ArticlePDF Available

Abstract and Figures

Nucleic acid (NA) based therapeutics have witnessed tremendous progress and breakthroughs in treating pathological conditions, including viral infections, neurological disorders, genetic diseases, and metabolic disorders. NAs such as plasmid DNA (pDNA), short interfering RNA (siRNA), microRNA (miRNA), and antisense oligonucleotides (ASOs) can be modified to revolutionize personalized medicine. Despite the great potential of NA-based therapeutics, their clinical transformation is significantly hampered by instability, degradation, and inefficient delivery to the targeted site in the in vivo system. Lipid-based delivery systems hold great potential to overcome these shortcomings to enhance the delivery and bioavailability, improve stability, and increase the therapeutic effect of the NAs by delivering them to the active site. This review emphasized various nucleic acid-based therapeutics and their enhanced and improved delivery using different nanocarriers. Ultimately, the importance of lipid-based nanocarriers for delivering NAs is discussed and provides perspective in this field.
This content is subject to copyright. Terms and conditions apply.
RSC
Pharmaceutics
REVIEW
Cite this: RSC Pharm., 2024, 1,645
Received 16th January 2024,
Accepted 5th August 2024
DOI: 10.1039/d4pm00009a
rsc.li/RSCPharma
Recent progress on nanosystems for nucleic acid
delivery
Shanka Walia
a
and Mohit J. Mehta *
b,c,d
Nucleic acid (NA) based therapeutics have witnessed tremendous progress and breakthroughs in treating
pathological conditions, including viral infections, neurological disorders, genetic diseases, and metabolic
disorders. NAs such as plasmid DNA (pDNA), short interfering RNA (siRNA), microRNA (miRNA), and anti-
sense oligonucleotides (ASOs) can be modied to revolutionize personalized medicine. Despite the great
potential of NA-based therapeutics, their clinical transformation is signicantly hampered by instability,
degradation, and inecient delivery to the targeted site in the in vivo system. Lipid-based delivery systems
hold great potential to overcome these shortcomings to enhance the delivery and bioavailability, improve
stability, and increase the therapeutic eect of the NAs by delivering them to the active site. This review
emphasized various nucleic acid-based therapeutics and their enhanced and improved delivery using
dierent nanocarriers. Ultimately, the importance of lipid-based nanocarriers for delivering NAs is dis-
cussed and provides perspective in this eld.
1. Introduction
Nucleic acids (NAs) are immensely recognized for their thera-
peutic potential during the COVID-19 pandemic. NAs, includ-
ing DNA, antisense oligonucleotides (ASO), aptamers, short
interfering RNA (siRNA), microRNA (miRNA), and messenger
RNA (mRNA), provide a highly versatile platform at the under-
lying level of transcription and translation for the treatment
of diseases as compared with traditional drugs.
1,2
Nucleic
acidshigh specificity and pharmacological eect oered un-
precedented opportunities to combat viral infections and
complicated diseases, including cancer, diabetes, cardio-
vascular diseases, genetic disorders, and acquired con-
ditions.
3
The majority of the NAs bind selectively to their
target molecules via complementary WatsonCrick base
pairing, making target molecule detection simple.
4
NAs are
good candidates for the personalized medicinal approach
because genes and their mutations can be easily questioned.
Hence, NAs can target patient-specific genes with minimal
o-target eects.
5
In contrast, pharmaceutical drug mole-
cules require extensive screening and medicinal chemical
optimization.
However, the intracellular uptake of NAs is quite challen-
ging because of their anionic nature and poor stability, which
makes the intracellular uptake of NAs dicult. Due to their
highly hydrophilic and polyvalent anionic properties, NAs are
degraded by extracellular enzymes and, thus, poorly absorbed
by cells. When injected intravenously, they tend to accumulate
in the liver and kidneys.
6
All these factors reduced the
eciency of the delivered cargo to elicit the desired response.
7
Furthermore, due to their high molecular weight, negative zeta
potential, and hydrophilicity, NAs cannot penetrate the cellular
membrane. However, clinical application is the primary goal
of any therapeutic molecule; hence, nanovehicles should
ensure adequate delivery of drugs to the targeted cell or organ
without causing any harmful eect on the healthy cells. The
therapeutic payload is delivered to the targeted cells via two
main approaches, namely, passive targeting and active target-
ing. The foundation of passive targeting is the enhanced per-
meability and retention (EPR) eect. For example, tumor vas-
culature is often leaky compared with healthy blood vessels.
This helps nanoparticles to penetrate the cancer cells more
eciently.
8
In contrast to this, active targeting facilitates
surface modifications of nanovehicles with specific ligands,
resulting in enhanced binding to the receptors of targeted
cells. Presently, a structure capable of improving the bio-
ecacy and controlled release of NAs at the desired site is of
the utmost importance. Nucleic acids possess versatile
physicochemical properties and tunability. These features
allow them to be easily functionalized with various bio-
a
Swami Sahajanand College of Computer Science, Plot No. 639, ISCON Mega City,
Gujarat, 364002, India
b
Nanomedicine Lab, Department of Biological Sciences and Bioengineering, Inha
University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
c
Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere
University, Tampere, 33520, Finland
d
Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere
University Hospital, Tampere, 33520, Finland. E-mail: mohitjmehta@gmail.com,
mohit.mehta@tuni.fi
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 645
molecules and nanoparticles (NPs) for enhanced therapeutic
eect with selective binding to the targeted molecules.
Dierent nanoparticle-based platforms such as metal NPs,
polymers, dendrimers, proteins, lipids, and other bio-
molecules are widely exploited in conjugation with NAs.
9,10
For
instance, liposomes and micelles, which have now received
FDA approval, were part of the first generation of nanoparticle-
based therapy.
11
Lipid-based nanocarriers were introduced as
promising tools to retain the structural integrity and thera-
peutic potential of NAs. Lipid-based nanocarriers oer a versa-
tile platform for NA encapsulation, which resulted in the clini-
cal translation of several NA therapeutics. This review aims to
provide an in-depth discussion on various nanosystems for
nucleic acid delivery and therapeutics. They oer a powerful
approach to address the limitations of traditional treatments,
and hence can potentially overcome these challenges.
2. Nucleic acid-based therapeutics
In the last decade, nucleic acid-based therapies have emerged
as promising approaches for treating multiple disorders by
regulating molecular pathways.
12
NAs target disease-causing
genes in a specific manner, enabling precise and personalized
treatment for life-threatening diseases.
13
NA-based thera-
peutics knock down, upregulate, or alter targeted gene
expression. Currently, several NA-based therapeutics are used
for the treatment of severe ailments (Table 1).
2.1 Plasmids
Plasmids are circular, double-stranded, high molecular weight
(<1000 to >200 000 bp) DNA constructs. They are often used as
vectors to introduce a foreign gene into a target cell for various
purposes such as gene therapy, vaccine production, and
protein expression.
14,15
The plasmid backbone contains the
gene for antibiotic resistance controlled by a prokaryotic pro-
moter (a prokaryotic origin of replication for plasmid replica-
tion),
14
and an expression cassette with a promoter to initiate
the transcription of a transgene that encodes a therapeutic
protein.
15
Plasmid DNA molecules typically contain several
regulatory signals, such as promoter and enhancer sequences,
responsible for regulating gene expression.
14
Promoters (cyto-
megalovirus (CMV) and Rous sarcoma virus or human alpha-
actin, human beta-actin promoter) commence gene transcrip-
tion by providing a recognition site for the RNA polymerase.
Enhancers play a vital role in the large-scale production of a
gene with specificity. They are localized either downstream or
upstream from the promoter site. Transcription eciency can
be elevated by selecting the promoter and enhancer of interest;
for example incorporating SV40 enhancer in the expression
plasmid enhanced muscle-specific gene expression.
26
Plasmids also play an essential role in generating vaccines for
genetic immunization. Plasmid-derived vaccines are easy to
produce if a genetic sequence of the concerned variant is
identified, and are also known to control the infection.
27
Pardridge et al. (2020) developed a plasmid DNA approach for
the treatment of NiemannPick C1 (NPC1), a lysosomal chole-
sterol storage disorder aecting the brain.
28
Plasmid DNA was
encapsulated in pegylated liposomes encoding the functional
human NPC1 gene. Quantitative PCR has shown the successful
delivery of pDNA and NPC1 mRNA expression in the brain,
liver, and spleen. Malardo and coworkers (2012) demonstrated
that injecting a low dose of pDNA (pcDNA3) in the Wistar rat
endotoxemia model has increased plasma vasopressin and
regulates blood pressure.
29
The rats treated with 1020 µg
pDNA showed lower levels of inflammatory cytokines, namely,
IL-6 and TNF-α.
2.2 Antisense oligonucleotide (ASO)
Antisense oligonucleotides (ASOs) of therapeutic importance
comprise single-stranded nucleic acids and are 1830 base
pairs in length.
30
ASOs form complementary base pairs with
their target RNA by classical Watson and Crick base pairing.
After binding with the target RNA, ASOs resulted in gene silen-
cing by altering or degrading the expression of the target RNA
via cleavage or blockage.
16,17
The various mechanisms adopted
by ASOs to degrade target RNA include (1) modified splicing
by skipping and inclusion of exons, and inhibition of 5cap
formation, (2) steric blockage of ribosomal functions as they
inhibit the binding of target RNA with ribosomes through the
translation arrest of the target RNA, and (3) induction of
RNAse H that recognizes ASORNA hybrids and degrade the
target RNA present in the hybrid.
31,32
ASOs are primarily admi-
nistered via transfection and transduction in vitro and
in vivo.
33
For example, Passini et al. (2011) used 2-O-2-methox-
yethyl-modified ASOs for the treatment of severe spinal muscu-
lar atrophy (SMA).
34
SMA is an autosomal recessive neuromus-
cular disorder related to the deficiency of survival motor
neuron (SMN) protein caused by mutations in the SMN1 gene.
ASO injection resulted in increased levels of SMN protein in
Table 1 Applications of dierent types of nucleic acid in clinical research
S. no. Nucleic acid Biomedical application Ref.
1 Plasmid DNA Gene cloning, gene therapy, DNA vaccine, protein expression, gene editing 14 and 15
2 Antisense oligonucleotides (ASOs) Targeting RNA functioning of genetic disorders, gene silencing 16 and 17
3 Aptamers Cell tracking, bacterial and viral protein sensing 18
4 siRNA and miRNA Gene therapy, antiviral therapy, drug discovery 1921
Biomarkers, cancer treatment
5 mRNA Vaccine production, cancer immunotherapy, gene editing 22 and 23
6 CpG DNA Immunotherapy, vaccine adjuvant, gene therapy, anti-inflammatory 24 and 25
Review RSC Pharmaceutics
646 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
SMA-infected mice. In vivo studies suggested improved thera-
peutic eects on muscle physiology, functioning, and the sur-
vival rate of motor neurons. Some examples of FDA-approved
ASO-based drugs include eteplirsen, golodirsen, and nusiner-
sen.
4
Several others are under clinical trials for the treatment
of neurological disorders, hepatitis B virus infections, solid
tumors, and renal diseases.
35
Although ASOs might have shown enhanced ecacy in vitro
and in vivo, they showed drawbacks related to low cellular
uptake and poor stability in body fluids that hindered their
potential from bench to bedside.
2.3 Aptamers
Aptamers are short single-stranded synthetic oligonucleotides
of 10100 nucleotides folded into three-dimensional
shapes.
36,37
Aptamers possess a remarkable tendency to bind
non-covalently to their target with high selectivity and speci-
ficity. Aptamers are widely explored to target biomolecules of
interest, including proteins, peptides, carbohydrates, anti-
bodies, small molecules, toxins, live cells, and even heavy
metals.
18
Thus, aptamers are potential candidates for cell
tracking, bacterial and viral protein sensing, medicine, and
analytical chemistry. Aptamers are primarily generated in vitro
from the systematic evolution of ligands by exponential enrich-
ment (SELEX).
38
Aptamers are often compared with antibodies
because of their similar function in binding the proteins with
specificity. Unfortunately, antibodies are fraught with signifi-
cant shortcomings, such as limited and unreproducible syn-
thesis, being expensive, and being prone to generating immu-
nogenicity. However, aptamers with versatile properties have
gained much attention in the research community since their
discovery in 1990.
39,40
Aptamers are a fascinating alternative to
conventional antibodies due to their facile and reproducible
production, small size, reduced immunogenic eect, physico-
chemical stability, longer shelf life, and in vitro chemical
synthesis.
41
2.4 Small interfering RNA and microRNA
Small interfering RNA (siRNA) and microRNA (miRNAs) are
representative modulators of the interference RNA (RNAi)
mechanism. RNAi comprises a group of agents that use
double-stranded RNA containing homologous sequences
complementary to the target gene and perform sequence-
specific gene silencing.
19
The primary function of RNAi is to
build a robust defense mechanism for protecting the genome
from mobile genetic material released from viruses, which, on
activation, produce abnormal dsRNA or RNA.
42
For example,
stable RNAi nanocomplexes with redox-sensitive glycol chito-
san derivatives were synthesized based on rolling circle tran-
scription. This nanocomplex ensured systemic and targeted
siRNA delivery with enhanced therapeutic eects in vivo via
the EPR eect.
43
A nanomedicine platform based on AuNPs co-
valently functionalized with siRNA duplexes was eciently
used to neutralize oncogene expression in glioblastoma multi-
forme.
44
In vivo studies indicated that the AuNPsiRNA nano-
complex penetrates the bloodbrain barrier. AuNPsiRNA
nanocomplex eectively knocked down endogenous
Bcl2L12 mRNA and protein levels and sensitized glioma cells
toward therapy-induced apoptosis by targeting the oncoprotein
Bcl2L12. Elbashir and coworkers (2001), demonstrated that 21-
nucleotide siRNA duplexes mediated the knockdown of
endogenous and heterologous genes in human embryonic
kidney (293) and HeLa cells.
45
In 2018, the FDA approved the
first siRNA-based drug, patisiran (Onpattro), to treat familial
amyloid polyneuropathy.
46
Other siRNA-based medications
approved by the FDA for clinical use include givosiran (to treat
acute hepatic porphyria), lumasiran (for the treatment of
primary hyperoxaluria type 1), and inclisiran (to treat athero-
sclerotic cardiovascular disease).
47
Vir Biotechnology and
Alnylam Pharmaceuticals recently developed ALN-COV (VIR
2703) based on siRNA therapeutics to cure SARS-CoV and
SARS-CoV-2 infections.
20
microRNA (miRNA) is an attractive therapeutic tool, particu-
larly in cancer treatment. miRNA interacts with the 3or 5
untranslated region of the targeted mRNA, resulting in degra-
dation and translational suppression.
21
Li et al. (2021) reported
intracellular miRNA imaging and gene silencing using a let-7a
miRNA-activated DNA nanomachine.
48
Multifunctional
miRNA-515 sponge-loaded magnetic nanodroplets combined
with ultrasound and magnetism were used to treat hepatocel-
lular carcinoma (HCC).
49
In vivo, studies showed the suppres-
sion of xenograft HCC because miRNA-515 upregulated the
expression of anti-oncogenes, namely CD22, P21, TIMP1,
NFKB, and E-cadherin in cancerous cells. However, no miRNA
drug is available and approved by the FDA for therapeutic pur-
poses. Most of the miRNA drugs are still under clinical trials;
for example, miravirsen (miR-122 inhibitor) for treating hepa-
titis (Hep) C has completed Phase II clinical trials. Similarly,
lademirsen, or RG-012 (miR-21 inhibitor), is in Phase II clini-
cal trials for treating Alport syndrome (NCT02855268).
MRG-110 has completed phase I clinical trials, and further
studies are underway to treat impaired wounds. MRG-110
(miRNA-92a inhibitor) improved wound healing in preclinical
models and can eectively treat impaired wound healing con-
ditions in diabetic patients. In short, several miRNA drugs are
under preclinical and clinical trials to treat various disorders.
2.5 Messenger RNA (mRNA)
Messenger RNA (mRNA) is a type of genetic material that con-
tains the information for producing proteins from DNA, the
genetic code found in the nucleus of a cell.
50
In recent years,
scientists have investigated the use of mRNA as a therapeutic
agent, specifically in the medical field. mRNA therapeutics,
commonly termed mRNA vaccines or mRNA drugs, are
designed from synthetic or modified mRNA molecules to
stimulate the production of specific proteins in the body to
treat diseases.
51
The Pfizer-BioNTech and Moderna COVID-19
vaccines, which employ mRNA to guide cells to generate the
spike protein present on the surface of the SARS-CoV-2 virus,
are among the most noteworthy milestones of mRNA thera-
pies.
22
The immune system then recognizes these spike pro-
teins as foreign moieties and stimulates an immune response
RSC Pharmaceutics Review
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 647
against the virus. This method has proved to be ecient in
preventing critical illness and death from COVID-19, and regu-
latory agencies around the globe have permitted its emergency
usage.
51,52
Additionally, mRNA therapies can cure many ailments,
including cancer, genetic abnormalities, and viral
infections.
23,53
For instance, mRNA is used in cancer immu-
notherapy to engineer T cells and natural killer (NK) cells with
antigen receptors and as a template for immunologically active
proteins in various immune and non-immune cells.
23
mRNA
therapeutics also have the potential to treat genetic disorders
by providing cells with functional copies of genes that are
missing or non-functional. Researchers have already demon-
strated the ability to use mRNA to correct genetic mutations in
animal models of certain diseases, such as inherited retinal
diseases. They are working on advancing these therapies to
clinical trials.
54
mRNA therapeutics have several advantages
over traditional drugs. Because mRNA does not integrate into
the genome, it does not have the potential to cause permanent
genetic changes.
55
Moreover, mRNA can be easily synthesized,
thus allowing the rapid development and production of new
therapeutics. Despite these advantages, some challenges still
need to be overcome to fully realize the potential of mRNA
therapeutics. One of the main challenges is ensuring mRNAs
safe and eective delivery to the targeted cells. Researchers are
developing various delivery methods, such as NPs and viral
vectors, to overcome this challenge.
56
Furthermore, mRNA
therapeutics are relatively new, and more research is needed to
fully understand their safety and ecacy.
55
Overall, mRNA
therapeutics represent a promising new approach to treating
and preventing various diseases and conditions. With contin-
ued research and development, mRNA therapeutics have the
potential to transform the way we think about medicine and
improve the lives of millions of people worldwide.
2.6 Cytosine-phosphate-guanine (CpG) DNA
Cytosine-phosphate-guanine (CpG) is a synthetic oligo-
nucleotide studied as a therapeutic agent for several undrug-
gable conditions. CpG activates the immune system by
binding to Toll-like receptor 9 (TLR9), expressed in immune
cells such as dendritic and B cells.
57
Consequently, these cells
are activated, and an immunological response is initiated.
Preclinical investigations demonstrated that CpG has anti-
tumor properties and enhances the immune response to
cancer cells, increasing tumor cell death.
24,58
It has been
reported that CpG has anti-inflammatory properties, making it
a suitable candidate for treating autoimmune illnesses such as
rheumatoid arthritis and multiple sclerosis.
59
CpG has been
shown to enhance the immune response to viral infections,
including influenza and herpes simplex virus, HIV,
60,61
hepa-
titis B and C,
62,63
and bacterial infections such as Streptococcus
pneumoniae.
25,64,65
In addition, CpG has been studied as a
potential treatment for allergies. In preclinical studies, CpG
has been found to reduce the severity of allergic reactions and
is currently being investigated as a treatment for allergies such
as asthma and allergic rhinitis.
6668
Overall, CpG is a promis-
ing therapeutic agent with many potential applications.
However, additional research is required to completely com-
prehend its mechanism of action and determine its human
safety and ecacy.
3. Classication of spherical
particles/nanoparticles for potential
complexation with nucleic acids
The development of NPs provides diverse platforms for deliver-
ing drugs with enhanced eects. However, it is reported that
NP morphology (viz., shape and size) can directly aect their
response in biological systems.
69
Amidst them, spherical NPs
such as metallic NPs, polymeric NPs, protein, nucleic acids,
and lipid-based complexes having a shape in common are the
most promising drug delivery agents and therapeutic probes.
It has been reported that under in vivo conditions, the NPs
behave dierently from biomolecules in therapeutic appli-
cations, particularly in vaccine delivery.
70
(1) The larger surface
area to volume ratio and permeability of these nanocomplexes
allow ecient and targeted delivery of the cargo (DNA/RNA
etc.) and reduce unwanted side eects.
71
(2) Nanocarriers can
be tailored to slowly release loaded their cargo, providing con-
trolled and sustained release within the targeted tissue or
organ. (3) In biological systems, small molecules face chal-
lenges such as degradation by enzymes and other biological
processes. Nanostructures can encapsulate the therapeutic
molecules, protecting them from degradation and ensuring
their eective and targeted delivery. This is especially impor-
tant for fragile molecules like DNA and RNA used in nucleic
acid vaccines.
72
(4) Naked DNA or RNA can sometimes trigger
unwanted immune responses. Nanocarriers can shield the
nucleic acids, overcoming such reactions and promoting a
more focused immune response toward the target antigen.
73,74
Overall, under in vivo conditions, nanostructures provide dis-
tinct advantages over small molecules. Their ability to deliver
drugs directly to target sites, oer controlled release, and
protect sensitive molecules makes them a powerful tool for
improved treatment ecacy and reduced side eects.
Table 2 shows the merits and demerits of these drug deliv-
ery vehicles. One can choose the ideal carrier to deliver par-
ticular drugs/genes based on these advantages and disadvan-
tages. A perfect carrier transports the drug to its target site and
releases it at the site of action. Several conditions must be
addressed, including specific and selective interactions,
sucient drug delivery, and sustained drug release at the tar-
geted area. For example, nanoparticles can be made from
various materials to encapsulate hydrophobic and hydrophilic
drugs with increased stability. Similarly, polymeric NPs can be
used to encapsulate hydrophobic and hydrophilic drugs and
can be designed to release drugs in a controlled manner over
time. Lipid-based drug delivery agents can encapsulate hydro-
phobic drugs, protecting them from degradation and helping
them reach the targeted area in the body.
