Back to Journals » Journal of Inflammation Research » Volume 18

RNA-Based Therapies in Kidney Diseases

Authors Hu L, Jin T, Zhang N, Ding J, Li L 

Received 7 November 2024

Accepted for publication 18 February 2025

Published 4 March 2025 Volume 2025:18 Pages 3143—3160

DOI https://doi.org/10.2147/JIR.S505252

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 3

Editor who approved publication: Dr Adam Bachstetter



Liang Hu,1,* Ting Jin,2,* Ning Zhang,3 Jin Ding,2 Lina Li3,4

1Department of Urology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321001, People’s Republic of China; 2Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321001, People’s Republic of China; 3College of Medicine, Jinhua University of Vocational Technology, Jinhua, Zhejiang, 321016, People’s Republic of China; 4Faculty of Medicine, Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Jin Ding; Lina Li, Email [email protected]; [email protected]

Abstract: Kidney diseases are major global health challenges, affecting over 750 million people worldwide. Despite significant efforts, effective treatment strategies are still insufficient. In recent years, RNA therapeutics have made substantial progress, and an increasing number of nucleic acid-based therapies have been approved, showing potential for treating various diseases (including kidney diseases). These therapies can target proteins, transcripts, and genes that were previously considered “undruggable”, allowing for the regulation of their expression and the expansion of therapeutic targets. Among RNA therapeutics, mRNA-based therapies are particularly promising because they can rapidly express therapeutic proteins, along with their design flexibility and potential to target previously inaccessible disease mechanisms. This review discussed various RNA-based strategies for developing new treatments, including antisense and RNA interference technologies, mRNA-based approaches, and CRISPR-Cas-mediated genome editing. Additionally, we highlighted the opportunities and challenges associated with the widespread application of these therapies in kidney disease treatment.

Keywords: RNA-based therapies, mRNA therapy, kidney diseases

Introduction

Kidney disease is a complex disease characterized by high morbidity and mortality, affecting over 750 million individuals worldwide and representing a significant global public health challenge.1 Recent studies have highlighted that the prevalence of chronic kidney disease (CKD) is increasing globally, becoming one of the leading risk factors for mortality worldwide.2 Although numerous studies have been conducted in recent years aimed at enhancing our understanding of kidney diseases, current research is still insufficient to elucidate the specific mechanisms involved and develop effective treatment strategies.3 Furthermore, emerging challenges (such as population aging, obesity, and climate change) are expected to exacerbate the global burden of kidney diseases, underscoring the urgency of addressing these issues.4,5 Consequently, there is a pressing need for more comprehensive research to address these existing gaps.

Recent advancements in RNA technology have significantly enhanced various fields (vaccine development, protein expression, etc)., providing a safe and efficient strategy for the expression or inhibition of specific genes. The RNA interference phenomenon, first described in the 1980s as a gene silencing mechanism triggered by double-stranded RNA, lays the foundation for small RNA (siRNA) technology. With breakthroughs made by scientists such as Karikó6 in nucleoside base modification, mRNA therapy has been gradually established as a cutting-edge and revolutionary treatment strategy. Furthermore, RNA technology encompasses antisense oligonucleotides (ASOs) and nucleic acid aptamers. ASOs consist of specific short, single-stranded DNA or RNA that complementarily bind to target mRNA sequences, interfering with and regulating the expression of mRNA. Since research began in the 1980s, ASOs have demonstrated the potential to alter gene expression and treat genetic diseases, cancer, and viral infections.7 Nucleic acid aptamers are single-stranded small molecules composed of RNA or DNA, exhibiting high specificity and affinity for their target molecules and thus exerting regulatory effects. Following the invention of the Systemic Evolution of Exponentially Enriched Ligands (SELEX) in the 1990s, aptamers with their easy synthesis, stability, and modifiable properties have been extensively studied across various fields, including diagnosis, treatment, and biological detection.8

In recent years, the potential of RNA-based therapies in kidney diseases has gained significant attention. Several studies have explored RNA-based therapies for inherited kidney diseases, which are often characterized by a lack of curative therapies. Bondue et al have reviewed RNA-based approaches for inherited kidney diseases, with a focus on the importance of developing efficient delivery vehicles (such as lipid nanoparticles) to protect RNA molecules from degradation and to enhance targeted delivery to kidney-specific cells.9 Moreover, they have also discussed strategies for improving the targeting of the glomerulus and tubules using antibodies, peptides, and small molecules, which are vital for kidney-targeted therapies.

In addition to inherited kidney diseases, RNA-based therapies have also shown promise in the treatment of acute kidney injury (AKI), a disease associated with high mortality rates. Haddad et al have explored the role of noncoding RNAs (ncRNAs) in the pathogenesis of ischemic AKI.10 In short, they have discussed how microRNAs, long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs) contribute to kidney injury, as well as their potential as biomarkers and therapeutic targets. This review highlighted the need for further exploration of ncRNAs, particularly their application in AKI-targeted therapy.

This review outlined the advantages and classifications of RNA-based therapies, with a focus on the applications and recent advancements of RNA-based therapies in the treatment of kidney diseases. This review aimed to promote research on RNA as a therapeutic agent for kidney diseases and to provide solutions to the challenges posed by insufficient treatment strategies in this area.

The Advantage of RNA-Based Therapies

By now, various RNA-based medications have been identified and applied in numerous areas (such as vaccines, treatments, and diagnostic tools) due to their significant benefits. This review highlighted several important advantages of these innovations compared to conventional therapeutic agents.

Targeting Multiple Genetic Components

The advantages of RNA-based therapy in targeting multiple genetic components are primarily reflected in its wide application range and high efficiency. RNA drugs can target almost any genetic component within cells, including certain targets that remain inaccessible to other technologies, including small molecules and antibodies that are often referred to as “undruggable” targets. For instance, many ncRNAs, especially small RNAs, possess few distinguishing features apart from their RNA sequences. Importantly, ncRNAs significantly outnumber proteins in the human genome11 playing a crucial role in the pathogenesis of various human diseases.12–14 Therefore, RNA drugs designed to target these molecules hold enormous therapeutic potential.

Several RNA drugs targeting ncRNAs have been developed and tested, showing promising results. For example, anti-miR21 is a therapeutic RNA drug that targets microRNA-21, which is involved in fibrosis and the progression of various cancers and diseases, including kidney diseases.15 Additionally, ASOs targeting lncRNAs have been explored for the treatment of kidney diseases, such as the use of ASOs targeting lncRNA KCNQ1OT1 in polycystic kidney disease models.16 These approaches underscore the significant therapeutic potential of ncRNA-targeting RNA therapeutics in treating various kidney conditions.

Moreover, prior studies have indicated that less than one-third of human proteins are amenable to effective targeting by small molecules.17,18 Current RNA therapeutics can precisely target these proteins both in vivo and in vitro.19,20 RNA therapy primarily exerts two mechanistic effects in treating undruggable targets: (1) utilizing coding RNA for protein or antigen expression, and (2) employing ncRNA to bind and regulate mRNA function (such as promoting degradation or inhibiting translation).21

Simple and Flexible Development and Manufacturing Process

In 2020, two breakthrough mRNA vaccines were released in quick succession in response to the COVID-19 pandemic, setting a record for the fastest vaccine development.22–24 Typically, the process of developing new small molecule or antibody drugs spans several years. However, RNA-based technologies facilitate rapid production due to their uniform action mechanism within cells. Moreover, RNA-based production technology has been optimized for the application of various RNA products. Notably, RNA-based production systems are automated, encompassing synthesis, isolation, and purification processes. A key advantage of mRNA therapeutics is the in vitro transcription (IVT) process, which is relatively simple and adaptable for large-scale manufacturing. The IVT process allows for rapid synthesis of mRNA from a DNA template, providing a scalable and flexible approach for producing therapeutic mRNA molecules. Consequently, RNA-based technology can be prepared for production in a shorter time than traditional pharmaceuticals. Compared with traditional small molecule drugs and protein-based therapies, mRNA therapeutics are faster and developed more easily, involving fewer steps in the production process. For instance, a drug based on siRNA aimed at targeting illness caused by overexpression of a certain gene in a specific organ may also be adapted to treat other ailments within that organ by merely modifying the siRNA sequence, thus facilitating personalized RNA treatment.

Long-Term Impact

Although natural RNA is readily degraded by nucleases, its stability can be significantly enhanced through various modifications during synthesis.25 One such breakthrough modification involves introducing pseudouridines into mRNA structure, which can stabilize the mRNA, reduce immune activation, and enhance translation efficiency. Moreover, when these RNAs are encapsulated in carriers (such as liposomes), they are effectively protected against nuclease attacks following systemic administration,26 thereby extending their lifespan. Another remarkable strategy involves the use of self-amplifying mRNA replicons, which can prolong the duration of mRNA drugs or vaccines and enhance immune responses.27,28

Recent studies have also emphasized the significant role of miRNAs in the progression of kidney diseases and their potential therapeutic benefits. For instance, miRNA clusters (such as miR-23a/27a/26a) have been proven to ameliorate renal tubulointerstitial fibrosis in diabetic nephropathy, highlighting their application prospects in RNA-based therapies for kidney diseases.29 Additionally, miRNA-429-3p has been found to play a pivotal role in ferroptosis regulation during CKD, presenting a potential target for therapeutic intervention.30

In addition to miRNAs, recent studies have also emphasized the significant roles of lncRNAs in the context of kidney diseases. For example, Xing et al31 have explored how lncRNAs regulate the fibroblast-to-myofibroblast transition, which is a critical process in renal fibrosis. Similarly, Lin et al32 have discovered that lncRNA AP001007 protects renal cells from injury in sepsis-induced AKI. Xie et al33 have identified lncRNA6524 as a key mediator of renal fibrosis through the Wnt/β-catenin signaling pathway. Furthermore, Li et al34 have demonstrated that the YY1-induced upregulation of lncRNA-ARAP1-AS2 promotes diabetic kidney fibrosis by altering glycolysis. Finally, Yang et al35 have shown that FTO modulates the m6A modification of lncRNA SNHG14, playing a crucial role in AKI. These findings emphasize the therapeutic potential of targeting lncRNAs in kidney diseases and provide promising new avenues for treatment strategies.

