CC BY 4.0 · Pharmaceutical Fronts 2024; 06(02): e101-e118
DOI: 10.1055/s-0044-1786681
Review Article

Advances in Tumor Targeting Biomimetic Drug Delivery Systems: A Promising Approach for Antitumor Therapy

Ziyi Mo
1   Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu, People's Republic of China
,
Jiao He
1   Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu, People's Republic of China
,
Man Li
1   Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu, People's Republic of China
,
Rong Guo
2   Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, People's Republic of China
,
Qin He
1   Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu, People's Republic of China
› Institutsangaben
Funding The work was supported by the National Natural Science Foundation (Grant No. 82173771) and the Key Project of Applied Basic Research in Sichuan province (Grant No. 22YYJC1231).
 

Abstract

Cancer is one of the most fatal diseases that attract numerous efforts and attention from researchers. Among plentiful therapeutic agents, chemotherapy is frequently used in treating virulent tumors, and its insistent administration is useful in the ablation of cancers; however, it also produces side effects. Biomimetic drug delivery systems (BDDSs) provide an alternative route for antitumor therapy. Their endogenous substances may be extracellular vesicles, living cells, cell membranes, etc., which optimize single-agent chemotherapy. They “upgrade” traditional drug delivery platforms by combining the original drug with itself, disguised as a Trojan Horse, to trick the immune system or tumor tissues to achieve higher targeting and lower immunogenicity. Herein, we review three BDDS strategies being used recently in antitumor drug development and their advances, aiming at providing general guidelines and opportunities in this field in the future.


#

Introduction

Since its discovery, cancer has been considered to be one of the most lethal diseases with a high mortality rate and a heavy social burden. Unfortunately, its occurrence is rising dramatically across the globe, preventing people from reaching a higher life expectancy.[1] It brings patients not only physical and mental pain but also a burden on their lives. Chemotherapy, especially combination chemotherapy, is the most prevalent strategy in cancer treatment,[2] which uses small molecules to eliminate tumors. However, these small-molecule drugs would cause some adverse reactions under normal circumstances, leading to a decrease in patient compliance.

With the widespread application of nanomaterials in the medical industry, nanoplatforms have been the most commonly used materials in drug delivery.[3] The platforms are usually meticulously fabricated. By manipulating these platforms, nano-drug delivery systems (NDDSs) can achieve enhanced permeability and retention (EPR), improved circulation capability, reduced toxicity as well as increased penetration of biological barriers when compared to traditional chemotherapy.[4] [5] [6] Thus, these drug delivery systems (DDSs) have the advantages of prolonging the half-life of drugs, boosting drug accumulation, and conducting controllable release at the target site, leading to minimized side effects on the nontargeted tissues.[7] These advantages give DDS an outstanding ability to treat various diseases, especially cancer, making it a benign influence in medical applications.

Nowadays, significant advancements have been made in the field, with multiple nanomaterials being adapted to DDS. They can be categorized as “hard nanomaterials” (including metals, metal oxides, carbon, graphene quantum dots, etc.) and “soft nanomaterials” (including liposomes and polymer nanoparticles [NPs], etc.).[8] Although the traditional DDS mentioned above facilitates better cancer treatment, setbacks and challenges still exist, including poor tumor penetration, high immunogenicity, rapid clearance of the reticuloendothelial system, unsatisfactory toxic side effects, etc.[9] Nevertheless, the “off-target” effect is the most critical one that needs to be addressed. The “off-target” effect, in which a drug relocates an unintended drug target or “noncanonical” target, is due to the similarity of the pharmacological target protein or pathway between the proper target sites and other parts of the body. After intravenous administration, the structure of the NDDS is greatly affected by the complex environment in the body, resulting in inefficient drug delivery and off-target biological distribution.[10] This “off-target” property damages normal tissue and organs, produces numerous side effects, reduces the quality of life of patients, and affects the effectiveness of chemotherapy to a certain extent.[11] [12] The unsolved limitations have triggered researchers to escalate and alter the original nanoplatforms.

Biomimetic DDSs (BDDSs) came into people's view. BDDS made full use of the “Trojan Horse” tactics by coating traditional DDS or chemotherapeutic drugs with cell membranes to better “deceive” the body's immune system and lesions (e.g. tumors, inflammations, etc.) to obtain higher targeting accuracy, longer circulating time, and better EPR effects and bioavailability. Drugs are encapsulated into extracellular vesicles (EVs) secreted by the host cells, or attached to intrinsic proteins or the living cells ([Scheme 1]). In general, compared to traditional DDS, BDDS significantly increases the proportion of “self-components” and improves drug delivery performances in terms of reducing the “off-target” effect, promoting precision therapy, reducing immunogenicity, and boosting drug accumulation at tumor sites by using the camouflage with autologous cells or components (e.g., the homing characteristics of tumor cells), and this high-potency medication is available in smaller doses, or longer duration of therapy, and triggers fewer side effects.

Zoom Image
Scheme 1 A brief introduction of the biomimetic drug delivery system.

EVs are natural drug carriers with well-regarded intrinsic abilities, including high stability with negatively charged surfaces and the ability to avoid clearance, and are now regarded as an acclaimed platform to deliver drugs to tumor sites.[13] [14] Then, the cell membrane-coated platforms show their advantages in interacting with biological substrates and provide DDS with desirable targeting ability.[15] In addition, living cell-based carriers are rich in surface ligands that can effectively interact with specific cells or tissues according to their different physiological functions, giving them the potential to target different tumors.[16] Given above, it is crucial to prevent damage to living cells during drug loading, and correspondingly, the immunogenicity of these living cell carriers is the greatest among the aforementioned BDDS. Herein, based on antitumor therapy, we select and summarize different kinds of BDDS exhibiting high tumor-targeted delivery and therapeutic efficiency to demonstrate the latest advancements in this field.


#

EVs for Targeted Drug Delivery

EVs are particles composed of artificially manufactured lipid bilayers, and secreted by a variety of cells currently identifiable in the human body,[17] and can be classified as exosomes (50–150 nm), microvesicles (150–500 nm, or even >10 μm), and exomeres (∼35 nm) according to the sizes.[18] EVs, as communicators between different cells in the body, are considered prospective drug carriers with therapeutic implications. The past decades have witnessed a great deal of enthusiasm and passion among researchers for the advantages of EVs in blocking various diseases, particularly malignant tumors.

EVs have a stronger enhanced delivery ability compared with traditional DDS-like liposomes. Liposomes can only rely on some coatings to avoid rapid clearance and a few targeting ligands to increase cellular uptake, while EVs can not only express various endogenous self-markers to escape the recognition by the immune system but also use these makers to interact with recipient cells and promote their internalization.[19] Piffoux et al modified EVs by fusion with liposomes.[20] They found that after the incorporation of EV, the uptake of modified liposomes was significantly increased (approximately 50%) compared to liposomes without EV incorporation, suggesting the good performance of EVs in enhancing cellular uptake of the carriers and also implying that EVs have better cellular uptake than liposomes.

EVs are internalized by various pathways, including passive membrane fusion and endocytosis. After being internalized, some EVs can escape from the endosomal/lysosomal pathway,[21] and at the same time have an immuno-evasive function and good biocompatibility.[22] The surface of EVs can be modified by a variety of engineering techniques to enhance cell-specificity or prevent nonspecific uptake,[23] to further strengthen their delivery ability. The Endosomal Sorting Complex Required for Transport (ESCRT) Pathway is the best-known pathway for EV production and cargo sorting, which facilitates the loading of certain proteins and RNAs with potential therapeutic options.[24] EVs with these characteristics have made certain progress in clinical trials.[25] For example, a phase I trial (NCT03608631) conducted by MD Anderson Cancer Center investigated the efficacy of mesenchymal stromal cell-derived exosomes with KrasG12D siRNA (iExosomes) in participants with pancreatic cancer whose KrasG12D mutation had spread to other parts of the body. In addition, a phase II trial (NCT01159288) to vaccinate tumor antigen-loaded dendritic cell (DC)-derived exosomes in patients with unresectable non-small cell lung cancer has now been completed.

Due to imperfection of isolation processes, the materials used in a large number of studies contain compounds from different kinds of EVs. Therefore, the article used the term EVs to symbolize any of the types we mentioned above.[26]

Mesenchymal Stem Cell-Derived EVs for Targeted Drug Delivery

First discovered in 1976,[27] the mesenchymal stem cell (MSC) is regarded as a kind of multipotent cell with differentiation potential and self-renewal abilities,[28] and can be isolated from a variety of different organs or tissues such as bone marrow, heart, lung, and adipose tissue,[29] with the characteristics of “double-edged sword.” On the one hand, MSCs have immunosuppressive and anti-inflammatory properties,[30] which inhibit the activation of T cells, and change the phenotype of macrophages and DCs,[31] and are the basis for the treatments of inflammatory disorders. On the other hand, it establishes a tumor microenvironment (TME), redounds tumor growth and metastasis,[32] and participates in TME formation.[33] Nonetheless, it still holds the ability to improve antitumor therapy due to its outstanding tumor tropism ability, low immunogenicity, and applicability for large-scale production.[34] However, the safety of using it for cell-based antitumor therapy remains debated in academia.

MSC-derived EVs (MEVs) are similar to MSCs in many aspects. However, it is worth noting that it has the advantages of small size, prominent half-life, inferior immunogenicity, and good penetration compared to the application of MSC as a delivery platform.[35] In mice experiments, the cell-like lipid membrane also provides them with down-regulated cytotoxicity. More importantly, MEVs seem to act as a “mailman” to consign the information including DNA, RNA, proteins, etc. to targeted cells,[36] and therefore bear the innate capacity to target malignancy sites in vivo,[37] making them natural vehicles for tumor-targeted drug delivery. The ability of MEVs to transport functional endogenous biomacromolecules has been confirmed by many researchers. Of all the components, nucleic acid is the most abundant. Jahangiri et al suggested that miRNA (miR)-100 and miR-143 were transferred from MEVs to human colorectal cancer cells, and inhibited cell proliferation and metastasis by manipulating the miR-100/mTOR/miR-143 axis.[38] Accordingly, Yao et al showed for the first time that circRNA circ-0030167, isolated from bone marrow MEVs, acts as a molecular sponge for miR-338-5p while increasing the expression of a tumor suppressor gene WIF1, thereby significantly arresting the progression of pancreatic cancer ([Fig. 1]).[39] In addition, their potential to upload exogenous compounds is being simultaneously investigated. Pascucci et al found that paclitaxel (PTX)-treated MSCs were characterized by high drug concentrations and low cell mortality, and were able to secrete EVs that also contain PTX to inhibit tumor cell proliferation and growth.[40] Meanwhile, Pinto et al examined the effectiveness of MEV-encapsulated meta-tetra(hydroxyphenyl)chlorin (mTHPC) in combination with photodynamic therapy (PDT) against peritoneal carcinomatosis. They reported that due to the outstanding tumor-targeted function of MEVs, few organs were harmed under the violent stimulation of PDT, and a stronger anti-metastasis effect was detected.[41] However, it is worth noting that MEVs, secreted by cells treated with doxorubicin (DOX), would enhance breast cancer resistance when delivered to the tumor,[42] suggesting that the antitumor effect may be related to the type of cancer and the drugs loaded into the EVs.

Zoom Image
Fig. 1 A schematic representation of the molecular mechanisms by which MSC-Exos affects cell proliferation and metastasis in CRC cells. CRC, colorectal cell. (Reproduced with permission from Jahangiri et al[38], copyright 2022 Elsevier B.V. All rights reserved.)

#

DC-Derived EVs for Targeted Drug Delivery

DCs are specialized antigen-presenting cells (APCs) that patrol humans' bodies and are responsible for adaptive immune responses,[43] including immune surveillance,[44] antigen presentation,[45], stimulation of naïve T cells,[46] etc. They are characterized by a stellate morphology and sustained expression of major histocompatibility complex class II (MHCII), and can uptake antigens and migrate to the draining lymph nodes to prime naïve T cells to generate a strong immune response.[47] Due to the great potential of DCs in eradicating tumors, a great deal of effort has been invested in the development of DC-based antitumor vaccines. However, vaccine manufacturers might face issues like high cost, relatively time-consuming,[48] and the risk of in vivo DCs being influenced by immunosuppressive factors produced by the tumor.[49] In this case, DC-derived EVs (DEVs) have many of the key immunostimulatory properties of DCs, with the benefits of a long shelf-life when frozen and relatively simple good manufacturing practices (GMP) handling, and therefore lower cost but the same or greater anticancer efficacy.[50] DEVs would be a better option for malignancy therapies.

DCs are capable of secreting EVs with multiple functions,[51] mainly targeting immune cells. Compared with DCs, DEVs have inherited characteristics from their parent cells, especially in terms of surface molecules such as costimulatory molecules, MHCI, MHCII, etc.[48] Membrane proteins bound to externalized phosphatidylserine, such as milk fat globule EGF factor 8, act as intermediary between DEVs and αvβ3 or αvβ5 integrins on the membranes of recipient cells, facilitating the penetration of DEVs into the target cells, and thus are essential for the enhancement of the targeted ability of DEVs.[52] Thus, proper modifications to the surface of DEVs could further enhance their tumor elimination effect. For example, by applying the DSPE–PEG–NHS linker, Fan and colleagues inserted an anti-CD3 antibody into the membrane of DEVs to activate T cell response while also embedding an anti-epidermal growth factor receptor (anti-EGFR) antibody to direct mature T cells to the tumor sites ([Fig. 2]).[53] Notably, Zhu et al showed that the direct conjugation of MUC1 glycopeptide (a molecule overexpressed in various cancers) on the surface of DEVs could upregulate the titers of immunoglobulin G antibody and inhibit tumor growth.[54] Infection of DCs with lentivirus-containing special genes prior to isolation of EVs is another tool for effective DEV surface engineering.[55] Despite investigation claiming that DEVs produced according to GMP can trigger proliferation and activation of natural killer (NK) cells to destroy tumors, this is mainly because the NK group 2 member D (NKG2D) ligands and IL-15Rα that are originally bound on the membrane,[56] artificially engineered surfaces of DEV, still proved to be a more effective way of targeted delivery.

Zoom Image
Fig. 2 Schematic diagram. (A) Schematic of the construction of anti-CD3 and anti-EGFR antibody-engineered tDC-Exo (Exo-OVA-aCD3/aEGFR). (B) MHC–antigen complex and a co-stimulating molecule CD86 on Exo-OVA-aCD3/aEGFR can be regarded as the CAR of CAR T cells which could activate endogenous T cells in vivo. (Reproduced with permission from Fan et al[53], copyright 2022 Elsevier B.V. All rights reserved.)

Unlike MEV strategies, which load drugs more often, DEVs are usually used to carry antigens to enhance the efficacy of immunotherapy. One of the most important jobs of DEVs is to demonstrate their immunostimulatory potential to stimulate cytotoxic T lymphocyte responses, thereby controlling the tumor as a “lifeless” antigen presentation machine. Therefore, DEVs and their antigen fragments need to maintain a certain degree of immunogenicity to achieve better therapeutic efficacy. Antigens not only from tumors but also from other immune adjuvant-loaded DEVs are also commonly used as vaccines to prevent and treat established tumors by activating naïve T cells in a (cross) presentation manner.[50] [57] Li et al reported that DEVs loaded with a multi-neoantigen peptide exhibited excellent antitumor responses in melanoma models, stimulating a broad-spectrum immune response and preventing immune escape.[48] Interestingly, Damo et al loaded DEVs with antigens from the melanoma model B16F10, and when these EVs were incubated with toll-like receptor agonist poly(I:C), they showed a proinflammatory Th1 response in tumor-bearing animals. This suggests that the antitumor effect may be related to their maturation environment.[58]


#

Tumor Cell-Derived EVs for Targeted Drug Delivery

Tumor cells usually secrete more EVs than other cells in the body due to the formation of a TME that creates hostile conditions like high acidity, hypoxia, genotoxic stress, etc. Survival pressure caused by therapeutic factors like applying chemotherapy drugs may also lead to increased secretion of tumor EVs (TEVs).[59] [60] EVs from those malignant cells are usually associated with tumor initiation, progression, metastasis drug resistance, etc., leading to poor patient prognosis.[61] [62] Specifically, TEVs have the role of inducing tumor immune tolerance, promoting tumor angiogenesis and vascular permeability, and developing chemoresistance during the natural generation by the tumor cells.[63] [64] [65] However, concerns arise when those exosomes are delivered to the body as free, circulating particles, where causing tumor metastasis is one of the biggest obstacles to the use of those TEVs as drug delivery platforms because lipid-rich TEVs can subvert normal cells and bring them into an abnormal state in integrin and miRNA-dependent manner.[66] [67] [68]

Despite this, TEVs have great potential for targeted drug delivery. The homing characteristics remain decisive. Qiao et al designed a “Trojan horses” platform to encapsulate DOX into TEVs on the basis that TEVs are more likely to interact with the tumor cells than other cells due to the shared protein and lipid composition. More importantly, the level of TEVs absorbed by tumor cells was closely related to their types, with the highest absorption efficiency occurring in TEVs secreted by the aimed tumor, indicating the magnitude of using specific TEVs in the preparation of drug carriers.[69] In addition, TEV, as an endogenous antigen itself, can be used to sensitize immune cells, such as DCs, to boost antitumor efficacy. Derived from the tumor cells, specific antigens shared by the TEVs are displayed on their surfaces and can be presented to CD8+ T cells by DCs through an MHCI-dependent way.[70] From this perspective, on the one hand, when TEVs are used to deliver therapeutic agents, it is important to minimize the immunogenicity of TEV to ensure that more of the drug can reach the tumor site. We can do this by inducing tumors by transplanting targeted cancer cell lines into nude mice and isolating TEVs from tumor cells harvested from the transplanted tumor. On the other hand, a certain degree of immunogenicity should be kept to stimulate immune cells to activate the intrinsic antitumor response. Finally, some chemotherapy drugs (e.g., DOX) have high selectivity toward TEVs and high stability in these carriers,[71] further confirming the excellent encapsulation ability of TEVs.

In summary, EVs are natural carriers secreted by multi-types of cells, with great potential for targeted drug delivery and broad application prospects. Their innate malignancy-targeting ability, tumor penetration redounding, and easy surface modification enable them to achieve comprehensive antitumor functions. Although EVs are superior to traditional DDS in at least the aspects mentioned above, their low yield and unsatisfactory retention time in the body still limit their clinical use. Therefore, further research studies are needed before entering clinical studies.


#
#

Cell-Membrane-Coated NPs for Targeted Drug Delivery

In addition to using cell-mediated drug delivery to treat cancers, cell membranes with naturally complex structures and functional properties have gradually attracted the attention of some researchers in the field of targeted administration. Different cell membranes prepared from erythrocytes, platelets, cancer cells, immune cells, etc., provide NPs with a series of cell-specific proteins that can be leveraged for dynamic and multiplex binding interactions, resulting in function-driven and broad-spectrum bio-activity.[72] [73] The prepared cell-membrane-coated coated NPs (CMCNs) are usually made up of two parts, including (1) synthetic NP cores, the inorganic or organic NPs that function as antitumor drugs, and (2) layer of natural cell membranes, camouflage clothing that mimics the antigenic diversity of the source cells. The combination of these two elements displays characteristics of the parent cells on artificial NPs.[74]

CMCNs are constructed following an effective top-down strategy, which has the potential to simplify the development of drug delivery platforms with the required performances that can be customized for a wide range of applications. The preparation of CMCNs mainly involves two stages: membrane derivation and membrane coating. The first step usually contains methods like homogenization, hypotonic lysis, and centrifugation, the latter possesses procedures like co-extrusion, sonication, microfluidic electroporation, cell membrane-templated polymerization, etc.[75] While natural membranes can be used directly as coatings for NPs, they can also be modified through lipid insertion, membrane fusion, or genetic engineering to form hybrid clothing to obtain better performance.[15] [75] During the manufacturing process, however, attention should be paid to NPs partially coated with membranes when those membranes were subjected to mechanical forces such as extrusion or sonication.[76]

In general, CMCNs provide more opportunities for custom-tailored therapies. In this review, we focus on membrane coats of erythrocytes, cancer cells, leukocytes, and the hybrid membranes, because they are wildly investigated.

Erythrocyte Membrane-Coated NPs for Targeted Drug Delivery

Erythrocyte membranes (EMs) are commonly used as a naturally mimicking material in drug delivery because of their unique benefits of extending from the original cells.[77] First, the human body contains a large amount of erythrocytes, and the raw materials are readily available, making the preparation process economic and elementarily achieved. Second, membrane proteins on EMs are crucial in helping them evade the immune system. CD47 is a protein highly expressed in EMs that interacts with signal regulatory protein-α (SIRPα) on macrophages to inhibit phagocytes.[78] [79] [80] However, there are still limitations, i.e., EMs from donors should be matched to the patients' blood type and Rh compatibility to reduce the chance of inducing alloimmunization, which hinders large-scale production and prevents researchers from translating this advanced strategy into clinical applications.[81] [82] There are fixed standards and specifications for EM extraction. Briefly, whole blood is centrifuged to remove serum and fluffy precipitation, then erythrocytes are subjected to hypotonic treatment to shed intracellular components. Subsequently, after washing, sonicating, or extruding through porous membranes, EMs can be coated on the NPs.[77] [80]

EM-based passive delivery is widely used by many researchers, such as combining EMs and photothermal therapy according to EM carriers without targeting pieces. Because under irradiation of near-infrared laser, EMs would be disrupted, thus increasing the release of the drug.[83] Unfortunately, the effectiveness of this strategy which mainly relies on the EPR effect for passive targeted delivery faces controversy due to the lack of tumor-specific adhesion molecules and low drug-loading capacity. EM-based nanocarriers require special structural modifications to enhance their tendency toward tumors and the ability to deliver drugs to the site.[84] RGD peptide is a tumor-penetrating and cell-internalizing peptide that interacts with αvβ3/αvβ5 integrin receptor overexpressed in tumor tissues. Among all the targeting molecules, RGD peptide is commonly used and shows good performance during drug delivery. Xie and colleagues coated tranexamic acid + DOX NPs with EMs modified by cRGD peptide to induce tumor thrombotic infarction by precisely targeting and damaging tumor vascular endothelium.[85] Instead of delivering chemotherapeutics directly to the tumor tissues, Wang's group first used EMs to cloak worm-like siRNA; however, in this case, the surface of the siRNA vector is positively charged, leading to the adsorption of serum proteins and a much shorter circulation time. By adjusting the ratio of siRNA to cationic bovine serum albumin (cBSA), the surface charge can be controlled to be negative at neutral pH but positive at low pH, resulting in the release of EMs released and lysosomal escape through the proton sponge effect. Therefore, RGD is also applied in the construction of carriers.[86] In addition, targeting molecule combinations, such as combining anti-EGFR and RGD peptide, also improves delivery.[87]

Interestingly, deformability is an important characteristic of natural erythrocytes, and EMs derived from different life stages will have different delivery functions. Deformability, pH, etc. are critical conditions that should be emphasized when using EMs as a basis for targeted delivery.[88]


#

Cancer Cell Membrane-Coated NPs for Targeted Drug Delivery

Cancer cell membranes (CCMs) are likewise considered effective drug delivery carriers. The anti-immune clearance and homotypic binding capacity of the malignant cells are of great importance during tumor formation and progression (including tumor growth and metastasis). CCMs have been reported to play a key role in fostering these competencies of tumors. Specific molecules such as integrin αvβ3, N-cadherin, epithelial cell adhesion molecule (EpCAM), and galectin-3 have been demonstrated to help cancer cells camouflage and recognize homologous cancer cells under the surveillance of the immune system to lower their immunogenicity. Therefore, CCMs should endow NPs with superior homotypic targeting and immune escape abilities.[89] [90] [91] In this case, CCMs should be the basis of the DDS. In addition, tumor cells, tumor cell membranes, and whole tumor lysates are considered to be perfect polyvalent antigens, and therefore rather than shipping drugs into the tumors, their membranes could be used as cancer vaccines, which is a feasible cancer therapy. For example, cloaking aluminum phosphate absorbing adjuvant CpG can provide comprehensive tumor antigens to APC and other relevant immune cells, and enhance specific antitumor immunity.[92]

Covering NPs with CCMs has proven to be a valid and commonly used method of drug delivery. The coated NPs gain a membrane phospholipid bilayer structure as well as cancer surface proteins to be decoys or Trojan horses for precise treatment of cancer. Jin et al demonstrated that simply fabricating CCMs on poly (lactic-co-glycolic acid) (PLGA) NPs can interfere with cancer cell − stromal cell interactions to reduce fibroblast-mediated invasion and metastasis while activating the following-up immune response ([Fig. 3]).[93]

Zoom Image
Fig. 3 (A) Schematic illustration of the preparation of cancer cell plasma membrane fraction-coated PLGA NPs (CCMF-PLGA NPs). (B) The purpose was to determine the ability of these cancer cell-mimicking NPs to disrupt cancer cell–stromal cell interactions, reduce metastasis, and prime the immune system for cancer immunotherapy. NPs, nanoparticles. (Reproduced with permission from Jin et al[93], copyright 2019 American Chemical Society.)

Compared to other NDDSs, CCM-coated NPs are more capable of crossing biological barriers such as the blood–brain barrier, blood–brain–tumor barrier, etc., without redundant decorations on CCMs themselves. Wang and colleagues designed a novel brain tumor imaging and surgical navigation system by coating Er-based lanthanide-doped NPs (LnNPs), NPs which have excellent near-infrared-IIb luminescence performance, with CCMs. Thanks to the excellent tumor-homing ability, the system can visualize brain tumor boundaries and guide surgical resection.[94] Some researchers, when trying to increase the accumulation of NPs at the tumor site, choose ligands like Asn-Gly-Arg (NGR) on the CCMs for better targeting.[95] In the process of surface modification, to improve the target capability of CCM, it is necessary to pay attention to the modification efficiency. Zheng et al reported that when decorating bladder cancer membranes that will subsequently be camouflaged on PLGA NPs, proceeding on live tumor cells before isolating the membranes will help to sustain the correct positioning of modifiers at the extracellular side of the membrane.[96]

As we mentioned before, these carriers may increase the risk of tumor metastasis and progression, and thus face some controversies. TEVs contain nucleic acids and proteins that are already present in parent tumor cells and may be a set of pro-tumor progression, pro-metastasis, and pro-drug resistance messengers. Encouragingly, CCMs with simple membrane structures can be a solution to this dilemma. In conclusion, CCM-coated NPs are foreseeable promising materials for future precise medicine manufacture.


#

Leukocyte Membrane-Coated NPs for Targeted Drug Delivery

Leukocytes can be divided into granulocytes and agranulocytes which can differentiate into neutrophils, eosinophils, and basophils, or monocytes and lymphocytes, respectively. Same as other cell membrane-coated NPs, leukocyte membranes (LMs) enable NPs to escape from the rapid clearance and increase their circulation time. Due to their high affinity for inflamed areas (e.g., tumor sites peculiarly), they are now regarded as a new carrier for targeted delivery of antitumor drugs.[97] NPs mimicking leukocytes, as well as other types of cell membrane-coated NPs, can mimic the interaction between leukocytes and cancer cells, thereby enhancing tumor therapeutic capacities ([Fig. 4]).[98]

Zoom Image
Fig. 4 Schematic summary of using LM-coated nanoparticles for medical applications. LM NPs are made by cloaking plasma membranes derived from natural lymphocytes onto synthetic cores. LM, leukocyte membrane; NP, nanoparticle. (Reproduced with permission from Wang et al[98], copyright 2022 Wiley-VCH GmbH.)

Interestingly, LM-coated NPs can be used for drug delivery, as well as for isolation and downstream studies of circulating tumor cells (CTCs). These NPs can bind to CTCs because they are homologous to leukocytes and simultaneously repel the living leukocytes to make up a high-density CTC environment. Zhou et al first prepared graphene nanosheets that combined Fe3O4 NPs and disguised them with LMs. By inserting lipid linkers onto the membranes, the antibodies could be conjugated to the LMs, resulting in high capture efficiency and enhanced anti-leukocyte absorption.[98] [99]

Macrophages are specialized APC with long blood half-life and specific binding ability with tumor tissue. In addition, their superior ability to recognize antigens, better cellular interactions, gradual drug release, and reduced toxicity in vivo contributed to their availability in antitumor drug development. This targeted nature makes macrophages and macrophage membranes (MMs) excellent materials for carrying therapeutic drugs for cancer treatment. By co-extruding NPs from the extracted MMs, successfully MM-coated NPs have a thin layer on the surface, are slightly larger in size, have a negatively charged surface zeta potential, and contain membrane proteins.[100] [101] Li et al created an MM-coated nano-gemcitabine system that restores the tumoricidal function of lymphocytes by upregulating PD-L1 expression. The study used MMs with tumor-tropism characteristics to enhance drug accumulation at tumor sites.[102] Tumor-associated macrophages (TAMs), living in the TME, have high levels of colony-stimulating factor 1 receptor on their surfaces, and CSF1 secreted by cancer cells significantly promotes the polarization of TAMs toward M2-macrophages, which are responsible for immunosuppressive characteristic in TME. Inspiringly, Chen et al fabricated TAM membrane-coated NPs that would selectively bind CSF1 as a mock TAM to eliminate primary tumor growth.[101]

In addition to macrophages, the membranes of T cells and NK cells have also been significantly studied. T cell, as an instinctive killer in humans, displays numerous receptors on its membrane aiming at recognizing abnormal cells like cancer cells. Inspired by chimeric antigen receptor (CAR) therapy, a number of immunotherapies based on T cell membranes (TMs) have been exploited. Different antibodies (e.g., anti-EGFR) can be decorated on the TMs while chemicals can be loaded inside the membranes, resulting in CAR-T like nano-robot that can be used to breach biological barriers and improve therapeutic outcomes.[103] Furthermore, Kang et al proposed a technique to combine different molecules including FasL, PD-1, LFA-1, and TGF-β1R to activate strong immunoresponses. T-cell-membrane-coated nanoparticles and anti-cancer drug loaded T-cell-membrane nanoparticles can perform tasks like “real” T cells via FasL and release drugs inside, what's more exciting is that those NPs are free from immunoexhaustion because they are not living cells and they can block the immune checkpoint interactions.[104]


#

Hybrid Cell Membrane-Coated NPs for Targeted Drug Delivery

Hybrid cell membranes (HCMs) are a new type of cloaking membrane for NPs that inherit unique characteristics from two-parent cell lines.[105] In contrast to single-cell membrane (SCM)-coated NPs, which can only be characterized from one type of cell, HCMs have the gifted ability to overcome the limitations of SCMs to enhance the functionality of nanomaterials.[106] The pivotal proteins and properties inherited from both of the parent cells enable multifunctional biomimetic nanomaterials to perform increasingly complex tasks in dynamic biological environments more effectively and safely.[107] When we prepare HCMs, we need one more step than SCMs, which is the fusion of two different membranes. This procedure can be accomplished by two respective methods. One is, of course, to extract the membranes before fusion and the other is to fuse cells before extraction. Among the two membranes used for constructing the drug delivery platform, one should at least offer the targeting ability and the other should offer favorable characteristics for drug administration and release ([Fig. 5]).[108]

Zoom Image
Fig. 5 Hybrid cell membrane-camouflaged nanoparticles (HMC@NPs) designed for cancer diagnosis and treatment. Cell membranes from different types of natural cells were extracted and leveraged to wrap around different nanoparticles for the theranostic of cancer. (Reproduced with permission from Chen et al[6], copyright 2020 Elsevier B.V. All rights reserved.)

