<<

CORE Metadata, citation and similar papers at core.ac.uk

Provided by Elsevier - Publisher Connector

Osteoarthritis and 23 (2015) 334e350

Review An educational review of cartilage repair: precepts & practice e myths & misconceptions e progress & prospects

* E.B. Hunziker , K. Lippuner a, M.J.B. Keel b, N. Shintani c

Departments of Osteoporosis, Orthopaedic Surgery and Clinical Research, Inselspital, University of Bern, Bern, Switzerland

article info summary

Article history: Objective: The repair of cartilaginous lesions within synovial joints is still an unresolved and weighty Received 10 July 2014 clinical problem. Although research activity in this area has been indefatigably sustained, no significant Accepted 12 December 2014 progress has been made during the past decade. The aim of this educational review is to heighten the awareness amongst students and scientists of the basic issues that must be tackled and resolved before Keywords: we can hope to escape from the whirlpool of stagnation into which we have fallen: cartilage repair Articular redivivus! Cartilage Design: Articular-cartilage lesions may be induced traumatically (e.g., by sports injuries and occupational Repair Tissue engineering accidents) or pathologically during the course of a degenerative disease (e.g., osteoarthritis). This review Osteoarthritis addresses the biological basis of cartilage repair and surveys current trends in treatment strategies, Review focussing on those that are most widely adopted by orthopaedic surgeons [viz., abrasive chondroplasty, microfracturing/microdrilling, osteochondral grafting and autologous- implantation (ACI)]. Also described are current research activities in the field of cartilage-tissue engineering, which, as a therapeutic principle, holds more promise for success than any other experimental approach. Results and Conclusions: Tissue engineering aims to reconstitute a tissue both structurally and func- tionally. This process can be conducted entirely in vitro, initially in vitro and then in vivo (in situ), or entirely in vivo. Three key constituents usually form the building blocks of such an approach: a matrix scaffold, cells, and signalling molecules. Of the proposed approaches, none have yet advanced beyond the phase of experimental development to the level of clinical induction. The hurdles that need to be sur- mounted for ultimate success are discussed. © 2014 Published by Elsevier Ltd on behalf of Osteoarthritis Research Society International.

Introduction Articular cartilage is a highly specialized tissue. Lesions therein seldom heal, or heal only partially under certain biological condi- The bony ends of mammalian synovial joints are lined with a tions. They are frequently associated with symptoms such as joint layer of articular cartilage (Fig. 1), which permits congruency be- pain, joint-locking phenomena and reduced or disturbed joint tween the two opposing skeletal elements, facilitates the transfer of function. Moreover, the progression of such lesions is generally e forces between these and their practically frictionless movement, believed to forebode the onset of osteoarthritis (OA)2 4. and acts as an absorber of weight during sustained static loading1. In Western countries, OA is one of the most common diseases suffered by elderly persons. It is a disabling joint disorder of multifactorial aetiology, and, consequently, of usually unknown * Address correspondence and reprint requests to: E.B. Hunziker, Center of origin in individual cases (primary OA). However, it can be Regenerative Medicine for Skeletal Tissues, Departments of Osteoporosis, Ortho- precipitated also by physical injury [e.g., by sports or occupational paedic Surgery and Clinical Research, Inselspital, University of Bern, Murtenstrasse accidents (secondary OA)]. The incidence of OA is positively 35, P.O. Box 54, 3010 Bern, Switzerland. Tel: 41-31-632-86-85; Fax: 41-31-632- 49-55. correlated not only with age but also with obesity, and due to the E-mail addresses: [email protected] (E.B. Hunziker), kurt.lippuner@ upward trends in both life-expectancy and youth overweight, a e insel.ch (K. Lippuner), [email protected] (M.J.B. Keel), nahoko.shintani@dkf. steady increase in the prevalence of the disease is expected5 7. unibe.ch (N. Shintani). Although joint pain is a common concomitant of OA, this symptom a Tel: 41-31-632-31-28; Fax: 41-31-632-95-96. b Tel: 41-31-632-22-13; Fax: 41-31-632-18-80. is not correlated with the size of the articular-cartilage lesion, and 8e10 c Tel: 41-31-632-09-37; Fax: 41-31-632-49-99. the mechanism underlying its generation is largely unknown .

http://dx.doi.org/10.1016/j.joca.2014.12.011 1063-4584/© 2014 Published by Elsevier Ltd on behalf of Osteoarthritis Research Society International. E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350 335

Lesioning of the articular-cartilage layer

As aforementioned, structural lesioning of an articular-cartilage layer can be induced either traumatically or during the course of a disease-based process. Once the process of lesioning has begun, it cannot be arrested, but progresses inexorably with time, the reason e for which is unknown14 17. Superficial cartilaginous lesions, viz., structural defects that are confined to the articular-cartilage layer itself and do not penetrate the subchondral- plate, represent one of the first overt signs of OA (namely, fibrillation of the carti- e laginous layer and loss of proteoglycans therefrom)18 20. Such lesions do not heal spontaneously21,22. With the lapse of time, they broaden and lengthen, such that what is initially a discrete condi- tion progresses insidiously towards a crippling disease. The reasons why lesions confined to the layer of adult articular cartilage do not heal lie mainly in the nature of this tissue's struc- ture and physiology: it is avascular, aneural and harbours but a sparse population of cells ()23,24, which endure and remain in their allotted positions throughout the life of the host (as is the case for neurons of the central nervous system and for cardiomyocytes). Under metabolically pathological conditions, and in the vicinity of traumatically-induced lesions, the chondrocytes respond but sluggishly to internal and external stimuli; they manifest only a limited proliferative capacity and a reduced e potential for the de-novo synthesis of an extracellular matrix25 27.

Spontaneous repair

From the point of view of its working status, a synovial joint (Fig. 2) can be considered as an organ. The articular-cartilage layer represents but one component of a functional unit, having struc- tural and physiological relationships with the synovial fluid, the synovial membrane, the subchondral-bone plate and the sub- chondral-, as well as with the trabecular bone and its vascular system. It is from these associated, highly-vascularized tissue-compartments that we might expect supportive activities for articular-cartilage repair21 e as well as destructive influences28. It was appreciated already at the beginning of the 20th century that if articular-cartilage lesions penetrate the layer of subchondral bone and the bone-marrow spaces e and as a consequence become Fig. 1. Light micrograph of vertically-sectioned articular cartilage, derived from the femoral of a 17-month-old bovine cow, illustrating its subdivision into su- perficial (S), transitional (T), upper-radial (UR), lower-radial (LR) and calcified-cartilage (C) zones; the latter abuts on the subchondral-bone plate (B). 1 mm-thick section, stained with Toluidine Blue. Bar: 200 mm.

The larger joints of the human body, for example, the knee, the hip and the shoulder, are most frequently affected by OA. During the early stages of the disease, a localized pattern of lesioning of the articular-cartilage layer is a characteristic occurrence11. However, even in unlesioned areas of the affected joint, the metabolism of the chondrocytes is disturbed12,13. Once initiated, the lesioning process is progressive, and no prophylactic (medicational) or e interventional measures are at hand to arrest it14 17. Likewise, no biologically-based treatment strategies are available to induce an efficacious healing of the structural defects. This review briefly surveys the biological basis of cartilage repair, as well as current trends in the treatment of articular-cartilage defects, and points out the misconceptional elements in these approaches. Particular emphasis is placed on research activities in the field of cartilage- tissue engineering. The major scientific and practical hurdles that need to be overcome for successful articular-cartilage repair are discussed; so, too, are the shortcomings of repair-evaluation pro- tocols. Suggestions for improvements in the analytical approach are made. Fig. 2. Schematic drawing of a normal synovial joint. 336 E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350 imbued with blood bearing diverse chondroprogenitor cells and type of cartilaginous repair tissue and lower incidences of tissue fibrin (which acts as the supportive scaffold of a blood clot) e a degeneration 12 months after surgery than did those who under- limited healing response may ensue26,29. But the repair tissue that went osteotomy alone. However, no difference in the clinical e is laid down by the blood-borne and bone-marrow-derived cells of outcome was observed after 2 (and up to 9) years45 49. diverse nature does not endure for more than a few months17,30,31, The idea of drilling holes through damaged areas of the and even if it persists for a longer period of time, it is deficient in articular-cartilage layer into the subchondral bone-marrow spaces e functional competence32 35. Despite this poor outlook, many of the was conceived by Pridie in 195939,50,51. As aforementioned, the surgical treatment strategies that are in clinical use today rationale behind this concept is to stimulate a spontaneous repair endeavour to foster this spontaneous repair reaction. Interventions reaction. But, perforce, the tissue that is formed is variable in of this kind are often categorized as “bone-marrow-stimulation” composition, structure and durability. The intervention is now e approaches (for review, see27,36 38). performed chiefly in young patients with osteoarthritic conditions, such as osteochondritis dissecans50. The aim is to bring symp- Current surgical strategies tomatic relief and to improve joint functionality52,53 by promoting a resurfacing of the articular layer with a fibrocartilaginous type of Treatment strategies that exploit the spontaneous repair response repair tissue. Clinical reports suggest that patients suffering from by a “stimulation of the bone marrow” generalized arthroses derive the greatest benefits from Pridie drilling54. However, this procedure appears to confer short-term These surgical approaches include abrasion chondroplasty22,29, benefits only, and only in patients of younger age; in the long-run Pridie drilling39 and microfracturing40 (recently refined to micro- (several years after surgery), these beneficial effects are lost55. drilling41,42). The aim of these techniques is to expose the lesioned The microfracturing technique e a modification of the Pridie cartilaginous tissue to the bone-marrow spaces, which, together approach e was elaborated by Steadman et al.40. After the removal with other vicinal compartments (such as the vascular and peri- of the calcified-cartilage layer, multiple holes, 1- to 2-mm in vascular spaces, the bony tissue itself, adipose tissue and the diameter and 3 to 5 mm apart, are created [with either a pointed synovium), are thereby stimulated. In essence, these interventions metallic (ice-pick-like) instrument40 or a narrow-calibre drill41,42] lead to a spontaneous repair response, which is based upon in the bone plate and the bony trabeculae. This process is associated surgically-induced bleeding from the subchondral-bone spaces and with bleeding and blood-clot formation (Fig. 3). The technique has subsequent blood-clot formation. The induction of such a sponta- been recently modified. The improved “microdrilling” version is neous repair response is well known from animal experiments. better controllable, more reproducible and less traumatic41,42,56. However, the ensuing healing response is highly variable and non- The microfracturing technique has thus developed into a “micro- reproducible, and the tissue that is formed is of a fibrous43, unen- Pridie” drilling approach and relies on the same biological princi- during nature26,30. These techniques were introduced several de- ples. The advantage of the latter technique was originally believed cades ago44, at which time no alternative or more efficacious to lie in the avoidance of the thermal necrosis that was thought strategies were available, and in the intervening years a consider- possible with abrasion and drilling. However, this concern has not able body of clinical experience has been gained with them. been confirmed either experimentally or clinically. Microdrilling is Various retrospective clinical investigations appertaining to now performed more widely than any other bone-marrow- e abrasion chondroplasty have been conducted45 48. The success stimulation technique. Albeit so, no prospective clinical trials rates are quite variable49 and depend upon many factors, such as have been undertaken that would either confirm or refute the the patient's age and physical-activity level, the severity of the claimed advantages of this over other bone-marrow-stimulation arthritic condition, and the follow-up period. One relevant clinical techniques. Microdrilling and microfracturing have been advo- trial has been conducted: patients suffering from OA and painful cated chiefly for young patients, in whom good results (improved knee conditions were subjected either to concomitant abrasion joint functionality and relief from pain in 60 to 80% of cases) have e chondroplasty and osteotomy (for realignment and pain relief) or to been reported56 59, probably owing to the larger numbers and the osteotomy alone. Individuals who underwent the combined higher activity-levels of the participating precursor-cell pools in approach manifested significantly higher levels of a “hyaline-like” these individuals, thereby leading to a more exuberant repair

