La terra è un bel posto peccato ci siano gli umani...
The earth is a beautiful place, too bad there are humans...
Source:
https://pubs.acs.org/doi/10.1021/acsnano.0c06109
Engineering Antiviral Vaccines
Xingwu Zhou,● Xing Jiang,● Moyuan Qu,● George E. Aninwene, II, Vadim Jucaud, James J. Moon,
Zhen Gu, Wujin Sun,* and Ali Khademhosseini*
Cite This: ACS Nano 2020, 14, 12370−12389 Read Online
ACCESS Metrics & More Article Recommendations
ABSTRACT: Despite the vital role of vaccines in fighting viral pathogens, effective
vaccines are still unavailable for many infectious diseases. The importance of vaccines
cannot be overstated during the outbreak of a pandemic, such as the coronavirus disease
2019 (COVID-19) pandemic. The understanding of genomics, structural biology, and
innate/adaptive immunity have expanded the toolkits available for current vaccine
development. However, sudden outbreaks and the requirement of population-level
immunization still pose great challenges in today’s vaccine designs. Well-established
vaccine development protocols from previous experiences are in place to guide the
pipelines of vaccine development for emerging viral diseases. Nevertheless, vaccine
development may follow different paradigms during a pandemic. For example, multiple
vaccine candidates must be pushed into clinical trials simultaneously, and manufacturing
capability must be scaled up in early stages. Factors from essential features of safety,
efficacy, manufacturing, and distributions to administration approaches are taken into
consideration based on advances in materials science and engineering technologies. In
this review, we present recent advances in vaccine development by focusing on vaccine discovery, formulation, and delivery
devices enabled by alternative administration approaches. We hope to shed light on developing better solutions for faster and
better vaccine development strategies through the use of biomaterials, biomolecular engineering, nanotechnology, and
microfabrication techniques.
KEYWORDS: COVID-19, pandemics, infectious disease, vaccine, immunotherapy, drug discovery, drug delivery, biomedical devices
T
he appearance of severe acute respiratory syndrome-
coronavirus 2 (SARS-CoV-2) has caused unprece-
dented global disruption to health, economy, and
social stability.1 SARS-CoV-2 causes coronavirus disease 2019
(COVID-19) in which individuals can suffer from mild to
severe symptoms once infected.2 Global scientific communities
are working together to fight the crisis caused by this
pandemic. The initial efforts to combat this pandemic included
identifying infected patients and ramping up clinical trials for
repurposing existing drugs.3 However, the development of
effective vaccines to prevent SARS-CoV-2 infection is believed
by many to be the most effective long-term solution.4
The pathway from identifying a virus to having a vaccine is
lengthy and expensive. After billions of dollars spent and
multiple years of trials, researchers generally face high rates of
failure in the traditional vaccine development paradigms
(Figure 1a).4 Despite rapid responses from the scientific
community and the pharmaceutical industry, vaccines have
historically not been ready even after an epidemic becomes
manageable, such as severe acute respiratory syndrome
(SARS), Zika, and Ebola.5 Considering economic factors,
funding for vaccine development may be reallocated after a
substantial drop in infection cases is observed, as in the 2015−
2016 Zika and SARS epidemics. The 2014−2016 Ebola
outbreak in Africa resulted in over 28,000 cases and 11,000
deaths.6 After continuous research funding support, in
December 2019, the FDA granted the first approval to Ervebo
(a single-dose, live attenuated virus vaccine from Merck) for
Ebola virus prevention.7 Benefiting from egg- and cell-based
platforms, vaccine approval for the most recent 2009 H1N1
pandemic was relatively fast following the peak of the outbreak,
and the vaccine was ultimately incorporated in the seasonal
influenza vaccine.8 Since the initiation and ending of epidemics
are highly unpredictable, the rapid deployment of vaccine
development may still be too slow for a sudden outbreak.4,5
To tackle a pandemic, a different vaccine development
process is needed to rapidly produce safe and effective vaccines
against novel viruses. This process is characterized by
overlapping phases, multiple potential vaccine candidates,
Received: July 21, 2020
Accepted: September 18, 2020
Published: October 1, 2020
Review
www.acsnano.org
© 2020 American Chemical Society
12370
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
This article is made available via the ACS COVID-19 subset for unrestricted RESEARCH re-use
and analyses in any form or by any means with acknowledgement of the original source.
These permissions are granted for the duration of the World Health Organization (WHO)
declaration of COVID-19 as a global pandemic.
Downloaded via 95.145.163.132 on November 9, 2022 at 06:37:05 (UTC).
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
and early ramp-up of manufacturing capacities (Figure 1a).4
After the initial release of the genetic sequence of SARS-CoV-
2, researchers worldwide started to develop vaccines for the
prevention of COVID-19. The first dose was administered into
humans by Moderna Inc. on March 16, 2020.9 This method
utilized lipid nanoparticles (LNPs) with a formulated mRNA
vaccine.9 This work was fast-tracked at an unprecedented pace,
even though mRNA-based vaccines have never been licensed
before.9 A snapshot of the landscape of COVID-19 vaccines
(Figure 1b) shows that around 31 vaccines have entered
clinical trials as of August 25, 2020.10 A high diversity of
vaccine candidates is seen in these forerunners, which includes
six mRNA-based, five inactivated virus-based, five nonreplicat-
ing viral vector-based, one replicating viral vector-based, four
DNA-based, and one cell culture-based vaccines as well as nine
protein-based vaccines (Figure 1b,c). The incomplete under-
standing of the SARS-CoV-2 interactions with the human
immune system became one of the major obstacles in vaccine
development. A recent report on a second COVID-19
infection in the same patient further raises the question of
protective immunity and immune memory. For instance, it is
still unknown how durable the immune memory response
induced by SARS-CoV-2 could be or what is the threshold of
antibodies titers that can protect the patients against
reinfection.11,12 In addition, protective immunity against
SARS-CoV-2 should also be comprehensively studied, where
the balance of cellular and humoral immunity, the ratio of
effector to memory T cells, the maintenance of memory B
cells, and functional features of activated T cells should be
characterized.11,12 These aspects could ultimately facilitate
vaccine designs and evaluations.
In this review, we aim to provide engineering insights on
vaccine development by covering vaccine discovery, vaccine
formulations in terms of different materials (lipid, polymer, and
inorganic particles), and vaccine delivery devices. We will
present how some of the existing challenges could potentially
be addressed by nano/micro/macroscale engineering ap-
proaches. We will also discuss some emerging technologies
that may contribute to future vaccine development pipelines.
VACCINE DISCOVERY
After a virus is identified, different categories of vaccines can be
developed, which includes virus-, viral vector-, nucleic acid-,
protein/peptide-, and/or cell-based vaccines (Figure 1c).13
Virus-based vaccines commonly require patient-derived
viruses, while other types can utilize the viral genomic
sequences to accelerate vaccine development. Previously
approved vaccines mostly fall into the virus-based and the
recombinant protein-based categories, but not into the nucleic
acid-based one (Figure 2a).14 The discovery process of each
vaccine type is distinct with corresponding pros and cons
(Figure 2b−d).
Virus-Based Vaccines. Virus-based vaccines, including the
weakened and inactivated types, are still the most widely used
to date. The effective prevention of the polio epidemic by
vaccination in the 20th century represented a landmark success
in medical history.18 This vaccination regimen included the use
of the inactivated polio vaccine (IPV) and oral polio vaccine
Figure 1. Summary of vaccine development paradigms and major types of vaccines. (a) Paradigms of vaccine development in a traditional
condition vs during a pandemic.5 (b) Snapshot of the vaccine landscape for COVID-19 with the different types of vaccine candidates in
various clinical stages as of August 25, 2020.10 (c) Schematic of major types of vaccines, including virus-based (weakened and inactivated),
viral vector-based (replicating and nonreplicating), nucleic acid-based (DNA and mRNA), protein and peptide-based, and cell-based.13
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12371
(OPV).18 The IPV was made by inactivating lab-cultured
poliovirus by formalin. The OPV was weakened live polio-
virus.18 Weakened viruses are highly immunogenic and are
commonly developed by the deletion of viral genes. Never-
theless, mutations could cause virulence reversion as observed
for vaccine-derived polioviruses that have been responsible for
recent polio outbreaks.19 This represents an obstacle for polio
eradication and, more importantly, a concern for the weakened
virus-based vaccines in general. Further genetic manipulation
strategies are therefore required to prevent this issue. For
instance, modifying the 5′ untranslated region (UTR), 2C
coding region, and 3D polymerase region resulted in a
genetically stabilized OPV strain that was less likely to regain
virulence and exhibited limited viral adaptability.20
On the contrary, inactivated virus vaccines generally do not
have virulence reversion issues. They also do not rely on
extensive genetic manipulations, but the inactivation process
may cause the loss of antigenicity of viral immunogens. This
vaccine type has been quickly adapted to cope with COVID-
19, where patient-derived viruses were inactivated with β-
propiolactone, followed with purification by chromatography,
and formulated with Al(OH)3 as the adjuvant.15 The vaccine
protected nonhuman primates from SARS-CoV-2 challenges
with no notable side effects.15 Advantages of these virus-based
methods include well-developed methodologies, well-estab-
lished regulation policies, and existing manufacturing facilities
from previous practices. Nevertheless, the requirement of virus
seeding and culture along with unknown safety issues
Figure 2. Vaccine discovery processes for different types of vaccines. (a) Timeline of previously approved antiviral vaccines.14 (b) Virus-
based vaccines require the seeding and culture of specific viruses derived from patients. By manipulating the genetic sequences of the virus
or inactivating the virus directly with chemicals, weakened and inactivated virus vaccines can be produced.15 (c) Viral vector-based and
nucleic acid-based vaccines are independent of virus culture and rely on the genetic sequence of the virus and/or the selection of
immunogenic sequences of the virus. By selecting commonly developed vectors, virus-specific sequences can be inserted.16 Nucleic acid
vaccines require further formulations for optimal efficacy. (d) Structure-based understanding of native viral proteins can validate the
expressed recombinant proteins and predict the peptide sequences desirable as vaccines. Protein/peptide vaccines require further
formulations for optimal efficacy.17
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12372
necessitates alternative practices for the development of other
vaccines.
Viral Vector-Based Vaccines. Viral vector-based vaccines
work by inserting viral antigen-encoding DNA sequences into
host cells, which leads to the expression of viral antigens by the
host cells.16 The rationale is based on the capability of viruses
to infect cells efficiently through viral genome integration
mechanisms.16 This method could also be problematic since
the genome of the host could be altered and cause other
diseases. The selection of viruses is therefore crucial in
balancing the efficacy (efficient infectivity) and safety (limited
pathogenicity). Another common obstacle is the host’s pre-
existing immunity to certain vectors that would mitigate the
efficacy of a vaccine. The selected viral vectors could be further
genetically modified to be nonreplicable by disrupting genes
responsible for replication. Adenoviruses (Ad) are the most
extensively used vectors for vaccines because of several
advantages: infectious to various cell types, efficient transgene
expression, high in vitro growth, lack of genome integration,
and genetic stability.21 For instance, Johnson & Johnson (J &
J) deployed the Ad26 vector to deliver gene encoding surface
proteins of the SARS-CoV-2, and CanSino Biological used the
Ad5 vector.22 The spike (S) glycoprotein of SARS-CoV-2 was
cloned with the tissue plasminogen activator signal peptide
gene in the early region 1 (E1) and early region 3 (E3) deleted
Ad5 vector. The E1 and E3 deletions render the Ad5 vector
nonreplicating. Even though rapid humoral and T cell
responses were induced by the vaccine, pre-existing anti-Ad5
immunity partially diminished the immune responses among
healthy adults.22 To address the pre-existing immunity to Ad,
chimpanzee Ad (ChAdOX1) represents an alternative solution
that provided broad protective immunity against the Middle
East Respiratory Syndrome Coronavirus (MERS-CoV).23 An
investigational vaccine for SARS-CoV-2 from the University of
Oxford is based on this vector.23 Their preclinical study
showed the prevention of SARS-CoV-2 pneumonia in rhesus
macaques with no evidence of subsequent immune-enhanced
diseases.23 However, viral loads were more significantly
reduced in the lower respiratory tract but not in the nasal
swabs. In general, because of previous vaccine programs, the
manufacturing capacity and protocols for this method are
already well-established; therefore, less effort may be needed to
shift toward the large-scale production of vector-based
vaccines. However, both the pre-existing immunity toward
certain vectors and previously demonstrated safety concerns
over the increased risk of human immunodeficient virus type 1
(HIV-1) acquisition should be carefully evaluated.22
Nucleic Acid-Based Vaccines. DNA- and mRNA-based
vaccines have the greatest potential for rapid development
because of their synthetic nature that circumvents the need for
cell culture or fermentation of viruses. The synthetic nature of
this method also results in a high safety profile and rapid
manufacturing ability.24 However, this method generally
requires further delivery formulations or better delivery devices
to boost vaccine efficacy.24 In contrast to RNA, the increased
stability of DNA decreases the need for frozen storage and low-
temperature transportation.24 One of the forerunners among
COVID-19 vaccines is DNA-based (Inovio).10 Similar DNA-
based vaccines have had promising results for the prevention of
MERS.25 In the discovery phase of DNA-based vaccines,
strategies such as codon/RNA optimization and the addition
of highly efficient immunoglobulin leader sequences are
needed to enhance the magnitude and breadth of the immune
response.26 For instance, Muthumani et al. designed a
consensus DNA sequence for the S protein by analyzing S
protein sequences, where sequences from clades A and B can
be both involved.26 To enhance the in vivo expression, the
immunoglobulin E (IgE) leader sequence was added to the
immunogen sequence. The insert was further incorporated into
the pVax1 vector, which allowed a high-level transient
expression of proteins of interest in mammalian cells.26 The
strategies of adding the IgE leader sequence and the use of the
pVax1 vector could be quickly adapted to the different virus
sequences as well.
Due to their high potency, rapid development, and low cost
for manufacturing, mRNA vaccines have emerged as a
promising alternative vaccination development strategy for
various infectious diseases and cancers.27 No mRNA-based
vaccine is currently FDA-approved. Several strategies have
been proposed to solve the common drawbacks of mRNA-
based vaccines, which include mRNA instability and high
innate immunogenicity.28 The 5′ and 3′ UTR of mRNA has
significant regulatory influences on both stability and trans-
latability.28 Half-life and expression can be enhanced when
these sequences come from viral genes: A 5′ cap is essential for
efficient protein expression, and a poly(A) tail affects mRNA
translation and stability.29 Furthermore, the replacement of
rare codons with frequently used synonymous codons also
enhances protein expression.30 Additional optimization strat-
egies include increasing G:C content and introducing modified
nucleosides.31,32 In spite of the innate immunostimulatory
effect of exogenous or unpurified mRNA, further investigations
are required to assess the advantages of their use for vaccines
specifically.33 Innate immune sensing mechanisms of mRNA
have resulted in the inhibition of antigen expression and a
decreased immune response.34 Therefore, further formulations
for vaccine delivery could potentially reduce innate mRNA-
associated immunogenicity and favor immune activation.
Subunit Protein/Peptide Vaccines. Unlike nucleic acid-
based vaccines, which produce viral protein after admin-
istration, the direct application of viral protein antigens may be
a more straightforward method to trigger immunity. This
approach currently represents the largest category of COVID-
19 vaccines under preclinical investigation.4,10 Full-length
proteins are advantageous because they can induce the
development of antibodies against multiple epitopes. Impor-
tantly, there is a higher probability that the developed
antibodies could bind to the native conformation of the viral
protein. However, this also increases the chance of inducing
nonspecific cross-reactive antibodies.35 In addition to the high
cost of recombinant protein technologies, the recombinant
protein products may not fully match all the molecular features
of the corresponding viral proteins, which may lead to
ineffective induction of broad neutralizing antibodies
(bNAb).35 Optimizing peptide sequences in the discovery
phase can improve the stabilization of epitopes associated with
bNAb while reducing the antigenicity of non-NAb epitopes.17
Given that only specific amino acid sequences of the full-
length protein antigens are responsible for effective immune
responses, minimally immunogenic peptides, mimicking B and
T cell epitopes, have been proposed for vaccines.17 Early
approaches used short peptide sequences (8−10 amino acids)
that could potentially be presented by major histocompatibility
complex (MHC) I molecules. Importantly, the memory
immune response of cytotoxic T cells could not be efficiently
induced with such T cell epitope-mimicking peptides.36 One
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12373
proposed rationale is that in vivo direct loading of short
peptides occurs for all cells expressing MHC molecules,
including T and B cells. Unlike DCs, they are incapable of
generating an effective immune response.37 These early
findings benefitted the design of synthetic long peptide
(SLP) vaccines where both helper T cells (TH) peptides and
toll-like receptor (TLR) ligand or peptides could be hybridized
to enhance the immune response. For example, Jackson et al.
demonstrated one single SLP vaccine that can target TLR 2
can serve as self-adjuvant and contain both MHC I- and II-
specific peptides (T cell epitopes).38 SLPs cannot directly bind
with MHC I and have to be internalized by DCs for further
presentation. Furthermore, in silico computer-based ap-
proaches can be used to benefit the discovery of peptide
vaccines by enabling quicker screening and more accurate
predictions of peptide sequences with high immunogenicity
and binding affinities to MHC I and II molecules.39 These
approaches have been used to accelerate the development
pipeline of vaccines, and particularly for emerging viral diseases
such as COVID-19. These computational approaches can
predict the binding affinity of specific peptides sequences to
either MHC I and II molecules or B cell receptors/antibodies.
