You are on page 1of 19

Chemistry and Physics of Lipids 226 (2020) 104837

Contents lists available at ScienceDirect

Chemistry and Physics of Lipids


journal homepage: www.elsevier.com/locate/chemphyslip

Review

Lipid nanocarriers for microRNA delivery T


a,b,c d d,
Marcel Scheideler , Ivan Vidakovic , Ruth Prassl *
a
Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
b
Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
c
German Center for Diabetes Research (DZD), Neuherberg, Germany
d
Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Biophysics, Medical University of Graz, Neue Stiftingtalstraße 6/IV, 8010 Graz, Austria

A R T I C LE I N FO A B S T R A C T

Keywords: Non-coding RNAs (ncRNAs) like microRNAs (miRNAs) or small interference RNAs (siRNAs) with their power to
Liposomes selectively silence any gene of interest enable the targeting of so far ‘undruggable’ proteins and diseases. Such
Lipoplexes RNA molecules have gained much attention from biotech and pharmaceutical companies, which led to the first
Lipid nanoparticles Food and Drug Administration (FDA) approved ncRNA therapeutic in 2018.
Oligonucleotides
However, the main barrier in clinical practice of ncRNAs is the lack of an effective delivery system that can
protect the RNA molecules from nuclease degradation, deliver them to specific tissues and cell types, and release
them into the cytoplasm of the targeted cells, all without inducing adverse effects.
For that reason, drug delivery approaches, formulations, technologies and systems for transporting phar-
macological ncRNA compounds to achieve a diagnostic or therapeutic effect in the human body are in demand.
Here, we review the development of therapeutic lipid-based nanoparticles for delivery of miRNAs, one class
of endogenous ncRNAs with specific regulatory functions. We outline challenges and opportunities for advanced
miRNA-based therapies, and discuss the complexity associated with the delivery of functional miRNAs. Novel
strategies are addressed how to deal with the most critical points in miRNA delivery, such as toxicity, specific
targeting of disease sites, proper cellular uptake and endosomal escape of miRNAs. Current fields of application
and various preclinical settings involving miRNA therapeutics are discussed, providing an outlook to future
clinical approaches.
Following the current trends and technological developments in nanomedicine exciting new delivery systems
for ncRNA-based therapeutics can be expected in upcoming years.

1. Introduction these molecules highly interesting candidates either as therapeutics or


as therapeutic targets. Given the diversity of ncRNA molecules, miRNA-
MicroRNAs (miRNAs) are the most studied class of non-coding RNA based therapeutics are applied either as synthetic double stranded small
molecules (ncRNAs) that play a central role as regulators of gene ex- RNA molecules, so called miRNA mimics, or as miRNA inhibitors/an-
pression. MiRNAs are single-stranded RNAs with 19–25 nucleotides in tagonists. The function of miRNA mimics is to restore or enhance
length that can diminish protein output. Approximately 2000 miRNAs miRNA abundance and function to be used in miRNA replacement
exist in humans, and it is now estimated that miRNAs regulate about therapy (Hosseinahli et al., 2018; Rothschild, 2014; Wang et al., 2016).
60% of all protein-coding genes (Friedman et al., 2009; Selbach et al., In contrast to miRNA mimics, miRNA inhibitors are single-stranded
2008). Hence, miRNAs are deeply involved in different cellular pro- antisense oligonucleotides, which have a complementary sequence to
cesses such as differentiation, proliferation, apoptosis or autophagy. A the miRNA thus preventing miRNA binding to its endogenous targets
dysregulation of miRNAs can cause numerous pathological disorders (Rupaimoole and Slack, 2017).
like cancer, atherosclerosis or cardiovascular diseases (Feinberg and MiRNAs in therapeutic applications typically require a delivery
Moore, 2016; Iorio and Croce, 2012; Rupaimoole et al., 2016a; system to improve their efficiency in vivo and to enhance their ther-
Rupaimoole and Slack, 2017). Especially, the capacity of miRNAs to apeutic index. Thus, miRNAs are encapsulated into various nano-
target multiple mRNAs that are altered in disease conditions makes carriers, which have the ability to provide a higher molecular stability


Corresponding author.
E-mail addresses: marcel.scheideler@helmholtz-muenchen.de (M. Scheideler), ivan.vidakovic@medunigraz.at (I. Vidakovic),
ruth.prassl@medunigraz.at (R. Prassl).

https://doi.org/10.1016/j.chemphyslip.2019.104837
Received 24 July 2019; Received in revised form 12 October 2019; Accepted 15 October 2019
Available online 02 November 2019
0009-3084/ © 2019 Elsevier B.V. All rights reserved.
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

and efficient protection from degradation by nucleases abundantly targeting moieties are important assets to improve particle stability,
present in serum (Chen et al., 2015). In principle, the nanoparticles circulation lifetime and tissue specificity. Particularly for miRNA
have to be specifically designed to permit an effective transport of high therapy the biggest challenges to be met are acceptable toxicity, tissue
concentrations of active miRNAs to the target organs and enable cel- specific targeting and proper cellular uptake as well as cytosolic release
lular entry by endocytosis followed by endosomal escape and cytosolic of the miRNA cargo.
release. Among non-viral delivery systems lipid-based nanoparticles are In this review, we focus on the design and applicability of lipid-
the most intensively investigated carrier systems (Cullis and Hope, based miRNA delivery systems rather than on specific miRNAs to be
2017; Kulkarni et al., 2018). transported, as we expect the delivery vehicles to be similarly effective
Some of the lipid-based formulations that are currently developed irrespective of which miRNA molecule is encapsulated. Furthermore, it
for the delivery of DNA/RNA or ncRNAs are already in clinical practice, can be expected that the physicochemical characteristics of the nano-
predominantly for cancer therapy (Campani et al., 2016). A great ad- particle will not significantly change using different sequences of nu-
vantage of lipid nanocarriers is the biodegradability and biocompat- cleic acids. Moreover, all systemically administered oligonucleotides
ibility of their individual components. Likewise, most lipid nano- (ONDs) face similar physiological hurdles that might impede their
particles show tolerable toxicity and low immunogenicity. The therapeutic applicability. In circulation, they have to escape enzymatic
nanoparticles are assembled from various lipids and vary in their degradation and rapid clearance by the immune system while they have
physicochemical characteristics attributable to differences in chemical to reach their target cells, accomplish cellular uptake to be released in
composition, size variability and particle surface properties. All these the cytosol (Kanasty et al., 2013; Tibbitt et al., 2016; Whitehead et al.,
parameters can be easily tuned and adjusted. Most commonly, the lipid 2009). Hence, the delivery systems appear to be adequate for both
nanoparticles are coated with polymers offering the opportunity to in- miRNA and siRNA molecules as well, making nanoparticles a universal
crease particle stability in plasma leading to prolonged half-life time in delivery vehicle for OND based drugs. Rather, we proceed from the
circulation (Barenholz, 2001). Another attractive approach to improve assumption that the cellular species and the target organ to be ad-
drug delivery efficacy is to conjugate ligands or targeting sequences dressed determine which kind of nanoparticle assembly is most ap-
onto the surface of the nanoparticles to achieve a more specific re- propriate. In this respect, liposomes, lipid nanoparticles and lipid-based
cognition at disease sites or target organs. Such ligands include peptide assemblies serve as flexible nanoparticle platform for the delivery of
sequences, proteins, specific antibodies or aptamers (Belfiore et al., drugs including nucleic acid derived therapeutics.
2018; Torchilin, 2005). Ligand conjugated liposomes enable an active
targeting of cell receptors or specifically surface exposed biomarkers. 2. Preparation, loading, functionalization and upscaling aspects
Alternatively, passive accumulation of nanoparticles in tumor tissues for lipid-based nanoparticles in ncRNA delivery
can be attained by the so called enhanced permeability and retention
(EPR) effect. Owing to the overall leaky structure of the tumor vascu- Lipid based nanoparticle technology for ncRNA delivery has to
lature, which is induced by gaps of varying size between the endothelial follow stringent criteria for robust and reliable production processes,
cells and widespread fenestrations of blood vessels, an enhanced ex- which have to be scalable and cost efficient. High encapsulation effi-
travasation of nanoparticles in the tumor microenvironment is gained ciencies and long-term stabilities are required. Other challenges in the
(Miao and Huang, 2015). The longer retention time for nanoparticles in development of ncRNA formulations are (1) to preserve the functional
tumor tissues is attributed to the lack of efficient lymphatic drainage activity of the ncRNA payload, (2) to protect the drug from degrada-
(Bazak et al., 2014). tion, (3) to confer high stability to the ncRNA molecules, (4) to enable
Particle size is another key factor for determining the in vivo fate of tissue-specific targeting; to reduce cytotoxicity, and (5) to enable safe
the particle involving cellular uptake mechanisms, biodistribution delivery to the diseased site with reduced risk of off-target effects (Li
profiles and clearance rates. Along these lines, therapeutic nano- and Rana, 2014; Singh and Peer, 2016; Slaby et al., 2017).
particles for intravenous applications need to have an optimal size that Usually, lipid nanoparticles are easy to prepare and scale-up pro-
typically ranges between 30–200 nm. Bigger particles are mostly taken cedures for large-scale manufacturing are readily available. Established
up by the RES for phagocytotic clearance, while smaller nanoparticles preparation methods for lipid based nanoparticles were recently re-
with the size of only a few nanometers are rapidly cleared by renal viewed (Patil and Jadhav, 2014; Pattni et al., 2015). Among them the
excretion (Choi et al., 2007). Apart from nanoparticle size other phy- most prominent techniques are conventional thin lipid film rehydration
sicochemical properties like chemical nature of lipids, charge or particle techniques combined with size extrusion procedures, ethanol-lipid di-
shape are relevant features for delivery purposes. For example, charged lution techniques or more recently microfluidics-based rapid mixing
nanoparticles might be opsonized and removed more rapidly from procedures (Maeki et al., 2018). An exemplary scheme for the pre-
circulation than their neutral counterparts (Carrstensen et al., 1992). paration of miRNA loaded liposomes using microfluidic devices is
Yet, different clearance rates are reported for negatively and positively presented in Fig. 1. The relatively high amount of ethanol (up to 40%
charged liposomes depending on their propensity to interact non- by volume) during the rapid mixing process facilitates the encapsula-
specifically with oppositely charged plasma components and their in- tion of higher amounts of oligonucleotides. With this single step man-
trinsic tendency for particle aggregation (Deshpande et al., 2013; Miller ufacturing process large amounts of small lipid nanoparticles (dia-
et al., 1998). Once in circulation, nanoparticles adsorb plasma proteins meter < 100 nm) can be produced within a few minutes (Cullis and
to become surrounded by the so-called protein corona, which might Hope, 2017). Subsequently, the formulations have to be dialysed to
readily affect their biodistribution and pharmacokinetics (Bertrand remove excess of ethanol and unbound ncRNA molecules. Dialysis is
et al., 2017). Often the protein corona impairs targeting specificity and essential to stabilize the nanoparticles, to inhibit aggregation and to
cellular uptake. On the other hand, for some lipid-based delivery sys- adjust the pH to neutral for lowering the surface charge whenever io-
tems it is essential to interact with plasma components like lipoproteins nizable cationic lipids have been used at low pH. The formulation can
after their entry in the bloodstream to be directed to hepatocytes in the be sterilized by filtration and freeze-dried for storage in the presence of
liver for the active treatment of liver diseases (Akinc et al., 2010; a sugar lyoprotectant (Stark et al., 2008).
Wolfrum et al., 2007). This exemplifies the fact that nanocarriers have Notably, the behavior of the nanoparticles might be influenced by
to be flexibly adopted to meet individual treatment requirements. the manufacturing technique. For some cases, it was shown that the
Taken together, for the successful delivery of miRNA the carriers same substances formulated into nanoparticles by microfluidics per-
need to be carefully designed and optimized in terms of lipid compo- formed much better than those formulated by extrusion techniques
sition, particle size, charge distribution, miRNA to lipid ratio and en- (Chen et al., 2012; Valencia et al., 2012; Wan et al., 2014).
capsulation efficiency. Moreover, surface coatings and coupling of To confer steric stabilization to the lipid formulation with the goal

2
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

therapeutics are discussed later on in this review article.

3. Lipid platform for nucleic acid delivery with specific focus on


miRNA

At present, various lipid systems have been explored for gene


transfection or OND delivery. In the following, some of them will be
discussed in more detail, supported by representative examples of their
potential therapeutic applications with special focus on miRNA de-
livery. Since significantly more research has emerged about siRNA
using advanced lipid formulations some studies are important to be
mentioned when discussing delivery strategies. Table 1 presents a se-
lected list of lipid nanocarriers currently used for miRNA delivery. A list
of lipids appropriate for gene transfection in nanomedicine technology
is given in Table 2. This review does not take into account approaches
using commercially available lipid-based transfection reagents as such
reagents are primarily intended for in vitro cell culture experiments.
Although currently significant efforts are being made to develop new
transfection reagents that can also be used for in vivo gene delivery in
preclinical settings.
Fig. 1. Schematic overview of a microfluidic system for the manufacturing of
miRNA loaded liposomes. Lipids dissolved in ethanol are mixed with an aqu-
3.1. Cationic lipoplexes
eous solution of miRNA at a constant flow rate within a microfluidic chip de-
vice. The liposomes are formed by self-assembly. Excess ethanol and non-en-
Among lipid-based nanoparticles cationic liposomes, often termed
capsulated free drug are removed by dialysis.
lipoplexes, are the most prominent and widely explored delivery ve-
hicles for DNA (Felgner et al., 1987; Zuidam et al., 1999), mRNA (Islam
to enhance circulation lifetime and to reduce recognition by the im- et al., 2015), siRNA (Xia et al., 2016) or OND (Garbuzenko et al., 2009).
mune system, the nanoparticles are coated with a polymer shell. In For the development of lipoplexes a great variety of different cationic
numerous studies, polyethylene glycol (PEG) with a mean molecular lipid molecules are available which are mostly used in combination
weight of 2000 is used as polymer chain conjugated to phosphatidy- with a zwitterionic “helper” lipid, such as 1,2-distearoyl-sn-glycero-3-
lethanolamine (PE) lipids. When the PEGylated lipid is used in a de- phosphoethanolamine (DOPE). In general, the cationic lipid molecules
fined concentration range between 3–5 mol % PEG2000-PE of total li- spontaneously interact with the negatively charged nucleic acid mole-
pids, a polymer coat is formed that tightly surrounds the lipid cules to form a stable complex via electrostatic interactions. Among
nanoparticle and sterically confers stabilization to the particles without such cationic lipids, 1,2-dioleoyl-3-trimethylammonium-propane
inducing particle deformation (Marsh et al., 2003). The inherent hy- chloride (DOTAP) is the most prominent one that was synthesized first
drophilicity and slightly negative charge of the PEG shell diminishes about 30 years ago (Leventis and Silvius, 1990). DOTAP has a qua-
particle uptake by the reticuloendothelial system (RES) and sig- ternary amino head group linked to a glycerol backbone with two C18:1
nificantly prolongs particle´s life-time in blood circulation (Zylberberg chains. At physiological pH-value DOTAP is fully protonated and due to
and Matosevic, 2016). its biodegradable ester bond DOTAP shows a relatively low toxicity.
However, it was shown that repeated injection of PEGylated lipo- Upon particle formation due to self-assembly of DOTAP molecules the
somes causes faster blood clearance rates (Abu Lila et al., 2013). Be- nucleic acid molecules become sandwiched between single lipid bi-
sides, PEGylation might hamper cellular uptake of cationic liposomes layers. The inter-lamellar spacing of DOTAP/OND lipoplexes was de-
by shielding the surface charges (Fang et al., 2017). To overcome this termined by small angle X-ray scattering (SAXS) and transmission
problem cleavable PEGylated lipids were developed that respond either electron microscopy (TEM) to be about 4.9 nm, and the DOTAP bilayer
to the acidic pH (Kale and Torchilin, 2007) or the reductive environ- thickness as 3.72 nm. The interbilayer aqueous layer thickness of about
ment presented in the tumor microenvironment (Wang et al., 2014a). 1.2 nm is appropriate to accommodate sufficient amounts of nucleic
Finally, functionalization of the lipid nanoparticle surface with acid molecules (Weisman et al., 2004). The positive surface charge of
targeting ligands like peptides, proteins, antibodies or aptamers can the nanoparticles further promotes the association with the negatively
assist to enhanced delivery efficacy by specifically targeting cell surface charged cell surface. N-[1-(2,3-dioleyloxy)propyl]-N,N,N-trimethy-
exposed proteins or intracellular organelles. For example, some mem- lammonium chloride (DOTMA) is structurally similar to DOTAP but has
brane active peptides like melittin or the anionic peptide GALA have an ether bond to link the head group to the acyl chains instead of an
the potential to disrupt the endosomal membrane after endocytosis ester bond. While DOTAP and DOTMA are monovalent cationic lipids,
facilitating endosomal escape of ncRNA therapeutics (Hou et al., 2015; 2,3-dioleyloxy-N-[2(sperminecarboxamido)ethyl]-N,N-dimethyl-l-pro-
Tai and Gao, 2017) panaminium trifluoroacetate (DOSPA) and dioctadecylamidoglycyl-
Typically, the targeting ligands are covalently coupled either to spermine (DOGS) are multivalent with different ammonium groups. In
lipid molecules or to the distal ends of the PEG chains. Commonly the DOSPA a multivalent spermine group is linked via a peptide bond to a
linkage is based on amide, thioester, carbamate or disulfide bonds. The quaternary amine conjugated to the hydrophobic alkyl chains, which
conjugation can take place directly by coupling the ligands to the lipid/ are monounsaturated. DOGS is structurally very similar to DOSPA but
PEG molecules which are then used for the preparation of the lipid lacks the quaternary amino group and has two saturated 18-carbon
nanoparticles. Alternatively, the ligand can be coupled to the surface of alkyl chains. Due to the high affinity of the spermine head group to
preformed liposomes. The coupling strategy strongly depends on the nucleic acids, the ONDs can be more efficiently bound. DOSPA and
chemical characteristics of the targeting ligand (Sapra et al., 2005). DOGS are the major components of the commercially available trans-
Targeting moieties recently used for the synthesis of targeted liposomes fection reagents Lipofectamine and Transfectam, respectively. Another
in gene therapy include cyclic (RGD) peptides, folate, transferrin, cationic lipid that is frequently used for transfection is DC-Chol (3ß-[N-
monoclonal antibodies (mABs) and aptamers (listed in detail by (N',N'-dimethylaminoethane)-carbamoyl]cholesterol hydrochloride)
(Zylberberg et al., 2017). The specific targeting approaches for miRNA that contains a tertiary amine in the dimethylethylenediamine head