Fig. 3. Schematic representation of the microfracturing technique. The floor of a chondral defect is perforated in several places using either a pointed instrument or a narrow- calibre drill. By this action, the marrow space of the subchondral bone is opened. Blood percolates into the defect e bearing with it local bone-marrow-derived MSCs e and therein forms a haematoma. E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350 337 response than in older ones. Since there is some evidence that lessen the burden on health-care systems, which would, in turn, microdrilling and microfracturing are most effective in patients render them more affordable to the average citizen in Western below 40 years of age60, it is questionable whether these tech- societies79. niques would be appropriate for OA-patients, who are usually older, whose spontaneous tissue-repair potential is impoverished, and in Transplantation strategies whom the number and the availability of bone-marrow-derived e stem cells is reduced61 63. Osteochondral autografting (mosaicplasty) With a view to improving the clinical outcome of microdrilling The idea of implanting chondral or osteochondral tissue itself and its reproducibility, this intervention has been combined with within articular-cartilage defects dates back to the beginning of the e the introduction of a thin-layered, blood-absorbing matrix (see last century80 82. And this concept still forms the basis of clinical e e Matrix scaffolds) of either collagen types I and III64,65 or chitosan strategies involving both autografts83 86 and allografts87 89. Of the glycerol phosphate41,66. However, available data afford little evi- autologous osteochondral treatment strategies currently in use, dence of any beneficial effect of this combined approach. The mosaicplasty (Fig. 4) is the one with the longest history. This disappointing results may reflect the relatively high bio- technique is used for large, full-thickness lesions, and involves the incompatibility of the implanted matrical material, which triggers introduction of several or multiple pieces (plugs) of autologous an inflammatory response. Indeed, when this combined approach graft-material into the defect void. The advantage of this treatment is adopted, the defect site is confronted not only with the extrinsic strategy is that it can be performed during a single intervention. matrix but also with the intrinsic one, namely, fibrin, which fills Upon exposure and surveillance of the diseased joint, the ortho- the area after the induction of bleeding, and which likewise triggers paedic surgeon is able to promptly decide from which regions an inflammatory response. Hence, inflammatory reactivity is autologous donor material should be removed and can then pro- augmented by the introduction of an extrinsic matrix. One possible ceed immediately with the autotransplantation (Fig. 4). Mosaic- way to improve the repair result that is achieved in the presence of plasty has for some years been the favoured technique of an extrinsic matrix is to enhance its biocompatibility and to mini- orthopaedic surgeons in Japan and Europe, but is of only limited mize its pro-inflammatory properties. Recent clinical studies that popularity in North America. Indeed, for various reasons (see have been conducted using a collagenous membrane with such below), the approach is now losing credibility among surgeons in e attributes have yielded fairly favourable results67 71. This type of all geographic regions. collagenous matrix is introduced into the defect void after a stim- One of the most serious drawbacks of the technique relates to ulation of the bone marrow by microdrilling. For this combined the destiny of the areas from which the autologous osteochondral approach, clinicians have coined the term autologous matrix- plugs are removed. In taking such tissue, the surgeon is artificially induced chondrogenesis (AMIC), which is a misleading misnomer, creating additional lesions within an already diseased joint (Fig. 4), since the carrier material is not in itself chondrogenic. The advan- and in so doing establishes additional sites of joint degeneration, tages of the AMIC-approach are that it involves no cell- thereby exacerbating rather than assuaging the local pathological transplantation, that it can be performed during a single surgical process. Given the not inconsiderable size of the osteochondral intervention, and that it is cost-effective. Prospective randomized plugs that are excised by the surgeon, the donor sites probably fail clinical trials of a comparative nature are currently underway to to heal. For this reason, some surgeons apply a matrical material to assess the practical usefulness of the AMIC-approach relative to the surgically-created lesions (donor sites), with a view to pro- that of ACI and/or matrix-associated ACI (MACI). moting a spontaneous repair response90. Similarly, we have but a A surer means of achieving an additional improvement in the poor conception of what occurs at the acceptor sites after the clinical outcome of microdrilling-based therapies would be to transplantation of autologous tissue-plugs. The drilling procedures introduce an on-the-site-prepared local population of autologous that are used to remove the osteochondral tissue expose chon- stem cells (either isolated or in the form of tissue-fragments), drocytes within the transplanted plug e as well as those bordering derived, for example, from the synovial membrane or the bone the cavity created at the donor site e to extremely high tempera- marrow. An approach that draws on the on-site preparation of tures and drying conditions, which are liable to impair their vitality. autologous material would involve fewer regulatory hurdles and The osteochondral plugs, once removed, are then press-fitted by circumvent prior laboratory processing, and would, therefore, be hammering into the subchondral-bone bed. This hammering pro- cost-effective. The reproducibility of the repair results could cess probably induces an injurious compression of the transplanted probably be improved by the introduction of a vehicle-borne cartilage91, which could precipitate the immediate death of the growth-factor cocktail to induce the desired chondrogenic differ- chondrocytic population91. Moreover, if the tissue is transferred entiation of the stem cells, which could be conceived as a from a low to a high-weight-bearing region of the joint, it will be e commercially-available, off-the-shelf product72 74, and which subjected to unphysiological conditions of mechanical loading for would have to be applied together with a functional or structural which it was not conceived, thereby accelerating its degeneration. barrier75,76 to prevent the up-growth of underlying bone into the The insertion of several serried plugs within the defect void, cartilaginous compartment. A concept that is based on the use of without taking measures to promote knitting between them, might fragments of synovial membrane would be the one most likely to also give rise to complications: the absence of structural bonding meet with success, since stem cells of synovial origin have a higher around the entire circumference of each plug will certainly pose chondrogenic potential than those derived from either the bone nutritional as well as functional problems from the outset, and marrow, the , adipose tissue or muscle, at least in vitro77. these will tend to prompt tissue degeneration (see Ref. 27). Only a Moreover, it would even be advantageous to introduce the cells in few attempts have so far been made to clarify these issues. Hence, the form of tissue-fragments rather than as isolates, since, when although mosaicplasty is still used, there exists no sound scientific supported by their physiological scaffold, their responsiveness to basis for the contention that patients subjected to this treatment e the applied chondrogenic differentiation factor(s) would be better strategy benefit in the long run92 98. assured78. New treatment strategies should be preferably conceived from the onset as single surgical interventions or Osteochondral allografting arthroscopic approaches e with a view to clinical practicability and For many decades now, allogeneic osteochondral grafts have to reduce the risk of infection or other complications, as well as to been used for the filling of articular-cartilage defects. The approach 338 E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350

Fig. 4. Schematic representation of the principle of mosaicplasty. Cylindrical plugs of osteochondral tissue are drilled from a non-weight-bearing area of the knee joint (using a special surgical instrument) and then press-fitted, side-by-side, within the damaged zone. The number of plugs that are removed will depend upon the surface diameter of the acceptor site. The denuded donor sites are either filled with osteochondral tissue that is removed from the damaged acceptor site during its surgical preparation for the plugs, or left empty. When viewed from above, the fitted plugs form a mosaic of circular profiles that are interspersed with empty spaces; hence the name “mosaicplasty”.

does not attempt to induce a cartilage-repair response, but repre- fact the brainchild of Grande et al., who tested it in a preclinical sents a means of substituting failed or lost tissue with healthy study with rabbits in 1989114. According to the ACI-technique, a articular cartilage, which is usually derived from cadavers80. biopsy of healthy articular cartilage is first arthroscopically har- The biological rationale for this approach is based on the vested from a low- or a non-load-bearing location of the diseased knowledge that large osteochondral defects do not heal sponta- joint. The cartilaginous tissue is then enzymatically digested to neously and that it is extremely difficult to elaborate a suitable release the chondrocytes, which are subsequently expanded in stimulatory system to promote effective repair in a compartment- culture, thereby generating more than 10 million cells from the few specific manner within the cartilaginous and the osseous carrels. hundred thousand that are contained within the biopsy. A sus- It has thus been deemed simpler to replace the lost tissue with pension of the expanded autologous chondrocytes is introduced e cadaverous osteochondral allografts99 101. Immunological prob- directly into the defect, beneath a surgically-sutured periosteal lems are inevitably to be counted amongst the drawbacks of this flap, which is usually removed from the medial tibia. Systems technique102,103. Nevertheless, patients with large osteochondral involving the application of freely-suspended cells are technically defects, such as those generated by tumour resection, osteonec- more complex than those in which matrix-embedded cells are rosis, extensive trauma, widespread OA or osteochondritis dis- grafted, since the careful placement and suturing of periosteal secans, benefit greatly from this treatment strategy104,105. flaps (or other types of material with a comparable function) are Clinical experience with this therapeutic option has been sur- required to ensure their retention and optimal integration with the prisingly good, immunological reactions being apparently less surrounding articular cartilage. Furthermore, the postoperative extensive in humans than in experimental animals103,106,107. Hu- delamination of such flaps e which can and does occur, even after man osteochondral allografts also survive for longer periods (some suturing115 e must be prevented by a temporary immobilization years), even after freezing or lyophilization87,88,102,108. Clinical (partial or complete) of the joint. More recently, a matrix- success rates have been reported to range from 65% to 85%, even associated variant of the ACI-approach (the so-called MACI-tech- e after monitoring periods of up to 10 years100,109 111. The major nique) was introduced116. According to this MACI-technique, the drawbacks that are associated with the implementation of this autologous chondrocytes are embedded within a matrix (e.g., methodology in clinical practice include the scarcity of fresh donor collagen or hyaluronan) rather than suspended in a fluid, thereby material and the storage of frozen tissue112. The small but ever- obviating the need for a periosteal flap. However, the use of a present risk of disease transmission must also be borne in mind111. matrix introduces a new set of problems, such as the issues of bioincompatibility, inflammation and degradation (see Trans- Autologous-chondrocyte implantation (ACI) plantation strategies, and Refs. 27,117). The ideal carrier118 has not yet been developed. Albeit so, this is still a goal towards which When the ACI-technique (Fig. 5) was introduced on the clinical researchers aspire, and efforts in this direction have not been scene by Brittberg et al. in 1994113, it took the orthopaedic world by thwarted by either the slowness of the progress or the set-backs of storm, generating much hope and enthusiasm. The concept was in failure. E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350 339

Fig. 5. Schematic representation of the ACI-technique. A fragment of articular cartilage is removed from the periphery of the joint that bears the lesion. This fragment of tissue is digested to release the chondrocytes, which are expanded in vitro for 11 to 21 days. A periosteal flap is then removed from the medial tibia and sutured over the lesion. A suspension of the expanded chondrocytes is injected beneath the flap into the lesion.

