Abstract
Context:
Articular cartilage possesses poor natural healing mechanisms, and a variety of non-cell-based and cell-based treatments aim to promote regeneration of hyaline cartilage.
Data Sources:
A review of the literature to December 2013 using PubMed with search criteria including the keywords stem cell, cell therapy, cell transplantation, cartilage, chondral, and chondrogenic.
Study Selection:
Forty-five articles were identified that employed local mesenchymal stem cell (MSC) therapy for joint disorders in humans. Nine comparative studies were identified, consisting of 3 randomized trials, 5 cohort studies, and 1 case-control study.
Study Type:
Clinical review.
Level of Evidence:
Level 4.
Data Extraction:
Studies were assessed for stem cell source, method of implantation, comparison groups, and concurrent surgical techniques.
Results:
Two studies comparing MSC treatment to autologous chondrocyte implantation found similar efficacy. Three studies reported clinical benefits with intra-articular MSC injection over non-MSC controls for cases undergoing debridement with or without marrow stimulation, although a randomized study found no significant clinical difference at 2-year follow-up but reported better 18-month magnetic resonance imaging and histologic scores in the MSC group. No human studies have compared intra-articular MSC therapy to non-MSC techniques for osteoarthritis in the absence of surgery.
Conclusion:
Mesenchymal stem cell–based therapies appear safe and effective for joint disorders in large animal preclinical models. Evidence for use in humans, particularly, comparison with more established treatments such as autologous chondrocyte implantation and microfracture, is limited.
Keywords: stem cells, cell therapy, cartilage, osteoarthritis, cell transplantation
Articular surface injury is a frequent problem, with recovery limited by incomplete natural healing mechanisms complicated by progression to osteoarthritis (OA), which leads to further pain and dysfunction.
This lack of effective healing of chondral defects has led to a need to develop therapies to restore the articular surface to near normal. Broadly, these may be considered as non-cell-based and cell-based.55,63 Cell-based therapies may be further subdivided into non–stem cell therapy or stem cell therapy.
For non–stem cells, the most frequently employed technique is autologous chondrocyte implantation (ACI), with further advancements employing a collagen rather than periosteal cover and third generation approaches utilizing cells seeded within bioscaffolds rather than injection as a cell suspension.11
The isolation of mesenchymal stem cells (MSCs) from a variety of tissues and their promise in in vitro and in animal models has led to their relatively recent implementation in humans.119 This review will focus on localized joint abnormalities such as chondral injury and OA.95
Methods
A search was conducted through PubMed using various combinations of the terms stem cell, cell therapy, cell transplantation, cartilage, chondral, and chondrogenic to December 2013 with no earlier limit. Only 9 comparative studies (Table 1) were identified among a total of 45 human reports of local stem cell therapy for joint disorders (see the Appendix, available at http://sph.sagepub.com/content/suppl).
Table 1.
Study | Cell Type | Level/Design | Number of Patients | Comparison/Controls | Disorder/Grade | Surgical Approach/Method of Stem Cell Implantation | Follow-up | Outcomes |
---|---|---|---|---|---|---|---|---|
Giannini et al (2010),39 Italy | BMC | Level 3 (cohort) | 25 MSC | 10 open ACI, 46 arthroscopic ACI | Talar osteochondral lesions, average 2.18 ± 0.5 cm2 | Arthroscopic: Debridement, platelet gel + collagen powder or HA membrane | 36 months | • In all groups AOFAS improved at 12 and 36 months • No significant difference between groups • Intact cartilage in all cases at arthroscopy • One-step BMC technique less than half the cost of 2-step arthroscopic ACI and less than one third of open |
Kim et al (2013),61 South Korea | SVF | Level 3 (cohort) | 31 MSC injection + surgery | 37 only surgery | Talar osteochondral lesions, 118.9 ± 47.9 mm2 in MSC group, 102.7 ± 31.4 mm2 surgery only | Intra-articular injection—supplement to arthroscopic debridement and microfracture | Mean 21.8 months (range, 12-44 months) | • Significantly greater improvement in MSC group compared with non-MSC for VAS, AOFAS, Roles and Maudsley score and Tegner activity scale at final follow-up |
Koh and Choi (2012),64 South Korea | Infrapatellar fat SVF | Level 4 (case-control) | 25 MSC injection + surgery and PRP | 25 surgery and PRP only | OA—knee, ICRS grade 3.7 ± 0.4 MSC and 2.8 ± 0.8 control | Intra-articular injection of MSC and PRP following arthroscopic debridement. Marrow stimulation procedures not performed | 1 year | • Suggestion of greater benefit from MSC as groups similar at final follow-up, but preoperative clinical scores (VAS, Tegner, Lysholm) and ICRS grade significantly worse for MSC group |
Lee et al (2012),69 Singapore | BM-MSC (culture expanded) | Level 3 (cohort) | 35 group 1 (arthro-scopic surgery + MSC injection) | 35 group 2 (open MSC implantation) | Full-thickness chondral defects—knee | 1: Arthroscopic debridement and microfracture, outpatient injection BM-MSC and HA2: Open debridement, cultured MSC sheet implantation beneath sutured periosteal patch, fibrin glue | 24.5 months | • Both groups significantly improved IKDC, Lysholm, VAS, and SF-36 scores • Injected group more improvement in IKDC and Lysholm scores than open, while improvement in VAS and SF-36 scores were similar |
Nejadnik et al (2010),84 Singapore | BM-MSC (culture expanded) | Level 3 (cohort) | 36 MSC | 36 ACI (periosteal cover) | Chondral defects/OA, ICRS grade III-IV, MSC average 4.6 cm2 (SD 3.53), ACI average 3.6 cm2 (SD 2.84) | Open surgical: debridement, subchondral bone intact, periosteal patch, cells implanted beneath patch, fibrin glue seal | 2 years | • No significant difference in IKDC, Tegner activity, and Lysholm scores • Physical role functioning significantly improved in stem cell group |
Saw et al (2013),102 Malaysia | PBSC | Level 2 (RCT) | 25 PBPC + HA | 25 HA only | Knee—chondral defects, ICRS grade III-IV | Intra-articular injection of PBPC + HA (group 1) or HA alone (group 2) × 8 injections following arthroscopic subchondral drilling | 24 months | • Biopsy at 18 months, 16 patients from each group, better histology PBSC (1066 vs 957) • MRI scores better at 18 months (9.9 vs 8.5) • No significant clinical difference with IKDC scores at 24 months |
Skowroński and Rutka (2013),105 Poland | BMC/PBSC | Level 3 (cohort) | 21 BMC | 25 PBSC | Osteochondral defects medial femoral condyle, >4 cm2, >6 mm deep | Open surgical: BMC or PBSC suspension injected under collagen membrane + fibrin glue following debridement and autologous iliac graft of osseous defect | 5 years | • KOOS, Lysholm, and VAS scales significantly better in PBSC group at 6 months and 1 year • Slight decrease in clinical scores at 5 years in both groups |
Varma et al (2010),117 India | BMC | Level 2 (RCT) | 25 MSC + surgery | 25 surgery only | OA—knee | Intra-articular injection following arthroscopic debridement | 6 months | • Significant improvements in ADLs, sports and recreational activity, and quality of life scores at 6 months MSC compared with controls |
Wakitani et al (2002, 2008),119,122 Japan | BM-MSC (culture expanded) | Level 2 (RCT) | 12 MSC | 12 non-MSC controls | OA—knee, Outerbridge IV, mean 14 × 35 mm | Open surgical: subchondral abrasion and drilling, collagen gel-sheet implant and periosteal cover + high tibial osteotomy | 64 months | • Arthroscopic and histologic scores better in MSC group at 28-95 weeks • No clinical difference then or at 64-month follow-up |
BM-MSC, bone marrow–derived mesenchymal stem cells; ACI, autologous chondrocyte implantation; SVF, stromal vascular fraction; PBSC, peripheral blood stem cells; RCT, randomized controlled trial; BMC, bone marrow concentrate; OA, osteoarthritis; HA, hyaluronic acid; PRP, platelet-rich plasma; SD, standard deviation; ADLs, activities of daily living; ICRS, International Cartilage Repair Society; MFC, medial femoral condyle; VAS, visual analog scale; AOFAS, American Orthopaedic Foot and Ankle Society; KOOS, Knee Injury and Osteoarthritis Outcome Score; IKDC, International Knee Documentation Committee; SF-36, Short Form-36.
