Application of Autological Adipose Tissue and its Products in the Treatment of Infected Wounds of Different Genesis


Cite item

Full Text

Abstract

Introduction. Transplantation of autologous adipose tissue and its products to stimulate the wound process is a rapidly developing area of ​​regenerative medicine. Adaptation of the technology to the conditions of infected wound treatment is a great challenge.
The aim of the study was to determine the most effective and safe technologies for obtaining, processing and transplanting autologous adipose tissue used to treat infected wounds.
Material and methods. The authors searched for relevant papers published in 2011 to 2021 in electronic databases PubMed, eLIBRARY, Embase, Cyberleninka, Web of Science. The analysis included 30 sources: 5 RCTs, 16 clinical studies, 3 case reports, 1 systematic review, 2 literature reviews, and 4 sources published before 2011 due to their fundamental and practical significance for the scope of the issue.
Results and discussions. Although the lipoaspiration procedure in patients with infected wounds is described as standard conventional one, it still has a number of significant features that distinguish it from the standard technology of aesthetic surgery. To treat lipoaspirate, both enzymatic and mechanical options were used with the same frequency. The main technique of introducing adipose tissue products was local intradermal injections into the edges and under the wound bed; in a few cases, applications on the wound surface were used, or a combination of these options. As recorded, the cellularity index of adipose tissue products ranged from (min) 1x104/ml to (max) 50x106/ml. It should also be noted that a unified terminology for adipose tissue products used for wound treatment has not yet been adopted, but the most common term is “stromal-vascular adipose tissue fraction”.
Conclusions. Systematization of literature data on the use of autologous adipose tissue products for wound treatment has shown that at present this technology is safe, provided with an accessible instrumental base, has positive results and is being actively developed. Further randomized clinical trials, involving extensive and multiple wounds, are required to determine the minimum effective and maximum tolerable dose in the treatment of wounds with adipose tissue products. In addition, it is promising to develop new means and approaches for delivering adipose tissue products in the treatment of infected wounds of various origins.

Full Text

Autologous fat grafting (lipografting) and its products to stimulate the wound process is a rapidly developing area of ​​regenerative medicine. Although some preclinical studies on the efficacy of lipografting in wound healing, both in animal and in vitro models, are encouraging, it remains to be definitively established whether transplantation of autologous adipose tissue and its products improves clinical wound healing outcomes [1]. In addition, lipografting is increasingly being studied as a component of the treatment of other surgical diseases associated with tissue trauma and infection: open fractures, damage to peripheral nerves, external intestinal fistulas, Crohn's disease [2-5]. The technology of lipografting is changing and developing along with the expansion of the range of its application. It is necessary to systematize the data on the adaptation of the traditional operation of adipose tissue transplantation to the new conditions of its application.

Purpose. By systematic analysis of the literature, to determine the most effective and safe technologies for obtaining, processing and transplanting autologous adipose tissue used to treat infected wounds.

Material and methods. Search methods. In electronic databases PubMed. eLIBRARY, Embase, Cyberleninka, Web of Science searched for relevant articles from 2011 to 2021. The keywords for the search were: “autologous fat grafting”, “lipografting”, “stromal vascular fraction”, “adipose derived stem cells”, “wounds”, “wound treatment”, “burns”, “burn treatment”, “chronic ulcers”, “treatment of chronic ulcers” in Russian and English. Search algorithms: 1) “transplantation of autologous adipose tissue” or “lipografting” or “fat grafting” or “stromal vascular fraction” or “adipose derived stem cells” + “wounds” or “wound treatment”; 2) “ autologous fat grafting ” or “lipografting” or “fat grafting” or “stromal vascular fraction” or “adipose derived stem cells” + “burns” or “burn treatment”; 3) " autologous fat grafting " or "lipografting" or "fat grafting" or "stromal vascular fraction" or " adipose derived stem cells " + " chronic ulcers " or "treatment of chronic ulcers ". Based on the specified keywords, 2292 sources were included in the preliminary analysis.

