Next Article in Journal
Development of an Improved Method for the Determination of Iodine/β-Cyclodextrin by Means of HPLC-UV: Validation and the Thyroid-Stimulating Activity Revealed by In Vivo Studies
Next Article in Special Issue
DENV-Mimetic Polymersome Nanoparticles Bearing Multi-Epitope Lipopeptides Antigen as the Next-Generation Dengue Vaccine
Previous Article in Journal
Ophthalmic In Situ Gels with Balsam Poplar Buds Extract: Formulation, Rheological Characterization, and Quality Evaluation
Previous Article in Special Issue
The Effect of a TLR4 Agonist/Cationic Liposome Adjuvant on Varicella-Zoster Virus Glycoprotein E Vaccine Efficacy: Antigen Presentation, Uptake, and Delivery to Lymph Nodes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mimicking Pathogens to Augment the Potency of Liposomal Cancer Vaccines

1
Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
2
Department of Pharmaceutics, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
3
Department of Biomaterials, Science and Technology, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, The Netherlands
4
Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
*
Author to whom correspondence should be addressed.
Pharmaceutics 2021, 13(7), 954; https://doi.org/10.3390/pharmaceutics13070954
Submission received: 20 May 2021 / Revised: 21 June 2021 / Accepted: 22 June 2021 / Published: 24 June 2021
(This article belongs to the Special Issue Liposomes for Vaccine Delivery)

Abstract

:
Liposomes have emerged as interesting vehicles in cancer vaccination strategies as their composition enables the inclusion of both hydrophilic and hydrophobic antigens and adjuvants. In addition, liposomes can be decorated with targeting moieties to further resemble pathogenic particles that allow for better engagement with the immune system. However, so far liposomal cancer vaccines have not yet reached their full potential in the clinic. In this review, we summarize recent preclinical studies on liposomal cancer vaccines. We describe the basic ingredients for liposomal cancer vaccines, tumor antigens, and adjuvants, and how their combined inclusion together with targeting moieties potentially derived from pathogens can enhance vaccine immunogenicity. We discuss newly identified antigen-presenting cells in humans and mice that pose as promising targets for cancer vaccines. The lessons learned from these preclinical studies can be applied to enhance the efficacy of liposomal cancer vaccination in the clinic.

1. Introduction

Liposomes are versatile drug delivery vehicles because of their ability to transfer both hydrophobic and hydrophilic substances. In addition, liposomes are known for their low immunogenicity, a good safety profile, and their biocompatibility. Hence, interest in liposomal delivery vehicles has emerged in various fields [1]. In the field of oncology, the last decades have been marked by major developments in combating cancer cells with chemotherapy or molecular targeted therapies (e.g., targeting of the proteasome, receptors for growth factors) [2]. However, the systemic (soluble) application of these therapies can have severe side effects, thereby limiting the therapeutic window [2]. In order to facilitate a more effective and safe therapy, liposomes then emerged as vehicles to directly deliver these drugs to cancer cells [3]. Furthermore, liposomes gained interest because of their potential use in modulating the tumor microenvironment by direct delivery of immunomodulatory drugs to tumors. Indeed, liposomes can enter tumors because of the enhanced permeability and retention effect caused by the relatively permeable (“leaky”) tumor vasculature [4]. Simultaneously, major developments in the field of immunology were made to engage the immune system against cancer [5,6]. As cancers gain mutations and start to aberrantly overexpress tissue-restricted proteins during malignant transformation, they are recognized by the immune system as “non-self”. Hence, similar to infectious agents, this opens a therapeutic window to harness the immune system in the fight against “non-self” [7]. Indeed, immunotherapies that reinvigorate the exhausted anti-cancer immune response (e.g., immune checkpoint blockade) have been successful in a limited number of cancers and patients, and it is envisaged that this could be enhanced by means of vaccination [8,9,10].
In contrast to vaccinations against infectious agents that require strong induction of (neutralizing) antibody responses, cancer vaccination strategies are more focused toward induction of cytotoxic CD8+ T cell responses that are capable of tumor cell killing [11]. This indicates that direct translation of methods used in vaccines against infectious diseases may be less effective in cancer vaccination. In addition, due to the low immunogenicity of cancer antigens, vaccine effectiveness is often suboptimal, pointing to the need to improve the potency of vaccine technologies [12]. Hence, liposomes, and also other nanoparticulate carriers, received great interest in immune-oncology as carriers of antigens and adjuvants. Optimized cancer vaccines in combination with immune checkpoint blockade can potentially synergistically induce antitumor immunity by eliciting highly effective de novo or reinvigorated T cell responses and, consequently, improved clinical responses [5,6,13].
In this review, we aim to provide an overview of preclinical studies that address the efficacy of liposomal cancer vaccines and relate it to immunological processes important for T cell priming. Although there is a wide variety of nanoparticulate carriers that can be used in vaccination strategies, we have limited ourselves to liposomes and occasionally include related lipoplexes, as the latter lipid-based carriers are often used and particularly designed for the delivery of nucleic acids encoding for antigens. In this review, we will first discuss the characteristics of the two major cancer vaccine components, antigen and adjuvant, and how the incorporation of these two components into liposomes enhances their immunogenicity or potency, respectively. Secondly, we discuss how the different administration routes and biochemical characteristics of liposomes influence uptake in secondary lymphoid organs, presentation by antigen-presenting cells (APCs) and T cell priming. We provide an overview of different types of APCs, including newly identified subsets, and discuss their functions in T cell priming. Finally, we describe how the incorporation of targeting moieties derived from pathogens leads to more efficient uptake by specific APCs and further enhances the immunogenicity of vaccines. In summary, liposomes that incorporate antigen, stimulatory adjuvant, and targeting moieties start to resemble pathogens, and this pathogen mimicry may provide perspectives for the improvement of liposomal cancer vaccines in the near future.

2. Antigen and Adjuvant Are the Basic Ingredients of Vaccines

2.1. Classification of Tumor Antigens

The immune system recognizes foreign antigens and can react to these molecules that are present in pathogens, infected, or malignantly transformed cells. One class of antigens that is present in cancer cells is normal proteins that are aberrantly expressed or overexpressed. These antigens are normally restricted to distinct tissues such as the testis or are only expressed at low levels and are termed cancer testis antigens (CTAs) and tumor associated antigens (TAAs). These antigens can be immunogenic and are potential cancer antigens that can be used for vaccination strategies [12]. Since these antigens are shared by multiple patients, they present as possible targets for “off the shelf” vaccination strategies. However, CTAs and TAAs are normal proteins and can therefore be classified as “self”. Tolerance to self-antigens is achieved in the thymus, in which T cells with a high affinity T cell receptor (TCR) for self-antigens are deleted. As a consequence, remaining T cells that may react to these antigens have a low affinity TCR, which may hamper their effectiveness. This is especially relevant for TAA-specific T cells, as tolerance induction for CTA-specific T cells is less stringent [12].
In addition to CTAs and TAAs, tumor cells are marked by a high mutational load that leads to an accumulation of proteins with mutations that are termed as neoantigens or tumor-specific antigens [14]. These mutations are highly specific for a patient or a model used and can be identified by means of high-throughput screening methods (e.g., next generation sequencing). Since no tolerance to newly developed mutations exist, neoantigens are considered to be highly immunogenic and can elicit responses from T cells with a high affinity TCR [15]. Thus, cancer antigens can be broadly divided in aberrant or overexpressed proteins shared by multiple patients and newly developed mutations that are highly patient-specific, and both types of antigens can be included in cancer vaccines.

2.2. Vaccine Antigen Can Be Incorporated in Various Forms

While CD8+ cytotoxic T cells recognize peptides in the context of major histocompatibility complex (MHC), vaccination strategies can make use of different forms of the antigen to prime the immune system, such as peptides, protein, DNA, or RNA. Initially, short peptides that represent viral epitopes were used to vaccinate against virally induced cancers [16]. However, short peptides were subsequently found to be prone to tolerance induction and, instead, longer peptides that do need processing, like protein antigen, were preferred [16].
In addition to long peptides and protein antigen, more recently major developments in nucleotide vaccines have been made. This is highlighted by the recent effectiveness of mRNA vaccines against COVID-19 [17]. This vaccination platform emerged from a longer standing interest in mRNA vaccines to treat cancer [18]. Indeed, an mRNA-based vaccination platform was used for cancer patients, where mRNA encoded multiple neoantigens that were present in their tumors [19]. In addition, mRNA vaccines can also be used to encode multiple TAAs [20,21]. The former possibility opens a window to personalized cancer vaccines and the latter one to “off the shelf” cancer vaccines. In order to augment the potency of these vaccines, mRNA that encodes CD4+ helper T cell epitopes may also be included in the vaccine formulation [22].
In comparison to peptide and/or protein antigen, mRNA molecules are known to be relatively instable, which may hamper their effectiveness. In order to overcome this hurdle, multiple modifications to the 5′-cap, poly(A) tail and 5′ and 3′ untranslated regions can be made to make the mRNA more stable and effective. These alterations enhance the magnitude of the response (translation efficiency) as well as the half-life of mRNA (duration of the response) [18]. Another potential drawback of these vaccines is the need for uptake of relatively large negatively charged mRNA molecules by cells, which is generally considered to be relatively inefficient. However, especially immature dendritic cells (DCs) are able to take up naked mRNA molecules, albeit to a low extent. Importantly, vaccination with naked mRNA successfully resulted in the priming of antigen-specific T cell responses upon intradermal or intranodal administration [23,24]. In order to enhance the stability and/or uptake of these vaccines, mRNA is often complexed with lipids to create lipoplexes, which can further augment vaccine effectiveness [20,21].

2.3. Cancer Vaccination Aims to Induce CD8+ T Cell Responses via Cross-Presenting DCs

In order to mount a cytotoxic T cell response specific to cancer cells, CD8+ T cells should be activated by DCs that present cancer antigens [25]. DCs can take up cancer antigens at the tumor site or in the draining lymph node (LN). Subsequently, DCs process and present these antigens in their MHC molecules, MHC class I or II to elicit responses by CD8+ or CD4+ T cells, respectively [26,27]. MHC class I is present on all nucleated cells and is usually loaded with endogenous peptides derived from the cytosol. However, DCs have the unique capacity to cross-present peptides in MHC class I. Here, peptides are derived from exogenous antigens that have been phagocytosed, such as dying cancer cells. This latter process is called cross-presentation and is restricted to DCs. However, different types of DCs exist and not all DCs have the same capacity for cross-presentation (further described in “Different APCs for T cell priming”) [28].
Two major routes have been described for cross-presentation, the cytosolic and vacuolar route [29,30]. Cross-presentation via the cytosolic pathway requires antigen translocation to the cytosol, proteasomal degradation of antigen, and subsequent TAP-dependent transport to the endoplasmic reticulum (ER) or back to the phagosome. Hence, antigen should escape, or be released, from phagosomal compartments to gain access to the cytosol. Although the mechanism of transport is under debate, recent work has shown that phagosomal rupture is a potential mechanism [31]. In contrast to the cytosolic route, cross-presentation via the vacuolar route occurs fully in endo-phagosomal compartments and is not dependent on cytosolic proteases, but is sensitive to inhibition of lysosomal proteolysis [26,32]. Interestingly, the mechanism of vaccine uptake influences antigen routing and subsequent cross-presentation (further described in “Active targeting leads to uptake by APCs of choice”) [33,34]. Thus, CD8+ T cell responses are elicited by cross-presenting DCs, and the efficiency of this process is largely determined by the targeted DC subtype and its capacity of cross-present.