A considerable disadvantage of ACI-based strategies is that Almost unbelievably, this possibility has not been excluded in the surgical intervention is required on two separate occasions, which published studies by the establishment of the appropriate controls is a costly process necessitating long-term patient planning. The [viz., an “empty” defect (negative control): no injection of chon- creation of additional lesions within the joint during the harvesting drocytes beneath a periosteal flap114,116; and a defect filled with of healthy cartilage is also a drawback. Although only a small biopsy another cell type (positive control), e.g., fibroblasts]. is taken from a non-weight-bearing area of the joint, some in- Neither microfracturing/microdrilling nor ACI is clinically vestigators claim that this may heighten the risk for the develop- effective in patients over 50 years of age, probably because the ment of OA119,120. It has also been suggested that the chondrocytes bone-marrow-derived (mesenchymal) stem cells (microfracturing could be removed from osteoarthritic rather than from normal technique) and the transplanted autologous chondrocytes (ACI- articular cartilage, or even from other tissue sources121,122. technique) suffer age-related losses in their potential to proliferate Although the ACI-technique is still highly popular, its superiority and differentiate125. Hence, especially for elderly persons, who are to simpler interventions, such as microfracturing or microdrilling, particularly prone to osteoarthritic lesioning of the articular- has not been established. Knutsen et al.123 have published the re- cartilage layer, there exists an undisputed need for an effective sults of a prospective randomized clinical trial in which patients cell-based repair strategy. We have to face the bleak fact that very with circumscribed cartilaginous lesions in the femoral condyle little progress in this area has been made since the bone-marrow- were treated either conventionally, namely, by microfracturing, or stimulation technique e of which microfracturing/microdrilling is a according to the ACI-technique. The findings of this 5-year trial variant e was introduced by Pridie in the 1950s44. However, one revealed the latter strategy to confer no advantage over the con- source of comfort is that cell-based cartilage-repair strategies, such ventional one. Although the repair tissue that was evident 1 year as the ACI-technique, do not elicit worse healing results than those after treatment according to the ACI-technique was reported to be that are based on a stimulation of the bone marrow. A refinement of structurally superior to that laid down after microfracturing, the the ACI-technique has been recently introduced, which involves a clinical outcome was the same124. An ongoing long-term follow-up selection from the harvested cells of specific sub-populations of analysis of the same cohort of patients has confirmed the earlier (characterized) chondrocytes with particularly high chondrogenic (5-year) findings: 15 years after treatment by microfracturing or potentials. However, 2 years after treatment according to the according to the ACI-technique, the clinical results were the same in refined ACI-technique, the clinical benefits were reported to be only each group, which argues in favour of the less traumatic, less risky incremental126. And 5 years after treatment by microfracturing or and less costly microfracturing approach (Gunnar Knutsen: per- according to the refined ACI-treatment, the clinical outcome was sonal communication). A possible explanation for these short- and ultimately the same in each patient-group127, despite some differ- long-term findings is, that from a biological and biomechanistic ences during the follow-up course128. point of view, the treatments are basically the same. When the ACI- Given the high degree of technological finesse that is now technique is performed, the floor and the walls of the defect are achievable, and against the backdrop of knowledge and experience usually “neatened” by shaving, which induces local bleeding and a that have been gained over the past 60 years with Pridie drilling spontaneous repair response from the bone-marrow spaces. and over the last 20 to 30 years with the ACI-approach [as well as 340 E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350 with its variations/modifications (e.g., MACI)], both strategies are sections, the key constituents of a tissue-engineering-based accessible to improvement. As aforementioned, in an attempt to approach to cartilage repair are described. improve the clinical outcome of microdrilling and its reproduc- ibility, the approach has been combined with the introduction of a Cell sources thin-layered, blood-absorbing matrix (enhanced microfracturing) of either collagen types I and III64,65, “manipulated” collagen (as in Success in tissue-engineering-based strategies would obviate the AMIC67) or chitosan glycerol phosphate41,66. The available data need for the transplantation of cartilaginous tissue. And if appro- afford evidence that the clinical results are reproducible68,71, and, priate precursor-cell pools could be identified, then the numerous possibly, that the long-term outcome (compared to the conven- problems that are associated with the use of donor (allogeneic) tional technique) may be improved70. As heretofore mentioned, the material would be circumvented. Precursor-cell pools of diverse introduction of a local population of autologous stem cells (e.g., in tissue origin are known to exist within adult mammalian organisms, the form of small tissue-fragments, which could be derived from a each of which has the potential to differentiate into cartilage- local source, such as the synovium) would probably be a fruitful producing chondrocytes and which can be used for trans- route to pursue in the future. However, the currently practiced plantation purposes. And in some instances, the tissue of origin has approaches, which involve the transplantation of either fragments the potential for self-repair after the removal of not insubstantial e of juvenile allogeneic cartilage129 131 or pieces of adult autologous quantities of material. The tissue sources include the cambial layers e articular cartilage129,132 134, are based on the false premiss that the of the and the periosteum, the bone-marrow stroma, e cells can migrate out of the tissue. Tissue-embedded chondrocytes the synovial membrane, muscle, fat143 145 and adult cartilage itself e are incapable of migration; they can undergo only indirect trans- (autologous or allogeneic chondrocytes146 148). Perinatal cells149,as location via a directional resorption of the surrounding matrical well as foetal or embryonic stem cells and , can also be coat and its neoformation135. Migration is possible only if the cells employed150. The search for suitable and hitherto untested types e undergo dedifferentiation (which occurs during the late stages of continues unabated151 153. Due to the limited availability of autol- osteoarthritis) and if the pericellular and territorial matrical com- ogous chondrocytes, the focus is now more on allogeneic sources. e partments are destroyed by excessive catabolic activity136 138. However, the use of allogeneic cells raises other problematic issues, Clinical studies that have dealt with these approaches129,133 suffer such as bioincompatibility and immunogenicity142,152. from fundamental shortcomings, which include insufficient Mesenchymal stem cells (MSCs) are now frequently used numbers of patients, the absence of a basis for comparison (e.g., experimentally for cartilage-tissue engineering, owing to their ca- e sham surgery), and the failure to instigate random-selection pro- pacity for self-renewal and their accessibility143 145. Amongst the cedures. The data that have been gleaned from such trials serve various tissue sources, bone-marrow-derived MSCs have been the only to demonstrate that the interventions can be safely imple- most intensively investigated, both in vitro and in vivo, since the mented; they do not permit the drawing of comparative conclu- time of their original isolation by Friedenstein in 1966154. Bone- sions. The same arguments hold true for clinical studies that have marrow-derived MSCs evince a high capacity for chondrogenesis evaluated the efficacy of approaches involving the application of a only in the presence of appropriate stimulation factors78,155,156.Ina e bone-marrow-aspiration concentrate (BMAC)139 141 or of platelet- clinical setting, Kuroda et al.157 treated a large full-thickness artic- rich-plasma (PRP), either alone or in combination with ular-cartilage defect within the femoral condyle of an athlete with BMAC140,141. PRP- and BMAC-based strategies are classical examples autologous bone-marrow-derived stromal cells. The bone marrow of empirical, “alchemistic” approaches to the engineering of carti- was aspirated from the patient's iliac crest 4 weeks before surgery laginous tissue. Using the former approach (PRP), an ill-defined and the cells were expanded in culture after their separation from broth of multifarious growth factors at unknown concentrations the erythrocytes. The undifferentiated cells were then embedded and with diverse e antagonistic as well as synergistic e workings is within a collagenous gel, which was transferred to the articular- administered; and using the latter (BMAC), a similarly ill-defined cartilage defect and covered with an autologous periosteal flap. soup of diverse cells in variable numbers is applied. Owing to Although the knee-outcome scores improved with time, biopsies inter-individual variations in the compositions of these pot-pourris, revealed the presence of fibro-, not of hyaline, cartilage. In this the exerted effects will be likewise variable and thus irreproduc- study, no exogenous agents were applied to guide the course of ible; and even if these are positively rated, an identification of the differentiation. This fact suggests that the formation of a hyaline key-player(s) is impossible. type of repair cartilage by MSCs requires the presence of appro- priate stimulation factors (see Signalling molecules), which may be Current research activities relating to tissue-engineering- locally absent, or present at insufficiently high levels to guide the based strategies process of differentiation in the desired direction. Although MSCs have been claimed by some investigators to furnish their own Many more research teams around the world are now moving supply of growth factors158,159 and to be conferred with the ca- away from an overly empirical approach to cartilage repair, which pacity for immunomodulation160, experimental experience gained was hitherto the norm, towards a more biologically rational one. over the past 20 years in the field of cartilage repair has revealed Current activities tend to focus on the elaboration of novel tissue- these activities e if indeed they exist e to be too low to support an engineering-based strategies142. Tissue engineering can be defined efficacious healing response. as the art of reconstituting mammalian tissues, both structurally and One of the limitations of bone-marrow-derived MSCs is that functionally. Such reconstructive processes can be conducted (1) their chondrogenic potential declines with increasing patient entirely in vitro, (2) initially in vitro and then in vivo (in situ), or (3) age125. MSCs that are derived from the synovium do not suffer from entirely in vivo. this drawback161. Furthermore, the chondrogenic potential of MSCs Three key constituents usually form the building blocks of a originating from the synovium appears to be superior to that of tissue-engineering-based approach: a matrix scaffold, cells, and MSCs originating from either the bone marrow, the periosteum, signalling molecules (such as growth factors or genes). Most of the adipose tissue or muscle, at least in vitro77. However, as yet, we are experience so far gained with these techniques has been derived still in the dark as to which tissue source of MSCs is ideal for the from in-vitro studies, although experiments with laboratory ani- clinical repair of articular cartilage, since convincing in-vivo data mals and humans have also been conducted. In the following have not been forthcoming to date. E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350 341

Table I programming of joint-surface shape or of locational differences in Chemical classes of matrix structure, composition and thickness23,24, is lacking. Knowledge of 1. Protein-based polymers this kind is indispensable for the fruitful development of more Fibrin sophisticated epigenetic and higher-level approaches to cartilage Collagen repair. Only against a background of improved knowledge and Gelatine understanding can we hope to engineer a mechanically-competent 2. Carbohydrate-based polymers Polylactic acid and durable type of articular cartilage. At present, research in this Polyglycolic acid area has attained a hyatal status and is moving in circles rather than Hyaluronan forward. Agarose Alginate Chitosan Matrix scaffolds 3. Artificial polymers Dacron (polyethylene terephthalate) The use of matrices as cell-carriers in cartilage-repair strategies fl fl Te on (polytetra uoroethylene) dates back to the 1960s163. Many different types of matrix have fi Carbon bres e Polyesterurethane been tested not only in vitro but also in experimental animals and Polybutyric acid human patients e for their capacity to facilitate or promote Polyethylmethacrylate articular-cartilage repair. These matrices can be broadly categorized Hydroxyapatite according to their chemical nature into protein-based polymers, 4. Within/between classes carbohydrate-based ones, artificial materials, and combinations of Cross-linkage Chemical modulations these (Table I). Even when a naked matrix (viz., one that carries no Geometrical modifications (to produce fibrillar forms or foams) cells or signalling substances) is deposited within an articular- Matrix combinations cartilage defect, it must be considered as an information-carrying Reproduced e in a slightly modified form e with permission from30. device: its three-dimensional structure will, according to its peculiar geometrical configuration, specifically guide, or at least influence, the process of tissue growth, whilst the chemical and Irrespective of the type of stem cell that is employed, tremen- physical properties of its surface will impact cell attachment and dous hurdles must still be surmounted before the goal of cartilage the binding of growth factors in the niche environment. Hence, as regeneration can be achieved. Our knowledge of the regulatory and an “active biologic”, such a naked matrix will elicit a biological signalling pathways that are implicated in the biology of stem cells response within a bodily tissue-compartment, which may or may generally, and more especially that govern their commitment to not be desirable, but which cannot be prevented unless specific differentiate along a specific course and to form a specific type of modulations of an appropriate nature are made to guide the pro- cartilaginous tissue, namely, fibro-, hyaline or elastic, is still very cess in the desired direction [e.g., by means of AMIC (see discussion incomplete. Information appertaining to the temporal course of above)]. The basic requirements of a matrix to be used in a chondrogenic differentiation and its regulation is also lacking. With biological environment are summarized in Table II (see also respect to induced pluripotent stem (iPS) cells162, for example, it Refs. 27,117,118). still remains to be ascertained whether the point in time at which Although some of the listed materials are already in clinical they are induced to undergo committed differentiation along a use164, most of them, and especially the synthetic ones, are still specific course is or is not programmed into their genetic appa- being tested in preclinical trials165. Moreover, using scaffolding ratus; and, if so, whether the “age”, viz., maturation state, of the materials that have been applied for awhile in the field of cartilage newly-formed cells and tissues matches that of the donor. The same repair, such as collagen166, fibrin167, hyaluronan168,169 and poly- questions arise also when dealing with foetal, embryonic or adult lactate170, as well as those that have appeared on the scene more stem cells. Moreover, we are still in the dark as to whether the recently, attempts are now being made either to produce them in tissue-differentiation and growth processes that ontogenetically nanofibrillar forms171, to enhance their porosity172, or to manu- span decades in time can indeed be speeded up therapeutically so facture them in a state that is conducive to injection173. The com- as to be completed within the matter of a few weeks only. mon aim of these endeavours is usually to create an absorbable, Furthermore, although our knowledge of the human genetic code is biomimetic material174, even when using synthetic polymers, such now fairly complete, information relating, for example, to the as polydioxanone175.

