Abstract
-
Current non-surgical managements of osteoarthritis (OA) do not change the clinical course or arrest the progression of the disease, while joint replacement is indicated for end-stage disease. Given these limitations, there is an unmet clinical demand for new treatment modalities that can improve the pain and quality of life of patients suffering from OA without surgery. The recent surge of interest in regenerative medicine (RM) for OA is based on these circumstances.
-
Unlike traditional medicine, RM products may be accompanied by many uncertainties and long-term consequences. Considering that OA directly affects quality of life rather than life and death, the ‘first do no harm’ principle is more important when applying RM technology to the disease.
-
Presently, culture-expanded mesenchymal stromal cells (MSCs) and orthobiologics, including bone marrow aspirate concentrate, stromal vascular fraction from adipose tissue and platelet-rich plasma have been applied to patients in clinical trials.
-
Results of randomized clinical trials using MSCs have demonstrated that structural improvement and reversal of the pathologic process in OA are not definitely shown, while symptomatic relief is apparent.
-
Orthobiologics seem to have efficiency comparable to those of culture-expanded MSCs. With the advantage of avoiding the approval process from regulation agencies, orthobiologics might provide a less expensive and handier option to culture-expanded MSCs.
-
High-quality data from a large number of patients and head-to-head comparisons of several RM products will be necessary to define the place of culture-expanded MSCs or orthobiologics for OA treatment and resolve the reimbursement issue.
Introduction
Regenerative medicine (RM) is an interdisciplinary field of medicine that focuses on biological repair, replacement or regeneration of injured or diseased tissues. According to Daar et al., RM is an ‘interdisciplinary field of research and clinical applications focused on repair, replacement or regeneration of cells, tissues or organs to restore impaired function resulting from any case’ (1, 2).
In a broad sense, RM comprises any medical endeavor to regenerate tissues, including small-molecule drugs, biological therapies, tissue-engineered devices or cell and gene therapy (3). Compared with traditional medicine, applying a product developed for RM to patients can be accompanied by greater uncertainties of long-term consequences, which have aroused ethical issues for clinical application. Using RM products in clinics may bring about complications that are not expected in conventional treatments. Given these differences from conventional medicine, traditional ethical standards established for conducting clinical research need reconsideration and adaptation for RM (4).
Considering that osteoarthritis (OA) directly affects quality of life rather than life and death, this ‘first do no harm’ principle is more important when applying RM technology to the disease. Potential serious adverse reactions, particularly an increased risk of carcinogenesis, should be assessed as the first priority. Long-term follow-up is particularly emphasized for individuals conducting clinical projects using cell-based products because of the possibility that these substances can persist in the body and potentially cause malignancy. It should be borne in mind that negligence and underreporting regarding negative outcomes can be especially costly and harmful when monitoring RM products (5).
Need for RM in OA
OA is a chronic disease manifested by joint pain and reduced mobility of affected patients, thereby lowering quality of life (6). Although OA is not a lethal disease by itself, it is nevertheless a serious disease that can exacerbate associated comorbidities, including cardiovascular and metabolic diseases. While OA pathology is generally summarized as the loss of joint cartilage and thickening of underlying subchondral bone, OA can be heterogeneous in clinical features and responses to different treatments. Unfortunately, current non-surgical managements for OA are essentially time-buying measures that neither change the clinical course nor arrest the progression of the disease. To find fundamental therapeutics for OA, several disease-modifying OA drugs (DMOADs) have been developed, with some of them undergoing clinical trials. However, none of them has yet brought clinical success demonstrated by both structural improvement and symptomatic relief. In advanced OA, surgical treatments, primarily joint replacement, are at present accepted as the standard treatment (7). While joint replacement may eventually deliver an impressive result for end-stage OA, not all patients are happy with surgical outcomes, with 20% of patients unsatisfied with their results. Joint replacement can also potentially bring about life-threatening complications, including pulmonary embolism and periprosthetic infection. For these reasons, many patients still prefer a non-surgical solution for OA if available. Therefore, there is an unmet clinical demand for new treatment modalities that can improve the pain and quality of life of patients suffering from OA without surgery, even at advanced stages. The recently increased interest in RM for OA is based on these circumstances (8).
Design of clinical studies on RM for osteoarthritis
Treatment target
It can be argued whether early- or advanced-stage OA patients should be preferred candidates for RM treatment: the former is based on grounds that RM can provide an efficient cure before irreversible damage occurs and the latter is based on a rationale that RM can be the last resort after all other available treatments have already been tried. In practice, it is mostly applied in Kellgren–Lawrence grade 2–3 patients who have clinically evident but not end-stage disease (for which joint replacement is indicated). In orthopedic disorders, including OA, structural damages as demonstrated by radiological changes are not always equal to patients’ clinical symptoms. To be considered as a disease-modifying regenerative treatment, both symptomatic improvement and evidence of tissue regeneration are necessary. Adequate basic science and preclinical research should be performed to establish the mode of action of an RM product before considering clinical application. Before RM interventions are brought to patients, careful pilot studies and randomized controlled trials (RCTs) are preferred to introduce evidence-based medicine as much as possible. Efforts should be made so that clinical studies on RM are flawless and properly designed (9).