2.4. Vaccine Adjuvants Are Essential to Induce an Immune Response

In addition to antigen, cancer vaccines should be supplemented with an adjuvant to mature DCs, which can subsequently provide essential costimulatory signals to naïve T cells during their priming. Adjuvant-induced DC maturation enhances the density of MHC–peptide complexes on the cell surface, but also stimulates expression of costimulatory markers such as CD40, CD80, and CD86, while simultaneously triggering cytokine secretion [35]. The maturation of DCs can be triggered by bacterial or viral infections, and cancer vaccines can include bacterial or viral components as adjuvant. These bacterial or viral components consist of pathogen-associated molecular patterns (PAMPs) that can be recognized by pattern recognition receptors, such as a toll-like receptors (TLRs), present on DCs [36,37]. TLRs can be found on the cell surface, but can also reside in endosomal compartments, dependent on the nature and localization of their ligand [36,37]. Importantly, TLRs are not ubiquitously expressed by all DC types [38]. In addition to TLRs, innate immune receptors in the cytosol, such as RIG-I, MDA-5, and STING, may be activated to provide immune activation [39,40,41]. When a cancer vaccine contains nucleotides (i.e., DNA or RNA) the vaccine may not require additional TLR ligands or other activation signals due to the intrinsic adjuvant effect caused by binding of nucleotides to TLRs [20].
Interestingly, next to the effects of adjuvant on DC maturation and costimulatory capacity, TLR stimulation was also found to promote fusion of phagosomes (containing exogenous antigen) with endosomal recycling compartments (containing MHC class I molecules). Thereby, exogenous antigen is brought in close proximity to MHC class I-containing compartments, which promotes antigen cross-presentation [42,43,44]. As this route of antigen loading is distinct from endogenous antigen loading on MHC class I (in the endoplasmatic reticulum), this points to a preferential stimulation of the cross-presentation route upon TLR stimulation [45].
In conclusion, two major prerequisites for successful cancer vaccines are inclusion of an antigen, to which the subsequent immune response should be directed, and an immune stimulating agent, adjuvant, to ensure proper DC maturation. Importantly, since many cell types can potentially take up vaccine components but cannot induce T cell responses, specific targeting of DC subsets with both vaccine antigen and adjuvant, taking heterogeneous expression of TLR and antigen processing capacity into account, can increase vaccine potency.

3. Liposomes as Delivery System

3.1. Liposomal Antigen Accumulation in Lymph Nodes

Since CD8+ T cell activation by DCs can only occur in secondary lymphoid organs (LN) or spleen, antigens that are administered in other tissues should be transported to these lymphoid organs. After antigen injection via conventional routes (e.g., intradermal (i.d.), intramuscular (i.m.), or subcutaneous (s.c.)), antigen can accumulate in the LN via local uptake at the site of injection by tissue-resident APCs that subsequently migrate to the LN. Alternatively, antigen itself may passively drain from the site of injection via the lymphatic system to the LN. The potential use of liposomes and their physicochemical properties largely determine how and where the immune system encounters antigen (Table 1). For example, the majority of freely administered (i.e., not encapsulated) protein antigen was found to drain from the site of injection within a day, while encapsulation of the antigen in cationic liposomes resulted in electrostatic interactions with the tissue and the retention of antigen at the site of injection (i.m.) over multiple days [46]. In fact, cationic liposomes appear to serve as a depot from which antigen is slowly released over time [46]. In contrast, neutral and anionic liposomes drain more easily upon i.m. injection to the LN [47,48]. The sustained release characteristics of a liposomal antigen depot are dependent on the charge, membrane fluidity, and retention time of the liposomes at the injection site [49]. In addition, direct interaction of the liposomes with tissue-resident APCs and subsequent internalization by these cells can also be an important determining factor [50]. Thus, the degree of liposome uptake is dependent on surface charge and membrane fluidity as they influence the interaction with the extracellular matrix and cells.
Surface stabilization of liposomes with polymers (e.g., polyethylene glycerol (PEG)) can result in enhanced draining of the liposome particles from the site of injection [51]. While relatively low mole percentages of PEG (1 mol %) were indeed found to improve draining to the regional lymphatics and subsequent localization in the LN, incorporation of a higher quantity of PEG (5 mol %) facilitated lymphatic drainage, but simultaneously hindered antigen accumulation in the draining LN due to lower antigen uptake by APCs [52]. PEG can also shield phospholipids in liposomal preparations, so that specific recognition of, for example, phosphatidylserine (PS), that is normally present on apoptotic cells and is a ligand for APCs, is hindered [47,53]. Similarly, PEG can shield specific targeting moieties on the liposome surface, and therefore liposomal stabilization with PEG may have an unfavorable effect on targeting capacity [54]. However, coupling of targeting ligands to the terminal ends of PEG can be utilized to overcome this [55]. Of note, liposomal surface decoration with PEG also makes the liposomes immunogenic [56]. Thus, the altered targeting efficiency and immunogenicity indicate that addition of PEG to liposomal preparations should be carefully assessed.
The size of liposome particles further influences the capacity of liposomes to drain to the LN as particles that were smaller than 150 nm drained to the regional lymphatics while larger particles stayed at the injection site after an s.c. injection [57,58]. Upon i.m. injection, smaller particles (100 nm) also drained rapidly, while larger particles (900 nm) were retained [47]. Thus, smaller particles tend to rapidly drain, while relatively large particles are not well equipped to diffuse to the lymphatic system and their localization in the LN is dependent on peripheral uptake and subsequent migration of tissue-resident immune cells [59].
Table 1. Physicochemical properties of liposomes determine liposomal behavior upon injection.
Table 1. Physicochemical properties of liposomes determine liposomal behavior upon injection.
SizeChargeAdministration RouteEffectReferences
~100 nmCationici.m.Depot formation[46]
~100 nmNeutral and anionici.m.Draining via the regional lymphatics to the LN[47]
‘Large’ 500–900 nmCationics.c.Retention at the site of injection[47,58]
‘Small’ 100–140 nmCationics.c.Enhanced draining to the LN as compared to larger counterparts[47,58]
~250 nmCationics.c.Liposomal surface decoration with PEG enhanced draining to the LN[52]
~300 nmNeutral or anionici.v.Antigen uptake in the spleen[20]
~160 nmCationici.v.Antigen sequestration in the lung[60]
Thus, the rate and extent of liposome drainage from the site of injection into the lymphatics are dependent on their physicochemical properties. In general, a cationic nature or large size of liposomes results in strong retention at the injection site and subsequent release of antigen (depot formation), while small, PEGylated, anionic, or neutral liposomes drain more easily to the LN (Table 1).

3.2. Liposomal Antigen Uptake in the Spleen

When liposomes are injected intravenously (i.v.), uptake or clearance of liposomes is mostly performed by phagocytic cells of the mononuclear phagocytic system (MPS) present in spleen and liver [61,62]. As the spleen is a lymphoid organ that mediates immune responses, i.v. administration is a potential route for vaccines to facilitate T cell priming [20,21,63]. Similar to other administration routes, physicochemical characteristics of the liposomes play a major role in their in vivo behavior. Hence, modifications of the surface of liposomes may be used to optimize splenic targeting. Indeed, i.v. administration of neutral or negatively charged lipoplexes resulted in considerable antigen uptake in the spleen [20]. However, administration of positively charged particles resulted in predominant uptake in the lung and, to a lesser extent, in the spleen and liver (Table 1) [60]. This difference in biodistribution between cationic and anionic lipoplexes was corroborated in a side-by-side comparison where cationic particles were found to predominantly accumulate in the lung and anionic particles in the spleen and liver [20,64]. Particle sequestration in the lung can be explained by rapid aggregation in the bloodstream induced by the interaction of cationic particles with circulating blood proteins and cells. This can result in rapid clearance from the bloodstream and can have serious side effects [65]. Importantly, the sequestration of these positively charged particles in the lung is likely dependent on the extent to which lipoplexes interact with blood-borne molecules and cells. When lipoplexes composed of different cationic lipids were assessed for their biodistribution, differences in the relative distribution over multiple organs (lungs, liver, spleen, and kidney) were found, indicating that sequestration is not solely dependent on charge, but also on the lipids used to complex with the nucleotides [66].
Thus, the physicochemical characteristics and the biodistribution profile of cancer vaccines that are intravenously administered should be carefully assessed and tailored to optimally induce a robust immune response.

3.3. The Route of Vaccine Administration Dictates Subsequent Responses

While the physicochemical characteristics of liposomes affect their subsequent interaction with tissues after immunization, alterations in the immunization route can also impact the quality and magnitude of the subsequent responses (Table 2) [67]. When cationic liposomes were evaluated, i.d. injection elicited superior anti-tumor responses as compared to s.c. immunization [68]. We observed a lower magnitude of the immune response upon s.c. immunization versus i.v. immunization when we evaluated anionic liposomes [54].
Although i.v. immunization is considered to be rather unconventional, i.v. administration of mRNA lipoplexes made it to the clinic and have been tested in patients with melanoma [20,21,63]. Lipoplexes were also tested for their effectiveness upon i.d., i.m., or s.c. administration. Intriguingly, the effectiveness of immunization with lipoplexes was found to be severely hampered upon s.c. or i.d. administration [69], which was in sharp contrast to the effectiveness of i.v. administration [20,21]. When the i.v. administration route was directly compared to the i.m./s.c. or to the i.d./s.c. route, respectively, both of these studies showed superior responses when lipoplexes were administered i.v. [70,71]. Intriguingly, s.c. delivery of lipoplexes was associated with release of type I interferon (IFN) that significantly hampered priming of cytotoxic T cells [69]. Conversely, the potent effect of i.v. administered lipoplexes was abrogated by blocking type I IFN signaling [71]. Hypothetically, sensing of either mRNA or IFN results in a cellular response that is accompanied by a lower translational activity, and thereby reduces the amount of protein antigen obtained with mRNA vaccines. In contrast, type I IFN has also been shown to promote DC maturation and to induce anti-cancer T cell responses [72,73]. In an attempt to shed light on these observations, van Hoecke and co-workers observed that a selective knockout (KO) of the type I IFN receptor (IFNR) on T cells improves immune responses upon s.c. administration of lipoplexes, but reduces T cell responses upon i.v. administration of lipoplexes [74]. In contrast to a selective IFNR-KO on T cells, a selective IFNR-KO on DCs was found to be of less importance [74]. This surprising finding indicates that the route of immunization has a major role in the potency of vaccination and that similar factors have a different role dependent on the route of immunization.
To conclude, these findings stress that successful vaccination with liposomes and lipoplexes is highly dependent on the route of administration and this should be carefully chosen on the basis of experimental in vivo studies.

3.4. Antigen Encapsulation Augments Antigen Immunogenicity

Uptake of soluble administered antigen is not solely restricted to professional APCs, which can hamper vaccine effectiveness, because steady-state scavenging non-professional APCs, such as liver sinusoid endothelial cells and lymphatic endothelial cells, are capable of antigen processing and presentation in MHC class I, leading to tolerance induction [75,76]. Furthermore, immunization with soluble minimal peptides that do not need processing also led to tolerance induction after an initial phase of activation. These minimal peptides directly bound to MHC class I molecules on non-activated cells that subsequently induced tolerance [16]. Therefore, cancer vaccine formulations should prevent erroneous off-targeting and use of soluble minimal peptides that may be taken up by non-professional APCs.
As previously mentioned, dependent on the size and lipid composition, liposomal encapsulation of antigen can lead to depot formation at the site of injection. Indeed, radioactively labeled antigen and liposomes were retained at the site of injection upon i.m. injection, in sharp contrast to antigen that was injected without liposomes and that was quickly cleared [77]. Interestingly, the difference in antigen retention also translated to differences in the magnitude of the immune response, as CD8+ T cell responses were observed when antigen was injected with liposomes, but not in absence of liposomes [77]. Indeed, because of the endured antigen accessibility and slower release characteristics, the immune reaction, as determined by CD8+ T cell responses, can increase in potency compared to administration of free peptide (Table 2) [78,79]. DCs were also shown to more efficiently present liposomal-associated peptide compared to soluble peptide [80,81]. When we assessed the peptide immunogenicity in vivo, we observed that incorporation of peptide in liposomes dramatically augmented the immunogenicity. While immunization with liposomal peptide resulted in potent CD8+ T cell responses, an equal dose of soluble antigen failed to elicit a noticeable immune response [54]. In agreement with these findings, encapsulation of peptide in liposomes was found to be an absolute requirement for anti-tumor activity [82]. In addition to peptide antigen, the immunogenicity of protein antigen was also found to be enhanced by incorporation in liposomes [83].
While the main focus in cancer vaccination is on CD8+ T cells, the role of CD4+ T cells should not be overlooked [84]. We observed that the priming of CD4+ T cells was also markedly enhanced upon immunization with liposomal, rather than soluble, peptide [54]. In an elaborate study, Varypataki and co-workers corroborated the importance of CD4+ T cell epitope inclusion and peptide encapsulation. They made use of OVA-derived synthetic long peptides (SLPs) containing either a CD8+ or a CD4+ T cell epitope. Immunization with mixed-in soluble SLPs and poly(I:C) as adjuvant of choice did not elicit a noticeable immune response. In contrast, when these SLPs with a CD8+ or CD4+ T cell epitope were loaded into liposomes the induction of antigen-specific immune responses upon immunization was markedly increased. In two different tumor models, enhanced immune responses and a delayed tumor growth were observed when antigens were administered in a liposomal context, further supporting the notion that antigen encapsulation is of importance for vaccine effectiveness [68].
Similar to protein or peptide antigen, formation of lipoplexes increased the potency of nucleotides as antigen in vitro [85]. While this is important for protection of nucleotide integrity, the uptake of RNA lipoplexes was also found to be enhanced [85,86].
Thus, for different types of antigen, liposomal encapsulation can facilitate slower antigen release and promote cellular antigen uptake, which leads to enhanced immune responses (Table 2).
Table 2. Liposomal encapsulation of antigen and adjuvant as well as surface modification with targeting moieties augments vaccine potency in vivo.
Table 2. Liposomal encapsulation of antigen and adjuvant as well as surface modification with targeting moieties augments vaccine potency in vivo.
Administration RouteAntigenAdjuvantTargeting (Moiety)Targeted or Activated CellEffectReferences
i.p.OVA proteinCpGPassiveN/AEnhanced responses compared to soluble vaccine components[87]
i.p.OVA proteinCpG or Poly ICPassiveDC and macrophageEnhanced responses upon combination of TLR ligands[88]
i.d.OVA proteinCpGPassiveN/AEnhanced responses compared to soluble vaccine components[89]
IntratumoralN/AaCD40 and CpGAntibody mediatedIntratumoral macrophage and DCEnhanced responses compared to soluble vaccine components[90]
InhalationN/ACyclic dinucleotidesPSDC and macrophageSynergized anti-tumor responses when combined with irradiation[91]
Hypodermic (not specified)Melanoma peptidesCpGMannoseDCEnhanced responses compared to soluble vaccine components[92]
s.c.OVA peptidePoly IC and Pam3CSK4PassiveN/AEnhanced responses compared to soluble peptide[78]
s.c.OVA proteinCyclic dinucleotidesPassiveDC and macrophageEnhanced responses compared to soluble vaccine adjuvant[93]
s.c.Human papilloma virus peptideCpGMannoseDCEnhanced responses compared to soluble vaccine components[82]
s.c.Melanoma antigen mRNAN/Amannose-mimicking head groupDCProtection from tumor challenge[94]
s.c.ErbB2 proteinPam(3)CAG, Pam(2)CAG and Pam(2)CGDMannoseDCEnhanced responses upon targeting[95]
i.v., s.c., i.m.OVA-encoding mRNAN/APassiveDC and macrophageEnhanced responses upon i.v. immunization[70]
i.v., s.c.OVA peptideSoluble poly IC and aCD40GM3CD169-expressing macrophagesEnhanced immune responses upon i.v. immunization, targeting and encapsulation of antigen[54]
i.v.Human papilloma virus peptideCpGPassivecDC and pDCEnhanced responses compared to soluble vaccine components[96]
i.v.OVA proteinSoluble poly IC and aCD40GM3CD169-expressing macrophagesEnhanced immune responses upon targeting and encapsulation of antigen[83]
i.v.OVA proteinα-GalactosylceramideGM3CD169-expressing macrophagesEnhanced CD8 T cell responses upon targeting[97]

3.5. Adjuvant Encapsulation Augments Its Potency

In addition to antigen, encapsulation of adjuvant in liposomes can have a beneficial effect on the subsequent immune response (Table 2). For example, the TLR 9 agonist, CpG, was found to be more potent when present in the aqueous core of liposomes [87,89]. Indeed, administration of soluble CpG did not yield antitumor responses [92]. This finding was corroborated in a similar experimental setting, where liposomal, but not soluble, CpG could upregulate maturation markers on cDC, elicit IFN-y secretion by T cells, and induce potent antitumor responses [96]. In an alternative approach, Kwong and co-workers did not only encapsulate CpG in their liposomes, but also linked an activating anti CD40-agonistic antibody to the surface of the liposomes. Similar to previous observations, the potency of CpG-containing and anti-CD40-targeted liposomes was enhanced when compared to the soluble components. Indeed, upon immunization with these liposomes tumor outgrowth in mice was significantly delayed [90]. Thus, the potency of adjuvant is markedly increased upon liposomal encapsulation, which may be explained by enhanced delivery to the APCs. Hypothetically, the enhanced activation through liposomal delivery of TLR ligands is explained by the resemblance to the physiological route of activation (triggered by pathogens) as compared to administration of soluble adjuvant [89].
The use of liposomes also allows for incorporation of hydrophobic adjuvant in the bilayer, such as monophosphoryl lipid A (MPLA. Indeed, liposomal delivery of MPLA augmented cross-presentation by DCs, compared to soluble MPLA [98,99]. Furthermore, the use of liposomes also provides the opportunity to incorporate multiple TLR ligands as stimulus. When TLR 4 and TLR 7 were stimulated following combined liposomal delivery, synergistic effects were observed [100]. Similarly, this was also detected when ligands of TLR 3 and TLR 9 were combined in the same liposome, as co-delivery resulted in almost complete protection from a tumor challenge [88].
While the use of TLR ligands in (anti-cancer) vaccines is already well established, various other adjuvants, such as ligands for the cytosolic sensor STING, have been more recently included in liposomes for achieving cellular activation and subsequent anti-tumor responses. The potency of cyclic dinucleotides, which act as second messengers in the STING activation pathway was also found to be enhanced upon delivery in liposomes [93]. Delivery of cyclic dinucleotides encapsulated within the liposomal aqueous phase enhanced its cellular association and protection from tumor rechallenge as compared to free cyclic dinucleotides [101]. Also in the absence of an antigen, delivery of adjuvant-containing nanoparticles may prove beneficial to activate immune cells [102]. When this strategy was combined with irradiation to release tumor antigens, indeed, liposomal-encapsulated cyclic dinucleotides increased the anti-tumor immune response [91].
In summary, the potency of various adjuvants is enhanced when delivered by means of a liposomal carrier (Table 2).

4. Enhanced Delivery of Antigen to APCs Is a Third Component to Improve Cancer Vaccines

4.1. Different APCs for T Cell Priming

DCs are the designated cells to internalize and present antigen to prime naïve T cells. Importantly, cross-priming of CD8+ T cells occurs in secondary lymphoid organs and is dependent on both cellular cross-presentation capacity and the expression of costimulatory molecules [35]. While various cell populations appear to be capable of antigen presentation, cross-presentation by DCs is known to be superior to that by other APCs. However, DCs are a heterogeneous subset of cells with different capacities to (cross-)present antigens. DCs can be broadly divided in type 1 or 2 conventional, or plasmacytoid DCs (cDC1, cDC2, and pDC) [103]. In the mouse, DC1s are marked by XCR1 and Clec9a, whereas cDC2s can be identified by a lack of these markers, the expression of Sirpα and a higher expression of CD11b [103]. In addition, cDC1s in the murine spleen express CD8α, while migratory peripheral tissue cDC1s are marked by CD103 [25]. While both cDC1s and cDC2s are able to internalize and process antigen, cDC1s are most well known for their capacity to cross-present dead cells and to activate CD8+ T cells [104]. cDC1s have a lower proteolytic activity in their endosomal pathway, efficiently transfer peptides to the cytosol, and express relatively high levels of proteins related to MHC class I mediated presentation of antigen, which allows for efficient cross-presentation [28,105,106]. Indeed, in Batf3-/- mice that lack cDC1s, an indispensable role for these cells in the initiation of an anti-tumor response was observed [107]. However, the type of antigen and environmental cues can dictate the extent to which various non-cDC1 DC subsets can cross-present antigen [108]. For example, pDCs, which are mostly well known for their capacity to produce type I IFN [103], gained the ability to cross-present antigen upon TLR stimulation [109,110]. In addition, cDC2s are mostly associated with antigen presentation in the context of MHC class II and activation of CD4+ T cells, but have been shown to be able to cross-present immune complexes and yeast particles [103,111,112]. In addition, recent work has shown that inflammatory cDC2s can also efficiently cross-present antigen [113].
The human counterparts of murine cDC1s and cDC2s are marked by the expression of XCR1, Clec9a, and BDCA3 versus BDCA1 (also known as CD141 and CD1c, respectively) [103]. In contrast to mice, cross-presentation in humans seems to be less restricted to cDC1s [114]. Indeed, human tissue-resident cDCs and pDCs were found to cross-present antigen in steady state. In addition, upon TLR stimulation blood-borne cDC1s and cDC2s were also able to cross-present [114]. Whether or not human DCs are equally potent to cross-prime T cells remains elusive [115]. Interestingly, the expression of Clec9a (DNGR-1) on cDC1s is evolutionarily conserved between mouse and human. DNGR-1 binds to freely accessible actin filaments that are present in dying cells [116]. Since this marker is exclusively expressed by cDC1s, this points to a major role for human cDC1s in the cross-presentation of antigen upon capture of dying cells [117,118,119]. Furthermore, in contrast to other human DC subsets, cDC1s express high levels of TLR 3 and stimulation of these cells with poly(I:C) induces potent maturation [120]. In the mouse, high expression of TLR 3 was determined to be important in the DNGR-1-mediated uptake and response to cell fragments that emerged from virally infected cells [121]. Therefore, the exclusive expression of DNGR-1 and TLR 3 in human cDC1s points toward an important role for these cells in uptake and subsequent cross-presentation of antigen derived from virally infected cells.
Recently, new human cDC subsets have been described that exceed the classical distinction between cDC1s and cDC2s. Especially within the classically defined CD1c-expressing cDC2 subset, more heterogeneity was observed [122]. For example, DC3 was identified as a DC subset that also expresses CD1c and therefore, sole discrimination of DC subsets on CD1c expression resulted in erroneous identification of DC3s as cDC2s. DC3 subset expresses a gene signature that is associated with acute and chronic inflammatory genes [123]. Interestingly, DC3s can be found in human tumors, can secrete high amounts of T cell polarizing cytokines, and are able to induce potent T cell responses [124,125]. In addition, two other subsets were described and defined as DC4 and DC5. Of these subsets, DC4 subset was not extensively described and is not marked by expression of either CD141 or CD1c, but does express certain markers also expressed by monocytes and may be a subset of non-classical monocytes [114,123]. DC5 subset is also referred to as Axl DC (AS DC), as it can be identified based on its expression of Axl, often in combination with Siglec-6 [123]. Since trajectory analysis showed that these cells can develop into other DC subtypes, these cells might be precursors and were also referred to as preDCs [126]. Here, we further refer to these cells as AS DCs. Interestingly, AS DCs can be found in the blood of healthy donors, but also in the blood of donors that suffer from a variety of cancers [127].
To summarize, cDC1s are designated as the major player in cross-priming, especially in mice. However, multiple DCs may play a role in cross-presentation and cross-priming, dependent on environmental cues. The recent identification of novel DC subsets, such as DC3s and AS DCs, opens new avenues in liposomal vaccination studies. Indeed, vaccination strategies may be tailored to optimally make use of the localization of DCs (e.g., blood-borne or tissue residency) as well as the capacity of DC subsets to cross-prime antigen [128].

4.2. Active Targeting Leads to Uptake by APCs of Choice

Although APCs are well known for their capacity to capture antigens, cancer vaccines may be functionalized to enhance the efficiency of uptake by specific types of APCs (Table 2). As mentioned, DCs (and especially cDC1s) may present as ideal targets for cancer vaccines, because of their excellent capacity to cross-present exogenous antigen [103]. In order to target DCs, Boks and co-workers decorated liposomes with Lewis X, which can act as ligand for the C-type lectin dendritic cell-specific intercellular adhesion molecule-3 grabbing non-integrin (DC-SIGN) on DCs and observed that surface coupling of targeting molecules enhanced liposomal binding and uptake by DC-SIGN-expressing target cells [99]. Additionally, mannose or a mannose-mimicking ligand can be used to target the mannose receptor (MR), to enhance liposome delivery to DCs [94]. Thomann and colleagues made use of this targeting strategy and prepared liposomes that contained ligands of TLR 1/2 or TLR 1/6 and a peptide antigen. Interestingly, additional incorporation of mannose to target DCs allowed lowering the dose of adjuvant used 100-fold, while maintaining similar anti-tumor effects [95]. Furthermore, Langerhans cells, APCs that are present in the skin, can be targeted via their expression of the C-type lectin receptor langerin. Modification of liposomes with one of its natural ligands, Lewis Y, or a glycomimetic ligand, resulted in enhanced uptake by Langerhans cells [129,130,131]. As an alternative target, multiple APCs expressed the receptor for phosphatidylserine (PS), which is exposed on dead, but not living, cells. Hence, inclusion of PS in the liposomal membrane can facilitate liposome uptake [91].
Next to the liposomal surface decoration with physiological ligands or a mimetic of these ligands for receptors on DCs, cell type specific antibodies can be conjugated to the surface of liposomes to specifically direct liposomes to the surface of a cell type of interest [132]. An alternative approach is to use the constant part of antibodies to target the Fc receptor on DCs [80,133].
While active targeting of liposomes can allow for tailored target cell delivery, it can also be used to influence subsequent intracellular antigen preservation and routing. Interestingly, antigen uptake via the MR or Fc receptors was shown to lead to endured colocalization of antigen with EEA1-positive early endosomal compartments, in sharp contrast to antigen that was taken up by scavenger receptors, which were quickly routed to lysosomes for degradation [33,34]. In contrast, uptake of antigen via DNGR-1 that is present on cDC1s was found to result in phagosome rupture and therefore efficient transfer of phagocytosed antigen to the cytosol for cross-presentation via the cytosolic route [31]. Thus, targeting of antigen-containing liposomes can facilitate enhanced presentation of the vaccine antigen to the immune system.
While we discussed that cDC1s, especially in mice but also in humans, appear to be highly relevant in cross-presentation of cell-associated antigen, to the best of our knowledge, liposomal targeting studies specifically directed to cDC1 markers, such as XCR1 and Clec9a, are still lacking. On a side note, cancer vaccine targeting to Clec9a was performed with antigen–antibody conjugates and antibody/antigen/poly (lactic-co-glycolic acid) (PLGA)nanoparticles and resulted in robust induction of antigen-specific T cell responses [134,135,136]. In conclusion, active targeting of liposomes by modification of their surface with targeting ligands can markedly increase the potency of vaccination.

4.3. Liposomal Vaccine Targeting to CD169-Expressing Cells in Mice and Men

As an alternative to DC targeting, recent studies investigated targeting to CD169-expressing splenic macrophages. These macrophages are strategically located in the spleen and are known to sequester blood-borne pathogens [137]. CD169-expressing macrophages recognize particulate antigens such as viruses and extracellular vesicles [138,139,140,141]. The CD169 receptor (also known as Siglec-1 or Sialoadhesin [142,143]) binds to α2,3-linked sialic acids that are present on glycoproteins or glycolipids, such as gangliosides [144]. Human immunodeficiency virus (HIV) buds of host lipid rafts are enriched in gangliosides. As a result, the viral envelope is decorated with ganglioside GM3 and indeed, these viruses are taken up by CD169-expressing cells [138,139,140]. In addition, artificial viral nanoparticles, which contained a gold core wrapped in a membrane exposing the physiological ligand for CD169, were taken up by CD169-expressing cells as an example of pathogen mimicry [145,146,147]. Next to the physiological ligand, decoration of liposomes with synthetic ligands for CD169 also resulted in efficient uptake by these cells [55,148]. Our group decorated liposomes with GM3 and detected robust targeting of liposomes to murine splenic CD169-expressing macrophages (Table 2) [54]. Of note, GM3-containing liposomes were minimally taken up by other cell types, for example, red pulp macrophages, that express low levels of CD169 [54]. Interestingly, CD169-expressing macrophages appear to closely collaborate with cDC1s upon liposomal immunization, where CD169-expressing macrophages capture and cDC1s cross-present liposomal antigen. Indeed, the presence of both cell types was essential in our liposomal vaccination strategy [83]. In addition, when liposomes were modified with α-Galactosylceramide (the natural ligand for invariant natural killer T (iNKT) cells), further enhancement of the liposomal efficacy for CD8+ T cell priming was observed [97]. Thus, liposome targeting to CD169-expressing splenic macrophages presents as an interesting strategy, as targeting is specific and upon liposome capture by CD169-expressing macrophages a potent immune response is elicited.
In addition to murine macrophages, human CD169-expressing splenic macrophages also bound liposomes that contained gangliosides [127]. Interestingly, in humans, CD169 is also constitutively expressed by the recently described AS DCs that are present in the blood [123]. As expected, AS DCs could also be targeted with ganglioside-containing liposomes. Upon delivery of antigen and adjuvant in liposomes, AS DCs presented peptide to CD8+ T cells and elicited IFN-y production by antigen-specific T cells [127].
Recently, the murine counterpart of AS DCs was termed transitional DC (tDC). Similar to AS DCs, tDCs were shown to express low levels of CD169 [149]. Since both CD169-expressing macrophages and tDCs are present in the spleen, it remains to be determined whether intravenously injected liposomes are taken up by tDCs next to CD169-expressing macrophages and whether they are involved in T cell priming [54].
Altogether, these studies using both mouse models and human cells demonstrated that targeting to CD169-expressing macrophages and DCs is an interesting approach to increase vaccine efficacy. Liposomes that contain components similar to those present in pathogens, such as gangliosides and nucleotides, can be used to target CD169-expressing APCs (Figure 1 and Table 2).

5. Conclusions and Future Perspectives

Recent studies have indicated that various liposomal characteristics as well as the route of immunization highly impact the subsequent immune responses and should be taken into account to improve liposomal cancer vaccines. Liposomes should be large and cationic in nature to yield depot formation from which antigens are slowly released, while smaller and anionic liposomes prove to be beneficial regarding their capability to enter the regional lymphatics to reach the LN. Recent studies using intravenous administration of liposomes or lipoplexes demonstrated the importance of an anionic nature of the particle (summarized in Table 1). Moreover, they indicated that this rather unconventional route of immunization in vaccination strategies can be more potent as compared to the more conventional immunization routes. While intravenous immunization is not feasible for vaccines that have to be delivered worldwide, such as COVID-19, this route may still be an option for cancer vaccines.
Numerous in vivo studies indicated that the immunogenicity and potency of delivered antigen and adjuvant, respectively, is markedly enhanced upon liposomal delivery. In addition, active targeting of liposomal cancer vaccines to professional APCs using ligands that that are also present in pathogens can further augment the potency of the immune responses (summarized in Table 2).
In our opinion, lessons learned from these preclinical studies in combination with our increasing knowledge of the immune system should be used for vaccine design in future (pre)clinical studies. Profiling of APC subsets revealed a previously underappreciated heterogeneity, in pathogen pattern recognition receptor-expression and also in the uptake of liposomal vaccines. Since liposomal surface decoration with targeting moieties can facilitate uptake by APCs of interest, the choice of vaccine adjuvant should also be based on the pathogen pattern recognition receptor-expression of the targeted APCs. In addition, recent identification of novel DC subsets (e.g., AS DCs) provides novel targeting strategies to be further explored.
In conclusion, the design of an optimal cancer vaccine should be inspired by the composition of pathogens in which antigen, adjuvant, and targeting moieties are present in a single particle, which may enhance the efficacy of liposomal cancer vaccines in the clinic.

Author Contributions

Writing—original draft preparation, M.K.N.T., J.M.M.d.H.; writing—review and editing, L.C., C.F.v.N., G.S.; visualization, M.K.N.T.; funding acquisition, G.S., J.M.M.d.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from NWO ZonMW (TOP 91218024) to J.M.M.d.H. and G.S, grants from the Dutch Cancer Society (VU2016-10449), from the Phospholipid Research Center (JDH-2020-082/1-1), from NWO ENW (ENPPS.KlEM.019.008), from Health Holland TKI-PPP to J.M.M.d.H, and a grant from the NUS School of Medicine Nanomedicine Research Programme (NUHSRO/2021/034/TRP/09/Nanomedicine) to G.S.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Allen, T.M.; Cullis, P.R. Liposomal drug delivery systems: From concept to clinical applications. Adv. Drug Deliv. Rev. 2013, 65, 36–48. [Google Scholar] [CrossRef]
  2. Holohan, C.; Van Schaeybroeck, S.; Longley, D.B.; Johnston, P.G. Cancer drug resistance: An evolving paradigm. Nat. Rev. Cancer 2013, 13, 714–726. [Google Scholar] [CrossRef]
  3. Tran, S.; DeGiovanni, P.; Piel, B.; Rai, P. Cancer nanomedicine: A review of recent success in drug delivery. Clin. Transl. Med. 2017, 6, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Shi, J.; Kantoff, P.W.; Wooster, R.; Farokhzad, J.S.O.C. Cancer nanomedicine: Progress, challenges and opportunities. Nat. Rev. Cancer 2017, 17, 20–37. [Google Scholar] [CrossRef]
  5. Melief, C.J.; van Hall, T.; Arens, R.; Ossendorp, F.; Van Der Burg, S.H. Therapeutic cancer vaccines. J. Clin. Investig. 2015, 125, 3401–3412. [Google Scholar] [CrossRef] [PubMed]
  6. van der Burg, S.H.; Arens, R.; Ossendorp, F.; van Hall, T.; Melief, C.J.M. Vaccines for established cancer: Overcoming the challenges posed by immune evasion. Nat. Rev. Cancer 2016, 16, 219–233. [Google Scholar] [CrossRef]
  7. Schumacher, T.N.; Scheper, W.; Kvistborg, P. Cancer Neoantigens. Annu. Rev. Immunol. 2019, 37, 173–200. [Google Scholar] [CrossRef] [PubMed]
  8. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
  9. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Herbst, R.S.; Soria, J.-C.; Kowanetz, M.; Fine, G.D.; Hamid, O.; Gordon, M.S.; Sosman, J.A.; McDermott, D.F.; Powderly, J.D.; Gettinger, S.N.; et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nat. Cell Biol. 2014, 515, 563–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Sultan, H.; Kumai, T.; Nagato, T.; Wu, J.; Salazar, A.M.; Celis, E. The route of administration dictates the immunogenicity of pep-tide-based cancer vaccines in mice. Cancer Immunol. Immunother. 2019, 68, 455–466. [Google Scholar] [CrossRef]
  12. Hollingsworth, R.E.; Jansen, K. Turning the corner on therapeutic cancer vaccines. NPJ Vaccines 2019, 4, 1–10. [Google Scholar] [CrossRef]
  13. Sahin, U.; Türeci, Ö. Personalized vaccines for cancer immunotherapy. Science 2018, 359, 1355–1360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Gubin, M.M.; Zhang, X.; Schuster, H.; Caron, E.; Ward, J.P.; Noguchi, T.; Ivanova, Y.; Hundal, J.; Arthur, C.D.; Krebber, W.J.; et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 2014, 515, 577–581. [Google Scholar] [CrossRef]
  15. Kreiter, S.; Castle, J.C.; Türeci, Ö.; Sahin, U. Targeting the tumor mutanome for personalized vaccination therapy. OncoImmunology 2012, 1, 768–769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. van Hall, T.; van der Burg, S.H. Mechanisms of peptide vaccination in mouse models: Tolerance, immunity, and hyperreactivity. Adv. Immunol. 2012, 114, 51–76. [Google Scholar] [PubMed]
  17. Polack, F.P.; Thomas, S.J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J.L.; Marc, G.P.; Moreira, E.D.; Zerbini, C.; et al. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N. Engl. J. Med. 2020, 383, 2603–2615. [Google Scholar] [CrossRef] [PubMed]
  18. Sahin, U.; Karikó, K.; Türeci, Ö. mRNA-based therapeutics—Developing a new class of drugs. Nat. Rev. Drug Discov. 2014, 13, 759–780. [Google Scholar] [CrossRef]
  19. Grabbe, S.; Haas, H.; Diken, M.; Kranz, L.M.; Langguth, P.; Sahin, U. Translating nanoparticulate-personalized cancer vaccines into clinical applications: Case study with RNA-lipoplexes for the treatment of melanoma. Nanomedicine 2016, 11, 2723–2734. [Google Scholar] [CrossRef] [PubMed]
  20. Kranz, L.M.; Diken, M.; Haas, H.; Kreiter, S.; Loquai, C.; Reuter, K.C.; Meng, M.; Fritz, D.; Vascotto, F.; Hefesha, H.; et al. Systemic RNA delivery to dendritic cells exploits antivi-ral defence for cancer immunotherapy. Nature 2016, 534, 396–401. [Google Scholar] [CrossRef]
  21. Sahin, U.; Oehm, P.; Derhovanessian, E.; Jabulowsky, R.A.; Vormehr, M.; Gold, M.; Maurus, D.; Schwarck-Kokarakis, D.; Kuhn, A.N.; Omokoko, T.; et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature 2020, 585, 107–112. [Google Scholar] [CrossRef] [PubMed]
  22. Salomon, N.; Vascotto, F.; Selmi, A.; Vormehr, M.; Quinkhardt, J.; Bukur, T.; Schrörs, B.; Löewer, M.; Diken, M.; Türeci, Ö.; et al. A liposomal RNA vaccine inducing neoanti-gen-specific CD4(+) T cells augments the antitumor activity of local radiotherapy in mice. Oncoimmunology 2020, 9, 1771925. [Google Scholar] [CrossRef] [PubMed]
  23. Selmi, A.; Vascotto, F.; Kautz-Neu, K.; Türeci, Ö.; Sahin, U.; Von Stebut, E.; Diken, M.; Kreiter, S. Uptake of synthetic naked RNA by skin-resident dendritic cells via macropinocytosis allows antigen expression and induction of T-cell responses in mice. Cancer Immunol. Immunother. 2016, 65, 1075–1083. [Google Scholar] [CrossRef] [PubMed]
  24. Kreiter, S.; Selmi, A.; Diken, M.; Koslowski, M.; Britten, C.M.; Huber, C.; Türeci, Ö.; Sahin, U. Intranodal Vaccination with Naked Antigen-Encoding RNA Elicits Potent Prophylactic and Therapeutic Antitumoral Immunity. Cancer Res. 2010, 70, 9031–9040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Böttcher, J.P.; e Sousa, C.R. The Role of Type 1 Conventional Dendritic Cells in Cancer Immunity. Trends Cancer 2018, 4, 784–792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Joffre, O.; Segura, E.; Savina, A.; Amigorena, S. Cross-presentation by dendritic cells. Nat. Rev. Immunol. 2012, 12, 557–569. [Google Scholar] [CrossRef]
  27. Jhunjhunwala, S.; Hammer, C.; Delamarre, L. Antigen presentation in cancer: Insights into tumour immunogenicity and immune evasion. Nat. Rev. Cancer 2021, 21, 298–312. [Google Scholar] [CrossRef]
  28. Dudziak, D.; Kamphorst, A.O.; Heidkamp, G.F.; Buchholz, V.R.; Trumpfheller, C.; Yamazaki, S.; Cheong, C.; Liu, K.; Lee, H.-W.; Park, C.G.; et al. Differential Antigen Processing by Dendritic Cell Subsets in Vivo. Science 2007, 315, 107–111. [Google Scholar] [CrossRef]
  29. Grotzke, J.E.; Sengupta, D.; Lu, Q.; Cresswell, P. The ongoing saga of the mechanism(s) of MHC class I-restricted cross-presentation. Curr. Opin. Immunol. 2017, 46, 89–96. [Google Scholar] [CrossRef]
  30. Colbert, J.D.; Cruz, F.M.; Rock, K.L. Cross-presentation of exogenous antigens on MHC I molecules. Curr. Opin. Immunol. 2020, 64, 1–8. [Google Scholar] [CrossRef]
  31. Canton, J.; Blees, H.; Henry, C.M.; Buck, M.D.; Schulz, O.; Rogers, N.C.; Childs, E.; Zelenay, S.; Rhys, H.; Domart, M.C.; et al. The receptor DNGR-1 signals for phagosomal rupture to promote cross-presentation of dead-cell-associated antigens. Nat. Immunol. 2021, 22, 140–153. [Google Scholar] [CrossRef] [PubMed]
  32. Tang-Huau, T.-L.; Gueguen, P.; Goudot, C.; Durand, M.; Bohec, M.; Baulande, S.; Pasquier, B.; Amigorena, S.; Segura, E. Human in vivo-generated mono-cytederived dendritic cells and macrophages cross-present antigens through a vacuolar pathway. Nat. Commun. 2018, 9, 2570. [Google Scholar] [CrossRef]
  33. Burgdorf, S.; Kautz, A.; Böhnert, V.; Knolle, P.A.; Kurts, C. Distinct Pathways of Antigen Uptake and Intracellular Routing in CD4 and CD8 T Cell Activation. Science 2007, 316, 612–616. [Google Scholar] [CrossRef]
  34. van Montfoort, N.; Camps, M.G.; Khan, S.; Filippov, D.V.; Weterings, J.J.; Griffith, J.M.; Geuze, H.J.; van Hall, T.; Verbeek, J.S.; Melief, C.J.; et al. Antigen storage compartments in ma-ture dendritic cells facilitate prolonged cytotoxic T lymphocyte cross-priming capacity. Proc. Natl. Acad. Sci. USA 2009, 106, 6730–6735. [Google Scholar] [CrossRef] [Green Version]
  35. Gardner, A.; Ruffell, B. Dendritic Cells and Cancer Immunity. Trends Immunol. 2016, 37, 855–865. [Google Scholar] [CrossRef] [Green Version]
  36. Akira, S.; Uematsu, S.; Takeuchi, O. Pathogen recognition and innate immunity. Cell 2006, 124, 783–801. [Google Scholar] [CrossRef] [Green Version]
  37. Kawai, T.; Akira, S. The role of pattern-recognition receptors in innate immunity: Update on Toll-like receptors. Nat. Immunol. 2010, 11, 373–384. [Google Scholar] [CrossRef]
  38. Schlitzer, A.; McGovern, N.; Ginhoux, F. Dendritic cells and monocyte-derived cells: Two complementary and integrated functional systems. Semin. Cell Dev. Biol. 2015, 41, 9–22. [Google Scholar] [CrossRef] [PubMed]
  39. Chen, Q.; Sun, L.; Chen, Z. Regulation and function of the cGAS–STING pathway of cytosolic DNA sensing. Nat. Immunol. 2016, 17, 1142–1149. [Google Scholar] [CrossRef] [PubMed]
  40. Corrales, L.; Glickman, L.H.; McWhirter, S.M.; Kanne, D.B.; Sivick, K.E.; Katibah, G.E.; Woo, S.-R.; Lemmens, E.; Banda, T.; Leong, J.J.; et al. Direct Activation of STING in the Tumor Microenvironment Leads to Potent and Systemic Tumor Regression and Immunity. Cell Rep. 2015, 11, 1018–1030. [Google Scholar] [CrossRef] [Green Version]
  41. Goubau, D.; Schlee, M.; Deddouche, S.; Pruijssers, A.J.; Zillinger, T.; Goldeck, M.; Schuberth, C.; Van Der Veen, A.G.; Fujimura, T.; Rehwinkel, J.; et al. Antiviral immunity via RIG-I-mediated recognition of RNA bearing 5′-diphosphates. Nat. Cell Biol. 2014, 514, 372–375. [Google Scholar] [CrossRef] [Green Version]
  42. Nair-Gupta, P.; Baccarini, A.; Tung, N.; Seyffer, F.; Florey, O.; Huang, Y.; Banerjee, M.; Overholtzer, M.; Roche, P.; Tampé, R.; et al. TLR Signals Induce Phagosomal MHC-I Delivery from the Endosomal Recycling Compartment to Allow Cross-Presentation. Cell 2014, 158, 506–521. [Google Scholar] [CrossRef] [Green Version]
  43. Weimershaus, M.; Mauvais, F.-X.; Saveanu, L.; Adiko, C.; Babdor, J.; Abramova, A.; Montealegre, S.; Lawand, M.; Evnouchidou, I.; Huber, K.J.; et al. Innate Immune Signals Induce Antero-grade Endosome Transport Promoting MHC Class I Cross-Presentation. Cell Rep. 2018, 24, 3568–3581. [Google Scholar] [CrossRef] [Green Version]
  44. Pauwels, A.-M.; Härtlova, A.; Peltier, J.; Driege, Y.; Baudelet, G.; Brodin, P.; Beyaert, R.; Hoffmann, E. Spatiotemporal Changes of the Phagosomal Pro-teome in Dendritic Cells in Response to LPS Stimulation. Mol. Cell Proteom. 2019, 18, 909–922. [Google Scholar] [CrossRef] [PubMed]
  45. Burgdorf, S.; Schölz, C.; Kautz, A.; Tampé, R.; Kurts, C. Spatial and mechanistic separation of cross-presentation and endogenous antigen presentation. Nat. Immunol. 2008, 9, 558–566. [Google Scholar] [CrossRef]
  46. Anderluzzi, G.; Schmidt, S.T.; Cunliffe, R.; Woods, S.; Roberts, C.W.; Veggi, D.; Ferlenghi, I.; O’Hagan, D.T.; Baudner, B.C.; Perrie, Y. Rational design of adjuvants for subunit vac-cines: The format of cationic adjuvants affects the induction of antigen-specific antibody responses. J. Control. Release 2021, 330, 933–944. [Google Scholar] [CrossRef]
  47. Khadke, S.; Roces, C.B.; Cameron, A.; Devitt, A.; Perrie, Y. Formulation and manufacturing of lymphatic targeting liposomes us-ing microfluidics. J. Control. Release 2019, 307, 211–220. [Google Scholar] [CrossRef] [PubMed]
  48. Henriksen-Lacey, M.; Christensen, D.; Bramwell, V.W.; Lindenstrøm, T.; Agger, E.M.; Andersen, P.; Perrie, Y. Liposomal cationic charge and antigen adsorption are important properties for the efficient deposition of antigen at the injection site and ability of the vaccine to induce a CMI response. J. Control. Release 2010, 145, 102–108. [Google Scholar] [CrossRef] [PubMed]
  49. Christensen, D.; Henriksen-Lacey, M.; Kamath, A.T.; Lindenstrøm, T.; Korsholm, K.S.; Christensen, J.P.; Rochat, A.F.; Lambert, P.H.; Andersen, P.; Siegrist, C.A.; et al. A cationic vaccine ad-juvant based on a saturated quaternary ammonium lipid have different in vivo distribution kinetics and display a distinct CD4 T cell-inducing capacity compared to its unsaturated analog. J. Control. Release 2012, 160, 468–476. [Google Scholar] [CrossRef] [PubMed]
  50. Kaur, R.; Henriksen-Lacey, M.; Wilkhu, J.; Devitt, A.; Christensen, D.; Perrie, Y. Effect of Incorporating Cholesterol into DDA:TDB Liposomal Adjuvants on Bilayer Properties, Biodistribution, and Immune Responses. Mol. Pharm. 2014, 11, 197–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Kaur, R.; Bramwell, V.W.; Kirby, D.J.; Perrie, Y. Pegylation of DDA:TDB liposomal adjuvants reduces the vaccine depot effect and alters the Th1/Th2 immune responses. J. Control. Release 2012, 158, 72–77. [Google Scholar] [CrossRef] [PubMed]
  52. Zhuang, Y.; Ma, Y.; Wang, C.; Hai, L.; Yan, C.; Zhang, Y.; Liu, F.; Cai, L. PEGylated cationic liposomes robustly augment vaccine-induced immune responses: Role of lymphatic trafficking and biodistribution. J. Control. Release 2012, 159, 135–142. [Google Scholar] [CrossRef] [PubMed]
  53. Schmidt, S.T.; Olsen, C.L.; Franzyk, H.; Wørzner, K.; Korsholm, K.S.; Rades, T.; Andersen, P.; Foget, C.; Christensen, D. Comparison of two different PEGylation strate-gies for the liposomal adjuvant CAF09: Towards induction of CTL responses upon subcutaneous vaccine administration. Eur. J. Pharm. Biopharm. 2019, 140, 29–39. [Google Scholar] [CrossRef]
  54. Nijen Twilhaar, M.K.; Czentner, L.; Grabowska, J.; Affandi, A.J.; Lau, C.Y.J.; Olesek, K.; Kalay, K.; van Nostrum, C.F.; van Kooyk, Y.; Storm, G.; et al. Optimization of Liposomes for Antigen Targeting to Splenic CD169(+) Macrophages. Pharmaceutics 2020, 12, 1138. [Google Scholar] [CrossRef] [PubMed]
  55. Edgar, L.J.; Kawasaki, N.; Nycholat, C.M.; Paulson, J.C. Targeted Delivery of Antigen to Activated CD169+ Macrophages Induces Bias for Expansion of CD8+ T Cells. Cell Chem. Biol. 2019, 26, 131–136.e4. [Google Scholar] [CrossRef]
  56. Mohamed, M.; Abu Lila, A.S.; Shimizu, T.; Alaaeldin, E.; Hussein, A.; Sarhan, H.A.; Szebeni, J.; Ishida, T. PEGylated liposomes: Immunological responses. Sci. Technol. Adv. Mater. 2019, 20, 710–724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Oussoren, C.; Storm, G. Liposomes to target the lymphatics by subcutaneous administration. Adv. Drug Deliv. Rev. 2001, 50, 143–156. [Google Scholar] [CrossRef]
  58. Carstens, M.G.; Camps, M.G.; Henriksen-Lacey, M.; Franken, K.; Ottenhoff, T.H.; Perrie, Y.; Bouwstra, J.A.; Ossendorp, F.; Jiskoot, W. Effect of vesicle size on tissue localization and immunogenicity of liposomal DNA vaccines. Vaccine 2011, 29, 4761–4770. [Google Scholar] [CrossRef]
  59. Zhang, R.; Billingsley, M.; Mitchell, M.J. Biomaterials for vaccine-based cancer immunotherapy. J. Control. Release 2018, 292, 256–276. [Google Scholar] [CrossRef]
  60. Verbeke, R.; Lentacker, I.; Wayteck, L.; Breckpot, K.; Van Bockstal, M.; Descamps, B.; Vanhove, C.; De Smedt, S.C.; Dewitte, H. Co-delivery of nucleoside-modified mRNA and TLR agonists for cancer immunotherapy: Restoring the immunogenicity of immunosilent mRNA. J. Control. Release 2017, 266, 287–300. [Google Scholar] [CrossRef]
  61. Petros, R.A.; DeSimone, J.M. Strategies in the design of nanoparticles for therapeutic applications. Nat. Rev. Drug Discov. 2010, 9, 615–627. [Google Scholar] [CrossRef]
  62. Moghimi, S.M.; Hunter, A.C.; Andresen, T.L. Factors Controlling Nanoparticle Pharmacokinetics: An Integrated Analysis and Perspective. Annu. Rev. Pharmacol. Toxicol. 2012, 52, 481–503. [Google Scholar] [CrossRef] [PubMed]
  63. Sahin, U.; Derhovanessian, E.; Miller, M.; Kloke, B.-P.; Simon, P.; Löwer, M.; Bukur, V.; Tadmor, A.D.; Luxemburger, U.; Schrörs, B.; et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 2017, 547, 222–226. [Google Scholar] [CrossRef] [PubMed]
  64. Rosigkeit, S.; Meng, M.; Grunwitz, C.; Gomes, P.; Kreft, A.; Hayduk, N.; Heck, R.; Pickert, G.; Ziegler, K.; Abassi, Y.; et al. Monitoring Translation Activity of mRNA-Loaded Nanoparticles in Mice. Mol. Pharm. 2018, 15, 3909–3919. [Google Scholar] [CrossRef] [PubMed]
  65. Perrie, Y.; Crofts, F.; Devitt, A.; Griffiths, H.R.; Kastner, E.; Nadella, V. Designing liposomal adjuvants for the next generation of vaccines. Adv. Drug Deliv. Rev. 2016, 99, 85–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Hattori, Y.; Nakamura, M.; Takeuchi, N.; Tamaki, K.; Shimizu, S.; Yoshiike, Y.; Taguchi, M.; Ohno, H.; Ozaki, K.-I.; Onishi, H. Effect of cationic lipid in cationic liposomes on siRNA delivery into the lung by intravenous injection of cationic lipoplex. J. Drug Target. 2019, 27, 217–227. [Google Scholar] [CrossRef] [PubMed]
  67. Heuts, J.; Jiskoot, W.; Ossendorp, F.; van der Maaden, K. Cationic Nanoparticle-Based Cancer Vaccines. Pharmaceutics 2021, 13, 596. [Google Scholar] [CrossRef] [PubMed]
  68. Varypataki, E.M.; Benne, N.; Bouwstra, J.; Jiskoot, W.; Ossendorp, F. Efficient Eradication of Established Tumors in Mice with Cationic Liposome-Based Synthetic Long-Peptide Vaccines. Cancer Immunol. Res. 2017, 5, 222–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. De Beuckelaer, A.; Pollard, C.; Van Lint, S.; Roose, K.; Van Hoecke, L.; Naessens, T.; Udhayakumar, V.K.; Smet, M.; Sanders, N.; Lienenklaus, S.; et al. Type I Interferons Interfere with the Ca-pacity of mRNA Lipoplex Vaccines to Elicit Cytolytic T Cell Responses. Mol. Ther. 2016, 24, 2012–2020. [Google Scholar] [CrossRef] [Green Version]
  70. Broos, K.; Van der Jeught, K.; Puttemans, J.; Goyvaerts, C.; Heirman, C.; Dewitte, H.; Verbeke, R.; Lentacker, I.; Thielemans, K.; Breckpot, K. Particle-mediated Intravenous Delivery of Antigen mRNA Results in Strong Antigen-specific T-cell Responses Despite the Induction of Type I Interferon. Mol. Ther. Nucleic Acids 2016, 5, e326. [Google Scholar] [CrossRef] [Green Version]
  71. Sayour, E.J.; De Leon, G.; Pham, C.; Grippin, A.; Kemeny, H.; Chua, J.; Huang, J.; Sampson, J.H.; Sanchez-Perez, L.; Flores, C.; et al. Systemic activation of antigen-presenting cells via RNA-loaded nanoparticles. OncoImmunology 2016, 6, e1256527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Fuertes, M.B.; Kacha, A.K.; Kline, J.; Woo, S.-R.; Kranz, D.M.; Murphy, K.M.; Gajewski, T.F. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8{alpha}+ dendritic cells. J. Exp. Med. 2011, 208, 2005–2016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Diamond, M.S.; Kinder, M.; Matsushita, H.; Mashayekhi, M.; Dunn, G.P.; Archambault, J.M.; Lee, H.; Arthur, C.D.; White, J.M.; Kalinke, U.; et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J. Exp. Med. 2011, 208, 1989–2003. [Google Scholar] [CrossRef] [PubMed]
  74. Van Hoecke, L.; Roose, K.; Ballegeer, M.; Zhong, Z.; Sanders, N.N.; De Koker, S.; Saelens, X.; van Lint, S. The Opposing Effect of Type I IFN on the T Cell Response by Non-modified mRNA-Lipoplex Vaccines Is Determined by the Route of Administration. Mol. Ther. Nucleic Acids 2020, 22, 373–381. [Google Scholar] [CrossRef]
  75. Limmer, A.; Ohl, J.; Kurts, C.; Ljunggren, H.-G.; Reiss, Y.; Groettrup, M.; Momburg, F.; Arnold, B.; Knolle, P.A. Efficient presentation of exogenous antigen by liver endothelial cells to CD8+ T cells results in antigen-specific T-cell tolerance. Nat. Med. 2000, 6, 1348–1354. [Google Scholar] [CrossRef] [Green Version]
  76. Hirosue, S.; Vokali, E.; Raghavan, V.R.; Rincon-Restrepo, M.; Lund, A.W.; Corthésy-Henrioud, P.; Capotosti, F.; Halin Winter, C.; Hugues, S.; Swartz, M.A. Steady-state antigen scav-enging, cross-presentation, and CD8+ T cell priming: A new role for lymphatic endothelial cells. J. Immunol. 2014, 192, 5002–5011. [Google Scholar] [CrossRef] [Green Version]
  77. Henriksen-Lacey, M.; Bramwell, V.W.; Christensen, D.; Agger, E.-M.; Andersen, P.; Perrie, Y. Liposomes based on dimethyldiocta-decylammonium promote a depot effect and enhance immunogenicity of soluble antigen. J. Control. Release 2010, 142, 180–186. [Google Scholar] [CrossRef]
  78. Varypataki, E.M.; Silva, A.L.; Barnier-Quer, C.; Collin, N.; Ossendorp, F.; Jiskoot, W. Synthetic long peptide-based vaccine formula-tions for induction of cell mediated immunity: A comparative study of cationic liposomes and PLGA nanoparticles. J. Control. Release 2016, 226, 98–106. [Google Scholar] [CrossRef]
  79. Heuts, J.; Varypataki, E.M.; van der Maaden, K.; Romeijn, S.; Drijfhout, J.W.; van Scheltinga, A.T.; Ossendorp, F.; Jiskoot, W. Cationic Liposomes: A Flexible Vaccine Delivery System for Physicochemically Diverse Antigenic Peptides. Pharm. Res. 2018, 35, 207. [Google Scholar] [CrossRef] [Green Version]
  80. Cruz, L.J.; Rueda, F.; Cordobilla, B.; Simón, L.; Hosta, L.; Albericio, F.; Domingo, J.C.; Rigau, L.H. Targeting Nanosystems to Human DCs via Fc Receptor as an Effective Strategy to Deliver Antigen for Immunotherapy. Mol. Pharm. 2010, 8, 104–116. [Google Scholar] [CrossRef]
  81. Bae, J.; Parayath, N.; Ma, W.; Amiji, M.; Munshi, N.; Anderson, K.C. BCMA peptide-engineered nanoparticles enhance induction and function of antigen-specific CD8(+) cytotoxic T lymphocytes against multiple myeloma: Clinical applications. Leukemia 2020, 34, 210–223. [Google Scholar] [CrossRef]
  82. Zhao, Y.; Wang, H.; Yang, Y.; Jia, W.; Su, T.; Che, Y.; Feng, Y.; Yuan, X.; Wang, X. Mannose-Modified Liposome Co-Delivery of Human Papillomavirus Type 16 E7 Peptide and CpG Oligodeoxynucleotide Adjuvant Enhances Antitumor Activity Against Established Large TC-1 Grafted Tumors in Mice. Int. J. Nanomed. 2020, 15, 9571–9586. [Google Scholar] [CrossRef] [PubMed]
  83. Grabowska, J.; Affandi, A.; van Dinther, D.; Twilhaar, M.N.; Olesek, K.; Hoogterp, L.; Ambrosini, M.; Heijnen, D.; Klaase, L.; Hidalgo, A.; et al. Liposome induction of CD8+ T cell responses depends on CD169+ macrophages and Batf3-dependent dendritic cells and is enhanced by GM3 inclusion. J. Control. Release 2021, 331, 309–320. [Google Scholar] [CrossRef] [PubMed]
  84. Aarntzen, E.H.J.G.; De Vries, I.J.M.; Lesterhuis, W.J.; Schuurhuis, D.; Jacobs, J.F.M.; Bol, K.; Schreibelt, G.; Mus, R.; De Wilt, J.H.W.; Haanen, J.B.A.G.; et al. Targeting CD4+ T-Helper Cells Improves the Induction of Antitumor Responses in Dendritic Cell–Based Vaccination. Cancer Res. 2013, 73, 19–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Guan, C.; Chernyak, N.; Dominguez, D.; Cole, L.; Zhang, B.; Mirkin, C.A. RNA-Based Immunostimulatory Liposomal Spherical Nucleic Acids as Potent TLR7/8 Modulators. Small 2018, 14, e1803284. [Google Scholar] [CrossRef]
  86. Persano, S.; Guevara, M.L.; Li, Z.; Mai, J.; Ferrari, M.; Pompa, P.P.; Shen, H. Lipopolyplex potentiates anti-tumor immunity of mRNA-based vaccination. Biomaterials 2017, 125, 81–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Erikçi, E.; Gursel, M.; Gürsel, İ. Differential immune activation following encapsulation of immunostimulatory CpG oligodeox-ynucleotide in nanoliposomes. Biomaterials 2011, 32, 1715–1723. [Google Scholar] [CrossRef]
  88. Bayyurt, B.; Tincer, G.; Almacioglu, K.; Alpdundar, E.; Gursel, M.; Gursel, I. Encapsulation of two different TLR ligands into lipo-somes confer protective immunity and prevent tumor development. Journal Control. Release 2017, 247, 134–144. [Google Scholar] [CrossRef]
  89. Bal, S.; Hortensius, S.; Ding, Z.; Jiskoot, W.; Bouwstra, J.A. Co-encapsulation of antigen and Toll-like receptor ligand in cationic liposomes affects the quality of the immune response in mice after intradermal vaccination. Vaccine 2011, 29, 1045–1052. [Google Scholar] [CrossRef] [Green Version]
  90. Kwong, B.; Liu, H.; Irvine, D.J. Induction of potent anti-tumor responses while eliminating systemic side effects via liposomeanchored combinatorial immunotherapy. Biomaterials 2011, 32, 5134–5147. [Google Scholar] [CrossRef] [Green Version]
  91. Liu, Y.; Crowe, W.N.; Wang, L.; Lu, Y.; Petty, W.J.; Habib, A.A.; Zhao, D. An inhalable nanoparticulate STING agonist synergizes with radiotherapy to confer long-term control of lung metastases. Nat. Commun. 2019, 10, 1–15. [Google Scholar] [CrossRef] [Green Version]
  92. Lai, C.; Duan, S.; Ye, F.; Hou, X.; Li, X.; Zhao, J.; Yu, X.; Hu, Z.; Tang, Z.; Mo, F.; et al. The enhanced antitumor-specific immune response with mannose- and CpG-ODN-coated liposomes delivering TRP2 peptide. Theranostics 2018, 8, 1723–1739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Hanson, M.C.; Crespo, M.P.; Abraham, W.; Moynihan, K.D.; Szeto, G.L.; Chen, S.H.; Melo, M.B.; Mueller, S.; Irvine, D.J. Nanoparticulate STING agonists are po-tent lymph node-targeted vaccine adjuvants. J. Clin. Investig. 2015, 125, 2532–2546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Garu, A.; Moku, G.; Gulla, S.K.; Chaudhuri, A. Genetic Immunization With In Vivo Dendritic Cell-targeting Liposomal DNA Vaccine Carrier Induces Long-lasting Antitumor Immune Response. Mol. Ther. 2016, 24, 385–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Thomann, J.-S.; Heurtault, B.; Weidner, S.; Brayé, M.; Beyrath, J.; Fournel, S.; Schuber, F.; Frisch, B. Antitumor activity of liposomal ErbB2/HER2 epitope peptide-based vaccine constructs incorporating TLR agonists and mannose receptor targeting. Biomaterials 2011, 32, 4574–4583. [Google Scholar] [CrossRef]
  96. Shen, K.-Y.; Liu, H.-Y.; Yan, W.-L.; Wu, C.-C.; Lee, M.-H.; Leng, C.-H.; Liu, S.J. Liposomal TLR9 Agonist Combined with TLR2 Ago-nist-Fused Antigen Can Modulate Tumor Microenvironment through Dendritic Cells. Cancers 2020, 12, 810. [Google Scholar] [CrossRef] [Green Version]
  97. Grabowska, J.; Stolk, D.A.; Nijen Twilhaar, M.K.; Ambrosini, M.; Storm, G.; van der Vliet, H.J.; de Gruijl, T.D.; van Kooyk, Y.; den Haan, J.M.M. Liposomal Nanovaccine Con-taining α-Galactosylceramide and Ganglioside GM3 Stimulates Robust CD8(+) T Cell Responses via CD169(+) Macrophages and Cdc1. Vaccines 2021, 9, 56. [Google Scholar] [CrossRef]
  98. Boks, M.A.; Bruijns, S.C.M.; Ambrosini, M.; Kalay, H.; van Bloois, L.; Storm, G.; de Gruil, T.D.; van Kooyk, Y. In situ Delivery of Tumor Antigen- and Adju-vant-Loaded Liposomes Boosts Antigen-Specific T-Cell Responses by Human Dermal Dendritic Cells. J. Investig. Dermatol. 2015, 135, 2697–2704. [Google Scholar] [CrossRef] [Green Version]
  99. Boks, M.A.; Ambrosini, M.; Bruijns, S.C.; Kalay, H.; van Bloois, L.; Storm, G.; Garcia-Vallejo, J.J.; van Kooyk, Y. MPLA incorporation into DC-targeting glycoliposomes favours anti-tumour T cell responses. J. Control. Release 2015, 216, 37–46. [Google Scholar] [CrossRef] [PubMed]
  100. Fox, C.B.; Sivananthan, S.J.; Duthie, M.S.; Vergara, J.; Guderian, J.A.; Moon, E.; Coblentz, D.; Reed, S.G.; Carter, D. A nanoliposome delivery system to synergisti-cally trigger TLR4 AND TLR7. J. Nanobiotechnol. 2014, 12, 17. [Google Scholar] [CrossRef] [Green Version]
  101. Koshy, S.T.; Cheung, A.S.; Gu, L.; Graveline, A.R.; Mooney, D.J. Liposomal Delivery Enhances Immune Activation by STING Ago-nists for Cancer Immunotherapy. Adv. Biosyst. 2017, 1, 1600013. [Google Scholar] [CrossRef]
  102. Nakamura, T.; Miyabe, H.; Hyodo, M.; Sato, Y.; Hayakawa, Y.; Harashima, H. Liposomes loaded with a STING pathway ligand, cyclic di-GMP, enhance cancer immunotherapy against metastatic melanoma. J. Control. Release 2015, 216, 149–157. [Google Scholar] [CrossRef] [Green Version]
  103. Guilliams, M.; Ginhoux, F.; Jakubzick, C.; Naik, S.H.; Onai, N.; Schraml, B.U.; Segura, E.; Tussiwand, R.; Yona, S. Dendritic cells, monocytes and macrophages: A unified nomenclature based on ontogeny. Nat. Rev. Immunol. 2014, 14, 571–578. [Google Scholar] [CrossRef]
  104. den Haan, J.M.; Lehar, S.M.; Bevan, M.J. CD8(+) but not CD8(−) dendritic cells cross-prime cytotoxic T cells in vivo. J. Exp. Med. 2000, 192, 1685–1696. [Google Scholar] [CrossRef]
  105. Savina, A.; Peres, A.; Cebrian, I.; Carmo, N.; Moita, C.; Hacohen, N.; Moita, L.F.; Amigorena, S. The small GTPase Rac2 controls phagosomal alkaliniza-tion and antigen crosspresentation selectively in CD8(+) dendritic cells. Immunity 2009, 30, 544–555. [Google Scholar] [CrossRef] [Green Version]
  106. Lin, M.L.; Zhan, Y.; Proietto, A.I.; Prato, S.; Wu, L.; Heath, W.R.; Villadangos, J.A.; Lew, A.M. Selective suicide of cross-presenting CD8+ dendritic cells by cytochrome c injection shows functional heterogeneity within this subset. Proc. Natl. Acad. Sci. USA 2008, 105, 3029–3034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Hildner, K.; Edelson, B.T.; Purtha, W.E.; Diamond, M.; Matsushita, H.; Kohyama, M.; Calderon, B.; Schraml, B.U.; Unanue, E.R.; Diamond, M.S.; et al. Batf3 Deficiency Reveals a Critical Role for CD8α+ Dendritic Cells in Cytotoxic T Cell Immunity. Science 2008, 322, 1097–1100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Nierkens, S.; Tel, J.; Janssen, E.; Adema, G.J. Antigen cross-presentation by dendritic cell subsets: One general or all sergeants? Trends Immunol. 2013, 34, 361–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Oberkampf, M.; Guillerey, C.; Mouriès, J.; Rosenbaum, P.; Fayolle, C.; Bobard, A.; Savina, A.; Ogier-Denis, E.; Enninga, J.; Amigorena, S.; et al. Mitochondrial reactive oxygen species reg-ulate the induction of CD8(+) T cells by plasmacytoid dendritic cells. Nat. Commun. 2018, 9, 2241. [Google Scholar] [CrossRef] [Green Version]
  110. Mouries, J.; Moron, G.; Schlecht-Louf, G.; Escriou, N.; Dadaglio, G.; Leclerc, C. Plasmacytoid dendritic cells efficiently cross-prime naive T cells in vivo after TLR activation. Blood 2008, 112, 3713–3722. [Google Scholar] [CrossRef]
  111. Backer, R.; Van Leeuwen, F.; Kraal, G.; Haan, J.M.M.D. CD8– dendritic cells preferentially cross-presentSaccharomyces cerevisiae antigens. Eur. J. Immunol. 2008, 38, 370–380. [Google Scholar] [CrossRef]
  112. den Haan, J.M.M.; Bevan, M.J. Constitutive versus activation-dependent cross-presentation of immune complexes by CD8(+) and CD8(-) dendritic cells in vivo. J. Exp. Med. 2002, 196, 817–827. [Google Scholar] [CrossRef] [PubMed]
  113. Bosteels, C.; Neyt, K.; Vanheerswynghels, M.; van Helden, M.J.; Sichien, D.; Debeuf, N.; De Prijck, S.; Bosteels, V.; Vandamme, N.; Martens, L.; et al. Inflammatory Type 2 cDCs Acquire Features of cDC1s and Macrophages to Orchestrate Immunity to Respiratory Virus Infection. Immunity 2020, 52, 1039–1056.e9. [Google Scholar] [CrossRef] [PubMed]
  114. Collin, M.; Bigley, V. Human dendritic cell subsets: An update. Immunology 2018, 154, 3–20. [Google Scholar] [CrossRef] [PubMed]
  115. Segura, E.; Amigorena, S. Chapter One—Cross-Presentation in Mouse and Human Dendritic Cells. In Advances in Immunology; Alt, F.W., Ed.; Academic Press: Cambridge, MA, USA, 2015; pp. 1–31. [Google Scholar]
  116. Ahrens, S.; Zelenay, S.; Sancho, D.; Hanč, P.; Kjær, S.; Feest, C.; Fletcher, G.; Durkin, C.; Postigo, A.; Skehel, M.; et al. F-Actin Is an Evolutionarily Conserved Damage-Associated Molecular Pattern Recognized by DNGR-1, a Receptor for Dead Cells. Immunity 2012, 36, 635–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Jongbloed, S.L.; Kassianos, A.; McDonald, K.J.; Clark, G.; Ju, X.; Angel, C.; Chen, C.-J.J.; Dunbar, P.R.; Wadley, R.B.; Jeet, V.; et al. Human CD141+ (BDCA-3)+ dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens. J. Exp. Med. 2010, 207, 1247–1260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Bachem, A.; Güttler, S.; Hartung, E.; Ebstein, F.; Schaefer, M.; Tannert, A.; Salama, A.; Movassaghi, K.; Opitz, C.; Mages, H.W.; et al. Superior antigen cross-presentation and XCR1 expression define human CD11c+CD141+ cells as homologues of mouse CD8+ dendritic cells. J. Exp. Med. 2010, 207, 1273–1281. [Google Scholar] [CrossRef] [Green Version]
  119. Crozat, K.; Guiton, R.; Contreras, V.; Feuillet, V.; Dutertre, C.A.; Ventre, E.; Vu Manh, T.P.; Baranek, T.; Storset, A.K.; Marvel, J.; et al. The XC chemokine receptor 1 is a conserved selec-tive marker of mammalian cells homologous to mouse CD8alpha+ dendritic cells. J. Exp. Med. 2010, 207, 1283–1292. [Google Scholar] [CrossRef]
  120. Colletti, N.J.; Liu, H.; Gower, A.C.; Alekseyev, Y.O.; Arendt, C.W.; Shaw, M.H. TLR3 Signaling Promotes the Induction of Unique Human BDCA-3 Dendritic Cell Populations. Front. Immunol. 2016, 7, 88. [Google Scholar] [CrossRef] [Green Version]
  121. Zelenay, S.; Keller, A.M.; Whitney, P.G.; Schraml, B.U.; Deddouche, S.; Rogers, N.C.; Schulz, O.; Sancho, D.; Reis e Sousa, C. The dendritic cell receptor DNGR-1 con-trols endocytic handling of necrotic cell antigens to favor cross-priming of CTLs in virus-infected mice. J. Clin. Investig. 2012, 122, 1615–1627. [Google Scholar] [CrossRef] [Green Version]
  122. Cytlak, U.; Resteu, A.; Pagan, S.; Green, K.; Milne, P.; Maisuria, S.; McDonald, D.; Hulme, G.; Filby, A.; Carpenter, B.; et al. Differential IRF8 Transcription Factor Requirement Defines Two Pathways of Dendritic Cell Development in Humans. Immunity 2020, 53, 353–370.e8. [Google Scholar] [CrossRef]
  123. Villani, A.C.; Satija, R.; Reynolds, G.; Sarkizova, S.; Shekhar, K.; Fletcher, J.; Griesbeck, M.; Butler, A.; Zheng, S.; Lazo, S.; et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 2017, 356, eaah4573. [Google Scholar] [CrossRef] [Green Version]
  124. Bourdely, P.; Anselmi, G.; Vaivode, K.; Ramos, R.N.; Missolo-Koussou, Y.; Hidalgo, S.; Tosselo, J.; Nuñez, N.; Richer, W.; Vincent-Salomon, A.; et al. Transcriptional and Functional Analy-sis of CD1c(+) Human Dendritic Cells Identifies a CD163(+) Subset Priming CD8(+)CD103(+) T Cells. Immunity 2020, 53, 335–352.e8. [Google Scholar] [CrossRef]
  125. Santegoets, S.J.; Duurland, C.L.; Jordanova, E.J.; van Ham, V.J.; Ehsan, I.; Loof, N.M.; Narang, V.; Dutertre, C.A.; Ginhoux, F.; van Egmond, S.L.; et al. CD163(+) cytokine-producing cDC2 stim-ulate intratumoral type 1 T cell responses in HPV16-induced oropharyngeal cancer. J. Immunother. Cancer 2020, 8, e001053. [Google Scholar] [CrossRef] [PubMed]
  126. See, P.; Dutertre, C.-A.; Chen, J.; Günther, P.; McGovern, N.; Irac, S.E.; Gunawan, M.; Beyer, M.; Händler, K.; Duan, K.; et al. Mapping the human DC lineage through the integration of high-dimensional techniques. Science 2017, 356, eaag3009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Affandi, A.J.; Grabowska, J.; Olesek, K.; Lopez Venegas, M.; Barbaria, A.; Rodríguez, E.; Mulder, P.P.G.; Pijffers, H.J.; Ambrosini, M.; Kalay, H.; et al. Selective tumor antigen vaccine de-livery to human CD169(+) antigen-presenting cells using ganglioside-liposomes. Proc. Natl. Acad. Sci. USA 2020, 117, 27528–27539. [Google Scholar] [CrossRef]
  128. Tacken, P.J.; De Vries, I.J.M.; Torensma, R.; Figdor, C. Dendritic-cell immunotherapy: From ex vivo loading to in vivo targeting. Nat. Rev. Immunol. 2007, 7, 790–802. [Google Scholar] [CrossRef]
  129. Stolk, D.A.; de Haas, A.; Vree, J.; Duinkerken, S.; Lübbers, J.; Van De Ven, R.; Ambrosini, M.; Kalay, H.; Bruijns, S.; Van Der Vliet, H.J.; et al. Lipo-Based Vaccines as an Approach to Target Dendritic Cells for Induction of T- and iNKT Cell Responses. Front. Immunol. 2020, 11, 990. [Google Scholar] [CrossRef] [PubMed]
  130. Wamhoff, E.-C.; Schulze, J.; Bellmann, L.; Rentzsch, M.; Bachem, G.; Fuchsberger, F.F.; Rademacher, J.; Hermann, M.; Del Frari, B.; van Dalen, R.; et al. A Specific, Glycomimetic Langerin Lig-and for Human Langerhans Cell Targeting. ACS Cent. Sci. 2019, 5, 808–820. [Google Scholar] [PubMed] [Green Version]
  131. Schulze, J.; Rentzsch, M.; Kim, D.; Bellmann, L.; Stoitzner, P.; Rademacher, C. A Liposomal Platform for Delivery of a Protein Antigen to Langerin-Expressing Cells. Biochemistry 2019, 58, 2576–2580. [Google Scholar] [CrossRef] [PubMed]
  132. Klauber, T.C.; Laursen, J.M.; Zucker, D.; Brix, S.; Jensen, S.S.; Andresen, T.L. Delivery of TLR7 agonist to monocytes and dendritic cells by DCIR targeted liposomes induces robust production of anti-cancer cytokines. Acta Biomater. 2017, 53, 367–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Rueda, F.; Eich, C.; Cordobilla, B.; Domingo, P.; Acosta, G.; Albericio, F.; Cruz, L.J.; Domingo, J.C. Effect of TLR ligands co-encapsulated with multiepitopic antigen in nanoliposomes targeted to human DCs via Fc receptor for cancer vaccines. Immunobiology 2017, 222, 989–997. [Google Scholar] [CrossRef] [PubMed]
  134. Masterman, K.A.; Haigh, O.L.; Tullett, K.M.; Leal-Rojas, I.M.; Walpole, C.; Pearson, F.E.; Cebon, J.; Schmidt, C.; O’Brien, L.; Rosendahl, N.; et al. Human CLEC9A antibodies deliver NY-ESO-1 antigen to CD141(+) dendritic cells to activate naïve and memory NY-ESO-1-specific CD8(+) T cells. J. Immunother. Cancer 2020, 8, e000691. [Google Scholar] [CrossRef] [PubMed]
  135. Pearson, F.E.; Tullett, K.M.; Leal-Rojas, I.M.; Haigh, O.L.; Masterman, K.-A.; Walpole, C.; Bridgeman, J.S.; McLaren, J.E.; Ladell, K.; Miners, K.; et al. Human CLEC9A antibodies deliver Wilms’ tumor 1 (WT1) antigen to CD141 + dendritic cells to activate naïve and memory WT1-specific CD8 + T cells. Clin. Transl. Immunol. 2020, 9, e1141. [Google Scholar] [CrossRef]
  136. Ghinnagow, R.; De Meester, J.; Cruz, L.J.; Aspord, C.; Corgnac, S.; Macho-Fernandez, E.; Soulard, D.; Fontaine, J.; Chaperot, L.; Charles, J.; et al. Co-delivery of the NKT agonist α-galactosylceramide and tumor antigens to cross-priming dendritic cells breaks tolerance to self-antigens and promotes antitumor responses. OncoImmunology 2017, 6, e1339855. [Google Scholar] [CrossRef]
  137. Mebius, R.E.; Kraal, G. Structure and function of the spleen. Nat. Rev. Immunol. 2005, 5, 606–616. [Google Scholar] [CrossRef]
  138. Akiyama, H.; Miller, C.; Patel, H.V.; Hatch, S.C.; Archer, J.; Ramirez, N.-G.P.; Gummuluru, S. Virus particle release from glycosphin-golipid-enriched microdomains is essential for dendritic cell-mediated capture and transfer of HIV-1 and henipavirus. J. Virol. 2014, 88, 8813–8825. [Google Scholar] [CrossRef] [Green Version]
  139. Puryear, W.B.; Yu, X.; Ramirez, N.P.; Reinhard, B.M.; Gummuluru, S. HIV-1 incorporation of host-cell-derived glycosphingolipid GM3 allows for capture by mature dendritic cells. Proc. Natl. Acad. Sci. USA 2012, 109, 7475–7480. [Google Scholar] [CrossRef] [Green Version]
  140. Puryear, W.B.; Akiyama, H.; Geer, S.D.; Ramirez, N.P.; Yu, X.; Reinhard, B.M.; Gummuluru, S. Interferon-Inducible Mechanism of Dendritic Cell-Mediated HIV-1 Dissemination Is Dependent on Siglec-1/CD169. PLOS Pathog. 2013, 9, e1003291. [Google Scholar] [CrossRef] [Green Version]
  141. Saunderson, S.C.; Dunn, A.C.; Crocker, P.; McLellan, A.D. CD169 mediates the capture of exosomes in spleen and lymph node. Blood 2014, 123, 208–216. [Google Scholar] [CrossRef]
  142. Crocker, P.R.; Paulson, J.C.; Varki, A. Siglecs and their roles in the immune system. Nat. Rev. Immunol. 2007, 7, 255–266. [Google Scholar] [CrossRef] [PubMed]
  143. Macauley, M.; Crocker, P.R.; Paulson, J.C. Siglec-mediated regulation of immune cell function in disease. Nat. Rev. Immunol. 2014, 14, 653–666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Grabowska, J.; Lopez-Venegas, M.A.; Affandi, A.J.; Haan, J.M.M.D. CD169+ Macrophages Capture and Dendritic Cells Instruct: The Interplay of the Gatekeeper and the General of the Immune System. Front. Immunol. 2018, 9, 2472. [Google Scholar] [CrossRef] [Green Version]
  145. Xu, F.; Bandara, A.; Akiyama, H.; Eshaghi, B.; Stelter, D.; Keyes, T.; Straub, J.E.; Gummuluru, S.; Reinhard, B.M. Membrane-wrapped nanoparticles probe divergent roles of GM3 and phosphatidylserine in lipid-mediated viral entry pathways. Proc. Natl. Acad. Sci. USA 2018, 115, E9041–E9050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Yu, X.; Feizpour, A.; Ramirez, N.-G.P.; Wu, L.; Akiyama, H.; Xu, F.; Gummuluru, S.; Reinhard, B.M. Glycosphingolipid-functionalized nanoparticles recapitu-late CD169-dependent HIV-1 uptake and trafficking in dendritic cells. Nat. Commun. 2014, 5, 4136. [Google Scholar] [CrossRef] [Green Version]
  147. Yu, X.; Xu, F.; Ramirez, N.-G.P.; Kijewski, S.D.G.; Akiyama, H.; Gummuluru, S.; Reinhard, B.M. Dressing up Nanoparticles: A Membrane Wrap to Induce Formation of the Virological Synapse. ACS Nano 2015, 9, 4182–4192. [Google Scholar] [CrossRef] [Green Version]
  148. Chen, W.C.; Kawasaki, N.; Nycholat, C.M.; Han, S.; Pilotte, J.; Crocker, P.R.; Paulson, J.C. Antigen Delivery to Macrophages Using Liposomal Nanoparticles Targeting Sialoadhesin/CD169. PLoS ONE 2012, 7, e39039. [Google Scholar] [CrossRef]
  149. Leylek, R.; Alcántara-Hernández, M.; Lanzar, Z.; Lüdtke, A.; Perez, O.A.; Reizis, B.; Idoyaga, J. Integrated Cross-Species Analysis Identi-fies a Conserved Transitional Dendritic Cell Population. Cell Rep. 2019, 29, 3736–3750.e8. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Pathogen mimicry of liposomal cancer vaccines augment their efficacy. (A) Pathogens harbor (1) antigenic proteins or peptide, from which an immune response can be elicited, (2) pathogen-associated molecular patterns (PAMPs) that activate the immune system, and (3) recognition molecules that allow for uptake by host cells. Liposomal cancer vaccines mimic pathogens by incorporation of (1) antigens, for example, protein/peptide, or mRNA, (2) TLR ligands or nucleotides to activate the immune system, and (3) modification of the liposomal surface with targeting moieties that allow for tailored interactions between the liposomal vaccine and immune cells. (B) Upon pathogen challenge or liposomal cancer vaccination, designated APCs take up and subsequently cross-present antigen in MHC class I to induce potent immune responses. Liposomal cancer vaccine mimicry can optimize vaccine interaction with the immune system that subsequently translates to robust antigen-specific immune responses.
Figure 1. Pathogen mimicry of liposomal cancer vaccines augment their efficacy. (A) Pathogens harbor (1) antigenic proteins or peptide, from which an immune response can be elicited, (2) pathogen-associated molecular patterns (PAMPs) that activate the immune system, and (3) recognition molecules that allow for uptake by host cells. Liposomal cancer vaccines mimic pathogens by incorporation of (1) antigens, for example, protein/peptide, or mRNA, (2) TLR ligands or nucleotides to activate the immune system, and (3) modification of the liposomal surface with targeting moieties that allow for tailored interactions between the liposomal vaccine and immune cells. (B) Upon pathogen challenge or liposomal cancer vaccination, designated APCs take up and subsequently cross-present antigen in MHC class I to induce potent immune responses. Liposomal cancer vaccine mimicry can optimize vaccine interaction with the immune system that subsequently translates to robust antigen-specific immune responses.
Pharmaceutics 13 00954 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Nijen Twilhaar, M.K.; Czentner, L.; van Nostrum, C.F.; Storm, G.; den Haan, J.M.M. Mimicking Pathogens to Augment the Potency of Liposomal Cancer Vaccines. Pharmaceutics 2021, 13, 954. https://doi.org/10.3390/pharmaceutics13070954

AMA Style

Nijen Twilhaar MK, Czentner L, van Nostrum CF, Storm G, den Haan JMM. Mimicking Pathogens to Augment the Potency of Liposomal Cancer Vaccines. Pharmaceutics. 2021; 13(7):954. https://doi.org/10.3390/pharmaceutics13070954

Chicago/Turabian Style

Nijen Twilhaar, Maarten K., Lucas Czentner, Cornelus F. van Nostrum, Gert Storm, and Joke M. M. den Haan. 2021. "Mimicking Pathogens to Augment the Potency of Liposomal Cancer Vaccines" Pharmaceutics 13, no. 7: 954. https://doi.org/10.3390/pharmaceutics13070954

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop