Elsevier

Advanced Drug Delivery Reviews

Volume 143, 15 March 2019, Pages 68-96
Advanced Drug Delivery Reviews

Concepts of nanoparticle cellular uptake, intracellular trafficking, and kinetics in nanomedicine

https://doi.org/10.1016/j.addr.2019.04.008Get rights and content

Abstract

Nanoparticle-based therapeutics and diagnostics are commonly referred to as nanomedicine and may significantly impact the future of healthcare. However, the clinical translation of these technologies is challenging. One of these challenges is the efficient delivery of nanoparticles to specific cell populations and subcellular targets in the body to elicit desired biological and therapeutic responses. It is critical for researchers to understand the fundamental concepts of how nanoparticles interact with biological systems to predict and control in vivo nanoparticle transport for improved clinical benefit. In this overview article, we review and discuss cellular internalization pathways, summarize the field`s understanding of how nanoparticle physicochemical properties affect cellular interactions, and explore and discuss intracellular nanoparticle trafficking and kinetics. Our overview may provide a valuable resource for researchers and may inspire new studies to expand our current understanding of nanotechnology-biology interactions at cellular and subcellular levels with the goal to improve clinical translation of nanomedicines.

Introduction

The design and medical application of nanoparticles for diagnosis and treatment of diseases represent an important area of current nanotechnology research. This research field has been widely referred to as nanomedicine [1]. In nanomedicine, researchers engineer nanoparticles, for example, as delivery vehicles for therapeutics or imaging agents with the ultimate goal to improve clinical outcomes [2]. To achieve this goal, researchers need to be able to efficiently deliver nanoparticles to diseased sites in the body with cellular specificity and oftentimes subcellular precision [3]. Such efficient and effective nanomedicine delivery requires full control over the nanoparticle transport in the body. However, this level of control has not been achieved yet and is one of the greatest challenges in nanomedicine research [4].

Addressing this challenge is a major quest in the field which emphasizes the need to better understand the fundamental concepts of how nanoparticles interact with biological systems [5]. These nanotechnology-biology (i.e., nano-bio) interactions are complex, dynamic, and multiparametric, which poses substantial obstacles for the engineering of effective nanomedicines [6]. Factors that contribute to this complexity are manifold and include: (i) a nanoparticle’s physicochemical properties, including size, shape, surface chemistry, composition, architecture, density, and modulus; (ii) the biological and biochemical environments, including type of organ/tissue, biomolecular milieu and composition, pH, and other biochemical factors; and (iii) the interplay and interactions between these individual nanoparticle properties and biological/biochemical parameters, including the kinetics of nano-bio interactions [7].

While researchers are able to synthesize colloidal nanoparticles in the laboratory with precise physicochemical properties and functions, these deliberately designed nanoparticle characteristics may change substantially upon introduction of nanoparticles into a biological environment [8,9]. This phenomenon can be observed, for example, when nanoparticles are administered into the body through intravenous injection. Upon contact with blood, serum proteins adsorb non-specifically onto the nanoparticle surface to form a so-called protein corona [10,11]. The protein corona alters nanoparticles’ physicochemical properties by providing them with an unintentional biological identity [12]. Ultimately, this biological identity determines a nanoparticle’s interactions with biological systems, including organs, tissues, cells, and subcellular organelles [[13], [14], [15], [16]]. Therefore, nanoparticle in vivo transport and biodistribution are largely controlled by this biological identity rather than the deliberately engineered synthetic nanoparticle characteristics [17,18].

The fact that a nanoparticle’s physicochemical properties may change significantly upon biological exposure imposes major challenges for the engineering of nanomedicines. To advance our current understanding and to develop fundamental concepts needed for the design of more effective nanomedicines, researchers have started to describe and decipher essential mechanisms of how nanoparticles interact with biological systems. These studies can be divided into three categories: (i) nanoparticle interactions at organ and tissue levels; (ii) nanoparticle interactions at cellular and subcellular levels; and (iii) nanoparticle interactions with biomolecules and biochemical parameters. We focus in our review article on the second category, i.e., cellular and subcellular interactions of nanoparticles, and refer interested readers to excellent overview articles and original papers that cover nano-bio interactions at organ, tissue, and biomolecular levels [11,[19], [20], [21], [22], [23], [24], [25]].

To maximize clinical benefits of nanomedicines while minimizing side effects, researchers require profound understanding of nanoparticles’ cellular and subcellular interactions [19]. An intriguing example is the engineering of nanoparticles that are able to distinguish between healthy and diseased cells through the use of precise biomolecular recognition strategies [26,27]. To achieve this level of cellular identification and discrimination, a nanoparticle surface can be decorated with specific biomolecular ligands that can recognize and bind to complementary cell surface receptors on targeted cells [28]. The idea behind this concept is that upon recognition nanoparticles may deliver their payloads (e.g., active pharmaceutical ingredients; APIs; and imaging agents) preferentially to diseased cells while leaving healthy cells mostly unaffected. As some types of nanoparticle payloads require delivery to specific intracellular targets for maximizing efficacy, it is critical for researchers to understand and explore nanoparticles’ cellular interactions, intracellular trafficking pathways, and corresponding kinetics to ensure targeted delivery [[29], [30], [31], [32], [33]].

