14
Recent Advances of Membrane-Cloaked Nanoplatforms for Biomedical Applications Xiangzhao Ai, Ming Hu, Zhimin Wang, Wenmin Zhang, ,Juan Li, Huanghao Yang, Jun Lin, § and Bengang Xing* ,,Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore, 637371 College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China § State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China ABSTRACT: In terms of the extremely small size and large specic surface area, nanomaterials often exhibit unusual physical and chemical properties, which have recently attracted considerable attention in bionanotechnology and nanomedicine. Currently, the extensive usage of nanotechnology in medicine holds great potential for precise diagnosis and eective therapeutics of various human diseases in clinical practice. However, a detailed understanding regarding how nanomedicine interacts with the intricate environment in complex living systems remains a pressing and challenging goal. Inspired by the diversied membrane structures and functions of natural prototypes, research activities on biomimetic and bioinspired membranes, especially for those cloaking nanosized platforms, have increased exponentially. By taking advantage of the exible synthesis and multiple functionality of nanomaterials, a variety of unique nanostructures including inorganic nanocrystals and organic polymers have been widely devised to substantially integrate with intrinsic biomoieties such as lipids, glycans, and even cell and bacteria membrane components, which endow these abiotic nanomaterials with specic biological functionalities for the purpose of detailed investigation of the complicated interactions and activities of nanomedicine in living bodies, including their immune response activation, phagocytosis escape, and subsequent clearance from vascular system. In this review, we summarize the strategies established recently for the development of biomimetic membrane-cloaked nanoplatforms derived from inherent host cells (e.g., erythrocytes, leukocytes, platelets, and exosomes) and invasive pathogens (e.g., bacteria and viruses), mainly attributed to their versatile membrane properties in biological uids. Meanwhile, the promising biomedical applications based on nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and eective vaccine development for disease prevention, have also been outlined. Finally, the potential challenges and future prospects of the biomimetic membrane- cloaked nanoplatforms are also discussed. INTRODUCTION Currently, the remarkable progress of nanomedicine based on extensive usage of nanotechnology has received considerable interest for its potential in precise diagnosis and eective therapeutics of various diseases including cancer, cardiovascular disease, as well as neurological disorders, 16 mostly owing to the unique physical and chemical properties of diverse nanomaterials in terms of the nanoscale size eect and high surface-to-volume ratio. 712 Despite the continuous progress in recent decades, a detailed understanding regarding how nanomedicine interacts with the intricate environment in living systems still remains a pressing and challenging goal. 1315 Many signicant eects of structures in nanomedicine designed for their unique bioactivity and function, especially for their dynamic transport pathways in the vascular system, clearance, phagocytic immune-mediated degradation, as well as the potential binding sites of nanomedicine in living bodies, have not yet been thoroughly elucidated. 1618 To this end, the well- designed nanoplatforms are highly demanded as advanced biotechnological tools to fully understand the detailed behaviors of nanomedicine in complicated physiopathological processes including disease surveillance, sensitive diagnosis, and targeted therapeutics. 1921 In fact, there are plenty of native prototypes (e.g., cells, virus, bacteria, etc.) with inherent properties to regulate diverse biological processes (e.g., growth, metabolism, immunity, etc.), which serve as a major source to motivate us in constructing biomimetic nanostructures for the investigation of nano- medicine bioactivities in vivo. 2224 Particularly, inspired by the diversied membrane structures and functions of these Special Issue: Biomimetic Materials Received: February 10, 2018 Revised: March 2, 2018 Published: March 6, 2018 Review pubs.acs.org/bc Cite This: Bioconjugate Chem. 2018, 29, 838-851 © 2018 American Chemical Society 838 DOI: 10.1021/acs.bioconjchem.8b00103 Bioconjugate Chem. 2018, 29, 838851

Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

Recent Advances of Membrane-Cloaked Nanoplatforms forBiomedical ApplicationsXiangzhao Ai,† Ming Hu,† Zhimin Wang,† Wenmin Zhang,†,‡ Juan Li,‡ Huanghao Yang,‡ Jun Lin,§

and Bengang Xing*,†,‡

†Division of Chemistry and Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University,Singapore, 637371‡College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China§State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences,Changchun, 130022, China

ABSTRACT: In terms of the extremely small size and large specific surfacearea, nanomaterials often exhibit unusual physical and chemical properties,which have recently attracted considerable attention in bionanotechnology andnanomedicine. Currently, the extensive usage of nanotechnology in medicineholds great potential for precise diagnosis and effective therapeutics of varioushuman diseases in clinical practice. However, a detailed understanding regardinghow nanomedicine interacts with the intricate environment in complex livingsystems remains a pressing and challenging goal. Inspired by the diversifiedmembrane structures and functions of natural prototypes, research activities onbiomimetic and bioinspired membranes, especially for those cloaking nanosizedplatforms, have increased exponentially. By taking advantage of the flexiblesynthesis and multiple functionality of nanomaterials, a variety of uniquenanostructures including inorganic nanocrystals and organic polymers havebeen widely devised to substantially integrate with intrinsic biomoieties such as lipids, glycans, and even cell and bacteriamembrane components, which endow these abiotic nanomaterials with specific biological functionalities for the purpose ofdetailed investigation of the complicated interactions and activities of nanomedicine in living bodies, including their immuneresponse activation, phagocytosis escape, and subsequent clearance from vascular system. In this review, we summarize thestrategies established recently for the development of biomimetic membrane-cloaked nanoplatforms derived from inherent hostcells (e.g., erythrocytes, leukocytes, platelets, and exosomes) and invasive pathogens (e.g., bacteria and viruses), mainly attributedto their versatile membrane properties in biological fluids. Meanwhile, the promising biomedical applications based onnanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective vaccine development fordisease prevention, have also been outlined. Finally, the potential challenges and future prospects of the biomimetic membrane-cloaked nanoplatforms are also discussed.

■ INTRODUCTION

Currently, the remarkable progress of nanomedicine based onextensive usage of nanotechnology has received considerableinterest for its potential in precise diagnosis and effectivetherapeutics of various diseases including cancer, cardiovasculardisease, as well as neurological disorders,1−6 mostly owing tothe unique physical and chemical properties of diversenanomaterials in terms of the nanoscale size effect and highsurface-to-volume ratio.7−12 Despite the continuous progress inrecent decades, a detailed understanding regarding hownanomedicine interacts with the intricate environment in livingsystems still remains a pressing and challenging goal.13−15

Many significant effects of structures in nanomedicine designedfor their unique bioactivity and function, especially for theirdynamic transport pathways in the vascular system, clearance,phagocytic immune-mediated degradation, as well as thepotential binding sites of nanomedicine in living bodies, havenot yet been thoroughly elucidated.16−18 To this end, the well-

designed nanoplatforms are highly demanded as advancedbiotechnological tools to fully understand the detailedbehaviors of nanomedicine in complicated physiopathologicalprocesses including disease surveillance, sensitive diagnosis, andtargeted therapeutics.19−21

In fact, there are plenty of native prototypes (e.g., cells, virus,bacteria, etc.) with inherent properties to regulate diversebiological processes (e.g., growth, metabolism, immunity, etc.),which serve as a major source to motivate us in constructingbiomimetic nanostructures for the investigation of nano-medicine bioactivities in vivo.22−24 Particularly, inspired bythe diversified membrane structures and functions of these

Special Issue: Biomimetic Materials

Received: February 10, 2018Revised: March 2, 2018Published: March 6, 2018

Review

pubs.acs.org/bcCite This: Bioconjugate Chem. 2018, 29, 838−851

© 2018 American Chemical Society 838 DOI: 10.1021/acs.bioconjchem.8b00103Bioconjugate Chem. 2018, 29, 838−851

Page 2: Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

natural biomoieties, research activity on biomimetic mem-branes, especially for those cloaking nanosized platforms, hasincreased exponentially in recent decades.25−28 So far, on thebasis of the flexible synthesis and multiple functionality ofnanomaterials, a variety of unique nanostructures includinginorganic nanocrystals (e.g., gold nanoparticles (AuNPs), etc.)and organic polymers (e.g., poly(lactic-co-glycolic acid)(PLGA) nanoparticles, etc.) have been widely devised tosubstantially integrate with intrinsic biomoieties such as lipids,glycans, and even cell and bacteria surface components.29−31

These hybridized nanoplatforms endow the abiotic nanoma-terials with specific biological functionalities for the explorationof complicated interactions and activities of nanomedicine inliving conditions, including immune response activation,phagocytosis escape, and subsequent clearance from thevascular system.32,33 Until now, various membrane-mimickingnanoplatforms based on several inherent host cells in biologicalfluids (e.g., erythrocytes, leukocytes, platelets, etc.) have beendeveloped to mimic the cell membrane function during manyessential physiological processes such as the specific cell−cellinteractions, intercellular recognition, adhesion, as well ascommunication.34−36 Moreover, considering the specificmembrane immunogenic antigens on various bacteria andviruses, the pathogen-mimicking nanoplatforms are alsoemerging as versatile vehicles to study the complex relation-ships between host immune system and invasive pathogens.37,38

In this review, we summarize recent advances of membrane-cloaked nanoplatforms to mimic the natural entities inbiological fluids ranging from inherent host cells (e.g.,erythrocytes, leukocytes, platelets, and exosomes) to invasivepathogens (e.g., bacteria and viruses) based on cell-membrane-mimicking and pathogen-mimicking strategies (Figure 1).

These approaches provide excellent means to in-depthexploration of the detailed information regarding how nano-medicine interacts with the surrounding environment and howto optimize their structures for improved theranostics in vivo.Last but not least, we also discuss the potential challenges andprospectives of these biomimetic membrane-cloaked nanoplat-forms for future development.