In this regard, theoretical B and T cell epitopes specific to
SARS-CoV-2 were synthesized to generate a multiepitope
protein.39 The antigenicity, allergenicity, physiochemical
parameter, and secondary and tertiary structures of these
multiepitope proteins were determined in silico. However, the
processing in the endosomal/lysosomal compartments of the
Figure 3. Action mechanisms of vaccine formulations. (a) Vaccines generate humoral and cellular immunity within lymph nodes (LNs): I.
DCs can process the antigens and present the peptide fragments via both MHC class I and class II molecules. II. B cells can directly
recognize the antigens via BCRs and present the antigenic peptide fragments by MHC class II to helper T cells (CD4+). Stimulated B cells
can subsequently initiate a humoral immune response. III. Cytotoxic T cells (CD8+) can recognize the antigenic peptide fragments presented
by MHC class I through TCRs and trigger the cellular immune response.12,45 (b) Intracellular response of DCs to antigen presentation for
different types of vaccines through PRRs. I. Vaccine formulations can be effectively internalized by cells, followed by II. Endosomal release.
Before III. MHC loading of antigen peptide, peptide vaccine undergoes enzymatic processing, DNA vaccine undergoes transcription and
translation, and mRNA vaccine undergoes translation.24,27,37
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12374
designed multiepitope SARS-CoV-2 antigens remains to be
addressed.
Cell-Based Vaccines. DCs are the most potent antigen-
presenting cells (APCs) at the interface of innate and adaptive
immunity to sense the “danger signals”, process the antigens,
and present the nonself-antigens.40 Unlike other types of
vaccines, DC vaccines are manipulated ex vivo to be antigen-
specific, and thus they are ready to initiate immune responses
after being reinfused in vivo. The DC vaccine is quality
controllable and natively able to traffic efficiently in vivo.
Different methodologies have been developed to produce
modified DC vaccines ex vivo, including loading DCs with
immunogenic peptides or proteins (subunit DCs), viral
transduction to express immunogenic peptides,41 or using
mRNA-based DCs.42 Different strategies may produce DC
vaccines with different potency, for instance, genetically
modified DCs may be more efficacious in immune activation
than subunit DC vaccines.41 In addition, supplementary
immunomodulatory genes could be incorporated to enhance
the potency. However, the process of producing cell-based
vaccines is labor-intensive and expensive.43 Thus, this method
could be the most challenging to be scaled up as a universal
vaccine for a global pandemic.43
VACCINE FORMULATIONS
Action Mechanisms. Vaccines are commonly adminis-
tered intramuscularly, making it less efficient in interacting
with the abundant immune cells present within the skin.
Alternative strategies, including oral or intranasal deliveries, are
also widely studied because of their ease of use.44 The
effectiveness of alternative strategies is based on mucosal
immunity.44 Nonetheless, different strategies rely on the
activation of the adaptive immune system, which involves the
interactions between T cells and APCs (DCs and B cells).12 As
shown in Figure 3a, APCs can internalize exogenous antigens
and process them into antigenic peptide fragments. The
peptide fragments can subsequently be loaded onto the MHC
II molecules and displayed on the surface of the APCs.12,45
CD4+ helper T cells could recognize peptides presented by
MHC class II through T cell receptors (TCRs), which could
induce the production of cytokines, such as IL-2, that are
essential to the normal function of immune cells.12,45 B cells
can directly capture antigens through B-cell receptors (BCRs).
The antigens can be internalized and subsequently processed
into antigenic peptides for presentation by MHC class II to
CD4+ helper T cells. This process is required for the clonal
expansion and differentiation of B cells into IgG antibody-
secreting plasma cells and memory B cells, which is classified as
humoral immunity.12,45 Generally, only endogenous antigens
could be degraded into peptides and loaded onto MHC class I
molecules for presentation to CD8+ T cells.12 Cross-
presentation is the ability of certain APCs to uptake, process,
and present extracellular antigens in an MHC class I-
dependent manner to CD8+ T cells. Cross-presentation can
be achieved by several cell types, including DCs (the primary
cell type), neutrophils, macrophages, and endothelial cells.12
The MHC class I presented peptide fragments can activate
CD8+ cytotoxic T cells through TCR recognition. Antigen-
specific CD8+ cytotoxic T cells can then recognize these
peptide fragments presented by target cells and kill them,
which refers to cellular immunity.12,45 Lastly, DCs are also
capable of detecting “danger signals” via an array of receptors
called pattern recognition receptors (PRRs). PRRs are capable
of recognizing pathogenic molecules as part of the innate
immunity for a response to virus infection by identifying
pathogen-associated molecular patterns (PAMPs) from viruses
or microbes. The understanding of innate immunity could
benefit the development of adjuvants to enhance the immune
response further. Together, the understanding of the action
mechanisms facilitates the design of vaccine formulations to
boost their efficacy.46
Requirement of Formulations. Based on the current
understanding of eliciting optimal immune responses, materi-
Figure 4. Materials-based vaccine formulations for important immunological functions. (a) Vaccine formulations that protect the active
“cargo” and enhance cellular uptake may be based on lipids, polymeric materials, and/or inorganic particles. (b) Vaccine formulations that
allow LN targeting by forming extra small-sized albumin-based vaccine complexes, tuning the net charge to be negative and incorporating
targeting ligands. (c) Vaccine formulations that promote immunostimulatory effects by using microbial wall-derived polysaccharides as
nanocarriers, co-delivering adjuvants with peptide antigens, and exploring innate immunostimulatory lipids.
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12375
als-based nanoengineering approaches could improve the
efficacy of vaccines. In this regard, different methodologies
could be applied based on different types of vaccines, with
either distinct or shared requirements. Virus and viral vector-
based vaccines generally do not require further formulations,
and their efficacy and safety profiles are largely dependent on
the purification and inactivation strategies, the optimization of
culturing of viral particles, and the choice of vectors.47 They
have efficient innate advantages in delivery due to their small
sizes, and they are easily recognized by the immune system.
Although inactivated or weakened virus-based vaccines still
make up most of the effective vaccines today, they are not
effective for all pathogens.47 Similarly, the potency of DC
vaccines is mostly dependent on how DCs are manipulated ex
vivo. Strategies in further potentiating cell-based immuno-
therapies, such as the decoration with nanoparticles (NPs),48
are mainly developed for cancer immunotherapies, but they
may be less suitable for large-scale manufacturing that is
needed for the production of antiviral vaccines.
In contrast, synthetic vaccines (nucleic acid-based and
protein/peptide-based ones) generally require additional
formulation designs for optimal efficacy.49 These synthetic
molecules all have essential secondary or higher-order
structures that could be disrupted in physiological con-
ditions.12 Various endogenous enzymes (such as nuclease or
protease) can digest these synthetic molecules before reaching
immunocompetent cells, such as APCs, or immunocompetent
sites, such as secondary lymphoid organs.12 Furthermore,
physiochemical features of these synthetic molecules may pose
challenges for cells to efficiently uptake and process them. For
instance, DNA and mRNA are too large to directly diffuse
across the cellular membrane and their dense negative charge
repels them from cell membranes, which further prevents
efficient cellular uptake.27 Even after penetrating the
membrane, the vaccine formulations need to reach the
endosomal compartments either to be processed into peptides
(protein/peptide-based vaccines), released for further tran-
scription (DNA-based vaccines), or translation (mRNA-based
vaccines). The resulting peptide fragments can be further
loaded onto MHC molecules and exported to the membrane
surface for antigen presentation to T cells (Figure 3b).50
We refer vaccine formulations to engineering approaches
that could facilitate the in vivo delivery of vaccines and enhance
immune responses.51 We categorize the vaccine formulations
in terms of commonly used materials: lipids, polymeric
materials, and inorganic particles (Figure 4). Commonly
pursued feature goals include protection of cargo, improving
loading capacity, and enhancing cellular uptake (Figure 4a).52
In addition, the ideal microenvironment for initiating and
amplifying immune responses is secondary lymphoid organs
Table 1. List of the Representative Formulations Used for Vaccines
materials formulations cargos (vaccine types) functions refs
lipids
ionizable lipid mRNA (glycoprotein 100 (gp100) and tyrosinase-
related protein 2 (TRP 2)) and adjuvants
lipopolysaccharide (LPS)
stabilization, enhanced cellular uptake 54
covalently cross-linked lipid
bilayers
protein/adjuvants (Monophosphoryl lipid A
(MPLA))
stabilization, sustained delivery, co-delivery 55
net negative charged formulations
by tuning cationic lipids and
mRNA ratio
mRNA (influenza virus hemagglutinin (HA) and
OVA)
LN targeting, stabilization 56
nanodiscs via sHDL peptide neoantigens/adjuvants (CpG); dual
adjuvants (CpG and MPLA)
co-delivery, enhanced uptake 57, 58
lipids with cyclic amino headgroup mRNA (OVA and E7) immunostimulatory function 59
polymers
modified dendrimer mRNA (H1N1 influenza, Ebola viruses, and
Toxoplasma gondii parasite)
enhanced loading, cellular uptake, and
endosomal release
60
stearic acid conjugated low MW
PEI
peptide (TRP 2) and mRNA (HIV-1) lowered charge density, higher
biocompatibility
61, 62
cyclodextrin conjugated PEI mRNA (HIV) lowered charge density, higher
biocompatibility
63
PLGA NPs protein (OVA) controlled antigen release 64
PEI/chitosan/poly lysine coated
PLGA/PLA
protein (HBV surface antigen) enhanced loading and cellular uptake,
intrinsic immunostimulatory function
65−67
PBAE and PLGA plasmid DNA (Luciferase) enhanced loading and lower cytotoxicity 68
ionizable amino polyester mRNA (luciferase) spleen targeting 69
oligopeptide-modified PBAE mRNA (enhanced green fluorescent protein) APC targeting 70
albumin and albumin-binding
lipids
peptide (OVA) /adjuvant (CpG) LN targeting 71
polysaccharide from microbial cell
wall and PEI
mRNA (OVA) immunostimulatory, enhanced loading and
endosomal release
72
inorganic
particles
aluminum hydroxide phosphoserine conjugated protein antigen (HIV
trimer)
immunostimulatory, sustained release 73
chemically functionalized Au NPs
with hydrophobicity
− intrinsic immunostimulatory function 74
aspect ratio optimized and surface
modified Au NRs
plasmid DNA (HIV-1) enhanced cellular uptake, intrinsic
immunostimulatory function
75, 76
sized optimized Au NPs peptide (OVA) and adjuvant (CpG) enhanced DC activation 77
mesoporous Si NPs adjuvant; protein (E2 of diarrhea virus) intrinsic immunostimulatory function 78, 79
antigen density tunable/Size
controllable QDs
peptide (self-antigen from multiple sclerosis) LN homing and higher density of antigens
leads to more effective tolerance, intrinsic
antiviral effects
80, 81
oxidized multiwalled CNTs protein (NY-ESO-1) and adjuvant (CpG) co-delivery, enhanced uptake 82
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12376
where APCs, T cells, and B cells are in close proximity for cell-
to-cell interactions. Therefore, targeting DC or LN can be one
type of approach to pursue (Figure 4b).53 To further boost
immune responses, immunostimulatory components or
adjuvants are usually required, and coformulation approaches
have been widely explored for developing immunostimulatory
vaccine formulations (Figure 4c). Relevant studies have been
summarized in Table 1.
Lipids. Lipid molecules are suitable for vaccine formula-
tions because of their high safety profile and tunable
physiochemical features.55,59 These advantages also make
them the leading platform in developing vaccines quickly.
Currently, there are two mRNA vaccine forerunners in
development for COVID-19 that are based on lipid
formulations (Moderna and BioNTech).10 Lipids are a class
of amphiphiles that contain hydrophilic head groups and
hydrophobic tails. Interactions between those molecules can
drive the formation of spherical vesicles to encapsulate vaccine
formulations either with one (unilamellar) or more (multi-
lamellar) lipids bilayers.55 Lipid molecules have also been
explored to achieve distinct biological functions.59 For
instance, cationic lipids are commonly used for efficient
cellular uptake and increase the loading of negatively charged
mRNA and DNA.56 DOTMA, DOTAP, and DOPE are
common examples.56 However, cationic lipids are commonly
associated with high toxicity and neutralization by anionic
serum proteins, which reduce the delivery efficacy.83 There-
fore, ionizable lipids that can change their charge state at
different pH conditions can be utilized to maintain efficacy, to
reduce toxicity, and to facilitate the endosomal release of
cargos.84 Some common strategies to improve the efficacy of
LNPs include enhancing cellular uptake and promoting
endosomal escape by incorporating cholesterol, polyethylene
glycol (PEG) lipids, or helper lipids DOPE.56 Hydrophobic
cholesterol can fill the gaps between lipid tails to stabilize the
vesicle. PEG lipids can shield the vesicles and protect them
from clearance when systemically delivered.56 Helper lipids
have unsaturated bonds to form an unstable hexagonal lamellar
phase that enhances the endosomal escape.85 In addition, good
biocompatibility of lipids makes it possible for expedited FDA
approvals.86 Therefore, LNPs can be optimized in vaccine
formulations to stabilize and enhance uptake of cargo, to
improve immunostimulatory functions for antigens, and for
targeted delivery. Oberli et al. utilized an ionizable lipid, a
phospholipid, cholesterol, and a PEG anchored lipid as LNP
formulations for mRNA vaccine delivery, which resulted in
stable nanoformulations, efficient antigen presentation, and
elicitation of a strong T cell response.54 However, the in vivo
potency of LNP-based vaccines could be limited by their
suboptimal physiological stability, which can cause the fast
release of antigens before initiating any immune response.
Moon et al. developed a hyperstabilized lipid-based vaccine
formulation by covalently cross-linking two lipid molecule
layers between their head groups using a mild chemical
condition that is compatible with loaded antigens.55 The
resulting vesicles are stabilized multilamellar structures that
could trap high levels of protein antigens. Combined with
PEGylation as a well-established stabilization strategy, the
hyperstabilized NPs can sustain the release of antigens up to 30
days in the presence of serum, which significantly enhanced the
humoral immune response.87 By adding the lipid-like mono-
phosphoryl lipid A (MPLA) as adjuvants, the vaccine
formulation can further enhance T cell-mediated humoral
responses.
In addition, LNPs formulations can be optimized for LN
targeting, thereby enhancing the immune response in situ. One
notable study by Kranz et al. demonstrated that a designed
mRNA-lipoplexes (RNA-LPX) could target DCs in vivo simply
by using lipid molecules.56 They established a library of RNA
lipoplexes by tuning the lipid to RNA ratios. Cationic lipids
DOTMA or DOTAP and the helper lipid DOPE or cholesterol
were used. The charge ratio, size, ζ potential, colloidal
properties, and RNA stability also varied with the shifting of
lipid to RNA ratios. Interestingly and unexpectedly, all
negatively charged particles displayed selective targeting to
the spleen. The negatively charged formulation (1.3:2
lipid:RNA) displayed effective targeting of DCs and formed
monodisperse NPs (∼300 nm) that protected encapsulated
mRNA. The authors demonstrated the in vivo efficacy using
RNA-LPX encoding for the influenza virus hemagglutinin
(HA), where DCs, NK, B, and T cells were activated by a
single intravenous injection of HA-LPX. They also observed an
enhanced IFN-α production by vaccination, which is a typical
hallmark of APCs sensing for antiviral environment develop-
ment induced by RNA virus infections through TLR3 and
TLR7.88
To enhance immunostimulatory effects, a common strategy
is to co-deliver adjuvants to stimulate immune responses. Kuai
et al. designed an antigen and adjuvant co-delivery nanodisc
platform based on synthetic high-density lipoprotein
(sHDL).89 One of the striking features of the nanodisc is its
extra-small size: ∼10 nm in diameter, which contributed to the
enhanced trafficking to LNs.57 Exploiting the endogenous role
of HDL as a cholesterol carrier, adjuvant CpG was modified
with cholesterol to efficiently insert into preformed nanodiscs.
This strategy also allowed the co-loading of more than one
adjuvant, which promoted a synergistic immune activation.58
Together, these features contributed to the sustained antigen
presentation by DCs. Recently, there is a growing interest in
exploring innate immunostimulatory functions of lipid
molecules. To expand the diversity of lipid molecules, Miao
et al. developed a library of lipids through a one-step three-
component reaction that included amines, isocyanides, and
alkyl ketones.59 This strategy significantly increased the
diversity of synthesized lipids compared to traditional two-
component reactions.59 They showed that the immunostimu-
latory effect of lipids was dependent on the head groups. The
identified lipids with cyclic amino head groups displayed
immune cell activation efficacy through the intracellular
stimulator of interferon genes (STING) pathway. The lipids
could be further formulated with mRNA to form LNPs as
mRNA vaccine formulations to efficiently deliver the
oligonucleotide products. Another advantage is that the
formulation could activate the mRNA-independent intra-
cellular STING pathway rather than TLRs, which could
reduce systemic toxicity. In addition, inhibition of the antigen
expression related to the TLRs binding by exogenous mRNA
was prevented to favor an optimal immune response.27
Polymers. Aside from the lipids system, polymers have
been widely explored for the delivery of different vaccine
candidates. Specifically, cationic and ionizable polymers are
promising in nucleic acid-based vaccine formulations.
Positively charged polymers can condense nucleic acids as
nanocomplexes for efficient cellular uptake and endosomal
release. In addition, early studies showed that cationic
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12377
materials tend to induce an inflammatory response.90 One
commonly applied polymer category is polyamines, such as
polyethylene imine (PEI) and its derivatives.91 Chahal et al.
utilized modified dendrimer for antigen-encoding mRNA
replicon complexation to form monodisperse NPs.60 This
delivery system, with self-amplifying mRNA, enabled immuni-
zation in mice, which protected them from various virus
challenges, including H1N1 influenza, Ebola virus, and the
Toxoplasma gondii parasite.60 However, their high molecular
weight (MW) and branched polymer structure (needed to
allow a massive loading of vaccine therapeutics) also lead to
severe cytotoxicity, which limits wider applications.92 Zhao et
al. developed a low MW PEI with modifications (stearic acid
and 2 kDa PEI conjugates) to form self-assembled micelles for
delivering mRNA encoding HIV-1 group-specific antigen
(gag).62 Li et al. used 2 kDa PEI conjugated with cyclodextrin
for intranasal delivery of HIV antigen-encoding mRNA that
was able to stimulate HIV-specific immune response.63
Cyclodextrin modification provided mucosal permeation and
mitigated cytotoxicity by lowering the charge density. Other
commonly used polymers include polyester-based, such as
poly(lactic-co-glycolic acid) (PLGA) and poly(lactic acid)
(PLA), because of their biocompatibility and biodegradabil-
ity.93 Several studies have demonstrated the delivery of protein
antigens in PLGA NPs with enhanced immune responses as
compared to soluble formulations.94,95 In addition, such solid
polymer particles can control the kinetics of antigens released
from the polymer core based on different biodegradation
profiles.64,96 However, nonmodified PLGA is generally
negatively charged, and as a result, it has a limited cellular
uptake efficiency and loading of anionic antigens.66 Therefore,
various coating or modification strategies (such as PEI,65,97
chitosan,66 and poly-L-lysine (PLL) coatings) have been
demonstrated to enable the delivery of vaccines and promote
interactions with negatively charged cellular membranes.67
Notably, the cationic polymeric delivery carrier itself could
induce T helper cell responses through TLR-4 dependent IL-
12 secretion.67,98 Poly(β-amino esters) (PBAE) is also
extensively used in delivering plasmids DNA or mRNA
vaccines because of its nontoxic and biodegradable nature.99
Fields et al. developed NPs formulations for plasmid DNA
delivery by combining PLGA and PBAE to reconcile the
drawbacks from each component: the low DNA loading
efficiency of PLGA and the cytotoxicity of PBAE.68 Other
polymeric materials have also been explored, such as
polysaccharides100 and block copolymers.101
In terms of targeted delivery to immune cells, versatility in
polymer chemistry shows great potential in developing tissue-
selective formulations. For instance, a large library of PBAE-
based polymers could be easily synthesized and screened for
desirable target polymers.69 Kowalski et al. designed an
ionizable amino polyester (APE) for mRNA delivery through
the ring opening of lactones with amino alcohols.69 Tissue-
selective delivery was achieved by different APE candidates,
including APCs within the spleen.69 Oligopeptides have been
used to modify PBAE to allow the tuning of the charge status
of NPs.102 Initially, this was done to optimize cargo loading
and mitigate cytotoxicity. Later, Fornaguera et al. demon-
strated that specific oligopeptide modifications lead to APCs
targeting within the spleen and exhibited an efficient
transfection of mRNA.70 Inspirations also come from using
native LN trafficking molecules. Clinically, visualization of
sentinel LNs is achieved by injecting dyes that bind to albumin
because of the LN trafficking capability of albumin particles.103
Inspired by the endogenous role of albumin, Liu et al. designed
a vaccine by conjugating the adjuvants or antigens to the
albumin-binding lipid tail.71 The vaccines displayed a notable
increased accumulation in the LNs, which decreased the
systemic distribution of the vaccines and prolonged the vaccine
bioavailability. These features limited the systemic toxicity
while increasing the potency of vaccination for OVA antigen,
HIV antigen, and human papillomavirus (HPV) antigen.71
Some polymeric delivery carriers have been demonstrated to
have immunostimulatory effects, such as polymers with
cationic charge and hydrophobic domains.104,105 In addition,
inspired by microbes’ immunostimulatory effect, Son et al.
designed sugar capsules that were derived from microbial cell
walls for vaccine NPs coating.72 The coating process was
achieved by using silicate NPs (Si NPs) as a template, followed
by its removal after the mRNA was loaded and the sugar
capsules were entirely coated. Notably, the removal of Si NPs
renders relatively flexible nanostructures that are beneficial for
lymphatic drainage and accumulation.72 The mRNA cargos
can be efficiently loaded by PEI coating due to electrostatic
interactions. The PEI coating was also expected to facilitate the
endosomal release for mRNA translation and antigen
presentation. The sugar capsule itself functioned as a strong
DC activator by eliciting the production of a variety of pro-
inflammatory factors, including IL-6, TNF-α, and IL-12 p40,
without the use of exogenous adjuvants.72
Inorganic Particles. Inorganic materials have been used in
vaccine formulations for a long time. Back in 1926, an
aluminum compound (alum) was discovered to be able to
precipitate the toxoid to enhance the immunogenicity.106 Alum
is still the most widely used adjuvant today. In spite of over 80
years of development, the underlying mechanisms for alum as
an adjuvant are still controversial.107 Popular explanations
include the “depot theory” for extended antigen release106 and
enhanced uptake by APCs through antigen absorption on
alum.108 Various immunity signaling and activation pathways
have also been identified to contribute to the function of the
adjuvants.109 Recently, Moyer et al. utilized the interaction
between alum and phosphoserine (pSer) to engineer a short
peptide-immunogen conjugate to significantly prolong the
immunogen release from pSer-immunogen:alum complexes.73
The complexes could form NPs for efficient trafficking to LNs
and initiated antigen processing and presentation by APCs.73
Site-specific pSer modification on the base of the HIV trimer
could enable the immunogen (HIV trimer) to be displayed
with a specific orientation, which could prevent the induction
of undesired NAbs.73 Apart from alum, other inorganic
materials have been proposed that may stimulate vaccine-
specific immune responses, such as silica crystal110 and calcium
phosphate.109
In addition to the traditional use as immunostimulatory
adjuvants, a wide range of biocompatible inorganic particles
have also been developed as vaccine formulations,111 including
quantum dots (QDs),80 gold nanoparticles (Au NPs),77
carbon nanotubes (CNTs),82 and Si NPs.79 The physical
properties of formulations, such as surface charge, hydro-
phobicity, and aspect ratios, are closely associated with the
efficacy of vaccines.112 Inorganic materials have a high degree
of tunability to allow them to fulfill the requirements of vaccine
formulations. Easy preparation and good storage stability are
additional advantages of inorganic particles.113 For example,
Au NPs have been chemically functionalized to investigate the
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12378
relationship between surface hydrophobicity and the associated
immune activity.74 It is also expected that other functional
groups may bring a pathway-dependent immune activation
that is specifically desirable for antiviral defense. Xu et al.
described surface-engineered Au nanorods (NRs) for plasmid
DNA vaccine delivery.75 With certain surface modifications
and aspect ratios, cellular toxicity of Au NRs can be lowered,
internalization can be enhanced, and intrinsic adjuvant features
can be utilized.76 In vivo vaccination with HIV-1 envelope
glycoprotein (Env) plasmids demonstrated enhanced cellular
and humoral immunity.75 Zhou et al. synthesized a series of Au
NPs (15 to 80 nm) for co-delivery of antigens (OVA peptides)
and adjuvants (CpG) by surface conjugation.77 Particles of 60
and 80 nm outperformed other sizes (15, 30, and 40 nm) in
activating DCs; this was most likely due to a higher payload of
antigens. Notably, they promoted DC homing to the LNs and
induced potent cellular immune response.77 High tunability is
also achievable in other organic particles, such as adjuvant-
based mesoporous Si NPs,78 mesoporous Si NP-based
antigen/adjuvant co-delivery systems,79 antigen density tuna-
ble QDs,80 size-controlled antiviral QDs,81 and surface
oxidized antigen/adjuvant-based multiwall CNTs.82
VACCINE DELIVERY DEVICES
If the vaccine formulations are considered as utilizing
nanoscale engineering, then vaccine delivery devices utilize
micro/macroscale engineering to improve vaccine efficiency
and efficacy. Beyond the optimization of the vaccine itself and
designing appropriate delivery carriers, the efficacy of a vaccine
also depends on pharmacokinetics (PK) of the antigens after
administration.114 When the body is exposed to an exogenous
antigen, both humoral and cellular immune responses are
initiated to neutralize it, which also leads to immunological
memory.12 During a virus infection, the replication process
typically takes one to several weeks to sustainably stimulate the
immune system with a continuous supplement of antigens.115
In contrast, typical vaccines show a rapid clearance and can
only be detected in LNs within days.114 Although still poorly
Figure 5. Summary and characteristics of various vaccine delivery devices. (a) Drug delivery devices developed for vaccine administration: I.
Traditional vaccine-containing solutions for bolus injection. II. Minimally invasive microneedle devices (including fast-dissolving, sustained-
released, and liquid-eluting hollow microneedles). III. Injectable drug delivery devices (such as microparticles, solid scaffolds, and hydrogel-
based). IV. Other devices for enhancing vaccine efficacy (such as electroporation and iontophoresis). (b) Schematic for representative PKs
of vaccine and administration features for different vaccine delivery devices: I. Traditional bolus injection is invasive and may require
multiple doses. II. Fast dissolving or liquid eluting MNs can be self-administrable and minimally invasive but still require multiple doses.125
III. Sustained released MNs can be both minimally invasive, with only one dose necessary,122 while scaffold and gel vaccines could be
injectable to avoid implantation and also only require one dose. IV. Pulse-released microparticles can mimic a multiple-dose regimen with
only one injection.127,128 (c) Schematic of representative mechanisms for vaccine delivery devices. I. Scaffold vaccines: The scaffold could
sustainably release recruiting factors to home DCs, and encapsulated antigens could further be processed by DCs in situ.129 II. Gel vaccines:
Hydrogel microenvironment can be engineered to support encapsulated cell vaccines (such as DC vaccines) by incorporating cell adhesive
peptides and supplementary factors.136
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12379
understood, the PK of a vaccine, which refers to the dynamics
of the antigens presence during immunization, has a profound
impact on immune response.87 One study indicated that
increasing the dosage of vaccines over 2 weeks at both priming
and boost stages induced a durable immune response and
increased antibody production compared to traditional bolus
immunization.87 Also, computational models suggested that
antigens can be better captured in germinal centers (GCs) if
antigen availability is extended.116 In addition to the
advantages of extended antigen availability, traditional
vaccination strategies often fail to yield the desired optimized
immune responses in a single dose. Furthermore, vaccine
regimes are often designed to be multiple shots, thus achieving
an even longer immune protection by rechallenging the
immune system with the same antigens in secondary
“boosting” shots administered after the primary shots.117
Therefore, vaccine delivery devices have been widely
investigated, to develop single-dose vaccines, minimal
invasively administered vaccines, and self-administrable
vaccines (Figure 5a,b). However, its potential can only be
realized if various design principles are met (Table 2).
Microneedles Vaccines. Transdermal microneedles
(MNs) patches have been proposed as a promising drug
delivery device for immunotherapies144−147 based on the
minimally invasive nature of MNs and the highly active
immune environment of the skin. Additionally, since MNs
patch-based vaccines can be easily administered without the
need for significant medical training, these vaccines can be
easily deployed both in areas with limited medical resources,
such as developing countries and during times of significant
medical crisis, as seen during an outbreak or pandemic. Early
studies include the use of vaccine-coated metal MNs118 and
fast-dissolving polymer-based MNs.119 Recently, the dissolving
MNs (carboxymethyl cellulose-based) were used for adjuvants
and Ad5-based vaccine co-delivery in an attempt to address
COVID-19.147 A 1 month storage of MNs vaccine at 4 °C
maintained the immunogenicity of Ad, where no significant
loss in antibody responses was shown in mice studies.147
Similar dissolving MNs are also being investigated to deliver
the SARS-CoV-2 subunit spike protein (S1).120 Potent virus-
specific antibody responses were elicited in mice as early as 2
weeks after the immunization with MNs. The dissolvable MNs
for influenza vaccine have been previously evaluated in a phase
I clinical trial compared to the traditional intramuscular
hypodermic injection of a single-dose vaccine.121 The MN
patch was well-tolerated and immunogenic after a single-dose
vaccination. In addition, self-administration exhibited efficacies
comparable to the administration by medical workers. The
dissolving feature leaves no sharp wastes, and self-admin-
istration could significantly relieve the pressure on medical
resources, which could be especially critical in coping with a
pandemic. Beyond these milestones, advances in micro-
fabrication, materials engineering, and vaccine formulation
optimization have provided additional opportunities to
enhance vaccine efficacies.148
Recent advances in microfabrication, such as 3D printing
and stereolithography, can allow for more sophisticated MNs
designs.125 Liquid-based vaccine formulations are still the most
widely used.125 However, liquid-based vaccine formulations are
incompatible with most solid-based MNs forms. Inspired by
the capability of snake-fangs to efficiently inject venom, Bae et
al. used an exposure lithography-based strategy to produce
MNs patches with snake-fang architectures for efficient liquid
Table 2. Summary of Typical Designs, Advantages, and Limitations of Different Vaccine Delivery Devices
delivery devices designs advantages limitations refs
MNs vaccine (minimally invasive; minimal require-
ment on medical professionals and medical
equipment)
fast-dissolving
MNs
fast-dissolving polymers as MNs matrix fast and enhanced transdermal delivery; could be cold-chain free multiple administrations; vaccine for-
mulations may get denatured from
MNs fabrication
118−121
sustained-re-
lease MNs
controlled biodegradable materials as
MNs matrix
potential to be single shot; protect the vaccine formulations in the
physiological environment; could be cold-chain free
vaccine formulations may get dena-
tured from MNs fabrication
122−124
liquid-eluting
MNs
hollow MNs with different architectures compatible with liquid vaccine formulation; fast and enhanced
transdermal delivery
multiple administrations; requirement
of cold-chain transport
125
injectable materials assisted vaccine (potential for
single-shot vaccines to reduce the requirement of
medical resources)
pulse-released
microparticle
materials with different biodegradation
profiles
optimal release for a potent immune responses; release kinetic can
be optimized by materials engineering
vaccine formulations may get dena-
tured during the fabrication
126−128
scaffold vac-
cine
solid scaffolds that could tune the release
of different immunomodulatory factors
continuously modulate the immune response in situ injectability need to be achieved to
avoid surgical implantation
129−134
hydrogel vac-
cine
biomimicking gellable materials with
tunable biophysical properties and high
cytocompatibility
compatible with various types of vaccines, including cell-based
vaccines; simple fabrication process and the water environment
help protect the cargos
in situ gelation may be weak to achieve
sustained release
135−143
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12380
formulations delivery.125 The design was compatible with most
existing vaccine formulations, and the patch can be applied
with the pressure of the thumb and achieve an efficient
transdermal delivery.125 Nevertheless, solid-based vaccines,
including MNs-based ones, also attracted significant attention
because of the decreased cost of vaccine manufacturing,
transport, and storage when compared to the liquid-based
formulations.149 One of the key challenges faced by these MNs
patches is the loss of vaccine activity during the drying or
heating process of MNs patch fabrication,150 where molecular
structures of vaccines may be disrupted.151 Different strategies
can be adopted, such as the use of lyoprotective polymers as
MNs matrices, which include silk fibroin,122 gelatin, dextran,
and PVP,152 or adding sugar molecules as stabilizing excipients,
including trehalose and sucrose.147,152 In the vaccine discovery
phase, the introduction of certain mutations can improve the
thermal stability of vaccines as well,148 which can be further
applied to specific MNs formulations. Additionally, a hyper-
stabilized vaccine formulation, such as the interbilayer-cross-
linked multilamellar vesicles, helped protect vaccines during
the drying process of MNs fabrication and have outperformed
other traditional delivery strategies in inducing a potent
antigen-specific immune response.123
MNs also provide advantages in tuning optimal release
profiles of vaccines. As mentioned earlier, the sustained
availability of vaccines is a desirable feature for immunization.
Vaccines targeting different infectious diseases may vary in
terms of optimal release profiles based on the unique features
of each virus. Initially, MNs for vaccination were mainly an
upgrade to traditional bolus delivery by using a fast-dissolving
MNs matrix,119 which requires multiple doses for sufficient
immunity. With the exploration of the desired PK of vaccines,
MNs can play a much more important role. For example, a silk
matrix with crystallized structures could be used as an MNs
matrix to sustainably release loaded vaccines over 2 weeks and
to elicit potent immune responses with one single admin-
istration.122 In addition, biodegradable polymers with known
degradation kinetics can be used to encapsulate the vaccines
and tune antigen release profiles. These polymers include
biodegradable cationic PBAE123 and PLGA.124
Injectable Material-Assisted Vaccines. Macroscale
biomaterials strategies have recently boosted the field of
immunotherapy.153 There are many advantages in controlled
release and encapsulation of various cargos, ranging from small
molecules to proteins and cells.153 These varied cargo elements
can help to meet the need for diverse types of vaccine
candidates. In terms of controlling PKs of vaccines, both
sustained and pulsatile releases could be possibly achieved.153
In addition, materials engineering can enable devices that do
not require surgically invasive administrations.153 Together,
they could minimize the requirement of medical resources
without mitigating the vaccine efficacy.
PLGA has also been developed for a single-shot vaccine
based on the tunability of its biodegradation profile.126 In the
case of polio vaccines, two to three administrations over several
months are required. Tzeng et al. demonstrated that IPV could
be encapsulated within PLGA microspheres stably and released
through two bursts that were one-month apart.126 They
examined the effect of adding cationic excipients (Eudragit E,
PLL, and branched PEI) to stabilize IPV antigens, protecting
them from denaturation by acidic byproducts, and modulate
degradation profiles of PLGA microspheres.126 Notably, a
single shot of PLGA microsphere-based vaccines could elicit
the production of NAb as potent as two bolus injections.126
Combined with the advances in microfabrication and additive
manufacturing, McHugh et al. fabricated microscale (∼400
μm) hollow PLGA-based microparticles that were inject-
able.127 The microparticles can be filled with vaccine
formulations, and the degradation rate of the microparticle
shell (a lactic/glycolic copolymer) can be tuned to achieve
controlled release spanning from a few days to a few
months.127 Such a design can accomplish the goals of general
transdermal vaccine injection and achieve temporally con-
trolled pulsed delivery of antigens in a single injection.127
Recently, a similar PLGA-based microparticles platform was
applied to release a STING agonist (cyclic guanosine
monophosphate-adenosine monophosphate (cGAMP)) in a
pulsatile manner over a long period (∼16 days).128 One single
injection of the pulsatile drug-releasing microparticles induced
potent immunity as effective as three separate injections. These
engineered platforms are versatile in terms of encapsulating
different cargos required by vaccine formulations and tuning
release kinetics correspondingly. However, even though the
use of FDA-approved PLGA could facilitate the clinical
translation to a certain degree, several design limitations
need to be overcome for translating PLGA-based drug delivery
devices into clinical applications. For instance, the pulsatile
release was achieved with hollow microparticles that have thick
PLGA shells to encapsulate the drugs in the hollow core for
controlled release. This design significantly sacrificed their
loading efficiency due to the limited volume of the core.
Clinically, patients need a high drug dosage, and this
formulation needs further optimization to meet the dosage
requirement. In addition, PLGA can create an acidic
microenvironment during biodegradation, damaging the
encapsulated drugs.126 Furthermore, the encapsulated vaccines
need to survive the fabrication process; brief heating requires
thermally stable vaccines.127,128 All these aspects could
compromise their applications for a broader collection of
drugs or vaccines.
Traditionally, ex vivo modification of antigen-specific DC
vaccines is a multistep process associated with high costs and
high levels of complexity.43 It was proposed that in situ DC
recruitment, activation, and further trafficking to LNs would be
more desirable compared to the ex vivo modification strategy
(Figure 5c).129 The early study by Ali et al. demonstrated the
use of macroporous scaffold vaccines based on PLGA, which
presented cytokines (GM-CSF), danger signals (CpG), and
antigens in a spatiotemporally defined manner. GM-CSF can
be sustainably released from the scaffolds over 20 days for
effective DC recruitment.129 By co-loading the CpG through
PEI-mediated electrostatic interactions, CpG was immobilized
on the scaffolds for local internalization by the recruited DCs.
This process mimicked an infection and was able to
continuously recruit and activate DCs.129 This specific scaffold
vaccine was surgically implanted; nevertheless, it encouraged
further development of injectable vaccines that work similarly.
Kim et al. demonstrated the use of mesoporous silica rods
(MSRs) with a high-aspect-ratio that could be injected with a
syringe needle.130 They could self-assemble in vivo as
macroporous scaffolds to initiate potent immune responses.130
The DCs could be recruited into the scaffolds where the
inflammatory signals (GM-CSF), adjuvants (CpG), and
antigens (OVA) were sustainably released from the scaffolds
for in situ DC activation and maturation. It was also
demonstrated that locally injected scaffold vaccines based on
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12381
MSRs could elicit strong systemic immune responses with the
increase in levels of antigen-specific B cells, helper T cells, and
cytotoxic T cells.130 A PEI coating was later added to the
design to enable antigen loading through adsorption for
enhanced immunogenicity.131 This specific system was
recently utilized for synthetic peptide vaccines and small
antigens.132 This strategy was able to bypass multiple
immunization requirements but sustained a more robust
antibody response, compared to traditional bolus or alum
vaccine formulations.132
Scaffold-based vaccines have displayed efficacy in inducing
systemic immune response through in situ immune modu-
lations. In vivo self-assembled scaffolds provide a good
alternative to bypass surgically implantation; however, the
self-assembling process is commonly undefined in terms of
how long it could take for self-assembling.133 Materials
engineering facilitates the fabrication of scaffolds maintaining
injectability. Bencherif et al. described a strategy for fabricating
injectable scaffolds with shape memory based on cryotropic
gelation.134 The cryogels were based on alginate methacrylate,
and they were capable of being reversibly deformed, over 90%
strain, after being chemically cross-linked in a frozen condition
(−20 °C).134 The ice crystals hindered the cross-linking in
frozen spots and therefore formed the macroporous pores that
not only enabled the shape memory for the gel but also
allowed the DCs infiltration.134
Hydrogels have been extensively explored in the field of
immunotherapy.153 They have advantages in terms of control-
lable delivery of multiple cargos, in situ immunomodulation,
and mild fabrication process for various vaccine formulations
(Figure 5c). Early pioneering work includes the loading of ex
vivo antigen-primed DC vaccines within Ca2+ cross-linked
alginate gel by Hori et al.135 Activated DCs have a short
lifespan, and continuous immune responses may be suppressed
if no sufficient host DCs and T cells could be recruited and
primed in vivo.135 Therefore, cell-based vaccines delivered by
gels are expected to improve cell survival and prolong the
presence of DCs.136 Both host DCs and cytotoxic T cells can
be recruited to the injection site by cytokines and chemokines
secreted from the injected DCs. In addition, the enhanced
efficacy of transplanted cells can be enhanced by supplement-
ing the gels with other immunomodulatory molecules, such as
cytokines (IL-2) or adjuvants (CpG).137 The increased
persistence of transplanted cells could be achieved by
designing gels mimicking ECM by the incorporation of cell-
adhesive peptides, such as RGD.154
Apart from cell-based vaccines, other types of vaccines could
also be incorporated into gel systems, where the release profiles
can be tuned. Roy et al. utilized the in situ cross-linking
mechanism of tetrafunctional poly(ethylene oxide) amine and
poly(ethylene oxide) succinimidyl glutarate to encapsulate
plasmid DNA.138 The gelation did not interfere with the
supercoiled structure of plasmid DNA, and the gelling ability
can be tuned by adjusting the concentrations of these two
components. Notably, gene expression levels were comparable
to bare DNA, but the duration of expression was significantly
longer. Other in situ gel systems explored for vaccination
include temperature-responsive gels139 and Michael-addition-
type hydrogels.140 Singh et al. engineered dextran and PEG-
based hydrogels that could be cross-linked in situ for antigen/
adjuvant delivery.141,142 Umeki et al. designed an immunosti-
mulatory CpG DNA-based hydrogel for antigen delivery.143
To achieve a sustained release of antigens, they proposed to
cationize the antigens by ethylenediamine conjugation where
antigens could form complexes with a negatively charged DNA
hydrogel matrix. The innate immunostimulatory function of
gel and prolonged antigen presence leads to an enhanced
antigen-specific immune response with less toxicity.143 In situ
gelling strategies or physically cross-linked gels from these
studies generally take minutes to hours, which renders limited
control on cargos release and material properties during the
gelling period.143 Therefore, the development of gel systems
that are both injectable and display defined cargo release/
biodegradation profiles could be desirable for vaccine
administration.
Other Delivery Devices. Many more investigations are
currently ongoing to engineer alternative drug delivery devices.
For instance, to promote the uptake of DNA vaccine by cells,
electroporation is one of the traditional and effective strategies
to enhance the uptake of plasmids.155 Different parameters,
such as electrical features (voltage, current, and frequency) and
length of the electrode (intradermal or intramuscular targets),
need to be optimized to ensure efficacy and safety and to
minimize pain. This process has recently demonstrated
promising clinical phase I results for MERS.25 The same
electroporation device (CELLECTRA) is also being inves-
tigated for a DNA vaccine of SARS-CoV-2 from Inovio.156 The
S protein of SARS-CoV-2 encoding DNA vaccine demon-
strated potent antigen-specific T cell responses and the
production of neutralization antibodies in preclinical mice
and guinea pigs studies.156 In order to incorporate the
advantages of MNs, electroactive MNs devices have also
been developed for the electroporation of DNA vaccine in a
minimally invasive manner.157 Similar devices using iontopho-
resis can also enhance the transdermal delivery of vaccines.158
Recently, Tadros et al. designed millimeter-scale star-shaped
particles, termed STAR particles, based on aluminum oxide for
topical vaccine delivery,159 and the minimally invasive STAR
particles elicited an immune response comparable to intra-
muscular injection.159 Even though STAR particles are simple
to manufacture and well-tolerated by the immune system, a
much higher dosage was administered compared to intra-
muscular injection. In addition, potential human-to-human
variability in administration and drug leftovers on the skin may
result in differential efficacy and undesirable toxicity.
In terms of reducing the demands for medical resources,
vaccines that could be intranasally or orally administered are
desired if their efficacy is comparable to transdermal delivery.44
The main strategy for intranasal or oral vaccine development is
currently limited to weakened viral vaccines mimicking natural
infections. Nonliving vaccines are safer but less immunogenic
and thus may benefit greatly from drug delivery formulations
and devices.160 A variety of drug delivery devices have been
proposed for oral vaccination delivery. For instance, the
micromotor-driven microparticles for oral delivery of anti-
gens.161,162 The micromotor vaccine can efficiently enter the
intestine to enhance the antigen uptake. Aran et al. designed a
needle-free oral microjet where the high-pressure liquid jet of
the vaccine was produced by a self-contained gas-producing
reaction.163 It was shown that a buccal immunity response was
enhanced, and this method was able to elicit a potent humoral
immune response both locally and systemically.163 Abramson
et al. engineered a self-orienting system for oral delivery that
could attach to the gastric wall by autonomously positioning to
maximize the delivery efficiency.164 Innovations in drug
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12382
delivery devices will significantly transform traditional
vaccination regimes.
SUMMARY AND FUTURE DIRECTIONS
The devastating disruptions caused by the sudden outbreak of
COVID-19 has urged vaccine development at an unprece-
dented speed.4 Within 70 days of the first release of the genetic
information on SARS-CoV-2, the first dose of a vaccine was
administered to human participants.9 As of August 25, 2020,
31 vaccine candidates are in various stages of clinical trials, and
around 142 candidates are in preclinical trials.10 Apart from the
high diversity of the vaccine candidates, many candidates are
based on different technological platforms, such as nano-
technology delivered mRNA vaccine, electroporation device-
enabled DNA vaccine, and MN-delivered vaccine. The
COVID-19 pandemic has accelerated the deployment of
different vaccine formulations and delivery devices. It is
reasonable to expect that more efforts will be dedicated to
further optimize the vaccine designs to help fight future
outbreaks.
Here, we reviewed recent research efforts in the field of
vaccine discovery, vaccine formulation development, and
vaccine delivery device designs. Because of the deployment
of various engineering technologies, it is expected that future
vaccines will not only need to demonstrate safety and efficacy
but also to be relatively convenient and simple to use for better
coping with population-level vaccinations or sudden outbreaks
worldwide. Nanoengineered formulations could enhance the
immune response by delivering the required components
together and maximizing their uptake by immunocompetent
cells. To expedite the future preparedness of vaccines for the
occurrence of a pandemic, synthetic vaccines have demon-
strated great potential for rapid clinical applications because
they are molecularly defined and independent of the time-
consuming culture of viruses, which can accelerate the initial
vaccine development. However, synthetic vaccines are also the
ones that need to be formulated the most, which constitutes
the major part of their time costs. To accelerate the
development of synthetic vaccines, the nanoengineered
formulations need to be simple and robust to manufacture in
terms of chemistry or the selection of raw materials.165,166 In
terms of vaccine delivery devices, self-administrable vaccines,
single-shot vaccines, and solid MN-based vaccines could
further enhance the availability of vaccines to a broader
population. For instance, self-administrable vaccines can be
mailed to patients at home without the need of medical
experts.165 Single-shot vaccines can be much more convenient
compared to the traditional immunization regimens that
require multiple shots. Furthermore, solid MN-based vaccines
that are cold chain free could simplify the vaccine trans-
portation and distribution process. All of these aspects could
be enabled by engineering approaches to accelerate the
development of vaccines at a pandemic speed.165,166 Although
combining the advantages of vaccine formulations and vaccine
delivery devices is promising, micro/macro-engineering still
needs to develop solutions that do not require “harsh”
conditions to be compatible with nanoengineered formula-
tions.167 Expectedly, platform technologies that can be quickly
adapted to address emerging viral diseases could speed up the
pace of vaccine development.
However, several challenges remain before these goals can
be fully achieved. First, a differently identified virus is unknown
in terms of its interactions with our immune system and which
type of immune responses are most desirable to trigger
prolonged immunity. An improved understanding of our
immune system and its interactions with viruses are the basis
for designing an effective vaccine, which also guides the steps
for designing corresponding vaccine administration regimens.
Second, innovative formulations or delivery devices for
vaccines are mostly in the preclinical stage, which have not
been approved by the FDA yet. For example, MN-based
vaccines showed encouraging results from a phase I trial of
dissolvable MN-based influenza vaccine and indicated a
promise of using it as an alternative strategy for vaccination.121
However, patient compliance with this approach and its
efficacy needs to be tested in a larger population, considering
only 100 human subjects are currently involved. Also, the
efficacy of vaccines by self-administration should be further
validated in comparison to those performed by medical
experts. In addition, the proposed formulations and delivery
devices should be easy to manufacture with minimal batch-to-
batch variability to simplify the quality control process. The
raw materials used could prioritize the FDA-approved ones to
simplify the regulatory process, and the required chemistry is
preferred to be simple and robust. Even though advanced
manufacturing technologies excel in reducing batch-to-batch
variability, a high cost may be associated. The improved
efficacy of a vaccine from these formulations and delivery
devices should not be complicated by the fabrication process
so that it can facilitate the clinical translation and regulatory
approvals.165
Additional challenges for vaccine development also exist
during a pandemic. Multiple vaccine candidates flush into
different stages of clinical trials that could burden the
regulatory process by the FDA for selecting promising
candidates and deciding final ones.4 Furthermore, human
trials are starting even before the most suitable animal models
for safety and efficacy are defined.168 Based on previous
successful preclinical and early stage clinical data for a similar
platform, animal studies are even skipped directly for human
administrations.10 Other engineering approaches such as
organs-on-a-chip169−171 or screening using organoids172 may
serve as a promising tool in future clinical trials for efficacy and
toxicity determination. Ethically controversial human challenge
trials by deliberately infecting humans with a virus have been
proposed and pursued to accelerate the process of vaccine
development.173 Therefore, potentially we should have vaccine
clinical trials specifically designed for different scenarios by
incorporating engineering advances and regulatory guidelines
readily outlined for unusual trials. In conclusion, human health
and societal stability are constantly challenged by sudden virus
outbreaks, and the rapid development of effective vaccines is
believed to be an ultimate solution. We expect to observe faster
and more efficient vaccine development paradigms in the
future by combining the advances in different engineering
fields.
AUTHOR INFORMATION
Corresponding Authors
Ali Khademhosseini −Department of Bioengineering, Center for
Minimally Invasive Therapeutics, Department of Chemical and
Biomolecular Engineering, California NanoSystems Institute,
Jonsson Comprehensive Cancer Center, and Department of
Radiological Sciences, University of California, Los Angeles, Los
Angeles, California 90095, United States; Terasaki Institute for
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12383
Biomedical Innovation, Los Angeles, California 90064, United
States; Email: khademh@terasaki.org
Wujin Sun −Department of Bioengineering, Center for
Minimally Invasive Therapeutics, and California NanoSystems
Institute, University of California, Los Angeles, Los Angeles,
California 90095, United States; Terasaki Institute for
Biomedical Innovation, Los Angeles, California 90064, United
States; orcid.org/0000-0002-3167-111X; Email: wsun@
terasaki.org
Authors
Xingwu Zhou −Department of Bioengineering and Center for
Minimally Invasive Therapeutics, University of California, Los
Angeles, Los Angeles, California 90095, United States;
Department of Pharmaceutical Sciences, University of Michigan,
Ann Arbor, Michigan 48109, United States
Xing Jiang −School of Nursing, Nanjing University of Chinese
Medicine, Nanjing 210023, China
Moyuan Qu −The Affiliated Stomatology Hospital, Zhejiang
University School of Medicine, Key Laboratory of Oral
Biomedical Research of Zhejiang Province, Zhejiang University
School of Stomatology, Hangzhou 310006, China
George E. Aninwene, II −Department of Bioengineering and
Center for Minimally Invasive Therapeutics, University of
California, Los Angeles, Los Angeles, California 90095, United
States
Vadim Jucaud −Terasaki Institute for Biomedical Innovation,
Los Angeles, California 90064, United States
James J. Moon −Department of Pharmaceutical Sciences,
University of Michigan, Ann Arbor, Michigan 48109, United
States
Zhen Gu −Department of Bioengineering, Center for Minimally
Invasive Therapeutics, California NanoSystems Institute, and
Jonsson Comprehensive Cancer Center, University of California,
Los Angeles, Los Angeles, California 90095, United States
Complete contact information is available at:
https://pubs.acs.org/10.1021/acsnano.0c06109
Author Contributions
●These authors contributed equally to this work.
Notes
The authors declare no competing financial interest.
ACKNOWLEDGMENTS
This work was supported by National Institute of Health
(CA214411, GM126831, and AR069564).
VOCABULARY
Coronavirus disease 2019 (COVID-19), a disease caused by
severe acute respiratory syndrome coronavirus 2 (SARS-CoV-
2) and has been declared as a pandemic by WHO since March
11, 2020; antiviral vaccine, a biological preventive preparation
strategy to train the immune systems to fight against viral
pathogens; vaccine formulations, biomaterial engineering
strategies aiming to boost the efficacy or reduce side effects
of vaccines through co-delivering adjuvants, targeting immune
cells, or protecting vaccines activities; vaccine delivery
devices, tools for administering the vaccine formulations, and
they can be used by medical workers or the patients
themselves; pattern recognition receptors (PRRs), proteins
that can recognize pathogenic molecules as part of innate
immunity in response to virus infection, including identifying
pathogen-associated molecular patterns from a virus; adju-
vants, supplementary agents that could be co-delivered with
vaccines to boost the immune response, minimize the dosage
of antigens, and maintain longer immunity; lymph nodes,
round or bean-shaped clusters of cells containing abundant
immune cells that can initiate the robust immune response
toward viral infections
REFERENCES
(1) Bedford, J.; Enria, D.; Giesecke, J.; Heymann, D. L.; Ihekweazu,
C.; Kobinger, G.; Lane, H. C.; Memish, Z.; Oh, M.-d.; Sall, A. A.;
Schuchat, A.; Ungchusak, K.; Wieler, L. H. COVID-19: Towards
Controlling of a Pandemic. Lancet 2020, 395, 1015−1018.
(2) Berlin, D. A.; Gulick, R. M.; Martinez, F. J. Severe Covid-19. N.
Engl. J. Med. 2020, DOI: 10.1056/NEJMcp2009575.
(3) Rome, B. N.; Avorn, J. Drug Evaluation during the Covid-19
Pandemic. N. Engl. J. Med. 2020, 382, 2282−2284.
(4) Lurie, N.; Saville, M.; Hatchett, R.; Halton, J. Developing Covid-
19 Vaccines at Pandemic Speed. N. Engl. J. Med. 2020, 382, 1969−
1973.
(5) Fauci, A. S.; Morens, D. M. Zika Virus in the Americas Yet
Another Arbovirus Threat. N. Engl. J. Med. 2016, 374, 601−604.
(6) Regules, J. A.; Beigel, J. H.; Paolino, K. M.; Voell, J.; Castellano,
A. R.; Hu, Z.; Muñoz, P.; Moon, J. E.; Ruck, R. C.; Bennett, J. W.;
Twomey, P. S.; Gutie
́
rrez, R. L.; Remich, S. A.; Hack, H. R.;
Wisniewski, M. L.; Josleyn, M. D.; Kwilas, S. A.; Van Deusen, N.;
Mbaya, O. T.; Zhou, Y.; et al. A Recombinant Vesicular Stomatitis
Virus Ebola Vaccine. N. Engl. J. Med. 2017, 376, 330−341.
(7) Mulangu, S.; Dodd, L. E.; Davey, R. T.; Tshiani Mbaya, O.;
Proschan, M.; Mukadi, D.; Lusakibanza Manzo, M.; Nzolo, D.;
Tshomba Oloma, A.; Ibanda, A.; Ali, R.; Coulibaly, S.; Levine, A. C.;
Grais, R.; Diaz, J.; Lane, H. C.; Muyembe-Tamfum, J.-J.; The, P. W.
G. A Randomized, Controlled Trial of Ebola Virus Disease
Therapeutics. N. Engl. J. Med. 2019, 381, 2293−2303.
(8) Fineberg, H. V. Pandemic Preparedness and Response
Lessons from the H1N1 Influenza of 2009. N. Engl. J. Med. 2014, 370,
1335−1342.
(9) Le, T. T.; Andreadakis, Z.; Kumar, A.; Roman, R. G.; Tollefsen,
S.; Saville, M.; Mayhew, S. The COVID-19 Vaccine Development
Landscape. Nat. Rev. Drug Discovery 2020, 19, 305−306.
(10) Draft Landscape of COVID-19 Candidate Vaccines. https://
www.who.int/publications/m/item/draft-landscape-of-covid-19-
candidate-vaccines (accessed 2020-08-28).
(11) Cox, R. J.; Brokstad, K. A. Not Just Antibodies: B Cells and T
Cells Mediate Immunity to COVID-19. Nat. Rev. Immunol. 2020,
581−582.
(12) Irvine, D. J.; Swartz, M. A.; Szeto, G. L. Engineering Synthetic
Vaccines Using Cues from Natural Immunity. Nat. Mater. 2013, 12,
978−990.
(13) Callaway, E. The Race for Coronavirus Vaccines: A Graphical
Guide. Nature 2020, 580, 576.
(14) Nabel, G. J. Designing Tomorrow’s Vaccines. N. Engl. J. Med.
2013, 368, 551−560.
(15) Gao, Q.; Bao, L.; Mao, H.; Wang, L.; Xu, K.; Yang, M.; Li, Y.;
Zhu, L.; Wang, N.; Lv, Z.; Gao, H.; Ge, X.; Kan, B.; Hu, Y.; Liu, J.;
Cai, F.; Jiang, D.; Yin, Y.; Qin, C.; Li, J.; et al. Development of an
Inactivated Vaccine Candidate for SARS-CoV-2. Science 2020, 369,
77−81.
(16) Ura, T.; Okuda, K.; Shimada, M. Developments in Viral Vector-
Based Vaccines. Vaccines 2014, 2, 624−641.
(17) Hos, B. J.; Tondini, E.; van Kasteren, S. I.; Ossendorp, F.
Approaches to Improve Chemically Defined Synthetic Peptide
Vaccines. Front. Immunol. 2018, 9, 884.
(18) Donlan, A. N.; Petri, W. A. Mucosal Immunity and the
Eradication of Polio. Science 2020, 368, 362−363.
(19) Macklin, G. R.; O’Reilly, K. M.; Grassly, N. C.; Edmunds, W. J.;
Mach, O.; Santhana Gopala Krishnan, R.; Voorman, A.; Vertefeuille, J.
F.; Abdelwahab, J.; Gumede, N.; Goel, A.; Sosler, S.; Sever, J.;
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12384
Bandyopadhyay, A. S.; Pallansch, M. A.; Nandy, R.; Mkanda, P.; Diop,
O. M.; Sutter, R. W. Evolving Epidemiology of Poliovirus Serotype 2
following Withdrawal of the Serotype 2 Oral Poliovirus Vaccine.
Science 2020, 368, 401−405.
(20) Yeh, M. T.; Bujaki, E.; Dolan, P. T.; Smith, M.; Wahid, R.;
Konz, J.; Weiner, A. J.; Bandyopadhyay, A. S.; Van Damme, P.; De
Coster, I.; Revets, H.; Macadam, A.; Andino, R. Engineering the Live-
Attenuated Polio Vaccine to Prevent Reversion to Virulence. Cell Host
Microbe 2020, 27, 736−751.
(21) Robert-Guroff, M. Replicating and Non-Replicating Viral
Vectors for Vaccine Development. Curr. Opin. Biotechnol. 2007, 18,
546−556.
(22) Zhu, F.-C.; Li, Y.-H.; Guan, X.-H.; Hou, L.-H.; Wang, W.-J.; Li,
J.-X.; Wu, S.-P.; Wang, B.-S.; Wang, Z.; Wang, L.; Jia, S.-Y.; Jiang, H.-
D.; Wang, L.; Jiang, T.; Hu, Y.; Gou, J.-B.; Xu, S.-B.; Xu, J.-J.; Wang,
X.-W.; Wang, W.; et al. Safety, Tolerability, and Immunogenicity of a
Recombinant Adenovirus Type-5 Vectored COVID-19 Vaccine: A
Dose-Escalation, Open-Label, Non-Randomised, First-in-Human
Trial. Lancet 2020, 395, 1845−1854.
(23) van Doremalen, N.; Haddock, E.; Feldmann, F.; Meade-White,
K.; Bushmaker, T.; Fischer, R. J.; Okumura, A.; Hanley, P. W.;
Saturday, G.; Edwards, N. J.; Clark, M. H. A.; Lambe, T.; Gilbert, S.
C.; Munster, V. J. A Single Dose of ChAdOx1MERS Provides
Protective Immunity in Rhesus Macaques. Sci. Adv. 2020, 6,
No. eaba8399.
(24) Gary, E. N.; Weiner, D. B. DNA Vaccines: Prime Time Is Now.
Curr. Opin. Immunol. 2020, 65, 21−27.
(25) Modjarrad, K.; Roberts, C. C.; Mills, K. T.; Castellano, A. R.;
Paolino, K.; Muthumani, K.; Reuschel, E. L.; Robb, M. L.; Racine, T.;
Oh, M.-d.; Lamarre, C.; Zaidi, F. I.; Boyer, J.; Kudchodkar, S. B.;
Jeong, M.; Darden, J. M.; Park, Y. K.; Scott, P. T.; Remigio, C.;
Parikh, A. P.; et al. Safety and Immunogenicity of an Anti-Middle East
Respiratory Syndrome Coronavirus DNA Vaccine: A Phase 1, Open-
Label, Single-Arm, Dose-Escalation Trial. Lancet Infect. Dis. 2019, 19,
1013−1022.
(26) Muthumani, K.; Falzarano, D.; Reuschel, E. L.; Tingey, C.;
Flingai, S.; Villarreal, D. O.; Wise, M.; Patel, A.; Izmirly, A.; Aljuaid,
A.; Seliga, A. M.; Soule, G.; Morrow, M.; Kraynyak, K. A.; Khan, A. S.;
Scott, D. P.; Feldmann, F.; LaCasse, R.; Meade-White, K.; Okumura,
A.; et al. A Synthetic Consensus Anti-Spike Protein DNA Vaccine
Induces Protective Immunity against Middle East Respiratory
Syndrome Coronavirus in Nonhuman Primates. Sci. Transl. Med.
2015, 7, 301−132.
(27) Pardi, N.; Hogan, M. J.; Porter, F. W.; Weissman, D. mRNA
Vaccines A New Era in Vaccinology. Nat. Rev. Drug Discovery
2018, 17, 261−279.
(28) Suschak, J. J.; Williams, J. A.; Schmaljohn, C. S. Advancements
in DNA Vaccine Vectors, Non-Mechanical Delivery Methods, and
Molecular Adjuvants to Increase Immunogenicity. Hum. Vaccines
Immunother. 2017, 13, 2837−2848.
(29) Gallie, D. R. The Cap and Poly (A) Tail Function
Synergistically to Regulate mRNA Translational Efficiency. Genes
Dev. 1991, 5, 2108−2116.
(30) Gustafsson, C.; Govindarajan, S.; Minshull, J. Codon Bias and
Heterologous Protein Expression. Trends Biotechnol. 2004, 22, 346−
353.
(31) Kariko,́ K.; Muramatsu, H.; Welsh, F. A.; Ludwig, J.; Kato, H.;
Akira, S.; Weissman, D. Incorporation of Pseudouridine into mRNA
Yields Superior Nonimmunogenic Vector with Increased Transla-
tional Capacity and Biological Stability. Mol. Ther. 2008, 16, 1833−
1840.
(32) Kariko,́ K.; Buckstein, M.; Ni, H.; Weissman, D. Suppression of
RNA Recognition by Toll-Like Receptors: the Impact of Nucleoside
Modification and the Evolutionary Origin of RNA. Immunity 2005,
23, 165−175.
(33) Chen, N.; Xia, P.; Li, S.; Zhang, T.; Wang, T. T.; Zhu, J. RNA
Sensors of the Innate Immune System and Their Detection of
Pathogens. IUBMB Life 2017, 69, 297−304.
(34) Kariko, K.; Muramatsu, H.; Ludwig, J.; Weissman, D.
Generating the Optimal mRNA for Therapy: HPLC Purification
Eliminates Immune Activation and Improves Translation of Nucleo-
side-Modified, Protein-Encoding mRNA. Nucleic Acids Res. 2011, 39,
142.
(35) Cohen, J. Designer Proteins Produce Potent HIV Defense.
Science 2015, 348, 1297.
(36) Bijker, M. S.; van den Eeden, S. J.; Franken, K. L.; Melief, C. J.;
Offringa, R.; van der Burg, S. H. CD8+ CTL Priming by Exact
Peptide Epitopes in Incomplete Freund’s Adjuvant Induces a
Vanishing CTL Response, Whereas Long Peptides Induce Sustained
CTL Reactivity. J. Immunol. 2007, 179, 5033−5040.
(37) Bijker, M. S.; van den Eeden, S. J.; Franken, K. L.; Melief, C. J.;
van der Burg, S. H.; Offringa, R. Superior Induction of Anti-Tumor
CTL Immunity by Extended Peptide Vaccines Involves Prolonged,
DC-Focused Antigen Presentation. Eur. J. Immunol. 2008, 38, 1033−
1042.
(38) Jackson, D. C.; Lau, Y. F.; Le, T.; Suhrbier, A.; Deliyannis, G.;
Cheers, C.; Smith, C.; Zeng, W.; Brown, L. E. A Totally Synthetic
Vaccine of Generic Structure That Targets Toll-Like Receptor 2 on
Dendritic Cells and Promotes Antibody or Cytotoxic T Cell
Responses. Proc. Natl. Acad. Sci. U. S. A. 2004, 101, 15440.
(39) Enayatkhani, M.; Hasaniazad, M.; Faezi, S.; Guklani, H.;
Davoodian, P.; Ahmadi, N.; Einakian, M. A.; Karmostaji, A.; Ahmadi,
K. Reverse Vaccinology Approach to Design a Novel Multi-Epitope
Vaccine Candidate against COVID-19: An In Silico Study. J. Biomol.
Struct. Dyn. 2020, 1−16.
(40) Kapsenberg, M. L. Dendritic-Cell Control of Pathogen-Driven
T-Cell Polarization. Nat. Rev. Immunol. 2003, 3, 984−993.
(41) Malowany, J. I.; McCormick, S.; Santosuosso, M.; Zhang, X.;
Aoki, N.; Ngai, P.; Wang, J.; Leitch, J.; Bramson, J.; Wan, Y.; Xing, Z.
Development of Cell-Based Tuberculosis Vaccines: Genetically
Modified Dendritic Cell Vaccine Is a Much More Potent Activator
of CD4 and CD8 T Cells Than Peptide- or Protein-Loaded
Counterparts. Mol. Ther. 2006, 13, 766−775.
(42) Benteyn, D.; Heirman, C.; Bonehill, A.; Thielemans, K.;
Breckpot, K. mRNA-Based Dendritic Cell Vaccines. Expert Rev.
Vaccines 2015, 14, 161−176.
(43) Perez, C. R.; De Palma, M. Engineering Dendritic Cell Vaccines
to Improve Cancer Immunotherapy. Nat. Commun. 2019, 10, 5408.
(44) Vela Ramirez, J. E.; Sharpe, L. A.; Peppas, N. A. Current State
and Challenges in Developing Oral Vaccines. Adv. Drug Delivery Rev.
2017, 114, 116−131.
(45) Melief, C. J. M.; van der Burg, S. H. Immunotherapy of
Established (Pre)malignant Disease by Synthetic Long Peptide
Vaccines. Nat. Rev. Cancer 2008, 8, 351−360.
(46) Napolitani, G.; Rinaldi, A.; Bertoni, F.; Sallusto, F.;
Lanzavecchia, A. Selected Toll-Like Receptor Agonist Combinations
Synergistically Trigger a T Helper Type 1-Polarizing Program in
Dendritic Cells. Nat. Immunol. 2005, 6, 769−776.
(47) Coffman, R. L.; Sher, A.; Seder, R. A. Vaccine Adjuvants:
Putting Innate Immunity to Work. Immunity 2010, 33, 492−503.
(48) Stephan, M. T.; Moon, J. J.; Um, S. H.; Bershteyn, A.; Irvine, D.
J. Therapeutic Cell Engineering with Surface-Conjugated Synthetic
Nanoparticles. Nat. Med. 2010, 16, 1035−1041.
(49) Yong, C. Y.; Ong, H. K.; Yeap, S. K.; Ho, K. L.; Tan, W. S.
Recent Advances in the Vaccine Development against Middle East
Respiratory Syndrome-Coronavirus. Front. Microbiol. 2019, 10, 1781.
(50) Hajj, K. A.; Whitehead, K. A. Tools for Translation: Non-Viral
Materials for Therapeutic mRNA Delivery. Nat. Rev. Mater. 2017, 2,
17056.
(51) Moyer, T. J.; Zmolek, A. C.; Irvine, D. J. Beyond Antigens and
Adjuvants: Formulating Future Vaccines. J. Clin. Invest. 2016, 126,
799−808.
(52) Sun, W.; Hu, Q.; Ji, W.; Wright, G.; Gu, Z. Leveraging
Physiology for Precision Drug Delivery. Physiol. Rev. 2017, 97, 189−
225.
(53) Moon, J. J.; Huang, B.; Irvine, D. J. Engineering Nano-and
Microparticles to Tune Immunity. Adv. Mater. 2012, 24, 3724−3746.
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12385
(54) Oberli, M. A.; Reichmuth, A. M.; Dorkin, J. R.; Mitchell, M. J.;
Fenton, O. S.; Jaklenec, A.; Anderson, D. G.; Langer, R.; Blankschtein,
D. Lipid Nanoparticle Assisted mRNA Delivery for Potent Cancer
Immunotherapy. Nano Lett. 2017, 17, 1326−1335.
(55) Moon, J. J.; Suh, H.; Bershteyn, A.; Stephan, M. T.; Liu, H.;
Huang, B.; Sohail, M.; Luo, S.; Ho Um, S.; Khant, H.; Goodwin, J. T.;
Ramos, J.; Chiu, W.; Irvine, D. J. Interbilayer-Crosslinked Multi-
lamellar Vesicles as Synthetic Vaccines for Potent Humoral and
Cellular Immune Responses. Nat. Mater. 2011, 10, 243−251.
(56) Kranz, L. M.; Diken, M.; Haas, H.; Kreiter, S.; Loquai, C.;
Reuter, K. C.; Meng, M.; Fritz, D.; Vascotto, F.; Hefesha, H.;
Grunwitz, C.; Vormehr, M.; Hüsemann, Y.; Selmi, A.; Kuhn, A. N.;
Buck, J.; Derhovanessian, E.; Rae, R.; Attig, S.; Diekmann, J.; et al.
Systemic RNA Delivery to Dendritic Cells Exploits Antiviral Defence
for Cancer Immunotherapy. Nature 2016, 534, 396−401.
(57) Irvine, D. J.; Read, B. J. Shaping Humoral Immunity to
Vaccines through Antigen-Displaying Nanoparticles. Curr. Opin.
Immunol. 2020, 65, 1−6.
(58) Kuai, R.; Sun, X.; Yuan, W.; Ochyl, L. J.; Xu, Y.; Hassani
Najafabadi, A.; Scheetz, L.; Yu, M.-Z.; Balwani, I.; Schwendeman, A.;
Moon, J. J. Dual TLR Agonist Nanodiscs as a Strong Adjuvant System
for Vaccines and Immunotherapy. J. Controlled Release 2018, 282,
131−139.
(59) Miao, L.; Li, L.; Huang, Y.; Delcassian, D.; Chahal, J.; Han, J.;
Shi, Y.; Sadtler, K.; Gao, W.; Lin, J.; Doloff, J. C.; Langer, R.;
Anderson, D. G. Delivery of mRNA Vaccines with Heterocyclic Lipids
Increases Anti-Tumor Efficacy by STING-Mediated Immune Cell
Activation. Nat. Biotechnol. 2019, 37, 1174−1185.
(60) Chahal, J. S.; Khan, O. F.; Cooper, C. L.; McPartlan, J. S.;
Tsosie, J. K.; Tilley, L. D.; Sidik, S. M.; Lourido, S.; Langer, R.; Bavari,
S.; Ploegh, H. L.; Anderson, D. G. Dendrimer-RNA Nanoparticles
Generate Protective Immunity against Lethal Ebola, H1N1 Influenza,
and Toxoplasma Gondii Challenges with a Single Dose. Proc. Natl.
Acad. Sci. U. S. A. 2016, 113, E4133−42.
(61) Zeng, Q.; Jiang, H.; Wang, T.; Zhang, Z.; Gong, T.; Sun, X.
Cationic Micelle Delivery of Trp2 Peptide for Efficient Lymphatic
Draining and Enhanced Cytotoxic T-Lymphocyte Responses. J.
Controlled Release 2015, 200, 1−12.
(62) Zhao, M.; Li, M.; Zhang, Z.; Gong, T.; Sun, X. Induction of
HIV-1 Gag Specific Immune Responses by Cationic Micelles
Mediated Delivery of Gag mRNA. Drug Delivery 2016, 23, 2596−
2607.
(63) Li, M.; Zhao, M.; Fu, Y.; Li, Y.; Gong, T.; Zhang, Z.; Sun, X.
Enhanced Intranasal Delivery of mRNA Vaccine by Overcoming the
Nasal Epithelial Barrier via Intra- and Paracellular Pathways. J.
Controlled Release 2016, 228, 9−19.
(64) Demento, S. L.; Cui, W.; Criscione, J. M.; Stern, E.; Tulipan, J.;
Kaech, S. M.; Fahmy, T. M. Role of Sustained Antigen Release from
Nanoparticle Vaccines in Shaping the T Cell Memory Phenotype.
Biomaterials 2012, 33, 4957−4964.
(65) Gu, P.; Wusiman, A.; Zhang, Y.; Liu, Z.; Bo, R.; Hu, Y.; Liu, J.;
Wang, D. Rational Design of PLGA Nanoparticle Vaccine Delivery
Systems to Improve Immune Responses. Mol. Pharmaceutics 2019, 16,
5000−5012.
(66) Chen, X.; Liu, Y.; Wang, L.; Liu, Y.; Zhang, W.; Fan, B.; Ma, X.;
Yuan, Q.; Ma, G.; Su, Z. Enhanced Humoral and Cell-Mediated
Immune Responses Generated by Cationic Polymer-Coated PLA
Microspheres with Adsorbed HBsAg. Mol. Pharmaceutics 2014, 11,
1772−1784.
(67) Chen, H.; Li, P.; Yin, Y.; Cai, X.; Huang, Z.; Chen, J.; Dong, L.;
Zhang, J. The Promotion of Type 1 T Helper Cell Responses to
Cationic Polymers in Vivo via Toll-Like Receptor-4 Mediated IL-12
Secretion. Biomaterials 2010, 31, 8172−8180.
(68) Fields, R. J.; Cheng, C. J.; Quijano, E.; Weller, C.; Kristofik, N.;
Duong, N.; Hoimes, C.; Egan, M. E.; Saltzman, W. M. Surface
Modified Poly(β Amino Ester)-Containing Nanoparticles for Plasmid
DNA Delivery. J. Controlled Release 2012, 164, 41−48.
(69) Kowalski, P. S.; Capasso Palmiero, U.; Huang, Y.; Rudra, A.;
Langer, R.; Anderson, D. G. Ionizable Amino-Polyesters Synthesized
via Ring Opening Polymerization of Tertiary Amino-Alcohols for
Tissue Selective mRNA Delivery. Adv. Mater. 2018, 30, 1801151.
(70) Fornaguera, C.; Guerra-Rebollo, M.; Ángel Laźaro, M.;
Castells-Sala, C.; Meca-Corte
́
s, O.; Ramos-Pe
́
rez, V.; Cascante, A.;
Rubio, N.; Blanco, J.; Borro
́
s, S. mRNA Delivery System for Targeting
Antigen-Presenting Cells in Vivo. Adv. Healthcare Mater. 2018, 7,
1800335.
(71) Liu, H.; Moynihan, K. D.; Zheng, Y.; Szeto, G. L.; Li, A. V.;
Huang, B.; Van Egeren, D. S.; Park, C.; Irvine, D. J. Structure-Based
Programming of Lymph-Node Targeting in Molecular Vaccines.
Nature 2014, 507, 519−522.
(72) Son, S.; Nam, J.; Zenkov, I.; Ochyl, L. J.; Xu, Y.; Scheetz, L.;
Shi, J.; Farokhzad, O. C.; Moon, J. J. Sugar-Nanocapsules Imprinted
with Microbial Molecular Patterns for mRNA Vaccination. Nano Lett.
2020, 20, 1499−1509.
(73) Moyer, T. J.; Kato, Y.; Abraham, W.; Chang, J. Y. H.; Kulp, D.
W.; Watson, N.; Turner, H. L.; Menis, S.; Abbott, R. K.; Bhiman, J.
N.; Melo, M. B.; Simon, H. A.; Herrera-De la Mata, S.; Liang, S.;
Seumois, G.; Agarwal, Y.; Li, N.; Burton, D. R.; Ward, A. B.; Schief,
W. R.; et al. Engineered Immunogen Binding to Alum Adjuvant
Enhances Humoral Immunity. Nat. Med. 2020, 26, 430−440.
(74) Moyano, D. F.; Goldsmith, M.; Solfiell, D. J.; Landesman-Milo,
D.; Miranda, O. R.; Peer, D.; Rotello, V. M. Nanoparticle
Hydrophobicity Dictates Immune Response. J. Am. Chem. Soc.
2012, 134, 3965−3967.
(75) Xu, L.; Liu, Y.; Chen, Z.; Li, W.; Liu, Y.; Wang, L.; Liu, Y.; Wu,
X.; Ji, Y.; Zhao, Y.; Ma, L.; Shao, Y.; Chen, C. Surface-Engineered
Gold Nanorods: Promising DNA Vaccine Adjuvant for HIV-1
Treatment. Nano Lett. 2012, 12, 2003−2012.
(76) Qiu, Y.; Liu, Y.; Wang, L.; Xu, L.; Bai, R.; Ji, Y.; Wu, X.; Zhao,
Y.; Li, Y.; Chen, C. Surface Chemistry and Aspect Ratio Mediated
Cellular Uptake of Au Nanorods. Biomaterials 2010, 31, 7606−7619.
(77) Zhou, Q.; Zhang, Y.; Du, J.; Li, Y.; Zhou, Y.; Fu, Q.; Zhang, J.;
Wang, X.; Zhan, L. Different-Sized Gold Nanoparticle Activator/
Antigen Increases Dendritic Cells Accumulation in Liver-Draining
Lymph Nodes and CD8+ T Cell Responses. ACS Nano 2016, 10,
2678−2692.
(78) Mercuri, L. P.; Carvalho, L. V.; Lima, F. A.; Quayle, C.; Fantini,
M. C. A.; Tanaka, G. S.; Cabrera, W. H.; Furtado, M. F. D.;
Tambourgi, D. V.; Matos, J. d. R.; Jaroniec, M.; Sant’Anna, O. A.
Ordered Mesoporous Silica SBA-15: A New Effective Adjuvant to
Induce Antibody Response. Small 2006, 2, 254−256.
(79) Mahony, D.; Cavallaro, A. S.; Mody, K. T.; Xiong, L.; Mahony,
T. J.; Qiao, S. Z.; Mitter, N. In Vivo Delivery of Bovine Viral
Diahorrea Virus, E2 Protein Using Hollow Mesoporous Silica
Nanoparticles. Nanoscale 2014, 6, 6617−6626.
(80) Hess, K. L.; Oh, E.; Tostanoski, L. H.; Andorko, J. I.; Susumu,
K.; Deschamps, J. R.; Medintz, I. L.; Jewell, C. M. Engineering
Immunological Tolerance Using Quantum Dots to Tune the Density
of Self-Antigen Display. Adv. Funct. Mater. 2017, 27, 1700290.
(81) Hu, Z.; Song, B.; Xu, L.; Zhong, Y.; Peng, F.; Ji, X.; Zhu, F.;
Yang, C.; Zhou, J.; Su, Y.; Chen, S.; He, Y.; He, S. Aqueous
Synthesized Quantum Dots Interfere with the NF-κB Pathway and
Confer Anti-Tumor, Anti-Viral and Anti-Inflammatory Effects.
Biomaterials 2016, 108, 187−196.
