Next Article in Journal
OX40-OX40L Inhibition for the Treatment of Atopic Dermatitis—Focus on Rocatinlimab and Amlitelimab
Next Article in Special Issue
Thymol-Loaded Eudragit RS30D Cationic Nanoparticles-Based Hydrogels for Topical Application in Wounds: In Vitro and In Vivo Evaluation
Previous Article in Journal
Integrated Analysis of Metabolomics and Lipidomics in Plasma of T2DM Patients with Diabetic Retinopathy
Previous Article in Special Issue
The Experimental Study of Periodontal Ligament Stem Cells Derived Exosomes with Hydrogel Accelerating Bone Regeneration on Alveolar Bone Defect
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Locoregional Lymphatic Delivery Systems Using Nanoparticles and Hydrogels for Anticancer Immunotherapy

1
Division of Biological Science and Technology, Yonsei University, Wonju 26493, Republic of Korea
2
Department of Food and Nutrition, Andong National University, Andong 36729, Republic of Korea
*
Author to whom correspondence should be addressed.
Pharmaceutics 2022, 14(12), 2752; https://doi.org/10.3390/pharmaceutics14122752
Submission received: 28 October 2022 / Revised: 22 November 2022 / Accepted: 7 December 2022 / Published: 8 December 2022

Abstract

:
The lymphatic system has gained significant interest as a target tissue to control cancer progress, which highlights its central role in adaptive immune response. Numerous mechanistic studies have revealed the benefits of nano-sized materials in the transport of various cargos to lymph nodes, overcoming barriers associated with lymphatic physiology. The potential of sustained drug delivery systems in improving the therapeutic index of various immune modulating agents is also being actively discussed. Herein, we aim to discuss design rationales and principles of locoregional lymphatic drug delivery systems for invigorating adaptive immune response for efficient antitumor immunotherapy and provide examples of various advanced nanoparticle- and hydrogel-based formulations.

1. Introduction

Immunotherapy has garnered attention in anticancer therapy owing to advances in biotechnology and expanded understanding of the complex immune networks in cancer progression [1]. Cancer cells create immunosuppressive “cold” tumor microenvironments to evade the body’s immune surveillance [2]. Immune checkpoint blockades (ICBs), such as antagonistic cytotoxic T-lymphocyte-associated protein (CTLA)-4, anti-programmed cell death (PD)-1, and anti-PD-ligand (PD-L)-1 monoclonal antibodies (mAbs), which are clinically approved by the U.S. Food and Drug Administration (FDA), are representative immunotherapeutics that reprogram these “cold” tumors to “hot” tumors that are not only characterized by abundant cytotoxic CD8+ T cells (CTLs) and natural killer (NK) cells, but are also responsive to these immune cells [2,3]. The therapeutic effects of chemotherapy, the most widely utilized anticancer methodology in the clinic, are found to be either mainly or partially dependent on direct or indirect immuno-modulatory effects of the drugs. As a result, recent re-evaluation and re-purposing of the traditional chemotherapeutics in terms of chemoimmunotherapy have accelerated [4,5]. Accordingly, the anticancer therapy paradigm has been shifting from being heavily focused on cancer cells to a comprehensive consideration of localized tumor microenvironment and systemic immune responses [1,2,4].
Current anticancer immunotherapy can be roughly categorized into two strategies: Initiation of anticancer immunity by improving the susceptibility of cancer cells to immune cells to exert anticancer effects in tumor microenvironments. This strategy includes the induction of immunogenic cell death (ICD) to release adenosine triphosphate (ATP), calreticulin (CRT), and high mobility group box protein 1 (HMGB-1) for the recruitment of dendritic cells (DCs) that phagocytize cancer cells or their antigens [4,6,7,8,9], suppression of immunosuppressive pathways of cancer cells [10] or cancer-associated ECM-resident cells [11], or inhibition of the signals associated with the immune checkpoints between cancer cells and immune cells [3,7,12,13,14]. Accordingly, nano-sized materials that can efficiently deliver drugs to tumor tissues via enhanced permeation and retention (EPR) serve as one of the attractive tools to achieve efficient anticancer immunotherapy [4,15,16,17].
The lymph nodes (LNs)-centered anticancer immunotherapy to achieve efficient expansion of immune cells in the secondary lymphoid tissues, followed by increased infiltration in tumor microenvironments serves as the second strategy [1,5,18,19,20,21,22,23,24,25,26,27,28,29,30]. LNs are the primary tissues for adaptive immunity. In fact, LNs instruct and expand antigen-specific CTLs via antigen-presenting cells (APCs), such as DCs [1,18,19,20,21,22]. The importance of LNs in adaptive anticancer immunotherapy has been emphasized recently. Francis et al. reported that intradermal (i.d.) injections of ICBs to the tissue ipsilateral (i.l.) to the tumor allows their preferential accumulation in draining LNs (dLNs) over tumor tissues, leading to similar therapeutic effects to intratumoral (i.t.) administration enabling tumor and tumor dLN (TdLN) accumulation [31]. Similarly, Manspeaker et al. demonstrated that locoregional i.l. administration for TdLNs targeting led to a higher anticancer therapeutic index than intravenous (i.v.) administration, mainly targeting the tumor microenvironment in the combination therapy with the PD-1 monoclonal antibody (aPD-1 mAb) and nanoparticles (NPs) loaded with paclitaxel (PTX) [5]. Even in the aforementioned immunotherapy strategy that primarily targets the tumor microenvironments, DCs migrated from peripheral tissues to the LNs after the uptake of antigens produced via natural apoptosis or by cytotoxic drugs, initiate and propagate the anticancer immunotherapy.
The pharmacokinetics of immuno-modulatory drugs govern therapeutic effects, side effects, and patient compliance regarding cancer immunotherapy [16,32,33,34,35,36,37,38]. A hydrogel (HG) is a 3D polymeric network that contains water and serves as one of the important classes of drug delivery systems for the sustained release of drugs. Indeed, i.t. administration of HGs has been reported to lead to prolonged accumulation of immuno-stimulatory drugs to the tumor microenvironment, leading to a higher anticancer therapeutic index than that obtained with bolus delivery [32,33,34]. In addition, locoregional i.l. administration of ICBs-loaded HGs resulted in prolonged and higher accumulations of mAbs in TdLNs, followed by improved anticancer immunotherapy [31,35].
After considering numerous reviews that enabled an extensive and intensive discussion of the design rationales and examples of biomaterials for the delivery of immuno-therapeutics to the tumor microenvironment [1,4,16,17,36,37,38], we aimed to discuss the principles, design rationales, and recent progress of lymphatic drug delivery systems to achieve efficient adaptive anticancer immunotherapy. First, the design rationales and examples of advanced NPs required to overcome physiological barriers of lymphatic systems were summarized in conjunction with detailed immune response. Second, several advanced HGs that enable prolonged adaptive immune response in LNs were discussed. Finally, hybrid HGs and NPs systems that allow sustained delivery of drug-loaded NPs to the LNs for efficient anticancer immunotherapy were introduced.

2. Advanced NPs for LNs Targeting and Adaptive Cancer Immunotherapy

2.1. Basic Criteria for the Lymphatic Delivery of NPs and Immunomodulatory Drugs

LNs-centered adaptive cancer immunotherapy requires a cascade pathway. First, NPs containing immuno-modulatory drugs should drain into the LNs [18,19,21]. In locoregional administration of NPs, such as i.d., i.t., subcutaneous (s.c), or intramuscular (i.m.), the transport of NPs to dLNs depends on interstitial flow, a bulk fluid flow for transit to the initial lymphatics within tissues [18,39]. NPs with a hydrodynamic size ranging from 10 to 50 nm can efficiently pass through the pores of the extracellular matrix of tissues in bulk [40,41,42,43,44,45]. However, the size threshold for delivery into the lymphatics can differ based on the flexibility and rigidity of the nanomaterials [40,41,42,43,44,45,46]. Further, NPs with negative surface charges display a higher transport to the lymphatics than NPs with positive charges, which is due to the negative charge of the ECM [43,47,48,49,50]. In this regard, the surface modification of polyethylene glycol (PEG) to facilitate anti-biofouling [51] and neutralization of surface charges can improve the lymphatic delivery efficacy of NPs [52,53,54,55]. In particular, lymph-homing and migratory APCs contribute to the cell-mediated lymphatic delivery of NPs not only via phagocytosis [27,41,42,44,52], but also selective interaction with NPs engineered for modification with mannose, mannan, hyaluronic acid, hyaluronan, and antibodies to bind to the receptors of APCs [56,57,58,59,60,61,62,63,64]. During systemic delivery of NPs, such as intravenous (i.v.) or intraperitoneal (i.p.), NPs in the bloodstream can access LNs via high endothelial venules (HEVs) enabling transmigration of lymphocytes interacting with peripheral node addressin (PANd) in LNs [18,19,65,66,67,68,69], which can be improved when NPs are decorated with PNAd-targeting antibodies [70,71,72]. Second, immuno-modulatory drugs that reach the afferent lymphatic vessels of LNs can access the LN parenchyma when NPs or drugs released from NPs are smaller than 70 kDa in size [73]. APCs that have already encountered NPs and immuno-modulatory drugs, or other migratory immune cells interacting with NPs containing immuno-modulatory drugs, facilitate cell-mediated delivery to the cortex and paracortex regions of LNs, where adaptive immune responses occur [74,75,76,77,78].

2.2. Advanced NPs to Exploit Passive Transport to the LNs

Passive transport involves the delivery of cargos to LNs by exploiting interstitial flow toward LNs within the ECM. Considering the physicochemical properties that enable transport through the ECM from peripheral tissues to LNs [18,39,40,41,42,43,44,45,46,47,48,49,50,51], efficient lymphatic delivery of small molecular immuno-modulatory drugs requires advanced drug delivery systems not only to tune the physicochemical properties of cargos comprising drugs and drug delivery systems, but also to allow the loading and release of drugs at LNs. Depending on the intrinsic properties of immuno-modulatory drugs, various strategies can be employed for drug loading, such as hydrophobic/hydrophilic interactions, electrostatic interactions, nucleotides hybridization, protein engineering, or chemical conjugation. Recent advancements in NP design for efficient lymphatic passive transport are discussed below.

2.2.1. Physical Load; Hydrophobic/Hydrophilic Interactions

Micelles are one of the representative drug delivery systems that allow the loading of hydrophobic drugs into the hydrophobic core of structures formed via the self-assembly of block-copolymers comprising hydrophilic and hydrophobic segments in an aqueous solution [79]. Thomas et al. utilized a micelle physically entangled by poly(propylene sulfide) (PPS) to explore the adjuvant potential of paclitaxel (PTX) in LNs. PTX-loaded PPS micelles (~30 nm) administered i.l. caused moderate suppression of B16F10 melanoma tumor growth, and were more efficient than those administered c.l. (i.d. administration to the tissue contralateral to the tumor) [80]. The mechanism underlying the therapeutic effects of PTX is ascribed to the activation (CD86+, CD40+ and/or MHCII+) and expansion of conventional DCs (cDCs, CD45+CD11b+CD11c+) in LNs [5,80,81]. In particular, i.l. or i.t. administration of free aPD-1 mAbs combined with PTX-loaded PPS micelles administered i.l. significantly expanded stem-like CD8+ T cells (Tcf-1+Tim-3-PD-1+ CD45+CD3+CD8+) systemically in triple-negative breast cancer (TNBC) E0771 tumor models after selective accumulation in TdLNs, leading to the control of distant and local tumors [5]. This series of projects suggest a strategy to re-purpose traditional drugs by exploiting micelles as lymphatic delivery systems [5,80,81].
Contrary to the micelles, lipid NPs and liposomes serve as a carrier for hydrophobic and hydrophilic drugs by incorporating them into the hydrophobic and hydrophilic segments of the self-assembled structures, respectively [23,82]. Hanson et al. utilized lipid NPs to demonstrate the potential of lymphatic accumulation of a stimulator of IFN gene (STING) agonists as a vaccine adjuvant [83]. Indeed, lipid NPs (150 nm) administered s.c. led to increased accumulation of the STING agonists in dLNs and mitigated systemic exposure of the drugs in blood. Compared to soluble STING agonists, the STING agonist-loaded lipid NPs significantly improved the expansion of antigen-specific (tetramer+) CD8+ T cells (CD45+CD3+CD8+) when co-administered with ovalbumin antigen (OVA), leading to a markedly higher anticancer effect in terms of survival and tumor growth [83].

