Nanoparticle interaction with plasma proteins as it relates to particle biodistribution, biocompatibility and therapeutic efficacy

https://doi.org/10.1016/j.addr.2009.03.009Get rights and content

Abstract

Proteins bind the surfaces of nanoparticles, and biological materials in general, immediately upon introduction of the materials into a physiological environment. The further biological response of the body is influenced by the nanoparticle–protein complex. The nanoparticle's composition and surface chemistry dictate the extent and specificity of protein binding. Protein binding is one of the key elements that affects biodistribution of the nanoparticles throughout the body. Here we review recent research on nanoparticle physicochemical properties important for protein binding, techniques for isolation and identification of nanoparticle-bound proteins, and how these proteins can influence particle biodistribution and biocompatibility. Understanding the nanoparticle–protein complex is necessary for control and manipulation of protein binding, and allows for improved engineering of nanoparticles with favorable bioavailability and biodistribution.

Introduction

Nanotechnology has recently gained attention as one of the critical research endeavors of the 21st century [1]. According to the National Nanotechnology Initiative, nanotechnology is the research and development of nanosystems, such as nanoparticles, on the scale of 1–100 nm [1]. Particles of this size are potentially useful tools for medicine and biology, as they are of commensurate size to important biological components (e.g. DNA, proteins, cell membranes) and thus able to interact in a sophisticated and controlled way at the cellular level. The ability to manipulate particular nanoparticle features, such as their physical, chemical, and biological properties, opens up a host of possibilities for researchers interested in rationally designing these nanoparticles for use in drug delivery, as image contrast agents, and for diagnostic purposes.

Nanoparticles for such medical applications are frequently given via parenteral administration. As with any foreign material, the body mounts a biological response to an administered nanoparticle. This response is the result of a complex interplay of factors, not just the intrinsic characteristics of the nanoparticle. In particular, most materials, upon contact with biological matrices, are immediately coated by proteins, leading to a protein “corona” [2], [3], [4]. Protein coronas are complex and variable. The complete plasma proteome is expected to contain as many as 3700 proteins [5], [6], of which approximately 50 have been identified in association with various nanoparticles [7], [8], [9], [10].

Certain components of the nanoparticle corona, called opsonins, may enhance uptake of the coated material by cells of the reticuloendothelial system (RES) [11], [12]. The presence of opsonins on the particle surface creates a “molecular signature” which is recognized by immune cells and determines the route of particle internalization [13], [14]. The route of internalization, then, may affect the eventual fate of the nanoparticle in the body (i.e. its rate of clearance from the bloodstream, volume of distribution, organ disposition, and rate and route of clearance from the body) [15], [16].

The effect of this protein corona may have special significance for nanoparticles, due to the increased importance of surface effects for particles of this size. Several studies have shown that biological responses to nanoparticles tend to scale with surface area rather than mass [17], [18], [19], [20], [21]. As things become smaller, their surface areas shrink much more slowly than their volumes, causing nanoscale materials to have far greater surface-to-volume ratios than larger particles. A larger surface-to-volume ratio also implies more proteins will bind a nanoparticle (relative to its mass) than a particle of larger size.

Even for larger particles, protein binding is established as one of the most important factors influencing biodistribution [22], [23], [24], [25]. In current preclinical testing of pharmaceutical molecules, evaluation of plasma protein binding is recognized as an important element in an assessment of drug efficacy, safety, and disposition [26], [27]. It has been shown to be important for understanding the pharmacokinetics and pharmacodynamics of the drug inside the body [26], [28], [29], [30]. It is also used to extrapolate preclinical data to models that predict potential drug efficacy and/or toxicity in humans [26]. For biomaterials used as medical implants, it has long been understood that the nature of the deposited protein layer onto these medical devices is responsible for the early immunological response in patients [6]. In fact, even late stage biological responses are influenced and dictated by the surface composition of the material and how this surface interacts with surrounding tissue.

Of course, there are other factors that play a role in determining how nanoparticles distribute within the body. These include particle properties such as size, shape, surface charge (zeta potential), solubility, surface modifications (including targeting), and route of administration. There have been numerous review articles dwelling on the importance of these factors and how each one can influence biodistribution [11], [31], [32], [33], [34], [35], [36], [37], [38]. What is missing, however, is an understanding of how these factors influence plasma protein binding to nanoparticles, and the importance of nanoparticle–protein interactions to biodistribution, biocompatibility, and therapeutic efficacy of nanoparticles. The mechanism of protein binding is not well understood, nor is it currently known how more or less protein binding influences the biological response to a given nanoparticle (e.g. uptake by phagocytic cells of the RES and clearance). However, it is clear that both the amount and identities of proteins on the particle surface play a part in affecting the biodistribution of nanoparticles. To fully understand the protein corona, one must understand not only which proteins are attached to the particle, but the kinetics, affinities, and stoichiometries of protein association and dissociation with the nanoparticle [2].

This review article will cover these aspects of protein binding to nanoparticles. We will discuss methods used to isolate and identify proteins bound to nanoparticles, what factors influence protein binding, how these factors can be manipulated to enhance or decrease protein binding for drug development, the kinetics of protein binding, and the importance of protein binding to drug delivery and distribution throughout the body.

Section snippets

Methods for nanoparticle separation from plasma

Many methods are available and have been used to identify particle-bound proteins. One common challenge is the isolation of the nanoparticle–protein complex from excess proteins without disrupting the complex or inducing additional protein binding [2], [39]. For most protein identification studies, nanoparticles are incubated with plasma so that the plasma levels are in excess of the available particle surface area. This nanoparticle/protein ratio is more representative of the true biological

Protein separation and identification

Once the nanoparticle–protein complex has been separated from excess plasma proteins, the proteins bound to the nanoparticle surface need to be individually separated and identified. A common technique for the separation of the proteins is two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) [23], [46], [47] (Fig. 1). To identify individual proteins, it is common practice to compare the 2D protein gels to a 2D master map of human plasma proteins [8], [9], [15], [23], [25], [47], [48],

Kinetics of protein binding

The composition of the protein corona on a given nanoparticle, at a given time, depends on the concentrations and kinetic properties of the proteins found in plasma. Therefore, it is important to not only determine which proteins are adsorbed onto the surface of the nanoparticle, but also understand the binding affinities and stoichiometries. Protein affinities for nanoparticle surfaces likely differ from their affinities to analogous bulk materials due to a variety of effects including size,

Properties that influence protein binding

The majority of studies examining the influence of protein binding on uptake have been conducted by either preincubating particles with bulk serum/plasma, or by preincubating particles with individual proteins or attaching individual proteins to the surface of the particles, and evaluating uptake by macrophages. This research has showed that neutrally charged particles have a distinctively slower opsonization rate than charged particles, demonstrating a direct correlation between surface charge

Effect of protein binding on nanoparticle biodistribution

Just as the properties of a nanoparticle influence the protein binding profile, the binding profile influences biodistribution. Protein binding can cause a change in nanoparticle size and surface charge [22], [31], [34], [58]. These changes affect the internalization process of these nanoparticles into macrophages and the overall distribution throughout the body. While many studies have considered the importance of physical characteristics (i.e. size, surface charge, etc.) for biodistribution

Use in medical applications

Knowledge of specific proteins that are adsorbed onto particle surfaces and their biological effects, including biodistribution and targeting, can be very useful when attempting to design drug candidates. Besides the ability to design particles to interact with particular proteins, one can attach these proteins specifically to a nanoparticle to obtain the desired targeting effect. A good example of this technology at work is the FDA approved drug Abraxane™. Abraxane is a nanoparticle

Conclusion

A first step in fully utilizing and developing nanoparticles as successful drug candidates is understanding the nature of the nanoparticle–protein complex and realizing that the nature of the proteins adsorbed onto the particles (encompassing the protein corona) influence nanoparticle uptake and traffic throughout the body. The ability to manipulate nanoparticles to achieve particular medical functions depends on this understanding. Designing nanoparticles to specifically adsorb certain

Acknowledgements

We are grateful to Allen Kane for assistance with illustrations. This project has been funded in whole or in part with federal funds from the National Cancer Institute, National Institutes of Health, under contract N01-CO-12400. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the US Government.

References (133)

  • R.E. Olson et al.

    Plasma protein binding of drugs

    Annu. Rep. Med. Chem.

    (1996)
  • M.J. Banker et al.

    Development and validation of a 96-well equilibrium dialysis apparatus for measuring plasma protein binding

    J. Pharm. Sci.

    (2003)
  • I. Kariv et al.

    Development of a high throughput equilibrium dialysis method

    J. Pharm. Sci.

    (2001)
  • S. Sarre et al.

    The use of microdialysis for the determination of plasma protein binding of drugs

    J. Pharm. Biomed. Anal.

    (1992)
  • W.H. De Jong et al.

    Particle size-dependent organ distribution of gold nanoparticles after intravenous administration

    Biomaterials

    (2008)
  • R.W. Mahley et al.

    Plasma lipoproteins: apolipoprotein structure and function

    J. Lipid Res.

    (1984)
  • M.A. Eriksson et al.

    Studies of drug binding to plasma proteins using a variant of equilibrium dialysis

    J. Pharm. Biomed. Anal.

    (2005)
  • F. Zsila et al.

    Selective plasma protein binding of antimalarial drugs to alpha1-acid glycoprotein

    Bioorg. Med. Chem.

    (2008)
  • K. Seehof et al.

    Interactions of nanoparticles with body proteins—improvement of 2D-PAGE-analysis by internal standard

    Int. J. Pharm.

    (2000)
  • R. Gref et al.

    ‘Stealth’ corona–core nanoparticles surface modified by polyethylene glycol (PEG): influences of the corona (PEG chain length and surface density) and of the core composition on phagocytic uptake and plasma protein adsorption

    Colloids Surf., B Biointerfaces

    (2000)
  • C. Lemarchand et al.

    Influence of polysaccharide coating on the interactions of nanoparticles with biological systems

    Biomaterials

    (2006)
  • M. Luck et al.

    Plasma protein adsorption on biodegradable microspheres consisting of poly(d,l-lactide-co-glycolide), poly(l-lactide) or ABA triblock copolymers containing poly(oxyethylene). Influence of production method and polymer composition

    J. Control. Release

    (1998)
  • M.T. Peracchia et al.

    Visualization of in vitro protein-rejecting properties of PEGylated stealth polycyanoacrylate nanoparticles

    Biomaterials

    (1999)
  • M. Ehrenberg et al.

    Binding between particles and proteins in extracts: implications for microrheology and toxicity

    Acta. Biomater.

    (2005)
  • M.E. Norman et al.

    Influence of block copolymers on the adsorption of plasma proteins to microspheres

    Biomaterials

    (1993)
  • S. Stolnik et al.

    The effect of surface coverage and conformation of poly(ethylene oxide) (PEO) chains of poloxamer 407 on the biological fate of model colloidal drug carriers

    Biochim. Biophys. Acta

    (2001)
  • S. Nagayama et al.

    Time-dependent changes in opsonin amount associated on nanoparticles alter their hepatic uptake characteristics

    Int. J. Pharm.

    (2007)
  • M.M. Frank et al.

    The role of complement in inflammation and phagocytosis

    Immunol. Today

    (1991)
  • D.E. Owens et al.

    Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles

    Int. J. Pharm.

    (2006)
  • S.M. Moghimi et al.

    Stealth liposomes and long circulating nanoparticles: critical issues in pharmacokinetics, opsonization and protein-binding properties

    Prog. Lipid Res.

    (2003)
  • L. Vroman et al.

    Interaction of high molecular weight kininogen, factor XII, and fibrinogen in plasma at interfaces

    Blood

    (1980)
  • S. Harnisch et al.

    Adsorption kinetics of plasma proteins on oil-in-water emulsions for parenteral nutrition

    Eur. J. Pharm. Biopharm.

    (2000)
  • H.S. Liu et al.

    The sorption of lysozyme and ribonuclease onto ferromagnetic nickel powder. 2. Desorption and competitive adsorption

    Colloids Surf., B Biointerfaces

    (1995)
  • S. Patil et al.

    Protein adsorption and cellular uptake of cerium oxide nanoparticles as a function of zeta potential

    Biomaterials

    (2007)
  • G. Yin et al.

    Impacts of the surface charge property on protein adsorption on hydroxyapatite

    Chem. Eng. J.

    (2002)
  • M. Roser et al.

    Surface-modified biodegradable albumin nano- and microspheres. II: Effect of surface charges on in vitro phagocytosis and biodistribution in rats

    Eur. J. Pharm. Biopharm.

    (1998)
  • D. Labarre et al.

    Interactions of blood proteins with poly(isobutylcyanoacrylate) nanoparticles decorated with a polysaccharidic brush

    Biomaterials

    (2005)
  • R.H. Muller et al.

    In vitro characterization of poly (methyl-methacrylate) nanoparticles and correlation to their in vivo fate

    J. Control. Release

    (1992)
  • M.E. Norman et al.

    Human serum-albumin as a probe for surface conditioning (opsonization) of block copolymer-coated microspheres

    Biomaterials

    (1992)
  • P. Asuri et al.

    The protein–nanomaterial interface

    Curr. Opin. Biotechnol.

    (2006)
  • E. Gabellieri et al.

    Dendrimer–protein interactions studied by tryptophan room temperature phosphorescence

    Biochim. Biophys. Acta

    (2006)
  • B. Klajnert et al.

    The effect of polyamidoamine dendrimers on human erythrocyte membrane acetylcholinesterase activity

    Bioelectrochemistry

    (2004)
  • D. Shcharbin et al.

    Effect of dendrimers on pure acetylcholinesterase activity and structure

    Bioelectrochemistry

    (2006)
  • J.C. Leroux et al.

    An investigation on the role of plasma and serum opsonins on the internalization of biodegradable poly(d,l-lactic acid) nanoparticles by human monocytes

    Life Sci.

    (1995)
  • R. Gref et al.

    The controlled intravenous delivery of drugs using peg-coated sterically stabilized nanospheres

    Adv. Drug Deliv. Rev.

    (1995)
  • L. Illum et al.

    The organ distribution and circulation time of intravenously injected colloidal carriers sterically stabilized with a block copolymer–poloxamine 908

    Life Sci.

    (1987)
  • Z. Panagi et al.

    Effect of dose on the biodistribution and pharmacokinetics of PLGA and PLGA-mPEG nanoparticles

    Int. J. Pharm.

    (2001)
  • National Nanotechnology Initiative
  • T. Cedervall et al.

    Understanding the nanoparticle–protein corona using methods to quantify exchange rates and affinities of proteins for nanoparticles

    Proc. Natl. Acad. Sci. U. S. A.

    (2007)
  • T. Cedervall et al.

    Detailed identification of plasma proteins adsorbed on copolymer nanoparticles

    Angew. Chem., Int. Ed. Engl.

    (2007)
  • Cited by (1538)

    View all citing articles on Scopus

    This review is part of the Advanced Drug Delivery Reviews theme issue on “Identifying and Assessing Biomaterial Nanotoxicity in Translational Research for Preclinical Drug Development.”

    View full text