Review RSC Pharmaceutics
648 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
Table 2 Merits and demerits of advanced delivery systems
S.
no Delivery agent Advantages Disadvantages Ref.
1 Inorganic NPs Ease of synthesis and surface functionalization Intracellular toxicity 75
Enhanced intracellular uptake Rapid elimination by the
reticuloendothelial system
Able to entrap both hydrophilic and hydrophobic drugs Lack of clinical trials
Enabled active and passive targeting Poor biodegradability
Photoluminescence properties May trigger the immune system
Stability (w.r.t. wide range of pH and temperatures)
Nobel metal (AuNPs,
AgNPs) NPs
Strong biocompatibility Expensive for large-scale applications 76
Tunable optical properties Potential toxicity concern
High surface-to-volume ratio Stability issues due to aggregation at
elevated temperaturesHigh binding anity
Quantum dots (QDs) High quantum yield High cytotoxicity 77
Resistance to photobleaching as compared with
traditional organic dyes
Exponential decrease in fluorescence and
blinking of dierent QDs
Size-dependent emission wavelength from visible to NIR
region
Instability and elevated hydrodynamic
diameter on interaction with serum
proteins
Silicon NPs (SiNPs) Good biocompatibility and biodegradable Low quantum yield 78
Tunable optical properties Potential toxicity concerns
Ease of functionalization
Thermal stability
Carbon nanomaterials
(CMs)
Tunable photoluminescence Toxicity concerns due to ROS generation,
inflammation, DNA damage 79
Photostability Limited understanding of the long-term
eect on human health
Ease of fabrication Limited control over size and structure
Economical
Eco-friendly, and biocompatible
Iron oxide NPs Biocompatibility Toxicity concerns 80
Magnetic properties Stability issues due to aggregation
High surface-to-volume ratio Long-term eects
Tunable properties Costly production
2 Polymeric NPs Biocompatible and biodegradable Burst eect 8183
Able to entrap both hydrophilic and hydrophobic drugs Limited drug loading
Ease of surface functionalization Deep knowledge of polymerreceptor
molecular interactions required
Controlled and sustained release Limited knowledge about long-term side
eectsProtect the drug from metabolic degradation
Prolonged residence time
3 Dendrimers Increasing solubility of highly lipophilic drugs Not a suitable candidate carrier for
hydrophilic drugs 84
Tunable physicochemical properties Cellular toxicity
Ease of surface functionalization for targeted drug
delivery
Elimination and metabolism depend on
the generation and materials
Covalently associating drugs High cost for their synthesis
4 Nanofibers Large surface area Toxicity 85 and
86Biocompatibility Degradation
Encapsulation and targeted delivery Expensive for large-scale production
Tissue engineering
5 Proteins/peptides Various anticancer eects Size influenced pharmacokinetics 83
High cell permeability Short half-life
Low systemic toxicity
Improved target selectivity
Bypass biological barriers
6 SNAs High specificity and potency Low yield and high cost of synthesis 87 and
88High drug loading eciency Low stability w.r.t. temperature and UV
exposure
Biocompatibility and prolonged circulation time Limited targeting ability
Dense packing and delivery of therapeutics Limited knowledge about their behavior
in in vivo systemsResistant to nuclear degradation
Eective gene regulation
7 Liposomes Biocompatible and biodegradable Poor stability 83
Able to entrap both hydrophilic and hydrophobic drugs Short shelf life and instability in
circulation
Controlled release A special storage system is needed
Protect the drug from metabolic degradation. Prolonged
circulation time
Batch-to-batch variation in the size of
liposomes
Low systemic toxicity
RSC Pharmaceutics Review
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 649
3.1 Inorganic NPs
Inorganic NPs are vital in biomedical applications, viz., bio-
imaging, therapy, diagnosis, and drug delivery.
89,90
Inorganic
NPs are mainly metal oxides, semiconductors, and noble
metals with sizes ranging from 1100 nm. These NPs pos-
sessed intrinsic physical, optical, magnetic, and electrical pro-
perties due to the characteristics of the core material.
Additionally, these properties can be controlled by tailoring
the size, shape, structure, and composition to achieve
enhanced sensing and therapeutic eects.
91
NPs gain positive
responses as nanocarriers because of their tunable properties,
improved ecacy, and decreased side eects by boosting their
targeting ability.
92,93
The loading can be done via four
methods: physical adsorption, electrostatic interaction, encap-
sulation inside the NP core, or covalent binding. For example,
gold NPs (AuNPs) are well known for their therapeutic and
delivery capabilities due to their stability, inertness, high
binding anity to thiols, amines, and disulfides, and ease of
functionalization with ligands such as drugs, proteins, and
nucleic acids. Excellent fluorescence due to the surface
plasmon resonance eect resulted in absorption in the visible
and NIR regions. Therefore, it is essential in diagnosis and
sensing applications.
94
Also, depending upon the shape and
size of the AuNPs, the free electrons on the surface of AuNPs
oscillate continually, granting them photothermal
properties.
95,96
Thiolate NAs can be easily conjugated to
AuNPs via covalent and electrostatic interactions between
sulfur and gold.
9
Gracezyk et al. (2021) developed an AuNP
carrier to deliver siRNA by functionalizing AuNPs with a thiol-
modified tectoRNAs trimer (structural RNA). They applied this
conjugate to regulate the CopGFP expression in MDA-MB-231
GFP/RFP cells (Fig. 1a).
97
The cellular uptake of AuNP: to
tectoRNA conjugate was determined by TEM studies, showing
Table 2 (Contd.)
S.
no Delivery agent Advantages Disadvantages Ref.
8 Lipid-based
nanocomplexes (LNPs and
SLNs)
Production on a large-scale industry level Limited drug loading for SLNs 84
Easy sterilization Instability in the bloodstream
Able to entrap both hydrophilic and hydrophobic drugs SLNs tend to gelation
Able to target specific cells or tissues Polymorphic transition for SLNs
Modulated and controlled release Particle size growth and drug expulsion
during storageLow toxicity due to their biocompatible and
biodegradable components and the absence of organic
solvents in their process
Protecting drugs from environmental conditions
Low cost compared with liposomes
Fig. 1 (a) Impact of AuNP:structural RNA complex on MDA-MB-231 GFP/RFP cells. In vitro studies show the regulation of CopGFP expression in the
targeted cells. Adapted with permission from ref. 97. Copyright 2021 ACS Chemical Society. (b) Confocal microscopy images of cells incubated with
(AC) nanoparticle dimers and (DF) non-targetingscrambled nanoparticle dimers. Sixteen HBE cells express only keratin 8, and MRC 5 cells
express only vimentin. A549 expresses both keratin 8 and vimentin. A uorescence signal corresponding to the presence of keratin 8 mRNA (A),
vimentin mRNA (B), and both vimentin and keratin 8 mRNA (C) are observed. When incubated with nanoparticle dimers designed with non-target-
ingscramble sequences, all three cell lines display no response. Adapted with permission from ref. 98. Copyright 2018 ACS Chemical Society.
Review RSC Pharmaceutics
650 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
their presence in the cytoplasm both inside the cytosol and
membrane structure. Also, the prepared AuNP:tecto RNA con-
jugates eectively regulated gene expression, as demonstrated
by GFP expression studies using fluorescence techniques.
Kyriazi et al. (2018) synthesized a DNAAuNP dimer-based
multifunctional platform for mRNA sensing and targeted drug
delivery.
98
The synthesized DNAAuNP dimer specifically
recognizes keratin 8 and vimentin mRNAs in keratin 8-expres-
sing 16HBE (epithelial cells), vimentin-expressing MRC 5
(mesenchymal cells), and A549, showing the expression of
both keratin 8 and vimentin respectively. Furthermore, two
anticancer drugs, doxorubicin (Dox), which detects keratin
8 mRNA, and mitoxantrone (MXT), which detects vimentin
mRNA, were intercalated into a DNA duplex, resulting in cell
death in response to specific mRNA signatures. Contrary to
this, the scrambled NPs, without having recognition sites for
targeted mRNA, failed to release the loaded drugs (Fig. 1b).
Another class of inorganic NPs includes semiconductor
quantum dots (QDs) due to their nanosize showing size-
tunable 3D quantum confinement eects, thus giving rise to
exquisite optical properties.
99101
QDs are typically made of
semiconducting materials such as group IIVI elements (CdS,
CdSe, CdTe, ZnS, ZnSe, ZnTe), group IIIV elements (InP or
InAs), group IIIIVI
2
elements (CuInS
2
, AgInS
2
), group IVVI
elements (PbS, PbSe, PbTe), or group IV elements (C, Si,
Ge).
102105
QDs are important fluorophores as they provide
emission in the UV, visible, and near-infrared ranges with an
impressive quantum yield compared with traditional organic
dyes, with drawbacks including photobleaching and poor
signal intensity.
106,107
All these properties of QDs make them
suitable probes for optical bioimaging and targeted molecular
sensing. For instance, Ma et al. (2019) designed a CdTe:Zn
2+
QD nanobeacon conjugated with black hole quencher (BHQ1)
and phosphorothioate co-modified DNA by hydrothermal syn-
thesis for single RNA detection and imaging (Fig. 2a(A)).
108
This nanobeacon was highly sensitive and eciently detected
low-abundance nucleic acids in live cells via FRET. QDs func-
tionalized with BHQ1, and single DNA were applied to detect
and image single HIV-1 RNAs in live HIV-1 integrated cells
(Fig. 2a(B, C)). Similarly, gene silencing in tumor cells of the
central nervous system was done using siRNA-loaded polyethyl-
enimine (PEI) functionalized CdSSe/ZnS QD-based nano-
carriers.
109
siRNA-loaded PEI-CdSSe/ZnS QDs eciently target
human telomerase reverse transcriptase (TERT). Two glioblas-
toma cell lines, U87 and U251, after transfection showed a
decrease in gene and protein expression levels of TERT with a
high level of gene transfection eciency within 48 h.
The rapid development of silicon nanostructures provides a
potential class of sensitive sensors and therapeutic agents for
real-time diagnosis and therapeutic applications.
110112
Silicon
NPs (SiNPs) are indirect band gap semiconductors and thus
exhibit longer excited-state lifetimes; after entering the cellular
system, SiNPs are biodegraded into silicic acid (nontoxic com-
pound) and easily excreted out of the body without showing
any sign of toxicity.
78,113
Chaix et al. (2019) reported amine-
functionalized porous SiNPs for the loading and improved
delivery of pDNA
114
(Fig. 2b). In vitro studies suggested that
the SiNPs showed better biocompatibility and successful trans-
fection of up to 10
7
RLU mg
1
proteins in HEK 293 cells.
Carbon nanomaterials (CNMs), also called green NMs, are
admirable fluorescent NMs with fascinating characteristics
such as tunable photoluminescence, photostability, ease of
fabrication, economical production, eco-friendliness, and bio-
compatibility.
115
In contrast to QDs, the mechanism behind
the fluorescence property of CNMs involves π-plasmon and
surface defects generated from radiative recombination of the
surface-confined electrons and holes.
116
Wang et al. (2014) syn-
thesized fluorescent carbon dots (CDs) for simultaneous
imaging and ecient siRNA delivery for cancer therapy.
117
PEI-functionalized CDs were applied for the adsorption of sur-
vivin siRNA. After transfection for two hours with siRNA-
loaded CDs/PEI complexes, MGC-803 gastric cancer cells
showed blue fluorescence in the cytoplasm, thus suggesting
the internalization of loaded siRNA into the cancerous cells
(Fig. 2c). Furthermore, the expression of survivin mRNA in
MGC-803 cells was downregulated to 96.4 ± 8.7 when exposed
to siRNA-loaded CD/PEI complexes. Iron oxide NPs (IONPs),
with their remarkable superparamagnetic properties and excel-
lent surface-to-volume ratio, have attracted worldwide inter-
est.
80
Magnetic NPs are widely used for magnetic particle
imaging, magnetic resonance imaging, hyperthermia, cell
tracking, targeted genes, and drug delivery.
118,119
Iron oxide
NPs are comparatively less toxic and cytocompatible, thus
allowing clinical translation. IONPs are usually synthesized
from iron oxides such as maghemite (γ-Fe
2
O
3
) and magnetite
(Fe
3
O
4
), metal alloys (e.g., FeCo and FePt), as well as the
doping of magnetically susceptible elements (e.g., MnFe
2
O
4
and CoFe
2
O
4
).
120,121
Ecient gene (pDNA) delivery to
mesenchymal stem cells was accomplished using positively
charged, PEI-coated IONPs.
122
The uniform and narrow-sized
(15 nm) IONPs exhibit good magnetic properties. In vitro
studies suggested that IONPs under the external magnetic
field eciently delivered 99% of the loaded pDNA within
30 min, followed by nuclear importing of the carried genes,
resulting in enhanced gene expression of the treated cells. The
caveolin-mediated pathway facilitated the internalization of
pDNA-loaded IONPs into the cytoplasm. Zhang et al. (2010)
designed PEI-coated IONPs and applied them to deliver inter-
fering RNA (siRNA) GFP plasmid via an external magnetic
force in 3D cell culture (NIH 3T3 cells).
123
The NPs facilitated
64% and 77% transfection eciency for siRNA and GFP
plasmid, respectively. These transfection complexes signifi-
cantly reduced the GFP-expressed cellsgrowth, with 8082%
silencing eciency. Furthermore, these complexes delivered
four toxic shRNA to the 3D cell culture, enhancing cell death
(4151%) (Fig. 2d).
Other commonly used inorganic NPs include silica NPs, which
are hydrophilic and porous, have access to surface functionali-
zation due to silane groups, and are widely used for drug delivery,
biomolecule conjugation, and many more applications.
However, for in vivo applications, the toxicity of these in-
organic NPs is an inevitable issue among researchers.
RSC Pharmaceutics Review
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 651
Additionally, the cellular uptake, circulation, and clearance of
NPs depend upon the physicochemical properties of the NPs.
After entering the bloodstream, NPs come across the biological
barrier composed of lipids, proteins, and other components.
The interaction of NPs with biomolecules resulted in the for-
mation of biomolecule coronas around the NPs before reach-
ing the target site, thus influencing the biological fate of the
NPs.
100
To circumvent these issues, researchers developed bio-
compatible surface modification approaches to inorganic NPs
to avoid or decrease the nonspecific interactions of these NPs
with biomolecules and increase prolonged accumulation at
the site of interest.
3.2 Polymeric NPs
Polymeric nanoparticles have shown great potential in recent
years due to their small size, ranging from 11000 nm.
Polymeric nanoparticles are mostly spherical with a solid
structure. The main advantages of polymeric NPs (PNPs)
Fig. 2 (a) Schematic illustration of QD-NBs for HIV-1 genomic RNA detection. (A) Schematic diagram of the QD-NBspreparation; schematic illus-
tration of QD-NBs for (B) HIV-1 genomic RNA detection in living cells, and (C) uorescence labeling of single virus particles. Adapted with per-
mission from ref. 108. Copyright 2019 American Chemical Society. (b) Reaction scheme for the chemical functionalization of SiNPs with histidine
and lysine and the complexation of pDNA. Adapted with permission from ref. 114. Copyright 2019 Royal Society of Chemistry. (c) Confocal laser
scanning microscopic images of MGC-803 cells incubated with Cdots@PEI or Cy3-siRNA for 2 h, or Cy3-siRNA-Cdots@PEI complexes for 2 and 5 h.
Adapted with permission from ref. 117. Copyright 2014 Springer Nature. (d) GFP silencing by PEI-coated SPMNs/GFP shRNA in GFP-transfected 3D
cell cultures. Adapted with permission from ref. 123. Copyright 2010 American Chemical Society.
Review RSC Pharmaceutics
652 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
include (1) the high molecular weight and polyvalent nature of
these molecules, which enable the encapsulation and delivery
of bulky and long-chain NAs,
124
(2) biocompatibility and bio-
degradability: PNPs easily break down inside the body into
water and carbohydrates and hence can be quickly eliminated
from the body,
125
(3) facilitation of successful loading and con-
trolled drug delivery,
126,127
(4) retention of the bioactivity of
drugs or biomolecules by evading the immune system, thus
enhancing their bioavailability and therapeutic potential, and
(5) the provision of a platform for ligand functionalization,
resulting in the targeted and stealthy delivery of drugs.
96
Polymeric nanoparticles can be synthesized by natural and
synthetic polymers.
128
Natural polymers, namely chitosan, hya-
luronic acid, starch, alginate, cellulose, and lignin, and syn-
thetic polymers, viz., polylactide-co-glycolide (PLGA), polylac-
tides (PLA), polyethylenimine (PEI), polyethylene glycol (PEG),
polycaprolactones, and polyacrylates, have been widely
explored for the synthesis of PNPs.
125,126,128
The most
common polymeric NPs include nanocapsules (polymeric cap-
sules surrounding a cavity) and nanospheres (solid matrix).
For instance, a nanovehicle based on cationic cyclodextrin-
polyethyleneimine 2k conjugate delivered mRNA encoding HIV
glycoprotein120 for treating HIV-1.
129
The delivery system
enhanced the intranasal delivery of mRNA by crossing the
nasal epithelial barrier through intracellular pathways, thus
resulting in a solid anti-HIV immune response. Biodegradable
chitosan-alginate 3D porous injectable gel was designed for
in vivo mRNA vaccine delivery.
130
Increased levels of IFN-α
secretion, luciferase reporter protein expression, and T-cell
proliferation were observed from mRNA lipoplex-loaded gel
scaolds compared with the systemic injection of naked
mRNA and mRNA:lipoplex.
A nanovehicle based on PLGA-encapsulated antisense
microRNA-21 (miRNA-21), known to be overexpressed in glio-
blastoma cells, was applied for the improved therapeutic eect
of temozolomide (TMZ) on glioblastoma cells (Fig. 3a).
131
Enhanced drug delivery and sustained gene silencing of
miRNA 21 were observed in glioblastoma cells, namely U87
MG, LN229, and T98G cells. This nanovehicle also showed a
significant decrease in cell viability (p< 0.001) with a 1.6-fold
increase in cell arrest at the G2/M phase in the TMZ-treated
cells (Fig. 3bd). Furthermore, the intracellular co-delivery of
the nanocomplex and TMZ in glioblastoma cells resulted in a
67% and 15% enhancement in the expression of miRNA-21
targeted phosphatase and tension homologue (PTEN) genes
and apoptosis-associated caspase-3 respectively (Fig. 3e). The
PLGASNA complex was designed to accommodate the chemo-
therapeutic drug coumarin spatially and nucleic acid to inde-
pendently improve the controlled loading and tunable release
of encapsulated moieties.
132
In vitro studies performed on
RAW blue cells to confirm the immunotherapeutic response of
PLGASNAs depicted dose- and time-dependent activation of
TLR9. PLGASNAs were cytocompatible at concentrations
ranging from 10 × 10
9
Mto2×10
6
M. Also, the cellular
uptake eciency of PLGASNAs was tenfold higher than their
linear counterpart and at a shorter time of 0.5 h. However,
PNPs also suer from drawbacks, including toxicity and a high
risk of particle aggregation. The FDA approves very few PNP-
based drugs for clinical applications.
133
Dendrimers or dense starsare globular, hyperbranched
3D structures, and their physicochemical properties can be
controlled for the desired application. Dendrimers can be syn-
thesized by divergent or convergent techniques.
134,135
The
Fig. 3 (a) Phase contrast microscopic images of control and antisense
miR-21 transfected cells treated with 500 μM TMZ. In vitro cell viability
MTT assays on control and antisense miR-21 transfected (b) U87 MG
cells, (c) LN 229 cells, and (d) T98G cells treated with dierent concen-
trations of TMZ. (e) Cellular pathway analysis of antisense miR-21 and
TMZ co-treatment on U87 MG cells. Adapted with permission from ref.
131. Copyright 2015 American Chemical Society. (f ) Schematic represen-
tation of PAMAM dendrimer/pDNA-p53 nanocarrier preparation for
cancer gene therapy. Adapted with permission from ref. 139. Copyright
2021, American Chemical Society.
RSC Pharmaceutics Review
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 653
remarkable properties of dendrimers make them suitable
vehicles for the delivery of nucleic acids. For instance, the poly-
cation nature of dendrimers provides multiple sites for the
electrostatic binding of the negatively charged phosphate back-
bone of NAs, resulting in solid DNA complexation. Also, the
formation of the dendrimerNA complex protects against
nuclease degradation of the attached NAs. It is speculated that
the presence of tertiary amines in the structure of dendrimers
enhanced the cellular uptake and delivery of NAs through
endosome escape via the sponge eect.
136
Other merits
include the attachment of biomolecules or ligands to the func-
tional groups available on the exterior surface. In contrast,
drugs and small cargo can be encapsulated in the interior of
the dendrimer.
133
Various dendrimers are studied for the
delivery of NAs, such as poly L-lysine, triazine, polyglycerol,
poly(propyleneimine), and poly(amidoamine) (PAMAM) based
dendrimers. At the same time, PAMAM is the most investi-
gated nanocarrier because of facile synthesis and functionali-
zation.
137
Palombarini and coworkers (2021) demonstrated the
targeted delivery of miRNA to myeloid leukemia cells (which
are otherwise challenging to transfect) by incorporating
nucleic acid with ferritin-poly(amidoamine) (PAMAM) dendri-
mer NPs.
138
The cellular internalization of this nanocomplex
drives morphological changes and enhances the expression of
the retinoic acid receptor, alpha (RARα), an early hallmark of
granulocytic dierentiation. Mekuria et al. (2021) developed a
nanocarrier by covalently binding PAMAM dendrimer with
4,4-dithiodibutryic acid (DA) to successfully deliver p53-pDNA.
The nanocarrier reflected a 2.3- and 2.1-fold increased gene
transfection in 4T1 and mouse breast cancer cells.
