Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Biomaterials 299 (2023) 122129

Contents lists available at ScienceDirect

Biomaterials
journal homepage: www.elsevier.com/locate/biomaterials

Sustained release of levobupivacaine from temperature-sensitive injectable


hydrogel for long-term local anesthesia in postoperative pain management
YuJun Zhang a, c, 1, Kun Shi b, 1, Xi Yang b, Wen Chen b, TianHong Wang a, c, Yi Kang a, c,
DeYing Gong a, c, ZhiYong Qian b, **, WenSheng Zhang a, c, *
a
Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
b
Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
c
Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China
Hospital, Sichuan University, 610041, China

A R T I C L E I N F O A B S T R A C T

Keywords: Postoperative pain is a major concern for most of the surgical patients, and an inadequate postoperative pain
Injectable thermo-sensitive hydrogel control may cause a series of complications. With an effective pain control and lesser side effects, local anes­
pH adjustment thetics are preferred for use in postoperative pain management. However, the action duration of current local
Long-acting local anesthetic
anesthetics is too short to meet the requirements of postoperative analgesia. In this study, an injectable levo­
Levobupivacaine
Postoperative analgesia
bupivacaine (LB)-loaded thermo-sensitive hydrogel system based on biodegradable poly(D,L-lactide)-poly
(ethylene glycol)-poly(D,L-lactide) (PLEL) was developed for long-acting local anesthetic, in which the soluble
charged cation form of LB (LB HCl) was partly alkalified to the poorly soluble base form (LB base). This hybrid LB
loaded PLEL system (hLB/PLEL) is a free flowable liquid at room temperature and changes into a semi-solid
hydrogel once injection in response to the physiological temperature. Then, the dissolved LB HCl could
release firstly from the hydrogel contributing to a quick work, and the insoluble LB base dissolved and released
gradually as the decrease of the pH during the biodegradation of PLEL hydrogel, resulting in a long-term LB
release in local. The drug release behavior, pharmacokinetic, and biocompatibility of the thermo-sensitive hLB/
PLEL were studied in vitro and in vivo. The anesthetic effects of hLB/PLEL system were evaluated in the rat models
of sciatic nerve block, subcutaneous infiltration anesthesia and postoperative pain as well. This hLB/PLEL system
generated a significantly prolonged analgesic effect in rat models, which produced approximately 7 times longer
duration than 0.75% LB HCl and effectively relieved the spontaneous pain for 3 days. In general, the presented
hLB/PLEL system can not only achieve a fast-acting but also sustainably release LB to block the nerve and
significantly extend the effect of local analgesia, which means a promising candidate for long-acting post­
operative pain management.

1. Introduction local anesthetics [3]. In recent years, multimodal analgesic strategies for
enhanced recovery after surgery have focused on reducing the use of
More than 80% of applicable patients experience have postoperative opioid agents to avoid opioid-related adverse effects such as drug
pain, and 75% of which has moderate to extreme pain scores [1]. addiction, gastrointestinal distress, sedation, ileus, pruritus, and respi­
Inadequate postoperative pain control may cause a series of complica­ ratory depression [3–5]. Furthermore, an important warning published
tions, such as prolonged hospital stay, chronic pain, and drug addiction by the US Food and Drug Administration indicated that opioid abuse was
[2]. The major drug categories for postoperative pain treatment are the most pressing public health crisis [6]. It is generally accepted that
opioid analgesics, nonsteroidal anti-inflammatory drugs (NSAIDs), and NSAIDs are not necessary for postoperative pain control because of their

Abbreviations: LB, Levobupivacaine; PLEL, poly(D,L-lactide)-poly(ethylene glycol)-poly(D,L-lactide); hLB/PLEL, hybrid LB loaded PLEL system.
* Corresponding author. Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of
Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, China.
** Corresponding author. Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China.
E-mail addresses: zhiyongqian@scu.edu.cn (Z. Qian), zhang_ws@scu.edu.cn (W. Zhang).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.biomaterials.2023.122129
Received 18 November 2022; Received in revised form 10 April 2023; Accepted 20 April 2023
Available online 1 May 2023
0142-9612/© 2023 Elsevier Ltd. All rights reserved.
Y. Zhang et al. Biomaterials 299 (2023) 122129

analgesic ceiling effect as well as a series of severe side effects, including For long-term local anesthesia in postoperative pain management,
gastrointestinal toxicities, cardiovascular risks, renal injuries and hep­ how to effectively prolong the action duration of local anesthetic re­
atotoxicity [7–11]. Thus, with effective pain control and fewer side ef­ mains a challenge currently. Several studies have reported that the
fects, local anesthetics are preferred for postoperative pain alkalization of local anesthetic benefited to extend the duration of action
management. The most serious degree of postoperative pain was within [28,29]. However, unexpected soft tissue necrosis at the injection site
72 h after surgery, which did not exceed 7 days [1]. However, the effects induced by the precipitate of the base form of local anesthetic was
of currently available local anesthetics do not exceed 12 h after subcu­ observed, which limited the application of this strategy [30,31]. To
taneous infiltration [12]. The development of a long-acting local anes­ solve this problem, drug delivery systems were tried to encapsulate the
thetic for postoperative pain management is urgently needed [13]. base form of local anesthetic. Although some of them have reported to
Local anesthetics-induced analgesic effect usually achieved through prolong the analgetic duration and avoid the risks of local toxicity to
reversible blockade of neuronal sodium channels to prevent the gener­ some extent, the effects are still far from satisfactory [25,32,33]. Thus,
ation and the conduction of nerve impulses [14]. The investigation developing a novel local anesthetic-based formulation for long-acting
strategies for a novel long-acting local anesthetic primarily includes the postoperative pain control should be urgently addressed.
invention of new chemical compounds and the development of Levobupivacaine (LB), the S(− )-enantiomer of bupivacaine, is a
slow-release drug delivery systems. A few new chemical compounds, common used long-acting local anesthetic for infiltration, nerve block,
such as QX-314 [15], QX-OH [16], Neosaxitoxin [17], and EN3427 [18] ophthalmic, epidural and intrathecal anesthesia. What’s more, levobu­
have been shown to produce long-acting local anesthesia in animal pivacaine has been proven to produce a greater length of action and a
models. Unfortunately, one of the most promising new chemical com­ lower risk of cardiotoxicity comparison to bupivacaine, thus attracted
pounds with the longest duration of action, EN3427, have been proven more and more attention [34]. Similar to the other local anesthetics,
to produce analgesia for only 24 h after a single injection. Slow-release levobupivacaine normally exist in two forms in solution, specifically
drug delivery systems developed by encapsulating a local anesthetic via presents as cycles through a rapid equilibrium between a charged
a carrier hold the potential to provide a prolonged period of analgesia cationic form (good solubility in water) and uncharged base form (base
effect and to overcome the short duration of local anesthetic effect [19]. form, poorly soluble in water). In this study, we proposed an injectable
Specifically, a variety of formulations, including microparticles [20], hybrid LB-loaded hydrogel (hLB/PLEL) system based on an biodegrad­
nanoparticles [21], liposomes [22], cyclodextrins [23] hydrogels [24] able thermosensitive hydrogel, in which the soluble charged cation form
and composite systems [25], have all been investigated to develop a (LB HCl) and insoluble base form (LB base) co-existed through adjusting
long-acting local anesthetic drug products. Typically, Exparel™ (Pacira the initial pH of the PLEL hydrogel system (Scheme 1). The thermo­
Pharmaceuticals, USA) is a liposomal encapsulation of 13.3 mg/mL of sensitive hydrogel was made of poly(D,L-lactide)-poly(ethylene glyco­
bupivacaine arranged in a microscopic honeycomb-like structure and is l)-poly(D,L-lactide) (PLEL) amphiphilic triblock copolymer, whose
delivered directly to the surgical site via injection, which provides biocompatibility and potential for local drug sustained delivery has been
post-surgical local analgesia for up to 72 h [26]. However, patients who demonstrated in our several previous studies already [35–37]. This
underwent total knee arthroplasty and received Exparel™ treatment did developed hLB/PLEL system is a free-flowable liquid at room tempera­
not report a clinically meaningful reduction in inpatient opioid pre­ ture and changes into a semi-solid hydrogel in situ once injected in
scription, resource utilization, or opioid-related complications [27]. In response to the physiological temperature. This characteristic is not only
short, the action duration of current local anesthetic-based formulations helpful for easier management but also conducive to avoid the local
is still too short to meet the requirements of postoperative pain precipitation of the LB base. More importantly, after administration, the
management. dissolved LB HCl could initially release from the hydrogel and

Scheme 1. Schematic illustration of levobupivacaine-loaded temperature-sensitive injectable hydrogel system (hLB/PLEL), which is used for sustained release of
anesthetic to achieve long-term local anesthesia in postoperative pain management.

