Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Orthop. Oct 18, 2024; 15(10): 908-917
Published online Oct 18, 2024. doi: 10.5312/wjo.v15.i10.908
Evidence-based orthobiologic practice: Current evidence review and future directions
Madhan Jeyaraman, Naveen Jeyaraman, Department of Orthopedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
Madhan Jeyaraman, Sathish Muthu, Department of Research Methods, Orthopedic Research Group, Coimbatore 641045, Tamil Nadu, India
Swaminathan Ramasubramanian, Sangeetha Balaji, Department of Orthopedic, Government Medical College, Omandurar Government Estate, Chennai 600002, Tamil Nadu, India
Sathish Muthu, Department of Orthopedics, Government Medical College and Hospital, Karur 639004, Tamil Nadu, India
Sathish Muthu, Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
ORCID number: Madhan Jeyaraman (0000-0002-9045-9493); Naveen Jeyaraman (0000-0002-4362-3326); Swaminathan Ramasubramanian (0000-0001-8845-8427); Sangeetha Balaji (0000-0002-1566-1333); Sathish Muthu (0000-0002-7143-4354).
Co-first authors: Madhan Jeyaraman and Naveen Jeyaraman.
Author contributions: Jeyaraman M conceptualized the manuscript; Jeyaraman N, Ramasubramanian S and Balaji S performed data analysis and wrote the manuscript; Muthu S performed image analysis. All authors have read and approved the final version of the manuscript.
Conflict-of-interest statement: Authors declare no conflicts of interest in publishing the manuscript.
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, MS, PhD, Assistant Professor, Research Associate, Department of Orthopedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Velappanchavadi, Chennai 600077, Tamil Nadu, India. madhanjeyaraman@gmail.com
Received: July 16, 2024
Revised: August 31, 2024
Accepted: September 11, 2024
Published online: October 18, 2024
Processing time: 87 Days and 3.5 Hours

Abstract

The field of orthopedic and regenerative medicine is rapidly evolving with the increasing utilization of orthobiologic. These biologically derived therapies, including platelet-rich plasma, mesenchymal stem cells, bone marrow aspirate concentrate, stromal vascular fraction (SVF), and autologous chondrocyte implantation, are gaining traction for their potential to enhance the body's natural healing processes. They offer a promising alternative to traditional surgical interventions for musculoskeletal injuries and degenerative conditions. Current evidence suggests significant benefits of orthobiologics in treating conditions like osteoarthritis, tendon injuries, and spinal disorders, yet inconsistencies in treatment protocols and outcomes persist. The global market for orthobiologics is projected to grow substantially, driven by advancements in biologic therapies such as adipose-derived stem cells and SVF, and the demand for minimally invasive treatments. Despite their promise, regulatory and ethical challenges, as well as the need for high-quality, standardized research, remain significant obstacles. Future directions in the field include advancements in delivery systems, personalized medicine approaches, and the exploration of novel sources like induced pluripotent stem cells, aiming for more targeted and effective treatments. Collaborative efforts are crucial to overcoming these challenges and ensuring the safe and effective application of orthobiologics in clinical practice.

Key Words: Orthobiologics; Platelet-rich plasma; Stem cells; Musculoskeletal regeneration; Regenerative medicine; Evidence-based medicine

Core Tip: Orthobiologics, including platelet-rich plasma, mesenchymal stem cells, bone marrow aspirate concentrate, stromal vascular fraction, and autologous chondrocyte implantation, show significant potential in enhancing musculoskeletal healing and reducing the need for invasive surgeries. Despite their growing popularity, inconsistencies in treatment protocols and evidence levels highlight the need for standardized, high-quality research. Future advancements in delivery systems, personalized medicine, and novel cell sources may further optimize their efficacy and safety.



INTRODUCTION

The field of orthopedic and regenerative medicine is witnessing a surge in interest and application of orthobiologics. These biologically derived therapies, such as platelet-rich plasma (PRP), Adipose tissue-derived Mesenchymal Stem Cells (AD-MSCs), and Bone Marrow Aspirate Concentrate (BMAC), are being recognized for their potential to significantly improve the treatment outcomes of musculoskeletal injuries and degenerative conditions. The promise of these therapies lies in their ability to enhance the body's natural healing processes, offering a less invasive alternative to traditional surgical methods.

Orthobiologics is a dynamic and rapidly advancing field dedicated to the application of biologically derived substances and techniques to enhance healing and regeneration in musculoskeletal tissues. This domain encompasses a diverse range of therapies, including cell-based treatments such as MSCs and cultured chondrocytes, blood-derived products like PRP, tissue grafts, growth factors, hormones, and extracellular matrix components such as hyaluronic acid. While the focus of this paper is primarily on specific cell-based therapies and blood-derived products, it is crucial to recognize the broader and continually evolving nature of the orthobiologics landscape.

Orthobiologics involves using biological substances to promote the repair and regeneration of musculoskeletal tissues. These therapies are increasingly employed to address a wide array of conditions, including osteoarthritis, tendon and ligament injuries, cartilage defects, muscle injuries, and bone fractures[1-4]. To provide a comprehensive understanding of the field, it is essential to outline the major categories of orthobiologics currently in use or under investigation:

Autologous peripheral blood-derived orthobiologics

Utilized in the form of PRP, platelet-rich fibrin, platelet lysate, autologous conditioned serum, autologous protein solution, autologous conditioned plasma, hyperacute serum, growth factor concentrates, plasma rich in growth factors, and gold-indued cytokines.

MSCs

These multipotent cells can be harvested from several sources namely: (1) Autologous (bone marrow, adipose tissue, umbilical cord, amniotic fluid, dental pulp, hair follicle, periosteum, menstrual blood, peripheral blood, and synovial fluid); and (2) allogenic.

Bone marrow-derived biologics

Utilized in the form of BMA and BMAC.

Adipose tissue-derived biologics

Utilized in the form of adipose-derived stem cells, stromal vascular fraction (SVF), micro-fat, nano-fat, microvascular fragments, and exosomes.

CULTURED CELLS
Autologous chondrocyte implantation

The patient's chondrocytes are cultured and reimplanted.

Cultured MSCs

Expanded under laboratory conditions to increase cell numbers.

Growth factors

Including bone morphogenetic proteins (BMPs), which are utilized to enhance bone healing.

Tissue grafts

Autologous or allogeneic grafts used for tissue repair and regeneration.

The clinical use of these orthobiologic techniques varies across the globe, influenced by differing regulatory frameworks. In the United States, the Food and Drug Administration has approved certain forms of PRP, Autologous Chondrocyte Implantation (ACI)/Matrix-Induced ACI (MACI), and specific growth factors such as some BMPs for orthopedic use. Stem cell therapies, particularly those involving significant manipulation of cells, remain largely experimental in many jurisdictions and are subject to ongoing clinical trials. In Europe, the regulatory landscape mirrors that of the United States, with approved applications for PRP, ACI/MACI, and specific growth factors. However, some countries, like Japan and South Korea, have more permissive regulations regarding stem cell therapies, enabling earlier clinical application of these treatments. It is important to note that the regulatory status and clinical adoption of these therapies are continuously evolving, with practices varying not only between countries but also among different medical institutions within the same country.

Despite the growing popularity and enthusiasm surrounding these therapies, the scientific validation of their efficacy and safety remains inconsistent and often controversial. Recent studies have shown promising results for the use of orthobiologics. For instance, PRP therapy has demonstrated significant potential in managing early knee osteoarthritis[5] and enhancing the healing process following rotator cuff repairs[6]. Similarly, stem cell therapies are being explored for their ability to regenerate cartilage in osteoarthritic joints[7] and improve recovery from tendon injuries[8]. However, the field faces challenges such as variability in treatment protocols, inconsistent outcomes, and regulatory concerns. The clinical application of orthobiologics also extends to chronic wound healing, where therapies like PRP have shown effectiveness in promoting tissue regeneration and reducing healing time[9]. Moreover, advancements in molecular biology and drug delivery systems are paving the way for more targeted and controlled release of bioactive molecules, enhancing the therapeutic potential of orthobiologics. This editorial aim to provide a concise overview of the current evidence levels in the field of orthobiologics.

CURRENT UTILIZATION OF ORTHOBIOLOGICS
Clinical applications

Orthobiologics are increasingly being utilized in various clinical scenarios to promote the healing and regeneration of musculoskeletal tissues. They are particularly prominent in the treatment of sports injuries, osteoarthritis, and spinal disorders. PRP, in particular, has demonstrated effectiveness in treating tendinopathies like tennis elbow and patellar tendinitis. It achieves this by delivering growth factors that promote healing and reduce inflammation. Similarly, stem cell therapies, especially those involving MSCs, are being explored for their potential to repair and regenerate damaged ligaments and tendons. These therapies show promise in accelerating recovery and reducing the necessity for surgical interventions[9,10]. In osteoarthritis management, orthobiologics offer a promising alternative to conventional treatments. PRP injections aim to reduce pain and inflammation, potentially delaying the need for joint replacement, while MSCs are being investigated for their cartilage regeneration potential[3,11-13]. Orthobiologics are gaining traction in the treatment of spinal disorders. BMP, for instance, are utilized in spinal fusion procedures to enhance bone growth and increase the success rate of the surgery. PRP and stem cell therapies are also being investigated for their potential to repair intervertebral discs and alleviate chronic back pain associated with degenerative disc disease[14].

Popularity and market growth

The popularity and market growth of orthobiologics are driven by several factors, including the increasing prevalence of musculoskeletal conditions, advancements in biological therapies, and a growing preference for minimally invasive treatments. The global orthobiologics market is projected to surpass USD 11.4 billion by 2032. This expansion is fueled by the increasing prevalence of musculoskeletal conditions and sports injuries, advancements in biologic therapies, a growing preference for minimally invasive treatments, and the aging population's shift towards more active lifestyles. Additionally, the demand for treatments that offer faster recovery and fewer complications compared to traditional surgical methods further bolsters the market's growth, establishing orthobiologic treatments as promising alternatives in regenerative medicine. Orthobiologics play an essential role in soft tissue healing, enhancing the repair of ligaments, tendons, and cartilage. For instance, viscosupplementation using hyaluronic acid derivatives is becoming a popular treatment for knee osteoarthritis, providing lubrication and cushioning for arthritic joints. Similarly, stem cell-based therapies are being adopted to treat various orthopedic conditions, including osteoarthritis and degenerative disc disease, due to their potential to regenerate damaged tissues and improve joint function. Despite the promising potential and growing market, the clinical efficacy of orthobiologics is still under investigation. The need for high-quality, reproducible research is critical to establishing standardized treatment protocols and ensuring the safety and effectiveness of these therapies in clinical practice.

Current evidence base

Table 1 summarizes the indications for orthobiologic products and the level of evidence supporting their usage across various musculoskeletal conditions. We used the Level of Evidence table to ascertain the level of evidence of the studies available for a given orthobiologic[15]. Further, we graded the studies for their quality, based on the bias in their study design into high, moderate, and low[16,17]. For example: Despite the availability of randomized controlled trials (RCTs) for a given orthobiologic if the quality parameters are not satisfied they are downgraded by one level. Orthobiologics such as PRP, BMAC/Microfragmented Adipose Tissue/(SVF/MFAT/AD-MSC), allogeneic MSC, and cultured chondrocytes are utilized in treating a range of disorders, including knee osteoarthritis, avascular necrosis of the femoral head, tendinopathies, adhesive capsulitis, plantar fasciitis, degenerative disc disease, fractures, anterior cruciate ligament (ACL) augmentation, meniscus repair, rotator cuff repair augmentation, ankle sprains, acute muscle injuries, ankle osteoarthritis, and carpal tunnel syndrome[18-78].

Table 1 Orthobiologic usage indications and the level of evidence supporting its usage.
Indication
Orthobiologic product
Level of evidence
Ref.
Knee osteoarthritisPRP1[18-20]
BMAC1[21,22]
SVF/MFAT/AD-MSC1[23-25]
Allogeneic MSC1[26-28]
Cultured chondrocytes (ACI/MACI)1[29]
Avascular necrosis of femoral headBMAC1[30-32]
SVF/MFAT/AD-MSC4[33,34]
Cultured osteoblasts4[35-37]
Lateral epicondylitisPRP1[1,38]
BMAC1[39]
Achilles tendinopathyPRP1[40,41]
BMAC1[39]
Patellar tendinopathyPRP1[42,43]
BMAC1[39]
Adhesive capsulitisPRP1[44,45]
Plantar fasciitis PRP1[46,47]
SVF4[48]
Degenerative disc diseasePRP1[49,50]
BMAC1[51-53]
FracturePRP1[54-56]
BMAC1[57-59]
ACL augmentPRP1[60,61]
BMAC1[62]
Meniscus repairPRP1[63-65]
BMAC4[66,67]
MFAT1[68]
Rotator cuff repair augmentPRP1[69,70]
BMAC1[39,71,72]
Ankle sprainPRP1[73]
Acute muscle injuriesPRP1[74]
Ankle osteoarthritis PRP1[75,76]
Carpal tunnel syndrome PRP 1[77,78]

For knee osteoarthritis, multiple orthobiologics such as PRP, BMAC, SVF/MFAT/AD-MSC, allogeneic MSC, and cultured chondrocytes demonstrate a high level of evidence (Level 1). Similarly, PRP and BMAC show strong support (Level 1) in the treatment of conditions like lateral epicondylitis, Achilles tendinopathy, patellar tendinopathy, adhesive capsulitis, plantar fasciitis, degenerative disc disease, fractures, ACL augmentation, and meniscus repair (with MFAT also showing Level 1 evidence). In contrast, avascular necrosis of the femoral head shows high-level evidence (Level 1) for BMAC, but a lower level of evidence (Level 4) for SVF/MFAT/AD-MSC and cultured osteoblasts. Other conditions like plantar fasciitis treated with SVF and meniscus repair with BMAC exhibit Level 4 evidence, suggesting the need for further research to validate their efficacy. The data suggest that PRP and BMAC are extensively supported by high-quality evidence across multiple indications, particularly in tendinopathies and joint-related conditions. SVF/MFAT/AD-MSC and cultured cells, while promising, require more rigorous research to establish their efficacy conclusively. This evidence highlights the progressive adoption and validation of orthobiologics in clinical practice, yet underscores the variability in the level of support depending on the specific condition as shown in Figure 1, and treatment modality as shown in Table 1. It is important to note that 'cultured chondrocytes' in the context of knee osteoarthritis treatment refers to techniques such as ACI or MACI, where a patient's cartilage cells are cultured and then reimplanted to repair cartilage defects.

Figure 1
Figure 1 Strength of evidence supporting the use of orthobiologics in various inflammatory conditions. ACL: Anterior cruciate ligament; PCL: Posterior cruciate ligament; TFCC: Triangular fibrocartilage complex.
Challenges and future directions

The development and application of orthobiologics face significant regulatory challenges. Regulatory bodies like the European Medicines Agency and the United States Food and Drug Administration require extensive clinical data to ensure safety and efficacy, but the rapid advancement of these therapies often outpaces regulatory frameworks. This mismatch can lead to delays in approval and commercialization[79-81]. Ethically, the use of stem cells and other biological materials raises concerns about patient safety, informed consent, and potential exploitation. For instance, the sourcing of stem cells, whether from autologous (self) or allogeneic (donor) origins, necessitates strict ethical oversight to prevent misuse and ensure that patients are fully aware of the risks and benefits. Additionally, there is an ongoing debate regarding the commercialization of stem cell therapies, with concerns about equity and access to these potentially life-changing treatments[82].

Despite the promising potential of orthobiologics, several research gaps need to be addressed. One of the key gaps is the lack of standardized protocols for the preparation and application of these therapies. The variability in methods for isolating and concentrating biological materials, such as PRP and stem cells, leads to inconsistent results across studies, making it challenging to draw definitive conclusions about their efficacy. Moreover, long-term safety and efficacy data are scarce. Most studies focus on short-term outcomes, and there is a need for longitudinal studies that follow patients over several years to understand the durability of the benefits and any potential long-term adverse effects. There is also a need for more high-quality RCTs to provide robust evidence that can guide clinical practice.