3
Table 1
Selected list of miRNA loaded lipid nanocarriers.
M. Scheideler, et al.

Lipid composition (molar ratio) miRNA /drug Cells / Organism Targets Diseases References

Cationic Lipoplexes
DOTMA/cholesterol/TPGS 49.5/49.5/1 pre-miRNA-133b A549 Mcl-1 Lung cancer Wu et al. (2011)
DOTMA/cholesterol/TPGS 49.5/49.5/1 miRNA-29b A549, CHO, human non-small cell lung CDK6, DNMT3B, myeloid cell leukemia Lung cancer Wu et al. (2013)
adenocarcinomas tissue samples, mouse sequence 1 (MCL1)
DDAB/cholesterol/TPGS 60/35/5 pre-miR-107 SCC15, SCC25, CAL27, UMSCC74A PKCε, CDK6, HIF1-β Head and neck squamous cell Piao et al. (2012)
carcinoma (HNSCC)
DOTAP/cholesterol/DSPE-PEG 2000-cyanur 1/ let-7a miR CRL-2081, CRL-5830, A549 EphA2 receptor RAS Lung cancer Malignant pleural Lee et al. (2013)
1/0.1 Ephrin-A1 conjugated mesothelioma
DOTAP/Chol/mPEG-DSPE 40/55/5 miRNA-101 SMMC‐7721, HepG2, Huh7 NLK, EZH2, STMN1, Mcl‐1, Rab5A, JunB Hepatocellular carcinoma Xu et al. (2017)
doxorubicin
DOTAP/DSPE-PEG2k/HSPC/cholesterol 9.3/ anti-miR-712 Immortalized mouse aortic endothelial cells TIMP3, MMPs, ADAMs Atherosclerosis Kheirolomoom et al.
3.1/52.6/35VHPK-peptide to target VCAM1 (2015)
Neutral or Anionic Lipoplexes
DOPE/linoleic acid/DMG-PEG 50/48/2 miRNA-29b Kasumi-1, MV4-11, THP-1, KG1, KG1a, OCI- DNMTs, CDK6, SP1, KIT, and FLT3 Acute myeloid leukemia (AML) Huang et al. (2013)
Transferrin conjugated AML3 and patient blasts
DOPE/linoleic acid/DMG-PEG 50/48/2 anti-miRNA-126 Blasts from AML patients; bone marrow cells Leukemia stem cells Acute myeloid leukemia (AML) Dorrance, et al. (2015)
Transferrin or anti-CD45.2 conjugated from wild type mice
DOPE/linoleic acid/DMG-PEG 50/48/2 PEI miR-181a KG1a, MV4-11, HL60, HEK 293 T and HEK KRAS, NRAS and MAPK1 RAS Acute myeloid leukemia (AML) Huang et al. (2016)
Transferrin conjugated 293 T N; blasts from AML patients
HSPC/CHOL/mPEG-DSPE/DSPE-mPEG2000- anti-miRNA-221 HepG2 P27kip1, PTEN, TIMP3 Hepatocellular carcinoma Zhang et al. (2015)
NH2 55/40/4/1 Transferrin conjugated
DOPE/linoleic acid/DMG-PEG 50/48/2 miRNA-1 Humanglioblastoma stem cells MET, EGFR Glioblastom Wang et al. (2014)

4
Transferrin conjugated
DOPC miR-520d-3p SKOV3ip1, HeyA8 Orthotopic metastasis model EphB2, EphA2 Ovarian cancer Nishimura et al. (2013)
EphA2-siRNA
DOPC/Tween 20 miR-2000 Ovarian, lung, renal, basal-like breast cancer interleukin-8, CXCL1 Cancer angiogenesis Pecot et al. (2013)
cells Tumor mouse models
EPC/ CHO/DSPE-PEG2000 49/50/1 Anti- anti-miR-1 Primary neonatal myocardial cells CX43, Kir2.1; Ischemic myocardium Ischemic arrhythmia Liu et al. (2014)
troponin 1 cardiac muscle antibody rat model
conjugated
Ionizable cationic lipid nanoparticles
DSPC/cholesterol/DODAP/ PEG-Cer16 miR-199b-5p HT-29, CaCo-2, SW480, MDA-MB231 T, MCF-7, Hes-1 Colon, breast, prostate, glioblastoma, De Antonellis et al. (2013)
25/45/20/10 PC-3, U-87, Daoy, ONS-76, UW-228 medulloblastoma cancer
DSPC/cholesterol/DODAP/ PEG-Cer16 25/45/ miR-34a multiple myeloma xenografts Erk-2 and Akt Multiple myeloma Di Martino et al. (2014)
20/10
EPC/cholesterol/DODMA/PEG-chol 35/15/45/ miR-122 HepG2, Sk-Hep-1 cells in mouse model Hepatocellular carcinoma Hsu et al. (2013)
5
DODAP/Lac-DOPE/DOPE/DMG-PEG/ anti-miR-155 HepG2 C/EBPβ, FOXP3 Hepatocellular carcinoma Zhang et al. (2013a)
gramicidin A 50/10/28/2/10
DODMA/DOTAP/DOPC/cholesterol/PEG-DPPE anti-miR-21 A549 xenograft mouse PTEN, DDAH1 Lung cancer Yung et al. (2016)
X/Y/36/20/4 (X + Y = 40)
DPPC/DOPE/cholesterol/DCP-TEPA 3/4/4/1 miR-92a HUVEC Integrin α5, mitogen-activated protein Cancer angiogenesis Ando et al. (2013)
kinase kinase 4, sphingosine-1-phosphate
receptor 1
Dlin-KC2-DMA/DSPC/cholesterol/DMG-PEG anti-miR-103 anti- ob/ob mice and diet-induced obese (DIO) Caveolin-1 Type 2 diabetes, obesity Trajkovski et al. (2011)
50/10/38.5/1.5 miR-107 C57BL/6 J mice Adipocytes, liver
YSK05/cholesterol/DMG-PEG 70/30/3 anti-miR-122 murine hepatoma cells, ICR mice liver AldoA, Bckdk, Ndrg3 Liver cancer Hatakeyama et al. (2013)
DSPC/DSDAP/PEG2000 64/30/6 miR-126 HUVEC, mouse SPRED1, PI3KR2 Ischemia Endo-Takahashi, et al.
(2014)
Chemistry and Physics of Lipids 226 (2020) 104837
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

Table 2
Representative lipids used for therapeutic delivery of miRNA.
Lipid Abbreviation Characteristics

1,2-distearoyl-sn-glycero-3-phosphoethanolamine DOPE Helper


1,2-dioleoyl-sn-glycero-3-phosphatidylcholine DOPC Neutral
1,2-distearoyl-sn-glycero-3-phosphocholine DSPC Neutral
1-palmitoyl-2-oleoyl-glycero-3-phosphocholine POPC Neutral
egg-phosphatidylcholine EPY Neutral
soy phosphatidylcholine SPC Neutral
Cholesterol Chol Sterol
cholesteryl hemissucinate CHEMS Sterol
α-(3′-O-cholesteryloxycarbonyl)-δ-(N-ethylmorpholine)-succinamide MoChol Amphoteric
1,2-dimyristoylglycerol-3-hemisuccinate DMGSucc Amphoteric
1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)] (ammonium salt) DSPE-PEG PEGylated
1,2-dimyristoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (ammonium salt) DMPE-PEG PEGylated
1,2-dimyristoyl-rac-glycero-3-methylpolyoxyethylene DMG-PEG PEGylated
1,2-dioleoyl-3-trimethylammonium-propane chloride DOTAP Cationic
1,2-dimyristoyl-trimethylammonium-propane chloride DMTAP Cationic
1,2,-distearoyl- trimethylammonium-propane chloride DSTAP Cationic
N-[1-(2,3-dioleyloxy)propyl]-N,N,N-trimethylammonium chloride DOTMA Cationic
2,3-dioleyloxy-N-[2(sperminecarboxamido)ethyl]-N,N-dimethyl-l-propanaminium trifluoroacetate DOSPA Cationic
dioctadecylamidoglycylspermine DOGS Cationic
3ß-[N-(N',N'-dimethylaminoethane)-carbamoyl]cholesterol hydrochloride DC-Chol Cationic
dimethyldioctadecyl ammonium bromide DDAB Cationic
1,2-distearoyl-3-dimethylamminopropane DSDAP Ionizable
1,2-dioleyloxy-3-dimethylaminopropane DODMA Ionizable
1,2-dioleoyl-3-dimethylammoniumpropane DODAP Ionizable
1,2-dilinoleyloxy-3-dimethylaminopropane DLinDMA Ionizable
1,2-dilinoleoyl-3-dimethylaminopropane DLinDAP Ionizable
1-linoleoyl-2-linoeyloxy-3- dimethylaminopropane DLin-2-DMAP Ionizable
1,2-dilinoleyl-carbamoyloxy-3-dimethylaminopropane DLin-C-DAP Ionizable
2,2-dilinoleyl-4-dimethylaminomethyl-[1,3]-dioxolane DLin-K-DMA Ionizable
2,2-dilinoleyl-4-(2-dimethy aminoethyl)-[1,3]-dioxolane DLin-KC2-DMA Ionizable
1,2-dilinoleylmethyl-4-dimethylaminobutyrate DLin-MC3-DMA Ionizable

group that is conjugated via an ester bond to a cholesterol moiety nanoparticles they can be loaded with large amounts of ONDs. These
(Balazs and Godbey, 2011). A big advantage of DC-Chol is its very low systems are special in terms of particle-endosomal membrane interac-
toxicity confirmed by measurements on various mammalian cell lines tions that are controlled by elasticity energetics and topologically ac-
(Gao and Huang, 1991). A list of the most largely used cationic lipids tivated mechanisms rather than by electrostatics (Kim and Leal, 2015).
for lipoplex formation is provided in Fig. 2. In the following we summarize some examples of lipoplex based
The aim of helper lipids incorporated into lipoplexes is to promote a delivery systems for miRNAs. Most of the formulations are intended for
pH-dependent conversion of a lamellar structure to a non-lamellar cancer therapy e.g. lipoplexes loaded with pre-miRNA-133b were ex-
phase due to the geometry of the helper lipid. DOPE, for example, has a amined for the treatment of lung cancer (Wu et al., 2011). The samples
conical shape with a cross-sectional area of the head group that is much were prepared by ethanol injection technique using a lipid mixture of
smaller than the hydrophobic tail region and thus the packing para- DOTMA/cholesterol/D-α-tocopheryl polyethylene glycol 1000 succi-
meter exceeds “1” (Israelachvili and Mitchell, 1975). Thus, DOPE tends nate (vitamin E TPGS). The final ethanol concentration in the solution
to induce a lamellar to inverted hexagonal phase transition at low pH was 10%. Vitamin E TPGS was added to increase particle stability and
through hydrogen bond formation, and so exerting a destabilizing effect circulation time in blood. The empty liposomes were incubated with
on endosomal membranes to facilitate fusion and cytosolic release of pre-miRNA-133b and used immediately after complexation (Jin et al.,
nucleic acids. The effects of fusogenic neutral helper lipids on lipoplex 2010). The efficacy of the lipoplexes was evaluated in A549 non-small
assembly and structural features relevant for endosomal uptake and cell lung cancer (NSCLC) cells overexpressing MCL-1 protein that could
release are reviewed elsewhere in more detail (Balazs and Godbey, be efficiently downregulated by the use of lipoplex encapsulated pre-
2011; Cheng and Lee, 2016; Wasungu and Hoekstra, 2006). miRNA-133b. The in vivo biodistribution study in mouse showed a 30%
Cholesterol can also be useful as helper lipid that resides in the accumulation of the lipoplex nanoparticles in lung tissues (Wu et al.,
hydrophobic acyl chain region in the lipid bilayer. Higher concentra- 2011). A very similar lipoplex formulation was loaded with miRNA-29b
tions of cholesterol (about 30–40%) incorporated in the lipid bilayer to reduce the expression of the oncogene cyclin-dependent protein ki-
significantly decrease the permeability of the lipid membrane making it nase 6 (CDK6) as a direct target of miRNA-29b in lung cancer (Wu et al.,
more rigid. Through the addition of cholesterol the liposomes become 2013). In this study, the mRNA expression of CDK6 was downregulated
stabilized in vivo and the exchange of lipid molecules between lipo- by ∼ 57% and the tumor growth was inhibited by ∼ 60% compared to
somes and lipoproteins or circulating cells is significantly reduced. In negative control. A comparable cationic formulation prepared with
addition, cholesterol shields the ONDs from nuclease degradation in- dimethyldioctadecyl ammonium bromide (DDAB), cholesterol and vi-
creasing in vivo transfection efficiency of lipoplexes (Sercombe et al., tamin E TPGS was used to transport pre-miR-107 to head and neck
2015). squamous cell carcinoma (HNSCC) cells (Piao et al., 2012). This for-
Another lipid formulation was developed to display a highly or- mulation significantly suppressed the tumorigenesis of HNSCC in in
dered bicontinuous cubic phase as internal structural motif revealing a vitro and in vivo experiments. In another example ephrin-A1 was con-
high colloidal stability combined with steric stabilization. Such nano- jugated to the surface of the DOTAP/cholesterol/DSPE-PEG-cyanur li-
particles called PEGylated cuboplexes are primarily composed of gly- poplexes to target ephrin type-A receptor 2 (EphA2) that is over-
cerol monooleate (GMO), DOTAP and a GMO lipid conjugated to expressed in aggressive malignancies including NSCLC cells (Lee et al.,
polyethylene glycol. Due to the cubic internal structure of the 2013). Ephrin-A1 inhibits proliferation and migration of cancer cells by