(82) Faria, P. C. B. d.; Santos, L. I. d.; Coelho, J. P.; Ribeiro, H. B.;
Pimenta, M. A.; Ladeira, L. O.; Gomes, D. A.; Furtado, C. A.;
Gazzinelli, R. T. Oxidized Multiwalled Carbon Nanotubes as Antigen
Delivery System to Promote Superior CD8+ T Cell Response and
Protection against Cancer. Nano Lett. 2014, 14, 5458−5470.
(83) Zhang, L.; Chan, J. M.; Gu, F. X.; Rhee, J.-W.; Wang, A. Z.;
Radovic-Moreno, A. F.; Alexis, F.; Langer, R.; Farokhzad, O. C. Self-
Assembled Lipid-Polymer Hybrid Nanoparticles: A Robust Drug
Delivery Platform. ACS Nano 2008, 2, 1696−1702.
(84) Walsh, C. L.; Nguyen, J.; Tiffany, M. R.; Szoka, F. C. Synthesis,
Characterization, and Evaluation of Ionizable Lysine-Based Lipids for
siRNA Delivery. Bioconjugate Chem. 2013, 24, 36−43.
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12386
(85) Jokerst, J. V.; Lobovkina, T.; Zare, R. N.; Gambhir, S. S.
Nanoparticle PEGylation for Imaging and Therapy. Nanomedicine
2011, 6, 715−728.
(86) Beltra
́
n-Gracia, E.; Lo
́
pez-Camacho, A.; Higuera-Ciapara, I.;
Vela
́
zquez-Ferna
́
ndez, J. B.; Vallejo-Cardona, A. A. Nanomedicine
Review: Clinical Developments in Liposomal Applications. Cancer
Nanotechnol. 2019, 10, 11.
(87) Tam, H. H.; Melo, M. B.; Kang, M.; Pelet, J. M.; Ruda, V. M.;
Foley, M. H.; Hu, J. K.; Kumari, S.; Crampton, J.; Baldeon, A. D.;
Sanders, R. W.; Moore, J. P.; Crotty, S.; Langer, R.; Anderson, D. G.;
Chakraborty, A. K.; Irvine, D. J. Sustained Antigen Availability during
Germinal Center Initiation Enhances Antibody Responses to
Vaccination. Proc. Natl. Acad. Sci. U. S. A. 2016, 113, E6639−E6648.
(88) Diebold, S. S.; Kaisho, T.; Hemmi, H.; Akira, S.; Reis e Sousa,
C. Innate Antiviral Responses by Means of TLR7-Mediated
Recognition of Single-Stranded RNA. Science 2004, 303, 1529−1531.
(89) Kuai, R.; Ochyl, L. J.; Bahjat, K. S.; Schwendeman, A.; Moon, J.
J. Designer Vaccine Nanodiscs for Personalized Cancer Immunother-
apy. Nat. Mater. 2017, 16, 489−496.
(90) Dobrovolskaia, M. A.; McNeil, S. E. Immunological Properties
of Engineered Nanomaterials. Nat. Nanotechnol. 2007, 2, 469−478.
(91) Akinc, A.; Thomas, M.; Klibanov, A. M.; Langer, R. Exploring
Polyethylenimine-Mediated DNA Transfection and the Proton
Sponge Hypothesis. J. Gene Med. 2005, 7, 657−663.
(92) Boussif, O.; Lezoualch, F.; Zanta, M. A.; Mergny, M. D.;
Scherman, D.; Demeneix, B.; Behr, J. P. A Versatile Vector for Gene
and Oligonucleotide Transfer into Cells in Culture and in Vivo:
Polyethylenimine. Proc. Natl. Acad. Sci. U. S. A. 1995, 92, 7297−7301.
(93) Anderson, J. M.; Shive, M. S. Biodegradation and
Biocompatibility of PLA and PLGA Microspheres. Adv. Drug Delivery
Rev. 2012, 64, 72−82.
(94) Sunshine, J. C.; Perica, K.; Schneck, J. P.; Green, J. J. Particle
Shape Dependence of CD8+ T Cell Activation by Artificial Antigen
Presenting Cells. Biomaterials 2014, 35, 269−277.
(95) Wang, Q.; Tan, M. T.; Keegan, B. P.; Barry, M. A.; Heffernan,
M. J. Time Course Study of the Antigen-Specific Immune Response
to a PLGA Microparticle Vaccine Formulation. Biomaterials 2014, 35,
8385−8393.
(96) Chan, J. M.; Zhang, L.; Yuet, K. P.; Liao, G.; Rhee, J.-W.;
Langer, R.; Farokhzad, O. C. PLGA-Lecithin-PEG Core-Shell
Nanoparticles for Controlled Drug Delivery. Biomaterials 2009, 30,
1627−1634.
(97) Sun, W.; Wang, J.; Hu, Q.; Zhou, X.; Khademhosseini, A.; Gu,
Z. CRISPR-Cas12a Delivery by DNA-Mediated Bioresponsive Editing
for Cholesterol Regulation. Sci. Adv. 2020, 6, No. eaba2983.
(98) Dong, L.; Xia, S.; Chen, H.; Chen, J.; Zhang, J. Anti-Arthritis
Activity of Cationic Materials. J. Cell. Mol. Med. 2010, 14, 2015−2024.
(99) Little, S. R.; Lynn, D. M.; Ge, Q.; Anderson, D. G.; Puram, S.
V.; Chen, J.; Eisen, H. N.; Langer, R. Poly-β Amino Ester-Containing
Microparticles Enhance the Activity of Nonviral Genetic Vaccines.
Proc. Natl. Acad. Sci. U. S. A. 2004, 101, 9534−9539.
(100) Liu, Z.; Jiao, Y.; Wang, Y.; Zhou, C.; Zhang, Z.
Polysaccharides-Based Nanoparticles as Drug Delivery Systems. Adv.
Drug Delivery Rev. 2008, 60, 1650−1662.
(101) Acharya, A. P.; Sinha, M.; Ratay, M. L.; Ding, X.; Balmert, S.
C.; Workman, C. J.; Wang, Y.; Vignali, D. A. A.; Little, S. R. Localized
Multi-Component Delivery Platform Generates Local and Systemic
Anti-Tumor Immunity. Adv. Funct. Mater. 2017, 27, 1604366.
(102) Dosta, P.; Segovia, N.; Cascante, A.; Ramos, V.; Borro
́
s, S.
Surface Charge Tunability as a Powerful Strategy to Control
Electrostatic Interaction for High Efficiency Silencing, Using Tailored
Oligopeptide-Modified Poly(Beta-Amino Ester)s (PBAEs). Acta
Biomater. 2015, 20, 82−93.
(103) Tsopelas, C.; Sutton, R. Why Certain Dyes Are Useful for
Localizing the Sentinel Lymph Node. J. Nucl. Med. 2002, 43, 1377−
1382.
(104) Seong, S.-Y.; Matzinger, P. Hydrophobicity: An Ancient
Damage-Associated Molecular Pattern That Initiates Innate Immune
Responses. Nat. Rev. Immunol. 2004, 4, 469−478.
(105) Shokouhi, B.; Coban, C.; Hasirci, V.; Aydin, E.; Dhanasingh,
A.; Shi, N.; Koyama, S.; Akira, S.; Zenke, M.; Sechi, A. S. The Role of
Multiple Toll-Like Receptor Signalling Cascades on Interactions
between Biomedical Polymers and Dendritic Cells. Biomaterials 2010,
31, 5759−5771.
(106) Glenny, A.; Pope, C.; Waddington, H.; Wallace, U.
Immunological Notes. xvii-xxiv. J. Pathol. Bacteriol. 1926, 29, 31−40.
(107) Levitz, S. M.; Golenbock, D. T. Beyond Empiricism:
Informing Vaccine Development through Innate Immunity Research.
Cell 2012, 148, 1284−1292.
(108) Mannhalter, J.; Neychev, H.; Zlabinger, G.; Ahmad, R.; Eibl,
M. Modulation of the Human Immune Response by the Non-Toxic
and Non-Pyrogenic Adjuvant Aluminium Hydroxide: Effect on
Antigen Uptake and Antigen Presentation. Clin. Exp. Immunol.
1985, 61, 143−151.
(109) McKee, A. S.; Munks, M. W.; Marrack, P. How Do Adjuvants
Work? Important Considerations for New Generation Adjuvants.
Immunity 2007, 27, 687−690.
(110) Hornung, V.; Bauernfeind, F.; Halle, A.; Samstad, E. O.; Kono,
H.; Rock, K. L.; Fitzgerald, K. A.; Latz, E. Silica Crystals and
Aluminum Salts Activate the NALP3 Inflammasome through
Phagosomal Destabilization. Nat. Immunol. 2008, 9, 847−856.
(111) Hess, K. L.; Medintz, I. L.; Jewell, C. M. Designing Inorganic
Nanomaterials for Vaccines and Immunotherapies. Nano Today 2019,
27, 73−98.
(112) Reddy, S. T.; van der Vlies, A. J.; Simeoni, E.; Angeli, V.;
Randolph, G. J.; O’Neil, C. P.; Lee, L. K.; Swartz, M. A.; Hubbell, J. A.
Exploiting Lymphatic Transport and Complement Activation in
Nanoparticle Vaccines. Nat. Biotechnol. 2007, 25, 1159−1164.
(113) Sokolova, V.; Epple, M. Inorganic Nanoparticles as Carriers of
Nucleic Acids into Cells. Angew. Chem., Int. Ed. 2008, 47, 1382−1395.
(114) Moon, J. J.; Suh, H.; Li, A. V.; Ockenhouse, C. F.; Yadava, A.;
Irvine, D. J. Enhancing Humoral Responses to a Malaria Antigen with
Nanoparticle Vaccines That Expand Tfh Cells and Promote Germinal
Center Induction. Proc. Natl. Acad. Sci. U. S. A. 2012, 109, 1080−
1085.
(115) Lin, W.-H. W.; Kouyos, R. D.; Adams, R. J.; Grenfell, B. T.;
Griffin, D. E. Prolonged Persistence of Measles Virus RNA Is
Characteristic of Primary Infection Dynamics. Proc. Natl. Acad. Sci. U.
S. A. 2012, 109, 14989−14994.
(116) Pape, K. A.; Catron, D. M.; Itano, A. A.; Jenkins, M. K. The
Humoral Immune Response Is Initiated in Lymph Nodes by B Cells
That Acquire Soluble Antigen Directly in the Follicles. Immunity
2007, 26, 491−502.
(117) McHugh, K. J.; Guarecuco, R.; Langer, R.; Jaklenec, A. Single-
Injection Vaccines: Progress, Challenges, and Opportunities. J.
Controlled Release 2015, 219, 596−609.
(118) Zhu, Q.; Zarnitsyn, V. G.; Ye, L.; Wen, Z.; Gao, Y.; Pan, L.;
Skountzou, I.; Gill, H. S.; Prausnitz, M. R.; Yang, C.; Compans, R. W.
Immunization by Vaccine-Coated Microneedle Arrays Protects
against Lethal Influenza Virus Challenge. Proc. Natl. Acad. Sci. U. S.
A. 2009, 106, 7968−7973.
(119) Sullivan, S. P.; Koutsonanos, D. G.; del Pilar Martin, M.; Lee,
J. W.; Zarnitsyn, V.; Choi, S.-O.; Murthy, N.; Compans, R. W.;
Skountzou, I.; Prausnitz, M. R. Dissolving Polymer Microneedle
Patches for Influenza Vaccination. Nat. Med. 2010, 16, 915−920.
(120) Kim, E.; Erdos, G.; Huang, S.; Kenniston, T. W.; Balmert, S.
C.; Carey, C. D.; Raj, V. S.; Epperly, M. W.; Klimstra, W. B.;
Haagmans, B. L.; Korkmaz, E.; Falo, L. D.; Gambotto, A. Microneedle
Array Delivered Recombinant Coronavirus Vaccines: Immunogenicity
and Rapid Translational Development. EBioMedicine 2020, 55,
102743.
(121) Rouphael, N. G.; Paine, M.; Mosley, R.; Henry, S.; McAllister,
D. V.; Kalluri, H.; Pewin, W.; Frew, P. M.; Yu, T.; Thornburg, N. J.;
Kabbani, S.; Lai, L.; Vassilieva, E. V.; Skountzou, I.; Compans, R. W.;
Mulligan, M. J.; Prausnitz, M. R.; Beck, A.; Edupuganti, S.; Heeke, S.;
et al. The Safety, Immunogenicity, and Acceptability of Inactivated
Influenza Vaccine Delivered by Microneedle Patch (TIV-MNP
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12387
2015): A Randomised, Partly Blinded, Placebo-Controlled, Phase 1
Trial. Lancet 2017, 390, 649−658.
(122) Boopathy, A. V.; Mandal, A.; Kulp, D. W.; Menis, S.; Bennett,
N. R.; Watkins, H. C.; Wang, W.; Martin, J. T.; Thai, N. T.; He, Y.;
Schief, W. R.; Hammond, P. T.; Irvine, D. J. Enhancing Humoral
Immunity via Sustained-Release Implantable Microneedle Patch
Vaccination. Proc. Natl. Acad. Sci. U. S. A. 2019, 116, 16473−16478.
(123) DeMuth, P. C.; Moon, J. J.; Suh, H.; Hammond, P. T.; Irvine,
D. J. Releasable Layer-by-Layer Assembly of Stabilized Lipid
Nanocapsules on Microneedles for Enhanced Transcutaneous
Vaccine Delivery. ACS Nano 2012, 6, 8041−8051.
(124) Zaric, M.; Lyubomska, O.; Touzelet, O.; Poux, C.; Al-Zahrani,
S.; Fay, F.; Wallace, L.; Terhorst, D.; Malissen, B.; Henri, S.; Power,
U. F.; Scott, C. J.; Donnelly, R. F.; Kissenpfennig, A. Skin Dendritic
Cell Targeting via Microneedle Arrays Laden with Antigen-
Encapsulated Poly-d, l-lactide-co-Glycolide Nanoparticles Induces
Efficient Antitumor and Antiviral Immune Responses. ACS Nano
2013, 7, 2042−2055.
(125) Bae, W.-G.; Ko, H.; So, J.-Y.; Yi, H.; Lee, C.-H.; Lee, D.-H.;
Ahn, Y.; Lee, S.-H.; Lee, K.; Jun, J.; Kim, H.-H.; Jeon, N. L.; Jung, W.;
Song, C.-S.; Kim, T.; Kim, Y.-C.; Jeong, H. E. Snake Fang-Inspired
Stamping Patch for Transdermal Delivery of Liquid Formulations. Sci.
Transl. Med. 2019, 11, No. eaaw3329.
(126) Tzeng, S. Y.; McHugh, K. J.; Behrens, A. M.; Rose, S.;
Sugarman, J. L.; Ferber, S.; Langer, R.; Jaklenec, A. Stabilized Single-
Injection Inactivated Polio Vaccine Elicits a Strong Neutralizing
Immune Response. Proc. Natl. Acad. Sci. U. S. A. 2018, 115, E5269−
E5278.
(127) McHugh, K. J.; Nguyen, T. D.; Linehan, A. R.; Yang, D.;
Behrens, A. M.; Rose, S.; Tochka, Z. L.; Tzeng, S. Y.; Norman, J. J.;
Anselmo, A. C.; Xu, X.; Tomasic, S.; Taylor, M. A.; Lu, J.; Guarecuco,
R.; Langer, R.; Jaklenec, A. Fabrication of Fillable Microparticles and
Other Complex 3D Microstructures. Science 2017, 357, 1138−1142.
(128) Lu, X.; Miao, L.; Gao, W.; Chen, Z.; McHugh, K. J.; Sun, Y.;
Tochka, Z.; Tomasic, S.; Sadtler, K.; Hyacinthe, A.; Huang, Y.; Graf,
T.; Hu, Q.; Sarmadi, M.; Langer, R.; Anderson, D. G.; Jaklenec, A.
Engineered PLGA Microparticles for Long-Term, Pulsatile Release of
STING Agonist for Cancer Immunotherapy. Sci. Transl. Med. 2020,
12, No. eaaz6606.
(129) Ali, O. A.; Huebsch, N.; Cao, L.; Dranoff, G.; Mooney, D. J.
Infection-Mimicking Materials to Program Dendritic Cells in Situ.
Nat. Mater. 2009, 8, 151−158.
(130) Kim, J.; Li, W. A.; Choi, Y.; Lewin, S. A.; Verbeke, C. S.;
Dranoff, G.; Mooney, D. J. Injectable, Spontaneously Assembling,
Inorganic Scaffolds Modulate Immune Cells in Vivo and Increase
Vaccine Efficacy. Nat. Biotechnol. 2015, 33, 64−72.
(131) Li, A. W.; Sobral, M. C.; Badrinath, S.; Choi, Y.; Graveline, A.;
Stafford, A. G.; Weaver, J. C.; Dellacherie, M. O.; Shih, T.-Y.; Ali, O.
A.; Kim, J.; Wucherpfennig, K. W.; Mooney, D. J. A Facile Approach
to Enhance Antigen Response for Personalized Cancer Vaccination.
Nat. Mater. 2018, 17, 528−534.
(132) Dellacherie, M. O.; Li, A.; Lu, B. Y.; Verbeke, C. S.; Gu, L.;
Stafford, A. G.; Doherty, E. J.; Mooney, D. J. Single-Shot Mesoporous
Silica Rods Scaffold for Induction of Humoral Responses against
Small Antigens. Adv. Funct. Mater. 2020, 30, 2002448.
(133) Shih, T.-Y.; Blacklow, S. O.; Li, A. W.; Freedman, B. R.;
Bencherif, S.; Koshy, S. T.; Darnell, M. C.; Mooney, D. J. Injectable,
Tough Alginate Cryogels as Cancer Vaccines. Adv. Healthcare Mater.
2018, 7, 1701469.
(134) Bencherif, S. A.; Sands, R. W.; Bhatta, D.; Arany, P.; Verbeke,
C. S.; Edwards, D. A.; Mooney, D. J. Injectable Preformed Scaffolds
with Shape-Memory Properties. Proc. Natl. Acad. Sci. U. S. A. 2012,
109, 19590−19595.
(135) Hori, Y.; Winans, A. M.; Huang, C. C.; Horrigan, E. M.;
Irvine, D. J. Injectable Dendritic Cell-Carrying Alginate Gels for
Immunization and Immunotherapy. Biomaterials 2008, 29, 3671−
3682.
(136) Guo, Z.; Zhang, M.; Tang, H.; Cao, X. Fas Signal Links Innate
and Adaptive Immunity by Promoting Dendritic-Cell Secretion of CC
and CXC Chemokines. Blood 2005, 106, 2033−2041.
(137) Hori, Y.; Winans, A. M.; Irvine, D. J. Modular Injectable
Matrices Based on Alginate Solution/Microsphere Mixtures That Gel
in Situ and Co-deliver Immunomodulatory factors. Acta Biomater.
2009, 5, 969−982.
(138) Roy, K.; Wang, D.; Hedley, M. L.; Barman, S. P. Gene
Delivery with In-Situ Crosslinking Polymer Networks Generates
Long-Term Systemic Protein Expression. Mol. Ther. 2003, 7, 401−
408.
(139) Wu, Y.; Wei, W.; Zhou, M.; Wang, Y.; Wu, J.; Ma, G.; Su, Z.
Thermal-Sensitive Hydrogel as Adjuvant-Free Vaccine Delivery
System for H5N1 Intranasal Immunization. Biomaterials 2012, 33,
2351−2360.
(140) Hiemstra, C.; van der Aa, L. J.; Zhong, Z.; Dijkstra, P. J.;
Feijen, J. Rapidly in Situ-Forming Degradable Hydrogels from
Dextran Thiols through Michael Addition. Biomacromolecules 2007,
8, 1548−1556.
(141) Singh, A.; Suri, S.; Roy, K. In-Situ Crosslinking Hydrogels for
Combinatorial Delivery of Chemokines and siRNA-DNA Carrying
Microparticles to Dendritic Cells. Biomaterials 2009, 30, 5187−5200.
(142) Singh, A.; Qin, H.; Fernandez, I.; Wei, J.; Lin, J.; Kwak, L. W.;
Roy, K. An Injectable Synthetic Immune-Priming Center Mediates
Efficient T-Cell Class Switching and T-Helper 1 Response against B
Cell Lymphoma. J. Controlled Release 2011, 155, 184−192.
(143) Umeki, Y.; Mohri, K.; Kawasaki, Y.; Watanabe, H.; Takahashi,
R.; Takahashi, Y.; Takakura, Y.; Nishikawa, M. Induction of Potent
Antitumor Immunity by Sustained Release of Cationic Antigen from a
DNA-Based Hydrogel with Adjuvant Activity. Adv. Funct. Mater.
2015, 25, 5758−5767.
(144) Prausnitz, M. R.; Mikszta, J. A.; Cormier, M.; Andrianov, A. K.
Microneedle-Based Vaccines. Curr. Top. Microbiol. Immunol. 2009,
333, 369−393.
(145) Chen, G.; Chen, Z.; Wen, D.; Wang, Z.; Li, H.; Zeng, Y.;
Dotti, G.; Wirz, R. E.; Gu, Z. Transdermal Cold Atmospheric Plasma-
Mediated Immune Checkpoint Blockade Therapy. Proc. Natl. Acad.
Sci. U. S. A. 2020, 117, 3687−3692.
(146) Ye, Y.; Wang, C.; Zhang, X.; Hu, Q.; Zhang, Y.; Liu, Q.; Wen,
D.; Milligan, J.; Bellotti, A.; Huang, L.; Dotti, G.; Gu, Z. A Melanin-
Mediated Cancer Immunotherapy Patch. Sci. Immunol. 2017, 2,
No. eaan5692.
(147) Erdos, G.; Balmert, S. C.; Carey, C. D.; Falo, G. D.; Patel, N.
A.; Zhang, J.; Gambotto, A.; Korkmaz, E.; Falo, L. D. Improved
Cutaneous Genetic Immunization by Microneedle Array Delivery of
an Adjuvanted Adenovirus Vaccine. J. Invest. Dermatol. 2020,
DOI: 10.1016/j.jid.2020.03.966.
(148) Lee, K.; Goudie, M. J.; Tebon, P.; Sun, W.; Luo, Z.; Lee, J.;
Zhang, S.; Fetah, K.; Kim, H.-J.; Xue, Y.; Darabi, M. A.; Ahadian, S.;
Sarikhani, E.; Ryu, W.; Gu, Z.; Weiss, P. S.; Dokmeci, M. R.;
Ashammakhi, N.; Khademhosseini, A., Non-Transdermal Micro-
needles for Advanced Drug Delivery. Adv. Drug Delivery Rev. 2019,
DOI: 10.1016/j.addr.2019.11.010.
(149) van der Maaden, K.; Jiskoot, W.; Bouwstra, J. Microneedle
Technologies for (Trans)dermal Drug and Vaccine Delivery. J.
Controlled Release 2012, 161, 645−655.
(150) Kim, Y.-C.; Quan, F.-S.; Compans, R. W.; Kang, S.-M.;
Prausnitz, M. R. Stability Kinetics of Influenza Vaccine Coated onto
Microneedles during Drying and Storage. Pharm. Res. 2011, 28, 135−
144.
(151) Sanders, R. W.; van Gils, M. J.; Derking, R.; Sok, D.; Ketas, T.
J.; Burger, J. A.; Ozorowski, G.; Cupo, A.; Simonich, C.; Goo, L.;
Arendt, H.; Kim, H. J.; Lee, J. H.; Pugach, P.; Williams, M.; Debnath,
G.; Moldt, B.; van Breemen, M. J.; Isik, G.; Medina-Ramírez, M.; et al.
HIV-1 Neutralizing Antibodies Induced by Native-Like Envelope
Trimers. Science 2015, 349, No. aac4223.
(152) Wang, W. Lyophilization and Development of Solid Protein
Pharmaceuticals. Int. J. Pharm. 2000, 203, 1−60.
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12388
(153) Dellacherie, M. O.; Seo, B. R.; Mooney, D. J. Macroscale
Biomaterials Strategies for Local Immunomodulation. Nat. Rev. Mater.
2019, 4, 379−397.
(154) Wu, Y.; Jane Grande-Allen, K.; West, J. L. Adhesive Peptide
Sequences Regulate Valve Interstitial Cell Adhesion, Phenotype and
Extracellular Matrix Deposition. Cell. Mol. Bioeng. 2016, 9, 479−495.
(155) Diehl, M. C.; Lee, J. C.; Daniels, S. E.; Tebas, P.; Khan, A. S.;
Giffear, M.; Sardesai, N. Y.; Bagarazzi, M. L. Tolerability of
Intramuscular and Intradermal Delivery by CELLECTRA® Adaptive
Constant Current Electroporation Device in Healthy Volunteers.
Hum. Vaccines Immunother. 2013, 9, 2246−2252.
(156) Smith, T. R. F.; Patel, A.; Ramos, S.; Elwood, D.; Zhu, X.; Yan,
J.; Gary, E. N.; Walker, S. N.; Schultheis, K.; Purwar, M.; Xu, Z.;
Walters, J.; Bhojnagarwala, P.; Yang, M.; Chokkalingam, N.; Pezzoli,
P.; Parzych, E.; Reuschel, E. L.; Doan, A.; Tursi, N.; et al.
Immunogenicity of a DNA Vaccine Candidate for COVID-19. Nat.
Commun. 2020, 11, 2601.
(157) Choi, S.-O.; Kim, Y.-C.; Lee, J. W.; Park, J.-H.; Prausnitz, M.
R.; Allen, M. G. Intracellular Protein Delivery and Gene Transfection
by Electroporation Using a Microneedle Electrode Array. Small 2012,
8, 1081−1091.
(158) Banga, A. K.; Bose, S.; Ghosh, T. K. Iontophoresis and
Electroporation: Comparisons and Contrasts. Int. J. Pharm. 1999,
179, 1−19.
(159) Tadros, A. R.; Romanyuk, A.; Miller, I. C.; Santiago, A.; Noel,
R. K.; O’Farrell, L.; Kwong, G. A.; Prausnitz, M. R. STAR Particles for
Enhanced Topical Drug and Vaccine Delivery. Nat. Med. 2020, 26,
341−347.
(160) Lamson, N. G.; Berger, A.; Fein, K. C.; Whitehead, K. A.
Anionic Nanoparticles Enable the Oral Delivery of Proteins by
Enhancing Intestinal Permeability. Nat. Biomed. Eng. 2020, 4, 84−96.
(161) Wei, X.; Beltra
́
n-Gaste
́
lum, M.; Karshalev, E.; Esteban-
Ferna
́
ndez de Ávila, B.; Zhou, J.; Ran, D.; Angsantikul, P.; Fang, R.
H.; Wang, J.; Zhang, L. Biomimetic Micromotor Enables Active
Delivery of Antigens for Oral Vaccination. Nano Lett. 2019, 19,
1914−1921.
(162) Karshalev, E.; Esteban-Ferna
́
ndez de Ávila, B.; Beltra
́
n-
Gaste
́
lum, M.; Angsantikul, P.; Tang, S.; Mundaca-Uribe, R.; Zhang,
F.; Zhao, J.; Zhang, L.; Wang, J. Micromotor Pills as a Dynamic Oral
Delivery Platform. ACS Nano 2018, 12, 8397−8405.
(163) Aran, K.; Chooljian, M.; Paredes, J.; Rafi, M.; Lee, K.; Kim, A.
Y.; An, J.; Yau, J. F.; Chum, H.; Conboy, I.; Murthy, N.; Liepmann, D.
An Oral Microjet Vaccination System Elicits Antibody Production in
Rabbits. Sci. Transl. Med. 2017, 9, No. eaaf6413.
(164) Abramson, A.; Caffarel-Salvador, E.; Khang, M.; Dellal, D.;
Silverstein, D.; Gao, Y.; Frederiksen, M. R.; Vegge, A.; Huba
́
lek, F.;
Water, J. J.; Friderichsen, A. V.; Fels, J.; Kirk, R. K.; Cleveland, C.;
Collins, J.; Tamang, S.; Hayward, A.; Landh, T.; Buckley, S. T.;
Roxhed, N.; et al. An Ingestible Self-Orienting System for Oral
Delivery of Macromolecules. Science 2019, 363, 611−615.
(165) Shin, M. D.; Shukla, S.; Chung, Y. H.; Beiss, V.; Chan, S. K.;
Ortega-Rivera, O. A.; Wirth, D. M.; Chen, A.; Sack, M.; Pokorski, J.
K.; Steinmetz, N. F. COVID-19 Vaccine Development and a Potential
Nanomaterial Path Forward. Nat. Nanotechnol. 2020, 15, 646−655.
(166) Weiss, C.; Carriere, M.; Fusco, L.; Capua, I.; Regla-Nava, J. A.;
Pasquali, M.; Scott, J. A.; Vitale, F.; Unal, M. A.; Mattevi, C.;
Bedognetti, D.; Merkoç
i, A.; Tasciotti, E.; Yilmazer, A.; Gogotsi, Y.;
Stellacci, F.; Delogu, L. G. Toward Nanotechnology-Enabled
Approaches against the COVID-19 Pandemic. ACS Nano 2020, 14,
6383−6406.
(167) Fang, J.; Hsueh, Y.-Y.; Soto, J.; Sun, W.; Wang, J.; Gu, Z.;
Khademhosseini, A.; Li, S. Engineering Biomaterials with Micro/
Nanotechnologies for Cell Reprogramming. ACS Nano 2020, 14,
1296−1318.
(168) Sun, J.; Zhuang, Z.; Zheng, J.; Li, K.; Wong, R. L.-Y.; Liu, D.;
Huang, J.; He, J.; Zhu, A.; Zhao, J.; Li, X.; Xi, Y.; Chen, R.; Alshukairi,
A. N.; Chen, Z.; Zhang, Z.; Chen, C.; Huang, X.; Li, F.; Lai, X.; et al.
Generation of a Broadly Useful Model for COVID-19 Pathogenesis,
Vaccination, and Treatment. Cell 2020, 182, 734−743.
(169) Si, L.; Bai, H.; Rodas, M.; Cao, W.; Oh, C. Y.; Jiang, A.;
Nurani, A.; Zhu, D. Y.; Goyal, G.; Gilpin, S. E.; Prantil-Baun, R.;
Ingber, D. E., Human Organs-on-Chips as Tools for Repurposing
Approved Drugs as Potential Influenza and COVID19 Therapeutics
in Viral Pandemics. bioRxiv, April 14, 2020, ver. 1. https://www.
biorxiv.org/content/10.1101/2020.04.13.039917v1 (accessed 2020-
08-25).
(170) Sun, W.; Luo, Z.; Lee, J.; Kim, H.-J.; Lee, K.; Tebon, P.; Feng,
Y.; Dokmeci, M. R.; Sengupta, S.; Khademhosseini, A. Organ-on-a-
Chip for Cancer and Immune Organs Modeling. Adv. Healthcare
Mater. 2019, 8, 1801363.
(171) Segaliny, A. I.; Li, G.; Kong, L.; Ren, C.; Chen, X.; Wang, J.
K.; Baltimore, D.; Wu, G.; Zhao, W. Functional TCR T Cell
Screening Using Single-Cell Droplet Microfluidics. Lab Chip 2018,
18, 3733−3749.
(172) Monteil, V.; Kwon, H.; Prado, P.; Hagelkrüys, A.; Wimmer, R.
A.; Stahl, M.; Leopoldi, A.; Garreta, E.; Hurtado del Pozo, C.;
Prosper, F.; Romero, J. P.; Wirnsberger, G.; Zhang, H.; Slutsky, A. S.;
Conder, R.; Montserrat, N.; Mirazimi, A.; Penninger, J. M. Inhibition
of SARS-CoV-2 Infections in Engineered Human Tissues Using
Clinical-Grade Soluble Human ACE2. Cell 2020, 181, 905−913.
(173) Shah, S. K.; Miller, F. G.; Darton, T. C.; Duenas, D.; Emerson,
C.; Lynch, H. F.; Jamrozik, E.; Jecker, N. S.; Kamuya, D.; Kapulu, M.;
Kimmelman, J.; MacKay, D.; Memoli, M. J.; Murphy, S. C.; Palacios,
R.; Richie, T. L.; Roestenberg, M.; Saxena, A.; Saylor, K.; Selgelid, M.
J.; et al. Ethics of Controlled Human Infection to Address COVID-19.
Science 2020, 368, 832−834.
ACS Nano www.acsnano.org Review
https://dx.doi.org/10.1021/acsnano.0c06109
ACS Nano 2020, 14, 12370−12389
12389
No comments:
Post a Comment
Note: only a member of this blog may post a comment.