Study inclusion  criteria. 1. Treatment of infected soft tissue wounds against the background of chronic circulatory disorders, diabetes mellitus; burns; fistulas; 2. Clinical presense of wound infection; 3. The use of autologous adipose tissue or its low-manipulation products in local treatment. Study exclusion  criteria: 1. Experimental studies on animals; 2. Cosmetic surgery; 3. Surgical treatment of scars. After applying inclusion and exclusion criteria, combining database results and removing duplicate entries, checking titles and abstracts, the remaining articles were read in full.

Results and their discussion.

As a result, 30 sources published over the past 10 years were analyzed. Of these: 5 RCTs, 16 clinical trials, 3 case reports, 1 systematic review, 2 literature reviews. 4 sources published earlier than 10 years are included, due to their fundamental and practical significance for the issue under consideration (Table 1),

Table 1. Summary data.

Author

Study design

Number of patients

Fat Processing

 

Cellularity index

Adipose derived product

1

Deng C[6]

Clinical trial

20

Mechanical (nanofat)

unknown

SVF-gel

2

Nilforoushzadeh MA[7]

Clinical trial

5

Enzymatic (collagenase)

1×104/ml

SVF-cells

3

Konstantinow A[8]

Controlled Clinical Trial

16

Enzymatic (collagenase, dyspase)

15x106 (6 ml)

SVF

4

Samoylov AS.[9]

Original Article

5

Enzymatic (collagenase)

from 10х106 to 50х106 /5 ml

SVF

5

Marino G[10]

Controlled Clinical Trial

20

Enzymatic (automatic)

3х105 /ml (5 ml)

ADSC

6

Bene MD[11]

Case series, prospective

9

Mechanical (centrifugation)

unknown

Purified fat (mid.layer)

7

Marangi GF[12]

Case series, prospective

42

Mechanical (centrifugation)

unknown

Fat

8

Del Papa N[13]

Case series, prospective

15

Mechanical (centrifugation)

unknown

Adipose derived tissue cells (mid.layer)

9

Piccolo NS[3]

Review

282

Mechanical (centrifugation)

unknown

SVF

10

Stasch T[14]

Prospective cohort study

26

without processing

unknown

Fat

11

Caviglia H[4]

Case series, prospective

6

Mechanical (centrifugation)

unknown

Concetrate fat cells

12

Kim JH[15]

Case series, retrospective

12

Enzymatic (collagenase)

unknown

SVF

13

Carstens MH[16]

Original Article

10

Enzymatic (collagenase)

unknown

SVF-cells

14

Chopinaud M[17]

Prospective pilot study

10

Mechanical (centrifugation)

unknown

Fat micrografts (mid.layer)

15

Cervelli V[18]

Case control study

10

Enzymatic (automatic)

5х104 /ml

SVF

16

Mizushima T[5]

Clinical Trial

6

Enzymatic (automatic)

3,5 ×106

Adipose derived regenerative cells

17

Giuggioli D[19]

Case Reports

1

without processing

unknown

Fat

18

Smith OJ[20]

Randomized Controlled Trial

18

without processing

unknown

Isolated lipoaspirate

19

Piccolo NS[21]

Review

27

Mechanical (centrifugation)

unknown

Fat

20

Williams EA[22]

Original Article

20

without processing

unknown

SVF

21

Biyao Z[23]

Case Reports

1

Mechanical (centrifugation)

unknown

------

22

Rangaswamy M[24]

Case Reports

3

without processing

unknown

Fat

23

Abouzaid AM[25]

Randomized control clinical trial

100

Mechanical (nanofat)

unknown

Nanofat

24

Carstens MH[26]

Сlinical trial

63

Enzymatic (automatic)

30 × 106

SVF

25

Tanios E[27]

Randomized Controlled Trial

100

Enzymatic (collagenase)

unknown

SVF-cells

26

Ryabkov MG.[28]