Table II Matrix requirements

Matrix property Biological basis

1. Porosity Cell migration 2. Carrier Lodgement and release of signalling substances 3. Adhesion Cell attachment 4. Biodegradability Physiological remodelling 5. Volume stability Smooth surface contour of repair tissue flush with that of native articular cartilage 6. Biocompatibility Good contact with the native-tissue compartment 7. Bonding Enhances interfacial integration between collagenous fibrils in repair- and native-tissue compartments 8. Internal cohesiveness Prevention of matrix outflow 9. Elasticity Resiliency during and following dynamic or static deformation 10. Structural anisotropy Promotion of native anisotropic tissue organization

Matrix properties specific to the mode of surgical application A. Fluid state during application with subsequent solidification in situ Arthroscopic implantation B. Stiff and amenable to press-fitting Arthrotomy (open surgery of a joint)

Reproduced e in a slightly modified form e with permission from30. 342 E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350

More recently, it has become the vogue to use biphasic scaf- There are three possible approaches to the engineering of folds176,177. These bilayers consist of two different materials: an articular cartilage. According to the first, the repair tissue is engi- upper one that is deemed to be conducive to chondrogenesis and neered completely in vitro, the fully-differentiated construct being which is destined for the cartilaginous compartment of osteochon- then implanted within the defect void. The advantage of this dral defects, and a lower one of a ceramic-like substance, which can approach lies in the fact that cell metabolism and differentiation be press-fitted as a plug into the subchondral bone176. Apart from the are subject to better control in vitro than in vivo, provided that the obvious issues that must be addressed when testing a scaffold, such appropriate bioreactor systems195,196, growth factors and delivery as biocompatibility, biodegradability and toxicity, its integration systems are available. Its drawbacks are, that the problems asso- with native cartilage is also a troublesome problem, and one that has ciated with the integration and the mechanical fixation of the not yet been overcome178. During the past decade, no matrices with repair tissue cannot be readily anticipated and solved. Moreover, basically new chemical features have been inaugurated. However, the simulated mechanical-loading conditions of the extracellular physical or chemical modifications to existing materials have been matrix may not be ideally suited to the specific needs of the pro- effected, with a view to enhancing the efficacy of clinically- spective repair site. In addition, biocompatibility and immunolog- established methodologies, such as microfracturing/micro- ical problems are frequently associated even with this in-vitro e drilling41,64 66, by AMIC68,70, or ACI (by MACI)116, or, in the context of approach and in the absence of a scaffold197. Furthermore, the more experimental tissue-engineering-based approaches, with the natural curvature of the joint surface poses a challenge to the aim of guiding cartilage-formation in a spatially-defined manner process of press-fitment whereby the engineered construct is [three-dimensional bioprinting (see discussion below)]. The dearth introduced into the defect void. of genuine innovation in this field partially reflects the unfavourable The second approach, which is more frequently adopted, aims to investment climate that prevails in the commercial world, which engineer only the basic building block, namely, a matrix scaffold encourages low-cost, incremental improvements in existing stra- containing a homogeneous population of cells, and signalling mol- tegies rather than revolutionary novelty. This is a regrettable ecules that are entrapped within an appropriate delivery system. circumstance, since it is precisely this flux of ingenuity that is now The vehicle-bound signalling molecules ensure that the desired needed to push research in a really fruitful direction. However, one differentiation process takes place in a controlled and timely positive aspect of this dilemmatic situation is that any newly- manner in vivo and that chondrocytic activity within the repair conceived therapeutic principle must now be not only of a truly tissue is sustained. According to this approach, the differentiation impressive nature but also backed up by rigorous experimentation and the remodelling of the repair tissue occur in vivo under physi- before any willingness to invest in clinical trials can be expected. ological conditions of mechanical loading. Repair tissue that is formed in situ is more likely to adhere to and integrate with native Signalling molecules articular cartilage than is that produced in vitro, and it will also adapt naturally to the contour of the synovial joint, provided that appro- One of the trends now foremost in the field of tissue engineering priate measures are taken to promote the bonding of the implant e relates to the elaboration of constructs that consist not only of a that has been generated in culture198 200. One of the disadvantages matrix and cells but also of signalling molecules to specifically of this “second” approach is that cell activity is more difficult to guide the course of differentiation in the desired direction. In most control on a long-term basis. Moreover, appropriate measures must cartilage-engineering strategies, the chosen cell type is applied be taken to avoid “contamination” from cells and signalling sub- together with free or appropriately-encapsulated growth factors. stances that are involved in the spontaneous healing response, since The efficacy of various growth factors to induce chondrogenesis has the tissue thereby formed would compromise the quality and the been evaluated mainly in vitro and to only a limited degree in vivo mechanical competence of the final repair-composite. (for reviews, see Refs. 179e183). In-vitro experiments have involved The third approach involves the direct application of exogenous various combinations of matrix, cell type and growth factors. These growth factors (entrapped within a matrix) to the defect site; these include collagenous matrices containing bone-marrow-derived then stimulate the intrinsic formation of cartilaginous tissue in stromal cells and transforming growth factor (TGF)-ß1184,185, situ72. An appropriate matrix is first introduced into the defect to agarose matrices containing chondrocytes and fibroblast growth define the space that is to be repaired. The laying of this physical factor (FGF)-2186, fibrinous matrices containing chondrocytes and “track” is necessary to guide the movements of the intrinsic pre- insulin-like growth factor (IGF)-1187, and polylactatous matrices cursor cells, which have a limited spatial awareness21. The growth containing perichondrial cells and TGF-ß1188. In some instances, factors are usually introduced into the matrix in two different states: MSCs have been cultured in the absence of a matrix (viz., under as a freely-soluble agent e to stimulate the immediate recruitment scaffold-free conditions), namely, as high-density pellets (or ag- of the intrinsic population of precursor cells from their site of origin, gregates)183,189,190. Under such conditions, not only growth factors, their migration into the defect area, and their proliferation therein21 but also dexamethasone, a synthetic glucocorticoid, is frequently e and in a vehicle-bound form for gradual delivery at a steady rate applied. However, this agent can elicit undesired effects in a that is sustained for several weeks e to stimulate the chondrogenic manner that depends upon the source of the cultured cells and the differentiation of the defect-filling population of cells72. Hence, an absence or presence of the natural (physiological) extracellular intelligent, internally-programmed matrix is applied to the defect. matrix78,155,156. Successful testing in vivo has been achieved using The freely-soluble signalling agent that is applied is usually IGF-1, equine191 and mature miniature-pig models72. The study that was bFGF or a TGF-b (at low concentration). As aforementioned, the conducted using the latter model has served to show how different growth factor in this form stimulates the recruitment of precursor the in-vivo situation may be from the in-vitro one. Under in-vitro cells from the synovium21, their migration into the defect space, and conditions, various types of growth factor have been shown to their subsequent proliferation therein. The vehicle-bound signalling support chondrogenic activities192, whereas in vivo, principally only agent that is applied to induce the chondrogenic differentiation of those of the TGF-b superfamily are effective193. A recently- the defect-filling population of cells is usually a member of the TGF- discovered compound, namely, kartogenin194, is now attracting b superfamily, for example, bone morphogenetic protein (BMP)-2, some attention. However, this molecule appears to exert a gener- BMP-9, BMP-13 or TGFb1 (at high concentration). Various drug- alized anabolic effect on chondrocytes rather than being specif- delivery systems have been employed, including microspheres, li- ically chondrogenic. posomes, microparticles and fibrillar networks. The principle E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350 343 underlying this approach was first published by our group in In this review, suggestions for fruitful directions of pursuit in the 1991201. A full description of the strategy appeared in the literature future have been made. But what is now needed is a revolutionary in 199621 and then, in an optimized form, in 200172. The approach turnaround in our conception of cartilage repair and a more thor- has since been summarized in several publications27,202. The prin- ough and systematic approach to its evaluation. We should not ciple has been recently confirmed by an independent group of in- stagnate, but break free of the bounds of our comfort zone as vestigators203 (2010) using TGF-b3 as the chemotactic and represented by the old well-worn ruts e the trusted, tried and chondrogenic differentiation agent, a hydrogel as the drug-release tested tracks of yore e the pursuit of which can contribute no more system, and three-dimensional-printed poly-ε-caprolactone-hy- than incremental or cosmetic information to the body of existing droxyapatite as the matrix. Unfortunately, the authors chose not to data that has accumulated over the past decade. cite the original pioneering work on this topic and coveted the idea A fundamental change in the general attitude of scientists, PhD- as a new one. Actually, the novelty of the study lay in the use of poly- students and supervisors towards the evaluation of cartilage repair ε-caprolactone-hydroxyapatite as a three-dimensionally-printed is also called for. In many laboratories, the analytical process is matrix. A matrix of this kind is implemented with a view to guiding conducted on an elementary e almost routine e and insufficiently tissue formation in a spatially-defined manner. As appertaining to discriminative level. Nowadays, repair cartilage is all too commonly articular cartilage, the aim is to reproduce the anisotropic organi- presented as a tissue that contains lacuna-mantled cells (chon- zation of the chondrocytes and their extracellular matrix that is so drocytes), that stains histochemically with a cationic dye (such as characteristic of the mature tissue and that is indispensable for its Toluidine Blue or Safranin O), and that manifests immunohisto- mechanical competence. Although three-dimensional bioprinting is chemical reactivity for type-II collagen. Attempts to quantify the an old technique (for a review of the topic, see Ref. 204), it has characteristics of the tissue are confined to parameters that insuf- recently undergone a revival: owing to technological advancements, ficiently describe the true nature of articular cartilage. These pa- sophisticated materials can nowadays be manufactured with rameters are gauged using subjective and biased scoring systems, greater facility and more cheaply than heretofore. such as those propounded by Mankin215, O'Driscoll216 and the The material of which the matrix is formed needs to be suffi- ICRS217. The parameters that are evaluated using these tools afford ciently porous to allow the infiltration of cells27,201 and also biode- no information relating to the maturation status of the repair gradable. Ideally, it should also have a well-defined anisotropic cartilage, viz., whether it is of a foetal, ontogenetically-transient, organization, which forces the cells to align themselves into vertical early postnatal or adult type; nor is any information gleaned as to columns during the process of remodelling205 and which is whether the repair cartilage is of a joint-specific type or not. important for the mechanical competence of the tissue as a whole, Similarly, these rudimentary evaluations yield no information as aforementioned. In this context, the three-dimensionally-printed respecting the metabolic status of the tissue, viz., whether the matrix that represented the sole novelty of the study by Lee et al.203 anabolic and the catabolic activities are in a state of equilibrium or may be advantagenous. not. Very often, the repair cartilage is sweepingly designated as One problem appertaining to the chondrogenic differentiation being “hyaline-like” without adequate supportive evidence. The of the defect-filling population of cells that we have recently repair cartilage needs to be subjected to a rigorous analysis: its addressed relates to the timely arrestation of the process prior to maturation status should be defined; so, too, should its architec- the stage of terminal hypertrophy and matrix mineralization73. This tonic and ultrastructural features, including the degree of anisot- undesired event can be avoided by the addition of TGF-b1 at a low ropy. These and other structural characteristics should be concentration73. Other investigators have achieved similar results quantified morphometrically, not using subjective scoring systems. e using alternative agents206 212. Another drawback of scoring-system-based approaches is that Consequently, to achieve optimal cartilage-repair results, mini- they attempt to assess too many parameters, whereas ideally, three mally three sets of growth-factor-stimulated “activities” need to be to maximally four pertinent ones should be evaluated. The impact e embraced in the applied cocktail of signalling agents72 74. This ne- of a repair strategy on the individual parameters can vary greatly cessity, which is not always recognized (cf203), renders the approach (ranging from positive to negative), and yet, in the end, an average unattractive to cost-conscious commercial parties and/or investors. score for all is calculated. Hence, there is the danger that positive Principally two novel growth-factor-based therapeutic strate- findings for some parameters will be “diluted” by negative effects gies are now at the conceptual stage of development: the afore- on others, and, in extreme cases, that falsely-negative results will mentioned one, which is designed to recruit cells with be generated; the reverse situation also holds true. Unfortunately, chondrogenic potential from a local intrinsic source, such as the one cannot escape from the reality that scoring-system-based ap- synovial membrane21,72,203; and a second approach, which involves proaches are subjective and biased, since they attempt to describe the injection into the defect site of MSCs that have been transfected the situation that appertains within the volume of a defect void with genes encoding the factors necessary for chondroprogenitor from two-dimensional images of histological sections without commitment213,214. Both approaches are still at a preliminary stage respecting the laws of random sampling and stereology. And it is of development; exhaustive short- and long-term preclinical precisely these laws that a morphometric analysis does respect. The studies79 are requisite prior to the launching of clinical trials. authors feel the necessity of dissuading investigators from imple- menting a scoring-system-based approach, which may not “go Scientific evaluation down well” with those who have used such regimes for many years. However, “a man should never be ashamed to own he has been After perusing this review article, the reader will no doubt be wrong, which is but saying, in other words, that he is wiser today left with the impression that in the area of cartilage repair, research than he was yesterday” (Alexander Pope: Miscellanies). In- activity over the past 10 years or so has turned around in circles, not vestigators should not be daunted by the prospect of undertaking a moved forward in excitingly new and fruitful directions e which is morphometric analysis. Once the basic principles of the approach a regrettable situation for hopeful patients. The circumstance is the have been grasped (for a simple introduction to these, see Ref. 218), less excusable in so far as the technological advancements that have all that is needed for an objective and unbiased evaluation of a few been made during this time span have greatly aided the probing of pertinent parameters are correctly-sampled tissue-sections and an many facets of cartilage biology, thereby permitting an enrichment appropriate grid-system. The investigator's task is then a simple of knowledge and understanding in the targeted disciplines. “counting” one; moreover, he or she is not burdened with the 344 E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350 responsibility of “judgement”, which is the case when a scoring Jakob and Gunnar Knutsen for their valuable input respecting the system is implemented. critical analysis of clinical studies. In addition to structural data, information appertaining to the post-translational metabolic status of the chondrocytes should be derived by monitoring the gene-expression levels of representative References anabolic and catabolic markers. The conception of truly novel therapeutic approaches to carti- 1. Hunziker EB. Articular cartilage structure in humans and lage repair is greatly encouraged. But all the ingenuity in the world experimental animals. In: Kuettner KE, Schleyerbach R, will be as wind-blown chaff unless we make a stand against the Peyron JG, Hascall VC, Eds. Articular Cartilage and Osteoar- quick-and-easy attitude that is now sadly enough the brand of thritis. New York: Raven Press; 1992:183e99. modern times. 2. Buckwalter JA, Mankin HJ. Articular cartilage. 2: degeneration and osteoarthrosis, repair, regeneration, and transplantation. Prospects for the future J Bone Joint Surg Am 1997;79A:612e32. 3. Gilbert JE. Current treatment options for the restoration of Current trends in the induction of cartilage repair seem to point articular cartilage. Am J Knee Surg 1998;11:42e6. towards a more tissue-engineering-based approach. The aim of 4. Messner K, Gillquist J. Cartilage repair. A critical review. Acta tissue engineering is to develop methodologies whereby bodily Orthop Scand 1996;67:523e9. defects can be repaired in such a manner as to restore the structure 5. Chaganti RK, Lane NE. Risk factors for incident osteoarthritis and the functional competence of the affected tissues. Tissues and of the hip and knee. Curr Rev Musculoskelet Med 2011;4: organs that do not heal spontaneously are induced to repair by 99e104. attempting to overcome the biological limitations that undermine 6. Monira Hussain S, Wang Y, Cicuttini FM, Simpson JA, this process in Nature. At the stage of its conception, a tissue- Giles GG, Graves S, et al. Incidence of total knee and hip engineering-based principle must be screened to ascertain the replacement for osteoarthritis in relation to the metabolic size of the patient population that will be targeted, its socioeco- syndrome and its components: a prospective cohort study. nomic impact, the feasibility and likely cost of its manufacture, and Semin Arthritis Rheum 2013;43:429e36. its surgical practicability. Hence, tissue engineering is an interdis- 7. Odum SM, Springer BD, Dennos AC, Fehring TK. National ciplinary enterprise79. obesity trends in total knee arthroplasty. J Arthroplasty There exists no magical formula for the engineering of a product 2013;28:148e51. that would be universally applicable to all bodily tissues. An engi- 8. Jones G. Osteoarthritis: where are we for pain and therapy in neered construct must be tailor-made on a tissue-specific basis. To 2013? Aust Fam Physician 2013;42:766e9. yield a product that will be beneficial to the targeted patient pop- 9. Haviv B, Bronak S, Thein R. The complexity of pain around the ulation, a tissue-engineering-based principle should be experi- knee in patients with osteoarthritis. Isr Med Assoc J 2013;15: mentally developed along systematic and, whenever possible, 178e81. rational lines, and in the light of a thorough understanding of the 10. Malfait AM, Schnitzer TJ. Towards a mechanism-based biological system in hand. approach to pain management in osteoarthritis. Nat Rev Rheumatol 2013;9:654e64. 11. Aigner T, Soder S. Typing, grading and staging of osteoar- Conclusion thritis: histopathological assessment of joint degeneration. Z Rheumatol 2008;67. 32e36, 38-40. We are still a long way from achieving our goal of cartilage 12. Aigner T, Vornehm SI, Zeiler G, Dudhia J, von der Mark K, regeneration. However, Nature affords the clues to this end if they Bayliss MT. Suppression of cartilage matrix gene expression are sought, and if we have the fortitude to pursue them rationally in upper zone chondrocytes of osteoarthritic cartilage. and systematically. Only by following such a course can we hope for Arthritis Rheum 1997;40:562e9. ultimate success in the generation of an endurable, structurally and 13. Chambers MG, Bayliss MT, Mason RM. Chondrocyte cytokine functionally competent type of cartilaginous repair tissue. and growth factor expression in murine osteoarthritis. Osteoarthritis Cartilage 1997;5:301e8. Contributions 14. Lefkoe TP, Trafton PG, Ehrlich MG, Walsh WR, Dennehy DT, Barrach HJ, et al. An experimental model of femoral condylar Conception and design: EBH, KL, MK, NS. defect leading to osteoarthrosis. J Orthop Trauma 1993;7: Analysis and interpretation of the data: EBH. 458e67. Drafting of the article: EBH, KL, MK, NS. 15. Gratz KR, Wong BL, Bae WC, Sah RL. The effects of focal Critical revision of the article for important intellectual content: articular defects on intra-tissue strains in the surrounding EBH. and opposing cartilage. Biorheology 2008;45:193e207. Final approval of the article: EBH, KL, MK, NS. 16. Hunziker EB, Stahli A. Surgical suturing of articular cartilage Obtaining of funding: EBH. induces osteoarthritis-like changes. Osteoarthritis Cartilage Administrative, technical, or logistic support: EBH. 2008;16:1067e73. Collection and assembly of data: EBH. 17. Simon TM, Aberman HM. Cartilage regeneration and repair testing in a surrogate large animal model. Tissue Eng Part B Competing interests Rev 2009;16:65e79. The authors have no conflicts to declare. 18. Madry H, Luyten FP, Facchini A. Biological aspects of early osteoarthritis. Knee Surg Sports Traumatol Arthrosc 2011;20: Acknowledgements 407e22. 19. Grenier S, Bhargava MM, Torzilli PA. An in vitro model for the The authors would like to thank Dr Yoshihiro Suzuki for his help pathological degradation of articular cartilage in osteoar- with the artwork, and are indebted to Hans Jorg€ Hauselmann,€ Roland thritis. J Biomech 2013;47:645e52. E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350 345