139
Furthermore, in vitro and in vivo studies showed an upregu-
lated expression of mRNA and p53 and p21 protein and down-
regulation of cyclin-D1 and CDK-4 protein, thus facilitating
cell cycle arrest in the G1 phase (Fig. 3f).
Dendrimers are ideal for delivering NAs with enhanced
stability and improved cellular internalization compared with
other therapeutics. However, regarding the safety concerns,
the toxicity of these dendrimers is still unaddressed. The
strong cationic amine groups on the surface of dendrimers
resulted in solid binding with the negatively charged cell mem-
brane, which led to destabilization, disruption of cell com-
ponents, and eventually lysis.
140
Nanofibers are a promising class of biomaterials for nucleic
acid delivery, oering several advantages including a large
surface area, biocompatibility, encapsulation and targeted
delivery (gene, growth factors, protein, and peptide delivery),
and scaolding for tissue engineering.
85
Nanofibers can be
synthesized by dierent methods, namely, phase separation,
electrospinning, and physical and chemical fabrication.
Various natural, semisynthetic, and synthetic polymers are
widely used for their synthesis. Furono et al. (2022) reported
plasmid DNA delivery using horseradish peroxidase cross-
linked gelatin nanofibers.
141
Nanofibers immobilized with
Lipofectamine/pDNA resulted in the transfection of pDNA
delivery in HEK293 cells. Furthermore, genome-editing mole-
cules including Cas9 protein and guide RNA (gRNA) were
expressed in nanofiber-treated HEK293 cells, resulting in gene
knock-in and knock-out. Polycaprolactone nanofiber-encapsu-
lated siRNA showed controlled release for up to 28 days with
successful transfection of the treated HEK 293 cells.
142
The
cells showed enhanced cellular uptake and GAPDH gene silen-
cing of 6181%. Nanofibrous scaolds made up of collagen
type 1 were used for the controlled and long-term delivery (at
least 5 months) of siRNA/silica NPs.
143
In vivo studies revealed
that the nanofiber-based scaolds showed more eective gene
silencing (p< 0.05) as compared with traditional bolus deliv-
ery. An in vivo biodistribution study revealed that siRNA stayed
confined up to 290 μm from the implants. As compared with
negative scrambled siRNA therapy, a reduction in fibrous cap-
sules of 45.8% was observed after 4 weeks.
Along with the advantages, nanofibers do have some draw-
backs that researchers are working to overcome.
86
(1)
Nanofibers may have inherent toxicity or cause inflammatory
responses in the body. (2) Depending on the material, nano-
fibers may degrade too quickly or too slowly, aecting the
release profile of the drugs they carry. (3) Manufacturing nano-
fibers for large-scale clinical use can be complex and
expensive.
3.3 Proteins/peptides
Proteins, a class of natural biomolecules, stand out as an
attractive biocompatible substitute for synthetic polymers in
nanomedicine due to their biodegradability, natural abun-
dance, mild synthesis, and fast metabolization.
144,145
Additionally, proteins showed remarkable chemical modifi-
cation properties due to their surface abundance of functional
groups such as carboxyl, amine, and hydroxyl. The amphiphi-
lic nature makes them amenable for small molecules, metallic
NPs, and hydrophobic and hydrophilic drugs.
145,146
Protein-
based NPs, when administered inside the body, showed weak
immunogenicity, were eciently degraded by the enzymes and
were eliminated through hepatic clearance. Abraxane is an
FDA-approved drug composed of paclitaxel-bound albumin-
based NPs used to treat metastatic breast cancer by inhibiting
the mitosis of cancer cells.
147,148
The drug avoids hypersensi-
tivity due to albumin protein, a major drawback of traditional
anticancer drugs. Protein NPs can be easily synthesized via
electrospray, emulsion, and desolvation processes using
natural proteins, namely albumin, fibroins, 30Kc19, gelatin,
lipoprotein, legumin, zein, gliadin, and ferritin proteins.
144,149
Peptides are short chains of amino acids linked by a
covalent amide bond (peptide bond). They exhibit remarkable
sequence and functional diversity, merits employed to design
spherical NPs via a self-assembly approach.
150
Several peptides
are currently used for biomarker imaging, targeted drug and
gene delivery, bioprinting, wound healing, and tissue
engineering.
151,152
Dierent peptides such as nuclear localiz-
ation, tumor-targeted, and cell penetration peptide sequences
are extensively reported for biomedical applications. Jia et al.
(2020) reported enhanced siRNA delivery and improved gene
silencing using hyaluronic acid (HA)-modified transmembrane
peptide octa-arginine (R8)-based R8-bipolar (HA-bibola/siRNA)
Review RSC Pharmaceutics
654 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
and R8-monobola (HA-bola/siRNA) amphiphilic nanocom-
plexes.
153
Cell viability studies reflected better cytocompatibil-
ity of HA-bibola/siRNA compared with HA-bola/siRNA and
control samples (PEI/siRNA) in 4T1 cells. Additionally, HA-
bibola/siRNA showed enhanced cell uptake eciency and
down-regulation of Bcl-2 protein expression due to the pres-
ence of cell-penetrating peptides on the surface of the nano-
complex. In vivo, studies reflected higher antitumor ecacy,
improved targeted ability, and increased Bcl-2 gene suppres-
sion in 4T1 tumor-bearing Balb/c mice.
3.4 Spherical nucleic acids (SNAs)
The primary function of nucleic acids includes storing and
transduction of genetic information. Nucleic acid-based (DNA,
RNA, CRISPR/Cas9 gene editing system) spherical NPs hold
great significance in the biosensing, therapy, and silencing of
various diseases ranging from viral infections to neurological
disorders, cardiovascular diseases, and cancer by taking
advantage of cellular pathways.
154156
In the present era, thera-
peutic technologies related to nucleic acids are at the forefront
of fighting the COVID-19 pandemic around the globe.
157,158
They consist of highly compact self-assembled oligonucleotide
layers oriented in 3D geometries based on typical Watson
Crick base-pairing.
119,120
Selective and precise base-pairing of
DNA/RNA NPs dierentiates them from traditional bio-
molecules, which might be otherwise dicult to design with
such ease.
159
Various chemical modifications (nucleotide
sequence, sugar, or phosphate backbone modification), bio-
compatibility, and programmable therapeutic approaches are
other fascinating properties of these nanostructures.
160
Most
nucleic acid-based NPs interact via complementary base
pairing with their target molecules, thus resulting in specific
and rapid action.
161
Spherical nucleic acids (SNAs) comprise
(1) the outer shell of densely packed nucleic acid radially
encasing the contents, and (2) an inner core of NPs. They were
initially made of DNA shells and AuNP cores.
161,162
Since then,
various inorganic nanoparticles such as Ag,
163
QDs, magnetic
NPs,
164
silica,
165
organic nanocomposites such as polymers,
proteins, and liposomes,
166,167
and hybrid structures of in-
organicorganic materials were used to explore dierent bio-
medical applications. The three-dimensional architecture of
SNAs provides them with unique physicochemical properties
and makes them superior to their linear counterparts.
87
The
spherical shape allows the dense packing of the oligo-
nucleotide into a limited space.
168
This compact structure of
SNAs leads to an enhanced electric charge, increasing the
stability of the SNAs and providing resistance to nuclease
degradation and prolonged cell accumulation via scavenger
receptor engagement and endocytosis.
169
SNAs can easily
evade the immune systems attack due to strong electric
charges because of the dense packing. Also, the interaction of
SNAs with receptors on the cell surface resulted in easy pene-
tration through the cell, tissue membranes, and even the
bloodbrain barrier.
170
Melamed et al. (2018) suggested that
the spherical architecture of SNAs functionalized with poly-
ethylenimine has improved siRNA-mediated GFP gene silen-
cing 10-fold.
171
Wang et al. (2019) reported an SNA-based
vaccine for treating a mouse tumor.
172
SNA co-delivered CpG
oligonucleotide (adjuvant) and peptide (antigen) to generate
an antitumor immune response. The results suggested that
the vaccine could increase the survival rate to 31 days, also
delaying tumor growth by 15 days. SNAs eciently promote
gene regulation for the treatment of skin diseases.
173
Randeria
et al. (2015) showed the application of siRNA-encapsulated
SNAs in impaired wound healing by downregulating ganglio-
side GM3 synthase (GM3S) in diabetic mice.
174
The expression
of GM3S was reduced by >80% at the wound site via the siRNA
pathway, and the wound was observed to heal within 12 days
in the SNA-treated mice.
174
Although nucleic acid-based drugs are in the infant stages
of clinical trials, they have revolutionized therapeuticsfate in
recent years. Despite the remarkable merits of SNA-based
therapeutics, challenges still hinder their further clinical and
translational applications.
4. Classication of lipid
nanocomplexes for ecient delivery
of NAs
The clinical application of NPs depends on their successful
delivery to the disease site. The eectiveness of synthesized
NPs in treating diseases depends upon their therapeutic
response, which can be achieved by delivering a required
dose to the targeted site. During their journey to a target site,
NPs must evade multiple biological barriers at dierent sites,
including (1) phagocytosis by liver cells while NPs are circu-
lating in the blood.
175
(2) Even after reaching the target site,
NP entry into the disease site is restricted by the endothelial
cell walls. (3) The immune system also recognizes and
destroys foreign components (NPs) and vectors (spherical
DNA/RNA containing genetic information).
176
(4) The intern-
alization of NPs in the cell membrane or nucleus enhanced
the eect of the former. The overall eectofthesebarriers
restricted the penetration of most NPs into the targeted cell
or organ and thus reduced their therapeutic eciency.
Recently, remarkable progress has been made in designing
and developing delivery systems that enhance the therapeutic
trajectory of drugs, inorganic NPs, and DNA/RNA-based
genetic drugs.
Lipid-based delivery cargos, namely micelles, liposomes,
and lipid nanoparticles (LNPs), are promising delivery agents
as they can protect nanomaterials from degradation and
enhance circulation by avoiding early clearance by the
immune system.
177,178
Lipids are widely used nonviral delivery
agents and are fascinating because of their easy synthesis,
characterization, greater payload, homogeneity, biodegradabil-
ity, and marginal toxicity profile. A broad range of lipid-based
NPs is utilized to deliver NAs, including cationic lipids, ioniz-
able lipids, zwitterionic lipids, LNPs, liposomes, and solid
lipid NPs (SLNs) (Table 3).
RSC Pharmaceutics Review
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 655
Lipid-based delivery agents, viz., micelles, liposomes, and
lipid NPs, have been extensively studied to encapsulate and
deliver NAs, primarily due to their ease of synthesis and inter-
action with NAs (Fig. 4a).
195
The essential components of FDA-
approved LNPs for NA delivery developed by pharmaceutical
companies include cationic or ionizable lipids, cholesterol, a
helper lipid, and a PEG-lipid (Fig. 4b and c), for example DLin-
MC3-DMA, PEG: PEG-2000-C-DMG (Alnylam), SM-102,
PEG-2000-DMG (Moderna), and ALC-0315(Pfizer/BioNTech)
ALC-0159 (Pfizer/BioNTech/Acuitas), along with DSPC and
cholesterol (Fig. 4d).
Moreover, lipid-based nanovehicles can be surface functio-
nalized to achieve the targeted delivery and release of thera-
peutics in a specific tissue or cell.
196,197
However, it is impor-
tant to consider the chemical properties of the lipids used in
NA delivery for ecient drug development. One of the critical
parameters of lipids is their surface charge. Dierently
charged lipids can have varying degrees of compatibility with
nucleic acids. In general, positively charged lipids tend to have
a higher degree of compatibility with negatively charged
nucleic acids, such as DNA and RNA, as electrostatic inter-
actions between the positive charges on the lipids and the
negative charges on the nucleic acids can help to stabilize the
interactions between the two. Conversely, negatively charged
lipids may have reduced compatibility with NAs, as the nega-
tive charges on the lipids may repel the negative charges on
the NAs. Also, it is well known that the charge of NPs alters
their tissue selectivity after intravenous administration. Taking
advantage of this approach, dierent tissue-targeted LNPs
have been designed by incorporating cationic or anionic lipids
into the general composition of LNPs to tune their charge.
198
For example, using zwitterionic lipids, which have both a posi-
tive and negative charge, has been shown to enhance the stabi-
lity of NPs and improve in vivo ecacy.
199
Additionally, using
lipids with a high melting point can help improve the NPs
stability and enhance their ability to penetrate cell mem-
branes.
200
Another critical factor to consider is the size of the
NPs formed by the lipids. Smaller NPs have been shown to
penetrate cell membranes more eectively and deliver NAs to
target cells.
201
However, it is also important to note that the
size of the NPs should not be so small that they are rapidly
cleared from the body by the immune system. Besides consid-
ering the lipidsstructural and chemical properties, the NPs
composition is also an essential consideration for NA deliv-
ery.
202
For example, using PEG as a surface coating on lipids
has been shown to improve the stability of the particles and
reduce their clearance from the body.
203
Additionally, using
other polymers, such as PEI or PLA, can also help improve the
stability and ecacy of the lipid-based delivery agents. Overall,
the design of lipids for NA delivery is a complex process that
requires a thorough understanding of the structural, chemical,
and composition-based factors that influence their ecacy.
Through the use of structureactivity relationships and careful
consideration of the properties of the lipids, it is possible to
design nanoparticles that can eectively deliver NAs to target
cells and improve the ecacy of gene therapy.
Table 3 A summary of lipid-based NA delivery systems and their biomedical applications
S.
no Type of lipid vehicle Composition of vehicle Cargo Method of
synthesis Treatment Ref.
1 Cationic DOTAP/cholesterol mRNA, pDNA, and oligonucleotide Thin-film
evaporation Ovarian cancer 179
2 Cationic 9322-O16B/Chol/DOPE mRNA Chemical B-cell lymphoma 180
3 Cationic DOPE-stearylated octaarginine (STR-R8), DOTMA-YSK05,
cholesterol-GALA peptide pDNA Chemical Ecient and selective delivery of pDNA to
the lungs 181
4 Ionizable cationic DLin-MC3-DMA/DLin-KC2-DMA/DODAP/DSPC/PEG-DSPE,
cholesterol siRNA Microfluidic pDNA transfection 182
5 Ionizable cationic C-12-200 (IL)/DOPE, cholesterol/PEG-lipid conjugate pDNA Microfluidic Cardiovascular diseases 183
6 Ionizable cationic DLin-MC3-DMA, DSPC, cholesterol, DMG-PEG2K mRNA Chemical Hepatic reticuloendothelial diseases 184
7 Ionizable DSPC, CHO, DMG-PEG
2000
siRNA Chemical Hyperlipidemia 185
8 Zwitterionic Phosphatidylcholine, DPPC, cholesterol pDNA Chemical pDNA transfection 186
9 Liposome DOTAP/DOPE/DSPE-PEG 6-Carboxyfluorescein-labeled 14-mer
oligonucleotide Ethanol dilution In vivo labeling of human microbiota 187
10 Cationic liposome DPPC/DOTAP/cholesterol GFP-mRNA Lipid film
hydration Neurodegenerative diseases 188
11 Protamine liposome DOTAP/cholesterol mRNA Chemical Colorectal cancer gene therapy 189
12 Lipid nanoparticles C-14-4 IL/DOPE/cholesterol/PEG mRNA Microfluidic Engineering of CAR T cells to kill cancer
cells 190
13 Lipid nanoparticles DLin-MC3-DMA/DAP/phospholipid/cholesterol/PEG Spherical DNA/RNA Ethanol dilution Organ-specific delivery of nucleic acid 191
14 Solid lipid NPs DOTAP/lecithin/cholesterol/lipopolysaccharide TNF-αsiRNA Ultrasonication Rheumatoid arthritis 192
15 Solid lipid NPs Stearic acid, soya lecithin pDNA, Dox Solvent
displacement Lung cancer therapy 193
16 Cationic solid lipid
NPs Peptide-cationic lipid CDO14 siRNA, upconversion NPs Thin-film
dispersion Bioimaging and gene therapy 194
Review RSC Pharmaceutics
656 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
4.1 Cationic lipids
Cationic lipids (CLs) are well characterized by their positively
charged hydrophilic head (such as quaternary ammonium,
amines), linker (such as ester, ether, disulfide), and negatively
charged hydrophobic tail (cholesterol, tocopherol). CLs are
well-explored for the delivery of negatively charged nucleic
acids. Cationic lipid, namely N-[1-(2,3-dioleyloxy) propyl]-N,N,
N-trimethylammonium chloride (DOTMA) bearing a quatern-
ary ammonium head group and unsaturated phosphatidyletha-
nolamine (DOPE), was first employed for nucleic acid delivery
for gene therapy by Felgners group in 1987.
204
The lipid
formed is still commercially available with Lepofectin and is
extensively used for the in vitro delivery of nucleic acids.
205
CLs
formed complexes with negatively charged phosphate groups
in the polynucleotidesDNA. The complexes are formed
through positively charged head groups via electrostatic inter-
actions, whereas hydrophobic tails exist as bilayers around
DNA.
206
This lipid envelope surrounding DNA provides resis-
tance against omnipresent nucleases and overcomes immuno-
Fig. 4 FDA-approved lipid-based structures contain some variation of the four essential components: cholesterol, a helper lipid, a PEG-lipid, and a
cationic or ionizable lipid. (a) Lipid-based structures can include micelles, which consist of a lipid monolayer, or liposomes, which consist of a
bilayer. Lipid nanoparticles comprise multiple lipid layers and lipid and nucleic acid microdomains. (b) and (c) In addition to the RNA payload, LNPs
often consist of cholesterol, a helper lipid, a PEG-lipid (all shown in part b), and a cationic or ionizable lipid (part c). (d) The molar ratios of the four
components constitute the FDA-approved Acuitas/BioNTech/Pzer COVID-19 vaccine and patisiran, which delivers siRNA to the liver. Adapted with
permission from ref. 195. Copyright 2022, Springer Nature.
RSC Pharmaceutics Review
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 657
genicity and mutagenicity. Additionally, it can easily combine
with other anionic sulfate groups of cell membrane proteogly-
cans, thus ensuring cell uptake. 1,2-Dioleoyl-3-(trimethyl-
ammonium) propane (DOTAP) and polyethylene-glycol-2000-
1,2-distearyl-3-sn-phosphatidylethanolamine (PEG-DSPE)
based cationic lipid was used to coat hydrophobic IONPs via
self-assembly synthesis.
207
The lipid-coated IONPs showed an
average size of 46 nm and improved delivery in three cell lines:
HeLa, PC-3, and Neuro-2a. Furthermore, in vivo, studies
revealed that the growth status of the tumor cells in Balb/c
mice was eciently monitored through MR images, and the
magnetic property of IONPs was retained up to 15 days after
administration. Sun et al. (2022) demonstrated the ecacy of
DOTAP/cholesterol-based CL NPs for successfully delivering
mRNA, pDNA, and oligonucleotide.
208
The nanocomplex was
stable for up to 60 days at 4 °C storage conditions without
aecting transfection ecacy. The eective DOTAP/cholesterol
CL NP:mRNA ratio for in vitro studies was 62.5 µM lipid to 1 μg
mRNA. It was speculated that a lower lipid concentration
carried a lower surface charge, resulting in decreased cell inter-
action and endosome escape. In contrast, a higher concen-
tration might lead to cytotoxicity and inhibition of mRNA dis-
sociation from the nanocomplex.
Based on the chemical structure of a positively charged
head group, lipids can be classified into six classes, namely
quaternary ammoniums, amines (primary to quaternary),
amino acids or peptides, guanidiniums, heterocyclic head-
groups, and some uncommon headgroups.
177,209
Headgroups
containing charge and dimensions impact cell interaction,
endosome escape, and access to the target cell.
210
Linkers typically comprise non-biodegradable (e.g., ethers
and carbamates) and widely used biodegradable (e.g., esters,
amides, and thiols) functional moieties. Linkers form a junc-
tion between the headgroup and tails.
211
The linker aects the
stability, biodegradability, cytotoxicity, and transfection
eciency of LNPs.
The negatively charged, lipophilic aliphatic chains of
sterols form the last part of cationic lipids. The hydrophobic
tails elicit NP formation and potency by maintaining fluidity,
hydrophobicity, and fusion with the cell membrane.
209
Nevertheless, despite the advantages CLs oer for the deliv-
ery of therapeutic cargo, researchers are motivated to continue
their development and optimization in preclinical research.
Their drawbacks include reduced in vivo ecacy, low circula-
tion time, rapid elimination by RES, o-target accumulation in
the negatively charged cellular system, and unacceptable tox-
icity related to inflammatory responses, which has challenged
their clinical acceptance.
212214
4.2 Ionizable lipids
Ionizable lipids (ILs) or ionizable cationic lipids are small
amphiphilic vesicles containing an ionizable protonatable ter-
tiary-amino head group, a spacer/linker, and a hydrophobic
moiety.
215
ILs showed the unique feature of electrostatic
charge dependency on the lipid pK
a
and pH of their environ-
mental surroundings and, thus, become positively charged at
acidic pH, ensuring the binding of therapeutic cargos while
maintaining a neutral charge at physiological pH, reducing
toxicity eects.
216,217
ILs with a suitable pK
a
can modify their
electrostatic charge to ensure prolonged circulation and cyto-
solic release of therapeutics, elevated transfection eciency,
access to targeted sites, and reduced toxicity. The transfection
eciency of ILs depends mainly on the pK
a
, with an optimal
pK
a
value of 6.26.5.
176,218
Ionizable lipids can be categorized into various subclasses
based on their structure.
(1) Unsaturated ILs: the degree of tail unsaturation sig-
nificantly impacts the fluidity and delivery eciency of ILs.
The high unsaturation of lipid tails leads to nonbilayer lipid
formation, enhancing membrane disruption and cargo
release. Some common unsaturated ILs include DLin-MC3-
DMA (MC3), A18-iso5-2DC18, OF02, etc.
(2) Polymeric ILs: the structural basis of polymeric ILs is
the substitution of free amines on cationic polymers with alkyl
tails, and their hydrophobic aggregation results in improved
particle formation. Common examples include (1,2 bis(triclo-
san-10,12-diynoyl)-snglycero-3-phosphocholine) (DC8,9PC),
7C1, G0-C14 etc.