1. ERYTHROCYTE-DERIVED NANOPLATFORMS

Typically, the specific interactions of nanomedicine in manyphysiological processes play significant roles in their biologicalactivities and therapeutic outcomes in living systems, includinghow to escape undesired phagocytosis and clearance by thehost immune system and how to localize at the targetedpathological regions without side effects.39−41 In line with thesefactors, the erythrocytes, commonly known as red blood cells(RBCs), may act as a valuable model for nanomedicine toexplore and mimic their specific properties in vivo.42−44

Normally, RBCs are capable of serving as oxygen carriersthroughout the body with prolonged circulation time (∼120days) in the vascular system.45 Moreover, the RBCs can easilyescape the phagocytic immune cell-controlled clearance anddegradation through the expression of several biomarkers onthe cell membrane including “don’t eat me” marker CD47 andsignal-regulatory protein α (SIRPα) receptors.46 Such remark-able properties of RBCs suggest a promising strategy allowingtraditional nanoparticles to achieve long circulation time andspecific membrane functions for potential utilization in livinganimals.47,48 For example, RBCs-mimicking nanoparticles havebeen designed by Zhang’s group for the development of novelbiomimetic and long-circulating nanoplatforms based on theirgreat biocompatibility and limited immunogenicity.49 Theyprovided a smart strategy to fabricate RBCs membrane-camouflaged nanoparticles (RBCs-NPs) in two steps: RBCsmembrane vesicle extrusion and vesicle−nanoparticle fusion(Figure 2a). Briefly, the isolated RBCs from whole bloodunderwent membrane rupture using hypotonic treatment toremove their intracellular components, and the emptied RBCswere washed and extruded through porous membranes tocreate erythrocyte-derived vesicles. The final core−shellstructure of RBCs-NPs was achieved by fusing the RBCvesicles with carboxyl-terminated PLGA nanoparticles viamechanical extrusion.50 Moreover, similar levels of proteincontent and expression of CD47 were demonstrated on RBC-NPs compared with native erythrocytes, and superiorcirculation half-life (39.6 h) was also achieved for RBC-NPsthan that in conventional polyethylene glycol (PEG) modifiednanoparticles (15.8 h) in mice. These results strongly indicatedthat RBCs-NPs could effectively prolong the circulation time inblood and mimic the specific membrane functions in livingconditions.Furthermore, toward the biomedical applications of eryth-

rocyte-derived nanoparticles, excellent selectivity is anotherdesirable feature that promises minimization of off-target sideeffects for effective disease diagnosis and therapeutics.51 So far,various chemical functionalization methods have beenemployed to modify nanoparticles with targeting ligands fortheir specific binding with overexpressed antigens (e.g.,carbohydrates, proteins, etc.) on the cell membranes at diseasedsites.52−54 In order to nondisruptively integrate targetingligands on the surface of RBCs-NPs, a lipid insertion strategy,which tethers targeting ligands to lipid molecules for RBCmembrane insertion, was recently developed based on theintrinsic fluidity and dynamic conformation of the phospholipidbilayer of cell membrane (Figure 2b).55 This approach couldnot only allow for the membrane functionalization of varioustargeting ligands at different molecular weights from small-molecule folate (441 Da) to macromolecule nucleolin-targetingaptamer AS1411 (9000 Da), but also achieve the adjustabilityof ligand density by controlling the lipid-tethered ligand input,

Figure 1. Development of biomimetic membrane-cloaked nanoplat-forms inspired by the natural entities in biological fluids ranging frominherent host cellular structures (erythrocytes, leukocytes, platelets,and exosomes) to invasive pathogens (bacteria and viruses).

Bioconjugate Chemistry Review

DOI: 10.1021/acs.bioconjchem.8b00103Bioconjugate Chem. 2018, 29, 838−851

839

Page 3: Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

which holds great promise to improve the selectivity ofbiomimetic nanoplatforms with reduced off-target side effects.Encouraged by these promising pioneer studies, similar RBC-

membrane-derived approaches have been applied in manyother nanostructures including polymer nanoparticles,56

AuNPs,57 mesoporous silica nanoparticles,44 upconversionnanocrystals,58 and magnetic nanomaterials.59 All these RBCs-NPs hold unique capabilities to evade macrophage uptake andavoid immune clearance in living systems. Interestingly, bytaking advantage of their specific membrane−antigen inter-action, RBCs-NPs have recently been employed as abiomimetic nanosponge to clear poisonous pathologicalantibodies and toxins in vivo.60−62 For instance, Zhang et al.demonstrated that RBCs-NPs could act as nanosponges toarrest membrane damaging staphylococcal alpha-hemolysin (α-toxin) in the bloodstream and to divert them away from theircellular targets.60 In a mouse model, the nanosponges couldprevent toxin-mediated hemolysis and reduce their toxicity byneutralization, which exhibited remarkable improvement insurvival rate of toxin-challenged mice (Figure 2c). Similarly,Zhang et al. also reported that the RBCs-NPs could abrogatethe effect of pathological antibody-induced anemia disease inwhich the immune system produces autoantibodies to attracthealthy erythrocytes.61 Different from the conventionalimmune suppression drugs, the RBCs-NPs could serve as analternative target for pathological antibodies to protect healthyerythrocytes from macrophage phagocytosis (Figure 2d). Theseinnovative studies clearly demonstrated that erythrocytemembrane-derived nanoparticles represented promising ther-apeutic nanoplatforms for the broad range of biomedicalapplications on the basis of their multifaceted interactions withinnate immune system in living animals.

2. LEUKOCYTE-DERIVED NANOPLATFORMS

Leukocytes, also known as white blood cells, are inherent cellsin the immune system that protect the living body against bothinfectious diseases and foreign invaders.63,64 When the bodytissues are damaged by infection or injury with inflammatoryresponse generation, leukocytes are recruited from thebloodstream to the inflammation sites to effectively kill thepathogens and remove them by phagocytosis.65,66 During thisphysiological process, the specific surface interactions betweenleukocytes and endothelia play crucial roles in the recruitmentof immune cells at the targeted disease regions, owing to theoverexpression of endothelial adhesion molecules (e.g.,integrins) to selectively bind with ligands expressed onleukocyte surfaces (such as selectins).67−69 Therefore, inorder to mimic the membrane functions of leukocytes, avariety of bioinspired nanoplatforms have been constructed toexplore the underlying mechanisms of leukocyte−endothelialinteractions during the inflammatory response.25,70−73 Basically,the initial investigations to endow nanoparticles with theintrinsic features of leukocytes mainly rely on the surfacefunctionalization with target ligands.72,74 For example, bymodification of the polymersome nanoparticle surface withspecific leukocytal carbohydrate ligands (sialyl Lewisx),Hammer et al. demonstrated that this promising leuko-polymersome could firmly adhere to cell surfaces coated withthe inflammatory adhesion molecules including P-selectin andactivated β2-integrin (LFA-1, Mac-1, ICAM-1),74 whichindicated significant effects to the kinetic and mechanicalproperties of leukocyte-endothelial rolling interactions.Despite the controllable physical and chemical parameters

(e.g., size, component, surface ligands functionalization,homogeneity, etc.) of the proposed strategy, it is still highlydemanding to reproduce the integrality and complexity of theleukocyte membrane.75,76 In recent years, researchers haveconsidered the possibility of efficient manipulation of the

Figure 2. Schematic illustration of erythrocyte-derived nanoplatforms. (a) RBCs-NPs fabrication procedures and TEM image. Scale bar: 50 nm. (b)Formation of targeted RBCs-NPs with lipid-tethered ligands. (c) Biomimetic nanosponges (right) and mechanism for neutralizing α-toxins (left).(d) Pathological antibodies opsonizing healthy RBCs for extravascular hemolysis via phagocytosis (left) and RBCs-NPs protecting RBCs byneutralizing antibodies (right). (Reprinted with permission from refs 49, 55, 60, and 61. Copyright 2011 and 2014 American Chemical Society, 2013Nature Publishing Group, and 2012 Royal Society of Chemistry.)

Bioconjugate Chemistry Review

DOI: 10.1021/acs.bioconjchem.8b00103Bioconjugate Chem. 2018, 29, 838−851

840

Page 4: Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

integrated leukocyte membrane to enable the transfer of severalsignificant leukocyte markers on the surface of nanoparticles,including superior endothelial adhesion molecules (e.g., LFA-1,Mac-1, etc.) and “self-recognition” proteins (e.g., CD45, CD47,etc.) for long circulation.77,78 For instance, Tasciotti et al.successfully integrated leukocyte plasma membrane onto ananoporous silicon (NPS) platform as hybrid leukocyte-likevectors (LLVs), which possessed specific leukocyte propertiesincluding biomarkers (CD45 and CD3z) and antigens (LFA-1or CD11a) on the vector surface (Figure 3a).77 Importantly,the promising LLVs have the potential to recognize andcommunicate with endothelial cells through receptor−ligandinteractions in an active and nondestructive manner (Figure3b), which could effectively improve the accumulation in tumorregions for further cancer therapy. Moreover, Zhang et al.demonstrated that grapefruit-derived nanovectors (GNV)coated with activated leukocyte membranes (IGNVs) couldsignificantly enhance their endothelial cell transmigrationcapability at inflammatory sites, and further effectively inhibittumor growth after encapsulation of doxorubicin (Dox) inIGNVs (Figure 3c).78 Intestinally, the targeted homingproperties of IGNVs toward inflammatory tumor tissuescould be blocked by some chemokine receptors includingLFA-1 and CXCR2, indicating that these receptors play keyroles in the recruitment and migration of leukocytes intoinflamed regions. These relevant studies demonstrated thatleukocyte-derived nanoparticles supply a versatile technique to

explore the detailed processes of leukocyte−endothelialinteractions during the inflammatory response in living systems.

3. PLATELET-DERIVED NANOPLATFORMS

As circulating sentinels in the bloodstream, platelets (alsoknown as thrombocytes) are key components in hemostasisand thrombosis during blood vessel injuries, and also performsignificant functions in the development of lymphaticvasculature and mediation of innate or adaptive immuneresponse.79−81 Moreover, platelets are involved in thepathological processes of multiple health issues includingcancer, inflammation, and infection, acting in a key role tomediate the platelet−cell interactions and their behaviors.82−84

However, extensive understanding of the detailed mechanismsand significant roles of platelets in these pathophysiologicalprocesses remain under unclear.85 Fortunately, recent studieshave demonstrated the outstanding merits of platelet-mimicking nanoparticles for the exploration of variouspathological pathways, including phagocytotic escape, immunesystem activation, and selective adhesion to damagedvasculature and tumor tissues.86,87 Until now, severalapproaches have been adopted to integrate platelets withvarious types of nanoparticles for the purpose of developmentof platelet-mimicking nanoplatforms. One initial strategy is totransfer platelet-derived surface moieties (e.g., proteins, glycans,etc.) with specific functions to synthetic liposome nanoparticles(plateletsomes). For example, by modifying the liposomebilayer moieties which contain over 15 kinds of platelet

Figure 3. Schematic illustration of leukocyte-derived nanoparticles. (a) LLV structure and possible interactions between the functional groups onNPS surface and membrane phospholipids. (b) TEM (top) and SEM (bottom) images of bare NPS (left) and leukocyte-derived NPS (LLV) (right).Scale bars: 100 nm (TEM) and 1 mm (SEM). (c) Synthesis procedures of IGNVs for targeted homing of therapeutic drugs to inflammatory sites(left). (Reproduced with permission from refs 77 and 78. Copyright 2013 Nature Publishing Group, and 2015 American Association for CancerResearch.)