Prospective clinical trial

34

Mechanical (sedimentation)

unknown

Lipoaspirate (mid.layer)

 

Physiological basis of the regenerative potential of autologous adipose tissue. Lipografting has been used as an aesthetic procedure in plastic surgery and cosmetology for more than 20 years. At the beginning of the period of surgical application, adipose tissue was used as a "filler", a plastic material to change defects in body contours. A new therapeutic potential of adipose tissue was discovered in 2001 by Zuk, Hedrick et al., when mesenchymal stem cells were isolated from it, later called adipose derived stem cells (ADSCs) [29]. As a result of intensive research, it was found that ADSCs are multipotent mesenchymal stem cells and are able to differentiate into various cells, including keratinocytes, fibroblasts, endotheliocytes. In addition, ADSCs have a wide variety of regenerative and metabolic properties, and are also able to secrete various paracrine factors: epidermal growth factor, transforming growth factor-β, hepatocyte growth factor, platelet-derived growth factor, basic fibroblast growth factor, placental growth factor, vascular endothelial growth factor, which initiate tissue repair and accelerate the process of wound healing due to the activation of neovasculogenesis [2]. It was these data that subsequently became the basis for the use of ADSCs in practical wound surgery, since they made it possible to more clearly define the goal and expected physiological effects of therapy.

Features of fat harvesting in patients with infected wounds. Adipose tissue sampling in most of the sources included in the review was described as a standard procedure performed according to the methodology proposed by Coleman [30]. This technique involves the use of a 10 ml Luer Lok syringe attached to a 3-3.5 mm cannula with two 3 mm side distal holes. The length of the cannula used is 10, 15 or 20 cm, depending on the sampling site. The most common lipoaspiration procedure was performed in the area of ​​the anterior abdominal wall. Only in 4 cases, adipose tissue sampling was performed from other areas: internal thigh [3, 5, 12] and from the buttocks [18]. Lipoaspiration was performed under sterile conditions of the operating room or procedure room, usually under local tumescent anesthesia, and in one case under spinal anesthesia [3] and under general anesthesia [18]. During lipoaspiration, standard or modified Klein's solution (physiological saline or Ringer's lactate and/or local anesthetic and/or adrenaline) [19] was used, which was necessary to reduce the trauma of the procedure for extracting adipose tissue, for local hemostasis, and for pain contol. A 3-5 mm long incision was made with a scalpel, and a blunt cannula was inserted to collect fat. The intake of adipose tissue was carried out by creating a slight negative pressure while retracting the syringe plunger and simultaneously performing reciprocating hand movements. The amount of adipose tissue obtained in one manipulation, in the analyzed literature, was indicated in 8 works and ranged from 30 ml (min) [8] to 400 ml (max) [18]. In 2 sources, to calculate the required amount of adipose tissue, the formula was proposed: 1 cm3 of adipose tissue per 1 cm2 of the wound surface [14, 19].

Although the lipoaspiration in patients with infected wounds is described by the authors as standard, it still has a number of significant features that distinguish it from the standard technology of aesthetic surgery. These features include: 1. The choice of a zone for obtaining adipose tissue is determined by the clinical situation, namely, the maximum distance from the zone of damaged skin. This is especially important in patients with extensive burns. 2. The amount of adipose tissue obtained in one manipulation may be limited due to extensive damage or severe exhaustion in patients with burn wounds.

Fat processing. Adipose derived products in wounds treatment. Adipose tissue products used in the treatment of wounds. Treatment options for lipoaspirate have varied in different sources. In 10 studies, the production of the final product from adipose tissue was carried out by an enzymatic method using collagenase [5, 7-10, 15, 16, 18, 26, 27], 4 of them used automatic Celution systems [5, 10, 18, 26] , and in one case, dispase was also used as an enzyme [8]. In 14 studies, lipoaspirate was subjected to minimal mechanical processing in the form of centrifugation for 1-3 min at a speed of 920-3500 rpm [3, 4, 11-13, 17, 21, 23] or sedimentation [28] followed by removal of the middle layer containing “purified fat”, or not processed at all [14, 19, 20, 22, 24]. In only 2 studies, lipoaspirate was mechanically emulsified by moving between two 10-cc syringes connected to each other with a mother-mother Luer-Lok connector and the so-called “nanofat” was obtained [6, 25]. The resulting products in 11 cases were designated as “stromal-vascular fraction (SVF) of adipose tissue” [3, 6-9, 15, 16, 18, 22, 26, 27], and in 8 of them enzymatic methods of processing lipoaspirate were used [ 7-9, 15, 16, 18, 26, 27], in 2 of them mechanical methods [3, 6] and in one were not processed [22]. Among the enzymatic methods, only 2 products had a name other than “SVF” - “solution of adipose derived stem cells” [10] and “adipose derived regenerative cells” [5]. Among the mechanical processing methods, with the exception of the two cases mentioned above, the authors used the terms: "fat", "purified fat", "cells from adipose tissue", "nanofat", "fat micrografts". Thus, despite the fact that most authors describe the technology for obtaining and processing adipose tissue as “standard”, the stages of the technology involve the same type of manipulation, but the products used for injection into the wound are designated very differently. A unified terminology for adipose derived products used in the wounds treatment has not yet been adopted. The most common is the term "stromal-vascular fraction of adipose tissue".

Application methods. The next stage of lipografting is the introduction of the resulting product into the area of the recipient wound. Judging by the analyzed sources, two methods of delivery are most often used in the treatment of wounds: local injections or application of adipose derived products directly to the wound surface (Table 2).

 

Table 2. Application methods of adipose derived products.

 

 

Author

Characteristic of wound

Application method

1.

Deng C[6]

Chronic wounds (>3 months)

Approximately 3/4 of the prepared SVF gel was injected directly into the base and edges of the wound, and the remaining gel covered the wound as a dressing, which was then covered with saline gauze.

2.

Nilforoushzadeh MA[7]

Fill-thickness neuropatic diabetic ulcers

A three-layer hydrogel skin substitute (inoculated with autologous keratinocytes, fibroblasts, and SVF) was transplanted onto the wounds.

3.

Konstantinow A[8]

Chronic venous and arterial ulcers from от 2 сm2 до 400 сm2.

4 ml of suspension of SVF was injected to a depth of 5-10 mm into the central area and the area adjacent to the ulcer in different directions. An additional 2.5 ml of the cell suspension was soaked in a collagen sponge cut to fit the wound and placed on the wound area.

4.

Samoylov AS.[9]

Thermal burns III–IV in TBSA from 1 to 80 %

Intradermal administration of autologous SVF AT was performed at 10–15 points around the damage to the skin.

5.

Marino G[10]

Chronic diabetic foot ulcers

Isolated cells were injected along the edges of the ulcer; cells were injected to a depth of 1 cm, trying to distribute them in all directions.

6.

Bene MD[11]

Digital ulcers

2–3 ml of fat was injected into the fingers using blunt cannulas, at the border of larger ulcers of different depths, or at the base of the fingers for smaller ulcers.

7.

Marangi GF[12]

Pressure ulcers.

1 cm3 of fat per 1 cm2 of the recipient zone was injected into the deep subcutaneous layer, where ischemic damage occurs, using a 3 mm diameter lipostructure cannula.

8.

Del Papa N[13]

Single diabetic foot ulcer (>5 months)

0.5–1 ml of autofraction was injected into the base of the affected finger by successive introduction of small aliquots in different directions from the injection site.

9.

Piccolo NS[3]

Burn wounds, diabetic foot ulcer, arterial ulcers

A 1 ml syringe is inserted through the needle puncture hole and (with force) is inserted directly under the wound bed or scar. The fat is then deposited retrograde over several "passes" until the entire area is entered. On average, 1.6–2.0 ml is injected for every 10 cm2 of area; 25–30 passes are required to inject 1 ml. Also injected through the wound and applied to the wound.

