header advert
Bone & Joint 360 Logo

Receive monthly Table of Contents alerts from Bone & Joint 360

Comprehensive article alerts can be set up and managed through your account settings

View my account settings

Visit Bone & Joint 360 at:

Loading...

Loading...

Full Access

Feature

Biologics in orthopaedics

Concepts and controversies



Download PDF

Background

Biological approaches to treat orthopaedic injuries seek to improve clinical outcomes by promoting tissue regeneration and healing. These approaches can be used in isolation or as an augment to surgical interventions. They include, but are not limited to, preparations of growth factors, autologous blood products, and cells. Despite considerable research effort, the majority of biological approaches have not yet achieved a sufficient evidence base to warrant widespread clinical application, and inappropriate use is a growing problem. This article aims to outline concepts underlying some of the key biological approaches, namely growth factors, autologous blood products (including platelet-rich plasma or PRP), and mesenchymal stem/stromal cells (MSCs), highlighting current challenges that limit clinical translation and application.

The race to the clinic has been at the expense of scientific understanding

There is a good rationale for using biological strategies to accelerate tissue healing. Nature uses cells and growth factors as the building blocks for tissue repair and, as such, approaches that seek to emulate this process seem, on the face of it, intuitive. Unfortunately, the race to clinical use has often been at the expense of scientific understanding or supportive data. Promising preliminary results from laboratory studies,1-3 modest regulatory barriers of certain autologous preparations, and public appetite for new treatments have all fuelled a rush to find new applications in which biological treatments may incur clinical benefit.1,2,4 Patient popularity has been further driven by public celebrity endorsements from athletes including Tiger Woods5 in golf and Peyton Manning6 in American football. Many biological strategies are now being used to treat a wide range of clinical conditions in mainstream orthopaedic practice, despite a lack of robust clinical evidence supporting efficacy.7 This is not only a problem in so-called ‘pirate clinics’ where some physicians are seizing an opportunity to make a fast buck, but also in larger teaching institutions. To put the problem into perspective, there are now over 400 complete or ongoing clinical trials evaluating the use of PRP and over 800 evaluating the use of MSCs in a range of clinical applications (see clinicaltrials.gov). Many of these trials have been designed and started with little knowledge of what preparations contain, and without comprehensive scientific understanding of the mechanisms by which it may produce benefit.7 There is an evident danger here that potentially beneficial treatments will be dismissed as ineffective simply because scientifically questionable and sub-optimized preparations are being rushed directly into a clinical setting. We will only truly know if biological therapies can be of therapeutic benefit if the scientific/clinical community accepts that shortcuts cannot be taken, and adopts a responsible approach to the use of biological therapies including the generation of both an evidence base to support their use and an understanding of the principles of use.

First consider the injury microenvironment

Biological approaches should always begin with an understanding of the underlying injury microenvironment. From the injury perspective, specific deficiencies in cells, cytokines, or the mechanical environment that contributes to pathology should be identified, and, from a regeneration perspective, opportunities to enhance particular components of the healing response can then be exploited. Once the biological targets for a specific scenario are identified, the ideal biological formulation should then be matched to the clinical setting. The effect of biological strategies relies on a complex interplay between the injury microenvironment and the biological preparations being delivered. Important injury factors contributing to variability include tissue type, and the mechanism and chronicity of injury. The therapeutic ‘needs’ of each injury will therefore be different. For example, ligaments are relatively acellular and avascular, while skeletal muscle is highly cellular and generously supplied with blood vessels. Ligaments are largely static constraints, while skeletal muscle is designed to contract. The archetypal cell within ligament (fibroblast) is the cell least welcome in the long term following skeletal muscle injury, and the presence of fibroblast-produced tissue (fibrosis) is a principal predictor of re-injury. This highlights that injuries affecting different tissue types, and of different chronicity, may not be best served by the same formulations, and treatments must therefore be individualized.

Growth factors and cytokines

Growth factors are part of the normal response to injury, acting to promote tissue regeneration and healing. Most growth factors are pleiotropic, causing multiple biological effects in a variety of cell types. Although the term ‘cytokine’ is sometimes used interchangeably with ‘growth factor’, cytokines are a unique family of growth factors secreted primarily from leucocytes that stimulate both the humeral and immune responses, as well as activation of phagocytes. Growth factors bind with specific receptor molecules on the surface of target cells and the presence of these receptors define a cell’s capacity to respond to the signals. A large number of growth factors have effects relevant to musculoskeletal regeneration and therefore represent potential therapeutic targets (Table 1), not just cytokines. It is imperative to note that not all factors will promote healthy regeneration in all tissues at all times – moreover, some may do more harm than good. For example, cues such as transforming growth factor beta (TGFβ)-1 that encourage the persistence of matrix-producing fibroblasts are undesirable in skeletal muscle,8 yet this cytokine has been shown to be beneficial for healing of ligament.9 TGFβ-1 also has angiogenic actions that may be of particular value in tissues where healing is delayed secondary to reduced vascularity.10

Table I.