(3) Biodegradable ILs: the main objective of using
dierent materials for biomedical applications includes
improved biocompatibility and biodegradability with reduced
toxicity and inappropriate accumulation inside the body.
202
Adding biocleavable ester bonds (simple structure, chemical
stability) to ILs provides biocompatibility via undergoing enzy-
matic hydrolysis in vivo. Maier et al. (2013) synthesized a
library of biodegradable ILs by incorporating ester bonds at
dierent lipid positions.
202
Recently, anti-SARS-CoV-2 vaccines have also used ILs for
their formation.
219
The vaccines mainly comprise mRNA
motifs and LNPs composed of pH-sensitive ILs. Kim et al.
(2021) reported on the target delivery of RNA using engineered
ILs into specific liver cells, namely hepatocytes and liver sinu-
soidal endothelial cells.
220
The delivery of ILs to the targeted
site was ensured by controlling the size and PEG:lipid ratio
(Fig. 5a). Moreover, active targeting was achieved by adding
mannose to the ILs. In vivo, gene silencing studies showed the
selectivity of the engineered ILs towards the targeted liver cells
(Fig. 5b).
ILs are widely explored by modifying their domains, includ-
ing hydrophobic tails, hydrophilic heads, or linkers. Walsh
et al. (2013) reported a new class of IL with a lysine head group
linked to long-chain dialkylamine via an amide linker for
siRNA delivery.
221
The resulting pH-dependent ILs contain a
carboxylate group and two ionizable amines. This ionizable
lipid exhibits electrostatic charge-dependent membrane dis-
ruption advantages, successful in vitro siRNA transfection, and
enhanced siRNA-mediated knockdown in transfected HeLa
cells. Nucleic acid delivery was also accomplished using
dierent aminoglycoside-derived ILs.
222,223
Lipid-based delivery systems are widely used for improved
chemotherapy for cancer treatment. Broma et al. (2019)
reported ILs as a Trojan horse in delivering AuNPs with a size
Review RSC Pharmaceutics
658 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
range of 5 nm for enhanced outcomes in the radiation therapy
of triple-negative MDA-MB-231 cells.
224
The ILs with the com-
position DLin-MC3-DMA/DSPC/cholesterol/PEG were used to
coat AuNPs. The complex of ILAuNPs showed a 73-fold
increase in the uptake of small-sized AuNPs in cancerous cells.
4.3 Zwitterionic lipids
The zwitterionic lipids (ZILs) contain an equal number of co-
valently bonded anionic and cationic moieties.
225
ZILs have
gained the utmost attention in biomedical applications,
including drug delivery and increased uptake of loaded mole-
cules by reducing the adsorption of proteins on nanocarriers
in serum.
226
Obata et al. (2010) designed endosomal-pH-
responsive liposomes functionalized with glutamic acid-based
zwitterionic lipids for enhanced drug delivery applications.
227
The liposomes showed a positive zeta potential at lower pH
and became negatively charged at basic pH due to the carboxyl
group moiety in the glutamic acid. Furthermore, synthesized
pH-responsive lipids reflected high fusogenic potential with
the anionic membrane of cancer cells. Thus, it ensures the
Fig. 5 (a) Preparation of LNPs derived from ionizable lipids. (b) In vivo evaluation of ILs showed potent luciferase expression. Ex vivo organ images
showed that LNPs were mostly taken up into the liver. Liver histology image and transfection eciency showed signicant tdTomato uorescence in
the hepatocyte. cv, central vein. Adapted with permission from ref. 220. Copyright 2021, American Association for the Advancement of Science. (b)
Synthesis and in vivo application of CP liposomes. (c) Tumor therapy ecacy of CP liposomes (DDCP-c, DMCP-c, DPCP-c, and folate-CP) with
DPPC as the control. Adapted with permission from ref. 228. Copyright 2021, American Chemical Society.
RSC Pharmaceutics Review
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 659
improved release of encapsulated Dox in HeLa cells and high
antitumor activity in vivo against a xenograft breast cancer
tumor. Cancer treatment with targeted and enhanced drug
delivery was ensured using a novel biorthogonal zwitterionic
lipid (choline phosphate (CP)) based liposome functionalized
with folic acid using a click reaction.
228
Furthermore, com-
pared with phosphatidylcholine (PC) based liposomes, supra-
molecular ionic pair interactions of zwitterionic lipid exhibit
adhesive characteristics with the cell membrane of cancer
cells, enhanced biocompatibility in normal cells, significantly
enhanced cytotoxicity and inhibition of tumor growth (Fig. 5c).
4.4 Liposomes
Liposomes are small spherical-shaped artificial vesicles syn-
thesized from cholesterol and nontoxic phospholipids.
Liposomes are widely explored as delivery agents due to their
size and ability to load both hydrophilic and hydrophobic
molecules.
229
Doxil was the first liposomal formulation
approved in 1995 by the FDA of the USA for the treatment of
refractory acquired immune deficiency syndrome (AIDS)-
related to Kaposis sarcoma.
230
The remarkable journey of lipo-
somes as delivery agents includes ligand-targeted delivery,
nucleic acid/gene delivery, and delivery of active drugs, poly-
mers, anesthetics, and antimicrobial agents.
231
Spherical lipo-
somes are readily taken up by irregular and distorted tumor
cells via an enhanced permeability and retention eect, result-
ing in elevated drug distribution at the tumor site.
232
Curcumin and metformin-loaded DSPE-PEG2000-hyaluronic
acid liposomes were designed to target hepatocellular tumors
and drug resistance.
233
In vitro and in vivo studies revealed that
this formulation exhibits more potent antiproliferation and
antimetastasis. The inhibition of drug resistance and tumor
growth was attributed to the down-regulation of multidrug re-
sistance-related P-glycoprotein and the inducing epithelial
mesenchymal transformation of tumor cells. Michel et al.
(2017) developed cationic liposomes to load mRNA and
improve cell transfection to treat alpha-1-antitrypsin
deficiency.
234
Liposomes showed a prolonged transfection
eect with negligible cytotoxicity in A549 cells. Liposomes had
a long-acting transfection eect on cells, resulting in increased
expression of a functional alpha-1-antitrypsin protein.
Dhaliwal et al. (2020) developed a cationic liposome-based
nanovehicle for intranasal delivery and potent mRNA transfec-
tion to a murine models brain.
188
The incubation of mRNA-
loaded cationic liposomes with J774.1 macrophage cells
showed stable GFP expression in the cytosol up to 24 h (Fig. 6a
and b). Furthermore, intranasal administration of mRNA-
loaded cationic liposomes in mice compared with control
Fig. 6 (a) GFP-mRNA transfection studies in J774A.1 macrophages using cationic liposomes at dierent concentrations. (b) Quantication of %
GFP-expressing cells. Lipofect_5 represents Lipofectamine (with 5 μg GFP-mRNA)-treated cells as a positive control, and the 0 μg GFP-mRNA/Lipo
group represents empty liposome-treated cells as a negative control (showed no GFP signal). (c) GFP-mRNA expression in mouse brain, 24 h post
intranasal administration of GFP-mRNA-loaded cationic liposomes. Adapted with permission from ref. 188. Copyright 2020, American Chemical
Society.
Review RSC Pharmaceutics
660 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
(GFP-mRNA) and vehicle (liposome)-treated groups showed sig-
nificantly higher GFP expression by 15% (p< 0.05) up to 24 h
(Fig. 6c).
Lei et al. (2020) demonstrated colorectal cancer gene
therapy using a protamine/liposome-based delivery carrier
loaded with IL-15 mRNA (CLPP-mIL-5 complex).
189
Cytokine
IL-15 can be a cancer gene therapy due to its immune stimu-
lation characteristics. In vitro studies showed the successful
delivery of mRNA in C26 cells. The accumulation of mRNA in
cancer cells induced cytotoxicity and lymphocyte stimulation.
Following systemic delivery in three C26 murine colon cancer-
bearing mice, CLPP-mIL-5 complex inhibits cancer rates up to
70%, 55%, and 69% in abdominal cavity metastasis tumor,
subcutaneous, and pulmonary metastasis models, respectively.
4.5 Lipid nanoparticles (LNPs)
Lipid NPs are the highly advanced clinically viable vector for
NA delivery with Onpattro (patisiran) approval for amyloidosis
treatment and their application in COVID-19 vaccine around
the globe.
177,235
Lipid NPs are composed of four dierent
lipids, namely (1) ionizable cationic lipid, (2) three neutral
helper lipids, namely phospholipid, (3) cholesterol, and (4)
PEGylated lipid.
236
In general, ionizable cationic lipids form
an electrostatic complex with anionic nucleic acid, enabling
intracellular uptake and endosomal escape. The stability and
fluidity of the complex are maintained by cholesterol.
Furthermore, phospholipids enhance the structural integrity
of the complex. PEGylated lipid is an important module that
maintains stability, enables cellular uptake, and protects the
lipidNA complex from being captured by the protein
corona.
237,238
Billingsley et al. (2022) developed a sequential
library of biocompatible lipid NPs via microfluidic mixing to
successfully deliver mRNA to T cells (Fig. 7a and b).
190
Their
study revealed that lipid NPs composed of C14-4 ionizable
lipid improved the delivery of mRNA by a 3-fold increase in
primary human T cells. Additionally, lipid NPs with minimal
cytotoxicity induced chimeric antigen receptor (CAR)
expression and eectively killed cancer cells (Fig. 7c). Ly et al.
(2022) developed lipid NPs using FDA-approved ionizable
lipids, namely, MC3, ALC-0315, and SM-102, and applied them
for the loading and delivery of self-amplifying mRNA (saRNA),
protein expression, and cytokine activation in vitro (triggering
dierent levels of IL-6 response).
236
Furthermore, it was found
that PEG facilitates the encapsulation and stability of saRNA,
which is otherwise complicated to preserve as compared with
mRNA. The type of ionizable lipid highly influenced the
protein expression. Protein expression was highest in
ALC-0315, followed by SM-102, whereas MC3 failed to show
potent protein expression. The study provides an insight into
lipid NPs required for specific applications, including the
delivery of heavy RNA molecules (saRNA, Cas9), therapies
related to protein replacement, and vaccine production.
4.6 Solid lipid nanoparticles
Solid lipid NPs (SLNs) have been extensively studied over the
past decade because they provide the combinatorial eect of
various carrier systems, including polymeric nanoparticles,
emulsions, and liposomes.
239
SLNs have changed the dimen-
sions of gene and drug delivery because of their advantages,
including biocompatibility and safety profile, avoidance of
organic solvents, ease of large-scale synthesis, water-based
technology, physicochemical stability, ability to encapsulate
proteins and nucleic acids along with hydrophilic, hydro-
phobic drugs and bioactive compounds, and controlled and
sustained release of loaded components.
240242
SLNs are
spherical nanoparticles with diameters of 501000 nm.
239,242
SLNs remain solid at room temperature because of the core of
SLNs. The core of SLNs is composed of solid lipids such as
glycerides, steroids, fatty acids, or waxes, whereas LNPs com-
prise solid and liquid crystalline lipids.
201,243
SLNs are
superior to lipid nanoparticles in protecting nucleic acids
Fig. 7 (a) Schematic of LNP synthesis, including the components used
to make LNPs via microuidic mixing and the expected resulting struc-
ture. (b) Visualization of the design process used to generate libraries A
and B with library A resulting from orthogonal DOE screening of a wide
range of excipient molar ratios, and library B examining more formu-
lations within a narrowed range of excipient ratios based on the results
from the library A screen. (c) Schematic of the CAR T cell production uti-
lizing either LNPs or EP for mRNA delivery to T cells. The treated T cell
populations may dier in viability and CAR potency depending on the
transfection method. Still, both can generate functional CAR T cells to
induce targeted cancer cell killing. Adapted with permission from ref.
190. Copyright 2022, American Chemical Society.
RSC Pharmaceutics Review
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 661
from degradation and leakage during storage. SLNs are syn-
thesized using physiological, biodegradable, and biocompati-
ble lipids in a solid state at room temperature, emulsifiers, or
a combination of pharmaceutical agents and solvents.
Dierent methods have been extensively investigated for the
synthesis of SLNs, such as emulsion-solvent evaporation,
244
microemulsion,
245
high-shear homogenization,
246
ultra-
sonication,
247
solvent injection methods,
248
nanoprecipitation
(solvent displacement technique) and so on.
193
SLNs possess a
solid lipid core loaded with the active ingredient and stabilized
with an outer surface of surfactant. Nonviral delivery of
plasmid DNA was eciently done using SLP-based cargos.
249
The key ingredients of the SLNs prepared via the solvent-emul-
sification method include cholesteryl oleate glycerol trioleate,
and DOPE cholesterol3β-[N-(dimethylaminoethane)carbamoyl
(DC)-cholesterol]. Furthermore, plasmid DNA was incorporated
into SLNPs. In vitro studies suggested improved transfection
eciency of plasmid DNA in dendritic cells. Song et al. (2017)
synthesized cationic solid lipid NPs (CSLNs) for the delivery of
siRNA and β-NaYF
4
:Yb,Er upconversion nanoparticles (UCNPs)
and applied these for bioimaging and gene therapy in A549
cells.
194
CSLPs were synthesized via the thin-film dispersion
method using a peptide-based cationic lipid, CDO14, and
further conjugated to UCNPs and siRNA by electrostatic inter-
action. Gene silencing, bioimaging, and cytocompatibility
studies demonstrated that encapsulated siRNA and UCNPs
could eciently be delivered and internalized into the cells
and displayed superior cellular imaging and gene silencing
with CSLPs (Fig. 8a & b). Cytotoxicity studies performed on
A549 cells with an incubation of 48 h showed that CLSPs were
cytocompatible, with cell viability higher than 90% (Fig. 8c).
4.7 Commercial history of lipid-based drug delivery agents
The commercial history of lipidic formulations for drug and
gene delivery can be traced back to the late 20th century when
the first liposomal drug, Doxil (doxorubicin hydrochloride
liposome injection), was approved by the US FDA in 1995.
250
Since then, lipid formulations have become a popular and
eective means of delivering a wide range of therapeutic
agents, including cancer drugs, anti-inflammatory agents, and
nucleic acids for gene therapy. Liposomes, LNPs, and spherical
structures can encapsulate drugs and protect them from rapid
clearance and degradation, enabling targeted delivery to cells
and tissues. The market for lipid-based drug delivery systems
has snowballed in recent years, with several new drugs and
gene therapies using lipidic formulations in development and
commercialization. One of the main advantages of lipidic for-
mulations is their ability to enhance the ecacy of drugs and
reduce their toxic side eects. All these advantages have led to
the approval of several lipid-based drugs for the treatment of
cancer, including Doxil,
250
Camptosar (lipid NPs),
200
and
DaunoXome (daunorubicin citrate liposome injection).
251
In
recent years, the use of lipidic formulations for gene delivery
has also become increasingly popular, leading to the develop-
Fig. 8 (a) Uptake of CSLNs into A549 cells. The uorescence microscope images after treatment with CSLNs (excited by 980 nm light) at concen-
trations of (a) 5 μgμL
1
, (b) 10 μgμL
1
, (c) 15 μgμL
1
and (d) 20 μgμL
1
. (b) Gene silencing mediated by CSLNs. (c) Cytotoxicity of CSLNs/siRNA and
UCNPs/siRNA in A549 cells by MTT assay. Adapted with permission from ref. 194. Copyright 2017, The Royal Society of Chemistry.
Review RSC Pharmaceutics
662 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
ment of new treatments for genetic diseases and other dis-
orders. During the outbreak of SARS-CoV-2, several LNP mRNA
vaccines were developed to fight the disease, such as
Comirnaty and Spikevax.
177,252
Lipid-based delivery systems
are widely accepted in clinical applications and have been
approved by the FDA to deliver various therapeutics, including
drugs and nucleic acids (Table 4).
5. Nanomaterial delivery vehicles for
nucleic acids: promise and challenges
Nucleic acids are widely explored for the treatment of a broad
spectrum of diseases including cancer, heart diseases, genetic
disorders, and viral and bacterial infections. However, the
applicability of nucleic acid in therapeutics is challenging due
to enzymatic degradation, and unfavorable physiocochemical
properties (hydrophilicity and high molecular weight, negative
charge) resulting in reduced cellular uptake, poor in vivo stabi-
lity, rapid excretion or uptake by non-targeted cells.
72,255
Suitable vectors that can overcome inadequate ecacy, suscep-
tibility to enzymatic degradation, low bioavailability, and o-
target side eects are required to increase the therapeutic
ecacy of nucleic acids. Nanomaterials oer exciting possibili-
ties for the therapeutic delivery of nucleic acids, like DNA or
RNA.
223
These nanocarriers hold immense potential for the
pharmaceutical landscape as they revolutionized gene therapy,
RNA interference, and vaccine development. Compared with
traditional methods of delivering nucleic acids into cells,
nanomaterials oer a multitude of advantages. (1) Nucleic
acids on their own have diculty penetrating cell membranes
and reaching their target sites. Nanomaterials can encapsulate
and protect nucleic acids from enzymatic degradation, facili-
tating their cellular uptake and delivery across biological bar-
riers.
234
(2) Nanocarriers can be engineered with specific mole-
cules on their surface that recognize and bind to particular cell
types. This targeted delivery allows researchers to focus the
eects of nucleic acids on the desired cells, reducing side
eects on healthy tissues.
249
(3) Nanocarriers can be designed
to release their nucleic acid cargo in a controlled manner over
time. This sustained release can be crucial for some therapies,
allowing for longer-lasting eects.
256
(4) Some nanocarriers
can be engineered to combine functionalities like delivering
therapeutic molecules and imaging capabilities, oering a
more comprehensive approach to treatment and monitor-
ing.
257
(5) Nanocarriers can be designed to release their cargo
in response to specific stimuli like pH changes or light
exposure.
237
This allows for a more controlled and localized
release of nucleic acids within the body.
While nanomaterials oer exciting possibilities for nucleic
acid delivery, there are also some limitations and potential
downsides to consider. (1) Despite targeting strategies, there is
a risk that nanocarriers could deliver nucleic acids to unin-
tended cell types. This can lead to unwanted side eects or
even toxicity.
74
(2) The bodys immune system may recognize
nanocarriers as foreign invaders and trigger an inflammatory
response. This can limit the eectiveness of the therapy or
cause complications.
258
(3) Developing and manufacturing
safe and eective nanocarriers for nucleic acid delivery can be
complex and expensive. This can hinder the accessibility and
aordability of these therapies. (4) Some nanomaterials may
not be eciently cleared from the body after delivering their
payload. This can raise concerns about potential long-term
eects.
259
Table 4 Overview of the FDA-approved lipid-based therapeutics
S.
no. Commercial
name Developer Date of
approval Lipid Drug Treatment Ref.
1 Diprivan Crucell Feb 23, 1989 Liposome Propofol Induction and maintenance of
anesthesia 251
2 Doxil® Janssen Nov 17, 1995 Liposome Dox Ovarian and breast cancer 250
3 Daunoxome® Galen Apr 8, 1996 Liposome Daunorubicin HIV-associated Kaposis sarcoma 251
4 AmBisome Gilead Sciences Nov 8, 1997 Liposome Amphotericin
BInvasive fungal infections 251
5 Cytosar-U® Pfizer Jan 8, 1999 Lipid NPs Cytarabine Leukemia and lymphoma 200
6 Visudyne Bausch and Lomb Apr 30, 2002 Liposomal Verteporfin Macular degeneration, pathologic, or
ocular histoplasmosis 251
7 Camptosar® Pfizer June 24, 2004 Lipid NPs Irinotecan Metastatic colon or rectum cancer 200
8 VinlonTalon Therapeutics,
Inc. Sept 8, 2012 Liposome Vincristine Acute lymphoblastic leukemia 200
9 Onivyde® Merrimack Oct 22, 2015 Liposome Irinotecan Various metastatic cancers, including
breast, pancreatic, sarcomas, or brain 251
10 Nocita® Aratana therapeutics August 12, 2016 Liposome Bupivacaine Anesthetic 253
11 Vyxeos
CPX-351® Jazz Pharmaceuticals Aug 3, 2017 Liposome Cytarabine :
daunorubicin Acute myeloid leukemia 251
12 Onpattro®
(patisiran) Alnylam
Pharmaceuticals Inc. Aug 8, 2018 Lipid NPs siRNA Hereditary transthyretin amyloidosis 235
13 Givosiran
(Givlaari®) Alnylam
Pharmaceuticals Inc. Nov 20, 2019 Lipid NPs siRNA Hepatic porphyria 254
14 Comirnaty® BioNTech, Pfizer Aug 23, 2021 Lipid NPs mRNA COVID-19 177
15 Spikevax® Moderna Jan 31, 2022 Lipid NPs mRNA COVID-19 252
RSC Pharmaceutics Review
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 663
6. Future strategies
Researchers are actively exploring several strategies to further
improve nanomaterial-based delivery vehicles for nucleic
acids, addressing the current limitations and unlocking their
full potential. (1) Attaching targeting ligands to the surface of
nanocarriers can significantly improve their specificity. These
ligands can bind to receptors present on specific cell types,
directing the delivery of nucleic acids to the desired location.
(2) Developing nanocarriers that respond to specific environ-
mental cues within the body. For example, carriers could
release their cargo in response to changes in pH or tempera-
ture, allowing for targeted release at the disease site.
260
(3)
Modifying the surface properties of nanocarriers to minimize
interactions with the immune system and reduce the risk of
inflammatory responses.
261
This can involve using biocompati-
ble coatings or manipulating surface charges. (4) Developing
nanocarriers from biodegradable polymers that are naturally
broken down and eliminated by the body, reducing long-term
accumulation concerns.
259
(4) Optimizing the rate at which
nucleic acids are released from nanocarriers. This can help to
minimize o-target eects and improve the overall therapeutic
ecacy. (5) Developing nanocarriers that can co-deliver nucleic
acids with other therapeutic agents, such as drugs or imaging
molecules, for combination therapy and improved treatment
outcomes.