20. Oakley SP, Lassere MN, Portek I, Szomor Z, Ghosh P, 39. Insall J. The Pridie debridement operation for osteoarthritis of Kirkham BW, et al. Biomechanical, histologic and macro- the knee. Clin Orthop Relat Res 1974:61e7. scopic assessment of articular cartilage in a sheep model of 40. Steadman JR, Rodkey WG, Singleton SB, Briggs KK. Micro- osteoarthritis. Osteoarthritis Cartilage 2004;12:667e79. fracture technique for full-thickness chondral defects: tech- 21. Hunziker EB, Rosenberg LC. Repair of partial-thickness de- nique and clinical results. Operative Tech Orthop 1997;7: fects in articular cartilage: cell recruitment from the synovial 300e4. membrane. J Bone Joint Surg Am 1996;78:721e33. 41. Hoemann CD, Sun J, McKee MD, Chevrier A, Rossomacha E, 22. Kim HK, Moran ME, Salter RB. The potential for regeneration Rivard GE, et al. Chitosan-glycerol phosphate/blood implants of articular cartilage in defects created by chondral shaving elicit repair integrated with porous sub- and subchondral abrasion. An experimental investigation in chondral bone in microdrilled rabbit defects. Osteoarthritis rabbits. J Bone Joint Surg Am 1991;73:1301e15. Cartilage 2007;15:78e89. 23. Hunziker EB, Quinn TM, Hauselmann HJ. Quantitative struc- 42. Chen H, Sun J, Hoemann CD, Lascau-Coman V, Ouyang W, tural organization of normal adult human articular cartilage. McKee MD, et al. Drilling and microfracture lead to different Osteoarthritis Cartilage 2002;10:564e72. bone structure and necrosis during bone-marrow stimulation 24. Quinn TM, Hauselmann HJ, Shintani N, Hunziker EB. Cell and for cartilage repair. J Orthop Res 2009;27:1432e8. matrix morphology in articular cartilage from adult human 43. Furukawa T, Eyre DR, Koide S, Glimcher MJ. Biochemical knee and ankle joints suggests depth-associated adaptations studies on repair cartilage resurfacing experimental defects to biomechanical and anatomical roles. Osteoarthritis Carti- in the rabbit knee. J Bone Joint Surg Am 1980;62:79e89. lage 2013;24:1904e12. 44. Pridie KH. A method of resurfacing osteoarthritic knee joints. 25. Hunziker EB, Rosenberg LC. Articular cartilage repair. In: J Bone Joint Surg Br 1959;41:618e9. McCarty DJ, Koopman WJ, Eds. Arthritis and Allied Conditions 45. Chen FS, Frenkel SR, Di Cesare PE. Repair of articular cartilage e A Textbook of Rheumatology. Philadelphia: Lea & Febiger; defects: part II. Treatment options. Am J Orthop (Belle Mead 1997:2027e38. NJ) 1999;28:88e96. 26. Shapiro F, Koide S, Glimcher MJ. Cell origin and differentia- 46. Moseley Jr JB, Wray NP, Kuykendall D, Willis K, Landon G. tion in the repair of full-thickness defects of articular carti- Arthroscopic treatment of osteoarthritis of the knee: a pro- lage. J Bone Joint Surg Am 1993;75:532e53. spective, randomized, placebo-controlled trial. Results of a 27. Hunziker EB. Articular cartilage repair: basic science and pilot study. Am J Sports Med 1996;24:28e34. clinical progress. A review of the current status and pros- 47. Singh S, Lee CC, Tay BK. Results of arthroscopic abrasion pects. Osteoarthritis Cartilage 2002;10:432e63. arthroplasty in osteoarthritis of the knee joint. Singapore 28. Bromley M, Woolley DE. Histopathology of the rheumatoid Med J 1991;32:34e7. lesion. Identification of cell types at sites of cartilage erosion. 48. Haspl M, Pecina M. [Treatment of gonarthrosis with arthro- Arthritis Rheum 1984;27:857e63. scopic abrasion]. Lijec Vjesn 1995;117:236e40. 29. Altman RD, Kates J, Chun LE, Dean DD, Eyre D. Preliminary 49. Bert JM. Role of abrasion arthroplasty and debridement in the observations of chondral abrasion in a canine model. Ann management of osteoarthritis of the knee. Rheum Dis Clin Rheum Dis 1992;51:1056e62. North Am 1993;19:725e39. 30. Jackson DW, Lalor PA, Aberman HM, Simon TM. Spontaneous 50. Muller B, Kohn D. [Indication for and performance of articular repair of full-thickness defects of articular cartilage in a goat cartilage drilling using the Pridie method]. Orthopade model. A preliminary study. J Bone Joint Surg Am 2001;83-A: 1999;28:4e10. 53e64. 51. Insall JN. Intra-articular surgery for degenerative arthritis of 31. Dell'accio F, Vincent TL. Joint surface defects: clinical course the knee. A report of the work of the late K. H. Pridie. J Bone and cellular response in spontaneous and experimental Joint Surg Br 1967;49:211e28. lesions. Eur Cell Mater 2010;20:210e7. 52. Goldman RT, Scuderi GR, Kelly MA. Arthroscopic treatment of 32. Metsaranta M, Kujala UM, Pelliniemi L, Osterman H, Aho H, the degenerative knee in older athletes. Clin Sports Med Vuorio E. Evidence for insufficient chondrocytic differentia- 1997;16:51e68. tion during repair of full-thickness defects of articular carti- 53. Beiser IH, Kanat IO. Subchondral bone drilling: a treatment lage. Matrix Biol 1996;15:39e47. for cartilage defects. J Foot Surg 1990;29:595e601. 33. Guilak F, Butler DL, Goldstein SA. Functional tissue engi- 54. Pedersen MS, Moghaddam AZ, Bak K, Koch JS. The effect of neering: the role of biomechanics in articular cartilage repair. bone drilling on pain in gonarthrosis. Int Orthop 1995;19: Clin Orthop Relat Res 2001:S295e305. 12e5. 34. Schmitt LC, Quatman CE, Paterno MV, Best TM, Flanigan DC. 55. Schmidt H, Hasse E. Arthroscopic surgical treatment of cir- Functional outcomes after surgical management of articular cumscribed cartilage damage with spongiolization or Pridie cartilage lesions in the knee: a systematic literature review to drilling. Beitr Orthop Traumatol 1989;36:35e7. guide post-operative rehabilitation. J Orthop Sports Phys 56. Sledge SL. Microfracture techniques in the treatment of Ther 2014:1e53. osteochondral injuries. Clin Sports Med 2001;20:365e77. 35. McMahon LA, O'Brien FJ, Prendergast PJ. Biomechanics and 57. Steadman JR, Briggs KK, Rodrigo JJ, Kocher MS, Gill TJ, mechanobiology in osteochondral tissues. Regen Med Rodkey WG. Outcomes of microfracture for traumatic chon- 2008;3:743e59. dral defects of the knee: average 11-year follow-up. 36. Buckwalter JA. Articular cartilage: injuries and potential for Arthroscopy 2003;19:477e84. healing. J Orthop Sports Phys Ther 1998;28:192e202. 58. Mithoefer K, Williams 3rd RJ, Warren RF, Potter HG, 37. Martin JA, Buckwalter JA. Aging, articular cartilage chon- Spock CR, Jones EC, et al. The microfracture technique for the drocyte senescence and osteoarthritis. Biogerontology 2002; treatment of articular cartilage lesions in the knee. A pro- 3:257e64. spective cohort study. J Bone Joint Surg Am 2005;87: 38. Buckwalter JA, Brown TD. Joint injury, repair, and remodel- 1911e20. ing: roles in post-traumatic osteoarthritis. Clin Orthop Relat 59. Asik M, Ciftci F, Sen C, Erdil M, Atalar A. The microfracture Res 2004:7e16. technique for the treatment of full-thickness articular 346 E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350

cartilage lesions of the knee: midterm results. Arthroscopy 77. Sakaguchi Y, Sekiya I, Yagishita K, Muneta T. Comparison of 2008;24:1214e20. human stem cells derived from various mesenchymal tissues: 60. Kreuz PC, Erggelet C, Steinwachs MR, Krause SJ, Lahm A, superiority of synovium as a cell source. Arthritis Rheum Niemeyer P, et al. Is microfracture of chondral defects in the 2005;52:2521e9. knee associated with different results in patients aged 40 78. Shintani N, Hunziker EB. Differential effects of dexametha- years or younger? Arthroscopy 2006;22:1180e6. sone on the chondrogenesis of mesenchymal stromal cells: 61. Caplan AI. Mesenchymal stem cells. J Orthop Res 1991;9: influence of microenvironment, tissue origin and growth 641e50. factor. Eur Cell Mater 2011;22:302e20. 62. Muraglia A, Cancedda R, Quarto R. Clonal mesenchymal 79. Hunziker E, Spector M, Libera J, Gertzman A, Woo SL, progenitors from human bone marrow differentiate in vitro Ratcliffe A, et al. Translation from research to applications. according to a hierarchical model. J Cell Sci 2000;113(Pt 7): Tissue Eng 2006;12:3341e64. 1161e6. 80. Axhausen G. Die histologischen und klinischen Gesetze der 63. Pittenger MF, Mosca JD, McIntosh KR. Human mesenchymal freien Osteoplastik auf Grund von Tierversuchen. Arch Klin stem cells: progenitor cells for cartilage, bone, fat and stroma. Chir 1909;88:23e145. Curr Top Microbiol Immunol 2000;251:3e11. 81. Lexer E. Substitution of whole or half joints from freshly 64. Gille J, Schuseil E, Wimmer J, Gellissen J, Schulz AP, Behrens P. amputated extremites by free plastic operation. Surg Gynecol Mid-term results of Autologous Matrix-Induced Chondro- Obstet 1908;6:601e7. genesis for treatment of focal cartilage defects in the knee. 82. Beresford WA. Chondroid Bone, Secondary Cartilage and Knee Surg Sports Traumatol Arthrosc 2010;18:1456e64. Metaplasia. Baltimore-Münich: Urban & Schwarzenburg; 65. Benthien JP, Behrens P. The treatment of chondral and 1981. osteochondral defects of the knee with autologous matrix- 83. Brent B. Auricular repair with autogenous rib cartilage grafts: induced chondrogenesis (AMIC): method description and two decades of experience with 600 cases. Plast Reconstr recent developments. Knee Surg Sports Traumatol Arthrosc Surg 1992;90:355e74 (Discussion 375e356). 2011;19:1316e9. 84. Hangody L, Kish G, Modis L, Szerb I, Gaspar L, Dioszegi Z, et al. 66. Marchand C, Chen G, Tran-Khanh N, Sun J, Chen H, Mosaicplasty for the treatment of osteochondritis dissecans Buschmann MD, et al. Microdrilled cartilage defects treated of the talus: two to seven year results in 36 patients. Foot with thrombin-solidified chitosan/blood implant regenerate Ankle Int 2001;22:552e8. a more hyaline, stable, and structurally integrated osteo- 85. Bobic V. Autologous osteo-chondral grafts in the manage- chondral unit compared to drilled controls. Tissue Eng Part A ment of articular cartilage lesions. Orthopade 1999;28: 2012;18:508e19. 19e25. 67. Steinwachs MR, Guggi T, Kreuz PC. Marrow stimulation 86. Girdler NM. Repair of articular defects with autologous techniques. Injury 2008;39(Suppl 1):S26e31. mandibular condylar cartilage. J Bone Joint Surg Br 1993;75: 68. Anders S, Volz M, Frick H, Gellissen J. A randomized, 710e4. controlled trial comparing autologous matrix-induced chon- 87. Bakay A, Csonge L, Papp G, Fekete L. Osteochondral resur- drogenesis (AMIC(R)) to microfracture: analysis of 1- and 2- facing of the knee joint with allograft. Clinical analysis of 33 year follow-up data of 2 Centers. Open Orthop J 2013;7: cases. Int Orthop 1998;22:277e81. 133e43. 88. Bell RS, Davis A, Allan DG, Langer F, Czitrom AA, Gross AE. 69. Wiewiorski M, Miska M, Kretzschmar M, Studler U, Bieri O, Fresh osteochondral allografts for advanced giant cell tumors Valderrabano V. Delayed gadolinium-enhanced MRI of at the knee. J Arthroplasty 1994;9:603e9. cartilage of the ankle joint: results after autologous matrix- 89. Czitrom AA, Keating S, Gross AE. The viability of articular induced chondrogenesis (AMIC)-aided reconstruction of cartilage in fresh osteochondral allografts after clinical osteochondral lesions of the talus. Clin Radiol 2013;68: transplantation. J Bone Joint Surg Am 1990;72:574e81. 1031e8. 90. Bartha L, Hamann D, Pieper J, Peters F, Riesle J, Vajda A, et al. 70. Bark S, Piontek T, Behrens P, Mkalaluh S, Varoga D, Gille J. A clinical feasibility study to evaluate the safety and efficacy Enhanced microfracture techniques in cartilage knee surgery: of PEOT/PBT implants for human donor site filling during fact or fiction? World J Orthop 2014;5:444e9. mosaicplasty. Eur J Orthop Surg Traumatol 2013;23:81e91. 71. Dhollander A, Moens K, Van der Maas J, Verdonk P, 91. Quinn TM, Grodzinsky AJ, Hunziker EB, Sandy JD. Effects of Almqvist KF, Victor J. Treatment of patellofemoral cartilage injurious compression on matrix turnover around individual defects in the knee by autologous matrix-induced chondro- cells in calf articular cartilage explants. J Orthop Res 1998;16: genesis (AMIC. Acta Orthop Belg 2014;80:251e9. 490e9. 72. Hunziker EB. Growth-factor-induced healing of partial- 92. Koh JL, Wirsing K, Lautenschlager E, Zhang LO. The effect of thickness defects in adult articular cartilage. Osteoarthritis graft height mismatch on contact pressure following osteo- Cartilage 2001;9:22e32. chondral grafting: a biomechanical study. Am J Sports Med 73. Shintani N, Siebenrock KA, Hunziker EB. TGF-ß1 enhances the 2004;32:317e20. BMP-2-induced chondrogenesis of bovine synovial explants 93. Kolker D, Murray M, Wilson M. Osteochondral defects of the and arrests downstream differentiation at an early stage of talus treated with autologous bone grafting. J Bone Joint Surg hypertrophy. PLoS One 2013;8:e53086. Br 2004;86:521e6. 74. Shintani N. Compositions for repair of lesions in cartilage and 94. Nam EK, Makhsous M, Koh J, Bowen M, Nuber G, Zhang LQ. bone. European Patent EP2175869B1 2008. Biomechanical and histological evaluation of osteochondral 75. Hunziker EB, Driesang IM. Functional barrier principle for transplantation in a rabbit model. Am J Sports Med 2004;32: growth-factor-based articular cartilage repair. Osteoarthritis 308e16. Cartilage 2003;11:320e7. 95. Tibesku CO, Szuwart T, Kleffner TO, Schlegel PM, Jahn UR, Van 76. Hunziker EB, Driesang IM, Saager C. Structural barrier prin- Aken H, et al. Hyaline cartilage degenerates after autologous ciple for growth factor-based articular cartilage repair. Clin osteochondral transplantation. J Orthop Res 2004;22: Orthop Relat Res 2001:S182e9. 1210e4. E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350 347