5
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

Fig. 2. Chemical structures of representative cationic lipids used for lipoplex formation.

downregulation of EphA2 expression via binding to EphA2 receptors on prevent atheroma formation in atherosclerosis (Kheirolomoom et al.,
the cell membrane. In addition, the tumor-suppressive properties of 2015). To prepare this formulation the hydrophilic anti-miR-712 in
ephrin-A1 are due to the expression of miRNA let-7a. Thus, the com- water and the hydrophobic DOTAP in chloroform were reacted by ad-
bination therapy using ephrin-A1 targeted lipoplexes delivering let-7a dition of methanol to induce the Bligh Dyer monophase for creating a
miRNA to NSCLC cells led to a significantly reduced proliferation rate, hydrophobic DOTAP-anti-miR-712 complex. The complex was ex-
migration and clonogenic expansion of lung cancer cells by down- tracted in the organic phase and added to a mixture of neutral and
regulating Ras mRNA as target (Lee et al., 2013). Only recently, the co- PEGylated lipids. Upon addition of water a stable water-in-oil micro-
delivery of doxorubicin and miR-101, a tumor suppressor miRNA, was emulsion was created. Gradual removal of chloroform resulted in the
achieved using cationic DOTAP liposomes to target hepatocellular formation of cationic lipoparticles with asymmetric lipid bilayers en-
carcinoma (HCC) cells. Synergistic anti-tumor effects of this combina- trapping anti-miR-712 within the core. The particles were about
tion therapy could be validated in vitro and in vivo (Xu et al., 2017). 150 nm in size and slightly negatively charged. To selectively target
Another example for combination therapy using cationic liposome na- vascular cell adhesion molecule 1 (VCAM1) the particles were deco-
nocarriers composed of DOPE, DSPE-PEG and a target modified cationic rated with a targeting peptide sequence (VHPK) by post insertion into
Kojic acid based lipid, which contains a endosomal pH-sensitive imi- the outer leaflet of the preformed miRNA loaded liposomes. Optical
dazole ring, was presented by (Reddy et al., 2016). These nanoparticles imaging validated disease-specific accumulation of anti-miR-712 that
were successful in delivering paclitaxel and Bcl-2 siRNA for the sy- was efficiently delivered to inflamed mouse aortic endothelial cells in
nergistic treatment of melanoma. Recent strategies for combinatorial vitro and in vivo in an Apo E−/− mouse model of atherosclerosis
therapeutic approaches applying co-delivery of ncRNA and anticancer (Kheirolomoom et al., 2015).
drugs are reviewed by (Babu et al., 2017). All these studies amply demonstrate that cationic liposomes – li-
Apart from tumor treatment, cationic lipoplexes containing anti- poplexes – might serve as potent delivery systems for miRNAs, how-
miR-712 were synthesized to target inflamed endothelial cells to ever, problems associated with potential cytotoxicity and

6
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

immunogenicity of cationic lipids or intolerable side effects might pose composed for egg-phosphatidylcholine (EPC)/CHOL/DSPE-PEG-mal
potential limitations and hamper their successful translation into the conjugated to anti-cardiac troponin 1 antibody to deliver anti-miR-1
clinic (Pecot et al., 2011). OND (AMO-1) to ischemic myocardium tissues. By silencing of miR-1 in
ischemic myocardium using AMO-1 loaded liposomes ischemic ar-
3.2. Neutral or negatively charged lipoplexes rhythmogenesis could be efficiently relieved in a rat model after myo-
cardial infarction (Liu et al., 2014).
Except for cationic lipoplexes an interesting approach concerns ac-
tively targeted neutral or negatively charged miRNA loaded lipoplexes 3.3. Ionizable lipids for cationic liposomes: aminolipids
(Huang et al., 2013). Thereby, transferrin (Tf) an 80 kDa glycoprotein is
used as active ligand to target transferrin receptors overexpressed on A new generation of ionizable cationic lipids with low pKa-values
certain cancer or leukemia cells. Potential benefits of neutral or anionic has been developed aiming to achieve a high encapsulation efficiency
formulations are their lower propensity to cause non-specific immune for nucleic acids at low pH values when the lipids are positively
responses and pro-inflammatory reactions (Lonez et al., 2012). Ad- charged. Since ionizable lipids are neutrally charged at physiological
ditionally, the overall neutral surface charge of the lipoplexes is less pH they have a lower toxicity than cationic lipids. Among the first io-
prone to opsonization, plasma protein binding and unspecific cellular nizable aminolipids reported in literature was 1,2-dioleoyl-3-dimethy-
uptake. The anionic lipoplexes are formed by ethanol injection tech- lammoniumpropane (DODAP) with an intrinsic pKa value between 6.6
nique using a lipid mixture of DOPE, linoleic acid and 1,2-dimyristoyl- and 7. However, the apparent pKa value of DODAP can be further ad-
rac-glycero-3-methylpolyoxyethylene (DMG-PEG). The negatively justed to much lower values by increasing the ionic strength of the
charged miRNA molecules are complexed and condensed with low solution (Maurer et al., 2001). When particles are formed at acidic pH
molecular weight polyethylenimine (PEI) before mixing with empty in the presence of up to 40% ethanol a very high encapsulation effi-
lipoplexes. Through sonication the miRNA-PEI core complex is loaded ciency can be achieved. At present, a broad range of ionizable cationic
into the lipoplexes. The particles are about 130 nm in size and slightly lipids with varying chemical structures have been synthesized and as-
negatively charged. The targeting ligand is incorporated by post in- sessed for their transfection efficiency. Representative examples are
sertion technique upon incubation of miRNA-lipoplexes with micelles DLinDMA (1,2-dilinoleyloxy-3-dimethylaminopropane), DLinDAP (1,2-
of DSPE-PEG conjugated with Tf. The Tf-targeted miR-29b loaded li- dilinoleoyl-3-dimethylaminopropane), DLin-2-DMAP (1-linoleoyl-2-li-
poplexes showed anti-leukemic activity by improving the survival rate noeyloxy-3- dimethylamino-propane), DLin-C-DAP (1,2-dilinoleyl-car-
in an acute myeloid leukemia (AML) mouse model (Huang et al., 2013). bamoyloxy-3-dimethylaminopropane), DLin-K-DMA (2,2-dilinoleyl-4-
Lipid nanoparticles with the same composition targeted either with Tf dimethylaminomethyl-[1,3]-dioxolane). The chemical structures of the
or anti-CD45.2 antibodies conjugated to DSPE-PEG2000 maleimide most prominent aminolipids are shown in Fig. 3.
were used to deliver antagomiR-126 to leukemia stem cell (LSC) en- An additional strategy is to add low amounts of poly(ethylene
riched cell subpopulations in AML (Dorrance et al., 2015). The results glycol)-lipids during the formulation process to prevent aggregation.
showed a reduction of LSCs in vivo in mice transplanted with human These nanoparticles termed stable nucleic acid lipid particles (SNALPs)
AML primary blasts taken from AML patients, most likely by depletion reveal a longer lifetime in circulation when administered intravenously
of the quiescent cell subpopulation. The same group was able to show (Semple et al., 2001). The efficiency of SNALPS for nucleic acid delivery
that Tf-targeted lipoplexes delivering miR-181a mimics can target the in vivo was assessed in a Factor VII mouse model (Semple et al., 2010).
RAS-MAPK-pathways presenting a novel promising therapeutic ap- The silencing potential of siRNA containing SNALPs with a lipid com-
proach for the treatment of AML and possibly also for other RAS-driven position of cationic aminolipid/1,2-distearoyl-sn-glycero-3-phos-
cancers (Huang et al., 2016). In another study Tf-targeted lipoplexes are phocholine (DSPC)/cholesterol/ PEG-lipid in a molar ratio of 40/10/
formulated with anti-miR-221 to target HepG2 cells overexpressing Tf- 40/10 was tested using Factor VII as target in hepatocytes. Thus, Factor
receptors. The anionic liposomes successfully accumulate at tumor sites VII becomes secreted in the circulation and could be measured directly
promising a potential therapy of human hepatocellular carcinoma one day after i.v. administration of SNALPs. Following a screening
(Zhang et al., 2015). Finally, Tf-targeted lipoplexes were successfully procedure DLin-KC2-DMA (2,2-dilinoleyl-4-(2-dimethy aminoethyl)-
applied to restore miR-1 levels in patient derived glioblastoma stem [1,3]-dioxolane) was identified as best performing lipid (Semple et al.,
cells (Wang et al., 2014b). 2010). The pKa value of 6.4 indicates that SNALPs based on DLin-KC2-
Nanoparticles based on 1,2-dioleoyl-sn-glycero-3-phosphatidylcho- DMA have limited surface charge in circulation, but become positively
line (DOPC) have been successfully developed for siRNA delivery. In charged in the acidified endosomes. More recently, studies on the use of
principle, DOPC and siRNA are complexed in the presence of excess SNALPs for miRNA delivery became available. One of such examples is
tertiary butanol (Landen et al., 2005). The neutral DOPC particles have miRNA 119b-5p that was efficiently encapsulated into DODAP based
certain advantages over charged particles. For instance, they are less particles composed of DSPC/cholesterol/DODAP/PEG-lipid. The for-
prone to aggregation in biological fluids, they do not preferentially mulation was tested in a range of different tumor cell lines and its
adhere to the endothelium, and are less frequently taken up by sca- delivery efficiency was demonstrated by a significant impairment of the
venger macrophages (Landen et al., 2005). The authors could show that transcription factor HES1 levels and cancer stem cell markers (de
siRNA-DOPC nanoparticles targeted with the oncoprotein EphA2 and Antonellis et al., 2013). The same lipid composition was used to in-
combined with paclitaxel dramatically reduce tumor growth compared corporate miR-34a (Di Martino et al., 2014). In this study, the activity
to treatment with paclitaxel and siRNA separately. The neutral DOPC- was tested against multiple myeloma xenografts in a mouse model in
based nanoparticles are equally efficient for the delivery of miRNA which a significant tumor growth inhibition could be achieved. To
mimics as shown in preclinical testing for tumor targeting (Joshi et al., address liver cancer a lipid formulation based on 1,2-dioleyloxy-3-di-
2014; Nishimura et al., 2013; Pecot et al., 2013; Rupaimoole et al., methylaminopropane (DODMA), which is a lipid with a protonatable
2016b). DOPC liposomes formulated with miRNA-506 mimics or tertiary amino head group, EPC, cholesterol and cholesterol conjugated
miRNA-520 applied in an ovarian cancer orthotopic mouse model re- PEG2000 was loaded with miR-122, a liver-specific tumor suppressor,
sulted in a decreased expression of the respective mRNA targets and to restore deregulated gene expression in HCC cells (Hsu et al., 2013).
significant tumor regression (Nishimura et al., 2013; Yang et al., 2013). Upon reaching the liver the nanoparticles could exit the intravascular
Similarly effective was the delivery of miR-2000 complexed with DOPC space to directly access hepatocytes by passive targeting as long as they
liposomes in orthotopic lung cancer models resulting in a significant are smaller than the pore size of fenestrated vasculature of the liver,
decrease in the primary tumor mass (Pecot et al., 2013). which is about 100–150 nm in diameter (Wisse et al., 2008). Active
Another promising study is based on conventional liposomes targeting of HCC cells was achieved using lactosylated lipid

7
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

Fig. 3. Chemical structures of aminolipids used for the formation of ionizable lipoplexes.

nanoparticles being composed of DODAP, DOPE, DMG-PEG and N- The delivery efficiency of such aminolipid containing formulations
lactobionyl (LAC)-DOPE, as targeting ligand to address asialoglyco- could be further improved by the incorporation of gramicidin A, a
protein receptors on HCC cells. The particles are prepared by ethanol hydrophobic peptide that is known for its channel forming properties in
injection method and loaded with anti-miR-155 by post incubation at lipid bilayers. Presumably, gramicidin A could provide additional ion
room temperature (Zhang et al., 2013a). transport and increase membrane permeability, in turn causing the

