Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Review

Plant Exosome-like Nanovesicles: Emerging


Therapeutics and Drug Delivery Nanoplatforms
Haseeb Anwar Dad,1 Ting-Wei Gu,1 Ao-Qing Zhu,1 Lu-Qi Huang,2 and Li-Hua Peng1
1College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, P.R. China; 2National Resource Centre for Chinese Materia Medica, China Academy of
Chinese Medical Sciences, Beijing 100700, P.R. China

Plant exosome-like nanovesicles, being innately replete with production costs. Addressing these challenges, PELNVs represent
bioactive lipids, proteins, RNA, and other pharmacologically themselves as avant-garde innocuous and eco-friendly robust and
active molecules, offer unique morphological and composi- feasible carriers for nanomedicine. In their comparison, synthesized
tional characteristics as natural nanocarriers. Furthermore, nanoparticles are linked with complications such as immunogenicity,
their compelling physicochemical traits underpin their modu- cytotoxicity, and complex fabrication process requirements.8
lative role in physiological processes, all of which have fostered Contrarily, PELNVs, owing to their natural source and composition,
the concept that these nanovesicles may be highly proficient in enjoy the perks of being undetected by the immune system, and hence
the development of next-generation biotherapeutic and drug enhanced circulation periods and higher bioavailability. Moreover,
delivery nanoplatforms to meet the ever-stringent demands unlike mammals, plants do not harbor zoonotic or human pathogens.
of current clinical challenges. This review systemically deals Thus, PELNVs also take the edge over mammalian cell-derived
with various facets of plant exosome-like nanovesicles ranging ELNVs with their non-immunogenic and innocuous traits. This pre-
from their origin and isolation to identification of morpholog- cedence is also owing to their capability to showcase efficient uptake
ical composition, biological functions, and cargo-loading at the target site, high output of delivery of therapeutic agents, and
mechanisms. Efforts are made to encompass their bio- cost-efficient production.9 PELNVs lodged in the paramural space
therapeutic roles by elucidating their immunological modu- of plants are identical in morphology and functionality to their
lating, anti-tumor, regenerative, and anti-inflammatory roles. mammalian analog and are recognized for their importance.10 They
We also shed light on re-engineering these nanovesicles into not only contribute in plant cell crosstalk but also to inter-kingdom
robust, innocuous, and non-immunogenic nanovectors for communication.11 Although there are only limited numbers of
drug delivery through multiple stringent biological hindrances PELNV-related reviews, possibly because of only a handful of original
to various targeted organs such as intestine and brain. Finally, research articles in this field, some of which can be found else-
recent advances centered around plant exosome-like nanove- where,12–14 unfortunately, none of them has covered all aspects of
sicles along with new insights into transdermal, transmem- PELNVs. Herein, we have attempted to comprehensively review
brane and targeting mechanisms of these vesicles are also and consolidate the position of PELNVs as entities poised to become
elucidated. We expect that the continuing development of plant next-generation therapeutic and drug delivery nanoplatforms for
exosome-like nanovesicle-based therapeutic and delivery nano- desired clinical outcomes. The present review is also meant to give
platforms will promote their clinical applications. a quick glance to readers of all investigations directed toward PELNVs
and PELNV-derived nanovectors and their isolation techniques along
with identification of morphological composition. More importantly,
Exosome-like nanovesicles (ELNVs), which are biological nanostruc- we elucidate the translation of their innate biological functions into
tures of 40–150 nm, are secreted by most types of cells and relay in- diversified therapeutic actions, and we also emphasize the immense
formation between cells and organisms across all three kingdoms of potential of manipulating them as a safe and sustainable drug delivery
life.1,2 Although earlier perceived to be cellular debris and hence nanoplatform, while highlighting the limitations and research gaps
undervalued, ELNVs are now acknowledged as crucial entities to for enhancing delivery objectives.
regulate physiological functions of multicellular organisms in an
intercellular transmission manner.3 Despite a growing appreciation
of the importance of ELNVs in mammals, and the fact that plant Biogenesis and Physiological Actions of PELNVs
ELNVs (PELNVs) were observed first around 15 years earlier than Evidence of the existence of PELNVs has been being built since 1967.7
mammalian ELNVs,4,5 nothing much was known about PELNVs In earlier findings, the existence of multivesicular bodies (MVBs) in
beyond the old microscopy images suggesting their existence.6,7
https://doi.org/10.1016/j.ymthe.2020.11.030.
When aiming to bloom new therapeutic approaches, scientists must
Correspondence: Li-Hua Peng, PhD, College of Pharmaceutical Sciences, Zhejiang
surmount numerous challenges, including issues with delivery, tissue University, 866# Yuhangtang Road, Hangzhou 310058, P.R. China.
specificity, potential off-target effects, safety, toxicity, and large-scale E-mail: lhpeng@zju.edu.cn

Molecular Therapy Vol. 29 No 1 January 2021 ª 2020 The American Society of Gene and Cell Therapy. 13
www.moleculartherapy.org

Review

Figure 1. Biogenesis of PELNVs and Their Functions


in Plant Physiology
PELNVs are formed by inward budding of MVBs with the
content in similar orientation as in the plasmalemma, and
fusion of MVBs with plasmalemma permits the release of
PELNVs.

extensively fraught with cell wall remodeling


enzymes and defense proteins (Figure 1).24
Mounting evidence highlights their role as being
the front line of plant defenses in plant infections
and in the developmental processes.25–27 In
recent years, in-depth understanding of PELNVs
gave scientists an impetus to direct their investi-
gations toward exploring the interspecies or
inter-kingdom communication role of PELNVs
and to look for developmental space in therapeu-
tics and drug delivery.

Preparation, Re-engineering, and Cargo


Loading of PELNVs
The farthermost objective of the isolation tech-
nique is to formulate PELNV-based nanostruc-
tures to accomplish the ultimate clinical outcome
for curing ailments.28 There are two types of
nanoplatforms that can be used for therapeutics
and drug delivery purposes, with one being
PELNVs obtained in their pristine form and the
other being PELNV-derived nanovectors. Here-
in, we elaborate the preparation methods of
both kinds along with cargo loading techniques
currently being investigated to equip them for
drug delivery.

Preparation of PELNVs
The overlapped size range and rather indistin-
guishable morphological similarities necessitate
plant cells and their fusion with plasmalemma led to jettison ELNVs the current efforts in designing a feasible isolation method of
into the extracellular space in higher plants.6,7 In the ensuing years, ELNVs from the masses of the subpopulation of extracellular vesi-
similar morphology vesicular structure was found to be involved in cles (EVs). In this consideration, the differential ultracentrifugation
the processes of cell differentiation causing secondary wall thick- (UC) technique is commonly used and has acquired the benchmark
ness.15–17 The evolution of understanding the biogenesis and secre- status in isolation and purification of ELNVs owing to its ease of use
tion of PELNVs can be found in the literature.18–22 In short, PELNV and inexpensiveness.29–32 The technique mechanism operates on the
biogenesis is initiated with formation of the trans-Golgi network or size and density variations of particles.28 PELNV source material or
early endosome, which matures multivesicular endosomes (MVEs) plant juice is put through a series of centrifugations with gradually
or MVBs (Figure 1). The characteristic intraluminal vesicles (ILVs), enhancing speed. Each subsequent speedy centrifugation is sub-
which actively and selectively incorporate RNAs, including mRNAs, jected to longer duration than the previous one to sediment the
microRNAs (miRNAs), and other non-coding RNAs along with higher density particles, and then pellets obtained in each centrifu-
lipids and DNA, dwell inside MVBs, which are ultimately integrated gation are disposed. In the next step, previously acquired superna-
with plasmalemma to release PELNVs.23 PELNVs, being an integral tant is subjected to a further higher speed of 100,000  g to retrieve
constituent of a plant’s physiological machinery, execute several the pellet accommodating the PELNVs, which is subsequently re-
crucial roles within their own kingdom. For example, PELNVs are suspended and washed in a small amount of phosphate-buffered

14 Molecular Therapy Vol. 29 No 1 January 2021


www.moleculartherapy.org

Review

Figure 2. Isolation and Preparation of PELNVs


PELNVs could be isolated and prepared by series of centrifugations, including ultracentrifugation and sucrose gradient ultracentrifugation.

saline (PBS) (Figure 2).33 The PELNV yield obtained after the basic Re-engineering of PELNVs
procedure of differential UC is usually contaminated with nucleic It is a grueling task to prepare uniform-sized PELNVs because they
acids and protein agglomerates.34 Therefore, for further purification, vary in size from approximately 50 to 500 nm between and even
homogenized suspension is subjected to sucrose gradient ultracen- within species, which is major hindrance in their exploitation for
trifugation (SGC) (8%, 15%, 30%, 45%, and 60%) at an elevated drug delivery. Moreover, efficient drug loading is one major concern
speed greater than 150,000  g for 120 min. This step holds para- that is not readily achievable in purified PELNVs in pristine form.
mount importance in obtaining uncontaminated PELNVs. PELNVs Hence, it is indispensable to conceive a method to formulate uni-
float in the sucrose bands depending on the elevated g-force form-sized nanoparticles repeatedly with competent drug loading.41
sedimentation and various speeds of sucrose gradients.35 Mostly, For the fortification and re-engineering of PELNVs as drug delivery
sucrose gradients of the 30%/45% layer are harvested as the pre- nanoplatforms, researchers have successfully adopted the Bligh and
eminent source of PELNVs. To obtain the ultra-pure PELNV yield, Dyer technique (a well-reported liquid-liquid extraction method) to
disposal of the pellets after 100,000  g UC and use of an additional extract nanolipids from PELNVs, and extracted nanolipids are
UC at 135,000  g manage to get rid of other extraneous EVs. processed through a 200-nm liposome extruder, which ultimately
Although this benefits in achieving purity of PELNVs, it drops re-engineers them into a uniform-size nanoplatform termed
the PELNV yield concentration.36,37 “PELNV-derived nanovectors.”42

Repeated pelleting of PELNVs, under high centrifugal force of differ- Despite that PELNVs intrinsically express some lipids and cell adhe-
ential UCs, may result in compromising the PELNV structural integ- sion molecules such as phosphatidic acid (PA) that naturally promote
rity and cause agglomeration.38 This complication may be averted by binding to particular recipient cells,43 most targeting achievements of
incorporating a thin layer of high-density iso-osmotic material as a PELNVs are through its natural biodistribution. PELNV-derived
cushion at the bottom of the centrifuge tube, and as a result PELNVs nanovectors also offer the possibility of tailoring their surface, which
will never spin out as pellets at the bottom and will give enhanced can broaden the scope of desired target ability.44 In this regard, spe-
yield and quality.28,39 In addition to differential centrifugation, cific bioactive molecules can be immobilized onto the surface of
commercially available ELNV isolation kits are also utilized for PELNV-based nanoplatforms to develop displays more compatible
ELNV isolation by researchers. However, this technique has been to cellular surface receptors. For example, functionalizing the surface
reported to co-precipitate other EVs alongside ELNVs as well as of PELNV-based nanoplatforms more specifically to tumors can
RNA-protein complexes.40 assist in intensifying anti-tumor characteristics associated with the

