Sunteți pe pagina 1din 95

C CUPRINS

Studii experimentale
Studii clinice
Meta-analize
pag.
pag.
pag.
1
23
45
Utilizari speciale pag. 59
Referinte pag. 75

Material realizat cu sprijinul Dr. Dan Laptoiu


Medic Primar Ortopedie Traumatologie
Doctor in Stiinte Medicale
Competenta Artroscopie
Spitalul Clinic Colentina Bucuresti
1
S
Studii experimentale

1
2
Acidul Hialuronic creste viabilitatea celulelor
derivate din tendon si expresia colagenului de
tip I in vitro: Studiu comparativ intre 4 tipuri de
Acid Hialuronic cu greutati moleculare diferite
Leonardo Osti, Martina Berardoco, Viviana di Giacommo, Grazziella di Bernardo, Francesco Oliva si
Anna Berardi

REZUMAT

BACKGROUND:
Acidul Hialuronic (AH) a fost deja aprobat de catre FDA (Food and Drug Administration)
in tratamentul osteoartritei (OA), in timp ce in tratamentul tendinopatiei subiectul este
inca dezbatut. In acest studiu scopul a fost evaluarea efectelor a 4 tipuri de Acid
Hialuronic cu greutate moleculara diferita asupra celulor derivate din tendon uman
de coafa de rotatori in ceea ce priveste viabilitatea celulara, proliferarea, apoptoza si
expresia colagenului de tip I si tip III.

METODE:
S-a dezvoltat un model in vitro de celule derivate din tendon uman de coafa de rotatori
pentru a studia efectele a 4 preparate(Ps) diferite de Acid Hialuronic (hialorunat de sodiu
(GM- greutate moleculara): 500-730 KDa – Hyalgan, 1000 kDa Artrosulfur HA, 1600 kDa
Hyalubrix si 2200 kDa Synolis – VA) in concentratii variate. A fost studiata morfologia
celulelor derivate din tendon dupa 0, 7 si 14 zile de cultura. Viabilitatea, prolifereraea
si apoptoza au fost evaluate dupa 0, 24 si 48 h de cultura. Expresia si sedimentarea
colagenului de tip I si de tip III au fost evaluate dupa 1,7 si 14 zile de cultura.

REZULTATE:
Toate tipurile de AH testate au crescut viabilitatea si proliferarea, dependent de doza.
Toate AHPs au redus dupa 24 de ore apoptoza comparativ cu celulele control. Mai mult
AHPs au stimulat sinteza de colagen de tip I intr-un trend dependent de doza dupa
14 zile, fara sa creasca colagenul de tip III; in plus in prezenta Synolis-VA expresia si
sedimentarea colagenului de tip I a fost semnificativ mai mare comparativ cu alte tipuri
de AH.

CONCLUZIA:
Preparatele de AH au crescut viabilitatea, proliferarea si expresia de colagen de tip I in
celulele derivate din tendon;

CUVINTE CHEIE:
Acid Hialuronic, Tendinopatie, Celule derivate din tendon uman, tendoane de coafa de
rotatori.

22
Hyaluronic Acid increases tendon derived cell
viability and collagen type I expression in vitro:
Comparative study of four different Hyaluronic
Acid preparations by molecular weight
Leonardo Osti, Martina Berardoco, Viviana di Giacommo, Grazziella di Bernardo, Francesco Oliva
and

Abstract
Background: Hyaluronic Acid (HA) has been already approved by Food and Drug Administration (FDA) for osteoarthritis (OA), while
its use in the treatment of tendinopathy is still debated. The aim of this study was to evaluate in human rotator cuff tendon derived
cells the effects of four different HA on cell viability, proliferation, apoptosis and the expression of collagen type I and collagen
type III.
Methods: An in vitro model was developed on human tendon derived cells from rotator cuff tears to study the effects of four
different HA preparations (Ps) (sodium hyaluronate MW: 500-730 KDa - Hyalgan®, 1000 kDa Artrosulfur HA®, 1600 KDa Hyalubrix®
and 2200 KDa Synolis-VA®) at various concentrations. Tendon derived cells morphology were evaluated after 0, 7 and 14 d of
culture. Viability, proliferation, apoptosis were evaluated after 0, 24 and 48 h of culture. The expression and deposition of collagen
type I and collagen type III were evaluated after 1, 7 and 14 d of culture.
Results: All HAPs tested increased viability and proliferation, in dose dependent manner. HAPs already reduce apoptosis at 24
h compared to control cells (without HAPs). Furthermore, HAPs stimulated the synthesis of collagen type I in a dose dependent
fashion over 14 d, without increase in collagen type III; moreover, in the presence of Synolis-VA® the expression and deposition of
collagen type I was significantly higher as compare with the other HAPs.
Conclusions: HAPs enhanced viability, proliferation and expression of collagen type I in tendon derived cells.

Background of the tendon derive cells, moreover, the pattern of gene


Non-traumatic rotator cuff tears are the most common shoulder expression is consistent with the presence of mixed
joint disease, and have age-associated incidence, since they population. [19]. Clinical studies in patients with rotator
are favored by the co-presence of metabolic diseases such cuff disease ranging from tendinopathy to rotator cuff
as diabetes, thyroid disorders and hypercholesterolemia tears detected a positive influence on the reduction of
[1–4]. Conservative treatment of tendinopathies has been pain and improved function with no consistent side-effects
increasingly supported by scientific evidence over the last recorded. Despite the increased awareness of the effective
twenty year [5]. Despite decades of study for HA in the role of HA in regenerative medicine, the therapeutic use of
conservative treatment of osteoarthritis [6], poor evidence HA for tendinopathies has been poorly studied on human
is present in the literature about the indication of this drug tenocytes in vitro.
for tendinopathies [7]. During tendinopathy and tendon In this study, was evaluated the effect of four different HAPs
acute rupture has been reported an higher incidence of by molecular weight on viability, metabolic activity, apoptosis
tenocyte apoptosis and decreased collagen synthesis [8]. and collagen type I and collagen type III expression on human
Failure of the healing response may occur in genetically- rotator cuff tendon tears derived cells.
predisposed patients, decreasing the resistance of ten-
don structures to mechanical load, resulting eventually in Methods
tendinopathy, or a tendon tear [4, 9, 10]. All the procedures described in this investigation were approved
Hyaluronic acid (HA) (or “hyaluronan”, or “sodium by the Ethical Committee of Rome Tor Vergata University. All
hyaluronate preparation”) is a high molecular weight the patients gave written informed consent to be included in
glycosa-miniglycan consists of the repetition of a the present study. Tendon samples were harvested from
disaccharide unit of an N-acetyl-glucosamine and a healthy area close to degenerative supraspinatus tendons
β-glucuronic acid [11]. Its most important physicochemical tear area biopsy specimen in 10 patients were operated
properties are its capacity to retain water, having a very arthroscopically for shoulder rotator cuff repair, with a
high hydration ratio, and its visco-elasticity. These two mean age of 63,6 ± 6,9 years. Trauma history, heavy smoking
properties are, however, interdependent. Changes in HA habit or systemic conditions such as thyroid disorders,
concentrations within the extracellular matrix modulate diabetes, gynecological condition, neoplasia, rheumatic
a variety of cellular functions, such as cell migration diseases, and any previous or concomitant rotator cuff disease
[12, 13], adhesion [14, 15], and proliferation [16–18]. were considered exclusion criteria.
Several important medical applications of HA have been
discovered for joints degeneration [7]. Additionally, high Tendon cell cultures
local concentration of HA causes release of endogenous Primary human tendon derived cell cultures were estab- lished
growth factors and stimulates cell–cell interaction, as previously described [20]. In brief, cells were iso- lated
resulting in faster cell proliferation during early stages of in from tissue sample by washing several times with phosphate
vitro culture. Additional effects reported in clinical animal buffered saline Dulbecco’s W/O Ca and Mg (PBS) + 1 %
studies are related to an accelerated healing process in penicillin/streptomycin (Invitrogen, Life Technologies, Carlsbad,
the tendons after repair, and decreased scar formation CA, USA). Small pieces of fresh tendon isolated were carefully
within the tendons. There has been a lack of specific dissected and mechanically disaggregated with the aid of fine
studies on human shoulder derived cells. Much of the watchmaker forceps to maximize the interface between tissue
study, has been limited by the lack of the exact phenotype and medium. Finally, the tendons were immediately placed on

33
Petri dishes of 60 mm in diameter (Greiner CELLSTAR dish, Aldrich), and in triplicates in 100 μl of α-MEM supplemented
Sigma-Aldrich, Saint Louis, MO, USA), containing 5 mL of with 10 % FCS . Cells were cultured as previous described [20].
α-MEM supplemented with 20% heat-inactivated foetal calf se- Briefly, after 24 h, cultured cells were exposed to 4 different
rum (FCS) and 1% L-glutamine and 1% penicillin/streptomycin hyaluronic acid: Hyalgan MW 500–730 KDa, Artrosulfur HA®
(Gibco, Invitrogen, Life Technologies) at 37 °C in 5% CO2 and MW 1000, Hyalubrix® MW 1600 KDa, Synolis-VA® MW 2200
air with a change medium every 2–3 d. Tenocytes were then KDa, their features are shown in Table 1. Three different doses
harvested by StemPro Accutase (Life technologies Carlsbad, of Hyalgan or Artrosulfur HA® (250 μg/ml, 500 μg/ml and 1000
CA, USA), and centrifugated at 1,500 rpm for 5 min when the μg/ml), one doses of Hyalubrix® or Synolis-VA® (1000 μg/ml).
cells migrated out of tendon pieces and reached 60–80 % of HAPs were dissolved in the same culture media used for the
confluence (19 day). Collected tendon derived cells were im- entire experiments (α-MEM supplemented with 10 % FCS)
mediately used for culture to avoid phenotype drift with further and the Ph was adjusted to 7. Untreated cells were used as
in vitro passages [21]. The phenotype of the tendon derived control. All the cells (HAPs treated and untreated) were cultured
cells had not demonstrated significant drift as evidence by the in 1 ml of medium. Alamar blue dye test (Serotec, Oxford, UK)
gene expression pattern by assessing the expression of gene was performed to assess cell viability after 0, 24, and 48 h of
for scleraxis and genes for collagens α1(I), α2(I) and α1(III) in culture, as previous described [16]. The absorbance was read
real-time PCR assays with specific primers (data not shown). spectrophotometrically at 570 and 600 nm wavelengths by
MicroPlate reader (BioRad, Hercules, CA). The results, obtained
as optical density (OD) data, were processed following
Tenocyte viability and proliferation manufacturer’s instructions and expressed as reduction
In vitro proliferation was determined by the Alamar Blue assay.
percentage. The calculation of the of the percentage of alamar
This test was used to measure the metabolism rate of the cells.
blue reduction is as follows according to the manufacture’s
The tendon derived cells were seeded with 5×103 vital cells
protocol:
per well in a 96-well plate (Greiner CELL- STAR dish, Sigma-

a further 24 h. A negative control was prepared by incubating


cells in the absence of both inducing agent and HAPs. The
PE Annexin V/Dead Cell Apoptosis Kit with SYTOX® Green
for Flow Cytometry (Invitrogen, Life Technologies) was used
In the formula ελ1 and ελ2 are constant representing the to detect apoptosis by flow cytometry, cells were harvested,
molar extinction coefficient of alamar blue at 540 nm and 630 and processed according to the manufacturer’s instruction.
nm, respectively, in the oxidized (εox) and the reduced (εred) This product detects the externalization of phosphatidylserine
forms. Aλ1 and Aλ2 represent absorbance of test wells at 540 nm in apoptotic cells using recombinant annexin V conjugated to
and 630 nm, respectively. A’λ1 and A’λ2 represent absorbance the orange fluorescent phycobili-protein R-PE, and dead cells
of negative control wells at 540 and 630 nm, respectively. using SYTOX® Green nucleic acid stain. After treatment with
The values of % alamar blue reduction were corrected for both probes, apoptotic cells show orange fluorescence, dead
background values of negative controls containing medium cells show green fluorescence, and live cells show little or no
without cells. fluorescence. Fluorescence-activated cell sorting analysis
Finally, in parallel trypan blue exclusion assay was per- was carried out using a FC500 flow cytometer (FL1 and FL3
formed. The tendon derived cells were seeded with 104 vital detector in a log mode) using the CXP analysis software
cells per well in a 24-well plate (Greiner CELLSTAR dish, Sigma- (Beckmann Coulter, FL, USA).
Aldrich), and in triplicates in 1 ml of α-MEM supplemented with
10 % FCS. After 0, 24 and 48 h the cultures were detached,
collected and counted (Nikon Instruments INC., Melville, NY, Immunofluorescence staining
USA) in the Burker chamber with vital dye Trypan Blue (Stem The tendon derived cells were seeded with 5×103 vital cells
Cells Technologies, Vancouver, Canada) to evaluate cell per well in a 2-well chamber slides (Thermo Fisher Scientific,
viability. Inc., Rochester, NY, USA), in triplicates and cultured as previous
described [23]. After 1, 7 and 14 d of culture the tendon derived
cells were fixed with pure acetone for 10 min at −20 °C. Then,
Apoptosis induction washed a few minutes with PBS. Cells were incubated for 30
Hydrogen peroxide (H2O2) was used as an inducer of apoptosis min at room temperature with PBS containing 5 % of Bovine
as previously described [22]. The tendon derived cells were Serum Albumin (BSA) (Kedrion Group S.P.A., Lucca, Italy) for
seeded with 105 vital cells per well in a 6-well plate in 4 ml of protein blockage. Primary antibodies for Anti-type I (1:2000),
α-MEM supplemented with 10 % FCS. After 24 h, the medium Anti-type III collagen molecules (1:500) (Sigma-Aldrich), and
was removed, and the cultured cells were treated with H2O2 secondary antibodies fluorochrome were diluted in PBS
(2 mM) in α-MEM and 10 % FCS with or without Hyalgan®, containing 5 % BSA.
Artrosulfur HA®, Hyalubrix® and Synolis-VA® (1000 μg/ml) for

44
containing 5 % BSA. Cells were incubated overnight at 4 °C Comparison of individual treatment was conducted using
with primary antibodies, 1 h with the appropriate secondary Student’s t test. Statistical significance in comparison with the
antibody fluorochrome at room temperature and then washed corresponding control values was indicated by *P < 0.05 versus
a few times with PBS containing 5 % BSA. Molecule’s staining control.
Alexa Fluor 488 (Life Tecnologies) was used for type I collagen
and Alexa Fluor 568 (Life Tecnologies) for type III collagen.
After washing with PBS plus 5 % BSA, slides were mounted
Results
with 25 μL VECTASHIELD® Hard Set Mounting Medium Tendon derived cells viability
and then were examinated with ECLIPSE Ti-U inverted, Tendon derived cells morphology was evaluated under a
fluorescent microscope (Nikon Instruments INC., Melville, NY, light microscope at 0, 7 and 14 d. The cells maintained their
USA). For image analysis all digital images were captured normal, bipolar, spindle shape and cell processes, during the
with NIS-Elements Imaging Software (Nikon Instruments whole study period for each of the sets of culture conditions;
INC.). As previously described [23, 24], slides were examined cellular morphology remained unaltered for up to 14 d in all
independently by two experienced operator and one researcher, the experimental groups (Fig. 1a). Results from the trypan
with a double-blind method. The total fluorescence intensity of blue exclusion assay showed that none of the HAPs reduced
the area ≥ 10 frames from each slides was determined. The cell viability (Fig. 1b). After 48 h of exposure, living cells are
intensity level was normalized with the control cells untreated. in similar numbers when exposed to HAPs compared to the
Fully automated image analysis improve the accuracy of control. Metabolic tests provide some information concerning
detection and categorization of collagen staining, making this the activity of cells (Fig. 1c). Alamar blue confirmed an
technique more sensitive, specific and thus suitable for use in increase in the metabolic activity of tendon derived cells
quality assurance results. for all the HAPs utilized, when compared to untreated cells
(Fig. 1c). In particular, all HAPs induced cell-activity most
effectively at 1000 μg/ml (Fig. 1c, Table 2). The highest
Statistical analysis increase was obtained at 48 h for all the HA treatments.
Data are typical results from a minimum of three replicated
However, as reported in Fig. 1c, there are no significant
independent experiments and are expressed as mean ± SD.
statistical differences between all the various HAPs.

Fig. 1 Effects of Hyalgan®, Artrosulfur HA®, Hyalubrix® and Synolis-VA® on morphologic change cell viability and cell metabolic
activity. Tendon derived cells were treated with different concentrations of Hyalgan, Artrosulfur HA®, Hyalubrix® and Synolis-VA®
for different time periods. a. after treatment for 14 d the morphology of the cells treated with all the HAPs was photographed
(1000ug/ml). Magnification, ×100. b. Absolute number of live cells was calculated using trypan blue exclusion. c. Cell metabolic
activity was evaluated with the Alamar Blue method. All tests and determinations were repeated in triplicate. The metabolic activity
rate was calculated by subtracting the background OD value (complete culture medium without cells) from the OD value from
each test well (see materials and methods section). Hyalgan®, Artrosulfur HA®, Hyalubrix®, Synolis-VA® increased cell activity
in a concentration-dependent pattern and with regard to time. No statistically significant difference were observed from control
according to Student’s t test

55
Apoptosis induction Immunofluorescence staining
To verify whether or not they counteracted apoptosis in tendon Next, we determined and measured the type of collagen
derived cells, the Annexin V experiment was performed. Cells deposited by tendon derived cells after stimulation with
were plated and H2O2 induction was performed for 24 h to Hyalgan®, Artrosulfur HA®, Hyalubrix®, or Synolis-VA®.
induce apoptosis. Concurrently, tendon derived cells were Collagen accumulation was evaluated by immunofluorescent
separately exposed (or not, for the untreated sample) to staining of cells cultured on chamber slides. Furthermore,
Hyalgan®, Artrosulfur HA®, Hyalubrix®, and Synolis-VA® the expression of collagen type I was higher in Synolis-
(1000 μg/ml). Staining cells simultaneously with PE-Annexin V VA® than in the presence of Hyalgan®, Artrosulfur HA®,
(red fluorescence) and the non-vital dye, Sytox Green (green or Hyalubrix®, and was significantly higher compared
fluorescence), to untreated cells used as a control (Fig. 2a, b). In detail,
allowed, using bivariate analysis, discrimination between intact immunofluorescent staining at day 7 only revealed pro-
cells (Annexix V−Sytox Green−), early apoptotic (Annexix duction of collagen type I from the tendon derived cells in
V+Sytox Green−) and late apoptotic (Annexix V+Sytox Green+) intracytoplasmic staining (Fig. 3a). Moreover, at day 14 ex-
and necrotic cells (Annexix V−Sytox pression and production of Collagen type I had increased
Green+) (Fig. 2a). The treatment of tendon derived cells with in the Hyalgan®, Artrosulfur HA®, Hyalubrix® and Synolis-
Hyalgan®, Artrosulfur HA®, Hyalubrix®, or Synolis-VA® VA® (Fig. 3a, b). Moreover, Synolis-VA® induced the
(all at 1000 μg/ml) caused a sizable decrease in apoptosis, most significant expression of collagen type I after 14
as clearly shown in Fig. 2b. The percentage of vital cells, d (Fig. 3, Table 2). Collagen type III was not found to be
at 24 h following Hyalgan®, Artrosulfur HA®, Hyalubrix®, present in any culture conditions (data not shown).
or Synolis-VA® exposure, increased compared to the
control (33.14, 24.01, 36.31, 33.04 and 22.25 % respectively)
(Annexix V−, Sytox green−; bottom left quadrant) (Fig. 2c,
Table 2).

66
Fig. 2 The apoptosis rate in tendon derived cells. a. The apoptosis was induced with H2O2 (apoptotic control
cells [ACtrCs]) and treated with Hyalgan®, Artrosulfur HA®, Hyalubrix® and Synolis-VA® (1000 ug/ml).
Apoptosis rate of cells in each sample were detected in a population of 10,000 cells analyzed by flow cytometry.
b. The percentage of apoptotic cells in Hyalgan®, Artrosulfur HA®, Hyalubrix® and Synolis-VA® decreased
compared to the proliferate control cells (PCtrCs) (without HA). Flow charts: (A1), upper left quadrant annexin
V-negative and sytox green-positive cells indicate necrotic cells; (A2), upper right quadrant, annexin V and sytox
green-positive cells represent late apoptotic cells. (A3), lower left quadrant annexin V-negative and sytox green-
negative cells indicate live cells; (A4), lower right quadrant, annexin V positive and sytox green-negative cells
represent early apoptotic cells. Both early and late apoptotic cells were calculated as the incidence of apoptotic
cell. The numbers indicate percentage of total gated cells (mean ± S.D.; n = 3). c. % of cells population of live
cells, early apoptotic cells, late apoptotic cells and necrotic cells.
Graph showing the average % of results obtained in 3 independent experiment. No significant differences were
detected (mean ± S.D.; n = 3)

77
Discussion I, in a dose dependent manner over 14 d. This increase in
These results suggest a binary role for HA on tendon expression leads to collagen synthesis and accumulation.
cells - directly, on tendon derived cells metabolic activ- It should be noted that Synolis-VA® induced the most
ity, and on tenocyte Collagen type I production. It was significant expression of collagen type I at 14d of culture.
highlighted that Hyalgan, Artrosulfur HA®, Hyalubrix® In contrast, no HAPs induced any expression of collagen
and Synolis-VA® regulate cell activity of tendon derived type III, which is normally less abundant in tendons, and
cells. All HAPs increased cell-metabolic activity and most only increases in concentration during the early phase of
effectively at 1000 μg/ml and at 48 h (Fig. 1c). HA modulate remodeling [27] and in tendinopathy [20, 28]. The lack of
a number of biological process including cell apoptosis. collagen type III production under HA stress should be
The results show a decrease rate of the apoptosis when considered a protective factor for tendons. The results
the tendon derived cells were exposed to the HAPs. obtained are consistent with a previous study of Yamada
Collagen metabolism has been reported to be affected by and coworkers, even if a different methods focused directly
HA [25, 26]. HA stimulated the synthesis of collagen type on the collagen type I and III proteins was used [29].

Fig. 3 Collagen type I, expression of tendon derived cells in vitro culture isolated from 9 healthy patients, and staining as described
in material and methods, section Immunofluorescence Staining. Representative images from 4 independent experiments. a.
Expression of collagen type I after 7 and 14 d of culture in green fluorescence and in regular light image showing the tendon derived
cells. b. Quantization of the immunohistochemistry in a. Synolis-VA® induced the expression of collagen type I significantly after 14
d of in vitro culture. The mean fluorescent intensity/pixel was measured and
expressed to the corresponding tendon derived cells. Collagen type I Intensity (Total Area was quantified by anti-collagen type I) it
was measured by Nikon software. Data are expressed as mean ± SD for 4 independent experiments for samples run in triplicate.
Scale bar (a.): 50 μm

88
Considering the results of this study, the three different molecular effect of HAPs on collagen type I and III. As soon as possible
weight of HAPs tested seems not exert any effects on tendon there is need to widen the knowledge of the effects of HAPs on
derived cells in vitro, while is clear the essential importance of the main proteins of the extracellular matrix of tendons.
the concentrations and of the timing of exposure. The most Despite we tested the three different molecular weight HA,
significant expression of collagen type I at 14 d of culture of perhaps more HAPs and different concentration need to be
Synolis-VA® can be explained to the presence of Sorbitol (4 tested in the same way we did to confirm which should have
%) that limits the HA degradation, allowing an higher local the best in vitro results.
concentration of the drug. Translating these considerations in Obviously, it is advocate randomized control studies on the
the clinical practice, HA are effectives on human tenocytes and use of HA in the conservative treatment of tendinopathy and in
extracellular matrix of rotator cuff, with no essential differences selected patients with rotator cuff tears, in order to understand
among HA available in the market. There are many biological and clarify best timing, doses, intervals of injections and, finally,
questions that remain to be answered, and translational full clinical confirmation of effectiveness.
factors to resolve. Although, this in vitro model shows some
role played from HA on tendon derived cells, the study have Conclusion
some limitations. First of all, probably these results cannot be In conclusion, HAPs in dose dependent manner but not
generalized for other tendon derived cells from other sources; related to the molecular weight, induces increase of
furthermore in vitro environment, rich of nutrients and oxygen cells activities, decrease of apoptosis of tendon derived
is very different from the diseased environment. The complexity cells Collagen type I protein secretion. Taken together,
of the extracellular matrix of tendons and its relationship with these results strengthen a physiological role of HA in the
tenocytes during physiological homeostasis, disease and homeostasis of tendons and has implications for regen-
healing process, attest that is reductive to investigate only the erative medicine.

Competing interests
The authors declare that they have no competing interests.

Authors’ contributions
LO and ACB made substantial contributions to the conception and design of this manuscript, data acquisition,
analysis and interpretation and the drafting of the manuscript. MB carried out the Tendon cell cultures, Tendon
derived cells viability and metabolic activity. VdG Apoptosis analyses. GDB participated in tendon derived
cells culture. ACB conceived the study and participated in its design and coordination. FO critically revised
the manuscript for important intellectual content, and gave the final approval of the version to be published. All
authors read and approved the final manuscript.

Authors’ information
Not applicable.

Acknowledgements
The laboratory “Stem Cells” is supported by Fondazione PCFF ONLUS. The authors also wish to thank Blood
Donors Association FIDAS Pescara.

Author details
1Unit of Arthoscopy and Sports Trauma Surgery, Hesperia Hospital, Modena, Italy.
2U.O.C. of Immunohaematology and Transfusion Medicine, Laboratory of Stem Cells, Spirito Santo Hospital, via
Fonte Romana 8, 65125 Pescara, Italy.
3Department of Pharmacy, University G. d’Annunzio, Chieti, Italy. 4U.O.C. of Immunohaematology and
Transfusion Medicine, Santo Spirito Hospital, Pescara,
Italy. 5Department of Orthopedics and Traumatology, University of Rome “Tor Vergata” School of Medicine,
Rome, Italy.

Received: 10 June 2015 Accepted: 24 September 2015

99
Evaluarea conceptelor actuale
Leziuni ale tendoanelor si tendinopatia:
Vindecarea si Recuperarea
Pankaj Sharma, Mrcs, and Nicola Maffulli, Md, Ms, Phd, Frcs(Orth)

- Afectiunile tendoanelor sunt frecvente si sunt responsabile pentru morbiditate


substantiala, atat în domeniul sportului cat si la locul de munca.
- Tendinopatia, spre deosebire de tendinita sau tendinoze, este cel mai bun termen
generic descriptiv pentru conditiile clinice in si in jurul tendoanele care rezulta din
utilizarea excesiva a lor.
- Tendinopatia este o problema dificila, care necesita gestionare de lunga durata, iar
pacientii raspund adesea slab la tratament.
- Degenerarea preexistenta este considerata factor de risc pentru ruptura de tendon
acuta.
- Au fost dezvoltate mai multe modalitati fizice pentru a trata tendinopatia. Exista dovezi
limitate si mixte la nivel inalt pentru a sprijini, utilizarea clinica a acestor modalitati.
- Sunt necesare cercetarile suplimentare si evaluare stiintifica inainte ca solutiile
biologice sa devina optiuni reale.

Tendoanele conecteaza muschii la nivelul osului si permit transmiterea fortelor generate


de acestia la nivel osos, ceea ce are ca rezultat miscarea articulatiei.
Leziunile de tendon produc o morbiditate considerabila, iar handicapul pe care il
provoaca poate dura mai multe luni in ciuda a ceea ce este considerat management 1
adecvat. Problemele cronice cauzate de suprasolicitarea tendoanelor, au o probabilitate
de aparitie de 30% dintre toate leziunile legate de alergare 2 , iar prevalenta tendinopatiei
de cot la jucatorii de tenis poate atinge 40% 3. Biologia de baza a celulelor tendoanelor
nu este inca pe deplin inteleasa, iar gestionarea leziunilor de tendon reprezinta o
provocare considerabila pentru clinicieni.
Acest articol descrie functia si structura tendoanelor, revizuieste patofiziologia leziunilor
de tendon si fazele de vindecare ale tendonului precum si posibile strategii pentru
optimizarea vindecarii si repararatiei tendoanelor.

10
10
THE JOURNAL OF BONE & JOINT SURGERY

JB&JS
Tendon Injury and tendinopathy : Healing and Repair
Pankaj Sharma and Nicola Maffulli
J. Bone Joint Surg. Am. 87:187-202, 2005. doi:10.2106/JBJS.D.01850

Letters to The Editor are available at


http://www.ejbjs.org/cgi/content/full/87/1/187#responses

subject Collections Articles on similar topics can be found in the following collections

Reprints and Permissions Adult Trauma (508 articles)


Trauma (302 articles)
Soft Tissue Trauma (218 articles)
Surgical Procedures (697 articles)
Physiology (61 articles)

Publisher Information The Journal of Bone and Joint Surgery


20 Pickering Street, Needham, MA 02492-3157
www.jbjs.org

11
11
Current Concepts Review
Tendon Injury and
Tendinopathy: Healing and
Repair
BY PANKAJ SHARMA, MRCS, AND NICOLA MAFFULLI, MD, MS, PHD, FRCS(ORTH)

- Tendon disorders are frequent and are responsible for substantial morbidity both in sports and
in the workplace.
- Tendinopathy, as opposed to tendinitis or tendinosis, is the best generic descriptive term for the
clinical conditions in and around tendons arising from overuse.
- Tendinopathy is a difficult problem requiring lengthy management, and patients often respond
poorly to treatment.
- Preexisting degeneration has been implicated as a risk factor for acute tendon rupture.
- Several physical modalities have been developed to treat tendinopathy. There is limited and
mixed high-level evidence to support the, albeit common, clinical use of these modalities.
- Further research and scientific evaluation are required before biological solutions become
realistic options.

Tendons connect muscle to bone and allow transmission of al cells and smooth muscle cells of arterioles. Tenocytes are ac-
forces generated by muscle to bone, resulting in joint move- tive in energy generation through the aerobic Krebs cycle, an-
ment. Tendon injuries produce considerable morbidity, and aerobic glycolysis, and the pentose phosphate shunt, and they
the disability that they cause may last for several months de- synthe- size collagen and all components of the extracellular
spite what is considered appropriate management1. Chronic matrix network6-8. With increasing age, metabolic pathways
problems caused by overuse of tendons probably account for shift from aerobic to more anaerobic energy production9,10.
30% of all running-related injuries2, and the prevalence of el- The oxygen consumption of tendons and ligaments is
bow tendinopathy in tennis players can be as high as 40%3. 7.5 lower than that of skeletal muscles11. The low meta-
The basic cell biology of tendons is still not fully understood, bolic rate and well-developed anaerobic energy-generation
and the management of tendon injury poses a considerable capacity are essential to carry loads and maintain tension for
challenge for clinicians. This article describes the function and long periods, reducing the risk of ischemia and subsequent
structure of tendons, reviews the pathophysiology of tendon necrosis.
injury and the phases of tendon healing, and reviews possible However a low metabolic rate results in slow healing
strategies for optimizing tendon healing and repair. after injury12.
The dry mass of human tendons is approximately 30% of
the total tendon mass, with water accounting for the remaining
Tendon Structure 70%. Collagen type I accounts for 65% to 80% and elastin ac-
Healthy tendons are brilliant white in color and have a fibroelas-
counts for approximately 2% of the dry mass of tendons6,13-15.
tic texture. Tendons demonstrate marked variation in form; they
Tenocytes and tenoblasts lie between the collagen fibers along
can be rounded cords, straplike bands, or flattened ribbons4.
the long axis of the tendon16.
Within the extracellular matrix network, tenoblasts and teno-
Collagen is arranged in hierarchical levels of increasing
cytes constitute about 90% to 95% of the cellular ele- ments
complexity, beginning with tropocollagen, a triple-helix poly-
of tendons5. Tenoblasts are immature tendon cells. They are
peptide chain, which unites into fibrils; fibers (primary bun-
spindle-shaped and have numerous cytoplasmic organelles,
dles); fascicles (secondary bundles); tertiary bundles; and the
reflecting their high metabolic activity5. As they mature, teno-
tendon itself (Fig. 1)17-19. Soluble tropocollagen molecules
blasts become elongated and transform into tenocytes5. Teno-
form cross-links to create insoluble collagen molecules, which
cytes have a lower nucleus-to-cytoplasm ratio than tenoblasts,
aggregate to form collagen fibrils. A collagen fiber is the small-
with decreased metabolic activity5. The remaining 5% to 10%
est tendon unit that can be tested mechanically and is visible
of the cellular elements of tendons consists of chondrocytes at
under light microscopy. Although collagen fibers are mainly
the bone attachment and insertion sites, synovial cells of the
tendon sheath, and vascular cells, including capillary endotheli-

12
12
oriented longitudinally, fibers also run transversely and hori- ultrafiltration membrane to produce synovial fluid33. The fibrous
zontally, forming spirals and plaits20-22. sheath forms condensations, the pulleys, which function as
The ground substance of the extracellular matrix network fulcrums to aid tendon function34.
surrounding the collagen and the tenocytes is composed of At the myotendinous junction, tendinous collagen fibrils are
proteoglycans, glycosaminoglycans, glycoproteins, and several inserted into deep recesses formed by myocyte processes,
other small molecules5. Proteoglycans are strongly hydrophilic, allowing the tension generated by intracellular contractile
enabling rapid diffusion of water-soluble molecules and the proteins of muscle fibers to be transmitted to the collagen
migration of cells. Adhesive glycoproteins, such as fibronectin fibrils35-39. This complex architecture reduces the tensile stress
and thrombospondin, participate in repair and regeneration exerted on the tendon during muscle contraction35. However,
processes in tendon20,23,24. Tenascin-C, another important the myotendinous junction still remains the weakest point of the
component of the tendon extracellular matrix network, is muscle-tendon unit35,39-42.
abundant in the tendon body and at the osteotendinous and The osteotendinous junction is composed of four zones: a
myotendinous junctions25,26. Tenascin-C contains a number of dense tendon zone, fibrocartilage, mineralized fibrocartilage,
repeating fibronectin type-III domains, and, fol- lowing stress- and bone43. The specialized structure of the osteotendinous
induced unfolding of these domains, it also functions as an junction prevents collagen or fiber bending, fraying, shearing,
elastic protein26,27. The expression of tenascin-C is regulated and failure44,45.
by mechanical strain and is upregulated in tendinopathy25,28,29.
Tenascin-C may play a role in collagen fiber alignment and Blood Supply
orientation30. Tendons receive their blood supply from three main sources:
The epitenon, a fine, loose connective-tissue sheats the intrinsic systems at the myotendinous junction and osteo-
containing the vascular, lymphatic and nerve supply to tendinous junction, and the extrinsic system through the
the tendon, covers the whole tendon and extends deep paratenon or the synovial sheath46,47. The ratio of blood supply
within it between the tertiary bundles as the endotenon. from the intrinsic systems to that from the extrinsic system
The endotenon is a thin reticular network of connective varies from tendon to tendon. For example, the central third
tissue investing each tendon fiber31-32. Superficially, the of the rabbit Achilles tendon receives 35% of its blood supply
epitenon is surrounded by paratenon, a looser areolar from the extrinsic system48,49. At the myotendinous junction,
connective tissue consisting of type I and type III collagen perimysial vessels from the muscle continue between the fasci-
fibrils, some elastic fibrils, and an inner lining synovial cles of the tendon25. However, blood vessels originating from
cells9. Synovial tendon sheaths are found in areas subjected to the muscle are unlikely to extend beyond the proximal third of
increased mechanical stress, such as tendons of the hands and the tendon46. The blood supply from the osteotendinous junc-
feet, where efficient lubrication is required. Synovial sheaths tion is sparse and is limited to the insertion zone of the ten-
consist of an outer fibrotic sheath and an inner synovial sheath, don, although vessels from the extrinsic system communicate
which consists of thin visceral and parietal sheets18. The inner with periosteal vessels at the osteotendinous junction5,46.
synovial sheath invests the tendon body and functions as an In tendons enveloped by sheaths to reduce friction,

Fig. 1
Anatomy of a normal tendon.

13
13
Fig. 2
Stress-strain curve demonstrating the basic physical properties of a tendon

branches from major vessels pass through the vincula (meso- the epitenon. Most nerve fibers do not actually enter the main
tenon) to reach the visceral sheet of the synovial sheath, where body of the tendon but terminate as nerve endings on its
they form a plexus18 that supplies the superficial part of the surface.
tendon, while some vessels from the vincula penetrate the Nerve endings of myelinated fibers function as specialized
epitenon. These penetrating vessels course in the endotenon mechanoreceptors to detect changes in pressure or tension.
septa and form a connection between the peritendinous and These mechanoreceptors, the Golgi tendon organs, are most
intratendinous vascular networks. numerous at the insertion of tendons into the muscle55,56.
In the absence of a synovial sheath, the paratenon provides Golgi tendon organs are essentially a thin delicate capsule of
the extrinsic component of the vasculature. Vessels entering connective tissue that encloses a group of branches of large
the paratenon course transversely and branch repeatedly to myelinated nerve fibers. These fibers terminate with a spray of
form a complex vascular network50. Arterial branches from the fiber endings between bundles of collagen fibers of the ten-
paratenon penetrate the epitenon to course in the endotenon don57,58. Unmyelinated nerve endings act as nociceptors, and
septa, where an intratendinous vascular network with abundant they sense and transmit pain. Both sympathetic and parasym-
anastomoses is formed5,51. pathetic fibers are present in tendon59.
Tendon vascularity is compromised at junctional zones and
sites of torsion, friction, or compression. In the Achilles tendon,
angiographic injections techniques have demonstrated
Biomechanics
Tendons transmit force from muscle to bone and act as a
a zone of hypovascularity 2 to 7 cm proximal to the
buffer by absorbing external forces to limit muscle damage60.
tendon injection46,52 . However, laser Doppler flowmetry
Tendons exhibit high mechanical strength, good flexibility, and
has demonstrated substantially reduced blood flow near the
an optimal level of elasticity to perform their unique role16,61,62.
Achilles tendon insertion, with an otherwise even blood flow
Tendons are viscoelastic tissues that display stress re- laxation
throughout the tendon53. A similar zone of hypovascularity is
and creep63,64.
present on the dorsal surface of the flexor digitorum profundus
The mechanical behavior of collagen depends on the number
tendon subjacent to the volar plate, within 1 cm of the tendon
and types of intramolecular and intermolecular bonds65.
insertion54.In general, tendon blood flow decreases with
A stress-strain curve helps to demonstrate the behavior of
increasing age and mechanical loading53.
tendon (Fig. 2). At rest, collagen fibers and fibrils display a
crimped configuration66. The initial concave portion of the
Tendon Innervation curve (toe region), where the tendon is strained up to 2%, rep-
Tendon innervation originates from cutaneous, muscular, and resents flattening of the crimp pattern13,67,68. Beyond this point,
peritendinous nerve trunks. At the myotendinous junction, tendons deform in a linear fashion as a result of intramolecular
nerve fibers cross and enter the endotenon septa. Nerve fibers sliding of collagen triple helices, and the fibers become more
form rich plexuses in the paratenon, and branches penetrate parallel69,70. If the strain remains <4%, the tendon behaves in an

14
14
elastic fashion and returns to its original length when unloaded71. frictional forces between the fibrils and causing localized fiber
Microscopic failure occurs when the strain exceeds 4%. damage96.
Beyond 8% and 10% strain, microscopic failure occurs The etiology of tendinopathy remains unclear, and many
from intrafibril damage by molecular slippage61,67,72. causes have been theorized17,89. Ischemia occurs when
X-ray diffraction studies have demonstrated that collagen fibril a tendon is under maximal tensile load. On relaxation,
elongation initially occurs as a result of molecular elongation, reperfusion occurs, generating oxygen free radicals97,98;
but as stress increases, the gap between molecules increases, this may cause tendon damage, resulting in tendinopathy
eventually leading to slippage of lateral adjoining molecules73. Peroxiredoxin 5 is an antioxidant enzyme that protects
After this, complete failure occurs rapidly, and the fibers recoil cells against damage from such reactive oxygen species.
into a tangled bud at the ruptured end60. Peroxiredoxin 5 is found in human tenocytes. Its expression
The tensile strength of tendons is related to thickness and is increased in tendinopathy, a finding that supports the
collagen content, and a tendon with an area of 1 cm2 is capable view that oxidative stress may play a role99. Hypoxia alone
of bearing 500 to 1000 kg31,74,75. During strenuous activities may also result in degeneration, as tendons rely on oxidative
such as jumping and weight-lifting, very high loads are placed energy metabolism to maintain cellular ATP levels100. During
on tendons76. Forces of 9 kN, corresponding to 12.5 times vigorous exercise, localized hypoxia may occur in tendons,
body weight, have been recorded in the human Achilles tendon with tenocyte death.
during running77-79. Since these forces exceed the single-load During locomotion, tendons store energy, 5% to 10% of
ultimate tensile strength of the tendon, the rate of loading may which is converted into heat101,102. In the equine superficial
also play an important role in tendon rupture67,79. Tendons are digital flexor tendon, temperatures of up to 45°C have been
at the highest risk for rupture if tension is applied quickly and recorded during galloping103. Although short periods at 45°C
obliquely, and the highest forces are seen during eccentric are unlikely to result in tenocyte death, repeated hyperthermic
muscle contraction65,80-84. insults and prolonged hyperthermia may compromise cell via-
bility and lead to tendon degeneration104,105.
Excessive tenocyte apoptosis, the physiological process
Tendon Injury often referred to as “programmed cell death,” has been
Tendon injuries can be acute or chronic and are caused by implicated in rotator cuff tendinopathy106. Application of
in- trinsic or extrinsic factors, either alone or in combination. In strain to tenocytes produces stress-activated protein kinases,
acute trauma, extrinsic factors predominate. which in turn trigger apoptosis107,108. Oxidative stress may play
a role in inducing apoptosis, but the precise details remain to
Tendon Rupture be elucidated109. There are more apoptotic cells in ruptured su-
An acceleartion-decelaration mechanism has been reported praspinatus tendons than in normal subscapularis tendons110.
in up to 90% of sports-related Achilles tendon ruptures85. Tendinopathic quadriceps femoris tendons exhibited a rate of
Malfunction of the normal protective inhibitory pathway of the spontaneous apoptosis that was 1.6 times greater than that of
musculotendinous unit may result in injury86. The etiology of normal tendons111.
tendon rupture remains unclear12. Degenerative tendinopathy In animal studies, local administration of cytokines and
is the most common histological finding in spontaneous ten- inflammatory prostaglandins produced a histological picture
don ruptures. Arner et al. reported degenerative changes in all of tendinopathy112,113. Application of cyclic strain increases
of their seventy-four patients with an Achilles tendon rupture, production of prostaglandin E2 (PGE2) in human patellar
and they hypothesized that those changes were due to intrinsic tenocytes , and it increases interleukin-6 (IL-6) secretion115 and
abnormalities that had been present before the rupture87. Kan- IL-1ß gene expression in human flexor tenocytes116. Human
nus and Jozsa found degenerative changes in 865 (97%) of 891 flexor tendon cells treated with IL-1ß produced increased
tendons that had spontaneously ruptured, whereas degenera- mRNA for cycloox-ygenase-2, matrix metalloproteinase-1
tive changes were seen in 149 (33%) of 445 control tendons10. (MMP-1), MMP-3, and PGE2 . IL-1ß released on mechanical
Tendon degeneration may lead to reduced tensile strength and stretching of rabbit Achilles tendons results in increased
a predisposition to rupture. Indeed, histological evaluation of production of MMP-3 (stromelysin-1)118. Hence, prolonged
ruptured Achilles tendons has demonstrated greater degenera- mechanical stimuli induce production of cytokines and
tion than was found in tendons that were chronically painful as inflammatory prostaglandins, which may be mediators of
a result of an overuse injury88. tendinopathy.
Ciprofloxacin also induces IL-1ß-mediated MMP-3 release,
Tendinopathy and use of fluoroquinolone is associated with tendon rupture
Overuse injuries generally have a multifactorial origin. Interac- and tendinopathy119-121. Fluoroquinolones inhibit tenocyte
tion between intrinsic and extrinsic factors is common in metabolism, reducing cell proliferation and collagen
chronic tendon disorders12. It has been claimed that intrinsic and matrix synthesis a mechanism that may induce
factors such as alignment and biomechanical faults play a tendinopathy122,123.
causative role in two-thirds of Achilles tendon disorders MMPs, a family of proteolytic enzymes124, are classified
in athletes19,90. In particular, hyperpronation of the foot according to their substrate, specificity, and primary structure.
has been linked with an increased prevalence of Achilles They have the combined ability to degrade the components
tendinopathy91,92. Excessive loading of tendons during vigorous of the extracellular matrix network and to facilitate tissue
physical training is regarded as the main pathological stimulus remodeling125-127. Downregulation of MMP-3 mRNA has
for degeneration93, and there may be a greater risk of excessive been reported in Achilles tendinopathy128,129. Alfredson et al.
loading inducing tendinopathy in the presence of intrinsic risk found, in addition to downregulation of MMP-3, upregulation
factors. Tendons respond to repetitive overload beyond the of MMP-2 (gelatinase A) and vascular endothelial growth
physiological threshold with either inflammation of their sheath factor (VEGF) in Achilles tendinopathy compared with control
or degeneration of their body, or both94. Different stresses samples129. Decreased MMP-3 and MMP-2 activity, but in-
induce different responses. Unless fatigue damage is actively creased MMP-1 (collagenase-1) activity, has been reported in
repaired, tendons will weaken and eventually rupture95. The ruptured supraspinatus tendons130. However, a rabbit model of
repair mechanism is probably mediated by resident tenocytes, supraspinatus tears showed increased expression of MMP-2
which maintain a fine balance between extracellular matrix and TIMP-1 (tissue inhibitor of metalloproteinase-1)131.
network production and degradation. Tendon damage may Failure to adapt to recurrent excessive loads may result in
even occur from stresses within the physiological limits, as release of cytokines by tenocytes, leading to further modula-
frequent cumulative microtrauma may not allow enough time tion of cell activity132. An increase in cytokine levels in response
for repair93. Microtrauma can also result from nonuniform stress to repeated injury or mechanical strain may induce MMP
within tendons, producing abnormal load concentrations and release, with degradation of the extracellular matrix network

