+ All Categories
Home > Documents > Targeted nanoparticles with novel non-peptidic ligands for oral delivery

Targeted nanoparticles with novel non-peptidic ligands for oral delivery

Date post: 04-Dec-2016
Category:
Upload: veronique
View: 213 times
Download: 0 times
Share this document with a friend
42
Targeted nanoparticles with novel non-peptidic ligands for oral delivery Anne des Rieux, Vincent Pourcelle, Patrice D. Cani, Jacqueline Marchand- Brynaert, V´ eronique Pr´ eat PII: S0169-409X(13)00029-X DOI: doi: 10.1016/j.addr.2013.01.002 Reference: ADR 12435 To appear in: Advanced Drug Delivery Reviews Accepted date: 30 January 2013 Please cite this article as: Anne des Rieux, Vincent Pourcelle, Patrice D. Cani, Jacqueline Marchand-Brynaert, V´ eronique Pr´ eat, Targeted nanoparticles with novel non-peptidic ligands for oral delivery, Advanced Drug Delivery Reviews (2013), doi: 10.1016/j.addr.2013.01.002 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Transcript
Page 1: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

�������� ����� ��

Targeted nanoparticles with novel non-peptidic ligands for oral delivery

Anne des Rieux, Vincent Pourcelle, Patrice D. Cani, Jacqueline Marchand-Brynaert, Veronique Preat

PII: S0169-409X(13)00029-XDOI: doi: 10.1016/j.addr.2013.01.002Reference: ADR 12435

To appear in: Advanced Drug Delivery Reviews

Accepted date: 30 January 2013

Please cite this article as: Anne des Rieux, Vincent Pourcelle, Patrice D. Cani,Jacqueline Marchand-Brynaert, Veronique Preat, Targeted nanoparticles with novelnon-peptidic ligands for oral delivery, Advanced Drug Delivery Reviews (2013), doi:10.1016/j.addr.2013.01.002

This is a PDF file of an unedited manuscript that has been accepted for publication.As a service to our customers we are providing this early version of the manuscript.The manuscript will undergo copyediting, typesetting, and review of the resulting proofbefore it is published in its final form. Please note that during the production processerrors may be discovered which could affect the content, and all legal disclaimers thatapply to the journal pertain.

Page 2: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

1

Targeted nanoparticles with novel non-peptidic ligands for oral

delivery

Anne des Rieux1, Vincent Pourcelle

2, Patrice D. Cani

3 Jacqueline Marchand-

Brynaert2, Véronique Préat

1*

1. Université catholique de Louvain, Louvain Drug Research Institute, Pharmaceutics and Drug Delivery

Research Group, Avenue E. Mounier, 73 B1.73.12, 1200 Brussels, Belgium

2. Université catholique de Louvain, Institute of Condensed Matter and Nanosciences, Laboratory of Solids,

Molecules and Reactivity, UCL L4.01.02, Place Louis Pasteur 1, 1348 Louvain-la-Neuve, Belgium

3. Université catholique de Louvain, Louvain Drug Research Institute, Metabolism and Nutrition Research

Group, Avenue E. Mounier, 73 B1.73.11, 1200 Brussels, Belgium

* corresponding author

Université catholique de Louvain

Louvain Drug Research Institute

Pharmaceutics and Drug Delivery Research Group

Avenue E. Mounier, 73 B1.73.12

1200 Brussels

Belgium

Tel : +32 2 764 73 09

Fax : +32 2 764 73 98

Email : [email protected]

Page 3: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

2

Abstract

Orally administered targeted nanoparticles have a large number of potential

biomedical applications and display several putative advantages for oral drug

delivery, such as the protection of fragile drugs or modification of drug

pharmacokinetics. These advantages notwithstanding, oral drug delivery by

nanoparticles remains challenging. The optimization of particle size and surface

properties and targeting by ligand grafting have been shown to enhance nanoparticle

transport across the intestinal epithelium. Here, different grafting strategies for non-

peptidic ligands, e.g., peptidomimetics, lectin mimetics, sugars and vitamins, that are

stable in the gastrointestinal tract are discussed. We demonstrate that the grafting of

these non-peptidic ligands allows nanoparticles to be targeted to M cells, enterocytes,

immune cells or L cells. We show that these grafted nanoparticles could be promising

vehicles for oral vaccination by targeting M cells or for the delivery of therapeutic

proteins. We suggest that targeting L cells could be useful for the treatment of type 2

diabetes or obesity.

Key words: Nanoparticles, oral delivery, ligand, non peptidic ligand, targeting

Page 4: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

3

Table of Content

1. INTRODUCTION ------------------------------------------------------------------------------- 4

1.1 CARRIERS AND GENERAL PROPERTIES -------------------------------------------------------------------- 4

1.2 MECHANISMS OF ORAL UPTAKE OF NANOPARTICLES ---------------------------------------------------- 5

1.3 TARGETING CELL RECEPTORS BY GRAFTING SPECIFIC LIGANDS TO THE NANOPARTICLE SURFACE ----- 7 1.3.1 Importance of receptor-mediated targeting ---------------------------------------------------------- 7 Limitations of peptidic and proteinic ligands ---------------------------------------------------------------- 7 1.3.2 ------------------------------------------------------------------------------------------------------------------------- 7 1.3.3 Advantages of non-peptidic small molecule ligands ------------------------------------------------ 8

2. GRAFTING OF NON-PEPTIDIC LIGANDS TO POLYMERS COMPOSING NANOPARTICLES --- 9

2.1 STRATEGIES FOR INTRODUCING NON-PEPTIDIC LIGANDS ONTO NANOPARTICLES ---------------------- 9

2.2 GRAFTING OF NON-PEPTIDIC LIGANDS TO A POLYMER ------------------------------------------------ 10

2.3 ANALYTICAL METHODS TO EVALUATE NON-PEPTIDIC LIGAND GRAFTING ----------------------------- 12

3. TARGETING OF NANOPARTICLES TO DIFFERENT INTESTINAL CELL TYPES --------------- 13

3.1 M CELLS -------------------------------------------------------------------------------------------------- 13

3.2 ENTEROCYTES -------------------------------------------------------------------------------------------- 17

3.3 GOBLET CELLS -------------------------------------------------------------------------------------------- 18

3.4 DENDRITIC CELLS ---------------------------------------------------------------------------------------- 18

3.5 ENTEROENDOCRINE CELLS AND THE IDENTIFICATION OF NOVEL POTENTIAL LIGANDS FOR

NANOPARTICLE DELIVERY --------------------------------------------------------------------------------------- 20

4. CONCLUSIONS REGARDING THE POTENTIAL BIOMEDICAL APPLICATIONS OF THE ORAL

DELIVERY OF NANOPARTICLES TARGETED WITH NON-PEPTIDIC LIGANDS ------------- 23

5. REFERENCES -------------------------------------------------------------------------------- 24

6. FIGURE LEGENDS --------------------------------------------------------------------------- 32

Page 5: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

4

1. Introduction

1.1 Carriers and general properties

The intestinal epithelium allows the absorption of nutrients, electrolytes and fluids

while acting as a defense system and an efficient barrier to macromolecules, toxins

and microorganisms. When particulate drug delivery systems are administered orally,

they are exposed to the harsh environment of the gastrointestinal tract. They must

cross the mucus layer before they come into contact with intestinal cells. Moreover,

they do not permeate easily across the intestinal barrier. However, fine-tuning the

size, shape and surface properties of the delivery system allows the enhancement of

drug-loaded nanoparticle (NP) absorption. A specific targeting effect can be achieved

by conjugating the ligands of receptors that are expressed at the apical site of

intestinal epithelial cells to the surfaces of the particulate delivery system. In other

words, the targeted delivery of drugs through the oral administration of particles

requires “smart” vehicles that are able to tolerate different conditions and cross

various barriers to entry via specific interactions with the targeted cell surface.

The general physicochemical properties required for orally delivered particles to

reach the attended sites of entry (e.g., gut-associated lymphoid tissue (GALT) or

enterocytes) or the desired sites of action (e.g., cancer cells or a zone of inflammation)

are beyond the scope of this paper and have been reviewed elsewhere [1-6]. This

review will focus on cell-NP-specific interactions that could be induced by specific

ligands that are present on the surfaces of the carriers. Nevertheless, we provide a

short overview of the influence of NP surface properties on their uptake by intestinal

cells because adequate surface chemistry promotes NP stability in gastrointestinal

(GI) tract fluids, increases their transit time in the gut and finally enhances their

ability to cross the mucosal barrier, including the mucus. This review will help to

identify the potential benefits of receptor targeting.

The nanometric particle size is of prime importance to allow the particle to cross the

mucus, avoid rapid clearance and finally enhance transmucosal transport. The

external shell should have a good hydrophilic and hydrophobic balance. Hydrophilic

molecules (i.e., PEGs, carbohydrates) are useful for the stabilization and diffusion of

the particles in the fluids and mucus, whereas a hydrophobic coating enhances cellular

and lymphatic uptake. The chemical functions of the materials play an important role

in the surface charge, the overall stability of the system and the non-specific

Page 6: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

5

interactions with surrounding media. Neutral surfaces facilitate the crossing of the

mucus barrier, whereas positively charged surfaces enhance interactions with the

mucus and negatively charged cell surfaces. In contrast, thiols, ammoniums and

lipophilic carbon chains reinforce adhesiveness and thus residency time by forming

disulfide bonds, electrostatic interactions and hydrophobic interactions, respectively.

Moreover, the presence of acidic or basic functional groups leads to pH-dependent

behaviors that are useful for targeting different portions of the GI tract.

In that context, degradable polymeric particles are particularly interesting [7]. Indeed,

polymers have well-defined properties (i.e., chain length, chemical functions, shape

and self-assembly) and can be derivatized using well-described chemistry. Therefore,

they constitute an interesting tool to produce tailored delivery vehicles. The most

widely described polymers for use in oral delivery are poly(glycolic acid) (PGA),

poly(lactic acid) (PLA) and their copolymers (e.g., PLGA, PLGA-PEG, and PLA-

PEG) [3,8], polyanhydrides [9], chitosan and its derivatives [10-12] and other

polysaccharides. As illustrated in Fig. 1, the surfaces of polymeric NPs can display

ligands that will bind to cell-specific receptors. Consequently, this review focuses

mainly on polymeric NPs.

1.2 Mechanisms of oral uptake of nanoparticles

The study of the mechanisms by which orally delivered drug loaded-NPs, whether

targeted or untargeted, are absorbed have attracted less attention than their design.

The design of new NPs for oral administration usually focuses on overcoming the

different barriers in the GI tract. The NPs and/or cargo must resist the harsh GI

environment, e.g., the low pH in the stomach and the degradative enzymes, but the

major barrier to their absorption remains the intestinal mucosa.