Moreover, recent studies have highlighted the therapeutic potential of certain chemical drugs and natural products in regulating lncRNAs for the treatment of kidney diseases. Luo et al36 have demonstrated that quercetin improves contrast-induced acute kidney injury (CI-AKI) through the HIF-1α/lncRNA NEAT1/HMGB1 pathway. This study highlighted the therapeutic potential of quercetin in regulating lncRNA NEAT1 to reduce cell injury and apoptosis in CI-AKI. Additionally, as revealed by Xie et al37 hederagenin (HDG), a natural compound derived from Astragalus membranaceus, alleviates cisplatin-induced AKI by inhibiting the lncRNA-A330074k22Rik/Axin2/β-catenin signaling pathway. These findings suggest that HDG may be used to modulate lncRNA-related pathways in AKI treatment.

No Genotoxic Risk Profile

Compared to DNA therapy, RNA therapy exhibits no significant genotoxic effects. DNA-based therapies involve delivering DNA molecules into cells via viral vectors, which carry the risk of integrating into the genome and inducing mutations. Genotoxicity is typically defined as the ability of a substance to cause genetic damage that leads to mutations, which may result in cancer or other genetic disorders. DNA-based therapies, especially those using integrating viral vectors, have been shown to carry the risk of insertional mutagenesis, where the inserted DNA can disrupt the host genome and potentially lead, to oncogenic effects. The use of RNA instead of DNA can effectively mitigate this potential risk. Specifically, RNA therapies do not integrate into the host genome, and their effects are transient, further minimizing the risk of permanent genetic alterations. Therefore, compared to DNA therapies, RNA therapies are considered to have a significantly lower genotoxic risk, as RNA does not permanently remain in the cells and does not induce persistent changes in genetic material.

RNA Therapies in Kidney Disease

Currently, the drug market is primarily composed of two categories of drugs: small molecules and proteins.38 However, these molecules have a limited range of disease targets for proteins or genes and cannot adequately address the required therapeutic needs.39 Based on the characteristics of RNA drugs, RNA therapeutics can be divided into three categories. The first category includes oligonucleotide therapeutics, which interact with pre-mRNA, mRNA, or proteins to modulate gene expression in cells, including ASOs, siRNA, miRNA, RNA aptamers, etc. The second category comprises RNA drugs that are translated into proteins, such as in vitro transcribed mRNA. The third category involves therapeutic genome editing, where RNA serves as a guide for effector proteins that modify cellular DNA sequences (Figure 1). This chapter summarized the current research progress and clinical applications of RNA therapy in kidney diseases.

Figure 1 The mechanism of RNA therapies. RNA-based drugs can target various stages of protein coding and gene expression. For instance, mRNA encoding Cas9 has the potential to edit the genome. ASO can modulate splicing, while mature mRNA can be targeted by ASO, siRNA, or miRNA. The function of proteins can be regulated through the binding of aptamers. Additionally, exogenous mRNA can be utilized to introduce specific proteins into cells, either to supplement deficient enzymes or to serve as antigens, cytokines, and other molecules that can trigger immune responses.

Oligonucleotide Therapy

Over the past few decades, oligonucleotide therapeutics (including ASOs, siRNA, miRNA, aptamers, and decoys) have emerged as increasingly important tools in nephrology and other fields.40–42 These promising drugs leverage specific mechanisms for therapeutic effects. For instance, siRNA promotes mRNA degradation by attaching to specific sequences within the coding area, thereby increasing its promise as a therapeutic tool. ASOs identify and attach to complementary sequences of DNA or RNA, supporting proper mRNA splicing, averting faulty protein production, or directing RNA toward degradation. Furthermore, aptamers are sequences of oligonucleotides that form specific three-dimensional shapes, enabling them to interact with various targets (such as proteins, cells, microorganisms, compounds, and additional nucleic acids). Their binding to proteins can inhibit protein-protein interactions, yielding therapeutic effects (Figure 1). A significant benefit of focusing on the kidney is that oligonucleotide therapies are rapidly removed from the bloodstream via renal filtration, improving their distribution within the kidney compared to other organs.43 This section presented the key strategies utilized in the creation of oligonucleotides-based therapies, as well as their recent progress in treating renal diseases, which are also detailed in Table 1.

Table 1 The Research and Application of RNA Drugs in Kidney Diseases

ASO Therapeutics

ASOs are single-stranded RNAs that hybridize complementarily to messenger RNA (mRNA) encoding proteins, thereby inhibiting their translation into protein. ASOs utilize various mechanisms of action; however, approved drugs can be classified into two broad categories based on their mechanisms (Figure 1).

The initial category of ASOs promotes the cleavage of target mRNA by attaching to a specific sequence. Generally, these ASOs are modified to contain a DNA-based central sequence that is bordered by chemically altered RNA. When a duplex is formed with the target RNA, the central segment creates a DNA-RNA hybrid that is detected by RNase H. This enzyme then cleaves the RNA sequence within the duplex, resulting in the degradation of the target RNA. As this drug class relies on RNase H, which is active in both the nucleus and cytoplasm, they can efficiently target non-coding elements,77 which is different from siRNA-based therapies that mainly operate within the cytoplasm. There are specific advantages in certain situations.78 By now, some ASO medications for kidney disease have shown encouraging outcomes in clinical trials. The 2.5th generation APOL1 ASO (IONIS-APOL1 Rx) was selected as a clinical candidate because it has demonstrated its reliable and strong efficacy in studies using genomic APOL1 transgenic mice. Subcutaneous administration of IONIS-APOL1 Rx into APOL1 G1 transgenic mice led to a reduction in APOL1 mRNA levels in the kidney and liver that was dependent on the dose, thereby effectively preventing interferon-induced proteinuria in a dose-dependent manner.79 This drug has been utilized in a first-in-human single ascending dose Phase I study (NCT04269031) aimed at evaluating the safety and pharmacokinetics of a single ascending dose of subcutaneous ASO (ION532, also referred to as AZD2373), although the results have not yet been published. Additionally, another clinical study is currently ongoing (NCT05351047). A Phase 2 trial evaluating the efficacy of an ASO targeting apolipoprotein(a) in patients with hyperlipoproteinemia(a) and cardiovascular diseases has shown promising results (NCT03070782). This suggests that it may also be beneficial for treating kidney disease where lipoprotein(a) levels are implicated. Furthermore, the overproduction of complement factor B (CFB) has been linked to the exacerbation of IgA nephropathy. In response, Ionis partnering with Roche has developed IONIS-FB-LRx, a ligand-conjugated ASO therapeutic designed to target FB aimed at reducing IgA production and alleviating the symptoms associated with IgA nephropathy (NCT04014335, NCT05797610). Additionally, a current study has been evaluating an ASO aimed at the renal sodium-glucose cotransporter 2 to reduce blood glucose levels in individuals diagnosed with type 2 diabetes (NCT00836225).

Several ASO drugs have demonstrated therapeutic potential in animal and cell models. For instance, Guha et al have utilized an ASO-gapmer, which specifically inhibits the expression of connective tissue growth factor (Ctgf) in a mouse model of diabetic nephropathy, thus alleviating the associated symptoms.47 Another study has employed an ASO-gapmer to silence Kras in a rat model of unilateral ureteral obstruction (UUO), significantly improving the pathological phenotype.48 Additionally, several ASOs have shown promising therapeutic effects in mouse models of polycystic kidney disease (PKD) by targeting mTORC1, mTORC2, angiotensinogen, and asparagine synthase (ASNS), thereby regulating their mRNA expressions.45,50,80 The potential of ASOs for targeted modulation of renal cell carcinoma (RCC) development and metastasis has also been explored in both in vitro and in vivo models. These ASOs can impede RCC progression by inhibiting the expression of tumor oncogenes such as vascular endothelial growth factor (VEGF), Ki67, and circEHD2.44,46,49,81,82 Collectively, these studies underscore the significant potential of novel ASO-based therapeutic strategies for the treatment of kidney diseases.

The second category of ASO therapies mainly influences the pre-mRNA splicing process via a steric hindrance mechanism. Numerous RNA-binding proteins engage with specific sequences in pre-mRNA, which in turn regulates additional splicing factors, leading to diverse alternative splicing patterns.83,84 Occasionally, these alternative splicing variations may play a role in disease onset. ASO drugs in this category can accurately target these sequences in pre-mRNA, thus impeding disease progression by modulating alternative splicing. This ability is distinctive to ASOs and offers new potential strategies for treating a range of genetic disorders. Multiple ASO therapies have been approved by the US Food and Drug Administration (FDA), such as nusinersen, eteplirsen, and golodirsen, aimed at modifying the splicing of target pre-mRNA. Regarding kidney diseases, studies have shown that exon skipping mediated by ASO can improve the ciliary structure and restore the placement of the CEP290 protein at the cilia’s base in individuals with Joubert syndrome harboring nonsense mutations in CEP290 exon 41. This strategy has also been confirmed through experiments using mouse models. The use of an ASO targeting technique can facilitate alternative splicing within intron 25 of the CEP290 gene in mice, which in turn aids in partially rescuing the complete CEP290 transcript and protein, ultimately enhancing renal ciliary length and alleviating cystic kidney disease symptoms.52 Furthermore, another type of ASO has been shown to inhibit EDA exon inclusion, thereby reducing TGFβ-induced profibrotic effects and enhancing cell survival.51 These findings indicate that ASO therapies hold significant potential for the treatment of alternative splicing-associated kidney diseases.

siRNA-Based Therapeutics

siRNAs are short double-stranded RNAs with typically 20–27 nucleotides in length, which specifically target and degrade mRNA in a sequence-dependent manner. siRNA technology has demonstrated considerable potential in the treatment of kidney diseases, not only helping to understand the molecular mechanisms underlying these conditions but also promoting various treatment strategies for kidney diseases.85 A previous study has shown that modulation of Mapk1 silencing can ameliorate glomerulosclerosis in a mouse model of glomerulonephritis,53 while the use of siRNA targeting Smad4 has been found effective in preventing renal fibrosis. These findings suggest that siRNA may serve as a pivotal therapeutic target for kidney disease.16

siRNA technology demonstrates significant therapeutic potential in certain kidney diseases. For example, the delivery of p38α MAPK and p65 siRNA to glomerular endothelial and mesangial cells in an IgA nephropathy mouse model via LNP carriers effectively alleviates proteinuria, inflammation, and excessive extracellular matrix deposition.54 In lupus nephritis, co-delivery of siRNA targeting dihydroartemisinin and HMGB1 produces a therapeutic effect by inhibiting the TLR4 signaling pathway.55 In cases of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis, siRNA targeting p65 reduces endothelial cell NF-κB activation and mitigates renal inflammation.56 In diabetic nephropathy, siRNA targeting NLRP3 and HDAC4 attenuates podocyte damage and renal lesion progression, respectively.57,58 Primary hyperoxaluria type 1 (PH1) is a hereditary kidney disease characterized by excessive oxalate production, which can lead to kidney stones, renal failure, and widespread toxicity. The siRNA drug (Oxlumo) developed by Alnylam was approved by the FDA in 2020. It reduces urinary oxalate excretion in PH1 patients by targeting the HAO1 gene.86,87 Clinical studies have indicated that after Oxlumo treatment for 6 months, most patients have achieved oxalate levels close to normal (NCT04152200).

Additionally, siRNA technology has demonstrated significant efficacy in AKI. In cases of ischemia- or nephrotoxicity-related kidney injury, siRNA targeting p53 has been shown to effectively protect proximal tubule cells and preserve kidney function.59 Furthermore, siRNA targeting meprin-1β and p53 has been effective in preventing kidney damage in a cisplatin-induced AKI model.60 However, siRNA targeting CD40 has been observed to reduce inflammation and promote kidney repair in a unilateral ureteral obstruction-induced AKI model.61 Glevova et al88 have assessed the potential of 53 various siRNA targets, mainly related to apoptosis, inflammation, and immune rejection pathways after transplant-related ischemia-reperfusion. Although this approach remains under development, it has achieved promising results in mouse models.89

Notably, siRNA drug delivery systems are also being continuously optimized, including polycationic cyclodextrin nanoparticles and E-selectin-targeted liposomes. These delivery systems can enhance the specificity and efficiency of siRNA in the kidney.90

miRNA-Based Therapeutics

miRNA consists of single-stranded ncRNA with approximately 22 nucleotides in length, which binds to the 3′ untranslated region (UTR) of target mRNA through partial complementarity, primarily mediating translation inhibition and mRNA decay. miRNA-based therapeutic approaches include the use of anti-miRNA oligonucleotides (AMOs), miRNA antagonists, miRNA sponges, miRNA mimics, and target site blockers (TSB) to modulate miRNA function.91,92

miRNAs play a crucial role in the regulation of post-transcriptional gene expression, with their abnormal expression closely associated with various diseases, particularly kidney diseases (such as AKI,93 kidney transplantation,94 polycystic kidney disease,95,96 and renal fibrosis.97,98 As novel biomarkers and therapeutic targets, miRNAs can be detected in plasma and urine exosomes, demonstrating their potential for disease diagnosis and monitoring.99,100 Moreover, as indicated by the results of animal models, the pathological processes of kidney disease can be ameliorated by regulating specific miRNAs, including anti-miR21, anti-miR192, miR204 mimics, miR211 mimics, and anti-miR107.15,62–64,68,101 Furthermore, miRNAs have also shown promise in the treatment of ischemic kidney injury.65–68

Although miRNA-based therapy holds significant promise, its clinical translation encounters several challenges, including the need for a comprehensive understanding of the miRNA regulatory network associated with the disease, cell or organ-specific regulation, and potential off-target effects. Currently, there are 4 miRNA-based therapies in clinical development, two of which specifically target kidney disease:93 the anti-miR21 drug RG012 [currently in phase 2 clinical trial for Alport nephropathy (NCT02855268)101 and RGLS4326 [an antagonist that inhibits miR17 in autosomal dominant polycystic kidney disease (ADPKD), currently in Phase 1 clinical trial (NCT04536688)102. These findings offer hope for future evaluation of the clinical utility of miRNA mimics and inhibitors aimed at key pathological renal pathways.

RNA Aptamer Therapy

Aptamers are special DNA or RNA oligonucleotides that bind to various molecular targets with high affinity and specificity, including proteins, inorganic molecules, and complex biosystems.103 They are selected through the SELEX method, involving multiple rounds of incubation and PCR amplification to identify the sequences with the strongest binding affinity and specificity.104 Aptamers have been widely used in biomedical and non-medical areas, such as pesticide residue detection.104 Their high target specificity offers a significant advantage in oligonucleotide-based therapies, addressing delivery problems and off-target effects.105,106

In renal diseases, aptamers have shown potential as diagnostic tools and targeted therapies. For example, aptamers selected by cell-SELEX specifically bind to pathophysiologically altered renal cells, demonstrating higher binding specificity compared to non-stimulated cells105 This study suggested aptamers may be valuable for renal disease treatment. Additionally, aptamers have been developed to target specific receptors and modulate intracellular pathways, and their effectiveness in treating renal injury have been demonstrated.69–71 Additionally, aptamers with high binding affinity and specificity to RCC cell lines have also been identified, which may be used in RCC identification and targeting72 Wang et al107 have employed cell-SELEX technology to develop the RCC nucleic acid aptamer probe W786-1 (Kd = (9.4 ± 2.0) nmol/L), with the RCC cell line 786-O as the target and 293T cells as the control. Following structural optimization, the short sequence W786-1S (48 nucleotides) was derived, retaining its ability to recognize 786-O cells and confirming that its target is a cell surface membrane protein. W786-1 exhibits strong binding affinity and selectivity at physiological temperatures, indicating its potential for RCC diagnosis and treatment. Additionally, Zhang et al72 have identified the nucleic acid aptamer SW-4 (Kd ≈ (45.92 ± 5.58) nmol/L) from the Swan Library, which exhibits high affinity and specificity for 786-O cells while showing no binding to HEK293T or HK-2 cells. The optimized SW-4b is internalized by target cells, and fluorescence imaging reveals its robust recognition capability, along with significant anti-proliferative activity against 786-O cells, effectively inhibiting the progression of the S phase of the cell cycle. SW-4b is a novel promising tool for RCC diagnosis and targeted therapy, and future studies involving tumor-bearing mice should be conducted to validate its tumor suppressive function.

However, limited studies have been reported on aptamers in renal diseases, and in vitro-SELEX-selected aptamers may have limitations when translated to in vivo organisms.108 The in vivo-SELEX approaches using animal models of renal diseases could lead to the development of aptamers with enhanced therapeutic or targeting potential.

mRNA‐based Therapeutics

With notable progress in mRNA-driven nanotechnology, groundbreaking therapies have been created for various illnesses, especially in the field of cancer treatment. mRNA-based therapies have shown significant potential in the realms of immunotherapy and vaccine development, and their potential applications are rapidly increasing. In this section, we outline the latest advancements in mRNA-based therapies that have been incorporated into preclinical and clinical studies aimed at combating kidney cancer. We provide an overview of the utilization of mRNA-encoding tumor antigens, cytokines, and tumor suppressors in the context of renal tumors.

Vaccines

Vaccines utilizing mRNA can be crafted according to the specific tumor antigens found in cancer cells, which subsequently elicit strong immune responses from both T cells and B cells against tumors in the body.108,109 Tumor antigens are primarily divided into two categories: tumor-associated autoantigens (TAAs) and tumor-specific antigens (TSAs.110 TAAs are generally found in high quantities in tumor cells, but they are also present in normal tissues, which leads to relatively limited tumor specificity and immunogenicity. Conversely, TSAs are neoantigens generated by mutations within tumor cells with greater tumor specificity and immunogenicity, although they tend to elicit lower tolerance in the organism.111 Since the groundbreaking introduction and assessment of the first mRNA cancer vaccine,112 extensive preclinical and clinical investigations have thoroughly demonstrated the potential of mRNA vaccines in the field of cancer therapy.

In tumor treatment, tumor-associated antigen (TAA) vaccines are among the most widely utilized. However, mammals typically exhibit high immune tolerance to individual TAAs. Therefore, the strategy of employing a combination of multiple TAAs is increasingly receiving attention, intending to enhance the therapeutic efficacy of cancer vaccines.73 For instance, a phase I/II trial73 involving 30 metastatic RCC patients has evaluated an mRNA-based vaccine formulation comprising a mixture of in vitro transcribed RNA encoding 6 distinct TAAs (including MUC1, CEA, Her2/neu, telomerase, survivin, and MAGE-A1). According to the survival analysis results, there is a significant correlation between patient survival and the immune response to the TAAs encoded by the naked mRNA vaccine, marking it as one of the pioneering vaccination studies in the realm of RCC.

Given the critical role of dendritic cells in inducing specific tumor antigen responses, they are an ideal and commonly utilized delivery vehicle for mRNA cancer vaccines. Dendritic cells primarily originate from the patient’s own body and can effectively generate an mRNA vaccine after transfection with the corresponding mRNA. For instance, during the development of rocapuldencel-T (AGS-003), a single-arm, open-label Phase II trial has investigated the combined use of AGS-003 and sunitinib in 21 patients with intermediate- or high-risk RCC. The primary endpoint of this trial is the complete response rate, while secondary endpoints include overall survival (OS), progression-free survival (PFS), and safety. As indicated by the results, approximately 62% of patients achieved clinical benefit.113

While previous clinical trials have demonstrated some efficacy in the domain of mRNA cancer vaccines, the overall results are not optimal. To further enhance the efficacy, Xu et al have identified 4 potential RCC-specific neoantigen candidate genes through a comprehensive analysis of the TCGA-kidney renal clear cell carcinoma (KIRC) dataset: DNA topoisomerase II alpha (TOP2A), neutrophil cytoplasmic factor 4 (NCF4), formin-like protein 1 (FMNL1), and docking protein 3 (DOK3). These genes exhibit upregulation and mutation characteristics in KIRC and are closely associated with patient survival and the activity of antigen-presenting cells. This study not only validated TOP2A, NCF4, FMNL1, and DOK3 as potentially effective targets for the development of KIRC mRNA vaccines, but also aimed to further classify the immune subtypes of KIRC and identify RIS2, which may derive greater benefit from mRNA vaccination within the cancer patient population.114

Cytokine

Cytokines are soluble proteins primarily released by leukocytes. They form a complex superfamily of regulatory proteins that can finely modulate the activity of other immune system target cells.115 Cytokines [such as interferons (IFNs) and interleukins (ILs)] are produced by eukaryotic cells in response to viral infections or various biological and synthetic inducers, exhibiting a wide range of biological and pharmacological properties.115,116 Notable examples of clinically significant cytokines include ILs, IFNs (α, β, γ), colony-stimulating factors (such as G-CSF and GM-CSF), and tumor necrosis factor.

In the 1980s, IFN-α and IL-2 were extensively studied in phase II clinical trials for their anti-tumor activity against RCC.74,117 In contrast, other cytokines (including IL-12, IL-6, IL-1, IFN-β, IFN-γ, and GM-CSF) did not demonstrate significant efficacy in subsequent phase II and Phase III clinical trials.74,118–136 As a result, 7 phase II studies evaluating high-dose IL-2 therapy were approved by the FDA.137 These studies continued to monitor patient toxicity and clinical outcomes and reported an overall response rate (ORR) of 15%, with 7% of patients achieving a complete response. Unfortunately, long-term disease control remains elusive for most patients.138 Therefore, further research is essential to investigate the application of cytokines in kidney diseases, so as to better understand their efficacy and potential.

Encode for Disease-Related Proteins

Tumor suppressor genes are essential biological regulators that inhibit tumor development by preventing abnormal cell proliferation. However, they are frequently lost or functionally inactive in various tumors, thereby promoting tumor formation and progression.139 Researchers have explored several strategies (including small molecule inhibitors, protein delivery technologies, and plasmid DNA-based gene transfection) to restore their function; however, these strategies still face significant limitations.139 Recently, the use of mRNA nanoparticles encoding tumor suppressors for systemic delivery has emerged as a promising avenue. This is particularly important in RCC, where tumor suppressor genes (such as P53 and HIF1α) play critical roles in maintaining cellular homeostasis and inhibiting tumor growth.140,141 Among them, the TP53 gene encodes the p53 protein (a major cancer preventive factor and has garnered particular attention regarding its function and regulatory mechanisms. Through self-assembly technology, TP53 mRNA can be efficiently encapsulated in nanoparticles, which significantly enhances stability and delivery efficiency, leading to high-level recovery of p53 protein and notable therapeutic effects in TP53 gene-deleted tumor models.139 This discovery not only provides new experimental evidence for understanding the tumor suppression mechanism but also suggests that this strategy may be applicable for RCC treatment, although further research and validation are necessary to confirm this potential.

mRNA-based cytokine therapies aim to modulate the levels of cytokines in a targeted manner. Compared with traditional cytokine therapies that aim to either enhance or suppress specific cytokines, mRNA therapy introduces mRNA-encoding cytokines to enhance their production in a controlled manner, providing a novel approach. For example, mRNA vaccines encoding cytokines (such as IL-12) are being explored to boost immune responses. However, the immunogenicity of mRNA therapy involving stimulation of innate immune responses may pose challenges in terms of unwanted side effects, including activation of the inflammatory pathways. Therefore, the interplay between mRNA-induced cytokine production and the immune response remains a key consideration for optimizing the therapeutic applications of mRNA-based cytokine therapies in treating kidney diseases (such as kidney cancer and autoimmune kidney diseases).

Additionally, mRNA-based therapies are also being explored for conditions such as cystinosis and methylmalonic acidemia. These therapies aim to introduce mRNA into cells to produce the necessary proteins that are otherwise deficient due to genetic mutations; mRNA-based therapies offer the potential for treating these diseases by directly correcting the molecular deficiencies.75,76

Therapeutic Genome Editing

CRISPR and CRISPR–Cas9 systems comprise the Cas9 nuclease and single-stranded guide RNA (sgRNA), which are widely utilized as gene editing tools due to their speed, accuracy, and efficiency.76,142 Compared with traditional therapies, CRISPR–Cas9 can circumvent the need for repeated administration and enhance therapeutic outcomes.143 The successful delivery of both two components (Cas9 and sgRNA) to target cells is crucial for effective therapy.144 The delivery methods of Cas9 include protein, DNA, or mRNA forms.144 Although direct delivery of Cas9 protein is relatively simple, it may induce adverse immune responses and encounter rapid degradation, resulting in a limited duration of gene editing efficacy.145 Additionally, the considerable mass (160 kDa) of the Cas9 protein poses difficulties for delivery systems, whether viral or non-viral.145 Common vectors for delivering Cas9 as DNA include plasmids and adeno-associated viruses (AAVs), which allow for prolonged expression of the protein but may result in increased off-target effects. AAVs exhibit low efficiency in editing tissues outside the liver and show high levels of hepatotoxicity, which increases the likelihood of off-target mutations occurring in non-cancerous cells.145 Recently, the use of in vitro transcribed mRNA has surfaced as an alternative transient method for delivering Cas9, which can significantly minimize off-target impacts and genotoxic concerns, thereby enhancing gene editing efficacy.146,147

The CRISPR/Cas9 system holds significant promise for the treatment of kidney diseases,40 as many such conditions (including autosomal dominant polycystic kidney disease and Alport syndrome) are caused by genetic mutations. Nevertheless, the application of gene editing in solid organs encounters challenges related to efficiently delivering the system to specific cells or tissues. Therefore, the use of this technology in kidney research is currently confined to the development of innovative in vitro models (utilizing human organoids) and in vivo models of kidney diseases. These models facilitate a deeper understanding of the molecular mechanisms underlying kidney disease and aid in the identification of novel disease progression-associated genes, as well as potential therapeutic targets.148,149

Another potential application of CRISPR/Cas9 technology is in the expansion of available sources for kidney transplants. Some researchers have proposed the possibility of cross-species organ transplantation, particularly using organs from pigs. However, this approach has resulted in severe immune responses in the human body, leading to rejection of donor organs. Nevertheless, CRISPR/Cas9 has emerged as a promising tool to address this challenge. Researchers have utilized CRISPR/Cas9 to genetically modify pig eggs, resulting in a lack of animal carbohydrate xenoantigens, which significantly reduces their recognition by human and non-human primate antibodies.150 Additionally, modifications have been made to induce MHC-I deletions in pigs.151 Higginbotham et al have demonstrated the feasibility of this method for the first time, achieving long-term transplantation success (over 125 days) from pigs to primates.152

Conclusions and Outlook

In recent years, the use of RNA-based approaches to treat kidney disease has gained significant importance, and several studies and drugs are currently in clinical development (Table 1). Although research has demonstrated that RNA-based therapies can effectively reach kidney cells and elicit therapeutic effects, targeted delivery within the body remains a significant challenge for the widespread application of these therapies. Currently, most ASO therapies utilize local delivery. However, when administered systemically, these drugs are frequently retained in various other organs, which may lead to off-target effects, side effects, and potentially renal toxicity. Additionally, the application of RNA technology still faces multiple challenges related to immunogenicity, stability, delivery efficiency, and intracellular effects.

These challenges, especially off-target effects and immunogenicity, remain significant barriers to the clinical use of RNA-based therapies. Although efforts are being made to improve RNA stability and reduce immune responses, addressing off-target effects is essential for the safe and effective use of these therapies. The use of RNA chemical modifications, advanced delivery systems, and kidney-specific ligands could enhance targeting precision and minimize unintended interactions with other organs, thereby reducing side effects. Further research into developing RNA delivery systems that minimize off-target effects and improve specificity is of great significance for the translation of RNA therapies from animal models to human patients.

When evaluating RNA therapies for specific renal diseases, various RNA-based approaches show promise depending on the disease’s characteristics. For example, ASOs can effectively target specific genetic mutations in inherited kidney diseases, such as polycystic kidney disease. siRNA therapies are promising for conditions like diabetic nephropathy and AKI by silencing harmful genes or pathways. Additionally, ncRNAs, such as miRNAs, hold potential for addressing fibrosis and inflammation-related kidney diseases. Ongoing clinical trials are essential to confirm the clinical viability of these approaches and identify optimal strategies for their use.

Recent advancements in RNA delivery systems, including improved lipid nanoparticles and other targeted vehicles, have shown promise in overcoming some of these challenges. Innovations in ncRNA therapies, particularly miRNAs and lncRNAs, could further improve the therapeutic outcomes of RNA-based treatments for kidney diseases. Clinical studies will help evaluate the practicality and clinical benefit of these innovations, guiding their integration into standard treatment protocols.

Thus, the potential applications of RNA-based therapies are vast. A deeper understanding of their specific roles in renal diseases will significantly contribute to advancing nephrology. Clinical evidence generated through these studies will be instrumental in establishing RNA therapies as a new treatment paradigm, with the potential to improve patient outcomes and reduce disease burden.

We have focused our discussion on RNA technologies and delivery strategies and have removed references to cytokine therapies to maintain the focus on RNA-based treatments for kidney diseases. Overall, this review emphasizes the critical role RNA-based therapies play in revolutionizing the treatment of kidney diseases, particularly with the continuous improvement of targeted delivery systems and specificity.

In summary, RNA technology not only accelerates the rapid development and production of mRNA vaccines but also introduces new research directions and treatment strategies in the field of nephrology. The continuous development of RNA-based therapies, alongside advances in delivery systems and targeting strategies that specifically address immunogenicity and off-target effects, may significantly enhance the treatment of kidney diseases. By assessing and tailoring RNA therapies to specific renal conditions, we can maximize their therapeutic potential and ultimately provide more effective and personalized treatments for kidney diseases. Integrating clinical trials with ongoing advancements in RNA technology will be key to fully realize the therapeutic potential of these treatments in clinical practice.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

This work was supported by the Key Project of Social Development of Jinhua Science and Technology Bureau of Zhejiang Province, China (No. 2021-3-079No. 2023-3-165No. 2021-3-154No. 2021-3-044).

Disclosure

The authors have no conflict of interest.

References

1. Arici M. Refugees with kidney disease: an increasing global challenge. Nat Rev Nephrol. 2021;17(6):366–367. doi:10.1038/s41581-020-00377-0

2. Francis A, Harhay MN, Ong ACM, et al. Chronic kidney disease and the global public health agenda: an international consensus. Nat Rev Nephrol. 2024;20(7):473–485. doi:10.1038/s41581-024-00820-6

3. Schreibing F, Kramann R. Mapping the human kidney using single-cell genomics. Nat Rev Nephrol. 2022;18(6):347–360. doi:10.1038/s41581-022-00553-4

4. Huang R, Fu P, Ma L. Kidney fibrosis: from mechanisms to therapeutic medicines. Signal Transduct Target Ther. 2023;8(1):129. doi:10.1038/s41392-023-01379-7

5. Kishi S, Kadoya H, Kashihara N. Treatment of chronic kidney disease in older populations. Nat Rev Nephrol. 2024;20(9):586–602. doi:10.1038/s41581-024-00854-w

6. Kariko K, Buckstein M, Ni H, et al. Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. Immunity. 2005;23(2):165–175. doi:10.1016/j.immuni.2005.06.008

7. Bennett CF. Therapeutic antisense oligonucleotides are coming of age. Annu Rev Med. 2019;70:307–321. doi:10.1146/annurev-med-041217-010829

8. Zhou JH, Rossi J. Aptamers as targeted therapeutics: current potential and challenges. Nat Rev Drug Discov. 2017;16(3):181–202. doi:10.1038/nrd.2016.199

9. Bondue T, van den Heuvel L, Levtchenko E, et al. The potential of RNA-based therapy for kidney diseases. Pediatr Nephrol. 2023;38(2):327–344. doi:10.1007/s00467-021-05352-w

10. Haddad G, Kölling M, Lorenzen JM. The hypoxic kidney: pathogenesis and noncoding RNA-based therapeutic strategies. Swiss Med Wkly. 2019;149:w14703. doi:10.4414/smw.2019.14703

11. Frankish A, Diekhans M, Ferreira AM, et al. GENCODE reference annotation for the human and mouse genomes. Nucleic Acids Res. 2019;47(D1):D766–D73. doi:10.1093/nar/gky955

12. Esteller M. Non-coding RNAs in human disease. Nat Rev Genet. 2011;12(12):861–874. doi:10.1038/nrg3074

13. Beermann J, Piccoli MT, Viereck J, et al. Non-coding RNAs in development and disease: background, mechanisms, and therapeutic approaches. Physiol Rev. 2016;96(4):1297–1325. doi:10.1152/physrev.00041.2015

14. Song J, Kim YK. Targeting non-coding RNAs for the treatment of retinal diseases. Mol Ther Nucleic Acids. 2021;24:284–293. doi:10.1016/j.omtn.2021.02.031

15. Chau BN, Xin C, Hartner J, et al. MicroRNA-21 promotes fibrosis of the kidney by silencing metabolic pathways. Sci Transl Med. 2012;4(121):121ra18. doi:10.1126/scitranslmed.3003205

16. Morishita Y, Yoshizawa H, Watanabe M, et al. siRNAs targeted to Smad4 prevent renal fibrosis in vivo. Sci Rep. 2014;4:6424. doi:10.1038/srep06424

17. Fauman EB, Rai BK, Huang ES. Structure-based druggability assessment--identifying suitable targets for small molecule therapeutics. Curr Opin Chem Biol. 2011;15(4):463–468. doi:10.1016/j.cbpa.2011.05.020

18. Finan C, Gaulton A, Kruger FA, et al. The druggable genome and support for target identification and validation in drug development. Sci Transl Med. 2017;9(383). doi:10.1126/scitranslmed.aag1166

19. Beck JD, Reidenbach D, Salomon N, et al. mRNA therapeutics in cancer immunotherapy. mol Cancer. 2021;20(1):69. doi:10.1186/s12943-021-01348-0

20. Tian Z, Liang G, Cui K, et al. Insight into the prospects for RNAi therapy of cancer. Front Pharmacol. 2021;12:644718. doi:10.3389/fphar.2021.644718

21. Le Huy B, Bui Thi Phuong H, Luong Xuan H. Advantages and disadvantages of RNA therapeutics. Prog mol Biol Transl Sci. 2024;203:151–164.

22. Jogalekar MP, Veerabathini A, Gangadaran P. Novel 2019 coronavirus: genome structure, clinical trials, and outstanding questions. Exp Biol Med. 2020;245(11):964–969. doi:10.1177/1535370220920540

23. Wang C, Horby PW, Hayden FG, et al. A novel coronavirus outbreak of global health concern. Lancet. 2020;395(10223):470–473. doi:10.1016/S0140-6736(20)30185-9

24. Jackson NAC, Kester KE, Casimiro D, et al. The promise of mRNA vaccines: a biotech and industrial perspective. NPJ Vaccines. 2020;5:11. doi:10.1038/s41541-020-0159-8

25. Roberts TC, Langer R, Wood MJA. Advances in oligonucleotide drug delivery. Nat Rev Drug Discov. 2020;19(10):673–694. doi:10.1038/s41573-020-0075-7

26. Thi TTH, Suys EJA, Lee JS, et al. Lipid-based nanoparticles in the clinic and clinical trials: from cancer nanomedicine to COVID-19 vaccines. Vaccines. 2021;9(4):359. doi:10.3390/vaccines9040359

27. Deering RP, Kommareddy S, Ulmer JB, et al. Nucleic acid vaccines: prospects for non-viral delivery of mRNA vaccines. Expert Opin Drug Deliv. 2014;11(6):885–899. doi:10.1517/17425247.2014.901308

28. Ulmer JB, Geall AJ. Recent innovations in mRNA vaccines. Curr Opin Immunol. 2016;41:18–22. doi:10.1016/j.coi.2016.05.008

29. Ji JL, Shi HM, Li ZL, et al. Satellite cell-derived exosome-mediated delivery of microRNA-23a/27a/26a cluster ameliorates the renal tubulointerstitial fibrosis in mouse diabetic nephropathy. Acta Pharmacol Sin. 2023;44(12):2455–2468. doi:10.1038/s41401-023-01140-4

30. Sone H, Lee TJ, Lee BR, et al. MicroRNA-mediated attenuation of branched-chain amino acid catabolism promotes ferroptosis in chronic kidney disease. Nat Commun. 2023;14(1):7814. doi:10.1038/s41467-023-43529-z

31. Xing X, Rodeo SA. Emerging roles of non-coding RNAs in fibroblast to myofibroblast transition and fibrotic diseases. Front Pharmacol. 2024;15:1423045. doi:10.3389/fphar.2024.1423045

32. Lin S, Ren Z, Li L, et al. LncRNA AP001007 protects human renal tubular epithelial HK-2 cells and kidney organoids from LPS-induced injury. Sci Rep. 2024;14(1):28578. doi:10.1038/s41598-024-79367-2

33. Xie Y, Zhang G, Pan J, et al. The LncRNA6524/miR-92a-2-5p/Dvl1/Wnt/β-catenin axis promotes renal fibrosis in the UUO mouse model. Arch Biochem Biophys. 2024;761:110175. doi:10.1016/j.abb.2024.110175

34. Li X, Ma TK, Wang M, et al. YY1-induced upregulation of LncRNA-ARAP1-AS2 and ARAP1 promotes diabetic kidney fibrosis via aberrant glycolysis associated with EGFR/PKM2/HIF-1α pathway. Front Pharmacol. 2023;14:1069348. doi:10.3389/fphar.2023.1069348

35. Yang N, Yan N, Bai Z, et al. FTO attenuates LPS-induced acute kidney injury by inhibiting autophagy via regulating SNHG14/miR-373-3p/ATG7 axis. Int Immunopharmacol. 2024;128:111483. doi:10.1016/j.intimp.2023.111483

36. Luo M, Liu Z, Hu Z, et al. Quercetin improves contrast-induced acute kidney injury through the HIF-1α/lncRNA NEAT1/HMGB1 pathway. Pharm Biol. 2022;60(1):889–898. doi:10.1080/13880209.2022.2058558

37. Xie KH, Liu XH, Jia J, et al. Hederagenin ameliorates cisplatin-induced acute kidney injury via inhibiting long non-coding RNA A330074k22Rik/Axin2/β-catenin signalling pathway. Int Immunopharmacol. 2022;112:109247. doi:10.1016/j.intimp.2022.109247

38. Kaczmarek JC, Kowalski PS, Anderson DG. Advances in the delivery of RNA therapeutics: from concept to clinical reality. Genome Med. 2017;9(1):60. doi:10.1186/s13073-017-0450-0

39. Deweerdt S. The natural history of an epidemic. Nature. 2019;573(7773):S10–S2. doi:10.1038/d41586-019-02686-2

40. Carton-Garcia F, Saande CJ, Meraviglia-Crivelli D, et al. Oligonucleotide-Based Therapies for Renal Diseases. Biomedicines. 2021;9(3):303. doi:10.3390/biomedicines9030303

41. Dhuri K, Bechtold C, Quijano E, et al. Antisense oligonucleotides: an emerging area in drug discovery and development. J Clin Med. 2020;9(6):2004. doi:10.3390/jcm9062004

42. Li H, Wang C, Che R, et al. A potential therapy using antisense oligonucleotides to treat autosomal recessive polycystic kidney disease. J Clin Med. 2023;12(4):1428.

43. Geary RS, Norris D, Yu R, et al. Pharmacokinetics, biodistribution and cell uptake of antisense oligonucleotides. Adv Drug Deliv Rev. 2015;87:46–51. doi:10.1016/j.addr.2015.01.008

44. Tan Y, Zheng T, Su Z, et al. Alternative polyadenylation reprogramming of MORC2 induced by NUDT21 loss promotes KIRC carcinogenesis. JCI Insight. 2023;8(18). doi:10.1172/jci.insight.162893

45. Clerici S, Podrini C, Stefanoni D, et al. Inhibition of asparagine synthetase effectively retards polycystic kidney disease progression. EMBO Mol Med. 2024;16(6):1379–1403. doi:10.1038/s44321-024-00071-9

46. He T, Zhang Q, Xu P, et al. Extracellular vesicle-circEHD2 promotes the progression of renal cell carcinoma by activating cancer-associated fibroblasts. mol Cancer. 2023;22(1):117. doi:10.1186/s12943-023-01824-9

47. Guha M, Xu ZG, Tung D, et al. Specific down-regulation of connective tissue growth factor attenuates progression of nephropathy in mouse models of type 1 and type 2 diabetes. FASEB J. 2007;21(12):3355–3368. doi:10.1096/fj.06-6713com

48. Wang JH, Newbury LJ, Knisely AS, et al. Antisense knockdown of Kras inhibits fibrosis in a rat model of unilateral ureteric obstruction. Am J Pathol. 2012;180(1):82–90. doi:10.1016/j.ajpath.2011.09.036

49. Shi W, Siemann DW. Inhibition of renal cell carcinoma angiogenesis and growth by antisense oligonucleotides targeting vascular endothelial growth factor. Br J Cancer. 2002;87(1):119–126. doi:10.1038/sj.bjc.6600416

50. Ravichandran K, Zafar I, He Z, et al. An mTOR anti-sense oligonucleotide decreases polycystic kidney disease in mice with a targeted mutation in Pkd2. Hum Mol Genet. 2014;23(18):4919–4931. doi:10.1093/hmg/ddu208

51. Phanish MK, Heidebrecht F, Jackson M, et al. Targeting alternative splicing of fibronectin in human renal proximal tubule epithelial cells with antisense oligonucleotides to reduce EDA+ fibronectin production and block an autocrine loop that drives renal fibrosis. Exp Cell Res. 2024;442(1):114186. doi:10.1016/j.yexcr.2024.114186

52. Ramsbottom SA, Molinari E, Srivastava S, et al. Targeted exon skipping of a CEP290 mutation rescues Joubert syndrome phenotypes in vitro and in a murine model. Proc Natl Acad Sci U S A. 2018;115(49):12489–12494. doi:10.1073/pnas.1809432115

53. Shimizu H, Hori Y, Kaname S, et al. siRNA-based therapy ameliorates glomerulonephritis. J Am Soc Nephrol. 2010;21(4):622–633. doi:10.1681/ASN.2009030295

54. Wang YF, Wu QS, Wang JD, et al. Co-delivery of p38α MAPK and p65 siRNA by novel liposomal glomerulus-targeting nano carriers for effective immunoglobulin a nephropathy treatment. J Control Release. 2020;320:457–468. doi:10.1016/j.jconrel.2020.01.024

55. Diao L, Li M, Tao J, et al. Therapeutic effects of cationic liposomes on lupus-prone MRL/lpr mice are mediated inhibition of TLR4-triggered B-cell activation. Nanomed-Nanotechnol. 2022;40:102491. doi:10.1016/j.nano.2021.102491

56. Choi M, Schreiber A, Eulenberg-Gustavus C, et al. Endothelial NF-κB Blockade Abrogates ANCA-Induced GN. J Am Society Nephrol. 2017;28(11):3190–3203. doi:10.1681/ASN.2016060690

57. Wu M, Yang ZF, Zhang CY, et al. Inhibition of NLRP3 inflammasome ameliorates podocyte damage by suppressing lipid accumulation in diabetic nephropathy. Metabolism. 2021;118.

58. Raval N, Jogi H, Gondaliya P, et al. Cyclo-RGD truncated polymeric nanoconstruct with dendrimeric templates for targeted HDAC4 gene silencing in a diabetic nephropathy mouse model. Mol Pharmaceut. 2021;18(2):641–666. doi:10.1021/acs.molpharmaceut.0c00094

59. Molitoris BA, Dagher PC, Sandoval RM, et al. siRNA targeted to p53 attenuates ischemic and cisplatin-induced acute kidney injury. J Am Soc Nephrol. 2009;20(8):1754–1764. doi:10.1681/ASN.2008111204

60. Alidori S, Akhavein N, Thorek DL, et al. Targeted fibrillar nanocarbon RNAi treatment of acute kidney injury. Sci Transl Med. 2016;8(331):331ra39. doi:10.1126/scitranslmed.aac9647

61. Narvaez A, Guiteras R, Sola A, et al. siRNA-silencing of CD40 attenuates unilateral ureteral obstruction-induced kidney injury in mice. PLoS One. 2019;14(4):e0215232. doi:10.1371/journal.pone.0215232

62. Putta S, Lanting L, Sun G, et al. Inhibiting microRNA-192 ameliorates renal fibrosis in diabetic nephropathy. J Am Soc Nephrol. 2012;23(3):458–469. doi:10.1681/ASN.2011050485

63. Li XY, Zhang K, Jiang ZY, et al. MiR-204/miR-211 downregulation contributes to candidemia-induced kidney injuries via derepression of Hmx1 expression. Life Sci. 2014;102(2):139–144. doi:10.1016/j.lfs.2014.03.010

64. Wang S, Zhang Z, Wang J, et al. MiR-107 induces TNF-alpha secretion in endothelial cells causing tubular cell injury in patients with septic acute kidney injury. Biochem Biophys Res Commun. 2017;483(1):45–51. doi:10.1016/j.bbrc.2017.01.013

65. Wei Q, Liu Y, Liu P, et al. MicroRNA-489 induction by hypoxia-inducible factor-1 protects against ischemic kidney Injury. J Am Soc Nephrol. 2016;27(9):2784–2796. doi:10.1681/ASN.2015080870

66. Wei Q, Sun H, Song S, et al. MicroRNA-668 represses MTP18 to preserve mitochondrial dynamics in ischemic acute kidney injury. J Clin Invest. 2018;128(12):5448–5464. doi:10.1172/JCI121859

67. Zhu G, Pei L, Lin F, et al. Exosomes from human-bone-marrow-derived mesenchymal stem cells protect against renal ischemia/reperfusion injury via transferring miR-199a-3p. J Cell Physiol. 2019;234(12):23736–23749. doi:10.1002/jcp.28941

68. Wilflingseder J, Jelencsics K, Bergmeister H, et al. miR-182-5p inhibition ameliorates ischemic acute kidney injury. Am J Pathol. 2017;187(1):70–79. doi:10.1016/j.ajpath.2016.09.011

69. Ranches G, Lukasser M, Schramek H, et al. In vitro selection of cell-internalizing DNA aptamers in a model system of inflammatory kidney disease. Mol Ther Nucleic Acids. 2017;8:198–210. doi:10.1016/j.omtn.2017.06.018

70. Matsui T, Higashimoto Y, Nishino Y, et al. RAGE-aptamer blocks the development and progression of experimental diabetic nephropathy. Diabetes. 2017;66(6):1683–1695. doi:10.2337/db16-1281

71. Um JE, Park JT, Nam BY, et al. Periostin-binding DNA aptamer treatment attenuates renal fibrosis under diabetic conditions. Sci Rep. 2017;7(1):8490. doi:10.1038/s41598-017-09238-6

72. Taguchi K, Yamagishi SI, Yokoro M, et al. RAGE-aptamer attenuates deoxycorticosterone acetate/salt-induced renal injury in mice. Sci Rep. 2018;8(1):2686. doi:10.1038/s41598-018-21176-5

73. He Q, Gao H, Tan D, et al. mRNA cancer vaccines: advances, trends and challenges. Acta Pharm Sin B. 2022;12(7):2969–2989. doi:10.1016/j.apsb.2022.03.011

74. Wirth MP. Immunotherapy for metastatic renal cell carcinoma. Urol Clin North Am. 1993;20(2):283–295. doi:10.1016/S0094-0143(21)00487-0

75. Bondue T, Berlingerio SP, Siegerist F, et al. CTNS mRNA as a potential treatment for nephropathic cystinosis. bioRxiv. 2023;2023–2024.

76. An D, Frassetto A, Jacquinet E, et al. Long-term efficacy and safety of mRNA therapy in two murine models of methylmalonic acidemia. EBioMedicine. 2019;45:519–528. doi:10.1016/j.ebiom.2019.07.003

77. Liang XH, Sun H, Nichols JG, et al. RNase H1-dependent antisense oligonucleotides are robustly active in directing RNA cleavage in both the cytoplasm and the nucleus. Mol Ther. 2017;25(9):2075–2092. doi:10.1016/j.ymthe.2017.06.002

78. Carthew RW, Sontheimer EJ. Origins and Mechanisms of miRNAs and siRNAs. Cell. 2009;136(4):642–655. doi:10.1016/j.cell.2009.01.035

79. Aghajan M, Booten SL, Althage M, et al. Antisense oligonucleotide treatment ameliorates IFN-gamma-induced proteinuria in APOL1-transgenic mice. JCI Insight. 2019;4(12). doi:10.1172/jci.insight.126124

80. Ravichandran K, Ozkok A, Wang Q, et al. Antisense-mediated angiotensinogen inhibition slows polycystic kidney disease in mice with a targeted mutation in Pkd2. Am J Physiol Renal Physiol. 2015;308(4):F349–57. doi:10.1152/ajprenal.00478.2014

81. Cenni E, Perut F, Zuntini M, et al. Inhibition of angiogenic activity of renal carcinoma by an antisense oligonucleotide targeting fibroblast growth factor-2. Anticancer Res. 2005;25(2A):1109–1113.

82. Zellweger T, Miyake H, July LV, et al. Chemosensitization of human renal cell cancer using antisense oligonucleotides targeting the antiapoptotic gene clusterin. Neoplasia. 2001;3(4):360–367. doi:10.1038/sj.neo.7900174

83. Chen M, Manley JL. Mechanisms of alternative splicing regulation: insights from molecular and genomics approaches. Nat Rev mol Cell Biol. 2009;10(11):741–754. doi:10.1038/nrm2777

84. Lee Y, Rio DC. Mechanisms and Regulation of Alternative Pre-mRNA Splicing. Annu Rev Biochem. 2015;84:291–323. doi:10.1146/annurev-biochem-060614-034316

85. Yang C, Zhang C, Zhao Z, et al. Fighting against kidney diseases with small interfering RNA: opportunities and challenges. J Transl Med. 2015;13(1):39. doi:10.1186/s12967-015-0387-2

86. Scott LJ, Keam SJ. Lumasiran: first Approval. Drugs. 2021;81(2):277–282. doi:10.1007/s40265-020-01463-0

87. Garrelfs SF, Frishberg Y, Hulton SA, et al. Lumasiran, an RNAi therapeutic for primary hyperoxaluria type 1. N Engl J Med. 2021;384(13):1216–1226. doi:10.1056/NEJMoa2021712

88. Glebova K, Reznik ON, Reznik AO, et al. siRNA technology in kidney transplantation: current status and future potential. BioDrugs. 2014;28(4):345–361. doi:10.1007/s40259-014-0087-0

89. Zheng X, Zang G, Jiang J, et al. Attenuating ischemia-reperfusion injury in kidney transplantation by perfusing donor organs with siRNA cocktail solution. Transplantation. 2016;100(4):743–752. doi:10.1097/TP.0000000000000960

90. Zuckerman JE, Gale A, Wu PW, et al. siRNA delivery to the glomerular mesangium using polycationic cyclodextrin nanoparticles containing siRNA. Nucleic Acid Ther. 2015;25(2):53–64. doi:10.1089/nat.2014.0505

91. Schena FP, Serino G, Sallustio F. MicroRNAs in kidney diseases: new promising biomarkers for diagnosis and monitoring. Nephrol Dial Transplant. 2014;29(4):755–763. doi:10.1093/ndt/gft223

92. Dammes N, Peer D. Paving the road for RNA therapeutics. Trends Pharmacol Sci. 2020;41(10):755–775. doi:10.1016/j.tips.2020.08.004

93. Brandenburger T, Lorenzen JM. Diagnostic and therapeutic potential of microRNAs in acute kidney injury. Front Pharmacol. 2020;11:657. doi:10.3389/fphar.2020.00657

94. Ledeganck KJ, Gielis EM, Abramowicz D, et al. MicroRNAs in AKI and kidney transplantation. Clin J Am Soc Nephrol. 2019;14(3):454–468. doi:10.2215/CJN.08020718

95. Ramalingam H, Yheskel M, Patel V. Modulation of polycystic kidney disease by non-coding RNAs. Cell Signal. 2020;71:109548. doi:10.1016/j.cellsig.2020.109548

96. Li D, Sun L. MicroRNAs and polycystic kidney disease. Kidney Med. 2020;2(6):762–770. doi:10.1016/j.xkme.2020.06.013

97. Fan Y, Chen H, Huang Z, et al. Emerging role of miRNAs in renal fibrosis. RNA Biol. 2020;17(1):1–12. doi:10.1080/15476286.2019.1667215

98. Lv W, Fan F, Wang Y, et al. Therapeutic potential of microRNAs for the treatment of renal fibrosis and CKD. Physiol Genomics. 2018;50(1):20–34. doi:10.1152/physiolgenomics.00039.2017

99. Peters LJF, Floege J, Biessen EAL, et al. MicroRNAs in chronic kidney disease: four candidates for clinical application. Int J mol Sci. 2020;21(18):6547. doi:10.3390/ijms21186547

100. Sun IO, Lerman LO. Urinary microRNA in kidney disease: utility and roles. Am J Physiol Renal Physiol. 2019;316(5):F785–F93. doi:10.1152/ajprenal.00368.2018

101. Gomez IG, MacKenna DA, Johnson BG, et al. Anti-microRNA-21 oligonucleotides prevent Alport nephropathy progression by stimulating metabolic pathways. J Clin Invest. 2015;125(1):141–156. doi:10.1172/JCI75852

102. Gale DP, Gross O, Wang F, et al. A randomized controlled clinical trial testing effects of lademirsen on kidney function decline in adults with Alport syndrome. Clin J Am Soc Nephrol. 2024;19(8):995–1004. doi:10.2215/CJN.0000000000000458

103. Lee EC, Valencia T, Allerson C, et al. Discovery and preclinical evaluation of anti-miR-17 oligonucleotide RGLS4326 for the treatment of polycystic kidney disease. Nat Commun. 2019;10(1):4148. doi:10.1038/s41467-019-11918-y

104. Ellington AD, Szostak JW. In vitro selection of RNA molecules that bind specific ligands. Nature. 1990;346(6287):818–822. doi:10.1038/346818a0

105. Lakhin AV, Tarantul VZ, Gening LV. Aptamers: problems, solutions and prospects. Acta Naturae. 2013;5(4):34–43. doi:10.32607/20758251-2013-5-4-34-43

106. He J, Peng T, Peng Y, et al. Molecularly engineering triptolide with aptamers for high specificity and cytotoxicity for triple-negative breast cancer. J Am Chem Soc. 2020;142(6):2699–2703. doi:10.1021/jacs.9b10510

107. Zhang H, Wang Z, Xie L, et al. Molecular recognition and in-vitro-targeted inhibition of renal cell carcinoma using a DNA aptamer. Mol Ther Nucleic Acids. 2018;12:758–768. doi:10.1016/j.omtn.2018.07.015

108. Sola M, Menon AP, Moreno B, et al. Aptamers against live targets: is in vivo SELEX finally coming to the edge? Mol Ther Nucleic Acids. 2020;21:192–204. doi:10.1016/j.omtn.2020.05.025

109. Pastor F, Berraondo P, Etxeberria I, et al. An RNA toolbox for cancer immunotherapy. Nat Rev Drug Discov. 2018;17(10):751–767. doi:10.1038/nrd.2018.132

110. Lang F, Schrors B, Lower M, et al. Author correction: identification of neoantigens for individualized therapeutic cancer vaccines. Nat Rev Drug Discov. 2024;23(2):156. doi:10.1038/s41573-023-00873-5

111. Mitchell DA, Batich KA, Gunn MD. Tetanus toxoid and CCL3 improve dendritic cell vaccines in mice and glioblastoma patients. Nature. 2015;519(7543):366–369. doi:10.1038/nature14320

112. Blass E, Ott PA. Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat Rev Clin Oncol. 2021;18(4):215–229. doi:10.1038/s41571-020-00460-2

113. Rittig SM, Haentschel M, Weimer J, et al. Long-term survival correlates with immunological responses in renal cell carcinoma patients treated with mRNA-based immunotherapy. Oncoimmunology. 2016;5(5):e1108511. doi:10.1080/2162402X.2015.1108511

114. Barata PC, Rini BI. Treatment of renal cell carcinoma: current status and future directions. CA Cancer J Clin. 2017;67(6):507–524. doi:10.3322/caac.21411

115. Leonard W. Type 1 cytokines and interferons and their receptors. Fundament Immunol. 2003.

116. Rosenberg S. Principles of cancer management: biologic therapy. Cancer. 1997;349.

117. Motzer RJ, Bander NH, Nanus DM. Renal-cell carcinoma. N Engl J Med. 1996;335(12):865–875. doi:10.1056/NEJM199609193351207

118. Kuebler J, Brown T, Goodman P. Continuous infusion recombinant gamma interferon (Clr-GIFN) for metastatic renal cell carcinoma. proceedings of the Proc Am Soc Clin Oncol; 1989.

119. Ellerhorst A, Kilbourn RG, Amato RJ, et al. Phase II trial of low dose γ -interferon in metastatic renal cell carcinoma. J Urol. 1994;152(3):841–845. doi:10.1016/S0022-5347(17)32587-9

120. Bruntsch U, De Mulder P, ten Bokkel Huinink W, et al. Phase II study of recombinant human interferon-γ in metastatic renal cell carcinoma. J Immunother. 1990;9(3):335–338.

121. Foon K, Doroshow J, Bonnem E, et al. A prospective randomized trial of α2b-interferon/γ-interferon or the combination in advanced metastatic renal cell carcinoma. J Immunother. 1988;7(6):540–545.

122. Aulitzky W, Gastl G, Aulitzky W, et al. Successful treatment of metastatic renal cell carcinoma with a biologically active dose of recombinant interferon-gamma. J Clin Oncol. 1989;7(12):1875–1884. doi:10.1200/JCO.1989.7.12.1875

123. Grups J, Frohmueller H. Cyclic interferon gamma treatment of patients with metastatic renal carcinoma. Br J Urol. 1989;64(3):218–220. doi:10.1111/j.1464-410X.1989.tb06000.x

124. Rinehart JJ, Young D, Laforge J, et al. Phase I/II trial of interferon-β-serine in patients with renal cell carcinoma; immunological and biological effects. Cancer Res. 1987;47(9):2481–2485.

125. Kish J, Ensley J, Alsarraf M, et al. Activity of serine inhibited recombinant-DNA interferon-beta (ifn-beta) in patients with metastatic and recurrent renal-cell cancer. Proceedings of the proceedings of the american association for cancer research; 1986. Amer assoc cancer research public ledger bldg, suite. 816. 150.

126. Nelson K, Wallenberg J, Todd M. High-dose intravenous therapy with beta-interferon in patients with renal cell cancer. proceedings of the Proc Am Assoc Cancer Res; 1989.

127. Kinney P, Triozzi P, Young D, et al. Phase II trial of interferon-beta-serine in metastatic renal cell carcinoma. J Clin Oncol. 1990;8(5):881–885. doi:10.1200/JCO.1990.8.5.881

128. Quesada JR, Kurzrock R, Sherwin SA, et al. Phase II studies of recombinant human interferon gamma in metastatic renal cell carcinoma. J Immunother. 1987;6(1):20–27.

129. Koiso K. Phase II study of recombinant human interferon gamma (S-6810) on renal cell carcinoma. Cancer. 1987;60:929–933.

130. Machida T, Koiso K, Takaku F, et al. Phase II study of recombinant human interferon gamma (S-6810) in renal cell carcinoma. Urological cooperative study group of recombinant human interferon gamma (S-6810). Gan to Kagaku Ryoho Cancer Chemother. 1987;14(2):440–445.

131. Rinehart JJ, Young D, Laforge J, et al. Phase I/II trial of recombinant gamma-interferon in patients with renal cell carcinoma: immunologic and biologic effects. J Immunother. 1987;6(3):302–312.

132. Garnick MB, Reich SD, Maxwell B, et al. Phase I/II study of recombinant interferon gamma in advanced renal cell carcinoma. J Urol. 1988;139(2):251–255. doi:10.1016/S0022-5347(17)42379-2

133. Rini BI, Stadler WM, Spielberger RT, et al. Granulocyte‐macrophage‐colony stimulating factor in metastatic renal cell carcinoma: a phase II trial. Cancer. 1998;82(7):1352–1358. doi:10.1002/(SICI)1097-0142(19980401)82:7<1352::AID-CNCR19>3.0.CO;2-5

134. Motzer RJ, Rakhit A, Schwartz LH, et al. Phase I trial of subcutaneous recombinant human interleukin-12 in patients with advanced renal cell carcinoma. Clin Cancer Res. 1998;4(5):1183–1191.

135. Stouthard J, Goey H, De Vries E, et al. Recombinant human interleukin 6 in metastatic renal cell cancer: a phase II trial. Br J Cancer. 1996;73(6):789–793. doi:10.1038/bjc.1996.137

136. Redman BG, Abubakr Y, Chou T-H, et al. Phase II trial of recombinant interleukin-1β in patients with metastatic renal cell carcinoma. J Immunother. 1994;16(3):211–215. doi:10.1097/00002371-199410000-00005

137. Fyfe G, Fisher RI, Rosenberg SA, et al. Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. J Clin Oncol. 1995;13(3):688–696. doi:10.1200/JCO.1995.13.3.688

138. Fisher RI, Rosenberg SA, Fyfe G. Long-term survival update for high-dose recombinant interleukin-2 in patients with renal cell carcinoma. Cancer J Sci Am. 2000;6:S55–7.

139. Sherr CJ. Principles of tumor suppression. Cell. 2004;116(2):235–246. doi:10.1016/S0092-8674(03)01075-4

140. Kong N, Tao W, Ling X, et al. Synthetic mRNA nanoparticle-mediated restoration of p53 tumor suppressor sensitizes p53-deficient cancers to mTOR inhibition. Sci Transl Med. 2019;11(523). doi:10.1126/scitranslmed.aaw1565

141. Shen C, Beroukhim R, Schumacher SE, et al. Genetic and functional studies implicate HIF1alpha as a 14q kidney cancer suppressor gene. Cancer Discov. 2011;1(3):222–235. doi:10.1158/2159-8290.CD-11-0098

142. Pickar-Oliver A, Gersbach CA. The next generation of CRISPR-Cas technologies and applications. Nat Rev mol Cell Biol. 2019;20(8):490–507. doi:10.1038/s41580-019-0131-5

143. Mali P, Yang L, Esvelt KM, et al. RNA-guided human genome engineering via Cas9. Science. 2013;339(6121):823–826. doi:10.1126/science.1232033

144. Jinek M, Chylinski K, Fonfara I, et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337(6096):816–821. doi:10.1126/science.1225829

145. Chew WL. Immunity to CRISPR Cas9 and Cas12a therapeutics. Wiley Interdiscip Rev Syst Biol Med. 2018;10(1). doi:10.1002/wsbm.1408

146. Wang X, Li M, Lee CM, et al. CRISPR/Cas9-based genome editing for disease modeling and therapy: challenges and opportunities for nonviral delivery. Chem Rev. 2017;117(15):9874–9906. doi:10.1021/acs.chemrev.6b00799

147. Zhang D, Wang GX, Yu XL, et al. Enhancing CRISPR/Cas gene editing through modulating cellular mechanical properties for cancer therapy. Nat Nanotechnol. 2022;17(7):777–+. doi:10.1038/s41565-022-01122-3

148. Liu C, Shi Q, Huang X, et al. mRNA-based cancer therapeutics. Nat Rev Cancer. 2023;23(8):526–543. doi:10.1038/s41568-023-00586-2

149. Cruz NM, Freedman BS. CRISPR gene editing in the kidney. Am J Kidney Dis. 2018;71(6):874–883. doi:10.1053/j.ajkd.2018.02.347

150. Higashijima Y, Hirano S, Nangaku M, et al. Applications of the CRISPR-Cas9 system in kidney research. Kidney Int. 2017;92(2):324–335. doi:10.1016/j.kint.2017.01.037

151. Estrada JL, Martens G, Li P, et al. Evaluation of human and non-human primate antibody binding to pig cells lacking GGTA1/CMAH/beta4GalNT2 genes. Xenotransplantation. 2015;22(3):194–202. doi:10.1111/xen.12161

152. Reyes LM, Estrada JL, Wang ZY, et al. Creating class I MHC-null pigs using guide RNA and the Cas9 endonuclease. J Immunol. 2014;193(11):5751–5757. doi:10.4049/jimmunol.1402059

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.