15
15
and eventual tendinopathy. Mechanical loading studies have The role played by inflammation in tendon rupture is less
varied with regard to the strain protocol used, and direct clear.
comparison of their results is often difficult. The amount and
frequency of application of strain may in fact determine the Pain in Tendinopathy
type and amount of cytokines released. Although an imbalance Classically, pain in tendinopathy was attributed to
in MMP activity has been demonstrated in tendinopathic and inflammation. However, chronically painful Achilles and
ruptured tendons, differences in expression of the various patellar tendons show no evidence of inflammation,
MMPs have been reported125-131. A differential temporal and many tendons with intratendinous lesions detected
sequence of MMP expression may occur, and MMP expression on magnetic resonance imaging or ultrasound are not
may differ between tendinopathic and ruptured tendons. painful149. Pain may originate from a combination
of mechanical and biochemical factors149. Tendon
Histological Changes in Tendinopathy degeneration with mechanical breakdown of collagen
The term “tendinosis” has been in use for nearly three could theoretically explain the pain, but clinical and surgical
decades to describe the pathological features of the observations have challenged this view149. Chemical irritants
extracellular matrix network in tendinopathy133. Despite and neurotransmitters may generate pain in tendinopathy,
that, most clinicians still use the term “tendinitis” or and microdialysis sampling has revealed a twofold increase
“tendonitis”, thus implying that the fundamental problem in lactate levels in tendons with tendinopathy compared
is inflammatory. We advocate the use of the term with those in controls156. Patients with chronic Achilles
“tendinopathy” as a generic descriptor of the clinical tendinopathy and patellar tendinopathy showed high
conditions in and around tendons arising from overuse, and concentrations of the neurotransmitter glutamate, with no
we suggest that the terms “tendinosis” and “tendinitis” significant elevation of the proinflammatory prostaglandin
be used only after histopathological examination134. PGE2157. However, the levels of PGE2 were consistently higher
Histological examination of tendinopathy shows disor- in the tendinopathic tendons than they were in controls, and it
dered, haphazard healing with an absence of inflammatory is possible that the results lacked significance because of the
cells, a poor healing response, noninflammatory intratendi- small sample size of the study.
nous collagen degeneration, fiber disorientation and Substance P functions as a neurotransmitter and neuro-
thinning, hypercellularity, scattered vascular ingrowth, modulator, and it is found in small unmyelinated sensory
and increased interfibrillar glycosaminoglycans18,135-137. nerve fibers158. A network of sensory innervation is present in
Frank inflammatory lesions and granulation tissue tendons, and substance P has been found both in tendino-
are infrequent and are mostly associated with tendon pathic Achilles tendons and in medial and lateral epicondy-
ruptures138. lopathy159-162. Sensory nerves transmit nociceptive information
Various types of degeneration may be seen in tendons, to the spinal cord, and increased levels of substance P correlate
but mucoid or lipoid degeneration is usually found in the with pain levels in rotator cuff disease162.
Achilles tendon18,139. Light microscopy of a tendon with An opioid system has been demonstrated in the Achilles
mucoid degeneration reveals large mucoid patches and tendons of rats163. Under normal conditions, there is probably
vacuoles between fibers. In lipoid degeneration, abnormal a balance between nociceptive and anti-nociceptive pep-
intratendinous accumulation of lipid occurs, with disruption tides164,165, with alteration of this equilibrium in pathological
of collagen fiber structure18,140,141. In rotator cuff tendinopathy, conditions164,165.
mucoid degeneration occurs, but fibrocartilaginous metaplasia,
often accompanied by calcium deposition, is also common147. Tendon Healing
Amyloid deposition in supraspinatus tendons with degenerative Studies of tendon healing predominantly have been per-
tears has also been reported148. formed on transected animal tendons or ruptured human ten-
Tendinosis can be viewed as a failure of the cell matrix dons, and their relevance to healing of tendinopathic human
to adapt to a variety of stresses as a result of an imbalance tendons remains unclear.
be- tween matrix degeneration and synthesis93,132. Macroscopi- Tendon healing occurs in three overlapping phases. In the
cally, the affected portions of the tendon are seen to have lost initial, inflammatory phase, erythrocytes and inflammatory
their normal glistening-white appearance and to have become cells, particularly neutrophils, enter the site of injury. In
gray-brown and amorphous. Tendon thickening, which can the first twenty-four hours, monocytes and macrophages pre-
be diffuse, fusiform, or nodular, occurs149. Tendinosis is often dominate and phagocytosis of necrotic materials occurs. Vaso-
clinically silent, and its only manifestation may be a rupture; active and chemotactic factors are released with increased
however, it may also coexist with symptomatic paratendin- vascular permeability, initiation of angiogenesis, stimulation of
opathy98,150-152. Mucoid degeneration, fibrosis, and vascular tenocyte proliferation, and recruitment of more inflammatory
pro- liferation with a slight inflammatory infiltrate have been cells166. Tenocytes gradually migrate to the wound, and type-III
reported in paratendinopathy12,153,154. Edema and hyperemia of collagen synthesis is initiated167.
the paratenon are seen clinically. A fibrinous exudate accumu- After a few days, the proliferative phase begins. Synthesis
lates within the tendon sheath, and crepitus may be felt on of type-III collagen peaks during this stage and lasts for a few
clinical examination149. weeks. Water content and glycosaminoglycan concentrations
In samples from 397 ruptured Achilles tendons, Kannus remain high during this stage167.
and Jozsa found no evidence of inflammation under light and After approximately six weeks, the remodeling phase
electron microscopy10. Arner et al. also found no neutrophilic commences, with decreased cellularity and decreased
infiltration in Achilles tendons on the first day after rupture, and collagen and glycosaminoglycan synthesis. The remodeling
they concluded that any inflammation seen at a later stage phase can be divided into a consolidation stage and a
occurred subsequent to the rupture87. In a recent study, im- maturation stage168. The consolidation stage begins at about
munohistochemical staining of neutrophils confirmed acute six weeks and continues for up to ten weeks. In this period,
inflammation in all of sixty ruptured Achilles tendons155. Col- the repair tissue changes from cellular to fibrous. Tenocyte
lagen degeneration and tenocyte necrosis may trigger an acute metabolism remains high during this period, and tenocytes and
inflammatory response, which further weakens the tendon, collagen fibers become aligned in the direction of stress169. A
predisposing it to rupture. higher proportion of type-I collagen is synthesized during this
In summary, tendinopathy shows features of disordered stage170. is synthesized during this stage170. After ten weeks,
healing, and inflammation is not typically seen. Although de- the maturation stage occurs, with gradual change of the
generative changes do not always lead to symptoms, preexist- fibrous tissue to scar-like tendon tissue over the course
ing degeneration has been implicated as a risk factor for acute of one year169,171. During the latter half of this stage, tenocyte
tendon rupture10,87,88. metabolism and tendon vascularity decline172.

16
16
Tendon healing can occur intrinsically, by proliferation of and protein extravasation209-211. In addition, substance P
epitenon and endotenon tenocytes, or extrinsically, by invasion enhances cellular release of prostaglandins, histamines, and
of cells from the surrounding sheath and synovium173-175. cytokines212,213. Levels of substance P and CGRP peak during
Epitenon tenoblasts initiate the repair process through prolifer- the proliferative phase, suggesting a possible role during that
ation and migration176-179. Healing of severed tendons can be phase.
achieved by cells from the epitenon alone, without reliance
on adhesions for vascularity or cellular support180,181. Internal Limitations of Healing
tenocytes contribute to the intrinsic repair process and secrete Adhesion formation after intrasynovial tendon injury poses a
larger and more mature collagen fibers than do epitenon major clinical problem214. Disruption of the synovial sheath at
cells182. Despite this, fibroblasts in the epitenon and tenocytes the time of the injury or surgery allows granulation tissue and
synthesize collagen during repair, and different cells probably tenocytes from surrounding tissue to invade the repair site.
produce different collagen types at different time-points. Exogenous cells predominate over endogenous tenocytes, al-
Initially, collagen is produced by epitenon cells, with endotenon lowing the surrounding tissue to attach to the repair site and
cells synthesizing collagen later183-187. The relative contribution resulting in adhesion formation.
of each cell type may be influenced by the type of trauma Despite remodeling, the biochemical and mechanical
sustained, the anatomical location, the presence of a synovial properties of healed tendon tissue never match those of intact
sheath, and the amount of stress induced by motion after repair tendon. In a study of transected sheep Achilles tendons that
has taken place188. had spontaneously healed, the rupture force was only 56.7%
Tenocyte function may vary depending on the region of of normal at twelve months215. One possible reason for this
origin. Cells from the tendon sheath produce less collagen and is the absence of mechanical loading during the period of
glycosaminoglycans than do epitenon and endotenon cells. immobilization.
However, fibroblasts from the flexor tendon sheath proliferate
more rapidly189,190. The variation in phenotypic expression of Current Strategies for Tendon Healing
tenocytes has not been extensively investigated, and this infor- Physical Modalities
mation may prove useful for optimizing repair strategies. Many physical modalities are used in the management of
Intrinsic healing results in better biomechanics and fewer ten- don disorders. However, although these modalities are in
complications;in particular, a normal gliding mechanism rou- tine clinical use, only a few controlled clinical trials have
within the tendon sheath is preserved191. In extrinsic been performed. Most of the evidence is still pre-clinical and,
healing, scar tissue results in adhesion formation, which at times, controversial.
disrupts tendon gliding192. Different healing patterns may Extracorporeal shock wave therapy applied to rabbit Achilles
predominate in particular locations; for example, extrinsic tendons, at a rate of 500 impulses of 14 kV in twenty
healing tends to prevail in torn rotator cuffs193. minutes, resulted in neovascularization and an increase in the
MMPs are important regulators of extracellular matrix angiogenesis-related markers such as nitric oxide synthase
network remodeling, and their levels are altered during ten- and VEGF216. Extracorporeal shock wave therapy promoted
don healing126-128. In a rat flexor tendon laceration model, the healing of experimental Achilles tendinopathy in rats217. The au-
expression of MMP-9 and MMP-13 (collagenase-3) peaked thors proposed that the healing improved because of an
between the seventh and fourteenth days after the surgery. increase in growth factor levels, as they had noted elevated
MMP-2, MMP-3, and MMP-14 (MT1-MMP) levels increased lev- els of transforming growth factor-ß1 (TGF-ß1) in the early
after the surgery and remained high until the twenty-eighth stage and persistently elevated levels of IGF-1217. In another
day194. These findings suggest that MMP-9 and MMP-13 study, seventy-four patients with chronic noncalcific rotator
participate only in collagen degradation, whereas MMP-2, cuff tendinopathy were randomized to receive either active
MMP- 3, and MMP-14 participate both in collagen degradation ex- tracorporeal shock wave therapy (1500 pulses of 0.12 mJ/
and in collagen remodeling. Wounding and inflammation also mm2) or sham treatment monthly for three months218. The
provoke release of growth factors and cytokines from platelets, mean duration of symptoms was 23.3 months in both groups.
polymorphonuclear leukocytes, macrophages, and other in- All patients were assessed for pain in the shoulder, including
flammatory cells195-200. These growth factors induce neovascu- night pain measured with a visual analogue score, and a dis-
larization and chemotaxis of fibroblasts and tenocytes and ability index was calculated before each treatment and at one
stimulate fibroblast and tenocyte proliferation as well as syn- and three months after the completion of the treatment. There
thesis of collagen201,202. were no significant differences between the two groups before
Nitric oxide is a short-lived free radical with many bio- treatment. Both groups showed marked and sustained im-
logical functions: it is bactericidal, it can induce apoptosis provements from two months onward, but the moderate doses
in inflammatory cells, and it causes angiogenesis and of extracorporeal shock wave therapy provided no added
vasodilation203-205. Nitric oxide may play a role in several benefit compared with the sham treatment.
aspects of tendon healing. Nitric oxide synthase is responsible In a double-blind, randomized, placebo-controlled trial
for synthesizing nitric oxide from L-arginine. Levels of nitric of 144 patients with calcific tendinopathy of the rotator cuff,
oxide synthase peaked after seven days and returned to patients received high-energy extracorporeal shock wave
baseline fourteen days after tenotomies of rat Achilles therapy, low-energy extracorporeal shock wave therapy, or a
tendons206. In that study, inhibition of nitric oxide synthase placebo (sham treatment)219. The two groups treated with ex-
reduced healing, resulted in a decreased cross-sectional area, tracorporeal shock wave therapy received the same cumulative
and reduced failure load206. The authors did not identify the energy dose. All patients received two treatment sessions ap-
specific isoforms of nitric oxide synthase. More recently, the proximately two weeks apart, followed by physical therapy.
same research group demonstrated a temporal expression of Both the high-energy and the low-energy extracorporeal shock
the three isoforms of nitric oxide synthase207. The inducible wave therapy resulted in an improvement in the mean Constant
isoform peaks on the fourth day, the endothelial isoform and Murley score at six months compared with the score after
peaks on the seventh day, and the neuronal isoform peaks on the sham treatment. Also, the patients who had re- ceived the
the twenty-first day207. Interestingly, in a rat Achilles tendon high-energy extracorporeal shock wave therapy had a higher
rupture model, nerve fiber formation peaked between two six-month Constant and Murley score than did the patients
and six weeks after the rupture, in concert with peak levels of who had received the low-energy extracorporeal shock wave
the neuronal isoform of nitric oxide synthase208. These nerve therapy. Compared with the placebo, both the high-energy and
fibers presumably deliver neuropeptides that act as chemical the low-energy extracorporeal shock wave therapy appeared to
messengers and regulators, and they may play an important provide a beneficial effect in terms of bet- ter shoulder function,
role in tendon healing. Substance P and calcitonin gene-related less self-rated pain, and diminished size of calcifications. Also,
peptide (CGRP) are proinflammatory and cause vasodilation the high-energy extracorporeal shock wave therapy appeared

17
17
to provide a beneficial effect in terms of better shoulder function, well. With these methods, sustained gene expression seems
less self-rated pain, and diminished size of calcifications. to last for about six weeks, possibly long enough for clinical
Also, the high-energy extracorporeal shock wave therapy applications254,255. Ex vivo gene transduction is possibly more
appeared to be superior to the low-energy therapy. However, efficient, but the techniques must be optimized.
caution should be exercised when using extra-corporeal shock Gene therapy can also alter the healing environment
wave therapy, as dose-dependent tendon damage, including of tendons in animal models of tendon repair. Adenoviral
fibrinoid necrosis, fibrosis, and inflammation, has been transduction of focal adhesion kinase (FAK) into partially
reported in rabbits220. lacerated chicken flexor tendons resulted in an expected
Pulsed magnetic fields with a frequency of 17 Hz im- increase in adhesion formation and a twofold increase in the
proved collagen fiber alignment in a rat Achilles tendinopathy work required for flexion compared with the results in control
model221. In another study, tenotomized rat Achilles tendons groups256. These differences were significant (p = 0.001). While
were sutured and then treated with low-intensity galvanic cur- tendon healing was not improved in this study, the re- sults
rent for fifteen minutes a day for two weeks222. Biomechanical did demonstrate that the healing environment and con- ditions
analysis revealed an increased force to breakage in the anode- could be manipulated.
stimulated group compared with controls and a cathode-stim- Bone morphogenetic protein-12 (BMP-12) is the human
ulated group. analogue of murine GDF-7257. BMP-12 increases the expression
Direct current applied to rabbit tendons in vitro in- of procollagen type-I and III genes in human patellar tenocytes,
creased type-I-collagen production and decreased adhesion and it is found at sites of tendon remodeling258. BMP-12
formation223. In a randomized trial, lacerated rabbit flexor ten- increased synthesis of type-I collagen by 30% in chicken
dons were repaired and then received pulsed electromagnetic flexor tenocytes, and application of tenocytes transfected with
field stimulation for six hours a day, starting six days after the the BMP-12 gene to a chicken flexor tendon laceration model
surgery and continuing until twenty-one days after the resulted in a twofold increase in tensile strength and load to
surgery224. At four weeks, no difference in adhesion formation failure at four weeks259.
was noted. Transfer of genes to tendons is feasible, and, as the healing
The effects of laser therapy on tendon healing have also environment can be manipulated for up to eight to ten weeks226,
been studied. Laser phototherapy increased collagen produc- this may be long enough to be clinically relevant. While the
tion in rabbits subjected to tenotomy and surgical repair225. above studies were conducted in tendon transection models,
In a placebo-controlled, double-blind, prospective study of delivery of substances such as platelet-derived growth factor-B
twenty-five patients with a total of forty-one digital flexor (PDGF-B), BMP-12, and decorin may improve healing of
tendon repairs, laser therapy reduced postoperative edema tendinopathy257-267; additional research in this area is required.
but provided no improvement with regard to pain relief, grip
strength, or functional results compared with controls226. Tissue Engineering with Mesenchymal Stem Cells
Radiofrequency coblation is a new application of bipo- Mesenchymal stem cells are capable of undergoing differentia-
lar radiofrequency energy used for volumetric tissue removal. tion into a variety of specialized mesenchymal tissues,
Under appropriate conditions, a small vapor layer forms on including bone, tendon, cartilage, muscle, ligament, fat,
the active electrode of the device. The electrical field of the and marrow stroma (Fig. 3)268. In adults, mesenchymal stem
energized electrode causes electrical breakdown of the vapor, cells are preva- lent in bone marrow, but they are also found
producing a highly reactive plasma that is able to break down in muscle, fat, and skin and around blood vessels269. The
most of the bonds found in soft-tissue molecules. Radiofre- differentiation of mesenchymal stem cells along a particular
quency coblation stimulates an angiogenic response in normal phenotypic pathway may be controlled by a master regulatory
rabbit Achilles tendons227. Rapid pain relief was reported in a gene, a concept formulated after the discovery of MyoD, a
preliminary uncontrolled prospective, nonrandomized, single- muscle transcription factor capable of inducing expression
center, single-surgeon study of twenty patients with tendinop- of a bank of muscle-specific genes270. However, MyoD may
athy of the Achilles tendon, patellar tendon, and common not be the only transcription factor responsible for myogenic
extensor origin227. Six months after the procedure, magnetic differentiation; Myf5, myogenin, and MRF4 may also play
resonance imaging showed complete or near complete resolu- a role271. Transcription factors that regulate adipogenic and
tion of the tendinopathy in ten of the twenty patients. osteogenic differentiation have also been identified, but no
transcription factors regulating tenocyte differentiation have yet
Cytokines and Growth Factors been identified272-275.
Increased levels of TGF-ß2 have been reported in tendino- Mesenchymal stem cells can be applied directly to the site of
pathic human Achilles tendons and in rabbit flexor tendons after injury or can be delivered on a suitable carrier matrix, which
injury228,229. TGF-ß results in scar formation and fibrosis, and functions as a scaffold while tissue repair takes place. In ex
TGF-ß1 expression is increased in patients with hypertrophic vivo, de novo tissue engineering with use of mesenchymal
scarring and keloids following a burn230,231. The response to stem cells, whole body tissues are constructed in the labora-
cytokines may be site-specific, and insulin-like growth factor-I tory and are subsequently implanted into patients. Tissue-
(IGF-I) induces a higher rate of collagen synthesis in rabbit engineered tendons could be used to bridge areas of tendon
flexor tendons than it does in rabbit Achilles tendons232. The loss or to replace severely degenerated regions276-279.
use of cytokines and growth factors to enhance tendon healing At present, tissue engineering is an emerging field, and many
remains largely experimental and has been restricted to in vitro issues, such as ideal scaffold materials, optimal cellseeding
studies and animal models233-249. The clinical use of growth density, and optimal culture conditions, need to be es-
factors for the treatment of tendon problems has not yet been tablished before it becomes a real option in the management
reported, to our knowledge. of tendon disorders. Effective vascularization and innervation
of implanted tissue-engineered constructs must take place for
Gene Therapy the constructs to be viable. Vascularization allows survival of
Gene therapy delivers genetic material (DNA) to cells, permit- the construct. Innervation is required for proprioception and
ting modification of cellular function, by means of viral or non- to maintain reflexes, mediated by Golgi tendon organs, to pro-
viral vectors or direct gene transfer250,251. Gene therapy enables tect tendons from excessive forces280,281.
the delivery of individual proteins to specific tissues and cells252.
Several animal studies have been done to investigate Prevention of Adhesions
the feasibility of gene transfer to tendons. For example, The most important factor implicated in adhesion formation
hemaggluti- nating virus of Japan (HVJ)-liposome constructs is trauma282. Tenocytes and tenoblasts are key cells in tendon
were used to deliver ß-galactosidase to rat patellar tendons253. healing. The actin isoform α-smooth muscle actin has been
In vivo and ex vivo gene transfer techniques have been used as identified in tendons and ligaments283,284.Tenocytes that express

18
18
smooth muscle actin are known as myofibroblasts. There is unclear whether hyaluronate has any effect on myofibroblast
are three essential morphological elements that define function or just acts as a mechanical barrier. Results may vary
myofibroblasts: stress fibers (actin microfilaments), well- with different doses of hyaluronate.
developed cell-stroma attachment sites (fibronexus), and 5-fluorouracil, an antimetabolite with anti-inflammatory
intercellular gap junctions285. The fibronexus is presumed to properties, inhibits fibroblast proliferation, with a greater effect
transfer tensile forces to the extracellular matrix network286. on synovial fibroblasts than on endotenon fibroblasts296,297.
Myofibroblasts are thought to play a role in extracellular matrix Lacerated chicken flexor tendons were repaired and were
network homeostasis in tendons and ligaments, and they may exposed to various doses of 5-fluorouracil for five minutes298.
well be responsible for the formation of tendon adhesions287. A dose of 25 mg/mL effectively preserved tendon gliding,
Many attempts have been made to reduce adhesion for- and, at three weeks after the surgery, there was no significant
mation by using materials acting as mechanical barriers such difference in excursion, maximal load, or work of flexion
as polyethylene or silicone or by using pharmacological agents between the repaired tendons and normal controls. Use of 50
such as indomethacin and ibuprofen, but no simple method is mg/mL of 5-fluorouracil produced inferior results, suggesting
widely used288-291. Hyaluronate is found in synovial fluid around that there is a therapeutic threshold beyond which 5-fluorouracil
tendon sheaths292. Its use decreased adhesion formation in may be detrimental to tendon healing.
repaired rabbit flexor tendons293,294 but resulted in no signifi- Despite many efforts, adhesion formation after tendon trauma
cant differences in adhesion formation in a rat Achilles tendon remains a clinical problem, with no ideal method of prevention.
model295. The absence of a synovial membrane around the With advances in the understanding of the mechanisms
Achilles tendon may explain this difference. A single dose of involved in adhesion formation, it may be possible to formulate
hyaluronate, at a concentration of 10 mL/mg, had no effect on improved strategies of prevention.
rabbit tenocyte proliferation or matrix synthesis296. Therefore, it

Fig. 3
Schematic representation of mesenchymal stem cell differentiation.

Mobilization and Mechanical Loading from stress-shielded rabbit patellar tendons displayed lower
In animal experiments, training has improved the tensile tensile strength and strain at failure than did control samples303.
strength, elastic stiffness, weight, and cross-sectional area of Immobilization reduces the water and proteoglycan content
tendons299,300. These effects can be explained by an increase of tendons and increases the number of reducible collagen
in collagen and extracellular matrix network synthesis by teno- crosslinks304,305. Immobilization results in tendon atrophy,
cytes300. There are little data on the effect of exercise on human but, as a result of the low metabolic rate and vascularity,
tendons, although intensively trained athletes are reported to these changes occur slowly301.
have thicker Achilles tendons than control subjects301. Most of After the inflammatory phase of healing, controlled stretching
our current knowledge is therefore based on the results of ani- is likely to increase collagen synthesis and improve fiber
mal studies. However, care must be taken when interpreting alignment, resulting in higher tensile strength306. Collagen that
animal studies, as the results in untrained animals cannot be remains unstressed during the proliferative and remodeling
directly compared with those in trained animals. Also, con- phases remains haphazard in organization and is weaker than
fined animals are likely to have reduced connective-tissue stressed collagen307. Experimental studies have demonstrated
mass and tendon tensile strength, and physical training may the beneficial effects of motion and mechanical loading
merely return these parameters to normal302. on tenocyte function. Repetitive motion increases DNA
Prolonged immobilization following musculoskeletal content and protein synthesis in human tenocytes308.
injury may have detrimental effects. Collagen fascicles Even fifteen minutes of cyclic biaxial mechanical strain applied

19
19
to human tenocytes results in cellular proliferation309. The optimization strategies discussed in this article are cur-
Application of a cyclic load to wounded avian flexor tendons rently at an early stage of development. While these emerging
results in migration of epitenon cells into the wound310. In technologies may develop into clinical treatment options, their
rabbit patellar tendons, application of a 4% strain provides full impact on tendon healing needs to be critically eval- uated
protection against degradation by bacterial collagenase311. in a scientific fashion.
Clinical studies have shown the benefit of early mobili-
zation following tendon repair, and several postoperative
mo- bilization protocols have been advocated312-316. The Pankaj Sharma, MRCS
precise mechanism by which cells respond to load remains Nicola Maffulli, MD, MS, PhD, FRCS(Orth)
Department of Trauma and Orthopaedics, Keele University School
to be elucidated. However, cells must respond to mechanical of Medicine, Thornburrow Drive, Hartshill, Stoke-on-Trent, Stafford- shire,
and chemical signals in a coordinated fashion. For example, ST4 7QB, United Kingdom. E-mail address for N. Maffulli: n.maffulli@keele.
intercellular communication by means of gap junctions is ac.uk
necessary to mount mitogenic and matrigenic responses in ex
vivo models317. The authors did not receive grants or outside funding in support of their
research or preparation of this manuscript. They did not receive pay- ments
or other benefits or a commitment or agreement to provide such benefits
Overview from a commercial entity. No commercial entity paid or directed, or agreed
Tendon injuries produce substantial morbidity, and at present to pay or direct, any benefits to any research fund, foundation, educational
there are only a limited number of scientifically proven man- institution, or other charitable or nonprofit organization with which the
agement modalities. A better understanding of tendon func- authors are affiliated or associated.
tion and healing will allow specific treatment strategies to be
developed. Many interesting techniques are being pioneered. doi:10.2106/JBJS.D.01850

20
20
21
21
22
S
Studii clinice

23
23
23
LYNEN 2012
Tratamentul tendinopatiilor cronice cu injectii cu hialuronat peritendon sub control
imagistic - studiu interventional, multicentric, prospectiv, cu un singur brat.

REZUMAT :

ABSTRACT:
Tendinopatiile cronice reprezinta o problema uzuala, pentru care metodele actuale de
tratament sunt adesea nesatisfacatoare. In studiul de fata a fost evaluata eficacitatea
clinica si siguranta a doua injectii peritendinoase cu solutie 2% de Acid Hialuronic ad-
ministrat la un interval de o saptamana (OSTENIL® tendon seringa preumpluta, TRB
CHEMEDICA AG, Germania Haar) la 35 de pacienti care sufereau de tendinopatia dure-
roasa persistenta timp de cel putin sase saptamani a tendonului Ahilean, a tendonului
extensor comun de la nivelul articulatiei mainii sau a tendoanelor peroniere. Rezultatele
au aratat ca prin tratament se obtine o reducere semnificativa a durerii si o imbunatatire
semnificativa a simptomelor legate de boala. Tolerabilitatea si siguranta produsului ex-
celente au fost in mod clar demonstrate in acest studiu.

CUVINTE CHEIE:
Tendinopatia, tendinoze, Acid Hialuronic, hialuronat, tendonul lui Ahile, tendon peronier,
epicondilita, cotul tenismenului;

24
24
25
25
WISSENSCHAFT / RESEARCH Original Article / Originalarbeit

N. Lynen1

Treatment of chronic tendinopathies


with peritendinous hyaluronan
injections under sonographic
guidance – an interventional,
prospective,single-arm, multicenter
study
Behandlung von chronischen Tendinosen durch ultraschallgesteuerte
peritendinöse Hyaluronsäureinjektion - Eine interventionelle,
prospektive, einarmige multizentrische Studie

Abstract: Chronic tendinopathies are a common problem, for which current treatment methods are often unsatisfactory. In the
present study, the clinical efficacy and safety of two peritendinous injections of a 2% solution of hyaluronic acid administered at
an interval of one week (OSTENIL® TENDON pre-filled syringe , TRB Chemedica AG, Haar Germany) was assessed in 35 patients
who suffered from painful tendinopathy of the Achilles tendon, the common wrist extensor tendon or the peroneal tendons persis-
tent for at least six weeks. The results showed that treatment achieves a significant pain reduction and a significant improvement in
disease-related symptoms. Excellent tolerability and safety of the product were clearly demonstrated in this study.

Keywords: Tendinopathy, tendinosis, hyaluronic acid, hyaluronan, Achilles tendon, peroneal tendon, epicondylitis, tennis elbow.

Introduction
Overuse injuries of tendons are a common cause of painful symptoms in the extremities, particularly in young and active
individuals. This is often due to increased physical stress from sports activities [1]. But tendinopathy can also occur as a
result of physical work or improper loading. Pathological changes in tendons typically have a protracted course lasting
6 months or longer [2], with a smooth transition from a healthy tendon, via acute inflammatory achillodynia to chronic
tendinopathy. Tendon pain that persists for a long period is particularly difficult to treat successfully.
Although a wide variety of conservative treatment methods are available, few have lasting success, especially for patients with
a long history of symptoms [3]. The most likely reason is that there is an aseptic inflammatory response only at the onset of the
disease process. As shown in recent studies, the role of the inflammatory component becomes relatively minor already at an
earlier stage of the disease, so that the anti-inflammatory characteristics of corticosteroids or nonsteroidal anti-inflammatory drugs
(NSAIDs) do not provide effective treatment at this stage [4, 5]. In the further course of the disease, fibrosis develops with increased
formation of collagen between tissue layers, which leads in turn to adhesion of the tendons and a painful reduction in the ability
of the tendon to glide freely. The adhesions also result in reduced nutrition of the bradytrophic tendons, since synovial fluid is no
longer distributed in sufficient quantities. This leads to metabolic disorders, with changes in fibre structure and dedifferentiation of
type I collagen to type III collagen as well as sprouting of nociceptive nerve endings.
Several studies have shown that peritendinous application of hyaluronic
acid (HA) is an effective therapeutic option for the treatment of chronic tendinopathy [6, 7, 8]. HA is the main component of synovial
fluid, which is found in the normal tendon sheath and surrounding tissues; synovial fluid contributes to the nutrition of the tendon [9].
The viscoelastic properties of HA also have significant therapeutic effects, since HA can help to reduce the surface
friction of tendons whose capacity to glide freely has been reduced by microtears and adhesions. Recent studies provide an
explanation of the physical mechanism by which surface friction of tendons is reduced: the application of HA together with the
formation of a network of the cells on the tendon surface result in the “gliding effect“ [10,11,12].
At the cellular level, a further effect is produced by expansion of the extracellular space. Because of its high anionic charge,
HA is able to bind a large quantity of water molecules, and the HA-water aggregate may swell to occupy as much as 10,000
times the volume of the HA molecule alone [13]. Due to the increased osmotic pressure, spaces form in the extracellular matrix

26
26
Figure 1
Box plot analysis, visual analogue scale

and between the cells, contributing to an increase in cell mobility and thus leading to increased cell migration [14]. Furthermore,
the tightly interlocked HA macromolecules act like a molecular sieve, which prevents free passage of inflammatory cells,
prostaglandins and cytokines, and at the same time channels the transport of nutritionally important metabolites from the
synovial cells to the tendon [9, 15].
In addition, HA provides an ideal environment for cell proliferation and differentiation due to its high affinity for the
extracellular matrix [16, 17]. The improved environmental conditions increase cellular activity, so that the cells produce increased
quantities of extracellular matrix, which in turn results in optimisation of the repair process [15]. HA also has an analgesic effect,
since it has a desensitising action on nociceptive receptors [18].
Due to the favourable characteristics of HA and the lack of satisfactory therapeutic alternatives, the present study was
designed to evaluate the efficacy and safety in patients with chronic tendinopathy of two peritendinous injections of HA (2
ml of 2% HA + 0.5% mannitol) at an interval of one week.

Patients and Methods


The study was an interventional, prospective, single-arm, multicenter trial.
The trial was approved by an independent ethics committee, and each patient gave written consent to participation before inclu-
sion in the trial.
A total of 35 patients aged between 18 and 75 years who suffered for at least 6 weeks from chronic tendinopathy in
the mid-portion of the Achilles tendon, of the peroneal tendons or enthesitis of the lateral epicondyle of the humerus were
included in the study. All patients had severe pain symptoms with a score of at least 40 mm on the visual analogue scale (Pain
VAS) according to Huskisson [19].
Patients were excluded from participation if they had a concomitant disease that might interfere with the parameters to be as-
sessed, if they had a severe systemic disease or if the trial product was contraindicated.
Patients were also excluded if they had been treated with systemic or topical steroids less than 4 weeks prior to the first examina-
tion, or with NSAIDs within the week prior to the start of the trial. Patients were not permitted to use any additional medications
(NSAIDs in particular) or physotherapy for the entire duration of the trial. Extreme sports as well as heavy physical activities that
could have had a negative impact on the symptoms were also prohibited.
After checking the inclusion and and exclusion criteria, each patient received a peritendinous injection (with ultrasound guidance)

27
27
Figure 2
Visual analog scale by tendon

of 2 ml of the trial product (40 mg HA + 10 mg of mannitol) CGI, patient questionnaire for all time time points, and the
around the affected tendon. A second injection was made VAS score at 1 and 12 weeks after the start of treatment.
after an interval of one week. During the treatment phase, An intra-group comparison for the primary endpoint
trial parameters were recorded before the first injection and was performed with the 2-sided Wilcoxon-Pratt test. The
then before the second injection. Further measurements alpha level was defined as alpha = 0.05. All secondary
were made at 5 and 12 weeks after the start of treatment. parameters were analysed with the 2-sided sign test, alpha
The time course of pain symptoms = 0.05, for intra-group comparisons between baseline and
was assessed with the Pain VAS (100 mm), in which 0 follow-up parameters.
represents ‘no pain’ and 100 represents ‘extreme pain’
[19]. The clinical parameters typical for tendinopathy
(redness, warmth, swelling, tenderness, crepitus during
movement and peritendinous effusion) were recorded
Results
at all time points. A 5-point ordinal scale was used to In the period from 3 February to 30 May 2011, a total of 35
assess intensity (0 = none, 1 = slight, 2 = moderate, 3 = patients with a mean age of 45.9 ± 10.6 years (± SD) were
substantial, 4 = extreme) of each clinical parameter. included in the study. Seventeen (51.4 %) of the patients
The overall impression of treatment success (clinical were female and 18 (48.6 %) were male.
global impression, CGI) was measured using a 7-point Tendinopathy was localised in the mid-portion of the
ordinal scale (1 = very much improved, 2 = much improved, Achilles tendon in 19 patients (54.3%), at the lateral
3 = slightly improved, 4 = unchanged, 5 = slightly worse, epicondyle of the humerus in 14 patients (40.0%) and in
6 = much worse, 7 = very much worse). This assessment the peroneal tendon in 2 patients (5.7%). Patients had
was made by both the investigator and the patient at 1, 5 suffered from chronic tendinopathy for a median period of
and 12 weeks after the start of treatment. 12.3 ± 17.9 months.
A patient questionnaire was used to assess the patient’s Assessment of pain symptoms with the VAS showed a
perception of the extent to which his daily life, leisure significant improvement compared to baseline
activities and work were restricted. This assessment was (p <0.0001) at 5 weeks after the start of treatment. At
made using a 5-point ordinal scale (0 = no restriction, 1 = the time of enrolment, patients rated pain with a median
slight restriction, 2 = moderate restriction, 3 = substantial intensity of 62 ± 18.9 mm, but at 5 weeks the score was
restriction, 4 = extreme restriction) at all time points. Safety only 21 ± 21.3 mm (Fig. 1).
aspects were evaluated by documenting all adverse The significant decrease in pain intensity was maintained
events. in the long term, since further significant improvement of
the VAS score was found at 12 weeks, with a median of 17
Statistical analysis ± 26.2 mm (p <0.0001).
Specific analysis by tendon showed a significant
Since this was a pilot study, there was no control group improvement of pain in all affected tendons (Fig. 2). The
and a statistical calculation of sample size was not most pronounced improvement was found in patients
performed. with Achilles tendinopathy. These patients reported pain
The number of participants was chosen to guarantee intensity before the first injection with a median VAS score
a reliable analysis. For assessment and analysis of the of 57 ± 19.1 mm, but during treatment this improved to a
safety and effectiveness of the trial product, data from all median of 19 ± 19.9 mm at 5 weeks and 13 ± 20.1 mm at
35 patients were analysed. Missing data were replaced 12 weeks after the start of treatment.
according to the ‘Last Observation Carried Forward’ Patients with tendinopathy localised to the lateral
(LOCF) principle. epicondyle of the humerus reported a median VAS score
The Pain VAS value at 5 weeks after the start of of 65 ± 19.9 mm at the initial assessment, which improved
treatment was used as the primary measure of efficacy. to 29 ± 21.0 mm and 29 ± 31.1 mm at the first and second
Secondary parameters included clinical parameters, the follow-up assessments.

28
28
Patients with affected peroneal tendons showed an [24]. It should also be noted that all patients enrolled in
improvement in the median VAS score from an initial value the trial had suffered from painful tendinopathy for at least
of 65 ± 19.8 mm to 26 ± 31.8 mm at 12 weeks after the 12 months, and their pain improved in a short time with
first treatment. HA treatment.
With regard to clinical parameters, complete resolution Similarly, there was also a significant improvement
of peritendinous fluid accumulation was found in all in the clinical findings associated with tendinopathy.
patients (100%) at 12 weeks after the start of treatment. In particular, there were significant improvements in
Almost all patients showed complete improvement of non-structural altera tions, although tissue swelling,
redness (96.9%), warmth (96.9%) and crepitation during an indication of structural reorganisation of the
movement (93.8%). Almost 2/3 of the patients (60%) tendon, was still present in two thirds of the patients
experienced a reduction in tenderness, while 66% of at the final examination at 12 weeks after the start of
patients had less swelling at 12 weeks after the start of treatment. Despite the swelling of the tendon and/or the
treatment. peritendinous tissues, there were no lasting symptoms of
The CGI was very well correlated for both the clinical inflammation or irritation. This may be explained by
investigators and the patients. The investigators reported a reduction in friction and peritendinous adhesions due to
clinical global improvement at in 88.6% of patients at one the HA injections [10, 11, 12].
week, at 5 weeks in 91.4% of patients and at 12 weeks in With regard to the long-term effects of the HA
93.8% of patients. treatment, it was found that even at the time of the follow-
The patients reported a clinical global improvement at one up at 12 weeks after the start of treatment, there was still
week in 82.9% of cases, at 5 weeks in 88.6% of cases and a lasting effect with clear improvement of the previous
at 12 weeks in 87.5% of cases. symptoms. This is probably due to the high concentration
One week after the first injection, many patients (45.7%) of HA that was used and stabilisation of the HA with the
reported a significant improvement, and five weeks after excipient mannitol. The high concentration of HA probably
the start of treatment, a large number (40.0%) reported a contributed to the excellent results achieved in the present
slight improvement compared to the initial findings. study. The favourable outcome of the treatment regimen
At the check-up at 12 weeks after the start of treatment, was sustained for a relatively long follow-up period,
the majority of patients reported that their symptoms were although it consisted of only two HA injections at an
‘much improved’ or ‘very muchimproved’ (31.3% each). interval of one week.
This positive assessment was also reflected in the
evaluation of the patient questionnaire. At all time points,
there was a significant improvement in the restriction of
daily activities (p <0.0001), leisure activities (p <0.0001)
and at work (p <0.0018). Summary and Clinical
A total of 14 adverse events were reported by 11
(31.4%) patients. None of these events were associated
Significance
with the trial product. The peritendinous injections of HA
were well tolerated by all patients. The results of this study indicate that two injections
of a 2% HA solution administered at an interval of one
week represent a very effective therapeutic option for
treatment of chronic tendinopathy. Treatment with
Discussion 2% hyaluronic acid resulted in a rapid and sustained
reduction in pain as well as a significant improvement
of clinical symptoms. Excellent tolerability and safety
Treatment of chronic tendinopathies often represents of the product were clearly demonstrated in this study.
a significant problem for the physician. Injections of None of the adverse events was associated with
corticosteroids, which are commonly used, have a the use of the trial product. Further clinical studies
particularly limited efficacy in chronic tendinopathies, with larger patient groups and a longer period of
and may have significant side effects. For example, assessment should be conducted in the near future to
symptoms often recur shortly after treatment with lipid- confirm the present findings.
soluble corticosteroids [20]. In particular, frequent use of
corticosteroids may be accompanied by peritendinous
crystalline deposits that can make the tendon brittle and
prone to rupture [21]. Regular administration of NSAIDs, Conflict of interest. Dr. Lynen acted as a medical expert
which are commonly used to treat tendinopathy, may have for this clinical trial.
gastrointestinal side effects among others.
HA is a naturally occurring biological substance, which
is a major component of ligaments, cartilage and synovial
structures [22]. Furthermore, HA is very effective in the
treatment of osteoarthritis, and has few side effects [23].
In the present study, 2 ml of a 2% HA solution were used.
Based on the therapeutic regimen for HA treatment of
osteoarthritis, two injections were performed at an interval
of one week. The injections were administered around the
tendon under ultrasound guidance.
In the evaluation of treatment outcomes, it was found Correspondence:
that treatment of painful chronic tendinopathy with
peritendinous injection of HA significantly reduced pain Dr. Nils Lynen
symptoms. This is probably due in part to the tightly Practice Centre
interlocked HA macromolecules, which specifically Nordrhein Orthopaedics and
prevent free passage of inflammatory cells, prostaglandins Traumatology Friedrich-Wilhelm-Platz 5–6
and cytokines [15], and partly to the effect of the mannitol, 52062 Aachen
added to the formulation which traps oxygen free radicals n.lynen@exinmo.de

29
29
Injectii subacromiale cu hialuronat de sodiu pentru
gestionarea tendinopatiei bazale a coafei de
rotatori: studiu comparativ cu terapia de reabilitare

REZUMAT :

BACKGROUND:
Tendinopatia bazala de coafa de rotatori (CR) este o cauza obisnuita a durerii si a disfunctiei
umarului. Literatura sugereaza ca dovezi, ca o combinatie de utilizare excesiva si compresiune
extrinseca poate induce tendinopatia cronica de CR. Scopul studiului a fost de a compara rezul-
tatele actuale ale preparatelor injectabile subacromial pe baza de hialuronat de sodiu cu terapia
de reabilitare.

MATERIALE SI METODE:
Am inrolat 48 pacienti (M/F : 26/22 ; varsta medie: 50 ani; umarul drept / stang : 29/19), cu dureri
de umar persistente timp de cel putin 4 luni. Criteriile de excludere au fost dupa cum urmeaza:
ruptura de CR, tendinita calcifiata, instabilitate glenohumerala, osteoartrita, boli reumatice, terapie
fizica si / sau injectare in cele 4 luni anterioare, chirurgie de umar, bloc nervos anestezic, trauma-
tisme, boli severe. Subiectii inclusi au primit fie doua injectii sub ghidaj ecografic subacromial de
Acid Hialuronic (AH) (2% pacienti, grupa AH) la momentul initial si la 14 zile, sau au suferit terapie
de reabilitare (23 pacienti, grupul Physio), inclusiv mobilizarea activa a umarului, intinderea de
tesut moale, pozitionare capului humeral si consolidarea muschilor propulsori timp de 30 de zile (3
sesiuni in fiecare saptamana). Evaluarea clinica a functiei umarului a fost realizata cu scorul bazat
pe scala analogica vizuala pentru durere (VAS) (0-100), scorul Oxford pentru umar (OSS), si Scorul
Constant-Murley (CS). In general, pacientii au fost examinati la momentul initial, saptamana 2,
saptamana 4, saptamana 12 si saptamana 24. Semnificatia statistica fost stabilita la 5% (p \ 0,05).

REZULTATE:
Reducerea durerii globale in grupul AH a fost statistic semnificativa in saptamana a-2-a (p \ 0,05)
saptamana 4 (p \ 0,05), saptamana 12 fata de valoarea initiala. In mod similar, subscorurile de
durere (pe timpul noptii si cu activitate) au fost in mod semnificativ mai scazute in saptamana 2 (p
\ 0,05), saptamana 4 (p \ 0,05), si respective in saptamana 12 (p \ 0,05). In grupul Physio, durerea
a scazut in mod semnificativ in saptamana 2 (p \ 0,05), dar nu s-a mentinut la saptamana 4 (p \
0,05), saptamana 12 (p \ 0,05), si saptamana 24 (p \ 0,05). Scorul CS si OSS in grupul HA a crescut
in mod semnificativ in saptamana 2 (p \ 0,05), saptamana 4 (p \ 0,05), si saptamana 12 (p \ 0,05).
O crestere nesemnificativa statistic a scorurilor clinice a fost gasit in saptamana 24 (p \ 0,05). O
imbunatatire semnificativa a CS si OSS s-a inregistrat in grupul Physio in saptamana 2 (p \ 0,05),
dar nu si in saptamanile 4, 12 si 24 (p \ 0,05).

CONCLUZII:
Preparatele injectabile subacromial cu AH ar putea fi o alternativa de tratament eficienta si sigura
pentru pacientii care sufera de tendinopatii de CR. Noi credem ca rezultatele acestui studiu sunt
incurajatoare, dar nu pentru un tratament de durata si am putea presupune ca o serie de trei pana
la patru injectii subacromial cu hialuronat de sodiu ar putea oferi beneficii clinice pe termen la
mediu si lung.

CUVINTE CHEIE:
Umar, injectie, hialuronat de sodiu, rotatori, tendinopatia.

30
30
Musculoskelet Surg (2013) 97 (Suppl 1):S49–S56
DOI 10.1007/s12306-013-0259-y

Ultrasound-guided subacromial injections of sodium hyaluronate


for the management of rotator cuff tendinopathy: a prospective
comparative study with rehabilitation therapy
G. Merolla • P. Bianchi • G. Porcellini

Received: 20 December 2012 / Accepted: 3 March 2013 / Published online: 21 April 2013
© Istituto Ortopedico Rizzoli 2013

Abstract
Background: Rotator cuff (RC) tendinopathy is a common cause of pain and shoulder dysfunction. The literature evidence
suggests that a combination of overuse and extrinsic compression may induce chronic RC tendinopathy. Aim of the current
study was to compare the results of subacromial sodium hyaluronate injections with rehabilitation therapy. Materials and
methods. We enrolled 48 patients (M/F: 26/22; mean age: 50 years; shoulder right/left: 29/19) with persistent shoulder pain for at
least 4 months. Exclusion criteria were as follows: RC tear, calcifying tendinitis, glenohumeral instability, osteoarthritis, rheumatic
diseases, physical therapy and/or injection in the previous 4 months, shoulder surgery, anesthetic nerve block, trauma, and severe
medical diseases. The included subjects received either two ultrasound-guided subacromial hyaluronic acid (HA) injections (25
patients, HA group) at baseline and 14 days, or underwent rehabilitation therapy (23 patients, Physio group) including active
shoulder mobilization, soft tissue stretching and humeral head positioner and propeller muscles strengthening for 30 days (3
sessions every week). Clinical assessment of shoulder function was performed with visual analog scale score for pain (0–100),
Oxford Shoulder Score (OSS), and Constant–Murley Score (CS). Overall, patients were examined at baseline, week 2, week 4, week
12, and week 24. Statistical significance was set at 5 % (p \ 0.05).

Results Reduction in overall pain in the HA group was statistically significant at week 2 (p \ 0.05) week 4 (p \ 0.05), week 12
comparing to baseline. Similarly, pain sub- scores (at night and with activity) were significantly lower at week 2 (p \ 0.05), week 4 (p
\ 0.05), and week 12 (p \ 0.05), respectively. In the Physio group, pain decreased significantly at week 2 (p \ 0.05) but not maintained
at week 4 (p [ 0.05), week 12 (p [ 0.05), and week 24 (p [ 0.05). CS and OSS in the HA group increased significantly at week 2 (p \
0.05), week 4 (p \ 0.05), and week 12 (p \ 0.05). A non-statistically significant increase in clinical scores was found at week 24 (p
[ 0.05). A significant improvement of CS and OSS we found in the Physio group at week 2 (p \ 0.05), but not at weeks 4, 12, and
24 (p [ 0.05).

Conclusions: Subacromial HA injections could be an effective and safe alternative treatment for patients suffering from
RC tendinopathy. We believe that the results of this study are encouraging but not lasting and we might suppose that a
series of three to four subacromial sodium hyaluronate injections could provide good mid and long-term clinical benefits.

Keywords: Shoulder • Injection • Sodium hyaluronate •Rotator cuff • Tendinopathy

G. Merolla (&) • G. Porcellini


Unit of Shoulder and Elbow Surgery, D. Cervesi Hospital,
Via L. V. Beethowen 46, Cattolica, Italy e-mail: giovanni.merolla@auslrn.net; giovannimerolla@hotmail.com

P. Bianchi
Department of Orthopedics, Traumatology, Rehabilitation
and Plastic Surgery, Second University of Naples, Naples, Italy

Introduction
Rotator cuff (RC) tendinopathy is a common source of pain and shoulder dysfunction that affects both the young and older
population [1] mainly in arduous overhead workers [2]. The available research findings emphasize the multi-factorial origin of
RC tendinopathy that begins as a failure in the tendon fibers due to the overuse and cyclic loading on their internal side
[1]. Under these conditions, the subunits of the RC tendons undergo internal compressive forces which induce the development

31
31
of fibrocartilage in these regions [3]; particularly, involving a study population of 60 patients with RC
fibrocartilage develops on the articular side of the tendinopathy aged from 47 to 53 years old, seen in
supraspinatus tendon close to its insertion, where the the outpatient office of our Shoulder and Elbow Unit
articular fibers are subjected to less strain than the non- between June 2010 and March 2011 (Table 1). Patients
articular side [4, 5]. Fibrocartilage is less capable of were considered eligible for subacromial therapy if they
withstanding tension load, and joint-side fibers have half were 18 years or older, had persistent shoulder pain for
the force of the bursal-side fibers [6]. Furthermore, the at least 4 months, clinical diagnosis of RC tendinopathy
supraspinatus and infraspinatus are inserted into a cable detected with MRI, no previous treatment with articular or
located 1.5 cm from the humeral insertion of the fibers [7], subacromial steroid injections within the last 4 months,
and the ‘‘crescent’’ region is located between the cable availability for the duration of the study. Patients were
and the humerus insertion where the tendon fibers are excluded if they refused to consent to such a proce-
thinner and close to the hypovascularized region and dure, had a positive history of shoulder trauma,
therefore are more susceptible to degeneration [8]. This partial or complete RC tears, calcifying tendinitis,
evidence suggests that a combination of overuse previous arthroscopic or open shoulder surgery, shoulder
and extrinsic compression may induce chronic RC instability, infections or neoplasm, symptomatic cervical
tendinopathy. Conservative therapies, including spine disease, rheumatoid arthritis or immune diseases,
rehabilitation, physical therapies analgesics, and gout and uric acid diseases, severe medical conditions,
non-steroid anti-inflammatory drugs (NSAIDs), are or were pregnant. Patients were also assessed for
commonly recommended to restore shoulder function their mental status and excluded if they presented with
in chronic RC tendinopathy. Physiotherapy is based cognitive limitations that could prevent them expressing
on the correction of the flexibility, motion, and strength a valid consent, or undergo subjective and objective
that are the cause of painful shoulder [9]. Subacromial evaluations. We also excluded subjects with known
supplementation with sodium hyaluronate or hyaluronic allergic or adverse reactions to previous nonsteroid or
acid (HA) has been proposed to treat RC dysfunction hyaluronan injections. All evaluations were performed by
[10] due to its action as a lubricant [11] and two experienced shoulder surgeons. The diagnosis of RC
suppressor of the joint inflammatory process [12]. To tendinopathy was performed by trained musculoskeletal
our knowledge, no studies in the literature have compared radiologists who depicted a high tendon signal intensity
physiotherapy and subacromial HA injection; therefore, the that was anatomically intact on the MRI in T2-weighted
purpose of the current research was to assess the images. During the screening, 10 patients were excluded
efficacy and safety of sodium hyaluronate injection because they refused the treatment or failed to comply
versus physiotherapy in a population of patients with with the inclusion criteria. We allowed a rescue medication
painful chronic RC tendinopathy. of oral paracetamol at a maximum dosage of 4 g/day,
and the amount taken by each patient was recorded
Materials and methods at the four follow-up visits. Withdrawal of rescue
medication (paracetamol) 24 h prior to each follow- up
Study population visit was recommended by the assessor and confirmed
by the patients during the examination. There were no
The study was approved by the Hospital Institutional statistically significant differences between the two groups
review board (Prot. 4232/2011/I.5/186), and all patients of treatment with respect to demographic characteristics
gave informed consent prior to enrollment. This was at baseline (Table 1).
a prospective non-randomized comparative study

Table 1
Demographic and anthropometric data of the population enrolled in the study

32
32
Fig. 1 Anatomical landmark with the two sites of injections drawn Fig. 2 The preparation is injected under ultrasound assistance
for the anterolateral approach to the subacromial space taking care to direct the needle medially and posteriorly, on the
insertion area of the supraspinatus tendon

Treatment groups Physiotherapy


We recruited an intent-to-treat population to be assigned We set a standard protocol of physiotherapy [9] for 30
to two treatment groups with the objective of gaining days, following three sessions a week. The loss of flexibility
an improvement in the pain score of 4 points and in the due to the glenohumeral internal rotation deficit (GIRD)
Constant–Murley scale of 10 points. According to these and the coracoid base muscles tightness was treated
parameters and a power value of 0.8, we had enrolled 50 using open stretch in the supine position and rotation
patients consecutively: the first half in the hyaluronic stretch exercises with the scapula stabilized. Scapula
acid group ( HA group, N= 25) and the second half in control was obtained performing exercises for lower
physiotherapy group (Physio group, N = 25). trapezius and serratus anterior with the arm below
90° abduction. RC activation exercises were allowed after
a stable scapular base was established and included
horizontal and vertical closed-chain exercises,
horizontal open-chain exercises, and diagonal closed-
chain exercises. The final sequence was completed with
open-chain plyometric exercises.

Ultrasound-guided injection Outcome measurements


The study material was a specific preparation (SportVisTM, Primary outcome measurements were the visual
MDT Int’l SA, Switzerland), whose main content is a analog scale (VAS) for pain.
STABHATM (Soft Tissue Adapted Biocompatible Hyalu- Secondary outcomes measurements were the
ronic Acid) of 1 million of daltons contained in a pre-filled Constant–Murley scale (CS) [13] and the Oxford
syringe (12 mg/1.2 ml). Shoulder Score OSS) [14, 15], the Patient Global
The procedure was performed in a standardized way Assessment (PGA), the tolerability and any adverse
in the outpatients’ office with the patient in the upright events after each injection. Pain included overall pain,
position using local disinfection, sterile drape, and marking pain at night, pain with activity, and pain without activity
the site of injection (Fig. 1). A shoulder surgeon (MG) with and was rated subjectively on a scale ranging from 1 (no
10 years of experience injected all the shoulders using pain) to 10 (severe pain). Patients were invited to write their
an ultrasound-guided anterolateral approach and the average pain rate for the previous 48 h. The CS included
same GE Logiq 7 ultrasound machine (7.5–14 MHz). a subjective ques- tionnaire for pain, the ability to perform
We first checked the tendon thickness and its superior daily living activity (DLA), an objective evaluation of active
limitant. Subsequently, a 22G needle was introduced range of motion (ROM) and strength. Pain was scored on a
beneath the anterolateral acromial edge above the tendon 15-point scale (0 severe pain, 15 no pain), while DLA was
(Fig. 2), taking care not to overcome the superior limitant. scored on a 20-point scale, with lower scores associated
The preparation was injected allowing it to spread over the with greater impairment on DLA. ROM was measured
superior tendon surface (Fig. 3). using a standard goniometer between the upper arm
and the upper part of the thorax. Shoulder strength was
assessed using the Lafayette handheld dynamometer
(Lafayette Instruments, Lafayette, Ind, USA) that has a
microprocessor with a resolution of 0.4 lb (0.2 kg) in the
range 0–50 pounds

33
33
(0–22.6 kg), 0.03 % accuracy with two calibration points: injection treatment with hyaluronic acid (HA group)
0.25 and 50 lbs (0.11 and 22.6 kg). Data were recorded from June to July 2010 completed the follow-up
and analyzed using SPSS version 10 software (SPSS Inc, evaluations (Table 1).
Chicago, IL, USA). We assigned 1 point for each 0.5 kg of Among the 25 patients who were assigned to the
strength registered. physiotherapy group (Physio group) from August to
The OSS is a 12-item patient-reported outcome October 2010, two cases were excluded: one case
measure specifically used for assessing the impact of because he refused to consent to the use of his data
RC tendin- opathy on the patient’s quality of life. Each for research pur- poses at the time of the enrollment
question on the OSS is scored 0–4, with 4 representing and one case due to incomplete evaluation (lost at last
the best; the twelve items are summed to produce overall follow-up examination) (Table 1).
scores ranging from 0 to 48. Interpreting the OSS: a score
0–19 indicates a severe shoulder condition, a score 20–29 Pain score and subscores
indicates a moderate to severe shoulder condition, a score
30–39 indicates a mild to moderate shoulder condition, a In the HA group, we registered a remarkable decrease
score 40–48 may indicate a satisfactory shoulder function. in overall pain score from baseline to the week 2 (p \
0.05), week 4 (p \ 005), and week 12 (p \ 0.05), and it
Follow-up was always statistically significant. The pain reduction
was recorded at week 24, but no statistically significant
Overall, patients were assessed immediately before dif- ference was found (p [ 0.05) (Table 2). Similarly, pain
the treatment (baseline), at the time of the second subscores (at night and with activity) were significantly
injection (week 2), and subsequently at week 4, week lower compared with baseline values, respectively, at
12, and week 24. The value of the subjective pain score week 2 (p \ 0.05), week 4 (p \ 0.05), and week 12 (p \ 0.05),
was recorded by each patient using a printed analogic while at week 24, such reduction was maintained but not
scale. Data were then entered on an electronic worksheet statistically significant (p [ 0.05) (Table 3).
(Microsoft Excel for Mac OS 2011) which calculated the In the Physio group at follow-up evaluation, we found
average values for pain overall and relative subscores. that pain decreased significantly at week 2 (p \ 0.05) while
CS and OSS were obtained as the numeric sum of the
subjective and objective evaluations using a dedicated
electronic version of both scales (www.orthopaedicscores.
com) that gave the final score at the end of the
questionnaire. Overall clinical scores were collected by
two independent observers who had not per- formed the
injections.

Statistical analysis
Statistical analysis was performed using the Kruskal–
Wallis test for the equality of populations and the ANOVA
test for the variables, setting the significance at 5 %.
Bravais–Pearson correlation coefficient was calculated to
analyze the variability of the two independent observations.

Results
Fig. 3 Ultrasound image showing the needle (red arow) placed in the
All the 25 patients with persistent shoulder pain due subacromial bursa, on the superior limitant of the supraspinatus ten-
don. The blue arrows emphasize the preparation that expanded above
to RC tendinopathy considered for subacromial
the tendon surface (color figure online)

34
34
higher values were registered at week 4 (p [ 0.05), week by all the patients enrolled for the study.
12 (p [ 0.05), and week 24 (p [ 0.05) (Table 2). Pain at
night and pain with activity had similar trend with a sig-
nificant decreasing at week 2 (p \ 0.05) and persistent Discussion
higher values at week 4 (p [ 0.05), week 12 (p [ 0.05), and
week 24 (p [ 0.05) (Table 3). Constant–Murley Score and Sodium hyaluronate is effective in managing acute
Oxford Shoulder Score The average values of CS and OSS or chronic ligaments and tendon injuries, like ankle
in the HA group increased significantly comparing to sprains and epicondylalgia [16, 17]. HA is believed
baseline at week 2 non-statistically significant increase to integrate into the extracellular fibrin matrix to
in clinical scores was found at week 24 (p [ 0.05). In the help realignment of fibrils thanks to electrostatic
Physio group, we significant difference compared with interactions. Thus, stability of form and function is
baseline values (p [ 0.05) (Tables 4, 5). then restored allowing healing to occur in structures
(ligaments and tendons) and shortening the rehabilitation
Comparison between HA and Physio groups process. Recent research findings demonstrated that
Overall pain and subscores repeated periarticular injections of the study materials
were more effective in pain relief and joint function
Overall pain and subscores were stratified to evaluate the improvement, compared to placebo or standard
difference in pain scores between the two groups of treat- conservative treatment for ankle sprains and
ment. There was no statistically significant difference lateral epicondylalgia. Long-term follow-up by
in overall pain scores between the groups at baseline investigators confirmed the therapeutic effects
and at week 2 (p \ 0.05). We found an average difference persist after 12 and 24 months [16, 17].
of 2.22 points at week 4 (p = 0.0149) and 2.16 points at Several trials using sodium hyaluronate in the treatment
week 12 (p = 0.0168), while the difference at week 24 was for chronic shoulder pain have been documented, on
not statistically significant (p \ 0.05) (Table 2). Subscores shoulder osteoarthritis [18], RC tears [19], peri-arthritis
for pain at night were significantly different at week 4 [20, 21] adhesive capsulitis [22], and chronic shoulder
(p = 0.0487), and remained at week 12, while the average pain of different etiologies [23]. The effectiveness of
difference found at week 24 was not significant (p \ 0.05). subacromial injections of HA alone in patients with
A similar trend was found for pain with activity which chronic RC tendinopathy is also reported in the literature.
registered a significant difference at week 4(p = 0.0318) In an open label multicenter study, Itokazu et al. [21]
and maintained at week 12 and but not at week observed a significant pain and range of motion they
24(p \ 0.05). Rescue medication consumption was found conclude that this treatment was effective in patients
to be similar at week 2 and 4 in both groups, and higher with periarthritis of the shoulder. Kim et al. [24],
from the week 12 to the week 24 in the Physio group. in a prospective randomized single-blind comparative
study on reduction and similar functional improvement
comparing to corticosteroid at 12 weeks. On the
Clinical scores contrary, respect to our results, Penning et al. [25]
reported a significant reduction in pain at short- and
(p \ 0.05), and maintained at week 24 follow-up but long-term follow-up with corticosteroid injections
without statistical significance (p \ 0.05). The OSS in compared with hyaluronic acid and in the long-
the two groups had the same values at baseline, but term placebo injections showed the best results.
significantly higher scorers registered in the HA group In the current study, physiotherapy was selected for
at weeks 4 and 12 (p \ 0.05) and such improvement the comparator because it is proven to be effective in
difference maintained at week 24 but with no shoulder tendinopathy management [26] and is recom-
significant difference (p \ 0.05). Interobserver agreement mended by the recent European guidelines (2008).
resulted in k values ranging from 0.81 to 1.85 for CS and A standard program of physiotherapy is recommended to
from 0.80 to 0.88 for OSS; good intraobserver agreement correct inflexibility, loss of strength and mobility in RC,
was registered with k = 0.85–0.90. and associated structures, but it is common that these
conditions persist despite a proper rehabilitation program
which is phased into a maintenance program to reduce
Patient global assessment and adverse events the risk of reinjury [9]. The current study compared directly
the effects of HA injections and physiotherapy, and we
The PGA showed good patient compliance with found that both treatments produced good results in pain
no serious adverse events registered during the and clinical scores in the short term, with a significant pain
experimentation. The anterolateral approach injections, decrease at week 2 of 3.3 points in the HA group and 2.8
with US assistance, proved to be safe and well tolerated

35
35
and clinical scores in the short term, with a significant pain decrease at week 2 of 3.3 points in the HA group and 2.8 points in the
Physio group. In midterm, the pain score maintained a significant improvement at week 4 and week 12 only in the HA group, while
the improvement found in the Physio group was not significant at week 4 and week 12 compared to the baseline. Pain subscores
at night and during activity in the HA group showed a significant reduction from the start of the study throughout follow-up, except
for week 24, while in the Physio group, the reduction in pain subscores was not significant from week 2 to week 24. CS and OSS
showed a course similar to overall pain, with a significant improvement in both treatment groups at week 2, persistent pain relief in
the HA group only at week 4 and week 12, and a return to lower but non-statistically significant scores with respect to baseline, at
week 24 in both groups. The consumption of rescue medication was higher in the Physio group from week 12 to week 24, but at the
end of the study, such difference became not significant. These results suggest that subacromial HA injections are effective in in-
ducing better clinical outcomes and pain relief compared to physiotherapy from the start of the study up to week 12, the therapeutic
effects became less important at 6 months when symptoms gradually returned. Stratifying the results, we found that five patients in
the Physio group had worsening VAS scores at the end of study, whereas this was seen in only two patients in the HA group. The
two patients in the HA group were young in age and have relative high daily activities on the shoulder in life, that is, regular fitness
training and therefore higher risk to develop overuse tendinopathy and subacromial bursitis. The procedure for shoulder injection
was safe without any adverse events registered in all the enrolled subjects.
We used the same anterolateral approach with ultra-sound assistance, to allow the injection on the superior limitant of the
supraspinatus tendon as accurate as possible [27]. Using this approach, we ensured that the HA preparation was injected on the
thinner crescent supraspinatus fibers, [7] located within the hypovascular region (critical zone) [8] and more at risk of degeneration
and tearing.
Our research findings confirmed the good early and midterm results of HA in RC tendinopathy and are consistent with the
outcomes of Itokazu et al. [23] and with those reported by Kim et al. [24] who described a good VAS and ASES scores in patients
treated with HA injections by using corticosteroids as a comparator.
The choice of two injections 2 week apart could explain the lower clinical scores observed at 6 months, and it might be reason-
able to expect that the long-term results may be more favorable if the number of injections was increased using protocols similar
to those already tested by other authors [24, 27]. The lack of randomization and placebo control, the limited sample size represent
the limitations of this research. Taking into account these limitations, we still have to note that the combination of physiotherapy
with subacromial injections showed the better results as suggested by the available literature evidence [28]. Physiotherapy can be
regarded as part of the conservative treatment to be undertaken in subjects with chronic RC tendinopathy, but this treatment alone
is not able to reduce pain and restore shoulder function in long term. A recent Cochrane review [29] concluded that subacromial
corticosteroid injection has insufficient overall evidence to support its usage. It might be beneficial for RC disease and intra-articular
injection for adhesive capsulitis, but their effect is small and not well maintained. We believe that a series of three to four subac-
romial sodium hyaluronate injections could provide good mid and long-term clinical benefits.

Conclusions

Despite the limitations of the current research, such as no randomization and relatively small sample size, we may conclude
that subacromial HA injections could be an effective and safe alternative treatment for patients suffering from RC
tendinopathy. Our results are encouraging but not lasting, and we might suppose that further investigation with a larger sample
size into the optimal injection regime and possible combination with current standard therapies, so as to provide long-term benefits,
are warranted.

Conflict of interest None.

36
36
Efectul injectarii subacromiale de Acid
Hialuronic in boala coafei de rotatori.
Studiu clinic dublu-orb controlat placebo
Alireza Moghtaderi, Sepideh Sajadiyeh, Saeid Khosrawi, Farnaz dehghan, Vahid Bateni

Departamentul de reabilitare si medicina fizica,Universitatea de medicina si radiologie

REZUMAT

BACKGROUND:
Afectiunile coafei de rotatori sunt o cauza obisnuita a durerii de umar. Exista deja studii
despre eficacitatea hialuronatului de sodiu injectabil in durerile de umar si de genunchi,
dar putine studii care sa demonstreze eficacitatea hialuronatului de sodiu injectat sub
ghidaj ecografic in afectiunile coafei de rotatori. Acest studiu evalueaza eficacitatea
ultrasonografiei ghidate a injectarii subacromiale cu hialuronat de sodiu, la pacientii cu
sindrom de impingment fara ruptura completa de coafa de rotatori.

MATERIALE SI METODE:
Acest studiu clinica prospectiv, dublu-orb, controlat placebo a fost efectuat pe 40 de
pacienti cu sindrom subacromial de impingement fara rupere completa a coafei de
rotatori. Pacientii au fost injectati aleator sub ghidaj ecografic in 2 grupuri: grupul studiu
de caz cu hialuronat de sodiu 20 mg (Fermathron™) si grupul control cu solutie salina
0,9%. Ambele grupuri au primit 3 injectii saptamanale. Scorul de durere (scorul visual
analog de durere - VAS) 100 mm a fost evaluat inainte de prima injectie si dupa o
saptamana dupa fiecare injectie. Scorul Constant a fost evaluat dupa prima si a 12-a
saptamana dupa ultima injectie. Datele au fost analizate statistic de testul t-independent.

REZULTATE:
In ambele grupuri scorul mediu VAS a scazut, dar mai semnificativ in grupul de studiu
(p<0.001). Scorul mediu Constant a fost semnificativ mai mare in grupul de studio la 12
saptamani dupa ultima injectie (p<0.001). Scorul Constant s-a imbunatatit 12 saptamani
dupa ultima injectie in ambele grupuri, cu un rezultat semnificativ mai bun in grupul de
studiu (p<0.001).

CONCLUZIA:
Injectiile subacromiale cu hialuronat de sodiu sunt eficiente in tratamentul afectiunii
coafei de rotatori fara ruptura completa.

CUVINTE CHEIE:
Scorul constant, afectiunea coafei rotatorilor, hialuronat de sodiu, injectii subacromiale,
ghidaj ultrasonografic.

37
37
Effect of subacromial sodium hyaluronate injection
on rotator cuff disease : A double-blind placebo-
controlled clinical trial
Alireza Moghtaderi, Sepideh Sajadiyeh, Saeid Khosrawi, Farnaz Dehghan, Vahid
Bateni1

Departments of Physical Medicine and Rehabilitation,1Radiology, Medical University of Isfahan, Isfahan, Iran

Abstract

Background
Rotator cuff disease is a common cause of shoulder pain. There are studies about the effectiveness of sodium hyaluronate injection
on shoulder and knee pain, but few studies demonstrating the efficacy of sodium hyaluronate ultrasonography guided injection
for rotator cuff disease. This study evaluates effectiveness of ultrasonography guided subacromial sodium hyaluronate injection in
patients with impingment syndrome without rotator cuff complete tear.

Materials and Methods


This prospective, double-blind, placebo controlled clinical trial study was performed among 40 patients with subacromial
impingement syndrome without complete tear of rotator cuff. Patients randomly injected ultrasonography guided in 2 groups: Case
group by 20 mg of sodium hyaluronate (Fermathron™) and control group by 0.9% normal saline. Both groups received 3 weekly
injections. The pain score (100 mm visual analogue score [VAS]) was evaluated before first injection and one week after each
injection. The constant score was evaluated before first and 12 week after last injection.
Data was analyzed statistically by Independent t-test.

Results
In both groups mean VAS has decreased, but more significantly in case group (P < 0.001). Mean constant score was significantly
higher in case group 12 weeks after last injection (P < 0.001). The constant score improved 12 weeks after the last injection in both
groups with a significantly better result in case group (P < 0.001).

Conclusion
Subacromial injections of sodium hyaluronate are effective in treating rotator cuff disease without complete tears.

Key Words
Constant score, rotator cuff disease, sodium hyaluronate, subacromial injections, ultrasonography guided

Address for correspondence:


Prof. Saeid Khoshrawi, Departments of Physical Medicine and Rehabilitation,1Radiology, Medical University of Isfahan, Shohadaye
Sofe, Isfahan, Iran
E-mail: khosrawi@med.mui.ac.ir
Received: 05.08.2012, Accepted: 09.02.2013

38
38
Introduction We included patients between 30 and 80 years old who
had shoulder pain, a positive Neer and Hawkins sign,
and a positive ultrasonographic diagnosis of rotator
Shoulder pain accounts for approximately 3 million visits cuff pathology without a complete tear, who did not
to physicians each year in the United States, and of these, respond to conservative treatments or rehabilitation
rotator cuff disease is the most common cause of shoulder for at least 6 months and signed the informed consent
pain necessitating a visit to a primary care physician.[1] form. We excluded patients who (1) had any rheumatic
Rotator cuff disease in particular is a common cause of disease, glenohumeral osteoarthritis, full-thickness
shoulder pain and weakness and comprises a significant rotator cuff tears, fractures, diabetes mellitus, infections,
proportion of the musculoskeletal complaints presenting or tumors, (2) had hypersensitivity to hyaluronate, (3)
to primary care doctors.[2] Treatments for rotator had participated in any other study within 6 months, (4)
cuff lesions without complete tears are mainly had received a subacromial injection within 8 weeks, (5)
conservative[3] and surgery should be considered were pregnant or planned to become pregnant, (6) were
if the patient fails to improve with a progressive at risk of complications of intra-articular injections such
non-operative therapy program of 3-6 months.[4] as patients who received anti-coagulant drugs. Forty
Subacromial injection of anesthetics or corticosteroids is patients randomly allocated in 2 groups: Hyaluronate
often used to treat patients with persistent symptoms after injection (case group) and normal saline injection (placebo
rehabilitation and use of non-steroidal anti-inflammatory group). According to demographic data gender and
drugs.[5] In patients who have failed conservative age, there were no significant differences between two
treatment, there is evidence that viscosupplementation groups. The test drug was Fermathron™, 20 mg/2 ml of
is beneficial for the treatment of knee pain caused by sodium hyaluronate. The placebo was 0.9% normal saline
osteoarthritis.[6] In a study, evaluating rehabilitative and solution, at 2 mL/syringe. Both groups received 3 weekly
infiltrative treatment with hyaluronic acid in elderly patients injections in a same protocol.
with rotator cuff tears found statistically significant results,
both for the reduction of pain and for the improvement of All patients screened for complete tear of rotator cuff
range of motion and autonomy in daily life activities.[7] with ultrasonography and all injections were performed
In one large placebo-controlled, randomized trial of 660 under ultrasonography guidance. Ultrasonography
patients with moderate to severe shoulder pain caused with a high-frequency (7-15 MHz) linear-array probe
by osteoarthritis, rotator cuff tear, or adhesive capsulitis, was performed by one radiologist with a SIEMENS G60
a significant improvement in pain relief was found in all (SIEMENS G 60s, MODEL: 7474922 made in Japan march
groups up to 13 weeks after viscosupplementation.[6] 2006) device.
Another study demonstrated that sodium hyaluronate
injection is effective for the treatment of osteoarthritis Sonographer stood behind the patient, scanning over
and persistent shoulder pain that is refractory to other its shoulder. The long head of biceps tendon was
conservative measurements.[8] Thirty patients with examined within the intertubercular groove, in both the
symptomatic osteoarthritis of the shoulder who failed transverse and longitudinal planes, with the patient’s
conservative treatment and were treated with intraarticular arm in a neutral position and the elbow flexed to 90°.
viscosupplementation demonstrated improvement in The presence of fluid around the tendon was noted, and a
function and pain levels.[9] Treatment with sodium search for fluid in the subdeltoid bursa was made. With the
hyaluronate is emerging as an alternative intraarticular shoulder externally rotated, the subscapularis tendon was
regimen for osteoarthritic knee joints.[10,11] Clinical brought into view and examined in both planes. Then, the
studies have reported the efficacy of hyaluronate injection patient was asked to rotate the shoulder internally. This
in treating shoulder pain.[12-14] In a randomized study, movement was achieved either by placing the forearm
sodium hyaluronate was found to be effective for patients behind the back with the palm facing posteriorly or by
with rotator cuff tears.[15] placing the palm on the upper buttock. The supraspinatus
and infraspinatus tendons were evaluated in this position
Ultrasound is a readily available and cheap modality in both the transverse and longitudinal planes.
for looking more specifically at the rotator cuff and
surrounding structures.[1] More recently, ultrasound The examination was completed by requesting the patient
guidance has become a commonly employed method to to place a hand on the contralateral shoulder. This position
perform diagnostic or therapeutic interventions.[16] allowed further assessment of the infraspinatus tendon
if required. With ultrasound, the normal tendon was an
Despite the long history and the fact that this is a echoic structure, whereas the cartilage and fluids were
common diagnosis made in clinical practice, the exact hypoechoic. Tendinitis was diagnosed when the tendon
cause and best treatment for rotator cuff disease still are lost its echogenicity and became diffusely hypoechoic.
being explored. Although, there are very few high-quality [17-19] Dynamic evaluation of abduction was performed
studies demonstrating the efficacy of sodium hyaluronate by observing the supraspinatus tendon and bursa
injection for rotator cuff disease, this study was performed longitudinally as they retract deep to the coracoacromial
to examine the effect of ultrasound-guided subacromial ligament. Bunching of tissue or buckling of the ligament
sodium hyaluronate injection in patients with impingement correlated with impingement.
syndrome without rotator cuff complete tear.
If ultrasonographic evidence of full-thickness
supraspinatus tendon tear was seen, patient excluded
fromourstudy. These features included non-visualization
MATERIALS AND METHODS of the distal supraspinatus tendon and dipping of the
deltoid muscle or abnormal anechoic disruption of the
This was a prospective, randomized, double-blind, supraspinatus tendon, which extended from the articular
placebo controlled clinical trial involved 48 patients to the bursa surface. The diagnosis of a partial rotator cuff
with subacromial impingement syndrome without tear was made when the hypoechoic or bursal herniation
a complete tear of rotator cuff. Eight patients were did not cross the full width of the tendon.[17-19]
excluded because of protocol violation. The reasons were Ultrasound guided injection was done from a lateral
violation of the selection criteria and refused injection. approach. We used 21 gage needle for injection of 2cc
FermathronTM.

39
39
As long as the needle was kept in the same axial plane lower in case group (P < 0.05). In both groups mean
as the probe, it followed accurately until reached the VAS has decreased every week during the treatment
superficial surface of the supraspinatus tendon. A but this decrement significantly was more in case
preliminary injection in this location confirmed the correct group (P < 0.001) [Table 1].
positioning in the bursa as evident by rapid flow of injected
material away from the needle tip. After insurance firm Although, it showed that there were no significant
placement within the bursa, injection was completed. differences between 2 groups in mean constant score
before treatment (P = 0.4) but mean constant score was
The pain score (100 mm visual analog score [VAS]) was significantly higher in case group 12 weeks after the
evaluated blindly by the different physiatrists before last injection (P < 0.001). The constant score improved
the first injection and 1 week after each injection. The at the 12 weeks after the last injection in both groups,
constant score[20] of shoulder also was evaluated blindly although the independent t-test revealed significantly
before the first injection and 12 week after last injection. A better results in case group (P < 0.001) [Table 2].
constant score of 80 points or more or an improvement in
the constant score by 10 points or more was considered No complications such as flare reaction, infection,
satisfactory. hemarthrosis and synovitis were reported by the
patients in either group. Of the patients in the placebo
Statistical analyses were performed using the SPSS group 15 had additional sodium hyaluronate injections and
statistical package version 13.0 (SPSS Inc., Chicago, IL, 6 were unavailable for follow-up. The remaining patients
USA). Independent sample t-test or Mann-Whitney U-test, were satisfied with their results so hadn’t any additional
paired t-test or Repeated Measure ANOVA test, and Chi- procedures.
square test were used to assess the differences between
stages, as appropriate. A P < 0.05 was considered Of the patients in the case group, 2 had additional
significant. injections 6 months after the last injection, 8 were lost to
follow-up and the remaining satisfied with their results.

Results Discussions
In this study, there were 20 patients in each group, in The literature on the efficacy of intraarticular
case group there were 8 male (40%) and 12 females (60%) viscosupplementation is primarily focused on the knee,
and the placebo group contained 6 males (30%) and 14 with limited studies of other joints. Although, the literature
females (70%). Chi-square test showed that they did not covering visco supplementation for knee osteoarthritis has
differ significantly in gender. been conflicting and has a known publication bias,[21,22]
meta-analyses suggest an improvement in pain compared
Using Independent t-test, revealed that mean VAS with placebo.[22] In patients who have failed conservative
scores did not differ significantly before treatment (P treatment, there is evidence that visco supplementation
= 0.617) but 1 week after the first, second and third is beneficial for the treatment of knee pain caused by
injection (W1, W2 and W3) mean VAS was significantly osteoarthritis.[21,22]

40
40
The clinical effects of sodium hyaluronate on pain associated with osteoarthritis of the knee are probably mediated by several
factors. In vitro and in vivo studies indicate that sodium hyaluronate can enhance prostaglandin synthesis and prevent
its release from the cell matrix. Regarding inflammation, sodium hyaluronate suppresses the production and activity of
pro-inflammatory mediators and proteases as well as altering the function of certain immune cells. Histological evidence
shows that sodium hyaluronate prevents the degradation of cartilage and may promote its regeneration.[23]

Chou et al. performed a randomized, double-blind, placebo-controlled study of sodium hyaluronate (ARTZ Dispo) treatment in
51 patients with rotator cuff lesions without complete tears. They had injections of 25 mg/wk. of sodium hyaluronate into the
subacromial bursa for 5 consecutive weeks in case group and 2.5 ml of normal saline with the same injection protocol for control
group.[24] We performed the study with patients and all the injections were performed ultrasound guided. Additionally, instead of
5 injections, we treated our patients with 3 injections of sodium hyaluronate. The results were the same.

A recent study assessed the hypothesis that injection of high-molecular weight hyaluronate in the treatment of subacromial
impingement syndrome is effective and sodium hyaluronate, compared with corticosteroid injection in the shoulder joint.
They found that subacromial hyaluronate injection to treat impingement syndrome produces similar pain and functional
improvement to corticosteroid at a short-term follow-up.[25]

The probable mechanism of action of sodium hyaluronate is an autocrine stimulation of its own synthesis in a positive
feedback manner in synoviocytes that has been shown in vitro.[26,27]

In our study, the case group had significantly better constant scores than the placebo group at the 12th week after the last
injection and improvement of VAS scores each week after the injections. It shows that although the injection of normal saline
solution into the subacromial bursa may relieve the symptoms of impingement syndrome but the results are not constant.
Furthermore, we know that signs and symptoms of rotator cuff disease may relieve gradually if extrinsic and intrinsic factors are
stabilized by resting or modification of activity.[28] It may be the cause of some degree of improvement after placebo injection.
Another theory is removing inflammatory mediators by normal saline. Sodium hyaluronate may have inhibitory effect on
the function of inflammatory cells and it may account for the deactivation of pain receptors.