All studies utilized autologous cells. BM-MSCs represent culture-expanded cells. Non-BM-MSC studies utilized non–culture expanded cells from a variety of sources. Levels of evidence are as per the Oxford 2011 Levels of Evidence.85
Three randomized trials,102,117,119 3 nonrandomized cohort studies,39,61,84 and 1 case-control study64 compared stem cell with non–stem cell procedures. A further 2 cohort studies compared different stem cells or implantation methods.69,105 Statistical analysis was not performed because of differences in study populations and methods.
Non-Cell-Based Treatment of Chondral Injury
Non-cell-based surgical treatment includes debridement, marrow stimulation by microfracture, abrasion or drilling of the subchondral bone plate, and osteochondral grafting (mosaicplasty).55,63,83
Abnormal cartilage in defects produces detrimental effects on adjacent and opposing cartilage, and debridement can improve symptoms and potentially minimize further chondral loss.28,55 Activation of the innate repair mechanism by injuries involving the subchondral bone plate, as opposed to partial-thickness chondral injury, provides the rationale for marrow stimulation techniques where multiple small holes are placed in the subchondral bone of the defect.55 The mechanism of action is thought to be due to the influx of chondroprogenitor cells.63
Cell-Based Therapy
The first report describing ACI in humans was by Brittberg et al10 in 1994, involving debridement, covering of the defect with a periosteal flap from the proximal medial tibia sutured to surrounding normal cartilage, and cultured chondrocyte injection beneath the periosteal flap.
Autologous chondrocyte implantation utilizes cultured, mature, autologous chondrocytes suspended in an injectable medium with newer variants using a collagen type I/III membrane (ACI-C, CACI, second generation) rather than periosteal cover (ACI-P, first generation).11 Third generation techniques such as matrix-induced autologous chondrocyte implantation (MACI) use cells seeded onto the rough side of a collagen type I/III membrane with a smoother side facing the articular cavity, usually fixed with fibrin glue and sometimes sutures.11
Characterized chondrocyte implantation (CCI) maximizes chondrogenic capacity through a controlled ex vivo process that produces clinically significant improvement with up to 4 years follow-up.115 Comparing CCI with microfracture in a randomized trial, Saris et al98,99 found improved tissue regeneration, although similar clinical outcomes, at 1 year but improved clinical outcome for CCI at 3 years.
These procedures can result in improved clinical, arthroscopic, and histologic features, with hyaline-like cartilage or fibrocartilage present in 43.9% of ACI-C and 36.4% of MACI grafts in 1 prospective, randomized study by Bartlett et al.4 In a randomized trial comparing ACI-P to MACI, Zeifang et al128 found better Lysholm and Gillquist scores at 12 and 24 months in the ACI-P group but no significant difference in International Knee Documentation Committee (IKDC), Tegner Activity Score, or Short Form–36 scores.
In a systematic review of cell-based therapy for chondral lesions from 1994 to 2009, Nakamura et al83 concluded that there was insufficient evidence to indicate superiority of cell-based therapy to non-cell-based treatments with relatively short-term follow-up and most studies demonstrating no convincing differences. Variable results have been obtained comparing ACI with microfracture, with some studies showing no significant difference and others suggesting superiority of ACI.83 Second and third generation techniques offer potential advantages, but longer term follow-up is required.11,83
Basad et al5 demonstrated significantly improved Lysholm, Tegner, patient ICRS (International Cartilage Repair Society), and surgeon ICRS scores with MACI compared with microfracture at 2 years in a randomized study. For patients undergoing ACI after failed microfracture, significantly higher failure rates were observed.91
The chondral defect site as well as level of sports activity and physical training may influence outcome.83 Surgical technique and experience also play a role. Disadvantages of ACI/MACI include healthy cartilage damage at the donor site and lack of suitable donor cartilage in elderly patients with degenerative changes.11,83
Stem Cell Therapy
Sources of Stem Cells
The stem cells with the greatest capacity for differentiation are embryonic stem cells (ESCs). In addition to ethical concerns, questions of safety have arisen because of the risk of teratoma formation.51 These concerns have prompted the search for alternative stem cell sources including adult cells and, more recently, induced pluripotential stem cells (iPSCs), although the teratoma risk currently persists with iPSCs.51,108 IPSCs from osteoarthritic cartilage undergo chondrogenic differentiation in vitro and show chondrogenesis after subcutaneous implantation in mice, but have not yet been used in in vivo articular surface repair.126
Mesenchymal stem cells are multipotential cells originally isolated from bone marrow but naturally existing in many tissues, often around blood vessels. They are defined by the expression of various cell surface molecules (eg, CD73, CD90, CD105), the capacity for self-renewal, and the ability to differentiate into osteogenic, chondrogenic, or adipogenic lineages.48,93 While this capacity already signifies their applicability to musculoskeletal conditions, they also possess potent anti-inflammatory/immunosuppressive properties,79 which may predict efficacy in OA.48,71
Mesenchymal stem cells are being isolated from an increasingly wider variety of human tissues, including bone marrow,93 adipose tissue,130 skeletal muscle,70 synovial membrane26,47 and synovial fluid,47 periosteum,27 peripheral blood,88 umbilical cord blood,124 endometrium,37 amniotic fluid,53 and placenta.54 The potential therapeutic value for MSCs in the treatment of joint disorders is multifactorial, including paracrine effects on regenerating native tissue and immunomodulatory effects.14,48
The cytokine-based immunosuppressive properties of MSCs potentially induce immune tolerance, prompting investigation in multiple sclerosis, foreign graft rejection, and rheumatoid arthritis.48,95 These immunomodulatory effects may help slow the progression of OA by targeting the inflammatory processes in its pathogenesis.79
So far, in the musculoskeletal system, MSCs derived from autologous bone marrow, subcutaneous adipose tissue, infrapatellar fat, and peripheral blood have been utilized in humans in treating osteochondral injury, OA, and rheumatoid arthritis.64,88,101,119
Bone Marrow–Derived Mesenchymal Stem Cells and Bone Marrow Concentrate
Hematopoietic stem cells (HSCs) and bone marrow–MSCs (BM-MSCs) represent different cells lines, with only BM-MSCs used for chondral regeneration. HSCs renew blood elements while MSCs can differentiate into mesenchymal elements, including cartilage.93 Most animal and human stem cell studies for cartilage repair used BM-MSCs. Initial human reports employed culture-expanded BM-MSCs,119 but subsequent publications utilized bone marrow concentrate (BMC) without expansion, allowing a same-day procedure.15,38 BMC contains nucleated cells with a small stem cell component derived from marrow aspirates after removal of most red cells and plasma by centrifugation.38 Both show benefits compared with controls in small, human studies.39,84,117,119,123
Technique-related differences in aspirate yields include site (anterior vs posterior iliac crest) and syringe size.92 Substantial variability also exists for MSC counts between patients.92 For these reasons, comparison between studies or patients within a study is difficult unless the sample is analyzed prior to implantation. Although cell numbers may be counted, characterization with surface markers is required to assess true stem cell counts.18,105 Reported transplanted BM-MSC counts range from 8 million25 to 45.6 million17 cells.
Outcome for femoral head osteonecrosis and tibial nonunion is proportionate to the number of transplanted progenitor cells.49,50 This remains to be shown in humans for cartilage, but the principle of improved healing with greater cell numbers is important. In vitro work shows that increasing initial seeding density of BM-MSC enhances chondrogenesis.52 Government regulation forms a barrier to using culture-expanded cells in some countries, including the United States, as the degree of ex vivo manipulation classifies the treatment in the same manner as a drug.20 Geographic locations of human studies are listed (see the Appendix, available at http://sph.sagepub.com/content/suppl). The US Food and Drug Administration has currently not approved any stem cell products for use in the United States other than cord blood–derived hematopoietic stem cells for certain indications.114 While the use of culture-expanded cells is prohibited, some clinics offer same day procedures using minimally manipulated cells such as BMC, concentrated at the point of care.20 The issue of stem cell regulation continues to be a subject of active debate.
An apparent disadvantage of BM-MSCs is that cell numbers diminish with age and exhibit reduced proliferative capacity and increased rates of apoptosis compared with BM-MSC from younger patients.106
Adipose-Derived Stem Cells and Stromal Vascular Fraction
Mesenchymal stem cells in adipose tissue arise from or form perivascular cells.13,130,131 Adipose tissue contains proportionally higher numbers of MSCs (approximately 10% of nucleated cells) than bone marrow and is amenable to liposuction without significant morbidity. In contrast to BM-MSCs, numbers do not decline with age but do decline with obesity.3 Stem cells may differ in numbers from abdominal adipose tissue compared with the hip or thigh, but proliferation and differentiation do not appear influenced by harvest site.56
As with bone marrow, adipose stem cells may be utilized in 2 major forms. Stromal vascular fraction (SVF) is a heterogeneous population of cells that may contain MSCs, fibroblasts, endothelial cells, leukocytes (lymphocytes and macrophages), and pericytes.8 Stem cells from SVF may be separated and expanded in vitro (adipose-derived stem cells [ADSCs]).
An advantage of SVF and BMC is elimination of the time lag between harvest and implantation, minimizing exposure to risks, reducing cost and logistical difficulties.39,88,95 However, the lack of cellular content identification of SVF is a major problem in evaluating clinical efficacy and patient responses. SVF contains large numbers of T regulatory (Treg) cells that may assist in immunosuppression and tolerance induction.95
Intra-articular SVF has been successfully utilized in dogs with elbow or hip OA but with suboptimal results in horses.8,9,35 Co-administration of non-infrapatellar fat-derived SVF showed superior clinical results at mean 21.8-month follow-up compared with non-MSC controls in a human study.61
Improved cartilage repair was seen with culture-expanded ADSCs compared with controls in rabbit full-thickness chondral defects with better integration and more hyaline cartilage formation, but their use in humans has not been reported.31
Infrapatellar Fat Pad–Derived Stem Cells
Infrapatellar fat differs in composition to subcutaneous adipose tissue, containing a large amount of collagenous tissue and possibly synoviocytes.127 While exhibiting characteristics of ADSCs,57 they have more similarities with fibrous synovium-derived cells than subcutaneous fat–derived cells, and possibly greater chondrogenic potential.78 In rabbits, cells cultured from infrapatellar fat showed promising results compared with controls.110
Infrapatellar SVF (not culture-expanded) has shown similar clinical findings in humans at 1 year compared with MSC free controls undergoing arthroscopic debridement, implying a potential benefit from MSC because of poorer preoperative clinical scores and ICRS grades.64
Peripheral Blood Mesenchymal Stem Cells/Progenitor Cells
Peripheral blood presents another source of MSCs, obtained with relative ease and no significant donor site morbidity.21 MSCs derived from human peripheral blood cells (PBSCs) exhibit similar in vitro chondrogenic potential to BM-MSCs, although they are far less concentrated in blood than in marrow.23 Use of granulocyte–colony stimulating factor (G-CSF) increases MSC numbers in peripheral blood.21,101 While generally well tolerated, rare risks of G-CSF in healthy donors include splenic rupture and adult respiratory distress syndrome, although the theoretical risk of hematologic malignancy remains to be proven in the healthy donor population.32
Following an initial pilot report,101 PBSCs have been assessed in a randomized study augmenting arthroscopic subchondral drilling with postprocedural injections of either PBSC and hyaluronic acid (HA) or HA alone, reporting improved histologic and MRI scores at 18 months but no significant clinical difference at 24 months.102
Other stem cell sources trialed in animals, but not humans, include periosteum, synovium, and skeletal muscle.73,76,90,118
Methods of Stem Cell Transplantation
Open Surgical Implantation of MSCs
Surgical implantation may be similar to ACI, with MSCs beneath a periosteal119 or collagen43 cover instead of cultured chondrocytes.45,60,67,74,84,103,104,109,119-123 A cell-seeded construct (analogous to MACI) may be used rather than suspended cells.11 An ideal scaffold is nontoxic, absorbable, mechanically sound, and promotes cell growth.46
Wakitani et al118 found that osteochondral progenitor cells from bone marrow or periosteum in type I collagen gel produced superior repair of full-thickness rabbit medial femoral condylar defects compared with empty defects or a cell-free collagen gel with hyaline cartilage formation and mechanically superior repair tissue. Macroscopic appearance at 24 weeks and histologic appearance at 12 and 24 weeks was less favorable than at 4 weeks postimplantation.118
Wakitani et al119 used culture-expanded BM-MSCs in collagen gel in medial femoral condylar defects of humans with OA at the time of high tibial osteotomy, with 12 patients randomized to each group. Subchondral abrasion was performed to facilitate bleeding. A BM-MSC–collagen gel sheet composite was applied to the defect and covered with autologous periosteum. The control group received the same treatment without BM-MSCs. BM-MSC patients demonstrated improved arthroscopic and histologic scores 28 to 95 weeks following treatment, with no clinical difference, including on repeat assessment at 64 months.119,123
Gobbi et al43 applied a 1-step open approach with BMC (nonexpanded) following debridement of knee chondral lesions in 15 patients using a collagen membrane cover. Most underwent associated procedures. Significant clinical improvement was noted at 6, 12, and 24 months (visual analog scale [VAS], Knee Injury and Osteoarthritis Outcome Score [KOOS], IKDC, SF-36, Tegner, Marx, Lysholm). Defect filling at MRI was complete in 12 cases and incomplete in 3 cases. Arthroscopic evaluation in 4 knees was normal to nearly normal, and histology in 3 patients showed hyaline-like features.43
Skowroński and Rutka105 showed significantly better KOOS, Lysholm, and VAS scores for PBSC over nonexpanded BMC in conjunction with autologous iliac bone grafting of medial femoral condylar osteochondral lesions at 6 months and 1 year, but commented that it could reflect double the transplanted cell numbers compared with the BMC group, with possible contribution from more stem cells provided by marrow stimulation in the G-CSF–treated PBSC group.
Regarding open foot and ankle chondral defect repairs, implantation of nonexpanded BMC-impregnated collagen matrix was reported by Richter and Zech94 in 25 patients who were followed up with at 2 years with significant improvements in VAS foot and ankle scores.
Arthroscopic Implantation of Stem Cells
Giannini et al38,40 followed 49 patients, aged 14 to 50 years, for 4 years after 1-step arthroscopic implantation of nonexpanded BMC for talar osteochondral lesions, with either collagen powder/platelet gel or HA membrane/platelet gel scaffolds. American Orthopaedic Foot and Ankle Society (AOFAS) scores improved, with best results at 24 months, but deteriorating at 36 and 48 months.
Stem Cell Versus Autologous Chondrocyte Implantation
Few studies directly compare stem cell treatment to ACI. Adachi et al1 showed similar results of B-galactosidase gene–transfected muscle-derived stem cells (MDSCs) with similarly transfected chondrocytes in rabbits. Testing a gellan gum hydrogel in rabbits, Oliveira et al86 noted improved hyaline cartilage formation with chondrogenically predifferentiated ADSCs compared with chondrocytes, but similar results between undifferentiated ADSCs and chondrocytes.
