1 s2.0 S2452199X21001092 Main
1 s2.0 S2452199X21001092 Main
1 s2.0 S2452199X21001092 Main
Bioactive Materials
journal homepage: www.sciencedirect.com/journal/bioactive-materials
A R T I C L E I N F O A B S T R A C T
Keywords: Cancer immunotherapy is an effective antitumor approach through activating immune systems to eradicate
Polysaccharides tumors by immunotherapeutics. However, direct administration of “naked” immunotherapeutic agents (such as
Drug delivery systems nucleic acids, cytokines, adjuvants or antigens without delivery vehicles) often results in: (1) an unsatisfactory
Nanomedicines
efficacy due to suboptimal pharmacokinetics; (2) strong toxic and side effects due to low targeting (or off-target)
Cancer immunotherapy
Anticancer efficacy
efficiency. To overcome these shortcomings, a series of polysaccharide-based nanoparticles have been developed
to carry immunotherapeutics to enhance antitumor immune responses with reduced toxicity and side effects.
Polysaccharides are a family of natural polymers that hold unique physicochemical and biological properties, as
they could interact with immune system to stimulate an enhanced immune response. Their structures offer
versatility in synthesizing multifunctional nanocomposites, which could be chemically modified to achieve high
stability and bioavailability for delivering therapeutics into tumor tissues. This review aims to highlight recent
advances in polysaccharide-based nanomedicines for cancer immunotherapy and propose new perspectives on
the use of polysaccharide-based immunotherapeutics.
1. Introduction new therapeutic targets [1,8,9] or new strategies [10–12] to improve the
efficacy and reduce the side effect of immunotherapy are becoming
Cancer is one of the major diseases with high prevalence, severe attractive in cancer treatment. However, since tumor immunity is a
symptoms and clinical manifestations, unfavorable treatment responses complex process that is not fully understood yet, this emerging immu
and poor prognosis, thus early diagnosis and effective treatment of notherapy is facing great challenges such as a low targeting efficacy,
cancer are a hotspot that has attracted great attention. Currently, clin which would reduce the therapeutic effect of administrated drugs, and
ically practical and effective treatment methods for cancer are focused intrinsic toxicities of immunotherapeutic drugs, which could cause se
on surgery, chemotherapy, radiotherapy and immunotherapy, which are vere inflammatory and autoimmune diseases [13–15]. In this context,
known as the four pillars of cancer treatment [1]. Among them, many attempts have been made to improve the efficiency/efficacy of
immunotherapy, using human immune systems to treat cancer, blocks cancer immunotherapy while minimizing its side effects [16,17]. One of
certain immune inhibitory checkpoints or pathways [2,3], promote T the most promising approaches is to apply nanomaterials as carriers for
cells killing ability towards tumor cells (i.e. CAR-T therapy) [4,5] and immunotherapeutic agents.
increase innate immune processes by tumor associated macrophages With the rapid growth of nanobiotechnology, nanomaterials have
(TAMs) and natural killer (NK) cells [6,7] to accomplish targeting im become more and more clinically applicable for medical treatment [18,
mune suppression or elimination of tumor cells. Some of these immu 19]. These nanomaterials with a size of 10–500 nm are able to increase
notherapy strategies have been adopted in clinical practices and the therapeutic efficacy and reduce the toxicity of therapeutic drugs by
demonstrated to be efficacious for cancer diseases. The emergence of encapsulating or conjugating them to form stabilized nanomedicines
https://doi.org/10.1016/j.bioactmat.2021.03.008
Received 13 January 2021; Received in revised form 19 February 2021; Accepted 2 March 2021
Available online 18 March 2021
2452-199X/© 2021 The Authors. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co. Ltd. This is an open access article under the CC
BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
[20,21]. These nanomedicines selectively penetrate into tumor tissues for cancer immunotherapy. Thus this review aims to provide an over
and achieve controlled, efficient and sustained drug release. All these view of various natural polysaccharide-based immunotherapeutic de
advantages of nanomaterials earn them a space in the arena of cancer livery nanosystems and their cancer immunotherapy applications. The
diseases treatment [22,23]. Studies [24–26] have also demonstrated types of polysaccharides, their delivery nanosystems for immunothera
that nanomaterials could encapsulate or conjugate immunotherapeutic peutic agents, mechanisms of immune-activation/suppression at the
antigens, adjuvants, genes or antibodies to act as cancer nanovaccines molecular level, as well as their immunotherapeutic effects are
that could improve the efficacy of cancer immunotherapy (Fig. 1) while described and discussed.
reducing toxicity and side effects of these managed drugs. Among these
applied nanomaterials are polysaccharide-based nanosystems. 2. Cancer immunotherapy and potential role of polysaccharides
Because polysaccharides are biocompatible and biodegradable, thus and their derivatives
relatively safe to be applied in medical practices, polysaccharide-based
nanosystems have attracted significant attention as delivery platforms 2.1. Current status of cancer immunotherapy
for the treatment of various diseases [27,28]. The effectiveness of
polysaccharide-based nanomedicines in improving the antitumor ther In response to tumor genesis and growth, living bodies can generate
apeutic efficacy has been demonstrated [29,30]. Polysaccharide-based immune responses to eliminate these tumor cells, this immune stimu
nanoparticles could bypass adenosine triphosphate (ATP) binding latory effect is usually insufficient to eradicate tumor cells completely,
cassette transporters and are internalized into targeting cells such as and tumor tissues continue to grow and metastasize [67–69]. External
microfold cells or CD44 overexpressing tumor cells [31,32]. Since sys immunostimulators and immunomodulators are often required to evoke
temic administration of many antitumor drugs such as cisplatin, pacli a strong immune reaction that could effectively suppress or eliminate
taxel, and doxorubicin would result in severe side effects like tumor cells [70–72]. . To achieve cancer immunotherapy, currently
hepatotoxicity, nephrotoxicity, neurotoxicity or hypersensitivity re there are three major immunity stimulating and enhancing methods for
actions [33,34], this targeting polysaccharide-based nanosystem could cancer, including immune cell therapy, antibody therapy and cytokine
deliver these antitumor drugs only into selective cells to increase their therapy. Immune cell therapy applies genetically modified immune cells
therapeutic efficacy, and reduce the toxicity and the incidence of side to patients to provoke antitumor responses. Chimeric antigen receptor T
effects. In addition, it has also been reported that some polysaccharides (CAR-T) cell therapy has been successfully commercialized for liquid
could stimulate intrinsic antitumor immune systems themselves cancer, and US Food and Drug Administration (FDA) approved CAR-T
[35–37], thus triggering more studies on polysaccharide-based nano therapeutics include Breyanzi (Juno Therapeutics), Kymriah™ (Novar
medicines for cancer immunotherapy. tis) and Yescarta™ (Kite Pharma). By transducing the CAR gene into T
Regarding to this, functional nanomaterials based on chitosan, hy cells through viral vectors, CAR-T cells could specifically recognize
aluronic acid (HA), dextran, alginate and other polysaccharides have tumor cells and initiate a strong immune attack towards them [73].
been explored as potential drug delivery platforms for immunothera Provenge (Sipuleucel-T) developed by Dendreon Pharmaceuticals is
peutic agents and a few polysaccharides have been acted as immune another approved cellular product for immune cell therapy, and den
adjuvants themselves for cancer treatment (Tables 1–3). A few review dritic cells (DCs) instead of T cells are used in this product [74].
articles about polysaccharide-based nanomaterials and their biological Monoclonal antibodies are used as immunotherapeutics for antibody
applications or polysaccharide-derived nanomedicines for cancer ther therapy. After formation of B-cell and myeloma-cell complexes with
apy have been published in the past years. Two reviews in 2015 [38,39] unique tumor antigens on myeloma cells, the generated monoclonal
and another two in 2018 [40,41] thoroughly discussed the immuno antibodies could specifically target tumor cells, resulting in strong tumor
logical responses of polysaccharides and their potential for immuno immune stimulation and modulation. This is achieved through
therapy. Another four reviews in the past 6 years on the other hand antibody-dependent cell-mediated cytotoxicity (ADCC) directly towards
provided an overview of how nanomaterials can be beneficial for cancer tumor cells, or by stimulating the complement system to activate the
immunotherapy [24,42–44]. However, none of these reviews have membrane attack complex. FDA approved therapeutics with this
specifically discussed the role of polysaccharide-based nanomedicines mechanism include Rituximab [75], Alemtuzumab [76], Ofatumumab
3359
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
Table 1
Polysaccharides in the nano-based cancer immunotherapy and their structure and advantages.
Polysaccharide Type Structure Advantages
LYVE-1: lymphatic vessel endothelial-1 receptor; RHAMM: receptor for hyaluronic acid -mediated motility.
Table 2
Polysaccharide-based nanoparticles as delivery vehicles for cancer immunotherapy.
Polysaccharide Nanomaterial Loaded Agents Therapeutic Effects References
Type
Chitosan polyaniline-glycol-chitosan R848 induce dendritic cell maturation,promote antitumor memory [46]
nanoparticles
chitosan/poly (γ-glutamic acid) interferon-γ induce dendritic cell maturation and macrophage activation [47]
nanoparticles
PEG = MT/PC nanoparticles VEGF-siRNA, PIGF- alter the microenvironment to be anti-tumoral [48]
siRNA
mannose-chitosan-stearic acid ovalbumin and CCR7 induce dendritic cell maturation,increase CD8+ T cell population [49]
nanomicelles pDNA
poly (ethylene glycol)- g-chitosan therapeutic T cells demonstrate a better antitumor efficacy of loaded T cells [50]
hydrogel
Hyaluronic acid HA-gold nanoparticles ovalbumin increase antigen presentation, induce CD8+ T cell proliferation [51]
HA-paclitaxel-marimastat liposomes HA-paclitaxel, alter the tumor microenvironment to suppress tumor growth, metastasis and [52]
marimastat angiogenesis
HA-based hydrogel artificial T cell enhance activation of antitumor CD8+ T cells [53]
stimulating matrix
Dextran pH-sensitive HA-dextran PD-1 antibody, achieve a better therapeutic efficacy of PD-1 antibody [54]
nanoparticles glucose oxidase
spermine modified acetalated nutlin-3a, GM-CSF induce dendritic cell maturation,increase CD8+ T cell population [55]
dextran nanoparticles
dextran-grafted-poly (histidine) BLZ-945 induce M1 macrophages,increase CD8+ T cell population [56]
copolymer micell
porous silicon@acetalated exogenous antigen induce dendritic cell maturation,promote Th-1 cell differentiation [57]
dextran@cancer cell membrane
Alginate mannose-modified alginate ovalbumin increase tumor antigen presentation,induce CD8 T cell proliferation
+
[58]
nanoparticles
Chondroitin chondroitin sulfate-chlorin e6-lipoic docetaxel achieve chemo-sonodynamic combination therapy,induce tumor-associated [59]
sulfate acid nanoplatform antigen release,promote dendritic cell recognition,increase CD8+ T cell
population
Cyclodextrin β-cyclodextrin-based covalent R848 induce M1 macrophages,suppress M2 macrophages [60]
crosslinking nanoparticles
PEG: polyethylene glycol; MT: trimethyl chitosan; PC: citraconic anhydride grafted poly (allylamine hydrochloride); VEGF: vascular endothelial growth factor; PIGF:
placental growth factor; HA: hyaluronic acid; PEI: poly (ethylenimine); GM-CSF: granulocyte-macrophage colony-stimulating factor; PD-L: programmed death ligand.
[77] and Elotuzumab [78]. Another immune modulating mechanism by cells to escape immune elimination. By blocking tumor-related immune
antibodies is to block immune checkpoints. These immune checkpoints checkpoint proteins from binding their receptors or partner proteins,
usually act as error correctors that prevent an overstressed immune immune checkpoint inhibitors could effectively restore the immune
system from harming healthy cells, but could also be utilized by tumor function towards tumor cells and even promote an enhanced immune
3360
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
3361
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
was reported to enhance the phagocytosis activity of macrophages and [145]. Due to its versatility to form nanosystems with different func
induce the release of pro-inflammatory cytokines [123]. This has been tions, along with its excellent biocompatibility, non-toxicity and
confirmed by in vitro and in vivo experiments [124,125]. Phosphoryla mucoadhesiveness for enhancing drug absorption and bioavailability,
tion of polysaccharide surface groups is believed to enhance the prolif chitosan is usually considered to be one of the most widely used poly
eration of lymphocytes, induce the maturation of B cells and increase the saccharides in the nanomedicine field [146–148]. The generated
ability of their antigen presentation and cytokines secretion, thus chitosan-based nanomedicines usually have high biodegradability,
effectively regulating the systemic immune function [126]. The addition excellent biocompatibility, bioactivity and polycationicity [149–151],
of selenium could promote lymphocyte proliferation, induce or promote thus they are effective nanocarriers for drug delivery applications
lymphocytes to produce interferon, IL-2 and other soluble immune [152–154]. As will be presented next, only recently, there are several
factors, which is supported by the study of Qin et al. [127]. However, the studies attempting to use chitosan-based nanomaterials to deliver
influence of carboxymethylation of polysaccharides on immunomodu immunotherapeutic agents for cancer treatment.
lation is quite dependent on the structure of the original poly
saccharides. Studies have demonstrated that carboxymethylation of 3.1. Chitosan-based nanosystems to deliver adjuvants
herbal polysaccharides from Astragalus mongholicus or the seeds of
Plantago asiatica L. could effectively enhance the immune activity of One of the most promising immunotherapeutic agents, which have
immune cells such as DCs [109,128], while carboxymethylation of been developed and also approved for clinical trials, are molecular ad
herbal polysaccharides from Ganoderma atrum [129] or Dendrobium juvants [155]. They enhance the efficiency of antigen delivery and
candidum [40] had not enhanced the immune activity or had an induce their recognition by antigen presenting cells (APC). By increasing
immunosuppressive effect. Therefore, structure modifications of poly immunogenicity, adjuvants could stimulate the anti-tumor immunity
saccharides need to be rationally designed for favorable immunomod and decrease the immune evasion of tumor cells. It has been reported
ulatory effects. that a few adjuvants could induce the expression of CD80 and CD86 on
When polysaccharides are applied as a drug delivery platform, the the surface of DCs, which are the markers of DC maturation. These
loaded immunotherapeutics should be aligned with the structure of their maturated DCs would increase their production of pro-inflammatory
delivery platforms. For example, it was reported that positively-charged cytokines that could boost downstream antitumor immune responses
particles for gene delivery have higher transfection efficiencies [156,157]. However, some adjuvants exhibit intrinsic immunogenicity
compared with neutral or negatively-charged delivery vectors [130]. thus they could be degraded before they achieve the modulatory effects
Therefore, chitosan, with a positive-charge property, has been applied to towards DCs. Some adjuvants could systemically modulate the immune
deliver nucleic acids for cancer immunotherapy more often than other environment besides the maturation of DC, which would cause severe
polysaccharides like HA or dextran or alginate, which are negatively side effects [158,159]. How to effectively and selectively deliver adju
charged. The surface charge of polysaccharide-based nanomaterials vants into DCs is crucial for adjuvant-based cancer immunotherapy. In
should be considered when they are used to load polypeptide-based or this regard, Chen et al. [46] developed nanomedicines with hydrophilic
protein-based antigens with different charges [131,132]. In addition, glycol-chitosan (GCS) as the backbone and conjugated hydrophobic
hydrophilicity or hydrophobicity of polysaccharides nanomaterials also polyaniline (PANI) with GCS to address the above challenge. These
plays a significant role in effectively loading and releasing immuno PANI-GCS nanomaterials were utilized as a biocompatible and water
therapeutics [133,134]. By taking the above factors and commercial soluble nanocarrier for successful delivery of resiquimod (R848), a
availability into consideration, chitosan, hyaluronic acid, dextran and TLR-7/8 agonist which has already been approved by FDA as an
alginate are the most widely applied polysaccharides in the immuno immunomodulatory agent for cancer clinical trials [160]. The
therapy. The structures of these polysaccharides are shown in Table 1 PANI-GCS-R848 nanomedicines were found to promote their maturation
and their application as delivery vectors in cancer immunotherapy is selectivity towards DCs (Fig. 2a). The PANI moiety attached to the GCS
summarized in Table 2. Although chitosan, hyaluronic acid, dextran and backbone had excellent light absorption and great photothermal effect,
alginate without any modification are generally believed to have low/no while hydrophobic interaction and π-π stacking of PANI/R848 could
immunogenicity, their derivatives are often reported to demonstrate enhance the loading efficiency of R848. These maturated DCs would
excellent immunomodulatory effects with enhanced immune responses secrete several pro-inflammatory cytokines such as IL-6 and tumor ne
[40,135–138] as shown in Table 3. Thus, these polysaccharides have crosis factor (TNF)-α, which are actively involved in the immunity
been widely employed to incorporate immunotherapeutic agents to mediation and modulation of anti-tumor effects [161], in a considerable
prepare novel nanomedicines for cancer immunotherapy. Most of these amount and for a prolonged time. A long-lasting memory of systemic
polysaccharides-based nanomedicines could act as antitumor vaccines. anti-tumor responses was also observed from their experiments. These
They are distributed in lymph nodes after subcutaneous injection and R848-loaded chitosan-based nanomedicines could be further systemat
target tumor-associated immune cells to achieve synergetic and ically examined as in situ vaccines for cancer treatment. Another TLR-9
enhanced cancer immunotherapy [41,139,140]. Some agonist CpG oligonucleotide has also drawn attention recently due to its
polysaccharides-based nanomedicines, especially HA-based nano excellent capability of maturating DCs [162,163].
medicines with their binding ability towards CD44-expressing cells, Nevertheless, the majority of oligonucleotides was reported to be
could also target the tumor microenvironment to make tumor cells more degraded by deoxyribonuclease (DNase) and excreted by the kidney
vulnerable to immune responses [141,142]. rapidly after administration through injection subcutaneously, which
significantly reduced their circulating time and the therapeutic dose,
3. Chitosan-based nanomedicines thus limiting their therapeutic applications [165,166]. To overcome this
issue, chlorin e6 (Ce6)-doped-azobenzene-glycol chitosan (GC)-PEG-
Chitosan is a positively-charged natural polysaccharide that is modified mesoporous silica nanoparticles (CAGE) were introduced to
derived from deacetylation of chitin, which is abundantly found in fungi deliver CpG oligonucleotides (Fig. 2b) [164]. These CAGE nanoparticles
cell walls and anthropods shells [143,144]. Chitosan is acid-soluble due were hypoxia-responsive and could protect the loaded oligonucleotide
to the amino groups of it possess an excellent protonation ability at a low from biodegradation or renal clearance, thus improving the intracellular
pH environment, which could lead to preferred “pH-responsive man uptake efficacy as well as the targeting efficiency of adjuvants and
ners” in acidic subcellular organelles such as endosomes and lysosomes. photosensitizers to DCs. Since the delivered agents could induce the
It had been reported that chitosan and their oligomers could interact generation of immunogenic debris, promote the maturation of DCs, help
with negatively-charged agents such as tripolyphosphate (TPP) to form a exposure of tumor-associated antigens of tumor cells and enhance the
series of nanosystems with various particle sizes and zeta potentials antigen presenting effect of DCs, an increased intracellular uptake and
3362
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
Fig. 2. Preparation of chitosan-based nanomedicines (a: R848@NPs; b: GC-CAGE complex) and their therapeutic application on cancer immunotherapy. The use of
these chitosan-based nanosystems to carry immunotherapeutic agents could significant inhibit the tumor growth (c) thus beneficial for both primary and metastatic
tumor treatment (d). Reproduced with permission from Refs. [46,164]. Copyright 2019 Elsevier and 2018 American Chemical Society, respectively.
