Abstract
Nanotechnology has been increasingly utilized in anticancer treatment owing to its ability of engineering functional nanocarriers that enhance therapeutic effectiveness while minimizing adverse effects. Inorganic nanoparticles (INPs) are prevalent nanocarriers to be customized for a wide range of anticancer applications, including theranostics, imaging, targeted drug delivery, and therapeutics, because they are advantageous for their superior biocompatibility, unique optical properties, and capacity of being modified via versatile surface functionalization strategies. In the past decades, the high adaptation of INPs in this emerging immunotherapeutic field makes them good carrier options for tumor immunotherapy and combination immunotherapy. Tumor immunotherapy requires targeted delivery of immunomodulating therapeutics to tumor locations or immunological organs to provoke immune cells and induce tumor-specific immune response while regulating immune homeostasis, particularly switching the tumor immunosuppressive microenvironment. This review explores various INP designs and formulations, and their employment in tumor immunotherapy and combination immunotherapy. We also introduce detailed demonstrations of utilizing surface engineering tactics to create multifunctional INPs. The generated INPs demonstrate the abilities of stimulating and enhancing the immune response, specific targeting, and regulating cancer cells, immune cells, and their resident microenvironment, sometimes along with imaging and tracking capabilities, implying their potential in multitasking immunotherapy. Furthermore, we discuss the promises of INP-based combination immunotherapy in tumor treatments.
Introduction
Cancer is the primary cause of mortality worldwide. Despite the development of several cancer treatment approaches, including surgery, radiotherapy, and chemotherapy, these treatments have shown unsatisfactory therapeutic results. Each therapeutic modality has limitations. Surgery is generally restricted to a limited surgical area, and the effectiveness of radiotherapy and chemotherapy is often compromised owing to difficulties in accurate and efficient targeting, leading to undesirable effects such as tissue injury, drug resistance, and systemic toxicity [1–4]. To overcome these difficulties, extensive research on developing other cancer treatment techniques is in progress, and immune system-reliant cancer immunotherapy has become a huge success in the last decade. The highlighted advantage of immunotherapy is that it induces the generation of circulating effector and memory cells, which not only eliminate primary cancer but also prominently suppress the progression of distant cancer, and efficiently prevent metastasis and reoccurrence of the same type of cancer [5–9]. Immunotherapy expands the targets beyond cancer tissues to cancer-related immune systems. Anchoring and regulating immune cells, including macrophages, DCs, T cells, and immune organs such as LNs, contributes to establishing an internal environment that combats cancer [10–13]. However, similar to other therapeutic techniques, direct administration of immunotherapeutic agents leads to nonspecific targeting, resulting in severe adverse effects. A high dose causes immune imbalance, while a low dose results in no therapeutic response. To address these limitations, nanomedicines have shown potential and promise in cancer immunotherapy. Nanomedicines are generally composed of functional nanocarriers and immunotherapeutic drugs, exhibiting enhanced therapeutic efficiency and reduced side effects, primarily through ligand-guided active drug delivery and specific stimulus-based drug release. Additionally, encapsulating immunotherapeutic agents in nanocarriers improves drug stability, increases half-life, and increases maximum tolerated dose of some drugs, such as antibodies [14–17]. Among several nanocarriers, inorganic nanoparticles (INPs) are undisputed pioneers because of their physicochemical advantages, particularly their specific optical, electrical, magnetic, and catalytic properties, which not only render them useful for drug delivery but also provide platforms for therapeutic, appended imaging, and diagnostic systems [18–22]. However, recent clinical results suggest that immunotherapy alone is insufficient for complete remission and shows more favorable results when combined with other forms of therapeutics. Immunotherapy is compatible with a wide range of conventional anticancer therapeutic techniques because cancer cell death induced by other strategies can aggravate the anticancer immune response [23–27].
In this review, we provide a concise overview of the engineering concepts and recent advancements in cancer immunotherapy (Fig. 1). Specifically, it focuses on the utilization of INPs as nanocarriers and their combination with immunotherapy. We systematically summarized the most representative INPs, providing a detailed explanation of their alterations and utilization in the field of cancer immunotherapy (Table 1). Furthermore, we enumerated the possible targets of nanoparticulated immunotherapeutic agents and discussed the benefits of employing nanocarrier-based cancer immunotherapy. Finally, we anticipate and consider future advancements in cancer immunotherapy.
Fig. 1.
Functionalization of common INPs and their application in combined immunotherapy. A single INP, particularly those with intrinsic properties such as PCE or superparamagnetism, can be modified with immunomodulators and conventional therapeutic agents (chemical drugs, PDT drugs, or radiation agents for synergistic or imaging-guided anticancer therapy with enhanced therapeutic outcomes).
Table 1.
Nanocarrier type | Functionalization | Application | Reference |
---|---|---|---|
Au nanocarriers | HA and OVA | Vaccination | [272] |
TLR7 ligand 2 and α-mannose | Serve as adjuvant for enhanced vaccination | [273] | |
Bcl-2 siRNA | Bcl-2 silencing for enhanced B cell function | [274] | |
Imatinib mesylate and anti-STAT3 siRNA | Enhanced inhibition of STAT3 protein for enhanced anticancer effect | [275] | |
PD-L1 siRNA | Down-regulated expression of PD-L1 protein for immune checkpoint blockade | [108] | |
aPD-1 and aPD-L1 | Bind to PD-1 and PD-L1 for immune checkpoint blockade | [276] | |
MSN | OVA and CpG | DC-targeted delivery and OVA-specific CTL activation for cancer growth suppression | [277] |
OVA and polyIC | Enhanced TAA uptake and DC maturation, and decreased necessary dose of polyIC for cancer immunotherapy | [278] | |
GM-CSF, CpG, and OVA | In situ self-assembly of a pocket form MS rods, and recruit, activate, and liberate immune cells (DC) for enhanced immunotherapy | [279] | |
IONP | MPLA | Immunotherapy and MRI imaging | [280] |
Carbon nanomaterials | Formed injectable hydrogel with PEI, OVA mRNA, and R848 | Prolonged period of vaccination | [70] |
Alum-Ure B | Served as adjuvant for enhanced immunotherapy | [281] | |
Neoantigen, CpG, and aPD-L1 | Enhanced and personalized cancer vaccination | [282] | |
CpG, aCD40, OVA | Cancer immunotherapy | [63] | |
Durvalumab, PEI, and siTrem2 | Cancer immunotherapy | [283] | |
PDMAEMA and siPD-L1 | Cancer immunotherapy | [284] | |
B16F10-Ag and CT26-Ag | Cancer immunotherapy | [285] | |
OVA-mRNA | Cancer immunotherapy | [286] | |
PD-L1 and E3 ligase | Degrade PD-L1, activate STING pathway to promote DC maturation, and reshape the immunosuppressive TME for cancer immunotherapy | [287] | |
CaP NP | Minicircle DNA encoding BsAbEPH | Fabrication of needle-type CaP nanomaterials for enhanced transfection and antitumor immunotherapy | [288] |
ATP and pOVA vaccine | Enhanced vaccination and promoted costimulatory | [76] | |
Zoledronic acid | Improvement of γδT cell proliferation and M1-bias macrophage polarization for enhanced cancer immunotherapy | [289] | |
DSPE-PEG2000 and aptamer-DNAzyme | Activated multiple inflammation-related signaling pathways, accelerated DAMP release, and depleted PD-L1 to enhance immunotherapy | [22] | |
MnNP | Being dopped on ZnO2 NP | Together formed NP with ZnO2 for synergistic cancer immunotherapy | [290] |
Curcumin and Ca+ | Activation of cGAS-STING signaling pathway and reprogramming of TME for enhanced cancer immunotherapy | [291] |
INPs for Immunotherapy
Gold NPs
Gold NPs (AuNPs) have emerged as leading nanocarriers for cancer therapy, in the past decade owing to their advantages from synthesis to functionalization. AuNPs can be easily synthesized with controllable sizes (from several nanometers to micrometers) and shapes (spherical, rod, star, and cubic), and their strong affinity between Au/sulfur and Au/amine endows AuNPs with surface-modifiable capacity. Upon functionalization and conjugation of molecules of different types and functions, such as polymers, biomolecules, and therapeutics, with accordance to the application, AuNPs can be further stabilized and rendered specific properties of targeting, theranostics, imaging, and drug delivery. Owing to the unique localized surface plasmon resonance (LSPR) property of AuNPs, which have strong light scattering and optical absorption, AuNPs are considered novel and efficient contrast agents for computed tomography (CT) imaging.
With increasing attention to immunotherapy in recent years, AuNPs have been predominantly used as vesicles to transport immunotherapeutic cargos (Fig. 2A). Although AuNPs are regarded as bioinert materials, many studies have shown that they can trigger an array of immune responses, alternating the anticancer immunotherapeutic effect. Specifically, one research suggested that AuNPs interacted with extracellular interleukin-1β (IL-1β) to block the production of inflammatory cytokines secreted by the human myeloid leukemia cell THP-1 [28]; another study demonstrated that AuNPs might impair toll-like receptor 9 (TLR9) signaling in macrophages via binding with high-mobility group box 1 (HMGB-1), a nucleic acid (NA) sensor that constitutively associate with TLR9 for activation [29]. The effect of AuNPs on immunomodulation is size and shape dependent. Smaller AuNPs (~5 nm) had a greater influence on regulating immune responses than larger AuNPs (~35 nm) [29,30]. In addition, compared to spherical and cubic AuNPs, rods could be endocytosed by antigen-presenting cells (APCs) involving macrophages and DCs more efficiently and showed stronger response to the secretion of the inflammasome-related cytokines IL-1β and IL-18. In contrast, larger spherical and cubic NPs stimulated increased production of pro-inflammatory cytokines, such as NF-R, IL-6, IL-12, and granulocyte-macrophage colony-stimulating factor (GM-CSF) [31]. The regulatory function of AuNPs in immune responses is predicted by their interaction with critical molecules and/or internalization by particular cells, which either strengthens or weakens the immune response. The behavior of AuNPs is significantly influenced by their shape, surface area, and aspect ratio. Therefore, AuNPs must be meticulously engineered and modified to maximize their application. Although AuNPs have been proven to be capable of modulating immune signaling, they cannot induce strong immune responses to combat tumors. To realize effective cancer suppression and elimination, AuNPs should be further decorated to amplify anticancer signals and/or alter the cancer microenvironment from immune evasive to immune active. The anticancer decoration strategies can be classified into 3 major categories: (a) decoration with cancer vaccines, including encapsulation of specific tumor-associated antigens (TAAs) and adjuvants, which boost APC activation and TAA presentation, promoting tumor-specific anticancer immune responses [32,33]; (b) decoration with genetic therapeutics, predominantly small NA fragments such as small interfering RNA (siRNA), to silence genes that induce immune suppression [34,35]; (c) decoration with antibodies, usually monoclonal antibodies (mAbs) that directly target molecules assisting cancer immune escape [36,37]. Polyinosinic-polycytidylic acid (polyIC) and cytosine-guanine (CpG) motif-containing oligodeoxynucleotides (ODNs) are 2 universal TLR agonists that have been extensively applied as adjuvants with antigens to boost immune responses. Spherical AuNPs with cationic surfaces were alternately layered with an anionic polyIC adjuvant and cationic SIINFEKL peptide antigen through stepwise electrostatic interactions to create polyelectrolyte multilayer (PEM) coatings. Upon intradermal injection, these immune PEMs (iPEMs) were trafficked to tumor-draining LNs and internalized by localized primary DCs, resulting in TLR signaling and antigen presentation. Subsequently, antigen presentation induced the proliferation of antigen-specific cytotoxic T lymphocytes (CTLs) and secretion of more specific effector cytokines, thereby accomplishing efficient vaccination [38]. Therapeutic techniques employing genetic drugs and antibodies share similar strategies, primarily targeting immune checkpoints, such as programmed cell death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1) and cytotoxic T-lymphocyte associated protein 4 (CTLA-4), to limit their exposure by either down-regulating gene expression or direct binding [37,39,40].
Fig. 2.
(A) Au nanocarriers carrying therapeutics for tumor vaccination and immunotherapy. Multilayer antigen and adjuvant self-assembly-coated AuNP as a vaccine to promote antigen-specific T cell response. Reproduced with permission from [38]. An AuNP-encapsulating, ROS and pH dual-responsive carrier were complexed with Cas9-PD-L1 for permanent disruption of the PD-L1 gene in cancer cells to facilitate the anticancer immunotherapy. Reproduced with permission from [263]. (B) MSN encapsulating biomolecules for immunotherapy. IL-13-loaded extra-large-pore MSN directs alternative macrophage activation for autoimmune disease treatment. Reproduced with permission from [42]. Anticancer chemotherapeutic agent and anti-PD-L1 antibody dual-encapsulated MSN for TME modulation and tumor progression inhibition. Reproduced with permission from [264]. (C) IONP loading biomolecules for immunotherapy and imaging. A small-molecule PD-1/PD-L1 inhibitor- and gas-forming NaCO3-loaded IONP with macrophage membrane wrapping showed pH-responsive gas production-induced drug release, which assists M2 reprograming and PD-1/PD-L1 pathway blockade for enhanced anticancer immunotherapy. Reproduced with permission from [265]. A magnetic metal micelle composed of a metal oxide Zn1.15Fe1.85O4/adjuvant core and an antigen-modified shell were fabricated for visualized LN trafficking and showed efficient CD8+ T cell provocation for promoted anticancer immunotherapy. Reproduced with permission from [266].
Silica NPs
The most well-known property of silica NPs, distinguishing them from other INPs, is their mesoporous structure. Mesoporous silica NPs (MSNs) have been designed with various surface decorations for loading and stimuli-responsive drug release, which prevents sensitive drugs from deactivation and digestion and achieves high targeting efficiency [41]. For example, MSNs loaded with IL-13 in their pores showed direct macrophage activation and IL-13 protection [42]. In addition, the mesoporous structure increases the surface area-to-volume ratio, resulting in a higher drug-loading efficiency [43]. Recently, due to the increasing interest in anticancer immunotherapy, MSNs have become potential candidates for trafficking immunotherapeutic payloads to targeted cells and tissues (Fig. 2B).
Similar to AuNPs, MSN-loaded immunotherapeutic cargos are primarily TAAs, genetic therapeutics, and mAb inhibitors [9,44,45]. MSNs demonstrated strong self-adjuvanticity when loaded with TAA for anticancer immunotherapy. A model antigen, ovalbumin (OVA)-decorated MSN, induced only a slightly weaker immune response with an approximately 25-fold lower antigen payload compared with a mixture of OVA and conventional QuilA adjuvant, suggesting that MSN itself is an excellent adjuvant. Intriguingly, the shape of MSNs strongly affects their adjuvanticity, and asymmetric rod-shaped MSN could induce higher expression of CD40 and CD86 maturation markers on APCs than that of spherical MSN, which can be ascribed to that rods were more efficiently internalized by APCs than spherical particles, similar to the findings of rod and spherical AuNPs [46]. Anticancer immune responses can be further enhanced by the co-delivery of antigens and adjuvants, such as OVA and CpG, using MSNs. In addition to the particle morphology, the pore size of MSN has also been reported to stimulate the immune response. MSNs of 80 nm in diameter fabricated for LN targeting were designed with small (7.8 nm, MSNs-S), medium (10.3 nm, MSNs-M), and large pores (12.9 nm, MSNs-L). MSNs with different pore sizes exhibited similar LN-targeting efficiency with significantly different immune activation. MSN-L induced the largest CD4+ and CD8+ T cell populations in LNs, along with the highest suppression of cancer growth as well as the highest survival rate in animals. This might be due to faster degradation of MSN-L and subsequent extensive antigen exposure to APCs located in the targeted LN [47]. Immune checkpoint mAbs and siRNAs account for a large number of antigens and genetic drugs used in immunotherapy. The mAbs aPD-1 and aPD-L1 inhibit immunosuppressive signals during the immune response by blocking the PD-1/PD-L1 axis. siRNA silencing of PD-1 reduced the expression of PD-1 at the source to stimulate immunosuppression induced by PD-1/PD-L1 interaction [48].
Iron oxide NPs
Iron oxide (IO) NPs are superior to other INPs because of their intrinsic superparamagnetic nature. IONPs can achieve guided targeting with predictable efficiency using an external magnet. As excellent magnetic resonance imaging (MRI) contrast agents, they enable high-contrast visualization in vitro and in vivo using MRI [49,50]. Therefore, IONPs have recently gained attention for imaging-guided immunotherapy (Fig. 2C). To this goal, IONPs should be modified with functional molecules, usually polymers with functional groups, to arrest therapeutic agents. The antitumorigenic cytokine interferon-γ (IFN-γ) was adsorbed onto dimercaptosuccinic acid (DMSA)-coated magnetic NPs (MNPs) for anticancer therapy. With external magnet application, IFN-γ-adsorbed MNPs could be delivered more specifically to the tumor site, limiting the off-target-induced cytotoxicity in healthy tissues boosting stronger immunogenicity of cancer cells, and inhibiting cancer angiogenesis [51]. Similarly, IONPs have been used to transport therapeutic proteins and NAs to immune organs and tissues. A multifunctional core–shell NP composed of a superparamagnetic Fe3O4 core and a photonic ZnO shell was designed to deliver carcinoembryonic antigen (CEA) to DCs while functioning as an imaging agent. The Fe3O4-ZnO NPs were internalized by DCs within a short period, and the NP-pulsed DC-immunized mice exhibited enhanced tumor antigen-specific T cell responses, retarded tumor progression, and higher survival rate. In addition, because of high MRI contrast of NPs, DC migration can be tracked during in vivo administration of NP-laden DCs [52]. Although IONPs are excellent materials for drug delivery and MRI imaging, their size should be considered during particle design to maximize T2 relaxivity, drug payload, and cellular uptake. Polyvinylpyrrolidone-coated IONPs (PVP-IOs) with average size of 7.6 to 65.3 nm for IONP cores were fabricated for MRI imaging and macrophage internalization evaluations. In vitro T2-weighted MRI images demonstrated that PVP-IOs with the maximum core size displayed the strongest contrast in aqueous solution compared with smaller PVP-IOs of the same molarity. However, in RAW264.7 macrophage cell lines, PVP-IOs with the medium core size of 36.8 nm exhibited the highest cellular uptake. Interestingly, the strongest MRI contrast of the liver was observed in mice that were intravenously administered PVP-IOs with a core size of 36.8 nm and an overall size of 100 nm [53]. Surface modification of IONP alters cellular internalization as well. IONPs with a core size of 12 to 14 nm and a surface coating of DMSA showed the highest T2 relaxivity and cell labeling efficiency when they were internalized by cells using the centrifugation-mediated internalization method compared with IONPs of larger or smaller core size with the same functionalization or IONPs with same-sized core but surface decoration with other biomolecules, such as dextran, diethylaminoethyl-dextran, carboxymethyl-dextran, and (3-aminopropyl) triethoxysilane [54]. In addition, relatively small IONPs not only enhance MRI imaging and cellular uptake but also provide larger surface for drug loading, which expedites disease treatment progress [55].
Carbon nanomaterials
Carbon nanomaterials (CNMs) are a remarkable type of drug delivery vehicles and have been employed in immunotherapy. Unlike other inorganic nanomaterials such as AuNPs, which lack degradability and cause bioaccumulation, CNM are considered biocompatible after suitable functionalization and is easily degraded or eliminated by the human body [56–58]. CNM can be classified into diverse types according to their structures; carbon nanotubes (CNTs), including single-walled CNT (SWCNT) and multi-walled CNT (MWCNT), have enormous potential for both cancer treatment and theranostics [59,60] (Fig. 3A). Flexible modifiability imparts CNT with the capacity to load various drugs and contrast agents, and the unique structure endows CNT with the inherent property of strong absorption in the near-infrared (NIR) region, making CNT an outstanding material for photothermal therapy (PTT) [61,62]. Similar to the majority of nanomaterials, CNT may enhance the effects of common adjuvants used in cancer immunotherapy, such as CpG. Regardless of the co-delivery of TAA, both MWCNT and SWCNT conjugated with CpG induced more potent immune responses in vivo [63,64]. Co-administration of CpG and TAA via CNT stimulated TAA-specific immune responses. Co-delivery of CpG, OVA, and CD40 significantly retarded the proliferation of OVA-expressing B16F10 cells in melanoma-bearing mouse models [63]. Not only well-dispersed CNT is a suitable drug carrier, but also some interesting studies have shown that bundled SWCNT can improve protein adsorption; T cell-stimulating antibodies or cytokines were absorbed by SWCNT bundles to evoke immune responses against tumors [65,66].
Fig. 3.
(A) Carbon-based nanomaterials delivering biomolecules for tumor vaccination and immunotherapy. CpG, aCD40, and OVA-loaded MWCNT demonstrate significantly improved OVA-specific immune response and efficient eradication of both orthotopic and pseudo-metastatic tumors. Reproduced with permission from [63]. mOVA- and R848-loaded GO-bPEI injectable hydrogel shows sustained release of nanovaccine, efficient antigen-specific DC activation, and significant tumor regression. Reproduced with permission from [70]. OVA-functionalized CD self-adjuvants with the modification of red-, yellow-, and green-colored luminescent for efficient DC maturation and tumor elimination possess potential for bioimaging and biosensing. Reproduced with permission from [71]. (B) CaP NP together loaded with genetic therapeutics for promoted drug release and immunotherapy. A sophisticated nanocarrier composed of CaP, IL-2 pDNA/PD-L1 siRNA, and thymine-capped PAMAM dendrimer was exploited for pH-responsive drug liberation. The complex showed promises in increased infiltration and activation of CD8+ T cells as well as ICB-promoted anticancer immunotherapy. Reproduced with permission from [267]. (C) MnNP with surface functionalization and mAb loading for immunotherapy. Engineered cancer cell membrane-wrapped manganese oxide (CM@Mn) nanoenzyme for TME targeting. CM@Mn induces hypoxia and releases Mn2+ in acidic TME, thereby stimulating ICD. Synergism with aPD-1 triggers strong systemic anticancer immunity suppressing distant tumor progression. Reproduced with permission from [268].
In addition to CNT, graphene oxide (GO) and carbon dots (CDs) are other CNM forms of interest in the field of immunotherapy. GO is advantageous because of its 2-dimensional (2D) structure, and its wide surface area and abundant hydroxyl and carboxyl groups enable easy functionalization via chemical conjugation. The high hydroxyl and carboxyl group content also contributes to the electronegativity of GO, indicating that GO is suitable for physical modification via electrostatic interactions [67,68]. In addition, GO can sequester molecules through π–π stacking [69]. GO has been electrostatically decorated with polyethylenimine (PEI) to arrest functional DNA or RNA as nanovaccine or gene therapy for immunotherapy. Messenger RNA (mRNA) encoding OVA along with the TLR7/8 agonist resiquimod (R848) was encapsulated in an injectable hydrogel formed by electrostatic interactions between low-molecular-weight PEI and GO. The hydrogel became unstable and gradually transformed into NPs upon liquid solution embedding, such as subcutaneous injection, which achieved a sustained release of mRNA- and adjuvant-containing NPs into the surrounding environment. The results validated that NP release was sustainable for 30 days when the hydrogel was administered in vivo, and the released NPs targeted LN and induced an immune response that efficiently inhibited tumor growth [70]. Based on the same strategy, GO was modified with PEI to electrostatically interact with OVA and CpG, and further layered with polyethylene glycol (PEG) to improve biocompatibility. This formulation along with NLG919, a indoleamine-2,3-dioxygenase inhibitor, was used to treat B16-OVA-melanoma tumor-bearing mice and showed synergistic inhibition of tumor growth by activation of immune system and regulation of tumor microenvironment (TME) [19]. CDs are known as the rising star of carbon-based nanomaterials, which have attracted considerable attention in recent years owing to their versatile properties, including ease of synthesis, manipulation, functionalization, and unique optical and electrical characterizations. The size of CDs is highly tunable from 0.5 to 10 nm per different precursors and synthetic approaches, which ultimately impact the inherent fluorescence of CDs. For immunotherapy applications, CDs have been employed as an adjuvant to interact with TAA via electrostatic interactions to form relatively large nanocomposites (CD-OVA) with a size of approximately 50 nm, as measured by transmission electron microscopy. The CD-OVA efficiently stimulated the expression of the costimulatory molecules CD80 and CD86 on DCs and enhanced the secretion of tumor necrosis factor-α (TNF-α). Upon in vivo application, CD-OVA serves as a vaccine that promotes cellular uptake and strengthens OVA process, resulting in potent inhibition of B16-OVA melanoma growth in C57BL/6 mice by inducing antigen-specific immune responses [71]. CDs have also been functionalized with amphiphilic molecules (ACDs) for RNA delivery. ACDs of 100 to 200 nm in hydrodynamic diameter and approximately 50 mV showed promise for gene delivery. ACDs condensed with green fluorescent protein (GFP)-mRNA showed excellent transfection efficiency within 6 h in different types of cell lines in vitro, and those condensed with OVA mRNA demonstrated significant anticancer efficiency and tumor recurrence prevention when intravenously administered to OVA melanoma-bearing mice.
Calcium phosphate NPs
Sharing similar compositions with natural bones and teeth, calcium phosphate (CaP) NPs have the potential to serve as biocompatible and biodegradable vehicles for the delivery of anticancer therapeutics. In particular, because of the inherent pH-dependent solubility of CaP NPs, they are considered promising for accelerated and accumulated release of bioactive drugs in acidic environments such as the TME or more acidic endolysosomes upon cell endocytosis [72] (Fig. 3B). Since not all drugs can target the endolysosomal compartment and some drugs are pH sensitive, CaP NP modification is required to protect them from degradation and promote escape from the endolysosomes to deliver the drug to the active site [73]. In addition to transporting common chemical therapeutics, CaP NPs are also ideal scaffolds for generating cancer nanovaccines. Like many other NPs, CaP NPs also exhibit satisfactory adjuvanticity in vaccination and immunotherapy, owing to their suitable diameter, ranging from 1 to 1,000 nm. Recently, disulfiram (DSF), a conventional drug used to treat alcoholism, was coloaded with cupric ions onto lipid-coated CaP NPs (LCP NPs) for cancer treatment. The LCP NPs degraded after accumulation in the tumor to liberate cupric ions and DSF, and the released cargos formed a cytotoxic metabolic complex that induced immunogenic cell death (ICD) of cancer cells, resulting in switching of the immunosuppressive TME to an immunocompetent state and enhancing systemic immune responses [74]. Another study fabricated manganese-calcium phosphate NPs (Mn/CaRis) and employed them as adjuvants to boost immune responses. The Mn/CaRis showed better performance than that of traditional aluminum adjuvants in augmenting both humoral and cellular immune responses to tumor vaccines, thus improving anticancer immunotherapy outcomes. Moreover, in addition to tumor vaccine, Mn/CaRis exhibited amplification effect on prophylactic vaccines as well [75]. Similarly, adenosine triphosphate (ATP) was functionalized on CaP NP (ACP) as both stabilizer and adjuvant and then applied to OVA to manufacture nanovaccine for cancer treatment. This formulation showed efficient DC activation and T cell priming in cellular test as well as tumor growth inhibition when administered to tumor-bearing mice [76].
Manganese NPs
Manganese NPs (MnNPs), specifically manganese oxide (MnO2) NPs, have emerged in the field of cancer immunotherapy in the last decade. The greatest strength of MnO2 NPs is that they can have various oxidation states with coordination numbers up to 7.5, making them strong oxidants capable of mediating several oxidation reactions [77]. Owing to this fundamental oxidative property, MnO2 NPs have been shown to trigger the conversion of endogenous hydrogen peroxide (H2O2) into reactive oxygen species (ROS) under acidic conditions such as the TME [78] (Fig. 3C). Moreover, several oxidation states of MnO2 NPs enable easy and thorough functionalization of these particles for specific purposes [79–81]. In addition to their flexible reactivity, MnO2- and Mn-doped NPs demonstrated high biocompatibility. In vivo administration of Mn-doped MnFe2O4 NPs showed no irreversible damage to major organs and were eliminated from the kidneys, spleen, and brain of mice on day 7 after injection [82]. Being a superior nanocarrier, MnNPs have been applied for drug delivery in diverse single and combined tumor therapies, including immunotherapy. Moreover, Mn NPs itself have an effect on immune activation. Mn NPs could serve as an oxygen supplier to reduce the expression of the pivotal immune checkpoint PD-L1 in cancer cells by overcoming the hypoxic environment at the tumor site, contributing to the reprogramming of the immunosuppressive TME [4]. Mn NPs were also validated to comprehensively stimulate the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway, a vital signaling pathway related to the activation of the innate immune response combating tumors through the secretion of IFN-I and pro-inflammatory cytokines and by releasing Mn2+ ions in the cytoplasm [83]. Similar to AuNPs, Mn NPs can also be employed as adjuvants and have displayed significantly stronger adjuvanticity than that of conventional alum adjuvants for the treatment of live coronavirus and pseudovirus infections in vitro [84]. In addition, because of their strong absorption coefficient and relaxivity, Mn NPs and Mn-doped NPs are suitable contrast agents for both CT and MRI, indicating that these NPs can be used for diagnosis and guided therapy [85,86].
Targeting
Immunotherapy targets vary greatly from biomolecules to immune organs, and each target has its own merits and drawbacks. In addition, the dosage should also be carefully considered, as a low dose is therapeutically ineffective, whereas a high dose causes severe side effects by systemic immune homeostasis imbalance, which is sometimes fatal.
Biomacromolecules
PD-1/PD-L1 axis
Programmed cell death protein-1 (PD-1) and its ligands PD-L1 and PD-L2 are the most common representative axes of immune checkpoints and have been used as immunotherapy targets for decades. PD-1 is mainly expressed in immune cells, including activated tumor-specific T cells, B cells, DCs, macrophages, natural killer cells, and monocytes, upon chronic antigen exposure [87–89]. PD-L1 is mainly expressed in cancer cells and some APCs and acts as a protumorigenic factor in cancer cells [90]. One major cause of tumor immune escape is the immunosuppressive TME, mainly attributed to the crosstalk between PD-1 and PD-1 ligands through inhibition of the activation, proliferation, and cytotoxic function of CTL [91]. As such, immune checkpoint blockade (ICB) therapy using immune checkpoint inhibitors (ICIs) to block the PD-1 and PD-L1 axes and interrupt their interaction for cancer immunotherapy has been developed as a new milestone, and some clinical trials have achieved promising results [1,92,93]. However, owing to individual differences, high off-target rates, elevated PD-1/PD-L1 inhibitor resistance, and immune-related adverse events (irAEs) caused by the disruption of immune homeostasis upon long-term medication, the universal application of PD-1/PD-L1 blockade drugs is far to go [94–101]. One method to ameliorate these undesired side effects is to introduce nanocarriers to encapsulate the inhibitors. Related research results have proven that the targeting efficiency and immune response can be significantly enhanced by nanocarrier delivery systems, along with reduced off-target rates and side effects [102–106]. For instance, an ingenious AuNP platform with amphiphilic organic ligand protection was synthesized to carry a large payload with less off-targeting uptake, which was demonstrated 40-fold higher cell uptake in the targeted CD8+ T cells compared to nontargeted cells [105]. Another example proposed a remotely controlled upconversion NP-based immunodevice that allowed and constrained the generation of effective immune response only within the tumor site without disturbing systemic immunity [106]. Inspired by the intelligent nanotherapeutic devices, nanocarriers have been used to load mAb therapeutics that can directly block the immune checkpoints. An MSNP was employed as a multifunctional platform that co-delivered a PLK1 inhibitor (volasertib) and an anti-PD-L1 antibody for synergistic anticancer therapy by targeting both PLK1 kinase and PD-L1 immune checkpoint. The inhibition of PLK1 kinase selectively kills cancer cells by affecting their mitosis, leading to the up-regulation of PD-L1 expression in surviving cancer cells for a more effective PD-L1 antibody attack [107]. As a result of synergy, the effective doses of both volasertib and PD-L1 antibody were significantly reduced. In addition to facilitating ICB therapy toward the patients that efficiently respond to PD-L1 ICIs, the multifunctional MSNP vehicle might also be potential in promoting the response of patients that possess insufficient PD-L1 to PD-L1 antibodies by up-regulating the PD-L1 expression level. Other metal-based NP or microparticles, such as IONPs, metal–organic frameworks (MOFs), and AuNPs, are also prevalent nanocarrier choices for delivering antibodies, RNAs, or both for PD-1/PD-L1 axis targeting [108–114]. In addition to directly targeting the PD-1/PD-L1 axis, TLR agonists can also be loaded together to amplify the anticancer immunity by eliciting both innate and adaptive immune responses and regulating the TME [16]. Some nanocarriers with specific optical or magnetic properties are capable of multitasking, such as immunotherapeutic drug delivery and imaging or dual therapy involving immunotherapy and PTT [14,115].
Cytotoxic T lymphocyte-associated protein 4
Cytotoxic T lymphocyte-associated protein 4 (CTLA-4) is another important immune checkpoint that has recently emerged as a target for cancer immunotherapy. It is a glycoprotein homologous to the CD28 costimulatory molecule and belongs to the immunoglobulin superfamily; it is expressed primarily by T cells and translocated to the cell surface for T cell receptor engagement upon T cell activation [8,116]. As a pivotal biomolecule that regulates T cell homeostasis and self-tolerance, CTLA-4 competes with CD28 to bind to the B7 family molecules, particularly CD80 and CD86 expressed on APCs, resulting in T cell anergy and a compromised anticancer immune response [117,118]. Since CTLA-4 has a superior binding affinity toward B7 compared with CD28, blocking CTLA-4 is considered a promising approach to restore T cell activity [119,120]. Nonetheless, ICI-mediated tumor therapy targeting CTLA-4 is infrequently applied in clinical setting because it shows delayed effects and is sometimes associated with severe side effects, including hepatitis, colitis, and thyroiditis, owing to overactive immune responses [94,121]. Similar to therapeutic agents targeting PD-1 and PD-L1, their effects on CTLA-4 have been developed as nanotherapeutics by conjugating the effective drugs to nanocarriers, particularly those with thermal conversion properties, because hyperthermia-mediated tumor ablation induces immunogenic cell death [26]. SWCNT showed a good capacity for photothermal conversion and were therefore modified with an anti-CTLA-4 antibody (aCTLA-4) for the combined PTT and ICB therapy. In addition to direct thermally induced tumor elimination, tumor cell death leads to elevated TAA release, resulting in the maturation and activation of DCs. In this case, conjugating aCTLA-4 further facilitated CD4+ and CD8+ T cell activation and infiltration at the tumor sites as well as decreased T regulatory cell (Treg) activity [122]. IONPs are also used for hyperthermia therapy because of its magnetocaloric properties [123]. By integrating localized magnetic hyperthermia (MHT) therapy with systemic ICB therapy using aCTLA-4, a more comprehensive anticancer effect could be achieved, with not only tumor regression but also robust inhibition of tumor metastasis and recurrence [124].
However, as previously indicated, the use of ICB treatment is currently limited due to its significant toxicity caused by the disruption of immune homeostasis. Furthermore, owing to inherent variability across individuals, the dosages administered to different patients exhibited significant variation. Hence, the development of a widely standardized ICB therapy and controlled ICI release approach has immense value and significance.
Cells
Targeting tumor-associated cells is an attractive option for effective immunotherapy. These cells are typically closely linked to the immune system, such as APCs and T cells, or are strongly connected to cancer cell or microenvironment, such as cancer-associated fibroblast (CAF).
Dendritic cells
DCs are prominent targets for immunotherapy owing to their crucial role as APCs in initiating a strong immune response mediated by CTL specific to tumor antigens. Briefly, DCs acquire and process tumor-specific antigens. They present the resultant peptide, together with major histocompatibility complex class I (MHC-I) or class II (MHC-II), on the surface of CD4+ or CD8+ T cells. Furthermore, mature and activated DCs not only display MHC–antigen complex but also exhibit increased expression of CD80 and CD86. This enhanced expression allows effective communication with T cells, facilitating the transmission of costimulatory signals [25,125,126]. In addition, T cell differentiation requires the presence of certain cytokines, including IL-2, to guide the process. Collectively, these processes stimulate specific immune responses against cancer [127]. Conventional DC-based immunotherapy involves the isolation of DCs from individuals, their maturation and activation in vitro using tumor antigens, and reintroduction into patients through transfusion [128,129]. However, these processes are laborious and challenging to implement. The groundbreaking DC-mediated immunotherapy, specifically DC vaccine, involves the presentation of tumor-specific antigens onto nanodevices, which are then directly administered in vivo to initiate a tumor-specific immune response. In some cases, adjuvants are also loaded to augment the immune response. The strategy of loading TAAs and/or adjuvants, usually CpG ODN, into INPs targeting and inducing DC activation for cancer immunotherapy has been extensively discussed in the previous section. Therefore, there is no need to further explore this topic in this context [5,130]. Silencing YTHDF1 protein in DCs is another efficient method, besides CpG stimulation, to increase the expression levels of CD80 and CD86 [12,131]. A nanovaccine platform with AuNP core and dendrimer coating was modified with mannose for DC targeting and compressed with siYTHDF1 to elevate the expression of costimulatory factors. Co-administration of a PD-L1 antibody boosted anticancer immune responses in an animal model with a melanoma tumor xenograft [132].
T cells
DCs can be considered as intermediate targets for immunotherapy because they are responsible for presenting TAAs to T cells, thereby triggering a specific immune response against tumors. T cells, specifically CTLs, are direct targets because they directly combat cancer cells. A common method for targeting and activating T cells involves the use of NPs that imitate DCs [132–134]. In this particular instance, NPs are typically disguised with a coating made from cancer cell-primed DC membrane. This coating layer contains not only all DC membrane protein information but also processes that are prepared for presentation [135]. In contrast to DCs, DC-mimic NPs are smaller in size. This allows them to crosstalk more precisely and abundantly with T cells, leading to more effective activation and stronger immune responses [136]. In addition, other functional biomolecules including cytokines and NAs, such as IL-2 or siCTLA-4, can be used together for modification of DC membrane on NPs, to promote CTL proliferation or decrease CTL exhaustion. Although organic NPs, particularly poly(lactic-co-glycolic acid)-based NPs, account for the majority of NP cores camouflaged with DC membranes, some INPs, including AuNPs and MSNP, have also been extensively used as platforms for DC membrane packaging for CTL targeting [137].
Another prevalent T cell target is Treg, an important T cell subtype, which plays a critical role in maintaining immune homeostasis, preventing autoimmunity, and suppressing allergy. Unlike CTL, which surface-expresses CTLA-4 only at the activation state, Treg consistently expresses CTLA-4 to regulate excessive overreaction of the immune system. However, in patients with cancer, the inherent property of Treg hinders effective immune surveillance against tumors by establishing an immunosuppressive TME together with other cells such as myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs), and CAF. Therefore, targeting Treg to switch the TME to immunocompetent state is of great significance. Nanodevices carrying siCTLA-4 or aCTLA-4 for ICB therapy have been widely employed and are not discussed in detail in this section [8,138].
Chimeric antigen receptor (CAR)-T cell therapy is another ground-breaking pillar of T cell-mediated immunotherapy. T cells are genetically modified with CARs that specifically target malignant tumors, particularly B cell malignancy, under the prevailing conditions. Conventional CAR-T cell therapy consists of 3 main steps: T cell separation, laboratory CAR engineering, and CAR-T cell refusion. The entire technique closely resembles conventional DC-based immunotherapy, including intricate manufacturing procedures, and carries a significant risk, hampering its widespread adoption in clinical practice [139]. Thus, similar to the activation of DCs in living organisms by utilizing NPs, genetic modification of T cells to express CARs can be accomplished in vivo through NP delivery. This approach represents a potential strategy for creating universally applicable targets for immunotherapy [140]. Despite the use of biomolecule-based nanovesicles, such as liposomes and exosomes, and polymer-based complexes, inorganic nanomaterials such as quantum dots, CNTs, AuNPs, and MSNPs have also shown significant potential for in vivo gene editing of T cells and have been experimented [141].
Macrophages
Macrophages are predominant components of the TME and are involved in tumor development. They can be polarized into 2 basic phenotypes under different stimulations: M1-type macrophage (M1) and M2-type macrophage (M2), which are also referred to as TAM [142,143]. Concisely, M1 can be stimulated by IFN-γ, lipopolysaccharide (LPS), TNF-α, or GM-CSF, which then triggers the activation of TLR signaling pathways [144]. They have beneficial effects on the elimination of infections and malignant cells. M1 macrophages exert their antitumor activity in several ways. They express high levels of antigen-presenting MHC complexes to activate the adaptive immune response to combat tumors [145,146]. They act directly on cancer cells by generating nitric oxide (NO), ROS, and reactive nitrogen species (RNS). Moreover, they promote inflammatory responses by secreting pro-inflammatory cytokines [13,147–149]. In contrast, M2 macrophages are activated by several cytokines. Accompanied by the increased production of polyamines and ornithine through the arginase pathway, they participate in parasite clearance and homeostasis, including tissue remodeling and regeneration, wound healing, and anti-inflammation [150,151]. Two approaches have been developed to regulate M2 macrophages in cancer immunotherapy. One approach involves depleting M2 macrophages to transform the TME into an M1-biased antitumor environment that can prevent tumor growth [152]. Another approach involves exploiting the plasticity of differentiated macrophages to convert M2 into M1 under specific circumstances [153]. Consequently, M2 macrophages can be repolarized to M1 macrophages and prevent tumors. Both approaches were accomplished using nanomodulators, employing either organic or inorganic NPs as platforms [154–156].
Cancer-associated fibroblasts
CAFs are another dominant and abundant cell type responsible for constructing and modifying the extracellular matrix (ECM) framework within the TME. Nevertheless, because of the high heterogeneity and deficiency of specific markers, identification of CAF remained ambiguous for long period. Fortunately, owing to recent technological advancements, such as single-cell RNA sequencing, CAF can now be divided into 2 main subtypes: myofibroblastic CAF (myCAF) and inflammatory CAF (iCAF) [157,158]. Although the exact role of CAF subtypes in tumor progression is not yet fully understood, several studies have demonstrated that myCAFs can induce the expression of PD-1 and CTLA-4 in Treg through a positive feedback loop, suggesting that myCAFs hold potential as viable targets for cancer immunotherapy [159,160]. CAFs exhibit higher metabolic activity than that of regular fibroblasts. Although fibroblasts are typically considered inert in normal tissues and can be activated to assist in wound healing and tissue regeneration, CAFs are more metabolically active. They induce the production of various growth factors and pro-inflammatory cytokines to promote angiogenesis and recruit immunosuppressive cells to establish a TME capable of assisting tumor immune evasion [161,162]. As a burgeoning target, CAFs have been tested using various strategies, including ICB therapy by reducing the exposure of PD-1 or CTLA-4, blockade of the critical surface marker CXCL12 and the corresponding receptor CXCR4, depletion of fibroblast activation protein (FAP) accompanied by anticancer vaccination, normalization of CAF to reduce the deposition of ECM that impedes CTL infiltration, inhibition of NOX4 to prevent the activation of CAF, and even inactivation of activated CAF [163,164]. These strategies can be integrated with nanocarriers to enhance their therapeutic effects.
Sites
Tumor-draining lymph node
As an immune cell-resident organ, LN is another eligible drug target in immunotherapy. LN-targeted delivery can be briefly categorized into 2 major types: direct intranodal injection and lymphatic drainage followed by other methods involving systemic, subcutaneous, intradermal, intramuscular, and intraperitoneal injections [6,165–167]. Intranodal delivery, as the name indicates, involves direct injection of drugs or platforms with drug payloads into the LN. The advantage of this approach is that it minimizes off-targeting and concentrates the therapeutics to the LN, which reinforces the exposure of the resident immune cells to the drug and contributes to enhanced interaction between them, and finally resulting in enhanced immune responses [11,168]. For instance, intranodal injection of tumor-specific antigens and adjuvants has been shown to be effective in enhancing anticancer immunity by enhancing their contact with APC and their subsequent processing and presentation [169]. In addition, tumor lysates are alternative agonists for priming tumor-specific immune responses via intranodal injection [170]. Immune cells can also be administered intranodally. In vivo reinfusion of antigen-pulsed DC is a typical immunotherapy approach that has exhibited significant anticancer effects; however, direct intranodal injection showed stronger T cell response induction than that of intravenous DC refusion [171,172]. In addition to activating the immune system, modulation of immune cells is another alternative for enhancing the immune response. Some cytokines can be directly injected into LNs to control the phenotype or function of immune cells. For example, IL-2 is essential for T cell proliferation and has therefore been delivered together with tumor antigens to strengthen adaptive immunity [173]. However, compared with other administration approaches, intranodal delivery is more invasive and less accessible [174]. Moreover, some therapeutics injected intranodally can be rapidly eliminated from LNs, ultimately leading to poor immunological modulation [165].
LN-targeted delivery of injected immunomodulators by direct afferent lymphatic drainage is highly dependent on their physicochemical properties, particularly their size. Although small-sized immunomodulatory agents can be easily drained into the LN, they are also rapidly cleared, and their poor retention time is insufficient to pulse anticancer responses [175–177]. In this case, larger NPs with immunomodulatory functionalization are a better choice. The optimum size of NPs for lymphatic drainage as well as LN retention is a diameter of 10 to 200 nm, and NPs of smaller or larger size exhibit poor LN retention or poor lymphatic drainage, respectively [10,175]. In addition to size, the hydrophobicity and surface charge of NPs also influence their LN-targeting capacity. Hydrophilic NPs with anionic or neutral surfaces are more likely to be drained [178,179]. To further facilitate immunological activation after lymphatic drainage, NPs can be modified with biomolecules, typically antibodies against specific immune cell surface markers, to enhance their interactions with immune cells. NPs modified with anti-CD11c showed enhanced interaction with DC, and those decorated with anti-CD8 facilitated CTL binding [10].
Tumor microenvironment
The TME and its role in the development and advancement of various cancers has been acknowledged over the last decade. The TME is a complex system consisting of cells derived from mesenchymal, endothelial, and hematopoietic lineages organized within the ECM. These cells interact closely with cancer cells and play a significant role in tumor development [180,181]. The interaction between the tumor and TME plays a crucial role in influencing the advancement of cancer, by either promoting or inhibiting its progression [182,183]. Although the TME initially has anti-malignant effects in early-stage tumors, many cancer cells can withstand this suppression and subsequently alter the TME to promote the malignant behavior [182,184]. The TME is complicated and diverse during the last stage of solid tumors. First, rapid proliferation of cancer cells induces several irreversible events, which cause metabolic remodeling of cancer cells and an adaptation of the TME to the new setting, and the TME infrastructure is influenced by various immune and nonimmune cell types. The complex TME induces disorganized biological processes, including secretion of various cytokines and chemokines, alteration of metabolites, hypoxia, angiogenesis, ECM remodeling, interstitial pressure, and pH changes [185–188]. These factors contribute to the development of a chronic inflammatory, pro-angiogenic, and immunosuppressive environment in cancer [189,190]. Over the last decade, the TME has been recognized as a favorable setting for the development of new and effective anticancer drugs. The basic TME targets can be categorized into 2 types. One targets TME cells that exert immunosuppressive effect, including tumor-infiltrating Treg, M2 macrophages, MDSC, CAF, professional APCs such as DCs, and tumor-combating effector cells, whose functions are inhibited by the immunosuppressive TME, involving CTL [191]. The other factor is the environment itself, including ECM, hypoxia, and acidic pH [192]. TME delivery approaches can be divided into 2 types: systemic and intratumoral. Systemic delivery is realized by intravenous injection, and efficient delivery can be achieved by active targeting that employs tumor-targeting molecules to anchor tumor sites or passive tumor accumulation is mediated by particle size and surface charge [193–195]. Intratumoral delivery is accomplished by direct injection of therapeutic substances inside the tumor. Compared to systemic delivery, intratumoral delivery has always shown better therapeutic effects because the proportion of injected drug for utilization is significantly elevated. However, as not all tumors are physically accessible in the absence of surgery, systemic delivery approach is universally applicable [196–198].
Combination Therapy
Although immunotherapy has demonstrated advantages in pulsing adaptive immune responses and preventing tumor recurrence, it is not universal for all types of tumors. In addition, the dose differs significantly depending on the individual, and an overdose can cause severe adverse effects [199,200]. To maximize the anticancer effect with reduced adverse effects, higher survival rate, and prolonged survival, combination therapy is occasionally preferred over single immunotherapy. Current therapeutic methods used in combination with immunotherapy include chemotherapy, PTT, photodynamic therapy (PDT), and radiation therapy [201–204]. The common combination therapy and their applications were summarized and listed as Table 2.
Table 2.
Combined therapeutic strategy | Carrier | Functionalization | Application | Reference |
---|---|---|---|---|
Chemotherapy | IONP | EBP, polyIC, DOX, | Enhanced tumor targeting and combined immunotherapy and chemotherapy | [292] |
CD | AEAA, DOX, Fe ion, LOS, and Asp-Ala-Thr-Gly-Pro-Ala peptide crosslinker | Combined immunotherapy with chemotherapy | [293] | |
CD | FA, Cu, and aPD-L1 | Combined ferroptosis and immunotherapy for tumor eradication | [294] | |
MnNP | DOX and phospholipid | Induction of immune cell maturation, activation, and up-regulation of cytokine release for enhanced combined immunotherapy and chemotherapy | [269] | |
PTT | AuNP | TAA and DC biomolecules | Stepwise intra-cancer cell and DC-generated AuNP for combined immunotherapy and PTT | [270] |
AuNP | CpG | Fabricate of hydrogel using complementary DNA chains for combined immunotherapy and PTT | [295] | |
IONP | MIQ and ICG | Combined immunotherapy with interventional PTT for PC treatment, together with MRI guidance and temperature monitoring | [296] | |
IONP | Being coated on platelets together with aPD-L1 | Combined immunotherapy with PTT for postsurgical prevention of tumor reoccurrence | [297] | |
IONP | PDA, PEG, RGD, anisamide, and R848 | Combined immunotherapy with PTT for anticancer therapy and MRI for tumor diagnosis | [298] | |
GO | PEI, PEG, and CpG | Combined immunotherapy with PTT | [299] | |
GO | MTX and SB-431542 | Combined immunotherapy with chemotherapy and PTT | [300] | |
CD | aCTLA-4 | Combined immunotherapy with PTT | [122] | |
CD | Al, mannose and CpG, and formed hydrogel with oxidized dextran | Combined immunotherapy with PTT for tumor eradication | [301] | |
MnNP | Being coated with mPEG-b-PHEP and IR780 dye | Combined immunotherapy with PTT | [302] | |
PDT | MSN | MMP-2, aPD-1, Ce6, and PTX | Facilitate tumor accumulation, cellular uptake, immune checkpoint blockade, and combine immunotherapy with chemotherapy and PDT | [303] |
Gas therapy | IONP | l-Arg and PAA | Reprogram M2 TAMs toward M1 TAMs, release proinflammatory cytokines, and recruit T cells, and combined gas therapy with immunotherapy for anticancer treatment | [304] |
Chemo-immunotherapy
Chemo-immunotherapy involves a combination of traditional chemotherapy and emerging immunotherapy. Although some chemotherapeutics can trigger dose-dependent immunosuppression, the combination of chemotherapy and immunotherapy in some cases may maximize the therapeutic efficacy through potential synergistic effect [23,24] (Fig. 4A). One major chemotherapy actively combined with immunotherapy is cytotoxic, because it induces ICD, which can boost the adaptive immune response [205]. The underlying mechanism is that the ICD of cancer cells leads to the release and relocation of damage-associated molecular patterns (DAMPs), which act as strong adjuvants to pulse immune cells and potentiate immunotherapy [206,207]. Typical ICD-associated DAMP progression includes cell surface expression of calreticulin (CRT), ATP discharge, and post-apoptotic release of HMGB1. Exposure to CRT leads to elevated endothelial expression of adhesion molecules, resulting in promotion of lymphocyte infiltration into the tumor tissue and enhanced cancer immunotherapy. Released ATP serves as a “find-me” signal to attract APCs and recruit DCs to migrate to LNs. In contrast, released HMGB1 interacts with TLR4 in DCs to decelerate the degradation of phagocytic antigens and augment their presentation [208–211]. In addition to the above DAMPs, the release of annexin A1, heat shock proteins 70 and 90 (HSP70 and HSP90), TLR3, TLR9, and other cytokines and chemokines also play roles in maturation and recruitment of APCs and T cells [212–214]. ICD is capable of enhancing adjuvanticity, and several studies have implied that it enables stronger antigenicity in cancer cells. Some chemotherapeutic drugs significantly up-regulate antigen expression. For example, gemcitabine and topotecan can induce elevated expression of human leukocyte antigen (HLA) through activation of the nuclear factor κB (NF-κB)/IFN-β/MHC-I signaling axis, which leads to amplification of tumor-specific immune response [215]. In addition to administering chemotherapeutic agents that are capable of inducing enhanced ICD to provoke immune cells, employing functionalized NPs for promoted and precise targeting as well as localized stimuli-responsive drug release also helps with specific immune response and reduced systemic toxicity. Although organic NPs are more prevalent carrier choices for chemo-immunotherapy, INPs still have a place in this field due to their unique properties. For instance, some INPs serve as more than carriers, and they can also be stimuli-susceptible drug precursors; some INP carriers not only deliver drugs but also impart imaging-guided functions [216–220]. An iron-carbonyl complex was designed for chemodynamic immunotherapy since it could be initiated by both glutathione and H2O2 to generate CO and ferrous ions, which could induce mitochondria damage as well as oxidative stress, resulting in ICD-mediated immune response [217]. Another example showed that prodrugs loaded on nanogapped AuNPs sensitive to pH and glutathione could work together with photoacoustic imaging to help with chemo-immunotherapy and imaging of the deep tumor area in the NIR-II window [216]. In some situations, elaborated INP platforms could be better choices for multi-tasking purposes with more concise fabrication steps in comparison to some organic NPs, making them promising candidates in combined immunotherapy.
Fig. 4.
(A) Immunotherapy combined with chemotherapy for tumor treatment. Manganese-based nanoactivator loaded with doxorubicin (DOX) and phospholipid shows prolonged systemic circulation and induces ICD via DOX as well as sensitize cGAS-STING pathway by released Mn2+ in cancer cells, thus boosting both innate and adaptive anticancer immune responses and eliminating tumor by combined chemotherapy and immunotherapy. Reproduced with permission from [269]. (B) Immunotherapy combined with phototherapy for tumor treatment. Immunological AuNPs generated stepwise in cancer cells and DCs presenting tumor antigens for combined PTT and immunotherapy, which exhibit enhanced biosafety, rapid hyperthermia induction, and significant evocation of tumor-specific immune response. Reproduced with permission from [270]. Manufacturing of visible light-triggered prodrug NPs with the modification of DOX and PS to enhance ICD and immune response by synergistic chemotherapy and PDT at the tumor site, and combining them with PD-L1 checkpoint blockade-mediated immunotherapy for tumor eradication and cancer recurrence prevention. Reproduced with permission from [271].
Radio-immunotherapy
Radiotherapy is another therapeutic approach that enhances the adjuvanticity and antigenicity of cancer cells to boost tumor-specific immunity. Therefore, they are suitable for combination with immunotherapy [221]. Similar to cytotoxic chemotherapy, radiotherapy can also induce ICD, followed by the surface translocation of CRT and release of annexin A1, HSP70, HSP90, HMGB1, and other molecules, resulting in enhanced recruitment of APCs and T cells [222–224]. Moreover, radiation down-regulates the expression of the macrophage “Do not eat me” signal, CD47, which collaborates with the elevated “eat me” signal CRT, to further enhance the tumor-specific antigen presentation [225]. In addition to ICD, radiation enhances the presentation of tumor antigens by up-regulating the expression of MHC-I and induces the damage and release of DNA to up-regulate the expression of the type I interferon pathway and boost immune responses via the cGAS/STING pathway [226,227]. In addition to directly damaging DNA, radiation induces ROS generation, which oxidizes DNA and proteins, and increases their antigenicity [228]. However, as radiation indiscriminately attacks cancer and normal cells, it also causes the death of immune cells that infiltrate the TME [229]. It also promotes the infiltration and aggregation of immunosuppressive MDSC and Treg and simultaneously reduces the penetration of CTL into the TME. Together, these circumstances lead to the generation of a more immunosuppressive TME, weakening the effect of immunotherapy and assisting tumor progression [230,231]. Despite being challenging, combining radiotherapy with immunotherapy is prospective to minimize undesirable side effects, and using INPs for image-guided drug delivery could further reduce the toxicity toward normal tissues [3,18,232–234]. Based on this concept, a radiation-responsive snowflake-like Au nanocarriers (S-AuNC) were loaded with aPD-L1 for synergistic radio-immunotherapy. External radiation not only induced ICD for immune evocation but also mediated S-AuNC deformation and aPD-L1 liberation for ICB-mediated immunotherapy. In addition, Au could be a splendid contrast agent for CT imaging, which achieved image-guided radiation, resulting in minimized systemic immune-related adverse effects [233]. Likewise, other INPs such as activation and guiding of irradiation by x-ray (AGuIX) NP were used as radiosensitizers and were together administered with aPD-L1 for synergistic therapy. Compared to conventional radiotherapy, AGuIX NP/aPD-L1-mediated radio-immunotherapy significantly increased the infiltration of effector CD8+ T cells and effectively alleviated the immunosuppressive TME [18]. Therefore, NP-mediated combined radio-immunotherapy showed superiority over either radiotherapy or immunotherapy, because its imaging-guided property could realize controlled treatment to maximize the outcome while minimizing the adverse effect.
Photo-immunotherapy
Phototherapy, which principally involves 2 typical categories, PTT and PDT, is an important cancer treatment modality. PTT and PDT offer significant benefits such as noninvasiveness, low systemic toxicity, and precise tumor targeting [235–237].
PTT entails injecting materials with a high photothermal conversion efficiency (PCE) into the human body, followed by tumor tissue accumulation via passive or active targeting. When materials are exposed to external light sources, such as NIR laser, the light energy is converted into heat, producing local hyperthermia and inducing cancer cell necrosis or apoptosis, leading to tumor elimination [238,239]. One of the most critical factors in inducing significant PTT is the selection of photothermal transduction agent (PTA) with efficient PCE. PTT encompasses different categories of converters, including inorganic photothermal converters such as Au nanomaterials and organic photothermal converters such as polydopamine NPs [240,241]. Compared with inorganic Au nanomaterials, which exhibits a high PCE of NIR, resulting in damage of normal tissues in the tumor surroundings, organic polydopamine NPs exhibit excellent compatibility with living organisms and are an optimal choice for PTT [242]. PTT not only directly ablates tumors but also contributes to ICD, which induces the release of DAMPs and TAAs from dying cancer cells to enhance the immune response. In addition, elevated temperature at the tumor site expedite blood circulation, which promotes the accumulation of systemically administered small molecules such as immunotherapeutic agents inside tumors [26]. Therefore, the combination of PTT and immunotherapy synergistically enhances primary tumor eradication and distant metastasis (Fig. 4B).
In contrast, the PDT regime is highly oxygen dependent. This includes the application of photodynamic molecules known as photosensitizer (PS), followed by the induction of singlet oxygen (1O2) generation by reacting with O2 or ROS via direct oxidation of biomolecules under photo-irradiation [243,244]. However, most conventional PS agents, such as chlorine or phthalocyanine, show poor solubility in water, limiting their in vivo application. To address this deficiency, nanocarriers are typically introduced to load PS agents for targeted trafficking with efficient drug delivery. In this case, PS agents are usually encapsulated in the core area via hydrophobic interaction or shielded by a hydrophilic layer in the intermediate phase of solid NPs [245–247]. The modified NPs were multitasked with cancer elimination through synergistic PDT and immunotherapy by cofunctionalizing with immunotherapeutic agents such as antigens, antibodies, adjuvants, and NAs (Fig. 4B).
Summary of Current Processes in Cancer Immunotherapy and Perspectives
INPs have been widely employed in conventional chemotherapy and phototherapy because of their biocompatibility, high drug-loading capacity, enhanced targeting efficiency, and unique physicochemical characteristics. However, unlike organic NPs, most of which are designed to degrade spontaneously, many INPs cannot be easily eliminated in vivo, making them arduous for clinical translation due to the potential toxicity. Nevertheless, clinically administering INP-based therapeutic agents is not an impossible gulf. There have been well-designed biodegradable INPs, such as MnO2, iron-carbonyl complex, calcium ore NPs, and so forth, as prodrugs or drug carriers for in vivo application. In addition, stimuli-triggered deformable INPs, partially degradable organic/inorganic hybrid NPs, and INPs with a diameter smaller than 5 nm can be easily renal cleared and therefore are promising candidate for clinical translation [4,18,83,84,217,233]. In fact, there have been a few approved INP drugs used for clinical treatment, and more have been successfully progressed to clinical trials [248].
With development in cancer immunotherapy and discovery of self-adjuvanticity of a large number of INPs, the application of INPs as carriers for immunotherapeutic agents, such as mAbs and genetic drugs that block immune checkpoints, has emerged in the last decade [249]. Immunotherapeutic agents regulate immunosuppressive signaling to switch the “cold” (immunosuppressive) TME to the “hot” (immunocompetent) state, ultimately provoking systemic anticancer immune signals. Inorganic hafnium oxide NP-based therapeutics (NBTXR3/Hensify) have been included in many clinical trials, and one of them has been approved for radiotherapy (CE Mark, 2019). Because this platform also triggered enhanced antitumor immune response, the NP has potential for immunotherapy and combined radio-immunotherapy, and its clinical trial for immunotherapy is also under progress (NCT03589339) [7,250,251]. However, several studies have shown that cancer immunotherapy is highly individually biased; some patients respond strongly to immunotherapeutic agents, whereas others have a weak or even no response to the same formulation [94,252]. Therefore, to achieve potent tumor elimination while preventing tumor metastasis and recurrence, combined therapies integrating immunotherapy with other anticancer therapeutic modalities have burgeoned [253–255]. In recent years, advanced clinical therapeutics have been explored for compatibility with immunotherapy for synergistic therapeutic effect, such as the combination of cryosurgery with immunotherapy, which provides a much safer technology and achieves complete tumor elimination, including operative cancers and distal and metastatic cancers of the same type [135,256,257]. In addition to combining surgery and co-delivering immunotherapeutic agents with drugs having specific anticancer effects on the same substance, anticancer outcomes can also be improved by proper nanocarrier selection. Nanocarriers with inherent characteristics, such as the ability to generate heat and produce ROS and singlet oxygen under irradiation, or properties of guided tumor targeting in the presence of an external force, are promising candidates to exert synergistic anticancer effect with modified immunotherapeutic agents [258,259]. Both Au and mesoporous silica nanocarriers possess inherent self-adjuvanticity and have been widely used for antigen delivery [12,260]. Au, IO, and carbon nanocarriers have demonstrated substantial PCE, enabling their use for integrating immunotherapy with PTT [261]. Likewise, Mn NPs can function as O2 suppliers and therefore can be used as carriers to enable hyperoxia-enhanced immunotherapy [4].
In addition to conventional immunotherapeutics, cell membranes, such as tumor and immune cell membranes presenting TAAs, have become novel stimuli that evoke specific immunity. Cell membrane-camouflaged nanocarriers contain complete TAA information, facilitating the induction of tumor-specific immunity; in addition, subcutaneous injection of small-sized nanocarriers enables more precise LN targeting and subsequent immune cell priming. Cell membrane-coated nanocarriers can accumulate in tumor tissues via ligand-mediated active targeting and size-dependent passive targeting when administered intravenously and then modulate the TME to stimulate the anticancer immune response. Additionally, augmenting targeting efficiency and reducing off-target effects can be achieved through the modification of nanocarriers with cell membranes engineered with synthetic ligands [135,177]. Nanocarriers with appropriate decoration also facilitate the in vivo generation of functional immune cells, such as CAR-T cells and M1 macrophages [140,262]. Considering the similar physicochemical (small size) and unique biological characteristics of various inorganic NPs, as well as ease of administration, effective targeting, and potential to enhance cancer elimination and prevent cancer recurrence when employed together with other anticancer therapeutic approaches, we anticipate that NPs hold great potential for advancements in anticancer therapy.
Acknowledgments
Funding: This work was supported by the National Research Foundation of Korea (NRF) grants funded by the Ministry of Science and ICT and the Ministry of Education in Republic of Korea (RS-2024-00339160 and RS-2023-00271205).
Competing interests: The authors declare that they have no competing interests.
Data Availability
Data availability is not applicable to this article as no new data were created or analyzed in this study.
References
- 1.Zhou X, Ni Y, Liang X, Lin Y, An B, He X, Zhao X. Mechanisms of tumor resistance to immune checkpoint blockade and combination strategies to overcome resistance. Front Immunol. 2022;13: Article 915094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Yu X, Fang C, Zhang K, Su C. Recent advances in nanoparticles-based platforms targeting the PD-1/PD-L1 pathway for cancer treatment. Pharmaceutics. 2022;14(8):1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Liu X, Yuan Z, Tang Z, Chen Q, Huang J, He L, Chen T. Selenium-driven enhancement of synergistic cancer chemo-/radiotherapy by targeting nanotherapeutics. Biomater Sci. 2021;9(13):4691–4700. [DOI] [PubMed] [Google Scholar]
- 4.Liu Y, Pan Y, Cao W, Xia F, Liu B, Niu J, Alfranca G, Sun X, Ma L, Fuente JM, et al. A tumor microenvironment responsive biodegradable CaCO3/MnO2- based nanoplatform for the enhanced photodynamic therapy and improved PD-L1 immunotherapy. Theranostics. 2019;9(23):6867–6884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Zhang J, Fan B, Cao G, Huang W, Jia F, Nie G, Wang H. Direct presentation of tumor-associated antigens to induce adaptive immunity by personalized dendritic cell-mimicking nanovaccines. Adv Mater. 2022;34(47):2205950. [DOI] [PubMed] [Google Scholar]
- 6.He A, Li X, Dai Z, Li Q, Zhang Y, Ding M, Wen Z, Mou Y, Dong H. Nanovaccine-based strategies for lymph node targeted delivery and imaging in tumor immunotherapy. J Nanobiotechnology. 2023;21(1):236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Hu Y, Paris S, Bertolet G, Barsoumian HB, Wang Q, Da Silva J, Patel NB, Nguyen N, Doss DJ, Huang A, et al. NBTXR3 improves the efficacy of immunoradiotherapy combining nonfucosylated anti-CTLA4 in an anti-PD1 resistant lung cancer model. Front Immunol. 2022;13:1022011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Esmaily M, Masjedi A, Hallaj S, Nabi Afjadi M, Malakotikhah F, Ghani S, Ahmadi A, Sojoodi M, Hassannia H, Atyabi F, et al. Blockade of CTLA-4 increases anti-tumor response inducing potential of dendritic cell vaccine. J Control Release. 2020;326:63–74. [DOI] [PubMed] [Google Scholar]
- 9.Feng Y, Xie X, Zhang H, Su Q, Yang G, Wei X, Li N, Li T, Qin X, Li S, et al. Multistage-responsive nanovehicle to improve tumor penetration for dual-modality imaging-guided photodynamic-immunotherapy. Biomaterials. 2021;275: Article 120990. [DOI] [PubMed] [Google Scholar]
- 10.Wang Y, Wang H. Lymph node targeting for immunotherapy. Immunooncol Technol. 2023;20: Article 100395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Morisaki T, Morisaki T, Kubo M, Morisaki S, Nakamura Y, Onishi H. Lymph nodes as anti-tumor immunotherapeutic tools: Intranodal-tumor-specific antigen-pulsed dendritic cell vaccine immunotherapy. Cancers. 2022;14(10):2438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Liu F, Tan L, Dai Z, Wang Y, Huang L, Zhang Y, Cheng Q, Li X, Liu M-D, Wang L, et al. Manganese-based microcrystals equipped with Ythdf1-targeted biomimetic nanovaccines for reinforced dendritic cell spatiotemporal orchestration. Nano Today. 2024;54: Article 102112. [Google Scholar]
- 13.Zou Z, Lin H, Li M, Lin B. Tumor-associated macrophage polarization in the inflammatory tumor microenvironment. Front Oncol. 2023;13:1103149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Meir R, Shamalov K, Betzer O, Motiei M, Horovitz-Fried M, Yehuda R, Popovtzer A, Popovtzer R, Cohen CJ. Nanomedicine for cancer immunotherapy: Tracking cancer-specific T-cells in vivo with gold nanoparticles and CT imaging. ACS Nano. 2015;9(6):6363–6372. [DOI] [PubMed] [Google Scholar]
- 15.Cevaal PM, Ali A, Czuba-Wojnilowicz E, Symons J, Lewin SR, Cortez-Jugo C, Caruso F. In vivo T cell-targeting nanoparticle drug delivery systems: Considerations for rational design. ACS Nano. 2021;15(3):3736–3753. [DOI] [PubMed] [Google Scholar]
- 16.Huang L, Ge X, Liu Y, Li H, Zhang Z. The role of toll-like receptor agonists and their nanomedicines for tumor immunotherapy. Pharmaceutics. 2022;14(6):1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Siegler EL, Kim YJ, Wang P. Nanomedicine targeting the tumor microenvironment: Therapeutic strategies to inhibit angiogenesis, remodel matrix, and modulate immune responses. J Cell Immunotherapy. 2016;2(2):69–78. [Google Scholar]
- 18.Song H, Sun H, He N, Xu C, Wang Y, Du L, Liu Y, Wang Q, Ji K, Wang J, et al. Gadolinium-based ultra-small nanoparticles augment radiotherapy-induced T-cell response to synergize with checkpoint blockade immunotherapy. Nanoscale. 2022;14(31):11429–11442. [DOI] [PubMed] [Google Scholar]
- 19.Zhang L, Xu L, Wang Y, Liu J, Tan G, Huang F, He N, Lu Z. A novel therapeutic vaccine based on graphene oxide nanocomposite for tumor immunotherapy. Chin Chem Lett. 2022;33:4089–4095. [Google Scholar]
- 20.Almeida JPM, Lin AY, Figueroa ER, Foster AE, Drezek RA. In vivo gold nanoparticle delivery of peptide vaccine induces anti-tumor immune response in prophylactic and therapeutic tumor models. Small. 2015;11(12):1453–1459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Almeida JPM, Figueroa ER, Drezek RA. Gold nanoparticle mediated cancer immunotherapy. Nanomedicine. 2014;10(3):503–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.An J, Liu M, Zhao L, Lu W, Wu S, Zhang K, Liu J, Zhang Z, Shi J. Boosting tumor immunotherapy by bioactive nanoparticles via Ca2+ interference mediated TME reprogramming and specific PD-L1 depletion. Adv Funct Mater. 2022;32(24):2201275. [Google Scholar]
- 23.Qi J, Jin F, You Y, Du Y, Liu D, Xu X, Wang J, Zhu L, Chen M, Shu G, et al. Synergistic effect of tumor chemo-immunotherapy induced by leukocyte-hitchhiking thermal-sensitive micelles. Nat Commun. 2021;12(1):4755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Pham LM, Poudel K, Ou W, Phung CD, Nguyen HT, Nguyen BL, Karmacharya P, Pandit M, Chang J-H, Jeong J-H, et al. Combination chemotherapeutic and immune-therapeutic anticancer approach via anti-PD-L1 antibody conjugated albumin nanoparticles. Int J Pharm. 2021;605: Article 120816. [DOI] [PubMed] [Google Scholar]
- 25.Zhao Y, Liu X, Liu X, Yu J, Bai X, Wu X, Guo X, Liu Z, Liu X. Combination of phototherapy with immune checkpoint blockade: Theory and practice in cancer. Front Immunol. 2022;13: Article 955920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Zeng W, Li Z, Chen H, Zeng X, Mei L. An optimal portfolio of photothermal combined immunotherapy. Cell Rep Phys Sci. 2022;3(6): Article 100898. [Google Scholar]
- 27.Wang C, Wu L, Yuan H, Yu H, Xu J, Chen S, Yan S, Wang X. A powerful antitumor “trident”: The combination of radio-, immuno- and anti-angiogenesis therapy based on mesoporous silica single coated gold nanoparticles. J Mater Chem B. 2023;11(4):879–889. [DOI] [PubMed] [Google Scholar]
- 28.Sumbayev VV, Yasinska IM, Garcia CP, Gilliland D, Lall GS, Gibbs BF, Bonsall DR, Varani L, Rossi F, Calzolai L. Gold nanoparticles downregulate interleukin-1β-induced pro-inflammatory responses. Small. 2013;9(3):472–477. [DOI] [PubMed] [Google Scholar]
- 29.Tsai C-Y, Lu S-L, Hu C-W, Yeh C-S, Lee G-B, Lei H-Y. Size-dependent attenuation of TLR9 signaling by gold nanoparticles in macrophages. J Immunol. 2012;188(1):68–76. [DOI] [PubMed] [Google Scholar]
- 30.Moyano DF, Goldsmith M, Solfiell DJ, Landesman-Milo D, Miranda OR, Peer D, Rotello VM. Nanoparticle hydrophobicity dictates immune response. J Am Chem Soc. 2012;134(9):3965–3967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Niikura K, Matsunaga T, Suzuki T, Kobayashi S, Yamaguchi H, Orba Y, Kawaguchi A, Hasegawa H, Kajino K, Ninomiya T, et al. Gold nanoparticles as a vaccine platform: Influence of size and shape on immunological responses in vitro and in vivo. ACS Nano. 2013;7(5):3926–3938. [DOI] [PubMed] [Google Scholar]
- 32.Liu Q, Zhou Y, Li M, Zhao L, Ren J, Li D, Tan Z, Wang K, Li H, Hussain M, et al. Polyethylenimine hybrid thin-shell hollow mesoporous silica nanoparticles as vaccine self-adjuvants for cancer immunotherapy. ACS Appl Mater Interfaces. 2019;11(51):47798–47809. [DOI] [PubMed] [Google Scholar]
- 33.Bai S, Jiang H, Song Y, Zhu Y, Qin M, He C, Du G, Sun X. Aluminum nanoparticles deliver a dual-epitope peptide for enhanced anti-tumor immunotherapy. J Control Release. 2022;344:134–146. [DOI] [PubMed] [Google Scholar]
- 34.Wu Y, Gu W, Li J, Chen C, Xu ZP. Silencing PD-1 and PD-L1 with nanoparticle-delivered small interfering RNA increases cytotoxicity of tumor-infiltrating lymphocytes. Nanomedicine. 2019;14(8):955–967. [DOI] [PubMed] [Google Scholar]
- 35.Feng Y, Wu J, Chen J, Lin L, Zhang S, Yang Z, Sun P, Li Y, Tian H, Chen X. Targeting dual gene delivery nanoparticles overcomes immune checkpoint blockade induced adaptive resistance and regulates tumor microenvironment for improved tumor immunotherapy. Nano Today. 2021;38: Article 101194. [Google Scholar]
- 36.Neek M, Tucker JA, Butkovich N, Nelson EL, Wang S-W. An antigen-delivery protein nanoparticle combined with anti-PD-1 checkpoint inhibitor has curative efficacy in an aggressive melanoma model. Adv Ther. 2020;3(12):2000122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Moon Y, Shim MK, Choi J, Yang S, Kim J, Yun WS, Cho H, Park JY, Kim Y, Seong J-K, et al. Anti-PD-L1 peptide-conjugated prodrug nanoparticles for targeted cancer immunotherapy combining PD-L1 blockade with immunogenic cell death. Theranostics. 2022;12(5):1999–2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zhang P, Chiu Y-C, Tostanoski LH, Jewell CM. Polyelectrolyte multilayers assembled entirely from immune signals on gold nanoparticle templates promote antigen-specific T cell response. ACS Nano. 2015;9(6):6465–6477. [DOI] [PubMed] [Google Scholar]
- 39.Jiang C, Zhang L, Xu X, Qi M, Zhang J, He S, Tian Q, Song S. Engineering a smart agent for enhanced immunotherapy effect by simultaneously blocking PD-L1 and CTLA-4. Adv Sci. 2021;8(20):2102500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zhang J, Liu D, Liu J, Han Y, Xu H, Leng X, Kong D, Liu L. Hybrid spherical nucleotide nanoparticles can enhance the synergistic anti-tumor effect of CTLA-4 and PD-1 blockades. Biomater Sci. 2020;8(17):4757–4766. [DOI] [PubMed] [Google Scholar]
- 41.Möller K, Bein T. Degradable drug carriers: Vanishing mesoporous silica nanoparticles. Chem Mater. 2019;31(12):4364–4378. [Google Scholar]
- 42.Park J, Choi SW, Cha BG, Kim J, Kang S-J. Alternative activation of macrophages through interleukin-13-loaded extra-large-pore mesoporous silica nanoparticles suppresses experimental autoimmune encephalomyelitis. ACS Biomater Sci Eng. 2021;7(9):4446–4453. [DOI] [PubMed] [Google Scholar]
- 43.Bavnhøj CG, Knopp MM, Madsen CM, Löbmann K. The role interplay between mesoporous silica pore volume and surface area and their effect on drug loading capacity. Int J Pharm X. 2019;1: Article 100008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Shahidi M, Abazari O, Dayati P, Bakhshi A, Zavarreza J, Modarresi MH, Haghiralsadat F, Rahmanian M, Naghib SM, Tofighi D. Multicomponent siRNA/miRNA-loaded modified mesoporous silica nanoparticles targeted bladder cancer for a highly effective combination therapy. Front Bioeng Biotechnol. 2022;10:949704. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 45.Nguyen TL, Cha BG, Choi Y, Im J, Kim J. Injectable dual-scale mesoporous silica cancer vaccine enabling efficient delivery of antigen/adjuvant-loaded nanoparticles to dendritic cells recruited in local macroporous scaffold. Biomaterials. 2020;239: Article 119859. [DOI] [PubMed] [Google Scholar]
- 46.Mahony D, Cavallaro AS, Stahr F, Mahony TJ, Qiao SZ, Mitter N. Mesoporous silica nanoparticles act as a self-adjuvant for ovalbumin model antigen in mice. Small. 2013;9(18):3138–3146. [DOI] [PubMed] [Google Scholar]
- 47.Hong X, Zhong X, Du G, Hou Y, Zhang Y, Zhang Z, Gong T, Zhang L, Sun X. The pore size of mesoporous silica nanoparticles regulates their antigen delivery efficiency. Sci Adv. 2024;6(25):eaaz4462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lin X, Li F, Guan J, Wang X, Yao C, Zeng Y, Liu X. Janus silica nanoparticle-based tumor microenvironment modulator for restoring tumor sensitivity to programmed cell death ligand 1 immune checkpoint blockade therapy. ACS Nano. 2023;17(15):14494–14507. [DOI] [PubMed] [Google Scholar]
- 49.Huang Y, Mao K, Zhang B, Zhao Y. Superparamagnetic iron oxide nanoparticles conjugated with folic acid for dual target-specific drug delivery and MRI in cancer theranostics. Mater Sci Eng C. 2017;70(Pt 1):763–771. [DOI] [PubMed] [Google Scholar]
- 50.Estelrich J, Escribano E, Queralt J, Busquets MA. Iron oxide nanoparticles for magnetically-guided and magnetically-responsive drug delivery. Int J Mol Sci. 2015;16(4):8070–8101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Mejías R, Pérez-Yagüe S, Gutiérrez L, Cabrera LI, Spada R, Acedo P, Serna CJ, Lázaro FJ, Villanueva Á, del P. Morales M, et al. Dimercaptosuccinic acid-coated magnetite nanoparticles for magnetically guided in vivo delivery of interferon gamma for cancer immunotherapy. Biomaterials. 2011;32(11):2938–2952. [DOI] [PubMed] [Google Scholar]
- 52.Cho N-H, Cheong T-C, Min JH, Wu JH, Lee SJ, Kim D, Yang J-S, Kim S, Kim YK, Seong S-Y. A multifunctional core–shell nanoparticle for dendritic cell-based cancer immunotherapy. Nat Nanotechnol. 2011;6(10):675–682. [DOI] [PubMed] [Google Scholar]
- 53.Huang J, Bu L, Xie J, Chen K, Cheng Z, Li X, Chen X. Effects of nanoparticle size on cellular uptake and liver MRI with polyvinylpyrrolidone-coated iron oxide nanoparticles. ACS Nano. 2010;4(12):7151–7160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Cortés-Llanos B, Ocampo SM, Cueva L, Calvo GF, Belmonte-Beitia J, Pérez L, Salas G, Ayuso-Sacido Á. Influence of coating and size of magnetic nanoparticles on cellular uptake for in vitro MRI. Nanomaterials. 2021;11(11):2888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jeon M, Lin G, Stephen ZR, Kato FL, Zhang M. Paclitaxel-loaded iron oxide nanoparticles for targeted breast cancer therapy. Adv Ther. 2019;2(12):1900081. [Google Scholar]
- 56.Zheng S, Tian Y, Ouyang J, Shen Y, Wang X, Luan J. Carbon nanomaterials for drug delivery and tissue engineering. Front Chem. 2022;10: Article 990362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Liu W, Speranza G. Functionalization of carbon nanomaterials for biomedical applications. C. 2019;5(4):72. [Google Scholar]
- 58.Debnath SK, Srivastava R. Drug delivery with carbon-based nanomaterials as versatile nanocarriers: Progress and prospects. Front Nanotechnol. 2021;3:644564. [Google Scholar]
- 59.Mashal A, Sitharaman B, Li X, Avti PK, Sahakian AV, Booske JH, Hagness SC. Toward carbon-nanotube-based theranostic agents for microwave detection and treatment of breast cancer: Enhanced dielectric and heating response of tissue-mimicking materials. IEEE Trans Biomed Eng. 2010;57(8):1831–1834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Golubewa L, Timoshchenko I, Romanov O, Karpicz R, Kulahava T, Rutkauskas D, Shuba M, Dementjev A, Svirko Y, Kuzhir P. Single-walled carbon nanotubes as a photo-thermo-acoustic cancer theranostic agent: Theory and proof of the concept experiment. Sci Rep. 2020;10(1):22174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Tang L, Zhang A, Mei Y, Xiao Q, Xu X, Wang W. NIR light-triggered chemo-phototherapy by ICG functionalized MWNTs for synergistic tumor-targeted delivery. Pharmaceutics. 2021;13(12):2145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Zhao Y, Zhao T, Cao Y, Sun J, Zhou Q, Chen H, Guo S, Wang Y, Zhen Y, Liang X-J, et al. Temperature-sensitive lipid-coated carbon nanotubes for synergistic photothermal therapy and gene therapy. ACS Nano. 2021;15(4):6517–6529. [DOI] [PubMed] [Google Scholar]
- 63.Hassan HAFM, Smyth L, Wang JT-W, Costa PM, Ratnasothy K, Diebold SS, Lombardi G, Al-Jamal KT. Dual stimulation of antigen presenting cells using carbon nanotube-based vaccine delivery system for cancer immunotherapy. Biomaterials. 2016;104:310–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Zhao D, Alizadeh D, Zhang L, Liu W, Farrukh O, Manuel E, Diamond DJ, Badie B. Carbon nanotubes enhance CpG uptake and potentiate antiglioma immunity. Clin Cancer Res. 2011;17(4):771–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Fadel TR, Steenblock ER, Stern E, Li N, Wang X, Haller GL, Pfefferle LD, Fahmy TM. Enhanced cellular activation with single walled carbon nanotube bundles presenting antibody stimuli. Nano Lett. 2008;8(7):2070–2076. [DOI] [PubMed] [Google Scholar]
- 66.Fadel TR, Look M, Staffier PA, Haller GL, Pfefferle LD, Fahmy TM. Clustering of stimuli on single-walled carbon nanotube bundles enhances cellular activation. Langmuir. 2010;26(8):5645–5654. [DOI] [PubMed] [Google Scholar]
- 67.Yu W, Sisi L, Haiyan Y, Jie L. Progress in the functional modification of graphene/graphene oxide: A review. RSC Adv. 2020;10(26):15328–15345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Aliyev E, Filiz V, Khan MM, Lee YJ, Abetz C, Abetz V. Structural characterization of graphene oxide: Surface functional groups and fractionated oxidative debris. Nanomaterials. 2019;9(8):1180 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Zhuang W-R, Wang Y, Cui P-F, Xing L, Lee J, Kim D, Jiang H-L, Oh Y-K. Applications of π-π stacking interactions in the design of drug-delivery systems. J Control Release. 2019;294:311–326. [DOI] [PubMed] [Google Scholar]
- 70.Yin Y, Li X, Ma H, Zhang J, Yu D, Zhao R, Yu S, Nie G, Wang H. In situ transforming RNA nanovaccines from polyethylenimine functionalized graphene oxide hydrogel for durable cancer immunotherapy. Nano Lett. 2021;21(5):2224–2231. [DOI] [PubMed] [Google Scholar]
- 71.Luo L, Liu C, He T, Zeng L, Xing J, Xia Y, Pan Y, Gong C, Wu A. Engineered fluorescent carbon dots as promising immune adjuvants to efficiently enhance cancer immunotherapy. Nanoscale. 2018;10(46):22035–22043. [DOI] [PubMed] [Google Scholar]
- 72.Qiu C, Wu Y, Guo Q, Shi Q, Zhang J, Meng Y, Xia F, Wang J. Preparation and application of calcium phosphate nanocarriers in drug delivery. Mater Today Bio. 2022;17: Article 100501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Cai R-Q, Liu D-Z, Cui H, Cheng Y, Liu M, Zhang B-L, Mei Q-B, Zhou S-Y. Charge reversible calcium phosphate lipid hybrid nanoparticle for siRNA delivery. Oncotarget. 2017;8(26):42772–42788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Li Q, Chao Y, Liu B, Xiao Z, Yang Z, Wu Y, Liu Z. Disulfiram loaded calcium phosphate nanoparticles for enhanced cancer immunotherapy. Biomaterials. 2022;291: Article 121880. [DOI] [PubMed] [Google Scholar]
- 75.Zhang X, Wei M, Zhang Z, Zeng Y, Zou F, Zhang S, Wang Z, Chen F, Xiong H, Li Y, et al. Risedronate-functionalized manganese-hydroxyapatite amorphous particles: A potent adjuvant for subunit vaccines and cancer immunotherapy. J Control Release. 2024;367:13–26. [DOI] [PubMed] [Google Scholar]
- 76.Sun B, Zhao X, Gu W, Cao P, Movahedi F, Wu Y, Xu ZP, Gu W. ATP stabilised and sensitised calcium phosphate nanoparticles as effective adjuvants for a DNA vaccine against cancer. J Mater Chem B. 2021;9(36):7435–7446. [DOI] [PubMed] [Google Scholar]
- 77.Haque S, Tripathy S, Patra CR. Manganese-based advanced nanoparticles for biomedical applications: Future opportunity and challenges. Nanoscale. 2021;13(39):16405–16426. [DOI] [PubMed] [Google Scholar]
- 78.Prasad P, Gordijo CR, Abbasi AZ, Maeda A, Ip A, Rauth AM, DaCosta RS, Wu XY. Multifunctional albumin–MnO2 nanoparticles modulate solid tumor microenvironment by attenuating hypoxia, acidosis, vascular endothelial growth factor and enhance radiation response. ACS Nano. 2014;8(4):3202–3212. [DOI] [PubMed] [Google Scholar]
- 79.Sobańska Z, Roszak J, Kowalczyk K, Stępnik M. Applications and biological activity of nanoparticles of manganese and manganese oxides in in vitro and in vivo models. Nanomaterials. 2021;11(5):1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sun Y, Wang Y, Liu Y, Wang H, Yang C, Liu X, Wang F. Integration of manganese dioxide-based nanomaterials for biomedical applications. Adv NanoBiomed Res. 2023;3(1):2200093. [Google Scholar]
- 81.Alsharif NB, Bere K, Sáringer S, Samu GF, Takács D, Hornok V, Szilagyi I. Design of hybrid biocatalysts by controlled heteroaggregation of manganese oxide and sulfate latex particles to combat reactive oxygen species. J Mater Chem B. 2021;9(24):4929–4940. [DOI] [PubMed] [Google Scholar]
- 82.Bellusci M, La Barbera A, Padella F, Mancuso M, Pasquo A, Grollino MG, Leter G, Nardi E, Cremisini C, Giardullo P, et al. Biodistribution and acute toxicity of a nanofluid containing manganese iron oxide nanoparticles produced by a mechanochemical process. Int J Nanomedicine. 2014;9:1919–1929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Deng Z, Xi M, Zhang C, Wu X, Li Q, Wang C, Fang H, Sun G, Zhang Y, Yang G, et al. Biomineralized MnO2 nanoplatforms mediated delivery of immune checkpoint inhibitors with STING pathway activation to potentiate cancer radio-immunotherapy. ACS Nano. 2023;17:4495–4506. [DOI] [PubMed] [Google Scholar]
- 84.Song T, Liao Y, Zuo Q, Liu N, Liu Z. MnO2 nanoparticles as a minimalist multimode vaccine adjuvant/delivery system to regulate antigen presenting cells for tumor immunotherapy. J Mater Chem B. 2022;10(18):3474–3490. [DOI] [PubMed] [Google Scholar]
- 85.Cai X, Zhu Q, Zeng Y, Zeng Q, Chen X, Zhan Y. Manganese oxide nanoparticles as MRI contrast agents in tumor multimodal imaging and therapy. Int J Nanomedicine. 2019;14:8321–8344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Yen T-YC, Abbasi AZ, He C, Lip H-Y, Park E, Amini MA, Adissu HA, Foltz W, Rauth AM, Henderson J, et al. Biocompatible and bioactivable terpolymer-lipid-MnO2 nanoparticle-based MRI contrast agent for improving tumor detection and delineation. Mater Today Bio. 2024;25: Article 100954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Zhao Y, Qu Y, Hao C, Yao W. PD-1/PD-L1 axis in organ fibrosis. Front Immunol. 2023;14:1145682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Dutta S, Ganguly A, Chatterjee K, Spada S, Mukherjee S. Targets of immune escape mechanisms in cancer: Basis for development and evolution of cancer immune checkpoint inhibitors. Biology. 2023;12(12):218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Xu S, Wang C, Yang L, Wu J, Li M, Xiao P, Xu Z, Xu Y, Wang K. Targeting immune checkpoints on tumor-associated macrophages in tumor immunotherapy. Front Immunol. 2023;14:1199631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Han Y, Liu D, Li L. PD-1/PD-L1 pathway: Current researches in cancer. Am J Cancer Res. 2020;10(3):727–742. [PMC free article] [PubMed] [Google Scholar]
- 91.Li X, Yang Y, Huang Q, Deng Y, Guo F, Wang G, Liu M. Crosstalk between the tumor microenvironment and cancer cells: A promising predictive biomarker for immune checkpoint inhibitors. Front Cell Dev Biol. 2021;9: Article 738373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Sharon E, Streicher H, Goncalves P, Chen HX. Immune checkpoint inhibitors in clinical trials. Chin J Cancer. 2014;33(9):434–444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Alturki NA. Review of the immune checkpoint inhibitors in the context of cancer treatment. J Clin Med. 2023;12(13):4301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Martins F, Sofiya L, Sykiotis GP, Lamine F, Maillard M, Fraga M, Shabafrouz K, Ribi C, Cairoli A, Guex-Crosier Y, et al. Adverse effects of immune-checkpoint inhibitors: Epidemiology, management and surveillance. Nat Rev Clin Oncol. 2019;16(9):563–580. [DOI] [PubMed] [Google Scholar]
- 95.Conroy M, Naidoo J. Immune-related adverse events and the balancing act of immunotherapy. Nat Commun. 2022;13(1):392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Grossman JE, Vasudevan D, Joyce CE, Hildago M. Is PD-L1 a consistent biomarker for anti-PD-1 therapy? The model of balstilimab in a virally-driven tumor. Oncogene. 2021;40(8):1393–1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Jiang C, Cao SR, Li N, Jiang L, Sun T. PD-1 and PD-L1 correlated gene expression profiles and their association with clinical outcomes of breast cancer. Cancer Cell Int. 2019;19:2333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Wu Y, Chen W, Xu ZP, Gu W. PD-L1 distribution and perspective for cancer immunotherapy—Blockade, knockdown, or inhibition. Front Immunol. 2019;10:2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wu Q, Jiang L, Li S-C, He Q-J, Yang B, Cao J. Small molecule inhibitors targeting the PD-1/PD-L1 signaling pathway. Acta Pharmacol Sin. 2021;42(1):1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Wang Z, Wu X. Study and analysis of antitumor resistance mechanism of PD1/PD-L1 immune checkpoint blocker. Cancer Med. 2020;9(21):8086–8121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Sun JY, Zhang D, Wu S, Xu M, Zhou X, Lu XJ, Ji J. Resistance to PD-1/PD-L1 blockade cancer immunotherapy: Mechanisms, predictive factors, and future perspectives. Biomark Res. 2020;8:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Wang G, Wu W, Yang H, Zhang P, Wang J-Y. Intact polyaniline coating as a conductive guidance is beneficial to repairing sciatic nerve injury. J Biomed Mater Res Part B Appl Biomater. 2020;108:128–142. [DOI] [PubMed] [Google Scholar]
- 103.Liu H, Xu C, Meng M, Li S, Sheng S, Zhang S, Ni W, Tian H, Wang Q. Metal-organic framework-mediated multifunctional nanoparticles for combined chemo-photothermal therapy and enhanced immunotherapy against colorectal cancer. Acta Biomater. 2022;144:132–141. [DOI] [PubMed] [Google Scholar]
- 104.Alsaiari SK, Qutub SS, Sun S, Baslyman W, Aldehaiman M, Alyami M, Almalik A, Halwani R, Merzaban J, Mao Z, et al. Sustained and targeted delivery of checkpoint inhibitors by metal-organic frameworks for cancer immunotherapy. Sci Adv. 2021;7:eabe7174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Yang YSS, Moynihan KD, Bekdemir A, Dichwalkar TM, Noh MM, Watson N, Melo M, Ingram J, Suh H, Ploegh H, et al. Targeting small molecule drugs to T cells with antibody-directed cell-penetrating gold nanoparticles. Biomater Sci. 2018;7:113–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Chu H, Zhao J, Mi Y, Di Z, Li L. NIR-light-mediated spatially selective triggering of anti-tumor immunity via upconversion nanoparticle-based immunodevices. Nat Commun. 2019;10(1):2839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Reda M, Ngamcherdtrakul W, Nelson MA, Siriwon N, Wang R, Zaidan HY, Bejan DS, Reda S, Hoang NH, Crumrine NA, et al. Development of a nanoparticle-based immunotherapy targeting PD-L1 and PLK1 for lung cancer treatment. Nat Commun. 2022;13(1):4261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Liu B, Cao W, Qiao G, Yao S, Pan S, Wang L, Yue C, Ma L, Liu Y, Cui D. Effects of gold nanoprism-assisted human PD-L1 siRNA on both gene down-regulation and photothermal therapy on lung cancer. Acta Biomater. 2019;99:307–319. [DOI] [PubMed] [Google Scholar]
- 109.Pandey R, Yang F-S, Sivasankaran VP, Lo Y-L, Wu Y-T, Chang C-Y, Chiu C-C, Liao Z-X, Wang L-F. Comparing the variants of iron oxide nanoparticle-mediated delivery of miRNA34a for efficiency in silencing of PD-L1 genes in cancer cells. Pharmaceutics. 2023;15(1):215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Ge R, Liu C, Zhang X, Wang W, Li B, Liu J, Liu Y, Sun H, Zhang D, Hou Y, et al. Photothermal-activatable Fe3O4 superparticle nanodrug carriers with PD-L1 immune checkpoint blockade for anti-metastatic cancer immunotherapy. ACS Appl Mater Interfaces. 2018;10(24):20342–20355. [DOI] [PubMed] [Google Scholar]
- 111.Ge Y-X, Zhang T-W, Zhou L, Ding W, Liang H-F, Hu Z-C, Chen Q, Dong J, Xue F-F, Yin X-F, et al. Enhancement of anti-PD-1/PD-L1 immunotherapy for osteosarcoma using an intelligent autophagy-controlling metal organic framework. Biomaterials. 2022;282: Article 121407. [DOI] [PubMed] [Google Scholar]
- 112.Yan C, Liu Y, Zhao G, Yang H, Lv H, Li G, Li Y, Fu Y, Sun F, Feng Y, et al. Inhalable metal–organic framework-mediated cuproptosis combined with PD-L1 checkpoint blockade for lung metastasis synergistic immunotherapy. Acta Pharm Sin B. 2024;14(5):2281–2297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Li Z, Bu J, Zhu X, Zhou H, Ren K, Chu PK, Li L, Hu X, Ding X. Anti-tumor immunity and ferroptosis of hepatocellular carcinoma are enhanced by combined therapy of sorafenib and delivering modified GO-based PD-L1 siRNAs. Biomater Adv. 2022;136: Article 212761. [DOI] [PubMed] [Google Scholar]
- 114.Liu Y, Yang J, Liu B, Cao W, Zhang J, Yang Y, Ma L, Fuente JM, Song J, Ni J, et al. Human iPS cells loaded with MnO2-based nanoprobes for photodynamic and simultaneous enhanced immunotherapy against cancer. Nanomicro Lett. 2020;12(1):127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Grippin AJ, Wummer B, Wildes T, Dyson K, Trivedi V, Yang C, Sebastian M, Mendez-Gomez HR, Padala S, Grubb M, et al. Dendritic cell-activating magnetic nanoparticles enable early prediction of antitumor response with magnetic resonance imaging. ACS Nano. 2019;13(12):13884–13898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Gardner D, Jeffery LE, Sansom DM. Understanding the CD28/CTLA-4 (CD152) pathway and its implications for costimulatory blockade. Am J Transplant. 2014;14(9):1985–1991. [DOI] [PubMed] [Google Scholar]
- 117.Chikuma S. CTLA-4, an essential immune-checkpoint for T-cell activation. Curr Top Microbiol Immunol. 2017;410:99–126. [DOI] [PubMed] [Google Scholar]
- 118.Rowshanravan B, Halliday N, Sansom DM. CTLA-4: A moving target in immunotherapy. Blood. 2018;131(1):58–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Crepeau RL, Ford ML. Challenges and opportunities in targeting the CD28/CTLA-4 pathway in transplantation and autoimmunity. Expert Opin Biol Ther. 2017;17(8):1001–1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Hajihassan Z, Afsharian NP, Ansari-Pour N. In silico engineering a CD80 variant with increased affinity to CTLA-4 and decreased affinity to CD28 for optimized cancer immunotherapy. J Immunol Methods. 2023;513: Article 113425. [DOI] [PubMed] [Google Scholar]
- 121.Ji H-H, Tang X-W, Dong Z, Song L, Jia Y-T. Adverse event profiles of anti-CTLA-4 and anti-PD-1 monoclonal antibodies alone or in combination: Analysis of spontaneous reports submitted to FAERS. Clin Drug Investig. 2019;39(3):319–330. [DOI] [PubMed] [Google Scholar]
- 122.Wang C, Xu L, Liang C, Xiang J, Peng R, Liu Z. Immunological responses triggered by photothermal therapy with carbon nanotubes in combination with anti-CTLA-4 therapy to inhibit cancer metastasis. Adv Mater. 2014;26(48):8154–8162. [DOI] [PubMed] [Google Scholar]
- 123.Li M, Lin C, Shen A, Ma X, Ni J, Wu J, Wang W, Wang P, Gao X. A smart responsive nanotheranostic system for MRI of tumor response to immunotherapy and enhanced synergism of thermo-immunotherapy. Adv Funct Mater. 2024;34(23):2315171. [Google Scholar]
- 124.Jiang H, Fu H, Guo Y, Hu P, Shi J. Evoking tumor associated macrophages by mitochondria-targeted magnetothermal immunogenic cell death for cancer immunotherapy. Biomaterials. 2022;289: Article 121799. [DOI] [PubMed] [Google Scholar]
- 125.Bol KF, Schreibelt G, Gerritsen WR, Vries IJM, Figdor CG. Dendritic cell–based immunotherapy: State of the art and beyond. Clin Cancer Res. 2016;22(8):1897–1906. [DOI] [PubMed] [Google Scholar]
- 126.Sabado RL, Balan S, Bhardwaj N. Dendritic cell-based immunotherapy. Cell Res. 2017;27(1):74–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Ghasemi A, Martinez-Usatorre A, Li L, Hicham M, Guichard A, Marcone R, Fournier N, Torchia B, Martinez Bedoya D, Davanture S, et al. Cytokine-armed dendritic cell progenitors for antigen-agnostic cancer immunotherapy. Nat Cancer. 2024;5(2):240–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Seong J, Jeong S, Kim S, Yun S, Baek Y, Kim K. Ex vivo activation of dendritic cells via coacervate-mediated exogenous tumor cell lysate delivery. Biomater Sci. 2023;11(13):4537–4548. [DOI] [PubMed] [Google Scholar]
- 129.Zhao Y, Qiao G, Wang X, Song Y, Zhou X, Jiang N, Zhou L, Huang H, Zhao J, Morse MA, et al. Combination of DC/CIK adoptive T cell immunotherapy with chemotherapy in advanced non-small-cell lung cancer (NSCLC) patients: A prospective patients’ preference-based study (PPPS). Clin Transl Oncol. 2019;21(6):721–728. [DOI] [PubMed] [Google Scholar]
- 130.Smith R, Wafa EI, Geary SM, Ebeid K, Alhaj-Suliman SO, Salem AK. Cationic nanoparticles enhance T cell tumor infiltration and antitumor immune responses to a melanoma vaccine. Sci Adv. 2024;8(29):eabk3150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Han D, Liu J, Chen C, Dong L, Liu Y, Chang R, Huang X, Liu Y, Wang J, Dougherty U, et al. Anti-tumour immunity controlled through mRNA m6A methylation and YTHDF1 in dendritic cells. Nature. 2019;566(7743):270–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Ouyang Z, Gao Y, Yang R, Shen M, Shi X. Genetic engineering of dendritic cells using partially zwitterionic dendrimer-entrapped gold nanoparticles boosts efficient tumor immunotherapy. Biomacromolecules. 2022;23(3):1326–1336. [DOI] [PubMed] [Google Scholar]
- 133.Kim WG, Choi B, Yang H-J, Han J-A, Jung H, Cho H, Kang S, Hong SY. Covalent conjugation of small-molecule adjuvants to nanoparticles induces robust cytotoxic T cell responses via DC activation. Bioconjug Chem. 2016;27(9):2007–2013. [DOI] [PubMed] [Google Scholar]
- 134.Hanlon DJ, Aldo PB, Devine L, Alvero AB, Engberg AK, Edelson R, Mor G. Enhanced stimulation of anti-ovarian cancer CD8+ T cells by dendritic cells loaded with nanoparticle encapsulated tumor antigen. Am J Reprod Immunol. 2011;65(6):597–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Zeng Z, Pu K. Improving cancer immunotherapy by cell membrane-camouflaged nanoparticles. Adv Funct Mater. 2020;30(43):2004397. [Google Scholar]
- 136.Cheng S, Xu C, Jin Y, Li Y, Zhong C, Ma J, Yang J, Zhang N, Li Y, Wang C, et al. Artificial mini dendritic cells boost T cell–based immunotherapy for ovarian cancer. Adv Sci. 2020;7(7):1903301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Raza F, Zafar H, Zhang S, Kamal Z, Su J, Yuan W-E, Mingfeng Q. Recent advances in cell membrane-derived biomimetic nanotechnology for cancer immunotherapy. Adv Healthc Mater. 2021;10(6):e2002081. [DOI] [PubMed] [Google Scholar]
- 138.Li C, Jiang P, Wei S, Xu X, Wang J. Regulatory T cells in tumor microenvironment: New mechanisms, potential therapeutic strategies and future prospects. Mol Cancer. 2020;19(1):116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Sterner RC, Sterner RM. CAR-T cell therapy: Current limitations and potential strategies. Blood Cancer J. 2021;11:69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Xin T, Cheng L, Zhou C, Zhao Y, Hu Z, Wu X. In-vivo induced CAR-T cell for the potential breakthrough to overcome the barriers of current CAR-T cell therapy. Front Oncol. 2022;12: Article 809754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Wang X, Meng F, Yen Y-T, Li R, Liu B. Nanotechnology-based CAR-T strategies for improving efficacy and safety of tumor immunotherapy. Adv Funct Mater. 2021;31(1):2004713. [Google Scholar]
- 142.Boutilier AJ, Elsawa SF. Macrophage polarization states in the tumor microenvironment. Int J Mol Sci. 2021;22(13):6995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Gao J, Liang Y, Wang L. Shaping polarization of tumor-associated macrophages in cancer immunotherapy. Front Immunol. 2022;13: Article 888713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Chen S, Saeed AFUH, Liu Q, Jiang Q, Xu H, Xiao GG, Rao L, Duo Y. Macrophages in immunoregulation and therapeutics. Signal Transduct Target Ther. 2023;8(1):207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Monnier M, Paolini L, Vinatier E, Mantovani A, Delneste Y, Jeannin P. Antitumor strategies targeting macrophages: The importance of considering the differences in differentiation/polarization processes between human and mouse macrophages. J Immunother Cancer. 2022;10(10):e005560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Duan Z, Luo Y. Targeting macrophages in cancer immunotherapy. Signal Transduct Target Ther. 2021;6(1):127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Kashfi K, Kannikal J, Nath N. Macrophage reprogramming and cancer therapeutics: Role of iNOS-derived NO. Cells. 2021;10(11):3194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Wang P, Geng J, Gao J, Zhao H, Li J, Shi Y, Yang B, Xiao C, Linghu Y, Sun X, et al. Macrophage achieves self-protection against oxidative stress-induced ageing through the Mst-Nrf2 axis. Nat Commun. 2019;10(1):755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Canton M, Sánchez-Rodríguez R, Spera I, Venegas FC, Favia M, Viola A, Castegna A. Reactive oxygen species in macrophages: Sources and targets. Front Immunol. 2021;12: Article 734229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Rath M, Müller I, Kropf P, Closs EI, Munder M. Metabolism via arginase or nitric oxide synthase: Two competing arginine pathways in macrophages. Front Immunol. 2014;5:532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Latour YL, Gobert AP, Wilson KT. The role of polyamines in the regulation of macrophage polarization and function. Amino Acids. 2020;52(2):151–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Shen K-Y, Song Y-C, Chen I-H, Chong P, Liu S-J. Depletion of tumor-associated macrophages enhances the anti-tumor immunity induced by a toll-like receptor agonist-conjugated peptide. Hum Vaccin Immunother. 2014;10(11):3241–3250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Liu J, Geng X, Hou J, Wu G. New insights into M1/M2 macrophages: Key modulators in cancer progression. Cancer Cell Int. 2021;21(1):389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Chen P-M, Pan W-Y, Luo P-K, Phung HN, Liu Y-M, Chiang M-C, Chang W-A, Tien T-L, Huang C-Y, Wu W-W, et al. Pollen-mimetic metal–organic frameworks with tunable spike-like nanostructures that promote cell interactions to improve antigen-specific humoral immunity. ACS Nano. 2021;15(4):7596–7607. [DOI] [PubMed] [Google Scholar]
- 155.Wang W, Wang P, Tang X, Elzatahry AA, Wang S, Al-Dahyan D, Zhao M, Yao C, Hung C-T, Zhu X, et al. Facile synthesis of uniform virus-like mesoporous silica nanoparticles for enhanced cellular internalization. ACS Cent. Sci. 2017;3(8):839–846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Luo Y-H, Wu ZW, Tsai H-T, Lin S-Y, Lin P. Endotoxin nanovesicles: Hydrophilic gold nanodots control supramolecular lipopolysaccharide assembly for modulating immunological responses. Nano Lett. 2015;15(10):6446–6453. [DOI] [PubMed] [Google Scholar]
- 157.Lavie D, Ben-Shmuel A, Erez N, Scherz-Shouval R. Cancer-associated fibroblasts in the single-cell era. Nat Cancer. 2022;3(7):793–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Han C, Liu T, Yin R. Biomarkers for cancer-associated fibroblasts. Biomark Res. 2020;8(1):64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Kieffer Y, Hocine HR, Gentric G, Pelon F, Bernard C, Bourachot B, Lameiras S, Albergante L, Bonneau C, Guyard A, et al. Single-cell analysis reveals fibroblast clusters linked to immunotherapy resistance in cancer. Cancer Discov. 2020;10(9):1330–1351. [DOI] [PubMed] [Google Scholar]
- 160.Thiery J. Modulation of the antitumor immune response by cancer-associated fibroblasts: Mechanisms and targeting strategies to hamper their immunosuppressive functions. Explor Target Antitumor Ther. 2022;3(5):598–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Liu T, Han C, Wang S, Fang P, Ma Z, Xu L, Yin R. Cancer-associated fibroblasts: An emerging target of anti-cancer immunotherapy. J Hematol Oncol. 2019;12(1):86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Zhang C, Fei Y, Wang H, Hu S, Liu C, Hu R, Du Q. CAFs orchestrates tumor immune microenvironment—A new target in cancer therapy. Front Pharmacol. 2023;14:1113378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Barrett RL, Puré E. Cancer-associated fibroblasts and their influence on tumor immunity and immunotherapy. eLife. 2020;9:e57243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Zhou W, Guo S, Liu M, Burow ME, Wang G. Targeting CXCL12/CXCR4 axis in tumor immunotherapy. Curr Med Chem. 2019;26(17):3026–3041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Andorko JI, Tostanoski LH, Solano E, Mukhamedova M, Jewell CM. Intra-lymph node injection of biodegradable polymer particles. J Vis Exp. 2014;(83): Article e50984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Kato S, Shirai Y, Sakamoto M, Mori S, Kodama T. Use of a lymphatic drug delivery system and sonoporation to target malignant metastatic breast cancer cells proliferating in the marginal sinuses. Sci Rep. 2019;9:13242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Li Z-Z, Zhong N-N, Cao L-M, Cai Z-M, Xiao Y, Wang G-R, Liu B, Xu C, Bu L-L. Nanoparticles targeting lymph nodes for cancer immunotherapy: Strategies and influencing factors. Small. 2024;20(19):2308731. [DOI] [PubMed] [Google Scholar]
- 168.Moon JJ, Huang B, Irvine DJ. Engineering nano- and microparticles to tune immunity. Adv Mater. 2012;24(28):3724–3746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Zhu G, Zhang F, Ni Q, Niu G, Chen X. Efficient nanovaccine delivery in cancer immunotherapy. ACS Nano. 2017;11(3):2387–2392. [DOI] [PubMed] [Google Scholar]
- 170.Lambert LA, Gibson GR, Maloney M, Durell B, Noelle RJ, Barth RJ Jr. Intranodal immunization with tumor lysate-pulsed dendritic cells enhances protective antitumor Immunity1. Cancer Res. 2001;61(2):641–646. [PubMed] [Google Scholar]
- 171.Lesimple T, Neidhard E-M, Vignard V, Lefeuvre C, Adamski H, Labarrière N, Carsin A, Monnier D, Collet B, Clapisson G, et al. Immunologic and clinical effects of injecting mature peptide-loaded dendritic cells by intralymphatic and intranodal routes in metastatic melanoma patients. Clin Cancer Res. 2006;12(24):7380–7388. [DOI] [PubMed] [Google Scholar]
- 172.Gilliet M, Kleinhans M, Lantelme E, Schadendorf D, Burg G, Nestle FO. Intranodal injection of semimature monocyte-derived dendritic cells induces T helper type 1 responses to protein neoantigen. Blood. 2003;102(1):36–42. [DOI] [PubMed] [Google Scholar]
- 173.Murthy V, Minehart J, Sterman DH. Local immunotherapy of cancer: Innovative approaches to harnessing tumor-specific immune responses. J Natl Cancer Inst. 2017;109(12):djx097. [DOI] [PubMed] [Google Scholar]
- 174.Sukhbaatar A, Mori S, Kodama T. Intranodal delivery of modified docetaxel: Innovative therapeutic method to inhibit tumor cell growth in lymph nodes. Cancer Sci. 2022;113(4):1125–1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Zinkhan S, Ogrina A, Balke I, Reseviča G, Zeltins A, Brot S, Lipp C, Chang X, Zha L, Vogel M, et al. The impact of size on particle drainage dynamics and antibody response. J Control Release. 2021;331:296–308. [DOI] [PubMed] [Google Scholar]
- 176.Kjellman P, Fredriksson S, Kjellman C, Strand S-E, In ’t Zandt R. Size-dependent lymphatic uptake of nanoscale-tailored particles as tumor mass increases. Future Sci OA. 2015;1(4):FSO60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Howard GP, Verma G, Ke X, Thayer WM, Hamerly T, Baxter VK, Lee JE, Dinglasan RR, Mao H-Q. Critical size limit of biodegradable nanoparticles for enhanced lymph node trafficking and paracortex penetration. Nano Res. 2019;12(4):837–844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Fromen CA, Rahhal TB, Robbins GR, Kai MP, Shen TW, Luft JC, DeSimone JM. Nanoparticle surface charge impacts distribution, uptake and lymph node trafficking by pulmonary antigen-presenting cells. Nanomedicine. 2016;12(3):677–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Ke X, Howard GP, Tang H, Cheng B, Saung MT, Santos JL, Mao H-Q. Physical and chemical profiles of nanoparticles for lymphatic targeting. Adv Drug Deliv Rev. 2019;151–152:72–93. [DOI] [PubMed] [Google Scholar]
- 180.Yuan Z, Li Y, Zhang S, Wang X, Dou H, Yu X, Zhang Z, Yang S, Xiao M. Extracellular matrix remodeling in tumor progression and immune escape: From mechanisms to treatments. Mol Cancer. 2023;22:48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Henke E, Nandigama R, Ergün S. Extracellular matrix in the tumor microenvironment and its impact on cancer therapy. Front Mol Biosci. 2020;6:160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19(11):1423–1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Neophytou CM, Panagi M, Stylianopoulos T, Papageorgis P. The role of tumor microenvironment in cancer metastasis: Molecular mechanisms and therapeutic opportunities. Cancers (Basel). 2021;13(9):2053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Baghban R, Roshangar L, Jahanban-Esfahlan R, Seidi K, Ebrahimi-Kalan A, Jaymand M, Kolahian S, Javaheri T, Zare P. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal. 2020;18(1):59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Serpa J. Metabolic remodeling as a way of adapting to tumor microenvironment (TME), a job of several holders. Adv Exp Med Biol. 2020;1219:1–34. [DOI] [PubMed] [Google Scholar]
- 186.Nakahara R, Maeda K, Aki S, Osawa T. Metabolic adaptations of cancer in extreme tumor microenvironments. Cancer Sci. 2023;114(4):1200–1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Babar Q, Saeed A, Tabish TA, Sarwar M, Thorat ND. Targeting the tumor microenvironment: Potential strategy for cancer therapeutics. Biochim Biophys Acta Mol Basis Dis. 2023;1869(6): Article 166746. [DOI] [PubMed] [Google Scholar]
- 188.Pitt JM, Marabelle A, Eggermont A, Soria J-C, Kroemer G, Zitvogel L. Targeting the tumor microenvironment: Removing obstruction to anticancer immune responses and immunotherapy. Ann Oncol. 2016;27(8):1482–1492. [DOI] [PubMed] [Google Scholar]
- 189.Wen Y, Zhu Y, Zhang C, Yang X, Gao Y, Li M, Yang H, Liu T, Tang H. Chronic inflammation, cancer development and immunotherapy. Front Pharmacol. 2022;13:1040163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Zhao H, Wu L, Yan G, Chen Y, Zhou M, Wu Y, Li Y. Inflammation and tumor progression: Signaling pathways and targeted intervention. Signal Transduct Target Ther. 2021;6(1):263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Tie Y, Tang F, Wei Y-Q, Wei X-W. Immunosuppressive cells in cancer: Mechanisms and potential therapeutic targets. J Hematol Oncol. 2022;15(1):61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Zhong S, Jeong J-H, Chen Z, Chen Z, Luo J-L. Targeting tumor microenvironment by small-molecule inhibitors. Transl Oncol. 2020;13(1):57–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Park D, Lee SJ, Park J-W. Aptamer-based smart targeting and spatial trigger–response drug-delivery systems for anticancer therapy. Biomedicines. 2024;12(1):187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Uthaman S, Huh KM, Park I-K. Tumor microenvironment-responsive nanoparticles for cancer theragnostic applications. Biomater Res. 2018;22:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Subhan MA, Parveen F, Filipczak N, Yalamarty SSK, Torchilin VP. Approaches to improve EPR-based drug delivery for cancer therapy and diagnosis. J Pers Med. 2023;13(3):389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Crittenden MR, Thanarajasingam U, Vile RG, Gough MJ. Intratumoral immunotherapy: Using the tumour against itself. Immunology. 2005;114:11–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Melero I, Castanon E, Alvarez M, Champiat S, Marabelle A. Intratumoural administration and tumour tissue targeting of cancer immunotherapies. Nat Rev Clin Oncol. 2021;18:558–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Marabelle A, Tselikas L, Baere T, Houot R. Intratumoral immunotherapy: Using the tumor as the remedy. Ann Oncol. 2017;28:xii33–xii43. [DOI] [PubMed] [Google Scholar]
- 199.Le Louedec F, Leenhardt F, Marin C, Chatelut É, Evrard A, Ciccolini J. Cancer immunotherapy dosing: A pharmacokinetic/pharmacodynamic perspective. Vaccines. 2020;8(4):632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Maker AV, Yang JC, Sherry RM, Topalian SL, Kammula US, Royal RE, Hughes M, Yellin MJ, Haworth LR, Levy C, et al. Intrapatient dose escalation of anti-CTLA-4 antibody in patients with metastatic melanoma. J Immunother. 2006;29(4):455–463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Xu P, Liang F. Nanomaterial-based tumor photothermal immunotherapy. Int J Nanomedicine. 2020;15:9159–9180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Lu Y, Sun W, Du J, Fan J, Peng X. Immuno-photodynamic therapy (IPDT): Organic photosensitizers and their application in cancer ablation. JACS Au. 2023;3:682–699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Sordo-Bahamonde C, Lorenzo-Herrero S, Gonzalez-Rodriguez AP, Martínez-Pérez A, Rodrigo JP, García-Pedrero JM, Gonzalez S. Chemo-immunotherapy: A new trend in cancer treatment. Cancers. 2023;15(11):2912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Zhang Z, Liu X, Chen D, Yu J. Radiotherapy combined with immunotherapy: The dawn of cancer treatment. Signal Transduct Target Ther. 2022;7(1):258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Zhai J, Gu X, Liu Y, Hu Y, Jiang Y, Zhang Z. Chemotherapeutic and targeted drugs-induced immunogenic cell death in cancer models and antitumor therapy: An update review. Front Pharmacol. 2023;14:1152934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Liu J, Li Z, Zhao D, Feng X, Wang C, Li D, Ding J. Immunogenic cell death-inducing chemotherapeutic nanoformulations potentiate combination chemoimmunotherapy. Mater Des. 2021;202: Article 109465. [Google Scholar]
- 207.Li Z, Lai X, Fu S, Ren L, Cai H, Zhang H, Gu Z, Ma X, Luo K. Immunogenic cell death activates the tumor immune microenvironment to boost the immunotherapy efficiency. Adv Sci. 2022;9(22):2201734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Gupta G, Borglum K, Chen H. Immunogenic cell death: A step ahead of autophagy in cancer therapy. J Cancer Immunol. 2021;3(1):47–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Chang X, Bian M, Liu L, Yang J, Yang Z, Wang Z, Lu Y, Liu W. Induction of immunogenic cell death by novel platinum-based anticancer agents. Pharmacol Res. 2023;187: Article 106556. [DOI] [PubMed] [Google Scholar]
- 210.Zhang J, Sun X, Zhao X, Yang C, Shi M, Zhang B, Hu H, Qiao M, Chen D, Zhao X. Combining immune checkpoint blockade with ATP-based immunogenic cell death amplifier for cancer chemo-immunotherapy. Acta Pharm Sin B. 2022;12(9):3694–3709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Del Prete A, Salvi V, Soriani A, Laffranchi M, Sozio F, Bosisio D, Sozzani S. Dendritic cell subsets in cancer immunity and tumor antigen sensing. Cell Mol Immunol. 2023;20(5):432–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Garg AD, Agostinis P. Cell death and immunity in cancer: From danger signals to mimicry of pathogen defense responses. Immunol Rev. 2017;280(1):126–148. [DOI] [PubMed] [Google Scholar]
- 213.Palanivelu L, Liu C-H, Lin L-T. Immunogenic cell death: The cornerstone of oncolytic viro-immunotherapy. Front Immunol. 2022;13:1038226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Fucikova J, Kepp O, Kasikova L, Petroni G, Yamazaki T, Liu P, Zhao L, Spisek R, Kroemer G, Galluzzi L. Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis. 2020;11(11):1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Larson AC, Knoche SM, Brumfield GL, Doty KR, Gephart BD, Moore-Saufley PR, Solheim JC. Gemcitabine modulates HLA-I regulation to improve tumor antigen presentation by pancreatic cancer cells. Int J Mol Sci. 2024;25(6):3211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Zhu R, Su L, Dai J, Li Z-W, Bai S, Li Q, Chen X, Song J, Yang H. Biologically responsive plasmonic assemblies for second near-infrared window photoacoustic imaging-guided concurrent chemo-immunotherapy. ACS Nano. 2020;14(4):3991–4006. [DOI] [PubMed] [Google Scholar]
- 217.Zhai T, Zhong W, Gao Y, Zhou H, Zhou Z, Liu X, Yang S, Yang H. Tumor microenvironment-activated nanoparticles loaded with an iron-carbonyl complex for chemodynamic immunotherapy of lung metastasis of melanoma in vivo. ACS Appl Mater Interfaces. 2021;13(33):39100–39111. [DOI] [PubMed] [Google Scholar]
- 218.Zhang F, Chen F, Yang C, Wang L, Hu H, Li X, Zheng X, Wang Z, Chang Z, Li T, et al. Coordination and redox dual-responsive mesoporous organosilica nanoparticles amplify immunogenic cell death for cancer chemoimmunotherapy. Small. 2021;17(26):e2100006. [DOI] [PubMed] [Google Scholar]
- 219.Shao D, Zhang F, Chen F, Zheng X, Hu H, Yang C, Tu Z, Wang Z, Chang Z, Lu J, et al. Biomimetic diselenide-bridged mesoporous organosilica nanoparticles as an X-ray-responsive biodegradable carrier for chemo-immunotherapy. Adv Mater. 2020;32(50):e2004385. [DOI] [PubMed] [Google Scholar]
- 220.Sun K, Hu J, Meng X, Lei Y, Zhang X, Lu Z, Zhang L, Wang Z. Reinforcing the induction of immunogenic cell death via artificial engineered cascade bioreactor-enhanced chemo-immunotherapy for optimizing cancer immunotherapy. Small. 2021;17(37):2101897. [DOI] [PubMed] [Google Scholar]
- 221.Lhuillier C, Rudqvist N-P, Elemento O, Formenti SC, Demaria S. Radiation therapy and anti-tumor immunity: Exposing immunogenic mutations to the immune system. Genome Med. 2019;11(1):40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Asadzadeh Z, Safarzadeh E, Safaei S, Baradaran A, Mohammadi A, Hajiasgharzadeh K, Derakhshani A, Argentiero A, Silvestris N, Baradaran B. Current approaches for combination therapy of cancer: The role of immunogenic cell death. Cancers. 2020;12(4):1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Schildkopf P, Frey B, Mantel F, Ott OJ, Weiss E-M, Sieber R, Janko C, Sauer R, Fietkau R, Gaipl US. Application of hyperthermia in addition to ionizing irradiation fosters necrotic cell death and HMGB1 release of colorectal tumor cells. Biochem Biophys Res Commun. 2010;391(1):1014–1020. [DOI] [PubMed] [Google Scholar]
- 224.Yang W, Xiu Z, He Y, Huang W, Li Y, Sun T. Bip inhibition in glioma stem cells promotes radiation-induced immunogenic cell death. Cell Death Dis. 2020;11(9):786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Xi Y, Chen L, Tang J, Yu B, Shen W, Niu X. Amplifying “eat me signal” by immunogenic cell death for potentiating cancer immunotherapy. Immunol Rev. 2024;321(1):94–114. [DOI] [PubMed] [Google Scholar]
- 226.Reits EA, Hodge JW, Herberts CA, Groothuis TA, Chakraborty M, Wansley EK, Camphausen K, Luiten RM, Ru AH, Neijssen J, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. 2006;203(5):1259–1271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Storozynsky Q, Hitt MM. The impact of radiation-induced DNA damage on cGAS-STING-mediated immune responses to cancer. Int J Mol Sci. 2020;21(22):8877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Cadet J, Wagner JR. DNA base damage by reactive oxygen species, oxidizing agents, and UV radiation. Cold Spring Harb Perspect Biol. 2013;5(2):a012559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Chargari C, Rassy E, Helissey C, Achkar S, Francois S, Deutsch E, Chapter Three—Impact of radiation therapy on healthy tissues. Int Rev Cell Mol Biol. 2023;376:69–98. [DOI] [PubMed]
- 230.Bergerud KMB, Berkseth M, Pardoll DM, Ganguly S, Kleinberg LR, Lawrence J, Odde DJ, Largaespada DA, Terezakis SA, Sloan L. Radiation therapy and myeloid-derived suppressor cells: Breaking down their cancerous partnership. Int J Radiat Oncol Biol Phys. 2024;119(1):42–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Ostrand-Rosenberg S, Horn LA, Ciavattone NG. Radiotherapy both promotes and inhibits myeloid-derived suppressor cell function: Novel strategies for preventing the tumor-protective effects of radiotherapy. Front Oncol. 2019;9:215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Pei P, Shen W, Zhou H, Sun Y, Zhong J, Liu T, Yang K. Radionuclide labeled gold nanoclusters boost effective anti-tumor immunity for augmented radio-immunotherapy of cancer. Nano Today. 2021;38: Article 101144. [Google Scholar]
- 233.Kim DH, Choi B, Choi H, Yu B. Synergistic local combination of radiation and anti-programmed death ligand 1 immunotherapy using radiation-responsive splintery metallic nanocarriers. ACS Nano. 2020;14(10):13115–13126. [DOI] [PubMed] [Google Scholar]
- 234.Zhen W, Weichselbaum RR, Lin W. Nanoparticle-mediated radiotherapy remodels the tumor microenvironment to enhance antitumor efficacy. Adv Mater. 2023;35(21):e2206370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Overchuk M, Weersink RA, Wilson BC, Zheng G. Photodynamic and photothermal therapies: Synergy opportunities for nanomedicine. ACS Nano. 2023;17(9):7979–8003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Cheng X, Wei Y, Jiang X, Wang C, Liu M, Yan J, Zhang L, Zhou Y. Insight into the prospects for tumor therapy based on photodynamic immunotherapy. Pharmaceuticals. 2022;15(11):1359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Liu Z, Xie Z, Li W, Wu X, Jiang X, Li G, Cao L, Zhang D, Wang Q, Xue P, et al. Photodynamic immunotherapy of cancers based on nanotechnology: Recent advances and future challenges. J. Nanobiotechnology. 2021;19(1):160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Li J, Wang J, Zhang J, Han T, Hu X, Lee MMS, Wang D, Tang BZ. A facile strategy of boosting photothermal conversion efficiency through state transformation for cancer therapy. Adv Mater. 2021;33(51):e2105999. [DOI] [PubMed] [Google Scholar]
- 239.Wang P, Chen B, Zhan Y, Wang L, Luo J, Xu J, Zhan L, Li Z, Liu Y, Wei J. Enhancing the efficiency of mild-temperature photothermal therapy for cancer assisting with various strategies. Pharmaceutics. 2022;14(11):2279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Chen J, Ma Y, Du W, Dai T, Wang Y, Jiang W, Wan Y, Wang Y, Liang G, Wang G. Furin-instructed intracellular gold nanoparticle aggregation for tumor photothermal therapy. Adv Funct Mater. 2020;30(50):2001566. [Google Scholar]
- 241.Lei W, Sun C, Jiang T, Gao Y, Yang Y, Zhao Q, Wang S. Polydopamine-coated mesoporous silica nanoparticles for multi-responsive drug delivery and combined chemo-photothermal therapy. Mater Sci Eng C Mater Biol Appl. 2019;105: Article 110103. [DOI] [PubMed] [Google Scholar]
- 242.Xu Z, Wang T, Liu J. Recent development of polydopamine anti-bacterial nanomaterials. Int J Mol Sci. 2022;23(13):7278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Maharjan PS, Bhattarai HK. Singlet oxygen, photodynamic therapy, and mechanisms of cancer cell death. J Oncol. 2022;2022:7211485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Gulati LK, Gulati GK, Kumar S. Photochromic materials as a photosensitizer in reversible reactive singlet oxygen generation. Dyes Pigm. 2022;199: Article 110104. [Google Scholar]
- 245.Thakur NS, Patel G, Kushwah V, Jain S, Banerjee UC. Facile development of biodegradable polymer-based nanotheranostics: Hydrophobic photosensitizers delivery, fluorescence imaging and photodynamic therapy. J Photochem Photobiol B. 2019;193:39–50. [DOI] [PubMed] [Google Scholar]
- 246.Camerin M, Moreno M, Marín MJ, Schofield CL, Chambrier I, Cook MJ, Coppellotti O, Jori G, Russell DA. Delivery of a hydrophobic phthalocyanine photosensitizer using PEGylated gold nanoparticle conjugates for the in vivo photodynamic therapy of amelanotic melanoma. Photochem Photobiol Sci. 2016;15:618–625. [DOI] [PubMed] [Google Scholar]
- 247.Liu Y, Ma K, Jiao T, Xing R, Shen G, Yan X. Water-insoluble photosensitizer nanocolloids stabilized by supramolecular interfacial assembly towards photodynamic therapy. Sci Rep. 2017;7:42978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Huang H, Feng W, Chen Y, Shi J. Inorganic nanoparticles in clinical trials and translations. Nano Today. 2020;35: Article 100972. [Google Scholar]
- 249.Twomey JD, Zhang B. Cancer immunotherapy update: FDA-approved checkpoint inhibitors and companion diagnostics. AAPS J. 2021;23(2):39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Hu Y, Welsh J, Paris S, Barsoumian H, Abana C, Gandhi S, Nguyen Q, Cortez M. 200 NBTXR3 nanoparticle with immunoradiation improves survival and generates long-term anti-tumor memory in an anti-PD1 resistant murine lung cancer model. J Immunother Cancer. 2020;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Shen C, Frakes JM, Niu J, Rosenberg A, Weiss J, Caudell JJ, Jameson KL, Said P, Seiwert TY. A phase I trial evaluating NBTXR3 activated by radiotherapy in combination with nivolumab or pembrolizumab in patients with advanced cancers. J Clin Oncol. 2021;39:2590. [Google Scholar]
- 252.He X, Xu C. Immune checkpoint signaling and cancer immunotherapy. Cell Res. 2020;30(8):660–669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Wang L, Geng H, Liu Y, Liu L, Chen Y, Wu F, Liu Z, Ling S, Wang Y, Zhou L. Hot and cold tumors: Immunological features and the therapeutic strategies. MedComm. 2023;4(5): Article e343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Liu Y-T, Sun Z-J. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics. 2021;11(11):5365–5386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Cremolini C, Vitale E, Rastaldo R, Giachino C. Advanced nanotechnology for enhancing immune checkpoint blockade therapy. Nanomaterials. 2021;11(3):661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Song W, Jia P, Zhang T, Dou K, Liu L, Ren Y, Liu F, Xue J, Hasanin MS, Qi H, et al. Cell membrane-camouflaged inorganic nanoparticles for cancer therapy. J Nanobiotechnology. 2022;20(1):289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Vijayan V, Uthaman S, Park I-K. Cell membrane-camouflaged nanoparticles: A promising biomimetic strategy for cancer theragnostics. Polymers. 2018;10(9):983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Shang L, Zhou X, Zhang J, Shi Y, Zhong L. Metal nanoparticles for photodynamic therapy: A potential treatment for breast cancer. Molecules. 2021;26(21):6532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Farzam OR, Mehran N, Bilan F, Aghajani E, Dabbaghipour R, Shahgoli GA, Baradaran B. Nanoparticles for imaging-guided photothermal therapy of colorectal cancer. Heliyon. 2023;9(11): Article e21334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Liao Z, Huang J, Lo P-C, Lovell JF, Jin H, Yang K. Self-adjuvanting cancer nanovaccines. J Nanobiotechnology. 2022;20(1):345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Choi J, Kim SY. Photothermally enhanced photodynamic therapy based on glutathione-responsive pheophorbide a-conjugated gold nanorod formulations for cancer theranostic applications. J Ind Eng Chem. 2020;85:66–74. [Google Scholar]
- 262.Raggi F, Pelassa S, Pierobon D, Penco F, Gattorno M, Novelli F, Eva A, Varesio L, Giovarelli M, Bosco MC. Regulation of human macrophage M1-M2 polarization balance by hypoxia and the triggering receptor expressed on myeloid cells-1. Front Immunol. 2017;8:1097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Liu J, Li G, Guo H, Ni C, Gao Y, Cao X, Xia J, Shi X, Guo R. Dual-responsive core–shell tecto dendrimers enable efficient gene editing of cancer cells to boost immune checkpoint blockade therapy. ACS Appl Mater Interfaces. 2023;15(10):12809–12821. [DOI] [PubMed] [Google Scholar]
- 264.Liu H, Han J, Lv Y, Zhao Z, Zheng S, Sun Y, Sun T. Isorhamnetin and anti-PD-L1 antibody dual-functional mesoporous silica nanoparticles improve tumor immune microenvironment and inhibit YY1-mediated tumor progression. J Nanobiotechnology. 2023;21:208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Kang Y, Yan J, Han X, Wang X, Wang Y, Song P, Su X, Rauf A, Jin X, Pu F, et al. Construction of hierarchically biomimetic iron oxide nanosystems for macrophage repolarization-promoted immune checkpoint blockade of cancer immunotherapy. ACS Appl Mater Interfaces. 2024;16(28):36131–36141. [DOI] [PubMed] [Google Scholar]
- 266.Li J, Ren H, Sun Y, Liu G, Yang X, Qiu Q, Ding Y, Lovell JF, Zhang Y. Magnetic metal micelles for enhanced delivery of self-immolating CD8+ T-cell epitopes for cancer immunotherapy. Chem Mater. 2021;33(24):9780–9794. [Google Scholar]
- 267.Huang K-W, Hsu F-F, Qiu JT, Chern G-J, Lee Y-A, Chang C-C, Huang Y-T, Sung Y-C, Chiang C-C, Huang R-L, et al. Highly efficient and tumor-selective nanoparticles for dual-targeted immunogene therapy against cancer. Sci Adv. 2024;6(3):eaax5032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Zhao Z, Dong S, Liu Y, Wang J, Ba L, Zhang C, Cao X, Wu C, Yang P. Tumor microenvironment-activable manganese-boosted catalytic immunotherapy combined with PD-1 checkpoint blockade. ACS Nano. 2022;16(12):20400–20418. [DOI] [PubMed] [Google Scholar]
- 269.Hou L, Tian C, Yan Y, Zhang L, Zhang H, Zhang Z. Manganese-based nanoactivator optimizes cancer immunotherapy via enhancing innate immunity. ACS Nano. 2020;14(4):3927–3940. [DOI] [PubMed] [Google Scholar]
- 270.Zhang D, Wu T, Qin X, Qiao Q, Shang L, Song Q, Yang C, Zhang Z. Intracellularly generated immunological gold nanoparticles for combinatorial photothermal therapy and immunotherapy against tumor. Nano Lett. 2019;19(9):6635–6646. [DOI] [PubMed] [Google Scholar]
- 271.Choi J, Shim MK, Yang S, Hwang HS, Cho H, Kim J, Yun WS, Moon Y, Kim J, Yoon HY, et al. Visible-light-triggered prodrug nanoparticles combine chemotherapy and photodynamic therapy to potentiate checkpoint blockade cancer immunotherapy. ACS Nano. 2021;15:12086–12098. [DOI] [PubMed] [Google Scholar]
- 272.Cao F, Yan M, Liu Y, Liu L, Ma G. Photothermally controlled MHC class I restricted CD8+ T-cell responses elicited by hyaluronic acid decorated gold nanoparticles as a vaccine for cancer immunotherapy. Adv Healthc Mater. 2018;7(10):1701439. [DOI] [PubMed] [Google Scholar]
- 273.Shinchi H, Yamaguchi T, Moroishi T, Yuki M, Wakao M, Cottam HB, Hayashi T, Carson DA, Suda Y. Gold nanoparticles coimmobilized with small molecule toll-like receptor 7 ligand and α-mannose as adjuvants. Bioconjug Chem. 2019;30(11):2811–2821. [DOI] [PubMed] [Google Scholar]
- 274.Hou W, Wei P, Kong L, Guo R, Wang S, Shi X. Partially PEGylated dendrimer-entrapped gold nanoparticles: A promising nanoplatform for highly efficient DNA and siRNA delivery. J Mater Chem B. 2016;4(17):2933–2943. [DOI] [PubMed] [Google Scholar]
- 275.Feng Q, Xiao K. Nanoparticle-mediated delivery of STAT3 inhibitors in the treatment of lung cancer. Pharmaceutics. 2022;14(12):2787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Ruan S, Xie R, Qin L, Yu M, Xiao W, Hu C, Yu W, Qian Z, Ouyang L, He Q, et al. Aggregable nanoparticles-enabled chemotherapy and autophagy inhibition combined with anti-PD-L1 antibody for improved glioma treatment. Nano Lett. 2019;19(11):8318–8332. [DOI] [PubMed] [Google Scholar]
- 277.Cha BG, Jeong JH, Kim J. Extra-large pore mesoporous silica nanoparticles enabling co-delivery of high amounts of protein antigen and toll-like receptor 9 agonist for enhanced cancer vaccine efficacy. ACS Cent Sci. 2018;4(4):484–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Wang X, Li X, Ito A, Sogo Y, Watanabe Y, Hashimoto K, Yamazaki A, Ohno T, Tsuji NM. Synergistic effects of stellated fibrous mesoporous silica and synthetic dsRNA analogues for cancer immunotherapy. Chem Commun. 2018;54(9):1057–1060. [DOI] [PubMed] [Google Scholar]
- 279.Kim J, Li WA, Choi Y, Lewin SA, Verbeke CS, Dranoff G, Mooney DJ. Injectable, spontaneously assembling, inorganic scaffolds modulate immune cells in vivo and increase vaccine efficacy. Nat Biotechnol. 2015;33(1):64–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Stater EP, Morcos G, Isaac E, Ogirala A, Hsu H-T, Longo VA, Grimm J. Translatable drug-loaded iron oxide nanophore sensitizes murine melanoma tumors to monoclonal antibody immunotherapy. ACS Nano. 2023;17(7):6178–6192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Xu L, Xiang J, Liu Y, Xu J, Luo Y, Feng L, Liu Z, Peng R. Functionalized graphene oxide serves as a novel vaccine nano-adjuvant for robust stimulation of cellular immunity. Nanoscale. 2016;8(6):3785–3795. [DOI] [PubMed] [Google Scholar]
- 282.Xu C, Hong H, Lee Y, Park KS, Sun M, Wang T, Aikins ME, Xu Y, Moon JJ. Efficient lymph node-targeted delivery of personalized cancer vaccines with reactive oxygen species-inducing reduced graphene oxide nanosheets. ACS Nano. 2020;14(10):13268–13278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Qiang N, Wei L, Tao Y, Jin W, Bin Y, DingHua Z. Construction of Durvalumab/carbon nanotube/PEI/aptamer-siRNA chimera for the immunotherapy of hepatocellular carcinoma. Biomed Mater. 2022;17(2):025015. [DOI] [PubMed] [Google Scholar]
- 284.Li D, Ahmed M, Khan A, Xu L, Walters AA, Ballesteros B, Al-Jamal KT. Tailoring the architecture of cationic polymer brush-modified carbon nanotubes for efficient siRNA delivery in cancer immunotherapy. ACS Appl Mater Interfaces. 2021;13(26):30284–30294. [DOI] [PubMed] [Google Scholar]
- 285.Liu H, Xie Z, Zheng M. Carbon dots and tumor antigen conjugates as nanovaccines for elevated cancer immunotherapy. Small. 2023;19(31):2206683. [DOI] [PubMed] [Google Scholar]
- 286.Chen P, He X, Hu Y, Tian X-L, Yu X-Q, Zhang J. Spleen-targeted mRNA delivery by amphiphilic carbon dots for tumor immunotherapy. ACS Appl Mater Interfaces. 2023;15(16):19937–19950. [DOI] [PubMed] [Google Scholar]
- 287.Su W, Tan M, Wang Z, Zhang J, Huang W, Song H, Wang X, Ran H, Gao Y, Nie G, et al. Targeted degradation of PD-L1 and activation of the STING pathway by carbon-dot-based PROTACs for cancer immunotherapy. Angew Chem Int Ed Engl. 2023;62(11): Article e202218128. [DOI] [PubMed] [Google Scholar]
- 288.Zhao J, Chen G, Pang X, Zhang P, Hou X, Chen P, Xie Y-W, He C-Y, Wang Z, Chen Z-Y. Calcium phosphate nanoneedle based gene delivery system for cancer genetic immunotherapy. Biomaterials. 2020;250: Article 120072. [DOI] [PubMed] [Google Scholar]
- 289.Anoop M, Nambiar AR, Nair SV, Koyakutty M, Ashokan A. Zoledronic acid conjugated calcium phosphate nanoparticles for applications in cancer immunotherapy. Mater Today Commun. 2022;30: Article 103065. [Google Scholar]
- 290.Zhou M, Liang S, Liu D, Ma K, Yun K, Yao J, Peng Y, Hai L, Zhang Q, Wang Z. Manganese-enriched zinc peroxide functional nanoparticles for potentiating cancer immunotherapy. Nano Lett. 2023;23(22):10350–10359. [DOI] [PubMed] [Google Scholar]
- 291.Luo G, Li X, Lin J, Ge G, Fang J, Song W, Xiao GG, Zhang B, Peng X, Duo Y, et al. Multifunctional calcium–manganese nanomodulator provides antitumor treatment and improved immunotherapy via reprogramming of the tumor microenvironment. ACS Nano. 2023;17(16):15449–15465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Mu Q, Lin G, Jeon M, Wang H, Chang F-C, Revia RA, Yu J, Zhang M. Iron oxide nanoparticle targeted chemo-immunotherapy for triple negative breast cancer. Mater Today. 2021;50:149–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.Hou L, Chen D, Wang R, Wang R, Zhang H, Zhang Z, Nie Z, Lu S. Transformable honeycomb-like nanoassemblies of carbon dots for regulated multisite delivery and enhanced antitumor chemoimmunotherapy. Angew Chem Int Ed Engl. 2021;60(12):6581–6592. [DOI] [PubMed] [Google Scholar]
- 294.Bao Y, Li G, Li S, Zhang H, Wu X, Yan R, Wang Z, Guo C, Jin Y. Multifunctional tumor-targeting carbon dots for tumor microenvironment activated ferroptosis and immunotherapy in cancer treatment. ACS Appl Mater Interfaces. 2023;15(49):56834–56845. [DOI] [PubMed] [Google Scholar]
- 295.Kumar S, Mongia A, Gulati S, Singh P, Diwan A, Shukla S. Emerging theranostic gold nanostructures to combat cancer: Novel probes for combinatorial immunotherapy and photothermal therapy. Cancer Treat Res Commun. 2020;25: Article 100258. [DOI] [PubMed] [Google Scholar]
- 296.Wang M, Li Y, Wang M, Liu K, Hoover AR, Li M, Towner RA, Mukherjee P, Zhou F, Qu J, et al. Synergistic interventional photothermal therapy and immunotherapy using an iron oxide nanoplatform for the treatment of pancreatic cancer. Acta Biomater. 2022;138:453–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297.Gao Y, Chen X, Wang B, Wang S, Wang J, Ren L, Jin W, Han H, Wang L. Engineering platelets with PDL1 antibodies and iron oxide nanoparticles for postsurgical cancer immunotherapy. ACS Appl Bio Mater. 2023;6(1):257–266. [DOI] [PubMed] [Google Scholar]
- 298.Yuan Z, He H, Zou J, Wang H, Chen Y, Chen Y, Lan M, Zhao Y, Gao F. Polydopamine-coated ferric oxide nanoparticles for R848 delivery for photothermal immunotherapy in breast cancer. Int J Pharm. 2023;644: Article 123249. [DOI] [PubMed] [Google Scholar]
- 299.Tao Y, Ju E, Ren J, Qu X. Immunostimulatory oligonucleotides-loaded cationic graphene oxide with photothermally enhanced immunogenicity for photothermal/immune cancer therapy. Biomaterials. 2014;35(37):9963–9971. [DOI] [PubMed] [Google Scholar]
- 300.Zhou F, Wang M, Luo T, Qu J, Chen WR. Photo-activated chemo-immunotherapy for metastatic cancer using a synergistic graphene nanosystem. Biomaterials. 2021;265: Article 120421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Liu S, Zhang M, Yu H, Sun X, Li Q, Yang M, Qiu X, Su H, Gong A, Du F. Immunoinducible carbon dot-incorporated hydrogels as a photothermal-derived antigen depot to trigger a robust antitumor immune response. ACS Appl Mater Interfaces. 2023;15(6):7700–7712. [DOI] [PubMed] [Google Scholar]
- 302.Li Z, Chu Z, Yang J, Qian H, Xu J, Chen B, Tian T, Chen H, Xu Y, Wang F. Immunogenic cell death augmented by manganese zinc sulfide nanoparticles for metastatic melanoma immunotherapy. ACS Nano. 2022;16(9):15471–15483. [DOI] [PubMed] [Google Scholar]
- 303.Chen Y, Ma H, Wang W, Zhang M. A size-tunable nanoplatform: Enhanced MMP2-activated chemo-photodynamic immunotherapy based on biodegradable mesoporous silica nanoparticles. Biomater Sci. 2021;9(3):917–929. [DOI] [PubMed] [Google Scholar]
- 304.Wu X, Cheng Y, Zheng R, Xu K, Yan J, Song P, Wang Y, Rauf A, Pan Y, Zhang H. Immunomodulation of tumor microenvironment by arginine-loaded iron oxide nanoparticles for gaseous immunotherapy. ACS Appl Mater Interfaces. 2021;13(17):19825–19835. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data availability is not applicable to this article as no new data were created or analyzed in this study.