The comparator in RM clinical trials for OA
In a clinical experiment to test the efficacy of a treatment method, randomization of treatment and control groups that allows an unbiased perspective of efficacy and safety is the favored method (10, 11). Given that an RCT provides the most reliable information on the ultimate value of a treatment, the control or comparators in RCTs can be designated as one of the three: i) standard conventional care, ii) placebo or iii) no intervention. Placebos are preferred to ensure validity of the clinical trial, particularly when clinical endpoints such as quality of life are assessed in the trial (12). By blinding, in which one gets a tested treatment or placebo, nonspecific effects such as placebo effects and reporting bias can be kept to a minimum (13). Particularly with RM trials, these confounding effects could be accentuated by excessive unjustified expectations on these ‘new’ technologies from both patients and investigators. When an RM intervention is a rather invasive one or when the trial is performed for patients with serious symptoms, conventional treatment is preferred as the control arm unless the intervention is an additive one to the main treatment.
Current status of RM in OA
Mesenchymal stromal/stem cells
Mesenchymal stromal/stem cells (MSCs) have been the most investigated RM products in translational studies or clinical applications for OA. While bone marrow-derived MSCs have been the mainstay sources, those isolated from fat or umbilical cord blood have also been tried as candidate sources because of their own merits. Anti-inflammatory and immunomodulatory properties of MSCs are known to provide joint environments better suited for the regeneration of damaged articular cartilage, either with transplanted MSCs or with endogenous cells mobilized by paracrine factors released from MSCs (14).
The original anticipation in cell therapy for cartilage regeneration was that implanted cells would survive, incorporate into chondral defects and differentiate to articular chondrocytes producing the extracellular matrix. Therefore, cell treatment would lead to structural improvement of the damaged joint by regenerating articular cartilage (15). However, it became obvious that intra-articularly (IA) administered cells survived only transiently and underwent rapid cell death (16). Most IA-administered MSCs survived from a few days to several weeks, depending on the mode of administration and the local environment (17, 18). Paracrine factors released from administered cells before undergoing apoptosis are thought to exert immunosuppressive and anti-inflammatory functions, as well as chondro-induction for host cells. Interestingly, stem cells survived longer when implanted focally rather than diffusely injected (19). Efforts to prolong cell survival and thereby exert extended paracrine effects and/or enhance engraftment with chondrogenic differentiation merit further inquiry and investigation.
Orthobiologics
Orthobiologics are biological substances known to promote repair/regeneration of musculoskeletal tissues. They are mostly autologous and comprise bone marrow aspirate concentrate (BMAC), stromal vascular fraction (SVF) from adipose tissue, platelet-rich plasma (PRP), platelet lysate, autologous conditioned serum or plasma, autologous protein solution and growth factor concentrates. Autologous BMAC, SVF and PRP have been used for OA treatment or cartilage regeneration, more frequently in private clinics than in academic centers. While the mode of action has not been thoroughly investigated as in MSCs, their regenerative effects can also be explained by paracrine factors secreted from cells (20, 21, 22, 23).
Because orthobiologics are categorized as minimally manipulated products, they do not undergo preliminary review by regulation agencies, which is the norm for the drug approval process and takes a long time and much cost in terms of human resources (24). Minimal regulation and increased availability of orthobiologics have led to aggressive marketing and overuse of these products in some countries. On the other hand, in the clinical setting, practitioners face scanty guiding information and the deficient evidence-based literature (25).
Exosomes or extracellular vesicles
The other new field in RM for OA is the use of exosomes or extracellular vesicles (eVs). They are lipid bilayer-limited particles that are naturally released from a cell and take part in cell-to-cell communication. Nucleic acids, proteins, metabolites, lipids and sometimes organelles from parent cells are contained in eVs. Given that the paracrine action of MSCs is principally attributed to secreted eVs, eVs isolated from MSCs can replace MSCs per se in RM for OA treatment. Being free from nuclear material, eVs can present unique advantages in passing through the regulatory process. On the other hand, very few translational studies, let alone clinical studies, have demonstrated the efficacy of eVs for OA treatment. In addition, isolating eVs in clinically meaningful doses is still a very costly procedure currently. However, advantages of eVs in passing the regulatory process and application will invigorate future research (26, 27).
Clinical results from RM products applied to treat OA
Culture-expanded MSCs
Sixteen peer-reviewed RCTs that used culture-expanded MSCs to treat OA were available from the literature search. Of them, nine studies investigated autologous MSCs while seven used allogeneic MSCs. Bone marrow-derived MSCs were used in seven RCTs (28, 29, 30, 31, 32, 33, 34, 35). Adipose tissue-derived MSCs were used in seven studies (36, 37, 38, 39, 40, 41, 42). Placenta tissues- or Wharton’s jelly-derived MSCs were used in two studies (43, 44). Fourteen studies showed clinical improvements compared with baseline values at the final follow-up. Thirteen trials found significantly better clinical results compared with control groups. Mean ages of treated patients ranged from 51 to 66 years, reflecting the age range with a high OA prevalence. In addition, the women (57%) were greater in number than the men (43%), owing to the higher OA prevalence in women (45). Cell doses differed widely from one study to another, ranging from 3.9 × 106 to 1.5 × 108 cells. Most studies showed that MSCs were effective at mid-to-high doses (>40 × 106 MSCs). There is not a clearly defined efficacious MSC dose range that reproducibly and consistently leads to better results compared to other doses (46).
Results of RCTs demonstrate that structural improvement and reversal of the pathologic process in OA are not definitely shown with IA-injected culture-expanded stem cells, while symptomatic relief improvement seems to be apparent. While long-term follow-up of patients is necessary to monitor ultimate effects of MSCs in OA treatment and detect unknown potential mechanisms, few studies have tracked patients beyond 1 year. While symptomatic relief is not sufficient to justify the high cost associated with RM treatments, including culture-expanded stem cells, definitive structural improvement that would last for years or decades and obviate or delay the need for joint arthroplasty is yet to be demonstrated for culture-expanded stem cell therapy (46).
BMAC
As BMAC can be harvested in outpatient settings without a culture process and does not need complex regulatory hurdles for application to the patient, it can provide an alternative to MSCs, provided that the therapeutic effect is comparable to that of MSCs. BMAC actually contains relatively few MSCs (only 0.001–0.01% of cellular contents) (47, 48). BMAC can be safely harvested from patients with few complications (49). Although several studies have investigated the use of BMAC in OA patients (50), evidence is insufficient to confirm its unequivocal efficacy. While no significant harm from BMAC application has been reported, its acceptance as a suitable therapy for knee OA is limited due to inadequate supporting data and variable results.
Two high-quality studies have demonstrated that a single IA injection of BMAC in platelet-poor plasma is not more effective than a saline placebo injection regarding function, MRI-based cartilage appearance or pain control (50, 51). On the contrary, another high-quality study showed that BMAC plus platelet product IA injection functionally outperformed exercise therapy, with all patients switching treatments within 3 months after the study began (52).
Several RCTs have compared BMAC injections with injections of other substances as comparators. Two studies concluded that both BMAC and hyaluronic acid (HA) improved pain and function at 1–2 years post-injection as compared with baseline, with BMAC achieving better results in pain scale than HA (53, 54). According to these articles, BMAC demonstrated comparable or slightly better results than HA for alleviating pain in particular, if not structural improvement (53, 54). One RCT that compared BMAC, HA and PRP reported clinical improvements with all three injectables, with BMAC showing superior results to the other two at a year after injection (55) while other RCT that compared BMAC and PRP found no difference between the two injectables after 2 years of follow-up (24). With the above findings, BMAC appears to exert similar effects on pain and function in OA patients as PRP does. However, whether BMAC is the orthobiologic superior to PRP remains unclear. Notably, a high-quality study that prospectively compared BMAC, PRP, HA and corticosteroid (CS) reported no differences in clinical parameters or significant changes in MRI findings after long-term follow-up over 1 year (56). These results suggest that strong placebo effects exist in injection therapy per se, raising questions about the long-term efficacy of BMAC.
In summary, BMAC injections do not show undisputedly superior results in pain relief or functional improvement compared to conventional therapies such as HA or CS, despite some studies reporting longer duration of effectiveness. The evidence of structural improvement, the original rationale of cell therapy, is also scarcely reported. In consideration of these findings, it is suggested that high-quality data from a large number of patients are needed to define the place of BMAC injection for OA treatment (7).
Stromal vascular fraction (SVF)
The SVF, a cellular mixture derived from lipoaspirate, can be obtained through mechanical or enzymatic separation (57, 58). The SVF comprises a diverse group of cells including adipose-derived MSCs (ADMSCs), also known as adipose stromal/stem cells (ASCs), preadipocytes, macrophages, lymphocytes, endothelial progenitors, fibroblasts, pericytes, smooth muscle cells and other uncharacterized cells, along with extracellular matrix (59, 60). The proportion of ASCs in the SVF may vary widely from less than 1% to more than 15%, depending on the donor’s age, sex, health status and the method utilized for harvesting it from adipose tissues (60). The SVF is particularly attractive due to its accessibility, low complication rate and minimal donor-site morbidity (60).
Compared to studies on BMAC, fewer randomized, prospective studies that offer unbiased information against placebo or other conventional treatment are available. The crucial advantage of the SVF over ASC is that it bypasses strict regulatory approval whereas its main disadvantage lies in its composition, which is a mixture of various cell types that can impede quality control. Consequently, allogeneic therapy with SVF is not recommended (61). Unlike BMAC, which requires only aspiration needle and centrifuge for processing, the production of the SVF requires a semi-laboratory facility to digest the tissue using enzyme treatment or a micro-fragmentation machine to mince the acquired fatty tissue. Subsequently, the SVF is less accessible to practicing clinicians compared to BMAC, even less so than PRP. Given this disadvantage, the value of the SVF will hinge on its competitive advantage in efficacy over other biologics.
Two controlled studies indicated that the SVF was superior to saline, which possesses a strong placebo effect merely from injection, in terms of pain and functional improvement. Compared to HA, the SVF demonstrates a more durable effect (62, 63). In addition, a few studies that utilized MRI in follow-up showed increased cartilage thickening with the SVF (64, 65, 66). On the other hand, a high-quality study demonstrated that none of the three orthobiologics – BMAC, SVF, PRP – outperformed corticosteroid injections in terms of functional improvement and MRI features (56). Two other studies comparing BMAC and SVF also found no differences in outcomes between the two orthobiologics (67, 68).
Injection volume, cell dose and product quality control are unsolved issues with the use of SVF for OA treatment. In addition, isolation processes, including collagenase digestion and mechanical dissociation, are not standardized and differ among studies. When compared to outcomes with culture-expanded ASCs, which also showed recognizable symptomatic improvement over a longer term than HA but failed to achieve significant structural improvement (38, 69, 70, 71, 72), the effects of SVF, which is a mixture of various cells, do not appear inferior. A study that compares SVF injection with ASC injection head-to-head may provide interesting data to resolve this issue.
Based on the limited literature available, it is suggested that SVF may offer better long-term symptomatic relief than placebo or HA, and its effects are comparable to those of BMAC. Prospective studies with improved control over cell isolation methods, dosing and patient selection are necessary to provide convincing evidence of SVF’s efficacy in treating OA.
PRP
PRP refers to plasma containing concentrated platelets. It is enriched in growth factors that can promote tissue repair, cellular growth and angiogenesis. It includes platelet-derived growth factor, transforming growth factor-beta and vascular endothelial growth factor (73). Like other orthobiologics, PRP has been considered as a disease-modifying therapeutic for OA due to its anti-inflammatory and tissue-regenerative potential (73). The process of obtaining PRP is simpler with less morbidity than the process of obtaining BMAC or SVF. PRP has been used in autologous base and associated with a low incidence of adverse effects (74, 75, 76).
Compared with BMAC and SVF, PRP has more published data on OA because it began to be applied earlier. Systematic reviews of randomized trials have shown a significant decrease in visual analog scale score for pain with improved functionality and quality of life following PRP treatment in knee OA patients (75, 76). On the other hand, PRP injection therapy did not significantly reduce pain symptoms in patients with hip OA. In addition, the analgesic effect of leukocyte-poor PRP was greater than that of leukocyte-rich PRP (75). While evidence remains mixed, PRP treatment was not associated with a significant increase in cartilage thickness (77). As with BMAC and SVF, the optimal dose and multiplicity of injection for PRP in knee OA remain debated. Inconsistencies remain on whether higher doses or repeated injections of PRP could enhance pain relief and improve functional results (74, 76).
Recent analysis of statistical fragility of RCTs assessing the use of PRP for treating knee OA reported that given statistical significance might be reversed with only a few changes in patient outcomes, meaning that the significance of results in these RCTs is not robust enough (78, 79). Of note, a large randomized, placebo-controlled clinical trial showed negative results of PRP. That study was performed for Kellgren and Lawrence grade 2 or 3 OA patients to evaluate effects of IA PRP injections on symptoms and joint structure with participant, injector and assessor blinded. IA injection of leukocyte-poor PRP compared to saline placebo injection did not result in a significant difference in symptoms or joint structure at 12 months (80). At present, on grounds of insufficient high-quality evidence about its efficacy for symptomatic and structural improvement, most clinical guidelines do not recommend PRP for knee OA. Guidelines emphasize the need for rigorous studies to justify the use of PRP over less expensive conventional treatments.
To sum up, though PRP shows promise, high-quality data that show unquestioned efficacy of PRP are insufficient and the variability in preparation methods, dosage and treatment protocols presents challenges in standardizing its clinical application. Future research should focus on RCTs that compare PRP in different formulations and combinations with established therapies to thoroughly evaluate its efficacy and establish guidelines.
Conclusion
As applying end products of RM development to patients can be accompanied by many uncertainties not seen in conventional medicine, long-term follow-up is important for individuals undergoing clinical trials using cell-based products. Before RM products are approved for clinical application to patients, careful pilot studies and RCTs are necessary to establish evidence-based medicine as much as possible.
Presently, culture-expanded MSCs and orthobiologics including BMAC, SVP and PRP have been applied to patients in clinical trials. Results of RCTs using culture-expanded MSCs have demonstrated that structural improvement and reversal of pathologic process in OA are not definitely shown while symptomatic relief seems to be apparent. Given that symptomatic relief is not sufficient to justify the high cost associated with culture-expanded stem cells, definitive structural improvement that could last for years/decades and obviate/delay the need for joint arthroplasty is yet to be demonstrated for culture-expanded MSCs therapy.
Orthobiologics may also offer better long-term symptomatic relief than control arms including HA, CS or placebo although a few high-quality studies have demonstrated no advantage over conventional treatments. Likewise, the evidence of structural improvement, the original rationale of cell therapy, is scarcely reported. Although direct head-to-head comparison is not available, efficacies of orthobiologics have been reported to be comparable to those of culture-expanded MSCs. With the advantage of avoiding the approval process from regulation agencies, orthobiologics might provide a less expensive and handier option to culture-expanded MSCs therapy. In consideration of these findings, it is suggested that high-quality data from a large number of patients and head-to-head comparison of several RM products will be necessary to define the place of culture-expanded MSCs or orthobiologics for OA treatment and resolve the reimbursement issue.
ICMJE Statement of Interest
The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the work.
Funding Statement
This work was supported by the research grant from the Korean Fund for RM (RS-2022-00070271).
References
- 1↑
de Windt TS , Niemansburg SL , Vonk LA , et al. Ethics in musculoskeletal regenerative medicine; guidance in choosing the appropriate comparator in clinical trials. Osteoarthr Cartil 2019 27 34–40. (https://doi.org/10.1016/j.joca.2018.08.022)
- 2↑
Daar AS & Greenwood HL . A proposed definition of regenerative medicine. J Tissue Eng Regen Med 2007 1 179–184. (https://doi.org/10.1002/term.20)
- 3↑
Cossu G , Birchall M , Brown T , et al. Lancet Commission: stem cells and regenerative medicine. Lancet 2018 391 883–910. (https://doi.org/10.1016/S0140-6736(17)31366-1)
- 4↑
Emanuel EJ , Wendler D & Grady C . What makes clinical research ethical? JAMA 2000 283 2701–2711. (https://doi.org/10.1001/jama.283.20.2701)
- 5↑
Saini P , Loke YK , Gamble C , et al. Selective reporting bias of harm outcomes within studies: findings from a cohort of systematic reviews. BMJ 2014 349 g6501. (https://doi.org/10.1136/bmj.g6501)
- 6↑
Felson DT , Zhang Y , Hannan MT , et al. The incidence and natural history of knee osteoarthritis in the elderly. The Framingham Osteoarthritis Study. Arthritis Rheum 1995 38 1500–1505. (https://doi.org/10.1002/art.1780381017)
- 7↑
Im GI . Bone marrow aspiration concentrate in the treatment of osteoarthritis: a review of its current clinical application. Tissue Eng Regen Med 2025 22 159–166. (https://doi.org/10.1007/s13770-024-00693-7)
- 8↑
Im GI . Current status of regenerative medicine in osteoarthritis. Bone Joint Res 2021 10 134–136. (https://doi.org/10.1302/2046-3758.102.BJR-2020-0517.R1)
- 9↑
Niemansburg SL , van Delden JJ , Oner FC , et al. Ethical implications of regenerative medicine in orthopedics: an empirical study with surgeons and scientists in the field. Spine J 2014 14 1029–1035. (https://doi.org/10.1016/j.spinee.2013.10.016)
- 10↑
European Medicines Agency . Choice of control group in clinical trials. 2001. (https://www.ema.europa.eu/en/ich-e10-choice-control-group-clinical-trials-scientific-guideline)
- 11↑
Food and Drug Administration . Guidance for industry E10 choice of control group and related issues in clinical trials. 2001. (https://www.fda.gov/regulatory-information/search-fda-guidance-documents/e10-choice-control-group-and-related-issues-clinical-trials)
- 12↑
Schulz KF & Grimes DA . Blinding in randomised trials: hiding who got what. Lancet 2002 359 696–700. (https://doi.org/10.1016/S0140-6736(02)07816-9)
- 13↑
Brittberg M , Lindahl A , Nilsson A , et al. Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. N Engl J Med 1994 331 889–895. (https://doi.org/10.1056/NEJM199410063311401)
- 14↑
Wang J , Zhou L , Zhang Y , et al. Mesenchymal stem cells - a promising strategy for treating knee osteoarthritis. Bone Joint Res 2020 9 719–728. (https://doi.org/10.1302/2046-3758.910.BJR-2020-0031.R3)
- 15↑
Im G-I & Kim T-K . Regenerative therapy for osteoarthritis: a perspective. Int J Stem Cells 2020 13 177–181. (https://doi.org/10.15283/ijsc20069)
- 16↑
Kim M-K , Ha C-W , In Y , et al. A multicenter, double-blind, phase III clinical trial to evaluate the efficacy and safety of a cell and gene therapy in knee osteoarthritis patients. Hum Gene Ther Clin Dev 2018 29 48–59. (https://doi.org/10.1089/humc.2017.249)
- 17↑
Im G-I . Perspective on intra-articular injection cell therapy for osteoarthritis treatment. Tissue Eng Regen Med 2019 16 357–363. (https://doi.org/10.1007/s13770-018-00176-6)
- 18↑
Im G-I . Tissue engineering in osteoarthritis: current status and prospect of mesenchymal stem cell therapy. BioDrugs 2018 32 183–192. (https://doi.org/10.1007/s40259-018-0276-3)
- 19↑
Park YB , Ha CW , Kim JA , et al. Single-stage cell-based cartilage repair in a rabbit model: cell tracking and in vivo chondrogenesis of human umbilical cord blood-derived mesenchymal stem cells and hyaluronic acid hydrogel composite. Osteoarthr Cartil 2017 25 570–580. (https://doi.org/10.1016/j.joca.2016.10.012)
- 20↑
Madhi MI , Yausep OE , Khamdan K , et al. The use of PRP in treatment of Achilles Tendinopathy: a systematic review of literature. Study design: systematic review of literature. Ann Med Surg 2020 55 320–326. (https://doi.org/10.1016/j.amsu.2020.04.042)
- 21↑
Barnett J , Bernacki MN , Kainer JL , et al. The effects of regenerative injection therapy compared to corticosteroids for the treatment of lateral Epicondylitis: a systematic review and meta-analysis. Arch Physiother 2019 9 12. (https://doi.org/10.1186/s40945-019-0063-6)
- 22↑
Johal H , Khan M , Yung SP , et al. Impact of platelet-rich plasma use on pain in orthopaedic surgery: a systematic review and meta-analysis. Sports Health 2019 11 355–366. (https://doi.org/10.1177/1941738119834972)
- 23↑
Li A , Wang H , Yu Z , et al. Platelet-rich plasma vs corticosteroids for elbow epicondylitis: a systematic review and meta-analysis. Medicine 2019 98 e18358. (https://doi.org/10.1097/MD.0000000000018358)
- 24↑
Food and Drug Administration . Regulatory considerations for human cells, tissues, and cellular and tissue-based products: minimal manipulation and homologous use; guidance for industry and food and drug administration staff; availability. 2020. (https://www.fda.gov/regulatory-information/search-fda-guidance-documents/regulatory-considerations-human-cells-tissues-and-cellular-and-tissue-based-products-minimal)
- 25↑
Marenah M , Li J , Kumar A , et al. Quality assurance and adverse event management in regenerative medicine for knee osteoarthritis: current concepts. J Clin Orthop Trauma 2019 10 53–58. (https://doi.org/10.1016/j.jcot.2018.09.005)
- 26↑
Miyaki S & Lotz MK . Extracellular vesicles in cartilage homeostasis and osteoarthritis. Curr Opin Rheumatol 2018 30 129–135. (https://doi.org/10.1097/BOR.0000000000000454)
- 27↑
Mianehsaz E , Mirzaei HR , Mahjoubin-Tehran M , et al. Mesenchymal stem cell-derived exosomes: a new therapeutic approach to osteoarthritis? Stem Cell Res Ther 2019 10 340. (https://doi.org/10.1186/s13287-019-1445-0)
- 28↑
Bastos R , Mathias M , Andrade R , et al. Intra-articular injection of culture-expanded mesenchymal stem cells with or without addition of platelet-rich plasma is effective in decreasing pain and symptoms in knee osteoarthritis: a controlled, double-blind clinical trial. Knee Surg Sports Traumatol Arthrosc 2020 28 1989–1999. (https://doi.org/10.1007/s00167-019-05732-8)
- 29↑
Emadedin M , Labibzadeh N , Liastani MG , et al. Intra-articular implantation of autologous bone marrow–derived mesenchymal stromal cells to treat knee osteoarthritis: a randomized, triple-blind, placebo-controlled phase 1/2 clinical trial. Cytotherapy 2018 20 1238–1246. (https://doi.org/10.1016/j.jcyt.2018.08.005)
- 30↑
Lamo-Espinosa JM , Mora G , Blanco JF , et al. Intra-articular injection of two different doses of autologous bone marrow mesenchymal stem cells versus hyaluronic acid in the treatment of knee osteoarthritis: long-term follow up of a multicenter randomized controlled clinical trial (phase I/II). J Transl Med 2018 16 213. (https://doi.org/10.1186/s12967-018-1591-7)
- 31↑
Lamo-Espinosa JM , Blanco JF , Sánchez M , et al. Phase II multicenter randomized controlled clinical trial on the efficacy of intra-articular injection of autologous bone marrow mesenchymal stem cells with platelet rich plasma for the treatment of knee osteoarthritis. J Transl Med 2020 18 356. (https://doi.org/10.1186/s12967-020-02530-6)
- 32↑
Lamo-Espinosa JM , Mora G , Blanco JF , et al. Intra-articular injection of two different doses of autologous bone marrow mesenchymal stem cells versus hyaluronic acid in the treatment of knee osteoarthritis: multicenter randomized controlled clinical trial (phase I/II). J Transl Med 2016 14 246. (https://doi.org/10.1186/s12967-016-0998-2)
- 33↑
Wong KL , Lee KBL , Tai BC , et al. Injectable cultured bone marrow-derived mesenchymal stem cells in varus knees with cartilage defects undergoing high tibial osteotomy: a prospective, randomized controlled clinical trial with 2 years' follow-up. Arthroscopy 2013 29 2020–2028. (https://doi.org/10.1016/j.arthro.2013.09.074)
- 34↑
Gupta PK , Chullikana A , Rengasamy M , et al. Efficacy and safety of adult human bone marrow-derived, cultured, pooled, allogeneic mesenchymal stromal cells (Stempeucel®): preclinical and clinical trial in osteoarthritis of the knee joint. Arthritis Res Ther 2016 18 301. (https://doi.org/10.1186/s13075-016-1195-7)
- 35↑
Vega A , Martín-Ferrero MA , Del Canto F , et al. Treatment of knee osteoarthritis with allogeneic bone marrow mesenchymal stem cells: a randomized controlled trial. Transplantation 2015 99 1681–1690. (https://doi.org/10.1097/TP.0000000000000678)
- 36↑
Lee WS , Kim HJ , Kim KI , et al. Intra-articular injection of autologous adipose tissue-derived mesenchymal stem cells for the treatment of knee osteoarthritis: a phase IIb, randomized, placebo-controlled clinical trial. Stem Cells Transl Med 2019 8 504–511. (https://doi.org/10.1002/sctm.18-0122)
- 37↑
Freitag J , Bates D , Wickham J , et al. Adipose-derived mesenchymal stem cell therapy in the treatment of knee osteoarthritis: a randomized controlled trial. Regen Med 2019 14 213–230. (https://doi.org/10.2217/rme-2018-0161)
- 38↑
Lu L , Dai C , Zhang Z , et al. Treatment of knee osteoarthritis with intra-articular injection of autologous adipose-derived mesenchymal progenitor cells: a prospective, randomized, double-blind, active-controlled, phase IIb clinical trial. Stem Cell Res Ther 2019 10 143. (https://doi.org/10.1186/s13287-019-1248-3)
- 39↑
Chen CF , Hu CC , Wu CT , et al. Treatment of knee osteoarthritis with intra-articular injection of allogeneic adipose-derived stem cells (ADSCs) ELIXCYTE®: a phase I/II, randomized, active-control, single-blind, multiple-center clinical trial. Stem Cell Res Ther 2021 12 562. (https://doi.org/10.1186/s13287-021-02631-z)
- 40↑
Kuah D , Sivell S , Longworth T , et al. Safety, tolerability and efficacy of intra-articular Progenza in knee osteoarthritis: a randomized double-blind placebo-controlled single ascending dose study. J Transl Med 2018 16 49. (https://doi.org/10.1186/s12967-018-1420-z)
- 41↑
Kim KI , Lee MC , Lee JH , et al. Clinical efficacy and safety of the intra-articular injection of autologous adipose-derived mesenchymal stem cells for knee osteoarthritis: a phase III, randomized, double-blind, placebo-controlled trial. Am J Sports Med 2023 51 2243–2253. (https://doi.org/10.1177/03635465231179223)
- 42↑
Sadri B , Hassanzadeh M , Abolfazl Bagherifard A , et al. Cartilage regeneration and inflammation modulation in knee osteoarthritis following injection of allogeneic adipose-derived mesenchymal stromal cells: a phase II, triple-blinded, placebo controlled, randomized trial. Stem Cell Res Ther 2023 14 162. (https://doi.org/10.1186/s13287-023-03359-8)
- 43↑
Matas J , Orrego M , Amenabar D , et al. Umbilical cord-derived mesenchymal stromal cells (MSCs) for knee osteoarthritis: repeated MSC dosing is superior to a single MSC dose and to hyaluronic acid in a controlled randomized phase I/II trial. Stem Cells Transl Med 2019 8 215–224. (https://doi.org/10.1002/sctm.18-0053)
- 44↑
Khalifeh Soltani S , Forogh B , Ahmadbeigi N , et al. Safety and efficacy of allogenic placental mesenchymal stem cells for treating knee osteoarthritis: a pilot study. Cytotherapy 2019 21 54–63. (https://doi.org/10.1016/j.jcyt.2018.11.003)
- 45↑
Moretti B , Spinarelli A , Varrassi G , et al. Influence of sex and gender on the management of late-stage knee osteoarthritis. Musculoskelet Surg 2022 106 457–467. (https://doi.org/10.1007/s12306-021-00725-8)
- 46↑
Im GI . Clinical updates in mesenchymal stromal cell therapy for osteoarthritis treatment. Expert Opin Biol Ther 2025 25 187–195. (https://doi.org/10.1080/14712598.2024.2446612)
- 47↑
Pittenger MF , Mackay AM , Beck SC , et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999 284 143–147. (https://doi.org/10.1126/science.284.5411.143)
- 48↑
Sampson S , Bemden AB-V & Aufiero D . Stem cell therapies for treatment of cartilage and bone disorders: osteoarthritis, avascular necrosis, and non-union fractures. PM R 2015 7 S26–S32. (https://doi.org/10.1016/j.pmrj.2015.01.023)
- 49↑
Aneja A , Adams SB , Foster JA , et al. AAOS technology overview summary: concentrated bone marrow aspirate for knee osteoarthritis. J Am Acad Orthop Surg 2023 31 e9–e13. (https://doi.org/10.5435/JAAOS-D-22-00228)
- 50↑
Shapiro SA , Arthurs JR , Heckman MG , et al. Quantitative T2 MRI mapping and 12-month follow-up in a randomized, blinded, placebo controlled trial of bone marrow aspiration and concentration for osteoarthritis of the knees. Cartilage 2019 10 432–443. (https://doi.org/10.1177/1947603518796142)
- 51↑
Shapiro SA , Kazmerchak SE , Heckman MG , et al. A prospective, single-blind, placebo-controlled trial of bone marrow aspirate concentrate for knee osteoarthritis. Am J Sports Med 2017 45 82–90. (https://doi.org/10.1177/0363546516662455)
- 52↑
Centeno C , Sheinkop M , Dodson E , et al. A specific protocol of autologous bone marrow concentrate and platelet products versus exercise therapy for symptomatic knee osteoarthritis: a randomized controlled trial with 2 year follow-up. J Transl Med 2018 16 355. (https://doi.org/10.1186/s12967-018-1736-8)
- 53↑
Goncars V , Jakobsons E , Blums K , et al. The comparison of knee osteoarthritis treatment with single-dose bone marrow-derived mononuclear cells vs. hyaluronic acid injections. Medicina 2017 53 101–108. (https://doi.org/10.1016/j.medici.2017.02.002)
- 54↑
Boffa A , Di Martino A , Andriolo L , et al. Bone marrow aspirate concentrate injections provide similar results versus viscosupplementation up to 24 months of follow-up in patients with symptomatic knee osteoarthritis. A randomized controlled trial. Knee Surg Sports Traumatol Arthrosc 2022 30 3958–3967. (https://doi.org/10.1007/s00167-021-06793-4)
- 55↑
Dulic O , Rasovic P , Lalic I , et al. Bone marrow aspirate concentrate versus platelet rich plasma or hyaluronic acid for the treatment of knee osteoarthritis. Medicina 2021 57 1193. (https://doi.org/10.3390/medicina57111193)
- 56↑
Mautner K , Gottschalk M , Boden SD , et al. Cell-based versus corticosteroid injections for knee pain in osteoarthritis: a randomized phase 3 trial. Nat Med 2023 29 3120–3126. (https://doi.org/10.1038/s41591-023-02632-w)
- 57↑
Cao L , Xiaoming F , Zhang Q , et al. An optimized method for adipose stromal vascular fraction isolation and its application in fat grafting. Aesthetic Plast Surg 2022 46 2500–2508. (https://doi.org/10.1007/s00266-021-02738-x)
- 58↑
Senesi L , De Francesco F , Farinelli L , et al. Mechanical and enzymatic procedures to isolate the stromal vascular fraction from adipose tissue: preliminary results. Front Cell Dev Biol 2019 7 88. (https://doi.org/10.3389/fcell.2019.00088)
- 59↑
Sun Y , Chen S , Zhang X , et al. Significance of cellular cross-talk in stromal vascular fraction of adipose tissue in neovascularization. Arterioscler Thromb Vasc Biol 2019 39 1034–1044. (https://doi.org/10.1161/ATVBAHA.119.312425)
- 60↑
Ude CC , Shah S , Ogueri KS , et al. Stromal vascular fraction for osteoarthritis of the knee regenerative engineering. Regen Eng Transl Med 2022 8 210–224. (https://doi.org/10.1007/s40883-021-00226-x)
- 61↑
Im GI . Current status of stromal vascular fraction from adipose tissue in the clinical application for osteoarthritis treatment. Tissue Eng Regen Med 2025 [In press]. (https://doi.org/10.1007/s13770-025-00722-z)
- 62↑
Garza JR , Campbell RE , Tjoumakaris FP , et al. Clinical efficacy of intra-articular mesenchymal stromal cells for the treatment of knee osteoarthritis: a double-blinded prospective randomized controlled clinical trial. Am J Sports Med 2020 48 588–598. (https://doi.org/10.1177/0363546519899923)
- 63↑
Tantuway V , Thomas W , Parikh MB , et al. Clinical outcome of minimally manipulated, mechanically isolated autologous adipose tissue-derived stromal vascular fraction (Sahaj Therapy®) in knee osteoarthritis-randomized controlled trial. Indian J Orthop 2023 57 1646–1658. (https://doi.org/10.1007/s43465-023-00981-9)
- 64↑
Ren B , Chang Y , Liu R , et al. Clinical phase I/II trial of SVF therapy for cartilage regeneration: a cellular therapy with novel 3D MRI imaging for evaluating chondral defect of knee osteoarthritis. Front Cell Dev Biol 2023 11 1106279. (https://doi.org/10.3389/fcell.2023.1106279)
- 65↑
Zhang S , Xu H , He B , et al. Mid-term prognosis of the stromal vascular fraction for knee osteoarthritis: a minimum 5-year follow-up study. Stem Cell Res Ther 2022 13 105. (https://doi.org/10.1186/s13287-022-02788-1)
- 66↑
Zhang Y , Bi Q , Luo J , et al. The effect of autologous adipose-derived stromal vascular fractions on cartilage regeneration was quantitatively evaluated based on the 3D-FS-SPGR sequence: a clinical trial study. BioMed Res Int 2022 2022 2777568. (https://doi.org/10.1155/2022/2777568)
- 67↑
Pintore A , Notarfrancesco D , Zara A , et al. Intra-articular injection of bone marrow aspirate concentrate (BMAC) or adipose-derived stem cells (ADSCs) for knee osteoarthritis: a prospective comparative clinical trial. J Orthop Surg Res 2023 18 350. (https://doi.org/10.1186/s13018-023-03841-2)
- 68↑
Mautner K , Gottschalk M , Boden SD , et al. Cell-based versus corticosteroid injections for knee pain in osteoarthritis: a randomized phase 3 trial. Nat Med 2023 29 3120–3126. (https://doi.org/10.1038/s41591-023-02632-w)
- 69↑
Kim KI , Lee MC , Lee JH , et al. Clinical efficacy and safety of the intra-articular injection of autologous adipose-derived mesenchymal stem cells for knee osteoarthritis: a phase III, randomized, double-blind, placebo-controlled trial. Am J Sports Med 2023 51 2243–2253. (https://doi.org/10.1177/03635465231179223)
- 70↑
Lee WS , Kim HJ , Kim KI , et al. Intra-articular injection of autologous adipose tissue-derived mesenchymal stem cells for the treatment of knee osteoarthritis: a phase IIb, randomized, placebo-controlled clinical trial. Stem Cells Transl Med 2019 8 504–511. (https://doi.org/10.1002/sctm.18-0122)
- 71↑
Freitag J , Bates D , Wickham J , et al. Adipose derived mesenchymal stem cell therapy in the treatment of knee osteoarthritis: a randomized controlled trial. Regen Med 2019 14 213–230. (https://doi.org/10.2217/rme-2018-0161)
- 72↑
Song Y , Du H , Dai C , et al. Human adipose-derived mesenchymal stem cells for osteoarthritis: a pilot study with long-term follow-up and repeated injections. Regen Med 2018 13 295–307. (https://doi.org/10.2217/rme-2017-0152)
- 73↑
Verma R , Kumar S , Garg P , et al. Platelet-rich plasma: a comparative and economical therapy for wound healing and tissue regeneration. Cell Tissue Bank 2023 24 285–306. (https://doi.org/10.1007/s10561-022-10039-z)
- 74↑
Khalilizad M , Emadian ST & Marzban Abbas Abadi M . Comparative efficacy of different doses of platelet-rich plasma injection in the treatment of knee osteoarthritis: a systematic review and network meta-analysis. J Orthop Surg Res 2025 20 221. (https://doi.org/10.1186/s13018-025-05650-1)
- 75↑
Xiong Y , Gong C , Peng X , et al. Efficacy and safety of platelet-rich plasma injections for the treatment of osteoarthritis: a systematic review and meta-analysis of randomized controlled trials. Front Med 2023 10 1204144. (https://doi.org/10.3389/fmed.2023.1204144)
- 76↑
Hong M , Cheng C , Sun X , et al. Efficacy and safety of intra-articular platelet-rich plasma in osteoarthritis knee: a systematic review and meta-analysis. BioMed Res Int 2021 2021 2191926. (https://doi.org/10.1155/2021/2191926)
- 77↑
Prodromidis AD , Charalambous CP , Moran E , et al. The role of platelet-rich plasma (PRP) intraarticular injections in restoring articular cartilage of osteoarthritic knees. A systematic review and meta-analysis. Osteoarthr Cartil Open 2022 4 100318. (https://doi.org/10.1016/j.ocarto.2022.100318)
- 78↑
Chan JP , Vrla M , Thompson C , et al. Statistical fragility of randomized controlled trials evaluating platelet-rich plasma use for knee osteoarthritis: a systematic review. Orthop J Sports Med 2023 11 23259671231187894. (https://doi.org/10.1177/23259671231187894)
- 79↑
Lameire DL , Abdel KH , Sritharan P , et al. Randomized controlled trials assessing continuous outcomes for the use of platelet-rich plasma in knee osteoarthritis are statistically fragile: a systematic review. Arthroscopy 2024 S0749-8063 00779-5. (https://doi.org/10.1016/j.arthro.2024.09.054)
- 80↑
Bennell KL , Paterson KL , Metcalf BR , et al. Effect of intra-articular platelet-rich plasma vs placebo injection on pain and medial tibial cartilage volume in patients with knee osteoarthritis: the RESTORE randomized clinical trial. JAMA 2021 326 2021–2030. (https://doi.org/10.1001/jama.2021.19415)