targeting efficiency would result in a higher DC maturation rate and a 3.2. Chitosan-based nanosystems to deliver cytokines
greater antigen presenting efficiency, reduce the incidence of tumor
evasion and promote a stronger immune response towards tumor cells. Another promising type of agents that are involved in immuno
In addition to protective and targeting delivery of the loaded adjuvants, therapy of cancer is cytokines. Cytokines could direct proliferation,
these chitosan-based nanomedicines could perform synergistically with activation and differentiation of the downstream immune cells and
photodynamic therapy to achieve multi-modality therapy of cancer. The induce them to express and secrete substances demonstrating tumor
PANI-GCS-R848 nanomedicines were reported to induce inhibition of suppression or killing effects [167,168]. Chitosan-based nanoparticles
tumor cell growth due to photo-generated hyperthermia and the have been explored for delivery of certain cytokines to achieve a better
immunoregulative effect from R848 [46], which enhanced the efficacy outcome of immunotherapy. Previous studies had demonstrated chito
of these chitosan-based nanomedicines for cancer treatment. san/poly (γ-glutamic acid) nanosystems and applied them to deliver
therapeutic agents such as hormones and chemokines [169–171], and
evaluated the maturation efficacy of their intrinsic macrophages and
DCs [172]. Recently these chitosan/poly (γ-glutamic acid) nanosystems
3363
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
had been further evaluated as a drug delivering platform to achieve More importantly, they noticed that these siRNA-loaded nanomedicines
targeting delivery of IFN-γ to DCs and macrophages [47]. They have could promote differentiation of T cells towards Th1 and suppress their
discovered that these IFN-γ-loaded chitosan/poly (γ-glutamic acid) differentiation towards regulatory T cells (Tregs). Since Th1 is one of
nanoparticles (IFN-γ-Ch/γ-PGA NPs) could increase the expression of main anti-tumor functioning T lymphocytes and the induction of Tregs is
CD40, CD83 and CD86 of DCs and human leukocyte antigen-DR one of the mechanisms for immunosuppression and immune evasion of
(HLA-DR), and the secretion of pro-inflammatory cytokines such as tumor cells [183–185], these siRNA-loaded PEG-chitosan-lactate nano
IL-6, IL-12/IL-23 (p40) and TNF-α [173]. Administration of these medicines could effectively stimulate anti-tumor immune responses
IFN-γ-Ch/γ-PGA NPs resulted in promotion of maturation and activation selectively at the tumor sites without systemic injection of A2AR an
of DCs. They also had a high internalization efficiency towards macro tagonists that may cause unwanted side effects through induction of Th1
phages, thus they activated certain monocyte-macrophage lineages and and suppression of Tregs. As the induction of antitumor immune re
increased the release of pro-inflammatory cytokines so as to enhance the sponses requires activation or inhibition of different receptors or
immune stimulatory effect [174,175]. These Ch/γ-PGA NPs could not signaling pathways, administration of multi-type nucleic acids targeting
only protect the cytokines from degradation and retain their bio at different pathways may have the potential to further enhance the
functions, but also deliver therapeutic agents towards targeting macro efficacy of cancer immunotherapy [186,187]. However, how to achieve
phages and DCs so as to increase the therapeutic efficacy and decrease co-delivery of different nucleic acids to targeting sites and simulta
their side effects, which provided a new and effective way to promote neously maintain their inherent bioactivity, is still a great challenge.
cytokine-based cancer immunotherapy. While Ch/γ-PGA NPs worked Recently, chitosan-based nanomaterials have been used to address this
well for delivery of IFN-γ, another study applied chitosan-modified se challenge. A novel nanomaterial composing of polyethylene glycol and
lenium nanoparticles to successfully deliver TNF–α, another critical mannose-modified trimethyl chitosan (PEG = MT) and citraconic an
pro-inflammatory cytokine that could induce and activate downstream hydride grafted poly (allylamine hydrochloride) (PC) was synthesized
effectors for antitumor responses [176]. According to in vitro and in vivo for co-delivery of both vascular endothelial growth factor (VEGF) siRNA
results, these nanoparticles could achieve stable and sustained release of (siVEGF) and placental growth factor (PIGF) siRNA (siPIGF) [48]. These
the loaded TNF-α derived polypeptides (P16) so that they could effec serum stable and “smart” pH-sensitive nanomedicines could accumulate
tively suppress proliferation of several types of tumor cells, while in tumor tissues and effectively target tumor-associated macrophages
demonstrated no toxic effect against normal non-tumorigenic epithelial and breast cancer cells without damages to other normal cells. Two
cells. Since P16 was previously reported to exhibit a limited therapeutic released siRNAs would synergize in effectively silencing certain genes,
efficacy due to its high renal clearance and hepatic metabolic rate [177], thus inhibiting proliferation of cancer cells and also changing the
this nanoparticle-based delivery could prolong the circulating time of microenvironment of tumor tissues from pro-oncogenic to anti-tumoral.
P16 and thus yielded a higher efficacy to affect the p38 MAPK/JNK It is worth noting that, by co-delivery of two siRNAs using this nano
signaling pathway, the G0/G1 cell cycle arrest, and the material, distant metastasis of the original tumor cells to lungs has also
caspase-dependent apoptosis pathway so as to suppress proliferation been significantly inhibited. Chitosan-based nanomaterials were also
and also induce apoptosis of tumor cells. It has been suggested that prepared to achieve co-delivery of not only nucleic acids, but a mixture
combination of multiple types of cytokines could achieve a synergistic of nucleic acids and therapeutic agents. For example, chitosan oligomers
immune effect, resulting in a better antitumor efficacy [168]. were mixed with TNF-α or CD40L plasmids to form DNA-loaded nano
Chitosan-based nanosystems could be employed to deliver other types of complexes [188]. Mammary carcinoma 4T1 cells were in vitro trans
cytokines or co-deliver multiple types of cytokines to enhance cancer fected with these chitosan-based nanomedicines and these transfected
immunotherapy. 4T1 cells could effectively induce the maturation of DCs and signifi
cantly increase the amount of pro-inflammatory cytokines secreted by
3.3. Chitosan-based nanosystems to deliver nucleic acids DCs. This immune process would further stimulate the proliferation of T
cells, enhance the production of IFN-γ while suppressing the release of
Apart from the delivery of the above discussed immunotherapeutic IL-4 of these T cells, thus playing a more active role in the tumor immune
agents, chitosan-based nanomaterials are also explored to deliver response.
nucleic acid (gene) for modulating immune responses for eradicating All the above studies indicated that the use of chitosan-based
cancer. The immune responsive nucleic acid (DNA or RNA) could nanocomplexes could protect the integrity and increase the therapeu
modify DCs or T cells genetically so that these genetically modified tic efficacy of the loaded plasmids, while exhibiting low/no toxicity to
immune cells could enhance their responses to tumor antigens normal cells. Since chitosan-based nanomaterials could also achieve
[178–180]. targeting endocytosis to avoid the recognition of P-glycoproteins [189],
In a recent study, chitosan-coated selenium nanoparticles were pre using this nanomaterial to deliver therapeutic agents may also help the
pared and conjugated with a folic acid-targeting moiety to deliver suppression of drug resistance. In addition to co-delivering cytokines
Photinus pyralis firefly luciferase (Fluc) mRNA [181]. These nano and plasmids, these chitosan-based nanoparticles were applied to
particles could efficiently bind and stabilize mRNA, protect them from deliver CD40, inducible co-stimulator ligand (ICOSL), and EGFP-N1
degradation from RNase and selectively deliver them into cancer cells. mRNA to DCs to enhance the antitumor effects of these immune cells
More importantly, these nanomedicines possessed low cytotoxicity to [190]. These mRNA-loaded chitosan nanocomplexes could induce a
normal cells but displayed much higher cytotoxicity to colorectal car higher expression of CD40, ICOSL, CD86, and MHC-II on the surface of
cinoma (Caco-2) and colon carcinoma (HT-29) cancer cells, which DCs, thus assisting in their maturation. The maturated DCs could pro
significantly reduced the incidence of side effects, promoting a safer and mote the secretion of pro-inflammatory cytokines, which would further
effective way to deliver nucleic acids for cancer immunotherapy. promote the proliferation of T cells and induction of Th1 differentiation
However, the experimental data from these chitosan-coated nano that could strengthen the anti-tumor immunity [191]. These
medicines were obtained at the cellular level and further evaluation of chitosan-based nanoparticles have demonstrated a high efficacy to
their in vivo delivery efficacy is still needed. Ali et al. [182] prepared deliver not only DNA or RNA, but also other different therapeutic agents
PEG-chitosan-lactate nanoparticles to load therapeutic siRNAs. In vitro to achieve synergistic effects between them and promote combinational
experiments demonstrated that these siRNA-loaded nanomedicines were therapy towards cancer treatment.
low toxic, stable in serum and could achieve controllable release of the
loaded siRNA within 60 h. Cellular and animal studies showed that these
PEG-chitosan-lactate nanomedicines could selectively target T cells at
the tumor site, enhanced T cell proliferation and reduced apoptosis.
3364
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
3.4. Chitosan-based nanosystems to deliver other types of cervical cancer [208], and breast cancer [209]. However, in vivo and
immunotherapeutic agents clinical trials are highly demanded to confirm these immune enhancing
effects towards cancer treatment. In addition to delivery of therapeutic
In addition to adjuvants, cytokines, and nucleic acids, there are other drugs, chitosan-based polymers have also the potential to deliver living
types of therapeutic drugs that could achieve the immunomodulatory cells to achieve immunotherapy of cancer. Tsao et al. [50]designed poly
effect for cancer treatment. Generally, cancer cells express a higher (ethylene glycol)-g-chitosan hydrogel to carry therapeutic T cells. They
amount of tumor-associated antigens than normal cells, and some of found that the chitosan-based hydrogels were compatible with T cells
these antigens are exclusively identified on tumor cells: tumor-specific and could retain their anti-tumor function. After loading T cells in the
antigens or neo-antigens [192,193]. But these antigens on tumor cells chitosan-based hydrogel, these T cells displayed a better therapeutic
are usually not directly recognized by the immune system due to im efficacy compared with controlled groups. It might be caused by the fact
mune evasion of tumor cells [194], thus delivery of exogenous that these hydrogels could provide an optimal pore size to enable better
tumor-associated antigens into cancer cells is a feasible approach for invasion of T cells [210]. This study provides a new way of delivering T
anti-tumor immune stimulation. However, administration of these cells for immunotherapy of cancer but this method is still in need of
“naked” antigens alone would result in their premature degradation in further studies, for example, to unmasking the mechanism of action for
the body fluid before they reach tumor sites. Concerns also arise that better efficacy.
non-targeting administration of these antigens would cause undesirable
systemic inflammatory reactions. Thus protective and selectively tar 3.5. The immunomodulatory effect of chitosan-based nanosystems
geting delivery platforms are needed to promote antigen-based cancer
immunotherapy [195,196]. Windberg et al. [197] applied poly Apart from delivery of immunotherapeutic agents, previous studies
peptide/Chit2DC (chitosan-deoxycholate) micelles to deliver an exoge have demonstrated that chitosan-based nanosystems without thera
nous MAGE-3 polypeptide antigen, which is a CD4+ and CD8+ T cell peutic drugs could also have immunomodulatory properties. It has been
epitope. In the animal model study, they demonstrated that these reported that chitosan-based nanosystems could significantly increase
nanoscale vaccines could induce the immune response towards MAGE-3 the secretion of IFN-γ by Th1 cells and stimulate the cell-mediated im
expressing tumor cells by increasing differentiation of cytotoxic T munity. Compared to chitosan oligosaccharides, the chitosan nano
lymphocyte (CTL) against MAGE-3 antigens, which would inhibit the particles showed an enhanced immunomodulatory effect (1.2–1.5-fold),
growth of tumor cells and promote apoptosis in tumor tissues. Another indicating the immunomodulatory effect could be tuned by aggregation
mannose-chitosan-stearic acid nanomicelles have also been developed of nanoparticles [211]. Wardani et al. [212] also applied chitosan
to deliver ovalbumin and CCR7 pDNA [49]. After these cargos-loaded nanoparticles to animal models and concluded that these nanoparticles
nanomaterials were applied to the tumor-bearing mice, they could could effectively stimulate immune responses and have a therapeutic
effectively promote the maturation of DCs and also induce the migration potential for immunotherapy.
of these immune cells to lymph nodes to generate an enhanced antigen A further study [61] examined the detailed tumor immunity induced
presenting process. A significant increase in the population of antitumor by chitosan-based nanosystems and it was found that they could stim
cytotoxic CD8+ T cells had been witnessed. Both novel chitosan-based ulate macrophages towards a pro-inflammatory profile, expressing less
nanomaterials have achieved safe and effective delivery of exogenous CD163 molecules and producing more secretory IL-12 p40 and TNF-α. In
antigens, and demonstrated the feasibility of applying tumor-associated addition, they discovered that these chitosan-based nanoparticles could
antigens in cancer immunotherapy. It is worth noting that a patent [198] stimulate DCs, increase their expression of co-stimulatory molecules and
has already been applied in which chitosan nanoparticles were utilized HLA-DR, promote the secretion of pro-inflammatory cytokines that
to deliver antigens. Their patent document demonstrated that the could stimulate antitumor effectors, and induced differentiation and
chitosan-antigen complex nanoparticles possessed superior immune activation of CD4+ and CD8+ T cells so as to modulate the whole tumor
response-stimulating effects and could be used in therapeutic immune response. Their in vivo study also confirmed these immuno
vaccination. modulatory results evidenced by inhibited growth of tumors and coun
Rajaei et al. studied the immunoregulatory effect of arteether teracted invasion of cancer cells. A more recent study [62] also achieved
delivered by folic acid-chitosan-Fe3O4 composite nanoparticles [199]. a similar immunostimulatory effect by application of chitosan-coated
These arteether-loaded chitosan-based nanomedicines were applied to copper oxide nanoparticles. In both in vitro study on breast cancer
4T1 cell lines and breast cancer-bearing mice and significant augmen cells (MCF-7) and cervical cancer cells (HeLa) and in vivo study on breast
tation in the production of cytokines IFN-γ and IL-4 was demonstrated. cancer (4 T1 cells induced) bearing mice, a great therapeutic efficacy of
Since IFN-γ and IL-4 could induce and activate downstream antitumor this nanomaterial has been demonstrated as it activated both Th1 and
effectors, the growth of 4T1 tumor cells was greatly inhibited and the Th2 cells, increased the production of pro-inflammatory cytokines and
tumor volume in the breast cancer-bearing mice was significantly expanded the CD4+ T cell population. Furthermore, a chitosan-based
shrunk. This experiment indicated that the folic acid-chitosan-Fe3O4 biopolymer, N-dihydrogalactochitosan, could synergize with photody
nanocomposite might be another promising arteether delivery platform namic therapy or cryoablation therapy to result in better antitumor ef
for cancer immunotherapy. Another promising immunotherapeutic ficacy. This synergetic effect of N-dihydrogalactochitosan was further
agent that could be used in cancer treatment is curcumin. It has been confirmed by its ability of direct tumor killing and prominent immune
reported that curcumin could affect several cell signaling pathways that modulation [213]. More recently, another chitosan micelle has been
are involved in tumorigenesis and cancer metastasis and has the po synthesized as antigen-capturing adjuvants that could achieve an
tential of converting Tregs into Th1 cells to avoid immune evasion of enhanced cancer immunity. It has been discovered that this
tumor cells and induce tumor killing responses [200–204], while the chitosan-based nanomedicine could effective target at tumor-draining
mechanism for the conversion is still unknown. One study has been lymph nodes and induce strong CD4+ and CD8+ T cell antitumor re
conducted to use chitosan-based mesoporous silica nanoparticles [205] sponses [115,214]. Based on previous positive findings, Moran et al.
to deliver curcumin to promote the immunotherapeutic efficiency [215] published a review to systemically examine the immunomodula
against cancer. Their in vitro results showed that these chitosan-based tory properties of chitosan polymers. In their review, they concluded
nanomedicines could effectively deliver curcumin and achieve stable that chitosan-based materials could activate both cGAS-STING DNA
and sustained controllable release of it in the U87MG glioblastoma sensing pathway and NLRP3 inflammasomes so as to induce differenti
tumor cells. Other studies also indicated that chitosan-based nano ation and activation of Th1 cells and suggested they could be applied as
systems could effectively deliver curcumin to tumor tissues thus increase a promising vaccine adjuvant for cancer immunotherapy.
their treatment efficacy in pancreatic cancer [206], colon cancer [207], Overall, chitosan-based nanomaterials have great potential to use in
3365
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
the immunotherapy of cancer. These nanomaterials could not only act as cells and macrophages could be activated and maturated to present
a nano-platform to carry immunotherapeutic agents to tumor tissues tumor killing properties and to produce pro-inflammatory cytokines
effectively and selectively so as to enhance anti-tumor immune re [226,227], immunotherapeutic drugs themselves are exogenous anti
sponses, but also display immunomodulatory effects as immune adju genic agents that could be recognized and eliminated by immune sys
vants themselves to induce tumor-killing immune responses. Despite tems before they reach tumor cells. That is partially the reason why
positive outcomes of their in vitro and in vivo studies, clinical trials are managing these immunotherapeutic agents alone usually results in a
required to examine in vivo safety and effectiveness of these chitosan- limited efficacy and often leads to unfavorable side effects [228–230].
based nanomaterials as potential immuno-therapeutics towards cancer HA, on the other hand, is a polysaccharide that demonstrates an
treatment. anti-inflammatory and anti-immunogenic function. It has been reported
that HA could provide an immunoprotective and immunomodulatory
4. Hyaluronic acid-based nanomedicines effect towards conjugated/encapsulated agents and protect them from
elimination by the immune system [137,231,232]. Although this
HA is negatively charged natural polysaccharides and they have been anti-inflammatory effect of HA may seem to be contradictory to the
widely used in the biological and medicinal field. It has a linear structure principles of immunotherapy, this effect results in a decrease in the
with repeated units of N-acetyl-D-glucosamine and D-glucuronic acid di- incidence of premature degradation of loaded drugs and a reduction in
saccharide bound via beta-linkages [216]. The hydroxyl, carboxylic and undesired immune responses. Since HA could selectively target CD44,
N-acetyl groups of HA allow further structural manipulation via chem LYVE-1 and RHAMM receptors that are usually overexpressed by tumor
ical reaction, which opens a door for broad applications of this material cells, HA-assisted delivery of exogenous immunotherapeutic drugs
[217]. Similar to chitosan, HA is a natural biomaterial that could be would increase their accumulation inside tumor tissues and selectively
found in many living organisms. HA is one of the main components of stimulate immune responses in the tumor microenvironment. Likewise,
the extracellular matrix, which are synthesized and secreted by inter HA could directly target some immune cells through CD44 targe
stitial cells such as fibroblasts [218]. While low-molecular-weight HA ting/binding [233]. HA, often as a subset to bind to CD44 on T cells
oligomers resulting from degradation by hyaluronidase was reported to (mouse), could inducibly stimulate PMA/ionomycin, CD3 antibodies
be immunostimulatory [214,219], HA polymers with a high molecular and specific antigens [234]. HA could also bind to CD4+ CD25+ T reg
weight have been demonstrated to possess great cytocompatibility and ulatory cells (human and mouse) and stimulate CD3+/− CD28 activation
biodegradability, low toxicity and no immunogenicity. Thus many [235]. These processes would result in an increased efficacy of loaded
studies have been conducted to develop HA-based medicines for immunotherapeutic drugs and decreased incidence of side effects
biomedical applications [220,221]. Many cancer cells overexpress (Fig. 3).
CD44, lymphatic vessel endothelial (LYVE)-1 receptors and receptor for
HA-mediated motility (RHAMM), which are HA-binding receptors [141,
4.2. Hyaluronic acid-based nanosystems to deliver tumor-associated
222], hence one approach to utilizing HA is to apply HA-based nano
antigens
materials as drug delivery carriers to selectively target tumor cells
[223–225]. Recently, the HA nanosystem-based targeting drug delivery
Using HA-based nanosystems to deliver tumor-associated antigens to
system has been developed for cancer immunotherapy.
enhance the immunity towards tumor cells is one of the most promising
applications of HA-based nanoparticles for cancer immunotherapy. The
4.1. Hyaluronic acid to induce immunoprotection use of HA-based nanomaterials was reported to extend the release pro
file of administrated immunomodulatory agents, thus effectively
Although immunotherapy of cancer diseases requires stimulation of enhancing their therapeutic efficacy [237]. Although several
patients’ immune responses so that certain immune cells such as DCs, T tumor-associated antigens [238,239] are helpful in cancer
Fig. 3. Hyaluronic acid-coated nanomedicines could selectively target at CD44+ cells, effectively deliver loaded drugs into the cytoplasm and promote the activation
of antigen presenting cells. Reproduced with permission from Ref. [236]. Copyright 2019 American Chemical Society.
3366
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
immunotherapy, ovalbumin (OVA) is the most widely used one due to its tissues [246]. Using this method, these PEI-HAase nanomaterials could
bioavailability and therapeutic efficacy [240,241]. However, similar to successfully deliver both OVA and CpG to induce a better efficacy of
other soluble antigens, administrating OVA alone usually does not result immunotherapy. This method sacrifices the targeting ability of HA but
in a satisfactory therapeutic effect. This is mostly caused by its intrinsic utilizes the penetrating ability of HAase to achieve enhanced intracel
immunogenicity and an insufficient dose delivered into immune cells. lular uptake and a sufficient amount of therapeutic agents in tumor site.
Thus it is important to develop delivery platforms to aid in in vivo de This concept has also been applied in another study [247]. However,
livery of these tumor-associated antigens [242]. Recently, a novel further examinations are required to elucidate which method, HA or
multifunctional micellar platform, consisting of self-assembled poly HAase-assisted delivery, could achieve a better immunotherapeutic
ethylenimine (PEI), vitamin E succinate (VES), HA and PEGylated HA efficacy.