Molecular Therapy Vol. 29 No 1 January 2021 15


www.moleculartherapy.org

Review

aforementioned nanoplatforms.45 By virtue of convenient surface compounds can lead toward their encapsulation by surmounting the
tailoring, ginger-derived ELNVs were functionalized with arrowtail surface-associated electrostatic forces of nanovesicles. Some findings
pRNA-3WJ and folic acid (FA) for ligand display and further used illustrated that siRNAs and DNAs, being strongly negatively charged
to systematically deliver genes to the tumor site.46 The most common molecules, could be laden into PELNVs and exhibited their desired
strategy for the surface functionalization of PELNV-based nanoplat- activity. However, the cargo loading efficiencies were found to be sub-
forms is the incubation method, which involves the mechanisms of par to those of positively charged molecules.58
hydrophobic interaction, the diffusion process, and electrostatic
interaction.47 In addition to this, some ligands that are immobilized Despite these substantial advantages, the peril of impairment of the
on the surface of PELNVs can act as tags for bio-imaging applica- innate structure of PELNVs during brief distortion for cargo loading
tions.48 It was reported that grapefruit-derived nanovectors were demands solution. As PELNVs are innately laden with some biomol-
labeled with lipophilic carbocyanine (DiR) dye that enabled their ecules, there might be the requirement of unloading some of the com-
in vivo tracking by fluorescence.9 Furthermore, click chemistry opens ponents of PELNVs. However, there has not been any attempt related
the door for bioconjugation of targeting macromolecules or small to unloading ELNVs. This proposition may help in augmenting the
molecules such as imaging agents to the surface of PELNVs via cova- ELNV-oriented delivery, lest ELNV integrity should remain intact
lent bonds.49 during unloading procedure. Moreover, massive challenge of success-
ful RNA loading onto PELNVs still persists, particularly in elevated
Careful and selective treatment of the surface of PELNVs can also levels for non-viral gene delivery. In this context, several studies
enhance the nucleic acid delivery capability of PELNVs. For instance, have demonstrated that physical electroporation can be exploited
the hybridization of polyethylenimine with grapefruit-derived nano- for cargo loading, in particular RNA loading onto ELNVs, and its
vectors not only enhanced gene delivery to brain tumor but also loading efficiency can be elevated by optimization of ELNVs and
diminished the cytotoxicity characteristic of polyethylenimine the siRNA ratio and voltage supplied during electroporation.59,60
considerably.9 Similarly, introduction of cell-penetrating peptide to However, electroporation fails to incorporate larger molecules such
the lipid bilayer structure of PELNVs has reportedly resulted in as mRNA and DNA with the same efficiency as for small RNAs.61
boosting the cellular uptake and tissue penetration.50 Moreover, to Besides this, other RNA loading techniques harnessing the biological
maximize the therapeutic functionalization of PELNV-based nano- process during ELNV formation can also be exploited such as passive
platforms, drug molecules can also be conjugated with their surface.46 loading in which augmented intracellular RNA load can be acquired
by transfecting the source cells with desired cargo via viral or non-
Cargo Loading Techniques viral nanovectors.62 These strategies have the potential to obviate
To achieve superior effects against disease, besides the endogenous the current procedural limitations associated with RNA loading
components, PELNVs could be laden with exogenous therapeutic onto PELNVs and will likely expand the horizons of gene delivery.
molecular drugs such as protein, expression vectors, siRNA, and
DNA.51,52 Different strategies have been devised to lade ELNVs with Characterization
therapeutic drug molecules (Figure 3). These methods first entail prep- Morphological Characterization
aration of PELNVs from plant and later loaded with drug molecules The flourishing advancements in ELNV-based nanotechnology de-
via direct manipulation of PELNVs. Mechanistically, for cargo loading mand precision and variety of characterization techniques to acquire
of ELNVs, passive and active cargo loading techniques are generally a more elaborative view of the morphological and physiochemical
used.53 The passive drug loading technique comprises an incubation traits of prepared PELNV-based nanoplatforms. It is important to
method, in which PELNVs are incubated with drug molecules at a devise and implement multiple characterization techniques to iden-
certain temperature. This drug encapsulation strategy is driven by tify the diverse subpopulations of EVs and is vital to the design of
diffusion action and lipophilic interaction between the drug molecule PELNV-based therapeutics or drug nanocarriers. Transmission
and lipid bilayer of PELNVs.54,55 In addition to passive loading, the electron microscopy (TEM) is used for ultrastructure analysis of the
sonication technique, an active cargo loading method, is used, which subcellular status of PELNVs.63,64 As an illustration, multifarious
distorts the ELNV membrane structure briefly for successful diffusion ELNVs derived from grapefruit, ginger, carrots, and tomatoes have
of cargo into ELNVs. The membrane structure of ENLVs reverts to its been reported.65 In addition, atomic force microscopy (AFM) can
intact integrity after the cargo loading. It also has been published that also be used to develop the structural and size-related understanding
this method takes precedence by boosting the cargo loading capability of individual PELNVs. AFM asserts its superiority to typical optical
up to 11-fold over passive cargo loading.56 In most conditions, microscopy by ascertaining resolution of fractions of nanome-
PELNVs retain a negatively charged surface that can attract oppositely ters.33,66,67 Size determination and zeta potential evaluation of
charged drugs such as doxorubicin and ultimately support the loading dispersed PELNVs can be done by dynamic light scattering (DLS),
of drug into their inner cavity by the sonication technique.57 also termed quasi-elastic light scattering, photon correlation spectros-
Conversely, it also has been reported that negatively charged com- copy.65 Being non-intrusive and ultra-sensitive, DLS needs only little
pounds such as FA and neutrally surfaced drugs such as curcumin sample volume to compute accurate and precise size, which makes
can also be ensnared in PELNVs despite modifying their biological it the benchmark technique in evaluation of size distribution of sus-
activities. Reported data suggested that lipophilic properties of these pension particulates in multiple scientific disciplines.68–70 In addition

16 Molecular Therapy Vol. 29 No 1 January 2021


www.moleculartherapy.org

Review

Figure 3. Re-engineering Method for PELNVs and Drug Loading Procedure


The Bligh and Dyer extraction method is used for nanolipid extraction from PELNVs to re-engineer them into nanovectors. The drug loading procedure comprises the
sonication technique for drug loading onto re-engineered nanovectors and an incubation method for drug loading onto pristine PELNVs.

to this, nanoparticle tracking analysis (NTA) is also gaining popu- trophoresis analyzer, and high-throughput small RNA sequencing
larity among researchers to quantify the number of ELNVs in a sam- have been established for deep analysis of the components of
ple volume and determine their size distribution. PELNVs.76,77 In particular, colorimetric assays such as bicincho-
ninic acid (BCA), fluorimetric assays, SDS-PAGE, and western blot-
Biochemical Characterization ting are popular for protein analysis.78 Another highly sophisticated
The chemical composition profiles of PELNVs maintain significant molecular characterization technique is Raman spectroscopy, which
discrepancies from those of mammalian-derived ELNVs in terms displays the chemical structure of PELNVs by developing a certain
of their protein, lipid, and RNA content.65 Compositional analyses spectrum of biomolecules such as peptides and nucleic acids
of lipids, nucleic acids, and proteins of PELNVs are appreciated residing in them.79 Moreover, microarray analysis, digital droplet
as important characterization criteria for the quality control of PCR, and next-generation sequencing techniques have been estab-
PELNVs.71,72 Plant and mammalian cell-derived ELNVs share lished for the RNA content analysis of PELNVs.80 For the lipidomic
mutual common techniques used for chemical component charac- analysis of PELNVs, the techniques most often used are a sulfo-
terization. For confirmation of the origin of ELNVs, immunoblot- phospho-vanillin assay and total reflection Fourier transform
ting of specific proteins is the most widely practised approach. infrared spectroscopy.81,82 Additionally, fluorescence measurement
With the advancements in technology, ELISA, sodium dodecyl of lipophilic fluorescent dyes such as DiR can also correspond to
sulfate (SDS)-polyacrylamide gel electrophoresis (PAGE),73 liquid lipid quantification, as those dyes will only give fluorescence when
chromatography-tandem massspectroscopy,74,75 a microfluidic elec- integrated into the lipid bilayer of PELNVs.83

Molecular Therapy Vol. 29 No 1 January 2021 17


www.moleculartherapy.org

Review

PELNVs generally have three major components: lipids, proteins, and mRNAs. However, naked miRNAs are unstable and prone to subjec-
nucleic acids.84 tion to immediate decomposition by ubiquitous RNases, and their ul-
timate packaging inside ELNVs circumvents their early obliteration
Identification of Lipids by RNases.63,99,100 Furthermore, a dietary plant’s characteristics along
Lipids are essential components of lipid bilayer structures of with nutrigenomic influences may redesign the miRNA constitution
PELNVs.85 Lipid profiling of PELNVs has revealed two distinct group and other components of PELNVs.101 Arabidopsis thaliana-derived
of lipids, phospholipids and glycerol lipids, which have been found to ELNVs are found to be laden with miRNAs, sRNAs (carrying 18–
be vital for PELNV stability, uptake, and other biological functions.86 24 nt), and tiny RNAs (tyRNAs) (RNA with 10–17 nt). Co-existence
Many PELNVs harbor PA, which modulates membrane fission and of the aforementioned RNAs and cargo in the apoplast indicates
fusion, being the dominant lipid mediator. It can also influence specific sorting mechanisms for RNA loading and then use for
PELNV uptake, as it triggers cytoskeleton rearrangement and protein delivery to distant sites.102 In addition, PELNVs often contain un-
regulation in vesicular endocytosis.43 Besides PA, PELNVs also amass usual noncoding RNAs, although it is not completely understood
diverse lipid content such as phosphatidylethanolamine (PE), phos- how ELNVs process them and what is their influence on uptake of
phatidylcholine (PC), phosphatidylinositol (PI), and diacylglycerol recipient cells and consequently in cell communication, which will
(DG). It is noteworthy that, among the aforementioned lipids, PC require further investigations to be elucidated.
has been suggested to have the capability to vector PELNVs from
the intestine to the liver in an augmented manner. It is also note- PELNVs as Nano-Therapeutics for Diseases
worthy that all PELNVs investigated so far were found to be devoid Biological functions of the PELNVs, in their natural pristine struc-
of any cholesterol content.51,87,88 Since several studies suggest that tural integrity with intact bioactive cargoes after mere isolation
PELNV lipids play a biologically active role in recipient cells, a thor- from plants, eventuate in mitigating pathological conditions in other
ough understanding of each lipid component that how it modulates kingdom’s species and provide multiple therapeutic alternatives (Fig-
its actions will be beneficial in the development of effective ure 4). The attributes that make PELNVs fascinating for therapy are
PELNV-based therapeutic strategies. their plant provenance and enhanced safety profile along with their
versatile therapeutic potential rooted in their active source plants.65
Identification of Proteins
According to the International Society for Extracellular Vesicles, sci- Immunological Modulation Effects
entists are encouraged to analyze vesicles with respect to proteins, Currently, PELNVs have revealed their salubrious immunological
which are found to be abundant or deficient in ELNVs, as proteins regulating actions on the gastrointestinal (GI) haemostasis.52 It is
are integral to their functionality.89 PELNV proteins can be mainly widely recognized that inflammation is a part of the innate immunity
categorized into transmembrane proteins and other plasmalemma- mechanism. If left uncontrolled, it can evolve into acute or chronic in-
associated proteins. Currently among transmembrane proteins, flammatory diseases and sometimes also turn into the deep-rooted
CD63, CD81, and CD9 have been identified as possible markers of cause of most chronic ailments.103 PELNVs have been found to exert
mammalian cell-derived ELNVs.90–92 However, markers of PELNVs their anti-inflammatory effect by regulating the immunological
have not been clearly reported yet and demand further investigations. actions of hosts. Moreover, miRNAs of PELNVs regulate the inter-
It was reported that protein contents of ginger-derived ELNVs kingdom communication between gut microbiota and the host
happen to be predominantly cytosolic, and fewer membrane trans- immune system, which is translated into the homeostatic balance
porter proteins were identified, such as aquaporins and chloride between immunity and gut microbiota (Figure 5). In fact, after inter-
channels.87 In addition, some reports have also suggested that nalization into the host’s recipient cells, PELNVs trigger a plethora of
grapes,86 lemons,30 and Arabidopsis93 have been found to harbor intracellular signaling cascades, which modulate cellular responses
heat shock proteins and aquaporin proteins. Other plasmalemma- and bring about tissue homeostasis.52 In the recurrent inflammatory
associated proteins mainly include metabolic enzymes, signal trans- disorders of intestine such as colitis, intestinal macrophages get
duction factors, adhesion factors, cytoskeleton proteins, and ubiqui- deprived of their tolerogenic traits. Wang et al.104 demonstrated
tin.94,95 There is a need of considerable research for identification that grapefruit-derived ELNVs have a propitious impact on intestinal
of a broad range of PELNV proteins, which possibly will reveal their immune homeostasis by fortifying the anti-inflammatory capability
roles in biological and pharmacological activities. of intestinal macrophages, which ultimately alleviate dextran sulfate
sodium (DSS)-induced colitis in mice with no toxicity. After the
Identification of RNAs successful uptake of grapefruit-derived ELNVs by intestinal macro-
The most fascinating trait of ELNVs is their ability to ferry various phages, these nanoplatforms upregulate the expression of heme-oxy-
RNA species among cells. RNAs in PELNVs mainly include genase-1 (HO-1) and interleukin (IL)-10 and suppress the production
mRNA, miRNA, and small RNA (sRNA), among which mRNA of IL-6, IL-1b, and tumor necrosis factor (TNF)-a. Moreover,
and non-coding miRNA can regulate the levels of RNA and proteins miRNAs of ginger- and grapefruit-derived ELNVs were also reported
in recipient cells, thus affecting cell morphology and function.96–98 to target the genes panel in gut probiotic Lactobacillus rhamnosus
Hitherto, most studies on ELNV-mediated communication have (LGG) alongside growing its number in mice gut and consequently
been concentrated on notable RNA species such as miRNAs and prompting anti-microbial immunity and wellbeing of the gut. Similar

