Sei sulla pagina 1di 13

Acta Biomaterialia 96 (2019) 55–67

Contents lists available at ScienceDirect

Acta Biomaterialia
journal homepage: www.elsevier.com/locate/actabiomat

Review article

Recent advances in polymer-based drug delivery systems for local


anesthetics
Bo Wang a,1, Shuo Wang b,1, Qi Zhang c, Yixuan Deng a, Xiang Li a, Liangyu Peng a, Xianghao Zuo d,
Meihua Piao e, Xin Kuang a,⇑, Shihou Sheng f,⇑, Yingjie Yu g,h,⇑
a
Department of Anesthesiology, The First Affiliated Hospital of the University of South China, Hengyang, Hunan 421001, China
b
Department of Pharmacy, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
c
Department of Chemical and Biomolecular Engineering, New York University, Brooklyn, NY 11201, United States
d
Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, NY 11794, United States
e
Department of Anesthesiology, The First Hospital of Jilin University, Changchun 130021, China
f
Department of Gastrointestinal Surgery, China–Japan Union Hospital of Jilin University, Changchun 130033, China
g
Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen 518039, China
h
Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA

a r t i c l e i n f o a b s t r a c t

Article history: Local anesthetics, which cause temporary loss of pain by inhibiting the transmission of nerve impulses,
Received 8 December 2018 have been widely used in clinical practice. However, neurotoxicity and short half-lives have significantly
Received in revised form 16 May 2019 limited their clinical applications. To overcome those barriers, numerous drug delivery systems (DDS)
Accepted 19 May 2019
have been designed to encapsulate local anesthetic agents, so that large doses can be released slowly
Available online 29 May 2019
and provide analgesia over a prolonged period. So far, multiple classes of local anesthetic carriers have
been investigated, with some of them already on the market. Among those, polymer-based delivery plat-
Keywords:
forms are the most extensively explored, especially in the form of polymeric nanoparticle carriers. This
Polymer
Drug delivery system
review gives a specific focus on the most commonly used natural and synthetic polymers for local anes-
Local anesthetics thetics delivery, owing to their excellent biocompatibility, biodegradability and versatility. State-of-the-
art studies concerning such polymer delivery systems have been discussed in depth. We also highlight
the impact of those delivery platforms as well as some key challenges that need to be overcome for their
broader clinical applications.

Statement of significance

Currently, local anesthetics have been widely used in clinically practices to prevent transmission of nerve
impulses. However, the applications of anesthetics are greatly limited due to their neurotoxicity and
short half-lives. Moreover, it is difficult to maintain frequent administrations which can cause poor com-
pliance and serious consequences. Numerous drug delivery systems have been developed to solve those
issues. In this review, we highlight the recent advances in polymer-based drug delivery systems for local
anesthetics. The advantages as well as shortcomings for different types of polymer-based drug delivery
systems are summarized in this paper. In the end, we also give prospects for future development of poly-
mer drug delivery systems for anesthetics.
Ó 2019 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

Abbreviations: LID, lidocaine; BUP, bupivacaine; BZC, benzocaine; LBP, levobupivacaine; ATC, articaine; HA, hyaluronic acid; CARR, carrageenan; CS, chitosan; AG, alginate;
GEL, gelatin; CEL, cellulose; CD, cyclodextrin; HPMC, hydroxypropyl methyl cellulose; PLA, polylactic acid; PEG, polyethylene glycol; PLGA, poly (lactic-co-glycolic acid); PCL,
polycaprolactone; PVA, poly (vinyl alcohol); LPSPs, lipid-protein-sugar particles; BVC LPNs, BUP-loaded lipid-polymer nanoparticles; AgNPs, silver nanoparticles; CHG, CS
glutamate; icLD, LID/multivalent ion complex; THB, tetrahydroxyborate; TAT, transcriptional transactivator peptide; LBL-LA/NLCs, Layer-by-layer-coated LID-loaded
nanostructured lipid nanoparticles; OQLCS, octadecyl-quaternized lysine modified CS.
⇑ Corresponding authors at: Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen 518039,
China (Y. Yu).
E-mail addresses: kxnhfy@126.com (X. Kuang), sheng89723711@163.com (S. Sheng), yuyingjie312@outlook.com (Y. Yu).
1
Authors contributed to the paper equally.

https://doi.org/10.1016/j.actbio.2019.05.044
1742-7061/Ó 2019 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
56 B. Wang et al. / Acta Biomaterialia 96 (2019) 55–67

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
2. Delivery systems developed for local anesthetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
3. Polymer-based delivery system for anesthetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
3.1. Natural polymer-based delivery systems for anesthetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
3.1.1. Hyaluronic acid. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
3.1.2. Chitosan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
3.1.3. Alginate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
3.1.4. Gelatin. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
3.1.5. Cellulose . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
3.1.6. Cyclodextrin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
3.2. Synthetic polymer-based delivery systems for anesthetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
3.2.1. Polylactic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
3.2.2. Poly (lactic-co-glycolic acid) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
3.2.3. Polycaprolactone. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
3.2.4. Polypeptide and peptide-drug conjugates. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
3.2.5. Poly (vinyl alcohol). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
3.3. Lipid and polymer complex delivery systems for anesthetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 62
3.4. Inorganic materials and polymer complex delivery systems for anesthetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
4. Conclusion and future perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
Acknowledgement. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65

1. Introduction value of plasma-drug concentration [15]. Till 2011, liposomal bupi-


vacaine (BUP) (ExparelÒ, Pacira Pharmaceuticals Inc., Parsippany,
Pain is a common symptom in clinical practice. Patients suffer- NJ, USA) was originally approved by food and Drug Administration
ing from pain reveal dysfunctional behaviors in both life and work (FDA) for use as a local anesthetic. Studies have shown that
[1,2]. Various factors, such as physical trauma, emotional trauma, ExparelÒ almost doubled the duration of action of sciatic never
societal and cultural pressures, can cause pain [3–6]. Although blockade as compare to bupicacaine alone [16]. It has been proved
the overall rate of perioperative mortality has declined signifi- that nanoparticles are an effective tool for postoperative pain relief
cantly in the past few decades, the anesthetic-related mortality with fewer side effects, suggesting a promising future of the DDS
rate in developing countries is still two times higher than that in for local anesthetics [17].
developed countries [7]. Hence, there is a growing demand in pre- To date, a number of different types of controlled released deliv-
venting and managing undesired pain from post-operative compli- ery systems have emerged. Among them, polymers are the most
cations. Currently, the main pharmacotherapeutic strategy that extensively explored materials for DDS owing to its excellent
aims to manage these complications involves the optimization of applicability [18–20]. In this review, the recent advances in
anesthesia delivery systems for patients undergoing surgical pro- polymer-based drug delivery systems for local anesthetics are dis-
cedures [1]. cussed in detail. The impact of them as well as some key challenges
Anesthetics provide several benefits including skeletal muscle that need to be overcome for clinical application have been
relaxation, lack of awareness, reduced anxiety, and suppression highlighted.
of undesirable reflexes [8]. They are mainly categorized into four
classes: preanesthetics, neuromuscular blockers, general anesthet-
ics and local anesthetics (Fig. 1) [9]. Clinically, local anesthetics can 2. Delivery systems developed for local anesthetics
be classified according to potency, speed of onset and duration of
anesthesia [10]. The chemical structure of clinically used local Current research on injectable or implantable local anesthetic
anesthetics contains three parts: a hydrophilic nitrogenous group, delivery systems aim to prolong anesthetic effects and reduce tox-
a lipophilic phenyl group, and an intermediate chain which can be icity. External delivery devices are inaccurate and limit the clinical
either an amide group or an ester group (Fig. 1) [11]. Despite all the use of new delivery systems. Hence, the development of a nanopar-
benefits in acute and chronic pain management, applying local ticle delivery system provides a safe and effective alternative in
anesthetics typically lead to sharp fluctuating levels of plasma- perioperative anesthesia treatment [11].
drug concentration. Consequently, it may cause patients to experi- The anesthetics most commonly studied are lidocaine (LID)
ence severe side effects [12]. In addition, frequent administrations and BUP [1,18,21]. Several local anesthetic delivery systems have
are difficult to maintain, which may result in poor compliance [13]. been developed using a variety of materials including lipid
Several topical anesthetics have been developed as commercial nanoparticles [22–26]), polymer-based nanoparticles (e.g., poly-
products to decrease the pain associated with superficial dermato- lactic acid (PLA) [27–30], poly (lactic-co-glycolic acid) (PLGA)
logic and laser procedures. However, a number of case reports indi- [31,32] or chitosan (CS) [33–35]), inorganic material based deliv-
cated that the non-FDA products were associated with adverse ery system (e.g., calcium [36,37] or silver [38]). In this review,
outcomes, due to inappropriately high anesthetic concentrations specific focus is given to polymer-based local anesthetic delivery
[14]. In order to address those challenges, researchers have devel- systems classified into four categories: natural polymer-based
oped versatile DDS that enable prolonged local anesthesia by delivery systems, synthetic polymer-based delivery systems,
incorporating anesthetics into biodegradable polymeric matrices, lipid/polymer complex delivery systems, and inorganic/polymer
from which drug diffusion is prolonged to maintain at an ideal complex delivery systems.
B. Wang et al. / Acta Biomaterialia 96 (2019) 55–67 57

