Systematic Reviews Open Access
Copyright ©The Author(s) 2020. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Jul 26, 2020; 12(7): 676-687
Published online Jul 26, 2020. doi: 10.4252/wjsc.v12.i7.676
Application and prospect of adipose stem cell transplantation in treating lymphedema
Zhu-Jun Li, Elan Yang, Yun-Zhu Li, Zheng-Yun Liang, Jiu-Zuo Huang, Nan-Ze Yu, Xiao Long, Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
ORCID number: Zhu-Jun Li (0000-0003-1978-432X); Elan Yang (0000-0003-3516-0576); Yun-Zhu Li (0000-0003-1667-7908); Zheng-Yun Liang (0000-0003-4907-8020); Jiu-Zuo Huang (0000-0002-0458-9006); Nan-Ze Yu (0000-0002-6296-6236); Xiao Long (0000-0003-0136-2508).
Author contributions: Li ZJ and Long X designed the report; Li ZJ, Li YZ, Liang ZY, Huang JZ, and Yu NZ collected and evaluated the data; Li ZJ and Yang E analyzed the data and wrote the paper.
Conflict-of-interest statement: All the authors declare that they have no competing interests.
PRISMA 2009 Checklist statement: The authors have read the PRISMA 2009 Checklist, and the manuscript was prepared and revised according to the PRISMA 2009 Checklist.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Xiao Long, MD, PhD, Professor, Surgeon, Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital of Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1, Shuaifuyuan, Dongcheng District, Beijing 100730, China. pumclongxiao@126.com
Received: February 18, 2020
Peer-review started: February 18, 2020
First decision: April 25, 2020
Revised: May 8, 2020
Accepted: May 28, 2020
Article in press: May 28, 2020
Published online: July 26, 2020

Abstract
BACKGROUND

Lymphedema is a chronic, debilitating and incurable disease that affects 0.13%-2% of the global population. Emerging evidence indicates that adipose-derived stem cells (ADSCs) might serve as suitable seed cells for lymphatic tissue engineering and lymphedema therapy.

AIM

To summarize applications of ADSCs for treating lymphedema in both animal studies and clinical trials.

METHODS

A systematic search was performed on four databases – PubMed, Clinicaltrials.gov, the evidence-based Cochrane Library, and OVID – using the following search string: (“lymphedema” or “lymphoedema” or “lymphangiogenesis”) and (“adipose-derived stem cells” or “adipose-derived stromal cells” or “adipose-derived regenerative cells”). A manual search was performed by skimming the references of relevant studies. Animal studies and clinical trials using adipose-derived cells for the treatment of any kind of lymphedema were included.

RESULTS

A total of eight research articles published before November 2019 were included for this analysis. Five articles focused on animal studies and another three focused on clinical trials. ADSC transplantation therapy was demonstrated to be effective against lymphedema in all studies. The animal studies found that coadministration of ADSCs and controlled-release vascular endothelial growth factor-C or platelet-rich plasma could improve the effectiveness of ADSC therapy. Three sequential clinical trials were conducted on breast cancer-related lymphedema patients, and all showed favorable results.

CONCLUSION

ADSC-based therapy is a promising option for treating lymphedema. Large-scale, multicenter randomized controlled trials are needed to develop more effective and durable therapeutic strategies.

Key Words: Lymphedema, Adipose-derived stem cells, Animal model, Clinical trial, Vascular endothelial growth factor-C, Lymphangiogenesis

Core tip: Lymphedema is a growing global health problem. Adipose-derived stem cells might serve as suitable seed cells for tissue engineering of lymphatic vessels in vitro and in vivo. A systematic search of publications on the application of adipose-derived stem cells in the treatment of lymphedema identified five animal studies and three clinical trials. All eight studies showed improvement of lymphedema after treatment with adipose-derived stem cells. Animal studies conducted with acute lymphedema mouse models provided data for finding the proper dose and methods of administration. Clinical trials were conducted on breast cancer-related lymphedema patients and are important references for further application.



INTRODUCTION

As the second circulatory system of the body, the lymphatic system functions to transport tissue fluid in the interstitial space back to the venous circulation system and maintain fluid homeostasis. Hypoplasia or dysfunction of lymphatic vasculature may result in “lymphedema”[1], which is characterized by retention of lymphatic fluid in the interstitial space, leading to a series of pathological changes, including tissue swelling, chronic inflammation, lipid deposition, and tissue fibrosis[2].

There are two types of lymphedema according to etiology[3]. Primary lymphedema comes from developmental or congenital abnormalities of the lymphatic system resulting in dysfunctional lymphatics, which could be symptomatic at birth or more commonly in adolescence. Secondary lymphedema is more common and results from trauma, obstruction, surgery, or infection involving the lymphatic system. Up to 250 million people in developing countries suffer from lymphedema, with the parasitic disease filariasis as the most prevalent cause[4]; lymphadenectomy and radiation therapy for cancer treatment are usually the major causes of lymphedema in developed countries.

Chronic lymphedema affects 0.13%-2% of the global population[5]. It is estimated that one out of six patients undergoing treatment for a solid tumor will eventually develop lymphedema[6]. For patients with breast cancer, 24%-49% of those receiving mastectomy will develop upper extremity lymphedema[7]. The persistence of the disease, burden of treatment, and likelihood of progression press heavy medical and socioeconomic burdens onto patients. Therefore, development of effective therapies for lymphedema is of vital importance.