The future of orthobiologics is promising, driven by advancements in biotechnology and personalized medicine. One exciting prospect is the development of more sophisticated delivery systems that can target biological materials precisely to the site of injury or disease. For instance, advances in nanotechnology and drug delivery systems could enhance the efficacy of growth factors and stem cells by ensuring sustained and controlled release. Personalized medicine also holds great potential for orthobiologics. By leveraging genomic and proteomic data, clinicians can tailor biologic therapies to the individual patient’s biological profile, improving outcomes and reducing the risk of adverse reactions. This approach aligns with the broader trend in medicine towards more personalized and precision-based treatments.

iPSCs represent a promising frontier for future orthobiologic therapies, offering several key advantages that could revolutionize the field. Derived from adult somatic cells, iPSCs circumvent the ethical concerns associated with embryonic stem cells while providing a patient-specific, autologous source that minimizes the risk of immune rejection and eliminates the need for immunosuppression. The scalability of iPSCs is another significant benefit, as established iPSC lines can be expanded indefinitely, potentially offering an unlimited source of cells for therapeutic applications. Moreover, iPSCs possess the capacity for directed differentiation into various cell types relevant to orthopedic tissues, such as osteoblasts, chondrocytes, and tenocytes. Additionally, patient-derived iPSCs enable the creation of in vitro models of orthopedic conditions, facilitating drug discovery and personalized treatment approaches. Future research directions in the field may focus on developing efficient and safe methods for generating clinical-grade iPSCs, optimizing protocols for their differentiation into specific orthopedic cell types, investigating the use of iPSC-derived organoids in tissue engineering, and exploring the integration of gene editing technologies like CRISPR to correct genetic defects in patient-derived cells before transplantation. Furthermore, long-term safety studies are essential to assess the risks of tumorigenicity and other potential adverse effects associated with iPSC-based therapies. Ongoing research into the mechanisms of action of different orthobiologic materials will likely yield new insights that can be translated into clinical practice. Understanding how these materials interact with the body at a molecular level will enable the development of more effective and safer treatments.

CONCLUSION

The field of orthobiologics shows immense potential in enhancing musculoskeletal healing and regeneration. Despite promising clinical applications and market growth, the evidence supporting their efficacy remains variable and sometimes inconsistent. High-quality, standardized research is critical to address these gaps, validate treatment protocols, and ensure safety and effectiveness. Regulatory and ethical challenges also demand attention to facilitate the responsible development and use of these therapies. Future advancements in delivery systems and personalized medicine hold promise for more targeted and effective treatments. The orthopaedic and regenerative medicine communities must prioritize rigorous, high-quality research to establish standardized protocols and robust evidence for orthobiologics. Regulatory bodies should streamline approval processes without compromising safety. Collaborative efforts are essential to overcome existing challenges and fully realize the therapeutic potential of orthobiologics, ultimately improving patient outcomes.

