Prospective Study Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Orthop. May 18, 2024; 15(5): 457-468
Published online May 18, 2024. doi: 10.5312/wjo.v15.i5.457
Efficacy of stromal vascular fraction for knee osteoarthritis: A prospective, single-centre, non-randomized study with 2 years follow-up
Madhan Jeyaraman, Naveen Jeyaraman, Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu 600077, India
Madhan Jeyaraman, Saurabh Kumar Jha, Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh 201310, India
Madhan Jeyaraman, Ashim Gupta, Department of Orthopaedics and Regenerative Medicine, South Texas Orthopaedic Research Institute, Laredo, TX 78045, United States
Tarun Jayakumar, Department of Orthopaedics, KIMS-Sunshine Hospital, Hyderabad, Telangana 500032, India
Swaminathan Ramasubramanian, Department of Orthopaedics, Government Medical College, Omandurar Government Estate, Chennai, Tamil Nadu 600002, India
Rajni Ranjan, Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh 201306, India
Saurabh Kumar Jha, Department of Zoology, Kalindi College, University of Delhi, New Delhi 110008, India
Ashim Gupta, Department of Orthopaedics and Regenerative Medicine, Regenerative Orthopaedics, Noida 201301, Uttar Pradesh, India
Ashim Gupta, Department of Orthopaedics and Regenerative Medicine, Future Biologics, Lawrenceville, GA 30043, United States
Ashim Gupta, Department of Orthopaedics and Regenerative Medicine, BioIntegrate, Lawrenceville, GA 30043, United States
ORCID number: Madhan Jeyaraman (0000-0002-9045-9493); Naveen Jeyaraman (0000-0002-4362-3326); Tarun Jayakumar (0000-0002-4305-1022); Swaminathan Ramasubramanian (0000-0001-8845-8427); Rajni Ranjan (0000-0003-2324-6970); Saurabh Kumar Jha (0000-0002-7437-0755); Ashim Gupta (0000-0003-1224-2755).
Co-corresponding authors: Madhan Jeyaram and Saurabh Kumar Jha.
Author contributions: Jeyaraman M and Jha SK contributed to conceptualization; Jeyaraman M, Jeyaraman N, and Ranjan R contributed to patient screening, acquired clinical data, performed the desired investigation and follow-up; Jeyaraman M, Jayakumar T, and Ramasubramanian S contributed to manuscript writing; Gupta A contributed to manuscript revision; Jeyaraman M, Gupta A, and Jha SK contributed to proofreading; Jha SK contributed to administration. All the authors have read and approved the final manuscript. Jeyaraman M and Jha SK have played important and indispensable roles in the experimental design, data interpretation and manuscript preparation as the co-corresponding authors. The collaboration between Jeyaraman M and Jha SK is crucial for the publication of this manuscript and other manuscripts still in preparation.
Institutional review board statement: The study was conducted following the Declaration of Helsinki and approved by the Ethics Committee of the School of Medical Sciences and Research, Sharda University (Ref no. SU/SMS&R/76-A/2021/113).
Informed consent statement: All study participants, or their legal guardian, provided written consent prior to study enrollment.
Conflict-of-interest statement: All authors of this manuscript having no conflicts of interest to disclose.
Data sharing statement: The data is contained within the manuscript.
CONSORT 2010 statement: The authors have read the CONSORT 2010 statement, and the manuscript was prepared and revised according to the CONSORT 2010 statement.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Madhan Jeyaraman, Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai, Tamil Nadu 600077, India. madhanjeyaraman@gmail.com
Received: January 23, 2024
Revised: March 21, 2024
Accepted: April 18, 2024
Published online: May 18, 2024

Abstract
BACKGROUND

Current osteoarthritis (OA) treatments focus on symptom relief without addressing the underlying disease process. In regenerative medicine, current treatments have limitations. In regenerative medicine, more research is needed for intra-articular stromal vascular fraction (SVF) injections in OA, including dosage optimization, long-term efficacy, safety, comparisons with other treatments, and mechanism exploration.

AIM

To compare the efficacy of intra-articular SVF with corticosteroid (ICS) injections in patients with primary knee OA.

METHODS

The study included 50 patients with Kellgren-Lawrence grades II and III OA. Patients were randomly assigned (1:1) to receive either a single intra-articular SVF injection (group A) or a single intra-articular ICS (triamcinolone) (group B) injection. Patients were followed up at 1, 3, 6, 12, and 24 months. Visual analog score (VAS) and International Knee Documentation Committee (IKDC) scores were administered before the procedure and at all follow-ups. The safety of SVF in terms of adverse and severe adverse events was recorded. Statistical analysis was performed with SPSS Version 26.0, IBM Corp, Chicago, IL, United States.

RESULTS

Both groups had similar demographics and baseline clinical characteristics. Follow-up showed minor patient loss, resulting in 23 and 24 in groups A and B respectively. Group A experienced a notable reduction in pain, with VAS scores decreasing from 7.7 to 2.4 over 24 months, compared to a minor reduction from 7.8 to 6.2 in Group B. This difference in pain reduction in group A was statistically significant from the third month onwards. Additionally, Group A showed significant improvements in knee functionality, with IKDC scores rising from 33.4 to 83.10, whereas Group B saw a modest increase from 36.7 to 45.16. The improvement in Group A was statistically significant from 6 months and maintained through 24 months.

CONCLUSION

Our study demonstrated that intra-articular administration of SVF can lead to reduced pain and improved knee function in patients with primary knee OA. More adequately powered, multi-center, double-blinded, randomised clinical trials with longer follow-ups are needed to further establish safety and justify its clinical use.