Based on this thread, many drug delivery platforms have emerged. Shen et al prepared a self-assemble Nano/ZnO and miR21 antagomir NPs and coated them with the membranes of LnNPs and cancer cells, which enabled NPs to have immune escape and homologous targeting abilities.[109] Novel frameworks, such as tetrahedral framework nucleic acid consisting of four single-stranded DNAs with ingeniously designed sequences, can also be modified with HCMs made from DSPE-PEOz liposomes and EMs, which can speed up drug the release process in acidic environments, as well as enhance circulating time and NP accumulation at tumor sites. In this study, in order to overcome the dilemma of the lack of targeting tendency of erythrocytes, an anti-HER2 aptamer was utilized to direct the NPs to the right place.[110] Besides, delivery systems combining the membranes of erythrocytes and cancer cells, liposomes and cancer cells, etc. have also been explored.[111] [112] Rather than delivering drugs directly to tumor cells, Zang et al shifted the focus to using cancer-associated fibroblasts to cut off the nutritional supply of tumors by delivering solid lipid NPs containing PTX and glycolysis inhibitor PFK15 coated with hybrid membranes. Evidence suggests that the hybrid biomimetic camouflage formed by breast CCMs and activated fibroblast membranes improves antitumor efficiency.[113] However, despite many attempts to explore NP coatings combining different membranes, much remains unknown. Therefore, further efforts in this field are still needed.


#
#

Living Cells for Targeted Drug Delivery

The study of cell-mediated drug delivery methods has been an attractive area in recent years, as these DDSs show great potential for targeted drug delivery. The traditional DDS may be fast and prematurely cleared by the mononuclear phagocytic system and loss of efficacy of PEG-modified NPs under continual administrations because of the stimulation such as the immune system.[114] [115] Compared with traditional DDS, cell-mediated drug delivery has the advantages of hypoimmunogenicity, good stability, biocompatibility, and extended circulating time ([Fig. 6]).[116] [117]

Zoom Image
Fig. 6 Advantages of nanoparticles and circulatory cells in drug delivery. Circulatory cells, including red blood cells, monocytes, platelets, and lymphocytes, have natural drug-delivery abilities. (Reproduced with permission from Anselmo et al[118], copyright 2014 Elsevier B.V. All rights reserved.)

In this case, in terms of different types of circulatory cells, including erythrocytes, leukocytes, platelets, and DCs, living unfettered cells can be used as efficient delivery machines to transport NPs, e.g., those synthetic carriers are bound to circulatory cells by loading the drugs into their internal volume or binding the drugs to their surface via covalent or noncovalent coupling.[118]

Erythrocytes Used for Cell-Mediated Targeted Drug Delivery

Erythrocytes are the most abundant cells in human blood, accounting for 40% of the total.[119] Mature cells are oxygen-carrying cells, disc-shaped, biconcave, and without a nucleus.[120] In the process of antitumor therapy, erythrocytes act as carriers of drugs and transfer the loading drug from the carrier cells (i.e., erythrocytes) to the malignant cells, which is characterized by long circulation time and better biodistribution.[121] Specifically, the “Don't eat me” sign displayed by CD47 and other receptors on their surfaces helps them avoid the degradation of the phagocytosis and assist the NDDS to escape from the clearance of the immune system at the same time.[122]

To establish a living erythrocyte-based DDS, several approaches have been commonly explored, known as erythrocyte hitchhiking (passive adsorption) and attachment. Traditional NDDSs already contain chemotherapeutics or protein pharmaceuticals or small-molecule drugs, and delivery in combination with erythrocytes is one of the methods we are currently exploring to enhance the efficacy of the carriers.

The erythrocyte hitchhiking is proposed to solve the problem that NPs injected into the body are rapidly eliminated by the liver and spleen, thus causing “off-target” effect when drugs are particularly targeted for the lungs and brain.[123] In erythrocyte hitchhiking, changes in shear stress are critical for triggering the release of NPs in microcapillaries. We found that the detachment rate of NP hitchhiking on the cell surfaces increases with elevated stress, implying that this strategy can be used for recognition between abnormal sites and normal sites for more efficient drug delivery.[124]

Erythrocyte attachment, on the other hand, is the most commonly used antitumor strategy. With different techniques, NDDS can be affixed to erythrocytes. Among them, lipid insertion, biotin–avidin bridges, EDC/NHS coupling, and antibody/ligand-receptor conjugation are the four commonly used methods to construct erythrocyte-based DDS ([Fig. 7]).[125]

Zoom Image
Fig. 7 A schematic diagram of (A) lipid insertion, (B) biotin–avidin bridges, (C) EDC/NHS coupling, (D) antibody/ligand-receptor conjugation, and (E) passive adsorption (hitchhiking) methods for refunctionalization of erythrocyte-based nanomedicine. (Reproduced with permission from Sun et al[125], copyright 2019 Ivyspring International Publisher.)

Liu et al reported an erythrocyte-based DDS that unites an oxygen-transporting function (correcting tumor hypoxia environment) with PDT-sensitive NPs through the interaction of avidin and biotin. They stated that owing to the erythrocytes, the DDS could achieve long tumor retention, therefore enhancing therapeutic efficacy.[126]

Efforts have also been put into combining the two approaches, and it is believed that we can fully utilize the advantages of each. Ferguson et al reported an intravascular nanocarrier that combines erythrocyte ligand conjugation and hitchhiking. By applying different kinds of antibodies or adhesion molecules, NPs can achieve erythrocyte hitchhiking at the beginning, and then due to the proximity between the erythrocytes and the NPs, the ligand–receptor bound to the targeted cells (e.g., tumor cells), thus releasing the NPs.[127]


#

Platelets Used for Cell-Mediated Targeted Drug Delivery

Platelets are disc-shaped, anucleate fragments derived from megakaryocytes that play a critical role in maintaining hemostasis and thrombosis in the body, including those in tumor vasculature. Increasing evidence suggests that platelets are also involved in other processes such as immune responses, angiogenesis, and lymphatic vessel development, which may be relevant to tumor occurrence, growth, and metastasis. Platelets generally have a lifespan of 7 to 10 days.[128] [129]

There is an interaction between cancers and platelets. Recent studies have pointed out that tumor cells promote platelet production and aggregation, leading to a number of cardiovascular diseases. Platelets, on the other hand, are responsible for cancer progression and metastatic dissemination by modulating the TME by releasing multiple growth factors and binding to the surfaces of tumor cells to protect CTCs from shear stress and shield them from recognition by immune cells.[130] [131] [132]

Platelets are natural carriers for antitumor drug delivery, and platelet-mediated platforms offer the advantages of readily available, well-tolerated, and relatively low direct production costs in comparison to traditional DDS.[133] These carriers can be hypoimmunogenic as they can be harvested from the patients themselves.[134] When an in situ tumor is seeking metastasis, selectins and integrins' interactions between the two substances are significant during platelet contact with tumor cells. This character makes platelet hitchhiking NPs possible.[135] On this basis, N3-mediated click chemistry can be utilized to fasten Granzyme B-loaded NPs on the surface of platelets, and by hitchhiking platelets, the system can respond to acidic TME and release NPs to attack tumor cells.[136] The platform is highly suitable for postsurgical cancer treatment. Platelets here are called “bridges” which fill the gap between NDDS and tumor cells.[137] At the same time, the use of platelets as a basic component of immunotherapy is also a hot area of research. Anti-PD-1 antibodies (aPD-1) are conjugated on the surfaces of platelets. When platelets are activated after arriving at tumor sites, aPD-1 is released in the form of platelet-derived microparticles that can subsequently bind to T cells to enhance the efficacy of immunotherapy.[138] [139]


#

Leukocytes Used for Cell-Mediated Targeted Drug Delivery

Leukocytes are important participants in the body's innate and adaptive immunity, which are involved in the protection of the body from infectious diseases and the removal of cell debris and foreign antigens.[140] [141] Leukocytes can be divided into five categories according to their physiological functions and characteristics: neutrophils (amount in blood: 50–70%), lymphocytes (25–35%), monocytes (2–8%), eosinophils (1–3%), and basophils (0.4–1%). Leukocytes provide an excellent opportunity for drug delivery due to their long-circulating time, tumor penetration tendencies, and the capability to cross biological barriers. While traditional NDDS is hindered by a variety of biological barriers, such as blood vessels, agents such as leukocytes hijacked by NPs can take full advantage of the targeting motility and transmigration ability of leukocytes to deliver NPs to inflammatory such as tumors.[142] [143] Similar to erythrocyte carriers, leukocyte-based platforms can be constructed by NPs, free drug hitchhiking, or internalization.[144] In this review, we focus on neutrophil- and lymphocyte-based DDSs.

Neutrophils are the largest group of leukocytes. Administration via activated neutrophils is a promising approach to antitumor drug delivery, as neutrophils have natural chemotaxis to inflammatory signals from progressing tumors. Using their sophisticated cellular machinery, neutrophils can penetrate the tumor-associated endothelium and infiltrate the TME.[145] There are two methods to construct DDS, including incubating neutrophils with drugs before administration or hijacking neutrophils in the bloodstream. For the former method, neutrophils act perfectly as Trojan Horses, releasing the therapeutic cargoes at the tumor site by releasing neutrophil extracellular traps (NETs).[146] Inflammation is one of the main causes of NET release. Thus, Ren et al designed a neutrophil-mediated liposome co-loaded with PTX and hydroxychloroquine to form NETs that “open” the cells and release the liposomes when activated by cytokines secreted by the tumor-related cells.[147] By inducing tumor cell death and inhibiting their autophagy at the same time, a better antitumor effect can be achieved. This technology can also be applied to deliver drugs to different organs such as bone marrow.[148] For the latter, antibody conjugation can be used to capture and internalize NPs via activated neutrophils after intravenous injection.[149] However, this method might encounter problems with the rapid clearance of injected NPs by the immune system and therefore requires further investigation.

T cells interfaced with antitumor agents have also been explored in recent years, and the distinctive features of cytotoxic T-lymphocytes (CTLs) have been exploited to design a T cell-based platform for cancer therapy. When CTLs interact with tumor cells, they release perforin and granzymes into the immunological synapse formed between the CTLs and target cells. The nanocapsules attached to the surfaces of the CTLs entered the synapse and met the perforin secreted by CTLs, leading to the disruption of the nanocapsule and release of encapsulated drugs in tandem with the lysis of the tumor cells.[150] In addition, innovative carriers, as shown in [Fig. 8], a TA-encapsulated polymeric micelle decorating with aPD-1 on the surface, which can bind circulating PD-1+ T cells to overcome immune checkpoint blockade resistance, are still under careful research.[151]

Zoom Image
Fig. 8 Schematic illustration for the antitumor mechanism of nanodrug in overcoming immune checkpoint blockade resistance in HCC. HCC, hepatocellular carcinoma. (Reproduced with permission from Wang et al[151], copyright 2023 BMJ Publishing Group Ltd & Society for Immunotherapy of Cancer.)

In summary, the use of living cells for the targeted delivery of antitumor agents or diagnostic reagents is an emerging approach that has made significant progress to date, but the challenges in this field cannot be ignored. First, the most common strategy is to separate carrier cells and load drugs into these cells before administration, which may affect the original characteristics of cells, and the drugs may undergo complex intracellular processes in living cells. In addition, drug-loading efficiency is relatively low due to the limited drug-cell binding capacity. Also, the leaking of drugs into the body from the exogenous cells is an urgent issue. Once these issues are addressed, this therapy could have a revolutionary impact on the process of eliminating tumors.


#
#

Summary

BDDSs mimic the structures and functions of various natural substances. BDDS, like well-trained and well-equipped plainclothes policemen, can achieve better targeting, improve the efficiency and accuracy of drug delivery, and make up for the shortcomings of existing NDDS. BDDS is able to better address the current problems faced by traditional chemotherapeutics and DDS, and thereby an ideal candidate for cancer treatment. In this article, the performances and advances of three BDDS, i.e. EVs, living cell-based drug carriers, and membrane-coated NPs, have been reviewed in antitumor therapies, and are summarized in [Table 1]. The emergence of BDDS breaks through the limitations of traditional nanomedicine, with higher targeting ability, longer circulating time, excellent capacity to cross biological barriers, and higher drug accumulation at the tumor sites, and brings more opportunities for targeted tumor therapies. In the future, biomimetic systems can produce unified and standardized formulations for most patients on the one hand, and can also design more adaptable drugs for specific patients, achieving personalized precision medicine on the other.

Table 1

Brief introduction of the current BDDS

BDDS type

Source cells

Advantages

Disadvantages

Tumor cell model

Pharmacological factors

Ref.

EVs

MSC

Smaller size, prolonged circulating time, industrial application, malignancy-targeting ability

Building tumor microenvironment,

immunosuppressive

behavior

CFPAC-1

Paclitaxel

[40]

PANC-1, MIA-PaCa-2, CFPAC-1

circRNA circ_0030167

[39]

B16/F10

PI3Kγ inhibitor

[154]

CT26

Photodynamic therapy

agent mTHPC

[41]

DCs

Boost the efficacy of immuno-

therapy

High cost, time-consuming

B16-OVA

Anti-CD3 antibody

membrane insertion

[53]

B16-MUC1

Tumor antigen

MUC1 glycopeptide

membrane conjugation

[54]

B16F10

CTX

[155]

B16F10

Tumor antigen-loaded inside

[58]

Tumor cell

Homing, immune cell sensitization, and high absorption

Induce tumor immune tolerance, promoting neoplastic angiogenesis

MDA-MB-231/luc

TLR3 agonist and ELANE

[156]

4T1

FeS2 nanozyme

[157]

HT1080, HeLa

Doxorubicin

[69]

CMCNs

Erythrocyte

Sufficient raw materials, immune escape

Induce alloimmunization, complex extraction process

B16F10

Worm-like siRNA

[86]

HepG2

Mesoporous silicon nanoparticle

[158]

Saos-2

DOX-loaded ZC NPs

[159]

Tumor cell

Homotypic targeting and immune escape abilities

Difficult extraction

SMMC-7721

Lenvatinib@PAE NPs

[160]

HeLa, MCF7, etc.

AuNPs

[161]

Leukocyte

Increased circulation time, high affinity toward tumors

Lack of economic manufacturing processes and high-standards of quality assurance

Kasumi-1, U937, MV4–11, etc.

Glycyrrhetinic acid/PLGA NPs

[162]

KYSE-150

DOX-loaded

lipid nanovector

[163]

Hybrid cell membrane

Multi-features from parent cells

Cell membrane damage caused by hybridization

SKBR3, BT474

Maytansine-loaded tetrahedral framework nucleic acid

[110]

Living cell drug delivery system

Erythrocyte

Long circulation time and immune escape

Lack of targeting molecules

Upconversion NPs, hitchhiking

[125]

Platelet

Low costs, hypoimmunogenic

May contribute to cancer progression and metastatic dissemination

C1498

Anti-PD-L1 antibody surface conjugation

[137] [138]

B16F10, CT26, 4T1

Leukocyte

Targeting movement and transmigration ability toward tumor cells

Interfered by the immunosuppressive factors secreted by tumor cells

4T1

Paclitaxel and hydroxychloroquine-loaded liposomes

[146]

Abbreviation: BDDS, biomimetic drug delivery system; CMCNs, cell-membrane-coated NPs; CTX, cyclophosphamide; DCs, dendritic cells; DOX, doxorubicin; EVs, extracellular vesicles; MSC, mesenchymal stem cell.


One of the most challenging problems faced by traditional small-molecule chemotherapy drugs and conventional DDS is the adverse effects caused by the “off-target” effect, relatively high dose, and high immunogenicity. BDDS offers a possible solution to these problems. BDDS carriers not only have a variety of natural targeting molecules on their surfaces that can precisely direct the drugs to the tumor sites but also have “self” characteristics that allow them to avoid rapid clearance by the immune system. In this case, the adverse effects can be minimized by applying smaller doses while achieving the same therapeutic effect as before or even better.

BDDSs are all obtained from bio-materials and carry a large number of biological macromolecules, so one of the key issues of BDDS is their immunogenicity. Overcoming the inherent immunogenicity of BDDS will be an urgent problem to be solved in large-scale industrial production in the future. We believe that by modifying cell biofilm, controlling the composition of biomimetic membranes, ensuring consistency of source, avoiding heterologous issues, etc., more BDDSs will enter clinical trials and even be on the market.

The advanced DDS still has many other challenges and difficulties in ensuring carrier separation and purification effect, unclear mechanism of functioning in vivo and clinical transformation. Questions such as “Will there be interactions between natural components of the human body and artificially modified components that are not conducive to drug delivery?,” “Can artificially processed natural materials still efficiently maintain the functions of their parent tissues or cells?,” or “Can current technology support the mass production and clinical application of BDDS?” still need to be answered. In addition, the safety of various materials currently used to construct BDDS has not been thoroughly and meticulously evaluated and has rarely been approved by the U.S. Food and Drug Administration, which to some extent limits the subsequent clinical conversion of such drugs.

To solve these problems, several new strategies have been proposed. Recently, EVs isolated from natural grapefruit and ginseng, instead of from mammals, were reported in antitumor therapy in combination chemotherapeutics. Those plant-derived EVs are low immunologically, green, renewable, and mass-producible, and are expected to ameliorate the shortcomings of EVs and bring new hopes for patients.[152] [153] Many researchers have been trying to combine traditional DDS with biomaterials to achieve better therapeutic effects by using hybridizing liposomes, cell membranes, and other materials. It seems that strategies applied to traditional DDS, such as modification of the NP surfaces using PLGA and targeted molecules, appear to be equally feasible with BDDS and could improve antitumor efficacy.

Despite the issues encountered with BDDS, as mentioned earlier, the advantages of these systems are also evident. EVs seem to be the most promising carrier for tumor diagnoses and treatment. To date, several EV-based drugs are in clinical trials. The large number of EVs currently points to new directions in the treatments of Crohn's disease, chronic obstructive pulmonary disease, empyrosis, etc. The smaller size and “lifeless” characteristics give them a better ability to evade immune clearance and stability compared to living cell-based nano-carriers. While the proteins and nucleic acids contained within them may be sources of immunogenicity and a driver of tumor metastasis (especially EVs derived from tumor cells), some of the endogenous substances in EVs have intrinsic antitumor activity, which to some extent gives them an advantage over cell membrane-coated NPs.

It is believed that with further research in the fields of materials science and molecular biology, BDDS will be more widely used and play an important role in weaponizing common substances in the body to fight tumors.


#
#

Conflict of Interest

None declared.

  • References

  • 1 Sung H, Ferlay J, Siegel RL. et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 71 (03) 209-249
  • 2 Pusuluri A, Wu D, Mitragotri S. Immunological consequences of chemotherapy: single drugs, combination therapies and nanoparticle-based treatments. J Control Release 2019; 305: 130-154
  • 3 Ma Q, Cao J, Gao Y. et al. Microfluidic-mediated nano-drug delivery systems: from fundamentals to fabrication for advanced therapeutic applications. Nanoscale 2020; 12 (29) 15512-15527
  • 4 Ho YJ, Chiang YJ, Kang ST, Fan CH, Yeh CK. Camptothecin-loaded fusogenic nanodroplets as ultrasound theranostic agent in stem cell-mediated drug-delivery system. J Control Release 2018; 278: 100-109
  • 5 Wu HH, Zhou Y, Tabata Y, Gao JQ. Mesenchymal stem cell-based drug delivery strategy: from cells to biomimetic. J Control Release 2019; 294: 102-113
  • 6 Chen HY, Deng J, Wang Y, Wu CQ, Li X, Dai HW. Hybrid cell membrane-coated nanoparticles: a multifunctional biomimetic platform for cancer diagnosis and therapy. Acta Biomater 2020; 112: 1-13
  • 7 Chi J, Ma Q, Shen Z. et al. Targeted nanocarriers based on iodinated-cyanine dyes as immunomodulators for synergistic phototherapy. Nanoscale 2020; 12 (20) 11008-11025
  • 8 Guan YH, Wang N, Deng ZW, Chen XG, Liu Y. Exploiting autophagy-regulative nanomaterials for activation of dendritic cells enables reinforced cancer immunotherapy. Biomaterials 2022; 282: 121434
  • 9 Bush LM, Healy CP, Javdan SB, Emmons JC, Deans TL. Biological cells as therapeutic delivery vehicles. Trends Pharmacol Sci 2021; 42 (02) 106-118
  • 10 Elsharkasy OM, Nordin JZ, Hagey DW. et al. Extracellular vesicles as drug delivery systems: why and how?. Adv Drug Deliv Rev 2020; 159: 332-343
  • 11 Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol 2021; 16 (07) 748-759
  • 12 Liu T, Gao C, Gu D, Tang H. Cell-based carrier for targeted hitchhiking delivery. Drug Deliv Transl Res 2022; 12 (11) 2634-2648
  • 13 Zhang X, Li N, Zhang S. et al. Emerging carrier-free nanosystems based on molecular self-assembly of pure drugs for cancer therapy. Med Res Rev 2020; 40 (05) 1754-1775
  • 14 Ayer M, Klok HA. Cell-mediated delivery of synthetic nano- and microparticles. J Control Release 2017; 259: 92-104
  • 15 Fang RH, Gao W, Zhang L. Targeting drugs to tumours using cell membrane-coated nanoparticles. Nat Rev Clin Oncol 2023; 20 (01) 33-48
  • 16 Sun S, Yang Y, Gao Z. et al. endogenous stimuli-responsive autonomous separation of dual-targeting DNA guided missile from nanospacecraft for intelligent targeted cancer therapy. ACS Appl Mater Interfaces 2022; 14 (40) 45201-45216
  • 17 Théry C, Witwer KW, Aikawa E. et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 2018; 7 (01) 1535750
  • 18 Zhang H, Lyden D. Asymmetric-flow field-flow fractionation technology for exomere and small extracellular vesicle separation and characterization. Nat Protoc 2019; 14 (04) 1027-1053
  • 19 van der Meel R, Fens MH, Vader P, van Solinge WW, Eniola-Adefeso O, Schiffelers RM. Extracellular vesicles as drug delivery systems: lessons from the liposome field. J Control Release 2014; 195: 72-85
  • 20 Piffoux M, Silva AKA, Wilhelm C, Gazeau F, Tareste D. Modification of extracellular vesicles by fusion with liposomes for the design of personalized biogenic drug delivery systems. ACS Nano 2018; 12 (07) 6830-6842
  • 21 Schulz-Siegmund M, Aigner A. Nucleic acid delivery with extracellular vesicles. Adv Drug Deliv Rev 2021; 173: 89-111
  • 22 Ou YH, Liang J, Czarny B. et al. Extracellular vesicle (EV) biohybrid systems for cancer therapy: recent advances and future perspectives. Semin Cancer Biol 2021; 74: 45-61
  • 23 Richter M, Vader P, Fuhrmann G. Approaches to surface engineering of extracellular vesicles. Adv Drug Deliv Rev 2021; 173: 416-426
  • 24 Sharma S, Masud MK, Kaneti YV. et al. Extracellular vesicle nanoarchitectonics for novel drug delivery applications. Small 2021; 17 (42) e2102220
  • 25 Roerig J, Schulz-Siegmund M. Standardization approaches for extracellular vesicle loading with oligonucleotides and biologics. Small 2023; 19 (40) e2301763
  • 26 Liu YR, Cheng YQ, Wang SB. et al. Therapeutic effects and perspective of stem cell extracellular vesicles in aging and cancer. J Cell Physiol 2021; 236 (07) 4783-4796
  • 27 Lan T, Luo M, Wei X. Mesenchymal stem/stromal cells in cancer therapy. J Hematol Oncol 2021; 14 (01) 195
  • 28 Keshtkar S, Azarpira N, Ghahremani MH. Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Res Ther 2018; 9 (01) 63
  • 29 Hass R, Kasper C, Böhm S, Jacobs R. Different populations and sources of human mesenchymal stem cells (MSC): a comparison of adult and neonatal tissue-derived MSC. Cell Commun Signal 2011; 9: 12
  • 30 Rani S, Ryan AE, Griffin MD, Ritter T. Mesenchymal stem cell-derived extracellular vesicles: toward cell-free therapeutic applications. Mol Ther 2015; 23 (05) 812-823
  • 31 Xu F, Fei Z, Dai H. et al. Mesenchymal stem cell-derived extracellular vesicles with high PD-L1 expression for autoimmune diseases treatment. Adv Mater 2022; 34 (01) e2106265
  • 32 Lai RC, Yeo RW, Lim SK. Mesenchymal stem cell exosomes. Semin Cell Dev Biol 2015; 40: 82-88
  • 33 Su C, Zhang J, Yarden Y, Fu L. The key roles of cancer stem cell-derived extracellular vesicles. Signal Transduct Target Ther 2021; 6 (01) 109
  • 34 Sohrabi B, Dayeri B, Zahedi E. et al. Mesenchymal stem cell (MSC)-derived exosomes as novel vehicles for delivery of miRNAs in cancer therapy. Cancer Gene Ther 2022; 29 (8–9): 1105-1116
  • 35 Sun Y, Liu G, Zhang K, Cao Q, Liu T, Li J. Mesenchymal stem cells-derived exosomes for drug delivery. Stem Cell Res Ther 2021; 12 (01) 561
  • 36 Crivelli B, Chlapanidas T, Perteghella S. et al; Italian Mesenchymal Stem Cell Group (GISM). Mesenchymal stem/stromal cell extracellular vesicles: from active principle to next generation drug delivery system. J Control Release 2017; 262: 104-117
  • 37 Xunian Z, Kalluri R. Biology and therapeutic potential of mesenchymal stem cell-derived exosomes. Cancer Sci 2020; 111 (09) 3100-3110
  • 38 Jahangiri B, Khalaj-Kondori M, Asadollahi E, Purrafee Dizaj L, Sadeghizadeh M. MSC-Derived exosomes suppress colorectal cancer cell proliferation and metastasis via miR-100/mTOR/miR-143 pathway. Int J Pharm 2022; 627: 122214
  • 39 Yao X, Mao Y, Wu D. et al. Exosomal circ_0030167 derived from BM-MSCs inhibits the invasion, migration, proliferation and stemness of pancreatic cancer cells by sponging miR-338-5p and targeting the Wif1/Wnt8/β-catenin axis. Cancer Lett 2021; 512: 38-50
  • 40 Pascucci L, Coccè V, Bonomi A. et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: a new approach for drug delivery. J Control Release 2014; 192: 262-270
  • 41 Pinto A, Marangon I, Méreaux J. et al. Immune reprogramming precision photodynamic therapy of peritoneal metastasis by scalable stem-cell-derived extracellular vesicles. ACS Nano 2021; 15 (02) 3251-3263
  • 42 Luo T, Liu Q, Tan A. et al. Mesenchymal stem cell-secreted exosome promotes chemoresistance in breast cancer via enhancing miR-21-5p-mediated S100A6 expression. Mol Ther Oncolytics 2020; 19: 283-293
  • 43 Zhu S, Yang N, Wu J. et al. Tumor microenvironment-related dendritic cell deficiency: a target to enhance tumor immunotherapy. Pharmacol Res 2020; 159: 104980
  • 44 Clark GJ, Silveira PA, Hogarth PM, Hart DNJ. The cell surface phenotype of human dendritic cells. Semin Cell Dev Biol 2019; 86: 3-14
  • 45 Bol KF, Schreibelt G, Gerritsen WR, de Vries IJ, Figdor CG. Dendritic cell-based immunotherapy: state of the art and beyond. Clin Cancer Res 2016; 22 (08) 1897-1906
  • 46 Macri C, Pang ES, Patton T, O'Keeffe M. Dendritic cell subsets. Semin Cell Dev Biol 2018; 84: 11-21
  • 47 Worbs T, Hammerschmidt SI, Förster R. Dendritic cell migration in health and disease. Nat Rev Immunol 2017; 17 (01) 30-48
  • 48 Li J, Li J, Peng Y, Du Y, Yang Z, Qi X. Dendritic cell derived exosomes loaded neoantigens for personalized cancer immunotherapies. J Control Release 2023; 353: 423-433
  • 49 Näslund TI, Gehrmann U, Qazi KR, Karlsson MC, Gabrielsson S. Dendritic cell-derived exosomes need to activate both T and B cells to induce antitumor immunity. J Immunol 2013; 190 (06) 2712-2719
  • 50 Xiong X, Ke X, Wang L. et al. Neoantigen-based cancer vaccination using chimeric RNA-loaded dendritic cell-derived extracellular vesicles. J Extracell Vesicles 2022; 11 (08) e12243
  • 51 Esser J, Gehrmann U, D'Alexandri FL. et al. Exosomes from human macrophages and dendritic cells contain enzymes for leukotriene biosynthesis and promote granulocyte migration. J Allergy Clin Immunol 2010; 126 (05) 1032-1040 , 1040.e1–1040.e4
  • 52 Pitt JM, Charrier M, Viaud S. et al. Dendritic cell-derived exosomes as immunotherapies in the fight against cancer. J Immunol 2014; 193 (03) 1006-1011
  • 53 Fan M, Liu H, Yan H. et al. A CAR T-inspiring platform based on antibody-engineered exosomes from antigen-feeding dendritic cells for precise solid tumor therapy. Biomaterials 2022; 282: 121424
  • 54 Zhu H, Wang K, Wang Z. et al. An efficient and safe MUC1-dendritic cell-derived exosome conjugate vaccine elicits potent cellular and humoral immunity and tumor inhibition in vivo . Acta Biomater 2022; 138: 491-504
  • 55 Lu Z, Zuo B, Jing R. et al. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol 2017; 67 (04) 739-748
  • 56 Viaud S, Terme M, Flament C. et al. Dendritic cell-derived exosomes promote natural killer cell activation and proliferation: a role for NKG2D ligands and IL-15Ralpha. PLoS One 2009; 4 (03) e4942
  • 57 Zitvogel L, Regnault A, Lozier A. et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med 1998; 4 (05) 594-600
  • 58 Damo M, Wilson DS, Simeoni E, Hubbell JA. TLR-3 stimulation improves anti-tumor immunity elicited by dendritic cell exosome-based vaccines in a murine model of melanoma. Sci Rep 2015; 5: 17622
  • 59 Tian X, Shen H, Li Z, Wang T, Wang S. Tumor-derived exosomes, myeloid-derived suppressor cells, and tumor microenvironment. J Hematol Oncol 2019; 12 (01) 84
  • 60 Jiang C, Zhang N, Hu X, Wang H. Tumor-associated exosomes promote lung cancer metastasis through multiple mechanisms. Mol Cancer 2021; 20 (01) 117
  • 61 Naseri M, Bozorgmehr M, Zöller M, Ranaei Pirmardan E, Madjd Z. Tumor-derived exosomes: the next generation of promising cell-free vaccines in cancer immunotherapy. OncoImmunology 2020; 9 (01) 1779991
  • 62 Mashouri L, Yousefi H, Aref AR, Ahadi AM, Molaei F, Alahari SK. Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol Cancer 2019; 18 (01) 75
  • 63 Gao Y, Xu H, Li N. et al. Renal cancer-derived exosomes induce tumor immune tolerance by MDSCs-mediated antigen-specific immunosuppression. Cell Commun Signal 2020; 18 (01) 106
  • 64 Ma Z, Wei K, Yang F. et al. Tumor-derived exosomal miR-3157-3p promotes angiogenesis, vascular permeability and metastasis by targeting TIMP/KLF2 in non-small cell lung cancer. Cell Death Dis 2021; 12 (09) 840
  • 65 Guo X, Sui R, Piao H. Tumor-derived small extracellular vesicles: potential roles and mechanism in glioma. J Nanobiotechnology 2022; 20 (01) 383
  • 66 Hoshino A, Costa-Silva B, Shen TL. et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015; 527 (7578) 329-335
  • 67 Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science 2020; 367 (6478) eaau6977
  • 68 Thakur A, Parra DC, Motallebnejad P, Brocchi M, Chen HJ. Exosomes: small vesicles with big roles in cancer, vaccine development, and therapeutics. Bioact Mater 2021; 10: 281-294
  • 69 Qiao L, Hu S, Huang K. et al. Tumor cell-derived exosomes home to their cells of origin and can be used as Trojan horses to deliver cancer drugs. Theranostics 2020; 10 (08) 3474-3487
  • 70 Andre F, Schartz NE, Movassagh M. et al. Malignant effusions and immunogenic tumour-derived exosomes. Lancet 2002; 360 (9329) 295-305
  • 71 Taghikhani A, Hassan ZM, Ebrahimi M, Moazzeni SM. microRNA modified tumor-derived exosomes as novel tools for maturation of dendritic cells. J Cell Physiol 2019; 234 (06) 9417-9427
  • 72 Gong H, Zhang Q, Komarla A. et al. Nanomaterial biointerfacing via mitochondrial membrane coating for targeted detoxification and molecular detection. Nano Lett 2021; 21 (06) 2603-2609
  • 73 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 (08) 3233-3254
  • 74 Zhen X, Cheng P, Pu K. Recent advances in cell membrane-camouflaged nanoparticles for cancer phototherapy. Small 2019; 15 (01) e1804105
  • 75 Dash P, Piras AM, Dash M. Cell membrane coated nanocarriers - an efficient biomimetic platform for targeted therapy. J Control Release 2020; 327: 546-570
  • 76 Liu L, Pan D, Chen S. et al. Systematic design of cell membrane coating to improve tumor targeting of nanoparticles. Nat Commun 2022; 13 (01) 6181
  • 77 Luk BT, Zhang L. Cell membrane-camouflaged nanoparticles for drug delivery. J Control Release 2015; 220 (Pt B): 600-607
  • 78 Wang Y, Chen X, He D, Zhou Y, Qin L. Surface-modified nanoerythrocyte loading DOX for targeted liver cancer chemotherapy. Mol Pharm 2018; 15 (12) 5728-5740
  • 79 Fu S, Liang M, Wang Y. et al. Dual-modified novel biomimetic nanocarriers improve targeting and therapeutic efficacy in glioma. ACS Appl Mater Interfaces 2019; 11 (02) 1841-1854
  • 80 Tao C, Nie X, Zhu W, Iqbal J, Xu C, Wang DA. Autologous cell membrane coatings on tissue engineering xenografts for suppression and alleviation of acute host immune responses. Biomaterials 2020; 258: 120310
  • 81 Han X, Wang C, Liu Z. Red blood cells as smart delivery systems. Bioconjug Chem 2018; 29 (04) 852-860
  • 82 Peng S, Ouyang B, Men Y. et al. Biodegradable zwitterionic polymer membrane coating endowing nanoparticles with ultra-long circulation and enhanced tumor photothermal therapy. Biomaterials 2020; 231: 119680
  • 83 Ye S, Wang F, Fan Z. et al. Light/pH-triggered biomimetic red blood cell membranes camouflaged small molecular drug assemblies for imaging-guided combinational chemo-photothermal therapy. ACS Appl Mater Interfaces 2019; 11 (17) 15262-15275
  • 84 Zhang Y, Xia Q, Wu T. et al. A novel multi-functionalized multicellular nanodelivery system for non-small cell lung cancer photochemotherapy. J Nanobiotechnology 2021; 19 (01) 245
  • 85 Xie H, Li W, Liu H. et al. Erythrocyte membrane-coated invisible acoustic-sensitive nanoparticle for inducing tumor thrombotic infarction by precisely damaging tumor vascular endothelium. Small 2022; 18 (30) e2201933
  • 86 Wang Y, Ji X, Ruan M. et al. Worm-like biomimetic nanoerythrocyte carrying siRNA for melanoma gene therapy. Small 2018; 14 (47) e1803002
  • 87 Zhang Z, Qian H, Huang J. et al. Anti-EGFR-iRGD recombinant protein modified biomimetic nanoparticles loaded with gambogic acid to enhance targeting and antitumor ability in colorectal cancer treatment. Int J Nanomedicine 2018; 13: 4961-4975
  • 88 Miao Y, Yang Y, Guo L. et al. Cell membrane-camouflaged nanocarriers with biomimetic deformability of erythrocytes for ultralong circulation and enhanced cancer therapy. ACS Nano 2022; 16 (04) 6527-6540
  • 89 Guo H, Zhang W, Wang L, Shao Z, Huang X. Biomimetic cell membrane-coated glucose/oxygen-exhausting nanoreactor for remodeling tumor microenvironment in targeted hypoxic tumor therapy. Biomaterials 2022; 290: 121821
  • 90 Pan WL, Tan Y, Meng W. et al. Microenvironment-driven sequential ferroptosis, photodynamic therapy, and chemotherapy for targeted breast cancer therapy by a cancer-cell-membrane-coated nanoscale metal-organic framework. Biomaterials 2022; 283: 121449
  • 91 Fang Z, Zhang M, Kang R, Cui M, Song M, Liu K. A cancer cell membrane coated nanoparticles-based gene delivery system for enhancing cancer therapy. Int J Pharm 2022; 629: 122415
  • 92 Gan J, Du G, He C. et al. Tumor cell membrane enveloped aluminum phosphate nanoparticles for enhanced cancer vaccination. J Control Release 2020; 326: 297-309
  • 93 Jin J, Krishnamachary B, Barnett JD. et al. Human cancer cell membrane-coated biomimetic nanoparticles reduce fibroblast-mediated invasion and metastasis and induce T-cells. ACS Appl Mater Interfaces 2019; 11 (08) 7850-7861
  • 94 Wang Z, Zhang M, Chi S, Zhu M, Wang C, Liu Z. Brain tumor cell membrane-coated lanthanide-doped nanoparticles for NIR-IIb luminescence imaging and surgical navigation of glioma. Adv Healthc Mater 2022; 11 (16) e2200521
  • 95 Chen M, Cui Y, Hao W. et al. Ligand-modified homologous targeted cancer cell membrane biomimetic nanostructured lipid carriers for glioma therapy. Drug Deliv 2021; 28 (01) 2241-2255
  • 96 Zheng B, Liu Z, Wang H. et al. R11 modified tumor cell membrane nanovesicle-camouflaged nanoparticles with enhanced targeting and mucus-penetrating efficiency for intravesical chemotherapy for bladder cancer. J Control Release 2022; 351: 834-846
  • 97 Mohale S, Kunde SS, Wairkar S. Biomimetic fabrication of nanotherapeutics by leukocyte membrane cloaking for targeted therapy. Colloids Surf B Biointerfaces 2022; 219: 112803
  • 98 Wang D, Wang S, Zhou Z. et al. White blood cell membrane-coated nanoparticles: recent development and medical applications. Adv Healthc Mater 2022; 11 (07) e2101349
  • 99 Zhou X, Luo B, Kang K. et al. Leukocyte-repelling biomimetic immunomagnetic nanoplatform for high-performance circulating tumor cells isolation. Small 2019; 15 (17) e1900558
  • 100 Xia Y, Rao L, Yao H, Wang Z, Ning P, Chen X. Engineering macrophages for cancer immunotherapy and drug delivery. Adv Mater 2020; 32 (40) e2002054
  • 101 Chen C, Song M, Du Y. et al. Tumor-associated-macrophage-membrane-coated nanoparticles for improved photodynamic immunotherapy. Nano Lett 2021; 21 (13) 5522-5531
  • 102 Li J, Wu Y, Wang J. et al. Macrophage membrane-coated nano-gemcitabine promotes lymphocyte infiltration and synergizes antiPD-L1 to restore the tumoricidal function. ACS Nano 2023; 17 (01) 322-336
  • 103 Wang W, Wu F, Mohammadniaei M. et al. Genetically edited T-cell membrane coated AIEgen nanoparticles effectively prevents glioblastoma recurrence. Biomaterials 2023; 293: 121981
  • 104 Kang M, Hong J, Jung M. et al. T-Cell-mimicking nanoparticles for cancer immunotherapy. Adv Mater 2020; 32 (39) e2003368
  • 105 Hao W, Cui Y, Fan Y. et al. Hybrid membrane-coated nanosuspensions for multi-modal anti-glioma therapy via drug and antigen delivery. J Nanobiotechnology 2021; 19 (01) 378
  • 106 Wu L, Li Q, Deng J. et al. Platelet-tumor cell hybrid membrane-camouflaged nanoparticles for enhancing therapy efficacy in glioma. Int J Nanomedicine 2021; 16: 8433-8446
  • 107 Zhao Y, Li A, Jiang L, Gu Y, Liu J. Hybrid membrane-coated biomimetic nanoparticles (HM@BNPs): a multifunctional nanomaterial for biomedical applications. Biomacromolecules 2021; 22 (08) 3149-3167
  • 108 Liao Y, Zhang Y, Blum NT, Lin J, Huang P. Biomimetic hybrid membrane-based nanoplatforms: synthesis, properties and biomedical applications. Nanoscale Horiz 2020; 5 (09) 1293-1302
  • 109 Shen T, Yang S, Qu X. et al. A bionic “Trojan horse”-like gene delivery system hybridized with tumor and macrophage cell membrane for cancer therapy. J Control Release 2023; 358: 204-218
  • 110 Ma W, Yang Y, Zhu J. et al. Biomimetic nanoerythrosome-coated aptamer-DNA tetrahedron/maytansine conjugates: pH-responsive and targeted cytotoxicity for HER2-positive breast cancer. Adv Mater 2022; 34 (46) e2109609
  • 111 Zhang W, Gong C, Chen Z, Li M, Li Y, Gao J. Tumor microenvironment-activated cancer cell membrane-liposome hybrid nanoparticle-mediated synergistic metabolic therapy and chemotherapy for non-small cell lung cancer. J Nanobiotechnology 2021; 19 (01) 339
  • 112 Chen H, Deng J, Yao X. et al. Bone-targeted erythrocyte-cancer hybrid membrane-camouflaged nanoparticles for enhancing photothermal and hypoxia-activated chemotherapy of bone invasion by OSCC. J Nanobiotechnology 2021; 19 (01) 342
  • 113 Zang S, Huang K, Li J. et al. Metabolic reprogramming by dual-targeting biomimetic nanoparticles for enhanced tumor chemo-immunotherapy. Acta Biomater 2022; 148: 181-193
  • 114 Nikfar M, Razizadeh M, Paul R, Muzykantov V, Liu Y. A numerical study on drug delivery via multiscale synergy of cellular hitchhiking onto red blood cells. Nanoscale 2021; 13 (41) 17359-17372
  • 115 Anselmo AC, Gupta V, Zern BJ. et al. Delivering nanoparticles to lungs while avoiding liver and spleen through adsorption on red blood cells. ACS Nano 2013; 7 (12) 11129-11137
  • 116 Su Y, Xie Z, Kim GB, Dong C, Yang J. Design strategies and applications of circulating cell-mediated drug delivery systems. ACS Biomater Sci Eng 2015; 1 (04) 201-217
  • 117 Yang L, Yang Y, Chen Y, Xu Y, Peng J. Cell-based drug delivery systems and their in vivo fate. Adv Drug Deliv Rev 2022; 187: 114394
  • 118 Anselmo AC, Mitragotri S. Cell-mediated delivery of nanoparticles: taking advantage of circulatory cells to target nanoparticles. J Control Release 2014; 190: 531-541
  • 119 Villa CH, Cines DB, Siegel DL, Muzykantov V. Erythrocytes as carriers for drug delivery in blood transfusion and beyond. Transfus Med Rev 2017; 31 (01) 26-35
  • 120 Mukthavaram R, Shi G, Kesari S, Simberg D. Targeting and depletion of circulating leukocytes and cancer cells by lipophilic antibody-modified erythrocytes. J Control Release 2014; 183: 146-153
  • 121 Li Y, Raza F, Liu Y. et al. Clinical progress and advanced research of red blood cells based drug delivery system. Biomaterials 2021; 279: 121202
  • 122 Parodi A, Molinaro R, Sushnitha M. et al. Bio-inspired engineering of cell- and virus-like nanoparticles for drug delivery. Biomaterials 2017; 147: 155-168
  • 123 Brenner JS, Pan DC, Myerson JW. et al. Red blood cell-hitchhiking boosts delivery of nanocarriers to chosen organs by orders of magnitude. Nat Commun 2018; 9 (01) 2684
  • 124 Wang S, Ma S, Li R. et al. Probing the interaction between supercarrier RBC membrane and nanoparticles for optimal drug delivery. J Mol Biol 2023; 435 (01) 167539
  • 125 Sun D, Chen J, Wang Y. et al. Advances in refunctionalization of erythrocyte-based nanomedicine for enhancing cancer-targeted drug delivery. Theranostics 2019; 9 (23) 6885-6900
  • 126 Wang P, Wang X, Luo Q. et al. Fabrication of red blood cell-based multimodal theranostic probes for second near-infrared window fluorescence imaging-guided tumor surgery and photodynamic therapy. Theranostics 2019; 9 (02) 369-380
  • 127 Ferguson LT, Hood ED, Shuvaeva T. et al. Dual affinity to RBCs and target cells (DART) enhances both organ- and cell type-targeting of intravascular nanocarriers. ACS Nano 2022; 16 (03) 4666-4683
  • 128 Yan M, Jurasz P. The role of platelets in the tumor microenvironment: from solid tumors to leukemia. Biochim Biophys Acta 2016; 1863 (03) 392-400
  • 129 Xu XR, Yousef GM, Ni H. Cancer and platelet crosstalk: opportunities and challenges for aspirin and other antiplatelet agents. Blood 2018; 131 (16) 1777-1789
  • 130 Liu Y, Zhang Y, Ding Y, Zhuang R. Platelet-mediated tumor metastasis mechanism and the role of cell adhesion molecules. Crit Rev Oncol Hematol 2021; 167: 103502
  • 131 Roweth HG, Battinelli EM. Lessons to learn from tumor-educated platelets. Blood 2021; 137 (23) 3174-3180
  • 132 Morris K, Schnoor B, Papa AL. Platelet cancer cell interplay as a new therapeutic target. Biochim Biophys Acta Rev Cancer 2022; 1877 (05) 188770
  • 133 Geranpayehvaghei M, Dabirmanesh B, Khaledi M. et al. Cancer-associated-platelet-inspired nanomedicines for cancer therapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2021; 13 (05) e1702
  • 134 Cacic D, Hervig T, Reikvam H. Platelets for advanced drug delivery in cancer. Expert Opin Drug Deliv 2023; 20 (05) 673-688
  • 135 Li S, Li L, Lin X, Chen C, Luo C, Huang Y. Targeted inhibition of tumor inflammation and tumor-platelet crosstalk by nanoparticle-mediated drug delivery mitigates cancer metastasis. ACS Nano 2022; 16 (01) 50-67
  • 136 Fan X, Wang K, Lu Q. et al. Surface-anchored tumor microenvironment-responsive protein nanogel-platelet system for cytosolic delivery of therapeutic protein in the post-surgical cancer treatment. Acta Biomater 2022; 154: 412-423
  • 137 Zhang Y, Zhu X, Chen X. et al. Activated platelets-targeting micelles with controlled drug release for effective treatment of primary and metastatic triple negative breast cancer. Adv Funct Mater 2019; 29 (13) 1806620
  • 138 Hu Q, Sun W, Wang J. et al. Conjugation of haematopoietic stem cells and platelets decorated with anti-PD-1 antibodies augments anti-leukaemia efficacy. Nat Biomed Eng 2018; 2 (11) 831-840
  • 139 Li Z, Ding Y, Liu J. et al. Depletion of tumor associated macrophages enhances local and systemic platelet-mediated anti-PD-1 delivery for post-surgery tumor recurrence treatment. Nat Commun 2022; 13 (01) 1845
  • 140 Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 2013; 14 (10) 1014-1022
  • 141 Mitchell MJ, King MR. Leukocytes as carriers for targeted cancer drug delivery. Expert Opin Drug Deliv 2015; 12 (03) 375-392
  • 142 Dong X, Chu D, Wang Z. Leukocyte-mediated delivery of nanotherapeutics in inflammatory and tumor sites. Theranostics 2017; 7 (03) 751-763
  • 143 Yang L, Zhang Y, Zhang Y. et al. Live macrophage-delivered doxorubicin-loaded liposomes effectively treat triple-negative breast cancer. ACS Nano 2022; 16 (06) 9799-9809
  • 144 Ye B, Zhao B, Wang K. et al. Neutrophils mediated multistage nanoparticle delivery for prompting tumor photothermal therapy. J Nanobiotechnology 2020; 18 (01) 138
  • 145 Hosseinalizadeh H, Mahmoodpour M, Razaghi Bahabadi Z, Hamblin MR, Mirzaei H. Neutrophil mediated drug delivery for targeted glioblastoma therapy: a comprehensive review. Biomed Pharmacother 2022; 156: 113841
  • 146 Wu Y, Han X, Zheng R. et al. Neutrophil mediated postoperative photoimmunotherapy against melanoma skin cancer. Nanoscale 2021; 13 (35) 14825-14836
  • 147 Ren K, He J, Qiu Y. et al. A neutrophil-mediated carrier regulates tumor stemness by inhibiting autophagy to prevent postoperative triple-negative breast cancer recurrence and metastasis. Acta Biomater 2022; 145: 185-199
  • 148 Luo Z, Lu Y, Shi Y. et al. Neutrophil hitchhiking for drug delivery to the bone marrow. Nat Nanotechnol 2023; 18 (06) 647-656
  • 149 Chu D, Dong X, Zhao Q, Gu J, Wang Z. Photosensitization priming of tumor microenvironments improves delivery of nanotherapeutics via neutrophil infiltration. Adv Mater 2017; 29 (27) 10 .1002/adma.201701021
  • 150 Jones RB, Mueller S, Kumari S. et al. Antigen recognition-triggered drug delivery mediated by nanocapsule-functionalized cytotoxic T-cells. Biomaterials 2017; 117: 44-53
  • 151 Wang X, Zhang Q, Zhou J. et al. T cell-mediated targeted delivery of tadalafil regulates immunosuppression and polyamine metabolism to overcome immune checkpoint blockade resistance in hepatocellular carcinoma. J Immunother Cancer 2023; 11 (02) e006493
  • 152 Niu W, Xiao Q, Wang X. et al. A biomimetic drug delivery system by integrating grapefruit extracellular vesicles and doxorubicin-loaded heparin-based nanoparticles for glioma therapy. Nano Lett 2021; 21 (03) 1484-1492
  • 153 Qiao Z, Zhang K, Liu J. et al. Biomimetic electrodynamic nanoparticles comprising ginger-derived extracellular vesicles for synergistic anti-infective therapy. Nat Commun 2022; 13 (01) 7164
  • 154 Han X, Bi L, Wu Y. et al. Genetically engineered exosomes for targetedly preventing premetastatic niche formation and suppressing postoperative melanoma lung metastasis. Nano Today 2022; 46: 101597
  • 155 Taieb J, Chaput N, Schartz N. et al. Chemoimmunotherapy of tumors: cyclophosphamide synergizes with exosome based vaccines. J Immunol 2006; 176 (05) 2722-2729
  • 156 Huang L, Rong Y, Tang X. et al. Engineered exosomes as an in situ DC-primed vaccine to boost antitumor immunity in breast cancer. Mol Cancer 2022; 21 (01) 45
  • 157 Huang C, Liu Z, Chen M. et al. Tumor-derived biomimetic nanozyme with immune evasion ability for synergistically enhanced low dose radiotherapy. J Nanobiotechnology 2021; 19 (01) 457
  • 158 Chen K, Wang Y, Liang H. et al. Intrinsic Biotaxi solution based on blood cell membrane cloaking enables fullerenol thrombolysis in vivo . ACS Appl Mater Interfaces 2020; 12 (13) 14958-14970
  • 159 Wu X, Zhang X, Feng W. et al. A targeted erythrocyte membrane-encapsulated drug-delivery system with anti-osteosarcoma and anti-osteolytic effects. ACS Appl Mater Interfaces 2021; 13 (24) 27920-27933
  • 160 Wu Y, Zhu R, Zhou M. et al. Homologous cancer cell membrane-camouflaged nanoparticles target drug delivery and enhance the chemotherapy efficacy of hepatocellular carcinoma. Cancer Lett 2023; 558: 216106
  • 161 Xie X, Hu X, Li Q. et al. Unraveling cell-type-specific targeted delivery of membrane-camouflaged nanoparticles with plasmonic imaging. Nano Lett 2020; 20 (07) 5228-5235
  • 162 Li Q, Su R, Bao X. et al. Glycyrrhetinic acid nanoparticles combined with ferrotherapy for improved cancer immunotherapy. Acta Biomater 2022; 144: 109-120
  • 163 Jun Y, Tang Z, Luo C. et al. Leukocyte-mediated combined targeted chemo and gene therapy for esophageal cancer. ACS Appl Mater Interfaces 2020; 12 (42) 47330-47341

Address for correspondence

Qin He, PhD
West China School of Pharmacy, Sichuan University
No. 17, Block 3, Southern Renmin Road, Chengdu 610041
People's Republic of China   

Publikationsverlauf

Eingereicht: 29. August 2023

Angenommen: 07. April 2024

Artikel online veröffentlicht:
22. Mai 2024

© 2024. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Sung H, Ferlay J, Siegel RL. et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 71 (03) 209-249
  • 2 Pusuluri A, Wu D, Mitragotri S. Immunological consequences of chemotherapy: single drugs, combination therapies and nanoparticle-based treatments. J Control Release 2019; 305: 130-154
  • 3 Ma Q, Cao J, Gao Y. et al. Microfluidic-mediated nano-drug delivery systems: from fundamentals to fabrication for advanced therapeutic applications. Nanoscale 2020; 12 (29) 15512-15527
  • 4 Ho YJ, Chiang YJ, Kang ST, Fan CH, Yeh CK. Camptothecin-loaded fusogenic nanodroplets as ultrasound theranostic agent in stem cell-mediated drug-delivery system. J Control Release 2018; 278: 100-109
  • 5 Wu HH, Zhou Y, Tabata Y, Gao JQ. Mesenchymal stem cell-based drug delivery strategy: from cells to biomimetic. J Control Release 2019; 294: 102-113
  • 6 Chen HY, Deng J, Wang Y, Wu CQ, Li X, Dai HW. Hybrid cell membrane-coated nanoparticles: a multifunctional biomimetic platform for cancer diagnosis and therapy. Acta Biomater 2020; 112: 1-13
  • 7 Chi J, Ma Q, Shen Z. et al. Targeted nanocarriers based on iodinated-cyanine dyes as immunomodulators for synergistic phototherapy. Nanoscale 2020; 12 (20) 11008-11025
  • 8 Guan YH, Wang N, Deng ZW, Chen XG, Liu Y. Exploiting autophagy-regulative nanomaterials for activation of dendritic cells enables reinforced cancer immunotherapy. Biomaterials 2022; 282: 121434
  • 9 Bush LM, Healy CP, Javdan SB, Emmons JC, Deans TL. Biological cells as therapeutic delivery vehicles. Trends Pharmacol Sci 2021; 42 (02) 106-118
  • 10 Elsharkasy OM, Nordin JZ, Hagey DW. et al. Extracellular vesicles as drug delivery systems: why and how?. Adv Drug Deliv Rev 2020; 159: 332-343
  • 11 Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol 2021; 16 (07) 748-759
  • 12 Liu T, Gao C, Gu D, Tang H. Cell-based carrier for targeted hitchhiking delivery. Drug Deliv Transl Res 2022; 12 (11) 2634-2648
  • 13 Zhang X, Li N, Zhang S. et al. Emerging carrier-free nanosystems based on molecular self-assembly of pure drugs for cancer therapy. Med Res Rev 2020; 40 (05) 1754-1775
  • 14 Ayer M, Klok HA. Cell-mediated delivery of synthetic nano- and microparticles. J Control Release 2017; 259: 92-104
  • 15 Fang RH, Gao W, Zhang L. Targeting drugs to tumours using cell membrane-coated nanoparticles. Nat Rev Clin Oncol 2023; 20 (01) 33-48
  • 16 Sun S, Yang Y, Gao Z. et al. endogenous stimuli-responsive autonomous separation of dual-targeting DNA guided missile from nanospacecraft for intelligent targeted cancer therapy. ACS Appl Mater Interfaces 2022; 14 (40) 45201-45216
  • 17 Théry C, Witwer KW, Aikawa E. et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles 2018; 7 (01) 1535750
  • 18 Zhang H, Lyden D. Asymmetric-flow field-flow fractionation technology for exomere and small extracellular vesicle separation and characterization. Nat Protoc 2019; 14 (04) 1027-1053
  • 19 van der Meel R, Fens MH, Vader P, van Solinge WW, Eniola-Adefeso O, Schiffelers RM. Extracellular vesicles as drug delivery systems: lessons from the liposome field. J Control Release 2014; 195: 72-85
  • 20 Piffoux M, Silva AKA, Wilhelm C, Gazeau F, Tareste D. Modification of extracellular vesicles by fusion with liposomes for the design of personalized biogenic drug delivery systems. ACS Nano 2018; 12 (07) 6830-6842
  • 21 Schulz-Siegmund M, Aigner A. Nucleic acid delivery with extracellular vesicles. Adv Drug Deliv Rev 2021; 173: 89-111
  • 22 Ou YH, Liang J, Czarny B. et al. Extracellular vesicle (EV) biohybrid systems for cancer therapy: recent advances and future perspectives. Semin Cancer Biol 2021; 74: 45-61
  • 23 Richter M, Vader P, Fuhrmann G. Approaches to surface engineering of extracellular vesicles. Adv Drug Deliv Rev 2021; 173: 416-426
  • 24 Sharma S, Masud MK, Kaneti YV. et al. Extracellular vesicle nanoarchitectonics for novel drug delivery applications. Small 2021; 17 (42) e2102220
  • 25 Roerig J, Schulz-Siegmund M. Standardization approaches for extracellular vesicle loading with oligonucleotides and biologics. Small 2023; 19 (40) e2301763
  • 26 Liu YR, Cheng YQ, Wang SB. et al. Therapeutic effects and perspective of stem cell extracellular vesicles in aging and cancer. J Cell Physiol 2021; 236 (07) 4783-4796
  • 27 Lan T, Luo M, Wei X. Mesenchymal stem/stromal cells in cancer therapy. J Hematol Oncol 2021; 14 (01) 195
  • 28 Keshtkar S, Azarpira N, Ghahremani MH. Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Res Ther 2018; 9 (01) 63
  • 29 Hass R, Kasper C, Böhm S, Jacobs R. Different populations and sources of human mesenchymal stem cells (MSC): a comparison of adult and neonatal tissue-derived MSC. Cell Commun Signal 2011; 9: 12
  • 30 Rani S, Ryan AE, Griffin MD, Ritter T. Mesenchymal stem cell-derived extracellular vesicles: toward cell-free therapeutic applications. Mol Ther 2015; 23 (05) 812-823
  • 31 Xu F, Fei Z, Dai H. et al. Mesenchymal stem cell-derived extracellular vesicles with high PD-L1 expression for autoimmune diseases treatment. Adv Mater 2022; 34 (01) e2106265
  • 32 Lai RC, Yeo RW, Lim SK. Mesenchymal stem cell exosomes. Semin Cell Dev Biol 2015; 40: 82-88
  • 33 Su C, Zhang J, Yarden Y, Fu L. The key roles of cancer stem cell-derived extracellular vesicles. Signal Transduct Target Ther 2021; 6 (01) 109
  • 34 Sohrabi B, Dayeri B, Zahedi E. et al. Mesenchymal stem cell (MSC)-derived exosomes as novel vehicles for delivery of miRNAs in cancer therapy. Cancer Gene Ther 2022; 29 (8–9): 1105-1116
  • 35 Sun Y, Liu G, Zhang K, Cao Q, Liu T, Li J. Mesenchymal stem cells-derived exosomes for drug delivery. Stem Cell Res Ther 2021; 12 (01) 561
  • 36 Crivelli B, Chlapanidas T, Perteghella S. et al; Italian Mesenchymal Stem Cell Group (GISM). Mesenchymal stem/stromal cell extracellular vesicles: from active principle to next generation drug delivery system. J Control Release 2017; 262: 104-117
  • 37 Xunian Z, Kalluri R. Biology and therapeutic potential of mesenchymal stem cell-derived exosomes. Cancer Sci 2020; 111 (09) 3100-3110
  • 38 Jahangiri B, Khalaj-Kondori M, Asadollahi E, Purrafee Dizaj L, Sadeghizadeh M. MSC-Derived exosomes suppress colorectal cancer cell proliferation and metastasis via miR-100/mTOR/miR-143 pathway. Int J Pharm 2022; 627: 122214
  • 39 Yao X, Mao Y, Wu D. et al. Exosomal circ_0030167 derived from BM-MSCs inhibits the invasion, migration, proliferation and stemness of pancreatic cancer cells by sponging miR-338-5p and targeting the Wif1/Wnt8/β-catenin axis. Cancer Lett 2021; 512: 38-50
  • 40 Pascucci L, Coccè V, Bonomi A. et al. Paclitaxel is incorporated by mesenchymal stromal cells and released in exosomes that inhibit in vitro tumor growth: a new approach for drug delivery. J Control Release 2014; 192: 262-270
  • 41 Pinto A, Marangon I, Méreaux J. et al. Immune reprogramming precision photodynamic therapy of peritoneal metastasis by scalable stem-cell-derived extracellular vesicles. ACS Nano 2021; 15 (02) 3251-3263
  • 42 Luo T, Liu Q, Tan A. et al. Mesenchymal stem cell-secreted exosome promotes chemoresistance in breast cancer via enhancing miR-21-5p-mediated S100A6 expression. Mol Ther Oncolytics 2020; 19: 283-293
  • 43 Zhu S, Yang N, Wu J. et al. Tumor microenvironment-related dendritic cell deficiency: a target to enhance tumor immunotherapy. Pharmacol Res 2020; 159: 104980
  • 44 Clark GJ, Silveira PA, Hogarth PM, Hart DNJ. The cell surface phenotype of human dendritic cells. Semin Cell Dev Biol 2019; 86: 3-14
  • 45 Bol KF, Schreibelt G, Gerritsen WR, de Vries IJ, Figdor CG. Dendritic cell-based immunotherapy: state of the art and beyond. Clin Cancer Res 2016; 22 (08) 1897-1906
  • 46 Macri C, Pang ES, Patton T, O'Keeffe M. Dendritic cell subsets. Semin Cell Dev Biol 2018; 84: 11-21
  • 47 Worbs T, Hammerschmidt SI, Förster R. Dendritic cell migration in health and disease. Nat Rev Immunol 2017; 17 (01) 30-48
  • 48 Li J, Li J, Peng Y, Du Y, Yang Z, Qi X. Dendritic cell derived exosomes loaded neoantigens for personalized cancer immunotherapies. J Control Release 2023; 353: 423-433
  • 49 Näslund TI, Gehrmann U, Qazi KR, Karlsson MC, Gabrielsson S. Dendritic cell-derived exosomes need to activate both T and B cells to induce antitumor immunity. J Immunol 2013; 190 (06) 2712-2719
  • 50 Xiong X, Ke X, Wang L. et al. Neoantigen-based cancer vaccination using chimeric RNA-loaded dendritic cell-derived extracellular vesicles. J Extracell Vesicles 2022; 11 (08) e12243
  • 51 Esser J, Gehrmann U, D'Alexandri FL. et al. Exosomes from human macrophages and dendritic cells contain enzymes for leukotriene biosynthesis and promote granulocyte migration. J Allergy Clin Immunol 2010; 126 (05) 1032-1040 , 1040.e1–1040.e4
  • 52 Pitt JM, Charrier M, Viaud S. et al. Dendritic cell-derived exosomes as immunotherapies in the fight against cancer. J Immunol 2014; 193 (03) 1006-1011
  • 53 Fan M, Liu H, Yan H. et al. A CAR T-inspiring platform based on antibody-engineered exosomes from antigen-feeding dendritic cells for precise solid tumor therapy. Biomaterials 2022; 282: 121424
  • 54 Zhu H, Wang K, Wang Z. et al. An efficient and safe MUC1-dendritic cell-derived exosome conjugate vaccine elicits potent cellular and humoral immunity and tumor inhibition in vivo . Acta Biomater 2022; 138: 491-504
  • 55 Lu Z, Zuo B, Jing R. et al. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol 2017; 67 (04) 739-748
  • 56 Viaud S, Terme M, Flament C. et al. Dendritic cell-derived exosomes promote natural killer cell activation and proliferation: a role for NKG2D ligands and IL-15Ralpha. PLoS One 2009; 4 (03) e4942
  • 57 Zitvogel L, Regnault A, Lozier A. et al. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat Med 1998; 4 (05) 594-600
  • 58 Damo M, Wilson DS, Simeoni E, Hubbell JA. TLR-3 stimulation improves anti-tumor immunity elicited by dendritic cell exosome-based vaccines in a murine model of melanoma. Sci Rep 2015; 5: 17622
  • 59 Tian X, Shen H, Li Z, Wang T, Wang S. Tumor-derived exosomes, myeloid-derived suppressor cells, and tumor microenvironment. J Hematol Oncol 2019; 12 (01) 84
  • 60 Jiang C, Zhang N, Hu X, Wang H. Tumor-associated exosomes promote lung cancer metastasis through multiple mechanisms. Mol Cancer 2021; 20 (01) 117
  • 61 Naseri M, Bozorgmehr M, Zöller M, Ranaei Pirmardan E, Madjd Z. Tumor-derived exosomes: the next generation of promising cell-free vaccines in cancer immunotherapy. OncoImmunology 2020; 9 (01) 1779991
  • 62 Mashouri L, Yousefi H, Aref AR, Ahadi AM, Molaei F, Alahari SK. Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol Cancer 2019; 18 (01) 75
  • 63 Gao Y, Xu H, Li N. et al. Renal cancer-derived exosomes induce tumor immune tolerance by MDSCs-mediated antigen-specific immunosuppression. Cell Commun Signal 2020; 18 (01) 106
  • 64 Ma Z, Wei K, Yang F. et al. Tumor-derived exosomal miR-3157-3p promotes angiogenesis, vascular permeability and metastasis by targeting TIMP/KLF2 in non-small cell lung cancer. Cell Death Dis 2021; 12 (09) 840
  • 65 Guo X, Sui R, Piao H. Tumor-derived small extracellular vesicles: potential roles and mechanism in glioma. J Nanobiotechnology 2022; 20 (01) 383
  • 66 Hoshino A, Costa-Silva B, Shen TL. et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015; 527 (7578) 329-335
  • 67 Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science 2020; 367 (6478) eaau6977
  • 68 Thakur A, Parra DC, Motallebnejad P, Brocchi M, Chen HJ. Exosomes: small vesicles with big roles in cancer, vaccine development, and therapeutics. Bioact Mater 2021; 10: 281-294
  • 69 Qiao L, Hu S, Huang K. et al. Tumor cell-derived exosomes home to their cells of origin and can be used as Trojan horses to deliver cancer drugs. Theranostics 2020; 10 (08) 3474-3487
  • 70 Andre F, Schartz NE, Movassagh M. et al. Malignant effusions and immunogenic tumour-derived exosomes. Lancet 2002; 360 (9329) 295-305
  • 71 Taghikhani A, Hassan ZM, Ebrahimi M, Moazzeni SM. microRNA modified tumor-derived exosomes as novel tools for maturation of dendritic cells. J Cell Physiol 2019; 234 (06) 9417-9427
  • 72 Gong H, Zhang Q, Komarla A. et al. Nanomaterial biointerfacing via mitochondrial membrane coating for targeted detoxification and molecular detection. Nano Lett 2021; 21 (06) 2603-2609
  • 73 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 (08) 3233-3254
  • 74 Zhen X, Cheng P, Pu K. Recent advances in cell membrane-camouflaged nanoparticles for cancer phototherapy. Small 2019; 15 (01) e1804105
  • 75 Dash P, Piras AM, Dash M. Cell membrane coated nanocarriers - an efficient biomimetic platform for targeted therapy. J Control Release 2020; 327: 546-570
  • 76 Liu L, Pan D, Chen S. et al. Systematic design of cell membrane coating to improve tumor targeting of nanoparticles. Nat Commun 2022; 13 (01) 6181
  • 77 Luk BT, Zhang L. Cell membrane-camouflaged nanoparticles for drug delivery. J Control Release 2015; 220 (Pt B): 600-607
  • 78 Wang Y, Chen X, He D, Zhou Y, Qin L. Surface-modified nanoerythrocyte loading DOX for targeted liver cancer chemotherapy. Mol Pharm 2018; 15 (12) 5728-5740
  • 79 Fu S, Liang M, Wang Y. et al. Dual-modified novel biomimetic nanocarriers improve targeting and therapeutic efficacy in glioma. ACS Appl Mater Interfaces 2019; 11 (02) 1841-1854
  • 80 Tao C, Nie X, Zhu W, Iqbal J, Xu C, Wang DA. Autologous cell membrane coatings on tissue engineering xenografts for suppression and alleviation of acute host immune responses. Biomaterials 2020; 258: 120310
  • 81 Han X, Wang C, Liu Z. Red blood cells as smart delivery systems. Bioconjug Chem 2018; 29 (04) 852-860
  • 82 Peng S, Ouyang B, Men Y. et al. Biodegradable zwitterionic polymer membrane coating endowing nanoparticles with ultra-long circulation and enhanced tumor photothermal therapy. Biomaterials 2020; 231: 119680
  • 83 Ye S, Wang F, Fan Z. et al. Light/pH-triggered biomimetic red blood cell membranes camouflaged small molecular drug assemblies for imaging-guided combinational chemo-photothermal therapy. ACS Appl Mater Interfaces 2019; 11 (17) 15262-15275
  • 84 Zhang Y, Xia Q, Wu T. et al. A novel multi-functionalized multicellular nanodelivery system for non-small cell lung cancer photochemotherapy. J Nanobiotechnology 2021; 19 (01) 245
  • 85 Xie H, Li W, Liu H. et al. Erythrocyte membrane-coated invisible acoustic-sensitive nanoparticle for inducing tumor thrombotic infarction by precisely damaging tumor vascular endothelium. Small 2022; 18 (30) e2201933
  • 86 Wang Y, Ji X, Ruan M. et al. Worm-like biomimetic nanoerythrocyte carrying siRNA for melanoma gene therapy. Small 2018; 14 (47) e1803002
  • 87 Zhang Z, Qian H, Huang J. et al. Anti-EGFR-iRGD recombinant protein modified biomimetic nanoparticles loaded with gambogic acid to enhance targeting and antitumor ability in colorectal cancer treatment. Int J Nanomedicine 2018; 13: 4961-4975
  • 88 Miao Y, Yang Y, Guo L. et al. Cell membrane-camouflaged nanocarriers with biomimetic deformability of erythrocytes for ultralong circulation and enhanced cancer therapy. ACS Nano 2022; 16 (04) 6527-6540
  • 89 Guo H, Zhang W, Wang L, Shao Z, Huang X. Biomimetic cell membrane-coated glucose/oxygen-exhausting nanoreactor for remodeling tumor microenvironment in targeted hypoxic tumor therapy. Biomaterials 2022; 290: 121821
  • 90 Pan WL, Tan Y, Meng W. et al. Microenvironment-driven sequential ferroptosis, photodynamic therapy, and chemotherapy for targeted breast cancer therapy by a cancer-cell-membrane-coated nanoscale metal-organic framework. Biomaterials 2022; 283: 121449
  • 91 Fang Z, Zhang M, Kang R, Cui M, Song M, Liu K. A cancer cell membrane coated nanoparticles-based gene delivery system for enhancing cancer therapy. Int J Pharm 2022; 629: 122415
  • 92 Gan J, Du G, He C. et al. Tumor cell membrane enveloped aluminum phosphate nanoparticles for enhanced cancer vaccination. J Control Release 2020; 326: 297-309
  • 93 Jin J, Krishnamachary B, Barnett JD. et al. Human cancer cell membrane-coated biomimetic nanoparticles reduce fibroblast-mediated invasion and metastasis and induce T-cells. ACS Appl Mater Interfaces 2019; 11 (08) 7850-7861
  • 94 Wang Z, Zhang M, Chi S, Zhu M, Wang C, Liu Z. Brain tumor cell membrane-coated lanthanide-doped nanoparticles for NIR-IIb luminescence imaging and surgical navigation of glioma. Adv Healthc Mater 2022; 11 (16) e2200521
  • 95 Chen M, Cui Y, Hao W. et al. Ligand-modified homologous targeted cancer cell membrane biomimetic nanostructured lipid carriers for glioma therapy. Drug Deliv 2021; 28 (01) 2241-2255
  • 96 Zheng B, Liu Z, Wang H. et al. R11 modified tumor cell membrane nanovesicle-camouflaged nanoparticles with enhanced targeting and mucus-penetrating efficiency for intravesical chemotherapy for bladder cancer. J Control Release 2022; 351: 834-846
  • 97 Mohale S, Kunde SS, Wairkar S. Biomimetic fabrication of nanotherapeutics by leukocyte membrane cloaking for targeted therapy. Colloids Surf B Biointerfaces 2022; 219: 112803
  • 98 Wang D, Wang S, Zhou Z. et al. White blood cell membrane-coated nanoparticles: recent development and medical applications. Adv Healthc Mater 2022; 11 (07) e2101349
  • 99 Zhou X, Luo B, Kang K. et al. Leukocyte-repelling biomimetic immunomagnetic nanoplatform for high-performance circulating tumor cells isolation. Small 2019; 15 (17) e1900558
  • 100 Xia Y, Rao L, Yao H, Wang Z, Ning P, Chen X. Engineering macrophages for cancer immunotherapy and drug delivery. Adv Mater 2020; 32 (40) e2002054
  • 101 Chen C, Song M, Du Y. et al. Tumor-associated-macrophage-membrane-coated nanoparticles for improved photodynamic immunotherapy. Nano Lett 2021; 21 (13) 5522-5531
  • 102 Li J, Wu Y, Wang J. et al. Macrophage membrane-coated nano-gemcitabine promotes lymphocyte infiltration and synergizes antiPD-L1 to restore the tumoricidal function. ACS Nano 2023; 17 (01) 322-336
  • 103 Wang W, Wu F, Mohammadniaei M. et al. Genetically edited T-cell membrane coated AIEgen nanoparticles effectively prevents glioblastoma recurrence. Biomaterials 2023; 293: 121981
  • 104 Kang M, Hong J, Jung M. et al. T-Cell-mimicking nanoparticles for cancer immunotherapy. Adv Mater 2020; 32 (39) e2003368
  • 105 Hao W, Cui Y, Fan Y. et al. Hybrid membrane-coated nanosuspensions for multi-modal anti-glioma therapy via drug and antigen delivery. J Nanobiotechnology 2021; 19 (01) 378
  • 106 Wu L, Li Q, Deng J. et al. Platelet-tumor cell hybrid membrane-camouflaged nanoparticles for enhancing therapy efficacy in glioma. Int J Nanomedicine 2021; 16: 8433-8446
  • 107 Zhao Y, Li A, Jiang L, Gu Y, Liu J. Hybrid membrane-coated biomimetic nanoparticles (HM@BNPs): a multifunctional nanomaterial for biomedical applications. Biomacromolecules 2021; 22 (08) 3149-3167
  • 108 Liao Y, Zhang Y, Blum NT, Lin J, Huang P. Biomimetic hybrid membrane-based nanoplatforms: synthesis, properties and biomedical applications. Nanoscale Horiz 2020; 5 (09) 1293-1302
  • 109 Shen T, Yang S, Qu X. et al. A bionic “Trojan horse”-like gene delivery system hybridized with tumor and macrophage cell membrane for cancer therapy. J Control Release 2023; 358: 204-218
  • 110 Ma W, Yang Y, Zhu J. et al. Biomimetic nanoerythrosome-coated aptamer-DNA tetrahedron/maytansine conjugates: pH-responsive and targeted cytotoxicity for HER2-positive breast cancer. Adv Mater 2022; 34 (46) e2109609
  • 111 Zhang W, Gong C, Chen Z, Li M, Li Y, Gao J. Tumor microenvironment-activated cancer cell membrane-liposome hybrid nanoparticle-mediated synergistic metabolic therapy and chemotherapy for non-small cell lung cancer. J Nanobiotechnology 2021; 19 (01) 339
  • 112 Chen H, Deng J, Yao X. et al. Bone-targeted erythrocyte-cancer hybrid membrane-camouflaged nanoparticles for enhancing photothermal and hypoxia-activated chemotherapy of bone invasion by OSCC. J Nanobiotechnology 2021; 19 (01) 342
  • 113 Zang S, Huang K, Li J. et al. Metabolic reprogramming by dual-targeting biomimetic nanoparticles for enhanced tumor chemo-immunotherapy. Acta Biomater 2022; 148: 181-193
  • 114 Nikfar M, Razizadeh M, Paul R, Muzykantov V, Liu Y. A numerical study on drug delivery via multiscale synergy of cellular hitchhiking onto red blood cells. Nanoscale 2021; 13 (41) 17359-17372
  • 115 Anselmo AC, Gupta V, Zern BJ. et al. Delivering nanoparticles to lungs while avoiding liver and spleen through adsorption on red blood cells. ACS Nano 2013; 7 (12) 11129-11137
  • 116 Su Y, Xie Z, Kim GB, Dong C, Yang J. Design strategies and applications of circulating cell-mediated drug delivery systems. ACS Biomater Sci Eng 2015; 1 (04) 201-217
  • 117 Yang L, Yang Y, Chen Y, Xu Y, Peng J. Cell-based drug delivery systems and their in vivo fate. Adv Drug Deliv Rev 2022; 187: 114394
  • 118 Anselmo AC, Mitragotri S. Cell-mediated delivery of nanoparticles: taking advantage of circulatory cells to target nanoparticles. J Control Release 2014; 190: 531-541
  • 119 Villa CH, Cines DB, Siegel DL, Muzykantov V. Erythrocytes as carriers for drug delivery in blood transfusion and beyond. Transfus Med Rev 2017; 31 (01) 26-35
  • 120 Mukthavaram R, Shi G, Kesari S, Simberg D. Targeting and depletion of circulating leukocytes and cancer cells by lipophilic antibody-modified erythrocytes. J Control Release 2014; 183: 146-153
  • 121 Li Y, Raza F, Liu Y. et al. Clinical progress and advanced research of red blood cells based drug delivery system. Biomaterials 2021; 279: 121202
  • 122 Parodi A, Molinaro R, Sushnitha M. et al. Bio-inspired engineering of cell- and virus-like nanoparticles for drug delivery. Biomaterials 2017; 147: 155-168
  • 123 Brenner JS, Pan DC, Myerson JW. et al. Red blood cell-hitchhiking boosts delivery of nanocarriers to chosen organs by orders of magnitude. Nat Commun 2018; 9 (01) 2684
  • 124 Wang S, Ma S, Li R. et al. Probing the interaction between supercarrier RBC membrane and nanoparticles for optimal drug delivery. J Mol Biol 2023; 435 (01) 167539
  • 125 Sun D, Chen J, Wang Y. et al. Advances in refunctionalization of erythrocyte-based nanomedicine for enhancing cancer-targeted drug delivery. Theranostics 2019; 9 (23) 6885-6900
  • 126 Wang P, Wang X, Luo Q. et al. Fabrication of red blood cell-based multimodal theranostic probes for second near-infrared window fluorescence imaging-guided tumor surgery and photodynamic therapy. Theranostics 2019; 9 (02) 369-380
  • 127 Ferguson LT, Hood ED, Shuvaeva T. et al. Dual affinity to RBCs and target cells (DART) enhances both organ- and cell type-targeting of intravascular nanocarriers. ACS Nano 2022; 16 (03) 4666-4683
  • 128 Yan M, Jurasz P. The role of platelets in the tumor microenvironment: from solid tumors to leukemia. Biochim Biophys Acta 2016; 1863 (03) 392-400
  • 129 Xu XR, Yousef GM, Ni H. Cancer and platelet crosstalk: opportunities and challenges for aspirin and other antiplatelet agents. Blood 2018; 131 (16) 1777-1789
  • 130 Liu Y, Zhang Y, Ding Y, Zhuang R. Platelet-mediated tumor metastasis mechanism and the role of cell adhesion molecules. Crit Rev Oncol Hematol 2021; 167: 103502
  • 131 Roweth HG, Battinelli EM. Lessons to learn from tumor-educated platelets. Blood 2021; 137 (23) 3174-3180
  • 132 Morris K, Schnoor B, Papa AL. Platelet cancer cell interplay as a new therapeutic target. Biochim Biophys Acta Rev Cancer 2022; 1877 (05) 188770
  • 133 Geranpayehvaghei M, Dabirmanesh B, Khaledi M. et al. Cancer-associated-platelet-inspired nanomedicines for cancer therapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2021; 13 (05) e1702
  • 134 Cacic D, Hervig T, Reikvam H. Platelets for advanced drug delivery in cancer. Expert Opin Drug Deliv 2023; 20 (05) 673-688
  • 135 Li S, Li L, Lin X, Chen C, Luo C, Huang Y. Targeted inhibition of tumor inflammation and tumor-platelet crosstalk by nanoparticle-mediated drug delivery mitigates cancer metastasis. ACS Nano 2022; 16 (01) 50-67
  • 136 Fan X, Wang K, Lu Q. et al. Surface-anchored tumor microenvironment-responsive protein nanogel-platelet system for cytosolic delivery of therapeutic protein in the post-surgical cancer treatment. Acta Biomater 2022; 154: 412-423
  • 137 Zhang Y, Zhu X, Chen X. et al. Activated platelets-targeting micelles with controlled drug release for effective treatment of primary and metastatic triple negative breast cancer. Adv Funct Mater 2019; 29 (13) 1806620
  • 138 Hu Q, Sun W, Wang J. et al. Conjugation of haematopoietic stem cells and platelets decorated with anti-PD-1 antibodies augments anti-leukaemia efficacy. Nat Biomed Eng 2018; 2 (11) 831-840
  • 139 Li Z, Ding Y, Liu J. et al. Depletion of tumor associated macrophages enhances local and systemic platelet-mediated anti-PD-1 delivery for post-surgery tumor recurrence treatment. Nat Commun 2022; 13 (01) 1845
  • 140 Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 2013; 14 (10) 1014-1022
  • 141 Mitchell MJ, King MR. Leukocytes as carriers for targeted cancer drug delivery. Expert Opin Drug Deliv 2015; 12 (03) 375-392
  • 142 Dong X, Chu D, Wang Z. Leukocyte-mediated delivery of nanotherapeutics in inflammatory and tumor sites. Theranostics 2017; 7 (03) 751-763
  • 143 Yang L, Zhang Y, Zhang Y. et al. Live macrophage-delivered doxorubicin-loaded liposomes effectively treat triple-negative breast cancer. ACS Nano 2022; 16 (06) 9799-9809
  • 144 Ye B, Zhao B, Wang K. et al. Neutrophils mediated multistage nanoparticle delivery for prompting tumor photothermal therapy. J Nanobiotechnology 2020; 18 (01) 138
  • 145 Hosseinalizadeh H, Mahmoodpour M, Razaghi Bahabadi Z, Hamblin MR, Mirzaei H. Neutrophil mediated drug delivery for targeted glioblastoma therapy: a comprehensive review. Biomed Pharmacother 2022; 156: 113841
  • 146 Wu Y, Han X, Zheng R. et al. Neutrophil mediated postoperative photoimmunotherapy against melanoma skin cancer. Nanoscale 2021; 13 (35) 14825-14836
  • 147 Ren K, He J, Qiu Y. et al. A neutrophil-mediated carrier regulates tumor stemness by inhibiting autophagy to prevent postoperative triple-negative breast cancer recurrence and metastasis. Acta Biomater 2022; 145: 185-199
  • 148 Luo Z, Lu Y, Shi Y. et al. Neutrophil hitchhiking for drug delivery to the bone marrow. Nat Nanotechnol 2023; 18 (06) 647-656
  • 149 Chu D, Dong X, Zhao Q, Gu J, Wang Z. Photosensitization priming of tumor microenvironments improves delivery of nanotherapeutics via neutrophil infiltration. Adv Mater 2017; 29 (27) 10 .1002/adma.201701021
  • 150 Jones RB, Mueller S, Kumari S. et al. Antigen recognition-triggered drug delivery mediated by nanocapsule-functionalized cytotoxic T-cells. Biomaterials 2017; 117: 44-53
  • 151 Wang X, Zhang Q, Zhou J. et al. T cell-mediated targeted delivery of tadalafil regulates immunosuppression and polyamine metabolism to overcome immune checkpoint blockade resistance in hepatocellular carcinoma. J Immunother Cancer 2023; 11 (02) e006493
  • 152 Niu W, Xiao Q, Wang X. et al. A biomimetic drug delivery system by integrating grapefruit extracellular vesicles and doxorubicin-loaded heparin-based nanoparticles for glioma therapy. Nano Lett 2021; 21 (03) 1484-1492
  • 153 Qiao Z, Zhang K, Liu J. et al. Biomimetic electrodynamic nanoparticles comprising ginger-derived extracellular vesicles for synergistic anti-infective therapy. Nat Commun 2022; 13 (01) 7164
  • 154 Han X, Bi L, Wu Y. et al. Genetically engineered exosomes for targetedly preventing premetastatic niche formation and suppressing postoperative melanoma lung metastasis. Nano Today 2022; 46: 101597
  • 155 Taieb J, Chaput N, Schartz N. et al. Chemoimmunotherapy of tumors: cyclophosphamide synergizes with exosome based vaccines. J Immunol 2006; 176 (05) 2722-2729
  • 156 Huang L, Rong Y, Tang X. et al. Engineered exosomes as an in situ DC-primed vaccine to boost antitumor immunity in breast cancer. Mol Cancer 2022; 21 (01) 45
  • 157 Huang C, Liu Z, Chen M. et al. Tumor-derived biomimetic nanozyme with immune evasion ability for synergistically enhanced low dose radiotherapy. J Nanobiotechnology 2021; 19 (01) 457
  • 158 Chen K, Wang Y, Liang H. et al. Intrinsic Biotaxi solution based on blood cell membrane cloaking enables fullerenol thrombolysis in vivo . ACS Appl Mater Interfaces 2020; 12 (13) 14958-14970
  • 159 Wu X, Zhang X, Feng W. et al. A targeted erythrocyte membrane-encapsulated drug-delivery system with anti-osteosarcoma and anti-osteolytic effects. ACS Appl Mater Interfaces 2021; 13 (24) 27920-27933
  • 160 Wu Y, Zhu R, Zhou M. et al. Homologous cancer cell membrane-camouflaged nanoparticles target drug delivery and enhance the chemotherapy efficacy of hepatocellular carcinoma. Cancer Lett 2023; 558: 216106
  • 161 Xie X, Hu X, Li Q. et al. Unraveling cell-type-specific targeted delivery of membrane-camouflaged nanoparticles with plasmonic imaging. Nano Lett 2020; 20 (07) 5228-5235
  • 162 Li Q, Su R, Bao X. et al. Glycyrrhetinic acid nanoparticles combined with ferrotherapy for improved cancer immunotherapy. Acta Biomater 2022; 144: 109-120
  • 163 Jun Y, Tang Z, Luo C. et al. Leukocyte-mediated combined targeted chemo and gene therapy for esophageal cancer. ACS Appl Mater Interfaces 2020; 12 (42) 47330-47341

Zoom Image
Scheme 1 A brief introduction of the biomimetic drug delivery system.
Zoom Image
Fig. 1 A schematic representation of the molecular mechanisms by which MSC-Exos affects cell proliferation and metastasis in CRC cells. CRC, colorectal cell. (Reproduced with permission from Jahangiri et al[38], copyright 2022 Elsevier B.V. All rights reserved.)
Zoom Image
Fig. 2 Schematic diagram. (A) Schematic of the construction of anti-CD3 and anti-EGFR antibody-engineered tDC-Exo (Exo-OVA-aCD3/aEGFR). (B) MHC–antigen complex and a co-stimulating molecule CD86 on Exo-OVA-aCD3/aEGFR can be regarded as the CAR of CAR T cells which could activate endogenous T cells in vivo. (Reproduced with permission from Fan et al[53], copyright 2022 Elsevier B.V. All rights reserved.)
Zoom Image
Fig. 3 (A) Schematic illustration of the preparation of cancer cell plasma membrane fraction-coated PLGA NPs (CCMF-PLGA NPs). (B) The purpose was to determine the ability of these cancer cell-mimicking NPs to disrupt cancer cell–stromal cell interactions, reduce metastasis, and prime the immune system for cancer immunotherapy. NPs, nanoparticles. (Reproduced with permission from Jin et al[93], copyright 2019 American Chemical Society.)
Zoom Image
Fig. 4 Schematic summary of using LM-coated nanoparticles for medical applications. LM NPs are made by cloaking plasma membranes derived from natural lymphocytes onto synthetic cores. LM, leukocyte membrane; NP, nanoparticle. (Reproduced with permission from Wang et al[98], copyright 2022 Wiley-VCH GmbH.)
Zoom Image
Fig. 5 Hybrid cell membrane-camouflaged nanoparticles (HMC@NPs) designed for cancer diagnosis and treatment. Cell membranes from different types of natural cells were extracted and leveraged to wrap around different nanoparticles for the theranostic of cancer. (Reproduced with permission from Chen et al[6], copyright 2020 Elsevier B.V. All rights reserved.)
Zoom Image
Fig. 6 Advantages of nanoparticles and circulatory cells in drug delivery. Circulatory cells, including red blood cells, monocytes, platelets, and lymphocytes, have natural drug-delivery abilities. (Reproduced with permission from Anselmo et al[118], copyright 2014 Elsevier B.V. All rights reserved.)
Zoom Image
Fig. 7 A schematic diagram of (A) lipid insertion, (B) biotin–avidin bridges, (C) EDC/NHS coupling, (D) antibody/ligand-receptor conjugation, and (E) passive adsorption (hitchhiking) methods for refunctionalization of erythrocyte-based nanomedicine. (Reproduced with permission from Sun et al[125], copyright 2019 Ivyspring International Publisher.)
Zoom Image
Fig. 8 Schematic illustration for the antitumor mechanism of nanodrug in overcoming immune checkpoint blockade resistance in HCC. HCC, hepatocellular carcinoma. (Reproduced with permission from Wang et al[151], copyright 2023 BMJ Publishing Group Ltd & Society for Immunotherapy of Cancer.)