Autologous, cultured BM-MSCs were compared with matrix-associated autologous chondrocyte transplantation in sheep, both suspended in collagen gel, with superior results for BM-MSCs at 1 year, particularly regarding integration with adjacent native cartilage.72
Arthroscopic, nonexpanded BMC implantation was retrospectively compared with open field and arthroscopic ACI.39 Significant clinical improvement was found in all 3 groups at 36 months, with no significant difference between them. Second-look arthroscopies demonstrated intact cartilage in all cases, with components of hyaline cartilage at biopsy. The 1-step arthroscopic BMC technique was less than half the cost of the 2-step arthroscopic ACI technique.39
Nejadnik et al84 matched 36 patients undergoing ACI-P with 36 patients receiving autologous, culture-expanded BM-MSCs, with cultured chondrocytes or BM-MSCs implanted beneath a sutured periosteal patch in similar techniques. No significant difference was shown between the 2 groups in terms of clinical outcome up to 24 months except for physical role functioning, which was better improved for the BM-MSC group. The BM-MSC therapy required only 1 surgery, reduced costs, and caused less donor site morbidity. In contrast to the ACI group, an age-related response was not evident with BM-MSCs.84
Intra-Articular Injection of MSCs
Rationale
Intra-articular injection holds several potential advantages, including reduced recovery time and less cost.15,25,33,58,64,101 Same day intra-articular administration of cells surgically obtained from the infrapatellar fat pad has been used to augment arthroscopic debridement.64 From a therapeutic perspective, intra-articular injection may be better matched to the pathogenesis of OA,79,100 although it may increase the risk of synovial proliferation.63
Cartilage Defects
In animal studies involving surgically created injuries to anterior cruciate ligaments, menisci, and articular cartilage, intra-articularly administered labeled BM-MSCs migrated to sites of injury.68,80,81 To enhance migration to the desired location, Kobayashi et al62 utilized an external magnetic force to direct magnetically labeled, intra-articularly injected BM-MSCs to experimentally created osteochondral defects in rabbit and swine patellae. In a further laboratory study, the magnetic force improved cell adhesion with no deleterious effects on cell proliferation for up to 3 weeks.82
Osteoarthritis
The anti-inflammatory and immunomodulatory effects of MSCs may retard the progression of OA. Intra-articular injections of stem cells slowed progression of surgically induced OA in goats following a single intra-articular dose of cultured BM-MSCs,81 in rabbits using infrapatellar fat pad–derived MSCs,110 and in rats using MDSCs with transduced genes.73 BM-MSCs may also prevent the onset of posttraumatic OA in mice when injected at the same time as experimentally created closed tibial plateau fracture.30
Studies of animals with spontaneous OA, as well as experimental OA, have also reported improvement following MSC injection.8,9,44,100 Cell labeling shows incorporation into damaged cartilage and partial cartilage regeneration in guinea pigs using cultured human BM-MSCs.100 Black et al8 reported significant clinical improvement in dogs with spontaneous OA of the coxofemoral joint following intra-articular SVF compared with placebo in a randomized, double-blinded, placebo-controlled trial. Improvements were also noted following a single humeroradial SVF injection for dogs with elbow OA, although with no control group in this study.9 Guercio et al44 found improved clinical benefit following ADSC injection in 4 dogs with lameness due humeroradial OA that had previously failed to respond to anti-inflammatory drugs. While most investigations appear to focus on restoration of articular cartilage, stem cell therapy may also benefit meniscal defects in animals and humans.17,81
Orozco et al87 followed 12 patients receiving intra-articular, autologous-expanded BM-MSCs (40 × 106 cells) for 1 year, demonstrating significantly improved VAS, Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), and Lequesne scores, with no significant difference in SF-36 and reduction in pain occurring within 3 months. Decreased poor quality cartilage on T2 mapping was seen in 11 patients. No human studies have yet compared MSC injection with other treatments in the absence of concurrent surgery.
Augmenting Surgery
Mesenchymal stem cell injection has been utilized as an adjunct to surgical techniques in humans64,69,101 and animals.75,107 In goats, BM-MSCs in alginate applied between osteochondral plugs during mosaicplasty was superior at 24 weeks compared with mosaicplasty alone, and better still using transforming growth factor-β1–transduced BM-MSCs.107 Comparing intra-articular BM-MSCs and HA to intra-articular HA alone following microfracture of full-thickness chondral defects in horses, McIlwraith et al75 noted significantly increased firmness and a non-significant trend for better overall repair quality with BM-MSCs.
The location of the infrapatellar fat pad makes it an attractive harvest target. Koh and Choi64 utilized intra-articular injection of nonexpanded infrapatellar fat cells combined with arthroscopic debridement and PRP in humans with knee OA, with similar 1-year clinical findings compared with controls receiving only PRP post-debridement but significantly worse preoperative clinical (Tegner, Lysholm, and VAS) scores and ICRS grades in the MSC group, favoring a benefit from MSC injection. Of the 25 MSC patients, 18 were reassessed at 2 years, with significantly improved clinical features (WOMAC, Lysholm, and VAS scores) as well as MRI scores compared with preoperative, including significant clinical improvement in patients with grade 3 compared with grade 4 OA.65 The fat pad was acquired at surgery, but the 3- to 4-hour processing necessitated a separate procedure that day.64
Kim et al61 used non-expanded buttock adipose cells (SVF) as an intra-articular supplement to arthroscopic debridement and microfracture of talar osteochondral lesions. Significantly better clinical scores were obtained with MSC (31 ankles) compared with arthroscopic surgery alone (37 ankles).
In a randomized trial, Varma et al117 compared 25 patients with mild to moderate knee OA undergoing arthroscopic debridement alone with 25 patients undergoing arthroscopic debridement followed by intra-articular, nonexpanded BMC injection. Significant improvements in activities of daily living, sports and recreational activity, and quality of life scores were seen at 6 months.117
Saw et al102 randomized 50 patients with ICRS grade 3-4 chondral defects undergoing arthroscopic debridement and subchondral drilling to a series of 8 injections of either non-expanded PBSC and HA or HA alone. Significantly better histologic scores (1066 vs 957) and MRI scores (9.9 vs 8.5) were reported at 18 months, with blinding of the reporting radiologist and pathologist, although no significant difference in IKDC scores at 24 months (74.8 vs 71.1).102
Lee et al69 compared 35 knee full-thickness chondral defects undergoing arthroscopic debridement and microfracture, followed by outpatient injection of culture-expanded BM-MSC and HA, with 35 matched patients receiving open implantation of BM-MSC sheets beneath a sutured periosteal cover. Both groups showed significantly improved IKDC, Lysholm, VAS, and SF-36 scores at up to 2 years. The arthroscopic-injected group experienced more improvement in IKDC and Lysholm scores compared with the open group but similar improvement in VAS and SF-36 scores. MRI at 1 year showed good defect filling and integration.69
Growth Factors, Platelet-Rich Plasma, Gene Therapy, and Hyaluronic ACID
Platelet-rich plasma (PRP) is a source of autologous growth factors and an effective treatment for elbow tendinopathy.24 A systematic review of intra-articular PRP injection for cartilage repair has shown safety in humans with potential pain reduction and improved function.36 Longer term follow-up is required before it can be recommended for OA therapy.36 After chemical induction of OA in rat knee joints, Mifune et al76 compared MDSCs expressing bone morphogenetic protein 4 (BMP-4) and sFlt-1 with and without PRP. Improved articular cartilage repair was seen at 4 and 12 weeks with the addition of PRP.
Hyaluronic acid, a glycosaminoglycan extracellular matrix constituent, has been used for human OA with MRI evaluation up to 24 months showing beneficial effects on cartilage preservation.125 Multiple animal studies have shown the combined use of stem cells and HA to produce better results than HA alone.68,75,79,81
Following in vitro expansion, stem cells may be induced via transforming growth factor-β1 or BMP-2 to undergo chondrogenic differentiation22,72 or can be uninduced.93,129 Encouraging results have been achieved with both approaches compared with controls.22,129 Comparing induced with uninduced cells in animal studies shows mixed results.22,72
Safety of Mesenchymal Stem Cell–Related Procedures
In vitro manipulation creates the opportunity for infection, necessitating antibiotic administration above the minimum inhibitory concentration for relevant organisms while not impeding MSC proliferation and differentiation.59
Malignancy has been flagged as a potential risk of MSC implantation but has not yet been shown in clinical practice.18,19,123 Miura et al77 found that fibrosarcoma developed from murine BM-MSCs after numerous in vitro passages. Tolar et al111 also identified sarcomatous transformation from mouse BM-MSCs expanded in vitro.
In 2005, Rubio et al97 reported that after long-term in vitro culture of 4 to 5 months, human ADSCs exhibited malignant transformation. The group retracted this article in 2010, unable to reproduce the findings, proposing potential cross-contamination.29 Another group described spontaneous transformation of BM-MSCs due to cross-contamination by immortalized cell lines, emphasizing the need for DNA fingerprinting.96,112
Bernardo et al7 found that human BM-MSCs did not demonstrate malignant transformation after long-term culture, showing telomeric shortening with progressive decline in proliferation until reaching senescence.
Discussion
Cell therapy represents promising treatment for many conditions, including joint disorders. The most widely practiced form, ACI and its newer variants, is capable of promoting cartilage repair and providing clinical benefit, although there is insufficient evidence to recommend these procedures over marrow stimulation techniques and osteochondral grafting.83 Only limited human data exist for use of MSCs, but both surgical implantation and intra-articular injection appear to be safe and exhibit reasonable efficacy. There is currently a paucity of randomized human trials.
Cell sources that do not require in vitro expansion, such as BMC or SVF, provide the opportunity for same day therapy by reducing the turnaround time from cell harvest to treatment.38,64 Intra-articular injection offers a reduction in postoperative recovery time.8,64,117 For chondral injury, MSC therapy may improve symptom control through anti-inflammatory and immunomodulatory effects.19
Conclusion
At present, there is no conclusive evidence to recommend cell therapy over non-cell-based procedures, but both treatments appear to offer beneficial results. Non–stem cell therapy such as ACI, mosaicplasty, and microfracture at present possesses more clinical evidence than MSC treatments.
Footnotes
The following authors declared potential conflicts of interest: Daniel Bates, BSc (Hons), BMed, is a consultant for Bio Stem Cell, Hong Kong and has stock/stock options with Prostem and Cell Medicine Australia; Richard Boyd, BSc (Hons), PhD, is a board member of and has stock options with Magellan.
References
- 1. Adachi N, Sato K, Usas A, et al. Muscle derived, cell based ex vivo gene therapy for treatment of full thickness articular cartilage defects. J Rheumatol. 2002;29:1920-1930. [PubMed] [Google Scholar]
- 2. Adachi N, Ochi M, Deie M, Ito Y. Transplant of mesenchymal stem cells and hydroxyapatite ceramics to treat severe osteochondral damage after septic arthritis of the knee. J Rheumatol. 2005;32:1615-1618. [PubMed] [Google Scholar]
- 3. Aust L, Devlin B, Foster SJ, et al. Yield of human adipose-derived stem cells from liposuction aspirates. Cytotherapy. 2004;6:7-14. [DOI] [PubMed] [Google Scholar]
- 4. Bartlett W, Skinner JA, Gooding CR, et al. Autologous chondrocyte implantation versus matrix-induced autologous chondrocyte implantation for osteochondral defects of the knee. J Bone Joint Surg Br. 2005;87:640-645. [DOI] [PubMed] [Google Scholar]
- 5. Basad E, Ishaque B, Bachmann G, Stürz H, Steinmeyer J. Matrix-induced autologous chondrocyte implantation versus microfracture in the treatment of cartilage defects of the knee: a 2-year randomised study. Knee Surg Sports Traumatol Arthrosc. 2010;18:519-527. [DOI] [PubMed] [Google Scholar]
- 6. Battaglia M, Rimondi E, Monti C, et al. Validity of T2 mapping in characterization of the regeneration tissue by bone marrow derived cell transplantation in osteochondral lesions of the ankle. Eur J Radiol. 2011;80:e132-e139. [DOI] [PubMed] [Google Scholar]
- 7. Bernardo ME, Zaffaroni N, Novara F, et al. Human bone marrow–derived mesenchymal stem cells do not undergo transformation after long-term in vitro culture and do not exhibit telomere maintenance mechanisms. Cancer Res. 2007;67:9142-9149. [DOI] [PubMed] [Google Scholar]
- 8. Black LL, Gaynor J, Gahring D, et al. Effect of adipose-derived mesenchymal stem and regenerative cells on lameness in dogs with chronic osteoarthritis of the coxofemoral joints: a randomized, double-blinded, multicentre, controlled trial. Vet Ther. 2007;8:272-284. [PubMed] [Google Scholar]
- 9. Black LL, Gaynor J, Adams C, et al. Effect of intraarticular injection of autologous adipose-derived mesenchymal stem and regenerative cells on clinical signs of chronic osteoarthritis of the elbow joint in dogs. Vet Ther. 2008;9:192-200. [PubMed] [Google Scholar]
- 10. Brittberg M, Lindahl A, Nilsson A, Ohlsson C, Isaksson O, Peterson L. Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. N Eng J Med. 1994;331:889-895. [DOI] [PubMed] [Google Scholar]
- 11. 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;38:1259-1271. [DOI] [PubMed] [Google Scholar]
- 12. 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] [PubMed] [Google Scholar]
- 13. Cai X, Lin Y, Hauschka PV, Grottkau BE. Adipose stem cells originate from perivascular cells. Biol Cell. 2011;103:435-447. [DOI] [PubMed] [Google Scholar]
- 14. Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. J Cell Biochem. 2006;98:1076-1084. [DOI] [PubMed] [Google Scholar]
- 15. Centeno CJ, Kisiday J, Freeman M, Schultz JR. Partial regeneration of the human hip via autologous bone marrow nucleated cell transfer: a case study. Pain Physician. 2006;9:253-256. [PubMed] [Google Scholar]
- 16. 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. 2008;11:343-353. [PubMed] [Google Scholar]
- 17. Centeno CJ, Busse D, Kisiday J, Keohan C, Freeman M, Karli D. Regeneration of meniscus cartilage in a knee treated with percutaneously implanted autologous mesenchymal stem cells. Med Hypotheses. 2008;71:900-908. [DOI] [PubMed] [Google Scholar]
- 18. Centeno CJ, Schultz JR, Cheever M, Robinson B, Freeman M, Marasco W. Safety and complications reporting on the re-implantation of culture-expanded mesenchymal stem cells using autologous platelet lysate technique. Curr Stem Cell Res Ther. 2010;5:81-93. [DOI] [PubMed] [Google Scholar]
- 19. Centeno CJ, Schultz JR, Cheever M, et al. Safety and complications reporting update on the re-implantation of culture-expanded mesenchymal stem cells using autologous platelet lysate technique. Curr Stem Cell Res Ther. 2011;6:368-378. [DOI] [PubMed] [Google Scholar]
- 20. Center for Biologics Evaluation and Research, Food and Drug Administration, US Department of Health and Human Services. Guidance for industry regulation of human cells, tissues, and cellular and tissue-based products (HCT/Ps): small entity compliance guide. August 2007. http://permanent.access.gpo.gov/LPS112358/LPS112358_ucm062592.pdf Accessed January 29, 2014.
- 21. Cesselli D, Beltrami AP, Rigo S, et al. Multipotent progenitor cells are present in human peripheral blood. Circ Res. 2009;104:1225-1234. [DOI] [PubMed] [Google Scholar]
- 22. Chang CH, Kuo TF, Lin FH, et al. Tissue engineering-based cartilage repair with mesenchymal stem cells in a porcine model. J Orthop Res. 2011;29:1874-1880. [DOI] [PubMed] [Google Scholar]
- 23. Chong PP, Selvaratnam L, Abbas AA, Kamarul T. Human peripheral blood derived mesenchymal stem cells demonstrate similar characteristics and chondrogenic differentiation potential to bone marrow derived mesenchymal stem cells. J Orthop Res. 2012;30:634-642. [DOI] [PubMed] [Google Scholar]
- 24. Creaney L, Wallace A, Curtis M, Connell D. Growth factor-based therapies provide additional benefit beyond physical therapy in resistant elbow tendinopathy: a prospective, single-blind, randomised trial of autologous blood injections versus platelet-rich plasma injections. Br J Sports Med. 2011;45:966-971. [DOI] [PubMed] [Google Scholar]
- 25. Davatchi F, Abdollahi BS, Mohyeddin M, Shahram F, Nikbin B. Mesenchymal stem cell therapy for knee osteoarthritis. Preliminary report of four patients. Int J Rheum Dis. 2011;14:211-215. [DOI] [PubMed] [Google Scholar]
- 26. De Bari C, Dell’Accio F, Luyten FP. Failure of in vitro–differentiated mesenchymal stem cells from the synovial membrane to form ectopic stable cartilage in vivo. Arthritis Rheum. 2004;50:142-150. [DOI] [PubMed] [Google Scholar]
- 27. De Bari C, Dell’Accio F, Vanlauwe J, et al. Mesenchymal multipotency of adult human periosteal cells demonstrated by single-cell lineage analysis. Arthritis Rheum. 2006;54:1209-1221. [DOI] [PubMed] [Google Scholar]
- 28. Dean DD, Martel-Pelletier J, Pelletier JP, Howell DS, Woessner JF., Jr. Evidence for metalloproteinase and metalloproteinase inhibitor imbalance in human osteoarthritic cartilage. J Clin Invest. 1989;84:678-685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. de la, Fuente R, Bernad A, Garcia-Castro J, Martin MC, Ciqudosa JC. Retraction: spontaneous human adult stem cell transformation. Cancer Res. 2010;70:6682. [DOI] [PubMed] [Google Scholar]
- 30. Diekman BO, Wu CL, Louer CR, et al. Intra-articular delivery of purified mesenchymal stem cells from C57BL/6 or MRL/MpJ superhealer mice prevents post-traumatic arthritis. Cell Transplant. 2013;22:1395-1408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Dragoo JL, Carlson G, McCormick F, et al. Healing full-thickness cartilage defects using adipose-derived stem cells. Tissue Eng. 2007;13:1615-1621. [DOI] [PubMed] [Google Scholar]
- 32. D’Souza A, Jaiyesimi I, Trainor L, Venuturumili P. Granulocyte colony-stimulating factor administration: adverse events. Transfus Med Rev. 2008;22:280-290. [DOI] [PubMed] [Google Scholar]
- 33. 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;15:422-428. [PubMed] [Google Scholar]
- 34. Enea D, Cecconi S, Calcagno S, et al. Single-stage cartilage repair in the knee with microfracture covered with a resorbable polymer-based matrix and autologous bone marrow concentrate. Knee. 2013;20:562-569. [DOI] [PubMed] [Google Scholar]
- 35. Frisbie DD, Kisiday JD, Kawcak CE, Werpy NM, McIlwraith CW. Evaluation of adipose-derived stromal vascular fraction or bone marrow–derived mesenchymal stem cells for treatment of osteoarthritis. J Orthop Res. 2009;27:1675-1680. [DOI] [PubMed] [Google Scholar]
- 36. Frizziero A, Giannotti E, Ferraro C, Masiero S. Platelet rich plasma intra-articular injections: a new therapeutic strategy for the treatment of knee osteoarthritis in sport rehabilitation. A systematic review. Sport Sci Health. 2012;8:15-22. [Google Scholar]
- 37. Gargett CE, Masuda H. Adult stem cells in the endometrium. Mol Hum Reprod. 2010;16:818-834. [DOI] [PubMed] [Google Scholar]
- 38. Giannini S, Buda R, Vannini F, Cavallo M, Grigolo B. One-step bone marrow–derived cell transplantation in talar osteochondral lesions. Clin Orthop Relat Res. 2009;467:3307-3320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. 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;41:1196-1203. [DOI] [PubMed] [Google Scholar]
- 40. Giannini S, Buda R, Battaglia M, et al. One-step repair in talar osteochondral lesions. 4-year clinical results and T2-mapping capability in outcome prediction. Am J Sports Med. 2013;41:511-518. [DOI] [PubMed] [Google Scholar]
- 41. Gigante A, Calcagno S, Cecconi S, Ramazzotti D, Manzotti S, Enea D. Use of collagen scaffold and autologous bone marrow concentrate as a one-step cartilage repair in the knee: histological results of second-look biopsies at 1 year follow-up. Int J Immunopathol Pharmacol. 2011;24(suppl 2):69-72. [DOI] [PubMed] [Google Scholar]
- 42. Gigante A, Cecconi S, Calcagno S, Busilacchi A, Enea D. Arthroscopic knee cartilage repair with covered microfracture and bone marrow concentrate. Arthrosc Tech. 2012;1:e175-e180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Gobbi A, Karnatzikos G, Scotti C, Mahajan V, Mazzucco L, Grigolo B. One-step cartilage repair with bone marrow aspirate concentrated cells and collagen matrix in full-thickness knee cartilage lesions: results at 2-year follow-up. Cartilage. 2011;2:286-299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Guercio A, Di Marco P, Casella S, et al. Production of canine mesenchymal stem cells from adipose tissue and their application in dogs with chronic osteoarthritis of the humeroradial joints. Cell Biol Int. 2012:36:189-194. [DOI] [PubMed] [Google Scholar]
- 45. Haleem AM, 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;1:253-261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Han Y, Wei Y, Wang S, Song Y. Cartilage regeneration using adipose-derived stem cells and the controlled-release hybrid microspheres. Joint Bone Spine. 2010;77:27-31. [DOI] [PubMed] [Google Scholar]
- 47. Harvanová D, Táthová T, Sarišsky M, Amrichová J, Rosocha J. Isolation and characterization of synovial mesenchymal stem cells. Folia Biol (Praha). 2011;57:119-124. [PubMed] [Google Scholar]
- 48. Heng TS, Dudakov JA, Khong DM, Chidgey AP, Boyd RL. Stem cells—meet immunity. J Mol Med (Berl). 2009;87:1061-1069. [DOI] [PubMed] [Google Scholar]
- 49. Hernigou P, Beaujean F. Treatment of osteonecrosis with autologous bone marrow grafting. Clin Orthop Relat Res. 2002;(405):14-23. [DOI] [PubMed] [Google Scholar]
- 50. Hernigou P, Poignard A, Beaujean F, Rouard H. Percutaneous autologous bone-marrow grafting for nonunions. Influence of the number and concentration of progenitor cells. J Bone Joint Surg Am. 2005;87:1430-1437. [DOI] [PubMed] [Google Scholar]
- 51. Ho PJ, Yen ML, Yet SF, Yen BL. Current applications of human pluripotent stem cells: possibilities and challenges. Cell Transplant. 2012;21:801-814. [DOI] [PubMed] [Google Scholar]
- 52. Huang CY, 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;278:428-436. [DOI] [PubMed] [Google Scholar]
- 53. in ‘t Anker PS, Scherjon SA, Kleijburg-van der Keur, et al. Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood. 2003;102:1548-1549. [DOI] [PubMed] [Google Scholar]
- 54. in ‘t Anker PS, Scherjon SA, Kleijburg-van der, Keur C, et al. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells. 2004;22:1338-1345. [DOI] [PubMed] [Google Scholar]
- 55. Jaiswal PK, Wong K, Khan WS. Operative treatment of knee cartilage injuries: a review of the current literature on non-cell-based and cell-based therapies. Br J Med Med Res. 2011;1:516-537. [Google Scholar]
- 56. Jurgens WJ, Oedayrajsingh-Varma MJ, Helder MN, et al. Effect of tissue-harvesting site on yield of stem cells derived from adipose tissue: implications for cell-based therapies. Cell Tissue Res. 2008;332:415-426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Jurgens WJ, van Dijk A, Doulabi BZ, et al. Freshly isolated stromal cells from the infrapatellar fat pad are suitable for a one-step surgical procedure to regenerate cartilage tissue. Cytotherapy. 2009;11:1052-1064. [DOI] [PubMed] [Google Scholar]
- 58. Jurgens WJ, Kroeze RJ, Bank RA, Ritt MJ, Helder MN. Rapid attachments of adipose stromal cells on resorbable polymeric scaffolds facilitates the one-step surgical procedure for cartilage and bone tissue engineering purposes. J Orthop Res. 2011;29:853-860. [DOI] [PubMed] [Google Scholar]
- 59. Kagiwada H, Fukuchi T, Machida H, Yamashita K, Ohgushi H. Effect of gentamicin on growth and differentiation of human mesenchymal stem cells. J Toxicol Pathol. 2008;21:61-67. [Google Scholar]
- 60. Kasemkijwattana C, Hongeng S, Kesprayura S, Rungsinaporn V, Chaipinyo K, Chansiri K. Autologous bone marrow mesenchymal stem cells implantation for cartilage defects: two case reports. J Med Assoc Thai. 2011;94:395-400. [PubMed] [Google Scholar]
- 61. Kim YS, Park EH, Kim YC, Koh YG. Clinical outcomes of mesenchymal stem cell injection with arthroscopic treatment in older patients with osteochondral lesions of the talus. Am J Sports Med. 2013;41:1090-1099. [DOI] [PubMed] [Google Scholar]
- 62. Kobayashi T, Ochi M, Yanada S, et al. A novel cell delivery system using magnetically labelled mesenchymal stem cells and an external magnetic device for clinical cartilage repair. Arthroscopy. 2008;24:69-76. [DOI] [PubMed] [Google Scholar]
- 63. Koga H, Engebretsen L, Brinchmann JE, Muneta T, Sekiya I. Mesenchymal stem cell-based therapy for cartilage repair: a review. Knee Surg Sports Traumatol Arthrosc. 2009;17:1289-1297. [DOI] [PubMed] [Google Scholar]
- 64. Koh YG, Choi YJ. Infrapatellar fat pad–derived mesenchymal stem cell therapy for knee osteoarthritis. Knee. 2012;19:902-907. [DOI] [PubMed] [Google Scholar]
- 65. Koh YG, Jo SB, Kwon OR, et al. Mesenchymal stem cell injections improve symptoms of knee osteoarthritis. Arthroscopy. 2013;29:748-755. [DOI] [PubMed] [Google Scholar]
- 66. Kon E, Vannini F, Buda R, et al. How to treat osteochondritis dissecans of the knee: surgical techniques and new trends. J Bone Joint Surg Am. 2012;94:e1(1-8). [DOI] [PubMed] [Google Scholar]
- 67. Kuroda R, Ishida K, Matsumoto T, et al. Treatment of a full-thickness articular cartilage defect in the femoral condyle of an athlete with autologous bone-marrow stromal cells. Osteoarthritis Cartilage. 2007;15:226-231. [DOI] [PubMed] [Google Scholar]
- 68. Lee KB, Hui JH, Song IC, Ardany L, Lee EH. Injectable mesenchymal stem cell therapy for large cartilage defects—a porcine model. Stem Cells. 2007;25:2964-2971. [DOI] [PubMed] [Google Scholar]
- 69. Lee KB, Wang VT, Chan YH, Hui JH. 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;41:511-517. [PubMed] [Google Scholar]
- 70. Lu SH, Yang AH, Wei CF, Chiang HS, Chancellor MB. Multi-potent differentiation of human purified muscle-derived cells: potential for tissue regeneration. BJU Int. 2010;105:1174-1180. [DOI] [PubMed] [Google Scholar]
- 71. Manferdini C, Maumus M, Gabusi E, et al. Adipose-derived mesenchymal stem cells exert antiinflammatory effects on chondrocytes and synoviocytes from osteoarthritis patients through prostaglandin E2. Arthritis Rheum. 2013;65:1271-1281. [DOI] [PubMed] [Google Scholar]
- 72. 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;39:1401-1412. [DOI] [PubMed] [Google Scholar]
- 73. Matsumoto T, Cooper GM, Gharaibeh B, et al. Cartilage repair in a rat model of osteoarthritis through intraarticular transplantation of muscle-derived stem cells expressing bone morphogenetic protein 4 and soluble Flt-1. Arthritis Rheum. 2009;60:1390-1405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Matsumoto T, Okabe T, Ikawa T, et al. Articular cartilage repair with autologous bone marrow mesenchymal cells. J Cell Physiol. 2010;225:291-295. [DOI] [PubMed] [Google Scholar]
- 75. McIlwraith CW, Frisbie DD, Rodkey WG, et al. Evaluation of intra-articular mesenchymal stem cells to augment healing of microfractured chondral defects. Arthroscopy. 2011;27:1552-1561. [DOI] [PubMed] [Google Scholar]
- 76. Mifune Y, Matsumoto T, Takayama K, et al. The effect of platelet-rich plasma on the regenerative therapy of muscle derived stem cells for articular cartilage repair. Osteoarthritis Cartilage. 2013;21:175-185. [DOI] [PubMed] [Google Scholar]
- 77. Miura M, Miura Y, Padilla-Nash HM, et al. Accumulated chromosomal instability in murine bone marrow mesenchymal stem cells leads to malignant transformation. Stem Cells. 2006;24:1095-1103. [DOI] [PubMed] [Google Scholar]
- 78. Mochizuki T, Muneta T, Sakaguchi Y, et al. Higher chondrocyte potential of fibrous synovium– and adipose synovium–derived cells compared with subcutaneous fat-derived cells: distinguishing properties of mesenchymal stem cells in humans. Arthritis Rheum. 2006;54:843-853. [DOI] [PubMed] [Google Scholar]
- 79. Mokbel AN, El-Tookhy OS, Shamaa AA, Rashed LA, Sabry D, El Sayed AM. Homing and reparative effect of intra-articular injection of autologous mesenchymal stem cells in osteoarthritic animal model. BMC Musculoskelet Disord. 2011;12:259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. 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. 2011;29:275-284. [PubMed] [Google Scholar]
- 81. Murphy JM, Fink DJ, Hunziker EB, Barry FP. Stem cell therapy in a caprine model of osteoarthritis. Arthritis Rheum. 2003;48:3464-3474. [DOI] [PubMed] [Google Scholar]
- 82. Nakamae T, Adachi N, Kobayashi T, et al. The effect of an external magnetic force on cell adhesion and proliferation of magnetically labelled mesenchymal stem cells. Sports Med Arthrosc Rehab Ther Technol. 2010;2:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Nakamura N, Miyama T, Engebretsen L, Yoshikawa H, Shino K. Cell-based therapy in articular cartilage lesions of the knee. Arthroscopy. 2009;25:531-552. [DOI] [PubMed] [Google Scholar]
- 84. Nejadnik H, Hui JH, Feng Choong EP, Tai BC, Lee EH. Autologous bone marrow-derived mesenchymal stem cells versus autologous chondrocyte implantation: an observational cohort study. Am J Sports Med. 2010;38:1110-1116. [DOI] [PubMed] [Google Scholar]
- 85. OCEBM Levels of Evidence Working Group. The Oxford 2011 Levels of Evidence. Oxford, England: Oxford Centre for Evidence-Based Medicine; 2011. [Google Scholar]
- 86. Oliveira JT, Gardel LS, Rada T, Martins L, Gomes ME, Reis RL. Injectable gellan gum hydrogels with autologous cells for the treatment of rabbit articular cartilage defects. J Orthop Res. 2010;28:1193-1199. [DOI] [PubMed] [Google Scholar]
- 87. Orozco L, Munar A, Soler R, et al. Treatment of knee osteoarthritis with autologous mesenchymal stem cells: a pilot study. Transplantation. 2013;95:1535-1541. [DOI] [PubMed] [Google Scholar]
- 88. Pak J. Regeneration of human bones in hip osteonecrosis and human cartilage in knee osteoarthritis with autologous adipose-tissue-derived stem cells: a case series. J Med Case Rep. 2011;5:296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Pak J, Lee JH, Lee SH. A novel biological approach to treat chondromalacia patellae. PLoS One. 2013;8:e64569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Pei M, He F, Boyce BM, Kish VL. Repair of full-thickness femoral condyle cartilage defects using allogeneic synovial cell-engineered tissue constructs. Osteoarthritis Cartilage. 2009;17:714-722. [DOI] [PubMed] [Google Scholar]
- 91. Pestka JM, Bode G, Salzmann G, Südkamp NP, Niemeyer P. Clinical outcome of autologous chondrocyte implantation for failed microfracture treatment of full-thickness cartilage defects of the knee joint. Am J Sports Med. 2012;40:325-331. [DOI] [PubMed] [Google Scholar]
- 92. Pierini M, Di Bella C, Dozza B, et al. The posterior iliac crest outperforms the anterior iliac crest when obtaining mesenchymal stem cells from bone marrow. J Bone Joint Surg Am. 2013;95:1101-1107. [DOI] [PubMed] [Google Scholar]
- 93. Pittenger MF, Mackay AM, Beck SC, et al. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143-147. [DOI] [PubMed] [Google Scholar]
- 94. Richter M, Zech S. Matrix-associated stem cell transplantation (MAST) in chondral defects of foot and ankle is effective. Foot Ankle Surg. 2013;19:84-90. [DOI] [PubMed] [Google Scholar]
- 95. Rodriguez JP, Murphy MP, Hong S, et al. Autologous stromal vascular fraction therapy for rheumatoid arthritis: rationale and clinical safety. Int Arch Med. 2012;5:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Rosland GV, Svendsen A, Torsvik A, et al. Long-term cultures of bone marrow-derived human mesenchymal stem cells frequently undergo spontaneous malignant transformation. Cancer Res. 2009;69:5331-5339. [DOI] [PubMed] [Google Scholar]
- 97. Rubio D, Garcia-Castro J, Martin MC, et al. Spontaneous human adult stem cell transformation. Cancer Res. 2005;65:3035-3039. [DOI] [PubMed] [Google Scholar]
- 98. Saris DB, Vanlauwe J, Victor J, et al. Characterized chondrocyte implantation results in better structural repair when treating symptomatic cartilage defects of the knee in a randomized controlled trial versus microfracture. Am J Sports Med. 2008;36:235-246. [DOI] [PubMed] [Google Scholar]
- 99. Saris DB, Vanlauwe J, Victor J, et al. Treatment of symptomatic cartilage defects of the knee: characterized chondrocyte implantation results in better clinical outcome at 36 months in a randomized trial compared to microfracture. Am J Sports Med. 2009;37(suppl 1):10S-19S. [DOI] [PubMed] [Google Scholar]
- 100. Sato M, Uchida K, Nakajima H, et al. Direct transplantation of mesenchymal stem cells into the knee joints of Hartley strain guinea pigs with spontaneous osteoarthritis. Arthritis Res Ther. 2012;14:R31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Saw KY, 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;27:493-506. [DOI] [PubMed] [Google Scholar]
- 102. Saw KY, Anz A, Siew-Yoke Jee C, et al. Articular cartilage regeneration with autologous peripheral blood stem cells versus hyaluronic acid: a randomized controlled trial. Arthroscopy. 2013;29:684-694. [DOI] [PubMed] [Google Scholar]
- 103. Skowroński J, Skowroński R, Rutka M. Cartilage lesions of the knee treated with blood mesenchymal stem cells - results. Ortop Traumatol Rehabil. 2012;14:569-577. [DOI] [PubMed] [Google Scholar]
- 104. Skowroński J, Skowroński R, Rutka M. Large cartilage lesions of the knee treated with bone marrow concentrate and collagen membrane—results. Ortop Traumatol Rehabil. 2013;15:69-76. [DOI] [PubMed] [Google Scholar]
- 105. Skowroński J, Rutka M. Osteochondral lesions of the knee reconstructed with mesenchymal stem cells—results. Ortop Traumatol Rehabil. 2013;15:195-204. [DOI] [PubMed] [Google Scholar]
- 106. Stolzing A, Jones E, McGonagle D, Scutt A. Age-related changes in human bone marrow-derived mesenchymal stem cells: consequences for cell therapies. Mech Ageing Dev. 2008;129:163-173. [DOI] [PubMed] [Google Scholar]
- 107. Sun J, Hou XK, 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;129:757-771. [DOI] [PubMed] [Google Scholar]
- 108. Takahashi K, Tanabe K, Ohnuki M, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861-872. [DOI] [PubMed] [Google Scholar]
- 109. Teo BJ, Buhary K, Tai BC, Hui JH. Cell-based therapy improves function in adolescents and young adults with patellar osteochondritis dissecans. Clin Orthop Relat Res. 2013;471:1152-1158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Toghraie FS, Chenari N, Gholipour MA, et al. Treatment of osteoarthritis with infrapatellar fat pad–derived mesenchymal stem cells in rabbit. Knee. 2011;18:71-75. [DOI] [PubMed] [Google Scholar]
- 111. Tolar J, Nauta AJ, Osborn MJ, et al. Sarcoma derived from cultured mesenchymal stem cells. Stem Cells. 2007;25:371-379. [DOI] [PubMed] [Google Scholar]
- 112. Torsvik A, Rosland GV, Svendsen A, et al. Spontaneous malignant transformation of human mesenchymal stem cells reflects cross-contamination: putting the research field on track [letter]. Cancer Res. 2010;70:6393-6396. [DOI] [PubMed] [Google Scholar]
- 113. Turajane T, Chaweewannakorn U, Larbpaiboonpong V, et al. Combination of intra-articular autologous activated peripheral blood stem cells with growth factor addition/preservation and hyaluronic acid in conjunction with arthroscopic microdrilling mesenchymal cell stimulation Improves quality of life and regenerates articular cartilage in early osteoarthritic knee disease. J Med Assoc Thai. 2013;96:580-588. [PubMed] [Google Scholar]
- 114. US Food and Drug Administration. Consumer information on stem cells. http://www.fda.gov/newsevents/publichealthfocus/ucm286218.htm Accessed December 3, 2013.
- 115. Vanlauwe JJ, Claes T, Van Assche D, Bellemans J, Luyten FP. Characterized chondrocyte implantation in the patellofemoral joint: an up to 4 year follow-up of a prospective cohort of 38 patients. Am J Sports Med. 2012;40:1799-1807. [DOI] [PubMed] [Google Scholar]
- 116. Vannini F, Battaglia M, Buda R, Cavallo M, Giannini S. “One step” treatment of juvenile osteochondritis dissecans in the knee: clinical results and T2 mapping characterization. Orthop Clin North Am. 2012;43:237-244. [DOI] [PubMed] [Google Scholar]
- 117. Varma HS, Dadarya B, Vidyarthi A. The new avenues in the management of osteo-arthritis of knee—stem cells. J Indian Med Assoc. 2010;108:583-585. [PubMed] [Google Scholar]
- 118. Wakitani S, Goto T, Pineda SJ, et al. Mesenchymal cell-based repair of large, full thickness defects of articular cartilage. J Bone Joint Surg Am. 1994;76:579-592. [DOI] [PubMed] [Google Scholar]
- 119. 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;10:199-206. [DOI] [PubMed] [Google Scholar]
- 120. Wakitani S, Mitsuoka T, Nakamura N, Toritsuka Y, Nakamura Y, Horibe S. Autologous bone marrow stromal cell transplantation for repair of full-thickness articular cartilage defects in human patellae: two case reports. Cell Transplant. 2004;13:595-600. [DOI] [PubMed] [Google Scholar]
- 121. 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. 2007;1:74-79. [DOI] [PubMed] [Google Scholar]
- 122. Wakitani S, Kawaguchi A, Tokuhara Y, Takaoka K. Present status of and future direction for articular cartilage repair. J Bone Miner Metab. 2008;26:115-122. [DOI] [PubMed] [Google Scholar]
- 123. Wakitani S, Okabe T, Horibe S, et al. Safety of autologous bone marrow–derived mesenchymal stem cell transplantation for cartilage repair in 41 patients with 45 joints followed for up to 11 years and 5 months. J Tissue Eng Regen Med. 2011;5:146-150. [DOI] [PubMed] [Google Scholar]
- 124. Wang JF, Wang LJ, Wu YF, et al. Mesenchymal stem/progenitor cells in human umbilical cord blood as support for ex vivo expansion of CD34(+) hematopoietic stem cells and for chondrogenic differentiation. Haematologica. 2004;89:837-844. [PubMed] [Google Scholar]
- 125. Wang Y, Hall S, Hanna F, et al. Effects of Hylan G-F 20 supplementation on cartilage preservation detected by magnetic resonance imaging in osteoarthritis of the knee: a two-year single-blind clinical trial. BMC Musculoskelet Disord. 2011;12:195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Wei Y, Zeng W, Wan R, et al. Chondrogenic differentiation of induced pluripotent stem cells from osteoarthritic chondrocytes in alginate matrix. Eur Cell Mater. 2012;23:1-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Wickham MQ, Erickson GR, Gimble JM, Vail TP, Guilak F. Multipotent stromal cells derived from the infrapatellar fat pad of the knee. Clin Orthop Relat Res. 2003;412:196-212. [DOI] [PubMed] [Google Scholar]
- 128. Zeifang F, Oberle D, Nierhoff C, Richter W, Moradi B, Schmitt H. Autologous chondrocyte implantation using the original periosteum-cover technique versus matrix-associated autologous chondrocyte implantation: a randomized clinical trial. Am J Sports Med. 2010;38:924-933. [DOI] [PubMed] [Google Scholar]
- 129. Zhang HN, Li L, Leng P, Wang YZ, Lv CY. Uninduced adipose-derived stem cells repair the defect of full-thickness hyaline cartilage. Chin J Traumatol. 2009;12:92-97. [PubMed] [Google Scholar]
- 130. Zuk PA, Zhu M, Ashjian P, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13:4279-4295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Zuk PA, Zhu M, Mizuno H, et al. Multilineage cells from human adipose tissue: implications for cell based therapies. Tissue Eng. 2001;7:211-228. [DOI] [PubMed] [Google Scholar]