Bioconjugate Chemistry Review

DOI: 10.1021/acs.bioconjchem.8b00103Bioconjugate Chem. 2018, 29, 838−851

841

Page 5: Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

membrane glycoproteins, such as GPIb, GPIIb-IIIa, and GPIV/III, Renzulli et al. reported a smart plateletsome with greathemostatic efficacy that presented a greater reduction (67%decrease) of tail bleeding in a thrombocytopenic rat model.88

Moreover, in order to exploit the specific interactions ofreceptors on the surfaces of platelets for targeting liposomedelivery, Marchant et al. modified an arginine-glycine-aspartic(RGD) peptide as a model ligand to target the integrin GPIIb-IIIa on activated platelets, which indicated that the peptides arecapable of directing liposomes to receptors expressed onpathologically stimulated vascular territories.89

Despite the expected results in hemostasis and targetedpayload delivery presented by artificial plateletsomes, it is still achallenging task to replicate the flexible shape and highlycomplex platelet−cell interactions.90−92 In order to fullypreserve the integrality of platelets, recent studies haveindicated the development of biomimetic nanoparticles thatcombined the plasma membrane of platelets with variousfunctional nanostructures. For instance, Zhang et al. developeda smart strategy utilizing platelet membrane-cloaked PLGA

nanoparticles (PNPs) for the specific clearance of antiplateletantibodies in blood for effective treatment of immunethrombocytopenia (Figure 4a).93 The PNPs could act asdecoys to strongly bind with pathological antiplatelet antibodiesand subsequently neutralize them with considerable therapeuticefficacy for immune thrombocytopenia purpura in a murinemodel. Furthermore, they also reported the PNPs which endowthe nanoplatform surface with platelets for the adherence ofseveral disease-relevant substrates (Figure 4b).94 The resultingPNPs contained specific integrin components (e.g., αIIb, α2,β1, etc.), transmembrane proteins (e.g., GPIbα, GPIV, CLEC-2, etc.), and immunomodulatory antigens (e.g., CD47, CD55,CD59, etc.), which could selectively adhere to the pathogens indamaged vasculature of living animals. Moreover, enhancedtherapeutic efficiency was determined to inhibit the growth ofneointima in a coronary restenosis rat model by loading withdocetaxel (Dtxl) and vancomycin (Van), which presented amultifaceted approach in developing an effective nanoplatformfor disease-targeting treatment. Such unique approaches basedon platelet-derived nanoplatforms provided promising feasi-

Figure 4. Platelet-derived nanoplatforms. (a) Scheme of PNP preparation and activation as decoys to neutralize pathological antiplatelet antibodiesfor the treatment of immune thrombocytopenia. (b) (Left) H&E-stained arterial cross sections of normal (top) and zoomed-in (bottom) tissues in arat model of coronary restenosis at different treatment groups: baseline, Dtxl-loaded PNPs, PNPs, and Dtxl. I: intima; M: media. Scale bar: 200 mm(top), 100 mm (bottom). (Right) SEM images of MRSA252 bacteria at different groups. Scale bar: 1 μm. (Reproduced with permission from refs 93and 94. Copyright 2016 Elsevier and 2015 Nature Publishing Group.)

Figure 5. Schematic illustration of exosome-derived nanoplatforms: (a) Preparation procedures of targeted exosomes for gene delivery. (b)Fabrication of exosome-like nanovesicles (BLNs) and their excellent targeting ligand-mediated affinity to the EGFR- or HER2-overexpressing tumorcells. (Reproduced with permission from refs 106 and 113. Copyright 2011 Nature Publishing Group and 2017 John Wiley & Sons).

Bioconjugate Chemistry Review

DOI: 10.1021/acs.bioconjchem.8b00103Bioconjugate Chem. 2018, 29, 838−851

842

Page 6: Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

bility to fabricate the investigations of platelet−cell interactionsin complicated pathophysiological processes including hemo-stasis, inflammation, and infection in living conditions.

4. EXOSOME-DERIVED NANOPLATFORMS

Exosomes, one type of intrinsic cell-derived small membranevesicle usually with diameter range of 40−100 nm, can besecreted by most cell types in biological fluids.95,96 Typically,the surfaces of exosomes consist of different kinds of biologicalcomponents, such as chaperone proteins, adhesion molecules,and metabolic enzymes,97,98 which exert their biological effectsin a highly diversified manner, including activation of targetedcell surface receptors via protein−ligand interactions, mergingof the membrane contents with the recipient cell membrane, ordirect delivery of proteins, mRNA, and lipid into recipientcells.99−101 Most of these features are determined by theirspecific surface protein expression originating from parentcells.102,103 Therefore, exosomes have been well recognized asan attractive nanoplatform for extensive biomedical applicationsdue to their versatile and alterable membrane functions.104−107

For example, Wood et al. recently produced dendritic cell-derived exosomes for targeted delivery of short interfering RNA(siRNA) into the mouse brain for Alzheimer’s diseasetreatment (Figure 5a).106 In order to reduce the immunoge-nicity and achieve a targeting effect, the dendritic cells wereengineered to produce natural exosomes with membraneprotein (Lamp2b) expression for selective fusion withneuron-specific RVG peptide. Upon loading exogenoussiRNA through electroporation, the RVG-targeted exosomescould effectively deliver GAPDH siRNA to neurons, microglia,and oligodendrocytes in the mouse brain after intravenousinjection. Moreover, efficient mRNA (∼60%) and protein(∼62%) knockdown of a therapeutic target in Alzheimer’sdisease (BACE1) was determined in vivo, which clearlydemonstrated the effective therapeutic effects mediated bysiRNA delivery nanoplatform with modification of exosome.

Furthermore, Kang et al. also prepared dual-functionalexosome-based drug delivery vehicles based on superparamag-netic nanoparticle clusters for effective tumor treatment.107

With a strong superparamagnetic property under an externalmagnetic field, the drug-loaded exosomes could be efficientlyaccumulated at desired tumor regions for significant inhibitionof tumor growth in living animals. This strategy endowed theexosomes with magnetism and could thus advance the potentialusage of exosomes in vivo.In spite of the promising perspectives of exosomes in

biological sciences, so far, how to rapidly produce, isolate, andpurify exosomes in sufficient amounts remains a technicalchallenge that requires more research effort.108,109 Moreover,natural exosomes usually have complicated surface compo-nents, which may raise potential concerns to interfere with theexosome−cell interactions during long-distance intercellularcommunication and targeted payload delivery.110,111 To addressthese issues, synthetic exosome-like nanoparticles, whichcombine desired membrane proteins with phospholipid bilayeron the surface of artificial exosomes, have been developed inrecent years.112−115 For instance, by utilizing biomimeticsynthesis strategies, Liu et al. presented biofunctionalizedliposome-like nanovesicles (BLNs) that are capable ofartificially encapsulating two different kinds of tumor targetingmoieties for effective drug delivery and cancer therapy in livingmice (Figure 5b).113 Upon genetic engineering with humanepidermal growth factor (hEGF) or anti-HER2 affibody astargeting ligands, the BLNs exhibited higher biological activityand selectivity toward EGF receptor-overexpressing cancercells, and enhanced therapeutic outcomes than clinicallyapproved liposomal-Dox in HER2-overexpressing BT474tumor xenograft models. In addition, Gho et al. also reportedexosome-mimetic nanovesicles with anticancer drug loading fortargeted delivery in chemotherapy of cancer.114 The hybridnanovesicles were prepared through breakdown of monocytesor macrophages by utilizing a serial extrusion with filters of

Figure 6. Schematic illustration of bacterial-mimicking nanoplatforms. (a) Modulation of antibacterial vaccines via BM-AuNPs. (b) TEM image andstability of BM-AuNPs (top); CD11c+ and INF-γ expressions from activated dendritic cells and T cells in vivo (bottom). (c) EGFPDNV productionfrom EGF expressing bacteria. (Reproduced with permission from refs 130 and 133. Copyright 2016 American Chemical Society and 2017 Elsevier.)

Bioconjugate Chemistry Review

DOI: 10.1021/acs.bioconjchem.8b00103Bioconjugate Chem. 2018, 29, 838−851

843

Page 7: Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

different pore sizes. Interestingly, compared with traditionalexosomes, these nanovesicles presented 100-fold higherproduction yield and exhibited excellent targeting capabilityby mimicking the topology of membrane proteins. Moreover,enhanced cell death and tumor growth inhibition without toxicside effects have also been identified in living mice, clearlysuggesting that the bioengineered nanovesicles could serve asnovel exosome-mimetics with selective tumor affinity forenhanced treatment. These simplified exosome nanostructuresnot only could be manufactured in a mass production mannerthrough standard technology in industry, but they also providedesired surface functionalization methods to achieve specificinvestigation of exosome−cell interactions in vitro and in vivo.

5. BACTERIAL-MIMICKING NANOPLATFORMSIt is well-known that there are a variety of microbes (e.g.,bacteria, viruses, fungi, and other tiny organisms) throughoutthe human body, which can definitely act as essentialcomponents of immunity and functional entities to influencefundamental metabolism and modulate cell host−microbeinteractions in living systems.116−118 As one type of importantmicrobes, bacterial species are actually of great practical interestto human beings, and they are essential for normal bodyfunctions including digestion and immune response. In general,a majority of bacteria are harmless owing to the protectiveeffects of the innate immune system; some are even particularlybeneficial in the gut.119,120 However, several kinds of extraneousbacteria are indeed pathogenic and induce various infectiousdiseases, including cholera, anthrax, tuberculosis, and syph-ilis.121−123 Normally, the diverse bacterial surface componentsplay critical roles in the pathogenesis of infectious disease sincethey mediate the specific activities of bacteria−cell interactionsin living conditions, including colonizing tissues, resistingphagocytosis, and activating immune responses.124,125 Withthese attractive features, the excellent bacterial-mimickingsystems that can avoid the pathogenicity of living bacteria aswell as preserve the integrality of bacterial membrane havebecome highly desirable in recent decades.126

Considering the promising ability of nanoparticles to mimicthe aforementioned key aspects of the cellular membrane,various bacteria-mimicking nanoparticles have been proposedin recent years for biomedical applications including thedevelopment of antibacterial vaccines and targeted deliveryvehicles.127−131 For instance, by using Escherichia coli as amodel pathogen, Zhang et al. developed a unique bacterialmembrane-cloaked gold nanoparticle (BM-AuNPs) as anexciting and robust antibacterial vaccine (Figure 6a).130 Thebacterial outer membrane vesicles (OMVs) were collected andfurther coated on the surface of small AuNPs (∼30 nm). Aftersubcutaneous injection, the BM-AuNPs induced rapidactivation and maturation of dendritic cells in lymph nodes ofvaccinated mice. Interestingly, the BM-AuNPs presented ahigher efficacy to elicit bacterium-specific B-cell and T-cellresponses in the vaccinated animals than those elicited byOMVs only, indicating that the synergistic action of bacterialmembranes and AuNP cores could benefit each other forenhanced immune responses (Figure 6b). These results clearlyshowed that the synthetic nanoparticles with natural bacterialmembrane modification hold great promise for fabricatingeffective antibacterial vaccines.Moreover, so far, the bacterial-mimicking strategy has also

been applied to establish the effective delivery vehicles towardenhanced targeting of diseases regions.132−135 For instance,

Gho et al. engineered one novel nanovesicle system by utilizingbacterial protoplast (a type of cells with wall structureremoved) as a unique cargo for targeted delivery (Figure6c).133 After removing the toxins in the outer wall of thebacteria, the bacterial protoplast-derived nanovesicles (PDNV)could be fabricated by serial extrusions on the basis ofnanosized membrane filters. The PDNV could selectivelydeliver chemotherapeutics (e.g., Dox) to tumor tissues viareceptor-mediated interactions through the specific surfaceexpression of tumor-targeting moieties, such as epidermalgrowth factor (EGF), etc., in in vivo experiments furtherindicated that the drug-loaded PDNV could not only efficientlyinhibit the tumor growth, but also reduce the chemo-therapeutics-induced adverse effects in the heart after systemicadministration to mice. These innovative studies demonstratedthat bacterial-mimicking nanomaterials provide great potentialto systematically understand the complicated bacteria−cellinteractions during the treatment of diverse infectious diseases.

6. VIRUS-MIMICKING NANOPLATFORMSAs a small infectious species, virus can replicate itself only whenit invades into the host including animals, plants, bacteria, andother organisms.136,137 Naturally pathogenic viruses possess theintrinsic ability to avoid immune system recognition and injecttheir genetic material (e.g., DNA or RNA) into a host for self-replication, which will cause severe infectious inflammation(e.g., AIDS, SARS, Ebola virus disease, etc.) and eventuallyresult in the death of the host.138−140 During the invasionprocesses, the outer membrane of the virus such as the capsid(a protein coat) or envelope (lipid bilayer) plays a significantrole in viral infection, including cell attachment and entry, generelease, and assembling of newly formed viruses.141−143

Therefore, the detailed understanding of the intricate virus−cell interactions will ultimately provide more information forthe design of innovative and effective therapeutics against viralinfection.So far, various viral vectors such as adenoviruses, retroviruses,

and lentiviruses, etc., have been utilized for successful clinicalapplications in the treatment of adenosine deaminase deficiencyand X-linked severe combined immunodeficiency.144−146

However, considering the case in which these viral vectorsare pathogenic and can be derived from viruses in naturalinfection, substantial concerns still occur regarding theirpotential issues in safety and immunogenicity.147 In order toachieve the benefits of viruses while greatly minimizing thesepotential issues of introducing pathogenic genes, extensiveresearch efforts have been engaged to design an initialgeneration of virus-like nanoparticles (VNPs) and virosomes,which are self-assembled nanoparticles and could mimic thecapsid and envelope structures with incorporation of functionalsurface glycoproteins in real viruses.148−152 For example,Sainsbury et al. reported the recombinant of novel VNPsbased on the capsid assembled by bluetongue virus structuralproteins (VP3 and VP7) from plant leaves (Figure 7a).152 TheVNPs presented specific capability to bind with cell surfacereceptors (e.g., αvβ3/β5 integrins) by integrating with cyclicRGD peptide, which could act as an attractive vehicle foreffective payload delivery including therapeutic drugs, contrastagents, proteins, and siRNA toward cancer treatment.Encouraged by these successful investigations, scientists

developed novel nanoparticles that mimic various naturalfeatures of viruses (e.g., surface antigens recognition,cytoplasmic capsid assembly, immune system escape, etc) to

Bioconjugate Chemistry Review

DOI: 10.1021/acs.bioconjchem.8b00103Bioconjugate Chem. 2018, 29, 838−851

844

Page 8: Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

explore the virus-cell interactions through the specific surfacemodifications of these assembled carriers.153−155 For example,inspired by viral capsid protein structures, Ni and Chau recentlyconstructed a biomimetic capsid assembled by syntheticpeptide with specific nanoribbon and nanococoon shapes andstriped surface patterns (Figure 7b).153 The rational design ofthis smart peptide contained different segments for DNAbinding and β-sheet assembly, which offered the capabilities ofartificial capsid with excellent stability, low permeability, andresistance to enzyme digestion for gene protection. Moreimportantly, this biomimetic strategy could regulate and controlthe properties of synthetic capsid by introducing diversefunctional groups into assembling blocks, which thereforeproduced a feasible model to understand the peptide/DNAinteraction during the capsid encapsulation process. Thesepromising studies suggested that virus-mimicking nanoparticlescould provide more beneficial information for effectivepharmaceutical development against viral infection and detailedunderstanding of virus activities in living animals.

■ CONCLUSION AND PERSPECTIVESCurrently, extensive nanomedicine holds great potential for theprecise diagnosis and effective therapeutics of various humandiseases in clinical practice. However, a detailed understanding

regarding how nanomedicine interact with the intricateenvironment in complex living systems, still remains challeng-ing. To this end, inspired by the diversified membranestructures and functions of natural prototypes, relevant researchhas increased exponentially for the development of membrane-mimicking nanoplatforms, which endow the abiotic nanoma-terials with specific biological functionalities to investigate thecomplicated interactions and activities of nanomedicine inhuman bodies. In this review, we focused on the strategiesestablished recently for the development of membrane-cloakednanoplatforms derived from inherent host cells (e.g.,erythrocytes, leukocytes, platelets, and exosomes) and invasivepathogens (e.g., bacteria and viruses), mainly attributed to theirversatile membrane properties in biological fluids. Therepresentative examples of different kinds of biomimeticmembrane-cloaked nanoplatforms in living system aresummarized in Table 1. Despite the wide exploitation ofdiverse membrane-mimicking strategies in recent decades, thereis still a long way toward conducting any clinical trials in thisresearch field.For example, these “cloaking” strategies could effectively

endow various nanoparticles with specific advantages of diversebiological membranes, such as long circulation time in blood(erythrocytes), great selectivity at inflamed endothelial regions(leukocytes), and excellent capacity to evade immune systemrecognition (viruses). However, numerous works are greatlyexpected to formulate these bioinpired nanoplatforms byavoiding their undesirable shortcomings, including complexsynthetic and purification routes (platelets), lack of stand-ardized protocol for preparation and isolation in sufficientamounts (erythrocytes and exosomes), and potential concernsregarding safety and immunogenicity in the human body(bacteria and viruses) (Table 2). Therefore, great challengesstill remain in this research area which require extensiveexploitation in the near future. First of all, although thesebiomimetic strategies based on various cell membranes andpathogens have been widely established in recent years, it is stilldifficult to maintain the integrality and functionality of naturalentities due to the requirement of multiple labor-intensiveprocesses during the fabrication of these membrane-mimickingnanoparticles, such as genetic engineering or prolonged ex vivohypotonic treatment.49,149 For example, the surface integralityof RBCs could be compromised during ex vivo producing ofRBC-coated nanoparticles, which may result in decreasingcirculation time and rapid clearance by the immune system.8,76

Therefore, researchers should pay more attention to optimizingthe membrane extraction techniques and particle−membranefusion procedures, which will minimize the structuralalterations for more accurate investigations of the relationshipsbetween the interfaces of nanotechnology and biology.Furthermore, even though several rational designs of

membrane-cloaked nanoplatforms are inspired by naturalbiomoieties including living cells and pathogens, the potentialimmunogenicity of these biomimetic nanostructures may stilloccur as undesired side effects and safety concerns, especiallyfor pathogen-mimicking nanoparticles.162 For example, a seriesof conformational changes of the membrane-anchored frag-ments (e.g., proteins, glycans, etc.) might be occurring duringthe period of membrane extraction or fusion with nanoparticles,which could be recognized as invaders to activate immuneresponse.163 Significantly, it is worth noting that some certaindegrees of immunogenicity could be beneficial to human healthwhen the pathogen-mimicking nanoparticles are designed as

Figure 7. Schematic illustration of virus-mimicking nanoplatforms. (a)Synthesis and isolation procedures of plant-based VNPs (top);recombined structure and TEM image of VNPs (bottom) assembledby virus proteins (VP3 and VP7). Scale bar: 200 nm. (b) Chemicalstructure of peptide and TEM images of self-assembled capsid withnanoribbon and nanococoon structures in the absence and presence ofDNA as templates. Scale bar: 100 nm. (Reproduced with permissionfrom refs 152 and 153. Copyright 2014 and 2017 American ChemicalSociety.)

Bioconjugate Chemistry Review

DOI: 10.1021/acs.bioconjchem.8b00103Bioconjugate Chem. 2018, 29, 838−851

845

Page 9: Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

vaccines to stimulate the adaptive immune responses. However,the potentially immunogenic components of pathogens thatmay induce unexpected immune response/reaction in vivomust be removed or inactivated, and their biosafety should bethoroughly addressed by the examinations in preclinicalstudies.164

In summary, the biomimetic membrane-cloaked nanoplat-forms, by integrating the diversified properties of variousbiological membranes and nanomaterials, provide a brightperspective for the investigations regarding the performance ofnanomedicine within the intricate environment during diversephysiological and pathological processes in living systems.

Along with all the innovative studies in these research areas, webelieve that these bioinspired strategies conjugated withattractive features of nanomaterials will promote the develop-ment of efficient and precise nanomedicine and finally have apromising outlook to benefit human health.

■ AUTHOR INFORMATIONCorresponding Author*E-mail: [email protected] Lin: 0000-0001-9572-2134Bengang Xing: 0000-0002-8391-1234

Table 1. Representative Examples of Biomimetic Membrane-Cloaked Nanoplatforms in Living Systems

membranesources corea size (nm) payloadb

loadingcapacity

targetingmoietiesc

circulation time (t1/2,h) %ID/g in organsd (24 h) ref

Erythrocytes PLGA 80 −− −− −− 39.6 L: 39, S: 26, 49G: 14, B: 10

MSNs 108 Ce6 21% −− 11.5 L: 40, S: 13, 41Dox 39% G: 15, T: 18

Bismuth (Bi) NPs 56 Bi 70% FA 17.1 L: 34, S: 19, 156K: 7, T: 9

Leukocytes GNV 200 Dox 75% CXCR2 LFA-1 −− L: 18, S: 14, 78K: 7, G: 8, T: 7

−− 120 PTX 76% LFA-1 CD45 −− L: 48, B: 12, 157K: 13, T: 7

liposome 115 DM1 96% α4β1- integrin 4.9 L: 62, G: 21, 158K: 9, S: 6

Platelets PLGA 113 Dtxl 2.1% surface- glycans 33.2 L: 42, S: 28, 94200 Van 4.0% K: 4, D: 17

nanogel 121 Dox −− TRAIL, 32.6 L: 19, S: 5, K: 159P-selectin 12, G: 7, T: 53

Exosomes −− 88 siRNA 25% Lamp2b −− L: 19, S: 16, 106M: 18, H:17

−− 105 ICG >70% hEGF 7.2 −− 113Dox anti-HER2

Bacteria −− 42 Dox 40% hEGF −− L: 7, S: 14, K: 1336, G: 6, T: 62

−− 90−133 siRNA 15% anti-HER2 −− −− 160Viruses −− 50−150 HA 6.5% HPV16 L2 −− L: 45, S: 42, 151

K: 12−− 33 Dox 3.9% RGD −− L: 14, S: 4, 161

K: 24, T: 37aMSNs: mesoporous silica nanoparticles. bCe6: chlorin e6, PTX: paclitaxel, DM1: emtansine, ICG: Indocyanine green, HA: hemagglutinin. cFA:folic acid, CXCR2: CXC chemokine receptor 2, LFA-1: Lymphocyte function-associated antigen 1 (LFA-1), TRAIL: tumor necrosis factor (TNF)-related apoptosis inducing ligand, HPV: human papillomavirus, RGD: Arg-Gly-Asp tripeptide. dL: liver, S: spleen, K: kidney, G: lung, B: blood, T:tumor, D: denuded artery, M: muscle, H: heart.

Table 2. Advantages and Disadvantages of Various Biomimetic Membrane-Cloaked Nanoplatforms

membranetypes advantages disadvantages

Erythrocytes Long circulation time in blood Time-consuming purification methodsSimple approaches for membrane functionalization Lack of standardized protocol for preparation, purification, and storage in

sufficient amountsLeukocytes Great selectivity at specific disease regions and regulation of

inflammatory responseInadequacy reproducing the integrality and complexity of leukocytemembrane

Platelets Favorable properties in treating hemostasis, hemorrhage andtargeted payloads delivery

Complex synthetic and purification routesLimited assessment of immunogenic potential

Exosomes Promising candidate for payload delivery Lack of standardized methods to rapidly produce, isolate, and purify exosomesin sufficient amountsLong-distance cell-to-cell communications

Bacteria Great antibacterial vaccines and targeted delivery vehicles Potential concerns regarding safety and immunogenicityViruses Excellent capacity in cellular targeting, entry, and avoiding immune

system recognitionPotential concerns regarding safety and immunogenicity

Bioconjugate Chemistry Review

DOI: 10.1021/acs.bioconjchem.8b00103Bioconjugate Chem. 2018, 29, 838−851

846

Page 10: Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

NotesThe authors declare no competing financial interest.

■ ACKNOWLEDGMENTSThe authors acknowledge the financial support by NTU-AIT-MUV NAM/16001, RG110/16 (S), Merlion 2017 Program(M4082162), JSPS-NTU Joint Research (M4082175) and (RG35/15) awarded in Nanyang Technological University,Singapore, and National Natural Science Foundation ofChina (NSFC) (No. 51628201).

■ REFERENCES(1) Veiseh, O., Tang, B. C., Whitehead, K. A., Anderson, D. G., andLanger, R. (2015) Managing diabetes with nanomedicine: challengesand opportunities. Nat. Rev. Drug Discovery 14, 45−57.(2) Shi, J., Kantoff, P. W., Wooster, R., and Farokhzad, O. C. (2017)Cancer nanomedicine: progress, challenges and opportunities. Nat.Rev. Cancer 17, 20−37.(3) Jain, R. K., and Stylianopoulos, T. (2010) Delivering nano-medicine to solid tumors. Nat. Rev. Clin. Oncol. 7, 653−664.(4) Ai, X., Ho, C. J. H., Aw, J., Attia, A. B. E., Mu, J., Wang, Y., Wang,X., Wang, Y., Liu, X., Chen, H., et al. (2016) In vivo covalent cross-linking of photon-converted rare-earth nanostructures for tumourlocalization and theranostics. Nat. Commun. 7, 10432−10440.(5) Chen, G., Roy, I., Yang, C., and Prasad, P. N. (2016)Nanochemistry and nanomedicine for nanoparticle-based diagnosticsand therapy. Chem. Rev. 116, 2826−2885.(6) Min, Y., Caster, J. M., Eblan, M. J., and Wang, A. Z. (2015)Clinical translation of nanomedicine. Chem. Rev. 115, 11147−11190.(7) Mehdi, A., Reye, C., and Corriu, R. (2011) From molecularchemistry to hybrid nanomaterials. Design and functionalization.Chem. Soc. Rev. 40, 563−574.(8) Mout, R., Moyano, D. F., Rana, S., and Rotello, V. M. (2012)Surface functionalization of nanoparticles for nanomedicine. Chem.Soc. Rev. 41, 2539−2544.(9) Chen, X., Li, C., Gratzel, M., Kostecki, R., and Mao, S. S. (2012)Nanomaterials for renewable energy production and storage. Chem.Soc. Rev. 41, 7909−7937.(10) Ai, X., Lyu, L., Zhang, Y., Tang, Y., Mu, J., Liu, F., Zhou, Y., Zuo,Z., Liu, G., and Xing, B. (2017) Remote Regulation of MembraneChannel Activity by Site-Specific Localization of Lanthanide-DopedUpconversion Nanocrystals. Angew. Chem., Int. Ed. 56, 3031−3035.(11) Barreto, J. A., O’Malley, W., Kubeil, M., Graham, B., Stephan,H., and Spiccia, L. (2011) Nanomaterials: applications in cancerimaging and therapy. Adv. Mater. 23, 18−44.(12) Dobrovolskaia, M. A., and McNeil, S. E. (2007) Immunologicalproperties of engineered nanomaterials. Nat. Nanotechnol. 2, 469−478.(13) De, M., You, C. C., Srivastava, S., and Rotello, V. M. (2007)Biomimetic interactions of proteins with functionalized nanoparticles:a thermodynamic study. J. Am. Chem. Soc. 129, 10747−10753.(14) Murillo, G., Blanquer, A., Vargas-Estevez, C., Barrios, L., Ibanez,E., Nogues, C., and Esteve, J. (2017) Electromechanical Nano-generator-Cell Interaction Modulates Cell Activity. Adv. Mater. 29,1605048.(15) Drees, C., Raj, A. N., Kurre, R., Busch, K. B., Haase, M., andPiehler, J. (2016) Engineered upconversion nanoparticles for resolvingprotein interactions inside living cells. Angew. Chem., Int. Ed. 55,11668−11672.(16) Zhang, X. Q., Xu, X., Bertrand, N., Pridgen, E., Swami, A., andFarokhzad, O. C. (2012) Interactions of nanomaterials and biologicalsystems: Implications to personalized nanomedicine. Adv. DrugDelivery Rev. 64, 1363−1384.(17) Nguyen, V. H., and Lee, B.-J. (2017) Protein corona: A newapproach for nanomedicine design. Int. J. Nanomed. 12, 3137−3151.(18) Corbo, C., Molinaro, R., Parodi, A., Toledano Furman, N. E.,Salvatore, F., and Tasciotti, E. (2016) The impact of nanoparticleprotein corona on cytotoxicity, immunotoxicity and target drugdelivery. Nanomedicine 11, 81−100.

(19) Geldert, A., Liu, Y., Loh, K. P., and Lim, C. T. (2017) Nano-biointeractions between carbon nanomaterials and blood plasma proteins:why oxygen functionality matters. NPG Asia Mater. 9, e422.(20) Pearson, R. M., Hsu, H.-j., Bugno, J., and Hong, S. (2014)Understanding nano-bio interactions to improve nanocarriers for drugdelivery. MRS Bull. 39, 227−237.(21) Lai, Z. W., Yan, Y., Caruso, F., and Nice, E. C. (2012) Emergingtechniques in proteomics for probing nano−bio interactions. ACSNano 6, 10438−10448.(22) Wang, H., Agarwal, P., Zhao, S., Yu, J., Lu, X., and He, X. (2015)A biomimetic hybrid nanoplatform for encapsulation and preciselycontrolled delivery of theranostic agents. Nat. Commun. 6, 10081−10093.(23) Balmert, S. C., and Little, S. R. (2012) Biomimetic Delivery withMicro-and Nanoparticles. Adv. Mater. 24, 3757−3778.(24) Ruiz-Hitzky, E., Darder, M., Aranda, P., and Ariga, K. (2010)Advances in biomimetic and nanostructured biohybrid materials. Adv.Mater. 22, 323−336.(25) Parodi, A., Quattrocchi, N., Van De Ven, A. L., Chiappini, C.,Evangelopoulos, M., Martinez, J. O., Brown, B. S., Khaled, S. Z., Yazdi,I. K., Enzo, M. V., et al. (2013) Synthetic nanoparticles functionalizedwith biomimetic leukocyte membranes possess cell-like functions. Nat.Nanotechnol. 8, 61−68.(26) Zan, G., and Wu, Q. (2016) Biomimetic and bioinspiredsynthesis of nanomaterials/nanostructures. Adv. Mater. 28, 2099−2147.(27) Patterson, D. P., Rynda-Apple, A., Harmsen, A. L., Harmsen, A.G., and Douglas, T. (2013) Biomimetic antigenic nanoparticles elicitcontrolled protective immune response to influenza. ACS Nano 7,3036−3044.(28) Zhang, P., Liu, G., and Chen, X. (2017) Nanobiotechnology:Cell membrane-based delivery systems. Nano Today 13, 7−9.(29) Tang, J., Shen, D., Caranasos, T. G., Wang, Z., Vandergriff, A.C., Allen, T. A., Hensley, M. T., Dinh, P. U., Cores, J., Li, T. S., et al.(2017) Therapeutic microparticles functionalized with biomimeticcardiac stem cell membranes and secretome. Nat. Commun. 8, 13724−13736.(30) Trantidou, T., Friddin, M., Elani, Y., Brooks, N. J., Law, R. V.,Seddon, J. M., and Ces, O. (2017) Engineering CompartmentalizedBiomimetic Micro-and Nanocontainers. ACS Nano 11, 6549−6565.(31) Fang, R. H., Jiang, Y., Fang, J. C., and Zhang, L. (2017) Cellmembrane-derived nanomaterials for biomedical applications. Bio-materials 128, 69−83.(32) Kroll, A. V., Fang, R. H., and Zhang, L. (2017) Biointerfacingand applications of cell membrane-coated nanoparticles. BioconjugateChem. 28, 23−32.(33) Gao, W., and Zhang, L. (2015) Coating nanoparticles with cellmembranes for targeted drug delivery. J. Drug Targeting 23, 619−626.(34) Parodi, A., Molinaro, R., Sushnitha, M., Evangelopoulos, M.,Martinez, J. O., Arrighetti, N., Corbo, C., and Tasciotti, E. (2017) Bio-inspired engineering of cell-and virus-like nanoparticles for drugdelivery. Biomaterials 147, 155−168.(35) Luk, B. T., and Zhang, L. (2015) Cell membrane-camouflagednanoparticles for drug delivery. J. Controlled Release 220, 600−607.(36) Narain, A., Asawa, S., Chhabria, V., and Patil-Sen, Y. (2017) Cellmembrane coated nanoparticles: next-generation therapeutics. Nano-medicine 12, 2677−2692.(37) Angsantikul, P., Fang, R. H., and Zhang, L. (2017) ToxoidVaccination Against Bacterial Infection Using Cell Membrane-CoatedNanoparticles. Bioconjugate Chem., DOI: 10.1021/acs.bioconj-chem.7b00692.(38) Angsantikul, P., Thamphiwatana, S., Gao, W., and Zhang, L.(2015) Cell membrane-coated nanoparticles as an emergingantibacterial vaccine platform. Vaccines 3, 814−828.(39) Zhai, Y., Su, J., Ran, W., Zhang, P., Yin, Q., Zhang, Z., Yu, H.,and Li, Y. (2017) Preparation and Application of Cell Membrane-Camouflaged Nanoparticles for Cancer Therapy. Theranostics 7,2575−2592.

Bioconjugate Chemistry Review

DOI: 10.1021/acs.bioconjchem.8b00103Bioconjugate Chem. 2018, 29, 838−851

847

Page 11: Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

(40) Rascol, E., Devoisselle, J. M., and Chopineau, J. (2016) Therelevance of membrane models to understand nanoparticles-cellmembrane interactions. Nanoscale 8, 4780−4798.(41) Su, J., Sun, H., Meng, Q., Zhang, P., Yin, Q., and Li, Y. (2017)Enhanced blood suspensibility and laser-activated tumor-specific drugrelease of theranostic mesoporous silica nanoparticles by functionaliz-ing with erythrocyte membranes. Theranostics 7, 523−537.(42) Gao, M., Liang, C., Song, X., Chen, Q., Jin, Q., Wang, C., andLiu, Z. (2017) Erythrocyte-Membrane-Enveloped Perfluorocarbon asNanoscale Artificial Red Blood Cells to Relieve Tumor Hypoxia andEnhance Cancer Radiotherapy. Adv. Mater. 29, 1701429.(43) Gao, L., Wang, H., Nan, L., Peng, T., Sun, L., Zhou, J., Xiao, Y.,Wang, J., Sun, J., Lu, W., et al. (2017) Erythrocyte membrane-wrappedpH sensitive polymeric nanoparticles for non-small cell lung cancertherapy. Bioconjugate Chem. 28, 2591−2598.(44) Ding, H., Lv, Y., Ni, D., Wang, J., Tian, Z., Wei, W., and Ma, G.(2015) Erythrocyte membrane-coated NIR-triggered biomimeticnanovectors with programmed delivery for photodynamic therapy ofcancer. Nanoscale 7, 9806−9815.(45) O’Neill, J. S., and Reddy, A. B. (2011) Circadian clocks inhuman red blood cells. Nature 469, 498−503.(46) Oldenborg, P. A., Zheleznyak, A., Fang, Y. F., Lagenaur, C. F.,Gresham, H. D., and Lindberg, F. P. (2000) Role of CD47 as a markerof self on red blood cells. Science 288, 2051−2054.(47) Zen, K., Guo, Y., Bian, Z., Lv, Z., Zhu, D., Ohnishi, H., Matozaki,T., and Liu, Y. (2013) Inflammation-induced proteolytic processing ofthe SIRPα cytoplasmic ITIM in neutrophils propagates a proin-flammatory state. Nat. Commun. 4, 2436−2446.(48) Zhao, X. W., van Beek, E. M., Schornagel, K., Van der Maaden,H., Van Houdt, M., Otten, M. A., Finetti, P., Van Egmond, M.,Matozaki, T., Kraal, G., et al. (2011) CD47-signal regulatory protein-α(SIRPα) interactions form a barrier for antibody-mediated tumor celldestruction. Proc. Natl. Acad. Sci. U. S. A. 108, 18342−18347.(49) Hu, C. M. J., Zhang, L., Aryal, S., Cheung, C., Fang, R. H., andZhang, L. (2011) Erythrocyte membrane-camouflaged polymericnanoparticles as a biomimetic delivery platform. Proc. Natl. Acad. Sci.U. S. A. 108, 10980−10985.(50) Hu, C. M. J., Fang, R. H., Luk, B. T., Chen, K. N., Carpenter, C.,Gao, W., Zhang, K., and Zhang, L. (2013) ‘Marker-of-self’function-alization of nanoscale particles through a top-down cellular membranecoating approach. Nanoscale 5, 2664−2668.(51) Allen, T. M. (2002) Ligand-targeted therapeutics in anticancertherapy. Nat. Rev. Cancer 2, 750−763.(52) Ulbrich, K., Hola, K., Subr, V., Bakandritsos, A., Tucek, J., andZboril, R. (2016) Targeted drug delivery with polymers and magneticnanoparticles: covalent and noncovalent approaches, release control,and clinical studies. Chem. Rev. 116, 5338−5431.(53) Ray, P. C., Khan, S. A., Singh, A. K., Senapati, D., and Fan, Z.(2012) Nanomaterials for targeted detection and photothermal killingof bacteria. Chem. Soc. Rev. 41, 3193−3209.(54) Gu, F., Zhang, L., Teply, B. A., Mann, N., Wang, A., Radovic-Moreno, A. F., Langer, R., and Farokhzad, O. C. (2008) Preciseengineering of targeted nanoparticles by using self-assembledbiointegrated block copolymers. Proc. Natl. Acad. Sci. U. S. A. 105,2586−2591.(55) Fang, R. H., Hu, C. M. J., Chen, K. N., Luk, B. T., Carpenter, C.W., Gao, W., Li, S., Zhang, D. E., Lu, W., and Zhang, L. (2013) Lipid-insertion enables targeting functionalization of erythrocyte membrane-cloaked nanoparticles. Nanoscale 5, 8884−8888.(56) Zhang, Y., Zhang, J., Chen, W., Angsantikul, P., Spiekermann, K.A., Fang, R. H., Gao, W., and Zhang, L. (2017) Erythrocytemembrane-coated nanogel for combinatorial antivirulence andresponsive antimicrobial delivery against Staphylococcus aureusinfection. J. Controlled Release 263, 185−191.(57) Gao, W., Hu, C. M. J., Fang, R. H., Luk, B. T., Su, J., and Zhang,L. (2013) Surface functionalization of gold nanoparticles with redblood cell membranes. Adv. Mater. 25, 3549−3553.(58) Rao, L., Meng, Q. F., Bu, L. L., Cai, B., Huang, Q., Sun, Z. J.,Zhang, W. F., Li, A., Guo, S. S., Liu, W., et al. (2017) Erythrocyte

membrane-coated upconversion nanoparticles with minimal proteinadsorption for enhanced tumor imaging. ACS Appl. Mater. Interfaces 9,2159−2168.(59) Rao, L., Bu, L. L., Xu, J. H., Cai, B., Yu, G. T., Yu, X., He, Z.,Huang, Q., Li, A., Guo, S. S., et al. (2015) Red blood cell membrane asa biomimetic nanocoating for prolonged circulation time and reducedaccelerated blood clearance. Small 11, 6225−6236.(60) Hu, C. M. J., Fang, R. H., Copp, J., Luk, B. T., and Zhang, L.(2013) A biomimetic nanosponge that absorbs pore-forming toxins.Nat. Nanotechnol. 8, 336−340.(61) Copp, J. A., Fang, R. H., Luk, B. T., Hu, C.-M. J., Gao, W.,Zhang, K., and Zhang, L. (2014) Clearance of pathological antibodiesusing biomimetic nanoparticles. Proc. Natl. Acad. Sci. U. S. A. 111,13481−13486.(62) Hu, C. M. J., Fang, R. H., Luk, B. T., and Zhang, L. (2013)Nanoparticle-detained toxins for safe and effective vaccination. Nat.Nanotechnol. 8, 933−938.(63) Vicente-Manzanares, M., and Sanchez-Madrid, F. (2004) Roleof the cytoskeleton during leukocyte responses. Nat. Rev. Immunol. 4,110−122.(64) Friedl, P., and Weigelin, B. (2008) Interstitial leukocytemigration and immune function. Nat. Immunol. 9, 960−969.(65) Bloes, D. A., Kretschmer, D., and Peschel, A. (2015) Enemyattraction: bacterial agonists for leukocyte chemotaxis receptors. Nat.Rev. Microbiol. 13, 95−104.(66) Ley, K., Laudanna, C., Cybulsky, M. I., and Nourshargh, S.(2007) Getting to the site of inflammation: the leukocyte adhesioncascade updated. Nat. Rev. Immunol. 7, 678−689.(67) Zahr, A., Alcaide, P., Yang, J., Jones, A., Gregory, M., Dela Paz,N. G., Patel-Hett, S., Nevers, T., Koirala, A., Luscinskas, F. W., et al.(2016) Endomucin prevents leukocyte-endothelial cell adhesion andhas a critical role under resting and inflammatory conditions. Nat.Commun. 7, 10363−10372.(68) Langer, H. F., and Chavakis, T. (2009) Leukocyte-endothelialinteractions in inflammation. J. Cell. Mol. Med. 13, 1211−1220.(69) Haskard, D. O., and Landis, R. C. (2002) Interactions betweenleukocytes and endothelial cells in gout: lessons from a self-limitinginflammatory response. Arthrit. Res. Ther. 4, S91.(70) Peer, D. (2012) Immunotoxicity derived from manipulatingleukocytes with lipid-based nanoparticles. Adv. Drug Delivery Rev. 64,1738−1748.(71) Chen, X., Wong, R., Khalidov, I., Wang, A. Y., Leelawattanachai,J., Wang, Y., and Jin, M. M. (2011) Inflamed leukocyte-mimeticnanoparticles for molecular imaging of inflammation. Biomaterials 32,7651−7661.(72) Robbins, G. P., Saunders, R. L., Haun, J. B., Rawson, J., Therien,M. J., and Hammer, D. A. (2010) Tunable leuko-polymersomes thatadhere specifically to inflammatory markers. Langmuir 26, 14089−14096.(73) Ai, X., Lyu, L., Mu, J., Hu, M., Wang, Z., and Xing, B. (2017)Synthesis of Core-shell Lanthanide-doped Upconversion Nanocrystalsfor Cellular Applications. J. Visualized Exp. 129, 56416 DOI: 10.3791/56416.(74) Hammer, D. A., Robbins, G. P., Haun, J. B., Lin, J. J., Qi, W.,Smith, L. A., Ghoroghchian, P. P., Therien, M. J., and Bates, F. S.(2008) Leuko-polymersomes. Faraday Discuss. 139, 129−141.(75) Park, S., Kang, S., Chen, X., Kim, E. J., Kim, J., Kim, N., Kim, J.,and Jin, M. M. (2013) Tumor suppression via paclitaxel-loaded drugcarriers that target inflammation marker upregulated in tumorvasculature and macrophages. Biomaterials 34, 598−605.(76) Molinaro, R., Corbo, C., Martinez, J. O., Taraballi, F.,Evangelopoulos, M., Minardi, S., Yazdi, I. K., Zhao, P., De Rosa, E.,Sherman, M., et al. (2016) Biomimetic proteolipid vesicles fortargeting inflamed tissues. Nat. Mater. 15, 1037−1046.(77) Parodi, A., Quattrocchi, N., Van De Ven, A. L., Chiappini, C.,Evangelopoulos, M., Martinez, J. O., Brown, B. S., Khaled, S. Z., Yazdi,I. K., Enzo, M. V., et al. (2013) Synthetic nanoparticles functionalizedwith biomimetic leukocyte membranes possess cell-like functions. Nat.Nanotechnol. 8, 61−68.

Bioconjugate Chemistry Review

DOI: 10.1021/acs.bioconjchem.8b00103Bioconjugate Chem. 2018, 29, 838−851

848

Page 12: Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

(78) Wang, Q., Ren, Y., Mu, J., Egilmez, N. K., Zhuang, X., Deng, Z.,Zhang, L., Yan, J., Miller, D., and Zhang, H. G. (2015) Grapefruit-derived nanovectors use an activated leukocyte trafficking pathway todeliver therapeutic agents to inflammatory tumor sites. Cancer Res. 75,2520−2529.(79) Semple, J. W., Italiano, J. E., and Freedman, J. (2011) Plateletsand the immune continuum. Nat. Rev. Immunol. 11, 264.(80) Ojha, A., Nandi, D., Batra, H., Singhal, R., Annarapu, G. K.,Bhattacharyya, S., Seth, T., Dar, L., Medigeshi, G. R., Vrati, S., et al.(2017) Platelet activation determines the severity of thrombocytope-nia in dengue infection. Sci. Rep. 7, 41697−41706.(81) von Hundelshausen, P., and Weber, C. (2007) Platelets asimmune cells: bridging inflammation and cardiovascular disease. Circ.Res. 100, 27−40.(82) Franco, A. T., Corken, A., and Ware, J. (2015) Platelets at theinterface of thrombosis, inflammation, and cancer. Blood 126, 582−588.(83) Jenne, C. N., and Kubes, P. (2015) Platelets in inflammationand infection. Platelets 26, 286−292.(84) Ai, X., Mu, J., and Xing, B. (2016) Recent Advances of Light-Mediated Theranostics. Theranostics 6, 2439−2457.(85) Papapanagiotou, A., Daskalakis, G., Siasos, G., Gargalionis, A.,and G. Papavassiliou, A. (2016) The role of platelets in cardiovasculardisease: molecular mechanisms. Curr. Pharm. Des. 22, 4493−4505.(86) Dehaini, D., Wei, X., Fang, R. H., Masson, S., Angsantikul, P.,Luk, B. T., Zhang, Y., Ying, M., Jiang, Y., and Kroll, A. V. (2017)Erythrocyte-platelet hybrid membrane coating for enhanced nano-particle functionalization. Adv. Mater. 29, 1606209.(87) Pawlowski, C. L., Li, W., Sun, M., Ravichandran, K., Hickman,D., Kos, C., Kaur, G., and Sen Gupta, A. (2017) Platelet microparticle-inspired clot-responsive nanomedicine for targeted fibrinolysis.Biomaterials 128, 94−108.(88) Rybak, M. E. M., and Renzulli, L. A. (1993) A liposome basedplatelet substitute, the plateletsome, with hemostatic efficacy.Biomater., Artif. Cells, Immobilization Biotechnol. 21, 101−118.(89) Lestini, B. J., Sagnella, S. M., Xu, Z., Shive, M. S., Richter, N. J.,Jayaseharan, J., Case, A. J., Kottke-Marchant, K., Anderson, J. M., andMarchant, R. E. (2002) Surface modification of liposomes for selectivecell targeting in cardiovascular drug delivery. J. Controlled Release 78,235−247.(90) Amo, L., Tamayo-Orbegozo, E., Maruri, N., Eguizabal, C.,Zenarruzabeitia, O., Rinon, M., Arrieta, A., Santos, S., Monge, J., Vesga,M. A., et al. (2014) Involvement of Platelet-Tumor Cell Interaction inImmune Evasion. Potential Role of Podocalyxin-Like Protein 1. Front.Oncol. 4, 245.(91) Kral, J. B., Schrottmaier, W. C., Salzmann, M., and Assinger, A.(2016) Platelet interaction with innate immune cells. Transfus. Med.Hemother. 43, 78−88.(92) Lam, F. W., Vijayan, K. V., and Rumbaut, R. E. (2015) Plateletsand their interactions with other immune cells. Compr. Physiol. 5,1265−1280.(93) Wei, X., Gao, J., Fang, R. H., Luk, B. T., Kroll, A. V., Dehaini, D.,Zhou, J., Kim, H. W., Gao, W., Lu, W., et al. (2016) Nanoparticlescamouflaged in platelet membrane coating as an antibody decoy forthe treatment of immune thrombocytopenia. Biomaterials 111, 116−123.(94) Hu, C. J., Fang, R. H., Wang, K., Luk, B. T., Thamphiwatana, S.,Dehaini, D., Nguyen, P., Angsantikul, P., Wen, C. H., Kroll, A. V., et al.(2015) Nanoparticle biointerfacing by platelet membrane cloaking.Nature 526, 118−121.(95) Trajkovic, K., Hsu, C., Chiantia, S., Rajendran, L., Wenzel, D.,Wieland, F., Schwille, P., Brugger, B., and Simons, M. (2008)Ceramide triggers budding of exosome vesicles into multivesicularendosomes. Science 319, 1244−1247.(96) Raposo, G., and Stoorvogel, W. (2013) Extracellular vesicles:exosomes, microvesicles, and friends. J. Cell Biol. 200, 373−383.(97) Conde-Vancells, J., Rodriguez-Suarez, E., Embade, N., Gil, D.,Matthiesen, R., Valle, M., Elortza, F., Lu, S. C., Mato, J. M., andFalcon-Perez, J. M. (2008) Characterization and comprehensive

proteome profiling of exosomes secreted by hepatocytes. J. ProteomeRes. 7, 5157−5166.(98) Thery, C., Zitvogel, L., and Amigorena, S. (2002) Exosomes:composition, biogenesis and function. Nat. Rev. Immunol. 2, 569−579.(99) Valadi, H., Ekstrom, K., Bossios, A., Sjostrand, M., Lee, J. J., andLotvall, J. O. (2007) Exosome-mediated transfer of mRNAs andmicroRNAs is a novel mechanism of genetic exchange between cells.Nat. Cell Biol. 9, 654−659.(100) Wolfers, J., Lozier, A., Raposo, G., Regnault, A., Thery, C.,Masurier, C., Flament, C., Pouzieux, S., Faure, F., Tursz, T., et al.(2001) Tumor-derived exosomes are a source of shared tumorrejection antigens for CTL cross-priming. Nat. Med. 7, 297−303.(101) Phinney, D. G., Di Giuseppe, M., Njah, J., Sala, E., Shiva, S., StCroix, C. M., Stolz, D. B., Watkins, S. C., Di, Y. P., Leikauf, G. D., et al.(2015) Mesenchymal stem cells use extracellular vesicles to outsourcemitophagy and shuttle microRNAs. Nat. Commun. 6, 8472−8486.(102) Camussi, G., Deregibus, M. C., Bruno, S., Cantaluppi, V., andBiancone, L. (2010) Exosomes/microvesicles as a mechanism of cell-to-cell communication. Kidney Int. 78, 838−848.(103) Camussi, G., Deregibus, M.-C., Bruno, S., Grange, C., Fonsato,V., and Tetta, C. (2011) Exosome/microvesicle-mediated epigeneticreprogramming of cells. Am. J. Cancer Res. 1, 98−110.(104) Azmi, A. S., Bao, B., and Sarkar, F. H. (2013) Exosomes incancer development, metastasis, and drug resistance: a comprehensivereview. Cancer Metastasis Rev. 32, 623−642.(105) Bobrie, A., Colombo, M., Raposo, G., and Thery, C. (2011)Exosome secretion: molecular mechanisms and roles in immuneresponses. Traffic 12, 1659−1668.(106) Alvarez-Erviti, L., Seow, Y., Yin, H., Betts, C., Lakhal, S., andWood, M. J. (2011) Delivery of siRNA to the mouse brain by systemicinjection of targeted exosomes. Nat. Biotechnol. 29, 341−345.(107) Qi, H., Liu, C., Long, L., Ren, Y., Zhang, S., Chang, X., Qian,X., Jia, H., Zhao, J., Sun, J., et al. (2016) Blood exosomes endowedwith magnetic and targeting properties for cancer therapy. ACS Nano10, 3323−3333.(108) Momen-Heravi, F., Bala, S., Kodys, K., and Szabo, G. (2015)Exosomes derived from alcohol-treated hepatocytes horizontallytransfer liver specific miRNA-122 and sensitize monocytes to LPS.Sci. Rep. 5, 9991.(109) David, G., and Zimmermann, P. (2016) Heparanase tailorssyndecan for exosome production. Mol. Cell. Oncol. 3, e1047556.(110) Yim, N., Ryu, S.-W., Choi, K., Lee, K. R., Lee, S., Choi, H.,Kim, J., Shaker, M. R., Sun, W., Park, J. H., et al. (2016) Exosomeengineering for efficient intracellular delivery of soluble proteins usingoptically reversible protein−protein interaction module. Nat. Commun.7, 12277−12285.(111) Plebanek, M. P., Angeloni, N. L., Vinokour, E., Li, J., Henkin,A., Martinez-Marin, D., Filleur, S., Bhowmick, R., Henkin, J., Miller, S.D., et al. (2017) Pre-metastatic cancer exosomes induce immunesurveillance by patrolling monocytes at the metastatic niche. Nat.Commun. 8, 1319−1330.(112) Luan, X., Sansanaphongpricha, K., Myers, I., Chen, H., Yuan,H., and Sun, D. (2017) Engineering exosomes as refined biologicalnanoplatforms for drug delivery. Acta Pharmacol. Sin. 38, 754−763.(113) Zhang, P., Zhang, L., Qin, Z., Hua, S., Guo, Z., Chu, C., Lin,H., Zhang, Y., Li, W., Zhang, X., et al. (2018) Genetically EngineeredLiposome-like Nanovesicles as Active Targeted Transport Platform.Adv. Mater. 30, 1705350.(114) Jang, S. C., Kim, O. Y., Yoon, C. M., Choi, D. S., Roh, T.-Y.,Park, J., Nilsson, J., Lotvall, J., Kim, Y. K., and Gho, Y. S. (2013)Bioinspired exosome-mimetic nanovesicles for targeted delivery ofchemotherapeutics to malignant tumors. ACS Nano 7, 7698−7710.(115) García-Manrique, P., Gutierrez, G., and Blanco-Lopez, M. C.(2018) Fully Artificial Exosomes: Towards New TheranosticBiomaterials. Trends Biotechnol. 36, 10−14.(116) Dethlefsen, L., McFall-Ngai, M., and Relman, D. A. (2007) Anecological and evolutionary perspective on human−microbe mutual-ism and disease. Nature 449, 811−818.

Bioconjugate Chemistry Review

DOI: 10.1021/acs.bioconjchem.8b00103Bioconjugate Chem. 2018, 29, 838−851

849

Page 13: Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

(117) Nauseef, W. M. (2007) How human neutrophils kill anddegrade microbes: an integrated view. Immunol. Rev. 219, 88−102.(118) Brussow, H. (2015) Microbiota and the human nature: knowthyself. Environ. Microbiol. 17, 10−15.(119) Zhang, Y. J., Li, S., Gan, R. Y., Zhou, T., Xu, D. P., and Li, H. B.(2015) Impacts of gut bacteria on human health and diseases. Int. J.Mol. Sci. 16, 7493−7519.(120) Burne, R. A., and Chen, Y. Y. M. (2000) Bacterial ureases ininfectious diseases. Microbes Infect. 2, 533−542.(121) Hill, D. A., and Artis, D. (2010) Intestinal bacteria and theregulation of immune cell homeostasis. Annu. Rev. Immunol. 28, 623−667.(122) Sethi, S., and Murphy, T. F. (2001) Bacterial infection inchronic obstructive pulmonary disease in 2000: a state-of-the-artreview. Clin. Microbiol. Rev. 14, 336−363.(123) Anderson, R. M., May, R. M., and Anderson, B. (1992)Infectious diseases of humans: dynamics and control, Vol. 28, WileyOnline Library.(124) García, B., Merayo-Lloves, J., Martin, C., Alcalde, I., Quiros, L.M., and Vazquez, F. (2016) Surface proteoglycans as mediators inbacterial pathogens infections. Front. Microbiol. 7, 220−230.(125) Sutherland, I. W. (1988) Bacterial surface polysaccharides:structure and function, in International Reviews in Cytology, pp 187−231, Elsevier.(126) Merz, C., Knoll, W., Textor, M., and Reimhult, E. (2008)Formation of supported bacterial lipid membrane mimics. Biointer-phases 3, 41−50.(127) Lai, M.-H., Clay, N. E., Kim, D. H., and Kong, H. (2015)Bacteria-mimicking nanoparticle surface functionalization with target-ing motifs. Nanoscale 7, 6737−6744.(128) Kuehn, M. J., and Kesty, N. C. (2005) Bacterial outermembrane vesicles and the host-pathogen interaction. Genes Dev. 19,2645−2655.(129) Poetsch, A., and Wolters, D. (2008) Bacterial membraneproteomics. Proteomics 8, 4100−4122.(130) Gao, W., Fang, R. H., Thamphiwatana, S., Luk, B. T., Li, J.,Angsantikul, P., Zhang, Q., Hu, C.-M. J., and Zhang, L. (2015)Modulating antibacterial immunity via bacterial membrane-coatednanoparticles. Nano Lett. 15, 1403−1409.(131) Siefert, A. L., Caplan, M. J., and Fahmy, T. M. (2016) Artificialbacterial biomimetic nanoparticles synergize pathogen-associatedmolecular patterns for vaccine efficacy. Biomaterials 97, 85−96.(132) Dehaini, D., Fang, R. H., and Zhang, L. (2016) Biomimeticstrategies for targeted nanoparticle delivery. Bioeng. Transl. Med. 1,30−46.(133) Kim, O. Y., Dinh, N. T. H., Park, H. T., Choi, S. J., Hong, K.,and Gho, Y. S. (2017) Bacterial protoplast-derived nanovesicles fortumor targeted delivery of chemotherapeutics. Biomaterials 113, 68−79.(134) Paukner, S., Kohl, G., and Lubitz, W. (2004) Bacterial ghosts asnovel advanced drug delivery systems: antiproliferative activity ofloaded doxorubicin in human Caco-2 cells. J. Controlled Release 94,63−74.(135) Kudela, P., Paukner, S., Mayr, U. B., Cholujova, D.,Schwarczova, Z., Sedlak, J., Bizik, J., and Lubitz, W. (2005) Bacterialghosts as novel efficient targeting vehicles for DNA delivery to thehuman monocyte-derived dendritic cells. J. Immunother. 28, 136−143.(136) Cohen, F. S. (2016) How viruses invade cells. Biophys. J. 110,1028−1032.(137) Orlova, E. V. (2009) How viruses infect bacteria? EMBO J. 28,797−798.(138) Perelson, A. S. (2002) Modelling viral and immune systemdynamics. Nat. Rev. Immunol. 2, 28−36.(139) Braciale, T. J., Sun, J., and Kim, T. S. (2012) Regulating theadaptive immune response to respiratory virus infection. Nat. Rev.Immunol. 12, 295−305.(140) McGill, J., Heusel, J. W., and Legge, K. L. (2009) Innateimmune control and regulation of influenza virus infections. J.Leukocyte Biol. 86, 803−812.

(141) McMichael, A. J., and Phillips, R. E. (1997) Escape of humanimmunodeficiency virus from immune control. Annu. Rev. Immunol.15, 271−296.(142) Nowak, M. A., and Bangham, C. R. (1996) Populationdynamics of immune responses to persistent viruses. Science 272, 74−79.(143) Jonjic, S., Babic, M., Polic, B., and Krmpotic, A. (2008)Immune evasion of natural killer cells by viruses. Curr. Opin. Immunol.20, 30−38.(144) Waehler, R., Russell, S. J., and Curiel, D. T. (2007) Engineeringtargeted viral vectors for gene therapy. Nat. Rev. Genet. 8, 573−587.(145) Kotterman, M. A., Chalberg, T. W., and Schaffer, D. V. (2015)Viral vectors for gene therapy: translational and clinical outlook. Annu.Rev. Biomed. Eng. 17, 63−89.(146) Bouard, D., Alazard-Dany, N., and Cosset, F. L. (2009) Viralvectors: from virology to transgene expression. Br. J. Pharmacol. 157,153−165.(147) Nayak, S., and Herzog, R. W. (2010) Progress and prospects:immune responses to viral vectors. Gene Ther. 17, 295−304.(148) Xu, L., Liu, Y., Chen, Z., Li, W., Liu, Y., Wang, L., Ma, L., Shao,Y., Zhao, Y., and Chen, C. (2013) Morphologically virus-like fullerenolnanoparticles act as the dual-functional nanoadjuvant for HIV-1vaccine. Adv. Mater. 25, 5928−5936.(149) Yoo, J. W., Irvine, D. J., Discher, D. E., and Mitragotri, S.(2011) Bio-inspired, bioengineered and biomimetic drug deliverycarriers. Nat. Rev. Drug Discovery 10, 521−535.(150) Ruff, Y., Moyer, T., Newcomb, C. J., Demeler, B., and Stupp, S.I. (2013) Precision templating with DNA of a virus-like particle withpeptide nanostructures. J. Am. Chem. Soc. 135, 6211−6219.(151) Zhang, P., Chen, Y., Zeng, Y., Shen, C., Li, R., Guo, Z., Li, S.,Zheng, Q., Chu, C., Wang, Z., et al. (2015) Virus-mimetic nanovesiclesas a versatile antigen-delivery system. Proc. Natl. Acad. Sci. U. S. A. 112,E6129−E6138.(152) Brillault, L., Jutras, P. V., Dashti, N., Thuenemann, E. C.,Morgan, G., Lomonossoff, G. P., Landsberg, M. J., and Sainsbury, F.(2017) Engineering recombinant virus-like nanoparticles from plantsfor cellular delivery. ACS Nano 11, 3476−3484.(153) Ni, R., and Chau, Y. (2014) Structural mimics of virusesthrough peptide/DNA co-assembly. J. Am. Chem. Soc. 136, 17902−17905.(154) Mammadov, R., Cinar, G., Gunduz, N., Goktas, M., Kayhan,H., Tohumeken, S., Topal, A. E., Orujalipoor, I., Delibasi, T., Dana, A.,et al. (2015) Virus-like nanostructures for tuning immune response.Sci. Rep. 5, 16728−16742.(155) Plummer, E. M., and Manchester, M. (2011) Viral nano-particles and virus-like particles: platforms for contemporary vaccinedesign. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 3, 174−196.(156) Deng, J., Xu, S., Hu, W., Xun, X., Zheng, L., and Su, M. (2018)Tumor targeted, stealthy and degradable bismuth nanoparticles forenhanced X-ray radiation therapy of breast cancer. Biomaterials 154,24−33.(157) Molinaro, R., Corbo, C., Martinez, J. O., Taraballi, F.,Evangelopoulos, M., Minardi, S., Yazdi, I. K., Zhao, P., De Rosa, E.,Sherman, M., et al. (2016) Biomimetic proteolipid vesicles fortargeting inflamed tissues. Nat. Mater. 15, 1037.(158) Cao, H., Dan, Z., He, X., Zhang, Z., Yu, H., Yin, Q., and Li, Y.(2016) Liposomes coated with isolated macrophage membrane cantarget lung metastasis of breast cancer. ACS Nano 10, 7738−7748.(159) Hu, Q., Sun, W., Qian, C., Wang, C., Bomba, H. N., and Gu, Z.(2015) Anticancer platelet-mimicking nanovehicles. Adv. Mater. 27,7043−7050.(160) Gujrati, V., Kim, S., Kim, S. H., Min, J. J., Choy, H. E., Kim, S.C., and Jon, S. (2014) Bioengineered bacterial outer membranevesicles as cell-specific drug-delivery vehicles for cancer therapy. ACSNano 8, 1525−1537.(161) Shan, W., Zhang, D., Wu, Y., Lv, X., Hu, B., Zhou, X., Ye, S., Bi,S., Ren, L., and Zhang, X. (2018) Modularized peptides modified HBcvirus-like particles for encapsulation and tumor-targeted delivery ofdoxorubicin. Nanomedicine 14, 725−734.

Bioconjugate Chemistry Review

DOI: 10.1021/acs.bioconjchem.8b00103Bioconjugate Chem. 2018, 29, 838−851

850

Page 14: Recent Advances of Membrane-Cloaked Nanoplatforms for … · 2018-05-05 · nanoplatforms inspired by diverse moieties, such as selective drug delivery in targeted sites and effective

(162) Fang, R. H., Luk, B. T., Hu, C. M. J., and Zhang, L. (2015)Engineered nanoparticles mimicking cell membranes for toxinneutralization. Adv. Drug Delivery Rev. 90, 69−80.(163) Green, D. R., Ferguson, T., Zitvogel, L., and Kroemer, G.(2009) Immunogenic and tolerogenic cell death. Nat. Rev. Immunol. 9,353−363.(164) Lu, Y., Aimetti, A. A., Langer, R., and Gu, Z. (2016)Bioresponsive materials. Nat. Rev. Mater. 2, 16075−16091.

Bioconjugate Chemistry Review

DOI: 10.1021/acs.bioconjchem.8b00103Bioconjugate Chem. 2018, 29, 838−851

851