3. Polymer-based delivery system for anesthetics


A Preanesthetic medications
Antacids Antiemetics Benzodiazepines
3.1. Natural polymer-based delivery systems for anesthetics
Anticholinergics Antihistamines Opioids
3.1.1. Hyaluronic acid
B General anesthetics: inhaled
Hyaluronic acid (HA) mainly exists in synovial fluid, heart
Desfl urane Halothane Isofl urane valves, vitreous of the eye, and extracellular matrix. It is a non-
F F F Cl Cl F
F
immunogenic naturally occurring mucopolysaccharide. Owing to
F F
F O
F F O F its excellent biocompatibility, biodegradability and viscoelastic
F Br
F F properties, HA has been widely used in various therapeutic appli-
Sevoflurane Nitrous oxide cations, including drug delivery, wound healing, tissue regenera-
F3C O F tion [39,40]. Furthermore, even if problems were to arise with
N N O N N O HA, it can be easily removed by digestion with hyaluronidase [41].
CF3 Particular attention has been focused on HA as a potential deliv-
ery for BUP. Diego et al. successfully conjugated BUP to HA deriva-
C General anesthetics: intravenous
tive Hylan B particles. In vitro drug release study showed the
Barbiturates Ketamine Opioids Etomidate duration of BUP prolonged to more than 16 h for HA-BUP particle,
HO
O
Cl O
which is significantly longer than that of free BUP (0.4 h). For
O
HN NH
O H in vivo study, compared with free BUP, HA-BUP exhibited five-
NH
O O H
N CH
3
N
fold longer block time in impairing motor function, thus proving
N
O HO this HA drug delivery system effectively prolonged the duration
Dexmedetomidine Propofol Benzodiazepines of local anesthesia [42]. HA was also combined with other compo-
R1
N
R
2
nents to make composite for biomedical application due to its
OH
NH
N
excellent compatibility. Ovidio et al. fabricated a multi-targeted
R7
N
R2' composite carrageenan (CARR)/HA based wafers loaded with LID
and silver nanoparticles (AgNPs). The AgNP loaded wafers effec-
tively killed all the bacteria tested after 6 h incubation. Further-
D Neuromuscular blockers
more, the mechanical hardness could be easily tuned by varying
Vecuroni um Pancuronium Rocuroni um the content of HA, providing good handling property for chronic
leg ulcer. This composite system exhibited multiple properties
O O
O O O O

N
H
H

H
N
N
H
H

H
N O
N
H N such as fast drug release and the effective antimicrobial activity,
Br-

indicating its potential application for chronic leg ulcer dressing


O H H
H O
H HO
O O H

(Fig. 2) [43].
Cisatracurium Succinylcholine
CH3 CH3
O O
CH3 H 3C O
2Cl-
H3C
O
N

O
O O

O
N
O
CH3
N
O
O
N
3.1.2. Chitosan
O
-
O-

O
O
O S O O S O
O
Chitosan (CS) is a natural polysaccharide used in biological
CH3 O
CH3 H 3C
O CH3
applications for decades. It can be fabricated into the form of pow-
ders, films, fiber meshes, membranes, beads, and hydrogels,
depending on their future application [44,45].
E Local anesthetics: amides
CS-based materials have been widely applied in oral mucosal
Bupivacaine Lidocaine Mepivacaine delivery systems due to its versatile property [46]. Several studies
H
N
N
H
N H related to CS loaded with LID have been carried out in buccal area.
N N
O
O
N Varshosaz et al. prepared films of LID with three different molecu-
O
lar weight of CS in various conditions. The result demonstrated
Ropivacaine Etidocaine Articaine
O that high concentration and molecular weight of CS significantly
S
H
N
H
N OCH3 increased the flux of LID through the films. However, this system
N N
O O NH has not been tested in vivo. Further clinical studies are necessary
O H
N to evaluate its clinical potential [47]. Pignatello et al. fabricated a
hydrogel for the buccal application of LID using CS glutamate
F Local anesthetics: esters (CHG). To evaluate its performance, CHG hydrogel were compared
Chloroprocaine Procaine Bezocaine with two commercial products. A much slower drug release rate
O O
O has been achieved with CHG hydrogel. In vivo pharmacological
N
O O
N
O activity study proved that CHG hydrogel has similar anesthetic
H 2N
H 2N Cl NH2 activity as commercial products, but with less tissue inflammation
Cocaine Tetracaine [48].
O
CH3 O
H 3C N O
N
O O

O N
H
3.1.3. Alginate
Alginates (AG) are water-soluble linear polysaccharides
Fig. 1. Classification of anesthetics. (A) Preanesthetic can be divided into six obtained from brown algae. It has been widely used in tissue engi-
categories. (B) and (C) represent general anesthetics delivered via inhalation and
intravenous injection. (D) Neuromuscular blockers facilitate tracheal intubation and
neering for pharmaceutical and biomedical applications as drug
surgery. (E) and (F) represent two most common chemical structures of local delivery systems due to its excellent biocompatibility and
anesthetics. biodegradability [49].
58 B. Wang et al. / Acta Biomaterialia 96 (2019) 55–67

LID release
100

LID release (% )
80

60
CARR
40 CARR/HA 10
CARR/HA 30
20 CARR/HA 50

0
0 1 2 3 4 5 6
Time (hours)

Antimicrobial activity

5.0 Control Wafer


CARR/AgNPs
4.0 CARR/AgNPs

Absorbance
CARR/AgNPs
CARR/AgNPs
3.0
Leg ulcer
2.0

1.0 * *
** **
Silver Nanoparticle ** ** ** ** ** **
Carrageenan 0.0 **

HA Lidocaine

Fig. 2. Scheme of composite CARR and HA dressings loaded with LID and AgNPs. Reprinted with permission from [43], copyrightÓ 2017 Polymer.

Propofol is a short-acting intravenously administered hypnotic encapsulated AG/CS nanoparticles [34]. Compared with single
agent which is used for induction of general anesthesia and proce- component system, AG/GS nanoparticles improved loading effi-
dural sedation. Commercial lipid emulsion of propofol (DiprivanÒ) ciency to 86%. Then, the nanoparticles were further applied to a
has certain drawbacks such as pain on injection and emulsion sciatic nerve blockade model. It has been demonstrated that a pro-
instability. In order to improve its stability and solubility, Najafa- longed duration of motor and sensory blockades was achieved with
badi et al. developed a facile method for preparation of (C8)- AG/GS nanoparticles in a sciatic nerve blockade model.
grafted alginate. The propofol encapsulated nanoparticles with
the size of less than 80 nm have been successfully prepared. This
nanoparticles exhibited great stability for more than 6 months. 3.1.4. Gelatin
The release profile of nanoparticle was similar to commercial pro- Gelatin (GEL) is derived from animal tissue consisting of min-
duct DiprivanÒ. Then sleep recovery test was applied on rats, the eral salts and proteins. It has been widely used in pharmaceutical
AG nanoparticle has similar anesthesia effect as DiprivanÒ. This and food industries [53]. Structurally, a typical GEL film incorpo-
study proved that AG nanoparticles served as a promising candi- rates a coil structure and triple-helixes (Fig. 3) [54].
date to replace DiprivanÒ. However, safety evaluation for AG The topical LID solution formulations prepared by using GEL,
nanoparticles should be the next step for its clinical application agar, and a food thickener as the delivery base, showed an excel-
[50]. lent analgesic effect against pain associated with needle insertion
Similar to CS, AGs are also incorporated with other materials, [55]. While GEL initially appeared an initial bursts issue. Specifi-
such as hydroxyapatite, to make multifunctional scaffold. Dubnika cally, a film system composed of sunflower oil and GEL could
et al. combined the hydroxyapatite with CS and AG to ensure LID release benzocaine (BZC) in a controlled manner, but with an initial
delivery up to 60 h [51]. The polymers are formed into gel by ionic burst [56]. Then, Huang et al. rectified this issue by dipping poly-
gelation method when mixed with cationic polymers (such as CS) lactic acid/polyethylene glycol (PLA/PEG) microspheres into a
or divalent cations (such as Ca2+) [52]. Grillo et al. developed BUP dilute GEL solution [57]. Consequently, a two-stage release system

Fig. 3. Scheme of GEL film structure. Left: amorphous coils; middle: triple helixes and coils; right: bundles of triple helixes and coils. Reprinted with permission from [54],
copyrightÓ 2012 Polymer.
B. Wang et al. / Acta Biomaterialia 96 (2019) 55–67 59

has been successfully developed to inhibit the initial burst release complexation revealed a significant enhancement of intensity
in drug release tests. and duration of anesthetic effect [71].
GEL as a composition of denatured collagens, frequently
changes its own structure during gelation, which substantially 3.2. Synthetic polymer-based delivery systems for anesthetics
affects the concentration, temperature and energy of GEL. Besides,
there are numerous hydrogen bonds on the side chains of amino 3.2.1. Polylactic acid
acids, which not only contribute to the formation of gelation, but PLA is a versatile material polymerized from lactic acid, it is
also increase the complexity of hydrophobic interactions between mainly used for biodegradable products such as plastic bags and
GEL and the carried drugs [58]. Therefore, more efforts should be planting cups [72].
made to produce a standardized DDS with GEL on a large scale. Firstly, PLA was used as the delivery system to develop BUP-
polyester microspheres to prolong percutaneous blockade of
3.1.5. Cellulose peripheral nerves [27]. Then, the key formulation variables that
Cellulose (CEL) is an extensively used biopolymer existed in liv- affected the release of BUP or BZC from different biodegradable
ing species, such as plants, animals, and bacteria. It has repeating drug delivery devices in the PLA solutions were investigated
units of ringed glucose molecules and a flat ribbon-like conforma- [28,29]. As a result, these nanocapsules exhibited completely dif-
tion. Furthermore, its abundant –OH side groups greatly facilitate ferent behaviors than those of the pure anesthetic in solution.
the grafting of chemical species to achieve different surface prop- Recently, LID-coated poly (L-lactide) (PLLA) microneedle arrays
erties, making it become an attractive materials for biomedical were fabricated based on micro-molding technique. A newly devel-
engineering [59,60]. oped dip-coating device enable LID to be coated only at the needle
Medhi et al. first utilized CEL to fabricate biopolymer for trans- tips and significantly reduced drug loss. LID coated on the arrays
dermal drug delivery. LID-loaded biopolymer composite micronee- was released rapidly into PBS within 2 min. Surprisingly, more effi-
dles from fish scale-nanocellulose were fabricated. This cient skin penetration was achieved with PLLA microneedle arrays
microneedles could pierce the stratum corneum and get dissolved compared to the commercial product EMLAÒ cream. This PLLA
in skin to release the loaded LID. A desirable drug loading capacity microneedle arrays could rapidly release LID in a painless manner,
has been achieved for LID [61]. In order to develop gel formulations which is beneficial for transdermal delivery (Fig. 4) [30]. In most
with suitable bioadhesion for transdermal drug delivery, the mepi- cases, instead of being used as a single formulation, PLA is often
vacaine gels contained vasoconstrictor and penetration enhancer copolymerized with PEG to form tunable micelles by changing
mixed with hydroxypropyl methyl CEL (HPMC) showed a pro- the ratio of PLA and PEG, thereby significantly enhancing the drug
longed local anesthetic action compared with control group [62]. incorporation efficiency [73,74].
Another possible application is based on the nasal administration.
A LID nasal gel was developed using HPMC as the base material to 3.2.2. Poly (lactic-co-glycolic acid)
transport LID directly from the nasal cavity into the central ner- PLGA has been among the most attractive polymers applied in
vous system [63]. In most cases, CEL is not the single component biomedical engineering for decades. It is approved by FDA for sev-
in delivery systems; researchers combined CEL with other poly- eral therapeutic applications due to its excellent biocompatibility,
mers like CS or AG. For example, a mucoadhesive bilayer device biodegradability and mechanical properties [75]. Hence, countless
made of CS and ethylcellulose showed promising potential in con- studies concerning application of PLGA in tissue engineering and
trolled delivery of anesthetics to the oral cavity [64]. drug delivery have been carried out.
Initially, Moraes et al. [76] prepared ropivacaine-loaded PLGA
3.1.6. Cyclodextrin nanospheres and found PLGA drug delivery system effectively
Cyclodextrins (CD) are cyclic oligosaccharides made of several reduced the toxicity of ropivacaine formulation. Then, they further
repeating dextrose units (a-, b, and c-CDs, respectively) joined applied this PLGA delivery system to encapsulate other anesthetics
through one to four bonds [65]. They have found applications in such as BUP and BZC, offering the possibility of prolonged anes-
food and pharmaceutical products for decades. thetic effect and reduced toxicity [31,77,78]. LID was also loaded
A plethora of research proved sustained-release of various kinds in PLGA microparticles and displayed sustainable delivery into
of local anesthetic encapsulated with CD. Although methyl and the cochlea, suggesting LID-PLGA microparticles could be used
ethyl thioether groups could be in the positions of a-, b-, and c- for the attenuation of peripheral tinnitus [79]. When using PLGA
CD, only the b-CD derivatives exhibit high affinity to anesthetic microparticles as a delivery system, the ‘‘microparticle mass/ bulk
drugs, like halothane and sevoflurane [66]. The complexing of fluid volume” ratio should be taken into account during in vitro
levobupivacaine (LBP) with maltosyl-b-CD was effective to use in drug release measurements [80].
intrathecal block and was able to extend the duration of anesthesia Combinational therapy has drawn tremendous attention owing
effects in a sciatic nerve block [67]. Moreover, other anesthetics to its synergistic effect. Zhang et al. developed an injectable PLGA
like LID or BUP can also be complexed with CD, both of which hydrogel/microsphere (GEL/MS) composite co-delivery system to
revealed to prolong local nerve block [68,69]. simultaneously encapsulate BUP and dexmedetomidine (DEX) for
The size of inner hydrophobic cavity depends on the number of synergistic analgesia. DEX served as a local anesthetic additive
glucopyranose units in CD. Hence, the loaded molecules can nei- drugs in this combinational therapy, which exhibited long-term
ther be too small, nor be too large [70]. At the same time, while vasoconstriction effect and improved the local anesthetic concen-
the inside of CD is hydrophobic, the outside is hydrophilic. Drug- tration at injection site. Neurobehavioral analyses showed that
in CD-in liposome system can be used as an excellent platform to the duration of sensory blockade in the GEL/(DEX-MS/BUP) group
improve solubility and stability of drug. It effectively resolved was 37 h, which is significantly longer than that of control groups.
the rapid release issue inherited from conventional single-use lipo- Pharmacokinetics and biodistribution study have been carried out
somes release system. In addition, it further improved drug avail- to evaluate the toxicity and the safety of this co-delivery system.
ability through skin rout by increasing drug solubility and Furthermore, the mechanism of DEX have been comprehensively
permeation across the skin [71]. One example of this application studied. The result indicated that the addition of DEX could greatly
is conducted by Maestrelli et al. who combined both BZC and reduce the amount of drug entering the bloodstream, therefore
butamben with hydroxypropyl-b-CD. The best solubility and disso- reducing the safety of the GEL/MS co-delivery system (Fig. 5). This
lution properties were achieved. When loaded in liposomes, the comprehensive study laid a solid foundation for its application.
60 B. Wang et al. / Acta Biomaterialia 96 (2019) 55–67

Fig. 4. Pristine PLA microneedle array (a) was observed by stereo microscopy. LID-coated PLA microneedle array was fabricated and observed by stereo microscopy (b) and
FE-SEM of 290.6 ± 45.9 mg LID was coated on the array (c). Reprinted with permission from [30], copyrightÓ 2017 Biomed. Microdevices.

Fig. 5. Schematic illustration of the preparation and in vivo nerve blockade effect of the Gel-microsphere system with BUP. Reprinted with permission from [82], copyrightÓ
2018 Biomaterials.

This system was then further applied for sustained released BUP. It in vivo performance of eugenol-loaded nanocapsule. The result
proves that this GEL/MS system is able to offer a precisely guided indicated that eugenol-loaded nanocapsule could effectively pre-
drug release and retention system, exhibiting great potential to vent septal bone resorption.
be used as an alternative for clinical pain management [81,82].
Therefore, the aforementioned studies proves that PLGA, as an 3.2.4. Polypeptide and peptide-drug conjugates
appealing biocompatible polymer, has broad application prospect Polypeptides are recombinant proteins genetically engineered
for anesthetic delivery. from cells. They are biodegradable and are made up of simple
amino acid residues [18,89]. Different amino acid sequences gener-
3.2.3. Polycaprolactone ate various structure, endowing the polypeptide with various spe-
Polycaprolactone (PCL) has been approved by the FDA as a cial properties [90,91].
suture material, a drug delivery device and an adhesion barrier Peptide-drug conjugates are prodrugs formed by covalent
[83]. It is an aliphatic polyester composed of repeating hexanoate attachment of the peptide and drug via a cleavable linker [92].
units, which is produced by ring opening polymerization of e- Among them, transcriptional transactivator peptide (TAT), as one
caprolactone [84]. of the most frequently used cell-penetrating peptides, has excel-
Silva de Melo et al. encapsulated articaine (ATC) with PEG-PCL lent ability to enhance the skin delivery of drugs. Wang et al. pre-
nanocapsules. This system was stable for up to 120 days of storage pared a LID loaded TAT-peptide-conjugated nanoparticle for
at ambient temperature. Satisfactory encapsulation efficiency was transdermal delivery. In vivo skin permeation studies were applied
achieved with values of around 60%. At the same time, the toxicity to evaluate the penetration depth in skin by monitoring the loca-
of this PEG-PCL nanocapsules was much less than that of free drug tion of fluorescent of calcein encapsulated in different vehicles.
group, indicating its clinical application for the carrier of ATC [85]. TAT-conjugated polymeric liposomes group achieved the best skin
In another study, PEG-PCL-PEG hydrogel has been applied to penetration ability, indicating its potential for transdermal formu-
encapsulate LID or LBP. It could produce longer anesthesia effects lation (Fig. 6) [93].
than that of the pure solution at the same dose [86,87]. Parmod In addition to the conventional used peptide, peptide with spe-
et al. applied eugenol loaded PCL in the treatment of periodontal cial property has been well explored recently. Leu-enkephalin
infections. Solvent displacement method was applied for the for- (LENK) has been regarded as a promising painkiller due to its high
mation of eugenol-loaded PCL nanocapsules [88]. Ligature- affinity toward d-opioid. Nevertheless, due to its pharmacokinetic
induced periodontitis model in rats was selected to evaluate issues, including plasma stability and blood-brain barrier perme-
B. Wang et al. / Acta Biomaterialia 96 (2019) 55–67 61

Fig. 6. Polypeptide or peptide-drug conjugates based local anesthetics delivery system. (A) Synthesis process of TAT-peptide-conjugated OQLCS and Schematic illustration of
the preparation of drug loaded TAT-liposomes. (B) In vivo mouse skin permeation of calcein encapsulated liposomes and calcein encapsulated TAT-conjugated liposomes.
Reprinted with permission from [93], copyrightÓ 2013 Int. J. Pharm.