The first barrier that must be crossed is the mucus bilayer, which covers and protects

the epithelium. The NPs must adhere to and cross this highly viscoelastic layer, which

is continuously secreted and cleared. Mucoadhesive NPs, particularly positively

charged NPs, have been designed to promote strong interactions with the mucus and

prolong the retention time of the NPs at the mucosal surface. However, the

penetration of and diffusion through the mucus is also critical, as mucoadhesive NPs

can be trapped in the loosely adherent mucus and thus rendered vulnerable to rapid

Page 7: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

6

clearance [13]. NPs that penetrate the mucus and release drugs and/or drug-loaded

NPs closer to the epithelium have been engineered. Similar to viruses and bacteria,

NPs that are capable of penetrating the mucus should i) be small to allow their

diffusion in the mucin mesh; ii) have a non-mucoadhesive surface; and iii) be densely

coated, with a net neutrally charged hydrophilic surface [13]. The group of J. Hanes

has designed PEGylated muco-inert NPs that can penetrate the mucus. They found

that mucus-penetrating particles could be designed by carefully modifying their

surface properties, particularly by attaching a dense, low-molecular-weight PEG

[6,14]. In conclusion, a balance between minimal mucoadhesion or interactions with

the mucus and mucus penetration is required [6]. This also applies to targeted NPs,

which must be PEGylated to allow their penetration into the mucus and subsequent

access to the targeted receptor. Interestingly, the mucus layer is reduced at the surface

of M cells.

The second barrier to overcome for drug-loaded NPs is their generally limited cellular

uptake and translocation. In vitro models of the intestinal epithelium associated with

specific inhibitors or markers of endocytosis have been used to better understand the

fate of NPs in the intestines and optimize their design. It is generally accepted that

NPs do not diffuse through the paracellular route, although NP components, such as

chitosan, can affect tight junctions [1,6,12]. Rather, NPs can be taken up by

phagocytosis, which is restricted to M cells, or by pinocytosis. The uptake and

transport of particles by M cells is significantly higher than their transport by

enterocytes. Pinocytosis can occur by macropinocytosis, clathrin- or caveolae-

mediated endocytosis or clathrin- and caveolae-independent endocytosis [6,15,16].

The mechanism of uptake will affect subsequent intracellular trafficking and

transcytosis. The physicochemical properties of the particles influence their uptake,

e.g., the smaller the particle, the higher the uptake [1]. One strategy to enhance NP

uptake by intestinal cells is to conjugate a ligand to the NP surface that will favor cell-

NP interactions and enhance NP internalization, primarily through clathrin-mediated

endocytosis.

Page 8: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

7

1.3 Targeting cell receptors by grafting specific ligands to the nanoparticle

surface

1.3.1 Importance of receptor-mediated targeting

Despite the promising results obtained for NPs with appropriate size and surface

properties, the absorption and/or in vivo therapeutic efficiency of drugs following the

oral administration of drug-loaded NPs typically remains low [1-6,17]. The mucus

penetration, cellular uptake, NP trafficking inside the cells and biological fate of the

delivered drugs are still not optimal. Some studies have shown that conjugating the

NP surface with specific ligands for epithelial receptors or antibodies might enhance

the specific cellular uptake and transepithelial transport of the NP [1,6,18]. Moreover,

in the constantly moving environment of the gut, particles are rapidly cleared, and

thus, strong associations (e.g., receptor-ligand interactions) would favor the

accumulation of particles at their sites of action or absorption [2].

A large variety of epithelial cell receptors have been investigated as potential targets

of delivery systems [1,6,18]. Based on this knowledge, studies on ligand (or

antibody)-modified oral drug delivery systems have emerged in the last decade,

leading to promising in vitro results but unclear and often disappointing in vivo

conclusions.

1.3.2 Limitations of peptidic and proteinic ligands

In addition to the challenges presented by interactions with mucus and cells and the

poor uptake of NPs by cells, the chemical nature of ligands grafted onto the NP

surface can also play a role in the particle fate. In the GI tract, ligands present on the

NP surface are exposed to different harsh chemical conditions, such as the acidic pH

of the stomach and enzymatic digestion, among others. Moreover, most of the ligands

employed are biomacromolecules (e.g., antibodies and bacterial proteins) that can

diminish the progression of NP in the mucus by increasing their size and the

incidence of non-specific interactions, and they may also induce steric hindrance

when they approach the receptor surface [2]. Some of these biomolecules are also

immunogenic (Fc).

One critical consideration for ligands is the chemical method used for their

introduction in the drug delivery system. While it seems obvious that covalent

conjugation is mandatory, the chemistries that can be used and the chemical

Page 9: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

8

properties of both the carrier and the ligand are not necessarily easy to combine.

Biomacromolecules often have a large number of hydroxyl, amino or thiol functional

groups and are delicate compounds. In addition, the chemical characterization of the

obtained bioconjugates is a demanding task that requires an entire development

process in itself. It is thus difficult to determine whether a ligand is attached to the

particles by the desired chemistry, how much is bound and whether it is still bioactive

after conjugation [19,20].

Another limitation of targeted drug delivery systems is the accessibility and adequate

bioactive conformation of ligands in the biological media. These are indeed difficult

questions to address because during the formulation of the NP, it is challenging to

assess where the ligands will be located (i.e., outside or inside the particles).

Moreover, in the GI environment, the carrier can be reorganized, and the

conformation of the ligands can change. Several recent studies have attempted to

answer some of these questions. Most notably, the water solubility of the ligand

influences its localization in the NP and, consequently, its targeting efficiency. For

instance, in a self-assembling process, a hydrophilic ligand is more efficient than a

hydrophobic ligand because of its greater representation in the outer shell [21].

Furthermore, the nature of the ligands and their surface densities, might also play a

role in the level and nature of the immune response when antigen-loaded NPs are

targeted to dendritic cells [22]. These are controversial questions, and it is not yet

clear whether higher ligand density will lead to improved targeting because targeting

also depends strongly on the aforementioned surface characteristics of the NP [23],

the way ligands are tethered [24,25] and the affinity of the ligand for the targeted

receptor [22,25,26].

1.3.3 Advantages of non-peptidic small molecule ligands

Considering the above-mentioned limitations, the use of small molecule ligands (i.e.,

molecules of less than ~1500 Da), such as sugar derivatives, peptidomimetics or

metabolites, seems to have several advantages over peptidic conjugates: i) they are

chemically resistant to GI conditions (or could be modified for this purpose); ii) they

are usually conformationally stable; iii) they do not induce steric hindrance at the

receptor surface; iv) they can be conjugated to the NP through simple chemical

procedures, and the obtained conjugates can be more easily characterized; v) they are

Page 10: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

9

easier and less expensive to produce; and vi) they do not have immunogenic effects.

The main small molecule ligands that have been investigated in oral delivery are

mannose derivatives, RGD or LDV peptide sequence mimetics, lectin mimetics, fatty

acids and vitamins (see table 1).

2. Grafting of non-peptidic ligands to polymers composing nanoparticles

2.1 Strategies for introducing non-peptidic ligands onto nanoparticles

The conjugation of small ligands to a wide variety of biomaterials has been

extensively studied for applications ranging from tissue engineering to the conception

of targeted imaging agents. In targeted nanoparticulate delivery systems, particularly

those designed for oral administration, the conjugation methods are more limited.

Careful attention should be paid to the localization and density of the ligands.

Moreover, the biodegradable polymers employed can be degraded during the grafting

procedure. The chemical bonds should resist hydrolysis by GI pH and enzymes.

Consequently, the conjugation strategy should consider i) the chemical reaction used

for the anchorage (i.e., how to conjugate the ligands); ii) the localization of the ligand

along the polymer backbone (i.e., along the chain or at its end); and iii) the timing of

the conjugation step with respect to the overall NP synthesis process (i.e., before or

after NP formulation).

For instance, PLA, PLGA and their derivatives suffer from transamination and

hydrolysis reactions, even under very mild conditions [27]. Consequently, gentle,

selective and efficient chemistries must be used for their functionalization. Click

chemistry with copper-catalyzed azide alkyne cycloaddition (CuAAC) is the method

of choice for this type of polymer because it is a very tolerant reaction that occurs in

organic solvents without requiring amines.

Otherwise, modular approaches must be considered, such as our “clip and click”

strategy, in which PEG is first functionalized through “clip photochemistry” grafting

and then attached to a terpolymer of poly(lactide-co-glycolide-co--caprolactone) by

CuAAC [28,29]. A common strategy for attaching ligands to PLGA-based NPs is to

use poly(-caprolactone) (PCL) or PCL-PEG, which have the same properties as

PLGA but are more chemically resistant. Ligand-PCL conjugates can subsequently be

introduced to PLGA-based NPs. To ensure the protection of PLGA while using more

conventional chemistry (e.g., carbodiimide activation), some authors have worked

Page 11: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

10

directly with the particles after their formulation. This allows the PLGA core to be

protected against degradation and ensures that the ligands are present on the external

surface, but the risk of particle leakage and loss of payload cannot be avoided. Other

carriers used for conjugation with small ligands include reactive polymers (e.g.,

poly(anhydride)) and stable compounds that can tolerate typical chemical treatments

(i.e., PEGs, lipids, and polysaccharides).

The methods of conjugation will also influence the localization of the ligand along the

polymer backbone. However, it has not been clearly established that ligand

localization at the end of a polymer chain will generate better nanoparticle targeting

than a ligand grafted along the carrier backbone [24]. Moreover, the final availability

of the ligand may depend on the self-assembly process and the ligand characteristic

[21].

2.2 Grafting of non-peptidic ligands to a polymer

The strategies for ligand grafting are schematized in Fig. 2 and will mainly be

illustrated with PLGA and PCL, two biodegradable polymers that are commonly used

for nanoparticle formulation. Most authors have chosen chain-end approaches with

copolymers bearing reactive functions at the chain end of the hydrophilic block that

can react with the ligand via usual coupling methods (e.g., reductive amination,

ammonium formation, carbodiimide coupling, and polymerization initiated by

ligands). Recently however, several interesting methods have been developed to

obtain ligands that are grafted along the polymer chain (i.e., click chemistry, clip

chemistry and reactive polymers).

Reactive polymers can be grafted directly. Because poly(anhydride) (i.e., poly(methyl

vinyl ether-co-maleic anhydride)) is a reactive polymer, it forms naturally covalent

conjugates through the reaction of the amine functional group of modified VB12 with

the anhydride functional groups of the polymer (Fig. 2A) [30].

Grafting of the RGD sequence to NPs has been investigated as a strategy to target

drug-loaded NP to integrins that are overexpressed by specific cells, particularly v3

in the angiogenic tumor endothelium [31] and 1 integrin in intestinal M cells of the

follicle-associated epithelium [32]. However, because RGD might be degraded in the

GI tract, non-peptidic analogues have also been investigated. RGD peptidomimetic

Page 12: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

11

(RGDp) sequences have been developed for potential applications in antiangiogenesis

treatment [33] or tissue engineering, but few have been used in targeted drug delivery

systems [34-38]. The works conducted by our group [32] [39] provide clear evidence

that RGDp-targeted PLGA NPs are superior to peptidic RGD-targeted NPs in

inducing an immune response against a model antigen (ovalbumin) in mice. These

RGDps are built on a tyrosine template [40] and mimic the bioactive conformation of

cRGDf(NMe)V inside the binding pocket of v3 integrin. These ligands were aimed

at biomaterial applications [41] and have a short oligo ethylene glycol (OEG) spacer

arm. They were conjugated via a “clip photochemistry” process to the PEG parts of a

PCL-PEG polymer before their incorporation with PLGA during the formulation of

the NPs [32,39,42]. The presence of the ligands in the shell of the NPs was confirmed

by surface chemistry analysis using X-ray photoelectron spectroscopy (XPS) [43].