In this review, we describe the field’s understanding of three distinct aspects of nanoparticle-cell interactions: (i) nanoparticle cellular uptake; (ii) nanoparticle intracellular trafficking; and (iii) underlying kinetics of these cellular and subcellular nano-bio interactions. We hope that our review of these important concepts provides a valuable resource to researchers in the nanomedicine field and inspires new research to further enrich our knowledge of cellular and subcellular nanoparticle interactions. With improved knowledge and understanding, better control over nanoparticle transport in the body may be achieved, which could ultimately result in improved clinical benefits of nanomedicines.

Section snippets

Cellular uptake of nanoparticles

Cellular uptake of nanoparticles involves highly regulated mechanisms with complex biomolecular interactions to overcome the cell plasma membrane. This biological membrane acts as a barrier and separates a cell’s interior from the outside environment. Structural and biomolecular membrane characteristics (i.e., phospholipid-based bilayer membrane littered with proteins and other biomolecules) result in an overall negative charge of the plasma membrane with few cationic domains and selective

Mediating nanoparticle uptake through material design

As shown in Table 1 for gold nanoparticles, cells internalize nanoparticles through multiple different uptake routes even if the nanomaterial is kept constant. These findings suggest that biological factors, including cell type, affect nanoparticle uptake pathways significantly (Table 1).

A study by Saha and coworkers showed that healthy and diseased cells uptake nanoparticles using different pathways. In more detail, healthy mammary epithelium cells and cancerous HeLa cells were each incubated

Intracellular trafficking of nanoparticles

After cellular internalization, nanoparticles undergo transport and trafficking to various intracellular destinations. If nanoparticle cellular uptake occurs via endocytic pathways, nanoparticles are confined within a membrane-lined vesicle, such as an endosome (Fig. 1). These vesicles transport throughout the cell in complex trafficking patterns. Currently used methods for probing the intracellular trafficking of nanoparticles include optical- and electron-based microscopy techniques, such as

Kinetics of nanoparticle-cell interactions

As discussed in previous chapters, nanoparticle cellular uptake and intracellular transport depend on both nanoparticle physicochemical properties (e.g., size, shape, composition, surface chemistry) and characteristics of the biological system, including cell type and function. While nanoparticle-cell interactions are prerequisites for effective application of nanomedicines in vitro and in vivo, the rates at which these interactions occur are complex and need to be investigated and understood (

Future directions and conclusions

Nanoparticles can be engineered from inorganic and organic materials with unique physical, chemical, and biological properties for applications in medicine. Once administered into the body, nanoparticles interact with different tissues and cells. While specific and efficient delivery of nanoparticles to diseased tissue sites and cells in the body is challenging [17,18], nanomedicine offers the potential to transform diagnostic and therapeutic strategies. However, more quantitative studies that

Acknowledgements

We thank Erika Green and Simon Liang for helpful discussions. We also thank Biorender for their technical support in creating some of the figures. HA and SW acknowledge funding support by the Vice President for Research of the University of Oklahoma (Junior Faculty Fellowship).

References (342)

  • B. Yameen et al.

    Insight into nanoparticle cellular uptake and intracellular targeting

    J. Control. Release

    (2014)
  • X. Chen et al.

    Nucleolin-mediated cellular trafficking of DNA nanoparticle is lipid raft and microtubule dependent and can be modulated by glucocorticoid

    Mol. Ther.

    (2011)
  • L.M. Stuart et al.

    Phagocytosis: elegant complexity

    Immunity

    (2005)
  • G. Sahay et al.

    Endocytosis of nanomedicines

    J. Control. Release

    (2010)
  • Y.N. Zhang et al.

    Nanoparticle–liver interactions: cellular uptake and hepatobiliary elimination

    J. Control. Release

    (2016)
  • Y. Li et al.

    Endocytosis of PEGylated nanoparticles accompanied by structural and free energy changes of the grafted polyethylene glycol

    Biomaterials

    (2014)
  • N.G. Sosale et al.

    “Marker of Self” CD47 on lentiviral vectors decreases macrophage-mediated clearance and increases delivery to SIRPA-expressing lung carcinoma tumors

    Mol. Ther. Methods Clin. Dev.

    (2016)
  • S. Hirosue et al.

    Antigen delivery to dendritic cells by poly(propylene sulfide) nanoparticles with disulfide conjugated peptides: cross-presentation and T cell activation

    Vaccine

    (2010)
  • P. Guterstam et al.

