Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

JID: ACTBIO

ARTICLE IN PRESS [m5G;August 2, 2021;15:11]


Acta Biomaterialia xxx (xxxx) xxx

Contents lists available at ScienceDirect

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

Full length article

Evaluation of the in vitro cytotoxicity and modulation of the


inflammatory response by the bioresorbable polymers
poly(D,L-lactide-co-glycolide) and poly(L-lactide-co-glycolide)
Lucy Geddes a, Efrosyni Themistou b, James F. Burrows c, Fraser J. Buchanan a,
Louise Carson c,∗
a
School of Mechanical and Aerospace Engineering, Queens University Belfast, Ashby Building, Belfast, BT9 5AG, Northern Ireland, UK
b
School of Chemistry and Chemical Engineering, Queens University Belfast, David Keir Building, Belfast, BT9 5AG, Northern Ireland, UK.
c
School of Pharmacy, Queens University Belfast, Belfast, BT9 7BL, Northern Ireland, UK

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

Article history: Bioresorbable polymers composed of poly(D,L-lactide-co-glycolide) (PDLLGA) and poly(L-lactide-co-


Received 3 December 2020 glycolide) (PLLGA) have become increasingly popular for the preparation of bone substitute constructs.
Revised 21 July 2021
However, there are reports of a delayed inflammatory reaction occurring months or years after implanta-
Accepted 22 July 2021
tion. Due to the long polymer degradation times, in vitro tests carried out at physiological temperature,
Available online xxx
37°C, tend to assess only the short-term biocompatibility of these materials. The aim of this work is to de-
Keywords: velop an in vitro protocol that can be used to assess the long-term cytotoxicity of bioresorbable polymers
Poly(lactide-co-glycolide) in a time efficient manner. This study used a previously developed and validated accelerated degradation
Biocompatibility protocol to obtain samples of PDLLGA and PLLGA at increasing levels of degradation. Samples were then
Cytotoxicity applied to standard ISO 10993-5 direct contact cytotoxicity testing and it was found that PDLLGA samples
Quasi Vivo showed increasing levels of cytotoxicity at the later stages of degradation, with PLLGA samples demon-
Lactate
strating significantly less cytotoxic behaviour. Following concern that accumulation of acidic degradation
Hydroxycarboxylic acid receptor 1
HCA1
products in a closed multi-well culture environment could overestimate cytotoxicity, we developed and
validated a new dynamic flow culture methodology, for testing the cytotoxicity of these degradable ma-
terials, by adapting a commercial “organ on a chip” flow culture system, Quasi Vivo®. In addition to
cytotoxicity testing, we have carried out profiling of inflammatory cytokines released by cells in response
to degraded PDLLGA and PLLGA, and have suggested mechanism by which lactide-based bioresorbable
materials could modulate the inflammatory response through the G-protein coupled receptor (GPCR), hy-
droxycarboxylic acid receptor 1 (HCA1 ).

Statement of significance

Bioresorbable materials naturally disintegrate over time when implanted into the body. They are often
used to make screws and clips for repair of broken bones. Unfortunately, some patients can react badly
to the material, resulting in painful inflammation. Biomaterials scientists are interested in developing
materials that are more compatible with the body. However, it is very difficult to predict the long-term
compatibility of bioresorbable materials in the lab. In our study, we have developed a method that will
allow us to study the effects of the materials as they continue to break down. This will help us under-
stand why the materials may cause inflammation, and will support research into the development of new
and improved materials for bone repair.
© 2021 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

1. Introduction

Bioresorbable polymers composed of poly(lactide) (PLA),



Corresponding author. poly(glycolide) (PGA) and their related copolymers are commonly
E-mail address: l.carson@qub.ac.uk (L. Carson). used in orthopaedic applications to help regenerate and repair

https://doi.org/10.1016/j.actbio.2021.07.049
1742-7061/© 2021 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

Please cite this article as: L. Geddes, E. Themistou, J.F. Burrows et al., Evaluation of the in vitro cytotoxicity and modulation of the
inflammatory response by the bioresorbable polymers poly(D,L-lactide-co-glycolide) and poly(L-lactide-co-glycolide), Acta Biomaterialia,
https://doi.org/10.1016/j.actbio.2021.07.049
JID: ACTBIO
ARTICLE IN PRESS [m5G;August 2, 2021;15:11]

L. Geddes, E. Themistou, J.F. Burrows et al. Acta Biomaterialia xxx (xxxx) xxx

injured bone tissue. Upon implantation, polymers degrade via ucts, the acidic environment created by the release of acidic by-
hydrolytic degradation, gradually transferring the load to the bone products, the geometry of the implant and the implant location
tissue, with metabolic pathways used to excrete the degradation [21–24]. However, it is difficult to determine the aetiology of this
products from the body [1–3]. The main benefit of bioresorbable response as it is difficult to isolate the factors that contribute to
polymers is the ability to tailor their physicochemical and me- this response.
chanical properties to meet the desired needs of the application. Medical devices must undergo a series of tests to ensure they
Poly(L-lactide) (PLLA) is commonly used for orthopaedic appli- are safe for clinical use, with these tests regulated by the Interna-
cations, however, reports of long polymer degradation times, tional Organisation for Standardisation (ISO) [25,26]. Cytotoxicity
in excess of 5 years, has led to research into faster degrading analysis is required for every medical device and is generally the
polymers, allowing replacement by regenerating tissue over more first step of in vitro biocompatibility testing. These tests provide a
realistic timeframes [4–8]. There is growing interest into the highly effective method to determine the cytotoxicity of a material
copolymerisation of poly(D,L-lactide) (PDLLA) and PLLA with PGA or substance on living cells, however, their use is limited as they
forming poly(D,L-lactide-co-glycolide) (PDLLGA) and poly(L-lactide- cannot be used to determine the cause of cellular death. Testing
co-glycolide) (PLLGA). The inherent properties of the amorphous of the in vitro immune response at the early stages of biomaterial
PDLLGA and semi-crystalline PLLGA, give the copolymers different development is currently not incorporated within standard proce-
characteristics and their mechanical, physiochemical and degrada- dures. Gaining insight into the in vitro cell response to biomaterials
tion properties can be controlled by altering their monomer unit would allow for early detection and prediction of potential adverse
ratios. reactions. Protein-based arrays can be used as a useful method to
The in vivo host response to implanted biomaterials is impor- evaluate how biomaterials interact with different cells within the
tant to be considered during the development of bioresorbable FBR [27]. The profiling of cytokines expressed by cells in contact
polymers. Upon implantation, a host reaction is triggered and re- with biomaterials can be determined using antibody arrays and an
sults in a cascade of regulated events. Following implantation, a enzyme-linked immunosorbent assay (ELISA).
provisional matrix is formed by protein adsorption on the sur- In vitro studies play an important role in the prediction of in
face of the biomaterial and is rich in cytokines, chemoattractants vivo behaviour by allowing rapid screening of biomaterials prior to
and growth factors. Acute inflammation follows and is charac- animal trials. However, in vitro tests tend to assess the short-term
terised by the presence of neutrophils, whose role involves the effects, with the long-term biocompatibility assessed using in vivo
removal of cellular debris via phagocytosis and the expression of trials. The development of extended in vitro biocompatibility test-
cytokines, which are involved in the recruitment of macrophages, ing for degradable biomaterials would be beneficial, as it would
such as macrophage inflammatory protein-1α (MIP-1α ) and mono- allow the early identification of potential issues before progressing
cyte chemoattractant protein-1 (MCP-1) [9–13]. This phase is fol- to animal or clinical studies and would also advance the under-
lowed by chronic inflammation, where inflammatory cells, such as standing of why adverse reactions occur. Despite clinical studies
monocytes, are recruited to the site of implantation. The acute and demonstrating adverse reactions to bioresorbable polymers, there
chronic inflammation phases are relatively short, usually lasting no are few in vitro studies that corroborate these results. The aim of
longer than two weeks. The resolution of these phases leads to this work is to develop an in vitro protocol that can be used to as-
the formation of granulation tissue, which can be identified by the sess the cytotoxicity of bioresorbable polymers in a time efficient
presence of macrophages, fibroblasts, and the proliferation of small manner.
blood vessels. The foreign body reaction (FBR) is identified by the Current in vitro ISO standard tests for cytotoxicity are carried
presence of macrophages and foreign body giant cells at the sur- out at 37°C in cell culture media (or other suitable diluent), in
face of the implant, together with the components of the gran- closed, static multi-well culture vessels. These tests only assess the
ulation tissue. The cellular composition of the FBR is largely de- short-term biocompatibility, whereas late-stage degradation, can
termined by the form, topography, and surface-to-volume ratio of take months to occur at physiological temperature. This timescale
the implanted biomaterial and can persist for its lifetime. The final is incompatible with cell culture-based assays.
stage of the healing process is fibrous capsule formation surround- In this study, we have used a previously developed and vali-
ing the biomaterial. This acts as protective barrier isolating the FBR dated accelerated degradation protocol to obtain polymer samples
and the biomaterial from the host tissue [13,14]. This is the body’s at increasing levels of degradation [28]. This utilised an elevated
natural healing mechanism and is essential for the successful in- temperature of 47°C, i.e. 10°C above physiological temperature.
tegration of the implanted biomaterial with its host environment. This previous study also compared elevated temperatures of 57°C
The progression of these stages occurs via complex cellular inter- and 70°C and concluded 47°C was the optimum temperature owing
actions, with crosstalk between different cascade systems control- to the fact that it was below the polymer’s Tg, and produced con-
ling the cellular response and behaviour. Development of a better sistent results over practicable timescales. Briefly, polymers were
understanding of these complex interactions will enhance the pro- incubated in sterile phosphate buffered saline at 47°C, and samples
gression of suitable bioresorbable polymers for use as medical de- were removed at predetermined time points, informed by physico-
vices. chemical characterisation. The samples were then applied to stan-
Biocompatibility is used to describe the desired interaction of dard ISO 10993-5 direct contact cytotoxicity testing [29]. Addition-
a biomaterial with its host environment, whereby the biomate- ally, we have developed a dynamic flow culture methodology for
rial or degradation products should not cause any adverse reac- testing these degradable materials, following concern that accumu-
tions or cytotoxicity after implantation [15]. PLA, PGA, and their lation of acidic degradation products in closed multi-well culture
related copolymers are widely considered biocompatible, however, could overestimate cytotoxicity. In an in vivo physiological environ-
throughout the literature there is evidence of a delayed inflamma- ment, diffusion of degradation products will occur, and there will
tory reaction occurring months or years after implantation. These be an exchange of tissue fluid in the peri-implant microenviron-
reactions generally present as a non-specific FBR and can range ment. To replicate the in vivo environment and increase physiolog-
in severity [5,16–20]. The occurrence of this reaction is not well ical relevance, we have adapted a commercial “organ on a chip”
understood as it is not specific to polymer type, implant loca- flow culture system, Quasi Vivo®, for testing the polymer samples.
tion or subject [5]. Numerous reasons for the reported incom- In addition to cytotoxicity testing, we have carried out profil-
patibility have been hypothesised, which include the presence of ing of inflammatory cytokines released by cells in response to de-
crystalline particle debris, the accumulation of degradation prod- graded PDLLGA and PLLGA, and have proposed a suggested mecha-

2
JID: ACTBIO
ARTICLE IN PRESS [m5G;August 2, 2021;15:11]

L. Geddes, E. Themistou, J.F. Burrows et al. Acta Biomaterialia xxx (xxxx) xxx

nism by which lactide-based bioresorbable materials could module The number average molecular weight (Mn ) of the samples was
the inflammatory response through the G-protein coupled receptor determined by gel permeation chromatography (GPC).
(GPCR), Hydroxycarboxylic acid receptor 1 (HCA1 ).
2.5. Cell culture
2. Materials and methods

L-929 murine fibroblasts (ATCC CCL-1) were maintained in Ea-


2.1. Materials
gle’s minimum essential medium (MEM) containing 10% foetal
bovine serum (FBS) and 50 U/mL penicillin/streptomycin (pen
PDLLGA 85:15 (PURASORB PDLG 8531, Corbion Biomaterials, the
strep). RAW264.7 (ATCC TIB-71) murine macrophages were main-
Netherlands) and PLLGA 85:15 (PURASORB PLG 8531, Corbion Bio-
tained in Dulbecco’s minimum essential medium (DMEM) contain-
materials, the Netherlands) in pellet form were used for this study.
ing 10% FBS and 50 U/mL pen strep. Both cell lines were cultured
Materials were stored in a freezer (-20°C) in sealed packaging until
under standard conditions of 37°C and 5% CO2 .
use.

2.2. Sample preparation 2.6. Cell viability assay

PDLLGA and PLLGA pellets were compression moulded (Collins 2.6.1. Static conditions
P200P), at 180°C for 6.5 minutes, into sheets with a volume of Cells were cultured in a 24-well plate for 24 hours in 1 mL
100 × 100 × 1 mm3 . PLLGA compression moulded sheets were of media to allow for a near confluent monolayer. The degraded
then annealed in an oven for 4 hours at 100°C (PDLLGA was not samples, in the transwell inserts, were placed into the wells of
annealed, as this was considered unnecessary due to its amor- the tissue culture plate, and cells incubated for a further 24 hours
phous nature). A laser cutting machine (FB 1800 50W) was used at 37°C. After this time, the transwell inserts were removed from
to cut the PDLLGA and PLLGA sheets into disc-shaped samples with the 24-well plate. The supernatants were removed from the cells,
an 8mm diameter. Samples were sterilised using electron beam ra- placed into microcentrifuge tubes, and stored at -80°C for later
diation (Steris, Ireland), with a dosage of 20 kGy. For experimen- analysis (i.e. antibody arrays and ELISA analysis described below).
tation under static cell culture conditions positive control mate- 500 μL of fresh MEM or DMEM were then added to the cells. All
rial was prepared by swelling poly(2-hydroxyethyl methacrylate) experimental steps were carried out under aseptic conditions in a
(pHEMA) in a solution of 0.1% benzalkonium chloride (BAK) in Class II microbiological safety cabinet. All cell culture experiments
phosphate buffered saline (PBS), and sterilising by autoclave. BAK were carried out in triplicate.
is a known cytotoxic substance and hydrogel polymers soaked in
BAK have been validated as a suitable positive control material for
cytotoxicity testing [30]. The negative controls were non-degraded 2.6.2. Development of flow conditions for cell culture using quasi
PDLLGA samples and non-degraded PLLGA samples. vivo® QV500 system
The Quasi Vivo® QV500 system (Kirkstall) is a flow system
2.3. In vitro degradation that can be used to culture cells in a way that better reflects the
dynamic nature of the physiological environment. This approach
PDLLGA and PLLGA samples were separated into three groups, utilises interconnected chambers with cell culture media pumped
the first group was used for characterisation, the second for through the system, allowing cells to be cultured under flow condi-
tests on L929 murine fibroblasts and the third for tests on tions. The components of the QV500 system were connected under
RAW264.7 murine macrophages. Under sterile conditions, samples sterile conditions in a Class II microbiological safety cabinet. The
were placed into transwell inserts (Millicell, PIHP01250), immersed system is designed to recirculate media; however, this was adapted
in 10 mL of PBS buffer (pH 7.4) and degraded in an oven at 47°C. for the current study to allow for a single pass of media though the
PDLLGA sample were degraded for 5, 7, 10 and 12 days and PLLGA system. Nine Quasi Vivo® chambers were connected in series and
was degraded for 28, 42, 56 and 70 days. each sample examined in triplicate (Fig. 1). A peristaltic pump (SP-
minipump compact, Shenchen Baoding) with variable speed con-
2.4. Characterisation trol was used with the QV500 system. The pump was calibrated
in order to achieve a flowrate of 75 μL/min; this flowrate was se-
Polymers were characterised in a previous study by the au- lected as it was used by Nithiananthan et al. [31] when using the
thors [28]. PDLLGA and PLLGA were assessed for changes in the QV500 system with fibroblast cell lines.
pH of the degradation media (PBS buffer), percentage mass loss,
percentage swelling and molecular weight loss during degradation 2.6.2.1. Flow conditions using quasi vivo® QV500 system. Prior to
at 47°C. Briefly, the pH of the degradation media (PBS buffer) was use, glass coverslips (13 mm diameter) were sterilised in ethanol,
recorded at each time point. Prior to degradation, the initial mass rinsed in PBS, primed in MEM, and placed into a 24-well plate.
of the samples was recorded. At each time point the samples (n=5) L929 fibroblasts were cultured, in static conditions, on coverslips
were retrieved from the PBS buffer and rinsed with distilled wa- in 1 mL of MEM at 37°C with 5% CO2 , for 24 hours. As per man-
ter. Surface moisture was removed, and the weight was recorded ufacturer’s instructions, before starting the experiment, the QV500
(wet mass). The dry mass was then determined after samples were system was washed with PBS buffer for 20 minutes, then primed
dried in a vacuum oven for 72 hours at 30°C and 600 mmHg. with cell culture media (MEM) for 20 minutes, with the peristaltic
Eqs. 1 and 2 were used to determine the percentage mass loss and pump set to its maximum flowrate. The media was then removed
swelling of the samples, respectively. from the system. Fig. 2 shows step-by-step images of the next
m0 − md stages of the experiment, wherein material samples are loaded
% Mass Loss = x100 (1)
m0 into each QV500 chamber. The closed system is then transferred
to the incubator and operated with a flowrate of 75 μL/min for 24
mw − md
% Swel l ing = x100 (2) hours. After this time, coverslips were removed from the system
md and placed into a 24-well plate, with 500 μL of fresh MEM added,
Where, m0 = initial mass; md = dry mass and mw = wet mass in preparation for the MTT cytotoxicity assay.

3
JID: ACTBIO
ARTICLE IN PRESS [m5G;August 2, 2021;15:11]

L. Geddes, E. Themistou, J.F. Burrows et al. Acta Biomaterialia xxx (xxxx) xxx

Fig. 1. Schematic representation of QV500 system (A) and real image of QV500 system (B).

Fig. 2. Step-by-step images of Quasi-Vivo® system set-up. The empty chamber (A) is opened and a coverslip containing cultured L929 fibroblasts is placed into the chamber
(B). A transwell insert, either empty (cells only) or containing the negative control or sample is then placed on top of the cells (C), 1 mL of MEM is added (D) and the
chamber is closed (E). This is repeated until all chambers contain samples.

2.6.3. MTT cytotoxicity assay contact with PDLLGA degraded for 10 days and PLLGA degraded
The viability of the cells is determined by the ability of the for 42 days. These time points were selected at which the mate-
mitochondrial enzyme succinate dehydrogenase to metabolically rials began to show slight reduction in cell viability. The controls
reduce the yellow water soluble 3-(4,5-dimethylthiazol-2-yl)-2,5- used for this study were the supernatants collected from the L929
diphenyltetrazoliumbromid (MTT) into blue/ violet formazan crys- fibroblasts and RAW264.7 macrophages cultured in cell medium
tals. The intensity of the blue/ violet colour is directly related to only. Supernatants from triplicate samples were pooled and pre-
the number of viable cells [29,32]. Following the treatments de- pared according to the manufacturer’s array procedure. The cy-
scribed in Sections 2.6.1 and 2.6.2, 100 μL of a filter-sterilised MTT tokine array was conducted according to the manufacturer’s in-
(5 mg/mL) solution was added to the cells in the 24-well plates. structions and chemiluminescence was detected using the Molec-
The plates were wrapped in tin foil to protect from light and ular Imager® ChemiDocTM XRS+ imaging system (BioRad). Images
placed in the incubator at 37°C for 2 hours. Media was then aspi- were analysed using Image Studio Lite version 5.2 software.
rated, and the formazan crystals formed were dissolved in 500 μL
of dimethyl sulfoxide (DMSO). The optical density was measured at
a wavelength of 570 nm using a FLUOstar Omega microplate reader 2.8. ELISA
(BMG Labtech). Eq. 3 was used to monitor the changes in cell vi-
ability. As stated by ISO 10993-5, if the cell viability is less than MCP-1 and MIP-1α , were selected for continued analysis using
70% the material has cytotoxic potential. The lower the percentage the DuoSet® ELISA Development System (R&D Systems), based on
viability the greater the cytotoxic potential of the material. To cal- the results of the antibody array, in Section 3.5. Cell conditioned
culate the percentage viability all the sample values are relative to media, collected under static conditions (Section 2.6.1), were anal-
a “cells only” blank control, as per Equation 3. ysed. These samples included the supernatants collected from L929
fibroblasts and RAW264.7 macrophages in contact with PDLLGA
ODs
Cel l V iabil ity (% ) = × 100% (3) non-degraded and degraded for 7, 10 and 12 days at 47°C and
ODb PLLGA non-degraded and degraded for 28, 42, 56 and 70 days at
where: ODs is the optical density of the test sample extracts and 47°C. The controls used were the supernatants collected from the
ODb is the optical density of the “cells only” blanks. L929 fibroblasts and RAW264.7 macrophages cultured in cell cul-
ture medium only. The samples collected from the L929 fibrob-
2.7. Antibody array lasts were analysed for MCP-1 and the samples collected from the
RAW264.7 macrophages were analysed for MIP-1α . The ELISA was
The relative expression levels of 40 mouse cytokines from carried out according to the manufacturer’s instructions. All ELISA
selected samples were determined using a Proteome ProfilerTM analysis was performed in duplicate wells, on three experimental
Mouse Cytokine Array, Panel A (R&D Systems). Four samples of cell replicates. The optical density was determined using a FLUOstar
conditioned media were selected based on the results of the MTT Omega microplate reader (BMG Labtech), measured at wavelengths
cytotoxicity assay, in Section 3.2. These included the supernatants of 450 nm and 570 nm. Wavelength correction was calculated by
collected from L929 fibroblasts and RAW264.7 macrophages after subtraction of the reading at 570 nm from the reading at 450 nm.

4
JID: ACTBIO
ARTICLE IN PRESS [m5G;August 2, 2021;15:11]

L. Geddes, E. Themistou, J.F. Burrows et al. Acta Biomaterialia xxx (xxxx) xxx

The calibration curve was generated using four-parameter logistic broblasts and macrophages. High cell viability values, between 86
(4-PL) curve fit. – 106%, were obtained in both cell lines for the 5- and 7- days
samples, which showed no significant difference to the negative
2.9. GPCR (HCAR1 ) activation and calcium mobilisation assays control. However, a significant decrease compared to the negative
control, was observed after 10 days, as viability was reduced to
GPCR activation was monitored via calcium signalling using the 53% and 14% in the fibroblasts and macrophages, respectively. Af-
proprietary Fluo-4 Direct TM Calcium Assay Kit. This kit contains ter 12 days, viability was reduced to 0% in both cell lines, therefore
a Ca2+ indicator, which is loaded into the cells and produces a demonstrating a high degree of cytotoxicity. When PLLGA degraded
large fluorescence increase upon binding Ca2+ . To prevent extru- for 42- and 70- days was placed in contact with the fibroblasts,
sion of the indicator out of the cell by organic anion transporters, cell viability reduced to 58% and 63%, respectively (Fig. 4b). These
probenecid is included in the kit formulation, and this helps to values are below the 70% viability threshold that is stated by ISO
reduce the baseline signal. Media removal and washing steps are 10993-5, below which the material displays cytotoxic potential. In
not required with the Fluo-4 Direct TM Calcium Assay Kit, as back- contrast, when PLLGA was placed onto the macrophages (Fig. 4b),
ground fluorescence attributable to the growth media is eliminated there was no cytotoxic effect observed at any time point, with high
by the addition of a suppression dye. cell viability values between 84–112%.
Cells were seeded at a density of 5 × 104 cells/well in black
sterile 96-well plates with optical bottom, in 100 μl of the ap- 3.3. Cytotoxicity and polymer degradation
propriate culture media, and allowed to recover for 24 hours. The
Fluo-4 Direct TM indicator was reconstituted as per kit instructions The degradation characteristics (change in pH of the degrada-
under aseptic conditions and added to each well in an equal vol- tion media, percentage mass loss, percentage swelling and change
ume to culture media. The plate was wrapped in aluminium foil in the number averaged molecular weight (Mn ) value) of PDLLGA
to protect from light, then incubated for 30 minutes at 37°C, and a and PLLGA, degraded at 47°C, were measured [28] and compared
further 30 minutes at room temperature. to the MTT assay results in Fig. 4. PDLLGA and PLLGA degrade via
To confirm GPCR receptor activation in response to lactate (and a mechanism of bulk degradation, which can be observed when
the presence of HCA1 ) 15 μM sodium L-lactate was added to in- degradation proceeds in two distinct phases; firstly, a reduction in
dicator loaded cells, and the fluorescence was monitored using a molecular weight, followed by mass loss. Fig. 5 shows that after 5
BMG Fluostar Optima® spectrofluorimeter, at wavelengths of 494 days, there was a significant reduction in the Mn value of PDLLGA
nm excitation and 516 nm emission. The response to 10 μL samples from 27 800 g/mol to 3380 g/mol indicating significant change in
of the polymer degradation media (as described in Section 2.3) the degree of polymerization of the polymer and break down of
was also evaluated, with degradation time points of 12 days for its chains to oligomers and monomers, with a mass loss of only 6%
PDLLGA, and 70 days for PLLGA selected for examination. This anal- after 12 days. There was a slight decrease in the pH of the degra-
ysis was carried out in triplicate. Sterile, calcium free PBS was used dation media from pH 7.4 to 6.3 after 12 days. However, there was
as a vehicle control and was not found to have any effect on recep- significant polymer swelling after 10 days (97%) and 12 days (57%),
tor activation (Supplementary Information, Figure S1). which corresponds to the reduction in cell viability in both the fi-
broblasts and macrophages at these time points. Fig. 6 shows that
2.10. Statistics
the Mn of PLLGA decreased steadily with degradation time from
135 0 0 0 g/mol to 22 70 0 g/mol after 42 days and to 20 0 0 g/mol af-
Statistical analysis was carried out using GraphPad Prism 5.0
ter 56 days, indicating that the high molecular weight chains have
software. The results are expressed as the mean ± standard devi-
almost completely broken down to shorter chains. There was ini-
ation. Statistical analysis for the static conditions MTT assay and
tially a small decrease in mass, 2%, after 42 days, followed by a
ELISA was performed using a one-way ANOVA with a post-hoc
larger reduction in mass, 19%, after 56 days, and a further reduc-
Tukey analysis. Statistical significance was set at p < 0.05. Statisti-
tion to 38% after 70 days. There was a slight decrease in the pH
cal analysis for the Quasi Vivo flow conditions was performed us-
of the degradation media from pH 7.4 to 6.0 after 42 days, and
ing a t-test. Statistical significance was set at p < 0.05.
a significant drop to pH 2.8 after 56 days. Swelling in PLLGA did
3. Results not occur immediately but increased with degradation time with a
swelling ratio value of 43% after 70 days, with a corresponding cell
3.1. Visual inspection viability of 63% in the L929 fibroblasts and 103% in the RAW264.7
macrophages.
PDLLGA underwent significant changes to its appearance dur-
ing degradation and changes occurred without considerable mass 3.4. MTT cytotoxicity assay using quasi vivo® flow conditions
loss, where, after 12 days, only 6% mass was lost from the samples
(Fig. 3a). After 5 days at 47°C the samples became visibly damaged, Fig. 7 compares the viability of fibroblasts in contact with
after 10 days the samples appeared visibly swollen with a hard PDLLGA degraded for 10 days and PLLGA degraded for 42 days, un-
exterior and viscous liquid interior, and after 12 days the sam- der static and flow conditions using the Quasi Vivo® QV500 sys-
ples were a viscous liquid. In contrast, the appearance of PLLGA tem. The samples were selected based on the results of the MTT
(Fig. 3b) remained the same until 56 days at 47°C, where the sam- assay, in Section 3.2, as after 10 days and 42 days, PDLLGA and
ples changed from a translucent off-white colour to opaque and PLLGA, respectively, began to show cytotoxic potential towards the
bright white. Throughout degradation, the samples appeared volu- fibroblasts. Fig. 7a shows that PDLLGA was cytotoxic after 10 days
metrically unchanged, however, when mass loss occurred, after 56 using both static and flow conditions, with cell viability reduced
days, the samples became fragile and disintegrated into a powder to 46% and 16%, respectively. Significant differences were observed
on handling. between the control and 10 days sample for both sets of condi-
tions. PLLGA showed cytotoxic potential, under static conditions,
3.2. MTT cytotoxicity assay for static conditions after 42 days, with cell viability reduced to 58% (Fig. 7b). How-
ever, cell viability was improved to 69% using the Quasi Vivo flow
The direct cytotoxicity assay (conducted as per ISO 10993-5) on conditions and the results showed no significant difference to the
pre-degraded PDLLGA (Fig. 4a) showed similar trends in both fi- control, suggesting that after 42 days degradation, the material

5
JID: ACTBIO
ARTICLE IN PRESS [m5G;August 2, 2021;15:11]

L. Geddes, E. Themistou, J.F. Burrows et al. Acta Biomaterialia xxx (xxxx) xxx

Fig. 3. Samples appearance and mass loss of a) PDLLGA and b) PLLGA after degradation at 47°C and drying.

Fig. 4. Cell viability of L929 fibroblasts and RAW264.7 macrophages versus degradation time of a) PDLLGA and b) PLLGA at 47°C. (∗ indicates significant difference to the
negative control (p<0.05), red line represents ISO 10993-5 cytotoxicity threshold, at 70% viability relative to blank control (cells only); Percentage cell viability values of the
samples may be greater than 100% in cases where the sample cells proliferate more than the “cells only” blank control).

showed less potential to induce cytotoxic effects under flow condi- MIP-1α , macrophage inflammatory protein-1β (MIP-1β ), SDF-
tions than those observed under the standard static experimental 1, interferon-inducible protein-10 (IP-10), interleukin-1ra (IL-1ra),
conditions. MCP-1, TNF-α and C-X-C motif chemokine ligand 9 (CXCL9). How-
ever, only MIP-1α and SDF-1 were detected in the treated sam-
3.5. Antibody array analysis ples. The results shows a pronounced downregulation of the rel-
ative expression levels of MIP-1α , MIP-1β , IP-10, IL-1ra, MCP-
Fig. 8a shows the relative expression levels of the inflam- 1 and TNF-α . Fig. 8b shows the relative expression levels of
matory cytokines detected in conditioned media from fibrob- the cytokines released from the fibroblasts and macrophages af-
lasts and macrophages after contact with PDLLGA degraded for ter contact with PLLGA degraded for 42 days. In the fibroblasts,
10 days. For fibroblasts, nine cytokines were detected in the four cytokines were detected in the control and treated sam-
control, including MCP-1, tissue inhibitor of metalloproteinase- ples, these include MCP-1, TIMP-1, M-CSF and SDF-1. The re-
1 (TIMP-1), macrophage colony-stimulating factor (M-CSF), stro- sults show an upregulation of MCP-1, a downregulation of TIMP-
mal cell-derived factor-1 (SDF-1), keratinocyte chemoattractant 1 and SDF and no change in M-CSF in the treated samples. In
(KC), interleukin-1α (IL-1α ), interferon-inducible T cell alpha the macrophages, four cytokines were also detected in the con-
chemoattractant (I-TAC), tumour necrosis factor-α (TNF-α ), inter- trol and treated samples, which include MIP-1α , SDF-1, IL-1ra and
feron gamma (IFN-γ ), and eleven cytokines were detected in MIP-1β . The results show that there was an increase in the rel-
the treated samples, with the addition of interleukin-17 (IL-17) ative expression levels of MIP-1α , SDF-1, IL-1ra and MIP-1β in
and interleukin-7 (IL-7) to those already observed in the con- the treated samples compared to the control. In both PDLLGA
trol. The results showed a downregulation in the relative ex- and PLLGA, MCP-1 was the most abundant cytokine in the fi-
pression levels of MCP-1 and KC in the treated samples. For broblasts and MIP-1α was the most abundant cytokine in the
the macrophages, eight cytokines were detected, which included macrophages.

6
JID: ACTBIO
ARTICLE IN PRESS [m5G;August 2, 2021;15:11]

L. Geddes, E. Themistou, J.F. Burrows et al. Acta Biomaterialia xxx (xxxx) xxx

Fig. 5. Relationship of cell viability and degradation time with a) pH of degradation media, b) Mass Loss, c) Swelling and d) Mn of PDLLGA.

3.6. ELISA analysis were observed after 28 days and 70 days, with values increasing
by 240% and 383%, respectively, compared to the control.
Fig. 9 shows the concentrations of the cytokines MCP-1 and
MIP-1α , in the fibroblast and macrophage conditioned media, re- 3.7. GPCR (HCAR1 ) activation and calcium mobilisation assays
spectively, after contact with PDLLGA and PLLGA. Concentrations analysis
of the cytokines MCP-1 and MIP-1α increased when in contact
with non-degraded PDLLGA and PDLLGA degraded for 7 days, in When a ligand binds to its G-protein coupled receptor (GPCR),
both cell lines (Fig. 9a). However, after 10 days, the concentration it undergoes a conformational change, which facilitates the acti-
of the cytokines decreased. The concentration of MCP-1 in the fi- vation of specific intracellular pathways. Guanosine diphosphate
broblasts decreased by 49% after 10 days and 28% after 12 days (GDP) bound to the Gα protein subunit is replaced by guanosine
relative to the control. The concentration of MIP-1α decreased by triphosphate (GTP). This initiates the dissociation of the Gα sub-
88% and 89% compared to the control, after 10 and 12 days, re- unit from the Gβγ subunit, which in turn opens calcium ion chan-
spectively. Statistical analysis showed that there was no significant nels and a surge of calcium is released into the cytoplasm [33].
change in the concentration of MCP-1 during the experiment, but The calcium mobilisation assay detects this influx of calcium using
there was a significant difference in the concentration of MIP-1α a fluorescent dye (Fluo-4).
in the macrophages after 10 and 12 days. This assay was used to determine whether GPCRs of the fibrob-
Somewhat different trends were observed when the fibroblasts lasts and macrophages, are activated in the presence of the degra-
and macrophages were in contact with PLLGA (Fig. 9b). The con- dation products released from PDLLGA and PLLGA during degrada-
centration of MCP-1 in the fibroblasts decreased with degradation tion in PBS buffer at 47°C. Observation of receptor activation would
time (Fig. 9bi). There was no significant difference in the concen- strongly point towards activation of the hydroxycarboxylic acid re-
tration of MCP-1 in the non-degraded PLLGA sample compared to ceptor 1 (HCA1 ), the agonist for which is lactic acid/lactate, and
the control, however, significant differences were observed after this is the only constituent of the degradation products known to
28, 42, 56 and 70 days. There was a 42% reduction in the concen- activate GPCRs. Receptor activation is observed, in Fig. 10, upon the
tration of MCP-1 after 28 days and a 60% reduction observed after addition of the degradation media of PDLLGA and PLLGA degraded
70 days. There was no statistically significant difference in the con- for 12- and 70- days, respectively. The curves show a sharp in-
centration of MIP-1α in the macrophages after contact with PLLGA crease in fluorescence, which gradually declines after reaching a
at any time point (Fig. 9bii). However, elevated levels of MIP-1α maximum. This is characteristic of GPCR activation [33]. Control

7
JID: ACTBIO
ARTICLE IN PRESS [m5G;August 2, 2021;15:11]

L. Geddes, E. Themistou, J.F. Burrows et al. Acta Biomaterialia xxx (xxxx) xxx

Fig. 6. Relationship of cell viability and degradation time with a) pH of degradation media, b) Mass Loss, c) Swelling and d) Mn of PLLGA.

Fig. 7. Comparison of the cell viability of L929 fibroblasts using static conditions compared to Quasi Vivo flow conditions when in contact with a) PDLLGA degraded for 10
days and b) PLLGA degraded for 42 days. (∗ indicates significant difference to the control p<0.05, Percentage viability is calculated relative to blank controls (cells only);
Percentage cell viability values of the samples may be greater than 100% in cases where the sample cells proliferate more than the “cells only” blank control).

8
JID: ACTBIO
ARTICLE IN PRESS [m5G;August 2, 2021;15:11]

L. Geddes, E. Themistou, J.F. Burrows et al. Acta Biomaterialia xxx (xxxx) xxx

Fig. 8. Relative expression levels of inflammatory cytokines detected in the supernatants collected from L929 fibroblasts and RAW264.7 macrophages after contact with
PDLLGA pre-degraded for 10 days and PLLGA pre-degraded for 42 days, and controls collected from the supernatants of L929 fibroblasts and RAW264.7 macrophages that
had no contact with the polymers.

curves for PBS (blank) and sodium L-lactate can be found in Sup- mer cytotoxicity may change during a more prolonged degradation
plementary Information (Figure S1). timeframe. Therefore, long-term cytotoxicity studies are needed to
assess the effects of these polymers and their degradation products
4. Discussion in the physiological environment. Previously, Sung et al. [38] com-
pared the biocompatibility of the fast degrading PDLLGA to slow
4.1. Correlating cytotoxicity with degradation behaviour degrading poly(ε -caprolactone) (PCL), by culturing mouse aortic
smooth muscle cells on PDLLGA and PCL polymer scaffolds and as-
The biocompatibility of bioresorbable polymers is generally sessed the in vitro cell viability after degradation for 1, 7, 14,21 and
tested using short-term in vitro cytotoxicity tests before proceeding 28 days at 37°C. The results showed that the cell viability in the
to in vivo trials [34]. These studies have shown polymers composed fast-degrading PDLLGA was lower than that of the slow degrading
of lactide and glycolide demonstrate satisfactory results [35,36]. PCL. It was suggested that the reduction in cell viability was due to
Yang et al. [37] carried out an MTT assay on L929 fibroblasts in- the increase in acidification of the local environment during poly-
cubated for up to 7 days with extract medium prepared using mer degradation. Despite the results showing differences in the cy-
PDLLGA (70:30). The results showed no cytotoxic effects at any totoxicity of the two polymers, the study did not consider that the
time point. However, this study did not account for how poly- fast-degrading PDLLGA was significantly more advanced in terms of

9
JID: ACTBIO
ARTICLE IN PRESS [m5G;August 2, 2021;15:11]

L. Geddes, E. Themistou, J.F. Burrows et al. Acta Biomaterialia xxx (xxxx) xxx

Fig. 9. Concentration of MCP-1 and MIP-1α in the supernatants collected from L929 fibroblasts and RAW264.7 macrophages in contact with PDLLGA and PLLGA after
degradation at 47°C. (∗ indicates significant difference to the control).

the relative course of its degradation, than the slow degrading PCL not provide any information on how a material interacts directly
after the 28 day period. For a true comparison, it is important to with the cells. To the best of the authors knowledge, no in vitro
evaluate the polymers at similar levels of degradation, considering study has demonstrated how the cytotoxicity of polymers com-
for example, mass and molecular weight loss. However, the slow posed of lactide and glycolide change during degradation, using
polymer degradation times at 37°C can make it difficult and im- a direct contact test. Direct contact tests are highly sensitive and
practical to obtain polymer samples for study at adequate extents can be used to detect even small levels of cytotoxicity in samples.
of degradation. One method to achieve this is to degrade the poly- One limitation of direct contact studies is the potential for me-
mers at an increased temperature. Ignatius et al. [39], evaluated chanical damage to the cell monolayer, therefore great care must
the biocompatibility of poly(D,L lactide) (PDLLA) 70:30 and PLLGA be taken with experimental technique when placing the material
90:10 using a series of in vitro tests, including an MTT assay on in contact with the cells. To overcome this, we have developed a
BALB 3T3 cells. The MTT assay was performed using the extracts protocol for pre-degrading polymers while cradled within a tran-
of PDLLA and PLLGA prepared in PBS buffer for 10 days at 37°C swell insert, which is then transferred directly from degradation
and 70°C. The extracts prepared at 70°C were used to simulate the media to cell culture. This method has been validated against con-
degradation products released after the long-term degradation of ventional direct contact methodology, showing no significant dif-
the polymers. To exclude pH concerns during the assay and to fo- ference in results, but with the advantage of convenience during
cus on the degradation products, samples were neutralised to pH sample handling, especially for degraded samples that become fri-
7.4. The results of the assay found that cell proliferation was sig- able or gel-like, and that are difficult to keep intact on handling
nificantly reduced when in contact with high concentrations of the (Supplementary information Figure S2). The aim of this study was
extracts prepared at 70°C. Although the conclusions of the study to develop an in vitro protocol that can be used to assess the cy-
found both polymers showed acceptable biocompatibility overall, totoxicity of bioresorbable polymers, at different stages of degra-
it was suggested that high concentrations of degradation products dation, in an accelerated period. The increased temperature ac-
were toxic to the cell lines. celerated degradation methodology, validated in a previous study
In vitro cytotoxicity tests are carried out by placing the test [28], was used to assess the long-term cytotoxicity of PDLLGA
material or extract solution directly onto the cultured cells [34]. and PLLGA. Both polymers were pre-degraded in PBS buffer at
Extract tests are useful in determining the cytotoxicity of leach- 47°C, before being placed on L929 fibroblasts and RAW264.7
able substances released from the biomaterial, however, they do macrophages.

10
JID: ACTBIO
ARTICLE IN PRESS [m5G;August 2, 2021;15:11]

L. Geddes, E. Themistou, J.F. Burrows et al. Acta Biomaterialia xxx (xxxx) xxx

Fig. 10. Calcium mobilisation assay of L929 fibroblasts and RAW264.7 macrophages response to polymer degradation media a) PDLLGA pre-degraded at 47°C for 12 days b)
PLLGA pre-degraded at 47°C for 70 days.

The results of this work demonstrate differences in the cyto- is no accumulation of acidic degradation products within the poly-
toxic potential of PDLLGA and PLLGA. The cytotoxicity of PDLLGA mer matrix. After 70 days, further mass loss occurs, however, there
increased significantly at the later stages of degradation, after 12 is an increase in polymer swelling. Swelling occurs via an influx of
days with 0% cell viability in both the fibroblasts and macrophages, the acidic degradation media, and a reduction in cell viability oc-
whereas PLLGA demonstrated low levels of cytotoxicity against fi- curs when the acidic degradation products are released from the
broblasts after 42 and 70 days, with 61% and 64% cell viabil- polymer matrix upon incubation with the fibroblasts. It is unclear
ity, respectively. The correlation of these results to the degrada- why the cytotoxic effect was only observed in the fibroblasts and
tion behaviour data is shown in Figs. 5 and 6. PDLLGA showed not the macrophages. However, it should be noted that the cyto-
a significant reduction in cell viability, in both fibroblasts and toxic effect observed for PLLGA was significantly lower than that
macrophages, at the later stages of degradation. Fig. 5 shows that of PDLLGA, which suggests PDLLGA has greater potential to dis-
there were no significant reductions in the pH of the degrada- play cytotoxicity compared to PLLGA. The molecular weight dis-
tion media or mass of the polymer after 12 days. However, a large tributions, from a previous study by Geddes et al. [28], revealed
reduction in the Mn was observed after 5 days, with substantial that PDLLGA is more acidic than PLLGA at the later stages of degra-
polymer swelling occurring after 10 days. This indicates that low dation as high amounts of oligomers and monomers are retained
molecular weight polymer chains are being trapped within the within the polymer matrix of PDLLGA. This could explain the dif-
polymer matrix. It is likely that these acidic degradation products ferences in cytotoxicity of the two polymers.
are released from the polymer during incubation with the cells, The temperature accelerated degradation methodology vali-
overwhelming the local microenvironment at the polymer/cell in- dated in a previous study [28], can be used as a method to pre-
terface and causing a significant reduction in cell viability. In com- degrade specimens at the accelerated temperature (47°C) where
parison, PLLGA did not become toxic to the macrophages but physiological temperature (37°C) would be impractical due to the
showed cytotoxic potential against the fibroblasts after 42 days and time duration required. It should be noted however that the model
70 days. As shown in Fig. 6, there was no significant mass loss after is only valid up to the point where mass loss occurs, so beyond
42 days, but there was a gradual decline in the molecular weight this point it will become a less reliable tool. The model devel-
of the polymer. This suggests that low molecular weight chains are oped found that the rate of hydrolysis at 47°C was increased 3.7-
retained within the polymer matrix. These acidic chains are sub- fold in PDLLGA and 21.9-fold in PLLGA compared to 37°C. Polymer
sequently released during incubation with the fibroblasts, causing degradation time is dependent on many factors, such as geometry,
a reduction in cell viability. After 56 days, mass is lost from the composition, crystallinity and implant location. It can, therefore, be
polymer and a significant decrease in the pH of the degradation difficult to give exact degradation times for every scenario and it
media demonstrates the release of the acidic degradation products is acknowledged that, in the in vivo environment, factors such as
from the polymer matrix. Therefore, when this sample is in con- phagocytosis combined with enzymic action may play a role in the
tact with fibroblasts, there is no reduction in cell viability as there later stages of degradation [40] . Few in vivo animal studies or in-

11
JID: ACTBIO
ARTICLE IN PRESS [m5G;August 2, 2021;15:11]

L. Geddes, E. Themistou, J.F. Burrows et al. Acta Biomaterialia xxx (xxxx) xxx

deed clinical studies have monitored PLLGA degradation up to the [8,23,51,52]. This study has demonstrated that methodology using
point of full resorption. Of four studies identified for PLLGA in vivo flow conditions can reveal an improved cytotoxicity profile of one
degradation, none continued to monitor up to full resorption, so polymer (PLLGA) but have no impact on another (PDLLGA). Poly-
there is no definitive answer as to when this occurs. What can be mers in the poly(lactide-co-glycolide) family degrade via heteroge-
determined from the literature [40–43], is that PLLGA is likely to neous bulk degradation. However, each polymer within this fam-
be substantially resorbed within 24 months in vivo. It should be ily exhibits different degradation characteristics which are depen-
noted, however, that additives such as ciprofloxacin [40] or trical- dent on a number of factors, such as polymer morphology, com-
cium phosphate (TCP) [41,43] may influence polymer degradation position and configurational structures, and the geometry of the
rate. Our previous work, using an in vivo rabbit model, showed a device [53,54]. Future research should consider how different poly-
slight, but non-significant reduction in degradation rate in PLLGA mer degradation mechanisms and characteristics will result in dif-
screws loaded with TCP, compared to screws composed of PLLGA ferences of physiological response.
polymer alone [47].
Throughout the literature, an inflammatory foreign body reac- 4.3. Further evaluation of the inflammatory response
tion for PDLLGA has been observed to occur between 14–243 days
[21,41,44–48]. Fewer incidences of foreign body reactions have The aim of the final stage of this study was to gain a better
been reported for PLLGA, with some reporting no foreign body re- understanding of the in vitro inflammatory response of fibroblasts
actions [40,42], and others reporting incidences of cyst formation and macrophages to PDLLGA and PLLGA at progressive stages of
occurring between 730–1460 days [41,43]. Based on the results of degradation. The results of the antibody array (Fig. 8) and ELISA
this in vitro study, the degradation times extrapolated to 37°C, in- (Fig. 9) demonstrate a general downregulation of inflammatory cy-
dicate adverse reactions would be predicted to occur 37 days after tokines at degradation levels where PDLLGA and PLLGA begin to
implantation for PDLLGA and after 920 days for PLLGA. This ex- show cytotoxic potential. Alternatively, when no cytotoxicity was
trapolation method corroborates the results observed in this study observed, there was either no significant change or an upregu-
with the adverse reactions noted by clinical case reports in the lit- lation in the expression of the inflammatory cytokines. This was
erature [21,41,43–48]. contrary to our expectations, as it was anticipated that materials
displaying cytotoxic potential would initiate an inflammatory re-
4.2. Development of an improved method to test for cytotoxicity sponse. Surprisingly, these results suggest an immunosuppressive
or anti-inflammatory effect of PDLLGA and PLLGA. Sung et al. [38],
In Section 4.1, fibroblasts and macrophages were cultured in compared the biocompatibility of fast degrading PDLLGA to the
traditional static 24-well plates. An MTT assay was carried out, af- slow degrading PCL. The polymer scaffolds were implanted subcu-
ter PDLLGA and PLLGA were placed on the cultured cells and in- taneously in wild-type 129/SvEv mice, harvested after 7, 14, 21 and
cubated for 24 hours. Multiwell plates, used in this way, have be- 28 days and assessed for the density of inflammatory cells within
come a standard in cell culture experiments. However, static con- the polymer. It was found that a significantly reduced number of
ditions are limited as they do not resemble the dynamic nature inflammatory cells migrated into PDLLGA than PCL and it was sug-
of the physiological environment. The QV500 system was inves- gested that the acidic environment, created by PDLLGA, inhibited
tigated as a technique that could be used to improve upon the cell migration. However, it is not clear what inflammatory cells
static multiwell plate conditions, and better mimic in vivo con- were detected. Britland et al. [55] found a dose dependent rela-
ditions. The transwell methodology used for the static conditions tionship between lactate concentration and cell viability using an
was adapted using the QV500 system, as the dimension of the MTT assay. It was observed that cell viability was reduced when
chambers (15 mm internal diameter, 10 mm height) are similar to cells were treated with a lactate concentration of 10 mM for hu-
those of the standard 24-well plate [49,50]. The system was con- man umbilical vein endothelial cells (HUVEC) and between 10 and
nected in series to allow for a continuous flow of media through 50 mM for 3T3 fibroblasts. However, the authors were not able to
each chamber. Therefore, degradation products, released into the determine the concentration threshold for when lactate made this
media, are continuously carried away and replenished with fresh change from being a positive influence on the healing process to
media, as would be the case in vivo, where degradation products becoming a detrimental presence.
would diffuse from the vicinity of the implanted material follow- In light of our results discussed above, we formed the hypoth-
ing a concentration gradient, and where there would be a con- esis that the degradation products of PDLLGA and PLLGA must be
tinuous exchange of tissue fluid. In this study, samples were as- active in an immunomodulatory capacity. Lactate/lactic acid, a ma-
sessed for cytotoxicity using the QV500 system and compared to jor degradation product, has been found to be biologically active
the response observed under static conditions (Fig. 7). The results and involved in the signalling pathways and regulatory functions
demonstrate that when PDLLGA reaches a certain stage of degrada- in many physiological and pathological conditions [56,57]. Lactate
tion it becomes inherently cytotoxic to the fibroblasts, even under is the conjugate base of lactic acid and is formed in the body as
flow conditions. This suggests that the amount of acidic degrada- the end product of anaerobic or aerobic glycolysis [55]. Anaerobic
tion products released from PDLLGA exceeds the clearing capacity glycolysis yields lactate under hypoxic conditions in normal cells,
of the flow system, resulting in a reduction in cell viability. PLLGA whereas, in aerobic glycolysis (Warburg effect [58]) lactate is pro-
showed improved cell viability using the flow system with no sig- duced, in the presence of oxygen, in rapidly dividing cells, for ex-
nificant difference between the degraded sample and the control, ample, in cancer cells or during wound healing [56,59]. Monocar-
whereas under static conditions a cytotoxic response was observed. boxylate transporters (MCTs) are used to transport lactate across
This indicates that the flow of fresh media through the chamber cell membranes, which is carried out by a series of reactions, in-
provides sufficient clearance of the acidic degradation products re- volving the conversion of lactate to pyruvate and alterations to
leased from PLLGA. The flow system is more indicative of the true the intercellular NADH/NAD+ ratio [59,60]. Lactate has tradition-
in vivo scenario where diffusion through, and replenishment of tis- ally been considered as an energy-rich metabolic fuel, but more
sue fluid in the peri-implant environment will result in the clear- recently its role as an important cell signalling molecule has been
ance of acidic degradation products. come to light. It was observed for the first time in 2009 that HCA1
Throughout the literature there are reports that implanting (also known as G-protein coupled receptor-81 or GPR81) could
polymers composed of lactide and glycolide in bone regions be activated by L-lactate [56,57,59,61]. The D-lactate enantiomer
with poor vascularity can result in an adverse reaction occurring has also been reported as a agonist/partial agonist of the receptor

12
JID: ACTBIO
ARTICLE IN PRESS [m5G;August 2, 2021;15:11]

L. Geddes, E. Themistou, J.F. Burrows et al. Acta Biomaterialia xxx (xxxx) xxx

[61–63]. The stimulation of HCA1 by lactate results in cell sig- tion of PDLLGA and PLLGA has demonstrated great potential for the
nalling by the downregulation of cAMP and the activation of non- assessment of bioresorbable polymers with long degradation times
canonical, β -arrestin-dependent signalling pathways [56,57]. This as the results extrapolated to 37°C corroborate with the clinical
method of signalling is different to MCT-driven transport and its data of adverse reactions occurring, in vivo, in the literature. Efforts
discovery has led to significant interest in the role of lactate as a were also made in this work to improve upon the static conditions
cell signalling molecule through the activation of HCA1 . Lactate has generally used for in vitro research. The results validate the feasi-
been shown to have an important role in the modulation of the bility of using the QV500 system as a method to better represent
immune response and inflammation of various pathophysiological the in vivo flow environment. This study has shown differences in
conditions. The activation of HCA1 has been shown to have an im- the cytotoxicity of PDLLGA and PLLGA and proposed explanations
munosuppressive effect on the regulation of intestinal inflamma- for the observed response. It is recommended that future research
tion by signalling the downregulation of inflammatory cytokines into orthopaedic devices focuses on PLLGA.
in colonic macrophages and dendritic cells [64]. The presence of The final section of this work demonstrates the immunosup-
lactate generated via aerobic glycolysis in cancer cells has been pressive and anti-inflammatory role of lactate. To the best of the
found to promote tumour growth and metastasis by acting as an authors knowledge, the effect of lactate in the clinical outcomes
immunosuppressant [59,65,66]. Lactate has also been shown to in- of PLA and PLGA used for orthopaedic devices has not been con-
crease collagen deposition and promote angiogenesis by inducing sidered in the literature, due to the recent discovery of the ligand
the release of proangiogenic factors, such as VEGF [67–70]. The activity of lactate at HCA1 receptor. It is likely lactate plays a sig-
therapeutic effect of poly(lactide-co-glycolide) (PLGA) polymers in nificant role in the cell signalling and regulatory functions at the
wound healing and as a drug delivery system has been reported site of polymer implantation. It is therefore important future re-
in the literature. Porporato et al. [69], reported that wound heal- search considers this effect when analysing the biological response
ing was accelerated in two strains of mice, by delivering a rate- of these polymers.
controlled release of lactate using PDLLGA 50:50. It was found that
a nearly 60% decrease in excisional wound closure, 10 days post
injury, and a 2.2-fold increase in angiogenesis in the mice treated Declaration of Competing Interest
with the polymer occurred. Similar results were also observed by
Chereddy et al. [71], with a 75% wound closure observed after 10 The authors declare that they have no known competing finan-
days, when the injury was treated with 1 mg of PLGA nanopar- cial interests or personal relationships that could have appeared to
ticles, however, it is unclear which enantiomer of lactide was influence the work reported in this paper.
used. They also observed a downregulation in the inflammatory
expressions of glutathione peroxidase (GPx) and nuclear factor-κ B
Acknowledgments
(NFκ B). These observations reported in the literature, and the re-
sults reported in this present study (inflammatory cytokine pro-
Corbion Biomaterials, The Netherlands, is acknowledged for the
filing, and media from PDLLGA and PLLGA degradation studies in-
kind gift of poly(D, L-lactide-co-glycolide) and poly(L-lactide-co-
ducing GPCR receptor activation, as demonstrated by calcium mo-
glycolide). This work has been funded by the Department for the
bilisation), have important implications for lactic acid-based biore-
Economy, Northern Ireland, via a PhD studentship awarded to LG.
sorbable materials used for medical devices, such as orthopaedic
fixation screws. These devices have to date been chosen for their
physical and mechanical characteristics, however their effects on Supplementary materials
host tissues may be much further reaching, given the proven cell
signalling role of lactate. Devices made from these polymers have Supplementary material associated with this article can be
the potential to be bio-active and influence inflammation and heal- found, in the online version, at doi:10.1016/j.actbio.2021.07.049.
ing at the site of implantation.
The results in this study support what has been revealed in
the literature in relation to the immunosuppressive effect of lac- References
tate. During the degradation of PDLLGA and PLLGA a downregula-
[1] M.S. Taylor, A.U. Daniels, K.P. Andriano, J. Heller, Six bioabsorbable polymers:
tion in the inflammatory cytokines in fibroblasts and macrophages In vitro acute toxicity of accumulated degradation products, J. Appl. Biomater.
was observed. The results from the calcium mobilisation assays 5 (1994) 151–157.
demonstrate that this observed effect is likely to be due to the ac- [2] J. Bergsma, Late degradation tissue response to poly(L-lactide) bone plates and
screws, Biomaterials 16 (1995) 25–31.
tivation of the GPCR, HCA1 , by the polymer degradation product, [3] P.B. Maurus, C.C. Kaeding, Bioabsorbable implant material review, Oper. Tech.
lactate/lactic acid, which in turn acts as a signalling molecule to Sports Med. 12 (2004) 158–160.
downregulate the cytokines. This effect is likely to have an impact [4] R.E. Cameron, A. Kamvari-Moghaddam, Synthetic bioresorbable polymers, in:
Degrad. Rate Bioresorbable Mater. Predict. Eval., 2008: pp. 43–66.
on the in vivo implantation of PLGA and PLA polymers. It is possi- [5] K. Ginjupalli, G.V. Shavi, R.K. Averineni, M. Bhat, N. Udupa, N.P. Upadhya,
ble that as a polymer degrades, in vivo, the lactate released initially Poly(α -hydroxy acid) based polymers: a review on material and degradation
has a therapeutic anti-inflammatory effect on the surrounding tis- aspects, Polym. Degrad. Stab. 144 (2017) 520–535.
[6] D.G. Weir NA, F.J. Buchanan, J.F. Orr, D.F. Farrar, Degradation of poly-L- lac-
sue. However, at the later stages of degradation, significant poly-
tide. Part 2: increased temperature accelerated degradation, Proc. IME H J. Eng.
mer erosion leads to a large release of acidic degradation products, Med. 218 (2004) 321–330.
which could be considered the ‘tipping-point’ at which lactate lev- [7] D. Farrar, Bioresorbable polymers in orthopaedics, Biomaterials 6 (2005) 1–4.
[8] J.C. Middleton, A.J. Tipton, Synthetic biodegradable polymers as orthopedic de-
els overwhelm the polymer/tissue environment.
vices, Biomaterials 21 (20 0 0) 2335–2346.
[9] J.C.Y. Chan, K. Burugapalli, J.L. Kelly, A.S. Pandit, in: Influence of Clinical Ap-
5. Conclusion plication on Bioresorbability: Host Response, Elsevier, 2008, pp. 267–318. in:
Degrad. Rate Bioresorbable Mater..
[10] J.M. Anderson, A. Rodriguez, D.T. Chang, Foreign body reaction to biomaterials,
The results observed in this study demonstrate the occurrence Semin. Immunol. 20 (2008) 86–100.
of a delayed inflammatory reaction at the late stages of polymer [11] Z. Sheikh, P.J. Brooks, O. Barzilay, N. Fine, M. Glogauer, Macrophages, for-
degradation. It can be difficult to extrapolate in vitro results to in eign body giant cells and their response to implantable biomaterials, Materials
(Basel) 8 (2015) 5671–5701.
vivo applications as the conditions are very different. However, the [12] E. Mortaz, S.D. Alipoor, I.M. Adcock, S. Mumby, L. Koenderman, Update on neu-
use of the increased temperature, 47°C, to accelerate the degrada- trophil function in severe inflammation, Front. Immunol. 9 (2018) 1–14.

13
JID: ACTBIO
ARTICLE IN PRESS [m5G;August 2, 2021;15:11]

L. Geddes, E. Themistou, J.F. Burrows et al. Acta Biomaterialia xxx (xxxx) xxx

[13] A.R. Amini, J.S. Wallace, S.P. Nukavarapu, Short-term and long-term effects of [41] H.E. Bourke, L.J. Salmon, A. Waller, C.S. Winalski, H.A. Williams, J.M. Linklater,
orthopedic biodegradable implants, J. Long. Term. Eff. Med. Implants. 21 (2011) A. Vasanji, J.P. Roe, L.A. Pinczewski, Randomized controlled trial of osteocon-
93–122. ductive fixation screws for anterior cruciate ligament reconstruction: a com-
[14] J.M. Anderson, G. Cook, B. Costerton, S.R. Hanson, A. Hensten-Pettersen, parison of the calaxo and milagro screws, Arthrosc. - J. Arthrosc. Relat. Surg.
N. Jacobsen, R.J. Johnson, R.N. Mitchell, M. Pasmore, F.J. Schoen, M. Shirtliff, 29 (2013) 74–82.
P. Stoodley, Host reactions to biomaterials and their evaluation, in: Biomater. [42] A.S. Xue, J.C. Koshy, W.M. Weathers, E.M. Wolfswinkel, Y. Kaufman, S.E. Sharabi,
Sci. (1996) 293–304. R.H. Brown, M.J. Hicks, L.H. Hollier, Local foreign-body reaction to commercial
[15] I.C.C. de Moraes Porto, in: Polymer Biocompatibility, in: Polymerization, InTech, biodegradable implants: an in vivo animal study, Craniomaxillofac. Trauma Re-
2012, pp. 47–62, doi:10.5772/47786. constr. 7 (2014) 27–34.
[16] C.G. Ambrose, T.O. Clanton, Bioabsorbable implants: review of clinical experi- [43] C.L. Cox, K.P. Spindler, J.P. Leonard, B.J. Morris, W.R. Dunn, E.K. Reinke, Do
ence in orthopedic surgery, Ann. Biomed. Eng. 32 (2004) 171–177. newer-generation bioabsorbable screws become incorporated into bone at two
[17] S. Konan, F.S. Haddad, A clinical review of bioabsorbable interference screws years after ACL reconstruction with patellar tendon graft? J. Bone Jt. Surg. 96
and their adverse effects in anterior cruciate ligament reconstruction surgery, (2014) 244–250.
Knee 16 (2009) 6–13. [44] A.C. Stahelin, A. Weiler, H. Rufenacht, R. Hoffmann, A. Geissmann, R. Feinstein,
[18] W.H. De Jong, J.E. Bergsma, J.E. Robinson, R.R.M. Bos, Tissue response to par- Clinical degradation and biocompatibility of different bioabsorbable interfer-
tially in vitro predegraded poly- L -lactide implants, Biomaterials 26 (2005) ence screws : a report of six cases, 13 (1997) 238–244.
1781–1791. [45] G. Lajtai, I. Noszian, K. Humer, F. Unger, G. Aitzetmuller, E. Orthner, Serial mag-
[19] O.M. Böstman, H.K. Pihlajamäki, Late foreign-body reaction to an intraosseuous netic resonance imaging evaluation of operative site after fixation of patellar
bioabsorbable polylactic acid screw, J. Bone Jt. Surg. 80-A (1998) 1791–1794. tendon graft with bioabsorbable interference screws in anterior cruciate liga-
[20] K.A. Athanasiou, G.G. Niederauer, C.M. Agrawal, Sterilization, toxicity, biocom- ment reconstruction, Arthroscopy 15 (1999) 709–718.
patibility and clinical applications of polylactic acid /polyglycolic acid copoly- [46] C.L. Cox, K.C. Homlar, J.L. Carey, K.P. Spindler, CALAXO osteoconductive inter-
mers, Biomaterials 17 (1996) 93–102. ference screw: the value of postmarket surveillance, J. Surg. Orthop. Adv. 19
[21] S. Konan, F.S. Haddad, The unpredictable material properties of bioabsorbable (2010) 121–124.
PLC interference screws and their adverse effects in ACL reconstruction [47] I. Palmer, S.A. Clarke, F.J. Buchanan, Enhanced release of calcium phosphate
surgery, Knee Surgery, Sport. Traumatol. Arthrosc. 17 (2009) 293–297. additives from bioresorbable orthopaedic devices using irradiation technology
[22] J.H. Kwak, J.A. Sim, S.H. Kim, K.C. Lee, B.K. Lee, Delayed intra-articular inflam- is non-beneficial in a rabbit model: an animal study, Bone Joint Res. 8 (2019)
matory reaction due to poly-L-lactide bioabsorbable interference screw used 266–274.
in anterior cruciate ligament reconstruction, arthrosc, J. Arthrosc. Relat. Surg. [48] J. Dujardin, H. Vandenneucker, J. Bellemans, Tibial cyst and intra-articu-
24 (2008) 243–246. lar granuloma formation after anterior cruciate ligament reconstruction us-
[23] P.K. Givissis, S.I. Stavridis, P.J. Papagelopoulos, P.D. Antonarakos, ing polylactide carbonate osteoconductive interference screws, arthrosc, J.
A.G. Christodoulou, Delayed foreign-body reaction to absorbable implants in Arthrosc. Relat. Surg. 24 (2008) 238–242.
metacarpal fracture treatment, Clin. Orthop. Relat. Res. 468 (2010) 3377–3383. [49] G. Mattei, S. Giusti, A. Ahluwalia, Design criteria for generating physiologically
[24] P. Tuompo, E.K. Partio, H. Pätiälä, K. Jukkala-Partio, E. Hirvensalo, P. Rokka- relevant in vitro models in bioreactors, Processes 2 (2014) 548–569.
nen, Causes of the clinical tissue response to polyglycolide and polylactide im- [50] P. Miranda-Azpiazu, S. Panagiotou, G. Jose, S. Saha, A novel dynamic multicel-
plants with an emphasis on the knee, Arch. Orthop. Trauma Surg. 121 (2001) lular co-culture system for studying individual blood-brain barrier cell types
261–264. in brain diseases and cytotoxicity testing, Sci. Rep. 8 (2018) 1–10.
[25] J.M. Anderson, Future challenges in the in vitro and in vivo evaluation of bio- [51] O. Böstman, H. Pihlajamäki, Clinical biocompatibility of biodegradable or-
material biocompatibility, Regen. Biomater. 3 (2016) 73–77. thopaedic implants for internal fixation: a review, Biomaterials 21 (20 0 0)
[26] W. Li, J. Zhou, Y. Xu, Study of the in vitro cytotoxicity testing of medical de- 2615–2621.
vices (Review), Biomed. Rep. (2015) 617–620. [52] O.M. Böstman, H.K. Pihlajamäki, Adverse tissue reactions to bioabsorbable fix-
[27] A. Lock, J. Cornish, D.S. Musson, The role of in vitro immune response assess- ation devices, Clin. Orthop. Relat. Res. (20 0 0) 216–227.
ment for biomaterials, J. Funct. Biomater. 10 (2019) 31. [53] S. Li, M. Vert, Biodegradation of Aliphatic Polyesters, in: Degrad. Polym. Princ.
[28] L. Geddes, L. Carson, E. Themistou, F. Buchanan, A comparison of the in- Appl., Springer, Dordrecht, 1995.
creased temperature accelerated degradation of Poly(D,L-lactide-co-glycolide) [54] S. Li, Hydrolytic degradation characteristics of aliphatic polyesters derived from
and Poly(L-lactide-co-glycolide), Polym. Test. 91 (2020) 106853. lactic and glycolic acids, J. Biomed. Mater. Res. (Appl. Biomater.) 48 (1998)
[29] International Organisation for Standardisation, Biological Evaluation of Medical 342–353.
Devices - Part 5: Tests for in vitro cytotoxicity, 2009. [55] S. Britland, O. Ross-Smith, H. Jamil, A.G. Smith, K. Vowden, P. Vowden, The
[30] C.K. Postnikoff, R. Pintwala, S. Williams, A.M. Wright, D. Hileeto, M.B. Gorbet, lactate conundrum in wound healing: clinical and experimental findings indi-
Development of a curved, stratified, in vitro model to assess ocular biocom- cate the requirement for a rapid point-of-care diagnostic, Biotechnol. Prog. 28
patibility, PLoS One 9 (2014) e96448. (2012) 917–924.
[31] S. Nithiananthan, A. Crawford, J.C. Knock, D.W. Lambert, S.A. Whawell, Physi- [56] K.K. Chereddy, V.L. Payen, V. Préat, PLGA polymer: from a classic drug car-
ological fluid flow moderates fibroblast responses to TGF-β 1, J. Cell. Biochem. rier to a novel therapeutic activity contributor, J. Control. Release. 289 (2018)
118 (2017) 878–890. 10–13.
[32] X. Wang, Y. Xia, L. Liu, M. Liu, N. Gu, H. Guang, F. Zhang, Comparison of MTT [57] S. Sun, H. Li, J. Chen, Q. Qian, Lactic acid: no longer an inert and end-product
assay, flow cytometry, and RT-PCR in the evaluation of cytotoxicity of five of glycolysis, Physiology 32 (2017) 453–463.
prosthodontic materials, J. Biomed. Mater. Res. - Part B Appl. Biomater. 95 B [58] M.V. Liberti, J.W. Locasale, The Warburg effect: how does it benefit cancer
(2010) 357–364. cells? (vol 41, pg 211, 2016), Trends Biochem. Sci. 41 (2016) 287.
[33] J. Caers, K. Peymen, N. Suetens, L. Temmerman, T. Janssen, L. Schoofs, I. Beets, [59] T.P. Brown, V. Ganapathy, Lactate/GPR81 signaling and proton motive force
Characterization of G protein-coupled receptors by a fluorescence-based cal- in cancer: role in angiogenesis, immune escape, nutrition, and Warburg phe-
cium mobilization assay, J. Vis. Exp. (2014) 1–10. nomenon, Pharmacol. Ther. 206 (2020) 107451.
[34] S. Anju, N. Prajitha, V.S. Sukanya, P.V. Mohanan, Complicity of degradable poly- [60] L. Ippolito, A. Morandi, E. Giannoni, P. Chiarugi, Lactate: a metabolic driver in
mers in health-care applications, Mater. Today Chem. 16 (2020) 100236. the tumour landscape, Trends Biochem. Sci. 44 (2019) 153–166.
[35] E. Díaz, I. Puerto, I. Sandonis, S. Ribeiro, S. Lanceros-Mendez, Hydrolytic degra- [61] C. Liu, J. Wu, J. Zhu, C. Kuei, J. Yu, J. Shelton, S.W. Sutton, X. Li, J.Y. Su,
dation and cytotoxicity of poly(lactic- co -glycolic acid)/multiwalled carbon T. Mirzadegan, C. Mazur, F. Kamme, T.W. Lovenberg, Lactate inhibits lipolysis
nanotubes for bone regeneration, J. Appl. Polym. Sci. 137 (2020) 48439. in fat cells through activation of an orphan G-protein-coupled receptor, GPR81,
[36] J.R. Sarasua, N. López-Rodríguez, E. Zuza, S. Petisco, B. Castro, M. Del Olmo, J. Biol. Chem. 284 (2009) 2811–2822.
T. Palomares, A. Alonso-Varona, Crystallinity assessment and in vitro cytotox- [62] W. Wagner, K.D. Kania, A. Blauz, W.M. Ciszewski, The lactate receptor
icity of polylactide scaffolds for biomedical applications, J. Mater. Sci. Mater. (HCAR1/GPR81) contributes to doxorubicin chemoresistance via ABCB1 trans-
Med. 22 (2011) 2513–2523. porter up-regulation in human cervical cancer HeLa cells, J. Physiol. Pharmacol.
[37] Y. Yang, J. Fang, W. Liu, Y. Zhao, T. Huang, J. Cui, Z. Zhou, In vitro degradation 68 (n.d.) 555–564.
behavior and biological properties of a novel maleated poly (D, L-lactide-co-g- [63] X. Castillo, K. Rosa, M.T. Wyss, K. Drandarov, A. Buck, L. Pellerin, B. Weber,
lycolide) for biomedical applications, J. Macromol. Sci. Part B Phys. 58 (2019) A probable dual mode of action for both L- and D-lactate neuroprotection in
209–218. cerebral ischemia, J. Cereb. Blood Flow Metab. 35 (2015) 1561–1569.
[38] H. Sung, C. Meredith, C. Johnson, Z.S. Galis, The effect of scaffold degrada- [64] P. Ranganathan, A. Shanmugam, D. Swafford, A. Suryawanshi, P. Bhattacharjee,
tion rate on three-dimensional cell growth and angiogenesis, Biomaterials 25 M.S. Hussein, P.A. Koni, P.D. Prasad, Z.B. Kurago, M. Thangaraju, V. Ganapathy,
(2004) 5735–5742. S. Manicassamy, GPR81, a cell-surface receptor for lactate, regulates intestinal
[39] A.A. Ignatius, L.E. Claes, In vitro biocompatibility of bioresorbable polymers: homeostasis and protects mice from experimental colitis, J. Immunol. (2018)
poly (L, DL-lactide) and poly (L-lactide-co-glycolide), Biomaterials 17 (1996) 1781–1789.
831–839. [65] C.L. Roland, T. Arumugam, D. Deng, S.H. Liu, B. Philip, S. Gomez, W.R. Burns,
[40] J. Tiainen, Y. Soini, E. Suokas, M. Veiranto, P. Törmälä, T. Waris, N. Ashammakhi, V. Ramachandran, H. Wang, Z. Cruz-Monserrate, C.D. Logsdon, Cell surface lac-
Tissue reactions to bioabsorbable ciprofloxacin-releasing polylactide-polygly- tate receptor GPR81 is crucial for cancer cell survival, Cancer Res. 74 (2014)
colide 80/20 screws in rabbits’ cranial bone, J. Mater. Sci. Mater. Med. 17 5301–5310.
(2006) 1315–1322.

14
JID: ACTBIO
ARTICLE IN PRESS [m5G;August 2, 2021;15:11]

L. Geddes, E. Themistou, J.F. Burrows et al. Acta Biomaterialia xxx (xxxx) xxx

[66] A. Calcinotto, P. Filipazzi, M. Grioni, M. Iero, A. De Milito, A. Ricupito, A. Cova, [69] P.E. Porporato, V.L. Payen, C.J. De Saedeleer, V. Préat, J.P. Thissen, O. Feron,
R. Canese, E. Jachetti, M. Rossetti, V. Huber, G. Parmiani, L. Generoso, M. Santi- P. Sonveaux, Lactate stimulates angiogenesis and accelerates the healing of su-
nami, M. Borghi, S. Fais, M. Bellone, L. Rivoltini, Modulation of microenviron- perficial and ischemic wounds in mice, Angiogenesis 15 (2012) 581–592.
ment acidity reverses anergy in human and murine tumor-infiltrating T lym- [70] T.K. Hunt, R.S. Aslam, S. Beckert, S. Wagner, Q.P. Ghani, M.Z. Hussain, S. Roy,
phocytes, Cancer Res. 72 (2012) 2746–2756. C.K. Sen, Aerobically derived lactate stimulates revascularization and tissue re-
[67] Q.P. Ghani, S. Wagner, M.Z. Hussain, T.K. Hunt, Role of ADP-ribosylation in pair via redox mechanisms, Antioxidants Redox Signal 9 (2007) 1115–1124.
wound repair. The contributions of Thomas K. Hunt, MD, Wound Repair Re- [71] K.K. Chereddy, R. Coco, P.B. Memvanga, B. Ucakar, A. Des Rieux, G. Vander-
gen. 11 (2003) 439–444. meulen, V. Préat, Combined effect of PLGA and curcumin on wound healing
[68] J. Song, K. Lee, S.W. Park, H. Chung, D. Jung, Y.R. Na, H. Quan, C.S. Cho, activity, J. Control. Release. 171 (2013) 208–215.
J.H. Che, J.H. Kim, J.H. Park, S.H. Seok, Lactic acid upregulates VEGF expression
in macrophages and facilitates choroidal neovascularization, Investig. Ophthal-
mol. Vis. Sci. 59 (2018) 3747–3754.

15

You might also like