Next Article in Journal
Comparative Assessment and High-Throughput Drug-Combination Profiling of TEAD-Palmitoylation Inhibitors in Hippo Pathway Deficient Mesothelioma
Previous Article in Journal
Biopsychosocial Variables in Male Schizophrenic Patients: A Comprehensive Comparison with Healthy Controls
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

RNA Combined with Nanoformulation to Advance Therapeutic Technologies

by
Eduarda Santos Lima
,
Déborah dos Santos
,
Atena Liriel Souza
,
Maria Eduarda Macedo
,
Mariana Evangelista Bandeira
,
Sérgio Santos Silva Junior
,
Bianca Sampaio Dotto Fiuza
,
Vinicius Pinto Costa Rocha
,
Larissa Moraes dos Santos Fonseca
,
Danielle Devequi Gomes Nunes
,
Katharine Valéria Saraiva Hodel
and
Bruna Aparecida Souza Machado
*
SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC (Integrated Manufacturing and Technology Campus), Salvador 41650-010, Brazil
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(12), 1634; https://doi.org/10.3390/ph16121634
Submission received: 4 October 2023 / Revised: 10 November 2023 / Accepted: 16 November 2023 / Published: 21 November 2023
(This article belongs to the Section Biopharmaceuticals)

Abstract

:
Nucleic acid-based therapies have the potential to address numerous diseases that pose significant challenges to more traditional methods. RNA-based therapies have emerged as a promising avenue, utilizing nanoformulation treatments to target a range of pathologies. Nanoformulation offers several advantages compared to other treatment modalities, including targeted delivery, low toxicity, and bioactivity suitable for drug loading. At present, various types of nanoformulations are available, such as liposomes, polymeric nanoparticles (NPs), magnetic NPs, nanoshells, and solid lipid nanoparticles (SLNs). RNA-based therapy utilizes intracellular gene nanoparticles with messenger RNA (mRNA) emerging prominently in cancer therapy and immunotechnology against infectious diseases. The approval of mRNA-based technology opens doors for future technological advancements, particularly self-amplifying replicon RNA (repRNA). RepRNA is a novel platform in gene therapy, comprising viral RNA with a unique molecular property that enables the amplification of all encoded genetic information countless times. As a result, repRNA-based therapies have achieved significant levels of gene expression. In this context, the primary objective of this study is to furnish a comprehensive review of repRNA and its applications in nanoformulation treatments, with a specific focus on encapsulated nanoparticles. The overarching goal is to provide an extensive overview of the use of repRNA in conjunction with nanoformulations across a range of treatments and therapies.

1. Introduction

While drug development has made significant progress over the years, there remains a pressing need for further advancements in this field [1]. Technological innovations and genomic research have opened numerous treatment possibilities [2]. Moreover, nucleic acid-based therapies hold the potential to address or even cure many diseases that have proven resistant to traditional treatment methods [2]. The discovery of small interfering RNA (siRNA) and antisense RNA, which effectively suppress the expression of specific genes in cells affected by viruses or cancer, has sparked increased interest in RNA treatments [3]. These advances encompass a range of mechanisms, including approaches such as inhibiting messenger RNA (mRNA) translation, gene expression interference via siRNA, self-amplifying replicon RNA (repRNA), catalytically active ribozymes, and protein-binding RNA molecules. Consequently, RNA-based therapies find applications in diagnostics and certain treatment modalities [4].
Synthetic mRNA serves as a versatile tool for producing proteins and peptides, finding extensive use in pharmaceutical applications, including cancer immunotherapy [5]. It is generated in a cell-free system through in vitro transcription (IVT) from a DNA template [6]. Acting as a natural ligand for various receptors, exogenous mRNA stimulates the release of type I interferon and pro-inflammatory cytokines, providing it with inherent adjuvanticity [5] On the other hand, repRNA, with sizes ranging from 12 to 15 kilobases, stem from viral genomes lacking at least one structural protein gene, enabling sustained antigen production without the risk of producing infectious progeny [7]. However, their susceptibility to RNase and inefficient uptake by dendritic cells represent key limitations. Compared to conventional mRNA, repRNA triggers prolonged gene expression lasting weeks to months after a single injection, making it a promising candidate for sustained antigen expression in vaccine design [8]. repRNA vaccines have emerged as promising candidates, showcasing exemplary safety and immunogenicity, and have already demonstrated encouraging results in various clinical trials [9,10].
Regarding siRNA, its mechanism relies on post-transcriptional gene silencing, known for its specificity in targeting disease-related genes [11]. However, siRNAs face challenges such as low cellular uptake and susceptibility to degradation, necessitating protective carriers for efficient delivery into target cells [12,13]. Various delivery systems, including nanocarriers, aptamers, peptides, sugars, proteins, and antibodies, have been developed to address these limitations [12,13]. In mammalian cells, siRNAs play a crucial role in RNA interference (RNAi), with minimal concentrations as low as 2000 siRNAs per cell being sufficient for effective gene knockdown [14]. Compared to DNA, RNA has several distinctive qualities that make it a potent biomaterial. Notably, RNA is capable of folding into several tertiary structures with unique activities [3]. The effectiveness of RNA therapy is significantly enhanced by its synergistic association with encapsulated nanoparticles, which facilitate targeted and controlled delivery, thereby increasing its therapeutic potential.
RNA nanotechnology offers several advantages for therapeutic applications. These advantages encompass passive targeting due to its nano-scale size and branched structure, precise synthesis control, limited nonspecific cell membrane crossing, high water solubility, and reduced immunogenicity. Additionally, RNA nanotechnology provides multivalency for module conjugation, favorable in vivo profiles, and specific delivery potential [15]. Furthermore, RNA’s classification as a chemical reagent implies more favorable regulatory processes compared to protein-based clinical reagents.
It is important to note that lipid-encapsulated nanoparticles containing small interfering double-stranded RNA (siRNA) have been employed to target transthyretin (TTR) mRNA, leading to the degradation of TTR deposits in patients with TTR-mediated hereditary amyloidosis [9]. This RNA interference drug, Patisiran (ONPATTRO™), has received approval in both the US and Europe for single intravenous infusion [16]. In recent years, mRNAs generated through IVT have gained prominence as promising candidates for the development of new drugs and vaccines [1,16].
Despite considerable progress in RNA nanotechnology for medical applications, several challenges persist. These include RNA’s chemical instability, the need for thermodynamic stability in RNA nanoparticles, construction costs, the requirement for favorable pharmacological profiles, specific cell targeting, and the prevention of degradation in the endocytic pathway [3]. Overcoming these challenges is crucial for realizing the full potential of RNA nanotechnology in therapeutics.
Consequently, intensive research efforts are currently underway in the fields of cancer immunotherapy, vaccine development for infectious diseases, gene editing, and protein replacement, with the goal of advancing our understanding in this rapidly evolving field [9] In light of these developments, this study seeks to provide a comprehensive review of RNA delivery systems, focusing particularly on encapsulated nanoparticles. The primary objective is to examine the significant levels of gene expression achieved through repRNA-based therapies within the broader context of nanoformulation treatments. The overarching goal is to offer an extensive overview of repRNA’s utilization in conjunction with nanoformulations, encompassing a range of treatments and therapies.

2. Nanotechnology’s Impact on Healthcare: Advancements, Applications, and RNA Nanotechnology

The prefix “nano”, derived from Greek, signifies something minuscule, measuring as small as less than one-millionth of a meter [17]. This minuscule scale is fundamental to nanotechnology’s core concept. Conversely, technology denotes the systematic study of processes within a specific domain [18]. However, nanotechnology takes this a step further. Following this initial definition, nanotechnology is regarded as a scientific discipline dedicated to comprehending matter at nanometric scales, typically ranging from 1 to 100 nanometers, with the aim of precise control and manipulation to usher in technological advancements across diverse domains [19].
Our civilization has greatly benefited from nanotechnology, which has made significant contributions to numerous prominent industries [20]. Among these industries, over the past two decades, nanotechnology’s advancement in the medical and healthcare sectors have played a crucial role in preventing, diagnosing, and treating various illnesses [20].
Within the realm of healthcare, the application of nanotechnology is termed “nanomedicine,” a field that has yielded a profound understanding of cellular mechanisms in living organisms [21]. This profound understanding has paved the way for various applications encompassing pharmaceutical therapy and the prevention, diagnosis, monitoring, and treatment of many diseases [22].
The implications of nanotechnology in scientific research are highly promising, transcending the limitations of conventional therapies by enhancing the mechanisms employed, particularly in safeguarding the desired drug [21]. To achieve these advancements, nanotechnology employs top-down and bottom-up assembly methods to create and utilize materials at the nanometer scale. Notably, DNA, RNA, and protein macromolecules possess inherent properties at the nanoscale, making them promising building blocks for the bottom-up construction of nanostructures and nanodevices [15].
Research into the folding and structure of RNA has a long history. However, it is important to note that RNA nanotechnology distinguishes itself from conventional RNA structure and folding studies [23]. RNA nanotechnology is the study of RNA-based structures with a primary framework at the nanoscale [24]. This unique focus necessitates a comprehensive understanding of not only intramolecular interactions and folding but also intermolecular interactions. A crucial aspect of RNA nanotechnology involves characterizing the physical, chemical, biological, and pharmacological properties of nanoparticles that researchers can homogenize through purification [23]. An important milestone in the field was the assembly of RNA dimer, trimer, and hexamer nanoparticles using reengineered RNA fragments derived from pRNA (packaging RNA), a vital component responsible for driving the DNA packaging motor in bacteriophage phi29 [25].
RNA nanotechnology, as opposed to conventional RNA biology research, focuses on using the characteristics of RNA to create structures for use in nanomedicine [24]. This approach is particularly promising for therapeutic applications, where nanoformulations offer synergistic actions and efficient delivery systems, notably in the treatment of diverse cancer types [26]. Beyond medicinal technology, a range of industrial sectors, including electronics, environmental science, food production, and textiles, have expressed keen interest in nanotechnology due to its potential to enhance product quality, safety, and durability [22].

2.1. Nanoformulation

Researchers engineer nanoformulations to optimize drug delivery, increase drug solubility, improve bioavailability, and enable targeted drug release, thereby enhancing the overall efficacy and safety of pharmaceuticals [26]. In terms of therapeutic effectiveness, researchers have observed that nanoparticles exhibit optimal internalization by cancer cells, especially in the treatment of diseases like breast cancer [27]. Their enhanced permeation into the target cells not only improves drug delivery, but also renders the nanoformulation more cytotoxic when compared to free drugs [28].
These nanodelivery technologies can also apply to phytocompounds. Various phytocompounds, including cannabidiol (CBD), have received attention for delivery using nanocarriers as a viable platform, with the aim of restricting the variety of negative consequences [29]. Nanodelivery technologies have improved the stability of phytocompounds, enhanced their absorption, shielded them from early enzymatic depletion or metabolism within the body, and prolonged their circulation duration [29].
Therefore, the significance of this technology lies in advancing the treatment of various diseases and offering expanded therapeutic possibilities [30]. Furthermore, its economic advantages are compelling, as it allows for easy industrial scale-up, making it a practical choice in this sector. These innovative nanoformulations hold the potential to revolutionize drug delivery and substantially improve patient outcomes across a wide range of medical conditions. With this in mind, Table 1 shows some nanoformulations and their main components that have already been approved for therapeutic use by regulatory agencies such as the FDA.

2.1.1. Liposomes

Nanoparticles serve as drug carriers, and one of the key applications of this nanotechnology is in disease therapy [31]. An excellent example is liposomes, which, as the name suggests, are lipid-based nanoparticles that are usually 50–500 nanometers in diameter size [32,33]. They consist of phospholipid and cholesterol bilayers, with their number varying depending on the classification and determined by their size and the number of lamellae [34].
Liposome vesicles are spherical and spontaneously form in aqueous media, offering versatility in terms of the drug and the route of administration [35]. The administered drug may be either polar (water-soluble), concentrating in the internal aqueous phase, or apolar (fat-soluble), containing compounds with this characteristic [36]. The variation in bilayers is crucial for their function, as these structures are essential for encapsulating drugs with different sizes and solubilities [37]. Furthermore, the choice of a specific liposome classification influences its targeting specificity towards organs, cells, and tissues [38].
The use of liposomes offers numerous advantages. Liposomes, with their unique properties, serve as flexible and adaptable nanocarriers for drug delivery [39]. Their amphiphilic nature enables the encapsulation of both hydrophilic and hydrophobic medications, enhancing their versatility, providing excellent biocompatibility and biodegradability, along with the isolation of the drug from the surrounding medium [39,40]. By preventing direct contact with healthy cells, liposomes can reduce the toxicity of chemotherapeutic drugs, while also enhancing the stability and bioavailability of these compounds [39]. Drug concentrations at sites of action are seven to nine times greater when employing drug-loaded liposomes as delivery vehicles than when using medicines alone without carriers [41]. However, there are still disadvantages to consider, such as their relatively large particle size and rapid clearance by the immune system [42].
Nevertheless, the applications of liposomes are extensive. They find use in protecting drugs from degradation in vivo, directing drug delivery to the desired site, controlling drug release, modifying and refining biodistribution, and increasing bioavailability and solubility [43]. As the first nano-delivery system, liposomes have achieved considerable clinical success, with well-consolidated and established pharmacokinetics and delivery pathways [16]. Consequently, liposomes have demonstrated high compatibility with RNA for complexed use in cancer immunotherapies [44]. These adaptable lipid-based nanoparticles continue to be a foundation in advancing drug delivery technologies.
While liposomal products approved for drug delivery have been predominantly parenteral, research into gene therapy has yielded liposomal formulations for restoring therapeutic function, demonstrating promising in vivo data [45]. Liposome-mediated gene delivery’s clinical efficacy hinges on the availability of clinically relevant vectors and robust, efficient methods for their delivery to the target cellular site [46,47]. Achieving an optimal balance between the molecular interactions of hydrophilicity and hydrophobicity is essential for effectively transferring genes to the target cells using liposome vectors [45,48]. Local gene delivery at sites of tumor metastases is anticipated to mitigate systemic side effects [49].
Conventional liposomes are an insufficient transfection technology because encapsulating DNA into them presents technical difficulties because of the size of the plasmid [50]. As an alternative, a technology based on cationic lipids and PE was developed in the late 1980s to neutralize the negative charge of plasmids, facilitating their more efficient capture and delivery into cells [47].
Because DNA is polyanionic, cationic lipids are usually used for gene delivery, whereas anionic liposomes are mostly used for other therapeutic macromolecule deliveries [51]. When cationic lipid solution is combined with neutral helper lipid solution, DNA may create a positively charged compound called a lipoplex [51,52]. Since it makes it relatively easy to entrap DNA or RNA in a liposome and facilitates efficient gene delivery to cells, DOTMA—one of the first synthesized and commercially available cationic lipids for gene delivery—has greatly increased the potential of nonviral agents for gene therapy [47,51,53]
Liposomes serve as versatile and adaptable nanocarriers for drug delivery, offering benefits such as enhanced biocompatibility and drug isolation. Despite their successes in cancer immunotherapies, challenges like large particle size and rapid immune system clearance persist. Further research is needed to optimize liposomal formulations and explore their integration with emerging technologies like repRNA for groundbreaking advancements in drug delivery and therapeutic applications.