    Elucidating cell-penetrating peptide mechanisms of action for membrane interaction, cellular uptake, and translocation utilizing the hydrophobic counter-anion pyrenebutyrate

    BBA Biomembr.

    (2009)
  • W.B. Kauffman et al.

    Mechanism matters: a taxonomy of cell penetrating peptides

    Trends Biochem. Sci.

    (2015)
  • J.M. Steinbach et al.

    Cell penetrating peptide-modified poly (lactic-co-glycolic acid) nanoparticles with enhanced cell internalization

    Acta Biomater.

    (2016)
  • B.Y.S. Kim et al.

    Engl. J. Med.

    (2010)
  • B. Pelaz et al.

    Diverse Applications of Nanomedicine

    ACS Nano

    (2017)
  • M. Björnmalm et al.

    Bridging bio-nano science and cancer nanomedicine

    ACS Nano

    (2017)
  • D. Rosenblum et al.

    Progress and challenges towards targeted delivery of cancer therapeutics

    Nat. Commun.

    (2018)
  • A.E. Nel et al.

    Understanding biophysicochemical interactions at the nano-bio interface

    Nat. Mater.

    (2009)
  • P.C. Ke et al.

    A Decade of the Protein Corona

    ACS Nano

    (2017)
  • J. Hühn et al.

    Selected standard protocols for the synthesis, phase transfer, and characterization of inorganic colloidal nanoparticles

    Chem. Mater.

    (2017)
  • C.D. Walkey et al.

    Understanding and controlling the interaction of nanomaterials with proteins in a physiological environment

    Chem. Soc. Rev.

    (2012)
  • A. Albanese et al.

    The effect of nanoparticle size, shape, and surface chemistry on biological systems

    Annu. Rev. Biomed. Eng.

    (2012)
  • M.P. Monopoli et al.

    Biomolecular coronas provide the biological identity of nanosized materials

    Nat. Nanotechnol.

    (2012)
  • C.C. Fleischer et al.

    Nanoparticle-cell interactions: molecular structure of the protein corona and cellular outcomes

    Acc. Chem. Res.

    (2014)
  • C.D. Walkey et al.

    Protein corona fingerprinting predicts the cellular interaction of gold and silver nanoparticles

    ACS Nano

    (2014)
  • C.D. Walkey et al.

    Nanoparticle size and surface chemistry determine serum protein adsorption and macrophage uptake

    J. Am. Chem. Soc.

    (2012)
  • Q. Dai et al.

    Quantifying the ligand-coated nanoparticle delivery to cancer cells in solid tumours

    ACS Nano

    (2018)
  • S. Wilhelm et al.

    Analysis of nanoparticle delivery to tumours

    Nat. Rev. Mater.

    (2016)
  • E. Blanco et al.

    Principles of nanoparticle design for overcoming biological barriers to drug delivery

    Nat. Biotechnol.

    (2015)
  • B. Du et al.

    Glomerular barrier behaves as an atomically precise bandpass filter in a sub-nanometre regime

    Nat. Nanotechnol.

    (2017)
  • B. Pelaz et al.

    Surface Functionalization of Nanoparticles with Polyethylene Glycol: Effects on Protein Adsorption and Cellular Uptake

    ACS Nano

    (2015)
  • S. Behzadi et al.

    Cellular uptake of nanoparticles: journey inside the cell

    Chem. Soc. Rev.

    (2017)
  • J. Xu et al.

    Dose dependencies and biocompatibility of renal clearable gold nanoparticles: from mice to non-human primates

    Angew. Chem. Int. Ed.

    (2018)
  • K.M. Tsoi et al.

    Mechanism of hard-nanomaterial clearance by the liver

    Nat. Mater.

    (2016)
  • V.P. Chauhan et al.

    Strategies for advancing cancer nanomedicine

    Nat. Mater.

    (2013)
  • D.B. Kirpotin et al.

    Antibody targeting of long-circulating lipidic nanoparticles does not increase tumor localization but does increase internalization in animal models

    Cancer Res.

    (2006)
  • M.E. Davis et al.

    Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles

    Nature

    (2010)
  • C.V. Pecot et al.

    RNA interference in the clinic: challenges and future directions

    Nat. Rev. Cancer

    (2011)
  • K. Lee et al.

    Nanoparticle delivery of Cas9 ribonucleoprotein and donor DNA in vivo induces homology-directed DNA repair

    Nat. Biomed. Eng.

    (2017)
  • M. Kodiha et al.

    Off to the organelles - killing cancer cells with targeted gold nanoparticles

    Theranostics

    (2015)
  • M.R.K. Ali et al.

    Nuclear membrane-targeted gold nanoparticles inhibit cancer cell migration and invasion

    ACS Nano

    (2017)
  • A. Panariti et al.

    The effect of nanoparticle uptake on cellular behavior: disrupting or enabling functions?

    Nanotechnol. Sci. Appl.

    (2012)
  • Cited by (570)

    View all citing articles on Scopus
    View full text