Next Article in Journal
Rational Design of Topical Semi-Solid Dosage Forms-How Far Are We?
Next Article in Special Issue
In Vitro and Preclinical Antitumor Evaluation of Doxorubicin Liposomes Coated with a Cholesterol-Based Trimeric β-D-Glucopyranosyltriazole
Previous Article in Journal
Development of an Alcohol Dilution–Lyophilization Method for the Preparation of mRNA-LNPs with Improved Storage Stability
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biomimetic Cell-Derived Nanoparticles: Emerging Platforms for Cancer Immunotherapy

1
Department of Pharmacy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
2
Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceutics 2023, 15(7), 1821; https://doi.org/10.3390/pharmaceutics15071821
Submission received: 2 June 2023 / Revised: 23 June 2023 / Accepted: 23 June 2023 / Published: 26 June 2023
(This article belongs to the Special Issue Lipid-Based Nanoparticles for Drug Delivery in Cancer)

Abstract

:
Cancer immunotherapy can significantly prevent tumor growth and metastasis by activating the autoimmune system without destroying normal cells. Although cancer immunotherapy has made some achievements in clinical cancer treatment, it is still restricted by systemic immunotoxicity, immune cell dysfunction, cancer heterogeneity, and the immunosuppressive tumor microenvironment (ITME). Biomimetic cell-derived nanoparticles are attracting considerable interest due to their better biocompatibility and lower immunogenicity. Moreover, biomimetic cell-derived nanoparticles can achieve different preferred biological effects due to their inherent abundant source cell-relevant functions. This review summarizes the latest developments in biomimetic cell-derived nanoparticles for cancer immunotherapy, discusses the applications of each biomimetic system in cancer immunotherapy, and analyzes the challenges for clinical transformation.

1. Introduction

Cancer immunotherapy, including immune checkpoint inhibitors (ICIs), adoptive cellular immunotherapy (ACT), cancer vaccines, and cytokine immunotherapy, is an effective treatment method that recognizes and kills cancer cells by activating the autoimmune system [1]. Although cancer immunotherapy has made some achievements in clinical cancer treatment, it is still restricted by systemic immunotoxicity, immune cell dysfunction, cancer heterogeneity, and the ITME [2,3]. In addition, how to accurately deliver therapeutic agents to tumors for enhanced immune responses and reduced systemic immunotoxicity is one of the urgent issues to be solved.
In the past decades, nanoparticles have gradually emerged as promising drug delivery carriers for chemotherapy, photothermal therapy (PTT), gene therapy, and immunotherapy due to their good design flexibility, reduction of side effects, and improvement of in vivo efficacy [4,5,6,7]. However, owing to the complexity of the blood environment in vivo, nanoparticles with high immunogenicity can be easily identified and removed by the mononuclear phagocytic system [8,9]. Engineered nanoparticles show promise for improving the biodistribution and metabolism of nanoparticles. For instance, coating nanoparticles with PEG can prolong their circulation half-life in the blood [10,11]. Nevertheless, long-term repeated PEGylated nanoparticles treatment can, in turn, accelerate their clearance and trigger more intense immunogenic stimulation [12,13]. Therefore, more effective solutions are needed. To address these issues, the application of biomimetic cell-derived nanoparticles is a promising strategy [14,15,16].
In the field of cancer diagnosis and therapy, nanoparticles coated with cell membranes combine the biological functions of cell membranes with the drug-loading capability of engineered core nanoparticles, thereby providing a series of unique advantages (Figure 1), including improved biological interface properties, lower immunogenicity, better biocompatibility, longer circulation, more efficient drug delivery, and elevated active-targeting [17,18,19,20]. Therefore, biomimetic cell-derived nanoparticles have been applied to the precision delivery of theranostic agents or to directly improve the therapeutic efficacy of cancer immunotherapy.
In this review, we underline the unique advantages of biomimetic cell-derived nanoparticles, summarize the latest developments in biomimetic cell-derived nanoparticles for cancer immunotherapy, and analyze the challenges of clinical translation of biomimetic cell-derived nanoparticles.

2. Biomimetic Cell-Derived Nanoparticles

Biomimetic cell-derived nanoparticles are synthetic nanoparticles camouflaged with natural or engineered cell membrane materials to trick the immune system and enhance tumor targeting [21,22]. The structure of biomimetic cell-derived nanoparticles is a core–shell structure, where the core is the nanoparticles delivering the therapeutic agents to the target site and the shell is the membrane materials extracted from different cells. These resulting biomimetic nanoparticles combine the physical and chemical properties of nanoparticles with the intrinsic properties of natural cell membranes, possessing the capacity to evade the immune system, prolong blood circulation, and actively target diagnostic and therapeutic agents to the targeted sites [17,18,19,20]. In 2011, Hu et al. constructed erythrocyte membrane-camouflaged nanoparticles by co-extruding PLGA polymeric nanoparticles and erythrocyte membrane-derived vesicles [23]. This is the first report of nanoparticles derived from cell membranes. Since then, researchers have designed various biomimetic cell-derived nanoparticles to their meet desired functions, and flexibly combined different types of nanoparticles with different sources of biomimetic membrane materials [24,25,26,27], such as cancer cell membranes, white blood cell or leukocyte membranes, stem cell membranes, platelet membranes, and bacteria membranes. The biomimetic cell-derived nanoparticles have been widely recognized for a variety of applications in drug delivery, disease diagnosis, immune modulation, and disease treatment [28,29,30].

2.1. Unique Function of Biomimetic Cell-Derived Nanoparticles

Biomimetic cell-derived nanoparticles inherit abundant important biological functions related to their source cells, including “self” labeling, biological targeting, cross-talk with the immune system, and region-specific homing. Most of these important functions are attributed to specific proteins or molecules on the surface of cell membranes.

2.1.1. Erythrocyte Membranes

Erythrocytes can prolong the systemic circulation time of wrapped nanoparticles due to some special membrane proteins [31], and the most important biomarker is CD47. Briefly, signal-regulatory protein alpha (SIRPα) on the surface of phagocytes interacts with CD47 expressed by erythrocytes, which disguises the immune cells as self and prevents immune phagocytes from phagocytizing erythrocytes [32]. In addition, CD59, C8 binding protein (C8bp), and complement receptor 1 (CR1) on the surface of erythrocytes have a role in defending against complement system attack [33]. Therefore, nanoparticles coated with erythrocyte membranes have a prolonged circulation half-life and are less immunogenic. Nevertheless, erythrocyte membranes have no tumor-targeting properties. To remedy this deficiency, researchers further utilize tumor-targeting ligands/peptides to modify erythrocyte membranes or construct erythrocyte membrane-based hybrid membranes to realize tumor targeting [34,35,36,37].

2.1.2. Immune Cell Membranes

Tumors are chronic inflammatory tissues that attract and recruit immune cells by secreting a variety of chemokines and cytokines [38,39]. Therefore, the adhesive property of immune cells can be exploited to actively target drugs for cancer treatment [40].
Dendritic cells (DCs) are professional antigen presenting cells (APCs). Thus, mature DC membrane-wrapped nanoparticles can thoroughly inherit the antigen-presenting function of DCs [41]. With this advantage, mature dendritic cell membrane-wrapped nanoparticles can specifically activate T cells due to the peptide/major histocompatibility complex (MHC) complexes on the surface of biomimetic nanoparticles [42]. In addition, costimulatory molecules and adhesion molecules on DC membranes, including integrins, CD44, CD40, and ICAM-3, can facilitate the interaction between T cells and DCs [43].
Macrophages can accumulate in the tumor microenvironment owing to some adhesion molecules and specific receptors, such as intercellular adhesion molecule-1, C-C chemokine receptor 2, and vascular cell adhesion molecule-1 (VCAM-1) [43]. In addition, macrophages can achieve active tumor targeting owing to the interaction between α4 integrins on macrophage membranes and VCAM-1 on tumor cell membranes [44].
T cells or cytotoxic T cells are also a subtype of leukocytes that can kill tumor cells. T cells have various properties, such as searching for antigens in the systemic circulation, activating cytolysis by recognizing a single peptide, and producing interferon-γ (IFN-γ, a cytokine with multiple antitumor properties), which make them attractive mediators of antitumor immunity [45,46]. Additionally, cytotoxic T cells can promote tumor cell apoptosis mediated by granule and receptors [47]. In addition, TCR complexes expressed on T cells can specifically bind to tumor-associated antigens with high affinity, which makes T cells more specific in tumor-targeting [48,49].
Natural killer (NK) cells play an important role in the recognition and killing of tumor cells owing to their innate capacity to monitor the abnormal expression of stress proteins and MHC-I [50]. Although NK cells lack tumor antigen-specific receptors on their surface, they have some alternative receptors that can recognize tumor cells, such as DNAM-1, NKG2D, and NKp46 [51,52]. Therefore, NK cell membrane-wrapped nanoparticles possess good tumor-targeting ability.

2.1.3. Platelet Membranes

Similar to erythrocytes, platelet cell membrane-wrapped nanoparticles also have a prolonged blood circulation time and are less immunogenic due to their reduced immunogenicity. Firstly, CD47 expressed on the surface of platelets can inhibit the uptake of platelet cell membrane-wrapped nanoparticles by macrophages [53]. Moreover, CD55 and CD59 expressed on the surface of platelet membranes, along with CD47, can avoid immune complement system attack [54,55]. In addition, P-selectin is overexpressed on the surface of platelets, which allows platelet membrane-derived nanoparticles to specifically bind to the CD44 receptors expressed on tumor cells [56]. Thus, nanoparticles can achieve aggressive tumor targeting and long circulation capability through platelet membrane coating.

2.1.4. Cancer Cell Membranes

Cancer cell membrane-camouflaged nanoparticles can enable immune escaping, prolong systemic circulation, and target homotypic tumors owing to a series of membrane proteins expressed by cancer cells [57,58]. CD47 on the tumor cell surface plays an important role in immune escape, especially in 4T1, MDA-MB-231, and MCF-7 [59,60,61,62]. Thomsen–Friedenreich antigen, E-cadherin, Galectin-3, N-cadherin, and epithelial cell adhesion molecules on the surface of tumor cells are essential for homologous targeting and adhesion [63,64,65]. Therefore, the application of cancer cell membranes for nanoparticle surface coating has the unique advantages of inherent immune escaping and homologous targeting and adhesion.

2.1.5. Stem Cell Membranes

Stem cell membranes are easy to isolate and have a various of molecular recognition sites, which can be used in biomimetic nanoparticles [66,67]. Chemokine receptors on mesenchymal stem cell membranes respond to ligand molecules on tumor cells, promoting the migration of mesenchymal stem cells to the tumor [68]. Moreover, P-selectin, E-selectin, and TGF-β expressed on the membrane of mesenchymal stem cells also influence the tumor tropism of mesenchymal stem cells [68,69]. Therefore, mesenchymal stem cell membrane-wrapped nanoparticles have also attracted increasing attention owing to their good biocompatibility, prolonged circulation time, and tumor targeting.

2.1.6. Bacteria Membranes

In the 1890s, William Coley used toxins made from attenuated bacteria to activate the anti-tumor immune system [70]. This is the first report of bacteria being used in cancer immunotherapy. Outer membrane vesicles (OMVs) produced by Gram-negative bacteria are composed of lipid bilayers and inherit various parent bacteria-derived components, such as enzymes, bacteria antigens, adhesins, and a variety of pathogen-associated molecular patterns (PAMPs) [71,72,73]. Among them, bacteria-derived antigens and PAMPs play a vital role in inducing the humoral and cellular anti-tumor immune responses [74,75]. Taking the advantages of OMVs, researchers have attempted to construct bacterial membrane-derived nanoparticles for cancer immunotherapy [76].

2.1.7. Extracellular Vesicles

Extracellular vesicles (EVs) are a group of nanoscale membrane-bound vesicles secreted by almost all eukaryotic cells [77,78]. The surface of EVs is rich in a variety of transmembrane proteins, such as ICAM-1, integrin, and tetraspanin, which give EVs the ability to target specific tissues or cells [79]. In addition, compared with traditional drug delivery platforms, EVs have excellent biocompatibility, low immunogenicity, phagocytosis avoidance, controllable biological characteristics, and the potential to cross natural barriers such as the blood–brain barrier [80]. Especially, EVs with immunomodulatory capacities, such as DC-derived EVs, NK-derived EVs, and T cell-derived EVs, can be used as effective therapeutics for cancer immunotherapy [81,82].

2.1.8. Hybrid Cell Membranes

Compared with single-cell membranes, hybrid cell membranes not only retain the physical and chemical characteristics of nanoparticles, but also endow nanoparticles with the biological functions of two or more derived cells, which makes biomimetic cell-derived nanoparticles more attractive [83]. Hybrid cell membrane-camouflaged nanoparticles can enhance the flexibility of nanoparticle functionality and thereby achieve better anti-tumor effects [84,85,86,87].

2.2. Fabrication of Biomimetic Cell-Derived Nanoparticles

The fabrication of biomimetic cell-derived nanoparticles mainly includes: (1) isolation and construction of parent cell membrane-derived vesicles and (2) fusion of the parent cell membrane with nanoparticle cores (Figure 2).

2.2.1. Isolation and Preparation of Parent Cell Membrane-Derived Vesicles

The isolation of cell membranes should be gentle in order to obtain biologically active membrane vesicles and usually involves cell lysis and membrane purification. Anucleated cells, such as platelets and erythrocytes, are isolated from whole blood by centrifugation. After that, the collected cells are lysed via repeated freeze–thaw or hypotonic treatment. Purified membranes are obtained by removing soluble proteins by centrifugation. Finally, the purified membranes are extruded through a polycarbonate porous membrane to gain parent cell membrane-derived vesicles [23,54,55]. Bacteria are coated with cell membranes and peptidoglycans, making membrane extraction more difficult. Luckily, Gram-negative bacteria can naturally produce OMVs, which can be directly separated from their culture via ultrafiltration [88,89]. Compared with anucleated cells, the extraction and purification of parent membranes from eukaryotic cells, such as stem cells and leukocytes, is more complicated. First, we have to collect source cells from the tissue, blood, or culture medium. Then, a variety of methods are used for cytolysis, including hypotonic solution treatment, repeated freeze–thaw, and/or mechanical rupture. Additionally, the obtained membranes are purified by discontinuous sucrose gradient centrifugation to remove nuclei, intracellular vesicles, and intracellular biomacromolecules [90,91].

2.2.2. Fusion of Parent Cell Membrane-Derived Vesicles with Nanoparticle Cores

Membrane extrusion and ultrasonic and microfluidic electroporation are commonly used methods to facilitate the coating of parent cell membrane-derived vesicles onto nanoparticle cores [2,14]. Mechanical extrusion is one of the commonly used methods reported in the literature. Briefly, the membrane vesicles and nanoparticles are extruded through the polycarbonate porous membrane with progressively smaller pore sizes. The fusion of vesicles and particles is achieved by applying mechanical forces to facilitate the passage of nanoparticles through the lipid bilayer of the membrane vesicles [92]. This approach can largely ensure the bioactivity of membrane proteins. Nevertheless, it is a time-consuming process [93]. Ultrasound is another effective method. When the nanoparticles and membrane vesicles are mixed together, the membranes surrounding the nanoparticles are reassembled by sonication. Compared to the extrusion method, this method is time-saving. However, the parameters of the ultrasound apparatus, including power, frequency, and duration, need to be optimized to guarantee fusion efficiency while avoiding protein inactivation. Microfluidic electroporation can also be utilized for coating different types of nanoparticles. Electromagnetic energy forms holes in cell membranes by a microfluidic chip, thereby promoting membrane vesicles to wrap nanoparticle cores [94]. In this process, the parameters also need to be optimized, such as pulse voltage, flow rate, and duration. The biomimetic cell-derived nanodrugs constructed by this method are completely coated, uniformly distributed, and highly reproducible. However, this technology comes at a high cost. In contrast to nanoparticle-templated membrane coating, Zhang and his colleagues reported a simple method to synthesize cell membrane-wrapped nanogels by cell membrane-templated polymerization [95]. Unlike the previously mentioned approaches, this new strategy uses membrane vesicles to ‘guide’ the growth of nanoparticle cores, thereby surmounting the “coatability” limitation of nanoparticles, as well as adding elevated controllability for the application of biomimetic cell-derived nanoparticles. The key challenge for this technique is how to efficiently and precisely inhibit the polymerization reaction outside the vesicles while maintaining the reaction activity inside the vesicles.

3. Application of Biomimetic Cell-Derived Nanoparticles in Cancer Immunotherapy

Considering the outstanding advantages, biomimetic cell-derived nanoparticles have been extensively investigated for cancer immunotherapy. For example, they can be used to deliver immunotherapeutic drugs and immune adjuvants to tumors or improve the effectiveness of cancer immunotherapy by reversing the ITME. In the following, we present the application of various biomimetic cell-derived nanoparticles in ICIs, ACT, cancer vaccines, modulating of the ITME, and combination therapy.

3.1. Immune Checkpoint Inhibitors

Immune checkpoint inhibitors (ICIs), such as programmed death ligand 1/programmed death protein 1 (PD-L1/PD-1) inhibitors, have shown efficacy in anti-cancer immune responses in various cancers [96,97]. However, the clinical use of ICIs is largely limited because only 10 to 30% of cancer patients respond positively to ICIs [98]. To improve the efficiency of ICIs, biomimetic cell-derived nanodrug delivery systems may have great potential.
Recent studies have found that some cancer cells release a mass of exosomes (named TEXs), which carry a large number of PD-L1s on their surface [99]. Moreover, exosomes expressing PD-L1 can competitively bind and exhaust PD-L1 Ab (aPD-L1), leading to drug resistance to aPD-L1 [100,101]. Thus, suppression of exosomes secreted by tumor cells can be a powerful anti-cancer strategy to improve the effectiveness of current aPD-L1. Herein, Yan and colleagues developed self-adaptive platelet cell membrane-wrapped nanoparticles to enable cascaded delivery of exosome-inhibiting siRNA (siRab) and aPD-L1, resulting in a robust anti-tumor immune response [102]. In their study, siRab effectively suppressed the production of TEXs, alleviated immunosuppression, and enhanced cytotoxic T lymphocyte infiltration to facilitate the on-demand release of aPD-L1 by biomimetic nanoparticles. As a result, the competitive exhaustion of aPD-L1 by TEXs was restrained. Additionally, siRab-mediated reversal of ITME combined with aPD-L1-based immunotherapy induced a robust anti-tumor response and immune memory.
Cancer cells secrete small extracellular vesicles (sEVs) to promote tumor progression, but conversely, immune cells secrete sEVs to prevent tumor progression [103,104,105]. Researchers have found that sEVs secreted by immune cells play a crucial role in anti-cancer immune responses by participating in the interaction between innate immunity and adaptive immunity [103,106]. Herein, Jung and colleagues generated interleukin-2-anchored T cell sEVs (IL2-sEVs) by attaching IL2 to the surface of T cells [107]. In their study, self-stimulation of T cells by IL2 altered the microRNA (miRNA) profile of T cell-derived sEVs. Among them, miR-223-3p and miR-181a-3p distinctly inhibited the sEV secretion and PD-L1 expression in melanoma cells, leading to an elevated immune response. Moreover, IL2-sEVs notably enhanced the therapeutic efficacy of aPD-L1 by reducing PD-L1 expression in vivo (Figure 3).
The accumulation of hypoxia-inducible factor-1α (HIF-1α) in the hypoxic tumor microenvironment has been confirmed to activate the downstream CD73–adenosine (CD73-ADO) pathway and lead to effector T cell exhaustion [108], which is a key disadvantage for the poor clinical efficacy of ICIs treatment [109]. Therefore, it is of great value to alleviate tumor hypoxia and block the CD73-ADO pathway to enhance the therapeutic effect of ICIs. Herein, Yuan and colleagues constructed cancer cell membrane-wrapped and matrix metallopeptidase-sensitive nanoparticles (CSG@B16F10) to co-deliver oxygen-generating agent catalase (CAT) and CD73siRNA, thus alleviating hypoxia and reshaping T cell exhaustion caused by the CD73-ADO pathway [110]. In their study, CAT improved cancer hypoxia by producing abundant endogenous oxygen, while CD73siRNA efficiently inhibited the expression of the target gene, synergistically down-regulating the level of CD73 and promoting the T cell-specific immune response. Moreover, CSG@B16F10 significantly enhanced the tumor immunotherapy efficacy and response rate of aPD-L1 by alleviating hypoxia and reversing the immunosuppressive microenvironment in vivo.
In another study, Li and colleagues also designed an intelligent biomimetic drug delivery system (mEHGZ) to convert an immunosuppressive microenvironment to an immunoresponsive one, which ultimately enhanced the sensitivity of aPD-L1 immunotherapy [111]. Different from alleviating tumor hypoxia, mEHGZ mainly amplified the generation of immunogenic cell death (ICD) by triggering a cascade reaction for reactive oxygen species (ROS) production, thereby enhancing the sensitivity of immunosuppressed tumors to aPD-L1. After cellular uptake of mEHGZ, a Fenton reaction was triggered by released hemin and glucose oxidase to facilitate ROS generation and increase endoplasmic reticulum stress, which collectively amplified the ICD effect. Furthermore, the induced powerful ICD effect promoted DC maturation and cytotoxic T lymphocyte infiltration, thereby activating the ITME. mEHGZ combined with aPD-L1 significantly inhibited tumor progression and lung metastasis in vivo, indicating that a robust ICD could substantially improve the efficacy of aPD-L1.
Additionally, a summary of biomimetic cell-derived nanoparticles applied in ICIs is displayed in Table 1.

3.2. Adoptive Cellular Immunotherapy

Adoptive cellular immunotherapy (ACT) refers to the in vivo transfusion of immune effector cells activated in vitro to activate the patients’ tumor-specific immune responses or to directly kill tumors. Despite the great potential of ACT to activate the anti-tumor immune responses, its clinical application is severely hampered by disadvantages such as off-target effects, poor infiltration, T-cell exhaustion, in vitro expansion and engineering, severe side effects, and great costs [124,125,126]. Therefore, there is a high demand for the construction of ACT mimics that can conquer the challenges mentioned above using some effective methods, and biomimetic cell-derived nanodrug delivery systems may be a good option.
Kang and colleagues constructed T cell-wrapped nanoparticles (TCMNPs) as a mimicry of CTLs to reactivate the exhausted T cells induced by the ITME [127]. Similar to CTLs, TCMNPs actively targeted tumors via adhesion proteins and directly eliminated tumor cells by releasing dacarbazine and triggering Fas-ligand-induced apoptosis. Unlike CTLs, TCMNPs were free from immunosuppressive cytokines and PD-L1 on cancer cells by blocking TGF-β1 and PD-L1 via TGF-β1 receptors or PD-1 proteins on TCMNPs, which ultimately restored the cytotoxic functions of exhausted T cells. Indeed, the significant inhibitory effect of TCMNPs on melanoma was attributed to the synergistic effect of chemotherapy, TGF-β blocking, and the PD-1/PD-L1 signaling blockade. In another study, Hong and colleagues constructed CD8+ T cell-derived exosomes (TCNVs) that also effectively prevented T-cell exhaustion and reversed ITME in the same way as TCMNPs [128]. Furthermore, TCNVs directly induced apoptosis via the delivery of granzyme B to tumor cells.
Compared with T cell-based adoptive immunotherapy, NK cells can directly kill tumor cells without causing graft-versus-host disease due to immunocompatibility, making them suitable for a variety of allogeneic and situations [129]. Nevertheless, the number of NK cells in the blood is relatively small, and the cytotoxic effect is relatively weak, which cannot meet the needs of current clinical treatment [130]. Herein, Wu and colleagues designed cancer cell membrane-wrapped magnetic nanoparticles (CM-Fe3O4@SiO2, CMNPs) for stimulating NK cells and enhancing NK cell-based immunotherapy [131]. In their study, CMNPs enhanced the expression of surface-activating hallmark receptors on NK cells and facilitated the production of cytotoxic cytokines such as granzyme and perforin, resulting in enhanced NK cell-mediated immunotherapy. In another study, Gong and colleagues constructed nanobody 7D12-engineered NK cells (7D12-NK92MI) through a glycoengineering approach to improve NK cell-based immunotherapy in EGFR-overexpressing solid tumors [132]. The obtained 7D12-NK92MI exhibited high targeting and affinity to EGFR-positive cancer cells, resulting in good tissue penetration and enhanced secretion of cytotoxic cytokines such as enzyme B, IL-2, and IFN-γ. In addition, 7D12-NK92MI exhibited more significant immunotherapy efficacy in EGFR-positive cancer cell-bearing mice.
DC-based ACT is another alternative treatment for T cell-mediated immunotherapy [133]. Nevertheless, the ITME can trigger the differentiation of tolerogenic DCs, which in turn induces the apoptosis of CD8+ T cells or activates regulatory T cells, eventually leading to tumor immune escape [134]. Herein, Sun and colleagues constructed smart DCs (iDCs) to restart the cancer immunity cycle by coating IR-797-loaded nanoparticles with mature DC membranes [135]. The obtained iDCs could effectively cross-prime T cells to secrete cytokines and migrate to lymph nodes to activate the initial T cell. Then, the activated T cells up-regulated the levels of heat shock proteins in cancer cells, thus making tumor cells more susceptive to heat stress. Moreover, radiation therapy with mild photothermal therapy effectively killed cancer cells and further induced the immune responses by releasing ROS as well as other immunomodulators. Consequently, the dying cancer cells combined with activated immune cells synergistically triggered powerful ICD and restarted the everlasting cancer immunity cycle, exhibiting a synergistic anticancer effect.
Additionally, a summary of biomimetic cell-derived nanoparticles applied in ACT is displayed in Table 2.

3.3. Cancer Vaccines

Cancer vaccines, including DNA vaccines, mRNA vaccines, peptides, tumor-specific proteins, tumor cells/lysates, and personalized vaccines coated with tumor cell membranes, can activate the anti-cancer immune responses by presenting tumor antigens and adjuvants to the host immune system [139,140,141]. Nevertheless, challenges in vaccine manufacturing, genetic heterogeneity among patients, limitations of immune response identification, and the disadvantages of the ITME seriously hampered the clinical application of cancer vaccines [139,142]. Therefore, nanovaccines as an alternative strategy in highly effective immunotherapy are attracting more and more attention [142,143,144].
Inspired by in situ vaccines, Xiong and colleagues constructed a personalized vaccine (R@P-IM) by camouflaging R837-loaded nanoparticles with calcinetin (CRT)-expressed cancer cell membranes containing tumor-associated antigens [145]. The engineered tumor-associated antigens constructed by ICD induction in vitro retained a complete antigen array while alleviating the acute systemic toxicity of chemotherapy in vivo. Meanwhile, the CRT expressed on the surface of the nanovaccine facilitated the internalization of the nanovaccine by DCs, consequently enhancing the immune responses. Subsequently, adjuvant R837 released from the nanovaccine excited toll-like receptor 7 (TLR7), which further activated DCs. In addition, the nanovaccine activated memory T cells for long-term protection, exhibiting a satisfactory immunotherapeutic efficacy for cancer therapy and prevention.
Although cancer cell membrane-derived nanovaccines can stimulate multiantigenic immunities with enhanced anticancer efficacy, once these nanovaccines enter the blood capillary via lymphatic capillaries, the adjuvants leakage usually triggers severe systemic inflammation. In order to avoid this high risk, there is an urgent need to construct biomimetic nanovaccines based on nanoparticle cores with weak or no immune stimulatory effects. Li and colleagues constructed a biomimetic nanovaccine (CCM@(PSiNPs@Au)) based on weak-immunostimulatory silicon@Au nanoparticles [146]. In their study, CCM@(PSiNPs@Au) with a weak immune stimulatory effect still efficiently delivered cancer cell membranes into DCs and triggered DC maturation, thereby leading to the activation of the downstream anti-tumor immune responses. Moreover, CCM@(PSiNPs@Au), as a photothermal therapeutic agent, combined with immunotherapies exhibited a satisfactory immunotherapeutic efficacy for cancer occurrence and development by activating the anti-cancer immune responses and reversing the ITME in vivo.
Neoantigens are abnormal proteins generated only by tumor cells, which are called tumor-specific antigens [147]. Therefore, neoantigens can prevent “off-target” destruction to normal tissues and are not affected by central or peripheral tolerance, making them promising candidate antigens for personalized cancer immunotherapies [148,149]. Meng and colleagues engineered bacteria to fabricate fusion neoantigens and constructed bacteria-derived neoantigen-bearing vesicles (BDVs-Neo) as an individualized cancer vaccine to trigger the systemic anti-tumor immune responses [150]. Then, BDVs-Neo and granulocyte-macrophage colony-stimulating factor (GM-CSF, an adjuvant) were injected subcutaneously within temperature-sensitive hydrogels. When combined with aPD-1, the sustained release of GM-CSF and BDVS-lipopolysaccharide (LPS) in hydrogels recruited DCs and provided long-term memory immunity by intensively enhancing the proliferation and activation of tumor-infiltrating lymphocytes (TILs) and clonal expansion of memory T cells (Figure 4).
Herpesvirus, a major human pathogen, can bind specifically to cancer cells by identifying tumor-associated antigens on the surface of tumor cells, and trigger strong and long-lasting anti-cancer immune responses by inducing mitochondrial DNA (mtDNA) stress [151]. In addition, Mn2+ is released from organelles and accumulates in the cytoplasm during herpesvirus infection, which promotes the antiviral innate immune responses by increasing the sensitivity of cGAS to mtDNA and facilitating STING activation [152,153]. Herein, encouraged by the strong innate immunity triggered by herpesvirus, engineering erythrocyte membrane-wrapped DNAzyme-loaded nanoparticles (Vir-ZM@TD) were constructed for cancer immunotherapy by simulating the structure and infection processes of herpesvirus [154]. Vir-ZM@TD not only effectively prolonged the blood circulation time of nanoparticles, but also closely mimicked a series of herpesvirus infection processes, including specific tumor targeting, effective endosomal escape mediated by membrane fusion, mitochondrial DNA stress triggered by transcription factor A, and Mn2+ release from organelles into the cytoplasm. Thus, the anti-cancer immune response triggered by the cGAS-STING innate pathway was effectively activated, and the complete tumor regression was about 68%.
Additionally, a summary of biomimetic cell-derived nanoparticles applied in cancer vaccines is displayed in Table 3.

3.4. Modulating the Immunosuppressive Tumor Microenvironment

The immunosuppressive tumor microenvironment (ITME) supports tumor escape from immune surveillance and is a major obstacle to immunotherapy [165,166]. Multiple complex factors contribute to the ITME (Figure 5). The low immunogenicity of tumors impede recognition by the immune system [167]. A variety of immunosuppressive cells and cytokines hinder anti-tumor immune responses via different pathways [168]. The extracellular matrix of the tumor prevents the infiltration of anti-tumor immune cells [169]. The hypoxia and abnormal metabolic activities of tumors are conducive to the immune escape of tumors [170,171,172]. Reversing the ITME is beneficial to the recruitment and activation of anti-tumor immune cells, which can promote immunotherapy, and biomimetic cell-derived nanodrug delivery systems may be a good choice.
Tumor-associated macrophages (TAMs) are an important part of the ITME [173]. TAMs can be polarized into M2-like phenotypes, which trigger the occurrence, progression, and recurrence of tumors [174]. Fortunately, TAMs have a certain degree of plasticity and can be converted to M1 type to inhibit tumor growth [175]. Wei and colleagues constructed a bacteria–nanoparticles complex (Ec-PR848) for TAM polarization and enhanced immunotherapy [176]. This smart biomimetic nanoparticle platform greatly repolarized M2-type macrophages towards M1-type macrophages, resulting in the generation of TNF-α, an increased expression of IL-6, as well as the activation of anti-cancer immunity. When supplemented with PLGA-DOX-triggered ICD, Ec-PR848 further weakened the degree of tumor immunosuppression and subsequently induced a strong anticancer immune response. In another study, Rao and colleagues show that SIRPα variants-overexpressed cancer cell membrane-wrapped magnetic nanoparticles (gCM-MNs) could effectively polarize M2 macrophages to M1 macrophages by the magnetic nanoparticle cores, facilitating macrophages to phagocytose cancer cells and enhancing anticancer T-cell immunity [177].
In addition to M2-type TAMs, the presence of tumor hypoxia is also significantly associated with the ITME [178]. Research shows that remodulating tumor hypoxia can facilitate cancer immunotherapy by stimulating anti-tumor T cells and NK cells, reducing macrophage recruitment and PD-L1 on cancer cells, and maintaining M1-TAMs polarization [179,180]. Herein, Wang and colleagues constructed a biomimetic drug delivery system (V(Hb)@DOX) for ITME remodulation by co-delivering oxygen and DOX using erythrocyte membrane camouflaged amphiphilic PCL nanoparticles [181]. The Hb moiety of V(Hb)@DOX effectively killed cancer cells by specifically targeting M2-type TAMs via the CD163 receptor, while the O2 released by Hb relieved cancer hypoxia and further enhanced the anti-cancer immunity by reducing the recruitment of M2-type TAMs, which synergistically reversed the ITME by down-regulating the expression of PD-L1 on cancer cells, reducing levels of immunosuppressive cytokines, increasing immunostimulant IFN-γ, enhancing CTL response, and inducing a robust memory immunity.
CAFs, as the main components of tumor stroma, provide a tremendous energy supply to tumor cells via the glycolytic pathway [182]. In addition, lactate produced by CAFs and cancer cells via glycolysis often results in the ITME [183,184]. Therefore, metabolic reprogramming by destroying the metabolic networks between CAFs and cancer cells may be a key to enhancing cancer immunotherapy. Zang and colleagues constructed a biomimetic nano-delivery system by using hybrid membranes of activated fibroblasts and cancer cells to coat solid lipid nanoparticles containing the glycolytic inhibitor PFK15 and chemotherapeutic drug PTX [185]. The obtained biomimetic nanoparticles (PTX/PFK15-SLN@ [4T1-3T3] NPs) possessed homologous targeting towards both CAFs and tumor cells. The encapsulated PFK15 effectively blocked glycolysis in both CAFs and tumor cells, thereby cutting off the energy supply of CAFs to tumor cells. Moreover, PTX/PFK15-SLN@ [4T1-3T3] significantly reduced lactate production in CAFs and cancer cells, thus reversing the ITME and thereby enhancing anti-cancer immunity.
Different from apoptosis which is generally considered to be an immune tolerance process, pyroptosis is a highly inflammatory programmed cell death (PCD) triggered by caspase-3, demonstrating a good opportunity to alleviate the ITME and facilitate the systemic immune responses [186,187]. Zhao and colleagues constructed a biomimetic nano-delivery system (BNP) by using a breast cancer membrane to coat PLGA nanoparticles containing indocyanine green (ICG) and decitabine (DCT) for photo-triggered cancer pyroptosis and cancer immunotherapy [188]. In their study, ICG induced a sharp increase in cytosolic Ca2+ concentration by NIR, which promoted the release of cytochrome c and subsequently activated caspase-3. DCT synergistically up-regulated the expression of gasdermin E by inhibiting DNA methylation, thereby enhancing the cleavage of gasdermin E by caspase-3 and causing robust cell pyroptosis. In particular, the cell pyroptosis triggered DC maturation, activated T cells, and exhibited powerful effects on primary and distant tumor immunotherapy.
Additionally, a summary of biomimetic cell-derived nanoparticles applied in modulating the ITME is displayed in Table 4.

3.5. Combination Therapy

Cancer is a complex disease with a variety of intricate factors, which makes it more difficult to treat different types of cancer with monotherapy [200]. It is suggested that combination therapy is a new trend for cancer treatment in the future. The combination of chemotherapy, radiotherapy, PDT, or other therapies with immunotherapy is the main combination treatment modality [201]. Additionally, biomimetic cell-derived nanocarriers play an important role in cancer combination therapy because of their unique superiorities.
Xiao and colleagues constructed biomimetic nanoparticles (PDA/GNS@aPD-L1) to combine PTT with PD-1/PD-L1 blockade for synergistic tumor inhibition in colorectal cancer [202]. Firstly, polydopamine-modified gold nanoparticles (PDA-GNS) were prepared as nanoparticle cores. Subsequently, cell membranes extracted from anti-PD-L1 scFv-engineered HEK 293T cells were applied to camouflage these PDA-GNs cores. Anti-PD-L1 scFv on the biomimetic nanoparticles not only blocked the PD-1/PD-L1 signal, but also promoted the aggregation of PDA-GNs at the tumor site. What is more, PDA-GNs-induced PTT triggered the release of tumor-associated antigens and reversed the ITME, which thereby greatly improved the outcome of ICI immunotherapy (Figure 6).
Lu and colleagues constructed tumor cells and DCs dual active-targeting biomimetic nanoparticles (AMR-MOF@AuPt) as a sono-immunotherapeutic nanoplatform for multimodal therapies [203]. In their study, the sono-responsive nanometal organic frameworks (MOFs) were successfully coated by engineering cancer cell membranes displaying anti-DEC205 antibodies. Anti-DEC205-anchored cell membranes directly targeted and activated DCs to facilitate tumor antigen cross-presentation, thereby triggering a cascade of T cell immune responses. More interestingly, AMR-MOF@AuPt-triggered SDT observably inhibited tumor growth through ROS and induced ICD of cancer cells, which further activated and proliferated cytotoxic T cells. This design allowed for multicellular engagement between tumor cells, DCs, and T cells, which thereby induced a robust systemic and long-term immunity to inhibit tumor relapse and distant metastasis.
Wang and colleagues constructed a PLGA-based biomimetic nano-delivery system (AFT/2-BP@PLGA@MD) which integrated small molecule targeted therapy with immunotherapy [204]. In this platform, the palmitoylation inhibitor 2-bromopalmitic acid (2-BP) was encapsulated in PLGA nanoparticles to enhance the treatment efficacy of afatinib against EGFR-TKIs non-sensitive cancer cells. Then, drug-loaded PLGA nanoparticles were wrapped with cancer cell membranes modified by a D-peptide antagonist (DPPA-1) to block PD-1/PD-L1. In their study, AFT/2-BP@PLGA@MD observably inhibited cancer occurrence and progression by directly killing cancer cells and triggering systemic T cell responses. Their findings indicated that small molecule targeted therapy combined with immunotherapy could break through their respective limitations and obtain stronger anti-cancer efficacy.
Li and colleagues constructed a multifunctional biomimetic delivery system (AuDRM) to realize the synergistic effect of starvation/PTT/immunotherapy [205]. Briefly, Au NPs were developed in the mesopores of mesoporous silica and then the obtained nanocomplexes were camouflaged with pH-responsive hybrid cancer cell membrane fragments. When AuDRM nanoparticles were efficiently accumulated at the tumor site, the pH-responsive membranes fell off, thereby facilitating the exposed Au NPs for starvation treatment and the released immunostimulant R837 for enhanced immunotherapy. Importantly, upon irradiation, Au NPs caused PTT and then triggered ICD. The resulting immunogenicity, together with the immunostimulant R837 released by AuDRM, stimulated stronger immune responses for cancer therapy.
To improve the safety and efficacy of OMVs in cancer treatment, Li and colleagues loaded OMVs into macrophages and used OMVs as platforms for co-delivery of DOX and Ce6 for combined tumor chemo/photodynamic/immunotherapy [206]. In their study, OMVs effectively polarized M2 macrophages to M1 macrophages and activated pyroptosis-related pathways, thereby enhancing antitumor immunity. The developed Ce6/DOX-OMVs@M demonstrated high safety and exhibited satisfactory immunotherapeutic efficacy for tumor ablation and metastasis (Figure 7).
Additionally, a summary of biomimetic cell-derived nanoparticles applied in combination therapy is displayed in Table 5.