2.1.2. Polymeric Nanoparticle

Polymeric nanoparticles (PNPs) are compounds trapped or adsorbed onto the surface of a polymeric core, and they exhibit variations in characteristics such as concentration, composition, size, shape, and surface properties [54]. The polymeric mass, also known as the nanosphere, entraps the drug within its core or adsorbs it onto its surface [55]. The study and application of this compound are continually expanding, with the most prevalent use being in gene therapy to combat current disorders [56].
The significant potential of this drug delivery type lies in the versatility of the polymers used, making them well-suited for meeting the requirements of various drug delivery systems [57]. Among their advantages, the use of PNPs helps target specific desired sites of action, reducing systemic concentrations and minimizing side effects [57]. Thus, the development of vaccines, such as those for SARS-COV-2, using PNPs in conjunction with self-amplifying RNA, stands as a promising example applied in recent years [58].
Two types of polymers, natural or synthetic, can formulate polymeric nanoparticles based on the therapy’s objectives, administration route, or polymer nature [59]. Natural polymers commonly selected include sodium alginate, albumin, chitosan, and gelatin, while synthetic options encompass polylactides, poly lactide co-glycolides (PLGA), polyglycosides (PGA), polyanhydrides, poly orthoesters (POE), and polycaprolactone (PCL) [55].
Polymer-based nanosystems comprise a range of structures, such as hydrogels, polymer micelles, polymerosomes, and polyplexes—which are complexes of cationic polymers and nucleic acids [60,61]. Polyplexes are polymeric systems in which the negatively charged nucleic acids and the cationic groups of the polymer interact electrostatically to include the condensed and/or complexed gene or siRNA [62,63]. The synthesis of nanoparticles with the nucleic acid payload in a compressed and protected state is the outcome of polyplex formation, which is dependent on the entropy-driven ionic interaction between the polyanionic nucleic acid and the multivalent cationic polymers [61].
However, it is important to note that polymeric nanoparticles offer several significant benefits, including the ability to provide controlled release to the target region, stabilize labile molecules (such as proteins), and allow surface modification with ligands for discreet and targeted medication administration [64]. These advantages make them an adjustable and promising option in the field of drug delivery.
Nonetheless, the limitations of polymeric nanoparticles include potential toxic degradation, the aggregation of toxic monomers, residual material attachment, and the possibility of a toxic degradation process within such systems [65]. It is worth noting that using nanoparticles for drug delivery may carry risks due to the increased reactivity and potential toxicity associated with smaller particle sizes [66]. However, various factors, including nanoparticle shape, chemical composition, hydrophobicity/hydrophilicity, and surface charge, may also influence these behaviors [66]. As such, careful consideration and research are essential to harness the full potential of polymeric nanoparticles while mitigating associated risks.
Polymeric nanoparticles offer diverse drug delivery options, targeting specific sites and minimizing side effects. While advantageous in controlled release and stability of labile molecules, they pose risks such as toxic degradation and potential toxicity linked to their small size. Careful research is vital to harness their potential and mitigate associated risks.

2.1.3. Magnetic Nanoparticle

This type of nanoparticle (NP) has garnered extensive research attention and investigation, resulting in its expanded applications [67]. Magnetic nanoparticles (MNPs) have received considerable recognition, primarily for their role in magnetic resonance therapy and magnetic hyperthermia. They offer advantages such as low production costs, high physicochemical stability, acceptable biocompatibility, and biodegradability [68].
Particles possessing both mass and electric charges exhibit magnetic phenomena. This group of particles includes protons, electrons, and holes, as well as positive and negative ions [69]. The functional principle of magnetic nanoparticles centers on their response to magnetic field variations over time, leading to energy transfer from the magnetic field to these nanoparticles [70].
Nanoparticles acquire magnetic properties in the presence of an external magnet; however, they revert to their nonmagnetic state once the external magnet is removed [69]. This characteristic prevents the particles from exhibiting magnetic behavior when no external field is applied. This phenomenon induces significant heating and efficient heat transfer, making them suitable for use as chemotherapeutic or radiotherapeutic agents for cancer cell destruction [71]. Moreover, this alternative has proven to be effective without the side effects associated with conventional cancer treatments [72].
Beyond their role in tumor catabolism via hyperthermia, magnetic nanoparticles find applications in various fields [73]. In biomedicine, they facilitate drug or radioisotope transport, serve as magnetic separators for labeled cells, and support various diagnostic mechanisms [74]. This technology can also play a crucial role in sensitive clinical diagnostics for viral RNA, such as SARS-CoV-2, achieved through the synthesis of aminoesters with carboxyl group-coated magnetic nanoparticles and the development of viral RNA extraction methods [75].
However, it is important to note that magnetic nanoparticles may have limitations in certain areas, like the environmental industry, due to their high cost and complex operational modifications [76].
The application of an external magnetic field organizes MNPs into a two-dimensional space, imposing a limitation on their concentration within a three-dimensional region for drug administration [77]. Furthermore, removing the external magnetic field makes it challenging to retain the magnetic particles within the targeted organ. Patients cannot experience continuous exposure to an external magnetic field, thereby limiting treatment efficacy, which depends on the frequency, strength, and duration of magnetic field exposure [77]. Despite these challenges, magnetic nanoparticles remain a promising technology with diverse applications, especially in the field of biomedicine.
Magnetic nanoparticles have garnered extensive research attention for their expanded applications in magnetic resonance therapy and magnetic hyperthermia. While they offer advantages such as biocompatibility and stability, challenges remain regarding complex operational modifications and the retention of magnetic properties post removal of the external magnetic field. Despite these challenges, MNPs continue to hold promise for significant advancements in the field of biomedicine, necessitating further research and the enhancement of their applications.

2.1.4. Inorganic Nanoparticles

Nanoshells, also referred to as core shells, consist of cores made of one material encased by an outer shell or coating [78]. Nanoshells typically comprise a dielectric core surrounded by a thin metallic shell, often made of gold, with a thickness ranging from 1 to 20 nm [79]. They can be divided into two main types: oxide nanoshells, originating from oxide core shell particles, including hollow nanoshells, and metal nanoshells, formed using dielectric cores and metallic shells, like silver and gold nanoshells [80]. A common method for producing nanoshells involves using silica as the core material and applying a layer of another substance to attach gold particles to the outer surface, forming the shell [81].
According to Ahmadi and Arami (2014) nanoshells have demonstrated significant potential in the field of biomedicine due to their stability, biocompatibility, photoluminescence, tunability, safety, and bioavailability. These applications include gene delivery, drug delivery, targeted therapy, tissue welding, and cancer imaging, among others. However, nanoshells that are conjugated with various molecules, such as antibodies, oligonucleotides, fluorophores, targeting ligands, therapeutic agents, polymers, and radioisotopes, offer enhanced efficacy for diagnostic and therapeutic purposes [82].
The purpose of creating nanoshells is to modify them to absorb light at medically advantageous wavelengths [78]. Nanoshells represent a recent addition to the realm of nanotechnology applications, offering tunable optical properties and the capability to easily conjugate with biomolecules [83]. They offer advantages such as resistance to heat and chemical denaturation, facilitating interactions with medications [84].
They also provide a photothermal effect, facilitating a synergistic combination of chemo–photothermal therapy [85]. The ability to set optical resonance allows for the application of photothermal therapy at various wavelengths of interest, potentially enhancing the mRNA transcription of genes responsible for inducing mitochondrial biogenesis, including sirtuin 1 [86]. This capability has significant implications for fields like muscle tissue engineering, regenerative medicine, and bionics [86].
Researchers have used these nanoshell types to eliminate cancer cells in animal models. When injected into a tumor and exposed to radiation, nanoshells heat up sufficiently to destroy the tumor cells [78]. Supporting this, studies have shown that the use of nanoparticles results in higher cancer cell lethality compared to using infrared irradiation alone [87]. However, it is important to note that due to their high surface energy, metal nanoshells can be prone to deactivation, and may exhibit a degree of instability [88].
Nanoshells exhibit great potential in biomedicine due to their stability and tunability, enabling applications in gene and drug delivery, targeted therapy, and cancer imaging. However, their high surface energy may lead to deactivation and instability, necessitating further research for addressing these concerns and fully unlocking their potential in biomedical applications.

2.1.5. Solid Lipid Nanoparticles (SLNs)

Originally known as lipospheres, solid lipid nanoparticles (SLNs) have gained recognition as promising pharmaceutical carriers for controlled drug delivery since 1990, particularly for poorly soluble and bioavailable substances [89,90]. These nanoparticles consist of a lipid matrix along with surfactants and co-surfactants, essential components that provide both stability and ligand properties, resulting in aqueous colloidal dispersions [91]. Lipid nanoparticles (LNPs) consist of a lipid bilayer that encases the RNA, often including additional components such as polyethylene glycol (PEG) and targeting ligands [92]. These components play a crucial role in enhancing stability, extending circulation duration, and enabling targeted delivery to specific cells or tissues [92]. The particle size, which can vary from 10 to 1000 nm, is determined by the chosen preparation method [93].
SLNs exhibit remarkable versatility, as they can encapsulate a wide range of substances, including drugs, vitamins, chemicals, and various xenobiotics [94]. Moreover, they possess the unique capability to transport both anticancer drugs and genes [94]. Comprised mainly of mono-, di-, and triglycerides or fatty acids, these lipid matrices remain in the solid state at typical body temperatures [95].
The choice of lipids in SLN preparation significantly influences their capacity to transport genes, with various lipid substances, such as glyceryl behenate and behenic acid, having been employed as solid lipid matrices [91].
This use of solid lipids is particularly advantageous, reducing drug mobility within the lipid matrix and preventing particle coalescence, thereby enhancing stability [96]. Furthermore, the application of SLNs in RNA delivery has not adversely affected RNA functionality, showing promise for both drug and gene delivery [97].
Ionizable lipids are considered crucial components of LNP-based RNA therapeutics because they are positively charged at a low pH to enhance the encapsulation of negatively charged RNA, and the charge becomes less positive or almost neutral at physiological pH [98]. A non-bilayer structure is formed by the interaction of these positively charged ionizable lipids with the negatively charged inner endosome membrane [99]
As the demand for advanced disease therapies grows, SLNs with complex internal structures provide improved stability and offer advantages over conventional biopharmaceuticals and other nanoscale carriers [94,100]. These advantages include minimal biotoxicity due to the biocompatible and biodegradable nature of the lipids used [101]. They can be produced without the need for organic solvents and provide excellent structural stability [101,102]. SLNs are versatile, allowing for both drug targeting and controlled drug release, and the incorporation of active compounds enhances their stability [101,103]. Additionally, SLNs are capable of encapsulating lipophilic and hydrophilic drugs, making them adaptable to various therapeutic applications [101,104]. Furthermore, producing SLNs on a large scale is simple, and they are sterilizable [101,105].
Despite these numerous advantages, SLNs also come with potential limitations, including relatively low drug-loading capacity depending on the environment and the physicochemical composition of the active ingredients, especially hydrophilic molecules [101,106]. Additionally, the polymorphic nature of the medication may lead to drug leakage from the carrier, particularly during storage, when phase transitions are likely to occur [101,107]. Another concern is the relatively high-water content in SLN dispersions, which can range from 70% to 99.9% [101,108].
While the absence of SLNs in clinical applications may be attributed to a lack of rational cost–benefit analysis, they still hold promise for the pharmacokinetic profiling of encapsulated drugs [109].
SLNs have shown promise in controlled drug delivery due to their enhanced stability and biocompatibility, but their drawbacks include low drug-loading capacity for hydrophilic molecules, potential drug leakage during storage, and high water content in SLN dispersions. Further research is needed to address these limitations and fully integrate SLNs into clinical practice.

2.2. Nanotoxicology Challenges and Opportunities

Nanotoxicology, while an expanding field, still has significant gaps, with existing research primarily focused on acute toxicity, necessitating an in-depth exploration of long-term toxicity and chronic exposure effects [110]. The comprehensive evaluation of the safety and toxicity of nanodrug delivery systems remains challenging due to various potential factors contributing to nanotoxicity and the potential for unpredictable interactions with biological systems [110,111].
The integration of validated analytic methodologies and well-designed experiments can elucidate toxicity mechanisms, ensuring the safe utilization of nanoformulations in therapeutic and diagnostic applications [112,113]. Advancements in nanobiotechnology, material synthesis, and computer simulation studies offer promising prospects for engineering nanoformulations that prioritize both safety and efficacy, thereby mitigating potential toxicities [112]. Notably, regulatory bodies such as EMA and FDA have established guidelines to oversee the application of nanoformulations in medicine [110].
Consequently, further toxicological investigations are imperative to enable an in-depth understanding of nanoparticle behavior, allowing for the enhancement of their properties to minimize toxicity and maximize therapeutic efficacy [111,114]. Studies have documented various toxic effects associated with different nanoparticles, including increased oxidative stress and potential intracellular infiltration by metal-based nanoparticles, as well as observed hepatotoxicity and nephrotoxicity from protein-based nanoparticles [111,115,116].