18 Molecular Therapy Vol. 29 No 1 January 2021


www.moleculartherapy.org

Review

Figure 4. Overview of Biological Functions of PELNVs from a Variety of Plant Sources and Their Translation into Therapeutic Applications
(A) Summary of the biological functions of PELNVs derived from plants. (B) Elucidation of therapeutic mechanisms of PELNVs in immunological modulation, tissue repair, and
intestinal transporters modulation.

investigations on broccoli, grapes, and carrots have also been re- there has been no report up to now of any immunogenic response
ported, which gave credence to the impressive role of PELNVs induced by PELNVs that makes them safe and biocompatible. Besides
in immunological modulation therapeutics.52,65 Interestingly, that, transgenic plant-derived virus-like particles have been

Molecular Therapy Vol. 29 No 1 January 2021 19


www.moleculartherapy.org

Review

Figure 5. Mechanism of PELNV miRNAs Regulating the Inter-kingdom Communication between Gut Microbiota and the Host Immune System
PELNV mRNA-mediated targeting of the LGG monooxygenase ycnE increase the yields of indole-3-carboxaldehyde (I3A), which leads to inducing the production of IL-22.
IL-22 can improve the barrier function and ameliorate colitis in mice.

demonstrated as an effective nanoplatform to produce vaccines owing tive stress reduction, and most importantly by activating TNF-related
to their ability to trigger a desired immunogenic response.105,106 apoptosis-inducing ligand (TRAIL)-mediated apoptotic cell death.112

Anti-tumor Activities Regenerative Effects


Cancerous tumors pose a grave danger to human health, and medic- Humans ingest PELNVs during the whole span of their life, but very
inal plants play important roles in the treatment of tumors to make up limited information has seen daylight regarding their functionality
for the shortcomings of modern medicine.107 The rise of miRNA and impact on the intestinal tract. Ju et al.86 explored the role of
research has led people to gradually realize that miRNA is closely grape-derived ELNVs on DSS-induced colitis in mouse models. It
related to tumor proliferation and apoptosis.108–110 With the further was shown that they have the capability to diffuse through and enter
study of ELNVs, more and more research has shown that plant- intestinal stem cells and thereupon induce the proliferation of Lgr5hi
derived miRNAs can span species and play a certain regulatory role intestinal stem cells and regenerate the intestinal epithelial tissue.
in the human body.111 For instance, ginger-derived ELNVs show Moreover, these nanoplatforms can upregulate the expression of
promising results in curtailing colorectal tumorigenesis by diminish- pluripotent stem cell markers such as SOX2, Oct4, and Klf4, which
ing pro-inflammatory cytokines and suppressing intestinal epithelial will prompt the homeostasis of intestinal epithelial tissue. In addition,
cell proliferation and apoptosis, as overexpression of cyclin D1 these findings also set the precedent for future implications of grape-
mRNA suggests early cancer onset and tumor progression. It was derived ELNVs for inducing the proliferation of Lgr5hi stem cells in
also found that ginger-derived ELNVs reduced cyclin D1 mRNA other ailments.113 Moreover, ginger-derived ELNVs exhibited the
levels in mouse colorectal cancer models, corroborating the anti- role in curtailing the expression of the secreted protein hemopexin
tumor action of ginger-derived ELNVs.87 In addition, Citrus limon and also influenced the expression of other mitochondrial and
L.-derived ELNVs were also explored for their role in the curtailment cytoplasmic proteins such as heat shock protein, axin, and kinesin
of tumor growth. They were able to curtail the in vivo tumor develop- to promote intestinal wound healing.87 In one remarkable investiga-
ment in chronic myeloid leukemia (CML) by tumor targeting, oxida- tion, wheat-derived ELNVs induced skin regeneration by triggering

20 Molecular Therapy Vol. 29 No 1 January 2021


www.moleculartherapy.org

Review

proliferative and migratory actions in a dose-dependent manner on required.122 On the one hand, such PEG coatings extend the circula-
epithelial, endothelial, and dermal fibroblasts in in vitro studies. tion time and help with the immune tolerance to some extent; on the
Moreover, expression of the mRNA level of collagen type I was other hand, these coatings can hamper the interaction between the
elevated significantly. Aside from these proliferative and migratory nanoparticle and target cells, thereby diminishing the drug bio-
actions, wheat-derived ELNVs were reported to be proangiogenic in distribution in target tissue.123 Moreover, it also has been reported
nature by triggering tube-like structure formation in a human umbil- that repeated administration of PEG-coated liposome has contributed
ical vein endothelial cell (HUVEC) line, which suggested that to formation of anti-PEG antibodies, resulting in ineffective ther-
wheat-derived ELNVs have the capability to induce vascular forma- apy.124,125 Furthermore, synthetic nanoparticles only serve the
tion during the wound healing process. These findings present exam- purpose of delivering drug molecules and do not contain any innate
ples for future exploration of wheat-derived ELNVs in in vivo studies therapeutic capability.
for wound healing development.114
In comparison, PELNVs rose to new heights during the last decade
Hepatoprotection with the discovery of their innate therapeutic capabilities in cross-
Researchers explored the use of ginger-derived ELNVs for amelioration kingdom communication such as anti-tumor, regenerative, and
of alcohol-induced liver damage in mice. Alcohol-derived metabolites anti-inflammatory activities. Moreover, PELNVs take the lead by of-
can stimulate pro-inflammatory cytokines, transforming growth factor, fering the merits of small size for deep tissue penetration, slightly
TNF-a, reactive oxygen species (ROS), and collagen, which lead to liver negative zeta potential for extended circulation,126 resemblance to
deterioration. Ginger-derived ELNVs suppress ROS, which contributes the plasma membrane, and pronounced physicochemical stability
to hepatoprotection. Owing to shogaols and other bioactive con- under varying pH values and temperatures.79,127 When set in com-
stituents, they can mediate the activation of nuclear factor erythroid parison with liposomes, their homing capability to tumor tissues is
2-related factor 2 (Nrf2). Reportedly, Nrf2 stimulation leads to the 10-fold higher, denoting a higher specificity for tumor targeting.128
expression of liver detoxifying and antioxidant genes, which collectively Contrary to mammalian or bacterial cell-derived ELNVs, PELNVs
contributes to hepatoprotection. In conclusion, ginger-derived ELNVs abate the potential perils of unwanted genetic or protein transfer
can induce hepatocyte metabolism and detoxifying and antioxidant de- and a detrimental immunogenic response before clinical applica-
fenses, which are vital for hepatoprotection.115 tion.129 Additionally, PELNV-derived nanovectors have reportedly
been successful in traversing mammalian hindrances such as
PELNVs as Drug Delivery Nanoplatforms diffusing through the blood-brain barrier (BBB), which avoids any
ELNVs are committed to ferry a payload between cells in their near excitation of inflammatory response or necrosis, unlike artificially
vicinity to faraway sites, but they can also deliver payloads to other fabricated liposomes.51 Moreover, lipid bilayer structure of PELNVs
species in inter-kingdom communication. Their unique design and shrouds their cargo securely and evades enzymatic decomposition
transport capability accentuate their potential for use in drug by proteinases and nucleases.130 All of these stellar features coupled
delivery.116 The advantages of PELNV-based nanoplatforms over with discovery of their intrinsic therapeutic actions make PELNVs
synthesized nanoplatforms and mammalian-derived ELNVs for an ideal for their advent into the arena of drug delivery applications.
drug delivery have been demonstrated. First, the selection of ELNVs Here, we shed light on PELNV-based nanoplatforms for targeted
as a drug delivery nanoplatform demands strict scrutiny because their drug delivery.
innate biological actions and derivation source play pivotal roles in
their immunogenic acceptance. For example, ELNVs derived from Colon-Targeted Delivery
mammalian cancerous cells pose the risk of transmitting pro- PELNVs proved themselves to be phenomenally biocompatible
cancerous traits to recipient cells.117 PELNVs and artificially synthe- through oral administration and successful in achieving targeted co-
sized nanoparticles such as liposomes and micelles share common lon drug delivery.86 This is because PELNVs are naturally capable of
essential traits, including a comparable lipid bilayer structure and enhancing the bioavailability of bioactive compounds via keeping
the ability to deliver both hydrophilic and hydrophobic cargo.118,119 the load stable and secure.131,132 One study showed that ELNVs
However, when set in comparison, there are stark differences that with their liquid-ordered phase membrane resist detergent lysis to
show that PELNVs fare better against artificially synthesized nano- a greater degree than do other subtypes of EVs.133 Moreover, it
particles in terms of being innocuous and having low immunogenic has also come to light that PELNVs are resistant to digestive en-
effects, enhanced cellular uptake, higher stability in the GI tract zymes such as pepsin, intestinal pancreatin, and bile extract solution.
(GIT), and specific target ability.120 Moreover, PELNV-based nano- This stellar feature helps them in circumventing the decomposition
platforms offer the benefit of a comparatively less complex prepara- of their cargo by the gastric or intestinal environment and delivering
tion method, whereas artificially synthesized nanoparticles such as it to the desired colon site securely. However, it is still not well
liposomes require complex fabrication processes involving mem- understood how PELNVs resist the harsh GI environment and ac-
brane extrusion and microemulsification, among others.121 More- quire the safe entry into intestinal cells while keeping their payload
over, additional rigorous modifications of synthetic nanoparticles intact. In one attempt, grapefruit-derived ELNVs were loaded with
for cargo loading and coating refinements, e.g., polyethylene glycol methotrexate to be aimed at intestinal macrophages. Upon oral de-
(PEG) coatings to develop satisfactory immune tolerance, are also livery of methotrexate-loaded conjugate, it was revealed that these

Molecular Therapy Vol. 29 No 1 January 2021 21


www.moleculartherapy.org

Review

Figure 6. Schematic Illustration of Transdermal Delivery of PELNVs


PELNVs could penetrate the stratum corneum though three pathways: transcellular routes, intercellular routes, and appendageal routes.

conjugates effectively targeted F4/80+ macrophages localized in the cally applied on skin, they will penetrate via a “lipid-rich channel”
intestinal lamina propria through micropinocytosis and clathrin- coating on hair follicles.135,136 There are two ways that PELNVs could
dependent cellular uptake pathways. After internalization these con- reach the hair shafts.137 PELNVs could reach the hair shafts by either
jugates made significant decline in the DSS colitis-associated weight entering hair matrix cells and moving farther by cell differentiation or
loss and reversed the shortening of colon length along with its via direct entry into hair shafts from the hair tip or else the side of hair
obvious anti-inflammatory response by curbing the production of shaft.138 In addition, PELNVs structurally resemble liposome (i.e., bi-
proinflammatory cytokines TNF-a, IL-1b, and IL-6 in mice. layered phospholipid structure and flexible diameters),139 the skin
Furthermore, the side effects of methotrexate significantly subsided penetration of which through trans-follicle routes has previously
when delivered by grapefruit-derived ELNVs as compared to free been reported by our group and others.140–142 In addition to the
methotrexate. Therefore, the compelling innate tendency of PELNVs trans-follicle route, another route to enter dermis is through penetra-
to augment anti-inflammatory actions of intestinal macrophages tion of the stratum corneum (SC). This route follows two pathways,
coupled with cargo delivery ability put forth an interesting solution i.e., transcellular and intercellular routes.135 Owing to the similarity
for the prevention or treatment of autoimmune diseases and colon of the membrane surface between PELNVs and mammalian cells,
cancer.104 Finally, yet importantly, PELNVs also show the potential PELNVs are thought to be efficient in passing through the SC both
of being exploited as a personalized oral delivery tool because grape- by the transcellular and intercellular routes through the lipid fusion
derived ELNVs were found to target intestinal cells, whereas grape- effect (Figure 6). In addition, the near-infrared-induced hyperthermia
fruit-derived ELNVs targeted macrophagess.86 Therefore, such re- effect can cause the configuration change of the membrane and hence
ports foreshadow the possible personalized modulation of the ac- increase the permeability of the SC, which could also enhance the
tions of recipient cells according to therapeutic goals. penetration of ELNVs. Recently, a study surfaced that gave credence
to the use of PELNVs in transdermal drug delivery. It was reported
Transdermal Delivery that broccoli-derived ELNVs, being highly lipophilic, represent a
There are generally two pathways for transdermal permeation of high entrapment efficacy and can be deployed to penetrate deeply
exogenous agents. One is the physical channels of skin appendages into the skin tissue. Enhanced fusion of broccoli-derived ELNVs
with adequate micrometer diameter to provide passage to nanove- with keratinocytes was corroborated by the transport of an encapsu-
sicles.134 Mechanistically, it is speculated that once PELNVs are topi- lated fluorescent agent into the cells and also by the presence of

22 Molecular Therapy Vol. 29 No 1 January 2021


www.moleculartherapy.org

Review

Figure 7. Modification of PELNVs for Targeted Drug Delivery


(A–D) Targeted drug delivery applications of (A) pristine PELNVs and (B) PELNV re-engineered nanovectors, with further elucidation of surface functionalization of pristine
PELNVs, and (C and D) surface modification of re-engineered nanovectors for achieving cargo loading and targeted delivery.

broccoli proteins in the plasmalemma of keratinocytes. Hence, model cells and inhibited their growth. It is noteworthy here that pro-
PELNVs can also be envisaged as an efficient conduit to transdermal nounced anti-tumor action was not solely due to doxorubicin, but it
drug delivery.143 Furthermore, considering enhanced skin perme- was also partly due to suppression of oxidative stress and pro-inflam-
ation and cargo entrapment efficacy, PELNVs can be exploited for matory cytokine levels induced by the inherent nature of ginger-
transdermal gene delivery for skin melanomas based on our past derived ELNVs. Additionally, it was suggested that ginger-derived
experience in transdermal gene delivery.110 nanovectors opportunistically triggered doxorubicin release in acidic
pH, exploiting the acidic extracellular microenvironment of tumors
Tumor-Targeted Delivery and consequently indicating diminished doxorubicin side effects.
The present chemotherapeutic approaches in the fight against cancer Furthermore, it was reported that they were innocuous, biocompat-
confront two interwoven impediments of low target specificity and ible, and able to be produced economically on a large scale. On a
higher toxicity. To tackle both of these challenges, great efforts have comparative investigation with artificially synthesized liposome,
been made toward development of multiple new drug delivery sys- they happen to be superior to liposome in terms of biocompatibility,
tems such as cell membrane capsules144 and multiple generations of safety, apoptosis, electric cell-substrate impedance, and pH-depen-
liposomes, but such efforts are still faced with issues of toxicity and dent profile for drug release.57
target specificity.145 Conversely, PELNV-derived re-engineered
nanovectors have proven themselves to be innocuous and non- In another investigation, grapefruit-derived nanovectors were
immunogenic with target ability. As a case in point, scientists have demonstrated for the co-delivery of FA and paclitaxel to target the
been successful in re-engineering nanovectors from ginger-derived CT26 colon cancer model (Figure 7B) and the human SW620 colon
ELNVs with the aim to deliver doxorubicin to colon tumor cells. cancer severe combined immunodeficiency (SCID) mouse model
These nanovectors efficiently internalized into Colon-26 tumor cells intravenously. Moreover, these nanovectors were reported to

Molecular Therapy Vol. 29 No 1 January 2021 23


www.moleculartherapy.org

Review

be innocuous with higher aiming efficacy, as they did not traverse the that ginger-derived ELNVs innately harbor 6-gingerol and 6-shogaol,
placental barrier when injected intravenously in pregnant mice. This which are pharmacologically bioactive molecules with prominent
paves the way for potential use of grapefruit-derived nanovectors in anti-oxidative and anti-tumor actions.112 Furthermore, researchers
pregnant women for delivering certain drugs in the future.51 In sub- successfully transferred siRNA-CD98 to intestinal epithelial tissue
sequent research by the same group, they carried out the modification by ginger-derived nanovectors, and they reported that these orally
of grapefruit-derived nanovectors by coating them with an inflamma- administered nanoplatforms impressively were retained in the upper
tory chemokine receptor-enriched plasma membrane (Figure 7C). As colon and ileum and diminished CD98 gene expression and curbed
a result, these nanoplatforms were targeted to the inflamed CT26 co- the inflammation caused by ulcerative colitis and also prevented co-
lon tumor and 4T1 breast tumor in mouse models to deliver the litis-associated cancer.58 Moreover, apart from efficient siRNA-
chemotherapeutic agent doxorubicin and anti-inflammatory drug CD98 delivery and its high activity, anti-inflammatory action could
curcumin. It was also reported that leukocyte function-associated an- also be partially attributed to structural components of ginger-derived
tigen-1 (LFA-1) and CXC chemokine receptor 2 (CXCR2) on the sur- nanovectors, which curb pro-inflammatory cytokines (TNF-a, IL-1b,
face of coated grapefruit-derived nanovectors effectuated the in- and IL-6) and promote the induction of anti-inflammatory cytokines
flamed tumor target specificity.130 It cannot be ignored that not (IL-10 and IL-22).88,112 Hence, it can be deduced that intrinsic ther-
only do PELNVs play the role of delivery vehicle for chemothera- apeutic action coupled with gene delivery capability raise the thera-
peutic drugs, but they also execute their inherent anti-tumor thera- peutic index of PELNV-based therapies.
peutic action simultaneously. However, it is noteworthy that the ther-
apeutic role is dependent on selection of the plant source of PELNVs. Cellular Uptake and Biodistribution of PELNVs
For instance, naringin, a key flavanone in grapefruit,146 can be hydro- The driving mechanisms behind the cellular internalization and cargo
lyzed by intestinal microbiota into its active metabolite naringenin, delivery of ELNVs into recipient cell cytosol are still poorly identified.
which displays anti-tumor action.146,147 Moreover, PELNVs have The pivotal step of ELNV internalization is the entry step into the
the ability to disrupt the cascade of several protein expressions vital recipient cell. ELNV cellular internalization into recipient cells,
for tumor development. For instance, PELNVs have been found to occurring either via a non-specific process such as micropinocytosis
promote anti-tumor action by inhibiting cyclin D1 mRNA expression or macropinocytosis, or through a specific receptor-dependent route,
and by activating cyclic guanosine monophosphate (cGMP)-depen- is still ambiguous. On the grounds that multifarious ELNVs share a
dent protein kinase, which ultimately assist in tumor suppression common surface proteome, it is speculated that one of them assumes
by inhibiting T cell transcription signaling.87 Additionally, innate the role of general ligand for the cell receptor, facilitating ELNV inter-
characteristics of PELNVs to upregulate the expression of pro- nalization.150 Additionally, ELNV cellular internalization has been re-
apoptotic molecules and suppression of angiogenic factors such as cy- ported to happen via endocytic pathways such as clathrin-dependent
tokines exert anti-proliferative and apoptotic actions on cancerous and clathrin-independent pathways. Even so, this ELNV feature lacks
cells while sparing normal cells.112 Therefore, considering all of these consensus among researchers. Currently, it is theorized that the deter-
aspects, it can be proposed that integration of natural PELNVs and minative factor behind the internalization route is recipient cell types
chemotherapy could be harnessed as a viable strategy to eradicate rather than ELNVs themselves.151
cancer in the future.
PELNV-based nanoplatforms laden with drug molecules have been
Gene Delivery reported to be introduced into experimental subject animals via
Previously, non-viral delivery vectors have been deployed to deliver oral,58 intravenous,46 intramuscular, and intranasal routes.51 There
nucleic acid-based drugs to disrupt the gene expression of multiple are multiple reported pathways involved in the cellular internaliza-
diseases. However, such vectors typically have the innate risks of tion of PELNV-based nanoplatforms such as phagocytosis, macro-
immunogenicity and toxicity.148,149 Although PELNVs are neophytes pinocytosis, clathrin-mediated endocytosis, and caveolae-mediated
in the arena of gene delivery, they provide a solution to aforemen- endocytosis (Figure 8).57,104 Biodistribution and bioavailability of
tioned complications and also go a notch above by traversing strin- these drug-laden nanoplatforms are influenced by multiple factors,
gent biological barriers such as the BBB. For instance, grapefruit- e.g., selection of administration route, source plant’s natural charac-
derived nanovectors were tasked to carry exogenous miRNA teristics, surface charge, and size dimension. In vivo biodistribution
(miR17) to mouse brain tumor via the intranasal route (Figure 7D), of several DiR-labeled PELNVs and re-engineered nanovectors have
as reports had suggested that major histocompatibility complex class been evaluated by fluorescently enabled high-imaging systems.
I (MHC class I) is among the prominent targeted genes of miR17. Af- Ginger-derived nanovectors were reported to be impressively re-
ter administration, grapefruit-derived nanovectors were able to tained in stomach, colon, and upper ileum even after 12 h of oral
achieve quick intracerebral tumor internalization for miR17 delivery, administration.58 Similarly, ginger-derived PELNVs prefer to
which downregulated the MHC class I expression of tumor cells by continue to dwell inside intestine upon oral administration.88 In
the activation of natural killer cells, thus inhibiting the tumor contrast, grapefruit-derived nanovectors were concentrated in mac-
growth.9 Similarly, Li et al.46 successfully silenced the survivin gene rophages of the lamina propria of cecum and colon of small intes-
expression in subcutaneous tumor cells via intravenous delivery of tine and later preferentially migrated to the liver as well.104 In addi-
siRNA by ginger-derived ELNVs (Figure 7A). It is highlighted here tion, upon intranasal delivery of DiR-labeled grapefruit-derived

24 Molecular Therapy Vol. 29 No 1 January 2021


www.moleculartherapy.org

Review

Figure 8. Elucidation of Multiple Pathways of Cellular Internalization/Uptake of PELNVs after Introduction into the Subject
Facilitated diffusion, endocytosis, and direct fusion are shown.

nanovectors in mice, elevated intensity of fluorescence in lungs and leoyl-3-trimethylammonium-propane)-DOPE (dioleoylphosphati-


brain was detected, thus demonstrating brain tumor target ability.51 dylethanolamine) liposomes. ALT, AST, and pro-inflammatory
Intravenously administered DiR-labeled ginger-derived nanovec- cytokine levels were reported to be notably elevated in liposomal-
tors and FA-coupled ginger-derived nanovectors exhibited treated mice and, surprisingly, no elevation was recorded in the
augmented levels of fluorescence in the liver of mice, suggesting a PELNV-treated mice group. In addition to this, no pathological
higher accumulation and probable contribution toward detoxifica- modification or necrosis at all was observed in any of the histological
tion. Meanwhile, relatively lower fluorescence was visible in other samples of liver, kidneys, spleen, and lungs in the PELNV-treated
organs such as heart, kidney, and spleen. Fluorescence intensity mice group.51 Moreover, in vitro MTT (3-(4,5-dimethylthiazol-2-
of DiR-labeled, FA-coupled ginger-derived nanovectors was found yl)-2,5-diphenyltetrazolium bromide) assays also revealed compara-
to be identifiable in blood circulation after 48 h. Moreover, it was ble result to earlier mentioned findings on PELNVs, where PELNVs
reported to be 2.8-fold higher than its counterpart, suggesting had far less impact on viability in both RAW 264.7 and colon-26
enhanced tumor target ability, drug delivery, and diminished sys- cell lines as compared to liposomes.58 These reports suggest that ar-
temic toxicity to normal organs.57 Based on these findings, it can tificially synthesized nanoplatforms induce an immune response in
be deduced that PELNV-based nanoplatforms are equipped to be the host, meaning that they are sequestered by filtering through or-
retained at the target site for the desired duration with no toxicity. gans, and, hence, clearance of these nanomaterials is enhanced as a
result. Conversely, during investigations, it has become explicit that
PELNV Toxicity and Immunogenicity PELNVs are phenomenally non-immunogenic and in fact have
An ideal drug delivery system must ensure its superlative non-toxicity strong supportive immunomodulatory actions that bring about tissue
and non-immunogenicity with no side effects in in vitro and in vivo as homeostasis and contribute to an organism’s wellbeing.
well.152,153 Even though there have been meagre attempts to find their
cytotoxic effects on living subjects, PELNVs show a fascinating In conclusion, PELNV-based nanoplatforms display a wide range of
biocompatible nature so far because of their natural provenance. advantages with respect to biocompatibility, stability, biodistribution,
For instance, in probes related to cytotoxic potential effects of grape- prolonged half-life, and cellular internalization. Additionally, one of
fruit-derived ELNVs in mice, several markers such as pro-inflamma- the key benefits of using PELNVs for drug delivery is their ability
tory cytokines and serum levels of liver enzymes, e.g., alanine trans- to reduce the side effects of a loaded drug when administered into
aminase (ALT) and aspartate transaminase (AST), were experimental subjects.104 All of these characteristics make them
quantitatively measured. Mice were pre-administered with grape- poised to provide a safe, robust, and feasible approach for targeted
fruit-derived ELNVs and commercially available DOTAP (1,2-dio- drug delivery.

