs using nanocarriers and nano-delivery systems are confirmed.
What is new here?
The enormous benefits of ncRNA therapies include ease of sequence design and creation, functional flexibility, charge and protection, and the opportunity for patient-specific management.
Competing Interests
The authors declare no competing interests.
Data Availability Statement
Not applicable.
Ethical Approval
Not applicable.
Funding Statement
This research received no external funding.
References
- 1.Tsagakis I, Douka K, Birds I, Aspden JL. Long non-coding RNAs in development and disease: conservation to mechanisms. J Pathol. 2020;250:480–95. doi: 10.1002/path.5405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Taft RJ, Pang KC, Mercer TR, Dinger M, Mattick JS. Non-coding RNAs: regulators of disease. J Pathol. 2010;220:126–39. doi: 10.1002/path.2638. [DOI] [PubMed] [Google Scholar]
- 3.Yu AD, Wang Z, Morris KV. Long noncoding RNAs: a potent source of regulation in immunity and disease. Immunol Cell Biol. 2015;93:277–83. doi: 10.1038/icb.2015.2. [DOI] [PubMed] [Google Scholar]
- 4.Haleem A, Javaid M, Singh RP, Rab S, Suman R. Applications of nanotechnology in medical field: a brief review. Glob Health J. 2023;7:70–7. doi: 10.1016/j.glohj.2023.02.008. [DOI] [Google Scholar]
- 5. Maurice PA, Hochella MF. Nanoscale particles and processes: a new dimension in soil science. In: Advances in Agronomy. Academic Press. 2008. p. 123-53. doi: 10.1016/s0065-2113(08)00605-6. [DOI]
- 6.Kreyling WG, Semmler-Behnke M, Chaudhry Q. A complementary definition of nanomaterial. Nano Today. 2010;5:165–8. doi: 10.1016/j.nantod.2010.03.004. [DOI] [Google Scholar]
- 7.Coradduzza D, Bellu E, Congiargiu A, Pashchenko A, Amler E, Necas A, et al. Role of nano-miRNAs in diagnostics and therapeutics. Int J Mol Sci. 2022;23:6836. doi: 10.3390/ijms23126836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Domingues C, Santos A, Alvarez-Lorenzo C, Concheiro A, Jarak I, Veiga F, et al. Where is nano today and where is it headed? A review of nanomedicine and the dilemma of nanotoxicology. ACS Nano. 2022;16:9994–10041. doi: 10.1021/acsnano.2c00128. [DOI] [PubMed] [Google Scholar]
- 9.Sargazi S, Mukhtar M, Rahdar A, Bilal M, Barani M, Díez-Pascual AM, et al. Opportunities and challenges of using high-sensitivity nanobiosensors to detect long noncoding RNAs: a preliminary review. Int J Biol Macromol. 2022;205:304–15. doi: 10.1016/j.ijbiomac.2022.02.082. [DOI] [PubMed] [Google Scholar]
- 10.Arshad R, Fatima I, Sargazi S, Rahdar A, Karamzadeh-Jahromi M, Pandey S, et al. Novel perspectives towards RNA-based nano-theranostic approaches for cancer management. Nanomaterials (Basel) 2021;11:3330. doi: 10.3390/nano11123330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Grillone K, Caridà G, Luciano F, Cordua A, Di Martino MT, Tagliaferri P, et al. A systematic review of non-coding RNA therapeutics in early clinical trials: a new perspective against cancer. J Transl Med. 2024;22:731. doi: 10.1186/s12967-024-05554-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Beylerli O, Gareev I, Sufianov A, Ilyasova T, Guang Y. Long noncoding RNAs as promising biomarkers in cancer. Noncoding RNA Res. 2022;7:66–70. doi: 10.1016/j.ncrna.2022.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bi Z, Li Q, Dinglin X, Xu Y, You K, Hong H, et al. Nanoparticles (NPs)-meditated lncRNA AFAP1-AS1 silencing to block Wnt/β-catenin signaling pathway for synergistic reversal of radioresistance and effective cancer radiotherapy. Adv Sci (Weinh) 2020;7:2000915. doi: 10.1002/advs.202000915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Shen C, Li J, Che G. Prognostic value of let-7 in lung cancer: systematic review and meta-analysis. Transl Cancer Res. 2020;9:6354–61. doi: 10.21037/tcr-20-1240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wang T, Li P, Wan T, Tu B, Li J, Huang F. TIGIT/PVR and lncRNA ANRIL dual-targetable PAMAM polymeric nanoparticles efficiently inhibited the hepatoma carcinoma by combination of immunotherapy and gene therapy. J Drug Target. 2021;29:783–91. doi: 10.1080/1061186x.2021.1879088. [DOI] [PubMed] [Google Scholar]
- 16.Zhang Y, Liu Q, Liao Q. CircHIPK3: a promising cancer-related circular RNA. Am J Transl Res. 2020;12:6694–704. [PMC free article] [PubMed] [Google Scholar]