3. Virus-Based Delivery System

Viral vectors, which make use of viruses’ ability to transfer genetic information into host cells, have shown promise in gene therapy systems [117]. Numerous altered viral vector genomes have been created, such as lentiviruses, herpes viruses, adenoviruses, adeno-associated viruses, retroviruses, human foamy viruses (HFVs), and herpes viruses [118,119] These modified genomes have certain limitations, including immunogenicity, toxin production, insertional mutagenesis, and constraints in transgenic capacity size [120,121]. Recent advancements have led to the design of viral vectors with specific receptors for the transfer of transgenes to non-natural target cells, a concept known as retargeting [120].
Retroviral vectors are commonly used for gene delivery in both somatic and germline gene therapies due to their ability to transfect dividing cells by passing through the nuclear pores of mitotic cells [122]. This feature renders retroviruses suitable candidates for in situ treatments, distinguishing them from adenoviral and lentiviral vectors [120]. These viruses integrate with the host genome, allowing the production of viral proteins, such as gag, pol, and env, that are extracted during gene delivery [123]. The coordinated design of packaging cell lines and retroviral expression vectors is the foundation of retroviral gene transfer technology, which makes it easier to package recombinant retroviral RNAs into infectious, replication-incompetent particles [124].
Lentiviruses, a subclass of retroviruses, have emerged as significant gene delivery vectors, distinguished by their unique capacity to naturally integrate with nondividing cells [125]. This characteristic sets them apart from other retroviruses, which can solely infect dividing cells [126]. The strong tropism of lentiviruses for neural stem cells has rendered them invaluable for ex vivo gene transfer in the central nervous system, exhibiting no significant immune responses or unwanted side effects [127,128]. These vectors offer several advantages, including the high-efficiency infection of both dividing and nondividing cells, long-term stable expression of a transgene, low immunogenicity, and the ability to accommodate larger transgenes [120,129]. Due to their ability to integrate the transgene into the host genome and enable long-term expression, lentiviral vectors have remained a compelling option for clinical gene delivery [130].
Adenoviral vectors, derived from various species and boasting over 100 different serotypes [131], exhibit a versatile nature that allows them to be employed for transferring genetic material into both dividing and non-dividing cells [128]. Notably, adenoviruses such as type 2 and 5 possess low host specificity, enabling their utilization in gene delivery across a wide range of tissues [120]. Large DNA particles up to 38 kb may be transported by these vectors, but unlike retroviruses, they do not integrate into the host genome, causing comparatively short-term gene activation [120,132]. Adenoviral vectors exist in different generations, with the initial generation involving the deletion of the E1 gene, which might trigger acute and chronic immune responses [130,133]. Subsequent generations have seen the removal of the E2 and E4 genes to mitigate the immune response [130].
These systems benefit from the structural protection of the virus, preventing DNA degradation via the lysosome [117]. RNA-based gene delivery has also gained attention, with systems such as oncoretroviral vectors and lentiviral vectors, offering transient gene expression [117,134]. Although viral gene delivery technologies provide therapeutic and continuous gene expression, there are still issues with manufacturing, immunogenicity, toxicity, and the requirement for extensive tuning [135]. Researchers continue to explore the optimal design of viral vectors, considering the specific virus type to be used [135,136].

4. Advantages and Challenges of Replicon RNA Therapy

The application of nucleic acid-based therapy offers promise in addressing diseases that traditional treatments struggle to manage [137,138]. Self-replicating RNA, known as replicon RNA (RepRNA), represents a novel frontier in gene therapy [137,139]. These RepRNA molecules, derived from RNA viruses with specific gene deletions, can amplify all encoded genetic information, resulting in high antigen expression levels [137]. This sets RepRNA apart from plasmid DNA-based vaccines, which depend on delivering initial genetic information to the nucleus [139].
mRNA therapy, another breakthrough, leverages nanoparticles as intracellular delivery systems for normal genes [138,140]. These synthetically manipulated mRNAs can replace missing or defective genes and express transient proteins to correct genetic disorders and promote beneficial mechanisms or pathways, ultimately aiming to cure or treat pathologies [16,141].
repRNA’s favorable characteristics are evident in its therapeutic potential [139]. RepRNA-based vaccines have proven safe and potent in clinical trials against various infectious diseases and cancers [142]. RepRNA serves as a template for increased RNA molecule translation, leading to more rounds of antigen production [143]. Unlike DNA, which requires entry into the cell nucleus, both mRNA and RepRNA perform protein translation in the cytoplasm, reducing the risk of gene interactions [139].
In the comprehensive exploration of nanoformulations integrated with repRNA technology, a multifaceted approach unfolds. The synergy between various nanoformulations and repRNA is visually depicted in Figure 1a, illustrating the diverse forms of association that empower this innovative therapeutic approach. From liposomes to polymeric nanoparticles, this mix enhances the pharmacological potential of repRNA. These nanoformulations also delve into the realm of immunomodulation, as illustrated in Figure 1b. They possess the remarkable capacity to either stimulate or suppress the immune response, opening the door to a range of therapeutic applications. Whether due to their potential for immunostimulation to combat infections or employing them for immunodepression in autoimmune disorders, the versatility of nanoformulations associated with repRNA holds immense promise in shaping the future of medical treatments [144,145].
The RepRNA production system is based on the loading of nanoparticles with RepRNA, followed by the efficient uptake of this formulation by target cells. These nanoparticles have a protective role against RNAses and degradation processes, ensuring the integrity of the encapsulated RNA [146]. Once inside the cellular cytoplasm, the nanoparticles underwent controlled disintegration, facilitating the amplification of RNA and a subsequent increase in the number of copies and protein expression [146]. This orchestrated process ultimately led to the presentation of RNA-derived antigens to the major histocompatibility complex (MHC), eliciting a robust immune response and highlighting the potential of our nanoparticle-based RNA delivery system.
Furthermore, RepRNA production is considered simpler and more cost-effective than mRNA production, as it generates superior immune responses with significantly lower RNA doses due to the presence of RNA replicons [139]. However, despite these advantages, RepRNA therapy still faces major challenges [1,16].
The sizeable nature of RepRNA presents a significant obstacle to efficient cytosolic delivery, particularly when compared to smaller conventional mRNA [8]. The significant challenges associated with large and intricate RepRNA vaccines primarily revolve around their vulnerability to ribonucleases (RNases) and the ineffective translation process within dendritic cells (DCs) [7]. Consequently, one of the primary hurdles to expanding the extensive potential of RepRNA vaccines lies in the imperative requirement for safe and effective in vivo delivery strategies [8]. These limitations underscore the critical need for innovative strategies to mitigate the impact of these obstacles and enhance the overall efficacy of RepRNA vaccines.
One significant challenge lies in the delivery of RNA, given its negative charge and large molecular size, making it difficult to traverse the anionic lipid bilayer of cells [137]. Additionally, its single-stranded nature renders it fragile and susceptible to degradation by RNase enzymes, hindering its intracellular entry and limiting its applications in gene therapy [147]. Recent scientific and technological advances have made strides in overcoming these RNA-associated obstacles, leading to the development of efficient delivery systems that protect RNA from degradation, break through cell membranes, and deliver RNA into the cytoplasm [148].
Another challenge is the route of administration, primarily intravenous (i.v.) delivery, which targets highly vascularized organs like the liver or spleen to achieve therapeutic protein expression in a sufficient number of transfected cells [149]. However, this approach requires prolonged administration times and results in high treatment costs, often necessitating hospitalization or monitoring in infusion centers. Therefore, researchers have increasingly explored subcutaneous or intramuscular applications to address these limitations [142].
In recent years, RepRNA has gained attention as a potential therapy for COVID-19, and its success has stimulated interest in its application for various other pathologies (Table 2).
Notably, McCullough and collaborators (2014) laid the foundation by demonstrating the transport of self-amplifying, cap-independent replicon RNA to dendritic cells (DCs), resulting in increased RepRNA translation and the activation of the immune response [156].
Garcia et al. (2018) conducted a study to evaluate the impact of macrophage migration inhibitory factor (PMIF) immunoneutralization on host responses. Their research revealed remarkable advancements, including the enhanced control of liver and blood-stage Plasmodium infection and complete protection against reinfection, through a novel RNA replicon-based vaccine. A cationic nanoemulsion was prepared by combining squalene, DOTAP, and sorbitan trioleate, and it was characterized for particle size, RNAse protection, and endotoxin, as previously described [150,157]. The complexation process was allowed to continue for at least 30 min before immunization [150]. Intriguingly, the study observed a delay in the onset of blood-stage patency following sporozoite infection [150]. Furthermore, vaccination against PMIF resulted in notable improvements in T follicular helper (Tfh) cell and germinal center responses, a reduction in the expression of Th1-associated inflammatory markers such as TNF-α, IL-12, and IFN-γ during the blood-stage infection, elevated anti-Plasmodium antibody levels, and the enhanced differentiation of antigen-experienced memory CD4 T cells and liver-resident CD8 T cells [150].
To facilitate the delivery of an alphavirus replicon encoding the highly neutralizing human monoclonal antibody (mAb) ZIKV-117, Erasmus et al. (2020) engineered a nanostructured lipid carrier (NLC). These NLC nanoparticles exhibit exceptional colloidal stability attributed to their hybrid core composed of liquid squalene and solid glyceryl trimyristate (Dynasan 114) [158]. Their studies have demonstrated robust protection, both as pre-exposure prophylaxis and post-exposure therapy, following alphavirus-driven expression of ZIKV-117 mRNA delivered via intramuscular injection (IM) [151]. The nanoparticle formulation was manufactured using a specific composition [151,158]. The oil phase was composed of squalene and glyceryl trimyristate, along with a non-ionic sorbitan ester surfactant, and the cationic lipid DOTAP. The aqueous phase consisted of a 10 mM sodium citrate trihydrate buffer containing the non-ionic PEGylated surfactant Tween 80. Additionally, the researchers successfully optimized the expression of human IgG from repRNA, enabling the production of protective levels in mice [151].
Furthermore, the same research group unveiled the LION/repRNA-CoV2S vaccine, exhibiting substantial neutralization of SARS-CoV-2 and inducing potent S-specific T cell responses in vaccinated mice and pigtail macaques [142]. According to Erasmus et al. (2020) LION is a highly stable cationic squalene emulsion containing 15 nm superparamagnetic iron oxide (Fe3O4) nanoparticles (SPIO) incorporated in the hydrophobic oil phase. This vaccine’s ability to stimulate memory T cell responses specific to SARS-CoV-2 in macaques may contribute to long-term protection against and recovery from SARS-CoV-2 infection [142].
Recent findings by Leventhal et al. (2022) have significantly improved the understanding of how vaccines against the Crimean–Congo hemorrhagic fever virus (CCHFV) provide protection. Vaccine RNA was synthesized in vitro and subsequently complexed to cationic nanocarrier, following established protocols as described previously by Erasmus et al. (2020). Their research has shed light on the involvement of humoral and cellular immunity in repRNA-mediated protection. Importantly, administering RNA through straightforward intramuscular vaccinations, particularly with the use of cationic emulsions, has emerged as an essential strategy for enhancing RNA stability and transport [152].
In the study conducted by Fotoran et al., tests were managed on a self-amplifying repRNA (repRNA) vaccine formulation, which was based on the SP6 Venezuelan equine encephalitis (VEE) vector incorporated into cationic liposomes. They generated three vaccines that encoded two reporter genes (GFP and nanoLuc) and the Plasmodium falciparum reticulocyte binding protein homologue 5 (PfRH5). The liposome–replicon complexes exhibited high transfection efficiencies and elicited antibodies capable of inhibiting the growth of the parasite in vitro [153].
To find out how mannans of different lengths (from mono to tetrasaccharide) affected the antibody response of a model repRNA replicon that encodes the respiratory syncytial virus fusion F protein, a new set of LNPs with changed surfaces was created by Goswami et al. (2021). As the mannose chain length increased, the vaccination priming response showed a steady improvement; nevertheless, the response to the booster dose peaked at a length greater than the disaccharide. Mannosylated lipid nanoparticles (MLNPs) were shown to exhibit higher amounts of IgG1 and IgG2a in comparison to LNPs. The potential of mannosylated samRNA LNPs for intramuscular and intradermal distribution is confirmed by this study [154].
Chahala et al. (2016) created a single-dose, completely synthetic, adjuvant-free dendrimer nanoparticle vaccination platform with a fast response, and encapsulated mRNA replicons that encode antigens. Protective immunity against a wide range of deadly pathogen threats, including as the Ebola virus, Toxoplasma gondii, and H1N1 influenza, can be produced via this mechanism [155]. The vaccine possesses the ability to induce both CD8+ T-cell and antibody responses and may be designed with various antigen-expressing replicons [155].

