You are on page 1of 11

Expert Review of Vaccines

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ierv20

Dendritic cell vaccine as a potential strategy to


end the COVID-19 pandemic. Why should it be Ex
Vivo?

Jonny Jonny, Terawan Agus Putranto, Enda Cindylosa Sitepu & Raoulian Irfon

To cite this article: Jonny Jonny, Terawan Agus Putranto, Enda Cindylosa Sitepu & Raoulian Irfon
(2022): Dendritic cell vaccine as a potential strategy to end the COVID-19 pandemic. Why should it
be Ex�Vivo?, Expert Review of Vaccines, DOI: 10.1080/14760584.2022.2080658

To link to this article: https://doi.org/10.1080/14760584.2022.2080658

© 2022 The Author(s). Published by Informa


UK Limited, trading as Taylor & Francis
Group.

Published online: 26 May 2022.

Submit your article to this journal

Article views: 5846

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ierv20
EXPERT REVIEW OF VACCINES
https://doi.org/10.1080/14760584.2022.2080658

REVIEW

Dendritic cell vaccine as a potential strategy to end the COVID-19 pandemic. Why
should it be Ex Vivo?
Jonny Jonny , Terawan Agus Putranto, Enda Cindylosa Sitepu and Raoulian Irfon
Cellcure Center, Gatot Soebroto Central Army Hospital, Jakarta, Indonesia

ABSTRACT ARTICLE HISTORY


Introduction: Developing a safe and efficacious vaccine that can induce broad and long-term immunity Received 29 March 2022
for SARS-CoV-2 infection is the most critical research to date. As the most potent APCs, dendritic cells Accepted 18 May 2022
(DCs) can induce a robust T cell immunity. In addition, DCs also play an essential role in COVID-19
KEYWORDS
pathogenesis, making them a potential vaccination target. However, the DCs-based vaccine with ex vivo
Cell immunotherapy; COVID-
loading has not yet been explored for COVID-19. 19; dendritic cell; SARS-CoV
Areas covered: This review aims to provide the rationale for developing a DCs-based vaccine with ex -2; T cells; vaccination
vivo loading of SARS-CoV-2 antigen. Here, we discuss the role of DCs in immunity and the effect of
SARS-CoV-2 infection on DCs. Then, we propose the mechanism of the DCs-based vaccine in inducing
immunity and highlight the benefits of ex vivo loading of antigen.
Expert opinion: We make the case that an ex vivo loaded DC-based vaccination is appropriate for
COVID-19 prevention.

1. Introduction developed for influenza vaccination in high-risk patients who


respond poorly to traditional vaccines, such as immunocom­
COVID-19 (coronavirus infection disease-2019) is a mild to
promised and cancer patients [15]. In light of this, this method
severe respiratory disease caused by SARS-CoV-2 (Severe
might be used for other infections such as SARS-CoV-2.
Acute Respiratory Syndrome Coronavirus-2) infection [1]. This
However, its potential as a COVID-19 vaccine has not been
disease is a primary health concern worldwide due to the
explored.
pandemic. Although several vaccines have already been
This review aims to provide the rationale for developing
approved for emergency use, herd immunity generally relies
a DC-based vaccine with ex vivo loading of SARS-CoV-2 anti­
on vaccines [2]. However, recent findings suggest that Variant
gen. First, we discuss the role of dendritic cells in immunity
of Concerns (VoC) might escape vaccine-induced immunity
against infection. Then, we review the effects of SARS-CoV-2
hindering its efficacy [3]. In addition, immunity waning raises
infection on dendritic cells. From here, we propose the
the concern for the long-term efficacy of currently available
mechanism of the DC-based vaccine with ex vivo loading of
vaccines [4,5]. Thus, discovering safe and efficacious vaccines
antigen in inducing immunity. Finally, we also highlight the
that lead to broader and long-term immunity remains
benefits of ex vivo loading of antigen.
challenging.
The DCs-based vaccine is one of the promising vaccine
candidates due to the vital role DCs play in the immunity 2. Dendritic cells in immunity
and pathogenesis of COVID-19 [6]. Ex vivo loading of antigens
2.1. Dendritic cell in innate immunity
to DCs is mainly developed for cancer therapy and chronic
viral infection. Dendritic cell-based vaccine is a novel vaccina­ The human body has a defense system that works during
tion approach. Studies of dendritic cell vaccines in cancer pathogen attacks. There are two types of immune response:
patients that have been conducted to date show promising innate and adaptive immune responses [16]. The innate
results [7–12]. For example, adding the autologous tumor immune response is the first line of defense that is activated
lysate-pulsed dendritic cell vaccine to standard therapy for instantly within minutes to hours after the pathogen invasion.
glioblastoma patients may prolong survival [9]. Other studies However, innate immune responses are generally nonspecific
conducted on Wilms tumor patients using autologous dendri­ and unadaptable. Meanwhile, the adaptive immune response
tic cells incubated with WT1 peptides showed induction of can recognize antigens specifically through diversified somatic
systemic tumor antigen-specific immune response, increased receptors on B cells and T cells. So that, although requiring
infiltration of CD8+ tumor T cells, and increased tumor PD-L1 a longer time to be acquired, the adaptive immune response
expression [8]. This method was also developed for HIV-1 and will cause a much faster and more effective response upon
Hepatitis B [13,14]. Furthermore, DCs-based vaccines were also antigen re-encounter [17].

CONTACT Jonny Jonny jonny_army@yahoo.com Cellcure Center, Gatot Soebroto Central Army Hospital, Jakarta, Indonesia
© 2022 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/4.0/),
which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in any way.
2 JONNY ET AL.

important for cells communication. For example, NK cells


Article highlights produce IFN-γ that modulates DCs function [25]. IFN-γ triggers
● DCs are essential in the innate immune response through the pro­
DC maturation, increases MHC I and II expression, triggers
duction of IFN-1 and immune cells modulation. antigen presentation, and boosts Interleukin 2 (IL-2) and
● DCs are the most important APCs that activate various T cells Cluster of Differentiation 80 (CD80) expression, which are
immune responses.
● SARS-CoV-2 can directly infect DCs causing inadequate innate and
vital in signaling T cell and B cell activation [26,27].
adaptive immune response. Furthermore, communication between NK and DC is bidirec­
● COVID-19 vaccination targeting DCs might result in better outcomes tional, leading to the amplification of pro-inflammatory cyto­
and potentially causes long-term and broader spectrum immunity.
● Ex vivo loading of antigens is an appropriate approach in manufac­
kines [28].
turing DCs-based vaccines for COVID-19. Because this approach M1 (inflammatory types) macrophages produce a variety of
bypasses the rigorous process of antigen presentation inside the inflammatory cytokines, including TNF-α [29]. TNF-α produced
body, results in high quality and quantity antigen-loaded mature
DCs and has a well-established safety profile. by these macrophages mediates DCs maturation and NK cells
activation [30]. In addition, TNF-α plays a role in immunoregu­
lation by limiting the expression of CXR2 receptors and the
TLR7/9-induced gene in pDC [31]. In sepsis, TNF-α decreases
The innate immune response consists of various modalities: autophagy function and causes DC dysfunction [32]. Taken
physical, chemical, and cellular defenses. Physical defense together, similar to NK cells, macrophages also modulate
protects the body from pathogens using epithelial tissue and DCs function via TNF-α. Crosstalks between these cells create
mucous membrane [18]. When a pathogen successfully pene­ a balanced cytokine milieu in the innate immune response.
trates this defense, the cellular component of innate immunity
reacts by destroying the pathogen through phagocytosis. The
primary phagocytic cells are derived from myeloid progenitor
2.2. Dendritic cell in adaptive immunity
cells, such as monocyte-macrophage cells, neutrophils, baso­
phils, and eosinophils. Meanwhile, Natural Killer (NK) cells are When the innate immune response cannot destroy antigens,
nonspecific cytotoxic, which destroy cells infected with viruses the body activates the adaptive immune response [33]
or intracellular pathogens [19]. Finally, Dendritic Cells (DCs) are (Figure 1). Adaptive immunity is antigen-dependent and spe­
also an essential cellular component in the innate immune cific. It can induce immunological memory that causes a faster
response, which have phagocytic abilities, produce inflamma­ reaction to antigen re-exposure. It is important to note that
tory mediators, and present antigen [20] (Figure 1). recent studies have found that immunological memory can
The effectiveness of the innate immune response in fight­ also occur in innate immunity to a certain extent [34].
ing viral infections depends on the production of IFN-1. IFN-1 However, it remains a fundamental feature of adaptive
suppresses the spread of the virus through the intrinsic anti­ immunity.
microbial activity of infected cells, promotes antigen presenta­ Adaptive immunity consists of T-cell mediated immunity,
tion, and induces NK cells activation [21]. Most IFN-1 is and B-cell mediated immunity [35]. B cells and T cells only
produced by plasmacytoid dendritic cells (pDC) [22]. pDC eliminate antigens recognized by their receptors [36]. So, the
recognizes the virus through Toll-Like Receptors (TLR) 7 and key to adaptive immunity is antigen recognition and presenta­
9 [23]. Stimulation of these receptors triggers massive IFN-1 tion by Antigen Presenting Cells (APCs). APCs stimulate the
release within 1–3 hours after viral infection, especially the expression of specific receptors on T cells and B cells.
IFN-α and IFN-β subtypes [24]. Compared to other APCs, DCs are more potent in inducing
In addition to IFN-1, cellular components of innate immu­ T cells [37,38]. Therefore, DCs are the most essential APCs that
nity express other types of inflammatory mediators that are bridge innate and adaptive immune responses.

Figure 1. DCs role in innate and adaptive immune response. Pathogens that pass through the physical barrier (epithelium and mucosa) activate the innate immune
response and adaptive immune response. First, monocytes-macrophages, neutrophils, and DC capture pathogens, then express antigens on cell membranes. next,
DCs as the primary APC migrate to the lymph nodes, where antigens are introduced to T and B cells, thus triggering memory development.
EXPERT REVIEW OF VACCINES 3

There are five types of DC subsets, namely Plasmacytoid involving the Endoplasmic Reticulum (ER) and the vacuolar
DCs (pDCs), Conventional DCs 1 (cDC1), Conventional DCs 2 pathway in the endocytic compartment [48,49]. Through the
(cDC2), Inflammatory DCs, and Langerhans cells [37]. pDC, cytophilic pathway, pathogenic proteins are translocated to
cDC1, and cDC2 are present constantly in the body [39]. ER. Inside ER, MHC-I binds those proteins to be translocated
While Inflammatory DCs are derived from monocyte precur­ back (retrotranslocation) to the cytosol [49]. Antigen presenta­
sors that differentiate into DCs in the event of infection, tissue tion by DCs also involves extracellular vesicles, thus enabling
damage, or the production of cytokines and chemokines [37]. cross-dressing (peptide-loaded MHC retrieval from dead cells
Inflammatory DCs are also called Monocyte Derived Dendritic or cells that have undergone apoptosis) [50].
Cells (MoDCs). Based on the path to access lymphoid organs, DCs with peptide-loaded MHC (pMHC) migrate to second­
the cDCs consist of two types [40]. First, Resident DCs, derived ary lymphatic organs to activate T cells. The migration of DCs
from bone marrow precursors within lymphoid organs, do not to the secondary lymphatic organs is regulated by chemokines
pass through peripheral tissues. Second, Migratory DC formed C-C Chemokine Ligand (CCL) 19 and 21 secreted by lymphatic
from precursors in peripheral tissues, after exposure to anti­ endothelial cells. It also relies on C-C Chemokine Receptor 7
gens, will transfer to lymphoid organs to further interact with (CCR7) for DCs positioning and initiation of specific immune
T cells to induce adaptive immunity. responses [51,52]. In secondary lymphoid organs, DCs actively
Although it has different classifications, phenotypes, and spread antigens to other DCs through cross-dressing, conse­
role specificities, the primary role of DCs is antigen presenta­ quently increasing the number of effective APCs [53]. So that,
tion. Even pDC, whose primary function is to produce IFN-1, T cells priming can still occur even if the donor DCs cannot
can be diversified into pDC with phenotypes that are able to directly present the antigen.
stimulate T cells [41]. The DCs will capture pathogenic parti­ T cell priming occurs in secondary lymphoid organs. This
cles during an infection, then undergo maturation and process requires complicated T cell and DC signaling to ensure
migrate to the local lymph nodes to activate T cells and contact. After migration to secondary lymphoid organs, DCs
B cells [42]. DCs also serves as surveillance cells that continu­ tend to settle, while T cells can scan through hundreds of DCs
ously take up antigen samples from the environment. by moving from one lymph node to another. Although DCs
Exogenous antigen uptake is done through endocytosis, pha­ are estimated to interact with 5000 T cells per hour, initiation
gocytosis, micropinocytosis, and receptor-mediated endocyto­ of T cell response remains highly dependent on the possibility
sis [43]. In addition, TLRs on DCs can recognize parts of of T cells encountered by DCs [52,54]. A study in mice found
pathogens. For example, TLR3 can recognize the double- that it takes at least 85 DCs to initiate T cell priming, so in
stranded ribonucleic acid (dsRNA), TLR7/8 can recognize sin­ order to induce a response, this threshold should be met [55].
gle-stranded ribonucleic acid (ssRNA), and TLR9 can recognize In addition to that, T cells priming requires three signals: 1)
5’ – C – phosphate – G – 3’ deoxyribonucleic acid (CpG DNA). Interaction of T Cell Receptors (TCR) with pMHC; 2) Co-
The interaction between pathogen-associated molecular pat­ stimulatory signals through the attachment of CD28 with
terns (PAMPS) or danger-associated molecular patterns CD80 and CD86; 3) Cytokines expressed by DCs such as IL-12
(DAMPS) and TLR causes activation of various transcription [52]. Damage to any of these signals will cause failure to
factors resulting in DCs maturation [44]. T priming.
At the time of maturation, DCs undergo morphological Antigens presented by MHC-I triggers CD8 + T cell
and phenotype changes. Immature DCs have a rounded response, while antigens presented by MHC-II triggers
shape and high phagocytosis ability but express fewer co- CD4 + T cell response. CD8 + T cells primarily kill host cells
stimulatory molecules. After interaction with pathogens, infected with viruses. Therefore, most CD8+ will proliferate
DCs transform their morphology to cells with longer den­ during acute viral infections after exposure to antigens pre­
drites and pseudopodia and express more co-stimulatory sented by DCs to assist viral clearance [56]. Meanwhile,
molecules such as CD80 and CD86. In addition, mature CD4 + T cells differentiate into various sub-types depending
DCs have weaker phagocytosis ability but much higher on the cytokines (e.g. Th1, Th2, Tfh, and Th17) [57]. Each sub-
migratory ability than immature DCs [45]. types has a specific function in mediating humoral and cellular
In the acute phase of infection, especially infections adaptive immunity. Moreover, after interaction with DCs, some
caused by intracellular viruses or bacteria, some monocyte T cells differentiate into T memory cells [52]. DCs are also
cells turn into MoDCs. IFN-γ primarily mediates the forma­ essential in germinal center (GC) development by facilitating
tion of MoDCs [46]. Moreover, MoDCs isolated from periph­ T cell follicular helper (Tfh) differentiation and directly migrat­
eral blood can differentiate into MoDC outside the body. ing to the light zone of GC [58].
This manipulation is done by incubating peripheral blood DCs have a vital role in the immune system, as previously
mononuclear cells (PBMCs) in the differentiation media such described. The role of DCs begins from innate immunity
as GM-CSF and IL-4 [47]. This method is the most widely through phagocytic abilities, inflammatory mediators produc­
used DCs-based vaccine manufacturing. tion, and antigen presentation. DCs are professional APCs that
As the primary APC, DCs have unique capabilities com­ connect innate and adaptive immune responses. In the adap­
pared to others. DCs can present exogenous antigens through tive immune response, DCs have several advantages in carry­
both MHC-I and MHC-II (cross-presentation). Cross- ing out their function as APCs: 1) DCs are much more potential
presentation can occur through the cytophilic pathway than other APCs; 2) Enable various antigen uptake pathways
4 JONNY ET AL.

and can process both endogenic and exogenic pathogens; 3) dysfunctional monocytes and neutrophils are associated with
The migration of DCs to secondary lymphoid organs and the severity of the disease [69]. In studies comparing acute
recruitment of T cells is a very efficient process so that the and convalescent-phase COVID-19 patients, low DCs and
probability for the occurrence of T cell priming is very high; 4) monocyte count values were obtained in the convalescent
Have cross-presentation capabilities to trigger a wide array of phase. Although, the number of T and NK lymphocyte cell
T cells response; 5) Lastly, Has cross-dressing capability to counts was increased. This finding indicates the possibility of
allow antigen transfer from other cells and active dissemina­ suppressing the production of monocytes, DC, NK, and T cells,
tion of antigens within secondary lymphatic organs. but NK and T cell suppression lasts shorter [70]. As explained
in the previous section, the interaction between NK cells,
macrophages, and DCs is vital in creating a cytokine induction
3. SARS-CoV-2 infection dysregulate dendritic cell
balance in the innate immune response. Suppression of these
Viruses that enter the body will try to avoid the immune cells creates an imbalance in immunoregulation. Disruptions
response. Some viruses develop ways to escape immune in the axiom of communication between these cells may cause
response by interfering with DCs functions. For example, it clinical worsening.
has been suggested that DCs dysregulation plays a vital role in There is an IFN-1 response abnormality in COVID-19
SARS-CoV-2 immune evasion [6]. DCs dysregulation causes patients, as evidenced by the low value of the IFN-1 and IFN-
various abnormalities throughout the innate and adaptive 3 [71]. This finding is likely related to pDCs, the primary source
immune response. In this section, we will highlight some of of IFN-1. A decrease of pDCs is found in patients infected with
the important findings. SARS-CoV-2 [70]. A timely IFN-1 response is necessary to acti­
vate an effective innate immunity. However, in coronaviruses
infection, there is a delay in innate immunity for the first ten
3.1. SARS-CoV-2 causes inadequate innate immune
days in which during this period a steady increase of viral load
response
occurs [72,73]. Ultimately, delayed IFN-1 response also leads to
SARS-CoV-2 is known to infect cells through the ACE2 receptor the accumulation of pathogenic macrophages that cause pul­
and relies on TMPRSS2 [59]. Several other receptors can be the monary infiltration, vascular leakage, and suboptimal T-cell
entry point of this virus, including CD147, L-SIGN, and DC-SIGN response [74].
[60–62]. DCSIGN receptors are widely expressed in DCs and Delayed IFN-1 response might be due to SARS-CoV-2
macrophages, while L-SIGN is mainly expressed in liver sinu­ infected pDC. Protein in SARS-CoV-2 such as ORF9b, Nsp14,
soid endothelial cells and lymphatic nodes [62]. The presence Nsp10, M, and Nsp1 may inhibit IFN-1 production. At the same
of such receptors suggests that this virus can directly infect time, other proteins such as ORF3a and ORF6 can inhibit the
DCs. Research on SARS-CoV infected MoDCs proves that these IFN-1 signaling pathway [72]. Furthermore, congenital
cells can be infected, but the infection does not cause cell abnormalities (inborn errors) of the IFN-1 related genes,
death [63]. In line with these findings, in vitro research proves namely inborn error in TLR-3 and IFN regulatory factor 7
that infected MoDC does not support viral replication [64]. (IRF-7), are found in critical patients [75]. In line with these
People infected by SARS-CoV-2 experience an increase in findings, pDC as the primary source IFN-1 expresses much IRF-
blood serums, pro-inflammatory cytokines, and chemokines 7 [24,76]. Furthermore, the presence of autoantibodies against
such as TNF, IL-6, IL-8, Macrophage Inflammatory Protein-1-α IFN-1 is also found in life-threatening COVID-19 [77]. So that,
(MIP-1), Monocyte Chemoattractant Protein-1 (MCP-1), and people with the IFN-1 genetic disorder become more suscep­
Interferon-inducible Protein-10 (IP-10) [65]. There is a strong tible to clinical worsening [75].
correlation between IP-10 and symptom severity [66]. In addi­
tion, critical patients have an increased inflammatory response
3.2. SARS-CoV-2 causes adaptive immunity disruption
and dysfunctional response related to IFN [67]. Infected DCs
are thought to be the source of these excessive pro- The adaptive immune response to SARS-CoV-2 relies heavily
inflammatory cytokines. Interestingly, a study shows that on specific T and B lymphocytes. In COVID-19 patients, there is
macrophages (not DCs) are the source of pro-inflammatory a tendency for lymphopenia, where the number of CD4 + and
cytokines when infected with SARS-CoV-2 [64,68]. However, CD8 + T cells were low [78]. Decreased number and fatigue of
infected MoDCs express IP-10, resulting in decreased IFN pro­ T cells are related to high mortality and severe COVID-19 [79–
duction and IFN associated genes [64]. Conflicting results on 81]. Low CD4 + T cell counts were also associated with the
this matter still require further exploration, particularly the longer length of ICU treatment [82]. One reason that might
immune cells that contribute most to cytokine storms. There explain low T cell count is the direct inhibition from cytokines.
are complex interactions between immune cells that have yet For example, increased serum IL-17 is found in critical patients
been explained. Since specific signaling between immune cells [67]. IL-17, which Th17 produces, is responsible for T cell
orchestrates innate immunity, it is crucial to identify the suppression. In addition, direct T cell inhibition from cytokines
checkpoint that causes this dysfunction. Therefore, there is such as IL-6, IL-10, or TNF may also contribute [78].
still a possibility that MoDC is related to the increase of pro- However, another elegant explanation is that the decreased
inflammatory cytokines. number and lower DCs maturation causes the lack of optimal
DCs communicates with NK cells and macrophages in the priming of T cells, resulting in delays of T cells movement.
induced innate immunity [26,27,30–32]. The myeloid compart­ Studies on 32 COVID-19 patients with severe symptoms trea­
ment of COVID-19 patients is abnormal. Immature and ted at Heidelberg University Hospital showed low CD8+ and
EXPERT REVIEW OF VACCINES 5

myeloid DCs counts of patients’ blood cultures [67]. The study approach that focuses on protecting and improving DCs can
results to look at the immune system profiles of COVID-19 result in faster virus clearance and a better prognosis.
patients in Prague with peripheral and culture blood isolation
methods showed decreased expression of HLA-DR and DCs
4. Dendritic cell-based vaccine against SARS-CoV-2
maturation markers such as CD80 and CD86 in all subsets of
DCs [83]. As explained in the previous section, T cell priming There are several reasons to develop DCs-based vaccines for
would not have occurred without DCs’ co-stimulatory the prevention of COVID-19. First, SARS-CoV-2 causes DCs
molecules. dysfunction, making DCs an appropriate vaccination target
Lastly, moDC infected with SARS-CoV-2 may cause T cell (Figure 3). In addition, stimulation of DCs results in a robust
death. Research shows increased Tumor Necrosis Factor- T cell response [46]. The response of T cells to SARS-CoV-2
related Apoptosis-Inducing Ligand (TRAIL) activity in MoDCs lasted longer than the humoral response, which is up to
of SARS-CoV patients where TRAIL expression is associated 10 months after infection [89]. Interestingly, research on SARS-
with the amount of SARS-CoV virus [84]. Single-Cell CoV showed that memory T cell responses to the virus could
Sequencing of blood mononuclear cells infected with SARS- survive up to 17 years after infection [90]. DCs-based vaccine
CoV-2 showed a similar thing: increased expression of TRAIL might elicit T cell response, which is protective against infec­
and its receptors on T cells [85]. TRAIL is a molecule that plays tion, this includes formation of memory T cells [46]. Unlike
a role in inducing cellular apoptosis pathways. The bond neutralizing antibody, which is expected to diminish over
between TRAIL and T cells leads to the Death-Inducing time, several findings suggest that T cell responses can be
Signaling Complex (DISC) recruitment, resulting in cell death retained for a long period, even years [89,90]. In this case,
[86]. Thus, infected DCs can directly affect T cell lymphopenia booster doses might only need to be given after a long
due to apoptosis activity through TRAIL. time, and might not even be necessary. Hence, DCs-based
Abnormalities in the adaptive immune response can also vaccine can be a way to acquire long-term immunity.
be caused through other pathways. For example, the SARS- Second, the DCs-based vaccine also might induce broader
CoV-2 virus can activate the nuclear factor kappa-light-chain spectrum immunity. Evidence shows that memory T cell
enhancer of activated B cells (NFkB). Furthermore, NFkB responses remain effective against VoC [91]. DCs-based vac­
induces tolerogenic phenotypes, which are immunosuppres­ cine can elicit such a response. In addition, DCs can trigger the
sive, through the production of IL-10 [87]. Therefore, in a state formation of germinal center (GC) response to form B cells
of viral infection, suppression of NFkB activity is needed to that can recognize variants [92]. DCs induces GC response in
increase the activity of the immune response mediated by IFN two ways: converting naïve T cells into follicular helper T cells
[88]. In contrast, there was an increase in the expression of the and migration of follicular DCs [58]. Thus, DCs-based vaccina­
Programmed Death-Ligand 1 (PD-L1) program, which plays tion is particularly suitable for fighting viruses with high muta­
a role in inhibiting the adaptive immune response [83]. tion rates such as SARS-CoV-2.
Failure to change from innate to the adaptive immune
response in SARS-CoV-2 infection led to worsening cases of
COVID-19 infection. Disruption of DCs’ ability to accommodate
5. Ex vivo approach: challenges and opportunities
such changes is an integral part of the pathogenesis of COVID- Immunization targeting DCs can be done via ex vivo loading
19 (Figure 2). In addition, DCs has an extensive role in the and in vivo targeting. In the ex vivo loading approach, antigens
immune response. Therefore, the COVID-19 vaccination are exposed outside the body to DCs isolated from peripheral

Figure 2. Immune Response to SARS-COV-2 Infection. SARS-CoV-2 can infect DC mainly via DC-SIGN receptors. Infected DC become dysfunctional, causing
inadequate IFN-1 response, decreased DC maturation, upregulation of pro-inflammatory cytokines, inhibition of adaptive immunity via PD-L1 and NFkB, and
T cell apoptosis via TRAIL. These cause the failure of innate and adaptive immunity.
6 JONNY ET AL.

Figure 3. DC-based vaccine with ex vivo loading of antigens. Antigen loaded mature DCs are injected back to body, then migrate to secondary lymphoid organ to
induce T cells. DCs are not infected by SARS-CoV-2 so that there’s no immunological dysfunction.

blood monocyte cells and then injected back into the body Some studies using ex vivo generated DCs for therapy of
[93]. This vaccine is specific and individual. Despite still being infections are summarized in Table 1. This approach is mostly
experimental, this approach is also currently being developed developed for chronic infection immunotherapy such as HIV-1,
for SARS-CoV-2 vaccination [94]. In contrast, in vivo targeting, Hepatitis B, Hepatitis C and HSV [13,14,97–99]. Ex vivo gener­
antigen delivery is done by conjugating the desired antigen ated DCs-based vaccine for prevention of viral infection is
with antibodies specific to DCs receptors (e.g. anti-CD40 or deemed unjustifiable due to the high cost of production.
C-type lectin receptor) [95]. Both of these methods have their This hinders the progress to understand the full scope of ex
respective advantages and disadvantages. However, the ex vivo generated DCs in inducing immunity. However, in the
vivo loading method is still the primary choice in developing case of COVID-19, the justification to develop this approach
dendritic cell-based vaccines. Several dendritic cell vaccine still require a comprehensive cost-benefit analysis. The efficacy
studies that entered the clinical trial employ the ex vivo load­ of currently available COVID-19 vaccines are decreasing over­
ing method [7–12,96]. time, and the emergence of variants are pushing for the

Table 1. Studies of DCs-based vaccines for chronic infections.


Study
(year) Population (N*, Duration) Treatment Result Ref**
Chen et al HBV-infected patients (12 Autologous PMBC Derived DCs Pulsed Suppression of HBV replication, Viral load reduction, HBeAg [14]
(2005) subjects, 1 year) With HbsAg elimination and promotion of HBeAg/anti- HBe transformation
Wang et al Hepatitis B-positive Autologous PMBC DCs No increase in [97]
(2015) Patients with Pulsed With Autologous Irradiated hepatic transaminases, bilirubin, prothrombin time, hepatitis B
Hepatocellular Carcinoma Tumor Stem Cells antigens, or viral DNA
(15 subjects, 8 weeks)
Mekonnen C57BL6 mice (7 subjects, Necrotized DC2.4 cell line expressing Increased the breadth of T-cell responses and enhanced the [108]
et al 2 weeks) HCV NS3 production of IL-2, TNF-α, and IFN-γ by effector memory CD4
(2020) + and CD8 + T cells, induced a greater influx and activation of
cross-presenting CD11c+ CD8α+ DC and necrosis-sensing Clec9A
+ DC in the draining lymph nodes, enhanced clearance of NS3-
positive hepatocytes from the livers
Lu et al Untreated HIV-1-infected Autologous PMBC derived DCs loaded Decreased plasma viral load, prolonged suppression of viral load for [13]
(2004) patients (18 subjects, with autologous aldrithiol- at least 1 year correlated with HIV-1-specifc interleukin-2 or
1 year) 2-inactivated HIV-1 interferon-γ-expressing CD4 + T cells and with HIV-1 gag–specific
perforin-expressing CD8+ effector cells
Surenaud Antiretroviral treated HIV- Autologous PMBC derived IFNα DCs Induced and/or expanded HIV-specific CD8 + T cells producing IFNγ, [98]
et al 1-infected patients (16 loaded with HIV-1 lipopolipeptide perforin, granzyme A and granzyme B, and also CD4 + T cells
(2019) subjects, 24 weeks) secreting IFNγ, IL-2 and IL- 13
Leplina Recurrent HSV-1 infected Autologous PMBC derived DCs loaded Reduction in the recurrence rate and significant enhancement of the [99]
(2016) patients (14 subjects, with HSV-1 recombinant viral inter-recurrent time, induction of an antigen-specific response to
9 months) glycoprotein D (HSV1gD) HCV1gD, reduced mitogenic responsiveness of mono- nuclear
cells
*N = Total number of subjects in the study, **Ref = Reference
EXPERT REVIEW OF VACCINES 7

discovery of next generation vaccines [4,5]. The potential in the S-protein of SARS-CoV-2 for 2 days. This process is carefully
inducing long-term and broad immunity by DCs-based vac­ controlled, and the quality of the DCs can be checked prior to
cine might eliminate the need for frequent booster vaccines, administration. High-quality mature DCs can migrate efficiently
thus in the long run the total cost of production and distribu­ to the secondary lymphoid organs, where they initiate the
tion of ex vivo generated DCs vaccine can be comparable to innate and adaptive immune response to SARS-CoV-2. In addi­
conventional vaccines. Another aspect that should be consid­ tion, there is evidence that DCs actively spread antigens to
ered is the feasibility of wide distribution due to the fact that other APCs through cross-dressing [53]. This ability increases
this vaccine production requires a highly trained staff in spe­ the adequate number of APCs and keeps T cell priming from
cific facilities. However, vaccines made point-of-care such as occurring even though the donor DCs cannot directly prime
hospitals and medical laboratories can be the solution to this T cells. Whereas in vivo DCs-based vaccination, the quality of
problem [100]. In conclusion, cost and practical limitation of DCs is affected by the difficulty of controlling each individual’s
this approach should not limit the potential benefit (Table 2). state of immunity. For example, there is a decrease in the
Furthermore, in the context of the SARS-CoV-2 vaccine, the quality of DCs in people with chronic diseases and the elderly
world is in a race against time [101]. Therefore, the ex vivo [103–105]. Furthermore, a DCs-related genetic disorder can
presentation of antigens is currently an appropriate method. impair DCs’ function [106]. Therefore, in vivo DCs targeting
Meanwhile, in vivo DCs targeting still needs to be refined. This vaccination in that population might be ineffective.
method also has the following advantages: Fourth, the body can tolerate autologous DCs vaccine with
First, the presentation of antigens outside the body will ex vivo antigen presentation. This method is proved safe from
bypass DCs antigen presentation process that is key in initiat­ previous studies and caused only negligible toxicity [7–12].
ing the immune response. The process of antigen presentation Side effects are limited to mild local and systemic reactions
in the body is highly complex. Not all antigens that enter the such as injection pain, joint pain, and flu-like syndrome. There
body are successfully taken up and presented through DCs. are no deaths or life-threatening events related to the auto­
Antigens must meet with DCs directly for the process to occur. logous DCs vaccine. When an antigen enters the body, the
This process is influenced by several factors: availability of immune system activates the signaling pathways distinguish­
dendritic cells, the presence of PRR (Pathogen Recognition ing self and non-self [107]. However, autologous DCs are
Receptors), a condition that supports DCs maturation and recognized as a part of the self so that the body can tolerate
differentiation, and activation of antigen processing pathways the material well without a reaction of foreign body rejection.
through the corresponding compartment [23,43,49].
Deficiency in one of these factors will make the failure of
6. Conclusion
antigen presentation by DCs. Ex vivo antigen presentation
will cut through such a complicated process. We have outlined that DCs are the most potent APCs that can
Second, ex vivo antigen presentation results in a sufficient activate a wide array of T-cell immunity. DCs play a crucial role
number of mature DCs for T cells priming. A study shows that in innate and adaptive immunity. SARS-CoV-2 can infect DCs
it takes at least 85 mature DCs to induce T cells response [55]. resulting in disruption of innate and adaptive immunity.
Moreover, large amounts of antigens are required because the Dendritic cell vaccine using ex vivo approach can result in
pMHC only lasts for a short time [40]. Appropriate cytokines better clinical outcome.
are also needed as maturation signals [43]. When done outside
the body, antigen presentation occurs in a controlled environ­
7. Expert opinion
ment, where the number of DCs, maturation signals, and
antigens are known. In addition, by isolating DCs ex vivo, the DCs can activate various T-cells responses and are essential in
antigen is presented only to the DCs and not other cells. Thus, T-cells fate determination [38]. In innate immunity, pDCs are
this method ensures the development of mature DCs in suffi­ the primary source of IFN-1. Crosstalk between DCs, NK cells,
cient quantities for T cells priming. and macrophages create the cytokine milieu suitable for
Third, the ex vivo antigen presentation method produces innate immune responses. Furthermore, DCs are responsible
high-quality DCs. DCs can be derived from peripheral blood for antigen presentation, which is key to adaptive immune
mononuclear cells when stimulated by IL-4 and GM-CSF response. T-cells priming by DC is a highly efficient process
[47,102]. In our study, DCs were obtained by incubation of orchestrated by various cytokines and chemokines for optimal
peripheral blood mononuclear cells for 5 days within differen­ cell positioning. On the other hand, SARS-CoV-2 can infect DCs
tiation media [100]. Those DCs are subsequently incubated with and causes dysfunction in innate and adaptive immunity [6].
As postulated in this review, this virus can directly infect DCs
resulting in abnormal cytokines production, T-cell depletion
Table 2. Advantages and disadvantages of Ex vivo loading. and fatigue, and adaptive immunity inhibition. Therefore, the
Advantages Disadvantages vaccination approach by targeting and protecting DCs might
(1) Bypasses complicated antigen pre­ (1) Relatively high cost (depends on result in faster viral clearance and better clinical outcomes.
sentation process. the number of production).
(2) Results in a sufficient number of (2) Require specific facilities for vac­ Here, we have outlined the rationale to develop DCs-based
mature DCs for T cells priming. cine production. vaccine for prevention of COVID-19. Although mainly developed
(3) Produces high-quality DCs. for treatment of cancer and chronic infections, this method can
(4) Safe and causes fewer adverse
events. also be extended for prevention of SARS-CoV-2 infection [7–
12,96]. Some previous studies show that DCs based vaccine
8 JONNY ET AL.

can induce immunity against viral infection through activation References


of T-cells immunity [13,14,98,99,108]. Interestingly, even when
1. Zhu N, Zhang D, Wang W, et al. A novel coronavirus from patients
the immune system is compromised such as in HIV infection, with Pneumonia in China, 2019. N Engl J Med. 2020;382:727–733.
DCs vaccines are still capable to induce T-cell response resulting 2. Randolph HE, Barreiro LB. Herd immunity: understanding
in reduction of viral load [13,98]. So that, it is possible that DCs COVID-19. Immunity. 2020;52:737–741.
vaccine will remain effective for people with weaker immunity, 3. Lopez Bernal J, Andrews N, Gower C, et al. Effectiveness of Covid-19
such as children, elderly, and people with chronic illnesses. vaccines against the B.1.617.2 (Delta) variant. N Engl J Med.
2021;385:585–594.
DCs are responsible for a wide array of T cell responses, 4. Sabino EC, Buss LF, Carvalho MPS, et al. Resurgence of COVID-19 in
thus utilizing DCs as vaccine carrier might induce a broad and Manaus, Brazil, despite high seroprevalence. Lancet.
long-term immunity for SARS-CoV-2 prevention [46,89,90]. 2021;397:452–455.
There is also a possibility of induction of GC response by 5. Wang P, Nair MS, Liu L, et al. Antibody resistance of SARS-CoV-2
DCs. Even though the mechanism is still unclear, GCs response variants B.1.351 and B.1.1.7. Nature. 2021;593:130–135.
6. Borges RC, Hohmann MS, Borghi SM. Dendritic cells in COVID-19
can induce production of antibody with broader neutralization immunopathogenesis: insights for a possible role in determining
capacity through clonal evolution [92]. So that, alongside disease outcome. Int Rev Immunol. 2021;40:108–125. 1–2
clinical efficacy, T-cell and GC response elicited by DCs should 7. Jiang N, Qiao G, Wang X, et al. Dendritic Cell/Cytokine-Induced
be taken into consideration in future studies. killer cell immunotherapy combined with S-1 in patients with
Currently, ex vivo loading is the most feasible method for advanced pancreatic cancer: a prospective study. Clin Cancer Res.
2017;23:5066–5073.
DC-based vaccine development. Most of the studies that 8. Zhang W, Lu X, Cui P, et al. Phase I/II clinical trial of a Wilms’ tumor
entered clinical trial employ this approach [7–12]. There are 1-targeted dendritic cell vaccination-based immunotherapy in
several advantages to this approach as previously mentioned. patients with advanced cancer. Cancer Immunol Immunother.
This method bypasses DCs antigen presentation in a highly 2019;68:121–130.
controllable environment so T cells priming can occur. This 9. Liau LM, Ashkan K, Tran DD, et al. First results on survival from
a large phase 3 clinical trial of an autologous dendritic cell vaccine
approach ensures the generation of a high-quality DCs in in newly diagnosed glioblastoma. J Transl Med. 2018;16:142.
sufficient quantities for T cells priming. Furthermore, ex vivo 10. Wang Q-T, Nie Y, Sun S-N, et al. Tumor-associated antigen-based
loading produces a vaccine with very few adverse reactions personalized dendritic cell vaccine in solid tumor patients. Cancer
due to its autologous properties. Immunol Immunother. 2020;69:1375–1387.
However, the cost of production of this approach is consider­ 11. Bulgarelli J, Tazzari M, Granato AM, et al. Dendritic cell vaccination
in metastatic melanoma turns “Non-T Cell inflamed” Into “T-Cell
ably high, but this depends on the number of productions. inflamed” Tumors. Front Immunol. 2019;10:2353.
Vaccine production by this method also requires certain facilities, 12. Lee JM, Lee M-H, Garon E, et al. Phase I Trial of Intratumoral
so wide distribution might not be practical. On the other hand, Injection of CCL21 Gene-Modified dendritic cells in lung cancer
the possibility of inducing long-term and broader immunity will elicits Tumor-Specific immune responses and CD8(+) T-cell
eliminate the need for frequent boosters. So that, the total cost infiltration. Clin Cancer Res. 2017;23:4556–4568.
13. Lu W, Arraes LC, Ferreira WT, et al. Therapeutic dendritic-cell vac­
of production might eventually become cost-effective in com­ cine for chronic HIV-1 infection. Nat Med. 2004;10:1359–1365.
parison to conventional vaccine. Production at point-of-care, 14. Chen M, Y-G L, Zhang D-Z, et al. Therapeutic effect of autologous
such as hospital and medical laboratories, can be the solution dendritic cell vaccine on patients with chronic hepatitis B: a clinical
to practical limitation of this approach. In conclusion, this study. World J Gastroenterol. 2005;11:1806–1808.
approach is appropriate for the COVID-19 vaccine, and the safety 15. Decker WK, Safdar A. Dendritic cell vaccines for the immunocom­
promised patient: prevention of influenza virus infection. Expert
profile of this approach should allow clinical study to proceed. Rev Vaccines. 2010;9:721–730.
16. Marshall JS, Warrington R, Watson W, et al. An introduction to
immunology and immunopathology. Allergy, Asthma Clin
Funding Immunol. 2018;14:5–14.
This paper was funded by Cellcure Center RSPAD Gatot Soebroto. 17. Varadé J, Magadán S, González-Fernández Á. Human immunology
and immunotherapy: main achievements and challenges. Cell Mol
Immunol. 2021;18:805–828.
18. Stambas J, Lu C, Tripp RA. Innate and adaptive immune responses
Declaration of interest in respiratory virus infection: implications for the clinic. Expert Rev
The authors have no relevant affiliations or financial involvement with any Respir Med. 2020;14:1141–1147.
organization or entity with a financial interest in or financial conflict with 19. Riera Romo M, Pérez-Martínez D, Castillo Ferrer C. Innate immunity
the subject matter or materials discussed in the manuscript. This includes in vertebrates: an overview. Immunology. 2016 April 05.
employment, consultancies, honoraria, stock ownership or options, expert 2016;148:125–139.
testimony, grants or patents received or pending, or royalties. 20. Bieber K, Autenrieth SE. Dendritic cell development in infection.
Mol Immunol. 2020;121:111–117.
21. Ivashkiv LB, Donlin LT. Regulation of type I interferon responses.
Reviewer disclosures Nat Rev Immunol. 2014;14:36–49.
22. Mitchell D, Chintala S, Dey M. Plasmacytoid dendritic cell in immu­
Peer reviewers on this manuscript have no relevant financial or other nity and cancer. J Neuroimmunol. 2018;322:63–73.
relationships to disclose.
23. Amon L, Lehmann CHK, Baranska A, et al. Transcriptional control of
dendritic cell development and functions. 1st (Amsterdam,
Nedherland: Elsevier Inc) ed. Int. Rev. Cell Mol. Biol. Elsevier Inc.;
ORCID 2019.
Jonny Jonny http://orcid.org/0000-0002-8564-7430 24. Reizis B. Plasmacytoid dendritic cells: development, regulation, and
Enda Cindylosa Sitepu http://orcid.org/0000-0002-8112-7198 function. Immunity. 2019;50:37–50.
EXPERT REVIEW OF VACCINES 9

25. Oth T, Vanderlocht J, Van Elssen CHMJ, et al. Pathogen- 50. Lindenbergh MFS, Stoorvogel W. Antigen presentation by extra­
Associated molecular patterns induced crosstalk between den­ cellular vesicles from professional Antigen-Presenting cells. Annu
dritic cells, T helper cells, and natural killer helper cells can Rev Immunol. 2018;36:435–459.
improve dendritic cell vaccination. Mediators Inflamm. 2016; 51. Tiberio L, Del Prete A, Schioppa T, et al. Chemokine and chemo­
2016. 1–12 tactic signals in dendritic cell migration review-article. Cell Mol
26. Lee AJ, Ashkar AA. The dual nature of type I and type II interferons. Immunol. 2018;15. 15
Front Immunol. 2018. p. 2061. 9 52. Ugur M, Mueller SN. T cell and dendritic cell interactions in lym­
27. Kwon B. IFN-γ in tissue-immune homeostasis and antitumor immu­ phoid organs: more than just being in the right place at the right
nity. Cell Mol Immunol. 2018;15:531–532. time. Immunol Rev. 2019;289:115–128.
28. Oth T, Habets THPM, Germeraad WTV, et al. Pathogen recognition 53. Gurevich I, Feferman T, Milo I, et al. Active dissemination of cellular
by NK cells amplifies the pro-inflammatory cytokine production of antigens by DCs facilitates CD8+ T-cell priming in lymph nodes. Eur
monocyte-derived DC via IFN-γ. BMC Immunol. 2018;19:1–13. J Immunol. 2017;47:1802–1818.
29. Kang S, Kumanogoh A. The spectrum of macrophage activation by 54. Miller MJ, Hejazi AS, Wei SH, et al. T cell repertoire scanning is
immunometabolism. Int Immunol. 2020;32:467–473. promoted by dynamic dendritic cell behavior and random T cell
30. Gupta U, Hira SK, Singh R, et al. Essential role of TNF-α in gamma motility in the lymph node. Proc Natl Acad Sci U S A.
c cytokine aided crosstalk between dendritic cells and natural killer 2004;101:998–1003.
cells in experimental murine lymphoma. Int Immunopharmacol. 55. Celli S, Day M, Müller AJ, et al. How many dendritic cells are
2020;78:106031. required to initiate a T-cell response? Blood. 2012;120:3945–3948.
31. Kobayashi S, Sakurai T, So T, et al. TNF Receptor–Associated factor 5 56. Schmidt ME, Varga SM. The CD8 T cell response to respiratory virus
limits function of plasmacytoid dendritic cells by controlling IFN infections. Front Immunol. 2018;9. 10.3389/fimmu.2018.00678
regulatory factor 5 expression. J Immunol. 2019;203:1447–1456. 57. Reed J, Wetzel SA. CD4 + T cell differentiation and activation.
32. Liu S-Q, Ren C, Yao R-Q, et al. TNF-α-induced protein 8-like 2 Methods Mol Biol. 2018 1803 . p. 335–351.
negatively regulates the immune function of dendritic cells by 58. Koutsakos M, Nguyen THO, Kedzierska K. With a little help from
suppressing autophagy via the TAK1/JNK pathway in septic mice. T follicular helper friends: humoral immunity to influenza
Cell Death Dis. 2021 12 ;1032. vaccination. J Immunol. 2019;202:360–367.
33. Murphy K, Weaver C. Janeway’s immunobiology. 9th ed. New York 59. Hirano T, Murakami M. COVID-19: a new virus, but a familiar recep­
and London: Garland Science : Taylor & Francis Group; 2017. tor and cytokine release syndrome. Immunity. 2020;52:731–733.
34. Netea MG, Schlitzer A, Placek K, et al. Innate and adaptive immune 60. Wang K, Chen W, Zhang Z, et al. CD147-spike protein is a novel
memory: an evolutionary continuum in the hosts response to route for SARS-CoV-2 infection to host cells. Signal Transduct
pathogens. Cell Host Microbe. 2019;25:13–26. Target Ther. 2020;1–10. 5
35. Sun L, Wang X, Saredy J, et al. Innate-adaptive immunity interplay 61. Taefehshokr N, Taefehshokr S, Hemmat N, et al. Covid-19: perspec­
and redox regulation in immune response. Redox Biol. tives on innate immune evasion. Front Immunol. 2020;11:580641.
2020;37:101759. 62. Cai G, Du M, Boss Y, et al. SARS-CoV-2 impairs dendritic cells and
36. Normann N, Tietz G, Kühn A, et al. Pathogen-specific antibody regulates DC-SIGN gene expression in tissues. Int J Mol Sci.
profiles in patients with severe systemic infections. Eur Cells 2021;22:1–20.
Mater. 2020;39:171–182. 63. Chu H, Zhou J, Ho-Yin Wong B, et al. Productive replication of
37. Balan S, Saxena M, Bhardwaj N. Dendritic cell subsets and locations. middle East respiratory syndrome coronavirus in
1st (Amsterdam, Netherland: Elsevier Inc) ed. Int. Rev. Cell Mol. Biol. monocyte-derived dendritic cells modulates innate immune
Elsevier Inc. 2019. response. Virology. 2014;454–455:197–205.
38. Constantino J, Gomes C, Falcão A, et al. Dendritic cell-based immu­ 64. Yang D, Chu H, Hou Y, et al. Attenuated interferon and proinflam­
notherapy: a basic review and recent advances. Immunol Res. matory response in SARS-CoV-2-infected human dendritic cells is
2017;65:798–810. associated with viral antagonism of STAT1 phosphorylation. J Infect
39. Collin M, Bigley V. Human dendritic cell subsets: an update. Dis. 2020;222:734–745.
Immunology. 2018;154:3–20. 65. Chi Y, Ge Y, Wu B, et al. Serum cytokine and chemokine profile in
40. Villadangos JA, Schnorrer P. Intrinsic and cooperative relation to the severity of coronavirus disease 2019 in China.
antigen-presenting functions of dendritic-cell subsets in vivo. Nat J Infect Dis. 2020;222:746–754.
Rev Immunol. 2007;7:543–555. 66. Huang C, Wang Y, Li X, et al. Clinical features of patients infected
41. Alculumbre SG, Saint-André V, Di Domizio J, et al. Diversification of with 2019 novel coronavirus in Wuhan, China. Lancet.
human plasmacytoid predendritic cells in response to a single 2020;395:497–506.
stimulus article. Nat Immunol. 2018;19:63–75. 67. Tiwari-Heckler S, Rauber C, Longhi MS, et al. Dysregulated host
42. Worbs T, Hammerschmidt SI, Förster R. . Nat. Rev. Immunol. (Berlin, response in severe acute respiratory syndrome coronavirus
Germany: Nature Publishing Group); 2017. p. 30–48. 2-Induced critical illness. Open Forum Infect Dis. 2021;8:1–9.
43. Hilligan KL, Ronchese F. Antigen presentation by dendritic cells and 68. Niles MA, Gogesch P, Kronhart S, et al. Macrophages and den­
their instruction of CD4+ T helper cell responses. Cell Mol Immunol. dritic cells are not the major source of Pro-Inflammatory cyto­
2020;17:587–599. kines upon SARS-CoV-2 infection. Front Immunol. 2021;12:1–12.
44. Fitzgerald KA, Kagan JC. Toll-like receptors and the control of 69. Schulte-Schrepping J, Reusch N, Paclik D, et al. Severe
immunity. Cell. 2020;180:1044–1066. COVID-19 is marked by a dysregulated myeloid cell
45. Kim MK, Kim J. Properties of immature and mature dendritic cells: compartment. Cell. 2020;182:1419–1440.e23.
phenotype, morphology, phagocytosis, and migration. RSC Adv. 70. Zhou R, Kkw T, Wong YC, et al. Acute SARS-CoV-2 infection
2019;9:11230–11238. impairs dendritic cell and T cell responses. Immunity.
46. Shin KS, Jeon I, Kim BS, et al. Monocyte-derived dendritic cells 2020;53:1–14.
dictate the memory differentiation of CD8+ T cells during acute 71. Blanco-Melo D, Nilsson-Payant BE, Liu WC, et al. Imbalanced host
infection. Front Immunol. 2019;10:5–7. response to SARS-CoV-2 drives development of COVID-19. Cell.
47. Gu Y, Zhao X, rong SX. Ex vivo pulsed dendritic cell vaccination 2020;181:1036–1045.e9.
against cancer. Acta Pharmacol Sin. 2020;41:959–969. 72. Ribero MS, Jouvenet N, Dreux M, et al. Interplay between
48. Blum JS, Wearsch PA, Cresswell P. Pathways of antigen processing. SARS-CoV-2 and the type I interferon response. PLoS Pathog.
Annu Rev Immunol. 2013;31:443–473. 2020;16:1–22.
49. Roche PA, Cresswell P. . Myeloid Cells Heal Dis A Synth (Wiley 73. Peiris JSM, Chu CM, Cheng VCC Clinical progression and viral load
Online Book). 2017;209–223:9781683670667 doi:10.1128/ in a community outbreak of coronavirus-associated SARS pneumo­
9781555819194.ch11. nia: a prospective study , et al. Lancet. , . 2003;361:1767–1772.
10 JONNY ET AL.

74. Channappanavar R, Fehr AR, Vijay R, et al. Dysregulated Type 91. Tarke A, Sidney J, Methot N, et al. Impact of SARS-CoV-2 variants on
I interferon and inflammatory Monocyte-Macrophage responses the total CD4+ and CD8+ T cell reactivity in infected or vaccinated
cause lethal Pneumonia in SARS-CoV-Infected mice. Cell Host individuals. Cell Reports Med. 2021;2:100355.
Microbe. 2016;19:181–193. 92. Laidlaw BJ, Ellebedy AH. The germinal centre B cell response to
75. Zhang Q, Liu Z, Moncada-Velez M, et al. Inborn errors of type I IFN SARS-CoV-2. Nat Rev Immunol. 2022;22:7–18.
immunity in patients with life-threatening COVID-19. Science (80). 93. Sabado RL, Balan S, Bhardwaj N. Dendritic cell-based
2020;370. immunotherapy. Cell Res. 2017;27:74–95.
76. Galati D, Zanotta S, Capitelli L, et al. A bird’s eye view on the role of 94. Martínez L, Malaina I, Salcines-Cuevas D, et al. First computational
dendritic cells in SARS-CoV-2 infection: perspectives for immune- design using lambda-superstrings and in vivo validation of
based vaccines. Allergy. 2021: 00. p. 1–11. SARS-CoV-2 vaccine. Sci Rep. 2022;12:2–13.
77. Bastard P, Rosen LB, Zhang Q, et al. Autoantibodies against type 95. Tacken PJ, De Vries IJM, Torensma R, et al. Dendritic-cell immu­
I IFNs in patients with life-threatening COVID-19. Science (80). notherapy: from ex vivo loading to in vivo targeting. Nat Rev
2020;370. Immunol. 2007. p. 790–802. 7
78. Chen Z, John Wherry E. T cell responses in patients with COVID-19. 96. Dillman RO, Cornforth AN, Nistor GI, et al. Randomized phase II trial
Nat Rev Immunol. 2020;20:529–536. of autologous dendritic cell vaccines versus autologous tumor cell
79. Du RH, Liang LR, Yang CQ, et al. Predictors of mortality for patients vaccines in metastatic melanoma: 5-year follow up and additional
with COVID-19 pneumonia caused by SARSCoV- 2: a prospective analyses. J Immunother Cancer. 2018;6:19.
cohort study. Eur Respir J. 2020;3:55. 97. Wang X, Bayer ME, Chen X, et al. Phase I trial of active specific
80. Xu B, yu FC, lu WANGA, et al. Suppressed T cell-mediated immunity immunotherapy with autologous dendritic cells pulsed with
in patients with COVID-19: a clinical retrospective study in Wuhan, autologous irradiated tumor stem cells in hepatitis B-positive
China. J Infect. 2020;81:e51–e60. patients with hepatocellular carcinoma. J Surg Oncol.
81. Diao B, Wang C, Tan Y, et al. Reduction and functional exhaustion 2015;111:862–867.
of T cells in patients with coronavirus disease 2019 (COVID-19). 98. Surenaud M, Montes M, Lindestam Arlehamn CS, et al. Anti-HIV
Front Immunol. 2020. p. 827. 11 potency of T-cell responses elicited by dendritic cell therapeutic
82. Chen J, Qi T, Liu L, et al. Clinical progression of patients with vaccination. PLoS Pathog. 2019;15:1–21.
COVID-19 in Shanghai, China. J Infect. 2020;80:e1–e6. 99. Leplina O, Starostina N, Zheltova O, et al. Dendritic cell-based
83. Parackova Z, Zentsova I, Bloomfield M. Disharmonic inflammatory vaccines in treating recurrent herpes labialis: results of pilot clinical
signatures in COVID-19 : augmented neutrophils’ but impaired study. Hum Vaccines Immunother. 2016;12:3029–3035.
monocytes’ and dendritic cells’ responsiveness. Cells. 2020;9. 10 100. Jonny J. Preventive dendritic cell vaccine, AV-COVID-19, in subjects
2206 not actively infected with COVID-19 [Internet]. Identifier
84. Law HKW, Cheung CY, Sia SF, et al. Toll-like receptors, chemokine NCT00103181. cited 2022 Apr 27]. Available from 2022 Apr 27:
receptors and death receptor ligands responses in SARS corona­ https://clinicaltrials.gov/ct2/show/NCT05007496.
virus infected human monocyte derived dendritic cells. BMC 101. Khuroo MS, Khuroo M, Khuroo MS, et al. COVID-19 vaccines: a race
Immunol. 2009;10:1–12. against time in the middle of death and devastation! J Clin Exp
85. Zhu L, Yang P, Zhao Y, et al. Single-Cell sequencing of peripheral Hepatol. 2020;10:610–621.
mononuclear cells reveals distinct immune response landscapes of 102. Sung SSJ. Monocyte-derived dendritic cells as antigen-presenting
COVID-19 and influenza patients. Immunity. 2020;53:685–696.e3. cells in T-cell proliferation and cytokine production. Methods Mol
86. von Karstedt S, Montinaro A, Walczak H. Exploring the TRAILs less Biol. 2019;2020:131–141.
travelled: TRAIL in cancer biology and therapy. Nat Rev Cancer. 103. Wong C, Goldstein DR. Impact of aging on antigen presentation cell
2017;17:352–366. function of dendritic cells. Curr Opin Immunol. 2013;25:535–541.
87. Catanzaro M, Fagiani F, Racchi M, et al. Immune response in 104. Zacca ER, Crespo MI, Acland RP, et al. Aging Impairs the ability
COVID-19: addressing a pharmacological challenge by targeting of conventional dendritic cells to cross-prime CD8+ T cells upon
pathways triggered by SARS-CoV-2. Signal Transduct Target Ther.
stimulation with a TLR7 Ligand. PLoS One. 2015;10:1–20.
2020;5. 10.1038/s41392-020-0191-1
105. Li S, Wu J, Zhu S, et al. Disease-associated plasmacytoid dendri­
88. DeDiego ML, Nieto-Torres JL, Regla-Nava JA, et al. Inhibition of NF-
tic cells. Front Immunol. 2017;8:1–12.
B-Mediated inflammation in severe acute respiratory syndrome
106. Asano T, Immunol S, Asano T, et al. X-linked recessive TLR7 defi­
Coronavirus-Infected mice increases survival. J Virol.
ciency in ~ 1 % of men under 60 years old with life-threatening
2014;88:913–924.
COVID-19. Sci Immunol. 2021;4348:1–31.
89. Jung JH, Rha MS, Sa M, et al. SARS-CoV-2-specific T cell memory is
sustained in COVID-19 convalescent patients for 10 months with 107. Forsdyke DR. Lymphocyte repertoire selection and intracellular
successful development of stem cell-like memory T cells. Nat self/non-self-discrimination: historical overview. Immunol Cell
Commun. 2021;12:1–12. Biol. 2015;93:297–304.
90. Le Bert N, Tan AT, Kunasegaran K, et al. SARS-CoV-2-specific T cell 108. Mekonnen ZA, Masavuli MG, Yu W, et al. Enhanced T cell responses
immunity in cases of COVID-19 and SARS, and uninfected controls. induced by a necrotic dendritic cell vaccine, expressing HCV NS3.
Nature. 2020;584:457–462. Front Microbiol. 2020;11:1–13.

You might also like