Footnotes

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

Peer-review model: Single blind

Specialty type: Orthopedics

Country of origin: India

Peer-review report’s classification

Scientific Quality: Grade C, Grade C

Novelty: Grade B, Grade B

Creativity or Innovation: Grade B, Grade B

Scientific Significance: Grade B, Grade B

P-Reviewer: Ignatyeva N S-Editor: Liu H L-Editor: A P-Editor: Zhao YQ

References
1.  Muthu S, Patel S, Gobbur A, Patil SC, Ks KH, Yadav V, Jeyaraman M. Platelet-rich plasma therapy ensures pain reduction in the management of lateral epicondylitis - a PRISMA-compliant network meta-analysis of randomized controlled trials. Expert Opin Biol Ther. 2022;22:535-546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 10]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
2.  Ranjan R, Kumar R, Jeyaraman M, Arora A, Kumar S, Nallakumarasamy A. Autologous platelet-rich plasma in the delayed union of long bone fractures - A quasi experimental study. J Orthop. 2023;36:76-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
3.  Jeyaraman M, Jeyaraman N, Jayakumar T, Ramasubramanian S, Ranjan R, Jha SK, Gupta A. Efficacy of stromal vascular fraction for knee osteoarthritis: A prospective, single-centre, non-randomized study with 2 years follow-up. World J Orthop. 2024;15:457-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
4.  Muthu S, Patel S, Selvaraj P, Jeyaraman M. Comparative analysis of leucocyte poor vs leucocyte rich platelet-rich plasma in the management of lateral epicondylitis: Systematic review & meta-analysis of randomised controlled trials. J Clin Orthop Trauma. 2021;19:96-107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
5.  Zhao K, Liu YS, Nie LY, Qian LN, Nie NF, Leptihn S, Bunpetch V, Xu JQ, Zou XH, Ouyang H. The influence of sample size and gender composition on the meta-analysis conclusion of platelet-rich plasma treatment for osteoarthritis. J Orthop Translat. 2020;22:34-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
6.  Poff G, Spencer E, Scott B, Sleadd R, Broyles J. Comparison of Clinical Outcomes after Platelet-Rich Plasma and Rotator Cuff Repair in High-Grade Intrasubstance Partial Rotator Cuff Tears. J Clin Med. 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
7.  Pabinger C, Lothaller H, Kobinia GS. Intra-articular injection of bone marrow aspirate concentrate (mesenchymal stem cells) in KL grade III and IV knee osteoarthritis: 4 year results of 37 knees. Sci Rep. 2024;14:2665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
8.  Cho WS, Chung SG, Kim W, Jo CH, Lee SU, Lee SY. Mesenchymal Stem Cells Use in the Treatment of Tendon Disorders: A Systematic Review and Meta-Analysis of Prospective Clinical Studies. Ann Rehabil Med. 2021;45:274-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
9.  Qu W, Wang Z, Hunt C, Morrow AS, Urtecho M, Amin M, Shah S, Hasan B, Abd-Rabu R, Ashmore Z, Kubrova E, Prokop LJ, Murad MH. The Effectiveness and Safety of Platelet-Rich Plasma for Chronic Wounds: A Systematic Review and Meta-analysis. Mayo Clin Proc. 2021;96:2407-2417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 25]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
10.  Cavallo C, Roffi A, Grigolo B, Mariani E, Pratelli L, Merli G, Kon E, Marcacci M, Filardo G. Platelet-Rich Plasma: The Choice of Activation Method Affects the Release of Bioactive Molecules. Biomed Res Int. 2016;2016:6591717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 139]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
11.  Han Y, Huang H, Pan J, Lin J, Zeng L, Liang G, Yang W, Liu J. Meta-analysis Comparing Platelet-Rich Plasma vs Hyaluronic Acid Injection in Patients with Knee Osteoarthritis. Pain Med. 2019;20:1418-1429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 63]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
12.  Jeyaraman M, Karthik KS, Choudary D, Jeyaraman N, Nallakumarasamy A, Ramasubramian S. Autologous Bone Marrow Aspiration Concentrate (BMAC) Therapy for Primary Knee Osteoarthritis-An Observational and Dose Escalation Study. Indian J Orthop. 2024;58:1016-1026.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
13.  Jeyaraman M, Jeyaraman N, Ramasubramanian S, Ranjan R, Jha SK, Gupta A. Bone Marrow Aspirate Concentrate for Treatment of Primary Knee Osteoarthritis: A Prospective, Single-Center, Non-randomized Study with 2-Year Follow-Up. Indian J Orthop. 2024;58:894-904.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
14.  Rodeo SA. Orthobiologics: Current Status in 2023 and Future Outlook. J Am Acad Orthop Surg. 2023;31:604-613.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
15.  Burns PB, Rohrich RJ, Chung KC. The levels of evidence and their role in evidence-based medicine. Plast Reconstr Surg. 2011;128:305-310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 936]  [Cited by in F6Publishing: 1224]  [Article Influence: 94.2]  [Reference Citation Analysis (0)]
16.  Sterne JAC, Savović J, Page MJ, Elbers RG, Blencowe NS, Boutron I, Cates CJ, Cheng HY, Corbett MS, Eldridge SM, Emberson JR, Hernán MA, Hopewell S, Hróbjartsson A, Junqueira DR, Jüni P, Kirkham JJ, Lasserson T, Li T, McAleenan A, Reeves BC, Shepperd S, Shrier I, Stewart LA, Tilling K, White IR, Whiting PF, Higgins JPT. RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ. 2019;366:l4898.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6581]  [Cited by in F6Publishing: 11432]  [Article Influence: 2286.4]  [Reference Citation Analysis (0)]
17.  Sterne JA, Hernán MA, Reeves BC, Savović J, Berkman ND, Viswanathan M, Henry D, Altman DG, Ansari MT, Boutron I, Carpenter JR, Chan AW, Churchill R, Deeks JJ, Hróbjartsson A, Kirkham J, Jüni P, Loke YK, Pigott TD, Ramsay CR, Regidor D, Rothstein HR, Sandhu L, Santaguida PL, Schünemann HJ, Shea B, Shrier I, Tugwell P, Turner L, Valentine JC, Waddington H, Waters E, Wells GA, Whiting PF, Higgins JP. ROBINS-I: a tool for assessing risk of bias in non-randomised studies of interventions. BMJ. 2016;355:i4919.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7683]  [Cited by in F6Publishing: 8769]  [Article Influence: 1096.1]  [Reference Citation Analysis (2)]
18.  Simental-Mendía M, Ortega-Mata D, Tamez-Mata Y, Olivo CAA, Vilchez-Cavazos F. Comparison of the clinical effectiveness of activated and non-activated platelet-rich plasma in the treatment of knee osteoarthritis: a systematic review and meta-analysis. Clin Rheumatol. 2023;42:1397-1408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
19.  Xiong Y, Gong C, Peng X, Liu X, Su X, Tao X, Li Y, Wen Y, Li W. Efficacy and safety of platelet-rich plasma injections for the treatment of osteoarthritis: a systematic review and meta-analysis of randomized controlled trials. Front Med (Lausanne). 2023;10:1204144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 16]  [Reference Citation Analysis (0)]
20.  Nie LY, Zhao K, Ruan J, Xue J. Effectiveness of Platelet-Rich Plasma in the Treatment of Knee Osteoarthritis: A Meta-analysis of Randomized Controlled Clinical Trials. Orthop J Sports Med. 2021;9:2325967120973284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
21.  Keeling LE, Belk JW, Kraeutler MJ, Kallner AC, Lindsay A, McCarty EC, Postma WF. Bone Marrow Aspirate Concentrate for the Treatment of Knee Osteoarthritis: A Systematic Review. Am J Sports Med. 2022;50:2315-2323.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 29]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
22.  Belk JW, Lim JJ, Keeter C, McCulloch PC, Houck DA, McCarty EC, Frank RM, Kraeutler MJ. Patients With Knee Osteoarthritis Who Receive Platelet-Rich Plasma or Bone Marrow Aspirate Concentrate Injections Have Better Outcomes Than Patients Who Receive Hyaluronic Acid: Systematic Review and Meta-analysis. Arthroscopy. 2023;39:1714-1734.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 14]  [Reference Citation Analysis (0)]
23.  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: 38]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
24.  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: 42]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
25.  Bolia IK, Bougioukli S, Hill WJ, Trasolini NA, Petrigliano FA, Lieberman JR, Weber AE. Clinical Efficacy of Bone Marrow Aspirate Concentrate Versus Stromal Vascular Fraction Injection in Patients With Knee Osteoarthritis: A Systematic Review and Meta-analysis. Am J Sports Med. 2022;50:1451-1461.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 18]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
26.  Han X, Yang B, Zou F, Sun J. Clinical therapeutic efficacy of mesenchymal stem cells derived from adipose or bone marrow for knee osteoarthritis: a meta-analysis of randomized controlled trials. J Comp Eff Res. 2020;9:361-374.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 25]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
27.  Jeyaraman M, Muthu S, Nischith DS, Jeyaraman N, Nallakumarasamy A, Khanna M. PRISMA-Compliant Meta-Analysis of Randomized Controlled Trials on Osteoarthritis of Knee Managed with Allogeneic vs Autologous MSCs: Efficacy and Safety Analysis. Indian J Orthop. 2022;56:2042-2059.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 8]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
28.  Iijima H, Isho T, Kuroki H, Takahashi M, Aoyama T. Effectiveness of mesenchymal stem cells for treating patients with knee osteoarthritis: a meta-analysis toward the establishment of effective regenerative rehabilitation. NPJ Regen Med. 2018;3:15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 68]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
29.  Dhillon J, Decilveo AP, Kraeutler MJ, Belk JW, McCulloch PC, Scillia AJ. Third-Generation Autologous Chondrocyte Implantation (Cells Cultured Within Collagen Membrane) Is Superior to Microfracture for Focal Chondral Defects of the Knee Joint: Systematic Review and Meta-analysis. Arthroscopy. 2022;38:2579-2586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 12]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
30.  Jindal K, Aggarwal S, Kumar P, Rathod P. Core decompression with bone marrow aspirate concentrate in post collapse avascular necrosis of hip: A systematic review and meta-analysis. J Clin Orthop Trauma. 2021;17:78-87.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
31.  Ulusoy İ, Yılmaz M, Kıvrak A. Efficacy of autologous stem cell therapy in femoral head avascular necrosis: a comparative study. J Orthop Surg Res. 2023;18:799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
32.  Jeyaraman M, Muthu S, Jain R, Khanna M. Autologous bone marrow derived mesenchymal stem cell therapy for osteonecrosis of femoral head: A systematic overview of overlapping meta-analyses. J Clin Orthop Trauma. 2021;13:134-142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
33.  Tantuway V, Jeyaraman M, Nallakumarasamy A, Prikh MB, Sharma AK, Sharma R. Functional Outcome Analysis of Autologous Stromal Vascular Fraction (SVF) (Sahaj Therapy(®)) Using Direct Sonication in Osteonecrosis of the Femoral Head (ONFH): A 6-Year Follow-Up Study. Indian J Orthop. 2024;58:68-78.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
34.  Zhang W, Zheng C, Yu T, Zhang H, Huang J, Chen L, Tong P, Zhen G. The therapeutic effect of adipose-derived lipoaspirate cells in femoral head necrosis by improving angiogenesis. Front Cell Dev Biol. 2022;10:1014789.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
35.  Patro BP, Jeyaraman N, Jayakumar T, Das G, Nallakumarasamy A, Jeyaraman M. Efficacy of Autologous Adult Live-Cultured Osteoblast (AALCO) Implantation in Avascular Necrosis of the Femoral Head: A Mid-Term Outcome Analysis. Indian J Orthop. 2024;58:1053-1063.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
36.  Sadat-Ali M, Al-Omran AS, AlTabash K, Acharya S, Hegazi TM, Al Muhaish MI. The clinical and radiological effectiveness of autologous bone marrow derived osteoblasts (ABMDO) in the management of avascular necrosis of femoral head (ANFH) in sickle cell disease (SCD). J Exp Orthop. 2022;9:18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
37.  Palekar G, Bhalodiya HP, Archik S, Trivedi K. Retrospective Study on Implantation of Autologous-Cultured Osteoblasts for the Treatment of Patients with Avascular Necrosis of the Femoral Head. Orthop Res Rev. 2021;13:15-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
38.  Chen XT, Fang W, Jones IA, Heckmann ND, Park C, Vangsness CT Jr. The Efficacy of Platelet-Rich Plasma for Improving Pain and Function in Lateral Epicondylitis: A Systematic Review and Meta-analysis with Risk-of-Bias Assessment. Arthroscopy. 2021;37:2937-2952.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
39.  Imam MA, Holton J, Horriat S, Negida AS, Grubhofer F, Gupta R, Narvani A, Snow M. A systematic review of the concept and clinical applications of bone marrow aspirate concentrate in tendon pathology. SICOT J. 2017;3:58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 48]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
40.  Nauwelaers AK, Van Oost L, Peers K. Evidence for the use of PRP in chronic midsubstance Achilles tendinopathy: A systematic review with meta-analysis. Foot Ankle Surg. 2021;27:486-495.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 25]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
41.  Arthur Vithran DT, Xie W, Opoku M, Essien AE, He M, Li Y. The Efficacy of Platelet-Rich Plasma Injection Therapy in the Treatment of Patients with Achilles Tendinopathy: A Systematic Review and Meta-Analysis. J Clin Med. 2023;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
42.  Dupley L, Charalambous CP. Platelet-Rich Plasma Injections as a Treatment for Refractory Patellar Tendinosis: A Meta-Analysis of Randomised Trials. Knee Surg Relat Res. 2017;29:165-171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 36]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
43.  Barman A, Sinha MK, Sahoo J, Jena D, Patel V, Patel S, Bhattacharjee S, Baral D. Platelet-rich plasma injection in the treatment of patellar tendinopathy: a systematic review and meta-analysis. Knee Surg Relat Res. 2022;34:22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 14]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
44.  Yu S, Hu R, Feng H, Huang D. Efficacy of platelet-rich plasma injection in the treatment of frozen shoulder: A systematic review and meta-analysis. J Back Musculoskelet Rehabil. 2023;36:551-564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
45.  Lin HW, Tam KW, Liou TH, Rau CL, Huang SW, Hsu TH. Efficacy of Platelet-Rich Plasma Injection on Range of Motion, Pain, and Disability in Patients With Adhesive Capsulitis: A Systematic Review and Meta-analysis. Arch Phys Med Rehabil. 2023;104:2109-2122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
46.  Hohmann E, Tetsworth K, Glatt V. Platelet-Rich Plasma Versus Corticosteroids for the Treatment of Plantar Fasciitis: A Systematic Review and Meta-analysis. Am J Sports Med. 2021;49:1381-1393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 27]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
47.  Fei X, Lang L, Lingjiao H, Wei C, Zhou X. Platelet-rich plasma has better mid-term clinical results than traditional steroid injection for plantar fasciitis: A systematic review and meta-analysis. Orthop Traumatol Surg Res. 2021;107:103007.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
48.  Wu B, Xiao S, Yang S, Wei Z, Deng C. A New Minimally Invasive Procedure for Treating Plantar Heel Pain: Stromal Vascular Fraction Gel Grafting. Ann Plast Surg. 2023;91:609-613.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
49.  Schneider BJ, Hunt C, Conger A, Qu W, Maus TP, Vorobeychik Y, Cheng J, Duszynski B, McCormick ZL. The effectiveness of intradiscal biologic treatments for discogenic low back pain: a systematic review. Spine J. 2022;22:226-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
50.  Chang MC, Park D. The Effect of Intradiscal Platelet-Rich Plasma Injection for Management of Discogenic Lower Back Pain: A Meta-Analysis. J Pain Res. 2021;14:505-512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
51.  Her YF, Kubrova E, Martinez Alvarez GA, D'Souza RS. The Analgesic Efficacy of Intradiscal Injection of Bone Marrow Aspirate Concentrate and Culture-Expanded Bone Marrow Mesenchymal Stromal Cells in Discogenic Pain: A Systematic Review. J Pain Res. 2022;15:3299-3318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
52.  Law L, Hunt CL, van Wijnen AJ, Nassr A, Larson AN, Eldrige JS, Mauck WD, Pingree MJ, Yang J, Muir CW, Erwin PJ, Bydon M, Qu W. Office-Based Mesenchymal Stem Cell Therapy for the Treatment of Musculoskeletal Disease: A Systematic Review of Recent Human Studies. Pain Med. 2019;20:1570-1583.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
53.  Yim RL, Lee JT, Bow CH, Meij B, Leung V, Cheung KM, Vavken P, Samartzis D. A systematic review of the safety and efficacy of mesenchymal stem cells for disc degeneration: insights and future directions for regenerative therapeutics. Stem Cells Dev. 2014;23:2553-2567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 69]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
54.  Jamal MS, Hurley ET, Asad H, Asad A, Taneja T. The role of Platelet Rich Plasma and other orthobiologics in bone healing and fracture management: A systematic review. J Clin Orthop Trauma. 2022;25:101759.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 11]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
55.  Li S, Xing F, Luo R, Liu M. Clinical Effectiveness of Platelet-Rich Plasma for Long-Bone Delayed Union and Nonunion: A Systematic Review and Meta-Analysis. Front Med (Lausanne). 2021;8:771252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 9]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
56.  Zhang Y, Xing F, Luo R, Duan X. Platelet-Rich Plasma for Bone Fracture Treatment: A Systematic Review of Current Evidence in Preclinical and Clinical Studies. Front Med (Lausanne). 2021;8:676033.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
57.  Palombella S, Lopa S, Gianola S, Zagra L, Moretti M, Lovati AB. Bone Marrow-Derived Cell Therapies to Heal Long-Bone Nonunions: A Systematic Review and Meta-Analysis-Which Is the Best Available Treatment? Stem Cells Int. 2019;2019:3715964.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
58.  Attia AK, Robertson GAJ, McKinley J, d'Hooghe PP, Maffulli N. Surgical Management of Jones Fractures in Athletes: Orthobiologic Augmentation: A Systematic Review and Meta-analysis of 718 Fractures. Am J Sports Med. 2023;51:2216-2228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
59.  Imam MA, Holton J, Ernstbrunner L, Pepke W, Grubhofer F, Narvani A, Snow M. A systematic review of the clinical applications and complications of bone marrow aspirate concentrate in management of bone defects and nonunions. Int Orthop. 2017;41:2213-2220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 59]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
60.  Zhu T, Zhou J, Hwang J, Xu X. Effects of Platelet-Rich Plasma on Clinical Outcomes After Anterior Cruciate Ligament Reconstruction: A Systematic Review and Meta-analysis. Orthop J Sports Med. 2022;10:23259671211061535.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 5]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
61.  Lv ZT, Zhang JM, Pang ZY, Wang Z, Huang JM, Zhu WT. The efficacy of platelet rich plasma on anterior cruciate ligament reconstruction: a systematic review and meta-analysis. Platelets. 2022;33:229-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
62.  Cao Y, Zhang Z, Song G, Ni Q, Zheng T, Li Y. Biological enhancement methods may be a viable option for ACL arthroscopic primary repair - A systematic review. Orthop Traumatol Surg Res. 2022;108:103227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
63.  Li Z, Weng X. Platelet-rich plasma use in meniscus repair treatment: a systematic review and meta-analysis of clinical studies. J Orthop Surg Res. 2022;17:446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
64.  Zaffagnini S, Poggi A, Reale D, Andriolo L, Flanigan DC, Filardo G. Biologic Augmentation Reduces the Failure Rate of Meniscal Repair: A Systematic Review and Meta-analysis. Orthop J Sports Med. 2021;9:2325967120981627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
65.  Xie Y, Xing Q, Wang S, Yang Z, Hu A, Wu Q. Can platelet-rich plasma enhance the effect of meniscus repair? A meta-analysis of randomized controlled trials Platelet-rich plasma and meniscus repair. J Orthop Surg (Hong Kong). 2022;30:10225536221131483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
66.  Massey PA, Sampognaro G, Starnes E, Lowery MT, Duncan M, Sherman WF, Zhang AS. Improved Outcomes After Reinforced Radial Meniscus Repair Augmented With Bone Marrow Aspirate Concentrate. Arthrosc Sports Med Rehabil. 2023;5:e843-e851.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
67.  Dancy ME, Marigi EM, Krych AJ, Werner BC, Camp CL. Impact of Biologic Augmentation on Revision Surgery Rates After Meniscus Repair: A Matched-Cohort Analysis of 3420 Patients. Orthop J Sports Med. 2023;11:23259671231186990.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
68.  Conte P, Anzillotti G, Di Matteo B, Gallese A, Vitale U, Marcacci M, Kon E. Orthobiologic injections for treating degenerative meniscus lesions: a matter of facts? Ten years of clinical experience in a systematic review. J Cartilage Joint Preser. 2023;3:100104.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Zhao JG, Zhao L, Jiang YX, Wang ZL, Wang J, Zhang P. Platelet-rich plasma in arthroscopic rotator cuff repair: a meta-analysis of randomized controlled trials. Arthroscopy. 2015;31:125-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 61]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
70.  Hurley ET, Lim Fat D, Moran CJ, Mullett H. The Efficacy of Platelet-Rich Plasma and Platelet-Rich Fibrin in Arthroscopic Rotator Cuff Repair: A Meta-analysis of Randomized Controlled Trials. Am J Sports Med. 2019;47:753-761.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 130]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
71.  Muthu S, Mogulesh C, Viswanathan VK, Jeyaraman N, Pai SN, Jeyaraman M, Khanna M. Is cellular therapy beneficial in management of rotator cuff tears? Meta-analysis of comparative clinical studies. World J Meta-Anal. 2022;10:162-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
72.  Liu F, Meng Q, Yin H, Yan Z. Stem Cells in Rotator Cuff Injuries and Reconstructions: A Systematic Review and Meta-Analysis. Curr Stem Cell Res Ther. 2019;14:683-697.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
73.  Milo AMM, Braganza CL. Platelet-Rich Plasma on Ankle Sprains - Efficacy on Pain Reduction and Shorter Return to Play: A Systematic Review of Available Randomized Control Trials. J Med University St Tomas. 2023;7:1153-1160.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Grassi A, Napoli F, Romandini I, Samuelsson K, Zaffagnini S, Candrian C, Filardo G. Is Platelet-Rich Plasma (PRP) Effective in the Treatment of Acute Muscle Injuries? A Systematic Review and Meta-Analysis. Sports Med. 2018;48:971-989.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 73]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
75.  Laohajaroensombat S, Prusmetikul S, Rattanasiri S, Thakkinstian A, Woratanarat P. Platelet-rich plasma injection for the treatment of ankle osteoarthritis: a systematic review and meta-analysis. J Orthop Surg Res. 2023;18:373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
76.  Ding SL, Ji LF, Zhang MZ, Xiong W, Sun CY, Han ZY, Wang C. Safety and efficacy of intra-articular injection of platelet-rich plasma for the treatment of ankle osteoarthritis: a systematic review and meta-analysis. Int Orthop. 2023;47:1963-1974.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 5]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
77.  Catapano M, Catapano J, Borschel G, Alavinia SM, Robinson LR, Mittal N. Effectiveness of Platelet-Rich Plasma Injections for Nonsurgical Management of Carpal Tunnel Syndrome: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Arch Phys Med Rehabil. 2020;101:897-906.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
78.  Dong C, Sun Y, Qi Y, Zhu Y, Wei H, Wu D, Li C. Effect of Platelet-Rich Plasma Injection on Mild or Moderate Carpal Tunnel Syndrome: An Updated Systematic Review and Meta-Analysis of Randomized Controlled Trials. Biomed Res Int. 2020;2020:5089378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
79.  Master Z, Matthews KRW, Abou-El-Enein M. Unproven stem cell interventions: A global public health problem requiring global deliberation. Stem Cell Reports. 2021;16:1435-1445.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
80.  Guleria I, de Los Angeles Muñiz M, Wilgo M, Bapat A, Cui W, Hsu YS, Jeyaraman M, Muthu S, Rodriguez F, Fesnak A, Celluzzi C, Sesok-Pizzini D, Reich-Slotky R, Spitzer T. How do I: Evaluate the safety and legitimacy of unproven cellular therapies? Transfusion. 2022;62:518-532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
81.  George B. Regulations and guidelines governing stem cell based products: Clinical considerations. Perspect Clin Res. 2011;2:94-99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 38]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
82.  Vaish A, Vaishya R. Stem cells in orthopaedics and sports injuries: A comprehensive review and future research directions. J Orthop Rep. 2024;3:100344.  [PubMed]  [DOI]  [Cited in This Article: ]