Lymphedema can be treated conservatively, surgically, or by a combination of them[2]. Conservative therapies consist of complex decongestive therapy, manual lymphatic drainage, and exercise[8]. Surgical therapies include liposuction, wedge resection, Charles procedure (radical excision for limb lymphedema), and lymphatic reconstruction or bypass techniques[9]. However, there is a lack of effective and feasible therapies which could radically cure lymphedema[10]. Therefore, elucidating the underlying pathophysiological mechanisms of lymphedema holds promise for the treatment of lymphedema.

Progenitor or stem cell-based therapies, which treat diseases through regeneration, have represented an alternative treatment method not only for lymphedema but for a wide spectrum of other diseases as well. Due to their abundant resources, easy access, pluripotent capacity, and harboring of few ethical and immunological issues, the adipose-derived stem cells (ADSCs) are considered as one of the most promising seed cell types for regenerative medicine[11-15]. In addition, ADSCs exhibit paracrine immunomodulatory and trophic effects in their local microenvironment. Emerging in vitro studies have investigated the possible mechanisms and benefit of ADSCs in the treatment of lymphedema, such as their capacity for differentiation into lymphatic endothelial cells (LECs)[16,17] and paracrine secretion of cytokines[18], chemokines[19] and exosomes[20], thereby promoting angiogenesis and modulating the immune response. Takeda et al[21] reported that, by secreting lymphangiogenic factors, ADSCs promote proliferation, migration, and tube formation of LECs in vitro. Deng et al[22] demonstrated that overexpression of Prox1 in human ADSCs (via lentiviral vectors) induces the differentiation of human ADSCs into stable lymphatic endothelial-like cells in vitro and that the differentiated cells form tube-like structures (as shown in tube formation assay). Yen et al[23] showed that ADSCs promote lymphangiogenesis under stimulation of vascular endothelial growth factor-C (VEGF-C), a key lymphangiogenic factor, or in response to inhibition of TGF-β1; moreover, stimulation of ADSCs with VEGF-C was found to markedly increase cellular proliferation and cellular survival after in vivo implantation and to induce the expression of podoplanin, a lymphangiogenic cell marker. Sun et al[19] reported that interleukin-7 enhanced the differentiation of ADSCs into LECs via AKT signaling pathways. Most recently, Saijo et al[24] revealed that paracrine effects of ADSCs promoted lymphangiogenesis in irradiated LECs. They reported that coculture with ADSCs and the use of ADSC-conditioned medium improved proliferation, migration, and tube formation of nonirradiated LECs. Furthermore, they demonstrated that irradiated ADSCs can exert similar alleviative effects to irradiated human dermal LECs[24].

Collectively, these research findings have suggested that ADSCs might serve as suitable seed cells for lymphatic tissue engineering and secondary lymphedema therapy. The aim of this review is to systematically summarize the application of ADSCs for lymphedema treatment in animal studies and in clinical trials. In addition, the future perspectives of ADSCs in lymphedema therapy are discussed.

MATERIALS AND METHODS

A systematic search was performed on four databases (PubMed, Clinicaltrials.gov, the evidence-based Cochrane library, and OVID) using the following search string: (“lymphedema” or “lymphoedema” or “lymphangiogenesis”) and (“adipose-derived stem cells” or “adipose-derived stromal cells” or “adipose-derived regenerative cells”). After duplicate removal, all studies were screened based on title and abstract. Furthermore, full-text versions of included studies were read for further evaluation. A manual search was also performed by skimming the references of included studies. The search process is presented in Figure 1.

Figure 1
Figure 1 Search process of articles regarding adipose-derived stem cell-based treatment of lymphedema studied in animal models and clinical trials.

The inclusion criteria were animal studies and clinical trials using adipose-derived cells for treatment of any kind of lymphedema, which had been published no later than November 2019. The exclusion criteria were non-English language, reviews, or in vitro studies. For animal studies, data retrieved were year of publication, first author, type of animal models, cell type used (freshly isolated or culture-expanded as well as autologous or allogeneic), cell dosage, cell characterization (cell count/viability, surface marker analysis, etc.), means and routes of transplantation (alone or in combination with growth factors, scaffolds, etc.), assessment types, and results. For clinical trials, data retrieved were year of publication, country of origin, disease treated, study design (randomized controlled trial, nonrandomized study, or case series/pilot study), number of participants, cell type used (freshly isolated or culture-expanded as well as autologous or allogeneic), cell dosage, cell characterization (cell count/viability, surface marker analysis, etc.), means and routes of transplantation (alone or in combination with growth factors, scaffolds, etc.), assessment types, and outcomes.

RESULTS

A total of eight research articles published before November 2019 met the inclusion criteria for this analysis. These included five articles focused on animal studies (Table 1) and three focused on clinical trials (Table 2).

Table 1 Adipose-derived stem cell-based treatment of lymphedema in animal studies.
YearRef.AnimalLocation and methodsGroupsCell type used for characterization and numberImplantation methodsAssessmentResults
2011Hwang et al[25]6-8-wk-old female BALB/c miceHindlimb, Circumferential incision and electrocautery5 groups (n = 5 for each group): Normal, control, hADSCs, VEGF-C hydrogel, hADSCs/VEGF-C hydrogelPKH-26-labeled hADSCs N/AIn combination with VEGF-C gelatin hydrogel subcutaneous injectionDermal edema depth measurement using Vernier calipersCo-administration of hADSCs and VEGF-C decreased dermal edema depth and increased lymphatic vessel intensity
H&E staining
IFC (LYVE-1) for lymphatic vessel intensity
2012Shimizu et al[26]7-8-wk-old male C57BL/6J miceTail 2 mm-wide circumferential annulus of the skin excision at 10 mm distal to the tail base, excluding a 4 mm2 dermal flap located at the ventral side3 groups (n = 12 for each group): Sham, PBS, and freshly isolated ADRCsFreshly isolated ADRCs 2 × 106Local injectionTail thickness measurementLocal injection of ADRCs significantly reduced lymphedema ADRC implantation accelerated lymphangiogenesis ADRC implantation enhanced recruitment of M2 macrophages, to serve as lymphatic endothelial progenitor cells
IHC analysis (LVYE-1)
IFC staining (LYVE-1, CD11-b, CD163)
2015Ackermann et al[27]10-wk-old male C57BL/6J miceTail Circumferential 5 mm-wide full thickness excision at a 10 mm distance from the base of the tail3 groups: Saline, PRP, and ASCAllogenic 3 passages FACS analyzed (CD31−/CD45−/CD29+/CD90+ cellsInjectionAngiogenesis (anti-CD31 staining)Wounds treated by PRP and ASC healed faster and showed a significantly increased epithelialization
Laser Doppler imaging for microcirculationApplication of PRP induced a significantly increased lymphangiogenesis, while application of ASC did not induce any significant change, in this regard.
Lymphangiogenesis (anti-LYVE1 staining)
Corrosion casting for microvascular architecture
Digital planimetry for wound healing
2015Yoshida et al[28]Male C57BL/6J miceRight hindlimb, 30-gray x irradiation, surgical lymph node dissection, and 2-mm gap creation4 groups (n = 20 for each group with different cell numbers)Allogenic up to 5 passages 0, 106, 105, 104Subcutaneous injectionCircumference measurementNumber of lymphatic vessels significantly increased at 2 wk
Near-infrared video camera for lymphatic flow assessment
IHC for quantitation of lymphatic vessels (LYVE, VEGF-C, VEGFR, and EGFP)No direct detection of ADSCs involving lymphangiogenesis by EGFP at 2 wk or chromosome FISH at 2 wk and 4 wk
XY chromosome FISH analysis
2017Hayashida et al[29]10-wk-old male C57BL/6J miceLeft hindlimb, 30-Gy X-ray irradiation, surgical lymph node dissection, and 5-mm gap creation4 groups (n = 5 for each group): Control, VLNT, ADSCs, VLNT-plus/ADSCs-plusAllogenic 1-3 passages at 104VLNT, subcutaneouslyNear-infrared video camera for lymphatic flow assessmentIncreased number of lymphatic vessels
Water-displacement plethysmometer for hind-paw volumetry testInduced lymphatic flow drainage to the circulatory system
IHC for tissue quantification of lymphatic vessels (LYVE-1, VEGF-C, and VEGF-R3)
B16 mouse melanoma cells for functional analysis of lymphatic vessels and nodes
Table 2 Adipose-derived stem cell treatment of lymphedema in clinical trials.
YearFirst authorStudy typePatientnumber,test/controlEdemalocationCell typeCell numberInjection method,location/depthFollow-up inmoassessmentResults
2016Toyserkani et al[30]Pilot study1/0Upper limbFreshly isolated autologous ADSCs plus fat graft4.07 × 107 plus 10 mL lipoaspirateAxillary region (8 points)4 mo In vitro cell characterization (cell count, viability, and surface marker)Great reduction in symptoms of arm heaviness and tension, reduction in need for compression therapy
Circumference measurementsVolume reduction in affected arm
Dual-energy X-ray absorptiometry scans Adverse eventsNo postoperative complications or adverse events
2017Toyserkani et al[31]Pilot study10/0Upper limbFreshly isolated autologous ADSCs plus fat graft5 × 107 plus 30 mL lipoaspirateAxilla (8 points)6 mo In vitro cell characterization (cell count, viability, and surface marker)A small volume reduction but not significant
Circumference measurementsPatient-reported outcomes improved significantly over time
Dual-energy X-ray absorptiometry scansHalf of the patients reduced their use of conservative management
Patient-reported outcome and safety questionnaire assessmentNo noteworthy serious adverse events
2019Toyserkani et al[32]Pilot study10/0Upper limbFreshly isolated autologous ADSCs plus fat graft5 × 107 plus 30 mL lipoaspirateAxilla (8 points)12 mo In vitro cell characterization (cell count, viability, and surface marker)No significant change in volume
Circumference measurementsPatient-reported outcomes improved significantly over time
Dual-energy X-ray absorptiometry scansFive patients reduced their use of conservative management
Patient-reported outcome and safety questionnaire assessmentQuantitative lymphoscintigraphy showed no improvement on the lymphedema-affected arms
Lymphoscintigraphy changesADRCs were well-tolerated and only minor transient adverse events related to liposuction were noted
Animal studies

A total of five animal studies were included in the analysis. Commonly-used animal models were mouse hindlimb or tail model of lymphedema. The surgical procedures were circumferential incision with or without radiation. The number of cells used for injection varied from 104 to 1010 each time. Various treatments were used in combination with cell injection, including controlled-release VEGF-C, platelet-rich plasma (PRP), or vascularized lymph node transfer. Treatment outcomes were evaluated by circumference, dermal edema depth, imaging (lymphangiography and photodynamic dye), and histochemical and immunohistochemical staining (for CD31, LYVE1, and VEGF receptor).

Hwang et al[25] established a mouse hindlimb model of lymphedema by electrocauterizing the lymph vessels in the thigh following a circumferential incision. In vivo study demonstrated that combination of human (h) ADSCs and VEGF-C hydrogel markedly alleviated dermal edema and increased lymphatic vessel density when compared with results achieved with hADSCs or VEGF-C hydrogel alone at various post-treatment time points (from 3-4 d to 4 wk post-treatment). In addition, the authors demonstrated the existence of hADSCs in all of the implantation sites in the hADSC/VEGF-C group with LECs phenotype. Their results also suggested that, in conjunction with hADSCs, VEGF-C-containing hydrogels could serve as suitable delivery vectors to improve lymphangiogenesis.