- 17.Li J, Esteban-Fernández de Ávila B, Gao W, Zhang L, Wang J. Micro/nanorobots for biomedicine: delivery, surgery, sensing, and detoxification. Sci Robot. 2017;2:eaam6431. doi: 10.1126/scirobotics.aam6431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Lin Z, Tang X, Wang L, Ling L. Prognostic and clinicopathological value of circPVT1 in human cancers: a meta-analysis. Cancer Rep (Hoboken) 2021;4:e1385. doi: 10.1002/cnr2.1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Shi Y, Huang Q, Kong X, Zhao R, Chen X, Zhai Y, et al. Current knowledge of long non-coding RNA HOTAIR in breast cancer progression and its application. Life (Basel) 2021;11:483. doi: 10.3390/life11060483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Chen L, Qian X, Wang Z, Zhou X. The HOTAIR lncRNA: a remarkable oncogenic promoter in human cancer metastasis. Oncol Lett. 2021;21:302. doi: 10.3892/ol.2021.12563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ren Y, Li RQ, Cai YR, Xia T, Yang M, Xu FJ. Effective codelivery of lncRNA and pDNA by pullulan-based nanovectors for promising therapy of hepatocellular carcinoma. Adv Funct Mater. 2016;26:7314–25. doi: 10.1002/adfm.201603041. [DOI] [Google Scholar]
- 22.Tai Z, Ma J, Ding J, Pan H, Chai R, Zhu C, et al. Aptamer-functionalized dendrimer delivery of plasmid-encoding lncRNA MEG3 enhances gene therapy in castration-resistant prostate cancer. Int J Nanomedicine. 2020;15:10305–20. doi: 10.2147/ijn.S282107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Gong N, Teng X, Li J, Liang XJ. Antisense oligonucleotide-conjugated nanostructure-targeting lncRNA MALAT1 inhibits cancer metastasis. ACS Appl Mater Interfaces. 2019;11:37–42. doi: 10.1021/acsami.8b18288. [DOI] [PubMed] [Google Scholar]
- 24.Goyal B, Yadav SRM, Awasthee N, Gupta S, Kunnumakkara AB, Gupta SC. Diagnostic, prognostic, and therapeutic significance of long non-coding RNA MALAT1 in cancer. Biochim Biophys Acta Rev Cancer. 2021;1875:188502. doi: 10.1016/j.bbcan.2021.188502. [DOI] [PubMed] [Google Scholar]
- 25.Li Y, Li F, Pan H, Huang X, Yu J, Liu X, et al. Targeted OUM1/PTPRZ1 silencing and synergetic CDT/enhanced chemical therapy toward uveal melanoma based on a dual-modal imaging-guided manganese metal-organic framework nanoparticles. J Nanobiotechnology. 2022;20:472. doi: 10.1186/s12951-022-01643-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yu Y, Hann SS. Novel tumor suppressor lncRNA growth arrest-specific 5 (GAS5) in human cancer. Onco Targets Ther. 2019;12:8421–36. doi: 10.2147/ott.S221305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kobayashi M, Sawada K, Miyamoto M, Shimizu A, Yamamoto M, Kinose Y, et al. Exploring the potential of engineered exosomes as delivery systems for tumor-suppressor microRNA replacement therapy in ovarian cancer. Biochem Biophys Res Commun. 2020;527:153–61. doi: 10.1016/j.bbrc.2020.04.076. [DOI] [PubMed] [Google Scholar]
- 28.Lemos AE, Matos AD, Ferreira LB, Gimba ER. The long non-coding RNA PCA3: an update of its functions and clinical applications as a biomarker in prostate cancer. Oncotarget. 2019;10:6589–603. doi: 10.18632/oncotarget.27284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Nguyen HT, Phung CD, Tran TH, Pham TT, Pham LM, Nguyen TT, et al. Manipulating immune system using nanoparticles for an effective cancer treatment: combination of targeted therapy and checkpoint blockage miRNA. J Control Release. 2021;329:524–37. doi: 10.1016/j.jconrel.2020.09.034. [DOI] [PubMed] [Google Scholar]
- 30.Rezasoltani M, Forouzesh F, Salehi Z, Zabihi MR, Rejali L, Nazemalhosseini-Mojarad E. Identification of LncPVT1 and CircPVT1 as prognostic biomarkers in human colorectal polyps. Sci Rep. 2023;13:13113. doi: 10.1038/s41598-023-40288-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Wang H, Wang Z, Chen W, Wang W, Shi W, Chen J, et al. Self-assembly of photosensitive and radiotherapeutic peptide for combined photodynamic-radio cancer therapy with intracellular delivery of miRNA-139-5p. Bioorg Med Chem. 2021;44:116305. doi: 10.1016/j.bmc.2021.116305. [DOI] [PubMed] [Google Scholar]
- 32.Yan J, Zhang Y, Zheng L, Wu Y, Wang T, Jiang T, et al. Let‐7i miRNA and platinum loaded nano‐graphene oxide platform for detection/reversion of drug resistance and synergetic chemical‐photothermal inhibition of cancer cell. Chin Chem Lett. 2022;33:767–72. doi: 10.1016/j.cclet.2021.08.018. [DOI] [Google Scholar]
- 33.Kidd SG, Carm KT, Bogaard M, Olsen LG, Bakken AC, Løvf M, et al. High expression of SCHLAP1 in primary prostate cancer is an independent predictor of biochemical recurrence, despite substantial heterogeneity. Neoplasia. 2021;23:634–41. doi: 10.1016/j.neo.2021.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ghaffari M, Kalantar SM, Hemati M, Dehghani Firoozabadi A, Asri A, Shams A, et al. Co-delivery of miRNA-15a and miRNA-16-1 using cationic PEGylated niosomes downregulates Bcl-2 and induces apoptosis in prostate cancer cells. Biotechnol Lett. 2021;43:981–94. doi: 10.1007/s10529-021-03085-2. [DOI] [PubMed] [Google Scholar]
- 35.Hanusek K, Poletajew S, Kryst P, Piekiełko-Witkowska A, Bogusławska J. piRNAs and PIWI proteins as diagnostic and prognostic markers of genitourinary cancers. Biomolecules. 2022;12:186. doi: 10.3390/biom12020186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hu Y, Lin J, Fang H, Fang J, Li C, Chen W, et al. Targeting the MALAT1/PARP1/LIG3 complex induces DNA damage and apoptosis in multiple myeloma. Leukemia. 2018;32:2250–62. doi: 10.1038/s41375-018-0104-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bautista-Sánchez D, Arriaga-Canon C, Pedroza-Torres A, De La Rosa-Velázquez IA, González-Barrios R, Contreras-Espinosa L, et al. The promising role of miR-21 as a cancer biomarker and its importance in RNA-based therapeutics. Mol Ther Nucleic Acids. 2020;20:409–20. doi: 10.1016/j.omtn.2020.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Vaidya AM, Sun Z, Ayat N, Schilb A, Liu X, Jiang H, et al. Systemic delivery of tumor-targeting siRNA nanoparticles against an oncogenic lncRNA facilitates effective triple-negative breast cancer therapy. Bioconjug Chem. 2019;30:907–19. doi: 10.1021/acs.bioconjchem.9b00028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Zheng YJ, Liang TS, Wang J, Zhao JY, Zhai SN, Yang DK, et al. MicroRNA-155 acts as a diagnostic and prognostic biomarker for oesophageal squamous cell carcinoma. Artif Cells Nanomed Biotechnol. 2020;48:977–82. doi: 10.1080/21691401.2020.1773479. [DOI] [PubMed] [Google Scholar]
- 40.Wang F, Wang J, Zhang H, Fu B, Zhang Y, Jia Q, et al. Diagnostic value of circulating miR-155 for breast cancer: a meta-analysis. Front Oncol. 2024;14:1374674. doi: 10.3389/fonc.2024.1374674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Elfiky AM, Mohamed RH, Abd El-Hakam FE, Yassin MA, ElHefnawi M. Targeted delivery of miR-218 via decorated hyperbranched polyamidoamine for liver cancer regression. Int J Pharm. 2021;610:121256. doi: 10.1016/j.ijpharm.2021.121256. [DOI] [PubMed] [Google Scholar]
- 42.Wei S, Gao J, Zhang M, Dou Z, Li W, Zhao L. Dual delivery nanoscale device for miR-451 and adriamycin co-delivery to combat multidrug resistant in bladder cancer. Biomed Pharmacother. 2020;122:109473. doi: 10.1016/j.biopha.2019.109473. [DOI] [PubMed] [Google Scholar]
- 43.Imani S, Wu RC, Fu J. MicroRNA-34 family in breast cancer: from research to therapeutic potential. J Cancer. 2018;9:3765–75. doi: 10.7150/jca.25576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Li XJ, Ren ZJ, Tang JH. MicroRNA-34a: a potential therapeutic target in human cancer. Cell Death Dis. 2014;5:e1327. doi: 10.1038/cddis.2014.270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wang S, Wo L, Zhang Z, Zhu C, Wang C, Wang Y, et al. Delivery of LINC00589 via mesoporous silica nanoparticles inhibits peritoneal metastasis in gastric cancer. Cancer Lett. 2022;549:215916. doi: 10.1016/j.canlet.2022.215916. [DOI] [PubMed] [Google Scholar]
- 46.Wang H, Ellipilli S, Lee WJ, Li X, Vieweger M, Ho YS, et al. Multivalent rubber-like RNA nanoparticles for targeted co-delivery of paclitaxel and MiRNA to silence the drug efflux transporter and liver cancer drug resistance. J Control Release. 2021;330:173–84. doi: 10.1016/j.jconrel.2020.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wang Y, Zeng G, Jiang Y. The emerging roles of miR-125b in cancers. Cancer Manag Res. 2020;12:1079–88. doi: 10.2147/cmar.S232388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Bi YN, Guan JP, Wang L, Li P, Yang FX. Clinical significance of microRNA-125b and its contribution to ovarian carcinogenesis. Bioengineered. 2020;11:939–48. doi: 10.1080/21655979.2020.1814660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chen Z, Guo X, Sun S, Lu C, Wang L. Serum miR-125b levels associated with epithelial ovarian cancer (EOC) development and treatment responses. Bioengineered. 2020;11:311–7. doi: 10.1080/21655979.2020.1736755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Masoudi Asil S, Ahlawat J, Guillama Barroso G, Narayan M. Nanomaterial based drug delivery systems for the treatment of neurodegenerative diseases. Biomater Sci. 2020;8:4109–28. doi: 10.1039/d0bm00809e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Chen Z, Liang Y, Feng X, Liang Y, Shen G, Huang H, et al. Vitamin-B12-conjugated PLGA-PEG nanoparticles incorporating miR-532-3p induce mitochondrial damage by targeting apoptosis repressor with caspase recruitment domain (ARC) on CD320-overexpressed gastric cancer. Mater Sci Eng C Mater Biol Appl. 2021;120:111722. doi: 10.1016/j.msec.2020.111722. [DOI] [PubMed] [Google Scholar]
- 52.Zhou X, You M, Wang F, Wang Z, Gao X, Jing C, et al. Multifunctional graphdiyne-cerium oxide nanozymes facilitate microRNA delivery and attenuate tumor hypoxia for highly efficient radiotherapy of esophageal cancer. Adv Mater. 2021;33:e2100556. doi: 10.1002/adma.202100556. [DOI] [PubMed] [Google Scholar]
- 53.Xu MD, Qi P, Du X. Long non-coding RNAs in colorectal cancer: implications for pathogenesis and clinical application. Mod Pathol. 2014;27:1310–20. doi: 10.1038/modpathol.2014.33. [DOI] [PubMed] [Google Scholar]
- 54.Sullenger BA, Nair S. From the RNA world to the clinic. Science. 2016;352:1417–20. doi: 10.1126/science.aad8709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Weng Y, Li C, Yang T, Hu B, Zhang M, Guo S, et al. The challenge and prospect of mRNA therapeutics landscape. Biotechnol Adv. 2020;40:107534. doi: 10.1016/j.biotechadv.2020.107534. [DOI] [PubMed] [Google Scholar]
- 56.Baptista B, Riscado M, Queiroz JA, Pichon C, Sousa F. Non-coding RNAs: emerging from the discovery to therapeutic applications. Biochem Pharmacol. 2021;189:114469. doi: 10.1016/j.bcp.2021.114469. [DOI] [PubMed] [Google Scholar]
- 57.Simonson B, Das S. MicroRNA therapeutics: the next magic bullet? Mini Rev Med Chem. 2015;15:467–74. doi: 10.2174/1389557515666150324123208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Jain CK, Srivastava P, Pandey AK, Singh N, Kumar RS. miRNA therapeutics in precision oncology: a natural premium to nurture. Explor Target Antitumor Ther. 2022;3:511–32. doi: 10.37349/etat.2022.00098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Santos M, Fidalgo A, Varanda AS, Oliveira C, Santos MAS. tRNA deregulation and its consequences in cancer. Trends Mol Med. 2019;25:853–65. doi: 10.1016/j.molmed.2019.05.011. [DOI] [PubMed] [Google Scholar]
- 60.Wang J, Ma G, Li M, Han X, Xu J, Liang M, et al. Plasma tRNA fragments derived from 5' ends as novel diagnostic biomarkers for early-stage breast cancer. Mol Ther Nucleic Acids. 2020;21:954–64. doi: 10.1016/j.omtn.2020.07.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Zhang F, Shi J, Wu Z, Gao P, Zhang W, Qu B, et al. A 3'-tRNA-derived fragment enhances cell proliferation, migration and invasion in gastric cancer by targeting FBXO47. Arch Biochem Biophys. 2020;690:108467. doi: 10.1016/j.abb.2020.108467. [DOI] [PubMed] [Google Scholar]
- 62.Green JA, Ansari MY, Ball HC, Haqqi TM. tRNA-derived fragments (tRFs) regulate post-transcriptional gene expression via AGO-dependent mechanism in IL-1β stimulated chondrocytes. Osteoarthritis Cartilage. 2020;28:1102–10. doi: 10.1016/j.joca.2020.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Xiao L, Wang J, Ju S, Cui M, Jing R. Disorders and roles of tsRNA, snoRNA, snRNA and piRNA in cancer. J Med Genet. 2022;59:623–31. doi: 10.1136/jmedgenet-2021-108327. [DOI] [PubMed] [Google Scholar]
- 64.Porter JJ, Heil CS, Lueck JD. Therapeutic promise of engineered nonsense suppressor tRNAs. Wiley Interdiscip Rev RNA. 2021;12:e1641. doi: 10.1002/wrna.1641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Boon RA, Jaé N, Holdt L, Dimmeler S. Long noncoding RNAs: from clinical genetics to therapeutic targets? J Am Coll Cardiol. 2016;67:1214–26. doi: 10.1016/j.jacc.2015.12.051. [DOI] [PubMed] [Google Scholar]
- 66.Nazari M, Babakhanzadeh E, Mollazadeh A, Ahmadzade M, Mohammadi Soleimani E, Hajimaqsoudi E. HOTAIR in cancer: diagnostic, prognostic, and therapeutic perspectives. Cancer Cell Int. 2024;24:415. doi: 10.1186/s12935-024-03612-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Yadav B, Pal S, Rubstov Y, Goel A, Garg M, Pavlyukov M, et al. LncRNAs associated with glioblastoma: from transcriptional noise to novel regulators with a promising role in therapeutics. Mol Ther Nucleic Acids. 2021;24:728–42. doi: 10.1016/j.omtn.2021.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Mahmoodi Chalbatani G, Dana H, Gharagouzloo E, Grijalvo S, Eritja R, Logsdon CD, et al. Small interfering RNAs (siRNAs) in cancer therapy: a nano-based approach. Int J Nanomedicine. 2019;14:3111–28. doi: 10.2147/ijn.S200253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Leong YX, Tan EX, Leong SX, Lin Koh CS, Thanh Nguyen LB, Ting Chen JR, et al. Where nanosensors meet machine learning: prospects and challenges in detecting disease X. ACS Nano. 2022;16:13279–93. doi: 10.1021/acsnano.2c05731. [DOI] [PubMed] [Google Scholar]
- 70. Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov 2021. 20: 101-24. doi: 10.1038/s41573-020-0090-8. [DOI] [PMC free article] [PubMed]
- 71.Gong J, He XX, Tian A. Emerging role of microRNA in hepatocellular carcinoma (review) Oncol Lett. 2015;9:1027–33. doi: 10.3892/ol.2014.2816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ulitsky I, Bartel DP. lincRNAs: genomics, evolution, and mechanisms. Cell. 2013;154:26–46. doi: 10.1016/j.cell.2013.06.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Zekri AN, Youssef AS, El-Desouky ED, Ahmed OS, Lotfy MM, Nassar AA, et al. Serum microRNA panels as potential biomarkers for early detection of hepatocellular carcinoma on top of HCV infection. Tumour Biol. 2016;37:12273–86. doi: 10.1007/s13277-016-5097-8. [DOI] [PubMed] [Google Scholar]
- 74.Falahi S, Rafiee-Pour HA, Zarejousheghani M, Rahimi P, Joseph Y. Non-coding RNA-based biosensors for early detection of liver cancer. Biomedicines. 2021;9:964. doi: 10.3390/biomedicines9080964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Shi KQ, Lin Z, Chen XJ, Song M, Wang YQ, Cai YJ, et al. Hepatocellular carcinoma associated microRNA expression signature: integrated bioinformatics analysis, experimental validation and clinical significance. Oncotarget. 2015;6:25093–108. doi: 10.18632/oncotarget.4437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zhang K, Sun Y, Wu S, Zhou M, Zhang X, Zhou R, et al. Systematic imaging in medicine: a comprehensive review. Eur J Nucl Med Mol Imaging. 2021;48:1736–58. doi: 10.1007/s00259-020-05107-z. [DOI] [PubMed] [Google Scholar]
- 77.Siddique S, Chow JCL. Application of nanomaterials in biomedical imaging and cancer therapy. Nanomaterials (Basel) 2020;10:1700. doi: 10.3390/nano10091700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Tadimety A, Closson A, Li C, Yi S, Shen T, Zhang JXJ. Advances in liquid biopsy on-chip for cancer management: technologies, biomarkers, and clinical analysis. Crit Rev Clin Lab Sci. 2018;55:140–62. doi: 10.1080/10408363.2018.1425976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Viswanath B, Kim S. Recent insights into the development of nanotechnology to detect circulating tumor cells. Trends Analyt Chem. 2016;82:191–8. doi: 10.1016/j.trac.2016.05.026. [DOI] [Google Scholar]
- 80.Shen Z, Wu A, Chen X. Current detection technologies for circulating tumor cells. Chem Soc Rev. 2017;46:2038–56. doi: 10.1039/c6cs00803h. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Wang J, Wuethrich A, Lobb RJ, Antaw F, Sina AA, Lane RE, et al. Characterizing the heterogeneity of small extracellular vesicle populations in multiple cancer types via an ultrasensitive chip. ACS Sens. 2021;6:3182–94. doi: 10.1021/acssensors.1c00358. [DOI] [PubMed] [Google Scholar]
- 82.Naranbat D, Herdes E, Tapinos N, Tripathi A. Review of microRNA detection workflows from liquid biopsy for disease diagnostics. Expert Rev Mol Med. 2025;27:e11. doi: 10.1017/erm.2025.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Huang M, Yang J, Wang T, Song J, Xia J, Wu L, et al. Homogeneous, low-volume, efficient, and sensitive quantitation of circulating exosomal PD-L1 for cancer diagnosis and immunotherapy response prediction. Angew Chem Int Ed Engl. 2020;59:4800–5. doi: 10.1002/anie.201916039. [DOI] [PubMed] [Google Scholar]
- 84.Dogra P, Shinglot V, Ruiz-Ramírez J, Cave J, Butner JD, Schiavone C, et al. Translational modeling-based evidence for enhanced efficacy of standard-of-care drugs in combination with anti-microRNA-155 in non-small-cell lung cancer. Mol Cancer. 2024;23:156. doi: 10.1186/s12943-024-02060-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Bartosik M, Jirakova L. Electrochemical analysis of nucleic acids as potential cancer biomarkers. Curr Opin Electrochem. 2019;14:96–103. doi: 10.1016/j.coelec.2019.01.002. [DOI] [Google Scholar]
- 86.Abdul Rashid JI, Yusof NA. The strategies of DNA immobilization and hybridization detection mechanism in the construction of electrochemical DNA sensor: a review. Sens Biosensing Res. 2017;16:19–31. doi: 10.1016/j.sbsr.2017.09.001. [DOI] [Google Scholar]
- 87.Juracek J, Peltanova B, Dolezel J, Fedorko M, Pacik D, Radova L, et al. Genome-wide identification of urinary cell-free microRNAs for non-invasive detection of bladder cancer. J Cell Mol Med. 2018;22:2033–8. doi: 10.1111/jcmm.13487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Ciui B, Jambrec D, Sandulescu R, Cristea C. Bioelectrochemistry for miRNA detection. Curr Opin Electrochem. 2017;5:183–92. doi: 10.1016/j.coelec.2017.09.014. [DOI] [Google Scholar]
- 89.Chen YX, Huang KJ, Niu KX. Recent advances in signal amplification strategy based on oligonucleotide and nanomaterials for microRNA detection-a review. Biosens Bioelectron. 2018;99:612–24. doi: 10.1016/j.bios.2017.08.036. [DOI] [PubMed] [Google Scholar]
- 90.Kilic T, Erdem A, Ozsoz M, Carrara S. microRNA biosensors: opportunities and challenges among conventional and commercially available techniques. Biosens Bioelectron. 2018;99:525–46. doi: 10.1016/j.bios.2017.08.007. [DOI] [PubMed] [Google Scholar]
- 91.Islam MN, Masud MK, Nguyen NT, Gopalan V, Alamri HR, Alothman ZA, et al. Gold-loaded nanoporous ferric oxide nanocubes for electrocatalytic detection of microRNA at attomolar level. Biosens Bioelectron. 2018;101:275–81. doi: 10.1016/j.bios.2017.09.027. [DOI] [PubMed] [Google Scholar]
- 92.Bar-Zeev M, Livney YD, Assaraf YG. Targeted nanomedicine for cancer therapeutics: towards precision medicine overcoming drug resistance. Drug Resist Updat. 2017;31:15–30. doi: 10.1016/j.drup.2017.05.002. [DOI] [PubMed] [Google Scholar]