Other targeting ligands, such as a Leu-Asp-Val (LDV) modified sequence and a LDV

peptidomimetic (LDVp) with an OEG spacer arm, initially designed to bind 41

integrins, have also been grafted onto PCL-PEG and incorporated into PLGA-based

NPs [39,44].

C-type lectins and mannose receptors are the most explored molecules for the

targeting of antigen-presenting cell (APC) receptors. Consequently, mannose-grafted

particles, intended to for dendritic cell or macrophage targeting, have been

investigated [45], including their activity when administered via the oral route. Recent

reviews [46-48] have more precisely detailed such chemistry. Thus, they will not be

described here, but some examples of the most relevant strategies should be

mentioned. For example, Rieger et al. produced PCL-PEG with a mannose residue on

the chain end of the PEG block [49,50]. They synthesized PCL-PEG terminating with

a secondary amine function that reacted with a mannose derivative equipped with a

small ethyl spacer ended by bromine to form an ammonium link. The obtained

copolymer easily formed positively charged micelles bearing mannose residues on

their surfaces (Fig. 2B). Similarly, Freichels et al. produced aldehyde-terminated

PCL-PEG that, through a reductive amination protocol with 2-aminoethyl-α -D-

mannopyroside, allowed for the production of end-capped mannosylated PCL-PEG

[51]. PCL-PEG micelles prepared with these modified polymers displayed a neutral

surface charge. Click chemistry can also be used to produce mannosylated PCL

bearing sugar residues along the entire polymer chain. For that purpose, Xu et al.

Page 13: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

12

produced PCL diblock polymers in which one block had azide functional groups that

could be engaged in a CuAAC reaction with propargylic sugar derivatives [52] (Fig.

2C). The large number of carbohydrates fixed on one block of the polymer confers

amphiphilic properties to this PCL-based polymer. We also successfully applied our

“clip photochemistry” process to the preparation of PCL-PEG grafted with mannose

(Fig. 2C) [42]. This mannosylated PCL-PEG was subsequently introduced into the

formulation of PLGA-based NPs [39].

Careful and demanding strategies are required to functionalize PLGA or PLA

polymers with mannose derivatives. For instance, Nagasaki used protected sugars to

induce the sequential ring-opening polymerization of ethylene oxide and DL-lactide

[53]. Deprotection yielded PLA-PEG chains that were end capped with carbohydrates

and demonstrated interesting micellization properties. Hamdy et al. synthesized

mannan-conjugated PLGA NPs by direct functionalization of the carboxylate

functions on the surface of preformed OVA-loaded NPs via a classical carbodiimide

protocol [54] (Fig. 2B). Similarly, Brandhonneur et al. produced PLGA particles

bearing different ligands (including lectin, RGD and mannose) and demonstrated their

enhanced uptake by macrophages in vitro [26]. The aforementioned “clip and click

strategy” allows the production of different functionalized PLGA-PEG with targeting

moieties randomly dispersed along the PEG block to produce mannosylated PLGA-

PEG copolymer with self-assembling properties [29].

2.3 Analytical methods to evaluate non-peptidic ligand grafting

Due to the uncertainties inherent in self-assembling processes, it is usually difficult to

predict where ligands will be localized. Consequently, new characterization methods

must be investigated in parallel with conjugation strategies.

The most common test to detect sugar moieties at the particle surface is to provoke

the aggregation of sugar-conjugated NPs in the presence of free soluble lectins and

further analyze them using isothermal titration calorimetry, enzyme-linked lectin

assays (ELLAs) [50], turbidimetry, surface plasmon resonance [55] or affinity column

retention assays [53]. NP aggregation upon binding to lectin is clear proof of the

presence of bioavailable sugar ligands. Quartz crystal microbalance with dissipation

(QCM-D) affinity binding assays have also been conducted, with mannosylated NPs

interacting with lectins immobilized on a QCM-D surface [29]. This test provides not

Page 14: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

13

only information about the availability of mannose for lectins on a surface but also an

opportunity to assess the relevance of the NP-receptor interaction under shear stress,

representing a “near to reality” evaluation of targeted NP behavior.

The chemical characterization of the NP surface can be performed by surface

chemistry techniques, such as X-ray photoelectron spectroscopy (XPS), but only

solids can be analyzed using this technique. NMR could be a good alternative for

studying particles in aqueous solution [20,56,57]. Indeed, in magnetic resonance, only

soluble components are detected. This provides valuable information regarding the

bioaccessibility of the constituents of the external shell of NPs, and the micellar

stability of the NP. Under appropriate conditions, NMR can also be used for

quantitative calculations of ligand accessibility and the lengths of the PEG chains

[58].

3. Targeting of nanoparticles to different intestinal cell types

Polymeric carriers protect antigens against degradation and inactivation in the harsh

gastro-intestinal environment and have the ability to enhance their transmucosal

transport [1,6,17]. Despite these advantages, the oral delivery of drug- or antigen-

loaded NPs remains challenging. The poor efficacy of these particulate systems has

been reported, possibly as a consequence of poor particle uptake. Thus, none of these

systems has reached the market. Methods for increasing the uptake and transcytosis of

orally delivered particles by specific cells represent a promising opportunity for

enhancing their efficacy. Due to their physiological functions, M cells are a

particularly attractive target for oral drug delivery [1,6,18,59,60]. Enterocytes, which

cover most of the gut surface and are responsible for absorption, could also be a good

target for NP uptake. Epithelial immune cells that sample particulate antigens and

microorganisms can also be exploited for NP uptake. Finally, endocrine cells, which

are rarely studied for NP targeting, could be potential new targets, mainly for local

drug delivery.

3.1 M cells

M cells are specialized epithelial cells that are located in the follicle-associated

epithelium (FAE) of Peyer’s patches or GALT [59-61] and are part of the mucosal

Page 15: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

14

immune system. They deliver samples of foreign material from the lumen to the

underlying organized mucosa lymphoid tissues to induce immune responses. They

have high transcytotic capabilities and are able to transport a broad range of materials,

such as bacteria, viruses, antigens and particles, from the intestinal lumen to the

underlying lymphoid tissues [1]. In addition, M cells have less glycocalyx and

reduced levels of membrane hydrolase activity, which can influence the fate of

protein-containing or protein-coated NPs, compared with normal epithelial cells.

Villous M cells located outside the FAE have also been observed [62], but the

transport of antigens and microorganisms across the intestinal mucosa is carried out

mainly by FAE-M cells [1]. Although they are less numerous than enterocytes, M

cells present enhanced transcytosis abilities, which makes them particularly

interesting for oral drug delivery applications. Therefore, M cells represent a potential

portal for the oral delivery of drug-loaded NPs, particularly peptides, proteins and for

the mucosal vaccination.

Although the role of M cells in particle uptake is well known, it is commonly believed

that their low proportions in the human GI tract (1% of the total intestinal surface) and

their variability among species (i.e., they represent 5% to 50% of the FAE surface and

express species-specific markers), individuals, physiological state and age decrease

the impact they could have on oral drug delivery [1]. However, in light of their

particle uptake capabilities, several groups have considered it worthwhile to work on

improving NP delivery through M cells, particularly with the aim of compensating for

the low number of M cells through more efficient targeting [1,18,32,39,59,60,63-69].

The main strategy has been to coat the NP surface with an M cell-targeting molecule.

This task is not trivial, especially considering the limited predictive value of the most

commonly employed mouse models and the difficulties in identifying markers that are

specific to human M cells.

The most investigated family of M cell-targeting molecules is the lectins. Lectins

constitute a structurally diverse group of proteins and glycoproteins that bind

reversibly and with relatively high affinity to specific carbohydrate residues present

on cell surface proteins or lipids [18]. In several studies, the Ulex europaeus

agglutinin-1 (UEA-1) lectin was grafted to the NP surface to target -L-fucose

residues expressed on the apical surface of M cells, yielding improvements in

nanoparticle transport across the intestinal barrier [66,67,70]. The conjugation of

Page 16: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

15

PLGA particles to Aleuria aurantia lectin (AAL) induced an increased expression of

IFN upon oral birch pollen immunotherapy [18] and effectively protected mice

against subcutaneous challenge with melanoma or prostate cancer cells [63].

However, the UEA-1 and AAL lectins, in addition to their potential immunogenicity,

are specifically expressed on mouse and not human M cells [60], limiting the

translation of these therapies to humans.

Many studies have been dedicated to the search for specific markers of human M

cells. Two lectins specific to the human FAE (galectin 9 and sialyl Lewis A antigen

(SLAA)) have been identified [65,71]. One anti-SLAA antibody out of the 41 tested

lectins and antibodies reacted strongly with human M cells and bound only weakly to

FAE enterocytes [59]. Galectin 9 has not been used as a targeting molecule for the

oral delivery of NP.

To target M cell receptors, it may be advantageous to replace lectins with small

molecule mimetics. Lambkin et al. identified UEA-1 lectin mimetics from a

combinatorial library [70,72]. These compounds are easy to synthesize and are based

on an oligo-lysine scaffold with 1 to 4 lysine units terminated by galloyl entities. The

lectin mimetics were coupled to a PEG construct through p-nitrophenyl-activated

esters to form a tetragalloyl-D-lysine dendrimer (TGDK) that was used to vectorize a

Rhesus CCR5-derived cyclopeptide antigen. Misumi et al. conducted in vivo studies

on macaques with this TGDK and found a significant stool IgA response and efficient

M cell transcytosis of the dendrimer, which induced neutralizing activity against SIV

infection [68]. TGDK was efficiently transported from the lumen of the intestinal tract

into Rhesus Peyer’s patches by M cells and then accumulated in germinal centers. In

addition, TGDK specifically bound to human M-like cells in vitro and was efficiently

transcytosed from the apical side to the basolateral side in the M-like cell model (Fig.

3).

M cells are considered the gateways for antigen entry to the underlying mucosal

tissues, and they are exploited by various enteric pathogens as a route of entry to the

underlying host tissue, predominantly through the hijacking of their endocytic

machinery [18]. The invasiveness of these viral and bacterial pathogens is mediated

by specific pathogen–host interactions, which could be adapted to deliver drug-loaded

NPs into the GALT [60,69]. Some important pathogen recognition receptors (PRRs),

such as Toll-like receptor-4 (TLR-4), platelet-activating factor receptor and 51

Page 17: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

16

integrin, are expressed on the surfaces of human and mouse M cells [32,39,73]. The

specific pathogen–host interactions are crucial for the translocation of bacteria across

the lumen. Consequently, targeting PRRs might be a suitable strategy for enhancing

the uptake of orally administered NPs by M cells [59].