8
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

swelling of the endosome and release of the trapped substances into value as important parameter to predict the ability of lipidoid based
cytoplasm, preventing its lysosomal degradation (Bolkent and Zierold, nanoparticles to mediate greater than 95% protein silencing in vivo.
2002). More precisely, lipidoids which were synthesized with amide bonds
A more sophisticated lipid formulation was designed for the de- linking lipid tails containing at least one tertiary amine and at least
livery of anti-miR-21 for lung cancer treatment (Yung et al., 2016). three substitution sites had the highest probability to facilitate potent
These nanoparticles, termed QTsomes, are composed of a mixture of gene silencing in mice. The pKa value appeared to be most influential in
quaternary and tertiary amino cationic lipids, neutral lipids, choles- determining in vivo efficacy of the nanoparticles. It was ruled out that
terol, and a PEG-lipid (DODMA/DOTAP/DOPC/CHOL/DPPE-PEG). The the pKa value should be equal or less than 5.4 (Whitehead et al., 2014).
efficiency was assessed in an A549 xenograft mouse model in terms of The most promising data achieved so far for lipidoids are summarized
tumor regression and a prolonged survival rate (Yung et al., 2016). For in a recent review by Dahlman (Dahlman et al., 2014b). As an example,
example, Ando et al. developed miRNA loaded polycationic QTsomes one lead lipidoid that was synthesized with alkyl acrylate tails con-
for anti-angiogenesis based cancer therapy (Ando et al., 2013). The li- taining ester groups, termed 304O13, efficiently targeted hepatocytes in
posomes were formulated from dicetyl phosphate-tetra- vivo with very low toxicity becoming hydrolyzed by liver enzymes.
ethylenepentamine (DCP-TEPA)/DPPC/DOPE and cholesterol. MiR- Interestingly, the targeting efficiency of the lipidoid nanoparticles to
92a, one of the miRNAs known to regulate angiogenesis, was grafted to liver cells was shown to be dependent on the binding to serum apoli-
cholesterol and bound to the preformed polycationic liposomes. The poprotein E, which is naturally endocytosed by hepatocytes. In case of
authors could show that the miR-92a loaded liposomes were efficiently lipidoids, apolipoprotein E functions as endogenous targeting ligand to
taken up by human umbilical vein endothelial cells (HUVECs) via mi- deliver siRNA-lipidoid nanoparticles to hepatocytes (Akinc et al.,
cropinocytosis and escaped from the endosomes into the cytoplasm 2010). The same effect could not be observed when using cationic li-
suppressing the expression of several proteins encoded by miR-92a posomes. Till now the relationship between nanoparticle features and
target genes (Ando et al., 2013). apolipoprotein E binding propensity remains elusive. In another ex-
Apart from tumor targeting, aminolipid based nanoparticles were ample, concurrent delivery of miR-34a and siRNA targeting Kirsten rat
successfully applied to target the liver for the treatment of metabolic sarcoma viral oncogene analog (KRAS) using polymer based lipid na-
diseases. Such particles were prepared from Dlin-KC2-DMA/DSPC/ noparticles resulted in an improved therapeutic response in combina-
cholesterol/DMG-PEG at a molar ratio 50/10/38.5/1.5 and loaded with tion therapy of lung cancer in a genetically engineered mouse tumor
a combination of antagomirs to silence both miR-103 and miR-107 model (Xue et al., 2014). A simultaneous silencing of multiple en-
(Trajkovski et al., 2011). Here, it was demonstrated that upon miR- dothelial genes was achieved. The nanoparticles were made of low
103/107 inactivation by antagomir loaded nanoparticles, caveolin-1 is molecular weight polyamines and lipids derived from a chemical li-
upregulated in adipocytes leading to improved glucose homeostasis and brary (Dahlman et al., 2014a, b).
insulin sensitivity in obese animals. The results clearly imply that miR- Briefly, small polyamines were conjugated to alkyl tails via an ep-
103/107 are promising therapeutic targets to be silenced by antagomir oxide ring-opening reaction. A variation of amine backbone, lipid
loaded lipid formulations when aiming to treat diabetes (Trajkovski length, and the molar ratio of lipids and amines generated a structurally
et al., 2011). A novel pH-sensitive lipid, termed YSK05, with a pKa diverse nanoparticle library of 600 compounds in ethanol. The com-
value of 6.40–6.45 was formulated with cholesterol and DMG-PEG at a pounds were mixed with DMPE-PEG2000 at a molar ratio of 80 to 20 to
molar ratio of 70/30/3, and loaded with anti-miR-122 to regulate liver form multilamellar structured nanoparticles with a size of about 50 nm
specific miRNA-122. Due to the enhanced escape of the antagomir that are neutral at physiological pH. Applying this formulation in
against miR-122 in the endosome a high activity in liver cells was combination therapy of miR-34a and siRNA against KRAS had a strong
achieved. Systemic administration of YSK05 containing liposomes in- impact on solid lung tumor growth (Xue et al., 2014). In a recent study,
duced the knock-down of miR-122 and an increase in target genes in lipidoid nanoparticles were synthesized by mixing lipidoids, cholesterol
the liver (Hatakeyama et al., 2013). and DOPE to deliver miR-335-5p, which specifically targets the 3’-UTR
In another example, miR-126 was entrapped in 1,2-distearoyl-3- sequence of Dickkopf-1 (DKK1) and regulates its gene expression (Sui
dimethylamminopropane (DSDAP), DSPC and PEG2000 containing et al., 2018). The in vitro and in vivo effects of miR-335-5p transfection
bubble liposomes to treat hindlimp ischemia (Endo-Takahashi et al., on the differentiation of stem cells toward the osteogenic lineage and on
2014). Bubble liposomes contain an ultrasound contrast gas for diag- enhancing calvarial critical-size defect healing in a murine model were
nostic imaging. In this study, therapeutic miR-126 delivery for angio- investigated. It was found that the novel lipidoid-miR-335-5p for-
genic treatment and ultrasound for detection in a hindlimp ischemia mulation is highly suitable to promote calvarial bone regeneration (Sui
model could be combined in a single bubble liposome formulation et al., 2018). In another approach, lipidoid like nanoparticles have been
(Endo-Takahashi et al., 2016). tested to deliver miRNA to human mesenchymal stem cells (hMSCs).
The optimal chemical structures for the lipids were derived from a small
3.4. Lipidoid based nanoparticles lipid library and used to deliver miR-9 to promote neuronal differ-
entiation of stem cells (Takeda, 2016). The nanoparticles were formed
Lipidoids are lipid-like molecules, which are produced by a high- by mixing synthetic double-stranded miRNA mimics with biodegrad-
throughput combinatorial approach (Akinc et al., 2008). So far, a li- able lipids in sodium acetate buffer. The biodegradable lipids were
brary of 1400 degradable lipidoid molecules has been synthesized by synthesized from amines and acrylates of different chain lengths con-
applying Michael addition chemistry. To do so, alkyl-amines were re- taining a disulfide bond that is degraded intracellularly to release the
acted with alkyl-acrylates of varying carbon chain tail length, which miRNA.
have ester groups that can be hydrolyzed by enzymes to make them
biodegradable (Whitehead et al., 2014). A general scheme for lipidoid 3.5. Amphoteric liposomes: Smarticles®
synthesis together with a representative example is provided in Fig. 4.
The efficiency of the lipidoid based particles in siRNA delivery was Amphoteric liposomes are composed of differently charged lipids
screened in hepatocytes and immune cell populations after i.v. ad- including pH-sensitive lipids and an additive. In principle, amphoteric
ministration to mice (Love et al., 2010). The nanoparticles were for- liposomes are cationic at low pH and neutral or anionic at neutral and
mulated from lipidoid molecules, cholesterol, DSPC and DMG-PEG with higher pH. Accordingly, they can be efficiently loaded with ONDs at
a molar ratio of 50/38.5/10/1.5. The particle size varied between 60 low pH. For example, one typical formulation of amphoteric liposomes
and 120 nm in diameter. Based on these screening results, Whitehead includes DOTAP, cholesteryl hemisuccinate (CHEMS), α-(3′-O-choles-
et al. identified four criteria, three structural requirements and the pKa teryloxycarbonyl)-δ-(N-ethylmorpholine)-succinamide (MoChol), 1,2-

9
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

Fig. 4. Scheme for lipidoid synthesis showing one representative example.

dimyristoylglycerol-3-hemisuccinate (DMGSucc) (Siepi et al., 2011). Bothell, WA, and exploited for the delivery of miRNA mimics for tumor
The authors demonstrate that the binding of counterions to charged treatment. Smarticles® are composed of different mixtures of 1-palmi-
lipids promotes the formation of lamellar membranes while ion deso- toyl-2-oleoyl-glycero-3-phosphocholine (POPC), DOTAP, DMGSucc and
rption causes membrane fusion. The amphoteric liposome delivery cholesterol. The particles are about 120 nm in diameter. At physiolo-
technology NOV340 was marketed as Smarticles® by Marina Biotech, gical pH Smarticles® are slightly negatively charged, which might

10
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

prevent their interactions with negatively charged cellular membranes inhibit lung tumor in mice (Trang et al., 2011; Wiggins et al., 2010).
in the endothelium. Once in the tumor the pH tends to be lower and the The same formulation was successfully applied to deliver synthetic
Smarticles® become positively charged thus facilitating their interaction miR-34a mimics to B-cell lymphoma in vivo to reduce tumor growth
with tumor cells. Smarticles® technology was first to enter the clinics with treatment options for aggressive lymphoma. The tumor-suppres-
delivering a miRNA mimic, miR-34a (MRX34). These studies are out- sive effects of miR-34a therapy are attributed to its anti-apoptotic
lined in more detail chapter 5. properties and to the specific knockdown of FoxP1 (Craig et al., 2012).

3.6. Liposome polycationic hybrid particles


3.8. Solid lipid nanoparticles
Apart from cationic lipids, other cationic excipients such as poly-
Solid lipid nanoparticles (SLNs) are colloidal particles harboring a
cationic peptides or stearylamine (SA) can be used to formulate lipid
solid lipid core matrix emulsified by a surfactant that stabilizes the lipid
carriers for gene delivery. Liposome-polycation-hyaluronic acid (LPH)
dispersion. As example, cationic SLNs composed of DDAB, glyceryl
particles containing hyaluronic acid (HA) and a cationic peptide like
monostearate (GMS), polyoxyethylene 50 stearate (Myrj53), choles-
protamine are produced by self-assembly based on charge-charge in-
terol, and soy phosphatidylcholine (SPC) were synthesized by film-ul-
teraction. As an example, HA and miRNA are first complexed with
trasonic techniques and loaded with miRNA by incubation for 30 min at
protamine, a polycationic peptide isolated from salmon sperm, such
room temperature (Shi et al., 2013). The particles have successfully
that the condensed core is negatively charged. This complex is then
proven to deliver miR-34a to cancer stem cells (CSC) in B16F10-CD44+
encapsulated in cationic liposomes composed of DOTAP/cholesterol by
bearing lung tumors in vitro inducing cell apoptosis and inhibiting cell
charge interaction. These nanoparticles are further coated with PEG
migration. In an in vivo study, an accumulation of miR-34a in lung
through post-insertion of PEG-lipids (e.g. DSPE-PEG) and ligand-mod-
tumor sites was observed (Shi et al., 2013). The same group has used
ified PEG-lipids (e.g. DSPE-PEG-anisamide), which specifically target
SLNs to load them with anti-miR-21 to suppress miR-21 in human lung
the sigma receptor overexpressed in B16-F10 melanoma cells. The
cancer cells (Shi et al., 2012). In this case, the antagomirs were first
targeted LPH nanoparticles (PEGylated with anisamide ligand) suc-
complexed with DDAB by a Bligh and Dyer extraction method before
cessfully silenced 80% of luciferase activity in the metastatic B16-F10
formation of SLNs through solvent diffusion (Reimer et al., 1995). In
tumor in the lung after a single i.v. injection (Chono et al., 2008). In
this case, an aqueous poloxamer 188 solution was used to form an oil in
another study, PEGylated LPH nanoparticles were surface functiona-
water emulsion prior to removal of the organic solvent. Cellular uptake
lized with cyclic RGD peptides and loaded with antisense inhibitors of
and activation properties of antagomir loaded SLNs were tested in
miR-296 to target αvβ3 integrin positive endothelial cells in tumor neo-
human lung adenocarcinoma A549 cells, including studies on antisense
vasculature (Liu et al., 2011). The potential of cyclic RGD targeted LPH
efficiency, cell migration and invasion (Shi et al., 2012).
nanoparticles for anti-angiogenic therapy using antagomirs was shown
in a matrigel plug assay in BALB/c nude mice (Liu et al., 2011). In a
separate study, the PEGylated LPH particles were modified with the 3.9. Nonionic surfactant vesicles
single-chain variable fragment (scFv) of the N-cadherin antibody (GC4)
to target lung metastasis in a syngeneic murine model (Chen et al., Nonionic surfactant vesicles are formed by self-assembly from
2010). The delivery efficacy to tumor metastasis was tested with both, nonionic surfactants and excipients, most likely cholesterol, and are
siRNA und miR-34a. MiR-34a is an important p53 regulated tumor often referred to as niosomes (Moghassemi and Hadjizadeh, 2014).
suppressor. Encapsulated in targeted LPH particles miR-34a induced Niosomes are promising carriers for drugs as hydrophilic and hydro-
apoptosis, inhibited survivin expression and downregulated MAPK phobic molecules can be incorporated in the aqueous interior of the
pathways in B16-F10 melanoma cells. Interestingly, when miR-34a and vesicles or within the closed surface bilayer, respectively. Cationic
siRNAs are co-formulated in LPH nanoparticles targeting GC4 a pro- niosomes formulated by nonionic surfactants such as sorbitan mono-
nounced inhibition of tumor growth was found in lung metastasis oleate (Span 80), polyoxyethylenesorbitan trioleate (Tween 85),
bearing mice (Chen et al., 2010). These data illustrate for the first time monopalmitin glycerol, cholesterol and DDAB have been validated as
the potential of combined delivery of siRNA and miRNA in a single efficient transporter for siRNA to cancer cells displaying a high loading
delivery system to target different oncogenic pathways and open up the efficiency combined with low toxicity (Obeid et al., 2017; Sun et al.,
avenue towards future personalized combination therapies in cancer 2015). Recently, a highly interesting approach describes cationic nio-
treatment. somes as theranostic platform for the delivery of siRNA/miRNA to
An SA based cationic liposome formulation was designed for the human mesenchymal stem cells (hMSCs) to promote their differentia-
delivery of anti-miR-191 to breast cancer cell lines. A combined treat- tion. Concurrently, a dye was encapsulated in the niosomes for in vivo
ment of SA liposome loaded with anti-miR-191 and anti-cancer drugs tracking of the transfected stem cells (Yang et al., 2018). The niosomes
markedly enhanced apoptotic cell death and suppressed the migration are made of Span 80, TGPS and DOTAP by ethanol injection technique
of cancer cells as shown in vitro in breast cancer cell lines (Sharma et al., incorporating the amphiphilic dye indocyanine green (ICG), which is
2017). self-quenched due to close proximity of the dye molecules but can be
visualized upon release in the near-infrared (NIR) region. siRNAs/
3.7. Neutral lipid emulsions miRNAs are complexed on the surface by electrostatic interaction with
the positive head groups of DOTAP. In this study anti-miR-138 was used
Neutral lipid emulsions (NLE) used for drug delivery usually consist as gene regulator to promote osteogenic differentiation in hMSCs. The
of a phosphatidylcholine, mostly DOPC, oils like middle-chain (MCT) or particles were efficiently taken up intracellularly resulting in specific
long-chain (LCT) triglycerides, polysorbates and antioxidants. NLEs gene silencing and NIR labeling of hMSCs upon niosome decomposi-
have some advantages over other lipid formulations. Similar to the tion. For in vivo stem cell tracking the NIR labeled transfected hMSCs
behavior of neutral liposomes, NLEs are highly stable in biological were injected subcutaneously into mice and imaged for seven days
fluids and do not tend to form aggregates due to electrostatic repulsion. following implantation. By this way, simultaneous long-term cell
They show low toxicity, are not filtered by the liver, do not adhere to tracking and in vivo gene silencing could be achieved within one par-
endothelium, and are not taken up by scavenging macrophages (Trang ticle. Following these promising results the authors conclude that
et al., 2011; Wiggins et al., 2010). A disadvantage of lipid emulsions is theranostic niosomes could present a novel platform for gene regulation
that they stay in circulation with limited cellular access. NLEs have in both stem cell research and regenerative medicine (Yang et al.,
been successfully applied for the systemic delivery of miRNA mimics to 2018).