- 93.Germain M, Caputo F, Metcalfe S, Tosi G, Spring K, Åslund AKO, et al. Delivering the power of nanomedicine to patients today. J Control Release. 2020;326:164–71. doi: 10.1016/j.jconrel.2020.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Chamundeeswari M, Jeslin J, Verma ML. Nanocarriers for drug delivery applications. Environ Chem Lett. 2019;17:849–65. doi: 10.1007/s10311-018-00841-1. [DOI] [Google Scholar]
- 95.Kumari P, Ghosh B, Biswas S. Nanocarriers for cancer-targeted drug delivery. J Drug Target. 2016;24:179–91. doi: 10.3109/1061186x.2015.1051049. [DOI] [PubMed] [Google Scholar]
- 96.Liu R, Luo C, Pang Z, Zhang J, Ruan S, Wu M, et al. Advances of nanoparticles as drug delivery systems for disease diagnosis and treatment. Chin Chem Lett. 2023;34:107518. doi: 10.1016/j.cclet.2022.05.032. [DOI] [Google Scholar]
- 97.Youn YS, Bae YH. Perspectives on the past, present, and future of cancer nanomedicine. Adv Drug Deliv Rev. 2018;130:3–11. doi: 10.1016/j.addr.2018.05.008. [DOI] [PubMed] [Google Scholar]
- 98.Bharali DJ, Mousa SA. Emerging nanomedicines for early cancer detection and improved treatment: current perspective and future promise. Pharmacol Ther. 2010;128:324–35. doi: 10.1016/j.pharmthera.2010.07.007. [DOI] [PubMed] [Google Scholar]
- 99.Shi J, Kantoff PW, Wooster R, Farokhzad OC. Cancer nanomedicine: progress, challenges and opportunities. Nat Rev Cancer. 2017;17:20–37. doi: 10.1038/nrc.2016.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Bastiancich C, Danhier P, Préat V, Danhier F. Anticancer drug-loaded hydrogels as drug delivery systems for the local treatment of glioblastoma. J Control Release. 2016;243:29–42. doi: 10.1016/j.jconrel.2016.09.034. [DOI] [PubMed] [Google Scholar]
- 101.Kemp JA, Shim MS, Heo CY, Kwon YJ. "Combo" nanomedicine: co-delivery of multi-modal therapeutics for efficient, targeted, and safe cancer therapy. Adv Drug Deliv Rev. 2016;98:3–18. doi: 10.1016/j.addr.2015.10.019. [DOI] [PubMed] [Google Scholar]
- 102.Agrahari V, Agrahari V, Chou ML, Chew CH, Noll J, Burnouf T. Intelligent micro-/nanorobots as drug and cell carrier devices for biomedical therapeutic advancement: promising development opportunities and translational challenges. Biomaterials. 2020;260:120163. doi: 10.1016/j.biomaterials.2020.120163. [DOI] [PubMed] [Google Scholar]
- 103.Alzhrani R, Alsaab HO, Petrovici A, Bhise K, Vanamala K, Sau S, et al. Improving the therapeutic efficiency of noncoding RNAs in cancers using targeted drug delivery systems. Drug Discov Today. 2020;25:718–30. doi: 10.1016/j.drudis.2019.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Abdellatif AA, Scagnetti G, Younis MA, Bouazzaoui A, Tawfeek HM, Aldosari BN, et al. Non-coding RNA-directed therapeutics in lung cancer: delivery technologies and clinical applications. Colloids Surf B Biointerfaces. 2023;229:113466. doi: 10.1016/j.colsurfb.2023.113466. [DOI] [PubMed] [Google Scholar]
- 105.Jung HN, Lee SY, Lee S, Youn H, Im HJ. Lipid nanoparticles for delivery of RNA therapeutics: current status and the role of in vivo imaging. Theranostics. 2022;12:7509–31. doi: 10.7150/thno.77259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Hosseini-Kharat M, Bremmell KE, Prestidge CA. Why do lipid nanoparticles target the liver? Understanding of biodistribution and liver-specific tropism. Mol Ther Methods Clin Dev. 2025;33:101436. doi: 10.1016/j.omtm.2025.101436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Piotrowski-Daspit AS, Kauffman AC, Bracaglia LG, Saltzman WM. Polymeric vehicles for nucleic acid delivery. Adv Drug Deliv Rev. 2020;156:119–32. doi: 10.1016/j.addr.2020.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Smith ES, Whitty E, Yoo B, Moore A, Sempere LF, Medarova Z. Clinical applications of short non-coding RNA-based therapies in the era of precision medicine. Cancers (Basel) 2022;14:1588. doi: 10.3390/cancers14061588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Connerty P, Moles E, de Bock CE, Jayatilleke N, Smith JL, Meshinchi S, et al. Development of siRNA-loaded lipid nanoparticles targeting long non-coding RNA LINC01257 as a novel and safe therapeutic approach for t(8;21) pediatric acute myeloid leukemia. Pharmaceutics. 2021;13:1681. doi: 10.3390/pharmaceutics13101681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Garbo S, Maione R, Tripodi M, Battistelli C. Next RNA therapeutics: the mine of non-coding. Int J Mol Sci. 2022;23:7471. doi: 10.3390/ijms23137471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Hassanzadeh A, Shamlou S, Yousefi N, Nikoo M, Verdi J. Genetically-modified stem cell in regenerative medicine and cancer therapy; a new era. Curr Gene Ther. 2022;22:23–39. doi: 10.2174/1566523221666210707125342. [DOI] [PubMed] [Google Scholar]