5. Advances and Perspectives

Various RNA-based therapies have emerged, including RNA-directed drugs involving chemically modified oligonucleotides, which represent a significant advancement in the field of RNA therapeutics [159]. The extensive range of diseases currently undergoing clinical trials highlights the potential of nanoparticles, especially lipid nanoparticles (LNPs), in the realm of RNA-based therapies [160]. Additionally, coupled nanoformulations have exhibited promise in various treatments (Table 3).
Massad-Massade et al. (2018) synthesized siRNA-SQ (squalene) and siRNA-SOLA (solanesol) and achieved the production of effective siRNA-nanoparticles using an innovative method that easily adapts to pharmaceutical development. They utilized nanoprecipitation in acetone/water to create the nanoparticles [161].
The simplicity and adaptability of the researchers’ proposed methodology, along with the increased synthesis yield, provide a novel approach for targeting overexpressed genes using siRNA–polyisoprenoid-conjugated nanoparticles, thereby reducing the risk of malignancies or genetic disorders [161,163,164,165].
The study conducted by Guo et al. in 2020 showed that intravenous administrations of RNA-paclitaxel nanoparticles, integrated with precise cancer-targeting ligands, efficiently addressed the solubility challenges of hydrophobic drugs in patient care. Notably, this strategy resulted in a significant suppression of breast cancer growth in mice, accompanied by negligible levels of toxicity and immune responses [162].
Furthermore, Song et al. (2020) designed mesoporous silica nanoparticles (MSN) loaded with myricetin (Myr) and multidrug resistance protein (MRP-1) siRNA and modified with folic acid (FA) to enhance therapeutic efficacy for non-small cell lung cancer (NSCLC) treatment. These nanoparticles exhibited sustained Myr release and improved uptake by lung cancer cells compared to non-targeted ones. The in vitro drug release data confirmed sustained release in FA-conjugated MSN with Myr and MRP-1 nanoparticles [166]. Treatment involving FA-conjugated MSN with Myr and MRP-1 significantly reduced the viability of lung cancer cell lines (A549 and NCI-H1299) and inhibited colony formation [166].
The potential of mRNA technology in various applications, including cancer immunotherapy and infectious disease vaccination, is evident, but its success depends on improving stability and reducing immunogenicity [2]. Moreover, due to electrostatic repulsion by cell membranes and vulnerability to RNases, mRNA requires protection and efficient transport to target cells. Over the years, researchers have harnessed lipid vesicles to safeguard and deliver RNA molecules, with advancements incorporating structural lipids to mimic cell membranes and shield the positive charge [171].
Recent approvals of RNA-based technologies, including Patisiran (Onpattro®), Givoriran from Alnylam, and the emergency-use mRNA-LNP vaccine for COVID-19 (Comirnaty®), underscore the safety and efficacy of LNP-based nucleic acid delivery [160,171,172].
Patisiran, an innovative medicine, uses a liposomal siRNA to selectively target transthyretin (TTR), a plasma transport protein for retinol-binding protein and thyroid hormone [167]. This RNA interference (RNAi) therapy treats polyneuropathy associated with transthyretin amyloidosis (ATTR) [168]. Patisiran’s therapeutic component consists of a novel siRNA formulated as a lipid nanoparticle, which effectively reduces TTR accumulation in tissues, leading to improvements in cardiac and neuropathic function [167,168].
Givosiran, developed by Alnylam Pharmaceuticals in Cambridge, Massachusetts, is a highly selective siRNA designed to reduce the occurrence of recurrent episodes in conditions like acute intermittent porphyria (AIP) by downregulating hepatic delta-aminolevulinic acid synthase 1 (ALAS1) enzyme expression [169,170]. This siRNA, which specifically targets the messenger RNA (mRNA) encoding ALAS1, effectively reduces induced ALAS1 levels, preventing the accumulation of potentially harmful porphyrin precursors such as porphobilinogen (PBG) and delta-aminolevulinic acid (ALA) [169,170]. Givosiran demonstrates selectivity for hepatocytes due to its linkage to N-acetylgalactosamine (GalNac), facilitating its uptake through asialoglycoprotein receptors (ASGPR) [169]. While Givosiran demonstrates efficacy in reducing recurrent episodes, it also has potential adverse effects, including hepatic, renal, and hyperhomocysteinemia risks [169].
The durability of mRNA-LNP vaccines depends on their chemical composition, with various mRNA vaccine candidates created during the development of vaccines targeting SARS-CoV-2 [173]. Among these candidates, three “conventional” mRNA vaccines, all encoding the entire S protein, have advanced to advanced clinical development stages. These vaccines are the Moderna mRNA-1273 vaccine, the BioNTech/Pfizer BNT162b2/Comirnaty vaccine, and the CureVac CVnCoV vaccine [173].
Nonetheless, while LNPs offer a highly customizable platform for nucleic acid delivery, particularly in mRNA vaccines, and exhibit potential in therapeutics for rare diseases and cancers, it is important to note that there is no one-size-fits-all solution for all diseases, necessitating ongoing optimization efforts [171]. Amid heightened public interest in mRNA vaccines, scientists have dedicated a wealth of scientific research to design and refine this technology, ushering in a new era in nanomedicine [160,171,172,174].

6. Conclusions

The rapid progress in the field of RNA-based therapeutics, driven by the resounding success of mRNA technology and the urgency brought about by the COVID-19 pandemic, has illuminated a promising path for repRNA technology. The recent approvals of mRNA-based products have not only demonstrated the regulatory acceptance, but also highlighted the immense potential of the RNA platform in modern medicine. As we find ourselves at the crossroads of innovation in nanomedicine, the potential of repRNA technology as a versatile tool against a myriad of diseases becomes evident. Its distinctive capacity for high-level antigen expression, simplified production processes, and the potential for lower therapeutic doses have positioned it as a formidable contender for the future of medical therapies. However, to fully realize its potential and ensure its place as a transformative technology, we must address several key challenges and consider specific future directions.
Future research should prioritize increasing the stability of repRNA to mitigate the different degradation possibilities, making its application more multifaceted. Improving resistance to degradation is pivotal for ensuring the sustained therapeutic effect of repRNA-based treatments. Moreover, to unlock the full potential of repRNA, researchers must continue to explore innovative methods for efficient and targeted delivery, overcoming obstacles in delivering repRNA to specific cells or tissues. As repRNA therapies advance, understanding and mitigating their immunogenicity remains a crucial area of study. A focus on minimizing immune responses and side effects is vital to ensure patient safety and treatment effectiveness. Beyond infectious diseases, repRNA holds promise in addressing genetic disorders, cancer, and chronic illnesses. Future research and development efforts should expand its applications. To facilitate the integration of repRNA into mainstream medical practice, a robust and clear regulatory framework must be established. This framework should encourage innovation while ensuring patient safety and equitable access. The momentum in RNA-based therapeutics is undeniable, and with sustained research and innovation, repRNA stands to become one of the most transformative medical technologies of our time. It offers the promise of more effective treatments and potential cures across a range of illnesses, making the future of medicine brighter and more hopeful.

Author Contributions

Conceptualization, E.S.L., D.d.S., A.L.S., M.E.M., M.E.B., S.S.S.J., B.S.D.F., L.M.d.S.F. and B.A.S.M.; methodology, B.S.D.F., K.V.S.H. and B.A.S.M.; software, K.V.S.H.; writing—original draft preparation, E.S.L., D.d.S., A.L.S., M.E.M., M.E.B., S.S.S.J. and B.A.S.M.; writing—review and editing, B.S.D.F., K.V.S.H., L.M.d.S.F. and B.A.S.M.; visualization, V.P.C.R., L.M.d.S.F., D.D.G.N., K.V.S.H. and B.A.S.M.; supervision, B.A.S.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by CNPq Brazil–“Conselho Nacional de Desenvolvimento Científico e Tecnológico” (Grant Number 424911/2021-3). BASM is acknowledged as a Technological Fellow supported by CNPq (Grant Number 306041/2021).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable.

Acknowledgments

The authors immensely thank the University Center SENAI/CIMATEC (National Service for Industrial Training (SENAI), Bahia–Brazil, and the National System of the Nanotechnology Laboratories (SisNANO/MCTI/Brazil) Plataforma Aberta CIMATEC Nano.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lundstrom, K. Nanoparticle-Based Delivery of Self-Amplifying RNA. Gene Ther. 2020, 27, 183–185. [Google Scholar] [CrossRef]
  2. Sergeeva, O.V.; Koteliansky, V.E.; Zatsepin, T.S. MRNA-Based Therapeutics–Advances and Perspectives. Biochemistry 2016, 81, 709–722. [Google Scholar] [CrossRef]
  3. Guo, P.; Haque, F.; Hallahan, B.; Reif, R.; Li, H. Uniqueness, Advantages, Challenges, Solutions, and Perspectives in Therapeutics Applying RNA Nanotechnology. Nucleic Acid. Ther. 2012, 22, 226–245. [Google Scholar] [CrossRef] [PubMed]
  4. Tenchov, R.; Bird, R.; Curtze, A.E.; Zhou, Q. Lipid Nanoparticles from Liposomes to MRNA Vaccine Delivery, a Landscape of Research Diversity and Advancement. ACS Nano 2021, 15, 16982–17015. [Google Scholar] [CrossRef] [PubMed]
  5. Beck, J.D.; Reidenbach, D.; Salomon, N.; Sahin, U.; Türeci, Ö.; Vormehr, M.; Kranz, L.M. MRNA Therapeutics in Cancer Immunotherapy. Mol. Cancer 2021, 20, 69. [Google Scholar] [CrossRef]
  6. Sahin, U.; Karikó, K.; Türeci, Ö. MRNA-Based Therapeutics-Developing a New Class of Drugs. Nat. Rev. Drug Discov. 2014, 13, 759–780. [Google Scholar] [CrossRef] [PubMed]
  7. Démoulins, T.; Englezou, P.C.; Milona, P.; Ruggli, N.; Tirelli, N.; Pichon, C.; Sapet, C.; Ebensen, T.; Guzmán, C.A.; McCullough, K.C. Self-Replicating RNA Vaccine Delivery to Dendritic Cells. In Methods in Molecular Biology; Humana Press Inc.: Totowa, NJ, USA, 2017; Volume 1499, pp. 37–75. [Google Scholar]
  8. Lin, G.; Yan, H.; Sun, J.; Zhao, J.; Zhang, Y. Self-Replicating RNA Nanoparticle Vaccine Elicits Protective Immune Responses against SARS-CoV-2. Mol. Ther. Nucleic Acids 2023, 32, 650–666. [Google Scholar] [CrossRef]
  9. Lundstrom, K. Self-Replicating RNA Viruses for RNA Therapeutics. Molecules 2018, 23, 3310. [Google Scholar] [CrossRef]
  10. Nakamura, A.; Kotaki, T.; Nagai, Y.; Takazawa, S.; Tokunaga, K.; Kameoka, M. Construction and Evaluation of a Self-Replicative RNA Vaccine against SARS-CoV-2 Using Yellow Fever Virus Replicon. PLoS ONE 2022, 17, e0274829. [Google Scholar] [CrossRef]
  11. Dana, H.; Mahmoodi Chalbatani, G.; Mahmoodzadeh, H.; Karimloo, R.; Rezaiean, O.; Moradzadeh, A.; Mehmandoost, N.; Moazzen, F.; Mazraeh, A.; Marmari, V.; et al. Molecular Mechanisms and Biological Functions of SiRNA. Int. J. Biomed. Sci. IJBS 2017, 13, 48–57. [Google Scholar] [CrossRef]
  12. Alshaer, W.; Zureigat, H.; Al Karaki, A.; Al-Kadash, A.; Gharaibeh, L.; Hatmal, M.M.; Aljabali, A.A.A.; Awidi, A. SiRNA: Mechanism of Action, Challenges, and Therapeutic Approaches. Eur. J. Pharmacol. 2021, 905, 174178. [Google Scholar] [CrossRef] [PubMed]
  13. Yu, B.; Tai, H.C.; Xue, W.; Lee, L.J.; Lee, R.J. Receptor-Targeted Nanocarriers for Therapeutic Delivery to Cancer. Mol. Membr. Biol. 2010, 27, 286–298. [Google Scholar] [CrossRef] [PubMed]
  14. Friedrich, M.; Aigner, A. Therapeutic SiRNA: State-of-the-Art and Future Perspectives. BioDrugs 2022, 36, 549–571. [Google Scholar] [CrossRef] [PubMed]
  15. Shu, Y.; Pi, F.; Sharma, A.; Rajabi, M.; Haque, F.; Shu, D.; Leggas, M.; Evers, B.M.; Guo, P. Stable RNA Nanoparticles as Potential New Generation Drugs for Cancer Therapy. Adv. Drug Deliv. Rev. 2014, 66, 74–89. [Google Scholar] [CrossRef]
  16. Li, M.; Li, Y.; Li, S.; Jia, L.; Wang, H.; Li, M.; Deng, J.; Zhu, A.; Ma, L.; Li, W.; et al. The Nano Delivery Systems and Applications of MRNA. Eur. J. Med. Chem. 2022, 227, 113910. [Google Scholar] [CrossRef]
  17. Antunes Filho, S.; Backx, B.P. Nanotecnologia e Seus Impactos Na Sociedade. Rev. Tecnol. Soc. 2020, 16, 1–15. [Google Scholar] [CrossRef]
  18. Bayda, S.; Adeel, M.; Tuccinardi, T.; Cordani, M.; Rizzolio, F. The History of Nanoscience and Nanotechnology: From Chemical-Physical Applications to Nanomedicine. Molecules 2020, 25, 112. [Google Scholar] [CrossRef]
  19. Fialkoski, D.; Malfatti, C.R.M. Nanotecnologia: Uma Prospecção Tecnológica No Âmbito Nacional e Internacional. Cad. Prospecção 2019, 12, 590. [Google Scholar] [CrossRef]
  20. Nunes, A.R.; Costa, E.C.; Alves, G.; Silva, L.R. Nanoformulations for the Delivery of Dietary Anthocyanins for the Prevention and Treatment of Diabetes Mellitus and Its Complications. Pharmaceuticals 2023, 16, 736. [Google Scholar] [CrossRef]
  21. Chakravarty, M.; Vora, A. Nanotechnology-Based Antiviral Therapeutics. Drug Deliv. Transl. Res. 2021, 11, 748–787. [Google Scholar] [CrossRef]
  22. Cui, F.; Liu, J.; Pang, S.; Li, B. Recent Advance in Tumor Microenvironment-Based Stimuli-Responsive Nanoscale Drug Delivery and Imaging Platform. Front. Pharmacol. 2022, 13, 929854. [Google Scholar] [CrossRef] [PubMed]
  23. Guo, P. The Emerging Field of RNA Nanotechnology. Nat. Nanotechnol. 2010, 5, 833–842. [Google Scholar] [CrossRef] [PubMed]
  24. Jasinski, D.; Haque, F.; Binzel, D.W.; Guo, P. Advancement of the Emerging Field of RNA Nanotechnology. ACS Nano 2017, 11, 1142–1164. [Google Scholar] [CrossRef] [PubMed]
  25. Guo, P.; Zhang, C.; Chen, C.; Garver, K.; Trottier, M. Inter-RNA Interaction of Phage Φ29 PRNA to form a Hexameric Complex for Viral DNA Transportation. Mol. Cell 1998, 2, 149–155. [Google Scholar] [CrossRef] [PubMed]
  26. Zafar, A.; Alruwaili, N.K.; Imam, S.S.; Alharbi, K.S.; Afzal, M.; Alotaibi, N.H.; Yasir, M.; Elmowafy, M.; Alshehri, S. Novel Nanotechnology Approaches for Diagnosis and Therapy of Breast, Ovarian and Cervical Cancer in Female: A Review. J. Drug Deliv. Sci. Technol. 2021, 61, 102198. [Google Scholar] [CrossRef]
  27. Maji, R.; Dey, N.S.; Satapathy, B.S.; Mukherjee, B.; Mondal, S. Preparation and Characterization of Tamoxifen Citrate Loaded Nanoparticles for Breast Cancer Therapy. Int. J. Nanomed. 2015, 9, 3107–3118. [Google Scholar] [CrossRef]
  28. Carvalho Lopes, J.; Pereira Torres, M.L. Utilização de Nanopartículas No Tratamento Do Câncer: Aspectos Gerais, Mecanismos de Ação Antineoplásicos e Aplicabilidades Tumorais. Rev. Bras. Cancerol. 2020, 65, e-13400. [Google Scholar] [CrossRef]
  29. Assadpour, E.; Rezaei, A.; Das, S.S.; Krishna Rao, B.V.; Singh, S.K.; Kharazmi, M.S.; Jha, N.K.; Jha, S.K.; Prieto, M.A.; Jafari, S.M. Cannabidiol-Loaded Nanocarriers and Their Therapeutic Applications. Pharmaceuticals 2023, 16, 487. [Google Scholar] [CrossRef]
  30. Lashkari, A.; Ranjbar, R. Nanoparticles and Nanoformulated Drugs as Promising Delivery System in Treatment of Microbial-Induced CNS Infection: A Systematic Review of Literature. J. Neurovirol. 2021, 27, 542–549. [Google Scholar] [CrossRef]
  31. Zanoni, E.T.; Cardoso, W.A.; Baesso, A.S.; Savi, G.D.; Folgueras, M.V.; Mendes, E.; Angioletto, E. Evaluation of Antimicrobial Activity and Adsorption of Silica Nanoparticles Doped with Cuo. Rev. Mater. 2019, 24, 1–11. [Google Scholar] [CrossRef]
  32. Frézard, F. Liposomes: From Biophysics to the Design of Peptide Vaccines. Braz. J. Med. Biol. Res. 1999, 32, 181–189. [Google Scholar] [CrossRef] [PubMed]
  33. Nsairat, H.; Khater, D.; Sayed, U.; Odeh, F.; Al Bawab, A.; Alshaer, W. Liposomes: Structure, Composition, Types, and Clinical Applications. Heliyon 2022, 8, e09394. [Google Scholar] [CrossRef] [PubMed]
  34. Najahi-Missaoui, W.; Arnold, R.D.; Cummings, B.S. Safe Nanoparticles: Are We There Yet? Int. J. Mol. Sci. 2021, 22, 385. [Google Scholar] [CrossRef] [PubMed]
  35. Coutinho, J.A. Nanopartículas na Entrega Eficaz e Segura de Fármacos Ao Cérebro Por via Nasal—Experiência Profissionalizante Na Vertente de Farmácia Comunitária, Hospitalar e Investigação. Master’s Thesis, University of Beira Interior, Covilhã, Portugal, 2015. [Google Scholar]
  36. Chaves, M.A.; Ferreira, L.S.; Baldino, L.; Pinho, S.C.; Reverchon, E. Current Applications of Liposomes for the Delivery of Vitamins: A Systematic Review. Nanomaterials 2023, 13, 1557. [Google Scholar] [CrossRef] [PubMed]
  37. Guimarães, D.; Cavaco-Paulo, A.; Nogueira, E. Design of Liposomes as Drug Delivery System for Therapeutic Applications. Int. J. Pharm. 2021, 601, 120571. [Google Scholar] [CrossRef]
  38. Alavi, M.; Hamidi, M. Passive and Active Targeting in Cancer Therapy by Liposomes and Lipid Nanoparticles. Drug Metab. Pers. Ther. 2019, 34, 20180032. [Google Scholar] [CrossRef] [PubMed]
  39. Raju, R.; Abuwatfa, W.H.; Pitt, W.G.; Husseini, G.A. Liposomes for the Treatment of Brain Cancer—A Review. Pharmaceuticals 2023, 16, 1056. [Google Scholar] [CrossRef]
  40. Andrade, G.M.; Júnior, W.T.d.S.; De Souza, E.L.; Porto, P.G.; Araújo, C.d.S.; Martins, C.S.; Dos Santos, S.S.; Peixoto, C.C.C. de M.; Hayakawa, T.M.; Rosario, J.F.A. Compilação de Dados Referentes a Síndrome de Grisel: Exposição de Evidências/Compilation of Data Relating to Grisel Syndrome: Exhibition of Evidence. Braz. J. Health Rev. 2022, 5, 5783–5789. [Google Scholar] [CrossRef]
  41. Rajasegaran, T.; How, C.W.; Saud, A.; Ali, A.; Lim, J.C.W. Targeting Inflammation in Non-Small Cell Lung Cancer through Drug Repurposing. Pharmaceuticals 2023, 16, 451. [Google Scholar] [CrossRef]
  42. Van Tran, V.; Moon, J.Y.; Lee, Y.C. Liposomes for Delivery of Antioxidants in Cosmeceuticals: Challenges and Development Strategies. J. Control. Release 2019, 300, 114–140. [Google Scholar] [CrossRef]
  43. Shah, S.; Dhawan, V.; Holm, R.; Nagarsenker, M.S.; Perrie, Y. Liposomes: Advancements and Innovation in the Manufacturing Process. Adv. Drug Deliv. Rev. 2020, 154–155, 102–122. [Google Scholar] [CrossRef]
  44. Sayour, E.J.; Mendez-Gomez, H.R.; Mitchell, D.A. Cancer Vaccine Immunotherapy with RNA-Loaded Liposomes. Int. J. Mol. Sci. 2018, 19, 2890. [Google Scholar] [CrossRef] [PubMed]
  45. Zylberberg, C.; Gaskill, K.; Pasley, S.; Matosevic, S. Engineering Liposomal Nanoparticles for Targeted Gene Therapy. Gene Ther. 2017, 24, 441–452. [Google Scholar] [CrossRef] [PubMed]
  46. Saffari, M.; Moghimi, H.R.; Dass, C.R. Barriers to Liposomal Gene Delivery: From Application Site to the Target. Iran. J. Pharm. Res. IJPR 2016, 15, 3. [Google Scholar]
  47. Ropert, C. Liposomes as a Gene Delivery System. Braz. J. Med. Biol. Res. 1999, 32, 163–169. [Google Scholar] [CrossRef]
  48. Guillot, A.J.; Martínez-Navarrete, M.; Garrigues, T.M.; Melero, A. Skin Drug Delivery Using Lipid Vesicles: A Starting Guideline for Their Development. J. Control. Release 2023, 355, 624–654. [Google Scholar] [CrossRef]
  49. Wolinsky, J.B.; Colson, Y.L.; Grinstaff, M.W. Local Drug Delivery Strategies for Cancer Treatment: Gels, Nanoparticles, Polymeric Films, Rods, and Wafers. J. Control. Release 2012, 159, 14–26. [Google Scholar] [CrossRef]
  50. Xu, L.; Anchordoquy, T. Drug Delivery Trends in Clinical Trials and Translational Medicine: Challenges and Opportunities in the Delivery of Nucleic Acid-Based Therapeutics. J. Pharm. Sci. 2011, 100, 38–52. [Google Scholar] [CrossRef] [PubMed]
  51. Balazs, D.A.; Godbey, W.T. Liposomes for Use in Gene Delivery. J. Drug Deliv. 2011, 2011, 326497. [Google Scholar] [CrossRef]
  52. Thapa, B.; Narain, R. Mechanism, Current Challenges and New Approaches for Non Viral Gene Delivery. In Polymers and Nanomaterials for Gene Therapy; Elsevier Inc.: Amsterdam, The Netherlands, 2016; pp. 1–27. ISBN 9780081005217. [Google Scholar]
  53. Uddin, S.N. Cationic Lipids Used in Non-Viral Gene Delivery Systems. Biotechnol. Mol. Biol. Rev. 2007, 2, 58–067. [Google Scholar]
  54. Zielinska, A.; Carreiró, F.; Oliveira, A.M.; Neves, A.; Pires, B.; Nagasamy Venkatesh, D.; Durazzo, A.; Lucarini, M.; Eder, P.; Silva, A.M.; et al. Polymeric Nanoparticles: Production, Characterization, Toxicology and Ecotoxicology. Molecules 2020, 25, 3731. [Google Scholar] [CrossRef] [PubMed]
  55. El-Say, K.M.; El-Sawy, H.S. Polymeric Nanoparticles: Promising Platform for Drug Delivery. Int. J. Pharm. 2017, 528, 675–691. [Google Scholar] [CrossRef] [PubMed]
  56. Rai, R.; Alwani, S.; Badea, I. Polymeric Nanoparticles in Gene Therapy: New Avenues of Design and Optimization for Delivery Applications. Polymers 2019, 11, 745. [Google Scholar] [CrossRef]
  57. Begines, B.; Ortiz, T.; Pérez-Aranda, M.; Martínez, G.; Merinero, M.; Argüelles-Arias, F.; Alcudia, A. Polymeric Nanoparticles for Drug Delivery: Recent Developments and Future Prospects. Nanomaterials 2020, 10, 1403. [Google Scholar] [CrossRef] [PubMed]
  58. Blakney, A.K.; McKay, P.F.; Hu, K.; Samnuan, K.; Jain, N.; Brown, A.; Thomas, A.; Rogers, P.; Polra, K.; Sallah, H.; et al. Polymeric and Lipid Nanoparticles for Delivery of Self-Amplifying RNA Vaccines. J. Control. Release 2021, 338, 201–210. [Google Scholar] [CrossRef] [PubMed]
  59. Leyva-Gómez, G.; Piñón-Segundo, E.; Mendoza-Muñoz, N.; Zambrano-Zaragoza, M.L.; Mendoza-Elvira, S.; Quintanar-Guerrero, D. Approaches in Polymeric Nanoparticles for Vaginal Drug Delivery: A Review of the State of the Art. Int. J. Mol. Sci. 2018, 19, 1549. [Google Scholar] [CrossRef]
  60. MacHtakova, M.; Thérien-Aubin, H.; Landfester, K. Polymer Nano-Systems for the Encapsulation and Delivery of Active Biomacromolecular Therapeutic Agents. Chem. Soc. Rev. 2022, 51, 128–152. [Google Scholar] [CrossRef]
  61. Lächelt, U.; Wagner, E. Nucleic Acid Therapeutics Using Polyplexes: A Journey of 50 Years (and Beyond). Chem. Rev. 2015, 115, 11043–11078. [Google Scholar] [CrossRef]
  62. Ita, K. Polyplexes for Gene and Nucleic Acid Delivery: Progress and Bottlenecks. Eur. J. Pharm. Sci. 2020, 150, 105358. [Google Scholar] [CrossRef]
  63. Vasile, C. Polymeric Nanomaterials: Recent Developments, Properties and Medical Applications. In Polymeric Nanomaterials in Nanotherapeutics; Elsevier: Amsterdam, The Netherlands, 2018; pp. 1–66. ISBN 9780128139325. [Google Scholar]
  64. Peltonen, L.; Singhal, M.; Hirvonen, J. Principles of Nanosized Drug Delivery Systems. In Nanoengineered Biomaterials for Advanced Drug Delivery; Elsevier: Amsterdam, The Netherlands, 2020; pp. 3–25. ISBN 9780081029855. [Google Scholar]
  65. Singh, N.; Joshi, A.; Pal Toor, A.; Verma, G. Drug delivery: Advancements and challenges. In Nanostructures for Drug Delivery; Elsevier: Amsterdam, The Netherlands, 2017; pp. 865–886. [Google Scholar]
  66. Elmowafy, M.; Shalaby, K.; Elkomy, M.H.; Alsaidan, O.A.; Gomaa, H.A.M.; Abdelgawad, M.A.; Mostafa, E.M. Polymeric Nanoparticles for Delivery of Natural Bioactive Agents: Recent Advances and Challenges. Polymers 2023, 15, 1123. [Google Scholar] [CrossRef]
  67. Stueber, D.D.; Villanova, J.; Aponte, I.; Xiao, Z.; Colvin, V.L. Magnetic Nanoparticles in Biology and Medicine: Past, Present, and Future Trends. Pharmaceutics 2021, 13, 943. [Google Scholar] [CrossRef]
  68. Martins, M.G. Encapsulamento de Nanopartículas Magnéticas Em Polímeros Acrílicos e Avaliação de Hipertermia Para Potencial Tratamento de Câncer. In Master’s Thesis in Pharmaceutical Sciences; Federal University of Rio de Janeiro: Rio de Janeiro, Brazil, 2017. [Google Scholar]
  69. Akbarzadeh, A.; Samiei, M.; Davaran, S. Magnetic Nanoparticles: Preparation, Physical Properties, and Applications in Biomedicine. Nanoscale Res. Lett. 2012, 7, 144. [Google Scholar] [CrossRef] [PubMed]
  70. Materón, E.M.; Miyazaki, C.M.; Carr, O.; Joshi, N.; Picciani, P.H.S.; Dalmaschio, C.J.; Davis, F.; Shimizu, F.M. Magnetic Nanoparticles in Biomedical Applications: A Review. Appl. Surf. Sci. Adv. 2021, 6, 100163. [Google Scholar] [CrossRef]
  71. Farzin, A.; Etesami, S.A.; Quint, J.; Memic, A.; Tamayol, A. Magnetic Nanoparticles in Cancer Therapy and Diagnosis. Adv. Healthc. Mater. 2020, 9, e1901058. [Google Scholar] [CrossRef] [PubMed]
  72. Spoială, A.; Ilie, C.I.; Motelica, L.; Ficai, D.; Semenescu, A.; Oprea, O.C.; Ficai, A. Smart Magnetic Drug Delivery Systems for the Treatment of Cancer. Nanomaterials 2023, 13, 876. [Google Scholar] [CrossRef]
  73. Mohammadi Ziarani, G.; Malmir, M.; Lashgari, N.; Badiei, A. The Role of Hollow Magnetic Nanoparticles in Drug Delivery. RSC Adv. 2019, 9, 25094–25106. [Google Scholar] [CrossRef]
  74. Shasha, C.; Krishnan, K.M. Nonequilibrium Dynamics of Magnetic Nanoparticles with Applications in Biomedicine. Adv. Mater. 2021, 33, e1904131. [Google Scholar] [CrossRef]
  75. Zhao, Z.; Cui, H.; Song, W.; Ru, X.; Zhou, W.; Yu, X. A Simple Magnetic Nanoparticles-Based Viral RNA Extraction Method for Efficient Detection of SARS-CoV-2. bioRxiv 2020. [Google Scholar] [CrossRef]
  76. Gao, F. An Overview of Surface-Functionalized Magnetic Nanoparticles: Preparation and Application for Wastewater Treatment. ChemistrySelect 2019, 4, 6805–6811. [Google Scholar] [CrossRef]
  77. Schneider-Futschik, E.K.; Reyes-Ortega, F. Advantages and Disadvantages of Using Magnetic Nanoparticles for the Treatment of Complicated Ocular Disorders. Pharmaceutics 2021, 13, 1157. [Google Scholar] [CrossRef]
  78. Hobson, D.W. Nanotechnology. In Comprehensive Biotechnology, 2nd ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2011; Volume 3, pp. 683–697. ISBN 9780080885049. [Google Scholar]
  79. Baghaban-Eslaminejad, M.; Oryan, A.; Kamali, A.; Moshiri, A. The Role of Nanomedicine, Nanotechnology, and Nanostructures on Oral Bone Healing, Modeling, and Remodeling. In Nanostructures for Oral Medicine; Elsevier Inc.: Amsterdam, The Netherlands, 2017; pp. 777–832. [Google Scholar]
  80. Dahman, Y. Nanoshells. In Nanotechnology and Functional Materials for Engineers; Elsevier Inc.: Amsterdam, The Netherlands, 2017; pp. 175–190. [Google Scholar]
  81. Wang, Y.C.; Rhéaume, É.; Lesage, F.; Kakkar, A. Synthetic Methodologies to Gold Nanoshells: An Overview. Molecules 2018, 23, 2851. [Google Scholar] [CrossRef] [PubMed]
  82. Ahmadi, A.; Arami, S. Potential Applications of Nanoshells in Biomedical Sciences. J. Drug Target. 2014, 22, 175–190. [Google Scholar] [CrossRef]
  83. Loo, C.; Lin, A.; Hirsch, L.; Lee, M.-H.; Halas, N.; West, J.; Drezek, R. Nanoshell-Enabled Photonics-Based Imaging and Therapy of Cancer. Technol. Cancer Res. Treat. 2004, 3, 33–40. [Google Scholar] [CrossRef] [PubMed]
  84. Muthuraman, A.; Rishitha, N.; Mehdi, S. Role of Nanoparticles in Bioimaging, Diagnosis and Treatment of Cancer Disorder. In Design of Nanostructures for Theranostics Applications; Elsevier: Amsterdam, The Netherlands, 2018; pp. 529–562. ISBN 9780128136690. [Google Scholar]
  85. Lee, S.Y.; Shieh, M.J. Platinum(II) Drug-Loaded Gold Nanoshells for Chemo-Photothermal Therapy in Colorectal Cancer. ACS Appl. Mater. Interfaces 2020, 12, 4254–4264. [Google Scholar] [CrossRef] [PubMed]
  86. Marino, A.; Arai, S.; Hou, Y.; Degl’Innocenti, A.; Cappello, V.; Mazzolai, B.; Chang, Y.T.; Mattoli, V.; Suzuki, M.; Ciofani, G. Gold Nanoshell-Mediated Remote Myotube Activation. ACS Nano 2017, 11, 2494–2505. [Google Scholar] [CrossRef] [PubMed]
  87. Nouri, S.; Mohammadi, E.; Mehravi, B.; Majidi, F.; Ashtari, K.; Neshasteh-Riz, A.; Einali, S. NIR Triggered Glycosylated Gold Nanoshell as a Photothermal Agent on Melanoma Cancer Cells. Artif. Cells Nanomed. Biotechnol. 2019, 47, 2316–2324. [Google Scholar] [CrossRef]
  88. Gao, C.; Lyu, F.; Yin, Y. Encapsulated Metal Nanoparticles for Catalysis. Chem. Rev. 2021, 121, 834–881. [Google Scholar] [CrossRef]
  89. Yaghmur, A.; Mu, H. Recent Advances in Drug Delivery Applications of Cubosomes, Hexosomes, and Solid Lipid Nanoparticles. Acta Pharm. Sin. B 2021, 11, 871–885. [Google Scholar] [CrossRef]
  90. Paliwal, R.; Paliwal, S.R.; Kenwat, R.; Das Kurmi, B.; Sahu, M.K. Solid Lipid Nanoparticles: A Review on Recent Perspectives and Patents. Expert. Opin. Ther. Pat. 2020, 30, 179–194. [Google Scholar] [CrossRef]
  91. De Jesus, M.B.; Zuhorn, I.S. Solid Lipid Nanoparticles as Nucleic Acid Delivery System: Properties and Molecular Mechanisms. J. Control. Release 2015, 201, 1–13. [Google Scholar] [CrossRef]
  92. Subhan, M.A.; Filipczak, N.; Torchilin, V.P. Advances with Lipid-Based Nanosystems for SiRNA Delivery to Breast Cancers. Pharmaceuticals 2023, 16, 970. [Google Scholar] [CrossRef] [PubMed]
  93. Mohanraj, V.J.; Chen, Y. Nanoparticles-A Review. Trop. J. Pharm. Res. 2006, 5, 561–573. [Google Scholar] [CrossRef]
  94. Newton, A.M.J. Sukhjinder Kaur Solid Lipid Nanoparticles for Skin and Drug Delivery. In Nanoarchitectonics in Biomedicine; Elsevier: Amsterdam, The Netherlands, 2019; pp. 295–334. ISBN 9780128162002. [Google Scholar]
  95. Mandal, A.; Bisht, R.; Pal, D.; Mitra, A.K. Diagnosis and Drug Delivery to the Brain: Novel Strategies. In Emerging Nanotechnologies for Diagnostics, Drug Delivery and Medical Devices; Elsevier Inc.: Amsterdam, The Netherlands, 2017; pp. 59–83. ISBN 9780323429979. [Google Scholar]
  96. Mirchandani, Y.; Patravale, V.B.; Brijesh, S. Solid Lipid Nanoparticles for Hydrophilic Drugs. J. Control. Release 2021, 335, 457–464. [Google Scholar] [CrossRef] [PubMed]
  97. Wang, H.L.; Wang, Z.G.; Liu, S.L. Lipid Nanoparticles for MRNA Delivery to Enhance Cancer Immunotherapy. Molecules 2022, 27, 5607. [Google Scholar] [CrossRef] [PubMed]
  98. Jung, H.N.; Lee, S.Y.; Lee, S.; Youn, H.; Im, H.J. Lipid Nanoparticles for Delivery of RNA Therapeutics: Current Status and the Role of in Vivo Imaging. Theranostics 2022, 12, 7509–7531. [Google Scholar] [CrossRef]
  99. Albertsen, C.H.; Kulkarni, J.A.; Witzigmann, D.; Lind, M.; Petersson, K.; Simonsen, J.B. The Role of Lipid Components in Lipid Nanoparticles for Vaccines and Gene Therapy. Adv. Drug Deliv. Rev. 2022, 188, 114416. [Google Scholar] [CrossRef]
  100. Guo, D.; Dou, D.; Li, X.; Zhang, Q.; Bhutto, Z.A.; Wang, L. Ivermection-Loaded Solid Lipid Nanoparticles: Preparation, Characterisation, Stability and Transdermal Behaviour. Artif. Cells Nanomed. Biotechnol. 2018, 46, 255–262. [Google Scholar] [CrossRef]
  101. Badilli, U.; Gumustas, M.; Uslu, B.; Ozkan, S.A. Lipid-Based Nanoparticles for Dermal Drug Delivery. In Organic Materials as Smart Nanocarriers for Drug Delivery; Elsevier: Amsterdam, The Netherlands, 2018; pp. 369–413. ISBN 9780128136638. [Google Scholar]
  102. Duong, V.A.; Nguyen, T.T.L.; Maeng, H.J. Preparation of Solid Lipid Nanoparticles and Nanostructured Lipid Carriers for Drug Delivery and the Effects of Preparation Parameters of Solvent Injection Method. Molecules 2020, 25, 4781. [Google Scholar] [CrossRef]
  103. Sguizzato, M.; Subroto, E.; Andoyo, R.; Indiarto, R. Solid Lipid Nanoparticles: Review of the Current Research on Encapsulation and Delivery Systems for Active and Antioxidant Compounds. Antioxidants 2023, 12, 633. [Google Scholar] [CrossRef]
  104. López, K.L.; Ravasio, A.; González-Aramundiz, J.V.; Zacconi, F.C. Solid Lipid Nanoparticles (SLN) and Nanostructured Lipid Carriers (NLC) Prepared by Microwave and Ultrasound-Assisted Synthesis: Promising Green Strategies for the Nanoworld. Pharmaceutics 2023, 15, 1333. [Google Scholar] [CrossRef]
  105. Satapathy, M.K.; Yen, T.L.; Jan, J.S.; Tang, R.D.; Wang, J.Y.; Taliyan, R.; Yang, C.H. Solid Lipid Nanoparticles (Slns): An Advanced Drug Delivery System Targeting Brain through Bbb. Pharmaceutics 2021, 13, 1183. [Google Scholar] [CrossRef] [PubMed]
  106. Mukherjee, S.; Ray, S.; Thakur, S. Solid Lipid Nanoparticles: A Modern Formulation Approach in Drug Delivery System. Indian. J. Pharm. Sci. 2009, 71, 349–358. [Google Scholar] [CrossRef] [PubMed]
  107. Chauhan, I.; Yasir, M.; Verma, M.; Singh, A.P. Nanostructured Lipid Carriers: A Groundbreaking Approach for Transdermal Drug Delivery. Adv. Pharm. Bull. 2020, 10, 150–165. [Google Scholar] [CrossRef] [PubMed]
  108. Das, S.; Chaudhury, A. Recent Advances in Lipid Nanoparticle Formulations with Solid Matrix for Oral Drug Delivery. AAPS PharmSciTech 2011, 12, 62–76. [Google Scholar] [CrossRef]
  109. Scioli Montoto, S.; Muraca, G.; Ruiz, M.E. Solid Lipid Nanoparticles for Drug Delivery: Pharmacological and Biopharmaceutical Aspects. Front. Mol. Biosci. 2020, 7, 587997. [Google Scholar] [CrossRef]
  110. Lopalco, A.; Denora, N. Nanoformulations for Drug Delivery: Safety, Toxicity, and Efficacy. In Methods in Molecular Biology; Humana Press Inc.: Totowa, NJ, USA, 2018; Volume 1800, pp. 347–365. [Google Scholar]
  111. Sharma, S.; Parveen, R.; Chatterji, B.P. Toxicology of Nanoparticles in Drug Delivery. Curr. Pathobiol. Rep. 2021, 9, 133–144. [Google Scholar] [CrossRef] [PubMed]
  112. Huang, H.J.; Lee, Y.H.; Hsu, Y.H.; Liao, C.T.; Lin, Y.F.; Chiu, H.W. Current Strategies in Assessment of Nanotoxicity: Alternatives to in Vivo Animal Testing. Int. J. Mol. Sci. 2021, 22, 4216. [Google Scholar] [CrossRef]
  113. Savage, D.T.; Hilt, J.Z.; Dziubla, T.D. In Vitro Methods for Assessing Nanoparticle Toxicity. In Methods in Molecular Biology; Humana Press Inc.: Totowa, NJ, USA, 2019; Volume 1894, pp. 1–29. [Google Scholar]
  114. Chenthamara, D.; Subramaniam, S.; Ramakrishnan, S.G.; Krishnaswamy, S.; Essa, M.M.; Lin, F.H.; Qoronfleh, M.W. Therapeutic Efficacy of Nanoparticles and Routes of Administration. Biomater. Res. 2019, 23, 20. [Google Scholar] [CrossRef]
  115. Gupta, R.; Xie, H. Nanoparticles in Daily Life: Applications, Toxicity and Regulations. J. Environ. Pathol. Toxicol. Oncol. 2018, 37, 209–230. [Google Scholar] [CrossRef]
  116. Elsaesser, A.; Howard, C.V. Toxicology of Nanoparticles. Adv. Drug Deliv. Rev. 2012, 64, 129–137. [Google Scholar] [CrossRef]
  117. Sung, Y.K.; Kim, S.W. Recent Advances in the Development of Gene Delivery Systems. Biomater. Res. 2019, 23, 8. [Google Scholar] [CrossRef] [PubMed]
  118. Eshka, S.F.A.; Bahador, M.; Gordan, M.M.; Karbasi, S.; Tabar, Z.M.; Basiri, M. A Systematic Review of Gene Editing Clinical Trials. medRxiv 2022. [Google Scholar] [CrossRef]
  119. Kontogiannis, O.; Karalis, V. On the in Vivo Kinetics of Gene Delivery Vectors. medRxiv 2022. [Google Scholar] [CrossRef]
  120. Nayerossadat, N.; Ali, P.; Maedeh, T. Viral and Nonviral Delivery Systems for Gene Delivery. Adv. Biomed. Res. 2012, 1, 27. [Google Scholar] [CrossRef]
  121. Wang, D.; Tai, P.W.L.; Gao, G. Adeno-Associated Virus Vector as a Platform for Gene Therapy Delivery. Nat. Rev. Drug Discov. 2019, 18, 358–378. [Google Scholar] [CrossRef]
  122. Abinaya, R.V.; Viswanathan, P. Biotechnology-Based Therapeutics. In Translational Biotechnology; Elsevier: Amsterdam, The Netherlands, 2021; pp. 27–52. [Google Scholar]
  123. Trobridge, G.D. Foamy Virus Vectors for Gene Transfer. Expert. Opin. Biol. Ther. 2009, 9, 1427–1436. [Google Scholar] [CrossRef]
  124. Deregowski, V.; Canalis, E. Gene Delivery by Retroviruses. In Osteoporosis. Methods in Molecular Biology; Humana Press: Totowa, NJ, USA, 2008; Volume 455. [Google Scholar]
  125. Cavalieri, V.; Baiamonte, E.; Lo Iacono, M. Non-Primate Lentiviral Vectors and Their Applications in Gene Therapy for Ocular Disorders. Viruses 2018, 10, 316. [Google Scholar] [CrossRef]
  126. Worgall, S.; Crystal, R.G. Gene Therapy. In Principles of Tissue Engineerin, 4th ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2013; pp. 657–686. ISBN 9780123983589. [Google Scholar]
  127. Wong, L.-F.; Goodhead, L.; Kyriacos, C.P.; Mitrophanous, K.A.; Kingsman, S.M.; Mazarakis, N.D. Lentivirus-Mediated Gene Transfer to the Central Nervous: Therapeutic and Research Applications. Hum. Gene Ther. 2006, 17, 1–9. [Google Scholar] [CrossRef]
  128. Lee, C.S.; Bishop, E.S.; Zhang, R.; Yu, X.; Farina, E.M.; Yan, S.; Zhao, C.; Zeng, Z.; Shu, Y.; Wu, X.; et al. Adenovirus-Mediated Gene Delivery: Potential Applications for Gene and Cell-Based Therapies in the New Era of Personalized Medicine. Genes. Dis. 2017, 4, 43–63. [Google Scholar] [CrossRef]
  129. Kasala, D.; Yoon, A.R.; Hong, J.; Kim, S.W.; Yun, C.O. Evolving Lessons on Nanomaterial-Coated Viral Vectors for Local and Systemic Gene Therapy. Nanomedicine 2016, 11, 1689–1713. [Google Scholar] [CrossRef]
  130. Ahmadi, S.E.; Soleymani, M.; Shahriyary, F.; Amirzargar, M.R.; Ofoghi, M.; Fattahi, M.D.; Safa, M. Viral Vectors and Extracellular Vesicles: Innate Delivery Systems Utilized in CRISPR/Cas-Mediated Cancer Therapy. Cancer Gene Ther. 2023, 30, 936–954. [Google Scholar] [CrossRef]
  131. Zhang, H.; Wang, H.; An, Y.; Chen, Z. Construction and Application of Adenoviral Vectors. Mol. Ther. Nucleic Acids 2023, 34, 102027. [Google Scholar] [CrossRef] [PubMed]
  132. Vargas, J.E.; Chicaybam, L.; Stein, R.T.; Tanuri, A.; Delgado-Cañedo, A.; Bonamino, M.H. Retroviral Vectors and Transposons for Stable Gene Therapy: Advances, Current Challenges and Perspectives. J. Transl. Med. 2016, 14, 288. [Google Scholar] [CrossRef] [PubMed]
  133. Capasso, C.; Garofalo, M.; Hirvinen, M.; Cerullo, V. The Evolution of Adenoviral Vectors through Genetic and Chemical Surface Modifications. Viruses 2014, 6, 832–855. [Google Scholar] [CrossRef] [PubMed]
  134. Vandendriessche, T.; Naldini, L.; Collen, D.; Chuah, M.K.L. Oncoretroviral and Lentiviral Vector-Mediated Gene Therapy. Methods Enzym. 2002, 346, 573–589. [Google Scholar]
  135. Cevher, E.; Demir, A.; Sefik, E. Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy. In Recent Advances in Novel Drug Carrier Systems; InTech: London, UK, 2012. [Google Scholar]
  136. Travieso, T.; Li, J.; Mahesh, S.; Mello, J.D.F.R.E.; Blasi, M. The Use of Viral Vectors in Vaccine Development. NPJ Vaccines 2022, 7, 75. [Google Scholar] [CrossRef]
  137. Minnaert, A.K.; Vanluchene, H.; Verbeke, R.; Lentacker, I.; De Smedt, S.C.; Raemdonck, K.; Sanders, N.N.; Remaut, K. Strategies for Controlling the Innate Immune Activity of Conventional and Self-Amplifying MRNA Therapeutics: Getting the Message Across. Adv. Drug Deliv. Rev. 2021, 176, 113900. [Google Scholar] [CrossRef]
  138. Maruggi, G.; Zhang, C.; Li, J.; Ulmer, J.B.; Yu, D. MRNA as a Transformative Technology for Vaccine Development to Control Infectious Diseases. Mol. Ther. 2019, 27, 757–772. [Google Scholar] [CrossRef]
  139. Ballesteros-Briones, M.C.; Silva-Pilipich, N.; Herrador-Cañete, G.; Vanrell, L.; Smerdou, C. A New Generation of Vaccines Based on Alphavirus Self-Amplifying RNA. Curr. Opin. Virol. 2020, 44, 145–153. [Google Scholar] [CrossRef]
  140. Blom, D.J.; Marais, A.D.; Moodley, R.; van der Merwe, N.; van Tonder, A.; Raal, F.J. RNA-Based Therapy in the Management of Lipid Disorders: A Review. Lipids Health Dis. 2022, 21, 41. [Google Scholar] [CrossRef]
  141. Magadum, A.; Kaur, K.; Zangi, L. MRNA-Based Protein Replacement Therapy for the Heart. Mol. Ther. 2019, 27, 785–793. [Google Scholar] [CrossRef]
  142. Erasmus, J.H.; Khandhar, A.P.; O’Connor, M.A.; Walls, A.C.; Hemann, E.A.; Murapa, P.; Archer, J.; Leventhal, S.; Fuller, J.T.; Lewis, T.B.; et al. An Alphavirus-Derived Replicon RNA Vaccine Induces SARS-CoV-2 Neutralizing Antibody and T Cell Responses in Mice and Nonhuman Primates. Sci. Transl. Med. 2020, 12, eabc9396. [Google Scholar] [CrossRef] [PubMed]
  143. McCullough, K.C.; Bassi, I.; Milona, P.; Suter, R.; Thomann-Harwood, L.; Englezou, P.; Démoulins, T.; Ruggli, N. Self-Replicating Replicon-Rna Delivery to Dendritic Cells by Chitosan-Nanoparticles for Translation in Vitro and in Vivo. Mol. Ther. Nucleic Acids 2014, 3, e173. [Google Scholar] [CrossRef] [PubMed]
  144. Blakney, A.K.; McKay, P.F.; Yus, B.I.; Aldon, Y.; Shattock, R.J. Inside out: Optimization of Lipid Nanoparticle Formulations for Exterior Complexation and in Vivo Delivery of SaRNA. Gene Ther. 2019, 26, 363–372. [Google Scholar] [CrossRef] [PubMed]
  145. Feng, X.; Xu, W.; Li, Z.; Song, W.; Ding, J.; Chen, X. Immunomodulatory Nanosystems. Adv. Sci. 2019, 6, 1900101. [Google Scholar] [CrossRef]
  146. Machado, B.A.S.; Hodel, K.V.S.; Fonseca, L.M.D.S.; Mascarenhas, L.A.B.; Andrade, L.P.C.d.S.; Rocha, V.P.C.; Soares, M.B.P.; Berglund, P.; Duthie, M.S.; Reed, S.G.; et al. The Importance of Rna-Based Vaccines in the Fight against COVID-19: An Overview. Vaccines 2021, 9, 1345. [Google Scholar] [CrossRef]
  147. Wadhwa, A.; Aljabbari, A.; Lokras, A.; Foged, C.; Thakur, A. Opportunities and Challenges in the Delivery of Mrna-Based Vaccines. Pharmaceutics 2020, 12, 102. [Google Scholar] [CrossRef]
  148. Cullis, P.R.; Hope, M.J. Lipid Nanoparticle Systems for Enabling Gene Therapies. Mol. Ther. 2017, 25, 1467–1475. [Google Scholar] [CrossRef]
  149. Kowalski, P.S.; Rudra, A.; Miao, L.; Anderson, D.G. Delivering the Messenger: Advances in Technologies for Therapeutic MRNA Delivery. Mol. Ther. 2019, 27, 710–728. [Google Scholar] [CrossRef]
  150. Garcia, A.B.; Siu, E.; Sun, T.; Exler, V.; Brito, L.; Hekele, A.; Otten, G.; Augustijn, K.; Janse, C.J.; Ulmer, J.B.; et al. Neutralization of the Plasmodium-Encoded MIF Ortholog Confers Protective Immunity against Malaria Infection. Nat. Commun. 2018, 9, 2714. [Google Scholar] [CrossRef]
  151. Erasmus, J.H.; Archer, J.; Fuerte-Stone, J.; Khandhar, A.P.; Voigt, E.; Granger, B.; Bombardi, R.G.; Govero, J.; Tan, Q.; Durnell, L.A.; et al. Intramuscular Delivery of Replicon RNA Encoding ZIKV-117 Human Monoclonal Antibody Protects against Zika Virus Infection. Mol. Ther. Methods Clin. Dev. 2020, 18, 402–414. [Google Scholar] [CrossRef] [PubMed]
  152. Leventhal, S.S.; Meade-White, K.; Rao, D.; Haddock, E.; Leung, J.; Scott, D.; Archer, J.; Randall, S.; Erasmus, J.H.; Feldmann, H.; et al. Replicating RNA Vaccination Elicits an Unexpected Immune Response That Efficiently Protects Mice against Lethal Crimean-Congo Hemorrhagic Fever Virus Challenge. Lancet 2022, 82, 104188. [Google Scholar] [CrossRef] [PubMed]
  153. Fotoran, W.L.; da Silva, J.R.; Glitz, C.; Ferreira, L.C.d.S.; Wunderlich, G. Establishment of an Antiplasmodial Vaccine Based on PfRH5-Encoding RNA Replicons Stabilized by Cationic Liposomes. Pharmaceutics 2023, 15, 1223. [Google Scholar] [CrossRef] [PubMed]
  154. Goswami, R.; O’hagan, D.T.; Adamo, R.; Baudner, B.C. Conjugation of Mannans to Enhance the Potency of Liposome Nanoparticles for the Delivery of Rna Vaccines. Pharmaceutics 2021, 13, 240. [Google Scholar] [CrossRef] [PubMed]
  155. Chahal, J.S.; Khan, O.F.; Cooper, C.L.; McPartlan, J.S.; Tsosie, J.K.; Tilley, L.D.; Sidik, S.M.; Lourido, S.; Langer, R.; Bavari, S.; et al. Dendrimer-RNA Nanoparticles Generate Protective Immunity against Lethal Ebola, H1N1 Influenza, and Toxoplasma Gondii Challenges with a Single Dose. Proc. Natl. Acad. Sci. USA 2016, 113, E4133–E4142. [Google Scholar] [CrossRef] [PubMed]
  156. McCullough, K.C.; Milona, P.; Thomann-Harwood, L.; Démoulins, T.; Englezou, P.; Suter, R.; Ruggli, N. Self-Amplifying Replicon RNA Vaccine Delivery to Dendritic Cells by Synthetic Nanoparticles. Vaccines 2014, 2, 735–754. [Google Scholar] [CrossRef]
  157. Brito, L.A.; Chan, M.; Shaw, C.A.; Hekele, A.; Carsillo, T.; Schaefer, M.; Archer, J.; Seubert, A.; Otten, G.R.; Beard, C.W.; et al. A Cationic Nanoemulsion for the Delivery of Next-Generation RNA Vaccines. Mol. Ther. 2014, 22, 2118–2129. [Google Scholar] [CrossRef]