2
Y. Zhang et al. Biomaterials 299 (2023) 122129

contributed to a quick nerve block for a rapid analgesic effect, while the 8.0). A predetermined amount of LB HCl was then added to the copol­
undissolved LB base dissolved and released gradually as the pH ymer solution and stirred at room temperature for 10 min to obtain a
decreased during the biodegradation of the PLEL hydrogel, resulting in a homogeneous suspension.
long-term local LB HCl release. In another words, this designed
hLB/PLEL system characterized by self-adjustable drug release due to its 2.2.3. Dynamic rheological study of hLB/PLEL
changing pH probably achieve fast-acting, long-term anesthesia, and The thermosensitive solution-hydrogel (sol-gel) phase transition of
less side effect at the same time. Here, the drug release behavior and hLB/PLEL was investigated using a rheometer (HAAKE Rheostress 6000,
biocompatibility of the thermosensitive hLB/PLEL were studied in vitro Thermo Scientific, USA) through a parallel plzate (diameter: 20 mm).
and in vivo detailedly. The anesthetic effects of the hLB/PLEL system The storage modulus (G′ ) and loss modulus (Gʺ) of hLB/PLELs with
were evaluated in sciatic nerve block, subcutaneous infiltration anes­ different PLEL concentrations and LB concentrations were measured as
thesia, and acute postoperative pain rat models systematically. functions of temperature from 20 ◦ C to 50 ◦ C. The heating rate was set at
2 ◦ C/min. The data were collected at frequency of 1.0 Hz and a
2. Materials and methods controlled stress of 1 Pa. Rheological analysis of the PLEL hydrogel (10
wt%, 15 wt%, 20 wt%) was conducted for comparison. The viscosity of
2.1. Materials blank solution with different polymer concentrations were tested around
room temperature.
Poly(ethylene glycol) (PEG, Mn = 1500) was purchased from Nanj­
ing Well Pharmaceutical co., Ltd. (China.). Stannous octoate (Sn(Oct)2), 2.2.4. Undissolved drug analyses
acetonitrile, formic acid, and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphe­ Because of the initial neutral pH of the PLEL, dissolved LB HCl and
nyltetrazolium bromide (MTT) were purchased from Sigma-Aldrich undissolved LB base co-existed in the hLB/PLEL system. The undissolved
(USA). D,L-lactide (D,L-LA) was purchased from Jinan Daigang Bioma­ LB base dissolved gradually with a decrease in pH during biodegradation
terial Co., Ltd. (China.). Levobupivacaine hydrochloride (LB HCl) was of the PLEL hydrogel. Therefore, the pH and undissolved LB base
purchased from Hubei Humanwell Pharmaceutical, Ltd. (China). Ropi­ amounts were detected at different time points. Briefly, hLB/PLEL
vacaine (purity: 94.2%) was purchased from the China National In­ samples with different drug concentrations were maintained at 37 ◦ C to
stitutes for Food and Drug Control. Dulbecco’s modified Eagle’s medium form a stable hydrogel. At predetermined intervals, three samples from
(DMEM), penicillin-streptomycin, and fetal bovine serum (FBS) were each group were collected and kept at room temperature for 20 min to
obtained from Gibco (USA). A live-dead cell staining kit was purchased obtain fluid samples. The pH of each sample was tested and centrifuged
from Keygen Biotech Corp., Ltd. (China.). Cy5.5 was purchased from the for 10 min at 3000 rpm at 4 ◦ C. The sediment was completely dissolved
Beijing Fanbo Biochemicals Company (China). Isoflurane and normal in acidic normal saline and quantitatively measured using HPLC with a
saline were purchased from Hebei Yipin Pharmaceutical Co., Ltd. reversed-phase C18 column (4.6 mm × 5 mm, 5 μm) at a wavelength of
(China) and Chengdu Qingshanlikang Pharmaceutical Co., Ltd. (China), 250 nm. A solution of 20 mM aqueous sodium bicarbonate and aceto­
respectively. nitrile (35:65, v/v) was used as the mobile phase for isocratic elution and
Sprague-Dawley rats (aged 7–9 weeks, weighing 240–280 g) were delivered at a flow rate of 1 mL/min. The pH change in the PLEL
purchased from Chengdu Dossy Biological Technology Co., Ltd. (China) hydrogel at 37 ◦ C was also detected.
and maintained in a controlled environment with a 12 h light/dark
cycle. Food and water were provided ad libitum. Before experiments
were initiated, the rats underwent a minimum 5 d adaptation period. All 2.3. In vitro cell experiments
experiments were approved by the Committee of Animal Care of the
West China Hospital, Sichuan University, China (2015015A) and carried 2.3.1. MTT test
out in accordance with the Guidance on the Care and Use of Laboratory HT22 and C2C12 cells were used to examine the in vitro cytotoxicity
Animals (U.S. National Institutes of Health Publication No. 80-23, of the PLEL hydrogel on nerve and muscle cells using the MTT assay.
revised in 1996). C2C12 and HT22 cells were purchased from the American Type Culture
Collection (ATCC, USA) and cultured in DMEM medium containing 10%
2.2. Preparation and characterization of levobupivacaine-loaded FBS, supplemented with 1% penicillin and streptomycin at 37 ◦ C in a
thermosensitive hydrogel humidified 5% CO2 atmosphere. Briefly, different concentrations of
PLEL polymer solution were prepared in DMEM medium, and the
2.2.1. PLEL copolymer synthesis leachate of the PLEL hydrogel was prepared by extraction in DMEM
The PLEL triblock copolymer was synthesized via ring-opening medium for 24 h and diluted sequentially. The cell suspensions were
copolymerization to prepare an injectable thermosensitive hydrogel, seeded in 96-well plates at a density of 3500 cells/well and incubated for
as described in our previous study [35]. Briefly, the amounts of PEG and 24 h. The medium was then replaced with fresh medium with different
D,L-LA were calculated, and Sn(Oct)2 was introduced into a nitrogen concentrations of the PLEL polymer or PLEL hydrogel leachates. After
atmosphere under a dry glass flask. The mixtures were kept at 140 ◦ C culturing for another 48 h at 37 ◦ C, the cells were subjected to the MTT
under mild agitation and nitrogen protection. After 12 h, the resulting assay, and the absorbance was measured using a microplate reader (Bio-
crude copolymer was washed with water at approximately 80 ◦ C for Rad) at 570 nm. The relative viability of the treated cells was calculated
purification. The sediment was then lyophilized to a constant weight and as a percentage of the untreated control group.
stored at − 20 ◦ C until needed. The molecular weight of the obtained
PLEL copolymer was characterized using nuclear magnetic resonance 2.3.2. Live/dead assay
spectroscopy (1H NMR, Varian 400 spectrometer, USA) and gel perme­ A live/dead assay was conducted to assess the biocompatibility of the
ation chromatography (GPC, Agilent 110 HPLC, USA). PLEL hydrogel using C2C12 and HT22 cells. The cells were cultured in
24-well plates (1 × 104 cells/well) for 24 h, and the medium was
2.2.2. Levobupivacaine loaded PLEL hydrogel preparation replaced with the prepared PLEL polymer solution (1 mg/mL) or PLEL
The composites of LB/PLEL and hLB/PLEL were fabricated by dis­ hydrogel leachate (100%). After 24 h or 48 h of culture, cells were
solving LB HCl in a PLEL copolymer solution. Briefly, the prepared PLEL stained with 2 μM Calcein AM and 8 μM PI (Live/Dead cell staining kit)
copolymer (10 wt%, 15 wt%, 20 wt%) was fully dissolved in phosphate for 30 min at room temperature. Fluorescent images were then observed
buffered saline (PBS) with different pH by mild agitation for 10 h to get a under a fluorescence microscope (Olympus, Tokyo, Japan) and images
transparent micelle solution with a different pH (pH 5.3, pH 7.0, and pH were analyzed using ImageJ 7.0 software.