ability, its clinical application has been restrained. Recently, Feng signal in the non-inflamed paw and brain area was obtained for
et al. designed a facile method to conjugate neuropeptide LENK LENK-SQ nanoparticles group, suggesting that LENK-SQ nanoparti-
to squalene, and then the corresponding (LENK-squalene) LENK- cles group can be easily delivered to inflamed area.
SQ were assembled to form nanoparticle. An animal model of
inflammatory hyperalgesia that mimics human clinical pain condi- 3.2.5. Poly (vinyl alcohol)
tions was applied to evaluate the antihyperalgesic properties of Poly (vinyl alcohol) (PVA) is one of the most widely used bio-
LENK-SQ nanoparticles [94]. The LENK-SQ nanoparticles exhibited compatible and water soluble polymers with rather low cost
an impressive antihyperalgesic effect that last twice as long as [95]. It has been widely used in drug delivery and tissue
morphine. Furthermore, biodistribution studies using in vivo fluo- engineering.
rescence imaging were applied to investigate the target delivery Effective topical anesthesia demand a special delivery formula
ability of LENK-SQ nanoparticles. When compared to a single which has sufficient viscous flow to enable it to exactly flow into
LENK-SQ molecular form, very low accumulation of fluorescence the wound, meanwhile exhibiting sufficient cohesive integrity to
62 B. Wang et al. / Acta Biomaterialia 96 (2019) 55–67

be removed in one piece. Most of hydrogels cannot meet such stan- Although lipid-based carriers have attracted biological proper-
dard. However PVA-tetrahydroxyborate (THB) hydrogel exhibits ties, they suffer lack of reliability and reproducibility during man-
this unique property due to its reversible nature of the cross- ufacture. In addition, the low weight-volume ratio decrease the
linking mechanism. Loughlin et al. successfully applied this PVA- payload that can be attached [105]. Hence, a strategy of lipid-
THB hydrogel system to load LID for drug delivery to exposed polymer hybrid nanoparticles (LPNs) combines the advantages of
epithelial surfaces [95]. Petrisor et al. demonstrated that PVA can biodegradable polymeric nanoparticles and biomimetic phospho-
effectively eliminate burst release effects and restrain rapid anes- lipids [106]. Researchers compared BUP-loaded LPNs (BVC LPNs)
thesia. When LID was loaded in PVA, the burst effect disappeared with BUP PLGA nanoparticles (BVC NPs) (Fig. 8). In vitro and
without the need to change the dosage of LID released [96]. McCar- in vivo studies illustrated that BVC LPNs had better anesthesia
ron et al. conducted a preliminary clinical trial to evaluate the per- effect and lower toxicity than free BUP nanoparticles, demonstrat-
formance of PVA and THB hydrogel as a means to anesthetize acute ing that the combination of both delivery systems was superior
lacerations prior to suturing [97]. Through a series of optimiza- than single delivery system [107].
tions, the PVA-THB system could resolve the problems of poor pro- Typically, anesthetics were first conjugated with polymers, then
duce flow with wound area and problematic removal due to the mixture was coated with liposomes. CD complexes are rapidly
hydrogel fracturing, indicating its promising clinical applicability metabolized into urine and induce toxicity. Therefore, a ternary
(Fig. 7) [98]. system consisting of proparacaine, CD, and liposome is used to
overcome these complications. Firstly, BZC and butamben were
3.3. Lipid and polymer complex delivery systems for anesthetics combined with CD. Then, they were loaded in liposomes [102]. Fer-
reira et al. found that drug released from this delivery system was
Lipids are of significant interest for drug delivery field thanks to sustained for a longer period of time without causing toxic effect
its aptitude to trap both hydrophilic and lipophilic drugs. Tremen- [108]. They further employed this delivery system with ropiva-
dous efforts have been devoted to reducing the drug toxicity and caine and observed that it caused less inflammatory response than
improving the targeting effect for lipid. In the field of topical anes- that of a plain liposomal delivery system.
thesia, DDS based on lipids are utilized for skin delivery, drug pro- However, in other studies, anesthetics were first covered by
tection, and controlled release [99–101]. Various strategies have lipid. Then the hybrids were coated by polymers. For example, a
been explored to increase drug release controllability, solubility, layer-by-layer technique was used in order to achieve a prolonged
and stability using multicomponent systems, such as CS-lipid and anesthetic effect and better bioavailability through transdermal
CD-liposome [26,102,103]. Kohane et al. prepared lipid-protein- administration. The LID was first loaded in core-shell nanostruc-
sugar particles (LPSPs) as BUP carriers [23,24,104]. LPSPs provided tured lipid nanoparticles. Afterwards, they were coated with CS
sensory blockade durations comparable to those from PLGA micro- and HA to achieve topical anesthesia. This polymer/lipid hybrid
spheres. Histological sections were applied to study tissue reaction nanoparticles combined the beneficial characteristics of both natu-
to both particles. They found out that tissue reaction to PLGA ral polymeric nanoparticles and liposomes, exhibiting better anes-
microspheres lasted considerably longer than that to LPSPs, thetic effect than their LID-liposomes counterparts and free drugs
demonstrating that LPSPs are more biocompatible since they are [26,109]. In addition to natural polymers, lipids can also be com-
composed of natural ingredients. bined with synthetic polymers. The BUP-loaded lipid-polymer

Fig. 7. Representative images of formulation F3 (10.0% PVA, 2.5% w/w THB) that displayed the most appropriate characteristics for clinical use by scoring highest in
qualitative assessments. (A) Wound prior to hydrogel application, (B) formulation F3 applied to the wound, and (C) removal of the formulation from the wound in one piece.
Reprinted with permission from [98], copyrightÓ 2011 Acad. Emerg. Med.

Fig. 8. Lipid/polymer complex delivery systems for local anesthetics. Scheme of the fabrication of LBL-LA/NLCs. Reproduced from P. Ma, T. Li, H. Xing, S. Wang, Y. Sun, X. Sheng,
K. Wang. Local anesthetic effects of bupivacaine loaded lipid-polymer hybrid nanoparticles: in vitro and in vivo evaluation. Biomed. Pharmacother. 2017; 89: 689–695. Copyright Ó
2017 Elsevier Masson SAS. All rights reserved.
B. Wang et al. / Acta Biomaterialia 96 (2019) 55–67 63

nanoparticles (BVC LPNs) with PLGA was fabricated as the core [110,111]. This icLD continuously released LID at a constant rate
[107]. In vitro and in vivo evaluation illustrated that BVC LPNs have for 24 h. In contrast, free LID showed a much faster initial burst
better anesthesia effect and lower toxicity than free BUP within the first 1 h (Fig. 9 (A)). For in vivo study, the duration of sci-
nanoparticles. atic nerve blockade in the icLD 100-mg group was up to 14 h,
which is dramatically longer than free LID group (Fig. 9B). An elec-
trophysiological evaluation was also applied to evaluate whether
3.4. Inorganic materials and polymer complex delivery systems for
the nerve signal would be blocked after application of drug solu-
anesthetics
tions. The result clearly indicated that the rats in icLD 100-mg
showed nerve blockade up to 20 h (Fig. 9C). In the end, neurotox-
Silver compounds have been widely used as antibacterial agents
icity was evaluated through immunocytochemical images, and
to prevent bacterial infections and was first applied in the hydrox-
the result showed that ATF3 expression was significantly reduced
yapatite structure. The porous hydroxyapatite/Ag scaffolds which
through encapsulation (Fig. 9D). Hence, both the vitro and in vivo
was coated with AG could control silver ion release by a wet pre-
study indicated that icLD offers a prolonged period of LID release,
cipitation method. These scaffolds could be used for bone tissue
and thus provided longer nerve blockade effect with less neurotox-
engineering, with both long-term local antibacteria (up to one
icity, proving its potential for clinical applications. More evaluation
year) and short-term anesthetic effects (LID release up to two
concerning the safety of this system is desirable before pushing its
weeks) [51]. Another AgNPs prepared by reducing silver ions with
application to the next stage [110].
glucose in alkaline medium showed stability over time through
adding tetraethyl orthosilicate and L-asparagine as stabilizers.
Nonetheless, AgNPs exhibited high sensitivity to the anesthetics 4. Conclusion and future perspective
(procaine, dibucaine, or tetracaine) [38].
Jang et al. designed a facile method to fabricate LID/multivalent This review sketches the essential role of polymer-based deliv-
ion complex (icLD) for sustained release via the gradual ionic ery systems for local anesthetics by highlighting state-of-the-art
exchange between multivalent ions and monovalent ions studies in the field. The specific features and applications of each

A B
14
120
Paw Withdrawal Latency (sec)

Normal
12 LD 10-mg
100
Cumulative Release (%)

icLD 50-mg
10 icLD 100-mg
80
8
60 6

40 4
LD
20 icLD 2

0
0 0 5 10 15 20 25
0 5 10 15 20 25
Time (hr) Time (hr)

C 0 hr 1 hr 5 hrs 10 hrs 20 hrs 30 hrs


D Hoechst ATF3 NeuN Merge

Normal Normal
5mV
10ms

LD LD
10-mg 10-mg

icLD icLD
50-mg 50-mg

icLD icLD
100-mg 100-mg

2 mm

Fig. 9. Inorganic materials and polymer complex delivery systems for anesthetics. (A) Patterns of cumulative release of LD from the free LD and the icLD powders. (B) Changes
in the latency of paw withdrawal at different time points after application of three drug solutions, * p < 0.05. (C) Electrophysiological recordings of compound muscle action
potentials conducted through the sciatic nerves in the normal and experimental groups at different time points. (D) Immunfluorescence images at 3 days after application of
the three drug solutions. Reprinted with permission from [110], copyrightÓ 2017 Eur. J. Pharm. Biopharm.
64 B. Wang et al. / Acta Biomaterialia 96 (2019) 55–67

kind of polymer-based DDS have been systematically summarized and inorganic nanoparticles. Therefore, both natural and synthetic
in Table 1. In general, natural polymers are superior in biodegrad- polymers exhibit their unique properties, providing various
ability, while synthetic polymers typically have a longer degrada- options for local anesthetics in need.
tion process in comparison, but possess more versatile Clinically used local anesthetics, such as LID and BUP, are
functionalities. In addition, the synthetic polymers can be easily known for their effectiveness. However, they also suffer from a
used in combination with other delivery systems such as lipids short duration of action (e.g., the effect of LID lasts only 1–2 h)

Table 1
Summary of polymer-based local anesthetic delivery systems.

Materials Anesthetics Comments Ref


HA BUP Free BUP (3 mg/kg) admixed with BUP (13 mg/kg) conjugated to Hylan B particles showed a four to 5-fold longer [42]
impairment of motor function over the free BUP formulations with a total block time of 19 h.
CARR-HA LID The LID release profiles for CARR and CARR/HA wafers showed controlled release over 6 h. The AgNPs/LID loaded wafers [43]
did not interfere with cell viability and growth.
CS LID A film prepared from 3% high MW CS and cross-linked by 0.1% of tripolyphosphate penta sodiu salt showed a high flux of [47]
LID, and relatively high bioadhesion and tensile strength.
LID Hydrogels for a controlled local release of LID using CHG with the addition of glycerin were produced. LID-loaded muco- [48]
adhesive hydrogels can be of aid in reducing the pain symptoms that characterize aphthosis and other mouth diseases.
tetracaine Albumin-CS microparticles loading tetracaine was found to significantly increase the duration of action of the drug up to [33]
4-fold.
AG-CS BUP The efficiency of association of BUP in AG/CS and AG/bis(2-ethylhexyl) sulfosuccinate nanoparticles was high, with [34]
values in excess of 75%. The release profile of BUP was modified when associated with the nanoparticles, being slower
and more sustained when compared with the kinetics of free BUP in both the cases.
PLGA-CS BUP A gel-based PLGA-CS-microparticles encapsulating BUP displayed sustained, tunable release of BUP up to 7 days. This [35]
formulation showed controlled release of local anesthetics to treat acute/subacute pain while avoiding enhanced
inflammation.
lipid-CS LID The anesthetic activity of LID-LPNs revealed a more rapid anesthetic effect in the first few minutes and also displayed [109]
sustained activity compared with the LID-liposomes. The sustained anesthetic effect of the LID-LPNs could bring about
better local anesthetic therapy effects than the LID-liposomes.
GEL BZC A film system composed of sunflower oil and GEL could release BZC in a controlled manner, but with an initial burst [56]
LID A LID nasal gel was developed using HPMC as the base material to transport LID directly from the nasal cavity into the [63]
central nervous system
mepivacaine Mepivacaine gel containing polyoxyethylene 2-oleyl ether and tetrahydrozoline produced a 2.36-fold increase in [62]
anesthetic activity compared to the control gel without any additives, suggesting that the bioadhesive mepivacaine-
HPMC gel containing permeation enhancer and vasoconstrictor could be developed for enhanced local anesthetic action.
CD LBP The complex of LBP with CD prolonged the anesthetic effect of LBP in both intrathecal and sciatic nerve blocks in rats. [67]
lipid-CD BZC When loaded in liposomes, the complexation of BZC and CD revealed a significant enhancement of intensity and [71]
duration of anesthetic effect
PLA BUP Developed an injectable local anesthetic preparation that provides 2–5 days blockade of the sciatic nerves of rat in vivo [27]
and plasma BUP levels are below the range associated with systemic toxicity.
BZC BZC-containing PLA nanocapsules have good chemical stability and colloidal over a 60-day period. the release profile of [29]
BZC in 73% PLA caused BZC water solubility to be increased six times.
LID The microneedle arrays were released rapidly into PBS within 2 min, and its storage stability lasted 3 weeks for varying [30]
temperatures. in vitro studies showed enhanced skin penetration and more efficient LID delivery into the skin compared
to EMLAÒ cream 1, 2, and 5 min after application.
PLA-PEG procaine PLA-PEG 30:5 nanoparticles enhance the incorporation efficiency of drug. [74]
PLGA BUP Sensory blockade duration for 50% (w/w) BUP was 840 min. BUP loaded-PLGA nanospheres increased cell viability, in [31,32]
comparison with the effect produced by free BUP, indicating a reduced toxicity.
LID Sensory blockade duration for 50% (w/w) LID was 255 min. LID and BUP-PLGA microspheres resulted in similar degrees [32]
of myotoxicity, irrespective of drug loading.
BZC BZC loaded PLGA nanocapsules released different amounts of drug after 1500 min in HEPES buffer, compared with free [77]
BZC
lipid-PLGA BUP Compared with BVC NPs, the release profile of BVC LPNs presented a low burst effect and kept sustained release for 96 h. [107]
BVC LPNs can reverse or reduce the cytotoxicity of free BVC at the same drug concentration.
PEG-PCL-PEG LID A LID-loaded PCEC hydrogel produced more enduring local antinociceptive effects compared with LID aqueous solution [86]
at the same dose.
LBP LBP in situ gels maintained good anesthesia effects even after 9 h of injection and rats’ stinging reaction maintained at a [87]
relatively low level
PEG-PCL ATC Suspensions of PEG-PCL nanocapsules loaded with ATC were moderately stable over a period of 120 days. Cytotoxicity [85]
assays confirmed that the encapsulation of ATC reduced its toxicity.
PVA-THB LID D-mannitol was effective in formulating LID into PVA-THB hydrogels. It can increase the solubility of LID and prevent the [95]
demixing effect seen in PVA-THB hydrogels brought about by LID concentrations that exceed 3.0% w/w. As temperature
increased through 37 to 50 °C, the solubility of LID was reduced progressively.
LID A formulation (10.0% PVA, 2.5% w/w THB) displayed the most appropriate characteristics in application and removal of [98]
adhesiveness. The release of LID was proportional to the concentration of LID incorporated.
LID PVA-THB hydrogels loaded with LID could be formulated without addition of a polyol modulator to a maximum [97]
concentration 1.5% w/w, and these hydrogels provided a sustained release over 24 h.
peptide-polymer LID The transdermal flux of LID-TAT-liposomes was approximately 4.17 and 1.75 times higher than that of LID solution and [93]
LID liposomes.
inorganic-polymer LID The icLD continuously released LID at a constant rate for 24 h with a mild initial burst. The duration of sciatic nerve [110]
blockade in the icLD 100-mg group was up to 14 h, which is longer than free LID group. the rats in icLD 100-mg showed
nerve blockade up to 20 h, providing longer anesthetic action.
B. Wang et al. / Acta Biomaterialia 96 (2019) 55–67 65

and safety issues, mainly in neurotoxicity and cardiotoxicity [112]. [16] J.B. McAlvin, R.F. Padera, S.A. Shankarappa, G. Reznor, A.H. Kwon, H.H. Chiang,
J. Yang, D.S. Kohane, Multivesicular liposomal bupivacaine at the sciatic
To overcome these barriers, polymeric DDS have been developed to
nerve, Biomaterials 35 (15) (2014) 4557–4564.
prolong duration time and reduce the side effects for local anes- [17] Y.C. Tong, A.D. Kaye, R.D. Urman, Liposomal bupivacaine and clinical
thetics [113]. Furthermore, synergistic analgesia could be achieved outcomes, Best Pract. Res. Clin. Anaesthesiol. 28 (1) (2014) 15–27.
with properly designed DDS that impart physicians with additional [18] A.F. Rogobete, M. Dragomirescu, O.H. Bedreag, D. Sandesc, C.A. Cradigati, M.
Sarandan, M. Papurica, S.E. Popovici, C. Vernic, G. Preda, New aspects of
clinical medication options. controlled release systems for local anaesthetics: a review, Trends Anaesth.
PLGA, PLA, and liposome/polymer are the most commonly used Crit. Care 9 (2016) 27–34.
DDS in local anesthetic delivery systems, whereas other materials [19] Q. Zhang, A. Prabhu, A. San, J.F. Al-Sharab, K. Levon, A polyaniline based
ultrasensitive potentiometric immunosensor for cardiac troponin complex
are still sparsely studied. For instance, gold nanoparticles have detection, Biosens. Bioelectron. 72 (2015) 100–106.
been utilized as DDS for decades and exhibited promising effects [20] D. Peer, J.M. Karp, S. Hong, O.C. Farokhzad, R. Margalit, R. Langer, Nanocarriers
in both photo-diagnostics and photothermal therapy [114–116]. as an emerging platform for cancer therapy, Nat. Nanotechnol. 2 (12) (2007)
751.
Moreover, researchers managed to promote the replacement of [21] V.V. Dinh, Y.S. Suh, H.K. Yang, Y.T. Lim, Spatiotemporal programing for the on-
painful injections by patient-friendly needle-free topical formula- demand release of bupivacaine based on an injectable composite hydrogel, J.
tions, which is advantageous and will have promising applications Pharm. Sci. 105 (12) (2016) 3634–3644.
[22] G.P. Joshi, G. Patou, V. Kharitonov, The safety of liposome bupivacaine
in local anesthetics [117]. following various routes of administration in animals, J. Pain Res. 8 (2015)
In summary, the latest advances of polymer nanocarriers have 781–789.
been comprehensively reviewed in this article. We detailed the [23] D.S. Kohane, M. Lipp, R.C. Kinney, N. Lotan, R. Langer, Sciatic nerve blockade
with lipid-protein-sugar particles containing bupivacaine, Pharm. Res. 17
arsenal of polymer nanocarriers for local anesthetics. Despite
(10) (2000) 1243–1249.
numerous studies on encapsulating anesthetics with polymer [24] D.S. Kohane, M. Lipp, R.C. Kinney, D.C. Anthony, D.N. Louis, N. Lotan, R. Langer,
nanocarriers are ongoing, few of them are projected to enter clin- Biocompatibility of lipid-protein-sugar particles containing bupivacaine in
ical stages. It remains a challenge to select an appropriate nanocar- the epineurium, J. Biomed. Mater. Res. 59 (3) (2002) 450–459.
[25] G. Colombo, R. Padera, R. Langer, D.S. Kohane, Prolonged duration local
rier since several factors can simultaneously affect delivery anesthesia with lipid-protein-sugar particles containing bupivacaine and
efficiency and circulation of nanoparticles. Therefore, more sys- dexamethasone, J. Biomed. Mater. Res., Part A 75 (2) (2005) 458–464.
tematic studies are desired to eventually generate clinically useful [26] L. Zhang, J. Wang, H. Chi, S. Wang, Local anesthetic lidocaine delivery system:
chitosan and hyaluronic acid-modified layer-by-layer lipid nanoparticles,
nanocarriers for local anesthetics delivery. Drug Deliv. 23 (9) (2016) 3529–3537.
[27] J. Curley, J. Castillo, J. Hotz, M. Uezono, S. Hernandez, J.O. Lim, J. Tigner, M.
Chasin, R. Langer, C. Berde, Prolonged regional nerve blockade. Injectable
Acknowledgement biodegradable bupivacaine/polyester microspheres, Anesthesiology 84 (6)
(1996) 1401–1410.
[28] H. Kranz, R. Bodmeier, Structure formation and characterization of injectable
This work was supported by Hunan Provincial and Municipal drug loaded biodegradable devices: in situ implants versus in situ
Joint Funds (NO. 2017JJ4049). microparticles, Eur. J. Pharm. Sci. 34 (2–3) (2008) 164–172.
[29] N.F.S. de Melo, R. Grillo, A.H. Rosa, L.F. Fraceto, N.L. Dias, E. de Paula, D.R. de
Araujo, Development and characterization of poli (L-lactide) nanocapsules
References containing benzocaine, Quim. Nova 33 (1) (2010) 65–69.
[30] S.H. Baek, J.H. Shin, Y.C. Kim, Drug-coated microneedles for rapid and painless
local anesthesia, Biomed. Microdevices 19 (1) (2017).
[1] P. Bhusal, J. Harrison, M. Sharma, D.S. Jones, A.G. Hill, D. Svirskis, Controlled
[31] C.M. Moraes, R. de Lima, A.H. Rosa, E. de Paula, L.F. Fraceto, Encapsulation of
release drug delivery systems to improve post-operative pharmacotherapy,
local anesthetic bupivacaine in biodegradable Poly(DL-lactide-co-glycolide)
Drug Deliv. Transl. Res. 6 (5) (2016) 441–451.
nanospheres: factorial design, characterization and cytotoxicity studies,
[2] R.J. Alencar de Castro, P.C. Leal, R.K. Sakata, Pain management in burn
Macromol. Symp. 281 (2009) 106–112.
patients, Braz. J. Anesthesiol. 63 (1) (2013) 149–158.
[32] J.B. Mcalvin, G. Reznor, S.A. Shankarappa, C.F. Stefanescu, D.S. Kohane, Local
[3] Y. Olsen, J.M. Sharfstein, Chronic pain, addiction, and Zohydro, N. Engl. J. Med.
toxicity from local anesthetic polymeric microparticles, Anesth. Analg. 116
370 (22) (2014) 2061–2063.
(4) (2013) 794–803.
[4] D. Dowell, T.M. Haegerich, R. Chou, CDC guideline for prescribing opioids for
[33] R.T. Addo, A. Siddig, R. Siwale, N.J. Patel, J. Akande, A.N. Uddin, M.J. D’Souza,
chronic pain–United States, 2016, J. Am. Med. Assoc. 315 (15) (2016) 1624–
Formulation, characterization and testing of tetracaine hydrochloride-loaded
1645.
albumin-chitosan microparticles for ocular drug delivery, J. Microencapsul.
[5] J. Gierthmühlen, R. Baron, Neuropathic pain, Semi. Neurol. 36 (5) (2016) 462–
27 (2) (2010) 95–104.
468.
[34] R. Grillo, N.F.S. de Melo, D.R. de Araujo, E. de Paula, A.H. Rosa, L.F. Fraceto,
[6] D.B. Gordon, O.A. de Leon-Casasola, C.L. Wu, K.A. Sluka, T.J. Brennan, R. Chou,
Polymeric alginate nanoparticles containing the local anesthetic bupivacaine,
Research gaps in practice guidelines for acute postoperative pain
J. Drug Target. 18 (9) (2010) 688–699.
management in adults: findings from a review of the evidence for an
[35] F. Taraballi, S. Minardi, B. Corradetti, I.K. Yazdi, M.A. Balliano, J.L. Van Eps, M.
american pain society clinical practice guideline, J. Pain 17 (2) (2016) 158–
Allegri, E. Tasciotti, Potential avoidance of adverse analgesic effects using a
166.
biologically ‘‘smart” hydrogel capable of controlled bupivacaine release, J.
[7] D. Bainbridge, J. Martin, M. Arango, D. Cheng, Perioperative and anaesthetic-
Pharm. Sci. 103 (11) (2014) 3724–3732.
related mortality in developed and developing countries: a systematic review
[36] H. Gautier, V. Chamblain, P. Weiss, C. Merle, J.M. Bouler, In vitro
and meta-analysis, Lancet 380 (9847) (2012) 1075–1081.
characterisation of calcium phosphate biomaterials loaded with lidocaine
[8] G. Kannan, S.P. Kambhampati, S.R. Kudchadkar, Effect of anesthetics on
hydrochloride and morphine hydrochloride, J. Mater. Sci.: Mater. Med. 21
microglial activation and nanoparticle uptake: implications for drug delivery
(12) (2010) 3141–3150.
in traumatic brain injury, J. Controll. Release 263 (2017) 192–199.
[37] E. Verron, O. Gauthier, P. Janvier, H.L. Guen, D. Holopherne, R. Cavagna, J.M.
[9] K. Whaken, Lippincott’s Illustrated Reviews: Pharmacology, Lippincott,
Bouler, Analgesic properties of calcium phosphate apatite loaded with
Williams & Wilkins, Philadelphia, 2014.
bupivacaine on postoperative pain, J. Biomed. Mater. Res., Part B 94B (1)
[10] K. Welin-Berger, J.A. Neelissen, J. Engblom, Physicochemical interaction of
(2010) 89–96.
local anesthetics with lipid model systems-correlation with in vitro
[38] A. Mocanu, R.D. Pasca, G. Tomoaia, C. Garbo, P.T. Frangopol, O. Horovitz, M.
permeation and in vivo efficacy, J. Controll. Release 81 (1–2) (2002) 33–43.
Tomoaiacotisel, New procedure to synthesize silver nanoparticles and their
[11] L. Zorzetto, P. Brambilla, E. Marcello, N. Bloise, M.D. Gregori, L. Cobianchi, A.
interaction with local anesthetics, Int. J. Nanomed. 8 (1) (2013) 3867–3874.
Peloso, M. Allegri, L. Visai, P. Petrini, From micro- to nanostructured
[39] E. Gilbert, A. Hui, S. Meehan, H.A. Waldorf, The basic science of dermal fillers:
implantable device for local anesthetic delivery, Int. J. Nanomed. 11 (2016)
past and present Part II: adverse effects, J. Drugs Dermatol. 11 (9) (2012)
2695–2709.
1069–1077.
[12] A. Swain, D.S. Nag, S. Sahu, D.P. Samaddar, Adjuvants to local anesthetics:
[40] J. Liang, D. Jiang, P.W. Noble, Hyaluronan as a therapeutic target in human
current understanding and future trends, World J. Clin. Cases 5 (8) (2017)
diseases, Adv. Drug Deliv. Rev. 97 (2016) 186–203.
307–323.
[41] R.J. Hirsch, H.J. Brody, J.D. Carruthers, Hyaluronidase in the office: a necessity
[13] F.F. Tu, Insights gained from a negative trial of steroid blocks for perineal
for every dermasurgeon that injects hyaluronic acid, J. Cosmet. Laser Ther. 9
pain, Br. J. Obstet. Gynaecol. 124 (2) (2016). 261-261.
(3) (2007) 182–185.
[14] J.F. Sobanko, C.J. Miller, T.S. Alster, Topical anesthetics for dermatologic
[42] D.A. Gianolio, M. Philbrook, L.Z. Avila, H. Macgregor, S.X. Duan, R. Bernasconi,
procedures: a review, Dermatol. Surg. 38 (5) (2012) 709–721.
M. Slavsky, S. Dethlefsen, P.K. Jarrett, R.J. Miller, Synthesis and evaluation of
[15] S.J. Holland, B.J. Tighe, P.L. Gould, Polymers for biodegradable medical
hydrolyzable hyaluronan-tethered bupivacaine delivery systems,
devices. 1. The potential of polyesters as controlled macromolecular release
Bioconjugate Chem. 16 (6) (2005) 1512–1518.
systems, J. Control. Release 4 (3) (1986) 155–180.
66 B. Wang et al. / Acta Biomaterialia 96 (2019) 55–67

[43] O. Catanzano, R. Docking, P. Schofield, J. Boateng, Advanced multi-targeted [74] T. Govender, T. Riley, T. Ehtezazi, M.C. Garnett, S. Stolnik, L. Illum, S.S. Davis,
composite biomaterial dressing for pain and infection control in chronic leg Defining the drug incorporation properties of PLA-PEG nanoparticles, Int. J.
ulcers, Carbohydr. Polym. 172 (2017) 40–48. Pharm. 199 (1) (2000) 95–110.
[44] A. Ali, S. Ahmed, A review on chitosan and its nanocomposites in drug [75] H.K. Makadia, S.J. Siegel, Poly lactic-co-glycolic acid (PLGA) as
delivery, Int. J. Biol. Macromol. 109 (2018) 273–286. biodegradable controlled drug delivery carrier, Polymers 3 (3) (2011)
[45] M. Dash, F. Chiellini, R.M. Ottenbrite, E. Chiellini, Chitosan-a versatile semi- 1377–1397.
synthetic polymer in biomedical applications, Prog. Polym. Sci. 36 (8) (2011) [76] C.M. Moraes, A.P. de Matos, R. de Lima, A.H. Rosa, E. de Paula, L.F. Fraceto,
981–1014. Initial development and characterization of PLGA nanospheres containing
[46] S. Harloff-Helleberg, L.H. Nielsen, H.M. Nielsen, Animal models for evaluation ropivacaine, J. Biol. Phys. 33 (5–6) (2007) 455–461.
of oral delivery of biopharmaceuticals, J. Control. Release 268 (2017) 57–71. [77] C.M. Moraes, A.P. de Matos, E. de Paula, A.H. Rosa, L.F. Fraceto, Benzocaine
[47] J. Varshosaz, S. Karimzadeh, Development of cross-linked chitosan films for loaded biodegradable poly-(D, L-lactide-co-glycolide) nanocapsules: factorial
oral mucosal delivery of lidocaine, Res. Pharm. Sci. 2 (1) (2008) 43–52. design and characterization, Mater. Sci. Eng. B-Adv. Funct. Solid-State Mater.
[48] R. Pignatello, L. Basile, G. Puglisi, Chitosan glutamate hydrogels with local 165 (3) (2009) 243–246.
anesthetic activity for buccal application, Drug Deliv. 16 (3) (2009) 176–181. [78] C. Ning, Y. Guo, L. Yan, J.P. Thawani, W. Zhang, C. Fu, T. Liu, J. Ding, On-
[49] K.I. Draget, G. Skjåkbraek, O. Smidsrød, Alginate based new materials, Int. J. Demand prolongation of peripheral nerve blockade through bupivacaine-
Biol. Macromol. 21 (1–2) (1997) 47–55. loaded hydrogels with suitable residence periods, ACS Biomater. Sci. Eng. 5
[50] A.H. Najafabadi, S. Azodi-Deilami, M. Abdouss, H. Payravand, S. Farzaneh, (2) (2018) 696–709.
Synthesis and evaluation of hydroponically alginate nanoparticles as novel [79] R.T. Horie, T. Sakamoto, T. Nakagawa, Y. Tabata, N. Okamura, N. Tomiyama, M.
carrier for intravenous delivery of propofol, J. Mater. Sci.: Mater. Med. 26 (3) Tachibana, J. Ito, Sustained delivery of lidocaine into the cochlea using poly
(2015) 1–11. lactic/glycolic acid microparticles, Laryngoscope 120 (2) (2010) 377–383.
[51] A. Dubnika, D. Loca, V. Rudovica, M.B. Parekh, L. Berzina-Cimdina, [80] D. Klose, F. Siepmann, J.F. Willart, M. Descamps, J. Siepmann, Drug release
Functionalized silver doped hydroxyapatite scaffolds for controlled from PLGA-based microparticles: effects of the ‘‘microparticle:bulk fluid”
simultaneous silver ion and drug delivery, Ceram. Int. 43 (4) (2017) 3698– ratio, Int. J. Pharm. 383 (1–2) (2010) 123–131.
3705. [81] W. Zhang, C. Ning, W. Xu, H. Hu, M. Li, G. Zhao, J. Ding, X. Chen, Precision-
[52] A. Doustgani, E.V. Farahani, M. Imani, A.H. Doulabi, Dexamethasone sodium guided long-acting analgesia by Gel-immobilized bupivacaine-loaded
phosphate release from chitosan nanoparticles prepared by ionic gelation microsphere, Theranostics 8 (12) (2018) 3331–3347.
method, J. Colloid Sci. Biotechnol. 1 (1) (2012) 42–50. [82] W. Zhang, W. Xu, C. Ning, M. Li, G. Zhao, W. Jiang, J. Ding, X. Chen, Long-acting
[53] A. Duconseille, T. Astruc, N. Quintana, F. Meersman, V. Sante-Lhoutellier, hydrogel/microsphere composite sequentially releases dexmedetomidine
Gelatin structure and composition linked to hard capsule dissolution: a and bupivacaine for prolonged synergistic analgesia, Biomaterials 181
review, Food Hydrocolloids 43 (43) (2015) 360–376. (2018) 378–391.
[54] M. Coppola, M. Djabourov, M. Ferrand, Unified phase diagram of gelatin films [83] R.M. Mohamed, K. Yusoh, A review on the recent research of polycaprolactone
plasticized by hydrogen bonded liquids, Polymer 53 (7) (2012) 1483–1493. (PCL), Adv. Mater. Res. 1134 (2016) 249–255.
[55] T. Ando, Y. Shimoo, M. Nakasato, H. Yoshida, Development and clinical [84] M. Labet, W. Thielemans, Synthesis of polycaprolactone: a review, Chem. Soc.
evaluation of new topical anesthetic formulations for dental care, Biol. Pharm. Rev. 38 (12) (2009) 3484–3504.
Bull. 39 (3) (2016) 423–427. [85] N.F.S. de Melo, E.V.R. Campos, C.M. Goncalves, E. de Paula, T. Pasquoto, R. de
[56] J. Mhando, A.L.W. Po, Two-stage release of benzocaine from sunflower oil/ Lima, A.H. Rosa, L.F. Fraceto, Development of hydrophilic nanocarriers for the
gelatin emulsion films, Int. J. Pharm. 59 (2) (1990) 165–170. charged form of the local anesthetic articaine, Colloids Surf B 121 (2014) 66–
[57] Y.Y. Huang, T.W. Chung, T.W. Tzeng, A method using biodegradable 73.
polylactides/polyethylene glycol for drug release with reduced initial burst, [86] C.Y. Gong, S.A. Shi, L. Wu, M.L. Gou, Q.Q. Yin, Q.F. Guo, P.W. Dong, F. Zhang, F.
Int. J. Pharm. 182 (1) (1999) 93–100. Luo, X. Zhao, Y.Q. Wei, Z.Y. Qian, Biodegradable in situ gel-forming controlled
[58] A. Duconseille, T. Astruc, N. Quintana, F. Meersman, V. Sante-Lhoutellier, drug delivery system based on thermosensitive PCL-PEG-PCL hydrogel. Part
Gelatin structure and composition linked to hard capsule dissolution: a 2: sol-gel-sol transition and drug delivery behavior, Acta Biomater. 5 (9)
review, Food Hydrocolloids 43 (2015) 360–376. (2009) 3358–3370.
[59] R.J. Moon, A. Martini, J. Nairn, J. Simonsen, J. Youngblood, Cellulose [87] Y.J. Zhao, L.C. Zhou, J. Liu, Z.G. Chen, L. Yang, H. Shi, Preparation and
nanomaterials review: structure, properties and nanocomposites, Chem. investigation of a novel levobupivacaine in situ implant gel for prolonged
Soc. Rev. 40 (7) (2011) 3941–3994. local anesthetics, Artif. Cell. Nanomed. Biotechnol. 45 (3) (2017) 404–408.
[60] X. Wang, Q. Zhang, C. Nam, M. Hickner, M. Mahoney, M.E. Meyerhoff, An [88] K. Pramod, M.R. Aji Alex, M. Singh, S. Dang, S.H. Ansari, J. Ali, Eugenol
ionophore-based anion-selective optode printed on cellulose paper, Angew. nanocapsule for enhanced therapeutic activity against periodontal infections,
Chem. 56 (39) (2017) 11826–11830. J. Drug Target. 24 (1) (2016) 24–33.
[61] P. Medhi, O. Olatunji, A. Nayak, C.T. Uppuluri, R.T. Olsson, B.N. Nalluri, D.B. [89] X. Yang, Y. Yu, X. Huang, Q. Chen, Y. Qiu, Delivery of platinum (II) drugs with
Das, Lidocaine-loaded fish scale-nanocellulose biopolymer composite bulky ligands in trans-geometry for overcoming cisplatin drug resistance,
microneedles, AAPS PharmSciTech 18 (5) (2017) 1488–1494. Mater. Sci. Eng. B 96 (2018).
[62] C.W. Cho, J.S. Choi, S.C. Shin, Enhanced local anesthetic action of mepivacaine [90] R. Price, A. Poursaid, H. Ghandehari, Controlled release from recombinant
from the bioadhesive gels, Pak. J. Pharm. Sci. 24 (1) (2011) 87–93. polymers, J. Control. Release 190 (2014) 304–313.
[63] K.L. Hu, N. Mei, L. Feng, X.G. Jiang, Hydrophilic nasal gel of lidocaine [91] J. Li, Y. Yu, K. Myungwoong, K. Li, J. Mikhail, L. Zhang, C.-C. Chang, D.
hydrochloride. 1st communication: preparation, formulation optimization Gersappe, M. Simon, C. Ober, Manipulation of cell adhesion and dynamics
and in vitro release study, Arzneim. Forsch. 59 (11) (2009) 543–549. using RGD functionalized polymers, J. Mater. Chem. B 5 (31) (2017) 6307–
[64] C. Remuñánlópez, A. Portero, J.L. Vilajato, M.J. Alonso, Design and evaluation 6316.
of chitosan/ethylcellulose mucoadhesive bilayered devices for buccal drug [92] Y. Wang, A.G. Cheetham, G. Angacian, H. Su, L. Xie, H. Cui, Peptide-drug
delivery, J. Control. Release 55 (2–3) (1998) 143–152. conjugates as effective prodrug strategies for targeted delivery, Adv. Drug
[65] V.J. Stella, Q. He, Cyclodextrins, Toxicol. Pathol. 36 (1) (2008) 30–42. Deliv. Rev. 110 (2017) 112–126.
[66] L.F. Becker, D.H. Schwarz, G. Wenz, Synthesis of uniform cyclodextrin [93] Y. Wang, W. Su, Q. Li, C. Li, H. Wang, Y. Li, Y. Cao, J. Chang, L. Zhang,
thioethers to transport hydrophobic drugs, Beilstein J. Org. Chem. 10 (2014) Preparation and evaluation of lidocaine hydrochloride-loaded TAT-
2920–2927. conjugated polymeric liposomes for transdermal delivery, Int. J. Pharm. 441
[67] K. Karashima, M. Taniguchi, T. Nakamura, M. Takasaki, K. Matsuo, M. Irikura, (1–2) (2013) 748–756.
T. Irie, Prolongation of intrathecal and sciatic nerve blocks using a complex of [94] J. Feng, S. Lepetre-Mouelhi, A. Gautier, S. Mura, C. Cailleau, F. Coudore, M.
levobupivacaine with maltosyl-beta-cyclodextrin in rats, Anesth. Analg. 104 Hamon, P. Couvreur, A new painkiller nanomedicine to bypass the blood-
(5) (2007) 1121–1128. brain barrier and the use of morphine, Sci. Adv. 5 (2) (2019) eaau5148.
[68] R. Suzuki, Y.C.P. Arai, K. Hamayasu, K. Fujita, K. Hara, T. Yamaguchi, S. [95] R.G. Loughlin, M.M. Tunney, R.F. Donnelly, D.F. Murphy, M. Jenkins, P.A.
Sasaguri, Complex of branched cyclodextrin and lidocaine prolonged the McCarron, Modulation of gel formation and drug-release characteristics of
duration of peripheral nerve block, J. Anesth. 23 (2) (2009) 295–297. lidocaine-loaded poly(vinyl alcohol)-tetraborate hydrogel systems using
[69] M. Jug, F. Maestrelli, M. Bragagni, P. Mura, Preparation and solid-state scavenger polyol sugars, Eur. J. Pharm. Biopharm. 69 (3) (2008) 1135–1146.
characterization of bupivacaine hydrochloride cyclodextrin complexes aimed [96] G. Petrisor, R.M. Ion, C.H. Brachais, J.P. Couvercelle, O. Chambin, Designing
for buccal delivery, J. Pharm. Biomed. Anal. 52 (1) (2010) 9–18. medical devices based on silicon polymeric material with controlled release
[70] L.F. Fraceto, R. Grillo, E. Sobarzo-Sánchez, Cyclodextrin inclusion complexes of local anesthetics, J. Macromol. Sci., Part A 49 (5) (2012) 439–444.
loaded in particles as drug carrier systems, Curr. Top. Med. Chem. 14 (4) [97] D.H. Abdelkader, M.A. Osman, S.A. El-Gizawy, A.M. Faheem, P.A. McCarron,
(2014) 518–525. Characterisation and in vitro stability of low-dose, lidocaine-loaded poly
[71] F. Maestrelli, M. González-Rodríguez, A. Rabasco, C. Ghelardini, P. Mura, New (vinyl alcohol)-tetrahydroxyborate hydrogels, Int. J. Pharm. 500 (1–2) (2016)
‘‘drug-in cyclodextrin-in deformable liposomes” formulations to improve the 326–335.
therapeutic efficacy of local anaesthetics, Int. J. Pharm. 395 (1–2) (2010) 222– [98] P.A. McCarron, D.J. Murphy, C. Little, J. McDonald, O.J. Kelly, M.G. Jenkins,
231. Preliminary clinical assessment of polyvinyl alcohol–tetrahydroxyborate
[72] K. Oksman, M. Skrifvars, J.F. Selin, Natural fibres as reinforcement in hydrogels as potential topical formulations for local anesthesia of
polylactic acid (PLA) composites, Compos. Sci. Technol. 63 (9) (2003) 1317– lacerations, Acad. Emerg. Med. 18 (4) (2011) 333–339.
1324. [99] S. Jain, N. Patel, M.K. Shah, P. Khatri, N. Vora, Recent advances in lipid-based
[73] A. Basu, K.R. Kunduru, S. Doppalapudi, A.J. Domb, W. Khan, Poly(lactic acid) vesicles and particulate carriers for topical and transdermal application, J.
based hydrogels, Adv. Drug Delivery Rev. 107 (2016) 192–205. Pharm. Sci. 106 (2) (2017) 423–445.
B. Wang et al. / Acta Biomaterialia 96 (2019) 55–67 67

[100] P.J. You, R. Yuan, C.Y. Chen, Design and evaluation of lidocaine- and [109] J.G. Wang, L.Z. Zhang, H.M. Chi, S.L. Wang, An alternative choice of lidocaine-
prilocaine-coloaded nanoparticulate drug delivery systems for topical loaded liposomes: lidocaine-loaded lipid-polymer hybrid nanoparticles for
anesthetic analgesic therapy: a comparison between solid lipid local anesthetic therapy, Drug Deliv. 23 (4) (2016) 1254–1260.
nanoparticles and nanostructured lipid carriers, Drug Des. Dev. Ther. 11 [110] Y.J. Jang, J.H. Lee, T.B. Seo, S.H. Oh, Lidocaine/multivalent ion complex as a
(2017) 2743–2752. potential strategy for prolonged local anesthesia, Eur. J. Pharm. Biopharm.
[101] C. Weldon, T. Ji, M.-T. Nguyen, A. Rwei, W. Wang, Y. Hao, C. Zhao, M. Mehta, B. 115 (2017) 113–121.
Y. Wang, J. Tsui, Nanoscale bupivacaine formulations to enhance the duration [111] S.Y. Na, S.H. Oh, T.H. Kim, J.A. Yoon, I.S. Lee, J.H. Lee, Sustained release of
and safety of intravenous regional anesthesia, ACS Nano 13 (1) (2018) 18–25. antibiotic complexed by multivalent ion: in vitro and in vivo study for the
[102] L.F. Cabeça, S.A. Fernandes, P.E. De, A.J. Marsaioli, Topology of a ternary treatment of peritonitis, Int. J. Pharm. 476 (1–2) (2014) 213–222.
complex (proparacaine-b-cyclodextrin-liposome) by STD NMR, Magn. Reson. [112] A. Paavola, J. Yliruusi, Y. Kajimoto, E. Kalso, T. Wahlström, P. Rosenberg,
Chem. 46 (9) (2008) 832–837. Controlled release of lidocaine from injectable gels and efficacy in rat sciatic
[103] Y. Li, T. Yang, Y. Yu, N. Shi, L. Yang, Z. Glass, J. Bolinger, I.J. Finkel, W. Li, Q. Xu, nerve block, Pharm. Res. 12 (12) (1995) 1997–2002.
Combinatorial library of chalcogen-containing lipidoids for intracellular [113] R. Qi, Y. Wang, P.M. Bruno, H. Xiao, Y. Yingjie, T. Li, S. Lauffer, W. Wei, Q. Chen,
delivery of genome-editing proteins, Biomaterials 178 (2018) 652–662. X. Kang, Nanoparticle conjugates of a highly potent toxin enhance safety and
[104] G. Colombo, R. Padera, R. Langer, D.S. Kohane, Prolonged duration local circumvent platinum resistance in ovarian cancer, Nat. Commun. 8 (1) (2017)
anesthesia with lipid-protein-sugar particles containing bupivacaine and 2166.
dexamethasone, J. Biomed. Mater. Res., Part A 75A (2) (2010) 458–464. [114] E. Boisselier, Gold nanoparticles in nanomedicine: preparations, imaging,
[105] R. Cohen, H. Kanaan, G.J. Grant, Y. Barenholz, Prolonged analgesia from diagnostics, therapies and toxicity, Chem. Soc. Rev. 38 (6) (2009) 1759–1782.
Bupisome and Bupigel formulations: from design and fabrication to [115] S. Wang, L. Shang, L. Li, Y. Yu, C. Chi, K. Wang, J. Zhang, R. Shi, H. Shen, G.I.
improved stability, J. Control. Release 160 (2) (2012) 346–352. Waterhouse, Metal-organic-framework-derived mesoporous carbon
[106] B. Mandal, H. Bhattacharjee, N. Mittal, H. Sah, P. Balabathula, L.A. Thoma, G.C. nanospheres containing porphyrin-like metal centers for conformal
Wood, Core-shell-type lipid-polymer hybrid nanoparticles as a drug delivery phototherapy, Adv. Mater. 28 (38) (2016) 8379–8387.
platform, nanomedicine: nanotechnol, Biol. Med. 9 (4) (2013) 474–491. [116] Y. Yu, Q. Xu, S. He, H. Xiong, Q. Zhang, W. Xu, V. Ricotta, L. Bai, Q. Zhang, Z. Yu,
[107] P. Ma, T. Li, H. Xing, S. Wang, Y. Sun, X. Sheng, K. Wang, Local anesthetic Recent advances in delivery of photosensitive metal-based drugs, Coord.
effects of bupivacaine loaded lipid-polymer hybrid nanoparticles: in vitro Chem. Rev. 387 (2019) 154–179.
and in vivo evaluation, Biomed. Pharmacother. 89 (2017) 689–695. [117] R.O. do Couto, C. Cubayachi, P.L. Calefi, R.V.F. Lopez, V. Pedrazzi, C.M. De
[108] L.E. Ferreira, B.V. Muniz, J. Burga-Sánchez, M.C. Volpato, P.E. De, E.A. Rosa, F.C. Gaitani, O. De Freitas, Combining amino amide salts in mucoadhesive films
Groppo, The effect of two drug delivery systems in ropivacaine cytotoxicity enhances needle-free buccal anesthesia in adults, J. Control. Release 266
and cytokine release by human keratinocytes and fibroblasts, J. Pharm. (2017) 205.
Pharmacol. 69 (2) (2017) 161–171.

Potrebbero piacerti anche