Next Article in Journal
Results of Adjustable Trans-Obturator Male System in Patients with Prostate Cancer Treated with Prostatectomy and Radiotherapy: A Multicenter Study
Next Article in Special Issue
SuperPATH—Current Status of Evidence and Further Investigations: A Scoping Review and Quality Assessment
Previous Article in Journal
Virtual Non-Contrast Spectral CT in Renal Masses: Is It Time to Discard Conventional Unenhanced Phase?
Previous Article in Special Issue
Office-Based Intraosseous Infiltrations of PRGF as an Effective Treatment for Knee Osteoarthritis: A Retrospective Observational Clinical Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

A Worldwide Analysis of Adipose-Derived Stem Cells and Stromal Vascular Fraction in Orthopedics: Current Evidence and Applications

by
Robert Ossendorff
1,*,†,
Alessandra Menon
2,3,4,†,
Frank A. Schildberg
1,
Pietro S. Randelli
2,3,5,
Sebastian Scheidt
1,
Christof Burger
1,
Dieter C. Wirtz
1 and
Davide Cucchi
1
1
Department of Orthopaedics and Trauma Surgery, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
2
Laboratory of Applied Biomechanics, Department of Biomedical Sciences for Health, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
3
U.O.C. 1° Clinica Ortopedica, ASST Gaetano Pini-CTO, Piazza Cardinal Ferrari 1, 20122 Milan, Italy
4
Dipartimento di Scienze Cliniche e di Comunità, Scuola di Specializzazione in Statistica Sanitaria e Biometria, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
5
Research Center for Adult and Pediatric Rheumatic Diseases (RECAP-RD), Department of Biomedical Sciences for Health, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2023, 12(14), 4719; https://doi.org/10.3390/jcm12144719
Submission received: 2 May 2023 / Revised: 15 June 2023 / Accepted: 11 July 2023 / Published: 17 July 2023
(This article belongs to the Special Issue Osteoarthritis: Diagnosis and Therapeutic Approaches)

Abstract

:
The biological enhancement of tissue regeneration and healing is an appealing perspective in orthopedics. We aimed to conduct a systematic review to describe the global distribution of studies investigating the use of adipose tissue derivates in orthopedics and to provide information on their quality and on the products available. The quality of the included studies was assessed using the modified Coleman Methodology Score (mCMS) and the Cochrane risk-of-bias tool for randomized trials. Eighty-two studies were included, with a total of 3594 patients treated. In total, 70% of the studies investigated the treatment of knee disorders, predominantly osteoarthritis; 26% of all studies dealt with expanded adipose-derived stem/stromal cells (ADSCs), 72% of which had stromal vascular fraction (SVF); 70% described the injection of adipose tissue derivates into the affected site; and 24% described arthroscopies with the addition of adipose tissue derivates. The mean mCMS for all studies was 51.7 ± 21.4 points, with a significantly higher score for the studies dealing with expanded ADSCs compared to those dealing with SVF (p = 0.0027). Our analysis shows high heterogeneity in terms of the types of performed procedures as well as the choice and processing of adipose tissue derivates.

1. Introduction

Strategies to enhance tissue regeneration and healing are appealing perspectives for orthopedic surgeons, aiming to reduce pain in degenerative joint conditions, postpone replacement procedures, restore painless function in tendon pathologies, and to improve results after reconstructive approaches. Efficient and effective healing is necessary to restore normal function after acute injuries or degenerative changes to cartilage and tendon tissue, but these tissues have limited natural healing capacity; therefore, biological enhancements, such as growth factors, stem cells, bioactive scaffolds, and tissue engineering approaches could play a crucial role by helping to overcome the limited regeneration potential of specific tissues where natural healing is slow or challenging. Furthermore, by tailoring treatments to individual patient needs, such as using autologous stem cells or customized tissue engineering scaffolds, a personalized approach can be achieved, with improved outcomes and reduced risks of adverse reactions [1,2,3,4,5,6].
New biological solutions have been proposed to achieve these goals. First, growth factors were proposed as a possible solution to stimulate tissue regeneration, followed by platelet-rich plasma (PRP), which was recognized as a possible autologous source of these molecules. Finally, mesenchymal stem/stromal cells (MSCs) gained popularity in regenerative medicine for their potential to enhance tissue healing. MSCs are believed to enhance tissue healing through the stimulation of local cells via the paracrine mechanism and anti-inflammatory and/or immunomodulatory activity, thus creating a suitable microenvironment for tissue repair and regeneration [7,8,9,10,11,12,13,14,15]. In recent years, the adipose tissue and its stromal vascular fraction (SVF) have been demonstrated to provide an easily accessible source for these cells and modulatory substances, with promising results in preclinical models [1]. Considering the low harvest morbidity for adipose tissue and its relative abundance in easily accessible regions of the human body, clinical publications describing the treatment of numerous orthopedic conditions with adipose tissue derivates have flourished. Furthermore, future applications of adipose tissue derivates in combination with bioengineering innovation, such as 3D bioprinting and computational simulation, hold great potential to advance regenerative medicine for osteoarthritis and tissue repair [16,17].
Lipoaspirate or tissue biopsy allow the effective and safe harvest of adipose tissue from the subcutaneous layers, which can then be processed in different ways to obtain specific therapeutic products [18]. These products can be distinguished in three major groups: expanded MSCs, enzymatically extracted SVF, and mechanically extracted SVF (microfragmented or microfractured adipose tissue). Expanded MSCs are obtained via the enzymatic digestion of adipose tissue and are cultured to obtain a well-characterized product with a high and pure concentration of MSCs. Non-expanded adipose tissue derivates contain a cocktail of MSCs, platelets, immune cells, cytokines, and growth factors, defined as SVF, and can be obtained via enzymatic or mechanical methods from unprocessed fat tissue [19]. However, higher regulative demands and costs currently limit the clinical application of expanded MSCs to a few specialized centers. Conversely, the popularity of non-expanded adipose tissue derivates has increased due to their high cost-effectiveness and the possibility of a point-of care application, although the cell yield and characterization has not yet been investigated for most available products. Nonetheless, clinical applications have flourished across the globe in non-specialized settings, leading to a multitude of clinical studies, not always following strict methodological rules. As a result, many publications dealing with different adipose tissue derivates and using different extraction protocols and commercially available devices are available, with publication quality ranging from rigorous clinical trials to case series of questionable scientific value. No current analysis of the quality of the volume of published literature has been performed.
The quality assessment of publications is important to advance scientific knowledge and its practical applications. In particular, the evaluation of methodologic quality is necessary to determine the scientific validity and robustness of research studies by evaluating the study design, methodology, data collection, analysis, and interpretation. Such an assessment has not yet been performed for studies dealing with human-adipose-derived stem/stromal cells (ADSCs) or SVF (including microfragmented adipose tissue) applications to ensure proper research is conducted using appropriate methods to provide meaningful and reliable results. We aimed to conduct a systematic review to analyze the global distribution of studies investigating the use of adipose tissue derivatives in orthopedics, to assess the quality thereof and provide information on the available products.

2. Materials and Methods

The Preferred Reporting Items for Systematic reviews and Meta-Analysis statement (PRISMA) was followed as a guideline for the study [20]. This study did not require ethical committee approval. The review was registered on the PROSPERO database (Registration number: CRD42022339795).

2.1. Search Strategy

To analyze the use of adipose tissue derivates in orthopedic surgery, a systematic review of the PubMed/Ovid Medline and EMBASE electronic databases was performed. The search string was constructed with the aid of an experienced librarian with expertise in electronic searches, and the search was performed on 25 May 2022. Two reviewers independently applied the predefined eligibility and inclusion criteria to the articles. The references of all the fully assessed articles and relevant review papers were also hand-searched to identify additional articles.

2.2. Exclusion and Inclusion Criteria and Study Selection

Randomized controlled trials and prospective cohort studies (level I and II studies) were included; retrospective comparative trials (level III studies) and therapeutic case series (level IV studies) were also considered for inclusion. Reviews, meta-analyses, expert opinions, and editorial pieces were excluded. Animal studies, in vitro studies, and biomechanical studies on human cadaver specimens were also excluded.
Studies enrolling human subjects of all ages receiving treatments of orthopedic pathologies with the application of adipose tissue derivates (unprocessed adipose tissue, non-expanded or expanded ADSCs, SVF, or microfragmented adipose tissue) were eligible for inclusion. The treatment of rheumatoid conditions, diseases of the hand, and applications in plastic surgery, as well as wound healing enhancement protocols were excluded. Studies presenting results of different therapeutical approaches were excluded unless it was possible to identify and isolate data from the subgroup of patients who received the application of adipose tissue derivates.

2.3. Quality Assessment

All the studies that met the final inclusion criteria were individually assessed for quality by two independent reviewers, first by assigning a level of evidence according to the recommendations by Marx et al. [21]. Second, the presence of national or international registration as a clinical trial was documented. Furthermore, methodologic quality was assessed to determine the scientific validity and robustness of research studies by evaluating the study design, methodology, data collection, analysis, and interpretation. For this purpose, the quality of all studies was evaluated using the modified Coleman Methodology Score (mCMS). This instrument is utilized to evaluate methodological quality in studies reporting surgical outcomes after patellar tendinopathy. Subsequent, modified versions of the CMS have been proposed to improve the analysis of the quality of studies on different anatomical districts or procedures [22,23,24,25,26].
Methodological quality was evaluated for each paper and graded as excellent (>85 points), good (70–84 points), fair (55–69 points), and poor (<55 points). Grouping, depending on adipose tissue preparation and its processing, was subsequently performed. Finally, the Cochrane risk-of-bias tool for randomized trials (Version 2) was used to assess the risk of bias of Level I and II randomized controlled trials [27]. This tool is widely used to assess the risk of bias in individual studies included in systematic reviews.

2.4. Data Extraction, Collected Variables, Grouping, and Analysis

Information regarding authors, journal and year of publication, study design and quality of evidence, country where the study was conducted, anatomical district of application, treated pathology, intervention, and specific characteristics of the adipose tissue derivate used were extracted and entered into a spreadsheet for analysis. In particular, the source of the adipose cells; the setting of the processing (specialized laboratory or point-of-care); the methods of extraction, if expansion and characterization were performed; and if a commercial device was used were considered relevant variables. The included articles were then categorized depending on the country the trial was performed in, the anatomical area of application, the type of adipose tissue preparation, and tissue processing (site, device, extraction, expansion, and characterization).

2.5. Statistical Analysis

Statistical analysis was performed using GraphPad Prism v. 6.0 software (GraphPad Software Inc., Boston, MA, USA) and Microsoft Excel Version 2306 (Microsoft Corporation, Redmond, WA, USA). The Shapiro–Wilk normality test was used to evaluate the normal distribution of the sample. Dichotomous variables were expressed in numbers of cases and frequencies; their differences were tested using the chi-square test or the Fisher’s exact test. Continuous variables were expressed as median and interquartile range (first and third quartiles) or mean ± standard deviation, as appropriate. The between-group differences for continuous variables were evaluated with the unpaired Student t test or Mann–Whitney test, according to the characteristics of the data distribution evaluated using the Shapiro–Wilk normality test. For all analyses, the significance level was set at p < 0.05.

3. Results

3.1. Review Process and Included Studies

The database search identified 5038 studies. After the removal of duplicates and title and abstract screening, 297 articles were selected. The full-text assessment with an additional hand search of references identified 82 studies that were included in this review (Figure 1).
A trend of an increasing number of publications was observed over the last few years, stagnating after 2019 (Figure 2). This could be related to the COVID-19 pandemic.
A total of 3594 patients were treated with adipose tissue derivates in the included studies.

3.2. Worldwide Distribution

Most of the included studies were performed in Asia, with South Korea being the leading country, followed by the United States and Italy; with 42 national studies, these three countries account for over the half of all publications on this topic. Only four multi-national studies (all Europe-based) were published (Figure 3).
Most of the procedures were performed in Europe (European Union (EU) and the United Kingdom (UK)) (2589 patients) followed by Asia (747) and the USA (180). A higher number of patients per study were enrolled in European studies (25.5 [15.50–77.25]) compared to those in Asia (18.00 [12.00–30.00]) and the USA (13.50 [4.75–28.25]), with no statistically significant difference. It should, however, be noted that, among the European-based studies, the largest (1128 patients) is a case series involving different arthritic joints (61.0% knee, 33.7% hip, and 5.3% other joints) of very low methodological quality [28].

3.3. Anatomical Area and Treated Pathologies

In total, 57 out of the 82 included studies were specifically dedicated to procedures around the knee (Figure 4). The number of enrolled patients per study was significantly higher for knees compared to others (knee: 20.00 [15.50–25.25]; other areas: 10.00 [1.00–21.5]; p = 0.0015).
Most of the available studies investigated the treatment of knee disorders, mainly knee osteoarthritis. Nevertheless, no uniform indication for treatment was encountered, with included Kellgren–Lawrence osteoarthritis grades ranging from 1 to 4. Besides knee osteoarthritis, one study investigated the treatment of juvenile osteochondritis dissecans of the patella [29]; one, patellar tendinopathy [30]; and one, ACL tears [31]. With 3183 treated patients, the knee was confirmed to be the most studied joint in terms of the number of recorded procedures. The Western Ontario and McMaster Universities Arthritis Index (WOMAC), the Knee Injury and Osteoarthritis Outcome Score (KOOS), a Visual Analog Scale, and the 36-Item Short Form Survey (SF-36) were the most frequently used outcome measures.
Regarding the hip joint, osteoarthritis was an indication for treatment [32,33], together with acetabular cartilage delamination in femoroacetabular impingement [34] and osteonecrosis of the femoral head [35,36]. Ankle osteoarthritis [37,38] and Achilles tendinopathy [39,40] were treated in two publications each, and a case report presented the results of treatment of an unstable osteochondral lesion of the talus [41]. Around the shoulder joint, regenerative approaches involving adipose tissue derivates were all directed to rotator cuff pathologies, with two studies dealing with full thickness tears [42,43], three with partial-thickness tears [44,45,46], and one with shoulder pain caused by refractory rotator cuff disease in wheelchair users [47]. The treatment of recalcitrant lateral elbow pain was the topic of three included studies involving the elbow [48,49,50]. Two included studies involved the spine and addressed degenerative disease and chronic discogenic low back pain [51,52]. Four studies reported on the application of adipose tissue derivates in multiple joints, mainly including osteoarthritis patients treated for knee and hip symptoms [28,53,54,55].

3.4. Procedures

Almost three-quarters of the included studies focused on injections of adipose tissue derivates into the affected joint or tendon, either alone or combined with PRP or hyaluronic acid (Figure 5).
Arthroscopic procedures with associated injections of adipose tissue derivates were reported in 24% of the cases; half of these were sham procedures (the debridement of arthritic joints, synovectomy, and partial meniscectomy for degenerative lesions), and half included microfractures, ACL reconstructions, and rotator cuff repairs. In two papers, open procedures augmented by the injection of adipose tissue derivates were described (subtalar arthrodesis [38] and high tibial osteotomy [56]).

3.5. Adipose Tissue Derivate Types

The preferred source for tissue harvest was autogenic subcutaneous fat. The abdomen, flanks, buttocks, and thighs were the mentioned sources of subcutaneous fat tissue. In three studies, autologous fat tissue was harvested during the surgical procedure from a local reservoir as follows: the infrapatellar fat pad in two studies (25 and 30 patients, respectively [57,58]) and the peritrochanteric area in one (17 patients [34]). The use of allogenic ADSCs was reported in four studies, for a total of 108 patients [38,48,59,60]. Figure 6 summarizes the distribution of graft sources in the included studies.
Approximately one-quarter (26%) of the included studies dealt with expanded ADSCs, whereas the rest investigated the effect of SVF (including microfragmented adipose tissue); no significant differences in the number of patients per study in these two groups were found (ADSCs: 18.00 [12.00–24.00]; SVF: 20.00 [6.00–35.00]; p > 0.05). One study described the use of unprocessed fat tissue (Figure 7).
Of the 60 studies (72%) dealing with SVF, 30 used enzymatic and 30 non-enzymatic extraction methods. A characterization of the used cells was performed in less than half of the included studies (39%) (Supplementary Figure S1).

3.6. Adipose Tissue Processing

Two-thirds of the included studies presented the application of adipose tissue derivates as a point-of-care procedure without cell expansion. The commercial products used in these 55 studies are reported in Figure 8, whereas relevant information on the site and type of processing is summarized in Supplementary Figure S1.

3.7. Quality of Evidence Appraisal

A level of evidence was assigned to each study according to the recommendations by Marx et al. [21] as follows: 15.9% of the included studies were classified as Level I and 19.5% as Level II trials. More than the half of the included studies were case reports or case series (Figure 9).
National or international clinical trial registration was obtained in 35% of the included studies. The level of evidence of studies using expanded ADSCs was significantly higher than that of studies using SVF (ADSCs: 2.00 [2.00–4.00]; SVF: 4.00 [2.00–4.00]; p < 0.001).
The mean mCMS for all included studies was 51.7 ± 21.4 points, with a significantly higher score for the studies dealing with expanded ADSCs compared to those dealing with SVF (mean mCMS: 63.9 ± 17.8 and 47.7 ± 21.4, respectively, p = 0.0027). A wide variability in the methodological quality of the studies was encountered, with most of the studies ranking in the “poor” or “fair” categories (76.8%). Reasons for low scores were mainly retrospective study designs, a lack of clearly reported diagnostic and inclusion criteria, as well as the low number of included patients and short follow-up. The average study quality in terms of mCMS was higher for Asian and USA-based studies (Asia: 56.2 ± 23.0, p = 0.0366) compared to the EU and the UK (USA: 55.9 ± 25.0, p: n.s.), when compared to the EU and the UK (EU and UK: 47.2 ± 17.6; Australia: 43.8 ± 18.8; Argentina: 39.5 ± 31.8). This trend was maintained when considering the percentage of studies with excellent or good (mCMS ≥ 70) methodological quality (Asia: 37.1%; USA: 30%; EU and UK: 6.7%; Australia: 20%; Argentina: 0%). The results of the Cochrane risk-of-bias analysis for Level I and II randomized studies are reported in Figure 10.
Considering these studies with a high level of evidence, the literature does not adequately investigate many of the aforementioned applications of adipose tissue derivates, such as symptomatic knee osteoarthritis, in particular, with Kellgren–Lawrence grade 2 and 3 (with controversial results in some randomized controlled trials) [58,61,62,63,64,65,66,67], partial- and full-thickness rotator cuff tears [42,45], and Achilles tendinopathy [39]. Promising results have been obtained in Level II trials on the treatment of lateral recalcitrant elbow pain [48] and degenerative disc disease [51,52] as well as in the augmentation of open-wedge high tibial osteotomy [56], yet these should be confirmed in Level I trials. All other previously listed treatments (osteochondritis dissecans of the patella, patellar tendinopathy, ACL tears, hip osteoarthritis, osteonecrosis of the femoral head, and ankle osteoarthritis) do not encounter any sound support in the currently available literature. These treatments should, therefore, be considered as experimental and only administered within adequately designed and monitored research protocols.

4. Discussion

Our systematic analysis of publications dealing with adipose tissue derivates in orthopedics showed high heterogeneity in terms of types of performed procedures as well as the choice and processing of adipose tissue derivates. Although high-quality studies have been produced, a high volume of publications show low methodological quality, especially for studies dealing with SVF derivates.
Only a few clinical applications were investigated with adequate high-level publications of symptomatic knee osteoarthritis, rotator cuff tears, Achilles tendinopathy, and subtalar arthrodesis. Promising results have been obtained for the treatment of lateral recalcitrant elbow pain and degenerative disc disease and in the augmentation of open-wedge high tibial osteotomy, yet these studies are biased by a lower level of evidence. No other application is currently supported by adequate high-quality literature, and care should be taken in the use thereof. Through permissive regulations, South Korea, the United States, and Italy are the countries in which the most studies were conducted.

4.1. Historical Remarks on MSC Sources for Orthopedic Applications

The term MSCs was introduced in 1991 to identify a group of multipotent, adult stem cells, which have the potential to differentiate into various types of mesenchymal tissues. These cells are characterized by a definite behavior in culture, a subset of surface markers, and differentiating abilities [70,71,72]. As opposed to the initial belief, more recent research demonstrates that, rather than through direct contribution to tissue regeneration through differentiation, MSCs move to sites of tissue injury and direct the regenerative response through the paracrine secretion of bioactive, trophic, and immunomodulatory agents [7,8,9,10,11,12,13,14]. These cells then contribute to create a suitable microenvironment for tissue repair, rather than repairing the tissue themselves; this suggests that an alternative acronym, “Medicinal Signaling Cells”, be considered more suitable than the actual activity of these cells [73].
Bone marrow has historically provided the classical source of MSCs used for orthopedic applications. This site is easy to access and provides a high number of MSCs, without related donor-site morbidity. During the last decade, several distinct populations of MSCs have been isolated from specific periarticular tissues (such as synovium, ligaments, tendons, and bursa), suggesting the possibility of perioperative extraction to avoid the violation of the iliac crest. Nevertheless, the clinical application of these cells remains limited due to the relatively small amount of tissue available for harvest and the need for expansion in a dedicated facility which might affect the functional phenotype of the cellular product [74,75,76,77,78,79,80,81]. Similar to periarticular tissues, adipose tissue also hosts local, resident MSCs. Furthermore, adipose tissue is frequently found in abundant quantities in easily surgically accessible regions of the human body. This triggered the development of techniques to obtain autologous adipose tissue samples with minimal patient morbidity for either processing in a dedicated laboratory or with point-of-care devices.

4.2. Adipose Tissue and Its Derivates for Regenerative Medicine Applications

ADSCs represent an appealing perspective for orthopedic surgeons, demonstrating the possibility of the enhancement of tissue regeneration and healing in preclinical studies [1,82]. Adipose tissue can be harvested in abundance from the subcutaneous tissue via lipoaspirate or tissue biopsy [18]. There are also a large variety of possibilities to process adipose tissue to a specific therapeutic product, which can be distinguished between (1) expanded MSC products and (2) non-expanded adipose tissue derivates, including enzymatically and mechanically obtained SVF.
ADSCs can be extracted from adipose tissue via enzymatic digestion and are plastic adherent when maintained in cell culture. Expanded ADSCs products are often characterized in clinical studies with typical surface markers, CD105, CD73, and CD90, as well as the absence (<2%) of CD45, CD34, CD14, CD19, and HLA-DR, and provide a high and pure concentration of therapeutic MSCs. Higher regulative demands and costs result in a lower number of worldwide studies being performed, but these studies are of a higher quality (Figure 7). Nevertheless, there is high heterogeneity in study protocols with different harvesting and cell culture methods, doses, repetitions, carriers, application methods, and co-treatments in musculoskeletal regenerative therapy [83]. Moreover, a limited or poor comprehension of the mesenchymal therapeutic source material may actually constitute the most salient rate-limiting step in achieving positive clinical outcomes. While adipose cells do indeed constitute an often-chosen resource, it is without question that other cell populations engender far greater plasticity—and thereby far greater therapeutic potential. Numerous basic science experiments point to sources other than adipose tissue as possessing both a marked ability to dedifferentiate as well as transdifferentiate. Moreover, the expansion of source cells, particularly those amplified in a monolayer, often delimits such cells from providing therapeutic utility. It may also well be the currently incomplete comprehension of the operative variables that actually influences the outcomes stem cells provide in the setting of senescent and degenerative morbidities.
Non-expanded adipose tissue derivates can be extracted from unprocessed fat tissue and contain a cocktail of MSCs, platelets, immune cells, cytokines, and growth factors, defined as SVF [19]. Cell isolation can be performed using enzymatic methods (collagenases or proteases) or, alternatively, the SVF can be separated from the connective tissue via mechanical methods such as centrifugation or microfragmentation. This last method leads to the production of microfragmented adipose tissue [19,84], which can be directly used in clinical application after tissue harvest, while the enzymatic SVF production time is dependent upon enzyme digestion. It is important to consider that different harvest and isolation methods can also affect cell viability and the functional phenotype of MSCs [18,85]. These effects are specifically reported after enzymatic digestion.
Alternatively, non-enzymatic processing leads to a lower cell yield, which also depends on the harvesting method [86]. Recently, non-expanded adipose tissue derivates have gained popularity in clinical applications (Figure 7). The advantages of these products are high cost-effectiveness and the possibility of a point-of care application without extensive cell culture. Nevertheless, the fraction of MSCs in these products is low compared to expanded MSC products, and there is a lack of systematic characterization and regulative guidelines [87]. Therefore, it remains questionable if these therapeutical non-expanded adipose tissue approaches can directly be compared with expanded and characterized ADSC products. Figure 11 summarizes the relevant features of the adipose tissue derivatives investigated in our review.

4.3. Limitations

This systematic analysis of publications dealing with human applications of adipose tissue derivates has some limitations. The first is directly related to the type and quality of the included studies, which leads to high heterogeneity. Different procedures for different conditions as well as different methods and measures are reported, which make the analysis of the outcomes more complex. This heterogeneity arises from the recent introduction of these applications in orthopedics, and the current lack of clear recommendations on their use, which leads to the flourishing of low-quality publications. This heterogeneity, especially for the methodologies of highlighted studies through quality appraisal, derives from the complexity of this unexplored research area and highlights the need for an improvement in methodological quality for in vivo studies dealing with adipose tissue derivates. A further limitation is related to the characteristics of the adipose cells used in the different studies. First, a characterization of the cells used was performed in less than half of the included studies (39%); furthermore, patient-related factors such as obesity and hormonal status may affect the characteristics of the harvested cells beyond their immunological phenotype, thus leading to slightly different effects when used in clinical applications. Finally, differences in national regulations and reimbursement systems limit the availability of some of the experimental procedures and the commercially available products in some countries, restricting researchers to a limited portfolio of adipose tissue derivates. The lack of regulations is reflected in the lack of a consensus on the use of such products in the international orthopedic community.

5. Conclusions

This systematic analysis of publications dealing with adipose tissue derivates in orthopedics shows a high heterogeneity in terms of types of performed procedures as well as the choice and processing of adipose tissue derivates. Although high-quality studies have been produced, a high volume of publications show low methodological quality, especially for studies dealing with SVF and microfragmented adipose tissue. Only few clinical applications were investigated with adequate high-level publications, mostly dealing with symptomatic knee osteoarthritis. Promising results with lower levels of evidence have been obtained for the treatment of lateral recalcitrant elbow pain and degenerative disc disease and in the augmentation of open-wedge high tibial osteotomy, yet further research is required to fill the current high-quality literature gap. No other application is currently supported by adequate high-quality literature, which means that extreme care and appropriate monitoring are advised.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm12144719/s1, Supplementary Figure S1: Relevant information on site (A), type of processing (B), expansion (C), and characterization (D) of the mesenchymal stem/stromal cells used in the included studies.

Author Contributions

R.O.: systematic review, original draft preparation; A.M.: systematic review, statistical analysis; F.A.S. and P.S.R.: discussion, manuscript correction; S.S., C.B. and D.C.W.: manuscript correction; D.C.: study design, systematic review, original draft preparation. All authors have read and agreed to the published version of the manuscript.

Funding

The APC was partially funded by the ON Foundation, Switzerland (ON project number 21-306).

Institutional Review Board Statement

Not applicable. The review was registered on the PROSPERO database (Registration number: CRD42022339795).

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The project (ON project number 21-306) was supported by a literature grant from the ON Foundation, Switzerland.

Conflicts of Interest

The authors, their immediate family, and any research foundation with which they are affiliated have not received any financial payments or other benefits from any commercial entity related to the subject of this article. P.S.R. declares consultancies for Arthrex, Depuy, Microport, and Medacta, outside the submitted work.

References

  1. Perucca Orfei, C.; Boffa, A.; Sourugeon, Y.; Laver, L.; Magalon, J.; Sánchez, M.; Tischer, T.; Filardo, G.; de Girolamo, L. Cell-Based Therapies Have Disease-Modifying Effects on Osteoarthritis in Animal Models. A Systematic Review by the ESSKA Orthobiologic Initiative. Part 1: Adipose Tissue-Derived Cell-Based Injectable Therapies. Knee Surg. Sport. Traumatol. Arthrosc. 2022, 31, 641–655. [Google Scholar] [CrossRef]
  2. Prakoso, A.T.; Basri, H.; Adanta, D.; Yani, I.; Ammarullah, M.I.; Akbar, I.; Ghazali, F.A.; Syahrom, A.; Kamarul, T. The Effect of Tortuosity on Permeability of Porous Scaffold. Biomedicines 2023, 11, 427. [Google Scholar] [CrossRef] [PubMed]
  3. Randelli, P.; Randelli, F.; Ragone, V.; Menon, A.; D’Ambrosi, R.; Cucchi, D.; Cabitza, P.; Banfi, G. Regenerative Medicine in Rotator Cuff Injuries. Biomed Res. Int. 2014, 2014, 129515. [Google Scholar] [CrossRef] [PubMed]
  4. Kim, K.-I.; Kim, M.-S.; Kim, J.-H. Intra-Articular Injection of Autologous Adipose-Derived Stem Cells or Stromal Vascular Fractions: Are They Effective for Patients with Knee Osteoarthritis? A Systematic Review with Meta-Analysis of Randomized Controlled Trials. Am. J. Sports Med. 2023, 51, 837–848. [Google Scholar] [CrossRef]
  5. Satija, N.K.; Singh, V.K.; Verma, Y.K.; Gupta, P.; Sharma, S.; Afrin, F.; Sharma, M.; Sharma, P.; Tripathi, R.P.; Gurudutta, G.U. Mesenchymal Stem Cell-Based Therapy: A New Paradigm in Regenerative Medicine. J. Cell. Mol. Med. 2009, 13, 4385–4402. [Google Scholar] [CrossRef] [Green Version]
  6. Dohan Ehrenfest, D.M.; Andia, I.; Zumstein, M.A.; Zhang, C.-Q.; Pinto, N.R.; Bielecki, T. Classification of Platelet Concentrates (Platelet-Rich Plasma-PRP, Platelet-Rich Fibrin-PRF) for Topical and Infiltrative Use in Orthopedic and Sports Medicine: Current Consensus, Clinical Implications and Perspectives. Muscles Ligaments Tendons J. 2014, 4, 3–9. [Google Scholar] [CrossRef] [PubMed]
  7. Angeline, M.E.; Rodeo, S.A. Biologics in the Management of Rotator Cuff Surgery. Clin. Sports Med. 2012, 31, 645–663. [Google Scholar] [CrossRef]
  8. Schmitt, A.; van Griensven, M.; Imhoff, A.B.; Buchmann, S. Application of Stem Cells in Orthopedics. Stem Cells Int. 2012, 2012, 394962. [Google Scholar] [CrossRef] [Green Version]
  9. Gnecchi, M.; Zhang, Z.; Ni, A.; Dzau, V.J. Paracrine Mechanisms in Adult Stem Cell Signaling and Therapy. Circ. Res. 2008, 103, 1204–1219. [Google Scholar] [CrossRef]
  10. Ménard, C.; Tarte, K. Immunoregulatory Properties of Clinical Grade Mesenchymal Stromal Cells: Evidence, Uncertainties, and Clinical Application. Stem Cell Res. Ther. 2013, 4, 64. [Google Scholar] [CrossRef] [Green Version]
  11. Shi, M.; Liu, Z.-W.; Wang, F.-S. Immunomodulatory Properties and Therapeutic Application of Mesenchymal Stem Cells. Clin. Exp. Immunol. 2011, 164, 1–8. [Google Scholar] [CrossRef]
  12. Soleymaninejadian, E.; Pramanik, K.; Samadian, E. Immunomodulatory Properties of Mesenchymal Stem Cells: Cytokines and Factors. Am. J. Reprod. Immunol. 2012, 67, 1–8. [Google Scholar] [CrossRef] [Green Version]
  13. Krasnodembskaya, A.; Song, Y.; Fang, X.; Gupta, N.; Serikov, V.; Lee, J.-W.; Matthay, M.A. Antibacterial Effect of Human Mesenchymal Stem Cells Is Mediated in Part from Secretion of the Antimicrobial Peptide LL-37. Stem Cells 2010, 28, 2229–2238. [Google Scholar] [CrossRef] [Green Version]
  14. Mei, S.H.J.; Haitsma, J.J.; Dos Santos, C.C.; Deng, Y.; Lai, P.F.H.; Slutsky, A.S.; Liles, W.C.; Stewart, D.J. Mesenchymal Stem Cells Reduce Inflammation While Enhancing Bacterial Clearance and Improving Survival in Sepsis. Am. J. Respir. Crit. Care Med. 2010, 182, 1047–1057. [Google Scholar] [CrossRef]
  15. Randelli, P.; Cucchi, D.; Cabitza, F.; Compagnoni, R.; Menon, A. Tendon-Derived Stem Cells for Rotator Cuff Repair. Oper. Tech. Orthop. 2016, 26, 147–154. [Google Scholar] [CrossRef]
  16. Potapov, I.; García-Prat, L.; Ravichandran, S.; Muñoz-Cánoves, P.; Del Sol, A. Computational Modelling of Stem Cell-Niche Interactions Facilitates Discovery of Strategies to Enhance Tissue Regeneration and Counteract Ageing. FEBS J. 2022, 289, 1486–1491. [Google Scholar] [CrossRef] [PubMed]
  17. Nedunchezian, S.; Banerjee, P.; Lee, C.-Y.; Lee, S.-S.; Lin, C.-W.; Wu, C.-W.; Wu, S.-C.; Chang, J.-K.; Wang, C.-K. Generating Adipose Stem Cell-Laden Hyaluronic Acid-Based Scaffolds Using 3D Bioprinting via the Double Crosslinked Strategy for Chondrogenesis. Mater. Sci. Eng. C 2021, 124, 112072. [Google Scholar] [CrossRef] [PubMed]
  18. Torres-Torrillas, M.; Rubio, M.; Damia, E.; Cuervo, B.; del Romero, A.; Peláez, P.; Chicharro, D.; Miguel, L.; Sopena, J.J. Adipose-Derived Mesenchymal Stem Cells: A Promising Tool in the Treatment of Musculoskeletal Diseases. Int. J. Mol. Sci. 2019, 20, 3105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Lana, J.F.S.D.; Lana, A.V.S.D.; da Fonseca, L.F.; Coelho, M.A.; Marques, G.G.; Mosaner, T.; Ribeiro, L.L.; Azzini, G.O.M.; Santos, G.S.; Fonseca, E.; et al. Stromal Vascular Fraction for Knee Osteoarthritis—An Update. J. Stem Cells Regen. Med. 2022, 18, 11–20. [Google Scholar] [CrossRef]
  20. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G. Preferred Reporting Items for Systematic Reviews and Meta-Analyses: The PRISMA Statement. Ann. Intern. Med. 2009, 151, 264–269, W64. [Google Scholar] [CrossRef] [Green Version]
  21. Marx, R.G.; Wilson, S.M.; Swiontkowski, M.F. Updating the Assignment of Levels of Evidence. J. Bone Jt. Surg. 2015, 97, 1–2. [Google Scholar] [CrossRef] [Green Version]
  22. Ramponi, L.; Yasui, Y.; Murawski, C.D.; Ferkel, R.D.; DiGiovanni, C.W.; Kerkhoffs, G.M.M.J.; Calder, J.D.F.; Takao, M.; Vannini, F.; Choi, W.J.; et al. Lesion Size Is a Predictor of Clinical Outcomes After Bone Marrow Stimulation for Osteochondral Lesions of the Talus: A Systematic Review. Am. J. Sports Med. 2017, 45, 1698–1705. [Google Scholar] [CrossRef] [PubMed]
  23. Abdul-Wahab, T.A.; Betancourt, J.P.; Hassan, F.; Al Thani, S.; Choueiri, H.; Jain, N.B.; Malanga, G.A.; Murrell, W.D.; Prasad, A.; Verborgt, O. Initial Treatment of Complete Rotator Cuff Tear and Transition to Surgical Treatment: Systematic Review of the Evidence. Muscle Ligaments Tendons J. 2019, 06, 35. [Google Scholar] [CrossRef]
  24. Patrick, C.M.; Fernandez, I.; Gonzalez, G.A.; Nesti, L.J.; Dunn, J.C. Analysis of the Quality of Prospective Randomized Controlled Trials for Treatment of Boxer’s Fractures. HAND 2021, 18, 294–299. [Google Scholar] [CrossRef]
  25. Jakobsen, R.B.; Engebretsen, L.; Slauterbeck, J.R. An Analysis of the Quality of Cartilage Repair Studies. J. Bone Joint Surg. Am. 2005, 87, 2232–2239. [Google Scholar] [CrossRef] [Green Version]
  26. Coleman, B.D.; Khan, K.M.; Maffulli, N.; Cook, J.L.; Wark, J.D. Studies of Surgical Outcome after Patellar Tendinopathy: Clinical Significance of Methodological Deficiencies and Guidelines for Future Studies. Victorian Institute of Sport Tendon Study Group. Scand. J. Med. Sci. Sports 2000, 10, 2–11. [Google Scholar] [CrossRef] [PubMed]
  27. Sterne, J.A.C.; Savović, J.; Page, M.J.; Elbers, R.G.; Blencowe, N.S.; Boutron, I.; Cates, C.J.; Cheng, H.-Y.; Corbett, M.S.; Eldridge, S.M.; et al. RoB 2: A Revised Tool for Assessing Risk of Bias in Randomised Trials. BMJ 2019, 366, l4898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Michalek, J.; Moster, R.; Lukac, L.; Proefrock, K.; Petrasovic, M.; Rybar, J.; Chaloupka, A.; Darinskas, A.; Michalek, J.; Kristek, J.; et al. Stromal Vascular Fraction Cells of Adipose and Connective Tissue in People with Osteoarthritis: A Case Control Prospective Multi-Centric Non-Randomized Study. Glob. Surg. 2017, 3, 163. [Google Scholar] [CrossRef]
  29. Russo, A.; Coco, V.; Zaffagnini, S. The Effect of Autologous Adipose Derived Mesenchymal Stem Cell Therapy on Juvenile Osteochondritis Dissecans of the Patella: A Case Study. J. Surg. Case Rep. 2020, 2020, rjaa274. [Google Scholar] [CrossRef] [PubMed]
  30. Khoury, M.A.; Chamari, K.; Tabben, M.; Alkhelaifi, K.; Ricardo, T.; Damián, C.; D’hooghe, P. Expanded Adipose Derived Mesenchymal Stromal Cells Are Effective in Treating Chronic Insertional Patellar Tendinopathy: Clinical and MRI Evaluations of a Pilot Study. J. Exp. Orthop. 2021, 8, 49. [Google Scholar] [CrossRef]
  31. Alentorn-Geli, E.; Seijas, R.; Martínez-De la Torre, A.; Cuscó, X.; Steinbacher, G.; Álvarez-Díaz, P.; Barastegui, D.; Navarro, J.; Serra-Renom, J.M.; Nishishinya, B.; et al. Effects of Autologous Adipose-Derived Regenerative Stem Cells Administered at the Time of Anterior Cruciate Ligament Reconstruction on Knee Function and Graft Healing. J. Orthop. Surg. 2019, 27, 2309499019867580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Heidari, N.; Slevin, M.; Zeinolabediny, Y.; Meloni, D.; Olgiati, S.; Wilson, A.; Noorani, A.; Azamfirei, L. Comparison of the Effect of MFAT and MFAT + PRP on Treatment of Hip Osteoarthritis: An Observational, Intention-to-Treat Study at One Year. J. Clin. Med. 2022, 11, 1056. [Google Scholar] [CrossRef] [PubMed]
  33. Pak, J.; Lee, J.H.; Park, K.S.; Lee, S. Efficacy of Autologous Adipose Tissue-Derived Stem Cells with Extracellular Matrix and Hyaluronic Acid on Human Hip Osteoarthritis. Biomed. Res. 2017, 28, 1654–1658. [Google Scholar]
  34. Ivone, A.; Fioruzzi, A.; Jannelli, E.; Castelli, A.; Ghiara, M.; Ferranti Calderoni, E.; Fontana, A. Micro-Fragmented Adipose Tissue Transplantation (MATT) for the Treatment of Acetabular Delamination. A Two Years Follow up Comparison Study with Microfractures. Acta Biomed. 2019, 90, 69–75. [Google Scholar] [CrossRef]
  35. Pak, J. Autologous Adipose Tissue-Derived Stem Cells Induce Persistent Bone-like Tissue in Osteonecrotic Femoral Heads. Pain Physician 2012, 15, 75–85. [Google Scholar] [CrossRef]
  36. Pak, J.; Lee, J.H.; Jeon, J.H.; Lee, S.H. Complete Resolution of Avascular Necrosis of the Human Femoral Head Treated with Adipose Tissue-Derived Stem Cells and Platelet-Rich Plasma. J. Int. Med. Res. 2014, 42, 1353–1362. [Google Scholar] [CrossRef]
  37. Niazi, N.; Islam, A.; Aljawadi, A.; Akbar, Z.; Pillai, A. Autologous Micro Fragmented Adipose Cell Therapy for End-Stage Ankle Osteoarthritis—Case Report and Review of Literature. SN Compr. Clin. Med. 2021, 3, 909–913. [Google Scholar] [CrossRef]
  38. Myerson, C.L.; Myerson, M.S.; Coetzee, J.C.; Stone McGaver, R.; Giveans, M.R. Subtalar Arthrodesis with Use of Adipose-Derived Cellular Bone Matrix Compared with Autologous Bone Graft: A Multicenter, Randomized Controlled Trial. J. Bone Joint Surg. Am. 2019, 101, 1904–1911. [Google Scholar] [CrossRef]
  39. Usuelli, F.G.; Grassi, M.; Maccario, C.; Vigano’, M.; Lanfranchi, L.; Alfieri Montrasio, U.; de Girolamo, L. Intratendinous Adipose-Derived Stromal Vascular Fraction (SVF) Injection Provides a Safe, Efficacious Treatment for Achilles Tendinopathy: Results of a Randomized Controlled Clinical Trial at a 6-Month Follow-Up. Knee Surg. Sport. Traumatol. Arthrosc. 2018, 26, 2000–2010. [Google Scholar] [CrossRef] [PubMed]
  40. Lo Presti, M.; Costa, G.G.; Agrò, G.; Vasco, C.; Zunarelli, P.; Zaffagnini, S. Adipose-Derived Stromal Vascular Fraction Injection in a Competitive High-Level Athlete Affected by Insertional Achilles Tendinopathy. J. Foot Ankle Surg. 2021, 60, 626–629. [Google Scholar] [CrossRef]
  41. Freitag, J.; Wickham, J.; Shah, K.; Tenen, A. Effect of Autologous Adipose-Derived Mesenchymal Stem Cell Therapy in the Treatment of an Osteochondral Lesion of the Ankle. BMJ Case Rep. 2020, 13, 234595. [Google Scholar] [CrossRef] [PubMed]
  42. Randelli, P.S.; Cucchi, D.; Fossati, C.; Boerci, L.; Nocerino, E.; Ambrogi, F.; Menon, A. Arthroscopic Rotator Cuff Repair Augmentation with Autologous Microfragmented Lipoaspirate Tissue Is Safe and Effectively Improves Short-Term Clinical and Functional Results: A Prospective Randomized Controlled Trial With 24-Month Follow-Up. Am. J. Sports Med. 2022, 50, 1344–1357. [Google Scholar] [CrossRef] [PubMed]
  43. Kim, Y.S.; Sung, C.H.; Chung, S.H.; Kwak, S.J.; Koh, Y.G. Does an Injection of Adipose-Derived Mesenchymal Stem Cells Loaded in Fibrin Glue Influence Rotator Cuff Repair Outcomes? A Clinical and Magnetic Resonance Imaging Study. Am. J. Sports Med. 2017, 45, 2010–2018. [Google Scholar] [CrossRef] [PubMed]
  44. Cherian, C.; Malanga, G.A.; Hogaboom, N.; Pollack, M.A.; Dyson-Hudson, T.A. Autologous, Micro-Fragmented Adipose Tissue as a Treatment for Chronic Shoulder Pain in a Wheelchair Using Individual with Spinal Cord Injury: A Case Report. Spinal Cord Ser. Cases 2019, 5, 46. [Google Scholar] [CrossRef]
  45. Hurd, J.L.; Facile, T.R.; Weiss, J.; Hayes, M.M.; Hayes, M.M.; Furia, J.P.; Maffulli, N.; Winnier, G.E.; Alt, C.; Schmitz, C.; et al. Safety and Efficacy of Treating Symptomatic, Partial-Thickness Rotator Cuff Tears with Fresh, Uncultured, Unmodified, Autologous Adipose-Derived Regenerative Cells (UA-ADRCs) Isolated at the Point of Care: A Prospective, Randomized, Controlled First-in-Hu. J. Orthop. Surg. Res. 2020, 15, 122. [Google Scholar] [CrossRef] [Green Version]
  46. Alt, E.; Rothoerl, R.; Hoppert, M.; Frank, H.-G.; Wuerfel, T.; Alt, C.; Schmitz, C. First Immunohistochemical Evidence of Human Tendon Repair Following Stem Cell Injection: A Case Report and Review of Literature. World J. Stem Cells 2021, 13, 944–970. [Google Scholar] [CrossRef] [PubMed]
  47. Hogaboom, N.; Malanga, G.; Cherian, C.; Dyson-Hudson, T. A Pilot Study to Evaluate Micro-Fragmented Adipose Tissue Injection under Ultrasound Guidance for the Treatment of Refractory Rotator Cuff Disease in Wheelchair Users with Spinal Cord Injury. J. Spinal Cord Med. 2021, 44, 886–895. [Google Scholar] [CrossRef] [PubMed]
  48. Lee, S.Y.; Kim, W.; Lim, C.; Chung, S.G. Treatment of Lateral Epicondylosis by Using Allogeneic Adipose-Derived Mesenchymal Stem Cells: A Pilot Study. Stem Cells 2015, 33, 2995–3005. [Google Scholar] [CrossRef]
  49. Freitag, J.; Shah, K.; Wickham, J.; Tenen, A. Effect of Autologous Adipose-Derived Mesenchymal Stem Cell Therapy in Combination with Autologous Platelet-Rich Plasma in the Treatment of Elbow Tendinopathy. BMJ Case Rep. 2020, 13, 234592. [Google Scholar] [CrossRef]
  50. Khoury, M.; Tabben, M.; Rolón, A.U.; Levi, L.; Chamari, K.; D’Hooghe, P. Promising Improvement of Chronic Lateral Elbow Tendinopathy by Using Adipose Derived Mesenchymal Stromal Cells: A Pilot Study. J. Exp. Orthop. 2021, 8, 6. [Google Scholar] [CrossRef]
  51. Comella, K.; Silbert, R.; Parlo, M. Effects of the Intradiscal Implantation of Stromal Vascular Fraction plus Platelet Rich Plasma in Patients with Degenerative Disc Disease. J. Transl. Med. 2017, 15, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Kumar, H.; Ha, D.-H.; Lee, E.-J.; Park, J.H.; Shim, J.H.; Ahn, T.-K.; Kim, K.-T.; Ropper, A.E.; Sohn, S.; Kim, C.-H.; et al. Safety and Tolerability of Intradiscal Implantation of Combined Autologous Adipose-Derived Mesenchymal Stem Cells and Hyaluronic Acid in Patients with Chronic Discogenic Low Back Pain: 1-Year Follow-up of a Phase I Study. Stem Cell Res. Ther. 2017, 8, 262. [Google Scholar] [CrossRef] [Green Version]
  53. Bright, B.; Bright, R.; Bright, P.; Limaye, A. Ankylosing Spondylitis, Chronic Fatigue and Depression Improved after Stromal Vascular Fraction Treatment for Osteoarthritis: A Case Report. J. Med. Case Rep. 2018, 12, 238. [Google Scholar] [CrossRef]
  54. Pak, J. Regeneration of Human Bones in Hip Osteonecrosis and Human Cartilage in Knee Osteoarthritis with Autologous Adipose-Tissue-Derived Stem Cells: A Case Series. J. Med. Case Rep. 2011, 5, 296. [Google Scholar] [CrossRef] [Green Version]
  55. Michalek, J.; Vrablikova, A.; Darinskas, A.; Lukac, L.; Prucha, J.; Skopalik, J.; Travnik, J.; Cibulka, M.; Dudasova, Z. Stromal Vascular Fraction Cell Therapy for Osteoarthritis in Elderly: Multicenter Case-Control Study. J. Clin. Orthop. Trauma 2019, 10, 76–80. [Google Scholar] [CrossRef] [PubMed]
  56. Koh, Y.-G.; Kwon, O.-R.; Kim, Y.-S.; Choi, Y.-J. Comparative Outcomes of Open-Wedge High Tibial Osteotomy with Platelet-Rich Plasma Alone or in Combination with Mesenchymal Stem Cell Treatment: A Prospective Study. Arthroscopy 2014, 30, 1453–1460. [Google Scholar] [CrossRef] [PubMed]
  57. Koh, Y.-G.; Choi, Y.-J. Infrapatellar Fat Pad-Derived Mesenchymal Stem Cell Therapy for Knee Osteoarthritis. Knee 2012, 19, 902–907. [Google Scholar] [CrossRef]
  58. Zhou, Y.; Li, H.; Xiang, D.; Shao, J.; Fu, Q.; Han, Y.; Zhu, J.; Chen, Y.; Qian, Q. The Clinical Efficacy of Arthroscopic Therapy with Knee Infrapatellar Fat Pad Cell Concentrates in Treating Knee Cartilage Lesion: A Prospective, Randomized, and Controlled Study. J. Orthop. Surg. Res. 2021, 16, 87. [Google Scholar] [CrossRef]
  59. Zhao, X.; Ruan, J.; Tang, H.; Li, J.; Shi, Y.; Li, M.; Li, S.; Xu, C.; Lu, Q.; Dai, C. Multi-Compositional MRI Evaluation of Repair Cartilage in Knee Osteoarthritis with Treatment of Allogeneic Human Adipose-Derived Mesenchymal Progenitor Cells. Stem Cell Res. Ther. 2019, 10, 308. [Google Scholar] [CrossRef] [Green Version]
  60. Lu, L.; Dai, C.; Du, H.; Li, S.; Ye, P.; Zhang, L.; Wang, X.; Song, Y.; Togashi, R.; Vangsness, C.T.; et al. Intra-Articular Injections of Allogeneic Human Adipose-Derived Mesenchymal Progenitor Cells in Patients with Symptomatic Bilateral Knee Osteoarthritis: A Phase I Pilot Study. Regen. Med. 2020, 15, 1625–1636. [Google Scholar] [CrossRef]
  61. Hong, Z.; Chen, J.; Zhang, S.; Zhao, C.; Bi, M.; Chen, X.; Bi, Q. Intra-Articular Injection of Autologous Adipose-Derived Stromal Vascular Fractions for Knee Osteoarthritis: A Double-Blind Randomized Self-Controlled Trial. Int. Orthop. 2019, 43, 1123–1134. [Google Scholar] [CrossRef] [PubMed]
  62. Peretti, G.M.; Ulivi, M.; De Girolamo, L.; Meroni, V.; Lombardo, M.D.; Mangiavini, L. Evaluation of the Use of Autologous Micro-Fragmented Adipose Tissue in the Treatment of Knee Osteoarthritis: Preliminary Results of a Randomized Controlled Trial. J. Biol. Regul. Homeost. Agents 2018, 32, 193–199. [Google Scholar]
  63. Freitag, J.; Bates, D.; Wickham, J.; Shah, K.; Huguenin, L.; Tenen, A.; Paterson, K.; Boyd, R. Adipose-Derived Mesenchymal Stem Cell Therapy in the Treatment of Knee Osteoarthritis: A Randomized Controlled Trial. Regen. Med. 2019, 14, 213–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Lee, W.-S.; Kim, H.J.; Kim, K.-I.; Kim, G.B.; Jin, W. 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. [Google Scholar] [CrossRef] [Green Version]
  65. Lu, L.; Dai, C.; Zhang, Z.; Du, H.; Li, S.; Ye, P.; Fu, Q.; Zhang, L.; Wu, X.; Dong, Y.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Qiao, Z.; Tang, J.; Yue, B.; Wang, J.; Zhang, J.; Xuan, L.; Dai, C.; Li, S.; Li, M.; Xu, C.; et al. Human Adipose-Derived Mesenchymal Progenitor Cells plus Microfracture and Hyaluronic Acid for Cartilage Repair: A Phase IIa Trial. Regen. Med. 2020, 15, 1193–1214. [Google Scholar] [CrossRef] [Green Version]
  67. Garza, J.R.; Campbell, R.E.; Tjoumakaris, F.P.; Freedman, K.B.; Miller, L.S.; Santa Maria, D.; Tucker, B.S. 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. [Google Scholar] [CrossRef]
  68. Song, Y.; Du, H.; Dai, C.; Zhang, L.; Li, S.; Hunter, D.J.; Lu, L.; Bao, C. 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. [Google Scholar] [CrossRef] [PubMed]
  69. Koh, Y.-G.; Kwon, O.-R.; Kim, Y.-S.; Choi, Y.-J.; Tak, D.-H. Adipose-Derived Mesenchymal Stem Cells with Microfracture Versus Microfracture Alone: 2-Year Follow-up of a Prospective Randomized Trial. Arthroscopy 2016, 32, 97–109. [Google Scholar] [CrossRef]
  70. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal Criteria for Defining Multipotent Mesenchymal Stromal Cells. The International Society for Cellular Therapy Position Statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  71. Krampera, M.; Galipeau, J.; Shi, Y.; Tarte, K.; Sensebe, L. Immunological Characterization of Multipotent Mesenchymal Stromal Cells-The International Society for Cellular Therapy (ISCT) Working Proposal. Cytotherapy 2013, 15, 1054–1061. [Google Scholar] [CrossRef] [Green Version]
  72. Horwitz, E.M.; Le Blanc, K.; Dominici, M.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.C.; Deans, R.J.; Krause, D.S.; Keating, A. Clarification of the Nomenclature for MSC: The International Society for Cellular Therapy Position Statement. Cytotherapy 2005, 7, 393–395. [Google Scholar] [CrossRef] [PubMed]
  73. Caplan, A.I. What’s in a Name? Tissue Eng. Part A 2010, 16, 2415–2417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Salmikangas, P.; Schuessler-Lenz, M.; Ruiz, S.; Celis, P.; Reischl, I.; Menezes-Ferreira, M.; Flory, E.; Renner, M.; Ferry, N. Marketing Regulatory Oversight of Advanced Therapy Medicinal Products (ATMPs) in Europe: The EMA/CAT Perspective. In Regulatory Aspects of Gene Therapy and Cell Therapy Products; Springer: Berlin/Heidelberg, Germany, 2015; pp. 103–130. [Google Scholar]
  75. Guess, A.J.; Daneault, B.; Wang, R.; Bradbury, H.; La Perle, K.M.D.; Fitch, J.; Hedrick, S.L.; Hamelberg, E.; Astbury, C.; White, P.; et al. Safety Profile of Good Manufacturing Practice Manufactured Interferon γ-Primed Mesenchymal Stem/Stromal Cells for Clinical Trials. Stem Cells Transl. Med. 2017, 6, 1868–1879. [Google Scholar] [CrossRef] [PubMed]
  76. Shin, D., II; Kim, M.; Park, D.Y.; Min, B.-H.; Yun, H.-W.; Chung, J.Y.; Min, K.J. Motorized Shaver Harvest Results in Similar Cell Yield and Characteristics Compared with Rongeur Biopsy During Arthroscopic Synovium-Derived Mesenchymal Stem Cell Harvest. Arthroscopy 2021, 37, 2873–2882. [Google Scholar] [CrossRef]
  77. Fu, W.; Li, Q.; Tang, X.; Chen, G.; Zhang, C.; Li, J. Mesenchymal Stem Cells Reside in Anterior Cruciate Ligament Remnants in Situ. Int. Orthop. 2016, 40, 1523–1530. [Google Scholar] [CrossRef]
  78. Randelli, P.; Conforti, E.; Piccoli, M.; Ragone, V.; Creo, P.; Cirillo, F.; Masuzzo, P.; Tringali, C.; Cabitza, P.; Tettamanti, G.; et al. Isolation and Characterization of 2 New Human Rotator Cuff and Long Head of Biceps Tendon Cells Possessing Stem Cell-Like Self-Renewal and Multipotential Differentiation Capacity. Am. J. Sports Med. 2013, 41, 1653–1664. [Google Scholar] [CrossRef]
  79. Tsai, C.C.; Huang, T.F.; Ma, H.L.; Chiang, E.R.; Hung, S.C. Isolation of Mesenchymal Stem Cells from Shoulder Rotator Cuff: A Potential Source for Muscle and Tendon Repair. Cell Transpl. 2013, 22, 413–422. [Google Scholar] [CrossRef]
  80. Utsunomiya, H.; Uchida, S.; Sekiya, I.; Sakai, A.; Moridera, K.; Nakamura, T. Isolation and Characterization of Human Mesenchymal Stem Cells Derived from Shoulder Tissues Involved in Rotator Cuff Tears. Am. J. Sports Med. 2013, 41, 657–668. [Google Scholar] [CrossRef] [PubMed]
  81. Moravcikova, E.; Meyer, E.M.; Corselli, M.; Donnenberg, V.S.; Donnenberg, A.D. Proteomic Profiling of Native Unpassaged and Culture-Expanded Mesenchymal Stromal Cells (MSC). Cytometry A 2018, 93, 894–904. [Google Scholar] [CrossRef] [Green Version]
  82. Tevlin, R.; DesJardins-Park, H.; Huber, J.; DiIorio, S.E.; Longaker, M.T.; Wan, D.C. Musculoskeletal Tissue Engineering: Adipose Derived Stromal Cell Implementation for the Treatment of Osteoarthritis. Biomaterials 2022, 286, 121544. [Google Scholar] [CrossRef]
  83. Ossendorff, R.; Walter, S.; Schildberg, F.; Khoury, M.; Salzmann, G. Controversies in Regenerative Medicine: Should Knee Joint Osteoarthritis Be Treated with Mesenchymal Stromal Cells? Eur. Cells Mater. 2022, 43, 89–111. [Google Scholar] [CrossRef]
  84. Aronowitz, J.A.; Lockhart, R.A.; Hakakian, C.S. Mechanical versus Enzymatic Isolation of Stromal Vascular Fraction Cells from Adipose Tissue. Springerplus 2015, 4, 713. [Google Scholar] [CrossRef] [Green Version]
  85. Walter, S.; Randau, T.; Hilgers, C.; Haddouti, E.-M.; Masson, W.; Gravius, S.; Burger, C.; Wirtz, D.; Schildberg, F. Molecular and Functional Phenotypes of Human Bone Marrow-Derived Mesenchymal Stromal Cells Depend on Harvesting Techniques. Int. J. Mol. Sci. 2020, 21, 4382. [Google Scholar] [CrossRef] [PubMed]
  86. Greenwood, V.; Clausen, P.; Matuska, A.M. Micro-Fragmented Adipose Tissue Cellular Composition Varies by Processing Device and Analytical Method. Sci. Rep. 2022, 12, 16107. [Google Scholar] [CrossRef]
  87. Bora, P.; Majumdar, A.S. Adipose Tissue-Derived Stromal Vascular Fraction in Regenerative Medicine: A Brief Review on Biology and Translation. Stem Cell Res. Ther. 2017, 8, 145. [Google Scholar] [CrossRef]
  88. Jo, C.H.; Lee, Y.G.; Shin, W.H.; Kim, H.; Chai, J.W.; Jeong, E.C.; Kim, J.E.; Shim, H.; Shin, J.S.; Shin, I.S.; et al. Intra-Articular Injection of Mesenchymal Stem Cells for the Treatment of Osteoarthritis of the Knee: A Proof-of-Concept Clinical Trial. Stem Cells 2014, 32, 1254–1266. [Google Scholar] [CrossRef] [PubMed]
  89. Pers, Y.-M.; Rackwitz, L.; Ferreira, R.; Pullig, O.; Delfour, C.; Barry, F.; Sensebe, L.; Casteilla, L.; Fleury, S.; Bourin, P.; et al. Adipose Mesenchymal Stromal Cell-Based Therapy for Severe Osteoarthritis of the Knee: A Phase I Dose-Escalation Trial. Stem Cells Transl. Med. 2016, 5, 847–856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Nguyen, P.D.; Tran, T.D.-X.; Nguyen, H.T.-N.; Vu, H.T.; Le, P.T.-B.; Phan, N.L.-C.; Vu, N.B.; Phan, N.K.; Van Pham, P. Comparative Clinical Observation of Arthroscopic Microfracture in the Presence and Absence of a Stromal Vascular Fraction Injection for Osteoarthritis. Stem Cells Transl. Med. 2017, 6, 187–195. [Google Scholar] [CrossRef]
  91. Tran, T.D.X.; Wu, C.-M.; Dubey, N.K.; Deng, Y.-H.; Su, C.-W.; Pham, T.T.; Thi Le, P.B.; Sestili, P.; Deng, W.-P. Time- and Kellgren–Lawrence Grade-Dependent Changes in Intra-Articularly Transplanted Stromal Vascular Fraction in Osteoarthritic Patients. Cells 2019, 8, 308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Tsubosaka, M.; Matsumoto, T.; Sobajima, S.; Matsushita, T.; Iwaguro, H.; Kuroda, R. Comparison of Clinical and Imaging Outcomes of Different Doses of Adipose-Derived Stromal Vascular Fraction Cell Treatment for Knee Osteoarthritis. Cell Transplant. 2021, 30, 9636897211067454. [Google Scholar] [CrossRef] [PubMed]
  93. Labarre, K.W.; Zimmermann, G. Infiltration of the Hoffa’s Fat Pad with Stromal Vascular Fraction in Patients with Osteoarthritis of the Knee -Results after One Year of Follow-Up. Bone Rep. 2022, 16, 101168. [Google Scholar] [CrossRef] [PubMed]
  94. Screpis, D.; Natali, S.; Farinelli, L.; Piovan, G.; Iacono, V.; de Girolamo, L.; Viganò, M.; Zorzi, C. Autologous Microfragmented Adipose Tissue for the Treatment of Knee Osteoarthritis: Real-World Data at Two Years Follow-Up. J. Clin. Med. 2022, 11, 1268. [Google Scholar] [CrossRef] [PubMed]
  95. Hudetz, D.; Borić, I.; Rod, E.; Jeleč, Ž.; Kunovac, B.; Polašek, O.; Vrdoljak, T.; Plečko, M.; Skelin, A.; Polančec, D.; et al. Early Results of Intra-Articular Micro-Fragmented Lipoaspirate Treatment in Patients with Late Stages Knee Osteoarthritis: A Prospective Study. Croat. Med. J. 2019, 60, 227–236. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A flow chart showing the selection of publications for the systematic review.
Figure 1. A flow chart showing the selection of publications for the systematic review.
Jcm 12 04719 g001
Figure 2. Publication trend over the last 12 years, showing a constant increase in the number of publications dealing with adipose-derived stem/stromal cells (ADSCs) or stromal vascular fraction (SVF) (including microfragmented adipose tissue) applications. For 2022, studies were included up to the review date (March 2022).
Figure 2. Publication trend over the last 12 years, showing a constant increase in the number of publications dealing with adipose-derived stem/stromal cells (ADSCs) or stromal vascular fraction (SVF) (including microfragmented adipose tissue) applications. For 2022, studies were included up to the review date (March 2022).
Jcm 12 04719 g002
Figure 3. Geographical distribution of the included studies. EU, European Union; UK, United Kingdom; USA, United States of America.
Figure 3. Geographical distribution of the included studies. EU, European Union; UK, United Kingdom; USA, United States of America.
Jcm 12 04719 g003
Figure 4. Anatomical areas of procedures performed in the included studies.
Figure 4. Anatomical areas of procedures performed in the included studies.
Jcm 12 04719 g004
Figure 5. Procedures performed in the included studies.
Figure 5. Procedures performed in the included studies.
Jcm 12 04719 g005
Figure 6. Source of adipose tissue considered in the included studies.
Figure 6. Source of adipose tissue considered in the included studies.
Jcm 12 04719 g006
Figure 7. Adipose tissue derivate types used in the included studies.
Figure 7. Adipose tissue derivate types used in the included studies.
Jcm 12 04719 g007
Figure 8. Commercial products used in the included studies.
Figure 8. Commercial products used in the included studies.
Jcm 12 04719 g008
Figure 9. Level of evidence of the included studies, according to Marx et al. [21].
Figure 9. Level of evidence of the included studies, according to Marx et al. [21].
Jcm 12 04719 g009
Figure 10. Cochrane risk-of-bias analysis for the included Level I and II randomized studies. R, bias arising from the randomization process; D, bias due to deviations from intended interventions; Mi, bias due to missing outcome data; Me, bias in measurement of the outcome; S, bias in selection of the reported result; O, overall risk of bias; green: low risk of bias; yellow: moderate risk of bias; red: high risk of bias [38,39,42,45,56,58,59,60,61,62,63,64,65,66,67,68,69].
Figure 10. Cochrane risk-of-bias analysis for the included Level I and II randomized studies. R, bias arising from the randomization process; D, bias due to deviations from intended interventions; Mi, bias due to missing outcome data; Me, bias in measurement of the outcome; S, bias in selection of the reported result; O, overall risk of bias; green: low risk of bias; yellow: moderate risk of bias; red: high risk of bias [38,39,42,45,56,58,59,60,61,62,63,64,65,66,67,68,69].
Jcm 12 04719 g010
Figure 11. Summary of the relevant features of the adipose tissue derivatives investigated in this systematic review. a: [63,64,65,66]; b: [38]; c: [45,61,67]; d: [39,42,58,62]; e: no Level I trials; f: [52,56,68,88,89]; g: [48,59,60]; h: [51,69,90,91,92]; i: [47,93,94,95]; j: no Level II trials; k: ARG, AUS, CHN, FRA, JPN, KOR, QAT, USA; l: CHN, KOR, USA; m: AUS, CHN, CZE, ESP, GER, JPN, KOR, VTN, USA; n: ARG, AUS, BEL, CHN, DNK, GER, GBR, HVR, ITA, USA; o: USA.
Figure 11. Summary of the relevant features of the adipose tissue derivatives investigated in this systematic review. a: [63,64,65,66]; b: [38]; c: [45,61,67]; d: [39,42,58,62]; e: no Level I trials; f: [52,56,68,88,89]; g: [48,59,60]; h: [51,69,90,91,92]; i: [47,93,94,95]; j: no Level II trials; k: ARG, AUS, CHN, FRA, JPN, KOR, QAT, USA; l: CHN, KOR, USA; m: AUS, CHN, CZE, ESP, GER, JPN, KOR, VTN, USA; n: ARG, AUS, BEL, CHN, DNK, GER, GBR, HVR, ITA, USA; o: USA.
Jcm 12 04719 g011
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ossendorff, R.; Menon, A.; Schildberg, F.A.; Randelli, P.S.; Scheidt, S.; Burger, C.; Wirtz, D.C.; Cucchi, D. A Worldwide Analysis of Adipose-Derived Stem Cells and Stromal Vascular Fraction in Orthopedics: Current Evidence and Applications. J. Clin. Med. 2023, 12, 4719. https://doi.org/10.3390/jcm12144719

AMA Style

Ossendorff R, Menon A, Schildberg FA, Randelli PS, Scheidt S, Burger C, Wirtz DC, Cucchi D. A Worldwide Analysis of Adipose-Derived Stem Cells and Stromal Vascular Fraction in Orthopedics: Current Evidence and Applications. Journal of Clinical Medicine. 2023; 12(14):4719. https://doi.org/10.3390/jcm12144719

Chicago/Turabian Style

Ossendorff, Robert, Alessandra Menon, Frank A. Schildberg, Pietro S. Randelli, Sebastian Scheidt, Christof Burger, Dieter C. Wirtz, and Davide Cucchi. 2023. "A Worldwide Analysis of Adipose-Derived Stem Cells and Stromal Vascular Fraction in Orthopedics: Current Evidence and Applications" Journal of Clinical Medicine 12, no. 14: 4719. https://doi.org/10.3390/jcm12144719

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop