Next Article in Journal
Magnetic Resonance Imaging of the Knee in the Presence of Bridging External Fixation: A Comparative Experimental Evaluation of Four External Fixators, Including Dolphix®
Next Article in Special Issue
Extracellular to Intracellular Body Water and Cognitive Function among Healthy Older and Younger Adults
Previous Article in Journal / Special Issue
Application and Surgical Technique of ACL Reconstruction Using Worldwide Registry Datasets: What Can We Extract?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Low Molecular Weight Hyaluronic Acid (500–730 Kda) Injections in Tendinopathies—A Narrative Review

by
Antonio Frizziero
1,*,
Filippo Vittadini
2,
Davide Bigliardi
1 and
Cosimo Costantino
1
1
Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
2
Department of Physical and Rehabilitation Medicine, Casa di Cura Policlinico S. Marco, 30100 Venice, Italy
*
Author to whom correspondence should be addressed.
J. Funct. Morphol. Kinesiol. 2022, 7(1), 3; https://0-doi-org.brum.beds.ac.uk/10.3390/jfmk7010003
Submission received: 2 December 2021 / Revised: 23 December 2021 / Accepted: 27 December 2021 / Published: 29 December 2021
(This article belongs to the Special Issue Role of Exercises in Musculoskeletal Disorders—4th Edition)

Abstract

:
Tendinopathies are common causes of pain and disability in general population and athletes. Conservative treatment is largely preferred, and eccentric exercise or other modalities of therapeutic exercises are recommended. However, this approach requests several weeks of consecutive treatment and could be discouraging. In the last years, injections of different formulations were evaluated to accelerate functional recovery in combination with usual therapy. Hyaluronic acid (HA) preparations were proposed, in particular LMW-HA (500–730 kDa) for its unique molecular characteristics in favored extracellular matrix homeostasis and tenocyte viability. The purpose of our review is to evaluate the state-of-the-art about the role of 500–730 kDa in tendinopathies considering both preclinical and clinical findings and encourage further research on this emerging topic.

1. Introduction

Tendinopathy is multifactorial pathology, that could lead to chronic pain and functional impairment [1]. The rotator cuff [2], the long head of the biceps [3], the wrist extensors and flexors [4], the adductors [5], the patellar tendon, the Achilles tendon [6] and the posterior tibial tendon [7] are the most affected tendons.
Tendinopathies are largely triggered by overload [8], but a great number (about one third) of patients do not practice regular physical activity [9].
Tendinopathies can occur in acute and chronic state and can be supported by different factors both intrinsic (age [10], body structure [11], nutrition [12], metabolic diseases [13,14], genetics [15]) and extrinsic (excessive and improper fatigue [8,16], detraining [17], and external damage [18,19,20]) factors. All these factors seem to be related to the appearance of overload tendinopathy although a cause–effect relationship has not yet been demonstrated.
Tendinopathies are characterized by disorganization of collagen fibers with dysregulation in extracellular matrix homeostasis, alteration in proteoglycan content, enhanced tenocyte apoptosis, and increase in the microvasculature and sensory nerve innervations [19,21]. Additionally, paratenon shows mucoid degeneration, fibrosis, and vascular proliferation with mild inflammatory infiltrate [22].
There are currently no treatments of choice for tendinopathies as no strong evidence has yet been found that one treatment is better than another [1,23,24]. Until a few years ago, eccentric exercise [25,26,27] and extracorporeal shock wave therapy (ESWT) [28] showed significant effects on both pain and functional recovery.
Recently, thanks also to the ultrasound high-definition evaluation and guide [29], injection therapy has also carved out an important role in the treatment of tendinopathies through different proposals with different levels of success, as hyaluronic acid [30], PRP [31], high volume injections [32], prolotherapy [33,34], autologous conditioned serum [35], sclerotherapy [34], percutaneous electrolysis [36], etc.
Hyaluronic acid (HA) is a polymer of disaccharides, which are composed of d-glucuronic acid and N-acetyl-d-glucosamine, linked via alternating β-(1→4) and β-(1→3) glycosidic bonds [37].
In particular, HA allows a smooth tendon gliding and provides nutrition to tendon itself, and it is an important component of tendon structure, being largely present in extracellular space.
HA can be classified by origin (rooster combs or bacterial fermentation), molecular organization (linear, cross-linked, or hybrid), and molecular weight [38]:
  • Low molecular weight: <1000 KDa;
  • Medium molecular weight: 1000 KDa < MW < 2000 KDa;
  • High molecular weight: >2000 KDa.
Most of the physiological effects of exogenous hyaluronic acid and its viscosity depend primarily on the molecular weight of the molecule, consequently influencing its possible applications [30].
Low molecular weight hyaluronic acid (500–730 KDa) formulations were the first proposed in clinical practice for knee osteoarthritis since 1990, considering “viscosupplementation” as purpose with high safety profile [39,40,41]. However, several preclinical and clinical studies increased the knowledge about the numerous possible effects of the interaction with articular tissues as cartilage subchondral bone, synovial membrane, and most recently on tendons.
In recent years, the role of low molecular weight HA (LMW-HA) is emerging among the possible treatments of tendon pathology, secondary to the growing scientific evidence in the preclinical field and to the greater sophistication of ultrasound and techniques to perform therapeutic interventions safely [42].
The purpose of the present study is to perform a descriptive review to highlight the state of the art on the efficacy of hyaluronic acid at low-molecular weight injections in the treatment of tendinopathies.

2. Materials and Methods

The present systematic review was conducted on common electronic databases including PubMed (MEDLINE), Pedro, and Cochrane Library. Each database was searched according to its specific syntax rules. Moreover, manual search of published and unpublished studies (conference abstracts, textbooks, ‘grey’ literature) was also conducted and reference lists of retrieved articles were screened. Search results were limited to studies written in English and published until August 2021. Studies were selected using the following keywords alone or in combination: “hyaluronic acid”, “low molecular weight hyaluronic acid 500–730 KDa”, “tendinopathy”, “tendon” and “injection”. The search results and the relative selection process are shown in the following flowchart (Figure 1).

Study Selection

The selection criteria were: (i) language of publications (English); (ii) study design (preclinical studies, RCT or prospective studies); (iii) sample, including adult patients with a diagnosis of Achilles, patellar or supraspinatus tendinopathy, lateral epicondylitis, or trigger digit; (iv) outcome measures, including pain and functional improvement, patient satisfaction, or return to sport; (v) treatment, consisting in hyaluronic acid injection, alone or associated with other therapies (i.e., exercise). Using the above-mentioned criteria, relevant studies were selected after reading the title and/or abstract, while publications that did not meet the criteria were excluded. Full text from articles selected for inclusion in the review was read, and the bibliographies were hand searched for further relevant publications. As a result, some studies were withdrawn after reading the full text, whereas others were retrieved from the bibliography of the selected articles.

3. Results

The initial search yielded 76 articles, of which 34 were identified as relevant after screening of titles/abstract. Twenty records were excluded because they were not experimental studies or pre-clinical studies. Two papers were excluded after reading full article for methodological issues.
Finally, 12 papers were retrieved from literature search. Among them, 8 clinical studies were selected. Only one compared LMW-HA against placebo; 4 were RTC; and 2 published observational study (Table 1 and Table 2).

3.1. In Vitro Studies

Osti et al. evaluated the effects of four different hyaluronic acid formulations on viability, metabolic activity, apoptosis, and collagen type I and collagen type III expression on human rotator cuff tendon tears derived cells. The authors found that all hyaluronic acid preparation were efficient in amelioration of cell-metabolic activity, decrease rate of the apoptosis of tendon derived cells and increase Collagen I production in dose dependent manner not related to the molecular weight [43].
Gallorini et al. investigated the antioxidant effects of the same four HA formulations and concluded that all the molecules have protective effects against the oxidative stress-related cytotoxicity. However, protective effects appear to be particularly efficient in tendon cells treated with LMW-HA (500–730 KDa) [44].

3.2. Pre-Clinical Studies

Salamanna et al. investigated the effects of repeated peri-patellar injections of LMW-HA (500–730 KDa) in sudden-detraining rat model tendinopathy, founding that HA allow the maintenance of tenocyte anabolic activity and proliferation in comparison to saline or no intervention [45].
Furthermore, in a second study Frizziero et al. demonstrated that repeated peri-patellar injections of LMW-HA may maintain the structural and functional properties of patellar tendon and enthesis in sudden detrained rats [46].

3.3. Clinical Studies

Meloni et al. found that five US-guided LMW-HA injections were effective compared to placebo in improving pain and function in 56 patients affected by supraspinatus tendinosis until 9 months [47].
Blaine et al. found that LMW-HA (500 to 730 kDa) injection is effective in the short term and well tolerated for the treatment of osteoarthritis and persistent shoulder pain (including rotator cuff tear) that is refractory to other standard non operative interventions [48].
Frizziero et al. observed that LMW-HA (500 to 730 kDa) and low-energy ESWT were both effective and safe in patients suffering from non-calcific rotator cuff tendinopathy until a 3-month follow-up. However, LMW-HA provide prompt clinical improvement compared to ESWT [28].
Fogli et al. have shown that three US-guided LMW-HA paratendinous injections induce significant progressive improvement in pain and function and US parameters (tendon thickness and neovascularization) in patients with Achilles or patellar tendinopathies [49].
Similar good results in terms of pain reduction and improvement of function in Achilles and patellar tendinopathy have also been observed by Frizziero et al. following US-guided paratendinous injection treatment [50].
Kanchanathepsak et al. compared the efficacy of US-guided injection of LMW-HA (500–730 KDa) and corticosteroids (triamcinolone acetate) in the treatment of trigger finger. The authors noted that both LMW-HA and corticosteroids provide a comparable therapeutic effect in the medium term. However, it is necessary to consider that corticosteroids have a cytotoxic effect on tendons [51].
Recently, Mohebbi et al. evaluated the clinical effects of HMW-HA (>2000 kDa) versus LMW-HA (500–730 KDa) single US-guided periarticular injection in the management of rotator cuff tendinopathy with persistent shoulder pain for more than 6 weeks. The pain, induration, and inflammation at the site of injection are less prominent for LMW-HA compared with HMW-HA. Even both preparations were effective in improving pain, joint function, and quality of life that last at least for 3 months, LMW-HA is less expensive and resulted more tolerable to the patient. LMW-HA seems also more effective than physiotherapy in alleviating pain and improving shoulder ROM and quality of life. Furthermore, patient compliance was higher with LMW-HA injection in relation to lower perceived pain [52].
Rezasoltani et al. compared the effects of LMW-HA injection with physiotherapy in patients with supraspinatus tendinopathy and concluded that injection therapy showed greater efficacy at a 3-month follow-up. In particular, hyaluronic acid is more successful in relieving pain [53].

4. Discussion

In past years, intra-articular injections of hyaluronic acid were a well-known and widely used option in the conservative treatment of osteoarthritis with positive results. On the other hand, the role of HA in tendinopathies was underestimated. Although further studies are needed to demonstrate the clinical efficacy and safety of hyaluronic acid in the management of tendinopathies, several studies demonstrated positive effects on tendinopathies with low molecular weight hyaluronic acid injections.
The more frequent performance of ultrasound evaluation and the possibility of positioning the therapeutic substance with extreme precision in the affected site have allowed for better results in the clinical field.
The results observed in in-vitro studies have showed protective effects against the oxidative stress-related cytotoxicity in the tendon derived cells treated with the three lower molecular weight HAs [43,44].
Osti et al. identified a protective effect of the tendon by hyaluronic acid, related to the very low expression for the production of collagen type III [43].
The encouraging effects of hyaluronic acid in conditions of tendon suffering have also been confirmed in pre-clinical studies [45,46]. Salamanna et al. [45] and Frizziero et al. [46] investigated the effects of repeated peri-patellar injections of low molecular weight HA (500–730 KDa) on tenocytes morphology, viability, proliferation, and metabolic activity and both the authors concluded that HA injections at the end of the I, II, III, and IV weeks in detrained tendinopathies rat model seems to help tenocytes to maintain their synthetic and metabolic activities, the structural and functional properties of the patellar tendon in ensuring safety.
Following the results just described where LMW-HA has shown the potential to determine amelioration of tendon behavior via change in tenocyte viability and proliferation and collagen composition were promoted clinical studies on human different tendon’s site (rotator cuff tendons, patellar tendon, Achilles tendon, etc.) with encouraging conclusions [47,48,49,50,51,52,53].
In fact, the results obtained by Meloni et al. [47] in rotator cuff pathology shows better results with hyaluronic acid than placebo, while Frizziero et al. [28] demonstrated faster efficacy of injections with low molecular weight hyaluronic acid compared to shock waves.
Thus, it could be speculated that LMW-HA should contribute to the restoration of the essential viscoelastic properties of tendon, leading to function improvement without side effects.
Mohebbi et al. [52] observed in a recent comparative study between ultrasound-guided injections with low molecular weight (500–730 KDa) and high molecular weight (>2000 KDa) that, with the same effectiveness, low molecular weight was more tolerated by patients.
Rezasoltani et al. [53] and Blaine et al. [48] confirmed the efficacy of low molecular weight hyaluronic acid (500–730 KDa) injection in pain relief and function amelioration.
Similar positive results on pain and functional recovery have been observed in the clinical experiences of Frizziero et al. [50] and Fogli et al. [49] in the Achilles tendon, patellar tendon, and epicondylitis.
Pathological conditions such as tenosynovitis also benefited from injection treatment with low molecular weight hyaluronic acid (500–730 KDa) in comparison to triamcinolone acetonide. Despite the corticosteroid produced more rapid remission of the symptoms, hyaluronic acid was also shown to be safer and produced a longer lasting effect [51].
All the studies have found that clinical effectiveness should be reached already at 1 month after the LMW-HA injections; this finding should be crucial in athletes that need safe and rapid return to sport participation.
Furthermore, it appears that LMW-HA injection was highly tolerated by patients, with only rare local, self-limiting side-effects. In specific, LMW-HA presented better tolerability and minor costs in comparison to HMW-HA formulations [52].

5. Conclusions

Tendinopathies are a group of pathologies that afflict the tendons and nowadays there are many current treatment options, but none has shown particular superiority. Among these therapeutic proposals and following favorable results observed with in vitro, pre-clinical, and clinical studies, the use of low molecular weight hyaluronic acid by injection should be encouraged compared to high-weight hyaluronic acid. The rapid action and the high safety profile make this treatment particularly suitable for athletes. To achieve extreme accuracy of positioning of hyaluronic acid, the use of ultrasound guidance is strongly recommended. Even the lower cost compared to hyaluronic acids with higher molecular weight can represent an additional incentive to its use.

6. Limitations of the Review

Despite the limitations of our study represented by the low number of papers in the literature and the small number of patients treated, the results obtained encourage the use of low molecular weight hyaluronic acid in tendinopathies. For these reasons, further studies are needed to produce more evidence of efficacy of low molecular weight hyaluronic acid in tendinopathies.

Author Contributions

Conceptualization, A.F.; methodology, A.F. and F.V.; validation, A.F., D.B. and C.C.; formal analysis, A.F.; investigation, A.F.; writing—original draft preparation, A.F. and D.B.; writing—review and editing, A.F. and F.V.; supervision, C.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The study does not report any data.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Challoumas, D.; Biddle, M.; Millar, N.L. Recent advances in tendinopathy. Fac. Rev. 2020, 9, 16. [Google Scholar] [CrossRef]
  2. Merolla, G.; Singh, S.; Paladini, P.; Porcellini, G. Calcific tendinitis of the rotator cuff: State of the art in diagnosis and treatment. J. Orthop. Traumatol. 2016, 17, 7–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Sarmento, M. Long head of biceps: From anatomy to treatment. Acta Reum. Port. 2014, 40, 26–33. [Google Scholar]
  4. Patrick, N.C.; Hammert, W.C. Hand and Wrist Tendinopathies. Clin. Sports Med. 2020, 39, 247–258. [Google Scholar] [CrossRef] [PubMed]
  5. Sirico, F.; Palermi, S.; Massa, B.; Corrado, B. Tendinopathies of the hip and pelvis in athletes: A narrative review. J. Hum. Sports Exerc. 2020, 15, S748–S762. [Google Scholar] [CrossRef]
  6. Tarantino, D.; Palermi, S.; Sirico, F.; Balato, G.; D’Addona, A.; Corrado, B. Achilles tendon pathologies: How to choose the best treatment. J. Hum. Sport Exerc. 2020, 15, S1300–S1321. [Google Scholar]
  7. Simpson, M.R.; Howard, T.M. Tendinopathies of the foot and ankle. Am. Fam. Physician 2009, 80, 1107–1114. [Google Scholar] [PubMed]
  8. Sharma, P.; Maffulli, N. Basic biology of tendon injury and healing. Surgeon 2005, 3, 309–316. [Google Scholar] [CrossRef]
  9. Waldecker, U.; Hofmann, G.; Drewitz, S. Epidemiologic investigation of 1394 feet: Coincidence of hindfoot malalignment and Achilles tendon disorders. Foot Ankle Surg. 2012, 18, 119–123. [Google Scholar] [CrossRef]
  10. Riley, G.P.; Curry, V.; DeGroot, J.; van El, B.; Verzijl, N.; Hazleman, B.L.; Bank, R.A. Matrix metalloproteinase activities and their relationship with collagen remodelling in tendon pathology. Matrix Biol. 2002, 21, 185–195. [Google Scholar] [CrossRef] [Green Version]
  11. Franceschi, F.; Papalia, R.; Paciotti, M.; Franceschetti, E.; Di Martino, A.; Maffulli, N.; Denaro, V. Obesity as a Risk Factor for Tendinopathy: A Systematic Review. Int. J. Endocrinol. 2014, 2014, 670262. [Google Scholar] [CrossRef]
  12. Longo, U.G.; Ronga, M.; Maffulli, N. Achilles tendinopathy. Sports Med. Arthrosc. Rev. 2009, 17, 112–126. [Google Scholar] [CrossRef] [PubMed]
  13. Oliva, F.; Piccirilli, E.; Berardi, A.C.; Frizziero, A.; Tarantino, U.; Maffulli, N. Hormones and tendinopathies: The current evidence. Br. Med Bull. 2016, 117, 39–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Frizziero, A.; Vittadini, F.; Gasparre, G.; Masiero, S. Impact of oestrogen deficiency and aging on tendon: Concise review. Muscle Ligaments Tendons J. 2019, 4, 324–328. [Google Scholar] [CrossRef]
  15. Maffulli, N.; Reaper, J.A.; Waterston, S.W.; Ahya, R. ABO Blood Groups and Achilles Tendon Rupture in the Grampian Region of Scotland. Clin. J. Sport Med. 2000, 10, 269–271. [Google Scholar] [CrossRef]
  16. Maffulli, N.; Sharma, P.; Luscombe, K.L. Achilles tendinopathy: Aetiology and management. J. R. Soc. Med. 2004, 97, 472–476. [Google Scholar] [CrossRef] [Green Version]
  17. Frizziero, A.; Fini, M.; Salamanna, F.; Veicsteinas, A.; Maffulli, N.; Marini, M. Effect of training and sudden detraining on the patellar tendon and its enthesis in rats. BMC Musculoskelet. Disord. 2011, 12, 20. [Google Scholar] [CrossRef]
  18. van der Linden, P.D.; Sturkenboom, M.C.J.M.; Herings, R.M.C.; Leufkens, H.M.G.; Rowlands, S.; Stricker, B.H.C. Increased risk of achilles tendon rupture with quinolone antibacterial use, especially in elderly patients taking oral corticosteroids. Arch. Intern. Med. 2003, 163, 1801–1807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Bisaccia, D.R.; Aicale, R.; Tarantino, D.; Peretti, G.; Maffulli, N. Biological and chemical changes in fluoroquinolone-associated tendinopathies: A systematic review. Br. Med Bull. 2019, 130, 39–49. [Google Scholar] [CrossRef] [PubMed]
  20. Di Meglio, F.; Sacco, A.M.; Belviso, I.; Romano, V.; Sirico, F.; Loiacono, C.; Palermi, S.; Pempinello, C.; Montagnani, S.; Nurzynska, D.; et al. Influence of Supplements and Drugs used for the Treatment of Musculoskeletal Disorders on Adult Human Tendon-Derived Stem Cells. Muscle Ligaments Tendons J. 2020, 10, 376. [Google Scholar] [CrossRef]
  21. Romero, E.A.S.; Pollet, J.; Pérez, S.M.; Pérez, J.L.A.; Fernández, A.C.M.; Pedersini, P.; Carballar, C.B.; Villafañe, J.H. Lower Limb Tendinopathy Tissue Changes Assessed through Ultrasound: A Narrative Review. Medicina 2020, 56, 378. [Google Scholar] [CrossRef] [PubMed]
  22. Ker, R.F. Dynamic tensile properties of the plantaris tendon of sheep (Ovisaries). J. Exp. Biol. 1981, 93, 283–302. [Google Scholar] [CrossRef]
  23. Cook, J.L.; Purdam, C.R. Is tendon pathology a continuum? A pathology model to explain the clinical presentation of load-induced tendinopathy. Br. J. Sports Med. 2009, 43, 409–416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Frizziero, A.; Bonsangue, V.; Trevisan, M.; Ames, P.R.J.; Masiero, S. Foot tendinopathies in rheumatic diseases: Etiopathogenesis, clinical manifestations and therapeutic options. Clin. Rheumatol. 2012, 32, 547–555. [Google Scholar] [CrossRef] [PubMed]
  25. Aicale, R.; Bisaccia, R.D.; Oliviero, A.; Oliva, F.; Maffulli, N. Current pharmacological approaches to the treatment of tendinopathy. Expert Opin. Pharmacother. 2020, 21, 1467–1477. [Google Scholar] [CrossRef]
  26. Frizziero, A.; Vittadini, F.; Fusco, A.; Giombini, A.; Masiero, S. Efficacy of eccentric exercise for lower limb tendinopathies in athletes. J. Sports Med. Phys. Fit. 2015, 56, 1352–1358. [Google Scholar]
  27. Frizziero, A.; Trainito, S.; Oliva, F.; Aldini, N.N.; Masiero, S.; Maffulli, N. The role of eccentric exercise in sport injuries rehabilitation. Br. Med Bull. 2014, 110, 47–75. [Google Scholar] [CrossRef] [Green Version]
  28. Frizziero, A.; Vittadini, F.; Barazzuol, M.; Gasparre, G.; Finotti, P.; Meneghini, A.; Maffulli, N.; Masiero, S. Extracorporeal shockwaves therapy versus hyaluronic acid injection for the treatment of painful non-calcific rotator cuff tendinopathies: Preliminary results. J. Sports Med. Phys. Fit. 2017, 57, 1162–1168. [Google Scholar] [CrossRef] [PubMed]
  29. Galletti, S.; Oliva, F.; Masiero, S.; Frizziero, A.; Galletti, R.; Schiavone, C.; Salini, V.; Abate, M. Sonoelastography in the diagnosis of tendinopathies: An added value. Muscle Ligaments Tendons J. 2015, 5, 325–330. [Google Scholar] [CrossRef]
  30. Frizziero, A.; Vittadini, F.; Bigliardi, D.; Costantino, C. The Role of Hyaluronic Acid Injection for the Treatment of Tendinopathy. Muscle Ligaments Tendons J. 2020, 10, 548. [Google Scholar] [CrossRef]
  31. Collins, T.; Alexander, D.; Barkatali, B. Platelet-rich plasma: A narrative review. EFORT Open Rev. 2021, 6, 225–235. [Google Scholar] [CrossRef]
  32. Kakkos, G.A.; Klontzas, M.E.; Koltsakis, E.; Karantanas, A.H. US-guided high-volume injection for Achilles tendinopathy. J. Ultrason. 2021, 21, e127–e133. [Google Scholar] [CrossRef]
  33. Arias-Vázquez, P.I.; Tovilla-Zárate, C.A.; González-Graniel, K.; Burad-Fonz, W.; González-Castro, T.B.; López-Narváez, M.L.; Castillo-Avila, R.G.; Arcila-Novelo, R. Efficacy of hypertonic dextrose infiltrations for pain control in rotator cuff tendinopathy: Systematic review and meta-analysis. Acta Reum. Port. 2021, 46, 156–170. [Google Scholar]
  34. Morath, O.; Beck, M.; Taeymans, J.; Hirschmüller, A. Sclerotherapy and prolotherapy for chronic patellar tendinopathies—A promising therapy with limited available evidence, a systematic review. J. Exp. Orthop. 2020, 7, 1–11. [Google Scholar] [CrossRef]
  35. Von Wehren, L.; Pokorny, K.; Blanke, F.; Sailer, J.; Majewski, M. Injection with autologous conditioned serum has better clinical results than eccentric training for chronic Achilles tendinopathy. Knee Surg. Sports Traumatol. Arthrosc. 2019, 27, 2744–2753. [Google Scholar] [CrossRef]
  36. Fernández, A.C.M.; Carballar, C.B.; Villafañe, J.H.; Pérez, M.M.; Pérez, J.L.; Díaz-Meco, R.; Jiménez, D.G.; Romero, E.A.S. A new ultrasound-guided percutaneous electrolysis and exercise treatment in patellar tendinopathy: Three case reports. Front. Biosci. 2021, 26, 1166–1175. [Google Scholar]
  37. Necas, J.; Bartosikova, L.; Brauner, P.; Kolar, J. Hyaluronic acid (hyaluronan): A review. Vet. Med. 2008, 53, 397–411. [Google Scholar] [CrossRef] [Green Version]
  38. Kaux, J.-F.; Samson, A.; Crielaard, J.-M. Hyaluronic acid and tendon lesions. Muscle Ligaments Tendons J. 2015, 5, 264–269. [Google Scholar] [CrossRef] [Green Version]
  39. Altman, R.; Hackel, J.; Niazi, F.; Shaw, P.; Nicholls, M. Efficacy and safety of repeated courses of hyaluronic acid injections for knee osteoarthritis: A systematic review. Semin. Arthritis Rheum. 2018, 48, 168–175. [Google Scholar] [CrossRef]
  40. Frizziero, A.; Maffulli, N.; Masiero, S.; Frizziero, L. Six-months pain relief and functional recovery after intra-articular injections with hyaluronic acid (mw 500–730 KDa) in trapeziometacarpal osteoarthritis. Muscle Ligaments Tendons J. 2019, 4, 256–261. [Google Scholar] [CrossRef]
  41. Foti, C.; Cisari, C.; Carda, S.; Giordan, N.; Rocco, A.; Frizziero, A.; Della Bella, G. A prospective observational study of the clinical efficacy and safety of intra-articular sodium hyaluronate in synovial joints with osteoarthritis. Eur. J. Phys. Rehabil. Med. 2011, 47, 407–415. [Google Scholar]
  42. Frizziero, A.; Vittadini, F.; Oliva, F.; Abatangelo, G.; Bacciu, S.; Berardi, A. IS Mu. LT Hyaluronic acid injections in musculoskeletal disorders guidelines. Muscles Ligaments Tendons J. 2018, 8, 364–398. [Google Scholar] [CrossRef] [Green Version]
  43. Osti, L.; Berardocco, M.; Di Giacomo, V.; Di Bernardo, G.; Oliva, F.; Berardi, A.C. 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. BMC Musculoskelet. Disord. 2015, 16, 284, Erratum in BMC Musculoskelet Disord. 2015, 16, 334. [Google Scholar] [CrossRef] [Green Version]
  44. Gallorini, M.; Berardi, A.C.; Berardocco, M.; Gissi, C.; Maffulli, N.; Cataldi, A.; Oliva, F. Hyaluronic acid increases tendon derived cell viability and proliferation in vitro: Comparative study of two different hyaluronic acid preparations by molecular weight. Muscle Ligaments Tendons J. 2017, 7, 208–214. [Google Scholar] [CrossRef]
  45. Salamanna, F.; Frizziero, A.; Pagani, S.; Giavaresi, G.; Curzi, D.; Falcieri, E.; Marini, M.; Abruzzo, P.M.; Martini, L.; Fini, M. Metabolic and cytoprotective effects ofin vivoperi-patellar hyaluronic acid injections in cultured tenocytes. Connect. Tissue Res. 2014, 56, 35–43. [Google Scholar] [CrossRef] [PubMed]
  46. Frizziero, A.; Salamanna, F.; Giavaresi, G.; Ferrari, A.; Martini, L.; Marini, M.; Veicsteinas, A.; Maffulli, N.; Masiero, S.; Fini, M.; et al. Hyaluronic acid injections protect patellar tendon from detraining-associated damage. Histol. Histopathol. 2015, 30, 1079–1088. [Google Scholar] [CrossRef]
  47. Meloni, F.; Milia, F.; Cavazzuti, M.; Doria, C.; Lisai, P.; Profili, S.; Meloni, G. 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. [Google Scholar] [CrossRef] [PubMed]
  48. Blaine, T.; Moskowitz, R.; Udell, J.; Skyhar, M.; Levin, R.; Friedlander, J.; Daley, M.; Altman, R. Treatment of Persistent Shoulder Pain with Sodium Hyaluronate: A Randomized, Controlled Trial. J. Bone Jt. Surg.-Am. Vol. 2008, 90, 970–979. [Google Scholar] [CrossRef]
  49. Fogli, M.; Giordan, N.; Mazzoni, G. Efficacy and safety of hyaluronic acid (500–730 kDa) Ultrasound-guided injections on painful tendinopathies: A prospective, open label, clinical study. Muscles Ligaments Tendons J. 2017, 7, 388–395. [Google Scholar] [CrossRef]
  50. Frizziero, A.; Oliva, F.; Vittadini, F.; Vetrano, M.; Bernetti, A.; Giordan, N.; Vulpiani, M.C.; Santilli, V.; Masiero, S.; Maffulli, N. Efficacy of ultrasound-guided hyaluronic acid injections in achilles and patellar tendinopathies: A prospective multicentric clinical trial. Muscles Ligaments Tendons J. 2019, 9, 305–313. [Google Scholar] [CrossRef] [Green Version]
  51. Kanchanathepsak, T.; Pichyangkul, P.; Suppaphol, S.; Watcharananan, I.; Tuntiyatorn, P.; Tawonsawatruk, T. Efficacy Comparison of Hyaluronic Acid and Corticosteroid Injection in Treatment of Trigger Digits: A Randomized Controlled Trial. J. Hand Surg. 2020, 25, 76–81. [Google Scholar] [CrossRef] [PubMed]
  52. Mohebbi, R.; Rezasoltani, Z.; Mir, M.; Mohebbi, M.; Vatandoost, S.; Esmaily, H. High-Versus Low-Molecular-Weight Hyaluronic Acid for the Treatment of Rotator Cuff Tendinopathy: A Triple-Blind Randomized Comparative Trial. Ann. Pharmacother. 2021, 55, 1203–1214. [Google Scholar] [CrossRef] [PubMed]
  53. Rezasoltani, Z.; Esmaily, H.; Dadarkhah, A.; Rousta, M.; Mohebbi, R.; Vashaei, F. Low Molecular-weight Hyaluronic Acid Versus Physiotherapy for the Treatment of Supraspinatus Tendinopathy: A Randomized Comparative Clinical Trial. J. Am. Acad. Orthop. Surg. 2021, 29, e979–e992. [Google Scholar] [CrossRef]
Figure 1. Summary of studies identification and selection.
Figure 1. Summary of studies identification and selection.
Jfmk 07 00003 g001
Table 1. Four papers were retrieved from literature search. Among them, 2 in vitro studies, 2 pre-clinical studies were selected.
Table 1. Four papers were retrieved from literature search. Among them, 2 in vitro studies, 2 pre-clinical studies were selected.
In Vitro Studies
ArticleSample DescriptionExperimental GroupControl GroupOutcome
Osti et al., 2015Human rotator cuff tears tendon-derived cells250 μg/mL GROUP-MW 500–730 KDa,
500 μg/mL GROUP-MW 500–730 KDa,
1000 μg/mL GROUP-MW 500–730 KDa,
250 μg/mL GROUP-MW 1000 KDa,
500 μg/mL GROUP-MW 1000 KDa,
1000 μg/mL GROUP-MW 1000 KDa,
1000 μg/mL GROUP-MW 1600 KDa,
1000 μg/mL GROUP-MW 2200 KDa,
Untreated cellsTenocyte viability and proliferation, Apoptosis induction, Immunofluorescence staining
Gallorini et al., 2019.Human supraspinatus tendon-derived cellsH2O2 GROUP: cultured cells were exposed to 2 mM H2O2
1000 KDa MW GROUP
2200 KDa MW GROUP
500–730 KDa MW GROUP
1600 KDa MW GROUP
H2O2+ HA GROUPS: cells were treated with different hyaluronic acids in the presence of 2 mM H2O2
Untreated cellsCytotoxicity-lactate dehydrogenase (LDH) release assay,
immunofluorescence staining of CD4, measurement of mitochondrial membrane potential (TMRE assay) by flow cytometry, cell lysis and protein extraction, immunoblotting, catalase activity
Pre-Clinical Studies
ArticleSample DescriptionExperimental GroupControl GroupOutcome
Salamanna et al., 201424 male Sprague-Dawley rats, aged 8 weeks, 280 ± 40 g body weight,
Patellar tendon
18 rats run on a treadmill 1 h a day, three times a week for 10 weeks. The other six rats underwent no training
TRAINED GROUP (6 rats)–euthanized at the end of the 10-week training.
Detrained group (12 rats)-euthanized after 10-week training + 4 weeks without exercise
DETRAINED-HA GROUP (6 of the 12 detrained rats)-at the end of the 1, 2, 3, and 4 weeks without exercise a peri-patellar infiltration of 300 μL of 20 mg/2 mL HA (500–730 KDa) was injected in the right patellar tendon
DETRAINED-NaCl GROUP (remaining 6 of the 12 detrained rats)-at the end of the 1, 2, 3, and 4 weeks without exercise, at the level of the right patellar tendon, received a peri-patellar infiltration of 300 μL of physiological saline solution (Fresenius Kabi)
UNTRAINED GROUP (6 rats)-euthanized without trainingTenocyte morphology and morphometric analysis (transmission electron microscopy), tenocyte viability and proliferation (Alamar blue dye test), tenocyte synthetic activity
Frizziero et al., 201524 male Sprague-Dawley rats, aged 8 weeks, 280 ± 40 g body weight,
Patellar tendon
18 rats run on a treadmill 1 h a day, three times a week for 10 weeks. The other six rats underwent no training
TRAINED GROUP (6 rats)–euthanized at the end of the 10-week training.
Detrained group (12 rats)-euthanized after 10-week training + 4 weeks without exercise
DETRAINED-HA GROUP (6 of the 12 detrained rats)-at the end of the 1, 2, 3, and 4 weeks without exercise a peri-patellar infiltration of 300 μL of 20 mg/2 mL HA (500–730 KDa, Fidia) was injected in the right patellar tendon
DETRAINED-NaCl GROUP (remaining 6 of the 12 detrained rats)-at the end of the 1, 2, 3, and 4 weeks without exercise, at the level of the right patellar tendon, received a peri-patellar infiltration of 300 μL of physiological saline solution (Fresenius Kabi)
UNTRAINED GROUP (6 rats)-euthanized without trainingTENDON ASSESSMENTS–modified semi-quantitative Movin grading scale (variables included: fiber structure, fiber arrangement, rounding of the nuclei, regional variation of cellularity, increased vascularity, collagen stainability, hyalinization), Tear density.
ENTHESIS ASSESSMENTS-semi-quantitative score (variables included: patellar enthesis structure; cell morphology in calcified cartilage; cell morphology in non-calcified cartilage; chondrocyte cluster formation in calcified cartilage; chondrocyte cluster formation in non-calcified cartilage; tidemark integrity between calcified and non-calcified cartilage; matrix staining; vascularization).
Table 2. Eight papers about clinical studies were selected from literature search.
Table 2. Eight papers about clinical studies were selected from literature search.
Clinical Studies
ArticleSample DescriptionPathologyInjection TechniqueExperimental GroupControl GroupFollow-UpOutcomeAdverse Events
Meloni et al., 2007Age = 31–71
N =56
Supraspinatus tendinosis (unresponsive to physical and medical therapy)5 periarticularinjections (1/week) under ultrasoundguide20 mg Sodium hyaluronate (MW = 500–730 KDa) together with 2 mL of 1% lidocaine and 2 mL of 0.9% sodium chloride solution4 mL of 0.9% sodium chloride solution, together with 2 mL of 1% lidocaineultrasound exam controls at 3, 6 and 12 months from the last injectionShoulder Range of motion (goniometer), Pain (VAS), degree of discomfortThere were no complications such as infections and no aggravations of symptoms compared with the pre-treatment state in either group
Blaine et al., 2008Age = 50–79
N = 79
Shoulder pain due to glenohumeral joint osteoarthritis, rotator cuff tear (partial or complete), and/or adhesive capsulitis for at least 6 months but less than 5 years’ refractory to standard treatments (physical therapy, corticosteroid injections and the administration of oral pain medications)5 intra-articular glenohumeralinjections (1/week)5 INJECTION HYALURONATE GROUP-2 mL sodium hyaluronate (molecular weight, 500–730 kDa)
3 INJECTION HYALURONATE GROUP-three injections of sodium hyaluronate (500–730 KDa) followed by two injections of phosphate-buffered saline solution
CONTROL GROUP-2 mL injections of phosphate-buffered saline solution7, 9, 13, 17, and 26 weeks after the initiation of therapyPain (VAS), maintenance of visual analogic scale pain relief, night pain improvement, patient global assessments, shoulder range of motion, general health questionnaire (short-form health survey-12)All treatments were well tolerated. The most frequently reported adverse event considered to be related to study treatment was injection-site pain.
Frizziero et al., 2017Age = 18–85
N = 34
Painful
non-calcific rotator cuff tendinopathies confirmed by Instrumental diagnosis (US or MRI)
3 sub-acromial space injections (1/week) under ultrasound guidance2 mL Hyaluronic acid (MW = 500–730 kDa)4 weeks (1/week) low energy extracorporeal shockwave therapy (MODULITH®); each session consisted of 1600 shots at a frequency of 4 Hz. The applied maximum energy (0.15 mJ/mm2) was adjusted on the basis of the patient’s tolerance, Mean session duration: 10 min.Parameters were evaluated at baseline (V0), at the end of the treatment (V1) and after 3 months of follow-up (V2)Disabilities of the arm (DASH score and Constant-Murley scales)No serious adverse events were recorded
Fogli et al., 2017Age = 33–59
N = 62
lateral elbow (26), Achilles (34) or patellar (11) tendinopathies3 peritendinous injections (1/week) under ecographic guide2 mL Hyaluronic acid (MW = 500–730 kDa) 7, 14, and 56 days after first treatmentPain (VAS), ultrasound assessment (changes in tendon thickness and neovascularization)No serious adverse events were recorded
Frizziero et al., 2019Age = 18–65
N = 35
Achilles (26) and patellar (9) mid-portion tendinopathies for ≥6 weeks duration and confirmed by ultrasound evaluation3 peritendinous injection (1/week) between paratenon and tendon under Ecographic guide20 mg/2 mL Hyaluronic acid (HyaloTend®, Fidia, MW = 500–730 KDa) 14, 45 and 90 days after the procedureVISA-A and VISA-P at 90 days of follow-up, pain (NRS-11), US parameters (tendon appearance and neovascularization), Patient Global Assessment (PGA), Clinical Observer Global Assessment (COGA), rescue medication consumption (paracetamol) and Health-related Quality of Life (EuroQoL EQ-5D-5L questionnaire)The treatment was well tolerated with only one adverse events in Achilles tendinopathy group, probably related to the injection procedure
Kanchanathepsak et al., 2020Age = 43–69
N = 66
Trigger digits1 peritendinous injection under ecographic guide1 mL hyaluronic acid (500–730 kD, 20 mg/2 mL)1 mL of 10 mg/mL of triamcinolone acetate1, 3, 6 monthsResidual symptoms (Quinell grading), Pain (VAS), disabilities of the Arm (DASH score)There was no major complication found in the study. Three patients complained about local discomfort at 1 week after injection without any sign of local inflammation.
Mohebbi et al., 2021Age = 16–70
N = 56
Rotator cuff tendinopathy (based on history, physical examination, and magnetic resonance imaging) with persistent shoulder pain for more than 6 weeks and less than 36 monthsSingle periarticular injection under ultrasound guidance20 mg (2 mL) LMW-HA 1% (500–730 kDa)20 mg (2 mL) HMW-HA 1% (>2000 kDa, Synogel, NikanTebKimia Pharmaceutical).baseline, 1, 4 weeks, and 3 months after the interventionsPain (VAS), shoulder range of motion (goniometer), quality of life (WHOQOL-Bref)No serious adverse events were recorded. Nine patients in the HMW-HA group and 3 participants in the LMW-HA group showed signs of inflammation at the site of injection. Overall, the LMW-HA group showed more tolerance to injection pain
Rezasoltani et al., 2021Age = 20–65
N = 51
Supraspinatus tendinopathy (based on history, physical examination, and magnetic resonance imaging) with persistent shoulder pain for more than 6 weeks and less than 3 monthsSingle subacromial injection under ultrasound guidance20 mg (2 mL) LMW-HA 1% (500–730 kDa).Physiotherapy group: 36 sessions, 3/week.Baseline, 1, 4, and 12 weeks postintervention.Pain (VAS), shoulder range of motion (goniometer), disabilities of the Arm (DASH score), quality of life (WHOQOL-Bref)No important adverse events occurred in the two groups
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Frizziero, A.; Vittadini, F.; Bigliardi, D.; Costantino, C. Low Molecular Weight Hyaluronic Acid (500–730 Kda) Injections in Tendinopathies—A Narrative Review. J. Funct. Morphol. Kinesiol. 2022, 7, 3. https://0-doi-org.brum.beds.ac.uk/10.3390/jfmk7010003

AMA Style

Frizziero A, Vittadini F, Bigliardi D, Costantino C. Low Molecular Weight Hyaluronic Acid (500–730 Kda) Injections in Tendinopathies—A Narrative Review. Journal of Functional Morphology and Kinesiology. 2022; 7(1):3. https://0-doi-org.brum.beds.ac.uk/10.3390/jfmk7010003

Chicago/Turabian Style

Frizziero, Antonio, Filippo Vittadini, Davide Bigliardi, and Cosimo Costantino. 2022. "Low Molecular Weight Hyaluronic Acid (500–730 Kda) Injections in Tendinopathies—A Narrative Review" Journal of Functional Morphology and Kinesiology 7, no. 1: 3. https://0-doi-org.brum.beds.ac.uk/10.3390/jfmk7010003

Article Metrics

Back to TopTop