Biological effects of a selected number of growth factors with activity relevant to musculoskeletal tissue engineering

Growth factor Selected biological effects
BMP2 MSC proliferation; promotes differentiation of MSCs to osteoblasts and chondrocytes
BMP4 Promotes differentiation of MSCs to osteoblasts and chondrocytes
BMP6 Promotes differentiation of MSCs to osteoblasts and chondrocytes
BMP7 Promotes differentiation of MSCs to osteoblasts and chondrocytes
CTGF Pro-angiogenic; promotes wound healing by increasing fibroblast proliferation
EGF MSC proliferation; regulates bone and cartilage formation
FGF1 Fibroblast proliferation; pro-angiogenic
FGF1 Pro-angiogenic; regulates osteogenic, myogenic, and chondrogenic differentiation
HGF Pro-angiogenic; antifibrotic; reduces local immune response
IGF1 Increases MSC proliferation and differentiation to osteoblasts and chondrocytes
IL-1 Pro-inflammatory
PDGF Promotes MSC proliferation and differentiation to osteoblasts; pro-inflammatory
NELL1 Induces osteoblastic differentiation of MSCs
SDF1α Pro-inflammatory; increases homing of MSCs
TGFβ-1 Fibroblast activation and proliferation; pro-angiogenic; MSC proliferation; chondrogenic and osteogenic differentiation
TGFβ-2 MSC proliferation; chondrogenic and osteogenic differentiation
TGFβ-3 MSC proliferation; chondrogenic and osteogenic differentiation
VEGF Stimulates angiogenesis; attracts macrophages and granulocytes
Wnt3a MSC proliferation and survival; regulates osteogenic and chondrogenic differentiation of MSCs
Wnt5a Regulates osteogenic and chondrogenic differentiation of MSCs
  1. BMP, bone morphogenetic protein; MSCs, mesenchymal stem/stromal cells; CTGF, connective tissue growth factor; EGF, endothelial growth factor; FGF, basic fibroblasts growth factor; HGF, hepatocyte growth factor; IGF, insulin-like growth factor; IL-1, interleukin 1; PDGF, platelet-derived growth factor; NELL1, NEL-like protein 1; SDF1α, stromal cell derived factor alpha; TGFβ, transforming growth factor beta; VEGF, vascular endothelial growth factor

Growth factors can be delivered either individually or in synergistic combinations. They can be used to pre-condition cells prior to delivery or combined with cells at the time of delivery.11 The number of commercially available purified cytokine products is low: platelet derived growth factor (PDGF) is the sole protein in three United States Food and Drug administration (FDA)-cleared products for patient use, while bone-morphogenetic protein (BMP)-2 is the sole protein in another FDA-approved product. BMP-2 has demonstrated beneficial effect on fracture healing in randomized controlled trials.12,13 Other agents such as fibroblast growth factor (FGF) have shown promise in clinical studies,14 while others, including TGF-β, have been evaluated in small animals.15,16

Autologous blood products/platelet-rich plasma

Not all PRP preparations are the same

The term PRP can be, and is, used to describe any autologous blood preparation with a platelet concentration higher than baseline. This is normally achieved through differential centrifugation, with collection of the PRP from above the white blood cell layer. A second spin is frequently performed and has the effect, in those preparations in which it is used, to further concentrate the platelets. Platelets are recognized to release a host of growth factors and cytokines that are able to induce pro-regenerative attributes in laboratory studies, including promoting proliferation and recruitment of progenitor cells, modulation of inflammatory responses, and stimulation of new blood vessel formation.17 In addition to platelets, these preparations can contain leucocytes, red blood cells, and over 300 different growth factors and cytokines in variable numbers (Table II).18,19 The heterogeneous nature of PRP preparations is widely underappreciated. There are over 20 commercially available systems, with considerable differences in the PRP composition even among the most widely used brands (Table III).17 In addition to cell composition, variability is introduced by different methods of ‘activating’ platelets prior to delivery to encourage release of their contained growth factors. PRP composition also varies considerably between individuals and even the same individuals can have variable amounts of growth factors in the PRP produced on a day-to-day basis, and this can even be affected by recent exercise or meals.20 Since the introduction of PRP, a number of other autologous blood products have emerged, including platelet poor plasma (PPP) and other autologous protein solutions. Regardless of the method of preparation, all autologous blood products should be considered highly variable and intrinsically different for each individual.