Shimizu et al[26] reported on the establishment of a mouse tail model of lymphedema. They made a 2 mm-wide circumferential excision on the skin 10 mm distal to the tail base and excluded a 4 mm2 dermal flap at the ventral side. They indicated that local injection of 2 × 106 freshly isolated autologous ADSCs could reduce lymphedema and accelerate lymphangiogenesis at the congestive lymphedema region. The authors also revealed that ADSCs could release VEGF-C to stimulate lymphangiogenesis and recruit bone marrow-derived M2 macrophages to serve as lymphatic endothelial progenitor cells.

Ackermann et al[27] assessed the effects of PRP and adipose stem cells on angiogenesis, microcirculation, lymphangiogenesis, microvascular architecture, and wound healing in a mouse tail lymphedema model. They found that treatment with PRP and adipose stem cells could accelerate wound healing and increase epithelialization. PRP application induced a remarkably improved lymphangio-genesis. Ultimately, the authors drew the conclusion that PRP and adipose stem cells can affect lymphangiogenesis and lymphedema development.

Yoshida et al[28] reported on the establishment of a mouse right hindlimb secondary lymphedema model, using 30 Gy X-ray irradiation 7 d before surgery to create a circumferential incision and a gap of approximately 2 mm, left open. Circumferential measurement, lymphatic flow assessment, and quantification of lymphatic vessels demonstrated that at 14 d post-treatment of local injection with 106 ADSCs, 105 ADSCs, or 104 ADSCs, the numbers of lymphatic vessels were significantly increased in the transplanted groups; the authors concluded that in secondary lymphedema, ADSCs could increase collecting vessels and reconstruct the lymphatic vascular network. A more recent study from the same research group[29] demonstrated that local implantation of 104 ADSCs in combination with vascularized lymph node transfer decreased tissue volume, increased lymphatic vessels density, and improved the lymphatic function in a slightly different mouse hindlimb model (left vs right hindlimb, 5 mm- vs 2 mm-wide gap, left open).

Clinical trials

A total of three clinical trials were included in the analysis. All these studies were conducted by the same research group in Denmark[25-27], focusing on upper limb and breast cancer-related lymphedema (BCRL), using freshly isolated autologous ADSCs with fat grafting. The follow-up period ranged from 4 mo to 1 year.

In 2016, Toyserkani et al[30] reported a pilot study using ADSC-assisted lipotransfer to treat lymphedema. A 48-year-old female patient developed BCRL after lymphadenectomy and radiation therapy. A total of 4.07 × 107 freshly isolated autologous ADSCs were administered, along with 10 mL of lipoaspirate (for fat-grafting), to the axillary region. At 4 mo post-treatment, the authors noted that the daily symptoms of arm heaviness and tension were greatly relieved, the needs for compression therapy were reduced, and the volume of the affected arm was decreased without postoperative complications or adverse events.

Furthermore, the authors had also performed a larger study to validate the feasibility and safety of this procedure in 2007[31], which was registered at Clinicaltrials.gov under the identifier NCT02592213 at phase 2 stage. In this pilot study, ten BCRL patients were included. Combined with a scar-releasing fat graft (about 30 mL) procedure, approximately 5 × 107 freshly isolated adipose-derived regenerative cells (ADRCs) were injected into the axillary region. During a 6-mo follow-up period, there was a small but not significant volume reduction. Five of the patients showed a reduced need for conservative treatment, and patient-reported follows-up improved significantly over time. Slight, temporary adverse events were observed, but were more likely caused by liposuction procedures rather than ADRC injection. Ultimately, the ADRCs were deemed as well-tolerated.

In 2019, the results of lymphoscintigraphic evaluation with 1-year follow-up were reported[32]. Consistent with the results of the 6-mo follow-up, ADRC injection improved lymphedema, as revealed by patient-reported outcomes without serious adverse events. However, there was no improvement in lymphoscintigraphic evaluation and no change in arm volume after the ADRC treatment. Now, this research group is recruiting patients for a randomized phase 3 trial, which is registered at Clinicaltrials.gov under the identifier NCT03776721. This study started December 17, 2018 and the estimated study completion date is September 1, 2021, designed to recruit 80 participants with a parallel assignment to evaluate the efficacy and safety of implantation of freshly isolated adipose-derived stromal cells in combination with fat grafting at the affected axillary region.

DISCUSSION

Lymphedema, characterized by tissue swelling, lipid deposition, and fibrosis due to excess accumulation of interstitial fluid and inadequate lymphatic drainage, affects 0.13%-2% of the global population[5] and remains a chronic, debilitating and incurable disease. Stem cell-based regenerative medicine has shown great promise for refractory diseases, such as inflammatory bowel diseases[33], heart failure[34], osteoarthritis[35], rheumatoid arthritis[36], and graft-versus-host disease[37]. With the properties of self-renewal, multipotential differentiation, paracrine, immunomodulatory, and trophic effects, and low immunogenicity alongside their practical advantages, ADSCs have become one of the most promising candidates for regenerative medicine.

