• No results found

University of Groningen Exploiting the Biomolecular Corona of Nanocarriers for Targeted drug Delivery to Endothelial Cells Aliyandi, Aldy

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Exploiting the Biomolecular Corona of Nanocarriers for Targeted drug Delivery to Endothelial Cells Aliyandi, Aldy"

Copied!
25
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Exploiting the Biomolecular Corona of Nanocarriers for Targeted drug Delivery to Endothelial Cells

Aliyandi, Aldy DOI:

10.33612/diss.133166778

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Aliyandi, A. (2020). Exploiting the Biomolecular Corona of Nanocarriers for Targeted drug Delivery to Endothelial Cells. University of Groningen. https://doi.org/10.33612/diss.133166778

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

1

Chapter 1

General Introduction

and Aim of the Thesis

(3)

1.1 Nanomedicine

1.1.1 Nanoparticles for drug delivery

Over the past few decades, nanoparticles have emerged as promising drug delivery systems for therapeutic and diagnostic purposes, with a particular focus on cancer therapy.1–9 Due to

their small sizes, nanoparticles can interact with other systems on the nano-scale, including proteins, cell surface receptors, and nucleic acids, making them suitable for biomedical applications. One important benefit of nanoparticles as drug carriers is that they can be used to increase the solubility of hydrophobic compounds or to carry high loads of hydrophilic drugs, while also providing protection against enzymatic degradation. By doing so, not only drug solubility and drug delivery but also their release profile can be designed in a more controlled manner. Moreover, nanoparticles can be engineered in many different ways by tuning their properties, such as surface (e.g. charge, hydrophobicity, and surface functionalization) and physical properties (e.g. size, shape, stiffness, and porosity), or their composition (e.g. lipid, polymeric, and inorganic) to meet various needs as illustrated in Figure.1. By varying nanoparticle design, various aspects of their outcomes in biological environments, their in vivo biodistribution and interactions with cells can be modulated, such as serum protein interactions, immune cell activation, pharmacokinetic profile, targeting ability, and also potential side effects.

1.1.2 The biomolecular corona of nanoparticles

The term “nanoparticle corona” was first introduced by Cedervall et al., and refers to the layer of biomolecules, such as proteins, lipids, sugar, and other biomolecules, that forms on the surface of nanoparticles once they are exposed to a biological environment (Figure 2).10 Since

proteins contribute to the majority of these biomolecules, the corona mainly comprises a protein layer, which is why it is generally called “protein corona”. These proteins can either be in a direct contact with the nanoparticle surface or indirectly bind via binding to other proteins already adsorbed.

(4)

1

Figure 1. Tunable physical and chemical properties of nanoparticles. The fate and therapeutic

outcome of nanoparticles are influenced by their structural features and chemical composition, including surface properties (e.g. charge and hydrophobicity), general physical properties (e.g, size, shape, and porosity), the types of material (e.g. lipid, inorganic, and polymeric), and targeting moieties (e.g. protein, antibodies, and nucleic acids). Adapted from Salvioni et al.7

The protein corona is a dynamic structure and its composition changes over time as nanoparticles travel to other biological environments. In general, the nanoparticle surface is first occupied by the most abundant proteins, and then quickly replaced by the proteins with higher affinity.11 Thus, it is also important to mention that although blood plasma consists of

thousands of different proteins, their abundance in the plasma does not necessarily reflect their abundance in the protein corona.12,13 Also, the type and amount of proteins adsorbed

vary depending on the properties of nanoparticles i.e. size, shape, surface charge, and rigidity, and of the environment and exposure conditions, such as for instance the serum type and its concentration, the temperature, presence of flow and shear stress, among many others factors.2,12,14–17 Furthermore, based on the binding affinity and exchange rate of the proteins,

the protein corona can be divided between a hard corona and a soft corona. The hard corona is composed of strongly bound proteins with a long residence time on the nanoparticle surface, while the soft corona comprises more loosely bound proteins characterized by fast exchange with the surrounding environment.

(5)

Figure 2. Formation of the protein corona on the nanoparticle surface and its impact on

nanoparticle-cell interactions. (A) When nanoparticles are exposed to biological environment, proteins and other biomolecules adsorb on the nanoparticle surface (A). Instead of bare nanoparticles, it is the nanoparticle-corona complex that interacts with cellular machinery. (B) The protein corona consists of strongly adsorbed proteins (k1) called the hard corona and loosely bound proteins (k2) called the soft corona. Sufficiently long-lived hard corona may lead nanoparticle interactions (recognition) with cell receptors (k3). The same biomolecule can be recognized by cell receptors (k4). Similarly, if present, bare nanoparticles may also interact with cell receptors (k5). Adapted from Monopoli et al.18

For nanomedicine applications, the formation of the protein corona is one crucial aspect that needs to be considered. This protein layer can alter nanoparticle properties and stability and it can also confer a new biological identity to the nanoparticles, which affects the subsequent interactions with cells.18–20 In fact, it is now widely known that the protein corona can affect

the way nanoparticles are recognized and processed by cells, and modulate their efficacy as well as their overall pharmacological and toxicological profiles.21–23 Moreover, different

studies have demonstrated that the protein corona can be specifically recognized by cell receptors (A more comprehensive review on protein corona interactions with cell receptors is given later in Chapter 2).24–27 More recent examples showed that not only the presence or

absence of the protein corona, but also its composition can affect the uptake mechanisms of the same nanoparticles by cells.21,27 Overall, all these findings have demonstrated that the

(6)

1

protein corona can have important implications for the success of nanomedicines and nanoparticle outcomes at cell and organism level. However, a detailed picture of how the protein corona affects the biological behavior of nanoparticles is still missing. The work presented in this Thesis aims at gaining a better understanding on how the protein corona mediates the interactions of nanoparticles with biological systems and how it can be exploited for improving nanomedicine targeting.

1.1.3 Targeting strategies of nanomedicine

Unlike small molecular drugs, which in many cases diffuse and partition into the body depending on their solubility, nanoparticles distribute in the body and are processed and internalized by cells using energy dependent mechanisms.28,29 As a result, by changing their

properties as previously mentioned, nanoparticles can be designed to target specific cell types. In the next sections, I will discuss briefly the current strategies to improve the delivery of nanomedicines to their target.

1.1.3.1 Plasma residence time

Generally, in order to improve drug delivery, nanoparticles are designed to have sufficiently prolonged circulation. To this end, several nanoparticle properties that can critically affect nanoparticle circulation time, such as size, shape, and surface charge need to be optimized. The size of nanoparticles needs to be smaller than the cut-off of the fenestrations in the neovasculature, but not too small to be excreted through the kidneys. Nanoparticles in blood adsorb opsonin proteins that are recognized by immune cells and induce nanoparticle clearance. By changing nanoparticle charge, their protein corona and opsonization profile can be varied, thus affecting their clearance and circulation time30–34 Positive surface charges are

generally recognized to have a negative impact on the blood circulation of nanoparticles by, for instance, increasing nanoparticle clearance by immune cells.30,34 Alternatively, coating

nanoparticles with stealth materials, such as polyethylene glycol (PEG) is commonly performed to reduce protein adsorption on the nanoparticle surface, thus minimizing unwanted interactions with immune cells and prolonging their plasma residence time.35–37

Other strategies, such as introducing zwitterionic modification or coating nanoparticles with cell membrane derived from blood cells (camouflaging strategy), have also been shown to reduce nanoparticle clearance by immune cells.38–40

(7)

1.1.3.2 Passive targeting

In passive targeting, nanoparticles are designed to accumulate in their target tissue via the so called enhanced permeation and retention (EPR effect) (see Figure 2A for the illustration), which is commonly used as a strategy to treat cancer.41,42 The EPR effect relies on the presence

of leaky vasculature, whose endothelium shows gaps between cells of around 100-700 nm of size.43 For example, the Kaposi sarcoma is a tumor with fenestrated blood vessels.44 Thus,

passive accumulation of nanotherapeutics into tumor can occur without any targeting ligands attached on the surface of the nanoparticles. However, it has been widely acknowledged that passive targeting based on EPR effect is not sufficient to treat cancers with more challenging microenvironments. For instance, an increased interstitial fluid pressure, hypoxia, and the presence of a dense extracellular matrix in certain malignancies can result in a reduced transport of nanoparticles into the tumor.45–47 Furthermore, not every tumor shows EPR and

heterogeneity in EPR among patients can further limit the success of passively targeted medicines.48 More recently, the overall passive targeting paradigm has been challenged by a

study that has suggested that 97% of nanoparticle transport to tumor is mediated by active transport and only 3% is actually mediated by the EPR effect.49 In addition, passive targeting

also allows for off-target accumulation of nanocarriers in other organs with fenestrated endothelium, such as the liver and spleen.50 In order to overcome some of these problems,

passive targeting is often combined with active targeting strategies, including the use of targeting ligands. In addition, the use of stimuli-responsive drug carriers provides alternative ways to deliver drugs specifically to their target: nanoparticles can be designed to release the therapeutic compounds in response to endogenous stimuli, such as lower pH or oxygen level in the tumor microenvironment, or external stimuli, such as light, heat, ultrasound, or magnetic fields.46,51,52

(8)

1

Figure 3. Schematic illustration of drug targeting strategies. (A) Extravasation of

nanotherapeutics through leaky vasculature leads to passive targeting, also known as EPR effect, to target tumor tissue. (B) Active targeting of tumor tissue or tumor endothelial cells using nanocarriers functionalized with targeting ligands. Adapted from Wicki et al.8

1.1.3.3 Active targeting

In the case of active targeting, targeting ligands are attached to the surface of nanocarriers. These ligands specifically bind to their receptor on the cell surface, ensuring the delivery and accumulation of a therapeutic compound to their desired targets. Therefore, the approach is aimed towards increasing its therapeutic effects while reducing its toxicity in other (non-targeted) cells. A wide array of ligands has been used for this purpose, including protein, peptides, antibodies, oligonucleotides, and small molecules such as vitamins.53 The

interactions between ligand-functionalized nanoparticles with their target receptors or antigens are enhanced by the presence of multiple copies of the ligands on the nanoparticle surface, which increases the avidity of the nanoparticles for their target.54

Currently, active targeting is envisioned as a promising complementary strategy to passive targeting to further improve the efficacy of nanomedicines. However, in order to do so, there are several important aspects that need to be considered. For instance, actively-targeted nanoparticles are required to be in the vicinity of their targeted cells in order to interact with them. In the case of tumors, where the blood flow is often low compared to those in other organs and poorly vascularized areas can be present, the increase in nanoparticle’s affinity for the targeted tumor cells cannot always compensate the lower delivery into the tumor due to poor vascularization.31 Therefore, as for passive targeting, actively targeted nanocarriers also

need to be designed and optimized to have extended blood circulation time to achieve optimal delivery.

(9)

Another challenge for actively targeting nanoparticles is the formation of the protein corona. As mentioned in the previous section, adsorption of proteins and other biomolecules to the surface of nanoparticles can alter the nanoparticle properties, including masking the targeting ligands, resulting in the loss of targeting capability.55,56 Because of this, the protein corona has

been initially considered as a hindrance for the targeting of nanocarriers. Strategies such as with the addition of a stealth layer using PEG or zwitterionic charges, as mentioned previously, have been widely used to reduce protein adsorption on the nanoparticle surface and limit similar effects on targeting.35,36,39 At the same time, a paradigm shift has also occurred, where

– in contrast - the protein corona is now extensively exploited as a new targeting strategy.57– 60 In fact, several studies have demonstrated that the protein corona can be recognized by cell

receptors, and others have reported that adsorption of certain corona proteins could enhance uptake to specific cells or transcytosis through the blood-brain barrier.24,25,27,61 As discussed

previously, formation of the protein corona is affected by the physicochemical properties of nanoparticles. Therefore, as a new means of achieving active targeting, nanoparticles can be tailored so that they may adsorb certain plasma proteins in their corona to ‘naturally’ target specific cell types.61,62 This kind of applications of corona are discussed in further detail in the

review of Chapter 2.

1.1.4 Current success and challenges of nanomedicine

Research on nanocarriers for drug delivery has been very active in the last decades as shown by the increasing number of publications in this field (Figure 5A).63 In fact, several

nanocarrier-based medicines have been successfully translated into the clinic, such as Doxil® (pegylated liposomal doxorubicin) and Abraxan® (nab-paclitaxel) for cancer therapy, and Feraheme® (super paramagnetic iron oxide nanoparticles) for iron replacement therapy. Passively targeted nanomedicines represent the majority of the clinically available nanomedicines. Doxil®, Abraxan®, for example, are based on passive targeting. These are the first-generation nanomedicine drugs, which allowed to improve the pharmacokinetics and biodistribution of poorly soluble drug by encapsulating them into a passively targeted drug carrier. In the case of Doxil, for instance, the blood circulation of conventional doxorubicin is improved by at least 250-fold thanks to the encapsulation in a liposome and addition of a pegylated surface, to make the liposome stealth, as explained in the previous sections.64

(10)

1

Figure 4. Nanocarriers in drug delivery. (A) Number of publications related to nanocarriers for

drug delivery from 1995-2019. (B) Analysis of median nanoparticle delivery to tumor in terms of %ID in various solid tumor models, indicating major delivery concerns in the delivery of nanocarrier-based medicines Adapted from Zhao et al (for Fig. 4A) and Wilhelm et al (for Fig. 4B).63,65

Despite the success of the first nanomedicines and high number of clinical trials of nanocarrier-based medicines, nanomedicine potential can be further improved.49,65–68 So far,

only relatively few nanomedicines made it to the clinic, and none of them is yet based on active targeting. In many cases, nanocarriers based on either passive or active targeting do not seem to offer significant therapeutic improvement compared to the free drug counterpart, and this still remains a great challenge in nanomedicine. A comprehensive study by Chan and co-workers, where they analyzed the accumulation of different types of nanoparticles that have been tested on different solid tumor models between 2005 and 2015, claimed that only 0.7% of the injected dose arrived at the tumor (Figure 5B).65 Moreover, Chan

and co-workers also recently performed a study to evaluate the concept of nanoparticle transport through the gap between endothelial cells. 49 In this study, as already mentioned

(11)

of human tumors, and imaging techniques, and mathematical simulation, they found that these gaps are not responsible for nanoparticle transport. Instead, 97% of nanoparticle accumulation in tumor is based on active transport via transcytosis through endothelial cells. Many other studies also have pointed out that the low targeting efficiency of nanocarriers is due to the presence of many biological barriers and the still limited understanding of how nanoparticles interact with biological systems.69–72 In this regard, several aspects such as

formation of the protein corona, ability to cross anatomical barriers, nanoparticle-cell interactions, and intracellular fate of nanoparticles have to be extensively investigated before more new nanomedicines may successfully step up to clinical trials.

1.2 Endothelial cell barriers in nanomedicine

With the current challenges faced by nanocarrier-based drug delivery systems, it is now acknowledged that a better understanding of how nanoparticles behave at cellular and molecular level is pivotal in order to improve further nanomedicine design and to achieve better targeting efficiency.71,73,74 As mentioned previously, the successful delivery of

nanomedicine is majorly hampered by the presence of several biological barriers. Therefore, understanding how nanoparticles interact with these barriers is one of the first key steps towards the successful design of nanomedicines.

One of the most critical barriers that nanomedicines need to overcome in order to reach their targeted organs or tissue is the endothelium lining the blood vessels.75,76 The recent study by

Chan et al, as discussed in the previous section, where it was shown that active transport through endothelial cells contributed to 97% of overall nanoparticle transport to tumor, further highlighted that endothelial cells represent a critical barrier for nanomedicine, and also for tumors. Endothelial cells control many important functions in the body, including maintenance of the vascular tone and blood pressure, as well as transport of blood components and nutrients to the underlying tissues. Furthermore, they also function as a barrier that separates blood and other exogenous materials, including drugs, from the extravascular tissues, and thereby controls the access of blood constituents to organs. In order to function as a barrier, endothelial cells are typically organized into cellular barriers expressing tight junction proteins, with clear spatial differences in shape, structure, and function between their apical and basolateral membrane.77 As a result, endothelial cells

(12)

1

express different endocytic pathways in the two membranes.78 In order to reach the

underlying tissue, nanoparticles can use paracellular transport or — more often — transcellular transport (transcytosis), where they have to be internalized from the apical membrane, and transported out through the basolateral membrane.79–85 Moreover, it is

known that the endothelial cell phenotype differs between various types of vasculature (veins, arteries, and capillaries), in different organs and also depending on physiological conditions (Figure 5).86–89 This large heterogeneity of endothelial barriers, each exhibiting unique

structural and functional properties, may also provide novel strategies that can be exploited for targeting of drug carriers to specific tissues or organs.90,91 Thus, in vitro models for different

endothelial cell barriers originating from different organs were prepared and used throughout the work as presented in this Thesis.

Figure 5. Heterogeneity of endothelial cells in various tissues. (A) The expression patterns of

tissue specific genes among all investigated tissue microvascular endothelial cells, and expanded on the right (B) with selected gene names. This heterogeneity can be exploited as a targeting strategy of drug carriers. Adapted from Chi et al.87

(13)

1.3 Aim of the thesis

Nano-sized materials offer great potential as drug carriers given their ability to exploit the cellular machinery in a fundamental new way compared to conventional drugs.28,92–95 As

already mentioned above, in order to achieve successful design and delivery of nanomedicines, a better understanding of how nanoparticles interact with biological systems is urgently needed.71,73,74 Among many important aspects in nano-bio interactions, the

formation of the protein corona and the ability of nanoparticles to cross endothelial cell barriers still remain among the major challenges that require deeper investigation.96–98

Within this context, the aim of the thesis is to gain better understanding of nanoparticle interactions with endothelial cells, and more specifically, to exploit the protein corona as a tool to discover novel targeting strategies to direct nanoparticles to specific endothelial cell types (see Figure 6 for the workflow overview).

Figure 6. Workflow diagram illustrating how the protein corona is exploited for targeting drug

carriers to specific endothelial cells. First, the effect of cell barrier formation on nanoparticle uptake is investigated (1). Similarly, the effect of endothelial cell heterogeneity between different organs on the uptake of different corona-coated silica nanoparticles is studied (2). The corona composition of each nanoparticle type is analyzed by mass spectrometry,

(14)

1

the uptake, and further evaluated (3). Finally, receptors interacting with the protein corona, including those involved in the uptake, are isolated using two different biotinylation-based approaches, analyzed and identified by mass spectrometry, and further evaluated (4).

In Chapter 2, I present a detailed review on our current understanding of protein corona interactions with cell receptors, illustrating the importance of this first step of nanoparticle-cell interactions and its implication for targeting of nanomedicines. Different methods currently applied to identify corona proteins correlating with higher or lower uptake by cells are discussed, together with methods to identify the cell receptors interacting with corona proteins and involved in nanoparticle uptake. This includes a description of the methods optimized in this Thesis to answer these questions.

In order to study nanoparticle-cell interactions with endothelial cell barriers in vitro, it is of great importance that the cell models used resemble as closely as possible physiological barriers. So far, a large majority of studies on nanoparticle-cell behavior has been performed using simple cell models, such as HeLa cells and other cells grown to various degrees of confluency.27,99,100 In the case of endothelial cells, a special attention is needed when using

them as a cell model. As mentioned previously, in vivo, endothelial cells are organized as a monolayer expressing tight junction proteins. Therefore, organized endothelial cell layers with proper expression of tight junction proteins may constitute an in vitro model that better reflects the barriers nanomedicine encounter in vivo compared to sub-confluent or even confluent cells. More importantly, organized cell barriers may take up and process nanoparticles differently in comparison to cells grown to different degrees of confluency as generally used in many in vitro studies. Therefore, in Chapter 3, in order to address this question, I have used primary human umbilical vein endothelial cells (HUVEC) and optimized procedures to differentiate them into an endothelial cell barrier. Then, I have investigated how the differentiation of endothelial cells into a cell barrier in vitro affects nanoparticle uptake and behavior in these cells.

As a next step, I have studied how endothelial cell heterogeneity affects nanoparticle uptake. As discussed in the previous section, endothelial cell phenotype differs in various organs and physiological conditions. Because of this, endothelial cells of different organs are likely to show different nanoparticle uptake behavior. We hypothesized that different endothelia

(15)

would show preferential uptake towards certain types of nanoparticles, and such differences could be exploited for targeting of nanocarriers. Based on this hypothesis, in Chapter 4 I have used recently established immortalized cell lines to prepare endothelial cell barriers derived from four different organs with high phenotypic differences between one another, namely brain (hCMEC/D3), lungs (HPMEC-ST1.6R), liver (TRP3), and kidney (ciGENC). Thus, the different endothelial barriers have been exposed to silica nanoparticles with three different surface functionalization and differences in uptake levels and uptake preferences have been investigated.

Having observed that endothelial cell heterogeneity affects nanoparticle uptake, in Chapter

5, I have used a panel of different nanoparticles to form different coronas and identify proteins

that are responsible in regulating nanoparticle uptake in the four endothelial cell barriers from different organs. As discussed in the previous section, several studies have shown that certain corona components can naturally target nanocarriers to specific cells, and that the protein corona can be recognized by cell receptors and mediate nanoparticle uptake.24,25,57,61,62 Since

the protein corona composition varies with nanoparticle physicochemical properties, tuning these properties allows to tune corona composition and, in this way, direct nanoparticles to specific cells. To this aim, I have studied the protein corona composition of a panel of nanoparticles of different sizes and surface functionalization, and performed a correlation analysis between the protein corona and their uptake profile in the four endothelial cell barriers. Thus, corona proteins affecting uptake have been identified and their role has been validated.

As a final step, in Chapter 6, using brain and liver endothelia as cell barrier models, I have characterized cell receptors involved in corona recognition and nanoparticle uptake. Given the ability of the protein corona to be recognized by cell receptors, it is of great interest to exploit further the protein corona to discover receptors that may allow targeted uptake in specific cells. However, given that many proteins constitute the protein corona, and may interact with multiple receptors, it is essential to develop methods to be able to identify which receptors are involved in this interaction. Currently, identifying the corona proteins recognized by cell receptors and the receptors involved in nanoparticle uptake remains difficult. The challenge mainly lies in the fact that only certain corona proteins, whose epitopes are accessible and not

(16)

1

covered by other proteins, may bind to cell receptors.26 So far, there are no available

systematic techniques to easily identify such corona proteins, and this makes it difficult to find interacting receptors. Therefore, in Chapter 6, I developed novel methods, based on biotinylation, mass spectrometry, and corona proteomic data, to address this challenge and identify all possible receptors that interact with the protein corona, as well as those that mediate nanoparticle uptake. These methods can be used also to exploit the corona as a tool to discover novel receptors for targeting.

Overall, the work presented elucidates important aspects on the effect of barrier formation and endothelial cell heterogeneity on nanoparticle uptake. Thus, different methods are presented and combined to identify cell receptors interacting with corona proteins and involved in nanoparticle uptake in different endothelial cell barriers. Endothelial cell heterogeneity and the protein corona can be exploited to develop novel strategies for nanomedicine targeting.

(17)

References

1. Wolfram J, Ferrari M. Clinical cancer nanomedicine. Nano Today. 2019;25:85-98. doi:10.1016/j.nantod.2019.02.005

2. Shi J, Kantoff PW, Wooster R, Farokhzad OC. Cancer nanomedicine: Progress, challenges and opportunities. Nat Rev Cancer. 2017;17(1):20-37. doi:10.1038/nrc.2016.108 3. D’Mello SR, Cruz CN, Chen ML, Kapoor M, Lee SL, Tyner KM. The evolving landscape of

drug products containing nanomaterials in the United States. Nat Nanotechnol. 2017;12(6):523-529. doi:10.1038/nnano.2017.67

4. Bertrand N, Wu J, Xu X, Kamaly N, Farokhzad OC. Cancer nanotechnology: The impact of passive and active targeting in the era of modern cancer biology. Adv Drug Deliv Rev. 2014;66:2-25. doi:10.1016/j.addr.2013.11.009

5. Prasad M, Lambe UP, Brar B, et al. Nanotherapeutics: An insight into healthcare and multi-dimensional applications in medical sector of the modern world. Biomed

Pharmacother. 2018;97:1521-1537. doi:10.1016/j.biopha.2017.11.026

6. Tran S, DeGiovanni P-J, Piel B, Rai P. Cancer nanomedicine: a review of recent success in drug delivery. Clin Transl Med. 2017;6(1):44. doi:10.1186/s40169-017-0175-0

7. Salvioni L, Rizzuto MA, Bertolini JA, Pandolfi L, Colombo M, Prosperi D. Thirty years of cancer nanomedicine: Success, frustration, and hope. Cancers (Basel). 2019;11(12):1855. doi:10.3390/cancers11121855

8. Wicki A, Witzigmann D, Balasubramanian V, Huwyler J. Nanomedicine in cancer therapy: Challenges, opportunities, and clinical applications. J Control Release. 2015;200:138-157. doi:10.1016/j.jconrel.2014.12.030

9. Audouy SAL, De Leij LFMH, Hoekstra D, Molema G. In vivo characteristics of cationic liposomes as delivery vectors for gene therapy. Pharm Res. 2002;19(11):1599-1605. doi:10.1023/A:1020989709019

10. Cedervall T, Lynch I, Lindman S, et al. Understanding the nanoparticle-protein corona using methods to quantify exchange rates and affinities of proteins for nanoparticles.

Proc Natl Acad Sci. 2007;104(7):2050-2055. doi:10.1073/pnas.0608582104

11. Vroman L, Adams AL. Findings with the recording ellipsometer suggesting rapid exchange of specific plasma proteins at liquid/solid interfaces. Surf Sci. 1969;16(C):438-446. doi:10.1016/0039-6028(69)90037-5

12. Tenzer S, Docter D, Rosfa S, et al. Nanoparticle size is a critical physicochemical determinant of the human blood plasma corona: A comprehensive quantitative proteomic analysis. ACS Nano. 2011;5(9):7155-7167. doi:10.1021/nn201950e

13. Walkey CD, Chan WCW. Understanding and controlling the interaction of nanomaterials with proteins in a physiological environment. Chem Soc Rev. 2012;41(7):2780-2799. doi:10.1039/C1CS15233E

(18)

1

surface properties determine the protein corona with possible implications for biological impacts. Proc Natl Acad Sci U S A. 2008;105(38):14265-14270. doi:10.1073/pnas.0805135105

15. Petros RA, Desimone JM. Strategies in the design of nanoparticles for therapeutic applications. Nat Rev Drug Discov. 2010;9(8):615-627. doi:10.1038/nrd2591

16. Nienhaus K, Wang H, Nienhaus GU. Nanoparticles for biomedical applications: exploring and exploiting molecular interactions at the nano-bio interface. Mater Today Adv. 2020;5:100036. doi:10.1016/j.mtadv.2019.100036

17. Vilanova O, Mittag JJ, Kelly PM, et al. Understanding the Kinetics of Protein-Nanoparticle Corona Formation. ACS Nano. 2016;10(12):10842-10850. doi:10.1021/acsnano.6b04858

18. Monopoli MP, Åberg C, Salvati A, Dawson KA. Biomolecular coronas provide the biological identity of nanosized materials. Nat Nanotechnol. 2012;7(12):779-786. doi:10.1038/nnano.2012.207

19. Ke PC, Lin S, Parak WJ, Davis TP, Caruso F. A Decade of the Protein Corona. ACS Nano. 2017:acsnano.7b08008. doi:10.1021/acsnano.7b08008

20. Pino P Del, Pelaz B, Zhang Q, Maffre P, Nienhaus GU, Parak WJ. Protein corona formation around nanoparticles - From the past to the future. Mater Horizons. 2014;1(3):301-313. doi:10.1039/c3mh00106g

21. Lesniak A, Fenaroli F, Monopoli MP, Åberg C, Dawson KA, Salvati A. Effects of the Presence or Absence of a Protein Corona on Silica Nanoparticle Uptake and Impact on Cells. ACS Nano. 2012;6(7):5845-5857. doi:10.1021/nn300223w

22. Zuhorn IS, Visser WH, Bakowsky U, Engberts JBFN, Hoekstra D. Interference of serum with lipoplex-cell interaction: Modulation of intracellular processing. Biochim Biophys

Acta - Biomembr. 2002;1560(1-2):25-36. doi:10.1016/S0005-2736(01)00448-5

23. Zyuzin M V., Yan Y, Hartmann R, et al. Role of the Protein Corona Derived from Human Plasma in Cellular Interactions between Nanoporous Human Serum Albumin Particles and Endothelial Cells. Bioconjug Chem. 2017;28(8):2062-2068. doi:10.1021/acs.bioconjchem.7b00231

24. Lara S, Alnasser F, Polo E, et al. Identification of Receptor Binding to the Biomolecular Corona of Nanoparticles. ACS Nano. 2017;11(2):1884-1893. doi:10.1021/acsnano.6b07933

25. Lara S, Perez-Potti A, Herda LM, Adumeau L, Dawson KA, Yan Y. Differential Recognition of Nanoparticle Protein Corona and Modified Low-Density Lipoprotein by Macrophage Receptor with Collagenous Structure. ACS Nano. 2018;12(5):4930-4937. doi:10.1021/acsnano.8b02014

26. Kelly PM, Åberg C, Polo E, et al. Mapping protein binding sites on the biomolecular corona of nanoparticles. Nat Nanotechnol. 2015;10(5):472-479. doi:10.1038/nnano.2015.47

(19)

27. Francia V, Yang K, Deville S, Reker-Smit C, Nelissen I, Salvati A. Corona Composition Can Affect the Mechanisms Cells Use to Internalize Nanoparticles. ACS Nano. 2019;13(10):11107-11121. doi:10.1021/acsnano.9b03824

28. Salvati A, Åberg C, dos Santos T, et al. Experimental and theoretical comparison of intracellular import of polymeric nanoparticles and small molecules: Toward models of uptake kinetics. Nanomedicine Nanotechnology, Biol Med. 2011;7(6):818-826. doi:10.1016/j.nano.2011.03.005

29. Zuhorn IS, Kalicharan R, Hoekstra D. Lipoplex-mediated transfection of mammalian cells occurs through the cholesterol-dependent clathrin-mediated pathway of endocytosis.

J Biol Chem. 2002;277(20):18021-18028. doi:10.1074/jbc.M111257200

30. He C, Hu Y, Yin L, Tang C, Yin C. Effects of particle size and surface charge on cellular uptake and biodistribution of polymeric nanoparticles. Biomaterials. 2010;31(13):3657-3666. doi:10.1016/j.biomaterials.2010.01.065

31. Bertrand N, Leroux JC. The journey of a drug-carrier in the body: An anatomo-physiological perspective. J Control Release. 2012;161(2):152-163. doi:10.1016/j.jconrel.2011.09.098

32. Salvador-Morales C, Zhang L, Langer R, Farokhzad OC. Immunocompatibility properties of lipid-polymer hybrid nanoparticles with heterogeneous surface functional groups.

Biomaterials. 2009;30(12):2231-2240. doi:10.1016/j.biomaterials.2009.01.005

33. Chonn A, Cullis PR, Devine D V. The role of surface charge in the activation of the classical and alternative pathways of complement by liposomes. J Immunol. 1991;146(12):4234-4241. http://www.ncbi.nlm.nih.gov/pubmed/2040798. Accessed May 4, 2020.

34. Xiao K, Li Y, Luo J, et al. The effect of surface charge on in vivo biodistribution of PEG-oligocholic acid based micellar nanoparticles. Biomaterials. 2011;32(13):3435-3446. doi:10.1016/j.biomaterials.2011.01.021

35. Harris JM, Martin NE, Modi M. Pegylation. Clin Pharmacokinet. 2001;40(7):539-551. doi:10.2165/00003088-200140070-00005

36. Howard MD, Jay M, Dziubla TD, Lu X. PEGylation of nanocarrier drug delivery systems: State of the art. J Biomed Nanotechnol. 2008;4(2):133-148. doi:10.1166/jbn.2008.021 37. Schöttler S, Becker G, Winzen S, et al. Protein adsorption is required for stealth effect

of poly(ethylene glycol)- and poly(phosphoester)-coated nanocarriers. Nat

Nanotechnol. 2016;11(4):372-377. doi:10.1038/nnano.2015.330

38. Yuan Y-Y, Mao C-Q, Du X-J, Du J-Z, Wang F, Wang J. Surface Charge Switchable Nanoparticles Based on Zwitterionic Polymer for Enhanced Drug Delivery to Tumor. Adv

Mater. 2012;24(40):5476-5480. doi:10.1002/adma.201202296

39. García KP, Zarschler K, Barbaro L, et al. Zwitterionic-coated “stealth” nanoparticles for biomedical applications: Recent advances in countering biomolecular corona formation and uptake by the mononuclear phagocyte system. Small. 2014;10(13):2516-2529.

(20)

1

doi:10.1002/smll.201303540

40. Luk BT, Zhang L. Cell membrane-camouflaged nanoparticles for drug delivery. J Control

Release. 2015;220:600-607. doi:10.1016/j.jconrel.2015.07.019

41. Gerlowski LE, Jain RK. Microvascular permeability of normal and neoplastic tissues.

Microvasc Res. 1986;31(3):288-305. doi:10.1016/0026-2862(86)90018-X

42. Matsumura Y, Maeda H. A New Concept for Macromolecular Therapeutics in Cancer Chemotherapy: Mechanism of Tumoritropic Accumulation of Proteins and the Antitumor Agent Smancs. Cancer Res. 1986;46(8):6387-6392. http://www.ncbi.nlm.nih.gov/pubmed/2946403. Accessed April 21, 2020.

43. Hobbs SK, Monsky WL, Yuan F, et al. Regulation of transport pathways in tumor vessels: Role of tumor type and microenvironment. Proc Natl Acad Sci U S A. 1998;95(8):4607-4612. doi:10.1073/pnas.95.8.4607

44. Lammers T, Kiessling F, Hennink WE, Storm G. Drug targeting to tumors: Principles, pitfalls and (pre-) clinical progress. J Control Release. 2012;161(2):175-187. doi:10.1016/j.jconrel.2011.09.063

45. Prabhakar U, Maeda H, K. Jain R, et al. Challenges and key considerations of the enhanced permeability and retention effect for nanomedicine drug delivery in oncology. In: Cancer Research. Vol 73. American Association for Cancer Research; 2013:2412-2417. doi:10.1158/0008-5472.CAN-12-4561

46. Jain RK, Stylianopoulos T. Delivering nanomedicine to solid tumors. Nat Rev Clin Oncol. 2010;7(11):653-664. doi:10.1038/nrclinonc.2010.139

47. Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic Targeting of the Stroma Ablates Physical Barriers to Treatment of Pancreatic Ductal Adenocarcinoma. Cancer Cell. 2012;21(3):418-429. doi:10.1016/j.ccr.2012.01.007 48. Maeda H. Toward a full understanding of the EPR effect in primary and metastatic

tumors as well as issues related to its heterogeneity. Adv Drug Deliv Rev. 2015;91:3-6. doi:10.1016/j.addr.2015.01.002

49. Sindhwani S, Syed AM, Ngai J, et al. The entry of nanoparticles into solid tumours. Nat

Mater. 2020;19(5):566-575. doi:10.1038/s41563-019-0566-2

50. Gaumet M, Vargas A, Gurny R, Delie F. Nanoparticles for drug delivery: The need for precision in reporting particle size parameters. Eur J Pharm Biopharm. 2008;69(1):1-9. doi:10.1016/j.ejpb.2007.08.001

51. Li HJ, Du JZ, Du XJ, et al. Stimuli-responsive clustered nanoparticles for improved tumor penetration and therapeutic efficacy. Proc Natl Acad Sci U S A. 2016;113(15):4164-4169. doi:10.1073/pnas.1522080113

52. Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol. 2007;2(12):751-760. doi:10.1038/nnano.2007.387

(21)

53. Saha RN, Vasanthakumar S, Bende G, Snehalatha M. Nanoparticulate drug delivery systems for cancer chemotherapy. Mol Membr Biol. 2010;27(7):215-231. doi:10.3109/09687688.2010.510804

54. Wang J, Tian S, Petros RA, Napier ME, Desimone JM. The complex role of multivalency in nanoparticles targeting the transferrin receptor for cancer therapies. J Am Chem Soc. 2010;132(32):11306-11313. doi:10.1021/ja1043177

55. Salvati A, Pitek AS, Monopoli MP, et al. Transferrin-functionalized nanoparticles lose their targeting capabilities when a biomolecule corona adsorbs on the surface. Nat

Nanotechnol. 2013;8(2):137-143. doi:10.1038/nnano.2012.237

56. Hadjidemetriou M, Al-Ahmady Z, Mazza M, Collins RF, Dawson K, Kostarelos K. In Vivo Biomolecule Corona around Blood-Circulating, Clinically Used and Antibody-Targeted Lipid Bilayer Nanoscale Vesicles. ACS Nano. 2015;9(8):8142-8156.

doi:10.1021/acsnano.5b03300

57. Ritz S, Schöttler S, Kotman N, et al. Protein Corona of Nanoparticles: Distinct Proteins Regulate the Cellular Uptake. Biomacromolecules. 2015;16(4):1311-1321. doi:10.1021/acs.biomac.5b00108

58. Walkey CD, Olsen JB, Song F, et al. Protein corona fingerprinting predicts the cellular interaction of gold and silver nanoparticles. ACS Nano. 2014;8(3):2439-2455. doi:10.1021/nn406018q

59. Nguyen VH, Lee B-J. Protein corona: a new approach for nanomedicine design. Int J

Nanomedicine. 2017;Volume 12:3137-3151. doi:10.2147/IJN.S129300

60. Liu R, Jiang W, Walkey CD, Chan WCW, Cohen Y. Prediction of nanoparticles-cell association based on corona proteins and physicochemical properties. Nanoscale. 2015;7(21):9664-9675. doi:10.1039/C5NR01537E

61. Kreuter J, Shamenkov D, Petrov V, et al. Apolipoprotein-mediated transport of nanoparticle-bound drugs across the blood-brain barrier. J Drug Target. 2002;10(4):317-325. doi:10.1080/10611860290031877

62. Caracciolo G, Cardarelli F, Pozzi D, et al. Selective Targeting Capability Acquired with a Protein Corona Adsorbed on the Surface of 1,2-Dioleoyl-3-trimethylammonium Propane/DNA Nanoparticles. ACS Appl Mater Interfaces. 2013;5(24):13171-13179. doi:10.1021/am404171h

63. Zhao Z, Ukidve A, Krishnan V, Mitragotri S. Effect of physicochemical and surface properties on in vivo fate of drug nanocarriers. Adv Drug Deliv Rev. 2019;143:3-21. doi:10.1016/j.addr.2019.01.002

64. Gabizon A, Shmeeda H, Barenholz Y. Pharmacokinetics of pegylated liposomal doxorubicin: Review of animal and human studies. Clin Pharmacokinet. 2003;42(5):419-436. doi:10.2165/00003088-200342050-00002

65. Wilhelm S, Tavares AJ, Dai Q, et al. Analysis of nanoparticle delivery to tumours. Nat

(22)

1

66. Torrice M. Does nanomedicine have a delivery problem? ACS Cent Sci. 2016;2(7):434-437. doi:10.1021/acscentsci.6b00190

67. Venditto VJ, Szoka FC. Cancer nanomedicines: So many papers and so few drugs! Adv

Drug Deliv Rev. 2013;65(1):80-88. doi:10.1016/j.addr.2012.09.038

68. Lammers T, Kiessling F, Ashford M, Hennink W, Crommelin D, Strom G. Cancer nanomedicine: Is targeting our target? Nat Rev Mater. 2016;1(9):16069. doi:10.1038/natrevmats.2016.69

69. Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol. 2015;33(9):941-951. doi:10.1038/nbt.3330 70. Wilczewska AZ, Niemirowicz K, Markiewicz KH, Car H. Nanoparticles as drug delivery

systems. Pharmacol Reports. 2012;64(5):1020-1037.

doi:10.1016/S1734-1140(12)70901-5

71. Time to deliver. Nat Biotechnol. 2014;32(10):961-961. doi:10.1038/nbt.3045

72. Cheon J, Chan W, Zuhorn I. The Future of Nanotechnology: Cross-disciplined Progress to Improve Health and Medicine. Acc Chem Res. 2019;52(9):2405. doi:10.1021/acs.accounts.9b00423

73. Iversen TG, Skotland T, Sandvig K. Endocytosis and intracellular transport of nanoparticles: Present knowledge and need for future studies. Nano Today. 2011;6(2):176-185. doi:10.1016/j.nantod.2011.02.003

74. Yameen B, Choi W Il, Vilos C, Swami A, Shi J, Farokhzad OC. Insight into nanoparticle cellular uptake and intracellular targeting. J Control Release. 2014;190:485-499. doi:10.1016/j.jconrel.2014.06.038

75. Zhang Y, Yang WX. Tight junction between endothelial cells: The interaction between nanoparticles and blood vessels. Beilstein J Nanotechnol. 2016;7(1):675-684. doi:10.3762/bjnano.7.60

76. Setyawati MI, Tay CY, Docter D, Stauber RH, Leong DT. Understanding and exploiting nanoparticles’ intimacy with the blood vessel and blood. Chem Soc Rev. 2015;44(22):8174-8199. doi:10.1039/c5cs00499c

77. Dejana E. Endothelial cell-cell junctions: Happy together. Nat Rev Mol Cell Biol. 2004;5(4):261-270. doi:10.1038/nrm1357

78. Sandvig K, van Deurs B. Delivery into cells: Lessons learned from plant and bacterial toxins. Gene Ther. 2005;12(11):865-872. doi:10.1038/sj.gt.3302525

79. Lin IC, Liang M, Liu TY, Monteiro MJ, Toth I. Cellular transport pathways of polymer coated gold nanoparticles. Nanomedicine Nanotechnology, Biol Med. 2012;8(1):8-11. doi:10.1016/j.nano.2011.09.014

80. Li CH, Shyu MK, Jhan C, et al. Gold nanoparticles increase endothelial paracellular permeability by altering components of endothelial tight junctions, and increase blood-brain barrier permeability in mice. Toxicol Sci. 2015;148(1):192-203.

(23)

doi:10.1093/toxsci/kfv176

81. Ghaffarian R, Bhowmick T, Muro S. Transport of nanocarriers across gastrointestinal epithelial cells by a new transcellular route induced by targeting ICAM-1. In: Journal of

Controlled Release. Vol 163. Elsevier; 2012:25-33. doi:10.1016/j.jconrel.2012.06.007

82. Ulbrich K, Knobloch T, Kreuter J. Targeting the insulin receptor: Nanoparticles for drug delivery across the blood-brain barrier (BBB). J Drug Target. 2011;19(2):125-132. doi:10.3109/10611861003734001

83. Georgieva J V., Brinkhuis RP, Stojanov K, et al. Peptide-mediated blood-brain barrier transport of polymersomes. Angew Chemie - Int Ed. 2012;51(33):8339-8342. doi:10.1002/anie.201202001

84. Georgieva J V., Kalicharan D, Couraud P-O, et al. Surface Characteristics of Nanoparticles Determine Their Intracellular Fate in and Processing by Human Blood–Brain Barrier Endothelial Cells In Vitro. Mol Ther. 2011;19(2):318-325. doi:10.1038/mt.2010.236 85. Georgieva J V., Hoekstra D, Zuhorn IS. Smuggling drugs into the brain: An overview of

ligands targeting transcytosis for drug delivery across the blood–brain barrier.

Pharmaceutics. 2014;6(4):557-583. doi:10.3390/pharmaceutics6040557

86. Langenkamp E, Molema G. Microvascular endothelial cell heterogeneity: General concepts and pharmacological consequences for anti-angiogenic therapy of cancer. Cell

Tissue Res. 2009;335(1):205-222. doi:10.1007/s00441-008-0642-4

87. Chi J-T, Chang HY, Haraldsen G, et al. Endothelial cell diversity revealed by global expression profiling. Proc Natl Acad Sci. 2003;100(19):10623-10628. doi:10.1073/pnas.1434429100

88. Aird WC. Endothelial cell heterogeneity. Cold Spring Harb Perspect Med. 2012;2(1):a006429-a006429. doi:10.1101/cshperspect.a006429

89. Ribatti D, Nico B, Vacca A, Roncali L, Dammacco F. Endothelial cell heterogeneity and organ specificity. J Hematotherapy Stem Cell Res. 2002;11(1):81-90. doi:10.1089/152581602753448559

90. Kowalski PS, Lintermans LL, Morselt HWM, et al. Anti-VCAM-1 and anti-E-selectin SAINT-O-somes for selective delivery of siRNA into inflammation-activated primary endothelial cells. Mol Pharm. 2013;10(8):3033-3044. doi:10.1021/mp4001124

91. Simone E, Ding B Sen, Muzykantov V. Targeted delivery of therapeutics to endothelium.

Cell Tissue Res. 2009;335(1):283-300. doi:10.1007/s00441-008-0676-7

92. Bareford L, Swaan P. Endocytic mechanisms for targeted drug delivery☆. Adv Drug Deliv

Rev. 2007;59(8):748-758. doi:10.1016/j.addr.2007.06.008

93. Chithrani BD, Ghazani AA, Chan WCW. Determining the Size and Shape Dependence of Gold Nanoparticle Uptake into Mammalian Cells. Nano Lett. 2006;6(4):662-668. doi:10.1021/nl052396o

(24)

1

2010;145(3):182-195. doi:10.1016/j.jconrel.2010.01.036

95. Duncan R, Richardson SCW. Endocytosis and intracellular trafficking as gateways for nanomedicine delivery: Opportunities and challenges. Mol Pharm. 2012;9(9):2380-2402. doi:10.1021/mp300293n

96. Pederzoli F, Tosi G, Vandelli MA, Belletti D, Forni F, Ruozi B. Protein corona and nanoparticles: how can we investigate on? Wiley Interdiscip Rev Nanomedicine

Nanobiotechnology. 2017;9(6):e1467. doi:10.1002/wnan.1467

97. Barui AK, Oh JY, Jana B, Kim C, Ryu J. Cancer-Targeted Nanomedicine: Overcoming the Barrier of the Protein Corona. Adv Ther. September 2019:1900124. doi:10.1002/adtp.201900124

98. Ge C, Tian J, Zhao Y, Chen C, Zhou R, Chai Z. Towards understanding of nanoparticle– protein corona. Arch Toxicol. 2015;89(4):519-539. doi:10.1007/s00204-015-1458-0 99. dos Santos T, Varela J, Lynch I, Salvati A, Dawson KA. Effects of transport inhibitors on

the cellular uptake of carboxylated polystyrene nanoparticles in different cell lines. Schnur JM, ed. PLoS One. 2011;6(9):e24438. doi:10.1371/journal.pone.0024438

100. Yang K, Mesquita B, Horvatovich P, Salvati A. Tuning liposome composition to modulate corona formation in human serum and cellular uptake. Acta Biomater. 2020;106:314-327. doi:10.1016/j.actbio.2020.02.018

(25)

Referenties

GERELATEERDE DOCUMENTEN

The current research has attempted to answer the following research question: Does perceived brand fit affect attitude towards the brand and message and purchase intention

The basal lamina is composed of different molecules which include integrins; they serve not only as the connection between the ECM and the epithelial cells but also as initiator

The research presented in this thesis was performed in the department of Nanomedicine and Drug Targeting at the Groningen Research Institute of Pharmacy (GRIP), University of

In order to test the effect of the development of a cell barrier on uptake and transport mechanisms, we also compared the expression levels of genes coding for key proteins

Deze methoden zijn gebaseerd op een combinatie van corona proteomics, op biotinylering-gebaseerde methoden, onderzoeken naar de opname van nanodeeltjes in cellen,

Additionally for Ingrid, and together with Henk Moorlag, I would also like to thank you both for your help and fruitful discussion on endothelial cells.. Additionally, I would like

1. In order to reach their target, nano-sized carriers and drugs need to overcome a series of barriers at organ, tissue, and cellular levels. Among these, endothelial cells

The Ministry of Environment, Forests and Climate Change (MoEFCC) reckoned in 2009 itself that ‘The Scheduled Tribes and Other Traditional Forest Dwellers (Recognition of Forest