10.

Stasch T[14]

Chronic ulcers of lower extremities; wound surface more 2 сm2; and etiology, include only venous, arterial, diabetic, postoperative, posttraumatic and pressure.

2 ml of lipoaspirate per 1 cm2 of the wound, followed by infiltration of lipoaspirate into the edges of the wound and injection under the lesion into the bottom of the cleaned ulcer, if possible.

11.

Caviglia H[4]

Cutaneous fistulas

The concentrate of fat cells is injected in small portions along the edge of the lesion.

12.

Kim JH[15]

Post-injections face skin necrosis

The final solution was injected into the lesion in the subcutaneous and dermal layers near the wound.

13.

Carstens MH[16]

Arterial ulcers of lower extremities

In the presence of an ulcer, SVF (3–4 ml) was infiltrated directly under the wound and along its edges.

14.

Chopinaud M[17]

Hypertensive ulcers

Insertion of fat micrografts around and under the wound.

15.

Cervelli V[18]

Posttraumatic ulcer of lower  extremities

Direct injection into the bottom of the ulcer or injection into the perilesional area

16.

Mizushima T[5]

Cutaneous fistula no more 10 ml.

1/4 volume of the concentrated cell solution was injected percutaneously into the tissues around the fistula, and 1/4 volume was injected endoscopically into the submucosal tissue around the fistula. The remaining half was mixed with thrombin solution and injected with fibrinogen solution. Finally, the fistula was filled with fibrin glue containing ADSC and the skin was sutured.

17.

Giuggioli D[19]

Stump-skin ulcer

From 0.5 to 1 ml of fat is injected into the edges of the wound and into the subcutaneous plane around the entire perimeter of the finger stump (crown-shaped

18.

Smith OJ[20]

Single diabetic foot ulcer >25 mm2 and <10000 mm2 (>3 months)

The incision was made at least 1 cm from the edge of the wound and the fat was infiltrated using a threading technique to allow the fat to enter the natural tissue planes. Approximately 2 ml of lipoaspirate was infiltrated per 1 cm2 of the wound.

19.

Piccolo NS[21]

Face burns and wounds.

Introduced through the needle puncture hole and immediately inserted under the wound bed. The fat is then deposited retrogradely, in several "passes", until the entire area is transplanted (through the required number of punctures along the periphery of the wound). On average, 1.8–2.5 cm3 of fat is injected for every 10 cm2, and 25–30 “passes” must be made to inject 1 cm3.

20.

Williams EA[22]

Chronic venous ulcers of lower extremities

Fat is injected using microcannulas to reimplant it around the edges of the ulcer.

21.

Biyao Z[23]

Diabetic foot ulcer

unknown

22.

Rangaswamy M[24]

Flaps-needed difficult wounds.

Three-layer matrix of platelet-rich fibrin gel, condensed liposuction own fat and porous collagen dressing.

23.

Abouzaid AM[25]

Superficial and deep burns

Single injection of autologous adipose tissue, followed by coating of the wound with nanofat.

24.

Carstens MH[26]

Diabetic foot ulcer.

SVF cells in a volume of 20 cm3 were injected into the subcutaneous tissues surrounding the ulcer along the perimeter using a series of injections of a small volume (0.5 cm3)

25.

Tanios E[27]

Chronic trophic ulcers.

Autologous stem cell injections every 3 weeks.

26

Ryabkov MG.[28]

Chronic trophic and pressure ulcers. posttraumatic wound

The middle fraction of lipoaspirate was moved to the surface of the wound or filled its cavity.

 

In 17 studies, the application of i adipose derived products was local intradermal injections into the edges and under the bed of the recipient wound [3-6, 8-22, 25, 27]. In 4 cases, in addition to injections, applications were additionally used on the wound surface [5, 6, 8, 25]. Only 4 studies used isolated application of adipose tissue products to the wound surface [3, 7, 24, 28]. It should be noted that extensive wounds of the skin were the subject of treatment and study of only two studies included in the analysis [8, 9]. In the rest, there were either no mentions of the size of the wound surface [3, 4, 6, 7, 10-13, 15-19, 21-28], or patients with single wounds of small sizes were included in the study [5, 14, 20]. This may explain the predominant use of local injections as a method of delivering the drug to the wound. In the treatment of extensive and/or multiple infected wounds, local injections, as a method of introducing products of autologous adipose tissue, may be unnecessarily time-consuming, traumatic, and, in case of extensive burns, technically unfeasible.

Dosage of autologous adipose derived products. For an objective assessment of the effectiveness of the use of any drug or product introduced into the human body, it is necessary to determine its dose, which makes it possible to unify the method of its use. Most products from adipose tissue, regardless of the method of processing, contain stem cells, so the cellularity of the product can be considered an objective characteristic, i.e. the content of cells in 1 ml. It should be noted that out of 26 sources included in the review, the cellularity of the administered product was objectified and indicated only in 7 cases [5, 7-10, 26]. The cellularity index ranged from (min) 1x104/ml [7] to (max) 50x106/ml [9]. However, a positive effect in the form of wound healing or trophic ulcers was achieved in all the considered works. Further randomized clinical trials are needed to accurately recommend the minimum effective and maximum tolerable doses for treating wounds with adipose tissue products.

Conclusions. Systematization of literature data on the use of autologous adipose derived products for wound treatment has shown that at present this technology is safe, provided with an accessible instrumental base, has positive results and is being actively developed. The technique for sampling adipose tissue is standard, but has a number of features in the presence of infected wounds in patients. Among the products of adipose tissue, there is still no single nomenclature, therefore, in order to unify the methods for their production and processing, it is necessary to approve a common terminology for all. Further randomized clinical trials, which will also include extensive and multiple wounds, are required to determine the minimum effective and maximum allowable dose in the treatment of wounds with adipose tissue products. In addition, it is promising to develop new means and methods for delivering adipose tissue products in the treatment of infected wounds of various origins.

×

About the authors

Nikita Anatolyevich Koloshein

University Clinic of the Federal State Budgetary Educational Institution of Higher Education "Volga Research Medical University" of the Ministry of Health of the Russian Federation

Author for correspondence.
Email: n.koloshein@mail.ru
ORCID iD: 0000-0002-4563-5478
SPIN-code: 8542-6969

Junior Researcher of the Burn Injury Study Group

Russian Federation, 603005, Russia, 10/1 Minin and Pozharsky Square

References

  1. Luck J, Smith OJ, Malik D, Mosahebi A. Protocol for a systematic review of autologous fat grafting for wound healing. Syst Rev. 2018;7(1):99. doi: 10.1186/s13643-018-0769-7.
  2. Pavlov VN, Kazixinurov AA, Kazixinurov RA, Pushkarev AM., Agaverdiev MA., Maksimova SYu., et all. Modern possibilities of clinical application of the adipose tissue-derived stromal vascular fraction. Review. FGBOU VO «Bashkirskij gosudarstvenny`j medicinskij universitet» Minzdrava Rossii, g. Ufa. 2020; 15:6 (90). (in Russ.)
  3. Piccolo NS, Piccolo MS, Piccolo MT. Fat grafting for treatment of burns, burn scars, and other difficult wounds. Clin Plast Surg. 2015; 42(2):263-83. doi: 10.1016/j.cps.2014.12.009.
  4. Caviglia H, Landro ME, Gallo E, Douglas Price AL, Galatro G, Neme D. Is it possible to use autologous adipose graft for wound repair in patients with coagulation disorders? Haemophilia. 2016; 22(2):298-302. doi: 10.1111/hae.12804.
  5. Mizushima T, Takahashi H, Takeyama H, Naito A, Haraguchi N, Uemura M. A clinical trial of autologous adipose-derived regenerative cell transplantation for a postoperative enterocutaneous fistula. Surg Today. 2016; 46(7):835-42. doi: 10.1007/s00595-015-1246-8.
  6. Deng C, Wang L, Feng J, Lu F. Treatment of human chronic wounds with autologous extracellular matrix/stromal vascular fraction gel: A STROBE-compliant study. Medicine (Baltimore). 2018; 97(32):e11667. doi: 10.1097/MD.0000000000011667.
  7. Nilforoushzadeh MA, Sisakht MM, Amirkhani MA, Seifalian AM, Banafshe HR, Verdi J. Engineered skin graft with stromal vascular fraction cells encapsulated in fibrin-collagen hydrogel: A clinical study for diabetic wound healing. J Tissue Eng Regen Med. 2020; 14(3):424-440. doi: 10.1002/term.3003.
  8. Konstantinow A, Arnold A, Djabali K, Kempf W, Gutermuth J, Fischer T. 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):2104-2118. doi: 10.1111/jdv.14489.
  9. Samoilov AS, Astrelina TA, Aksenenko AV, Kobzeva IV, Suchkova YuB, Nikitina VA, et all. Application of cell technologies in thermal burn damage to skin (Practical experience in State Research Center— Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency of Russia). Saratovskii nauchno-meditsinskii zhurnal. 2019; 15 (4): 999–1004. (in Rus.)
  10. Marino G, Moraci M, Armenia E, Orabona C, Sergio R, De Sena G, et all. Therapy with autologous adipose-derived regenerative cells for the care of chronic ulcer of lower limbs in patients with peripheral arterial disease. J Surg Res. 2013; 185(1):36-44. doi: 10.1016/j.jss.2013.05.024.
  11. Bene MD, Pozzi MR, Rovati L, Mazzola I, Erba G, Bonomi S. Autologous fat grafting for scleroderma-induced digital ulcers. An effective technique in patients with systemic sclerosis. Plast Chir. 2014; 46(4):242-7. doi: 10.1055/s-0034-1376970.
  12. Marangi GF, Pallara T, Cagli B. Treatment of early-stage pressure ulcers by using autologous adipose tissue grafts. Plast Surg Int. 2014:817283. doi: 10.1155/2014/817283.
  13. Del Papa N, Di Luca G, Sambataro D, Zaccara E, Maglione W, Gabrielli A, et all. Regional implantation of autologous adipose tissue-derived cells induces a prompt healing of long-lasting indolent digital ulcers in patients with systemic sclerosis. Cell Transplant. 2015;24(11):2297-305. doi: 10.3727/096368914X685636.
  14. Stasch T, Hoehne J, Huynh T, De Baerdemaeker R, Grandel S, Herold C. Débridement and Autologous Lipotransfer for Chronic Ulceration of the Diabetic Foot and Lower Limb Improves Wound Healing. Plast Reconstr Surg. 2015; 136(6):1357-1366. doi: 10.1097/PRS.0000000000001819.
  15. Kim JH, Park SH, Lee BH, Jeong HS, Yang HJ, Suh IS. Early Intervention with Highly Condensed Adipose-Derived Stem Cells for Complicated Wounds Following Filler Injections. Aesthetic Plast Surg. 2016;40(3):428-434. doi: 10.1007/s00266-016-0636-7.
  16. Carstens MH, Gómez A, Cortés R, Turner E, Pérez C, Ocon M. Non-reconstructable peripheral vascular disease of the lower extremity in ten patients treated with adipose-derived stromal vascular fraction cells. Stem Cell Res. 2017; 18:14-21. doi: 10.1016/j.scr.2016.12.001.
  17. Chopinaud M, Labbé D, Creveuil C, Marc M, Bénateau H, Mourgeon B. Autologous Adipose Tissue Graft to Treat Hypertensive Leg Ulcer: A Pilot Study. Dermatology. 2017;233(2-3):234-241.
  18. Cervelli V, Gentile P, De Angelis B, Calabrese C, Di Stefani A, Scioli MG. Application of enhanced stromal vascular fraction and fat grafting mixed with PRP in post-traumatic lower extremity ulcers. Stem Cell Res. 2011; 6(2):103-11. doi: 10.1016/j.scr.2010.11.003.
  19. Giuggioli D, Spinella A, Cocchiara E, de Pinto M, Pinelli M, Parenti L, et all. Autologous fat grafting in the treatment of a scleroderma stump-skin ulcer: a case report. Case Reports Plast Surg Hand Surg. 2021; 12;8(1):18-22. doi: 10.1080/23320885.2021.1881521.
  20. Smith OJ, Leigh R, Kanapathy M. Fat grafting and platelet-rich plasma for the treatment of diabetic foot ulcers: A feasibility-randomised controlled trial. Int Wound J. 2020;17(6):1578-1594. doi: 10.1111/iwj.13433.
  21. Piccolo NS, Piccolo MS, de Paula Piccolo N, de Paula Piccolo P, de Paula Piccolo N, Daher RP, et all. Fat Grafting for Treatment of Facial Burns and Burn Scars. Clin Plast Surg. 2020; 47(1):119-130. doi: 10.1016/j.cps.2019.08.015.
  22. Williams EA, Thaller SR. The Role of Fat Grafting in the Treatment of Keloid Scars and Venous Ulcers. J Craniofac Surg. 2019; 30(3):696-697. doi: 10.1097/SCS.0000000000005208.
  23. Biyao Z, Gang X, Hai J, Chenwang D, Xuan L. Autologous fat grafting combined with negative pressure wound therapy in severe diabetic foot ulcer: a case study. J Wound Care. 2021; 1;30(Sup4):S38-S40. doi: 10.12968/jowc.2021.30.Sup4.S38.
  24. Rangaswamy M. Regenerative Wound Healing by Open Grafting of Autologous Fat and PRP-Gel - A New Concept and Potential Alternative to Flaps. Plast Reconstr Surg Glob Open. 2021; 25;9(1):e3349. doi: 10.1097/GOX.0000000000003349.
  25. Abouzaid AM, El Mokadem ME, Aboubakr AK, Kassem MA, Al Shora AK, Solaiman A. Effect of autologous fat transfer in acute burn wound management: A randomized controlled study. Burns. 2021; 1:S0305-4179(21)00298-9. doi: 10.1016/j.burns.2021.10.011.
  26. Carstens MH, Quintana FJ, Calderwood ST, Sevilla JP, Ríos AB, Rivera CM, et all. Treatment of chronic diabetic foot ulcers with adipose-derived stromal vascular fraction cell injections: Safety and evidence of efficacy at 1 year. Stem Cells Transl Med. 2021; 10(8):1138-1147. doi: 10.1002/sctm.20-0497.
  27. Tanios E, Ahmed TM, Shafik EA, Sherif MF, Sayed D, Gaber N, et all. Efficacy of adipose-derived stromal vascular fraction cells in the management of chronic ulcers: a randomized clinical trial. Regen Med. 2021; 16(11):975-988. doi: 10.2217/rme-2020-0207.
  28. Ryabkov MG, Spiridonov AA, Beschastnov VV, Tixonova OA, Leontev AE. Treatment of chronic wounds by combination of autodermatoplasty and lipotransfer. Klinicheskaya i e`ksperimental`naya xirurgiya. Zhurnal imeni akademika B.V. Petrovskogo. 2018; 4(22): 17–23. doi: 10.24411/2308-1198-2018-14002. (in Russ.)
  29. 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.
  30. Coleman WP 3rd. Fat transplantation. Dermatol Clin. 1999; 17(4):891-8, viii. doi: 10.1016/s0733-8635(05)70136-4.

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright (c) 2022 Koloshein N.A.

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies