Skip to main content
Erschienen in: Journal of Orthopaedic Surgery and Research 1/2017

Open Access 01.12.2017 | Review

The use of mesenchymal stem cells for cartilage repair and regeneration: a systematic review

verfasst von: Andy Goldberg, Katrina Mitchell, Julian Soans, Louise Kim, Razi Zaidi

Erschienen in: Journal of Orthopaedic Surgery and Research | Ausgabe 1/2017

Abstract

Background

The management of articular cartilage defects presents many clinical challenges due to its avascular, aneural and alymphatic nature. Bone marrow stimulation techniques, such as microfracture, are the most frequently used method in clinical practice however the resulting mixed fibrocartilage tissue which is inferior to native hyaline cartilage. Other methods have shown promise but are far from perfect. There is an unmet need and growing interest in regenerative medicine and tissue engineering to improve the outcome for patients requiring cartilage repair. Many published reviews on cartilage repair only list human clinical trials, underestimating the wealth of basic sciences and animal studies that are precursors to future research. We therefore set out to perform a systematic review of the literature to assess the translation of stem cell therapy to explore what research had been carried out at each of the stages of translation from bench-top (in vitro), animal (pre-clinical) and human studies (clinical) and assemble an evidence-based cascade for the responsible introduction of stem cell therapy for cartilage defects.

Main body of abstract

This review was conducted in accordance to PRISMA guidelines using CINHAL, MEDLINE, EMBASE, Scopus and Web of Knowledge databases from 1st January 1900 to 30th June 2015. In total, there were 2880 studies identified of which 252 studies were included for analysis (100 articles for in vitro studies, 111 studies for animal studies; and 31 studies for human studies). There was a huge variance in cell source in pre-clinical studies both of terms of animal used, location of harvest (fat, marrow, blood or synovium) and allogeneicity. The use of scaffolds, growth factors, number of cell passages and number of cells used was hugely heterogeneous.

Short conclusions

This review offers a comprehensive assessment of the evidence behind the translation of basic science to the clinical practice of cartilage repair. It has revealed a lack of connectivity between the in vitro, pre-clinical and human data and a patchwork quilt of synergistic evidence. Drivers for progress in this space are largely driven by patient demand, surgeon inquisition and a regulatory framework that is learning at the same pace as new developments take place.
Abkürzungen
ACI
Autologous chondrocyte implantation
AMIC
Autologous matrix-enhanced chondrogenesis
AOFAS
American Orthopaedic Foot & Ankle Society
FACS
Fluorescence-activated cell sorting
HA
Hydroxyapatite
IKDC
International Knee Documentation Committee
iPSCs
Induced pluripotent stem cells
KOOS
Knee and Osteoarthritis Outcome Score
MACI
Matrix-induced autologous chondrocyte implantation
MeSH
Medical Subject Headings
MSC
Mesenchymal stem cells
OA
Osteoarthritis
PBS
Phosphate-buffered saline
PBSC
Peripheral blood stem cells
PRP
Platelet rich plasma
qPCR
Real-time polymerase chain reaction
RCT
Randomised controlled trial
VAS
Visual analogue scale
WOMAC
Western Ontario and McMaster Universities Arthritis Index

Background

Articular cartilage is a highly specialised tissue acting as a shock absorber, enabling synovial joints to articulate with low frictional forces. Due to its avascular, aneural and alymphatic state, it has a limited repair potential [1]. Surgical options to manage damaged articular cartilage include arthroscopic debridement [25], bone marrow stimulation techniques [68], chondrocyte implantation [913], osteochondral autografts (mosaicplasty) [2, 14, 15], osteochondral allograft [1618] and, in the presence of osteoarthritis, joint replacement [19].
Bone marrow stimulation techniques, such as microfracture, are the most frequently used method in clinical practice for treating small symptomatic lesions of the articular cartilage [68]. However, the resulting tissue has shown to be a mixed fibrocartilage tissue [2022] with varying amounts of type II collagen [8, 21, 23, 24] and inferior to native hyaline cartilage. Fibrocartilage is vulnerable to shear stresses and prone to breaking down over time [20]. Subchondral osseous overgrowth has also been reported after microfracture [25, 26]. Osteochondral grafts can lead to donor site morbidity and healing seams at the recipient site [27, 28]. Autologous chondrocyte implantation (ACI) [9, 10] and its later evolution, matrix-induced autologous chondrocyte implantation (MACI), offered great promise with 80% of patients showing good or excellent results at 10 years [29] but at best results in hyaline-like repair and has experienced complications such as graft failure, periosteal hypertrophy and delamination [30, 31]. In addition, it has also been reported that cells may lose their phenotype during expansion [32, 33].
There is therefore a growing interest in regenerative medicine, which can broadly be thought of as two main types: cell therapy, where cells are injected directly into the blood or into tissues, and tissue engineering, where cell-scaffold combinations are used to repair or regenerate tissues.
Stem cells are cells that have the ability to divide and develop into many different cell types in the body and can be categorised as pluripotent and multipotent. Pluripotent stem cells are often harvested from embryonic sources and can develop into any type of cell in the body whereas multipotent stem cells are generally taken from adults and can divide and develop into a more limited range of cell types. When stem cells divide, the new cells can either remain stem cells or develop into a new type of cell with a more specific function (Table 1).
Table 1
Table describing the three main properties of stem cells
Stem cell properties
 
• They are unspecialized (“blank slates” that can become specific types of cells).
 
• They can develop into specialized cell types (cells that do specific work in the body).
 
• They are capable of surviving over long periods and divide to make additional stem cells.
 
Mesenchymal stem cells (MSCs) are a form of multipotent cells that may offer an alternative to cartilage repair techniques not hampered by availability and donor site morbidity.
The introduction of stem cell therapies into clinical practice however is a form of translational research, which as per any “bench-to-bedside” pathway now has enormous governance issues [34, 35] and is highly regulatory across four phases (Table 2) and by the Tissues and Cells Directive (2004/23/EC) https://​www.​hta.​gov.​uk/​policies/​eu-tissue-and-cells-directives.
Table 2
Description of the different phases of clinical trials
Clinical trial phases (http://www.nlm.nih.gov/services/ctphases.html)
 
Phase I: Safety Studies or First-In-Man. Researchers test a new drug or treatment in a small group of people for the first time to evaluate its safety, determine a safe dosage range, and identify side effects.
 
Phase II: Uncontrolled Efficacy Studies. The drug or treatment is given to a larger group of people to see if it is effective and to further evaluate its safety.
 
Phase III: Randomised Clinical Trials. The drug or treatment is given to large groups of people to confirm its effectiveness, monitor side effects, compare it to commonly used treatments, and collect information that will allow the drug or treatment to be used safely.
 
Phase IV: Post-Market Surveillance. Studies are done after the drug or treatment has been marketed to gather information on the drug’s effect in various populations and any side effects associated with long-term use.
 
Many published reviews on cartilage repair only list human clinical trials [13, 3646], underestimating the wealth of basic sciences and animal studies that are precursors to future research and may be relevant in clinical practice further down the line. In addition, true translation would imply that all of the clinical studies would have supporting pre-clinical data.
We therefore set out to perform a systematic review of the literature to assess the translation of stem cell therapy to explore what research had been carried out at each of the stages of translation from bench-top (in vitro), animal (pre-clinical), and human studies (clinical) and assemble an evidence-based cascade for the responsible introduction of stem cell therapy for cartilage defects. In particular, we wanted to focus on the key burning questions pertaining to cartilage repair such as cell source, dosage (how many cells should be used), requirement for scaffolds and the role for extrinsic growth factors.

Main text

Search methodology

This review was conducted in accordance to PRISMA guidelines [47] using CINHAL, MEDLINE, EMBASE, Scopus and Web of Knowledge databases from 1st January 1900 to 30th June 2015.
The keywords used in the selection were “(“mesenchymal stem cells”[All Fields] OR “mesenchymal stem cells”[MeSH Terms] OR “mesenchymal”[All Fields] OR “stem cells”[All Fields] OR “Stem Cells”[MeSH Terms] OR “MSC”[All Fields]) AND (“Articular Cartilage”[MeSH Terms] OR “articular”[All Fields] OR “cartilage”[All Fields] OR “cartilage”[MeSH Terms]) AND (“healing”[All Terms] OR “repair”[All Terms] OR “Regeneration”[MeSH Terms] OR “regeneration”[All Fields] OR “tissue engineering”[MeSH Terms] OR “tissue engineering”[All Fields]) AND (“defect”[All Terms]) AND (“chond*”[All Terms])”.
All review and non-English studies were excluded. For analysis, only original research studies were included. Any duplicates were excluded. Initially, KM and JS independently screened studies’ title and abstract. Those included had the full text reviewed. Any disparities were discussed with the senior author (AJG). The references of eligible studies were also searched and included where relevant.
Unpublished trial databases (e.g. ClinicalTrials.gov) were reviewed as the grey literature using popular search engines, including Google. The keywords used for registered clinical trials in clinical trial databases were “stem cells”, “cartilage” and “orthopaedics”.
Eligible studies were drafted into tables tabulating the key data.

Results

The initial search identified 2880 study articles, of which 239 were included for analysis. The PRISMA flow diagram is shown in Fig. 1.

In vitro studies

MSC source
A list of cell sources used in the in vitro studies is shown in Table 3. The commonest being human MSCs (66%) followed by rabbit MSCs (15%). The majority of the studies used bone marrow-derived MSCs (63%) followed by adipose tissue (33%). Two studies used commercial cell lines [48, 49].
Table 3
Cell species and cell sources
Cell species
No. of studies
References
Cell Source
No. of studiesa
References
Human
73
[48, 50, 52, 53, 168236]
Bone marrow
62
[48, 5053, 164, 168, 170173, 177180, 182185, 187, 188, 192, 195197, 203, 206210, 212, 216, 217, 219, 221, 223, 227, 230, 232235, 237255]
Rabbit
17
[240242, 246, 249, 252, 255265]
Adipose
36
[66, 169, 175, 176, 181, 186, 189, 193, 194, 199, 201, 202, 211, 214, 216, 218220, 224, 228, 229, 231, 235, 242, 256, 257, 260269]
Bovine
5
[51, 164, 243, 245, 270]
Synovium
9
[174, 191, 200, 213, 222, 226, 258, 259, 270]
Rat/mouse
5
[239, 250, 266, 269, 271]
Umbilical cord blood
3
[205, 236, 190]
Porcine
3
[247, 248, 268]
Commercial cell line
2
[215, 271]
Equine
3
[238, 253, 254]
Placental
2
[198, 225]
Goat
1
[244]
Embryonic
1
[216]
Ovine
2
[237, 251]
Not stated
0
 
Not stated
1
[267]
   
aSome studies used cells from more than one cell source
Scaffold
Within the in vitro studies, 26 different types of natural scaffold and 9 types of synthetic scaffolds were identified with a further 18 different types of hybrids, the most popular being a fibrin-polyurethane scaffold (Table 4).
Table 4
Types of scaffolds
Number of studies using types of scaffold
Natural
Synthetic
Hybrid
Growth factor combined
None used
47
14
22
6
29
Scaffold
No. of studies
References
Types of scaffolds used
Natural scaffolds
 Type I collagen hydrogel
6
[185, 190, 211, 226, 241, 251]
 Agarose hydrogel
4
[53, 247, 248, 268]
 Alginate bead
3
[223, 231, 271]
 Fibrin hydrogel
3
[208, 211, 263]
 Silk fibroin
3
[198, 216, 256]
 Chitosan microspheres
2
[260, 262]
 Hyaluronic acid
2
[195, 237]
 Cartilage-derived matrix
2
[193, 238]
 K-carrageenan
2
[169, 199]
 Chitosan
2
[168, 216]
 Hyaluronic acid hydrogel
2
[164, 245]
 Gelatin-based scaffold
2
[176, 233]
 Devitalised cartilage ECM
1
[220]
 Bead in bead alginate polysaccharide capsules
1
[221]
 Atelocollagen gel
1
[225]
 Fibrin disk
1
[254]
 Methacrylated hyaluronic acid
1
[164]
 Gelatin microspheres
1
[260]
 Decellularised cell matrix
1
[191]
 Collagen type I microspheres
1
[52]
 Alginate microbeads
1
[266]
 Alginate disks
1
[270]
 Platelet rich plasma
1
[242]
 Free oligosaccharide chondroitin sulphate C
1
[205]
 Collagen type I sponge
1
[237]
 3D printed chitosan
1
[181]
Synthetic scaffolds
 Polycaprolactone
3
[197, 207, 209]
 PLGA
3
[194, 204, 257]
 Polylactic acid
2
[230, 232]
 PVA
1
[244]
 PGA
1
[178]
 Poly-DL-lactide-co-glycolide
1
[194]
 Polylactide-co-caprolactone
1
[214]
 GFOGER modified PEG hydrogel
1
[183]
 OPF hydrogel
1
[240]
Hybrid scaffolds
 Fibrin–polurethane hydrogel
4
[50, 188, 192, 267]
 Esterified hyaluronan and gelatin polymer
2
[212, 255]
 TruFit CB (PLGA, calcium sulphate and polycolide)
1
[187]
 PCL–HA bilayer
1
[243]
 PEGDG–crosslinked hyaluronic acid
1
[202]
 Polylactic acid–alginate
1
[232]
 Sodium alginate–hyaluronic acid
1
[189]
 Chitosan–collagen type I
1
[258]
 Polyvinylalcohol–polycaprolactone
1
[246]
 Tricalcium phosphate-collagen-hyaluronan
1
[180]
 Poly-L-lactic acid–hydroxyapatite
1
[215]
 Collagen type I–polylactic acid
1
[217]
 Polylactic acid–polyglycolic acid with fibrin
1
[261]
 Collagen–polyglycolic acid
1
[252]
 Chondroitin sulphate C–collagen type II
1
[236]
 Fibrin hydrogel with chondroitin sulphate
1
[263]
 Chitosan-demineralised bone matrix
1
[239]
 Alginate foam-chondroitin sulphate
1
[170]
Growth factor combined with scaffolds
 TGF-β1-loaded microspheres with chitosan microspheres
1
[262]
 TGF-β1 releasing chitosan-collagen hydrogel
1
[174]
 PEOT/PBT TGF-β1 loaded scaffolds
1
[173]
 TGF-β1-activated chitosan/gelatin
1
[249]
 PLGA nanospheres with TGF-β1
1
[172]
 TGF-β1 loaded Gelatin Microspheres
1
[175]
Growth factors
The commonest used growth factors were TGF-β and the bone morphogenetic protein (BMP) family. A list of growth factors used can be seen in Table 5.
Table 5
Number of in vitro studies using different growth factors
Growth factor
No. of studies (%)
References
Growth factor
No. of studies (%)
References
TGF-β1
48 (44%)
[50, 169175, 189, 190, 192, 193, 195, 199, 202, 208, 210, 211, 213, 214, 216, 217, 220, 222224, 228, 230232, 234, 235, 244, 246, 249, 252256, 258, 260263, 266, 267, 270]
SOX-5
1 (1%)
[204]
TGF-β3
32 (29%)
[51, 162, 164, 168, 177, 181184, 197, 200, 205207, 218, 223225, 227, 237, 239, 240, 245, 247, 248, 250, 251, 257, 259, 267, 268, 270]
SOX-6
1 (1%)
[204]
BMP-2
13 (12%)
[188, 202, 213, 219, 225227, 229, 264, 265, 267, 270, 271]
WNT3A
1 (1%)
[171]
FGF
9 (8%)
[171, 183, 193, 197, 198, 213, 225, 246, 258]
IL-1
1 (1%)
[197]
IGF-1
7 (6%)
[179, 184, 192, 213, 224, 254, 265]
EGF
1(1%)
[193]
BMP-6
7 (6%)
[181, 216, 219, 224, 227, 250, 266]
OP-1
1 (1%)
[222]
TGF-β2
4 (4%)
[209, 219, 238, 270]
AA2P
1 (1%)
[266]
GDF-5
3 (3%[
[48, 186, 269]
IL-10
1 (1%)
[178]
SOX-9
2 (2%)
[204, 221]
TNFα
1 (1%)
[178]
BMP-4
2 (2%)
[227, 271]
PRP
1 (1%)
[242]
DEX
2 (2%)
[224, 266]
IWP2
1 (1%)
[171]
BMP-7
1 (1%)
[219]
None
15 (14%)
[52, 176, 180, 185, 187, 191, 194, 196, 201, 212, 215, 233, 236, 241, 243]
PDGF
1 (1%)
[202]
   
Cell seeding and passage
There was wide heterogeneity in cell seeding density and there appeared to be no standard form of measurement. Li et al. [50] examined three different seeding densities: 2, 5 and 10 × 106 cells/scaffold, and found that scaffolds seeded with 5 × 106 cells per scaffold induced the highest chondrogenesis; however, other groups [5153] found that a higher seeding density results in better chondrogenesis.
Apart from 26 studies which did not state cell passage number, most studies used MSC of an early passage, anything between uncultured fresh (passage zero (P0) and five times passaged cells (P5). One study used cells of P6 [54], and another study used cells between P4 and P7 [48]. No relationship was apparent between chondrogenesis and number of passages.
Length of study
The length of each in vitro study can be seen in Table 6. The majority of studies were short-term models; 27 studies (25%) ended between 1 and 2 weeks, 35 studies (33%) ended between 2 and 3 weeks and 15 studies (14%) ended between 3 and 4 weeks.
Table 6
Length of studies
Length of study
No. of studies
References
Up to 1 week
9
[172, 203, 210, 212, 224, 229, 239, 266, 270]
1–2 weeks
27
[50, 170, 174, 178, 182, 189, 192, 194, 198, 202, 215, 218, 220, 223, 228, 234, 235, 237, 240, 249, 254, 260265]
2–3 weeks
36
[52, 53, 168, 169, 173, 175, 179, 180, 183186, 190, 191, 195, 196, 199, 200, 204, 205, 209, 213, 217, 225, 226, 230, 232, 233, 236, 246, 250, 256, 258, 269, 271]
3–4 weeks
15
[51, 176, 181, 188, 193, 201, 211, 216, 219, 221, 241, 251, 253, 255, 257]
4–5 weeks
7
[171, 177, 206, 214, 231, 259, 267]
5–6 weeks
10
[48, 187, 208, 222, 238, 244, 247, 248, 252, 268]
6–7 weeks
1
[207]
7–8 weeks
1
[197]
8–9 weeks
3
[164, 243, 245]
Not stated
1
[242]
Method of assessment
A range of techniques was used to assess chondrogenesis within the in vitro studies. These techniques consisted of histology, immunohistochemistry, qPCR, biochemical analysis, imagery and mechanical testing. The techniques used are summarised in Table 7.
Table 7
Types of techniques used to assess chondrogenesis of MSCs
Type of techniques
No. of studies (%)
References
Histology
87 (79%)
[48, 5053, 164, 168170, 173175, 177179, 181187, 191195, 197201, 204211, 213217, 219222, 226, 229, 230, 232238, 240248, 250, 252264, 267271]
Immunohistochemistry
78 (71%)
[48, 50, 52, 53, 168171, 173175, 178183, 185191, 193, 194, 197, 198, 201, 203205, 207, 212215, 217, 218, 220, 221, 224, 226, 228238, 241, 242, 244, 246248, 250259, 264, 265, 267271]
qPCR
70 (64%)
[53, 168, 169, 173, 174, 176, 178186, 188, 190, 192194, 196, 199, 200, 202205, 207209, 211, 214, 216220, 222232, 235, 236, 239, 240, 242, 246, 249251, 256, 258, 259, 261263, 265267, 269271]
Biochemical analysis
64 (58%)
[48, 5052, 164, 168, 170172, 176, 177, 179, 180, 182184, 188, 189, 191, 192, 197, 199, 200, 202, 204, 205, 209, 212, 214, 216219, 222224, 226, 227, 233240, 244, 245, 247249, 252, 254, 257, 260266, 268270]
Imaging (confocal, SEM, TEM)
24 (22%)
[52, 172, 176, 180, 185, 187, 194, 198, 208, 215217, 225, 226, 230, 232, 241, 242, 249, 252, 255, 262, 263, 265]
Mechanical testing
15 (14%)
[51, 52, 164, 169, 175, 193, 197, 207, 220, 245, 247, 248, 256, 257, 268]

Animal studies (pre-clinical)

One hundred eleven animal studies were included of which 109 were controlled laboratory studies, one was a pilot study [49] and one was a longitudinal case study on a race horse [55]. The commonest animal studied with 59 studies was rabbit (53%). The different species of animals studied is shown in Table 8.
Table 8
Different species of animals used to assess reparative effect of MSCs on cartilage defect
Animals
No. of studies (%)
References
Rabbits
57 (51%)
[49, 54102, 134, 150154, 160, 161, 207, 272324]
Pigs
16 (14%)
[61, 62, 6872, 87, 90, 153, 273, 276, 279, 290, 308310]
Rats
13 (12%)
[60, 7882, 91, 152, 160, 278, 286, 311, 312]
Sheep
8 (7%)
[89, 272, 282, 283, 313316]
Goats
5 (5%)
[49, 95, 100, 101, 318]
Horses
4 (4%)
[55, 96, 98, 317]
Dogs
4 (4%)
[86, 97, 151, 287]
Monkeys
2 (2%)
[319, 320]
Guinea pigs
1 (<1%)
[281]
Donkeys
1 (<1%)
[57]
Defect
The size of the defect varied from 2 to 25 mm2 in the smaller animals and from 1 to 64 mm2 in the larger animals. All but two studies [56, 57] used the knee for defect creation.
Stem cell type
Bone marrow-derived stem cells were used in 84 studies (75%). Thirteen studies (11%) used adipose stem cells [54, 5869], six (5%) used synovia [7075] and three (2%) used periostium-derived MSCs [7678]. Three studies (3%) used embryonic stem cell-derived MSCs [7981] whereas 2 studies (2%) used muscle-derived MSCs [82, 83]. One group showed promising results of allogenic MSCs in a rabbit model when compared to autologous cells, although numbers were small [84, 85]. Another used compared autologous chondroprogenitor cells and allogenic chondroprogenitor cells against controls in an equine model and reported that repair tissue quality in the allogenic cell group was not superior to that in the control (fibrin only) group and also showed poorer radiographic changes in the allogenic group [23].
Cell culture, dose and delivery
There was much variation in the number of cells implanted and the number of cell passages from 3–10 or more [79, 86].
The number of cells varied from 4 × 103 – 1 × 1010. The majority of studies used between 106 and 108 cells. Some did not specify the number of cells implanted. Two studies suggested that improved chondrogenesis occurs with a higher implanted cell number [75, 87], although others suggested that the high cell numbers increase the risk of synovitis [75] and synovial proliferation [88].
The cells were transplanted into the defect both as cell therapy (injection directly into the joint) (17 studies, 15%) or by tissue engineering (cell-scaffold combinations) (94 studies, 85%). Fifteen studies [49, 65, 72, 75, 81, 86, 8997] used a mixture of solutions prepared from hyaluronic acid [65, 92, 9497], phosphate buffer solution [91], plasma [75], basal medium with chondrogenesis [89], collagen acid [93], sodium alginate [86] or a growth factor medium [90]. Two studies used MSCs only [49, 72].
Scaffold
Ninety-two studies (82%) used a scaffold. The material used was a synthetic polymer either collagen based, fibrinogen glue or a synthetic protein (e.g. rHuBMP-2) in 62 (56%) studies (Table 9).
Table 9
Table showing the types of scaffold used in animal studies
Scaffold type
No. of studies
References
No Scaffold
19 (17%)
[49, 54, 61, 70, 7275, 81, 86, 8991, 97, 100, 102, 280282, 284]
Poly (lactide-co-glycoside) PLGA
17 (16%)
[56, 59, 62, 63, 83, 88, 150, 153, 160, 277, 285, 286, 289292, 316]
Fibrin/Fribrin glue
11 (9%)
[55, 64, 7678, 152, 278, 293, 308, 317, 318]
Hydrogel
9 (8%)
[65, 69, 81, 94, 279, 288, 314, 321, 323]
Collagen
9 (8%)
[79, 80, 134, 276, 299, 301, 309, 320, 322]
Hyaluronic acid
7 (6%)
[57, 92, 95, 96, 273, 304, 324]
Alginate beads
4 (3%)
[65, 84, 101, 294]
Tissue membrane
4 (3%)
[82, 98, 303, 305]
Polyglycolic acid
3 (3%)
[99, 161, 274]
PGA/PLA
3 (3%)
[68, 290, 296]
Hylauronan crosslinked matrix
2 (2%)
[154, 297]
Poly-L-lactide-co-caprolactone
2 (2%)
[275, 300]
Polycaprolactone cartilage (PCL)
2 (2%)
[87, 272]
Animal-origin osteochondral plug scaffold
2 (2%)
[272, 298]
Chitosan microspheres and fibrin glue
1 (<1%)
[60]
Gel carries (collagen/HA/Fibrogen)
1 (<1%)
[71]
Polychoxanone/poly(vinyl alcholo) PDO/PVA
1 (<1%)
[302]
Cartilage aggregate
1 (<1%)
[306]
Collagen/glycosaminoglycan porous titanium biphasic scaffold
1 (<1%)
[151]
Articular chondrocyte seeded matrix associated autologous chondrocyte transplant (MACT)
1 (<1%)
[313]
MSC-ADM (accellulo-dermal matrix)
1 (<1%)
[319]
Hyaff-11 scaffold
1 (<1%)
[295]
Porous-gelatin-chonroitin hyaluronate
1 (<1%)
[291]
Bone protein 7 PCL
1 (<1%)
[66]
Human acellular amniotic membrane
1 (<1%)
[307]
Pluronic-F 127
1 (<1%)
[102]
Tricalcium phosphate
1 (<1%)
[315]
Agarose
1 (<1%)
[311]
GCH-GCBB
1 (<1%)
[93]
ACHMS (atelocollagen honeycomb-shaped membrane)
1 (<1%)
[58]
Magnet
1 (<1%)
[310]
Human cartilage extra cellular matrix 3D porous acellular
1 (<1%)
[67]
Growth factors
Thirty-two studies (29%) assessed the effect of growth factors on MSC chondrogenesis. Seventeen out of 38 (44%) used TGF-β1/3 (Table 10), the majority of which show a positive effect on chondrogenesis.
Table 10
Table showing growth factors used in animal studies
Growth factor
No. of studies
References
TGF-β3/1/2
17 (15%)
[56, 65, 66, 70, 76, 85, 90, 100, 280, 282, 285, 287, 290, 291, 309, 311, 323]
CDMP–1
2 (2%)
[56, 134]
FGF-2
2 (2%)
[90, 304]
Ad-hTGF-B1
1 (<1%)
[321]
AdBMP–2
1 (<1%)
[78]
chABC
1 (<1%)
[74]
PRP
1 (<1%)
[75]
Gene modified MSCs (gene modification to BcL-xL gene)
1 (<1%)
[299]
hiGF-1-DNA
1 (<1%)
[101]
AdIGF–1
1 (<1%)
[78]
rHuBMP–2
1 (<1%)
[82]
Ham-F-12
1 (<1%)
[303]
NaO11
1 (<1%)
[277]
NSC23766-Rac1 inhibitor
1 (<1%)
[60]
Associated procedures
Ten of the studies compared MSC treatment against other surgical modalities such as debridement [55], microfracture [49, 91, 96, 98, 99] and mosaicplasty [77, 100102].
Outcome measures
There were a variety of outcome measures used to analyse the results of the studies. The majority of studies (79%) used evidence of hyaline-like cartilage as being a positive outcome (Tables 11 and 12).
Table 11
Outcome measures used in animal studies (some studies used more than one outcome measure)
Outcome score
No. of studies using the score (%)
References
Histology scores
111 (100%)
[49, 54102, 134, 150154, 160, 161, 272324]
International Cartilage Repair Society Score
26 (23%)
[49, 60, 61, 63, 66, 69, 72, 74, 79, 89, 92, 94, 98, 99, 272, 282, 283, 289, 305, 306, 310, 313, 314, 316, 319, 324]
Wakitani score
21 (19%)
[58, 62, 67, 68, 72, 73, 80, 82, 97, 151, 273, 277, 279, 284, 285, 290, 299, 304, 310, 321]
O’Driscoll score
2018%
[49, 71, 81, 84, 85, 93, 100, 160, 272, 276, 290, 296298, 302, 306, 308, 313, 314, 322]
Functional scores/mechanical
11 (10%)
[55, 57, 62, 67, 69, 81, 101, 277, 287, 290, 315]
MRI scores
5 (5%)
[63, 69, 96, 101, 316]
Arthroscopy scores
5 (5%)
[72, 96, 310, 317, 318]
Macroscopic osteoarthritis score
3 (3%)
[57, 281, 295]
Pineda score
3 (3%)
[290, 293, 309]
Schreiber score
2 (2%)
[101, 300]
Britternberg score
2 (2%)
[84, 85]
Slochagg score
1 (<1%)
[300]
Moran score
1 (<1%)
[64]
Gill score
1 (<1%)
[95]
Table 12
Analysis technique used on repaired tissue
Analysis used
No. of studies (%)
References
Hyaline-like cartilage
88 (79%)
[49, 5456, 58, 59, 61, 62, 6469, 7173, 75, 76, 7889, 92, 95, 97, 98, 100, 101, 134, 150152, 154, 160, 161, 273280, 285302, 304, 305, 307, 309, 310, 312, 314324]
Collagen type II
84 (76%)
[54, 5659, 62, 6573, 7588, 90, 91, 9396, 98, 100102, 134, 150154, 160, 161, 272276, 278282, 284288, 292, 294296, 300, 302306, 308, 309, 311, 313315, 317319, 321, 323]
Cluster Chondrocytes
34 (31%)
[57, 60, 62, 63, 72, 74, 77, 78, 80, 81, 83, 84, 91, 97, 102, 151, 152, 160, 161, 273, 276, 280, 281, 283, 291, 292, 296, 297, 304, 312, 318, 319, 322, 324]
Glycosaminoglycan
40 (36%)
[49, 62, 65, 6771, 7375, 81, 85, 87, 94, 96101, 160, 272, 274, 279, 282, 286, 288, 290, 291, 296, 300, 301, 308, 309, 311, 312, 315, 319, 323]
Genes
22 (20%)
[56, 60, 61, 63, 64, 66, 78, 80, 82, 90, 94, 96, 134, 275, 277, 283, 285, 294, 311, 316, 321, 323]
Proteoglycan
8 (7%)
[56, 63, 84, 95, 98, 287, 294, 295]

Human studies (clinical)

Thirty-one published studies by 15 different groups looked at clinical applications of MSCs. One used allogenic stem cells [103] and the rest autologous stem cells. The types of studies can be seen in Tables 13 and 14.
Table 13
Number of publications for each study type and phase
Category
No. of studies (total 28)
References
Phases of clinical studies
 Pilot/feasibility study incl. case report
15 (54%)
[104108, 118, 119, 122, 124129, 133]
 Phase 1 (safety assessment)
8 (26%)
[109112, 116, 123, 130, 131]
 Phase 2 (efficacy assessment)
8 (26%)
[103, 113115, 117, 120, 121, 132]
 Phase 3 (large scale efficacy assessment through a multi-centre RCT)
0 (0%)
 Phase 4 (post-market surveillance)
0 (0%)
Table 14
Summary of the published clinical studies
Category
No. of studies
References
Cell source
 Bone marrow
22 (71%)
[103105, 109, 111113, 115118, 120, 122128, 130132]
 Adipose
5 (16%)
[106108, 110, 114]
 Peripheral blood
2 (6%)
[119, 121]
 Synovium
2 (6%)
[129, 133]
Cell delivery
 Arthroscopic implantation
  Hyaluronic acid membrane
2 (6%)
[117, 130]
  Hyaluronic acid with fibrin glue or platelet gel
2 (6%)
[116, 128]
  Polyglycolic acid/hyaluronan
2 (6%)
[127, 131]
  Collagen with platelet gel
1 (3%)
[116]
  Fibrin glue
1 (3%)
[108]
  HYAFF 11 scaffold
1 (3%)
[132]
  Acetate Ringer solution
1 (3%)
[133]
  Unspecified
1 (3%)
[107]
Intra-articular injection
  PBS only
2 (6%)
[104, 110]
  PBS with HA
2 (6%)
[119, 121]
  Autologous serum
2 (6%)
[115, 123]
  Ringer lactate solution
3 (10%)
[103, 111, 112]
  PBS with serum albumin
1 (3%)
[105]
  HA and PRP
1 (3%)
[106]
  PRP
1 (3%)
[114]
  Commercial serum
1 (3%)
[109]
Transplantation by open surgery
  Collagen
6 (21%)
[103, 113, 118, 122, 124, 126, 129]
  Ascorbic acid-mediated sheet
2 (7%)
[120, 123]
  Fibrin glue
1 (4%)
[125]
Cell dose
 Less than 10 million
8 (26%)
[105, 107, 108, 114, 120, 122, 124, 129]
 10–20 million
5 (16%)
[113, 118, 119, 123, 125]
 Over 20 million
7 (23%)
[103, 104, 109112, 133]
 Unspecified
11 (35%)
[106, 115117, 121, 126128, 130132]
Follow-up
 Up to 6 months
4 (13%)
[104106, 110]
 Up to 12 months
6 (19%)
[103, 109, 111, 124, 125, 127]
 Up to 2 years
11 (35%)
[107, 113116, 120, 121, 128131]
 Up to 3 years
7 (23%)
[108, 112, 117, 119, 122, 126, 132]
 Over 3 years
2 (6%)
[118, 133]
Assessments
 Radiology (MRI, X-ray)
24 (77%)
[103106, 109112, 115117, 119, 121125, 127133]
 Arthroscopic assessment incl. histology
17 (54%)
[107, 108, 113, 116122, 124126, 130133]
 IKDC
10 (32%)
[107, 108, 115, 121, 122, 126, 128, 130132]
 VAS pain
12 (39%)
[103106, 109112, 114, 129, 131, 132]
 Tegner activity scale
8 (26%)
[107, 108, 114, 115, 129, 131133]
 Lysholm
6 (19%)
[114, 115, 125, 128, 131, 133]
 KOOS
5 (16%)
[126, 128130, 132]
 Function (no scoring systems or unspecified)
4 (13%)
[104106, 109]
 ICRS cartilage injury evaluation package
3 (10%)
[120, 123, 125]
 Clinical symptoms/outcomes (no scoring system or unspecified)
3 (10%)
[105, 109, 124]
 (Revised) Hospital for special surgery knee-rating scale
2 (6%)
[113, 125]
 Functional Rating Index
2 (6%)
[104, 106]
 WOMAC
5 (16%)
[103, 109112]
 AOFAS score
2 (6%)
[112, 116, 117]
 Knee Society Score
1 (3%)
[110]
 Harris Hip Score
1 (3%)
[112]
Concomitant procedures
 Subchondral bone marrow stimulation (multiple perforation, drilling, abrasion chondroplasty)
11 (35%)
[113, 115, 118, 119, 121123, 125, 127, 128, 131]
 Debridement, synovectomy, excision of degenerative tears (no subchondral bone marrow stimulation)
8 (26%)
[107, 108, 114, 116, 117, 124, 130, 133]
 ACL reconstruction, meniscus repair, osteotomy, or patella alignment, ACL calcification removal, trochlear resurfacing, osteochondral fragment fixation
8 (26%)
[115, 123, 126, 129133]
 None
6 (19%)
[103, 105, 106, 110112]
 Not specified
3 (10%)
[104, 109, 120]
Previous procedures
 Microfractures/multiple perforation/multiple drilling
6 (19%)
[104, 116, 117, 122, 125, 130]
 Menisectomy
6 (19%)
[103, 111, 124, 129, 131, 133]
 ACL reconstruction
4 (13%)
[103, 111, 131, 133]
 Multiple (microfracture, debridement)
1 (3%)
[119]
 ACI
2 (6%)
[116, 117]
 None
6 (19%)
[106108, 110, 114, 118]
 Not specified
9 (29%)
[105, 109, 112, 115, 120, 121, 126, 128, 132]
PBS phosphate-buffered saline, HA hyaluronic acid, PRP plate-rich-plasma, RCT randomised controlled study, KOOS Knee and Osteoarthritis Outcome Score, IKDC score International Knee Documentation Committee Score, WOMAC the Western Ontario and McMaster Universities Arthritis Index, AOFAS the American Orthopaedic Foot & Ankle Society
There were 52 unpublished clinical trials, majority of which are early phase studies (I–II; 63%) and only 5 trials were phase II/III. Table 15 shows a summary of these clinical trials.
Table 15
Clinical trials (unpublished/on-going) registered in ClinicalTrials.gov
Title
Cell source
Country
Clinical trial phase
Condition
Study design
Enrolment
Follow-up
Arm(s)
Cell delivery
Primary outcomes
Study status (on 8.3.2016)
ClinicalTrials.gov Identifier
Autologous cells
Mesenchymal Stem Cells in Knee Cartilage Injuries
Bone marrow
Jordan
II
Advanced knee articular cartilage injury
Non-randomized parallel assignment; double blind
13
12 months
Culture expanded MSCs alone vs. MSC with platelet lysate
Intra-articular injection
Therapeutic benefit
Completed in August 2015; no publication found
NCT02118519
Adult Stem Cell Therapy for Repairing Articular Cartilage in Gonarthrosis
Bone marrow
Spain
I/II
Gonarthrosis grade 2–3
Open label; single group assignment
15
12 months
Culture expanded MSCs (40 million cells)
Articular injection
Feasibility/safety
Completed in January 2013; no publication found
NCT01227694
Autologous Bone Marrow Mesenchymal Stem Cells Transplantation for Articular Cartilage Defects Repair
Bone marrow
UK
I/II
Knee articular cartilage defects
Randomized parallel assignment; double blind
10
12 months
MSCs (fresh or cultured unspecified)
Intra-articular injection
Change in WOMAC
Unknown
(estimated study completion date; July 2014)
NCT01895413
Mesenchymal Stem Cell for Osteonecrosis of the Femoral Head
Bone marrow
China
0
Osteochondritis of the femoral head
Open label single group assignment
15
5 years
Culture expanded MSC and bone marrow nuclear cells
Infusion through medial femoral circumflex artery, lateral femoral circumflex artery and obturator artery
Femoral head blood-supply artery angiographies; femoral head necrosis
Unknown
(estimated study completion date; August 2015)
NCT00813267
The Effects of Intra-articular Injection of Mesenchymal Stem Cells in Knee Joint Osteoarthritis
Bone marrow
Iran
II
Knee joint osteoarthritis
Single centre, randomised, placebo controlled, double blind
40
3 months
Culture-expanded MSCs vs. placebo
Intra-articular injection
Changes in WOMAC physical function and VAS pain
Completed in November 2012; no publication found
NCT01504464
Safety and Efficacy of Autologous Bone Marrow Stem Cells for Treating Osteoarthritis
Bone marrow
India
I/II
Knee OA Kellgren and Lawrence classification 3–4
Open label single group assignment; multi-centre
10
1 year
MSCs (fresh or culture-expanded unspecified)
Unknown
WOMAC pain score and safety
On-going
(estimated study completion date; January 2012)
NCT01152125
Treatment of Knee Osteoarthritis by Intra-articular Injection of Bone Marrow Mesenchymal Stem Cells
Bone marrow
Spain
I/II
Knee OA
Randomised parallel assignment; open label
30
12 months
Culture-expanded MSCs (10 million or 100 million cells) and hyaluronic acid (HyalOne®) vs. HyalOne®
Intra-articular injection
Pain and function (VAS, WOMAC, KOOS, EuroQol, SF-16, Lequesne), radiographic
On-going
(estimated study completion date; February 2015)
NCT02123368
Intra-Articular Autologous Bone Marrow Mesenchymal Stem Cells Transplantation to Treat Mild to Moderate Osteoarthritis
Bone marrow
Malaysia
II
Mild to moderate OA based on Kellgren-Lawrence radiographic classification
Randomised parallel assignment; open label
50
12 months
MSCs (fresh or culture-expanded unspecified) in hyaluronic acid “Orthovisc” vs. hyaluronic acid
Intra-articular implantation
Changes in cartilage thickness (MRI)
Unknown (estimated study completion date; March 2014)
NCT01459640
Treatment of Osteoarthritis by Intra-articular Injection of Bone Marrow Mesenchymal Stem Cells With Platelet Rich Plasma (CMM-PRGF/ART)
Bone marrow
Spain
I/II
Knee OA
Randomised parallel assignment; open label; multi-centre
38
12 months
Culture-expanded MSCs with PRP (PRGF®) vs. PRGF® only
Intra-articular injection
Pain and function (VAS, WOMAC, KOOS, EuroQol, SF-16, Lequesne), radiographic
On-going (estimated study completion date; June 2017)
NCT02365142
Mesenchymal Stem Cells Enhanced With PRP Versus PRP In OA Knee (MSCPRPOAK)
Bone marrow
India
I/II
Knee OA grade 1–2 Ahlbacks radiographic staging
Randomised parallel assignment double blinded
24
6 months
Culture-expanded MSCs (10 million cells) with autologous PRP vs. PRP only
Injected by lateral approach
VAS pain
Unknown
(estimated study completion date; June 2014)
NCT01985633
Side Effects of Autologous Mesenchymal Stem Cell Transplantation in Ankle Joint Osteoarthritis
Bone marrow
Iran
I
Severe ankle OA
Single group assignment open label
6
6 months
Culture-expanded MSCs
Intra-articular injection
Safety
Completed in September 2011; no publication found
NCT01436058
Human Autologous MSCs for the Treatment of Mid to Late Stage Knee OA
Bone marrow
Canada
I/II
Mid- to late-stage knee OA
Single group assignment, open label
12
1 year
Culture-expanded MSCs (1 million, 10 million or 50 million cells)
Injection
Safety
On-going
(estimated study completion date; February 2021)
NCT02351011
A Controlled Surveillance of the Osteoarthritic Knee Microenvironment With Regenexx® SD Treatment
Bone marrow
USA
NA
Knee OA Kellgren-Lawrence grade 2 or greater
Observational cohort study
20
6 weeks
Regenexx® SD (bone marrow concentrate)
Injection
Temporal median change in protein concentration or percentage of cellular subpopulations
On-going
(estimated study completion date; March 2016)
NCT02370823
The Effect of Platelet-rich Plasma in Patients With Osteoarthritis of the Knee
Bone marrow
Iran
III
Knee OA grade 2 and above (radiographic)
Randomised, parallel assignment, placebo controlled, double blinded
50
2 year
Bone marrow aspirate vs. placebo (saline)
Intra-articular injection
VAS pain, WOMAC physical activity, cartilage repair (MRI)
Completed in April 2014; no publication found
NCT02582489
Outcomes Data of Bone Marrow Stem Cells to Treat Hip and Knee Osteoarthritis
Bone marrow
USA
NA
Hip and knee OA
Observational cohort study
12
1 year
Bone marrow concentrate
Injection
VAS pain, Harris Hip Score or Knee Society Score, Physician Global Assessment
Completed in March 2014; no publication found
NCT01601951
Use of Autologous Bone Marrow Aspirate Concentrate in Painful Knee Osteoarthritis (BMAC)
Bone marrow
USA
II
Bilateral knee OA Kellgren-Lawrence grade 1–3
Randomised, parallel assignment, placebo controlled, single blinded
25
12 months
Bone marrow concentrate vs. placebo (saline)
Injection
Safety
On-going
(estimated study completion date; December 2016)
NCT01931007
Autologous Stem Cells in Osteoarthritis
Bone marrow
Mexico
I
Knee OA Kellgren-Lawrence radiographic scale grade 2–3
Randomised parallel assignment, open label
61
6 months
Hematopoietic stem cells (fresh) vs. acetaminophen (750 mg orally TID)
Infusion
Safety
Completed in May 2014; no publication found
NCT01485198
The Use of Autologous Bone Marrow Mesenchymal Stem Cells in the Treatment of Articular Cartilage Defects
Bone marrow
Egypt
Not given
An isolated osteochondral defect with no more than grade 1 or 2 Outerbridge
Single group assignment, open label
25
12 months
Culture-expanded MSCs
Open surgery or arthroscopy
Clinical scores and radiological images
Unknown
(estimated study completion date; December 2014)
NCT00891501
Autologous Transplantation of Mesenchymal Stem Cells (MSCs) and Scaffold in Full-thickness Articular Cartilage
Bone marrow
Iran
I
Full-thickness chondral defects
Single group assignment, open label
6
12 months
Culture-expanded MSCs mixed with collagen I scaffold
Unspecified
Knee cartilage defects
Completed in December 2010; no publication found
NCT00850187
“One-step” Bone Marrow Mononuclear Cell Transplantation in Talar Osteochondral Lesions (BMDC)
Bone marrow
USA
III
ICRS grade 3–4 Osteochondral lesions of the talar dome
Single group assignment, open label
140
24 months
Bone marrow concentrate
Arthroscopy
American Orthopaedic Foot and Ankle Society hindfoot score
On-going (estimated completion date; June 2016)
NCT02005861
Transplantation of Bone Marrow Stem Cells Stimulated by Proteins Scaffold to Heal Defects Articular Cartilage of the Knee
Bone marrow
France
0
Knee OA ICRS classification grade 4
Single group assignment, open label
50
1 year
Freshly isolated bone marrow mononuclear cells mixed with protein scaffold
Arthroscopy (one step procedure)
IKS
Unknown
(estimated completion date; December 2014))
NCT01159899
INSTRUCT for Repair of Knee Cartilage Defects
Bone marrow
The Netherlands
Not given
Knee articular cartilage defect
Single group assignment, open label; multi-centre
40
1 year
INSTRUCT scaffold (biodegradable scaffold seeded with autologous primary chondrocytes and bone marrow cells)
Arthrotomy
Safety and lesion filling
Completed in June 2014; no publication found
NCT01041885
HyaloFAST Trial for Repair of Articular Cartilage in the Knee (FastTRACK)
Bone marrow
Hungary
Not given
Knee articular cartilage defect
Randomised, parallel assignment, placebo controlled, single blinded, multi-centre
200
2 years
Hyalofast® scaffold with bone marrow aspirate concentrate vs. microfracture
One-step arthroscopic procedure
Changes in KOOS
On-going (estimated study completion date; June 2020)
NCT02659215
Autologous Adipose Stem Cells and Platelet Rich Plasma Therapy for Patients With Knee Osteoarthritis
Adipose
Vietnam
I/II
Idiopathic or secondary knee OA grade 2–3 radiographic severity
non-randomised unblinded
16
12 months
Stromal vascular fraction (10–50 million cells) and platelet rich plasma (PRP)
Injection
Safety
Completed in December 2015; no publication found
NCT02142842
Effectiveness and Safety of Autologous ADRC for Treatment of Degenerative Damage of Knee Articular Cartilage
Adipose
Russia
I/II
Knee OA (degenerative damage of knee articular cartilage)
Single group assignment, open label
12
24 weeks
Adipose-derived regenerative cells (ADRC) extracted using Celution 800/CRS System (Cytori Therapeutics, Inc.)
Intra-articular injection
Safety
On-going (estimated study completion date; December 2016)
NCT02219113
Autologous Adipose-Derived Stromal Cells Delivered Intra-articularly in Patients With Osteoarthritis
Adipose
USA
I/II
OA
Single group assignment, open label, multi-centre
500
6 months
MSCs in PRP
Intra-articular injection
Pain score, functional rating index, visual analogue scale (VAS), physical therapy (PT) and range of motion (53), quality of life scores, reduction in analgesics, adverse events
On-going (estimated study completion date; December 2016)
NCT01739504
Mesenchymal Stem Cell Treatment for Primary Osteoarthritis Knee
Adipose
Taiwan
I
Bilateral primary OA Kellgren and Lawrence grade 2–3 as determined by X-ray
Single group assignment, open label,
10
12 months
MSCs (8–10 million cells)
Intra-articular injections
Safety
On-going (estimated study completion date; December 2016)
NCT02544802
Autologous Adipose Tissue-Derived Mesenchymal Progenitor Cells Therapy for Patients With Knee Osteoarthritis
Adipose
China
II
Knee OA
Single group assignment, double blinded
48
6 months
Fresh MSCs (10 million, 20 million, 50 million cells twice) vs. placebo (PBS)
Intra-articular injection
WOMAC score
Completed in December 2013; no publication found
NCT01809769
Clinical Trial of Autologous Adipose Tissue-Derived Mesenchymal Progenitor Cells (MPCs) Therapy for Knee Osteoarthritis
Adipose
China
II
Knee OA
Randomised, parallel assignment, placebo controlled, single blinded
48
12 months
Culture-expanded MSCs vs. sodium hyaluronate
Intra-articular injection
WOMAC
On-going (estimated study completion date; July 2016)
NCT02162693
Outcomes Data of Adipose Stem Cells to Treat Osteoarthritis
Adipose
USA
NA
Knee OA
Observational cohort study
50
12 months
Cellular concentrate
Unknown
KOOS, HOOS
On-going (estimated study completion date; September 2017)
NCT02241408
Clinical Trial to Evaluate Efficacy and Safety of JOINTSTEM in Patients With Degenerative Arthritis
Adipose
Korea
II/III
Knee OA
Randomised parallel assignment, double blinded
120
24 weeks
MSCs (100 million cells) vs. sodium chloride
Injection
WOMAC
On-going (estimated study completion date; July 2017)
NCT02658344
ADIPOA–Clinical Study
Adipose
France
I
Moderate or severe knee OA
Non-randomised parallel assignment, open label
12
1 year
MSCs (2 million, 10 million, 50 million cells)
Intra-articular injection
Safety
Completed in December 2014; no publication found
NCT01585857
Safety and Clinical Effectiveness of A3 SVF in Osteoarthritis
Adipose
USA
Not given
OA
Single group assignment, open label
30
1 year
Stromal vascular fraction with activated platelet
Injection
Pain and inflammation–WOMAC scores, comprehensive inflammation blood panel
On-going
(estimated study completion date; September 2015)
NCT01947348
Safety and Clinical Outcomes Study: SVF Deployment for Orthopaedic, Neurologic, Urologic, and Cardio-pulmonary Conditions
Adipose
USA
Not given
Neurodegenerative diseases, OA, erectile dysfunction, autoimmune diseases, cardiomyopathies or emphysema
Single group assignment, open label
3000
36 months
Stromal vascular fraction
Intra-venous, intra-articular, and soft tissue injection
Safety
On-going
(estimated study completion date; March 2018)
NCT01953523
Microfracture Versus Adipose-Derived Stem Cells for the Treatment of Articular Cartilage Defects
Adipose
USA
Not given
Knee OA
Randomised, parallel assignment, double blind
90
24 months
Fibrin glue + acellular collagen dermal matrix + DSCs, + additional layer of fibrin glue vs. microfracture
Arthroscopy
KOOS
On-going (estimated study completion date; December 2020)
NCT02090140
Autologous Mesenchymal Stem Cells vs. Chondrocytes for the Repair of Chondral Knee Defects (ASCROD)
Adipose
Spain
I/II
Articular cartilage lesion of the femoral condyle
Randomised, parallel assignment, open label
30
18 months
Cultured stem cells vs. cultured autologous chondrocytes
Unknown
Hyaline cartilage production for chondral knee lesions repair
Unknown (estimated study completion date; June 2012)
NCT01399749
A Phase 2 Study to Evaluate the Efficacy and Safety of JointStem in Treatment of Osteoarthritis
Adipose
USA
II
Knee OA
Randomised, parallel assignment, double blinded
45
6 months
Joint stem adipose-derived (MSCs) vs. Synvisc-One (hyaluronic acid)
 
Cartilage volume, cartilage articular surface area, cartilage thickness, subchondral bone surface curvature (MRI)
On-going (estimated study completion date; September 2017)
NCT02674399
Allogenic cells
Treatment of Knee Osteoarthritis With Allogenic Mesenchymal Stem Cells (MSV_allo)
Bone marrow
Spain
I/II
Knee OA grade 2–4 of Kellgren and Lawrence
Randomised, parallel assignment, double blinded
30
1 years
Culture-expanded MSCs (40 million cells) vs. hyaluronic acid
Intra-articular transplantation
Safety
Completed in June 2014; published in August 2015
NCT01586312
(Linked to study NCT01183728)
Clinical Trial of Allogenic Adipose Tissue-Derived Mesenchymal Progenitor Cells Therapy for Knee Osteoarthritis
Adipose
China
I
Degenerative arthritis by radiographic criteria of Kellgren Lawrence
Randomised, parallel assignment, double blind
18
48 weeks
10 million MSCs vs. 20 million MSCs
Intra-articular injection
WOMAC
On-going (estimated study completion date; July 2017)
NCT02641860
Clinical Study of Umbilical Cord Tissue Mesenchymal Stem Cells (UC-MSC) for Treatment of Osteoarthritis
Umbilical Cord
Panama
I/II
Modified Kellgren-Lawrence classification grade 2–4 radiographic OA severity.
Randomised, parallel assignment, open label
40
12 months
Single intra-articular injection of MSCs vs.
IV injections of MSC for 3 days
Intra-articular injection; IV
Safety
On-going (estimated study completion date; March 2017)
NCT02237846
Safety and Feasibility Study of Mesenchymal Trophic Factor (MTF) for Treatment of Osteoarthritis
Umbilical Cord
Panama
I/II
Modified Kellgren-Lawrence classification grade 2–4 radiographic OA severity.
Non-Randomised, single group assignment,
open label
40
12 months
Intra-articular injection of allogeneic MTF from UC-MSC vs. 12 subcutaneous MTF injections, once per week
Intra-articular injection; subcutaneous injection
Safety
On-going (estimated study completion date; June 2017)
NCT02003131
A Study to Assess Safety and Efficacy of Umbilical Cord-derived Mesenchymal Stromal Cells in Knee Osteoarthritis
Umbilical Cord
Chile
I/II
Kellgren-Lawrence classification grade 1–3 radiographic OA severity
Randomised, parallel assignment, double blind
30
12 months
MSCs (single dose of 20 million MSCs or double dose at 6 month interval) vs. hyaluronic acid
Intra-articular injection
Safety
On-going (estimated study completion date; December 2016)
NCT02580695
Human Umbilical Cord Mesenchymal Stem Cell Transplantation in Articular Cartilage Defect
Umbilical Cord
China
I
Kellgren-Lawrence classification grade 2–4 radiographic OA severity
Single group assignment, open label
20
12 months
20 million cells every month for 4 months
Intra-articular injection
Safety
On-going (estimated study completion date; December 2016)
NCT02291926
Evaluation of Safety and Exploratory Efficacy of CARTISTEM®, a Cell Therapy Product for Articular Cartilage Defects
Umbilical cord blood
Korea
I/II
Focal, full-thickness grade 3–4 articular cartilage defects
Single group assignment, open label
12
12 months
CARTISTEM® (cultured UC MSCs mixed with sodium hyaluronate)
Unknown
Safety
On-going (estimated study completion date; May 2017)
NCT01733186
Study to Compare the Efficacy and Safety of Cartistem® and Microfracture in Patients With Knee Articular Cartilage Injury or Defect
Umbilical cord blood
Korea
III
Knee Articular Cartilage Injury or Defect
Randomised, parallel assignment, open label
104
48 weeks
CARTISTEM® (cultured UC MSCs mixed with sodium hyaluronate) vs. Microfracture
Surgery
CRS cartilage repair assessment
Completed in January 2011; no publication found
NCT01041001
Follow-Up Study of CARTISTEM® vs. Microfracture for the Treatment of Knee Articular Cartilage Injury or Defect
Umbilical cord blood
Korea
III
Knee articular cartilage injury or defect
Randomised, parallel assignment, open label
103
60 months
CARTISTEM® (cultured UC MSCs mixed with sodium hyaluronate) vs. microfracture
Unknown
IKDC, VAS pain, WOMAC
On-going (estimated study completion date; May 2015)
NCT01626677
Injections of FloGraft Therapy, Autologous Stem Cells, or Platelet Rich Plasma for the Treatment of Degenerative Joint Pain
Amniotic fluid
USA
NA
Pain associated with one of the following conditions: lumbar facet degeneration, degenerative condition causing upper extremity joint pain or degenerative condition causing lower extremity joint pain
Cohort observational study
300
24 weeks
FloGraftTM (allogenic amniotic fluid-derived allograft) vs. autologous BMMSCs vs. platelet rich plasma
Injection
Pain
On-going (estimated study completion date; June 2016)
NCT01978639
IMPACT: Safety and Feasibility of a Single-stage Procedure for Focal Cartilage Lesions of the Knee
Unspecified
The Netherlands
I/II
Full-thickness articular cartilage lesion on the femoral condyle or trochlea
Single-group assignment, open label
35
18 months
Autologous chondrons (chondrocytes with their pericellular matrix) and allogeneic MSCs in the fibrin glue carrier
Unspecified (single stage surgery)
Safety
On-going (Estimated Study Completion Date: August 2015)
NCT02037204
Allogeneic Mesenchymal Stem Cells in Osteoarthritis
Unspecified
India
II
Kellgren and Lawrence classification grade 2–3 radiographic OA severity
Randomised, double blind, multi-centre
60
2 years
Culture-expanded MSCs in 2 ml plasmalyte + 2 ml, hyaluronan vs. 2 ml, plasmalyte + 2 ml, hyaluronan
Intra-articular
Safety and tolerability
Unknown (estimated study completion date; July 2014
NCT01453738
Allogeneic Mesenchymal Stem Cells for Osteoarthritis
Unspecified
Malaysia
II
Kellgren and Lawrence classification grade 2–3 OA
Randomised, double blind, multi-centre
72
1 year
Culture-expanded MSCs in 2 ml plasmalyte + 2 ml, hyaluronan vs. 2 ml, plasmalyte + 2 ml, hyaluronan
Intra-articular
Safety and tolerability
Unknown (estimated study completion date; February 2013)
NCT01448434
Autologous or allogenic unspecified
Transplantation of Bone Marrow Derived Mesenchymal Stem Cells in Affected Knee Osteoarthritis by Rheumatoid Arthritis
Bone marrow
II/III
Iran
Rheumatoid arthritis
Randomised, parallel assignment, open label
60
6 months
MSCs vs. saline
Intra-articular injection
Pain
Completed in December 2011; no publication found
NCT01873625
Safety and Efficacy Study of MSB-CAR001 in Subjects 6 Weeks Post an Anterior Cruciate Ligament Reconstruction
Unknown
I/II
Australia
Anterior cruciate ligament injury
Randomised, parallel assignment, double blind
24
2 year
MSB-CAR001 (a preparation of MSCs) with hyaluronan vs. hyaluronan alone
Injection
Safety
Unknown
NCT01088191
Defects
The majority of studies (42%) used MSCs to treat knee osteoarthritis [103115]. The rest of the studies looked at knee cartilage defects except for two which studied the ankle talar dome [116, 117]. One study used MSCs to treat knee osteoarthritis (OA), knee OA and ankle OA [112].
Of the knee cartilage defects, the patients were heterogeneous with varying defect sizes and locations, including the patellae [118121], patella-femoral joints [122, 123], femoral condyle [113, 119121, 123132], trochlear [119121] and tibial plateau [121]; and several had multiple defect sites [105, 120, 123, 128].
Previous treatment and associated procedures
The majority of patients who received MSC treatment had undergone previous arthroscopy [103, 104, 118, 119, 122, 124, 130], failed debridement [113, 118, 119, 121123, 125, 127, 131] or bone marrow stimulation [114, 116, 117, 126].
Cell harvest source
Twenty-one studies (68%) used bone marrow-derived MSCs from the anterior or posterior superior iliac spine [103105, 109, 111113, 115118, 120, 122128, 130132]. Five studies (18%) used adipose-derived MSCs [106108, 110, 114], two studies (7%) used synovium-derived MSCs [129, 133] and two studies (7%) used peripheral blood progenitor cells collected by apheresis [119, 121].
Cell stage
Twenty studies (61%) culture-expanded their cells [103105, 107113, 115, 118, 120, 122126, 129, 133], whereas 11 studies (39%) used fresh concentrated stem cells from bone marrow [116, 117, 127, 128, 130132], fat tissues [106, 114] or peripheral blood [119, 121] in a one stage-procedure. In studies using bone marrow concentrate, approximately 60 ml of bone marrow aspirate was harvested and concentrated down to a volume of 2–4 ml before use [116, 117, 127, 130132]. In studies using culture-expanded cells, the majority used cells from early passages, P1–P3 [103, 105, 109, 110, 112, 113, 115, 118, 120, 122125, 129]. One study reported the use of cells at a late passage (P5) [104] ,and five studies did not specify a passage number [107, 108, 111, 126, 133].
Thirteen studies (42%) confirmed the phenotype of cells before clinical application [105, 108110, 112, 115, 119, 120, 122125, 129]. Commonly used surface markers to select MSCs were CD29, CD44, CD73, CD90 and CD105. Also CD14, CD34 and HLA-DR were used to eliminate non-MSCs.
Cell dose and delivery
The number of cells applied (dose) varied from 2–57 million for bone marrow-derived MSCs [103105, 109, 111113, 118, 120, 122125, 129] and from 1.2–100 million for adipose-derived MSCs [107, 108, 110, 114]. For synovial MSCs, 8–77 million cells were used [129, 133], and for peripheral blood progenitor cells, 20 million cells were used [119]. Also, the methods for implantation varied from arthroscopic implantation (35%) [107, 108, 116, 117, 127, 128, 130133], intra-articular injection [103106, 109112, 114, 115, 119, 121, 123] or open surgery (29%) [113, 118, 120, 122126, 129].
In the cell therapy studies, the cells were suspended with a variety of different co-stimulators, including hydroxyapatite (HA) [106, 119, 121, 123], platelet rich plasma (PRP) [106, 114] and platelet lysate [104]. Some studies also administered multiple injections of stem cells [119, 121] and/or further injection of HA [115, 119, 121, 123], PRP [106, 114] or nucleated cells [104] following a stem cell injection.
The most frequently used scaffolds were type I collagen of porcine or bovine origin [113, 118, 122, 124, 126, 129], followed by ascorbic acid sheet [120, 123] and platelet-rich fibrin glue mixture [108, 125].
Rehabilitation
Early continuous passive motion was employed in 14 studies [113, 117122, 124127, 129131]. Six studies did not report details on post-operation rehabilitation [104106, 109, 116, 132]. Three studies aimed for full weight bearing very early by week 4 [107, 108, 122] whereas 11 studies (40%) aimed for full weight bearing by the 6th–8th week [113, 117121, 124, 125, 127, 131, 133]. No study addressed the effect of rehabilitation on the quality of the repair.
Outcomes
Most commonly used outcome measures for treatment efficacy were radiological (77%) [103106, 109112, 115117, 119, 121, 123125, 127134] and arthroscopic assessment (61%) [107, 108, 113, 116122, 124126, 130133]. Most commonly used patient-reported outcomes are International Knee Documentation Committee (IKDC) score (36%), followed by a visual analogue scale (VAS) pain (39%) and Tegner activity scale (29%).
Adverse effects
None of the studies reported any severe adverse effects related to the MSC treatment. Two group reported minor adverse events including mild pain and effusion after the injections, which persisted for no more than 7 days [103, 114].

Conclusions

There is a growing fascination with the role of mesenchymal stem cells in cartilage repair.
As early as the 1950s, Pridie showed fibrocartilaginous repair through subchondral drilling [135137]. Initially, Pridie drilling was reported as a treatment for osteoarthritis [135, 138] and was often associated with many additional procedures such as synovectomy and trimming of osteophytes.
Since Pridie’s initial experiments, the process of marrow stimulation techniques or exposure of mesenchymal stem cells from cancellous bone has changed its guise on several occasions.
Ficat in 1979 described “Spongialization” in which the cancellous bed was exposed in 85 patients with chondral lesions of the patella with encouraging results [139]. Johnson et al. [140] described abrasion arthroplasty and encouraged its use especially in younger patients [141, 142]. Other authors had less positive outcomes [143146]. Dandy wrote an entertaining article on abrasion arthroplasty where he highlighted that at least in the treatment of osteoarthritis, its effects could relate to the arthroscopic washout, rest or even the placebo effects of the charismatic surgeon [147]. The final evolution of marrow stimulation was the term “Microfracture” enabled by commercially manufactured bone picks used to breach the subchondral bone [8]. Marrow-stimulating technique procedures, in particular microfracture, are now considered the first-line treatment for full-thickness cartilage lesions and have demonstrated good to excellent results in 60–80% of patients [148, 149].
Cartilage repair has evolved from marrow stimulation techniques through to chondrocyte transplant and now stem cells at rapid pace. An ideal translational pipeline would demonstrate how in vitro data was used to inform a pre-clinical model, which would later form a phase I/IIa first-in-man study and subsequently a phase III clinical trial. This would of course be the safe and responsible method by which novel therapies are brought to the market.
This systematic review is the first of its kind to explore the full spectrum of evidence from in vitro studies, through animal studies to human clinical trials, and yet, we found little evidence of connectivity between in vitro, animal and then human work. In fact, we did not find a single group that had carried out and reported studies in all three categories.
Indeed, even from groups, which showed a seemingly hierarchical approach to translation, discrepancies became apparent. For example, Saw et al. from Korea used a pre-clinical goat model to repair cartilage defects using HA plus bone marrow-derived cells [150] and then moved into a first-in-man study, but in doing so, elected to change from bone marrow aspirate to peripheral blood and justified this change because it was easier to harvest peripheral blood than marrow [151].
There are several sources of cells that have been used in cartilage repair including bone marrow, peripheral blood, synovium, adipose tissue and umbilicus (Table 14) without any clear evidence of superiority of one over the other.

One stage vs. two stages

As two stage procedures involving cell culture are expensive and cumbersome, there is an increasing push towards a single stage stem cell treatment. In this situation there is some supportive pre-clinical data [91, 95, 98, 152154], but there does not appear to be a pre-clinical study that directly compares bone marrow concentrates against cultured MSCs.
Several groups have reported the use of bone marrow concentrates in clinical practice [116, 117, 127, 128, 130132], in which the buffy coat is used containing the nucleated cells, of which a few will be stem cells.
Briefly, the patient has approximately 60 mL of bone marrow harvested from the iliac crest which is then spun down in a cell centrifuge (SmartPrep, Harvest Technologies Corp., USA, or IOR-G1, Novagenit, Mezzolombardo, TN, Italy) to provide 6 mL of concentrate containing nucleated cells. A small amount of the nucleated cells are then placed onto a hyaluronic acid membrane (Hyalofast, Fidia Advanced Biopolymers, Italy) or collagen membrane (IOR-G1, Novagenit, Mezzolombardo, TN, Italy) as a scaffold, which is then arthroscopically placed into the cartilage defect which had been pre-prepared using a burr or drill. The construct is then held with a platelet gel obtained from a harvest of 120 mL of patient’s venous blood taken the day before surgery (Vivostat system, (Vivolution, Denmark)) [118]. The results of the first 30 patients have been reported as showing improvements in MRI and arthroscopic appearance as well as clinical scores at 3 years follow-up [118].
This new technique is of course an evolution of the autologous matrix-enhanced chondrogenesis (AMIC) which used the stem cells from the adjacent marrow (and not pre-harvested bone marrow concentrates) within either collagen patches [155157] or polyglycolic acid–hyaluronan-based scaffolds [158, 159].
There has also been a further step taken to avoid bone marrow harvest in which peripheral blood has been used in knee chondral lesions. In an RCT, arthroscopic subchondral drilling was followed by postoperative intra-articular injections of hyaluronic acid (HA) with and without peripheral blood stem cells (PBSC). Fifty patients were studied and randomised 1 week after surgery to receive either 8 injections of HA or 8 injections of HA plus PBSC. Those that underwent PBSC received stimulation with filgrastim, which contains recombinant human granulocyte colony-stimulating factor prior to harvest [106, 151]. At 18 month follow-up, they reported no adverse effects and improved MRI findings in the PBSC group compared to HA alone, took biopsies of 16 of the 25 patients in each group and claimed better tissue morphology in the PBSC group, as graded by the International Cartilage Repair Society Visual Assessment Scale II. Interestingly, however, the same group’s pre-clinical used bone marrow aspirates and not peripheral blood [150].

Autologous vs. allogenic

There is an increasing interest in allogenic cells to avoid donor site morbidity and to reduce cost. The pre-clinical data with regards to allogenic cells is conflicting. One group showed promising results of allogenic MSCs in a rabbit model when compared to autologous cells, although numbers were small [160, 161]. Another group compared autologous chondroprogenitor cells and allogenic chondroprogenitor cells against controls in an equine model and reported inferior repair in the allogenic cell group [23]. Despite conflicting pre-clinical data, human studies using allogenic cells began in Korea in 2009. A phase I/IIa study to assess safety and efficacy of a combination of human umbilical cord blood-derived mesenchymal stem cells and sodium hyaluronate (CARTISTEM® (MEDIPOST Co., Ltd., Korea)) was performed in knee chondral defects (NCT01041001). A parallel phase 3, open-label, multi-centre RCT comparing CARTISTEM® and microfracture in knee chondral defects was carried out in Korea and the USA (NCT01733186). Results are still pending.
Another area of huge controversy is the actual dose of cells that should be used. In vitro between 50,000 cells/mL and 100 billion cells/ml have been studied. In pre-clinical animal studies, this ranged from 1000 to 1 billion cells/mL, and in human studies, the reported range has been 1.2 million cells/mL–24 million cells/mL.
It remains unclear what the most appropriate cell dose should be, with some groups reporting that a higher cell number leads to a better repair [52, 71, 87, 95, 162164], but Zhao et al. [99] highlighted the limitation to cell saturation and survival, and thus, there may be a top limit to cell number that can be used to aid repair.
A multitude of methods for cell delivery have also been adopted, from direct joint injection or embedded in a plethora of scaffolds, such as type I collagen gels of porcine or bovine origin, ascorbic acid sheets or fibrin glues (Table 14).
In vitro and in pre-clinical studies, a plethora of growth factors have been studied including TGF-β1 and TGF-β2 and BMP-7 but none of these have been included in human clinical trials (Table 5).
It is clear that the relationship between cell passage, cell dose, the use of scaffolds and growth factors and the efficacy of MSC treatment is still to be established.

Future

There is no question that the field of cartilage repair accelerates at rapid pace, and it is clear that the single stage procedures are likely to win over two stage procedures to save costs and reduce the burden on both provider and the patient. The reduction of donor site morbidity is a further driver helping direct progress.
The concept of cell banks of allogenic cells clearly meets all of the above criteria, but the lack of good supporting pre-clinical and long-term safety and efficacy data does little to pacify potential pitfalls of this direction. The fact that the phase 3 RCT of allogenic umbilical stem cells was allowed to be registered (NCT01041001) before the same group registered their phase I/IIa safety study (NCT01733186) intimates that sometimes clinical pace exceeds that of the regulators to lay down new ground.
Tools are likely to be introduced to the operating theatre that might improve the efficacy of treatment, such as fluorescence-activated cell sorting (FACS) machines which can isolate MSCs from the buffy coat of bone marrow aspirate by their cell surface markers. At present, this technology is expensive and complicated and ways to reduce cost and make the process simple are required before they could enter the operating theatre.
Induced pluripotent stem cells (iPSCs) are adult somatic cells that have been genetically reprogrammed to an embryonic stem cell-like state by being forced to express genes and factors important for maintaining the defining properties of embryonic stem cells [165].
These cells show unlimited self-renewal, and some in vitro studies have shown chondrogenic differentiation by iPSCs from human chondrocytes biopsied from osteoarthritic knees [166] and cartilage formation from human neural stem cells [167]. However, this work is at a very early stage, and aside from the ethical considerations, much research into control of cell phenotype and cell fate to alleviate concerns for cancer risk are required before this technology is ready to move into the pre-clinical and clinical realms.
In conclusion, this review is a comprehensive assessment of the evidence base to date behind the translation of basic science to the clinical practice of cartilage repair. We have revealed a lack of connectivity between the in vitro, pre-clinical and human data and a patchwork quilt of synergistic evidence. It appears that the drivers for progress in this space are largely driven by patient demand, surgeon inquisition, and a regulatory framework that is learning at the same pace as new developments take place. We strongly recommend funding body commission studies that have a clear translational purpose in order to drive the science towards patient benefit.

Acknowledgements

None

Funding

There was no external funding for this work.

Availability of data and materials

Not applicable

Authors’ contributions

All authors were involved in the conception and design of the study or acquisition of the data or analysis and interpretation of the data and contributed to drafting the article or revising it critically for important intellectual content. All authors read and approved the final manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable
Not applicable
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
8.
Zurück zum Zitat Steadman JR, Briggs KK, Rodrigo JJ, Kocher MS, Gill TJ, Rodkey WG. Outcomes of microfracture for traumatic chondral defects of the knee: average 11-year follow-up. Arthroscopy. 19(5):477-84. doi:10.1053/jars.2003.50112. Steadman JR, Briggs KK, Rodrigo JJ, Kocher MS, Gill TJ, Rodkey WG. Outcomes of microfracture for traumatic chondral defects of the knee: average 11-year follow-up. Arthroscopy. 19(5):477-84. doi:10.​1053/​jars.​2003.​50112.
9.
Zurück zum Zitat Peterson L, Menche D, Grande D PM. Chondrocyte transplantation: an experimental model in the rabbit. Trans Orthop Res Soc. 1984;9:218. Peterson L, Menche D, Grande D PM. Chondrocyte transplantation: an experimental model in the rabbit. Trans Orthop Res Soc. 1984;9:218.
13.
Zurück zum Zitat Brittberg M. Cell carriers as the next generation of cell therapy for cartilage repair: a review of the matrix-induced autologous chondrocyte implantation procedure. Am J Sports Med. 2010;38(6):1259–71. doi:10.1177/0363546509346395.PubMedCrossRef Brittberg M. Cell carriers as the next generation of cell therapy for cartilage repair: a review of the matrix-induced autologous chondrocyte implantation procedure. Am J Sports Med. 2010;38(6):1259–71. doi:10.​1177/​0363546509346395​.PubMedCrossRef
15.
Zurück zum Zitat Bentley G, Biant LC, Vijayan S, Macmull S, Skinner JA, Carrington RWJ. Minimum ten-year results of a prospective randomised study of autologous chondrocyte implantation versus mosaicplasty for symptomatic articular cartilage lesions of the knee. J Bone Joint Surg Br. 2012;94(4):504–9. doi:10.1302/0301-620X.94B4.27495.PubMedCrossRef Bentley G, Biant LC, Vijayan S, Macmull S, Skinner JA, Carrington RWJ. Minimum ten-year results of a prospective randomised study of autologous chondrocyte implantation versus mosaicplasty for symptomatic articular cartilage lesions of the knee. J Bone Joint Surg Br. 2012;94(4):504–9. doi:10.​1302/​0301-620X.​94B4.​27495.PubMedCrossRef
19.
Zurück zum Zitat Gossec L, Paternotte S, Maillefert JF, et al. The role of pain and functional impairment in the decision to recommend total joint replacement in hip and knee osteoarthritis: an international cross-sectional study of 1909 patients. Report of the OARSI-OMERACT Task Force on total joint replacement. Osteoarthritis Cartilage. 2011;19(2):147–54. doi:10.1016/j.joca.2010.10.025.PubMedCrossRef Gossec L, Paternotte S, Maillefert JF, et al. The role of pain and functional impairment in the decision to recommend total joint replacement in hip and knee osteoarthritis: an international cross-sectional study of 1909 patients. Report of the OARSI-OMERACT Task Force on total joint replacement. Osteoarthritis Cartilage. 2011;19(2):147–54. doi:10.​1016/​j.​joca.​2010.​10.​025.PubMedCrossRef
21.
Zurück zum Zitat Mithoefer K, Williams RJ, Warren RF, Wickiewicz TL, Marx RG. High-impact athletics after knee articular cartilage repair: a prospective evaluation of the microfracture technique. Am J Sports Med. 2006;34(9):1413–8. doi:10.1177/0363546506288240.PubMedCrossRef Mithoefer K, Williams RJ, Warren RF, Wickiewicz TL, Marx RG. High-impact athletics after knee articular cartilage repair: a prospective evaluation of the microfracture technique. Am J Sports Med. 2006;34(9):1413–8. doi:10.​1177/​0363546506288240​.PubMedCrossRef
22.
23.
Zurück zum Zitat Frisbie DD, Trotter GW, Powers BE, et al. Arthroscopic subchondral bone plate microfracture technique augments healing of large chondral defects in the radial carpal bone and medial femoral condyle of horses. Vet Surg. 28(4):242-55. Available at: http://www.ncbi.nlm.nih.gov/pubmed/10424704. Accessed 12 June 2016. Frisbie DD, Trotter GW, Powers BE, et al. Arthroscopic subchondral bone plate microfracture technique augments healing of large chondral defects in the radial carpal bone and medial femoral condyle of horses. Vet Surg. 28(4):242-55. Available at: http://​www.​ncbi.​nlm.​nih.​gov/​pubmed/​10424704. Accessed 12 June 2016.
26.
Zurück zum Zitat Minas T, Gomoll AH, Rosenberger R, Royce RO, Bryant T. Increased failure rate of autologous chondrocyte implantation after previous treatment with marrow stimulation techniques. Am J Sports Med. 2009;37(5):902–8. doi:10.1177/0363546508330137.PubMedCrossRef Minas T, Gomoll AH, Rosenberger R, Royce RO, Bryant T. Increased failure rate of autologous chondrocyte implantation after previous treatment with marrow stimulation techniques. Am J Sports Med. 2009;37(5):902–8. doi:10.​1177/​0363546508330137​.PubMedCrossRef
30.
Zurück zum Zitat Wood JJ, Malek MA, Frassica FJ, et al. Autologous cultured chondrocytes: adverse events reported to the United States Food and Drug Administration. J Bone Joint Surg Am. 2006;88(3):503–7. doi:10.2106/JBJS.E.00103.PubMed Wood JJ, Malek MA, Frassica FJ, et al. Autologous cultured chondrocytes: adverse events reported to the United States Food and Drug Administration. J Bone Joint Surg Am. 2006;88(3):503–7. doi:10.​2106/​JBJS.​E.​00103.PubMed
33.
Zurück zum Zitat Takata N, Furumatsu T, Abe N, Naruse K, Ozaki T. Comparison between loose fragment chondrocytes and condyle fibrochondrocytes in cellular proliferation and redifferentiation. J Orthop Sci. 2011;16(5):589–97. doi:10.1007/s00776-011-0128-1.PubMedCrossRef Takata N, Furumatsu T, Abe N, Naruse K, Ozaki T. Comparison between loose fragment chondrocytes and condyle fibrochondrocytes in cellular proliferation and redifferentiation. J Orthop Sci. 2011;16(5):589–97. doi:10.​1007/​s00776-011-0128-1.PubMedCrossRef
36.
Zurück zum Zitat Bartlett W, Skinner JA, Gooding CR, et al. Autologous chondrocyte implantation versus matrix-induced autologous chondrocyte implantation for osteochondral defects of the knee: a prospective, randomised study. J Bone Joint Surg Br. 2005;87(5):640–5. doi:10.1302/0301-620X.87B5.15905.PubMedCrossRef Bartlett W, Skinner JA, Gooding CR, et al. Autologous chondrocyte implantation versus matrix-induced autologous chondrocyte implantation for osteochondral defects of the knee: a prospective, randomised study. J Bone Joint Surg Br. 2005;87(5):640–5. doi:10.​1302/​0301-620X.​87B5.​15905.PubMedCrossRef
39.
Zurück zum Zitat Kon E, Verdonk P, Condello V, et al. Matrix-assisted autologous chondrocyte transplantation for the repair of cartilage defects of the knee: systematic clinical data review and study quality analysis. Am J Sports Med. 2009;37 Suppl 1:156S–66S. doi:10.1177/0363546509351649.PubMedCrossRef Kon E, Verdonk P, Condello V, et al. Matrix-assisted autologous chondrocyte transplantation for the repair of cartilage defects of the knee: systematic clinical data review and study quality analysis. Am J Sports Med. 2009;37 Suppl 1:156S–66S. doi:10.​1177/​0363546509351649​.PubMedCrossRef
48.
Zurück zum Zitat Zhang B, Yang S, Sun Z, et al. Human mesenchymal stem cells induced by growth differentiation factor 5: an improved self-assembly tissue engineering method for cartilage repair. Tissue Eng Part C Methods. 2011;17(12):1189–99. doi:10.1089/ten.tec.2011.0011.PubMedCrossRef Zhang B, Yang S, Sun Z, et al. Human mesenchymal stem cells induced by growth differentiation factor 5: an improved self-assembly tissue engineering method for cartilage repair. Tissue Eng Part C Methods. 2011;17(12):1189–99. doi:10.​1089/​ten.​tec.​2011.​0011.PubMedCrossRef
49.
Zurück zum Zitat Nam HY, Karunanithi P, Loo WC, et al. The effects of staged intra-articular injection of cultured autologous mesenchymal stromal cells on the repair of damaged cartilage: a pilot study in caprine model. Arthritis Res Ther. 2013;15(5):R129. doi:10.1186/ar4309.PubMedPubMedCentralCrossRef Nam HY, Karunanithi P, Loo WC, et al. The effects of staged intra-articular injection of cultured autologous mesenchymal stromal cells on the repair of damaged cartilage: a pilot study in caprine model. Arthritis Res Ther. 2013;15(5):R129. doi:10.​1186/​ar4309.PubMedPubMedCentralCrossRef
51.
53.
Zurück zum Zitat Huang C-YC, Reuben PM, D’Ippolito G, Schiller PC, Cheung HS. Chondrogenesis of human bone marrow-derived mesenchymal stem cells in agarose culture. Anat Rec A Discov Mol Cell Evol Biol. 2004;278(1):428–36. doi:10.1002/ar.a.20010.PubMedCrossRef Huang C-YC, Reuben PM, D’Ippolito G, Schiller PC, Cheung HS. Chondrogenesis of human bone marrow-derived mesenchymal stem cells in agarose culture. Anat Rec A Discov Mol Cell Evol Biol. 2004;278(1):428–36. doi:10.​1002/​ar.​a.​20010.PubMedCrossRef
54.
Zurück zum Zitat Wang ZJ, An RZ, Zhao JY, et al. Repair of articular cartilage defects by tissue-engineered cartilage constructed with adipose-derived stem cells and acellular cartilaginous matrix in rabbits. Genet Mol Res. 2014;13(2):4599–606. doi:10.4238/2014.June.18.2.PubMedCrossRef Wang ZJ, An RZ, Zhao JY, et al. Repair of articular cartilage defects by tissue-engineered cartilage constructed with adipose-derived stem cells and acellular cartilaginous matrix in rabbits. Genet Mol Res. 2014;13(2):4599–606. doi:10.​4238/​2014.​June.​18.​2.PubMedCrossRef
55.
Zurück zum Zitat Raheja LF, Galuppo LD, Bowers-Lepore J, Dowd JP, Tablin F, Yellowley CE. Treatment of bilateral medial femoral condyle articular cartilage fissures in a horse using bone marrow-derived multipotent mesenchymal stromal cells. J Equine Vet Sci. 2011;31(3):147–54. doi:10.1016/j.jevs.2010.12.009.CrossRef Raheja LF, Galuppo LD, Bowers-Lepore J, Dowd JP, Tablin F, Yellowley CE. Treatment of bilateral medial femoral condyle articular cartilage fissures in a horse using bone marrow-derived multipotent mesenchymal stromal cells. J Equine Vet Sci. 2011;31(3):147–54. doi:10.​1016/​j.​jevs.​2010.​12.​009.CrossRef
58.
Zurück zum Zitat Masuoka K, Asazuma T, Hattori H, et al. Tissue engineering of articular cartilage with autologous cultured adipose tissue-derived stromal cells using atelocollagen honeycomb-shaped scaffold with a membrane sealing in rabbits. J Biomed Mater Res B Appl Biomater. 2006;79(1):25–34. doi:10.1002/jbm.b.30507.PubMedCrossRef Masuoka K, Asazuma T, Hattori H, et al. Tissue engineering of articular cartilage with autologous cultured adipose tissue-derived stromal cells using atelocollagen honeycomb-shaped scaffold with a membrane sealing in rabbits. J Biomed Mater Res B Appl Biomater. 2006;79(1):25–34. doi:10.​1002/​jbm.​b.​30507.PubMedCrossRef
59.
60.
Zurück zum Zitat Zhu S, Chen P, Wu Y, et al. Programmed application of transforming growth factor β3 and Rac1 inhibitor NSC23766 committed hyaline cartilage differentiation of adipose-derived stem cells for osteochondral defect repair. Stem Cells Transl Med. 2014;3(10):1242–51. doi:10.5966/sctm.2014-0042.PubMedPubMedCentralCrossRef Zhu S, Chen P, Wu Y, et al. Programmed application of transforming growth factor β3 and Rac1 inhibitor NSC23766 committed hyaline cartilage differentiation of adipose-derived stem cells for osteochondral defect repair. Stem Cells Transl Med. 2014;3(10):1242–51. doi:10.​5966/​sctm.​2014-0042.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Lu C-H, Yeh T-S, Yeh C-L, et al. Regenerating cartilages by Engineered ASCs: prolonged TGF-β3/BMP-6 expression improved articular cartilage formation and restored zonal structure. Mol Ther. 2014;22(1):186–95. doi:10.1038/mt.2013.165.CrossRef Lu C-H, Yeh T-S, Yeh C-L, et al. Regenerating cartilages by Engineered ASCs: prolonged TGF-β3/BMP-6 expression improved articular cartilage formation and restored zonal structure. Mol Ther. 2014;22(1):186–95. doi:10.​1038/​mt.​2013.​165.CrossRef
66.
Zurück zum Zitat Im G-I, Lee JH. Repair of osteochondral defects with adipose stem cells and a dual growth factor-releasing scaffold in rabbits. J Biomed Mater Res B Appl Biomater. 2010;92(2):552–60. doi:10.1002/jbm.b.31552.PubMed Im G-I, Lee JH. Repair of osteochondral defects with adipose stem cells and a dual growth factor-releasing scaffold in rabbits. J Biomed Mater Res B Appl Biomater. 2010;92(2):552–60. doi:10.​1002/​jbm.​b.​31552.PubMed
67.
Zurück zum Zitat Kang H, Peng J, Lu S, et al. In vivo cartilage repair using adipose-derived stem cell-loaded decellularized cartilage ECM scaffolds. J Tissue Eng Regen Med. 2014;8(6):442–53. doi:10.1002/term.1538.PubMedCrossRef Kang H, Peng J, Lu S, et al. In vivo cartilage repair using adipose-derived stem cell-loaded decellularized cartilage ECM scaffolds. J Tissue Eng Regen Med. 2014;8(6):442–53. doi:10.​1002/​term.​1538.PubMedCrossRef
68.
Zurück zum Zitat Gong L, Zhou X, Wu Y, et al. Proteomic analysis profile of engineered articular cartilage with chondrogenic differentiated adipose tissue-derived stem cells loaded polyglycolic acid mesh for weight-bearing area defect repair. Tissue Eng Part A. 2014;20(3-4):575–87. doi:10.1089/ten.TEA.2013.0205.PubMed Gong L, Zhou X, Wu Y, et al. Proteomic analysis profile of engineered articular cartilage with chondrogenic differentiated adipose tissue-derived stem cells loaded polyglycolic acid mesh for weight-bearing area defect repair. Tissue Eng Part A. 2014;20(3-4):575–87. doi:10.​1089/​ten.​TEA.​2013.​0205.PubMed
69.
Zurück zum Zitat de Girolamo L, Niada S, Arrigoni E, et al. Repair of osteochondral defects in the minipig model by OPF hydrogel loaded with adipose-derived mesenchymal stem cells. Regen Med. 2015;10(2):135–51. doi:10.2217/rme.14.77.PubMedCrossRef de Girolamo L, Niada S, Arrigoni E, et al. Repair of osteochondral defects in the minipig model by OPF hydrogel loaded with adipose-derived mesenchymal stem cells. Regen Med. 2015;10(2):135–51. doi:10.​2217/​rme.​14.​77.PubMedCrossRef
70.
Zurück zum Zitat Pei M, He F, Li J, Tidwell JE, Jones AC, McDonough EB. Repair of large animal partial-thickness cartilage defects through intraarticular injection of matrix-rejuvenated synovium-derived stem cells. Tissue Eng Part A. 2013;19(9-10):1144–54. doi:10.1089/ten.TEA.2012.0351.PubMedCrossRef Pei M, He F, Li J, Tidwell JE, Jones AC, McDonough EB. Repair of large animal partial-thickness cartilage defects through intraarticular injection of matrix-rejuvenated synovium-derived stem cells. Tissue Eng Part A. 2013;19(9-10):1144–54. doi:10.​1089/​ten.​TEA.​2012.​0351.PubMedCrossRef
71.
72.
74.
Zurück zum Zitat Lee J-C, Min HJ, Lee S, Seong SC, Lee MC. Effect of chondroitinase ABC on adhesion and behavior of synovial membrane-derived mesenchymal stem cells in rabbit partial-thickness chondral defects. J Orthop Res. 2013;31(8):1293–301. doi:10.1002/jor.22353.PubMedCrossRef Lee J-C, Min HJ, Lee S, Seong SC, Lee MC. Effect of chondroitinase ABC on adhesion and behavior of synovial membrane-derived mesenchymal stem cells in rabbit partial-thickness chondral defects. J Orthop Res. 2013;31(8):1293–301. doi:10.​1002/​jor.​22353.PubMedCrossRef
77.
Zurück zum Zitat Hui JHP, Chen F, Thambyah A, Lee EH. Treatment of chondral lesions in advanced osteochondritis dissecans: a comparative study of the efficacy of chondrocytes, mesenchymal stem cells, periosteal graft, and mosaicplasty (osteochondral autograft) in animal models. J Pediatr Orthop. 24(4):427-33. Available at: http://www.ncbi.nlm.nih.gov/pubmed/15205626. Accessed 12 June 2016. Hui JHP, Chen F, Thambyah A, Lee EH. Treatment of chondral lesions in advanced osteochondritis dissecans: a comparative study of the efficacy of chondrocytes, mesenchymal stem cells, periosteal graft, and mosaicplasty (osteochondral autograft) in animal models. J Pediatr Orthop. 24(4):427-33. Available at: http://​www.​ncbi.​nlm.​nih.​gov/​pubmed/​15205626. Accessed 12 June 2016.
78.
Zurück zum Zitat Gelse K, von der Mark K, Aigner T, Park J, Schneider H. Articular cartilage repair by gene therapy using growth factor-producing mesenchymal cells. Arthritis Rheum. 2003;48(2):430–41. doi:10.1002/art.10759.PubMedCrossRef Gelse K, von der Mark K, Aigner T, Park J, Schneider H. Articular cartilage repair by gene therapy using growth factor-producing mesenchymal cells. Arthritis Rheum. 2003;48(2):430–41. doi:10.​1002/​art.​10759.PubMedCrossRef
79.
Zurück zum Zitat Zhang S, Jiang YZ, Zhang W, et al. Neonatal desensitization supports long-term survival and functional integration of human embryonic stem cell-derived mesenchymal stem cells in rat joint cartilage without immunosuppression. Stem Cells Dev. 2013;22(1):90–101. doi:10.1089/scd.2012.0116.PubMedCrossRef Zhang S, Jiang YZ, Zhang W, et al. Neonatal desensitization supports long-term survival and functional integration of human embryonic stem cell-derived mesenchymal stem cells in rat joint cartilage without immunosuppression. Stem Cells Dev. 2013;22(1):90–101. doi:10.​1089/​scd.​2012.​0116.PubMedCrossRef
81.
Zurück zum Zitat Chung J, Song M, Ha C-W, Kim J-A, Lee C-H, Park Y-B. Comparison of articular cartilage repair with different hydrogel-human umbilical cord blood-derived mesenchymal stem cell composites in a rat model. Stem Cell Res Ther. 2014;5(2):39. doi:10.1186/scrt427.PubMedPubMedCentralCrossRef Chung J, Song M, Ha C-W, Kim J-A, Lee C-H, Park Y-B. Comparison of articular cartilage repair with different hydrogel-human umbilical cord blood-derived mesenchymal stem cell composites in a rat model. Stem Cell Res Ther. 2014;5(2):39. doi:10.​1186/​scrt427.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Nawata M, Wakitani S, Nakaya H, et al. Use of bone morphogenetic protein 2 and diffusion chambers to engineer cartilage tissue for the repair of defects in articular cartilage. Arthritis Rheum. 2005;52(1):155–63. doi:10.1002/art.20713.PubMedCrossRef Nawata M, Wakitani S, Nakaya H, et al. Use of bone morphogenetic protein 2 and diffusion chambers to engineer cartilage tissue for the repair of defects in articular cartilage. Arthritis Rheum. 2005;52(1):155–63. doi:10.​1002/​art.​20713.PubMedCrossRef
84.
Zurück zum Zitat Tay LX, Ahmad RE, Dashtdar H, et al. Treatment outcomes of alginate-embedded allogenic mesenchymal stem cells versus autologous chondrocytes for the repair of focal articular cartilage defects in a rabbit model. Am J Sports Med. 2012;40(1):83–90. doi:10.1177/0363546511420819.PubMedCrossRef Tay LX, Ahmad RE, Dashtdar H, et al. Treatment outcomes of alginate-embedded allogenic mesenchymal stem cells versus autologous chondrocytes for the repair of focal articular cartilage defects in a rabbit model. Am J Sports Med. 2012;40(1):83–90. doi:10.​1177/​0363546511420819​.PubMedCrossRef
85.
Zurück zum Zitat Dashtdar H, Rothan HA, Tay T, et al. A preliminary study comparing the use of allogenic chondrogenic pre-differentiated and undifferentiated mesenchymal stem cells for the repair of full thickness articular cartilage defects in rabbits. J Orthop Res. 2011;29(9):1336–42. doi:10.1002/jor.21413.PubMedCrossRef Dashtdar H, Rothan HA, Tay T, et al. A preliminary study comparing the use of allogenic chondrogenic pre-differentiated and undifferentiated mesenchymal stem cells for the repair of full thickness articular cartilage defects in rabbits. J Orthop Res. 2011;29(9):1336–42. doi:10.​1002/​jor.​21413.PubMedCrossRef
86.
Zurück zum Zitat Igarashi T, Iwasaki N, Kawamura D, et al. Repair of articular cartilage defects with a novel injectable in situ forming material in a canine model. J Biomed Mater Res A. 2012;100(1):180–7. doi:10.1002/jbm.a.33248.PubMedCrossRef Igarashi T, Iwasaki N, Kawamura D, et al. Repair of articular cartilage defects with a novel injectable in situ forming material in a canine model. J Biomed Mater Res A. 2012;100(1):180–7. doi:10.​1002/​jbm.​a.​33248.PubMedCrossRef
87.
89.
Zurück zum Zitat Al Faqeh H, Nor Hamdan BMY, Chen HC, Aminuddin BS, Ruszymah BHI. The potential of intra-articular injection of chondrogenic-induced bone marrow stem cells to retard the progression of osteoarthritis in a sheep model. Exp Gerontol. 2012;47(6):458–64. doi:10.1016/j.exger.2012.03.018.PubMedCrossRef Al Faqeh H, Nor Hamdan BMY, Chen HC, Aminuddin BS, Ruszymah BHI. The potential of intra-articular injection of chondrogenic-induced bone marrow stem cells to retard the progression of osteoarthritis in a sheep model. Exp Gerontol. 2012;47(6):458–64. doi:10.​1016/​j.​exger.​2012.​03.​018.PubMedCrossRef
91.
Zurück zum Zitat Nishimori M, Deie M, Kanaya A, Exham H, Adachi N, Ochi M. Repair of chronic osteochondral defects in the rat. A bone marrow-stimulating procedure enhanced by cultured allogenic bone marrow mesenchymal stromal cells. J Bone Joint Surg Br. 2006;88(9):1236–44. doi:10.1302/0301-620X.88B9.17810.PubMedCrossRef Nishimori M, Deie M, Kanaya A, Exham H, Adachi N, Ochi M. Repair of chronic osteochondral defects in the rat. A bone marrow-stimulating procedure enhanced by cultured allogenic bone marrow mesenchymal stromal cells. J Bone Joint Surg Br. 2006;88(9):1236–44. doi:10.​1302/​0301-620X.​88B9.​17810.PubMedCrossRef
92.
Zurück zum Zitat Espinosa M, Vaisman A, Nazal N, Figueroa D, Gallegos M, Conget P. Intraarticular administration of dexamethasone after mesenchymal stem cells implantation does not improve significantly the treatment of preestablished full-thickness chondral defect in a rabbit model. Cartilage. 2013;4(2):144–52. doi:10.1177/1947603512472696.PubMedPubMedCentralCrossRef Espinosa M, Vaisman A, Nazal N, Figueroa D, Gallegos M, Conget P. Intraarticular administration of dexamethasone after mesenchymal stem cells implantation does not improve significantly the treatment of preestablished full-thickness chondral defect in a rabbit model. Cartilage. 2013;4(2):144–52. doi:10.​1177/​1947603512472696​.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Deng T, Lv J, Pang J, Liu B, Ke J. Construction of tissue-engineered osteochondral composites and repair of large joint defects in rabbit. J Tissue Eng Regen Med. 2014;8(7):546–56. doi:10.1002/term.1556.PubMed Deng T, Lv J, Pang J, Liu B, Ke J. Construction of tissue-engineered osteochondral composites and repair of large joint defects in rabbit. J Tissue Eng Regen Med. 2014;8(7):546–56. doi:10.​1002/​term.​1556.PubMed
94.
Zurück zum Zitat Wan W, Li Q, Gao H, et al. BMSCs laden injectable amino-diethoxypropane modified alginate-chitosan hydrogel for hyaline cartilage reconstruction. J Mater Chem B. 2015;3(9):1990–2005. doi:10.1039/C4TB01394H.CrossRef Wan W, Li Q, Gao H, et al. BMSCs laden injectable amino-diethoxypropane modified alginate-chitosan hydrogel for hyaline cartilage reconstruction. J Mater Chem B. 2015;3(9):1990–2005. doi:10.​1039/​C4TB01394H.CrossRef
97.
Zurück zum Zitat Mokbel A, El-Tookhy O, Shamaa AA, Sabry D, Rashed L, Mostafa A. Homing and efficacy of intra-articular injection of autologous mesenchymal stem cells in experimental chondral defects in dogs. Clin Exp Rheumatol. 29(2):275-84. Available at: http://www.ncbi.nlm.nih.gov/pubmed/21385540. Accessed 12 June 2016. Mokbel A, El-Tookhy O, Shamaa AA, Sabry D, Rashed L, Mostafa A. Homing and efficacy of intra-articular injection of autologous mesenchymal stem cells in experimental chondral defects in dogs. Clin Exp Rheumatol. 29(2):275-84. Available at: http://​www.​ncbi.​nlm.​nih.​gov/​pubmed/​21385540. Accessed 12 June 2016.
98.
Zurück zum Zitat Fortier LA, Potter HG, Rickey EJ, et al. Concentrated bone marrow aspirate improves full-thickness cartilage repair compared with microfracture in the equine model. J Bone Joint Surg Am. 2010;92(10):1927–37. doi:10.2106/JBJS.I.01284.PubMedCrossRef Fortier LA, Potter HG, Rickey EJ, et al. Concentrated bone marrow aspirate improves full-thickness cartilage repair compared with microfracture in the equine model. J Bone Joint Surg Am. 2010;92(10):1927–37. doi:10.​2106/​JBJS.​I.​01284.PubMedCrossRef
99.
Zurück zum Zitat Zhao Q, Wang S, Tian J, et al. Combination of bone marrow concentrate and PGA scaffolds enhance bone marrow stimulation in rabbit articular cartilage repair. J Mater Sci Mater Med. 2013;24(3):793–801. doi:10.1007/s10856-012-4841-x.PubMedCrossRef Zhao Q, Wang S, Tian J, et al. Combination of bone marrow concentrate and PGA scaffolds enhance bone marrow stimulation in rabbit articular cartilage repair. J Mater Sci Mater Med. 2013;24(3):793–801. doi:10.​1007/​s10856-012-4841-x.PubMedCrossRef
100.
Zurück zum Zitat Sun J, Hou X-K, Li X, et al. Mosaicplasty associated with gene enhanced tissue engineering for the treatment of acute osteochondral defects in a goat model. Arch Orthop Trauma Surg. 2009;129(6):757–71. doi:10.1007/s00402-008-0761-0.PubMedCrossRef Sun J, Hou X-K, Li X, et al. Mosaicplasty associated with gene enhanced tissue engineering for the treatment of acute osteochondral defects in a goat model. Arch Orthop Trauma Surg. 2009;129(6):757–71. doi:10.​1007/​s00402-008-0761-0.PubMedCrossRef
104.
Zurück zum Zitat Centeno CJ, Busse D, Kisiday J, Keohan C, Freeman M, Karli D. Increased knee cartilage volume in degenerative joint disease using percutaneously implanted, autologous mesenchymal stem cells. Pain Physician. 11(3):343-53. Available at: http://www.ncbi.nlm.nih.gov/pubmed/18523506. Accessed 13 June 2016. Centeno CJ, Busse D, Kisiday J, Keohan C, Freeman M, Karli D. Increased knee cartilage volume in degenerative joint disease using percutaneously implanted, autologous mesenchymal stem cells. Pain Physician. 11(3):343-53. Available at: http://​www.​ncbi.​nlm.​nih.​gov/​pubmed/​18523506. Accessed 13 June 2016.
107.
109.
Zurück zum Zitat Emadedin M, Aghdami N, Taghiyar L, et al. Intra-articular injection of autologous mesenchymal stem cells in six patients with knee osteoarthritis. Arch Iran Med. 2012;15(7):422-8. doi:012157/AIM.0010. Emadedin M, Aghdami N, Taghiyar L, et al. Intra-articular injection of autologous mesenchymal stem cells in six patients with knee osteoarthritis. Arch Iran Med. 2012;15(7):422-8. doi:012157/AIM.0010.
110.
Zurück zum Zitat Jo CH, Lee YG, Shin WH, et al. Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof-of-concept clinical trial. Stem Cells. 2014;32(5):1254–66. doi:10.1002/stem.1634.PubMedCrossRef Jo CH, Lee YG, Shin WH, et al. Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: a proof-of-concept clinical trial. Stem Cells. 2014;32(5):1254–66. doi:10.​1002/​stem.​1634.PubMedCrossRef
112.
Zurück zum Zitat Emadedin M, Ghorbani Liastani M, Fazeli R, et al. Long-term follow-up of intra-articular injection of autologous mesenchymal stem cells in patients with knee, ankle, or hip osteoarthritis. Arch Iran Med. 2015;18(6):336-44. doi:015186/AIM.003. Emadedin M, Ghorbani Liastani M, Fazeli R, et al. Long-term follow-up of intra-articular injection of autologous mesenchymal stem cells in patients with knee, ankle, or hip osteoarthritis. Arch Iran Med. 2015;18(6):336-44. doi:015186/AIM.003.
113.
Zurück zum Zitat Wakitani S, Imoto K, Yamamoto T, Saito M, Murata N, Yoneda M. Human autologous culture expanded bone marrow mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees. Osteoarthritis Cartilage. 2002;10(3):199–206. doi:10.1053/joca.2001.0504.PubMedCrossRef Wakitani S, Imoto K, Yamamoto T, Saito M, Murata N, Yoneda M. Human autologous culture expanded bone marrow mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees. Osteoarthritis Cartilage. 2002;10(3):199–206. doi:10.​1053/​joca.​2001.​0504.PubMedCrossRef
115.
Zurück zum Zitat Wong KL, Lee KBL, Tai BC, Law P, Lee EH, Hui JHP. Injectable cultured bone marrow-derived mesenchymal stem cells in varus knees with cartilage defects undergoing high tibial osteotomy: a prospective, randomized controlled clinical trial with 2 years’ follow-up. Arthroscopy. 2013;29(12):2020–8. doi:10.1016/j.arthro.2013.09.074.PubMedCrossRef Wong KL, Lee KBL, Tai BC, Law P, Lee EH, Hui JHP. Injectable cultured bone marrow-derived mesenchymal stem cells in varus knees with cartilage defects undergoing high tibial osteotomy: a prospective, randomized controlled clinical trial with 2 years’ follow-up. Arthroscopy. 2013;29(12):2020–8. doi:10.​1016/​j.​arthro.​2013.​09.​074.PubMedCrossRef
117.
Zurück zum Zitat Giannini S, Buda R, Cavallo M, et al. Cartilage repair evolution in post-traumatic osteochondral lesions of the talus: from open field autologous chondrocyte to bone-marrow-derived cells transplantation. Injury. 2010;41(11):1196–203. doi:10.1016/j.injury.2010.09.028.PubMedCrossRef Giannini S, Buda R, Cavallo M, et al. Cartilage repair evolution in post-traumatic osteochondral lesions of the talus: from open field autologous chondrocyte to bone-marrow-derived cells transplantation. Injury. 2010;41(11):1196–203. doi:10.​1016/​j.​injury.​2010.​09.​028.PubMedCrossRef
118.
119.
Zurück zum Zitat Saw K-Y, Anz A, Merican S, et al. Articular cartilage regeneration with autologous peripheral blood progenitor cells and hyaluronic acid after arthroscopic subchondral drilling: a report of 5 cases with histology. Arthroscopy. 2011;27(4):493–506. doi:10.1016/j.arthro.2010.11.054.PubMedCrossRef Saw K-Y, Anz A, Merican S, et al. Articular cartilage regeneration with autologous peripheral blood progenitor cells and hyaluronic acid after arthroscopic subchondral drilling: a report of 5 cases with histology. Arthroscopy. 2011;27(4):493–506. doi:10.​1016/​j.​arthro.​2010.​11.​054.PubMedCrossRef
120.
Zurück zum Zitat Nejadnik H, Hui JH, Feng Choong EP, Tai B-C, Lee EH. Autologous bone marrow-derived mesenchymal stem cells versus autologous chondrocyte implantation: an observational cohort study. Am J Sports Med. 2010;38(6):1110–6. doi:10.1177/0363546509359067.PubMedCrossRef Nejadnik H, Hui JH, Feng Choong EP, Tai B-C, Lee EH. Autologous bone marrow-derived mesenchymal stem cells versus autologous chondrocyte implantation: an observational cohort study. Am J Sports Med. 2010;38(6):1110–6. doi:10.​1177/​0363546509359067​.PubMedCrossRef
122.
Zurück zum Zitat Wakitani S, Nawata M, Tensho K, Okabe T, Machida H, Ohgushi H. Repair of articular cartilage defects in the patello-femoral joint with autologous bone marrow mesenchymal cell transplantation: three case reports involving nine defects in five knees. J Tissue Eng Regen Med. 1(1):74-9. doi:10.1002/term.8. Wakitani S, Nawata M, Tensho K, Okabe T, Machida H, Ohgushi H. Repair of articular cartilage defects in the patello-femoral joint with autologous bone marrow mesenchymal cell transplantation: three case reports involving nine defects in five knees. J Tissue Eng Regen Med. 1(1):74-9. doi:10.​1002/​term.​8.
123.
Zurück zum Zitat Lee KBL, Wang VTZ, Chan YH, Hui JHP. A novel, minimally-invasive technique of cartilage repair in the human knee using arthroscopic microfracture and injections of mesenchymal stem cells and hyaluronic acid—a prospective comparative study on safety and short-term efficacy. Ann Acad Med Singapore. 2012;41(11):511–7. Available at: http://www.ncbi.nlm.nih.gov/pubmed/23235728. Accessed 16 June 2016.PubMed Lee KBL, Wang VTZ, Chan YH, Hui JHP. A novel, minimally-invasive technique of cartilage repair in the human knee using arthroscopic microfracture and injections of mesenchymal stem cells and hyaluronic acid—a prospective comparative study on safety and short-term efficacy. Ann Acad Med Singapore. 2012;41(11):511–7. Available at: http://​www.​ncbi.​nlm.​nih.​gov/​pubmed/​23235728. Accessed 16 June 2016.PubMed
124.
125.
Zurück zum Zitat Haleem AM, El Singergy AA, Sabry D, et al. The clinical use of human culture-expanded autologous bone marrow mesenchymal stem cells transplanted on platelet-rich fibrin glue in the treatment of articular cartilage defects: a pilot study and preliminary results. Cartilage. 2010;1(4):253–61. doi:10.1177/1947603510366027.PubMedPubMedCentralCrossRef Haleem AM, El Singergy AA, Sabry D, et al. The clinical use of human culture-expanded autologous bone marrow mesenchymal stem cells transplanted on platelet-rich fibrin glue in the treatment of articular cartilage defects: a pilot study and preliminary results. Cartilage. 2010;1(4):253–61. doi:10.​1177/​1947603510366027​.PubMedPubMedCentralCrossRef
126.
128.
Zurück zum Zitat Shetty AA, Kim SJ, Shetty V, et al. Autologous bone-marrow mesenchymal cell induced chondrogenesis: single-stage arthroscopic cartilage repair. Tissue Eng Regen Med. 2014;11(3):247–53. doi:10.1007/s13770-014-0061-4.CrossRef Shetty AA, Kim SJ, Shetty V, et al. Autologous bone-marrow mesenchymal cell induced chondrogenesis: single-stage arthroscopic cartilage repair. Tissue Eng Regen Med. 2014;11(3):247–53. doi:10.​1007/​s13770-014-0061-4.CrossRef
129.
Zurück zum Zitat Akgun I, Unlu MC, Erdal OA, et al. Matrix-induced autologous mesenchymal stem cell implantation versus matrix-induced autologous chondrocyte implantation in the treatment of chondral defects of the knee: a 2-year randomized study. Arch Orthop Trauma Surg. 2015;135(2):251–63. doi:10.1007/s00402-014-2136-z.PubMedCrossRef Akgun I, Unlu MC, Erdal OA, et al. Matrix-induced autologous mesenchymal stem cell implantation versus matrix-induced autologous chondrocyte implantation in the treatment of chondral defects of the knee: a 2-year randomized study. Arch Orthop Trauma Surg. 2015;135(2):251–63. doi:10.​1007/​s00402-014-2136-z.PubMedCrossRef
130.
Zurück zum Zitat Buda R, Vannini F, Cavallo M, Grigolo B, Cenacchi A, Giannini S. Osteochondral lesions of the knee: a new one-step repair technique with bone-marrow-derived cells. J Bone Joint Surg Am. 2010;92 Suppl 2:2–11. doi:10.2106/JBJS.J.00813.PubMedCrossRef Buda R, Vannini F, Cavallo M, Grigolo B, Cenacchi A, Giannini S. Osteochondral lesions of the knee: a new one-step repair technique with bone-marrow-derived cells. J Bone Joint Surg Am. 2010;92 Suppl 2:2–11. doi:10.​2106/​JBJS.​J.​00813.PubMedCrossRef
132.
Zurück zum Zitat Gobbi A, Chaurasia S, Karnatzikos G, Nakamura N. Matrix-induced autologous chondrocyte implantation versus multipotent stem cells for the treatment of large patellofemoral chondral lesions: a nonrandomized prospective trial. Cartilage. 2015;6(2):82–97. doi:10.1177/1947603514563597.PubMedPubMedCentralCrossRef Gobbi A, Chaurasia S, Karnatzikos G, Nakamura N. Matrix-induced autologous chondrocyte implantation versus multipotent stem cells for the treatment of large patellofemoral chondral lesions: a nonrandomized prospective trial. Cartilage. 2015;6(2):82–97. doi:10.​1177/​1947603514563597​.PubMedPubMedCentralCrossRef
134.
Zurück zum Zitat Katayama R, Wakitani S, Tsumaki N, et al. Repair of articular cartilage defects in rabbits using CDMP1 gene-transfected autologous mesenchymal cells derived from bone marrow. Rheumatology (Oxford). 2004;43(8):980–5. doi:10.1093/rheumatology/keh240.CrossRef Katayama R, Wakitani S, Tsumaki N, et al. Repair of articular cartilage defects in rabbits using CDMP1 gene-transfected autologous mesenchymal cells derived from bone marrow. Rheumatology (Oxford). 2004;43(8):980–5. doi:10.​1093/​rheumatology/​keh240.CrossRef
148.
Zurück zum Zitat Mithoefer K, Williams RJ, Warren RF, et al. The microfracture technique for the treatment of articular cartilage lesions in the knee. A prospective cohort study. J Bone Joint Surg Am. 2005;87(9):1911–20. doi:10.2106/JBJS.D.02846.PubMed Mithoefer K, Williams RJ, Warren RF, et al. The microfracture technique for the treatment of articular cartilage lesions in the knee. A prospective cohort study. J Bone Joint Surg Am. 2005;87(9):1911–20. doi:10.​2106/​JBJS.​D.​02846.PubMed
149.
Zurück zum Zitat Steadman JR, Briggs KK, Rodrigo JJ, et al. Outcomes of microfracture for traumatic chondral defects of the knee: average 11-year follow-up. Arthrosc J Arthrosc Relat Surg. 2003;19(5):477–84. doi:10.1053/jars.2003.50112.CrossRef Steadman JR, Briggs KK, Rodrigo JJ, et al. Outcomes of microfracture for traumatic chondral defects of the knee: average 11-year follow-up. Arthrosc J Arthrosc Relat Surg. 2003;19(5):477–84. doi:10.​1053/​jars.​2003.​50112.CrossRef
151.
152.
Zurück zum Zitat Oshima Y, Watanabe N, Matsuda K, Takai S, Kawata M, Kubo T. Fate of transplanted bone-marrow-derived mesenchymal cells during osteochondral repair using transgenic rats to simulate autologous transplantation. Osteoarthritis Cartilage. 2004;12(10):811–7. doi:10.1016/j.joca.2004.06.014.PubMedCrossRef Oshima Y, Watanabe N, Matsuda K, Takai S, Kawata M, Kubo T. Fate of transplanted bone-marrow-derived mesenchymal cells during osteochondral repair using transgenic rats to simulate autologous transplantation. Osteoarthritis Cartilage. 2004;12(10):811–7. doi:10.​1016/​j.​joca.​2004.​06.​014.PubMedCrossRef
155.
Zurück zum Zitat Gille J, Schuseil E, Wimmer J, Gellissen J, Schulz AP, Behrens P. Mid-term results of autologous matrix-induced chondrogenesis for treatment of focal cartilage defects in the knee. Knee Surg Sports Traumatol Arthrosc. 2010;18(11):1456–64. doi:10.1007/s00167-010-1042-3.PubMedCrossRef Gille J, Schuseil E, Wimmer J, Gellissen J, Schulz AP, Behrens P. Mid-term results of autologous matrix-induced chondrogenesis for treatment of focal cartilage defects in the knee. Knee Surg Sports Traumatol Arthrosc. 2010;18(11):1456–64. doi:10.​1007/​s00167-010-1042-3.PubMedCrossRef
156.
Zurück zum Zitat Dhollander AAM, De Neve F, Almqvist KF, et al. Autologous matrix-induced chondrogenesis combined with platelet-rich plasma gel: technical description and a five pilot patients report. Knee Surg Sports Traumatol Arthrosc. 2011;19(4):536–42. doi:10.1007/s00167-010-1337-4.PubMedCrossRef Dhollander AAM, De Neve F, Almqvist KF, et al. Autologous matrix-induced chondrogenesis combined with platelet-rich plasma gel: technical description and a five pilot patients report. Knee Surg Sports Traumatol Arthrosc. 2011;19(4):536–42. doi:10.​1007/​s00167-010-1337-4.PubMedCrossRef
157.
Zurück zum Zitat Kusano T, Jakob RP, Gautier E, Magnussen RA, Hoogewoud H, Jacobi M. Treatment of isolated chondral and osteochondral defects in the knee by autologous matrix-induced chondrogenesis (AMIC). Knee Surg Sports Traumatol Arthrosc. 2012;20(10):2109–15. doi:10.1007/s00167-011-1840-2.PubMedCrossRef Kusano T, Jakob RP, Gautier E, Magnussen RA, Hoogewoud H, Jacobi M. Treatment of isolated chondral and osteochondral defects in the knee by autologous matrix-induced chondrogenesis (AMIC). Knee Surg Sports Traumatol Arthrosc. 2012;20(10):2109–15. doi:10.​1007/​s00167-011-1840-2.PubMedCrossRef
159.
Zurück zum Zitat Siclari A, Mascaro G, Gentili C, Cancedda R, Boux E. A cell-free scaffold-based cartilage repair provides improved function hyaline-like repair at one year. Clin Orthop Relat Res. 2012;470(3):910–9. doi:10.1007/s11999-011-2107-4.PubMedCrossRef Siclari A, Mascaro G, Gentili C, Cancedda R, Boux E. A cell-free scaffold-based cartilage repair provides improved function hyaline-like repair at one year. Clin Orthop Relat Res. 2012;470(3):910–9. doi:10.​1007/​s11999-011-2107-4.PubMedCrossRef
160.
Zurück zum Zitat Huade Li H, Qiang Zheng Q, Yuxiang Xiao Y, Jie Feng J, Zhongli Shi Z, Zhijun PZ. Rat cartilage repair using nanophase PLGA/HA composite and mesenchymal stem cells. J Bioact Compat Polym. 2009;24(1):83–99. doi:10.1177/0883911508100655.CrossRef Huade Li H, Qiang Zheng Q, Yuxiang Xiao Y, Jie Feng J, Zhongli Shi Z, Zhijun PZ. Rat cartilage repair using nanophase PLGA/HA composite and mesenchymal stem cells. J Bioact Compat Polym. 2009;24(1):83–99. doi:10.​1177/​0883911508100655​.CrossRef