3
Y. Zhang et al. Biomaterials 299 (2023) 122129

2.4. In vivo histological analyses and immunohistochemistry analyses 2.6. Pharmacokinetic study

2.4.1. Histological analyses 2.6.1. Sample preparation and liquid chromatography-mass spectrometry
Male rats (n = 8) were anesthetized with 2% isoflurane in oxygen analysis
delivered in an induction chamber and placed them in the right lateral After the sciatic nerve block, male rats (n = 6 in each group) were
position, following the method published in our previous study [16] A used to investigate the pharmacokinetics of 0.75% LB, 4% LB, and 4%
23G needle with a syringe was inserted into the center of the line, linking hLB/PLEL. Blood samples (0.2 mL) were collected from the caudal vein
the greater trochanter and the ischial tuberosity of the left leg. Once the and transferred to heparinized polypropylene tubes. Subsequently,
needle made bone contact, 0.2 mL of normal saline or the PLEL solution plasma was collected after centrifugation at 800×g for 10 min. The
was administered. The animals were euthanized with an overdose of sampling time points were as follows: pre-dose; 0.17, 0.33, 0.5, 0.67,
pentobarbital sodium after the 1, 3, 7, and 14 days of sciatic nerve block 0.83, 1, 2, 3, 4, 5, 6, and 8 h after dosing. According to the method
(n = 2 per day). The sciatic nerves and adjacent muscles were collected described in a previous study [40], plasma samples were deproteinized
and soaked in 10% buffered formalin. Hematoxylin-eosin (H&E) stain­ using acetonitrile solution with an internal standard (ropivacaine;
ing was performed [16], and the degree of inflammation of the sciatic 50:150, v/v). The mixture was then fully vortexed and centrifuged for
nerve and muscle of each rat was scored by two blinded investigators in 10 min at 28,621×g in 4 ◦ C, and the resultant supernatant was diluted
accordance with an established scoring standard [38,39]: 0 = normal; 1 with ultrapure water (50:50, v/v). The liquid chromatography-mass
= 0%–25% of area involved; 2 = 25%–50% of area involved; 3 = 50%– spectrometry system consisted of an Agilent 6460 triple quadrupole
75% of area involved; and 4 = 75%–100% of area involved. mass spectrometer and an electrospray ionization source (Agilent
Technologies, CA, USA). Chromatographic separation was performed
2.4.2. Immunohistochemistry analyses using an Agilent Extend C18 column (100 mm × 3 mm, 3.5 μm) at 35 ◦ C
The inflammatory factors, including interleukin-1 (IL-1), IL-6, and with an isocratic elution containing 0.05% formic acid and acetonitrile
tumor necrosis factor-α (TNF-α), was further carried out to evaluate the at a volume ratio of 78:22 and a flow velocity of 0.3 mL/min. The total
inflammatory response caused by the PLEL [25]. The sciatic nerve and running time for each sample was 5 min. Mass spectrometry was per­
surrounding muscles were dewaxed and rehydrated in gradual ethanol formed in the positive ionization mode under the following conditions:
series, followed by immersion in boiling citrate solution (pH = 6) for sheath gas flow rate, 11.0 L/min; sheath gas heater temperature, 300 ◦ C;
antigen retrieval for 60 min. Then, the sections were blocked with nebulizer pressure, 45 psi; capillary voltage, 3500 V. MassHunter soft­
donkey serum for 30 min at 37 ◦ C. The antibody solutions of IL-1, IL-6, ware was used to analyze the data (B.04.00 Build 4.0.479.0, Agilent
and TNF-α (1:1000; Abcam Company, USA) were added and incubated Technologies).
overnight at 4 ◦ C. Next day, the sections were rinsed with PBS and
incubated with secondary antibody solution (1:100; Life Technologies 2.6.2. Pharmacokinetic analysis
Co., Ltd., USA) for 45 min at 37 ◦ C. Then, the cell nuclei were stained A non-compartmental pharmacokinetic model was used to calculate
with DAPI. The sections were visualized under CaseViewer (3DHISTECH the value of maximum concentration (Cmax), the time to acquire Cmax
Co., Ltd., HU), and semi-quantitatively analyzed by ImageJ software (Tmax), half-life (t1/2), and area under curve (AUC) from zero to the last
(National Institutes of Health, USA). time point (AUC0-last), and from zero to infinity (AUC0-∞) by using DAS
software (version 3.2.1, Drug and Statistics, China).

2.5. Drug release of hLB/PLEL 2.7. Anesthetic effects of hLB/PLEL system in rat models

2.5.1. In vitro drug release 2.7.1. Sciatic nerve blockade model


Dialysis method was used to study the in vitro drug release behavior Male rats (n = 8 in each group) were randomly divided into eight
of LB/PLEL and hLB/PLEL. Briefly, 1 mL of the prepared 0.75% LB so­ groups: (1) normal saline, (2) PLEL, (3) 0.75% LB, (4) 0.75% hLB/PLEL,
lution or hLB/PLEL solutions with different LB concentrations (0.75%, (5) 1% hLB/PLEL, (6) 2% hLB/PLEL, (7) 3% hLB/PLEL, and (8) 4% hLB/
2%, and 4%) and 0.75% LB solution with different PLEL concentrations PLEL groups. The method of sciatic nerve blockade was elaborated in the
(10 wt%, 15 wt%, and 20 wt%) were transferred to a dialysis bag 2.4.1 section. In accordance with a previous study [16], thermal noci­
(molecular weight cut-off = 8000). The dialysis bags were placed in test ception was used to evaluate sensory function, which was determined in
tubes and equilibrated at 37 ◦ C for 30 min to form a stable hydrogel. the same anatomic area by using a modified hotplate at 56 ◦ C (model
Thereafter, 20 mL of pre-warmed phosphate-buffered saline (PBS, pH RB-200 Hotplate Analgesia Meter; Chengdu Technology & Market Co.,
7.4) was added to the test tubes, and the samples were incubated at 37 Ltd., China), and the cutoff value was set to 6 s to prevent injury or

C in a shaking incubator at 100 rpm. At predetermined intervals, the hyperalgesia. The time taken by the rats to withdraw the paw was
release medium was collected and replaced with 20 mL of fresh pre- measured using a stopwatch. Motor function was determined by
warmed PBS. The in vitro release of the LB solution from the dialysis measuring the extensor muscle force using a digital balance (model
bag was studied using the same method. The amount of LB released into HZT-B5000, Huazhi Scientific Instrument Co., Ltd., Fuzhou, China). The
the medium was measured using HPLC, as described previously. The rat was held upright over a digital balance, with the hind limbs extended
studies were conducted three times, and all data were averaged over to the platform. When the force was less than 60 g, motor function was
three individual results. considered to be blocked. The measurements were recorded at pre-dose,
10, 30, 60 min, then hourly until 12 h, then six hourly until 24 h, and
2.5.2. In vivo drug release then once every 12 h until sensory and motor functions were fully
To intuitively observe the in vivo drug release of the PLEL hydrogel, recovered. At each point, thermal nociception was measured only once;
water-soluble Cy5.5 was used as a substitute for LB. After the rats were three repeated measurements of extensor muscle force were obtained
anesthetized with 2% isoflurane in oxygen, 0.2 mL of Cy5.5 solution or and averaged.
the Cy5.5 loaded PLEL solution (Cy5.5/PLEL, 20 wt%) was dorsally and The animals were euthanized with an overdose of pentobarbital so­
injected around the sciatic nerve with a 23G needle. Rats were anes­ dium after the 1, 3, 7, and 14 days of sciatic nerve block (n = 2 per day).
thetized at time intervals of 0.25, 8, 24, 48, and 72 h, and fluorescence The sciatic nerves and adjacent muscles were collected and soaked in
images were acquired using an IVIS Lumina III imaging system (Perki­ 10% buffered formalin. Hematoxylin-eosin (H&E) staining was per­
nElmer, Caliper Life Sciences, MA, USA; excitation = 673 nm, emission formed [16], and the degree of inflammation of the sciatic nerve and
= 692 nm). The images were analyzed using the onboard software. muscle of each rat was scored by two blinded investigators in

4
Y. Zhang et al. Biomaterials 299 (2023) 122129

accordance with an established scoring standard [38,39]: 0 = normal; 1 average molecular weight of the prepared copolymer was 4520 (PEG/
= 0%–25% of area involved; 2 = 25%–50% of area involved; 3 = 50%– PDLLA ratio: 1500: 3020), and the molecular weight was distributed in a
75% of area involved; and 4 = 75%–100% of area involved. unimodal manner, indicating that the PLEL copolymer was successfully
synthesized.
2.7.2. Subcutaneous infiltration anesthesia model LB is a clinic long-acting local anesthetic. With the similarly physi­
Male rats (n = 8 in per group) were randomly divided into two cochemical property of other local anesthetics, LB consist of two forms
groups: (1) 4% LB and (2) 4% hLB/PLEL. Cutaneous trunci muscle reflex in solution, and which exist in a rapid equilibrium between the basic
(CTMR) was used to measure the effect of subcutaneous infiltration uncharged form (LB base, poorly soluble in water) and the charged
anesthesia effect [16]. After the rats were anesthetized with 2% isofl­ cationic form (LB HCl, good soluble in water). The amount of LB that
urane in oxygen, they were administered subcutaneously with 0.2 mL of exists in solution as LB base is determined by its acid dissociation con­
the test solution in the lateral thoracispinal site, which was marked with stant (pKa) and the pH of solvent, and then it can change to soluble LB
a nontoxic pen for the pinprick responsiveness (PPR) measurement. HCl when the pH changes from alkalinity or neutral or to acidic [42]. In
Then, the standardized stimulus to determine PPR was measured using clinical practice, LB commonly is used at the highest concentration of
an 18G needle affixed to a 26 g von Frey filament, and six pinpricks were 0.75%. Thus, we used the 0.75% LB HCl solution as a positive control
tested at pre-dose, 10, 30 min, 1, 2, 3, 4, 5, 6, 7, 8, 10, 12, 24, 36, and 48 when optimized the LB-loaded PLEL hydrogel systems in this study. As
h until the CTMR fully recovered. For each stimulation, it was accepted shown in Fig. 1A, the thermo-sensitive behavior of three types of
that PPR = 1 if the lateral thoracispinal muscles induced a skin move­ LB-loaded PLEL hydrogel composites (0.75% LB) made by PLEL solu­
ment over the back and that PPR = 0 if reflex movement was absent. tions with different pH were tested firstly. The LB-loaded PLEL hydrogel
composites made by acidic was transparent and homogeneous, while the
2.7.3. Acute postoperative pain model ones made by neutral and alkaline PLEL solution were cloudy, suggested
Male rats (n = 6 in per group) were randomly divided into four a hybrid of LB HCl form and LB base form. Both the LB/PLEL made by
groups: (1) saline, (2) PLEL, (3) 0.75% LB, and (4) 4% hLB/PLEL. The acidic or neutral PLEL solution were free flowable liquids at room
commonly used incisional pain model was used as described in a pre­ temperature and changed into a semi-solid gel rapidly around body
vious study [41]. Briefly, rats were anesthetized with 2% isoflurane in temperature, which were similar to the sol-gel of blank PLEL hydrogel.
oxygen, and the left leg was prepared using an iodophor solution for However, in the case of LB/PLEL made by alkaline PLEL solution, a
disinfection. Then, a 1 cm longitudinal incision, starting 0.5 cm from the longer time was needed to be hydrogel at 37 ◦ C, and the hydrogel is
proximal edge of the heel and extending toward the toes, was made with unstable and weak. Therefore, we chose the acidic and neutral PLEL
a number 11 blade through the skin and fascia. The plantaris muscle was solution to prepare LB-loaded hydrogel systems for further study, which
elevated and incised in all rats. Finally, the skin was sutured using a 5-0 were denoted as LB/PLEL and hLB/PLEL, respectively. In addition, We
nylon surgical sutures. After surgery, 25 μL of the drug was injected noticed that the pH of the PLEL hydrogel and hLB/PLEL were decreased
adjacent to the wound, and the rats were placed in their cages for with time when the samples were kept at 37 ◦ C, which may be caused by
recovery. the generation of D,L-lactide and low-molecular-weight oligomers dur­
The mechanical withdrawal threshold and pain score by weight were ing the biodegradation of the PLEL polymer (Fig. 1B). As a result of the
used to measure pain behavior. The mechanical withdrawal threshold pH decrease of hydrogel, the hLB/PLEL samples became clearer with
was determined by placing the rats on a plastic mesh floor (12 mm × 12 time and almost transparent after 96 h, suggesting the gradual dissolu­
mm grid) and using von Frey filaments starting with 0.4 g and tion of the insoluble LB base (Fig. 1C). Simultaneously, the hLB/PLEL
continuing until a withdrawal response occurred or the cutoff value (26 samples presented stable and homogeneous hydrogels at 37 ◦ C without
g) was reached. The testing site was located in the same area adjacent to precipitation. Generally, drug delivery systems made by physical
the wound near the heel. The pain score by weight was determined by hydrogels exhibit an initial burst release owing to the quick diffusion of
placing the rats on a plastic mesh floor and allowing them to acclimate small-molecule drugs. The dynamic dissolution of the LB base in
for at least 15 min. Each rat was observed once every 5 min for 1 h. The hLB/PLEL may relieve the initial burst release and extend the period of
scoring standard depended on the foot position: 0 = the wound was drug release. Thus, the drug release behavior of LB/PLEL and hLB/PLEL
blanched or distorted; 2 = the foot was completely off the mesh; was studied in vitro. As expected, hLB/PLEL showed the slowest drug
otherwise, the score was 1. The total pain score at 1 h represents the release compared with the LB/PLEL and free LB solution groups, sug­
degree of spontaneous pain. The time points for pain behavior mea­ gesting the potential for long-term local anesthesia (Fig. 1D). Consid­
surements were baseline, 2 h after surgery, once daily in the first 3 days ering the possible effect of hydrogel concentration on drug release,
after surgery, and 7 days after surgery. different concentrations of hydrogel (10 wt%, 15 wt%, and 20 wt%)
were used to prepare hLB/PLEL as well. With the increase of PLEL
2.8. Statistical analyses concentration from 10 wt% to 20 wt%, the slow-release effect of
hLB/PLEL was improved gradually (Fig. 1E), implying that a higher
Homogeneity-variance data were assessed using ANOVA followed by polymer concentration benefits to the sustained drug release. Although
Dunnett’s test to compare the treatment groups with the control. we have tried to make hLB/PLEL using PLEL hydrogel in 25 wt% to get a
Otherwise, the data were assessed using the Kruskal-Wallis test followed delaying drug release, it failed to carry out. Because the 25 wt% PLEL
by the Wilcoxon signed-rank test to compare with the control, and was found to have a higher viscosity at room temperature (Fig. S1),
Bonferroni correction was used to adjust the p values. The level of sta­ which is not only unfriendly to the dissolution and dispersion of LB, but
tistical significance was set at p < 0.05. Data were analyzed using SPSS also difficult to inject in vivo and sterilize by filtration. Thus, hLB/PLEL
Statistics (version 23, Chicago, IL, USA). with a concentration of 15 wt% and 20 wt% seemed to be the superior
candidates for further formulation optimization.
3. Results and discussion Encouraged by the in vitro drug release results, we preliminarily
evaluated the local anesthetic effect of hLB/PLEL and LB/PLEL using a
3.1. Preparation and characterization of levobupivacaine-loaded PLEL rat sciatic nerve block model (Fig. 1F). The duration of sensory blockade
hydrogel (5.88 ± 1.13 h) in 0.75% hLB/PLEL treated group showed a significant
difference with that of 0.75% LB/PLEL group (4.63 ± 0.74 h, p = 0.02)
The biodegradable PLEL copolymer was prepared via ring-opening and 0.75% LB (3.13 ± 0.35 h). For motor blockade, 0.75% hLB/PLEL
copolymerization of D,L-LA initiated by PEG, using Sn(Oct)2 as the (6.38 ± 1.69 h) also produced a significantly longer duration than
catalyst. According to the 1H NMR spectrum and GPC measurements, the 0.75% LB/PLEL (4.63 ± 0.74 h, p = 0.018) and 0.75% LB (3.25 ± 0.46

5
Y. Zhang et al. Biomaterials 299 (2023) 122129

Fig. 1. The sol-gel phase transition behavior of levobupivacaine-loaded thermosensitive hydrogel system. (A) Thermosensitive sol-gel phase transition of blank PLEL
hydrogel and LB-loaded hydrogel systems. (B) Changes in pH of PLEL hydrogel and hLB/PLEL upon time. (C) Transparency changes of hLB/PLEL upon time. (D) In
vitro drug release of different LB-loaded hydrogel systems. (E) In vitro drug release of hLB/PLEL with different concentrations of PLEL hydrogel. (F) Schematic
representation of rat sciatic nerve block model. (G) Duration of sensory blockade and motor blockade on the rat sciatic nerve block model after treated with different
LB preparations. (H) Effect of local anesthesia of hLB/PLEL with different polymer concentrations on the rat sciatic nerve block model. Data are presented as mean
± SD.

h) (Fig. 1G). These results indicate that hLB/PLEL containing a hybrid of hydrogel concentration. As shown in Fig. 2B, increase in LB encapsula­
LB HCl and LB base is superior in sustainably releasing LB and extending tion resulted in an increase in the thermoresponsive temperature. Spe­
the effect of local anesthesia. Furthermore, we compared the local cifically, when the LB concentration reached to 3% and 4%, the
blockade effect on the sensory and motor blockade functions of hLB/ thermoresponsive temperature of hLB/PLEL samples at polymer con­
PLEL made with 15 wt% and 20 wt% PLEL hydrogels (Fig. 1H). Both the centrations of 20 wt% were around 34 ◦ C, while that of samples in 10 wt
sensory and motor blockade durations in rats treated with hLB/PLEL % and 15 wt% hydrogels were above 37 ◦ C. As an injectable in-site
(20 wt% PLEL) were longer than those in rats treated with hLB/PLEL sustained drug delivery system, enough LB encapsulation and appro­
(15 wt% PLEL). Therefore, we chose the hLB/PLEL with 20 wt% PLEL priate gelation temperature less than 37 ◦ C of the hLB/PLEL is expected.
for further studies. hLB/PLEL made by 20 wt% PLEL showed the more potential to resist the
drug concentration limitation. However, when the LB concentration
reached to 5%, the gelation temperature of hLB/20 wt% PLEL was above
3.2. Sol-gel phase transition and dynamic drug composition of hLB/PLEL 37 ◦ C and its G′ at 37 ◦ C was measured as less than 2 Pa, which was not
applicable for in vivo drug delivery any more (Fig. 2B and C). Thus, an
Dynamic rheological measurements were performed to study the hLB/20 wt% PLEL with LB encapsulation of less than 4% was preferred
mechanical properties of the PLEL hydrogel and hLB/PLEL during the here.
sol-gel phase transition. According to the results in Fig. 2A and Fig. S2, The initial concentration of LB base in hLB/PLEL with 0.75%, 2%,
both the blank PLEL and hLB/PLEL formulations showed an evident and 4% LB were 2.23 ± 0.43 mg/mL, 7.19 ± 0.11 mg/mL, and 9.34 ±
change in storage modulus (G′ ) upon temperature, characterized as a 0.21 mg/mL, respectively (Fig. 2D). This difference could be ascribed to
rapidly growing after the temperature increasing to the thermores­ the pH of the hLB/PLEL and the amount of the loaded LB HCl. We further
ponsive temperature. It is worth mentioning that the gelation temper­ investigated the pH and LB base dynamic changes in hLB/PLEL samples
ature of hLB/PLEL is closely related to the loaded LB concentration and

6
Y. Zhang et al. Biomaterials 299 (2023) 122129

Fig. 2. Rheological properties and dynamic drug composition of hLB/PLEL. (A) Changes in G′ of hLB/PLEL (20 wt%) with different LB concentrations. (B) Ther­
moresponsive temperature of hLB/PLEL with different polymer concentration and LB concentrations. (C) G′ of hLB/20 wt% PLEL with different LB concentrations at
37 ◦ C. (D) The initial concentration of LB base in hLB/PLEL with different drug loading. (E) Changes in pH of hLB/PLEL systems upon time. (F) Concentration
changes in LB base of hLB/PLEL systems upon time. Data are presented as mean ± SD.

with different LB encapsulations at 37 ◦ C. The results are shown in 3.3. Drug release of hLB/PLEL
Fig. 2E and F. The initial pH of the hLB/PLEL decreased with the LB
encapsulation, and the pH of all the samples decreased with time. The in vitro drug release behavior of hLB/PLEL was evaluated under
Accordingly, the amount of LB base decreased when time went on, conditions mimicking the physiological microenvironment. Fig. 3A
suggesting the gradual dissolution of the LB base. shows that LB was released from the hLB/PLEL formulation in a sus­
tained manner over an extended period, whereas the free LB solution
showed a quick release within the first 2 h. The release profiles of hLB/
PLEL presented a biphasic pattern, with an initial burst release in the

Fig. 3. Drug release from the PLEL hydrogel both in vivo and in vitro. (A) In vivo drug release behavior of hLB/PLEL. (B) Real-time intravital fluorescence images of
local retention of Cy5.5 after injected around the sciatic nerve of rats. (C) Quantitative analysis of the fluorescence intensity of local Cy5.5 at different time. (D)
Results of pharmacokinetic study of hLB/PLEL systems on rat sciatic nerve block model. Data are presented as mean ± SD.

7
Y. Zhang et al. Biomaterials 299 (2023) 122129

first 2 h, followed by a sustained release period, suggesting a swelling and cytotoxicity of the PLEL hydrogel on the nerve and muscle cells were
diffusion-controlled process [35]. Logically, the initial burst of LB can assessed using HT22 and C2C12 cells in vitro. According to the results of
contribute to quick work after injection, and the subsequent sustained the MTT test in Fig. 4A, the PLEL polymer solution showed mild cyto­
drug release is preferred for a long-lasting action. Moreover, the increase toxicity to both HT22 and C2C12 cells, whose viability was more than
in LB encapsulation from 0.75% to 4% seemed to have benefited the 90%, even at a high polymer concentration of 1000 μg/mL. The viability
long-term drug release, which may have resulted from the dynamic of HT22 and C2C12 cells was approximately 90% after co-culture with
dissolution of the insoluble fraction during the pH decrease of the the PLEL hydrogel leachate for 48 h (Fig. 4B). Similar results were
hLB/PLEL system. observed in the live/dead assay as shown in Fig. 4C. Compared with the
To intuitively observe the drug release capacity of the PLEL hydrogel untreated group, the addition of the PLEL polymer or hydrogel leachate
in vivo, the water-soluble fluorescence chromogenic agent Cy5.5, was did not affect the proliferation and morphology of HT22 and C2C12
used as a substitute for LB, and noninvasive intravital fluorescence im­ cells. These results indicate that the PLEL hydrogel had little cytotoxicity
aging was used to monitor the local retention of Cy5.5 in real-time, as on the nerve and muscle cells, indicating that it is a safe biomaterial for
shown in Fig. 3B. After injected around the sciatic nerve of rats, the the local delivery of anesthetics.
fluorescence of Cy5.5, observed mainly at the injection site, showed a Mild tissue compatibility is of considerable importance for the clin­
persistent maintenance and slow decay for longer than 72 h in the ical application of local anesthetic formulations. We evaluated the in
Cy5.5/PLEL treatment group. In contrast, the fluorescence intensity in vivo local toxicity of the PLEL hydrogel on the sciatic nerve and adjacent
the Cy5.5 solution group decayed rapidly with time owing to its rapid muscles using a sciatic nerve blockade model in rats. Rats were eutha­
clearance in vivo. According to the quantitative analysis of the fluores­ nized 1,3, 7, and 14 d after PLEL hydrogel administration, and sciatic
cence intensity, the fluorescence of Cy5.5/PLEL was stronger than that nerves with adjacent tissues were collected for macroscopic examination
of the Cy5.5 solution during all observations, indicating that PLEL and H&E staining. As shown in Fig. 4D, on macroscopic examination, all
hydrogel holds the potential to prolong the drug release and extend the rats injected with PLEL exhibited gelatinous white deposits of hydrogel
local drug retention in the model of sciatic nerve blockade (Fig. 3C). surrounding the sciatic nerve. The PLEL hydrogel was biodegradable,
and most of it degraded within 7 d. Moreover, these findings show that
3.4. Pharmacokinetic study of hLB/PLEL the degradation rate of the PLEL hydrogel was increased with time. This
phenomenon may be related to the degradation of the PLEL generated
Local anesthetics were administered and absorbed into the circula­ acidic products (lactic acid), which led to a pH decrease in the local
tion through blood flow. The systemic concentration of local anesthetics microenvironment and expedited degradation of the PLEL hydrogel.
is closely related to their concentration in local tissues. Therefore, we PLEL did not cause significant pathological and morphological changes
determined the plasma concentration and concentration-time profiles in the rats (Fig. 4D). The total score of inflammation in the sciatic nerve
for 0.75% LB, 4% LB, and 4% hLB/PLEL application near the rat sciatic and muscle demonstrated that PLEL had a mild tissue compatibility
nerve. The data are shown in Fig. 3D and the computed pharmacokinetic (Fig. S3A). Then, the results of immunofluorescent staining and semi-
parameters are listed in Table 1. As shown in the plasma concentration- quantitative analyses of the inflammatory factors are shown in
time curve, the maximum concentration of 4% LB (993.03 ± 55.88 ng/ Fig. 4E. The red fluorescence representing the inflammatory factors was
mL) was significantly higher than that of 0.75% LB (377.77 ± 16.57 ng/ observed mainly in the muscle around the sciatic nerve. As shown in
mL, p < 0.001). However, the Cmax of 4% hLB/PLEL was 614.28 ± Figs. S3B–D, the fluorescence intensities of the rats treated with PLEL
54.19 ng/mL, which was not significantly different from that of 0.75% hydrogel were not significantly different from those treated with saline.
LB (p = 0.216). Meanwhile, the maximum concentrations were observed The observed inflammatory response was moderate, which was consis­
at 1.00 ± 0.22 h for 0.75% LB, 1.00 ± 0.21 h for 4% LB, and 1.50 ± 0.47 tent with the results of H&E staining. Additionally, no statistical dif­
h for 4% hLB/PLEL after dosing. The hydrogel was found to slowly ferences were found between these groups, indicating that the
release LB. The calculated AUC0-8 of 4% LB–hydrogel was 2820.70 ± inflammatory response to the PLEL hydrogel was mild. This quality
243.63 ng/mL*h, which was reduced at 37.69% than 4% LB (4526.61 ± makes it highly desirable for drug delivery applications.
182.48 ng/mL*h). Given the blood volume in the rats [43], we did not
continue to collect the blood samples to determine the time to reach the 3.6. Anesthetic effects of hLB/PLEL in sciatic nerve blockade model
minimal concentration in the 4% LB and 4% hLB/PLEL groups. How­
ever, we calculated the AUC0-∞ of 4% LB and 4% hLB/PLEL, which were For local anesthetics, the duration of anesthesia was determined
5877.52 ± 422.85 ng/mL*h and 5642.31 ± 313.34 ng/mL*h, respec­ using two principal factors: concentration and volume [44]. However, a
tively. These results preliminarily indicate that the PLEL hydrogel sys­ concentration that exceeds the limitation will induce local and even
tem could release LB slowly in vivo for long-lasting local drug delivery in systematic toxicity. Therefore, the duration and histopathological ex­
vivo. aminations of different concentrations of hLB/PLEL were measured in a
rat sciatic nerve blockade model to determine the efficacy and safety of
hLB/PLELs for local anesthesia. In pain research, the effect of sex on
3.5. In vitro cytotoxicity and in vivo biocompatibility of PLEL hydrogel
nociceptive processing has received attention. Previous studies have
shown that the response to pain stimulation varies between sexes [45].
Considering the local delivery of anesthetics, the biocompatibility
Therefore, to eliminate the effect of sex, all rats in the in vivo pharma­
cological studies were male.
Table 1
The duration of effect in different concentrations of LB in the PLEL
Systemic pharmacokinetic parameters in sciatic nerve block (Mean ± SEM).
hydrogel was measured. The sensory and motor functions of rats injec­
Pharmacokinetic 0.75% LB 4% LB solution 4% hLB/PLEL ted with saline and PLEL hydrogel did not differ at the baseline, which is
parameters solution
not presented. The onset time of rats injected with LB or hLB/PLEL was
Cmax (ng/mL) 377.77 ± 993.03 ± 55.88 614.28 ± 54.19 determined in the first 10 min post-dosing, indicating that the sustained-
16.57
release formulation could achieve the minimum effective concentration
Tmax (h) 1.00 ± 0.22 1.00 ± 0.21 1.50 ± 0.47
t1/2 (h) 1.13 ± 0.22 3.54 ± 0.35 7.58 ± 3.38 quickly (Fig. 5A). However, the duration of 0.75% and 1% hLB/PLEL-
AUC0–8 (ng/mL*h) 949.59 ± 4526.61 ± 2820.70 ± induced sensory and motor blockades were not significantly different
50.18 182.48 243.63 from that of 0.75% LB (Fig. 5B). Then, rats treated with 2%, 3%, or 4%
AUC0-∞ (ng/mL*h) 937.39 ± 5877.52 ± 5642.31 ± hLB/PLEL presented sensory blockades lasting for 7.88 ± 1.54 h (p =
52.11 422.85 313.34
0.036), 15.17 ± 1.83 h (p = 0.001), and 17.17 ± 2.24 h (p = 0.001),

8
Y. Zhang et al. Biomaterials 299 (2023) 122129

Fig. 4. In vitro Cytotoxicity and in vivo


biocompatibility of PLEL hydrogel. (A)
Cytotoxicity of PLEL copolymer against
HT22 cells and C2C12 cells after co-
culture for 48 h. Data are presented as
mean ± SD. (n = 4). (B) Cell viability of
HT22 cells and C2C12 cells after treated
with PLEL hydrogel leachate for 48 h.
Data are presented as mean ± SD. (n =
3). (C) Results of live/dead assay at
different treatment time. (Scale bar:
100 μm) (D) Macroscopic examination
(green arrow: hydrogel; red arrow:
sciatic nerve) and representative histo­
logical images of the sciatic nerves and
adjacent muscles (with H&E staining).
The scale bars in the H&E staining panel
represent 500 μm. (E) Immunohisto­
chemistry analyses of sciatic nerve and
surrounding muscles after treated with
PLEL hydrogel. Red fluorescence in­
dicates the inflammatory factors and
blue fluorescence represents the nuclei.
Scale bar = 200 μm. (For interpretation
of the references to colour in this figure
legend, the reader is referred to the Web
version of this article.)

respectively, which were significantly longer than that induced by hLB/PLEL was increased to 38.33 h. Then, the sensory (n = 1, 33%) and
0.75% LB (2.63 ± 0.16 h, Fig. 5B). Prolonged sensory blockade had a motor (n = 2, 67%) function of rats in 8% hLB/PLEL group did not re­
linear relationship with LB concentration. Similar to the sensory turn to normal even after the 14 d sciatic nerve block. However,
blockade, the duration of motor blockade induced by hLB/PLEL dis­ macroscopic changes, including muscle stiffness of the hind limb and
played a concentration-dependent relationship (Fig. 5C and D). The abnormal gait, were observed in rats injected with 6% and 8% hLB/
hLB/PLEL with LB at concentration of 2%–4% (8.42 ± 1.54 h at 2%, p = PLEL, which may have been induced by neuropathological injury. Thus,
0.047; 16.50 ± 2.16 h at 3%, p = 0.007; and 20.33 ± 2.84 h at 4%, p = 4% LB was regarded as the maximum safe concentration of hLB/PLEL.
0.001, respectively) produced a significantly longer effect than that Mild tissue compatibility is of considerable importance for the clin­
induced by 0.75% LB (3.00 ± 0 h). Then, the 6% hLB/PLEL and 8% hLB/ ical application of local anesthetic formulations. The rats were eutha­
PLEL were further measured to determine whether it is safe to increase nized for histological analysis 1, 3, 7, and 14 d after dosing (n = 2 in per
the concentration of LB in the PLEL hydrogel (n = 3). The duration of 6% day). Macroscopic examination and representative histological images

9
Y. Zhang et al. Biomaterials 299 (2023) 122129

Fig. 5. Anesthetic effects and tissue toxicity of hLB/PLEL in sciatic nerve blockade model. (A) The actual measurement value of paw withdraw latency. (B) Time to
recovery from sciatic nerve block for sensory blockade. (C) The actual measurement value of extensor postural thrust force. (D) Time to recovery from sciatic nerve
block for motor blockade. (E) The representative histological images of the sciatic nerves and adjacent muscles of hLB/PLEL (with H&E staining). The scale bars in
the H&E staining panel represent 500 μm. (F) The sciatic nerve inflammation score of hLB/PLEL. (G) The muscle inflammation score of hLB/PLEL.

10
Y. Zhang et al. Biomaterials 299 (2023) 122129

of the sciatic nerves and adjacent muscles are presented in Fig. 5E. We site, provides post-surgical local analgesia for up to 72 h, and is
compared the total score of inflammation in the sciatic nerve and muscle approved for postoperative pain [26]. In a previous study, the effect of
within 14 days in the 0.75% LB and 0.75%–4% hLB/PLEL groups the clinically available Exparel™ lasted only for 4–5 h in a rat sciatic
(Fig. 5F and G). The degree of inflammation in the local tissue of hLB/ blockade model. The effect 1% hLB/PLEL in the sciatic nerve block
PLEL was not significant than that recorded for LB at the maximum model lasted for 5.29 ± 0.61 h, which was not significantly different
clinical concentration. with that of Exparel™ (contains 1.31% bupivacaine). However, 4%
Exparel™ (Pacira Pharmaceuticals, USA) is a liposomal encapsula­ hLB/PLEL produced an anesthesia duration four times longer than that
tion of 13.3 mg/mL of bupivacaine arranged in a microscopic of Exparel™. Meanwhile, we compared 0.75%–4% hLB/PLEL with
honeycomb-like structure, which is delivered directly to the surgical 0.75% LB in terms of sciatic nerve and muscle inflammation. The

Fig. 6. Anesthetic effects of hLB/PLEL in subcutaneous infiltration blockade model and acute postoperative pain model. (A) Schematic representation of rat sub­
cutaneous infiltration blockade model. (B) The actual measurement value of cutaneous trunci muscle reflex (pinprick responsiveness, PPR). (C) Time to recovery from
subcutaneous infiltration blockade. *: P < 0.05. (D) Schematic representation of rat acute postoperative pain model. (E) The actual measurement value of withdrawal
threshold (mechanical pain). *: compared with saline (*: P < 0.05; **: P < 0.01; ***: P < 0.001); #: compared with PLEL (#: P < 0.05; ##: P < 0.01; ###: P <
0.001); &: compared with levobupivacaine (&: P < 0.05). (F) The actual measurement value of cumulative pain score (spontaneous pain). *: compared with saline (*:
P < 0.05; **: P < 0.01; ***: P < 0.001); #: compared with PLEL (#: P < 0.05; ##: P < 0.01; ###: P < 0.001); &: compared with levobupivacaine (&: P < 0.05).

11
Y. Zhang et al. Biomaterials 299 (2023) 122129

degrees of inflammation in the local tissue of 4% hLB/PLEL were 4. Conclusions


considerably not more than that recorded for LB at the clinically
maximal concentration. Based on these results, 4% hLB/PLEL may be a In this study, a biodegradable thermosensitive hydrogel was fabri­
possible method for the prolonged postoperative pain management in cated to release LB for postoperative analgesia. LB existed in the neutral
clinical settings. base form and the charged cation form due to the chemical construction
of LB and the initial pH of the PLEL hydrogel. After local injection, the
3.7. Anesthetic effects of hLB/PLEL in subcutaneous infiltration blockade dissolved LB HCl was first released from the hydrogel, and the insoluble
model LB base dissolved and was released gradually as the pH decreased during
biodegradation of the PLEL hydrogel, resulting in long-term LB release.
We used a sciatic nerve blockade model to consider the 4% hLB/PLEL Furthermore, the PLEL hydrogel showed mild cytotoxicity to nerve and
as the optimal concentration and to determine the longest duration with muscle cells, and histological analyses demonstrated that the degree of
mild tissue toxicity in rats. The subcutaneous anesthetic effect of 4% inflammation in local tissues of the hLB/PLEL was acceptable.
hLB/PLEL and the reason for the extended duration were verified using Furthermore, the hLB/PLEL system significantly extended the effect of
the well-established CTMR model, and the effects of 4% hLB/PLEL and local analgesia in sciatic nerve block, subcutaneous infiltration anes­
4% LB were explored (Fig. 6A). The measured values are shown in thesia, and acute postoperative pain models in rats, suggesting that it is a
Fig. 6B. The 4% hLB/PLEL produced a subcutaneous anesthetic effect promising candidate for local anesthetic delivery and postoperative pain
with a significantly longer duration than that recorded with 4% LB (34 management.
± 2 h vs. 18 ± 3 h, p = 0.003, Fig. 6C). The concentration of local
anesthetic is a major factor affecting the duration of anesthesia [46]. Credit author statement
Thus, we chose 4% LB as the control group to determine whether the
long-acting local anesthesia was generated by the slow-release effect and YuJun Zhang, Conceptualization, Methodology, Investigation, Re­
not by the increased concentration. However, 4% hLB/PLEL produced a sources, Data Curation, Writing - Original Draft; Kun Shi, Conceptuali­
nearly two-fold longer duration than that of 4% LB. Thus, the prolonged zation, Methodology, Investigation, Resources, Data Curation, Writing -
duration of this drug delivery system was not the result of drug Original Draft; Xi Yang, Investigation, Data Curation; Wen Chen,
concentration. Investigation, Data Curation; TianHong Wang, Investigation, Data
Curation; Yi Kang, Investigation, Data Curation; DeYing Gong, Investi­
3.8. Efficiency evaluation of hLB/PLEL in acute postoperative pain model gation, Data Curation; WenSheng Zhang, Conceptualization, Method­
ology, Resources, Supervision, Project administration, Writing - Review
Previously used animal models, including sciatic nerve and subcu­ & Editing Supervision; ZhiYong Qian, Conceptualization, Methodology,
taneous infiltration blocks, are standard models of neurobehavioral Resources, Supervision, Project administration, Writing - Review &
assessment for drug development, but they are not used for studying Editing Supervision.
postoperative pain. Therefore, surgical incision of the hind paw in rats
was used to estimate the effect of hLB/PLEL (Fig. 6D) [41]. The baseline Declaration of competing interest
of paw withdrawal threshold for von Frey hair stimulation was not
significantly different between the two groups (Fig. 6E). However, the The authors declare that they have no known competing financial
incised rats injected with 4% hLB/PLEL showed a higher paw with­ interests or personal relationships that could have appeared to influence
drawal threshold than the rats injected with 0.75% LB and control the work reported in this paper.
groups. Skin incision and treatment with 4% hLB/PLEL resulted in a
mild spontaneous pain after incision (Fig. 6F). Postoperative pain is Data availability
characterized by constant spontaneous pain near the surgical site and
acute exacerbation due to movement or other forms of stimulation. The Data will be made available on request.
pain scores measured in this study were related to either touch-evoked
pain or ongoing pain during the post-operative period. Therefore, our Acknowledgments
findings that a single postoperative dose of 4% hLB/PLEL attenuates the
pain scores is valuable from a therapeutic perspective. This work was supported by the National Natural Science Foundation
The common types of local anesthesia are topical anesthesia, infil­ of China (81973274, U21A20417, 31800797), the Sichuan Science and
tration anesthesia, and peripheral nerve blockade. In this study, the rat Technology Program, China (2020YJ0053, 2022YFS0333,
sciatic nerve block is the standard animal model used to simulate the 2022YFS0308), the China Postdoctoral Science Foundation
method of peripheral nerve blockade in human for surgeries at or below (2020M673235), the Post-Doctor Research Project, West China Hospi­
the knee. Then, the infiltration anesthesia is to anesthetize nerve ter­ tal, Sichuan University, China (2019HXBH012, 2018HXBH066), the
minal in a finite area of tissue by the injection of local anesthetics Science and Technology Department of Chengdu City, China (2021-
nearby. Contrast to nerve block, in which nerve axons are the target and YF05-00855-SN), and the 1•3•5 project for disciplines of excellence,
the injection may take place in an area removed from the surgical site. West China Hospital, Sichuan University, China (ZYGD18002,
Infiltration anesthesia alone can provide adequate analgesia for ZYJC21068).
superficial procedures (such as inguinal herniorrhaphy, breast and
anorectal surgery, shoulder and knee arthroscopy) but is vastly Appendix A. Supplementary data
underutilized in clinical practice. However, advantages of recom­
mended wound infiltration with local anesthetics are safety, simplicity, Supplementary data to this article can be found online at https://doi.
and enhanced postoperative analgesia [47], and the effect of hLB/PLEL org/10.1016/j.biomaterials.2023.122129.
was evaluated in acute postoperative pain rat model by wound infil­
tration. Thus, multiple animal models were used to demonstrate the References
sufficient feasibility of the proposed approach and significantly
advanced pain management. [1] R. Chou, D.B. Gordon, O.A. de Leon-Casasola, J.M. Rosenberg, S. Bickler,
T. Brennan, T. Carter, C.L. Cassidy, E.H. Chittenden, E. Degenhardt, S. Griffith,
R. Manworren, B. McCarberg, R. Montgomery, J. Murphy, M.F. Perkal, S. Suresh,
K. Sluka, S. Strassels, R. Thirlby, E. Viscusi, G.A. Walco, L. Warner, S.J. Weisman,
C.L. Wu, Management of postoperative pain: a clinical practice guideline from the