ADSCs are isolated from the aqueous fraction – known as the stromal vascular fraction (SVF) – by means of enzymatic digestion of lipoaspirate (liposuction product)[38,39]. As the major source of ADSCs, SVF is a heterogeneous cell group composed of ADSCs, endothelial precursor cells, endothelial cells, macrophages, smooth muscle cells, lymphocytes, pericytes, and pre-adipocytes derived from fat tissue. Recent advances have shown the role and efficacy of SVF and ADSCs in improvement of tissue regeneration, especially in plastic reconstruction[40], such as breast reconstruction[41,42], wound healing[43,44], scars[45,46], and soft tissue defects[47]. The SVF is more easily obtained, regardless of cell separation and culture conditions. Therefore, therapeutic cellular products are obtained immediately with minimal contact with reagents, making it not only technically easier but also relatively safer. At the same time, the unique heterogeneous cell components of SVF may achieve better treatment results in comparative animal studies. It is worth noting that SVF might be suitable for autologous therapy only, due to the existence of various cell types that might cause immunological rejection[48], whereas ADSCs are useful in both allogenic therapy and autologous therapy.

As mentioned in the first part of this review, emerging in vitro studies have already definitively demonstrated the advantage of ADSCs in lymphangiogenesis and treatment of lymphedema. All five animal studies included in this systematic review were performed on mouse models and the ADSCs were injected immediately or shortly after lymphedema induction. These studies examined the effect of ADSCs on acute lymphedema and exhibited improvement at 4-6 wk. However, the anatomy, physiology, and healing capacity are quite different between mice and human beings, bringing up the need for larger animal models in order to simulate chronic lymphedema in human beings more accurately. Further preclinical studies with better animal models are needed to determine whether ADSC-based therapies could fulfil expectations and be extrapolated to clinical use in patients. As is commonly known, to observe lymphatics in mice, it is usually necessary to visualize the blue dye-stained lymphatic vessels in dissected tissues or tissue sectioning for immunohistological staining for the lymphatic markers, such as VEGFR-3, PROX1, and LYVE1, to quantitatively identify lymphatic vessel intensity and lymphangiogenesis. Advances in near-infrared fluoroscopy lymphatic imaging[49], magnetic resonance imaging agents based on nanotechnology, and gene reporter technologies have paved roads for depicting functions of lymphatic vasculature. Though not all lymphatic imaging methods are suitable for both clinical and preclinical trials, their effective combination will provide new tools for lymphedema translational medicine[50].

Therapy of lymphedema has benefitted substantially from the most recent advances in lymphatic vessel engineering and regenerative medicine, including cell-seeded scaffolds for vessel reconstruction, implantation of stem cells, prolymphangiogenic growth factors, or a combination of these technologies. As mentioned above, when implanted into the injury site in a lymphedema mouse model, the gelatin hydrogel containing controlled-release VEGF-C could encapsulate hADSCs, increase vessel density, and improve dermal edema, leading to efficient application in lymphatic vessel regeneration[25]. It has been reported that SVFs and ADSCs could improve wound healing when used alone or in combination with PRP and hyaluronic acid, specifically in healing of lower-extremity soft- and hard-tissue wounds[43] and in severe hidradenitis suppurativa wounds[47]. Many other studies confirmed that SVFs and ADSCs can improve wound healing when used alone or in combination with PRP and fat graft[41,45,51,52]. Considering the similar biological processes and biomolecular pathways shared between wound healing and lymphedema, such as inflammation and angiogenesis, the efforts towards establishing ADSCs-based therapy for lymphedema will benefit from these studies. Certainly, the research efforts towards development of cell isolation and expansion methods and of three-dimensional scaffolds[53], nanoparticles, and targeted delivery will help to optimize and maximize the efficacy of ADSC-assisted therapy. Standardized platforms with minimized manipulations and maximized efficacy will facilitate both up-scaling and the ease of clinical translation.

Over the past few years, concerns regarding the safety of stem cell application have also been raised. Indeed, Toyserkani et al[54] performed a systematic review, including over 1400 patients who received ADSC treatment and were followed for 4 wk to 3 years, in order to assess the safety of ADSC therapy, concentrating on the risks of thromboembolical, immunological, and oncological safety concerns. They reported that the observed adverse outcomes were relevant to liposuction, injuries from implantation, or the underlying condition, rather than the ADSC therapy itself. Although the ADSC therapy has shown a favorable safety performance, more reliable and rigorous safety assessment approaches are still encouraged for further study and clinical practice.

In conclusion, the results of in vitro studies, animal models, and clinical trials characterize ADSC-based treatment as a promising option and one that can be used within biologically rational and controllable environments for the treatment of lymphedema. However, further investigations in larger animal models and larger-scale, multicenter randomized clinical trials with more reliable and rigorous safety assessments are needed to develop more effective and durable therapeutic strategies.

ARTICLE HIGHLIGHTS
Research background

Lymphedema is a chronic, debilitating and incurable disease that affects 0.13%-2% of the global population. Emerging evidence indicates that adipose-derived stem cells (ADSCs) might serve as suitable seed cells for lymphatic tissue engineering and lymphedema therapy. Here, we appraise the in vivo evidence for the application of ADSCs for lymphedema treatments.

Research motivation

Emerging research findings have suggested that ADSCs might serve as suitable seed cells for tissue engineering of lymphatic vessels in vitro and potentially in the treatment of secondary lymphedema in vivo. It is critical that we systematically summarize the application of ADSCs for lymphedema treatments in animal studies and in clinical trials and discuss the future perspectives.

Research objectives

The main objectives of this review are to systematically summarize the application of ADSCs for lymphedema treatments as shown in animal studies and clinical trials. In addition, the future perspectives of ADSCs in lymphedema therapy are discussed.

Research methods

A systematic search was performed on four databases – PubMed, Clinicaltrials.gov, the evidence-based Cochrane library, and OVID – using the following search string: (“lymphedema” or “lymphoedema” or “lymphangiogenesis”) and (“adipose-derived stem cells” or “adipose-derived stromal cells” or “adipose-derived regenerative cells”). A manual search was performed by skimming the references of relevant studies. Animal studies and clinical trials using adipose-derived cells for the treatment of any kind of lymphedema were included.

Research results

A total of eight research articles published before November 2019 were included for this analysis. Five articles focused on animal studies and another three focused on clinical trials. ADSC transplantation therapy was demonstrated to be effective against lymphedema in all studies. The animal studies found that coadministration of ADSCs and controlled-release vascular endothelial growth factor-C or platelet-rich plasma could improve the effectiveness of ADSC therapy. Three sequential clinical trials were conducted on breast cancer-related lymphedema patients, and all showed favorable results.

Research conclusions

The results of in vitro studies, animal models, and clinical trials characterize ADSC-based treatment as a promising option and one that can be used within biologically rational and controllable environments for the treatment of lymphedema.

Research perspectives

Further preclinical studies in larger animal models and large-scale, multicenter randomized controlled clinical trials with more reliable and rigorous safety assessments are needed to develop more effective and durable therapeutic strategies. Advances in lymphatic imaging methods will provide opportunities for lymphedema translational medicine as well.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Cell and tissue engineering

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Gentile P S-Editor: Wang J L-Editor: Wang TQ E-Editor: Xing YX

References
1.  Dayan JH, Ly CL, Kataru RP, Mehrara BJ. Lymphedema: Pathogenesis and Novel Therapies. Annu Rev Med. 2018;69:263-276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 92]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
2.  Hespe GE, Nores GG, Huang JJ, Mehrara BJ. Pathophysiology of lymphedema-Is there a chance for medication treatment? J Surg Oncol. 2017;115:96-98.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
3.  Grada AA, Phillips TJ. Lymphedema: Pathophysiology and clinical manifestations. J Am Acad Dermatol. 2017;77:1009-1020.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 193]  [Article Influence: 27.6]  [Reference Citation Analysis (1)]
4.  Carlson JA. Lymphedema and subclinical lymphostasis (microlymphedema) facilitate cutaneous infection, inflammatory dermatoses, and neoplasia: A locus minoris resistentiae. Clin Dermatol. 2014;32:599-615.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 66]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
5.  Hartiala P, Saarikko AM. Lymphangiogenesis and Lymphangiogenic Growth Factors. J Reconstr Microsurg. 2016;32:10-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
6.  Cormier JN, Askew RL, Mungovan KS, Xing Y, Ross MI, Armer JM. Lymphedema beyond breast cancer: a systematic review and meta-analysis of cancer-related secondary lymphedema. Cancer. 2010;116:5138-5149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 290]  [Cited by in F6Publishing: 305]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
7.  Brorson H, Svensson H. Liposuction combined with controlled compression therapy reduces arm lymphedema more effectively than controlled compression therapy alone. Plast Reconstr Surg. 1998;102:1058-67; discussion 1068.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 101]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
8.  Li L, Yuan L, Chen X, Wang Q, Tian J, Yang K, Zhou E. Current Treatments for Breast Cancer-Related Lymphoedema: A Systematic Review. Asian Pac J Cancer Prev. 2016;17:4875-4883.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 16]  [Reference Citation Analysis (0)]
9.  Allen RJ, Cheng MH. Lymphedema surgery: Patient selection and an overview of surgical techniques. J Surg Oncol. 2016;113:923-931.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 90]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
10.  Qi S, Pan J. Cell-based therapy for therapeutic lymphangiogenesis. Stem Cells Dev. 2015;24:271-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
11.  Si Z, Wang X, Sun C, Kang Y, Xu J, Wang X, Hui Y. Adipose-derived stem cells: Sources, potency, and implications for regenerative therapies. Biomed Pharmacother. 2019;114:108765.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 159]  [Article Influence: 31.8]  [Reference Citation Analysis (0)]
12.  Sheykhhasan M, Wong JKL, Seifalian AM. Human Adipose-Derived Stem Cells with Great Therapeutic Potential. Curr Stem Cell Res Ther. 2019;14:532-548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 26]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
13.  Suh A, Pham A, Cress MJ, Pincelli T, TerKonda SP, Bruce AJ, Zubair AC, Wolfram J, Shapiro SA. Adipose-derived cellular and cell-derived regenerative therapies in dermatology and aesthetic rejuvenation. Ageing Res Rev. 2019;54:100933.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 34]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
14.  Torres-Torrillas M, Rubio M, Damia E, Cuervo B, Del Romero A, Peláez P, Chicharro D, Miguel L, Sopena JJ. Adipose-Derived Mesenchymal Stem Cells: A Promising Tool in the Treatment of Musculoskeletal Diseases. Int J Mol Sci. 2019;20:3105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 50]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
15.  Mazini L, Rochette L, Admou B, Amal S, Malka G. Hopes and Limits of Adipose-Derived Stem Cells (ADSCs) and Mesenchymal Stem Cells (MSCs) in Wound Healing. Int J Mol Sci. 2020;21:1306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 239]  [Cited by in F6Publishing: 210]  [Article Influence: 52.5]  [Reference Citation Analysis (0)]
16.  Beerens M, Aranguren XL, Hendrickx B, Dheedene W, Dresselaers T, Himmelreich U, Verfaillie C, Luttun A. Multipotent Adult Progenitor Cells Support Lymphatic Regeneration at Multiple Anatomical Levels during Wound Healing and Lymphedema. Sci Rep. 2018;8:3852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
17.  Conrad C, Niess H, Huss R, Huber S, von Luettichau I, Nelson PJ, Ott HC, Jauch KW, Bruns CJ. Multipotent mesenchymal stem cells acquire a lymphendothelial phenotype and enhance lymphatic regeneration in vivo. Circulation. 2009;119:281-289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 120]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
18.  Kinashi H, Ito Y, Sun T, Katsuno T, Takei Y. Roles of the TGF-β⁻VEGF-C Pathway in Fibrosis-Related Lymphangiogenesis. Int J Mol Sci. 2018;19:2487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 46]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
19.  Sun Y, Lu B, Deng J, Jiang Z, Cao W, Dai T, Li S. IL-7 enhances the differentiation of adipose-derived stem cells toward lymphatic endothelial cells through AKT signaling. Cell Biol Int. 2019;43:394-401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
20.  Wang X, Wang H, Cao J, Ye C. Exosomes from Adipose-Derived Stem Cells Promotes VEGF-C-Dependent Lymphangiogenesis by Regulating miRNA-132/TGF-β Pathway. Cell Physiol Biochem. 2018;49:160-171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 38]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
21.  Takeda K, Sowa Y, Nishino K, Itoh K, Fushiki S. Adipose-derived stem cells promote proliferation, migration, and tube formation of lymphatic endothelial cells in vitro by secreting lymphangiogenic factors. Ann Plast Surg. 2015;74:728-736.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 51]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
22.  Deng J, Dai T, Sun Y, Zhang Q, Jiang Z, Li S, Cao W. Overexpression of Prox1 Induces the Differentiation of Human Adipose-Derived Stem Cells into Lymphatic Endothelial-Like Cells In Vitro. Cell Reprogram. 2017;19:54-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
23.  Yan A, Avraham T, Zampell JC, Haviv YS, Weitman E, Mehrara BJ. Adipose-derived stem cells promote lymphangiogenesis in response to VEGF-C stimulation or TGF-β1 inhibition. Future Oncol. 2011;7:1457-1473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 62]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
24.  Saijo H, Suzuki K, Yoshimoto H, Imamura Y, Yamashita S, Tanaka K. Paracrine Effects of Adipose-Derived Stem Cells Promote Lymphangiogenesis in Irradiated Lymphatic Endothelial Cells. Plast Reconstr Surg. 2019;143:1189e-1200e.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
25.  Hwang JH, Kim IG, Lee JY, Piao S, Lee DS, Lee TS, Ra JC, Lee JY. Therapeutic lymphangiogenesis using stem cell and VEGF-C hydrogel. Biomaterials. 2011;32:4415-4423.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 67]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
26.  Shimizu Y, Shibata R, Shintani S, Ishii M, Murohara T. Therapeutic lymphangiogenesis with implantation of adipose-derived regenerative cells. J Am Heart Assoc. 2012;1:e000877.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
27.  Ackermann M, Wettstein R, Senaldi C, Kalbermatten DF, Konerding MA, Raffoul W, Erba P. Impact of platelet rich plasma and adipose stem cells on lymphangiogenesis in a murine tail lymphedema model. Microvasc Res. 2015;102:78-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
28.  Yoshida S, Hamuy R, Hamada Y, Yoshimoto H, Hirano A, Akita S. Adipose-derived stem cell transplantation for therapeutic lymphangiogenesis in a mouse secondary lymphedema model. Regen Med. 2015;10:549-562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
29.  Hayashida K, Yoshida S, Yoshimoto H, Fujioka M, Saijo H, Migita K, Kumaya M, Akita S. Adipose-Derived Stem Cells and Vascularized Lymph Node Transfers Successfully Treat Mouse Hindlimb Secondary Lymphedema by Early Reconnection of the Lymphatic System and Lymphangiogenesis. Plast Reconstr Surg. 2017;139:639-651.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 36]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
30.  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;5:857-859.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
31.  Toyserkani NM, Jensen CH, Andersen DC, Sheikh SP, Sørensen JA. Treatment of Breast Cancer-Related Lymphedema with Adipose-Derived Regenerative Cells and Fat Grafts: A Feasibility and Safety Study. Stem Cells Transl Med. 2017;6:1666-1672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
32.  Toyserkani NM, Jensen CH, Tabatabaeifar S, Jørgensen MG, Hvidsten S, Simonsen JA, Andersen DC, Sheikh SP, Sørensen JA. Adipose-derived regenerative cells and fat grafting for treating breast cancer-related lymphedema: Lymphoscintigraphic evaluation with 1 year of follow-up. J Plast Reconstr Aesthet Surg. 2019;72:71-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
33.  Dave M, Mehta K, Luther J, Baruah A, Dietz AB, Faubion WA. Mesenchymal Stem Cell Therapy for Inflammatory Bowel Disease: A Systematic Review and Meta-analysis. Inflamm Bowel Dis. 2015;21:2696-2707.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 70]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
34.  Lipinski MJ, Luger D, Epstein SE. Mesenchymal Stem Cell Therapy for the Treatment of Heart Failure Caused by Ischemic or Non-ischemic Cardiomyopathy: Immunosuppression and Its Implications. Handb Exp Pharmacol. 2017;243:329-353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 6]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
35.  Freitag J, Bates D, Boyd R, Shah K, Barnard A, Huguenin L, Tenen A. Mesenchymal stem cell therapy in the treatment of osteoarthritis: reparative pathways, safety and efficacy - a review. BMC Musculoskelet Disord. 2016;17:230.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 172]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
36.  Keerthi N, Chimutengwende-Gordon M, Sanghani A, Khan W. The potential of stem cell therapy for osteoarthritis and rheumatoid arthritis. Curr Stem Cell Res Ther. 2013;8:444-450.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
37.  Jurado M, De La Mata C, Ruiz-García A, López-Fernández E, Espinosa O, Remigia MJ, Moratalla L, Goterris R, García-Martín P, Ruiz-Cabello F, Garzón S, Pascual MJ, Espigado I, Solano C. Adipose tissue-derived mesenchymal stromal cells as part of therapy for chronic graft-versus-host disease: A phase I/II study. Cytotherapy. 2017;19:927-936.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 35]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
38.  Gentile P, Piccinno MS, Calabrese C. Characteristics and Potentiality of Human Adipose-Derived Stem Cells (hASCs) Obtained from Enzymatic Digestion of Fat Graft. Cells. 2019;8:282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 59]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
39.  Gentile P, Calabrese C, De Angelis B, Pizzicannella J, Kothari A, Garcovich S. Impact of the Different Preparation Methods to Obtain Human Adipose-Derived Stromal Vascular Fraction Cells (AD-SVFs) and Human Adipose-Derived Mesenchymal Stem Cells (AD-MSCs): Enzymatic Digestion Versus Mechanical Centrifugation. Int J Mol Sci. 2019;20:5471.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 63]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
40.  Gentile P, Scioli MG, Bielli A, Orlandi A, Cervelli V. Concise Review: The Use of Adipose-Derived Stromal Vascular Fraction Cells and Platelet Rich Plasma in Regenerative Plastic Surgery. Stem Cells. 2017;35:117-134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 102]  [Article Influence: 12.8]  [Reference Citation Analysis (1)]
41.  Gentile P, Scioli MG, Orlandi A, Cervelli V. Breast Reconstruction with Enhanced Stromal Vascular Fraction Fat Grafting: What Is the Best Method? Plast Reconstr Surg Glob Open. 2015;3:e406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 70]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
42.  Gentile P, Casella D, Palma E, Calabrese C. Engineered Fat Graft Enhanced with Adipose-Derived Stromal Vascular Fraction Cells for Regenerative Medicine: Clinical, Histological and Instrumental Evaluation in Breast Reconstruction. J Clin Med. 2019;8:504.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 79]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
43.  Cervelli V, Lucarini L, Spallone D, Palla L, Colicchia GM, Gentile P, De Angelis B. Use of platelet-rich plasma and hyaluronic acid in the loss of substance with bone exposure. Adv Skin Wound Care. 2011;24:176-181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 68]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
44.  Gentile P, Garcovich S. Concise Review: Adipose-Derived Stem Cells (ASCs) and Adipocyte-Secreted Exosomal microRNA (A-SE-miR) Modulate Cancer Growth and proMote Wound Repair. J Clin Med. 2019;8:855.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 102]  [Article Influence: 20.4]  [Reference Citation Analysis (0)]
45.  Gentile P, De Angelis B, Pasin M, Cervelli G, Curcio CB, Floris M, Di Pasquali C, Bocchini I, Balzani A, Nicoli F, Insalaco C, Tati E, Lucarini L, Palla L, Pascali M, De Logu P, Di Segni C, Bottini DJ, Cervelli V. Adipose-derived stromal vascular fraction cells and platelet-rich plasma: basic and clinical evaluation for cell-based therapies in patients with scars on the face. J Craniofac Surg. 2014;25:267-272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 153]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
46.  Gentile P, Scioli MG, Bielli A, Orlandi A, Cervelli V. Comparing different nanofat procedures on scars: role of the stromal vascular fraction and its clinical implications. Regen Med. 2017;12:939-952.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 64]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
47.  Nicoli F, Balzani A, Lazzeri D, Gentile P, Chilgar RM, Di Pasquali C, Nicoli M, Bocchini I, Agovino A, Cervelli V. Severe hidradenitis suppurativa treatment using platelet-rich plasma gel and Hyalomatrix. Int Wound J. 2015;12:338-343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 58]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
48.  Bora P, Majumdar AS. Adipose tissue-derived stromal vascular fraction in regenerative medicine: a brief review on biology and translation. Stem Cell Res Ther. 2017;8:145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 285]  [Article Influence: 40.7]  [Reference Citation Analysis (0)]
49.  De Vrieze T, Vos L, Gebruers N, Tjalma WAA, Thomis S, Neven P, Nevelsteen I, De Groef A, Vandermeeren L, Belgrado JP, Devoogdt N. Protocol of a randomised controlled trial regarding the effectiveness of fluoroscopy-guided manual lymph drainage for the treatment of breast cancer-related lymphoedema (EFforT-BCRL trial). Eur J Obstet Gynecol Reprod Biol. 2018;221:177-188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
50.  Sevick-Muraca EM, Kwon S, Rasmussen JC. Emerging lymphatic imaging technologies for mouse and man. J Clin Invest. 2014;124:905-914.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 93]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
51.  Gentile P, Kothari A, Casella D, Calabrese C. Fat Graft Enhanced With Adipose-Derived Stem Cells in Aesthetic Breast Augmentation: Clinical, Histological, and Instrumental Evaluation. Aesthet Surg J. 2019;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 57]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
52.  Smith OJ, Jell G, Mosahebi A. The use of fat grafting and platelet-rich plasma for wound healing: A review of the current evidence. Int Wound J. 2019;16:275-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
53.  Scioli MG, Bielli A, Gentile P, Cervelli V, Orlandi A. Combined treatment with platelet-rich plasma and insulin favours chondrogenic and osteogenic differentiation of human adipose-derived stem cells in three-dimensional collagen scaffolds. J Tissue Eng Regen Med. 2017;11:2398-2410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 92]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
54.  Toyserkani NM, Jørgensen MG, Tabatabaeifar S, Jensen CH, Sheikh SP, Sørensen JA. Concise Review: A Safety Assessment of Adipose-Derived Cell Therapy in Clinical Trials: A Systematic Review of Reported Adverse Events. Stem Cells Transl Med. 2017;6:1786-1794.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 81]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]