262
(6) Creating nanocarriers that can be tracked
within the body using imaging techniques, allowing research-
ers to monitor their delivery pathway and optimize targeting
strategies. (7) Developing ecient and cost-eective pro-
duction methods for well-defined and consistent nanocarriers,
facilitating their wider clinical application.
263
By addressing
these challenges and focusing on these future strategies,
researchers can unlock the full potential of nanomaterial-
based delivery vehicles for nucleic acids. This has the potential
to revolutionize how we treat various diseases, oering more
targeted, eective, and safer therapies based on gene therapy,
RNAi, and DNA vaccines.
7. Conclusion
Nucleic acid-based therapeutics has experienced the trans-
formation from concept into clinical reality in recent years.
The first FDA-approved siRNA-based drug, Onpattro (2018),
provides a platform for further developing NA therapeutics to
cure rare conditions, cancers, and infectious diseases.
235,264
Since then, significant progress has been made to improve
their therapeutic eect, further amplified in the development
of the SARS-Cov-2 vaccine worldwide.
157
However, clinical
availability usually requires modification of NAs and their car-
riers to increase nuclease resistance and enhance cellular
uptake. SNAs with densely packed 3D structures have overcome
some challenges without further modification. SNAs have
minimized the nuclease degradation of delivered NAs.
191
Despite this, significant issues still need to be addressed, most
notably in eectively providing therapeutic NAs to the targeted
site in the required quantity. The stability of SNA structures
can be improved by tuning them with other NPs, resulting in
tissue/organ-specific delivery. Among all the classes of delivery
agents, lipid-based nanocomplexes have shown ideal delivery
eciency at the clinical level.
265
The clinical trials of the
dierent diseases have proved the importance of lipid-based
NPs in NA therapeutics. Over the past decade, several NA thera-
peutics have been in clinical trials, and only a few enjoyed
commercial successes. These points highlight the challenges
in translating new drugs from animals to humans. With the
success story of Onpattro, lipid-based technology is accepted
worldwide and is ready to uplift the next wave of NAs thera-
peutics for vaccination, gene therapy, and protein production.
Undoubtedly, the field of NAs is undergoing a massive expan-
sion, and their potential applications in the treatment of
chronic disorders, immunotherapy, and personalized medi-
cine will ensure their development to revolutionize the bio-
medical field in the near future.
Abbreviations
DOTAP 1,2-Dioleoyl-3-(trimethylammonium) propane
ASO Antisense oligonucleotides
CNMs Carbon nanomaterials
CLs Cationic lipids
FDA Food and drug administration
AuNPs Gold NPs
GFP Green fluorescent protein
ILs Ionizable lipids
IONPs Iron oxide NPs
LNPs Lipid NPs
mRNA Messenger RNA
miRNA MicroRNA
NPs Nanoparticles
NAs Nucleic acids
pDNA Plasmid DNA
PAMAM Poly(amidoamine)
PEG Polyethylene glycol
PEG-DSPE Polyethylene-glycol-2000-1,2-distearyl-3-sn-
phosphatidylethanolamine
PEI Polyethyleneimine
PLGA Polylactide-co-glycolide
PNPs Polymeric NPs
QDs Quantum dots
siRNA Short interfering RNA
SiNPs Silicon NPs
SLNPs Solid lipid NPs
SNAs Spherical nucleic acids
UCNPs Upconversion nanoparticles
ZILs Zwitterionic lipids.
Author contributions
All the authors read and approved the manuscript.
Review RSC Pharmaceutics
664 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
Data availability
No primary research results, software or code has been
included and no new data were generated or analysed as part
of this review.
Conicts of interest
The authors declare that they have no competing interests.
Acknowledgements
This project has received funding from the European Unions
Horizon Europe research and innovation program under Marie
Skłodowska-Curie grant agreement No. 101103113. M. J.
Mehta was supported by the National Research Foundation of
Korea grant (No. 2021R1A2C1007668 and RS-2024-00398030)
funded by the Korean government.
References
1 C. E. Dunbar, K. A. High, J. K. Joung, D. B. Kohn,
K. Ozawa and M. Sadelain, Gene therapy comes of age,
Science, 2018, 359(6372), eaan4672.
2 A. Gupta, J. L. Andresen, R. S. Manan and R. Langer,
Nucleic acid delivery for therapeutic applications, Adv.
Drug Delivery Rev., 2021, 178, 113834.
3 B. B. Mendes, J. Conniot, A. Avital, D. Yao, X. Jiang,
X. Zhou, et al., Nanodelivery of nucleic acids, Nat. Rev.
Methods Primers, 2022, 2(1), 121.
4 T. C. Roberts, R. Langer and M. J. A. Wood, Advances in
oligonucleotide drug delivery, Nat. Rev. Drug Discovery,
2020, 19(10), 673694.
5 J. Kim, C. Hu, C. Moufawad El Achkar, L. E. Black,
J. Douville, A. Larson, et al., Patient-Customized
Oligonucleotide Therapy for a Rare Genetic Disease, N.
Engl. J. Med., 2019, 381(17), 16441652.
6 Y. Yamada, Nucleic Acid DrugsCurrent Status, Issues,
and Expectations for Exosomes, Cancers, 2021, 13(19),
5002.
7 C. H. Jones, C. K. Chen, A. Ravikrishnan, S. Rane and
B. A. Pfeifer, Overcoming Nonviral Gene Delivery Barriers:
Perspective and Future, Mol. Pharmaceutics, 2013, 10(11),
40824098.
8 M. Chehelgerdi, M. Chehelgerdi, O. Q. B. Allela,
R. D. C. Pecho, N. Jayasankar, D. P. Rao, et al., Progressing
nanotechnology to improve targeted cancer treatment:
overcoming hurdles in its clinical implementation, Mol.
Cancer, 2023, 22, 169.
9 A. Graczyk, R. Pawlowska, D. Jedrzejczyk and A. Chworos,
Gold Nanoparticles in Conjunction with Nucleic Acids as
a Modern Molecular System for Cellular Delivery,
Molecules, 2020, 25(1), 204.
10 M. J. Mehta, H. J. Kim, S. B. Lim, M. Naito and K. Miyata,
Recent Progress in the Endosomal Escape Mechanism
and Chemical Structures of Polycations for Nucleic Acid
Delivery, Macromol. Biosci., 2024, 24(4), 2300366.
11 J. K. Patra, G. Das, L. F. Fraceto, E. V. R. Campos,
M. P. Rodriguez-Torres del, L. S. Acosta-Torres, et al.,
Nano based drug delivery systems: recent developments
and future prospects, J. Nanobiotechnol., 2018, 16(1), 71.
12 T. De Serres-Bérard, S. Ait Benichou, D. Jauvin,
M. Boutjdir, J. Puymirat and M. Chahine, Recent Progress
and Challenges in the Development of Antisense
Therapies for Myotonic Dystrophy Type 1, Int. J. Mol. Sci.,
2022, 23(21), 13359.
13 B. Hu, L. Zhong, Y. Weng, L. Peng, Y. Huang, Y. Zhao,
et al., Therapeutic siRNA: state of the art, Signal
Transduction Targeted Ther., 2020, 5(1), 125.
14 P. D. Williams and P. A. Kingston, Plasmid-mediated gene
therapy for cardiovascular disease, Cardiovasc. Res., 2011,
91(4), 565576.
15 C. Uherek and W. Wels, DNA-carrier proteins for targeted
gene delivery, Adv. Drug Delivery Rev., 2000, 44(23), 153
166.
16 C. Rinaldi and M. J. A. Wood, Antisense oligonucleotides:
the next frontier for treatment of neurological disorders,
Nat. Rev. Neurol., 2018, 14(1), 921.
17 A. J. Ward, M. Norrbom, S. Chun, C. F. Bennett and
F. Rigo, Nonsense-mediated decay as a terminating
mechanism for antisense oligonucleotides, Nucleic Acids
Res., 2014, 42(9), 58715879.
18 A. Ruscito and M. C. DeRosa, Small-Molecule Binding
Aptamers: Selection Strategies, Characterization, and
Applications, Front. Chem., 2016, 4, DOI: 10.3389/
fchem.2016.00014.
19 A. Levanova and M. M. Poranen, RNA Interference as a
Prospective Tool for the Control of Human Viral
Infections, Front. Microbiol., 2018, 9, DOI: 10.3389/
fmicb.2018.02151.
20 P. P. Mehta and V. S. Dhapte-Pawar, Novel and
Evolving Therapies for COVID-19 Related Pulmonary
Complications, Am.J.Med.Sci., 2021, 361(5), 557
566.
21 D. P. Bartel, MicroRNAs: genomics, biogenesis, mecha-
nism, and function, Cell, 2004, 116(2), 281297.
22 Z. Wang, F. Schmidt, Y. Weisblum, F. Muecksch,
C. O. Barnes, S. Finkin, et al., mRNA vaccine-elicited anti-
bodies to SARS-CoV-2 and circulating variants, Nature,
2021, 592(7855), 616622.
23 J. D. Beck, D. Reidenbach, N. Salomon, U. Sahin, Ö Türeci
and M Vormehr, , et al., mRNA therapeutics in cancer
immunotherapy, Mol. Cancer, 2021, 20(1), 69.
24 B. Jahrsdörfer and G. J. Weiner, CpG oligodeoxynucleo-
tides for immune stimulation in cancer immunotherapy,
Curr. Opin. Investig. Drugs, 2003, 4(6), 686690.
25 A. Dalpke, S. Zimmermann and K. Heeg, CpG DNA in the
Prevention and Treatment of Infections, BioDrugs, 2002,
16(6), 419431.
RSC Pharmaceutics Review
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 665
26 S. Li, F. C. MacLaughlin, J. G. Fewell, M. Gondo, J. Wang,
F. Nicol, et al., Muscle-specific enhancement of gene
expression by incorporation of SV40 enhancer in the
expression plasmid, Gene. Ther., 2001, 8(6), 494497.
27 R. Lassaunière, C. Polacek, G. J. Gram, A. Frische,
J. L. Tingstedt, M. Krüger, et al., Preclinical evaluation of a
candidate naked plasmid DNA vaccine against
SARS-CoV-2, npj Vaccines, 2021, 6(1), 113.
28 D. Jiang, H. Lee and W. M. Pardridge, Plasmid DNA gene
therapy of the Niemann-Pick C1 mouse with transferrin
receptor-targeted Trojan horse liposomes, Sci. Rep., 2020,
10(1), 13334.
29 T. Malardo, M. E. Batalhão, A. Panunto-Castelo,
L. P. Almeida, E. Padilha, I. C. Fontoura, et al., Low-dose
plasmid DNA treatment increases plasma vasopressin and
regulates blood pressure in experimental endotoxemia,
BMC Immunol., 2012, 13(1), 59.
30 F. Wada, T. Yamamoto, T. Kobayashi, K. Tachibana,
K. R. Ito, M. Hamasaki, et al., Drug discovery and develop-
ment scheme for liver-targeting bridged nucleic acid anti-
sense oligonucleotides, Mol. Ther.Nucleic Acids, 2021, 26,
957969.
31 D. Di Fusco, V. Dinallo, I. Marafini, M. M. Figliuzzi,
B. Romano and G. Monteleone, Antisense
Oligonucleotide: Basic Concepts and Therapeutic
Application in Inflammatory Bowel Disease, Front.
Pharmacol., 2019, 10, DOI: 10.3389/fphar.2019.00305.
32 D. R. Scoles, E. V. Minikel and S. M. Pulst, Antisense oligo-
nucleotides: A primer, Neurol.: Genet., 2019, 5(2), e323.
33 T. P. Prakash, An Overview of Sugar-Modified
Oligonucleotides for Antisense Therapeutics, Chem.
Biodiversity, 2011, 8(9), 16161641.
34 M. A. Passini, J. Bu, A. M. Richards, C. Kinnecom,
S. P. Sardi, L. M. Stanek, et al., Antisense oligonucleotides
delivered to the mouse CNS ameliorate symptoms of
severe spinal muscular atrophy, Sci. Transl. Med., 2011,
3(72), 72ra18.
35 M. Amanat, C. L. Nemeth, A. S. Fine, D. G. Leung and
A. Fatemi, Antisense Oligonucleotide Therapy for the
Nervous System: From Bench to Bedside with Emphasis
on Pediatric Neurology, Pharmaceutics, 2022, 14(11), 2389.
36 A. D. Keefe, S. Pai and A. Ellington, Aptamers as thera-
peutics, Nat. Rev. Drug Discovery, 2010, 9(7), 537550.
37 R. Piyush, K. Rajarshi, A. Chatterjee, R. Khan and S. Ray,
Nucleic acid-based therapy for coronavirus disease 2019,
Heliyon, 2020, 6(9), e05007.
38 B. N. Gawande, J. C. Rohlo, J. D. Carter, I. von Carlowitz,
C. Zhang, D. J. Schneider, et al., Selection of DNA apta-
mers with two modified bases, Proc. Natl. Acad.
Sci. U. S. A., 2017, 114(11), 28982903.
39 C. Tuerk and L. Gold, Systematic evolution of ligands by
exponential enrichment: RNA ligands to bacteriophage T4
DNA polymerase, Science, 1990, 249(4968), 505510.
40 A. D. Ellington and J. W. Szostak, In vitro selection of RNA
molecules that bind specific ligands, Nature, 1990,
346(6287), 818822.
41 J. Zhang, L. P. Smaga, N. S. R. Satyavolu, J. Chan and
Y. Lu, DNA Aptamer-Based Activatable Probes for
Photoacoustic Imaging in Living Mice, J. Am. Chem. Soc.,
2017, 139(48), 1722517228.
42 A. Fire, S. Xu, M. K. Montgomery, S. A. Kostas, S. E. Driver
and C. C. Mello, Potent and specific genetic interference
by double-stranded RNA in Caenorhabditis elegans,
Nature, 1998, 391(6669), 806811.
43 H. Nam, S. H. Ku, H. Y. Yoon, K. Kim, I. C. Kwon,
S. H. Kim, et al., Enhancing Systemic Delivery of
Enzymatically Generated RNAi Nanocomplexes for Cancer
Therapy, Adv. Ther., 2019, 2(6), 1900014.
44 S. A. Jensen, E. S. Day, C. H. Ko, L. A. Hurley, J. P. Luciano,
F. M. Kouri, et al., Spherical Nucleic Acid Nanoparticle
Conjugates as an RNAi-Based Therapy for Glioblastoma,
Sci. Transl. Med., 2013, 5(209), 209ra152209ra152.
45 S. M. Elbashir, J. Harborth, W. Lendeckel, A. Yalcin,
K. Weber and T. Tuschl, Duplexes of 21-nucleotide RNAs
mediate RNA interference in cultured mammalian cells,
Nature, 2001, 411(6836), 494498.
46 Y. Weng, H. Xiao, J. Zhang, X. J. Liang and Y. Huang,
RNAi therapeutic and its innovative biotechnological evol-
ution, Biotechnol. Adv., 2019, 37(5), 801825.
47 M. I. Sajid, M. Moazzam, Y. Cho, S. Kato, A. Xu, J. J. Way,
et al., siRNA Therapeutics for the Therapy of COVID-19
and Other Coronaviruses, Mol. Pharmaceutics, 2021, 18(6),
21052121.
48 L. Li, Y. Ren, X. Wen, Q. Guo, J. Wang, S. Li, et al.,
Endogenous miRNA-Activated DNA Nanomachine for
Intracellular miRNA Imaging and Gene Silencing, Anal.
Chem., 2021, 93(41), 1391913927.
49 W. Dong, P. Wu, M. Qin, S. Guo, H. Liu, X. Yang, et al.,
Multipotent miRNA Sponge-Loaded Magnetic
Nanodroplets with Ultrasound/Magnet-Assisted Delivery
for Hepatocellular Carcinoma Therapy, Mol. Pharmaceutics,
2020, 17(8), 28912910.
50 S. Qin, X. Tang, Y. Chen, K. Chen, N. Fan, W. Xiao, et al.,
mRNA-based therapeutics: powerful and versatile tools to
combat diseases, Signal Transduction Targeted Ther., 2022,
7, 166.
51 N. Chaudhary, D. Weissman and K. A. Whitehead, mRNA
vaccines for infectious diseases: principles, delivery and
clinical translation, Nat. Rev. Drug Discovery, 2021, 20(11),
817838.
52 E. V. Bailey and F. A. Wilson, Vaccine Uptake in the US
After Full Food and Drug Administration Approval of the
BNT162b2 mRNA COVID-19 Vaccine, JAMA Netw. Open,
2022, 5(4), e226108.
53 C. Zhang, G. Maruggi, H. Shan and J. Li, Advances in
mRNA Vaccines for Infectious Diseases, Front. Immunol.,
2019, 10, DOI: 10.3389/fimmu.2019.00594.
54 J. Devoldere, K. Peynshaert, H. Dewitte, C. Vanhove, L. De
Groef, L. Moons, et al., Non-viral delivery of chemically
modified mRNA to the retina: Subretinal versus intra-
vitreal administration, J. Controlled Release, 2019, 307,
315330.
Review RSC Pharmaceutics
666 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
55 N. Pardi, M. J. Hogan, F. W. Porter and D. Weissman,
mRNA vaccinesa new era in vaccinology, Nat. Rev. Drug
Discovery, 2018, 17(4), 261279.
56 S. Xu, K. Yang, R. Li and L. Zhang, mRNA Vaccine Era-
Mechanisms, Drug Platform and Clinical Prospection,
Int. J. Mol. Sci., 2020, 21(18), 6582.
57 J. E. Wooldridge and G. J. Weiner, CpG DNA and cancer
immunotherapy: orchestrating the antitumor immune
response, Curr. Opin. Oncol., 2003, 15(6), 440445.
58 A. M. Krieg, Antitumor applications of stimulating toll-
like receptor 9 with CpG oligodeoxynucleotides, Curr.
Oncol. Rep., 2004, 6(2), 8895.
59 P. P. Ho, P. Fontoura, P. J. Ruiz, L. Steinman and
H. Garren, An immunomodulatory GpG oligonucleotide
for the treatment of autoimmunity via the innate and
adaptive immune systems, J. Immunol., 2003, 171(9),
49204926.
60 S. Alipour, A. Mahdavi and A. Abdoli, The eects of
CpG-ODNs and Chitosan adjuvants on the elicitation of
immune responses induced by the HIV-1-Tat-based candi-
date vaccines in mice, Pathogens and Disease, 2017, 75(2),
ftx013.
61 H. Kim, W. Zhang, J. Hwang, E. K. An, Y. K. Choi,
E. Moon, et al., Carrier-free micellar CpG interacting with
cell membrane for enhanced immunological treatment of
HIV-1, Biomaterials, 2021, 277, 121081.
62 H. Zhao, Q. Han, A. Yang, Y. Wang, G. Wang, A. Lin, et al.,
CpG-C ODN M362 as an immunoadjuvant for HBV thera-
peutic vaccine reverses the systemic tolerance against
HBV, Int. J. Biol. Sci., 2022, 18(1), 154165.
63 J. G. McHutchison, B. R. Bacon, S. C. Gordon, E. Lawitz,
M. Shiman, N. H. Afdhal, et al., Phase 1B, randomized,
double-blind, dose-escalation trial of CPG 10101 in
patients with chronic hepatitis C virus, Hepatology, 2007,
46(5), 13411349.
64 N. Hanagata, CpG oligodeoxynucleotide nanomedicines
for the prophylaxis or treatment of cancers, infectious dis-
eases, and allergies, Int. J. Nanomed., 2017, 12, 515531.
65 T. H. Kim, D. Kim, H. Lee, M. H. Kwak, S. Park, Y. Lee and
H. J. Kwon, CpG-DNA induces bacteria-reactive IgM
enhancing phagocytic activity against Staphylococcus
aureus infection, BMB Rep., 2019, 52(11), 635640.
66 R. Q. Prado, T. B. Bertolini, A. R. Piñeros, A. F. Gembre,
S. G. Ramos, C. L. Silva, et al., Attenuation of experimental
asthma by mycobacterial protein combined with CpG
requires a TLR9-dependent IFN-γ-CCR2 signalling circuit,
Clin. Exp. Allergy, 2015, 45(9), 14591471.
67 H. T. Li, Z. G. Chen, H. Liu, J. Ye, X. L. Zou, Y. H. Wang,
et al., Treatment of allergic rhinitis with CpG oligodeoxy-
nucleotides alleviates the lower airway outcomes of com-
bined allergic rhinitis and asthma syndrome via a mecha-
nism that possibly involves in TSLP, Exp. Lung Res., 2016,
42(6), 322333.
68 D. E. Fonseca and J. N. Kline, Use of CpG oligonucleotides
in treatment of asthma and allergic disease, Adv. Drug
Delivery Rev., 2009, 61(3), 256262.
69 H. Hadji and K. Bouchemal, Eect of micro- and nano-
particle shape on biological processes, J. Controlled
Release, 2022, 342,93110.
70 R. A. Petros and J. M. DeSimone, Strategies in the design
of nanoparticles for therapeutic applications, Nat. Rev.
Drug Discovery, 2010, 9(8), 615627.
71 F. U. Din, W. Aman, I. Ullah, O. S. Qureshi, O. Mustapha,
S. Shafique, et al.,Eective use of nanocarriers as drug
delivery systems for the treatment of selected tumors,
Int. J. Nanomed., 2017, 12, 72917309.
72 Y. Qin, L. Ou, L. Zha, Y. Zeng and L. Li, Delivery of nucleic
acids using nanomaterials, Mol. Biomed., 2023, 4, 48.
73 A. Baker, J. Lorch, D. VanderWeele and B. Zhang, Smart
Nanocarriers for the Targeted Delivery of Therapeutic
Nucleic Acid for Cancer Immunotherapy, Pharmaceutics,
2023, 15(6), 1743.
74 H. J. Vaughan, J. J. Green and S. Y. Tzeng, Cancer-
Targeting Nanoparticles for Combinatorial Nucleic Acid
Delivery, Adv. Mater., 2020, 32(13), e1901081.
75 Y. Jiang, S. Huo, J. Hardie, X. J. Liang and V. M. Rotello,
Progress and perspective of inorganic nanoparticle-based
siRNA delivery systems, Expert Opin. Drug Delivery, 2016,
13(4), 547559.
76 R. Zhao, J. Xiang, B. Wang, L. Chen and S. Tan, Recent
Advances in the Development of Noble Metal NPs for
Cancer Therapy, in Bioinorganic Chemistry and
Applications, ed. V. De Matteis, 2022, vol. 2022, pp. 114.
77 C. Amri, A. K. Shukla and J. H. Lee, Recent Advancements
in Nanoparticle-Based Optical Biosensors for Circulating
Cancer Biomarkers, Materials, 2021, 14(6), 1339.
78 S. Walia, A. Guliani and A. Acharya, A Theragnosis Probe
Based on BSA/HSA-Conjugated Biocompatible Fluorescent
Silicon Nanomaterials for Simultaneous in Vitro
Cholesterol Euxing and Cellular Imaging of Macrophage
Cells, ACS Sustainable Chem. Eng., 2017, 5(2), 14251435.
79 D. Holmannova, P. Borsky, T. Svadlakova, L. Borska and
Z. Fiala, Carbon Nanoparticles and Their Biomedical
Applications, Appl. Sci., 2022, 12(15), 7865.
80 A. Ali, H. Zafar, M. Zia, I. ul Haq, A. R. Phull, J. S. Ali,
et al., Synthesis, characterization, applications, and chal-
lenges of iron oxide nanoparticles, Nanotechnol. Sci. Appl.,
2016, 9,4967.
81 A. Gagliardi, E. Giuliano, E. Venkateswararao, M. Fresta,
S. Bulotta, V. Awasthi, et al., Biodegradable Polymeric
Nanoparticles for Drug Delivery to Solid Tumors, Front.
Pharmacol., 2021, 12, DOI: 10.3389/fphar.2021.601626.
82 S. Adepu and S. Ramakrishna, Controlled Drug Delivery
Systems: Current Status and Future Directions, Molecules,
2021, 26(19), 5905.
83 S. Burgio, L. Noori, A. Marino Gammazza, C. Campanella,
M. Logozzi, S. Fais, et al., Extracellular Vesicles-Based
Drug Delivery Systems: A New Challenge and the
Exemplum of Malignant Pleural Mesothelioma, Int. J. Mol.
Sci., 2020, 21(15), 5432.
84 E. Kahraman, S. Güngör and Y. Özsoy, Potential enhance-
ment and targeting strategies of polymeric and lipid-
RSC Pharmaceutics Review
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 667
based nanocarriers in dermal drug delivery, Ther. Delivery,
2017, 8(11), 967985.
85 A. Hiwrale, S. Bharati, P. Pingale and A. Rajput,
Nanofibers: A current era in drug delivery system, Heliyon,
2023, 9(9), e18917.
86 S. M. S. Shahriar, J. Mondal, M. N. Hasan, V. Revuri,
D. Y. Lee and Y. K. Lee, Electrospinning Nanofibers for
Therapeutics Delivery, Nanomaterials, 2019, 9(4), 532.
87 Y. Song, W. Song, X. Lan, W. Cai and D. Jiang, Spherical
nucleic acids: Organized nucleotide aggregates as versatile
nanomedicine, Aggregate, 2022, 3(1), e120.
88 F. Xu, Q. Xia and P. Wang, Rationally Designed DNA
Nanostructures for Drug Delivery, Front. Chem., 2020, 8,
751.
89 Y. Wei, X. Mo, P. Zhang, Y. Li, J. Liao, Y. Li, et al.,
Directing Stem Cell Dierentiation via Electrochemical
Reversible Switching between Nanotubes and Nanotips of
Polypyrrole Array, ACS Nano, 2017, 11(6), 59155924.
90 Z. Gao, T. Ma, E. Zhao, D. Docter, W. Yang, R. H. Stauber,
et al., Small is Smarter: Nano MRI Contrast Agents
Advantages and Recent Achievements, Small, 2016, 12(5),
556576.
91 M. Jiao, P. Zhang, J. Meng, Y. Li, C. Liu, X. Luo, et al.,
Recent advancements in biocompatible inorganic nano-
particles towards biomedical applications, Biomater. Sci.,
2018, 6(4), 726745.
92 Á. Artiga, I. Serrano-Sevilla, L. D. Matteis, S. G. Mitchell
and J. M. de la Fuente, Current status and future perspec-
tives of gold nanoparticle vectors for siRNA delivery,
J. Mater. Chem. B, 2019, 7(6), 876896.
93 S. Wilhelm, A. J. Tavares, Q. Dai, S. Ohta, J. Audet,
H. F. Dvorak, et al., Analysis of nanoparticle delivery to
tumours, Nat. Rev. Mater., 2016, 1(5), 112.
94 J. Ou, Z. Zhou, Z. Chen and H. Tan, Optical Diagnostic
Based on Functionalized Gold Nanoparticles, Int. J. Mol.
Sci., 2019, 20(18), E4346.
95 J. Wang, A. M. Potocny, J. Rosenthal and E. S. Day, Gold
Nanoshell-Linear Tetrapyrrole Conjugates for Near
Infrared-Activated Dual Photodynamic and Photothermal
Therapies, ACS Omega, 2020, 5(1), 926940.
96 From the American Association of Neurological Surgeons
(AANS), American Society of Neuroradiology (ASNR),
Cardiovascular and Interventional Radiology Society of
Europe (CIRSE), Canadian Interventional Radiology
Association (CIRA), Congress of Neurological Surgeons
(CNS), European Society of Minimally Invasive
Neurological Therapy (ESMINT), European Society of
Neuroradiology (ESNR), European Stroke Organization
(ESO), Society for Cardiovascular Angiography and
Interventions (SCAI), Society of Interventional Radiology
(SIR), Society of NeuroInterventional Surgery (SNIS),
World Stroke Organization (WSO), D. Sacks, B. Baxter,
B. C. V. Campbell, J. S. Carpenter, C. Cognard, et al.,
Multisociety Consensus Quality Improvement Revised
Consensus Statement for Endovascular Therapy of Acute
Ischemic Stroke, Int. J. Stroke, 2018, 13(6), 612632.
97 A. Graczyk, R. Pawlowska and A. Chworos, Gold
Nanoparticles as Carriers for Functional RNA
Nanostructures, Bioconjugate Chem., 2021, 32(8), 1667
1674.
98 M. E. Kyriazi, D. Giust, A. H. El-Sagheer, P. M. Lackie,
O. L. Muskens, T. Brown, et al., Multiplexed mRNA
Sensing and Combinatorial-Targeted Drug Delivery Using
DNA-Gold Nanoparticle Dimers, ACS Nano, 2018, 12(4),
33333340.
99 K. D. Wegner, P. T. Lanh, T. Jennings, E. Oh, V. Jain,
S. M. Fairclough, et al., Influence of Luminescence
Quantum Yield, Surface Coating, and Functionalization of
Quantum Dots on the Sensitivity of Time-Resolved FRET
Bioassays, ACS Appl. Mater. Interfaces, 2013, 5(8), 2881
2892.
100 I. V. Martynenko, A. P. Litvin, F. Purcell-Milton,
A. V. Baranov, A. V. Fedorov and Y. K. Gunko, Application
of semiconductor quantum dots in bioimaging and bio-
sensing, J. Mater. Chem. B, 2017, 5(33), 67016727.
101 S. Walia, C. Sharma and A. Acharya, Biocompatible
Fluorescent Nanomaterials for Molecular Imaging
Applications, in Nanomaterial - Based Biomedical
Applications in Molecular Imaging, Diagnostics and Therapy,
ed. A. Acharya, Springer, Singapore, 2020. p. 2753. DOI:
10.1007/978-981-15-4280-0_3.
102 D. Aldakov, A. Lefrançois and P. Reiss, Ternary and qua-
ternary metal chalcogenide nanocrystals: synthesis, pro-
perties and applications, J. Mater. Chem. C, 2013, 1(24),
37563776.
103 Y. Lou, Y. Zhao, J. Chen and J. J. Zhu, Metal ions optical
sensing by semiconductor quantum dots, J. Mater. Chem.
C, 2013, 2(4), 595613.
104 S. Liu and X. Su, The synthesis and application of IIIIVI
type quantum dots, RSC Adv., 2014, 4(82), 4341543428.
105 G. Xu, S. Zeng, B. Zhang, M. T. Swihart, K. T. Yong and
P. N. Prasad, New Generation Cadmium-Free Quantum
Dots for Biophotonics and Nanomedicine, Chem. Rev.,
2016, 116(19), 1223412327.
106 O. Chen, H. Wei, A. Maurice, M. Bawendi and P. Reiss,
Pure colors from coreshell quantum dots, MRS Bull.,
2013, 38(9), 696702.
107 M. A. Cotta, Quantum Dots and Their Applications: What
Lies Ahead?, ACS Appl. Nano Mater., 2020, 3(6), 4920
4924.
108 Y. Ma, G. Mao, W. Huang, G. Wu, W. Yin, X. Ji, et al.,
Quantum Dot Nanobeacons for Single RNA Labeling and
Imaging, J. Am. Chem. Soc., 2019, 141(34), 1345413458.
109 G. Lin, T. Chen, J. Zou, Y. Wang, X. Wang, J. Li, et al.,
Quantum Dots-siRNA Nanoplexes for Gene Silencing in
Central Nervous System Tumor Cells, Front. Pharmacol.,
2017, 8, DOI: 10.3389/fphar.2017.00182.
110 X. Ji, H. Wang, B. Song, B. Chu and Y. He, Silicon
Nanomaterials for Biosensing and Bioimaging Analysis,
Front. Chem., 2018, 6, 38.
111 Y. Park, J. Yoo, M. H. Kang, W. Kwon and J. Joo,
Photoluminescent and biodegradable porous silicon
Review RSC Pharmaceutics
668 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
nanoparticles for biomedical imaging, J. Mater. Chem. B,
2019, 7(41), 62716292.
112 F. Peng, Y. Su, Y. Zhong, C. Fan, S. T. Lee and Y. He,
Silicon Nanomaterials Platform for Bioimaging,
Biosensing, and Cancer Therapy, Acc. Chem. Res., 2014,
47(2), 612623.
113 C. J. T. Robidillo and J. G. C. Veinot, Functional Bio-in-
organic Hybrids from Silicon Quantum Dots and
Biological Molecules, ACS Appl. Mater. Interfaces, 2020,
12(47), 5225152270.
114 A. Chaix, E. Cueto-Diaz, A. Delalande, N. Knezevic,
P. Midoux, J. O. Durand, et al., Amino-acid functionalized
porous silicon nanoparticles for the delivery of pDNA,
RSC Adv., 2019, 9(55), 3189531899.
115 T. Y. Wang, C. Y. Chen, C. M. Wang, Y. Z. Tan and
W. S. Liao, Multicolor Functional Carbon Dots via One-
Step Refluxing Synthesis, ACS Sens., 2017, 2(3), 354363.
116 G. E. LeCroy, S. K. Sonkar, F. Yang, L. M. Veca, P. Wang,
K. N. Tackett, et al., Toward Structurally Defined Carbon
Dots as Ultracompact Fluorescent Probes, ACS Nano, 2014,
8(5), 45224529.
117 Q. Wang, C. Zhang, G. Shen, H. Liu, H. Fu and D. Cui,
Fluorescent carbon dots as an ecient siRNA nanocarrier
for its interference therapy in gastric cancer cells,
J. Nanobiotechnol., 2014, 12(1), 58.
118 S. M. Dadfar, D. Camozzi, M. Darguzyte, K. Roemhild,
P. Varvarà, J. Metselaar, et al., Size-isolation of superpara-
magnetic iron oxide nanoparticles improves MRI, MPI
and hyperthermia performance, J. Nanobiotechnol., 2020,
18(1), 22.
119 G. Tang, J. He, J. Liu, X. Yan and K. Fan, Nanozyme for
tumor therapy: Surface modification matters, Exploration,
2021, 1(1), 7589.
120 Y. W. Jun, J. H. Lee and J. Cheon, Chemical Design of
Nanoparticle Probes for High-Performance Magnetic
Resonance Imaging, Angew. Chem., Int. Ed., 2008, 47(28),
51225135.
121 Y. Jun, J. Seo and J. Cheon, Nanoscaling Laws of Magnetic
Nanoparticles and Their Applicabilities in Biomedical
Sciences, Acc. Chem. Res., 2008, 41(2), 179189.
122 Q. Xu, T. Zhang, Q. Wang, X. Jiang, A. Li, Y. Li, et al.,
Uniformly sized iron oxide nanoparticles for ecient gene
delivery to mesenchymal stem cells, Int. J. Pharm., 2018,
552(1), 443452.
123 H. Zhang, M. Y. Lee, M. G. Hogg, J. S. Dordick and
S. T. Sharfstein, Gene Delivery in Three-Dimensional Cell
Cultures by Superparamagnetic Nanoparticles, ACS Nano,
2010, 4(8), 47334743.
124 P. S. Kowalski, A. Rudra, L. Miao and D. G. Anderson,
Delivering the Messenger: Advances in Technologies for
Therapeutic mRNA Delivery, Mol. Ther., 2019, 27(4), 710
728.
125 N. Kamaly, B. Yameen, J. Wu and O. C. Farokhzad,
Degradable Controlled-Release Polymers and Polymeric
Nanoparticles: Mechanisms of Controlling Drug Release,
Chem. Rev., 2016, 116(4), 26022663.
126 A. Zielińska, F. Carreiró, A. M. Oliveira, A. Neves, B. Pires,
D. N. Venkatesh, et al., Polymeric Nanoparticles:
Production, Characterization, Toxicology and
Ecotoxicology, Molecules, 2020, 25(16), E3731.
127 R. A. Youness, A. M. Al-Mahallawi, F. H. Mahmoud,
H. Atta, M. Braoudaki and S. A. Fahmy, Oral Delivery of
Psoralidin by Mucoadhesive Surface-Modified Bilosomes
Showed Boosted Apoptotic and Necrotic Eects against
Breast and Lung Cancer Cells, Polymers, 2023, 15(6), 1464.
128 A. Fabozzi, F. D. Sala, M. di Gennaro, N. Solimando,
M. Pagliuca and A. Borzacchiello, Polymer based nano-
particles for biomedical applications by microfluidic tech-
niques: from design to biological evaluation, Polym.
Chem., 2021, 12(46), 66676687.
129 M. Li, M. Zhao, Y. Fu, Y. Li, T. Gong, Z. Zhang, et al.,
Enhanced intranasal delivery of mRNA vaccine by over-
coming the nasal epithelial barrier via intra- and paracel-
lular pathways, J. Controlled Release, 2016, 228,919.
130 J. Yan, R. Chen, H. Zhang and J. D. Bryers, Injectable
Biodegradable Chitosan-Alginate 3D Porous Gel Scaold
for mRNA Vaccine Delivery, Macromol. Biosci., 2019, 19(2),
e1800242.
131 J. S. Ananta, R. Paulmurugan and T. F. Massoud,
Nanoparticle-Delivered Antisense MicroRNA-21 Enhances
the Eects of Temozolomide on Glioblastoma Cells, Mol.
Pharmaceutics, 2015, 12(12), 45094517.
132 S. Zhu, H. Xing, P. Gordiichuk, J. Park and C. A. Mirkin,
PLGA Spherical Nucleic Acids, Adv. Mater., 2018, 30(22),
1707113.
133 M. J. Mitchell, M. M. Billingsley, R. M. Haley,
M. E. Wechsler, N. A. Peppas and R. Langer, Engineering
precision nanoparticles for drug delivery, Nat. Rev. Drug
Discovery, 2021, 20(2), 101124.
134 M. A. Hutnick, S. Ahsanuddin, L. Guan, M. Lam,
E. D. Baron and J. K. Pokorski, PEGylated Dendrimers as
Drug Delivery Vehicles for the Photosensitizer Silicon
Phthalocyanine Pc 4 for Candidal Infections,
Biomacromolecules, 2017, 18(2), 379385.
135 V. Percec, M. Peterca, A. E. Dulcey, M. R. Imam,
S. D. Hudson, S. Nummelin, et al., Hollow spherical
supramolecular dendrimers, J. Am. Chem. Soc., 2008,
130(39), 1307913094.
136 L. Palmerston Mendes, J. Pan and V. P. Torchilin,
Dendrimers as Nanocarriers for Nucleic Acid and Drug
Delivery in Cancer Therapy, Molecules, 2017, 22(9), 1401.
137 J. Wu, W. Huang and Z. He, Dendrimers as Carriers for
siRNA Delivery and Gene Silencing: A Review, Sci. World J.,
2013, 2013, e630654.
138 F. Palombarini, S. Masciarelli, A. Incocciati, F. Liccardo,
E. Di Fabio, A. Iazzetti, et al., Self-assembling ferritin-den-
drimer nanoparticles for targeted delivery of nucleic acids
to myeloid leukemia cells, J. Nanobiotechnol., 2021, 19(1), 172.
139 S. L. Mekuria, J. Li, C. Song, Y. Gao, Z. Ouyang, M. Shen,
et al., Facile Formation of PAMAM Dendrimer
Nanoclusters for Enhanced Gene Delivery and Cancer
Gene Therapy, ACS Appl. Bio Mater., 2021, 4(9), 71687175.
RSC Pharmaceutics Review
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 669
140 M. Labieniec-Watala and C. Watala, PAMAM Dendrimers:
Destined for Success or Doomed to Fail? Plain and
Modified PAMAM Dendrimers in the Context of
Biomedical Applications, J. Pharm. Sci., 2015, 104(1), 214.
141 K. Furuno, K. Suzuki and S. Sakai, Gelatin-Based
Electrospun Nanofibers Cross-Linked Using Horseradish
Peroxidase for Plasmid DNA Delivery, Biomolecules, 2022,
12(11), 1638.
142 H. Cao, X. Jiang, C. Chai and S. Y. Chew, RNA interference
by nanofiber-based siRNA delivery system, J. Controlled
Release, 2010, 144(2), 203212.
143 C. Pinese, J. Lin, U. Milbreta, M. Li, Y. Wang, K. W. Leong,
et al., Sustained delivery of siRNA/mesoporous silica
nanoparticle complexes from nanofiber scaolds for long-
term gene silencing, Acta Biomater., 2018, 76, 164177.
144 S. Hong, D. W. Choi, H. N. Kim, C. G. Park, W. Lee and
H. H. Park, Protein-Based Nanoparticles as Drug Delivery
Systems, Pharmaceutics, 2020, 12(7), 604.
145 A. Jain, S. K. Singh, S. K. Arya, S. C. Kundu and S. Kapoor,
Protein Nanoparticles: Promising Platforms for Drug
Delivery Applications, ACS Biomater. Sci. Eng., 2018, 4(12),
39393961.
146 F. F. An and X. H. Zhang, Strategies for Preparing
Albumin-based Nanoparticles for Multifunctional
Bioimaging and Drug Delivery, Theranostics, 2017, 7(15),
36673689.
147 I. Cucinotto, L. Fiorillo, S. Gualtieri, M. Arbitrio, D. Ciliberto,
N. Staropoli, et al., Nanoparticle Albumin Bound Paclitaxel in
the Treatment of Human Cancer: Nanodelivery Reaches
Prime-Time?, J. Drug Delivery, 2013, 2013, 905091.
148 W. Ma, A. Saccardo, D. Roccatano, D. Aboagye-Mensah,
M. Alkaseem, M. Jewkes, et al., Modular assembly of pro-
teins on nanoparticles, Nat. Commun., 2018, 9(1), 1489.
149 M. Mendes, J. Sousa, A. Pais and C. Vitorino, 4 - Clinical
applications of nanostructured drug delivery systems:
From basic research to translational medicine, in Core-
Shell Nanostructures for Drug Delivery and Theranostics, ed.
M. L. Focarete and A. Tampieri, Woodhead Publishing,
2018. p. 43116. (Woodhead Publishing Series in
Biomaterials). Available from: https://www.sciencedirect.
com/science/article/pii/B9780081021989000041.
150 G. Wei, Y. Wang, X. Huang, H. Hou and S. Zhou, Peptide-
Based Nanocarriers for Cancer Therapy, Small Methods,
2018, 2(9), 1700358.
151 K. Han, Z. Ma and H. Han, Functional peptide-based
nanoparticles for photodynamic therapy, J. Mater. Chem.
B, 2017, 6(1), 2538.
152 M. Abbas, H. H. Susapto and C. A. E. Hauser, Synthesis
and Organization of Gold-Peptide Nanoparticles for
Catalytic Activities, ACS Omega, 2022, 7(2), 20822090.
153 N. Jia, J. Ma, Y. Gao, H. Hu, D. Chen, X. Zhao, et al.,
HA-Modified R8-Based Bola-Amphiphile Nanocomplexes
for Eective Improvement of siRNA Delivery Eciency,
ACS Biomater. Sci. Eng., 2020, 6(4), 20842093.
154 M. Chandler and K. A. Afonin, Smart-Responsive Nucleic
Acid Nanoparticles (NANPs) with the Potential to
Modulate Immune Behavior, Nanomaterials, 2019, 9(4),
611.
155 A. J. Mukalel, R. S. Riley, R. Zhang and M. J. Mitchell,
Nanoparticles for nucleic acid delivery: Applications in
cancer immunotherapy, Cancer Lett., 2019, 458, 102112.
156 G. Huang, C. Su, L. Wang, Y. Fei and J. Yang, The
Application of Nucleic Acid ProbeBased Fluorescent
Sensing and Imaging in Cancer Diagnosis and Therapy,
Front. Chem., 2021, 9, DOI: 10.3389/fchem.2021.705458.
157 J. A. Kulkarni, D. Witzigmann, S. B. Thomson, S. Chen,
B. R. Leavitt, P. R. Cullis, et al., The current landscape of
nucleic acid therapeutics, Nat. Nanotechnol., 2021, 16(6),
630643.
158 W. Ma, Y. Zhan, Y. Zhang, C. Mao, X. Xie and Y. Lin,
The biological applications of DNA nanomaterials: current
challenges and future directions, Sig. Transduct. Target
Ther., 2021, 6, 351. Available from: https://pubmed.ncbi.
nlm.nih.gov/34620843/.
159 S. Walia, A. R. Chandrasekaran, B. Chakraborty and
D. Bhatia, Aptamer-Programmed DNA Nanodevices for
Advanced, Targeted Cancer Theranostics, ACS Appl. Bio
Mater., 2021, 4(7), 53925404.
160 U. Singh, V. Morya, A. Rajwar, A. R. Chandrasekaran,
B. Datta, C. Ghoroi and D. Bhatia, DNA-Functionalized
Nanoparticles for Targeted Biosensing and Biological
Applications, ACS Omega, 2020, 5(48), 3076730774, Available
from: https://pubmed.ncbi.nlm.nih.gov/33324786/.
161 N. L. Rosi, D. A. Giljohann, C. S. Thaxton, A. K. R. Lytton-
Jean, M. S. Han and C. A. Mirkin, Oligonucleotide-modi-
fied gold nanoparticles for intracellular gene regulation,
Science, 2006, 312(5776), 10271030.
162 H. Li, B. Zhang, X. Lu, X. Tan, F. Jia, Y. Xiao, et al.,
Molecular spherical nucleic acids, Proc. Natl. Acad.
Sci. U. S. A., 2018, 115(17), 43404344.
163 Y. W. Cao, R. Jin and C. A. Mirkin, DNA-Modified
CoreShell Ag/Au Nanoparticles, J. Am. Chem. Soc., 2001,
123(32), 79617962.
164 Y. Ji and J. B. Lee, DNA microsponge-templated growth of
metal nanoparticles for signal-enhanced colorimetric
detection, Appl. Surf. Sci., 2021, 569, 151028.
165 K. L. Young, A. W. Scott, L. Hao, S. E. Mirkin, G. Liu and
C. A. Mirkin, Hollow spherical nucleic acids for intracellu-
lar gene regulation based upon biocompatible silica
shells, Nano Lett., 2012, 12(7), 38673871.
166 B. Deng, B. Ma, Y. Ma, P. Cao, X. Leng, P. Huang, et al.,
Doxorubicin and CpG loaded liposomal spherical nucleic
acid for enhanced Cancer treatment, J. Nanobiotechnol.,
2022, 20(1), 140.
167 C. E. Callmann, C. D. Kusmierz, J. W. Dittmar, L. Broger
and C. A. Mirkin, Impact of Liposomal Spherical Nucleic
Acid Structure on Immunotherapeutic Function, ACS
Cent. Sci., 2021, 7(5), 892899.
168 C. A. Mirkin, C. Laramy and K. Skakuj, The Power of
Spheres, Nature, 2019, 576(7785), S3S7.
169 C. H. J. Choi, L. Hao, S. P. Narayan, E. Auyeung and
C. A. Mirkin, Mechanism for the endocytosis of spherical
Review RSC Pharmaceutics
670 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
nucleic acid nanoparticle conjugates, Proc. Natl. Acad.
Sci. U. S. A., 2013, 110(19), 76257630.
170 S. C. P. Williams, Spherical nucleic acids: A whole new
ball game, Proc. Natl. Acad. Sci. U. S. A., 2013, 110(33),
1323113233.
171 J. R. Melamed, N. L. Kreuzberger, R. Goyal and E. S. Day,
Spherical Nucleic Acid Architecture Can Improve the
Ecacy of Polycation-Mediated siRNA Delivery, Mol.
Ther.Nucleic Acids, 2018, 12, 207219.
172 S. Wang, L. Qin, G. Yamankurt, K. Skakuj, Z. Huang,
P. C. Chen, et al., Rational vaccinology with spherical
nucleic acids, Proc. Natl. Acad. Sci. U. S. A., 2019, 116(21),
1047310481.
173 H. Tr and P. As, Gene Regulation Using Spherical Nucleic
Acids to Treat Skin Disorders, Pharmaceuticals, 2020,
13(11), Available from: https://pubmed.ncbi.nlm.nih.gov/
33147737/.
174 P. S. Randeria, M. A. Seeger, X. Q. Wang, H. Wilson,
D. Shipp, C. A. Mirkin, et al., siRNA-based spherical
nucleic acids reverse impaired wound healing in diabetic
mice by ganglioside GM3 synthase knockdown, Proc. Natl.
Acad. Sci. U. S. A., 2015, 112(18), 55735578.
175 W. Poon, B. R. Kingston, B. Ouyang, W. Ngo and
W. C. W. Chan, A framework for designing delivery
systems, Nat. Nanotechnol., 2020, 15(10), 819829.
176 P. R. Cullis and M. J. Hope, Lipid Nanoparticle Systems
for Enabling Gene Therapies, Mol. Ther., 2017, 25(7),
14671475.
177 Y. Eygeris, M. Gupta, J. Kim and G. Sahay, Chemistry of
Lipid Nanoparticles for RNA Delivery, Acc. Chem. Res.,
2022, 55(1), 212.
178 L. Miao, L. Li, Y. Huang, D. Delcassian, J. Chahal, J. Han,
et al., Delivery of mRNA vaccines with heterocyclic lipids
increases anti-tumor ecacy by STING-mediated immune
cell activation, Nat. Biotechnol., 2019, 37(10), 11741185.
179 M. Sun, U. J. Dang, Y. Yuan, A. M. Psaras, O. Osipitan,
T. A. Brooks, et al., Optimization of DOTAP/chol Cationic
Lipid Nanoparticles for mRNA, pDNA, and
Oligonucleotide Delivery, AAPS PharmSciTech, 2022, 23(5),
135.
180 Z. Ye, J. Chen, X. Zhao, Y. Li, J. Harmon, C. Huang, et al.,
In Vitro Engineering Chimeric Antigen Receptor
Macrophages and T Cells by Lipid Nanoparticle-Mediated
mRNA Delivery, ACS Biomater. Sci. Eng., 2022, 8(2), 722
733.
181 Y. Hagino, I. A. Khalil, S. Kimura, K. Kusumoto and
H. Harashima, GALA-Modified Lipid Nanoparticles for
the Targeted Delivery of Plasmid DNA to the Lungs, Mol.
Pharmaceutics, 2021, 18(3), 878888.
182 A. Algarni, E. H. Pilkington, E. J. A. Suys, H. Al-Wassiti,
C. W. Pouton and N. P. Truong, In vivo delivery of plasmid
DNA by lipid nanoparticles: the influence of ionizable cat-
ionic lipids on organ-selective gene expression, Biomater.
Sci., 2022, 10(11), 29402952.
183 S. Scalzo, A. K. Santos, H. A. Ferreira, P. A. Costa,
P. H. Prazeres, N. J. da Silva, et al., Ionizable Lipid
Nanoparticle-Mediated Delivery of Plasmid DNA in
Cardiomyocytes, IJN, 2022, 17, 28652881.
184 R. Pattipeiluhu, G. Arias-Alpizar, G. Basha, K. Y. T. Chan,
J. Bussmann, T. H. Sharp, et al., Anionic Lipid
Nanoparticles Preferentially Deliver mRNA to the Hepatic
Reticuloendothelial System, Adv. Mater., 2022, 34(16),
2201095.
185 B. Hu, B. Li, K. Li, Y. Liu, C. Li, L. Zheng, et al.,
Thermostable ionizable lipid-like nanoparticle (iLAND)
for RNAi treatment of hyperlipidemia, Sci. Adv., 2022,
8(7), eabm1418.
186 M. R. Mohammadabadi, M. El-Tamimy, R. Gianello and
M. R. Mozafari, Supramolecular assemblies of zwitterionic
nanoliposome-polynucleotide complexes as gene transfer
vectors: Nanolipoplex formulation and in vitro character-
isation, J. Liposome Res., 2009, 19(2), 105115.
187 L. Moreira, N. M. Guimarães, S. Pereira, R. S. Santos,
J. A. Loureiro, M. C. Pereira, et al., Liposome Delivery of
Nucleic Acids in Bacteria: Toward In Vivo Labeling of
Human Microbiota, ACS Infect. Dis., 2022, 8(7), 12181230.
188 H. K. Dhaliwal, Y. Fan, J. Kim and M. M. Amiji, Intranasal
Delivery and Transfection of mRNA Therapeutics in the
Brain Using Cationic Liposomes, Mol. Pharmaceutics,
2020, 17(6), 19962005.
189 S. Lei, X. Zhang, K. Men, Y. Gao, X. Yang, S. Wu, et al.,
Ecient Colorectal Cancer Gene Therapy with
IL-15 mRNA Nanoformulation, Mol. Pharmaceutics, 2020,
17(9), 33783391.
190 M. M. Billingsley, A. G. Hamilton, D. Mai, S. K. Patel,
K. L. Swingle, N. C. Sheppard, et al., Orthogonal Design of
Experiments for Optimization of Lipid Nanoparticles for
mRNA Engineering of CAR T Cells, Nano Lett., 2022, 22(1),
533542.
191 A. J. Sinegra, M. Evangelopoulos, J. Park, Z. Huang and
C. A. Mirkin, Lipid Nanoparticle Spherical Nucleic Acids
for Intracellular DNA and RNA Delivery, Nano Lett., 2021,
21(15), 65846591.
192 A. M. Aldayel, H. L. OMary, S. A. Valdes, X. Li,
S. G. Thakkar, B. E. Mustafa, et al., Lipid nanoparticles
with minimum burst release of TNF-αsiRNA show strong
activity against rheumatoid arthritis unresponsive to
methotrexate, J. Controlled Release, 2018, 283, 280289.
193 Y. Han, P. Zhang, Y. Chen, J. Sun and F. Kong, Co-delivery
of plasmid DNA and doxorubicin by solid lipid nano-
particles for lung cancer therapy, Int. J. Mol. Med., 2014,
34(1), 191196.
194 C. Song, S. Zhang, Q. Zhou, L. Shi, L. Du, D. Zhi, et al.,
Bifunctional cationic solid lipid nanoparticles of β-NaYF4:
Yb,Er upconversion nanoparticles coated with a lipid for
bioimaging and gene delivery, RSC Adv., 2017, 7(43),
2663326639.
195 K. Paunovska, D. Loughrey and J. E. Dahlman, Drug deliv-
ery systems for RNA therapeutics, Nat. Rev. Genet., 2022,
23(5), 265280.
196 A. G. Niculescu, A. C. Bîrcăand A. M. Grumezescu, New
Applications of Lipid and Polymer-Based Nanoparticles
RSC Pharmaceutics Review
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 671
for Nucleic Acids Delivery, Pharmaceutics, 2021, 13(12),
2053.
197 C. Zeng, C. Zhang, P. G. Walker and Y. Dong, Formulation
and Delivery Technologies for mRNA Vaccines, Curr. Top.
Microbiol. Immunol., 2022, 440,71110.
198 Q. Cheng, T. Wei, L. Farbiak, L. T. Johnson, S. A. Dilliard
and D. J. Siegwart, Selective organ targeting (SORT) nano-
particles for tissue-specific mRNA delivery and CRISPR
Cas gene editing, Nat. Nanotechnol., 2020, 15(4), 313320.
199 J. Kim, A. Jozic and G. Sahay, Naturally Derived
Membrane Lipids Impact Nanoparticle-Based Messenger
RNA Delivery, Cell. Mol. Bioeng., 2020, 13(5), 463474.
200 X. Hou, T. Zaks, R. Langer and Y. Dong, Lipid nano-
particles for mRNA delivery, Nat. Rev. Mater., 2021, 6(12),
10781094.
201 C. Zhang, Y. Ma, J. Zhang, J. C. T. Kuo, Z. Zhang, H. Xie,
et al., Modification of Lipid-Based Nanoparticles: An
Ecient Delivery System for Nucleic Acid-Based
Immunotherapy, Molecules, 2022, 27(6), 1943.
202 M. A. Maier, M. Jayaraman, S. Matsuda, J. Liu, S. Barros,
W. Querbes, et al., Biodegradable lipids enabling rapidly
eliminated lipid nanoparticles for systemic delivery of
RNAi therapeutics, Mol. Ther., 2013, 21(8), 15701578.
203 A. Spadea, M. Jackman, L. Cui, S. Pereira, M. J. Lawrence,
R. A. Campbell, et al., Nucleic Acid-Loaded Lipid
Nanoparticle Interactions with Model Endosomal
Membranes, ACS Appl. Mater. Interfaces, 2022, 14(26),
3037130384.
204 P. L. Felgner, T. R. Gadek, M. Holm, R. Roman,
H. W. Chan, M. Wenz, et al., Lipofection: a highly
ecient, lipid-mediated DNA-transfection procedure,
Proc. Natl. Acad. Sci. U. S. A., 1987, 84(21), 74137417.
205 M. Schlich, R. Palomba, G. Costabile, S. Mizrahy,
M. Pannuzzo, D. Peer, et al., Cytosolic delivery of nucleic
acids: The case of ionizable lipid nanoparticles, Bioeng.
Transl. Med., 2021, e10213.
206 K. A. Mislick and J. D. Baldeschwieler, Evidence for the
role of proteoglycans in cation-mediated gene transfer,
Proc. Natl. Acad. Sci. U. S. A., 1996, 93(22), 1234912354.
207 H. C. Huang, P. Y. Chang, K. Chang, C. Y. Chen,
C. W. Lin, J. H. Chen, et al., Formulation of novel lipid-
coated magnetic nanoparticles as the probe for in vivo
imaging, J. Biomed. Sci., 2009, 16(1), 86.
208 M. Sun, U. J. Dang, Y. Yuan, A. M. Psaras, O. Osipitan,
T. A. Brooks, et al., Optimization of DOTAP/chol Cationic
Lipid Nanoparticles for mRNA, pDNA, and Oligonucleotide
Delivery, AAPS PharmSciTech, 2022, 23(5), 135.
209 Y. Zhang, C. Sun, C. Wang, K. E. Jankovic and Y. Dong,
Lipids and Lipid Derivatives for RNA Delivery, Chem. Rev.,
2021, 121(20), 1218112277.
210 D. Zhi, S. Zhang, S. Cui, Y. Zhao, Y. Wang and D. Zhao,
The headgroup evolution of cationic lipids for gene deliv-
ery, Bioconjugate Chem., 2013, 24(4), 487519.
211 F. Ponti, M. Campolungo, C. Melchiori, N. Bono and
G. Candiani, Cationic lipids for gene delivery: many
players, one goal, Chem. Phys. Lipids, 2021, 235, 105032.
212 S. Cui, Y. Wang, Y. Gong, X. Lin, Y. Zhao, D. Zhi, et al.,
Correlation of the cytotoxic eects of cationic lipids with
their headgroups, Toxicol. Res., 2018, 7(3), 473479.
213 S. J. H. Soenen, A. R. Brisson and M. De Cuyper,
Addressing the problem of cationic lipid-mediated tox-
icity: The magnetoliposome model, Biomaterials, 2009,
30(22), 36913701.
214 R. J. C. Bose, Y. Arai, J. C. Ahn, H. Park and S. H. Lee,
Influence of cationic lipid concentration on properties of
lipid-polymer hybrid nanospheres for gene delivery,
Int. J. Nanomed., 2015, 10, 53675382.
215 A. L. Bailey and P. R. Cullis, Modulation of membrane
fusion by asymmetric transbilayer distributions of amino
lipids, Biochemistry, 1994, 33(42), 1257312580.
216 L. Miao, J. Lin, Y. Huang, L. Li, D. Delcassian, Y. Ge, et al.,
Synergistic lipid compositions for albumin receptor
mediated delivery of mRNA to the liver, Nat. Commun.,
2020, 11(1), 2424.
217 M. M. Żak and L. Zangi, Lipid Nanoparticles for Organ-
Specific mRNA Therapeutic Delivery, Pharmaceutics, 2021,
13(10), 1675.
218 S. C. Semple, S. K. Klimuk, T. O. Harasym, N. Dos Santos,
S. M. Ansell, K. F. Wong, et al.,Ecient encapsulation of
antisense oligonucleotides in lipid vesicles using ioniz-
able aminolipids: formation of novel small multilamellar
vesicle structures, Biochim. Biophys. Acta, 2001, 1510(12),
152166.
219 M. Paloncýová, P. Čechová, M. Šrejber, P. Kühro and
M. Otyepka, Role of Ionizable Lipids in SARS-CoV-2
Vaccines As Revealed by Molecular Dynamics Simulations:
From Membrane Structure to Interaction with mRNA
Fragments, J. Phys. Chem. Lett., 2021, 12(45), 11199
11205.
220 M. Kim, M. Jeong, S. Hur, Y. Cho, J. Park, H. Jung, et al.,
Engineered ionizable lipid nanoparticles for targeted
delivery of RNA therapeutics into dierent types of cells in
the liver, Sci. Adv., 2021, 7(9), eabf4398.
221 C. L. Walsh, J. Nguyen, M. R. Tiany and F. C. Szoka,
Synthesis, characterization, and evaluation of ionizable
lysine-based lipids for siRNA delivery, Bioconjugate Chem.,
2013, 24(1), 3643.
222 X. Yu, S. Liu, Q. Cheng, T. Wei, S. Lee, D. Zhang, et al.,
Lipid-Modified Aminoglycosides for mRNA Delivery to the
Liver, Adv. Healthcare Mater., 2020, 9(7), 1901487.
223 T. Colombani, P. Peuziat, L. Dallet, T. Haudebourg,
M. Mével, M. Berchel, et al., Self-assembling complexes
between binary mixtures of lipids with dierent linkers
and nucleic acids promote universal mRNA, DNA and
siRNA delivery, J. Controlled Release, 2017, 249, 131142.
224 K. Bromma, K. Rieck, J. Kulkarni, C. OSullivan, W. Sung,
P. Cullis, et al., Use of a lipid nanoparticle system as a
Trojan horse in delivery of gold nanoparticles to human
breast cancer cells for improved outcomes in radiation
therapy, Cancer Nanotechnol., 2019, 10(1), 1.
225 A. Erfani, J. Seaberg, C. P. Aichele and J. D. Ramsey,
Interactions between Biomolecules and Zwitterionic
Review RSC Pharmaceutics
672 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
Moieties: A Review, Biomacromolecules, 2020, 21(7), 2557
2573.
226 D. Montizaan, K. Yang, C. Reker-Smit and A. Salvati,
Comparison of the uptake mechanisms of zwitterionic
and negatively charged liposomes by HeLa cells,
Nanomedicine, 2020, 30, 102300.
227 Y. Obata, S. Tajima and S. Takeoka, Evaluation of pH-
responsive liposomes containing amino acid-based
zwitterionic lipids for improving intracellular drug deliv-
ery in vitro and in vivo, J. Controlled Release, 2010, 142(2),
267276.
228 W. Wang, S. Jiang, S. Li, X. Yan, S. Liu, X. Mao, et al.,
Functional Choline Phosphate Lipids for Enhanced Drug
Delivery in Cancer Therapy, Chem. Mater., 2021, 33(2),
774781.
229 M. Zhang, A. Hussain, H. Yang, J. Zhang, X. J. Liang and
Y. Huang, mRNA-based modalities for infectious disease
management, Nano Res., 2023, 16(1), 672691.
230 T. M. Allen and P. R. Cullis, Liposomal drug delivery
systems: from concept to clinical applications, Adv. Drug
Delivery Rev., 2013, 65(1), 3648.
231 A. Akbarzadeh, R. Rezaei-Sadabady, S. Davaran, S. W. Joo,
N. Zarghami, Y. Hanifehpour, et al., Liposome: classifi-
cation, preparation, and applications, Nanoscale Res. Lett.,
2013, 8(1), 102.
232 M. N. Vu, P. Rajasekhar, D. P. Poole, S. Y. Khor,
N. P. Truong, C. J. Nowell, et al., Rapid assessment of
nanoparticle extravasation in a microfluidic tumor model,
ACS Appl. Nano Mater., 2019, 2(4), 18441856.
233 X. Gong, J. Wu, J. Wen, X. Ding, N. Xu, M. Sun, et al.,
Dual-Ligand-Modified Nanoscale Liposomes Loaded with
Curcumin and Metformin Inhibit Drug Resistance and
Metastasis of Hepatocellular Carcinoma, ACS Appl. Nano
Mater., 2022, 5(5), 70637077.
234 T. Michel, D. Luft, M. K. Abraham, S. Reinhardt,
M. L. Salinas Medina, J. Kurz, et al., Cationic Nanoliposomes
Meet mRNA: Ecient Delivery of Modified mRNA Using
Hemocompatible and Stable Vectors for Therapeutic
Applications, Mol. Ther.Nucleic Acids, 2017, 8,459468.
235 H. Wood, FDA approves patisiran to treat hereditary trans-
thyretin amyloidosis, Nat. Rev. Neurol., 2018, 14(10), 570
570.
236 H. H. Ly, S. Daniel, S. K. V. Soriano, Z. Kis and
A. K. Blakney, Optimization of Lipid Nanoparticles for
saRNA Expression and Cellular Activation Using a Design-
of-Experiment Approach, Mol. Pharmaceutics, 2022, 19(6),
18921905.
237 A. R. Ferhan, S. Park, H. Park, H. Tae, J. A. Jackman and
N. J. Cho, Lipid Nanoparticle Technologies for Nucleic
Acid Delivery: A Nanoarchitectonics Perspective, Adv.
Funct. Mater., 2022, 2203669.
238 B. L. Mui, Y. K. Tam, M. Jayaraman, S. M. Ansell, X. Du,
Y. Y. C. Tam, et al., Influence of Polyethylene Glycol
Lipid Desorption Rates on Pharmacokinetics and
Pharmacodynamics of siRNA Lipid Nanoparticles, Mol.
Ther.Nucleic Acids, 2013, 2, e139.