96. Huntley JS, Bush PG, McBirnie JM, Simpson AH, Hall AC. with autologous chondrocyte transplantation. N Engl J Med Chondrocyte death associated with human femoral osteo- 1994;331:889e95. chondral harvest as performed for mosaicplasty. J Bone Joint 114. Grande DA, Pitman MI, Peterson L, Menche D, Klein M. The Surg Am 2005;87:351e60. repair of experimentally produced defects in rabbit articular 97. Baumbach K, Petersen JP, Ueblacker P, Schroder J, Gopfert C, cartilage by autologous chondrocyte transplantation. Stork A, et al. The fate of osteochondral grafts after autolo- J Orthop Res 1989;7:208e18. gous osteochondral transplantation: a one-year follow-up 115. Driesang IM, Hunziker EB. Delamination rates of tissue flaps study in a minipig model. Arch Orthop Trauma Surg used in articular cartilage repair. J Orthop Res 2000;18: 2008;128:1255e63. 909e11. 98. Kock NB, Hannink G, van Kampen A, Verdonschot N, van 116. Jones CW, Willers C, Keogh A, Smolinski D, Fick D, Yates PJ, Susante JL, Buma P. Evaluation of subsidence, chondrocyte et al. Matrix-induced autologous chondrocyte implantation survival and graft incorporation following autologous osteo- in sheep: objective assessments including confocal arthros- chondral transplantation. Knee Surg Sports Traumatol copy. J Orthop Res 2008;26:292e303. Arthrosc 2011;19:1962e70. 117. Hunziker EB. Articular cartilage repair: are the intrinsic bio- 99. Paccola CA, Xavier CA, Goncalves RP. Fresh immature artic- logical constraints undermining this process insuperable? ular cartilage allografts. A study on the integration of chon- Osteoarthritis Cartilage 1999;7:15e28. dral and osteochondral grafts both in normal and in papain- 118. Hunziker EB. Biologic repair of articular cartilage. Defect treated knee joints of rabbits. Arch Orthop Trauma Surg models in experimental animals and matrix requirements. 1979;93:253e9. Clin Orthop Relat Res 1999:S135e46. 100. Meyers MH, Jones RE, Bucholz RW, Wenger DR. Fresh 119. Lee CR, Grodzinsky AJ, Hsu HP, Martin SD, Spector M. Effects autogenous grafts and osteochondral allografts for the of harvest and selected cartilage repair procedures on the treatment of segmental collapse in osteonecrosis of the hip. physical and biochemical properties of articular cartilage in Clin Orthop Relat Res 1983:107e12. the canine knee. J Orthop Res 2000;18:790e9. 101. Czitrom AA, Langer F, McKee N, Gross AE. Bone and cartilage 120. Hjelle K, Solheim E, Strand T, Muri R, Brittberg M. Articular allotransplantation. A review of 14 years of research and cartilage defects in 1,000 knee arthroscopies. Arthroscopy clinical studies. Clin Orthop Relat Res 1986:141e5. 2002;18:730e4. 102. Langer F, Gross AE, West M, Urovitz EP. The immunogenicity 121. Dehne T, Karlsson C, Ringe J, Sittinger M, Lindahl A. Chon- of allograft knee joint transplants. Clin Orthop Relat Res drogenic differentiation potential of osteoarthritic chon- 1978:155e62. drocytes and their possible use in matrix-associated 103. Stevenson S. The immune response to osteochondral allo- autologous chondrocyte transplantation. Arthritis Res Ther grafts in dogs. J Bone Joint Surg Am 1987;69:573e82. 2009;11:R133. 104. Gross AE. Cartilage resurfacing: filling defects. J Arthroplasty 122. Giannini S, Buda R, Grigolo B, Vannini F, De Franceschi L, 2003;18:14e7. Facchini A. The detached osteochondral fragment as a source 105. Chahal J, Gross AE, Gross C, Mall N, Dwyer T, Chahal A, et al. of cells for autologous chondrocyte implantation (ACI) in the Outcomes of osteochondral allograft transplantation in the ankle joint. Osteoarthritis Cartilage 2005;13:601e7. knee. Arthroscopy 2013;29:575e88. 123. Knutsen G, Drogset JO, Engebretsen L, Grontvedt T, Isaksen V, 106. Hickey MJ, Ohta I, Shigetomi M, Hurley JV, Kuwata N, Ludvigsen TC, et al. A randomized trial comparing autologous O'Brien BM. Vascularized heterotopic osteochondral allo- chondrocyte implantation with microfracture. Findings at grafts in a rat model following long-term immunosuppres- five years. J Bone Joint Surg Am 2007;89:2105e12. sion. J Reconstr Microsurg 1994;10:255e60. 124. Saris DB, Vanlauwe J, Victor J, Haspl M, Bohnsack M, 107. Stevenson S, Dannucci GA, Sharkey NA, Pool RR. The fate of Fortems Y, et al. Characterized chondrocyte implantation articular cartilage after transplantation of fresh and cry- results in better structural repair when treating symptomatic opreserved tissue-antigen-matched and mismatched osteo- cartilage defects of the knee in a randomized controlled trial chondral allografts in dogs. J Bone Joint Surg Am 1989;71: versus microfracture. Am J Sports Med 2008;36:235e46. 1297e307. 125. Zheng H, Martin JA, Duwayri Y, Falcon G, Buckwalter JA. 108. Marco F, Lopez-Oliva F, Fernandez Fernandez-Arroyo JM, de Impact of aging on rat bone marrow-derived stem cell Pedro JA, Perez AJ, Leon C, et al. Osteochondral allografts for chondrogenesis. J Gerontol A Biol Sci Med Sci 2007;62: osteochondritis dissecans and osteonecrosis of the femoral 136e48. . Int Orthop 1993;17:104e8. 126. Van Assche D, Staes F, Van Caspel D, Vanlauwe J, Bellemans J, 109. McDermott AG, Langer F, Pritzker KP, Gross AE. Fresh small- Saris DB, et al. Autologous chondrocyte implantation versus fragment osteochondral allografts. Long-term follow-up microfracture for knee cartilage injury: a prospective ran- study on first 100 cases. Clin Orthop Relat Res 1985:96e102. domized trial, with 2-year follow-up. Knee Surg Sports 110. Meyers MH, Akeson W, Convery FR. Resurfacing of the knee Traumatol Arthrosc 2009;18:486e95. with fresh osteochondral allograft. J Bone Joint Surg Am 127. Vanlauwe J, Saris DB, Victor J, Almqvist KF, Bellemans J, 1989;71:704e13. Luyten FP. Five-year outcome of characterized chondrocyte 111. Mahomed MN, Beaver RJ, Gross AE. The long-term success of implantation versus microfracture for symptomatic cartilage fresh, small fragment osteochondral allografts used for defects of the knee: early treatment matters. Am J Sports Med intraarticular post-traumatic defects in the knee joint. Or- 2011;39:2566e74. thopedics 1992;15:1191e9. 128. Saris DB, Vanlauwe J, Victor J, Almqvist KF, Verdonk R, 112. Sherman SL, Garrity J, Bauer K, Cook J, Stannard J, Bugbee W. Bellemans J, et al. Treatment of symptomatic cartilage defects Fresh osteochondral allograft transplantation for the knee: of the knee: characterized chondrocyte implantation results current concepts. J Am Acad Orthop Surg 2014;22:121e33. in better clinical outcome at 36 months in a randomized trial 113. Brittberg M, Lindahl A, Nilsson A, Ohlsson C, Isaksson O, compared to microfracture. Am J Sports Med 2009;37(Suppl Peterson L. Treatment of deep cartilage defects in the knee 1):10Se9S. 348 E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350

