scispace - formally typeset
Search or ask a question
Journal ArticleDOI

Erythrocyte-Platelet Hybrid Membrane Coating for Enhanced Nanoparticle Functionalization.

TL;DR: A new type of biological coating is created by fusing membrane material from two different cells, providing a facile method for further enhancing nanoparticle functionality, and opens the door for the creation of biocompatible, custom‐tailored biomimetic nanoparticles with varying hybrid functionalities.
Abstract: Cell-membrane-coated nanoparticles have recently been studied extensively for their biological compatibility, retention of cellular properties, and adaptability to a variety of therapeutic and imaging applications. This class of nanoparticles, which has been fabricated with a variety of cell membrane coatings, including those derived from red blood cells (RBCs), platelets, white blood cells, cancer cells, and bacteria, exhibit properties that are characteristic of the source cell. In this study, a new type of biological coating is created by fusing membrane material from two different cells, providing a facile method for further enhancing nanoparticle functionality. As a proof of concept, the development of dual-membrane-coated nanoparticles from the fused RBC membrane and platelet membrane is demonstrated. The resulting particles, termed RBC-platelet hybrid membrane-coated nanoparticles ([RBC-P]NPs), are thoroughly characterized, and it is shown that they carry properties of both source cells. Further, the [RBC-P]NP platform exhibits long circulation and suitability for further in vivo exploration. The reported strategy opens the door for the creation of biocompatible, custom-tailored biomimetic nanoparticles with varying hybrid functionalities, which may be used to overcome the limitations of current nanoparticle-based therapeutic and imaging platforms.

Content maybe subject to copyright    Report

Erythrocyte-Platelet Hybrid Membrane Coating for Enhanced
Nanoparticle Functionalization
Diana Dehaini, Xiaoli Wei, Ronnie H. Fang, Sarah Masson, Pavimol Angsantikul, Brian T.
Luk, Yue Zhang, Man Ying, Yao Jiang, Ashley V. Kroll, Weiwei Gao, and Liangfang Zhang
*
Department of NanoEngineering and Moores Cancer Center, University of California, San Diego,
La Jolla, CA 92093, U.S.A
Table of content entry
Biomimetic dual membrane-functionalized nanoparticles, incorporating the natural properties of
two different cell types, are fabricated by a facile process employing fused cell membranes. The
resulting hybrid cell membrane-coated nanoparticles retain protein markers from each source cell
and combine the unique functions of both. The reported approach opens the door for the
fabrication of biocompatible nanocarriers with increasingly complex functionality.
Keywords
nanomedicine; biomimetic nanoparticle; membrane fusion; long circulation; targeted delivery
Nanotechnology has increasingly been employed for the design of drug delivery and
imaging modalities in order to further improve efficacy in the clinic. There are many classes
of nanoscale delivery vehicles, including liposomes, polymeric nanoparticles, and inorganic
zhang@ucsd.edu, Tel: +1-858-246-0999.
HHS Public Access
Author manuscript
Adv Mater
. Author manuscript; available in PMC 2018 April 01.
Published in final edited form as:
Adv Mater
. 2017 April ; 29(16): . doi:10.1002/adma.201606209.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

systems among many others.
[1–4]
For applications such as cancer treatment, the use of such
nanoparticle platforms has the potential to enable greatly enhanced potency while
minimizing the systemic toxicity associated with traditional therapeutics.
[5–9]
Recently, a
new class of drug nanocarrier has been reported that is fabricated by combining synthetic
nanoparticulate cores with a biologically derived membrane coating.
[10–13]
These
biomimetic, cell membrane-coated nanoparticles directly leverage the versatility and
complexity of cellular membrane, which has been crafted by nature through the process of
evolution to perform specific functions, especially with regards to biointerfacing.
[14–17]
By
translocating the entire membrane from a cell onto the surface of a nanoparticle, all
biologically relevant surface moieties are transferred, including those that can potentially be
used for immune evasion and targeting, two highly desirable properties. Additionally, it may
be possible to take advantage of cell-specific functionalities that are known to exist, but have
not been well characterized on a fundamental biological level.
[18–20]
The first platform of this type employed red blood cells (RBCs), also known as erythrocytes,
as the source material in order to achieve lengthened blood residency, and the resulting RBC
membrane-coated nanoparticles (RBCNPs) displayed significantly enhanced circulation
half-lives compared with a nanoparticle stabilized by a layer of polyethylene glycol
(PEG).
[21]
It was later demonstrated that these cell membrane-coated nanoparticles exhibited
right-side-out membrane orientation and included immunomodulatory markers such as
CD47 at the same density as the original RBCs.
[22]
RBCNPs have also been used for novel
detoxification applications, including for toxic nerve agents and bacterial toxins.
[23–26]
More
recently, many different cell types other than RBCs, including platelets, white blood cells,
cancer cells, stem cells, and even bacteria, have been used to source the membrane material,
each with its own set of unique characteristics.
[14,18,27–29]
Of note, expansion of the cell
membrane-coated platform to other cells has enabled the fabrication of naturally targeted
drug carriers.
[30]
This includes the use of cancer cells for homotypic delivery to the tumor of
origin, or platelets, which play a role in a wide array of different diseases that range from
atherosclerosis to bacterial infection.
While cell membrane coating is a powerful method of enhancing nanoparticle utility, it is
often desirable to introduce additional functionality depending on the specific application.
For example, while RBCNPs can circulate for extended periods of time, the addition of a
targeting ligand can help to improve localization to the desired target, such as a tumor.
[31]
Herein, we report on an approach for increasing the number of functions that can be
performed by a single membrane-coated nanocarrier
via
the simultaneous derivation of
functionality from multiple cell types. This novel approach, which involves the fusion of
natural cell membranes from different origins, represents a facile and effective means of
fashioning nanoparticles that can perform increasingly complex tasks within biologically
relevant contexts. Specifically, we aimed to combine the functionality of human-derived
RBCs and platelets (Figure 1A). The resulting RBC-platelet hybrid membrane-coated
nanoparticles ([RBC-P]NPs) were thoroughly characterized, and it was demonstrated that
they retained functionality that is characteristic of each individual cell type. Finally, we show
that these dual membrane-coated nanoparticles displayed
in vivo
properties consistent with
expectations, warranting further study.
Dehaini et al.
Page 2
Adv Mater
. Author manuscript; available in PMC 2018 April 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

In order to fabricate the [RBC-P]NPs, the first step was to verify that it was possible to fuse
RBC membrane and platelet membrane together, the product of which would then be used as
the base material for downstream nanoparticle preparation. To test for fusion, platelet
membrane was doped with two different dyes that constituted a Förster resonance energy
transfer (FRET) pair and added to increasing amounts of RBC membrane under elevated
temperature and stirring (Figure 1B). It was observed that, as the amount of RBC material
increased, there was a recovery of fluorescence at the lower emission wavelength around
534 nm, indicating the interspersing of the two membrane materials weakening the FRET
interactions in the original platelet membrane. Using a 1:1 protein weight ratio of RBC
membrane to platelet membrane for further study, we fabricated a batch of the hybrid [RBC-
P]NPs. RBC membrane labeled with a red fluorescent dye was fused with platelet
membrane that was labeled with a green fluorescent dye, and the resultant material was
coated onto preformed poly(lactic-
co
-glycolic acid) (PLGA) cores. When a dilute solution
of the [RBC-P]NPs was immobilized in glycerol and viewed under confocal microscopy,
significant colocalization of fluorescent signals was observed (Figure 1C). In stark contrast,
a mixture of RBCNPs and platelet membrane-coated nanoparticles (PNPs) fabricated with
the individual fluorescently labeled membranes exhibited distinct red and green punctates. It
was further demonstrated that the RBC and platelet membranes were retained on the [RBC-
P]NPs at a ratio nearly identical to the 1:1 input (Figure 1D). The results indicate that it was
indeed possible to fuse the two types of natural cell membrane and incorporate the material
of both onto the same nanoparticle.
Dynamic light scattering (DLS) was used to compare the [RBC-P]NPs with each single
membrane formulation of RBCNPs or PNPs (Figure 2A,B). It was observed that bare PLGA
cores, which were originally 80 nm in size, exhibited a very uniform increase in size of
approximately 20 nm after coating with each membrane. Additionally, the surface zeta
potential increased by over 10 mV for each of the three nanoparticle types compared with
PLGA cores, approaching the value of approximately −25 mV observed for each of the
different membrane vesicles. This phenomenon is commonly seen after membrane coating
and indicates shielding of the highly negative cores with the less negative outer membrane
surface.
[14,21]
When visualized under transmission electron microscopy (TEM), the
RBCNPs, PNPs, and hybrid [RBC-P]NPs all displayed a characteristic core-shell structure
that was consistent with the sizing results (Figure 2C). A set of assays were then carried out
to assess nanoparticle stability in various media. To determine stability of the hybrid
formulation in solution over time, samples were stored in either water or 1× phosphate
buffered saline (PBS, pH 7.4) and the size was measured over time (Figure 3A,B). The
[RBC-P]NPs, in addition to RBCNPs and PNPs, exhibited stable size over the 3 week
duration of the study in both solutions. In contrast, bare PLGA cores, which are only
stabilized by charge repulsion, immediately aggregated in PBS to the micron range.
Additionally, [RBC-P]NPs displayed little change in absorbance before and after incubation
with 100% serum, while bare PLGA cores again exhibited a large increase (Figure 3C).
Regarding long-term storage, the [RBC-P]NP formulation exhibited near identical size both
before freeze-drying and after resuspension (Figure 3D). Taken together, the characterization
data give strong physical evidence for successful coating by the fused membrane material,
Dehaini et al.
Page 3
Adv Mater
. Author manuscript; available in PMC 2018 April 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

with results in line with what would be expected given the properties of the single
membrane-coated RBCNPs and PNPs.
To analyze the overall protein content of the [RBC-P]NPs, SDS-PAGE was used to run RBC
membrane, platelet membrane, hybrid membrane, and all corresponding nanoparticle
formulations followed by Coomassie staining for visualization (Figure 4A). Compared with
RBCNPs and PNPs, the [RBC-P]NPs had a profile that represented the union of the two
single membrane formulations. As expected, all nanoparticles had protein profiles that
largely mirrored that of the corresponding membrane. To analyze specific protein markers,
western blotting analysis was carried out (Figure 4B). CD235a, a major RBC
sialoglycoprotein also known as glycophorin A,
[32]
as well as the blood group A antigen,
were present on RBCNPs and also to a lesser degree on [RBC-P]NPs. CD41 and CD61,
which collectively form integrin αIIbβ3,
[33]
are important for platelet adhesion as well as
activation, and both were present on PNPs and also to a lesser degree on [RBC-P]NPs.
Meanwhile, CD47,
[34,35]
an immunomodulatory protein responsible for inhibiting
macrophage uptake that is expressed by both cell types, was found at a near equivalent
degree on RBCNPs, PNPs, and [RBC-P]NPs. Additionally, immunogold staining followed
by TEM imaging provided visual evidence that a single [RBC-P]NP could simultaneously
present both RBC-specific and platelet-specific markers (Figure 4C). Ultimately, the protein
analysis carried out here indicates that functionalization with the fusion membrane can
bestow both RBC and platelet surface proteins onto the hybrid [RBC-P]NPs, although the
exact nature of the intraparticle distribution of individual protein markers at the molecular
level remains unknown.
Upon confirming successful transference of markers unique to both source cells, we next
sought to characterize the hybrid membrane coating on a functional level. Both RBCNPs
and PNPs, given the presence of immunomodulatory markers on their surface, have been
shown to be adept at minimizing macrophage uptake.
[14,22,36]
An uptake study using human
THP-1 monocytes differentiated into macrophage-like cells was thus carried out to evaluate
this property in [RBC-P]NPs (Figure 5A). Using flow cytometric analysis after incubation
with the macrophage-like cells, [RBC-P]NPs had low uptake consistent with both RBCNPs
and PNPs, whereas PLGA cores, without immunomodulatory membrane coatings, exhibited
a high degree of uptake. As an RBC-specific functional marker, we next assessed the level of
acetylcholinesterase activity (Figure 5B). The enzyme is important for regulating
neurotransmitter concentrations, and RBCNPs have been shown to be potent decoys capable
of neutralizing lethal doses of poisonous compounds that can deactivate the protein.
[25]
It
was confirmed that [RBC-P]NPs had an intermediate level of activity compared with
RBCNPs and PNPs, the latter of which had a negligible amount of activity. This was further
reflected in detoxification capacity, as the amount of dichlorvos, a model organophosphate,
left in solution after incubation with each nanoformulation was inversely related to
acetylcholinesterase activity (Figure 5C). Regarding platelet-specific functionality, the
cancer targeting properties of the hybrid [RBC-P]NPs was first evaluated. PNPs, given the
large number of disease-relevant binding markers present on their surface, have been shown
to be effective at targeting certain metastatic cancers.
[37]
Using both flow cytometry as well
as fluorescent imaging, it was demonstrated that [RBC-P]NPs bound to highly metastatic
MDA-MB-231 human breast cancer cells within the spectrum between RBCNPs and PNPs,
Dehaini et al.
Page 4
Adv Mater
. Author manuscript; available in PMC 2018 April 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

which exhibited low and high amounts of binding, respectively (Figure 5D,E). On the other
hand, normal HFF-1 human foreskin fibroblasts showed significantly lowered binding by all
nanoparticle types (Figure 5D), and this was further confirmed by a co-culture imaging
study using [RBC-P]NPs (Figure 5F). After binding to cancer cells, it is expected that the
hybrid nanoparticles would be endocytosed via the clathrin-dependent pathway.
[37]
To assess
in vivo
targeting ability, a mouse model of atherosclerosis, which platelets are known to
interact with, was employed. Fluorescence imaging of aortas collected after intravenous
nanoparticle administration revealed a trend consistent to what was observed for the
in vitro
studies (Figure 5G). Overall, the ability of [RBC-P]NPs to incorporate the functional
properties of both RBCs and platelets is encouraging and provides a strong indication that
other cell-specific functions should likewise be present on the dual membrane-coated
nanoparticles.
Finally, to evaluate the [RBC-P]NP formulation for potential
in vivo
application, the
circulation and biodistribution of the nanoparticles were evaluated using a mouse model.
Murine-derived RBCs and platelets, along with fluorescently labeled PLGA cores, were
used as the starting materials to fabricate RBCNPs, PNPs, and [RBC-P]NPs. To test the
circulation half-life, nanoparticles were administered intravenously via the tail vein, and
blood was sampled at increasing timepoints to evaluate remaining nanoparticle
concentration (Figure 6A). After fitting to a two-phase decay model, it was apparent that all
three formulations exhibited very similar circulation profiles. Indeed, both RBCNPs and
PNPs have previously been reported to circulate for extended periods of time upon
administration.
[21,37]
Numerical analysis indicated that RBCNPs, PNPs, and [RBC-P]NPs
had one-phase half-lives of 5.7, 5.7, and 6.4 hours and two-phase elimination half-lives of
42.4, 38.3, and 51.8 hours, respectively. All values were within error of each other (see
Experimental Section). To analyze biodistribution, nanoparticles were again administered
intravenously, and the mice were euthanized 24 hours afterwards in order to collect the
heart, lungs, kidneys, liver, and spleen for fluorescence analysis (Figure 6B). All three
nanoformulations, including the [RBC-P]NPs, displayed similar organ-level localization,
with the majority of the nanoparticles found in the liver and spleen as a result of
reticuloendothelial uptake. This pattern is consistent with what has been previously reported
for similar nanoformulations.
[14,21]
The uniformity in circulation and distribution profiles
between all experimental groups can likely be explained by the fact that, under normal
conditions, both RBCs and platelets are inherently designed to persist in the bloodstream for
extended periods of time; in healthy individuals, there is an absence of reactive substrates
and molecules that can contribute to accelerated clearance of either cell type.
In conclusion, we have successfully fabricated a new class of cell membrane-coated
nanocarrier that combines the function of two different cell types. It was confirmed that it is
possible to fuse RBC membrane and platelet membrane together, using it as a coating
material to fabricate hybrid [RBC-P]NPs. Physically, the resultant particles were similar to
pure, single membrane formulations of either RBCNPs or PNPs. On a protein and functional
level, the [RBC-P]NPs represented a cross between the two parent nanoparticle types,
retaining properties once only exclusive to either. The reported method of bestowing
nanoparticles with enhanced functionality provides a facile and natural alternative to
synthetic post-functionalization strategies. Importantly, the dual membrane-coated
Dehaini et al.
Page 5
Adv Mater
. Author manuscript; available in PMC 2018 April 01.
Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Citations
More filters
Journal ArticleDOI
TL;DR: There is still significant room for development, as researchers continue to refine existing workflows while finding new and exciting applications that can take advantage of this developing technology, cell‐membrane‐coating nanotechnology.
Abstract: Nanoparticle-based therapeutic, prevention, and detection modalities have the potential to greatly impact how diseases are diagnosed and managed in the clinic. With the wide range of nanomaterials available, the rational design of nanocarriers on an application-specific basis has become increasingly commonplace. Here, a comprehensive overview is provided on an emerging platform: cell-membrane-coating nanotechnology. As a fundamental unit of biology, cells carry out a wide range of functions, including the remarkable ability to interface and interact with their surrounding environment. Instead of attempting to replicate such functions via synthetic techniques, researchers are now directly leveraging naturally derived cell membranes as a means of bestowing nanoparticles with enhanced biointerfacing capabilities. This top-down technique is facile, highly generalizable, and has the potential to greatly augment existing nanocarriers. Further, the introduction of a natural membrane substrate onto nanoparticles surfaces has enabled additional applications beyond those traditionally associated with nanomedicine. Despite its relative youth, there exists an impressive body of literature on cell membrane coating, which is covered here in detail. Overall, there is still significant room for development, as researchers continue to refine existing workflows while finding new and exciting applications that can take advantage of this developing technology.

908 citations

Journal ArticleDOI
05 Jul 2017-ACS Nano
TL;DR: A cancer targeted cascade bioreactor was constructed by embedding glucose oxidase and catalase in the cancer cell membrane-camouflaged porphyrin metal-organic framework of PCN-224 to enhance its cancer targeting and retention abilities and displayed amplified synergistic therapeutic effects of long-term cancer starvation therapy and robust PDT.
Abstract: Selectively cuting off the nutrient supply and the metabolism pathways of cancer cells would be a promising approach to improve the efficiency of cancer treatment. Here, a cancer targeted cascade bioreactor (designated as mCGP) was constructed for synergistic starvation and photodynamic therapy (PDT) by embedding glucose oxidase (GOx) and catalase in the cancer cell membrane-camouflaged porphyrin metal–organic framework (MOF) of PCN-224 (PCN stands for porous coordination network). Due to biomimetic surface functionalization, the immune escape and homotypic targeting behaviors of mCGP would dramatically enhance its cancer targeting and retention abilities. Once internalized by cancer cells, mCGP was found to promote microenvironmental oxygenation by catalyzing the endogenous hydrogen peroxide (H2O2) to produce oxygen (O2), which would subsequently accelerate the decomposition of intracellular glucose and enhance the production of cytotoxic singlet oxygen (1O2) under light irradiation. Consequently, mCGP d...

593 citations

Journal ArticleDOI
TL;DR: The roles of nanoparticles and hybrid nanoparticles for drug delivery in chemotherapy, targeted therapy, and immunotherapy are discussed and the targeting mechanism of nanoparticle-based drug delivery as well as its function on reversing drug resistance are described.
Abstract: Nanotechnology has been extensively studied and exploited for cancer treatment as nanoparticles can play a significant role as a drug delivery system. Compared to conventional drugs, nanoparticle-based drug delivery has specific advantages, such as improved stability and biocompatibility, enhanced permeability and retention effect, and precise targeting. The application and development of hybrid nanoparticles, which incorporates the combined properties of different nanoparticles, has led this type of drug-carrier system to the next level. In addition, nanoparticle-based drug delivery systems have been shown to play a role in overcoming cancer-related drug resistance. The mechanisms of cancer drug resistance include overexpression of drug efflux transporters, defective apoptotic pathways, and hypoxic environment. Nanoparticles targeting these mechanisms can lead to an improvement in the reversal of multidrug resistance. Furthermore, as more tumor drug resistance mechanisms are revealed, nanoparticles are increasingly being developed to target these mechanisms. Moreover, scientists have recently started to investigate the role of nanoparticles in immunotherapy, which plays a more important role in cancer treatment. In this review, we discuss the roles of nanoparticles and hybrid nanoparticles for drug delivery in chemotherapy, targeted therapy, and immunotherapy and describe the targeting mechanism of nanoparticle-based drug delivery as well as its function on reversing drug resistance.

338 citations

Journal ArticleDOI
25 May 2018-ACS Nano
TL;DR: The DOX-loaded [RBC-B16]-coated CuS NP platform exhibits excellent synergistic photothermal/chemotherapy with about 100% melanoma tumor growth inhibition rate and may contribute to personalized nanomedicine of various tumors by combining the RBCs with a homotypic cancer membrane accordingly on the surface of the nanoparticle.
Abstract: Cellular-membrane-coated nanoparticles have increasingly been pursued to leverage the natural cell functions for enhancing biocompatibility and improved therapeutic efficacy. Taking advantage of specialized cell membranes or combining functions from different membrane types facilitates the strengthening of their functionality. Herein, we fuse membrane materials derived from red blood cells (RBCs) and melanoma cells (B16-F10 cells) to create a hybrid biomimetic coating (RBC–B16), and RBC–B16 hybrid membrane camouflaged doxorubicin (DOX)-loaded hollow copper sulfide nanoparticles (DCuS@[RBC–B16] NPs) are fabricated for combination therapy of melanoma. The DCuS@[RBC–B16] NPs are comprehensively characterized, showing the inherent properties of the both source cells. Compared to the bare CuS NPs, the DCuS@[RBC–B16] NPs exhibit highly specific self-recognition to the source cell line in vitro and achieve markedly prolonged circulation lifetime and enhanced homogeneous targeting abilities in vivo inherited from...

335 citations

Journal ArticleDOI
TL;DR: The therapeutic potential of a macrophage-like nanoparticle for sepsis control is demonstrated through a powerful two-step neutralization process: endotoxin neutralization in the first step followed by cytokine sequestration in the second step.
Abstract: Sepsis, resulting from uncontrolled inflammatory responses to bacterial infections, continues to cause high morbidity and mortality worldwide. Currently, effective sepsis treatments are lacking in the clinic, and care remains primarily supportive. Here we report the development of macrophage biomimetic nanoparticles for the management of sepsis. The nanoparticles, made by wrapping polymeric cores with cell membrane derived from macrophages, possess an antigenic exterior the same as the source cells. By acting as macrophage decoys, these nanoparticles bind and neutralize endotoxins that would otherwise trigger immune activation. In addition, these macrophage-like nanoparticles sequester proinflammatory cytokines and inhibit their ability to potentiate the sepsis cascade. In a mouse Escherichia coli bacteremia model, treatment with macrophage mimicking nanoparticles, termed MΦ-NPs, reduced proinflammatory cytokine levels, inhibited bacterial dissemination, and ultimately conferred a significant survival advantage to infected mice. Employing MΦ-NPs as a biomimetic detoxification strategy shows promise for improving patient outcomes, potentially shifting the current paradigm of sepsis management.

322 citations


Cites methods from "Erythrocyte-Platelet Hybrid Membran..."

  • ...With a lipid-like structure, they can be introduced onto MΦ-NPs through methods such as lipid insertion (38) or membrane hybridization (39), both of which have been validated for functionalizing nanoparticles coated with different cell membranes....

    [...]

References
More filters
Journal ArticleDOI
TL;DR: Lipidic nanoparticles are the first nanomedicine delivery system to make the transition from concept to clinical application, and they are now an established technology platform with considerable clinical acceptance.

3,497 citations

Journal ArticleDOI
27 Jan 2009-ACS Nano
TL;DR: This work focuses on the application of nanotechnology to drug delivery and highlights several areas of opportunity where current and emerging nanotechnologies could enable entirely novel classes of therapeutics.
Abstract: Nanotechnology is the engineering and manufacturing of materials at the atomic and molecular scale. In its strictest definition from the National Nanotechnology Initiative, nanotechnology refers to structures roughly in the 1−100 nm size regime in at least one dimension. Despite this size restriction, nanotechnology commonly refers to structures that are up to several hundred nanometers in size and that are developed by top-down or bottom-up engineering of individual components. Herein, we focus on the application of nanotechnology to drug delivery and highlight several areas of opportunity where current and emerging nanotechnologies could enable entirely novel classes of therapeutics.

2,783 citations

Journal ArticleDOI
TL;DR: This review explores recent work directed towards more targeted treatment of cancer, whether through more specific anti-cancer agents or through methods of delivery, including delivery by avoiding the reticuloendothelial system, utilizing the enhanced permeability and retention effect and tumor-specific targeting.

2,389 citations

Journal ArticleDOI
TL;DR: A top-down biomimetic approach in particle functionalization is reported by coating biodegradable polymeric nanoparticles with natural erythrocyte membranes, including both membrane lipids and associated membrane proteins for long-circulating cargo delivery.
Abstract: Efforts to extend nanoparticle residence time in vivo have inspired many strategies in particle surface modifications to bypass macrophage uptake and systemic clearance. Here we report a top-down biomimetic approach in particle functionalization by coating biodegradable polymeric nanoparticles with natural erythrocyte membranes, including both membrane lipids and associated membrane proteins for long-circulating cargo delivery. The structure, size and surface zeta potential, and protein contents of the erythrocyte membrane-coated nanoparticles were verified using transmission electron microscopy, dynamic light scattering, and gel electrophoresis, respectively. Mice injections with fluorophore-loaded nanoparticles revealed superior circulation half-life by the erythrocyte-mimicking nanoparticles as compared to control particles coated with the state-of-the-art synthetic stealth materials. Biodistribution study revealed significant particle retention in the blood 72 h following the particle injection. The translocation of natural cellular membranes, their associated proteins, and the corresponding functionalities to the surface of synthetic particles represents a unique approach in nanoparticle functionalization.

1,614 citations

Journal ArticleDOI
16 Jun 2000-Science
TL;DR: It is shown that CD47 (integrin-associated protein) functions as a marker of self on murine red blood cells and may represent a potential pathway for the control of hemolytic anemia.
Abstract: The immune system recognizes invaders as foreign because they express determinants that are absent on host cells or because they lack "markers of self" that are normally present. Here we show that CD47 (integrin-associated protein) functions as a marker of self on murine red blood cells. Red blood cells that lacked CD47 were rapidly cleared from the bloodstream by splenic red pulp macrophages. CD47 on normal red blood cells prevented this elimination by binding to the inhibitory receptor signal regulatory protein alpha (SIRPalpha). Thus, macrophages may use a number of nonspecific activating receptors and rely on the presence or absence of CD47 to distinguish self from foreign. CD47-SIRPalpha may represent a potential pathway for the control of hemolytic anemia.

1,556 citations

Related Papers (5)