239 Y. Duan, A. Dhar, C. Patel, M. Khimani, S. Neogi,
P. Sharma, et al., A brief review on solid lipid nano-
particles: part and parcel of contemporary drug delivery
systems, RSC Adv., 2020, 10(45), 2677726791.
240 S. Scioli Montoto, G. Muraca and M. E. Ruiz, Solid Lipid
Nanoparticles for Drug Delivery: Pharmacological and
Biopharmaceutical Aspects, Front. Mol. Biosci., 2020, 7,
DOI: 10.3389/fmolb.2020.587997.
241 P. Ghasemiyeh and S. Mohammadi-Samani, Solid lipid
nanoparticles and nanostructured lipid carriers as
novel drug delivery systems: applications, advantages
and disadvantages, Res. Pharm. Sci., 2018, 13(4), 288
303.
242 R. K. Shirodkar, L. Kumar, S. Mutalik and S. Lewis, Solid
Lipid Nanoparticles and Nanostructured Lipid Carriers:
Emerging Lipid Based Drug Delivery Systems, Pharm.
Chem. J., 2019, 53(5), 440453.
243 R. Tenchov, R. Bird, A. E. Curtze and Q. Zhou, Lipid
Nanoparticles-From Liposomes to mRNA Vaccine Delivery,
a Landscape of Research Diversity and Advancement, ACS
Nano, 2021, 15(11), 1698217015.
244 S. Beg, A. K. Malik, M. J. Ansari, A. A. Malik, A. M. A. Ali,
A. Theyab, et al., Systematic Development of Solid Lipid
Nanoparticles of Abiraterone Acetate with Improved Oral
Bioavailability and Anticancer Activity for Prostate
Carcinoma Treatment, ACS Omega, 2022, 7(20), 16968
16979.
245 M. Kotmakçı, H. Akbaba, G. Erel, G. Ertan and
G. Kantarcı, Improved Method for Solid Lipid
Nanoparticle Preparation Based on Hot Microemulsions:
Preparation, Characterization, Cytotoxicity, and
Hemocompatibility Evaluation, AAPS PharmSciTech, 2017,
18(4), 13551365.
246 E. B. Souto, S. Doktorovova, A. Zielinska and A. M. Silva,
Key production parameters for the development of solid
lipid nanoparticles by high shear homogenization, Pharm.
Dev. Technol., 2019, 24(9), 11811185.
247 H. B. Abo-zalam, E. S. El-Denshary, R. M. Abdelsalam,
I. A. Khalil, M. M. Khattab and M. A. Hamzawy,
Therapeutic advancement of simvastatin-loaded solid
lipid nanoparticles (SV-SLNs) in treatment of hyperlipide-
mia and attenuating hepatotoxicity, myopathy and apop-
tosis: Comprehensive study, Biomed. Pharmacother., 2021,
139, 111494.
248 V. A. Duong, T. T. L. Nguyen and H. J. Maeng, Preparation
of Solid Lipid Nanoparticles and Nanostructured Lipid
Carriers for Drug Delivery and the Eects of Preparation
Parameters of Solvent Injection Method, Molecules, 2020,
25(20), E4781.
249 A. Penumarthi, D. Parashar, A. N. Abraham, C. Dekiwadia,
I. Macreadie, R. Shukla, et al., Solid lipid nanoparticles
mediate non-viral delivery of plasmid DNA to dendritic
cells, J. Nanopart. Res., 2017, 19(6), 210.
250 Y. C. Barenholz, Doxil®The first FDA-approved nano-
drug: Lessons learned, J. Controlled Release, 2012, 160(2),
117134.
RSC Pharmaceutics Review
© 2024 The Author(s). Published by the Royal Society of Chemistry RSC Pharm.,2024,1,645674 | 673
251 A. C. Anselmo and S. Mitragotri, Nanoparticles in the
clinic: An update, Bioeng. Transl. Med., 2019, 4(3), e10143.
252 Commissioner O of the. Moderna COVID-19 Vaccines,
FDA, 2023, Available from: https://www.fda.gov/emer-
gency-preparedness-and-response/coronavirus-disease-
2019-covid-19/moderna-covid-19-vaccines.
253 T. T. H. Thi, E. J. A. Suys, J. S. Lee, D. H. Nguyen,
K. D. Park and N. P. Truong, Lipid-Based Nanoparticles in
the Clinic and Clinical Trials: From Cancer
Nanomedicine to COVID-19 Vaccines, Vaccines, 2021, 9(4),
359.
254 A. J. Debacker, J. Voutila, M. Catley, D. Blakey and
N. Habib, Delivery of Oligonucleotides to the Liver with
GalNAc: From Research to Registered Therapeutic Drug,
Mol. Ther., 2020, 28(8), 17591771.
255 A. I. S. van den Berg, C. O. Yun, R. M. Schielers and
W. E. Hennink, Polymeric delivery systems for nucleic
acid therapeutics: Approaching the clinic, J. Controlled
Release, 2021, 331, 121141.
256 Y. Guo, X. Cao, X. Zheng, S. J. Abbas, J. Li and W. Tan,
Construction of nanocarriers based on nucleic acids and
their applications in nanobiology delivery systems, Natl.
Sci. Rev., 2022, 9(5), nwac006.
257 H. Sabit, M. Abdel-Hakeem, T. Shoala, S. Abdel-Ghany,
M. M. Abdel-Latif, J. Almulhim, et al., Nanocarriers: A
Reliable Tool for the Delivery of Anticancer Drugs,
Pharmaceutics, 2022, 14(8), 1566.
258 S. Khatun, C. L. Putta, A. Hak and A. K. Rengan,
Immunomodulatory nanosystems: An emerging strategy
to combat viral infections, Biomater. Biosyst., 2023, 9,
100073.
259 S. Su and P. M. Kang, Systemic Review of Biodegradable
Nanomaterials in Nanomedicine, Nanomaterials, 2020,
10(4), 656.
260 J. M. Benns, J. S. Choi, R. I. Mahato, J. S. Park and
S. W. Kim, pH-Sensitive Cationic Polymer Gene Delivery
Vehicle: N-Ac-poly(l-histidine)-graft-poly(l-lysine) Comb
Shaped Polymer, Bioconjugate Chem., 2000, 11(5), 637
645.
261 Y. Huang, X. Guo, Y. Wu, X. Chen, L. Feng, N. Xie, et al.,
Nanotechnologys frontier in combatting infectious and
inflammatory diseases: prevention and treatment, Signal
Transduction Targeted Ther., 2024, 9(1), 150.
262 L. Sun, H. Liu, Y. Ye, Y. Lei, R. Islam, S. Tan, et al., Smart
nanoparticles for cancer therapy, Signal Transduction
Targeted Ther., 2023, 8, 418.
263 M. Chehelgerdi, M. Chehelgerdi, O. Q. B. Allela,
R. D. C. Pecho, N. Jayasankar, D. P. Rao, et al., Progressing
nanotechnology to improve targeted cancer treatment:
overcoming hurdles in its clinical implementation, Mol.
Cancer, 2023, 22, 169.
264 Z. Hejdankova, V. Vanek, F. Sedlak, J. Prochazka,
A. Diederichs, S. Kereïche, et al., Lipid Nanoparticles for
Broad-Spectrum Nucleic Acid Delivery, Adv. Funct. Mater.,
2021, 31(47), 2101391.
265 E. Samaridou, J. Heyes and P. Lutwyche, Lipid nano-
particles for nucleic acid delivery: Current perspectives,
Adv. Drug Delivery Rev., 2020, 154155,3763.
Review RSC Pharmaceutics
674 |RSC Pharm.,2024,1,645674 © 2024 The Author(s). Published by the Royal Society of Chemistry
... Metallic-polymeric hybrids, embedding magnetic or gold NPs in polymer matrices, enable simultaneous imaging and therapy. These systems have demonstrated superior performance in both diagnostics and treatment, offering new opportunities for theranostic applications [48]. ...
Article
Full-text available
This review aims to address the significant challenges of treating central nervous system (CNS) disorders such as neurodegenerative diseases, strokes, spinal cord injuries, and brain tumors. These disorders are difficult to manage due to the complexity of disease mechanisms and the protective blood–brain barrier (BBB), which restricts drug delivery. Recent advancements in nanoparticle (NP) technologies offer promising solutions, with potential applications in drug delivery, neuroprotection, and neuroregeneration. By examining current research, we explore how NPs can cross the BBB, deliver medications directly to targeted CNS regions, and enhance both diagnostics and treatment. Key NP strategies, such as passive targeting, receptor-mediated transport, and stimuli-responsive systems, demonstrate encouraging results. Studies show that NPs may improve drug delivery, minimize side effects, and increase therapeutic effectiveness in models of Alzheimer’s, Parkinson’s, stroke, and glioblastoma. NP technologies thus represent a promising approach for CNS disorder management, combining drug delivery and diagnostic capabilities to enable more precise and effective treatments that could significantly benefit patient outcomes.
Article
Full-text available
Inflammation-associated diseases encompass a range of infectious diseases and non-infectious inflammatory diseases, which continuously pose one of the most serious threats to human health, attributed to factors such as the emergence of new pathogens, increasing drug resistance, changes in living environments and lifestyles, and the aging population. Despite rapid advancements in mechanistic research and drug development for these diseases, current treatments often have limited efficacy and notable side effects, necessitating the development of more effective and targeted anti-inflammatory therapies. In recent years, the rapid development of nanotechnology has provided crucial technological support for the prevention, treatment, and detection of inflammation-associated diseases. Various types of nanoparticles (NPs) play significant roles, serving as vaccine vehicles to enhance immunogenicity and as drug carriers to improve targeting and bioavailability. NPs can also directly combat pathogens and inflammation. In addition, nanotechnology has facilitated the development of biosensors for pathogen detection and imaging techniques for inflammatory diseases. This review categorizes and characterizes different types of NPs, summarizes their applications in the prevention, treatment, and detection of infectious and inflammatory diseases. It also discusses the challenges associated with clinical translation in this field and explores the latest developments and prospects. In conclusion, nanotechnology opens up new possibilities for the comprehensive management of infectious and inflammatory diseases.
Article
Full-text available
Combining existing drug therapy is essential in developing new therapeutic agents in disease prevention and treatment. In preclinical investigations, combined effect of certain known drugs has been well established in treating extensive human diseases. Attributed to synergistic effects by targeting various disease pathways and advantages, such as reduced administration dose, decreased toxicity, and alleviated drug resistance, combinatorial treatment is now being pursued by delivering therapeutic agents to combat major clinical illnesses, such as cancer, atherosclerosis, pulmonary hypertension, myocarditis, rheumatoid arthritis, inflammatory bowel disease, metabolic disorders and neurodegenerative diseases. Combinatorial therapy involves combining or co-delivering two or more drugs for treating a specific disease. Nanoparticle (NP)-mediated drug delivery systems, i.e., liposomal NPs, polymeric NPs and nanocrystals, are of great interest in combinatorial therapy for a wide range of disorders due to targeted drug delivery, extended drug release, and higher drug stability to avoid rapid clearance at infected areas. This review summarizes various targets of diseases, preclinical or clinically approved drug combinations and the development of multifunctional NPs for combining therapy and emphasizes combinatorial therapeutic strategies based on drug delivery for treating severe clinical diseases. Ultimately, we discuss the challenging of developing NP-codelivery and translation and provide potential approaches to address the limitations. This review offers a comprehensive overview for recent cutting-edge and challenging in developing NP-mediated combination therapy for human diseases.
Article
Full-text available
The increasing number of approved nucleic acid therapeutics demonstrates the potential for the prevention and treatment of a broad spectrum of diseases. This trend underscores the significant impact and promise of nucleic acid-based treatments in the field of medicine. Nevertheless, employing nucleic acids as therapeutics is challenging due to their susceptibility to degradation by nucleases and their unfavorable physicochemical characteristics that hinder delivery into cells. Appropriate vectors play a pivotal role in improving nucleic acid stability and delivering nucleic acids into specific cells. The maturation of delivery systems has led to breakthroughs in the development of therapeutics based on nucleic acids such as DNA, siRNA, and mRNA. Non-viral vectors have gained prominence among the myriad of nanomaterials due to low immunogenicity, ease of manufacturing, and simplicity of cost-effective, large-scale production. Here, we provide an overview of the recent advancements in nanomaterials for nucleic acid delivery. Specifically, we give a detailed introduction to the characteristics of polymers, lipids, and polymer-lipid hybrids, and provide comprehensive descriptions of their applications in nucleic acid delivery. Also, biological barriers, administration routes, and strategies for organ-selective delivery of nucleic acids are discussed. In summary, this review offers insights into the rational design of next-generation delivery vectors for nucleic acid delivery.
Article
Full-text available
Smart nanoparticles, which can respond to biological cues or be guided by them, are emerging as a promising drug delivery platform for precise cancer treatment. The field of oncology, nanotechnology, and biomedicine has witnessed rapid progress, leading to innovative developments in smart nanoparticles for safer and more effective cancer therapy. In this review, we will highlight recent advancements in smart nanoparticles, including polymeric nanoparticles, dendrimers, micelles, liposomes, protein nanoparticles, cell membrane nanoparticles, mesoporous silica nanoparticles, gold nanoparticles, iron oxide nanoparticles, quantum dots, carbon nanotubes, black phosphorus, MOF nanoparticles, and others. We will focus on their classification, structures, synthesis, and intelligent features. These smart nanoparticles possess the ability to respond to various external and internal stimuli, such as enzymes, pH, temperature, optics, and magnetism, making them intelligent systems. Additionally, this review will explore the latest studies on tumor targeting by functionalizing the surfaces of smart nanoparticles with tumor-specific ligands like antibodies, peptides, transferrin, and folic acid. We will also summarize different types of drug delivery options, including small molecules, peptides, proteins, nucleic acids, and even living cells, for their potential use in cancer therapy. While the potential of smart nanoparticles is promising, we will also acknowledge the challenges and clinical prospects associated with their use. Finally, we will propose a blueprint that involves the use of artificial intelligence-powered nanoparticles in cancer treatment applications. By harnessing the potential of smart nanoparticles, this review aims to usher in a new era of precise and personalized cancer therapy, providing patients with individualized treatment options.
Article
Full-text available
The use of nanotechnology has the potential to revolutionize the detection and treatment of cancer. Developments in protein engineering and materials science have led to the emergence of new nanoscale targeting techniques, which offer renewed hope for cancer patients. While several nanocarriers for medicinal purposes have been approved for human trials, only a few have been authorized for clinical use in targeting cancer cells. In this review, we analyze some of the authorized formulations and discuss the challenges of translating findings from the lab to the clinic. This study highlights the various nanocarriers and compounds that can be used for selective tumor targeting and the inherent difficulties in cancer therapy. Nanotechnology provides a promising platform for improving cancer detection and treatment in the future, but further research is needed to overcome the current limitations in clinical translation. Graphical Abstract
Article
Full-text available
Nanofibers have a large area of surface variable 3D topography, porosity, and adaptable surface functions. Several researchers are researching nanofiber technology as a potential solution to the current problems in several fields. It manages cardiovascular disorders, infectious diseases, gastrointestinal tract-associated diseases, neurodegenerative diseases, pain treatment, contraception, and wound healing. The nanofibers are fabricated using various fabrication techniques, such as electrospinning, phase separation, physical Fabrication, and chemical fabrication. Depending on their intended use, nanofibers are manufactured using a variety of polymers. It comprises natural polymers, semi-synthetic polymers, synthetic polymers, metals, metal oxides, ceramics, carbon, nonporous materials, mesoporous materials, hollow structures, core-shell structures, biocomponents, and multi-component materials. Nanofiber composites are a good alternative for targeted gene delivery, protein and peptide delivery, and growth factor delivery. Thus, nanofibers have huge potential in drug delivery, which enables them to be used for various applications and can revolutionize these therapeutic areas. This review systematically studied nanofibers' history, advantages, disadvantages, types, and polymers used in nanofiber technology. Further, polymers and their types used in the preparation of nanofibers were summarised. Mainly review article focuses on the fabrication method, i.e., electrospinning and its types. Finally, the article discussed the applications and recent advancements of nanofabrication technology.
Article
Full-text available
A wide variety of therapeutic approaches and technologies for delivering therapeutic agents have been investigated for treating cancer. Recently, immunotherapy has achieved success in cancer treatment. Successful clinical results of immunotherapeutic approaches for cancer treatment were led by antibodies targeting immune checkpoints, and many have advanced through clinical trials and obtained FDA approval. A major opportunity remains for the development of nucleic acid technology for cancer immunotherapy in the form of cancer vaccines, adoptive T-cell therapies, and gene regulation. However, these therapeutic approaches face many challenges related to their delivery to target cells, including their in vivo decay, the limited uptake by target cells, the requirements for nuclear penetration (in some cases), and the damage caused to healthy cells. These barriers can be avoided and resolved by utilizing advanced smart nanocarriers (e.g., lipids, polymers, spherical nucleic acids, metallic nanoparticles) that enable the efficient and selective delivery of nucleic acids to the target cells and/or tissues. Here, we review studies that have developed nanoparticle-mediated cancer immunotherapy as a technology for cancer patients. Moreover, we also investigate the crosstalk between the function of nucleic acid therapeutics in cancer immunotherapy, and we discuss how nanoparticles can be functionalized and designed to target the delivery and thus improve the efficacy, toxicity, and stability of these therapeutics.
Article
Full-text available
This study aims to design and optimize chitosan-coated bilosomal formulations loaded with psoralidin (Ps-CS/BLs) with improved physicochemical properties, oral bioavailability, and boosted apoptotic and necrotic effects. In this regard, uncoated bilosomes loaded with Ps (Ps/BLs) were nanoformulated using the thin-film hydration technique using different molar ratios of phos-phatidylcholine (PC), cholesterol (Ch), Span 60 (S60), and sodium deoxycholate (SDC) (1:0.4:0.2:0.125, 1:0.4:0.2:0.25, and 1:0.4:0.2:0.5, respectively). The best-optimized formulation with respect to size, PDI, zeta potential, and EE% was selected and then coated with chitosan at two different concentrations (0.125 and 0.25 w/v%), forming Ps-CS/BLs. The optimized Ps/BLs and Ps-CS/BLs showed a spherical shape and relatively homogenous size with negligible apparent agglomerations. Additionally , it was demonstrated that coating Ps/BLs with chitosan has significantly increased the particle size from 123.16 ± 6.90 in the case of Ps/BLs to 183.90 ± 15.93 nm in the case of Ps-CS/BLs. In addition, Ps-CS/BLs exhibited higher zeta potential (+30.78 ± 1.44 mV) as compared to Ps/BLs (−18.59 ± 2.13 mV). Furthermore, Ps-CS/BL showed enhanced entrapment efficiency (EE%) of 92.15 ± 7.20% as compared to Ps/BLs (68.90 ± 5.95%). Moreover, Ps-CS/BLs exhibited a more sustained release behavior of Ps compared to Ps/BLs over 48 h, and both formulations were best obeying the Higuchi diffusion model. More importantly, Ps-CS/BLs displayed the highest mucoad-hesive efficiency% (74.89 ± 3.5%) as compared to Ps/BLs (26.78 ± 2.9%), indicating the ability of the designed nanoformulation to improve oral bioavailability and extend the residence time inside the gastrointestinal tract upon oral administration. Moreover, upon evaluating the apoptotic and necrotic effects of free Ps and Ps-CS/BLs on human breast cancer cell lines (MCF-7) and human lung adenocarcinoma cell lines (A549), there was a dramatic increase in the percentages of the apoptotic and necrotic cell compared to the control and free Ps. Our findings suggest the possible oral use of Ps-CS/BLs in hampering breast and lung cancers.
Article
Full-text available
The viral infection spreads with the assistance of a host. Traditional antiviral therapies cannot provide long-term immunity against emerging and drug-resistant viral infections. Immunotherapy has evolved as an efficient approach for disease prevention and treatment, which include cancer, infections, inflammatory, and immune disorders. Immunomodulatory nanosystems can dramatically enhance therapeutic outcomes by combating many therapeutic challenges, such as poor immune stimulation and off-target adverse effects. Recently, immunomodulatory nanosystems have emerged as a potent antiviral strategy to intercept viral infections effectively. This review introduces major viral infections with their primary symptoms, route of transmission & targeted organ, and different stages of the viral life cycle with respective traditional blockers. The IMNs have an exceptional capacity for precisely modulating the immune system for therapeutic applications. The nano sized immunomodulatory systems permit the immune cells to interact with infectious agents enhancing lymphatic drainage and endocytosis by the over-reactive immune cells in the infected areas. Immune cells that can be modulated upon viral infection via various immunomodulatory nanosystems have been discussed. Advancement in theranostics can yield an accurate diagnosis, adequate treatment, and real-time screening of viral infections. Nanosystem-based drug delivery can continue to thrive in diagnosing, treating, and preventing viral infections. The curative medicine for remerging and drug-resistant viruses remains challenging, though certain systems have expanded our perception and initiated a new research domain in antiviral treatments.
Article
Nucleic acid‐based therapies are seeing a spiralling surge. Stimuli‐responsive polymers, especially pH‐responsive ones, are gaining widespread attention because of their ability to efficiently deliver nucleic acids. These polymers can be synthesized and modified according to target requirements, such as delivery sites and the nature of nucleic acids. In this regard, the endosomal escape mechanism of polymer–nucleic acid complexes (polyplexes) remains a topic of considerable interest owing to various plausible escape mechanisms. This review describes current progress in the endosomal escape mechanism of polyplexes and state‐of‐the‐art chemical designs for pH‐responsive polymers. We also discuss the importance of the acid dissociation constant (i.e., p K a ) in designing the new generation of pH‐responsive polymers, along with assays to monitor and quantify the endosomal escape behavior. Further, we address the use of machine learning in p K a prediction and polymer design to find novel chemical structures for pH responsiveness. This review will facilitate the design of new pH‐responsive polymers for advanced and efficient nucleic acid delivery. This article is protected by copyright. All rights reserved