Molecular Therapy Vol. 29 No 1 January 2021 25


www.moleculartherapy.org

Review

Clinical Trials nisms that govern PELNV biogenesis, release, and functions remains
There are currently three PELNV-based clinical trials registered by limited. Applicable isolation techniques are still time-consuming in
the same sponsor, i.e., the University of Louisville. Grape-derived acquiring pure PELNVs and demand to be rectified to achieve the
ELNVs are currently in clinical trials and being tested on humans commercial and therapeutic translations of PELNVs. There is a dire
for averting oral mucositis associated with chemoradiation therapy need for standardization of qualitative and quantitative analyses to
for head and neck cancer (ClinicalTrials.gov: NCT01668849). develop better comparative interpretations of results. In addition,
Furthermore, aloe- and ginger-derived ELNVs are also under human there are still unresolved concerns of regulatory affairs of PELNV
clinical trials for the treatment and mitigation of symptoms (e.g., in- therapeutic and drug delivery applications as there has been no agree-
sulin resistance and chronic inflammation) associated with polycystic ment yet on how they should be classified for approval. Similarly, no
ovary syndrome (PCOS) (ClinicalTrials.gov: NCT03493984). In addi- consensus has been reached thus far related to routes followed by
tion to their role as an active pharmaceutical ingredient, clinical trials PELNVs for internalization or for cargo delivery to recipient cell, as
for harnessing these PELNVs as drug delivery nanoplatforms are well as how PELNV uptake or delivery induces transformations in
underway where they are being investigated for encapsulating the recipient cells.
hydrophobic drug curcumin for delivery to normal colon tissue and
colon tumor (ClinicalTrials.gov: NCT01294072). One major limitation pertaining to PELNVs is their inadequacy to
carry any high quantity of drug load. The membrane fusion tech-
The ELNV-mediated therapeutic approach is expected to be the nique145 could be harnessed to amalgamate artificially synthesized
cornerstone of a paradigm shift toward advanced cancer therapies. liposomes with PELNVs to achieve augmented cargo loading
In particular, rising interest in anti-tumor action of PELNVs is due capability. Resultant hybrid nanoplatforms may offer tractable
to their dual functionality, i.e., an intrinsic therapeutic capability physicochemical characteristics owing to feasible manipulation of
and the ability to deliver chemotherapeutic drugs. The anti-tumor ac- the integrated liposomal part. Additionally, these hybrid nanoplat-
tion of PELNVs is highlighted by their ability to interfere in the forms could be explored for delivery of CRISPR-Cas9 expression vec-
expression of multiple proteins implicated in tumor progression. tors for gene therapy. This technique might be helpful in obviating
Additionally, TRAIL-mediated apoptotic cell death, anti-angiogenic immunogenicity, cytotoxicity, and long-term expression because of
actions, and pharmacologically active components of PELNVs also the small size of PELNVs and their ability to traverse stringent biolog-
cast a spotlight on their anti-neoplastic actions.87,112 In addition, ical barriers and escape from the mononuclear phagocyte system
owing to elevated permeability and the retention effect, PELNVs (MPS).
tend to lodge inside tumor tissues containing ill-formed blood vessels
than they do in normal tissues.128 Furthermore, PELNVs also display In addition, our current knowledge of PELNV physiology, diver-
the possibility of re-engineering them with tumor-specific targeting sity, internalization, and cargo delivery is still limited to yield
proteins, peptides, or antibodies for precise therapy. At the same clear mechanistic conclusions about precisely how PELNVs
time, clinical translation of ELNVs is still thought to be limited by interact with and modify acceptor cells. Determining whether
the lack of appropriate, scalable, and cost- and time-effective nano- internalization occurs through macropinocytosis or micropinocy-
technologies for the purification and loading of EVs. Moreover, as tosis and/or receptor-mediated pathways, and whether these pro-
part of daily nutrition, clinical utilization of PELNVs may pose cesses result in cargo delivery, will be essential to understand and
different concerns than human cell-based medicinal EV products. control PELNV uptake and delivery of cargoes for clinical pur-
Notably, some doubts pertaining to cell-to-cell transmission of the poses. Intrinsic biochemical traits endow PELNVs with profound
plant toxin trichosanthin via ELNVs have been raised.154 Thus, potential of implementation into the drug delivery system.
possible perils linked to the transfer of toxins and allergens via PELNVs are already innately laden with bioactive compounds.
PELNVs should be addressed before considering them for clinical Further weaponizing these nanovesicles with drugs poses perils
studies. of interactions between innate bioactive components and exoge-
nously loaded drugs. Hence, this demands extensive and in-depth
Challenges and Perspectives investigations for developing design strategies to load drug mole-
PELNVs are important conveyers of information between cells cules into these nanovesicles without any potential interactions.
through the transmission of various proteins, bioactive lipids, and ge- Moreover, an additional process of unloading innate bioactive
netic information to alter the phenotype and function of recipient compounds from nanovesicles prior to drug loading could be
cells. Thus, PELNVs have now been implicated in numerous biolog- used; however, the unloading step should not compromise surface
ical and pathological processes not only between plant cells, but also morphology, size, and biological functions. Most therapeutic ef-
between cells of other species. However, there still lies ambiguity as to fects achieved by PELNVs are through their natural bio-
whether communication to recipient cells is specific or stochastic. distribution. However, PELNVs could be imparted with target
Moreover, their exploitation as nanotherapeutics or to deliver pro- specificity by investigating surface tailoring of PELNVs to selec-
teins, nucleic acids, and other drug cargoes across various biological tively amass at far away and difficult-to-target tumors without
barriers has been garnering much attention recently. However, until causing any immunogenicity. Second, PELNVs can be used as
recently, our understanding of the cellular and molecular mecha- an exemplary vehicle in the future for drugs that need to be

26 Molecular Therapy Vol. 29 No 1 January 2021


www.moleculartherapy.org

Review

transported across the BBB because they can traverse the BBB icles as transdermal nanoplatforms for photo-TRAIL-programmed therapy against
melanoma. Sci. Adv. 6, eaba2735.
and obviate toxicity and rapid drug clearance complexities by
MPS associated with other nanocarriers. Third, as PELNVs struc- 3. Shao, H., Im, H., Castro, C.M., Breakefield, X., Weissleder, R., and Lee, H. (2018).
New technologies for analysis of extracellular vesicles. Chem. Rev. 118, 1917–1950.
turally resemble liposomes along with their ability to penetrate
4. Harding, C., Heuser, J., and Stahl, P. (1983). Receptor-mediated endocytosis of
skin, they can be envisaged as potential delivery nanoplatforms transferrin and recycling of the transferrin receptor in rat reticulocytes. J. Cell
for transcutaneous vaccination as well.136 In order to acquire ver- Biol. 97, 329–339.
satile biomedical applications of PELNVs, the concept of amal- 5. Pan, B.-T., and Johnstone, R.M. (1983). Fate of the transferrin receptor during
gamation of natural organic PELNVs with inorganic nanomateri- maturation of sheep reticulocytes in vitro: selective externalization of the receptor.
als such as magnetic nanoparticles endowed with photothermal Cell 33, 967–978.

conversion traits is propounded. These envisioned hybrid nano- 6. Marchant, R., and Robards, A. (1968). Membrane systems associated with the plas-
malemma of plant cells. Ann. Bot. 32, 457–471.
platforms might provide the opportunity to explore the theranos-
7. Halperin, W., and Jensen, W.A. (1967). Ultrastructural changes during growth and
tic multifunctionality with augmented targeted drug delivery embryogenesis in carrot cell cultures. J. Ultrastruct. Res. 18, 428–443.
coupled with localized hyperthermia under the influence of exter-
8. De Jong, W.H., and Borm, P.J. (2008). Drug delivery and nanoparticles: applications
nally manipulated factors such as magnetic field. and hazards. Int. J. Nanomedicine 3, 133–149.
9. Zhuang, X., Teng, Y., Samykutty, A., Mu, J., Deng, Z., Zhang, L., Cao, P., Rong, Y.,
In conclusion, ELNVs have shown many advantages, especially in Yan, J., Miller, D., and Zhang, H.G. (2016). Grapefruit-derived nanovectors deliv-
biocompatibility, cellular uptake, and targeting capability. Current ering therapeutic miR17 through an intranasal route inhibit brain tumor progres-
sion. Mol. Ther. 24, 96–105.
synthetic drug delivery systems, including inorganic and polymeric
10. An, Q., van Bel, A.J., and Hückelhoven, R. (2007). Do plant cells secrete exosomes
nanoparticles, are intrinsically foreign materials and have much
derived from multivesicular bodies? Plant Signal. Behav. 2, 4–7.
more potential toxicity and immunogenicity. In comparison,
11. Liang, H., Zhang, S., Fu, Z., Wang, Y., Wang, N., Liu, Y., Zhao, C., Wu, J., Hu, Y.,
PELNVs, which are natural and endogenic, are considered as much Zhang, J., et al. (2015). Effective detection and quantification of dietetically absorbed
more biocompatible with multiple types of biofunctions along with plant microRNAs in human plasma. J. Nutr. Biochem. 26, 505–512.
the parent cells. We have made some great strides during the last 12. Woith, E., Fuhrmann, G., and Melzig, M.F. (2019). Extracellular vesicles—connect-
decade toward understanding the biomedical potential of PELNVs, ing kingdoms. Int. J. Mol. Sci. 20, 5695.
but in fact we have just started to scratch the surface of what they 13. Reiner, A.T., and Somoza, V. (2019). Extracellular vesicles as vehicles for the deliv-
can do. Certainly, we will encounter some challenges regarding the ery of food bioactives. J. Agric. Food Chem. 67, 2113–2119.

future implementations of PELNVs in therapeutics and drug delivery 14. Akuma, P., Okagu, O.D., and Udenigwe, C.C. (2019). Naturally occurring exosome
vesicles as potential delivery vehicle for bioactive compounds. Front. Sustain. Food
exploration. Nevertheless, these impediments will be resolved, as has Syst. 3, 23.
been witnessed in any newly burgeoning field. It is envisaged that the
15. McMullen, C., Gardner, W., and Myers, G. (1977). Ultrastructure of cell-wall thick-
multidisciplinary approach of integrating biological science, drug de- enings and paramural bodies induced by barley stripe mosaic virus. Phytopathology
livery techniques, nanotechnologies, and engineering will steer the 67, 462–467.
course for development of PELNV-based therapeutics and novel de- 16. Chafe, S.C. (1974). Cell wall structure in the xylem parenchyma of Cryptomeria.
livery nanoplatforms. Protoplasma 81, 63–76.
17. Robards, A., and Kidwai, P. (1969). Vesicular involvement in differentiating plant
vascular cells. New Phytol. 68, 343–349.
ACKNOWLEDGMENTS
18. Tanchak, M., and Fowke, L. (1987). The morphology of multivesicular bodies in soy-
The study was supported by the National Key Research and Develop- bean protoplasts and their role in endocytosis. Protoplasma 138, 173–182.
ment Program of China (2018YFC1105404), the Key Project at 19. An, Q., Hückelhoven, R., Kogel, K.H., and van Bel, A.J. (2006). Multivesicular bodies
Central Government Level (2060302), and the National Natural Sci- participate in a cell wall-associated defence response in barley leaves attacked by the
ence Foundation of China (81473145). pathogenic powdery mildew fungus. Cell. Microbiol. 8, 1009–1019.
20. Collins, N.C., Thordal-Christensen, H., Lipka, V., Bau, S., Kombrink, E., Qiu, J.-L.,
Hückelhoven, R., Stein, M., Freialdenhoven, A., Somerville, S.C., and Schulze-Lefert,
AUTHOR CONTRIBUTIONS
P. (2003). SNARE-protein-mediated disease resistance at the plant cell wall. Nature
L.-H.P. and L.-Q.H. conceived the project. H.A.D., T.-W.G., and 425, 973–977.
A.-Q.Z. wrote the manuscript. All authors discussed the contents 21. Meyer, D., Pajonk, S., Micali, C., O’Connell, R., and Schulze-Lefert, P. (2009).
and commented on the manuscript. Extracellular transport and integration of plant secretory proteins into pathogen-
induced cell wall compartments. Plant J. 57, 986–999.