2.2.2. Physical Load; Electrostatic Interactions

Nucleotides and nucleic acids contain phosphate backbones that have negative charges, enabling electrostatic complexation with positive charged-materials for the development of nano-sized particles that have been explored as a non-viral gene delivery option [16,27]. Similarly, nucleotide adjuvants or nucleic acids encoding antigens can be efficiently formulated into NPs via electrostatic complexation for efficient lymphatic delivery. Solid lipid NPs (SLP) containing cationic phospholipids (152 nm) could not only physically load mRNA encoding OVA, tyrosinase-related protein 2 (TRP-2), or point-mutated version of glycoprotein 100 (gp100) that are melanoma self-antigens but also efficiently drain into the dLNs [84], ultimately resulting in improved antigen-specific (tetramer+) CD8+ T cell (CD45+CD3+CD8+) expansion and efficient tumor regression. Nucleotide-based adjuvants can be delivered to lymphoid tissues using cationic nanomaterials, thereby mitigating systemic non-specific immune response. For example, the s.c. administration of gelatin NPs (272 nm) cationized by quaternary amino groups to electrostatically load CpG oligonucleotides, a Toll-like receptor 9 (TLR9) ligand, led to their selective accumulation in dLNs over the spleen, resulting in the efficient activation of CpG-positive DCs in LNs, suppression of tumor growth, and prevention of splenic follicle destruction [85]. In addition to NPs comprising organic materials, such as lipids and polymers [84,85,86,87], inorganic materials can be utilized to electrostatically load and deliver nucleotides and nucleic acids to the LNs for efficient anticancer immunotherapy. The benefits of inorganic NPs relative to organic NPs include the ease of tuning the size and surface charges and rendering additional functions, such as imaging, photothermal and photodynamic properties, and intrinsic adjuvant effects. For example, triethoxypropylaminosilane-functionalized silica NPs (SiNPs, 30 nm) [88], polyethyleneimine (PEI)-functionalized mesoporous silica NPs (MSN, ~200 nm) [89], and aluminum hydroxide NPs (~85 nm) [90] were explored as CpG lymphatic delivery systems for efficient anticancer immunotherapy; however, their intrinsic adjuvant effects [91] during delivery to the LNs need to be further investigated. The lymphatic transport mechanism of these rigid NPs with large sizes must also be investigated [40,41,42,43,44,45,46].
In non-viral gene delivery systems, the size of polyplex NPs is inversely proportional to the multivalent electrostatic interactions governed by Coulomb’s Law. Short-length monomeric small interfering RNA (siRNA) resulted in NPs with a size of ~460 nm with linear PEI (LPEI). The formation of a multimerized siRNA through self-crosslinking between siRNA via reducible disulfide bonds could lead to NPs with a size of ~82 nm [92]. The compact structures of NPs in multimerized siRNA are attributed to the higher charge density and more flexible structure than those of monomeric siRNA. Accordingly, polyplex NPs are often developed using multimerized nucleotide-based adjuvants for efficient lymphatic delivery. The Xiaoyuan Chen group utilized rolling circle amplification (RCA) to construct polymerized CpG with additional polymerized shRNA encoding signal transducer and activator of transcription 3 (Stat3) to suppress antigen-specific T cell tolerance [93,94]. Microflowers comprising poly(CpG) and poly(shStat3) can shrink from ~1200 nm to NPs of ~250 nm when complexed with positively charged PPT-g-PEG polymer, leading to efficient presentation of antigens on DCs (SIINFEKL+ CD45+CD11c+) in LNs after i.l. administration (Figure 1) [93]. In another study, poly(CpG) microflowers were produced on primers conjugated to the PEG segments of PEG-PLA micelles, enabling the co-encapsulation of a hydrophobic peptide neoantigen to MC38 tumor (herein, adpgk) and a hydrophobic adjuvant (herein, a TLR 7/8 agonist R848) in the core of micelles [94]. The resulting micelle-based microflowers containing poly(CpG), adpgk, and R848 with ~2200 nm size can be condensed into NPs with a size of ~170 nm when complexed with PPT-g-PEG. The resulting polyplex micelles were found to have higher LN accumulations than their free formulations following s.c. administration, ultimately leading to a higher expansion of LN-resident DCs (CD45+CD11c+) and macrophages (CD45+F4/80+), and systemic antigen-specific (dextramer+) CD8+ T cells (CD45+CD3+CD8+), and efficient tumor regression [94]. The combined administration of aPD-1 mAbs could also improve the therapeutic index [94].

2.2.3. DNA/RNA Engineering; Spherical Nucleic Acids (SNA) and DNA/RNA Origami

Spherical nucleic acids (SNAs), which were first introduced by Chad Mirkin’s group, comprise an NP core and a densely packed nucleic acid layer [95]. SNAs exhibit distinguished properties from bare nucleic acids in terms of cellular uptake, stability against enzymatic degradation, and adsorption with endogenous biomolecules [95,96]. SNA with a 13 nm gold NP (AuNP) core and a CpG shell displayed significant accumulation in the subcapsular sinus of dLNs, but not in the parenchyma [97]. Nevertheless, the CpG-decorated SNA induced the release of higher amounts of interleukin (IL)-12 from dLNs than free CpG, which significantly suppressed tumor growth when combined with an antigenic OVA protein or SIINFEKL peptides [97]. In a study utilizing liposomal SNA (~120 nm) where cholesterol-modified DNA is embedded into the phospholipid membrane of core liposome (80–90 nm), the effects of liposomal membrane fluidity on lymphatic delivery of SNA were examined by varying the lipids with different phase transition temperature (Tc) [98]. Despite the identical NP size and surface chemistry, a liposomal SNA with higher Tc accumulated in a higher quantity in dLNs than that with lower Tc at an early time point (2 h after s.c. injection), which led to more efficient suppression of tumor growth [98]. However, the accumulation of liposomal SNA was equivalent at 24 h, regardless of Tc [98]. As membrane fluidity is reversely proportional to Tc, these results imply that a rigid core is beneficial in the rapid lymphatic delivery of SNA, followed by LNs-mediated anticancer immunotherapy. The James Moon groups reported a liposomal nanodisc, in which cholesterol-modified CpG was embedded on the surficial phospholipid membrane and cysteine-functionalized neoantigen peptides were chemically conjugated to the thiol-functionalized phospholipids (Figure 2) [99]. Nanodiscs injected s.c. facilitated higher accumulation of CpG and antigen peptides in the dLNs, enabling efficient antigen-specific (tetramer+) CD8+ T-mediated anticancer therapy [99]. As numerous kinds of core NPs can facilitate the loading of various hydrophobic and hydrophilic adjuvants and antigens, SNAs with various functions are expected to serve as a platform for lymphatic-mediated anticancer immunotherapy [100,101].
DNA and RNA have been exploited as building blocks for well-defined nanostructures owing to their Watson–Crick base pairing [95,102]. One of the interesting fields include DNA/RNA origamis that fold long single-stranded DNA/RNA scaffolds into an arbitrary structure using short DNA/RNA complementary to the scaffold as a staple [102]. Previously, a DNA tetrahedron structure was demonstrated to display a higher accumulation in dLNs than double stranded linear DNA [103]. The Baoquan Ding groups reported an interesting DNA nanodevice that enabled structural transformation from tubular to sheet under intracellular endosomal pH using DNA origami (Figure 3) [104]. Multiple staple strands were utilized to fabricate a rectangle-shaped DNA origami from M13 bacteriophage DNA, which was further exploited to load tumor peptide antigen-DNA conjugates as an antigen and CpG and double-stranded RNA (dsRNA) as adjuvants via hybridization [104]. pH-responsive DNA was also employed to hybridize the DNA in long edges of rectangle-shaped DNA origami, enabling the formation of a tubular nanostructure with diameter and length of ~19 nm and ~90 nm, respectively, and pH-responsive transformation [104]. Compared to the physical mixture of therapeutics, including peptide antigens, CpG, and dsRNA, the resulting DNA nanodevices could drain into dLNs ~5 times more, eliciting higher expansion and activation (CD86+) of DCs (CD45+CD11c+), followed by enrichment of antigen-specific (tetramer+) CTL (IFN-γ+ CD45+CD3+CD8+) [104]. Accordingly, robust antitumor immunotherapy in various tumor models, including MC38, B16F10, and B16F10-OVA, was achieved [104].

2.2.4. Self-Assembled or Recombinant Protein

In addition to their general therapeutic roles, including as antigens or therapeutic antibodies, proteins can serve as a lymphatic drug delivery platform. Owing to size-dependent LN accumulation, self-assembled OVA NPs (~80 nm) were developed by cleaving the disulfide bonds of an OVA antigen protein to change its charges to positive and then complex it with CpG [105]. Coordination among OVA antigen proteins, manganese (Mn2+), and meso-2,6-diaminopimelic acid (DAP) was found to facilitate the preparation of nanocomposites with a size of 150 nm [106]. The increased size of these self-assembled antigen NPs led to a significantly higher LN accumulation of antigens than bare antigen proteins [105,106], followed by successful suppression of tumor growth [106].
Proteins vary in size, 3D structures, and surface characteristics, which critically affect lymphatic delivery. Advances in genetic recombination and protein engineering technology enable not only the preparation of recombinant proteins to present antigens or antibodies but also the tuning of physicochemical properties for self-assembly. Lee et al. demonstrated that human ferritin (hFTN, ~11 nm) exhibited a 3-to-7-fold higher accumulation than Escherichia coli DNA binding protein (DPS, 9.5 nm), Thermoplasma acidophilum proteasome (PTS, 13.4 nm), and Hepatitis B virus capsid (HBVC, 32.3 nm) [107]. Genetic fusion of RFP and hFTN subunits afforded an engineered hFTN to present a model tumor antigen (herein, red-fluorescence protein (RFP)), which induced conformational changes in proteins. The diameter of the engineered hFTN increased to ~22 nm. The hFTN-RFP protein NPs efficiently drained into the LNs, inducing efficient antitumor immunotherapy against RFP-expressing tumor-bearing models [107]. Owing to the efficient lymphatic transport of hFTN, Kim et al. developed hFTN fusion proteins to present a PD-1 (Figure 4) [108]. The resultant nano-cages had a larger size (~22 nm) than wild-type hFTN (wtNCs, ~13 nm), and they displayed not only a higher affinity against PD-L1 and PD-L2 than soluble PD-1 (sPD-1) but also higher accumulation in dLNs than the monomeric form of sPD-1 and wtNCs. Accordingly, efficient activation (CD86+, CD40+) of DCs (CD45+CD11c+) and CD8+ T (CD45+CD3+CD8+) cells-mediated inhibition of tumor growth occurred [108]. A further study to examine the mechanism whereby the properties of hFTN facilitate the efficient lymphatic delivery is expected to contribute to the development of advanced lymphatic delivery systems using protein scaffolds.

2.2.5. Chemical Conjugation

Drugs with functional groups that can participate in a chemical reaction can be conjugated to various drug delivery systems, including iron oxide NPs, quantum dots, graphene oxide, liposomes, micelles, etc. [109,110,111,112,113]. Examples include conjugations of CpG and siStat3 to linkers on the quantum dots (~150 nm) via a disulfide bond [109], amide conjugations of antigen peptides to lipids to enable hydrophobic interactions with oleic acids on iron oxide NPs (30–45 nm) [110], amide conjugations of antigen peptides to PEG-(poly(maleic anhydride-alt-1-octadecene)) loaded on graphene oxide (20–30 nm) via hydrophobic interactions [111], conjugations of antigen peptides to lipids comprising liposomes (~23 nm) via an amide bond [112], and conjugations of antigen peptides to poly(L-histidine)-poly(ethylene glycol) micelles (~60 nm) via a disulfide bond [113], ultimately leading to a high accumulation of immunomodulatory drugs in the LNs, followed by efficient antitumor immunotherapy. The other examples are discussed in overall manuscript.

2.3. Advanced NPs for Active Transport to the LNs

In contrast to passive transport, active transport involves the lymphatic transport of cargos via immune cells. In particular, peripheral DCs encountered immuno-stimulatory agents and antigens can initiate adaptive antitumor immune response in dLNs owing to the intrinsic lymph-homing ability of maturating and activating APCs. A representative strategy involves the construction or functionalization of NPs with mannose & mannan, or hyaluronic acid & hyaluronan with mannose-receptors or CD44, respectively, which are expressed on the surfaces of APCs, such as DCs and macrophages [56,57,58,59,60,61,62,63,64,114,115,116,117]. Ligand-mediated improvement of the lymphatic delivery of cargos was clearly demonstrated with mannose-decorated liposomes (~160 nm) loading poly(I:C) electrostatically [56], tumor cell lysate-loaded chitosan NPs (~120 nm) functionalized with mannose [114], micelles (~95 nm) comprising stearic acid-grafted and mannose-modified chitosan [115], NPs (~310 nm) comprising alginates grafting mannose and OVA proteins [116], mannose-functionalized pH-sensitive micelles (30–50 nm) loading antigens via disulfide bonds [117], and TLR7/8 agonists and OVA proteins loaded poly(lactide-co-glycolide) (PLGA) NPs (~225 nm), whose surface was emulsified with hyaluronic acid [62]. Of note, in these NPs, the size-dependent passive transport mechanisms cannot be omitted from the uptake of drugs and NPs in lymph-homing APCs and APCs-mediated CD8+ T cell instructions in LNs. Indeed, by comparing a mannan nanocapsule and a silica NP decorated with mannan, a higher quantity of nanocapsule was found to be accumulated in the dLNs than that of silica NPs, despite similar zeta potential and size (both ~220 nm). These results were ascribed to the high deformability of nanocapsules to pass through a 50–100 nm pore-sized membrane; silica NPs cannot efficiently pass through such membrane [61]. The study findings also implied the partial, but significant contribution of passive lymphatic transport pathways, even in the case of NPs decorated with ligands, to the interaction with lymph-homing APCs.
The intrinsic biocompatibility of mammalian cell membranes has promoted the development of membrane-cloaked NPs for efficient lymphatic delivery and antitumor immunotherapy [118]. For example, red blood cell (RBC) membranes for the camouflage of PLGA NPs chemically loading antigenic peptide (herein hgp10025–33) were modified with mannose (~160 nm) by embedding lipids-PEG-mannose to improve lymphatic active transport of the NPs [119]. Compared to the controls, including PLGA NPs and RBC-cloaked PLGA NPs, the mannose-RBC-cloaked PLGA NPs were more susceptible to uptake by APCs and accumulation into dLNs, and induced more efficient CD8+ T (CD45+CD3+CD8+) cell-mediated antitumor effects after i.d. administration [119]. In this regard, cancer cell membranes containing tumor-associated antigens could be exploited as a substitute for biomaterials containing mammalian cell membranes and antigens [120]. In particular, DC membranes containing CCR7, L-selectin, and integrins can be an interesting source for camouflaging NPs to exhibit homologous targeting to LNs [121]. Indeed, functionalization with DC membranes significantly promoted the LN accumulation of OVA protein-loading micelles synthesized using stearic acid- and histidine-grafted chitosan polymers (~150 nm), and improved the activation (CD86+ and CD40+) of DCs (CD45+CD11c+) in dLNs and expansion of CD8+ T cells (CD45+CD3+CD8+) in tumors [121]. DC membranes can be engineered to present antigens on major histocompatibility complex (MHC) and CD28 co-stimulator before being used for cloaking NPs (Figure 5) [122]. Further functionalization of matured DC membrane-camouflaged NPs (~180 nm) with CD3 antibodies was reported to enable significant regression of tumor growth owing to efficient lymphatic accumulation and retention of NPs via the combined lymph-homing effects of DC membranes and T cell binding effects of the CD3 antibodies [122]. The resultant NPs containing imiquimod led to efficient antitumor effects, with significant activation of DCs (CD45+CD11c+ CD80+CD86+) and macrophages (CD80hi CD45+CD11b+F4/80+) and expansion of CD8+ T cells (CD45+CD3+CD8+) [122]. Based on these criteria, fused membranes from DCs and cancer cells have been explored in the development of membrane-cloaked NPs for antitumor immunotherapy [123,124].
Numerous studies have exploited the extracellular vesicles (EVs) secreted from various cell types as a drug delivery platform and therapeutic, as EVs contain miRNA, bioactive lipids, and proteins that can control intercellular communications [125]. As EVs are formed through fusion of the cell vacuole with cell membranes, they can contain various membrane proteins on their surface. Accordingly, EVs from matured and activated APCs could elicit LN-homing ability and immuno-stimulatory functions in antitumor immunotherapy [126,127]. Indeed, EVs released from DCs maturated and activated by OVA antigens and poly(I:C) (30–100 nm) were revealed to contain CD80 and MHC, which stimulate the activation (CD69+) of CD8+ T cells (CD45+CD3+CD8+). The EVs could efficiently drain into LNs, and their efficacy was improved with a CTLA-4 mAb decoration on the membranes by anchoring lipids-PEG-aCTLA-4 (Figure 6) [126]. In particular, the resulting aCTLA-4 decorated EVs enabled long-term antitumor immunotherapy with effector memory T cells (CD45+CD3+CD8+CD44+CD62L-) [126]. Similarly, EVs purified from M1 macrophages polarized by LPS and IFN interferon-γ (IFN-γ) (50 nm) [127] displayed significant LNs accumulation, induced the release of proinflammatory cytokines (IL-6) in dLNs, and elicited antitumor immunotherapeutic effects when co-treated with antigens.