In summary, Sodium hyaluronate may contribute to healing and pain reduction in rotator cuff disease. Further studies are
required to find the exact mechanisms of action of this drug.

41
41
42
43
44
44
M
Meta-analize

45
45
45
Acidul Hialuronic si leziunile de tendon

Jean-François Kaux1,2,3
Antoine Samson1, 2
Jean-Michel Crielaard1,2,3
1 Department of Physical Medicine and Sports Traumatology,
CHU Liege, Belgium
2 Multidisciplinary Department for Medicine and Sports
Traumatology (SPORTS), CHU Liege, Belgium
3 Department of Sports and Rehabilitation Sciences,
University of Liege, Belgium

Autor correspondent:
Jean-Francois Kaux
Department of Physical Medicine and Sports Traumatology,
CHU and University of Liege
Avenue de l’Hopital, B35
4000 Liege, Belgium
E-mail: jfkaux@chu.ulg.ac.be

REZUMAT

BACKGROUND:
Introducere: recent, proprietatile vascoelastice ale acidului hialuronic pe tesutul
conjunctiv lichid au fost propuse ca tratament in tendinopatii. Cateva studii fundamentale
arata rezultate incurajatoare in ceea ce priveste abilitatea acidului hialuronic de a
contribui la alunecarea tendonului si de a reduce aderentele, ca si la imbunatatirea
organizarii arhitecturale a tendonului. Unele observatii sustin deasemenea folosirea lui
clinic pentru a imbunatati durerea si functia. Aceasta revizuire a literaturii analizeaza
studii legate de folosirea Acidului Hialuronic in tratamentul tendinopatiilor.

METODE:
Aceasta revizuire a fost construita folosind baza de date Medline prin Pubmed, Scopus
si Google Scholar. Cuvintele cheie folosite in cercetare au fost: Acid Hialuronic, tendon
si tendinopatie.

REZULTATE:
In total au fost selectate pentru relevanta si calitate stiintifica 28 de articole (in engleza
si Franceza) legate de aplicarea Acidului Hialuronic pe tendoane, incluzand 13 pentru
partea de in vitro, 7 pentru in vivo pe animale si 8 pentru sectiunea umana.

CONCLUZIA:
Studiile preclinice demonstreaza rezultate incurajatoare: Acidul Hialuronic permite
alunecarea tendoanelor, reduce aderentele, creaza o mai buna oraganizarea arhitecturala
a tendonului si limiteaza inflamatia. Aceste observatii de laborator par sa fie sustinute
de rezultatele clinice limitate, dar incurajatoare pe termen scurt asupra durerii si
functionalitatii. Cu toate acestea, sunt inca necesare studii randomizate controlate.
46
46
Hyaluronic acid and tendon lesions
Jean-François Kaux1,2,3
Antoine Samson1, 2
Jean-Michel Crielaard1,2,3
1 Department of Physical Medicine and Sports Traumatology,
CHU Liege, Belgium
2 Multidisciplinary Department for Medicine and Sports
Traumatology (SPORTS), CHU Liege, Belgium
3 Department of Sports and Rehabilitation Sciences,
University of Liege, Belgium

Autor correspondent:
Jean-Francois Kaux
Department of Physical Medicine and Sports Traumatology,
CHU and University of Liege
Avenue de l’Hopital, B35
4000 Liege, Belgium
E-mail: jfkaux@chu.ulg.ac.be

Summary
Introduction: recently, the viscoelastic properties of hyaluronic acid (HA) on liquid connective tissue have been proposed for the
treatment of tendinopathies. Some fundamental studies show encouraging results on hyaluronic acid’s ability to promote tendon
gliding and reduce adhesion as well as to improve tendon architectural organ- isation. Some observations also support its use in
a clinical setting to improve pain and function. This literature review analyses studies relating to the use of hyaluronic acid in the
treatment of tendinopathies.
Methods: this review was constructed using the Medline database via Pubmed, Scopus and Google Scholar. The key words
hyaluronic acid, tendon and tendinopathy were used for the research.
Results: in total, 28 articles (in English and French) on the application of hyaluronic acid to tendons were selected for their relevance
and scientific quality, including 13 for the in vitro part, 7 for the in vivo animal part and 8 for the human section.
Conclusions: preclinical studies demonstrate encouraging results: HA permits tendon gliding, reduces adhesions, creates better
tendon architectural organisation and limits inflammation. These laboratory observations appear to be supported by limited but
encouraging short-term clinical results on pain and function. However, controlled randomised studies are still needed.

KEY WORDS: hyaluronic acid, tendinopathy, tendon, healing, cicatrization.

Introduction
Tendon lesions are very common in sportsmen and physical workers1. Their physiopathology is multifactorial, including
intrinsic and extrinsic factors, which can be modified or not2. Even if detrained tendons present the same histological structure
as chronic tendinopathies, however, this condition remains an overuse pathology which is often difficult to treat2. For this reason,
various new treatment have been developed1 such as platelet-rich plasma (PRP)3 or growth factors4 infiltrations.
Recently, the anti-inflammatory and lubrication properties of HA have also been suggested for the treatment of these
pathologies5.
Hyaluronic acid (HA) is a polysaccharide with a high molecular weight (between 100 kDa and 10 MDa), mainly found in the
extracellular matrix where it plays different roles at various places in the organism5. Its half-life is only three to five minutes in
the circulation, less than a day in tissue, and between one and three weeks in cartilage. This speed of renewal means that this
polymer is constantly synthesised and degrad- ed5. HA can be categorised into three groups based on molecular weight:
- low molecular weight: MW < 1MDa;
- medium molecular weight: 1MDa < MW < 2MDa;
- high molecular weight: MW > 2MDa.
HA ensures the viscoelasticity of liquid connective tissues such as the synovial joint and the corpus vitreum of the
eye5. As a result of this ability, its role in the body is, among other things, to bind to water and to lubricate mobile parts of
the body such as the joints and muscles5. Intra-articular injections of synthetic HA are widely used in the medical treatment of
osteoarthritis6-8.
Industrially, HA is obtained through two different procedures:
- extraction from rooster combs, after grinding and chemical treatment5;
- purification and bacterial fermentation5.
The viscosity of the gel which is obtained is proportional to the length (molecular weight, expressed in Daltons) of the polymer fibres.
This viscosity will determine how quickly the product degrades after implantation.
One major absolute contraindication is hypersensitivity to one of the ingredients. Potential undesirable side-effects are pain, a
feeling of heat, haematoma, redness and swelling.

47
47
Material and methods

This literature review was constructed using the Med-line database via Pubmed, Scopus and Google Scholar, according to the
recommendations of the Journal9. Articles published in English and French, up to the 30th of September 2015, were found
using the following key words alone or in combination: hyaluronic acid, hyaluronan, tendon, tendinopathy, treatment (in English)
and acide hyaluronique, tendon, tendinopathie, traitement (in French). In total, only 28 articles on HA as applied to tendons were
selected for their relevance and scientific quality: 13 for the in vitro part, 7 for the animal in vivo part and 8 for the human section.

Results

In vitro experiments

Even if the first published paper studying the effect of HA on tendons of rabbits has not shown any effect on the functional strength
of tendons after healing in vitro10, several Authors have sought to identify the ability of HA to improve tendon gliding. Sun et al.
used 30 canine peroneus longus, randomised into five groups, and immersed for 30 seconds in one of the following five
solutions: saline solution, gelatin, carbodiimide and HA (cd-HA) and gelatin (cd-gelatin) and cd-gelatin HA11. The tendons
were first placed in an incubator at 100% humidity at 37°C, before being wrapped in damp cloths for one hour. Gliding resistance
for each tendon was measured before the surfaces of the tendons were coated in the various solutions. After 500 cycles, gliding
resistance for the tendons coated in saline solution increased significantly, by a factor of 10, while the cd-gelatin and cd-
gelatin-HA did not significantly change. In addition, under microscopy, the surface of the tendons in the saline, gelatin and
cd-HA solutions were rough, presenting a network of disorganised fibres. In contrast, the surface of the tendons immersed
in the gelatin-cd-HA solution appeared smoother than in the other groups. These observations were confirmed by the study
of Nishida et al., which suggest that HA may improve the gliding function of canine flexor tendons12.
Taguchi et al. wanted to identify whether these results could be transposed to human tendons13. They used 16 human
extrasynovial palmaris longus tendons which were divided into two and then immersed randomly in one of the following four
solutions: saline solution, carbodiimide-gelatin-HA (cg-gelatin-HA), carbodiimide-gelatin-lubrucin and cd-gelatin-HA-lubrucin. The
results, after 1,000 flexion/extension cycles, show that the tendons treated with saline solution presented significantly higher
gliding resistance than other groups. These findings, suggesting that HA may limit tendon adhesions, has already been
observed in a previous study of Akasaka et al14.
In addition to its effects on tendon biomechanics, the researchers looked at the influence of HA on tendon
regeneration.
Yamada et al. sought to determine the influence of HA on cell proliferation and mRNA expression of type I and type III
procollagens15. They used tendons from 10 subjects suffering from rotator cuff disease who had undergone surgical repair to the
cuff. For each fragment, the tendon tissue was separated from the synovial tissue, cut into small pieces and cultured in solutions
containing various concentrations of HA (1.0 to 5.0 mg/mL). The results showed HA binding to its receptors and a reduction
in cell proliferation in a dose-dependent manner in comparison with untreated cells. In addition, the expression level of
procollagen α1 (I) mRNA did not significantly reduce in contrast to the expression of procollagen α1 (III) mRNA in a dose-dependent
manner.
Another study by Shimpuku et al. analysed the suture site of the supracoracoid tendon in chickens after injection of HA16.
As well as confirming that HA treatment does not increase the production of α1 (I) procollagen, the team was able to
show early restoration of tendon continuity at the laceration site in comparison with the control group treated with saline
solution. Wiig et al. had compared the effect of HA on cell proliferation and synthesis of matrix component in intrasynovial deep
extensor tendons and extrasynovial peroneus tendons of rabbits17. They observed that HA modulates cell proliferation unequally
in intra and extrasynovial tendons, but without affecting the matrix component synthesis. This can be partly explained by
a difference in biochemical composition and cellular capacities of tendons and by a possible difference in HA sensitivity
and in its mechanism of action.
Osti et al. had compared 4 different HA preparation by their molecular weight on human tendon derived cells from rotator cuff
tears18. They observed that all the HA increased the cell viability and the proliferation at 24 hours compared to control
cells (without HA). Furthermore, the synthesis of collagen type I was stimulated over 14 days, without increase in collagen type III.
However, these observations were significantly higher in the high molecular weight HA groups.
Neumann et al. looked at the effects of HA on proinflammatory action caused by advanced glycation endproducts (AGEs)19. The
team compared the efficacy of a preparation of HA with a ‘high’ molecular weight (HMW-HA - >1.2MDa) to a preparation of HA with
a low molecular weight (LLW-HA - 0.2-0.5MDa) on the production of AGEs and activation of proinflammatory cytokines on murine
macrophages. Their results show that HMW-HA, with a molecular weight greater than 1.2 MDa, inhibits the inflammation
sig- nalling pathway (NF-kB) caused by AGEs and that this effect reduces with the molecular weight of HA. Moreover, the
results show that low molecular weight HA (<0.5MDa) is proinflammatory.
Tanimoto et al. studied the effect of proinflammatory cytokines on HA-synthetase (HAS) mRNA expression using rabbit synovial
membrane cells, cultured in inflammatory conditions IL1-β and TNF-α were used20. Their results showed that proinflammatory
cytokines increase HAS mRNA expression and, consequently, may contribute to the accumulation and fragmentation of
HA. Low molecular weight HA molecules no longer played their lubricating role in an optimal way. A study by Smith et al. highlighted
the effect of adding exongenous HA to endogenous HA synthesis in synovial fibroblasts21. Human synovial fibroblasts obtained
dur- ing biopsies or arthroplasties were cultured. Different molecular weights of HA were then added.
The results clearly showed that in vitro HA synthesis by these cells increased when the concentration and molecular weight
of the added HA was increased. The recent study of Nakamura et al., comparing the effects of corticosteroids or HA used in
subacromial injection on cell proliferation, in human tendon fibroblasts from rotator cuff tears and in rats after surgical lesion,
demonstrated that HA possesses anti-inflammatory and anti-adhesive activities in tendon and synovial fibroblasts. In
contrast to CS, HA caused no adverse effects and appears to be relatively safe22.

48
48
Animal experiments

Zhao et al. wanted to identify the effect of HA on the reduction of adhesions and on functional improvement after failed tendon
reconstruction23. To simulate this situation, 28 flexor tendons from the 2nd and 5th digits of dogs were lacerated and repaired. One
graft tendon in each dog was treated with an injection of saline solution, and the other with an injection of HA (1%) combined with
lubricin (cd-HA-lubricin). Six weeks after the surgical operation, the allografts treated with cd-HA-lubricin showed decreased
adhesions at the proximal extremity, improved digit function and increased graft gliding ability. However, rupture resistance
at the proximal and distal repair sites was lower in the treated group than in the control group. Moreover, histological analysis
showed delayed healing in the treated group. A study by Oryan et al. aimed to assess the effects of high molecular weight (1.6MDa)
HA (0.9%) on the healing of flexor tendons in the digits of rabbits after lesion and repair through surgical suture24. Injections
were administered to 40 rabbits, randomly divided into two equal groups of injured treated with HA and injured untreated. The
results showed a significant reduction in the diameter of the treated tendons and an increase in echogenicity. In addition,
the dry weight of the injured tendons had significantly increased in comparison with the non-treated tendons. The treatment also
significantly improved tenoblast numbers and the diameter and density of collagen fibrils. Yoshida et al. also demonstrated
that high molecular weight HA (2700 kDa) was effective for pain relief and for partial restoration of the patellar tendinopathy
in a rat model, compared to saline or nothing (control group)25.
The Liang et al. team wanted to show the effect of the presence of tenocytes when injecting HA into the Achilles tendons of rats
at an early healing stage26. After transection and suture of the Achilles tendon, 96 rats were randomised into three groups: one
group (THA) received an injection of 1.0 kDa HA (1.5%) with tenocytes (5x105), one HA group received the same injection without
tenocytes, and a control group received a saline solution injection.
A significant difference in recovery, tendon stiffness and locomotive ability was observed in the THA group. The THA group
also had a shorter healing time, ending the inflammatory phase and starting the proliferation phase from day 7, while
the non-treated and HA groups were still in the inflammatory phase of the healing process on day 7. Acceleration of the
inflammatory process through injection of THA may be the main reason for this functional improvement. In addition, the few
differences measured between the tendons treated with HA and the tendons injected with saline solution may be explained
by the low molecular weight (1.0 kDa) of HA used. This study shows that adding tenocytes to HA injections may positively
influence tendon repair by reducing inflammatory response time. However, the use of low molecular weight HA in this study
makes comparisons with THA difficult. Moreover, the use of THA appears to be more appropriate for surgical interventions
on tendons than in tendinopathies, given the constraints of removing tenocytes.
The recent series of Salamanna et al. comparing the mechanobiology of patellar tendons of rats randomized into 3 groups
(untrained, trained and detrained), demonstrate that discontinuing training activity in the short term alters tenocyte synthetic
and metabolic activity and that repeated peri-patellar infiltrations of HA during detraining allow the maintenance of
tenocyte anabolic activity27: higher proliferation rate and viability, and increased synthesis of C-terminal-propeptide of type I
collagen, fibronectin, aggrecan, tenascin-c and matrix-metalloproteinase-3. These results are reinforced by those of Frizziero et al.,
comparing the same groups, who demonstrated that HA is effective in the maintenance of the structural properties of patellar
tendon and enthesis in detrained rats28.
Contrary of the in vitro observation, Yamaguchi et al. suggested that both hyaluronic acid and steroid injections suppressed of
inflammation (evaluated by the expression of calcitonin generelated peptide - CGRP) in a rotator cuff of rats, which thought to
be provided pain relief (analyzed by the gait analysis)29. While there were no significant differences, the suppression of CGRP
expression and the improved gait after hyaluronic acid and steroid injections suggested that both methods were effective for rat
rotator cuff tear model.

Human experiments
Meloni et al. examined the effect of periarticular injection of HA in patients suffering from a supraspinatus tendinopathy30.
A total of 56 patients participated in the study and were randomised into two equal groups: one group treated with an injection
containing 20 mg of HA (concentration: 20mg/2ml, molecular weight: 0.5-0.7 mDa) in 2 ml of lidocaine (1%) and 2 ml of physiological
liquid (0.9%) and a control group receiving an injection of 4 ml of physiological solution (0.9%). Lidocaine is used to reduce pain
due to the injection. After the first injection, the injection was repeated once a week for four weeks. During this period, analgesics
were prescribed for three days after each injection. After one year, 19 of the 28 patients who had received HA were satisfied
with their treatment, while all patients in the control group said they were dissatisfied after their injections. Joint extension
remained identical throughout the follow-up. At months 1 and 3 following the injection, a significant improvement in pain in
the treated group in comparison with the control group was noted. However, after one year, this pain had increased in the
treated group. Moreover, no significant difference in tendon structure was observed during the ultrasound scan between
the two groups either at the start or at the end of the follow-up.
Petrella et al. treated 330 competitive racquet sports athletes presenting with chronic lateral epicondylosis for more than three
months31. Two injections were administered one week apart, into the subcutaneous tissue and muscle one centimeter from the
lateral epi- condyle toward the primary point of pain using a two-dimensional fanning technique: the treated group received an
injection of 1.2 ml of HA (high but unspecified molecular weight) at 1% and the control group received a saline solution of 1.2 ml.
Follow-up examinations were conducted after 7, 14, 30, 90 and 356 days and consisted of a VAS at rest and during measurement
of grip strength using a Jamar dynamometer, an elbow disability scale, a functional scale, a satisfaction scale and a measure of time
to return to pain-free sport. After one year, there was a significant improvement in pain at rest and grip. Moreover, the scores on all
scales had significantly improved in the treated group in comparison with the control group. The time to return to pain-free
sport was assessed at 18 days (+/-11 days) in the treated group.
The efficiency of an HA injection for treating patellar tendinopathies was determined in fifty patients32 with stage 2 or 3 using
Blazina’s classification33. The injected solution contained 2.5 mg of HA with a molecular weight of 900 kDa in 2.5 ml of physiological
liquid and 1 ml of lidocaine. The effectiveness was assessed on the basis of pain during sport, using the modified Roles and
Maudsley scores34. After treatment, 54% of cases were ‘excellent’, and 40% of cases were ‘good’. The results of this study
are based on the subjective views of the athletes and their trainers. With a view to reducing post-surgical adhesions of digital
flexor tendons, Riccio et al. studied 45 patients randomly assigned to two groups: a control receiving a placebo (n=10), and a
treated group receiving an injection of HA (high but unspecified molecular weight) (n=26).

49
49
All patients were assessed at 30, 60, 90 and 180 days after surgery by testing active motion, with functional capacity being assessed
using the quick-DASH questionnaire and the number of working days lost after surgery. Patients in the treated group showed
better recovery of digital motion at all time intervals (improvement of 27% in the HA group compared to 10% in the control
group, and 43 to 28% respectively at day 180), and returned to work earlier. However, no significant difference could be
shown in terms of functional score between the two groups.
More recently, HA injections has been used to treat the flexor muscle in the index finger after surgical suture36. 11 were treated
with three HA injections (0.4 ml of high molecular weight: 1.0-2.9 MDa and high concentration: 15 mg/ml) and 11 received a placebo
saline solution of identical volume. Three injections were administered: one at the time of the surgical intervention and two others
one week apart. The two groups followed the same rehabilitation protocol after the operation. The results showed no differences
between the two groups in terms of extension after three weeks. However, at three months and over the long term, significant
functional improvement of total active range of joint motions (p=0.002 and p=0.001 respectively) was noted in the treated group in
com- parison with the placebo.
The short and long-term effect of intra-articular injection of HA in patients suffering from supraspinatus tendinopathy37
was assessed in 24 patients randomised into two groups, each received three injections of 2 ml of HA (16 mg/2 ml
with a molecular weight of 6.0 MDa), following traditional physical therapy treatment. TENS current, ultrasound therapy,
thermotherapy (hotpack) and exercises at home (movements, stretches and eccentric exercises). Follow-up consisted of algo-
functional score, measurement of active and passive joint extension conducted at three weeks, three months and four years post-
injection. A significant improvement in most of the parameters measured was seen within both groups, but no significant
difference was observed for the pre-injection VAS in comparison with the VAS conducted at three weeks and three months
post-injection.
The series of Merolla et al. enrolled 48 patients with rotator cuff tendinopathy for more than 4 months randomized into 2 groups: one
group with 2 ultrasound-guided subacromial injection of HA (N=25) 14 days apart and one group with only a rehabilitation
pro-gram (N=23)38. The follow-up went up to 6 months. They observed that subacromial HA injections was more effective
on pain and algofunctional scores than rehabilitation alone, and this injection was a safe treatment for patients suffering
from rotator cuff tendinopathy.
A recent study of Kumai et al. evaluated the effects of a 2,5 mL HA (high-molecular-weight: 2700 kDa) injections, over a period of
1 week, in a total of 61 patients with enthesopathies: 16 with lateral epicondylitis,14 with patellar tendinopathy, 15 with insertional
Achilles tendinopathy, and 16 with plantar fasciitis39. They observed a significant decrease of pain (VAS) for the 4 sites (-2.20 ± 2.26
for the 4 sites overall), suggesting the efficacy of injections of HA for enthesopathies in the short-term.

Discussion
Tendinopathies are very common condition in sports and working population1. Tendinopathy and tenosynovitis disturbs tendon
gliding. The application of HA can reduce tendon glide resistance10-14. Several earlier studies have shown the link between
the inhibition of fibroblast proliferation and a reduction in type III collagen concentration in the tendon, with a reduction
in the formation of adhesions at the tendon healing site 40-42. By limiting the proliferation of fibroblasts and by stabilizing
the concentration of type III collagen, HA may reduce the risk of adhesions10-12. It is still discuss if HA can stimulate or not
the synthesis of type I collagen to improve the tendon structure and resistance5,18, but it is demonstrated that HA improve cell
viability and proliferation17,18. HA appears to inhibit the expression of key intermediaries for the inflammatory signaling pathways
(NF-kB), in a dose-dependent manner19,43. By reducing the expression of proinflammatory factors, adding exogenous HA
reduces the fragmentation of endogenous HA and stimulates synovial synthesis of endogenous HA16,26,36.
Concerning the therapeutic protocol for HA injections for tendinopathies, comparatively to other recent treatments for these
conditions, where post-injection rehabilitation could be crucial44, such as PRP for example45, only 1 or 2 injections alone are
described for HA treatments30-32,35-39. However, comparatively to PRP treatments, where it exist a wide variability in the
administered product46,47, the molecular weight and the concentration of HA are known18; high molecular weight HA seems to
have a better effect on tendons18 in the short-term.

Conclusion
Some in vitro and animal in vivo studies demonstrate encouraging results in terms of the ability of HA to encourage tendon
gliding and to reduce adhesions as well as creating better tendon architectural organisation. Moreover, the injection of
HA may limit the proinflammatory effect by restoring viscoelasticity and by stimulating the endogenous synthesis of HA. These
laboratory observations appear to be supported by limited but encouraging short-term clinical results on pain and function as well
as on the return to sport. However, controlled randomised studies are still needed.

50
50
Utilizarea Acidului Hialuronic dupa interventia
chirugicala la nivelul tendonului si in tendinopatii
Michele Abate, Cosima Schiavone, si Vincenzo Salini

Departamentul de Medicina si Stiintele Imbatranirii, Universitatea G. d’Annunzio,


Chieti-Pescara
Via dei Vestini 31, 66013 Chieti, Italia
Corespondenta tebuie adresata catre Michele Abate; m.abate@unich.it
Primit in 11 February 2014; Acceptat in 17 April 2014; Publicat 8 May 2014
Editor Academic: Lorenzo Cavagna
Copyright c 2014 Michele Abate et al.

Vascosuplimetarea cu Acid Hialuronic este sigur si eficace in managementul osteoar-


tritei, dar utilizarea lui in tratamentul afectiunilor tendoanelor a primit mai putina atentie.
Scopul acestei analize este sa sumarizeze cunostintele actuale despre acest subiect
evaluand studii experimentale si clinice. A fost realizata o cautare a articolelor in limba
engleza folosind termenii cheie: ”Acid Hialuronic” sau “vascosuplimentare”, combinat
cu “tendon”, “tendinopatie”, “aderente” sau “alunecare” luate separate. In aproape
toate studiile experimentale efectuate dupa procedurile chirugicale pentru leziunile
de tendon sau folosite in tratamentul tendinopatiilor cronice, folosind diversi compusi
cu Acid Hialuronic, au fost observate rezultate positive (formarea redusa de tesut de
granulatie si cicatricial dupa reparatia tendonului, mai putine aderente si rezistenta la
alunecare, si o imbunatatire a vindecarii tisulare). Dupa interventia chirurgicala asupra
tendonului flexor, s-a inregistrat o functie totala de miscare mai mare, precum si a de-
getelor cu o reintoarcere mai rapida la activitatile zilnice si la munca. Similar la pacientii
care sufereau de afectiuni ale tendoanelor umarului, cotului si patelei, durerea a fost
redusa si functia imbunatatita. Efectul pozitiv al Acidului Hialuronic poate fi atribuit ac-
tivitatii anti-inflamatorii, imbunatatirii proliferarii celulare si a depunerii de colagen, pe
langa actiunea lubrifianta pe suprafata de alunecare a tendonului.

51
51
Hindawi Publishing Corporation
BioMed Research International
Volume 2014, Article ID 783632, 6 pages
http://dx.doi.org/10.1155/2014/783632

Review Article
The Use of Hyaluronic Acid after
Tendon Surgery and in Tendinopathies

Michele Abate, Cosima Schiavone, and Vincenzo Salini


Department of Medicine and Science of Aging, University G. d’Annunzio, Chieti-Pescara,
Via dei Vestini 31, 66013 Chieti, Italy
Correspondence should be addressed to Michele Abate; m.abate@unich.it
Received 11 February 2014; Accepted 17 April 2014; Published 8 May 2014
Academic Editor: Lorenzo Cavagna
Copyright © 2014 Michele Abate et al.This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Viscosupplementation with hyaluronic acid is safe and effective in the management of osteoarthritis, but its use in the treatment of
tendon disorders has received less attention.The aim of this review is to summarize the current knowledge on this topic, evaluating
experimental and clinical trials. A search of English-language articles was performed using the key search terms “hyaluronic
acid” or “viscosupplementation” combined with “tendon,” “tendinopathy,“ “adhesions,“ or “gliding,“ independently. In quite all
the experimental studies, performed after surgical procedures for tendon injuries or in the treatment of chronic tendinopathies,
using different hyaluronic acid compounds, positive results (reduced formation of scars and granulation tissue after tendon repair,
less adhesions and gliding resistance, and improved tissue healing)were observed. In a limited number of cases, hyaluronic acid
has been employed in clinical practice. After flexor tendon surgery, a greater total active motion and fingers function, with an
earlier return to work and daily activities, were observed. Similarly, in patients suffering from elbow, patellar, and shoulder tendons
disorders, pain was reduced, and function improved.The positive effect of hyaluronic acid can be attributed to the anti-inflammatory
activity, enhanced cell proliferation, and collagen deposition, besides the lubricating action on the sliding surface of the tendon.

Introduction actively secreted by the tendon sheath and, as for joints, it is


an important component of the synovial fluid, which allows
a smooth tendon gliding, and provides nutrition to tendon
Hyaluronic acid (HA) is an important component of articular itself [3]. Moreover, it is an important component of tendon
cartilage; it is present as a coat around chondrocytes, where structure, being largely present in extracellular space.
it bounds to aggrecan monomers, which imbibe water and The aim of this narrative review is to summarize the current
are responsible for the resilience of cartilage (i.e., resistance knowledge on this topic, evaluating experimental data
to compression) [1]. Moreover, HA is a major component and clinical trials. A search of English-language articles
of the synovial fluid, and, along with lubricin, it is one of was performed in Pubmed, Web of Knowledge (WOK),
the fluid’s main lubricating components. The biological and EMBASE using the key search terms “hyaluronic
activities of HA are very complex: (a) it inhibits matrix acid” or “viscosupplementation” combined with “tendon,”
metalloproteinases (MMPs) and the phagocytic activity “tendinopathy,” “adhesions,” or “gliding,” independently.
of macrophages and leukocytes; (b) it promotes the Bibliographies were hand-searched to include any applicable
release of prostaglandins and the production of tissue studies that were not captured by our search.
inhibitor of MMP-1 and favours the normalization of Articles were eligible if they provided specific information
native hyaluronan synthesis; (c) it acts as free radicals related to the correlation between hyaluronic acid and
scavenger and stimulates proteoglycans synthesis by tendon disorders.
chondrocytes; (d) finally, it is provided of protective
effects on chondrocytes or cartilage explants from
degradation by enzymes [1]. Rationale
Several clinical trials have shown that viscosupplementation
therapy with HA is safe and effective in themanagement HA has been mostly used after a surgical procedure
of osteoarthritis (OA) resistant to conventional therapies for tendon injuries or in the treatment of chronic
[2]. This treatment has been approved by Food and Drug tendinopathies.
Administration for knee osteoarthritis, whereas for the Flexor tendon injuries of the hand are common, especially
other joints there are very promising results but not in the young and working-age population [4]. After surgical
conclusive evidence. repair, the finger motion can be dramatically impaired by
Despite the positive results inOA, its use in the treatment of oedema, and later by excessive scar formation, and/or by
tendon disorders has received less attention. Actually, HA is adhesions between the tendon and sheat or other tissues [5].

52
52
To limit these negative outcomes, a proper balance compared to low molecular weight HA [7]. Indeed, in the
between protection and mobilization is required. Indeed, experiments carried out by Kolodzinskyi et al. [7] on the
especially in Zone II, a region where the flexor digitorum isolated canine peroneus longus (PL) the gliding resistance
profundus and superficialis tendons reside within a of the tendon, treated with Hylan G-F 20, decreased
fibroosseous pulley system, immobilization is useful to significantly compared to untreated controls.
allow tendon union but often results in adhesions that Other authors have supposed that adhesions could be
impair the physiologic gliding [4]. Postoperative protected reduced with more stable HA derivatives and HA could
mobilization, either active or passive, is usually performed, be fixed to the tendon surface, and its removal could be
but early motion may have detrimental effects, contributing minimized [8–10, 19, 20]. Several HA derivatives have been
to gap formation or tendon rupture, while late mobilization evaluated.
may favour adhesions [6]. The carbodiimide-derivatized HA (cd-HA) is less
In complicated injuries, primary repair of a flexor tendon is soluble in water than normal HA and therefore has an
often impossible. In these situations, tendon grafts, using increased tissue residence time [19, 20]. Preliminary
extrasynovial tendons (e.g., palmaris longus or plantaris), experiments, erformed on intact tendon grafts in vitro,
are often used to reconstruct severely scarred or missing showed that the preparation improved gliding and
tendons [7]. The extrasynovial grafts have the advantage that the effect was increased when gelatin was added
of being easily harvested, but the higher gliding resistance [8]. Experiments were therefore performed on isolated
compared with intrasynovial tendons, due to adhesion flexor digitorum profundus tendons in dogs, submitted
formation, represents a main disadvantage on their use to dissection, followed by surgical repair.These studies
[8]. For this reason, physical barriers and biological showed that, from the 50th mechanical stress cycle
techniques have been employed to reduce tendon onwards, the gliding resistance was significantly lower in
adhesionswithout adversely affecting the healing the cd-HA gel group than in the control group, without any
process [9, 10]. In this context, HA has been considered difference in the breaking strength nor in tendon stiffness.
as a useful therapeutic tool. These results were reported to an increased residence
However, in a limited number of cases, HA has been time of HA on the tendon surface, favouring the cross-
also employed in the treatment of tendinopathies [11– linking of the HA mixture to tendon proteins [20].
13].HAis an essential component of the tendon itself. It Further studies were carried out by Zhao et al. [18], who
iswell known that, after experimental damage, tendon studied a compound, characterized by a cd-HA and
healing process proceeds along a complex pathway gelatin, added to lubricin, amucinous glycoprotein
beginning with inflammation and cellular proliferation present in the peritendinous fluid. Flexor digitorum
and followed by tissue formation and maturation, profundus tendons from the 2nd and 5th digits of one
with each phase lasting for days, weeks, and months, forepaw of six dogs were transected and repaired. One
respectively [14]. tendon in each paw was then treated with HA; the other
Briefly, in the damaged area, the injury molecular products repaired tendon was not treated.
stimulate an acute inflammatory reaction with the secretion Following tendon repair, a forearm cast was applied to fully
of cytokines, reactive oxygen species, cationic peptides, immobilize the operated forelimb. After 3 and 7 weeks, the
or proteases. This phase is followed by cell proliferation, dogs were euthanized and the tendons were evaluated.
collagen and matrix deposition, and tissue remodeling and The adhesion formation was assessed in two regions,
the final outcome is scar tissue formation, which can between the tendon and the sheath and between the
partially restore tendon function. Indeed, the mechanical tendon and the bone, using a semiquantitative scoring
properties are compromised for years, due to composition system (none, mild,moderate, and severe).The normalized
of the extracellular matrix and its organization. work of flexion, the gliding resistance, and the maximum
Basic research has shown that the activity of breaking force of treated tendons were also recorded. In
hyaluronidase 2 is increased in granulation tissue theHA-lubricin group, the normalized work of flexion was
during the healing of equine superficial digital flexor significantly lower, as well as the prevalence of severe
tendon injuries, suggesting that HA plays a relevant adhesions. However, also the maximum breaking force
role in controlling the healing process in equine was reduced, suggesting that the positive effects of the
tendonitis [15]. compound on adhesions were counteracted by some
impairment of tendon healing. The hypothesis was that
lubricin could enter the repair site, blocking the adhesion
of the healing tendon ends to each other. The authors
Experimental Studies suggest that a gelatin patch, sealing the tendon
and carrying growth factors, could provide a better
Several studies have been performed to evaluate protection of the lacerated tendon ends from lubricin
the efficacy of HA on adhesions, gliding resistance, leakage and enhance teh flexor tendon intrinsic
and tendon healing [7–10, 16–21]. Different healing[21].
experimentalmodels have been used: animal species Another HA-derived biomaterial is Seprafilm, a biore-
(dog, chicken, rabbit, rat, and horse), tendons (intra- or sorbable membrane composed of chemically modified
extrasynovial; auto- or allograft), and procedures of HA and carboxymethylcellulose [9]. This device,
induced tendon damage (surgical, collagenase, or steroid however, suffers from several drawbacks, such as the
lesion). Moreover, a lot of HA compounds (low and high short in vivo residence time and the difficulty to handle
molecular weight HA, cross-linked HA) have been studied when wrapping the tendon, because the material loses
in the form of gel, membrane, or scaffold, administered its integrity. In contrast, the Carbylan TM-SX (a new
intraand peritendinously, both in vitro and in vivo. carboxylated and thiol-modified HA-derivative) films
Quite all the authors have shown that HA reduces are elastic, robust, and easy to handle and can maintain
the formation of scars and granulation tissue and their integrity during manipulation. This preparation
prevents adhesions after tendon repair. The efficacy has been shown to be more effective in reducing the
seems in some way dependent on the HA used. Indeed, peritendinous adhesions, following partial thickness
the native HA has a limited action because of a very short injury in rabbits, compared to Seprafilm and Carbylan
half-life and a too rapid elimination to serve as a physical TM-SX sprayable gel [10]. The favourable activity was
barrier between the tendon and the sheath [3]. A slight attributed to the inhibitory activity of thiol-modified HA
superiority was found when using the high molecular on cell attachement, spreading and proliferation with the
weight HA (Hylan G-F 20) which has a longer half-life compound being negatively charged, hydrophilic, and

53
53
lacking peptidic epitopes that activate integrins. Seprafilm antiadhesion membrane could be effective
The few studies, at present performed in humans in human in restoring the morphological properties of injured
isolated tendons, confirming experimental research, suggest superficial digital flexor tendon of rabbits and might be
that tendon surface treatment using HA (cd-HA or added helpful for future clinical trial studies in tendon ruptures.
with lubricin) can reduce the excursion resistance in
the tendon-pulley unit in the intra- and extrasynovial
tendons, facilitating postoperative rehabilitation and Clinical Trials
improving the clinical outcome [22, 23].
These studies, taken together, suggest that HA Experimental evidence suggests that, in the clinical setting,
diminishes the excursion resistance after tendon repair it may be possible to inject HA within the flexor sheath
and can be useful to prevent adhesion until the synovial during the postoperative period in patients at risk for
surface is fully developed. adhesions or with noteworthy oedema.
Less experimental work has been performed to assess In a multicenter randomized controlled trial, Hyaloglide,
whether HA can be effective in tendon healing [15, 17]. a thick and sticky gel which adheres to the surface of the
In Achilles tendonitis, induced by repeated injections of tendon, was applied after flexor tenolysis in Zone II following
corticosteroids in rats, the effects of Hylan G-F 20 were failed flexor tendon repair [4]. 30, 60, 90, and 180 days after
investigated by means of histopathology [17]. For the tendon, surgery, patients in the study group, compared with not
thickness, staining affinity, nuclear appearance, and fibrillar treated controls, showed a greater total active motion and
appearance were taken into account, whereas, for paraten- finger function, with an earlier return to work and daily
on fibrosis and oedema, capillary changes and inflamma- activities. Noteworthy, Hyaloglide did not affect tendon and
tion were assessed, according to a semiquantitative scor- wound healing and did not increase the complications rate.
ing system. Compared to controls, injected with saline, Similarly, O¨ zgenel and Eto¨z [5] investigated the efficacy of
Hylan-injected tendons and paratenons showed significantly high molecular HA injections versus placebo (saline) on
lower scores, especially after 75 days. These positive functional outcomes after Zone II flexor tendon repair.
effects were attributed mainly to the anti-inflammatory Three doses were given (one at the time of tenorrhaphy
activity of the agent, which inhibits, both in vitro and in and two at one-week intervals) around the tenorrhaphy
vivo, the leukocyte function (phagocytosis, adherence, and site, and a rehabilitation program was started on the 3rd
mitogen-induced stimulation). Moreover, it was supposed postoperative day. Range of motion (active and passive)
that HA, according to studies in chicken embryos and in and functional outcome (Strickland classification) were
endothelial cell cultures, could inhibit vasculogenesis, assessed. In the short term (3 weeks), no difference
explaining the reduced vascular score at the calcaneal between the two groups was observed; however, at 3
insertion [17]. months and in the long term, a HA. Noteworthy, no
The positive role of HA in tendon healing has been confirmed adverse events (signs of inflammation or tendon rupture)
by the effects of the Seprafilm antiadhesion mem- brane, were seen in any cases.
composed of sodium-hyaluronate plus glucosamine Besides the prevention of adhesions after flexor tendons
HCl-chondroitin sulfate [30]. In rabbits, submitted to ten- repair, the therapeutic efficacy of HA has been evaluated in
don rupture and ensuing surgical repair, after 84 days, the other tendon diseases.
treatment significantly enhanced the maturation rate of the In an open-label randomized study, patients with trigger
tenoblasts, fibrillogenesis, the diameters of the collagen finger were randomly divided into two groups: (1) HA plus
fibrils, and fibrillar density. These findings suggest that corticosteroid and (2) open surgical release [11].

54
54
At 6 and 12 months, 93% of patients of the first group had treated with HA injections and compared to placebo or
complete symptom resolution, compared to 73% submitted active treatments (steroids or physical therapies). The main
to surgery. results of these trials are summarized in Table 1. A superior
In 50 athletic patients with patellar tendinopathy (stage 2 or therapeutic effect was observed in comparison to placebo,
3 according to Blazina’s classification), a mixture of 25 but no significant difference was shown when steroids or
mg HA and 1 mL of 1% lidocaine was injected blindly at physical therapy was used as controls.
the proximal interface between the posterior surface of
the patellar tendon and the infrapatellar fat pad or into the
region of maximum tenderness [12]. A week after the first Conclusions
injection, other injections were done on the patient’s request;
conservative treatments (exercises and instrumental thera- Experimental research and pivotal clinical trials suggest
pies) were also prescribed. An average of 2 injections/case that HA is effective in preventing adhesions after flexor
(range 1–11; total 135) was performed with 12-month (range tendons surgery. Different preparations and procedures
3–49 months) interval between injections. After treatment, are at present under study to define the best option.
54% of patients were rated in excellent conditions Promising results have been also observed in the treat-
(return to previous athletic activities), while 40% in good ment of tendinopathies. In general terms, the positive effect
conditions complained of some degree of limitation. The relies on the anti-inflammatory activity of HA, enhanced cell
open design and the subjective evaluation methods used (no proliferation, and collagen deposition, besides the lubricating
imaging) are important limitations of this study. action on the sliding surface of the tendon. However, it must
Petrella et al. [13] determined the efficacy of periarticular be underlined that in the majority of studies the drug was
HA injections in patients suffering from tennis elbow. not injected inside the degenerated tendon, but nearby and/
In this prospective randomized clinical trial, 165 or into the articular space. It can be speculated that the
subjects with chronic lateral epicondylosis received 2 modifications of the synovial fluid can exert a positive effect
blind injections of 1.2 cc of HA (once a week) into the on the tendon itself, but it cannot be excluded that the
subcutaneous tissue and muscle, from the lateral epicondyle clinical improvement may secondary to the positive action
toward the primary point of pain. Saline solution was injected on osteoarthritis frequently associated.
in the control group (166 patients). Pain, both at rest and
after grip testing, was significantly reduced in the study Conflict of Interests
group compared to controls after 30, 90, and 365 days. The The authors declare no conflict of interests.
treatment was highly satisfactory and resulted in earlier
return to pain-free sport. Authors’ Contribution
The experience of HA use in rotator cuf f tendinopathies All of the authors participated in the work and agree to the
is more consistent [24–29]. Patients with different rotator submission of the paper to the journal.
cuff diseases (full or partial thickness tear, tendinosis) were

55
55
5
6
77
U
Utilizari speciale

859
8
Injectia unica cu Hialuronat in Managementul
Tendinopatiei insertionale Ahiliene in comparatie
cu injectiile cu corticosteroid si tratamentele
conservative non-invazive

Lauren Gorelick1, Ayala Rozano-Gorelick2, Anwar Saab2, Edward Ram3


1Orthopaedic and Hand Surgeon, Assuta Medical Center, Haifa, Israel.
2Clalit Health Services- Bar Ilan Medical Faculty, Zefat.
3Division of General Surgery, Rabin Medical Center- Golda Campus, Sackler School of Medicine,
Tel Aviv University,
Israel.
*Autor corespondent:
Lauren Gorelick, MD
Email: gorelick@netvision.net.il

REZUMAT

Tendonul lui Ahile este cel mai puternic si mai gros tendon din organism si joaca un rol
important în biomecanica extremitatii inferioare. El poate rezista la forte mari in timpul
exercitiilor fizice precum si la torsiunea excesiva si intindere.Tendinopatia Ahiliana
(TA) nu are un mecanism patofiziologic cunoscut si nici o intelegere clara a procesului
patologic. In acest moment se folosesc pentru tratamentul tendinoptiei Ahiliene ,repausul
si exercitii specifice, antiinflamatoarele nesteroidiene, injectiile cu corticosteroizi si
tratamentele chirugicale. Niciuna dintre ele nu au o eficacitate dominanta pentru nicio
perioda de timp. In ultimul timp, injectiile cu hialuronat au fost din ce in ce mai utilizate
pentru tratamentul diferitelor procese degenerative ale articulatiilor si tesuturilor moi.
Autorii au evaluat 56 de pacienti cu TA insertionala tratati intre 2007 si 2012. Toti
pacientii au fost umariti timp de un an. Pacientii au fost divizati in 3 grupuri: primul grup
tratat prin injectarea de corticosteroizi, al doilea grup tratat prin injectie cu hialuroniat
in doza unica si al treilea grup tratat prin repaus, atela, AINS si fizioterapie. Cei mai
multi pacienti care au fost tratati cu o singura injectie cu hialuronat au aratat rezultate
de la bine la excelent pentru o lunga perioada de timp, in comparatie cu injectiile cu
corticosteroizi si pacientii tratati in mod conservator, in functie de scorurile Fadi si VAS.
In concluzie, se pare ca terapia de injectare a hialuronatului este superior fata de terapia
cu corticosteroizi si un tratament conservator non-invaziv pentru TA insertionala.

CUVINTE CHEIE:
tendonul lui Ahile, tendinopatia insertionala Ahiliana, injectare de corticosteroizi, injectia
hialuronatului

60
9
See discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/280556510

Single Hyaluronate Injection in the


Management of Insertional Achilles
Tendinopathy in Comparison to Corticosteroid
Injections and Non-invasive Conservative
Treatments

Article · July 2015

Viscosupplementation with hyaluronic acid is safe and effective in the management of osteoarthritis, but its use in the treatment of
tendon disorders has received less attention.The aim of this review is to summarize the current knowledge on this topic, evaluating
experimental and clinical trials. A search of English-language articles was performed using the key search terms “hyaluronic
acid” or “viscosupplementation” combined with “tendon,” “tendinopathy,“ “adhesions,“ or “gliding,“ independently. In quite all
the experimental studies, performed after surgical procedures for tendon injuries or in the treatment of chronic tendinopathies,
using different hyaluronic acid compounds, positive results (reduced formation of scars and granulation tissue after tendon repair,
less adhesions and gliding resistance, and improved tissue healing)were observed. In a limited number of cases, hyaluronic acid
has been employed in clinical practice. After flexor tendon surgery, a greater total active motion and fingers function, with an
earlier return to work and daily activities, were observed. Similarly, in patients suffering from elbow, patellar, and shoulder tendons
disorders, pain was reduced, and function improved.The positive effect of hyaluronic acid can be attributed to the anti-inflammatory
activity, enhanced cell proliferation, and collagen deposition, besides the lubricating action on the sliding surface of the tendon.

Available from: Lauren Gorelick


Retrieved on: 19 April 2016

61
10
Scholars Bulletin
(A Multidisciplinary Bi-weekly Journal)
An Official Publication of “Scholars Middle East Publishers”,
Dubai, United Arab Emirates
Website: http://scholarsbulletin.com/

Single Hyaluronate Injection in the Management of


Insertional Achilles
Tendinopathy in Comparison to Corticosteroid
Injections and Non-invasive
Conservative Treatments
Lauren Gorelick1, Ayala Rozano-Gorelick2, Anwar Saab2, Edward Ram3
1Orthopaedic and Hand Surgeon, Assuta Medical Center, Haifa, Israel.
2Clalit Health Services- Bar Ilan Medical Faculty, Zefat.
3Division of General Surgery, Rabin Medical Center- Golda Campus, Sackler School of Medicine, Tel Aviv University, Israel.

*Corresponding Author:
Lauren Gorelick, MD
Email: gorelick@netvision.net.il

Abstract:
The Achilles tendon is the strongest and thickest tendon in the body and plays an important role in the biomechanics of the lower
extremity. It can withstand high forces during sporting exercises and excessive torsion and
stretching. Achilles Tendinopathy (AT) has no pathophysiological theory and no clear understanding of the pathological process.
Rest and specific exercises, NSAIDs, steroid injections and surgical treatments are currently used to treat AT. Not one of them
has a dominant efficacy for any length of time. Lately, hyaluronate injections have increasingly been used for the treatment of
different degenerative processes of joints and soft tissues. The authors reviewed 56 patients with insertional AT treated between
2007 and 2012. All patients were followed up for one year. Patients were divided into three groups: the first group treated by
the corticosteroid injection, the second group treated by single hyaluronate injection and the third group treated by rest, splint,
NSAIDs, and physiotherapy. Most patients that treated by a single hyaluronate injection showed good to excellent results for a long
time in comparison with corticosteroids injections and the conservatively treated patients, according to FADI and VAS scores. In
conclusion, it seems that hyaluronate injection therapy is superior to steroid therapy and non-invasive conservative treatment for
insertional AT.

Keywords:
Achilles tendon, Achilles insertional tendinopathy, corticosteroid injection, hyaluronate injection

Introduction insertional tendinitis, paratenonitis, retrocalcaneal


bursitis, and subcutaneous Achilles tendon bursitis. It
Achilles tendinopathy (AT) is becoming a more common occurs in combinations of the above in most cases. The wide
complaint due to increased medical awareness and spectrum of nonsurgical treatment modalities (the rest,
greater physical activities of the general population. It activity modification, specific exercises, use of a brace,
can be the reason for substantial pain and loss of function NSAIDs, steroid injections, shockwave therapy) indicate
of the affected limb for a long time. AT is considered an that not one of them is strongly effective. AT, therefore,
overload injury at the point of attachment at the calcaneus is difficult to manage and up to 29% of AT patients may
and two to six cm. proximally of that point at the region of require surgery[2]. Specific exercises show the most
maximum torsion. Chronic stress can lead to a defective evidence of effectiveness in the treatment of AT[3].
arrangement of collagen fibers in the AT and result in Injection of corticosteroids have strong potential for
pain and limited function. At this time, there is no pain relief for a short period but is problematic because
pathophysiologic theory with a clear understanding of of potentially severe adverse effects[4]. Rupture of the
the pathological process of this entity. There first appears Achilles’ tendon following intratendinous injections has
the evidence of the existence of the TNF-α system in the been reported. There is evidence supporting the use
human AT. Findings are confirmed by mRNA and protein of hyaluronate injections in patients with ankle sprain,
levels as well as biochemically. The TNF-α system was adhesive capsulitis of the shoulder, patients after flexor
in principle confined to the tenocytes. The connection tendon injury, trochanteric bursitis, and rotator cuff
between tenocyte morphology and the expression disorder, although the mechanism of the effect has
pattern of TNF-α, TNFR1, and TNFR2 suggests that the not yet been clarified[5-10]. There is some evidence
TNF-α system may be involved in tenocyte activation in in experimental laboratory studies that hyaluronate
Achilles tendinosis[1]. AT disorders are multifactorial increases proliferation of vascular endothelial growth
and include tendinosis, tendinosis with partial rupture, factor (VEGF) and collagen type 4 during the six weeks

62
11
after injection, and decrease the amount of adhesion Statistical Analysis
tissue[11]. Hyaluronic acid dose-dependently inhibited Statistical analysis was performed using the SPSS statistical
cell proliferation and decreased the expression level of program. ANOVA with post-hoc analysis was used.
mRNA for adhesion-related procollagens and cytokines Between-group differences in the primary outcome analyzed
[6, 9]. In another study, Hylan G-F 20 has shown a based on the intention-to-treat principle. Between-group
promising curative effect on the tendon and paratenon differences in continuous outcome measure analyzed with
in an experimental study in rats, and Hylan-injected repeated measures for general linear models. A difference at
tendons and paratenons demonstrated significant the 0.05 level is termed significant.
improvement in comparison to saline injection especially
after 75 days[12]. A single hyaluronate injection RESULTS
resulted in similar improvements in pain in each of FADI Score
the four enthesopathies-lateral epicondylitis, patellar
tendinopathy, insertional AT, and plantar fasciitis[13]. The FADI score evaluated for the entire cohort. The FADI
No published studies have been found that have followed score increased from 26.3 to 67. 91 after six months and
patients who were administered hyaluronate in AT in 75.179 after a year. The pre-treatment average score was
comparison to other kinds of conservative treatments. similar in the three groups. A significant difference was found
Corticosteroid injection beneficial in the short term for between the groups (ANOVA, p < 0.05). The average score
the treatment of tendinopathy but may be worse than of patients treated by corticosteroid injection was 55.39
other treatments in the intermediate and long term[14]. as compared with 64.77 for the hyaluronate injected
group and 48.96 for the conservative treatment group.
The use of hyaluronate injections for different kinds of There was a time interaction with inter-group difference
tendinopathy has been routine in this author’s practice for in time. In the corticosteroid group, the average score
the last seven years. This study evaluates of three groups significantly improved from 26.95 to 73.95 at six months but
of patients treated with Hyaluronate and compared with decreased at 12 months (65.26). The time-related behavior
corticosteroid injection and non-invasive therapy. was different in the hyaluronate group with a pre-injection
average of 24.90 increasing to 72.85 at six months, and to
METHODS 96.55 at 12 months. The conservative treatment group had
Patients a time-related behavior similar to the corticosteroid group at
the one-year follow-up (Figure 1).
This study evaluates of a consecutive series of 56 patients
treated for Achilles insertional tendinopathy. M/W VAS Scale
ratio was 21/35. The average age was 54 (range 20-85 VAS scale declined over time for the entire cohort from
years). 17 patients treated by rest, splint, NSAIDs, and 10 to 2.68 after a year. There was a significant inter-
physiotherapy. 19 patients treated with corticosteroid group difference of the VAS scores with average VAS of
injection(betamethasone dipropionate five mg. and the conservative treatment group higher (5.46) than the
betamethasone sodium phosphate two mg.). 20 patients corticosteroids treated group (4.74) and the hyaluronate
of the third group treated with a standard single group (3.88). There is a time-dependent intergroup
sodium hyaluronate 2% (40 mg/2.0 ml) injection (Ostenil difference with a steep decline from pre-treatment to the six
Tendon, TRB Chemedica International S.A., 1211 Geneva, weeks mark, and an insignificant decrease in conservatively
Switzerland). treated group at later time points of three months, six
months and one year. There is an insignificant increase of
Achilles tendinopathy diagnosis. the VAS scale in the corticosteroids treated group at three
Insertional AT is characterized by “deep pain at the tendon- months and a significant increase at six and twelve months
bone junction of the posterior calcaneus point over a bony (Figure 2). The time dependence of the hyaluronate groups
prominence or spurs located at the posterior superior aspect is different with a continuing significant drop during the one
of the calcaneus”[15]. High-resolution sonography made year of follow-up.
of all patients for diagnosis of AT as a relatively reliable
diagnostic method in Achillodynia [16]. DISCUSSION
The mechanism of action of hyaluronate in Achilles
Inclusion Criteria. tendinopathy appears to be complicated. The basic pathology
All patients were newly diagnosed with insertional AT without underlying the symptoms is not yet fully understood. The
any previous treatment before being included in the study. disorder is apparently complex as a spectrum of diseases
ranging from inflammation of the paratendinous tissue to
Exclusion Criteria. structural degeneration of the tendon. Hyaluronate plays
•All patients treated in the past and having received any a significant role during wound healing and regeneration.
intervention due to insertional AT. It has anti-inflammatory effects as well as the possibility
•Patients who operated on in the same area. that its presence will restore normal tissue function. It
•Patients that had a systemic or rheumatological disease, enhances muscle progenitor cell recruitment and inhibits
tarsal tunnel syndrome, moderate to severe osteoarthritis premature myotube fusion, implicating a role for these
of the ankle and foot, lumbar stenosis or disc disease, and glycosaminoglycans in functional repair[17].
instability of the ankle.
•Patients who were unable to understand the questionnaires. The current study appears to demonstrate that there is
an ameliorative effect of hyaluronate injections in AT. The
pain relief appears to be significant with a large effect
Treatment Effect Assessment size for the hyaluronate group. The effect of steroids
Patients were assessed using a back-translated FADI (Foot appears to be short lived, and this concurs with the
and Ankle Disability Index) version (Hebrew, Russian and findings comparing effects of steroids and hyaluronic
Arabic). The questionnaire was self-filled by the patients, acid in knee osteoarthritis[18]. Corticosteroid injection
and a research assistant verified questionnaire filling. Also, may be worse than other treatments in the intermediate
a standardized VAS (Visual Analogue Scale) score was filled and long term. There is no consensus as to whether
out in answer to the question:”What number would you give local steroid injections have a therapeutic role in the
your pain right now?” treatment of AT. However, they may incur a risk of tendon
damage[19].

63
12
Figure 1: The FADI score is similarly affected by injection therapy in the hyaluronate and steroid at six months, while the amelio-
rative effect of steroids appears to be short-lived. No difference between steroid and non-invasive treated groups at long time
follow-up.

Figure- 2: The VAS score response to injection therapy is similar to that of the FADI score but in decreased direction. Note the
similar effects of steroids and non-invasive therapy at twelve months of the follow-up.

Corticosteroid injection may be worse than other CONCLUSION


treatments in the intermediate and long term. There is The current study appears to indicate that single
no consensus as to whether local steroid injections have hyaluronate injection is efficacious for the treatment of
a therapeutic role in the treatment of AT. However, they insertional AT. The effect size is larger with hyaluronate
may incur a risk of tendon damage [19]. injections and lasts longer. Limitations of the current study
were the small size of the groups and the fact that the patient
The current study appears to indicate that hyaluronate were not randomized. A further prospective randomized
injection is an acceptable alternative to steroid injections clinical trial should be done prior to making a clear-cut
for the treatment of AT. This alternative treatment recommendation that hyaluronate injections are superior to
may be more useful in a diabetic patient without any other conservative modalities and may provide an alternative
complications and side effects of steroids[20] for clinicians.

64
13
Managementul Cotului Tenismenului cu
injectiile periarticulare cu hialuronat
Robert J Petrella1*, Anthony Cogliano2, Joseph Decaria3, Naem Mohamed4, Robert Lee5

REZUMAT

OBIECTIV:
Sa determine eficacitatea si siguranta injectiilor periarticulare cu Acid Hialuronic in epicondilita
laterala (cotul tenismenului).
Design: studiu clinic prospectiv randomizat, in ingrijirea primara din medicina sportiva.
Pacienti: trei sute treizeci si unu atleti consecutivi si competitivi ai sportului cu racheta cu
epicondilita laterala cronica (>3 luni) li s-au administrat 2 injectii (prima injectie la baseline)
in tesutul subcutanat si muschi la 1 cm distanta de epicondilul lateral spre punctul de
durere primar, folosind tehnica bidimensionala de tatonare. Cea de-a doua injectie a fost
administrata o saptamana mai tarziu.

MASURAREA OBIECTIVELOR:
Evaluarile au fost facute la baseline, zilele 7, 14, 30, 90 si 356. Masuratorile de eficacitate
au inclus scara vizuala analogica (VAS) de durere in repaus (0-100 mm) si apoi o evaluare
a rezistentei de prindere (0-100 mm). Puterea de prindere a fost determinata folosind un
dinamometru Jamar cu brat hidraulic. Alte evaluari au inclus evaluare globala a leziunii
cotului (scala categorica de puncte; 1= fara dizabilitati, 5 = dizabilitatea maxima), evaluarea
functiei/activitatii normale a pacientilor (scala categorica de 5 puncte), evaluarea satisfactiei
pacientilor/medicilor (scala categorica de 10 puncte), timpul pana la reintoarcere la sport fara
durere si fara dizabilitati si efecte adverse conform definitiei WHO. Diferenta dintre grupuri a
fost determinata folosind ANOVA – intentie de tratament.

REZULTATE:
Varsta medie a populatiei de studiu a fost 49 de ani (+/- 12 ani). 165 de pacienti au fost
randomizati pe Acid Hialuronic si 166 au fost randomizati pe grupul de control. Schimbarea
in scala VAS pentru durere a fost -6.7 (=/- 2.0) pentru AH vs -1.3 (+/- 1.5) pentru grupul
control (p<0.001). Scorul VAS post handgrip a fost de 7.8 (+/-1.3) vs +0.3 (± 2.0) (p < 0.001)
care corespunde unei imbunatatiri semnificative a fortei de strangere de 2.6 kg in grupul
cu Acid Hialuronic vs grupul control (p < 0.01). De asemenea au fost inregistrate imbunatiri
semnificative ale evaluarii globale a leziunii cotului (p < 0.02),a functionalitatii si activitatii
normale (p < 0.05) si a evaluarii satisfactiei pacientilor/medicilor (p<0.05) favorizand grupul
cu AH. Timpul de reintoarecere la sport fara durere si fara dizabilitate a fost de 18 (+/- 11) zile
in grupul cu AH si nu a fost atins in grupul control. Modificarile de VAS au fost mentinute in
grupul cu AH la fiecare vizita de urmarire in timp ce celor din grupul control indicele a scazut
semnificativ de la baseline. Evaluarile satisfactiei pacientului si medicului au continuat sa fie
in favoarea grupului cu AH, la vizitele de urmarire ulterioare.

CONCLUZIE:
Tratamentul periarticular cu AH pentru cotul tenismenului a fost semnificativ mai bun decat
in grupul control in ameliorarea durerii in repaus si dupa testarea aderentei maxime. In plus,
tratamentul cu AH a fost considerat foarte satisfacator de catre pacienti si medici si a dus la
o reintoarcere mai buna la sportul fara durere, comparativ cu grupul control.

65
14
Petrella et al. Sports Medicine, Arthroscopy, Rehabilitation, Therapy & Technology
2010, 2:4 http://www.smarttjournal.com/content/2/1/4

RESEARC H Open
Access
Management of Tennis Elbow with sodium
hyaluronate periarticular injections
Robert J Petrella1*, Anthony Cogliano2, Joseph Decaria3, Naem Mohamed4, Robert Lee5

Abstract
Objectives: To determine the efficacy and safety of peri-articular hyaluronic acid injections in chronic lateral epicondylosis
(tennis elbow).
Design: Prospective randomized clinical trial in primary care sport medicine.
Patients: Three hundred and thirty one consecutive competitive racquette sport athletes with chronic (>3 months) lateral
epicondylosis were administered 2 injections (first injection at baseline) into the subcutaneous tissue and muscle 1 cm. from
the lateral epicondyle toward the primary point of pain using a two-dimensional fanning technique. A second injection was
administered 1 week later.
Outcomes measures: Assessments were done at baseline, days 7, 14, 30, 90 and 356. Efficacy measures included
patient’s visual analogue scale (VAS) of pain at rest (0-100 mm) and following assessment of grip strength (0-100 mm). Grip strength
was determined using a jamar hydraulic hand dynamometer. Other assessments included patients’ global assessment of elbow
injury (5 point categorical scale; 1 = no disability, 5 = maximal disability), patients’ assessment of normal function/activity (5 point
categorical scale), patients/physician satisfaction assessment (10 point categorical scale), time to return to pain-free and disability-
free sport and adverse events as per WHO definition. Differences between groups were determined using an intent-to-treat ANOVA.
Results: Average age of the study population was 49 years (± 12 years). One hundred and sixty-five patients were randomized to
the HA and 166 were randomized to the control groups. The change in VAS pain was -6.7 (± 2.0) for HA vs -1.3 (± 1.5) for control
(p < 0.001). The VAS post handgrip was -7.8 (± 1.3) vs +0.3 (± 2.0) (p < 0.001) which corresponded to a significant improvement in
grip of 2.6 kg in the HA vs control groups (p < 0.01).
Statistically significant improvement in patients’ global assessment of elbow injury (p < 0.02), patients’ assessment of normal
function/activity (p < 0.05) and patients/physician satisfaction assessment (p < 0.05) were also observed favoring the HA group.
Time to return to pain-free and disability-free sport was 18 (± 11) days in the HA group but was not achieved in the control group.
VAS changes were maintained in the HA group at each followup while those in the control significantly declined from baseline.
Assessment of patient and physician satisfaction continued to favor the HA group at subsequent followup.
Conclusion: Peri-articular HA treatment for tennis elbow was significantly better than control in improving pain at rest and after
maximal grip testing. Further, HA treatment was highly satisfactory by patients and physicians and resulted in better return to pain
free sport compared to control.

CONCLUSION efficacy in terms of pain and function with low incidence


Chronic tennis elbow or lateral epicondylosis produces of side effect and treatment of chronic tennis elbow.
symptoms of pain and functional disability. Typical Tennis elbow (lateral epicondylosis), a common cause of
treatments include RICE for acute exacerbations as well chronic elbow pain and wrist extensor dysfunction in adults,
as oral or topical NSAIDs, bracing and physical therapy. affects 1-3% of the general population each year[2]. Tennis
However, there is no consensus on treatment while players have been reported to account for 5- 8% of all cases,
efficacy of existing treatments is poor. Intra-articular although between 40-50% of all tennis players will be afflicted
hyaluronic acid (HA) has shown efficacy equivalent with the condition at some time[3]. Lateral epicondylosis
to NSAID in the treatment of osteoarthritis while is most prevalent in the fourth decade of life and the
it’s periarticular efficacy and safety have recently been syndrome is rarely seen in individuals under the age of 30.
reported for soft tissue use in acute ankle sprain. Hence, Localized tenderness around the lateral epicondyle generally
many patients, particularly those who require more rapid characterizes the condition, and pain can be reproduced by
improvement to return to sport or work activity, or those resisted extension of the wrist or the middle finger with the
in whom previous therapies have not achieved expected elbow in a straight position. The injury results in elbow pain
results, would benefit from a more rapid alleviation of that usually heals spontaneously, although it can become
symptoms, while still achieving the longer term benefits of a source of chronic pain and morbidity if left untreated. On
hyaluronic acid that have been reported in other soft tissue average, a typical episode of lateral epicondylosis lasts 6-24
indications. months[4]. The primary lesion and epicondylosis consists
Previous studies regarding treatment of chronic tennis of micro ruptures and result in inflammatory granulation
elbow have shown lack of consensus as well as variable tissue in the tendinous portion of the origins of the forearm
efficacy and high incidence of adverse effects[1]. musculature just distal to the epicondyle of the humerus. The
Hyaluronic acid has been used in soft tissue application lesion is found primarily in the extensor carpi radialis brevis
for acute ankle sprain with high degree of efficacy and (ECRB) origin, with less frequent involvement of the extensor
very limited side effect. Hence, given the biocompatibility carpi radialis longus (ECRL) and the anterior portion of the
of HA in treatment of acute ankle sprain we may show extensor digitorum communis[5]. Nirschl [6] maintains that

66
15
angiofibroblastic hyperplasia, resulting from avascular phosphate buffered saline contained in a prefilled syringe;
compromise and subsequent micro tears in the origin of 1.2 cc) [165 patients] versus 166 patients administered 1.2
the ECRB, is the basic cause of chronic lateral epicondy- cc saline placebo. Treatment course was randomized and
losis. While the muscle fibres have adequate blood sup- consisted of 2 injections (1 at baseline and a second at 7
ply and good healing potential, the tendon fibres attached days). Injections were administered using blinded syringes
to the periosteum are relatively avascular and are prone to affixed to a 27- gauge, 1-inch needle. Skin was prepped
ischemic stress and thus slow to heal [7]. Recent studies of using betadine 1%. Injections were delivered by the study
chronic tennis elbow have not found any significant evidence physician using a standard approach along the lateral
of inflammatory processes and the term epicondylosis epicondyle with the affected arm flexed and resting on a firm
has been suggested as a more appropriate term than surface. Injections were be administered into the soft tissue
epicondylitis [8]. 1 cm from the lateral epicondyle at the point of greatest
There is currently no consensus on the optimum pain in two planes using a fanning technique [13] (Fig. 1)
treatment, but numerous options are available. The whereby contents were injected on withdrawl of the needle
best available scientific evidence suggests that topical from the point of maximal tenderness in a single puncture.
and possibly oral non-steroidal anti-inflammatory All patients had radiograph to exclude other pathologies
drugs may be the most useful for short-term pain at the discretion of the study physician (ie to exclude
relief. Corticosteroid injections may be beneficial as a fracture). None took any other treatment modalities during
temporary measure but carry the risk of possible the observation period or 48 hours prior to assessments.
adverse affects [9,10]. Symptoms usually start with an ache Assessments included general demographics, comorbidities
in the extensor aspect of the forearm with certain movements and previous treatments.
that become localized in the lateral epicondyle. Visible Patients rated pain on a 10 cm VAS, with 0 representing
swelling is not a common feature in lateral epicondylosis no pain and 10 representing maximal pain. Patients’
and, if present, should suggest some other pathology. glo-bal assessment of elbow injury (5 point categorical
Elbow radiographs are usually normal and seldom helpful. scale; 1 = no disability, 5 = maximal disability),
Ectopic calcification of the lateral epicondyle appears in patients’ assessment of normal function/activity (5 point
approximately 25% of the cases but its presence does categori-cal scale; 1 = no change in function/activity,
not appear to alter prognosis. Differential diagnosis should 5 = maxi-mal change in normal function/activity) and
include referred pain from the cervical spine, rheumatoid physician’s global assessment of elbow injury (5 point
arthritis, radial tunnel syndrome and compression of the categorical scale; 1 = no impact of injury on function,
posterior interosseous nerve [11]. 5 = maximal impact of injury on function) were also
Hyaluronic acid is a natural occurring biological collected. Global assessments have not been validated
substance, which has been shown to have a positive but have been used previously by our and other groups to
effect in inter-articular administration for osteoarthritis link the findings to implementation into routine practice.
[12] as well as more recently periarticular use in acute Time to return to pain and disability-free sport and adverse
ankle sprain[13]. These studies have shown improved pain events were determined from review of a patient diary. After
and functional range of the arthritic as well as the soft tissue enroll- ment, patients were randomized (1:1) to one of two
injury with high degree of patient satisfaction and few adverse treatments using a computer-generated randomization
events. Previous studies of topical NSAID and botulinum schedule: HA or placebo.
toxin for the treatment of epilateral epicondylitis [14,15] have Follow-up examinations were completed at Day 14 (± 2
shown improved pain at 1-3 months post injection. However, days), Day 30 (± 2 days), Day 90 (± 2 days) and at Day 356
this was associated with rash, mild gastric upset [14] and (± 7 days). Patients will assess pain on a VAS at rest and
digit paresis and weakness of finger extension [15]. Further, after assessment of grip strength. Grip strength will be
efficacy was not assessed long term. Hence, given the lack determined using a jamar hydraulic hand dynamometer
of consensus, the high rate of adverse effects and lack of (Sammons Preston, Bolingbrook, Illinois). Assessment
long term followup with current options, use of HA, which is will be conducted with the patient’s elbow fully extended,
relatively free of these adverse effects and has been used in shoulder in neutral position and the dynamometer’s handle
ankle sprain with long term efficacy[16], could prove to be an in the middle position.
option for patients with chronic tennis elbow. Patients will perform three grip tests on the affected arm with
Hyaluronic acid is an unbranched, high molecular weight a mean score calculated and used for analysis.
polysaccharide distributed throughout the body, especially During the study, including the follow-up period, the
as a major component of synovial fluid cartilage and patients received usual care including RICE (rest, ice,
surrounding structures of arthroidial joints. compression and elevation). Use of any analgesics was
The primary role of the HA in these tissues is to prohibited and all concomitant medication use was recorded
maintain their viscoelastic structural and functional in the patient’s diary. Specifically, no NSAID, corticosteroid
characteristics. Given the long term efficacy and safety or topical analgesics were allowed during the study. ASA at
of periarticular HA in acute ankle sprain, it would suggest the dose of 325 mg and less for cardiovascular prophylaxis
similar protective effects of HA in chronic lateral was allowed.
epicondylosis. Patients were assessed for pain on a VAS at rest and after
We are unaware of any other published studies that have assessment of grip strength. Patients’ global assessment of
prospectively followed patients who were adminis- tered elbow injury (5 point categorical scale), patients’ assessment
HA in lateral epicondylosis for treatment of tennis elbow in of normal function/activity (5 point categorical scale), and
clinical practice. physician’s global assessment of elbow injury (5 point
Hence, we hypothesized that HA administered in the soft categorical scale) was performed. Also, patients/physician
tissue of lateral epicondylosis for chronic tennis elbow satisfaction assessment (10 point categorical scale; 1 = no
will be well-tolerated with few adverse events and result satisfaction with the procedure, 10 = very high satisfaction
in improved clinical pain and function outcomes from with the procedure) and review of a patient diary for adverse
baseline to long term followup compared to patients events and return to pain and disability-free sport was
administered placebo. performed.
The primary outcome measures were an improvement
Methodology on the VAS-pain at rest in the affected elbow and
We collected data on 331 consecutive patients administered VAS-pain immediately following grip strength testing.
HA (a clear solution of sterile 1% sodium hyaluronate in a Secondary outcome measures included patients’ global
assessment of elbow injury (5 point categorical scale),

67
16
patients’ assessment of normal function/activity (5 treat (ITT) population. The ITT population was patients who
point categorical scale), physician’s global assessment received at least 1 injection of double-blind study therapy.
of elbow injury (5 point categorical scale), patients/ For the efficacy analysis, all patients were counted in the
physician satisfaction assessment (10 point categorical group they were randomized to irrespective of their actual
scale), time to return to pain and disability-free sport, treatment assignment.
concomitant medication use and adverse events.
The patients were recruited and screened in 4 sites Statistical Power and Sample Size
(Canadian Center for Activity and Aging; University For the primary analysis of the change from baseline in elbow
of Western Ontario-Fowler-Kennedy Sport Medicine pain we estimated a sample size of 160 per group using a
Center; U Waterloo Sport Medicine Center; U Guelph 40% difference in resting VAS scores at Day 30 between
Sport Medicine Center). Patients eligible for the study were treatment and control, a potential dropout rate of 25% and
18 or older, with clinically or radiographic diagnosis of 95% confidence assuming a standard deviation of <10 mm
tennis elbow, and who were newly referred to the medical of the mean deviation, an a of 5%, and a b level of 10%.
outpatient clinics at the author’s institution which are primary
sport medicine referral centers serving a population of 1.5
million patients. Inclusion criteria were pain at the lateral side Results
of the elbow that had persisted more than 3 months and Both groups (HA = 165 vs placebo = 166) were similar for age
pain at the lateral epicondyle during resisted dorsiflexion of (49 ± 15 vs 47 ± 11) and gender (55 vs 53% male) distribution
the wrist with the elbow in full extension. Exclusion criteria (Table 2). There was also no difference among groups in the
were previous local injection treatments (ie. corticosteroid duration of their symptoms (26 vs 33 months). There were no
injections or acupuncture), nerve entrapment or systemic serious adverse events reported throughout the study. Three
neuromuscular disorders. patients (1.8%) in the HA and 5 patients (4%) in the control
The diagram below provides a description of the over-all reported pain during injection. No other adverse events were
study design (Table 1). reported. No subjects withdrew from the study during the
The analyses of the efficacy endpoints used an intent-to- treatment phase.

Figure 1 Localization and administration of HA.

6817
Mean baseline rest VAS was similar (8.5 ± 1.1 cm and 8.4 Discussion
± 1.6 cm) for HA and control respectively. VAS pain at There is currently no consensus in the management of
rest and after grip testing was significantly better in the chronic tennis elbow. Several, reviews have included var-
HA vs control (Table 2) at D 30. This was associated with ious therapies targeting local or systemic interventions.
significantly greater grip strength, patient global satisfaction Further, these therapies have not shown ability to shorten
and assessment of normal elbow function in the HA group the disability period nor have they shown long term benefit.
vs control (Table 2). Physician global assessment of elbow In our study, patients who received HA for lateral
injury was significantly better for the HA vs the control (Table epicondylosis (tennis elbow) had significantly greater
2). These differences persisted at each follow up assessment improvement in VAS pain at rest and after grip testing
(90 and 356 days). Time to return to pain-free and disability- than control that persisted to 356 days followup. The
free sport was 18 (± 11) days in the HA group (in 147 treatment was highly satisfactory to patients and
patients; 89% response rate) but was not achieved in any of physicians and was associated with very few minor
the control group patients. and transient adverse effects. Given the less than
optimal treatment options for tennis elbow and given
the associated chronic morbidity associated with this
condition, periarticular injection with HA may provide
an alternative for clinicians and their patients.

69
18
Eficacitatea si siguranta pe termen lung ale
Acidului Hialuronic periarticular in entorsa acuta
de glezna
Michael J. Petrella, PhD; Anthony Cogliano, MD; Robert J. Petrella, MD, PhD

REZUMAT

Obiectivele acestui studiu au fost sa determine eficacitatea si siguranta pe termen lung


a injectiilor cu Acid Hialuronic periarticular in entorsa laterala acuta de glezna. Studiu
prospectiv randomizat controlat placebo in medicina primara sportiva si practica de
urgenta au inclus 158 de atleti competitivi care sufereau de o entorsa laterala acuta
de glezna de Grad 1 sau grad 2 si care au fost randomizati in primele 48 de ore de la
accidentare. Pacientii au fost randomizati la baseline pe injectie periarticulara cu Acid
Hialuronic + terapia standard de ingrijire (odihna,gheata,ridicare si compresie) (RICE) sau
injectie placebo (PL) + terapie standard de ingrijire (RICE) la evaluarea baseline si in ziua
a 4 dupa accidentare. Urmarirea a fost efectuata la 30, 90, si 712 de zile dupa tratament.
Evaluarile la baseline si zilele 4,8, 30 , 90 si 712 au inclus scala vizuala analoga de durere
(VAS)(0-10 cm) a greutatii si a mersului 20 m, evaluarea globala a accidentarii gleznei
(scala categorica de 5 puncte), satisfactia pacientului cu tratament (scala categorica de
5 puncte) timpul de reintoarcere la sportul fara durere si dizabilitate, entorsa recurenta
de glezna, numarul total de zile de absenta din activitatea sportiva primara, si efecte
adverse (EA). Timpul pana la interventie a fost de 39 +/- 4 ore fara diferente intre grupuri.
O reducere semnificativa a VAS pe greutate si mers a fost observata la toate vizitele
de urmarire la grupul cu AH comparat cu placebo (p<0.001). Timpul de reintoarcere
la sport fara durere si fara dizabilitate a fost de 11 (+/-8) versus 17 (+/-) zile pentru
AH si placebo, respective (p<0.05). La 24 de luni, in grupul placebo vs AH au fost 2
versus 0 fracturi de membru inferior, 16 versus 7 a-2-a entorsa de glezna,(p<0.05), 3
versus 1 a 3-a entorsa de glezna, I un numar semnificativ mai mare zile de lipsa din
activitatea sportiva (41 versus 21; p <0.002). Satisfactia pacientilor a fost semnificativ
mai mare la pacientii din grupul cu AH versus placebo la toate evaluarile de urmarire.
Niciun eveniment advers grav nu a fost inregistrat in decursul urmaririi. Tratamentul
periarticular cu Acid Hialuronic in entorsa acuta de glezna a fost inalt satisfacatoare pe
termen scurt si lung versus placebo. Aceasta asociata cu trecerea durerii, intoarecerea
mai rapida la sport, mai putine entorse recurente de glezna, mai putine zile de lipsa de
la sport cu putine efecte advserse in 24 de luni.

CUVINTE CHEIE
entorsa de glezna, Acid Hialuronic, eficacitatea si siguranta pe termen lung

19
70
Long-Term Efficacy and Safety of Periarticular
Hyaluronic Acid in Acute Ankle Sprain
Michael J. Petrella, PhD; Anthony Cogliano, MD; Robert J. Petrella, MD, PhD

Abstract
The objectives of this study were to determine the long-term efficacy and safety of periarticular hyaluronic acid injections in acute
lateral ankle sprain. A randomized controlled prospective trial in a primary sport medicine and emergency practice involved 158
competitive athletes who suffered an acute Grade 1 or 2 lateral ankle sprain, and who were randomized within 48 hours of injury.
Patients were randomized at baseline to periarticular injection with hyaluronic acid (HA) + standard of care (rest, ice, elevation,
and compression [RICE]) or placebo injection (PL) + standard of care (RICE) treatment at baseline assessment and Day 4 post
injury. Follow-up was at 30, 90, and 712 days post treatment. Assessments at baseline and Days 4, 8, 30, 90, and 712 included
visual analog scale (VAS) (0-10 cm) pain on weight bearing and walking 20 m, patient global assessment of ankle injury (5-point
categorical scale), patient satisfaction with treatment (5-point categorical scale), time to return to pain-free and disability-free sport,
recurrent ankle sprain, total number of days missing from primary sport activity, and adverse events (AEs). Time to intervention
was 39 ± 4 hours with no difference between groups. A significant reduction in VAS pain on both weight bearing and walking was
observed at all follow-up assessments for HA compared with PL (P < 0.001). Time to pain-free and disability-free return to sport was
11 (± 8) versus 17 (± 8) days for HA and PL, respectively (P < 0.05). At 24 months, in the PL versus HA group, there were 2 versus 0
lower limb fractures, 16 versus 7 second ankle sprains (P < 0.05), 3 versus 1 third ankle sprains, and a significantly greater number
of days missing primary sport activity (41 versus 21; P < 0.002). Significantly greater patient satisfaction was also observed for HA
versus PL at all follow-up assessments. No serious AEs were recorded throughout follow-up. Periarticular HA treatment for acute
ankle sprain was highly satisfactory in the short and long term versus PL. This was associated with reduced pain, more rapid return
to sport, fewer recurrent ankle sprains, fewer missed days from sport, with few associated AEs to 24 months.
Keywords: ankle sprain; hyaluronic acid; long-term efficacy and safety

Michael J. Petrella, PhD1 Anthony Introduction


Cogliano, MD1 Ankle sprains are among the most common of all sports injuries, with approximately
Robert J. Petrella, MD, PhD1 2 million people per year seeking medical treatment.1–3 An epidemiological study of
professional, competitive, and recreational athletes found a prevalence of ankle sprain
1Faculties of Medicine and Health as high as 73%4 or a crude incidence rate of at least 52.7 per 10 000.5 Ankle sprains
Sciences, University of Western are also the cause of significant morbidity in the longer term. A recent review by van Rijn
Ontario, London, Canada et al6 found up to 33% of patients still experienced pain after 1 year, while only 36% to
85% reported full recovery up to 3 years later. There is also a wide variation in subjective
instability, of up to 33% to 53%. Hence, the impact of ankle sprain is of considerable
Correspondence: concern to long-term function and performance of athletes beyond the acute event.
Robert J. Petrella, MD, PhD, Dept. Compounding the impact of ankle sprain is the absence of a standard treatment
of Family and Physical approach that directly targets the injury. The American Academy of Orthopaedic
Medicine & Rehabilitation, Surgeons (AAOS) does recommend an initial rehabilitation program (up to 3 weeks)
Schulich School of Medicine, with nonsteroidal anti-inflammatory drugs (NSAIDs), rest, ice, compression, and
Lawson Health Research Institute, elevation (RICE), as well as protected weight bearing, early mobilization, and
University of Western Ontario, 801 isometric exercise.7 Further, while NSAIDs may effectively reduce the pain and swelling
Commissioner’s Road East, associated with acute ankle sprain,8–12 this may not alter the clinical course of ankle
London, Ontario, Canada N6C 5J1 sprain regarding return to sport and may also cause significant adverse events (AEs)
Tel: 519-685-4292 (ext 42633) including gastrointestinal intolerance and serious events such as ulcers and bleeding.
Fax: 519-685-4071 The conservative treatment approach recommended by the AAOS may limit disability to
E-mail: petrella@uwo.ca an average of 8 days for a Grade 1 and 15 days for a Grade 2.3–4 However, the longer-
term efficacy in terms of recurrent sprains and return to sport is limited. In one study of
ankle sprain, pain and dysfunction was found to persist 6 to 18 months (average, 12.8 months) after initial ankle sprain in 73% of
patients, with 40% reporting inability to walk 1 mile and 11% continuing to use medications for ankle symptoms,14 while in a long-
term follow-up study, nearly 40% of patients reported residual long-term symptoms and dysfunction 6.5 years after initial ankle
sprain.15 Since recurrence rates are high ranging from 3% to 34% of the patients for periods ranging from 2 weeks to 96 months
post injury,6 the current approach to ankle sprain could benefit from new modalities and interventions.
Some approaches to longer-term prevention of recurrent sprains have focused on prophylactic external bracing and rehabilitative
training but have shown variable results.13 Further, prophylactic approaches may impact performance and even increase risk
of other injury. What is missing is a targeted approach to ankle sprain that would provide superior efficacy, low risk of AEs, and
improved rates of recurrence.
Hyaluronic acid is a naturally occurring biological substance which has been shown to have a positive clinical impact in
intra-articular as well as intradermal indications.16 As hyaluronic acid relieves pain and stiffness related to its rheologic
modification of intra-articular matrix as well as “filling” intradermal space, it may be hypothesized to have a similar effect
on the extra-articular complex including ligamentous structures affected by acute ankle sprain, including structural and
inflammatory interruption. Further, hyaluronic acid injected locally at the site of injury would not precipitate systemic risk of AEs.
We previously reported the short-term efficacy and safety of periarticular hyaluronic acid in acute ankle sprain versus placebo.17
Periarticular hyaluronic acid was not only superior in terms of pain relief and patient satisfaction, but resulted in a faster return-
to-sport activity. In the current study, we report for the first time the long-term efficacy, safety, return-to-sport, and subsequent
recurrence rate of ankle sprain at 2 years post injury.

20
71
Methods
This randomized controlled study was conducted between March 2003 and December 2005 with follow-up to June 2008 in
3 primary care sport medicine facilities in Ontario, Canada. The study and follow-up was approved by the institutional ethics
committee and was conducted according to the Declaration of Helsinki Good Clinical Practice guidelines. All patients signed
informed consent prior to participation.
The study methodology was previously described.17 Briefly, the study included a screening phase where patients were assessed
based on selected inclusion and exclusion criteria. Prior to enrollment, a diagnosis of first or second-degree ankle sprain was
made by athletic trainers affiliated with university athletic programs, emergency physicians at affiliated local hospitals, and family
physicians in the referral base for the 3 sport medicine clinics. Enrolled patients were required to report to the sport medicine
clinics within 48 hours of their injury. Pain severity at enrollment was assessed using a pain visual analog scale (VAS) and
included eligibility of a VAS at rest of > 4.5 cm (0–10 cm). Patients were excluded if they had bilateral ankle sprain, third-degree
ankle sprain, or had previous sprain in the last 6 months. An x-ray of the ankle joint was performed prior to enrollment
to exclude other pathologies. Patients were randomized (1:1) to 1 of the 2 treatment groups using a computer-generated
randomization schedule: periarticular hyaluronic acid (HA) (MW range, 750–1 million kDal, 20 mg) + usual standard of care
RICE or periarticular placebo (PL) + usual standard of care RICE. The first dose of study treatment was administered on
Day 1 (within 48 hours of injury) and the second dose was administered on Day 4 (±1 day).
Assessments were done at baseline, and Days 4, 8, 30, 90, and 712 (Table 1). Efficacy measures included patients’ VAS
of pain on weight bearing (0–10 cm) and walking 20 m (0–10 cm), patients’ global assessment of ankle injury (5-point
categorical scale), patients’ assessment of return to normal function/activity in sport (5-point categorical scale), patients’
satisfaction assessment (10-point categorical scale), number of recurrent ankle sprains, number of other injuries, number
of days missing from incident sport due to ankle sprain and AEs as per World Health Organization (WHO) definition.
Following measurement of the outcome assessments, those randomized to HA treatment received a single injection of HA
(0.7–1.2 mL) or placebo (normal saline 0.7–1.2 mL). Injections were performed using previously18,19 described blinded syringes
affixed to a 27-gauge, 1-inch needle.17 Briefly, skin was prepped using Betadine 1%. Injections were delivered by the study
physician using a standard approach along the anterior talofibular ligament using clinical landmarks.

The injection (1.2 mL total) was delivered during a single penetration along 3 planes from anteroposterior, medial, and lateral from
the proximal ligamentous landmark. Assessments and injections were repeated on Day 4 (± 1 day). All randomized patients received
standard care consisting of RICE, assistive devices as determined by the study physician including crutches, taping, or bracing
but not physiotherapy, and oral or topical medications such as NSAIDs. These latter interventions could be used after Day 8 at the
patient’s discretion.
Rescue medication (500 mg acetaminophen tablets, up to 4 tablets daily) was allowed in both groups but not for the 24 hours
prior to study visit. Patients were free to withdraw at any time during the trial.
Follow-up assessments were completed at Days 8 (± 2 days), 30 (± 7 days), 90 (± 7 days), and 712 (± 7 days). Adverse events
and concomitant medications were assessed throughout the patients’ participation in the study.
Demographic and baseline data were compared within the 2 groups using Student’s t-tests for continuous and χ2 statistics for
noncontinuous variables. Statistical analysis was based on the intent to treat (ITT) population. Efficacy and safety variables were
analyzed between groups using appropriate statistical methods including Student’s t-test for quantitative, χ2 test for nominal, and
Mann-Whitney U test for ordinal variables. The data analysis was performed using the SAS® version 8.2 (SAS Institute, Cary, NC).
All statistical tests were 2-tailed at a 5% level of significance.

Results
A total of 499 patients were screened and the ITT population was 158 patients (Figure 1). The average age was 26 ± 7 years
and 24 ± 8 years for the HA and PL groups, respectively, with equal male to female representation between groups. Thirty percent
of sprains were first events and 65% were Grade 1 with no difference between groups. There was no difference between the groups

72
21
at baseline. We obtained 100% compliance with the injection series throughout the treatment phase and 70% to 75% retention at
2 years (Figure 1). Time to intervention was not different between groups (39 ± 4 hours).
Theprimaryefficacycriterionwasthedecreasefrombaseline to visit 2 (Day 8 ± 1) in weight-bearing pain calculated in the ITT population
(Table 2). This, and changes in walking pain, were -3.16 ± 1.18 cm and -1.83 ± 1.1 cm (%) (weight-bearing pain) and -4.99 ± 2.02
cm and -3.76 ± 2.43 cm (%) (walking pain) in the HA and PL groups, respectively (P < 0.0001), giving an inter-group difference of
1.31 cm and 1.23 cm in favor of treatment.
The differences between groups were also significant at visit 3 (Day 30), visit 4 (Day 90), and visit 5 (Day 712) in favor of the treatment
group (Table 2).
Globally, all efficacy parameters improved during the study in both groups. However, intergroup comparisons showed a statistically
significant difference in favor of HA on most efficacy parameters (Table 2). For parameters where such a difference was not obtained
for all visits, the improvement was more marked in the HA than in the control. The results for the secondary efficacy variables were
therefore globally consistent with those concerning the primary outcomes.
Fewer recurrent sprains were observed in the HA (7) versus PL (16) group at 2 years (Figure 2). Further, this was associated with
fewer days missing (21) versus (41) for the HA and PL groups, respectively (Figure 3). There was a small but nonsignificant difference
favoring HA in number of total MSK injuries (18 vs 24) reported at 2 years, suggesting that missing days from sport in patients post
ankle sprain is primarily driven by recurrent ankle sprain.

Abbreviations: HA, hyaluronic acid group; PL, usual care group; SD, standard deviation.
Sprain grade is percent presenting with Grade 1 sprain;VAS pain on weight bearing is the score decrease from baseline in cm on 10-point Likert scale;VAS pain on walking is the score decrease from
baseline in cm on 10-point Likert scale; patient global assessment of ankle injury is self reported score from 1–5 where 1 = very poor assessment of injury on health and 5 = very good assessment
of injury on health; patient assessment of return to normal activity in sport is self reported score from 1 = severely restricted to 5 = normal activity; patient satisfaction with treatment is self reported
score from 1 = not satisfied to 10 = completely satisfied with treatment; adverse events are the self reported adverse events at each study visit.
*Statistically significant differences within treatment groups for these parameters.

73
22
Patient satisfaction with treatment scores among subjects showed that 75% versus 48% and 77% versus 57% were satisfied at
Days 4 and 8 while 95% versus 76% were satisfied at Day 30, 93% vs 67% at Day 90, and 95% versus 60% at Day 712 (χ2 test,
P < 0.001) (Table 2).
Three AEs were observed among all subjects and consisted of pain requiring the self-medication with NSAID (2 HA, 1 PL) including
1 mild erythema and pain (HA) at the injection site not requiring further intervention at Day 4, and 3 pain and 1 mild erythema at the
injection site in the HA vs 2 erythema, 1 pain and 1 swelling at Day 8 in PL (Table 2). The pain AEs in the 2 groups did not differ in
intensity. At 712 days, 3 pain AEs for HA and 5 pain AEs for PL were reported (Table 2). There were no serious AEs.
No difference in concomitant treatment or physical therapy between groups was observed to Day 90. At Day 712 more patients in
PL had used at least 7 days of NSAID compared with HA (P < 0.001) and more had utilized physical therapy (P < 0.001).

Discussion
Conservative treatment of acute ankle sprain may limit disability to an average of 8 days for a Grade 1 and 15 days for a
Grade 2,3,4 but this treatment is not ideal as it remains that a significant number of patients will have ongoing disability
including instability, recurrent sprains, and time away from sport. Further, while NSAIDs effectively reduce the pain and
swelling associated with acute ankle sprain,8–12 they are nonselective and may cause significant AEs including gastro-
intestinal intolerance, serious events such as ulcers and bleeding, as well as potential negative cardio-renal effects.
Hence, therapeutic options for ankle sprains have been limited while both the short and long-term recovery suggest that
alternative treatments, particularly those that improve return-to-sport with minimal sequelae, are needed.
Hyaluronic acid is a naturally occurring biological substance representing an unbranched, high-molecular-weight
polysaccharide as a major component of ligamentous, cartilaginous, and synovial ultrastructure16 with no demonstrated
effect on gastrointestinal and platelet function. It has proven efficacy and safety in the treatment of osteoarthritis.20 We
have previously reported the short-term efficacy and safety of periarticular administration of HA in acute ankle sprain
compared with standard of care treatment.17

In the current study, we extend our previous findings that periarticular HA was associated with fewer recurrent ankle sprains and
fewer injury days missing from sport compared with PL. This is the first study to our knowledge, regardless of management, that
has demonstrated impact on future ankle injury and morbidity from a standardized treatment of acute ankle sprain. The primary
efficacy criterion was a decrease in pain in the first 8 days in the ITT population. Based on this criterion, periarticular HA was found
to be significantly more effective than control. This change of 3 cm is considered clinically significant.11 Furthermore, almost all
the secondary criteria, including patient global satisfaction, were also improved during the trial compared with control, even up to
90 days.
Further, clinical implications of our results were confirmed by the fact that the number of patients who withdrew for AE or lack
of efficacy was small for both groups, and that our short-term results are consistent with other treatments of ankle sprain with
topical NSAIDs 10,21,22 and oral NSAIDs and COX-2 inhibitors.9,12 Importantly, we reported a very low incidence of AEs with
high satisfaction both in the short and longer-term. Further, less use of concomitant medication for ankle pain and fewer physical
therapy sessions was observed at 2 years in the HA versus PL group, suggesting that this new treatment modality could have added
value to clinicians and their patients. Currently, those with ankle instability after an acute sprain may be advised that wearing a
prophylactic external brace to reduce the risk of a future sprain, although the results including costs and performance to date are
not clear.13 Investigation of the costs and benefits of periarticular HA versus conventional therapy requires attention to return to-
sport and recurrent ankle injury across the range of sport settings, including amateur to professional, to allow clinicians and athletes
to make informed decisions regarding their therapy options.

Summary
Our results showed superior short and long-term therapeutic response of periarticular HA in patients with acute ankle
sprain versus standard of care treatment. Further, periarticular HA resulted in fewer recurrent ankle sprains, less use of
concomitant treatments, and fewer days missing from sport 2 years after the initial injury. Given that this treatment is easily
performed in primary care sport medicine and has high patient satisfaction, these results suggest that periarticular HA in
acute ankle should be considered an important alternative by physicians and their patients.

Acknowledgments
Theauthorsacknowledgetheparticipationofstudyinvestigators at the Fowler-Kennedy Sport Medicine Clinic, London, Canada.

Conflict of Interest Statement


Michael J. Petrella, PhD, Anthony Cogliano, MD, and Robert
J. Petrella, MD, PhD declare no conflicts of interest.

74
23
Referinte
Hyaluronic acid increases tendon derived cell viability and collagen type
I expression in vitro: Comparative study of four different Hyaluronic acid
preparations by molecular weight (pag. 3)

References
1. Oliva F, Osti L, Padulo J, Maffulli N. Epidemiology of the rotator cuff tears: a new incidence related to thyroid disease. Muscles Ligaments Tendons J.
2014;4(3):309–14.
2. Osti L, Rizzello G, Panascì M, Denaro V, Maffulli N. Full thickness tears: retaining the cuff. Sports Med Arthosc. 2011;19(4):409–19.
3. Oliva F, Berardi AC, Misiti S, Maffulli N. Thyroid hormones and tendon: current views and future perspectives. Concise review. Muscles Ligaments Tendons
J. 2013;3(3):201–3.
4. Klatte-Schulz F, Pauly S, Scheibel M, Greiner S, Gerhardt C, Schmidmaier G, et al. Influence of age on the cell biological characteristics and the stimulation
potential of male human tenocyte-like cells. Eur Cell Mater. 2012;24:74–89.
5. Loppini M, Maffulli N. Conservative management of tendinopathy: an evidence-based approach. Muscles Ligaments Tendons J. 2012;1(4):134–7.
6. Rutjes AW, Jüni P, da Costa BR, Trelle S, Nüesch E, Reichenbach S. Viscosupplementation for osteoarthitis of the knee: a systematic review and meta-
analysis. Ann Intern Med. 2012;157(3):180–91.
7. Abate M, Schiavone C, Salini V. The use of hyaluronic acid after tendon surgery and in tendinopathies. Biomed Res Int. 2014;2014:783632.
8. Giai Via A, De Cupis M, Spoliti M, Oliva F. Clinical and biological aspects of rotator cuff tears. Muscles Ligaments Tendons J. 2014;3(4):359.
9. Yuan J, Wang MX, Murrell GA. Cell death and tendinopathy. Clin Sports Med. 2003;22(4):693–701.
10. Benson RT, McDonnell SM, Knowles HJ, Rees JL, Carr AJ, Hulley PA. Tendinopathy and tears of the rotator cuff are associated with hypoxia and apop-
tosis. J Bone Joint Surg Br. 2010;92(3):448–53.
11. Meyer K. Chemical structure of hyaluronic acid. Fed Proc. 1958;17(4):1075–7.
12. Melrose J, Numata Y, Ghosh P. Biotinylated hyaluronan: a versatile and highly sensitive probe capable of detecting nanogram levels of hyaluronan binding
proteins (hyaladherins) on electroblots by a novel affinity detection procedure. Electrophoresis. 1996;17(1):205.
13. Chen WY, Grant ME, Schor AM, Schor SL. Differences between adult and fetal fibroblasts in the regulation of hyaluronate synthesis: correlation with
migratory activity. J Cell Sci. 1989;94(Pt 3):577–84.
14. Klein ES, Asculai SS, Ben-Ari GY. Effects of hyaluronic acid on fibroblast behavior in peritoneal injury. J Surg Res. 1996;61(2):473–6.
15. Hall CL, Wang C, Lange LA, Turley EA. Hyaluronan and the hyaluronan receptor RHAMM promote focal adhesion turnover and transient tyrosine kinase
activity. J Cell Biol. 1994;126(2):575–88.
16. Wiig M, Abrahamsson SO, Lundborg G. Effects of hyaluronan on cell proliferation and collagen synthesis: a study of rabbit flexor tendons in vitro. J Hand
Surg Am. 1996;21(4):599–604.
17. Bernard E, Hornebeck W, Robert L. Effect of hyaluronan on the elastase-type activity of human skin fibroblasts. Cell Biol Int. 1994;18(10):967–71.
18. Matuoka K, Namba M, Mitsui Y. Hyaluronate synthetase inhibition by normal and transformed human fibroblasts during growth reduction. J Cell Biol.
1987;104(4):1105–15.
19. Dean BJ, Snelling SJ, Dakin SG, Murphy RJ, Javaid MK, Carr AJ. Differences in glutamate receptors and inflammatory cell numbers are associated with
the resolution of pain in human rotator cuff tendinopathy. Arthritis Res Ther. 2015;17:176.
20. Oliva F, Berardi AC, Misiti S, Verga Falzacappa C, Iacone A, Maffulli N. Thyroid hormones enhance growth and counteract apoptosis in human tenocytes
isolated from rotator cuff tendons. Cell Death Disease. 2013;4:e705.
21. Yao L, Bestwick CS, Bestwick LA, Maffulli N, Aspden RM. Phenotypic drift in human tenocyte culture. Tissue Eng. 2006;12(7):1843–9.
22. Yuan J, Murrell GA, Trickett A, Wang MX. Involvement of cytochome c release and caspase-3 activation in the oxidative stress-induced apoptosis in hu-
man tendon fibroblasts. Biochimica et Biophysica Acta. 2003;1641(1):35–41.
23. Berardi AC, Oliva F, Berardocco M, la Rovere M, Accorsi P, Maffulli N. Thyroid hormones increase collagen type Iand cartilage oligomeric matrix protein
(COMP) expression in vitro human tenocytes. Muscles Ligaments Tendons J.
2014;4(3):285–91.
24. Hanna S, Khalil B, Nasrallah A, Saykali BA, Sobh R, Nasser S, et al. StarD13 is a tumor suppressor in breast cancer that regulates cell motility and invasion.
Int J Oncol. 2014;44(5):1499–511.
25. Evanko SP, Tammi MI, Tammi RH, Wight TN. Hyaluronan-dependent pericellular matrix. Adv Drug Deliv Rev. 2007;59(13):1351–65.
26. Karna E, Miltyk W, Surazyński A, Pałka JA. Protective effect of hyaluronic acid on interleukin-1-induced deregulation of beta1-integrin and insulin-like
growth factor-I receptor signaling and collagen biosynthesis in cultured human chondrocytes. Mol Cell Biochem. 2008;308(1–2):57–64.
27. Oliva F, Gatti S, Porcellini G, Forsyth NR, Maffulli N. Growth factors and tendon healing. Med Sport Sci. 2012;57:53–64.
28. Maffulli N, Ewen SW, Waterston SW, Reaper J, Barrass V. Tenocytes from ruptured and tendinopathic achilles tendons produce greater quantities of type
III collagen than tenocytes from normal achilles tendons. An in vitro model of human tendon healing. Am J Sports Med. 2000;28(4):499–505.
29. Yamada T, Gotoh M, Nakama K, Mitsui Y, Higuchi F, Nagata K. Effects of hyaluronan on cell proliferation and mRNA expression of procollagens alpha 1 (I)
and alpha 1 (III) in tendon-derived fibroblasts from patients with rotator cuff disease: an in vitro study. Am J Sports Med. 2007;35(11):1870–6.

75
24
Tendon Injury and Tendinopathy: Healing and Repair (pag. 11)

References
1. Almekinders LC, Almekinders SV. Outcome in the treatment of chronic overuse sports injuries: a retrospective study. J Orthop Sports Phys Ther.
1994;19:157-61.
2. James SL, Bates BT, Osternig LR. Injuries to runners. Am J Sports Med. 1978; 6:40-50.
3. Gruchow HW, Pelletier D. An epidemiologic study of tennis elbow. Incidence, recurrence, and effectiveness of prevention strategies. Am J Sports Med.
1979; 7:234-8.
4. Benjamin M, Ralphs J. Functional and developmental anatomy of tendons and ligaments. In: Gordon SL, Blair SJ, Fine LJ, editors. Repetitive motion
disorders of the upper extremity. Rosemont, IL: American Academy of Orthopaedic Surgeons; 1995. p 185-203.
5. Kannus P, Jozsa L, Jarvinnen M. Basic science of tendons. In: Garrett WE Jr, Speer KP, Kirkendall DT, editors. Principles and practice of orthopaedic sports
medicine. Philadelphia: Lippincott Williams and Wilkins; 2000. p 21-37.
6. O’Brien M. Structure and metabolism of tendons. Scand J Med Sci Sports. 1997;7:55-61.
7. Jozsa L, Balint JB, Reffy A, Demel Z. Histochemical and ultrastructural study of adult human tendon. Acta Histochem. 1979;65:250-7.
8. Kvist M, Jozsa L, Jarvinen MJ, Kvist H. Chronic Achilles paratenonitis in athletes: a histological and histochemical study. Pathology. 1987;19:1-11.
9. Kvist M, Jozsa L, Jarvinen M, Kvist H. Fine structural alterations in chronic Achilles paratenonitis in athletes. Pathol Res Pract. 1985;180:416-23.
10. Kannus P, Jozsa L. Histopathological changes preceding spontaneous rupture of a tendon. A controlled study of 891 patients. J Bone Joint Surg Am.
1991; 73:1507-25.
11. Vailas AC, Tipton CM, Laughlin HL, Tcheng TK, Matthes RD. Physical activity and hypophysectomy on the aerobic capacity of ligaments and tendons. J
Appl Physiol. 1978;44:542-6.
12. Williams JG. Achilles tendon lesions in sport. Sports Med. 1986;3:114-35.
13. Hess GP, Cappiello WL, Poole RM, Hunter SC. Prevention and treatment of overuse tendon injuries. Sports Med. 1989;8:371-84.
14. Jozsa L, Lehto M, Kannus P, Kvist M, Reffy A, Vieno T, Jarvinen M, Demel S, Elek E. Fibronectin and laminin in Achilles tendon. Acta Orthop Scand.
1989; 60:469-71.
15. Tipton CM, Matthes RD, Maynard JA, Carey RA. The influence of physical activity on ligaments and tendons. Med Sci Sports. 1975;7:165-75.
16. Kirkendall DT, Garrett WE. Function and biomechanics of tendons. Scand J Med Sci Sports. 1997;7:62-6.
17. Astrom M. On the nature and etiology of chronic achilles tendinopathy [the- sis]. Lund, Sweden: University of Lund; 1997.
18. Jozsa LG, Kannus P. Human tendons: anatomy, physiology, and pathology. Champaign, IL: Human Kinetics; 1997.
19. Movin T, Kristoffersen-Wiberg M, Shalabi A, Gad A, Aspelin P, Rolf C. Intraten- dinous alterations as imaged by ultrasound and contrast medium-enhanced
mag- netic resonance in chronic achillodynia. Foot Ankle Int. 1998;19:311-7.
20. Jozsa L, Kannus P, Balint JB, Reffy A. Three-dimensional ultrastructure of hu- man tendons. Acta Anat (Basel). 1991;142:306-12.
21. Balint BJ, Jozsa L. [Investigation on the construction and spacial structure of human tendons (author’s transl)]. Magy Traumatol Orthop Helyreallito Seb.
1978;21:293-9. Hungarian.
22. Hueston JT, Wilson WF. The aetiology of trigger finger explained on the basis of intratendinous architecture. Hand. 1972;4:257-60.
23. Lawler J. The structural and functional properties of thrombospondin. Blood. 1986;67:1197-209.
24. Miller RR, McDevitt CA. Thrombospondin in ligament, meniscus and interver- tebral disc. Biochim Biophys Acta. 1991;1115:85-8.
25. Riley GP, Harrall RL, Cawston TE, Hazleman BL, Mackie EJ. Tenascin-C and human tendon degeneration. Am J Pathol. 1996;149:933-43.
26. Kannus P, Jozsa L, Jarvinen TA, Jarvinen TL, Kvist M, Natri A, Jarvinen M. Location and distribution of non-collagenous matrix proteins in musculoskeletal
tissues of rat. Histochem J. 1998;30:799-810.
27. Oberhauser AF, Marszalek PE, Erickson HP, Fernandez JM. The molecular elasticity of the extracellular matrix protein tenascin. Nature. 1998;393:181-5.
28. Mehr D, Pardubsky PD, Martin JA, Buckwalter JA. Tenascin-C in tendon re- gions subjected to compression. J Orthop Res. 2000;18:537-45.
29. Jarvinen TA, Jozsa L, Kannus P, Jarvinen TL, Kvist M, Hurme T, Isola J, Kalimo H, Jarvinen M. Mechanical loading regulates tenascin-C expression in the
osteo- tendinous junction. J Cell Sci. 1999;112:3157-66.
30. Mackie EJ, Ramsey S. Expression of tenascin in joint-associated tissues during development and postnatal growth. J Anat. 1996;188:157-65.
31. Elliott DH. Structure and function of mammalian tendon. Biol Rev Camb Philos Soc. 1965;40:392-421.
32. Kastelic J, Galeski A, Baer E. The multicomposite structure of tendon. Con- nect Tissue Res. 1978;6:11-23.
33. Lundborg G, Myrhage R. The vascularization and structure of the human digi- tal tendon sheath as related to flexor tendon function. An angiographic and
histo- logical study. Scand J Plast Reconstr Surg. 1977;11:195-203.
34. Doyle JR. Anatomy of the finger flexor tendon sheath and pulley system. J Hand Surg [Am]. 1988;13:473-84.
35. Kvist M, Jozsa L, Kannus P, Isola J, Vieno T, Jarvinen M, Lehto M. Morphology and histochemistry of the myotendineal junction of the rat calf muscles.
His- tochemical, immunohistochemical and electron-microscopic study. Acta Anat (Basel). 1991;141:199-205.
36. Michna H. A peculiar myofibrillar pattern in the murine muscle-tendon junc- tion. Cell Tissue Res.1983;233:227-31.
37. Trotter JA, Baca JM. A stereological comparison of the muscle-tendon junc- tions of fast and slow fibers in the chicken. Anat Rec. 1987;218:256-66.
38. Tidball JG, Daniel TL. Myotendinous junctions of tonic muscle cells: structure and loading. Cell Tissue Res. 1986;245:315-22.
39. Tidball JG. Myotendinous junction injury in relation to junction structure and molecular composition. Exerc Sport Sci Rev. 1991;19:419-45.
40. Nikolaou PK, Macdonald BL, Glisson RR, Seaber AV, Garrett WE Jr. Biome- chanical and histological evaluation of muscle after controlled strain injury.
Am J Sports Med. 1987;15:9-14.
41. Garrett WE Jr. Muscle strain injuries: clinical and basic aspects. Med Sci Sports Exerc. 1990;22:436-43.
42. Jarvinen M, Kannus P, Kvist M, Isola J, Lehto M, Jozsa L. Macromolecular composition of the myotendinous junction. Exp Mol Pathol. 1991;55:230-7.
43. Benjamin M, Ralphs JR. Fibrocartilage in tendons and ligaments—an adapta- tion to compressive load. J Anat. 1998;193:481-94.
44. Benjamin M, Qin S, Ralphs JR. Fibrocartilage associated with human tendons and their pulleys. J Anat. 1995;187:625-33.

76
25
45. Evans EJ, Benjamin M, Pemberton DJ. Fibrocartilage in the attachment zones of the quadriceps tendon and patellar ligament of man. J Anat.
1990;171:155-62.
46. Carr AJ, Norris SH. The blood supply of the calcaneal tendon. J Bone Joint Surg Br. 1989;71:100-1.
47. Kvist M, Hurme T, Kannus P, Jarvinen T, Maunu VM, Jozsa L, Jarvinen M. Vas- cular density at the myotendinous junction of the rat gastrocnemius muscle
after immobilization and remobilization. Am J Sports Med. 1995;23:359-64.
48. Kvist M, Jozsa L, Jarvinen M. Vascular changes in the ruptured Achilles ten- don and paratenon. Int Orthop. 1992;16:377-82.
49. Naito M, Ogata K. The blood supply of the tendon with a paratenon. An exper- imental study using hydrogen washout technique. Hand. 1983;15:9-14.
50. Reynolds NL, Worrell TW. Chronic Achilles peritendinitis: etiology, pathophysi- ology, and treatment. J Orthop Sports Phys Ther. 1991;13:171-6.
51. Field PL. Tendon fibre arrangement and blood supply. Aust NZ J Surg. 1971; 40:298-302.
52. Niculescu V, Matusz P. The clinical importance of the calcaneal tendon vascu- lature (tendo calcaneus). Morphol Embryol (Bucur). 1988;34:5-8.
53. Astrom M. Laser Doppler flowmetry in the assessment of tendon blood flow. Scand J Med Sci Sports. 2000;10:365-7.
54. Leversedge FJ, Ditsios K, Goldfarb CA, Silva MJ, Gelberman RH, Boyer MI. Vascular anatomy of the human flexor digitorum profundus tendon insertion.
J Hand Surg [Am]. 2002;27:806-12.
55. Lephart SM, Pincivero DM, Giraldo JL, Fu FH. The role of proprioception in the management and rehabilitation of athletic injuries. Am J Sports Med.
1997;25:130-7.
56. Fitzgerald MJT. Neuroanatomy: basic and clinical. 2nd ed. Philadelphia: Bal- liere Tindall; 1992.
57. Brodal A. Neurological anatomy in relation to clinical medicine. 3rd ed. New York: Oxford University Press; 1981.
58. Barr ML, Kiernan JA. The human nervous system: an anatomical viewpoint. 5th ed. Philadelphia: Lippincott; 1988.
59. Ackermann PW, Li J, Finn A, Ahmed M, Kreicbergs A. Autonomic innervation of tendons, ligaments and joint capsules. A morphologic and quantitative
study in the rat. J Orthop Res. 2001;19:372-8.
60. Best TM, Garrett WE. Basic science of soft tissue: muscle and tendon. In: DeLee JC, Drez D Jr, editors. Orthopaedic sports medicine: principles and
prac- tice. Philadelphia: WB Saunders; 1994. p 1.
61. O’Brien M. Functional anatomy and physiology of tendons. Clin Sports Med. 1992;11:505-20.
62. Oxlund H. Relationships between the biomechanical properties, composi- tion and molecular structure of connective tissues. Connect Tissue Res. 1986;
15:65-72.
63. Carlstedt CA, Nordin M. Biomechanics of tendons and ligaments. In: Nordin M, Frankel VH, editors. Basic biomechanics of the musculoskeletal system.
2nd ed. Philadelphia: Lea and Febiger; 1989. p 59-74.
64. Viidik A. Tendons and ligaments. In: Comper W, editor. Extracellular matrix. Volume 1. Amsterdam: Harwood Academic Publishers; 1996. p 303-27.
65. Fyfe I, Stanish WD. The use of eccentric training and stretching in the treat- ment and prevention of tendon injuries. Clin Sports Med. 1992;11:601-24.
66. Diamant J, Keller A, Baer E, Litt M, Arridge RG. Collagen; ultrastructure and its relation to mechanical properties as a function of aging. Proc R Soc Land
B Biol Sci. 1972;180:293-315.
67. Butler DL, Grood ES, Noyes FR, Zernicke RF. Biomechanics of ligaments and tendons. Exerc Sport Sci Rev. 1978;6:125-81.
68. Viidik A. Functional properties of collagenous tissues. Int Rev Connect Tissue Res. 1973;6:127-215.
69. Zernicke RF, Loitz BJ. Exercise-related adaptations in connective tissue. In: Komi PV, editor. The encyclopaedia of sports medicine. Strength and power
in sport. Volume 3. Boston: Blackwell Scientific Publications; 2002. p 93-113.
70. Mosler E, Folkhard W, Knorzer E, Nemetschek-Gansler H, Nemetschek T, Koch MH. Stress-induced molecular rearrangement in tendon collagen. J Mol
Biol. 1985;182:589-96.
71. Curwin S, Stanish WD. Tendinitis, its etiology and treatment. Lexington, MA: Collamore Press; 1984.
72. Kastelic J, Baer E. Deformation in tendon collagen. Symp Soc Exp Biol. 1980;34:397-435.
73. Sasaki N, Shukunami N, Matsushima N, Izumi Y. Time-resolved X-ray diffrac- tion from tendon collagen during creep using synchrotron radiation. J
Biomech. 1999;32:285-92.
74. Oakes BW, Singleton C, Haut RC. Correlation of collagen fibril morphology and tensile modulus in the repairing and normal rabbit patella tendon. Trans
Or- thop Res Soc. 1998;23:24.
75. Shadwick RE. Elastic energy storage in tendons: mechanical differences re- lated to function and age. J Appl Physiol. 1990;68:1033-40.
76. Zernicke RF, Garhammer J, Jobe FW. Human patellar-tendon rupture. J Bone Joint Surg Am. 1977;59:179-83.
77 Komi PV, Salonen M, Jarvinen M, Kokko O. In vivo registration of Achilles tendon forces in man. I. Methodological development. Int J Sports Med. 1987;
8 Suppl 1:3-8.
78. Komi PV. Relevance of in vivo force measurements to human biomechanics. J Biomech. 1990;23 Suppl 1:23-34.
79. Komi PV, Fukashiro S, Jarvinen M. Biomechanical loading of Achilles tendon during normal locomotion. Clin Sports Med. 1992;11:521-31.
80. Barfred T. Experimental rupture of the Achilles tendon. Comparison of various types of experimental rupture in rats. Acta Orthop Scand. 1971;42:528-43.
81. Barfred T. Experimental rupture of the Achilles tendon. Comparison of experi- mental ruptures in rats of different ages and living under different conditions.
Acta Orthop Scand. 1971;42:406-28.
82. Barfred T. Kinesiological comments on subcutaneous ruptures of the Achilles tendon. Acta Orthop Scand. 1971;42:397-405.
83. Komi PV. Physiological and biomechanical correlates of muscle function: ef- fects of muscle structure and stretch-shortening cycle on force and speed.
Exerc Sport Sci Rev. 1984;12:81-121.
84. Stanish WD, Curwin S, Rubinovich M. Tendinitis: the analysis and treatment for running. Clin Sports Med. 1985;4:593-609.
85. Soldatis JJ, Goodfellow DB, Wilber JH. End-to-end operative repair of Achilles tendon rupture. Am J Sports Med. 1997;25:90-5.
86. Inglis AE, Scott WN, Sculco TP, Patterson AH. Ruptures of the tendo achillis. An objective assessment of surgical and non-surgical treatment. J Bone
Joint Surg Am. 1976;58:990-3.
87. Arner O, Lindholm A, Orell SR. Histologic changes in subcutaneous rupture of the Achilles tendon; a study of 74 cases. Acta Chir Scand. 1959;116:
484-90.
88. Tallon C, Maffulli N, Ewen SW. Ruptured Achilles tendons are significantly more degenerated than tendinopathic tendons. Med Sci Sports Exerc. 2001;
33:1983-90.
89. Kvist M. Achilles tendon overuse injuries: a clinical and pathophysiological study in athletes [Thesis]. Turku, Finland: University of Turku; 1991.
90. Kvist M. Achilles tendon injuries in athletes. Sports Med. 1994;18:173-201.
91. Nigg BM. The role of impact forces and foot pronation: a new paradigm. Clin J Sport Med. 2001;11:2-9.
92. Clement DB, Taunton JE, Smart GW. Achilles tendinitis and peritendinitis: eti- ology and treatment. Am J Sports Med. 1984;12:179-84.
93. Selvanetti A, Cipolla M, Puddu G. Overuse tendon injuries: basic science and classification. Oper Tech Sports Med. 1997;5:110-7.
94. Benazzo F, Maffulli N. An operative approach to Achilles tendinopathy. Sports Med Arthroscopy Rev. 2000;8:96-101.
95. Ker RF. The implications of the adaptable fatigue quality of tendons for their construction, repair and function. Comp Biochem Physiol A Mol Integr Physiol.
2002;133:987-1000.
96. Arndt AN, Komi PV, Bruggemann GP, Lukkariniemi J. Individual muscle contri- butions to the in vivo achilles tendon force. Clin Biomech (Bristol, Avon).
1998; 13:532-41.
97. Goodship AE, Birch HL, Wilson AM. The pathobiology and repair of tendon and ligament injury. Vet Clin North Am Equine Pract. 1994;10:323-49.
98. Bestwick CS, Maffulli N. Reactive oxygen species and tendon problems: re- view and hypothesis. Sports Med Arthroscopy Rev. 2000;8:6-16.
99. Wang MX, Wei A, Yuan J, Clippe A, Bernard A, Knoops B, Murrell GA. Antioxi- dant enzyme peroxiredoxin 5 is upregulated in degenerative human tendon.
Bio- chem Biophys Res Commun. 2001;284:667-73.
100. Birch HL, Rutter GA, Goodship AE. Oxidative energy metabolism in equine tendon cells. Res Vet Sci. 1997;62:93-7.
101. Ker RF. Dynamic tensile properties of the plantaris tendon of sheep (Ovis ar- ies). J Exp Biol. 1981;93:283-302.
102. Riemersma DJ, Schamhardt HC. In vitro mechanical properties of equine tendons in relation to cross-sectional area and collagen content. Res Vet Sci.
1985;39:263-70.
103. Wilson AM, Goodship AE. Exercise-induced hyperthermia as a possible mechanism for tendon degeneration. J Biomech. 1994;27:899-905.
104. Arancia G, Crateri Trovalusci P, Mariutti G, Mondovi B. Ultrastructural changes induced by hyperthermia in Chinese hamster V79 fibroblasts. Int J Hy-
perthermia. 1989;5:341-50.
105. Birch HL, Wilson AM, Goodship AE. The effect of exercise-induced localised hyperthermia on tendon cell survival. J Exp Biol. 1997;200:1703-8.
106. Yuan J, Wang MX, Murrell GA. Cell death and tendinopathy. Clin Sports Med. 2003;22:693-701.

77
26
111. Machner A, Baier A, Wille A, Drynda S, Pap G, Drynda A, Mawrin C, Buhling F, Gay S, Neumann W, Pap T. Higher susceptibility to Fas ligand induced
apoptosis and altered modulation of cell death by tumor necrosis factor-alpha in periarticu- lar tenocytes from patients with knee joint osteoarthritis. Arthritis
Res Ther. 2003;5:R253-61.
112. Stone D, Green C, Rao U, Aizawa H, Yamaji T, Niyibizi C, Carlin G, Woo SL. Cytokine-induced tendinitis: a preliminary study in rabbits. J Orthop Res.
1999; 17:168-77.
113. Sullo A, Maffulli N, Capasso G, Testa V. The effects of prolonged peritendi- nous administration of PGE1 to the rat Achilles tendon: a possible animal
model of chronic Achilles tendinopathy. J Orthop Sci. 2001;6:349-57.
114. Wang JH, Jia F, Yang G, Yang S, Campbell BH, Stone D, Woo SL. Cyclic me- chanical stretching of human tendon fibroblasts increases the production
of pros- taglandin E2 and levels of cyclooxygenase expression: a novel in vitro model study. Connect Tissue Res. 2003;44:128-33.
115. Skutek M, van Griensven M, Zeichen J, Brauer N, Bosch U. Cyclic mechani- cal stretching enhances secretion of Interleukin 6 in human tendon
fibroblasts. Knee Surg Sports Traumatol Arthrosc. 2001;9:322-6.
116. Tsuzaki M, Bynum D, Almekinders L, Yang X, Faber J, Banes AJ. ATP modu- lates load-inducible IL-1beta, COX 2, and MMP-3 gene expression in human
ten- don cells. J Cell Biochem. 2003;89:556-62.
117. Tsuzaki M, Guyton G, Garrett W, Archambault JM, Herzog W, Almekinders L, Bynum D, Yang X, Banes AJ. IL-1 beta induces COX2, MMP-1, -3 and -13,
ADAMTS- 4, IL-1 beta and IL-6 in human tendon cells. J Orthop Res. 2003;21:256-64.
118. Archambault J, Tsuzaki M, Herzog W, Banes AJ. Stretch and interleukin- 1beta induce matrix metalloproteinases in rabbit tendon cells in vitro. J Orthop
Res. 2002;20:36-9.
119. Corps AN, Harrall RL, Curry VA, Fenwick SA, Hazleman BL, Riley GP. Cipro- floxacin enhances the stimulation of matrix metalloproteinase 3 expression
by interleukin-1beta in human tendon-derived cells. A potential mechanism of fluoroquinolone-induced tendinopathy. Arthritis Rheum. 2002;46:3034-40.
120. Gold L, Igra H. Levofloxacin-induced tendon rupture: a case report and re- view of the literature. J Am Board Fam Pract. 2003;16:458-60.
121. van der Linden PD, Sturkenboom MC, Herings RM, Leufkens HG, Stricker BH. Fluoroquinolones and risk of Achilles tendon disorders: case-control
study. BMJ. 2002;324:1306-7.
122. Corps AN, Curry VA, Harrall RL, Dutt D, Hazleman BL, Riley GP. Ciprofloxacin reduces the stimulation of prostaglandin E(2) output by interleukin-1beta
in hu- man tendon-derived cells. Rheumatology (Oxford). 2003;42:1306-10.
123. Williams RJ 3rd, Attia E, Wickiewicz TL, Hannafin JA. The effect of ciprofloxa- cin on tendon, paratenon, and capsular fibroblast metabolism. Am J
Sports Med. 2000;28:364-9.
124. Murphy G, Knauper V, Atkinson S, Butler G, English W, Hutton M, Stracke J, Clark I. Matrix metalloproteinases in arthritic disease. Arthritis Res. 2002;4
Suppl 3:S39-49.
125. Nagase H, Woessner JF. Matrix metalloproteinases. J Biol Chem. 1999;274:21491-4.
126. Vu TH, Werb Z. Matrix metalloproteinases: effectors of development and normal physiology. Genes Dev. 2000;14:2123-33.
127. Birkedal-Hansen H. Proteolytic remodeling of extracellular matrix. Curr Opin Cell Biol. 1995;7:728-35.
128. Ireland D, Harrall R, Curry V, Holloway G, Hackney R, Hazleman B, Riley G. Multiple changes in gene expression in chronic human Achilles tendinopathy.
Ma- trix Biol. 2001;20:159-69.
129. Alfredson H, Lorentzon M, Backman S, Backman A, Lerner UH. cDNA-arrays and real-time quantitative PCR techniques in the investigation of chronic
Achilles tendinosis. J Orthop Res. 2003;21:970-5.
130. Riley GP, Curry V, DeGroot J, van El B, Verzijl N, Hazleman BL, Bank RA. Ma- trix metalloproteinase activities and their relationship with collagen
remodelling in tendon pathology. Matrix Biol. 2002;21:185-95.
131. Choi HR, Kondo S, Hirose K, Ishiguro N, Hasegawa Y, Iwata H. Expression and enzymatic activity of MMP-2 during healing process of the acute
supraspina- tus tendon tear in rabbits. J Orthop Res. 2002;20:927-33.
132. Leadbetter WB. Cell-matrix response in tendon injury. Clin Sports Med. 1992;11:533-78.
133. Puddu G, Ippolito E, Postacchini F. A classification of Achilles tendon dis- ease. Am J Sports Med. 1976;4:145-50.
134. Maffulli N, Khan KM, Puddu G. Overuse tendon conditions: time to change a confusing terminology. Arthroscopy. 1998;14:840-3.
135. Khan KM, Maffulli N. Tendinopathy: an Achilles’ heel for athletes and clini- cians. Clin J Sport Med. 1998;8:151-4.
136. Movin T, Gad A, Reinholt FP, Rolf C. Tendon pathology in long-standing achill- odynia. Biopsy findings in 40 patients. Acta Orthop Scand. 1997;68:170-5.
137. Astrom M, Rausing A. Chronic Achilles tendinopathy. A survey of surgical and histopathologic findings. Clin Orthop. 1995;316:151-64.
138. Maffulli N, Barrass V, Ewen SW. Light microscopic histology of achilles ten- don ruptures. A comparison with unruptured tendons. Am J Sports Med.
2000;28:857-63.
139. Jarvinen M, Jozsa L, Kannus P, Jarvinen TL, Kvist M, Leadbetter W. Histo- pathological findings in chronic tendon disorders. Scand J Med Sci Sports.
1997;7:86-95.
140. Burry HC, Pool CJ. Central degeneration of the achilles tendon. Rheumatol- ogy Rehabilitation. 1973;12:177-81.
141. Burry HC, Pool CJ. The pathology of the painful heel. Br J Sports Med. 1971;6:9-12.
142. Colosimo AJ, Bassett FH 3rd. Jumper’s knee. Diagnosis and treatment. Or- thop Rev. 1990;19:139-49.
143. Fritschy D, Wallensten R. Surgical treatment of patellar tendinitis. Knee Surg Sports Traumatol Arthrosc. 1993;1:131-3.
144. Cook JL, Khan KM, Harcourt PR, Grant M, Young DA, Bonar SF. A cross sec- tional study of 100 athletes with jumper’s knee managed conservatively
and sur- gically. The Victorian Institute of Sport Tendon Study Group. Br J Sports Med. 1997;31:332-6.
145. Raatikainen T, Karpakka J, Puranen J, Orava S. Operative treatment of par- tial rupture of the patellar ligament. A study of 138 cases. Int J Sports Med.
1994;15:46-9.
146. Yu JS, Popp JE, Kaeding CC, Lucas J. Correlation of MR imaging and patho- logic findings in athletes undergoing surgery for chronic patellar tendinitis.
AJR Am J Roentgenol. 1995;165:115-8.
147. Fukuda H, Hamada K, Yamanaka K. Pathology and pathogenesis of bursal- side rotator cuff tears viewed from en bloc histologic sections. Clin Orthop.
1990;254:75-80.
148. Cole AS, Cordiner-Lawrie S, Carr AJ, Athanasou NA. Localised deposi- tion of amyloid in tears of the rotator cuff. J Bone Joint Surg Br. 2001; 83:561-4.
149. Khan KM, Cook JL, Bonar F, Harcourt P, Astrom M. Histopathology of com- mon tendinopathies. Update and implications for clinical management.
Sports Med. 1999;27:393-408.
150. Kvist M. Achilles tendon injuries in athletes. Ann Chir Gynaecol. 1991;
151. Almekinders LC, Temple JD. Etiology, diagnosis, and treatment of tendoni- tis: an analysis of the literature. Med Sci Sports Exerc. 1998;30:1183-90.
152. Jozsa L, Kannus P. Histopathological findings in spontaneous tendon rup- tures. Scand J Med Sci Sports. 1997;7:113-8.
153. Nelen G, Martens M, Burssens A. Surgical treatment of chronic Achilles tendinitis. Am J Sports Med. 1989;17:754-9.
154. Clancy WG Jr, Neidhart D, Brand RL. Achilles tendonitis in runners: a report of five cases. Am J Sports Med. 1976;4:46-57.
155. Cetti R, Junge J, Vyberg M. Spontaneous rupture of the Achilles tendon is preceded by widespread and bilateral tendon damage and ipsilateral
inflamma- tion: a clinical and histopathologic study of 60 patients. Acta Orthop Scand. 2003;74:78-84.
156. Alfredson H, Bjur D, Thorsen K, Lorentzon R, Sandstrom P. High intratendi- nous lactate levels in painful chronic Achilles tendinosis. An investigation
using microdialysis technique. J Orthop Res. 2002;20:934-8.
157. Alfredson H, Thorsen K, Lorentzon R. In situ microdialysis in tendon tissue: high levels of glutamate, but not prostaglandin E2 in chronic Achilles tendon
pain. Knee Surg Sports Traumatol Arthrosc. 1999;7:378-81.
158. Zubrzycka M, Janecka A. Substance P: transmitter of nociception (minire- view). Endocr Regul. 2000;34:195-201.
159. Ljung BO, Forsgren S, Friden J. Sympathetic and sensory innervations are heterogeneously distributed in relation to the blood vessels at the extensor
carpi radialis brevis muscle origin of man. Cells Tissues Organs. 1999;165:45-54.
160. Gotoh M, Hamada K, Yamakawa H, Inoue A, Fukuda H. Increased substance P in subacromial bursa and shoulder pain in rotator cuff diseases. J Orthop
Res. 1998;16:618-21.
161. Ackermann PW, Finn A, Ahmed M. Sensory neuropeptidergic pattern in ten- don, ligament and joint capsule. A study in the rat. Neuroreport.
1999;13:2055-60.
162. Ljung BO, Alfredson H, Forsgren S. Neurokinin 1-receptors and sensory neuropeptides in tendon insertions at the medial and lateral epicondyles of the
humerus. Studies on tennis elbow and medial epicondylalgia. J Orthop Res. 2004;22:321-7.
163. Ackermann PW, Spetea M, Nylander I, Ploj K, Ahmed M, Kreicbergs A. An opioid system in connective tissue: a study of achilles tendon in the rat. J
His- tochem Cytochem. 2001;49:1387-95.

7827
164. Brodin E, Gazelius B, Panopoulos P, Olgart L. Morphine inhibits substance P release from peripheral sensory nerve endings. Acta Physiol Scand. 1983;1
17:567-70.
165. Yaksh TL. Substance P release from knee joint afferent terminals: modula- tion by opioids. Brain Res. 1988;458:319-24.
166. Murphy PG, Loitz BJ, Frank CB, Hart DA. Influence of exogenous growth fac- tors on the synthesis and secretion of collagen types I and III by explants
of nor- mal and healing rabbit ligaments. Biochem Cell Biol. 1994;72:403-9.
167. Oakes BW. Tissue healing and repair: tendons and ligaments. In: Frontera WR, editor. Rehabilitation of sports injuries: scientific basis. Boston: Blackwell
Science; 2003. p 56-98.
168. Tillman LJ, Chasan NP. Properties of dense connective tissue and wound healing. In: Hertling D, Kessler RM, editors. Management of common musculo-
skeletal disorders: physical therapy principles and methods. 3rd ed. Philadelphia: Lippincott; 1996. p 8-21.
169. Hooley CJ, Cohen RE. A model for the creep behaviour of tendon. Int J Biol Macromol. 1979;1:123-32.
170. Abrahamsson SO. Matrix metabolism and healing in the flexor tendon. Ex- perimental studies on rabbit tendon. Scand J Plast Reconstr Surg Hand Surg
Suppl. 1991;23:1-51.
171. Farkas LG, McCain WG, Sweeney P, Wilson W, Hurst LN, Lindsay WK. An exper- imental study of changes following silastic rod preparation of a new
tendon sheath and subsequent tendon grafting. J Bone Joint Surg Am. 1973;55:1149-58.
172. Amiel D, Akeson W, Harwood FL, Frank CB. Stress deprivation effect on met- abolic turnover of medial collateral ligament collagen. A comparison
between nine- and 12-week immobilization. Clin Orthop. 1983;172:265-70.
173. Manske PR, Lesker PA. Biochemical evidence of flexor tendon participation in the repair process—an in vitro study. J Hand Surg [Br]. 1984;9:117-20.
174. Gelberman RH, Manske PR, Vande Berg JS, Lesker PA, Akeson WH. Flexor tendon repair in vitro: a comparative histologic study of the rabbit, chicken,
dog, and monkey. J Orthop Res. 1984;2:39-48.
175. Potenza AD. Tendon healing within the flexor digital sheath in the dog. Am J Orthop. 1962;44:49-64.
176. Gelberman RH, Amiel D, Harwood F. Genetic expression for type I procol- lagen in the early stages of flexor tendon healing. J Hand Surg [Am]. 1992;
17:551-8.
177. Garner WL, McDonald JA, Kuhn C 3rd, Weeks PM. Autonomous healing of chicken flexor tendons in vitro. J Hand Surg [Am]. 1988;13:697-700.
178. Manske PR, Gelberman RH, Lesker PA. Flexor tendon healing. Hand Clin. 1985;1:25-34.
179. Mast BA, Haynes JH, Krummel TM, Diegelmann RF, Cohen IK. In vivo degra- dation of fetal wound hyaluronic acid results in increased fibroplasia,
collagen deposition, and neovascularization. Plast Reconstr Surg. 1992;89:503-9.
180. Gelberman RH, Manske PR, Akeson WH, Woo SL, Lundborg G, Amiel D. Flexor tendon repair. J Orthop Res. 1986;4:119-28.
181. Tokita Y, Yamaya A, Yabe Y. [An experimental study on the repair and restora- tion of gliding function after digital flexor tendon injury. I. Repair of the
sutured digital flexor tendon within digital sheath]. Nippon Seikeigeka Gakkai Zasshi. 1974;48:107-127. Japanese.
182. Fujita M, Hukuda S, Doida Y. [Experimental study of intrinsic healing of the flexor tendon: collagen synthesis of the cultured flexor tendon cells of the
ca- nine]. Nippon Seikeigeka Gakkai Zasshi. 1992;66:326-33. Japanese.
183. Ingraham JM, Hauck RM, Ehrlich HP. Is the tendon embryogenesis process resurrected during tendon healing? Plast Reconstr Surg. 2003;112:844-54.
184. Lundborg G, Rank F. Experimental studies on cellular mechanisms involved in healing of animal and human flexor tendon in synovial environment. Hand.
1980;12:3-11.
185. Lundborg G, Hansson HA, Rank F, Rydevik B. Superficial repair of severed flexor tendons in synovial environment. An experimental, ultrastructural study
on cellular mechanisms. J Hand Surg [Am]. 1980;5:451-61.
186. Russell JE, Manske PR. Collagen synthesis during primate flexor tendon re- pair in vitro. J Orthop Res. 1990;8:13-20.
187. Becker H, Graham MF, Cohen IK, Diegelmann RF. Intrinsic tendon cell prolif- eration in tissue culture. J Hand Surg [Am]. 1981;6:616-9.
188. Koob TJ. Biomimetic approaches to tendon repair. Comp Biochem Physiol A Mol Integr Physiol. 2002;133:1171-92.
189. Klein MB, Pham H, Yalamanchi N, Chang J. Flexor tendon wound healing in vitro: the effect of lactate on tendon cell proliferation and collagen
production. J Hand Surg [Am]. 2001;26:847-54. Erratum in: J Hand Surg [Am]. 2002; 27:740.
190. Riederer-Henderson MA, Gauger A, Olson L, Robertson C, Greenlee TK Jr. Attachment and extracellular matrix differences between tendon and synovial
fi- broblastic cells. In Vitro. 1983;19:127-33.
191. Koob TJ, Summers AP. Tendon—bridging the gap. Comp Biochem Physiol A Mol Integr Physiol. 2002;133:905-9.
192. Strickland JW. Flexor tendons: acute injuries. In: Green DP, Hotchkiss RN, Pedersen WC, editors. Green’s operative hand surgery. 4th ed. New York:
Churchill Livingstone; 1999. p 1851-97.
193. Uhthoff HK, Sarkar K. Surgical repair of rotator cuff ruptures. The impor- tance of the subacromial bursa. J Bone Joint Surg Br. 1991;73:399-401.
194. Oshiro W, Lou J, Xing X, Tu Y, Manske PR. Flexor tendon healing in the rat: a histologic and gene expression study. J Hand Surg [Am]. 2003;28:814-23.
195. Evans CH. Cytokines and the role they play in the healing of ligaments and tendons. Sports Med. 1999;28:71-6.
196. Sciore P, Boykiw R, Hart DA. Semiquantitative reverse transcription-poly- merase chain reaction analysis of mRNA for growth factors and growth factor
re- ceptors from normal and healing rabbit medial collateral ligament tissue. J Orthop Res. 1998;16:429-37.
197. Chang J, Most D, Stelnicki E, Siebert JW, Longaker MT, Hui K, Lineaweaver WC. Gene expression of transforming growth factor beta-1 in rabbit zone II
flexor tendon wound healing: evidence for dual mechanisms of repair. Plast Reconstr Surg. 1997;100:937-44.
198. Chang J, Most D, Thunder R, Mehrara B, Longaker MT, Lineaweaver WC. Mo- lecular studies in flexor tendon wound healing: the role of basic fibroblast
growth factor gene expression. J Hand Surg [Am]. 1998;23:1052-8.
199. Woo SL, Hildebrand K, Watanabe N, Fenwick JA, Papageorgiou CD, Wang JH. Tissue engineering of ligament and tendon healing. Clin Orthop. 1999;367
Suppl:S312-23.
200. Natsu-ume T, Nakamura N, Shino K, Toritsuka Y, Horibe S, Ochi T. Temporal and spatial expression of transforming growth factor-beta in the healing
patellar ligament of the rat. J Orthop Res. 1997;15:837-43.
201. 2001;19:136-42.
202. Ackermann PW. Peptidergic innervation of periarticular tissue [thesis]. Stockholm, Sweden: Karolinska Institute; 2001.
203. Nakamura-Craig M, Smith TW. Substance P and peripheral inflammatory hyperalgesia. Pain. 1989;38:91-8.
204. Maggi CA. Tachykinins and calcitonin gene-related peptide (CGRP) as co- transmitters released from peripheral endings of sensory nerves. Prog
Neurobiol. 1995;45:1-98.
205. Brain SD, Williams TJ, Tippins JR, Morris HR, MacIntyre I. Calcitonin gene- related peptide is a potent vasodilator. Nature. 1985;313:54-6.
206. Vasko MR, Campbell WB, Waite KJ. Prostaglandin E2 enhances bradykinin- stimulated release of neuropeptides from rat sensory neurons in culture. J
Neuro- sci. 1994;14:4987-97.
207. Schaible HG, Grubb BD. Afferent and spinal mechanisms of joint pain. Pain. 1993;55:5-54.
208. Manske PR. Flexor tendon healing. J Hand Surg [Br]. 1988;13:237-45.
209. Bruns J, Kampen J, Kahrs J, Plitz W. Achilles tendon rupture: experimental results on spontaneous repair in a sheep-model. Knee Surg Sports Traumatol
Ar- throsc. 2000;8:364-9.
210. Wang CJ, Wang FS, Yang KD, Weng LH, Hsu CC, Huang CS, Yang LC. Shock wave therapy induces neovascularization at the tendon-bone junction. A
study in rabbits. J Orthop Res. 2003;21:984-9.
211. Chen YJ, Wang CJ, Yang KD, Kuo YR, Huang HC, Huang YT, Sun YC, Wang FS. Extracorporeal shock waves promote healing of collagenase-induced
Achilles tendinitis and increase TGF-beta1 and IGF-I expression. J Orthop Res. 2004;22:854-61.
212. Speed CA, Richards C, Nichols D, Burnet S, Wies JT, Humphreys H, Hazle- man BL. Extracorporeal shock-wave therapy for tendonitis of the rotator cuff.
A double-blind, randomised, controlled trial. J Bone Joint Surg Br. 2002;84:509-12.
213. Gerdesmeyer L, Wagenpfeil S, Haake M, Maier M, Loew M, Wortler K, Lampe R, Seil R, Handle G, Gassel S, Rompe JD. Extracorporeal shock wave
ther- apy for the treatment of chronic calcifying tendonitis of the rotator cuff: a random- ized controlled trial. JAMA. 2003;290:2573-80.
214. Rompe JD, Kirkpatrick CJ, Kullmer K, Schwitalle M, Krischek O. Dose- related effects of shock waves on rabbit tendo Achillis. A sonographic and histo-
logical study. J Bone Joint Surg Br. 1998;80:546-52.
215. Lee EW, Maffulli N, Li CK, Chan KM. Pulsed magnetic and electromagnetic fields in experimental achilles tendonitis in the rat: a prospective randomized
study. Arch Phys Med Rehabil. 1997;78:399-404.
216. Owoeye I, Spielholz NI, Fetto J, Nelson AJ. Low-intensity pulsed galvanic cur- rent and the healing of tenotomized rat achilles tendons: preliminary report
using load-to-breaking measurements. Arch Phys Med Rehabil. 1987;68:415-8.
217. Fujita M, Hukuda S, Doida Y. The effect of constant direct electrical cur- rent on intrinsic healing in the flexor tendon in vitro. An ultrastructural study
of differing attitudes in epitenon cells and tenocytes. J Hand Surg [Br]. 1992; 17:94-8.

7928
218. Greenough CG. The effect of pulsed electromagnetic fields on flexor tendon healing in the rabbit. J Hand Surg [Br]. 1996;21:808-12.
219. Reddy GK, Stehno-Bittel L, Enwemeka CS. Laser photostimulation of collagen production in healing rabbit Achilles tendons. Lasers Surg Med. 1998;22:281-7.
220. Ozkan N, Altan L, Bingol U, Akln S, Yurtkuran M. Investigation of the supple- mentary effect of GaAs laser therapy on the rehabilitation of human digital flexor tendons.
J Clin Laser Med Surg. 2004;22:105-10.
221. Tasto JP, Cummings J, Medlock V, Harwood F, Hardesty R, Amiel D. The ten- don treatment center: new horizons in the treatment of tendinosis. Arthroscopy. 2003;19
Suppl 1:213-23.
222. Chang J, Most D, Stelnicki E, Siebert JW, Longaker MT, Hui K, Lineaweaver WC. Gene expression of transforming growth factor beta-1 in rabbit zone II flexor tendon
wound healing: evidence for dual mechanisms of repair. Plast Reconstr Surg. 1997;100:937-44.
223. Banes AJ, Tsuzaki M, Hu P, Brigman B, Brown T, Almekinders L, Lawrence WT, Fischer T. PDGF-BB, IGF-I and mechanical load stimulate DNA synthesis in avian
tendon fibroblasts in vitro. J Biomech. 1995;28:1505-13.
224. Ghahary A, Shen YJ, Scott PG, Gong Y, Tredget EE. Enhanced expression of mRNA for transforming growth factor-beta, type I and type III procollagen in hu- man post-
burn hypertrophic scar tissues. J Lab Clin Med. 1993;122:465-73.
225. Peltonen J, Hsiao LL, Jaakkola S, Sollberg S, Aumailley M, Timpl R, Chu ML, Uitto J. Activation of collagen gene expression in keloids: co-localization of type I and VI
collagen and transforming growth factor-beta 1 mRNA. J Invest Dermatol. 1991;97:240-8.
226. Abrahamsson SO, Lohmander S. Differential effects of insulin-like growth factor-I on matrix and DNA synthesis in various regions and types of rabbit ten- dons. J Orthop
Res. 1996;14:370-6.
227. Chang J, Thunder R, Most D, Longaker MT, Lineaweaver WC. Studies in flexor tendon wound healing: neutralizing antibody to TGF-beta1 increases post- operative
range of motion. Plast Reconstr Surg. 2000;105:148-55.
228. Tsuzaki M, Brigman BE, Yamamoto J, Lawrence WT, Simmons JG, Mohap- atra NK, Lund PK, Van Wyk J, Hannafin JA, Bhargava MM, Banes AJ. Insulin-like growth
factor-I is expressed by avian flexor tendon cells. J Orthop Res. 2000;18:546-56.
229. Murphy DJ, Nixon AJ. Biochemical and site-specific effects of insulin-like growth factor I on intrinsic tenocyte activity in equine flexor tendons. Am J Vet Res.
1997;58:103-9.
230. Dahlgren LA, van der Meulen MC, Bertram JE, Starrak GS, Nixon AJ. Insulin- like growth factor-I improves cellular and molecular aspects of healing in a colla- genase-
induced model of flexor tendinitis. J Orthop Res. 2002;20:910-9.
231. Dowling BA, Dart AJ, Hodgson DR, Rose RJ, Walsh WR. Recombinant equine growth hormone does not affect the in vitro biomechanical properties of equine superficial
digital flexor tendon. Vet Surg. 2002;31:325-30.
232. Ferrara N. Role of vascular endothelial growth factor in the regulation of an- giogenesis. Kidney Int. 1999;56:794-814.
233. Neufeld G, Cohen T, Gengrinovitch S, Poltorak Z. Vascular endothelial growth factor (VEGF) and its receptors. FASEB J. 1999;13:9-22.
234. Pufe T, Petersen W, Tillmann B, Mentlein R. The angiogenic peptide vascular endothelial growth factor is expressed in foetal and ruptured tendons. Virchows Arch.
2001;439:579-85.
235. Bidder M, Towler DA, Gelberman RH, Boyer MI. Expression of mRNA for vas- cular endothelial growth factor at the repair site of healing canine flexor tendon. J Orthop
Res. 2000;18:247-52.
236. Boyer MI, Watson JT, Lou J, Manske PR, Gelberman RH, Cai SR. Quantita- tive variation in vascular endothelial growth factor mRNA expression during early flexor
tendon healing: an investigation in a canine model. J Orthop Res. 2001; 19:869-72.
237. Yang R, Thomas GR, Bunting S, Ko A, Ferrara N, Keyt B, Ross J, Jin H. Ef- fects of vascular endothelial growth factor on hemodynamics and cardiac perfor- mance. J
Cardiovasc Pharmacol. 1996;27:838-44.
238. Zhang F, Liu H, Stile F, Lei MP, Pang Y, Oswald TM, Beck J, Dorsett-Martin W, Lineaweaver WC. Effect of vascular endothelial growth factor on rat Achilles ten- don
healing. Plast Reconstr Surg. 2003;112:1613-9.
239. Chang SC, Hoang B, Thomas JT, Vukicevic S, Luyten FP, Ryba NJ, Kozak CA, Reddi AH, Moos M Jr. Cartilage-derived morphogenetic proteins. New members of the
transforming growth factor-beta superfamily predominantly expressed in long bones during human embryonic development. J Biol Chem. 1994;269:28227-34.
240. Wolfman NM, Hattersley G, Cox K, Celeste AJ, Nelson R, Yamaji N, Dube JL, DiBlasio-Smith E, Nove J, Song JJ, Wozney JM, Rosen V. Ectopic induction of ten- don
and ligament in rats by growth and differentiation factors 5, 6, and 7, mem- bers of the TGF-beta gene family. J Clin Invest. 1997;100:321-30.
241. Mikic B, Schalet BJ, Clark RT, Gaschen V, Hunziker EB. GDF-5 deficiency in mice alters the ultrastructure, mechanical properties and composition of the Achilles
tendon. J Orthop Res. 2001;19:365-71.
242. Forslund C, Rueger D, Aspenberg P. A comparative dose-response study of cartilage-derived morphogenetic protein (CDMP)-1, -2 and -3 for tendon healing in rats. J
Orthop Res. 2003;21:617-21.
243. Forslund C, Aspenberg P. Improved healing of transected rabbit Achilles ten- don after a single injection of cartilage-derived morphogenetic protein-2. Am J Sports Med.
2003;31:555-9.
244. Nakamura N, Timmermann SA, Hart DA, Kaneda Y, Shrive NG, Shino K, Ochi T, Frank CB. A comparison of in vivo gene delivery methods for antisense therapy in
ligament healing. Gene Ther. 1998;5:1455-61.
245. Nakamura N, Shino K, Natsuume T, Horibe S, Matsumoto N, Kaneda Y, Ochi
T. Early biological effect of in vivo gene transfer of platelet-derived growth factor (PDGF)-B into healing patellar ligament. Gene Ther. 1998;5:1165-70.
246. Hannallah D, Peterson B, Lieberman JR, Fu FH, Huard J. Gene therapy in or- thopaedic surgery. Instr Course Lect. 2003;52:753-68.
247. Nakamura N, Horibe S, Matsumoto N, Tomita T, Natsuume T, Kaneda Y, Shino K, Ochi T. Transient introduction of a foreign gene into healing rat patellar ligament. J
Clin Invest. 1996;97:226-31.
248. Gerich TG, Kang R, Fu FH, Robbins PD, Evans CH. Gene transfer to the rab- bit patellar tendon: potential for genetic enhancement of tendon and ligament healing.
Gene Ther. 1996;3:1089-93.
249. Gerich TG, Kang R, Fu FH, Robbins PD, Evans CH. Gene transfer to the pa- tellar tendon. Knee Surg Sports Traumatol Arthrosc. 1997;5:118-23.
250. Lou J, Kubota H, Hotokezaka S, Ludwig FJ, Manske PR. In vivo gene transfer and overexpression of focal adhesion kinase (pp125 FAK) mediated by recombi- nant
adenovirus-induced tendon adhesion formation and epitenon cell change. J Orthop Res. 1997;15:911-8.
251. Wolfman NM, Celeste AJ, Cox K, Hattersley G, Nelson R, Yamaji N, DiBlasio- Smith E, Nove J, Song JJ, Wozney JM, Rosen V. Preliminary characterization of the
biological activities of rhBMP-12. J Bone Miner Res.1995;10:S148.
252. Fu SC, Wong YP, Chan BP, Pau HM, Cheuk YC, Lee KM, Chan KM. The roles of bone morphogenetic protein (BMP) 12 in stimulating the proliferation and ma- trix
production of human patellar tendon fibroblasts. Life Sci. 2003;72:2965-74.
253. Lou J, Tu Y, Burns M, Silva MJ, Manske P. BMP-12 gene transfer augmenta- tion of lacerated tendon repair. J Orthop Res. 2001;19:1199-202.
254. Nakamura N, Shino K, Natsuume T, Horibe S, Matsumoto N, Kaneda Y, Ochi
T. Early biological effect of in vivo gene transfer of platelet-derived growth factor (PDGF)-B into healing patellar ligament. Gene Ther. 1998;5:1165-70.
255. Marchant JK, Hahn RA, Linsenmayer TF, Birk DE. Reduction of type V collagen using a dominant-negative strategy alters the regulation of fibrillogenesis and results in
the loss of corneal-specific fibril morphology. J Cell Biol. 1996;135:1415-26.
256. Adachi E, Hayashi T. In vitro formation of hybrid fibrils of type V collagen and type I collagen. Limited growth of type I collagen into thick fibrils by type V col- lagen.
Connect Tissue Res. 1986;14:257-66.
257. Niyibizi C, Kavalkovich K, Yamaji T, Woo SL. Type V collagen is increased dur- ing rabbit medial collateral ligament healing. Knee Surg Sports Traumatol Ar- throsc.
2000;8:281-5.
258. Shimomura T, Jia F, Niyibizi C, Woo SL. Antisense oligonucleotides reduce synthesis of procollagen alpha1 (V) chain in human patellar tendon fibroblasts: potential
application in healing ligaments and tendons. Connect Tissue Res. 2003;44:167-72.
259. Hart DA, Nakamura N, Marchuk L, Hiraoka H, Boorman R, Kaneda Y, Shrive NG, Frank CB. Complexity of determining cause and effect in vivo after antisense gene
therapy. Clin Orthop. 2000;379 Suppl:S242-51.
260. Nakamura N, Timmermann SA, Hart DA, Kaneda Y, Shrive NG, Shino K, Ochi T, Frank CB. A comparison of in vivo gene delivery methods for antisense therapy in
ligament healing. Gene Ther. 1998;5:1455-61.
261. Nakamura N, Hart DA, Boorman RS, Kaneda Y, Shrive NG, Marchuk LL, Shino K, Ochi T, Frank CB. Decorin antisense gene therapy improves functional healing of
early rabbit ligament scar with enhanced collagen fibrillogenesis in vivo. J Orthop Res. 2000;18:517-23.
262. Caplan AI. The mesengenic process. Clin Plast Surg. 1994;21:429-35.
263. Caplan AI, Bruder SP. Mesenchymal stem cells: building blocks for molecular medicine in the 21st century. Trends Mol Med. 2001;7:259-64.
264. Buckingham ME. Muscle: the regulation of myogenesis. Curr Opin Genet Dev. 1994;4:745-51.
265. Dennis JE, Charbord P. Origin and differentiation of human and murine stroma. Stem Cells. 2002;20:205-14.
266. Tontonoz P, Hu E, Spiegelman BM. Stimulation of adipogenesis in fibroblasts by PPAR gamma 2, a lipid-activated transcription factor. Cell. 1994;79:1147-56. Erratum
in: Cell. 1995;80:following 957.

8029
267. Fajas L, Fruchart JC, Auwerx J. Transcriptional control of adipogenesis. Curr Opin Cell Biol. 1998;10:165-73.
268. Ducy P, Karsenty G. Genetic control of cell differentiation in the skeleton. Curr Opin Cell Biol. 1998;10:614-9.
269. Satomura K, Krebsbach P, Bianco P, Gehron Robey P. Osteogenic imprinting up- stream of marrow stromal cell differentiation. J Cell Biochem.
2000;78:391-403.
270. Pelinkovic D, Lee JY, Engelhardt M, Rodosky M, Cummins J, Fu FH, Huard J. Muscle cell-mediated gene delivery to the rotator cuff. Tissue Eng.
2003;9:143-51.
271. Young RG, Butler DL, Weber W, Caplan AI, Gordon SL, Fink DJ. Use of mes- enchymal stem cells in a collagen matrix for Achilles tendon repair. J Orthop
Res. 1998;16:406-13.
272. Awad HA, Butler DL, Boivin GP, Smith FN, Malaviya P, Huibregtse B, Caplan AI. Autologous mesenchymal stem cell-mediated repair of tendon. Tissue
Eng. 1999;5:267-77.
273. Cao Y, Liu Y, Liu W, Shan Q, Buonocore SD, Cui L. Bridging tendon defects using autologous tenocyte engineered tendon in a hen model. Plast Reconstr
Surg. 2002;110:1280-9.
274. Powers SK, Howley ET. Exercise physiology: theory and application to fitness and performance. 4th ed. Boston: McGraw Hill; 2001.
275. Baechle TR, Earle R. Essentials of strength training and conditioning. Cham- paign, IL: Human Kinetics; 2000.
276. Jaibaji M. Advances in the biology of zone II flexor tendon healing and adhe- sion formation. Ann Plast Surg. 2000;45:83-92.
277. Ippolito E, Natali PG, Postacchini F, Accinni L, De Martino C. Ultrastructural and immunochemical evidence of actin in the tendon cells. Clin Orthop.
1977;126:282-4.
278. Murray MM, Spector M. Fibroblast distribution in the anteromedial bundle of the human anterior cruciate ligament: the presence of alpha-smooth muscle
ac- tin-positive cells. J Orthop Res. 1999;17:18-27.
279. Schurch W, Seemayer TA, Gabbiani G. The myofibroblast: a quarter century after its discovery. Am J Surg Pathol. 1998;22:141-7.
280. Serini G, Gabbiani G. Mechanisms of myofibroblast activity and phenotypic modulation. Exp Cell Res. 1999;250:273-83.
281. Weiler A, Unterhauser FN, Bail HJ, Huning M, Haas NP. Alpha-smooth muscle actin is expressed by fibroblastic cells of the ovine anterior cruciate
ligament and its free tendon graft during remodeling. J Orthop Res. 2002;20:310-7.
282. Hunter JM, Salisbury RE. Flexor-tendon reconstruction in severely damaged hands. A two-stage procedure using a silicone-dacron reinforced gliding
prosthe- sis prior to tendon grafting. J Bone Joint Surg Am. 1971;53:829-58.
283. Nishimura K, Nakamura RM, diZerega GS. Ibuprofen inhibition of postsurgi- cal adhesion formation: a time and dose response biochemical evaluation
in rab- bits. J Surg Res. 1984;36:115-24.
284. Nishimura K, Shimanuki T, diZerega GS. Ibuprofen in the prevention of ex- perimentally induced postoperative adhesions. Am J Med. 1984;77:102-6.
285. Szabo RM, Younger E. Effects of indomethacin on adhesion formation after repair of zone II tendon lacerations in the rabbit. J Hand Surg [Am].
1990;15:480- 3. Erratum in: J Hand Surg [Am]. 1990;15:802.
292. Hagberg L, Heinegard D, Ohlsson K. The contents of macromolecule solutes in flexor tendon sheath fluid and their relation to synovial fluid. A quantitative
analysis. J Hand Surg [Br]. 1992;17:167-71.
293. Hagberg L, Gerdin B. Sodium hyaluronate as an adjunct in adhesion preven- tion after flexor tendon surgery in rabbits. J Hand Surg [Am].1992;17:935-41.
294. Tuncay I, Ozbek H, Atik B, Ozen S, Akpinar F. Effects of hyaluronic acid on postoperative adhesion of tendo calcaneus surgery: an experimental study
in rats. J Foot Ankle Surg. 2002;41:104-8.
295. Wiig M, Abrahamsson SO, Lundborg G. Tendon repair—cellular activities in rabbit deep flexor tendons and surrounding synovial sheaths and the effects
of hyaluronan: an experimental study in vivo and in vitro. J Hand Surg [Am]. 1997; 22:818-25.
296. Khan U, Occleston NL, Khaw PT, McGrouther DA. Single exposures to 5- fluorouracil: a possible mode of targeted therapy to reduce contractile scarring
in the injured tendon. Plast Reconstr Surg. 1997;99:465-71.
297. Khan U, Kakar S, Akali A, Bentley G, McGrouther DA. Modulation of the for- mation of adhesions during the healing of injured tendons. J Bone Joint
Surg Br. 2000;82:1054-8.
298. Moran SL, Ryan CK, Orlando GS, Pratt CE, Michalko KB. Effects of 5- fluorouracil on flexor tendon repair. J Hand Surg [Am]. 2000;25:242-51.
299. Kannus P, Józsa KP, Renstrom P, Järvinen M, Kvist M, Lehto M, Oja P, Vuori I. The effects of training, immobilization and remobilization on musculoskeletal
tis- sue. 1: training and immobilization. Scand J Med Sci Sports. 1992;2:100-18.
300. Kannus P, Jozsa L, Natri A, Jarvinen M. Effects of training, immobilization and remobilization on tendons. Scand J Med Sci Sports. 1997;7:67-71.
301. Maffulli N, King JB. Effects of physical activity on some components of the skeletal system. Sports Med. 1992;13:393-407.
302. Butler DL, Grood ES, Noyes FR, Zernicke RF. Biomechanics of ligaments and tendons. Exerc Sport Sci Rev. 1978;6:125-81.
303. Yamamoto E, Hayashi K, Yamamoto N. Mechanical properties of collagen fascicles from stress-shielded patellar tendons in the rabbit. Clin Biomech
(Bris- tol, Avon). 1999;14:418-25.
304. Akeson WH, Woo SL, Amiel D, Coutts RD, Daniel D. The connective tissue response to immobility: biochemical changes in periarticular connective tissue
of the immobilized rabbit knee. Clin Orthop. 1973;93:356-62.
305. Akeson WH, Amiel D, Mechanic GL, Woo SL, Harwood FL, Hamer ML. Col- lagen cross-linking alterations in joint contractures: changes in the reducible
cross-links in periarticular connective tissue collagen after nine weeks of immobi- lization. Connect Tissue Res. 1977;5:15-9.
306. Kellett J. Acute soft tissue injuries—a review of the literature. Med Sci Sports Exerc. 1986;18:489-500.
307. Houglum P. Soft tissue healing and its impact on rehabilitation. J Sports Rehabil. 1992;1:19-39.
308. Almekinders LC, Baynes AJ, Bracey LW. An in vitro investigation into the effects of repetitive motion and nonsteroidal antiinflammatory medication on
human tendon fibroblasts. Am J Sports Med. 1995;23:119-23.
309. Zeichen J, van Griensven M, Bosch U. The proliferative response of isolated human tendon fibroblasts to cyclic biaxial mechanical strain. Am J Sports
Med. 2000;28:888-92.
310. Tanaka H, Manske PR, Pruitt DL, Larson BJ. Effect of cyclic tension on lacer- ated flexor tendons in vitro. J Hand Surg [Am]. 1995;20:467-73.
311. Nabeshima Y, Grood ES, Sakurai A, Herman JH. Uniaxial tension inhibits ten- don collagen degradation by collagenase in vitro. J Orthop Res.
1996;14:123-30.
312. Buckwalter JA. Activity vs. rest in the treatment of bone, soft tissue and joint injuries. Iowa Orthop J. 1995;15:29-42.
313. Buckwalter JA. Effects of early motion on healing of musculoskeletal tis- sues. Hand Clin. 1996;12:13-24.
314. Chow JA, Thomes LJ, Dovelle S, Monsivais J, Milnor WH, Jackson JP. Con- trolled motion rehabilitation after flexor tendon repair and grafting. A multi-
centre study. J Bone Joint Surg Br. 1988;70:591-5.
315. Cullen KW, Tolhurst P, Lang D, Page RE. Flexor tendon repair in zone 2 fol- lowed by controlled active mobilisation. J Hand Surg [Br]. 1989;14:392-5.
316. Elliot D, Moiemen NS, Flemming AF, Harris SB, Foster AJ. The rupture rate of acute flexor tendon repairs mobilized by the controlled active motion
regimen. J Hand Surg [Br]. 1994;19:607-12.
317. Banes AJ, Horesovsky G, Larson C, Tsuzaki M, Judex S, Archambault J, Zer- nicke R, Herzog W, Kelley S, Miller L. Mechanical load stimulates expression
of novel genes in vivo and in vitro in avian flexor tendon cells. Osteoarthritis Carti- lage. 1999;7:141-53.

8130
Treatment of chronic tendinopathies with peritendinous hyaluronan injec-
tions under sonographic guidance – an interventional, prospective,
single-arm, multicenter study (pag. 26)

References

1. Alfredson H. Chronic midportion Achilles tendinopathy: an update on research and treatment. Clin Sports Med 2003, 22: 727–741
2. Knobloch K. Aus nach Sportverletzungen? Moderne Diagnostik, Therapie und Präventionsmöglichkeiten. [Out of action after
sports injuries? Modern diagnostics, treatment and prevention options]. Balingen: Spitta, 2009
3. Sharma P, Maffulli N. Tendon injury and tendinopathy: healing and repair. J Bone Joint Surg Am 2005; 87: 187–202
4. Alfredson H, Lorentzon R. Chronic tendon pain: no signs of chemical inflammation but high concentrations of the neurotransmit-
ter glutamate. Implications for treatment? Curr Drug Targets 2002; 3: 43–54
5. Langberg H, Ellingsgaard H, Madsen T et al. Eccentric rehabilitation exercise increases peritendinous type 1 collagen synthesis
in humans with Achilles tendinosis. Scand J Med Sci Sports 2007; 17: 61–66
6. Meloni F, Milia F, Cavazzuti M et al. Clinical evaluation of sodium hyaluronate in the treatment of patients with supraspinatus
tendinosis under echographic guide: experimental study of periarticular injections. Eur J Radiol 2008; 68: 170–173
7. Petrella RJ, Cogliano A, Decaria J, Mohamed N, Lee R. Management of tennis elbow with sodium hyaluronate periarticular injec-
tions. Sports Medicine, Arthroscopy, Rehabilitation, Therapy & Technology 2010, 2: 1–6
8. Petrella MJ, Cogliano A, Petrella RJ. Original research: long-term efficacy and safety of periarticular hyaluronic acid in acute ankle
sprain. Phys Sportsmed 2009; 37: 64–70
9. Amadio PC. Friction of the gliding surface. Implications for tendon surgery and rehabilitation. J Hand Ther 2005, 18: 112–119
10. Akasaka T, Nishida J, Araki S, Shimamura T, Amadio PC, An KN. Hyaluronic acid diminishes the resistance to excursion after
flexor tendon repair: an in vitro biomechanical study. J Biomech 2005; 38: 503–507
11. Akasaka T, Nishida J, Imaeda T, Shimamura T, Amadio PC, An KN. Effect of hyaluronic acid on the excursion resistance of
tendon graft: a biomechanical in vitro study in a modified human model. Clin Biomech 2006; 21: 810–815
12. Momose T, Amadio PC, Zobitz ME, Zhao C, An KN. Effect of paratenon and repetitive motion on the gliding resistance of tendon
of extrasynovial origin. Clin Anat 2002; 15: 199–205.
13. Laurent TC, Fraser JR. Hyaluronan. FASEB J 1992; 6: 2397–2404
14. Abatangelo G, Cortivo R, Martelli M, Vecchia P. Cell detachment mediated by hyaluronic acid. Exp Cell Res 1982; 137: 73–78
15. Nicodemus GD, Bryant SJ. Cell encapsulation in biodegradable hydrogels for tissue engineering applications. Tissue Eng Part
B Rev. 2008; 14: 149–165
16. Hardwick C, Hoare K, Owens R et al. Molecular cloning of a novel hyaluronan receptor that mediates tumor cell motility. J Cell
Biol 1992; 117(6): 1343–1350
17. Toole BP, Turner RE, Banerjee SD. Hyaluronan- binding protein in chondrogenesis and angiogenesis in the developing limb.
Prog Clin Biol Res. 1993; 383B: 437–444
18. Huskisson EC, Donnelly S. Hyaluronic acid in the treatment of osteoarthritis of the knee. Rheumatology (Oxford) 1999; 38:
602–607
19. Huskisson EC. Measurement of pain. J Rheumatol 1982; 9: 768–769
20. van Ark M, Zwerver J, van den Akker- Scheek I. Injection treatments for patellar tendinopathy. Br J Sports Med 2011; 45:
1068–1076
21. Shrier I, Matheson GO, Kohl HW. Achilles tendonitis: are corticosteroid injections useful or harmful? Clin J Sport Med 1996; 6:
245–250
22. Balazs EA, Denlinger JL. Viscosupplementation: a new concept in the treatment of osteoarthritis. J Rheumatol Suppl 1993;
39: 3–9
23. Bellamy N, Campbell J, Robinson V, Gee T, Bourne R, Wells G. Viscosupplementation for the treatment of osteoarthritis of the
knee. Cochrane Database Syst Rev 2006; 19: CD 005321
24. Mendoza G, Álvarez AI, Pulido MM et. al. Inhibitory effects of different antioxidants on hyaluronan depolymerization. Carbohy-
drate Research 2007; 342: 96–102

82
31
Ultrasound-guided subacromial injections of sodium hyaluronate for the
management of rotator cuff tendinopathy: a prospective comparative
study with rehabilitation therapy (pag. 31)

References

1. Lewis JS (2009) Rotator cuff tendinopathy/subacromial impingement syndrome: is it time for a new method of assess- ment? Br J Sports Med 43:259–264
2. Herberts P, Kadefors R, Hogfors C, Sigholm G (1984) Shoulder pain and heavy manual labor. Clin Orthop 191:166–178
3. Vogel KG (2003) Tendon structure and response to changing mechanical load. J Musculoskelet Neuronal Interact 3:323–325
4. Loehr JF, Uhthoff HK (1987) The pathogenesis of degenerative rotator cuff tears. Orthop Trans 11:237
5. Bey MJ, Song HK, Wehrli FW et al (2002) Intratendinous strain fields of the intact supraspinatus tendon: the effect of glenohu- meral joint position and
tendon region. J Orthop Res 20:869–874
6. Nakajima T, Rokuuma N, Hamada K et al (1994) Histological and biomechanical characteristics of the supraspinatus tendon. J Should Elb Surg 3:79–87
7. Burkhart SS, Esch JC, Jolson RS (1993) The rotator crescent and rotator cable: an anatomic description of the shoulder’s ‘‘sus- pension bridge’’. Arthros-
copy 9:611–616
8. Codman E (1934) The shoulder: rupture of the supraspinatus tendon and other lesions in or about the subacromial bursa. Thomas Todd Company, Boston
9. Kibler WB (2003) Rehabilitation of rotator cuff tendinopathy. Clin Sports Med 22:837–847
10. Takagishi N, Minamikawa H, Iwamoto H, Kida H, Okuyama S, Tanaka M et al (1998) Clinical evaluation of SPH (high molec- ular weight sodium hyaluronate)
on periarthritis of the shoulder. Jpn J Pharmacol Ther 16:3553–3567
11. Iwata H (1993) Pharmacologic and clinical aspects of intraar- ticular injection of hyaluronate. Clin Orthop 289:285–291
12. Hakansson L, Hallgren R, Venge P (1980) Effect of hyaluronic acid on phagocytosis of opsonized latex particles. Scand J Immunol 11:649–653
13. Constant C, Murley A (1987) A clinical method of functional assessment of the shoulder. Clin Orthop 214:160–164
14. Dawson J, Fitzpatrick R, Carr A (1996) Questionnaire on the perceptions of patients about shoulder surgery. J Bone Joint Surg Br 78:593–600
15. Dawson J, Rogers K, Fitzpatrick R, Carr A (2009) The Oxford shoulder score revisited. Arch Orthop Trauma Surg 129:119–123
16. Petrella RJ, Petrella MJ, Cogliano A (2007) Periarticular hyalu- ronic acid in acute ankle sprain. Clin J Sport Med 17:251–257
17. Petrella R, Cogliano A, Decaria J, Mohamed N, Lee R (2010) Management of tennis elbow with sodium hyaluronate periar- ticular injections. Sports Med
Arthrosc Rehabill Ther Technol 2:4
18. Merolla G, Sperling JW, Paladini P, Porcellini G (2011) Efficacy of Hylan G-F 20 versus 6-methylprednisolone acetate in painful shoulder osteoarthritis: a
retrospective controlled trial. Muscul- oskelet Surg 95:215–224
19. Shibata Y, Modorikawa K, Emoto G, Naito M (2001) Clinical evaluation of sodium hyaluronate for the treatment of patients with rotator cuff tear. J Should
Elbow Surg 10:209–216
20. Ramonda R, Franceschini M, Leardini G (1998) Treatment of shoulder periarthritis with hyaluronic acid. Riv Ital Biol Med 1(suppl 2):51
21. Itokazu M, Matsunaga T (1995) Clinical evaluation of high- molecular-weight sodium hyaluronate for the treatment of patients with periarthritis of the
shoulder. Clin Ther 17:946–955
22. Rovetta G, Monteforte P (1998) Intraarticular injection of sodium hyaluronate plus steroid versus steroid in adhesive capsulitis of the shoulder. Int J Tissue
React 20:125–130
23. Blaine T, Moskowitz R, Udell J, Skyhar M, Levin R, Friedlander J, Daley M, Altman R (2008) Treatment of persistent shoulder pain with sodium hyaluronate:
a randomized, controlled trial. A multicenter study. J Bone Joint Surg Am 90:970–979
24. Kim YS, Park JY, Lee CS, Lee SJ (2012) Does hyaluronate injection work in shoulder disease in early stage? A multicenter, randomized, single blind and
open comparative clinical study. J Should Elb Surg 21:722–727
25. Penning LI, de Bie RA, Walenkamp GH (2012) The effectiveness of injections of hyaluronic acid or corticosteroid in patients with subacromial impingement:
a three-arm randomised controlled trial. J Bone Joint Surg Br 94:1246–1252
26. Littlewood C, Ashton J, Chance-Larsen K, May S, Sturrock B (2012) Exercises for rotator cuff tendinopathy: a systematic review. Physiotherapy 98(2):101–
109
27. Meloni F, Milia F, Cavazzuti M, Doria C, Lisai P, Profili S, Meloni GB (2008) Clinical evaluation of sodium hyaluronate in the treatment of patients with
sopraspinatus tendinosis under echographic guide: experimental study of periarticular injections. Eur J Rad 68:170–173
28. Crawshaw DP, Helliwell PS, Hensor EM, Hay EM, Aldous SJ, Conaghan PG (2010) Exercise therapy after corticosteroid injection for moderate to severe
shoulder pain: large pragmatic randomised trial. BMJ 340:c3037
29. Buchbinder R, Green S, Youd JM (2003) Corticosteroid injections for shoulder pain. Cochrane Database Syst Rev 1:CD004016

32
83
Effect of subacromial sodium hyaluronate injection on rotator cuff
disease : A double-blind placebo-controlled clinical trial (pag. 38)

References

1. Praemer AF, Rice D. Musculoskeletal conditions in the United States. Rosemont, IL. American Academy of Orthopaedic Surgeons; 1999;2:27-33.
2. Lin JC, Weintraub N, Aragaki DR. Nonsurgical treatment for rotator cuff injury in the elderly. J Am Med Dir Assoc 2008;9:626-32.
3. Andrews JR. Diagnosis and treatment of chronic painful shoulder: Review of nonsurgical interventions. Arthroscopy 2005;21:333-47.
4. Frontera WR, Silver JK, Rizzo TD. Rotator cuff tendinitis. Essentials of Physical Medicine and Rehabilitation. 2nd ed. Philadelphia: Saunders; 2008. p. 75.
5. Meloni F, Milia F, Cavazzuti M, Doria C, Lisai P, Profli S, et al. Clinical evaluation of sodium hyaluronate in the treatment of patients with sopraspinatus
tendinosis under echographic guide: Experimental study of periarticular injections. Eur J Radiol 2008;68:170-3.
6. Braddom RL. Peripheral joint and soft tissue injection techniques. Physical Medicine and Rehabilitation. 4th ed. Philadelphia: Saunders; 2011. p. 520
7. Costantino C, Olvirri S. Rehabilitative and infiltrative treatment with hyaluronic acid in elderly patients with rotator cuff tears. Acta Biomed 2009;80:225-9.
8. Blaine T, Moskowitz R, Udell J, Skyhar M, Levin R, Friedlander J, et al. Treatment of persistent shoulder pain with sodium hyaluronate: A randomized,
controlled trial. A multicenter study. J Bone Joint Surg Am 2008;90:970-9.
9. Silverstein E, Leger R, Shea KP. The use of intra-articular hylan G-F 20 in the treatment of symptomatic osteoarthritis of the shoulder: A preliminary study.
Am J Sports Med 2007;35:979-85.
10. Wu JJ, Shih LY, Hsu HC, Chen TH. The double-blind test of sodium hyaluronate (ARTZ) on osteoarthritis knee. Zhonghua Yi Xue Za Zhi (Taipei) 1997;59:99-
106.
11. Dahl LB, Dahl IM, Engström-Laurent A, Granath K. Concentration and molecular weight of sodium hyaluronate in synovial fluid from patients with rheu-
matoid arthritis and other arthropathies. Ann Rheum Dis 1985;44:817-22.
12. Itokazu M, Matsunaga T. Clinical evaluation of high-molecular-weight sodium
hyaluronate for the treatment of patients with periarthritis of the shoulder. Clin Ther 1995;17:946-55.
13. Takagishi N, Minamikawa H, Iwamoto H, Kida H, Okuyama S, Tanaka M, et al. Clinical evaluation of SPH (high molecular weight sodium hyaluronate) on
periarthritis of the shoulder. Jpn J Pharmacol Ther 1998;16:3553-67.
14. Yamamoto R, Namiki O, Iwata H, Shinmei M, Mikasa M, Ogawa N. Randomized cooperative study of sodium hyaluronate (SPH) on perarthritis scapulo-
humeralis. Jpn J Pharmacol Ther 1988;19:713-33.
15. Shibata Y, Midorikawa K, Emoto G, Naito M. Clinical evaluation of sodium hyaluronate for the treatment of patients with rotator cuff tear. J Shoulder Elbow
Surg 2001;10:209-16.
16. Adler RS, Sofka CM. Percutaneous ultrasound-guided injections in the musculoskeletal system. Ultrasound Q 2003;19:3-12.
17. Kluger R, Mayrhofer R, Kröner A, Pabinger C, Pärtan G, Hruby W, et al. Sonographic versus magnetic resonance arthrographic evaluation of full-thickness
rotator cuff tears in millimeters. J Shoulder Elbow Surg 2003;12:110-6.
18. Kim HA, Kim SH, Seo YI. Ultrasonographic fndings of painful shoulders and correlation between physical examination and ultrasonographic rotator cuff
tear. Mod Rheumatol 2007;17:213-9.
19. Teefey SA, Petersen B, Prather H. Shoulder Ultrasound vs MRI for rotator cuff pathology. PM R 2009;1:490-5.
20. Constant CR, Murley AH. A clinical method of functional assessment of the shoulder. Clin Orthop Relat Res 1987;214:160-4.
21. Bellamy N, Campbell J, Robinson V, Gee T, Bourne R, Wells G. Viscosupplementation for the treatment of osteoarthritis of the knee. Cochrane
Database Syst Rev 2006;19:CD005321.
22. Lo GH, LaValley M, McAlindon T, Felson DT. Intra-articular hyaluronic acid in treatment of knee osteoarthritis: A meta-analysis. JAMA 2003;290:3115-21.
23. Moreland LW. Intra-articular hyaluronan (hyaluronic acid) and hylans for the treatment of osteoarthritis: Mechanisms of action. Arthritis Res Ther
2003;5:54-67.
24. Chou WY, Ko JY, Wang FS, Huang CC, Wong T, Wang CJ, et al. Effect of sodium hyaluronate treatment on rotator cuff lesions without complete tears: A
randomized, double-blind, placebo-controlled study. J Shoulder Elbow Surg 2010;19:557-63.
25. Kim YS, Park JY, Lee CS, Lee SJ. Does hyaluronate injection work in shoulder disease in early stage? A multicenter, randomized, single blind and open
comparative clinical study. J Shoulder Elbow Surg 2012;21:722-7.
26. Ghosh P. The role of hyaluronic acid (hyaluronan) in health and disease: Interactions with cells, cartilage and components of synovial fluid. Clin Exp
Rheumatol 1994;12:75-82.

84
33
Hyaluronic acid and tendon lesions (pag. 47)
References
1. Kaux JF, Forthomme B, Le Goff C, Crielaard JM, Croisier JL. Current Opinions on Tendinopathy. J Sports Sci Med. 2011; 10:238-253.
2. Kaux JF, Crielaard JM. Tendon et tendinopathie. J Traumatol Sport. 2014;31:235-240.
3. Kaux JF, Drion P, Croisier JL, Crielaard JM. Tendinopathies and platelet-rich plasma (PRP): from pre-clinical experiments to therapeutic use. J Stem Cells
Regen Med. 2015;11:7-17.
4. Kaux JF, Janssen L, Drion P, et al. Vascular Endothelial Growth Factor-111 (VEGF-111) and tendon healing: preliminary results in a rat model of tendon
injury. Muscles Ligaments Tendons J. 2014;8:24-28.
5. Necas J, Bartosikova L, Brauner P, Kolar J. Hyaluronic acid (hyaluronan): a review, Veterinarni Medicina. 2008;53:397-411.
6. Jordan KM, Arden NK, Doherty M, et al. EULAR Recommendations 2003: an evidence based approach to the management of knee osteoarthritis: Report
of a Task Force of the Standing Committee for International Clinical Studies Including Therapeutic Trials (ESCISIT). Ann Rheum Dis. 2003;62:1145-1155.
7. Zhang W, Doherty M, Arden N, et al. EULAR evidence based recommendations for the management of hip osteoarthritis: report of a task force of the EULAR
Standing Committee for International Clinical Studies Including Therapeutics (ESCISIT). Ann Rheum Dis. 2005;64:669-681.
8. Zhang W, Moskowitz RW, Nuki G, et al. OARSI recommendations for the management of hip and knee osteoarthritis, part I: critical appraisal of existing
treatment guidelines and systematic review of current research evidence. Osteoarthritis Cartilage. 2007;15:981-1000.
9. Padulo S, Oliva F, Frizziero A, Maffulli N. Muscles, Ligaments and Tendons Journal. Basic principles and recommendations in clinical and field science
research. MLTJ. 2013;4:250-252.
10. Salti NI, Tuel RJ, Mass DP. Effect of hyaluronic acid on rabbit profundus flexor tendon healing in vitro. J Surg Res. 1993; 55:411-415.
11. Sun YL, Yang C, Amadio PC, Zhao C, Zobitz ME, An KN. Reducing friction by chemically modifying the surface of extrasynovial tendon grafts. J Orthop
Res. 2004;22:984-989.
12. Nishida J, Araki S, Akasaka T, Toba T, Shimamura T, Amadio PC, An KN. Effect of hyaluronic acid on the excursion resistance of tendon grafts. A biome-
chanical study in a canine model in vitro. J Bone Joint Surg Br. 2004;86:918-924.
13. Taguchi M, Zhao C, Sun YL, Jay GD, An KN, Amadio PC. The Effect of Surface Treatment Using Hyaluronic Acid and Lubricin on the Gliding Resistance
of Human Extrasynovial Tendons In Vitro. J Hand Surg Am. 2009;34:1276-1281.
14. Akasaka T, Nishida J, Imaeda T, Shimamura T, Amadio PC, An KN. Effect of hyaluronic acid on the excursion resistance of tendon graft: a biomechanical
in vitro study in a modified human model. Clin Biomech (Bristol, Avon). 2006;21:810-815.
15. Yamada T, Gotoh M, Nakama K, Mitsui Y, Higuchi F, Nagata K. Effects of hyaluronan on cell proliferation and mRNA expression of procollagens alpha 1 (I)
and alpha 1 (III) in tendon-derived fibroblasts from patients with rotator cuff disease: an in vitro study. Am J Sports Med. 2007;35:1870-1876.
16. Shimpuku E, Hamada K, Handa A, et al. Molecular effects of sodium hyaluronate on the healing of avian supracoracoid ten- don tear: according to in situ
hybridization and real-time polymerase chain reaction. J Orthop Res. 2007;25:173-184.
17. Wiig M, Abrahamsson SO. Hyaluronic acid modulates cell proliferation unequally in intrasynovial and extrasynovial rabbit tendons in vitro. J Hand Surg
Br. 2000;25:183-187.
18. Osti L, Berardocco M, di Giacomo V, Di Bernardo G, Oliva F, Berardi AC. Hyaluronic acid increases tendon derived cell viability and collagen type I expres-
sion in vitro: Comparative study of four different Hyaluronic acid preparations by molecular weight. BMC Musculoskelet Disord. 2015;16(1):284.
19. Neumann A, Schinzel R, Palm D, Riederer P, Münch G. High molecular weight hyaluronic acid inhibits advanced glycation endproduct-induced NF-κB
activation and cytokine expression. Febs Letters. 1999;453:283-287.
20. Tanimoto K, Ohno S, Fujimoto K, et al. Proinflammatory cy- tokines regulate the gene expression of hyaluronic acid synthetase in cultured rabbit synovial
membrane cells. Connect Tissue Res. 2001;42:187-195.
21. Smith MM, Ghosh P. The synthesis of hyaluronic acid by human synovial fibroblasts is influenced by the nature of the hyaluronate in the extracellular
environment. Rheumatol Int. 1987;7:113-122.
22. Nakamura H, Gotoh M, Kanazawa T, et al. Effects of corticosteroids and hyaluronic acid on torn rotator cuff tendons in vitro and in rats. J Orthop Res.
2015;33:1523-1530.
23. Zhao C, Wei Z, Kirk RL, et al. The Effects of Bio-Lubricating Molecules on Flexor Tendon Reconstruction in A Canine Allograft Model In Vivo. Plast Reconstr
Surg. 2014;133:628e- 637e.
24. Oryan A, Moshiri A, Meimandi Parizi AH, Raayat Jahromi A. Repeated administration of exogenous Sodium-hyaluronate improved tendon healing in an in
vivo transection model. J Tis- sue Viability. 2012;21:88-102.
25. Yoshida M, Funasaki H, Kubota M, Marumo K. Therapeutic effects of high molecular weight hyaluronan injections for tendinopathy in a rat model. J
Orthop Sci. 2015;20(1):186-195.
26. Liang JI, Lin PC, Chen MY, Hsieh TH, Chen JJ, Yeh ML. The effect of tenocyte/hyaluronic acid therapy on the early recovery of healing Achilles tendon in
rats. J Mater Sci Mater Med. 2014;25:217-227.
27. Salamanna F, Frizziero A, Pagani S, et al. Metabolic and cyto-protective effects of in vivo peri-patellar hyaluronic acid injections in cultured tenocytes.
Connect Tissue Res. 2015;56:35-43.
28. Frizziero A, Salamanna F, Giavaresi G, et al. Hyaluronic acid injections protect patellar tendon from detraining-associated damage. Histol Histopathol.
2015;30:1079-1088.
29. Yamaguchi T,Ochiai N,Sasaki Y,et al.Efficacy of hyaluronic acid or steroid injections for the treatment of a rat model of rotator cuff injury.J Orthop Res. 2015.
30. Meloni F, Milia F, Cavazzuti M, et al. Clinical evaluation of sodium hyaluronate in the treatment of patients with sopraspinatus tendinosis under echographic
guide: Experimental study of periarticular injections. Eur J Radiol. 2008;68:170-173.
31. Petrella RJ, Cogliano A, Decaria J, Mohamed N, Lee R. Management of Tennis Elbow with sodium hyaluronate periarticular injections. Sports Med
Arthrosc Rehabil Ther Technol. 2010;2;2-4.
32. Muneta T, Koga H, Ju YJ, Mochizuki T,Sekiya I. Hyaluronan injection therapy for athletic patients with patellar tendinopathy. J Orthop Sci.2012;17:425 431.
33. Blazina ME, Kerlan RK, Jobe FW, Carter VS, Carlson GJ. Jumper’s knee. Orthop Clin North Am. 1973;4:665-678.
34. Roles NC, Maudsley RH. Radial tunnel syndrome: resistant tennis elbow as a nerve entrapment. J Bone Joint Surg Br. 1972;54(3):499-508.
35. Riccio M, Battiston B, Pajardi G, et al. Efficiency of Hyaloglide in the prevention of the recurrence of adhesions after tenolysis of flexor tendons in zone II:
a randomized, controlled, multi-centre clinical trial. J Hand Surg Eur. 2010;35:130-138.
36. Özgenel GY, Etöz A. Effects of repetitive injections of hyaluronic acid on peritendinous adhesions after flexor tendon repair: a preliminary randomized,
placebo-controlled clinical trial. Ulus Travma Acil Cerrahi Derg. 2012;18:11-17.
37. Ozgen M, Fırat S, Sarsan A, Topuz O, Ardıç F, Baydemir C. Short and long-term results of clinical effectiveness of sodium hyaluronate injection in supraspi-
natus tendinitis. Rheumatol Int. 2012;32:137-144.
38. Merolla G, Bianchi P, Porcellini G. Ultrasound-guided subacromial injections of sodium hyaluronate for the management of rotator cuff tendinopathy: a
prospective comparative study with rehabilitation therapy. Musculoskelet Surg. 2013;97 Sup- pl 1:49-56.
39. Kumai T, Muneta T, Tsuchiya A, et al. The short-term effect after a single injection of high-molecular-weight hyaluronic acid in patients with enthesopathies
(lateral epicondylitis, patellar tendinopathy, insertional Achilles tendinopathy, and plantar fasciitis): a preliminary study. J Orthop Sci. 2014;19(4):603-611.
40. Al-Qattan MM. Factors in the pathogenesis of Dupuytren’s contracture. J Hand Surg [Am]. 2006;31:1527-1534.
41. Tomonaga A, Hamada K, Gotoh M, Yamakawa H, Kobayashi K, Fukuda H. Expression of procollagen alpha 1 type III mRNA in rotator cuff tears. Tokai J
Exp Clin Med. 2000;25(3):125-134.
42. Wiig M, Abrahamsson SO, Lundborg G. Effects of hyaluronan on cell proliferation and collagen synthesis: a study of rabbit flexor tendons in vitro. J Hand
Surg Am. 1997;22:818-825.
43. Moreland LW.Intra-articular hyaluronan(hyaluronic acid)and hylans for the treatment of osteoarthritis:mechanisms of action.Arthritis Res Ther.2003;5:54-67
44. Kaux JF, Forthomme B, Namurois MH, et al. Description of a standardized rehabilitation program based on sub-maximal eccentric following a platelet-rich
plasma infiltration for jumper’s knee. Muscles Ligaments Tendons J. 2014;4:85-89.
45. Kaux JF,Bouvard M,Lecut C,et al.Reflections about the optimisation of the treatment of tendinopathies with PRP.Muscles Ligaments Tendons J.2015;5:1 4.
46. Kaux JF, Le Goff C, Renouf J, et al. Comparison of the platelet concentrations obtained in platelet-rich plasma (PRP) between the GPS™ II and GPS™ III
systems. Pathol Biol (Paris). 2011;59:275-277.
47. Kaux JF, Le Goff C, Seidel L, et al. Comparative study of five techniques of preparation of platelet-rich plasma. Pathol Biol (Paris). 2011;59:157-160.

34
85
The Use of Hyaluronic Acid after Tendon Surgery and in Tendinopathies
(pag. 52)

References
1[1] M. Abate, D. Pulcini, A. di Iorio, and C. Schiavone, “Viscosup- plementation with intra-articular hyaluronic acid for treatment of osteoarthritis in the
elderly,” Current Pharmaceutical Design, vol. 16, no. 6, pp. 631–640, 2010.
[2] A. W. Rutjes, P. Ju¨ni, B. R. da Costa, S. Trelle, E. Nu¨esch, and S. Reichenbach, “Viscosupplementation for osteoarthritis of the knee: a systematic review
and meta-analysis,” Annals of Internal Medicine, vol. 157, no. 3, pp. 180–191, 2012.
[3] L. Hagberg, D. Heinegard, and K. Ohlsson, “The contents of macromolecule solutes in flexor tendon sheath fluid and their relation to synovial fluid; A
quantitative analysis,” Journal of Hand Surgery, vol. 17, no. 2, pp. 167–171, 1992.
[4] M. Riccio, B. Battiston, G. Pajardi et al., “Efficiency of hyaloglide in the prevention of the recurrence of adhesions after tenolysis of flexor tendons in zone
II: a randomized, controlled, multi- centre clinical trial,” Journal of Hand Surgery: European Volume, vol. 35, no. 2, pp. 130–138, 2010.
[5] G. Y. O¨ zgenel and A. Eto¨z, “Effects of repetitive injections of hyaluronic acid on peritendinous adhesions after flexor tendon repair: a preliminary rand-
omized, placebo-controlled clinical trial,” Ulusal Travma ve Acil Cerrahi Dergisi, vol. 18, no. 1, pp. 11–17, 2012.
[6] M. Corradi, M. Bellan, M. Frattini, G. Concari, S. Tocco, and F. Pogliacomi, “The four-strand staggered suture for flexor tendon repair: in vitro biomechanical
study,” Journal of Hand Surgery, vol. 35, no. 6, pp. 948–955, 2010.
[7] M. N. Kolodzinskyi, C. Zhao, Y. L. Sun et al., “The effects of hylan g-f 20 surface modification on gliding of extrasynovial canine tendon grafts in vitro,”
Journal of Hand Surgery, vol. 38, no. 2, pp. 231–236, 2013.
[8] T. Momose, P. C. Amadio, M. E. Zobitz, C. Zhao, and K.-N. An, “Effect of paratenon and repetitive motion on the gliding resistance of tendon of extrasyno-
vial origin,” Clinical Anatomy, vol. 15, no. 3, pp. 199–205, 2002.
[9] S. I¸sik, S. Oztu¨rk, S. Gu¨rses et al., “Prevention of restrictive adhesions in primary tendon repair by HA-membrane: experi- mental research in chickens,”
British Journal of Plastic Surgery, vol. 52, no. 5, pp. 373–379, 1999.
[10] Y. Liu, A. Skardal, X. Z. Shu, and G. D. Prestwich, “Prevention of peritendinous adhesions using a hyaluronan-derived hydrogel film following partial-
thickness flexor tendon injury,” Journal of Orthopaedic Research, vol. 26, no. 4, pp. 562–569, 2008.
[11] L. Callegari, E. Spano´, A. Bini, F. Valli, E. Genovese, and C. Fugazzola, “Ultrasound-guided injection of a corticosteroid and hyaluronic acid: a potential
new approach to the treatment of trigger finger,” Drugs in R and D, vol. 11, no. 2, pp. 137–145, 2011.
[12] T. Muneta, H. Koga, Y. J. Ju, T. Mochizuki, and I. Sekiya, “Hyaluronan injection therapy for athletic patients with patellar tendinopathy,” Journal of Ortho-
paedic Science, vol. 17, no. 4, pp. 425–431, 2012.
[13] R. J. Petrella, A. Cogliano, J. Decaria, N. Mohamed, and R. Lee, “Management of Tennis Elbow with sodium hyaluronate periar- ticular injections,” Sports
Medicine, Arthroscopy, Rehabilitation, Therapy and Technology, vol. 2, no. 1, article 4, 2010.
[14] I. Andia, M. Sanchez, and N. Maffulli, “Tendon healing and platelet-rich plasma therapies,” Expert Opinion on Biological Therapy, vol. 10, no. 10, pp.
1415–1426, 2010.
[15] J. W. Foland, G. W. Trotter, B. E. Powers, R. H. Wrighley, and F.
W. Smith, “Effect of sodium hyaluronate in collagenase-induced superficial digital flexor tendinitis in horses,” American Journal of Veterinary Research, vol.
53, no. 12, pp. 2371–2376, 1992.
[16] H. Tatari, E. Skiak, H. Destan, C¸. Uluku¸s, E. O¨ zer, and S. Satogˇlu, “Effect of hylan G-F 20 in achilles’ tendonitis: an experimental study in rats,” Archives
of Physical Medicine and Rehabilitation, vol. 85, no. 9, pp. 1470–1474, 2004.
[17] I. Tuncay, H. Ozbek, B. Atik, S. Ozen, and F. Akpinar, “Effects of hyaluronic acid on postoperative adhesion of tendo calcaneus surgery: an experimental
study in rats,” Journal of Foot and Ankle Surgery, vol. 41, no. 2, pp. 104–108, 2002.
[18] C. Zhao, Y. L. Sun, R. L. Kirk et al., “Effects of a lubricin- containing compound on the results of flexor tendon repair in a canine model in vivo,” The Journal
of Bone & Joint Surgery, vol. 92, no. 6, pp. 1453–1461, 2010.
[19] C. Yang, P. C. Amadio, Y.-I. Sun, C. Zhao, M. E. Zobitz, and K.-N. An, “Tendon surface modification by chemically modified HA coating after flexor
digitorum profundus tendon repair,” Journal of Biomedical Materials Research B: Applied Biomaterials, vol. 68, no. 1, pp. 15–20, 2004.
[20] Y.-L. Sun, C. Yang, P. C. Amadio, C. Zhao, M. E. Zobitz, and K.-
N. An, “Reducing friction by chemically modifying the surface of extrasynovial tendon grafts,” Journal of Orthopaedic Research, vol. 22, no. 5, pp. 984–989,
2004.
[21] A. R. C. Jones and C. R. Flannery, “Bioregulation of lubricin expression by growth factors and cytokines,” European Cells and Materials, vol. 13, pp.
40–45, 2007.
[22] T. Akasaka, J. Nishida, T. Imaeda, T. Shimamura, P. C. Amadio, and K.-N. An, “Effect of hyaluronic acid on the excursion resistance of tendon graft: a
biomechanical in vitro study in a modified human model,” Clinical Biomechanics, vol. 21, no. 8, pp. 810–815, 2006.
[23] M. Taguchi, C. Zhao, Y.-L. Sun, G. D. Jay, K.-N. An, and P. C. Amadio, “The effect of surface treatment using hyaluronic acid and lubricin on the gliding
resistance of human extrasynovial tendons in vitro,” Journal of Hand Surgery, vol. 34, no. 7, pp. 1276–1281, 2009.
[24] Y. Shibata, K. Midorikawa, G. Emoto, and M. Naito, “Clinical evaluation of sodium hyaluronate for the treatment of patients with rotator cuff tear,” Journal
of Shoulder and Elbow Surgery, vol. 10, no. 3, pp. 209–216, 2001.
[25] F. Meloni, F. Milia, M. Cavazzuti et al., “Clinical evaluation of sodium hyaluronate in the treatment of patients with sopraspinatus tendinosis under
echographic guide: experi- mental study of periarticular injections,” European Journal of Radiology, vol. 68, no. 1, pp. 170–173, 2008.
[26] C. Costantino and S. Olvirri, “Rehabilitative and infiltrative treatment with hyaluronic acid in elderly patients with rotator cuff tears,” Acta Biomedica de
l’Ateneo Parmense, vol. 80, no. 3, pp. 225–229, 2009.
[27] W.-Y. Chou, J.-Y. Ko, F.-S. Wang et al., “Effect of sodium hyaluronate treatment on rotator cuff lesions without complete tears: a randomized, double-blind,
placebo-controlled study,”Journal of Shoulder and Elbow Surgery, vol. 19, no. 4, pp. 557– 563, 2010.
[28] M. Ozgen, S. Fırat, A. Sarsan, O. Topuz, F. Ardıc¸, and C. Baydemir, “Short- and long-term results of clinical effectiveness of sodium hyaluronate injection
in supraspinatus tendinitis,” Rheumatology International, vol. 32, no. 1, pp. 137–144, 2012.
[29] G. Merolla, P. Bianchi, and G. Porcellini, “Ultrasound-guided subacromial injections of sodium hyaluronate for the manage- ment of rotator cuff tendinopa-
thy: a prospective comparative study with rehabilitation therapy,” Musculoskeletal Surgery, vol. 97, supplement 1, pp. 49–56, 2013.
[30] A. Menderes, F. Mola, V. Tayfur, H. Vayvada, and A. Barutc¸u, “Prevention of peritendinous adhesions following flexor tendon injury with seprafilm,” Annals
of Plastic Surgery, vol. 53, no. 6, pp. 560–564, 2004.

86
35
Single Hyaluronate Injection in the Management of Insertional Achilles
Tendinopathy in Comparison to Corticosteroid Injections and Non-
invasive Conservative Treatments (pag. 61)

References
1. Gaida, J. E., Bagge, J., Purdam, C., Cook, J., Alfredson, H., & Forsgren, S. (2012). Evidence of the TNF-α System in the Human Achilles Tendon: Expression
of TNF-α and TNF Receptor at both Protein and mRNA Levels in the Tenocytes. Cells Tissues Organs, 196(4), 339-352.
2. Zafar, M. S., Mahmood, A., & Maffulli, N. (2009). Basic science and clinical aspects of achilles tendinopathy. Sports medicine and arthroscopy review,
17(3), 190-197.
3. Magnussen, R. A., Dunn, W. R., & Thomson, A. B. (2009). Nonoperative treatment of midportion Achilles tendinopathy: a systematic review. Clinical Journal
of Sport Medicine, 19(1), 54-64.
4. BursITI, T. (2008). Musculoskeletal injections: a review of the evidence. Am Fam Physician, 78(8), 971-976.
5. Petrella, R. J., Petrella, M. J., & Cogliano, A. (2007). Periarticular hyaluronic acid in acute ankle sprain. Clinical Journal of Sport Medicine, 17(4), 251-257.
6. Nago, M., Mitsui, Y., Gotoh, M., Nakama, K., Shirachi, I., Higuchi, F., & Nagata, K. (2010). Hyaluronan modulates cell proliferation and mRNA expression
of adhesion-related procollagens and cytokines in glenohumeral synovial/capsular fibroblasts in adhesive capsulitis. Journal of Orthopaedic Research, 28(6),
726-731.
7. Ozgenel, G. Y., & Etöz, A. (2012). Effects of repetitive injections of hyaluronic acid on peritendinous adhesions after flexor tendon repair: a preliminary
randomized, placebo-controlled clinical trial. Ulus Travma Acil Cerrahi Derg, 18(1), 11-17.
8. Gorelick, L., Rozano-Gorelick, A., Robinson, D., Marcus, O., Joubran, S., & Ram, E. (2013). Treatment of Hip Trochanteric Bursitis Using Hyaluronate
Injections, 3(2), 125-129.
9. Mitsui, Y., Gotoh, M., Nakama, K., Yamada, T., Higuchi, F., & Nagata, K. (2008). Hyaluronic acid inhibits mRNA expression of proinflammatory cytokines and
cyclooxygenase-2/prostaglandin E2 production via CD44 in interleukin-1-stimulated subacromial synovial fibroblasts from patients with rotator cuff disease.
Journal of Orthopaedic Research, 26(7), 1032-1037.
10. Pereira, A. A., López, B. M., & de la Serna, A. R. (2015). A Comparative Study between Hyaluronic Acid and Corticosteroids for the Treatment of the Greater
Trochanteric Pain Syndrome. Open Journal of Rheumatology and Autoimmune Diseases, 5(03), 57-62.
11. Halici, M., Karaoglu, S., Canoz, O., Kabak, S., & Baktir, A. (2004). Sodium hyaluronate regulating angiogenesis during Achilles tendon healing. Knee
Surgery, Sports Traumatology, Arthroscopy, 12(6), 562-567.
12. Tatari, H., Skiak, E., Destan, H., Ulukuş, Ç., Özer, E., & Satoğlu, S. (2004). Effect of hylan GF 20 in Achilles’ tendonitis: an experimental study in rats.
Archives of physical medicine and rehabilitation, 85(9), 1470-1474.
13. Kumai, T., Muneta, T., Tsuchiya, A., Shiraishi, M., Ishizaki, Y., Sugimoto, K., ... & Takakura, Y. (2014). The short-term effect after a single injection of high-
molecular-weight hyaluronic acid in patients with enthesopathies (lateral epicondylitis, patellar tendinopathy, insertional Achilles tendinopathy, and plantar
fasciitis): a preliminary study. Journal of Orthopaedic Science, 19(4), 603-611.
14. Hart, L. (2011). Corticosteroid and other injections in the management of tendinopathies: a review. Clinical Journal of Sport Medicine, 21(6), 540-541.
15. Southerland, J. T., Boberg, J. S., Downey, M. S., Nakra, A., & Rabjohn, L. V. (2012). McGlamry’s comprehensive textbook of foot and ankle surgery. Lip-
pincott Williams & Wilkins., p- 1129-1130.
16. Lehtinen, A., Peltokallio, P., & Taavitsainen, M. (1993). Sonography of Achilles tendon correlated to operative findings. In Annales chirurgiae et gynaeco-
logiae, 83(4), 322-327.
17. Calve, S., Isaac, J., Gumucio, J. P., & Mendias, C.L. (2012). Hyaluronic acid, HAS1, and HAS2 are significantly upregulated during muscle hypertrophy.
American Journal of Physiology-Cell Physiology, 303(5), C577-C588.
18. Caborn, D., Rush, J., Lanzer, W., Parenti, D., Murray, C., & Synvisc 901 Study Group. (2004). A randomized, single-blind comparison of the efficacy and
tolerability of hylan GF 20 and triamcinolone hexacetonide in patients with osteoarthritis of the knee. The Journal of rheumatology, 31(2), 333-343.
19. Metcalfe, D., Achten, J., & Costa, M. L. (2009). Glucocorticoid injections in lesions of the Achilles tendon. Foot & ankle international, 30(7), 661- 665.
20. Gorelick, L, Rozano-Gorelick, A, Saab A, & Ram, E. (2014). Results of treatment of greater trochanteric pain syndrome in diabetic patients by single
hyaluronic acid injection. International Journal of Humanities, Arts, Medicine and Sciences, 2(9), 53-60.

36
87
Management of Tennis Elbow with sodium hyaluronate periarticular
injections (pag. 66)

Author details
1Dept Medicine, Canadian Centre for Activity and Aging, 801 Commissioners Road, London, N6C5J1, Canada. 2Dept Medicine, Fowler-Kennedy Sport Medi-
cine Clinic, 1490 Richmond St, London, N6C2 M3, Canada. 3Dept Kinesiology, Canadian Centre for Activity and Aging, 801 Commissioners Road, London,
N6C5J1, Canada. 4Dept Family Medicine, 801 Commissioners
Road, London, N6C5J1, Canada. 5Sport Medicine, University of Waterloo, 1100 University Ave, Waterloo, Canada.

Authors’ contributions
RJP contributed to the study design, data collection, analysis and manuscript preparation.
AC contributed to the data collection.
JD contributed to the study design, data collection and manuscript preparation
NM contributed to the data collection. RL contributed to the data collection.
All authors read and approved of the final manuscript.

Competing interests
The authors declare that they have no competing interests.

Received: 4 October 2009


Accepted: 2 February 2010 Published: 2 February 2010

References
1. Rabago D, Best TM, Zgierska A, Zeisig E, Ryan M, Crane D: A systematic review of four injection therapies for lateral epicondylosis: prolotherapy, polido-
canol, whole blood and platelet rich plasma. BJSM 2009, 43:471-481.
2. Verhaar JA: Tennis elbow. Anatomical, epidemiological and therapeutic aspects. Int Orthop 1994, 18:263-267.
3. Wadsworth DG: Tennis elbow: Conservative, surgical and manipulative treatment. BMJ 1987, 294:621-623.
4. Hudak PL, Cole DC, Haines AT: Understanding prognosis to improve rehabilitation: Example of lateral elbow pain. Arch Phys Med Rehabil 1996, 77:586-
593.
5. Ljung BO, Lieber RL, Fridén J: Wrist extensor muscle pathology in lateral epicondylitis. J Hand Surg Britain 1999, 24:177-183.
6. Nirschl RP: Soft tissue injuries about the elbow. Clin Sports Med 1986, 5:637-652.
7. Noteboom T, Cruver R, Keller J, Kellogg B, Nitz AJ: Tennis elbow: A review.
J Orthop Sports Phys Ther 1994, 19:357-366.
8. Vicenzino B, Wright A: Lateral epicondylagia: A review of epidemiology, pathophysiology, etiology and natural history. Phys Ther Rev 1996, 1:23-34.
9. Assendelft WJJ, Hay EM, Adshead R, Bouter LM: Corticosteroid injections for lateral epicondylitis: A systematic review. British J Gen Pract 1996, 46:209-
216.
10. Smidt N, Windt van der DA, Assendelft WJ, Deville WL, Korthals-de Bos IB, Bouter LM: Corticosteroid injections, physical therapy or a wait and see policy
for lateral epicondylitis: A randomized controlled trial. Lancet 2002, 359:657-662.
11. Assendelft W, Green S, Buchbinder R, Struijs P, Smidt N: Tennis elbow. Clin Evid 2004, , 11: 1633-1644.
12. Petrella RJ, DiSilvestro MD, Hildebrand C: Sodium hyaluronate effects on pain and physical functioning in osteoarthritis of the knee: a randomised,
double-blind, placebo-controlled clinical trial. Archives of Internal Medicine 2002, 162:292-298.
13. Petrella RJ, Petrella MJ, Cogliano A: Periarticular hyaluronic acid in acute ankle sprain. Clinical Journal of Sports Medicine 2007, 17(4):251-257.
14. Burnham R, Gregg R, Healy P, Steadward R: The effectiveness of topical diclofenac for lateral epicondylitis. Clin J Sport Med 1998, 8:78-81.
15. Wong SM, Hui ACF, Tong P-Y, Poon DWF, Yu E, Wong LKS: Treatment of lateral epicondylitis with botulinum toxin. A randomized, double-blind, placebo-
controlled trial. Ann Int Med 2005, 143:793-797.
16. Petrella MJ, Cogliano A, Petrella RJ: Long-term efficacy and safety of periarticular hyaluronic acid in acute ankle sprain. The Physician and Sportsmedicine
2009, 37(1).

doi:10.1186/1758-2555-2-4
Cite this article as: Petrella et al.: Management of Tennis Elbow with
sodium hyaluronate periarticular injections. Sports Medicine, Arthroscopy,
Rehabilitation, Therapy & Technology 2010 2:4.

88
37
Long-Term Efficacy and Safety of Periarticular Hyaluronic Acid in Acute
Ankle Sprain (pag. 71)

References
1. Perlman M, Leveille D, DeLeonibus J, et al. Inversion lateral ankle trauma: differential diagnosis, review of the literature, and prospective study. J Foot
Surg. 1987;26(2):95–135.
2. Garrick JG. Epidemiologic perspective: symposium on ankle and foot problems in the athlete. Clin Sports Med. 1982;1(1):13–18.
3. Liu SH, Nguyen TM. Ankle sprains and other soft tissue injuries. Curr Opin Rheumatol. 1999;11(2):132–137.
4. Yeung MS, Chan KM, So CH, et al. An epidemiological survey on ankle sprain. Br J Sports Med. 1994;28(2):112–116.
5. McCaig LF, Nghi L. National Hospital Ambulatory Medical Care Survey: 2000 Emergency Department Summary. Atlanta, GA: Centers for Disease Control
and Prevention. 2002;326:1–31.
6. van Rijn RM, van Os AG, Bernsen RM, Luijsterburg PA, Koes BW, Bierma-Zeinstra SM. What is the clinical course of acute ankle sprains? A systematic
literature review. Am J Med. 2008;121(4):324–331.
7. Colville MR, Amendola N, Butters MA, et al. Clinical Guideline on Ankle Injury. Rosemont, IL: American Academy of Orthopaedic Surgeons; 1999:7.
8. Cook RJ, Sackett DL. The number needed to treat: a clinically useful measure of treatment effect. BMJ. 1995;310(6977):452–454.
9. Ekman EF, Fiechtner JJ, Levy S, Fort JG. Efficacy of celecoxib versus ibuprofen in the treatment of acute ankle pain: a multicenter, double-blind, rand-
omized controlled trial in acute ankle sprain. Am J Orthop. 2002;31(8):445–451.
10. Fioravanti A, Cicero MR, Nerucci F, Manopulo R, Marcolongo R. Double-blind controlled clinical study of the efficacy and tolerability of diclofenac-
N-(2-hydroxyethyl)-pyrrolidine lecithin gel compared with diclofenac-N-(2-hydroxyethyl)-pyrrolidine gel in patients with peri and extraarticular inflammatory
diseases. Drugs Exp Clin Res. 1999:25(5): 235–240.
11. Gallagher EJ, Liebman M, Bijur PE. Prospective validation of clinically important changes in pain severity measured on a visual analog scale. Ann Emerg
Med. 2001;38(6):633–638.
12. Petrella R, Ekman EF, Schuller R, Fort JG. The efficacy of celecoxib, a COX-2-specific inhibitor, and naproxen in the management of acute ankle sprain:
results of a double-blind, randomized controlled trial. Clin J Sport Med. 2004;(4)14:225–231.
13. Handoll HH, Rowe BH, Quinn KM, de Bie R. Interventions for preventing ankle ligament injuries. Cochrane Database Syst Rev. 2001(3);CD000018.
14. Bridgman SA, Clement D, Downing A, Walley G, Phair I, Maffulli N. Population based epidemiology of acute ankle sprains attending accident and emer-
gency units in the West Midlands of England, and a survey of UK practice for severe ankle sprains. Emerg Med J. 2003;20(6):508–510.
15. Gerber JP, Williams GN, Scoville CR, Arciero RA, Taylor DC. Persistent disability associated with ankle sprains: a prospective examination of an athletic
population. Foot Ankle Int. 1998;19(10):653–660.
16. Balazs EA, Denlinger JL. Viscosupplemantation: a new concept in the treatment of osteoarthritis. J Rheumatol Suppl. 1993;39:3–9.
17. Petrella RJ, Petrella MJ, Cogliano A. Periarticular hyaluronic acid in acute ankle sprain. Clin J Sport Med. 2007;17(4):251–257.
18. Petrella RJ. Periarticular hyaluronic acid for acute ankle sprain. Ann Rheum Dis. 2005;64:385.
19. Petrella, RJ, DiSilvestro MD, Hildebrand, C. Effects of hyaluronate sodium on pain and physical functioning in osteoarthritis of the knee: a randomized,
double-blind, placebo-controlled clinical trial. Arch Intern Med. 2002;162(3):292–298.
20. Bellamy N, Campbell J, Robinson V, Gee T, Bourne R, Wells G. Viscosupplementation for the treatment of osteoarthritis of the knee. Cochrane Database
Syst Rev. 2006;19(2):CD005321.
21. Mahler P, Mahler F, Duruz H, Ramazzina M, Liguori V, Mautone G. Double-blind, randomized, controlled study on the efficacy and safety of a novel di-
clofenac epolamine gel formulated with lecithin for the treatment of sprains, strains and contusions. Drugs Exp Clin Res. 2003;29(1):45–52.
22. Mazieres B, Rouanet S, Velicy J, Scarsi C, Reiner V. Topical ketoprofen patch (100 mg) for the treatment of ankle sprain: a randomized, double-blind,
placebo-controlled study. Am J Sports Med. 2005;33(4):515–523.

38
89
39
40

S-ar putea să vă placă și