ArticlePDF AvailableLiterature Review

Evaluating the quality of studies reporting on clinical applications of stromal vascular fraction: A systematic review and proposed reporting guidelines (CLINIC-STRA-SVF)

Authors:

Abstract and Figures

Background: The stromal vascular fraction (SVF) has been widely explored in a number of therapeutic applications in several specialties. Its therapeutic potential is being increasingly demonstrated, although its mechanism of action is still unclear. Objective: To evaluate the quality of studies reporting on clinical applications of SVF. Method: This is a systematic literature review that followed the PRISMA guidelines with the search of the studies from December 1, 2012, to December 1, 2022, in the following databases: MEDLINE, LILACS and EMBASE. The level of evidence of the studies was assessed using the GRADE system, and the rigor used in the publication of the results was assessed in relation to adherence to the guidelines indicated by the EQUATOR Network Group. The CLINIC - STRA-SVF reporting guideline was developed after the completion of this systematic review. Results: A total of 538 articles were found, and 77 articles were selected after reading the titles and abstracts and removing duplicates. Then, 15 studies were removed for not meeting the inclusion criteria, leaving 62 studies. The CLINIC - STRA-SVF was developed and consists of 33 items and two tables. Conclusion: There is scientific evidence, although mostly with a low level of evidence, that the use of SVF in clinical applications is safe and effective. The information published in these studies should be standardized, and the CLINIC - STRA-SVF reporting guideline proposed in this study may assist in the design, conduct, recording and reporting of clinical trials and others clinical studies involving the SVF.
Content may be subject to copyright.
Review
Evaluating the quality of studies reporting on clinical applications of
stromal vascular fraction: A systematic review and proposed reporting
guidelines (CLINIC-STRA-SVF)
Marcio Yuri Ferreira
a
,
*
, Jos
e da Conceiç~
ao Carvalho Junior
b
, Lydia Masako Ferreira
b
a
Translational Surgery Graduate Program of Universidade Federal de S~
ao Paulo - Unifesp, S~
ao Paulo, SP, Brazil
b
Plastic Surgery Division, Universidade Federal de S~
ao Paulo - Escola Paulista de Medicina, SP, Brazil
article info
Article history:
Received 14 June 2023
Received in revised form
26 July 2023
Accepted 13 August 2023
Keywords:
Stromal vascular fraction
Adipose-derived stem cells
Regenerative medicine
Scientic rigor
Guideline
abstract
Background: The stromal vascular fraction (SVF) has been widely explored in a number of therapeutic
applications in several specialties. Its therapeutic potential is being increasingly demonstrated, although
its mechanism of action is still unclear.
Objective: To evaluate the quality of studies reporting on clinical applications of SVF.
Method: This is a systematic literature review that followed the PRISMA guidelines with the search of the
studies from December 1, 2012, to December 1, 2022, in the following databases: MEDLINE, LILACS and
EMBASE. The level of evidence of the studies was assessed using the GRADE system, and the rigor used in
the publication of the results was assessed in relation to adherence to the guidelines indicated by the
EQUATOR Network Group. The CLINIC eSTRA-SVF reporting guideline was developed after the
completion of this systematic review.
Results: A total of 538 articles were found, and 77 articles were selected after reading the titles and
abstracts and removing duplicates. Then, 15 studies were removed for not meeting the inclusion criteria,
leaving 62 studies. The CLINIC eSTRA-SVF was developed and consists of 33 items and two tables.
Conclusion: There is scientic evidence, although mostly with a low level of evidence, that the use of SVF
in clinical applications is safe and effective. The information published in these studies should be
standardized, and the CLINIC eSTRA-SVF reporting guideline proposed in this study may assist in the
design, conduct, recording and reporting of clinical trials and others clinical studies involving the SVF.
©2023, The Japanese Society for Regenerative Medicine. Production and hosting by Elsevier B.V. This is
an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/
4.0/).
Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ............................ 333
2. Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ............................ 333
3. Results and discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . ............................ 334
3.1. Therapeutic applications . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . ...............................................335
3.2. Prevalence of studies that reported clinical benefits . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ...............................335
3.3. Summary study details . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ...............................................335
3.4. Route of administration of the SVF . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..........................................335
3.5. Design, methodological rigor and level of evidence of the studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ....................336
3.6. Follow-up ...................................................................................................................337
Abbreviations: ADSCs, adipose tissue stem cells; VEGF, vascular endothelial growth factor; HGF, hepatocytic growth factor; PDGFB, platelet-derived growth factor subunit
B; bFGF, basic broblast growth factor; SVF, stromal vascular fraction; BM-MSCs, bone marrow-derived stem cells; EPCs, endothelial progenitor cells; CLINICeSTRA-SVF,
Strengthening Reporting in Therapeutic Applications of the Stromal Vascular Fraction Guideline; FDA, United States Food and Drug Administration; AEs, Adverse events.
*Corresponding author: Pedro de Toledo St., 650-2ºFloor, P.O. Box: 04039-002., S~
ao Paulo, SP, Brazil.
E-mail address: marcioferreiramed@gmail.com (M.Y. Ferreira).
Peer review under responsibility of the Japanese Society for Regenerative Medicine.
Contents lists available at ScienceDirect
Regenerative Therapy
journal homepage: http://www.elsevier.com/locate/reth
https://doi.org/10.1016/j.reth.2023.08.003
2352-3204/©2023, The Japanese Society for Regenerative Medicine. Production and hosting by Elsevier B.V. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
Regenerative Therapy 24 (2023) 332e342
3.7. Details and scientific rigor applied in the evaluation of the AEs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ...............................337
3.8. Details about the SVF: characterization, viability, total cells, and nomenclature used by the author . . . . . . . . . . . . . . . . . . . . . . ..............338
3.9. Studies with therapeutics with any relationship with oncological pathologies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ....................338
3.10. CLINIC eSTRA-SVF Guideline development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . ...............................338
3.11. Present, future, needs and encouraging highlights . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . ...............................339
3.12. Study limitations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ...............................................340
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ............................ 340
Financial disclosure statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ............................ 340
Declaration of competing interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ..........................................340
References .................................................................... .....................................................340
1. Introduction
One of the most important characteristics of adipose tissue, in
addition to its thermoregulation, impact absorption and energy
storage, is that it is a source of regenerative cells [1]. In 1893,
Neuber et al. removed fat from the forearm and used it to ll in
volume and correct irregularities in the contour of the face caused
by a scar, and reported excellent cosmetic results [2]. Two decades
later, Morrestin was the rst to clinically and benecially apply
regenerative adipose tissue cells by transferring adipose tissue
particles to the wounds of soldiers injured in World War I [3]. Since
then, fat has been increasingly investigated, and hundreds of
studies have been published. Today, the scientic medical com-
munity continues to develop, modify, rene and innovate tech-
niques for exploiting the potential of adipose tissue [4].
In 2001, Zuk et al. demonstrated in vitro that populations of cells
isolated from adipose tissue obtained by liposuction can differen-
tiate into adipocytes, chondrocytes, osteocytes and myocytes [1].
Today, it is known that in addition to this ability, adipose tissue
stem cells (ADSCs), have antiapoptotic [5], antioxidant, anti-
inammatory and immunomodulatory properties [6].
These cells also exhibit angiogenic function, through the regu-
lation of the expression of vascular endothelial growth factor
(VEGF), hepatocytic growth factor (HGF), platelet-derived growth
factor subunit B (PDGFB) and basic broblast growth factor (bFGF)
genes [7] and may also aid wound healing by stimulating collagen
synthesis by broblasts [8].
Through the processing of fat, which can be enzymatic or me-
chanical, it is possible to obtain the stromal vascular fraction (SVF),
which is a rich source of ADSCs [9]. In addition, the SVF represents a
convenient source of ADSCs because it is easily obtained and yields
a higher proportion of stem cells than other methods of isolating
mesenchymal stem cells [9]. The eld of application of ADSCs is
greater than that of bone marrow-derived stem cells (BM-MSCs) in
regenerative medicine, due to their greater availability and because
they involve an easier isolation technique than BM-MSCs [10]. In
addition, 1 g of fat contains 1000 times more stem cells than 1 g of
bone marrow, and the quality and proliferation capacity of these
cells do not decrease with age [1,12].
The SVF includes cells of the immune system, especially T helper
lymphocytes, pericytes, broblasts, endothelial cells and stromal
precursor cells [13,14]. The two main stem cell phenotypes present
in SVF are endothelial progenitor cells (EPCs) and ADSCs; the main
actions of SVF are pro-angiogenic, anti-apoptotic, antibrotic,
immunomodulatory, anti-inammatory and trophic. Some of these
actions can be attributed to the presence of ADSCs (2e10%), while
others are associated with the vascular niche, and, alternatively, the
interactions between all cellular phenotypes present in SVF
together with the various growth factors [15].
The SVF is an important source of blood vessel-associated
cellular phenotypes, including pericytes (adventitial location),
endothelial progenitors (luminal location), mesenchymal pro-
genitors, supra-adventitial adipose stromal cells (vessel sheath),
and endothelial cells [16].
The SVF has been widely explored by various medical specialties
in various clinical situations. Because its use is a promising possi-
bility for clinical applications, there is a need to investigate all
clinical studies, compare them and understand the benets of the
SVF to formulate appropriate protocols for its use. The objective of
the present study was to evaluate the quality of studies reporting
on clinical applications of SVF.
2. Methods
The present study is a systematic literature review performed in
the Medline, Lilacs, and Embase databases following the Preferred
Reporting Items for Systematic Reviews and Meta-Analyses
(PRISMA) guidelines [17], therefore complying with the Enhancing
the Quality and Transparency Of health Research (EQUATOR) guide-
lines [18], covering studies published in the period from January 12,
2012 to January 12, 2022, in Portuguese, Spanish and/or English.
In the Medline database, the following search strategy was used:
(stromal vascular fraction[MeSH Terms]) AND (clinical applica-
tionOR wound healing[MeSH Terms] OR burn OR face rejuve-
nationOR cicatrix [MeSH Terms] OR therapeutics [MeSH Terms] OR
cardiovascular diseases[MeSH Terms] OR musculoskeletal dis-
eases[MeSH Terms] OR respiratory tract diseases[MeSH Terms]
OR female urogenital diseases[MeSH Terms] OR male urogenital
diseases[MeSH Terms] OR Eye Diseases[MeSH Terms] OR
regenerative medicine[MeSH Terms] OR autoimmune diseases
[MeSH Terms] OR neurodegenerative diseases[MeSH Terms] OR
pressure ulcer[MeSH Terms] OR plantar fat pad atrophy).
The following was used in the Embase database: stromal
vascular fractionand clinical application. And the following was
used in the Lilacs database: stromal vascular fraction.
Studies were included if they were clinical trials, prospective
and retrospective cohorts, case series and case reports, only in
humans and that used enzymatically extracted stromal vascular
fraction. We excluded experimental studies, secondary studies,
studies that did not use freshly isolated SVF, studies that were
retracted for ethical reasons, studies that used SVF extracted by
ultrasound or mechanical cavitation, and studies that did not
describe the method of SVF processing.