Table II.

Fractions of PRP and their contents

PRP fraction Content or releasate
Plasma
Proteins Albumin, fibrinogen, globulins, complement, clotting factors
Electrolytes Sodium, chloride, potassium, calcium
Hormones IGF1, ACTH, HGH estrogens, progesterone, androgens
Platelets
Alpha granules Growth factors (over 300 including TGFβ1, IGF1, bFGF, PDGF, PDAF, PF4, EGF, VEGF, CTGF, HGF, SDF1α)
Clotting factors: Factor V, vWF, fibrinogen
Dense granules ADP, calcium, serotonin
Lysosomes Lysosomal enzymes
Leucocytes
Neutrophils Cytokines (e.g. IL-4, IL-8, TNFα), proteases, bactericidal molecules, lysozyme
Eosinophils Growth factors (including VEGF, PDGF, TGFα, TGFβ, interleukins), plasminogen
Basophils Histamine, proteases, heparin, leukotrienes
Monocytes Growth factors (including TGFβ, IL-1, FGF, PDGF)
Red blood cells Hemoglobin, nitric oxide, ATP, S-nitrosothiols, and free radicals
  1. IGF1, insulin-like growth factor 1; ACTH, adrenocorticotropic hormone; HGH, human growth hormone; TGFβ, transforming growth factor beta; bFGF, basic fibroblastic growth factor; PDGF, platelet derived growth factor; PDAF, platelet-derived angiogenesis factor; PF4, platelet factor 4; EGF, endothelial growth factor; VEGF, vascular endothelial growth factor; CTGF, connective tissue growth factor; HGF, hepatocyte growth factor; SDF1α, stromal cell derived factor; vWF, von Willebrand factor; ADP, adenosine diphosphate; IL, interleukin; TNFα, tumour necrosis factor alpha; TGFα, transforming growth factor alpha; ATP, adenosine triphosphate

Table III.

Differences in platelet count, white cell count and neutrophil count of selected commercial platelet-rich plasma (PRP) systems

PRP preparation Platelets (fold change) Leucocytes (fold change) Neutrophils (fold change)
GPS (Zimmer Biomet)* ×8.21 ×4.87 ×2.04
Magellan autologous platelet system (Medtronic perfusion systems, now Arteriocyte) ×9.29 ×5.4 ×1.99
Arthrex ACP Double Syringe (Arthrex) ×2.40 ×0.54 ×0.06
Genesis (EmCyte) ×5.53 ×4.30 ×2.86
Smart prep 2 system (Harvest autologous hemobiologics) ×7.25 ×4.3 ×1.57
Regen (Stryker) ×1.70 ×0.95 N/A
  1. *

    Data provided by Zimmer Biomet

  1. Data provided by Arthrex

  1. N/A, not available

Matching conditions with the ‘ideal’ formulation

Autologous blood products should be selected based on the formulation most suited to the pathology being treated. PRP preparations can be potentially and theoretically manipulated in this way by enriching or depleting certain growth factors or cell types. In osteoarthritis, a meta-analysis of six randomized controlled trials (evidence level 1) and three prospective comparative studies (evidence level 2) with a total of 1055 patients indicates that leucocyte-poor PRP, but not leucocyte-rich PRP, can result in symptomatic improvement over treatment with hyaluronic acid or placebo control.21 This may relate to the significant increase in anti-inflammatory mediators in leucocyte-poor PRP (interleukin (IL)-1ra, IL-4, IL-10), and the significant increase in inflammatory mediators seen in leucocyte-rich PRP (IL-1β, IL-6, interferon (IFN)-γ, tumour necrosis factor (TNF)-α).22 Conversely, multiple randomized control trials and meta-analyses have found that leucocyte-rich PRP is an efficacious treatment for lateral epicondylitis (tennis elbow).23-25 At present, there is little evidence to suggest an optimal formulation for acute ligament or muscle injury. Interestingly, laboratory studies suggest that autologous blood preparations depleted of platelets (platelet-poor plasma) may have therapeutic benefit in the setting of muscle injury.26

Current studies fail to characterize PRP and under-report methods

A major challenge in interpreting the currently published studies is the inadequate reporting of experimental detail or compositions of PRP formulations used.27 This precludes proper interpretation of results, prevents comparison between studies, and does not permit others to replicate experimental or clinical conditions to verify results. A recent systematic review concluded that only 16% of published clinical studies provided any quantitative metrics of the composition of PRP delivered.27 To address inadequacies in reporting, Murray et al28 facilitated an international expert consensus to establish minimum reporting standards for studies evaluating PRP. These are now approved by the EQUATOR network of journals (including The BMJ and The Lancet) and a growing number of orthopaedic journals.29

Cell therapies

Autologous chondrocyte implantation

Autologous chondrocyte implantation (ACI) is currently the only cell therapy recommended by the National Institute for Clinical Excellence (NICE) to treat musculoskeletal defects.30 ACI involves the harvesting of chondrocytes from a healthy non-weight-bearing portion of the knee followed by implantation of culture-expanded autologous chondrocytes under a periosteal flap (first-generation ACI) or a bioabsorbable collagen membrane (second-generation ACI), or onto a porcine membrane (MACI).31 MACI has now received approval from the FDA and is widely available in Europe. MACI was recently recommended by NICE as an option for treating large (> 2 cm2) symptomatic articular cartilage defects of the knee. Confirmation of cell viability, identity, and potency are critical quality assessments of seeded cells, and it is, of course, important that a characterized strain of chondrocytes, selected for their improved ability to generate hyaline cartilage, is used. The recently published five-year outcomes of the SUMMIT (Superiority of MACI Implant Versus Microfracture Treatment) randomized controlled clinical trial demonstrated that symptomatic cartilage knee defects 3 cm2 or larger can be treated safely with MACI resulting in clinically and statistically improved outcomes at five years compared with microfracture alone.32

Mesenchymal stem/stromal cells (MSCs)

Mesenchymal stem (or stromal) cells (MSCs) are multipotent cells with the capacity to multiply (‘self-renew’) and differentiate into osteocytes, chondrocytes, and other mesodermal cell types. They also release pro-regenerative growth factors and cytokines with immune modulating effects. Friedenstein et al33 first described cells from bone marrow that could replicate and become bone in the 1960s. Observations that these cells could also form fat, cartilage, and muscle led to the term ‘mesenchymal stem cells’ being introduced in 1991.34 The International Society for Cellular Therapy (ISCT) have since proposed minimum defining criteria that included a propensity to adhere to laboratory culture plastic and the capacity to differentiate into bone, cartilage, and fat and to express certain cell surface markers (Fig. 1).35

Fig. 1 
            International Society for Cell Therapy (ISCT) defining criteria for mesenchymal stem/stromal cells (MSCs).

Fig. 1

International Society for Cell Therapy (ISCT) defining criteria for mesenchymal stem/stromal cells (MSCs).

It has since been reported that MSCs reside around blood vessels in almost every tissue, including subcutaneous fat, and adipose tissue isolated from the knee fat pad.36 While all these populations fulfil ISCT criteria, they have distinctive features reflecting their tissue of origin. For example, MSCs from bone marrow exposed to TGF-β form chondrocytes and make cartilage extracellular matrix, while fast-derived MSCs require both TGF-β and bone BMP-6 to make cartilage.37 A timeline of the discovery of MSCs and their application in clinical studies is outlined in Figure 2. Interestingly, MSCs were being used in clinical trials long before the native identity of MSCs within blood vessel walls was known.

Fig. 2 
            A timeline of mesenchymal stem/stromal cells. Note that clinical trials using these cells have been running long before it was known where these cells resided within native tissues.

Fig. 2

A timeline of mesenchymal stem/stromal cells. Note that clinical trials using these cells have been running long before it was known where these cells resided within native tissues.

MSC preparations

Irrespective of their tissue of origin, MSC preparations can be broadly categorized on how separated they are from other cell types (homogeneity) and whether they have been exposed to laboratory culture. Although bone marrow represents the most widely used source (Fig. 3), these concepts can be equally applied to MSCs isolated from different tissues including adipose tissue which offers an attractive alternative because it is plentiful and largely dispensable.38 The yield of MSCs harvested from adipose tissues is reported to be higher than from bone marrow.39-40 The infrapatellar fat pad is also recognized as a source for adipose-derived stem cells. When considering commercial systems, clinicians should consider where in this framework any product fits as aspects of preparation are known to reflect regenerative characteristics.