162.
Zurück zum Zitat Charles Huang C-Y, Reuben PM, D’Ippolito G, Schiller PC, Cheung HS. Chondrogenesis of human bone marrow-derived mesenchymal stem cells in agarose culture. Anat Rec. 2004;278A(1):428–36. doi:10.1002/ar.a.20010.CrossRef Charles Huang C-Y, Reuben PM, D’Ippolito G, Schiller PC, Cheung HS. Chondrogenesis of human bone marrow-derived mesenchymal stem cells in agarose culture. Anat Rec. 2004;278A(1):428–36. doi:10.​1002/​ar.​a.​20010.CrossRef
163.
Zurück zum Zitat Koga H, Muneta T, Nagase T, et al. Comparison of mesenchymal tissues-derived stem cells for in vivo chondrogenesis: suitable conditions for cell therapy of cartilage defects in rabbit. Cell Tissue Res. 2008;333(2):207–15. doi:10.1007/s00441-008-0633-5.PubMedCrossRef Koga H, Muneta T, Nagase T, et al. Comparison of mesenchymal tissues-derived stem cells for in vivo chondrogenesis: suitable conditions for cell therapy of cartilage defects in rabbit. Cell Tissue Res. 2008;333(2):207–15. doi:10.​1007/​s00441-008-0633-5.PubMedCrossRef
167.
Zurück zum Zitat Medvedev SP, Grigor’eva EV, Shevchenko AI, et al. Human induced pluripotent stem cells derived from fetal neural stem cells successfully undergo directed differentiation into cartilage. Stem Cells Dev. 2011;20(6):1099–112. doi:10.1089/scd.2010.0249.PubMedCrossRef Medvedev SP, Grigor’eva EV, Shevchenko AI, et al. Human induced pluripotent stem cells derived from fetal neural stem cells successfully undergo directed differentiation into cartilage. Stem Cells Dev. 2011;20(6):1099–112. doi:10.​1089/​scd.​2010.​0249.PubMedCrossRef
168.
Zurück zum Zitat Millan C, Cavalli E, Groth T, Maniura-Weber K, Zenobi-Wong M. Engineered microtissues formed by schiff base crosslinking restore the chondrogenic potential of aged mesenchymal stem cells. Adv Healthc Mater. 2015;4(9):1348–58. doi:10.1002/adhm.201500102.PubMedCrossRef Millan C, Cavalli E, Groth T, Maniura-Weber K, Zenobi-Wong M. Engineered microtissues formed by schiff base crosslinking restore the chondrogenic potential of aged mesenchymal stem cells. Adv Healthc Mater. 2015;4(9):1348–58. doi:10.​1002/​adhm.​201500102.PubMedCrossRef
169.
Zurück zum Zitat Popa EG, Caridade SG, Mano JF, Reis RL, Gomes ME. Chondrogenic potential of injectable κ-carrageenan hydrogel with encapsulated adipose stem cells for cartilage tissue-engineering applications. J Tissue Eng Regen Med. 2015;9(5):550–63. doi:10.1002/term.1683.PubMedCrossRef Popa EG, Caridade SG, Mano JF, Reis RL, Gomes ME. Chondrogenic potential of injectable κ-carrageenan hydrogel with encapsulated adipose stem cells for cartilage tissue-engineering applications. J Tissue Eng Regen Med. 2015;9(5):550–63. doi:10.​1002/​term.​1683.PubMedCrossRef
170.
173.
Zurück zum Zitat Rey-Rico A, Venkatesan JK, Sohier J, Moroni L, Cucchiarini M, Madry H. Adapted chondrogenic differentiation of human mesenchymal stem cells via controlled release of TGF-β1 from poly(ethylene oxide)-terephtalate/poly(butylene terepthalate) multiblock scaffolds. J Biomed Mater Res A. 2015;103(1):371–83. doi:10.1002/jbm.a.35181.PubMedCrossRef Rey-Rico A, Venkatesan JK, Sohier J, Moroni L, Cucchiarini M, Madry H. Adapted chondrogenic differentiation of human mesenchymal stem cells via controlled release of TGF-β1 from poly(ethylene oxide)-terephtalate/poly(butylene terepthalate) multiblock scaffolds. J Biomed Mater Res A. 2015;103(1):371–83. doi:10.​1002/​jbm.​a.​35181.PubMedCrossRef
176.
Zurück zum Zitat Focaroli S, Teti G, Salvatore V, et al. Chondrogenic differentiation of human adipose mesenchimal stem cells: influence of a biomimetic gelatin genipin crosslinked porous scaffold. Microsc Res Tech. 2014;77(11):928–34. doi:10.1002/jemt.22417.PubMedCrossRef Focaroli S, Teti G, Salvatore V, et al. Chondrogenic differentiation of human adipose mesenchimal stem cells: influence of a biomimetic gelatin genipin crosslinked porous scaffold. Microsc Res Tech. 2014;77(11):928–34. doi:10.​1002/​jemt.​22417.PubMedCrossRef
177.
179.
Zurück zum Zitat Frisch J, Venkatesan J, Rey-Rico A, et al. Influence of insulin-like growth factor I overexpression via recombinant adeno-associated vector gene transfer upon the biological activities and differentiation potential of human bone marrow-derived mesenchymal stem cells. Stem Cell Res Ther. 2014;5(4):103. doi:10.1186/scrt491.PubMedPubMedCentralCrossRef Frisch J, Venkatesan J, Rey-Rico A, et al. Influence of insulin-like growth factor I overexpression via recombinant adeno-associated vector gene transfer upon the biological activities and differentiation potential of human bone marrow-derived mesenchymal stem cells. Stem Cell Res Ther. 2014;5(4):103. doi:10.​1186/​scrt491.PubMedPubMedCentralCrossRef
180.
Zurück zum Zitat Meng F, He A, Zhang Z, et al. Chondrogenic differentiation of ATDC5 and hMSCs could be induced by a novel scaffold-tricalcium phosphate-collagen-hyaluronan without any exogenous growth factors in vitro. J Biomed Mater Res Part A. 2014;102(8):2725–35. doi:10.1002/jbm.a.34948.CrossRef Meng F, He A, Zhang Z, et al. Chondrogenic differentiation of ATDC5 and hMSCs could be induced by a novel scaffold-tricalcium phosphate-collagen-hyaluronan without any exogenous growth factors in vitro. J Biomed Mater Res Part A. 2014;102(8):2725–35. doi:10.​1002/​jbm.​a.​34948.CrossRef
188.
Zurück zum Zitat Neumann AJ, Alini M, Archer CW, Stoddart MJ. Chondrogenesis of human bone marrow-derived mesenchymal stem cells is modulated by complex mechanical stimulation and adenoviral-mediated overexpression of bone morphogenetic protein 2. Tissue Eng Part A. 2013;19(11-12):1285–94. doi:10.1089/ten.TEA.2012.0411.PubMedCrossRef Neumann AJ, Alini M, Archer CW, Stoddart MJ. Chondrogenesis of human bone marrow-derived mesenchymal stem cells is modulated by complex mechanical stimulation and adenoviral-mediated overexpression of bone morphogenetic protein 2. Tissue Eng Part A. 2013;19(11-12):1285–94. doi:10.​1089/​ten.​TEA.​2012.​0411.PubMedCrossRef
189.
Zurück zum Zitat Kim D-H, Kim D-D, Yoon I-S. Proliferation and chondrogenic differentiation of human adipose-derived mesenchymal stem cells in sodium alginate beads with or without hyaluronic acid. J Pharm Investig. 2013;43(2):145–51. doi:10.1007/s40005-013-0059-2.CrossRef Kim D-H, Kim D-D, Yoon I-S. Proliferation and chondrogenic differentiation of human adipose-derived mesenchymal stem cells in sodium alginate beads with or without hyaluronic acid. J Pharm Investig. 2013;43(2):145–51. doi:10.​1007/​s40005-013-0059-2.CrossRef
191.
192.
Zurück zum Zitat Petrou M, Niemeyer P, Stoddart MJ, et al. Mesenchymal stem cell chondrogenesis: composite growth factor-bioreactor synergism for human stem cell chondrogenesis. Regen Med. 2013;8(2):157–70. doi:10.2217/rme.13.3.PubMedCrossRef Petrou M, Niemeyer P, Stoddart MJ, et al. Mesenchymal stem cell chondrogenesis: composite growth factor-bioreactor synergism for human stem cell chondrogenesis. Regen Med. 2013;8(2):157–70. doi:10.​2217/​rme.​13.​3.PubMedCrossRef
195.
Zurück zum Zitat Spoliti M, Iudicone P, Leone R, De Rosa A, Rossetti FR, Pierelli L. In vitro release and expansion of mesenchymal stem cells by a hyaluronic acid scaffold used in combination with bone marrow. Muscles Ligaments Tendons J. 2012;2(4):289–94. Available at: http://www.ncbi.nlm.nih.gov/pubmed/23738312. Accessed June 22, 2016.PubMed Spoliti M, Iudicone P, Leone R, De Rosa A, Rossetti FR, Pierelli L. In vitro release and expansion of mesenchymal stem cells by a hyaluronic acid scaffold used in combination with bone marrow. Muscles Ligaments Tendons J. 2012;2(4):289–94. Available at: http://​www.​ncbi.​nlm.​nih.​gov/​pubmed/​23738312. Accessed June 22, 2016.PubMed
198.
Zurück zum Zitat Li F, Chen Y-Z, Miao Z-N, Zheng S, Jin J. Human placenta-derived mesenchymal stem cells with silk fibroin biomaterial in the repair of articular cartilage defects. 2012. Li F, Chen Y-Z, Miao Z-N, Zheng S, Jin J. Human placenta-derived mesenchymal stem cells with silk fibroin biomaterial in the repair of articular cartilage defects. 2012.
199.
Zurück zum Zitat Popa E, Reis R, Gomes M. Chondrogenic phenotype of different cells encapsulated in κ-carrageenan hydrogels for cartilage regeneration strategies. Biotechnol Appl Biochem. 59(2):132-41. doi:10.1002/bab.1007. Popa E, Reis R, Gomes M. Chondrogenic phenotype of different cells encapsulated in κ-carrageenan hydrogels for cartilage regeneration strategies. Biotechnol Appl Biochem. 59(2):132-41. doi:10.​1002/​bab.​1007.
201.
Zurück zum Zitat Musumeci G, Lo Furno D, Loreto C, et al. Mesenchymal stem cells from adipose tissue which have been differentiated into chondrocytes in three-dimensional culture express lubricin. Exp Biol Med. 2011;236(11):1333–41. doi:10.1258/ebm.2011.011183.CrossRef Musumeci G, Lo Furno D, Loreto C, et al. Mesenchymal stem cells from adipose tissue which have been differentiated into chondrocytes in three-dimensional culture express lubricin. Exp Biol Med. 2011;236(11):1333–41. doi:10.​1258/​ebm.​2011.​011183.CrossRef
203.
Zurück zum Zitat Liu TM, Guo XM, Tan HS, Hui JH, Lim B, Lee EH. Zinc-finger protein 145, acting as an upstream regulator of SOX9, improves the differentiation potential of human mesenchymal stem cells for cartilage regeneration and repair. Arthritis Rheum. 2011;63(9):2711–20. doi:10.1002/art.30430.PubMedCrossRef Liu TM, Guo XM, Tan HS, Hui JH, Lim B, Lee EH. Zinc-finger protein 145, acting as an upstream regulator of SOX9, improves the differentiation potential of human mesenchymal stem cells for cartilage regeneration and repair. Arthritis Rheum. 2011;63(9):2711–20. doi:10.​1002/​art.​30430.PubMedCrossRef
206.
Zurück zum Zitat García-Álvarez F, Alegre-Aguarón E, Desportes P, et al. Chondrogenic differentiation in femoral bone marrow-derived mesenchymal cells (MSC) from elderly patients suffering osteoarthritis or femoral fracture. Arch Gerontol Geriatr. 52(2):239-42. doi:10.1016/j.archger.2010.03.026. García-Álvarez F, Alegre-Aguarón E, Desportes P, et al. Chondrogenic differentiation in femoral bone marrow-derived mesenchymal cells (MSC) from elderly patients suffering osteoarthritis or femoral fracture. Arch Gerontol Geriatr. 52(2):239-42. doi:10.​1016/​j.​archger.​2010.​03.​026.
208.
Zurück zum Zitat Baumgartner L, Arnhold S, Brixius K, Addicks K, Bloch W. Human mesenchymal stem cells: influence of oxygen pressure on proliferation and chondrogenic differentiation in fibrin glue in vitro. J Biomed Mater Res Part A. 2009;9999A(3):NA-NA. doi:10.1002/jbm.a.32577. Baumgartner L, Arnhold S, Brixius K, Addicks K, Bloch W. Human mesenchymal stem cells: influence of oxygen pressure on proliferation and chondrogenic differentiation in fibrin glue in vitro. J Biomed Mater Res Part A. 2009;9999A(3):NA-NA. doi:10.​1002/​jbm.​a.​32577.
209.
Zurück zum Zitat Kim H-J, Lee J-H, Im G-I. Chondrogenesis using mesenchymal stem cells and PCL scaffolds. J Biomed Mater Res Part A. 2009;9999A(2):NA-NA. doi:10.1002/jbm.a.32414. Kim H-J, Lee J-H, Im G-I. Chondrogenesis using mesenchymal stem cells and PCL scaffolds. J Biomed Mater Res Part A. 2009;9999A(2):NA-NA. doi:10.​1002/​jbm.​a.​32414.
212.
Zurück zum Zitat Angele P, Müller R, Schumann D, et al. Characterization of esterified hyaluronan-gelatin polymer composites suitable for chondrogenic differentiation of mesenchymal stem cells. J Biomed Mater Res Part A. 2009;91A(2):416–27. doi:10.1002/jbm.a.32236.CrossRef Angele P, Müller R, Schumann D, et al. Characterization of esterified hyaluronan-gelatin polymer composites suitable for chondrogenic differentiation of mesenchymal stem cells. J Biomed Mater Res Part A. 2009;91A(2):416–27. doi:10.​1002/​jbm.​a.​32236.CrossRef
215.
217.
Zurück zum Zitat Heymer A, Bradica G, Eulert J, Nöth U. Multiphasic collagen fibre-PLA composites seeded with human mesenchymal stem cells for osteochondral defect repair: an in vitro study. J Tissue Eng Regen Med. 2009;3(5):389–97. doi:10.1002/term.175.PubMedCrossRef Heymer A, Bradica G, Eulert J, Nöth U. Multiphasic collagen fibre-PLA composites seeded with human mesenchymal stem cells for osteochondral defect repair: an in vitro study. J Tissue Eng Regen Med. 2009;3(5):389–97. doi:10.​1002/​term.​175.PubMedCrossRef
218.
Zurück zum Zitat Pilgaard L, Lund P, Duroux M, et al. Effect of oxygen concentration, culture format and donor variability on in vitro chondrogenesis of human adipose tissue-derived stem cells. Regen Med. 2009;4(4):539–48. doi:10.2217/rme.09.28.PubMedCrossRef Pilgaard L, Lund P, Duroux M, et al. Effect of oxygen concentration, culture format and donor variability on in vitro chondrogenesis of human adipose tissue-derived stem cells. Regen Med. 2009;4(4):539–48. doi:10.​2217/​rme.​09.​28.PubMedCrossRef
219.
220.
Zurück zum Zitat Cheng N-C, Estes BT, Awad HA, Guilak F. Chondrogenic differentiation of adipose-derived adult stem cells by a porous scaffold derived from native articular cartilage extracellular matrix. Tissue Eng Part A. 2009;15(2):231–41. doi:10.1089/ten.tea.2008.0253.PubMedCrossRef Cheng N-C, Estes BT, Awad HA, Guilak F. Chondrogenic differentiation of adipose-derived adult stem cells by a porous scaffold derived from native articular cartilage extracellular matrix. Tissue Eng Part A. 2009;15(2):231–41. doi:10.​1089/​ten.​tea.​2008.​0253.PubMedCrossRef
222.
Zurück zum Zitat Miyamoto C, Matsumoto T, Sakimura K, Shindo H. Osteogenic protein-1 with transforming growth factor-β1: potent inducer of chondrogenesis of synovial mesenchymal stem cells in vitro. J Orthop Sci. 2007;12(6):555–61. doi:10.1007/s00776-007-1176-4.PubMedCrossRef Miyamoto C, Matsumoto T, Sakimura K, Shindo H. Osteogenic protein-1 with transforming growth factor-β1: potent inducer of chondrogenesis of synovial mesenchymal stem cells in vitro. J Orthop Sci. 2007;12(6):555–61. doi:10.​1007/​s00776-007-1176-4.PubMedCrossRef
224.
Zurück zum Zitat Estes BT, Wu AW, Guilak F. Potent induction of chondrocytic differentiation of human adipose-derived adult stem cells by bone morphogenetic protein 6. Arthritis Rheum. 2006;54(4):1222–32. doi:10.1002/art.21779.PubMedCrossRef Estes BT, Wu AW, Guilak F. Potent induction of chondrocytic differentiation of human adipose-derived adult stem cells by bone morphogenetic protein 6. Arthritis Rheum. 2006;54(4):1222–32. doi:10.​1002/​art.​21779.PubMedCrossRef
226.
Zurück zum Zitat Yokoyama A, Sekiya I, Miyazaki K, Ichinose S, Hata Y, Muneta T. In vitro cartilage formation of composites of synovium-derived mesenchymal stem cells with collagen gel. Cell Tissue Res. 2005;322(2):289–98. doi:10.1007/s00441-005-0010-6.PubMedCrossRef Yokoyama A, Sekiya I, Miyazaki K, Ichinose S, Hata Y, Muneta T. In vitro cartilage formation of composites of synovium-derived mesenchymal stem cells with collagen gel. Cell Tissue Res. 2005;322(2):289–98. doi:10.​1007/​s00441-005-0010-6.PubMedCrossRef
227.
Zurück zum Zitat Sekiya I, Larson BL, Vuoristo JT, Reger RL, Prockop DJ. Comparison of effect of BMP-2, -4, and -6 on in vitro cartilage formation of human adult stem cells from bone marrow stroma. Cell Tissue Res. 2005;320(2):269–76. doi:10.1007/s00441-004-1075-3.PubMedCrossRef Sekiya I, Larson BL, Vuoristo JT, Reger RL, Prockop DJ. Comparison of effect of BMP-2, -4, and -6 on in vitro cartilage formation of human adult stem cells from bone marrow stroma. Cell Tissue Res. 2005;320(2):269–76. doi:10.​1007/​s00441-004-1075-3.PubMedCrossRef
231.
235.
236.
Zurück zum Zitat Chen W-C, Wei Y-H, Chu I-M, Yao C-L. Effect of chondroitin sulphate C on the in vitro and in vivo chondrogenesis of mesenchymal stem cells in crosslinked type II collagen scaffolds. J Tissue Eng Regen Med. 2013;7(8):665–72. doi:10.1002/term.1463.PubMedCrossRef Chen W-C, Wei Y-H, Chu I-M, Yao C-L. Effect of chondroitin sulphate C on the in vitro and in vivo chondrogenesis of mesenchymal stem cells in crosslinked type II collagen scaffolds. J Tissue Eng Regen Med. 2013;7(8):665–72. doi:10.​1002/​term.​1463.PubMedCrossRef
237.
Zurück zum Zitat Bornes TD, Jomha NM, Mulet-Sierra A, et al. Hypoxic culture of bone marrow-derived mesenchymal stromal stem cells differentially enhances in vitro chondrogenesis within cell-seeded collagen and hyaluronic acid porous scaffolds. Stem Cell Res Ther. 2015;6(1):84. doi:10.1186/s13287-015-0075-4.PubMedPubMedCentralCrossRef Bornes TD, Jomha NM, Mulet-Sierra A, et al. Hypoxic culture of bone marrow-derived mesenchymal stromal stem cells differentially enhances in vitro chondrogenesis within cell-seeded collagen and hyaluronic acid porous scaffolds. Stem Cell Res Ther. 2015;6(1):84. doi:10.​1186/​s13287-015-0075-4.PubMedPubMedCentralCrossRef
239.
240.
Zurück zum Zitat Lam J, Lu S, Meretoja VV, Tabata Y, Mikos AG, Kasper FK. Generation of osteochondral tissue constructs with chondrogenically and osteogenically predifferentiated mesenchymal stem cells encapsulated in bilayered hydrogels. Acta Biomater. 2014;10(3):1112–23. doi:10.1016/j.actbio.2013.11.020.PubMedCrossRef Lam J, Lu S, Meretoja VV, Tabata Y, Mikos AG, Kasper FK. Generation of osteochondral tissue constructs with chondrogenically and osteogenically predifferentiated mesenchymal stem cells encapsulated in bilayered hydrogels. Acta Biomater. 2014;10(3):1112–23. doi:10.​1016/​j.​actbio.​2013.​11.​020.PubMedCrossRef
241.
Zurück zum Zitat Zhang L, Yuan T, Guo L, Zhang X. An in vitro study of collagen hydrogel to induce the chondrogenic differentiation of mesenchymal stem cells. J Biomed Mater Res Part A. 2012;100A(10):2717–25. doi:10.1002/jbm.a.34194.CrossRef Zhang L, Yuan T, Guo L, Zhang X. An in vitro study of collagen hydrogel to induce the chondrogenic differentiation of mesenchymal stem cells. J Biomed Mater Res Part A. 2012;100A(10):2717–25. doi:10.​1002/​jbm.​a.​34194.CrossRef
243.
Zurück zum Zitat Giannoni P, Lazzarini E, Ceseracciu L, Barone AC, Quarto R, Scaglione S. Design and characterization of a tissue-engineered bilayer scaffold for osteochondral tissue repair. J Tissue Eng Regen Med. 2015;9(10):1182–92. doi:10.1002/term.1651.PubMedCrossRef Giannoni P, Lazzarini E, Ceseracciu L, Barone AC, Quarto R, Scaglione S. Design and characterization of a tissue-engineered bilayer scaffold for osteochondral tissue repair. J Tissue Eng Regen Med. 2015;9(10):1182–92. doi:10.​1002/​term.​1651.PubMedCrossRef
246.
248.
250.
Zurück zum Zitat Eslaminejad MB, Taghiyar L, Falahi F. Co-culture of mesenchymal stem cells with mature chondrocytes: producing cartilage construct for application in cartilage regeneration. Iran J Med Sci. 2015;34(4):251–8. Eslaminejad MB, Taghiyar L, Falahi F. Co-culture of mesenchymal stem cells with mature chondrocytes: producing cartilage construct for application in cartilage regeneration. Iran J Med Sci. 2015;34(4):251–8.
254.
Zurück zum Zitat Worster AA, Brower-Toland BD, Fortier LA, Bent SJ, Williams J, Nixon AJ. Chondrocytic differentiation of mesenchymal stem cells sequentially exposed to transforming growth factor-beta1 in monolayer and insulin-like growth factor-I in a three-dimensional matrix. J Orthop Res. 2001;19(4):738–49. doi:10.1016/S0736-0266(00)00054-1.PubMedCrossRef Worster AA, Brower-Toland BD, Fortier LA, Bent SJ, Williams J, Nixon AJ. Chondrocytic differentiation of mesenchymal stem cells sequentially exposed to transforming growth factor-beta1 in monolayer and insulin-like growth factor-I in a three-dimensional matrix. J Orthop Res. 2001;19(4):738–49. doi:10.​1016/​S0736-0266(00)00054-1.PubMedCrossRef
255.
Zurück zum Zitat Angele P, Kujat R, Nerlich M, Yoo J, Goldberg V, Johnstone B. Engineering of osteochondral tissue with bone marrow mesenchymal progenitor cells in a derivatized hyaluronan-gelatin composite sponge. 2007. http://dx.doi.org/101089/ten19995545. Accessed June 2016. Angele P, Kujat R, Nerlich M, Yoo J, Goldberg V, Johnstone B. Engineering of osteochondral tissue with bone marrow mesenchymal progenitor cells in a derivatized hyaluronan-gelatin composite sponge. 2007. http://​dx.​doi.​org/​101089/​ten19995545. Accessed June 2016.
256.
Zurück zum Zitat Ding X, Zhu M, Xu B, et al. Integrated trilayered silk fibroin scaffold for osteochondral differentiation of adipose-derived stem cells. ACS Appl Mater Interfaces. 2014;6(19):16696–705. doi:10.1021/am5036708.PubMedCrossRef Ding X, Zhu M, Xu B, et al. Integrated trilayered silk fibroin scaffold for osteochondral differentiation of adipose-derived stem cells. ACS Appl Mater Interfaces. 2014;6(19):16696–705. doi:10.​1021/​am5036708.PubMedCrossRef
257.
Zurück zum Zitat Lu C-H, Lin K-J, Chiu H-Y, et al. Improved chondrogenesis and engineered cartilage formation from TGF-β3-expressing adipose-derived stem cells cultured in the rotating-shaft bioreactor. Tissue Eng Part A. 2012;18(19-20):2114–24. doi:10.1089/ten.TEA.2012.0010.PubMedCrossRef Lu C-H, Lin K-J, Chiu H-Y, et al. Improved chondrogenesis and engineered cartilage formation from TGF-β3-expressing adipose-derived stem cells cultured in the rotating-shaft bioreactor. Tissue Eng Part A. 2012;18(19-20):2114–24. doi:10.​1089/​ten.​TEA.​2012.​0010.PubMedCrossRef
262.
Zurück zum Zitat Han Y, Wei Y, Wang S, Song Y. Regenerative and technological section enhanced chondrogenesis of adipose-derived stem cells by the controlled release of transforming growth factor-NL 1 from hybrid microspheres. Gerontology. 2009;55:592–9. doi:10.1159/000235547.PubMedCrossRef Han Y, Wei Y, Wang S, Song Y. Regenerative and technological section enhanced chondrogenesis of adipose-derived stem cells by the controlled release of transforming growth factor-NL 1 from hybrid microspheres. Gerontology. 2009;55:592–9. doi:10.​1159/​000235547.PubMedCrossRef
263.
267.
Zurück zum Zitat Froelich K, Setiawan LE, Technau A, et al. Influence of different growth factors on chondrogenic differentiation of adipose-derived stem cells in polyurethane-fibrin composites. Int J Artif Organs. 2012;35(12):1047–60. doi:10.5301/ijao.5000132.PubMedCrossRef Froelich K, Setiawan LE, Technau A, et al. Influence of different growth factors on chondrogenic differentiation of adipose-derived stem cells in polyurethane-fibrin composites. Int J Artif Organs. 2012;35(12):1047–60. doi:10.​5301/​ijao.​5000132.PubMedCrossRef
270.
Zurück zum Zitat Park Y, Sugimoto M, Watrin A, Chiquet M, Hunziker EB. BMP-2 induces the expression of chondrocyte-specific genes in bovine synovium-derived progenitor cells cultured in three-dimensional alginate hydrogel. Osteoarthritis Cartilage. 2005;13(6):527–36. doi:10.1016/j.joca.2005.02.006.PubMedCrossRef Park Y, Sugimoto M, Watrin A, Chiquet M, Hunziker EB. BMP-2 induces the expression of chondrocyte-specific genes in bovine synovium-derived progenitor cells cultured in three-dimensional alginate hydrogel. Osteoarthritis Cartilage. 2005;13(6):527–36. doi:10.​1016/​j.​joca.​2005.​02.​006.PubMedCrossRef
275.
Zurück zum Zitat Shao X, Goh JCH, Hutmacher DW, Lee EH, Zigang G. Repair of large articular osteochondral defects using hybrid scaffolds and bone marrow-derived mesenchymal stem cells in a rabbit model. Tissue Eng. 2006;12(6):1539–51. doi:10.1089/ten.2006.12.1539.PubMedCrossRef Shao X, Goh JCH, Hutmacher DW, Lee EH, Zigang G. Repair of large articular osteochondral defects using hybrid scaffolds and bone marrow-derived mesenchymal stem cells in a rabbit model. Tissue Eng. 2006;12(6):1539–51. doi:10.​1089/​ten.​2006.​12.​1539.PubMedCrossRef
276.
277.
Zurück zum Zitat Zhu S, Zhang B, Man C, Ma Y, Liu X, Hu J. Combined effects of connective tissue growth factor-modified bone marrow-derived mesenchymal stem cells and NaOH-treated PLGA scaffolds on repair of articular cartilage defect in rabbits. Cell Transplant. 2013. doi:10.3727/096368913X6697790. Zhu S, Zhang B, Man C, Ma Y, Liu X, Hu J. Combined effects of connective tissue growth factor-modified bone marrow-derived mesenchymal stem cells and NaOH-treated PLGA scaffolds on repair of articular cartilage defect in rabbits. Cell Transplant. 2013. doi:10.​3727/​096368913X669779​0.
278.
Zurück zum Zitat Oshima Y, Watanabe N, Matsuda K, Takai S, Kawata M, Kubo T. Behavior of transplanted bone marrow-derived GFP mesenchymal cells in osteochondral defect as a simulation of autologous transplantation. J Histochem Cytochem. 2005;53(2):207–16. doi:10.1369/jhc.4A6280.2005.PubMedCrossRef Oshima Y, Watanabe N, Matsuda K, Takai S, Kawata M, Kubo T. Behavior of transplanted bone marrow-derived GFP mesenchymal cells in osteochondral defect as a simulation of autologous transplantation. J Histochem Cytochem. 2005;53(2):207–16. doi:10.​1369/​jhc.​4A6280.​2005.PubMedCrossRef
279.
Zurück zum Zitat Lim CT, Ren X, Afizah MH, et al. Repair of osteochondral defects with rehydrated freeze-dried oligo[poly(ethylene glycol) fumarate] hydrogels seeded with bone marrow mesenchymal stem cells in a porcine model. Tissue Eng Part A. 2013;19(15-16):1852–61. doi:10.1089/ten.TEA.2012.0621.PubMedCrossRef Lim CT, Ren X, Afizah MH, et al. Repair of osteochondral defects with rehydrated freeze-dried oligo[poly(ethylene glycol) fumarate] hydrogels seeded with bone marrow mesenchymal stem cells in a porcine model. Tissue Eng Part A. 2013;19(15-16):1852–61. doi:10.​1089/​ten.​TEA.​2012.​0621.PubMedCrossRef
280.
Zurück zum Zitat Li T, Nina F, Xiaozuo T, Xiaopeng L, Zhuo W, Na L. Chondrogenic differentiation of mesenchymal stem cells for repairing articular cartilage. J Clin Rehabil Tissue Eng Res. 2009;13(46):9041–4. Li T, Nina F, Xiaozuo T, Xiaopeng L, Zhuo W, Na L. Chondrogenic differentiation of mesenchymal stem cells for repairing articular cartilage. J Clin Rehabil Tissue Eng Res. 2009;13(46):9041–4.
281.
284.
Zurück zum Zitat Ishihara K, Nakayama K, Akieda S, Matsuda S, Iwamoto Y. Simultaneous regeneration of full-thickness cartilage and subchondral bone defects in vivo using a three-dimensional scaffold-free autologous construct derived from high-density bone marrow-derived mesenchymal stem cells. J Orthop Surg Res. 2014;9:98. doi:10.1186/s13018-014-0098-z.PubMedPubMedCentralCrossRef Ishihara K, Nakayama K, Akieda S, Matsuda S, Iwamoto Y. Simultaneous regeneration of full-thickness cartilage and subchondral bone defects in vivo using a three-dimensional scaffold-free autologous construct derived from high-density bone marrow-derived mesenchymal stem cells. J Orthop Surg Res. 2014;9:98. doi:10.​1186/​s13018-014-0098-z.PubMedPubMedCentralCrossRef
285.
286.
Zurück zum Zitat Xue D, Zheng Q, Zong C, et al. Osteochondral repair using porous poly(lactide-co-glycolide)/nano-hydroxyapatite hybrid scaffolds with undifferentiated mesenchymal stem cells in a rat model. J Biomed Mater Res A. 2010;94(1):259–70. doi:10.1002/jbm.a.32691.PubMedCrossRef Xue D, Zheng Q, Zong C, et al. Osteochondral repair using porous poly(lactide-co-glycolide)/nano-hydroxyapatite hybrid scaffolds with undifferentiated mesenchymal stem cells in a rat model. J Biomed Mater Res A. 2010;94(1):259–70. doi:10.​1002/​jbm.​a.​32691.PubMedCrossRef
287.
Zurück zum Zitat Wayne JS, McDowell CL, Shields KJ, Tuan RS. In vivo response of polylactic acid-alginate scaffolds and bone marrow-derived cells for cartilage tissue engineering. Tissue Eng. 11(5-6):953-63. doi:10.1089/ten.2005.11.953. Wayne JS, McDowell CL, Shields KJ, Tuan RS. In vivo response of polylactic acid-alginate scaffolds and bone marrow-derived cells for cartilage tissue engineering. Tissue Eng. 11(5-6):953-63. doi:10.​1089/​ten.​2005.​11.​953.
288.
Zurück zum Zitat Bal BS, Rahaman MN, Jayabalan P, et al. In vivo outcomes of tissue-engineered osteochondral grafts. J Biomed Mater Res B Appl Biomater. 2010;93(1):164–74. doi:10.1002/jbm.b.31571.PubMed Bal BS, Rahaman MN, Jayabalan P, et al. In vivo outcomes of tissue-engineered osteochondral grafts. J Biomed Mater Res B Appl Biomater. 2010;93(1):164–74. doi:10.​1002/​jbm.​b.​31571.PubMed
289.
Zurück zum Zitat Qi Y, Du Y, Li W, Dai X, Zhao T, Yan W. Cartilage repair using mesenchymal stem cell (MSC) sheet and MSCs-loaded bilayer PLGA scaffold in a rabbit model. Knee Surg Sports Traumatol Arthrosc. 2014;22(6):1424–33. doi:10.1007/s00167-012-2256-3.PubMedCrossRef Qi Y, Du Y, Li W, Dai X, Zhao T, Yan W. Cartilage repair using mesenchymal stem cell (MSC) sheet and MSCs-loaded bilayer PLGA scaffold in a rabbit model. Knee Surg Sports Traumatol Arthrosc. 2014;22(6):1424–33. doi:10.​1007/​s00167-012-2256-3.PubMedCrossRef
290.
291.
Zurück zum Zitat Fan H, Hu Y, Qin L, Li X, Wu H, Lv R. Porous gelatin-chondroitin-hyaluronate tri-copolymer scaffold containing microspheres loaded with TGF-beta1 induces differentiation of mesenchymal stem cells in vivo for enhancing cartilage repair. J Biomed Mater Res A. 2006;77(4):785–94. doi:10.1002/jbm.a.30647.PubMedCrossRef Fan H, Hu Y, Qin L, Li X, Wu H, Lv R. Porous gelatin-chondroitin-hyaluronate tri-copolymer scaffold containing microspheres loaded with TGF-beta1 induces differentiation of mesenchymal stem cells in vivo for enhancing cartilage repair. J Biomed Mater Res A. 2006;77(4):785–94. doi:10.​1002/​jbm.​a.​30647.PubMedCrossRef
296.
297.
Zurück zum Zitat Løken S, Jakobsen RB, Arøen A, et al. Bone marrow mesenchymal stem cells in a hyaluronan scaffold for treatment of an osteochondral defect in a rabbit model. Knee Surg Sports Traumatol Arthrosc. 2008;16(10):896–903. doi:10.1007/s00167-008-0566-2.PubMedCrossRef Løken S, Jakobsen RB, Arøen A, et al. Bone marrow mesenchymal stem cells in a hyaluronan scaffold for treatment of an osteochondral defect in a rabbit model. Knee Surg Sports Traumatol Arthrosc. 2008;16(10):896–903. doi:10.​1007/​s00167-008-0566-2.PubMedCrossRef
298.
Zurück zum Zitat Tan W, Zha Z, Zhang J, Zheng L, Liang Y XJ. Animal-origin osteochondral scaffold combined with bone marrow mesenchymal stem cells/chondrocytes for repair of composite osteochondral defects in rabbit knee joints. J Clin Rehabil Tissue Eng Res. 2011;15(12). doi:10.3969/j.issn.1673-8225.2011.12.043. Tan W, Zha Z, Zhang J, Zheng L, Liang Y XJ. Animal-origin osteochondral scaffold combined with bone marrow mesenchymal stem cells/chondrocytes for repair of composite osteochondral defects in rabbit knee joints. J Clin Rehabil Tissue Eng Res. 2011;15(12). doi:10.​3969/​j.​issn.​1673-8225.​2011.​12.​043.
299.
Zurück zum Zitat Hu B, Ren J-L, Zhang J-R, Ma Q, Liu Y-P, Mao T-Q. Enhanced treatment of articular cartilage defect of the knee by intra-articular injection of Bcl-xL-engineered mesenchymal stem cells in rabbit model. J Tissue Eng Regen Med. 2010;4(2):105–14. doi:10.1002/term.212.PubMedCrossRef Hu B, Ren J-L, Zhang J-R, Ma Q, Liu Y-P, Mao T-Q. Enhanced treatment of articular cartilage defect of the knee by intra-articular injection of Bcl-xL-engineered mesenchymal stem cells in rabbit model. J Tissue Eng Regen Med. 2010;4(2):105–14. doi:10.​1002/​term.​212.PubMedCrossRef
300.
Zurück zum Zitat Xie J, Han Z, Naito M, et al. Articular cartilage tissue engineering based on a mechano-active scaffold made of poly(L-lactide-co-epsilon-caprolactone): in vivo performance in adult rabbits. J Biomed Mater Res B Appl Biomater. 2010;94(1):80–8. doi:10.1002/jbm.b.31627.PubMed Xie J, Han Z, Naito M, et al. Articular cartilage tissue engineering based on a mechano-active scaffold made of poly(L-lactide-co-epsilon-caprolactone): in vivo performance in adult rabbits. J Biomed Mater Res B Appl Biomater. 2010;94(1):80–8. doi:10.​1002/​jbm.​b.​31627.PubMed
302.
Zurück zum Zitat Jeong W-K, Oh S-H, Lee J-H, Im G-I. Repair of osteochondral defects with a construct of mesenchymal stem cells and a polydioxanone/poly(vinyl alcohol) scaffold. Biotechnol Appl Biochem. 2008;49(Pt 2):155–64. doi:10.1042/BA20070149.PubMedCrossRef Jeong W-K, Oh S-H, Lee J-H, Im G-I. Repair of osteochondral defects with a construct of mesenchymal stem cells and a polydioxanone/poly(vinyl alcohol) scaffold. Biotechnol Appl Biochem. 2008;49(Pt 2):155–64. doi:10.​1042/​BA20070149.PubMedCrossRef
304.
Zurück zum Zitat Kayakabe M, Tsutsumi S, Watanabe H, Kato Y, Takagishi K. Transplantation of autologous rabbit BM-derived mesenchymal stromal cells embedded in hyaluronic acid gel sponge into osteochondral defects of the knee. Cytotherapy. 2006;8(4):343–53. doi:10.1080/14653240600845070.PubMedCrossRef Kayakabe M, Tsutsumi S, Watanabe H, Kato Y, Takagishi K. Transplantation of autologous rabbit BM-derived mesenchymal stromal cells embedded in hyaluronic acid gel sponge into osteochondral defects of the knee. Cytotherapy. 2006;8(4):343–53. doi:10.​1080/​1465324060084507​0.PubMedCrossRef
306.
Zurück zum Zitat Yoshioka T, Mishima H, Sakai S, Uemura T. Long-term results of cartilage repair after allogeneic transplantation of cartilaginous aggregates formed from bone marrow-derived cells for large osteochondral defects in rabbit knees. Cartilage. 2013;4(4):339–44. doi:10.1177/1947603513494003.PubMedPubMedCentralCrossRef Yoshioka T, Mishima H, Sakai S, Uemura T. Long-term results of cartilage repair after allogeneic transplantation of cartilaginous aggregates formed from bone marrow-derived cells for large osteochondral defects in rabbit knees. Cartilage. 2013;4(4):339–44. doi:10.​1177/​1947603513494003​.PubMedPubMedCentralCrossRef
308.
Zurück zum Zitat Jung M, Kaszap B, Redöhl A, et al. Enhanced early tissue regeneration after matrix-assisted autologous mesenchymal stem cell transplantation in full thickness chondral defects in a minipig model. Cell Transplant. 2009;18(8):923–32. doi:10.3727/096368909X471297.PubMedCrossRef Jung M, Kaszap B, Redöhl A, et al. Enhanced early tissue regeneration after matrix-assisted autologous mesenchymal stem cell transplantation in full thickness chondral defects in a minipig model. Cell Transplant. 2009;18(8):923–32. doi:10.​3727/​096368909X471297​.PubMedCrossRef
309.
311.
Zurück zum Zitat Coleman RM, Schwartz Z, Boyan BD, Guldberg RE. The therapeutic effect of bone marrow-derived stem cell implantation after epiphyseal plate injury is abrogated by chondrogenic predifferentiation. Tissue Eng Part A. 2013;19(3-4):475–83. doi:10.1089/ten.TEA.2012.0125.PubMedCrossRef Coleman RM, Schwartz Z, Boyan BD, Guldberg RE. The therapeutic effect of bone marrow-derived stem cell implantation after epiphyseal plate injury is abrogated by chondrogenic predifferentiation. Tissue Eng Part A. 2013;19(3-4):475–83. doi:10.​1089/​ten.​TEA.​2012.​0125.PubMedCrossRef
313.
Zurück zum Zitat Marquass B, Schulz R, Hepp P, et al. Matrix-associated implantation of predifferentiated mesenchymal stem cells versus articular chondrocytes: in vivo results of cartilage repair after 1 year. Am J Sports Med. 2011;39(7):1401–12. doi:10.1177/0363546511398646.PubMedCrossRef Marquass B, Schulz R, Hepp P, et al. Matrix-associated implantation of predifferentiated mesenchymal stem cells versus articular chondrocytes: in vivo results of cartilage repair after 1 year. Am J Sports Med. 2011;39(7):1401–12. doi:10.​1177/​0363546511398646​.PubMedCrossRef
315.
Zurück zum Zitat Guo X, Wang C, Zhang Y, et al. Repair of large articular cartilage defects with implants of autologous mesenchymal stem cells seeded into beta-tricalcium phosphate in a sheep model. Tissue Eng. 10(11-12):1818-29. doi:10.1089/ten.2004.10.1818. Guo X, Wang C, Zhang Y, et al. Repair of large articular cartilage defects with implants of autologous mesenchymal stem cells seeded into beta-tricalcium phosphate in a sheep model. Tissue Eng. 10(11-12):1818-29. doi:10.​1089/​ten.​2004.​10.​1818.
316.
Zurück zum Zitat Caminal M, Moll X, Codina D, et al. Transitory improvement of articular cartilage characteristics after implantation of polylactide:polyglycolic acid (PLGA) scaffolds seeded with autologous mesenchymal stromal cells in a sheep model of critical-sized chondral defect. Biotechnol Lett. 2014;36(10):2143–53. doi:10.1007/s10529-014-1585-3.PubMedCrossRef Caminal M, Moll X, Codina D, et al. Transitory improvement of articular cartilage characteristics after implantation of polylactide:polyglycolic acid (PLGA) scaffolds seeded with autologous mesenchymal stromal cells in a sheep model of critical-sized chondral defect. Biotechnol Lett. 2014;36(10):2143–53. doi:10.​1007/​s10529-014-1585-3.PubMedCrossRef
317.
Zurück zum Zitat Wilke MM, Nydam DV, Nixon AJ. Enhanced early chondrogenesis in articular defects following arthroscopic mesenchymal stem cell implantation in an equine model. J Orthop Res. 2007;25(7):913–25. doi:10.1002/jor.20382.PubMedCrossRef Wilke MM, Nydam DV, Nixon AJ. Enhanced early chondrogenesis in articular defects following arthroscopic mesenchymal stem cell implantation in an equine model. J Orthop Res. 2007;25(7):913–25. doi:10.​1002/​jor.​20382.PubMedCrossRef
321.
Zurück zum Zitat Qi B, Yu A, Zhu S, Zhou M, Wu G. Chitosan/poly(vinyl alcohol) hydrogel combined with Ad-hTGF-β1 transfected mesenchymal stem cells to repair rabbit articular cartilage defects. Exp Biol Med (Maywood). 2013;238(1):23–30. doi:10.1258/ebm.2012.012223.CrossRef Qi B, Yu A, Zhu S, Zhou M, Wu G. Chitosan/poly(vinyl alcohol) hydrogel combined with Ad-hTGF-β1 transfected mesenchymal stem cells to repair rabbit articular cartilage defects. Exp Biol Med (Maywood). 2013;238(1):23–30. doi:10.​1258/​ebm.​2012.​012223.CrossRef
322.
Zurück zum Zitat Qi Y, Zhao T, Xu K, Dai T, Yan W. The restoration of full-thickness cartilage defects with mesenchymal stem cells (MSCs) loaded and cross-linked bilayer collagen scaffolds on rabbit model. Mol Biol Rep. 2012;39(2):1231–7. doi:10.1007/s11033-011-0853-8.PubMedCrossRef Qi Y, Zhao T, Xu K, Dai T, Yan W. The restoration of full-thickness cartilage defects with mesenchymal stem cells (MSCs) loaded and cross-linked bilayer collagen scaffolds on rabbit model. Mol Biol Rep. 2012;39(2):1231–7. doi:10.​1007/​s11033-011-0853-8.PubMedCrossRef
323.
Zurück zum Zitat Park JS, Woo DG, Yang HN, et al. Chondrogenesis of human mesenchymal stem cells encapsulated in a hydrogel construct: neocartilage formation in animal models as both mice and rabbits. J Biomed Mater Res A. 2010;92(3):988–96. doi:10.1002/jbm.a.32341.PubMed Park JS, Woo DG, Yang HN, et al. Chondrogenesis of human mesenchymal stem cells encapsulated in a hydrogel construct: neocartilage formation in animal models as both mice and rabbits. J Biomed Mater Res A. 2010;92(3):988–96. doi:10.​1002/​jbm.​a.​32341.PubMed
324.
Zurück zum Zitat Liu Y, Shu XZ, Prestwich GD. Osteochondral defect repair with autologous bone marrow-derived mesenchymal stem cells in an injectable, in situ, cross-linked synthetic extracellular matrix. Tissue Eng. 2006;12(12):3405–16. doi:10.1089/ten.2006.12.3405.PubMedCrossRef Liu Y, Shu XZ, Prestwich GD. Osteochondral defect repair with autologous bone marrow-derived mesenchymal stem cells in an injectable, in situ, cross-linked synthetic extracellular matrix. Tissue Eng. 2006;12(12):3405–16. doi:10.​1089/​ten.​2006.​12.​3405.PubMedCrossRef
Metadaten
Titel
The use of mesenchymal stem cells for cartilage repair and regeneration: a systematic review
verfasst von
Andy Goldberg
Katrina Mitchell
Julian Soans
Louise Kim
Razi Zaidi
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Journal of Orthopaedic Surgery and Research / Ausgabe 1/2017
Elektronische ISSN: 1749-799X
DOI
https://doi.org/10.1186/s13018-017-0534-y

Weitere Artikel der Ausgabe 1/2017

Journal of Orthopaedic Surgery and Research 1/2017 Zur Ausgabe

Arthropedia

Grundlagenwissen der Arthroskopie und Gelenkchirurgie. Erweitert durch Fallbeispiele, Videos und Abbildungen. 
» Jetzt entdecken

Update Orthopädie und Unfallchirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.