(mPEG-g-HA), was introduced to load OVA to form immunotherapeutic Besides HA (ase)-PEI nanosystems, nanosystems made from HA, OVA
nano-micelles (OVA@mPEG-g-HA/VES-g-PEI) [243] (Fig. 4). It has been and gold (Au) particles have also been applied for immunotherapy [51]
discovered that HA in these micelles could decrease the cytotoxicity of (Fig. 5). It has been reported that after triggering with laser irradiation,
the attached PEI by neutralizing its positive charge and also assist in the these nanomaterials could achieve a similar immune stimulation process
targeting delivery of the nanomedicines into HA receptor-overexpressed to enhance the tumor immunity. During this process, near infrared (NIR)
tumor cells. This HA receptor-targeted delivery would further increase laser irradiation could trigger not only the rupture of endosomes and
cellular uptake of the micelles into tumor cells. Once this endocytosis lysosomes, but also the production of ROS which could enhance the
process is completed, hyaluronidases would degrade the HA shell of the proteasome activity and boost the antigen presenting process [248].
micelles so as to expose the inner OVA-loaded PEI structures. This Moreover, these loaded OVA and NIR irradiation generated ROS could
process could also benefit for the endosome/lysosome-mediated OVA promote the enhancement in CD8+ T cells-mediated anti-tumor immune
release and then promote antigen presentation. These responses, endowing this HA-Au nanomaterial as an excellent platform
OVA@mPEG-g-HA/VES-g-PEI-treated tumor cells could express MHC-I candidate to boost antitumor immunity, but ensure the targeting de
peptide epitope OVA257–264 on their surfaces so that DCs could recog livery of antigens with negligible toxicity. Alternatively, pH-sensitive
nize and then trigger differentiation and activation of antigen-specific HA derivative-modified liposomes have been developed to achieve tar
CD8+ T cells to achieve the tumor killing responses [243]. geted delivery of exogenous antigens to DCs [236]. These
However, although these HA-based micelles could promote the anti- antigen-loaded nanomaterials could increase the production of
tumor immunity, this immune stimulatory effect does not last longer, anti-tumor cytokines by Th1 cells and induce strong anti-tumor re
and the long-term immune enhancement is still relatively weak [244, sponses, leading to effective tumor growth inhibition in the
245]. Thus future modification of these micelles is needed to extend the tumor-bearing mice.
stimulatory time. Interestingly, another PEI-derived nanoparticles have Although above studies have explored the use of HA-based nano
also been adopted to deliver therapeutic agents. Instead of HA, these systems to deliver sole antigens, a recent study [249] attempted to
nanoparticles were combined with HAase to break down extracellular employ levodopa and poly (ε-caprolactone-co-lactide)ester-functional
HA so as to increase the permeability of these nanoparticles to tumor ized HA hydrogels to co-deliver both OVA and granulocyte–macrophage
Fig. 4. Synthesis of OVA-loaded mPEG-g-HA/VES-g-PEI micelle (a) its therapeutic efficacy on cancer immunotherapy. This antigen loaded HA-based nanomedicine
could targetedly deliver OVA into the tumor cells and induce an enhanced cytotoxic T lymphocyte (CTL)-mediated immune response (b). This HA-based nano
medicine could effectively induce the maturation of DCs (c) and increase the expression of tumor-associated antigens to activate CTLs (d), thus killing tumor cells (e).
*p < 0.01 compared with the OVA group in (c) and (d); *p < 0.01 compared with all other groups in (e). Reproduced with permission from Ref. [243], Copyright
2019 Elsevier.
3367
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
Fig. 5. Synthesis of HA-OVA-AuNPs complexes (a) and its therapeutic efficacy on cancer immunotherapy. This HA-based nanoparticle could be used as nanovaccine
and effectively enhance both MHC-I and MHC-II antigen presentation process (b), thus demonstrating a comprehensive anti-tumor immune promoting ability to
significantly inhibit the tumor growth (c). **p < 0.01; ***p < 0.001. Reproduced with permission from Ref. [51]. Copyright 2018 WILEY-VCH Verlag GmbH &
Co. KGaA.
colony-stimulating factor (GM-CSF). The stable release of the loaded memory T cells to secret antitumor IFN-γ when encountered with spe
drugs from the hydrogels suppressed the growth of tumor cells. It is cific antigens. Importantly, this inexpensive HA nanoparticle-based de
worth noting that subcutaneous injection was utilized as the adminis livery system could enhance the bioavailability of CpG ODNs and reduce
tration route, which could effectively avoid potential side effects due to the risk of side effects caused by positively-charged cationic lipids,
systemic drug administration. The HA-based nanomaterials were also which made these adjuvant-loaded nanomedicines safer and more
employed to deliver antigens in a needle-free administration route. affordable for cancer immunotherapy. In addition to delivering CpG
Bussio et al. [250] found that OVA-loaded HA nanovaccines could be ODN with Poly I: C, in another recent study [258], HA-based nano
absorbed through skin thus no needles were involved in this process to particles were employed to load CpG along with another therapeutic
avoid risks of injuries and infections. However, more studies are needed hypoxia inducible factor: signaling inhibitor Acriflavine (ACF)
to confirm the in vivo therapeutic efficacy of the HA-based nanovaccines [259–261]. In this study, HA coated metal–organic framework-based
through this needle-free approach. nanoparticles were synthesized through H2TCPP and zirconium ions
as a delivery platform (PCN-ACF-CpG@HA). Due to the binding ability
of HA, these PCN-ACF-CpG@HA could selectively target cancer cells
4.3. Hyaluronic acid-based nanosystems to deliver other
which usually overexpress CD44 receptors. The synergistic therapeutic
immunotherapeutic agents
effect of the loaded CpG and ACF has been demonstrated by inducing
significant antitumor immune responses and inhibiting growth and
Besides tumor-associated antigens, HA-based nanosystems have also
metastasis of tumor mass. This opens a new avenue to using HA-based
been employed for delivering other types of therapeutic agents to pro
nanomedicines for cancer immunotherapy at a low cost.
mote immunotherapy of cancer diseases.
Although chemotherapy and immunotherapy are two different
CpG oligodeoxynucleotide (CpG ODN) and polyinosinic-
methods for cancer treatment, different types of therapeutic agents with
polycytidylic acid (Poly I: C) are both members of pathogen-associated
distinct antitumor mechanisms could be combined to achieve a greater
molecule pattern (PAMP) families that could be recognized by TLR-9
tumor inhibition efficacy. Nevertheless, co-delivery of different types of
and -3. The former is usually found in bacteria and the latter is a syn
therapeutic agents together and maintenance of their own biofunctions
thetic analogue of double-stranded RNAs from viral-infected mamma
remains a challenge. Lv et al. [52] used HA-based nanoparticles to
lian cells [251–254]. The combination of these two substances was
deliver both chemotherapeutic and immunotherapeutic agents to ach
reported to demonstrate synergistic effects to stimulate T cell responses
ieve a combinational therapy. They used a HA-paclitaxel (HA-PTX)
and thus were explored as promising immune adjuvants for immuno
prodrug and marimastat (MATT)-loaded thermosensitive liposomes
therapeutic vaccines [255,256]. Despite their effectiveness, this com
(MATT-LTSLs) to from self-assembled nanoparticles and applied them to
bination would result in severe side effects. Using phosphorothioate to
the treatment of breast cancer. The results showed that these
modify the phosphodiester backbone could be one possible solution, but
CD44-targeting nanomedicines could selectively deliver the loaded
this solution would inevitably increase the cost for this therapy. Liu et al.
drugs (HA-PTX and MATT) into the tumor microenvironment, inhibit
[257] developed HA-modified cationic lipid-poly (lactic-co-glycolic
the matrix metalloproteinase (MMP) and expression of TGFes and TNC,
acid) (PLGA) hybrid nanoparticles to deliver both Poly I:C and natural
and suppress tumor growth, metastasis and angiogenesis. This provides
phosphodiester CpG ODNs. They discovered that these adjuvant-loaded
another effective way to promote the nanomaterial-based combination
nanomedicines could significantly induce the maturation of DCs and
cancer therapy. Camptothecin (CPT) is a chemotherapy drug that sup
increase the population of antitumor CD4+IFN-γ+, CD8+IFN-γ+ and
presses the activity of DNA enzyme topoisomerase I in tumor cells so as
CD8+CD107a+ T cells, while inhibiting the development of immuno
to achieve remarkable inhibition effect against tumor cells [262,263].
suppressive myeloid-derived suppressors cells. Furthermore, they found
Sun et al. [264] found that CPT-conjugated HA nanomedicines could
these adjuvant-loaded nanomedicines could increase the population of
have synergistic effects to increase the immunotherapeutic efficacy.
memory CD4+ and cytotoxic T cells and shorten the intervals of these
3368
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
Although 4T1 tumor cells usually express a low level of PD-L1 and they understanding of tumor immunity and the emergence of new thera
do not have great response to the anti-PD-L1 therapy, these peutic concepts and new immunotherapeutic drugs, more applications
CPT-conjugated HA nanomedicines could sensitize the tumor microen of HA-based nanomedicines for cancer immunotherapy will be further
vironment so as to enhance the immune checkpoint blockade therapy. discovered and evaluated.
The effect of improving the sensitivity of the tumor microenvironment to
immunotherapeutic drugs by a chemotherapeutic agent was also 5. Dextran-based nanomedicines
confirmed from previous combinational therapeutic studies [265–267].
It is believed that the incorporation of chemotherapeutic agents could Dextran is a polysaccharide with an α-1,6-glycosidic bonding chain.
enhance the proliferation of long-term and effective tumor anti It is water-soluble with great biocompatibility and biodegradability and
gen–specific T lymphocytes, thus achieving a synergic antitumor effi has low toxicity and non-immunogenicity. Modifications of dextran
cacy from chemotherapy and immunotherapy [268,269]. However, the through the hydroxyl groups in the backbone chain would allow in
detailed mechanisms remain to be discovered. teractions with selective receptors and other therapeutic ligands or
Herbal extracts such as curcumin and baicalin are another type of agents, and remain their biocompatibility, which makes it another
potential immunotherapeutic agents, which had been delivered by using excellent candidate for nano-delivery of therapeutic drugs to achieve
HA-derived nanoparticles delivery platforms to achieve effective cancer efficient treatment of diseases [282–285]. Furthermore, it has been
chemotherapy [270–273]. Recently, the HA-based nanoparticles were indicated that dextran could be utilized as an alternative for PEGylation
employed to deliver herbal extracts so as to promote cancer immuno to prevent interactions of nanomedicines with opsonin [147,286]. Due
therapy. Wang et al. [274] developed quercetin-dithiodipropionic to these advantages of dextran, it is widely used for the formation of
acid-oligomeric hyaluronic acid-mannose-ferulic acid (Que-S-S-oHA- nanomedicines. Recently, a number of dextran-based nanomedicines
Man-FA, QHMF) to form dandelion-like nanomicelles for delivering both have been developed for cancer immunotherapy.
curcumin and baicalin into tumor tissues. They demonstrated that these
drug-loaded nanomicelles could easily penetrate through vascular bar 5.1. Dextran-based nanosystems to deliver immunotherapeutic agents
riers and enter tumor tissues. Further examinations revealed that these
drug-loaded nanomicelles could reprogram the tumor-associated mac Wang et al. [54] developed a microneedle patch to deliver anti-PD-1
rophages from a pro-tumor M2 phenotype into a tumor-killing M1 antibody into tumor tissues to achieve immunomodulation (Fig. 6). In
phenotype, thus exhibiting a higher tumor growth inhibition effect their study, the microneedle patch consisted of biocompatible HA and
compared with free curcumin and baicalin. Moreover, increasing pH-sensitive, anti-PD-1 and glucose oxidase-loaded dextran nano
secretion of cytokines TNF-α and IL-6 indicated that baicalin could be particles. The drug-loaded nanoformulations were applied to the treat
employed as an adjuvant to enhance the immunomodulatory efficacy. ment of melanoma in model mice. The microneedle patch could
Recently, hyaluronate-polylactide (HA-PLA) nanoparticles have also effectively deliver PD-1 antibodies into the tumor microenvironment
been applied to encapsulate curcumin [275]. Instead of promoting and achieve sustained release of them. They discovered that the thera
antitumor immunity, these drug-loaded nanomedicines could repro peutic efficacy of this delivering strategy was greater than that of
gram macrophages from a M1 phenotype to a M2 phenotype so as to administrating free anti-PD-1 antibodies. This microneedle
reduce the production of pro-inflammatory cytokines and suppress the patch-assisted immunotherapy enhancement technology has already
inflammation response. Although some studies stated that curcumin been filed as a patent by this research group [287]. Another immuno
could promote inflammatory immune responses towards cancer [276, therapeutic agent (type B CpG DNAs) has been conjugated to dextran
277], curcumin has also been widely used in the treatment of autoim polymers and the formulated nanomedicines were applied to
mune diseases which rely on the inflammatory immune inhibition effect tumor-bearing mice [288] (Fig. 7a). These dextran-CpG conjugates were
of curcumin [278–280]. Thus the therapeutic mechanism of action of discovered to significantly enhance the antigen presenting process and
these curcumin-loaded HA-based nanomedicines for tumor immune increase the population of CD8+ T cells to achieve improved tumor
responses still remains controversial. killing immune responses. Yuba et al. [289] suggested that dextran and
Besides the above-mentioned therapeutic agents, a US patent docu its derivatives could be added to construct pH-sensitive liposomes that
ment [281] reported HA-based layer-by-layer nanoparticles to load cy could increase the safety and efficacy of the delivered antigens. More
tokines for the treatment of cancer. The proposed nanosystems were recently, Shin et al. [290] introduced a carboxymethyl dextran
composed of a liposomal core, a bilayer poly (L-arginine) coating and an (CMD)-based polymeric conjugate to deliver exogenous antigens
HA and poly (L-glutamic acid) polymer coating. These nanoparticles (Fig. 7b). Ovalbumin (OVA) was loaded into this polymeric conjugate
were applied to encapsulate and deliver cytokines such as IL-12 and with CMD as the backbone. This drug-loaded nanomaterial was applied
found that this cytokine-loaded nanomedicine formulation could to tumor-bearing mice. Compared with free OVA, this CMD-OVA con
significantly increase the secretion of antitumor IFN-γ by splenocytes, jugate could target tumor tissues and accumulate there with a prolonged
and the tumor growth of both MC38 tumor-bearing and HM1 retention period, effectively promoting the antigen presentation process
tumor-bearing mice had been effectively suppressed. However, although and stimulating greater anti-tumor immune responses. In addition to
cytokines have been greatly appreciated in cancer immunotherapy these immunotherapy strategies, a few studies on dextran-based nano
[168], the strategy of using HA-containing nanoparticles to deliver cy systems have been attempted to deliver therapeutic agents to achieve
tokines to enhance the immunotherapy efficacy directly is still at the reprogramming of tumor-associated macrophages. In a recent study
early stage. Recently HA-based hydrogels were developed to deliver conducted by Wang et al. [56], they synthesized a novel
artificial T cell stimulating matrix, which is not cytokines but a mixture erythrocyte-cancer cell hybrid membrane camouflaged pH-responsive
of substances (ECM, Anti-CD28, MHC, etc) crucial for T cell activation. dextran-grafted-poly (histidine) copolymer micelle to deliver BLZ-945,
Antitumor immune responses stimulation was achieved [53]. Further a CSF-1R inhibitor specific for tumor-associated macrophages. Their
investigations are needed to evaluate the efficacy and clinical applica preclinical results showed that this nanomedicine could effectively
bility of this immunotherapy method. reprogram the tumor microenvironment with increased M1 macro
Overall, HA-based nanosystems, with their selective targeting ability phages and elevated CD8+ T cells, thus resulting in inhibition of tumor
to tumor cell receptors and their immunoprotective effects to protect the growth. Besides, dextran-based nanomaterials have M2-TAMs targeting
conjugated/enveloped therapeutic agents, provide a simple and effec ability. Huang et al. developed a dextran-based tumor-targeting nucleic
tive way to deliver exogenous drugs with minimal side and toxic effects, acid delivery system derived from a tumor
which makes them a very promising candidate for delivery of immu microenvironment-responsive carrier and a TAMs-specific receptor
notherapeutic agents towards cancer immunotherapy. With a deep [291]. All these recent studies demonstrated dextran-based
3369
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
Fig. 6. The structure of anti-PD-1therapeutics loaded dextran-based nanoneedle patch MN-Gox-aPD1 (a: Schematic diagram; b: SEM image) and its therapeutic
efficacy on cancer immunotherapy. This nanoneedle patch could effectively inhibit the growth of tumor tissues and reduce the tumor size, resulting in a decreased
tumor signal in bioluminescence imaging in vivo (c, d). *p < 0.05. Reproduced with permission from Ref. [54]. Copyright 2016 American Chemical Society.
nanomaterials as an excellent candidate for targeting delivery of various demonstrated the intrinsic immune stimulating properties of dextran-
immunotherapeutic agents with low apparent toxicity. On the basis of based nanosystems. Fontana et al. designed porous silicon@acetalated
these positive results, further studies could be conducted to optimize the dextran@cancer cell membrane (TOPSi@AcDEX@CCM) nanovaccines
structure of these dextran-based nanomaterials for delivering different for immunotherapy of cancer [57]. In their study, thermally-oxidized
therapeutic agents to achieve a combinational therapy with better out porous silicon was coated by acetalated dextran polymers and the
comes. In this context, Bauleth-Ramos et al. [55] used nanostructure was co-extruded with cancer cell membrane particles to
spermine-modified acetalated dextran nanosystems for co-delivery of form core-shell-structured nanovaccines. These nanovaccines could
cis-imidazoline nutlin-3a, a chemotherapeutic agent, and cytokine significantly increase the expression of CD80 and CD86 on antigen
GM-CSF, an immunotherapeutic agent. These dextran-based nano presenting cells and induce T cells activation on both KG1 and BDCM
medicines could release the loaded cargos in a pH-dependent manner cells, which could result in a greater immunostimulation efficacy. In
and also help in promoting endosomal/lysosomal escape of the deliv addition, the authors noticed that these nanovaccines could induce the
ered drugs. Their in vitro study revealed that these nanomedicines could polarization of T cells towards Th1 cells to secret a greater amount of
selectively target wild-type p53 cancer cells and exhibit killing effects on anti-tumor IFN-γ and IL-2 as well. Furthermore, this nanovaccine system
them, but displayed no toxicity to immune cells. These nanomedicines could also be used to deliver exogenous antigens such as Trp2 to further
could also maturate DCs by increasing the expression of CD83 and CD86 improve the immunotherapeutic efficacy [57]. Moreover, Bamberger
on their surfaces, which would enhance their antigen presenting abili and their colleagues [63] developed a linear low molecular-weight
ties, induce proliferation and activation of the down-stream immune dextran attached with spermine-modified acetalated dextran nano
cells like cytotoxic CD8+ T cells, promote the secretion of IL-1β and particles. These surface-modified dextran nanosystems could selectively
reduce the expression of IL-10. Based on these findings, acetalated target DCs and trigger the activating signaling pathways of DCs, which
dextran nanoparticles-loaded injectable alginate cryogel was also could significantly stimulate activation of these antigen presenting cells.
introduced as an in situ cancer vaccine [292]. This peritumoral inject A similar immunostimulatory effect was also found for B cells and
able cancer vaccine was reported to demonstrate cytotoxicity towards macrophages [63,293,294]. However, in this study, it was also indicated
tumor cells and induce immunogenic cell death, which potentially that these dextran-based nanoparticles would induce cytotoxic effects
decreased cancer recurrence. on immune cells, which is the main drawback of applying them for
cancer immunotherapy. Further studies are needed to reduce the cyto
toxicity of these dextran-based nanosystems to immune cells and
5.2. The immunostimulatory effects of dextran-based nanosystems discover more clinical applications for cancer immunotherapy.
3370
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
Fig. 7. Synthesis of type B CpG DNAs conjugated dextran-based nanoparticles (a) and carboxymethyl dextran-ovalbumin (b) and their therapeutic efficacy on cancer
immunotherapy. Through improving the reorganization of tumor associated antigens by antigen presenting cells (c), these dextran-based nanomedicines could
effectively enhance anti-tumor immunity thus resulting in the decreased tumor sizes (d). **p < 0.01. Reproduced with permission from Refs. [288,290]. Copyright
2017 American Chemical Society and 2018 BMC Open Access, respectively.
3371
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
Fig. 8. Preparation of MAN-ALG (a) and ALG = OVA (b). ALG/ALG = OVA nanoparticles could significantly inhibit tumor cell proliferation by displaying a lower
tumor volume (c) and a smaller tumor weight (d). **p < 0.01. Reproduced with permission from Ref. [58], Copyright 2017 Elsevier.
explored in nano-scale drug delivery systems. Kyogoku et al. [309] these chondroitin sulfate derivative-modified liposomes could selec
discovered that cholesteryl-pullulan-melanoma antigen gene-A4 nano tively deliver tumor-associated OVA antigens into DCs and increase the
gels were effective in promoting tumor immune responses and they have production of anti-tumor cytokines (Fig. 9). Their in vivo study also
entered a phase I + II clinical trial as a vaccine for cancer treatment. demonstrated a significant tumor growth inhibition effect of the
However, frequent administration of this vaccine may result in some OVA-chondroitin liposomes on growth and metastasis of tumors in the
undesirable changes in the anti-tumor immunity. Miura et al. [310] mice. Liu et al. also synthesized a chondroitin sulfate-based lipoic acid
designed cholesterol-bearing pullulan (CHP) self-assembly nanogels to nanoplatform that was triple-responsive to redox, enzyme and ultra
deliver OVA antigens. It has been discovered that this nanogel could sound [59]. By conjugating Ce6 and loading docetaxel, this nano
selectively deliver OVA into the lymphatic system to be recognized by conjugate could not only achieve a combinational therapy of
APCs. The anionic charge of the nanogel contributed to the enhanced sonodynamic therapy and chemotherapy, but also recruit cytotoxic
interactivity of the loaded OVA with APCs, which induced strong lymphocytes into tumor tissues, resulting in an enhanced antitumor
adaptive immune responses towards tumor cells with enhanced activa immune response. During the sonodynamic therapeutic process with
tion of the Th1 immune pathway and the increased population of CD8+ this nanomedicine, ROS were generated in the tumor tissues. These ROS
T cells. A previous study indicated that pullulan-based nanosystems would activate innate and adaptive immune responses, promote the
could exhibit high affinity towards asialoglycoprotein receptors on he antigen release from tumor cells that DCs could recognize to induce and
patocytes [311], and they could be a promising targeting delivery activate CD8+ T cells [59].
platform for hepatocellular carcinoma. More studies on directly β-cyclodextrin (CD), a low molecular-weight polysaccharide, has
applying pullulan-based nanomedicines for hepatocellular carcinoma been also reported to be applied in the immunotherapeutic drug delivery
treatment are still demanded. [316,317]. It has seven glucopyranose units with a polar and hydro
Chondroitin sulfate is another polysaccharide that could be used in philic external surface and a relatively nonpolar and hydrophobic in
the drug delivery system. It has been reported that modified chondroitin ternal surface, and this structure feature has been considered for
sulfate could exhibit low hydrophilicity to protect the loaded drugs since delivery of immunotherapeutic agents. In a recent study, β-cyclodextrin
the structure of chondroitin sulfate is similar to that of HA [312–314]. was covalently reacted with lysine to form crosslinked CD nanoparticles
Okubo et al. [315]developed chondroitin sulfate-based pH-responsive (CDNPs) [60]. These nanoparticles were able to load R848 and deliver it
liposomes to deliver OVA antigens for tumor therapy. They found that to tumor tissues. These drug-loaded CDNPs could selectively target
3372
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
Fig. 9. Synthesis of chondroitin sulfate derivatives that could modify liposomes (a) and bioreaction of chondroitin sulfate derivatives-modified liposomes for
stimulation of cancer immunity in subcutaneous tissue (b) and tumor tissue (c). Reproduced with permission from Ref. [315], Copyright 2019 American Chemi
cal Society.
tumor-associated macrophages and induce alteration of macrophages immunotherapy has been studied. Xia et al. [326] utilized
from a pro-tumor M2 phenotype into an anti-tumor M1 phenotype, low-molecular-weight heparin to coat dendrimer-based core-shell
resulting in suppression of tumor growth and protection of animals nanocomposites to deliver CpG ODNs for cancer
against tumor recurrence. Furthermore, it has also been discovered that chemo-immunotherapy. According to their study, this
these R848 loaded-cyclodextrin nanomedicines along with anti-PD-1 nanomaterials-based combination therapy could effectively enhance the
agents could demonstrate synergistic effects to achieve an enhanced maturation of DCs and increase the population of cytotoxic CD8+ T cells.
immunotherapeutic efficacy [60]. The coating of low-molecular-weight heparin would prevent the
Pectin is a dietary component in plant-based foods such as fruits and epithelial-mesenchymal-like transition of tumor cells and suppress their
vegetables. It serves as one main component of plant cell walls from inhibition effect by damaging the arrangement of their actin cytoskel
where it can be extracted. Modified pectin materials are reported to etons, thus presenting as a promising candidate for chemo- and immu
possess excellent biological properties which have been applied in the notherapy of cancer diseases. Another study applied a similar core-shell
treatment of cancer [318,319]. Hira et al. [320]synthesized pectin-guar nanostructure with gambogic acid, heparin and CpG ODN to treat he
gum-zinc oxide nanocomposites that could be used in cancer immuno patocellular carcinoma [327]. It has been reported that these nano
therapy. In their study, these nanocomposites were shown to induce medicines could effectively increase the population of cytotoxic T cells,
activation of tumor cell killing processes and improve the tumor cell induce differentiation of Th1 cells and promote Th1-based antitumor
killing capacities of peripheral blood lymphocytes. It increased the immune responses, which contribute to cancer immunotherapy.
production of anti-tumor cytokines IFN-h, IL-2 and TNF-α which further With their excellent biocompatibility and targeting ability, inulin
led to the killing of tumor cells and inhibition of tumor growth. A deep and their derivatives have also been studied as a nanoplatform to deliver
understanding of pectin-based nanomaterials and their immunotherapy therapeutic agents to target certain cells or tissues [328–330]. Although
applications will pave the way for clinical trials. the application of inulin-based nanomaterials to cancer immunotherapy
Another polysaccharide heparin has a long history of uses in clinical is still at a very early stage, inulin acetate-based polymers have been
practice [321–323]. Its anticoagulation, anti-platelet aggregation and reported to deliver exogenous antigens into DCs effectively [331]. These
anti-thrombus functions make them versatile for a variety of clinical OVA-loaded inulin acetate-based polymers could be selectively recog
conditions [324,325]. Recently, application of heparin in cancer nized by DCs through TLR4 and induce the maturation of these cells. In
3373
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
vivo experiments demonstrated that these nanomedicines could signifi nanoparticles to achieve radiotherapeutic applications [65]. Through
cantly increase the titers of serum antibodies such as IgG1 and IgG2a and this conjugation, these nanoparticles could not only increase the sensi
the amount of cytokines like IL-4 and IL-10 to present strong immune tivity of radiotherapy, but also prominently promote activation and
responses towards tumor cells. By managing these OVA-loaded inulin maturation of DCs and increase the population of CD8+ T cells, which
acetate polymers, inhibition of metastasis of melanoma cells to lungs has further led to increased secretion of pro-inflammatory cytokines to
been witnessed in the experimented animals [331]. The targeting ability enhance antitumor immune responses. These GLP-Bi inhibited the
to DCs and biosafety of these inulin-based nanosystems make them safe tumor growth simultaneously and suppressed metastasis [65]. In a more
and effective carriers to deliver immunotherapeutic agents. recent study [332] galactoxyloglucan extracted from Tamarindus indica
Herbal extraction polysaccharide is another substance which has seeds was utilized to improve the stability and pH tolerance of copper
recently been applied in cancer immunotherapy (Fig. 10). Zhang et al. nanoparticles. According to this study, these
[64] used polysaccharide extracts from a natural herb (Ganoderma galactoxyloglucan-processed nanomedicines could significantly induce
lucidum) and gold to form gold-Ganoderma lucidum polysaccharide antitumor immune responses so as to reduce the tumor burden. Their
(GLP-Au) nanoparticles. Through their in vitro experiments, these excellent biodegradability, biocompatibility and immune stimulatory
GLP-Au nanoparticles could effectively promote the maturation of DCs effect warrant further investigations of their applications in cancer
and increase the transcription of anti-tumor cytokines. These nano immunotherapy. Astragalus membranaceus extracted polysaccharide is
systems also induced differentiation and activation of CD4+ and CD8+ T another herbal polysaccharide to be synthesized as nanomedicines for
cells. In vivo studies revealed that GLP-Au nanoparticles could signifi cancer immunotherapy [333]. These herbal nanomedicines induced the
cantly suppress tumor growth and its pulmonary metastasis. A signifi radiation-induced abscopal effect and promoted the systemic anti-tumor
cant increase in the number of CD4+/CD44+ memory T cells towards immunity. Anti-tumor immune memory has also been enhanced so that
tumor immunity has been also witnessed. These Ganoderma lucidum not only primary tumor growth has been inhibited, but growth of sec
polysaccharide has also been used to conjugate bismuth sulfide (GLP-Bi) ondary tumor remote to primary lesions was also suppressed. These
Fig. 10. Synthesis of GLP-Bi nanoparticles (a) and GLP-Au nanoparticles (b). GLP-based nanoparticles could effectively inhibit tumor growth (c) through the
interaction with immune cells (d). Reproduced with permission from Refs. [64,65]. Copyright 2018 Elsevier and 2019 American Chemical Society, respectively.
3374
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
natural herb-based nanomedicines may be feasible for both radio anti-angiogenic and immunosuppressive to 20 kDa to be angiogenic,
therapy and immunotherapy against cancers. More recently, cationic immune-stimulatory and inflammatory. The immunotherapeutic effect
polysaccharide derived from Lepidium meyenii Walpers has also been of HA-based NPs with different molecular weights should be investi
proven effective for tumor immunotherapy. This herb-extract nano gated in clinical trials [214]. Furthermore, clinical trials for cancer
medicine could re-educate tumor associated macrophages to M1 immunotherapy have their own unique considerations, and clinical trial
phenotype, which alter the immunosuppressive tumor microenviron designs should be tuned for these polysaccharide-based nanomedicines.
ment into strong antitumor immune-activated, leading to the effective For example, due to safety concerns of immunotherapeutics, the clas
tumor growth inhibition [66]. Nevertheless, since many herbal extracts sical phase I 3 + 3 dose escalation may not be appropriate for continuous
were reported to display short-term or long-term toxicities, whether assessment of the relationship between their efficacy and toxicity.
these herbal polysaccharide-based nanomaterials would generate tox However, an adaptive and seamless phase I/II combined trial with
icities to normal tissues, especially in the long-term, is still in need of multiple cohorts and continuous assessment of their efficacy and toxicity
long-term assessments. is recommended [336]. Furthermore, patient selection, clinical mea
surements, administration protocols, endpoint determination and anal
8. Conclusion and future perspectives ysis methods should be adjusted and harmonized in order to obtain safe,
effective and regulatory body-acceptable clinical trial results
In summary, this review discussed recent progress of polysaccharide- [336–339]. The issues mentioned above may inspire future inves
based nanomaterials and their applications in cancer immunotherapy. tigations/explorations on the development and improvement of
These natural or modified nanomaterials, with their excellent physico polysaccharide-based nanomaterials, and these immunotherapeutic
chemical and biological properties, could be used as nano-carrier plat drug formulations would become mature for clinical cancer treatment.
forms to deliver immunotherapeutic agents such as adjuvants,
cytokines, nucleic acids, and exogenous tumor-associated antigens. The Declaration of competing interest
polysaccharide nanomaterial-based drug delivery method could help to
achieve targeting delivery of immunotherapeutic agents to immune cell The authors declare no conflicts of interest.
subtypes and effectively improve the therapeutic efficacy of the loaded
agents. Some of these polysaccharide-based nanomaterials could even Acknowledgement
play a role as an immunomodulatory agent in addition to enhancing the
immunotherapeutic efficacy. This work was supported by the National Natural Science Foundation
Despite these encouraging outcomes of polysaccharide-based nano of China (51873120, 51673127, 81621003), National Science and
materials for immunotherapy, a few key challenges of these Technology Major Project of China (2017ZX09304023), Ruilong Sheng,
polysaccharide-based nanomaterials need to be overcome, such as Helena Tomás and João Rodrigues appreciate the support from
rational modification of polysaccharides, quantitative control of their Fundação para a Ciência e a Tecnologia (Base Fund UIDB/00674/2020,
molecular weight and the degree of modification, and incorporation of CQM, Portuguese Government Funds) and ARDITI-Agência Regional
multi-stimuli-responsive moieties to achieve smart and programmable para o Desenvolvimento da Investigação Tecnologia e Inovação through
immunotherapeutic effects. Especially, some essential aspects of the project M1420-01-0145-FEDER-000005-Centro de Química da
polysaccharide-based nanomaterials in immunotherapy are waiting to Madeira-CQM+ (Madeira 14–20 Program) and ARDITI-2017-ISG-003.
be investigated in future studies, including: 1) mechanistical studies of
polysaccharide-based immunotherapeutics towards “precision” immu Abbreviations
nomedicines. For efficient immune cell-activation and tumor targeting,
the mechanisms of polysaccharide-based nanoformulations and their ACF Acriflavine
immune-response properties need to be systematically elucidated, such APC Antigen presenting cell
as quantitative activation of immune cells (e.g. DCs, T cells, B cells and ATP Adenosine triphosphate
macrophages), intracellular localization of these nanoformulations and Caco-2 Colorectal carcinoma
intercellular localization of these immune-substances, especially, CAR-T cell Chimeric antigen receptor T cell
related up/down-regulation of immune-responsive genes, proteins/en Ce6 Chlorin e6
zymes; 2) Polysaccharide-based combinational cancer immunotherapy. CMD Carboxymethyl dextran
Since tumorigenesis mechanisms differ individually, a single therapeutic CpG ODN Oligodeoxynucleotide
agent may not be able to trigger enough immune responses to achieve CPT Cptothecin
satisfactory outcomes for cancer treatment. Therapy by combining CTL Cytotoxic T lymphocyte
multiple immunotherapeutic agents, namely combinational immuno DC Dendritic cell
therapy, is becoming an important strategy for tumor/cancer treatment. DNase Deoxyribonuclease
Some aforementioned studies have attempted to apply polysaccharide- FDA Food and Drug Administration
based nanomaterials to deliver two different types of therapeutic Fluc Luciferase
agents for combinational therapy, but up to now, the number of related GCS Glycol-chitosan
studies is very scarce. Moreover, how different immunotherapeutics are GM-CSF Granulocyte–macrophage colony-stimulating factor
combined rationally, quantitatively, compatibly and synergistically into HA Hyaluronic acid
one nanosystem to achieve high-performance combinational immuno HLA-DR Human leukocyte antigen-DR
therapy remains a great challenge; 3) Clinical trials of the HMGA High mobility group protein A
polysaccharide-based nanotheraputics from animal-models to human ICOSL Inducible co-stimulator ligand
beings. Although several studies stated that their polysaccharide-based IFN Interferon
nanomaterials have demonstrated a satisfactory efficacy without IL Interleukin
obvious toxicities, there are still concerns on whether these nano LPH Lipid-protamine-hyaluronic acid
medicines could be administrated safely and effectively in the human LYVE Lymphatic vessel endothelial
body [334,335] since most of these experiments were conducted only in MAN Mannose
animal models while still in the absence of clinical trial data in the MAPK Mitogen-activated protein kinase
human body. A recent study has shown that naturally-derived HA had a MATT Marimastat
wide range of molecular weights, ranging from a size of 104 kDa to be MMP Matrix metalloproteinase
3375
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
NF-κB Nuclear factor kappa B [18] J. Shen, Z. Lu, J. Wang, T. Zhang, J. Yang, Y. Li, G. Liu, X. Zhang, Advances of
nanoparticles for leukemia treatment, ACS Biomater. Sci. Eng. 6 (2020)
NIR Near infrared ray
6478–6489.
NK cell Natural killer cell [19] J. Li, L. Chen, X. Xu, Y. Fan, X. Xue, M. Shen, X. Shi, Targeted combination of
NO Nitric oxide antioxidative and anti-inflammatory therapy of rheumatoid arthritis using
OVA Ovalbumin multifunctional dendrimer-entrapped gold nanoparticles as a platform, Small 16
(2020) 2005661.
PAMP Pathogen-associated molecule pattern [20] M. Gonçalves, S. Mignani, J. Rodrigues, H. Tomás, A glance over doxorubicin
PANI Polyaniline based-nanotherapeutics: from proof-of-concept studies to solutions in the market,
PD-1 Programmed cell death protein 1 J. Contr. Release 317 (2020) 347–374.
[21] R. Zhao, Z. Lu, J. Yang, L. Zhang, Y. Li, X. Zhang, Drug delivery system in the
PD-L1 Programmed death ligand 1 treatment of diabetes mellitus, Front. Bioeng. Biotech. 8 (2020) 880.
PEG Polyethylene glycol [22] Y. Chao, Q. Chen, Z. Liu, Smart injectable hydrogels for cancer immunotherapy,
PEI Polyethylenimine Adv. Funct. Mater. 30 (2019) 1902785.
[23] S. Mignani, X. Shi, A. Steinmetz, J.-P. Majoral, Multivalent copper(II)-conjugated
PIGF Placental growth factor phosphorus dendrimers with noteworthy in vitro and in vivo antitumor activities:
PLA Polylactide a concise overview, Mol. Pharm. 18 (2021) 65–73.
PLGA Poly(lactic-co-glycolic acid) [24] M.S. Goldberg, Immunoengineering: how nanotechnology can enhance cancer
immunotherapy, Cell 161 (2015) 201–204.
PRR Pattern recognition receptor [25] M. Yang, J. Li, P. Gu, X. Fan, The application of nanoparticles in cancer
PTX Paclitaxel immunotherapy: targeting tumor microenvironment, Bioact. Mater. 6 (2021)
RHAMM Receptor for HA-mediated motility 1973–1987.
[26] P. Gong, Y. Wang, P. Zhang, Z. Yang, W. Deng, Z. Sun, M. Yang, X. Li, G. Ma,
ROS Reactive oxygen species
G. Deng, S. Dong, L. Cai, W. Jiang, Immunocyte membrane-coated nanoparticles
Th T helper for cancer immunotherapy, Cancers 13 (2020) 77.
TLR Toll-like receptors [27] T. Miao, J. Wang, Y. Zeng, G. Liu, X. Chen, Polysaccharide-based controlled
TNF Tumor necrosis factor release systems for therapeutics delivery and tissue engineering: from bench to
bedside, Adv. Sci. 5 (2018) 1700513.
TPP Tripolyphosphate [28] Z. Cui, N.C. Ni, J. Wu, G.-Q. Du, S. He, T.M. Yau, R.D. Weisel, H.-W. Sung, R.-
Treg Regulatory T cell K. Li, Polypyrrole-chitosan conductive biomaterial synchronizes cardiomyocyte
VEGF Vascular endothelial growth factor contraction and improves myocardial electrical impulse propagation,
Theranostics 8 (2018) 2752–2764.
VES Vitamin E succinate [29] E. Ghassami, J. Varshosaz, S. Taymouri, Redox sensitive polysaccharide based
nanoparticles for improved cancer treatment: a comprehensive review, Curr.
References Pharmaceut. Des. 24 (2018) 3303–3319.
[30] H. Cai, Y. Xiang, Y. Zeng, Z. Li, X. Zheng, Q. Luo, H. Zhu, Q. Gong, Z. Gu, Y. Liu,
H. Zhang, K. Luo, Cathepsin B-responsive and gadolinium-labeled branched
[1] S. Burugu, A.R. Dancsok, T.O. Nielsen, Emerging targets in cancer
glycopolymer-PTX conjugate-derived nanotheranostics for cancer treatment, Acta
immunotherapy, in: Semin. Cancer Biol., Elsevier, 2018, pp. 39–52.
Pharm. Sin. B 11 (2021) 544–559.
[2] A. Ribas, J.D. Wolchok, Cancer immunotherapy using checkpoint blockade,
[31] Y. Wu, F. Li, X. Zhang, Z. Li, Q. Zhang, W. Wang, D. Pan, X. Zheng, Z. Gu,
Science 359 (2018) 1350–1355.
H. Zhang, Q. Gong, K. Luo, Tumor microenvironment-responsive PEGylated
[3] J.J. Havel, D. Chowell, T.A. Chan, The evolving landscape of biomarkers for
heparin-pyropheophorbide-a nanoconjugates for photodynamic therapy,
checkpoint inhibitor immunotherapy, Nat. Rev. Canc. 19 (2019) 133–150.
Carbohydr. Polym. 255 (2021) 117490.
[4] C.H. June, R.S. O’Connor, O.U. Kawalekar, S. Ghassemi, M.C. Milone, CAR T cell
[32] X. Zhang, Y. Wu, Z. Li, W. Wang, Y. Wu, D. Pan, Z. Gu, R. Sheng, H. Tomás,
immunotherapy for human cancer, Science 359 (2018) 1361–1365.
H. Zhang, J. Rodrigues, Q. Gong, K. Luo, Glycodendron/pyropheophorbide-a
[5] H. Pan, W. Li, Z. Chen, Y. Luo, W. He, M. Wang, X. Tang, H. He, L. Liu, M. Zheng,
(Ppa)-functionalized hyaluronic acid as a nanosystem for tumor photodynamic
X. Jiang, T. Yin, R. Liang, Y. Ma, L. Cai, Click CAR-T cell engineering for robustly
therapy, Carbohydr. Polym. 247 (2020) 116749.
boosting cell immunotherapy in blood and subcutaneous xenograft tumor, Bioact.
[33] Z. Livshits, R.B. Rao, S.W. Smith, An approach to chemotherapy-associated
Mater. 6 (2020) 951–962.
toxicity, Emerg. Med. Clin. 32 (2014) 167–203.
[6] M. Daher, K. Rezvani, Next generation natural killer cells for cancer
[34] V. Narayan, D. Vaughn, Pharmacokinetic and toxicity considerations in the use of
immunotherapy: the promise of genetic engineering, Curr. Opin. Immunol. 51
neoadjuvant chemotherapy for bladder cancer, Expet Opin. Drug Metabol.
(2018) 146–153.
Toxicol. 11 (2015) 731–742.
[7] F. Souza-Fonseca-Guimaraes, J. Cursons, N.D. Huntington, The emergence of
[35] S. Zong, J. Li, Z. Ye, X. Zhang, L. Yang, X. Chen, M. Ye, Lachnum polysaccharide
natural killer cells as a major target in cancer immunotherapy, Trends Immunol.
suppresses S180 sarcoma by boosting anti-tumor immune responses and skewing
40 (2019) 142–158.
tumor-associated macrophages toward M1 phenotype, Int. J. Biol. Macromol. 144
[8] J. Sprooten, P. Agostinis, A.D. Garg, Type I interferons and dendritic cells in
(2020) 1022–1033.
cancer immunotherapy, Int. Rev. Cell Mol. Biol. 348 (2019) 217–262.
[36] Y. Jin, Y. Mu, S. Zhang, P. Li, F. Wang, Preparation and evaluation of the adjuvant
[9] Y.S. Lee, K.J. Radford, The role of dendritic cells in cancer, Int. Rev. Cell Mol.
effect of curdlan sulfate in improving the efficacy of dendritic cell-based vaccine
Biol. 348 (2019) 123–178.
for antitumor immunotherapy, Int. J. Biol. Macromol. 146 (2020) 273–284.
[10] J. Voeller, A.K. Erbe, J. Slowinski, K. Rasmussen, P.M. Carlson, A. Hoefges,
[37] S. Bi, W. Huang, S. Chen, C. Huang, C. Li, Z. Guo, J. Yang, J. Zhu, L. Song, R. Yu,
S. VandenHeuvel, A. Stuckwisch, X. Wang, S.D. Gillies, Combined innate and
Cordyceps militaris polysaccharide converts immunosuppressive macrophages
adaptive immunotherapy overcomes resistance of immunologically cold
into M1-like phenotype and activates T lymphocytes by inhibiting the PD-L1/PD-
syngeneic murine neuroblastoma to checkpoint inhibition, J. Immunother.
1 axis between TAMs and T lymphocytes, Int. J. Biol. Macromol. 150 (2020)
Cancer 7 (2019) 1–13.
261–280.
[11] Q. Chen, C. Wang, X. Zhang, G. Chen, Q. Hu, H. Li, J. Wang, D. Wen, Y. Zhang,
[38] S.S. Ferreira, C.P. Passos, P. Madureira, M. Vilanova, M.A. Coimbra, Structure-
Y. Lu, In situ sprayed bioresponsive immunotherapeutic gel for post-surgical
function relationships of immunostimulatory polysaccharides: a review,
cancer treatment, Nat. Nanotechnol. 14 (2019) 89–97.
Carbohydr. Polym. 132 (2015) 378–396.
[12] T. Zhou, J. Peng, Y. Hao, K. Shi, K. Zhou, Y. Yang, C. Yang, X. He, X. Chen,
[39] P. Li, F. Wang, Polysaccharides: candidates of promising vaccine adjuvants, Drug
Z. Qian, The construction of a lymphoma cell-based, DC-targeted vaccine, and its
Discov. Ther. 9 (2015) 88–93.
application in lymphoma prevention and cure, Bioact. Mater. 6 (2021) 697–711.
[40] F. Chen, G. Huang, Preparation and immunological activity of polysaccharides
[13] L.A. Emens, P.A. Ascierto, P.K. Darcy, S. Demaria, A.M.M. Eggermont, W.
and their derivatives, Int. J. Biol. Macromol. 112 (2018) 211–216.
L. Redmond, B. Seliger, F.M. Marincola, Cancer immunotherapy: opportunities
[41] F. Seidi, R. Jenjob, T. Phakkeeree, D. Crespy, Saccharides, oligosaccharides, and
and challenges in the rapidly evolving clinical landscape, Eur. J. Canc. 81 (2017)
polysaccharides nanoparticles for biomedical applications, J. Contr. Release 284
116–129.
(2018) 188–212.
[14] F. Kroschinsky, F. Stolzel, S. von Bonin, G. Beutel, M. Kochanek, M. Kiehl,
[42] R.S. Riley, C.H. June, R. Langer, M.J. Mitchell, Delivery technologies for cancer
P. Schellongowski, New drugs, new toxicities: severe side effects of modern
immunotherapy, Nat. Rev. Drug Discov. 18 (2019) 175–196.
targeted and immunotherapy of cancer and their management, Crit. Care 21
[43] B. Fu, X. Huang, J. Deng, D. Gu, Q. Mei, M. Deng, S. Tang, M. Lü, Application of
(2017) 89.
multifunctional nanomaterials in cancer vaccines, Oncol. Rep. 39 (2018)
[15] P. Cukier, F.C. Santini, M. Scaranti, A.O. Hoff, Endocrine side effects of cancer
893–900.
immunotherapy, Endocr. Relat. Canc. 24 (2017) T331–T347.
[44] R. Mahjub, S. Jatana, S.E. Lee, Z. Qin, G. Pauli, M. Soleimani, S. Madadi, S.-D. Li,
[16] C.W. Shields IV, L.L.W. Wang, M.A. Evans, S. Mitragotri, Materials for
Recent advances in applying nanotechnologies for cancer immunotherapy,
immunotherapy, Adv. Mater. 32 (2020) 1901633.
J. Contr. Release 288 (2018) 239–263.
[17] N.T. Trac, E.J. Chung, Peptide-based targeting of immunosuppressive cells in
[45] H.Y. Yoon, S.T. Selvan, Y. Yang, M.J. Kim, D.K. Yi, I.C. Kwon, K. Kim, Engineering
cancer, Bioact. Mater 5 (2020) 92–101.
nanoparticle strategies for effective cancer immunotherapy, Biomaterials 178
(2018) 597–607.
3376
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
[46] P.-M. Chen, W.-Y. Pan, C.-Y. Wu, C.-Y. Yeh, C. Korupalli, P.-K. Luo, C.-J. Chou, [69] E.L. Hopewell, C. Cox, S. Pilon-Thomas, L.L. Kelley, Tumor-infiltrating
W.-T. Chia, H.-W. Sung, Modulation of tumor microenvironment using a TLR-7/8 lymphocytes: streamlining a complex manufacturing process, Cytotherapy 21
agonist-loaded nanoparticle system that exerts low-temperature hyperthermia (2019) 307–314.
and immunotherapy for in situ cancer vaccination, Biomaterials 230 (2019) [70] J.G. Egen, W. Ouyang, L.C. Wu, Human anti-tumor immunity: insights from
119629. immunotherapy clinical trials, Immunity 52 (2020) 36–54.
[47] F. Castro, M.L. Pinto, R. Almeida, F. Pereira, A.M. Silva, C.L. Pereira, S.G. Santos, [71] J. Ye, Y. Yang, J. Jin, M. Ji, Y. Gao, Y. Feng, H. Wang, X. Chen, Y. Liu, Targeted
M.A. Barbosa, R.M. Gonçalves, M.J. Oliveira, Chitosan/poly (γ-glutamic acid) delivery of chlorogenic acid by mannosylated liposomes to effectively promote
nanoparticles incorporating IFN-γ for immune response modulation in the context the polarization of TAMs for the treatment of glioblastoma, Bioact. Mater. 5
of colorectal cancer, Biomater. Sci. 7 (2019) 3386–3403. (2020) 694–708.
[48] Y. Song, C. Tang, C. Yin, Combination antitumor immunotherapy with VEGF and [72] P.S. Hegde, D.S. Chen, Top 10 challenges in cancer immunotherapy, Immunity 52
PIGF siRNA via systemic delivery of multi-functionalized nanoparticles to tumor- (2020) 17–35.
associated macrophages and breast cancer cells, Biomaterials 185 (2018) [73] F. Marofi, R. Motavalli, V.A. Safonov, L. Thangavelu, A.V. Yumashev,
117–132. M. Alexander, N. Shomali, M.S. Chartrand, Y. Pathak, M. Jarahian, S. Izadi,
[49] X. Yang, K. Lian, T. Meng, X. Liu, J. Miao, Y. Tan, H. Yuan, F. Hu, Immune A. Hassanzadeh, N. Shirafkan, S. Tahmasebi, F.M. Khiavi, CAR T cells in solid
adjuvant targeting micelles allow efficient dendritic cell migration to lymph tumors: challenges and opportunities, Stem Cell Res. Ther. 12 (2021) 81.
nodes for enhanced cellular immunity, ACS Appl. Mater. Interfaces 10 (2018) [74] S.F. Slovin, Chapter 56 - sipuleucel-T – a model for immunotherapy trial
33532–33544. development, in: J.H. Mydlo, C.J. Godec (Eds.), Prostate Cancer, second ed.,
[50] C.-T. Tsao, F.M. Kievit, A. Ravanpay, A.E. Erickson, M.C. Jensen, R.G. Ellenbogen, Academic Press, San Diego, 2016, pp. 523–531.
M. Zhang, Thermoreversible poly (ethylene glycol)-g-chitosan hydrogel as a [75] G.A. Leget, M.S. Czuczman, Use of rituximab, the new FDA-approved antibody,
therapeutic T lymphocyte depot for localized glioblastoma immunotherapy, Curr. Opin. Oncol. 10 (1998) 548–551.
Biomacromolecules 15 (2014) 2656–2662. [76] C. Nabhan, The emerging role of alemtuzumab in chronic lymphocytic leukemia,
[51] F. Cao, M. Yan, Y. Liu, L. Liu, G. Ma, Photothermally controlled MHC class I Clin. Lymphoma & Myeloma 6 (2005) 115–121.
restricted CD8+ T-cell responses elicited by hyaluronic acid decorated gold [77] A. Osterborg, Ofatumumab, a human anti-CD20 monoclonal antibody, Expet
nanoparticles as a vaccine for cancer immunotherapy, Adv. Healthc. Mater. 7 Opin. Biol. Ther. 10 (2010) 439–449.
(2018) 1701439. [78] A. Markham, Elotuzumab: first global approval, Drugs 76 (2016) 397–403.
[52] Y. Lv, C. Xu, X. Zhao, C. Lin, X. Yang, X. Xin, L. Zhang, C. Qin, X. Han, L. Yang, [79] F. Cameron, G. Whiteside, C.M. Perry, Ipilimumab: first global approval, Drugs 71
Nanoplatform assembled from a CD44-targeted prodrug and smart liposomes for (2011) 1093–1104.
dual targeting of tumor microenvironment and cancer cells, ACS Nano 12 (2018) [80] M.A. Postow, M.K. Callahan, J.D. Wolchok, Immune checkpoint blockade in
1519–1536. cancer therapy, J. Clin. Oncol. 33 (2015) 1974–1982.
[53] J.W. Hickey, Y. Dong, J.W. Chung, S.F. Salathe, H.C. Pruitt, X. Li, C. Chang, A. [81] L.A. Raedler, Opdivo (Nivolumab): second PD-1 inhibitor receives FDA approval
K. Fraser, C.A. Bessell, A.J. Ewald, Engineering an artificial T-cell stimulating for unresectable or metastatic melanoma, Am. Health Drug Benef. 8 (2015)
matrix for immunotherapy, Adv. Mater. 31 (2019) 1807359. 180–183.
[54] C. Wang, Y. Ye, G.M. Hochu, H. Sadeghifar, Z. Gu, Enhanced cancer [82] R.M. Poole, Pembrolizumab: first global approval, Drugs 74 (2014) 1973–1981.
immunotherapy by microneedle patch-assisted delivery of anti-PD1 antibody, [83] A. Markham, Atezolizumab: first global approval, Drugs 76 (2016) 1227–1232.
Nano Lett. 16 (2016) 2334–2340. [84] E.S. Kim, Avelumab: first global approval, Drugs 77 (2017) 929–937.
[55] T. Bauleth-Ramos, M.A. Shahbazi, D. Liu, F. Fontana, A. Correia, P. Figueiredo, [85] Y.Y. Syed, Durvalumab: first global approval, Drugs 77 (2017) 1369–1376.
H. Zhang, J.P. Martins, J.T. Hirvonen, P. Granja, Nutlin-3a and cytokine Co- [86] A. Markham, S.T. Duggan, Cemiplimab: first global approval, Drugs 78 (2018)
loaded spermine-modified acetalated dextran nanoparticles for cancer chemo- 1841–1846.
immunotherapy, Adv. Funct. Mater. 27 (2017) 1703303. [87] M. von Locquenghien, C. Rozalén, T. Celià-Terrassa, Interferons in cancer
[56] Y. Wang, Z. Luan, C. Zhao, C. Bai, K. Yang, Target delivery selective CSF-1R immunoediting: sculpting metastasis and immunotherapy response, J. Clin.
inhibitor to tumor-associated macrophages via erythrocyte-cancer cell hybrid Invest. 131 (2021), e143296.
membrane camouflaged pH-responsive copolymer micelle for cancer [88] A. Blaauboer, K. Sideras, C.H.J. van Eijck, L.J. Hofland, Type I interferons in
immunotherapy, Eur. J. Pharmaceut. Sci. 142 (2020) 105136. pancreatic cancer and development of new therapeutic approaches, Crit. Rev.
[57] F. Fontana, M.A. Shahbazi, D. Liu, H. Zhang, E. Mäkilä, J. Salonen, J.T. Hirvonen, Oncol. Hematol. 159 (2020) 103204.
H.A. Santos, Multistaged nanovaccines based on porous silicon@ acetalated [89] Y. Yang, A. Lundqvist, Immunomodulatory effects of IL-2 and IL-15; Implications
dextran@ cancer cell membrane for cancer immunotherapy, Adv. Mater. 29 for cancer immunotherapy, Cancers 12 (2020) 3586.
(2017) 1603239. [90] K.G. Nguyen, M.R. Vrabel, S.M. Mantooth, J.J. Hopkins, E.S. Wagner, T.
[58] C. Zhang, G. Shi, J. Zhang, H. Song, J. Niu, S. Shi, P. Huang, Y. Wang, W. Wang, A. Gabaldon, D.A. Zaharoff, Localized interleukin-12 for cancer immunotherapy,
C. Li, Targeted antigen delivery to dendritic cell via functionalized alginate Front. Immunol. 11 (2020) 575597.
nanoparticles for cancer immunotherapy, J. Contr. Release 256 (2017) 170–181. [91] R. Weber, C. Groth, S. Lasser, I. Arkhypov, V. Petrova, P. Altevogt, J. Utikal,
[59] M. Liu, A.R. Khan, J. Ji, G. Lin, X. Zhao, G. Zhai, Crosslinked self-assembled V. Umansky, IL-6 as a major regulator of MDSC activity and possible target for
nanoparticles for chemo-sonodynamic combination therapy favoring antitumor, cancer immunotherapy, Cell. Immunol. 359 (2021) 104254.
antimetastasis management and immune responses, J. Contr. Release 290 (2018) [92] J. Summers, M.H. Cohen, P. Keegan, R. Pazdur, FDA drug approval summary:
150–164. bevacizumab plus interferon for advanced renal cell carcinoma, Oncol. 15 (2010)
[60] C.B. Rodell, S.P. Arlauckas, M.F. Cuccarese, C.S. Garris, R. Li, M.S. Ahmed, R. 104–111.
H. Kohler, M.J. Pittet, R. Weissleder, TLR7/8-agonist-loaded nanoparticles [93] G.C. Sim, L. Radvanyi, The IL-2 cytokine family in cancer immunotherapy,
promote the polarization of tumour-associated macrophages to enhance cancer Cytokine Growth Factor Rev. 25 (2014) 377–390.
immunotherapy, Nat. Biomed. Eng. 2 (2018) 578–588. [94] D. Jorgovanovic, M. Song, L. Wang, Y. Zhang, Roles of IFN-γ in tumor progression
[61] F. Castro, M.L. Pinto, A.M. Silva, C.L. Pereira, S.G. Santos, M.A. Barbosa, O. and regression: a review, Biomark. Res. 8 (2020) 49.
D. Wever, K. Serre, R.M. Gonçalves, M.J. Oliveira, PO-420 Harnessing chitosan/ [95] R. Upadhyay, J.A. Boiarsky, G. Pantsulaia, J. Svensson-Arvelund, M.J. Lin,
Poly(Y)-glutamic acid nanoparticles for immunomodulation at the tumour A. Wroblewska, S. Bhalla, N. Scholler, A. Bot, J.M. Rossi, N. Sadek, S. Parekh,
microenvironment, ESMO Open 3 (2018) A395. A. Laganà, A. Baccarini, M. Merad, B.D. Brown, J.D. Brody, A critical role for fas-
[62] A. Dey, S. Manna, S. Kumar, S. Chattopadhyay, B. Saha, S. Roy, mediated off-target tumor killing in T cell immunotherapy, Cancer Discov 11
Immunostimulatory effect of chitosan conjugated green copper oxide (2021) 599–613.
nanoparticles in tumor immunotherapy, Cytokine 127 (2020) 154958. [96] A.E. Andrea, A. Chiron, S. Bessoles, S. Hacein-Bey-Abina, Engineering next-
[63] D. Bamberger, D. Hobernik, M. Konhäuser, M. Bros, P.R. Wich, Surface generation CAR-T cells for better toxicity management, Int. J. Mol. Sci. 21 (2020)
modification of polysaccharide-based nanoparticles with PEG and dextran and 8620.
the effects on immune cell binding and stimulatory characteristics, Mol. Pharm. [97] W. Li, A. Peng, H. Wu, Y. Quan, Y. Li, L. Lu, M. Cui, Anti-cancer nanomedicines: a
14 (2017) 4403–4416. revolution of tumor immunotherapy, Front. Immunol. 11 (2020) 601497.
[64] S. Zhang, G. Pang, C. Chen, J. Qin, H. Yu, Y. Liu, X. Zhang, Z. Song, J. Zhao, [98] Y. Yu, M. Shen, Q. Song, J. Xie, Biological activities and pharmaceutical
F. Wang, Effective cancer immunotherapy by Ganoderma lucidum applications of polysaccharide from natural resources: a review, Carbohydr.
polysaccharide-gold nanocomposites through dendritic cell activation and Polym. 183 (2018) 91–101.
memory T cell response, Carbohydr. Polym. 205 (2019) 192–202. [99] A. Behera, D. Rai, S.S. Kulkarni, Total syntheses of conjugation-ready
[65] H. Yu, Y. Yang, T. Jiang, X. Zhang, Y. Zhao, G. Pang, Y. Feng, S. Zhang, F. Wang, trisaccharide repeating units of Pseudomonas aeruginosa O11 and
Y. Wang, Effective radiotherapy in tumor assisted by Ganoderma lucidum Staphylococcus aureus Type 5 capsular polysaccharide for vaccine development,
polysaccharide-conjugated bismuth sulfide nanoparticles through J. Am. Chem. Soc. 142 (2020) 456–467.
radiosensitization and dendritic cell activation, ACS Appl. Mater. Interfaces 11 [100] L. Liu, S. Nie, M. Xie, Tumor microenvironment as a new target for tumor
(2019) 27536–27547. immunotherapy of polysaccharides, Crit. Rev. Food Sci. Nutr. 56 (Suppl 1) (2016)
[66] T. Guo, Y. Yang, M. Gao, Y. Qu, X. Guo, Y. Liu, X. Cui, C. Wang, Lepidium meyenii S85–S94.
Walpers polysaccharide and its cationic derivative re-educate tumor-associated [101] I.A. Schepetkin, M.T. Quinn, Botanical polysaccharides: macrophage
macrophages for synergistic tumor immunotherapy, Carbohydr. Polym. 250 immunomodulation and therapeutic potential, Int. Immunopharm. 6 (2006)
(2020) 116904. 317–333.
[67] J. Galon, D. Bruni, Tumor immunology and tumor evolution: intertwined [102] X. Liu, J. Xie, S. Jia, L. Huang, Z. Wang, C. Li, M. Xie, Immunomodulatory effects
histories, Immunity 52 (2020) 55–81. of an acetylated Cyclocarya paliurus polysaccharide on murine macrophages
[68] F.S. Varn, D.W. Mullins, H. Arias-Pulido, S. Fiering, C. Cheng, Adaptive immunity RAW264.7, Int. J. Biol. Macromol. 98 (2017) 576–581.
programmes in breast cancer, Immunology 150 (2017) 25–34.
3377
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
[103] M. Zhang, W. Wu, Y. Ren, X. Li, Y. Tang, T. Min, F. Lai, H. Wu, Structural [130] L.M. Kranz, M. Diken, H. Haas, S. Kreiter, C. Loquai, K.C. Reuter, M. Meng,
characterization of a novel polysaccharide from Lepidium meyenii (Maca) and D. Fritz, F. Vascotto, H. Hefesha, C. Grunwitz, M. Vormehr, Y. Hüsemann,
analysis of its regulatory function in macrophage polarization in vitro, J. Agric. A. Selmi, A.N. Kuhn, J. Buck, E. Derhovanessian, R. Rae, S. Attig, J. Diekmann, R.
Food Chem. 65 (2017) 1146–1157. A. Jabulowsky, S. Heesch, J. Hassel, P. Langguth, S. Grabbe, C. Huber, Ö. Türeci,
[104] F. Su, M. Sun, Y. Geng, (1)H-NMR Metabolomics Analysis of the effects of sulfated U. Sahin, Systemic RNA delivery to dendritic cells exploits antiviral defence for
polysaccharides from masson pine pollen in RAW264.7 macrophage cells, cancer immunotherapy, Nature 534 (2016) 396–401.
Molecules 24 (2019) 1841. [131] M. Björnmalm, K.J. Thurecht, M. Michael, A.M. Scott, F. Caruso, Bridging
[105] L. Tong, L. Wang, X. Zhou, K. Zhong, L. Liu, F. Wang, S. Zhou, Antitumor activity bio–nano science and cancer nanomedicine, ACS Nano 11 (2017) 9594–9613.
of Dendrobium devonianum polysaccharides based on their immunomodulatory [132] R.S. Riley, C.H. June, R. Langer, M.J. Mitchell, Delivery technologies for cancer
effects in S180 tumor-bearing mice, RSC Adv. 6 (2016) 40250–40257. immunotherapy, Nat. Rev. Drug Discov. 18 (2019) 175–196.
[106] W. Xu, R. Guan, F. Shi, A. Du, S. Hu, Structural analysis and immunomodulatory [133] J. Conniot, J.M. Silva, J.G. Fernandes, L.C. Silva, R. Gaspar, S. Brocchini, H.
effect of polysaccharide from Atractylodis macrocephalae Koidz. on bovine F. Florindo, T.S. Barata, Cancer immunotherapy: nanodelivery approaches for
lymphocytes, Carbohydr. Polym. 174 (2017) 1213–1223. immune cell targeting and tracking, Front. Chem. 2 (2014) 105. -105.
[107] Z.Y. Zhu, J.Y. Zhang, F. Liu, L. Chen, L.J. Chen, Y. Tang, Characterization and [134] R.B. Rigon, M.H. Oyafuso, A.T. Fujimura, M.L. Gonçalez, A.H. do Prado, M.P.
lymphocyte proliferation activity of an oligosaccharide degraded from Astragalus D. Gremião, M. Chorilli, Nanotechnology-based drug delivery systems for
polysaccharide, MedChemComm 8 (2017) 1521–1530. melanoma antitumoral therapy: a review, BioMed Res. Int. 2015 (2015), 841817.
[108] L. Xu, W. Zhang, L. Zeng, J.-O. Jin, Rehmannia glutinosa polysaccharide induced [135] L. Liu, S. Nie, M. Xie, Tumor microenvironment as a new target for tumor
an anti-cancer effect by activating natural killer cells, Int. J. Biol. Macromol. 105 immunotherapy of polysaccharides, Crit. Rev. Food Sci. Nutr. 56 (2016) S85–S94.
(2017) 680–685. [136] H.B.T. Moran, J.L. Turley, M. Andersson, E.C. Lavelle, Immunomodulatory
[109] S. Kikete, L. Luo, B. Jia, L. Wang, G. Ondieki, Y. Bian, Plant-derived properties of chitosan polymers, Biomaterials 184 (2018) 1–9.
polysaccharides activate dendritic cell-based anti-cancer immunity, [137] F. Zamboni, S. Vieira, R.L. Reis, J.M. Oliveira, M.N. Collins, The potential of
Cytotechnology 70 (2018) 1097–1110. hyaluronic acid in immunoprotection and immunomodulation: chemistry,
[110] G. Pang, S. Zhang, X. Zhou, H. Yu, Y. Wu, T. Jiang, X. Zhang, F. Wang, Y. Wang, L. processing and function, Prog. Mater. Sci. 97 (2018) 97–122.
W. Zhang, Immunoactive polysaccharide functionalized gold nanocomposites [138] E. Yudiati, A. Isnansetyo, Murwantoko, Ayuningtyas, Triyanto, C.R. Handayani,
promote dendritic cell stimulation and antitumor effects, Nanomedicine 14 Innate immune-stimulating and immune genes up-regulating activities of three
(2019) 1291–1306. types of alginate from Sargassum siliquosum in Pacific white shrimp, Litopenaeus
[111] J. Li, A. Aipire, H. Zhao, P. Yuan, J. Li, Pleurotus ferulae polysaccharides improve vannamei, Fish Shellfish Immunol. 54 (2016) 46–53.
the antitumor efficacy of therapeutic human papillomavirus dendritic cell-based [139] S.K. Shukla, A.K. Mishra, O.A. Arotiba, B.B. Mamba, Chitosan-based
vaccine, Hum. Vaccines Immunother. 15 (2019) 611–619. nanomaterials: a state-of-the-art review, Int. J. Biol. Macromol. 59 (2013) 46–58.
[112] J. Du, J. Li, J. Zhu, C. Huang, S. Bi, L. Song, X. Hu, R. Yu, Structural [140] W. Song, S. Musetti, L. Huang, Nanomaterials for cancer immunotherapy,
characterization and immunomodulatory activity of a novel polysaccharide from Biomaterials 148 (2017) 16–30.
Ficus carica, Food Funct. 9 (2018) 3930–3943. [141] F. Dosio, S. Arpicco, B. Stella, E. Fattal, Hyaluronic acid for anticancer drug and
[113] X. Sun, G. Stefanetti, F. Berti, D.L. Kasper, Polysaccharide structure dictates nucleic acid delivery, Adv. Drug Deliv. Rev. 97 (2016) 204–236.
mechanism of adaptive immune response to glycoconjugate vaccines, Proc. Natl. [142] T.G. Dacoba, A. Olivera, D. Torres, J. Crecentecampo, M.J. Alonso, Modulating
Acad. Sci. U.S.A. 116 (2019) 193–198. the immune system through nanotechnology, Semin. Immunol. 34 (2017)
[114] H.T. Wang, L.C. Yang, H.C. Yu, M.L. Chen, H.J. Wang, T.J. Lu, Characteristics of 78–102.
fucose-containing polysaccharides from submerged fermentation of Agaricus [143] R. Jayakumar, D. Menon, K. Manzoor, S. Nair, H. Tamura, Biomedical
blazei Murill, J. Food Drug Anal. 26 (2018) 678–687. applications of chitin and chitosan based nanomaterials—a short review,
[115] F.V. Loures, E.F. Araújo, C. Feriotti, S.B. Bazan, V.L.G. Calich, TLR-4 cooperates Carbohydr. Polym. 82 (2010) 227–232.
with Dectin-1 and mannose receptor to expand Th17 and Tc17 cells induced by [144] C.K. Saurabh, A. Adnan, M.N. Fazita, M. Syakir, Y. Davoudpour, M. Rafatullah,
Paracoccidioides brasiliensis stimulated dendritic cells, Front. Microbiol. 6 (2015) C. Abdullah, M. Haafiz, R. Dungani, A review on chitosan-cellulose blends and
261. -261. nanocellulose reinforced chitosan biocomposites: properties and their
[116] S. Zhang, S. Nie, D. Huang, J. Huang, Y. Wang, M. Xie, Polysaccharide from applications, Carbohydr. Polym. 150 (2016) 216–226.
Ganoderma atrum evokes antitumor activity via Toll-like receptor 4-mediated NF- [145] M. Agarwal, M.K. Agarwal, N. Shrivastav, S. Pandey, R. Das, P. Gaur, Preparation
κB and mitogen-activated protein kinase signaling pathways, J. Agric. Food of chitosan nanoparticles and their in-vitro characterization, Int. J. Life Sci. Sci.
Chem. 61 (2013) 3676–3682. Res. 4 (2018) 1713–1720.
[117] X. Jia, Y. Liang, C. Zhang, K. Wang, Y. Tu, M. Chen, P. Li, J.B. Wan, C. He, [146] M.H. Periayah, A.S. Halim, A.Z.M. Saad, Chitosan: a promising marine
Polysaccharide PRM3 from Rhynchosia minima root enhances immune function polysaccharide for biomedical research, Phcog. Rev. 10 (2016) 39–42.
through TLR4-NF-κB pathway, Biochim. Biophys. Acta Gen. Subj. 1862 (2018) [147] W. Khan, E. Abtew, S. Modani, A.J. Domb, Polysaccharide based nanoparticles,
1751–1759. Isr. J. Chem. 58 (2018) 1315–1329.
[118] R. Zhang, Q. Yu, G. Shi, R. Liu, W. Zhang, X. Zhao, G. Li, M. Ge, chTL R4 pathway [148] D. Sahoo, S. Sahoo, P. Mohanty, S. Sasmal, P.L. Nayak, Chitosan: a new versatile
activation by Astragalus polysaccharide in bursa of Fabricius, BMC Vet. Res. 13 bio-polymer for various applications, Des. Monomers Polym. 12 (2009) 377–404.
(2017) 119. [149] P.S. Esbah Tabaei, M. Asadian, R. Ghobeira, P. Cools, M. Thukkaram, P.
[119] L. Yang, L. Zhang, Chemical structural and chain conformational characterization G. Derakhshandeh, S. Abednatanzi, P. Van Der Voort, K. Verbeken, C. Vercruysse,
of some bioactive polysaccharides isolated from natural sources, Carbohydr. H. Declercq, R. Morent, N. De Geyter, Combinatorial effects of coral addition and
Polym. 76 (2009) 349–361. plasma treatment on the properties of chitosan/polyethylene oxide nanofibers
[120] A. Mueller, J. Raptis, P.J. Rice, J. Kalbfleisch, R.D. Stout, H.E. Ensley, intended for bone tissue engineering, Carbohydr. Polym. 253 (2021) 117211.
W. Browder, D.L. Williams, The influence of glucan polymer structure and [150] H. Du, M. Liu, X. Yang, G. Zhai, The design of pH-sensitive chitosan-based
solution conformation on binding to (1→3)-β-d-glucan receptors in a human formulations for gastrointestinal delivery, Drug Discov. Today 20 (2015)
monocyte-like cell line, Glycobiology 10 (2000) 339–346. 1004–1011.
[121] S. Li, Q. Xiong, X. Lai, X. Li, M. Wan, J. Zhang, Y. Yan, M. Cao, L. Lu, J. Guan, [151] H. Du, M. Liu, X. Yang, G. Zhai, The role of glycyrrhetinic acid modification on
Molecular modification of polysaccharides and resulting bioactivities, Compr. preparation and evaluation of quercetin-loaded chitosan-based self-aggregates,
Rev. Food Sci. Food Saf. 15 (2016) 237–250. J. Colloid Interface Sci. 460 (2015) 87–96.
[122] G. Huang, H. Huang, The derivatization and antitumor mechanisms of [152] M.A. Elgadir, M.S. Uddin, S. Ferdosh, A. Adam, A.J.K. Chowdhury, M.Z.I. Sarker,
polysaccharides, Future Med. Chem. 9 (2017) 1931–1938. Impact of chitosan composites and chitosan nanoparticle composites on various
[123] J. Jiang, F.-Y. Meng, Z. He, Y.-L. Ning, X.-H. Li, H. Song, J. Wang, R. Zhou, drug delivery systems: a review, J. Food Drug Anal. 23 (2015) 619–629.
Sulfated modification of longan polysaccharide and its immunomodulatory and [153] K. Divya, M. Jisha, Chitosan nanoparticles preparation and applications, Environ.
antitumor activity in vitro, Int. J. Biol. Macromol. 67 (2014) 323–329. Chem. Lett. 16 (2018) 101–112.
[124] D. Wang, Y. Hu, Y. Fan, J. Wang, S. Abula, L. Guo, J. Zhang, S.K. Khakame, B. [154] J. He, X. Hu, J. Cao, Y. Zhang, J. Xiao, l. Peng, D. Chen, C. Xiong, L. Zhang,
K. Dang, Immuno-enhancing activity of sulfated Auricularia auricula Chitosan-coated hydroxyapatite and drug-loaded polytrimethylene carbonate/
polysaccharides, Carbohydr. Polym. 89 (2012) 1117–1122. polylactic acid scaffold for enhancing bone regeneration, Carbohydr. Polym. 253
[125] G.M. Jose, G.M. Kurup, The efficacy of sulfated polysaccharides from Padina (2021) 117198.
tetrastromatica in modulating the immune functions of RAW 264.7 cells, Biomed. [155] J.d.S. Apostólico, V.A.S. Lunardelli, F.C. Coirada, S.B. Boscardin, D.S. Rosa,
Pharma. 88 (2017) 677–683. Adjuvants: classification, modus operandi, and licensing, J. Immunol. Res. 2016
[126] L. Lin, J. Yang, Y. Yang, H. Zhi, X. Hu, D. Chai, Y. Liu, X. Shen, J. Wang, Y. Song, (2016) 1459394.
Phosphorylation of Radix Cyathula officinalis polysaccharide improves its [156] R.L. Sabado, S. Balan, N. Bhardwaj, Dendritic cell-based immunotherapy, Cell
immune-enhancing activity, J. Carbohydr. Chem. 39 (2020) 50–62. Res. 27 (2017) 74–95.
[127] T. Qin, J. Chen, D. Wang, Y. Hu, J. Zhang, M. Wang, S. Qiu, Z. Gao, R. Liu, Y. Yu, [157] P.M. Santos, L.H. Butterfield, Dendritic cell-based cancer vaccines, J. Immunol.
Selenylation modification can enhance immune-enhancing activity of Chinese 200 (2018) 443–449.
angelica polysaccharide, Carbohydr. Polym. 95 (2013) 183–187. [158] N. Petrovsky, Comparative safety of vaccine adjuvants: a summary of current
[128] L.M. Jiang, S.P. Nie, H.L. Zhou, D.F. Huang, M.Y. Xie, Carboxymethylation evidence and future needs, Drug Saf. 38 (2015) 1059–1074.
enhances the maturation-inducing activity in dendritic cells of polysaccharide [159] W.S. Bowen, A.K. Svrivastava, L. Batra, H. Barsoumian, H. Shirwan, Current
from the seeds of Plantago asiatica L, Int. Immunopharm. 22 (2014) 324–331. challenges for cancer vaccine adjuvant development, Expert Rev. Vaccines 17
[129] Y. Chen, H. Zhang, Y. Wang, S. Nie, C. Li, M. Xie, Acetylation and (2018) 207–215.
carboxymethylation of the polysaccharide from Ganoderma atrum and their
antioxidant and immunomodulating activities, Food Chem. 156 (2014) 279–288.
3378
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
[160] M.A. Shetab Boushehri, A. Lamprecht, TLR4-based immunotherapeutics in [186] F. Pastor, P. Berraondo, I. Etxeberria, J. Frederick, U. Sahin, E. Gilboa, I. Melero,
cancer: a review of the achievements and shortcomings, Mol. Pharm. 15 (2018) An RNA toolbox for cancer immunotherapy, Nat. Rev. Drug Discov. 17 (2018)
4777–4800. 751–767.
[161] K.S. Michelsen, A. Aicher, M. Mohaupt, T. Hartung, S. Dimmeler, C.J. Kirschning, [187] C. Chin, E.S. Lunking, M. de la Fuente, N.G. Ayad, Immunotherapy and epigenetic
R.R. Schumann, The role of Toll-like receptors (TLRs) in bacteria-induced pathway modulation in glioblastoma multiforme, Front. Oncol. 8 (2018) 521.
maturation of murine dendritic cells-PGN and LTA are inducers of DC maturation [188] S. Daneshmandi, S. Shahrokhi, Engineering tumor cells with tumor necrosis factor
and require TLR2, J. Biol. Chem. 276 (2001) 25680–25686. α (TNF-α) or CD40 ligand (CD40L) genes induce anti-tumor immune responses,
[162] T. Adamus, M. Kortylewski, The revival of CpG oligonucleotide-based cancer Int. J. Pept. Res. Therapeut. 25 (2019) 427–436.
immunotherapies, Contemp. Oncol. 22 (2018) 56–60. [189] J.Y. Yhee, H. Koo, D.E. Lee, K. Choi, I.C. Kwon, K. Kim, Multifunctional chitosan
[163] C.Y. Lai, G.Y. Yu, Y. Luo, R. Xiang, T.H. Chuang, Immunostimulatory activities of nanoparticles for tumor imaging and therapy. Chitosan for Biomaterials I,
CpG-oligodeoxynucleotides in teleosts: toll-like receptors 9 and 21, Front. Springer, 2011, pp. 139–161.
Immunol. 10 (2019) 179. [190] S. Daneshmandi, A.A. Pourfathollah, M. Forouzandeh-Moghaddam, Enhanced
[164] S. Im, J. Lee, D. Park, A. Park, Y.-M. Kim, W.J. Kim, Hypoxia-triggered CD40 and ICOSL expression on dendritic cells surface improve anti-tumor
transforming immunomodulator for cancer immunotherapy via immune responses; effectiveness of mRNA/chitosan nanoparticles,
photodynamically enhanced antigen presentation of dendritic cell, ACS Nano 13 Immunopharmacol. Immunotoxicol. 40 (2018) 375–386.
(2018) 476–488. [191] A. Gardner, B. Ruffell, Dendritic cells and cancer immunity, Trends Immunol. 37
[165] N. Hanagata, CpG oligodeoxynucleotide nanomedicines for the prophylaxis or (2016) 855–865.
treatment of cancers, infectious diseases, and allergies, Int. J. Nanomed. 12 [192] C. Fu, H. Zhao, Y. Wang, H. Cai, Y. Xiao, Y. Zeng, H. Chen, Tumor-associated
(2017) 515–531. antigens: tn antigen, sTn antigen, and T antigen, Hla 88 (2016) 275–286.
[166] H. Zhang, X.D. Gao, Nanodelivery systems for enhancing the immunostimulatory [193] C.C. Liu, H. Yang, R. Zhang, J.J. Zhao, D.J. Hao, Tumour-associated antigens and
effect of CpG oligodeoxynucleotides, Mater. Sci. Eng. C-Mater. Biol. Appl. 70 their anti-cancer applications, Eur. J. Canc. Care 26 (2017), e12446.
(2017) 935–946. [194] S.R. Woo, L. Corrales, T.F. Gajewski, Innate immune recognition of cancer, Annu.
[167] H.M. Xu, Th1 cytokine-based immunotherapy for cancer, Hepatobiliary Pancreat. Rev. Immunol. 33 (2015) 445–474.
Dis. Int. 13 (2014) 482–494. [195] T.N. Schumacher, R.D. Schreiber, Neoantigens in cancer immunotherapy, Science
[168] P. Berraondo, M.F. Sanmamed, M.C. Ochoa, I. Etxeberria, M.A. Aznar, J.L. Perez- 348 (2015) 69–74.
Gracia, M.E. Rodriguez-Ruiz, M. Ponz-Sarvise, E. Castanon, I. Melero, Cytokines [196] J.L. Gulley, R.A. Madan, R. Pachynski, P. Mulders, N.A. Sheikh, J. Trager, C.
in clinical cancer immunotherapy, Br. J. Canc. 120 (2019) 6–15. G. Drake, Role of antigen spread and distinctive characteristics of immunotherapy
[169] K. Sonaje, Y.-J. Chen, H.-L. Chen, S.-P. Wey, J.-H. Juang, H.-N. Nguyen, C.- in cancer treatment, J. Natl. Cancer Inst. 109 (2017) djw261.
W. Hsu, K.-J. Lin, H.-W. Sung, Enteric-coated capsules filled with freeze-dried [197] J. Windberg, R. Zhang, MAGE-3 peptide amphiphile micelle vaccine promote
chitosan/poly (γ-glutamic acid) nanoparticles for oral insulin delivery, anti-tumor immunity in mice with stomach cancer, bioRxiv (2019) 609214.
Biomaterials 31 (2010) 3384–3394. [198] A. Hanefeld, M. Weigandt, M. Wolf, P. Knolle, M. Schroeder, R. Scherliess,
[170] D.-W. Tang, S.-H. Yu, Y.-C. Ho, F.-L. Mi, P.-L. Kuo, H.-W. Sung, Heparinized P. Walden, A. Diedrich, H. Steckel, R.B. Baleeiro, Antigen-loaded chitosan
chitosan/poly (γ-glutamic acid) nanoparticles for multi-functional delivery of nanoparticles for immunotherapy, in: Google Patents, 2017.
fibroblast growth factor and heparin, Biomaterials 31 (2010) 9320–9332. [199] H. Rajaei, M. Jahromi, M. Ali, N. Khoramabadi, Z. Mohammad Hassan,
[171] Z. Keresztessy, M. Bodnár, E. Ber, I. Hajdu, M. Zhang, J.F. Hartmann, T. Minko, Immunoregulatory properties of arteether in folic acid-chitosan-Fe3O4 composite
J. Borbély, Self-assembling chitosan/poly-γ-glutamic acid nanoparticles for nanoparticle in 4T1 cell line and mice bearing breast cancer, Immunoregulation 1
targeted drug delivery, Colloid Polym. Sci. 287 (2009) 759–765. (2019) 207–220.
[172] F. Castro, M.L. Pinto, A.M. Silva, C.L. Pereira, G.Q. Teixeira, M. Gomez-Lazaro, S. [200] S. Bhattacharyya, D. Md Sakib Hossain, S. Mohanty, G. Sankar Sen,
G. Santos, M.A. Barbosa, R.M. Goncalves, M.J. Oliveira, Pro-inflammatory S. Chattopadhyay, S. Banerjee, J. Chakraborty, K. Das, D. Sarkar, T. Das, G. Sa,
chitosan/poly (γ-glutamic acid) nanoparticles modulate human antigen- Curcumin reverses T cell-mediated adaptive immune dysfunctions in tumor-
presenting cells phenotype and revert their pro-invasive capacity, Acta Biomater. bearing hosts, Cell. Mol. Immunol. 7 (2010) 306–315.
63 (2017) 96–109. [201] J. Odot, P. Albert, A. Carlier, M. Tarpin, J. Devy, C. Madoulet, In vitro and in vivo
[173] P. Berraondo, M.F. Sanmamed, M.C. Ochoa, I. Etxeberria, M.A. Aznar, J.L. Pérez- anti-tumoral effect of curcumin against melanoma cells, Int. J. Canc. 111 (2004)
Gracia, M.E. Rodríguez-Ruiz, M. Ponz-Sarvise, E. Castañón, I. Melero, Cytokines 381–387.
in clinical cancer immunotherapy, Br. J. Canc. 120 (2019) 6–15. [202] F. Milano, L. Mari, W. van de Luijtgaarden, K. Parikh, S. Calpe, K. Krishnadath,
[174] M. Tariq, J. Zhang, G. Liang, L. Ding, Q. He, B. Yang, Macrophage polarization: Nano-curcumin inhibits proliferation of esophageal adenocarcinoma cells and
anti-cancer strategies to target tumor-associated macrophage in breast cancer, enhances the T cell mediated immune response, Front. Oncol. 3 (2013) 137.
J. Cell. Biochem. 118 (2017) 2484–2501. [203] R. Shafabakhsh, M.H. Pourhanifeh, H.R. Mirzaei, A. Sahebkar, Z. Asemi,
[175] M. Najafi, N. Hashemi Goradel, B. Farhood, E. Salehi, M.S. Nashtaei, H. Mirzaei, Targeting regulatory T cells by curcumin: a potential for cancer
N. Khanlarkhani, Z. Khezri, J. Majidpoor, M. Abouzaripour, M. Habibi, I. immunotherapy, Pharmacol. Res. 147 (2019) 104353.
R. Kashani, K. Mortezaee, Macrophage polarity in cancer: a review, J. Cell. [204] J.Y. Zou, C.H. Su, H.H. Luo, Y.Y. Lei, B. Zeng, H.S. Zhu, Z.G. Chen, Curcumin
Biochem. 120 (2019) 2756–2765. converts Foxp3+ regulatory T cells to T helper 1 cells in patients with lung
[176] Q. Yan, X. Chen, H. Gong, P. Qiu, X. Xiao, S. Dang, A. Hong, Y. Ma, Delivery of a cancer, J. Cell. Biochem. 119 (2018) 1420–1428.
TNF-α–derived peptide by nanoparticles enhances its antitumor activity by [205] N. Ahmadi Nasab, H. Hassani Kumleh, M. Beygzadeh, S. Teimourian,
inducing cell-cycle arrest and caspase-dependent apoptosis, Faseb. J. 32 (2018) M. Kazemzad, Delivery of curcumin by a pH-responsive chitosan mesoporous
6948–6964. silica nanoparticles for cancer treatment, Artif. Cell. Nanomed. Biotechnol. 46
[177] Y. Ma, S. Zhao, S. Shen, S. Fang, Z. Ye, Z. Shi, A. Hong, A novel recombinant slow- (2018) 75–81.
release TNF α-derived peptide effectively inhibits tumor growth and angiogensis, [206] G. Arya, M. Das, S.K. Sahoo, Evaluation of curcumin loaded chitosan/PEG
Sci. Rep. 5 (2015) 13595. blended PLGA nanoparticles for effective treatment of pancreatic cancer, Biomed.
[178] M. Khedri, H. Rafatpanah, K. Abnous, P. Ramezani, M. Ramezani, Cancer Pharma. 102 (2018) 555–566.
immunotherapy via nucleic acid aptamers, Int. Immunopharm. 29 (2015) [207] A. Anitha, M. Sreeranganathan, K.P. Chennazhi, V.-K. Lakshmanan,
926–936. R. Jayakumar, In vitro combinatorial anticancer effects of 5-fluorouracil and
[179] S. Iurescia, D. Fioretti, M. Rinaldi, Targeting cytosolic nucleic acid-sensing curcumin loaded N, O-carboxymethyl chitosan nanoparticles toward colon cancer
pathways for cancer immunotherapies, Front. Immunol. 9 (2018) 711. and in vivo pharmacokinetic studies, Eur. J. Pharm. Biopharm. 88 (2014)
[180] L.M. Kranz, M. Diken, H. Haas, S. Kreiter, C. Loquai, K.C. Reuter, M. Meng, 238–251.
D. Fritz, F. Vascotto, H. Hefesha, Systemic RNA delivery to dendritic cells exploits [208] M.A. Khan, M. Zafaryab, S.H. Mehdi, I. Ahmad, M. Rizvi, A. Moshahid,
antiviral defence for cancer immunotherapy, Nature 534 (2016) 396–401. Physicochemical characterization of curcumin loaded chitosan nanoparticles:
[181] F. Maiyo, M. Singh, Folate-targeted mRNA delivery using chitosan-functionalized implications in cervical cancer, Anti-Cancer Agents Med. Chem. 18 (2018)
selenium nanoparticles: potential in cancer immunotherapy, Pharmaceuticals 12 1131–1137.
(2019) 164. [209] W. Song, X. Su, D. Gregory, W. Li, Z. Cai, X. Zhao, Magnetic alginate/chitosan
[182] A. Masjedi, H. Hassannia, F. Atyabi, A. Rastegari, M. Hojjat-Farsangi, A. Namdar, nanoparticles for targeted delivery of curcumin into human breast cancer cells,
H. Soleimanpour, G. Azizi, A. Nikkhoo, G. Ghalamfarsa, Downregulation of A2AR Nanomaterials 8 (2018) 907.
by siRNA loaded PEG-chitosan-lactate nanoparticles restores the T cell mediated [210] S. Rao, Y. Lin, Y. Du, L. He, G. Huang, B. Chen, T. Chen, Designing
anti-tumor responses through blockage of PKA/CREB signaling pathway, Int. J. multifunctionalized selenium nanoparticles to reverse oxidative stress-induced
Biol. Macromol. 133 (2019) 436–445. spinal cord injury by attenuating ROS overproduction and mitochondria
[183] L. Wu, M. Jiang, Y. Sun, X. Yang, Y. Hou, J. Fan, S. Liu, Dissecting the spatial dysfunction, J. Mater. Chem. B 7 (2019) 2648–2656.
heterogeneity of circulating tumor cells reveals CCL5-Treg-mediated immune [211] J. Mudgal, P.P. Mudgal, M. Kinra, R. Raval, Immunomodulatory role of chitosan-
evasion in hepatocellular carcinoma, Can. Res. 78 (2018) 4601. based nanoparticles and oligosaccharides in cyclophosphamide-treated mice,
[184] F. Ahmetlić, T. Riedel, N. Hömberg, V. Bauer, N. Trautwein, A. Geishauser, Scand. J. Immunol. 89 (2019), e12749.
T. Sparwasser, S. Stevanović, M. Röcken, R. Mocikat, Regulatory T cells in an [212] G. Wardani, S.A. Sudjarwo, Immunostimulatory activity of chitosan nanoparticles
endogenous mouse lymphoma recognize specific antigen peptides and contribute on wistar albino rats, Phcog. J. 10 (2018) 892–898.
to immune escape, Cancer Immunol. Res. 7 (2019) 600–608. [213] M. Korbelik, J. Banáth, W. Zhang, T. Hode, S.S. Lam, P. Gallagher, J. Zhao,
[185] Z. Yu, H. Zhao, X. Feng, H. Li, C. Qiu, X. Yi, H. Tang, J. Zhang, Long non-coding H. Zeng, W.R. Chen, N-dihydrogalactochitosan-supported tumor control by
RNA FENDRR acts as a miR-423-5p sponge to suppress the regulatory T cells- photothermal therapy and photothermal therapy-generated vaccine,
mediated immune escape of hepatocellular carcinoma cells, Mol. Ther. Nucleic J. Photochem. Photobiol. B Biol. 204 (2020) 111780.
Acids 17 (2019) 516–529.
3379
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
[214] M. Swierczewska, H.S. Han, K. Kim, J.H. Park, S. Lee, Polysaccharide-based ovalbumin and toll-like receptor ligand monophosphoryl lipid A in poly (D, L-
nanoparticles for theranostic nanomedicine, Adv. Drug Deliv. Rev. 99 (2016) lactic-co-glycolic acid) nanoparticles, J. Biomed. Mater. Res. 81 (2007) 652–662.
70–84. [242] K.T. Gause, A.K. Wheatley, J. Cui, Y. Yan, S.J. Kent, F. Caruso, Immunological
[215] H.B. Moran, J.L. Turley, M. Andersson, E.C. Lavelle, Immunomodulatory principles guiding the rational design of particles for vaccine delivery, ACS Nano
properties of chitosan polymers, Biomaterials 184 (2018) 1–9. 11 (2017) 54–68.
[216] N.M. Salwowska, K.A. Bebenek, D.A. Żądło, D.L. Wcisło-Dziadecka, [243] M. He, L. Huang, X. Hou, C. Zhong, Z.A. Bachir, M. Lan, R. Chen, F. Gao, Efficient
Physiochemical properties and application of hyaluronic acid: a systematic ovalbumin delivery using a novel multifunctional micellar platform for targeted
review, J. Cosmet. Dermatol. 15 (2016) 520–526. melanoma immunotherapy, Int. J. Pharm. 560 (2019) 1–10.
[217] W.M. Payne, D. Svechkarev, A. Kyrychenko, A.M. Mohs, The role of hydrophobic [244] N. Zhang, P.R. Wardwell, R.A. Bader, Polysaccharide-based micelles for drug
modification on hyaluronic acid dynamics and self-assembly, Carbohydr. Polym. delivery, Pharmaceutics 5 (2013) 329–352.
182 (2018) 132–141. [245] J. Wei, Y. Long, R. Guo, X. Liu, X. Tang, J. Rao, S. Yin, Z. Zhang, M. Li, Q. He,
[218] G. Mattheolabakis, L. Milane, A. Singh, M.M. Amiji, Hyaluronic acid targeting of Multifunctional polymeric micelle-based chemo-immunotherapy with immune
CD44 for cancer therapy: from receptor biology to nanomedicine, J. Drug Target. checkpoint blockade for efficient treatment of orthotopic and metastatic breast
23 (2015) 605–618. cancer, Acta Pharm. Sin. B 9 (2019) 819–831.
[219] R.A. Stern, Hyaluronan catabolism: a new metabolic pathway, Eur. J. Cell Biol. 83 [246] X. Guan, J. Chen, Y. Hu, L. Lin, P. Sun, H. Tian, X. Chen, Highly enhanced cancer
(2004) 317–325. immunotherapy by combining nanovaccine with hyaluronidase, Biomaterials 171
[220] G. Huerta-Ángeles, F. Ondreáš, M. Brandejsová, K. Kopecká, H. Vagnerová, (2018) 198–206.
J. Kulhánek, T. Drmota, Formulation of hyaluronan grafted with dodecanoic acid [247] H. Wang, X. Han, Z. Dong, J. Xu, J. Wang, Z. Liu, Hyaluronidase with pH-
as a potential ophthalmic treatment, Carbohydr. Polym. 246 (2020) 116578. responsive dextran modification as an adjuvant nanomedicine for enhanced
[221] G. Huerta-Ángeles, M. Brandejsová, P. Štěpán, V. Pavlík, J. Starigazdová, P. Orzol, photodynamic-immunotherapy of cancer, Adv. Funct. Mater. 29 (2019) 1902440.
K. Kopecká, P. Halamková, J. Kulhánek, V. Velebný, Retinoic acid grafted to [248] Z. Zou, H. Chang, H. Li, S. Wang, Induction of reactive oxygen species: an
hyaluronan for skin delivery: synthesis, stability studies, and biological emerging approach for cancer therapy, Apoptosis 22 (2017) 1321–1335.
evaluation, Carbohydr. Polym. 231 (2020) 115733. [249] H.T.T. Duong, T. Thambi, Y. Yin, S.H. Kim, T.L. Nguyen, V.G. Phan, J. Kim, J.
[222] Z. Cai, H. Zhang, Y. Wei, F. Cong, Hyaluronan-inorganic nanohybrid materials for H. Jeong, D.S. Lee, Degradation-regulated architecture of injectable smart
biomedical applications, Biomacromolecules 18 (2017) 1677–1696. hydrogels enhances humoral immune response and potentiates antitumor activity
[223] K.Y. Choi, H.S. Han, E.S. Lee, J.M. Shin, B.D. Almquist, D.S. Lee, J.H. Park, in human lung carcinoma, Biomaterials 230 (2020) 119599.
Hyaluronic acid–based activatable nanomaterials for stimuli-responsive imaging [250] J.I. Bussio, C. Molina-Perea, J.V. González-Aramundiz, Hyaluronic acid
and therapeutics: beyond CD44-mediated drug delivery, Adv. Mater. 31 (2019) nanocapsules as a platform for needle-free vaccination, Pharmaceutics 11 (2019)
1803549. 246.
[224] J.H. Kim, M.J. Moon, D.Y. Kim, S.H. Heo, Y.Y. Jeong, Hyaluronic acid-based [251] J. Vollmer, A.M. Krieg, Immunotherapeutic applications of CpG
nanomaterials for cancer therapy, Polymers 10 (2018) 1133. oligodeoxynucleotide TLR9 agonists, Adv. Drug Deliv. Rev. 61 (2009) 195–204.
[225] M. Zheng, Z. Yang, S. Chen, H. Wu, Y. Liu, A. Wright, J.-W. Lu, X. Xia, A. Lee, [252] P. Yu, J. Yan, W. Wu, X. Tao, X. Lu, S. Liu, W. Zhu, A CpG oligodeoxynucleotide
J. Zhang, Bioreducible zinc (II)–dipicolylamine functionalized hyaluronic acid enhances the immune response to rabies vaccination in mice, Virol. J. 15 (2018)
mediates safe siRNA delivery and effective glioblastoma RNAi therapy, ACS 174.
Applied Bio Materials 2 (2018) 362–369. [253] S. Beck, I.A. Buhimschi, T.L. Summerfield, W.E. Ackerman, O. Guzeloglu-Kayisli,
[226] P. Gotwals, S. Cameron, D. Cipolletta, V. Cremasco, A. Crystal, B. Hewes, U.A. Kayisli, G. Zhao, F. Schatz, C.J. Lockwood, C.S. Buhimschi, Toll-like receptor
B. Mueller, S. Quaratino, C. Sabatos-Peyton, L. Petruzzelli, Prospects for 9, maternal cell-free DNA and myometrial cell response to CpG
combining targeted and conventional cancer therapy with immunotherapy, Nat. oligodeoxynucleotide stimulation, Am. J. Reprod. Immunol. 81 (2019), e13100.
Rev. Canc. 17 (2017) 286–301. [254] A.N. de la Torre, S. Contractor, I. Castaneda, C.S. Cathcart, D. Razdan, D. Klyde,
[227] M.H. Spitzer, Y. Carmi, N.E. Reticker-Flynn, S.S. Kwek, D. Madhireddy, M. P. Kisza, S.F. Gonzales, A.M. Salazar, A phase I trial using local regional
M. Martins, P.F. Gherardini, T.R. Prestwood, J. Chabon, S.C. Bendall, Systemic treatment, nonlethal irradiation, intratumoral and systemic polyinosinic-
immunity is required for effective cancer immunotherapy, Cell 168 (2017) polycytidylic acid polylysine carboxymethylcellulose to treat liver cancer: in
487–502. search of the abscopal effect, J. Hepatocell. Carcinoma 4 (2017) 111–121.
[228] S. Farkona, E.P. Diamandis, I.M. Blasutig, Cancer immunotherapy: the beginning [255] C. Liu, X. Chu, P. Sun, X. Feng, W. Huang, H. Liu, Y. Ma, Synergy effects of
of the end of cancer? BMC Med. 14 (2016) 73. Polyinosinic-polycytidylic acid, CpG oligodeoxynucleotide, and cationic peptides
[229] F. Kroschinsky, F. Stölzel, S. von Bonin, G. Beutel, M. Kochanek, M. Kiehl, to adjuvant HPV E7 epitope vaccine through preventive and therapeutic
P. Schellongowski, Intensive Care in Hematological and Oncological Patients immunization in a TC-1 grafted mouse model, Hum. Vaccines Immunother. 14
(iCHOP) Collaborative Group. New drugs, new toxicities: severe side effects of (2018) 931–940.
modern targeted and immunotherapy of cancer and their management, Crit. Care [256] B. Bayyurt, G. Tincer, K. Almacioglu, E. Alpdundar, M. Gursel, I. Gursel,
21 (2017) 89. Encapsulation of two different TLR ligands into liposomes confer protective
[230] P. Cukier, F.C. Santini, M. Scaranti, A.O. Hoff, Endocrine side effects of cancer immunity and prevent tumor development, J. Contr. Release 247 (2017)
immunotherapy, Endocr. Relat. Canc. 24 (2017) T331–T347. 134–144.
[231] H. Knopf-Marques, M. Pravda, L. Wolfova, V. Velebny, P. Schaaf, N.E. Vrana, [257] C. Liu, X. Chu, M. Yan, J. Qi, H. Liu, F. Gao, R. Gao, G. Ma, Y. Ma, Encapsulation
P. Lavalle, Hyaluronic acid and its derivatives in coating and delivery systems: of Poly I: C and the natural phosphodiester CpG ODN enhanced the efficacy of a
applications in tissue engineering, regenerative medicine and hyaluronic acid-modified cationic lipid-PLGA hybrid nanoparticle vaccine in TC-
immunomodulation, Adv. Healthc. Mater. 5 (2016) 2841–2855. 1-grafted tumors, Int. J. Pharm. 553 (2018) 327–337.
[232] M. Litwiniuk, A. Krejner, M.S. Speyrer, A.R. Gauto, T. Grzela, Hyaluronic acid in [258] Z. Cai, F. Xin, Z. Wei, M. Wu, X. Lin, X. Du, G. Chen, D. Zhang, Z. Zhang, X. Liu,
inflammation and tissue regeneration, Wounds 28 (2016) 78–88. Photodynamic therapy combined with antihypoxic signaling and cpg adjuvant as
[233] S.S.M. Lee-Sayer, Y. Dong, A.A. Arif, M. Olsson, K.L. Brown, P. Johnson, The an in situ tumor vaccine based on metal–organic framework nanoparticles to
where, when, how, and why of hyaluronan binding by immune cells, Front. boost cancer immunotherapy, Adv. Healthc. Mater. 9 (2020) 1900996.
Immunol. 6 (2015) 150. [259] K. Lee, H. Zhang, D.Z. Qian, S. Rey, J.O. Liu, G.L. Semenza, Acriflavine inhibits
[234] N. Maeshima, G.F. Poon, M. Dosanjh, J. Felberg, S.S. Lee, J.L. Cross, HIF-1 dimerization, tumor growth, and vascularization, Proc. Natl. Acad. Sci. U.S.
D. Birkenhead, P. Johnson, Hyaluronan binding identifies the most proliferative A. 106 (2009) 17910–17915.
activated and memory T cells, Eur. J. Immunol. 41 (2011) 1108–1119. [260] A. Mangraviti, T. Raghavan, F. Volpin, N. Skuli, D. Gullotti, J. Zhou, L. Asnaghi,
[235] P.L. Bollyky, J.D. Lord, S.A. Masewicz, S.P. Evanko, J.H. Buckner, T.N. Wight, G. E. Sankey, HIF-1α- Targeting acriflavine provides long term survival and
T. Nepom, Cutting edge: high molecular weight hyaluronan promotes the radiological tumor response in brain cancer therapy, Sci. Rep. 7 (2017) 14978.
suppressive effects of CD4+CD25+ regulatory T cells, J. Immunol. 179 (2007) [261] J. Fan, X. Yang, Z. Bi, Acriflavine suppresses the growth of human osteosarcoma
744–747. cells through apoptosis and autophagy, Tumor Biol. 35 (2014) 9571–9576.
[236] M. Miyazaki, E. Yuba, H. Hayashi, A. Harada, K. Kono, Development of pH- [262] E. Martino, S. Della Volpe, E. Terribile, E. Benetti, M. Sakaj, A. Centamore,
responsive hyaluronic acid-based antigen carriers for induction of antigen-specific A. Sala, S. Collina, The long story of camptothecin: from traditional medicine to
cellular immune responses, ACS Biomater. Sci. Eng. 5 (2019) 5790–5797. drugs, Bioorg. Med. Chem. Lett. 27 (2017) 701–707.
[237] C.G. Park, C.A. Hartl, D. Schmid, E.M. Carmona, H. Kim, M. Goldberg, Extended [263] G. L Beretta, L. Gatti, P. Perego, N. Zaffaroni, Camptothecin resistance in cancer:
release of perioperative immunotherapy prevents tumor recurrence and insights into the molecular mechanisms of a DNA-damaging drug, Curr. Med.
eliminates metastases, Sci. Transl. Med. 10 (2018), eaar1916. Chem. 20 (2013) 1541–1565.
[238] B.K. Helmich, R.W. Dutton, The role of adoptively transferred CD8 T cells and [264] W. Sun, Y. Du, X. Liang, C. Yu, J. Fang, W. Lu, X. Guo, J. Tian, Y. Jin, J. Zheng,
host cells in the control of the growth of the EG7 thymoma: factors that determine Synergistic triple-combination therapy with hyaluronic acid-shelled PPy/CPT
the relative effectiveness and homing properties of Tc1 and Tc2 effectors, nanoparticles results in tumor regression and prevents tumor recurrence and
J. Immunol. 166 (2001) 6500–6508. metastasis in 4T1 breast cancer, Biomaterials 217 (2019) 119264.
[239] C. Solbrig, J. Saucier-Sawyer, V. Cody, W. Saltzman, D. Hanlon, Polymer [265] Y. Chen, M. Chang, W. Cheng, Metronomic chemotherapy and immunotherapy in
nanoparticles for immunotherapy from encapsulated tumor-associated antigens cancer treatment, Canc. Lett. 400 (2017) 282–292.
and whole tumor cells, Mol. Pharm. 4 (2007) 47–57. [266] C. Brignone, M. Gutierrez, F. Mefti, E. Brain, R. Jarcau, F. Cvitkovic, N. Bousetta,
[240] M. Yang, Y. Mine, Novel T-cell epitopes of ovalbumin in BALB/c mouse: potential J. Medioni, J. Gligorov, C. Grygar, M. Marcu, F. Triebel, First-line
for peptide-immunotherapy, Biochem. Biophys. Res. Commun. 378 (2009) chemoimmunotherapy in metastatic breast carcinoma: combination of paclitaxel
203–208. and IMP321 (LAG-3Ig) enhances immune responses and antitumor activity,
[241] S. Hamdy, P. Elamanchili, A. Alshamsan, O. Molavi, T. Satou, J. Samuel, J. Transl. Med. 8 (2010), 71.
Enhanced antigen-specific primary CD4+ and CD8+ responses by codelivery of
3380
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
[267] L. Zitvogel, L. Apetoh, F. Ghiringhelli, G. Kroemer, Immunological aspects of [297] H. Choukaife, A.A. Doolaanea, M. Alfatama, Alginate nanoformulation: influence
cancer chemotherapy, Nat. Rev. Immunol. 8 (2008) 59–73. of process and selected variables, Pharmaceuticals 13 (2020) 335.
[268] I. Mellman, G. Coukos, G. Dranoff, Cancer immunotherapy comes of age, Nature [298] P. Severino, C.F.D. Silva, L.N. Andrade, D.M.D.L. Oliveira, J.C. De Campos, E.
480 (2011) 480–489. B. Souto, Alginate nanoparticles for drug delivery and targeting, Curr.
[269] L.A. Emens, G. Middleton, The interplay of immunotherapy and chemotherapy: Pharmaceut. Des. 25 (2019) 1312–1334.
harnessing potential synergies, Cancer Immunol. Res. 3 (2015) 436–443. [299] A. Thomas, K. Harding, K. Moore, Alginates from wound dressings activate
[270] S.P. Singh, M. Sharma, P.K. Gupta, Cytotoxicity of curcumin silica nanoparticle human macrophages to secrete tumour necrosis factor-α, Biomaterials 21 (2000)
complexes conjugated with hyaluronic acid on colon cancer cells, Int. J. Biol. 1797–1802.
Macromol. 74 (2015) 162–170. [300] H. Zhang, J. Bi, H. Yi, T. Fan, Q. Ruan, L. Cai, Y.H. Chen, X. Wan, Silencing c-Rel
[271] Z. Yang, N. Sun, R. Cheng, C. Zhao, J. Liu, Z. Tian, Hybrid nanoparticles coated in macrophages dampens Th1 and Th17 immune responses and alleviates
with hyaluronic acid lipoid for targeted co-delivery of paclitaxel and curcumin to experimental autoimmune encephalomyelitis in mice, Immunol. Cell Biol. 95
synergistically eliminate breast cancer stem cells, J. Mater. Chem. B 5 (2017) (2017) 593–600.
6762–6775. [301] H.C. Bygd, K.M. Bratlie, The effect of chemically modified alginates on
[272] H.-Y. Seok, N.S. Rejinold, K.M. Lekshmi, K. Cherukula, I.-K. Park, Y.-C. Kim, CD44 macrophage phenotype and biomolecule transport, J. Biomed. Mater. Res. 104
targeting biocompatible and biodegradable hyaluronic acid cross-linked zein (2016) 1707–1719.
nanogels for curcumin delivery to cancer cells: in vitro and in vivo evaluation, [302] L. Zhu, F. Ge, L. Yang, W. Li, S. Wei, Y. Tao, G. Du, Alginate particles with
J. Contr. Release 280 (2018) 20–30. ovalbumin (OVA) peptide can serve as a carrier and adjuvant for immune therapy
[273] P. Kesharwani, L. Xie, S. Banerjee, G. Mao, S. Padhye, F.H. Sarkar, A.K. Iyer, in B16-OVA cancer model, Med. Sci. Monit. Basic Res. 23 (2017) 166–172.
Hyaluronic acid-conjugated polyamidoamine dendrimers for targeted delivery of [303] J. Yuan, L. Guo, S. Wang, D. Liu, X. Qin, L. Zheng, C. Tian, X. Han, R. Chen, R. Yin,
3, 4-difluorobenzylidene curcumin to CD44 overexpressing pancreatic cancer Preparation of self-assembled nanoparticles of ε-polylysine-sodium alginate: a
cells, Colloids Surf. B Biointerfaces 136 (2015) 413–423. sustained-release carrier for antigen delivery, Colloids Surf. B Biointerfaces 171
[274] B. Wang, W. Zhang, X. Zhou, M. Liu, X. Hou, Z. Cheng, D. Chen, Development of (2018) 406–412.
dual-targeted nano-dandelion based on an oligomeric hyaluronic acid polymer [304] S.A. Bencherif, R.W. Sands, O.A. Ali, W.A. Li, S.A. Lewin, T. Braschler, T.S. Shih,
targeting tumor-associated macrophages for combination therapy of non-small C.S. Verbeke, D. Bhatta, G. Dranoff, Injectable cryogel-based whole-cell cancer
cell lung cancer, Drug Deliv. 26 (2019) 1265–1279. vaccines, Nat. Commun. 6 (2015) 7556. -7556.
[275] R. Farajzadeh, Y. Pilehvar-Soltanahmadi, M. Dadashpour, S. Javidfar, J. Lotfi- [305] N. Shah, A.J. Najibi, T. Shih, A.S. Mao, A. Sharda, D.T. Scadden, D.J. Mooney,
Attari, H. Sadeghzadeh, V. Shafiei-Irannejad, N. Zarghami, Nano-encapsulated A biomaterial-based vaccine eliciting durable tumour-specific responses against
metformin-curcumin in PLGA/PEG inhibits synergistically growth and hTERT acute myeloid leukaemia, Nat. Biomed. Eng. 4 (2020) 40–51.
gene expression in human breast cancer cells, Artif. Cell. Nanomed. Biotechnol. [306] K.-C. Cheng, A. Demirci, J.M. Catchmark, Pullulan: biosynthesis, production, and
46 (2018) 917–925. applications, Appl. Microbiol. Biotechnol. 92 (2011) 29–44.
[276] S. Bose, A.K. Panda, S. Mukherjee, G. Sa, Curcumin and tumor immune-editing: [307] R.S. Singh, N. Kaur, J.F. Kennedy, Pullulan and pullulan derivatives as promising
resurrecting the immune system, Cell Div. 10 (2015) 6. biomolecules for drug and gene targeting, Carbohydr. Polym. 123 (2015)
[277] A. Deguchi, Curcumin targets in inflammation and cancer, Endocr. Metab. 190–207.
Immune Disord. Drug Targets 15 (2015) 88–96. [308] R.S. Singh, N. Kaur, V. Rana, J.F. Kennedy, Pullulan: a novel molecule for
[278] A.A. Momtazi-Borojeni, S.M. Haftcheshmeh, S.A. Esmaeili, T.P. Johnston, biomedical applications, Carbohydr. Polym. 171 (2017) 102–121.
E. Abdollahi, A. Sahebkar, Curcumin: a natural modulator of immune cells in [309] N. Kyogoku, H. Ikeda, T. Tsuchikawa, T. Abiko, A. Fujiwara, T. Maki,
systemic lupus erythematosus, Autoimmun. Rev. 17 (2018) 125–135. Y. Yamamura, M. Ichinokawa, K. Tanaka, N. Imai, Time-dependent transition of
[279] L. Vecchi Brumatti, A. Marcuzzi, P.M. Tricarico, V. Zanin, M. Girardelli, A. the immunoglobulin G subclass and immunoglobulin E response in cancer
M. Bianco, Curcumin and inflammatory bowel disease: potential and limits of patients vaccinated with cholesteryl pullulan-melanoma antigen gene-A4
innovative treatments, Molecules 19 (2014) 21127–21153. nanogel, Oncol. Lett. 12 (2016) 4493–4504.
[280] E. Abdollahi, A.A. Momtazi, T.P. Johnston, A. Sahebkar, Therapeutic effects of [310] R. Miura, S.-i. Sawada, S.-a. Mukai, Y. Sasaki, K. Akiyoshi, Antigen delivery to
curcumin in inflammatory and immune-mediated diseases: a nature-made jack-of- antigen presenting cells for adaptive immune response by self-assembled anionic
all-trades? J. Cell. Physiol. 233 (2018) 830–848. polysaccharide nanogel vaccines, Biomacromolecules 21 (2019) 621–629.
[281] A.E. Barberio, S.C. Echavarria, M.B. Melo, T. Tokatlian, E.C. Dreaden, P. [311] J.-H. Kang, Y. Tachibana, W. Kamata, A. Mahara, M. Harada-Shiba, T. Yamaoka,
T. Hammond, D.J. Irvine, Layer-by-layer nanoparticles for cytokine therapy in Liver-targeted siRNA delivery by polyethylenimine (PEI)-pullulan carrier, Bioorg.
cancer treatment, in: Google Patents, 2019. Med. Chem. 18 (2010) 3946–3950.
[282] F. Chen, G. Huang, Preparation and application of dextran and its derivatives as [312] L. Zhao, M. Liu, J. Wang, G. Zhai, Chondroitin sulfate-based nanocarriers for
carriers, Int. J. Biol. Macromol. 145 (2020) 827–834. drug/gene delivery, Carbohydr. Polym. 133 (2015) 391–399.
[283] G. Huang, H. Huang, Application of dextran as nanoscale drug carriers, Nano 13 [313] M. Liu, H. Du, A.R. Khan, J. Ji, A. Yu, G. Zhai, Redox/enzyme sensitive
(2018) 3149–3158. chondroitin sulfate-based self-assembled nanoparticles loading docetaxel for the
[284] M. Zhang, Y. Huang, W. Pan, X. Tong, Q. Zeng, T. Su, X. Qi, J. Shen, inhibition of metastasis and growth of melanoma, Carbohydr. Polym. 184 (2018)
Polydopamine-incorporated dextran hydrogel drug carrier with tailorable 82–93.
structure for wound healing, Carbohydr. Polym. 253 (2021) 117213. [314] M. Liu, H. Du, G. Zhai, Self-assembled nanoparticles based on chondroitin sulfate-
[285] Y. Zhang, P. Li, H. Pan, L. Liu, M. Ji, N. Sheng, C. Wang, L. Cai, Y. Ma, Retinal- deoxycholic acid conjugates for docetaxel delivery: effect of degree of substitution
conjugated pH-sensitive micelles induce tumor senescence for boosting breast of deoxycholic acid, Colloids Surf. B Biointerfaces 146 (2016) 235–244.
cancer chemotherapy, Biomaterials 83 (2016) 219–232. [315] M. Okubo, M. Miyazaki, E. Yuba, A. Harada, Chondroitin sulfate-based pH-
[286] I. Wasiak, A. Kulikowska, M. Janczewska, M. Michalak, I.A. Cymerman, sensitive polymer-modified liposomes for intracellular antigen delivery and
A. Nagalski, P. Kallinger, W.W. Szymanski, T. Ciach, Dextran nanoparticle induction of cancer immunity, Bioconjugate Chem. 30 (2019) 1518–1529.
synthesis and properties, PLoS ONE 11 (2016), e0146237. [316] A.P. Sherje, B.R. Dravyakar, D. Kadam, M. Jadhav, Cyclodextrin-based
[287] Z. Gu, C. Wang, Y. Ye, Enhanced cancer immunotherapy by microneedle patch- nanosponges: a critical review, Carbohydr. Polym. 173 (2017) 37–49.
assisted delivery, in: Google Patents, 2019. [317] X. Yao, P. Huang, Z. Nie, Cyclodextrin-based polymer materials: from controlled
[288] W. Zhang, M. An, J. Xi, H. Liu, Targeting CpG adjuvant to lymph node via dextran synthesis to applications, Prog. Polym. Sci. 93 (2019) 1–35.
conjugate enhances antitumor immunotherapy, Bioconjugate Chem. 28 (2017) [318] Y. Luo, K. Pan, Q. Zhong, Casein/pectin nanocomplexes as potential oral delivery
1993–2000. vehicles, Int. J. Pharm. 486 (2015) 59–68.
[289] E. Yuba, Design of pH-sensitive polymer-modified liposomes for antigen delivery [319] Y. Liu, Y. Zong, Z. Yang, M. Luo, G. Li, W. Yingsa, Y. Cao, M. Xiao, T. Kong, J. He,
and their application in cancer immunotherapy, Polym. J. 48 (2016) 761–771. Dual-targeted controlled delivery based on folic acid modified pectin-based
[290] J.M. Shin, S.H. Song, N.V. Rao, E.S. Lee, H. Ko, J.H. Park, A carboxymethyl nanoparticles for combination therapy of liver cancer, ACS Sustain. Chem. Eng. 7
dextran-based polymeric conjugate as the antigen carrier for cancer (2019) 3614–3623.
immunotherapy, Biomater. Res. 22 (2018) 21. [320] I. Hira, A. Kumar, R. Kumari, A.K. Saini, R.V. Saini, Pectin-guar gum-zinc oxide
[291] Z. Huang, Z. Zhang, Y. Jiang, D. Zhang, J. Chen, L. Dong, J. Zhang, Targeted nanocomposite enhances human lymphocytes cytotoxicity towards lung and
delivery of oligonucleotides into tumor-associated macrophages for cancer breast carcinomas, Mater. Sci. Eng. C-Mater. Biol. Appl. 90 (2018) 494–503.
immunotherapy, J. Contr. Release 158 (2012) 286–292. [321] J. Hirsh, Heparin, N. Engl. J. Med. 324 (1991) 1565–1574.
[292] T. Bauleth-Ramos, T.Y. Shih, M.A. Shahbazi, A.J. Najibi, A.S. Mao, D. Liu, [322] K. Einhäupl, A. Villringer, S. Mehraein, C. Garner, M. Pellkofer, R. Haberl,
P. Granja, H.A. Santos, B. Sarmento, D.J. Mooney, Acetalated dextran H. Pfister, P. Schmiedek, W. Meister, Heparin treatment in sinus venous
nanoparticles loaded into an injectable alginate cryogel for combined thrombosis, Lancet 338 (1991) 597–600.
chemotherapy and cancer vaccination, Adv. Funct. Mater. 29 (2019) 1903686. [323] H. Du, M. Liu, A. Yu, J. Ji, G. Zhai, Insight into the role of dual-ligand
[293] Y. Singh, V.K. Pawar, J.G. Meher, K. Raval, A. Kumar, R. Shrivastava, modification in low molecular weight heparin based nanocarrier for targeted
S. Bhadauria, M.K. Chourasia, Targeting tumor associated macrophages (TAMs) delivery of doxorubicin, Int. J. Pharm. 523 (2017) 427–438.
via nanocarriers, J. Contr. Release 254 (2017) 92–106. [324] B. Mulloy, J. Hogwood, E. Gray, R. Lever, C.P. Page, Pharmacology of heparin and
[294] M. Cully, Re-educating tumour-associated macrophages with nanoparticles, Nano related drugs, Pharmacol. Rev. 68 (2016) 76–141.
16 (2017) 369–370. [325] A. Onishi, K. St Ange, J.S. Dordick, R.J. Linhardt, Heparin and anticoagulation,
[295] S. Jana, K. Kumar Sen, A. Gandhi, Alginate based nanocarriers for drug delivery Front. Biosci. 21 (2016) 1372–1392.
applications, Curr. Pharmaceut. Des. 22 (2016) 3399–3410. [326] C. Xia, S. Yin, S. Xu, G. Ran, M. Deng, L. Mei, X. Tang, J. Rao, M. Li, Z. Zhang, Low
[296] M. Lopes, B. Abrahim, F. Veiga, R. Seica, L.M. Cabral, P. Arnaud, J.C. Andrade, A. molecular weight heparin-coated and dendrimer-based core-shell nanoplatform
J. Ribeiro, Preparation methods and applications behind alginate-based particles, with enhanced immune activation and multiple anti-metastatic effects for
Expet Opin. Drug Deliv. 14 (2017) 769–782. melanoma treatment, Theranostics 9 (2019) 337–354.
3381
Y. Zeng et al. Bioactive Materials 6 (2021) 3358–3382
[327] F. Shao, M. Zhang, L. Xu, D. Yin, M. Li, Q. Jiang, Q. Zhang, Y. Yang, Multiboosting [334] Z. Zhang, Y. Du, T. Liu, K.-H. Wong, T. Chen, Systematic acute and subchronic
of cancer immunotherapy by a core–shell delivery system, Mol. Pharm. 17 (2020) toxicity evaluation of polysaccharide–protein complex-functionalized selenium
338–348. nanoparticles with anticancer potency, Biomater. Sci. 7 (2019) 5112–5123.
[328] L. Zhang, Y. Li, C. Wang, G. Li, Y. Zhao, Y. Yang, Synthesis of methylprednisolone [335] H.-p. Zhao, Y. Zhang, Z. Liu, J.-y. Chen, S.-y. Zhang, X.-d. Yang, H.-l. Zhou, Acute
loaded ibuprofen modified inulin based nanoparticles and their application for toxicity and anti-fatigue activity of polysaccharide-rich extract from corn silk,
drug delivery, Mater. Sci. Eng. C-Mater. Biol. Appl. 42 (2014) 111–115. Biomed. Pharmacother. 90 (2017) 686–693.
[329] C. Scialabba, M. Licciardi, N. Mauro, F. Rocco, M. Ceruti, G. Giammona, Inulin- [336] S. Kruger, B.L. Cadilha, M. Von Bergweltbaildon, S. Endres, S. Kobold, Challenges
based polymer coated SPIONs as potential drug delivery systems for targeted in clinical trial design for T cell-based cancer immunotherapy, Clin. Pharmacol.
cancer therapy, Eur. J. Pharm. Biopharm. 88 (2014) 695–705. Ther. 107 (2020) 47–49.
[330] L. Zhang, G. Li, M. Gao, X. Liu, B. Ji, R. Hua, Y. Zhou, Y. Yang, RGD-peptide [337] S.F. Slovin, Emerging treatments in management of prostate cancer: biomarker
conjugated inulin-ibuprofen nanoparticles for targeted delivery of Epirubicin, validation and endpoints for immunotherapy clinical trial design, ImmunoTargets
Colloids Surf. B Biointerfaces 144 (2016) 81–89. Ther. 3 (2013) 1–8.
[331] M.K. Rajput, S.S. Kesharwani, S. Kumar, P. Muley, S. Narisetty, H. Tummala, [338] E.T. Roussos Torres, A.L. Epstein, Adopting an alternative structure for clinical
Dendritic cell-targeted nanovaccine delivery system prepared with an immune- trials in immunotherapy, Expert Rev. Anticancer Ther. (2021) 1–3.
active polymer, ACS Appl. Mater. Interfaces 10 (2018) 27589–27602. [339] A. Cesano, M.A. Cannarile, S. Gnjatic, B. Gomes, J. Guinney, V. Karanikas,
[332] M.M. Joseph, N. Hari, R.K. Pillai, A.J. Nair, S. Therakathinal T, Galactoxyloglucan M. Karkada, J.M. Kirkwood, B. Kotlan, G.V. Masucci, E. Meeusen, A. Monette,
endowed biogenic nano-immunobiotics arrests microbial growth and elicits A. Naing, V. Thorsson, N. Tschernia, E. Wang, D.K. Wells, T.L. Wyant, S. Rutella,
antitumor immunity, ACS Applied Bio Materials 3 (2020) 801–814. Society for Immunotherapy of Cancer clinical and biomarkers data sharing
[333] G. Pang, C. Chen, Y. Liu, T. Jiang, H. Yu, Y. Wu, Y. Wang, F.-J. Wang, Z. Liu, L. resource document: volume II—practical challenges, J. Immunothera. Cancer 8
W. Zhang, Bioactive polysaccharide nanoparticles improve radiation-induced (2020), e001472.
abscopal effect through manipulation of dendritic cells, ACS Appl. Mater.
Interfaces 11 (2019) 42661–42670.
3382