Key Words: Knee osteoarthritis, Adipose tissue, Stromal vascular fraction, Regenerative medicine, Orthobiologics

Core Tip: The clinical study presents a comprehensive analysis of the efficacy of stromal vascular fraction (SVF) in managing knee osteoarthritis, highlighting its superiority over corticosteroid injections in reducing pain and improving joint function. The research emphasizes the need for standardizing SVF therapy protocols to enhance its clinical application.



INTRODUCTION

Osteoarthritis (OA) is a highly prevalent degenerative joint disease that affects approximately 43 million people, contributing to the 11th most debilitating disease worldwide[1]. Its incidence is most common in people aged 60 and over, with a higher prevalence in women than men[2,3]. Among the different types of OA, knee OA is the most frequently encountered, with a prevalence of 181.2 cases per 100000 people[4]. Knee OA significantly impairs quality of life and presents substantial clinical and financial challenges due to its associated symptoms of pain, stiffness, and restricted joint mobility/range of motion (ROM)[4,5]. The disease is characterized by the degradation of articular cartilage, the formation of peripheral osteophytes, subchondral cysts, and bone sclerosis[6,7]. While the current therapeutic approaches for OA mainly focus on symptomatic relief through lifestyle modifications, physical therapy, and pharmacological interventions such as nonsteroidal anti-inflammatory drugs (NSAIDs), opioids, corticosteroids, and viscosupplementation, these treatments often provide temporary relief without addressing the underlying root cause of disease progression[8-11].

Given the limited intrinsic capacity for cartilage repair in the knee, there has been growing interest in exploring regenerative medicine as a promising avenue for managing knee OA. In particular, cellular therapy utilizing mesenchymal stem/stromal cells (MSCs) has emerged as a potent option, especially when traditional therapies are ineffective[12,13]. Autologous MSCs can be derived from various sources, including bone marrow and adipose tissue[14].

Adipose-derived mesenchymal stem cells (AD-MSCs), or Adipose-derived stem cells (ASCs), are a subset of mesenchymal stem cells traditionally viewed as biologically inert but later recognized for their dynamic role in adipose tissue, especially after the discovery of adipokines[15-17]. These cells are conveniently harvested from abundant adipose tissue and share regenerative properties akin to other MSCs, making them promising for regenerative medicine and tissue engineering applications[15,18-20]. They can differentiate into various mesodermal tissues like bone, cartilage, muscle, and adipose tissue[14,21,22]. AD-MSCs are categorized based on size: (1) Macro fat, with particles larger than 1 mm; (2) minifat, with particles between 0.5 and 1 mm; (3) micro fat, with particles between 0.2 and 0.5 mm; and (4) nanofat, with particles smaller than 0.2 mm, primarily containing SVF cells and free lipids. The stromal vascular fraction (SVF) is a mixed cellular product from adipose tissue, containing ASCs, endothelial cells, pericytes, fibroblasts, and immune cells. SVF-gel, an extracellular matrix/SVF blend, retains viable SVF cells and adipose tissue structure[23,24]. SVF has demonstrated superior safety and consistent efficacy in improving pain and functional outcomes[13].

The SVF, a heterogeneous cell population derived from adipose tissue, has been identified as a promising candidate for regenerative medicine in OA[12]. Preclinical models have shown promising results regarding the therapeutic potential of SVF, which is rich in AD-MSCs, in terms of cartilage regeneration[25]. Clinical studies have also begun to explore the use of SVF, with some reporting significant improvements in pain, ROM, functional rating, and functional outcome scores in OA patients receiving intra-articular SVF injections[26-29]. These positive outcomes were often accompanied by low donor-site morbidity and minimal adverse events[26]. Comparative studies have further supported the therapeutic potential of SVF, highlighting its superiority over other treatments such as hyaluronic acid in terms of pain relief, functionality, and structural improvement[30,31]. The lack of standardized SVF products and variability in concomitant treatments present challenges in interpreting and comparing different clinical studies[29,32].

This study acknowledges the existing heterogeneity in the literature regarding the effectiveness of SVF in OA management, which can be attributed to variations in SVF formulation protocols employed across different studies[33,34]. To address this variability, our research presents a meticulously standardized SVF formulation protocol and conducts a comprehensive assessment of its therapeutic efficacy. This scholarly endeavor is positioned to lay a robust foundation for forthcoming studies in the field of OA management, with the overarching goal of promoting a uniform and effective application of SVF therapy in clinical practice.

MATERIALS AND METHODS

A prospective, longitudinal, comparative study was conducted at a single tertiary care institute between September 2021 to August 2023 after obtaining institutional ethical committee clearance [SU/SMS&R/76-A/2021/113]. A total of 50 adult patients with primary OA knee were eligible for recruitment in this study. Patients having grade 2 or grade 3 OA knee according to the Kellgren-Lawrence (K-L) scale who showed no symptomatic relief from the conservative line of management (NSAIDs, physiotherapy, weight loss) for 6 months and were fit for the procedure were included in the study. Exclusion criteria consisted of patients with grade 1 or grade 4 OA knee (K-L scale), severe deformity (> 10° in the coronal or sagittal plane), body mass index (BMI) > 35, concomitant meniscal/ligament involvement, inflammatory arthritis, rheumatological or other systemic diseases, malignancy, patients on immunosuppressive medication, previous history of knee surgeries or intra-articular injections of steroids or hyaluronic acid in the past 6 months, previous history of intra articular stem cell procedures post-traumatic arthritis and infections.

Group allocation

After obtaining informed consent, patients were divided into two groups in an alternating manner. Group A (n = 25) comprised patients who were allocated to receive a single injection of SVF intra-articularly, with a dosage of 10 cc containing 5.0 × 107 cells/10cc and 85% viability. On the other hand, Group B (n = 25) consisted of patients who received a single intra-articular corticosteroid injection (40 mg of triamcinolone (Tricort 40 mg, Cadila Pharmaceuticals Ltd., India).

SVF preparation

Under strict aseptic precautions, a stab incision on the lower part of the abdomen on both iliac fossa just above the ASIS using a No.11 blade was made for cannula entry after local infiltration with 1% lidocaine with epinephrine 1:100000. 40 mL of 2% lidocaine plus 1 mL of 1% epinephrine was added to a 1000 mL bag of 0.9% normal saline and was infiltrated with the tumescent anaesthesia on the infraumbilical area of the abdomen. Approximately 200 cc of adipose tissue was aspirated. Aspirated adipose tissue was washed with saline and subjected to centrifugation at 3000 rpm for 10 mins. The oil layer was discarded and adipose mass was subjected to fragmentation by intra-syringing (30 passages) and then subjected to centrifugation at 3000 rpm for 10 mins. The pellet separated was the SVF which contains the heterogeneous population of cells. The harvesting of adipose tissue to prepare SVF and intra-articular SVF implantation was performed with minimal manipulation of cells under the same surgical sitting.

Intra-articular SVF implantation (Group A)

Under strict aseptic precaution, the prepared autologous SVF (10 cc with a cellular dosage of 5.0 × 107 cells with a viability of > 85% (range: 82%–91% SVF cells) was implanted intra-articularly into each knee joint through the medial infrapatellar fossa with the knee flexed to 90 degrees.

Intra-articular corticosteroid injection (Group B)

Under strict aseptic precaution, the corticosteroid (40 mg of triamcinolone) was injected intra-articularly into each knee joint through the medial infrapatellar fossa.

Post-operative protocol

Patients were started on full-weight-bearing mobilization immediately post-surgery. Physiotherapy included quadriceps and hamstring strengthening exercises and patients were permitted to do light activities as tolerated. All patients were made to avoid oral NSAIDs for at least 4 weeks following the procedure.

Outcome measures

All 50 patients were followed up regularly at 1, 3, 6, 12, and 24 months after the procedure. Patients were evaluated using a VAS pain score (0-10, with 10 being the most severe); and an International Knee Documentation Committee (IKDC) score (ranging from 0 to 100, with 100 representing the highest possible level of function and lowest degree of symptoms) both pre-operatively and at all follow-ups in-person via direct examination. Complications in both the groups and overall patient satisfaction at the final follow-up of 2 years were also documented along with magnetic resonance observation of cartilage repair tissue 2.0 knee score (MOCART 2.0) (0-100, with a higher score indicating better cartilage repair).

Statistical analysis

Statistical analysis was performed using a 2-tailed independent samples t-test for continuous data parameters such as VAS and IKDC scores. The Chi-square test or Fisher’s Exact test was used for categorical data parameters. Statistical analysis was performed using SPSS Version 26 (International Business Machines- IBM, Armonk, NY, United States). Assuming a power of 80% and 95% confidence intervals, a P value less than 0.05 was considered significant.

RESULTS

A total of 50 patients were recruited during the study. Demographic and baseline clinical characteristics of the study population are summarized in Table 1. The mean age of the participants in the study was 48.23 12.07 years (Range: 31-70) with almost equal gender distribution. Both groups were comparable in terms of age, gender, BMI, and Kellgren-Lawrence grading with no statistical difference between the groups. Throughout the study, two patients from Group A (SVF) and one patient from Group B (Triamcinolone) were not contactable and lost to follow-up leaving a total of 47 patients (Group A, n = 23 and Group B, n = 24) eligible for final analysis.

Table 1 Demographic characteristics of the study population, n (%).
Parameters
Group A (n = 25)
Group B (n = 25)
P value
Age (yr)0.95
31–40 3 (12)4 (16)
41–50 6 (24)5 (20)
51–60 7 (28)8 (32)
61–70 9 (36)8 (32)
Sex 0.57
Male 12 (48)14 (56)
Female 13 (52)11 (44)
BMI 0.94
18.5-24.99 (36)9 (36)
25.0-29.98 (32)9 (36)
30.0-35.08 (32)7 (28)
K-L grading 0.58
II 11 (44)13 (52)
III 14 (56)12 (48)
Visual analog scale score

Group A exhibited a substantial reduction in visual analog scale (VAS) scores over 24 months, decreasing from 7.7 ± 1.1 pre-procedure to 2.4 ± 0.88 at 24 months (P < 0.001). Conversely, Group B also saw a decrease in VAS scores, but with a less pronounced change, going from 7.8 ± 0.8 pre-procedure to 6.2 ± 1.1 at 24 months. There were no significant differences in VAS scores between the two groups at baseline (P = 0.71). However, starting from the 3-month mark, significant differences emerged. Group A had a VAS score of 6.5 ± 1.8 compared to 7.6 ± 1.6 in Group B (P = 0.03). This difference became more pronounced at 6 months (4.3 ± 1.4 in Group A vs 6.8 ± 1.2 in Group B, P < 0.001) and remained highly significant at 12 and 24 months (P < 0.001) (Table 2 and Figure 1A).

Figure 1
Figure 1 Graphical chart. A: Representation of Visual Analog Score over various time-points; B: Representation of International Knee Documentation Committee scores over various time-points. VAS: Visual Analog Score; IKDC: International Knee Documentation Committee.
Table 2 Visual analog score in various time-points.
Time-point
Group A (n = 23)
Group B (n = 24)
Standard error
95%CI
t
P value
Pre-procedure 7.7 ± 1.17.8 ± 0.80.272-0.44-0.640.370.71
1 month Follow-up 7.0 ± 1.87.2 ± 1.00.41-0.62-1.020.480.561
3 months Follow-up6.5 ± 1.87.6 ± 1.60.480.13-2.062.280.03
6 months Follow-up4.3 ± 1.46.8 ± 1.20.361.75-3.246.78< 0.001
12 months Follow-up2.3 ± 0.206.1 ± 0.800.163.47-4.1323.04< 0.001
24 months Follow-up2.4 ± 0.886.2 ± 1.10.273.25-4.3413.97< 0.001
IKDC score

Group A showed substantial improvement in their IKDC scores, increasing from an initial mean of 33.4 ± 10.20 to 83.10 ± 18.25 at the 24-month follow-up. In contrast, Group B had a less notable increase, starting at 36.7 ± 9.17 and reaching 45.16 ± 12.35 at 24 months. There was no significant difference in baseline IKDC scores (P = 0.23). However, starting at 6 months, Group A exhibited significantly higher IKDC scores compared to Group B (62.6 ± 13.27 vs 50.16 ± 12.35, P = 0.001) (Table 3 and Figure 1B). This difference continued to widen at 12 and 24 months (P < 0.001), emphasizing the sustained superiority of Group A's outcomes. The improvement in Group A was statistically significant from 6 months and maintained through 24 months.

Table 3 International Knee Documentation Committee score in various time-points.
Time-point
Group A (n = 23)
Group B (n = 24)
Standard error
95%CI
t
P value
Pre-procedure 33.4 ± 10.2036.7 ± 9.172.743-2.22-8.821.200.23
1 month Follow-up 58.14 ± 17.2356.6 ± 18.255.02-11.63-8.55-0.3070.76
3 months Follow-up60.3 ± 14.2154.2 ± 13.253.88-13.91-1.71-1.570.12
6 months Follow-up62.6 ± 13.2750.16 ± 12.353.62-19.72 to -5.15-3.4310.001
12 months Follow-up78.3 ± 12.2448.76 ± 12.173.45-36.48 to -22.59-8.56< 0.001
24 months Follow-up83.10 ± 18.2545.16 ± 12.354.40-46.80 to -29.07-8.61< 0.001
Adverse effects

Throughout the study duration, no patients in either group reported complications related to the procedure, including donor site morbidity, local reactions around the knee, hematoma, or paraesthesia, underscoring the safety profile of both treatments.

Radiological analysis

At baseline, both groups demonstrated similar MOCART 2.0 scores with Group A showing a mean score of 52.7 ± 6.2 and Group B having a mean score of 53.3 ± 4.7. The difference between the groups was not statistically significant (t = 0.37, P = 0.71), indicating comparable conditions prior to the commencement of the respective treatments, as detailed in Table 4 and Figure 2. Remarkably, at the 24-month follow-up, the SVF group exhibited a significant improvement in MOCART 2.0 scores, averaging 91.8 ± 5.1. In contrast, the corticosteroid group presented a more modest improvement, with an average score of 61.2 ± 8.4. The difference between the groups at this time-point was statistically significant (t = -15.01, P < 0.001) with a 95% confidence interval ranging from -34.70 to -26.49, as reported in Table 4. This substantial difference indicates a markedly superior efficacy of SVF therapy over corticosteroids in improving KOA over a 24-month period. A representational case from group A and group B are depicted in Figures 3 and 4 respectively.

Figure 2
Figure 2 Magnetic resonance observation of cartilage repair tissue 2.0 knee score at baseline and 24-month follow-up.
Figure 3
Figure 3 A representational case from group A. A: Pre-procedural radiograph of bilateral knees (anteroposterior (AP) view on standing position) showing decreased medial joint line in bilateral knees suggestive of Kellgren-Lawrence grade II knee osteoarthritis (OA); B: Pre-procedural T2W magnetic resonance imaging (MRI) (coronal section) showing hyperintensity with thinned out cartilage along the medial femoral condyle close to the medial meniscus suggestive of OA knee; C: 2-years follow-up radiograph of bilateral knees (AP view on standing position) showing subtle increase along with the maintenance of medial joint line in bilateral knees; D: 2-years follow-up T2W MRI (coronal image) showing increased cartilaginous thickness with relatively maintained cartilaginous signal indicating response to stromal vascular fraction therapy.
Figure 4
Figure 4 A representational case from group B. A: Pre-procedural radiograph of bilateral knees (anteroposterior (AP) view on standing position) showing decreased medial joint line in bilateral knees suggestive of Kellgren Lawrence grade II knee osteoarthritis (OA); B: Pre-procedural T2W magnetic resonance imaging (MRI) (coronal section) showing hyperintensity with thinned out cartilage along the medial femoral condyle suggestive of OA knee; C: 2-years follow-up radiograph of bilateral knees (AP view on standing position) showing no improvement in the thickness of medial joint line in bilateral knees; D: 2-years follow-up T2W MRI (coronal image) showing no cartilaginous thickness indicating response to corticosteroids therapy.
Table 4 Magnetic resonance observation of cartilage repair tissue 2.0 knee score at baseline and 24-month follow-up.
Time-point
Group A (n = 23)
Group B (n = 24)
Standard error
95%CI
t
P value
Baseline52.7 ± 6.253.3 ± 4.71.60-2.62-3.820.370.71
24 months follow-up91.8 ± 5.161.2 ± 8.42.03-34.70 to -26.49-15.01< 0.001
DISCUSSION

This research critically examines the utilization of SVF in the management of knee OA, a chronic and debilitating condition that affects millions worldwide. The evidence generated in this research is part of an expanding body of evidence that underscores both the safety and efficacy of SVF in the management of OA knee. Our study aligns with a growing consensus that AD-MSCs and SVF hold significant promise in the treatment of OA[13,22,35,36]. Previous studies report a range of positive outcomes from adipose-derived cell therapy, similar to our results[13,22,35,36]. These include reductions in pain, improvements in joint function, and enhanced quality of life. By adding our findings to this growing pool of research, we contribute further evidence to the potential of intra-articular administration of SVF in knee OA patients.

Our study utilized lipoaspirates as the source of AD-MSCs and SVF, similar to previous research[12,37,38]. However, the methodology employed for the extraction and application of these cells varied considerably across studies. Some studies used enzymatic digestion for the extraction of AD-MSCs, while others like ours relied on mechanical methods[12,37,39]. The heterogeneity in cell preparation methodologies demonstrably impacted the purity, viability, and efficacy of the resultant cells, consequently influencing treatment outcomes. In response to this variability, initiatives were undertaken to achieve standardization of the cell formulation, thereby enhancing the repeatability and reproducibility of results. This standardization was underpinned by meticulous characterization of the cellular components. Our results demonstrate that adipose-derived therapies, especially the administration of SVF, can be a promising approach to managing knee OA, an outcome that corroborates the evidence from several previous investigations[37-39].

In light of the findings from our study, there is clear evidence pointing towards a substantial improvement in MOCART 2.0 Scores among participants in Group A, who were treated with SVF, when compared to Group B, who received corticosteroid treatment. This observed improvement from baseline to the 24-month follow-up underscores the potential efficacy of SVF as a therapeutic intervention for knee osteoarthritis (Figure 5). Our results resonate with the existing body of literature, particularly a systematic review focusing on SVF therapy for knee osteoarthritis[29,40]. This comprehensive review highlighted noteworthy enhancements in both pain levels, as assessed by the VAS, and functional outcomes, as measured using the WOMAC. Such congruence between our study and the broader scientific consensus further strengthens the case for SVF as a viable treatment option for this debilitating condition.

Figure 5
Figure 5 Graphical abstract demonstrating the summary of the study. SVF: Stromal vascular fraction; VAS: Visual analog score; IKDC: International knee documentation committee; MOCART: Magnetic resonance observation of cartilage repair tissue 2.0 knee score.

Our study has employed Patient Reported Outcome Measures (PROMs) extensively, which aligns with the increasing recognition of their importance in assessing therapeutic interventions[41-44]. PROMs provide insights into the patient's perception of their symptoms and the effects of treatment on their quality of life, enabling a more holistic evaluation of therapy effectiveness.

Our study, along with previous works, reveals inconsistencies in the duration of improvements following adipose-derived therapy[13,41,45]. These discrepancies underscore the necessity for a standardized protocol to effectively evaluate the long-term efficacy of these therapies. A significant observation in our research, aligning with Nguyen et al[46], is the variability in patient responses to AD-MSCs. This highlights the critical role of individual differences in regenerative medicine, emphasizing the need for a personalized approach to treatment. Factors such as age, overall health status, degree of tissue damage, and genetic factors are posited to significantly influence the therapeutic effectiveness of AD-MSCs and SVF, a theory also supported by findings from Koh et al[47].

Our findings are particularly relevant in the context of managing knee OA. Currently, the focus in knee OA treatment is predominantly on symptom control, with limited options for altering disease progression. The potential of adipose-derived therapies, as suggested by our study and others[29,33,38,48], lies in their ability to not only provide symptomatic relief but also possibly modify the disease trajectory. This represents a significant advancement in the current treatment paradigm for knee OA.

Moreover, our research substantiates the robust safety profile of adipose-derived therapies, consistent with previous studies[29,38,48-53]. While we observed minimal adverse events, which were transient and self-limiting, it's imperative to note that these findings are based on short-term studies. The long-term safety of these therapies remains an area for future research. In addition, our study offers fresh insights into the safety profiles of AD-MSCs vs SVF, an area not extensively explored in existing literature[50,53]. Despite the promising results, there remains a call, echoed by several other studies, for more comprehensive and rigorous research to fully ascertain the efficacy and safety of adipose-derived therapies[22,46,48,51,53,54]. This includes addressing potential risks associated with their extraction, processing, and application[22,53,54]. The emergence of novel approaches, such as nanofat grafting discussed by Jeyaraman et al[55], further underscores the potential of adipose-derived therapies, warranting continued exploration and validation.

Our research indicates that adipose-derived therapies show promise for treating knee OA, but further studies are needed to address unanswered questions and challenges. Future research should focus on standardizing treatment protocols, defining patient selection criteria, examining factors that influence individual responses, and ensuring safety to establish their clinical relevance. A balanced perspective is essential as we explore the potential of these therapies in regenerative medicine. Emphasizing rigorous study designs, comprehensive outcome measures, and standardized assessment methods will enhance the comparability of study results and improve clinical decision-making. Additionally, considering patient-specific factors, such as age and comorbidities, is crucial in choosing the most appropriate therapy, like AD-MSCs or SVF[54,56]. Hence, future research should strive to elucidate these factors' role in influencing treatment outcomes, unravel the complexities around dosing, and ascertain the long-term safety and efficacy of these therapies. Through such comprehensive research efforts, we can harness the full potential of these promising therapies, opening new horizons in the management of knee OA, and solidifying the potential of adipose-derived therapies as a key player in regenerative medicine.

Despite the promising findings from our study and others, it is crucial to acknowledge the limitations inherent in the existing body of evidence. The selection of patients who might benefit most from adipose-derived therapies requires clarification. It is plausible that the degree of OA severity could influence treatment outcomes. For example, patients with less advanced OA may have a better potential for tissue regeneration and could, therefore, show greater benefits from adipose-derived therapies than those with more advanced diseases. Studies exploring this possibility could provide essential guidance for clinicians when considering patient eligibility for these therapies. As Schmitz astutely pointed out, there are substantial methodological inconsistencies and flaws in many published studies on this topic[57]. Many investigations, including ours, have been limited by factors such as the lack of a control group, small sample sizes, and short follow-up durations. Such limitations could potentially introduce bias and confound the results, limiting the generalizability of the findings. The preliminary findings are promising but require cautious interpretation, emphasizing the need for rigorous, large-scale, and longer-term randomized controlled trials. These studies are essential for generating high-quality evidence to guide clinical practice. Additionally, further research is necessary to understand the mechanistic pathways underlying the therapeutic effects of AD-MSCs and SVF, which can improve the therapies' effectiveness and safety. The clinical application of adipose-derived therapies faces challenges, including regulatory obstacles, lack of standardized protocols, and high costs, hindering their widespread adoption. To facilitate the translation of research into clinical use, concerted efforts should address these barriers. Collaborative efforts from researchers, clinicians, and policymakers are essential to advance the field of adipose-derived therapies for knee OA. This study represents a step in this direction and encourages further investigations to build upon its findings.

CONCLUSION

Our study demonstrates that SVF therapy provides notable functional and pain improvement for patients with mild to moderate knee OA. In comparison to the short-term benefits offered by intra-articular corticosteroid injections, SVF shows the potential for longer-lasting results. However, to ensure the widespread integration of SVF therapy into routine clinical practice, larger randomized controlled studies involving a larger cohort are necessary to validate the efficacy and safety of SVF therapy and provide more comprehensive evidence for its adoption in the management of knee OA.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Cell and tissue engineering

Country/Territory of origin: India

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Papazafiropoulou A, Greece S-Editor: Liu JH L-Editor: A P-Editor: Zhao YQ

References
1.  Sen R, Hurley JA.   Osteoarthritis. In: StatPearls. Treasure Island (FL): StatPearls Publishing, 2023. Available from: http://www.ncbi.nlm.nih.gov/books/NBK482326/.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Li C, Zheng Z. Males and Females Have Distinct Molecular Events in the Articular Cartilage during Knee Osteoarthritis. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
3.  Tschon M, Contartese D, Pagani S, Borsari V, Fini M. Gender and Sex Are Key Determinants in Osteoarthritis Not Only Confounding Variables. A Systematic Review of Clinical Data. J Clin Med. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 59]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
4.  Gupta A, Maffulli N. Allogenic Umbilical Cord Tissue for Treatment of Knee Osteoarthritis. Sports Med Arthrosc Rev. 2022;30:162-165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
5.  Gupta A, Sharma SP, Potty AG. Combination of Platelet-Rich Plasma and Hyaluronic Acid vs. Platelet-Rich Plasma Alone for Treatment of Knee Osteoarthritis. Biomedicines. 2023;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
6.  Kaspiris A, Hadjimichael AC, Lianou I, Iliopoulos ID, Ntourantonis D, Melissaridou D, Savvidou OD, Papadimitriou E, Chronopoulos E. Subchondral Bone Cyst Development in Osteoarthritis: From Pathophysiology to Bone Microarchitecture Changes and Clinical Implementations. J Clin Med. 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
7.  Man GS, Mologhianu G. Osteoarthritis pathogenesis - a complex process that involves the entire joint. J Med Life. 2014;7:37-41.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Hermann W, Lambova S, Muller-Ladner U. Current Treatment Options for Osteoarthritis. Curr Rheumatol Rev. 2018;14:108-116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 134]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
9.  Cai Z, Cui Y, Wang J, Qi X, He P, Bu P, Xu Y, Li Y. A narrative review of the progress in the treatment of knee osteoarthritis. Ann Transl Med. 2022;10:373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
10.  Malik KN, Camp N, Chan J, Ballard M. Interventional Techniques for the Management of Knee Osteoarthritis: A Literature Review. Cureus. 2023;15:e47133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
11.  Primorac D, Molnar V, Matišić V, Hudetz D, Jeleč Ž, Rod E, Čukelj F, Vidović D, Vrdoljak T, Dobričić B, Antičević D, Smolić M, Miškulin M, Ćaćić D, Borić I. Comprehensive Review of Knee Osteoarthritis Pharmacological Treatment and the Latest Professional Societies' Guidelines. Pharmaceuticals (Basel). 2021;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 35]  [Article Influence: 11.7]  [Reference Citation Analysis (1)]
12.  Jeyaraman M, Muthu S, Ganie PA. Does the Source of Mesenchymal Stem Cell Have an Effect in the Management of Osteoarthritis of the Knee? Meta-Analysis of Randomized Controlled Trials. Cartilage. 2021;13:1532S-1547S.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 34]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
13.  Muthu S, Patil SC, Jeyaraman N, Jeyaraman M, Gangadaran P, Rajendran RL, Oh EJ, Khanna M, Chung HY, Ahn BC. Comparative effectiveness of adipose-derived mesenchymal stromal cells in the management of knee osteoarthritis: A meta-analysis. World J Orthop. 2023;14:23-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 4]  [Cited by in F6Publishing: 12]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
14.  Muthu S, Jeyaraman M, Jain R, Gulati A, Jeyaraman N, Prajwal GS, Mishra PC. Accentuating the sources of mesenchymal stem cells as cellular therapy for osteoarthritis knees-a panoramic review. Stem Cell Investig. 2021;8:13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
15.  Bunnell BA. Adipose Tissue-Derived Mesenchymal Stem Cells. Cells. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 56]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
16.  Xu ZH, Xiong CW, Miao KS, Yu ZT, Zhang JJ, Yu CL, Huang Y, Zhou XD. Adipokines regulate mesenchymal stem cell osteogenic differentiation. World J Stem Cells. 2023;15:502-513.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
17.  Ceccarelli S, Pontecorvi P, Anastasiadou E, Napoli C, Marchese C. Immunomodulatory Effect of Adipose-Derived Stem Cells: The Cutting Edge of Clinical Application. Front Cell Dev Biol. 2020;8:236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 92]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
18.  Si Z, Wang X, Sun C, Kang Y, Xu J, Hui Y. Adipose-derived stem cells: Sources, potency, and implications for regenerative therapies. Biomed Pharmacother. 2019;114:108765.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 176]  [Article Influence: 35.2]  [Reference Citation Analysis (0)]
19.  Yuan X, Li L, Liu H, Luo J, Zhao Y, Pan C, Zhang X, Chen Y, Gou M. Strategies for improving adipose-derived stem cells for tissue regeneration. Burns Trauma. 2022;10:tkac028.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
20.  Minteer D, Marra KG, Rubin JP. Adipose-derived mesenchymal stem cells: biology and potential applications. Adv Biochem Eng Biotechnol. 2013;129:59-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 74]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
21.  Ong WK, Chakraborty S, Sugii S. Adipose Tissue: Understanding the Heterogeneity of Stem Cells for Regenerative Medicine. Biomolecules. 2021;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 41]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
22.  Sharma S, Muthu S, Jeyaraman M, Ranjan R, Jha SK. Translational products of adipose tissue-derived mesenchymal stem cells: Bench to bedside applications. World J Stem Cells. 2021;13:1360-1381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (1)]
23.  Vasilyev VS, Borovikova AA, Vasilyev SA, Khramtsova NI, Plaksin SA, Kamyshinsky RA, Presnyakov MY, Eremin II.   Features and Biological Properties of Different Adipose Tissue Based Products. Milli-, Micro-, Emulsified (Nano-) Fat, SVF, and AD-Multipotent Mesenchymal Stem Cells. In: Kalaaji A, editor. Plastic and Aesthetic Regenerative Surgery and Fat Grafting: Clinical Application and Operative Techniques. Cham: Springer International Publishing, 2022: 91–107.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Yang Z, Jin S, He Y, Zhang X, Han X, Li F. Comparison of Microfat, Nanofat, and Extracellular Matrix/Stromal Vascular Fraction Gel for Skin Rejuvenation: Basic Research and Clinical Applications. Aesthet Surg J. 2021;41:NP1557-NP1570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
25.  Zhuang WZ, Lin YH, Su LJ, Wu MS, Jeng HY, Chang HC, Huang YH, Ling TY. Mesenchymal stem/stromal cell-based therapy: mechanism, systemic safety and biodistribution for precision clinical applications. J Biomed Sci. 2021;28:28.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 89]  [Article Influence: 29.7]  [Reference Citation Analysis (0)]
26.  Ferreira MY, Carvalho Junior JDC, Ferreira LM. Evaluating the quality of studies reporting on clinical applications of stromal vascular fraction: A systematic review and proposed reporting guidelines (CLINIC-STRA-SVF). Regen Ther. 2023;24:332-342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
27.  Tantuway V, Thomas W, Parikh MB, Sharma R, Jeyaraman N, Jeyaraman M. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
28.  Yokota N, Yamakawa M, Shirata T, Kimura T, Kaneshima H. Clinical results following intra-articular injection of adipose-derived stromal vascular fraction cells in patients with osteoarthritis of the knee. Regen Ther. 2017;6:108-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 55]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
29.  Boada-Pladellorens A, Avellanet M, Pages-Bolibar E, Veiga A. Stromal vascular fraction therapy for knee osteoarthritis: a systematic review. Ther Adv Musculoskelet Dis. 2022;14:1759720X221117879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
30.  Mehling B, Hric M, Salatkova A, Vetrak R, Santora D, Ovariova M, Mihalyova R, Manvelyan M. A Retrospective Study of Stromal Vascular Fraction Cell Therapy for Osteoarthritis. J Clin Med Res. 2020;12:747-751.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
31.  Copcu HE, Oztan S. Not Stromal Vascular Fraction (SVF) or Nanofat, but Total Stromal-Cells (TOST): A New Definition. Systemic Review of Mechanical Stromal-Cell Extraction Techniques. Tissue Eng Regen Med. 2021;18:25-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
32.  Vargel İ, Tuncel A, Baysal N, Hartuç-Çevik İ, Korkusuz F. Autologous Adipose-Derived Tissue Stromal Vascular Fraction (AD-tSVF) for Knee Osteoarthritis. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 7]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
33.  Mautner K, Gottschalk M, Boden SD, Akard A, Bae WC, Black L, Boggess B, Chatterjee P, Chung CB, Easley KA, Gibson G, Hackel J, Jensen K, Kippner L, Kurtenbach C, Kurtzberg J, Mason RA, Noonan B, Roy K, Valentine V, Yeago C, Drissi H. Cell-based versus corticosteroid injections for knee pain in osteoarthritis: a randomized phase 3 trial. Nat Med. 2023;29:3120-3126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 4]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
34.  Anil U, Markus DH, Hurley ET, Manjunath AK, Alaia MJ, Campbell KA, Jazrawi LM, Strauss EJ. The efficacy of intra-articular injections in the treatment of knee osteoarthritis: A network meta-analysis of randomized controlled trials. Knee. 2021;32:173-182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 30]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
35.  Shanmugasundaram S, Vaish A, Chavada V, Murrell WD, Vaishya R. Assessment of safety and efficacy of intra-articular injection of stromal vascular fraction for the treatment of knee osteoarthritis-a systematic review. Int Orthop. 2021;45:615-625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 18]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
36.  Yang WT, Ke CY, Yeh KT, Huang SG, Lin ZY, Wu WT, Lee RP. Stromal-vascular fraction and adipose-derived stem cell therapies improve cartilage regeneration in osteoarthritis-induced rats. Sci Rep. 2022;12:2828.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
37.  Pak J, Chang JJ, Lee JH, Lee SH. Safety reporting on implantation of autologous adipose tissue-derived stem cells with platelet-rich plasma into human articular joints. BMC Musculoskelet Disord. 2013;14:337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 113]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
38.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 102]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
39.  Jevotovsky DS, Alfonso AR, Einhorn TA, Chiu ES. Osteoarthritis and stem cell therapy in humans: a systematic review. Osteoarthritis Cartilage. 2018;26:711-729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 101]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
40.  Zhang S, Xu H, He B, Fan M, Xiao M, Zhang J, Chen D, Tong P, Mao Q. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 10]  [Reference Citation Analysis (0)]
41.  Zhang Y, Chen X, Tong Y, Luo J, Bi Q. Development and Prospect of Intra-Articular Injection in the Treatment of Osteoarthritis: A Review. J Pain Res. 2020;13:1941-1955.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
42.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
43.  Koh YG, Choi YJ. Infrapatellar fat pad-derived mesenchymal stem cell therapy for knee osteoarthritis. Knee. 2012;19:902-907.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 285]  [Cited by in F6Publishing: 277]  [Article Influence: 23.1]  [Reference Citation Analysis (0)]
44.  Frazier T, March K, Garza JR, Bunnell BA, Darr KF, Rogers E, Hamel K, Gimble JM. Non-homologous use of adipose-derived cell and tissue therapies: Osteoarthritis as a case study. Bone Rep. 2022;17:101601.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
45.  Pham PV, Bui KHT, Duong TD, Nguyen NT, Nguyen TD, Le VT, Mai VT, Phan NLC, Ngoc DML and NK. Symptomatic knee osteoarthritis treatment using autologous adipose derived stem cells and platelet-rich plasma: a clinical study. Biomed Rese Therap. 2015;1:02-02.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
46.  Nguyen PD, Tran TD, Nguyen HT, Vu HT, Le PT, Phan NL, Vu NB, Phan NK, 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 65]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
47.  Koh YG, Kwon OR, Kim YS, Choi YJ. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 148]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
48.  Koh YG, Choi YJ, Kwon SK, Kim YS, Yeo JE. Clinical results and second-look arthroscopic findings after treatment with adipose-derived stem cells for knee osteoarthritis. Knee Surg Sports Traumatol Arthrosc. 2015;23:1308-1316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 166]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
49.  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 Sports Traumatol Arthrosc. 2023;31:641-655.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
50.  Kim YS, Choi YJ, Suh DS, Heo DB, Kim YI, Ryu JS, Koh YG. Mesenchymal stem cell implantation in osteoarthritic knees: is fibrin glue effective as a scaffold? Am J Sports Med. 2015;43:176-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 111]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
51.  Islam MS, Ebrahimi-Barough S, Al Mahtab M, Shirian S, Aghayan HR, Arjmand B, Allahverdi A, Ranjbar FE, Sadeg AB, Ai J. Encapsulation of rat bone marrow-derived mesenchymal stem cells (rBMMSCs) in collagen type I containing platelet-rich plasma for osteoarthritis treatment in rat model. Prog Biomater. 2022;11:385-396.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
52.  Hudetz D, Borić I, Rod E, Jeleč Ž, Radić A, Vrdoljak T, Skelin A, Lauc G, Trbojević-Akmačić I, Plečko M, Polašek O, Primorac D. The Effect of Intra-articular Injection of Autologous Microfragmented Fat Tissue on Proteoglycan Synthesis in Patients with Knee Osteoarthritis. Genes (Basel). 2017;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 70]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
53.  Delanois RE, Etcheson JI, Sodhi N, Henn RF 3rd, Gwam CU, George NE, Mont MA. Biologic Therapies for the Treatment of Knee Osteoarthritis. J Arthroplasty. 2019;34:801-813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 42]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
54.  Koh YG, Jo SB, Kwon OR, Suh DS, Lee SW, Park SH, Choi YJ. Mesenchymal stem cell injections improve symptoms of knee osteoarthritis. Arthroscopy. 2013;29:748-755.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 264]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
55.  Jeyaraman M, Muthu S, Sharma S, Ganta C, Ranjan R, Jha SK. Nanofat: A therapeutic paradigm in regenerative medicine. World J Stem Cells. 2021;13:1733-1746.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (2)]
56.  Gupta PK, Chullikana A, Rengasamy M, Shetty N, Pandey V, Agarwal V, Wagh SY, Vellotare PK, Damodaran D, Viswanathan P, Thej C, Balasubramanian S, Majumdar AS. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 192]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
57.  Schmitz C, Alt C, Pearce DA, Furia JP, Maffulli N, Alt EU. Methodological Flaws in Meta-Analyses of Clinical Studies on the Management of Knee Osteoarthritis with Stem Cells: A Systematic Review. Cells. 2022;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]