DECLARATION OF INTERESTS 22. Regente, M., Corti-Monzón, G., Maldonado, A.M., Pinedo, M., Jorrín, J., and de la
Canal, L. (2009). Vesicular fractions of sunflower apoplastic fluids are associated
The authors declare no competing interests. with potential exosome marker proteins. FEBS Lett. 583, 3363–3366.
23. Li, X., Bao, H., Wang, Z., Wang, M., Fan, B., Zhu, C., and Chen, Z. (2018). Biogenesis
REFERENCES and function of multivesicular bodies in plant immunity. Front. Plant Sci. 9, 979.
1. Colombo, M., Raposo, G., and Théry, C. (2014). Biogenesis, secretion, and intercel- 24. Regente, M., Pinedo, M., San Clemente, H., Balliau, T., Jamet, E., and de la Canal, L.
lular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. (2017). Plant extracellular vesicles are incorporated by a fungal pathogen and inhibit
Biol. 30, 255–289. its growth. J. Exp. Bot. 68, 5485–5495.
2. Peng, L.-H., Wang, M.-Z., Chu, Y., Zhang, L., Niu, J., Shao, H.-T., Yuan, T.J., Jiang, 25. Hansen, L.L., and Nielsen, M.E. (2017). Plant exosomes: using an unconventional
Z.H., Gao, J.Q., and Ning, X.H. (2020). Engineering bacterial outer membrane ves- exit to prevent pathogen entry? J. Exp. Bot. 69, 59–68.

Molecular Therapy Vol. 29 No 1 January 2021 27


www.moleculartherapy.org

Review

26. Cai, Q., Qiao, L., Wang, M., He, B., Lin, F.-M., Palmquist, J., Huang, S.D., and Jin, H. 47. Donoso-Quezada, J., Ayala-Mar, S., and González-Valdez, J. (2020). State-of-the-art
(2018). Plants send small RNAs in extracellular vesicles to fungal pathogen to silence exosome loading and functionalization techniques for enhanced therapeutics: a re-
virulence genes. Science 360, 1126–1129. view. Crit. Rev. Biotechnol. 40, 804–820.
27. de la Canal, L., and Pinedo, M. (2018). Extracellular vesicles: a missing component in 48. Richardson, J.J., and Ejima, H. (2019). Surface engineering of extracellular vesicles
plant cell wall remodeling. J. Exp. Bot. 69, 4655–4658. through chemical and biological strategies. Chem. Mater. 31, 2191–2201.
28. Li, P., Kaslan, M., Lee, S.H., Yao, J., and Gao, Z. (2017). Progress in exosome isola- 49. Smyth, T., Petrova, K., Payton, N.M., Persaud, I., Redzic, J.S., Graner, M.W., Smith-
tion techniques. Theranostics 7, 789–804. Jones, P., and Anchordoquy, T.J. (2014). Surface functionalization of exosomes us-
29. Pérez-Bermúdez, P., Blesa, J., Soriano, J.M., and Marcilla, A. (2017). Extracellular ing click chemistry. Bioconjug. Chem. 25, 1777–1784.
vesicles in food: experimental evidence of their secretion in grape fruits. Eur. J. 50. Susa, F., Limongi, T., Dumontel, B., Vighetto, V., and Cauda, V. (2019). Engineered
Pharm. Sci. 98, 40–50. extracellular vesicles as a reliable tool in cancer nanomedicine. Cancers (Basel) 11,
30. Pocsfalvi, G., Turiák, L., Ambrosone, A., Del Gaudio, P., Puska, G., Fiume, I., 1979.
Silvestre, T., and Vékey, K. (2018). Protein biocargo of citrus fruit-derived vesicles 51. Wang, Q., Zhuang, X., Mu, J., Deng, Z.-B., Jiang, H., Zhang, L., Xiang, X., Wang, B.,
reveals heterogeneous transport and extracellular vesicle populations. J. Plant Yan, J., Miller, D., and Zhang, H.G. (2013). Delivery of therapeutic agents by nano-
Physiol. 229, 111–121. particles made of grapefruit-derived lipids. Nat. Commun. 4, 1867.
31. Xiao, J., Feng, S., Wang, X., Long, K., Luo, Y., Wang, Y., Ma, J., Tang, Q., Jin, L., Li, 52. Deng, Z., Rong, Y., Teng, Y., Mu, J., Zhuang, X., Tseng, M., Samykutty, A., Zhang, L.,
X., and Li, M. (2018). Identification of exosome-like nanoparticle-derived Yan, J., Miller, D., et al. (2017). Broccoli-derived nanoparticle inhibits mouse colitis
microRNAs from 11 edible fruits and vegetables. PeerJ 6, e5186. by activating dendritic cell AMP-activated protein kinase. Mol. Ther. 25, 1641–1654.
32. Witwer, K.W., Buzás, E.I., Bemis, L.T., Bora, A., Lässer, C., Lötvall, J., Nolte-’t Hoen, 53. György, B., Hung, M.E., Breakefield, X.O., and Leonard, J.N. (2015). Therapeutic ap-
E.N., Piper, M.G., Sivaraman, S., Skog, J., et al. (2013). Standardization of sample plications of extracellular vesicles: clinical promise and open questions. Annu. Rev.
collection, isolation and analysis methods in extracellular vesicle research. Pharmacol. Toxicol. 55, 439–464.
J. Extracell. Vesicles 2, 20360.
54. Vashisht, M., Rani, P., Onteru, S.K., and Singh, D. (2017). Curcumin encapsulated in
33. Théry, C., Amigorena, S., Raposo, G., and Clayton, A. (2006). Isolation and charac- milk exosomes resists human digestion and possesses enhanced intestinal perme-
terization of exosomes from cell culture supernatants and biological fluids. Curr. ability in vitro. Appl. Biochem. Biotechnol. 183, 993–1007.
Protoc. Cell Biol. 30, 3.22.21–3.22.29.
55. Luan, X., Sansanaphongpricha, K., Myers, I., Chen, H., Yuan, H., and Sun, D. (2017).
34. Chiou, N., and Ansel, K. (2016). Improved exosome isolation by sucrose gradient
Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta
fractionation of ultracentrifuged crude exosome pellets. Protoc. Exch. , Published
Pharmacol. Sin. 38, 754–763.
online August 23, 2016. https://doi.org/10.1038/protex.2016.057.
56. Fuhrmann, G., Serio, A., Mazo, M., Nair, R., and Stevens, M.M. (2015). Active
35. Zeringer, E., Barta, T., Li, M., and Vlassov, A.V. (2015). Strategies for isolation of
loading into extracellular vesicles significantly improves the cellular uptake and
exosomes. Cold Spring Harb. Protoc. 2015, 319–323.
photodynamic effect of porphyrins. J. Control. Release 205, 35–44.
36. Arntz, O.J., Pieters, B.C., Oliveira, M.C., Broeren, M.G., Bennink, M.B., de Vries, M.,
57. Zhang, M., Xiao, B., Wang, H., Han, M.K., Zhang, Z., Viennois, E., Xu, C., and
van Lent, P.L., Koenders, M.I., van den Berg, W.B., van der Kraan, P.M., and van de
Merlin, D. (2016). Edible ginger-derived nano-lipids loaded with doxorubicin as a
Loo, F.A. (2015). Oral administration of bovine milk derived extracellular vesicles
novel drug-delivery approach for colon cancer therapy. Mol. Ther. 24, 1783–1796.
attenuates arthritis in two mouse models. Mol. Nutr. Food Res. 59, 1701–1712.
58. Zhang, M., Wang, X., Han, M.K., Collins, J.F., and Merlin, D. (2017). Oral admin-
37. Munagala, R., Aqil, F., Jeyabalan, J., and Gupta, R.C. (2016). Bovine milk-derived
istration of ginger-derived nanolipids loaded with siRNA as a novel approach for
exosomes for drug delivery. Cancer Lett. 371, 48–61.
efficient siRNA drug delivery to treat ulcerative colitis. Nanomedicine (Lond.) 12,
38. Linares, R., Tan, S., Gounou, C., Arraud, N., and Brisson, A.R. (2015). High-speed 1927–1943.
centrifugation induces aggregation of extracellular vesicles. J. Extracell. Vesicles 4,
59. Alvarez-Erviti, L., Seow, Y., Yin, H., Betts, C., Lakhal, S., and Wood, M.J. (2011).
29509.
Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes.
39. Jasinski, D.L., Schwartz, C.T., Haque, F., and Guo, P. (2015). Large scale purification Nat. Biotechnol. 29, 341–345.
of RNA nanoparticles by preparative ultracentrifugation. Methods Mol. Biol. 1297,
67–82. 60. Wahlgren, J., De L Karlson, T., Brisslert, M., Vaziri Sani, F., Telemo, E.,
Sunnerhagen, P., and Valadi, H. (2012). Plasma exosomes can deliver exogenous
40. Gardiner, C., Di Vizio, D., Sahoo, S., Théry, C., Witwer, K.W., Wauben, M., and Hill, short interfering RNA to monocytes and lymphocytes. Nucleic Acids Res. 40, e130.
A.F. (2016). Techniques used for the isolation and characterization of extracellular
vesicles: results of a worldwide survey. J. Extracell. Vesicles 5, 32945. 61. Lamichhane, T.N., Raiker, R.S., and Jay, S.M. (2015). Exogenous DNA loading into
extracellular vesicles via electroporation is size-dependent and enables limited gene
41. van der Meel, R., Fens, M.H., Vader, P., van Solinge, W.W., Eniola-Adefeso, O., and delivery. Mol. Pharm. 12, 3650–3657.
Schiffelers, R.M. (2014). Extracellular vesicles as drug delivery systems: lessons from
the liposome field. J. Control. Release 195, 72–85. 62. Ohno, S., Takanashi, M., Sudo, K., Ueda, S., Ishikawa, A., Matsuyama, N., Fujita, K.,
Mizutani, T., Ohgi, T., Ochiya, T., et al. (2013). Systemically injected exosomes tar-
42. Loureiro, J.A., Andrade, S., Duarte, A., Neves, A.R., Queiroz, J.F., Nunes, C., Sevin, geted to EGFR deliver antitumor microRNA to breast cancer cells. Mol. Ther. 21,
E., Fenart, L., Gosselet, F., Coelho, M.A., and Pereira, M.C. (2017). Resveratrol and 185–191.
grape extract-loaded solid lipid nanoparticles for the treatment of Alzheimer’s dis-
ease. Molecules 22, 277. 63. Chevillet, J.R., Kang, Q., Ruf, I.K., Briggs, H.A., Vojtech, L.N., Hughes, S.M., Cheng,
H.H., Arroyo, J.D., Meredith, E.K., Gallichotte, E.N., et al. (2014). Quantitative and
43. Wang, X., Devaiah, S.P., Zhang, W., and Welti, R. (2006). Signaling functions of stoichiometric analysis of the microRNA content of exosomes. Proc. Natl. Acad. Sci.
phosphatidic acid. Prog. Lipid Res. 45, 250–278. USA 111, 14888–14893.
44. Liu, C., and Su, C. (2019). Design strategies and application progress of therapeutic
64. van der Pol, E., Hoekstra, A.G., Sturk, A., Otto, C., van Leeuwen, T.G., and
exosomes. Theranostics 9, 1015–1028.
Nieuwland, R. (2010). Optical and non-optical methods for detection and character-
45. Armstrong, J.P., Holme, M.N., and Stevens, M.M. (2017). Re-engineering extracel- ization of microparticles and exosomes. J. Thromb. Haemost. 8, 2596–2607.
lular vesicles as smart nanoscale therapeutics. ACS Nano 11, 69–83.
65. Mu, J., Zhuang, X., Wang, Q., Jiang, H., Deng, Z.B., Wang, B., Zhang, L., Kakar, S.,
46. Li, Z., Wang, H., Yin, H., Bennett, C., Zhang, H.G., and Guo, P. (2018). Arrowtail Jun, Y., Miller, D., and Zhang, H.G. (2014). Interspecies communication between
RNA for ligand display on ginger exosome-like nanovesicles to systemic deliver plant and mouse gut host cells through edible plant derived exosome-like nanopar-
siRNA for cancer suppression. Sci. Rep. 8, 14644. ticles. Mol. Nutr. Food Res. 58, 1561–1573.

28 Molecular Therapy Vol. 29 No 1 January 2021


www.moleculartherapy.org

Review

66. Palanisamy, V., Sharma, S., Deshpande, A., Zhou, H., Gimzewski, J., and Wong, D.T. fication based on amide and CH stretching vibrations. Biochim. Biophys. Acta
(2010). Nanostructural and transcriptomic analyses of human saliva derived exo- Biomembr. 1859, 459–466.
somes. PLoS ONE 5, e8577. 83. Benmoussa, A., Ly, S., Shan, S.T., Laugier, J., Boilard, E., Gilbert, C., and Provost, P.
67. Sharma, S., Rasool, H.I., Palanisamy, V., Mathisen, C., Schmidt, M., Wong, D.T., and (2017). A subset of extracellular vesicles carries the bulk of microRNAs in commer-
Gimzewski, J.K. (2010). Structural-mechanical characterization of nanoparticle exo- cial dairy cow’s milk. J. Extracell. Vesicles 6, 1401897.
somes in human saliva, using correlative AFM, FESEM, and force spectroscopy. 84. Jeppesen, D.K., Fenix, A.M., Franklin, J.L., Higginbotham, J.N., Zhang, Q.,
ACS Nano 4, 1921–1926. Zimmerman, L.J., Liebler, D.C., Ping, J., Liu, Q., Evans, R., et al. (2019).
68. Zhang, M.Z., Li, C., Fang, B.Y., Yao, M.H., Ren, Q.Q., Zhang, L., and Zhao, Y.D. Reassessment of exosome composition. Cell 177, 428–445.e18.
(2014). High transfection efficiency of quantum dot-antisense oligonucleotide 85. Ren, J., He, W., Zheng, L., and Duan, H. (2016). From structures to functions: in-
nanoparticles in cancer cells through dual-receptor synergistic targeting. sights into exosomes as promising drug delivery vehicles. Biomater. Sci. 4, 910–921.
Nanotechnology 25, 255102.
86. Ju, S., Mu, J., Dokland, T., Zhuang, X., Wang, Q., Jiang, H., Xiang, X., Deng, Z.B.,
69. Zhang, M.Z., Yu, R.N., Chen, J., Ma, Z.Y., and Zhao, Y.D. (2012). Targeted quantum Wang, B., Zhang, L., et al. (2013). Grape exosome-like nanoparticles induce intesti-
dots fluorescence probes functionalized with aptamer and peptide for transferrin re- nal stem cells and protect mice from DSS-induced colitis. Mol. Ther. 21, 1345–1357.
ceptor on tumor cells. Nanotechnology 23, 485104.
87. Zhang, M., Viennois, E., Prasad, M., Zhang, Y., Wang, L., Zhang, Z., Han, M.K.,
70. Zhang, M.Z., Yu, Y., Yu, R.N., Wan, M., Zhang, R.Y., and Zhao, Y.D. (2013). Xiao, B., Xu, C., Srinivasan, S., and Merlin, D. (2016). Edible ginger-derived nano-
Tracking the down-regulation of folate receptor-a in cancer cells through target spe- particles: a novel therapeutic approach for the prevention and treatment of inflam-
cific delivery of quantum dots coupled with antisense oligonucleotide and targeted matory bowel disease and colitis-associated cancer. Biomaterials 101, 321–340.
peptide. Small 9, 4183–4193.
88. Teng, Y., Ren, Y., Sayed, M., Hu, X., Lei, C., Kumar, A., Hutchins, E., Mu, J., Deng,
71. Stanly, C., Fiume, I., Capasso, G., and Pocsfalvi, G. (2016). Isolation of exosome-like Z., Luo, C., et al. (2018). Plant-derived exosomal microRNAs shape the gut micro-
vesicles from plants by ultracentrifugation on sucrose/deuterium oxide (D2O) den- biota. Cell Host Microbe 24, 637–652.e8.
sity cushions. Methods Mol. Biol. 1459, 259–269.
89. Lötvall, J., Hill, A.F., Hochberg, F., Buzás, E.I., Di Vizio, D., Gardiner, C., Gho, Y.S.,
72. Woith, E., and Melzig, M.F. (2019). Extracellular vesicles from fresh and dried Kurochkin, I.V., Mathivanan, S., Quesenberry, P., et al. (2014). Minimal experi-
plants—simultaneous purification and visualization using gel electrophoresis. Int. mental requirements for definition of extracellular vesicles and their functions: a po-
J. Mol. Sci. 20, 357. sition statement from the International Society for Extracellular Vesicles.
73. Lamparski, H.G., Metha-Damani, A., Yao, J.-Y., Patel, S., Hsu, D.-H., Ruegg, C., and J. Extracell. Vesicles 3, 26913.
Le Pecq, J.B. (2002). Production and characterization of clinical grade exosomes 90. de Gassart, A., Géminard, C., Février, B., Raposo, G., and Vidal, M. (2003). Lipid
derived from dendritic cells. J. Immunol. Methods 270, 211–226. raft-associated protein sorting in exosomes. Blood 102, 4336–4344.
74. Andriolo, G., Provasi, E., Lo Cicero, V., Brambilla, A., Soncin, S., Torre, T., Milano, 91. Théry, C., Zitvogel, L., and Amigorena, S. (2002). Exosomes: composition, biogen-
G., Biemmi, V., Vassalli, G., Turchetto, L., et al. (2018). Exosomes from human car- esis and function. Nat. Rev. Immunol. 2, 569–579.
diac progenitor cells for therapeutic applications: development of a GMP-grade 92. Kowal, J., Arras, G., Colombo, M., Jouve, M., Morath, J.P., Primdal-Bengtson, B.,
manufacturing method. Front. Physiol. 9, 1169. Dingli, F., Loew, D., Tkach, M., and Théry, C. (2016). Proteomic comparison defines
75. Pachler, K., Lener, T., Streif, D., Dunai, Z.A., Desgeorges, A., Feichtner, M., Öller, novel markers to characterize heterogeneous populations of extracellular vesicle
M., Schallmoser, K., Rohde, E., and Gimona, M. (2017). A Good Manufacturing subtypes. Proc. Natl. Acad. Sci. USA 113, E968–E977.
Practice-grade standard protocol for exclusively human mesenchymal stromal 93. Rutter, B.D., and Innes, R.W. (2017). Extracellular vesicles isolated from the leaf
cell-derived extracellular vesicles. Cytotherapy 19, 458–472. apoplast carry stress-response proteins. Plant Physiol. 173, 728–741.
76. Watson, D.C., Yung, B.C., Bergamaschi, C., Chowdhury, B., Bear, J., Stellas, D., 94. Arpin, M., Chirivino, D., Naba, A., and Zwaenepoel, I. (2011). Emerging role for
Morales-Kastresana, A., Jones, J.C., Felber, B.K., Chen, X., and Pavlakis, G.N. ERM proteins in cell adhesion and migration. Cell Adhes. Migr. 5, 199–206.
(2018). Scalable, cGMP-compatible purification of extracellular vesicles carrying
95. Schaefer, A., Nethe, M., and Hordijk, P.L. (2012). Ubiquitin links to cytoskeletal dy-
bioactive human heterodimeric IL-15/lactadherin complexes. J. Extracell. Vesicles
namics, cell adhesion and migration. Biochem. J. 442, 13–25.
7, 1442088.
96. Zhao, Z., Yu, S., Li, M., Gui, X., and Li, P. (2018). Isolation of exosome-like nano-
77. Samoil, V., Dagenais, M., Ganapathy, V., Aldridge, J., Glebov, A., Jardim, A., and
particles and analysis of microRNAs derived from coconut water based on small
Ribeiro, P. (2018). Vesicle-based secretion in schistosomes: analysis of protein
RNA high-throughput sequencing. J. Agric. Food Chem. 66, 2749–2757.
and microRNA (miRNA) content of exosome-like vesicles derived from
Schistosoma mansoni. Sci. Rep. 8, 3286. 97. Zhao, L., Gu, C., Gan, Y., Shao, L., Chen, H., and Zhu, H. (2020). Exosome-mediated
siRNA delivery to suppress postoperative breast cancer metastasis. J. Control.
78. Théry, C., Witwer, K.W., Aikawa, E., Alcaraz, M.J., Anderson, J.D.,
Release 318, 1–15.
Andriantsitohaina, R., Antoniou, A., Arab, T., Archer, F., Atkin-Smith, G.K., et al.
(2018). Minimal information for studies of extracellular vesicles 2018 98. Ha, D., Yang, N., and Nadithe, V. (2016). Exosomes as therapeutic drug carriers and
(MISEV2018): a position statement of the International Society for Extracellular delivery vehicles across biological membranes: current perspectives and future chal-
Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 7, 1535750. lenges. Acta Pharm. Sin. B 6, 287–296.

79. Smith, Z.J., Lee, C., Rojalin, T., Carney, R.P., Hazari, S., Knudson, A., Lam, K., Saari, 99. Kosaka, N., Izumi, H., Sekine, K., and Ochiya, T. (2010). microRNA as a new im-
H., Ibañez, E.L., Viitala, T., et al. (2015). Single exosome study reveals subpopula- mune-regulatory agent in breast milk. Silence 1, 7.
tions distributed among cell lines with variability related to membrane content. 100. Zhou, Q., Li, M., Wang, X., Li, Q., Wang, T., Zhu, Q., Zhou, X., Wang, X., Gao, X.,
J. Extracell. Vesicles 4, 28533. and Li, X. (2012). Immune-related microRNAs are abundant in breast milk exo-
80. Ramirez, M.I., Amorim, M.G., Gadelha, C., Milic, I., Welsh, J.A., Freitas, V.M., somes. Int. J. Biol. Sci. 8, 118–123.
Nawaz, M., Akbar, N., Couch, Y., Makin, L., et al. (2018). Technical challenges of 101. Parasramka, M.A., Ho, E., Williams, D.E., and Dashwood, R.H. (2012). MicroRNAs,
working with extracellular vesicles. Nanoscale 10, 881–906. diet, and cancer: new mechanistic insights on the epigenetic actions of phytochem-
81. Osteikoetxea, X., Balogh, A., Szabó-Taylor, K., Németh, A., Szabó, T.G., Pálóczi, K., icals. Mol. Carcinog. 51, 213–230.
Sódar, B., Kittel, Á., György, B., Pállinger, É., et al. (2015). Improved characterization 102. Baldrich, P., Rutter, B.D., Karimi, H.Z., Podicheti, R., Meyers, B.C., and Innes, R.W.
of EV preparations based on protein to lipid ratio and lipid properties. PLoS ONE (2019). Plant extracellular vesicles contain diverse small RNA species and are en-
10, e0121184. riched in 10- to 17-nucleotide “tiny” RNAs. Plant Cell 31, 315–324.
82. Mihály, J., Deák, R., Szigyártó, I.C., Bóta, A., Beke-Somfai, T., and Varga, Z. (2017). 103. Kalluri, R., and LeBleu, V.S. (2020). The biology, function, and biomedical applica-
Characterization of extracellular vesicles by IR spectroscopy: fast and simple classi- tions of exosomes. Science 367, eaau6977.

Molecular Therapy Vol. 29 No 1 January 2021 29


www.moleculartherapy.org

Review

104. Wang, B., Zhuang, X., Deng, Z.-B., Jiang, H., Mu, J., Wang, Q., Xiang, X., Guo, H., Influence of PEG coating on the biodistribution and tumor accumulation of pH-
Zhang, L., Dryden, G., et al. (2014). Targeted drug delivery to intestinal macro- sensitive liposomes. Drug Deliv. Transl. Res. 9, 123–130.
phages by bioactive nanovesicles released from grapefruit. Mol. Ther. 22, 522–534. 124. Garay, R.P., El-Gewely, R., Armstrong, J.K., Garratty, G., and Richette, P. (2012).
105. Chen, Q., and Lai, H. (2013). Plant-derived virus-like particles as vaccines. Hum. Antibodies against polyethylene glycol in healthy subjects and in patients treated
Vaccin. Immunother. 9, 26–49. with PEG-conjugated agents. Expert Opin. Drug Deliv. 9, 1319–1323.
106. Pillet, S., Couillard, J., Trépanier, S., Poulin, J.-F., Yassine-Diab, B., Guy, B., Ward, 125. Abu Lila, A.S., Kiwada, H., and Ishida, T. (2013). The accelerated blood clearance
B.J., and Landry, N. (2019). Immunogenicity and safety of a quadrivalent plant- (ABC) phenomenon: clinical challenge and approaches to manage. J. Control.
derived virus like particle influenza vaccine candidate—two randomized phase II Release 172, 38–47.
clinical trials in 18 to 49 and 350 years old adults. PLoS ONE 14, e0216533. 126. Vader, P., Mol, E.A., Pasterkamp, G., and Schiffelers, R.M. (2016). Extracellular ves-
107. Efferth, T., Li, P.C., Konkimalla, V.S.B., and Kaina, B. (2007). From traditional icles for drug delivery. Adv. Drug Deliv. Rev. 106 (Pt A), 148–156.
Chinese medicine to rational cancer therapy. Trends Mol. Med. 13, 353–361. 127. Rupert, D.L.M., Claudio, V., Lässer, C., and Bally, M. (2017). Methods for the phys-
108. Ingenito, F., Roscigno, G., Affinito, A., Nuzzo, S., Scognamiglio, I., Quintavalle, C., ical characterization and quantification of extracellular vesicles in biological sam-
and Condorelli, G. (2019). The role of exo-miRNAs in cancer: a focus on therapeutic ples. Biochim. Biophys. Acta, Gen. Subj. 1861 (1 Pt A), 3164–3179.
and diagnostic applications. Int. J. Mol. Sci. 20, 4687. 128. Smyth, T.J., Redzic, J.S., Graner, M.W., and Anchordoquy, T.J. (2014). Examination
109. Deng, S.-Z., Lai, M.-F., Li, Y.-P., Xu, C.-H., Zhang, H.-R., and Kuang, J.-G. (2020). of the specificity of tumor cell derived exosomes with tumor cells in vitro. Biochim.
Human marrow stromal cells secrete microRNA-375-containing exosomes to regu- Biophys. Acta 1838, 2954–2965.
late glioma progression. Cancer Gene Ther. 27, 203–215. 129. Schuh, C.M.A.P., Cuenca, J., Alcayaga-Miranda, F., and Khoury, M. (2019).
Exosomes on the border of species and kingdom intercommunication. Transl.
110. Niu, J., Chu, Y., Huang, Y.-F., Chong, Y.-S., Jiang, Z.-H., Mao, Z.-W., Peng, L.H., and
Res. 210, 80–98.
Gao, J.Q. (2017). Transdermal gene delivery by functional peptide-conjugated
cationic gold nanoparticle reverses the progression and metastasis of cutaneous mel- 130. Wang, Q., Ren, Y., Mu, J., Egilmez, N.K., Zhuang, X., Deng, Z., Zhang, L., Yan, J.,
anoma. ACS Appl. Mater. Interfaces 9, 9388–9401. Miller, D., and Zhang, H.G. (2015). Grapefruit-derived nanovectors use an activated
leukocyte trafficking pathway to deliver therapeutic agents to inflammatory tumor
111. Liu, Y.-C., Chen, W.L., Kung, W.-H., and Huang, H.-D. (2017). Plant miRNAs
sites. Cancer Res. 75, 2520–2529.
found in human circulating system provide evidences of cross kingdom RNAi.
BMC Genomics 18 (Suppl 2 ), 112. 131. Sun, D., Zhuang, X., Xiang, X., Liu, Y., Zhang, S., Liu, C., Barnes, S., Grizzle, W.,
Miller, D., and Zhang, H.G. (2010). A novel nanoparticle drug delivery system:
112. Raimondo, S., Naselli, F., Fontana, S., Monteleone, F., Lo Dico, A., Saieva, L., Zito,
the anti-inflammatory activity of curcumin is enhanced when encapsulated in exo-
G., Flugy, A., Manno, M., Di Bella, M.A., et al. (2015). Citrus limon-derived nano-
somes. Mol. Ther. 18, 1606–1614.
vesicles inhibit cancer cell proliferation and suppress CML xenograft growth by
inducing TRAIL-mediated cell death. Oncotarget 6, 19514–19527. 132. Shandilya, S., Rani, P., Onteru, S.K., and Singh, D. (2017). Small interfering RNA in
milk exosomes is resistant to digestion and crosses the intestinal barrier in vitro.
113. Sato, T., van Es, J.H., Snippert, H.J., Stange, D.E., Vries, R.G., van den Born, M., J. Agric. Food Chem. 65, 9506–9513.
Barker, N., Shroyer, N.F., van de Wetering, M., and Clevers, H. (2011). Paneth cells
constitute the niche for Lgr5 stem cells in intestinal crypts. Nature 469, 415–418. 133. Osteikoetxea, X., Sódar, B., Németh, A., Szabó-Taylor, K., Pálóczi, K., Vukman,
K.V., Tamási, V., Balogh, A., Kittel, Á., Pállinger, É., and Buzás, E.I. (2015).
_ Yıldırım, E., and Kala, E.Y. (2019).
114. xSahin, F., Koçak, P., Güneş, M.Y., Özkan, I., Differential detergent sensitivity of extracellular vesicle subpopulations. Org.
In vitro wound healing activity of wheat-derived nanovesicles. Appl. Biochem. Biomol. Chem. 13, 9775–9782.
Biotechnol. 188, 381–394.
134. LaVan, D.A., McGuire, T., and Langer, R. (2003). Small-scale systems for in vivo
115. Zhuang, X., Deng, Z.-B., Mu, J., Zhang, L., Yan, J., Miller, D., Feng, W., McClain, drug delivery. Nat. Biotechnol. 21, 1184–1191.
C.J., and Zhang, H.G. (2015). Ginger-derived nanoparticles protect against
135. Gu, T.W., Wang, M.Z., Niu, J., Chu, Y., Guo, K.R., and Peng, L.H. (2020). Outer
alcohol-induced liver damage. J. Extracell. Vesicles 4, 28713.
membrane vesicles derived from E. coli as novel vehicles for transdermal and tumor
116. Samanta, S., Rajasingh, S., Drosos, N., Zhou, Z., Dawn, B., and Rajasingh, J. (2018). targeting delivery. Nanoscale 12, 18965–18977.
Exosomes: new molecular targets of diseases. Acta Pharmacol. Sin. 39, 501–513.
136. Li, N., Peng, L.-H., Chen, X., Nakagawa, S., and Gao, J.-Q. (2011). Transcutaneous
117. Schillaci, O., Fontana, S., Monteleone, F., Taverna, S., Di Bella, M.A., Di Vizio, D., vaccines: novel advances in technology and delivery for overcoming the barriers.
and Alessandro, R. (2017). Exosomes from metastatic cancer cells transfer amoeboid Vaccine 29, 6179–6190.
phenotype to non-metastatic cells and increase endothelial permeability: their
137. Hoffman, R.M. (1998). Topical liposome targeting of dyes, melanins, genes, and
emerging role in tumor heterogeneity. Sci. Rep. 7, 4711.
proteins selectively to hair follicles. J. Drug Target. 5, 67–74.
118. Çagdaş, M., Sezer, A.D., and Bucak, S. (2014). Liposomes as potential drug carrier 138. Toll, R., Jacobi, U., Richter, H., Lademann, J., Schaefer, H., and Blume-Peytavi, U.
systems for drug delivery. In Application of Nanotechnology in Drug Delivery, (2004). Penetration profile of microspheres in follicular targeting of terminal hair
A.D. Sezer, ed. (London: IntechOpen), pp. 1–100. follicles. J. Invest. Dermatol. 123, 168–176.
119. Wang, M.-Z., Niu, J., Ma, H.-J., Dad, H.A., Shao, H.-T., Yuan, T.-J., and Peng, L.H. 139. Lu, M., Zhao, X., Xing, H., Xun, Z., Zhu, S., Lang, L., Yang, T., Cai, C., Wang, D., and
(2020). Transdermal siRNA delivery by pH-switchable micelles with targeting effect Ding, P. (2018). Comparison of exosome-mimicking liposomes with conventional
suppress skin melanoma progression. J. Control. Release 322, 95–107. liposomes for intracellular delivery of siRNA. Int. J. Pharm. 550, 100–113.
120. Fujita, D., Arai, T., Komori, H., Shirasaki, Y., Wakayama, T., Nakanishi, T., and 140. Li, N., Peng, L.-H., Chen, X., Zhang, T.-Y., Shao, G.-F., Liang, W.-Q., and Gao, J.Q.
Tamai, I. (2018). Apple-derived nanoparticles modulate expression of organic- (2014). Antigen-loaded nanocarriers enhance the migration of stimulated
anion-transporting polypeptide (OATP) 2B1 in Caco-2 cells. Mol. Pharm. 15, Langerhans cells to draining lymph nodes and induce effective transcutaneous im-
5772–5780. munization. Nanomedicine (Lond.) 10, 215–223.
121. Fan, Y., and Zhang, Q. (2013). Development of liposomal formulations: from 141. Ciotti, S.N., and Weiner, N. (2002). Follicular liposomal delivery systems.
concept to clinical investigations. Asian J. Pharm. Sci. 8, 81–87. J. Liposome Res. 12, 143–148.
122. Theek, B., Baues, M., Ojha, T., Möckel, D., Veettil, S.K., Steitz, J., van Bloois, L., 142. Jung, S., Otberg, N., Thiede, G., Richter, H., Sterry, W., Panzner, S., and Lademann,
Storm, G., Kiessling, F., and Lammers, T. (2016). Sonoporation enhances liposome J. (2006). Innovative liposomes as a transfollicular drug delivery system: penetration
accumulation and penetration in tumors with low EPR. J. Control. Release 231, into porcine hair follicles. J. Invest. Dermatol. 126, 1728–1732.
77–85.
143. Yepes-Molina, L., Martínez-Ballesta, M.C., and Carvajal, M. (2020). Plant plasma
123. Nunes, S.S., Fernandes, R.S., Cavalcante, C.H., da Costa César, I., Leite, E.A., Lopes, membrane vesicles interaction with keratinocytes reveals their potential as carriers.
S.C.A., Ferretti, A., Rubello, D., Townsend, D.M., de Oliveira, M.C., et al. (2019). J. Adv. Res. 23, 101–111.

30 Molecular Therapy Vol. 29 No 1 January 2021


www.moleculartherapy.org

Review

144. Peng, L.-H., Zhang, Y.-H., Han, L.-J., Zhang, C.-Z., Wu, J.-H., Wang, X.-R., Gao, particles as unique non-viral vectors for gene delivery to mesenchymal stem cells
J.Q., and Mao, Z.W. (2015). Cell membrane capsules for encapsulation of chemo- with antibacterial activity. Biomaterials 103, 137–149.
therapeutic and cancer cell targeting in vivo. ACS Appl. Mater. Interfaces 7,
150. Mathieu, M., Martin-Jaular, L., Lavieu, G., and Théry, C. (2019). Specificities of
18628–18637.
secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell
145. Sercombe, L., Veerati, T., Moheimani, F., Wu, S.Y., Sood, A.K., and Hua, S. communication. Nat. Cell Biol. 21, 9–17.
(2015). Advances and challenges of liposome assisted drug delivery. Front.
Pharmacol. 6, 286. 151. Costa Verdera, H., Gitz-Francois, J.J., Schiffelers, R.M., and Vader, P. (2017).
Cellular uptake of extracellular vesicles is mediated by clathrin-independent endo-
146. Amaro, M.I., Rocha, J., Vila-Real, H., Eduardo-Figueira, M., Mota-Filipe, H.,
cytosis and macropinocytosis. J. Control. Release 266, 100–108.
Sepodes, B., et al. (2009). Anti-inflammatory activity of naringin and the bio-
synthesised naringenin by naringinase immobilized in microstructured materials 152. Kalaydina, R.-V., Bajwa, K., Qorri, B., Decarlo, A., and Szewczuk, M.R. (2018).
in a model of DSS-induced colitis in mice. Food Res. Int. 42, 1010–1017. Recent advances in “smart” delivery systems for extended drug release in cancer
therapy. Int. J. Nanomedicine 13, 4727–4745.
147. Dou, W., Zhang, J., Sun, A., Zhang, E., Ding, L., Mukherjee, S., Wei, X., Chou, G.,
Wang, Z.T., and Mani, S. (2013). Protective effect of naringenin against experi- 153. Peng, L.-H., Niu, J., Zhang, C.-Z., Yu, W., Wu, J.-H., Shan, Y.-H., Wang, X.R., Shen,
mental colitis via suppression of Toll-like receptor 4/NF-kB signalling. Br. J. Nutr. Y.Q., Mao, Z.W., Liang, W.Q., and Gao, J.Q. (2014). TAT conjugated cationic noble
110, 599–608. metal nanoparticles for gene delivery to epidermal stem cells. Biomaterials 35, 5605–
148. Yin, H., Kanasty, R.L., Eltoukhy, A.A., Vegas, A.J., Dorkin, J.R., and Anderson, D.G. 5618.
(2014). Non-viral vectors for gene-based therapy. Nat. Rev. Genet. 15, 541–555. 154. Abrami, L., Brandi, L., Moayeri, M., Brown, M.J., Krantz, B.A., Leppla, S.H., and van
149. Peng, L.-H., Huang, Y.-F., Zhang, C.-Z., Niu, J., Chen, Y., Chu, Y., Jiang, Z.H., Gao, der Goot, F.G. (2013). Hijacking multivesicular bodies enables long-term and exo-
J.Q., and Mao, Z.W. (2016). Integration of antimicrobial peptides with gold nano- some-mediated long-distance action of anthrax toxin. Cell Rep. 5, 986–996.

Molecular Therapy Vol. 29 No 1 January 2021 31

You might also like