12
Y. Zhang et al. Biomaterials 299 (2023) 122129

American pain society, the American society of regional anesthesia and pain hydrogel for prolonged relief of pain at the surgical wound, Int. J. Pharm. 558
medicine, and the American society of anesthesiologists’ committee on regional (2019) 225–230.
anesthesia, executive committee, and administrative council, J. Pain 17 (2) (2016) [25] W. Zhang, W. Xu, C. Ning, M. Li, G. Zhao, W. Jiang, J. Ding, X. Chen, Long-acting
131–157. hydrogel/microsphere composite sequentially releases dexmedetomidine and
[2] H. Kehlet, T.S. Jensen, C.J. Woolf, Persistent postsurgical pain: risk factors and bupivacaine for prolonged synergistic analgesia, Biomaterials 181 (2018) 378–391.
prevention, Lancet (London, England) 367 (9522) (2006) 1618–1625. [26] A.L. Balocco, P.G.E. Van Zundert, S.S. Gan, T.J. Gan, A. Hadzic, Extended release
[3] E.C. Wick, M.C. Grant, C.L. Wu, Postoperative multimodal analgesia pain bupivacaine formulations for postoperative analgesia: an update, Curr. Opin.
management with nonopioid analgesics and techniques: a review, JAMA Surg 152 Anaesthesiol. 31 (5) (2018) 636–642.
(7) (2017) 691–697. [27] L. Pichler, J. Poeran, N. Zubizarreta, C. Cozowicz, E.C. Sun, M. Mazumdar, S.
[4] R.A. Gabriel, M.W. Swisher, J.F. Sztain, T.J. Furnish, B.M. Ilfeld, E.T. Said, State of G. Memtsoudis, Liposomal bupivacaine does not reduce inpatient opioid
the art opioid-sparing strategies for post-operative pain in adult surgical patients, prescription or related complications after knee arthroplasty: a database analysis,
Expet Opin. Pharmacother. (2019) 1–13. Anesthesiology 129 (4) (2018) 689–699.
[5] J. Li, T. Zhang, J. Sun, F. Ren, H. Jia, Z. Yu, J. Cheng, W. Shi, Synthesis and [28] P.J. Sarvela, M.P. Paloheimo, P.H. Nikki, Comparison of pH-adjusted bupivacaine
evaluation of peptide–fentanyl analogue conjugates as dual μ/δ-opioid receptor 0.75% and a mixture of bupivacaine 0.75% and lidocaine 2%, both with
agonists for the treatment of pain, Chin. Chem. Lett. 33 (8) (2022) 4107–4110. hyaluronidase, in day-case cataract surgery under regional anesthesia, Anesth.
[6] C.D. Soelberg, R.E. Brown Jr., D. Du Vivier, J.E. Meyer, B.K. Ramachandran, The Analg. 79 (1) (1994) 35–39.
US opioid crisis: current federal and state legal issues, Anesth. Analg. 125 (5) [29] D.M. Coventry, J.G. Todd, Alkalinisation of bupivacaine for sciatic nerve blockade,
(2017) 1675–1681. Anaesthesia 44 (6) (1989) 467–470.
[7] C. Eccleston, T.E. Cooper, E. Fisher, B. Anderson, N.M. Wilkinson, Non-steroidal [30] Q.J. Milner, B.C. Guard, J.G. Allen, Alkalinization of amide local anaesthetics by
anti-inflammatory drugs (NSAIDs) for chronic non-cancer pain in children and addition of 1% sodium bicarbonate solution, Eur. J. Anaesthesiol. 17 (1) (2000)
adolescents, Cochrane Database Syst. Rev. 8 (2017) CD012537. 38–42.
[8] D.S. James, The multisystem adverse effects of NSAID therapy, J. Am. Osteopath. [31] I. Balga, H. Gerber, X.H. Schorno, F. Aebersold Keller, H.P. Oehen, Bupivacaine
Assoc. 99 (11_suppl) (1999) S1–S7. crystal deposits after long-term epidural infusion, Anaesthesist 62 (7) (2013)
[9] T. Dreischulte, D.R. Morales, S. Bell, B. Guthrie, Combined use of nonsteroidal anti- 543–548.
inflammatory drugs with diuretics and/or renin-angiotensin system inhibitors in [32] C. Weldon, T. Ji, M.T. Nguyen, A. Rwei, W. Wang, Y. Hao, C. Zhao, M. Mehta, B.
the community increases the risk of acute kidney injury, Kidney Int. 88 (2) (2015) Y. Wang, J. Tsui, R.P. Marini, D.S. Kohane, Nanoscale bupivacaine formulations to
396–403. enhance the duration and safety of intravenous regional anesthesia, ACS Nano 13
[10] A. Arfe, L. Scotti, C. Varas-Lorenzo, F. Nicotra, A. Zambon, B. Kollhorst, T. Schink, (1) (2019) 18–25.
E. Garbe, R. Herings, H. Straatman, R. Schade, M. Villa, S. Lucchi, V. Valkhoff, [33] R. Ohri, J.C. Wang, L. Pham, P.D. Blaskovich, D. Costa, G. Nichols, W. Hildebrand,
S. Romio, F. Thiessard, M. Schuemie, A. Pariente, M. Sturkenboom, G. Corrao, C, N. Scarborough, C. Herman, G.R. Strichartz, Prolonged amelioration of
Safety of Non-steroidal Anti-inflammatory Drugs Project, Non-steroidal anti- experimental postoperative pain by bupivacaine released from microsphere-coated
inflammatory drugs and risk of heart failure in four European countries: nested hernia mesh, Reg. Anesth. Pain Med. 39 (2) (2014) 97–107.
case-control study, BMJ 354 (2016) i4857. [34] M. Sanford, G.M. Keating, Levobupivacaine: a review of its use in regional
[11] W.P. Hooper, H. Gerber, Monte Carlo simulations of laser-generated sea surface anaesthesia and pain management, Drugs 70 (6) (2010) 761–791.
aureole, Appl. Opt. 27 (24) (1988) 5111–5118. [35] K. Shi, B. Xue, Y. Jia, L. Yuan, R. Han, F. Yang, J. Peng, Z. Qian, Sustained co-
[12] K. Lobo, C. Donado, L. Cornelissen, J. Kim, R. Ortiz, R.W. Peake, M. Kellogg, M. delivery of gemcitabine and cis-platinum via biodegradable thermo-sensitive
E. Alexander, D. Zurakowski, K.E. Kurgansky, J. Peyton, A. Bilge, K. Boretsky, M. hydrogel for synergistic combination therapy of pancreatic cancer, Nano Res. 12
E. McCann, C.B. Berde, J. Cravero, A phase 1, dose-escalation, double-blind, block- (6) (2019) 1389–1399.
randomized, controlled trial of safety and efficacy of Neosaxitoxin alone and in [36] K. Shi, Y.L. Wang, Y. Qu, J.F. Liao, B.Y. Chu, H.P. Zhang, F. Luo, Z.Y. Qian,
combination with 0.2% bupivacaine, with and without epinephrine, for cutaneous Synthesis, characterization, and application of reversible PDLLA-PEG-PDLLA
anesthesia, Anesthesiology 123 (4) (2015) 873–885. copolymer thermogels in vitro and in vivo, Sci. Rep. 6 (2016), 19077.
[13] C.M. Santamaria, A. Woodruff, R. Yang, D.S. Kohane, Drug delivery systems for [37] Y. Pan, Y. Xiao, Y. Hao, K. Shi, M. Pan, Z. Qian, An injectable mPEG-PDLLA
prolonged duration local anesthesia, Mater. Today 20 (1) (2017) 22–31. microsphere/PDLLA-PEG-PDLLA hydrogel composite for soft tissue augmentation,
[14] M.A. Gropper, R.D. Miller, L.I. Eriksson, L.A. Fleisher, J.P. Wiener-Kronish, N. Chin. Chem. Lett. 33 (5) (2022) 2486–2490.
H. Cohen, K. Leslie, Miller’s Anesthesia, 2-volume Set E-Book, Elsevier Health [38] R.F. Padera, J.Y. Tse, E. Bellas, D.S. Kohane, Tetrodotoxin for prolonged local
Sciences, 2019. anesthesia with minimal myotoxicity, Muscle Nerve 34 (6) (2006) 747–753.
[15] C.R. Ries, R. Pillai, C.C. Chung, J.T. Wang, B.A. MacLeod, S.K. Schwarz, QX-314 [39] Q. Yin, Y. Zhang, R. Lv, D. Gong, B. Ke, J. Yang, L. Tang, W. Zhang, T. Zhu, A fixed-
produces long-lasting local anesthesia modulated by transient receptor potential dose combination, QXOH/levobupivacaine, produces long-acting local anesthesia
vanilloid receptors in mice, Anesthesiology 111 (1) (2009) 122–126. in rats without additional toxicity, Front. Pharmacol. 10 (243) (2019).
[16] Y. Zhang, J. Yang, Q. Yin, L. Yang, J. Liu, W. Zhang, QX-OH, a QX-314 derivative [40] Y. Zhang, D. Gong, Q. Zheng, J. Liu, W. Zhang, LC-MS/MS method for preclinical
agent, produces long-acting local anesthesia in rats, Eur. J. Pharmaceut. Sci. 105 pharmacokinetic study of QX-OH, a novel long-acting local anesthetic, in sciatic
(2017) 212–218. nerve blockade in rats, J. Pharm. Biomed. Anal. 146 (2017) 161–167.
[17] A.J. Rodriguez-Navarro, N. Lagos, M. Lagos, I. Braghetto, A. Csendes, J. Hamilton, [41] T.J. Brennan, E.P. Vandermeulen, G.F. Gebhart, Characterization of a rat model of
C. Figueroa, D. Truan, C. Garcia, A. Rojas, V. Iglesias, L. Brunet, F. Alvarez, incisional pain, Pain 64 (3) (1996) 493–501.
Neosaxitoxin as a local anesthetic: preliminary observations from a first human [42] J.C. Shah, M. Maniar, pH-Dependent solubility and dissolution of bupivacaine and
trial, Anesthesiology 106 (2) (2007) 339–345. its relevance to the formulation of a controlled release system, J. Contr. Release 23
[18] M. Banerjee, A. Baranwal, S. Saha, A. Saha, T. Priestley, EN3427: a novel cationic (3) (1993) 261–270.
aminoindane with long-acting local anesthetic properties, Anesth. Analg. 120 (4) [43] K.H. Diehl, R. Hull, D. Morton, R. Pfister, Y. Rabemampianina, D. Smith, J.
(2015) 941–949. M. Vidal, C. van de Vorstenbosch, A, European Federation of Pharmaceutical
[19] B. Wang, S. Wang, Q. Zhang, Y. Deng, X. Li, L. Peng, X. Zuo, M. Piao, X. Kuang, Industries, M. European Centre for the Validation of Alternative, A good practice
S. Sheng, Y. Yu, Recent advances in polymer-based drug delivery systems for local guide to the administration of substances and removal of blood, including routes
anesthetics, Acta Biomater. 96 (2019) 55–67. and volumes, J. Appl. Toxicol. 21 (1) (2001) 15–23.
[20] R. Ohri, J.C. Wang, P.D. Blaskovich, L.N. Pham, D.S. Costa, G.A. Nichols, W. [44] D.N. Franz, R.S. Perry, Mechanisms for differential block among single myelinated
P. Hildebrand, N.L. Scarborough, C.J. Herman, G.R. Strichartz, Inhibition by local and non-myelinated axons by procaine, J. Physiol. 236 (1) (1974) 193–210.
bupivacaine-releasing microspheres of acute postoperative pain from hairy skin [45] J.A. DeLeo, M.D. Rutkowski, Gender differences in rat neuropathic pain sensitivity
incision, Anesth. Analg. 117 (3) (2013) 717–730. is dependent on strain, Neurosci. Lett. 282 (3) (2000) 197–199.
[21] E.V. Ramos Campos, N.F. Silva de Melo, V.A. Guilherme, E. de Paula, A.H. Rosa, D. [46] D.G. Littlewood, D.B. Scott, J. Wilson, B.G. Covino, Comparative anaesthetic
R. de Araujo, L.F. Fraceto, Preparation and characterization of poly(epsilon- properties of various local anaesthetic agents in extradural block for labour, Br. J.
caprolactone) nanospheres containing the local anesthetic lidocaine, Anaesth. 49 (1) (1977) 75–79.
J. Pharmaceut. Sci. 102 (1) (2013) 215–226. [47] J.S. Khan, D.I. Sessler, M.T.V. Chan, C.Y. Wang, I. Garutti, W. Szczeklik, A. Turan,
[22] M. Burbridge, R.A. Jaffe, Exparel(R): a new local anesthetic with special safety J.W. Busse, D.N. Buckley, J. Paul, M. McGillion, C. Fernandez-Riveira, S.
concerns, Anesth. Analg. 121 (4) (2015) 1113–1114. K. Srinathan, H. Shanthanna, I. Gilron, M. Jacka, P. Jackson, J. Hankinson,
[23] J.W. Hu, M.W. Yen, A.J. Wang, I.M. Chu, Effect of oil structure on cyclodextrin- P. Paniagua, S. Pettit, P.J. Devereaux, Persistent incisional pain after noncardiac
based Pickering emulsions for bupivacaine topical application, Colloids Surf. B surgery: an international prospective cohort study, Anesthesiology 135 (4) (2021)
Biointerfaces 161 (2018) 51–58. 711–723.
[24] K.S. Oh, C. Hwang, H.Y. Lee, J.S. Song, H.J. Park, C.K. Lee, I. Song, T.H. Lim,
Preclinical studies of ropivacaine extended-release from a temperature responsive

13

You might also like