11
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

4. Structural and functional aspects of miRNA delivery cancer therapy to target receptors overexpressed in many cancer cells.
Other ligands for nanoparticle targeting in cancer therapy are anti-
4.1. Structural organization of oligonucleotide loaded lipid-based bodies against VEGF, VCAM, matrix metalloproteases (MMPs) or in-
nanoparticles tegrins (Zylberberg and Matosevic, 2016). To some extent these ligands
are also promising to target inflamed vascular endothelium or angio-
Currently little information is available about the internal structural genesis. Endothelial cell adhesion molecules, like VCAM-1, E-selectin,
organization of OND or siRNA loaded lipid-based nanoparticles lacking P-selectin are preferentially exposed on the surface of activated en-
specific data on miRNAs. However, due to the compositional and dothelium and can be targeted by antibodies covalently conjugated to
structural similarity of ONDs the mechanism of interaction accom- the delivery system (Howard et al., 2014). As example, E-selectin
panying the self-assembling process of the lipid particles as well as the coupled immunoliposomes are readily taken up by receptor-mediated
inner molecular organization should rather depend on the lipid com- endocytosis by E-selectin expressing interleukin-1 activated endothelial
position, the molar lipid to nucleic acid ratio and the preparation cells (Jubeli et al., 2012; Kessner et al., 2001). Peptide ligands primarily
technique than on the specific OND to be incorporated. For lipid na- belong to the class of cell penetrating peptides (CPP) that translocate
noparticles containing ionizable cationic lipids produced by rapid mi- through the membrane. Thus, cyclic-RGD, TAT-peptides or octa-argi-
crofluidic mixing processes an electron-dense nanostructured core was nine peptides have been shown to be very effective for gene delivery
identified in the particle´s interior (Leung et al., 2012). The internal and transfection (Kibria et al., 2011; Levchenko et al., 2003; Zhang
structure organization and the localization of the siRNA payload was et al., 2006). A new approach explores aptamers selected against cell
deduced from cryo-transmission electron microscopy (cryoTEM), P31- surface receptors as targeting sequences for siRNA loaded SNALPS
NMR, fluorescent energy resonance transfer (FRET), density gradient (Wilner and Levy, 2016).
centrifugation, and molecular modeling (Cullis and Hope, 2017). The In general, the targeting ligands are conjugated to the surface of the
particles were composed of DLinKC2-DMA, DSPC, cholesterol and a nanoparticles to be recognized by specific receptors or proteins ex-
PEG2000-lipid. For microfluidics, the lipids were dissolved in ethanol pressed on the cell surface. The ligands can be coupled covalently by
and mixed with an aqueous acidic solution of ONDs, followed by ex- chemical reaction either to head group functionalized phospholipids or
tensive dialysis to remove ethanol (Belliveau et al., 2012). Depending the distal end group of functionalized PEG chains anchored to lipid
on the mixing conditions homogenous monodisperse lipid nanoparticle molecules (Nobs et al., 2004). The active transport across the cell
of tunable sizes between 20–100 nm and a very high encapsulation membrane permits tissue accumulation of the nanoparticles in diseased
range could be produced (Belliveau et al., 2012). Leung et al. found that sites before drug release occurs within intracellular compartments.
the lipid nanoparticles had a highly structured electron dense inner core
in which the ionizable lipids are arranged as inverted micellar struc- 4.3. Cellular uptake and intracellular trafficking
tures complexing siRNA molecules. The outer shell is formed by a
monolayer consisting predominantly of neutral lipids and PEGylated Effective cellular internalization and proper intracellular release are
lipids. Viger-Gravel et al. (Viger-Gravel et al., 2018) investigated a re- particularly relevant for RNA based therapeutics, which require cyto-
lated lipid nanoparticle system composed of the ionizable cationic lipid solic delivery for their bioactivity (Schroeder et al., 2010; Whitehead
(DLin-MC3-DMA), DSPC, cholesterol, and DMPE-PEG2000. Again, the et al., 2009). In case of cationic liposomes, the particles interact with
particles were synthesized by microfluidics and measured by dynamic the negatively charged cell surface preferentially via proteoglycans and
nuclear polarization enhanced NMR spectroscopy. With this technique sialic acids or by unspecific charge-mediated interactions with cellular
the internal spatial location of the single components could be detected. receptors. The uptake occurs most likely by endocytosis (for a com-
The authors found a more homogenous distribution throughout the prehensive review see (Zuhorn and Hoekstra, 2002)). Following en-
particles comprising a thin outer shell formed by DSPC and PEG-lipid docytosis, the membrane-like structure of the positively charged lipo-
and an interior being made up by evenly dispersed inverted micelles of somes allows the exchange of charged lipids between the endosomal
ionizable cationic lipid and cholesterol. In general, cationic liposomes membrane and the liposomes facilitating intracellular release of the
of defined size can be manufactured by microfluidic rapid mixing nucleic acids (for a review see (Leung et al., 2014)). In particular, fu-
through carefully tuning the manufacturing process parameters. Thus, sogenic helper lipids added in the formulation can destabilize the en-
homogeneous lipoplex suspensions in a size range from 20 to 100 nm dosomal membrane by conformational lipid transition at the low en-
can be produced by adjusting the total flow rate and the flow rate ratio dosomal pH. For instance, DOPE adopts an inverted hexagonal phase at
(i.e. the ratio between the organic solvent containing lipids and aqueous low pH, which readily causes fusion of DOPE containing liposomes with
buffer containing ONDs). A very general presentation of the structure of the endosomal membrane (Hafez and Cullis, 2001; Hafez et al., 2001).
OND-loaded unilamellar liposomes in comparison to lipid nanoparticles The fusion process destabilizes the liposomes and their cargo is directly
containing cationic ionizable lipids is presented in Fig. 5. released into the cytoplasm (Torchilin, 2007). Pure cationic liposomes
such as DOTAP liposomes efficiently incorporate miRNA/siRNA, but
4.2. Targeted active transport due to their intracellular stability the release rate of active substances
and endosomal escape capacities might be limited. The particles remain
Active transport across biological barriers is specifically important entrapped inside the endosome and are trafficked towards the lyso-
in cases when passive targeting depending on the EPR effect is not vi- somes for degradation (Vermeulen et al., 2018a). Potential cellular
able. The strategy of passive targeting has been successfully used to uptake and release scenarios for miRNA lipoplexes are drawn in Fig. 6.
reach sites of leaky vasculature typical for tumors, sites of inflamma- In case of pH-sensitive lipid nanoparticles the escape mechanism
tion, infection or angiogenesis. Active targeting is a more specific ap- was suggested to follow the so called “proton sponge” effect. As the pH
proach to address particular molecules uniquely present or enriched in value inside the endosome drops the nanoparticles become protonated,
cells, tissues or pathological sites of interest (Howard et al., 2014). As which causes an increased influx of protons and counter ions, such as
targeting ligands for liposomes a broad variety of molecules including chloride, increasing the osmotic pressure in the endosomes. The os-
proteins, antibodies, antibody fragments, peptides, aptamers or small motic gradient leads to water influx causing rupture and release of its
molecules has been explored. The reader is referred to some re- contents into the cytoplasm (Paliwal et al., 2015). Current evidence
presentative reviews on active targeting approaches for liposomes suggests that the osmotic pressure by itself is not enough to cause ly-
(Byrne et al., 2008; Deshpande et al., 2013; Pattni et al., 2015; Petrilli sosomal rupture (Wolfram et al., 2015; Won et al., 2009). At low pH
et al., 2014; Sawant and Torchilin, 2012; Torchilin, 2007). In brief, hydrophobic or electrostatically-driven adsorption of polycation mole-
transferrin and folic acid are amongst the most promising ligands in cules to the endosome membranes are likely to occur. The osmotic

12
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

Fig. 5. Schematic of oligonucleotide loaded lipoplexes/lipid nanoparticles formulated by rapid mixing. The lipoplex nanoparticles (A) are composed of a mixture of
neutral lipids/fusogenic lipids, PEGylated lipids, cholesterol and cationic lipids. Sterically stabilized liposomes are formed. (B) For lipid nanoparticles containing
ionizable cationic lipids that are formed by an ethanol loading formulation process, a structural scheme of inverted micelles surrounded by a PEGylated lipid
monolayer was suggested (Cullis and Hope, 2017).

swelling of the cell membrane further facilitates the protrusion of endosomal rupture is abolished.
polycations into the lipid bilayer thereby destabilizing the membrane Although there is still controversy in the literature upon endosomal
causing leakiness. Once the membrane becomes leaky the osmotic escape mechanisms it is important to note that the phenomena of in-
pressure cannot be maintained any longer and proton sponge-based tracellular trafficking are dependent on various parameters including

Fig. 6. Proposed cellular uptake and release mechanisms for miRNA loaded lipoplexes.

13
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

the composition of the nanoparticles, the cell type, and the endosomal limited treatment options. Importantly, miRNA expression profiles have
size and leakiness. For further details, the reader is referred to a recent to play a central role in the gene regulation of the disease to be ther-
topical review on the pro and cons of the proton sponge hypothesis apeutically manipulated (Rupaimoole and Slack, 2017). Forthcoming
(Vermeulen et al., 2018b). progress in biomarker discovery will certainly lead to new develop-
ments of miRNA therapeutics offering personalized treatment options
4.4. Cytotoxicity and safety for patients. Hence, it can be expected that novel strategies to use un-
ique OND based nanopharmaceuticals addressing unmet clinical needs,
The applicability of cationic liposomes in vivo is often hampered by will further attract the attention from biotech and pharmaceutical
their inherent dose-dependent toxicity. Multivalent cationic liposomes companies (Conde and Artzi, 2015; Pérez and Carlucci, 2018). Al-
are generally more toxic than monovalent cationic lipids. Even the though the commercial development remains a challenge (Ragelle
hydrocarbon chain length could influence the cytotoxicity of cationic et al., 2017), an increasing number of nanomedicines have received
lipids (Filion and Phillips, 1997). These authors found that the toxicity regulatory approval within the last few years. This also led to the first
of liposomes to macrophages decreases in mixtures containing DOPE Food and Drug Administration (FDA) approved RNAi therapeutic
and cationic lipids based on diacyltrimethylammonium propane from (Mullard, 2018). This siRNA therapeutics with the trade name patisiran
DOPE/DOTAP (dioleoyl-) > DOPE/DMTAP (dimyristoyl-) > DOPE/ was registered by Alnylam Pharmaceuticals in 2018 for the treatment of
DPTAP (dipalmitoyl-) > DOPE/DSTAP (distearoyl-) acyl chains. By in- hereditary transthyretin-mediated (ATTR) amyloidosis, a rare and po-
corporation of polymer conjugated lipids in the lipid formulation the tentially fatal disease (Adams et al., 2018). In partisiran the siRNA is
toxicity can be significantly reduced (Xue et al., 2015). For neutral li- formulated as hepatotropic lipid nanoparticle for intravenous (IV) ad-
posomes no toxicity has been reported so far (Landen et al., 2005). ministration. The lipid nanoparticle is composed of a modified ionizable
Negatively charged liposomes are of low toxicity, but they can induce cationic lipid (dilinoleylmethyl-4-dimethylaminobutyrate, DLin-MC3-
immunogenic response, and are less likely to cross negatively charged DMA) in combination with DSPC, cholesterol and a PEG-lipid.
cell membranes (Schroeder et al., 2010). Cationic lipids might also MRX34 is the first and only miRNA therapeutic that has entered a
activate the complement system, which results in binding of comple- phase 1 clinical trial in patients with multiple cancers including pri-
ment components to liposomes. Liposomes are then directed to specific mary liver cancer or metastatic cancer with liver involvement in 2013.
receptors found in the lung or in Kupffer cells in the liver (Landesman- The miRNA mimic is intended to restore the lost suppressor function of
Milo and Peer, 2012). Treatment with cationic liposomes might also miR-34 on the p53 and wnt/β-catenin cellular pathways (Baumann and
provoke a pro-inflammatory response by inducing Th1 cytokine ex- Winkler, 2014). The therapeutic MRX34 directly regulates at least 24
pression. As liposomes might trigger the innate immune system and known oncogenes, such as those involved in the cell cycle and pro-
macrophage clearing mechanisms (Szebeni and Moghimi, 2009), po- liferation, anti-apoptosis, metastasis, chemoresistance, cancer cell self-
tential toxicities associated with lipid carriers need to be addressed renewal and oncogenic transcription (Bader, 2012). The miRNA mimics
carefully before a translation in clinical practice can be implemented. In are delivered by the NOV340 technology (Smarticles®), as described
particular, a careful selection of lipids and formulation strategies might above. The miRNA mimics and the lipids are mixed under acidic con-
help to get rid of adverse cytotoxic effects. Given recent advances in the ditions, when the amphoteric lipids are cationic. At physiological pH
chemical synthesis of novel lipid compounds, new generations of lipid the complexes are slightly anionic to reduce the interaction propensity
formulations were designed which show significantly lower toxicity in with negatively charged cell membranes. Since the pH tends to be lower
vivo. However, immunogenicity and cytotoxicity issues are still pro- in tumor environment the Smarticles® become cationic and increasingly
blematic for clinical applications, in particular if high doses and fre- adhere to tumor cells. The efficacy of miR-34a loaded particles to target
quent repetitive dosing are required (Zatsepin et al., 2016). For ex- the liver and to promote tumor regression was verified in a clinically
ample, the clinical trial for the treatment of hypercholesterolemia using relevant mouse model of HCC (Bader, 2012). Mirna Therapeutics
siRNA/LNPs to target ApoB production was terminated due to potential (Mirna Therapeutics, Inc., Austin, TX, USA) has initiated a preclinical
immunogenic stimulation (see clinical trial number: NCT00927459). development program to support manufacturing of cGMP material of
Continued success depends on the improving the safety profile while MRX34. A phase 1 clinical study using MRX34 was launched in patients
maintaining efficient delivery (Zatsepin et al., 2016). with advanced solid tumors critically evaluating dosage issues, safety
aspects, pharmacokinetics and off-target effects (Beg et al., 2017). 74
5. Clinical developments: translation of lipid based miRNA patients with various solid tumors, including HCC (n = 14) were en-
therapeutics to the clinics rolled. MRX34 was given intravenously twice a week for three weeks in
four week cycles. The maximal tolerated dose, the optimal dosing re-
Today, several lipid-based technologies are successfully established gime and occurring adverse side effects were assessed. The results show
as proof of principle for miRNA delivery in cellular and pre-clinical that MRX34 therapy is feasible and tolerable, but only under adequate
settings. Thus, a considerable number of studies has been conducted dexamethasone pretreatment. It was detected that most of the adverse
over the years and the results of all these studies provide important effects are potentially attributed to the carrier independent of the
information regarding the therapeutic potential of the carrier system miRNA mimic. Unfortunately, in 2016 Mirna Therapeutics was forced
and its cargo to enable the translation into clinical development (Wang to halt the clinical study following multiple immune-related adverse
et al., 2015). However, clinical translation of the miRNA technology has events diagnosed in patients dosed with MRX34. Interestingly, the same
proven challenging. Problems associated with the establishment of liposomal carrier used in combination with a single-stranded DNA oli-
scaling-up procedures for cGMP production, molecular stability and gonucleotide for Bcl-2 targeted therapy (PTN2258, Sierra Oncology
robustness of the therapeutic delivery system, quality control me- Plymouth, MI; former ProNAi Therapeutics Inc.), which was tested in
chanisms and higher costs associated with the production of innovative multiple advanced solid tumor (phase I) and in patients with relapsed
nanopharmaceuticals remain and have to be overcome before clinical refractory non-Hodgkin lymphoma and diffuse large B cell lymphoma
applications can be successfully realized (Chakraborty et al., 2017; (phase II) did not show related severe side effects. PTN2258 has been
Zhang et al., 2013b). From the idea to the market the development of granted orphan drug destination by the FDA in 2016. Since the focus of
nanopharmaceuticals takes about ten to twelve years as shown in a the company has changed from RNA knockdown approaches to tar-
tentative timeline in Fig. 7 (Ragelle et al., 2017). In view of this long geting DNA damage response the development of PNT2258 was ter-
and costly way, some general considerations should be taken into ac- minated in 2017. Despite substantial progress, some disappointing re-
count. When designing novel carrier systems for miRNA therapeutics sults from clinical human studies indicate that substantial
researcher should focus on untreatable or poorly treated diseases with improvements of the carrier systems for nucleic acid therapeutics are in

14
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

Fig. 7. Tentative time line for pharmaceutical developments from the initial design of the drug delivery system to the final commercialized nanopharmaceuticals.

demand and that the molecular mechanisms of nanoparticle interac- nanoparticle to form a stable polymer corona surrounding the particle
tions with blood components, cellular recognition, uptake, release and surface. Fourth, targeting ligands can be included in the formulation to
metabolic response still need to be investigated and understood in more improve specific recognition and uptake by target cells and tissues.
detail. In addition, combination products that enable smart delivery of Apart from efficient and safe delivery, proper pharmacokinetic and
ncRNA therapeutics together with existing potent drugs, may present biodistribution profiles in clinical relevant models, stability and up-
the next revolution in therapy (Conde and Artzi, 2015; Ragelle et al., scaling issues, questions of cost efficiency hamper rapid translation of
2017). nanopharmaceuticals to the clinic. In this vein, advances in the design
of proper delivery systems may address a number of shortcomings as-
sociated with conventional medicine but the complexity of the in-
6. Concluding remarks
novative systems will certainly pose new challenges for clinical ap-
proval (Eifler and Thaxton, 2011). MiRNA based therapeutic
Indeed, there is a strong need of site-specific delivery systems that
approaches, applied either as miRNA mimics in miRNA replacement
integrate several features as high drug loading capacity, stability, im-
therapy with the aim to restore the function of miRNAs, or alternatively
proved circulation time, reduced toxicity and protection of its cargo
as miRNA antagonists for specific targeting and silencing of miRNAs are
from rapid degradation. In circulation an enhanced transport to the
still at the very beginning. Nevertheless, the initial stages have already
target tissue, proper cellular uptake and drug release are anticipated.
been completed with the first FDA approval for a lipid nanoparticle
Lipid-based nanoparticles are very promising candidates for therapeutic
ncRNA drug. Due to the fact that there is a huge number of so far
delivery. However, many physicochemical and structural properties
‘undruggable’ proteins and diseases, it can be expected that an in-
have to be taken into consideration when designing a lipid nanocarrier,
creasing number of studies will address these challenges to establish
especially for sensitive drugs like ncRNAs. First, to ensure efficient
unified delivery systems for siRNA/miRNA with specific targeting to
complexation of the negatively charged nucleic acids with lipids posi-
cell types and organs for clinical applications.
tively charged molecules are very helpful. By electrostatic interactions
the particles can be loaded during formation via lipid self-assembly. As
positively charged nanoparticles are often toxic, pH sensitive lipids 7. Perspectives
were developed, which are neutral at physiological pH but are posi-
tively charged under low pH conditions present in endosome or tumor Microfluidics has emerged as a powerful tool for the controlled
microenvironment. Second, the lipid carriers need to have a certain synthesis of lipid-based delivery systems with high encapsulation effi-
size, which is in a range between 50 to 200 nm to avoid renal filtration ciencies for ncRNA molecules. This technology allows a robust, fast and
or rapid clearance by the immune system. However, the optimal par- large scale production of highly homogeneous formulations under GMP
ticle size strongly depends on the application and the target tissue. conditions. In combination with advanced and standardized physico-
Third, to enhance circulation time and to reduce carrier uptake by RES, chemical techniques reliable quality control assurance can be guaran-
the nanoparticles might be decorated and coated with polymers. teed. The development of efficient targeted ncRNA delivery systems for
Accordingly, PEGylated lipids are incorporated into the lipid clinical settings will depend on both, the proper synthesis of new lipid

15
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

components and novel targeting ligands, in close association with bio- Nucleic Acids 1, e37.
marker research to address disease targets more specifically. We believe Bertrand, N., Grenier, P., Mahmoudi, M., Lima, E.M., Appel, E.A., Dormont, F., Lim, J.M.,
Karnik, R., Langer, R., Farokhzad, O.C., 2017. Mechanistic understanding of in vivo
that combination treatments uniting classical drugs with gene ther- protein corona formation on polymeric nanoparticles and impact on pharmacoki-
apeutics within one single lipid nanocarrier will finally promote the netics. Nat. Commun. 8, 777.
clinical success. Bolkent, S., Zierold, K., 2002. Effects of the ionophores valinomycin, ionomycin and
gramicidin A on the element compartmentation in cultured rat hepatocytes. Toxicol.
At the end the reader is referred to a recent comment by Twan In Vitro 16, 159–165.
Lammers and colleagues, who state that a rigorous, realistic and holistic Byrne, J.D., Betancourt, T., Brannon-Peppas, L., 2008. Active targeting schemes for na-
rethinking is needed to improve nanomedicine performance and in- noparticle systems in cancer therapeutics. Adv. Drug Deliv. Rev. 60, 1615–1626.
Campani, V., Salzano, G., Lusa, S., De Rosa, G., 2016. Lipid nanovectors to deliver RNA
crease patient benefit (Lammers et al., 2016). The authors recommend oligonucleotides in cancer. Nanomaterials (Basel, Switzerland) 6.
to think beyond delivery and targeting, and to focus on combination Carrstensen, H., Muller, R.H., Muller, B.W., 1992. Particle size, surface hydrophobicity
therapies, protocols for patient selection and ways to enable faster and and interaction with serum of parenteral fat emulsions and model drug carriers as
parameters related to RES uptake. Clin. Nutr. 11, 289–297.
more efficient clinical translation. This notion is especially true for new
Chakraborty, C., Sharma, A.R., Sharma, G., Doss, C.G.P., Lee, S.S., 2017. Therapeutic
biomolecular drugs, nucleic acid based therapeutics, and the design of miRNA and siRNA: moving from bench to clinic as next generation medicine. Mol.
advanced carrier systems. Ther. Nucleic Acids 8, 132–143.
Chen, D., Love, K.T., Chen, Y., Eltoukhy, A.A., Kastrup, C., Sahay, G., Jeon, A., Dong, Y.,
Whitehead, K.A., Anderson, D.G., 2012. Rapid discovery of potent siRNA-containing
Declaration of Competing Interest lipid nanoparticles enabled by controlled microfluidic formulation. J. Am. Chem. Soc.
134, 6948–6951.
The authors declare that there are no conflicts of interest. Chen, Y., Gao, D.-Y., Huang, L., 2015. In vivo delivery of miRNAs for cancer therapy:
challenges and strategies. Adv. Drug Deliv. Rev. 81, 128–141.
Chen, Y., Zhu, X., Zhang, X., Liu, B., Huang, L., 2010. Nanoparticles modified with tumor-
Acknowledgements targeting scFv deliver siRNA and miRNA for cancer therapy. Mol. Ther. 18,
1650–1656.
Cheng, X., Lee, R.J., 2016. The role of helper lipids in lipid nanoparticles (LNPs) designed
The authors are grateful to the Austrian Funding Agency (FWF), for oligonucleotide delivery. Adv. Drug Deliv. Rev. 99, 129–137.
Austria for financial support. Choi, H.S., Liu, W., Misra, P., Tanaka, E., Zimmer, J.P., Itty Ipe, B., Bawendi, M.G.,
Frangioni, J.V., 2007. Renal clearance of quantum dots. Nat. Biotechnol. 25,
1165–1170.
References Chono, S., Li, S.D., Conwell, C.C., Huang, L., 2008. An efficient and low im-
munostimulatory nanoparticle formulation for systemic siRNA delivery to the tumor.
Abu Lila, A.S., Kiwada, H., Ishida, T., 2013. The accelerated blood clearance (ABC) J. Control. Release 131, 64–69.
phenomenon: clinical challenge and approaches to manage. J. Control. Release 172, Conde, J., Artzi, N., 2015. Are RNAi and miRNA therapeutics truly dead? Trends
38–47. Biotechnol. 33, 141–144.
Adams, D., Gonzalez-Duarte, A., O’Riordan, W.D., Yang, C.C., Ueda, M., Kristen, A.V., Craig, V.J., Tzankov, A., Flori, M., Schmid, C.A., Bader, A.G., Müller, A., 2012. Systemic
Tournev, I., Schmidt, H.H., Coelho, T., Berk, J.L., Lin, K.P., Vita, G., Attarian, S., microRNA-34a delivery induces apoptosis and abrogates growth of diffuse large B-
Plante-Bordeneuve, V., Mezei, M.M., Campistol, J.M., Buades, J., Brannagan 3rd, cell lymphoma in vivo. Leukemia 26, 2421.
T.H., Kim, B.J., Oh, J., Parman, Y., Sekijima, Y., Hawkins, P.N., Solomon, S.D., Cullis, P.R., Hope, M.J., 2017. Lipid nanoparticle systems for enabling gene therapies.
Polydefkis, M., Dyck, P.J., Gandhi, P.J., Goyal, S., Chen, J., Strahs, A.L., Nochur, S.V., Mol. Ther. 25, 1467–1475.
Sweetser, M.T., Garg, P.P., Vaishnaw, A.K., Gollob, J.A., Suhr, O.B., 2018. Patisiran, Dahlman, J.E., Barnes, C., Khan, O., Thiriot, A., Jhunjunwala, S., Shaw, T.E., Xing, Y.,
an RNAi therapeutic, for hereditary transthyretin amyloidosis. N. Engl. J. Med. 379, Sager, H.B., Sahay, G., Speciner, L., Bader, A., Bogorad, R.L., Yin, H., Racie, T., Dong,
11–21. Y., Jiang, S., Seedorf, D., Dave, A., Sandu, K.S., Webber, M.J., Novobrantseva, T.,
Akinc, A., Querbes, W., De, S., Qin, J., Frank-Kamenetsky, M., Jayaprakash, K.N., Ruda, V.M., Lytton-Jean, A.K.R., Levins, C.G., Kalish, B., Mudge, D.K., Perez, M.,
Jayaraman, M., Rajeev, K.G., Cantley, W.L., Dorkin, J.R., Butler, J.S., Qin, L., Racie, Abezgauz, L., Dutta, P., Smith, L., Charisse, K., Kieran, M.W., Fitzgerald, K.,
T., Sprague, A., Fava, E., Zeigerer, A., Hope, M.J., Zerial, M., Sah, D.W., Fitzgerald, Nahrendorf, M., Danino, D., Tuder, R.M., von Andrian, U.H., Akinc, A., Schroeder, A.,
K., Tracy, M.A., Manoharan, M., Koteliansky, V., Fougerolles, A., Maier, M.A., 2010. Panigrahy, D., Kotelianski, V., Langer, R., Anderson, D.G., 2014a. In vivo endothelial
Targeted delivery of RNAi therapeutics with endogenous and exogenous ligand-based siRNA delivery using polymeric nanoparticles with low molecular weight. Nat.
mechanisms. Mol. Ther. 18, 1357–1364. Nanotechnol. 9, 648–655.
Akinc, A., Zumbuehl, A., Goldberg, M., Leshchiner, E.S., Busini, V., Hossain, N., Dahlman, J.E., Kauffman, K.J., Langer, R., Anderson, D.G., 2014b. Nanotechnology for in
Bacallado, S.A., Nguyen, D.N., Fuller, J., Alvarez, R., Borodovsky, A., Borland, T., vivo targeted siRNA delivery. Adv. Genet. 88, 37–69.
Constien, R., de Fougerolles, A., Dorkin, J.R., Narayanannair Jayaprakash, K., de Antonellis, P., Liguori, L., Falanga, A., Carotenuto, M., Ferrucci, V., Andolfo, I.,
Jayaraman, M., John, M., Koteliansky, V., Manoharan, M., Nechev, L., Qin, J., Racie, Marinaro, F., Scognamiglio, I., Virgilio, A., De Rosa, G., Galeone, A., Galdiero, S.,
T., Raitcheva, D., Rajeev, K.G., Sah, D.W., Soutschek, J., Toudjarska, I., Vornlocher, Zollo, M., 2013. MicroRNA 199b-5p delivery through stable nucleic acid lipid par-
H.P., Zimmermann, T.S., Langer, R., Anderson, D.G., 2008. A combinatorial library of ticles (SNALPs) in tumorigenic cell lines. Naunyn Schmiedebergs Arch. Pharmacol.
lipid-like materials for delivery of RNAi therapeutics. Nat. Biotechnol. 26, 561–569. 386, 287–302.
Ando, H., Okamoto, A., Yokota, M., Shimizu, K., Asai, T., Dewa, T., Oku, N., 2013. Deshpande, P.P., Biswas, S., Torchilin, V.P., 2013. Current trends in the use of liposomes
Development of a miR-92a delivery system for anti-angiogenesis-based cancer for tumor targeting. Nanomedicine (London, England) 8, 1509–1528.
therapy. J. Gene Med. 15, 20–27. Di Martino, M.T., Campani, V., Misso, G., Gallo Cantafio, M.E., Gulla, A., Foresta, U.,
Babu, A., Munshi, A., Ramesh, R., 2017. Combinatorial therapeutic approaches with RNAi Guzzi, P.H., Castellano, M., Grimaldi, A., Gigantino, V., Franco, R., Lusa, S.,
and anticancer drugs using nanodrug delivery systems. Drug Dev. Ind. Pharm. 43, Cannataro, M., Tagliaferri, P., De Rosa, G., Tassone, P., Caraglia, M., 2014. In vivo
1391–1401. activity of miR-34a mimics delivered by stable nucleic acid lipid particles (SNALPs)
Bader, A.G., 2012. miR-34 - a microRNA replacement therapy is headed to the clinic. against multiple myeloma. PLoS One 9, e90005.
Front. Genet. 3, 120. Dorrance, A.M., Neviani, P., Ferenchak, G.J., Huang, X., Nicolet, D., Maharry, K.S., Ozer,
Balazs, D.A., Godbey, W., 2011. Liposomes for use in gene delivery. J. Drug Deliv. 2011, H.G., Hoellarbauer, P., Khalife, J., Hill, E.B., Yadav, M., Bolon, B.N., Lee, R.J., Lee,
326497. L.J., Croce, C.M., Garzon, R., Caligiuri, M.A., Bloomfield, C.D., Marcucci, G., 2015.
Barenholz, Y., 2001. Liposome application: problems and prospects. Curr. Opin. Colloid Targeting leukemia stem cells in vivo with antagomiR-126 nanoparticles in acute
Interface Sci. 6, 66–77. myeloid leukemia. Leukemia 29, 2143–2153.
Baumann, V., Winkler, J., 2014. miRNA-based therapies: strategies and delivery plat- Eifler, A.C., Thaxton, C.S., 2011. Nanoparticle therapeutics: FDA approval, clinical trials,
forms for oligonucleotide and non-oligonucleotide agents. Future Med. Chem. 6, regulatory pathways, and case study. Methods Mol. Biol. 726, 325–338.
1967–1984. Endo-Takahashi, Y., Negishi, Y., Nakamura, A., Ukai, S., Ooaku, K., Oda, Y., Sugimoto, K.,
Bazak, R., Houri, M., Achy, S.E., Hussein, W., Refaat, T., 2014. Passive targeting of na- Moriyasu, F., Takagi, N., Suzuki, R., Maruyama, K., Aramaki, Y., 2014. Systemic
noparticles to cancer: a comprehensive review of the literature. Mol. Clin. Oncol. 2, delivery of miR-126 by miRNA-loaded Bubble liposomes for the treatment of hin-
904–908. dlimb ischemia. Sci. Rep. 4, 3883.
Beg, M.S., Brenner, A.J., Sachdev, J., Borad, M., Kang, Y.K., Stoudemire, J., Smith, S., Endo-Takahashi, Y., Negishi, Y., Suzuki, R., Maruyama, K., Aramaki, Y., 2016. MicroRNA
Bader, A.G., Kim, S., Hong, D.S., 2017. Phase I study of MRX34, a liposomal miR-34a imaging in combination with diagnostic ultrasound and bubble liposomes for
mimic, administered twice weekly in patients with advanced solid tumors. Invest. MicroRNA delivery. Methods Mol. Biol. 1372, 209–213.
New Drugs 35, 180–188. Fang, Y., Xue, J., Gao, S., Lu, A., Yang, D., Jiang, H., He, Y., Shi, K., 2017. Cleavable
Belfiore, L., Saunders, D.N., Ranson, M., Thurecht, K.J., Storm, G., Vine, K.L., 2018. PEGylation: a strategy for overcoming the "PEG dilemma" in efficient drug delivery.
Towards clinical translation of ligand-functionalized liposomes in targeted cancer Drug Deliv. 24, 22–32.
therapy: challenges and opportunities. J. Control. Release 277, 1–13. Feinberg, M.W., Moore, K.J., 2016. MicroRNA regulation of atherosclerosis. Circ. Res.
Belliveau, N.M., Huft, J., Lin, P.J., Chen, S., Leung, A.K., Leaver, T.J., Wild, A.W., Lee, 118, 703–720.
J.B., Taylor, R.J., Tam, Y.K., Hansen, C.L., Cullis, P.R., 2012. Microfluidic synthesis of Felgner, P.L., Gadek, T.R., Holm, M., Roman, R., Chan, H.W., Wenz, M., Northrop, J.P.,
highly potent limit-size lipid nanoparticles for in vivo delivery of siRNA. Mol. Ther. Ringold, G.M., Danielsen, M., 1987. Lipofection: a highly efficient, lipid-mediated