For instance, M cells take up many enteropathogenic microorganisms, such as

Yersinia, via the high-affinity interaction between invasin and the 51 integrins that

are overexpressed at the apical pole of human M cells [32,74,75]. Interactions with

5 integrin occur mainly through RGD sequences. We have demonstrated that

grafting RGD to the PEG chains of PLGA-based NPs significantly increases the in

vitro transport of these NPs by human M-like cells [32] (Fig. 4A) and slightly

enhance the IgG immune response after oral immunization [32]. We hypothesized

that these phenomena were due to a partial degradation of the RGD peptide during its

trafficking through the GI tract. Therefore, an RGD peptidomimetic (RGDp) was

grafted onto PEGylated PLGA-based NPs. RGDp significantly increased the transport

of NPs across an in vitro model of human M cells (Fig. 4B), and intraduodenal

immunization with RGDp-labeled NPs elicited a higher production of IgG antibodies

than the intramuscular injection of free ovalbumin or the intraduodenal administration

of either non-targeted or RGD-NPs [39]. NPs conjugated to LDVp also exhibited

greater transport by M cells in vitro and showed promising immune responses

compared to untargeted NPs, suggesting that these LDV ligands might have bound to

1 integrins on the apical surface of M cells or other integrins homing lymphocytes in

the gut [76].

The Clostridium perfringens enterotoxin (CPE) receptor (claudin 4) is a tight-junction

transmembrane protein that plays a role in establishing transepithelial electrical

resistance in the mucosal epithelium in addition to its function as a receptor for CPE

[77]. Claudin 4 is highly expressed in M cells and is conserved between mouse and

human Peyer’s patch. Targeting PLGA NPs to claudin 4 enhanced their in vivo uptake

and mucosal IgA responses [78].

Recently, Hase et al. [79,80] reported that glycoprotein 2 (GP2) is specifically

expressed at the apical surface of human and murine M cells and serves as a

transcytotic receptor for fimH+ bacteria (e.g., Escherichia coli, Salmonella enterica

and Yersinia). Thus, the GP2-dependent transcytosis pathway could provide a new

target for the development of M cell-targeted nanosystems.

Page 18: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

17

Reovirus in mice and poliovirus and HIV in humans use specific receptors to target

and cross the FAE [81-83]. Reovirus can invade intestinal M cells in rodents and

rabbits through interactions between its outer capsid protein σ1 with α(2,3)-linked

sialic acid containing glycoconjugates of the apical membrane [82]. The incorporation

of recombinant σ1 into liposomes or an OVA-σ1 fusion protein enhanced binding

to rat Peyer’s patch [84]. HIV-1 can adhere to M cells in mice and rabbits prior to its

endocytosis and transport across the epithelial barrier [85]. A lymphotropic (X4)

HIV-1 strain crosses M cell monolayers and infects the underlying CD4+ target cells.

This transport requires both the lactosyl cerebroside and CXCR4 receptors, which are

expressed on the apical surface of Caco-2 and M cells [81]. In contrast, a monotropic

(R5) HIV-1 strain is unable to cross M cell monolayers and infect underlying

monocytes. Caco-2 and M cells do not express CCR5, but the transfection of these

cells with CCR5 cDNA restores the transport of the R5 virus [81].

3.2 Enterocytes

The importance of enterocytes should not be overlooked; these cells vastly outnumber

M cells and can transcytose many macromolecules, such as cholera toxin (CT) and F4

fimbriae, as well as inert particles [86-89]. For instance, several Escherichia coli

strains express F4 fimbriae on their surface and bind to specific F4 receptors (F4Rs).

The expression of these receptors on the surface of porcine enterocytes is necessary to

induce a protective mucosal immunity following the oral administration of purified F4

fimbriae to piglets [88]. The conjugation of heterologous antigens to F4 fimbriae has

been shown to induce enhanced mucosal antigen-specific antibody responses upon

oral administration, but to date, no study evaluating the influence of F4 fimbriae

targeting on the oral transport of nanocarriers has been reported. The incorporation of

flagellin-rich Salmonella enteritidis extracts into Gantrez AN NPs induced

bioadhesion in the ileum during “Salmonella-like” gut colonization [90].

TLR-4, the receptor for LPS, mediates bacterial translocation through enterocytes

[91]. The rat enterocyte cell line IEC-6 internalized LPS-coated latex beads in a

TLR4-dependent manner, indicating that LPS-coated particles may provide yet

another alternative for the targeted delivery of NPs to the intestinal epithelium [18].

Some lectins (e.g., wheat germ agglutinin (WGA), concanavalin A (ConA) and

tomato-derived lectin) bind to enterocytes, with a relatively high affinity for the

Page 19: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

18

specific carbohydrate residues present on cell surface proteins or lipids [18,92-94].

They have been grafted onto the surfaces of nanoparticles for oral vaccines or drug

delivery.

Some authors have proposed using certain metabolic pathways as routes for NP

uptake. This strategy was attempted with vitamin B12 (VB12) or B1 (thiamine)-

coated NPs. VB12 forms a complex with the intrinsic factor (IF) in the stomach,

which is subsequently recognized by IF receptors on ileal epithelial cells, resulting in

the endocytosis of VB12. VB12-targeted micelles exhibited better in vitro uptake and

transport of a hydrophobic drug in a model intestinal cell monolayer in comparison to

untargeted micelles [30,95]. Similarly, Salman et al. also produced thiamine-

poly(anhydride) NPs, which could be advantageous because thiamine can be

administered at a higher daily dose than VB12. Encouraging immunization results

were also obtained with these thiamine-coated vehicles in mice, although no direct

comparison with VB12 NPs was made [96].

3.3 Goblet cells

Goblet cells represent the second largest population of intestinal epithelial cells, but

they are rarely chosen as a target for orally delivered nanocarriers. Recently, a CKS

peptide identified using a random phage display technique was found to have a

specific affinity for goblet cells [97]. Orally administered insulin-loaded trimethyl

chitosan chloride NPs that were modified with the CSK targeting peptide induced

enhanced transport via clathrin- and caveolae-dependent endocytosis and produced a

better hypoglycemic effect than non-targeted NPs [98].

Furthermore, Listeria monocytogenes is transcytosed across the intestinal barrier by

binding to E-cadherin, which is luminally accessible on goblet cells, suggesting that

targeting E-cadherin could be a promising strategy for delivering NP to goblet cells

[99].

3.4 Dendritic cells

Dendritic cells (DCs) represent the most potent APCs. They are found throughout the

intestine and can be divided into two major subsets, which can be distinguished based

on the expression of CD103 (the E chain of the E7 integrin), the receptor for the

epithelial cell adhesion molecule E-cadherin, and CX3CR1 [100]. Intestinal DCs have

Page 20: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

19

been proposed to be involved in the induction of protective immunity against

pathogens, tolerance to commensal bacteria and tolerance to food antigens and self-

antigens. Thus, they represent a potent target for oral vaccination strategies because

vaccine interactions with DCs can be enhanced by targeting DC surface molecules

[18,101]. There are very few, if any, DC-specific markers, but these cells possess a

broad spectrum of cell surface receptors that are involved in endocytosis and the

induction of immune responses, such as C-type lectins, scavenger receptors, TLRs

and Fc receptors (FcRs: FcgR, FcaR and FceR, which bind to IgG, IgA and IgE,

respectively) [102].

The outcome of the immune response can differ depending on the targeted receptor.

TLR ligands induce strong DC activation and thus have potent adjuvant properties.

The C-type lectin DEC-205 and mannose receptors are more involved in enhancing

endocytosis, although DC activation has been achieved with DEC-205 targeting

[101]. The FcRs are a family of membrane glycoproteins that bind the Fc fragment

of IgG and activate a signaling pathway that can regulate the adaptive immune

response when cross-linked with antigen-antibody immune complexes.

[26,54,100,103].

Mannan and other mannosylated structures enhance antigen endocytosis by DCs and

induce immune responses when grafted to a vaccine carrier surface [104,105].

Mannosylated liposomes bind to DC-SIGN and the mannose receptor CD206,

resulting in enhanced antigen-specific cell proliferation relative to antigen alone or

non-targeted liposomes, and they protect mice from lethal challenge when delivered

intraperitoneally. Mannosylated, PEGylated PLGA NPs elicited higher antigen-

specific IgG serum responses in mice upon intraduodenal administration than non-

grafted NPs [39]. They also enhanced NP uptake in a mouse model of colitis [106].

Targeting intestinal DCs enhances the endocytosis of antigen-loaded carriers and can

thus improve immune responses, but most of the studies investigating this

phenomenon have been performed in vitro or in murine models. Therefore, further

studies are required to i) evaluate vaccine carriers targeting intestinal DCs of higher

species (pigs and primates), ii) analyze the behavior of antigen-loaded carriers in the

GI tract and iii) deepen our understanding of the interactions between vaccine carriers

and intestinal DCs.

Page 21: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

20

3.5 Enteroendocrine cells and the identification of novel potential ligands for

nanoparticle delivery

The enteroendocrine system constitutes the largest endocrine organ. Enteroendocrine

cells are scattered throughout the GI tract in the epithelium among enterocytes. These

cells are typically conically shaped, with a large base from which gut hormones are

released into the blood from secretory granules. These cells are distributed along the

GI tract, and the apical pole facing the gut lumen possesses microvilli. Strikingly,

these cells are among the least understood cells in the body. However, new molecular

genetic techniques have led to important advances, thereby highlighting novel aspects

of enteroendocrine biology [107]. Enteroendocrine cells represent approximately 1%

of all epithelial cells in the intestine and are subdivided into more than 10 different

cell types based on their main secretory products and localization along the GI tract

(e.g., ileal/colonic L cells). Multiple biological functions are physiologically regulated

by gut hormones (e.g., food intake, gastric emptying, gut motility, gut barrier function

and glucose metabolism). Among the enteroendocrine cells, L cells have attracted

particular interest because of the pleiotropic effects of their secreted peptides.

In the gut, the posttranslational processing of proglucagon in endocrine L cells gives

rise to the major proglucagon-derived peptides (GLP-1, GLP-2, oxyntomodulin and

glicentin) [108]. These peptides are rapidly secreted in response to food intake, and

their production is modulated according to the nutrient (i.e., lipid, carbohydrate, and

protein) [109]. L cells secrete another anorexigenic peptide, PYY. Similar to GLP-1

administration, PYY injection delays gastric emptying and pancreatic and gastric

secretions. GLP-2 is co-secreted by L cells with GLP-1. GLP-2 assists in the

maintenance of the physiological gut barrier function (i.e., protects against gut

microbiota) and facilitates the digestion and absorption of ingested nutrients. In

addition, GLP-2 regulates the stimulation of intestinal epithelial cell proliferation

[110] and constitutes a key target in the maintenance of gut barrier function.

Given the location of these cells (the ileum and colon), targeting L cells and thereby

the endogenous production of these hormones remains challenging. Therefore,

strategies devoted to targeting specific receptors involved in L cell physiology, thus

leading to the production of hormones, such as GLP-1, GLP-2 and PYY, could be a

promising potential method to locally influence the mechanisms involved in high-

impact diseases, such as diabetes, obesity and inflammatory diseases.

Page 22: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

21

Recent studies have shown that L cells express various G-protein-coupled receptors

(GPCRs) that are activated by a wide variety of endogenous ligands found in the gut

lumen. GPCRs are involved in a large number of physiological processes and could

serve as targets for coated NPs. Potential ligands for these receptors include specific

lipids from short-chain fatty acids (i.e., GPR43, GPR41 and GPR109a) [111,112] and

long-chain fatty acids (GPR40 and GPR120) [113]. Interestingly, GPR43 and GPR41

expression is well conserved across species (human, pigs, and rodents) [114]; both

receptors are expressed in L cells and have been shown to directly control GLP-1 and

PYY secretion (Fig. 5) [115]. Although a direct link between GPR43 or GPR41 and

GLP-2 secretion has not been established, we have demonstrated that changing the

composition of the gut microbiota using specific nutrients that increase Short-chain

fatty acids (SCFA) (fermentable carbohydrate) stimulate endogenous GLP-2

production (Fig. 5) and protect against gut permeability and associated inflammation

[116]. Moreover, a similar treatment increases PYY and GLP-1 secretion.

Bioactive lipids belonging to the N-acylethanolamine family that are part of the

endocannabinoid system [117,118] could also activate the L cell-specific receptor

GPR119, thereby increasing GLP-1, GLP-2 and PYY secretion. Finally, TGR5 (also

known as M-BAR, GPBAR-1 or GPR131) can be activated by bile acids [119].

SCFAs are present in the gut lumen (ileum and colon) at a high concentration

(approximately 100 mM) [120]. These fatty acids are mainly produced through the

metabolic activity of the gut microbiota (undigested carbohydrate fermentation). Over

the last 30 years, numerous roles have been attributed to SCFAs, including the

harvesting of energy from undigested food, the regulation of epithelial cell

proliferation, electrolyte uptake and smooth muscle contraction [112], and SCFAs

could thus be used as NP ligands to act on L cell metabolism.

Another potential target is the GPR109A receptor, which binds to the ketone body β-

hydroxybutyrate and butyrate [114,115]. The GPR109A receptor is highly expressed

in the gut lumen, in which the concentration of butyrate reaches approximately 20

mM, thereby activating this receptor. The intestinal GPR109A receptor could

therefore be targeted by butyrate-labeled NPs.

The postprandial satiety effect of dietary lipids and free fatty acids stimulates several

gut peptides that control food intake. Growing evidence suggests that these effects are

mediated through two different receptors, GPR40 and GPR120. Both receptors are

Page 23: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

22

activated by medium- to long-chain free fatty acids that stimulate gut peptide

secretion [113]. It is worth noting that similar strategies and ligands might be used to

target L cells (i.e., GLP-2 production) and improve gut barrier function.

Bile acids are not only byproducts of cholesterol metabolism but also key metabolic

regulators that act through TGR5 [121]. For instance, Katsuma et al. discovered that

bile acids promote GLP-1 secretion through a TGR5-dependent mechanism [122],

thereby suggesting a novel role of bile acids in energy metabolism and glucose

homeostasis. Interestingly, a recent study has demonstrated the relevance of targeting

TGR5 in experimental colitis [123]. TGR5 activation has been shown to exert a

peripheral immune-modulatory effect in macrophages, and this study also showed that

TGR5 activation through a targeting strategy restores tight-junction protein

distribution, leading to reduced gut permeability [123]. For decades, ciprofloxacin has

been used for the treatment of Gram-negative bacterial infections occurring in the

context of Crohn’s disease. Cipriani et al have demonstrated that ciprofloxacin also

acts as a TGR5 agonist, thereby contributing to improvement of the inflammatory

status [123]. This last study supports the concept that the TGR5 receptor might be

targeted by not only specific bile acids but also synthetic molecules, such as

ciprofloxacin. While they are effective for treating infections in Crohn’s disease

patients, the wide use of antibiotics plays a minor role in the maintenance therapy for

these patients. Therefore, we propose that targeting TGR5 using NPs carrying a lower

dose of ciprofloxacin might be useful to target colon cells or macrophages to treat

inflammatory bowel disorder, preventing the major adverse effects linked to the

antibiotic activities of this compound.

In conclusion, although endogenous non-peptidic ligands have helped to identify the

different GPCRs described here, numerous synthetic agonists are also currently being

studied and may constitute a source of therapeutic agents that merit future

consideration [124]. Moreover, it is worth noting that in addition to their roles in the

control of gut peptide secretion, most of these receptors are also expressed in immune

cells, such as macrophages. Therefore, we propose that the oral delivery of NPs with

non-peptidic ligands targeting GPCRs of L cells could lead to new treatments, acting

locally on obesity, type 2 diabetes and intestinal inflammation. Thus, by exploring the

wide variety of L cell stimulation-dependent effects (e.g., GLP-1, GLP-2 and PYY

secretion) on GI tract physiology and peripheral metabolism (energy and glucose

Page 24: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

23

homeostasis), we propose to provide valuable insight into some novel beneficial

therapeutic targets.

4. Conclusions regarding the potential biomedical applications of the oral

delivery of nanoparticles targeted with non-peptidic ligands

Orally administered targeted NPs have a large number of potential biomedical

applications and exhibit several putative advantages for oral drug delivery, such as the

protection of fragile drugs or the potential for the modification of drug

pharmacokinetics. Despite these advantages, the oral delivery of drugs by NPs

remains challenging. To achieve efficient drug delivery, NPs must i) avoid rapid

mucus clearance; ii) penetrate the mucus layer; and iii) be extensively taken up by the

intestinal epithelium. The optimization of particle size and surface properties and the

targeting of specific cells by ligand grafting have been shown to enhance NP transport

across the intestinal epithelium.

In particular, targeted NPs with novel non-peptidic ligands for oral delivery have been

investigated. The main advantage of these particles is that their targeting properties

are improved by the use of ligands that are not degraded in the GI tract, unlike

peptidic/proteinic ligands, and are not limited to receptors, which interact only with

proteins. The use of targeted NPs will also depend on the type of cells/receptors

targeted by cell-specific targeting.

To the best of our knowledge, no clinical studies on the oral delivery of antigens or

drugs with targeted NPs are ongoing because until recently, the potential benefits of

these particles have been outweighed by the associated pitfalls. In particular, the

sophisticated design and associated high cost of the synthesis of the grafted polymer

and the high cost of NP production might be major limitations for their development

as pharmaceuticals. Due to their high cost of manufacture, targeted NPs must provide

significant added value for unmet pharmaceutical and medical needs. For example,

they will not be used for the solubilization of BCS class II or IV drugs; rather, their

use will likely be limited to punctual applications (e.g., vaccines) or the treatment of

diseases that are currently lacking satisfactory therapies. To a lesser extent, the lack of

control/robustness of NP absorption might also hinder their use in several biomedical

Page 25: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

24

applications. Thus, they should be used for applications that do not require a fine-

tuning of the delivered dose (e.g., vaccines and type 2 diabetes), and they would

likely not be suitable for insulin delivery for type 1 diabetes treatment.

Based on the literature review, we propose (as illustrated in Fig. 1) that the oral

delivery of NPs that are targeted using non-peptidic ligands would be useful to further

investigate i) oral vaccination targeting M cells; ii) the oral delivery of therapeutic

proteins and peptides by targeting enterocytes; and iii) the specific targeting of L cells

for the treatment of type 2 diabetes, obesity and inflammatory diseases.

5. References

[1] A. des Rieux, V. Fievez, M. Garinot, Y. J. Schneider, and V. Préat,

Nanoparticles as potential oral delivery systems of proteins and vaccines: a

mechanistic approach, J. Control Release, 116 (2006) 1-27.

[2] A. T. Florence, Nanoparticle uptake by the oral route: Fulfilling its potential?,

Drug Discov.Today: Technologies, 2 (2005) 75-81.

[3] E. A. McNeela and E. C. Lavelle, Recent Advances in Microparticle and

Nanoparticle Delivery Vehicles for Mucosal Vaccination

Mucosal Vaccines, in: P. A. Kozlowski (Ed.), Springer Berlin Heidelberg,

2012, pp. 75-99.

[4] R. J. Mrsny, Oral drug delivery research in Europe, J. Control Release, 161

(2012) 247-253.

[5] K. P. O'Donnell and R. O. Williams, Nanoparticulate systems for oral drug

delivery to the colon, Int. J. Nanotechn, 8 (2011) 4-20.

[6] L. Plapied, N. Duhem, A. des Rieux, and V. Préat, Fate of polymeric

nanocarriers for oral drug delivery, Current Opinion in Colloid Interface

Science, 16 (2011) 228-237.

[7] R. A. Petros and J. M. Desimone, Strategies in the design of nanoparticles for

therapeutic applications, Nat. Rev. Drug Discov., 9 (2010) 615-627.

[8] J. M. Lu, X. Wang, C. Marin-Muller, H. Wang, P. H. Lin, Q. Yao, and C.

Chen, Current advances in research and clinical applications of PLGA-based

nanotechnology, Expert. Rev. Mol. Diagn., 9 (2009) 325-341.

[9] M. Agueros, S. Espuelas, I. Esparza, P. Calleja, I. Penuelas, G. Ponchel, and J.

M. Irache, Cyclodextrin-poly(anhydride) nanoparticles as new vehicles for

oral drug delivery, Expert. Opin. Drug Deliv., 8 (2011) 721-734.

[10] M. Amidi, E. Mastrobattista, W. Jiskoot, and W. E. Hennink, Chitosan-based

delivery systems for protein therapeutics and antigens, Adv. Drug Deliv. Rev.,

62 (2010) 59-82.

[11] J. H. Park, G. Saravanakumar, K. Kim, and I. C. Kwon, Targeted delivery of

low molecular drugs using chitosan and its derivatives, Adv. Drug Deliv.

Rev., 62 (2010) 28-41.

[12] M. Werle, H. Takeuchi, and A. Bernkop-Schnurch, Modified chitosans for

oral drug delivery, J. Pharm. Sci., 98 (2009) 1643-1656.

Page 26: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

25

[13] L. M. Ensign, C. Schneider, J. S. Suk, R. Cone, and J. Hanes, Mucus

penetrating nanoparticles: biophysical tool and method of drug and gene

delivery, Adv. Mater., 24 (2012) 3887-3894.

[14] S. K. Lai, Y. Y. Wang, and J. Hanes, Mucus-penetrating nanoparticles for

drug and gene delivery to mucosal tissues, Adv. Drug Deliv. Rev., 61 (2009)

158-171.

[15] M. C. Chen, K. Sonaje, K. J. Chen, and H. W. Sung, A review of the prospects

for polymeric nanoparticle platforms in oral insulin delivery, Biomaterials, 32

(2011) 9826-9838.

[16] H. Hillaireau and P. Couvreur, Nanocarriers' entry into the cell: relevance to

drug delivery, Cell Mol. Life Sci., 66 (2009) 2873-2896.

[17] A. T. Florence, "Targeting" nanoparticles: The constraints of physical laws

and physical barriers, J. Control Release, 164 (2012) 115-124.

[18] B. Devriendt, B. G. De Geest, B. M. Goddeeris, and E. Cox, Crossing the

barrier: Targeting epithelial receptors for enhanced oral vaccine delivery, J.

Control Release, 160 (2012) 431-439.

[19] N. Desai, Challenges in development of nanoparticle-based therapeutics,

AAPS. J., 14 (2012) 282-295.

[20] B. Zhang and B. Yan, Analytical strategies for characterizing the surface

chemistry of nanoparticles, Anal. Bioanal. Chem, 396 (2010) 973-982.

[21] P. M. Valencia, M. H. Hanewich-Hollatz, W. Gao, F. Karim, R. Langer, R.

Karnik, and O. C. Farokhzad, Effects of ligands with different water

solubilities on self-assembly and properties of targeted nanoparticles,

Biomaterials, 32 (2011) 6226-6233.

[22] A. Bandyopadhyay, R. L. Fine, S. Demento, L. K. Bockenstedt, and T. M.

Fahmy, The impact of nanoparticle ligand density on dendritic-cell targeted

vaccines, Biomaterials, 32 (2011) 3094-3105.

[23] J. Park, T. Mattessich, S. M. Jay, A. Agawu, W. M. Saltzman, and T. M.

Fahmy, Enhancement of surface ligand display on PLGA nanoparticles with

amphiphilic ligand conjugates, J. Control Release, 156 (2011) 109-115.

[24] R. R. Sawant, R. M. Sawant, A. A. Kale, and V. P. Torchilin, The architecture

of ligand attachment to nanocarriers controls their specific interaction with

target cells, J. Drug Target, 16 (2008) 596-600.

[25] S. Wang and E. E. Dormidontova, Nanoparticle design optimization for

enhanced targeting: Monte Carlo simulations, Biomacromolecules, 11 (2010)

1785-1795.

[26] N. Brandhonneur, F. Chevanne, V. Vie, B. Frisch, R. Primault, M. F. Le

Potier, and C. P. Le, Specific and non-specific phagocytosis of ligand-grafted

PLGA microspheres by macrophages, Eur. J. Pharm. Sci., 36 (2009) 474-485.

[27] T. I. Croll, A. J. O'Connor, G. W. Stevens, and J. J. Cooper-White,

Controllable surface modification of poly(lactic-co-glycolic acid) (PLGA) by

hydrolysis or aminolysis I: physical, chemical, and theoretical aspects,

Biomacromolecules, 5 (2004) 463-473.

[28] H. Freichels, V. Pourcelle, C. S. Le Duff, J. Marchand-Brynaert, and C.

Jérôme, "Clip" and "click" chemistries combination: toward easy PEGylation

of degradable aliphatic polyesters, Macromol. Rapid Commun., 32 (2011)

616-621.

[29] H. Freichels, V. Pourcelle, R. Auzely-Velty, J. Marchand-Brynaert, and C.

Jérôme, Synthesis of poly(lactide-co-glycolide-co-epsilon-caprolactone)-graft-

Page 27: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

26

mannosylated poly(ethylene oxide) copolymers by combination of "clip" and

"click" chemistries, Biomacromolecules, 13 (2012) 760-768.

[30] H. H. Salman, C. Gamazo, P. C. de Smidt, G. Russell-Jones, and J. M. Irache,

Evaluation of bioadhesive capacity and immunoadjuvant properties of vitamin

B(12)-Gantrez nanoparticles, Pharm. Res., 25 (2008) 2859-2868.

[31] F. Danhier, A. L. Breton, and V. Préat, RGD-Based Strategies To Target

Alpha(v) Beta(3) Integrin in Cancer Therapy and Diagnosis, Mol. Pharm., 9

(2012) 2961-2973.

[32] M. Garinot, V. Fievez, V. Pourcelle, F. Stoffelbach, des Rieux A., L. Plapied,

I. Theate, H. Freichels, C. Jérôme, J. Marchand-Brynaert, Y. J. Schneider, and

V. Préat, PEGylated PLGA-based nanoparticles targeting M cells for oral

vaccination, J. Control Release, 120 (2007) 195-204.

[33] D. Heckmann, A. Meyer, B. Laufer, G. Zahn, R. Stragies, and H. Kessler,

Rational design of highly active and selective ligands for the alpha5beta1

integrin receptor, Chembiochem., 9 (2008) 1397-1407.

[34] C. B. Carlson, P. Mowery, R. M. Owen, E. C. Dykhuizen, and L. L. Kiessling,

Selective tumor cell targeting using low-affinity, multivalent interactions,

ACS Chem Biol., 2 (2007) 119-127.

[35] O. M. Merkel, O. Germershaus, C. K. Wada, P. J. Tarcha, T. Merdan, and T.

Kissel, Integrin alphaVbeta3 targeted gene delivery using RGD

peptidomimetic conjugates with copolymers of PEGylated poly(ethylene

imine), Bioconjug. Chem, 20 (2009) 1270-1280.

[36] D. Pan, J. L. Turner, and K. L. Wooley, Shell Cross-Linked Nanoparticles

Designed To Target Angiogenic Blood Vessels via avß3 ReceptorêÆLigand

Interactions, Macromolecules, 37 (2004) 7109-7115.

[37] P. M. Winter, A. H. Schmieder, S. D. Caruthers, J. L. Keene, H. Zhang, S. A.

Wickline, and G. M. Lanza, Minute dosages of alpha(nu)beta3-targeted

fumagillin nanoparticles impair Vx-2 tumor angiogenesis and development in

rabbits, FASEB J., 22 (2008) 2758-2767.

[38] X. B. Xiong, Y. Huang, W. L. Lu, X. Zhang, H. Zhang, T. Nagai, and Q.

Zhang, Enhanced intracellular delivery and improved antitumor efficacy of

doxorubicin by sterically stabilized liposomes modified with a synthetic RGD

mimetic, J. Control Release, 107 (2005) 262-275.

[39] V. Fievez, L. Plapied, des Rieux A., V. Pourcelle, H. Freichels, V. Wascotte,

M. L. Vanderhaeghen, C. Jérôme, A. Vanderplasschen, J. Marchand-Brynaert,

Y. J. Schneider, and V. Préat, Targeting nanoparticles to M cells with non-

peptidic ligands for oral vaccination, Eur. J. Pharm. Biopharm., 73 (2009) 16-

24.

[40] V. Rerat, G. Dive, A. A. Cordi, G. C. Tucker, R. Bareille, J. Amedee, L.

Bordenave, and J. Marchand-Brynaert, alphavbeta3 Integrin-targeting Arg-

Gly-Asp (RGD) peptidomimetics containing oligoethylene glycol (OEG)

spacers, J. Med. Chem, 52 (2009) 7029-7043.

[41] M. Rémy, R. Bareille, V. Rerat, C. Bourget, J. Marchand-Brynaert, and L.

Bordenave, Polyethylene terephthalate membrane grafted with

peptidomimetics: endothelial cell compatibility and retention under shear

stress, Journal of Biomaterials Science, Polymer Edition, (2012) 1-18.

[42] V. Pourcelle, H. Freichels, F. Stoffelbach, R. Auzely-Velty, C. Jérôme, and J.

Marchand-Brynaert, Light induced functionalization of PCL-PEG block

copolymers for the covalent immobilization of biomolecules,

Biomacromolecules, 10 (2009) 966-974.

Page 28: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

27

[43] V. Pourcelle, S. Devouge, M. Garinot, V. Préat, and J. Marchand-Brynaert,

PCL-PEG-based nanoparticles grafted with GRGDS peptide: preparation and

surface analysis by XPS, Biomacromolecules, 8 (2007) 3977-3983.

[44] M. Momtaz, V. Rerat, S. Gharbi, E. Gerard, V. Pourcelle, and J. Marchand-

Brynaert, A graftable LDV peptidomimetic: design, synthesis and application

to a blood filtration membrane, Bioorg. Med. Chem Lett., 18 (2008) 1084-

1090.

[45] J. M. Irache, H. H. Salman, C. Gamazo, and S. Espuelas, Mannose-targeted

systems for the delivery of therapeutics, Expert. Opin. Drug Deliv., 5 (2008)

703-724.

[46] H. Freichels, R. Jérôme, and C. Jérôme, Sugar-labeled and PEGylated

(bio)degradable polymers intended for targeted drug delivery systems,

Carbohydrate Polymers, 86 (2011) 1093-1106.

[47] K. Jain, P. Kesharwani, U. Gupta, and N. K. Jain, A review of glycosylated

carriers for drug delivery, Biomaterials, 33 (2012) 4166-4186.

[48] S. Slavin, J. Burns, D. M. Haddleton, and C. R. Becer, Synthesis of

glycopolymers via click reactions, Eur. Polym. J., 47 (2011) 435-446.

[49] J. Rieger, F. Stoffelbach, D. Cui, A. Imberty, E. Lameignere, J. L. Putaux, R.

Jérôme, C. Jérôme, and R. Auzely-Velty, Mannosylated poly(ethylene oxide)-

b-poly(epsilon-caprolactone) diblock copolymers: synthesis, characterization,

and interaction with a bacterial lectin, Biomacromolecules, 8 (2007) 2717-

2725.

[50] J. Rieger, H. Freichels, A. Imberty, J. L. Putaux, T. Delair, C. Jérôme, and R.

Auzely-Velty, Polyester nanoparticles presenting mannose residues: toward

the development of new vaccine delivery systems combining biodegradability

and targeting properties, Biomacromolecules, 10 (2009) 651-657.

[51] H. Freichels, R. Auzely-Velty, P. Lecomte, and C. Jérôme, Easy

functionalization of amphiphilic poly(ethylene oxide)-b-poly(e-caprolactone)

copolymer micelles with unprotected sugar: synthesis and recognition by

lectins, Polym. Chem., 3 (2012) 1436-1445.

[52] N. Xu, Wang R., Du F.S., and Li Z.C., Synthesis of amphiphilic biodegradable

glycocopolymers based on poly(-caprolactone) by ring-opening

polymerization and click chemistry, J. Polym. Sci. A Polym. Chem., 47 (2009)

3583-3594.

[53] Y. Nagasaki, K. Yasugi, Y. Yamamoto, A. Harada, and K. Kataoka, Sugar-

installed block copolymer micelles: their preparation and specific interaction

with lectin molecules, Biomacromolecules, 2 (2001) 1067-1070.

[54] S. Hamdy, A. Haddadi, A. Shayeganpour, J. Samuel, and A. Lavasanifar,

Activation of antigen-specific T cell-responses by mannan-decorated PLGA

nanoparticles, Pharm. Res., 28 (2011) 2288-2301.

[55] E. Jule, Y. Nagasaki, and K. Kataoka, Lactose-installed poly(ethylene glycol)-

poly(d,l-lactide) block copolymer micelles exhibit fast-rate binding and high

affinity toward a protein bed simulating a cell surface. A surface plasmon

resonance study, Bioconjug. Chem., 14 (2003) 177-186.

[56] F. Du, B. Zhang, H. Zhou, B. Yan, and L. Chen, Structure elucidation of

nanoparticle-bound organic molecules by 1H NMR, Trends Anal. Chem., 28

(2009) 88-95.

[57] Y. R. Du, S. Zhao, and L. F. Shen, Nuclear magnetic resonance studies of

micelles, Annual Reports on NMR Spectroscopy, Academic Press, 2002, pp.

145-194.

Page 29: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

28

[58] V. Pourcelle, C.S. Le Duff, H. Freichels, C. Jérôme, and J. Marchand-

Brynaert, Clickable PEG conjugate obtained by “clip photochemistry:

Synthesis and characterization by quantitative 19F NMR, J. Fluor.Chem., 140

(2012) 62-69.

[59] A. Azizi, A. Kumar, F. Diaz-Mitoma, and J. Mestecky, Enhancing oral

vaccine potency by targeting intestinal M cells, PLoS. Pathog., 6 (2010)

e1001147.

[60] D. J. Brayden, M. A. Jepson, and A. W. Baird, Keynote review: intestinal

Peyer's patch M cells and oral vaccine targeting, Drug Discov. Today, 10

(2005) 1145-1157.

[61] S. C. Corr, C. C. Gahan, and C. Hill, M-cells: origin, morphology and role in

mucosal immunity and microbial pathogenesis, FEMS Immunol. Med.

Microbiol., 52 (2008) 2-12.

[62] S. L. Demento, A. L. Siefert, A. Bandyopadhyay, F. A. Sharp, and T. M.

Fahmy, Pathogen-associated molecular patterns on biomaterials: a paradigm

for engineering new vaccines, Trends Biotechnol., 29 (2011) 294-306.

[63] A. Akalkotkar, S. A. Tawde, L. Chablani, and M. J. D'Souza, Oral delivery of

particulate prostate cancer vaccine: in vitro and in vivo evaluation, J. Drug

Target, 20 (2012) 338-346.

[64] V. Fievez, L. Plapied, C. Plaideau, D. Legendre, des Rieux A., V. Pourcelle,

H. Freichels, C. Jérôme, J. Marchand, V. Préat, and Y. J. Schneider, In vitro

identification of targeting ligands of human M cells by phage display, Int J.

Pharm., 394 (2010) 35-42.

[65] E. Gullberg and J. D. Soderholm, Peyer's patches and M cells as potential sites

of the inflammatory onset in Crohn's disease, Ann. N. Y. Acad. Sci., 1072

(2006) 218-232.

[66] P. N. Gupta, K. Khatri, A. K. Goyal, N. Mishra, and S. P. Vyas, M-cell

targeted biodegradable PLGA nanoparticles for oral immunization against

hepatitis B, J. Drug Target, 15 (2007) 701-713.

[67] P. N. Gupta and S. P. Vyas, Investigation of lectinized liposomes as M-cell

targeted carrier-adjuvant for mucosal immunization, Colloids Surf. B

Biointerfaces, 82 (2011) 118-125.

[68] S. Misumi, M. Masuyama, N. Takamune, D. Nakayama, R. Mitsumata, H.

Matsumoto, N. Urata, Y. Takahashi, A. Muneoka, T. Sukamoto, K. Fukuzaki,

and S. Shoji, Targeted delivery of immunogen to primate M cells with

tetragalloyl lysine dendrimer, J. Immunol., 182 (2009) 6061-6070.

[69] P. C. Tyrer, F. A. Ruth, J. M. Kyd, D. C. Otczyk, and A. W. Cripps, Receptor

mediated targeting of M-cells, Vaccine, 25 (2007) 3204-3209.

[70] C. Hamashin, L. Spindler, S. Russell, A. Schink, I. Lambkin, D. O'Mahony, R.

Houghten, and C. Pinilla, Identification of novel small-molecule Ulex

europaeus I mimetics for targeted drug delivery, Bioorg. Med. Chem, 11

(2003) 4991-4997.

[71] J. F. Pielage, C. Cichon, L. Greune, M. Hirashima, T. Kucharzik, and M. A.

Schmidt, Reversible differentiation of Caco-2 cells reveals galectin-9 as a

surface marker molecule for human follicle-associated epithelia and M cell-

like cells, Int J. Biochem. Cell Biol., 39 (2007) 1886-1901.

[72] I. Lambkin, C. Pinilla, C. Hamashin, L. Spindler, S. Russell, A. Schink, R.

Moya-Castro, G. Allicotti, L. Higgins, M. Smith, J. Dee, C. Wilson, R.

Houghten, and D. O'Mahony, Toward targeted oral vaccine delivery systems:

Page 30: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

29

selection of lectin mimetics from combinatorial libraries, Pharm. Res., 20

(2003) 1258-1266.

[73] S. S. Feng, L. Mei, P. Anitha, C. W. Gan, and W. Zhou, Poly(lactide)-vitamin

E derivative/montmorillonite nanoparticle formulations for the oral delivery of

Docetaxel, Biomaterials, 30 (2009) 3297-3306.

[74] E. Gullberg, M. Leonard, J. Karlsson, A. M. Hopkins, D. Brayden, A. W.

Baird, and P. Artursson, Expression of specific markers and particle transport

in a new human intestinal M-cell model, Biochem. Biophys. Res. Commun.,

279 (2000) 808-813.

[75] E. Gullberg, A. V. Keita, S. Y. Salim, M. Andersson, K. D. Caldwell, J. D.

Soderholm, and P. Artursson, Identification of cell adhesion molecules in the

human follicle-associated epithelium that improve nanoparticle uptake into the

Peyer's patches, J. Pharmacol. Exp. Ther., 319 (2006) 632-639.

[76] G. Gorfu, J. Rivera-Nieves, and K. Ley, Role of beta7 integrins in intestinal

lymphocyte homing and retention, Curr. Mol. Med., 9 (2009) 836-850.

[77] T. E. Rajapaksa, M. Stover-Hamer, X. Fernandez, H. A. Eckelhoefer, and D.

D. Lo, Claudin 4-targeted protein incorporated into PLGA nanoparticles can

mediate M cell targeted delivery, J. Control Release, 142 (2010) 196-205.

[78] T.E. Rajapaksa, M. Stover-Hamer, X. Fernandez, H.A. Eckelhoefer, D.D. Lo,

Claudin 4-targeted protein incorporated into PLGA nanoparticles can mediate

M cell targeted delivery, J Control Release, 142 (2010) 196–205

[79] K. Hase, K. Kawano, T. Nochi, G. S. Pontes, S. Fukuda, M. Ebisawa, K.

Kadokura, T. Tobe, Y. Fujimura, S. Kawano, A. Yabashi, S. Waguri, G.

Nakato, S. Kimura, T. Murakami, M. Iimura, K. Hamura, S. Fukuoka, A. W.

Lowe, K. Itoh, H. Kiyono, and H. Ohno, Uptake through glycoprotein 2 of

FimH(+) bacteria by M cells initiates mucosal immune response, Nature, 462

(2009) 226-230.

[80] H. Ohno and K. Hase, Glycoprotein 2 (GP2): grabbing the FimH bacteria into

M cells for mucosal immunity, Gut Microbes., 1 (2010) 407-410.

[81] G. Fotopoulos, A. Harari, P. Michetti, D. Trono, G. Pantaleo, and J. P.

Kraehenbuhl, Transepithelial transport of HIV-1 by M cells is receptor-

mediated, Proc. Natl. Acad. Sci. U. S. A, 99 (2002) 9410-9414.

[82] A. Helander, K. J. Silvey, N. J. Mantis, A. B. Hutchings, K. Chandran, W. T.

Lucas, M. L. Nibert, and M. R. Neutra, The viral sigma1 protein and

glycoconjugates containing alpha2-3-linked sialic acid are involved in type 1

reovirus adherence to M cell apical surfaces, J. Virol., 77 (2003) 7964-7977.

[83] A. B. Hutchings, A. Helander, K. J. Silvey, K. Chandran, W. T. Lucas, M. L.

Nibert, and M. R. Neutra, Secretory immunoglobulin A antibodies against the

sigma1 outer capsid protein of reovirus type 1 Lang prevent infection of

mouse Peyer's patches, J. Virol., 78 (2004) 947-957.

[84] W. Rubas, A. C. Banerjea, H. Gallati, P. P. Speiser, and W. K. Joklik,

Incorporation of the reovirus M cell attachment protein into small unilamellar

vesicles: incorporation efficiency and binding capability to L929 cells in vitro,

J. Microencapsul., 7 (1990) 385-395.

[85] H. M. Amerongen, R. Weltzin, C. M. Farnet, P. Michetti, W. A. Haseltine,

and M. R. Neutra, Transepithelial transport of HIV-1 by intestinal M cells: a

mechanism for transmission of AIDS, J. Acquir. Immune. Defic. Syndr., 4

(1991) 760-765.

[86] V. Snoeck, Van den Broeck W., V. De Colvenaer, F. Verdonck, B. Goddeeris,

and E. Cox, Transcytosis of F4 fimbriae by villous and dome epithelia in F4-

Page 31: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

30

receptor positive pigs supports importance of receptor-dependent endocytosis

in oral immunization strategies, Vet. Immunol. Immunopathol., 124 (2008)

29-40.

[87] P. Tiels, F. Verdonck, A. Coddens, B. Goddeeris, and E. Cox, The excretion

of F18+ E. coli is reduced after oral immunisation of pigs with a FedF and F4

fimbriae conjugate, Vaccine, 26 (2008) 2154-2163.

[88] Van den Broeck W., E. Cox, and B. M. Goddeeris, Receptor-dependent

immune responses in pigs after oral immunization with F4 fimbriae, Infect.

Immun., 67 (1999) 520-526.

[89] F. Verdonck, V. Snoeck, B. M. Goddeeris, and E. Cox, Cholera toxin

improves the F4(K88)-specific immune response following oral immunization

of pigs with recombinant FaeG, Vet. Immunol. Immunopathol., 103 (2005)

21-29.

[90] H. H. Salman, C. Gamazo, M. A. Campanero, and J. M. Irache, Salmonella-

like bioadhesive nanoparticles, J. Control Release, 106 (2005) 1-13.

[91] M. D. Neal, C. Leaphart, R. Levy, J. Prince, T. R. Billiar, S. Watkins, J. Li, S.

Cetin, H. Ford, A. Schreiber, and D. J. Hackam, Enterocyte TLR4 mediates

phagocytosis and translocation of bacteria across the intestinal barrier, J.

Immunol., 176 (2006) 3070-3079.

[92] Y. Liu, P. Wang, C. Sun, N. Feng, W. Zhou, Y. Yang, R. Tan, Z. Chen, S.

Wu, and J. Zhao, Wheat germ agglutinin-grafted lipid nanoparticles:

preparation and in vitro evaluation of the association with Caco-2 monolayers,

Int J. Pharm., 397 (2010) 155-163.

[93] N. Mishra, S. Tiwari, B. Vaidya, G. P. Agrawal, and S. P. Vyas, Lectin

anchored PLGA nanoparticles for oral mucosal immunization against hepatitis

B, J. Drug Target, 19 (2011) 67-78.

[94] Y. Yin, D. Chen, M. Qiao, X. Wei, and H. Hu, Lectin-conjugated PLGA

nanoparticles loaded with thymopentin: ex vivo bioadhesion and in vivo

biodistribution, J. Control Release, 123 (2007) 27-38.

[95] M. F. Francis, M. Cristea, and F. M. Winnik, Exploiting the vitamin B12

pathway to enhance oral drug delivery via polymeric micelles,

Biomacromolecules, 6 (2005) 2462-2467.

[96] H. H. Salman, C. Gamazo, M. Agueros, and J. M. Irache, Bioadhesive

capacity and immunoadjuvant properties of thiamine-coated nanoparticles,

Vaccine, 25 (2007) 8123-8132.

[97] S. K. Kang, J. H. Woo, M. K. Kim, S. S. Woo, J. H. Choi, H. G. Lee, N. K.

Lee, and Y. J. Choi, Identification of a peptide sequence that improves

transport of macromolecules across the intestinal mucosal barrier targeting

goblet cells, J. Biotechnol., 135 (2008) 210-216.

[98] Y. Jin, Y. Song, X. Zhu, D. Zhou, C. Chen, Z. Zhang, and Y. Huang, Goblet

cell-targeting nanoparticles for oral insulin delivery and the influence of

mucus on insulin transport, Biomaterials, 33 (2012) 1573-1582.

[99] G. Nikitas, C. Deschamps, O. Disson, T. Niault, P. Cossart, and M. Lecuit,

Transcytosis of Listeria monocytogenes across the intestinal barrier upon

specific targeting of goblet cell accessible E-cadherin, J. Exp. Med., 208

(2011) 2263-2277.

[100] M. D. Joshi, W. J. Unger, G. Storm, K. Y. van, and E. Mastrobattista,

Targeting tumor antigens to dendritic cells using particulate carriers, J.

Control Release, 161 (2012) 25-37.

Page 32: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

31

[101] B. Devriendt, B. G. De Geest, and E. Cox, Designing oral vaccines targeting

intestinal dendritic cells, Expert. Opin. Drug Deliv., 8 (2011) 467-483.

[102] D. Raghuwanshi, V. Mishra, K. Kaur, and M. R. Suresh, A simple approach

for enhanced immune response using engineered dendritic cell targeted

nanoparticles, Vaccine, (2012).

[103] B. Carrillo-Conde, E. H. Song, A. Chavez-Santoscoy, Y. Phanse, A. E.

Ramer-Tait, N. L. Pohl, M. J. Wannemuehler, B. H. Bellaire, and B.

Narasimhan, Mannose-functionalized "pathogen-like" polyanhydride

nanoparticles target C-type lectin receptors on dendritic cells, Mol. Pharm., 8

(2011) 1877-1886.

[104] Z. Ghotbi, A. Haddadi, S. Hamdy, R. W. Hung, J. Samuel, and A.

Lavasanifar, Active targeting of dendritic cells with mannan-decorated PLGA

nanoparticles, J. Drug Target, 19 (2011) 281-292.

[105] J. Holmgren and C. Czerkinsky, Mucosal immunity and vaccines, Nat. Med.,

11 (2005) S45-S53.

[106] R. Coco, L. Plapied, V. Pourcelle, C. Jérôme, D. J. Brayden, Y. J. Schneider,

and V. Préat, Drug delivery to inflamed colon by nanoparticles: Comparison

of different strategies, Int J. Pharm., (2012).

[107] F. Reimann, A. M. Habib, G. Tolhurst, H. E. Parker, G. J. Rogers, and F. M.

Gribble, Glucose sensing in L cells: a primary cell study, Cell Metab, 8 (2008)

532-539.

[108] S. Mojsov, G. Heinrich, I. B. Wilson, M. Ravazzola, L. Orci, and J. F.

Habener, Preproglucagon gene expression in pancreas and intestine diversifies

at the level of post-translational processing, J. Biol. Chem, 261 (1986) 11880-

11889.

[109] D. J. Drucker, The biology of incretin hormones, Cell Metab., 3 (2006) 153-

165.

[110] K. J. Rowland and P. L. Brubaker, The "cryptic" mechanism of action of

glucagon-like peptide-2, Am. J. Physiol Gastrointest. Liver Physiol, 301

(2011) G1-G8.

[111] A. J. Brown, S. M. Goldsworthy, A. A. Barnes, M. M. Eilert, L. Tcheang, D.

Daniels, A. I. Muir, M. J. Wigglesworth, I. Kinghorn, N. J. Fraser, N. B. Pike,

J. C. Strum, K. M. Steplewski, P. R. Murdock, J. C. Holder, F. H. Marshall, P.

G. Szekeres, S. Wilson, D. M. Ignar, S. M. Foord, A. Wise, and S. J. Dowell,

The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by

propionate and other short chain carboxylic acids, J. Biol. Chem., 278 (2003)

11312-11319.

[112] M. Thangaraju, G. A. Cresci, K. Liu, S. Ananth, J. P. Gnanaprakasam, D. D.

Browning, J. D. Mellinger, S. B. Smith, G. J. Digby, N. A. Lambert, P. D.

Prasad, and V. Ganapathy, GPR109A is a G-protein-coupled receptor for the

bacterial fermentation product butyrate and functions as a tumor suppressor in

colon, Cancer Res., 69 (2009) 2826-2832.

[113] S. Edfalk, P. Steneberg, and H. Edlund, Gpr40 is expressed in enteroendocrine

cells and mediates free fatty acid stimulation of incretin secretion, Diabetes,

57 (2008) 2280-2287.

[114] S. Karaki, H. Tazoe, H. Hayashi, H. Kashiwabara, K. Tooyama, Y. Suzuki,

and A. Kuwahara, Expression of the short-chain fatty acid receptor, GPR43, in

the human colon, J. Mol. Histol., 39 (2008) 135-142.

Page 33: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

32

[115] H. Tazoe, Y. Otomo, S. Karaki, I. Kato, Y. Fukami, M. Terasaki, and A.

Kuwahara, Expression of short-chain fatty acid receptor GPR41 in the human

colon, Biomed. Res., 30 (2009) 149-156.

[116] P. D. Cani, S. Possemiers, W. T. Van de, Y. Guiot, A. Everard, O. Rottier, L.

Geurts, D. Naslain, A. Neyrinck, D. M. Lambert, G. G. Muccioli, and N. M.

Delzenne, Changes in gut microbiota control inflammation in obese mice

through a mechanism involving GLP-2-driven improvement of gut

permeability, Gut, 58 (2009) 1091-1103.

[117] L. M. Lauffer, R. Iakoubov, and P. L. Brubaker, GPR119 is essential for

oleoylethanolamide-induced glucagon-like peptide-1 secretion from the

intestinal enteroendocrine L-cell, Diabetes, 58 (2009) 1058-1066.

[118] G. G. Muccioli, D. Naslain, F. Backhed, C. S. Reigstad, D. M. Lambert, N. M.

Delzenne, and P. D. Cani, The endocannabinoid system links gut microbiota

to adipogenesis, Mol. Syst. Biol., 6 (2010) 392.

[119] C. Thomas, A. Gioiello, L. Noriega, A. Strehle, J. Oury, G. Rizzo, A.

Macchiarulo, H. Yamamoto, C. Mataki, M. Pruzanski, R. Pellicciari, J.

Auwerx, and K. Schoonjans, TGR5-mediated bile acid sensing controls

glucose homeostasis, Cell Metab., 10 (2009) 167-177.

[120] J. H. Cummings, E. W. Pomare, W. J. Branch, C. P. Naylor, and G. T.

Macfarlane, Short chain fatty acids in human large intestine, portal, hepatic

and venous blood, Gut, 28 (1987) 1221-1227.

[121] Y. Kawamata, R. Fujii, M. Hosoya, M. Harada, H. Yoshida, M. Miwa, S.

Fukusumi, Y. Habata, T. Itoh, Y. Shintani, S. Hinuma, Y. Fujisawa, and M.

Fujino, A G protein-coupled receptor responsive to bile acids, J. Biol. Chem.,

278 (2003) 9435-9440.

[122] S. Katsuma, A. Hirasawa, and G. Tsujimoto, Bile acids promote glucagon-like

peptide-1 secretion through TGR5 in a murine enteroendocrine cell line STC-

1, Biochem. Biophys. Res. Commun., 329 (2005) 386-390.

[123] S. Cipriani, A. Mencarelli, M. G. Chini, E. Distrutti, B. Renga, G. Bifulco, F.

Baldelli, A. Donini, and S. Fiorucci, The bile acid receptor GPBAR-1 (TGR5)

modulates integrity of intestinal barrier and immune response to experimental

colitis, PLoS One, 6 (2011) e25637.

[124] S. Talukdar, J. M. Olefsky, and O. Osborn, Targeting GPR120 and other fatty

acid-sensing GPCRs ameliorates insulin resistance and inflammatory diseases,

Trends Pharmacol. Sci., 32 (2011) 543-550.

6. Figure legends

Figure 1. Schematic representation of polymeric nanoparticle targeting for oral

drug delivery as a function of cell type. Different pathways for the transport of

nanoparticles through enterocytes, M cells, goblet cells and L cells are represented by

orange arrows.

Page 34: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

33

Figure 2. Commonly used chemical principles for the conjugation of non-

peptidic ligands to (A) reactive polymers or (B) amphiphilic block copolymers. (C)

Chemical strategies for grafting ligands (or polymeric block-bearing ligands) along a

main polymer chain.

Figure 3. Tetragalloyl-D-lysine dendrimer (TGDK) targeting to M cells. (A) The

association of TGDK (red) with Rhesus Peyer’s patch follicle-associated epithelium.

(B) The penetration of TGDK (green) into Rhesus Peyer’s patch M cells (nuclear

staining by DAPI is shown in blue). (C) The transcytosis of TGDK through a Caco-

2/Raji B monolayer () and Caco-2 monolayer (o).

Figure 4. The influence of ligand grafting on nanoparticle transport across

monolayers of Caco-2 monoculture or Caco-2 and Raji B cell coculture,

mimicking the follicle-associated epithelium with M cells. Pegylated PLGA-based

nanoparticles of approximately 225 nm containing 15% PCL-PEG grafted to RGD, an

RGD peptidomimetic (RGDp), an LDV derivative (LDVd), an LDV peptidomimetic

(LDVp) or mannose (Man) were loaded with ovalbumin. Transport after (A) 90 min

of incubation at 37°C or (B) preincubation with (inhibitor +) or without (inhibitor -)

an anti-integrin 1 monoclonal antibody before adding nanoparticles for 90 min. The

number of particles transported is expressed as the apparent permeability coefficient

(Papp).

Figure 5. Schematic overview of the secretory function of L cells and the

receptors that can be targeted by nanoparticles with non-peptidic ligands.

Enteroendocrine L cells express a wide variety of GPCRs. The main endogenous and

physiological ligands of the different receptors are indicated at the top of the figure.

LCFA, long-chain fatty acid; SCFA: short-chain fatty acid; OEA:

oleoylethanolamide. Dietary lipids may be sensed by GPR40 and GRP120 expressed

on enteroendocrine cells. The gut microbiota contribute to the fermentation of various

non-digested compounds and produce specific metabolites, such as acetate,

propionate and butyrate, which have been described as specific ligands for GPR41, 43

and 109A receptors. Bioactive lipids, such as N-oleoylethanolamide, serve as ligands

of the GPR119 receptor. TGR5 (also known as M-BAR, GPBAR-1 or GPR131) is

Page 35: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

34

targeted by bile acids. Given the effects of gut peptides secreted by L cells (GLP-1,

GLP-2 and PYY), targeting L cells might help to treat obesity, type 2 diabetes and

inflammatory bowel diseases.

Page 36: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

35

Page 37: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

36

Figure 1

Page 38: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

37

Figure 2

Page 39: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

38

Figure 3

Page 40: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

39

Figure 4

Page 41: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

40

Figure 5

Page 42: Targeted nanoparticles with novel non-peptidic ligands for oral delivery

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPT

41

Graphical Abstract


Recommended