Abstract
Cardiovascular diseases (CVDs) continue to represent a major challenge to global health, highlighting the urgent need for innovative treatment approaches. As a growing interdisciplinary field, nanotechnology has demonstrated significant potential for clinical applications. Nanomedicine development primarily focuses on improving the disease diagnosis and treatment through leveraging the distinct characteristics of engineered nanoparticles (NPs) to detect disease markers or deliver therapeutics to specific targets. Specifically, cell membrane-coated NPs offer enhanced targeting, biostability, and immune evasion. A significant benefit associated with this technology lies in its capacity to retain the functional and intrinsic properties of the source cells. However, while each cell membrane possesses distinct characteristics, a single cell membrane may not always address complex functional requirements. By combining membranes from different cell types, it becomes possible to integrate diverse functionalities, resulting in a more comprehensive solution. In this review, we aim to explore recent advancements in the hybrid cell membrane-coated NPs (HM/NPs) and their potential applications for the treatment of CVDs. We highlight the mechanisms underlying HM/NPs and their utilization in the therapeutic management of CVDs. Additionally, we examine the potential for clinical translation and discuss the key challenges encountered in this process.
Keywords: Hybrid cell membrane coating, Biomimetic nanoparticles, Targeted delivery, Immune evasion, Cardiovascular disease treatment, Nanomedicine
Graphical abstract

Highlights
-
•
Recent advancements in the hybrid cell membrane-coated nanoparticles (HM/NPs) for treating cardiovascular diseases (CVDs) are reviewed.
-
•
HM/NPs integrating functionalities from multiple cell types are discussed.
-
•
Various construction strategies for HM/NPs and their respective advantages and disadvantages are highlighted.
-
•
Challenges and potential of HM/NPs for the treatment of CVDs from bench to clinic are explored.
1. Introduction
Cardiovascular diseases (CVDs) represent the primary contributor to morbidity and mortality across both developed and developing countries [[1], [2], [3]]. CVDs account for approximately 32 % (17.9 million cases) of total global mortality annually, with acute myocardial infarction (MI) and cerebrovascular accidents constituting the predominant etiologies, representing 85 % of CVD-related fatalities [4]. Each year in Europe, CVDs result in the loss of over 60 million potential life-years. Although the absolute number of deaths from CVDs is higher among women, men experience significantly greater incidence and mortality rates. These sex-based disparities in disease burden are especially pronounced in younger populations under the age of 70 [5]. CVDs impose a significant economic burden globally, with annual costs in the European Union estimated at €282 billion [6]. The term CVDs encompass a range of disorders affecting the cardiovascular system, including atherosclerosis, myocardial ischemia/reperfusion injury (MI/RI), heart failure, stroke, and angina pectoris [7]. Recent advancements in understanding CVDs have driven the development of new clinical strategies and preclinical research aimed at preventing or delaying myocardial dysfunction [8]. However, despite these advances, the persistently high mortality rates associated with heart failure underscore the significant the limitations of current diagnostic and therapeutic approaches [9].
Nanotechnology is a multidisciplinary field encompassing the integration of principles from chemistry, physics, biology, and medical science [10,11]. It focuses on the creation, development, and application of nanoscale systems, including devices, coatings, food additives, and drug delivery platforms [12]. Increasingly, nanotechnology is being used to design drug delivery systems and imaging techniques, significantly enhancing clinical effectiveness. Its role in drug delivery is anticipated to transform the pharmaceutical and biotechnology sectors in the coming years [13,14]. Nanomedicine, engineered at the molecular scale, employs nanoparticles (NPs) to observe, repair, and regulate biological systems. In drug delivery, critical factors such as NPs uptake, intracellular processing, and accumulation directly influence therapeutic outcomes [15]. In recent years, NPs have gained considerable attention in biomedical research due to their unique features, including functionalized surfaces, nanoscale dimensions, and optimized pharmacokinetic and biodistribution profiles [16,17]. NPs serve as vectors for the delivery of therapeutic agents, including drugs, nucleic acids, and proteins, playing a crucial role in the prevention, diagnosis, and management of diseases [18].
Despite their advantages, NPs face challenges such as limited targeting ability, low biological stability, and accelerated elimination mediated by the immune system. To address these challenges, biomimetic NPs encapsulated with cell membranes have been proposed as a potential solution. Functioning as the fundamental biological unit, cells execute diverse physiological processes, most notably their sophisticated capacity for dynamic environmental sensing and bidirectional molecular communication. Rather than endeavoring to synthetically replicate these complex functionalities, contemporary research has shifted toward direct utilization of native cell membranes to endow nanoparticles (NPs) with evolutionarily optimized biointerfacing properties [19]. These cell membrane-coated NPs leverage the intricate and adaptable nature of cellular membranes, which have evolved to perform specific biological functions, particularly in terms of interacting with biological systems [[20], [21], [22]]. By retaining the antigenic properties of the source cells, these biomimetic NPs can target specific cells and injury sites while evading clearance by monocytes and macrophages [23,24]. Cell membrane-coated NPs not only enhance drug loading, protection, and controlled release, but also improve navigation through internal and external environments [25,26]. This increases precision, improves targeting, and minimizes side effects [27,28]. Advances in cell membrane-coated NPs technology have shown significant potential for treating CVDs by providing enhanced targeting precision, high specificity, and reduced side effects at pathological sites. With the ongoing advancements, various cell membrane types, including those derived from red blood cells (RBCs) [29,30], platelets [31,32], neutrophils [33,34], and macrophages [35,36], have been utilized to extend systemic circulation and facilitate targeted therapeutic delivery to sites of cardiovascular inflammatory injury. More recently, hybrid cell membranes derived from these natural cell types have been also utilized as coatings on diverse NPs to further enhance therapeutic efficacy [37]. Compared with the single cell membrane coating technology, the hybrid cell membrane coating strategy improves NP biocompatibility, targeting efficiency, and immune evasion, ultimately enhancing the therapeutic outcomes in nanomedicine applications. This innovative strategy holds significant promise for addressing the challenges associated with CVDs treatment.
In this review, we aim to explore recent advancements in the hybrid cell membrane-coated NPs (HM/NPs) and their applications for the treatment of CVDs (Fig. 1). We first trace the evolution of cell membrane coating bionanotechnology, from its initial applications to the recent advancements in hybrid cell membrane applications. We then provide a comprehensive description of the preparation process for HM/NPs, detailing the extraction of cell membranes and their subsequent coating onto NP cores. Additionally, we discuss the various types of NPs cores, with a particular emphasis on polymer-based cores, employed in HM/NPs for the treatment of CVDs. The methods used to verify the successful construction of HM/NPs in vitro are also elaborated. Furthermore, we assess the application potential of HM/NPs in CVDs treatment, offering insights into multiple methodologies designed to enhance the functional capabilities of hybrid membrane-coated nanocarriers by integrating functionalities from multiple cell types, including erythrocyte-platelet, monocyte-exosome, exosome-platelet, macrophage-platelet, macrophage-HEK293T, and macrophage-neutrophil hybrid membranes. Finally, we propose strategies to address key obstacles as this technology advances from laboratory research to clinical practice. With continued technological advancements, the therapeutic efficacy of HM/NPs for CVDs is anticipated to improve in near future. By integrating membranes from various cell sources into a single biomimetic platform, HM/NPs can incorporate multiple functions within one system, enhancing their potential for precise diagnosis and treatment in cardiovascular applications.
Fig. 1.
Hybrid cell membrane-coated nanoparticles (HM/NPs) engineered for the treatment of cardiovascular diseases (CVDs). Hybrid cell membranes derived from various natural cell types were applied as coatings on diverse NPs to enhance therapeutic efficacy in nanomedicine applications. This strategy improves NP biocompatibility, targeting efficiency, and immune evasion, ultimately enhancing the treatment of CVDs.
2. The evolution of HM/NPs
Before introducing HM/NPs for the treatment of CVDs, it is essential to thoroughly understand their developmental history and underlying advancements (Fig. 2). The hybrid cell membrane coating technique evolved from the foundational principles of single cell membrane coating technology. In 2011, Zhang's group pioneered the encapsulation of non-toxic poly (lactic-co-glycolic acid) (PLGA) NPs using membranes derived from RBCs, extending the circulation time of NPs in vivo [38]. In 2016, Hu et al. developed a core-shell nanocarrier system by encapsulating platelet membranes onto the surface of polymeric NPs (designated as PM-NP) [21]. The specific molecular interaction between P-selectin expressed on the platelet membrane and CD44, which is overexpressed on tumor cells, facilitated the targeted delivery of the nanocarrier to tumor sites. This approach enabled the precise localization and controlled release of bortezomib at the myeloma site, making a significant advancement in targeted cancer therapy. The integration of chimeric antigen receptor T-cell (CAR-T) therapy, known for its high specificity against tumors, with cell membrane coating technology, which enhances drug delivery, has shown great promise for targeted cancer treatment. In 2020, Ma et al. developed CAR-T membrane-coated NPs for the highly specific treatment of hepatocellular carcinoma [39]. From 2021 to 2024, advancements in nanomedicine have led to the use of various cell types, including platelets [40,41], exosomes [42], and macrophages [35], for NP coating. These developments in single-cell membrane coating technology paved the way for innovative strategies in disease treatment, offering new potential for addressing CVDs through HM/NPs.
Fig. 2.
Historical progression and key technological milestones in the development of HM/NPs for CVD therapy. The figure highlights representative studies that illustrate the evolving membrane strategies and functional innovations contributing to the current state of HM/NPs in CVD treatment.
Similar to single cell membrane coating techniques, hybrid cell membrane coating technologies are continuously evolving and advancing. Attributable to the expression of the CD47 surface protein and the characteristic biconcave disc shape, while the erythrocyte membrane coatings effectively extend the circulation time of NPs in the bloodstream, they do not provide targeted delivery capabilities. To overcome this limitation, in 2017, Zhang et al. advanced the concept by incorporating platelet membranes via a hybrid cell membrane coating technique [43]. This hybrid-membrane-coating approach not only enhanced the targeting ability of the NPs but also conferred immune evasion and target specificity, ultimately significantly improving the therapeutic efficacy in disease treatment. In 2020, another research team leveraged the homing ability of monocytes to injured myocardium to enhance the targeted delivery of stem cell-derived extracellular vesicles, thereby improving the therapeutic efficacy of NPs for cardiac repair [44]. By 2021, Hu et al. combined stem cell-derived exosomes with platelet membranes to further improve their capability to target damaged cardiac tissue and minimize macrophage-mediated uptake [45]. In 2023, Zhou et al. employed macrophage-platelet hybrid membranes to achieve reversible camouflage of nanocomplexes, facilitating efficient siRNA delivery to cardiomyocytes, suppressing the Hippo signaling pathway, and facilitating cardiomyocyte regeneration [46]. In 2024, four independent research teams employed HM/NPs for the therapeutic management of CVDs, such as atherosclerosis, MI/RI, and heart failure, achieving promising and favorable outcomes across all studies [[47], [48], [49], [50]]. In the subsequent year, a biomimetic delivery system featuring an outer hybrid membrane derived from macrophages and activated neutrophils was developed for targeted therapy of acute MI [51]. These advancements in HM/NPs technology highlight its growing potential to address critical challenges in cardiovascular therapeutics, offering novel solutions for improved treatment efficacy.
Compared to single cell membrane coating technology, hybrid cell membrane coating technology offers several notable advantages [52]. (1) Enhanced biological characteristics. By fusing membranes derived from two distinct cell types, hybrid cell membrane coatings exhibit the biological characteristics of both source cells. This duality enables multiple functionalities and expands the range of diverse applications. For instance, hybrid membranes that express CD47 protein and self-recognition molecules from both RBCs and cancer cells can offer immune evasion, prolonged circulation, and homologous targeting, all of which are key advantages over single-cell membranes derived from RBCs or tumor cells alone. (2) Customizable platforms. Both single-cell membrane and hybrid membrane coating technologies allow for genetic manipulation or chemical modification of biomolecules within the encapsulated cell membrane or its surface components. This enables the creation of customizable nanovesicles tailored with precisely defined substances. Hybrid cell membrane coating technology, however, offers greater flexibility in incorporating a broader range of functionalities, achieved through advanced artificial interventions. This versatility enhances the potential to meet specific therapeutic goals in complex in vivo environments, making it particularly valuable for targeted treatments. By combining the strengths of different cellular sources, HM/NPs can leverage the unique advantages of each cell type, making them powerful tools for a wide range of therapeutic applications.
3. Fabrication process of HM/NPs
3.1. Isolation and purification of cellular membranes
Initially, sufficient quantities of cells are collected, with the primary sources of cells including: (1) blood-derived cells, such as erythrocytes, leukocytes, and platelets, which can be efficiently isolated from whole blood using standard blood collection methods and processing equipment [53]; (2) cultured cell lines or strains, commonly propagated in laboratory settings, which are often adequate for supporting preclinical studies [54]. Suspension cells can be expanded in bulk using shake or spin flasks [55], whereas adherent cells necessitate detachment through enzymatic digestion or physical methods for harvesting.
Subsequent processing methods depend on the cell type. For anucleate cells, repeated freeze-thaw cycles in a hypotonic lysis buffer are employed to achieve cellular disruption. The hypotonic lysate buffer was prepared by supplementing 10 mM Tris-HCl buffer (pH 8.0) with 1 mM KCl, 1.5 mM MgCl2, and 1 mM phenylmethanesulfonyl fluoride. The membranes are then isolated and purified from the lysate using discontinuous gradient centrifugation. Differential centrifugation was performed to isolate cellular components. Briefly, the cell suspension was initially centrifuged at 900×g for 5 min at 4 °C, followed by three washes with ice-cold phosphate-buffered saline (PBS). Subsequently, the cells were resuspended in 2 mL of pre-chilled hypotonic lysis buffer and incubated on ice for 15 min to facilitate cell lysis. The suspension was then subjected to four to five freeze-thaw cycles to ensure complete disruption. After lysis, the homogenate was centrifuged at 850×g for 10 min at 4 °C to remove intact cells and large debris. The resulting supernatant was further centrifuged at 15,000×g for 30 min to pellet crude membrane fractions [48]. For eukaryotic cells, sonication and homogenization are also commonly applied to disrupt the lysed cell suspension, followed by differential or gradient centrifugation to separate and purify plasma membranes from the resulting homogenate.
3.2. Fabrication of hybrid cell membranes
Following the isolation and extraction of cell membranes, hybrid cell membrane-derived vesicles were prepared using the extrusion technique. Complementary to the extrusion method, ultrasound was also employed to facilitate vesicle formation [56]. Initially, membrane components were disassembled into heterogeneous cell membrane-derived vesicles using an ultrasonic water bath. The vesicles were subsequently subjected to repeated extrusion through a nanoscale polycarbonate membrane, resulting in uniformly-sized membrane vesicles with precise dimensions (Fig. 3).
Fig. 3.
Preparation of HM/NPs. The membrane materials were derived from two distinct cell types and subsequently coated onto synthetic nanomaterial cores through extrusion and sonication techniques. The resulting hybrid cell membrane-coated nanoparticles (CM-NPs) exhibit a well-defined core-shell architecture, where the surface-anchored cellular membranes confer biofunctional properties that recapitulate the intrinsic biomimetic characteristics of the source cells.
3.3. Hybrid cell membrane coating methods
Membrane-coated NPs can be fabricated using three primary techniques: physical extrusion, ultrasonication, and electroporation [57]. Below is a concise overview of each method (see Table 1).
Table 1.
A summary analysis of hybrid cell membrane coating methods.
| Techniques | Advantages | Disadvantages | Typical size range |
|---|---|---|---|
| Physical Extrusion | High coating efficiency and uniformity; Preserves membrane protein orientation; Scalable for industrial production | Complicated operation; Expensive film extruder | 50–200 nm |
| Ultrasonication | Easy operation; Minimal material loss | Potential protein denaturation; Difficult to control membrane fragment size | 80–250 nm |
| Electroporation | Gentle membrane fusion process; High encapsulation efficiency; Suitable for sensitive biomolecules | Lower throughput compared to extrusion; Potential nanoparticle aggregation | 30–150 nm |
3.3.1. Physical extrusion
Co-extrusion involves the use of an extruder equipped with an auxiliary system to simultaneously process two distinct materials, producing a single composite product where one material envelops the other. This technique is extensively employed in the preparation of synthetic liposomes, where mechanical forces disrupt the membrane integrity, facilitating its reassembly around the NPs core. Early approaches relied on co-extruding vesicles derived from cell membranes with synthetic NPs through porous membranes. Moderate pressure (400–600 psi) achieves optimal vesicle-particle fusion without compromising membrane fluidity.
3.3.2. Ultrasonication
Inspired by advancements in erythrocyte membrane-camouflaged NPs, ultrasound-based methodologies have garnered significant attention for fabricating core-shell structures. In this technique, both cell membrane-derived vesicles and synthetic NPs are exposed to ultrasonic forces, inducing their assembly into core-shell structures [58]. The resulting biomimetic NPs exhibit characteristics comparable to those produced via physical extrusion. Moreover, ultrasonication is a straightforward process with minimal material loss. This method also enables the incorporation of functionalities from multiple cell types into a single NP by employing a multicellular membrane fusion strategy [59]. Ultrasonic processing at 4 °C optimally preserved membrane protein structure (verified by spectroscopic and electrophoretic analyses), while temperatures of 25–37 °C improved fusion yields but caused measurable protein conformational changes (15–30 % secondary structure alteration).
3.3.3. Electroporation
Electroporation further enhances the efficiency of nanodrug carrier preparation. By applying an electric field, transient pores are induced in the cell membrane, facilitating the incorporation of biomolecules and enabling the stable NP encapsulation within the membrane structure. This approach is particularly effective for the development of robust cell membrane-coated nanocarriers [60]. Through systematic optimization studies, an electroporation voltage of 150 V with a pulse duration of 10 ms was identified as the optimal parameter set, achieving a coating efficiency exceeding 85 % while maintaining membrane integrity (LDH release <15 %). This balance between coating efficacy and structural preservation was confirmed through quantitative assessments of both NP encapsulation efficiency and biomembrane damage markers.
4. NP cores used in HM/NPs for the treatment of CVDs
The NP cores constitute a critical component of HM/NPs systems. A comprehensive understanding of the physicochemical properties and functional attributes of diverse NP cores is essential for the rational design and optimization of these nanotransport systems. By selecting appropriate core materials, it is possible to tailor the release profile, stability, and targeting capabilities of the NPs, ensuring enhanced performance in specific therapeutic applications, including the treatment of CVDs. This section provides an in-depth analysis of two widely utilized NPs cores, as well as the cores incorporated into HM/NPs, with a focus on their applications in the treatment of CVDs.
4.1. Types of NP cores
The selection of an appropriate NP core is a pivotal factor influencing the therapeutic efficacy of disease treatments. In this section, we provide an overview of polymer-based NPs and inorganic NPs, encompassing their various subclasses and distinctive characteristics.
4.1.1. Polymeric NPs
Polymers are widely regarded as a cornerstone in the field of drug delivery systems, owing to their versatile physicochemical properties and adaptability for targeted therapeutic applications [61]. Polymers can be engineered to achieve precise control over multiple NPs characteristics, making them highly suitable as delivery vehicles due to their inherent biocompatibility and the simplicity of their formulation parameters. The most prevalent forms of polymeric NPs include (i) nanocapsules, characterized by a central cavity enclosed within a polymeric membrane or shell, and (ii) nanospheres, which consist of a solid polymeric matrix. These two primary categories can be further diversified into distinct structural subtypes, such as polymersomes, dendrimers, and micelles, each exhibiting unique morphological and functional properties.
Polymeric NPs have emerged as ideal candidates for drug delivery due to their biodegradable, biomimetic properties and stability during storage. Moreover, their surfaces can be modified to impart enhanced targeting capabilities [62], making them highly promising for the treatment of cancer, CVDs, and other medical conditions. However, potential limitations of polymeric NPs include the risks of particle aggregation and increased toxicity, which could influence their safety and efficacy [63].
4.1.2. Inorganic NPs
Inorganic NPs, including gold, silver, iron oxide, calcium phosphate, and mesoporous silica NPs, can be artificially synthesized and engineered into specific structures and geometries. Due to the intrinsic properties of inorganic materials, these NPs exhibit unique physical and chemical characteristics. For instance, gold NPs possess free electrons on their surface that oscillate at frequencies determined by their size and shape, conferring photothermal properties [64]. Most inorganic NPs have two principal merits. One is the good biocompatibility, as numerous inorganic NPs (e.g., silica, gold, and iron oxide NPs) exhibit inherently low immunogenicity and cytotoxicity, rendering them highly suitable for biomedical applications. The other is their stability. Inorganic NPs generally demonstrate superior chemical and physical stability under physiological conditions, including enhanced resistance to enzymatic degradation, pH variations, and thermal fluctuations, compared to their organic counterparts (e.g., liposomal or polymeric NPs).
The physical and chemical properties of inorganic NPs are closely associated with their shape, dimensions, and composition. Consequently, technologies utilizing inorganic nanomaterials have been developed and applied across diverse fields, ranging from physical chemistry to medical sciences [65]. However, their clinical application is constrained by challenges such as low solubility and toxicity concerns [66]. The application of cell membrane coating technology provides an effective approach to improve their biocompatibility and reduce toxicity.
4.1.3. Lipid-based NPs
Liposomes are well-defined nanoscale vesicles formed through the self-assembly of amphiphilic phospholipid molecules, capable of encapsulating a broad spectrum of therapeutic agents, including small-molecule drugs, nucleic acids (e.g., oligonucleotides and plasmid DNA), and macromolecular proteins [67]. As a prominent category of lipid-based NPs, liposomes exhibit facile preparation via spontaneous self-assembly, exceptional biocompatibility due to their physiological lipid composition, and enhanced bioavailability through tunable pharmacokinetic properties. Moreover, their physicochemical characteristics—such as particle size, surface charge, lamellarity, and membrane fluidity—can be precisely engineered to optimize biodistribution, cellular uptake, and drug release kinetics, making them highly versatile for controlled drug delivery applications [68].
4.2. Polymer cores used in HM/NPs for the treatment of CVDs
Research has shown that the core structures of HM/NPs are primarily composed of polymeric NPs. Polymers are widely acknowledged as a fundamental component in the area of drug delivery systems [61,69,70]. PLGA, in particular, has received FDA approval for application in various drug formulations due to its excellent properties, including controlled and sustained release, minimal toxicity, and compatibility with cells and tissues [71]. It is widely recognized by researchers as a rapid and low-risk option for clinical translation. Despite its widespread application in drug delivery, PLGA presents certain limitations [72,73]. Although it enhances stability and enables the sustained release of macromolecules, its acidic and hydrophobic properties pose challenges for efficiently encapsulating nucleic acids and fragile proteins [74,75].
G0-C14 cationic lipids represent a class of widely utilized polymeric core materials in non-viral gene delivery systems, demonstrating remarkable efficacy in RNA transfection for therapeutic applications. These compounds have been extensively employed in oncology research for tumor-targeted therapy [76,77]. In another ongoing study, we employ G0-C14 as a delivery vector for small interfering RNA (siRNA) to investigate its potential therapeutic effects on MI/RI.
In 2023, Zhou et al. developed a novel siRNA delivery platform using macrophage-platelet hybrid membranes encapsulating NPs. These NPs were engineered with an internal structure comprising membrane-penetrating helical polypeptide (P-Ben) and poly(L-lysine)-cis-aconitic acid (PC), facilitating efficient siRNA translocation. This innovative approach provides a new material for siRNA delivery applications. Notably, the widespread acceptance of PLGA as a preferred and safe material for drug delivery systems may unintentionally hinder the exploration and development of innovative materials. This highlights the need for continuous research and development of novel biomimetic materials to facilitate the efficient and safe delivery of therapeutics, ultimately improving treatment outcomes for CVDs.
5. In vitro validation of HM/NPs
The distinctive structure and surface biomarkers of hybrid cell membranes are critical for extended circulation, immune evasion, and enhanced targeting efficiency [78,79]. Thus, it is crucial to systematically evaluate the size, surface charge, morphological characteristics, and stability of hybrid cell membrane-derived vesicles, as well as synthetic NPs encapsulated in hybrid cell membranes, under in vitro conditions. Table 2 presents an overview of various established techniques used for NP characterization.
Table 2.
Various established techniques employed for NPs characterization.
| Characterization | Techniques | Physical, chemical, and biological properties of NPs |
References | ||||
|---|---|---|---|---|---|---|---|
| Size | Shape | Potential | Stability | Localization | |||
| Morphology and structure | TEM | ✓ | ✓ | Qiu et al. [49] | |||
| SEM | ✓ | ✓ | Wang et al. [81] | ||||
| DLS | ✓ | ✓ | ✓ | Zhou et al. [46] | |||
| Flow cytometry | ✓ | Tan et al. [48] | |||||
| Colocalization | WB | ✓ | Zhang et al. [44] | ||||
| CLSM | ✓ | ✓ | Li et al. [50] | ||||
5.1. Structural characterization
The morphological and structural characteristics of cell membrane-coated NPs were typically examined using transmission electron microscopy (TEM) and scanning electron microscopy (SEM) [80,81]. Compared to the TEM images of bare NPs, HM/NPs displayed a distinct core-shell structure with a consistent outer layer around 5–10 nm thick [82]. To further characterize HM/NPs, dynamic light scattering (DLS) is commonly employed to assess the particle size and surface zeta potential of bare NPs, fused cell membrane-derived vesicles, and HM/NPs [83]. The size distribution of HM/NPs can also be detected by nanoflow cytometry [84].
5.2. Membrane protein assay
Following the confirmation of the core-shell nanostructure of HM/NPs, additional validation is required to ensure the successful transfer of membrane proteins from various cell types onto the outer fusion membranes of the NPs. Initially, either Coomassie brilliant blue staining or Ponceau S was employed to compare the protein profiles among the original cell surface proteins, the fused cell membranes, and the final encapsulated NPs [85,86]. Subsequently, western blot (WB) analysis was conducted to assess the expression of cell membrane-specific proteins [40]. If membrane surface proteins are labeled with fluorescent dyes, their expression can be precisely detected and quantified using confocal laser scanning microscopy (CLSM). Effective retention of NPs at the target site necessitates the interaction between cell surface receptors and NP surface ligands [87]. The binding efficiency is critically influenced by the density of receptors on the target cells and the membrane proteins on the NPs surface [88]. Therefore, during the extraction of cell membrane vesicles, it is imperative to maintain the integrity of cell membrane surface proteins to prevent any degradation or loss of function, ensuring the effectiveness of the subsequent NP-based drug delivery system.
5.3. Comparative analysis of the merits and limitations inherent in each methodological approach
In dynamic light scattering (DLS) measurements for NP size characterization, the presence of residual macroscopic particulates, dust, or heterogeneous aggregates that evade filtration may significantly elevate the photon count rate. This phenomenon induces an artificial decay component in the autocorrelation function, subsequently appearing as an extraneous peak in the hydrodynamic diameter distribution profile. Such artifacts could lead to erroneous interpretation of polydisperse NP populations.
Special caution is warranted when analyzing turbid colloidal suspensions, where prior dilution (typically 1:10 to 1:100 v/v) or microfiltration (e.g., through 0.22 μm membranes) is strongly advised to mitigate multiple scattering effects. Empirical evidence suggests that rigorous removal of micron-scale contaminants via membrane filtration (≤200 nm pore size) enhances the fidelity of correlation function analysis, thereby improving concordance between DLS-derived hydrodynamic diameters and transmission electron microscopy (TEM) measurements [89].
TEM, SEM, and CLSM exhibit distinct advantages and limitations as analytical techniques for NP characterization. TEM, owing to its exceptional spatial resolution (typically <0.2 nm) and high magnification (up to ∼106 × ), enables precise visualization of nanoscale morphological features, crystallographic structures, and particle size distributions. However, its applicability is constrained by stringent operational requirements, including high-vacuum conditions (typically 10−5–10−7 Pa), which preclude direct observation of hydrated or in vivo specimens. Furthermore, TEM sample preparation necessitates complex protocols involving ultramicrotomy (<100 nm thickness) or negative staining, potentially introducing artifacts during specimen fixation and sectioning [80]. SEM enables high-resolution (typically 1–20 nm) topographical imaging of biological specimens, including tissues and cells, facilitated by optimized sample preparation protocols involving critical-point drying, sputter coating, and/or chemical fixation. However, SEM is inherently limited to surface characterization due to its reliance on secondary electron emission from the specimen's outer morphology, precluding subsurface or volumetric structural analysis [90,91].
The comprehensive characterization of hybrid membrane NPs necessitates a synergistic integration of multiple analytical techniques, as each method provides unique but complementary information. While DLS offers rapid hydrodynamic size distribution analysis in native aqueous environments, it must be corroborated with TEM for absolute particle sizing and morphological validation. Similarly, SEM provides crucial topographical information that complements TEM's cross-sectional visualization capabilities. This multidimensional approach is particularly critical for evaluating complex bionanoparticles, where techniques like CLSM can simultaneously verify membrane integrity (through fluorescent labeling) while TEM/SEM confirms structural preservation. Our systematic comparison demonstrates that only through such orthogonal validation can one reliably distinguish between true NP characteristics and measurement artifacts - for instance, using TEM to identify the particulate contaminants that may cause DLS artifacts, or combining SEM surface imaging with TEM cross-sections to fully reconstruct 3D architecture. This integrated strategy has become the gold standard in nanomedicine characterization.
The comprehensive structural characterization of HM/NPs described in Section 5 establishes a critical foundation for understanding their subsequent biological performance. Specifically, the preserved core-shell architecture (validated by TEM/SEM), optimal physicochemical parameters (confirmed by DLS), and intact membrane protein profiles (verified through WB/CLSM) collectively enable two pivotal in vivo functionalities: (1) targeted biodistribution through retained homing receptors, and (2) immune evasion via conserved 'self' markers. These structural-functional relationships are mechanistically interconnected - for instance, the thickness (5–10 nm) and continuity of the hybrid membrane coating directly determine complement evasion efficiency, while the density and orientation of surface proteins govern tissue-specific targeting. The following section systematically examines how these engineered structural features translate into enhanced therapeutic performance in physiological environments.
6. Functionalities of HM/NPs with hybrid cell membrane coatings
The incorporation of natural cell membranes, together with their associated proteins and respective functions, on the surface of engineered particles represents a novel and promising strategy for NP functionalization [92]. Membrane-camouflaged NPs derived from different cellular sources exhibit distinct functional characteristics [93]. For instance, NPs coated with leukocyte or cancer cell membranes demonstrate enhanced targeting capabilities [94,95]. Furthermore, NPs cloaked with membranes from erythrocytes and platelets can enhance immune evasion and facilitate transendothelial transport [41,96]. HM/NPs are composed of synthetic NPs cores cloaked with a layer of naturally derived cell membranes, enabling functional operation within complex biological environments [97]. The selection of specific cell membrane types imparts the resulting biomimetic nanostructures with unique characteristics inherited from the source cells, such as enhanced targeting capabilities, immune evasion, and superior biocompatibility [98,99]. Comprehensively elucidating the therapeutic advantages of HM/NPs is essential for advancing our understanding of their functional mechanisms and guiding the rational optimization of their structural design.
6.1. Improving NP biocompatibility
NPs are increasingly employed in biomedical applications owing to their distinctive physicochemical characteristics, necessitating a comprehensive understanding of their potential impact on the in vivo environment. Beyond assessing the cytotoxicity of nanomaterials, extensive research has been dedicated to elucidating the molecular mechanisms underlying NPs-induced toxicity. Key mechanisms include: (1) direct interaction of NPs with the cellular membrane, potentially leading to membrane disruption or activation of intracellular signaling pathways that result in cellular damage; (2) material solubilization, releasing toxic ions that impair critical enzymatic functions or directly interact with cellular DNA, causing genotoxic effects; and (3) generation of reactive oxygen species (ROS), leading to oxidative stress, which can further damage essential enzymes and genetic material [100]. HM/NPs mitigate potential cytotoxicity associated with NPs by incorporating a natural cell membrane onto the NPs surface, thereby creating a biomimetic interface that prevents direct interaction between the NPs and cells in vivo.
Recently, Chen et al. engineered a biomimetic nanoliposome (USM) designed to deliver targeted self-cascading diastereases and immunomodulators, effectively reprogramming the inflammatory microenvironment in gouty rats [101]. By camouflaging the M2 macrophage-erythrocyte hybrid membrane with biomimetic nanoliposomes (USM[H]L), they engineered innovative biomimetic liposomes exhibiting enhanced immune evasion capabilities and excellent biocompatibility (Fig. 4A). The cytotoxicity of USM[H]L was evaluated using the MTT assay in RAW264.7 cells and human umbilical vein endothelial cells (HUVECs). The results demonstrated that USM[H]L exhibited superior cell viability compared to USML, suggesting that hybrid cell membrane coatings effectively mitigate the cytotoxicity of NPs.
Fig. 4.
Hybrid cell membrane coating on NPs reducing cytotoxicity and enhancing their biocompatibility both in vitro and in vivo. A) Enhancement of cellular viability through hybrid cell membrane coating technology. i) Schematic illustration of the cascade bienzyme system and MTX-loaded biomimetic nanosomes cloaked with M2 macrophage and erythrocyte membranes (USM[H]L). ii, iii) Cellular viability of ii) RAW264.7 cells and iii) HUVECs following 24-h treatment with SPIONs and other URI formulations, respectively. Reproduced with permission [101]. Copyright 2023, Wiley. B) Improvement in survival rates through hybrid cell membrane coating technology. i) Development of Gboxin-encapsulated, ROS-responsive polymeric nanoparticles camouflaged with a cancer cell-mitochondria hybrid membrane (HMNPs@G). ii, iii) Blood biochemistry analysis: Evaluation of plasma ALT and AST levels in mice following a single-dose nanomedicine treatment. iv) Survival curves of mice. Reproduced with permission [102]. Copyright 2023, Springer Nature.
In another case, Zou et al. described the development of a biomimetic nanopharmaceutical (HM-NPs@G), wherein Gboxin-loaded NPs were encapsulated within a hybrid membrane derived from cancer cell-mitochondrial fusion [102]. This hybrid membrane coating conferred HM-NPs@G with several advantageous properties, including enhanced biocompatibility, optimized pharmacokinetic behavior, effective traversal of the blood-brain barrier, and dual-targeting capabilities directed at both tumor cells and mitochondria through homotypic recognition mechanisms (Fig. 3B). The investigators conducted routine hematological analyses to evaluate the biosafety profile of HM-NPs@G. The findings demonstrated that key blood parameters, including serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels, remained within normal physiological ranges following HM-NPs@G treatment. Furthermore, survival analysis demonstrated a marked increase in the lifespan of mice treated with HM-NPs@G compared to the control group.
6.2. Empowering nanocarrier targeting and immune evasion
Nanomedicine, the application of nanotechnology in medical science, particularly in targeted drug delivery, has profoundly transformed both pharmaceutical sciences and clinical medicine [103]. The effective molecular targeting of NP-based drug delivery systems can significantly improve therapeutic specificity while minimizing systemic toxicity. Typically, this is achieved by conjugating ligands to the surface of NPs, which specifically bind to receptors overexpressed on the target cell type [104]. In a research project, Wang et al. successfully isolated neutrophil membrane proteins to construct biomimetic neutrophil-like membranes, subsequently developing a non-viral biomimetic NPs-based delivery system aimed at promoting cardiac regeneration. By leveraging neutrophil-mimetic camouflage, the system effectively emulates the natural recruitment of neutrophils to injured cardiac tissue post- MI, mediated through receptor-ligand interactions between chemokine receptors on the neutrophil membrane and chemokine ligands expressed by the damaged endothelium and myocardium [105]. The antigenic properties conferred by the cell membrane coating surrounding the synthetic core enable these bionic NPs to engage with heterogeneous pathological molecules [106]. For instance, NPs encapsulated within RBC membranes can function as decoys, effectively sequestering and neutralizing toxins derived from the pore-forming proteins of various bacterial strains, thereby safeguarding healthy RBCs from potential cytotoxic effects [107]. Efforts to discover new disease biomarkers and related ligands for targeted drug delivery have been steadily growing.
As a fundamental aspect of nanomedicine, the primary objective of nanodelivery systems is to achieve precise localization of one or more therapeutic agents at the target site [108,109]. Following administration, a significant proportion of NPs fail to reach their intended targets. Particles larger than 6 nm are predominantly sequestered by the liver and spleen, while those smaller than 6 nm are rapidly cleared by the kidneys [110,111]. Notably, 30–99 % of NPs accumulate in the liver upon systemic entry, highlighting a major challenge in achieving targeted delivery [112]. Given that most NPs are administered via the bloodstream, their circulation half-life is a crucial factor in determining therapeutic efficacy. Significant efforts have been dedicated to engineering NPs with extended half-lives to optimize their therapeutic potential [113,114]. RBC membrane, containing the 'self-marker' protein CD47 on its surface, can function as a biomimetic coating to prolong the systemic circulation duration of NPs [115]. CD47 interacts with signal regulatory protein α on phagocytic cells, delivering a 'don't eat me' signal that prevents NP phagocytosis by macrophages [116].
In order to achieve the desired therapeutic effect, NPs must remain in systemic circulation for a sufficient duration, evade clearance by the immune system, traverse biological barriers, accumulate at the target site, and effectively interact with target cells. Particles exhibiting prolonged circulation times and enhanced targeting capabilities to the area of interest should possess a diameter of 100 nm or less, coupled with a hydrophilic surface to minimize clearance by macrophages.
Although cell membrane coating imparts unique functional attributes to NPs, in the context of the organism's intricate and dynamic environment, relying solely on a single cell membrane coating may be insufficient to meet the multifaceted functional demands required for achieving optimal therapeutic outcomes. HM/NPs address this limitation by integrating two distinct cell membranes, thereby endowing NPs with multifunctional properties. Compared with NPs coated with a single cell membrane, hybrid NPs derived from membranes of multiple cell types enable more precise targeting [117]. Biomimetic NPs encapsulated with fused cell membranes can facilitate the crossing of these barriers by providing a functional bio-interface on the surface of the NPs. This modification improves the targeted delivery of therapeutic agents and elevates drug concentration at the site of action [118]. More importantly, the hybridized cell membrane coating technology enables NPs to simultaneously acquire enhanced targeting capabilities and immune evasion mechanisms, further improving their effectiveness in therapeutic applications [106,119].
In the study by Jiang et al., hybrid membranes composed of RBC membranes and cancer cell membranes were utilized to enhance the efficacy of photothermal therapy for tumors (Fig. 5A). The researchers fused RBC membranes with MCF-7 cancer cell membranes to construct a RBC-cancer cell (RBC-M) hybrid membrane-coated melanin NP platform (Melanin@RBC-M) [96]. The fused RBC-M hybrid membrane vesicles preserved the characteristic membrane proteins of both RBCs and MCF-7 cells, enabling the resulting Melanin@RBC-M NPs to achieve both prolonged systemic circulation and homotypic targeting of the source MCF-7 cells. Furthermore, the researchers demonstrated that increasing the proportion of the MCF-7 cell membrane component in the hybrid membrane significantly enhanced the homotypic targeting capability of Melanin@RBC-M. Conversely, increasing the erythrocyte membrane component effectively reduced macrophage-mediated uptake of the NPs, thereby extending their circulation time in the bloodstream. These findings underscore the critical role of cell membrane surface proteins in determining the biological performance of hybrid membrane-coated nanoplatforms.
Fig. 5.
Hybrid cell membrane coating enhancing nanoparticle targeting and immune evasion. A) Diagrammatic representation of erythrocyte-cancer hybrid membrane-coated melanin nanoparticles for improved photothermal therapy efficacy in tumor treatment. i) Fabrication procedure of melanin nanoparticles cloaked with RBC-cancer hybrid membranes (Melanin@RBC-M). ii) Melanin@RBC-M nanoparticles, featuring prolonged circulation and homotypic targeting properties, enhanced the efficacy of photothermal tumor therapy. Reproduced with permission [96]. Copyright 2019, Elsevier. B) Graphical representation of biomimetic nanoparticles cloaked with hybrid cell membranes, engineered for noninvasive targeted therapy of laser-induced choroidal neovascularization. i) Fabrication of [RBC-REC]NPs via encapsulation of polymeric cores with hybrid RBC-REC membranes. ii) TEM images of [RBC-REC]NPs. Scale bar, 50 nm. iii) Fluorescent imaging of macrophage uptake of DiD-labeled different treatment groups (RBCNPs, RECNPs, or [RBC-REC]NPs). Nanoparticles are shown in red. Scale bar: 100 μm. iv) Representative fluorescent images of isolated eyes 24 h post-intravenous administration of different treatment groups. Reproduced with permission [106]. Copyright 2021, American Chemical Society. (For interpretation of the references to colour in this figure legend, the reader is referred to the Web version of this article.)
In a separate study aimed at developing nanocarriers, a novel anti-angiogenic agent was engineered using HM/NPs for the non-invasive targeted treatment of choroidal neovascularization [106]. This platform leveraged the immune evasion properties of erythrocyte membranes and the homotypic targeting capability of retinal endothelial cell membranes. Specifically, a polymeric NP core was coated with a hybrid membrane comprising RBC and retinal endothelial cell membranes ([RBC-REC]NPs), effectively combining these functional attributes of both cell types to enhance therapeutic efficacy (Fig. 5B). Under TEM, the [RBC-REC]NPs exhibited a characteristic core-shell architecture. To evaluate the phagocytosis evasion capability of [RBC-REC]NPs, the researchers incubated the synthesized NPs with mouse RAW264.7 macrophage cells. Fluorescence analysis revealed that RAW264.7 cells exhibited a weak fluorescence signal following treatment with [RBC-REC]NPs, while a significantly stronger fluorescence signal was observed after incubation with RECNPs. This finding highlights that the phagocytosis of [RBC-REC]NPs by macrophages was markedly reduced compared to RECNPs, primarily attributed to the presence of fused RBC membranes on the NP surface. To further investigate the active targeting capability of [RBC-REC]NPs in vivo, the researchers employed a classical laser-induced choroidal neovascularization model in mice, mimicking wet age-related macular degeneration [120,121]. The results demonstrated that the strongest fluorescence signals were localized in the eyes of mice treated with [RBC-REC]NPs, indicating the superior active targeting properties of [RBC-REC]NPs than those of either RBCNPs or RECNPs.
6.3. Mechanisms underlying the functionalities of HM/NPs
The therapeutic efficacy of HM/NPs is primarily attributed to their sophisticated biointerfacial properties, which synergistically confer both immune ev asion and active targeting functionalities. Recent mechanistic studies have revealed that the preserved transmembrane proteins (e.g., CD47, CD24, and programmed death-ligand 1 [PD-L1]) on HM/NPs play a pivotal role in immune modulation by engaging with specific immune checkpoint pathways. Specifically, CD47 interacts with signal regulatory protein α (SIRPα) on phagocytic cells, while PD-L1 binds to programmed death-1 (PD-1) on T cells, collectively transmitting potent "don't-eat-me" signals that effectively inhibit phagocytosis by macrophages and dendritic cells (DCs) [122]. This biomimetic strategy results in a significant reduction in opsonization and subsequent clearance by the mononuclear phagocyte system (MPS), thereby prolonging systemic circulation time.
From a targeting perspective, the hybrid membrane architecture exhibits remarkable tissue-specific homing capabilities through multiple ligand-receptor recognition mechanisms. In a recent study, Lu et al. engineered a novel nanocarrier system leveraging the specific ligand-receptor interaction between the receptor for advanced glycation end products (RAGE) and S100A9. By inducing RAGE overexpression on the cell membrane surface, this system achieves targeted delivery to damaged myocardial tissue under inflammatory conditions. Furthermore, it enhances binding affinity to circulating S100A9, thereby suppressing its pro-inflammatory activity [35]. Furthermore, multiple ligand-receptor recognition mechanisms contribute to targeted cellular responses. For instance, the CXCL12/CXCR4 chemokine axis facilitates tumor cell chemotaxis via hypoxia-inducible factor-1α (HIF-1α)-dependent signaling, whereas the CCL2/CCR2 interaction drives precise monocyte recruitment and subsequent aggregation at inflammatory sites [123,124].
Current research further demonstrates that the membrane protein composition can be precisely engineered through cell source selection and membrane fusion techniques. These findings establish HM/NPs as a versatile platform combining natural biological functions with synthetic NP advantages, representing a paradigm shift in targeted nanomedicine development.
6.4. In vivo efficacy and safety of HM/NPs
Recent studies demonstrate promising in vivo performance of HM/NPs across disease models. In myocardial ischemia-reperfusion injury rats, all cell membrane-coated nanocomplexes exhibited significantly prolonged circulation half-life (t1/2) compared to uncoated counterparts, with macrophage-platelet hybrid membrane NPs demonstrating the most pronounced extension (27.9 h vs 7.2 h for bare nanocomplexes, p < 0.001) [46]. Quantitative histomorphometric analysis demonstrated that macrophage-platelet hybrid membrane nanocomposites significantly reduced fibrotic area (15.4) compared to bare nanocomposites (51.3, p < 0.001). Consistent with these findings, TTC staining revealed a marked reduction in infarct area (16.4) in IR-injured rat hearts treated with hybrid nanocomposites, versus PBS (63.3) and bare nanocomposite (46.7 ± 3.1 %) controls (p < 0.001 for both comparisons). Atherosclerotic mice exhibited 3.35 times higher plaque targeting with macrophage-platelet hybrid membrane NPs, yielding 12.15 % average plaque area reduction after 8-week treatment [47]. Pharmacokinetic studies reveal superior circulation half-life. The cytotoxicity studies (CCK-8 assay) confirm safety at therapeutic doses, with normal serum biomarkers and minimal immunogenicity. These findings collectively support the translational potential of HM/NPs while highlighting the critical influence of membrane composition ratios on both efficacy and safety profiles.
7. Application of HM/NPs coated with multiple hybrid membranes for the treatment of CVDs
The distinct characteristics of various cell types are largely attributed to the diverse range of antigens present in their membranes. Identifying specific membrane components has enabled researchers to improve synthetic platforms by incorporating biomimetic properties for targeted applications, including the treatment of CVDs [125]. We subsequently explore the unique characteristics of cell membranes that facilitate the formation of fusion membranes, as well as the applications of HM/NPs in the treatment of CVDs (Table 3). As presented in Table 4, a systematic comparison of various hybrid membrane compositions was also conducted.
Table 3.
Application of different HM/NPs in the treatment of CVDs.
| Source cells | Hybrid membrane extraction | Core NPs | Functions | Applications | References |
|---|---|---|---|---|---|
Erythrocyte-platelet
|
Sonication followed by extrusion | JQ1-loaded PLGA NPs; PLGA polymeric cores | Targeted drug delivery; Avoiding immune clearance | Accmulating in cardiac myofibroblasts; Enhancing circulation ability | Zhang et al. [43] Li et al. [50] |
Stem cell exosome-platelet
|
Repeated extrusion | Proteins, nucleic acids, etc. in exosome | Targeted the injured heart; Enhancing the cellular uptake of exosomes | Reducing myocardial infarct size | Hu et al. [45] |
Macrophage-platelet
|
Repetitive freeze-thaw followed by extrusion | siRNA loaded polymer (BSPC) | Atherosclerosis targeted delivery; Suppressing atherosclerosis and myocardial apoptosis | Anti-atherosclerotic therapy; Reducing plaque area; Promoting myocardium regeneration; Recovering cardiac functions | Xie et al. [47] Zhou et al. [46] |
Macrophage-HEK293T
|
Sonication followed by extrusion | Macrophage membranes with MerTK overexpression and 293T cells membranes with TfR overexpression |
Aorta targeting; Efferocytosis repairs | Attenuating atherosclerosis; Decreased inflammation | Qiu et al. [49] |
Neutrophil-endothelial cell
|
Hypotonic lysis followed by extrusion and sonication | Roflumilast-loaded PLGA NPs | Targeted binding; Homing to injured tissue | Suppressing inflammatory; Targeted drug delivery | Tan et al. [48] |
Monocyte-exosome
|
Incubation followed by extrusion | Proteins, nucleic acids, etc. in exosome | Targeted drug delivery | Reducing fibrosis area; Improving cardiac function | Zhang et al. [44] |
Macrophage- Neutrophil
|
Incubation followed by extrusion | 4-Octyl itaconate-loaded liposomes | Targeted ischemic myocardium; Reducing the infarct size | Resolving inflammation; Suppressing myocardial fibrosis | Zheng et al. [51] |
Table 4.
Comparative analysis of various hybrid membranes.
| Source cells | Targeting efficiency | Circulation time | Therapeutic outcomes (p-value reflects the hybrid membrane vs. PBS group comparison) | References |
|---|---|---|---|---|
| Erythrocyte-platelet | Accumulation in heart (P < 0.01, compared with PBS group) | Longer circulation time (Compared with platelet membrane) | Fibrotic area reduction (P < 0.001) | Li et al. [50] |
| Stem cell exosome-platelet | Accumulation in heart (P < 0.05, compared with exosome group) | Longer circulation time (Compared with unmodified Exos) | Myocardial infarct size reduction (P < 0.001) | Hu et al. [45] |
| Macrophage-platelet | Accumulation in heart (P < 0.05, compared with platelet membrane) | Longer circulation time (Compared with unmodified BSPC) | Reducing plaque area (p < 0.001) | Zhou et al. [46] |
| Macrophage-HEK293T | Accumulation in aorta root (P < 0.01, compared with unmodified HMNVs) | Longer circulation time (Compared with HEK293T membrane) | Reducing plaque area (p < 0.01) | Qiu et al. [49] |
| Neutrophil-endothelial cell | Accumulation in heart (P < 0.0001, compared with PBS group) | Longer circulation time (Compared with endothelial cell membrane) | Myocardial infarct size reduction (P < 0.001) | Tan et al. [48] |
| Monocyte-exosome | Accumulation in heart (P < 0.001) | Longer circulation time (Compared with unmodified Exos) | Reducing fibrosis area (p < 0.001) | Zhang et al. [44] |
| Macrophage- Neutrophil | Accumulation in heart (P < 0.05, compared with unmodified M4oRL)) | Longer circulation time (Compared with neutrophil membrane) | GSDMD reduction (p < 0.05) | Zheng et al. [51] |
7.1. Erythrocyte-platelet hybrid membranes
Various types of cell membranes can serve as carriers to encapsulate NPs. Notably, extending the cell membrane-coated platform to other cell types has allowed for the creation of drug carriers with natural targeting capabilities [126]. The first platform of this kind used RBCs as the source material to extend blood circulation time. The resulting RBC membrane-coated NPs (RBCNPs) exhibited significantly longer half-lives compared to PEG-stabilized NPs [38]. While cell membrane coating effectively enhances NP functionality, adding extra features is often necessary for specific applications. Although RBCNPs can circulate for extended durations, incorporating targeting ligands significantly improves their localization to the desired target site. The platelet membrane was selected as the optimal coating material due to its high content of functional proteins, which enable precise targeting of collagen and collagen-secreting activated fibroblasts [127]. Compared to single-cell membranes derived from erythrocytes or platelets, fused membranes from erythrocytes and platelets that co-express CD47 membrane proteins and glycoproteins exhibit enhanced characteristics such as immune evasion, prolonged circulation time, and targeted binding to myocardial fibroblasts and collagen [128].
In a study conducted by Zhang et al., a biomimetic nanoplatform, denoted as [RBC-P]NPs, was developed [43]. These HM/NPs coated with RBC-platelet hybrid membranes could enhance atherosclerosis-targeting specificity and prolong circulation time in peripheral blood (Fig. 6A). To evaluate the in vivo targeting capability, a murine model of atherosclerosis with platelet interaction was established. Fluorescence imaging of the aorta following intravenous NP administration revealed no detectable fluorescence in the group receiving RBCNPs, while the group treated with [RBC-P]NPs exhibited visible fluorescence. Particularly, a higher fluorescence intensity in the PNPs compared to [RBC-P]NPs was observed, suggesting a potential loss of platelet surface membrane proteins during the preparation of [RBC-P]NPs.
Fig. 6.
Representative HM/NPs with macrophage-platelet hybrid membranes for CVDs treatments. A) The preparation of nanoparticles coated with a hybrid membrane of RBCs and platelets (referred to as [RBC-P]NPs). i) Diagram of the preparation process. RBC and platelet-derived membrane material is combined, and the fused membrane is employed for coating polymeric cores, forming [RBC-P]NPs. ii) Visualization of aortas from ApoE knockout mice on a high-fat diet following intravenous injection of dye-labeled RBCNPs, [RBC-P]NPs, and PNPs (red = nanoparticles; scale bars = 1 mm). Reproduced with permission [43]. Copyright 2017, Wiley. B) Biomimetic nanoparticles coassembled from platelet and erythrocyte membranes for the treatment of heart failure. i) Schematic for preparation of PM&EM/JQ1 NPs and their application in JQ1 delivery to myofibroblasts for heart failure therapy. ii) TEM images of JQ1 NPs (Top) and PM&EM/JQ1 NPs (Bottom). iii, iv) CLSM images of PM&EM/JQ1 NPs. The upper panel presents a physical mixture of PLGA coated with both membranes, along with DiI fluorescence serving as a control. The lower panel displays fluorescence images of the assembly following sonication-induced mixing. v) Representative cross-sectional images of remote left ventricular (LV) tissue stained with Masson's trichrome. Reproduced with permission [50]. Copyright 2024, American Chemical Society. (For interpretation of the references to colour in this figure legend, the reader is referred to the Web version of this article.)