  158. Erasmus, J.H.; Khandhar, A.P.; Guderian, J.; Granger, B.; Archer, J.; Archer, M.; Gage, E.; Fuerte-Stone, J.; Larson, E.; Lin, S.; et al. A Nanostructured Lipid Carrier for Delivery of a Replicating Viral RNA Provides Single, Low-Dose Protection against Zika. Mol. Ther. 2018, 26, 2507–2522. [Google Scholar] [CrossRef]
  159. Crooke, S.T.; Witztum, J.L.; Bennett, C.F.; Baker, B.F. RNA-Targeted Therapeutics. Cell Metab. 2018, 27, 714–739. [Google Scholar] [CrossRef]
  160. Samaridou, E.; Heyes, J.; Lutwyche, P. Lipid Nanoparticles for Nucleic Acid Delivery: Current Perspectives. Adv. Drug Deliv. Rev. 2020, 154–155, 37–63. [Google Scholar] [CrossRef]
  161. Massaad-Massade, L.; Boutary, S.; Caillaud, M.; Gracia, C.; Parola, B.; Gnaouiya, S.B.; Stella, B.; Arpicco, S.; Buchy, E.; Desmaële, D.; et al. New Formulation for the Delivery of Oligonucleotides Using “Clickable” SiRNA-Polyisoprenoid-Conjugated Nanoparticles: Application to Cancers Harboring Fusion Oncogenes. Bioconjug Chem. 2018, 29, 1961–1972. [Google Scholar] [CrossRef] [PubMed]
  162. Guo, S.; Vieweger, M.; Zhang, K.; Yin, H.; Wang, H.; Li, X.; Li, S.; Hu, S.; Sparreboom, A.; Evers, B.M.; et al. Ultra-Thermostable RNA Nanoparticles for Solubilizing and High-Yield Loading of Paclitaxel for Breast Cancer Therapy. Nat. Commun. 2020, 11, 972. [Google Scholar] [CrossRef] [PubMed]
  163. Ansari, M.A.; Thiruvengadam, M.; Farooqui, Z.; Rajakumar, G.; Sajid Jamal, Q.M.; Alzohairy, M.A.; Almatroudi, A.; Alomary, M.N.; Chung, I.M.; Al-Suhaimi, E.A. Nanotechnology, in Silico and Endocrine-Based Strategy for Delivering Paclitaxel and MiRNA: Prospects for the Therapeutic Management of Breast Cancer. Semin. Cancer Biol. 2021, 69, 109–128. [Google Scholar] [CrossRef] [PubMed]
  164. Régina, A.; Demeule, M.; Ché, C.; Lavallée, I.; Poirier, J.; Gabathuler, R.; Béliveau, R.; Castaigne, J.P. Antitumour Activity of ANG1005, a Conjugate between Paclitaxel and the New Brain Delivery Vector Angiopep-2. Br. J. Pharmacol. 2008, 155, 185–197. [Google Scholar] [CrossRef] [PubMed]
  165. Ma, P.; Mumper, R.J. Paclitaxel Nano-Delivery Systems: A Comprehensive Review. J. Nanomed. Nanotechnol. 2013, 4, 6. [Google Scholar] [CrossRef]
  166. Song, Y.; Zhou, B.; Du, X.; Wang, Y.; Zhang, J.; Ai, Y.; Xia, Z.; Zhao, G. Folic Acid (FA)-Conjugated Mesoporous Silica Nanoparticles Combined with MRP-1 SiRNA Improves the Suppressive Effects of Myricetin on Non-Small Cell Lung Cancer (NSCLC). Biomed. Pharmacother. 2020, 125, 109561. [Google Scholar] [CrossRef]
  167. Urits, I.; Swanson, D.; Swett, M.C.; Patel, A.; Berardino, K.; Amgalan, A.; Berger, A.A.; Kassem, H.; Kaye, A.D.; Viswanath, O. A Review of Patisiran (ONPATTRO) for the Treatment of Polyneuropathy in People with Hereditary Transthyretin Amyloidosis. Neurol. Ther. 2020, 9, 301–315. [Google Scholar] [CrossRef]
  168. Zhang, X.; Goel, V.; Attarwala, H.; Sweetser, M.T.; Clausen, V.A.; Robbie, G.J. Patisiran Pharmacokinetics, Pharmacodynamics, and Exposure-Response Analyses in the Phase 3 APOLLO Trial in Patients With Hereditary Transthyretin-Mediated (HATTR) Amyloidosis. J. Clin. Pharmacol. 2020, 60, 37–49. [Google Scholar] [CrossRef]
  169. Majeed, C.N.; Ma, C.D.; Xiao, T.; Rudnick, S.; Bonkovsky, H.L. Spotlight on Givosiran as a Treatment Option for Adults with Acute Hepatic Porphyria: Design, Development, and Place in Therapy. Drug Des. Devel Ther. 2022, 16, 1827–1845. [Google Scholar] [CrossRef] [PubMed]
  170. Ventura, P.; Bonkovsky, H.L.; Gouya, L.; Aguilera-Peiró, P.; Montgomery Bissell, D.; Stein, P.E.; Balwani, M.; Anderson, D.K.E.; Parker, C.; Kuter, D.J.; et al. Efficacy and Safety of Givosiran for Acute Hepatic Porphyria: 24-Month Interim Analysis of the Randomized Phase 3 ENVISION Study. Liver Int. 2022, 42, 161–172. [Google Scholar] [CrossRef]
  171. Eygeris, Y.; Gupta, M.; Kim, J.; Sahay, G. Chemistry of Lipid Nanoparticles for RNA Delivery. Acc. Chem. Res. 2022, 55, 2–12. [Google Scholar] [CrossRef] [PubMed]
  172. Ho, W.; Gao, M.; Li, F.; Li, Z.; Zhang, X.Q.; Xu, X. Next-Generation Vaccines: Nanoparticle-Mediated DNA and MRNA Delivery. Adv. Healthc. Mater. 2021, 10, 2001812. [Google Scholar] [CrossRef]
  173. Schoenmaker, L.; Witzigmann, D.; Kulkarni, J.A.; Verbeke, R.; Kersten, G.; Jiskoot, W.; Crommelin, D.J.A. mRNA-Lipid Nanoparticle COVID-19 Vaccines: Structure and Stability. Int. J. Pharm. 2021, 601, 120586. [Google Scholar] [CrossRef] [PubMed]
  174. Revia, R.A.; Stephen, Z.R.; Zhang, M. Theranostic Nanoparticles for RNA-Based Cancer Treatment. Acc. Chem. Res. 2019, 52, 1496–1506. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of the use of nanoformulations associated with repRNA: (a) examples of nanoformulations and forms of association with repRNA; (b) immunomodulatory potential of nanoparticles associated with repRNA and therapeutic application, either by immunostimulation or immunodepression. The information contained in this illustration was based on the work of Blakney [144] and Feng et al. [145]. Created with BioRender.com (accessed on 2 October 2023).
Figure 1. Overview of the use of nanoformulations associated with repRNA: (a) examples of nanoformulations and forms of association with repRNA; (b) immunomodulatory potential of nanoparticles associated with repRNA and therapeutic application, either by immunostimulation or immunodepression. The information contained in this illustration was based on the work of Blakney [144] and Feng et al. [145]. Created with BioRender.com (accessed on 2 October 2023).
Pharmaceuticals 16 01634 g001
Table 1. Nanoformulations and their main components approved for therapeutic use by FDA and regulatory agencies.
Table 1. Nanoformulations and their main components approved for therapeutic use by FDA and regulatory agencies.
ClassificationName
(Trade Tame)
Main ComponentDelivery RouteIndicationApproval
(Year)
LiposomeAmBisomeLiposomal amphotericin BIntravenousFungal/protozoal infectionsFDA (1997)
EMA (2006)
ANVISA (1997)
Doxil/ CaelyxLiposomal doxorubicinIntravenousAntineoplastic agents (ovarian and breast cancer; multiple myeloma; Karposi’s Sarcoma)FDA (1995, 2005, 2008)
EMA (1996)
ANVISA (2011)
Myocet
(Myoce liposomal)
Liposomal doxorubicinIntravenousAntineoplastic agents (breast neoplasms)FDA (2000)
EMA (2000)
VisudyneLiposomal verteporfinIntravenousOphthalmic agents (myopia; ocular histoplasmosis; macular degeneration, wet age-related)FDA (2000)
ANVISA (2004)
EMA (2007)
MarqiboLiposomal vincristineIntravenousAntineoplastic agents (hematologic malignancies and solid tumors)FDA (2012)
Onivyde
(Onivyde pegylated liposomal)
Liposomal irinotecanIntravenousAntineoplastic agents (pancreatic cancer)FDA (2015)
EMA (2016)
Polymer-based nanoparticlesEligardLeuprolide acetate and polymer (PLGH (poly (DL-Lactide-co-glycolide)))SubcutaneousAntineoplastic agents (prostate cancer)FDA (2002)
ANVISA (2006)
MirceraMethoxy polyethylene glycol-epoetin betaSubcutaneous/ IntravenousAnemia associated with chronic kidney diseaseFDA (2007)
EMA (2007)
ANVISA (2008)
CimziaPEGylated antibody fragment (Certolizumab)SubcutaneousAnti-inflammatory action (Crohn’s disease;
rheumatoid arthritis; psoriatic arthritis; ankylosing spondylitis)
FDA (2008, 2009, 2013)
EMA (2009)
ANVISA (2017)
PegIntronPEGylated IFN alpha-2b proteinSubcutaneousImmunomodulator
(hepatitis C)
FDA (2001)
EMA (2000)
ANVISA (2011)
Magnetic nanoparticlesNanoThermIron oxide coated with amino silaneIntratumoral injectionAntineoplastic agents (glioblastoma)FDA (2010)
EMA (2013)
FerahemeIron oxide and a polyglucose sorbitol carboxymethyetherIntravenousTreatment of anemiaFDA (2009)
EMA (2012)
Lipid nanoparticlePatisiran (Onpattro)Phospholipids, cholesterol, ionizable cationic lipid (DLin-MC3-DMA), and polyethylene glycol-modified lipidIntravenousPolyneuropathyFDA (2018)
Table 2. Application of repRNA associated with a nanocarrier for the therapy of pathologies.
Table 2. Application of repRNA associated with a nanocarrier for the therapy of pathologies.
CarrierRNA RepliconResultsReference
Cationic nanocarrierRepRNA PMIF (macrophage migration inhibitory factor and cytokine).It improved host cellular and humoral immunity against Plasmodium infection in the liver and blood and conferred complete protection against malaria reinfection in murine mice.[150]
Nanostructured lipid transporters (NLCs)RepRNA ZIKV-117 mAb.Rapid protection against Zika virus infection in mice.[151]
Lipid InOrganic Nanoparticles (LION)LION/repRNA-CoV2SLION/repRNA-CoV2S vaccine intramuscularly to mice, a significant amount of anti-SARS-CoV-2 S protein IgG antibody isotypes, resembling a Type 1 T helper cell response, were produced.[142]
Cationic nanocarrierRepRNA CCHFV (Crimean–Congo hemorrhagic fever virus) encoding NP (nucleoprotein), GPC (glycoprotein precursor) or bothIt provided robust protection against Crimean–Congo hemorrhagic fever virus in lethal mice.[152]
Cationic liposomessamPfRH5 replicon (Plasmodium falciparum reticulocyte binding protein homologue 5)The liposome–replicon complexes showed high transfection efficiencies. They elicited antibodies capable of inhibiting the growth of the parasite in vitro[153]
Mannosylation of lipid nanoparticles (LNPs)Self-amplifying mRNA encoded an influenza (hemagglutinin)Compared to LNPs, mannnosylated lipid nanoparticles (MLNPs) showed higher levels of IgG1 and IgG2a.[154]
Polymeric nanoparticle Nanoparticle (MDNP)-delivered VEEV replicon RNA encoding the hemagglutinin protein (HA) of an H1N1 influenza virus (A/WSN/33) or the Ebola virus (EBOV) glycoprotein (GP)The vaccine elicits both CD8+ T-cell and antibody responses and can be created with numerous antigen-expressing replicons.[155]
Table 3. Applications of RNA conjugated with nanoparticles used as therapy for diseases.
Table 3. Applications of RNA conjugated with nanoparticles used as therapy for diseases.
TechnologyApplicationType of NanocarrierReferences
siRNA-SQ and siRNA-SOLACancers Harboring Fusion OncogenesPolyisoprenoid chains[161]
RNA-paclitaxelBreast cancer treatmentLipidic[162,163,164,165]
MRP-1 siRNANon-small cell lung cancerMesoporous silica[166]
ALN-18328 (Patisiran)Transthyretin amyloidosis (ATTR)Lipid nanoparticle[167,168]
GalNAc-siRNA (Givosiran)Acute hepatic porphyriaLipid nanoparticle[169,170]
siRNA-LNPVaccine for COVID-19Lipid nanoparticle
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lima, E.S.; dos Santos, D.; Souza, A.L.; Macedo, M.E.; Bandeira, M.E.; Junior, S.S.S.; Fiuza, B.S.D.; Rocha, V.P.C.; dos Santos Fonseca, L.M.; Nunes, D.D.G.; et al. RNA Combined with Nanoformulation to Advance Therapeutic Technologies. Pharmaceuticals 2023, 16, 1634. https://doi.org/10.3390/ph16121634

AMA Style

Lima ES, dos Santos D, Souza AL, Macedo ME, Bandeira ME, Junior SSS, Fiuza BSD, Rocha VPC, dos Santos Fonseca LM, Nunes DDG, et al. RNA Combined with Nanoformulation to Advance Therapeutic Technologies. Pharmaceuticals. 2023; 16(12):1634. https://doi.org/10.3390/ph16121634

Chicago/Turabian Style

Lima, Eduarda Santos, Déborah dos Santos, Atena Liriel Souza, Maria Eduarda Macedo, Mariana Evangelista Bandeira, Sérgio Santos Silva Junior, Bianca Sampaio Dotto Fiuza, Vinicius Pinto Costa Rocha, Larissa Moraes dos Santos Fonseca, Danielle Devequi Gomes Nunes, and et al. 2023. "RNA Combined with Nanoformulation to Advance Therapeutic Technologies" Pharmaceuticals 16, no. 12: 1634. https://doi.org/10.3390/ph16121634

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

Article Metrics

Back to TopTop