  1. Minimally manipulated ‘heterogenous’ preparations (e.g. bone marrow aspirate concentrate, or BMAC). Although MSCs make up a small minority of cells within bone marrow (less than 1/10 000 cells) or fat, preparations that have undergone ‘minimal manipulation’ processing techniques such as centrifugation or mechanical disruption only (e.g. concentrated bone marrow aspirate) have been used directly with the aim of harnessing the potential of contained progenitors.41 These strategies are widely available and now represent the majority of clinical studies evaluating MSCs in the clinical literature.42 They are attractive to clinicians and patients because unlike culture preparations, harvest, and delivery can occur under the same procedure. Minimally manipulated preparations can also be utilized from fat.43 However, available studies demonstrate that these heterogeneous populations, including inflammatory cells, hematopoietic cells, endothelial cells, and nonviable cells, may result in poor and inconsistent tissue formation compared with enriched MSC preparations, as the contaminating cells may have inhibitory effects on the MSCs.44-47

  2. Culture-derived MSCs. A period of laboratory culture can be used to both enrich for MSCs, and increase their number, as MSCs tend to outgrow other cells in these conditions.48 A number of clinical studies have demonstrated that MSC implantation can be used effectively to repair and regenerate bone49 and cartilage.50 Promising results have been demonstrated in the treatment of chondral51,52 and osteochondral defects,53,54 osteonecrosis of the femoral head55 and to aid spinal fusion56 to name a few. However, controversy remains as to whether long-term culture is associated with genetic instability and a reduction in therapeutic potency.57

  3. MSCs purified through affinity-based separation. Using fluorescent activated cell sorting (FACS) or magnetic activated cell sorting (MACS), MSCs can now be rapidly separated from other cells in suspension using antibodies that target known cell surface markers of MSCs. This process does not require extended periods of laboratory culture and harvest and delivery of pure populations of MSCs can theoretically be performed under the same anaesthetic. Using this strategy, 200 ml of lipoaspirate can theoretically yield 31 million MSCs, sufficient for healing a mid-diaphyseal femoral defect measuring 2 cm in diameter.38 In addition, for the need for robust clinical trials, this strategy is currently limited by a lack of formal accreditation from the relevant regulatory bodies and high costs.

Fig. 3 
            Mesenchymal stem/stromal cell (MSC) preparations can be broadly categorized based on whether the preparations are heterogenous (contained MSCs remain mixed with other cell types), the preparations have been enriched for MSCs through laboratory culture, or the MSCs have been purified through cell sorting techniques. Similar categories of cells can be prepared from multiple different tissue types including bone marrow (shown here), adipose tissue and periosteum.

Fig. 3

Mesenchymal stem/stromal cell (MSC) preparations can be broadly categorized based on whether the preparations are heterogenous (contained MSCs remain mixed with other cell types), the preparations have been enriched for MSCs through laboratory culture, or the MSCs have been purified through cell sorting techniques. Similar categories of cells can be prepared from multiple different tissue types including bone marrow (shown here), adipose tissue and periosteum.

Ambiguous nomenclature and misrepresentation of biologics

There is growing concern about the misrepresentation of uncharacterized, minimally manipulated cell preparations as ‘stem cells’ leading to widespread clinical use of unproven biological therapies. Misinformation is frequently delivered by direct-to-consumer marketing. From 2014 to 2016, approximately 90 to 100 new stem cell business websites appeared per year, many of which offered services without any orthopaedic surgeon involvement. There is concern that the current environment may erode the trust in biological therapies and the investment and grant funding required to bring legitimate cell therapies to patients.58 There is a clear need to define terminology to clearly distinguish uncharacterized cell products from rigorously characterized, culture expanded, and purified ‘stem cell’ and progenitor cell populations.

Summary

The pro-regenerative characteristics of growth factors and regenerative cell types hold great promise for musculoskeletal tissue engineering. Early favourable results from in vitro work are now being supported by level 1 studies in a limited number of clinical settings. Irrespective of the setting, clinical use should be based on a comprehensive understanding of injury, environment, and the biological therapy being delivered.


email:

References

1. Zaky SH , Ottonello A , Strada P , Cancedda R , Mastrogiacomo M . Platelet lysate favours in vitro expansion of human bone marrow stromal cells for bone and cartilage engineering. J Tissue Eng Regen Med2008;2:472-481.CrossrefPubMed Google Scholar

2. Mishra A , Tummala P , King A et al. . Buffered platelet-rich plasma enhances mesenchymal stem cell proliferation and chondrogenic differentiation. Tissue Eng Part C Methods2009;15:431-435.CrossrefPubMed Google Scholar

3. Tawonsawatruk T , West CC , Murray IR et al. . Adipose derived pericytes rescue fractures from a failure of healing—non-union. Sci Rep2016;6:22779. Google Scholar

4. No authors listed. US Department of Health and Human Services: Human cells, tissues, and cellular and tissue-based products. 21 CFR Part1271. 2018. http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?CFRPart=1271&showFR=1 (date last accessed 12October2018). Google Scholar

5. Cox L . Tiger admits to platelet-rich plasma therapy, what's that?ABC News. 2010. http://abcnews.go.com/Health/Technology/tiger-woods-admits-platelet-rich-plasma-therapy/story?id=10303312 (date last accessed 12October2018). Google Scholar

6. Regalado A . The NFL has a problem with stem cell treatments. Technol Rev2014. https://www.technologyreview.com/s/533171/the-nfl-has-a-problem-with-stem-cell-treatments/ Google Scholar

7. Murray IR , LaPrade RF . Platelet-rich plasma: renewed scientific understanding must guide appropriate use. Bone Joint Res2016;5:92-94.CrossrefPubMed Google Scholar

8. Huard J , Li Y , Fu FH . Muscle injuries and repair: current trends in research. J Bone Joint Surg [Am]2002;84-A:822-32.PubMed Google Scholar

9. Beye JA , Hart DA , Bray RC , McDougall JJ , Salo PT . Injury-induced changes in mRNA levels differ widely between anterior cruciate ligament and medial collateral ligament. Am J Sports Med2008;36:1337-1346.CrossrefPubMed Google Scholar

10. Maerz T , Herkowitz H , Baker K . Molecular and genetic advances in the regeneration of the intervertebral disc. Surg Neurol Int2013;4:S94-S105.CrossrefPubMed Google Scholar

11. Caplan AI , Correa D . The MSC: an injury drugstore. Cell Stem Cell2011;9:11-15.CrossrefPubMed Google Scholar

12. Friedlaender GE , Perry CR , Cole JD et al. . Osteogenic protein-1 (bone morphogenetic protein-7) in the treatment of tibial nonunions. J Bone Joint Surg [Am]2001;83-A:S151-S158.PubMed Google Scholar

13. Govender S , Csimma C , Genant HK , Valentin-Opran A ; BMP-2 Evaluation in Surgery for Tibial Trauma (BESTT) Study Group. Recombinant human bone morphogenetic protein-2 for treatment of open tibial fractures: a prospective, controlled, randomized study of four hundred and fifty patients. J Bone Joint Surg [Am]2002;84-A:2123-2134.CrossrefPubMed Google Scholar

14. Akita S , Akino K , Tanaka K , Anraku K , Hirano A . A basic fibroblast growth factor improves lower extremity wound healing with a porcine-derived skin substitute. J Trauma2008;64:809-815.CrossrefPubMed Google Scholar

15. Guo X , Zheng Q , Kulbatski I et al. . Bone regeneration with active angiogenesis by basic fibroblast growth factor gene transfected mesenchymal stem cells seeded on porous beta-TCP ceramic scaffolds. Biomed Mater2006;1:93-99.CrossrefPubMed Google Scholar

16. Park JS , Yang HJ , Woo DG et al. . Chondrogenic differentiation of mesenchymal stem cells embedded in a scaffold by long-term release of TGF-beta 3 complexed with chondroitin sulfate. J Biomed Mater Res A2010;92:806-816.CrossrefPubMed Google Scholar

17. Andia I , Maffulli N . Platelet-rich plasma for managing pain and inflammation in osteoarthritis. Nat Rev Rheumatol2013;9:721-730.CrossrefPubMed Google Scholar

18. Castillo TN , Pouliot MA , Kim HJ , Dragoo JL . Comparison of growth factor and platelet concentration from commercial platelet-rich plasma separation systems. Am J Sports Med2011;39:266-271.CrossrefPubMed Google Scholar

19. Coppinger JA , Cagney G , Toomey S et al. . Characterization of the proteins released from activated platelets leads to localization of novel platelet proteins in human atherosclerotic lesions. Blood2004;103:2096-2104.CrossrefPubMed Google Scholar

20. Mazzocca AD , McCarthy MB , Chowaniec DM et al. . Platelet-rich plasma differs according to preparation method and human variability. J Bone Joint Surg [Am]2012;94-A:308-316.CrossrefPubMed Google Scholar

21. Riboh JC , Saltzman BM , Yanke AB , Fortier L , Cole BJ . Effect of leukocyte concentration on the efficacy of platelet-rich plasma in the treatment of knee osteoarthritis. Am J Sports Med2016;44:792-800.CrossrefPubMed Google Scholar

22. Braun HJ , Kim HJ , Chu CR , Dragoo JL . The effect of platelet-rich plasma formulations and blood products on human synoviocytes: implications for intra-articular injury and therapy. Am J Sports Med2014;42:1204-1210.CrossrefPubMed Google Scholar

23. Gosens T , Peerbooms JC , van Laar W , den Oudsten BL . Ongoing positive effect of platelet-rich plasma versus corticosteroid injection in lateral epicondylitis: a double-blind randomized controlled trial with 2-year follow-up. Am J Sports Med2011;39:1200-1208.CrossrefPubMed Google Scholar

24. Mishra AK , Skrepnik NV , Edwards SG et al. . Efficacy of platelet-rich plasma for chronic tennis elbow: a double-blind, prospective, multicenter, randomized controlled trial of 230 patients. Am J Sports Med2014;42:463-471.CrossrefPubMed Google Scholar

25. Fitzpatrick J , Bulsara M , Zheng MH . The effectiveness of platelet-rich plasma in the treatment of tendinopathy: a meta-analysis of randomized controlled clinical trials. Am J Sports Med2017;45:226-233.CrossrefPubMed Google Scholar

26. Miroshnychenko O , Chang WT , Dragoo JL . The Use of platelet-rich and platelet-poor plasma to enhance differentiation of skeletal myoblasts: implications for the use of autologous blood products for muscle regeneration. Am J Sports Med2017;45:945-953.CrossrefPubMed Google Scholar

27. Chahla J , Cinque ME , Piuzzi NS et al. . A call for standardization in platelet-rich plasma preparation protocols and composition reporting: a systematic review of the clinical orthopaedic literature. J Bone Joint Surg [Am]2017;99-A:1769-1779.CrossrefPubMed Google Scholar

28. Murray IR , Geeslin AG , Goudie EB , Petrigliano FA , LaPrade RF . Minimum information for studies evaluating biologics in orthopaedics (MIBO): platelet-rich plasma and mesenchymal stem cells. J Bone Joint Surg [Am]2017;99-A:809-819.CrossrefPubMed Google Scholar

29. No authors listed. Minimum information for studies evaluating biologics in orthopaedics (MIBO): platelet-rich plasma and mesenchymal stem cells. Equator Network. 2018. https://www.equator-network.org/reporting-guidelines/minimum-information-for-studies-evaluating-biologics-in-orthopaedics-mibo-platelet-rich-plasma-and-mesenchymal-stem-cells (date last accessed 12October2018).CrossrefPubMed Google Scholar

30. No authors listed. National Institute for Health and Care Excellence (2017) Autologous chondrocyte implantation for treating symptomatic articular cartilage defects of the knee (NICE Guideline TA477). 2017. https://www.nice.org.uk/guidance/ta477 (date last accessed 12October2018). Google Scholar

31. Murray IR , Benke MT , Mandelbaum BR . Management of knee articular cartilage injuries in athletes: chondroprotection, chondrofacilitation, and resurfacing. Knee Surg Sports Traumatol Arthrosc2016;24:1617-1626.CrossrefPubMed Google Scholar

32. Brittberg M , Recker D , Ilgenfritz J , Saris DBF ; SUMMIT Extension Study Group. Matrix-applied characterized autologous cultured chondrocytes versus microfracture: five-year follow-up of a prospective randomized trial. Am J Sports Med2018;46:1343-1351.CrossrefPubMed Google Scholar

33. Friedenstein AJ , Piatetzky-Shapiro II , Petrakova KV . Osteogenesis in transplants of bone marrow cells. J Embryol Exp Morphol1966;16:381-390.PubMed Google Scholar

34. Caplan AI . Mesenchymal stem cells. J Orthop Res1991;9:641-650.CrossrefPubMed Google Scholar

35. Dominici M , Le Blanc K , Mueller I et al. . Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy2006;8:315-317.CrossrefPubMed Google Scholar

36. Crisan M , Yap S , Casteilla L et al. . A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell2008;3:301-313.CrossrefPubMed Google Scholar

37. Hennig T , Lorenz H , Thiel A et al. . Reduced chondrogenic potential of adipose tissue derived stromal cells correlates with an altered TGFbeta receptor and BMP profile and is overcome by BMP-6. J Cell Physiol2007;211:682-691.CrossrefPubMed Google Scholar

38. Murray IR , Corselli M , Petrigliano FA , Soo C , Péault B . Recent insights into the identity of mesenchymal stem cells: implications for orthopaedic applications. Bone Joint J2014;96-B:291-298.CrossrefPubMed Google Scholar

39. Aust L , Devlin B , Foster SJ et al. . Yield of human adipose-derived adult stem cells from liposuction aspirates. Cytotherapy2004;6:7-14.CrossrefPubMed Google Scholar

40. Zuk PA , Zhu M , Ashjian P et al. . Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell2002;13:4279-4295.CrossrefPubMed Google Scholar

41. Bonab MM , Alimoghaddam K , Talebian F . Aging of mesenchymal stem cell in vitro. BMC Cell Biol2006;7:14.CrossrefPubMed Google Scholar

42. Murray IR , Robinson PG , West CC et al. . Reporting standards in clinical studies evaluating bone marrow aspirate concentrate: a systematic review. Arthroscopy2018;34:1366-1375.CrossrefPubMed Google Scholar

43. Coccè V , Brini A , Giannì AB et al. . A nonenzymatic and automated closed-cycle process for the isolation of mesenchymal stromal cells in drug delivery applications. Stem Cells Int2018;2018:4098140.CrossrefPubMed Google Scholar

44. Müller AM , Mehrkens A , Schäfer DJ et al. . Towards an intraoperative engineering of osteogenic and vasculogenic grafts from the stromal vascular fraction of human adipose tissue. Eur Cell Mater2010;19:127-135.CrossrefPubMed Google Scholar

45. Cheung WK , Working DM , Galuppo LD , Leach JK . Osteogenic comparison of expanded and uncultured adipose stromal cells. Cytotherapy2010;12:554-562.CrossrefPubMed Google Scholar

46. Rajashekhar G , Traktuev DO , Roell WC et al. . IFATS collection: Adipose stromal cell differentiation is reduced by endothelial cell contact and paracrine communication: role of canonical Wnt signaling. Stem Cells 2008;26:2674-2681.CrossrefPubMed Google Scholar

47. Meury T , Verrier S , Alini M . Human endothelial cells inhibit BMSC differentiation into mature osteoblasts in vitro by interfering with osterix expression. J Cell Biochem2006;98:992-1006.CrossrefPubMed Google Scholar

48. Chen WC , Park TS , Murray IR et al. . Cellular kinetics of perivascular MSC precursors. Stem Cells Int2013;2013:983059.CrossrefPubMed Google Scholar

49. Griffin M , Iqbal SA , Bayat A . Exploring the application of mesenchymal stem cells in bone repair and regeneration. J Bone Joint Surg [Br]2011;93-B:427-434.CrossrefPubMed Google Scholar

50. Mohal JS , Tailor HD , Khan WS . Sources of adult mesenchymal stem cells and their applicability for musculoskeletal applications. Curr Stem Cell Res Ther2012;7:103-109.CrossrefPubMed Google Scholar

51. 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. Cartilage2010;1:253-261.CrossrefPubMed Google Scholar

52. Wakitani S , Imoto K , Yamamoto T . Human autologous culture expanded bone marrow mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees. Osteoarthritis Cartilage2002;10:199-206.CrossrefPubMed Google Scholar

53. Buda R , Vannini F , Cavallo M et al. . One-step arthroscopic technique for the treatment of osteochondral lesions of the knee with bone-marrow-derived cells: three years results. Musculoskelet Surg2013;97:145-151.CrossrefPubMed Google Scholar

54. 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 Med2013;41:511-518.CrossrefPubMed Google Scholar

55. Kawate K , Yajima H , Ohgushi H et al. . Tissue-engineered approach for the treatment of steroid-induced osteonecrosis of the femoral head: transplantation of autologous mesenchymal stem cells cultured with beta-tricalcium phosphate ceramics and free vascularized fibula. Artif Organs2006;30:960-962.CrossrefPubMed Google Scholar

56. Muschler GF , Matsukura Y , Nitto H et al. . Selective retention of bone marrow-derived cells to enhance spinal fusion. Clin Orthop Relat Res2005;432:242-251.CrossrefPubMed Google Scholar

57. Røsland GV , Svendsen A , Torsvik A et al. . Long-term cultures of bone marrow-derived human mesenchymal stem cells frequently undergo spontaneous malignant transformation. Cancer Res2009;69:5331-5339.CrossrefPubMed Google Scholar

58. Knoepfler PS , Turner LG . The FDA and the US direct-to-consumer marketplace for stem cell interventions: a temporal analysis. Regen Med2018;13:19-27.CrossrefPubMed Google Scholar