Review Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Jun 26, 2025; 17(6): 103775
Published online Jun 26, 2025. doi: 10.4252/wjsc.v17.i6.103775
Challenges in the clinical translation of stromal vascular fraction therapy in regenerative medicine
Naveen Jeyaraman, Sandeep Shrivastava, Arulkumar Nallakumarasamy, Department of Orthopaedics, Datta Meghe Institute of Higher Education and Research, Wardha 442004, Maharashtra, India
Naveen Jeyaraman, Ravi Velamoor Rangarajan, Arulkumar Nallakumarasamy, Swaminathan Ramasubramanian, Avinash Gandi Devadas, Madhan Jeyaraman, Department of Regenerative Medicine, Mother Cell Regenerative Centre, Tiruchirappalli 620017, Tamil Nadu, India
Secunda Rupert, Department of Regenerative Medicine and Research, Government Stanley Hospital, Chennai 600001, Tamil Nadu, India
Madhan Jeyaraman, Department of Orthopaedics, ACS Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600077, Tamil Nadu, India
ORCID number: Naveen Jeyaraman (0000-0002-4362-3326); Ravi Velamoor Rangarajan (0009-0006-7303-8474); Arulkumar Nallakumarasamy (0000-0002-2445-2883); Swaminathan Ramasubramanian (0000-0001-8845-8427); Madhan Jeyaraman (0000-0002-9045-9493).
Author contributions: Jeyaraman N designed the study; Jeyaraman N and Nallakumarasamy A analyzed the articles for review; Jeyaraman N, Nallakumarasamy A, and Ramasubramanian S wrote the manuscript; Shrivastava S, Rangarajan RV, Devadas AG, Rupert S, and Jeyaraman M finalized the manuscript.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
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, Velappanchavadi, Chennai 600077, Tamil Nadu, India. madhanjeyaraman@gmail.com
Received: December 3, 2024
Revised: March 13, 2025
Accepted: May 7, 2025
Published online: June 26, 2025
Processing time: 208 Days and 1.1 Hours

Abstract

Stromal vascular fraction (SVF) therapy is a promising regenerative medicine strategy derived from adipose tissue, containing a heterogeneous mix of cells, including adipose-derived stem, endothelial, and immune cells. Despite its potential in treating conditions like osteoarthritis, chronic wounds, and myocardial ischemia, significant challenges impede its clinical translation. Key obstacles include biological variability in SVF composition, unclear mechanisms of action, regulatory ambiguities, and the technical difficulty of ensuring standardized and scalable isolation methods. Furthermore, patient-specific factors, ethical concerns, and the need for comprehensive efficacy assessment complicate clinical application. Addressing these challenges requires advancements in technology, regulatory flexibility, interdisciplinary collaboration, and personalized therapeutic approaches. Innovations such as automated isolation systems, advanced biomaterials, and CRISPR-based gene editing are potential solutions to improve the therapeutic reliability of SVF. A structured roadmap, including preclinical research, regulatory approval, and post-market surveillance, is proposed to advance SVF therapies from the laboratory to clinical practice. Future directions should focus on large-scale clinical trials, biomarker development, real-world evidence generation, and standardization of protocols to enhance the safety, efficacy, and accessibility of SVF, ultimately realizing its potential as a versatile therapeutic in regenerative medicine.

Key Words: Stromal vascular fraction; Regenerative medicine; Cell therapy; Clinical translation; Adipose-derived stem cells

Core Tip: Stromal vascular fraction (SVF) therapy, derived from adipose tissue and containing a mix of cells, shows promise in treating conditions like osteoarthritis and chronic wounds. However, challenges such as biological variability, unclear mechanisms, regulatory issues, and scalability hinder clinical translation. Addressing these challenges requires technological advancements, regulatory flexibility, and interdisciplinary collaboration. Innovations like automated isolation, advanced biomaterials, and CRISPR-based editing could enhance reliability. A structured roadmap, including preclinical research and large-scale trials, is essential to advance SVF therapies. Future efforts should focus on standardizing protocols and generating real-world evidence to maximize the potential of SVF in regenerative medicine.



INTRODUCTION

Stromal vascular fraction (SVF) represents a heterogeneous mixture of cells derived from adipose tissue, encompassing a variety of cellular components such as mesenchymal stem cells (MSCs), endothelial cells, pericytes, and a multitude of immune cells (Table 1)[1]. SVF contains adipose-derived stem cells (ADSCs), which are a subset of MSCs known for their regenerative capabilities, alongside endothelial progenitor cells that contribute to angiogenesis, and various immune cells that play roles in immunomodulation and inflammation control[2-5]. This complex cellular milieu positions SVF as a promising candidate in regenerative medicine, offering multifaceted therapeutic potential across a spectrum of diseases and conditions. The therapeutic applications of SVF span numerous medical disciplines. In orthopedics, SVF has been employed to treat osteoarthritis, where its anti-inflammatory and regenerative properties help reduce joint pain and enhance mobility[1,6,7]. In cardiology, SVF therapy has shown promise in myocardial ischemia, facilitating myocardial tissue regeneration and improving cardiac function[8,9]. In the realm of plastic and reconstructive surgery, SVF is utilized to improve graft survival and enhance tissue regeneration, particularly in breast reconstruction procedures[10-13]. Furthermore, the immunomodulatory effects of SVF have been explored in wound healing, where it accelerates tissue repair and reduces inflammation, demonstrating its versatility in clinical applications.

Table 1 Cellular composition and roles of stromal vascular fraction.
Cell type
Proportion in SVF
Primary functions
Therapeutic implications
Adipose-derived stem cellsAbout 30%-40%Multipotent differentiation, paracrine signaling, immunomodulationTissue regeneration, anti-inflammatory effects, angiogenesis
Endothelial cellsAbout 10%-15%Formation of new blood vessels (angiogenesis), maintenance of vascular integrityEnhancing blood supply to damaged tissues, improving graft survival
PericytesAbout 5%-10%Stabilization of blood vessels, support of endothelial cellsVascular stabilization, promoting tissue repair
Immune cellsAbout 20%-30%Modulation of immune responses, inflammation controlImmunosuppression in autoimmune diseases, reducing chronic inflammation
Smooth muscle cellsAbout 5%Regulation of blood vessel contraction and blood flowVascular remodeling, support of angiogenesis
FibroblastsAbout 5%Extracellular matrix production, wound healingEnhancing tissue structure and integrity during regeneration
Other cellsAbout 5%-10%Various supportive and regulatory rolesDiverse contributions to tissue repair and immune modulation

Currently, SVF therapy is being explored in both clinical and preclinical settings, with a growing body of evidence supporting its efficacy and safety. Clinical studies, such as those conducted by Onoi et al[14] and Kim et al[15] have demonstrated the beneficial effects of SVF injections in reducing symptoms of hip osteoarthritis and enhancing cartilage repair, respectively. These studies highlight the potential of SVF in improving patient outcomes with minimal adverse effects.

Additionally, randomized controlled trials, like that by Zhang et al[16] have shown that SVF outperforms traditional treatments like hyaluronic acid in managing knee osteoarthritis, offering prolonged effectiveness and reduced risk of clinical failure. Despite these successes, the clinical translation of SVF therapy is not entirely devoid of limitations. Variability in SVF composition, lack of standardized protocols, regulatory hurdles, and technical challenges in SVF isolation and processing are significant impediments[1]. Moreover, the long-term efficacy and safety of SVF therapies remain under investigation, necessitating further research to fully establish their therapeutic potential[5,8,17]. The progression from understanding the cellular composition of SVF to its diverse clinical applications highlights the promising therapeutic potential of the heterogeneous cell population.

However, despite encouraging clinical outcomes across multiple specialties, significant challenges remain in standardizing and optimizing SVF therapy for widespread clinical adoption. This review aimed to provide a comprehensive analysis of the key challenges hindering the clinical translation of SVF therapy in regenerative medicine. All studies included in this review underwent verification for appropriate ethical approval documentation, patient consent procedures, and adherence to international research standards.

BIOLOGICAL CHALLENGES
Heterogeneity of SVF composition

One of the foremost challenges in the clinical application of SVF therapy is the inherent heterogeneity in its cellular composition[4,18,19]. SVF derived from adipose tissue comprises a diverse array of cell types, including ADSCs, endothelial cells, pericytes, and various immune cells. This variability can be attributed to differences in donor characteristics, such as age, gender, body mass index, and metabolic health, as well as variations in isolation and processing techniques. For instance, studies like those conducted by Garza et al[20] and Jeon et al[21] have highlighted significant variability in SVF composition across different samples. Garza et al[20] reported dose-dependent improvements in Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) scores in knee osteoarthritis patients, indicating that the therapeutic efficacy of SVF is influenced by the concentration and proportion of specific cell types within the fraction. Similarly, Jeon et al[21] demonstrated that SVF combined with fat grafting yielded higher fat retention rates, suggesting that the cellular composition of SVF plays a crucial role in tissue regeneration outcomes. This heterogeneity poses substantial challenges for the reproducibility of therapeutic outcomes. Variations in cell populations can lead to inconsistent efficacy, making it difficult to standardize treatments and predict patient responses[22-25]. Moreover, the lack of uniformity complicates the comparison of results across different studies, thereby hindering the establishment of robust clinical protocols.

Mechanism of action

Understanding the precise mechanisms through which SVF exerts its therapeutic effects is pivotal to optimize its clinical application. The therapeutic potential of SVF is largely attributed to the paracrine signaling of its constituent cells[8,26]. ADSCs secrete a plethora of cytokines, growth factors, and extracellular vesicles that modulate the microenvironment, promoting angiogenesis, reducing inflammation, and facilitating tissue repair[27-29]. In addition, SVF promotes fibroblast proliferation and contributes to extracellular matrix remodeling by promoting collagen synthesis and matrix metalloproteinase activity, which degrades damaged tissue and supports structural integrity[30]. Key factors such as vascular endothelial growth factor, transforming growth factor, and tumor necrosis factor play significant roles in these processes. Furthermore, SVF reduces chronic inflammation by secreting anti-inflammatory cytokines like interleukin-10 and shifting macrophages from the pro-inflammatory M1 phenotype to the reparative M2 phenotype[5]. This modulation limits fibrosis and promotes functional tissue regeneration. Angiogenesis is stimulated through the upregulation of endothelial-specific CD31 expression and the release of microRNA-containing exosomes that enhance vascular stability[31]. SVF also operates effectively in hypoxic and low-nutrient conditions, increasing epidermal growth factor and transforming growth factor-β1 secretion to sustain cell proliferation[30].

Despite this knowledge, the exact pathways and interactions governing the therapeutic actions of SVF remain partially elucidated. For example, Onoi et al[14] observed that SVF injections in the hip joint led to significant reductions in osteoarthritis symptoms, presumably through immunomodulatory and regenerative effects. However, the specific cellular interactions and molecular pathways involved in mediating these benefits are not fully understood. This gap in knowledge impedes the targeted enhancement of SVF therapies. Without a comprehensive understanding of how SVF components interact and contribute to tissue regeneration, it becomes challenging to manipulate these factors for improved therapeutic outcomes. Furthermore, the complexity of the cellular interactions of SVF necessitates advanced analytical techniques to dissect the underlying mechanisms, which are currently limited.

Longevity and efficacy

Another critical biological challenge is the limited lifespan and sustained efficacy of SVF cells post-transplantation. Once introduced into the target tissue, SVF cells must survive, integrate, and function effectively within the local environment to exert their therapeutic benefits[22,32-34]. In allogeneic scenarios, factors such as immune rejection, apoptosis, and unfavorable microenvironmental conditions can compromise cell survival and functionality. Zhang et al[16] demonstrated that SVF provided prolonged pain management and improved knee functionality for an average of 61.5 months, compared to the 30.3 months of hyaluronic acid. This suggests that SVF can offer sustained therapeutic benefits; however, the mechanisms underlying this prolonged efficacy are not entirely clear. Understanding the factors that influence cell longevity and efficacy is essential to enhance the therapeutic potential of SVF. Optimizing cell survival and integration involves several strategies, including preconditioning of SVF cells, use of biomaterials to support cell delivery, and genetic modification to enhance cell resilience[1]. Additionally, identifying biomarkers that predict cell viability and therapeutic outcomes can aid in patient stratification and personalized treatment approaches. Addressing these aspects is crucial to ensure the long-term success of SVF therapies in clinical settings. Biological challenges in SVF therapy are depicted in Figure 1.

Figure 1
Figure 1 Biological and mechanistic challenges in stromal vascular fraction therapy. SVF: Stromal vascular fraction.
REGULATORY CHALLENGES
Classification and standardization issues

The regulatory landscape for SVF therapies is complex and varies significantly across different jurisdictions. Regulatory agencies such as the United States Food and Drug Administration (FDA) and the European Medicines Agency (EMA) classify SVF products based on their intended use, processing methods, and cellular composition. Typically, SVF is categorized as a biological product or a medical device, depending on the extent of manipulation it undergoes before clinical application[35-38]. One of the primary challenges is the lack of standardized methods for SVF isolation, characterization, and storage. For instance, studies like those by Perdomo-Pantoja et al[39] and Choi et al[40] have utilized different enzymatic digestion and mechanical separation techniques to isolate SVF, leading to variability in cell yield and composition. This lack of standardization complicates the regulatory approval process, as consistent and reproducible manufacturing practices are essential to meet regulatory standards.

Moreover, the classification of SVF as either a minimally manipulated product or an advanced therapy medicinal product has significant implications for its regulatory pathway[41]. The FDA, for example, requires rigorous testing and approval processes for advanced therapy medicinal products, which can be time-consuming and costly. Conversely, if SVF is classified as minimally manipulated, it may be subject to less stringent regulations, potentially facilitating faster clinical translation but raising concerns about product consistency and safety.

Automated isolation systems have emerged as a promising approach to optimizing SVF processing by enhancing efficiency and standardization. Automated systems minimize variability, reduce contamination risks, and improve reproducibility compared to manual methods. Studies highlight that closed, enzyme-based systems ensure a higher yield of viable cells while maintaining sterility, thereby complying with good manufacturing practices (GMP)[42]. Additionally, integrating real-time monitoring technologies into these systems allows for precise control of enzymatic digestion, filtration, and centrifugation parameters, ensuring consistent SVF composition[1].

Advanced biomaterials for cell delivery further enhance the therapeutic potential of SVF by improving engraftment and cell viability. Hydrogels, for instance, provide a three-dimensional scaffold that supports cell adhesion and proliferation, optimizing the regenerative capacity of SVF in wound healing and cartilage repair applications[43]. Furthermore, incorporating bioactive molecules, such as growth factors or cytokines, within these scaffolds enhances SVF-mediated tissue regeneration by modulating the local microenvironment[30].

Flow cytometry-based characterization of SVF cell populations allows for the standardization of therapeutic cell doses and ensures the presence of key regenerative cell types while eliminating contaminants[5]. Additionally, cytokine profiling can be employed to assess the functional activity of SVF, providing insights into its immunomodulatory and anti-inflammatory properties[44]. Implementing these optimization strategies will enhance the clinical applicability of SVF in regenerative medicine.

Regulatory pathways and approval

Navigating the regulatory pathways for SVF therapies poses a substantial barrier to market entry and clinical application (Table 2). The FDA’s guidelines for cell-based therapies necessitate comprehensive preclinical and clinical data demonstrating safety and efficacy. This includes detailed characterization of SVF products, quality control measures, and standardized manufacturing protocols[38]. The stringent requirements can delay the approval process and increase the financial burden on researchers and developers. Similarly, the EMA mandates that SVF therapies undergo rigorous evaluation before receiving marketing authorization within the European Union. This involves adherence to GMP, robust clinical trial data, and thorough safety assessments. The differences in regulatory frameworks between the FDA and EMA further complicate the global translation of SVF therapies, as developers must tailor their compliance strategies to meet diverse regulatory standards[45]. The regulatory landscape is further complicated by the rapid evolution of SVF therapies and the emergence of novel processing techniques. Regulatory agencies must continuously update their guidelines to accommodate advancements in cell therapy, necessitating ongoing dialogue between developers and regulators to ensure alignment and facilitate the approval process.

Table 2 Comparison of regulatory frameworks for stromal vascular fraction therapy.
Regulatory agency
Classification of SVF
Key requirements for approval
Pathway to market
FDA (United States)Biological product or medical deviceComprehensive preclinical and clinical data. GMP. Detailed characterization of SVFInvestigational New Drug application. Biologics license application or device approval
EMA (EU)Advanced therapy medicinal productRobust clinical trial data. Compliance with GMP. Safety and efficacy demonstrationMarketing authorization application. Scientific advice and protocol assistance
Health CanadaBiological drugEvidence of safety, efficacy, and quality. Adherence to GMPClinical trial application. New drug submission
TGA (Australia)Biological therapeuticDemonstrated safety and efficacy. Quality manufacturing processesTGA submission
PMDA (Japan)Regenerative medicine productExtensive clinical data. Compliance with good clinical practiceClinical trial notification. Marketing authorization
Safety and ethical concerns

Ensuring the safety of SVF therapies is paramount, given the potential risks associated with cell-based treatments. While studies such as those by Kwon et al[46] and Aletto et al[47] have reported minimal adverse effects following SVF administration, concerns remain regarding immune reactions, tumorigenicity, and the transmission of latent viruses when allogeneic usage is considered. The immunomodulatory properties of SVF cells, while beneficial to reduce inflammation, may also pose risks of unintended immune suppression or activation, necessitating careful monitoring and risk assessment.

Ethical considerations related to the sourcing of adipose tissue for SVF isolation also present significant challenges. The procurement of adipose tissue must adhere to ethical standards, ensuring informed consent and the protection of donor rights. Additionally, sourcing from vulnerable populations raises ethical concerns about exploitation and consent validity, necessitating stringent ethical guidelines and oversight mechanisms. The commercialization of SVF therapies also raises ethical questions regarding accessibility and equity. As SVF treatments are often expensive and not widely covered by insurance, there is a risk of creating disparities in access to these advanced therapies. Ensuring equitable access and preventing exploitation by unregulated clinics offering overpriced or unproven treatments is essential to maintain ethical standards in regenerative medicine[45]. Regulatory challenges in SVF therapy are depicted in Figure 2.

Figure 2
Figure 2 Regulatory challenges in stromal vascular fraction therapy. SVF: Stromal vascular fraction.
TECHNICAL AND LOGISTICAL CHALLENGES
SVF isolation and processing

The isolation and processing of SVF from adipose tissue involve several technical steps, each presenting its own set of challenges. Common methods include enzymatic digestion using collagenase and mechanical separation techniques such as centrifugation or filtration (Table 3)[44,48-51]. For example, Kim et al[15] utilized enzymatic digestion to isolate SVF cells, achieving a rapid and cost-effective preparation suitable for clinical applications. However, ensuring reproducibility and sterility across different clinical settings remains a significant hurdle. Variations in enzymatic digestion protocols, centrifugation speeds, and filtration techniques can lead to inconsistent cell yields and compositions, impacting therapeutic outcomes[52-54]. Additionally, maintaining sterility throughout the isolation process is critical to prevent contamination and ensure patient safety, necessitating stringent aseptic techniques and quality control measures. The variability in isolation methods complicates the standardization of SVF products, as different techniques may yield distinct cell populations with varying therapeutic potentials. This inconsistency hinders the ability to establish uniform treatment protocols and complicates the comparison of results across different studies and clinical trials.

Table 3 Comparison of stromal vascular fraction isolation methods.
Isolation method
Technique description
Advantages
Challenges
Enzymatic digestionUses enzymes like collagenase to break down adipose tissueHigh yield of viable cells. Effective separation of SVFPotential for enzymatic damage. Regulatory concerns with enzyme use
Mechanical separationUtilizes physical methods like centrifugation and filtrationMinimal enzymatic manipulation. Reduced regulatory barriersLower cell yield. Potential for contamination
Combination methodsCombines enzymatic and mechanical techniquesBalances cell yield and purity. Enhanced reproducibilityIncreased complexity. Higher cost
Automated systemsEmploys automated devices for consistent processingStandardization across batches. Improved sterilityHigh initial investment. Limited accessibility
Non-enzymatic techniquesUses methods like pressure-based separationAvoids use of enzymes. Potentially lower costVariable cell yield. Less efficient separation
Scalability and cost

Scaling up SVF processing for widespread clinical use presents both technical and financial challenges. The costs associated with SVF isolation, including enzymatic reagents, specialized equipment, and personnel training, can be substantial[55-57]. For instance, prospective studies by Mehling et al[58] and Aletto et al[47] have demonstrated the effectiveness of SVF therapies, yet the high costs of processing limit their accessibility and scalability. Emerging technologies aimed at reducing costs and simplifying procedures offer potential solutions. Innovations such as automated SVF isolation systems and cost-effective enzymatic reagents are being developed to streamline the processing workflow and minimize expenses. Additionally, advancements in microfluidics and bioreactor technologies hold promise to enhance the efficiency and scalability of SVF production, making it more feasible for large-scale clinical applications[59,60]. Despite these advancements, achieving cost-effective and scalable SVF processing requires ongoing research and investment. Addressing these challenges is crucial to make SVF therapies more accessible and economically viable, thereby facilitating their integration into mainstream clinical practice.

Quality control and storage

Ensuring the quality and viability of SVF cell products is essential to maintain therapeutic efficacy and patient safety. Quality control measures encompass various aspects, including cell viability assays, phenotypic characterization, and functional assessments. For example, the study by Perdomo-Pantoja et al[39] highlights the importance of robust quality control protocols in ensuring comparable fusion mass volume and radiographic fusion rates when SVF is combined with bone graft substitutes. Storage of SVF cells presents additional logistical challenges when culture of adipose-derived MSCs is contemplated[61,62]. Short-term storage typically involves cryopreservation techniques to maintain cell viability and functionality, while long-term storage requires optimized protocols to prevent cell degradation and ensure preservation of therapeutic potential[63-65]. Variability in storage conditions of SVF, such as temperature fluctuations and thawing protocols, can adversely affect cell quality, necessitating standardized storage procedures to maintain consistency[66-70]. Moreover, the integration of quality control and storage protocols within clinical workflows demands meticulous coordination and resource allocation. Implementing comprehensive quality assurance programs and investing in advanced storage facilities are imperative to ensure the reliability and consistency of SVF therapies. Technical and logistical challenges in SVF therapy are depicted in Figure 3.

Figure 3
Figure 3 Technical and logistical challenges in stromal vascular fraction therapy. SVF: Stromal vascular fraction.
CLINICAL AND TRANSLATIONAL CHALLENGES
Patient-specific factors

The efficacy and safety of SVF therapies are influenced by a myriad of patient-specific factors, including age, comorbidities, immune status, and overall health (Table 4). For instance, the study by Mehling et al[58] revealed that SVF cell therapy was more effective in patients with stage III arthritis compared to other stages, indicating that disease severity and patient condition significantly impact therapeutic outcomes. Age-related factors, such as the decline in stem cell function and regenerative capacity, can affect the efficacy of SVF therapies in older patients[71-74]. Comorbid conditions like diabetes and cardiovascular diseases may also alter the cellular environment, impacting SVF cell survival and integration.

Table 4 Patient-specific factors affecting stromal vascular fraction therapy.
Factor
Impact on SVF therapy efficacy
Considerations for clinical trials
AgeOlder patients may have reduced stem cell function and regenerative capacityStratify participants by age; analyze age-related efficacy differences
BMIObesity can affect adipose tissue quality and SVF cell compositionControl for BMI in study designs; assess cell quality based on BMI
ComorbiditiesConditions like diabetes and cardiovascular diseases may impair SVF integrationExclude or specifically include certain comorbidities; subgroup analyses
Immune statusImmunosuppressed patients may have altered SVF cell behaviorMonitor immune markers; consider immunomodulatory effects
GenderPotential differences in SVF cell composition and hormonal influencesEnsure balanced gender representation; analyze gender-specific outcomes
Medication useCertain medications may influence SVF cell viability and functionRecord and control for concurrent medication use; adjust protocols accordingly
Lifestyle factorsSmoking, alcohol use, and physical activity can affect tissue healingCollect detailed lifestyle data; adjust for confounding variables
Genetic factorsGenetic variations may influence SVF cell behavior and therapeutic responseConsider genetic screening; explore personalized approaches

Moreover, variations in immune status, whether due to immunosuppressive therapies or underlying autoimmune conditions, can influence the immunomodulatory effects of SVF cells, potentially leading to variable therapeutic responses[22]. These patient-specific factors necessitate tailored treatment approaches and personalized protocols to optimize SVF therapy outcomes. Additionally, these factors influence clinical study design, requiring stratification and careful patient selection to account for the diverse factors affecting efficacy and safety.

Customized SVF treatments account for patient-specific factors like age, health, comorbidities, and cellular regenerative potential. Elderly or chronically ill patients may have reduced cell viability, affecting therapy effectiveness. SVF volume is tailored to treatment goals, with larger volumes used for cartilage regeneration or joint function. Assessing prior treatment responses helps predict success, and in vitro manipulation of cells can enhance regenerative properties. Standardized protocols to harvest, process, and apply autologous SVF are essential for consistent, effective outcomes[1,5].

Outcome measures and efficacy assessment

Evaluating the efficacy of SVF therapies poses significant challenges due to the limitations of current outcome measures in clinical trials. Traditional metrics, such as pain reduction and functional improvement scores, provide valuable insights but may not fully capture the underlying biological changes induced by SVF treatments. For example, the WOMAC score used in Garza et al[20] provides a measure of pain and function in osteoarthritis patients, yet it does not directly assess the cellular or molecular mechanisms at play. To overcome these limitations, there is a need for the development of more comprehensive and objective biomarkers that can accurately reflect the therapeutic effects of SVF.

Biomarkers related to tissue regeneration, angiogenesis, and inflammation modulation can provide a more nuanced understanding of the impact of SVF[75-78]. Functional metrics, such as imaging-based assessments of tissue repair and biochemical markers of inflammation, can complement traditional outcome measures, offering a more holistic evaluation of therapeutic efficacy. Moreover, the integration of advanced analytical techniques, such as proteomics and genomics, can facilitate the identification of specific molecular signatures associated with successful SVF therapy outcomes. These improvements in efficacy assessment are crucial to establish robust evidence of the therapeutic benefits of SVF and to optimize treatment protocols based on objective data.

Interdisciplinary collaboration

The successful clinical translation of SVF therapies requires robust collaboration across multiple disciplines, including clinicians, researchers, regulatory experts, and industry stakeholders[79-82]. Interdisciplinary collaboration fosters the exchange of knowledge, facilitates the development of standardized protocols, and accelerates the translation of research findings into clinical practice. However, current gaps in communication and integration between different sectors hinder the clinical translation process. For instance, researchers may develop innovative SVF processing techniques without adequate consideration of regulatory requirements, leading to delays in approval and implementation. Similarly, clinicians may lack access to the latest research developments, limiting their ability to incorporate novel SVF therapies into patient care effectively. Enhancing interdisciplinary collaboration involves establishing platforms for regular communication, fostering joint research initiatives, and promoting the alignment of objectives between academic institutions and industry partners. Creating integrated networks that include regulatory agencies can also ensure that therapeutic developments follow regulatory standards, facilitating smoother clinical translation[83]. Moreover, fostering a culture of shared responsibility and mutual understanding among different stakeholders can bridge existing gaps, promoting a more cohesive approach to SVF therapy development and implementation. Addressing these collaborative challenges is essential to overcome the barriers to clinical translation and realize the full potential of SVF therapies in regenerative medicine. Clinical and translational challenges in SVF therapy are depicted in Figure 4.

Figure 4
Figure 4 Clinical and translational challenges in stromal vascular fraction therapy.
ETHICAL AND SOCIAL CONSIDERATIONS

The deployment of SVF therapies in clinical practice raises several ethical concerns that must be carefully addressed to ensure responsible and equitable application. One primary ethical issue is patient consent, particularly concerning the sourcing of adipose tissue. Ensuring that patients provide informed consent, fully understanding the procedures, potential risks, and expected outcomes, is fundamental to ethical clinical practice[84-86].

Commercialization of adipose-derived cells also presents ethical dilemmas. The high costs associated with SVF therapies can limit access to affluent populations, exacerbating existing healthcare disparities. These challenges necessitate stringent donor screening, regulatory adherence, and ethical business practices to balance profit motives with patient welfare[1]. Additionally, in vitro expansion of autologous SVF for personalized therapies requires careful ethical oversight, ensuring transparent patient consent and responsible clinical application to uphold high ethical standards in regenerative medicine[1]. Furthermore, the proliferation of unregulated clinics offering SVF treatments without sufficient evidence of efficacy and safety raises concerns about exploitation and patient safety. Such practices can erode public trust in regenerative medicine and undermine the ethical foundations of clinical research and application.

Accessibility is another critical ethical consideration. Ensuring that SVF therapies are accessible to diverse populations, regardless of socioeconomic status, is essential to promote equity in healthcare[87,88]. Efforts must be made to prevent the commodification of SVF therapies and to ensure that treatments are distributed based on medical need rather than financial capability. Ethical sourcing of adipose tissue, particularly from vulnerable populations, requires stringent oversight to prevent exploitation and ensure donor rights are respected. Establishing ethical guidelines and regulatory frameworks that prioritize patient welfare and equitable access are crucial to address these ethical implications.

Managing patient expectations is vital for the ethical and effective application of SVF therapies. Patients may have high expectations regarding the potential benefits of SVF treatments, influenced by media reports, anecdotal evidence, and marketing by clinics offering unproven therapies[89]. These expectations can lead to disappointment and erosion of trust if the outcomes do not meet patient anticipations. Educating patients about the current state of SVF research, including its potential benefits and limitations, is essential to foster realistic expectations. Clear communication regarding the experimental nature of SVF therapies, the uncertainties surrounding long-term efficacy and safety, and the variability in treatment outcomes can help patients make informed decisions[90,91]. Accurate information dissemination through healthcare providers, patient education materials, and public health campaigns can mitigate the risk of exaggerated claims and prevent patients from pursuing unregulated or ineffective treatments. Additionally, developing standardized informational resources and guidelines for clinicians can enhance the quality and consistency of patient education, ensuring that individuals receive reliable and evidence-based information about SVF therapies[92,93]. Addressing patient expectations and promoting education not only enhance the ethical application of SVF therapies but also contribute to better patient outcomes and satisfaction by aligning treatment goals with achievable results.

FUTURE DIRECTIONS AND POTENTIAL SOLUTIONS

Advancements in technology and scientific research offer promising avenues to address the challenges associated with SVF therapy. Emerging technologies such as CRISPR-based gene editing[94,95] and the development of advanced biomaterials[96,97] can enhance the efficacy and safety of SVF treatments (Table 5). For example, gene editing techniques could be employed to modify SVF cells to improve regenerative capabilities or reduce immunogenicity, thereby enhancing their therapeutic potential.

Table 5 Innovative approaches to overcome stromal vascular fraction therapy challenges.
Innovative approach
Description
Potential benefits
Current status
CRISPR-based gene editingModifying SVF cells to enhance regenerative capabilities and reduce immunogenicityImproved cell survival and integration. Enhanced therapeutic efficacyPreclinical research; limited clinical application
Advanced biomaterialsDevelopment of scaffolds and hydrogels to support SVF cell delivery and functionEnhanced cell retention and viability. Controlled release of growth factorsEmerging technologies; ongoing research
Single-cell sequencingAnalyzing individual SVF cells to understand cellular heterogeneity and mechanismsIdentification of key therapeutic cell populations. Personalized treatment strategiesActive research; integration into SVF characterization
Automated isolation systemsUtilizing automated devices for consistent and sterile SVF processingStandardized cell yields. Reduced contamination riskCommercially available; increasing adoption
Bioreactor cultivationCulturing progenitor cells in SVF in bioreactors to scale-up production and enhance cell propertiesLarge-scale production. Enhanced cell functionalityPilot studies; early-stage commercialization
Potency assays developmentCreating assays to evaluate SVF cell efficacy and functionality before clinical useImproved quality control. Predictive markers for therapeutic outcomesOngoing development; essential for regulatory approval

Biomaterials, such as scaffolds and hydrogels, can provide supportive environments for SVF cells, promoting their survival, integration, and functionality within target tissues[98-100]. These materials can also be engineered to deliver growth factors and other bioactive molecules in a controlled manner, facilitating sustained therapeutic effects and enhancing tissue regeneration. Advancements in single-cell sequencing and proteomics can deepen our understanding of the cellular composition and mechanistic pathways of SVF, enabling the identification of specific cell populations and factors that drive therapeutic outcomes[101]. This knowledge can inform the development of more targeted and effective SVF therapies, optimizing their clinical application.

Regulatory science must evolve to accommodate the unique challenges posed by SVF therapies, striking a balance between fostering innovation and ensuring patient safety. Potential changes in regulatory policies could include the development of specific guidelines for SVF isolation and characterization, streamlined approval processes for minimally manipulated cell products, and the establishment of standardized protocols for SVF manufacturing and quality control[102,103]. Collaborative efforts between regulatory bodies, researchers, and industry stakeholders can facilitate the creation of harmonized regulatory frameworks that support the safe and efficient translation of SVF therapies into clinical practice. Additionally, adopting adaptive regulatory approaches that allow for iterative modifications based on emerging evidence can enhance the responsiveness of regulatory systems to scientific advancements. Implementing national registries and databases to track SVF therapy outcomes can also inform regulatory decisions, providing real-world evidence of safety and efficacy[104]. These registries can support post-market surveillance, enabling the identification and mitigation of potential risks and the continuous improvement of SVF therapy protocols[105].

A structured roadmap is essential to advance SVF therapies from research to widespread clinical application, progressing through key stages from preclinical research to post-market surveillance (Table 6). In the initial preclinical phase, comprehensive studies are necessary to clarify mechanisms of action, refine cell processing techniques, and assess both the safety and efficacy of SVF therapies within appropriate animal models. This foundational research should emphasize understanding the interactions between SVF cells and target tissues, with a focus on identifying biomarkers predictive of therapeutic outcomes. Once these initial studies have established a solid foundation, the next stage involves meticulous clinical trial design. Designing trials with robust methodologies, such as adequate sample sizes, randomized controlled structures, and standardized outcome measures, is essential to produce reliable evidence of the therapeutic potential of SVF.

Table 6 Roadmap for clinical translation of stromal vascular fraction therapy.
Stage
Key activities
Goals
Milestones
Preclinical researchElucidate mechanisms of action. Optimize SVF isolation and processing techniques. Conduct animal studiesUnderstand SVF biology. Ensure safety and efficacyIdentification of key therapeutic pathways. Optimized protocols
Clinical trial designDevelop robust trial protocols. Define inclusion/exclusion criteria. Standardize outcome measuresGenerate reliable efficacy and safety dataSuccessful completion of Phase I/II trials
Regulatory approvalEngage with regulatory bodies. Compile comprehensive data packages. Ensure GMP complianceAchieve regulatory compliance. Facilitate market entrySubmission of IND/BLA or MAA Approval from FDA/EMA
Manufacturing scale-upImplement GMP-compliant manufacturing. Establish quality control systems. Scale production capacityEnsure consistent and high-quality SVF productsGMP certification. Scalable production lines
Market entryLaunch approved SVF therapies. Develop distribution channels. Train clinical practitionersIntroduce SVF therapies to the market. Ensure accessibilitySuccessful product launch. Initial clinical adoption
Post-market surveillanceMonitor long-term safety and efficacy. Collect real-world evidence. Implement continuous improvementEnsure ongoing patient safety. Refine treatment protocolsEstablishment of national registries. Regular safety reports

Additionally, clinical trials should incorporate patient stratification based on specific variables like disease severity, age, and comorbidities to account for differences in treatment responses and enhance the relevance of results. Early engagement with regulatory authorities is a critical component of the development process, as it facilitates alignment with regulatory expectations, potentially expediting the approval timeline. Ensuring compliance with GMP and providing rigorous, comprehensive data on both safety and efficacy are essential to obtain regulatory approval. Following successful regulatory submission, the final phase centers on post-market surveillance, which establishes the mechanisms for continuous monitoring of SVF therapy outcomes in real-world clinical settings. This ongoing surveillance plays a pivotal role in identifying rare adverse events, evaluating the durability of therapeutic benefits, and supporting iterative improvements to SVF treatment protocols based on emerging clinical insights.

Advancing the clinical translation of SVF therapies requires targeted research efforts across several critical domains (Tables 7 and 8). Mechanistic studies are essential to investigate the specific cellular and molecular mechanisms through which SVF exerts its therapeutic effects, as understanding the interactions between SVF cells and the host tissue microenvironment is fundamental to optimizing therapeutic strategies[106-108]. Additionally, conducting large-scale, multi-center clinical trials can generate robust evidence of the efficacy and safety of SVF across diverse patient populations. Such trials should focus on addressing current limitations, including small sample sizes and brief follow-up periods, to improve the generalizability and reliability of the findings.

Table 7 Suggested research directions for stromal vascular fraction therapy.
Research area
Description
Objectives
Expected outcomes
Mechanistic studiesInvestigate cellular and molecular pathways through which SVF exerts therapeutic effectsUnderstand SVF interactions with host tissuesIdentification of key therapeutic targets
Large-scale clinical trialsConduct multi-center, randomized controlled trials with larger and diverse patient populationsValidate efficacy and safety across different demographicsRobust evidence for regulatory approval and clinical guidelines
Biomarker developmentIdentify and validate biomarkers that predict therapeutic outcomes and monitor SVF efficacy in addition to the identified surface markersEnhance personalized treatment approachesImproved patient stratification and treatment optimization
Standardization of protocolsDevelop and validate standardized methods for SVF isolation, processing, and administrationEnsure reproducibility and consistency in SVF therapiesHarmonized clinical protocols and regulatory compliance
Personalized medicine approachesTailor SVF therapies based on individual patient characteristics and disease profilesOptimize therapeutic efficacy and minimize adverse effectsPersonalized treatment plans enhancing patient outcomes
Real-world evidence generationCollect and analyze data from SVF therapies in routine clinical settings to complement clinical trial dataUnderstand long-term effects and practical applicationsComprehensive understanding of SVF therapy performance in diverse settings
Integration with other therapiesExplore combination therapies involving SVF and other regenerative or pharmacological treatmentsEnhance overall therapeutic efficacySynergistic treatment strategies improving patient outcomes
Table 8 Ethical and social considerations in stromal vascular fraction therapy.
Ethical/social aspect
Description
Implications
Strategies to address
Informed consentEnsuring patients are fully aware of the procedures, risks, and benefits of SVF therapyEthical obligation to respect patient autonomyComprehensive consent forms. Clear communication of risks and benefits
Equitable accessEnsuring SVF therapies are accessible to all segments of the population regardless of socioeconomic statusPreventing healthcare disparitiesImplementing insurance coverage. Subsidized treatment options
Commercialization and exploitationAvoiding the commercialization of SVF therapies that exploit vulnerable patientsProtecting patients from unproven and overpriced treatmentsRegulatory oversight. Strict marketing guidelines
Donor rights and ethical sourcingEnsuring ethical procurement of adipose tissue, particularly from vulnerable populationsRespecting donor autonomy and preventing exploitationEthical sourcing protocols. Regulatory compliance
Public perception and trustMaintaining public trust in regenerative medicine amidst unregulated clinic practicesBuilding and sustaining confidence in SVF therapiesPublic education campaigns. Transparent clinical practices
Managing patient expectationsAligning patient expectations with realistic therapeutic outcomesReducing disappointment and maintaining trustClear communication of therapy limitations and realistic outcomes
Regulatory complianceAdhering to ethical and legal standards in SVF therapy development and applicationEnsuring ethical integrity and patient safetyOngoing regulatory training. Implementing best practice guidelines

Generating real-world evidence is another key component, as analyzing outcomes from SVF therapies in practical, real-world settings can offer valuable insights into the long-term effects and applicability of SVF treatments. This type of evidence can inform clinical guidelines, influence regulatory decisions, and shape patient care practices. Furthermore, the standardization of protocols for SVF isolation, processing, and administration is crucial to achieve consistent therapeutic outcomes and ensure regulatory compliance. Collaborative efforts to establish consensus guidelines can promote uniformity and reproducibility across studies and clinical applications. Personalized medicine approaches are also worth exploring, as tailoring SVF treatments based on individual patient characteristics and disease profiles can enhance therapeutic efficacy and reduce adverse effects. By focusing on these specific research areas, the field can overcome existing challenges and unlock the full potential of SVF therapies in regenerative medicine.

CONCLUSION

SVF shows immense promise in regenerative medicine, offering a versatile therapeutic option across various medical fields. Its diverse cellular composition, including ADSCs and immune cells, supports tissue regeneration, immunomodulation, and anti-inflammatory effects, beneficial for conditions like osteoarthritis and chronic wounds. However, challenges span biological, regulatory, technical, clinical, ethical, and social areas. Biologically, the heterogeneity and mechanisms of SVF require further research for standardization. Regulatory ambiguities necessitate clearer guidelines, while technical hurdles demand scalable, sterile, and cost-effective isolation processes. Clinically, patient-specific factors and outcome measures need tailored approaches, and ethical issues such as equitable access and informed consent are critical. Advancements in technology, regulatory science, and interdisciplinary collaboration are vital to fully realize the clinical potential of SVF.

Footnotes

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

Peer-review model: Single blind

Specialty type: Cell and tissue engineering

Country of origin: India

Peer-review report’s classification

Scientific Quality: Grade B, Grade B, Grade C, Grade C

Novelty: Grade B, Grade B, Grade C, Grade C

Creativity or Innovation: Grade B, Grade B, Grade B, Grade B

Scientific Significance: Grade B, Grade B, Grade B, Grade B

P-Reviewer: Apolonio EVP; Luo HC; Yang L S-Editor: Wang JJ L-Editor: Filipodia P-Editor: Zhang XD

References
1.  Jeyaraman N, Shrivastava S, Ravi VR, Nallakumarasamy A, Pundkar A, Jeyaraman M. Understanding and controlling the variables for stromal vascular fraction therapy. World J Stem Cells. 2024;16:784-798.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 6]  [Reference Citation Analysis (2)]
2.  Biniazan F, Stoian A, Haykal S. Adipose-Derived Stem Cells: Angiogenetic Potential and Utility in Tissue Engineering. Int J Mol Sci. 2024;25:2356.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 16]  [Reference Citation Analysis (0)]
3.  Kilinc MO, Santidrian A, Minev I, Toth R, Draganov D, Nguyen D, Lander E, Berman M, Minev B, Szalay AA. The ratio of ADSCs to HSC-progenitors in adipose tissue derived SVF may provide the key to predict the outcome of stem-cell therapy. Clin Transl Med. 2018;7:5.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 27]  [Cited by in RCA: 34]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
4.  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.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 28]  [Cited by in RCA: 25]  [Article Influence: 6.3]  [Reference Citation Analysis (2)]
5.  Goncharov EN, Koval OA, Igorevich EI, Encarnacion Ramirez MJ, Nurmukhametov R, Valentinovich KK, Montemurro N. Analyzing the Clinical Potential of Stromal Vascular Fraction: A Comprehensive Literature Review. Medicina (Kaunas). 2024;60:221.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 11]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
6.  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.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 2]  [Cited by in RCA: 6]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
7.  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.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
8.  Gareev I, Beylerli O, Ilyasova T, Ahmad A, Shi H, Chekhonin V. Therapeutic application of adipose-derived stromal vascular fraction in myocardial infarction. iScience. 2024;27:109791.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
9.  Yan W, Xia Y, Zhao H, Xu X, Ma X, Tao L. Stem cell-based therapy in cardiac repair after myocardial infarction: Promise, challenges, and future directions. J Mol Cell Cardiol. 2024;188:1-14.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 27]  [Reference Citation Analysis (0)]
10.  Mantovani GP, Marra C, De Maria F, Pinto V, De Santis G. Adipose-derived stromal vascular fraction (SVF) for the treatment of androgenic alopecia (AGA): a systematic review. Acta Biomed. 2023;94:e2023236.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
11.  Zocchi ML, Prantl L, Oliinyk D, Knoedler L, Siegmund A, Ahmad N, Duscher D, Larcher L, Raposio E, Pagani A. Potential benefits of adipose–derived SVF and MSCs to regenerate damaged tissues from alloplastic synthetic materials. Eur J Plast Surg. 2024;47:48.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
12.  Malekzadeh H, Tirmizi Z, Arellano JA, Egro FM, Ejaz A. Application of Adipose-Tissue Derived Products for Burn Wound Healing. Pharmaceuticals (Basel). 2023;16:1302.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 9]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
13.  Cai W, Yu LD, Tang X, Shen G. The Stromal Vascular Fraction Improves Maintenance of the Fat Graft Volume: A Systematic Review. Ann Plast Surg. 2018;81:367-371.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 15]  [Cited by in RCA: 24]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
14.  Onoi Y, Matsumoto T, Sobajima S, Tsubosaka M, Hayashi S, Matsushita T, Iwaguro H, Kuroda R. Clinical use of autologous adipose-derived stromal vascular fraction cell injections for hip osteoarthritis. Regen Ther. 2023;24:94-102.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
15.  Kim YS, Oh SM, Suh DS, Tak DH, Kwon YB, Koh YG. Arthroscopic Implantation of Adipose-Derived Stromal Vascular Fraction Improves Cartilage Regeneration and Pain Relief in Patients With Knee Osteoarthritis. Arthrosc Sports Med Rehabil. 2023;5:e707-e716.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
16.  Zhang S, Chen J, Zhang H, Hu S, Tong P, Shen J. Multiple intra-articular injections of autologous stromal vascular fractions for the treatment of multicompartmental osteoarthritis in both the tibiofemoral and patellofemoral joint: a single-blind randomized controlled study. Postgrad Med J. 2024;100:399-406.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
17.  Wu V, Klein-Nulend J, Bravenboer N, Ten Bruggenkate CM, Helder MN, Schulten EAJM. Long-Term Safety of Bone Regeneration Using Autologous Stromal Vascular Fraction and Calcium Phosphate Ceramics: A 10-Year Prospective Cohort Study. Stem Cells Transl Med. 2023;12:617-630.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
18.  Zinger G, Gronovich Y, Lotan AM, Sharon-Gabbay R. Pilot Study for Isolation of Stromal Vascular Fraction with Collagenase Using an Automated Processing System. Int J Mol Sci. 2024;25:7148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
19.  Ramakrishnan VM, Boyd NL. The Adipose Stromal Vascular Fraction as a Complex Cellular Source for Tissue Engineering Applications. Tissue Eng Part B Rev. 2018;24:289-299.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 114]  [Cited by in RCA: 103]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
20.  Garza JR, Campbell RE, Tjoumakaris FP, Freedman KB, Miller LS, Santa Maria D, Tucker BS. Clinical Efficacy of Intra-articular Mesenchymal Stromal Cells for the Treatment of Knee Osteoarthritis: A Double-Blinded Prospective Randomized Controlled Clinical Trial. Am J Sports Med. 2020;48:588-598.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 74]  [Cited by in RCA: 132]  [Article Influence: 26.4]  [Reference Citation Analysis (0)]
21.  Jeon HJ, Choi DH, Lee JH, Lee JS, Lee J, Park HY, Yang JD. A Prospective Study of the Efficacy of Cell-Assisted Lipotransfer with Stromal Vascular Fraction to Correct Contour Deformities of the Autologous Reconstructed Breast. Aesthetic Plast Surg. 2021;45:853-863.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
22.  Dykstra JA, Facile T, Patrick RJ, Francis KR, Milanovich S, Weimer JM, Kota DJ. Concise Review: Fat and Furious: Harnessing the Full Potential of Adipose-Derived Stromal Vascular Fraction. Stem Cells Transl Med. 2017;6:1096-1108.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 65]  [Cited by in RCA: 77]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
23.  Kostecka A, Kalamon N, Skoniecka A, Koczkowska M, Skowron PM, Piotrowski A, Pikuła M. Adipose-derived mesenchymal stromal cells in clinical trials: Insights from single-cell studies. Life Sci. 2024;351:122761.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
24.  Roshdy OH, Abdallah WI, Farid CI, Mehanna RA, Bayoumi NH, Ismail AI. Stromal vascular fraction improves the durability of autologous fat temple augmentation-A split-face randomized study using ultrasound biomicroscopy. J Plast Reconstr Aesthet Surg. 2022;75:1870-1877.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 10]  [Cited by in RCA: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
25.  Cremona M, Rusconi G, Ferrario A, Mariotta L, Gola M, Soldati G. Processing Adipose Tissue Samples in a GMP Environment Standardizes the Use of SVF in Cell Therapy Treatments: Data on 302 Patients. Biomedicines. 2023;11:2533.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
26.  Dykstra JA, Blue ED, Negrão de Assis PL, Weimer JM, Kota DJ. Human adipose-derived stromal vascular fraction: characterization, safety and therapeutic potential in an experimental mouse model of articular injury. J Stem Cells Regen Med. 2020;16:16-25.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
27.  Krawczenko A, Klimczak A. Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells and Their Contribution to Angiogenic Processes in Tissue Regeneration. Int J Mol Sci. 2022;23:2425.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 12]  [Cited by in RCA: 49]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
28.  Cai Y, Li J, Jia C, He Y, Deng C. Therapeutic applications of adipose cell-free derivatives: a review. Stem Cell Res Ther. 2020;11:312.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 48]  [Cited by in RCA: 130]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
29.  Jia Q, Zhao H, Wang Y, Cen Y, Zhang Z. Mechanisms and applications of adipose-derived stem cell-extracellular vesicles in the inflammation of wound healing. Front Immunol. 2023;14:1214757.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 23]  [Reference Citation Analysis (0)]
30.  Cai Y, Zhang F, Feng J, Wu B, Li H, Xiao S, Lu F, Wei Z, Deng C. Long-term follow-up and exploration of the mechanism of stromal vascular fraction gel in chronic wounds. Stem Cell Res Ther. 2023;14:163.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
31.  Agaverdiev M, Shamsov B, Mirzoev S, Vardikyan A, Ramirez ME, Nurmukhametov R, Beilerli A, Zhang B, Gareev I, Pavlov V. MiRNA regulated therapeutic potential of the stromal vascular fraction: Current clinical applications - A systematic review. Noncoding RNA Res. 2023;8:146-154.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
32.  Al-Kharboosh R, Perera JJ, Bechtle A, Bu G, Quinones-Hinojosa A. Emerging point-of-care autologous cellular therapy using adipose-derived stromal vascular fraction for neurodegenerative diseases. Clin Transl Med. 2022;12:e1093.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
33.  Seo Y, Shin TH, Kim HS. Current Strategies to Enhance Adipose Stem Cell Function: An Update. Int J Mol Sci. 2019;20:3827.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 98]  [Cited by in RCA: 104]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
34.  Bertozzi N, Simonacci F, Grieco MP, Grignaffini E, Raposio E. The biological and clinical basis for the use of adipose-derived stem cells in the field of wound healing. Ann Med Surg (Lond). 2017;20:41-48.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 64]  [Cited by in RCA: 68]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
35.  François P, Giraudo L, Veran J, Bertrand B, Dumoulin C, Aboudou H, Grimaud F, Vogtensperger M, Velier M, Arnaud L, Lyonnet L, Simoncini S, Guillet B, Dignat-George F, Magalon J, Sabatier F. Development and Validation of a Fully GMP-Compliant Process for Manufacturing Stromal Vascular Fraction: A Cost-Effective Alternative to Automated Methods. Cells. 2020;9:2158.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
36.  Rodriguez J, Pratta AS, Abbassi N, Fabre H, Rodriguez F, Debard C, Adobati J, Boucher F, Mallein-Gerin F, Auxenfans C, Damour O, Mojallal A. Evaluation of Three Devices for the Isolation of the Stromal Vascular Fraction from Adipose Tissue and for ASC Culture: A Comparative Study. Stem Cells Int. 2017;2017:9289213.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 30]  [Cited by in RCA: 31]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
37.  Marks PW. Clear Evidence of Safety and Efficacy Is Needed for Stromal Vascular Fraction Products: Commentary on "Arguments for a Different Regulatory Categorization and Framework for Stromal Vascular Fraction". Stem Cells Dev. 2020;29:263-265.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
38.  El-Kadiry AE, Rafei M, Shammaa R. Cell Therapy: Types, Regulation, and Clinical Benefits. Front Med (Lausanne). 2021;8:756029.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 29]  [Cited by in RCA: 114]  [Article Influence: 28.5]  [Reference Citation Analysis (0)]
39.  Perdomo-Pantoja A, Holmes C, Cottrill E, Rindone AN, Ishida W, Taylor M, Tomberlin C, Lo SL, Grayson WL, Witham TF. Comparison of Freshly Isolated Adipose Tissue-derived Stromal Vascular Fraction and Bone Marrow Cells in a Posterolateral Lumbar Spinal Fusion Model. Spine (Phila Pa 1976). 2021;46:631-637.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
40.  Choi JS, Chae DS, Ryu HA, Kim SW. Transplantation of human adipose tissue derived-SVF enhance liver function through high anti-inflammatory property. Biochim Biophys Acta Mol Cell Biol Lipids. 2019;1864:158526.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 7]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
41.  Muthu S, Jeyaraman M, Kotner MB, Jeyaraman N, Rajendran RL, Sharma S, Khanna M, Rajendran SNS, Oh JM, Gangadaran P, Ahn BC. Evolution of Mesenchymal Stem Cell Therapy as an Advanced Therapeutic Medicinal Product (ATMP)-An Indian Perspective. Bioengineering (Basel). 2022;9:111.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 17]  [Cited by in RCA: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
42.  Lee SJ, Lee CR, Kim KJ, Ryu YH, Kim E, Han YN, Moon SH, Rhie JW. Optimal Condition of Isolation from an Adipose Tissue-Derived Stromal Vascular Fraction for the Development of Automated Systems. Tissue Eng Regen Med. 2020;17:203-208.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
43.  Sananta P, I Gede Made Oka R, Suryanto Dradjat PR, Suroto H, Mustamsir E, Kalsum U, Andarini S. Adipose-derived stromal vascular fraction prevent bone bridge formation on growth plate injury in rat (in vivo studies) an experimental research. Ann Med Surg (Lond). 2020;60:211-217.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 4]  [Cited by in RCA: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
44.  Oberbauer E, Steffenhagen C, Wurzer C, Gabriel C, Redl H, Wolbank S. Enzymatic and non-enzymatic isolation systems for adipose tissue-derived cells: current state of the art. Cell Regen. 2015;4:7.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 85]  [Cited by in RCA: 108]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
45.  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.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
46.  Kwon H, Lee S, Kim J, Song SH. Efficacy and safety of stromal vascular fraction on scar revision surgery: a prospective study. J Dermatolog Treat. 2023;34:2171260.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
47.  Aletto C, Giordano L, Quaranta M, Zara A, Notarfrancesco D, Maffulli N. Short-term results of intra-articular injections of stromal vascular fraction for early knee osteoarthritis. J Orthop Surg Res. 2022;17:310.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 13]  [Reference Citation Analysis (0)]
48.  Mundluru VK, Naidu MJ, Mundluru RT, Jeyaraman N, Muthu S, Ramasubramanian S, Jeyaraman M. Non-enzymatic methods for isolation of stromal vascular fraction and adipose-derived stem cells: A systematic review. World J Methodol. 2024;14:94562.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (6)]
49.  Senesi L, De Francesco F, Farinelli L, Manzotti S, Gagliardi G, Papalia GF, Riccio M, Gigante A. Mechanical and Enzymatic Procedures to Isolate the Stromal Vascular Fraction From Adipose Tissue: Preliminary Results. Front Cell Dev Biol. 2019;7:88.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 54]  [Cited by in RCA: 48]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
50.  You X, Gao J, Yao Y. Advanced methods to mechanically isolate stromal vascular fraction: A concise review. Regen Ther. 2024;27:120-125.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
51.  Yaylacı S, Kaçaroğlu D, Hürkal Ö, Ulaşlı AM. An enzyme-free technique enables the isolation of a large number of adipose-derived stem cells at the bedside. Sci Rep. 2023;13:8005.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
52.  Aronowitz JA, Lockhart RA, Hakakian CS. Mechanical versus enzymatic isolation of stromal vascular fraction cells from adipose tissue. Springerplus. 2015;4:713.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 148]  [Cited by in RCA: 164]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
53.  Chen W, He Z, Li S, Wu Z, Tan J, Yang W, Li G, Pan X, Liu Y, Lyu FJ, Li W. The Effect of Tissue Stromal Vascular Fraction as Compared to Cellular Stromal Vascular Fraction to Treat Anal Sphincter Incontinence. Bioengineering (Basel). 2022;10:32.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
54.  Gentile P, Calabrese C, De Angelis B, Pizzicannella J, Kothari A, Garcovich S. Impact of the Different Preparation Methods to Obtain Human Adipose-Derived Stromal Vascular Fraction Cells (AD-SVFs) and Human Adipose-Derived Mesenchymal Stem Cells (AD-MSCs): Enzymatic Digestion Versus Mechanical Centrifugation. Int J Mol Sci. 2019;20:5471.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 95]  [Cited by in RCA: 86]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
55.  Andia I, Maffulli N, Burgos-Alonso N. Stromal vascular fraction technologies and clinical applications. Expert Opin Biol Ther. 2019;19:1289-1305.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 38]  [Cited by in RCA: 75]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
56.  van Dongen JA, Tuin AJ, Spiekman M, Jansma J, van der Lei B, Harmsen MC. Comparison of intraoperative procedures for isolation of clinical grade stromal vascular fraction for regenerative purposes: a systematic review. J Tissue Eng Regen Med. 2018;12:e261-e274.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 52]  [Cited by in RCA: 63]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
57.  van Dongen JA, Stevens HP, Parvizi M, van der Lei B, Harmsen MC. The fractionation of adipose tissue procedure to obtain stromal vascular fractions for regenerative purposes. Wound Repair Regen. 2016;24:994-1003.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 42]  [Cited by in RCA: 53]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
58.  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.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
59.  Maged A, Abdelbaset R, Mahmoud AA, Elkasabgy NA. Merits and advances of microfluidics in the pharmaceutical field: design technologies and future prospects. Drug Deliv. 2022;29:1549-1570.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 19]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
60.  Ryu H, Jeon TJ, Kim SM. Editorial Perspective: Advancements in Microfluidics and Biochip Technologies. Micromachines (Basel). 2025;16:77.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
61.  Jović D, Preradović L, Jović F, Kremenović M, Lukić D, Antonić M, Unčanin N, Jović M. Optimizing adipose-derived stromal vascular fraction storage: Temperature and time impact on cell viability in regenerative medicine. Medicine (Baltimore). 2024;103:e39859.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
62.  Dewi DA, Chouw A, Sartika CR, Facicilia G, Dirgantara Y, Pratiwi ED, Utami ER, Riswandani J, Wulandari D. Comparison of viability and proliferation potency of stromal vascular fraction stored at different temperature. Cytotherapy. 2019;21:S86.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
63.  Shaik S, Wu X, Gimble J, Devireddy R. Effects of Decade Long Freezing Storage on Adipose Derived Stem Cells Functionality. Sci Rep. 2018;8:8162.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 35]  [Cited by in RCA: 37]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
64.  Svalgaard JD, Juul S, Vester-Glovinski PV, Haastrup EK, Ballesteros OR, Lynggaard CD, Jensen AK, Fischer-Nielsen A, Herly M, Munthe-Fog L. Lipoaspirate Storage Time and Temperature: Effects on Stromal Vascular Fraction Quality and Cell Composition. Cells Tissues Organs. 2020;209:54-63.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6]  [Cited by in RCA: 15]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
65.  Tao Y, Zhao ZN, Xiang XJ, Liang ZX, Zhao Y. SVF-GEL Cryopreserved for Different Times Exhibits Varied Preservation and Regeneration Potential After Transplantation in a Mouse Model. Aesthetic Plast Surg. 2023;47:842-851.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
66.  Wu YD, Li M, Liao X, Li SH, Yan JX, Fan L, She WL, Song JX, Liu HW. Effects of storage culture media, temperature and duration on human adiposederived stem cell viability for clinical use. Mol Med Rep. 2019;19:2189-2201.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 12]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
67.  Gonda K, Shigeura T, Sato T, Matsumoto D, Suga H, Inoue K, Aoi N, Kato H, Sato K, Murase S, Koshima I, Yoshimura K. Preserved proliferative capacity and multipotency of human adipose-derived stem cells after long-term cryopreservation. Plast Reconstr Surg. 2008;121:401-410.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 91]  [Cited by in RCA: 97]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
68.  González-Fernández ML, Pérez-Castrillo S, Ordás-Fernández P, López-González ME, Colaço B, Villar-Suárez V. Study on viability and chondrogenic differentiation of cryopreserved adipose tissue-derived mesenchymal stromal cells for future use in regenerative medicine. Cryobiology. 2015;71:256-263.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 17]  [Cited by in RCA: 18]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
69.  Matsumoto D, Shigeura T, Sato K, Inoue K, Suga H, Kato H, Aoi N, Murase S, Gonda K, Yoshimura K. Influences of preservation at various temperatures on liposuction aspirates. Plast Reconstr Surg. 2007;120:1510-1517.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 51]  [Cited by in RCA: 49]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
70.  Zhang F, Ren H, Shao X, Zhuang C, Chen Y, Qi N. Preservation media, durations and cell concentrations of short-term storage affect key features of human adipose-derived mesenchymal stem cells for therapeutic application. PeerJ. 2017;5:e3301.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 11]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
71.  Rowe G, Heng DS, Beare JE, Hodges NA, Tracy EP, Murfee WL, LeBlanc AJ. Stromal Vascular Fraction Reverses the Age-Related Impairment in Revascularization following Injury. J Vasc Res. 2022;59:343-357.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
72.  Choudhery MS, Badowski M, Muise A, Pierce J, Harris DT. Donor age negatively impacts adipose tissue-derived mesenchymal stem cell expansion and differentiation. J Transl Med. 2014;12:8.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 408]  [Cited by in RCA: 374]  [Article Influence: 34.0]  [Reference Citation Analysis (0)]
73.  Ahmed AS, Sheng MH, Wasnik S, Baylink DJ, Lau KW. Effect of aging on stem cells. World J Exp Med. 2017;7:1-10.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 100]  [Cited by in RCA: 124]  [Article Influence: 15.5]  [Reference Citation Analysis (3)]
74.  Dos-Anjos Vilaboa S, Navarro-Palou M, Llull R. Age influence on stromal vascular fraction cell yield obtained from human lipoaspirates. Cytotherapy. 2014;16:1092-1097.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 51]  [Cited by in RCA: 64]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
75.  Szűcs D, Monostori T, Miklós V, Páhi ZG, Póliska S, Kemény L, Veréb Z. Licensing effects of inflammatory factors and TLR ligands on the regenerative capacity of adipose-derived mesenchymal stem cells. Front Cell Dev Biol. 2024;12:1367242.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
76.  Mitchell JB, McIntosh K, Zvonic S, Garrett S, Floyd ZE, Kloster A, Di Halvorsen Y, Storms RW, Goh B, Kilroy G, Wu X, Gimble JM. Immunophenotype of human adipose-derived cells: temporal changes in stromal-associated and stem cell-associated markers. Stem Cells. 2006;24:376-385.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 822]  [Cited by in RCA: 1050]  [Article Influence: 52.5]  [Reference Citation Analysis (0)]
77.  Debnath T, Chelluri LK. Standardization and quality assessment for clinical grade mesenchymal stem cells from human adipose tissue. Hematol Transfus Cell Ther. 2019;41:7-16.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 29]  [Cited by in RCA: 37]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
78.  Haack-Sørensen M, Follin B, Juhl M, Brorsen SK, Søndergaard RH, Kastrup J, Ekblond A. Culture expansion of adipose derived stromal cells. A closed automated Quantum Cell Expansion System compared with manual flask-based culture. J Transl Med. 2016;14:319.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 37]  [Cited by in RCA: 48]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
79.  Zocchi ML, Vindigni V, Pagani A, Pirro O, Conti G, Sbarbati A, Bassetto F. Regulatory, ethical, and technical considerations on regenerative technologies and adipose-derived mesenchymal stem cells. Eur J Plast Surg. 2019;42:531-548.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 15]  [Cited by in RCA: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
80.  François P, Rusconi G, Arnaud L, Mariotta L, Giraudo L, Minonzio G, Veran J, Bertrand B, Dumoulin C, Grimaud F, Lyonnet L, Casanova D, Giverne C, Cras A, Magalon G, Dignat-George F, Sabatier F, Magalon J, Soldati G. Inter-center comparison of good manufacturing practices-compliant stromal vascular fraction and proposal for release acceptance criteria: a review of 364 productions. Stem Cell Res Ther. 2021;12:373.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
81.  Bensemmane L, Milliat F, Treton X, Linard C. Systemically delivered adipose stromal vascular fraction mitigates radiation-induced gastrointestinal syndrome by immunomodulating the inflammatory response through a CD11b(+) cell-dependent mechanism. Stem Cell Res Ther. 2023;14:325.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 4]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
82.  Silva KR, Côrtes I, Liechocki S, Carneiro JR, Souza AA, Borojevic R, Maya-Monteiro CM, Baptista LS. Characterization of stromal vascular fraction and adipose stem cells from subcutaneous, preperitoneal and visceral morbidly obese human adipose tissue depots. PLoS One. 2017;12:e0174115.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 35]  [Cited by in RCA: 58]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
83.  Lopez-Navas L, Torrents S, Sánchez-Pernaute R, Vives J. Compliance in Non-Clinical Development of Cell-, Gene-, and Tissue-Based Medicines: Good Practice for Better Therapies. Stem Cells Transl Med. 2022;11:805-813.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
84.  West CC, Murray IR, González ZN, Hindle P, Hay DC, Stewart KJ, Péault B. Ethical, legal and practical issues of establishing an adipose stem cell bank for research. J Plast Reconstr Aesthet Surg. 2014;67:745-751.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 10]  [Cited by in RCA: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
85.  Grizzle WE. Issues in the Use of Human Tissues to Support Precision Medicine. J Health Care Poor Underserved. 2019;30:66-78.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 7]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
86.  Pietrzykowski T, Smilowska K. The reality of informed consent: empirical studies on patient comprehension-systematic review. Trials. 2021;22:57.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 14]  [Cited by in RCA: 96]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
87.  Williams JS, Walker RJ, Egede LE. Achieving Equity in an Evolving Healthcare System: Opportunities and Challenges. Am J Med Sci. 2016;351:33-43.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 44]  [Cited by in RCA: 93]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
88.  Stanford FC. The Importance of Diversity and Inclusion in the Healthcare Workforce. J Natl Med Assoc. 2020;112:247-249.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 105]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
89.  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.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
90.  Sharkiya SH. Quality communication can improve patient-centred health outcomes among older patients: a rapid review. BMC Health Serv Res. 2023;23:886.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 81]  [Reference Citation Analysis (0)]
91.  Simpkin AL, Armstrong KA. Communicating Uncertainty: a Narrative Review and Framework for Future Research. J Gen Intern Med. 2019;34:2586-2591.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 62]  [Cited by in RCA: 105]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
92.  Paterick TE, Patel N, Tajik AJ, Chandrasekaran K. Improving health outcomes through patient education and partnerships with patients. Proc (Bayl Univ Med Cent). 2017;30:112-113.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 59]  [Cited by in RCA: 173]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
93.  Jeyaraman N, Jeyaraman M, Ramasubramanian S, Balaji S, Muthu S. Voices that matter: The impact of patient-reported outcome measures on clinical decision-making. World J Methodol. 2025;15:98066.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (12)]
94.  Razavi Z, Soltani M, Souri M, van Wijnen AJ. CRISPR innovations in tissue engineering and gene editing. Life Sci. 2024;358:123120.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
95.  Hsu MN, Chang YH, Truong VA, Lai PL, Nguyen TKN, Hu YC. CRISPR technologies for stem cell engineering and regenerative medicine. Biotechnol Adv. 2019;37:107447.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 40]  [Cited by in RCA: 59]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
96.  Chen FM, Liu X. Advancing biomaterials of human origin for tissue engineering. Prog Polym Sci. 2016;53:86-168.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 824]  [Cited by in RCA: 648]  [Article Influence: 72.0]  [Reference Citation Analysis (0)]
97.  Rahmati M, Pennisi CP, Budd E, Mobasheri A, Mozafari M. Biomaterials for Regenerative Medicine: Historical Perspectives and Current Trends. Adv Exp Med Biol. 2018;1119:1-19.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 15]  [Cited by in RCA: 19]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
98.  Deptuła M, Zawrzykraj M, Sawicka J, Banach-Kopeć A, Tylingo R, Pikuła M. Application of 3D- printed hydrogels in wound healing and regenerative medicine. Biomed Pharmacother. 2023;167:115416.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 24]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
99.  Nugud A, Alghfeli L, Elmasry M, El-Serafi I, El-Serafi AT. Biomaterials as a Vital Frontier for Stem Cell-Based Tissue Regeneration. Front Cell Dev Biol. 2022;10:713934.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
100.  Revete A, Aparicio A, Cisterna BA, Revete J, Luis L, Ibarra E, Segura González EA, Molino J, Reginensi D. Advancements in the Use of Hydrogels for Regenerative Medicine: Properties and Biomedical Applications. Int J Biomater. 2022;2022:3606765.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 66]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
101.  Zhang Y, Wang J, Yu C, Xia K, Yang B, Zhang Y, Ying L, Wang C, Huang X, Chen Q, Shen L, Li F, Liang C. Advances in single-cell sequencing and its application to musculoskeletal system research. Cell Prolif. 2022;55:e13161.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 1]  [Cited by in RCA: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
102.  Choudhary RK, Choudhary S, Tripathi A. Emergence of the stromal vascular fraction and secretome in regenerative medicine. World J Stem Cells. 2024;16:896-899.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
103.  Agostini F, Rossi FM, Aldinucci D, Battiston M, Lombardi E, Zanolin S, Massarut S, Parodi PC, Da Ponte A, Tessitori G, Pivetta B, Durante C, Mazzucato M. Improved GMP compliant approach to manipulate lipoaspirates, to cryopreserve stromal vascular fraction, and to expand adipose stem cells in xeno-free media. Stem Cell Res Ther. 2018;9:130.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 28]  [Cited by in RCA: 39]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
104.  Jenio FZ, Scholes C, Marenah M, Li J, Cowley M, Ebrahimi M, Harrison-Brown M, Murrell WD. Quality in practice: implementation of a clinical outcomes registry in regenerative medicine. Ann Transl Med. 2019;7:130.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
105.  Abou-El-Enein M, Grainger DW, Kili S. Registry Contributions to Strengthen Cell and Gene Therapeutic Evidence. Mol Ther. 2018;26:1172-1176.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 25]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
106.  Lapuente JP, Dos-Anjos S, Blázquez-Martínez A. Intra-articular infiltration of adipose-derived stromal vascular fraction cells slows the clinical progression of moderate-severe knee osteoarthritis: hypothesis on the regulatory role of intra-articular adipose tissue. J Orthop Surg Res. 2020;15:137.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 22]  [Cited by in RCA: 35]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
107.  Goncharov EN, Koval OA, Nikolaevich Bezuglov E, Engelgard M, Igorevich EI, Velentinovich Kotenko K, Encarnacion Ramirez MJ, Montemurro N. Comparative Analysis of Stromal Vascular Fraction and Alternative Mechanisms in Bone Fracture Stimulation to Bridge the Gap between Nature and Technological Advancement: A Systematic Review. Biomedicines. 2024;12:342.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 6]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
108.  Piemonti L, Scholz H, de Jongh D, Kerr-Conte J, van Apeldoorn A, Shaw JAM, Engelse MA, Bunnik E, Mühlemann M, Pal-Kutas K, Scott WE 3rd, Magalon J, Kugelmeier P, Berishvili E. The Relevance of Advanced Therapy Medicinal Products in the Field of Transplantation and the Need for Academic Research Access: Overcoming Bottlenecks and Claiming a New Time. Transpl Int. 2023;36:11633.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]