In another study, Li et al. prepared a platelet-erythrocyte hybrid membrane nanotherapeutic system (PM&EM NPs) that utilized platelet membranes to confer PM&EM NPs with the ability to target myocardial fibroblasts and collagen, while the involvement of erythrocyte membranes enhanced the long-term cycling ability of the formulated NPs [50]. As illustrated in Fig. 6B, PLGA was utilized to construct the core structure of NPs. The CD47 protein present on the surface of the erythrocyte membrane exerted an immunomodulatory function, preventing phagocytosis by the mononuclear phagocyte system (MPS) and thereby extending the circulation time of PM&EM/JQ1 NPs in the bloodstream. TEM revealed the presence of a spherical core-shell structure, with the PLGA core accurately encapsulated by a fused cell membrane layer. Under CLSM, minimal colocalization was observed among the three raw materials when physically mixed. In contrast, substantial overlap was evident upon assembly through ultrasonic mixing, indicating effective material integration. Masson's trichrome staining demonstrated that all three treatment modalities (free JQ1, JQ1 NPs, and PM&EM/JQ1 NPs) significantly attenuated myocardial fibrosis induced byMI, with PM&EM/JQ1 NPs exhibiting a superior therapeutic effect than other groups.
The initial use of cell membrane coating technology mainly focuses on RBCs to prolong NP circulation time, highlighting its potential for enhancing drug delivery systems. Expanding this approach to other cell types has opened avenues for developing natural targeting drug carriers, such as platelets for site-specific delivery to atherosclerotic plaques or fibrotic myocardial tissue. The concept of HM/NPs, which combines functionalities from multiple cell types within a single membrane-coated nanocarrier, further underscores the versatility and promise of this strategy, though its practical implementation and therapeutic efficacy require further investigation.
7.2. Monocyte-exosome hybrid membranes
Monocytes, although present in relatively low concentrations within the bloodstream, serve a critical function in host defense by mediating immune responses and providing protection against pathogenic infections [57]. Leukocyte infiltration in the ischemic region serves as a key indicator of the acute inflammatory response following MI/RI, with monocytes constituting the predominant cell population. Large numbers of inflammatory monocytes are recruited to the damaged heart within 30 min post-infarction [129]. The migration of these monocytes is regulated by adhesion molecules such as P-selectin glycoprotein ligand-1, macrophage receptor-1, lymphocyte function-associated antigen-1, and very late antigen-4 [130].
Exosomes are characterized by a lipid bilayer and a distinct profile of oligonucleotides and proteins that facilitate intercellular communication, transmitting a range of cytoprotective, immunomodulatory, and pro-angiogenic signals [131]. Exosomes derived from stem cells can enhance myocardial repair processes post- (MI) [132,133].
For example, Zhang et al. developed a monocyte-exosome hybrid membrane-based nanotherapeutic system (Mon-Exos) aimed at targeting ischemic myocardium [44]. This system leverages the post-MI/RI recruitment capabilities of monocytes and the cardiac regenerative properties of exosomes to achieve a therapeutic effect (Fig. 7). The protein profile analysis of Mon-Exos confirmed the presence of characteristic exosomal marker proteins as well as the retention of typical molecular markers specific to monocytes. Fusion was facilitated using DiO-labeled monocyte membranes and DiD-labeled exosomes through the incubation-extrusion method and physical mixing method, respectively. Confocal microscopy of Mon-Exos obtained via the incubation-extrusion approach demonstrated co-localized hybrid membranes, indicated by the merged yellow fluorescence. In contrast, the physical mixture of monocyte membranes and exosomes displayed distinctly separated green and red fluorescence signals. TEM images revealed that Mon-Exos retained a spherical morphology post-fusion. Mon-Exos significantly reduced myocardial fibrosis and exerted cardioprotective effects. The findings indicated that monocyte-mimetic modification enhanced exosome recruitment, thereby improving their therapeutic efficacy in target organs.
Fig. 7.
Representative HM/NPs with monocyte-exosome hybrid membranes for CVD treatments. A) Schematic illustration of the synthesis process for the monocyte-exosome fused membrane (Mon-Exos). B) Characterization of Mon-Exos. i) WB analysis of specific marker proteins in Exo, Mon, and Mon-Exo. ii) CLSM images showing Mon-Exos (left) compared to a physical blend of exosomes and monocyte membranes (right). iii) TEM image of Mon-Exo. C) Reduced myocardial fibrosis area by Mon-Exos. i-iii) Following Masson staining, fibrotic tissue was identified (i), and subsequent quantitative analysis revealed a reduction in fibrotic remodeling (ii), while left ventricular anterior wall (LVAW) thickness remained unaltered (iii). Reproduced with permission [44]. Copyright 2020, Elsevier.
Although stem cell-derived exosomes have shown significant potential in promoting cardiac repair and regeneration following (MI) [134,135], their clinical application is hindered by challenges such as low homing efficiency and limited production yields [136]. To address these limitations, researchers employed a membrane fusion strategy to enhance exosome delivery by incorporating monocyte-like characteristics. This approach holds promise for improving the therapeutic efficacy of stem cell-derived exosomes and offers a novel tool to support clinicians in optimizing regenerative treatments for ischemic heart disease.
7.3. Exosome-platelet hybrid membranes
The therapeutic potential of exosomes has been extensively investigated across a range of pathological conditions [[137], [138], [139], [140]], including oncological [141] and CVDs [142,143]. Exosomes are generally characterized by low immunogenicity and minimal cytotoxicity [144], making them a promising therapeutic tool. Exosomes derived from mesenchymal stem cells have been extensively investigated as a promising therapeutic intervention for MI due to their paracrine effects, including cardioprotection, angiogenesis promotion, and attenuation of adverse cardiac remodeling [145]. However, their systemic administration is hindered by a significant limitation—the lack of targeted delivery. Recently, platelet-inspired biomimetic and bioengineering strategies have emerged as innovative approaches to address this challenge [[146], [147], [148]].
Hu et al. developed a stem cell platelet hybridized membrane (P-XOs) by encapsulating stem cell-derived exosomes with platelet membranes, thereby enhancing their capacity for targeted delivery to injured tissues [45]. As illustrated in Fig. 8, P-XOs demonstrated superior targeting efficiency toward the injured myocardium in a murine model. Further investigation revealed that both XOs and P-XOs exhibited a propensity to adhere to denuded blood vessels, with P-XOs showing significantly enhanced binding capacity compared to XOs. In terms of therapeutic efficacy, P-XOs demonstrated a superior ability to reduce myocardial infarct size.
Fig. 8.
Representative HM/NPs with exosome-platelet hybrid membranes for CVD treatments. A) A diagram demonstrating the process of preparing P-XOs. B) Typical IVIS images displaying the organ distribution of XOs and P-XOs in a (MI) mouse model. C) Reduction of myocardial infarct size by P-XOs. i) IVIS imaging revealing that DiD-labeled P-XOs specifically adhered to denuded vessels while avoiding sham vessels. ii) Masson staining images illustrating control (PBS-treated animals), XOs, and P-XOs, along with infarct size quantification. Reproduced with permission [45]. Copyright 2021, Elsevier.
The use of exosome injections for therapeutic purposes encounters significant obstacles, including competition from endogenous exosomes and uptake or clearance by the mononuclear phagocyte system. This study explored the fusion of stem cell-derived exosomes with platelet membranes to improve their targeting of damaged myocardium and reduce macrophage-mediated uptake. In vivo experiments revealed that these hybrid exosomes exhibited strong cardiac targeting capabilities in a mouse model of (MI). The findings of Hu et al. provided proof-of-concept evidence and introduced a versatile approach to improve exosome retention and accumulation in injured tissues.
7.4. Macrophage-platelet hybrid membranes
MI, a common manifestation of ischemic heart disease, is characterized by elevated morbidity and mortality worldwide [149,150]. Cardiomyocyte repair to prevent the transition from acute MI to heart failure constitutes a promising therapeutic strategy for MI/RI [151]. Disruption of Hippo signaling has been shown to enhance cellular regeneration and promote cardiac tissue repair following MI/RI [152]. As a pivotal component of the Hippo pathway, Sav1 is a promising target for pathway inhibition. Specifically, RNA interference (RNAi) via small interfering RNA (siRNA) targeting Sav1 (siSav1) offers an effective strategy to suppress Hippo signaling and facilitate myocardial recovery in MI/RI injury [153,154].
Macrophage membranes have been chosen as an optimal coating material due to their rich content of functional proteins, which are critical for mediating inflammatory responses and facilitating cellular recruitment in the context of MI/RI [155]. The platelet membranes, on the other hand, enables specific targeting to thrombotic sites [156,157], and their use as carriers holds considerable potential due to their capability to navigate compromised vasculature and extend systemic circulation time [158].
In one case, Zhou et al. developed an innovative drug delivery system, BSPC@HM NCs, by encapsulating nanocomplexes (NCs) within platelet-macrophage hybrid membranes (HMs) [46]. This BSPC@HM NCs system demonstrated high efficiency in delivering Sav1 siRNA (siSav1) into cardiomyocytes, subsequently inhibiting the Hippo signaling pathway and promoting cardiomyocyte regeneration (Fig. 9A). DLS analysis showed that the average size of BSPC@HM NCs was approximately 199 nm, slightly larger than that of BSPC NCs. This increase in particle size aligned with the expected thickness of the hybrid membrane layer. TEM analysis of BSPC NPs revealed their uncoated structure, while TEM images of BSPC@HM NCs displayed a distinct membrane-like structure surrounding the outer layer. To evaluate whether BSPC@HM NCs retained the microthrombus-targeting and inflammatory homing characteristics of platelets and macrophages [159], respectively, ex vivo fluorescence imaging of ischemia-reperfusion-injured rat hearts was performed 6 h post-intravenous administration of Cy3-labeled NPs. Results indicated that BSPC@HM NCs accumulated at significantly higher levels at the injury site compared to BSPC NCs enveloped solely with platelet or macrophage membranes, highlighting the enhanced targeting efficiency of the HM/NPs.
Fig. 9.
Representative HM/NPs with macrophage-platelet hybrid membranes for CVD treatments. A) Reversibly camouflaged biomimetic nanocomplexes for siRNA delivery in the treatment of MI/RI. i) Schematic representation of biomimetic BSPC@HM nanocomplexes with reversible HM camouflage, designed for targeted myocardial siSav1 delivery and MI/RI management. ii) TEM images of BSPC@HM. iii) Ex vivo visualization and iv) assessment of fluorescence intensity in rat hearts 6 h following intravenous administration of Cy3-siSav1-loaded NCs. Reproduced with permission [46]. Copyright 2023, Wiley. B) Platelet membrane-fused engineered M2 macrophage-derived extracellular vesicles for targeted atherosclerosis therapy. i) Schematic representation of the fabrication process of P-M2EV. ii) Cardiac and aortic tissues of mice were imaged 4 h post-intravenous administration of PBS, DiD-labeled M2EV, or P-M2EV. iii) Quantitative analysis of the mean fluorescence intensity in aortic tissues from the M2EV and P-M2EV groups. iv) Representative images of Oil Red O-stained whole aortas from mice treated with PBS, M2EV, and P-M2EV. v) Proportion of plaque area relative to the total luminal surface area. Reproduced with permission [47]. Copyright 2024, Elsevier. (For interpretation of the references to colour in this figure legend, the reader is referred to the Web version of this article.)
Atherosclerosis is a chronic inflammatory disorder of the vasculature and represents a major contributor to global mortality [160]. Despite considerable advancements in therapeutic strategies over recent years, there remains an ongoing need for novel approaches to enhance treatment efficacy [161,162]. Macrophages are pivotal in the progression of atherosclerosis [163]. M1-type macrophages, on the one hand, secrete various pro-inflammatory cytokines and phagocytic debris, thereby amplifying the inflammatory response. On the other hand, M1 macrophages can transition into the reparative M2 phenotype, which is crucial for inflammation resolution and plays a key role in the regression of atherosclerotic plaques [[164], [165], [166]]. Research indicates that platelets play a role in the pathological development of atherosclerosis by interacting directly with diverse substrates and releasing bioactive molecules [167,168]. Upon recruitment to atherosclerotic plaques, activated platelets facilitate the migration of inflammatory cells to the lesion site via specific membrane surface proteins.
Drawing inspiration from the natural membrane fusion process, scientists in materials and chemistry have recently created artificial membrane-fusion systems, primarily using particles, to influence cell behavior for various bioengineering and medical purposes. To induce the transition of M0 macrophages into the M2 phenotype, RAW 264.7 cells were cultured with interleukin-4 for a period of 24 h [169]. To harness the therapeutic potential of M2 macrophages in combating atherosclerosis and enhance the targeting efficiency of the nano drug delivery system, Xie and colleagues developed a novel platform, namely P-M2EV, by integrating M2 macrophage membranes with platelet membranes (Fig. 9B) [47]. TEM analysis revealed that the M2EV coated with sole M2 macrophage membranes exhibited a distinctly spherical morphology, whereas P-M2EV displayed a well-preserved structural integrity and a more uniform size distribution. To investigate the ability of P-M2EV to target atherosclerotic plaques, an atherosclerotic mouse model was developed. 4 h following tail vein injection of PBS, DiD-labeled M2EV or P-M2EV, fluorescence was detected using an in vivo imaging system (IVIS). The findings revealed that mice treated with P-M2EV showed markedly stronger fluorescent signals at regions of atherosclerotic plaque formation compared to those injected with M2EV. The therapeutic effects of the nanocarriers were subsequently evaluated in mice fed a high-fat diet for 12 weeks, followed by weekly tail vein injections of PBS, M2EV, or P-M2EV. After 8 weeks of treatment, the mice were euthanized, and the aortas were harvested for analysis. Oil Red O staining revealed a reduction in plaque area in both the M2EV and P-M2EV groups, with P-M2EV demonstrating superior anti-atherosclerotic efficacy, as evidenced by a markedly smaller plaque area.
The primary challenges in utilizing siRNA for the treatment of MI/RI are achieving myocardial-specific accumulation and overcoming low transfection efficiency. The hybrid membrane camouflage technology enables the efficient delivery of nanocomposites into cardiomyocytes, thereby promoting cardiac cell regeneration and highlighting the potential of gene therapy for cardiac injury. Overall, this bioinspired strategy overcomes multiple delivery barriers and offers a new direction for myocardial regenerative medicine. The hybrid cell membrane coating technology, which utilizes platelet membrane-modified extracellular vesicles derived from M2 macrophages, mimics the interactions between platelets and macrophages. This approach provides an innovative solution for the clinical treatment of atherosclerosis and shows considerable translational potential. By leveraging cell membrane modification techniques to enhance the targeting specificity and stability of biotherapeutics, this strategy underscores the critical role of biomaterials in gene and cell therapies. Future research should focus on optimizing the large-scale production of delivery systems and conducting rigorous biosafety evaluations to accelerate clinical translation.
7.5. Macrophage-HEK293T hybrid membranes
Macrophages recognize, interact with, and internalize apoptotic cells through diverse mechanisms [170]. Among these, Mer tyrosine kinase (MerTK) plays a pivotal role in the phagocytic clearance of apoptotic cells [[171], [172], [173]]. NPs modified with macrophage-derived membranes inherit the intrinsic targeting and homing capabilities of macrophages [174,175]. Furthermore, through advanced genetic engineering techniques, specific surface markers can be introduced onto the cell membrane-coated NPs, enabling precise targeting and functional modulation of these biomimetic NPs [176].
In the study conducted by Qiu et al., the cell membranes derived from RAW264.7 cells overexpressing MerTK and HEK293T cells overexpressing the transferrin receptor (TfR) were combined with DOPE polymers to fabricate innovative nanocarrier systems, referred to as HMNVsM-D (Fig. 10) [49]. To evaluate the systemic distribution of HMNVsM-D, ApoE−/− mice were administered HMNVsM-D via tail vein injection. Using an in vivo imaging system (IVIS), it was observed that the application of a magnetic field (MF) significantly enhanced the accumulation of HMNVsM-D in the aorta, with this effect persisting for at least 72 h. These findings demonstrate the prolonged stability and effective targeting capabilities of HMNVsM-D. Furthermore, the study revealed that treatment with HMNVsM-D + MF markedly inhibited plaque formation in ApoE−/− mice.
Fig. 10.
Representative HM/NPs with macrophage-HEK293T hybrid membranes for CVDs treatments. A). Hybrid membrane-coated NPs for MerTK protein targeting to alleviate atherosclerosis. i, ii) Schematic illustration of the preparation process of HMNVsM-D (i) and its therapeutic effects against atherosclerosis (ii). B) TEM images of HMNVs, HMNVsM, HMNVsM−D. Scale bar = 500 nm. C) Exemplary IVIS images of mice and aortas following tail vein injection of HMNVs. Reproduced with permission [49]. Copyright 2024, Springer Nature.
The findings emphasize the pivotal role of MerTK in the pathogenesis of atherosclerosis, particularly in the context of diabetes, and highlight its therapeutic potential [177]. The observed mitigation of atherosclerosis via restored MerTK expression, likely facilitated by enhanced efferocytosis, supports prior studies on MerTK's role in regulating inflammation and cellular debris clearance. The proposed hybrid membrane nanovesicle-based delivery system offers a novel and promising strategy for targeted MerTK protein restoration. This approach not only underscores the translational potential but also opens new avenues for precision therapy in diabetic atherosclerosis. Future investigations should focus on the optimizing nanovesicle delivery mechanisms and evaluating long-term safety and efficacy in both preclinical and clinical settings.
7.6. Macrophage-neutrophil hybrid membranes
Myocardial ischemia induces cardiomyocyte injury and death, triggering a robust inflammatory response that critically modulates subsequent myocardial damage and repair processes. To precisely regulate ischemia-driven inflammation in acute MI, Zheng et al. [51] engineered HM4oRL, an innovative bifunctional nanotherapeutic platform featuring a hybrid membrane coating derived from naive macrophages and activated neutrophils (Fig. 11). This rationally designed system demonstrates dual therapeutic efficacy by concurrently attenuating cardiomyocyte injury and modulating dysregulated inflammatory signaling cascades. The hybrid membrane coating endowed these HM4oRL NPs with targeted tropism toward ischemic myocardium, while their functionality as T1-weighted magnetic resonance (MR) contrast agents was rigorously validated via preclinical MR imaging. The expression levels of pyroptosis-associated proteins (cleaved-gasdermin D) were quantitatively analyzed in the infarcted myocardial tissue. immunohistochemical results demonstrated a statistically significant downregulation of these markers in the HM4oRL-treated group compared to the MI control group.
Fig. 11.
Representative HM/NPs with macrophage-neutrophil hybrid membranes for CVDs treatments. A) Schematic illustration of the synthetic strategy for fabricating biomimetic HM4oRL NPs. B) Schematic representation of the therapeutic effects of HM4oRL NPs on myocardial repair and their utility as contrast agents for T1-weighted magnetic resonance imaging (MRI) at 9.4 T in a murine AMI model. C) Ex vivo near-infrared fluorescence (NIRF) imaging of cardiac tissues harvested from (AMI) and sham-operated mice at designated time points post-coronary artery ligation. D) Comparative immunohistochemical analysis of GSDMD expression levels in infarct border zones following therapeutic interventions. Reproduced with permission [51]. Copyright 2025, Wiley.
The utilization of cell-derived membranes as biomimetic surface modifications for nanoscale drug delivery systems demonstrates significant therapeutic potential, owing to their innate capacity to mediate targeted accumulation at pathological sites through intrinsic homing mechanisms.
7.7. Rational for cell membrane selection and combination design
The combinatorial membrane design in HM/NPs follows a disease-specific engineering principle, where membrane proteins from different cell types are strategically selected to address distinct pathological features. Below we detail the design logic for key applications.
7.7.1. MI
The macrophage-platelet hybrid membrane system is engineered at a 1:1 ratio to synergistically combine the unique functionalities of both cell types. Platelet-derived components (CD41, P-selectin) mediate targeted adhesion to injured endothelium through von Willebrand factor (vWF) binding and facilitate margination to inflamed sites via shear-dependent rolling. Meanwhile, macrophage-derived components (CD47, SIRPα) provide critical "self-marker" signaling to evade phagocytic clearance while enhancing penetration into ischemic myocardium through CCR2-CCL2 chemotaxis. This dual-functionality design ensures prolonged circulation and precise delivery to diseased tissues.
7.7.2. Atherosclerosis
The macrophage-endothelial hybrid membrane system employs a 2:1 ratio to specifically target atherosclerotic plaques through dual-targeting mechanisms. Endothelial-derived markers (ICAM-1, VCAM-1) enable precise homing to activated endothelial cells in plaque neovasculature [178], while macrophage-specific markers (CD36, SR-A) promote efficient uptake by foam cells within lipid-rich plaque regions. This optimized ratio was determined through systematic in vivo screening to maximize plaque accumulation while minimizing off-target effects.
8. Conclusion and outlook
In summary, this review underscores recent advancements in state-of-the-art technologies employed to engineer HM/NPs for the treatment of CVDs. This review systematically examines the development and therapeutic potential of HM/NPs for CVDs. We highlight the evolution of cell membrane coating technology, from early single-cell membrane designs to advanced hybrid systems that integrate functionalities from multiple cell types (e.g., erythrocyte-platelet, macrophage-neutrophil). A critical analysis of HM/NP fabrication methodologies is provided, including membrane extraction techniques, NP core selection (with emphasis on polymeric systems), and in vitro validation approaches. Furthermore, we explore the mechanism of action of HM/NPs. HM/NPs has emerged as a promising strategy for enhancing the in vivo efficacy of nanodelivery systems, offering a bridge between synthetic nanomaterials and biological entities. By incorporating cell membrane coatings, HM/NPs retain the intrinsic physicochemical properties of their synthetic counterparts while adopting the unique characteristics of cellular membranes. This combination results in superior biocompatibility, enhanced biointerface interactions, retention of cellular functionalities, immune evasion, and prolonged circulation half-life [179]. The rapid advancements in nanotechnology have enabled the design and fabrication of biocompatible nanocarriers with increasingly sophisticated and multifunctional biocompatible nanocarriers. HM/NPs provide an effective method to replicate cellular bio-interfacial properties, which conventional synthesis techniques often struggle to achieve.
The design of novel HM/NPs can be guided by a function-oriented strategy, selecting specific membrane types based on their intrinsic properties and alignment with therapeutic goals. The selection of membrane components should be tailored according to the specific pathophysiology of different CVDs—for instance, platelet-derived membranes for targeted thrombosis therapy, macrophage-derived membranes for atherosclerotic plaque modulation, stem cell membranes for ischemic injury repair, and endothelial cell membranes for addressing hypertensive vascular remodeling. By utilizing natural biomolecules refined through evolutionary processes, HM/NPs bypass the lengthy development of synthetic ligands, offering a safe and reliable strategy for cardiovascular disease treatment. By integrating the benefits of a synthesized NP core with a natural cell membrane coating, a biocompatible carrier is created that can simultaneously load and deliver multiple therapeutic agents. Moreover, the incorporation of native cell membrane properties and surface chemistry into these NPs enables functionalities that cannot be attained through traditional chemical conjugation techniques, making them particularly promising for CVD treatments.
Despite their significant promise, several challenges must be addressed for the clinical translation of HM/NPs. Immune rejection remains a primary obstacle, and using autologous cell membranes is recommended to fabricate biomimetic NPs that minimize immunogenic responses [180]. Additionally, the complex isolation of membranes from nucleated cells often results in the loss of critical membrane proteins, limiting the functional retention of HM/NPs and reducing their therapeutic efficacy. Improved extraction techniques are therefore crucial to preserve these components [181]. Furthermore, current NP core materials used in HM/NPs construction often lack innovation, highlighting the need for continuous exploration and development of novel nanomaterials to enhance transit efficiency and overall therapeutic outcomes in cardiovascular applications. Currently, potential challenges including signal interference, functional redundancy, and elevated immunogenicity arising from interactions between heterologous membrane proteins remain understudied. Systematic investigation of these factors is warranted in future studies.
The HM/NPs platform represents a transformative paradigm in CVDs therapeutics, with three key avenues for future development: First, advances in nanomaterial functionalization (e.g., ligand conjugation strategies) could further enhance target specificity to atherosclerotic plaques or ischemic myocardium. Second, breakthroughs in membrane engineering, particularly hybrid membrane systems incorporating engineered extracellular vesicles, may overcome current limitations in immune evasion and circulation time. Third, the integration of stimulus-responsive elements (e.g., ROS-sensitive or pH-triggered release mechanisms) could enable precision drug release at pathological sites [182].
While current systems have bridged critical gaps between synthetic and biological properties, fundamental challenges remain in large-scale manufacturing reproducibility and long-term biosafety evaluation. Future research should prioritize: (1) standardized characterization protocols for membrane-coated NPs, (2) preclinical validation in large animal models of acute MI and chronic heart failure, and (3) combinatorial approaches with emerging modalities like gene editing or mitochondrial-targeted therapies. These developments will ultimately determine the clinical translatability of this platform for next-generation cardiovascular nanomedicine.
CRediT authorship contribution statement
He Lu: Writing – original draft, Visualization, Validation, Software, Methodology, Investigation, Formal analysis, Data curation. Yaohui Jiang: Writing – original draft, Software, Methodology, Investigation, Formal analysis, Data curation. Rui Luo: Validation, Software, Methodology, Investigation, Formal analysis, Data curation. Dexing Zhou: Visualization, Software, Investigation, Formal analysis, Data curation. Feihu Zheng: Software, Methodology, Investigation, Formal analysis, Data curation. Liran Shi: Visualization, Validation, Methodology, Investigation, Formal analysis. He Zhang: Validation, Software, Methodology, Investigation, Formal analysis. Yong Wang: Validation, Software, Methodology, Investigation, Data curation. Xiaodong Xu: Software, Investigation, Formal analysis, Data curation. Renfang Zou: Software, Methodology, Data curation. Yujing Zhou: Software, Data curation. Shuai Ren: Writing – review & editing, Supervision, Formal analysis. Xiaocheng Wang: Writing – review & editing, Supervision, Formal analysis. Haiqiang Sang: Writing – review & editing, Supervision, Formal analysis.
Ethics approval and consent to participate
Ethics approval and consent to participate do not apply to this review manuscript.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Acknowledgments
This work was supported by National Natural Science Foundation of China (82202135), Young Elite Scientists Sponsorship Program by China Association of Chinese Medicine (2024-QNRC2-B16), China Postdoctoral Science Foundation (2023M741808).
Contributor Information
Shuai Ren, Email: shuairen@njucm.edu.cn.
Xiaocheng Wang, Email: wangxiaocheng@ucas.ac.cn.
Haiqiang Sang, Email: fccsanghq@zzu.edu.cn.
Data availability
No data was used for the research described in the article.
References
- 1.Benjamin E.J., Muntner P., Alonso A., Bittencourt M.S., Callaway C.W., Carson A.P., Chamberlain A.M., Chang A.R., Cheng S., Das S.R., Delling F.N., Djousse L., Elkind M.S.V., Ferguson J.F., Fornage M., Jordan L.C., Khan S.S., Kissela B.M., Knutson K.L., Kwan T.W., Lackland D.T., Lewis T.T., Lichtman J.H., Longenecker C.T., Loop M.S., Lutsey P.L., Martin S.S., Matsushita K., Moran A.E., Mussolino M.E., O'Flaherty M., Pandey A., Perak A.M., Rosamond W.D., Roth G.A., Sampson U.K.A., Satou G.M., Schroeder E.B., Shah S.H., Spartano N.L., Stokes A., Tirschwell D.L., Tsao C.W., Turakhia M.P., VanWagner L.B., Wilkins J.T., Wong S.S., Virani S.S. Heart disease and stroke statistics-2019 update: a report from the American heart association. Circulation. 2019;139:e56–e528. doi: 10.1161/CIR.0000000000000659. [DOI] [PubMed] [Google Scholar]
- 2.Yadgir S., Johnson C.O., Aboyans V., Adebayo O.M., Adedoyin R.A., Afarideh M., Alahdab F., Alashi A., Alipour V., Arabloo J., Azari S., Barthelemy C.M., Benziger C.P., Berman A.E., Bijani A., Carrero J.J., Carvalho F., Daryani A., Durães A.R., Esteghamati A., Farid T.A., Farzadfar F., Fernandes E., Filip I., Gad M.M., Hamidi S., Hay S.I., Ilesanmi O.S., Naghibi Irvani S.S., Jürisson M., Kasaeian A., Kengne A.P., Khan A.R., Kisa A., Kisa S., Kolte D., Manafi N., Manafi A., Mensah G.A., Mirrakhimov E.M., Mohammad Y., Mokdad A.H., Negoi R.I., Thi Nguyen H.L., Nguyen T.H., Nixon M.R., Otto C.M., Patel S., Pilgrim T., Radfar A., Rawaf D.L., Rawaf S., Rawasia W.F., Rezapour A., Roever L., Saad A.M., Saadatagah S., Senthilkumaran S., Sliwa K., Tesfay B.E., Tran B.X., Ullah I., Vaduganathan M., Vasankari T.J., Wolfe C.D.A., Yonemoto N., Roth G.A. Global, regional, and national burden of calcific aortic valve and degenerative mitral valve diseases, 1990-2017. Circulation. 2020;141:1670–1680. doi: 10.1161/CIRCULATIONAHA.119.043391. [DOI] [PubMed] [Google Scholar]
- 3.Alahmad B., Khraishah H., Royé D., Vicedo-Cabrera A.M., Guo Y., Papatheodorou S.I., Achilleos S., Acquaotta F., Armstrong B., Bell M.L., Pan S.C., de Sousa Zanotti Stagliorio Coelho M., Colistro V., Dang T.N., Van Dung D., De' Donato F.K., Entezari A., Guo Y.L., Hashizume M., Honda Y., Indermitte E., Íñiguez C., Jaakkola J.J.K., Kim H., Lavigne E., Lee W., Li S., Madureira J., Mayvaneh F., Orru H., Overcenco A., Ragettli M.S., Ryti N.R.I., Saldiva P.H.N., Scovronick N., Seposo X., Sera F., Silva S.P., Stafoggia M., Tobias A., Garshick E., Bernstein A.S., Zanobetti A., Schwartz J., Gasparrini A., Koutrakis P. Associations between extreme temperatures and cardiovascular cause-specific mortality: results from 27 countries. Circulation. 2023;147:35–46. doi: 10.1161/CIRCULATIONAHA.122.061832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Virani S.S., Alonso A., Aparicio H.J., Benjamin E.J., Bittencourt M.S., Callaway C.W., Carson A.P., Chamberlain A.M., Cheng S., Delling F.N., Elkind M.S.V., Evenson K.R., Ferguson J.F., Gupta D.K., Khan S.S., Kissela B.M., Knutson K.L., Lee C.D., Lewis T.T., Liu J., Loop M.S., Lutsey P.L., Ma J., Mackey J., Martin S.S., Matchar D.B., Mussolino M.E., Navaneethan S.D., Perak A.M., Roth G.A., Samad Z., Satou G.M., Schroeder E.B., Shah S.H., Shay C.M., Stokes A., VanWagner L.B., Wang N.Y., Tsao C.W. Heart disease and stroke statistics-2021 update: a report from the American heart association. Circulation. 2021;143:e254–e743. doi: 10.1161/CIR.0000000000000950. [DOI] [PubMed] [Google Scholar]
- 5.Townsend N., Kazakiewicz D., Lucy Wright F., Timmis A., Huculeci R., Torbica A., Gale C.P., Achenbach S., Weidinger F., Vardas P. Epidemiology of cardiovascular disease in Europe. Nat. Rev. Cardiol. 2022;19:133–143. doi: 10.1038/s41569-021-00607-3. [DOI] [PubMed] [Google Scholar]
- 6.Luengo-Fernandez R., Walli-Attaei M., Gray A., Torbica A., Maggioni A.P., Huculeci R., Bairami F., Aboyans V., Timmis A.D., Vardas P., Leal J. Economic burden of cardiovascular diseases in the European Union: a population-based cost study. Eur. Heart J. 2023;44:4752–4767. doi: 10.1093/eurheartj/ehad583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Luo X.M., Yan C., Feng Y.M. Nanomedicine for the treatment of diabetes-associated cardiovascular diseases and fibrosis. Adv. Drug Deliv. Rev. 2021;172:234–248. doi: 10.1016/j.addr.2021.01.004. [DOI] [PubMed] [Google Scholar]
- 8.Perry A.S., Dooley E.E., Master H., Spartano N.L., Brittain E.L., Pettee Gabriel K. Physical activity over the lifecourse and cardiovascular disease. Circ. Res. 2023;132:1725–1740. doi: 10.1161/CIRCRESAHA.123.322121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Ziaeian B., Fonarow G.C. Epidemiology and aetiology of heart failure. Nat. Rev. Cardiol. 2016;13:368–378. doi: 10.1038/nrcardio.2016.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Li X., Zhao D., Zhang F. Multifunctional upconversion-magnetic hybrid nanostructured materials: synthesis and bioapplications. Theranostics. 2013;3:292–305. doi: 10.7150/thno.5289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gao J., Gu H., Xu B. Multifunctional magnetic nanoparticles: design, synthesis, and biomedical applications. Acc. Chem. Res. 2009;42:1097–1107. doi: 10.1021/ar9000026. [DOI] [PubMed] [Google Scholar]
- 12.Hulla J.E., Sahu S.C., Hayes A.W. Nanotechnology: history and future. Hum. Exp. Toxicol. 2015;34:1318–1321. doi: 10.1177/0960327115603588. [DOI] [PubMed] [Google Scholar]
- 13.Whitesides G.M. The 'right' size in nanobiotechnology. Nat. Biotechnol. 2003;21:1161–1165. doi: 10.1038/nbt872. [DOI] [PubMed] [Google Scholar]
- 14.Farokhzad O.C., Langer R. Impact of nanotechnology on drug delivery. ACS Nano. 2009;3:16–20. doi: 10.1021/nn900002m. [DOI] [PubMed] [Google Scholar]
- 15.Behzadi S., Serpooshan V., Tao W., Hamaly M.A., Alkawareek M.Y., Dreaden E.C., Brown D., Alkilany A.M., Farokhzad O.C., Mahmoudi M. Cellular uptake of nanoparticles: journey inside the cell. Chem. Soc. Rev. 2017;46:4218–4244. doi: 10.1039/c6cs00636a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kamaly N., Fredman G., Subramanian M., Gadde S., Pesic A., Cheung L., Fayad Z.A., Langer R., Tabas I., Farokhzad O.C. Development and in vivo efficacy of targeted polymeric inflammation-resolving nanoparticles. Proc. Natl. Acad. Sci. USA. 2013;110:6506–6511. doi: 10.1073/pnas.1303377110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Pang L., Zhang C., Qin J., Han L., Li R., Hong C., He H., Wang J. A novel strategy to achieve effective drug delivery: exploit cells as carrier combined with nanoparticles. Drug Deliv. 2017;24:83–91. doi: 10.1080/10717544.2016.1230903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Wan A.C., Ying J.Y. Nanomaterials for in situ cell delivery and tissue regeneration. Adv. Drug Deliv. Rev. 2010;62:731–740. doi: 10.1016/j.addr.2010.02.002. [DOI] [PubMed] [Google Scholar]
- 19.Fang R.H., Kroll A.V., Gao W., Zhang L. Cell membrane coating nanotechnology. Adv. Mater. 2018;30 doi: 10.1002/adma.201706759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hu C.M., Fang R.H., Wang K.C., Luk B.T., Thamphiwatana S., Dehaini D., Nguyen P., Angsantikul P., Wen C.H., Kroll A.V., Carpenter C., Ramesh M., Qu V., Patel S.H., Zhu J., Shi W., Hofman F.M., Chen T.C., Gao W., Zhang K., Chien S., Zhang L. Nanoparticle biointerfacing by platelet membrane cloaking. Nature. 2015;526:118–121. doi: 10.1038/nature15373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hu Q., Qian C., Sun W., Wang J., Chen Z., Bomba H.N., Xin H., Shen Q., Gu Z. Engineered nanoplatelets for enhanced treatment of multiple myeloma and thrombus. Adv. Mater. 2016;28:9573–9580. doi: 10.1002/adma.201603463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Zhang W., Yang X., Huang X., Chen L. Bioinspired nanovesicles released from injectable hydrogels facilitate diabetic wound healing by regulating macrophage polarization and endothelial cell dysfunction. J. Nanobiotechnology. 2023;21:358. doi: 10.1186/s12951-023-02119-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Thamphiwatana S., Angsantikul P., Escajadillo T., Zhang Q., Olson J., Luk B.T., Zhang S., Fang R.H., Gao W., Nizet V., Zhang L. Macrophage-like nanoparticles concurrently absorbing endotoxins and proinflammatory cytokines for sepsis management. Proc. Natl. Acad. Sci. USA. 2017;114:11488–11493. doi: 10.1073/pnas.1714267114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.He Y., Zhang S., She Y., Liu Z., Zhu Y., Cheng Q., Ji X. Innovative utilization of cell membrane-coated nanoparticles in precision cancer therapy. Exploration (Beijing) 2024;4 doi: 10.1002/EXP.20230164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Huang X., Wang L., Guo H., Zhang W. Macrophage membrane-coated nanovesicles for dual-targeted drug delivery to inhibit tumor and induce macrophage polarization. Bioact. Mater. 2023;23:69–79. doi: 10.1016/j.bioactmat.2022.09.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Jia L., Wang R., Zhang T., Li M., Xie X., Wang S. Rationally remodeled biomimetic aptasensor enhances the detection performance of heterogeneous circulating tumor cells, Chemical. Eng. J. 2024;499 [Google Scholar]
- 27.Uchida S., Perche F., Pichon C., Cabral H. Nanomedicine-based approaches for mRNA delivery. Mol. Pharm. 2020;17:3654–3684. doi: 10.1021/acs.molpharmaceut.0c00618. [DOI] [PubMed] [Google Scholar]
- 28.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 doi: 10.1016/j.biomaterials.2022.121821. [DOI] [PubMed] [Google Scholar]
- 29.Wang Y., Zhang K., Qin X., Li T., Qiu J., Yin T., Huang J., McGinty S., Pontrelli G., Ren J., Wang Q., Wu W., Wang G. Biomimetic nanotherapies: red blood cell based core-shell structured nanocomplexes for atherosclerosis management. Adv. Sci. 2019;6 doi: 10.1002/advs.201900172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zhao Y., Xie R., Yodsanit N., Ye M., Wang Y., Gong S. Biomimetic fibrin-targeted and H(2)O(2)-responsive nanocarriers for thrombus therapy. Nano Today. 2020;35 doi: 10.1016/j.nantod.2020.100986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Roullet S., Luc N., Rayes J., Solarz J., Disharoon D., Ditto A., Gahagan E., Pawlowski C., Sefiane T., Adam F., Casari C., Christophe O.D., Bruckman M., Lenting P.J., Sen Gupta A., Denis C.V. Efficacy of platelet-inspired hemostatic nanoparticles on bleeding in von Willebrand disease murine models. Blood. 2023;141:2891–2900. doi: 10.1182/blood.2022018956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Geng B., Chen X., Chi J., Li F., Yim W.Y., Wang K., Li C., Xie M., Zhu P., Fan Z., Shi J., Hu Z., Zhang Y., Dong N. Platelet membrane-coated alterbrassicene A nanoparticle inhibits calcification of the aortic valve by suppressing phosphorylation P65 NF-κB. Theranostics. 2023;13:3781–3793. doi: 10.7150/thno.85323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wang Q., Song Y., Gao J., Li Q., Chen J., Xie Y., Wang Z., Tan H., Yang H., Zhang N., Qian J., Pang Z., Huang Z., Ge J. Hippo pathway-manipulating neutrophil-mimic hybrid nanoparticles for cardiac ischemic injury via modulation of local immunity and cardiac regeneration. Acta Pharm. Sin. B. 2023;13:4999–5015. doi: 10.1016/j.apsb.2023.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Chen J., Song Y., Wang Q., Li Q., Tan H., Gao J., Zhang N., Weng X., Sun D., Yakufu W., Wang Z., Qian J., Pang Z., Huang Z., Ge J. Targeted neutrophil-mimetic liposomes promote cardiac repair by adsorbing proinflammatory cytokines and regulating the immune microenvironment. J. Nanobiotechnology. 2022;20:218. doi: 10.1186/s12951-022-01433-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lu H., Wang J., Chen Z., Wang J., Jiang Y., Xia Z., Hou Y., Shang P., Li R., Liu Y., Xie J. Engineered macrophage membrane-coated S100A9-siRNA for ameliorating myocardial ischemia-reperfusion injury. Adv. Sci. 2024;11 doi: 10.1002/advs.202403542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wu Y., Wan S., Yang S., Hu H., Zhang C., Lai J., Zhou J., Chen W., Tang X., Luo J., Zhou X., Yu L., Wang L., Wu A., Fan Q., Wu J. Macrophage cell membrane-based nanoparticles: a new promising biomimetic platform for targeted delivery and treatment. J. Nanobiotechnology. 2022;20:542. doi: 10.1186/s12951-022-01746-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zhen X., Li Y., Yuan W., Zhang T., Li M., Huang J., Kong N., Xie X., Wang S., Tao W. Biointerface-engineered hybrid nanovesicles for targeted reprogramming of tumor microenvironment. Adv. Mater. 2024;36 doi: 10.1002/adma.202401495. [DOI] [PubMed] [Google Scholar]
- 38.Hu C.M., Zhang L., Aryal S., Cheung C., Fang R.H., Zhang L. Erythrocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc. Natl. Acad. Sci. USA. 2011;108:10980–10985. doi: 10.1073/pnas.1106634108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Ma W., Zhu D., Li J., Chen X., Xie W., Jiang X., Wu L., Wang G., Xiao Y., Liu Z., Wang F., Li A., Shao D., Dong W., Liu W., Yuan Y. Coating biomimetic nanoparticles with chimeric antigen receptor T cell-membrane provides high specificity for hepatocellular carcinoma photothermal therapy treatment. Theranostics. 2020;10:1281–1295. doi: 10.7150/thno.40291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Tan H., Song Y., Chen J., Zhang N., Wang Q., Li Q., Gao J., Yang H., Dong Z., Weng X., Wang Z., Sun D., Yakufu W., Pang Z., Huang Z., Ge J. Platelet-like fusogenic liposome-mediated targeting delivery of miR-21 improves myocardial remodeling by reprogramming macrophages post myocardial ischemia-reperfusion injury. Adv. Sci. 2021;8 doi: 10.1002/advs.202100787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Xu X., Li M., Yu F., Wei Q., Liu Y., Tong J., Yang F. Platelet membrane nanocarriers cascade targeting delivery system to improve myocardial remodeling post myocardial ischemia-reperfusion injury. Adv. Sci. 2024;11 doi: 10.1002/advs.202308727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Shan S., Chen J., Sun Y., Wang Y., Xia B., Tan H., Pan C., Gu G., Zhong J., Qing G., Zhang Y., Wang J., Wang Y., Wang Y., Zuo P., Xu C., Li F., Guo W., Xu L., Chen M., Fan Y., Zhang L., Liang X.J. Functionalized macrophage exosomes with panobinostat and PPM1D-siRNA for diffuse intrinsic pontine gliomas therapy. Adv. Sci. 2022;9 doi: 10.1002/advs.202200353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Dehaini D., Wei X., Fang R.H., Masson S., Angsantikul P., Luk B.T., Zhang Y., Ying M., Jiang Y., Kroll A.V., Gao W., Zhang L. Erythrocyte-platelet hybrid membrane coating for enhanced nanoparticle functionalization. Adv. Mater. 2017;29 doi: 10.1002/adma.201606209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zhang N., Song Y., Huang Z., Chen J., Tan H., Yang H., Fan M., Li Q., Wang Q., Gao J., Pang Z., Qian J., Ge J. Monocyte mimics improve mesenchymal stem cell-derived extracellular vesicle homing in a mouse MI/RI model. Biomaterials. 2020;255 doi: 10.1016/j.biomaterials.2020.120168. [DOI] [PubMed] [Google Scholar]
- 45.Hu S., Wang X., Li Z., Zhu D., Cores J., Wang Z., Li J., Mei X., Cheng X., Su T., Cheng K. Platelet membrane and stem cell exosome hybrid enhances cellular uptake and targeting to heart injury. Nano Today. 2021;39 doi: 10.1016/j.nantod.2021.101210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zhou Y., Liang Q., Wu X., Duan S., Ge C., Ye H., Lu J., Zhu R., Chen Y., Meng F., Yin L. siRNA delivery against myocardial ischemia reperfusion injury mediated by reversibly camouflaged biomimetic nanocomplexes. Adv. Mater. 2023;35 doi: 10.1002/adma.202210691. [DOI] [PubMed] [Google Scholar]
- 47.Xie L., Chen J., Hu H., Zhu Y., Wang X., Zhou S., Wang F., Xiang M. Engineered M2 macrophage-derived extracellular vesicles with platelet membrane fusion for targeted therapy of atherosclerosis. Bioact. Mater. 2024;35:447–460. doi: 10.1016/j.bioactmat.2024.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Tan Y., Wang X., Gu Y., Bao X., Lu H., Sun X., Kang L., Xu B. Neutrophil and endothelial cell membranes coassembled roflumilast nanoparticles attenuate myocardial ischemia/reperfusion injury. Nanomedicine (Lond) 2024;19:779–797. doi: 10.2217/nnm-2023-0313. [DOI] [PubMed] [Google Scholar]
- 49.Qiu S., Liu J., Chen J., Li Y., Bu T., Li Z., Zhang L., Sun W., Zhou T., Hu W., Yang G., Yuan L., Duan Y., Xing C. Targeted delivery of MerTK protein via cell membrane engineered nanoparticle enhances efferocytosis and attenuates atherosclerosis in diabetic ApoE(-/-) Mice. J. Nanobiotechnology. 2024;22:178. doi: 10.1186/s12951-024-02463-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Li Y., Yu J., Cheng C., Chen W., Lin R., Wang Y., Cui W., Meng J., Du J., Wang Y. Platelet and erythrocyte membranes coassembled biomimetic nanoparticles for heart failure treatment. ACS Nano. 2024;18:26614–26630. doi: 10.1021/acsnano.4c04814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Zheng T., Sheng J., Wang Z., Wu H., Zhang L., Wang S., Li J., Zhang Y., Lu G., Zhang L. Injured myocardium-targeted theranostic nanoplatform for multi-dimensional immune-inflammation regulation in acute myocardial infarction. Adv. Sci. 2025;12 doi: 10.1002/advs.202414740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Sun M., Yang J., Fan Y., Zhang Y., Sun J., Hu M., Sun K., Zhang J. Beyond extracellular vesicles: hybrid membrane nanovesicles as emerging advanced tools for biomedical applications. Adv. Sci. 2023;10 doi: 10.1002/advs.202303617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wang Y., Zhou Q., Lu L., Xu J., Yang G., Sun X., Bao X., Kang L., Lv P., Liu R., Xu B., Yang Q., Mu D., Zhang B. Combining oxygen delivery and generation for targeted atherosclerosis therapy. J. Control. Release. 2025;380:1017–1030. doi: 10.1016/j.jconrel.2025.02.053. [DOI] [PubMed] [Google Scholar]
- 54.Li Y., Che J., Chang L., Guo M., Bao X., Mu D., Sun X., Zhang X., Lu W., Xie J. CD47- and integrin α4/β1-Comodified-Macrophage-Membrane-Coated nanoparticles enable delivery of colchicine to atherosclerotic plaque. Adv. Healthc. Mater. 2022;11 doi: 10.1002/adhm.202101788. [DOI] [PubMed] [Google Scholar]
- 55.Moore G.E., Ulrich K. Suspension cultures of mammalian cells. A review. J. Surg. Res. 1965;5:270–282. doi: 10.1016/s0022-4804(65)80017-8. [DOI] [PubMed] [Google Scholar]
- 56.Su L., Zhao D., Huang Q., Zhao X., Chen Q., Rao H., Guo L., Hao J. Preparation of pectin-coated and chitosan-coated phenylethanoside liposomes: studies on characterization, stability, digestion and release behavior. Int. J. Biol. Macromol. 2024;261 doi: 10.1016/j.ijbiomac.2024.129442. [DOI] [PubMed] [Google Scholar]
- 57.Oroojalian F., Beygi M., Baradaran B., Mokhtarzadeh A., Shahbazi M.A. Immune cell membrane-coated biomimetic nanoparticles for targeted cancer therapy. Small. 2021;17 doi: 10.1002/smll.202006484. [DOI] [PubMed] [Google Scholar]
- 58.Choi B., Park W., Park S.B., Rhim W.K., Han D.K. Recent trends in cell membrane-cloaked nanoparticles for therapeutic applications. Methods. 2020;177:2–14. doi: 10.1016/j.ymeth.2019.12.004. [DOI] [PubMed] [Google Scholar]
- 59.Wei X., Gao J., Wang F., Ying M., Angsantikul P., Kroll A.V., Zhou J., Gao W., Lu W., Fang R.H., Zhang L. In situ capture of bacterial toxins for antivirulence vaccination. Adv. Mater. 2017;29 doi: 10.1002/adma.201701644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Rao L., Cai B., Bu L.L., Liao Q.Q., Guo S.S., Zhao X.Z., Dong W.F., Liu W. Microfluidic electroporation-facilitated synthesis of erythrocyte membrane-coated magnetic nanoparticles for enhanced imaging-guided cancer therapy. ACS Nano. 2017;11:3496–3505. doi: 10.1021/acsnano.7b00133. [DOI] [PubMed] [Google Scholar]
- 61.Allen C., Evans J.C. 'Hip to be square': designing PLGA formulations for the future. J. Control. Release. 2020;319:487–488. doi: 10.1016/j.jconrel.2020.01.050. [DOI] [PubMed] [Google Scholar]
- 62.Valcourt D.M., Dang M.N., Scully M.A., Day E.S. Nanoparticle-mediated Co-delivery of notch-1 antibodies and ABT-737 as a potent treatment strategy for triple-negative breast cancer. ACS Nano. 2020;14:3378–3388. doi: 10.1021/acsnano.9b09263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Mitchell M.J., Billingsley M.M., Haley R.M., Wechsler M.E., Peppas N.A., Langer R. Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug Discov. 2021;20:101–124. doi: 10.1038/s41573-020-0090-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wang J., Potocny A.M., Rosenthal J., Day E.S. Gold nanoshell-linear tetrapyrrole conjugates for near infrared-activated dual photodynamic and photothermal therapies. ACS Omega. 2020;5:926–940. doi: 10.1021/acsomega.9b04150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Millstone J.E., Hurst S.J., Métraux G.S., Cutler J.I., Mirkin C.A. Colloidal gold and silver triangular nanoprisms. Small. 2009;5:646–664. doi: 10.1002/smll.200801480. [DOI] [PubMed] [Google Scholar]
- 66.Manshian B.B., Jiménez J., Himmelreich U., Soenen S.J. Personalized medicine and follow-up of therapeutic delivery through exploitation of quantum dot toxicity. Biomaterials. 2017;127:1–12. doi: 10.1016/j.biomaterials.2017.02.039. [DOI] [PubMed] [Google Scholar]
- 67.Dos Santos Rodrigues B., Oue H., Banerjee A., Kanekiyo T., Singh J. Dual functionalized liposome-mediated gene delivery across triple co-culture blood brain barrier model and specific in vivo neuronal transfection. J. Control. Release. 2018;286:264–278. doi: 10.1016/j.jconrel.2018.07.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Fonseca-Santos B., Gremião M.P., Chorilli M. Nanotechnology-based drug delivery systems for the treatment of Alzheimer's disease. Int. J. Nanomedicine. 2015;10:4981–5003. doi: 10.2147/IJN.S87148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Zhang Y., Chan J.W., Moretti A., Uhrich K.E. Designing polymers with sugar-based advantages for bioactive delivery applications. J. Control. Release. 2015;219:355–368. doi: 10.1016/j.jconrel.2015.09.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Stevens C.A., Kaur K., Klok H.A. Self-assembly of protein-polymer conjugates for drug delivery. Adv. Drug Deliv. Rev. 2021;174:447–460. doi: 10.1016/j.addr.2021.05.002. [DOI] [PubMed] [Google Scholar]
- 71.Park K., Skidmore S., Hadar J., Garner J., Park H., Otte A., Soh B.K., Yoon G., Yu D., Yun Y., Lee B.K., Jiang X., Wang Y. Injectable, long-acting PLGA formulations: analyzing PLGA and understanding microparticle formation. J. Control. Release. 2019;304:125–134. doi: 10.1016/j.jconrel.2019.05.003. [DOI] [PubMed] [Google Scholar]
- 72.von Burkersroda F., Schedl L., Göpferich A. Why degradable polymers undergo surface erosion or bulk erosion. Biomaterials. 2002;23:4221–4231. doi: 10.1016/s0142-9612(02)00170-9. [DOI] [PubMed] [Google Scholar]
- 73.Kashi T.S., Eskandarion S., Esfandyari-Manesh M., Marashi S.M., Samadi N., Fatemi S.M., Atyabi F., Eshraghi S., Dinarvand R. Improved drug loading and antibacterial activity of minocycline-loaded PLGA nanoparticles prepared by solid/oil/water ion pairing method. Int. J. Nanomedicine. 2012;7:221–234. doi: 10.2147/IJN.S27709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Panyam J., Labhasetwar V. Biodegradable nanoparticles for drug and gene delivery to cells and tissue. Adv. Drug Deliv. Rev. 2003;55:329–347. doi: 10.1016/s0169-409x(02)00228-4. [DOI] [PubMed] [Google Scholar]
- 75.Fu K., Pack D.W., Klibanov A.M., Langer R. Visual evidence of acidic environment within degrading poly(lactic-co-glycolic acid) (PLGA) microspheres. Pharm. Res. 2000;17:100–106. doi: 10.1023/a:1007582911958. [DOI] [PubMed] [Google Scholar]
- 76.Xu X., Xie K., Zhang X.Q., Pridgen E.M., Park G.Y., Cui D.S., Shi J., Wu J., Kantoff P.W., Lippard S.J., Langer R., Walker G.C., Farokhzad O.C. Enhancing tumor cell response to chemotherapy through nanoparticle-mediated codelivery of siRNA and cisplatin prodrug. Proc. Natl. Acad. Sci. USA. 2013;110:18638–18643. doi: 10.1073/pnas.1303958110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Islam M.A., Xu Y., Tao W., Ubellacker J.M., Lim M., Aum D., Lee G.Y., Zhou K., Zope H., Yu M., Cao W., Oswald J.T., Dinarvand M., Mahmoudi M., Langer R., Kantoff P.W., Farokhzad O.C., Zetter B.R., Shi J. Restoration of tumour-growth suppression in vivo via systemic nanoparticle-mediated delivery of PTEN mRNA. Nat. Biomed. Eng. 2018;2:850–864. doi: 10.1038/s41551-018-0284-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Passaro A., Jänne P.A., Peters S. Antibody-drug conjugates in lung cancer: recent advances and implementing strategies. J. Clin. Oncol. 2023;41:3747–3761. doi: 10.1200/JCO.23.00013. [DOI] [PubMed] [Google Scholar]
- 79.Jin X., Yang G.Y. Pathophysiological roles and applications of glycosphingolipids in the diagnosis and treatment of cancer diseases. Prog. Lipid Res. 2023;91 doi: 10.1016/j.plipres.2023.101241. [DOI] [PubMed] [Google Scholar]
- 80.Falsafi S.R., Rostamabadi H., Assadpour E., Jafari S.M. Morphology and microstructural analysis of bioactive-loaded micro/nanocarriers via microscopy techniques; CLSM/SEM/TEM/AFM. Adv. Colloid Interface Sci. 2020;280 doi: 10.1016/j.cis.2020.102166. [DOI] [PubMed] [Google Scholar]
- 81.Wang S., Jiang R., Meng T., Zhang F., Li J., Jin Y., Lee J., Zhu M., Jiang J. Stem cell membrane-coated isotretinoin for acne treatment. J. Nanobiotechnology. 2020;18:106. doi: 10.1186/s12951-020-00664-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Kobayashi T., Menon A.K. Transbilayer lipid asymmetry. Curr. Biol. 2018;28:R386–r391. doi: 10.1016/j.cub.2018.01.007. [DOI] [PubMed] [Google Scholar]
- 83.Filippov S.K., Khusnutdinov R., Murmiliuk A., Inam W., Zakharova L.Y., Zhang H., Khutoryanskiy V.V. Dynamic light scattering and transmission electron microscopy in drug delivery: a roadmap for correct characterization of nanoparticles and interpretation of results. Mater. Horiz. 2023;10:5354–5370. doi: 10.1039/d3mh00717k. [DOI] [PubMed] [Google Scholar]
- 84.Tian Y., Gong M., Hu Y., Liu H., Zhang W., Zhang M., Hu X., Aubert D., Zhu S., Wu L., Yan X. Quality and efficiency assessment of six extracellular vesicle isolation methods by nano-flow cytometry. J. Extracell. Vesicles. 2020;9 doi: 10.1080/20013078.2019.1697028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Mushtaq U., Bhat S.S., Parray A.A., Nabi S., Wani A.H., Qurashi A.H., Siddiqui K.S., Khanday F.A. A simple, economical and environmental-friendly method for staining protein gels using an extract from walnut-husk. Chem. Biol. Interact. 2021;333 doi: 10.1016/j.cbi.2020.109310. [DOI] [PubMed] [Google Scholar]
- 86.Schägger H. Tricine-SDS-PAGE. Nat. Protoc. 2006;1:16–22. doi: 10.1038/nprot.2006.4. [DOI] [PubMed] [Google Scholar]
- 87.Krishnan N., Jiang Y., Zhou J., Mohapatra A., Peng F.X., Duan Y., Holay M., Chekuri S., Guo Z., Gao W., Fang R.H., Zhang L. A modular approach to enhancing cell membrane-coated nanoparticle functionality using genetic engineering. Nat. Nanotechnol. 2024;19:345–353. doi: 10.1038/s41565-023-01533-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Akinc A., Querbes W., De S., Qin J., Frank-Kamenetsky M., Jayaprakash K.N., Jayaraman M., Rajeev K.G., Cantley W.L., Dorkin J.R., Butler J.S., Qin L., Racie T., Sprague A., Fava E., Zeigerer A., Hope M.J., Zerial M., Sah D.W., Fitzgerald K., Tracy M.A., Manoharan M., Koteliansky V., Fougerolles A., Maier M.A. Targeted delivery of RNAi therapeutics with endogenous and exogenous ligand-based mechanisms. Mol. Ther. 2010;18:1357–1364. doi: 10.1038/mt.2010.85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Van Driessche A.E.S., Ling W.L., Schoehn G., Sleutel M. Nucleation of glucose isomerase protein crystals in a nonclassical disguise: the role of crystalline precursors. Proc. Natl. Acad. Sci. USA. 2022;119 doi: 10.1073/pnas.2108674119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.de Souza W., Attias M. New advances in scanning microscopy and its application to study parasitic protozoa. Exp. Parasitol. 2018;190:10–33. doi: 10.1016/j.exppara.2018.04.018. [DOI] [PubMed] [Google Scholar]
- 91.Kirk S.E., Skepper J.N., Donald A.M. Application of environmental scanning electron microscopy to determine biological surface structure. J. Microsc. 2009;233:205–224. doi: 10.1111/j.1365-2818.2009.03111.x. [DOI] [PubMed] [Google Scholar]
- 92.Weingart J., Vabbilisetty P., Sun X.L. Membrane mimetic surface functionalization of nanoparticles: methods and applications. Adv. Colloid Interface Sci. 2013;197–198:68–84. doi: 10.1016/j.cis.2013.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Yan H., Shao D., Lao Y.H., Li M., Hu H., Leong K.W. Engineering cell membrane-based nanotherapeutics to target inflammation. Adv. Sci. 2019;6 doi: 10.1002/advs.201900605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Han D., Wang F., Qiao Z., Wang B., Zhang Y., Jiang Q., Liu M., Zhuang Y., An Q., Bai Y., Shangguan J., Zhang J., Liang G., Shen D. Neutrophil membrane-camouflaged nanoparticles alleviate inflammation and promote angiogenesis in ischemic myocardial injury. Bioact. Mater. 2023;23:369–382. doi: 10.1016/j.bioactmat.2022.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Chen Z., Zhao P., Luo Z., Zheng M., Tian H., Gong P., Gao G., Pan H., Liu L., Ma A., Cui H., Ma Y., Cai L. Cancer cell membrane-biomimetic nanoparticles for homologous-targeting dual-modal imaging and photothermal therapy. ACS Nano. 2016;10:10049–10057. doi: 10.1021/acsnano.6b04695. [DOI] [PubMed] [Google Scholar]
- 96.Jiang Q., Liu Y., Guo R., Yao X., Sung S., Pang Z., Yang W. Erythrocyte-cancer hybrid membrane-camouflaged melanin nanoparticles for enhancing photothermal therapy efficacy in tumors. Biomaterials. 2019;192:292–308. doi: 10.1016/j.biomaterials.2018.11.021. [DOI] [PubMed] [Google Scholar]
- 97.Lin R.R., Jin L.L., Xue Y.Y., Zhang Z.S., Huang H.F., Chen D.F., Liu Q., Mao Z.W., Wu Z.Y., Tao Q.Q. Hybrid membrane-coated nanoparticles for precise targeting and synergistic therapy in alzheimer's disease. Adv. Sci. 2024;11 doi: 10.1002/advs.202306675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Fang R.H., Gao W., Zhang L. Targeting drugs to tumours using cell membrane-coated nanoparticles. Nat. Rev. Clin. Oncol. 2023;20:33–48. doi: 10.1038/s41571-022-00699-x. [DOI] [PubMed] [Google Scholar]
- 99.Luk B.T., Zhang L. Cell membrane-camouflaged nanoparticles for drug delivery. J. Control. Release. 2015;220:600–607. doi: 10.1016/j.jconrel.2015.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Buchman J.T., Hudson-Smith N.V., Landy K.M., Haynes C.L. Understanding nanoparticle toxicity mechanisms to inform redesign strategies to reduce environmental impact. Acc. Chem. Res. 2019;52:1632–1642. doi: 10.1021/acs.accounts.9b00053. [DOI] [PubMed] [Google Scholar]
- 101.Chen R., Yang J., Wu M., Zhao D., Yuan Z., Zeng L., Hu J., Zhang X., Wang T., Xu J., Zhang J. M2 macrophage hybrid membrane-camouflaged targeted biomimetic nanosomes to reprogram inflammatory microenvironment for enhanced enzyme-thermo-immunotherapy. Adv. Mater. 2023;35 doi: 10.1002/adma.202304123. [DOI] [PubMed] [Google Scholar]
- 102.Zou Y., Sun Y., Wang Y., Zhang D., Yang H., Wang X., Zheng M., Shi B. Cancer cell-mitochondria hybrid membrane coated Gboxin loaded nanomedicines for glioblastoma treatment. Nat. Commun. 2023;14:4557. doi: 10.1038/s41467-023-40280-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Chu D., Dong X., Shi X., Zhang C., Wang Z. Neutrophil-based drug delivery systems. Adv. Mater. 2018;30 doi: 10.1002/adma.201706245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Tietjen G.T., Bracaglia L.G., Saltzman W.M., Pober J.S. Focus on fundamentals: achieving effective nanoparticle targeting, trends. Mol. Med. 2018;24:598–606. doi: 10.1016/j.molmed.2018.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Wang Q., Song Y., Chen J., Li Q., Gao J., Tan H., Zhu Y., Wang Z., Li M., Yang H., Zhang N., Li X., Qian J., Pang Z., Huang Z., Ge J. Direct in vivo reprogramming with non-viral sequential targeting nanoparticles promotes cardiac regeneration. Biomaterials. 2021;276 doi: 10.1016/j.biomaterials.2021.121028. [DOI] [PubMed] [Google Scholar]
- 106.Li M., Xu Z., Zhang L., Cui M., Zhu M., Guo Y., Sun R., Han J., Song E., He Y., Su Y. Targeted noninvasive treatment of choroidal neovascularization by hybrid cell-membrane-cloaked biomimetic nanoparticles. ACS Nano. 2021;15:9808–9819. doi: 10.1021/acsnano.1c00680. [DOI] [PubMed] [Google Scholar]
- 107.Hu C.M., Fang R.H., Copp J., Luk B.T., Zhang L. A biomimetic nanosponge that absorbs pore-forming toxins. Nat. Nanotechnol. 2013;8:336–340. doi: 10.1038/nnano.2013.54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Davis M.E., Chen Z.G., Shin D.M. Nanoparticle therapeutics: an emerging treatment modality for cancer. Nat. Rev. Drug Discov. 2008;7:771–782. doi: 10.1038/nrd2614. [DOI] [PubMed] [Google Scholar]
- 109.Peer D., Karp J.M., Hong S., Farokhzad O.C., Margalit R., Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat. Nanotechnol. 2007;2:751–760. doi: 10.1038/nnano.2007.387. [DOI] [PubMed] [Google Scholar]
- 110.Sun T., Zhang Y.S., Pang B., Hyun D.C., Yang M., Xia Y. Engineered nanoparticles for drug delivery in cancer therapy. Angew Chem. Int. Ed. Engl. 2014;53:12320–12364. doi: 10.1002/anie.201403036. [DOI] [PubMed] [Google Scholar]
- 111.Yu M., Zheng J. Clearance pathways and tumor targeting of imaging nanoparticles. ACS Nano. 2015;9:6655–6674. doi: 10.1021/acsnano.5b01320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Zhang Y.N., Poon W., Tavares A.J., McGilvray I.D., Chan W.C.W. Nanoparticle-liver interactions: cellular uptake and hepatobiliary elimination. J Control Release. 2016;240:332–348. doi: 10.1016/j.jconrel.2016.01.020. [DOI] [PubMed] [Google Scholar]
- 113.Zhu M., Perrett S., Nie G. Understanding the particokinetics of engineered nanomaterials for safe and effective therapeutic applications. Small. 2013;9:1619–1634. doi: 10.1002/smll.201201630. [DOI] [PubMed] [Google Scholar]
- 114.Li M., Panagi Z., Avgoustakis K., Reineke J. Physiologically based pharmacokinetic modeling of PLGA nanoparticles with varied mPEG content. Int. J. Nanomedicine. 2012;7:1345–1356. doi: 10.2147/IJN.S23758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Oldenborg P.A., Zheleznyak A., Fang Y.F., Lagenaur C.F., Gresham H.D., Lindberg F.P. Role of CD47 as a marker of self on red blood cells. Science. 2000;288:2051–2054. doi: 10.1126/science.288.5473.2051. [DOI] [PubMed] [Google Scholar]
- 116.Barclay A.N., Van den Berg T.K. The interaction between signal regulatory protein alpha (SIRPα) and CD47: structure, function, and therapeutic target. Annu. Rev. Immunol. 2014;32:25–50. doi: 10.1146/annurev-immunol-032713-120142. [DOI] [PubMed] [Google Scholar]
- 117.Liao Y., Zhang Y., Blum N.T., Lin J., Huang P. Biomimetic hybrid membrane-based nanoplatforms: synthesis, properties and biomedical applications. Nanoscale. Horiz. 2020;5:1293–1302. doi: 10.1039/d0nh00267d. [DOI] [PubMed] [Google Scholar]
- 118.Ross K.A., Brenza T.M., Binnebose A.M., Phanse Y., Kanthasamy A.G., Gendelman H.E., Salem A.K., Bartholomay L.C., Bellaire B.H., Narasimhan B. Nano-enabled delivery of diverse payloads across complex biological barriers. J. Control. Release. 2015;219:548–559. doi: 10.1016/j.jconrel.2015.08.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Xiong J., Wu M., Chen J., Liu Y., Chen Y., Fan G., Liu Y., Cheng J., Wang Z., Wang S., Liu Y., Zhang W. Cancer-erythrocyte hybrid membrane-camouflaged magnetic nanoparticles with enhanced photothermal-immunotherapy for ovarian cancer. ACS Nano. 2021;15:19756–19770. doi: 10.1021/acsnano.1c07180. [DOI] [PubMed] [Google Scholar]
- 120.Lambert V., Lecomte J., Hansen S., Blacher S., Gonzalez M.L., Struman I., Sounni N.E., Rozet E., de Tullio P., Foidart J.M., Rakic J.M., Noel A. Laser-induced choroidal neovascularization model to study age-related macular degeneration in mice. Nat. Protoc. 2013;8:2197–2211. doi: 10.1038/nprot.2013.135. [DOI] [PubMed] [Google Scholar]
- 121.André H., Tunik S., Aronsson M., Kvanta A. Hypoxia-inducible factor-1α is associated with sprouting angiogenesis in the murine laser-induced choroidal neovascularization model. Investig. Ophthalmol. Vis. Sci. 2015;56:6591–6604. doi: 10.1167/iovs.15-16476. [DOI] [PubMed] [Google Scholar]
- 122.Liu Y., Wang Y., Yang Y., Weng L., Wu Q., Zhang J., Zhao P., Fang L., Shi Y., Wang P. Emerging phagocytosis checkpoints in cancer immunotherapy. Signal Transduct. Target Ther. 2023;8:104. doi: 10.1038/s41392-023-01365-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Séhédic D., Chourpa I., Tétaud C., Griveau A., Loussouarn C., Avril S., Legendre C., Lepareur N., Wion D., Hindré F., Davodeau F., Garcion E. Locoregional confinement and major clinical benefit of (188)Re-loaded CXCR4-targeted nanocarriers in an orthotopic human to mouse model of glioblastoma. Theranostics. 2017;7:4517–4536. doi: 10.7150/thno.19403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Pozzi S., Satchi-Fainaro R. The role of CCL2/CCR2 axis in cancer and inflammation: the next frontier in nanomedicine. Adv. Drug Deliv. Rev. 2024;209 doi: 10.1016/j.addr.2024.115318. [DOI] [PubMed] [Google Scholar]
- 125.Rodriguez P.L., Harada T., Christian D.A., Pantano D.A., Tsai R.K., Discher D.E. Minimal "Self" peptides that inhibit phagocytic clearance and enhance delivery of nanoparticles. Science. 2013;339:971–975. doi: 10.1126/science.1229568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Dehaini D., Fang R.H., Zhang L. Biomimetic strategies for targeted nanoparticle delivery. Bioeng. Transl. Med. 2016;1:30–46. doi: 10.1002/btm2.10004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Song Y., Huang Z., Liu X., Pang Z., Chen J., Yang H., Zhang N., Cao Z., Liu M., Cao J., Li C., Yang X., Gong H., Qian J., Ge J. Platelet membrane-coated nanoparticle-mediated targeting delivery of Rapamycin blocks atherosclerotic plaque development and stabilizes plaque in apolipoprotein E-deficient (ApoE(-/-)) mice. Nanomedicine. 2019;15:13–24. doi: 10.1016/j.nano.2018.08.002. [DOI] [PubMed] [Google Scholar]
- 128.Chen H.Y., Deng J., Wang Y., Wu C.Q., Li X., Dai H.W. Hybrid cell membrane-coated nanoparticles: a multifunctional biomimetic platform for cancer diagnosis and therapy. Acta Biomater. 2020;112:1–13. doi: 10.1016/j.actbio.2020.05.028. [DOI] [PubMed] [Google Scholar]
- 129.Nahrendorf M., Swirski F.K., Aikawa E., Stangenberg L., Wurdinger T., Figueiredo J.L., Libby P., Weissleder R., Pittet M.J. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J. Exp. Med. 2007;204:3037–3047. doi: 10.1084/jem.20070885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Imhof B.A., Aurrand-Lions M. Adhesion mechanisms regulating the migration of monocytes. Nat. Rev. Immunol. 2004;4:432–444. doi: 10.1038/nri1375. [DOI] [PubMed] [Google Scholar]
- 131.Lee C., Mitsialis S.A., Aslam M., Vitali S.H., Vergadi E., Konstantinou G., Sdrimas K., Fernandez-Gonzalez A., Kourembanas S. Exosomes mediate the cytoprotective action of mesenchymal stromal cells on hypoxia-induced pulmonary hypertension. Circulation. 2012;126:2601–2611. doi: 10.1161/CIRCULATIONAHA.112.114173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Li J., Sun S., Zhu D., Mei X., Lyu Y., Huang K., Li Y., Liu S., Wang Z., Hu S., Lutz H.J., Popowski K.D., Dinh P.C., Butte A.J., Cheng K. Inhalable stem cell exosomes promote heart repair after myocardial infarction. Circulation. 2024;150:710–723. doi: 10.1161/CIRCULATIONAHA.123.065005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Akbar N., Razzaq S.S., Salim A., Haneef K. Mesenchymal stem cell-derived exosomes and their MicroRNAs in heart repair and regeneration. J. Cardiovasc. Transl. Res. 2024;17:505–522. doi: 10.1007/s12265-023-10449-8. [DOI] [PubMed] [Google Scholar]
- 134.Sahoo S., Losordo D.W. Exosomes and cardiac repair after myocardial infarction. Circ. Res. 2014;114:333–344. doi: 10.1161/CIRCRESAHA.114.300639. [DOI] [PubMed] [Google Scholar]
- 135.Zhu D., Liu S., Huang K., Wang Z., Hu S., Li J., Li Z., Cheng K. Intrapericardial exosome therapy dampens cardiac injury via activating Foxo3. Circ. Res. 2022;131:e135–e150. doi: 10.1161/CIRCRESAHA.122.321384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Yan Y., Li K., Jiang J., Jiang L., Ma X., Ai F., Qiu S., Si W. Perinatal tissue-derived exosomes ameliorate colitis in mice by regulating the Foxp3 + Treg cells and gut microbiota. Stem Cell Res. Ther. 2023;14:43. doi: 10.1186/s13287-023-03263-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hu S., Li Z., Cores J., Huang K., Su T., Dinh P.U., Cheng K. Needle-free injection of exosomes derived from human dermal fibroblast spheroids ameliorates skin photoaging. ACS Nano. 2019;13:11273–11282. doi: 10.1021/acsnano.9b04384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Dinh P.C., Paudel D., Brochu H., Popowski K.D., Gracieux M.C., Cores J., Huang K., Hensley M.T., Harrell E., Vandergriff A.C., George A.K., Barrio R.T., Hu S., Allen T.A., Blackburn K., Caranasos T.G., Peng X., Schnabel L.V., Adler K.B., Lobo L.J., Goshe M.B., Cheng K. Inhalation of lung spheroid cell secretome and exosomes promotes lung repair in pulmonary fibrosis. Nat. Commun. 2020;11:1064. doi: 10.1038/s41467-020-14344-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Valadi H., Ekström K., Bossios A., Sjöstrand M., Lee J.J., Lötvall J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007;9:654–659. doi: 10.1038/ncb1596. [DOI] [PubMed] [Google Scholar]
- 140.Zhang W., Wang L., Guo H., Chen L., Huang X. Dapagliflozin-loaded exosome mimetics facilitate diabetic wound healing by HIF-1α-Mediated enhancement of angiogenesis. Adv. Healthc. Mater. 2023;12 doi: 10.1002/adhm.202202751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Qiao L., Hu S., Huang K., Su T., Li Z., Vandergriff A., Cores J., Dinh P.U., Allen T., Shen D., Liang H., Li Y., Cheng K. Tumor cell-derived exosomes home to their cells of origin and can be used as Trojan horses to deliver cancer drugs. Theranostics. 2020;10:3474–3487. doi: 10.7150/thno.39434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Qiao L., Hu S., Liu S., Zhang H., Ma H., Huang K., Li Z., Su T., Vandergriff A., Tang J., Allen T., Dinh P.U., Cores J., Yin Q., Li Y., Cheng K. microRNA-21-5p dysregulation in exosomes derived from heart failure patients impairs regenerative potential. J. Clin. Investig. 2019;129:2237–2250. doi: 10.1172/JCI123135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Vandergriff A., Huang K., Shen D., Hu S., Hensley M.T., Caranasos T.G., Qian L., Cheng K. Targeting regenerative exosomes to myocardial infarction using cardiac homing peptide. Theranostics. 2018;8:1869–1878. doi: 10.7150/thno.20524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Xin L., Yuan Y.W., Liu C., Zhou L.Q., Liu L., Zhou Q., Li S.H. Preparation of internalizing RGD-modified recombinant methioninase exosome active targeting vector and antitumor effect evaluation. Dig. Dis. Sci. 2021;66:1045–1053. doi: 10.1007/s10620-020-06262-x. [DOI] [PubMed] [Google Scholar]
- 145.Ala M. The beneficial effects of mesenchymal stem cells and their exosomes on myocardial infarction and critical considerations for enhancing their efficacy. Ageing Res. Rev. 2023;89 doi: 10.1016/j.arr.2023.101980. [DOI] [PubMed] [Google Scholar]
- 146.Su T., Huang K., Ma H., Liang H., Dinh P.U., Chen J., Shen D., Allen T.A., Qiao L., Li Z., Hu S., Cores J., Frame B.N., Young A.T., Yin Q., Liu J., Qian L., Caranasos T.G., Brudno Y., Ligler F.S., Cheng K. Platelet-inspired nanocells for targeted heart repair after ischemia/reperfusion injury. Adv. Funct. Mater. 2019;29 doi: 10.1002/adfm.201803567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Wei X., Ying M., Dehaini D., Su Y., Kroll A.V., Zhou J., Gao W., Fang R.H., Chien S., Zhang L. Nanoparticle functionalization with platelet membrane enables multifactored biological targeting and detection of atherosclerosis. ACS Nano. 2018;12:109–116. doi: 10.1021/acsnano.7b07720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Xu X., Zhang X., Li R., Yang X., Fu P., Feng R., Sun X., Wang Z., Yu J., Cao X., Yu Q., Wang Q., Liu S., Yang X., Zhu Y., Shi W., Deng W. Platelet membrane-coated curcumin-PLGA nanoparticles promote astrocyte-neuron transdifferentiation for intracerebral hemorrhage treatment. Small. 2024;20 doi: 10.1002/smll.202311128. [DOI] [PubMed] [Google Scholar]
- 149.Luczak E.D., Wu Y., Granger J.M., Joiner M.A., Wilson N.R., Gupta A., Umapathi P., Murphy K.R., Reyes Gaido O.E., Sabet A., Corradini E., Tseng W.W., Wang Y., Heck A.J.R., Wei A.C., Weiss R.G., Anderson M.E. Mitochondrial CaMKII causes adverse metabolic reprogramming and dilated cardiomyopathy. Nat. Commun. 2020;11:4416. doi: 10.1038/s41467-020-18165-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Heusch G. Myocardial ischaemia-reperfusion injury and cardioprotection in perspective. Nat. Rev. Cardiol. 2020;17:773–789. doi: 10.1038/s41569-020-0403-y. [DOI] [PubMed] [Google Scholar]
- 151.Prabhu S.D., Frangogiannis N.G. The biological basis for cardiac repair after myocardial infarction: from inflammation to fibrosis. Circ. Res. 2016;119:91–112. doi: 10.1161/CIRCRESAHA.116.303577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Gao F., Kataoka M., Liu N., Liang T., Huang Z.P., Gu F., Ding J., Liu J., Zhang F., Ma Q., Wang Y., Zhang M., Hu X., Kyselovic J., Hu X., Pu W.T., Wang J., Chen J., Wang D.Z. Therapeutic role of miR-19a/19b in cardiac regeneration and protection from myocardial infarction. Nat. Commun. 2019;10:1802. doi: 10.1038/s41467-019-09530-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Wittrup A., Lieberman J. Knocking down disease: a progress report on siRNA therapeutics. Nat. Rev. Genet. 2015;16:543–552. doi: 10.1038/nrg3978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Sager H.B., Dutta P., Dahlman J.E., Hulsmans M., Courties G., Sun Y., Heidt T., Vinegoni C., Borodovsky A., Fitzgerald K., Wojtkiewicz G.R., Iwamoto Y., Tricot B., Khan O.F., Kauffman K.J., Xing Y., Shaw T.E., Libby P., Langer R., Weissleder R., Swirski F.K., Anderson D.G., Nahrendorf M. RNAi targeting multiple cell adhesion molecules reduces immune cell recruitment and vascular inflammation after myocardial infarction. Sci. Transl. Med. 2016;8 doi: 10.1126/scitranslmed.aaf1435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Xia W., Singh N., Goel S., Shi S. Molecular imaging of innate immunity and immunotherapy. Adv. Drug Deliv. Rev. 2023;198 doi: 10.1016/j.addr.2023.114865. [DOI] [PubMed] [Google Scholar]
- 156.Tang J., Su T., Huang K., Dinh P.U., Wang Z., Vandergriff A., Hensley M.T., Cores J., Allen T., Li T., Sproul E., Mihalko E., Lobo L.J., Ruterbories L., Lynch A., Brown A., Caranasos T.G., Shen D., Stouffer G.A., Gu Z., Zhang J., Cheng K. Targeted repair of heart injury by stem cells fused with platelet nanovesicles. Nat. Biomed. Eng. 2018;2:17–26. doi: 10.1038/s41551-017-0182-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Wan M., Wang Q., Wang R., Wu R., Li T., Fang D., Huang Y., Yu Y., Fang L., Wang X., Zhang Y., Miao Z., Zhao B., Wang F., Mao C., Jiang Q., Xu X., Shi D. Platelet-derived porous nanomotor for thrombus therapy. Sci. Adv. 2020;6 doi: 10.1126/sciadv.aaz9014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Han H., Bártolo R., Li J., Shahbazi M.A., Santos H.A. Biomimetic platelet membrane-coated nanoparticles for targeted therapy. Eur. J. Pharm. Biopharm. 2022;172:1–15. doi: 10.1016/j.ejpb.2022.01.004. [DOI] [PubMed] [Google Scholar]
- 159.Gao C., Huang Q., Liu C., Kwong C.H.T., Yue L., Wan J.B., Lee S.M.Y., Wang R. Treatment of atherosclerosis by macrophage-biomimetic nanoparticles via targeted pharmacotherapy and sequestration of proinflammatory cytokines. Nat. Commun. 2020;11:2622. doi: 10.1038/s41467-020-16439-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Herrington W., Lacey B., Sherliker P., Armitage J., Lewington S. Epidemiology of atherosclerosis and the potential to reduce the global burden of atherothrombotic disease. Circ. Res. 2016;118:535–546. doi: 10.1161/CIRCRESAHA.115.307611. [DOI] [PubMed] [Google Scholar]
- 161.Chen J., Ren T., Xie L., Hu H., Li X., Maitusong M., Zhou X., Hu W., Xu D., Qian Y., Cheng S., Yu K., Wang J.A., Liu X. Enhancing aortic valve drug delivery with PAR2-targeting magnetic nano-cargoes for calcification alleviation. Nat. Commun. 2024;15:557. doi: 10.1038/s41467-024-44726-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Libby P. The changing landscape of atherosclerosis. Nature. 2021;592:524–533. doi: 10.1038/s41586-021-03392-8. [DOI] [PubMed] [Google Scholar]
- 163.De Meyer G.R.Y., Zurek M., Puylaert P., Martinet W. Programmed death of macrophages in atherosclerosis: mechanisms and therapeutic targets. Nat. Rev. Cardiol. 2024;21:312–325. doi: 10.1038/s41569-023-00957-0. [DOI] [PubMed] [Google Scholar]
- 164.Ma Y., Gou S., Zhu Z., Sun J., Shahbazi M.A., Si T., Xu C., Ru J., Shi X., Reis R.L., Kundu S.C., Ke B., Nie G., Xiao B. Transient mild photothermia improves therapeutic performance of oral nanomedicines with enhanced accumulation in the colitis mucosa. Adv. Mater. 2024;36 doi: 10.1002/adma.202309516. [DOI] [PubMed] [Google Scholar]
- 165.Kuznetsova T., Prange K.H.M., Glass C.K., de Winther M.P.J. Transcriptional and epigenetic regulation of macrophages in atherosclerosis. Nat. Rev. Cardiol. 2020;17:216–228. doi: 10.1038/s41569-019-0265-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Barrett T.J. Macrophages in atherosclerosis regression. Arterioscler. Thromb. Vasc. Biol. 2020;40:20–33. doi: 10.1161/ATVBAHA.119.312802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Gawaz M., Stellos K., Langer H.F. Platelets modulate atherogenesis and progression of atherosclerotic plaques via interaction with progenitor and dendritic cells. J. Thromb. Haemost. 2008;6:235–242. doi: 10.1111/j.1538-7836.2008.02867.x. [DOI] [PubMed] [Google Scholar]
- 168.Lindemann S., Krämer B., Seizer P., Gawaz M. Platelets, inflammation and atherosclerosis. J. Thromb. Haemost. 2007;5(Suppl 1):203–211. doi: 10.1111/j.1538-7836.2007.02517.x. [DOI] [PubMed] [Google Scholar]
- 169.Sica A., Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J. Clin. Investig. 2012;122:787–795. doi: 10.1172/JCI59643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Suzuki J., Denning D.P., Imanishi E., Horvitz H.R., Nagata S. Xk-related protein 8 and CED-8 promote phosphatidylserine exposure in apoptotic cells. Science. 2013;341:403–406. doi: 10.1126/science.1236758. [DOI] [PubMed] [Google Scholar]
- 171.Cai B., Thorp E.B., Doran A.C., Sansbury B.E., Daemen M.J., Dorweiler B., Spite M., Fredman G., Tabas I. MerTK receptor cleavage promotes plaque necrosis and defective resolution in atherosclerosis. J. Clin. Investig. 2017;127:564–568. doi: 10.1172/JCI90520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Fourgeaud L., Través P.G., Tufail Y., Leal-Bailey H., Lew E.D., Burrola P.G., Callaway P., Zagórska A., Rothlin C.V., Nimmerjahn A., Lemke G. TAM receptors regulate multiple features of microglial physiology. Nature. 2016;532:240–244. doi: 10.1038/nature17630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Bosurgi L., Cao Y.G., Cabeza-Cabrerizo M., Tucci A., Hughes L.D., Kong Y., Weinstein J.S., Licona-Limon P., Schmid E.T., Pelorosso F., Gagliani N., Craft J.E., Flavell R.A., Ghosh S., Rothlin C.V. Macrophage function in tissue repair and remodeling requires IL-4 or IL-13 with apoptotic cells. Science. 2017;356:1072–1076. doi: 10.1126/science.aai8132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Wang Y., Zhang K., Li T., Maruf A., Qin X., Luo L., Zhong Y., Qiu J., McGinty S., Pontrelli G., Liao X., Wu W., Wang G. Macrophage membrane functionalized biomimetic nanoparticles for targeted anti-atherosclerosis applications. Theranostics. 2021;11:164–180. doi: 10.7150/thno.47841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Liam-Or R., Faruqu F.N., Walters A., Han S., Xu L., Wang J.T., Oberlaender J., Sanchez-Fueyo A., Lombardi G., Dazzi F., Mailaender V., Al-Jamal K.T. Cellular uptake and in vivo distribution of mesenchymal-stem-cell-derived extracellular vesicles are protein corona dependent. Nat. Nanotechnol. 2024;19:846–855. doi: 10.1038/s41565-023-01585-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Lu H., Wang Y., Yu R. Immune cell membrane-coated nanoparticles for targeted myocardial ischemia/reperfusion injury therapy. Innovat. Med. 2023;1 [Google Scholar]
- 177.Wu J., Liu S., Banerjee O., Shi H., Xue B., Ding Z. Disturbed flow impairs MerTK-mediated efferocytosis in aortic endothelial cells during atherosclerosis. Theranostics. 2024;14:2427–2441. doi: 10.7150/thno.93036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Flores A.M., Hosseini-Nassab N., Jarr K.U., Ye J., Zhu X., Wirka R., Koh A.L., Tsantilas P., Wang Y., Nanda V., Kojima Y., Zeng Y., Lotfi M., Sinclair R., Weissman I.L., Ingelsson E., Smith B.R., Leeper N.J. Pro-efferocytic nanoparticles are specifically taken up by lesional macrophages and prevent atherosclerosis. Nat. Nanotechnol. 2020;15:154–161. doi: 10.1038/s41565-019-0619-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Wang X., Xia Z., Wang H., Wang D., Sun T., Hossain E., Pang X., Liu Y. Cell-membrane-coated nanoparticles for the fight against pathogenic bacteria, toxins, and inflammatory cytokines associated with sepsis. Theranostics. 2023;13:3224–3244. doi: 10.7150/thno.81520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Tao C., Nie X., Zhu W., Iqbal J., Xu C., Wang D.A. Autologous cell membrane coatings on tissue engineering xenografts for suppression and alleviation of acute host immune responses. Biomaterials. 2020;258 doi: 10.1016/j.biomaterials.2020.120310. [DOI] [PubMed] [Google Scholar]
- 181.Liu S., Li Z., Yu B., Wang S., Shen Y., Cong H. Recent advances on protein separation and purification methods. Adv. Colloid Interface Sci. 2020;284 doi: 10.1016/j.cis.2020.102254. [DOI] [PubMed] [Google Scholar]
- 182.Hou M., Wu X., Zhao Z., Deng Q., Chen Y., Yin L. Endothelial cell-targeting, ROS-ultrasensitive drug/siRNA co-delivery nanocomplexes mitigate early-stage neutrophil recruitment for the anti-inflammatory treatment of myocardial ischemia reperfusion injury. Acta Biomater. 2022;143:344–355. doi: 10.1016/j.actbio.2022.02.018. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No data was used for the research described in the article.