- 112.Gavas S, Quazi S, Karpiński TM. Nanoparticles for cancer therapy: current progress and challenges. Nanoscale Res Lett. 2021;16:173. doi: 10.1186/s11671-021-03628-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Di Mauro V, Barandalla-Sobrados M, Catalucci D. The noncoding-RNA landscape in cardiovascular health and disease. Noncoding RNA Res. 2018;3:12–9. doi: 10.1016/j.ncrna.2018.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Renganathan A, Felley-Bosco E. Long noncoding RNAs in cancer and therapeutic potential. In: Rao MR, ed. Long Non-Coding RNA Biology. Singapore: Springer. 2017. p. 199-222. doi: 10.1007/978-981-10-5203-3_7. [DOI] [PubMed]
- 115.Mokhtarzadeh A, Alibakhshi A, Hashemi M, Hejazi M, Hosseini V, de la Guardia M, et al. Biodegradable nano-polymers as delivery vehicles for therapeutic small non-coding ribonucleic acids. J Control Release. 2017;245:116–26. doi: 10.1016/j.jconrel.2016.11.017. [DOI] [PubMed] [Google Scholar]
- 116.Vilaca A, de Windt LJ, Fernandes H, Ferreira L. Strategies and challenges for non-viral delivery of non-coding RNAs to the heart. Trends Mol Med. 2023;29:70–91. doi: 10.1016/j.molmed.2022.10.002. [DOI] [PubMed] [Google Scholar]
- 117.Wei C, Xu Y, Shen Q, Li R, Xiao X, Saw PE, et al. Role of long non-coding RNAs in cancer: from subcellular localization to nanoparticle-mediated targeted regulation. Mol Ther Nucleic Acids. 2023;33:774–93. doi: 10.1016/j.omtn.2023.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Yoo J, Park C, Yi G, Lee D, Koo H. Active targeting strategies using biological ligands for nanoparticle drug delivery systems. Cancers (Basel) 2019;11:640. doi: 10.3390/cancers11050640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Moku G, Gopalsamuthiram VR, Hoye TR, Panyam J. Surface modification of nanoparticles: methods and applications. In: Pinson J, Thiry D, eds. Surface Modification of Polymers. John Wiley & Sons. 2019. p. 317-46. doi: 10.1002/9783527819249.ch11. [DOI]
- 120.Lu J, Wang J, Ling D. Surface engineering of nanoparticles for targeted delivery to hepatocellular carcinoma. Small. 2018;14:1702037. doi: 10.1002/smll.201702037. [DOI] [PubMed] [Google Scholar]
- 121.Zhao R, Fu J, Zhu L, Chen Y, Liu B. Designing strategies of small-molecule compounds for modulating non-coding RNAs in cancer therapy. J Hematol Oncol. 2022;15:14. doi: 10.1186/s13045-022-01230-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Yu AM, Jian C, Yu AH, Tu MJ. RNA therapy: are we using the right molecules? Pharmacol Ther. 2019;196:91–104. doi: 10.1016/j.pharmthera.2018.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Luo M, Lee LK, Peng B, Choi CH, Tong WY, Voelcker NH. Delivering the promise of gene therapy with nanomedicines in treating central nervous system diseases. Adv Sci (Weinh) 2022;9:e2201740. doi: 10.1002/advs.202201740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Abdelmaksoud NM, Sallam AM, Abulsoud AI, El-Dakroury WA, Abdel Mageed SS, Al-Noshokaty TM, et al. Unraveling the role of miRNAs in the diagnosis, progression, and therapeutic intervention of Alzheimer's disease. Pathol Res Pract. 2024;253:155007. doi: 10.1016/j.prp.2023.155007. [DOI] [PubMed] [Google Scholar]
- 125.Yoon J, Shin M, Lee JY, Lee SN, Choi JH, Choi JW. RNA interference (RNAi)-based plasmonic nanomaterials for cancer diagnosis and therapy. J Control Release. 2022;342:228–40. doi: 10.1016/j.jconrel.2022.01.012. [DOI] [PubMed] [Google Scholar]
- 126.Buocikova V, Rios-Mondragon I, Pilalis E, Chatziioannou A, Miklikova S, Mego M, et al. Epigenetics in breast cancer therapy-new strategies and future nanomedicine perspectives. Cancers (Basel) 2020;12:3622. doi: 10.3390/cancers12123622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Duan H, Liu Y, Gao Z, Huang W. Recent advances in drug delivery systems for targeting cancer stem cells. Acta Pharm Sin B. 2021;11:55–70. doi: 10.1016/j.apsb.2020.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Liu S. Epigenetics advancing personalized nanomedicine in cancer therapy. Adv Drug Deliv Rev. 2012;64:1532–43. doi: 10.1016/j.addr.2012.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Almughem FA, Aldossary AM, Tawfik EA, Alomary MN, Alharbi WS, Alshahrani MY, et al. Cystic fibrosis: overview of the current development trends and innovative therapeutic strategies. Pharmaceutics. 2020;12:616. doi: 10.3390/pharmaceutics12070616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Slack FJ, Chinnaiyan AM. The role of non-coding RNAs in oncology. Cell. 2019;179:1033–55. doi: 10.1016/j.cell.2019.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Miller AD. Nanomedicine therapeutics and diagnostics are the goal. Ther Deliv. 2016;7:431–56. doi: 10.4155/tde-2016-0020. [DOI] [PubMed] [Google Scholar]
- 132.Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, et al. Tumor abnormality-oriented nanomedicine design. Chem Rev. 2023;123:10920–89. doi: 10.1021/acs.chemrev.3c00062. [DOI] [PubMed] [Google Scholar]
- 133.Kulkarni JA, Thomson SB, Zaifman J, Leung J, Wagner PK, Hill A, et al. Spontaneous, solvent-free entrapment of siRNA within lipid nanoparticles. Nanoscale. 2020;12:23959–66. doi: 10.1039/d0nr06816k. [DOI] [PubMed] [Google Scholar]
- 134.Zhang M, Yang D, Dong C, Huang H, Feng G, Chen Q, et al. Two-dimensional MXene-originated in situ nanosonosensitizer generation for augmented and synergistic sonodynamic tumor nanotherapy. ACS Nano. 2022;16:9938–52. doi: 10.1021/acsnano.2c04630. [DOI] [PubMed] [Google Scholar]
- 135.Liang Y, Lehrich BM, Zheng S, Lu M. Emerging methods in biomarker identification for extracellular vesicle-based liquid biopsy. J Extracell Vesicles. 2021;10:e12090. doi: 10.1002/jev2.12090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Lam K, Leung A, Martin A, Wood M, Schreiner P, Palmer L, et al. Unsaturated, trialkyl ionizable lipids are versatile lipid-nanoparticle components for therapeutic and vaccine applications. Adv Mater. 2023;35:e2209624. doi: 10.1002/adma.202209624. [DOI] [PubMed] [Google Scholar]
- 137.Gai C, Liu C, Wu X, Yu M, Zheng J, Zhang W, et al. MT1DP loaded by folate-modified liposomes sensitizes erastin-induced ferroptosis via regulating miR-365a-3p/NRF2 axis in non-small cell lung cancer cells. Cell Death Dis. 2020;11:751. doi: 10.1038/s41419-020-02939-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Ye H, Chu X, Cao Z, Hu X, Wang Z, Li M, et al. A novel targeted therapy system for cervical cancer: co-delivery system of antisense lncRNA of MDC1 and oxaliplatin magnetic thermosensitive cationic liposome drug carrier. Int J Nanomedicine. 2021;16:1051–66. doi: 10.2147/ijn.S258316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Rupaimoole R, Lee J, Haemmerle M, Ling H, Previs RA, Pradeep S, et al. Long noncoding RNA ceruloplasmin promotes cancer growth by altering glycolysis. Cell Rep. 2015;13:2395–402. doi: 10.1016/j.celrep.2015.11.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Sprangers AJ, Hao L, Banga RJ, Mirkin CA. Liposomal spherical nucleic acids for regulating long noncoding RNAs in the nucleus. Small. 2017;13:1602753. doi: 10.1002/smll.201602753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Yang X, Wen Y, Liu S, Duan L, Liu T, Tong Z, et al. LCDR regulates the integrity of lysosomal membrane by hnRNP K-stabilized LAPTM5 transcript and promotes cell survival. Proc Natl Acad Sci U S A. 2022;119:e2110428119. doi: 10.1073/pnas.2110428119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Jia F, Li Y, Deng X, Wang X, Cui X, Lu J, et al. Self-assembled fluorescent hybrid nanoparticles-mediated collaborative lncRNA CCAT1 silencing and curcumin delivery for synchronous colorectal cancer theranostics. J Nanobiotechnology. 2021;19:238. doi: 10.1186/s12951-021-00981-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Li RQ, Ren Y, Liu W, Pan W, Xu FJ, Yang M. MicroRNA-mediated silence of onco-lncRNA MALAT1 in different ESCC cells via ligand-functionalized hydroxyl-rich nanovectors. Nanoscale. 2017;9:2521–30. doi: 10.1039/c6nr09668a. [DOI] [PubMed] [Google Scholar]
- 144.Shao L, Wang R, Sun Y, Yue Z, Sun H, Wang X, et al. Delivery of microRNA-let-7c-5p by biodegradable silica nanoparticles suppresses human cervical carcinoma cell proliferation and migration. J Biomed Nanotechnol. 2020;16:1600–11. doi: 10.1166/jbn.2020.2989. [DOI] [PubMed] [Google Scholar]
- 145.Lo YL, Lin HC, Tseng WH. Tumor pH-functionalized and charge-tunable nanoparticles for the nucleus/cytoplasm-directed delivery of oxaliplatin and miRNA in the treatment of head and neck cancer. Acta Biomater. 2022;153:465–80. doi: 10.1016/j.actbio.2022.09.027. [DOI] [PubMed] [Google Scholar]
- 146.Hao J, Yan Q, Li Z, Liu X, Peng J, Zhang T, et al. Multifunctional miR181a nanoparticles promote highly efficient radiotherapy for rectal cancer. Mater Today Adv. 2022;16:100317. doi: 10.1016/j.mtadv.2022.100317. [DOI] [Google Scholar]
- 147.Ma Y, Lin H, Wang P, Yang H, Yu J, Tian H, et al. A miRNA-based gene therapy nanodrug synergistically enhances pro-inflammatory antitumor immunity against melanoma. Acta Biomater. 2023;155:538–53. doi: 10.1016/j.actbio.2022.11.016. [DOI] [PubMed] [Google Scholar]
- 148.Sasso JM, Ambrose BJB, Tenchov R, Datta RS, Basel MT, DeLong RK, et al. The progress and promise of RNA medicine─an arsenal of targeted treatments. J Med Chem. 2022;65:6975–7015. doi: 10.1021/acs.jmedchem.2c00024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Xu M, Qin Z, Chen Z, Wang S, Peng L, Li X, et al. Nanorobots mediated drug delivery for brain cancer active targeting and controllable therapeutics. Discov Nano. 2024;19:183. doi: 10.1186/s11671-024-04131-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Kong X, Gao P, Wang J, Fang Y, Hwang KC. Advances of medical nanorobots for future cancer treatments. J Hematol Oncol. 2023;16:74. doi: 10.1186/s13045-023-01463-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Not applicable.