16
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

DNA-transfection procedure. Proc. Natl. Acad. Sci. U. S. A. 84, 7413–7417. nanoparticles. J. Control. Release 161, 600–608.
Filion, M.C., Phillips, N.C., 1997. Toxicity and immunomodulatory activity of liposomal Lee, H.Y., Mohammed, K.A., Kaye, F., Sharma, P., Moudgil, B.M., Clapp, W.L., Nasreen,
vectors formulated with cationic lipids toward immune effector cells. Biochim. N., 2013. Targeted delivery of let-7a microRNA encapsulated ephrin-A1 conjugated
Biophys. Acta 1329, 345–356. liposomal nanoparticles inhibit tumor growth in lung cancer. Int. J. Nanomedicine 8,
Friedman, R.C., Farh, K.K., Burge, C.B., Bartel, D.P., 2009. Most mammalian mRNAs are 4481–4494.
conserved targets of microRNAs. Genome Res. 19, 92–105. Leung, A.K., Hafez, I.M., Baoukina, S., Belliveau, N.M., Zhigaltsev, I.V., Afshinmanesh, E.,
Gao, X., Huang, L., 1991. A novel cationic liposome reagent for efficient transfection of Tieleman, D.P., Hansen, C.L., Hope, M.J., Cullis, P.R., 2012. Lipid nanoparticles
mammalian cells. Biochem. Biophys. Res. Commun. 179, 280–285. containing siRNA synthesized by microfluidic mixing exhibit an electron-dense na-
Garbuzenko, O.B., Saad, M., Betigeri, S., Zhang, M., Vetcher, A.A., Soldatenkov, V.A., nostructured core. J. Phys. Chem. C Nanomater. Interfaces 116, 18440–18450.
Reimer, D.C., Pozharov, V.P., Minko, T., 2009. Intratracheal versus intravenous li- Leung, A.K., Tam, Y.Y., Cullis, P.R., 2014. Lipid nanoparticles for short interfering RNA
posomal delivery of siRNA, antisense oligonucleotides and anticancer drug. Pharm. delivery. Adv. Genet. 88, 71–110.
Res. 26, 382–394. Levchenko, T.S., Rammohan, R., Volodina, N., Torchilin, V.P., 2003. Tat peptide-medi-
Hafez, I.M., Cullis, P.R., 2001. Roles of lipid polymorphism in intracellular delivery. Adv. ated intracellular delivery of liposomes. Meth. Enzymol. 372, 339–349.
Drug Deliv. Rev. 47, 139–148. Leventis, R., Silvius, J.R., 1990. Interactions of mammalian cells with lipid dispersions
Hafez, I.M., Maurer, N., Cullis, P.R., 2001. On the mechanism whereby cationic lipids containing novel metabolizable cationic amphiphiles. Biochim. Biophys. Acta 1023,
promote intracellular delivery of polynucleic acids. Gene Ther. 8, 1188–1196. 124–132.
Hatakeyama, H., Murata, M., Sato, Y., Takahashi, M., Minakawa, N., Matsuda, A., Li, Z., Rana, T.M., 2014. Therapeutic targeting of microRNAs: current status and future
Harashima, H., 2013. The systemic administration of an anti-miRNA oligonucleotide challenges. Nat. Rev. Drug Discov. 13, 622–638.
encapsulated pH-sensitive liposome results in reduced level of hepatic microRNA-122 Liu, M., Li, M., Sun, S., Li, B., Du, D., Sun, J., Cao, F., Li, H., Jia, F., Wang, T., Chang, N.,
in mice. J. Control. Release. Yu, H., Wang, Q., Peng, H., 2014. The use of antibody modified liposomes loaded
Hosseinahli, N., Aghapour, M., Duijf, P.H.G., Baradaran, B., 2018. Treating cancer with with AMO-1 to deliver oligonucleotides to ischemic myocardium for arrhythmia
microRNA replacement therapy: a literature review. J. Cell. Physiol. 233, 5574–5588. therapy. Biomaterials 35, 3697–3707.
Hou, K.K., Pan, H., Schlesinger, P.H., Wickline, S.A., 2015. A role for peptides in over- Liu, X.Q., Song, W.J., Sun, T.M., Zhang, P.Z., Wang, J., 2011. Targeted delivery of anti-
coming endosomal entrapment in siRNA delivery - A focus on melittin. Biotechnol. sense inhibitor of miRNA for antiangiogenesis therapy using cRGD-functionalized
Adv. 33, 931–940. nanoparticles. Mol. Pharm. 8, 250–259.
Howard, M., Zern, B.J., Anselmo, A.C., Shuvaev, V.V., Mitragotri, S., Muzykantov, V., Lonez, C., Vandenbranden, M., Ruysschaert, J.M., 2012. Cationic lipids activate in-
2014. Vascular targeting of nanocarriers: perplexing aspects of the seemingly tracellular signaling pathways. Adv. Drug Deliv. Rev. 64, 1749–1758.
straightforward paradigm. ACS Nano 8, 4100–4132. Love, K.T., Mahon, K.P., Levins, C.G., Whitehead, K.A., Querbes, W., Dorkin, J.R., Qin, J.,
Hsu, S.H., Yu, B., Wang, X., Lu, Y., Schmidt, C.R., Lee, R.J., Lee, L.J., Jacob, S.T., Ghoshal, Cantley, W., Qin, L.L., Racie, T., Frank-Kamenetsky, M., Yip, K.N., Alvarez, R., Sah,
K., 2013. Cationic lipid nanoparticles for therapeutic delivery of siRNA and miRNA to D.W., de Fougerolles, A., Fitzgerald, K., Koteliansky, V., Akinc, A., Langer, R.,
murine liver tumor. Nanomedicine 9, 1169–1180. Anderson, D.G., 2010. Lipid-like materials for low-dose, in vivo gene silencing. Proc.
Huang, X., Schwind, S., Santhanam, R., Eisfeld, A.K., Chiang, C.L., Lankenau, M., Yu, B., Natl. Acad. Sci. U. S. A. 107, 1864–1869.
Hoellerbauer, P., Jin, Y., Tarighat, S.S., Khalife, J., Walker, A., Perrotti, D., Maeki, M., Kimura, N., Sato, Y., Harashima, H., Tokeshi, M., 2018. Advances in micro-
Bloomfield, C.D., Wang, H., Lee, R.J., Lee, L.J., Marcucci, G., 2016. Targeting the fluidics for lipid nanoparticles and extracellular vesicles and applications in drug
RAS/MAPK pathway with miR-181a in acute myeloid leukemia. Oncotarget 7, delivery systems. Adv. Drug Deliv. Rev. 128, 84–100.
59273–59286. Marsh, D., Bartucci, R., Sportelli, L., 2003. Lipid membranes with grafted polymers:
Huang, X., Schwind, S., Yu, B., Santhanam, R., Wang, H., Hoellerbauer, P., Mims, A., physicochemical aspects. Biochim. Biophys. Acta 1615, 33–59.
Klisovic, R., Walker, A.R., Chan, K.K., Blum, W., Perrotti, D., Byrd, J.C., Bloomfield, Maurer, N., Wong, K.F., Stark, H., Louie, L., McIntosh, D., Wong, T., Scherrer, P., Semple,
C.D., Caligiuri, M.A., Lee, R.J., Garzon, R., Muthusamy, N., Lee, L.J., Marcucci, G., S.C., Cullis, P.R., 2001. Spontaneous entrapment of polynucleotides upon electro-
2013. Targeted delivery of microRNA-29b by transferrin-conjugated anionic lipopo- static interaction with ethanol-destabilized cationic liposomes. Biophys. J. 80,
lyplex nanoparticles: a novel therapeutic strategy in acute myeloid leukemia. Clin. 2310–2326.
Cancer Res. 19, 2355–2367. Miao, L., Huang, L., 2015. Exploring the tumor microenvironment with nanoparticles.
Iorio, M.V., Croce, C.M., 2012. MicroRNA dysregulation in cancer: diagnostics, mon- Cancer Treat. Res. 166, 193–226.
itoring and therapeutics. A comprehensive review. EMBO Mol. Med. 4, 143–159. Miller, C.R., Bondurant, B., McLean, S.D., McGovern, K.A., O’Brien, D.F., 1998. Liposome-
Islam, M.A., Reesor, E.K., Xu, Y., Zope, H.R., Zetter, B.R., Shi, J., 2015. Biomaterials for cell interactions in vitro: effect of liposome surface charge on the binding and en-
mRNA delivery. Biomater. Sci. 3, 1519–1533. docytosis of conventional and sterically stabilized liposomes. Biochemistry 37,
Israelachvili, J.N., Mitchell, D.J., 1975. A model for the packing of lipids in bilayer 12875–12883.
membranes. Biochim. Biophys. Acta 389, 13–19. Moghassemi, S., Hadjizadeh, A., 2014. Nano-niosomes as nanoscale drug delivery sys-
Jin, Y., Liu, S., Yu, B., Golan, S., Koh, C.G., Yang, J., Huynh, L., Yang, X., Pang, J., tems: an illustrated review. J. Control. Release 185, 22–36.
Muthusamy, N., Chan, K.K., Byrd, J.C., Talmon, Y., Lee, L.J., Lee, R.J., Marcucci, G., Mullard, A., 2018. FDA approves landmark RNAi drug. Nat. Rev. Drug Discov. 17, 613.
2010. Targeted delivery of antisense oligodeoxynucleotide by transferrin conjugated Nishimura, M., Jung, E.J., Shah, M.Y., Lu, C., Spizzo, R., Shimizu, M., Han, H.D., Ivan, C.,
pH-sensitive lipopolyplex nanoparticles: a novel oligonucleotide-based therapeutic Rossi, S., Zhang, X., Nicoloso, M.S., Wu, S.Y., Almeida, M.I., Bottsford-Miller, J.,
strategy in acute myeloid leukemia. Mol. Pharm. 7, 196–206. Pecot, C.V., Zand, B., Matsuo, K., Shahzad, M.M., Jennings, N.B., Rodriguez-Aguayo,
Joshi, H.P., Subramanian, I.V., Schnettler, E.K., Ghosh, G., Rupaimoole, R., Evans, C., C., Lopez-Berestein, G., Sood, A.K., Calin, G.A., 2013. Therapeutic synergy between
Saluja, M., Jing, Y., Cristina, I., Roy, S., Zeng, Y., Shah, V.H., Sood, A.K., microRNA and siRNA in ovarian cancer treatment. Cancer Discov. 3, 1302–1315.
Ramakrishnan, S., 2014. Dynamin 2 along with microRNA-199a reciprocally regulate Nobs, L., Buchegger, F., Gurny, R., Allemann, E., 2004. Current methods for attaching
hypoxia-inducible factors and ovarian cancer metastasis. Proc. Natl. Acad. Sci. U. S. targeting ligands to liposomes and nanoparticles. J. Pharm. Sci. 93, 1980–1992.
A. 111, 5331–5336. Obeid, M.A., Elburi, A., Young, L.C., Mullen, A.B., Tate, R.J., Ferro, V.A., 2017.
Jubeli, E., Moine, L., Vergnaud-Gauduchon, J., Barratt, G., 2012. E-selectin as a target for Formulation of nonionic surfactant vesicles (NISV) prepared by microfluidics for
drug delivery and molecular imaging. J. Control. Release 158, 194–206. therapeutic delivery of siRNA into Cancer cells. Mol. Pharm. 14, 2450–2458.
Kale, A.A., Torchilin, V.P., 2007. Design, synthesis, and characterization of pH-sensitive Paliwal, S.R., Paliwal, R., Vyas, S.P., 2015. A review of mechanistic insight and appli-
PEG-PE conjugates for stimuli-sensitive pharmaceutical nanocarriers: the effect of cation of pH-sensitive liposomes in drug delivery. Drug Deliv. 22, 231–242.
substitutes at the hydrazone linkage on the ph stability of PEG-PE conjugates. Patil, Y.P., Jadhav, S., 2014. Novel methods for liposome preparation. Chem. Phys. Lipids
Bioconjug. Chem. 18, 363–370. 177, 8–18.
Kanasty, R., Dorkin, J.R., Vegas, A., Anderson, D., 2013. Delivery materials for siRNA Pattni, B.S., Chupin, V.V., Torchilin, V.P., 2015. New developments in liposomal drug
therapeutics. Nat. Mater. 12, 967–977. delivery. Chem. Rev. 115, 10938–10966.
Kessner, S., Krause, A., Rothe, U., Bendas, G., 2001. Investigation of the cellular uptake of Pecot, C.V., Calin, G.A., Coleman, R.L., Lopez-Berestein, G., Sood, A.K., 2011. RNA in-
E-Selectin-targeted immunoliposomes by activated human endothelial cells. Biochim. terference in the clinic: challenges and future directions. Nat. Rev. Cancer 11, 59–67.
Biophys. Acta 1514, 177–190. Pecot, C.V., Rupaimoole, R., Yang, D., Akbani, R., Ivan, C., Lu, C., Wu, S., Han, H.D.,
Kheirolomoom, A., Kim, C.W., Seo, J.W., Kumar, S., Son, D.J., Gagnon, M.K., Ingham, Shah, M.Y., Rodriguez-Aguayo, C., Bottsford-Miller, J., Liu, Y., Kim, S.B., Unruh, A.,
E.S., Ferrara, K.W., Jo, H., 2015. Multifunctional nanoparticles facilitate molecular Gonzalez-Villasana, V., Huang, L., Zand, B., Moreno-Smith, M., Mangala, L.S., Taylor,
targeting and miRNA delivery to inhibit atherosclerosis in ApoE(-/-) mice. ACS Nano M., Dalton, H.J., Sehgal, V., Wen, Y., Kang, Y., Baggerly, K.A., Lee, J.S., Ram, P.T.,
9, 8885–8897. Ravoori, M.K., Kundra, V., Zhang, X., Ali-Fehmi, R., Gonzalez-Angulo, A.M., Massion,
Kibria, G., Hatakeyama, H., Ohga, N., Hida, K., Harashima, H., 2011. Dual-ligand mod- P.P., Calin, G.A., Lopez-Berestein, G., Zhang, W., Sood, A.K., 2013. Tumour angio-
ification of PEGylated liposomes shows better cell selectivity and efficient gene de- genesis regulation by the miR-200 family. Nat. Commun. 4, 2427.
livery. J. Control. Release 153, 141–148. Pérez, S.E., Carlucci, A.M., 2018. Lipid-based siRNA nanodelivery systems: a learning
Kim, H., Leal, C., 2015. Cuboplexes: topologically active siRNA delivery. ACS Nano 9, process for improving transfer from concepts to clinical applications. Curr. Clin.
10214–10226. Pharmacol. 13, 142–163.
Kulkarni, J.A., Cullis, P.R., van der Meel, R., 2018. Lipid nanoparticles enabling gene Petrilli, R., Eloy, J.O., Marchetti, J.M., Lopez, R.F., Lee, R.J., 2014. Targeted lipid na-
therapies: from concepts to clinical utility. Nucleic Acid Ther. 28, 146–157. noparticles for antisense oligonucleotide delivery. Curr. Pharm. Biotechnol. 15,
Lammers, T., Kiessling, F., Ashford, M., Hennink, W., Crommelin, D., Storm, G., 2016. 847–855.
Cancer nanomedicine: Is targeting our target? Nat. Rev. Mater. 1, 16069. Piao, L., Zhang, M., Datta, J., Xie, X., Su, T., Li, H., Teknos, T.N., Pan, Q., 2012. Lipid-
Landen Jr, C.N., Chavez-Reyes, A., Bucana, C., Schmandt, R., Deavers, M.T., Lopez- based nanoparticle delivery of Pre-miR-107 inhibits the tumorigenicity of head and
Berestein, G., Sood, A.K., 2005. Therapeutic EphA2 gene targeting in vivo using neck squamous cell carcinoma. Mol. Ther. 20, 1261–1269.
neutral liposomal small interfering RNA delivery. Cancer Res. 65, 6910–6918. Ragelle, H., Danhier, F., Preat, V., Langer, R., Anderson, D.G., 2017. Nanoparticle-based
Landesman-Milo, D., Peer, D., 2012. Altering the immune response with lipid-based drug delivery systems: a commercial and regulatory outlook as the field matures.

17
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

Expert Opin. Drug Deliv. 14, 851–864. 623–629.


Reddy, T.L., Garikapati, K.R., Reddy, S.G., Reddy, B.V., Yadav, J.S., Bhadra, U., Bhadra, Vermeulen, L.M.P., Brans, T., Samal, S.K., Dubruel, P., Demeester, J., De Smedt, S.C.,
M.P., 2016. Simultaneous delivery of Paclitaxel and Bcl-2 siRNA via pH-Sensitive Remaut, K., Braeckmans, K., 2018a. Endosomal Size and Membrane Leakiness
liposomal nanocarrier for the synergistic treatment of melanoma. Sci. Rep. 6, 35223. Influence Proton Sponge-Based Rupture of Endosomal Vesicles. ACS Nano 12,
Reimer, D.L., Zhang, Y., Kong, S., Wheeler, J.J., Graham, R.W., Bally, M.B., 1995. 2332–2345.
Formation of novel hydrophobic complexes between cationic lipids and plasmid Vermeulen, L.M.P., De Smedt, S.C., Remaut, K., Braeckmans, K., 2018b. The proton
DNA. Biochemistry 34, 12877–12883. sponge hypothesis: Fable or fact? Eur. J. Pharm. Biopharm. 129, 184–190.
Rothschild, S.I., 2014. microRNA therapies in cancer. Mol. Cell. Ther. 2, 7. Viger-Gravel, J., Schantz, A., Pinon, A.C., Rossini, A.J., Schantz, S., Emsley, L., 2018.
Rupaimoole, R., Calin, G.A., Lopez-Berestein, G., Sood, A.K., 2016a. miRNA deregulation Structure of lipid nanoparticles containing siRNA or mRNA by dynamic nuclear po-
in Cancer cells and the tumor microenvironment. Cancer Discov. 6, 235–246. larization-enhanced NMR spectroscopy. J. Phys. Chem. B 122, 2073–2081.
Rupaimoole, R., Ivan, C., Yang, D., Gharpure, K.M., Wu, S.Y., Pecot, C.V., Previs, R.A., Wan, C., Allen, T.M., Cullis, P.R., 2014. Lipid nanoparticle delivery systems for siRNA-
Nagaraja, A.S., Armaiz-Pena, G.N., McGuire, M., Pradeep, S., Mangala, L.S., based therapeutics. Drug Deliv. Transl. Res. 4, 74–83.
Rodriguez-Aguayo, C., Huang, L., Bar-Eli, M., Zhang, W., Lopez-Berestein, G., Calin, Wang, H., Jiang, Y., Peng, H., Chen, Y., Zhu, P., Huang, Y., 2015. Recent progress in
G.A., Sood, A.K., 2016b. Hypoxia-upregulated microRNA-630 targets Dicer, leading microRNA delivery for cancer therapy by non-viral synthetic vectors. Adv. Drug
to increased tumor progression. Oncogene 35, 4312–4320. Deliv. Rev. 81, 142–160.
Rupaimoole, R., Slack, F.J., 2017. MicroRNA therapeutics: towards a new era for the Wang, K., Kievit, F.M., Zhang, M., 2016. Nanoparticles for cancer gene therapy: recent
management of cancer and other diseases. Nat. Rev. Drug Discov. advances, challenges, and strategies. Pharmacol. Res. 114, 56–66.
Sapra, P., Tyagi, P., Allen, T.M., 2005. Ligand-targeted liposomes for cancer treatment. Wang, S., Wang, H., Liu, Z., Wang, L., Wang, X., Su, L., Chang, J., 2014a. Smart pH- and
Curr. Drug Deliv. 2, 369–381. reduction-dual-responsive folate-PEG-coated polymeric lipid vesicles for tumor-trig-
Sawant, R.R., Torchilin, V.P., 2012. Challenges in development of targeted liposomal gered targeted drug delivery. Nanoscale 6, 7635–7642.
therapeutics. AAPS J. 14, 303–315. Wang, X., Huang, X., Yang, Z., Gallego-Perez, D., Ma, J., Zhao, X., Xie, J., Nakano, I., Lee,
Schroeder, A., Levins, C.G., Cortez, C., Langer, R., Anderson, D.G., 2010. Lipid-based L.J., 2014b. Targeted delivery of tumor suppressor microRNA-1 by transferrin-con-
nanotherapeutics for siRNA delivery. J. Intern. Med. 267, 9–21. jugated lipopolyplex nanoparticles to patient-derived glioblastoma stem cells. Curr.
Selbach, M., Schwanhausser, B., Thierfelder, N., Fang, Z., Khanin, R., Rajewsky, N., 2008. Pharm. Biotechnol. 15, 839–846.
Widespread changes in protein synthesis induced by microRNAs. Nature 455, 58–63. Wasungu, L., Hoekstra, D., 2006. Cationic lipids, lipoplexes and intracellular delivery of
Semple, S.C., Akinc, A., Chen, J.X., Sandhu, A.P., Mui, B.L., Cho, C.K., Sah, D.W.Y., genes. J. Control. Release 116, 255–264.
Stebbing, D., Crosley, E.J., Yaworski, E., Hafez, I.M., Dorkin, J.R., Qin, J., Lam, K., Weisman, S., Hirsch-Lerner, D., Barenholz, Y., Talmon, Y., 2004. Nanostructure of ca-
Rajeev, K.G., Wong, K.F., Jeffs, L.B., Nechev, L., Eisenhardt, M.L., Jayaraman, M., tionic lipid-oligonucleotide complexes. Biophys. J. 87, 609–614.
Kazem, M., Maier, M.A., Srinivasulu, M., Weinstein, M.J., Chen, Q.M., Alvarez, R., Whitehead, K.A., Dorkin, J.R., Vegas, A.J., Chang, P.H., Veiseh, O., Matthews, J., Fenton,
Barros, S.A., De, S., Klimuk, S.K., Borland, T., Kosovrasti, V., Cantley, W.L., Tam, Y.K., O.S., Zhang, Y., Olejnik, K.T., Yesilyurt, V., Chen, D., Barros, S., Klebanov, B.,
Manoharan, M., Ciufolini, M.A., Tracy, M.A., de Fougerolles, A., MacLachlan, I., Novobrantseva, T., Langer, R., Anderson, D.G., 2014. Degradable lipid nanoparticles
Cullis, P.R., Madden, T.D., Hope, M.J., 2010. Rational design of cationic lipids for with predictable in vivo siRNA delivery activity. Nat. Commun. 5, 4277.
siRNA delivery. Nat. Biotechnol. 28, 172–U118. Whitehead, K.A., Langer, R., Anderson, D.G., 2009. Knocking down barriers: advances in
Semple, S.C., Klimuk, S.K., Harasym, T.O., Dos Santos, N., Ansell, S.M., Wong, K.F., siRNA delivery. Nat. Rev. Drug Discov. 8, 129–138.
Maurer, N., Stark, H., Cullis, P.R., Hope, M.J., Scherrer, P., 2001. Efficient en- Wiggins, J.F., Ruffino, L., Kelnar, K., Omotola, M., Patrawala, L., Brown, D., Bader, A.G.,
capsulation of antisense oligonucleotides in lipid vesicles using ionizable aminolipids: 2010. Development of a lung cancer therapeutic based on the tumor suppressor
formation of novel small multilamellar vesicle structures. Biochim. Biophys. Acta microRNA-34. Cancer Res. 70, 5923–5930.
1510, 152–166. Wilner, S.E., Levy, M., 2016. Synthesis and characterization of aptamer-targeted SNALPs
Sercombe, L., Veerati, T., Moheimani, F., Wu, S.Y., Sood, A.K., Hua, S., 2015. Advances for the delivery of siRNA. Methods Mol. Biol. 1380, 211–224.
and challenges of liposome assisted drug delivery. Front. Pharmacol. 6. Wisse, E., Jacobs, F., Topal, B., Frederik, P., De Geest, B., 2008. The size of endothelial
Sharma, S., Rajendran, V., Kulshreshtha, R., Ghosh, P.C., 2017. Enhanced efficacy of anti- fenestrae in human liver sinusoids: implications for hepatocyte-directed gene
miR-191 delivery through stearylamine liposome formulation for the treatment of transfer. Gene Ther. 15, 1193–1199.
breast cancer cells. Int. J. Pharm. 530, 387–400. Wolfram, J., Zhu, M., Yang, Y., Shen, J., Gentile, E., Paolino, D., Fresta, M., Nie, G., Chen,
Shi, S., Han, L., Gong, T., Zhang, Z., Sun, X., 2013. Systemic delivery of microRNA-34a for C., Shen, H., Ferrari, M., Zhao, Y., 2015. Safety of nanoparticles in medicine. Curr.
cancer stem cell therapy. Angew. Chem. Int. Ed. Engl. 52, 3901–3905. Drug Targets 16, 1671–1681.
Shi, S.J., Zhong, Z.R., Liu, J., Zhang, Z.R., Sun, X., Gong, T., 2012. Solid lipid nano- Wolfrum, C., Shi, S., Jayaprakash, K.N., Jayaraman, M., Wang, G., Pandey, R.K., Rajeev,
particles loaded with anti-microRNA oligonucleotides (AMOs) for suppression of K.G., Nakayama, T., Charrise, K., Ndungo, E.M., Zimmermann, T., Koteliansky, V.,
microRNA-21 functions in human lung cancer cells. Pharm. Res. 29, 97–109. Manoharan, M., Stoffel, M., 2007. Mechanisms and optimization of in vivo delivery of
Siepi, E., Lutz, S., Meyer, S., Panzner, S., 2011. An ion switch regulates fusion of charged lipophilic siRNAs. Nat. Biotechnol. 25, 1149–1157.
membranes. Biophys. J. 100, 2412–2421. Won, Y.Y., Sharma, R., Konieczny, S.F., 2009. Missing pieces in understanding the in-
Singh, M.S., Peer, D., 2016. RNA nanomedicines: the next generation drugs? Curr. Opin. tracellular trafficking of polycation/DNA complexes. J. Control. Release 139, 88–93.
Biotechnol. 39, 28–34. Wu, Y., Crawford, M., Mao, Y., Lee, R.J., Davis, I.C., Elton, T.S., Lee, L.J., Nana-Sinkam,
Slaby, O., Laga, R., Sedlacek, O., 2017. Therapeutic targeting of non-coding RNAs in S.P., 2013. Therapeutic delivery of MicroRNA-29b by cationic lipoplexes for lung
cancer. Biochem. J. 474, 4219–4251. Cancer. Mol. Ther. Nucleic Acids 2, e84.
Stark, B., Andreae, F., Mosgoeller, W., Edetsberger, M., Gaubitzer, E., Koehler, G., Prassl, Wu, Y., Crawford, M., Yu, B., Mao, Y., Nana-Sinkam, S.P., Lee, L.J., 2011. MicroRNA
R., 2008. Liposomal vasoactive intestinal peptide for lung application: protection delivery by cationic lipoplexes for lung cancer therapy. Mol. Pharm. 8, 1381–1389.
from proteolytic degradation. Eur. J. Pharm. Biopharm. 70, 153–164. Xia, Y., Tian, J., Chen, X., 2016. Effect of surface properties on liposomal siRNA delivery.
Sui, L., Wang, M., Han, Q., Yu, L., Zhang, L., Zheng, L., Lian, J., Zhang, J., Valverde, P., Biomaterials 79, 56–68.
Xu, Q., Tu, Q., Chen, J., 2018. A novel Lipidoid-MicroRNA formulation promotes Xu, F., Liao, J.Z., Xiang, G.Y., Zhao, P.X., Ye, F., Zhao, Q., He, X.X., 2017. MiR-101 and
calvarial bone regeneration. Biomaterials 177, 88–97. doxorubicin codelivered by liposomes suppressing malignant properties of hepato-
Sun, M., Yang, C., Zheng, J., Wang, M., Chen, M., Le, D.Q.S., Kjems, J., Bunger, C.E., cellular carcinoma. Cancer Med. 6, 651–661.
2015. Enhanced efficacy of chemotherapy for breast cancer stem cells by simulta- Xue, H.Y., Guo, P., Wen, W.C., Wong, H.L., 2015. Lipid-based nanocarriers for RNA de-
neous suppression of multidrug resistance and antiapoptotic cellular defense. Acta livery. Curr. Pharm. Des. 21, 3140–3147.
Biomater. 28, 171–182. Xue, W., Dahlman, J.E., Tammela, T., Khan, O.F., Sood, S., Dave, A., Cai, W., Chirino,
Szebeni, J., Moghimi, S.M., 2009. Liposome triggering of innate immune responses: a L.M., Yang, G.R., Bronson, R., Crowley, D.G., Sahay, G., Schroeder, A., Langer, R.,
perspective on benefits and adverse reactions. J. Liposome Res. 19, 85–90. Anderson, D.G., Jacks, T., 2014. Small RNA combination therapy for lung cancer.
Tai, W., Gao, X., 2017. Functional peptides for siRNA delivery. Adv. Drug Deliv. Rev. 110- Proc. Natl. Acad. Sci. U. S. A. 111, E3553–3561.
111, 157–168. Yang, C., Gao, S., Song, P., Dagnaes-Hansen, F., Jakobsen, M., Kjems, J., 2018.
Takeda, Y.S., 2016. Synthetic bioreducible lipid‐based nanoparticles for miRNA delivery Theranostic niosomes for efficient siRNA/MicroRNA delivery and activatable near-
to mesenchymal stem cells to induce neuronal differentiation. Bioeng. Transl. Med. 1, infrared fluorescent tracking of stem cells. ACS Appl. Mater. Interfaces 10,
160–167. 19494–19503.
Tibbitt, M.W., Dahlman, J.E., Langer, R., 2016. Emerging frontiers in drug delivery. J. Yang, D., Sun, Y., Hu, L., Zheng, H., Ji, P., Pecot, C.V., Zhao, Y., Reynolds, S., Cheng, H.,
Am. Chem. Soc. 138, 704–717. Rupaimoole, R., Cogdell, D., Nykter, M., Broaddus, R., Rodriguez-Aguayo, C., Lopez-
Torchilin, V.P., 2005. Recent advances with liposomes as pharmaceutical carriers. Nat. Berestein, G., Liu, J., Shmulevich, I., Sood, A.K., Chen, K., Zhang, W., 2013.
Rev. Drug Discov. 4, 145–160. Integrated analyses identify a master microRNA regulatory network for the me-
Torchilin, V.P., 2007. Targeted pharmaceutical nanocarriers for cancer therapy and senchymal subtype in serous ovarian cancer. Cancer Cell 23, 186–199.
imaging. AAPS J. 9, E128–147. Yung, B.C., Li, J., Zhang, M., Cheng, X., Li, H., Yung, E.M., Kang, C., Cosby, L.E., Liu, Y.,
Trajkovski, M., Hausser, J., Soutschek, J., Bhat, B., Akin, A., Zavolan, M., Heim, M.H., Teng, L., Lee, R.J., 2016. Lipid nanoparticles composed of quaternary amine-tertiary
Stoffel, M., 2011. MicroRNAs 103 and 107 regulate insulin sensitivity. Nature 474, amine cationic lipid combination (QTsome) for therapeutic delivery of AntimiR-21
649–653. for lung Cancer. Mol. Pharm. 13, 653–662.
Trang, P., Wiggins, J.F., Daige, C.L., Cho, C., Omotola, M., Brown, D., Weidhaas, J.B., Zatsepin, T.S., Kotelevtsev, Y.V., Koteliansky, V., 2016. Lipid nanoparticles for targeted
Bader, A.G., Slack, F.J., 2011. Systemic delivery of tumor suppressor microRNA siRNA delivery - going from bench to bedside. Int. J. Nanomedicine 11, 3077–3086.
mimics using a neutral lipid emulsion inhibits lung tumors in mice. Mol. Ther. 19, Zhang, C., Tang, N., Liu, X., Liang, W., Xu, W., Torchilin, V.P., 2006. siRNA-containing
1116–1122. liposomes modified with polyarginine effectively silence the targeted gene. J.
Valencia, P.M., Farokhzad, O.C., Karnik, R., Langer, R., 2012. Microfluidic technologies Control. Release 112, 229–239.
for accelerating the clinical translation of nanoparticles. Nat. Nanotechnol. 7, Zhang, M., Zhou, X., Wang, B., Yung, B.C., Lee, L.J., Ghoshal, K., Lee, R.J., 2013a.

18
M. Scheideler, et al. Chemistry and Physics of Lipids 226 (2020) 104837

Lactosylated gramicidin-based lipid nanoparticles (Lac-GLN) for targeted delivery of 167–179.


anti-miR-155 to hepatocellular carcinoma. J. Control. Release 168, 251–261. Zuidam, N.J., Hirsch-Lerner, D., Margulies, S., Barenholz, Y., 1999. Lamellarity of ca-
Zhang, W., Peng, F., Zhou, T., Huang, Y., Zhang, L., Ye, P., Lu, M., Yang, G., Gai, Y., Yang, tionic liposomes and mode of preparation of lipoplexes affect transfection efficiency.
T., Ma, X., Xiang, G., 2015. Targeted delivery of chemically modified anti-miR-221 to Biochim. Biophys. Acta 1419, 207–220.
hepatocellular carcinoma with negatively charged liposomes. Int. J. Nanomedicine Zylberberg, C., Gaskill, K., Pasley, S., Matosevic, S., 2017. Engineering liposomal nano-
10, 4825–4836. particles for targeted gene therapy. Gene Ther. 24, 441.
Zhang, Y., Wang, Z., Gemeinhart, R.A., 2013b. Progress in microRNA delivery. J. Control. Zylberberg, C., Matosevic, S., 2016. Pharmaceutical liposomal drug delivery: a review of
Release 172, 962–974. new delivery systems and a look at the regulatory landscape. Drug Deliv. 23,
Zuhorn, I.S., Hoekstra, D., 2002. On the mechanism of cationic amphiphile-mediated 3319–3329.
transfection. To fuse or not to fuse: is that the question? J. Membr. Biol. 189,

19

You might also like