2.4. Advanced NPs for Deeper Access to the LNs

After transport to the LNs, immuno-modulatory drugs should access the LN parenchyma where adaptive immune cells reside. However, LN conduits and cortex function as size-dependent exclusion barriers to allow access to solutes that are less than 70 kDa (~several nm in size) [73], which could lead to a size conflict for efficient passive transport [40,41,42,43,44,45,46]. The Susan N. Thomas groups reported a multistage drug delivery system comprising a 30 nm pluronic F127 micelle (30 nm), whose core was stabilized with physically entangled PPS and shell was decorated with a CpG via a linker to exhibit first order degradation independent of pH and solvents (Figure 7) [128]. This size change for the cargo allowed the drug-conjugating NPs to efficiently drain into the LNs via size-dependent passive transport and subsequently deliver drugs to the LN parenchyma [128]. Owing to the efficient delivery of CpG into the LN parenchyma, higher activation (CD40+ and/or CD86+) of conventional DCs (cDCs, CD45+CD11c+B220-), plasmacytoid DCs (pDCs, CD45+CD11c+B220+), and B cells (CD45+CD11b-CD3-B220+) was observed with NPs with multistage drug delivery compared to that with the controls [128]. Accordingly, higher expansion of T cells (CD45+CD3+B220-), which significantly suppressed EL-4 lymphoma tumor growth in LNs, was recorded [128]. This strategy outlines an approach to re-purpose numerous trojan horse and size-transformable NPs previously reported for deep penetration into the tumor tissues [129,130,131] for LNs-mediated anticancer immunotherapy.

3. Advanced HGs for LNs Accumulation and Adaptive Cancer Immunotherapy

According to numerous reports, i.t. administration of HGs facilitates the sustained release of incorporated immunotherapeutics into tumor microenvironments, enabling efficient and/or prolonged anticancer immune responses in tumors [132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164] or tumors and TdLNs [165,166,167,168,169]. The immunotherapeutics released from HGs in a sustained manner would also accumulate in the TdLNs. Some studies attempted to determine the effects of sustained release on the lymphatic accumulation of immunotherapeutics, immuno-modulations in LNs, and overall therapeutic effects [31,170,171,172]. An HG prepared via crosslinking between aldehyde-functionalized mannan and N, O-carboxymethyl chitosan led to higher accumulations of OVA antigens in dLN at 24 h and 48 h after its locoregional s.c. administration than free OVA and Alum/OVA formulations, which promoted and prolonged the production of antigen-specific immunoglobulin G (IgG), despite a slight delay in tumor growth [170]. In addition, mPEG-b-poly(L-valine) HGs prolonged and enhanced the lymphatic accumulation of tumor cell lysates that were incorporated in the HGs and released from the injection sites (Figure 8) [171]. Accordingly, significant activation (MHCII+, CD86+) of DCs (CD45+CD11c+) in dLNs and expansion of CD8+ T cells (CD45+CD3+CD8+) in dLNs, spleen, and tumors were achieved when poly(I:C) and tumor cell lysates were incorporated in the HGs administered s.c. to the tissue i.l. to the tumor [171]. Compared to the control with free formulations, tumor growth could be significantly delayed with HG [171]. Similarly, silk HGs administered s.c. to the tissue i.l. to the tumor facilitated improved accumulation of the immunotherapeutics (herein, neoantigens, CpG, and STING agonist 2, 3-cyclic-GMP-AMP (cGAMP)) in dLNs, leading to significant suppression of tumor growth with the expansion of activated DCs (CD86+CD80+ CD45+CD11c+) in dLNs and activated CD8+ T cells (41BB+ CD45+CD3+CD8+) in tumors [172]. These studies highlight the importance of the pharmacokinetics of immunotherapeutic drugs in LNs, where adaptive immune responses mainly occur. However, the mechanism whereby HG-mediated sustained release improves lymphatic accumulations of cargos has not been clearly revealed.

4. Hybrid HGs and NPs for LNs Targeting and Adaptive Cancer Immunotherapy

Hybrid systems have been developed to exploit the potential of HGs and NPs for improved accumulation of immunotherapeutics in LNs. Yin et al. mixed graphene oxide (GO, ~200 nm) with linear PEI (LPEI) to form HGs encapsulating R848 via π−π stacking with GO and OVA-mRNA via electrostatic interactions with LPEI [173]. HG-released NPs (~220 nm) formulated via an electrostatic interaction between R848-loaded GO (negative charges) and OVA-mRNA/LPEI polyplex (positive charges), were released in a sustained manner, and led to markedly higher uptake of cargos into the dLNs when injected s.c. [173]. The prolonged lymphatic delivery of R848 and OVA-mRNA resulted in the most efficient tumor suppression with CTLs (IFN-γ+ CD45+CD3+CD8+) compared to that of the controls, including saline, free R848 and OVA-mRNA, and OVA-mRNA-loaded HG [173]. Our group reported a novel polymeric HG to release drug-loaded micelles in situ without using additional NPs (Figure 9) [174]. F127-Gelatin was prepared by crosslinking F127 to gelatin, which increased the hydrophobicity, enabling the formation of a thermosensitive HG at very low concentrations (4–7 wt.%) compared with the precursor F127 (>15 wt.%) [174]. Interestingly, the F127-Gelatin polymers comprising thermosensitive HGs were released as micelles (~30 nm) loaded with aCTLA-4 mAb in a sustained manner [174]. Sustained release and a size appropriate for passive lymphatic transport facilitated the efficient and prolonged accumulation of aCTLA-4 mAb, ultimately enabling efficient anticancer immunotherapy [174]. The concept of NPs and HG hybrid systems is expected to be more extensively explored for efficient locoregional anticancer immunotherapy in the near future.

5. Conclusions and Perspectives

From a traditional pharmaceutical perspective, anticancer drugs and their delivery formulations are mainly systemically administered to selectively reach metastatic tumors or deep tumor tissues during blood circulation. Locoregional administration via the i.t. route is occasionally employed to mitigate off-target systemic delivery and improve tumoral accumulations of drugs when tumors are located to be easily accessed to the treatment.
Advances in biotechnology and expanded knowledge on immunology help highlight lymphatic systems as important targets for adaptive anticancer immunotherapy. However, the systemic administration routes have limitations, such as inefficient lymphatic delivery compared to locoregional delivery [5,31]. Accordingly, in this review, we discussed emerging locoregional drug delivery strategies that target lymphoid tissues instead of tumor and tumor microenvironments for cancer immunotherapy. The physiology of lymphatic systems motivate the use of NPs for the accumulation of immuno-modulatory drugs at LNs, where adaptive immune responses mainly occur, especially via passive lymphatic transport mechanisms. Further, sophisticated engineering of NPs facilitates the exploitation of active lymphatic transport pathways of cargos, which utilize the intrinsic lymph-homing ability of peripheral APCs. In addition to NPs, HGs that release drugs in a sustained manner have demonstrated benefits such as improved, efficient, and prolonged lymphatic accumulation of drugs, avoidance of repeated administrations, and achievement of dose sparing, thereby leading to efficient anticancer immunotherapy. However, the mechanisms associated with sustained locoregional release and lymphatic drainage need to be further addressed. In particular, hybrid NP and HG systems have been explored as a potential platform to combine the benefits of both NPs and HGs.
There are several issues to be addressed in locoregional anticancer immunotherapy targeting LNs. First, several tumors lack tertiary lymphoid structures [175,176,177,178] where are primary target tissues of locoregional anticancer immunotherapy to instruct and expand effector immune cells. Second, although locoregional delivery allows immune responses specific to the tumors and dLNs and mitigates the systemic immune-related adverse toxicity, it is unclear whether it is beneficial to the treatment of metastatic tumors, compared to systemic delivery to allow the wide distribution of cargos to various LNs presented throughout the body [179,180,181]. Therefore, innovations in the design of NPs and HGs for lymphatic delivery and further investigations to address the issues would maximize therapeutic effectiveness, improve patient compliance, and increase safety in the clinic through locoregional anticancer immunotherapy.

Author Contributions

Investigation and writing—original draft preparation, K.J.C. and J.K., writing—review and editing, and validation, Y.-E.C. and J.K., and supervision, J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

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. Nam, J.; Son, S.; Park, K.S.; Zou, W.; Shea, L.D.; Moon, J.J. Cancer nanomedicine for combination cancer immunotherapy. Nat. Rev. Mater. 2019, 4, 398–414. [Google Scholar] [CrossRef]
  2. Zhang, J.; Huang, D.; Saw, P.E.; Song, E. Turning cold tumors hot: From molecular mechanisms to clinical applications. Trends Immunol. 2022, 43, 524–545. [Google Scholar] [CrossRef] [PubMed]
  3. Francis, D.M.; Thomas, S.N. Progress and opportunities for enhancing the delivery and efficacy of checkpoint inhibitors for cancer immunotherapy. Adv. Drug Deliv. Rev. 2017, 114, 33–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kim, J.; Manspeaker, M.P.; Thomas, S.N. Augmenting the synergies of chemotherapy and immunotherapy through drug delivery. Acta Biomater. 2019, 88, 1–4. [Google Scholar] [CrossRef] [PubMed]
  5. Manspeaker, M.P.; O’Melia, M.J.; Thomas, S.N. Elicitation of stem-like CD8+ T cell responses via lymph node-targeted chemoimmunotherapy evokes systemic tumor control. J. Immunother. Cancer 2022, 10, e005079. [Google Scholar] [CrossRef] [PubMed]
  6. Fucikova, J.; Kepp, O.; Kasikova, L.; Petroni, G.; Yamazaki, T.; Liu, P.; Zhao, L.; Spisek, R.; Kroemer, G.; Galluzzi, L. Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis. 2020, 11, 1013. [Google Scholar] [CrossRef] [PubMed]
  7. Liu, J.; Zhao, Z.; Qiu, N.; Zhou, Q.; Wang, G.; Jiang, H.; Piao, Y.; Zhou, Z.; Tang, J.; Shen, Y. Co-delivery of IOX1 and doxorubicin for antibodyindependent cancer chemo-immunotherapy. Nat. Commun. 2021, 12, 2425. [Google Scholar] [CrossRef] [PubMed]
  8. Lu, J.; Liu, X.; Liao, Y.-P.; Salazar, F.; Sun, B.; Jiang, W.; Chang, C.H.; Jiang, J.; Wang, X.; Wu, A.M.; et al. Nano-enabled pancreas cancer immunotherapy using immunogenic cell death and reversing immunosuppression. Nat. Commun. 2017, 8, 1811. [Google Scholar] [CrossRef] [Green Version]
  9. Liu, P.; Zhao, L.; Pol, J.; Levesque, S.; Petrazzuolo, A.; Pfirschke, C.; Engblom, C.; Rickelt, S.; Yamazaki, T.; Iribarren, K.; et al. Crizotinib-induced immunogenic cell death in non-small cell lung cancer. Nat. Commun. 2019, 10, 1486. [Google Scholar] [CrossRef] [Green Version]
  10. Imbert, C.; Montfort, A.; Fraisse, M.; Marcheteau, E.; Gilhodes, J.; Martin, E.; Bertrand, F.; Marcellin, M.; Burlet-Schiltz, O.; de Peredo, A.G.; et al. Resistance of melanoma to immune checkpoint inhibitors is overcome by targeting the sphingosine kinase-1. Nat. Commun. 2020, 11, 437. [Google Scholar] [CrossRef]
  11. Rivas, E.I.; Linares, J.; Zwick, M.; Gomez-Llonin, A.; Guiu, M.; Labernadie, A.; Badia-Ramentol, J.; Llado, A.; Bardia, L.; Perez-Nunez, I.; et al. Targeted immunotherapy against distinct cancer-associated fibroblasts overcomes treatment resistance in refractory HER2+ breast tumors. Nat. Commun. 2022, 13, 5310. [Google Scholar] [CrossRef] [PubMed]
  12. Ma, S.; Zhao, Y.; Lee, W.C.; Ong, L.-T.; Lee, P.L.; Jiang, Z.; Oguz, G.; Niu, Z.; Liu, M.; Goh, J.Y.; et al. Hypoxia induces HIF1α-dependent epigenetic vulnerability in triple negative breast cancer to confer immune effector dysfunction and resistance to anti-PD-1 immunotherapy. Nat. Commun. 2022, 13, 4118. [Google Scholar] [CrossRef] [PubMed]
  13. Deng, Y.; Xia, X.; Zhao, Y.; Zhao, Z.; Martinez, C.; Yin, W.; Yao, J.; Hang, Q.; Wu, W.; Zhang, J.; et al. Glucocorticoid receptor regulates PD-L1 and MHCI in pancreatic cancer cells to promote immune evasion and immunotherapy resistance. Nat. Commun. 2021, 12, 7041. [Google Scholar] [CrossRef] [PubMed]
  14. Kang, T.H.; Park, J.H.; Yang, A.; Park, H.J.; Lee, S.E.; Kim, Y.S.; Jang, G.-Y.; Farmer, E.; Lam, B.; Park, Y.-M.; et al. Annexin A5 as an immune checkpoint inhibitor and tumor-homing molecule for cancer treatment. Nat. Commun. 2020, 11, 1137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Maeda, H. Toward a full understanding of the EPR effect in primary and metastatic tumors as well as issues related to its heterogeneity. Adv. Drug Deliver. Rev. 2015, 91, 3–6. [Google Scholar] [CrossRef] [PubMed]
  16. Musetti, S.; Huang, L. Nanoparticle-Mediated Remodeling of the Tumor Microenvironment to Enhance Immunotherapy. ACS Nano 2018, 12, 11740–11755. [Google Scholar] [CrossRef]
  17. Yang, F.; Shi, K.; Jia, Y.-P.; Hao, Y.; Peng, J.-R.; Qian, Z.-Y. Advanced biomaterials for cancer immunotherapy. Acta Pharmacol. Sin. 2020, 41, 911–927. [Google Scholar] [CrossRef]
  18. Kim, J.; Archer, P.A.; Thomas, S.N. Innovations in lymph node targeting nanocarriers. Semin. Immunol. 2021, 56, 101534. [Google Scholar] [CrossRef]
  19. Schudel, A.; Francis, D.M.; Thomas, S.N. Material design for lymph node drug delivery. Nat. Rev. Mater. 2019, 4, 415–428. [Google Scholar] [CrossRef]
  20. Manspeaker, M.P.; Thomas, S.N. Lymphatic immunomodulation using engineered drug delivery systems for cancer immunotherapy. Adv. Drug Deliver. Rev. 2020, 160, 19–35. [Google Scholar] [CrossRef]
  21. Thomas, S.N.; Rohner, N.A.; Edwards, E.E. Implications of Lymphatic Transport to Lymph Nodes in Immunity and Immunotherapy. Annu. Rev. Biomed. Eng. 2016, 18, 207–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Sestito, L.F.; Thomas, S.N. Biomaterials for Modulating Lymphatic Function in Immunoengineering. ACS Pharmacol. Transl. Sci. 2019, 2, 293–310. [Google Scholar] [CrossRef] [PubMed]
  23. Cai, S.; Yang, Q.; Bagby, T.R.; Forrest, L. Lymphatic drug delivery using engineered liposomes and solid lipid nanoparticles. Adv. Drug Deliver Rev. 2011, 63, 901–908. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ravizzini, G.; Turkbey, B.; Barrett, T.; Kobayashi, H.; Choyke, P.L. Nanoparticles in sentinel lymph node mapping. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2009, 1, 610–623. [Google Scholar] [CrossRef]
  25. Niu, G.; Chen, X. Lymphatic Imaging: Focus on Imaging Probes. Theranostics 2015, 5, 686–697. [Google Scholar] [CrossRef] [Green Version]
  26. Hameed, S.; Chen, H.; Irfan, M.; Bajwa, S.Z.; Khan, W.S.; Baig, S.M.; Dai, Z. Fluorescence Guided Sentinel Lymph Node Mapping: From Current Molecular Probes to Future Multimodal Nanoprobes. Bioconjugate Chem. 2019, 30, 13–28. [Google Scholar] [CrossRef]
  27. Jiang, H.; Wang, Q.; Sun, X. Lymph node targeting strategies to improve vaccination efficacy. J. Control. Release 2017, 267, 47–56. [Google Scholar] [CrossRef]
  28. Trac, N.; Chung, E.J. Overcoming physiological barriers by nanoparticles for intravenous drug delivery to the lymph nodes. Exp. Biol. Med. 2021, 246, 2358–2371. [Google Scholar] [CrossRef]
  29. Gutjahr, A.; Phelip, C.; Collen, A.-L.; Monge, C.; Boisgard, A.-S.; Paul, S.; Verrier, B. Biodegradable Polymeric Nanoparticles-Based Vaccine Adjuvants for Lymph Nodes Targeting. Vaccines 2016, 4, 34. [Google Scholar] [CrossRef] [Green Version]
  30. Sahdev, P.; Ochyl, L.J.; Moon, J.J. Biomaterials for Nanoparticle Vaccine Delivery Systems. Pharm. Res. 2014, 31, 2563–2582. [Google Scholar] [CrossRef]
  31. Francis, D.M.; Manspeaker, M.P.; Schudel, A.; Sestito, L.F.; O’Melia, M.J.; Kissick, H.T.; Pollack, B.P.; Waller, E.K.; Thomas, S.N. Blockade of immune checkpoints in lymph nodes through locoregional delivery augments cancer immunotherapy. Sci. Transl. Med. 2020, 12, eaay3575. [Google Scholar] [CrossRef]
  32. Yu, S.; Wang, C.; Yu, J.; Wang, J.; Lu, Y.; Zhang, Y.; Zhang, X.; Quanyin, H.; Sun, W.; He, C.; et al. Injectable Bioresponsive Gel Depot for Enhanced Immune Checkpoint Blockade. Adv. Mater. 2018, 30, 1801527. [Google Scholar] [CrossRef] [PubMed]
  33. Ruan, H.; Hu, Q.; Wen, D.; Chen, Q.; Chen, G.; Lu, Y.; Wang, J.; Cheng, H.; Lu, W.; Gu, Z. A Dual-Bioresponsive Drug-Delivery Depot for Combination of Epigenetic Modulation and Immune Checkpoint Blockade. Adv. Mater. 2019, 31, 1806957. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, L.; Zhou, J.; Hu, L.; Han, X.; Zou, X.; Chen, Q.; Chen, Y.; Liu, Z.; Wang, C. In Situ Formed Fibrin Scaffold with Cyclophosphamide to Synergize with Immune Checkpoint Blockade for Inhibition of Cancer Recurrence after Surgery. Adv. Funct. Mater. 2019, 30, 1906922. [Google Scholar] [CrossRef]
  35. Kim, J.; Francis, D.M.; Thomas, S.N. In Situ Crosslinked Hydrogel Depot for Sustained Antibody Release Improves Immune Checkpoint Blockade Cancer Immunotherapy. Nanomaterials 2021, 11, 471. [Google Scholar] [CrossRef]
  36. Leach, D.G.; Young, S.; Hartgerink, J.D. Advances in immunotherapy delivery from implantable and injectable biomaterials. Acta Biomater. 2019, 88, 15–31. [Google Scholar] [CrossRef]
  37. Cai, L.; Xu, J.; Yang, Z.; Tong, R.; Dong, Z.; Wang, C.; Leong, K.W. Engineered biomaterials for cancer immunotherapy. MeComm 2020, 1, 35–46. [Google Scholar] [CrossRef]
  38. Kim, J.; Choi, Y.; Kim, D.-H.; Yoon, H.Y.; Kim, K. Injectable Hydrogel-Based Combination Cancer Immunotherapy for Overcoming Localized Therapeutic Efficacy. Pharmaceutics 2022, 14, 1908. [Google Scholar] [CrossRef]
  39. Swartz, M.A.; Fleury, M.E. Interstitial Flow and Its Effects in Soft Tissues. Annu. Rev. Biomed. Eng. 2007, 9, 229–256. [Google Scholar] [CrossRef] [Green Version]
  40. Rohner, N.A.; Thomas, S.N. Flexible Macromolecule versus Rigid Particle Retention in the Injected Skin and Accumulation in Draining Lymph Nodes Are Differentially Influenced by Hydrodynamic Size. ACS Biomater. Sci. Eng. 2017, 3, 153–159. [Google Scholar] [CrossRef]
  41. Rohner, N.A.; Thomas, S.N. Melanoma growth effects on molecular clearance from tumors and biodistribution into systemic tissues versus draining lymph nodes. J. Control. Release 2016, 223, 99–108. [Google Scholar] [CrossRef] [Green Version]
  42. O’Melia, M.J.; Rohner, N.A.; Manspeaker, M.P.; Francis, D.M.; Kissick, H.T.; Thomas, S.N. Quality of CD8+ T cell immunity evoked in lymph nodes is compartmentalized by route of antigen transport and functional in tumor context. Sci. Adv. 2020, 6, eabd7134. [Google Scholar] [CrossRef]
  43. Reddy, S.T.; Berk, D.A.; Jain, R.K.; Swartz, M.A. A sensitive in vivo model for quantifying interstitial convective transport of injected macromolecules and nanoparticles. J. Appl. Physiol. 2006, 101, 1162–1169. [Google Scholar] [CrossRef] [Green Version]
  44. Archer, P.A.; Sestito, L.F.; Manspeaker, M.P.; O’Melia, M.J.; Rohner, N.A.; Schudel, A.; Mei, Y.; Thomas, S.N. Quantitation of lymphatic transport mechanism and barrier influences on lymph node-resident leukocyte access to lymph-borne macromolecules and drug delivery systems. Drug Deliv. Transl. Res. 2021, 11, 2328–2343. [Google Scholar] [CrossRef]
  45. Howard, G.P.; Verma, G.; Ke, X.; Thayer, W.M.; Hamerly, T.; Baxter, V.K.; Lee, J.E.; Dinglasan, R.R.; Mao, H.-Q. Critical size limit of biodegradable nanoparticles for enhanced lymph node trafficking and paracortex penetration. Nano Res. 2019, 12, 837–844. [Google Scholar] [CrossRef]
  46. Zhang, Y.-N.; Lazarovits, J.; Poon, W.; Ouyang, B.; Nguyen, L.N.M.; Kingston, B.R.; Chan, W.C.W. Nanoparticle Size Influences Antigen Retention and Presentation in Lymph Node Follicles for Humoral Immunity. Nano Lett. 2019, 19, 7226–7235. [Google Scholar] [CrossRef]
  47. Nakamura, T.; Kawai, M.; Sato, Y.; Maeki, M.; Tokeshi, M.; Harashima, H. The Effect of Size and Charge of Lipid Nanoparticles Prepared by Microfluidic Mixing on Their Lymph Node Transitivity and Distribution. Mol. Pharm. 2020, 17, 944–953. [Google Scholar] [CrossRef]
  48. Wu, X.; Li, Y.; Chen, X.; Zhou, Z.; Pang, J.; Luo, X.; Kong, M. A surface charge dependent enhanced Th1 antigen-specific immune response in lymph nodes by transfersome-based nanovaccine-loaded dissolving microneedle-assisted transdermal immunization. J. Mater. Chem. B 2019, 7, 4854–4866. [Google Scholar] [CrossRef]
  49. Baek, J.-S.; Cho, C.-W. Surface modification of solid lipid nanoparticles for oral delivery of curcumin: Improvement of bioavailability through enhanced cellular uptake, and lymphatic uptake. Eur. J. Pharm. Biopharm. 2017, 117, 132–140. [Google Scholar] [CrossRef]
  50. Nishimoto, Y.; Nagashima, S.; Nakajima, K.; Ohira, T.; Sato, T.; Izawa, T.; Yamate, J.; Higashikawa, K.; Kuge, Y.; Ogawa, M.; et al. Carboxyl-, sulfonyl-, and phosphate-terminal dendrimers as a nanoplatform with lymph node targeting. Int. J. Pharm. 2020, 576, 119021. [Google Scholar] [CrossRef]
  51. Knop, K.; Hoogenboom, R.; Fischer, D.; Schubert, U.S. Poly(ethylene glycol) in Drug Delivery: Pros and Cons as Well as Potential Alternatives. Angew. Chem. Int. Ed. 2010, 49, 6288–6308. [Google Scholar] [CrossRef]
  52. Tomasetti, L.; Liebl, R.; Wastl, D.S.; Breunig, M. Influence of PEGylation on nanoparticle mobility in different models of the extracellular matrix. Eur. J. Pharm. Biopharm. 2016, 108, 145–155. [Google Scholar] [CrossRef]
  53. Mueller, S.N.; Tian, S.; DeSimone, J.M. Rapid and Persistent Delivery of Antigen by Lymph Node Targeting PRINT Nanoparticle Vaccine Carrier To Promote Humoral Immunity. Mol. Pharm. 2015, 12, 1356–1365. [Google Scholar] [CrossRef] [Green Version]
  54. Zukancic, D.; Suys, E.J.; Pilkington, E.H.; Algarni, A.; Al-Wassiti, H.; Truong, N.P. The Importance of Poly(ethylene glycol) and Lipid Structure in Targeted Gene Delivery to Lymph Nodes by Lipid Nanoparticles. Pharmaceutics 2020, 12, 1068. [Google Scholar] [CrossRef]
  55. McCright, J.; Skeen, C.; Yarmovsky, J.; Maisel, K. Nanoparticles with dense poly(ethylene glycol) coatings with near neutral charge are maximally transported across lymphatics and to the lymph nodes. Acta Biomater. 2022, 145, 146–158. [Google Scholar] [CrossRef]
  56. Kim, D.; Wu, Y.; Shim, G.; Oh, Y.-K. Lipid Nanoparticle-Mediated Lymphatic Delivery of Immunostimulatory Nucleic Acids. Pharmaceutics 2021, 13, 490. [Google Scholar] [CrossRef]
  57. Uthaman, S.; Kim, H.S.; Revuri, V.; Min, J.-J.; Lee, Y.-K.; Huh, K.M.; Park, I.-K. Green synthesis of bioactive polysaccharide-capped gold nanoparticles for lymph node CT imaging. Carbohydr. Polym. 2018, 181, 27–33. [Google Scholar] [CrossRef]
  58. Vu-Quang, H.; Yoo, M.-K.; Jeong, H.-J.; Lee, H.-J.; Muthiah, M.; Rhee, J.H.; Lee, J.-H.; Cho, C.-S.; Jeong, Y.Y.; Park, I.-K. Targeted delivery of mannan-coated superparamagnetic iron oxide nanoparticles to antigen-presenting cells for magnetic resonance-based diagnosis of metastatic lymph nodes in vivo. Acta Biomater. 2011, 7, 3935–3945. [Google Scholar] [CrossRef]
  59. Vu-Quang, H.; Muthiah, M.; Kim, Y.-K.; Cho, C.-S.; Namgung, R.; Kim, W.J.; Rhee, J.H.; Kang, S.H.; Jun, S.Y.; Choi, Y.-J.; et al. Carboxylic mannan-coated iron oxide nanoparticles targeted to immune cells for lymph node-specific MRI in vivo. Carbohydr. Polym. 2012, 88, 780–788. [Google Scholar] [CrossRef]
  60. Muthiah, M.; Vu-Quang, H.; Kim, Y.-K.; Rhee, J.H.; Kang, S.H.; Jun, S.Y.; Choi, Y.-J.; Jeong, Y.Y.; Cho, C.-S.; Park, I.-K. Mannose-poly(ethylene glycol)-linked SPION targeted to antigen presenting cells for magnetic resonance imaging on lymph node. Carbohydr. Polym. 2013, 92, 1586–1595. [Google Scholar] [CrossRef]
  61. Son, S.; Nam, J.; Zenkov, I.; Ochyl, L.J.; Xu, Y.; Scheetz, L.; Shi, J.; Farokhzad, O.C.; Moon, J.J. Sugar-Nanocapsules Imprinted with Microbial Molecular Patterns for mRNA Vaccination. Nano Lett. 2020, 20, 1499–1509. [Google Scholar] [CrossRef]
  62. Liang, X.; Li, X.; Duan, J.; Chen, Y.; Wang, X.; Pang, L.; Kong, D.; Song, B.; Li, C.; Yang, J. Nanoparticles with CD44 Targeting and ROS Triggering Properties as Effective in Vivo Antigen Delivery System. Mol. Pharm. 2018, 15, 508–518. [Google Scholar] [CrossRef]
  63. Kim, S.-Y.; Heo, M.B.; Hwang, G.-S.; Jung, Y.; Choi, D.Y.; Park, Y.-M.; Lim, Y.T. Multivalent Polymer Nanocomplex Targeting Endosomal Receptor of Immune Cells for Enhanced Antitumor and Systemic Memory Response. Angew. Chem. 2015, 127, 8257–8261. [Google Scholar] [CrossRef]
  64. Rosalia, R.A.; Cruz, L.J.; van Duikeren, S.; Tromp, A.T.; Silva, A.L.; Jiskoot, W.; de Gruijl, T.; Lowik, C.; Oostendorp, J.; van der Burg, S.H.; et al. CD40-targeted dendritic cell delivery of PLGA-nanoparticle vaccines induce potent anti-tumor responses. Biomaterials 2015, 40, 88–97. [Google Scholar] [CrossRef]
  65. Baekkevold, E.S.; Yamanaka, T.; Palframan, R.T.; Carlsen, H.S.; Reinholt, F.P.; von Andrian, U.H.; Brandtzaeg, P.; Haraldsen, G. The CCR7 Ligand ELC (CCL19) Is Transcytosed in High Endothelial Venules and Mediates T Cell Recruitment. J. Exp. Med. 2001, 193, 1105–1111. [Google Scholar] [CrossRef] [Green Version]
  66. Palframan, R.T.; Jung, S.; Cheng, G.; Weninger, W.; Luo, Y.; Dorf, M.; Littman, D.R.; Rollins, B.J.; Zweerink, H.; Rot, A.; et al. Inflammatory Chemokine Transport and Presentation in HEV: A Remote Control Mechanism for Monocyte Recruitment to Lymph Nodes in Inflamed Tissues. J. Exp. Med. 2001, 194, 1361–1373. [Google Scholar] [CrossRef] [Green Version]
  67. Bahmani, B.; Uehara, M.; Jiang, L.; Ordikhani, F.; Banouni, N.; Ichimura, T.; Solhjou, Z.; Furtmuller, G.J.; Brandacher, G.; Alvarez, D.; et al. Targeted delivery of immune therapeutics to lymph nodes prolongs cardiac allograft survival. J. Clin. Investig. 2018, 128, 4770–4786. [Google Scholar] [CrossRef]
  68. Ager, A. High endothelial venules and Other Blood vessels: Critical Regulators of Lymphoid Organ Development and Function. Front. Immunol. 2017, 8, 45. [Google Scholar] [CrossRef]
  69. Yoneyama, H.; Matsuno, K.; Zhang, Y.; Nishiwaki, T.; Kitabatake, M.; Ueha, S.; Narumi, S.; Morikawa, S.; Ezaki, T.; Lu, B.; et al. Evidence for recruitment of plasmacytoid dendritic cell precursors to inflamed lymph nodes through high endothelial venules. Int. Immunol. 2004, 16, 915–928. [Google Scholar] [CrossRef]
  70. Jiang, L.; Jung, S.; Zhao, J.; Kasinath, V.; Ichimura, T.; Joseph, J.; Fiorina, P.; Liss, A.S.; Shah, K.; Annabi, N.; et al. Simultaneous targeting of primary tumor, draining lymph node, and distant metastases through high endothelial venule-targeted delivery. Nano Today 2021, 36, 101045. [Google Scholar] [CrossRef]
  71. Azzi, J.; Yin, Q.; Uehara, M.; Ohori, S.; Tang, L.; Cai, K.; Ichimura, T.; McGrath, M.; Maarouf, O.; Kefaloyianni, E.; et al. Targeted Delivery of Immunomodulators to Lymph Nodes. Cell Rep. 2016, 15, 1202–1213. [Google Scholar] [CrossRef] [Green Version]
  72. Bahmani, B.; Uehara, M.; Ordikhani, F.; Li, X.; Jiang, L.; Banouni, N.; Ichimura, T.; Kasinath, V.; Eskandari, S.K.; Annabi, N.; et al. Ectopic high endothelial venules in pancreatic ductal adenocarcinoma: A unique site for targeted delivery. EBioMedicine 2018, 38, 79–99. [Google Scholar] [CrossRef] [Green Version]
  73. Roozendaal, R.; Mempel, T.R.; Pitcher, L.A.; Gonzalez, S.F.; Verschoor, A.; Mebius, R.E.; von Andrian, U.H.; Carroll, M.C. Conduits Mediate Transport of Low-Molecular-Weight Antigen to Lymph Node Follicles. Immunity 2009, 30, 264–276. [Google Scholar] [CrossRef] [Green Version]
  74. De Koker, S.; Cui, J.; Vanparijs, N.; Albertazzi, L.; Grooten, J.; Caruso, F.; De Geest, B.G. Engineering Polymer Hydrogel Nanoparticles for Lymph Node-Targeted Delivery. Angew. Chem. 2016, 128, 1356–1361. [Google Scholar] [CrossRef] [Green Version]
  75. Klechevsky, E.; Morita, R.; Liu, M.; Cao, Y.; Coquery, S.; Thompson-Snipes, L.; Briere, F.; Chaussabel, D.; Zurawski, G.; Palucka, A.K.; et al. Functional Specializations of Human Epidermal Langerhans Cells and CD14+ Dermal Dendritic Cells. Immunity 2008, 29, 497–510. [Google Scholar] [CrossRef] [Green Version]
  76. Chandrasekaran, S.; Chan, M.F.; Li, J.; King, M.R. Super natural killer cells that target metastases in the tumor draining lymph nodes. Biomaterials 2016, 77, 66–76. [Google Scholar] [CrossRef] [Green Version]
  77. Schmid, D.; Park, C.G.; Hartl, C.A.; Subedi, N.; Cartwright, A.N.; Bou Puerto, R.; Zheng, Y.; Maiarana, J.; Freeman, G.J.; Wucherpfennig, K.W.; et al. T cell-targeting nanoparticles focus delivery of immunotherapy to improve antitumor immunity. Nat. Commun. 2017, 8, 1747. [Google Scholar] [CrossRef] [Green Version]
  78. Zhang, X.; Wang, C.; Wang, J.; Hu, Q.; Longworthy, B.; Ye, Y.; Sun, W.; Lin, J.; Wang, T.; Fine, J.; et al. PD-1 Blockade Cellular Vesicles for Cancer Immunotherapy. Adv. Mater. 2018, 30, 1707112. [Google Scholar] [CrossRef]
  79. Kataoka, K.; Harada, A.; Nagasaki, Y. Block copolymer micelles for drug delivery: Design, characterization and biological significance. Adv. Drug Deliver. Rev. 2012, 64, 37–48. [Google Scholar] [CrossRef]
  80. Thomas, S.N.; Vokali, E.; Lund, A.W.; Hubbell, J.A.; Swartz, M.A. Targeting the tumor-draining lymph node with adjuvanted nanoparticles reshapes the anti-tumor immune response. Biomaterials 2014, 35, 814–824. [Google Scholar] [CrossRef]
  81. Kim, J.; Sestito, L.F.; Im, S.; Kim, W.J.; Thomas, S.N. Poly(cyclodextrin)-Polydrug Nanocomplexes as Synthetic Oncolytic Virus for Locoregional Melanoma Chemoimmunotherapy. Adv. Funct. Mater. 2020, 30, 1908788. [Google Scholar] [CrossRef]
  82. Horejs, C. From lipids to lipid nanoparticles to mRNA vaccines. Nat. Rev. Mater. 2021, 6, 1075–1076. [Google Scholar] [CrossRef]
  83. 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 potent lymph node–targeted vaccine adjuvants. J. Clin. Investig. 2015, 125, 2532–2546. [Google Scholar] [CrossRef] [Green Version]
  84. Oberli, M.A.; Reichmuth, A.M.; Dorkin, J.R.; Mitchell, M.J.; Fenton, O.S.; Jaklenec, A.; Anderson, D.G.; Langer, R.; Blankschtein, D. Lipid Nanoparticle Assisted mRNA Delivery for Potent Cancer Immunotherapy. Nano Lett. 2017, 17, 1326–1335. [Google Scholar] [CrossRef]
  85. Bourquin, C.; Anz, D.; Zwiorek, K.; Lanz, A.-L.; Fuchs, S.; Wigel, S.; Wurzenberger, C.; von der Borch, P.; Golic, M.; Moder, S.; et al. Targeting CpG Oligonucleotides to the Lymph Node by Nanoparticles Elicits Efficient Antitumoral Immunity. J. Immunol. 2008, 181, 2990–2998. [Google Scholar] [CrossRef] [Green Version]
  86. Zeng, Q.; Li, H.; Jiang, H.; Yu, J.; Wang, Y.; Ke, H.; Gong, T.; Zhang, Z.; Sun, X. Tailoring polymeric hybrid micelles with lymph node targeting ability to improve the potency of cancer vaccines. Biomaterials 2017, 122, 105–113. [Google Scholar] [CrossRef]
  87. Kim, S.-Y.; Noh, Y.-W.; Kang, T.H.; Kim, J.-E.; Kim, S.; Um, S.H.; Oh, D.-B.; Park, Y.-M.; Lim, Y.T. Synthetic vaccine nanoparticles target to lymph node triggering enhanced innate and adaptive antitumor immunity. Biomaterials 2017, 130, 56–66. [Google Scholar] [CrossRef]
  88. An, M.; Li, M.; Xi, J.; Liu, H. Silica Nanoparticle as a Lymph Node Targeting Platform for Vaccine Delivery. ACS Appl. Mater. Interfaces 2017, 9, 23466–23475. [Google Scholar] [CrossRef]
  89. Lu, Y.; Yang, Y.; Gu, Z.; Zhang, J.; Song, H.; Xiang, G.; Yu, C. Glutathione-depletion mesoporous organosilica nanoparticles as a self-adjuvant and Co-delivery platform for enhanced cancer immunotherapy. Biomaterials 2018, 175, 82–92. [Google Scholar] [CrossRef]
  90. Jiang, H.; Wang, Q.; Li, L.; Zeng, Q.; Li, H.; Gong, T.; Zhang, Z.; Sun, X. Turning the Old Adjuvant from Gel to Nanoparticles to Amplify CD8+ T Cell Responses. Adv. Sci. 2018, 5, 1700426. [Google Scholar] [CrossRef]
  91. Mody, K.T.; Popat, A.; Mahony, D.; Cavallaro, A.S.; Yu, C.; Mitter, N. Mesoporous silica nanoparticles as antigen carriers and adjuvants for vaccine delivery. Nanoscale 2013, 5, 5167–5179. [Google Scholar] [CrossRef]
  92. Mok, H.; Lee, S.H.; Park, J.W.; Park, T.G. Multimeric small interfering ribonucleic acid for highly efficient sequence-specific gene silencing. Nat. Mater. 2010, 9, 272–278. [Google Scholar] [CrossRef]
  93. Zhu, G.; Mei, L.; Vishwasrao, H.D.; Jacobson, O.; Wang, Z.; Liu, Y.; Yung, B.C.; Fu, X.; Jin, A.; Niu, G.; et al. Intertwining DNA-RNA nanocapsules loaded with tumor neoantigens as synergistic nanovaccines for cancer immunotherapy. Nat. Commun. 2017, 8, 1482. [Google Scholar] [CrossRef] [Green Version]
  94. Ni, Q.; Zhang, F.; Liu, Y.; Wang, Z.; Yu, G.; Liang, B.; Niu, G.; Su, T.; Zhu, G.; Lu, G.; et al. A bi-adjuvant nanovaccine that potentiates immunogenicity of neoantigen for combination immunotherapy of colorectal cancer. Sci. Adv. 2020, 6, eaaw6071. [Google Scholar] [CrossRef] [Green Version]
  95. Sun, X.; Liu, H. Nucleic Acid Nanostructure Assisted Immune Modulation. ACS Appl. Bio Mater. 2020, 3, 2765–2778. [Google Scholar] [CrossRef]
  96. Cutler, J.I.; Auyeung, E.; Mirkin, C.A. Spherical Nucleic Acids. J. Am. Chem. Soc. 2012, 134, 1376–1391. [Google Scholar] [CrossRef]
  97. Radovic-Moreno, A.F.; Chernyak, N.; Mader, C.C.; Nallagatla, S.; Kang, R.S.; Hao, L.; Walker, D.A.; Halo, T.L.; Merkel, T.J.; Rische, C.H.; et al. Immunomodulatory spherical nucleic acids. Proc. Natl. Acad. Sci. USA 2015, 112, 3892–3897. [Google Scholar] [CrossRef] [Green Version]
  98. Callmann, C.E.; Kusmeierz, C.D.; Dittmar, J.W.; Broger, L.; Mirkin, C.A. Impact of Liposomal Spherical Nucleic Acid Structure on Immunotherapeutic Function. ACS Cent. Sci. 2021, 7, 892–899. [Google Scholar] [CrossRef]
  99. Kuai, R.; Ochyl, L.J.; Bahjat, K.S.; Schwendeman, A.; Moon, J.J. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nat. Mater. 2017, 16, 489–496. [Google Scholar] [CrossRef]
  100. Callmann, C.E.; Cole, L.E.; Kusmierz, C.D.; Huang, Z.; Horiuchi, D.; Mirkin, C.A. Tumor cell lysate-loaded immunostimulatory spherical nucleic acids as therapeutics for triple-negative breast cancer. Proc. Natl. Acad. Sci. USA 2020, 117, 17543–17550. [Google Scholar] [CrossRef]
  101. Chen, P.; Wang, D.; Wang, Y.; Zhang, L.; Wang, Q.; Liu, L.; Li, J.; Sun, X.; Ren, M.; Wang, R.; et al. Maximizing TLR9 Activation in Cancer Immunotherapy with DualAdjuvanted Spherical Nucleic Acids. Nano Lett. 2022, 22, 4058–4066. [Google Scholar] [CrossRef]
  102. Wang, P.; Meyer, T.A.; Pan, V.; Dutta, P.K.; Ke, Y. The Beauty and Utility of DNA Origami. Chem 2017, 2, 359–382. [Google Scholar] [CrossRef] [Green Version]
  103. Kim, K.-R.; Lee, Y.-D.; Lee, T.; Kim, B.-S.; Kim, S.; Ahn, D.-R. Sentinel lymph node imaging by a fluorescently labeled DNA tetrahedron. Biomaterials 2013, 34, 5226–5235. [Google Scholar] [CrossRef]
  104. Liu, S.; Jiang, Q.; Zhao, X.; Zhao, R.; Wang, Y.; Wang, Y.; Liu, J.; Shang, Y.; Zhao, S.; Wu, T.; et al. A DNA nanodevice-based vaccine for cancer immunotherapy. Nat. Mater. 2021, 20, 421–430. [Google Scholar] [CrossRef]
  105. Xi, X.; Zhang, L.; Lu, G.; Gao, X.; Wei, W.; Ma, G. Lymph Node-Targeting Nanovaccine through Antigen-CpG Self-Assembly Potentiates Cytotoxic T Cell Activation. J. Immunol. Res. 2018, 2018, 3714960. [Google Scholar] [CrossRef]
  106. Zhao, H.; Xu, J.; Li, Y.; Guan, X.; Han, X.; Xu, Y.; Zhou, H.; Peng, R.; Wang, J.; Liu, Z. Nanoscale Coordination Polymer Based Nanovaccine for Tumor Immunotherapy. ACS Nano 2019, 13, 13127–13135. [Google Scholar] [CrossRef]
  107. Lee, B.-R.; Ko, H.K.; Ryu, J.H.; Ahn, K.Y.; Lee, Y.-H.; Oh, S.J.; Na, J.H.; Kim, T.W.; Byun, Y.; Kwon, I.C.; et al. Engineered Human Ferritin Nanoparticles for Direct Delivery of Tumor Antigens to Lymph Node and Cancer Immunotherapy. Sci. Rep. 2016, 6, 35182. [Google Scholar] [CrossRef] [Green Version]
  108. Kim, G.B.; Sung, H.-D.; Nam, G.-H.; Kim, W.; Kim, S.; Kang, D.; Lee, E.J.; Kim, I.-S. Design of PD-1-decorated nanocages targeting tumor-draining lymph node for promoting T cell activation. J. Control. Release 2021, 333, 328–338. [Google Scholar] [CrossRef]
  109. Kim, J.H.; Noh, Y.-W.; Heo, M.B.; Cho, M.Y.; Lim, Y.T. Multifunctional Hybrid Nanoconjugates for Efficient In Vivo Delivery of Immunomodulating Oligonucleotides and Enhanced Antitumor Immunity. Angew. Chem. Int. Ed. 2012, 51, 9670–9673. [Google Scholar] [CrossRef]
  110. Ruiz-de-Angulo, A.; Zabaleta, A.; Gomez-Vallejo, V.; Llop, J.; Mareque-Rivas, J.C. Microdosed Lipid-Coated 67 Ga-Magnetite Enhances Antigen-Specific Immunity by Image Tracked Delivery of Antigen and CpG to Lymph Nodes. ACS Nano 2016, 10, 1602–1618. [Google Scholar] [CrossRef]
  111. Xu, C.; Hong, H.; Lee, Y.; Park, K.S.; Sun, M.; Wang, T.; Aikins, M.E.; Xu, Y.; Moon, J.J. Efficient Lymph Node-Targeted Delivery of Personalized Cancer Vaccines with Reactive Oxygen Species-Inducing Reduced Graphene Oxide Nanosheets. ACS Nano 2020, 14, 13268–13278. [Google Scholar] [CrossRef] [PubMed]
  112. Chu, Y.; Qian, L.; Ke, Y.; Feng, X.; Chen, X.; Liu, F.; Yu, L.; Zhang, L.; Tao, Y.; Xu, R.; et al. Lymph node-targeted neoantigen nanovaccines potentiate anti-tumor immune responses of post-surgical melanoma. J. Nanobiotechnology 2022, 20, 190. [Google Scholar] [CrossRef] [PubMed]
  113. Jiang, D.; Gao, T.; Liang, S.; Mu, W.; Fu, S.; Liu, Y.; Yang, R.; Zhang, Z.; Liu, Y.; Zhang, N. Lymph Node Delivery Strategy Enables the Activation of Cytotoxic T Lymphocytes and Natural Killer Cells to Augment Cancer Immunotherapy. ACS Appl. Mater. Interfaces 2021, 13, 22213–22224. [Google Scholar] [CrossRef]
  114. Shi, G.-N.; Zhang, C.-N.; Xu, R.; Niu, J.-F.; Song, H.-J.; Zhang, X.-Y.; Wang, W.-W.; Wang, Y.-M.; Li, C.; Wei, X.-Q.; et al. Enhanced antitumor immunity by targeting dendritic cells with tumor cell lysate-loaded chitosan nanoparticles vaccine. Biomaterials 2017, 113, 191–202. [Google Scholar] [CrossRef]
  115. Yang, X.; Yu, T.; Zeng, Y.; Lian, K.; Zhou, X.; Li, S.; Qiu, G.; Jin, X.; Yuan, H.; Hu, F. Tumor-draining lymph node targeting chitosan micelles as antigen-capturing adjuvants for personalized immunotherapy. Carbohydr. Polym. 2020, 240, 116270. [Google Scholar] [CrossRef] [PubMed]
  116. Zhang, C.; Shi, G.; Zhang, J.; Song, H.; Niu, J.; Shi, S.; Huang, P.; Wang, Y.; Wang, W.; Li, C.; et al. Targeted antigen delivery to dendritic cell via functionalized alginate nanoparticles for cancer immunotherapy. J. Control. Release 2017, 256, 170–181. [Google Scholar] [CrossRef] [PubMed]
  117. Lv, S.; Song, K.; Yen, A.; Peeler, D.J.; Nguyen, D.C.; Olshefsky, A.; Sylvestre, M.; Srinivasan, S.; Stayton, P.S.; Pun, S.H. Well-Defined Mannosylated Polymer for Peptide Vaccine Delivery with Enhanced Antitumor Immunity. Adv. Healthc. Mater. 2022, 11, 2101651. [Google Scholar] [CrossRef]
  118. Zeng, Z.; Pu, K. Improving Cancer Immunotherapy by Cell Membrane-Camouflaged Nanoparticles. Adv. Funct. Mater. 2020, 30, 2004397. [Google Scholar] [CrossRef]
  119. Guo, Y.; Wang, D.; Song, Q.; Wu, T.; Zhuang, X.; Bao, Y.; Kong, M.; Qi, Y.; Tan, S.; Zhang, Z. Erythrocyte Membrane-Enveloped Polymeric Nanoparticles as Nanovaccine for Induction of Antitumor Immunity against Melanoma. ACS Nano 2015, 9, 6918–6933. [Google Scholar] [CrossRef]
  120. Yang, R.; Xu, J.; Xu, L.; Sun, X.; Chen, Q.; Zhao, Y.; Peng, R.; Liu, Z. Cancer Cell Membrane-Coated Adjuvant Nanoparticles with Mannose Modification for Effective Anticancer Vaccination. ACS Nano 2018, 12, 5121–5129. [Google Scholar] [CrossRef]
  121. Yang, X.; Yu, T.; Zeng, Y.; Lian, K.; Zhou, X.; Ke, J.; Li, Y.; Yuan, H.; Hu, F. pH-Responsive Biomimetic Polymeric Micelles as Lymph NodeTargeting Vaccines for Enhanced Antitumor Immune Responses. Biomacromolecules 2020, 21, 2818–2828. [Google Scholar] [CrossRef]
  122. Xiao, P.; Wang, J.; Zhao, Z.; Liu, X.; Sun, X.; Wang, D.; Li, Y. Engineering Nanoscale Artificial Antigen-Presenting Cells by Metabolic Dendritic Cell Labeling to Potentiate Cancer Immunotherapy. Nano Lett. 2021, 21, 2094–2103. [Google Scholar] [CrossRef] [PubMed]
  123. Liu, W.-L.; Zou, M.-Z.; Liu, T.; Zeng, J.-Y.; Li, X.; Yu, W.-Y.; Li, C.-X.; Ye, J.-J.; Song, W.; Feng, J.; et al. Cytomembrane nanovaccines show therapeutic effects by mimicking tumor cells and antigen presenting cells. Nat. Commun. 2019, 10, 3199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Ma, J.; Liu, F.; Sheu, W.C.; Meng, Z.; Xie, Y.; Xu, H.; Li, M.; Chen, A.T.; Liu, J.; Bao, Y.; et al. Copresentation of Tumor Antigens and Costimulatory Molecules via Biomimetic Nanoparticles for Effective Cancer Immunotherapy. Nano Lett. 2020, 20, 4084–4094. [Google Scholar] [CrossRef] [PubMed]
  125. Herrmann, I.K.; Wood, M.J.A.; Fuhrmann, G. Extracellular vesicles as a next-generation drug delivery platform. Nat. Nanotech. 2021, 16, 748–759. [Google Scholar] [CrossRef] [PubMed]
  126. Phung, C.D.; Pham, T.T.; Nguyen, H.T.; Nguyen, T.T.; Ou, W.; Jeong, J.-H.; Choi, H.-G.; Ku, S.K.; Yong, C.S.; Kim, J.O. Anti-CTLA-4 antibody-functionalized dendritic cell-derived exosomes targeting tumor-draining lymph nodes for effective induction of antitumor T-cell responses. Acta Biomater. 2020, 115, 371–382. [Google Scholar] [CrossRef]
  127. Cheng, L.; Wang, Y.; Huang, L. Exosomes from M1-Polarized Macrophages Potentiate the Cancer Vaccine by Creating a Proinflammatory Microenvironment in the Lymph Node. Mol. Ther. 2017, 25, 1665–1675. [Google Scholar] [CrossRef] [Green Version]
  128. Schudel, A.; Chapman, A.P.; Yau, M.-K.; Higginson, C.J.; Francis, D.M.; Manspeaker, M.P.; Avecilla, A.R.C.; Rohner, N.A.; Finn, M.G.; Thomas, S.N. Programmable multistage drug delivery to lymph nodes. Nat. Nanotech. 2020, 15, 491–499. [Google Scholar] [CrossRef]
  129. Xue, X.; Huang, Y.; Bo, R.; Jia, B.; Wu, H.; Yuan, Y.; Wang, Z.; Ma, Z.; Jing, D.; Xu, X.; et al. Trojan Horse nanotheranostics with dual transformability and multifunctionality for highly effective cancer treatment. Nat. Commun. 2018, 9, 3653. [Google Scholar] [CrossRef] [Green Version]
  130. Li, H.-J.; Du, J.-Z.; Liu, J.; Du, X.-J.; Shen, S.; Zhu, Y.-H.; Wang, X.; Ye, X.; Nie, S.; Wang, J. Smart Superstructures with Ultrahigh pHSensitivity for Targeting Acidic Tumor Microenvironment: Instantaneous Size Switching and Improved Tumor Penetration. ACS Nano 2016, 10, 6753–6761. [Google Scholar] [CrossRef]
  131. Chen, B.; Dai, W.; He, B.; Zhang, H.; Wang, X.; Wang, Y.; Zhang, Q. Current Multistage Drug Delivery Systems Based on the Tumor Microenvironment. Theranostics 2017, 7, 538–558. [Google Scholar] [CrossRef] [PubMed]
  132. Huynh, V.; Jesmer, A.H.; Shoaib, M.M.; D’Angelo, A.D.; Rullo, A.F.; Wylie, R.G. Improved Efficacy of Antibody Cancer Immunotherapeutics through Local and Sustained Delivery. ChemBioChem 2019, 20, 747–753. [Google Scholar] [CrossRef] [PubMed]
  133. Zhou, Y.; Ye, T.; Ye, C.; Wan, C.; Yuan, S.; Liu, Y.; Li, T.; Jiang, F.; Lovell, J.F.; Jin, H.; et al. Secretions from hypochlorous acid-treated tumor cells delivered in a melittin hydrogel potentiate cancer immunotherapy. Bioact. Mater. 2022, 9, 541–553. [Google Scholar] [CrossRef] [PubMed]
  134. Cao, Y.; Zhou, Y.; Pan, J.; Zhong, X.; Ding, J.; Jing, X.; Sun, S.K. A general strategy towards an injectable microwave-sensitive immune hydrogel for combined percutaneous microwave ablation and immunotherapy. Chem. Eng. J. 2021, 422, 130111. [Google Scholar] [CrossRef]
  135. Chen, Q.; Zhou, S.; Ding, Y.; Chen, D.; Dahiru, N.S.; Tang, H.; Xu, H.; Ji, M.; Wang, X.; Li, Z.; et al. A bio-responsive, cargo-catchable gel for postsurgical tumor treatment via ICD-based immunotherapy. J. Control. Release 2022, 346, 212–225. [Google Scholar] [CrossRef]
  136. Jin, H.; Wan, C.; Zou, Z.; Zhao, G.; Zhang, L.; Geng, Y.; Chen, T.; Huang, A.; Jiang, F.; Feng, J.-P.; et al. Tumor Ablation and Therapeutic Immunity Induction by an Injectable Peptide Hydrogel. ACS Nano 2018, 12, 3295–3310. [Google Scholar] [CrossRef]
  137. Yang, F.; Shi, K.; Hao, Y.; Jia, Y.; Liu, Q.; Chen, Y.; Pan, M.; Yuan, L.; Yu, Y.; Qian, Z. Cyclophosphamide loaded thermo-responsive hydrogel system synergize with a hydrogel cancer vaccine to amplify cancer immunotherapy in a prime-boost manner. Bioact. Mater. 2021, 6, 3036–3048. [Google Scholar] [CrossRef]
  138. Lee, J.; Le, Q.-V.; Yang, G.; Oh, Y.-K. Improved Efficacy of Antibody Cancer Immunotherapeutics through Local and Sustained Delivery. Biomaterials 2019, 218, 119359. [Google Scholar] [CrossRef]
  139. He, H.; Fei, Z.; Guo, T.; Hou, Y.; Li, D.; Wang, K.; Ren, F.; Fan, K.; Zhou, D.; Xie, C.; et al. Bioadhesive injectable hydrogel with phenolic carbon quantum dot supported Pd single atom nanozymes as a localized immunomodulation niche for cancer catalytic immunotherapy. Biomaterials 2022, 280, 121272. [Google Scholar] [CrossRef]
  140. Huo, W.; Yang, X.; Wang, B.; Cao, L.; Fang, Z.; Li, Z.; Liu, H.; Liang, X.-j.; Zhang, J.; Jin, Y. Biomineralized hydrogel DC vaccine for cancer immunotherapy: A boosting strategy via improving immunogenicity and reversing immune-inhibitory microenvironment. Biomaterials 2022, 288, 121722. [Google Scholar] [CrossRef]
  141. Wang, X.; Ye, L.; He, W.; Teng, C.; Sun, S.; Lu, H.; Li, S.; Lv, L.; Cao, X.; Yin, H.; et al. In situ targeting nanoparticles-hydrogel hybrid system for combined chemo-immunotherapy of glioma. J. Control. Release 2022, 345, 786–797. [Google Scholar] [CrossRef] [PubMed]
  142. Ren, X.; Wang, N.; Zhou, Y.; Song, A.; Jin, G.; Li, Z.; Luan, Y. An injectable hydrogel using an immunomodulating gelator for amplified tumor immunotherapy by blocking the arginase pathway. Acta Biomater. 2021, 124, 179–190. [Google Scholar] [CrossRef] [PubMed]
  143. Ding, M.; Fan, Y.; Lv, Y.; Liu, J.; Yu, N.; Kong, D.; Sun, H.; Li, J. A prodrug hydrogel with tumor microenvironment and near-infrared light dual-responsive action for synergistic cancer immunotherapy. Acta Biomater. 2022, 149, 334–346. [Google Scholar] [CrossRef] [PubMed]
  144. Wan, C.; Sun, Y.; Hu, Y.; Huang, J.; Lu, L.; Gao, Y.; Zi, H.; He, Q.; Sun, J.; Lovell, J.F.; et al. Peptide hydrogels loaded with irradiated tumor cell secretions enhance cancer immunotherapy. Nano Today 2021, 41, 101323. [Google Scholar] [CrossRef]
  145. Xiao, Z.; Wang, D.; Wang, C.; Chen, Z.; Huang, C.; Yang, Y.; Xie, L.; Zhang, L.; Xu, L.; Zhang, M.-R.; et al. PEIGel: A biocompatible and injectable scaffold with innate immune adjuvanticity for synergized local immunotherapy. Mater. Today Bio 2022, 15, 100297. [Google Scholar] [CrossRef]
  146. Qi, J.; Ding, T.; Liu, T.; Xia, X.; Wu, S.; Liu, J.; Chen, Q.; Zhang, D.; Zhao, H. Inosine-Based Supramolecular Hydrogel for Highly Efficient PD-L1 Blockade Therapy via Mediating CD8+ T Cells. Adv. Funct. Mater. 2022, 32, 2204273. [Google Scholar] [CrossRef]
  147. Sun, S.; Gu, W.; Wu, H.; Zhao, Q.; Qian, S.; Xiao, H.; Yang, K.; Liu, J.; Jin, Y.; Hu, C.; et al. Immunostimulant In Situ Hydrogel Improves Synergetic Radioimmunotherapy of Malignant Glioblastoma Relapse Post-Resection. Adv. Funct. Mater. 2022, 32, 2205038. [Google Scholar] [CrossRef]
  148. Si, X.; Ji, G.; Ma, S.; Xu, Y.; Zhao, J.; Zhang, Y.; Huang, Z.; Tang, Z.; Song, W.; Chen, X. In–Situ-Sprayed Dual-Functional Immunotherapeutic Gel for Colorectal Cancer Postsurgical Treatment. Adv. Healthc. Mater. 2021, 10, 2100862. [Google Scholar] [CrossRef]
  149. Meng, Z.; Zhou, X.; Xu, J.; Han, X.; Dong, Z.; Wang, H.; Zhang, Y.; She, J.; Xu, L.; Wang, C.; et al. Light-Triggered In Situ Gelation to Enable Robust Photodynamic-Immunotherapy by Repeated Stimulations. Adv. Mater. 2019, 31, 1900927. [Google Scholar] [CrossRef]
  150. Sun, L.; Shen, F.; Tian, L.; Tao, H.; Xiong, Z.; Xu, J.; Liu, Z. ATP-Responsive Smart Hydrogel Releasing Immune Adjuvant Synchronized with Repeated Chemotherapy or Radiotherapy to Boost Antitumor Immunity. Adv. Mater. 2021, 33, 2007910. [Google Scholar] [CrossRef]
  151. Li, S.; Zhu, C.; Zhou, X.; Chen, L.; Bo, X.; Shen, Y.; Guan, X.; Han, X.; Shan, D.; Sun, L.; et al. Engineering ROS-Responsive Bioscaffolds for Disrupting Myeloid Cell-Driven Immunosuppressive Niche to Enhance PD-L1 Blockade-Based Postablative Immunotherapy. Adv. Sci. 2022, 9, 2104619. [Google Scholar] [CrossRef] [PubMed]
  152. Wang, F.; Xu, D.; Su, H.; Zhang, W.; Sun, X.; Monroe, M.K.; Chakroun, R.W.; Wang, Z.; Dai, W.; Oh, R.; et al. Supramolecular prodrug hydrogelator as an immune booster for checkpoint blocker–based immunotherapy. Sci. Adv. 2020, 6, eaaz8985. [Google Scholar] [CrossRef] [PubMed]
  153. Gu, J.; Zhao, G.; Yu, J.; Xu, P.; Yan, J.; Jin, Z.; Chen, S.; Wang, Y.; Zhang, L.W.; Wang, Y. Injectable pH-responsive hydrogel for combinatorial chemoimmunotherapy tailored to the tumor microenvironment. J. Nanobiotechnology 2022, 20, 372. [Google Scholar] [CrossRef] [PubMed]
  154. Li, Z.; Ding, Y.; Liu, J.; Wang, J.; Mo, F.; Wang, Y.; Chen-Mayfield, T.-J.; Sondel, P.M.; Hong, S.; Hu, Q. Depletion of tumor associated macrophages enhances local and systemic platelet-mediated anti-PD-1 delivery for post-surgery tumor recurrence treatment. Nat. Commun. 2022, 13, 1845. [Google Scholar] [CrossRef] [PubMed]
  155. Wang, F.; Su, H.; Xu, D.; Dai, W.; Zhang, W.; Wang, Z.; Anderson, C.F.; Zheng, M.; Oh, R.; Wan, F.; et al. Tumour sensitization via the extended intratumoural release of a STING agonist and camptothecin from a self-assembled hydrogel. Nat. Biomed. Eng. 2020, 4, 1090–1101. [Google Scholar] [CrossRef]
  156. Park, J.; Wrzesinski, S.H.; Stern, E.; Look, M.; Criscione, J.; Ragheb, R.; Jay, S.M.; Demento, S.L.; Agawu, A.; Limon, P.L.; et al. Combination delivery of TGF-β inhibitor and IL-2 by nanoscale liposomal polymeric gels enhances tumour immunotherapy. Nat. Mater. 2012, 11, 895–905. [Google Scholar] [CrossRef]
  157. Hou, X.-l.; Dai, X.; Yang, J.; Zhang, B.; Zhao, D.-h.; Li, C.-q.; Yin, Z.-y.; Zhao, Y.-d.; Liu, B. Injectable polypeptide-engineered hydrogel depot for amplifying the anti-tumor immune effect induced by chemo-photothermal therapy. J. Mater. Chem. B 2020, 8, 8623–8633. [Google Scholar] [CrossRef]
  158. Yan, J.; Zhang, Z.; Zhan, X.; Chen, K.; Pu, Y.; Liang, Y.; He, B. In situ injection of dual-delivery PEG based MMP-2 sensitive hydrogels for enhanced tumor penetration and chemo-immune combination therapy. Nanoscale 2021, 13, 9577–9589. [Google Scholar] [CrossRef]
  159. Song, H.; Yang, P.; Huang, P.; Zhang, C.; Kong, D.; Wang, W. Injectable polypeptide hydrogel-based co-delivery of vaccine and immune checkpoint inhibitors improves tumor immunotherapy. Theranostics 2019, 9, 2299–2314. [Google Scholar] [CrossRef]
  160. Dai, X.; Meng, J.; Deng, S.; Zhang, L.; Wan, C.; Lu, L.; Huang, J.; Hu, Y.; Zhang, Z.; Li, Y.; et al. Targeting CAMKII to reprogram tumor-associated macrophages and inhibit tumor cells for cancer immunotherapy with an injectable hybrid peptide hydrogel. Theranostics 2020, 10, 3049–3063. [Google Scholar] [CrossRef]
  161. Liang, X.; Li, L.; Li, X.; He, T.; Gong, S.; Zhu, S.; Zhang, M.; Wu, Q.; Gong, C. A spontaneous multifunctional hydrogel vaccine amplifies the innate immune response to launch a powerful antitumor adaptive immune response. Theranostics 2021, 11, 6936–6949. [Google Scholar] [CrossRef] [PubMed]
  162. Li, C.; Liu, Y.; Li, D.; Wang, Q.; Zhou, S.; Zhang, H.; Wang, Y.; He, Z.; Liu, H.; Sun, J. Promising alternatives of CD47 monoclonal antibody: An injectable degradable hydrogel loaded with PQ912 for postoperative immunotherapy effectively blocks CD47-SIRPα signal. Theranostics 2022, 12, 4581–4598. [Google Scholar] [CrossRef] [PubMed]
  163. Zhang, Y.; Feng, Z.; Liu, J.; Li, H.; Su, Q.; Zhang, J.; Huang, P.; Wang, W.; Liu, J. Polarization of tumor-associated macrophages by TLR7/8 conjugated radiosensitive peptide hydrogel for overcoming tumor radioresistance. Bioact. Mater. 2022, 16, 359–371. [Google Scholar] [CrossRef] [PubMed]
  164. Liu, M.; Cao, Z.; Zhang, R.; Chen, Y.; Yang, X. Injectable Supramolecular Hydrogel for Locoregional Immune Checkpoint Blockade and Enhanced Cancer Chemo-Immunotherapy. ACS Appl. Mater. Interfaces 2021, 13, 33874–33884. [Google Scholar] [CrossRef] [PubMed]
  165. Zhang, H.; Liu, K.; Gong, Y.; Zhu, W.; Zhu, J.; Pan, F.; Chao, Y.; Xiao, Z.; Liu, Y.; Wang, X.; et al. Vitamin C supramolecular hydrogel for enhanced cancer immunotherapy. Biomaterials 2022, 287, 121673. [Google Scholar] [CrossRef]
  166. Wu, Y.; Li, Q.; Shim, G.; Oh, Y.-K. Melanin-loaded CpG DNA hydrogel for modulation of tumor immune microenvironment. J.Control. Release 2021, 330, 540–553. [Google Scholar] [CrossRef]
  167. Kremenovic, M.; Chan, A.A.; Feng, B.; Bariswyl, L.; Robatel, S.; Gruber, T.; Tang, L.; Lee, D.J.; Schenk, M. BCG hydrogel promotes CTSS-mediated antigen processing and presentation, thereby suppressing metastasis and prolonging survival in melanoma. J. Immunother. Cancer 2022, 10, e004133. [Google Scholar] [CrossRef]
  168. Li, Y.; Fang, M.; Zhang, J.; Wang, J.; Song, Y.; Shi, J.; Li, W.; Wu, G.; Ren, J.; Wang, Z.; et al. Hydrogel dual delivered celecoxib and anti-PD-1 synergistically improve antitumor immunity. Oncoimmunology 2016, 5, e1074374. [Google Scholar] [CrossRef]
  169. Wang, B.; Chen, J.; Caserto, J.S.; Wang, X.; Ma, M. An in situ hydrogel-mediated chemo-immunometabolic cancer therapy. Nat. Commun. 2022, 13, 3821. [Google Scholar] [CrossRef]
  170. He, T.; Liang, X.; Li, L.; Gong, S.; Li, X.; Zhang, M.; Zhu, S.; Xiao, H.; Wu, Q.; Gong, C. A spontaneously formed and self-adjuvanted hydrogel vaccine triggers strong immune responses. Mater. Des. 2021, 197, 109232. [Google Scholar] [CrossRef]
  171. Song, H.; Huang, P.; Niu, J.; Shi, G.; Zhang, C.; Kong, D.; Wang, W. Injectable polypeptide hydrogel for dual-delivery of antigen and TLR3 agonist to modulate dendritic cells in vivo and enhance potent cytotoxic T-lymphocyte response against melanoma. Biomaterials 2018, 159, 119–129. [Google Scholar] [CrossRef] [PubMed]
  172. Zhao, Q.; Wang, Y.; Zhao, B.; Chen, H.; Cai, Z.; Zheng, Y.; Zeng, Y.; Zhang, D.; Liu, X. Neoantigen Immunotherapeutic-Gel Combined with TIM-3 Blockade Effectively Restrains Orthotopic Hepatocellular Carcinoma Progression. Nano Lett. 2022, 22, 2048–2058. [Google Scholar] [CrossRef] [PubMed]
  173. Yin, Y.; Li, X.; Ma, H.; Zhang, J.; Yu, D.; Zhao, R.; Yu, S.; Nie, G.; Wang, H. In Situ Transforming RNA Nanovaccines from Polyethylenimine Functionalized Graphene Oxide Hydrogel for Durable Cancer Immunotherapy. Nano Lett. 2021, 21, 2224–2231. [Google Scholar] [CrossRef] [PubMed]
  174. Kim, J.; Francis, D.M.; Sestito, L.F.; Archer, P.A.; Manspeaker, M.P.; O’Melia, M.J.; Thomas, S.N. Thermosensitive hydrogel releasing nitric oxide donor and anti-CTLA-4 micelles for anti-tumor immunotherapy. Nat. Commun. 2022, 13, 1479. [Google Scholar] [CrossRef]
  175. O’Melia, M.J.; Manspeaker, M.P.; Thomas, S.N. Tumor-draining lymph nodes are survival niches that support T cell priming against lymphatic transported tumor antigen and effects of immune checkpoint blockade in TNBC. Cancer Immunol. Immunother. 2021, 70, 2179. [Google Scholar] [CrossRef] [PubMed]
  176. Engelhard, V.H.; Rodriguez, A.B.; Mauldin, I.S.; Woods, A.N.; Peske, J.D.; Slingluff, C.L., Jr. Immune Cell Infiltration and Tertiary Lymphoid Structures as Determinants of Antitumor Immunity. J. Immunol. 2018, 200, 432–442. [Google Scholar] [CrossRef] [Green Version]
  177. Joshi, N.S.; Akama-Garren, E.H.; Lu, Y.; Lee, D.-Y.; Chang, G.P.; Li, A.; DuPage, M.; Tammela, T.; Kerper, N.R.; Farago, A.F.; et al. Regulatory T Cells in Tumor-Associated Tertiary Lymphoid Structures Suppress Anti-tumor T Cell Responses. Immunity 2015, 43, 579–590. [Google Scholar] [CrossRef] [Green Version]
  178. Mohamed, L.; Elsaka, A.; Zamzam, Y.; Elkady, A. Tumor Infiltrating Lymphocytes and Tertiary Lymphoid Structure as Prognostic and Predictive Factor for Neoadjuvant Chemotherapy in Stage II & III Breast Cancer. Arch. Can. Res. 2018, 6, 17. [Google Scholar]
  179. Tseng, Y.-C.; Xu, Z.; Guley, K.; Yuan, H.; Huang, L. Lipidecalcium phosphate nanoparticles for delivery to the lymphatic system and SPECT/CT imaging of lymph node metastases. Biomaterials 2014, 35, 4688–4698. [Google Scholar] [CrossRef] [Green Version]
  180. Videira, M.A.; Botelho, M.F.; Santos, A.C.; Gouveia, L.F.; Pedroso De Lima, J.J.; Almeida, A.J. Lymphatic Uptake of Pulmonary Delivered Radiolabelled Solid Lipid Nanoparticles. J. Drug Target. 2002, 10, 607–613. [Google Scholar] [CrossRef]
  181. Mohammad, A.K.; Amayreh, L.K.; Mazzara, J.M.; Reineke, J.J. Rapid Lymph Accumulation of Polystyrene Nanoparticles Following Pulmonary Administration. Pharm. Res. 2013, 30, 424–434. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic illustration of the preparation and actions of NPs. (A) Microflowers were first synthesized via self-assembly of polymerized DNA and shRNA synthesized using the RCA of DNA templates to encode CpG and Stat3 shRNA. (B,C) (B) Cationic polymers allowed the shrinking of microflowers to develop NPs capable of (C) lymphatic drainage for adaptive immune response. The figure is adapted from [93] with permission from Springer Nature. Copyright 2017 Springer Nature.
Figure 1. Schematic illustration of the preparation and actions of NPs. (A) Microflowers were first synthesized via self-assembly of polymerized DNA and shRNA synthesized using the RCA of DNA templates to encode CpG and Stat3 shRNA. (B,C) (B) Cationic polymers allowed the shrinking of microflowers to develop NPs capable of (C) lymphatic drainage for adaptive immune response. The figure is adapted from [93] with permission from Springer Nature. Copyright 2017 Springer Nature.
Pharmaceutics 14 02752 g001
Figure 2. Schematic illustration of the preparation and actions of SNA with liposomal nanodisc cores. (A) Antigens were conjugated to liposomal nanodisc via a disulfide bond and cholesterol-CpG were embedded on the phospholipid layers to afford an SNA structure where CpG was exposed to the nanodisc. (B) The nanodisc was drained into the LNs to instruct the antigen-specific CD8+ T cells in LNs for anticancer immunotherapy. The figure is adapted from [99] with permission from Springer Nature. Copyright 2017 Springer Nature.
Figure 2. Schematic illustration of the preparation and actions of SNA with liposomal nanodisc cores. (A) Antigens were conjugated to liposomal nanodisc via a disulfide bond and cholesterol-CpG were embedded on the phospholipid layers to afford an SNA structure where CpG was exposed to the nanodisc. (B) The nanodisc was drained into the LNs to instruct the antigen-specific CD8+ T cells in LNs for anticancer immunotherapy. The figure is adapted from [99] with permission from Springer Nature. Copyright 2017 Springer Nature.
Pharmaceutics 14 02752 g002
Figure 3. Schematic illustration of the preparation and actions of pH-sensitive DNA nanodevice. (A) Rectangle-shaped DNA was prepared using staple strands and M13 bacteriophage DNA, which were further engineered to load tumor peptide-antigen-DNA conjugate antigens and double stranded RNA (dsRNA) adjuvants. pH-sensitive locking strands were then employed to generate tubular DNA nanostructures. (B) Mechanism of actions of the DNA nanostructures. pH-sensitive locking DNA was dehybridized to open the tubular structure, exposing the antigens and dsRNA outside to stimulate DC activation and presentation of antigens after draining into the LNs, followed by uptake by DCs, leading to antigen-specific CD8+ T cell response for robust anticancer immunotherapy. The figure is adapted from [104] with permission from Springer Nature. Copyright 2021 Springer Nature.
Figure 3. Schematic illustration of the preparation and actions of pH-sensitive DNA nanodevice. (A) Rectangle-shaped DNA was prepared using staple strands and M13 bacteriophage DNA, which were further engineered to load tumor peptide-antigen-DNA conjugate antigens and double stranded RNA (dsRNA) adjuvants. pH-sensitive locking strands were then employed to generate tubular DNA nanostructures. (B) Mechanism of actions of the DNA nanostructures. pH-sensitive locking DNA was dehybridized to open the tubular structure, exposing the antigens and dsRNA outside to stimulate DC activation and presentation of antigens after draining into the LNs, followed by uptake by DCs, leading to antigen-specific CD8+ T cell response for robust anticancer immunotherapy. The figure is adapted from [104] with permission from Springer Nature. Copyright 2021 Springer Nature.
Pharmaceutics 14 02752 g003
Figure 4. Schematic illustration of the preparation and lymphatic drainage of protein-fusion nanocages (PdNCs). (A) Self-assembled PdNCs (right) from a fusion protein (mid) containing hFTN and aPD-1 were prepared; these PdNCs were synthesized from a plasmid vector (left). (B) SDS-PAGE of the plasmid vector. (C) Representative TEM image of the PdNCs. (D) DLS size distribution of PdNCs (left) and control nanocages self-assembled from wild hFTN (right). (E) Quantification of the LN accumulation of PdNCs using ex vivo IVIS. * p < 0.05 was analyzed by one-way ANOVA with Tukey’s post-hoc. Figures are reproduced from [108] with permission from Elsevier. Copyright 2021 Elsevier.
Figure 4. Schematic illustration of the preparation and lymphatic drainage of protein-fusion nanocages (PdNCs). (A) Self-assembled PdNCs (right) from a fusion protein (mid) containing hFTN and aPD-1 were prepared; these PdNCs were synthesized from a plasmid vector (left). (B) SDS-PAGE of the plasmid vector. (C) Representative TEM image of the PdNCs. (D) DLS size distribution of PdNCs (left) and control nanocages self-assembled from wild hFTN (right). (E) Quantification of the LN accumulation of PdNCs using ex vivo IVIS. * p < 0.05 was analyzed by one-way ANOVA with Tukey’s post-hoc. Figures are reproduced from [108] with permission from Elsevier. Copyright 2021 Elsevier.
Pharmaceutics 14 02752 g004
Figure 5. Schematic illustration of the preparation and lymphatic drainage of DC-membrane cloaking NPs. (A) Preparation of aCD3 functionalized DC-membrane cloaking NPs by coating imiquimod-loaded PLGA NP with the DC membrane and conjugating aCD3 with the membrane. (B) The NPs drained into the LNs and elicited an antitumor immune response after s.c. administration. The figure is adapted from [122] with permission from American Chemical Society. Copyright 2021 American Chemical Society.
Figure 5. Schematic illustration of the preparation and lymphatic drainage of DC-membrane cloaking NPs. (A) Preparation of aCD3 functionalized DC-membrane cloaking NPs by coating imiquimod-loaded PLGA NP with the DC membrane and conjugating aCD3 with the membrane. (B) The NPs drained into the LNs and elicited an antitumor immune response after s.c. administration. The figure is adapted from [122] with permission from American Chemical Society. Copyright 2021 American Chemical Society.
Pharmaceutics 14 02752 g005
Figure 6. Schematic illustration of the preparation, characterization, lymphatic drainage, and therapeutic effects of activated DC-derived exosomes. (A) Activated DC-derived exosomes were prepared using poly(I:C) and OVA and further decorated with aCTLA-4. (B) DLS size distribution of exosomes. (C) TEM images of exosomes. (D) Quantification of the LN accumulation of exosomes. (E) Tumor growth curves with exosomes. * p < 0.05, ** p < 0.01, and *** p < 0.001 were analyzed by one-way ANOVA. Figures are reproduced from [126] with permission from Elsevier. Copyright 2020 Elsevier.
Figure 6. Schematic illustration of the preparation, characterization, lymphatic drainage, and therapeutic effects of activated DC-derived exosomes. (A) Activated DC-derived exosomes were prepared using poly(I:C) and OVA and further decorated with aCTLA-4. (B) DLS size distribution of exosomes. (C) TEM images of exosomes. (D) Quantification of the LN accumulation of exosomes. (E) Tumor growth curves with exosomes. * p < 0.05, ** p < 0.01, and *** p < 0.001 were analyzed by one-way ANOVA. Figures are reproduced from [126] with permission from Elsevier. Copyright 2020 Elsevier.
Pharmaceutics 14 02752 g006
Figure 7. Schematic illustration of the (A) preparation of multistage drug delivery systems and (B) distribution of cargos in dLNs. Figures are reproduced from [128] with permission from Springer Nature. Copyright 2020 Springer Nature.
Figure 7. Schematic illustration of the (A) preparation of multistage drug delivery systems and (B) distribution of cargos in dLNs. Figures are reproduced from [128] with permission from Springer Nature. Copyright 2020 Springer Nature.
Pharmaceutics 14 02752 g007
Figure 8. Lymphatic drainage and immune profiles of the tumor cell lysates (TCL) and poly(I:C)-loaded hydrogels. (A) Chemical structure of mPEG-poly(L-Valine). (B) Image of a hydrogel. (C) IVIS images of antigens-Cy7 administered and released from hydrogels. (D) Quantification of fluorescence signals at the injection site over time. (E) Quantification of the fluorescence signals of antigens-Cy7 at LNs. (F) Ex vivo IVIS images of the LNs at 96 h after injection. (GI) Populations of (G) CD8+ T, (H) CD80+ DCs, and (I) MHCII+ DCs in the dLNs. ** p < 0.01, *** p < 0.001, and **** p < 0.0001 were analyzed by students’ t-test. Figures are reproduced from [171] with permission from Elsevier. Copyright 2018 Elsevier.
Figure 8. Lymphatic drainage and immune profiles of the tumor cell lysates (TCL) and poly(I:C)-loaded hydrogels. (A) Chemical structure of mPEG-poly(L-Valine). (B) Image of a hydrogel. (C) IVIS images of antigens-Cy7 administered and released from hydrogels. (D) Quantification of fluorescence signals at the injection site over time. (E) Quantification of the fluorescence signals of antigens-Cy7 at LNs. (F) Ex vivo IVIS images of the LNs at 96 h after injection. (GI) Populations of (G) CD8+ T, (H) CD80+ DCs, and (I) MHCII+ DCs in the dLNs. ** p < 0.01, *** p < 0.001, and **** p < 0.0001 were analyzed by students’ t-test. Figures are reproduced from [171] with permission from Elsevier. Copyright 2018 Elsevier.
Pharmaceutics 14 02752 g008
Figure 9. Characterization, lymphatic drainage, and anticancer immunotherapeutic effects of ICB mAbs-loading micelles in situ released from thermosensitive hydrogels. (A) Representative SEM image of F127-Gelatin thermosensitive hydrogels. (B) Cumulative release of aCTLA-4 mAbs from the hydrogel in response to MMP-9. (C) Representative image of the F127-Gelatin micelle in situ released from the hydrogel. (D) FRET assay to demonstrate the load of aCTLA-4 mAbs on the in situ micelle via confirmation of the FRET signal between FITC-labeled F127-Gelatin polymer and TRITC-labeled aCTLA-4. (EH) Quantification of the fluorescence signals of AF647-labeled aCTLA-4 mAbs released from the hydrogel at LNs and tumor after i.t. injection into the primary tumor (1o tumor). (I) 1o and (J) 2o tumor growth curves with the hydrogel formulations. * p < 0.1, ** p < 0.05, *** p < 0.01, **** p < 0.001, and ***** p < 0.0001 were analyzed by one-way ANOVA with Tukey post-hoc for E-H, two-way ANOVA with Tukey post-hoc for B, and ANOVA using linear mixed-effects model for I and J. Figures are reproduced from [174] with permission from Springer Nature. Copyright 2022 Springer Nature.
Figure 9. Characterization, lymphatic drainage, and anticancer immunotherapeutic effects of ICB mAbs-loading micelles in situ released from thermosensitive hydrogels. (A) Representative SEM image of F127-Gelatin thermosensitive hydrogels. (B) Cumulative release of aCTLA-4 mAbs from the hydrogel in response to MMP-9. (C) Representative image of the F127-Gelatin micelle in situ released from the hydrogel. (D) FRET assay to demonstrate the load of aCTLA-4 mAbs on the in situ micelle via confirmation of the FRET signal between FITC-labeled F127-Gelatin polymer and TRITC-labeled aCTLA-4. (EH) Quantification of the fluorescence signals of AF647-labeled aCTLA-4 mAbs released from the hydrogel at LNs and tumor after i.t. injection into the primary tumor (1o tumor). (I) 1o and (J) 2o tumor growth curves with the hydrogel formulations. * p < 0.1, ** p < 0.05, *** p < 0.01, **** p < 0.001, and ***** p < 0.0001 were analyzed by one-way ANOVA with Tukey post-hoc for E-H, two-way ANOVA with Tukey post-hoc for B, and ANOVA using linear mixed-effects model for I and J. Figures are reproduced from [174] with permission from Springer Nature. Copyright 2022 Springer Nature.
Pharmaceutics 14 02752 g009
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cho, K.J.; Cho, Y.-E.; Kim, J. Locoregional Lymphatic Delivery Systems Using Nanoparticles and Hydrogels for Anticancer Immunotherapy. Pharmaceutics 2022, 14, 2752. https://doi.org/10.3390/pharmaceutics14122752

AMA Style

Cho KJ, Cho Y-E, Kim J. Locoregional Lymphatic Delivery Systems Using Nanoparticles and Hydrogels for Anticancer Immunotherapy. Pharmaceutics. 2022; 14(12):2752. https://doi.org/10.3390/pharmaceutics14122752

Chicago/Turabian Style

Cho, Kyeong Jin, Young-Eun Cho, and Jihoon Kim. 2022. "Locoregional Lymphatic Delivery Systems Using Nanoparticles and Hydrogels for Anticancer Immunotherapy" Pharmaceutics 14, no. 12: 2752. https://doi.org/10.3390/pharmaceutics14122752

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