4. Conclusions and Challenges

With new advances in the construction of nano-based drug delivery systems, researchers have successfully developed various of biomimetic cell-derived nanoparticles. This review highlights the recent developments in biomimetic cell-derived nanoparticles for cancer immunotherapy.
Benefiting from ideal characteristics, biomimetic cell-derived nanoparticles can effectively deliver immunotherapeutic agents and/or immunostimulants to tumor sites, reverse the ITME to an immune-supportive one, trigger ICD, induce the release of tumor-associated antigens, facilitate DCs maturation and tumor antigen presentation, as well as activate T and NK cells, which can observably promote the immune responses and improve the efficacy of cancer immunotherapy. The critical analysis of abundant evidence acquired from existing research suggests that biomimetic cell-derived nanoparticles with cell membrane camouflage can be effectively used for cancer bio-imaging, prevention, diagnosis, and treatment (Figure 8).
Although the advantages of biomimetic cell-derived nanoparticles have promoted the development of cancer immunotherapy, many challenges still need to be overcome before their clinical transformation.
Firstly, the anti-cancer efficacy and long-term safety of these emerging biomimetic nanoparticles have not been fully established in humans, which may be one of the key scientific questions for scientists to address in the future. Moreover, the pharmaceutical spectrum of these biomimetic nanoparticles can be further expanded to the delivery of different types of drugs to manage other types of diseases.
Secondly, the isolation of the cell membrane mainly includes cell lysis, cell contents removal, and purification. The removal of cytoplasmic components by differential centrifugation in the current studies may not completely remove all cytoplasmic components and may instead lead to the loss of membrane fragments. In addition, gentle extraction is required because functional proteins on the cell surface are very susceptible to denaturation and inactivation. How to extract high-purity and intact cell membranes to make sure that the cell membranes can completely inherit the biological functions of the source cells deserves further study.
Thirdly, the extrusion method generally used to prepare biomimetic nanoparticles is time-consuming, inefficient, and difficult to scale up in industrial production. Moreover, the uncontrollable stability of the production process and the high cost of the manufacturing method are also significant challenges. Therefore, the high-quality control and large-scale preparation of biomimetic nanoparticles are urgent problems to be solved.
Fourthly, current complexity and reproducibility issues are also major challenges associated with biomimetic nanoparticles. In general, the more complicated the preparation processes, the worse the reproducibility. Researchers should prioritize the scale-up of biomimetic nanoparticles in the early stages of nanoparticle design, and the preparation methods and technologies used to construct biomimetic nanoparticles should be simple, reliable, and mature enough to meet the requirements of large-scale production. In addition, the introduction of new preparation methods, such as microfluidic electroporation, is also a future direction. Moreover, surface membrane components are known to play a key role in the design of biomimetic nanoparticles, but the guidelines on which kind of cell membrane may work better for a specific application are not available. According to the current literatures, immune cell membranes and cancer cell membranes are mainly used to fabricate biomimetic nanovaccines for cancer immunotherapy, EVs are mainly used to treat neurological diseases or deliver nucleic acid drugs to treat genetic diseases and cancer, and platelet membrane-derived nanoparticles can effectively locate atherosclerotic plaques for atherosclerosis detection.
Lastly, tumor heterogeneity is a common challenge in the successful treatment of cancer. In cancer immunotherapy, the response of the immune system against antigen-positive cells can lead to selective pressure towards antigen-negative cells, which is a common cause of cancer recurrence. Combined with high-level techniques, developing some individualized biomimetic nanomedicines and improving their efficacy in cancer immunotherapy may be effective problem-solving strategies.
Although the clinical translation of biomimetic cell-derived nanoparticles faces many challenges, it is undeniable that biomimetic cell-derived nanoparticles have unique advantages and great potential in cancer immunotherapy.

Author Contributions

T.H., Data curation, Formal analysis, Investigation, Writing—original draft. Y.H., Data curation, Funding acquisition, Writing—original draft. J.L., Visualization. C.S., Validation. F.W., Conceptualization, Resources, Software, Writing—review and editing. Z.H., Conceptualization, Funding acquisition, Project administration, Supervision, Writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Science Fund for Excellent Young Scholars (No. 32122052) and the 1.3.5 project for disciplines of excellence—clinical research incubation project, West China Hospital, Sichuan University (No. 2021HXFH064).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable for this article.

Acknowledgments

This work was supported by National Key Clinical Specialties Construction Program.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Yang, Y. Cancer immunotherapy: Harnessing the immune system to battle cancer. J. Clin. Investig. 2015, 125, 3335–3337. [Google Scholar] [CrossRef] [Green Version]
  2. Zeng, Y.; Li, S.; Zhang, S.; Wang, L.; Yuan, H.; Hu, F. Cell membrane coated-nanoparticles for cancer immunotherapy. Acta Pharm. Sin. B 2022, 12, 3233–3254. [Google Scholar] [CrossRef]
  3. Peterson, C.; Denlinger, N.; Yang, Y. Recent Advances and Challenges in Cancer Immunotherapy. Cancers 2022, 14, 3972. [Google Scholar] [CrossRef]
  4. Alhaj-Suliman, S.O.; Wafa, E.I.; Salem, A.K. Engineering nanosystems to overcome barriers to cancer diagnosis and treatment. Adv. Drug Deliv. Rev. 2022, 189, 114482. [Google Scholar] [CrossRef]
  5. Yang, K.; Yang, Z.; Yu, G.; Nie, Z.; Wang, R.; Chen, X. Polyprodrug Nanomedicines: An Emerging Paradigm for Cancer Therapy. Adv. Mater. 2021, 34, e2107434. [Google Scholar] [CrossRef]
  6. Hegde, M.; Naliyadhara, N.; Unnikrishnan, J.; Alqahtani, M.S.; Abbas, M.; Girisa, S.; Sethi, G.; Kunnumakkara, A.B. Nanoparticles in the diagnosis and treatment of cancer metastases: Current and future perspectives. Cancer Lett. 2023, 556, 216066. [Google Scholar] [CrossRef]
  7. Shukla, A.; Maiti, P. Nanomedicine and versatile therapies for cancer treatment. MedComm 2022, 3, e163. [Google Scholar] [CrossRef]
  8. Llop, J.; Lammers, T. Nanoparticles for Cancer Diagnosis, Radionuclide Therapy and Theranostics. ACS Nano 2021, 15, 16974–16981. [Google Scholar] [CrossRef]
  9. Zhou, Y.; Dai, Z. New Strategies in the Design of Nanomedicines to Oppose Uptake by the Mononuclear Phagocyte System and Enhance Cancer Therapeutic Efficacy. Chem. Asian J. 2018, 13, 3333–3340. [Google Scholar] [CrossRef]
  10. Ibrahim, M.; Ramadan, E.; Elsadek, N.E.; Emam, S.E.; Shimizu, T.; Ando, H.; Ishima, Y.; Elgarhy, O.H.; Sarhan, H.A.; Hussein, A.K.; et al. Polyethylene glycol (PEG): The nature, immunogenicity, and role in the hypersensitivity of PEGylated products. J. Control. Release 2022, 351, 215–230. [Google Scholar] [CrossRef]
  11. Shi, D.; Beasock, D.; Fessler, A.; Szebeni, J.; Ljubimova, J.Y.; Afonin, K.A.; Dobrovolskaia, M.A. To PEGylate or not to PEGylate: Immunological properties of nanomedicine’s most popular component, polyethylene glycol and its alternatives. Adv. Drug Deliv. Rev. 2022, 180, 114079. [Google Scholar] [CrossRef]
  12. Kozma, G.T.; Shimizu, T.; Ishida, T.; Szebeni, J. Anti-PEG antibodies: Properties, formation, testing and role in adverse immune reactions to PEGylated nano-biopharmaceuticals. Adv. Drug Deliv. Rev. 2020, 154–155, 163–175. [Google Scholar] [CrossRef]
  13. Estape Senti, M.; de Jongh, C.A.; Dijkxhoorn, K.; Verhoef, J.J.F.; Szebeni, J.; Storm, G.; Hack, C.E.; Schiffelers, R.M.; Fens, M.H.; Boross, P. Anti-PEG antibodies compromise the integrity of PEGylated lipid-based nanoparticles via complement. J. Control. Release 2022, 341, 475–486. [Google Scholar] [CrossRef]
  14. Hussain, Z.; Rahim, M.A.; Jan, N.; Shah, H.; Rawas-Qalaji, M.; Khan, S.; Sohail, M.; Thu, H.E.; Ramli, N.A.; Sarfraz, R.M.; et al. Cell membrane cloaked nanomedicines for bio-imaging and immunotherapy of cancer: Improved pharmacokinetics, cell internalization and anticancer efficacy. J. Control. Release 2021, 335, 130–157. [Google Scholar] [CrossRef]
  15. Dhas, N.; Garcia, M.C.; Kudarha, R.; Pandey, A.; Nikam, A.N.; Gopalan, D.; Fernandes, G.; Soman, S.; Kulkarni, S.; Seetharam, R.N.; et al. Advancements in cell membrane camouflaged nanoparticles: A bioinspired platform for cancer therapy. J. Control. Release 2022, 346, 71–97. [Google Scholar] [CrossRef]
  16. 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, e2002081. [Google Scholar] [CrossRef]
  17. Zhang, F.; Mundaca-Uribe, R.; Askarinam, N.; Li, Z.; Gao, W.; Zhang, L.; Wang, J. Biomembrane-Functionalized Micromotors: Biocompatible Active Devices for Diverse Biomedical Applications. Adv. Mater. 2022, 34, e2107177. [Google Scholar] [CrossRef]
  18. Chen, X.; Liu, B.; Tong, R.; Zhan, L.; Yin, X.; Luo, X.; Huang, Y.; Zhang, J.; He, W.; Wang, Y. Orchestration of biomimetic membrane coating and nanotherapeutics in personalized anticancer therapy. Biomater. Sci. 2021, 9, 590–625. [Google Scholar] [CrossRef]
  19. Shen, M.; Wu, X.; Zhu, M.; Yi, X. Recent advances in biological membrane-based nanomaterials for cancer therapy. Biomater. Sci. 2022, 10, 5756–5785. [Google Scholar] [CrossRef]
  20. Jha, A.; Nikam, A.N.; Kulkarni, S.; Mutalik, S.P.; Pandey, A.; Hegde, M.; Rao, B.S.S.; Mutalik, S. Biomimetic nanoarchitecturing: A disguised attack on cancer cells. J. Control. Release 2021, 329, 413–433. [Google Scholar] [CrossRef]
  21. Zhao, J.; Ruan, J.; Lv, G.; Shan, Q.; Fan, Z.; Wang, H.; Du, Y.; Ling, L. Cell membrane-based biomimetic nanosystems for advanced drug delivery in cancer therapy: A comprehensive review. Colloids Surf. B Biointerfaces 2022, 215, 112503. [Google Scholar] [CrossRef]
  22. Fang, R.H.; Gao, W.; Zhang, L. Targeting drugs to tumours using cell membrane-coated nanoparticles. Nat. Rev. Clin. Oncol. 2022, 20, 33–48. [Google Scholar] [CrossRef]
  23. Hu, C.M.; Zhang, L.; Aryal, S.; Cheung, C.; Fang, R.H.; Zhang, L. Erythrocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc. Natl. Acad. Sci. USA 2011, 108, 10980–10985. [Google Scholar] [CrossRef] [Green Version]
  24. Liu, H.; Su, Y.Y.; Jiang, X.C.; Gao, J.Q. Cell membrane-coated nanoparticles: A novel multifunctional biomimetic drug delivery system. Drug Deliv. Transl. Res. 2023, 13, 716–737. [Google Scholar] [CrossRef]
  25. Chen, L.; Hong, W.; Ren, W.; Xu, T.; Qian, Z.; He, Z. Recent progress in targeted delivery vectors based on biomimetic nanoparticles. Signal Transduct. Target. Ther. 2021, 6, 225. [Google Scholar] [CrossRef]
  26. Zhang, M.; Cheng, S.; Jin, Y.; Zhang, N.; Wang, Y. Membrane engineering of cell membrane biomimetic nanoparticles for nanoscale therapeutics. Clin. Transl. Med. 2021, 11, e292. [Google Scholar] [CrossRef]
  27. Krishnan, N.; Peng, F.X.; Mohapatra, A.; Fang, R.H.; Zhang, L. Genetically engineered cellular nanoparticles for biomedical applications. Biomaterials 2023, 296, 122065. [Google Scholar] [CrossRef]
  28. Meng, Z.; Zhang, Y.; Zhou, X.; Ji, J.; Liu, Z. Nanovaccines with cell-derived components for cancer immunotherapy. Adv. Drug Deliv. Rev. 2022, 182, 114107. [Google Scholar] [CrossRef]
  29. Shi, Y.; Qian, H.; Rao, P.; Mu, D.; Liu, Y.; Liu, G.; Lin, Z. Bioinspired membrane-based nanomodulators for immunotherapy of autoimmune and infectious diseases. Acta Pharm. Sin. B 2022, 12, 1126–1147. [Google Scholar] [CrossRef]
  30. Wu, Z.; Zhang, H.; Yan, J.; Wei, Y.; Su, J. Engineered biomembrane-derived nanoparticles for nanoscale theranostics. Theranostics 2023, 13, 20–39. [Google Scholar] [CrossRef]
  31. Shi, Y.; Lu, A.; Wang, X.; Belhadj, Z.; Wang, J.; Zhang, Q. A review of existing strategies for designing long-acting parenteral formulations: Focus on underlying mechanisms, and future perspectives. Acta Pharm. Sin. B 2021, 11, 2396–2415. [Google Scholar] [CrossRef]
  32. Castro, F.; Martins, C.; Silveira, M.J.; Moura, R.P.; Pereira, C.L.; Sarmento, B. Advances on erythrocyte-mimicking nanovehicles to overcome barriers in biological microenvironments. Adv. Drug Deliv. Rev. 2021, 170, 312–339. [Google Scholar] [CrossRef]
  33. Xia, Q.; Zhang, Y.; Li, Z.; Hou, X.; Feng, N. Red blood cell membrane-camouflaged nanoparticles: A novel drug delivery system for antitumor application. Acta Pharm. Sin. B 2019, 9, 675–689. [Google Scholar] [CrossRef]
  34. Bidkar, A.P.; Sanpui, P.; Ghosh, S.S. Transferrin-Conjugated Red Blood Cell Membrane-Coated Poly(lactic-co-glycolic acid) Nanoparticles for the Delivery of Doxorubicin and Methylene Blue. ACS Appl. Nano Mater. 2020, 3, 3807–3819. [Google Scholar] [CrossRef]
  35. Choi, M.J.; Lee, Y.K.; Choi, K.C.; Lee, D.H.; Jeong, H.Y.; Kang, S.J.; Kim, M.W.; You, Y.M.; Im, C.S.; Lee, T.S.; et al. Tumor-Targeted Erythrocyte Membrane Nanoparticles for Theranostics of Triple-Negative Breast Cancer. Pharmaceutics 2023, 15, 350. [Google Scholar] [CrossRef]
  36. Long, Y.; Wang, Z.; Fan, J.; Yuan, L.; Tong, C.; Zhao, Y.; Liu, B. A hybrid membrane coating nanodrug system against gastric cancer via the VEGFR2/STAT3 signaling pathway. J. Mater. Chem. B 2021, 9, 3838–3855. [Google Scholar] [CrossRef]
  37. Huo, Y.Y.; Song, X.; Zhang, W.X.; Zhou, Z.L.; Lv, Q.Y.; Cui, H.F. Thermosensitive Biomimetic Hybrid Membrane Camouflaged Hollow Gold Nanoparticles for NIR-Responsive Mild-Hyperthermia Chemo-/Photothermal Combined Tumor Therapy. ACS Appl. Bio Mater. 2022, 5, 5113–5125. [Google Scholar] [CrossRef]
  38. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
  39. Li, R.; He, Y.; Zhang, S.; Qin, J.; Wang, J. Cell membrane-based nanoparticles: A new biomimetic platform for tumor diagnosis and treatment. Acta Pharm. Sin. B 2018, 8, 14–22. [Google Scholar] [CrossRef]
  40. Oroojalian, F.; Beygi, M.; Baradaran, B.; Mokhtarzadeh, A.; Shahbazi, M.A. Immune Cell Membrane-Coated Biomimetic Nanoparticles for Targeted Cancer Therapy. Small 2021, 17, e2006484. [Google Scholar] [CrossRef]
  41. 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, 1903301. [Google Scholar] [CrossRef]
  42. Xu, C.; Jiang, Y.; Han, Y.; Pu, K.; Zhang, R. A Polymer Multicellular Nanoengager for Synergistic NIR-II Photothermal Immunotherapy. Adv. Mater. 2021, 33, e2008061. [Google Scholar] [CrossRef]
  43. Gong, P.; Wang, Y.; Zhang, P.; Yang, Z.; Deng, W.; Sun, Z.; Yang, M.; Li, X.; Ma, G.; Deng, G.; et al. Immunocyte Membrane-Coated Nanoparticles for Cancer Immunotherapy. Cancers 2020, 13, 77. [Google Scholar] [CrossRef]
  44. Khatoon, N.; Zhang, Z.; Zhou, C.; Chu, M. Macrophage membrane coated nanoparticles: A biomimetic approach for enhanced and targeted delivery. Biomater. Sci. 2022, 10, 1193–1208. [Google Scholar] [CrossRef]
  45. Gaber, T.; Chen, Y.; Krauss, P.L.; Buttgereit, F. Metabolism of T Lymphocytes in Health and Disease. Int. Rev. Cell Mol. Biol. 2019, 342, 95–148. [Google Scholar] [CrossRef]
  46. Notarbartolo, S.; Abrignani, S. Human T lymphocytes at tumor sites. Semin. Immunopathol. 2022, 44, 883–901. [Google Scholar] [CrossRef]
  47. Farhood, B.; Najafi, M.; Mortezaee, K. CD8(+) cytotoxic T lymphocytes in cancer immunotherapy: A review. J. Cell Physiol. 2019, 234, 8509–8521. [Google Scholar] [CrossRef]
  48. Yaman, S.; Ramachandramoorthy, H.; Oter, G.; Zhukova, D.; Nguyen, T.; Sabnani, M.K.; Weidanz, J.A.; Nguyen, K.T. Melanoma Peptide MHC Specific TCR Expressing T-Cell Membrane Camouflaged PLGA Nanoparticles for Treatment of Melanoma Skin Cancer. Front. Bioeng. Biotechnol. 2020, 8, 943. [Google Scholar] [CrossRef]
  49. Raskov, H.; Orhan, A.; Christensen, J.P.; Gogenur, I. Cytotoxic CD8(+) T cells in cancer and cancer immunotherapy. Br. J. Cancer 2021, 124, 359–367. [Google Scholar] [CrossRef]
  50. Chen, B.Q.; Zhao, Y.; Zhang, Y.; Pan, Y.J.; Xia, H.Y.; Kankala, R.K.; Wang, S.B.; Liu, G.; Chen, A.Z. Immune-regulating camouflaged nanoplatforms: A promising strategy to improve cancer nano-immunotherapy. Bioact. Mater. 2023, 21, 1–19. [Google Scholar] [CrossRef]
  51. Han, B.; Mao, F.Y.; Zhao, Y.L.; Lv, Y.P.; Teng, Y.S.; Duan, M.; Chen, W.; Cheng, P.; Wang, T.T.; Liang, Z.Y.; et al. Altered NKp30, NKp46, NKG2D, and DNAM-1 Expression on Circulating NK Cells Is Associated with Tumor Progression in Human Gastric Cancer. J. Immunol. Res. 2018, 2018, 6248590. [Google Scholar] [CrossRef] [Green Version]
  52. Jain, N.; Shahrukh, S.; Famta, P.; Shah, S.; Vambhurkar, G.; Khatri, D.K.; Singh, S.B.; Srivastava, S. Immune cell-camouflaged surface-engineered nanotherapeutics for cancer management. Acta Biomater. 2023, 155, 57–79. [Google Scholar] [CrossRef]
  53. Han, H.; Bartolo, R.; Li, J.; Shahbazi, M.A.; Santos, H.A. Biomimetic platelet membrane-coated nanoparticles for targeted therapy. Eur. J. Pharm. Biopharm. 2022, 172, 1–15. [Google Scholar] [CrossRef]
  54. Hu, C.M.; Fang, R.H.; Wang, K.C.; Luk, B.T.; Thamphiwatana, S.; Dehaini, D.; Nguyen, P.; Angsantikul, P.; Wen, C.H.; Kroll, A.V.; et al. Nanoparticle biointerfacing by platelet membrane cloaking. Nature 2015, 526, 118–121. [Google Scholar] [CrossRef] [Green Version]
  55. Hu, Q.; Sun, W.; Qian, C.; Wang, C.; Bomba, H.N.; Gu, Z. Anticancer Platelet-Mimicking Nanovehicles. Adv. Mater. 2015, 27, 7043–7050. [Google Scholar] [CrossRef]
  56. Fabricius, H.A.; Starzonek, S.; Lange, T. The Role of Platelet Cell Surface P-Selectin for the Direct Platelet-Tumor Cell Contact During Metastasis Formation in Human Tumors. Front. Oncol. 2021, 11, 642761. [Google Scholar] [CrossRef]
  57. Chen, Z.; Zhao, P.; Luo, Z.; Zheng, M.; Tian, H.; Gong, P.; Gao, G.; Pan, H.; Liu, L.; Ma, A.; et al. Cancer Cell Membrane-Biomimetic Nanoparticles for Homologous-Targeting Dual-Modal Imaging and Photothermal Therapy. ACS Nano 2016, 10, 10049–10057. [Google Scholar] [CrossRef]
  58. Harris, J.C.; Scully, M.A.; Day, E.S. Cancer Cell Membrane-Coated Nanoparticles for Cancer Management. Cancers 2019, 11, 1836. [Google Scholar] [CrossRef] [Green Version]
  59. Tian, H.; Luo, Z.; Liu, L.; Zheng, M.; Chen, Z.; Ma, A.; Liang, R.; Han, Z.; Lu, C.; Cai, L. Cancer Cell Membrane-Biomimetic Oxygen Nanocarrier for Breaking Hypoxia-Induced Chemoresistance. Adv. Funct. Mater. 2017, 27, 1703197. [Google Scholar] [CrossRef]
  60. Zhu, J.Y.; Zheng, D.W.; Zhang, M.K.; Yu, W.Y.; Qiu, W.X.; Hu, J.J.; Feng, J.; Zhang, X.Z. Preferential Cancer Cell Self-Recognition and Tumor Self-Targeting by Coating Nanoparticles with Homotypic Cancer Cell Membranes. Nano Lett. 2016, 16, 5895–5901. [Google Scholar] [CrossRef]
  61. Sun, H.; Su, J.; Meng, Q.; Yin, Q.; Chen, L.; Gu, W.; Zhang, P.; Zhang, Z.; Yu, H.; Wang, S.; et al. Cancer-Cell-Biomimetic Nanoparticles for Targeted Therapy of Homotypic Tumors. Adv. Mater. 2016, 28, 9581–9588. [Google Scholar] [CrossRef]
  62. Pei, X.; Pan, X.; Xu, X.; Xu, X.; Huang, H.; Wu, Z.; Qi, X. 4T1 cell membrane fragment reunited PAMAM polymer units disguised as tumor cell clusters for tumor homotypic targeting and anti-metastasis treatment. Biomater. Sci. 2021, 9, 1325–1333. [Google Scholar] [CrossRef]
  63. Yue, X.S.; Murakami, Y.; Tamai, T.; Nagaoka, M.; Cho, C.S.; Ito, Y.; Akaike, T. A fusion protein N-cadherin-Fc as an artificial extracellular matrix surface for maintenance of stem cell features. Biomaterials 2010, 31, 5287–5296. [Google Scholar] [CrossRef]
  64. Iurisci, I.; Cumashi, A.; Sherman, A.A.; Tsvetkov, Y.E.; Tinari, N.; Piccolo, E.; D’Egidio, M.; Adamo, V.; Natoli, C.; Rabinovich, G.A.; et al. Synthetic inhibitors of galectin-1 and -3 selectively modulate homotypic cell aggregation and tumor cell apoptosis. Anticancer Res. 2009, 29, 403–410. [Google Scholar]
  65. Osta, W.A.; Chen, Y.; Mikhitarian, K.; Mitas, M.; Salem, M.; Hannun, Y.A.; Cole, D.J.; Gillanders, W.E. EpCAM is overexpressed in breast cancer and is a potential target for breast cancer gene therapy. Cancer Res. 2004, 64, 5818–5824. [Google Scholar] [CrossRef] [Green Version]
  66. Wu, H.H.; Zhou, Y.; Tabata, Y.; Gao, J.Q. Mesenchymal stem cell-based drug delivery strategy: From cells to biomimetic. J. Control. Release 2019, 294, 102–113. [Google Scholar] [CrossRef]
  67. Khosravi, N.; Pishavar, E.; Baradaran, B.; Oroojalian, F.; Mokhtarzadeh, A. Stem cell membrane, stem cell-derived exosomes and hybrid stem cell camouflaged nanoparticles: A promising biomimetic nanoplatforms for cancer theranostics. J. Control. Release 2022, 348, 706–722. [Google Scholar] [CrossRef]
  68. Wang, M.; Xin, Y.; Cao, H.; Li, W.; Hua, Y.; Webster, T.J.; Zhang, C.; Tang, W.; Liu, Z. Recent advances in mesenchymal stem cell membrane-coated nanoparticles for enhanced drug delivery. Biomater. Sci. 2021, 9, 1088–1103. [Google Scholar] [CrossRef]
  69. Takayama, Y.; Kusamori, K.; Nishikawa, M. Mesenchymal stem/stromal cells as next-generation drug delivery vehicles for cancer therapeutics. Expert Opin. Drug Deliv. 2021, 18, 1627–1642. [Google Scholar] [CrossRef]
  70. Starnes, C.O. Coley’s toxins. Nature 1992, 360, 23. [Google Scholar] [CrossRef]
  71. Sartorio, M.G.; Pardue, E.J.; Feldman, M.F.; Haurat, M.F. Bacterial Outer Membrane Vesicles: From Discovery to Applications. Annu. Rev. Microbiol. 2021, 75, 609–630. [Google Scholar] [CrossRef]
  72. Liu, J.; Liew, S.S.; Wang, J.; Pu, K. Bioinspired and Biomimetic Delivery Platforms for Cancer Vaccines. Adv. Mater. 2022, 34, e2103790. [Google Scholar] [CrossRef]
  73. Li, M.; Zhou, H.; Yang, C.; Wu, Y.; Zhou, X.; Liu, H.; Wang, Y. Bacterial outer membrane vesicles as a platform for biomedical applications: An update. J. Control. Release 2020, 323, 253–268. [Google Scholar] [CrossRef]
  74. Holay, M.; Guo, Z.; Pihl, J.; Heo, J.; Park, J.H.; Fang, R.H.; Zhang, L. Bacteria-Inspired Nanomedicine. ACS Appl. Bio Mater. 2021, 4, 3830–3848. [Google Scholar] [CrossRef]
  75. Krishnan, N.; Kubiatowicz, L.J.; Holay, M.; Zhou, J.; Fang, R.H.; Zhang, L. Bacterial membrane vesicles for vaccine applications. Adv. Drug Deliv. Rev. 2022, 185, 114294. [Google Scholar] [CrossRef]
  76. Long, Q.; Zheng, P.; Zheng, X.; Li, W.; Hua, L.; Yang, Z.; Huang, W.; Ma, Y. Engineered bacterial membrane vesicles are promising carriers for vaccine design and tumor immunotherapy. Adv. Drug Deliv. Rev. 2022, 186, 114321. [Google Scholar] [CrossRef]
  77. Herrmann, I.K.; Wood, M.J.A.; Fuhrmann, G. Extracellular vesicles as a next-generation drug delivery platform. Nat. Nanotechnol. 2021, 16, 748–759. [Google Scholar] [CrossRef]
  78. Najafi, S.; Majidpoor, J.; Mortezaee, K. Extracellular vesicle-based drug delivery in cancer immunotherapy. Drug Deliv. Transl. Res. 2023, 1–17. [Google Scholar] [CrossRef]
  79. Wu, P.; Zhang, B.; Ocansey, D.K.W.; Xu, W.; Qian, H. Extracellular vesicles: A bright star of nanomedicine. Biomaterials 2021, 269, 120467. [Google Scholar] [CrossRef]
  80. Liu, S.; Wu, X.; Chandra, S.; Lyon, C.; Ning, B.; Jiang, L.; Fan, J.; Hu, T.Y. Extracellular vesicles: Emerging tools as therapeutic agent carriers. Acta Pharm. Sin. B 2022, 12, 3822–3842. [Google Scholar] [CrossRef]
  81. Liu, C.; Wang, Y.; Li, L.; He, D.; Chi, J.; Li, Q.; Wu, Y.; Zhao, Y.; Zhang, S.; Wang, L.; et al. Engineered extracellular vesicles and their mimetics for cancer immunotherapy. J. Control. Release 2022, 349, 679–698. [Google Scholar] [CrossRef]
  82. Ruan, S.; Greenberg, Z.; Pan, X.; Zhuang, P.; Erwin, N.; He, M. Extracellular Vesicles as an Advanced Delivery Biomaterial for Precision Cancer Immunotherapy. Adv. Healthc. Mater. 2022, 11, e2100650. [Google Scholar] [CrossRef]
  83. Chen, H.Y.; Deng, J.; Wang, Y.; Wu, C.Q.; Li, X.; Dai, H.W. Hybrid cell membrane-coated nanoparticles: A multifunctional biomimetic platform for cancer diagnosis and therapy. Acta Biomater. 2020, 112, 1–13. [Google Scholar] [CrossRef]
  84. Wang, D.; Liu, C.; You, S.; Zhang, K.; Li, M.; Cao, Y.; Wang, C.; Dong, H.; Zhang, X. Bacterial Vesicle-Cancer Cell Hybrid Membrane-Coated Nanoparticles for Tumor Specific Immune Activation and Photothermal Therapy. ACS Appl. Mater. Interfaces 2020, 12, 41138–41147. [Google Scholar] [CrossRef]
  85. Liu, Y.; Sukumar, U.K.; Kanada, M.; Krishnan, A.; Massoud, T.F.; Paulmurugan, R. Camouflaged Hybrid Cancer Cell-Platelet Fusion Membrane Nanovesicles Deliver Therapeutic MicroRNAs to Presensitize Triple-Negative Breast Cancer to Doxorubicin. Adv. Funct. Mater. 2021, 31, 2103600. [Google Scholar] [CrossRef]
  86. Zhang, T.; Liu, H.; Li, L.; Guo, Z.; Song, J.; Yang, X.; Wan, G.; Li, R.; Wang, Y. Leukocyte/platelet hybrid membrane-camouflaged dendritic large pore mesoporous silica nanoparticles co-loaded with photo/chemotherapeutic agents for triple negative breast cancer combination treatment. Bioact. Mater. 2021, 6, 3865–3878. [Google Scholar] [CrossRef]
  87. Liu, W.L.; Zou, M.Z.; Liu, T.; Zeng, J.Y.; Li, X.; Yu, W.Y.; Li, C.X.; Ye, J.J.; Song, W.; Feng, J.; et al. Cytomembrane nanovaccines show therapeutic effects by mimicking tumor cells and antigen presenting cells. Nat. Commun. 2019, 10, 3199. [Google Scholar] [CrossRef] [Green Version]
  88. Gao, W.; Fang, R.H.; Thamphiwatana, S.; Luk, B.T.; Li, J.; Angsantikul, P.; Zhang, Q.; Hu, C.M.; Zhang, L. Modulating antibacterial immunity via bacterial membrane-coated nanoparticles. Nano Lett. 2015, 15, 1403–1409. [Google Scholar] [CrossRef] [Green Version]
  89. Patel, R.B.; Ye, M.; Carlson, P.M.; Jaquish, A.; Zangl, L.; Ma, B.; Wang, Y.; Arthur, I.; Xie, R.; Brown, R.J.; et al. Development of an In Situ Cancer Vaccine via Combinational Radiation and Bacterial-Membrane-Coated Nanoparticles. Adv. Mater. 2019, 31, e1902626. [Google Scholar] [CrossRef]
  90. Dash, P.; Piras, A.M.; Dash, M. Cell membrane coated nanocarriers—An efficient biomimetic platform for targeted therapy. J. Control. Release 2020, 327, 546–570. [Google Scholar] [CrossRef]
  91. Le, Q.V.; Lee, J.; Lee, H.; Shim, G.; Oh, Y.K. Cell membrane-derived vesicles for delivery of therapeutic agents. Acta Pharm. Sin. B 2021, 11, 2096–2113. [Google Scholar] [CrossRef]
  92. Luk, B.T.; Hu, C.M.; Fang, R.H.; Dehaini, D.; Carpenter, C.; Gao, W.; Zhang, L. Interfacial interactions between natural RBC membranes and synthetic polymeric nanoparticles. Nanoscale 2014, 6, 2730–2737. [Google Scholar] [CrossRef] [Green Version]
  93. Xu, C.H.; Ye, P.J.; Zhou, Y.C.; He, D.X.; Wei, H.; Yu, C.Y. Cell membrane-camouflaged nanoparticles as drug carriers for cancer therapy. Acta Biomater. 2020, 105, 1–14. [Google Scholar] [CrossRef]
  94. Rao, L.; Cai, B.; Bu, L.L.; Liao, Q.Q.; Guo, S.S.; Zhao, X.Z.; Dong, W.F.; Liu, W. Microfluidic Electroporation-Facilitated Synthesis of Erythrocyte Membrane-Coated Magnetic Nanoparticles for Enhanced Imaging-Guided Cancer Therapy. ACS Nano 2017, 11, 3496–3505. [Google Scholar] [CrossRef]
  95. Zhang, J.; Gao, W.; Fang, R.H.; Dong, A.; Zhang, L. Synthesis of Nanogels via Cell Membrane-Templated Polymerization. Small 2015, 11, 4309–4313. [Google Scholar] [CrossRef] [Green Version]
  96. Lee, J.B.; Kim, H.R.; Ha, S.J. Immune Checkpoint Inhibitors in 10 Years: Contribution of Basic Research and Clinical Application in Cancer Immunotherapy. Immune. Netw. 2022, 22, e2. [Google Scholar] [CrossRef]
  97. Pisibon, C.; Ouertani, A.; Bertolotto, C.; Ballotti, R.; Cheli, Y. Immune Checkpoints in Cancers: From Signaling to the Clinic. Cancers 2021, 13, 4573. [Google Scholar] [CrossRef]
  98. Akbay, E.A.; Koyama, S.; Carretero, J.; Altabef, A.; Tchaicha, J.H.; Christensen, C.L.; Mikse, O.R.; Cherniack, A.D.; Beauchamp, E.M.; Pugh, T.J.; et al. Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Discov. 2013, 3, 1355–1363. [Google Scholar] [CrossRef] [Green Version]
  99. Daassi, D.; Mahoney, K.M.; Freeman, G.J. The importance of exosomal PDL1 in tumour immune evasion. Nat. Rev. Immunol. 2020, 20, 209–215. [Google Scholar] [CrossRef]
  100. Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef]
  101. Ye, L.; Zhu, Z.; Chen, X.; Zhang, H.; Huang, J.; Gu, S.; Zhao, X. The Importance of Exosomal PD-L1 in Cancer Progression and Its Potential as a Therapeutic Target. Cells 2021, 10, 3247. [Google Scholar] [CrossRef]
  102. Yan, J.; Liu, X.; Wu, F.; Ge, C.; Ye, H.; Chen, X.; Wei, Y.; Zhou, R.; Duan, S.; Zhu, R.; et al. Platelet Pharmacytes for the Hierarchical Amplification of Antitumor Immunity in Response to Self-Generated Immune Signals. Adv. Mater. 2022, 34, e2109517. [Google Scholar] [CrossRef]
  103. Yan, W.; Jiang, S. Immune Cell-Derived Exosomes in the Cancer-Immunity Cycle. Trends Cancer 2020, 6, 506–517. [Google Scholar] [CrossRef]
  104. Marar, C.; Starich, B.; Wirtz, D. Extracellular vesicles in immunomodulation and tumor progression. Nat. Immunol. 2021, 22, 560–570. [Google Scholar] [CrossRef]
  105. Kugeratski, F.G.; Kalluri, R. Exosomes as mediators of immune regulation and immunotherapy in cancer. FEBS J. 2021, 288, 10–35. [Google Scholar] [CrossRef]
  106. Mittal, S.; Gupta, P.; Chaluvally-Raghavan, P.; Pradeep, S. Emerging Role of Extracellular Vesicles in Immune Regulation and Cancer Progression. Cancers 2020, 12, 3563. [Google Scholar] [CrossRef]
  107. Jung, D.; Shin, S.; Kang, S.M.; Jung, I.; Ryu, S.; Noh, S.; Choi, S.J.; Jeong, J.; Lee, B.Y.; Kim, K.S.; et al. Reprogramming of T cell-derived small extracellular vesicles using IL2 surface engineering induces potent anti-cancer effects through miRNA delivery. J. Extracell. Vesicles 2022, 11, e12287. [Google Scholar] [CrossRef]
  108. Masjedi, A.; Hassannia, H.; Atyabi, F.; Rastegari, A.; Hojjat-Farsangi, M.; Namdar, A.; Soleimanpour, H.; Azizi, G.; Nikkhoo, A.; Ghalamfarsa, G.; et al. Downregulation of A2AR by siRNA loaded PEG-chitosan-lactate nanoparticles restores the T cell mediated anti-tumor responses through blockage of PKA/CREB signaling pathway. Int. J. Biol. Macromol. 2019, 133, 436–445. [Google Scholar] [CrossRef]
  109. Pezzuto, A.; Carico, E. Role of HIF-1 in Cancer Progression: Novel Insights. A Review. Curr. Mol. Med. 2018, 18, 343–351. [Google Scholar] [CrossRef]
  110. Yuan, C.S.; Teng, Z.; Yang, S.; He, Z.; Meng, L.Y.; Chen, X.G.; Liu, Y. Reshaping hypoxia and silencing CD73 via biomimetic gelatin nanotherapeutics to boost immunotherapy. J. Control. Release 2022, 351, 255–271. [Google Scholar] [CrossRef]
  111. Li, Z.; Cai, H.; Li, Z.; Ren, L.; Ma, X.; Zhu, H.; Gong, Q.; Zhang, H.; Gu, Z.; Luo, K. A tumor cell membrane-coated self-amplified nanosystem as a nanovaccine to boost the therapeutic effect of anti-PD-L1 antibody. Bioact. Mater. 2023, 21, 299–312. [Google Scholar] [CrossRef]
  112. Tan, Y.N.; Huang, J.D.; Li, Y.P.; Li, S.S.; Luo, M.; Luo, J.; Lee, A.W.; Fu, L.; Hu, F.Q.; Guan, X.Y. Near-Infrared Responsive Membrane Nanovesicles Amplify Homologous Targeting Delivery of Anti-PD Immunotherapy against Metastatic Tumors. Adv. Healthc. Mater. 2022, 11, e2101496. [Google Scholar] [CrossRef]
  113. Li, Y.; Zhao, R.; Cheng, K.; Zhang, K.; Wang, Y.; Zhang, Y.; Li, Y.; Liu, G.; Xu, J.; Xu, J.; et al. Bacterial Outer Membrane Vesicles Presenting Programmed Death 1 for Improved Cancer Immunotherapy via Immune Activation and Checkpoint Inhibition. ACS Nano 2020, 14, 16698–16711. [Google Scholar] [CrossRef]
  114. Bai, S.; Lu, Z.; Jiang, Y.; Shi, X.; Xu, D.; Shi, Y.; Lin, G.; Liu, C.; Zhang, Y.; Liu, G. Nanotransferrin-Based Programmable Catalysis Mediates Three-Pronged Induction of Oxidative Stress to Enhance Cancer Immunotherapy. ACS Nano 2022, 16, 997–1012. [Google Scholar] [CrossRef]
  115. Li, J.; Tang, W.; Yang, Y.; Shen, Q.; Yu, Y.; Wang, X.; Fu, Y.; Li, C. A Programmed Cell-Mimicking Nanoparticle Driven by Potato Alkaloid for Targeted Cancer Chemoimmunotherapy. Adv. Healthc. Mater. 2021, 10, e2100311. [Google Scholar] [CrossRef]
  116. 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, e2004385. [Google Scholar] [CrossRef]
  117. Guo, Y.; Fan, Y.; Wang, Z.; Li, G.; Zhan, M.; Gong, J.; Majoral, J.P.; Shi, X.; Shen, M. Chemotherapy Mediated by Biomimetic Polymeric Nanoparticles Potentiates Enhanced Tumor Immunotherapy via Amplification of Endoplasmic Reticulum Stress and Mitochondrial Dysfunction. Adv. Mater. 2022, 34, e2206861. [Google Scholar] [CrossRef]
  118. Tang, Q.; Sun, S.; Wang, P.; Sun, L.; Wang, Y.; Zhang, L.; Xu, M.; Chen, J.; Wu, R.; Zhang, J.; et al. Genetically Engineering Cell Membrane-Coated BTO Nanoparticles for MMP2-Activated Piezocatalysis-Immunotherapy. Adv. Mater. 2023, 35, e2300964. [Google Scholar] [CrossRef]
  119. Sun, M.; Shi, W.; Wu, Y.; He, Z.; Sun, J.; Cai, S.; Luo, Q. Immunogenic Nanovesicle-Tandem-Augmented Chemoimmunotherapy via Efficient Cancer-Homing Delivery and Optimized Ordinal-Interval Regime. Adv. Sci. 2022, 10, e2205247. [Google Scholar] [CrossRef]
  120. Chen, X.; Ling, X.; Xia, J.; Zhu, Y.; Zhang, L.; He, Y.; Wang, A.; Gu, G.; Yin, B.; Wang, J. Mature dendritic cell-derived dendrosomes swallow oxaliplatin-loaded nanoparticles to boost immunogenic chemotherapy and tumor antigen-specific immunotherapy. Bioact. Mater. 2022, 15, 15–28. [Google Scholar] [CrossRef]
  121. Jiang, Q.; Wang, K.; Zhang, X.; Ouyang, B.; Liu, H.; Pang, Z.; Yang, W. Platelet Membrane-Camouflaged Magnetic Nanoparticles for Ferroptosis-Enhanced Cancer Immunotherapy. Small 2020, 16, e2001704. [Google Scholar] [CrossRef]
  122. Zheng, X.; Yan, J.; You, W.; Li, F.; Diao, J.; He, W.; Yao, Y. De Novo Nano-Erythrocyte Structurally Braced by Biomimetic Au(I)-peptide Skeleton for MDM2/MDMX Predation toward Augmented Pulmonary Adenocarcinoma Immunotherapy. Small 2021, 17, e2100394. [Google Scholar] [CrossRef]
  123. Li, L.; Miao, Q.; Meng, F.; Li, B.; Xue, T.; Fang, T.; Zhang, Z.; Zhang, J.; Ye, X.; Kang, Y.; et al. Genetic engineering cellular vesicles expressing CD64 as checkpoint antibody carrier for cancer immunotherapy. Theranostics 2021, 11, 6033–6043. [Google Scholar] [CrossRef]
  124. DeFrancesco, L. CAR-T cell therapy seeks strategies to harness cytokine storm. Nat. Biotechnol. 2014, 32, 604. [Google Scholar] [CrossRef]
  125. Biederstadt, A.; Rezvani, K. Engineering the next generation of CAR-NK immunotherapies. Int. J. Hematol. 2021, 114, 554–571. [Google Scholar] [CrossRef]
  126. Gauthier, J.; Turtle, C.J. Insights into cytokine release syndrome and neurotoxicity after CD19-specific CAR-T cell therapy. Curr. Res. Transl. Med. 2018, 66, 50–52. [Google Scholar] [CrossRef]
  127. Kang, M.; Hong, J.; Jung, M.; Kwon, S.P.; Song, S.Y.; Kim, H.Y.; Lee, J.R.; Kang, S.; Han, J.; Koo, J.H.; et al. T-Cell-Mimicking Nanoparticles for Cancer Immunotherapy. Adv. Mater. 2020, 32, e2003368. [Google Scholar] [CrossRef]
  128. Hong, J.; Kang, M.; Jung, M.; Lee, Y.Y.; Cho, Y.; Kim, C.; Song, S.Y.; Park, C.G.; Doh, J.; Kim, B.S. T-Cell-Derived Nanovesicles for Cancer Immunotherapy. Adv. Mater. 2021, 33, e2101110. [Google Scholar] [CrossRef]
  129. Spits, H.; Bernink, J.H.; Lanier, L. NK cells and type 1 innate lymphoid cells: Partners in host defense. Nat. Immunol. 2016, 17, 758–764. [Google Scholar] [CrossRef]
  130. Raza, A.; Rossi, G.R.; Janjua, T.I.; Souza-Fonseca-Guimaraes, F.; Popat, A. Nanobiomaterials to modulate natural killer cell responses for effective cancer immunotherapy. Trends Biotechnol. 2023, 41, 77–92. [Google Scholar] [CrossRef]
  131. Wu, D.; Shou, X.; Zhang, Y.; Li, Z.; Wu, G.; Wu, D.; Wu, J.; Shi, S.; Wang, S. Cell membrane-encapsulated magnetic nanoparticles for enhancing natural killer cell-mediated cancer immunotherapy. Nanomedicine 2021, 32, 102333. [Google Scholar] [CrossRef]
  132. Gong, L.; Li, Y.; Cui, K.; Chen, Y.; Hong, H.; Li, J.; Li, D.; Yin, Y.; Wu, Z.; Huang, Z. Nanobody-Engineered Natural Killer Cell Conjugates for Solid Tumor Adoptive Immunotherapy. Small 2021, 17, e2103463. [Google Scholar] [CrossRef]
  133. Saxena, M.; Balan, S.; Roudko, V.; Bhardwaj, N. Towards superior dendritic-cell vaccines for cancer therapy. Nat. Biomed. Eng. 2018, 2, 341–346. [Google Scholar] [CrossRef]
  134. Fucikova, J.; Palova-Jelinkova, L.; Bartunkova, J.; Spisek, R. Induction of Tolerance and Immunity by Dendritic Cells: Mechanisms and Clinical Applications. Front. Immunol. 2019, 10, 2393. [Google Scholar] [CrossRef] [Green Version]
  135. Sun, Z.; Deng, G.; Peng, X.; Xu, X.; Liu, L.; Peng, J.; Ma, Y.; Zhang, P.; Wen, A.; Wang, Y.; et al. Intelligent photothermal dendritic cells restart the cancer immunity cycle through enhanced immunogenic cell death. Biomaterials 2021, 279, 121228. [Google Scholar] [CrossRef]
  136. Lu, Y.; Li, L.; Du, J.; Chen, J.; Xu, X.; Yang, X.; Ding, C.; Mao, C. Immunotherapy for Tumor Metastasis by Artificial Antigen-Presenting Cells via Targeted Microenvironment Regulation and T-Cell Activation. ACS Appl. Mater. Interfaces 2021, 13, 55890–55901. [Google Scholar] [CrossRef]
  137. Zhao, S.; Duan, J.; Lou, Y.; Gao, R.; Yang, S.; Wang, P.; Wang, C.; Han, L.; Li, M.; Ma, C.; et al. Surface specifically modified NK-92 cells with CD56 antibody conjugated superparamagnetic Fe(3)O(4) nanoparticles for magnetic targeting immunotherapy of solid tumors. Nanoscale 2021, 13, 19109–19122. [Google Scholar] [CrossRef]
  138. Chen, F.; Geng, Z.; Wang, L.; Zhou, Y.; Liu, J. Biomimetic Nanoparticles Enabled by Cascade Cell Membrane Coating for Direct Cross-Priming of T Cells. Small 2022, 18, e2104402. [Google Scholar] [CrossRef]
  139. Hu, Z.; Ott, P.A.; Wu, C.J. Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat. Rev. Immunol. 2018, 18, 168–182. [Google Scholar] [CrossRef]
  140. Banchereau, J.; Palucka, K. Immunotherapy: Cancer vaccines on the move. Nat. Rev. Clin. Oncol. 2018, 15, 9–10. [Google Scholar] [CrossRef]
  141. Hollingsworth, R.E.; Jansen, K. Turning the corner on therapeutic cancer vaccines. NPJ Vaccines 2019, 4, 7. [Google Scholar] [CrossRef] [Green Version]
  142. Zhang, R.; Billingsley, M.M.; Mitchell, M.J. Biomaterials for vaccine-based cancer immunotherapy. J. Control. Release 2018, 292, 256–276. [Google Scholar] [CrossRef]
  143. Viswanath, D.I.; Liu, H.C.; Huston, D.P.; Chua, C.Y.X.; Grattoni, A. Emerging biomaterial-based strategies for personalized therapeutic in situ cancer vaccines. Biomaterials 2022, 280, 121297. [Google Scholar] [CrossRef]
  144. Wu, Y.; Feng, L. Biomaterials-assisted construction of neoantigen vaccines for personalized cancer immunotherapy. Expert Opin. Drug. Deliv. 2023, 20, 323–333. [Google Scholar] [CrossRef]
  145. Xiong, X.; Zhao, J.; Pan, J.; Liu, C.; Guo, X.; Zhou, S. Personalized Nanovaccine Coated with Calcinetin-Expressed Cancer Cell Membrane Antigen for Cancer Immunotherapy. Nano Lett. 2021, 21, 8418–8425. [Google Scholar] [CrossRef]
  146. Li, J.; Huang, D.; Cheng, R.; Figueiredo, P.; Fontana, F.; Correia, A.; Wang, S.; Liu, Z.; Kemell, M.; Torrieri, G.; et al. Multifunctional Biomimetic Nanovaccines Based on Photothermal and Weak-Immunostimulatory Nanoparticulate Cores for the Immunotherapy of Solid Tumors. Adv. Mater. 2022, 34, e2108012. [Google Scholar] [CrossRef]
  147. Jiang, T.; Shi, T.; Zhang, H.; Hu, J.; Song, Y.; Wei, J.; Ren, S.; Zhou, C. Tumor neoantigens: From basic research to clinical applications. J. Hematol. Oncol. 2019, 12, 93. [Google Scholar] [CrossRef] [Green Version]
  148. Blass, E.; Ott, P.A. Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat. Rev. Clin. Oncol. 2021, 18, 215–229. [Google Scholar] [CrossRef]
  149. Xie, N.; Shen, G.; Gao, W.; Huang, Z.; Huang, C.; Fu, L. Neoantigens: Promising targets for cancer therapy. Signal Transduct. Target. Ther. 2023, 8, 9. [Google Scholar] [CrossRef]
  150. Meng, F.; Li, L.; Zhang, Z.; Lin, Z.; Zhang, J.; Song, X.; Xue, T.; Xing, C.; Liang, X.; Zhang, X. Biosynthetic neoantigen displayed on bacteria derived vesicles elicit systemic antitumour immunity. J. Extracell. Vesicles 2022, 11, e12289. [Google Scholar] [CrossRef]
  151. West, A.P.; Khoury-Hanold, W.; Staron, M.; Tal, M.C.; Pineda, C.M.; Lang, S.M.; Bestwick, M.; Duguay, B.A.; Raimundo, N.; MacDuff, D.A.; et al. Mitochondrial DNA stress primes the antiviral innate immune response. Nature 2015, 520, 553–557. [Google Scholar] [CrossRef] [Green Version]
  152. Lv, M.; Chen, M.; Zhang, R.; Zhang, W.; Wang, C.; Zhang, Y.; Wei, X.; Guan, Y.; Liu, J.; Feng, K.; et al. Manganese is critical for antitumor immune responses via cGAS-STING and improves the efficacy of clinical immunotherapy. Cell Res. 2020, 30, 966–979. [Google Scholar] [CrossRef]
  153. Chen, C.; Tong, Y.; Zheng, Y.; Shi, Y.; Chen, Z.; Li, J.; Liu, X.; Zhang, D.; Yang, H. Cytosolic Delivery of Thiolated Mn-cGAMP Nanovaccine to Enhance the Antitumor Immune Responses. Small 2021, 17, e2006970. [Google Scholar] [CrossRef]
  154. Zhao, X.; Wang, Y.; Jiang, W.; Wang, Q.; Li, J.; Wen, Z.; Li, A.; Zhang, K.; Zhang, Z.; Shi, J.; et al. Herpesvirus-Mimicking DNAzyme-Loaded Nanoparticles as a Mitochondrial DNA Stress Inducer to Activate Innate Immunity for Tumor Therapy. Adv. Mater. 2022, 34, e2204585. [Google Scholar] [CrossRef]
  155. Li, T.; Chen, G.; Xiao, Z.; Li, B.; Zhong, H.; Lin, M.; Cai, Y.; Huang, J.; Xie, X.; Shuai, X. Surgical Tumor-Derived Photothermal Nanovaccine for Personalized Cancer Therapy and Prevention. Nano Lett. 2022, 22, 3095–3103. [Google Scholar] [CrossRef]
  156. Zhou, H.; He, H.; Liang, R.; Pan, H.; Chen, Z.; Deng, G.; Zhang, S.; Ma, Y.; Liu, L.; Cai, L. In situ poly I:C released from living cell drug nanocarriers for macrophage-mediated antitumor immunotherapy. Biomaterials 2021, 269, 120670. [Google Scholar] [CrossRef]
  157. Ma, X.; Kuang, L.; Yin, Y.; Tang, L.; Zhang, Y.; Fan, Q.; Wang, B.; Dong, Z.; Wang, W.; Yin, T.; et al. Tumor-Antigen Activated Dendritic Cell Membrane-Coated Biomimetic Nanoparticles with Orchestrating Immune Responses Promote Therapeutic Efficacy against Glioma. ACS Nano 2023, 17, 2341–2355. [Google Scholar] [CrossRef]
  158. Jiang, Y.; Krishnan, N.; Zhou, J.; Chekuri, S.; Wei, X.; Kroll, A.V.; Yu, C.L.; Duan, Y.; Gao, W.; Fang, R.H.; et al. Engineered Cell-Membrane-Coated Nanoparticles Directly Present Tumor Antigens to Promote Anticancer Immunity. Adv. Mater. 2020, 32, e2001808. [Google Scholar] [CrossRef]
  159. Li, Y.; Zhang, H.; Wang, R.; Wang, Y.; Li, R.; Zhu, M.; Zhang, X.; Zhao, Z.; Wan, Y.; Zhuang, J.; et al. Tumor Cell Nanovaccines Based on Genetically Engineered Antibody-Anchored Membrane. Adv. Mater. 2023, 35, e2208923. [Google Scholar] [CrossRef]
  160. Liu, S.; Wu, J.; Feng, Y.; Guo, X.; Li, T.; Meng, M.; Chen, J.; Chen, D.; Tian, H. CD47KO/CRT dual-bioengineered cell membrane-coated nanovaccine combined with anti-PD-L1 antibody for boosting tumor immunotherapy. Bioact. Mater. 2023, 22, 211–224. [Google Scholar] [CrossRef]
  161. Liu, C.; Liu, X.; Xiang, X.; Pang, X.; Chen, S.; Zhang, Y.; Ren, E.; Zhang, L.; Liu, X.; Lv, P.; et al. A nanovaccine for antigen self-presentation and immunosuppression reversal as a personalized cancer immunotherapy strategy. Nat. Nanotechnol. 2022, 17, 531–540. [Google Scholar] [CrossRef]
  162. Park, W.; Seong, K.Y.; Han, H.H.; Yang, S.Y.; Hahn, S.K. Dissolving microneedles delivering cancer cell membrane coated nanoparticles for cancer immunotherapy. RSC Adv. 2021, 11, 10393–10399. [Google Scholar] [CrossRef]
  163. Li, W.; Fan, J.X.; Zheng, D.W.; Zhang, X.Z. Tumor Antigen Loaded Nanovaccine Induced NIR-Activated Inflammation for Enhanced Antigen Presentation During Immunotherapy of Tumors. Small 2022, 18, e2205193. [Google Scholar] [CrossRef]
  164. Hu, S.; Ma, J.; Su, C.; Chen, Y.; Shu, Y.; Qi, Z.; Zhang, B.; Shi, G.; Zhang, Y.; Zhang, Y.; et al. Engineered exosome-like nanovesicles suppress tumor growth by reprogramming tumor microenvironment and promoting tumor ferroptosis. Acta Biomater. 2021, 135, 567–581. [Google Scholar] [CrossRef]
  165. Yan, W.L.; Lang, T.Q.; Yuan, W.H.; Yin, Q.; Li, Y.P. Nanosized drug delivery systems modulate the immunosuppressive microenvironment to improve cancer immunotherapy. Acta Pharmacol. Sin. 2022, 43, 3045–3054. [Google Scholar] [CrossRef]
  166. Kim, J.; Hong, J.; Lee, J.; Fakhraei Lahiji, S.; Kim, Y.H. Recent advances in tumor microenvironment-targeted nanomedicine delivery approaches to overcome limitations of immune checkpoint blockade-based immunotherapy. J. Control. Release 2021, 332, 109–126. [Google Scholar] [CrossRef]
  167. Westcott, P.M.K.; Sacks, N.J.; Schenkel, J.M.; Ely, Z.A.; Smith, O.; Hauck, H.; Jaeger, A.M.; Zhang, D.; Backlund, C.M.; Beytagh, M.C.; et al. Low neoantigen expression and poor T-cell priming underlie early immune escape in colorectal cancer. Nat. Cancer 2021, 2, 1071–1085. [Google Scholar] [CrossRef]
  168. Zhu, Y.; Yu, X.; Thamphiwatana, S.D.; Zheng, Y.; Pang, Z. Nanomedicines modulating tumor immunosuppressive cells to enhance cancer immunotherapy. Acta Pharm. Sin. B 2020, 10, 2054–2074. [Google Scholar] [CrossRef]
  169. Huang, J.; Zhang, L.; Wan, D.; Zhou, L.; Zheng, S.; Lin, S.; Qiao, Y. Extracellular matrix and its therapeutic potential for cancer treatment. Signal Transduct. Target. Ther. 2021, 6, 153. [Google Scholar] [CrossRef]
  170. Wu, Q.; You, L.; Nepovimova, E.; Heger, Z.; Wu, W.; Kuca, K.; Adam, V. Hypoxia-inducible factors: Master regulators of hypoxic tumor immune escape. J. Hematol. Oncol. 2022, 15, 77. [Google Scholar] [CrossRef]
  171. Barsoum, I.B.; Smallwood, C.A.; Siemens, D.R.; Graham, C.H. A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res. 2014, 74, 665–674. [Google Scholar] [CrossRef] [Green Version]
  172. Leone, R.D.; Emens, L.A. Targeting adenosine for cancer immunotherapy. J. Immunother. Cancer 2018, 6, 57. [Google Scholar] [CrossRef] [Green Version]
  173. Qiu, S.Q.; Waaijer, S.J.H.; Zwager, M.C.; de Vries, E.G.E.; van der Vegt, B.; Schroder, C.P. Tumor-associated macrophages in breast cancer: Innocent bystander or important player? Cancer Treat. Rev. 2018, 70, 178–189. [Google Scholar] [CrossRef] [Green Version]
  174. Fu, L.Q.; Du, W.L.; Cai, M.H.; Yao, J.Y.; Zhao, Y.Y.; Mou, X.Z. The roles of tumor-associated macrophages in tumor angiogenesis and metastasis. Cell Immunol. 2020, 353, 104119. [Google Scholar] [CrossRef]
  175. Funes, S.C.; Rios, M.; Escobar-Vera, J.; Kalergis, A.M. Implications of macrophage polarization in autoimmunity. Immunology 2018, 154, 186–195. [Google Scholar] [CrossRef] [Green Version]
  176. Wei, B.; Pan, J.; Yuan, R.; Shao, B.; Wang, Y.; Guo, X.; Zhou, S. Polarization of Tumor-Associated Macrophages by Nanoparticle-Loaded Escherichia coli Combined with Immunogenic Cell Death for Cancer Immunotherapy. Nano Lett. 2021, 21, 4231–4240. [Google Scholar] [CrossRef]
  177. Rao, L.; Zhao, S.K.; Wen, C.; Tian, R.; Lin, L.; Cai, B.; Sun, Y.; Kang, F.; Yang, Z.; He, L.; et al. Activating Macrophage-Mediated Cancer Immunotherapy by Genetically Edited Nanoparticles. Adv. Mater. 2020, 32, e2004853. [Google Scholar] [CrossRef]
  178. Sitkovsky, M.V.; Hatfield, S.; Abbott, R.; Belikoff, B.; Lukashev, D.; Ohta, A. Hostile, hypoxia-A2-adenosinergic tumor biology as the next barrier to overcome for tumor immunologists. Cancer Immunol. Res. 2014, 2, 598–605. [Google Scholar] [CrossRef] [Green Version]
  179. Yang, G.; Xu, L.; Chao, Y.; Xu, J.; Sun, X.; Wu, Y.; Peng, R.; Liu, Z. Hollow MnO2 as a tumor-microenvironment-responsive biodegradable nano-platform for combination therapy favoring antitumor immune responses. Nat Commun. 2017, 8, 902. [Google Scholar] [CrossRef] [Green Version]
  180. Chen, Q.; Chen, J.; Yang, Z.; Xu, J.; Xu, L.; Liang, C.; Han, X.; Liu, Z. Nanoparticle-Enhanced Radiotherapy to Trigger Robust Cancer Immunotherapy. Adv. Mater. 2019, 31, e1802228. [Google Scholar] [CrossRef]
  181. Wang, Y.; Yu, J.; Luo, Z.; Shi, Q.; Liu, G.; Wu, F.; Wang, Z.; Huang, Y.; Zhou, D. Engineering Endogenous Tumor-Associated Macrophage-Targeted Biomimetic Nano-RBC to Reprogram Tumor Immunosuppressive Microenvironment for Enhanced Chemo-Immunotherapy. Adv. Mater. 2021, 33, e2103497. [Google Scholar] [CrossRef]
  182. Chen, X.; Song, E. Turning foes to friends: Targeting cancer-associated fibroblasts. Nat. Rev. Drug Discov. 2019, 18, 99–115. [Google Scholar] [CrossRef]
  183. Martinez-Outschoorn, U.E.; Lisanti, M.P.; Sotgia, F. Catabolic cancer-associated fibroblasts transfer energy and biomass to anabolic cancer cells, fueling tumor growth. Semin. Cancer Biol. 2014, 25, 47–60. [Google Scholar] [CrossRef]
  184. Lyssiotis, C.A.; Kimmelman, A.C. Metabolic Interactions in the Tumor Microenvironment. Trends Cell Biol. 2017, 27, 863–875. [Google Scholar] [CrossRef] [Green Version]
  185. Zang, S.; Huang, K.; Li, J.; Ren, K.; Li, T.; He, X.; Tao, Y.; He, J.; Dong, Z.; Li, M.; et al. Metabolic reprogramming by dual-targeting biomimetic nanoparticles for enhanced tumor chemo-immunotherapy. Acta Biomater. 2022, 148, 181–193. [Google Scholar] [CrossRef]
  186. Shi, J.; Gao, W.; Shao, F. Pyroptosis: Gasdermin-Mediated Programmed Necrotic Cell Death. Trends Biochem. Sci. 2017, 42, 245–254. [Google Scholar] [CrossRef]
  187. Wang, Y.; Gao, W.; Shi, X.; Ding, J.; Liu, W.; He, H.; Wang, K.; Shao, F. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature 2017, 547, 99–103. [Google Scholar] [CrossRef]
  188. Zhao, P.; Wang, M.; Chen, M.; Chen, Z.; Peng, X.; Zhou, F.; Song, J.; Qu, J. Programming cell pyroptosis with biomimetic nanoparticles for solid tumor immunotherapy. Biomaterials 2020, 254, 120142. [Google Scholar] [CrossRef]
  189. You, Q.; Wang, F.; Du, R.; Pi, J.; Wang, H.; Huo, Y.; Liu, J.; Wang, C.; Yu, J.; Yang, Y.; et al. m(6) A Reader YTHDF1-Targeting Engineered Small Extracellular Vesicles for Gastric Cancer Therapy via Epigenetic and Immune Regulation. Adv. Mater. 2023, 35, e2204910. [Google Scholar] [CrossRef]
  190. Bahmani, B.; Gong, H.; Luk, B.T.; Haushalter, K.J.; DeTeresa, E.; Previti, M.; Zhou, J.; Gao, W.; Bui, J.D.; Zhang, L.; et al. Intratumoral immunotherapy using platelet-cloaked nanoparticles enhances antitumor immunity in solid tumors. Nat. Commun. 2021, 12, 1999. [Google Scholar] [CrossRef]
  191. Xia, C.; Bai, W.; Deng, T.; Li, T.; Zhang, L.; Lu, Z.; Zhang, Z.; Li, M.; He, Q. Sponge-like nano-system suppresses tumor recurrence and metastasis by restraining myeloid-derived suppressor cells-mediated immunosuppression and formation of pre-metastatic niche. Acta Biomater. 2023, 158, 708–724. [Google Scholar] [CrossRef]
  192. Chen, C.; Song, M.; Du, Y.; Yu, Y.; Li, C.; Han, Y.; Yan, F.; Shi, Z.; Feng, S. Tumor-Associated-Macrophage-Membrane-Coated Nanoparticles for Improved Photodynamic Immunotherapy. Nano Lett. 2021, 21, 5522–5531. [Google Scholar] [CrossRef]
  193. Chen, A.; Yang, F.; Kuang, J.; Xiong, Y.; Mi, B.B.; Zhou, Y.; Hu, J.J.; Song, S.J.; Wan, T.; Wan, Z.Z.; et al. A Versatile Nanoplatform for Broad-Spectrum Immunotherapy by Reversing the Tumor Microenvironment. ACS Appl. Mater. Interfaces 2021, 13, 45335–45345. [Google Scholar] [CrossRef]
  194. Yue, Y.; Li, F.; Li, Y.; Wang, Y.; Guo, X.; Cheng, Z.; Li, N.; Ma, X.; Nie, G.; Zhao, X. Biomimetic Nanoparticles Carrying a Repolarization Agent of Tumor-Associated Macrophages for Remodeling of the Inflammatory Microenvironment Following Photothermal Therapy. ACS Nano 2021, 15, 15166–15179. [Google Scholar] [CrossRef]
  195. Wang, T.; Zhang, H.; Qiu, W.; Han, Y.; Liu, H.; Li, Z. Biomimetic nanoparticles directly remodel immunosuppressive microenvironment for boosting glioblastoma immunotherapy. Bioact. Mater. 2022, 16, 418–432. [Google Scholar] [CrossRef]
  196. Wang, Y.; Chen, B.; He, Z.; Tu, B.; Zhao, P.; Wang, H.; Asrorov, A.; Muhitdinov, B.; Jiang, J.; Huang, Y. Nanotherapeutic macrophage-based immunotherapy for the peritoneal carcinomatosis of lung cancer. Nanoscale 2022, 14, 2304–2315. [Google Scholar] [CrossRef]
  197. Feng, Q.; Li, Y.; Wang, N.; Hao, Y.; Chang, J.; Wang, Z.; Zhang, X.; Zhang, Z.; Wang, L. A Biomimetic Nanogenerator of Reactive Nitrogen Species Based on Battlefield Transfer Strategy for Enhanced Immunotherapy. Small 2020, 16, e2002138. [Google Scholar] [CrossRef]
  198. Liu, L.; Wang, Y.; Guo, X.; Zhao, J.; Zhou, S. A Biomimetic Polymer Magnetic Nanocarrier Polarizing Tumor-Associated Macrophages for Potentiating Immunotherapy. Small 2020, 16, e2003543. [Google Scholar] [CrossRef]
  199. Jiang, Q.; Qiao, B.; Lin, X.; Cao, J.; Zhang, N.; Guo, H.; Liu, W.; Zhu, L.; Xie, X.; Wan, L.; et al. A hydrogen peroxide economizer for on-demand oxygen production-assisted robust sonodynamic immunotherapy. Theranostics 2022, 12, 59–75. [Google Scholar] [CrossRef]
  200. Drost, J.; van Jaarsveld, R.H.; Ponsioen, B.; Zimberlin, C.; van Boxtel, R.; Buijs, A.; Sachs, N.; Overmeer, R.M.; Offerhaus, G.J.; Begthel, H.; et al. Sequential cancer mutations in cultured human intestinal stem cells. Nature 2015, 521, 43–47. [Google Scholar] [CrossRef]
  201. Jiang, Q.; Xie, M.; Chen, R.; Yan, F.; Ye, C.; Li, Q.; Xu, S.; Wu, W.; Jia, Y.; Shen, P.; et al. Cancer cell membrane-wrapped nanoparticles for cancer immunotherapy: A review of current developments. Front. Immunol. 2022, 13, 973601. [Google Scholar] [CrossRef]
  202. Xiao, Y.; Zhu, T.; Zeng, Q.; Tan, Q.; Jiang, G.; Huang, X. Functionalized biomimetic nanoparticles combining programmed death-1/programmed death-ligand 1 blockade with photothermal ablation for enhanced colorectal cancer immunotherapy. Acta Biomater. 2023, 157, 451–466. [Google Scholar] [CrossRef]
  203. Lu, Z.; Bai, S.; Jiang, Y.; Wu, S.; Xu, D.; Zhang, J.; Peng, X.; Zhang, H.; Shi, Y.; Liu, G. Amplifying Dendritic Cell Activation by Bioinspired Nanometal Organic Frameworks for Synergistic Sonoimmunotherapy. Small 2022, 18, e2203952. [Google Scholar] [CrossRef]
  204. Wang, X.; Zhu, X.; Li, B.; Wei, X.; Chen, Y.; Zhang, Y.; Wang, Y.; Zhang, W.; Liu, S.; Liu, Z.; et al. Intelligent Biomimetic Nanoplatform for Systemic Treatment of Metastatic Triple-Negative Breast Cancer via Enhanced EGFR-Targeted Therapy and Immunotherapy. ACS Appl. Mater. Interfaces 2022, 14, 23152–23163. [Google Scholar] [CrossRef]
  205. Li, Z.; Rong, L. A Homotypic Membrane-Camouflaged Biomimetic Nanoplatform with Gold Nanocrystals for Synergistic Photothermal/Starvation/Immunotherapy. ACS Appl. Mater. Interfaces 2021, 13, 23469–23480. [Google Scholar] [CrossRef]
  206. Li, Y.; Wu, J.; Qiu, X.; Dong, S.; He, J.; Liu, J.; Xu, W.; Huang, S.; Hu, X.; Xiang, D.X. Bacterial outer membrane vesicles-based therapeutic platform eradicates triple-negative breast tumor by combinational photodynamic/chemo-/immunotherapy. Bioact. Mater. 2023, 20, 548–560. [Google Scholar] [CrossRef]
  207. Pan, P.; Dong, X.; Chen, Y.; Ye, J.J.; Sun, Y.X.; Zhang, X.Z. A heterogenic membrane-based biomimetic hybrid nanoplatform for combining radiotherapy and immunotherapy against breast cancer. Biomaterials 2022, 289, 121810. [Google Scholar] [CrossRef]
  208. Cao, H.; Gao, H.; Wang, L.; Cheng, Y.; Wu, X.; Shen, X.; Wang, H.; Wang, Z.; Zhan, P.; Liu, J.; et al. Biosynthetic Dendritic Cell-Exocytosed Aggregation-Induced Emission Nanoparticles for Synergistic Photodynamic Immunotherapy. ACS Nano 2022, 16, 13992–14006. [Google Scholar] [CrossRef]
  209. Fu, L.; Zhang, W.; Zhou, X.; Fu, J.; He, C. Tumor cell membrane-camouflaged responsive nanoparticles enable MRI-guided immuno-chemodynamic therapy of orthotopic osteosarcoma. Bioact. Mater. 2022, 17, 221–233. [Google Scholar] [CrossRef]
  210. Mu, X.; Zhang, M.; Wei, A.; Yin, F.; Wang, Y.; Hu, K.; Jiang, J. Doxorubicin and PD-L1 siRNA co-delivery with stem cell membrane-coated polydopamine nanoparticles for the targeted chemoimmunotherapy of PCa bone metastases. Nanoscale 2021, 13, 8998–9008. [Google Scholar] [CrossRef]
  211. Yang, C.; Ming, Y.; Zhou, K.; Hao, Y.; Hu, D.; Chu, B.; He, X.; Yang, Y.; Qian, Z. Macrophage Membrane-Camouflaged shRNA and Doxorubicin: A pH-Dependent Release System for Melanoma Chemo-Immunotherapy. Research 2022, 2022, 9768687. [Google Scholar] [CrossRef]
  212. Song, X.; Qian, R.; Li, T.; Fu, W.; Fang, L.; Cai, Y.; Guo, H.; Xi, L.; Cheang, U.K. Imaging-Guided Biomimetic M1 Macrophage Membrane-Camouflaged Magnetic Nanorobots for Photothermal Immunotargeting Cancer Therapy. ACS Appl. Mater. Interfaces 2022, 14, 56548–56559. [Google Scholar] [CrossRef]
  213. Huang, X.; Chen, L.; Lin, Y.; Tou, K.I.; Cai, H.; Jin, H.; Lin, W.; Zhang, J.; Cai, J.; Zhou, H.; et al. Tumor targeting and penetrating biomimetic mesoporous polydopamine nanoparticles facilitate photothermal killing and autophagy blocking for synergistic tumor ablation. Acta Biomater. 2021, 136, 456–472. [Google Scholar] [CrossRef]
  214. Cheng, Y.; Chen, Q.; Guo, Z.; Li, M.; Yang, X.; Wan, G.; Chen, H.; Zhang, Q.; Wang, Y. An Intelligent Biomimetic Nanoplatform for Holistic Treatment of Metastatic Triple-Negative Breast Cancer via Photothermal Ablation and Immune Remodeling. ACS Nano 2020, 14, 15161–15181. [Google Scholar] [CrossRef]
  215. Sun, Z.; Liu, J.; Li, Y.; Lin, X.; Chu, Y.; Wang, W.; Huang, S.; Li, W.; Peng, J.; Liu, C.; et al. Aggregation-Induced-Emission Photosensitizer-Loaded Nano-Superartificial Dendritic Cells with Directly Presenting Tumor Antigens and Reversed Immunosuppression for Photodynamically Boosted Immunotherapy. Adv. Mater. 2023, 35, e2208555. [Google Scholar] [CrossRef]
  216. Yao, Y.; Chen, H.; Tan, N. Cancer-cell-biomimetic nanoparticles systemically eliminate hypoxia tumors by synergistic chemotherapy and checkpoint blockade immunotherapy. Acta Pharm. Sin. B 2022, 12, 2103–2119. [Google Scholar] [CrossRef]
  217. Wang, H.; Wang, K.; He, L.; Liu, Y.; Dong, H.; Li, Y. Engineering antigen as photosensitiser nanocarrier to facilitate ROS triggered immune cascade for photodynamic immunotherapy. Biomaterials 2020, 244, 119964. [Google Scholar] [CrossRef]
  218. Hu, C.; Lei, T.; Wang, Y.; Cao, J.; Yang, X.; Qin, L.; Liu, R.; Zhou, Y.; Tong, F.; Umeshappa, C.S.; et al. Phagocyte-membrane-coated and laser-responsive nanoparticles control primary and metastatic cancer by inducing anti-tumor immunity. Biomaterials 2020, 255, 120159. [Google Scholar] [CrossRef]
  219. Dai, J.; Wu, M.; Wang, Q.; Ding, S.; Dong, X.; Xue, L.; Zhu, Q.; Zhou, J.; Xia, F.; Wang, S.; et al. Red blood cell membrane-camouflaged nanoparticles loaded with AIEgen and Poly(I:C) for enhanced tumoral photodynamic-immunotherapy. Natl. Sci. Rev. 2021, 8, nwab039. [Google Scholar] [CrossRef]
Figure 1. The unique advantages of biomimetic cell-derived nanoparticles. By Figdraw.
Figure 1. The unique advantages of biomimetic cell-derived nanoparticles. By Figdraw.
Pharmaceutics 15 01821 g001
Figure 2. The fabrication of biomimetic cell-derived nanoparticles. By Figdraw.
Figure 2. The fabrication of biomimetic cell-derived nanoparticles. By Figdraw.
Pharmaceutics 15 01821 g002
Figure 3. IL2-sEVs to enhance the anti-cancer efficacy of aPD-L1. (a) The construction of IL2-sEVs. (b) IL2-sEVs altered the miRNA profile of T cell-derived sEVs. (c) IL2-sEVs down-regulated PD-L1 levels in melanoma cells. (d) IL2-sEVs inhibited sEV secretion. (e,f) IL2-sEVs enhanced the therapeutic efficacy of aPD-L1. (g) Qualitative analysis of PD-L1 in tumor tissues after combined aPD-L1 treatment. ns: no significant, * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001. Reproduced under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License [107].
Figure 3. IL2-sEVs to enhance the anti-cancer efficacy of aPD-L1. (a) The construction of IL2-sEVs. (b) IL2-sEVs altered the miRNA profile of T cell-derived sEVs. (c) IL2-sEVs down-regulated PD-L1 levels in melanoma cells. (d) IL2-sEVs inhibited sEV secretion. (e,f) IL2-sEVs enhanced the therapeutic efficacy of aPD-L1. (g) Qualitative analysis of PD-L1 in tumor tissues after combined aPD-L1 treatment. ns: no significant, * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001. Reproduced under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License [107].
Pharmaceutics 15 01821 g003
Figure 4. BDVs-Neo vaccine combined with aPD-L1 for immunotherapy. (a) The preparation of BDVs-Neo vaccine. (b,c) DC activity and maturation induced by BDVs-Neo in lymph nodes. (d) BDVs-Neo combined with aPD-L1 for anti-tumor recurrence. (e,f) Synergistic mechanism of BDVs-Neo in combination with aPD-1. (g) BDVs-Neo vaccine combined with aPD-L1 for lung metastasis. (#1) Gel-PBS, (#2) Gel-Blank BDVs, (#3) Gel-Normal-M33-M47 BDVs, (#4) Gel-Mutation-M33-M47 BDVs, (#5) aPD-1, (#6) Gel-Mutation-M33-M47 BDVs + aPD-1. NS: no significant, * p < 0.05, ** p < 0.01, *** p < 0.001. Reproduced under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License [150].
Figure 4. BDVs-Neo vaccine combined with aPD-L1 for immunotherapy. (a) The preparation of BDVs-Neo vaccine. (b,c) DC activity and maturation induced by BDVs-Neo in lymph nodes. (d) BDVs-Neo combined with aPD-L1 for anti-tumor recurrence. (e,f) Synergistic mechanism of BDVs-Neo in combination with aPD-1. (g) BDVs-Neo vaccine combined with aPD-L1 for lung metastasis. (#1) Gel-PBS, (#2) Gel-Blank BDVs, (#3) Gel-Normal-M33-M47 BDVs, (#4) Gel-Mutation-M33-M47 BDVs, (#5) aPD-1, (#6) Gel-Mutation-M33-M47 BDVs + aPD-1. NS: no significant, * p < 0.05, ** p < 0.01, *** p < 0.001. Reproduced under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License [150].
Pharmaceutics 15 01821 g004
Figure 5. The immunosuppressive tumor microenvironment (ITME) and biomimetic cell-derived nanoparticles for reversing the ITME. By Figdraw.
Figure 5. The immunosuppressive tumor microenvironment (ITME) and biomimetic cell-derived nanoparticles for reversing the ITME. By Figdraw.
Pharmaceutics 15 01821 g005
Figure 6. PDA/GNS@aPD-L1 nanoparticles for cancer immunotherapy by combining PTT with PD-1/PD-L1 blockade. (a) The preparation of PDA/GNS@aPD-L1. (b) PDA/GNS@aPD-L1 binding capability for PD-L1-expressing cells. (c) PTT effects of PDA/GNS@aPD-L1. Scale bar: 100 μm. (d) Anti-cancer effect of PDA/GNS@aPD-L1 in primary tumor. (e,f) PDA/GNS@aPD-L1 reversed the ITME by decreasing the number of Treg and MDSC cells at the tumor site. (g) Anti-cancer effect of PDA/GNS@aPD-L1 in distal tumor. G1, PBS group; G2, PDA-GNS + NIR group; G3, PDA/GNS@Free NPs group; G4, PDA/GNS@aPD-L1 NPs group; G5, aPD-L1 mAb group; G6, aPD-L1 NVs + PDA-GNS + NIR group; G7, aPD-L1 mAb + PDA-GNS + NIR group; G8, PDA/GNS@aPD-L1 NPs + NIR group. ns: no significant, * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001. Reproduced under the terms of the CC BY-NC-ND license [202].
Figure 6. PDA/GNS@aPD-L1 nanoparticles for cancer immunotherapy by combining PTT with PD-1/PD-L1 blockade. (a) The preparation of PDA/GNS@aPD-L1. (b) PDA/GNS@aPD-L1 binding capability for PD-L1-expressing cells. (c) PTT effects of PDA/GNS@aPD-L1. Scale bar: 100 μm. (d) Anti-cancer effect of PDA/GNS@aPD-L1 in primary tumor. (e,f) PDA/GNS@aPD-L1 reversed the ITME by decreasing the number of Treg and MDSC cells at the tumor site. (g) Anti-cancer effect of PDA/GNS@aPD-L1 in distal tumor. G1, PBS group; G2, PDA-GNS + NIR group; G3, PDA/GNS@Free NPs group; G4, PDA/GNS@aPD-L1 NPs group; G5, aPD-L1 mAb group; G6, aPD-L1 NVs + PDA-GNS + NIR group; G7, aPD-L1 mAb + PDA-GNS + NIR group; G8, PDA/GNS@aPD-L1 NPs + NIR group. ns: no significant, * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001. Reproduced under the terms of the CC BY-NC-ND license [202].
Pharmaceutics 15 01821 g006
Figure 7. Ce6/DOX-OMVs@M for combined tumor chemo/photodynamic/immunotherapy. (a) The preparation of Ce6/DOX-OMVs@M. (b) Generation of ROS by Ce6/DOX-OMVs@M in 4T1 cells. Scale bar: 100 μm. (c) Anti-tumor effect of Ce6/DOX-OMVs@M. (d) Ce6/DOX-OMVs@M for lung metastasis. (e) OMVs activated pyroptosis in tumors in vivo. * p < 0.05, ** p < 0.01, *** p < 0.001. Reproduced under the terms of the CC BY-NC-ND license [206].
Figure 7. Ce6/DOX-OMVs@M for combined tumor chemo/photodynamic/immunotherapy. (a) The preparation of Ce6/DOX-OMVs@M. (b) Generation of ROS by Ce6/DOX-OMVs@M in 4T1 cells. Scale bar: 100 μm. (c) Anti-tumor effect of Ce6/DOX-OMVs@M. (d) Ce6/DOX-OMVs@M for lung metastasis. (e) OMVs activated pyroptosis in tumors in vivo. * p < 0.05, ** p < 0.01, *** p < 0.001. Reproduced under the terms of the CC BY-NC-ND license [206].
Pharmaceutics 15 01821 g007
Figure 8. Biomimetic cell-derived nanoparticles for cancer bio-imaging, prevention, diagnosis, and treatment. By Figdraw.
Figure 8. Biomimetic cell-derived nanoparticles for cancer bio-imaging, prevention, diagnosis, and treatment. By Figdraw.
Pharmaceutics 15 01821 g008
Table 1. Application of biomimetic cell-derived nanoparticles in ICIs.
Table 1. Application of biomimetic cell-derived nanoparticles in ICIs.
Cell MembraneCore NanoparticleCharacteristics of the Core NanoparticleDrug/Imaging Agent EncapsulatedCancer Models and the Reason for Selecting Each Cancer Cell LineBiomedical ApplicationRef.
Platelet membraneRab27 siRNA (siRab) polycationic nanocomplexes and aPD-L1 nanogelsDesired siRNA encapsulation and cell-transfection abilities, good biocompatibility, rapid clearance by macrophagessiRab and aPD-L1B16F10 cells, leading tumorsiRab silenced Rab27a, inhibited exosome secretion, relieved immunosuppression, and sensitized ICI.[102]
T cell-derived sEVs----Membrane-bound IL2B16F10 and B16F10-luc-g5 cells, leading tumorIL2-sEVs inhibited sEV secretion and PD-L1 expression by cancer cells through altering the miRNA profile of T cell-derived sEVs, and, consequently, sensitized ICI.[107]
Cancer cell membraneGelatin nanoparticlesGood biocompatibility, matrix metallopeptidase (MMP)-sensitive, low passive targeting efficiency, rapid clearance by macrophagesCAT and CD73 siRNAB16F10 cells, homologous targetingCAT relieved tumor hypoxia, while CD73 siRNA silenced CD73 protein and promoted T cell-specific immune response, which synergically sensitized ICI.[110]
Calreticulin over-expressed cancer cell membraneZeolitic imidazolate framework (ZIF-8) nanoparticlesLarge surface area, pH-induced biodegradability, low passive targeting efficiencyEpirubicin (EPI), glucose oxidase (Gox) and hemin4T1 cells, homologous targetingThe biomimetic delivery system displayed an amplified ICD and activated the tumor immune microenvironment, boosting the therapeutic efficacy of ICI.[111]
Cancer cell membraneThermosensitive lipid nanoparticlesThermosensitive, controlled drug release, good biocompatibility, low passive targeting efficiencyPD-1/PD-L1 inhibitor BMS202 and IR7804T1 and MCF-7 cells, homologous targeting, and multiple mouse modelsThe biomimetic delivery system inhibited cancer-associated fibroblasts (CAFs), increased the penetration of TILs, blocked the pathway of PD-1/PD-L1, and then sensitized ICI.[112]
Bacterial outer membrane----Membrane-bound PD1 ectodomainB16F10 and CT26 cells, multiple mouse modelsThe biomimetic delivery system could effectively block the PD-1/PD-L1 pathway, protect T cells, and sensitize ICI.[113]
HEK 293T cell membraneIR820-dihydroartemisinin (DHA) complexespH-sensitive, excellent biodegradability, sonodynamic therapy (SDT) nanoplatform, low passive targeting efficiency, rapid clearance by macrophagesIR820 and DHAHep1–6 cells, one of the major tumorsDHA-mediated chemo-dynamic therapy and IR820-induced sonodynamic therapy synergistically achieved a robust ICD effect and sensitized ICI.[114]
Erythrocyte membraneLiposomesGood biocompatibility and biodegradability, pH-sensitive, low passive targeting efficiency, rapid clearance by macrophagesPaclitaxel (PTX)PC-3 cells, one of the major tumorsThe biomimetic delivery system reinforced active tumor-targeting behavior and sensitized ICI.[115]
Cancer cell membraneMesoporous organosilica nanoparticles (MONs)Good biocompatibility, large surface area, X-ray-induced biodegradability, low passive targeting efficiencyDoxorubicin (DOX)4T1 cells, homologous targetingThe biomimetic delivery system exhibited enhanced DOX-mediated ICD and sensitized ICI.[116]
Cancer cell membranePolymeric nanoparticlesRedox-responsive, good biocompatibility, low passive targeting efficiencyToyocamycin (Toy) and generation 3 phosphorus dendrimer-copper (II) complexes (1G3-Cu)B16F10 cells, homologous targetingToy-triggered amplification of ER stress and 1G3-Cu-mediated mitochondrial dysfunction synergistically induced significant ICD, which thereby sensitized ICI.[117]
aPD-L1 over-expressed HEK 293T cell membrane anchored with a cleavable peptide by matrix metallopeptidase 2 (MMP2)Barium titanate (BTO) nanoparticlesPiezocatalysis materials, generating a cascade of redox reactions under US, rapid clearance by macrophagesBTO and membrane-bound aPD-L1B16F10 cells, leading tumorThe membrane-bound aPD-L1 could effectively block the PD-1/PD-L1 pathway, while the encapsulated BTO could achieve a piezocatalysis effect and induce tumor antigen release, which synergistically improved ICI therapy.[118]
Cancer cell-derived EVs with PD-L1 knockoutLiposomesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesDOX4T1 cells, homologous targetingThe biomimetic delivery system up-regulated the expression of PD-L1 in the tumor and amplified the ICD mediated by DOX, which thereby sensitized ICI.[119]
Mature dendritic cell membraneRedox-responsive nanoparticlesRedox-responsive, low passive targeting efficiency, rapid clearance by macrophagesOxaliplatin (OXA) prodrugsCT26 cells, homologous targetingThe biomimetic delivery system effectively sensitized the TME to ICI by immunogenic chemotherapy and tumor antigen-specific immunotherapy.[120]
Platelet membraneMagnetic nanoparticlesGood biocompatibility, effective ferroptosis, magnetic targeting, rapid clearance by macrophagesSulfasalazine (SAS)4T1 cells, classic tumor modelThe biomimetic delivery system-mediated ferroptosis elicited an effective immune response and sensitized ICI.[121]
Erythrocyte membraneOligomeric Au (I)-PMIV complexesGood biocompatibility, effective, self-assembling, low passive targeting efficiency, rapid clearance by macrophagesPMIV (a peptide that can degrade MDM2/MDMX)NCI-H1650 cells, LUAD-patient-derived xenograft mice model, and LUAD-PDX mice model, multiple mouse modelsThe biomimetic delivery system potently restored p53 and p73, and sensitized ICI.[122]
HEK 293T cell membrane expressing CD64----Membrane-bound aPD-L1 and cyclophosphamideB16F10-luci cells, leading tumorThe biomimetic delivery system restrained the Tregs and invigorated Ki67+CD8+ T cells, then improved ICI therapy.[123]
Table 2. Application of biomimetic cell-derived nanoparticles in ACT.
Table 2. Application of biomimetic cell-derived nanoparticles in ACT.
Cell MembraneCore NanoparticleCharacteristics of the Core NanoparticleDrug/Imaging Agent EncapsulatedCancer Models and the Reason for Selecting Each Cancer Cell LineBiomedical ApplicationRef.
T cell membranePoly(lactic-co-glycolic) acid (PLGA) nanoparticlesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesDacarbazine (DTIC)B16F10 cells, DTIC for metastatic melanoma treatmentThe biomimetic delivery system could induce Fas-ligand-mediated apoptosis and scavenge TGF-β1 and PD-L1 to restore the function of CTLs. [127]
T cell membrane over-expressing PD-1, TGF-β receptor, and granzyme B----Granzyme BLLC cancer cells, immunotherapy for lung carcinomaThe biomimetic delivery system prevented CD8+ T cell exhaustion mediated by PD-L1/TGF-β, and promoted tumor cell apoptosis via granzyme B. [128]
Cancer cell membraneMagnetic nanoparticlesGood biocompatibility, magnetic targeting, rapid clearance by macrophages--HepG2 and A375 cells, homologous targeting, and multiple mouse modelsThe biomimetic delivery system effectively activated NK cells and enhanced NK cell-based ACT through the enhanced secretion of soluble cytotoxic effectors. [131]
NK92MI cell membrane equipped with the anti-EGFR nanobody 7D12------EGFRpositive cancer cell lines, including LoVo, MDA-MB-468, A549, and A431 cells, 7D12 for specifically recognizing EGFRpositive cancer cellsThe biomimetic delivery system exhibited high targeting for EGFR-positive cancer cells, caused enhanced secretion of cytotoxic effectors, and improved NK cell-based ACT. [132]
Mature DC membranePolymeric nanoparticlesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesIR-7974T1 cells, specific recognition between tumor specific antigens and tumor cellsMature DC membrane facilitated effective cross-priming of T cells, recruited T cells, and produced immunostimulatory cytokines, while mild PTT mediated by IR-797 induced a greater degree of tumoricidal effect, which consequently amplified ICD and reinitiated the self-sustaining cycle of cancer immune responses. [135]
Cancer cell membraneMnOx nanoparticles functionalized with anti-CD3/CD28 mAbsGood biocompatibility, low passive targeting efficiency, rapid clearance by macrophagesanti-CD3/CD28 mAbsB16F10 cells, homologous targetingThe biomimetic delivery system not only efficiently promoted the expansion and activation of CD8+ T cells and DCs but also reversed the immunosuppressive microenvironment to promote T cell survival. [136]
NK92MI cell membraneAnti-CD56 antibodies modified Fe3O4 nanoparticlesGood biocompatibility, magnetic targeting, rapid clearance by macrophages--K562, B16F10, and H22 cells, NK92 cell possessing a broader-spectrum anti-cancer activityThe biomimetic delivery system up-regulated the secretion of granzyme B and IFN-γ at the tumor site, which thereby significantly improved the antitumor effect. [137]
Mature DC membraneCancer cell membrane coated PLGA nanoparticlesGood biocompatibility and biodegradability, high active targeting efficiency--HPV E6- and E7-expressing TC-1, B16-OVA, and Hepa 1–6 cells, multiple mouse modelsThe biomimetic delivery system could directly cross-prime T cells without the help of APCs and elicit robust antigen-specific T cell immunotherapy. [138]
Table 3. Application of biomimetic cell-derived nanoparticles in cancer vaccines.
Table 3. Application of biomimetic cell-derived nanoparticles in cancer vaccines.
Cell MembraneCore NanoparticleCharacteristics of the Core NanoparticleDrug/Imaging Agent EncapsulatedCancer Models and the Reason for Selecting Each Cancer Cell LineBiomedical ApplicationRef.
CRT-expressed cancer cell membranePLGA nanoparticlesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesAdjuvant R837Luc-4T1 cells, homologous targetingThe CRT expressed on the surface of the nanovaccine facilitated the internalization of the nanovaccine by DCs. R837 released from the nanovaccine excited TLR7 and further activated DCs. [145]
Cancer cell membraneSilicon@Au nanocompositesGood biocompatibility, low passive targeting efficiency, rapid clearance by macrophagesAu4T1 cells, homologous targetingThe biomimetic delivery system based on nanocomposites with weak immune stimulation could still induce DCs maturation to activate the downstream anti-tumor immune responses. [146]
BDVs presenting the neoantigens----Adjuvant GM-CSFB16F10-luc cells, leading tumorThe biomimetic delivery system efficiently recruited DCs and promoted the proliferation of TILs and memory T cells. [150]
Erythrocyte membraneManganese-doped imidazolate frameworks-90 nanoparticlesGood biocompatibility, low passive targeting efficiency, rapid clearance by macrophagesDNAzyme4T1 cells, classic tumor modelThe biomimetic delivery system closely mimicked the process of herpesvirus infection, ultimately initiating cGAS-STING pathway-mediated innate immunotherapy. [154]
Cancer cell membraneMesoporous polydopamine nanoparticlesGood biocompatibility and biodegradability, PTT nanoplatform, low passive targeting efficiency, rapid clearance by macrophagesAdjuvant R8484T1 cells, homologous targetingThe biomimetic delivery system effectively stimulated the immune response and demonstrated excellent photothermal immunotherapy, which significantly promoted the activation and maturation of lymph node DCs, and stimulated CD8+ T cells and memory T cells. [155]
Bone marrow-derived macrophage (BMDM) membranePoly (lactic-co-glycolic acid) nanoparticles (PLP NPs)Good biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesAdjuvant Poly I: C (PIC)4T1 cells, classic tumor modelThe PIC released from the biomimetic nanoparticles could not only directly induce tumor cell apoptosis, but also polarize exogenous BMDMs into killing M1 macrophages and activate endogenous APCs to trigger robust anti-tumor immune responses. [156]
Tumor-antigen activated DC membranePLGA nanoparticlesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesRapamycin (RAPA)C6-LUC cells, biomimetic nanoparticles possessing the potential to cross the BBBThe biomimetic delivery system could activate CD8+ T cells directly or indirectly to reconstitute the glioma tumor immune microenvironment and increase the proportion of NK cells to reduce the exhausted T cells, resulting in a significant anti-glioma efficacy. [157]
CD80 engineered cancer cell-membrane------B16-OVA cells, homologous targetingThe biomimetic nanoparticle platform could directly activate T cells without the help of professional APCs. [158]
Anti-CD40 scFv-anchored cancer cell membranePLGA nanoparticlesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophages--MC38 and Panc02 cells, homologous targeting, and multiple mouse modelsThe biomimetic nanoparticle platform effectively promoted DCs maturation in CD40-humanized transgenic mice, improved the engagement and expansion of cognate T-cell immune responses, and facilitated subsequent adaptive immune responses. [159]
CD47KO/CRT dual-bioengineered cancer cell membraneHyperbranched PEI25k nanoparticlesGood biocompatibility, large surface area, low passive targeting efficiency, rapid clearance by macrophagesUnmethylated cytosine-phosphate-guanine (CpG) adjuvantB16F10 cells, homologous targetingThe biomimetic nanoparticle platform significantly stimulated APCs, resulting in the activation of CD8+ T cells and intense anti-tumor immune responses. [160]
MHC-I-Ag-anchored mature DC membrane----aPD-1B16F10 cells, leading tumorThe biomimetic nanoparticle platform could present neoantigens to CD8+ T cells directly, resulting in strong CTL responses. In addition, the immunosuppressive reversal function of anti-PD-1 antibody was enhanced by CD28/B7 co-stimulation. [161]
Cancer cell membranePoloxamer 407 nanoparticlesGood biocompatibility, low passive targeting efficiency, rapid clearance by macrophagesAdjuvant R837HCT116 cells, homologous targetingThe biomimetic nanoparticle platform was presented with APCs to secret immunofactors and to activate the lymphatic immune network, resulting in significant tumor regression. [162]
Cancer cell membraneManganese dioxide (MnO2) nanoparticlesGood biocompatibility, low passive targeting efficiency, rapid clearance by macrophagesPolythiopheneB16F10 cells, homologous targetingThe biomimetic nanoparticle platform could stimulate a specific T cell anti-tumor immune response by promoting APCs maturation, autologous tumor antigen presentation, as well as generating local microinflammation. [163]
Cancer cell membrane expressing fibroblast activation protein-α (FAP)------CT26, B16F10, LLC, and 4T1 cells, 90% of human tumor tissues overexpressing FAP and multiple mouse modelsThe biomimetic nanoparticle platform suppressed tumor growth by triggering robust and specific T cell anti-tumor immune responses. Moreover, the biomimetic nanoparticle platform facilitated tumor ferroptosis by releasing IFN-γ from CTLs and eliminating FAP+CAFs. [164]
Table 4. Application of biomimetic cell-derived nanoparticles in modulating the ITME.
Table 4. Application of biomimetic cell-derived nanoparticles in modulating the ITME.
Cell MembraneCore NanoparticleCharacteristics of the Core NanoparticleDrug/Imaging Agent EncapsulatedCancer Models and the Reason for Selecting Each Cancer Cell LineBiomedical ApplicationRef.
Bacterial outer membranePLGA nanoparticlesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesDOX and R8484T1 cells, classic tumor modelThe biomimetic nanoparticle platform could polarize M2 macrophages into M1 macrophages, promote the secretion of TNF-α and IL-6, which thereby activated the anti-tumor immune responses. [176]
Cancer cell membrane overexpressing SIRPα variantsMagnetic nanoparticlesGood biocompatibility, magnetic targeting, rapid clearance by macrophages--B16F10 and 4T1 cells, homologous targeting, and multiple mouse modelsIn this system, the gCM shell genetically overexpressing SIRPα variants efficiently blocked the CD47-SIRPα pathway, while the nanoparticle cores polarized M2 macrophages into M1 macrophages, which thereby synergistically facilitated macrophage phagocytosis of tumor cells and triggered T-cell immune response. [177]
Erythrocyte membraneAmphiphilic PCL nanoparticlesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesDOX4T1 cells, classic tumor modelThe Hb moiety of the biomimetic nanoparticle platform could effectively kill the tumor cells. In addition, the O2 released by Hb alleviated cancer hypoxia and further augmented the anti-tumor immune response. [181]
4T1 and 3T3 hybrid cell membraneSolid lipid nanoparticlesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesPTX and glycolysis inhibitor PFK154T1 cells, homologous targetingThe biomimetic nanoparticle platform possessed outstanding dual-targeting ability. The encapsulated PFK15 could inhibit glycolysis in both CAFs and cancer cells, resulting in elevated immune responses. [185]
Cancer cell membranePLGA nanoparticlesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesICG and DCT4T1 cells, homologous targetingICG induced a sharp increase in cytosolic Ca2+ concentration, promoted cytochrome c release and subsequent caspase-3 activation. DCT synergistically up-regulated the expression of gasdermin E by inhibiting DNA methylation, enhancing the cleavage of gasdermin E by caspase-3, and causing robust cancer pyroptosis. [188]
Macrophage cell-derived sEVs----siRNA against YTHDF1MGC-803 and HGC-27 cells, YTHDF1 promoting tumor progression in a variety of cancers and multiple mouse modelsThis nanosystem efficiently depleted YTHDF1 expression and stimulated strong cytotoxic T lymphocytes responses through hampering frizzled7 translation and inactivating the Wnt/β-catenin pathway in an m6A dependent manner. [189]
Platelet membranePLA nanoparticlesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesR848MC38 and 4T1 cells, multiple mouse modelsThe biomimetic nanoparticle platform promoted the robust activation of APCs and increased immune infiltration, resulting in complete tumor regression. [190]
Activated neutrophils membraneRedox-responsive polymer nanoparticlesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesDOX4T1 cells, classic tumor modelThe biomimetic nanoparticle platform could prevent the recruitment and functions of MDSCs, thereby inhibiting tumor recurrence and metastasis. [191]
Tumor-associated macrophage membrane with high macrophage colony-stimulating factor 1 receptor (CSF1R)Rare-earth-upconversion nanoparticlesGood biocompatibility, “nanotransducer”, theranostic nanoplatform, low passive targeting efficiency, rapid clearance by macrophagesRose Bengal4T1 cells, classic tumor modelThe biomimetic nanoparticle platform could block the interaction between TAMs and cancer cells via depleting the CSF1 secreted by cancer cells. In addition, the biomimetic nanoparticle platform-mediated photodynamic therapy (PDT) could polarize M2 macrophages into M1 macrophages. [192]
Erythrocyte membraneCopper peroxide (CP) nanoparticlesGood biocompatibility, PDT nanoplatform, low passive targeting efficiency, rapid clearance by macrophagesH2O2 and protoporphyrin (PpIX)B16F10 cells, leading tumorThe biomimetic nanoparticle platform could relieve tumor hypoxia and polarize M2 macrophages into M1 macrophages. Furthermore, the ROS generated by photosensitizers could cause the ICD of tumor cells. [193]
Macrophage cell membranePolydopamine nanoparticlesGood biocompatibility and biodegradability, PTT nanoplatform, low passive targeting efficiency, rapid clearance by macrophagesTMP195 (a TAMs repolarization agent)4T1 cells, classic tumor modelThe loaded TMP195 significantly polarized M2 macrophages into M1 macrophages and subsequently remodeled the ITME in residual tumor after PTT, which thereby rescued CTLs. [194]
Cancer cell membraneCu2-xSe nanoparticlesGood biocompatibility, effective ferroptosis, low passive targeting efficiency, rapid clearance by macrophagesIndoximod (IND) and JQ1GL261 cells, homologous targetingThe smart nanoparticle platform could remodel the ITME by increasing M1 macrophages and preventing Tregs cell infiltration. In addition, it also served as a checkpoint inhibitor to inhibit the expression of PD-L1 on cancer cells. [195]
M1-like macrophage cell membrane----CelastrolLLC and GL261 cells, multiple mouse modelsM1 macrophages not only acted as drug delivery carriers for celastrol but also as therapeutic agents. In turn, celastrol could maintain the anticancer polarization of M1 macrophages. [196]
Erythrocyte membraneTiO2 nanoparticlesGood biocompatibility, SDT nanoplatform, low passive targeting efficiency, rapid clearance by macrophagesHb and RRx-0014T1 cells, classic tumor modelUpon reaching the hypoxic TME, the Hb in this smart nanoparticle platform was deoxygenated, further initiating a series of reactions that generated a large number of RNS in cascade fashion. Subsequently, oxygen compensation by Hb enhanced TiO2-mediated ROS for SDT, and the resulting highly active RNS triggered ICD in cancer cells. [197]
M1-like macrophage cell membraneMagnetic polymer nanoparticlesGood biocompatibility and biodegradability, magnetic targeting, rapid clearance by macrophagesR8374T1 cells, classic tumor modelThe intracellular uptake of this smart nanoparticle platform could greatly polarize M2 macrophages into M1 macrophages with the synergy of R837 and Fe3O4, alleviating the immunosuppression of the TME for immune recovery. [198]
Cancer cell membraneFe-doped polydiaminopyridine nanoparticlesGood biocompatibility, SDT nanoplatform, low passive targeting efficiency, rapid clearance by macrophagesCe64T1 cells, homologous targetingThe prepared smart nanoparticle platform could significantly reduce the unnecessary consumption of H2O2 in the TME, while US irradiation could promote the exposure of Fe-PDAP and provide more adequate O2 supply, enabling efficient ROS production and DCs maturation. [199]
Table 5. Application of biomimetic cell-derived nanoparticles in combination therapy.
Table 5. Application of biomimetic cell-derived nanoparticles in combination therapy.
Cell MembraneCore NanoparticleCharacteristics of the Core NanoparticleDrug/Imaging Agent EncapsulatedCancer Models and the Reason for Selecting Each Cancer Cell LineBiomedical ApplicationRef.
Anti-PD-L1 scFv-anchored HEK 293T cell membranePolydopamine-modified gold nanoparticlesGood biocompatibility and biodegradability, PTT nanoplatform, low passive targeting efficiency, rapid clearance by macrophagesAu and membrane-bound aPD-L1 scFvMC38 cells, immunotherapy for colorectal cancerThe developed smart nanoparticle platform combined PD-1/PD-L1 blockade with PTT, which triggered the immune stimulatory responses and remodeled the ITME.[202]
Anti-DEC205 anchored cancer cell membranePorphyrin-based metal organic frameworks (MOFs)Good biocompatibility and biodegradability, SDT nanoplatform, low passive targeting efficiency, rapid clearance by macrophagesR848 and AuPtHep1-6 cells, homologous targetingAMR-MOF@AuPt-mediated multimodal therapies based on sonoimmunotherapy induced systemic immune responses and long-term memory immunity.[203]
Cancer cell membrane anchored with a cleavable peptide by MMP2PLGA nanoparticlesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesAfatinib (AFT) and 2-bromopalmitate (2-BP)4T1 cells, homologous targetingThe developed smart nanoparticle platform combined targeted therapy with immunotherapy, which remarkably prevented tumor growth and metastasis.[204]
Hybrid pH-sensitive membraneMesoporous silica nanoparticlesGood biocompatibility and biodegradability, large surface area, low passive targeting efficiency, rapid clearance by macrophagesR837 and Au4T1 cells, homologous targetingThe developed smart nanoparticle platform realized the synergistic effect of starvation/PTT/immunotherapy and induced a long-term immune memory effect.[205]
Macrophage cell membraneBacterial outer membrane vesiclesGood biocompatibility and biodegradability, potent anti-cancer potential, low targeting efficiencyCe6 and DOX4T1 cells, classic tumor modelThe developed smart nanoparticle platform realized the synergism of PDT/chemo-/immunotherapy.[206]
Hybrid cell membraneGlutathione (GSH) decorated Te nanoparticlesGood biocompatibility, radiotherapy sensitization, radiotherapy nanoplatform, low passive targeting efficiency, rapid clearance by macrophagesGTe4T1 cells, homologous targetingThe developed smart nanoparticle platform realized the synergism of radiotherapy and immunotherapy.[207]
DC-derived sEVsMBPN-TCyP nanoparticle complexesGood biocompatibility, PDT nanoplatform, mitochondrion-targeting, rapid clearance by macrophagesMBPN-TCyP (an AIE photosensitizer)4T1 and CT26 cells, multiple mouse modelsThe developed smart nanoparticle platform realized the synergism of PDT/immunotherapy and inhibited the self-renewal of cancer stem cells for tumor suppression.[208]
Cancer cell membraneHollow manganese dioxide (HMnO2) nanoparticlesGood biocompatibility, GSH-sensitive, excellent loading capacity, low passive targeting efficiency, rapid clearance by macrophagesGinsenoside Rh2 (Rh2)K7M2 cells, homologous targetingThe developed smart nanoparticle platform realized attractive immuno-chemo-dynamic combined therapeutic efficiency.[209]
Stem cell membranePolydopamine nanoparticlesGood biocompatibility and biodegradability, PTT nanoplatform, low passive targeting efficiency, rapid clearance by macrophagesDOX and PD-L1 siRNAPC-3 cells, one of the major tumorsThe developed smart nanoparticle platform showed attractive immuno-chemo-dynamic combined therapeutic efficiency for PCa bone metastases.[210]
M1-like macrophage cell membranePolyethylenimine (PEI) nanoparticlesGood biocompatibility, large surface area, rapid clearance by macrophagesDOX and short-hairpin RNA (shRNA) targeting Ptpn2B16F10 cells, leading tumorThe developed smart nanoparticle platform realized the synergism of chemotherapy and gene immunotherapies.[211]
M1-like macrophage cell membraneMagnetic photothermal nanocomplexesGood biocompatibility, PTT nanoplatform, low passive targeting efficiency, rapid clearance by macrophagesDOX4T1 cells, classic tumor modelThe developed smart nanoparticle platform enhanced ROS generation and exhibited outstanding chemo-phototherapy efficacy.[212]
Cancer cell membraneMesoporous polydopamine nanoparticlesGood biocompatibility and biodegradability, PTT nanoplatform, low passive targeting efficiency, rapid clearance by macrophagesChloroquine (CQ, an autophagy inhibitor)RM-1 cells, homologous targetingThe developed smart nanoparticle platform facilitated PTT and autophagy blockade for synergistic tumor elimination.[213]
Hybrid cell membranePolydopamine nanoparticlesGood biocompatibility and biodegradability, PTT nanoplatform, low passive targeting efficiency, rapid clearance by macrophages--B16F10 cells, homologous targetingThe developed smart nanoparticle platform realized the synergism of OMV-mediated immunotherapy and hollow polydopamine-mediated PTT.[84]
Anti-PD-1 peptide AUNP-12 modified cancer cell membraneDendritic large-pore mesoporous silica nanoparticlesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesCopper sulfide (CuS) and R8484T1 cells, homologous targetingThe developed smart nanoparticle platform realized the synergism of PDT and immunotherapy.[214]
Cancer cell membrane expressing CD86 and anti-LAG3Polymeric nanoparticlesGood biocompatibility, PDT nanoplatform, mitochondrion-targeting, rapid clearance by macrophagesFs (an AIE photosensitizer)4T1 cells, homologous targetingThe developed smart nanoparticle platform realized the synergism of PDT and immunotherapy by directly presenting tumor antigens and reversed immunosuppression.[215]
Cancer cell membraneLiposomesGood biocompatibility and biodegradability, pH-sensitive, low passive targeting efficiency, rapid clearance by macrophagesRA-V (a chemotherapeutic drug) and BMS202CT26 cells, homologous targetingThe developed smart nanoparticle platform systemically eliminated hypoxia tumors by synergistic chemotherapy and checkpoint blockade immunotherapy.[216]
Cancer cell membraneOvalbumin antigen (OVA) nanoparticlesGood biocompatibility and biodegradability, rapid clearance by macrophagesCe6B16-OVA cells, homologous targetingThe developed smart nanoparticle platform facilitated ROS-triggered immune cascade for photodynamic immunotherapy.[217]
M1-like macrophage cell membraneNanoparticle complexesGood biocompatibility, laser-sensitive, size-changeable, rapid clearance by macrophagesCe6, DOX, and d indoleamine 2,3-dioxygenase 1 inhibitor4T1 and B16F10 cells, multiple mouse modelsThe developed smart nanoparticle platform realized the synergism of PDT/chemo-/immunotherapy.[218]
Erythrocyte membranePLGA nanoparticlesGood biocompatibility and biodegradability, low passive targeting efficiency, rapid clearance by macrophagesP2-PPh3 (an AIE photosensitizer) and Poly (I:C)B16F10 cells, leading tumorThe developed smart nanoparticle platform combined the PDT properties of P2-PPh3 with the immunotherapy properties of Poly (I:C).[219]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hu, T.; Huang, Y.; Liu, J.; Shen, C.; Wu, F.; He, Z. Biomimetic Cell-Derived Nanoparticles: Emerging Platforms for Cancer Immunotherapy. Pharmaceutics 2023, 15, 1821. https://doi.org/10.3390/pharmaceutics15071821

AMA Style

Hu T, Huang Y, Liu J, Shen C, Wu F, He Z. Biomimetic Cell-Derived Nanoparticles: Emerging Platforms for Cancer Immunotherapy. Pharmaceutics. 2023; 15(7):1821. https://doi.org/10.3390/pharmaceutics15071821

Chicago/Turabian Style

Hu, Tingting, Yuezhou Huang, Jing Liu, Chao Shen, Fengbo Wu, and Zhiyao He. 2023. "Biomimetic Cell-Derived Nanoparticles: Emerging Platforms for Cancer Immunotherapy" Pharmaceutics 15, no. 7: 1821. https://doi.org/10.3390/pharmaceutics15071821

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

Article Metrics

Back to TopTop