After reading the title and abstract, all retrieved articles were
tabulated and included in a database. The selection and reading
process of the articles were performed by two of the authors (MYF
and JC). All articles selected by the two authors were denitively
included in the systematic review. The articles selected by only one
of the authors were analyzed by the senior author (LMF) and
included only when there was agreement among the threeauthors.
The full text was obtained for the full evaluation and nal inclusion
of the articles.
M.Y. Ferreira, J.C. Carvalho Junior and L.M. Ferreira Regenerative Therapy 24 (2023) 332e342
333
The level of evidence of the studies was classied by the authors
according to the levels determined by the Grading Recommenda-
tions, Assessment, Development and Evaluations (GRADE) System
[19]. The studies were evaluated regarding adherence to the
EQUATOR Network Guidelines [18]; specically, each type of study
design was evaluated according to the appropriate guidelines:
randomized clinical trials eCONSORT [20]; clinical trials e
Guidelines for reporting non-randomised studies [21]; case studies
series ePROCESS [22]; case reports eCARE [23], and observational
studies eSTROBE [24].
The studies were evaluated in relation to the methodology for
evaluating the manifestations and monitoring of adverse events
(AEs), following the recommendations of the study Reporting
Adverse Events in Plastic Surgery: A Systematic Review of Ran-
domized Controlled Trials[25].
The preparation of the CLINIC eSTRA-SVF eStrengthening
Reporting in Therapeutic Applications of the Stromal Vascular
Fraction Guideline was performed following the AGREE [26]
recommendations, in addition to How to develop a reporting
guideline[27] of the EQUATOR Network [18], and was guided by
the results obtained in the evaluation of the studies included in this
review. Before starting the preparation of the Guideline, a search on
the Equator Network [18] platform was performed to verify the
existence of published or under development reporting guidelines
applied to publications of clinical studies involving the stromal
vascular fraction, through December 2022.
The data contained in the included articles were independently
extracted by the researchers and evaluated together to obtain
consensus on the information therein.
3. Results and discussion
In total, 538 articles were initially identied. Of these, 77 articles
were selected after reading the titles and abstracts and removing
duplicates. Then, 15 studies were removed because they met the
exclusion criteria, leaving 62 studies [28e89]. (Fig. 1) After reading
Fig. 1. Prisma ow diagram.
M.Y. Ferreira, J.C. Carvalho Junior and L.M. Ferreira Regenerative Therapy 24 (2023) 332e342
334
and completely analyzing these articles, all of them were included
in the present review.
3.1. Therapeutic applications
A total of 41 different therapeutic applications were found in the
selected studies, as shown in Table 1. The most prevalent thera-
peutic applications in the studies were osteoarthritis of the knee
(n ¼9; 14.5%) and volume restoration in breast reconstruction
(n ¼6; 9.7%). The identied studies showed that several specialties
have a promising future in the use of SVF, especially in relation to
pathologies in which the available treatments do not offer a cure.
The therapeutic objectives ranged from the use of SVF for cosmetic/
aesthetic purposes such as facial rejuvenation, which enhances
aesthetic results in procedures involving fat grafting, to therapeutic
procedures in osteoarticular, neurological, cardiovascular diseases
and vocal cord injuries, infertility, neuromas, post-trauma and
poststroke sequelae, autoimmune diseases (e.g., systemic sclerosis
and ankylosing spondylitis), neurodegenerative diseases (e.g., Par-
kinson's and Alzheimer's), chronic and refractory stulas and ul-
cers, erectile dysfunction, etc. (Table 1)
3.2. Prevalence of studies that reported clinical benets
The data extracted from the studies included in this review are
encouraging regarding the use of SVF. Most studies reported sta-
tistically signicant clinical benets in the results (n ¼56; 90.3%); 3
studies were designed to evaluate safety only [63,82,89], and the
remaining 3 studies reported that the use of SVF did not offer
benets in improving pain in 5 cases of painful neuromas [59]or
volume retention in breast reconstruction [53,77], although in the
latter application, another study reported the efcacy [81] and
another demonstrated the safety of SVFs [82]. (Table 2).
Although the mechanism of action of the SVF, in its various
clinical applications and methods of application, is not yet fully
understood, these studies demonstrate the therapeutic potential of
the SVF.
3.3. Summary study details
The year of publication of the studies, the country where the
study was conducted and the journal in which the article was
published are shown in Table 3. The years with the highest number
of published articles were 2020 (n ¼13; 21%) and 2016 (n ¼10;
16.1%), the countries with the highest number of published articles
were South Korea (n ¼12; 19.9%) and the USA (n ¼11; 16.6%), and
the journals with the largest number of published articles were
Stem Cell Res Ther (n ¼6; 9.6%) and Aesthet Surg J (n ¼5; 8%).
3.4. Route of administration of the SVF
The most prevalent routes of administration in the included
studies were subcutaneous (n ¼16; 25.8%) and intra-articular
(n ¼15; 24.2%). Two intravenous applications were performed
and demonstrated safety, with no reports of adverse effects [54,62].
Innovative forms of SVF application have been described, such as
intramyocardial, intraventricular (brain), suburethral, intravocal
folds, intracavernous, intratumoral, intrauterine and enriching
dermis grafts (Table 4).
In this review, the therapeutic applications of the SVF were used
alone and in combination (e.g., SVF þPRP - Platelet-Rich Plasma -,
SVF þL-PRP - leukocyte- and platelet-rich plasma -), SVF þFat
Graft, SVF þACAM2000); the routes were intravenous in a total of
26 patients, intraperitoneal in 4 patients, intratumoral in 17 pa-
tients, intracavernous in 17 patients, intramyocardial in 28 patients,
intrauterine in 6 patients, and intracerebral in 31 patients (with a
total of 113 applications were performed); among all of these, no
serious adverse effects were reported during the follow-up. One
patient received intravenous application of a thawed SVF after
cryopreservation, and with a 3-year follow-up, the study reported
an absence of AEs. One patient received a combination of SVF þPRP
that was frozen for 10 days for treatment of a refractory foot ulcer
that remained unhealed for 2 years, and the study reported treat-
ment efcacy and the absence of AEs.
Table 1
Therapeutic applications.
No. (%)
Therapeutic objective
Osteoarthritis of the knee 9 (14,5%)
Volume restauration in breast reconstruction 6 (9,7%)
Perianal stulas of Crohn's disease 3 (4,8%)
Systemic sclerosis 3 (4,8%)
Androgenetic Alopecia 2 (3,2%)
Atrophic acne scars 2 (3,2%)
Diabetic ulcer 2 (3,2%)
Peripheral vascular disease 2 (3,2%)
Alopecia areata 1 (1,6%)
Asherman's Syndr 1 (1,6%)
Avascular necrosis femoral head 1 (1,6%)
Bone regeneration 1 (1,6%)
Breast aumengtation 1 (1,6%)
Chondromalacia patellae 1 (1,6%)
Chronic fatigue and depression in Ankilosing Spondylitis 1 (1,6%)
Contracted nose erev. rhinoplast 1 (1,6%)
Contracted scars 1 (1,6%)
Cosmetic and Recons approachs 1 (1,6%)
Cryptoglandular Fistulae-in-Ano 1 (1,6%)
Degenerative disc disease 1 (1,6%)
Erectile Disfunction 1 (1,6%)
Infraorbital Skin Rejuvenation 1 (1,6%)
Ischemic cardiopathy 1 (1,6%)
Lymphedema 1 (1,6%)
Neurodegenerative Disorders 1 (1,6%)
Nose after BCC excision 1 (1,6%)
Osteonecrosis of the jaw 1 (1,6%)
Orthopedic pathologies 1 (1,6%)
Painful neuroma 1 (1,6%)
Parkinson's disease 1 (1,6%)
Photoaging 1 (1,6%)
Posterior lumbar interbody fusion 1 (1,6%)
Rejuvenation 1 (1,6%)
Salvage of Ischemic Fasciocutaneous ap 1 (1,6%)
Scarred Vocal Folds 1 (1,6%)
Solid neoplasies 1 (1,6%)
Stroke rehabilitation 1 (1,6%)
Treatment of amputed site 1 (1,6%)
Treatment of ulcus cruris of venous/mixed venous arterial 1 (1,6%)
Urinary incontinence 1 (1,6%)
X ray injury 1 (1,6%)
Table 2
Authors reported benets?
No. (%)
Yes 56 (90,3%)
No 3 (4,8%)
Only safety 3 (4,8%)
M.Y. Ferreira, J.C. Carvalho Junior and L.M. Ferreira Regenerative Therapy 24 (2023) 332e342
335
In all these applications, the safety of the applications was re-
ported, although at different levels of evidence, providing more
condence for the scientic community to establish new trials that
demand such applications.
3.5. Design, methodological rigor and level of evidence of the
studies
Only 6 (9.7%) of the studies were randomized clinical trials, and
26 (41.4%) were nonrandomized clinical trials, demonstrating that
approximately 50% of the studies included in this review were case
series, case reports, and cohorts (Table 5). These data demonstrate
the low quality of the available evidence because without robust
and randomized studies, the evidence obtained will always be
limited or have a low level of condence. This low level of con-
dence was also the result obtained in the application of the Grade
System [20] classication on the included studies, which resulted in
56 (90.3%) articles with a low level of evidence and 6 (9.68%) arti-
cles with a high level of evidence.
The EQUATOR Network [16] is an initiative recognized and
endorsed by scientic medical journals worldwide. The platform
provides guidelines, materials and tools that offer solutions
necessary for studies to be prepared and published with the
maximum scientic rigor possible by the medical community and
an extensive list of guidelines that can be used in each design and/
or study area. The group is dened as an umbrellaorganization
that brings together researchers, medical journal editors, peer re-
viewers, developers of reporting guidelines, research funding
bodies and other collaborators with mutual interest in improving
the quality of research publications and of research itself[18]. The
data extracted from the studies regarding adherence to the
Table 3
Summary details of the 62 studies.
No. (%)
Year of publication
2022 6 (9,6%)
2021 5 (8%)
2020 13 (21%)
2019 5 (8%)
2018 9 (14,5%)
2017 6 (9,6%)
2016 10 (16,1%)
2015 5 (8%)
2014 1 (1,6%)
2013 1 (1,6%)
Country
South Korea 12 (19,3%)
USA 11 (17,7%)
France 7 (11,3%)
China 5 (8%)
Nicaragua 4 (6,4%)
Belgium 2 (3,2%)
Brazil 2 (3,2%)
Denmark 2 (3,2%)
Germany 2 (3,2%)
Iran 2 (3,2%)
Australia 1 (1,6%)
Greece 1 (1,6%)
Italy 1 (1,6%)
Japan 1 (1,6%)
Lebanon 1 (1,6%)
Pakistan 1 (1,6%)
Poland 1 (1,6%)
Spain 1 (1,6%)
Switzerland 1 (1,6%)
Taiwan 1 (1,6%)
The Neterlands 1 (1,6%)
Thailand 1 (1,6%)
Vietnam 1 (1,6%)
Journal
Stem Cell Res Ther 6 (9,6%)
Aesthet Surg J 5 (8%)
J Cosmet Dermatol 3 (4,8%)
J Transl Med 3 (4,8%)
Stem Cells Transl Med 3 (4,8%)
Aesth Plast Surg 2 (3,2%)
Ann Plast Surg 2 (3,2%)
BMC Musculoskelet Disord 2 (3,2%)
Cells 2 (3,2%)
Plast Reconstr Surg 2 (3,2%)
Stem Cell Res 2 (3,2%)
Surg Innov 1 (1,6%)
Adv Clin Exp Med 1 (1,6%)
Am J Sports Med 1 (1,6%)
Ann Rheum Dis 1 (1,6%)
Biomed Res Int 1 (1,6%)
Curr Res Transl Med 1 (1,6%)
EBioMedicine 1 (1,6%)
Eur J Obstet Gynecol Reprod Biol 1 (1,6%)
Eur Rev Med Pharmacol Sci 1 (1,6%)
Gastroenterology 1 (1,6%)
Health Phys 1 (1,6%)
Int J Low Extrem Wounds 1 (1,6%)
Int Orthop 1 (1,6%)
J Clin Neurosci 1 (1,6%)
J Eur Acad Dermatol Venereol 1 (1,6%)
J Int Med Res 1 (1,6%)
J Med Case Rep 1 (1,6%)
J Orthop Surg Res 1 (1,6%)
J Plast Reconstr Aesthet Surg 1 (1,6%)
J Stomatol Oral Maxillofac Surg 1 (1,6%)
JAMA Otolaryngo Head Neck Sur 1 (1,6%)
Medicine (Baltimore) 1 (1,6%)
Microsurgery 1 (1,6%)
Mol Biol Rep 1 (1,6%)
PLoS One 1 (1,6%)
Table 3 (continued )
No. (%)
Regen Med 1 (1,6%)
Reprod Sci 1 (1,6%)
Rheumatology (Oxford) 1 (1,6%)
Stem Cells Int 1 (1,6%)
Surg Cosmet Dermatol 1 (1,6%)
Table 4
Route of administration.
No. (%)
Subcutaneous 16 (25,8%)
Intra-articular 15 (24,2%)
Intradermic 7 (11,3%)
Injected in the wound 5 (8%)
Peri/intra-stular 4 (6,45%)
Between muscles 2 (3,2%)
Engineered dermis graft 1 (1,6%)
Into the corpo cavernosum 1 (1,6%)
Intradiscal 1 (1,6%)
Intralesional (contracted nose) 1 (1,6%)
Intramiocardic 1 (1,6%)
Intramuscular 1 (1,6%)
Intrauterine 1 (1,6%)
Intravenous 1 (1,6%)
Intravenous and/or intra-tumoral and/or intra-peritoneal 1 (1,6%)
Intraventricular (brain) 1 (1,6%)
Intra-vocal folds 1 (1,6%)
Peri-fascial and peri-chondral 1 (1,6%)
Suburethral 1 (1,6%)
M.Y. Ferreira, J.C. Carvalho Junior and L.M. Ferreira Regenerative Therapy 24 (2023) 332e342
336
EQUATOR guidelines and the guidelines suggested by the group
show that almost none of the studies related to therapeutic appli-
cations adhered to these guidelines; only 3 of the 62 studies
adhered to them, resulting in a 95.2% nonadherence rate. The three
studies that adhered to the study used the CONSORT (n ¼1; Mattei
et al. [45]) and STROBE (n ¼2; Moon KC et al. [50] and Lee JW et al.
[58]) recommendations.
The results of the GRADE classication and the adherence of the
studies to the EQUATOR guidelines demonstrate that the studies
related to the clinical applications of SVF are of low quality.
3.6. Follow-up
The follow-up time, the description of the presence or absence
of AEs, and the methodology used to assess the presence of AEs are
determining factors in the publication of the safety of the new
therapies studied. Regarding the follow-up, most studies involved a
period between 12 and 36 months (n ¼40; 64.5%), and the mean
was 18.6 months (Table 6). The studies were accessed to extract
data related to the methodology for evaluating the AEs according to
previously published guidelines [25]. Among the 62 studies, 59
(95.16%) did not use a validated method to access and classify the
AEs of the established therapies, and 55 (88.8%) did not dene in
advance which AEs would be evaluated during the follow-up.
Furthermore, none of the 62 (100%) reported who the researcher
or collaborator responsible for documenting the AEs was, and a
total of 17 (27.4%) did not mention the presence or absence of AEs,
which should be considered a fundamental error in studies that
seek to describe clinical applications of new therapies (Table 7).
Of the 62 studies in this review, only 20 (32.26%) reported the
presence of AEs, among which 17 studies declared that the AEs
were somehow directly related to the type of intervention per-
formed, such as surgical site infection, knee inammation after
intra-articular application, and pain resulting from liposuction. No
serious adverse events, such as death, thromboembolic events,
neoplastic transformations or hypersensitivity reactions that could
generate a risk of death and/or hemodynamic instability or
neurological changes related to the SVF were reported.
3.7. Details and scientic rigor applied in the evaluation of the AEs
Of the 62 studies, 59 (95.16%) did not use a validated method to
assess the AEs of the established therapies, 55 (88.8%) did not
dene in advance which AEs would be evaluated during the follow-
up, 62 (100%) did not report who the researcher or collaborator
responsible for documenting the AEs was, 42 (67.74%) reported that
there were no AEs and 20 (32.26%) reported AEs. Of the 20 authors
who reported AEs, 17 (85% of the reported AEs) reported that the
effects were related to the instituted procedure and not directly
related to the SVF, and 3 (4.8% of all studies) reported that the AEs
were directly related to the SVF. None of the authors reported
neoplastic transformation during the follow-up of the studies, as
shown in Table 7.
Among the studies, a total of 17 (27.4%) did not mention the
presence or absence of AEs. The studies that used methods to
evaluate AEs used the following guidelines/methods: MOSES [93],
MedDRA [94] and the Common Terminology Criteria for Adverse
Events (CTCAE) [95].
The lack of validated methodologies and the lack of publication
of the methodology and the people involved in the evaluation of
the AEs hinder the reproducibility of the studies, which may make
the published evidence of safety questionable and incapable of
demonstrating its reliability per se using the published methods.
As important as it is to describe the adverse events, it is also
necessary that the authors adopt a method of identication, follow-
up, classication and publication of these events and that they
make this clear in the publication, ensuring that, from the study
Table 5
Methodological details of the studies.
No. (%)
Design of the study
Randomized clinical trial 6 (9,7%)
Non-randomized clinical trial 26 (41,4%)
Case series 9 (14,5%)
Case report 10 (16,1%)
Case-Control 1 (1,6%)
Retrospective Cohort 3 (4,8%)
Prospective Cohort 7 (11,3%)
EQUATOR network recommen-dations
Yes 3 (4,8%)
No 59 (95,2%)
GRADE classication
Low 56 (90,3%)
High 6 (9,68%)
Adoppted EQUATOR recommedend guideline
None 59 (95,2%)
CONSORT 1 (1,6%)
Nonrandomized guideline 0 (0%)
PROCESS 0 (0%)
CARE 0 (0%)
STROBE 2 (1,6%)
Other guidelines adopted
GPP eGuidelines 2022 [90] 1 (1,6%)
AIOM - follow-up guidelines [91] 1 (1,6%)
ICH-GCP [92] 2 (1,6%)
Tabela 6
Follow-up.
No. (%)
<1 months 1 (1,6%)
1e5 months 4 (%)
6e11 months 14 (%)
12e23 months 25 (%)
24e36 months 15 (%)
More than 37 months 3 (%)
Table 7
Details about adverse events (AEs).
No. (%)
A validated method to access AE was used?
No 59 (95,16%)
Yes 3 (4,8%)
Anticipated AE clearly dened
No 55 (88,7%)
Yes 7 (11,3%)
Was it clear who would be documenting any AE?
No 62 (100%)
Yes 0 (%)
Ocurrence of AE reported?
No 42 (67,74%)
Yes 20 (32,26%)
Authors declared that AE reported was common
and no serious AE of the type of the
proccedure and/or not related direct to SVF
17 (85% of 20)
Statement saying there where or no AE?
No 17 (27,4%)
Yes 45 (72,6%)
Total number of AE that authors declared that
there was a proba-ble relationship with SVF
3 (4,8%)
Neoplasy transformation reported 0 (0%)
Method for access AE 3 (4,8%)
CTCAE 1
MOSES 1
MedDRA 1
M.Y. Ferreira, J.C. Carvalho Junior and L.M. Ferreira Regenerative Therapy 24 (2023) 332e342
337
protocol, the AEs are accessed according to previously published
guidelines and following validated methods [25].
3.8. Details about the SVF: characterization, viability, total cells, and
nomenclature used by the author
Of the 62 studies, 28 (45.16%) did not report the total nucleated
cells in the SVF, 36 (58%) did not report the cell markers present in
the SVF used, and 32 (51.61%) did not describe the viability of the
SVF cells (Table 8). There are numerous variables that inuence the
viability and quantity of cells present in the SVF, and to compare
doses, therapeutic effects, and adverse effects, it is necessary to
know exactly which and how many cells were present in the SVF
used and their corresponding viabilities.
Among the studies that published the markers of the cells
present in the SVF, there was a great difference in the way they
were published and in the number of markers that were evaluated.
To allow a comparison and abetter understanding of the SVF and its
clinical applications, works that involve the SVF must evaluate and
publish the markers of the cells present in a standardized manner
as previously described [15,96,97].
More than 60% of the SVF cells is CD34
þ
, a marker present in the
cells of the vascular niche. The composition of the SVF may vary
depending on the processing protocol [98e100], and in the studies
included, numerous protocols were used. The International Feder-
ation for Adipose Therapeutics (IFATS) and the International Society
for Cellular Therapy (ISCT) have provided guidelines and recom-
mendations for the minimum basic characterization of the SVF, i.e.,
CD45
CD235a-CD31
CD34
þ
[97].
Regarding the nomenclature used by the authors to describe the
stromal vascular fraction, a total of 7 different acronyms were used:
SVF (n ¼49; 79%), ADSVF (n ¼7; 11.3%), AD-SVF (n ¼1), ADSVCs
(n ¼1), ADRCs (n ¼1), ASCs (n ¼1) and ADSCs (n ¼1) (Table 8).
This difference in the use of acronyms to refer to the stromal
vascular fraction makes standardization and better indexing of
these articles, as well as future searches in databases, impossible,
generating confusion that can and should be avoided. On January
01, 2022, the term stromal vascular fractionwas indexed as a
medical subject heading (MeSH) term and thus should be used in
future clinical studies involving the use of SVF.
3.9. Studies with therapeutics with any relationship with
oncological pathologies
An important concern in the context of the therapeutic appli-
cations of stem cells is the carcinogenic potential and the concern
generated, especially in patients who have or had affected by
neoplasms.
In this review, six of the included studies reported 184 onco-
logical related patients who were exposed to SVF therapies
[38,50,54,57,63,81], with a mean follow-up of 30.8 months and a
range of 6e96 months. None of the studies reported neoplastic
transformations and/or an increase in the number of relapses, thus
reinforcing the safety of the use of SVF (Table 9). These studies are
fundamental for the advancement of the application of SVF in new
therapies, serving as a basis for demonstrating evidence on the
safety of the SVF to regulatory agencies such as the FDA conferring
safety for the physician and the patient who will be treated with
such therapy.
3.10. CLINIC eSTRA-SVF Guideline development
No reporting guidelines published or under development were
identied on the EQUATOR platform with regard to studies
involving the SVF.
At the conclusion of this systematic review, the CLINIC eSTRA-
SVF guideline was developed, producing a checklist with 33 items
and two tables that includes the fundamental information that
should be published in studies evaluating therapies related to SVF
(Tables 10e12).
The guideline was developed with the intention of being
applicable to the complete elaboration of a study, from the writing
of the initial project and research protocol to the execution during
the follow-up, and through the publication of the collected
information.
The present study investigated the clinical scope and the quality
of studies reporting the use of enzymatic-extracted stromal
vascular fraction in clinical application, through the level of evi-
dence of the publications, the methodologies used to perform the
research and publish the results, the quality of the information
related to the SVF, as well as the results obtained in these studies
and the safety of the use of the SVF in these applications. In an era
where regenerative medicine and cell therapies have emerged
promising therapeutic solutions for various pathologies and
regenerative applications, the worldwide medical-scientic com-
munity has begun to increasingly exploit the potential of ADSCs and
a worldwide dedication to investigating the relationship between
the fat, the stromal vascular fraction and its regenerative potential
in a wide range of preclinical and clinical studies in an expanding
Table 8
Details about SVF.
No. (%)
Was reported the total nucleated cells in SVF?
No 28 (45,16%)
Yes 34 (54,84%)
Was reported the markers of cells present in SVF?
No 36 (58,06%)
Yes 26 (41,94%)
Was reported the viability of SVF cells?
No 32 (51,61%)
Yes 30 (48,39%)
Nomenclature used for SVF
SVF 49 (79,03%)
ADSVF 7 (11,3%)
AD-SVF 1 (1,6%)
ADSVCs 1 (1,6%)
ADRCs 1 (1,6%)
ASCs 1 (1,6%)
ADSCs 2 (3,2%)
Table 9
Oncologic related studies.
No. (%)
Total patients exposed to SVF therapeutic 184
Medium follow-up 30,8 months
Range follow-up 6e96 months
Jeon HJ et al. [39]
N;¼20
Follow eup 12
Moon KC et al. [51]
N;¼77 (30 SVF group)
Follow-up 35 months/range - 1-8y
Minev BR et al. [55]
N;¼26
Follow-up 6 months
Calabrese et al. [58]
N;¼169 (41 SVF)
Follow - up At least 60 months
Mazur S et al. [64]
N;¼298 (56 SVF)
Follow-up 36 months
Tissiani LAL and Alonso N [82].
N;¼19 (11 SVF group)
Follow-up 36 months
M.Y. Ferreira, J.C. Carvalho Junior and L.M. Ferreira Regenerative Therapy 24 (2023) 332e342
338
global market [4,101e109]. This can be explained by its greater
availability and ease of extraction or its higher cell yield relative to
other forms of stem cell extraction [9e11]; thus, SVF has become
the focus of attention, discussion and investigation by both clinical
and surgical specialties. However, the literature is still recent, and
publications on clinical applications are not standardized and still
lack the necessary scientic rigor. This lack of standardization
makes reliable comparisons impossible, and thus, a systematic re-
view can aid in this regard by demonstrating the current scientic
evidence regarding the use, risks, efcacy, safety and other pecu-
liarities involved in the various possibilities of the application of the
SVF. Thus, in this panorama, the key aspects that will enable
progress in the methodological standardization of future publica-
tions on the clinical applications of SVF are emphasized.
3.11. Present, future, needs and encouraging highlights
The applications found in the articles included in this study,
their results and safety, confer legitimacy to the establishment of
new therapeutic hypotheses and deepening our understanding of
the investigations already conducted. The use of the SVF for the
benet of patients, especially for diseases for which we currently do
not have well-established effective therapies such as neurodegen-
erative diseases (e.g., Parkinson's, Alzheimer's, Multiple Sclerosis),
autoimmune diseases (e.g., scleroderma, ankylosing spondylitis),
heart diseases involving areas of tissue necrosis, erectile dysfunc-
tion and many others, is encouraging, opening the possibility for
new studies to test new applications and further exploring the
regenerative, immunomodulatory and anti-inammatory potential
of SVF.
New studies involving the clinical applications of the SVF should
focus on increasing the scientic rigor of investigations and pub-
lication of results, providing the necessary reproducibility, greater
transparency and level of condence in the published evidence
[110 e112 ].
In summary, it was evident that there was a lack of standardi-
zation in publications and basic information related to SVF,
including the techniques used for collection, extraction, and
application, patient follow-up, and other factors. This lack of stan-
dardization hinders the availability of consistent information and
limits the ability to make comparisons and produce stronger sci-
entic evidence. Some studies, did not publish information such as
the age of the patients or the location of fat collection, which is
basic information for this type of study. As concluded by Kelleyet al.
[113 ], unless the data are comparable and the original studies are
of high-quality, meta-analysis should be avoided in favor of a
comprehensive systematic review. Inappropriate pooling of data
should be avoided to preserve the quality of the research and to
Table 10
CLINIC eSTRA-SVF checklist.
Section Item Checklist Item
Study Design 1a Study conducted in accordance with EQUATOR network guidelines?
1b CONSORT (RCT), Guidelines for Reporting non-randomised studies (NRCT), STROBE (cohort, case-control or cross-sectional),
PROCESS (case series) or CARE (case report)
1c Validated method was used to access Adverse Events?
1d Structured summary of trial design, methods, results, and conclusions (for specic guidance see CONSORT for abstracts)
Therapeutic objective 2a Clear description of the therapeutic goal
2b Clear description if it's the rst study in that scope or all previous studies published
2c Clear description about the outcomes that will be evaluated
General details 3a Objective description of the method of the outcome assessment (Validated tools, assessment method proposed by the author
and the reason it was chosen)
3b Description of the age and gender of the patients
3c Exact description of the fat harvest site
3d Complete description of the fat processing method (specifying whether it was extracted manually or by an automated method.
What device was used.)
3e Detailed description of the enzyme and concentration used for enzymatic digestion
3f Description of the amount of fat harvested, amount of fat processed by the enzymatic process, amount of SVF obtained, and ratio
of SVF yield/mL of fat processed.
3g Description of the time interval between fat collection, start and end of enzymatic processing
3h Description regarding the form of SVF used: immediately after processing cryopreserved other manipulations (describe which
manipulation was done)
3i The nomenclature used was SVF - Stromal Vascular Fraction?
SVF Charac-terization 4a Description of the SVF characteristics according to Table 12
4b Description of the characterization of the cells according to the markers
4c Characterization of the cells present in SVF by the markers
Application Details 5a Description of the application method with number of applications
5b Description of the applied quantity and/or combination used
5c Description of the interval time between fat collection and application of SVF
Pos-op 6a Description of dressing used
6b Description of post-procedure care/orientations
Follow-up 7a Description of the patients follow-up time
7b Description of the interval and number of evaluations performed during the patients' follow-up
Adverse events (AEs) 8a What validated method was used to access AEs?
8b How was classied the severity of the AEs?
8c The patients were informed about the possible and probable AEs?
8d Description of the evolution of the AEs and how it was treated
Outcome Results 9a Objective description of the results
Funding 10a Sources of funding and other support (such as supply of devices or drugs, products related with SVF), role of funders
Table 11
Model for evaluation and reporting the characteristics of stromal vascular fraction.
Stromal Vascular Fraction Mean/range
Adipose Tissue Harversted (cm [3])
Volume isolated (ml)
Number of viable nucleated cells (VNC) (millions) obtained
Recovery rate (VNC/cm [3] adipose tissue)
Number of VNC injected or applied (millions)
Viability (%)
M.Y. Ferreira, J.C. Carvalho Junior and L.M. Ferreira Regenerative Therapy 24 (2023) 332e342
339
avoid unjustiable conclusions. Adherence to the principles of the
scientic methods outlined will improve publication acceptance
rates and the overall quality of literature in the eld.This is also a
concept that should be taken into account in the acquisition of
robust scientic evidence related to the SVF.
In addition to this context, the reproducibility of the studies
must be guaranteed by the author during the project preparation
period, during the research and at the time of publication of the
results. Corroborating the ndings of Ascha et al. [111], this study
demonstrates that research reproducibility is a mandatory
component of the scientic method and is necessary for ensuring
the accuracy of the scienticndings and for cultivating trans-
parency and reproducibility in scientic research.
In this context, the authors of this review, driven by the evident
need for standardization as observed during the preparation of this
systematic review, proposed the creation of the CLINIC eSTRA-SVF
Guideline. The checklist aims to be easy to apply, and the authors
encourage researchers to prepare their research projects and pro-
tocols using the guideline for the development of a highly scien-
tically rigorous method. At the time of publication, the authors
should seek to ll in as many items in the guideline as possible
according to the reality and possibilities of their research projects to
ensure transparency, reproducibility, standardization and progress
in the construction of evidence related to the clinical applications of
the SVF.
Randomized clinical trials and prospective cohort studies, both
highly rigorous in methodological design, in the publication of re-
sults and detailed information, and validated methodologies for
evaluating the adverse effects of the clinical applications of SVF
should be encouraged, following the CLINIC eSTRA-SVF guideline
recommendations in the reporting of these studies.
3.12. Study limitations
Although this systematic review was prepared following the
PRISMA guidelines and sought through the EQUATOR guidelines
and the GRADE system to infer the level of evidence and identify
the global state of the clinical applications of SVF, it was difcult to
compare the information due to the low level of rigor in the
methodological approaches and the lack of standardization,
reproducibility and transparency of the included studies.
4. Conclusion
There is scientic evidence of the safety and effectiveness in the
clinical application of the SVF; however, the level of evidence of
most of the studies was low, and much of the evidence was pub-
lished in articles that do not follow the reporting methods used by
the scientic community worldwide and that do not use validated
methodologies for the evaluation of adverse effects. It is necessary
to standardize the information published in the studies involving
therapies with SVF, and the CLINIC eSTRA-SVF reporting guideline
proposed in this study may assist in the design, conduct, recording
and reporting of clinical trials, case series, and case reports
involving the SVF. The use of the CLINIC eSTRA-SVF guideline will
ensure the construction of studies and the publication of evidence
that guarantee the reproducibility, comparisons and transparency
necessary for future scientically rigorous comparisons to be per-
formed by the world scientic community and thus the construc-
tion and accumulation of better evidence for the benet of patients.
Financial disclosure statement
The authors have nothing to disclose.
Declaration of competing interest
None.
References
[1] Zuk PA, Zhu M, Mizuno H, et al. Multilineage cells from human adipose
tissue: implications for cell-based therapies. Tissue Eng 2001;7(2):211e28.
[2] XX Neuber G. Über die Wiederanheilung vollst
andig vom K
orper getrennter,
die ganze Fettschicht enthaltender Hautstücke. Zentralblatt Chir 1893;30:16.
[3] Morestin H. Quelques cas de greffes graisseuse appliquees aIa chirurgie
reparatrice. Bull Mem Soc Chir (Paris) 1915;41:1631.
[4] Shauly O, Gould DJ, Ghavami A. Fat grafting: basic science, techniques, and
patient management. Plast Reconstr Surg Glob Open 2022;10(3):e3987.
[5] Rehman J, Traktuev D, Li J, et al. Secretion of angiogenic and antiapoptotic
factors by human adipose stromal cells. Circulation 2004;109(10):1292e8.
[6] Kim WS, Park BS, Sung JH, et al. Wound healing effect of adipose-derived
stem cells: a critical role of secretory factors on human dermal broblasts.
J Dermatol Sci 2007;48(1):15e24.
[7] Nakagami H, Maeda K, Morishita R, et al. Novel autologous cell therapy in
ischemic limb disease through growth factor secretion by cultured adipose
tissue-derived stromal cells. Arterioscler Thromb Vasc Biol 2005;25(12):
2542e7.
[8] Atalay S, Coruh A, Deniz K. Stromal vascular fraction improves deep partial
thickness burn wound healing. Burns 2014;40(7):1375e83.
[9] Guo J, Widgerow AD, Banyard D, et al. Strategic sequences in fat graft sur-
vival. Ann Plast Surg 2015;74(3):376e82.
[10] Mazini L, Rochette L, Amine M, Malka G. Regenerative capacity of adipose
derived stem cells (ADSCs), comparison with mesenchymal stem cells
(MSCs). Int J Mol Sci 2019;20(10):2523.
[11] Oryan A, Kamali A, Moshiri A, Baghaban Eslaminejad M. Role of mesen-
chymal stem cells in bone regenerative medicine: what is the evidence? Cells
Tissues Organs 2017;204(2):59e83.
[12] Beane OS, Fonseca VC, Cooper LL, Koren G, Darling EM. Impact of aging on
the regenerative properties of bone marrow-, muscle-, and adipose-derived
mesenchymal stem/stromal cells. PLoS One 2014;9(12):e115963.
[13] McIntosh KR, Frazier T, Rowan BG, Gimble JM. Evolution and future pros-
pects of adipose-derived immunomodulatory cell therapeutics. Expet Rev
Clin Immunol 2013;9(2):175e84.
[14] Thirumala S, Gimble JM, Devireddy RV. Cryopreservation of stromal vascular
fraction of adipose tissue in a serum-free freezing medium. J Tissue Eng
Regen Med 2010;4(3):224e32.
[15] Andia I, Maffulli N, Burgos-Alonso N. Stromal vascular fraction technologies
and clinical applications. Expet Opin Biol Ther 2019;19(12):1289e305.
[16] Zimmerlin L, Donnenberg VS, Rubin JP, Donnenberg AD. Mesenchymal
markers on human adipose stem/progenitor cells. Cytometry 2013;83(1):
134e40.
[17] Page MJ, McKenzie JE, Bossuyt PM, et al. The PRISMA 2020 statement: an
updated guideline for reporting systematic reviews. Int J Surg 2021;88:
105906.
[18] Equator Network. About us. Available at: http://www.equator-network.org/
about-us/. Accessed December 1, 2022.
[19] Guyatt GH, Oxman AD, Vist GE, et al. GRADE: an emerging consensus on
rating quality of evidence and strength of recommendations. BMJ
2008;336(7650):924e6.
[20] Schulz KF, Altman DG, Moher D, CONSORT Group. CONSORT 2010 statement:
updated guidelines for reporting parallel group randomised trials. . Pub-
lished 2010 Mar 23 BMJ 2010;340:c332. https://doi.org/10.1136/bmj.c332.
[21] Reeves BC, Gaus W. Guidelines for reporting non-randomised studies. Forsch
Komplementarmed Klass Naturheilkd 2004;11(Suppl 1):46e52.
[22] Agha RA, Sohrabi C, Mathew G, et al. The PROCESS 2020 guideline: updating
consensus preferred reporting of case series in surgery (PROCESS) guidelines.
Int J Surg 2020;84:231e5.
[23] Gagnier JJ, Kienle G, Altman DG, et al. The CARE guidelines: consensus-based
clinical case reporting guideline development. BMJ Case Rep 2013;2013:
bcr2013201554.
Table 12
Model for evaluation and reporting markers of the SVF cells.
Subpopulation Marker (%)
Hematopoietic cells CD45
þ
Monocytes CD45þ/CD14þ/CD206-
Macrophages CD45þ/CD14þ/CD206þ
Progenitor Endothelial Cells CD45-/CD34þ/CD31þ
Pericytes CD45-/CD34-/CD146þ
Supra-adventitial stromal cells
Early mesenchymal stem cells
CD45-/CD34þ/CD31-
CD45-/CD34þ/CD73þ/CD90þ
M.Y. Ferreira, J.C. Carvalho Junior and L.M. Ferreira Regenerative Therapy 24 (2023) 332e342
340
[24] von Elm E, Altman DG, Egger M, et al. The Strengthening the Reporting of
Observational Studies in Epidemiology (STROBE) statement: guidelines for
reporting observational studies. Lancet 2007;370(9596):1453e7.
[25] Morzycki AD, Hudson AS, Samargandi OA, Bezuhly M, Williams JG. Reporting
adverse events in plastic surgery: a systematic review of randomized
controlled trials. Plast Reconstr Surg 2019;143(1):199ee208e.
[26] Brouwers MC, Kerkvliet K, Spithoff K; AGREE Next Steps Consortium. The
AGREE Reporting Checklist: a tool to improve reporting of clinical practice
guidelines [published correction appears in BMJ. 2016 Sep 06;354:i4852].
BMJ. 2016;352:i1152.
[27] Equator Network. Developing a reporting guideline. https://www.equator-
network.org/toolkits/developing-a-reporting-guideline/. [Accessed 1
December 2022].
[28] Maene A, Deniz G, Bouland C, Lagneaux L, Philippart P, Buxant F. Suburethral
implantation of autologous regenerative cells for female stress urinary in-
continence management: results of a pilot study. Eur J Obstet Gynecol
Reprod Biol 2022;278:38e44.
[29] Nilforoushzadeh MA, Heidari-Kharaji M, Alavi S, et al. Transplantation of
autologous fat, stromal vascular fraction (SVF) cell, and platelet-rich plasma
(PRP) for cell therapy of atrophic acne scars: clinical evaluation and bio-
metric assessment. J Cosmet Dermatol 2022;21(5):2089e98.
[30] Guillo L, Grimaud F, Houser F, et al. Three-year outcome of local injection of
autologous stromal vascular fraction cells and microfat in refractory perianal
stulas of Crohn's disease. Stem Cell Res Ther 2022;13(1):67.
[31] Zhang Y, Bi Q, Luo J, Tong Y, Yu T, Zhang Q. The effect of autologous adipose-
derived stromal vascular fractions on cartilage regeneration was quantita-
tively evaluated based on the 3D-FS-SPGR sequence: a clinical trial study.
BioMed Res Int 2022;2022:2777568.
[32] Behrangi E, Moradi S, Ghassemi M, et al. The investigation of the efcacy and
safety of stromal vascular fraction in the treatment of nanofat-treated acne
scar: a randomized blinded controlled clinical trial. Stem Cell Res Ther
2022;13(1):298. Published 2022 Jul 15.
[33] Zhang S, Xu H, He B, et al. Mid-term prognosis of the stromal vascular
fraction for knee osteoarthritis: a minimum 5-year follow-up study. Stem
Cell Res Ther 2022;13(1):105.
[34] Pattayadeekul T, Pawcsuntorn T, Nararatwanchai T. The efcacy and safety of
autologous stromal vascular fraction transplantation for infraorbital skin
rejuvenation: a clinical prospective study. J Cosmet Dermatol 2022;21(1):
220e6.
[35] Kim SJ, Kim MJ, Lee YJ, et al. Innovative method of alopecia treatment by
autologous adipose-derived SVF. Stem Cell Res Ther 2021;12(1):486.
[36] Yin S, Yang X, Bi H, Zhao Z. Combined use of autologous stromal vascular
fraction cells and platelet-rich plasma for chronic ulceration of the diabetic
lower limb improves wound healing. Int J Low Extrem Wounds 2021;20(2):
135e42.
[37] Ahn TH, Lee W, Kim HM, Cho SB, Yang EJ. Use of autologous adipose-derived
stromal vascular fractions in revision rhinoplasty for severe contractures in
asian patients. Plast Reconstr Surg 2021;147(3):401ee11e.
[38] Jeon HJ, Choi DH, Lee JH, et al. A prospective study of the efcacy of cell-
assisted lipotransfer with stromal vascular fraction to correct contour de-
formities of the autologous reconstructed breast. Aesthetic Plast Surg
2021;45(3):853e63.
[39] Garza JR, Campbell RE, Tjoumakaris FP, et al. Clinical efcacy of intra-
articular mesenchymal stromal cells for the treatment of knee osteoar-
thritis: a double-blinded prospective randomized controlled clinical trial. Am
J Sports Med 2020;48(3):588e98.
[40] Simunec D, Salari H, Meyer J. Treatment of grade 3 and 4 osteoarthritis with
intraoperatively separated adipose tissue-derived stromal vascular fraction:
a comparative case series. Cells 2020;9(9):2096.
[41] Bouland C, Meuleman N, Widelec J, et al. Case reports of medication-related
osteonecrosis of the jaw (MRONJ) treated with uncultured stromal vascular
fraction and L-PRF. J Stomatol Oral Maxillofac Surg 2021;122(2):212e8.
[42] Choi UY, Kim KT, Kim KG, et al. Safety and tolerability of stromal vascular
fraction combined with
b
-tricalcium phosphate in posterior lumbar inter-
body fusion: phase I clinical trial. Cells 2020;9(10):2250. Published 2020 Oct
8.
[43] Carstens MH, Zelaya M, Calero D, Rivera C, Correa D. Adipose-derived stro-
mal vascular fraction (SVF) cells for the treatment of non-reconstructable
peripheral vascular disease in patients with critical limb ischemia: a 6-year
follow-up showing durable effects. Stem Cell Res 2020;49:102071.
[44] Lapuente JP, Dos-Anjos S, Bl
azquez-Martínez A. Intra-articular inltration 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(1):137.
[45] Mattei A, Bertrand B, Jouve E, et al. Feasibility of rst injection of autologous
adipose tissue-derived stromal vascular fraction in human scarred vocal
folds: a nonrandomized controlled trial. JAMA Otolaryngol Head Neck Surg
2020;146(4):355e63.
[46] Carstens M, Haq I, Martinez-Cerrato J, Dos-Anjos S, Bertram K, Correa D.
Sustained clinical improvement of Parkinson's disease in two patients with
facially-transplanted adipose-derived stromal vascular fraction cells. J Clin
Neurosci 2020;81:47e51.
[47] Lee HB, Park SW, Kim IK, Kim JH, Kim DY, Hwang KC. Adipose tissue derived
stromal vascular fraction as an adjuvant therapy in stroke rehabilitation:
case reports. Medicine (Baltim) 2020;99(34):e21846.
[48] Tsubosaka M, Matsumoto T, Sobajima S, Matsushita T, Iwaguro H, Kuroda R.
The inuence of adipose-derived stromal vascular fraction cells on the
treatment of knee osteoarthritis. BMC Muscoskel Disord 2020;21(1):207.
[49] Butt G, Hussain I, Ahmad FJ, Choudhery MS. Stromal vascular fraction-
enriched platelet-rich plasma therapy reverses the effects of androgenetic
alopecia. J Cosmet Dermatol 2020;19(5):1078e85.
[50] Moon KC, Chung HY, Han SK, Jeong SH, Dhong ES. Tissue-engineered dermis
grafts using stromal vascular fraction cells on the nose: a retrospective case-
control study. J Plast Reconstr Aesthetic Surg 2020;73(5):965e74.
[51] Lee SY, Shin JE, Kwon H, Choi DH, Kim JH. Effect of autologous adipose-
derived stromal vascular fraction transplantation on endometrial regenera-
tion in patients of asherman's syndrome: a pilot study. Reprod Sci
2020;27(2):561e8.
[52] Duma C, Kopyov O, Kopyov A, et al. Human intracerebroventricular (ICV)
injection of autologous, non-engineered, adipose-derived stromal vascular
fraction (ADSVF) for neurodegenerative disorders: results of a 3-year phase 1
study of 113 injections in 31 patients. Mol Biol Rep 2019;46(5):5257e72.
[53] Chiu CH. Does stromal vascular fraction ensure a higher survival in autolo-
gous fat grafting for breast augmentation? A volumetric study using 3-
dimensional laser scanning. Aesthetic Surg J 2019;39(1):41e52.
[54] Minev BR, Lander E, Feller JF, et al. First-in-human study of TK-positive
oncolytic vaccinia virus delivered by adipose stromal vascular fraction
cells. J Transl Med 2019;17(1):271. Published 2019 Aug 19.
[55] Hong Z, Chen J, Zhang S, et al. Intra-articular injection of autologous adipose-
derived stromal vascular fractions for knee osteoarthritis: a double-blind
randomized self-controlled trial. Int Orthop 2019;43(5):1123e34.
[56] Serrero M, Grimaud F, Philandrianos C, Vis
ee C, Sabatier F, Grimaud JC. Long-
term safety and efcacy of local microinjection combining autologous
microfat and adipose-derived stromal vascular fraction for the treatment of
refractory perianal stula in crohn's disease. Gastroenterology 2019;156(8):
2335e2337.e2.
[57] Calabrese C, Kothari A, Badylak S, et al. Oncological safety of stromal vascular
fraction enriched fat grafting in two-stage breast reconstruction after nipple
sparing mastectomy: long-term results of a prospective study. Eur Rev Med
Pharmacol Sci 2018;22(15):4768e77.
[58] Lee JW, Park SH, Lee SJ, Kim SH, Suh IS, Jeong HS. Clinical impact of highly
condensed stromal vascular fraction injection in surgical management of
depressed and contracted scars. Aesthetic Plast Surg 2018;42(6):1689e98.
[59] Anderi R, Makdissy N, Azar A, Rizk F, Hamade A. Cellular therapy with human
autologous adipose-derived adult cells of stromal vascular fraction for alo-
pecia areata. Stem Cell Res Ther 2018;9(1):141. Published 2018 May 15.
[60] Calcagni M, Zimmermann S, Scaglioni MF, Giesen T, Giovanoli P, Fakin RM.
The novel treatment of SVF-enriched fat grafting for painful end-neuromas
of supercial radial nerve. Microsurgery 2018;38(3):264e9.
[61] Bourne DA, Thomas RD, Bliley J, et al. Amputation-site soft-tissue restoration
using adipose stem cell therapy. Plast Reconstr Surg 2018;142(5):1349e52.
[62] Bright B, Bright R, Bright P, Limaye A. Ankylosing spondylitis, chronic fatigue
and depression improved after stromal vascular fraction treatment for
osteoarthritis: a case report. J Med Case Rep 2018;12(1):238.
[63] Mazur S, Zołoci
nska A, Siennicka K, Janik-Kosacka K, Chrapusta A, Pojda Z.
Safety of adipose-derived cell (stromal vascular fraction - SVF) augmentation
for surgical breast reconstruction in cancer patients. Adv Clin Exp Med
2018;27(8):1085e90.
[64] Didangelos T, Koliakos G, Kouzi K, et al. Accelerated healing of a diabetic foot
ulcer using autologous stromal vascular fraction suspended in platelet-rich
plasma. Regen Med 2018;13(3):277e81.
[65] Philandrianos C, Serrero M, Grimaud F, et al. First clinical case report of local
microinjection of autologous fat and adipose-derived stromal vascular frac-
tion for perianal stula in Crohn's disease. Stem Cell Res Ther 2018;9(1):4.
[66] Comella K, Silbert R, Parlo M. Effects of the intradiscal implantation of
stromal vascular fraction plus platelet rich plasma in patients with degen-
erative disc disease [published correction appears in J Transl Med. 2017 May
22;15(1):108] J Transl Med 2017;15(1):12.
[67] Carstens MH, G
omez A, Cort
es R, et al. Non-reconstructable peripheral
vascular disease of the lower extremity in ten patients treated with adipose-
derived stromal vascular fraction cells. Stem Cell Res 2017;18:14e21.
[68] Carstens MH, Mendieta M, P
erez C, Villareal E, Garcia R. Assisted salvage of
ischemic fasciocutaneous ap using adipose-derived mesenchymal stem
cells: in-situ revascularization. Aesthetic Surg J 2017;37(suppl_3):S38e45.
[69] Konstantinow A, Arnold A, Djabali K, et al. Therapy of ulcus cruris of venous
and mixed venous arterial origin with autologous, adult, native progenitor
cells from subcutaneous adipose tissue: a prospective clinical pilot study.
J Eur Acad Dermatol Venereol 2017;31(12):2104e18.
[70] Nguyen PD, Tran TD, Nguyen HT, et al. Comparative clinical observation of
arthroscopic microfracture in the presence and absence of a stromal vascular
fraction injection for osteoarthritis. Stem Cells Transl Med 2017;6(1):187e95.
[71] Daumas A, Magalon J, Jouve E, et al. Long-term follow-up after autologous
adipose-derived stromal vascular fraction injection into ngers in systemic
sclerosis patients. Curr Res Transl Med 2017;65(1):40e3.
[72] Signor KC, Steiner D, Roth D, Batista Júnior ML, Souza LG, Santos KB. Stromal
vascular fraction, a new therapy in photoaging: a comparative controlled
study. Surgical &Cosmetic Dermatology 2016;8(2).
[73] Guillaume-Jugnot P, Daumas A, Magalon J, et al. Autologous adipose-derived
stromal vascular fraction in patients with systemic sclerosis: 12-month
follow-up. Rheumatology 2016;55(2):301e6.
M.Y. Ferreira, J.C. Carvalho Junior and L.M. Ferreira Regenerative Therapy 24 (2023) 332e342
341
[74] Comella K, Parcero J, Bansal H, et al. Effects of the intramyocardial implan-
tation of stromal vascular fraction in patients with chronic ischemic car-
diomyopathy. J Transl Med 2016;14(1):158. Published 2016 Jun 2.
[75] Haahr MK, Jensen CH, Toyserkani NM, et al. Safety and potential effect of a
single intracavernous injection of autologous adipose-derived regenerative
cells in patients with erectile dysfunction following radical prostatectomy:
an open-label phase I clinical trial. EBioMedicine 2016;5:204e10.
[76] Toyserkani NM, Jensen CH, Sheikh SP, Sørensen JA. Cell-assisted lipotransfer
using autologous adipose-derived stromal cells for alleviation of breast
cancer-related lymphedema. Stem Cells Transl Med 2016;5(7):857e9.
[77] Jung HK, Kim CH, Song SY. Prospective 1-year follow-up study of breast
augmentation by cell-assisted lipotransfer. Aesthetic Surg J 2016;36(2):
179e90.
[78] Fodor PB, Paulseth SG. Adipose derived stromal cell (ADSC) injections for
pain management of osteoarthritis in the human knee joint. Aesthetic Surg J
2016;36(2):229e36.
[79] Prins HJ, Schulten EA, Ten Bruggenkate CM, Klein-Nulend J, Helder MN. Bone
regeneration using the freshly isolated autologous stromal vascular fraction
of adipose tissue in combination with calcium phosphate ceramics. Stem
Cells Transl Med 2016;5(10):1362e74.
[80] Iddins CJ, Cohen SR, Goans RE, et al. Case report: industrial X-ray injury
treated with non-cultured autologous adipose-derived stromal vascular
fraction (SVF). Health Phys 2016;111(2):112e6.
[81] Tissiani LA, Alonso N. A prospective and controlled clinical trial on stromal
vascular fraction enriched fat grafts in secondary breast reconstruction. Stem
Cell Int 2016;2016:2636454.
[82] Aronowitz JA, Lockhart RA, Hakakian CS, Hicok KC. Clinical safety of stromal
vascular fraction separation at the point of care. Ann Plast Surg 2015;75(6):
666e71.
[83] Granel B, Daumas A, Jouve E, et al. Safety, tolerability and potential efcacy of
injection of autologous adipose-derived stromal vascular fraction in the
ngers of patients with systemic sclerosis: an open-label phase I trial. Ann
Rheum Dis 2015;74(12):2175e82.
[84] Wang L, Luo X, Lu Y, Fan ZH, Hu X. Is the resorption of grafted fat reduced in
cell-assisted lipotransfer for breast augmentation? Ann Plast Surg
2015;75(2):128e34.
[85] Borowski DW, Gill TS, Agarwal AK, Tabaqchali MA, Garg DK, Bhaskar P.
Adipose tissue-derived regenerative cell-enhanced lipolling for treatment
of cryptoglandular stulae-in-ano: the ALFA technique. Surg Innovat
2015;22(6):593e600.
[86] Schendel SA. Enriched autologous facial fat grafts in aesthetic surgery: 3D
volumetric results. Aesthetic Surg J 2015;35(8):913e9.
[87] Pak J, Lee JH, Jeon JH, Lee SH. Complete resolution of avascular necrosis of the
human femoral head treated with adipose tissue-derived stem cells and
platelet-rich plasma. J Int Med Res 2014;42(6):1353e62.
[88] Pak J, Lee JH, Lee SH. A novel biological approach to treat chondromalacia
patellae. PLoS One 2013;8(5):e64569.
[89] Pak J, Chang JJ, Lee JH, Lee SH. Safety reporting on implantation of autologous
adipose tissue-derived stem cells with platelet-rich plasma into human
articular joints. BMC Muscoskel Disord 2013;14:337.
[90] DeTora LM, Toroser D, Sykes A, et al. Good publication practice (GPP)
guidelines for company-sponsored biomedical research: 2022 update. Ann
Intern Med 2022;175(9):1298e304.
[91] AIOM eAssociazone Italiana Oncologia Medica. Follow-up schedule guide-
line. https://www.aiom.it/. [Accessed 1 December 2022].
[92] European Medicine Agency. ICH E6 (R2) Good clinical practice - scientic
guideline. https://www.ema.europa.eu/en/ich-e6-r2-good-clinical-practice-
scientic-guideline#current-versionsection. [Accessed 1 December 2022].
[93] Department of Social and Health Services eWashington State. Moses e
monitoring of side effects scale. Available at: https://www.dshs.wa.gov/sites/
default/les/forms/pdf/10-334.pdf, . [Accessed 1 December 2022].
[94] MedDRA MSSO. Medical dictionary for regulatory activities. Version 21.0.
https://www.meddra.org/. [Accessed 1 December 2022].
[95] National Institutes of Health, Division of Cancer Treatment and
Diagnosis. Common Terminology criteria for adverse events (CTCAE)
version 5.0. Published, https://ctep.cancer.gov/protocoldevelopment/
electronic_applications/docs/ctcae_v5_quick_reference_5x7.pdf. [Accessed 1
December 2022].
[96] Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for dening mul-
tipotent mesenchymal stromal cells. The International Society for Cellular
Therapy position statement. Cytotherapy 2006;8(4):315e7.
[97] Bourin P, Bunnell BA, Casteilla L, et al. Stromal cells from the adipose tissue-
derived stromal vascular fraction and culture expanded adipose tissue-
derived stromal/stem cells: a joint statement of the International Federa-
tion for Adipose Therapeutics and Science (IFATS) and the International
Society for Cellular Therapy (ISCT). Cytotherapy 2013;15(6):641e8.
[98] Cond
e-Green A, Kotamarti VS, Sherman LS, et al. Shift toward mechanical
isolation of adipose-derived stromal vascular fraction: review of upcoming
techniques. Plast Reconstr Surg Glob Open 2016;4(9):e1017.
[99] 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.
[100] 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(1):e261e74.
[101] Aronowitz JA, Ellenhorn JDI. Adipose stromal vascular fraction isolation: a
head-to-head comparison of four commercial cell separation systems. Plast
Reconstr Surg 2013;132(6):932ee9e.
[102] Zanata F, Bowles A, Frazier T, et al. Effect of cryopreservation on human
adipose tissue and isolated stromal vascular fraction cells: in vitro and
in vivo analyses. Plast Reconstr Surg 2018;141(2):232ee43e.
[103] Tonnard P, Verpaele A, Carvas M. Fat grafting for facial rejuvenation with
nanofat grafts. Clin Plast Surg 2020;47(1):53e62.
[104] Lamblet H, Ferreira LM. Fat obtained from plastic surgery procedures-stem
cells derived from adipose tissue and their potential in technological inno-
vation: a narrative literature review and perspective on dissociative
methods. Eur J Plast Surg 2022;45(5):701e31.
[105] Thomas-Porch C, Li J, Zanata F, et al. Comparative proteomic analyses of
human adipose extracellular matrices decellularized using alternative pro-
cedures. J Biomed Mater Res 2018;106(9):2481e93.
[106] Gimble JM, Ray SP, Zanata F, et al. Adipose derived cells and tissues for
regenerative medicine. ACS Biomater Sci Eng 2017;3(8):1477e82.
[107] Ghiasloo M, De Wilde L, Singh K, et al. A systematic review on extracellular
vesicles-enriched fat grafting: a shifting paradigm. Aesthetic Surg J
2021;41(11):NP1695e705.
[108] Vyas KS, Vasconez HC, Morrison S, et al. Fat graft enrichment strategies: a
systematic review. Plast Reconstr Surg 2020;145(3):827e41.
[109] Tauer JT, Al-Jallad H, Umebayashi M, et al. Characterization and functional
analysis of the adipose tissue-derived stromal vascular fraction of pediatric
patients with osteogenesis imperfecta. Sci Rep 2022;12(1):2414.
[110] Rohrich RJ, So Weinstein A. You want to improve your plastic surgery pa-
pers? Introducing PRS' friendly EQUATOR reporting guidelines. Plast
Reconstr Surg 2015;136(1):205e8.
[111] Ascha M, Ascha MS, Gatherwright J. The importance of reproducibility in
plastic surgery research. Plast Reconstr Surg 2019;144(1):242e8.
[112] Rohrich RJ, Cohen JM, Savetsky IL, Avashia YJ, Chung KC. Evidence-Based
medicine in plastic surgery: from then to now. Plast Reconstr Surg
2021;148(4):645ee9e.
[113] Kelley BP, Chung KC. Developing, conducting, and publishing appropriate
systematic review and meta-analysis articles. Plast Reconstr Surg
2018;141(2):516e25.
M.Y. Ferreira, J.C. Carvalho Junior and L.M. Ferreira Regenerative Therapy 24 (2023) 332e342
342
... Preclinical models have shown promising results regarding the therapeutic potential of SVF, which is rich in AD-MSCs, in terms of cartilage regeneration [25]. Clinical studies have also begun to explore the use of SVF, with some reporting significant improvements in pain, ROM, functional rating, and functional outcome scores in OA patients receiving intra-articular SVF injections [26][27][28][29]. These positive outcomes were often accompanied by low donor-site morbidity and minimal adverse events [26]. ...
... Clinical studies have also begun to explore the use of SVF, with some reporting significant improvements in pain, ROM, functional rating, and functional outcome scores in OA patients receiving intra-articular SVF injections [26][27][28][29]. These positive outcomes were often accompanied by low donor-site morbidity and minimal adverse events [26]. Comparative studies have further supported the therapeutic potential of SVF, highlighting its superiority over other treatments such as hyaluronic acid in terms of pain relief, functionality, and structural improvement [30,31]. ...
Article
Full-text available
BACKGROUND Current osteoarthritis (OA) treatments focus on symptom relief without addressing the underlying disease process. In regenerative medicine, current treatments have limitations. In regenerative medicine, more research is needed for intra-articular stromal vascular fraction (SVF) injections in OA, including dosage optimization, long-term efficacy, safety, comparisons with other treatments, and mechanism exploration. AIM To compare the efficacy of intra-articular SVF with corticosteroid (ICS) injections in patients with primary knee OA. METHODS The study included 50 patients with Kellgren-Lawrence grades II and III OA. Patients were randomly assigned (1:1) to receive either a single intra-articular SVF injection (group A) or a single intra-articular ICS (triamcinolone) (group B) injection. Patients were followed up at 1, 3, 6, 12, and 24 months. Visual analog score (VAS) and International Knee Documentation Committee (IKDC) scores were administered before the procedure and at all follow-ups. The safety of SVF in terms of adverse and severe adverse events was recorded. Statistical analysis was performed with SPSS Version 26.0, IBM Corp, Chicago, IL, United States. RESULTS Both groups had similar demographics and baseline clinical characteristics. Follow-up showed minor patient loss, resulting in 23 and 24 in groups A and B respectively. Group A experienced a notable reduction in pain, with VAS scores decreasing from 7.7 to 2.4 over 24 months, compared to a minor reduction from 7.8 to 6.2 in Group B. This difference in pain reduction in group A was statistically significant from the third month onwards. Additionally, Group A showed significant improvements in knee functionality, with IKDC scores rising from 33.4 to 83.10, whereas Group B saw a modest increase from 36.7 to 45.16. The improvement in Group A was statistically significant from 6 months and maintained through 24 months. CONCLUSION Our study demonstrated that intra-articular administration of SVF can lead to reduced pain and improved knee function in patients with primary knee OA. More adequately powered, multi-center, double-blinded, randomised clinical trials with longer follow-ups are needed to further establish safety and justify its clinical use.
... The MSC and EPC coculture enhances both angiogenesis and osteogenesis (Sun et al., 2016). The AT-SVF, cell source we used in the current study, contains these two cell populations and is considered safe to use in clinics (Ferreira et al., 2023). In 2013, Jurgens et al. showed in an animal model that the osteogenic potential of uncultured SVF was superior to the osteogenic potential of AT-MSC (Jurgens et al., 2013). ...
Article
Full-text available
Introduction: Medication-related osteonecrosis of the jaw (MRONJ) poses a significant challenge considering the absence of a “gold standard” treatment. Cell-based therapy and tissue engineering offer promising therapeutic alternatives. This study aimed to harness the regenerative properties of adipose-tissue stromal vascular fraction (AT-SVF) and leukocyte-platelet-rich fibrin (L-PRF) for MRONJ treatment. AT-SVF contains mesenchymal stromal cells (MSC) and endothelial progenitor cells (EPC), which promote bone formation, while the L-PRF scaffold can serve as a three-dimensional scaffold for the AT-SVF and support tissue healing through growth factor release. Materials and methods: The protocol involved applying autologous AT-SVF within an L-PRF matrix following surgical debridement. Age, gender, body mass index, comorbidities, underlying oncological condition, prescribed antiresorptive treatment: BP or DMB, antiresorptive treatment duration, antiresorptive treatment potential discontinuation, number of MRONJ lesion, MRONJ location, MRONJ stage, MRONJ trigger factor were assessed for each patient. Patients underwent the procedure and were monitored for a minimum of 6 months based on clinical, biological and medical imaging criteria. Results: Nine patients, with a total of ten MRONJ lesions, participated in the study. Six patients were female, and three were male, with a mean age of 68 ± 8 years. Four patients had multiple myeloma (MM), three had metastatic breast cancer, and two had metastatic prostate cancer. Seven MRONJ cases were classified as stage II, and three were classified as stage III. Soft tissue completely healed within a month after treatment in nine cases, with no clinical improvement observed in the remaining case. During follow-up, no sign of MRONJ recurrence was observed. Tridimensional medical imaging revealed bone healing 6 months after the surgical procedure. Immunophenotyping confirmed the presence of MSC and EPC in the AT-SVF: 12,6 ± 4,5% CD31⁺, 20.5 ± 7,8% CD34⁺, 34,4 ± 7,3% CD146⁺ and 54,6 ± 7,4% CD45⁺. Conclusion: This prospective study introduces a potential new treatment approach for MRONJ using autologous AT-SVF within an L-PRF scaffold. Our results are encouraging and suggest the need for further investigation with a larger patient cohort to better understand the underlying mechanisms.
Article
Full-text available
Background Acne is the most common skin disorder which is known as a chronic inflammatory disease with psychological burden and reduced quality of life. Adipose tissue-derived stromal vascular fraction (SVF) is recognized as a source of regenerative cells and improves the quality of skin by increasing collagen content. To date, a few studies have been performed on the therapeutic role of SVF in the treatment of acne scars. Methods This randomized, single-blinded clinical trial was performed on 7 patients with acne scars. In all patients, the initial grade of acne (volume, area and depth) was evaluated and ultrasound of the relevant scar was performed to evaluate neocollagenesis. As a spilt face study, for treating the scars, we used nanofat subcutaneously on one side of the face (control group) and combination of nanofat subcutaneously and SVF intradermally on the opposite side (intervention group). The patients were evaluated for severity of acne by visioface after one month, also for thickness of epidermis and dermis by ultrasound after one month and three months. Results All of the apparent findings of scars improved in two groups after one month, but these changes were significant just for the group treated with SVF ( p value < 0.05). Epidermal, dermal and complete thicknesses during the first month in both control and intervention groups were significantly increased ( p value < 0.05) but between the first and third months, there was no significant difference in the variables ( p value > 0.05). The findings showed that dermal and complete thicknesses of the skin in the first month were different between two groups significantly ( p value: 0.042 and 0.040, respectively). Conclusion The use of SVF in the treatment of patients with acne scars accelerates the improvement of volume, area and depth of the scar by increasing collagen content and the dermal thickness, so it can be used as a potentially effective treatment for these patients.
Article
Full-text available
In this review, a summary of the rich history of autologous fat grafting is provided, and a comprehensive summary of the science and theory behind autologous adipocyte transplantation, as well as the techniques commonly used is described. These include recipient site preparation, harvesting, processing, and engraftment. In addition, important considerations for preoperative and postoperative management are discussed to maximize graft retention. Special considerations in grafting to the breast, face, and buttocks are also summarized.
Article
Full-text available
Background The short-term safety and efficacy of stromal vascular fraction (SVF) in treating knee osteoarthritis (KOA) have been extensively studied but the mid-term and long-term prognoses remain unknown. Methods 126 KOA patients were recruited and randomly assigned to SVF group and hyaluronic acid (HA) group (control group). The scores of visual analogue scale (VAS) and the Western Ontario and McMaster University Osteoarthritis Index (WOMAC) were assessed and compared between the two groups 1, 2, 3, and 5 years after treatment. The endpoint was defined as surgeries related to KOA or clinical scores exceeding the patient acceptable symptom state (PASS). Results The VAS and WOMAC scores in the SVF group were significantly better than those in the HA group during the 5-year follow-up after treatment. The average responsive time to SVF treatment (61.52 months) was significantly longer than HA treatment (30.37 months). The adjusted Cox proportional hazards model showed that bone marrow lesion (BML) severity, body mass index (BMI) and treatment were independent risk factors and that the use of SVF reduced the risk of clinical failure by 2.602 times. The cartilage volume was reduced in both the SVF and control groups at 5 years but reduced less in the SVF group. Conclusions Up to 5 years after SVF treatment, acceptable clinical state was present for approximately 60% of patients. BML severity and BMI were independent predictors of the prognosis. Trial Registry : This study was retrospectively registered at Chinses Clinical Trial Registry with identifier ChiCTR2100052818 and was approved by ethics committee of the First Affiliated Hospital of Zhejiang Chinese Medical University, number 2013-X-063.
Article
Full-text available
Background Throughout its illustrious history, plastic surgery has searched for novel regenerative therapies and procedures. Recently, interest has emerged in using adipose tissue-derived stem cells (ASCs) in an ethical, easy, and reproducible manner. ASCs are generally not administered alone but as a constituent of the stromal vascular fraction (SVF) in clinical practice. Herein, we searched for innovative fat collection and ASC isolation technologies and applications and evaluated each study’s relevance to plastic surgery. Methods A narrative literature review was carried out using the MEDLINE/PubMed databases. Studies published from January 1993 to August 2020 and written in English, Portuguese, or Spanish were considered. Results The selection process yielded 33 articles for subsequent review, involving exploratory, selective, and interpretive reading, material choice, and text analysis. Twenty-three articles employed enzymatic dissociation methods to isolate ASCs, and 25 employed liposuction as the plastic surgery technique. Moreover, articles describing new devices (n = 2), techniques (n = 4), computational models (n = 1), tissue scaffolds (n = 21), and therapies and/or treatments (n = 5) were identified. Conclusions Given the importance of fat tissue for plastic surgery purposes, innovative ASC isolation and liposuction technologies could change how the surgeon conducts surgeries and improve surgical outcomes. Furthermore, many articles investigating tissue scaffolds demonstrate the importance of this area of research and development in plastic surgery and regenerative medicine. Continued efforts in the identified research areas will eventually bring in vivo human plastic surgery applications and regenerative medicine into the operating room. Level of evidence: Not gradable.
Article
Full-text available
Pediatric patients with Osteogenesis Imperfecta (OI), a heritable connective tissue disorder, frequently suffer from long bone deformations. Surgical correction often results in bone non-unions, necessitating revision surgery with autogenous bone grafting using bone-marrow-derived stem cells (BM-SC) to regenerate bone. BM-SC harvest is generally invasive and limited in supply; thus, adipose tissue's stromal vascular fraction (SVF) has been introduced as an alternative stem cell reservoir. To elucidate if OI patients' surgical site dissected adipose tissue could be used as autologous bone graft in future, we investigated whether the underlying genetic condition alters SVF's cell populations and in vitro differentiation capacity. After optimizing SVF isolation, we demonstrate successful isolation of SVF of pediatric OI patients and non-OI controls. The number of viable cells was comparable between OI and controls, with about 450,000 per gram tissue. Age, sex, type of OI, disease-causing collagen mutation, or anatomical site of harvest did not affect cell outcome. Further, SVF-containing cell populations were similar between OI and controls, and all isolated SVF's demonstrated chondrogenic, adipogenic, and osteogenic differentiation capacity in vitro. These results indicate that SVF from pediatric OI patients could be used as a source of stem cells for autologous stem cell therapy in OI.
Article
Full-text available
Perianal fistulas in Crohn's disease are frequent and disabling, with a major impact on patients' quality of life. Cell-based therapy using mesenchymal stem cells represents new hope for these patients, but long-term efficacy remains challenging. In a pilot study, including patients with refractory complex perianal fistulas, autologous adipose-derived stromal vascular fraction (ADSVF) combined with microfat achieved combined remission in 60% of cases, with a good safety profile at 1 year. The purpose of this study is to assess whether these results were maintained at longer term. The safety and efficacy data of the ten patients were evaluated retrospectively 3 years after injection on the basis of clinical and radiological data. MRI were analysed according to the MAGNIFI-CD score. No adverse event was attributed to the experimental stem-cell treatment. Combined remission was achieved in 7 patients (70%) and associated with a significant improvement in the MAGNIFI-CD MRI score. In conclusion, the safety and efficacy of ADSVF and microfat injection in Crohn's disease fistulas were maintained at 3 years, demonstrating that this innovative strategy is effective in producing a long-lasting healing effect. The ongoing multicentre randomized placebo-controlled trial (NCT04010526) will be helpful to define the place for this approach in the current therapeutic arsenal.
Article
Full-text available
Background: Numerous reports confirmed the safety and clinical efficacy of autologous adipose-derived stromal vascular fractions (SVF), which have recently been used to treat osteoarthritis (OA). However, there is still no consensus as to whether SVF can promote cartilage regeneration. Herein, the purpose of our study was to evaluate the effectiveness of SVF versus hyaluronic acid (HA) in cartilage regeneration by establishing a cartilage model based on the three-dimensional fat-suppressed spoiled gradient recalled echo (3D-FS-SPGR) sequence. Methods: Patients with symptomatic OA were recruited in our research, who were randomized into two groups. Meanwhile, patients in Kellgren-Lawrence (K-L) grades 2 and 3 were distinguished in each group. In the test group, patients received SVF injections of the knee, while patients in the control group received the same dose of HA. Each patient underwent the 3D-FS-SPGR sequence to establish a cartilage model at baseline, 6 months, and 12 months, respectively. The cartilage was characterized into six regions, and relevant parameters of the cartilage model were counted. Clinical and radiographic scores were recorded in one-year follow-up. Results: In all regions, the thickness and volume of cartilage defect and the volume of healthy cartilage were improved to some extent in the test group, especially the medial femoral condyle (MF) and medial tibial condyle (MT). In grades 2 and 3, the thickness and volume of cartilage defect decreased by 0.92 ± 0.18 mm and 1.03 ± 0.23 mm and 84.00 ± 32.30 mm3 and 130.30 ± 49.56 mm3 in MF and by 0.96 ± 0.22 mm and 0.99 ± 0.14 mm and 64.18 ± 21.40 mm3 and 95.11 ± 19.93 mm3 in MT, respectively. No such phenomenon was observed in the control group. Meanwhile, the SVF-treated knees showed significant improvement in clinical and radiographic scores at 12 months. Nevertheless, these scores of the control group became worse at 12-month follow-up visit. Conclusion: Taken together, this study shows that intra-articular injection of SVF markedly improved the clinical symptoms without adverse events, thereby repairing the damaged articular cartilage through cartilage regeneration.
Article
Objectives To assess the feasibility and the safety of treating female stress urinary incontinence (SUI) with suburethral implantation of a mixture of the stromal vascular fraction from adipose tissue and leukocyte-and platelet-rich-fibrin. Methods Patients with SUI were treated with a mixture of stromal vascular fraction and leukocyte-and platelet-rich fibrin. The stromal vascular fraction was obtained from enzymatic digestion of autologous adipose-tissue and added to an leukocyte-and platelet-rich-fibrin membrane. The mixture was transvaginally implanted into the suburethral area. A fraction of the Stromal vascular fraction sample was used for cellular characterization. Patients were followed for 9 months. Every 3 months, the patients were clinically evaluated with a cough- stress test and a validated-questionnaire. An MRI was performed preoperatively and 3 months after the procedure to assess tissue changes. Results Ten patients received the surgical procedure. The validated-questionnaire revealed a subjective SUI improvement in nine patients 3 months after the operation and in seven patients 9 months after the operation. Eight, six, and four patients achieved a negative cough-stress test 3, 6 and 9 months post-injection, respectively. Flow cytometric analysis of stromal vascular fraction cell phenotypes revealed predominantly mesenchymal and endothelial cell heterogeneity. In total, we injected 0,18×10⁶ to 13,6×10⁶ cells. No adverse events were observed peri- or postoperatively. Conclusion These preliminary results suggest that the suburethral implantation of a combination of SVF and L-PRF is a feasible and safe modality for treating female SUI. However, evidence is lacking and further research are needed to clarify the respective roles of SVF and L-PRF in female SUI treatment.
Article
These updated Good Publication Practice (GPP) guidelines include recommendations for publishing company-sponsored biomedical research. The GPP guidelines apply to peer-reviewed or peer-oriented biomedical publications, such as manuscripts, meeting presentations, posters, and abstracts, as well as enhanced content, such as plain-language summaries. The current GPP guidelines incorporate guidance on ethics and transparency as well as the planning, development, review, and approval of biomedical publications and policies and procedures that describe these practices. Supplemental materials lay out processes for steering committees, publication plans, publication working groups, determining authorship, and documentation. Information about new topics, such as alliances and working with patients, has been included where appropriate within these supplemental materials. Incorporating the principles and best practices presented in these GPP guidelines will result in increased transparency and a firm ethical footing. This guidance is also intended to enable the compliant incorporation of new and emerging publication tools for the ethical publication of company-sponsored research.
Article
Evidence-based medicine, as described by Dr. Sackett, is defined as the "conscientious, explicit, and judicious use of current best evidence, combined with individual clinical expertise and patient preferences and values, in making decisions about the care of individual patients." In the late 2000s, seminal articles in Clinics in Plastic Surgery and Plastic and Reconstructive Surgery introduced evidence-based medicine's role in plastic surgery and redefined varying levels of evidence. The American Society of Plastic Surgery sponsored the Colorado Springs Evidence-Based Medicine Summit that set forth a consensus statement and action plan regarding the increased incorporation of evidence-based medicine into the field; this key meeting ushered a new era among plastic surgeons worldwide. Over the past decade, Plastic and Reconstructive Surgery has incorporated evidence-based medicine into the Journal through an increase in articles with level I and II evidence, new sections of the Journal, and the introduction of validated tools to help authors perform prospective and randomized studies that ultimately led to best practices used today. Plastic surgery is a specialty built on problem-solving and innovation, values starkly in-line with evidence-based medicine. Evidence-based medicine is becoming more ingrained in our everyday practice and plastic surgery culture; however, we must work actively to ensure that we continue this trend. In the next decade, we will possibly see that level I and II evidence articles start to inhabit many of our journal issues.