Skip to main content
Kidney360 logoLink to Kidney360
. 2024 Feb 28;5(4):618–630. doi: 10.34067/KID.0000000000000400

Targeting the Kidneys at the Nanoscale: Nanotechnology in Nephrology

Anastasiia Vasylaki 1, Pratyusha Ghosh 1, Edgar A Jaimes 2,3, Ryan M Williams 1,4,
PMCID: PMC11093552  PMID: 38414130

Abstract

Kidney diseases, both acute and chronic, are a substantial burden on individual and public health, and they continue to increase in frequency. Despite this and an intense focus on the study of disease mechanisms, few new therapeutic approaches have extended to the clinic. This is in part due to poor pharmacology of many, if not most, therapeutics with respect to the sites of kidney disease within the glomerulus or nephron. Considering this, within the past decade, and more pointedly over the past 2 years, there have been substantial developments in nanoparticle systems to deliver therapeutics to the sites of kidney disease. Here, we provide a broad overview of the various classes of nanomaterials that have been developed to improve therapeutic development for kidney diseases, the strategy used to provide kidney accumulation, and briefly the disease models they focused on, if any. We then focus on one specific system, polymeric mesoscale nanoparticles, which has broadly been used over 13 publications, demonstrating targeting of the tubular epithelium with 26-fold specificity compared with other organs. While there have been several nanomedicines that have advanced to the clinic in the past several decades, including mRNA-based coronavirus disease vaccines and others, none have focused on kidney diseases specifically. In total, we are confident that the rapid advancement of nanoscale-based kidney targeting and a concerted focus by clinicians, scientists, engineers, and other stakeholders will push one or more of these technologies into clinical trials over the next decade.

Keywords: AKI, CKD, gene therapy, glomerulus, kidney, pharmacokinetics, proximal tubule

Introduction

Kidney Disease Therapies

Kidney diseases, both acute and chronic, are major and growing health burdens worldwide as risk factors, such as diabetes and hypertension, become more prevalent.13 AKI can arise from various etiologies, including chemotherapeutics or antibiotics, sepsis, ischemia, or others, and occurs in up to 67% of critically ill patients.4,5 CKD is hallmarked by glomerular and/or tubular fibrosis and progressive dysfunction, with causes ranging from diabetes, hypertension, or others, and it affects up to 10% of the population.6 In both AKI and CKD, depending on the severity, etiology, and baseline reserve, progression to ESKD can occur.

Therapeutics for both AKI and CKD are limited. ESKD is typically only manageable through RRT—dialysis or kidney transplant.7 There are no optimal pharmacologic interventions to prevent AKI or progression of CKD.7 BP control, management of underlying conditions such as diabetes, and avoidance of nephrotoxic drugs are key to avoid CKD progression. Renin angiotensin aldosterone system blockade, including angiotensin-converting enzyme inhibitors and angiotensin receptor blockers, as well as sodium–glucose cotransporter-2 blockade, slow kidney disease progression but no treatment has consistently reversed kidney injury.810 However, in many cases, these therapies fail to directly address underlying renal tissue damage and reverse progression of kidney dysfunction.11 There have been many clinical trials of potential pharmacologic agents to treat AKI or CKD; however, none have proven highly effective and safe in patient populations. This is in part due to a historical lack of strong therapeutic target identification, which is the subject of intense ongoing study. It is further due to difficulty in targeting effective therapies directly to the site of kidney damage,7 as normal kidney function is in opposition to typical drug uptake.

Nanomedicine

Nanomedicine is the development of clinically translational diagnostic or therapeutic engineered materials with at least one dimension on the nanoscale (1–1000 nm).12,13 One of the most prominent applications of nanomedicine is in drug and gene delivery, as these materials possess unique properties allowing them to improve pharmacologic characteristics of encapsulated therapeutics. Nanocarriers can be used to solubilize poorly water-soluble therapeutics and to achieve controlled drug release, resulting in lower frequency of dosing and increased patient compliance. Nanoparticles (NPs) are also able to preserve the integrity of easily degradable cargoes, such as biologics. Finally, in some circumstances nanomedicine enables drug targeting to specific organs and tissues, thus increasing drug concentration at a disease site, improving its efficacy, and reducing adverse effects in healthy tissues.14,15

Nanomedicines entered the pharmaceutical market almost three decades ago with the US Food and Drug Administration (FDA) approval of Doxil—the first NP therapeutic for cancer treatment.16 Since then, 30 other NP drugs have reached the clinic in the United States and Europe, and over 70 therapeutics are currently being investigated in clinical trials.17 NPs are used for various indications, including cancer therapies, iron replacement, fungal infections, macular degeneration, genetic liver disorders, and as imaging agents.1719 The most recent breakthrough in the nanomedicine field was the development of lipid NP mRNA vaccines.20 Lipid NP delivery of unstable mRNA protects the mRNA molecule, thus allowing cell uptake and preventing immune responses.2124 Undoubtedly, coronavirus disease 2019 NP vaccines have made a major effect on global health care, but they also advanced the possibility of clinical translation of other NP drug delivery systems by establishing their safety for use in wide populations and demonstrating efficacy in delivering advanced molecular therapeutics.25 Thus, the field of nanomedicine holds great promise for enabling the implementation of next-generation therapies and addressing previously unmet health care needs.

Those nanomaterials described above are made of lipid NPs,26,27 although there are several other broad categories of materials. Indeed, there are other FDA-approved nanomedicines on the basis of polymeric materials, proteins, and inorganic NPs.28 Iron oxide NPs have been widely used as iron-replaced therapies in anemic patients with CKD, with examples such as ferumoxytol and others.29,30 This is largely because colloidal nanoscale iron oxide is more stable than other means of administration. In addition, while those examples above are primarily therapeutic or prophylactic in nature, nanomaterials are widely used in the clinic for imaging and diagnostic examples. One common use of nanotechnology in a diagnostic application is to provide contrast (the “pink color”) in rapid coronavirus disease antigen tests or at-home pregnancy tests—which is achieved by gold nanoscale particles in a laminar flow immunoassay.31,32 In each case, the specific material choice is important as it may be fine-tuned for the expected function and safety, such as the cargo used and the specific disease indication.

Here, we broadly outlined the current state of the literature on the development of nanotechnologies for kidney diseases. By and large, these have been evaluated in rodent models of disease, and we were excited to see an explosion of new technologies and papers over just the past 2 years. We are confident that this burgeoning interest and ongoing preclinical successes will eventually translate to patient-centered therapeutic and diagnostic development.

Development of New NP Systems That Target the Kidneys is Growing

With the increasing prevalence of kidney diseases, NP-targeted kidney delivery presents a promising approach to improve the pharmacology of therapeutics with respect to the kidneys. Given these benefits, kidney-targeted drug delivery systems have gained interest from numerous research groups and have been covered in a number of review articles. Several papers have reviewed kidney-targeted treatments used for particular kidney conditions, including various etiologies of AKI and CKD, renal cell carcinoma, renal fibrosis, and other kidney diseases.3341 Another group of review articles has focused on nanomedicines targeting different parts of the nephron,42,43 specific cell types,37,39,44,45 and subcellular localization.44 Kidney targeting has also been described in relation to different drug carrier types, such as antibody conjugates, small molecule prodrugs, protein and peptide carriers, polymeric carriers, and NPs.37,43,46 For NP-based kidney treatments, passive and active targeting mechanisms have been detailed.39,44,47 Furthermore, a number of reviews have compiled the physicochemical properties of NPs that lead to kidney targeting, including the effect of size, shape, surface charge, composition, and surface modifications.39,41,4649 Thus, a growing body of literature on kidney-targeted drug delivery systems suggests the increasing importance of this approach in kidney disease management.

Given the recognized importance of kidney diseases and the substantial lack of therapeutic options for them, there has been a rapid increase in the number of novel NP systems designed to treat them. We performed a substantial literature search to identify these systems, finding publications primarily in materials and nanotechnology-based journals, with few disease-focused contexts (Table 1). Our search may have missed several published systems and therefore is likely not comprehensive, but any exclusions are unintentional. While we found a few systems (approximately five) which were published before 2015, the broad majority of publications we found were published within the past 2 years. Indeed, of the 35 particle systems in Table 1, 66% were published in 2021–2023. We outlined NP systems which the authors primarily described as kidney-targeted or that were used for kidney disease therapy. We should note, however, that a description of kidney targeting does not mean the particles exclusively localize in the kidneys nor does it mean that those particles are safe or effective in kidney disease therapy.

Table 1.

Nanoparticle systems which have shown some kidney localization or been used to treat kidney disease

Nanomaterial Tissue Within the Kidney Kidney Localization Mechanism Context or Demonstration
Lipid NPs/liposomes
 Liposome loaded with prednisolone50 Glomerular mesangium Cationic lipid shows selective affinity to the anionic cell surface and ECM in glomerular mesangial lesions Decreased deposition of IgA and C3 in glomeruli of ddY mice
 Phospholipid NPs with surface peptides and celastrol loading51 Glomerular podocytes Size, charge, and peptide modification mediated delivery shows affinity to VCAM1 Improved drug delivery to glomeruli, reduced toxicity of celastrol, reduced glomerular injury, and alleviated CKD in a mouse model through anti-inflammatory effect of the drug selectively delivered to endothelial cells and podocytes
 Liposomes loaded with triptolide52 Glomerular mesangium Cationic lipid TRX-20 shows high affinity to mesangial cells Triptolide-mediated immunosuppression, anti-inflammatory effects in membranous nephropathy rat model
 Liposomes coated with octa-arginine and loaded with siRNA against MAPK and p6553 Glomerular mesangium Size-based (110 nm) penetration through glomerular endothelium pores and retention in the glomerulus due to cationic charge and size larger than podocytes foot processes Reduced proteinuria, inflammation and ECM deposition in mouse model
 DSPE-PEG2000-folate and DSPE-PEG2000-methoxy amphiphile NPs54 Tubular epithelium Size-specific and folate-mediated passage through glomerulus Kidney accumulation in healthy mice
 Micelles with ([KKEEE]3K) kidney targeting peptide55 (same group as the above entry) Tubular epithelium ([KKEEE]3K) binds to megalin leading to receptor-mediated endocytosis Kidney accumulation in healthy mice
 Stearamine NPs loaded with enzymatic permanganate NPs56 Tubular epithelium Inflammation-mediated enhanced kidney accumulation Anti-inflammatory and antiapoptotic effects in the kidneys of IRI-AKI mice
Polymeric NPs
 PLL-PEG complexed with siRNA against MAPK157 Glomerular mesangium Penetration through glomerular endothelium fenestrae due to 10–20 nm size Reduced proteinuria and protein expression in mouse model of GN
 CDP-based siRNA NPs58 GBM Binding and disassembly by components of the renal filtration barrier influenced by NP size (10–100 nm), positive zeta potential, and electrostatically driven self-assembly GBM accumulation in healthy mice
 Chitosan NPs with metformin59 Tubular epithelium Megalin-mediated endocytosis Antiapoptotic, anti-inflammatory, and antifibrotic effect in ureteralUUO mice
 Chitosan/siRNA NPs targeting AQP160 Tubular epithelium Megalin-mediated endocytosis AQP1 gene silencing in healthy mice
 Chitosan/siRNA NPs targeting COX-261 Renal macrophages Phagocytic uptake by macrophages Prevention of unilateral ureteral obstruction-induced kidney damage
 PEG-PLGA NPs loaded with dexamethasone acetate62 Glomerular mesangium Penetration through glomerular endothelium fenestrae due to 90 nm diameter Glomerular mesangium targeting in healthy rats
 Polymeric nanosponges based on a phosphoester that scavenges ROS63 Tubular epithelium Increased microvascular permeability in AKI kidneys Treatment of AKI by downregulation of ROS, inflammation, and reduction of cell apoptosis in a mouse model
 Amphiphilic PAMAM polymer loaded with rosmarinic acid64 Tubular epithelium Serine binding to KIM-1 and charge-mediated passage through GFB Protection of cells from oxidative stress, decreased inflammatory response for therapy of AKI in a mouse model
 PEG-PCL-PEI copolymer loaded with rhein65 Glomerular mesangium Size-based penetration through glomerular endothelial membrane due to increased pore size in kidney disease Kidney targeting, improvement of fibrinogen levels, and kidney function markers in diabetic nephropathy mouse model
 Copolymer loaded with curcumin66 Tubular epithelium Size, charge-based penetration through GFB Alleviate mitochondrial injury, protect cells, and kidneys from oxidative stress in cisplatin-induced AKI mouse model
 PVP loaded with curcumin67 Tubular epithelium Size below renal excretion threshold (<10 nm) Lessened kidney damage and restored kidney function in cisplatin-induced AKI mouse model
 Hyaluronic acid conjugated to bilirubin loaded with a calcium chelator68 Tubular epithelium CD44 binding capacity of Hyaluronic acid Inhibition of activation of endoplasmic reticulum stress cascade, regulation of apoptosis pathway, reduced inflammatory response in AKI rat model
 PEGylated gambogic acid NPs69 Tubular epithelium Passage through GFB and enhanced retention in injured kidneys (<10 nm) Protection of cells from oxidative stress damage, improving renal damage by antiapoptotic and anti-inflammatory activity in cisplatin- and rhabdomyolysis-induced AKI mouse models
 Polyplex with siRNA against PCX70 Tubular epithelium CXCR4-mediated transport to tubules p53 gene silencing, improving kidney function and reduction in kidney damage therefore reducing AKI in cisplatin-induced and IRI-AKI mouse models
 Modified chitosan NPs loaded with siRNA against p5371 (same group as the above entry) Tubular epithelium Preferential internalization by injured tubule cells through CXCR4-mediated uptake Decreased kidney apoptosis, macrophage and neutrophil infiltration, improved kidney function in IRI-AKI mouse model
 PAMAM dendrimer modified with serine72 Tubular epithelium Size-based glomerular filtration and active transport (<10 nm) Kidney targeting in healthy mice
 Copolymer of sorbitol and PEI loaded with plasmid DNA73 Tubular epithelium Size-based endocytosis and targeting to vimentin Alport syndrome mouse model showed enhanced transfection efficiency and uptake by cells
 Pluronic NPs modified with folate loaded with triptolide74 Tubular epithelium Folate receptor-mediated endocytosis Reduced acute tubular injury index and renal function indexes in IRI-AKI mouse model
 Polyplex with albumin loaded with celastrol75 Tubular epithelium Megalin receptor-mediated internalization Improved kidney function markers and renal injury in IRI-AKI mouse model
 PLGA NPs loaded with oltipraz76 Tubular epithelium PLGA NPs with 100 nm diameter cross through impaired GFB Reduced tubular necrosis and collagen deposition, improved renal function and renal fibrosis in AKI mice model
Protein, peptide, and nucleic acid NPs
 Celastrol-albumin NPs77 Glomerular mesangium Size-based (95 nm) penetration through glomerular endothelium leading to accumulation in mesangial cells Alleviation of proteinuria, inflammation, and ECM deposition in rat GN model
 Albumin NP loaded with farnesyl thiosalicylic acid78 Glomerular mesangium Size-based (100 nm) penetration through glomerular endothelium and accumulation in mesangial space Alleviation of renal fibrosis in UUO-induced renal fibrosis mouse model
 DNA origami nanostructures79 Tubular epithelium Glomerular endothelial fenestrae filtration influenced by morphology and size Amelioration of rhabdomyolysis-induced AKI in mice by ROS scavenging
 Small-sized DNA tetrahedrons with p53 siRNA80 Tubular epithelium Size-based filtration through GBM and endocytosis into tubular cells (<10 nm) siRNA-induced gene downregulation in AKI mouse model
 Tetrahedral nucleic acid nanostructure loaded with typhaneoside81 Tubular epithelium Size near renal excretion threshold and enhanced retention in AKI Increased apoptotic and antioxidative function with kidney function restoration in IRI-AKI mouse model
 Crotamine (cell-penetrating peptide)/siRNA nanocomplexes82 Tubular epithelium Syndecan-1–mediated internalization in the brush border zone of PTECs Accumulation in PTECs of healthy mice after IP administration
 L-serine-modified Poly-l-Lysine with radiotracer83 Tubular epithelium Size-based filtration in the glomerulus and absorption in the lumen of proximal tubule Reduced renal tumor growth and nephrotoxicity in a mouse model
 Protein-based nanocage84 Tubular epithelium Size-based filtration through glomerular endothelium and reabsorption by proximal tubules Mitigation of proximal tubular damage in a mouse model of sepsis-induced kidney injury
Inorganic NPs
 Perfluorocarbon NP with collagen IV loaded with prednisone85 GBM Col4‐targeting ligand selectively binds to collagen IV on GBM Decreased IgG and C3 deposition, reduced proteinuria, improved GFR presentation, reduced glomerular pathology in lupus nephritis mouse model
 Quantum dots that chelate iron86 Tubular epithelium Size-based penetration through glomerular endothelial membrane (<10 nm) Decreased ferroptosis and apoptosis, removal of iron species and ROS in cisplatin-induced AKI mouse model
 Selenium NPs with albumin87 Tubular epithelium Endocytosis by RTEC Suppression of inflammasome and cytokines in IRI-AKI mouse model
 Iron oxide NPs loaded with nicotinamide88 Tubular epithelium Size-based penetration through glomerular endothelial membrane and NRK1-mediated cellular uptake Repair of renal structure, restoration of eGFR and hemoglobin elevation in AKI mouse model
 Gold NPs89 Tubular epithelium Size-based penetration through GFB (<10 nm) Reduction of ROS and apoptosis in a mouse model of subclinical AKI
 Gold NPs with PEG90 Glomerular mesangium Size-based targeting (approximately 75±25 nm) Mesangium targeting in healthy mice
 Hydrogenated germanene nanosheet91 Tubular epithelium DNA-like framework and negative surface charge and PEG modification-based accumulation Antioxidative protection against ROS in AKI mouse model
 Carbon nanotubes with siRNA conjugated against p53, Mep1b, Ctr1, and EGFP92 Tubular epithelium Glomerular filtration and reabsorption at proximal tubular cell brush border Prevention of cisplatin-induced AKI in murine model
 Mesoporous silica NP loaded with BAPTA93 Tubular epithelium Size-based penetration through glomerular membrane and active targeting with KIM-1 targeted peptide Antiapoptotic and anti-inflammatory effect restoring kidney function in a rat IRI-AKI model
 Zeolite imidazolate NPs coated with tubular epithelial cell membrane94 Tubular epithelium Active targeting through RTEC membrane coating and modification with kidney targeting peptide Attenuation of oxidative and inflammatory damage and recovery of renal function in sepsis-induced AKI murine model

“Kidney localization mechanism” does not necessarily signify that the kidney is the only, or primary, site of tissue accumulation in the body. AQP1, aquaporin 1; BAPTA, O,O′-bis(2-aminophenyl)ethyleneglycol-N,N,N′,N'-tetraacetic acid; CD44, cluster of differentiation 44, a cell surface glycoprotein; CDP, cyclodextrin-containing polymer; CLT, celastrol, an active ingredient with anti-inflammatory, antioxidant, immunosuppressive, and antitumor effects56; COX-2, cyclooxygenase type 2; Ctr1, copper transport protein 1; CXCR4, C-X-C chemokine receptor 4; DSPE-PEG2000, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000]; ECM, extracellular matrix; EGFP, enhanced green fluorescent protein; ER, endoplasmic reticulum; FBG, fasting blood glucose; GBM, glomerular basement membrane; GFB, glomerular filtration barrier; IV, intravenous; IRI, ischemia–reperfusion-induced; KIM-1, kidney injury molecule-1; MAPK, mitogen-activated protein kinase; Mep1b, meprin-1β; NP, nanoparticle; NRK1, nicotinamide riboside kinase 1; Oltipraz, a drug for treatment of AKI and renal fibrosis81; PAMAM, polyamidoamine; PCX, polymeric CXCR4 antagonist; PEG-PCL-PEI, polyethyleneglycol-co-polycaprolactone-co-polyethylenimine; PEG-PLGA, polyethylene glycol-poly(lactic-coglycolic acid); PEI, polyethylenimine; PLGA, poly(lactic-co-glycolic acid); PLL-PEG, poly(l-lysine)-poly(ethylene glycol); PTEC, proximal tubular epithelial cells; PVP, polyvinylpyrrolidone; RTEC, renal tubular epithelial cell; ROS, reactive oxygen species; siRNA, short interfering RNA; TP, triptolide, a drug with immunosuppressive properties57; TRX-20, 3,5-dipentadecyloxybenzamidine hydrochloride; UUO, unilateral uretral obstruction; VCAM1, vascular cell adhesion molecule 1ureteral.

Generally, the NP systems that we found can be broken down among the broader classes of NPs (Figure 1 and Table 1): (1) lipid NPs or liposomes, (2) polymer NPs, (3) protein or nucleic acid NPs, and (4) inorganic NPs. This follows broad trends in the nanotechnology field and gives some insight as to the diversity of approaches currently being studied in the “kidney nanomedicine revolution.” Several of these systems have been tracked by the investigators over several publications, including particles which pass through glomerular fenestrations allowing component breakdown and tubular delivery,58,90,95 size-based carbon nanotube penetration through the glomerulus and reabsorption by proximal tubules which has been studied in mice and nonhuman primates,92,96 and lipid micellar particles with active targeting agents.47,54,55

Figure 1.

Figure 1

NP systems and their uses in kidney diseases. Examples of major classes of NPs used in kidney-specific targeting are shown on the left. On the right, the nephron is shown with examples of strategies that have been used to target the glomerulus and tubules. CD44, cluster of differentiation 44, a cell surface glycoprotein; CXCR4, chemokine receptor type 4, a stromal-derived factor 1 receptor; NP, nanoparticle; PAMAM, polyamidoamine, a hyperbranched polymer dendrimer; PCL-PEI, polycaprolactone-polyethylene imine, an amphiphilic block copolymer; PLGA-PEG, poly (lactic-co-glycolic acid)-polyethylene glycol, an amphiphilic block copolymer; PVP, polyvinylpyrrolidone, a hydrophilic polymer; SWCNT, single-walled carbon nanotube, a nanoscale carbon tubule.

The kidney-targeting mechanisms, or mechanisms for enhanced renal accumulation, fall into two broad categories (Figure 1): (1) passive targeting through largely size-based accumulation and (2) active targeting, wherein a specific molecule that binds to renal cells was used. Several passive targeting approaches were used, including: (1) small NPs (<10 nm) which pass through the glomerular filtration barrier and are reabsorbed in the nephron, (2) somewhat larger NPs which pass through the glomerular endothelial fenestrations and arrest in the glomerular mesangium (typically up to 80 nm), (3) somewhat larger (approximately 100 nm) NPs which pass through only disrupted glomerular filtration barriers and are reabsorbed in the nephron, and (4) NPs which do not interact with the glomerulus and cross from the peritubular endothelium into the tubular epithelium. Active targeting of NPs to the kidneys has taken several different approaches, including serine modifications which purportedly bind kidney injury molecule-1,64,71,83,93 charge-based binding to the megalin receptor,55,59,60,75 or other cell surface receptor or basement membrane binding.51,68,70,85

Regarding therapeutic cargoes, we found that many of the active pharmaceutical ingredients (APIs) involved short interfering RNA (siRNA) delivery—about 10 of those we found. Many others incorporated natural products, often with reactive oxygen species scavenging capabilities, or the material itself acting as a reactive oxygen species scavenger.63,79 The disease models which were investigated have similarly been wide-ranging. AKI mouse models of several etiologies have been investigated, as well as fibrotic CKD and glomerular nephropathies. A few, but not many, studies investigated therapeutic efficacy in renal carcinoma models, while some performed biodistribution studies in healthy animals. In most cases, positive therapeutic efficacy was reported as it relates to one or several renal function outcomes, and no toxicity was found if reported.

Polymeric Mesoscale NPs—Tubular Kidney Targeting at the Meso-Nanoscale

As we outlined above, there are a variety of NP systems that either target the kidneys with some selectivity or demonstrate therapeutic efficacy in kidney disease models or both. The compilation of these systems and their potential use has been described in an array of recent review articles, which we overviewed above. One such system, polymeric mesoscale NPs (MNPs), has demonstrated both the most substantial renal selectivity, as well as the most flexibility in therapeutic payload and disease indication (Figure 2). To date, there have been 13 publications based on this delivery platform, with several directly from our group, some with collaborators, and some from other groups independently. Generally, if the authors ascribed their kidney targeting mechanism to the particles being in the mesoscale range, we considered those here as opposed to above.

Figure 2.

Figure 2

MNPs target the proximal tubular epithelium and are therapeutically effective in kidney disease. (A) Scanning electron micrograph of MNPs. (B) IVIS fluorescence image focused on the kidneys overlain on a CT of a mouse injected with fluorescent dye-loaded MNPs (reprinted with permission from Williams et al.97 Copyright 2015 American Chemical Society). (C) Fluorescence micrograph of renal tissue after injection of MNPs. Blue is nuclei, green is proximal tubular epithelial lumen, and red is anti-PEG staining for MNPs. (reprinted from Han 2020 Kidney International with permission from Elsevier98). (D) BP of db/db mice treated with control siRNA-loaded MNPs or IL-1β siRNA-loaded MNPs. (reprinted with permission from Veiras et al. 2022 Circulation Research with permission from Elsevier99). CT, computed tomography; MNP, mesoscale nanoparticle; siRNA, short interfering RNA.

In our prior work, we published that polymeric NPs in the so-called “mesoscale” range, approximately 300–500 nm in diameter, demonstrated substantial kidney targeting.97,101 These particles were initially formulated via nanoprecipitation using FDA-approved di-block polymers poly(lactic-co-glycolic acid) conjugated to polyethylene glycol. The first two publications on MNPs were focused on understanding their pharmacology, primarily using a hydrophobic Cy5 (cyanine) fluorescent dye cargo. Experiments investigated routes and concentrations of administration, with almost all studies using an intravenous dose of 10–100 mg/kg MNP. Using in vivo and ex vivo imaging coupled with immunohistochemistry, published studies determined that MNPs localize to the kidneys 26-fold greater than any other organ investigated.97,101,102 Proximal tubular epithelial cells were the primary tissue of localization, with some distal tubular localization (approximately 2:1 proximal:distal) and no glomerular uptake. Those studies concluded that MNPs were not filtered by the glomerulus, but instead transcytosed across the peritubular epithelium into the basolateral membrane of tubular epithelial cells. Furthermore, these experiments found no evidence of toxicity either systemically or localized in the kidneys. Generally, these particles and their cargoes have demonstrated long renal retention times, up to several days or weeks, with controlled release and extended pharmacodynamic profiles. Given this safety and kidney targeting profile, MNPs were further used as carriers for therapeutic cargoes in kidney diseases.

Diversity of MNP Cargoes: Small Molecules, Peptides, and Nucleic Acids/siRNA

Polymeric MNPs which target the kidneys with high selectivity have been demonstrated to encapsulate and deliver a broad range of various cargoes (Table 2). Modifications from those original dye-loaded MNP formulations were required to ensure that the size (300–500 nm diameter in all studies) and overall surface chemistry (PEGylated) were similar to ensure kidney targeting.

Table 2.

Pharmacologic and therapeutic studies using polymeric mesoscale nanoparticles

MNP Payload Disease Target/Use Therapeutic Outcomes (Compared with Baseline Control) Pharmacokinetics Notes
Fluorescent Cy5 dye97 Healthy mice/pharmacology N/A Seven-fold kidney targeting, tubular specific by IHC (PEG) Initial discovery and characterization, compared with free dye
Fluorescent Cy5 dye101 Healthy mice/pharmacology N/A 26-fold kidney targeting by IV and dose modulation, PTEC specific, no toxic effects in liver, serum, kidneys Dose and route modulation, pharmacology study, compared with free dye
ODN2088 selective TLR9 antagonist, nucleic acid98 Ischemic AKI 60%–90% reduction in creatinine, BUN, H&E injury, NGAL, infiltration of macrophages and neutrophils, apoptosis, cytokine signaling 30-fold kidney targeting (Cy5 dye) in healthy and IR mice, PTEC specific by IF (PEG) Compared with free drug control with positive benefits
Triptolide small molecule, Cy7 dye102 Ischemic AKI 80%–100% reduction in H&E injury, creatinine, BUN, C3, apoptosis Eight-fold kidney targeting (drug LC-MS and Cy7 fluorescence); no toxic effects in liver or other organs, protective effect from drug toxicity First replication by external groups, compared with free drug with benefits
NEMO binding peptide103 Ischemic AKI 40%–60% reduction in creatinine, BUN, H&E injury, NGAL, apoptosis, neutrophil infiltration, cytokine signaling PTEC specific by IHC (PEG) with no staining in other organs Similar results compared with NEMO-deleted mice
Emodin small molecule, Cy7 dye104 UUO CKD Reversal of fibrosis by H&E (not quantified) Primarily kidney targeting (not quantified), no in vitro toxicity
Sirolimus small molecule105 ADPKD rats (Pkhd1PCK/PCK) 75%–95% reduction in renal cyst volume, pS6/S6 ratio Increased body and heart weight compared with free sirolimus, indicated less toxicity compared with free drug Published conference proceedings, stronger performance than free drug
Formoterol small molecule106,107 Healthy mice/pharmacology Increase in mitochondrial biogenesis in the renal cortex, none in the heart Proximal tubular localization by IF (PEG), 15-fold enhancement of drug targeting compared with free drug
Formoterol small molecule, Cy5 dye108 Ischemic AKI 80%–100% reduction in creatinine, KIM-1, NGAL, fibrosis, increase in mitochondrial production No effects on heart rate or BP (free drug showed effects) Size 463–493 nm. Compared effects with free drug control at higher dose with benefits
Edaravone small molecule100,109 Cisplatin-induced AKI 70%–100% reduction in creatinine and BUN, NGAL, oxidative stress by IHC (nitrotyrosine) PTEC-specific targeting by IHC (PEG), renal accumulation compared with none with free drug Preprint demonstrates kidney targeting in flank xenograft and lung tumor-bearing mice
Renalase agonist peptide RP81110 Cisplatin-induced CKD in renalase knockout and WT mice 70% reduction in creatinine, KIM-1, inflammation, cell death PTEC-specific targeting by IHC (RP81)
siRNA against PDL1111 Hypertensive CKD in mouse DOCA+salt model 100% reduction in PDL1 expression, reduction in CD8 T cells, baseline BP No effects on BP of MNPs alone, no MNP-induced T cell infiltration, no siRNA-induced KD in lung, liver, or spleen Showed results similar to IFNγ knockout mice, single dose showed KD 18 d later
siRNA against IL-1β99 Diabetic kidney disease (db/db mice) 75%–100% reduction in IL-1β expression compared with baseline, baseline BP, and inflammation, salt sensitivity No effects on body weight, glucose, insulin; no siRNA-induced KD in plasma, heart, aorta, iver, spleen; no effects on BP alone Showed results similar to knockout of IL-1 receptor, MNPs dosed twice/every other week
siRNA against OCT 1 and 2, p53, PKCδ, γGT112 Cisplatin-induced AKI 50%–80% reduction in creatinine, BUN, fibrosis (PAS), target KD No cell toxicity MNPs were chitosan with PEG coating

ADPKD, autosomal dominant polycystic kidney disease; DOCA, deoxycorticosteroid acetate; γGT, γ-glutamyl transpeptidase; H&E, hematoxylin and eosin stain; IF, immunofluorescence; IHC, immunohistochemistry; IR, ischemia–reperfusion; IV, intravenous; KD, knockdown; KIM-1, kidney injury molecule-1; LC-MS, liquid chromatography-mass spectrometry; MNP, mesoscale nanoparticle; NEMO, nuclear factor-κB essential modulator; N/A, not applicable; NGAL, neutrophil gelatinaseassociated lipocalin; OCT, organic cation transporter; PAS, periodic acid–Schiff staining; PDL1, programmed death-ligand 1; PKCδ, protein kinase C delta; PTEC, proximal tubular epithelial cell; siRNA, short interfering RNA; TLR9, toll-like receptor 9; UUO, unilateral uretral obstruction; WT, wild type.

Several small molecule cargoes have been successfully encapsulated and delivered to the kidneys via MNPs. Encapsulation of small molecules is relatively straightforward in the nanoprecipitation process for drugs, with minor modifications to account for drug solubility. This includes hydrophobic small molecules, such as Cy5 and Cy7 dyes, that have been used for imaging and biodistribution studies.97,101,102,104,108 It also includes hydrophobic APIs formoterol,106108 triptolide,102 emodin,104 and rapamycin,105 as well as slightly hydrophilic edaravone.100 These small molecules cover several mechanisms of action: β2-adrenergic receptor agonist formoterol; anti-inflammatory natural products triptolide and emodin; redox scavenging for edaravone; and mammalian target of rapamycin inhibitor rapamycin. It is important to note that several of the above APIs were directly compared with free drug without MNP delivery or indirectly compared with prior studies. In each case, MNP-enabled delivery to the kidneys demonstrated a favorable therapeutic and safety profile compared with the free drug alone.

Biologically active nucleic acid cargoes have also been demonstrated to be therapeutically efficacious in kidney diseases following MNP-encapsulated delivery. Slight modifications in formulation are typically necessary for such large hydrophilic APIs, usually incorporating a solvent-aqueous phase mixture before nanoprecipitation. The first such example was an oligodinucleotide toll-like receptor 9 antagonist ODN2088.98 There have since been three different publications demonstrating therapeutic siRNA-loaded MNP formulation and kidney delivery: siRNA targeting the inflammatory cytokine IL-1β,100 the CD8 T-cell regulating programmed death-ligand 1,111 and simultaneous delivery of an siRNA cocktail against organic cation transporters—1 and 2, p53, protein kinase δ, and γ-glutamyl transpeptidase.112 It is exciting that, in all cases, kidney-specific knockdown of the siRNA target was demonstrated, in one case up to 3 weeks after a single dose, with no knockdown in other organs observed.111 In addition, loading of mRNA into MNPs and expression of a reporter protein (mCherry) in renal tubular cells in vitro has been reported.113 The ability to load and deliver functional nucleic acids to the kidneys, and generally anywhere beyond the liver, is an extremely important and exciting development in gene delivery science. The plug-and-play nature of siRNA and mRNA, coupled with recent developments in those fields, potentiates many therapeutic applications across a broad range of kidney diseases.

An additional category of demonstrated MNP-loaded APIs is peptides and proteins. There have been two manuscripts published using these cargoes: a peptide binding to nuclear factor-κB essential modulator103 and a peptide agonist of renalase.110 In both cases, MNP-targeted peptide delivery demonstrated superior efficacy to free peptide, although ongoing work is necessary to validate full-protein delivery.

Therapeutic Efficacy in Kidney Disease Models: AKI and CKD of Various Etiologies

While there have been several types of cargoes loaded into MNPs, they have similarly been applied to kidney disease models of several etiologies. Both acute and chronic models have been investigated, with a variety of dosing strategies, including single-dose and multidose regimens. Furthermore, basic pharmacology has been studied in both healthy and diseased rodents, with original discovery and validation of MNPs occurring in hairless immunocompetent and wild type mouse strains.97,101,106,107 Interestingly, kidney-specific targeting has been demonstrated in nude and nod-scid gamma mice bearing flank and orthotopic lung tumors in work designed to avoid NP localization to tumors in chemotherapy-associated AKI.109 It should be noted that most studies have been performed in mice to date, with one initial work in rats.

AKI models have been the most widely investigated for MNP application. The most common AKI etiology has been the ischemia–reperfusion model, with therapeutic effects stemming from a toll-like receptor 9 antagonist oligodinucleotide, nuclear factor-κB essential modulator binding peptide, formoterol, and the small molecule triptolide.98,102,103,108 Cisplatin-induced models of AKI are also commonly used in the field, with successful demonstration of both an siRNA cocktail and small molecule edaravone delivery.100,112 Although the siRNA cocktail delivery and triptolide delivery studies were administered daily for 3 days, the other AKI studies described here were single administrations associated with model initiation or up to 24 hours after.

MNP-targeted delivery and efficacy has also been demonstrated in CKD models. These included the unilateral ureteral obstruction model,104 a chronic cisplatin-induced model,110 hypertensive deoxycorticosteroid acetate+salt mice,111 and hypertensive diabetic (db/db) mice (Figure 2D).100 Notably, initial studies in a polycystic kidney disease model have also been published as a proffered abstract using an autosomal recessive polycystic kidney disease rat model, administering rapamycin-MNPs twice weekly for 8 weeks. In these models, only the programmed death-ligand 1–targeted siRNA-MNPs were administered just once (deoxycorticosteroid acetate+salt), while the anti-IL-1β siRNA-MNPs were administered every other week for 6 weeks (db/db mice), emodin-MNPs were administered daily (unilateral uretral obstruction), and renalase peptide MNPs were dosed weekly for 4 weeks (cisplatin CKD).

Conclusions and Call to Action

Here, we described the state of the art regarding several highly promising NP systems for kidney-targeted drug delivery, with a specific focus on one system that has demonstrated the broadest utility to date, albeit for tubule-specific targeting. Many of the studies have considerably strong results in preclinical rodent models of disease. Notably, several of these studies extended beyond basic formulation and demonstration of the materials, instead focusing on using kidney-targeted NPs as chemical biology tools to better understand underlying disease processes through targeted pathway inhibition.114 However, substantial work remains to translate these toward clinical utility. Proceeding to investigational new drug applications and clinical trials will be disease-specific and investigator-driven, meaning decisions on which cargoes to use and pathways to target must be made rationally based on substantial preclinical evidence. This will include full preclinical pharmacology and toxicology studies in both rodents and larger mammals, such as pigs or nonhuman primates. Design and carry forward of such a potential agent will require buy-in and collaboration of industry, clinicians, the venture community, and others. We are confident, however, that given the recent clinical successes of nanomedicines, that this class of therapies will soon make an effect on the kidney disease community.

Acknowledgments

The authors acknowledge all members of the Williams and Jaimes Labs for their assistance, insight, and discussion. We appreciate assistance with artwork by P. Jena.

Footnotes

A.V. and P.G. contributed equally to this work.

Disclosures

R.M. Williams reports the following: Consultancy: Goldilocks Therapeutics, Inc.; Ownership Interest: Goldilocks Therapeutics, Inc.; Patents or Royalties: Patents pending regarding kidney targeting NP formulation, no royalties received. Advisory or Leadership Role: Goldilocks Therapeutics, Inc. advisory role; and Other Interests or Relationships: Goldilocks Therapeutics, Inc. All remaining authors have nothing to disclose.

Funding

R.M. Williams: Office of Extramural Research, National Institutes of Health (CA132378), Oak Ridge Associated Universities Ralph E. Powe Junior Faculty Enhancement Award, Professional Staff Congress of The City University of New York (PSC-CUNY) Enhanced Award, and The City College of New York Grove School of Engineering and Department of Biomedical Engineering. E.A. Jaimes: CCNY-MSKCC Partnership for Cancer Research (CA137788).

Author Contributions

Conceptualization: Pratyusha Ghosh, Edgar A. Jaimes, Anastasiia Vasylaki, Ryan M. Williams.

Funding acquisition: Edgar A. Jaimes, Ryan M. Williams.

Visualization: Anastasiia Vasylaki, Ryan M. Williams.

Writing – original draft: Pratyusha Ghosh, Edgar A. Jaimes, Anastasiia Vasylaki, Ryan M. Williams.

Writing – review & editing: Pratyusha Ghosh, Edgar A. Jaimes, Anastasiia Vasylaki, Ryan M. Williams.

References

  • 1.Chertow GM, Burdick E, Honour M, Bonventre JV, Bates DW. Acute kidney injury, mortality, length of stay, and costs in hospitalized patients. J Am Soc Nephrol. 2005;16(11):3365–3370. doi: 10.1681/ASN.2004090740 [DOI] [PubMed] [Google Scholar]
  • 2.Silver SA, Chertow GM. The economic consequences of acute kidney injury. Nephron. 2017;137(4):297–301. doi: 10.1159/000475607 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Wang HE, Muntner P, Chertow GM, Warnock DG. Acute kidney injury and mortality in hospitalized patients. Am J Nephrol. 2012;35(4):349–355. doi: 10.1159/000337487 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hoste EA Kellum JA Karz NM, et al. Epidemiology of acute kidney injury. In: Cardiorenal Syndromes in Critical Care. Karger Publishers; 2010:1–8. [Google Scholar]
  • 5.Abuelo JG. Normotensive ischemic acute renal failure. N Engl J Med. 2007;357(8):797–805. doi: 10.1056/NEJMra064398 [DOI] [PubMed] [Google Scholar]
  • 6.Kovesdy CP. Epidemiology of chronic kidney disease: an update 2022. Kidney Int. 2022;12(1):7–11. doi: 10.1016/j.kisu.2021.11.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bellomo R, Kellum JA, Ronco C. Acute kidney injury. Lancet. 2012;380(9843):756–766. doi: 10.1016/S0140-6736(11)61454-2 [DOI] [PubMed] [Google Scholar]
  • 8.Ruggenenti P, Cravedi P, Remuzzi G. Mechanisms and treatment of CKD. J Am Soc Nephrol. 2012;23(12):1917–1928. doi: 10.1681/ASN.2012040390 [DOI] [PubMed] [Google Scholar]
  • 9.USRDS. Annual Data Report: Atlas of Chronic Kidney Disease and End Stage Renal Disease in the United States. National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases; 2013. [Google Scholar]
  • 10.Go AS, Chertow GM, Fan D, McCulloch CE, Hsu Cy. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N Engl J Med. 2004;351(13):1296–1305. doi: 10.1056/NEJMoa041031 [DOI] [PubMed] [Google Scholar]
  • 11.Jo SK, Rosner MH, Okusa MD. Pharmacologic treatment of acute kidney injury: why drugs haven’t worked and what is on the horizon. Clin J Am Soc Nephrol. 2007;2(2):356–365. doi: 10.2215/CJN.03280906 [DOI] [PubMed] [Google Scholar]
  • 12.Silva GA. Introduction to nanotechnology and its applications to medicine. Surg Neurol. 2004;61(3):216–220. doi: 10.1016/j.surneu.2003.09.036 [DOI] [PubMed] [Google Scholar]
  • 13.Soares S, Sousa J, Pais A, Vitorino C. Nanomedicine: principles, properties, and regulatory issues. Front Chem. 2018;6:360. doi: 10.3389/fchem.2018.00360 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gao J, Karp JM, Langer R, Joshi N. The future of drug delivery. Chem Mater. 2023;35(2):359–363. doi: 10.1021/acs.chemmater.2c03003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Williams RM, Jaimes EA, Heller DA. Nanomedicines for kidney diseases. Kidney Int. 2016;90(4):740–745. doi: 10.1016/j.kint.2016.03.041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Barenholz YC. Doxil®-the first FDA-approved nano-drug: lessons learned. J Control Release. 2012;160(2):117–134. doi: 10.1016/j.jconrel.2012.03.020 [DOI] [PubMed] [Google Scholar]
  • 17.Anselmo AC, Mitragotri S. Nanoparticles in the clinic: an update post COVID-19 vaccines. Bioeng Transl Med. 2021;6(3):e10246. doi: 10.1002/btm2.10246 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Mullard A. FDA approves landmark RNAi drug. Nat Rev Drug Discov. 2018;17(9):613–614. doi: 10.1038/nrd.2018.152 [DOI] [PubMed] [Google Scholar]
  • 19.Akinc A Maier MA Manoharan M, et al. The Onpattro story and the clinical translation of nanomedicines containing nucleic acid-based drugs. Nat Nanotechnol. 2019;14(12):1084–1087. doi: 10.1038/s41565-019-0591-y [DOI] [PubMed] [Google Scholar]
  • 20.Reichmuth AM, Oberli MA, Jaklenec A, Langer R, Blankschtein D. mRNA vaccine delivery using lipid nanoparticles. Ther Deliv. 2016;7(5):319–334. doi: 10.4155/tde-2016-0006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Let’s talk about lipid nanoparticles. Nat Rev Mater. 2021;6(2):99.doi: 10.1038/s41578-021-00281-4 [DOI] [Google Scholar]
  • 22.Department of Health and Human Services, C.f.D.C. Centers for Disease Control and Prevention. COVID Data Tracker; 2023. Accessed September 29, 2023. https://covid.cdc.gov/covid-data-tracker [Google Scholar]
  • 23.Pfizer. COVID-19 Vaccine Equity. Accessed September 29, 2023. https://www.pfizer.com/science/coronavirus/vaccine/working-to-reach-everyone-everywhere [Google Scholar]
  • 24.Moderna. Global Vaccination: The Way Forward. Accessed September 29, 2023. https://www.modernatx.com/en-US/media-center/all-media/blogs/global-vaccination-way-forward [Google Scholar]
  • 25.Milane L, Amiji M. Clinical approval of nanotechnology-based SARS-CoV-2 mRNA vaccines: impact on translational nanomedicine. Drug Deliv Transl Res. 2021;11(4):1309–1315. doi: 10.1007/s13346-021-00911-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Musielak E, Feliczak-Guzik A, Nowak I. Synthesis and potential applications of lipid nanoparticles in medicine. Materials. 2022;15(2):682. doi: 10.3390/ma15020682 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Tenchov R, Bird R, Curtze AE, Zhou Q. Lipid nanoparticles─from liposomes to mRNA vaccine delivery, a landscape of research diversity and advancement. ACS Nano. 2021;15(11):16982–17015. doi: 10.1021/acsnano.1c04996 [DOI] [PubMed] [Google Scholar]
  • 28.Mitchell MJ, Billingsley MM, Haley RM, Wechsler ME, Peppas NA, Langer R. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov. 2021;20(2):101–124. doi: 10.1038/s41573-020-0090-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.McCormack PL. Ferumoxytol: in iron deficiency anaemia in adults with chronic kidney disease. Drugs. 2012;72(15):2013–2022. doi: 10.2165/11209880-000000000-00000 [DOI] [PubMed] [Google Scholar]
  • 30.Lu M, Cohen MH, Rieves D, Pazdur R. FDA report: Ferumoxytol for intravenous iron therapy in adult patients with chronic kidney disease. Am J Hematol. 2010;85(5):315–319. doi: 10.1002/ajh.21656 [DOI] [PubMed] [Google Scholar]
  • 31.Ardekani LS, Thulstrup PW. Gold nanoparticle-mediated lateral flow assays for detection of host antibodies and COVID-19 proteins. Nanomaterials. 2022;12(9):1456. doi: 10.3390/nano12091456 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wang J Drelich AJ Hopkins CM, et al. Gold nanoparticles in virus detection: recent advances and potential considerations for SARS-CoV-2 testing development. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2022;14(1):e1754. doi: 10.1002/wnan.1754 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yang K, Shang Y, Yang N, Pan S, Jin J, He Q. Application of nanoparticles in the diagnosis and treatment of chronic kidney disease. Front Med (Lausanne). 2023;10:1132355. doi: 10.3389/fmed.2023.1132355 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chen Q Nan Y Yang Y, et al. Nanodrugs alleviate acute kidney injury: manipulate RONS at kidney. Bioactive Mater. 2023;22:141–167. doi: 10.1016/j.bioactmat.2022.09.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bishop B, Sharma S, Scott EA. Nanomedicine in kidney disease. Curr Opin Nephrol Hypertens. 2023;32(4):366–376. doi: 10.1097/MNH.0000000000000897 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Paluszkiewicz P, Martuszewski A, Zaręba N, Wala K, Banasik M, Kepinska M. The application of nanoparticles in diagnosis and treatment of kidney diseases. Int J Mol Sci. 2021;23(1):131. doi: 10.3390/ijms23010131 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Li H, Dai W, Liu Z, He L. Renal proximal tubular cells: a new site for targeted delivery therapy of diabetic kidney disease. Pharmaceuticals. 2022;15(12):1494. doi: 10.3390/ph15121494 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Gu X Liu Z Tai Y, et al. Hydrogel and nanoparticle carriers for kidney disease therapy: trends and recent advancements. Prog Biomed Eng. 2022;4(2):022006. doi: 10.1088/2516-1091/ac6e18 [DOI] [Google Scholar]
  • 39.Oroojalian F Charbgoo F Hashemi M, et al. Recent advances in nanotechnology-based drug delivery systems for the kidney. J Control Release. 2020;321:442–462. doi: 10.1016/j.jconrel.2020.02.027 [DOI] [PubMed] [Google Scholar]
  • 40.Ma Y, Cai F, Li Y, Chen J, Han F, Lin W. A review of the application of nanoparticles in the diagnosis and treatment of chronic kidney disease. Bioactive Mater. 2020;5(3):732–743. doi: 10.1016/j.bioactmat.2020.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Jiang D, Rosenkrans ZT, Ni D, Lin J, Huang P, Cai W. Nanomedicines for renal management: from imaging to treatment. Acc Chem Res. 2020;53(9):1869–1880. doi: 10.1021/acs.accounts.0c00323 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Huang X, Ma Y, Li Y, Han F, Lin W. Targeted drug delivery systems for kidney diseases. Front Bioeng Biotechnol. 2021;9:683247. doi: 10.3389/fbioe.2021.683247 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Liu C-P, Hu Y, Lin JC, Fu HL, Lim LY, Yuan ZX. Targeting strategies for drug delivery to the kidney: from renal glomeruli to tubules. Med Res Rev. 2019;39(2):561–578. doi: 10.1002/med.21532 [DOI] [PubMed] [Google Scholar]
  • 44.Li Z Fan X Fan J, et al. Delivering drugs to tubular cells and organelles: the application of nanodrugs in acute kidney injury. Nanomedicine. 2023;18(21):1477–1493. doi: 10.2217/nnm-2023-0200 [DOI] [PubMed] [Google Scholar]
  • 45.Chen Z, Peng H, Zhang C. Advances in kidney-targeted drug delivery systems. Int J Pharm. 2020;587:119679. doi: 10.1016/j.ijpharm.2020.119679 [DOI] [PubMed] [Google Scholar]
  • 46.Hauser PV, Chang HM, Yanagawa N, Hamon M. Nanotechnology, nanomedicine, and the kidney. Appl Sci. 2021;11(16):7187. doi: 10.3390/app11167187 [DOI] [Google Scholar]
  • 47.Huang Y, Wang J, Jiang K, Chung EJ. Improving kidney targeting: the influence of nanoparticle physicochemical properties on kidney interactions. J Control Release. 2021;334:127–137. doi: 10.1016/j.jconrel.2021.04.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Adhipandito CF, Cheung SH, Lin YH, Wu SH. Atypical renal clearance of nanoparticles larger than the kidney filtration threshold. Int J Mol Sci. 2021;22(20):11182. doi: 10.3390/ijms222011182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Du B, Yu M, Zheng J. Transport and interactions of nanoparticles in the kidneys. Nat Rev Mater. 2018;3(10):358–374. doi: 10.1038/s41578-018-0038-3 [DOI] [Google Scholar]
  • 50.Liao J, Hayashi K, Horikoshi S, Ushijima H, Kimura J, Tomino Y. Effect of steroid-liposome on immunohistopathology of IgA nephropathy in ddY mice. Nephron. 2001;89(2):194–200. doi: 10.1159/000046067 [DOI] [PubMed] [Google Scholar]
  • 51.Wu Q Wang J Wang Y, et al. Targeted delivery of celastrol to glomerular endothelium and podocytes for chronic kidney disease treatment. Nano Res. 2022;15(4):3556–3568. doi: 10.1007/s12274-021-3894-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Yuan Z-X Jia L Lim LY, et al. Renal-targeted delivery of triptolide by entrapment in pegylated TRX-20-modified liposomes. Int J Nanomedicine. 2017;12:5673–5686. doi: 10.2147/IJN.S141095 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Wang Y Wu Q Wang J, 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 [DOI] [PubMed] [Google Scholar]
  • 54.Tripathy N, Wang J, Tung M, Conway C, Chung EJ. Transdermal delivery of kidney-targeting nanoparticles using dissolvable microneedles. Cell Mol Bioeng. 2020;13(5):475–486. doi: 10.1007/s12195-020-00622-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wang J Poon C Chin D, et al. Design and in vivo characterization of kidney-targeting multimodal micelles for renal drug delivery. Nano Res. 2018;11(10):5584–5595. doi: 10.1007/s12274-018-2100-2 [DOI] [Google Scholar]
  • 56.Choi HS Mathew AP Uthaman S, et al. Inflammation-sensing catalase-mimicking nanozymes alleviate acute kidney injury via reversing local oxidative stress. J Nanobiotechnol. 2022;20(1):205. doi: 10.1186/s12951-022-01410-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.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 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Zuckerman JE, Choi CHJ, Han H, Davis ME. Polycation-siRNA nanoparticles can disassemble at the kidney glomerular basement membrane. Proc Natl Acad Sci U S A. 2012;109(8):3137–3142. doi: 10.1073/pnas.1200718109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Sun H Shi K Zuo B, et al. Kidney-targeted drug delivery system based on metformin-grafted chitosan for renal fibrosis therapy. Mol Pharm. 2022;19(9):3075–3084. doi: 10.1021/acs.molpharmaceut.1c00827 [DOI] [PubMed] [Google Scholar]
  • 60.Gao S Hein S Dagnæs-Hansen F, et al. Megalin-Mediated specific uptake of chitosan/siRNA nanoparticles in mouse kidney proximal tubule epithelial cells enables AQP1 gene silencing. Theranostics. 2014;4(10):1039–1051. doi: 10.7150/thno.7866 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Yang C Nilsson L Cheema MU, et al. Chitosan/siRNA nanoparticles targeting cyclooxygenase type 2 attenuate unilateral ureteral obstruction-induced kidney injury in mice. Theranostics. 2015;5(2):110–123. doi: 10.7150/thno.9717 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Li S, Zeng YC, Peng K, Liu C, Zhang ZR, Zhang L. Design and evaluation of glomerulus mesangium-targeted PEG-PLGA nanoparticles loaded with dexamethasone acetate. Acta Pharmacol Sinica. 2019;40(1):143–150. doi: 10.1038/s41401-018-0052-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.He S Chen C Li F, et al. A polymeric nanosponge as a broad-spectrum reactive oxygen species scavenger for acute kidney injury treatment. Nano Lett. 2023;23(19):8978–8987. doi: 10.1021/acs.nanolett.3c02531 [DOI] [PubMed] [Google Scholar]
  • 64.Li J, Duan Q, Wei X, Wu J, Yang Q. Kidney-targeted nanoparticles loaded with the natural antioxidant rosmarinic acid for acute kidney injury treatment. Small. 2022;18(48):2204388. doi: 10.1002/smll.202204388 [DOI] [PubMed] [Google Scholar]
  • 65.Chen D, Han S, Zhu Y, Hu F, Wei Y, Wang G. Kidney-targeted drug delivery via rhein-loaded polyethyleneglycol-co-polycaprolactone-co-polyethylenimine nanoparticles for diabetic nephropathy therapy. Int J Nanomedicine. 2018;13:3507–3527. doi: 10.2147/IJN.S166445 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Lan T Guo H Lu X, et al. Dual-responsive curcumin-loaded nanoparticles for the treatment of cisplatin-induced acute kidney injury. Biomacromolecules. 2022;23(12):5253–5266. doi: 10.1021/acs.biomac.2c01083 [DOI] [PubMed] [Google Scholar]
  • 67.Wei H Jiang D Yu B, et al. Nanostructured polyvinylpyrrolidone-curcumin conjugates allowed for kidney-targeted treatment of cisplatin induced acute kidney injury. Bioact Mater. 2023;19:282–291. doi: 10.1016/j.bioactmat.2022.04.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Wang Y Pu M Yan J, et al. 1,2-Bis(2-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid acetoxymethyl ester loaded reactive oxygen species responsive hyaluronic acid–bilirubin nanoparticles for acute kidney injury therapy via alleviating calcium overload mediated endoplasmic reticulum stress. ACS Nano. 2023;17(1):472–491. doi: 10.1021/acsnano.2c08982 [DOI] [PubMed] [Google Scholar]
  • 69.Li Y Wang G Wang T, et al. PEGylated gambogic acid nanoparticles enable efficient renal-targeted treatment of acute kidney injury. Nano Lett. 2023;23(12):5641–5647. doi: 10.1021/acs.nanolett.3c01235 [DOI] [PubMed] [Google Scholar]
  • 70.Tang W Chen Y Jang HS, et al. Preferential siRNA delivery to injured kidneys for combination treatment of acute kidney injury. J Control Release. 2022;341:300–313. doi: 10.1016/j.jconrel.2021.11.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Tang W Panja S Jogdeo CM, et al. Modified chitosan for effective renal delivery of siRNA to treat acute kidney injury. Biomaterials. 2022;285:121562. doi: 10.1016/j.biomaterials.2022.121562 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Matsuura S Katsumi H Suzuki H, et al. l-Serine–modified polyamidoamine dendrimer as a highly potent renal targeting drug carrier. Proc Natl Acad Sci U S A. 2018;115(41):10511–10516. doi: 10.1073/pnas.1808168115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Mathew AP, Uthaman S, Bae EH, Lee JY, Park IK. Vimentin targeted nano-gene carrier for treatment of renal diseases. J Korean Med Sci. 2021;36(49):e333. doi: 10.3346/jkms.2021.36.e333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Huang C Zeng T Li J, et al. Folate receptor-mediated renal-targeting nanoplatform for the specific delivery of triptolide to treat renal ischemia/reperfusion injury. ACS Biomater Sci Eng. 2019;5(6):2877–2886. doi: 10.1021/acsbiomaterials.9b00119 [DOI] [PubMed] [Google Scholar]
  • 75.Qin S, Wu B, Gong T, Zhang ZR, Fu Y. Targeted delivery via albumin corona nanocomplex to renal tubules to alleviate acute kidney injury. J Control Release. 2022;349:401–412. doi: 10.1016/j.jconrel.2022.07.013 [DOI] [PubMed] [Google Scholar]
  • 76.Yu H Lin T Chen W, et al. Size and temporal-dependent efficacy of oltipraz-loaded PLGA nanoparticles for treatment of acute kidney injury and fibrosis. Biomaterials. 2019;219:119368. doi: 10.1016/j.biomaterials.2019.119368 [DOI] [PubMed] [Google Scholar]
  • 77.Guo L Luo S Du Z, et al. Targeted delivery of celastrol to mesangial cells is effective against mesangioproliferative glomerulonephritis. Nat Commun. 2017;8(1):878. doi: 10.1038/s41467-017-00834-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Huang H Liu Q Zhang T, et al. Farnesylthiosalicylic acid-loaded albumin nanoparticle alleviates renal fibrosis by inhibiting Ras/Raf1/p38 signaling pathway. Int J Nanomedicine. 2021;16:6441–6453. doi: 10.2147/IJN.S318124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Jiang D Ge Z Im HJ, et al. DNA origami nanostructures can exhibit preferential renal uptake and alleviate acute kidney injury. Nat Biomed Eng. 2018;2(11):865–877. doi: 10.1038/s41551-018-0317-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Thai HBD Kim KR Hong KT, et al. Kidney-targeted cytosolic delivery of siRNA using a small-sized mirror DNA tetrahedron for enhanced potency. ACS Cent Sci. 2020;6(12):2250–2258. doi: 10.1021/acscentsci.0c00763 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Yan R, Cui W, Ma W, Li J, Liu Z, Lin Y. Typhaneoside-tetrahedral framework nucleic acids system: mitochondrial recovery and antioxidation for acute kidney injury treatment. ACS Nano. 2023;17(9):8767–8781. doi: 10.1021/acsnano.3c02102 [DOI] [PubMed] [Google Scholar]
  • 82.Campeiro JDA, Dam WA, Hayashi MAF, van den Born J. Crotamine/siRNA nanocomplexes for functional downregulation of syndecan-1 in renal proximal tubular epithelial cells. Pharmaceutics. 2023;15(6):1576. doi: 10.3390/pharmaceutics15061576 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Katsumi H Kitada S Yasuoka S, et al. L-Serine-Modified poly-L-lysine as a biodegradable kidney-targeted drug carrier for the efficient radionuclide therapy of renal cell carcinoma. Pharmaceutics. 2022;14(9):1946. doi: 10.3390/pharmaceutics14091946 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Uchida M Maier B Waghwani HK, et al. The archaeal Dps nanocage targets kidney proximal tubules via glomerular filtration. J Clin Invest. 2019;129(9):3941–3951. doi: 10.1172/JCI127511 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wei J Grabau R Zhang J, et al. Development of a glomerular specific targeted nanoparticles for lupus nephritis. FASEB J. 2019;33(S1):749.3. doi: 10.1096/fasebj.2019.33.1_supplement.749.3 [DOI] [Google Scholar]
  • 86.Zhu Z Liu X Li P, et al. Renal clearable quantum dot–drug conjugates modulate labile iron species and scavenge free radicals for attenuating chemotherapeutic drug-induced acute kidney injury. ACS Appl Mater Inter. 2023;15(18):21854–21865. doi: 10.1021/acsami.3c00714 [DOI] [PubMed] [Google Scholar]
  • 87.Wang S Chen Y Han S, et al. Selenium nanoparticles alleviate ischemia reperfusion injury-induced acute kidney injury by modulating GPx-1/NLRP3/Caspase-1 pathway. Theranostics. 2022;12(8):3882–3895. doi: 10.7150/thno.70830 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Duan R Li Y Zhang R, et al. Reversing acute kidney injury through coordinated interplay of anti‐inflammation and iron supplementation. Adv Mater. 2023;35(28):2301283. doi: 10.1002/adma.202301283 [DOI] [PubMed] [Google Scholar]
  • 89.Peres RA Silva-Aguiar RP Teixeira DE, et al. Gold nanoparticles reduce tubule-interstitial injury and proteinuria in a murine model of subclinical acute kidney injury. Biochim Biophys Acta Gen Subj. 2023;1867(4):130314. doi: 10.1016/j.bbagen.2023.130314 [DOI] [PubMed] [Google Scholar]
  • 90.Choi CHJ, Zuckerman JE, Webster P, Davis ME. Targeting kidney mesangium by nanoparticles of defined size. Proc Natl Acad Sci U S A. 2011;108(16):6656–6661. doi: 10.1073/pnas.1103573108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Chen Z Qi F Qiu W, et al. Hydrogenated germanene nanosheets as an antioxidative defense agent for acute kidney injury treatment. Adv Sci. 2022;9(33):2202933. doi: 10.1002/advs.202202933 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Alidori S Akhavein N Thorek DLJ, et al. Targeted fibrillar nanocarbon RNAi treatment of acute kidney injury. Sci Transl Med. 2016;8(331):331ra39. doi: 10.1126/scitranslmed.aac9647 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Yan J, Wang Y, Zhang J, Liu X, Yu L, He Z. Rapidly blocking the calcium overload/ROS production feedback loop to alleviate acute kidney injury via microenvironment-responsive BAPTA-AM/BAC Co‐delivery nanosystem. Small. 2023;19(17):2206936. doi: 10.1002/smll.202206936 [DOI] [PubMed] [Google Scholar]
  • 94.Huang Z, Chun C, Li X. Kidney targeting peptide-modified biomimetic nanoplatforms for treatment of acute kidney injury. J Control Release. 2023;358:368–381. doi: 10.1016/j.jconrel.2023.04.042 [DOI] [PubMed] [Google Scholar]
  • 95.Zuckerman JE, Gale A, Wu P, Ma R, Davis ME. 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 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Alidori S Thorek DLJ Beattie BJ, et al. Carbon nanotubes exhibit fibrillar pharmacology in primates. PLoS One. 2017;12(8):e0183902. doi: 10.1371/journal.pone.0183902 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Williams RM Shah J Ng BD, et al. Mesoscale nanoparticles selectively target the renal proximal tubule epithelium. Nano Lett. 2015;15(4):2358–2364. doi: 10.1021/nl504610d [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Han SJ Williams RM D’Agati V, et al. Selective nanoparticle-mediated targeting of renal tubular Toll-like receptor 9 attenuates ischemic acute kidney injury. Kidney Int. 2020;98(1):76–87. doi: 10.1016/j.kint.2020.01.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Veiras LC Bernstein EA Cao D, et al. Tubular IL-1β induces salt sensitivity in diabetes by activating renal macrophages. Circ Res. 2022;131(1):59–73. doi: 10.1161/CIRCRESAHA.121.320239 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Williams RM Shah J Mercer E, et al. Kidney-targeted redox scavenger therapy prevents cisplatin-induced acute kidney injury. Front Pharmacol. 2021;12:790913. doi: 10.3389/fphar.2021.790913 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Williams RM Shah J Tian HS, et al. Selective nanoparticle targeting of the renal tubules. Hypertension. 2018;71(1):87–94. doi: 10.1161/HYPERTENSIONAHA.117.09843 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Deng X Zeng T Li J, et al. Kidney-targeted triptolide-encapsulated mesoscale nanoparticles for high-efficiency treatment of kidney injury. Biomater Sci. 2019;7(12):5312–5323. doi: 10.1039/C9BM01290G [DOI] [PubMed] [Google Scholar]
  • 103.Han SJ Williams RM Kim M, et al. Renal proximal tubular NEMO plays a critical role in ischemic acute kidney injury. JCI Insight. 2020;5(19):e139246. doi: 10.1172/jci.insight.139246 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Tan L Deng X Lai X, et al. Mesoscale nanoparticles encapsulated with emodin for targeting antifibrosis in animal models. Open Chem. 2020;18(1):1207–1216. doi: 10.1515/chem-2020-0163 [DOI] [Google Scholar]
  • 105.Mrug M Heller DA Kapadia CH, et al. Efficacy and adverse effects of a novel mesoscale nanoparticle-guided sirolimus delivery strategy in a Pkhd1PCK rat model. J Am Soc Nephrol. 2021;32:298. https://www.asn-online.org/education/kidneyweek/2021/program-abstract.aspx?controlId=3606981 [Google Scholar]
  • 106.Vallorz EL Blohm-Mangone K Schnellmann RG, et al. Formoterol PLGA-PEG nanoparticles induce mitochondrial biogenesis in renal proximal tubules. AAPS J. 2021;23(4):88. doi: 10.1208/s12248-021-00619-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Vallorz EL Encinas-Basurto D Schnellmann RG, et al. Design, development, physicochemical characterization, and in vitro drug release of formoterol PEGylated PLGA polymeric nanoparticles. Pharmaceutics. 2022;14(3):638. doi: 10.3390/pharmaceutics14030638 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Vallorz EL Janda J Mansour HM Schnellmann RG, et al. Kidney targeting of formoterol containing polymeric nanoparticles improves recovery from ischemia reperfusion-induced acute kidney injury in mice. Kidney Int. 2022;102(5):1073–1089. doi: 10.1016/j.kint.2022.05.032 [DOI] [PubMed] [Google Scholar]
  • 109.Williams RM Shah J Mercer E, et al. Edaravone-loaded mesoscale nanoparticles treat cisplatin-induced acute kidney injury. BioRxiv. 2020. doi: 10.1101/2020.01.24.919134 [DOI] [Google Scholar]
  • 110.Guo X Xu L Velazquez H, et al. Kidney-targeted renalase agonist prevents cisplatin-induced chronic kidney disease by inhibiting regulated necrosis and inflammation. J Am Soc Nephrol. 2022;33(2):342–356. doi: 10.1681/ASN.2021040439 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Benson LN Liu Y Wang X, et al. The IFNγ-PDL1 pathway enhances CD8T-DCT interaction to promote hypertension. Circ Res. 2022;130(10):1550–1564. doi: 10.1161/CIRCRESAHA.121.320373 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Aydin E, Cebeci A, Lekesizcan A. Prevention of cisplatin-induced nephrotoxicity by kidney-targeted siRNA delivery. Int J Pharm. 2022;628:122268.doi: 10.1016/j.ijpharm.2022.122268 [DOI] [PubMed] [Google Scholar]
  • 113.Skelton R Roach A Prudhomme LE, et al. Formulation of lipid-free polymeric mesoscale nanoparticles encapsulating mRNA. Pharmaceut Res. 2022;39(11):1–9. doi: 10.1007/s11095-022-03398-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Williams RM Chen S Langenbacher RE, et al. Harnessing nanotechnology to expand the toolbox of chemical biology. Nat Chem Biol. 2021;17(2):129–137. doi: 10.1038/s41589-020-00690-6 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Kidney360 are provided here courtesy of American Society of Nephrology

RESOURCES