129. Farr J, Cole BJ, Sherman S, Karas V. Particulated articular chondrocytes embedded in collagen gel. J Bone Joint Surg Br cartilage: CAIS and DeNovo NT. J Knee Surg 2012;25:23e9. 1989;71:74e80. 130. Farr J, Tabet SK, Margerrison E, Cole BJ. Clinical, Radiographic, 148. McCarthy HE, Bara JJ, Brakspear K, Singhrao SK, Archer CW. and histological outcomes after cartilage repair with par- The comparison of equine articular cartilage progenitor cells ticulated juvenile articular cartilage: a 2-year prospective and bone marrow-derived stromal cells as potential cell study. Am J Sports Med 2014;42:1417e25. sources for cartilage repair in the horse. Vet J 2012;192: 131. Cerrato R. Particulated juvenile articular cartilage allograft 345e51. transplantation for osteochondral lesions of the talus. Foot 149. Yates KE, Glowacki J. Altered expression of connective tissue Ankle Clin 2013;18:79e87. genes in postnatal chondroinduced human dermal fibro- 132. Frisbie DD, Lu Y, Kawcak CE, DiCarlo EF, Binette F, blasts. Connect Tissue Res 2003;44:121e7. McIlwraith CW. In vivo evaluation of autologous cartilage 150. Farr J, Yao JQ. Chondral defect repair with particulated ju- fragment-loaded scaffolds implanted into equine articular venile cartilage allograft. Cartilage 2011;2:346e53. defects and compared with autologous chondrocyte im- 151. Steinert AF, Ghivizzani SC, Rethwilm A, Tuan RS, Evans CH, plantation. Am J Sports Med 2009;37(Suppl 1):71Se80S. Noth U. Major biological obstacles for persistent cell-based 133. Cole BJ, Farr J, Winalski CS, Hosea T, Richmond J, regeneration of articular cartilage. Arthritis Res Ther Mandelbaum B, et al. Outcomes after a single-stage proce- 2007;9:213. dure for cell-based cartilage repair: a prospective clinical 152. Wakitani S, Kawaguchi A, Tokuhara Y, Takaoka K. Present safety trial with 2-year follow-up. Am J Sports Med 2011;39: status of and future direction for articular cartilage repair. 1170e9. J Bone Miner Metab 2008;26:115e22. 134. Ahmed TA, Hincke MT. Strategies for articular cartilage lesion 153. Oldershaw RA. Cell sources for the regeneration of articular repair and functional restoration. Tissue Eng Part B Rev cartilage: the past, the horizon and the future. Int J Exp Pathol 2009;16:305e29. 2012;93:389e400. 135. Hunziker EB, Schenk RK. Physiological mechanisms adopted 154. Friedenstein AJ, Piatetzky II S, Petrakova KV. Osteogenesis in by chondrocytes in regulating longitudinal bone growth in transplants of bone marrow cells. J Embryol Exp Morphol rats. J Physiol 1989;414:55e71. 1966;16:381e90. 136. Mankin HJ. The reaction of articular cartilage to injury and 155. Johnstone B, Hering TM, Caplan AI, Goldberg VM, Yoo JU. osteoarthritis (second of two parts). N Engl J Med 1974;291: In vitro chondrogenesis of bone marrow-derived mesen- 1335e40. chymal progenitor cells. Exp Cell Res 1998;238:265e72. 137. Sandell LJ, Aigner T. Articular cartilage and changes in 156. Yoo JU, Barthel TS, Nishimura K, Solchaga L, Caplan AI, arthritis. An introduction: cell biology of osteoarthritis. Goldberg VM, et al. The chondrogenic potential of human Arthritis Res 2001;3:107e13. bone-marrow-derived mesenchymal progenitor cells. J Bone 138. Aigner T, Kurz B, Fukui N, Sandell L. Roles of chondrocytes in Joint Surg Am 1998;80:1745e57. the pathogenesis of osteoarthritis. Curr Opin Rheumatol 157. Kuroda R, Ishida K, Matsumoto T, Akisue T, Fujioka H, 2002;14:578e84. Mizuno K, et al. Treatment of a full-thickness articular carti- 139. Gobbi A, Karnatzikos G, Sankineani SR. One-step surgery with lage defect in the femoral condyle of an athlete with autol- multipotent stem cells for the treatment of large full- ogous bone-marrow stromal cells. Osteoarthritis Cartilage thickness chondral defects of the knee. Am J Sports Med 2007;15:226e31. 2014;42:648e57. 158. Haynesworth SE, Baber MA, Caplan AI. Cytokine expression 140. Betsch M, Schneppendahl J, Thuns S, Herten M, Sager M, by human marrow-derived mesenchymal progenitor cells Jungbluth P, et al. Bone marrow aspiration concentrate and in vitro: effects of dexamethasone and IL-1 alpha. J Cell platelet rich plasma for osteochondral repair in a porcine Physiol 1996;166:585e92. osteochondral defect model. PLoS One 2013;8:e71602. 159. Mayer H, Bertram H, Lindenmaier W, Korff T, Weber H, 141. Smyth NA, Murawski CD, Haleem AM, Hannon CP, Savage- Weich H. Vascular endothelial growth factor (VEGF-A) Elliott I, Kennedy JG. Establishing proof of concept: platelet- expression in human mesenchymal stem cells: autocrine and rich plasma and bone marrow aspirate concentrate may paracrine role on osteoblastic and endothelial differentiation. improve cartilage repair following surgical treatment for J Cell Biochem 2005;95:827e39. osteochondral lesions of the talus. World J Orthop 2012;3: 160. Aggarwal S, Pittenger MF. Human mesenchymal stem cells 101e8. modulate allogeneic immune cell responses. Blood 2005;105: 142. Richter W. Cell-based cartilage repair: illusion or solution for 1815e22. osteoarthritis. Curr Opin Rheumatol 2007;19:451e6. 161. De Bari C, Dell'Accio F, Tylzanowski P, Luyten FP. Multipotent 143. Chen FH, Rousche KT, Tuan RS. Technology Insight: adult mesenchymal stem cells from adult human synovial mem- stem cells in cartilage regeneration and tissue engineering. brane. Arthritis Rheum 2001;44:1928e42. Nat Clin Pract Rheumatol 2006;2:373e82. 162. Takahashi K, Yamanaka S. Induction of pluripotent stem cells 144. Ahmed N, Stanford WL, Kandel RA. Mesenchymal stem and from mouse embryonic and adult fibroblast cultures by progenitor cells for cartilage repair. Skeletal Radiol 2007;36: defined factors. Cell 2006;126:663e76. 909e12. 163. Chesterman PJ, Smith AU. Homotransplantation of articular 145. Arthur A, Zannettino A, Gronthos S. The therapeutic appli- cartilage and isolated chondrocytes. An experimental study cations of multipotential mesenchymal/stromal stem cells in in rabbits. J Bone Joint Surg Br 1968;50:184e97. skeletal tissue repair. J Cell Physiol 2009;218:237e45. 164. Iwasa J, Engebretsen L, Shima Y, Ochi M. Clinical application 146. Grande DA, Singh IJ, Pugh J. Healing of experimentally pro- of scaffolds for cartilage tissue engineering. Knee Surg Sports duced lesions in articular cartilage following chondrocyte Traumatol Arthrosc 2009;17:561e77. transplantation. Anat Rec 1987;218:142e8. 165. Kerker JT, Leo AJ, Sgaglione NA. Cartilage repair: synthetics 147. Wakitani S, Kimura T, Hirooka A, Ochi T, Yoneda M, Yasui N, and scaffolds: basic science, surgical techniques, and clinical et al. Repair of rabbit articular surfaces with allograft outcomes. Sports Med Arthrosc 2008;16:208e16. E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350 349

166. Chajra H, Rousseau CF, Cortial D, Ronziere MC, Herbage D, differentiation of bone marrow mesenchymal cells. Exp Cell Mallein-Gerin F, et al. Collagen-based biomaterials and Res 1999;250:485e98. cartilage engineering. Application to osteochondral defects. 185. Angele P, Kujat R, Nerlich M, Yoo J, Goldberg V, Johnstone B. Biomed Mater Eng 2008;18:S33e45. Engineering of osteochondral tissue with bone marrow 167. Ahmed TA, Dare EV, Hincke M. Fibrin: a versatile scaffold for mesenchymal progenitor cells in a derivatized hyaluronan- tissue engineering applications. Tissue Eng Part B Rev gelatin composite sponge. Tissue Eng 1999;5:545e54. 2008;14:199e215. 186. Weisser J, Rahfoth B, Timmermann A, Aigner T, Brauer R, von 168. Weidenbecher M, Henderson JH, Tucker HM, Baskin JZ, der Mark K. Role of growth factors in rabbit articular cartilage Awadallah A, Dennis JE. Hyaluronan-based scaffolds to repair by chondrocytes in agarose. Osteoarthritis Cartilage tissue-engineer cartilage implants for laryngotracheal 2001;9(Suppl A):S48e54. reconstruction. Laryngoscope 2007;117:1745e9. 187. Fortier LA, Lust G, Mohammed HO, Nixon AJ. Coordinate 169. Kang JY, Chung CW, Sung JH, Park BS, Choi JY, Lee SJ, et al. upregulation of cartilage matrix synthesis in fibrin cultures Novel porous matrix of hyaluronic acid for the three- supplemented with exogenous insulin-like growth factor-I. dimensional culture of chondrocytes. Int J Pharm 2009;369: J Orthop Res 1999;17:467e74. 114e20. 188. Dounchis JS, Goomer RS, Harwood FL, Khatod M, Coutts RD, 170. Pulliainen O, Vasara AI, Hyttinen MM, Tiitu V, Valonen P, Amiel D. Chondrogenic phenotype of perichondrium-derived Kellomaki M, et al. Poly-L-D-lactic acid scaffold in the repair of chondroprogenitor cells is influenced by transforming porcine knee cartilage lesions. Tissue Eng 2007;13:1347e55. growth factor-beta 1. J Orthop Res 1997;15:803e7. 171. Li WJ, Chiang H, Kuo TF, Lee HS, Jiang CC, Tuan RS. Evaluation 189. Adkisson HD, Gillis MP, Davis EC, Maloney W, Hruska KA. of articular cartilage repair using biodegradable nanofibrous In vitro generation of scaffold independent neocartilage. Clin scaffolds in a swine model: a pilot study. J Tissue Eng Regen Orthop Relat Res 2001:S280e94. Med 2009;3:1e10. 190. Oda K, Mori K, Imai S, Uenaka K, Matsusue Y. Comparison of 172. Ikeda R, Fujioka H, Nagura I, Kokubu T, Toyokawa N, Inui A, repair between cartilage and osteocartilage defects in rabbits et al. The effect of porosity and mechanical property of a using similarly manipulated scaffold-free cartilage-like con- synthetic polymer scaffold on repair of osteochondral de- structs. J Orthop Sci 2014;19(4):637e45. fects. Int Orthop 2009;33:821e8. 191. Nixon AJ, Fortier LA, Williams J, Mohammed H. Enhanced 173. Funayama A, Niki Y, Matsumoto H, Maeno S, Yatabe T, repair of extensive articular defects by insulin-like growth Morioka H, et al. Repair of full-thickness articular cartilage factor-I-laden fibrin composites. J Orthop Res 1999;17: defects using injectable type II collagen gel embedded with 475e87. cultured chondrocytes in a rabbit model. J Orthop Sci 192. van den Berg WB, van der Kraan PM, Scharstuhl A, van 2008;13:225e32. Beuningen HM. Growth factors and cartilage repair. Clin 174. Williams RJ, Gamradt SC. Articular cartilage repair using a Orthop Relat Res 2001:S244e50. resorbable matrix scaffold. Instr Course Lect 2008;57: 193. Hunziker EB, Driesang IM, Morris EA. Chondrogenesis in 563e71. cartilage repair is induced by members of the transforming 175. Jeong WK, Oh SH, Lee JH, Im GI. Repair of osteochondral growth factor-beta superfamily. Clin Orthop Relat Res 2001: defects with a construct of mesenchymal stem cells and a S171e81. polydioxanone/poly(vinyl alcohol) scaffold. Biotechnol Appl 194. Johnson K, Zhu S, Tremblay MS, Payette JN, Wang J, Biochem 2008;49:155e64. Bouchez LC, et al. A stem cell-based approach to cartilage 176. O'Shea TM, Miao X. Bilayered scaffolds for osteochondral repair. Science 2012;336:717e21. tissue engineering. Tissue Eng Part B Rev 2008;14:447e64. 195. Wu F, Dunkelman N, Peterson A, Davisson T, De La Torre R, 177. Lu HH, Subramony SD, Boushell MK, Zhang X. Tissue engi- Jain D. Bioreactor development for tissue-engineered carti- neering strategies for the regeneration of orthopedic in- lage. Ann N Y Acad Sci 1999;875:405e11. terfaces. Ann Biomed Eng 2010;38:2142e54. 196. Freed LE, Martin I, Vunjak-Novakovic G. Frontiers in tissue 178. Khan IM, Gilbert SJ, Singhrao SK, Duance VC, Archer CW. engineering. In vitro modulation of chondrogenesis. Clin Cartilage integration: evaluation of the reasons for failure of Orthop Relat Res 1999:S46e58. integration during cartilage repair. A review. Eur Cell Mater 197. Brehm W, Aklin B, Yamashita T, Rieser F, Trub T, Jakob RP, 2008;16:26e39. et al. Repair of superficial osteochondral defects with an 179. Reddi AH. Morphogenesis and tissue engineering of bone and autologous scaffold-free cartilage construct in a caprine cartilage: inductive signals, stem cells, and biomimetic bio- model: implantation method and short-term results. Osteo- materials. Tissue Eng 2000;6:351e9. arthritis Cartilage 2006;14:1214e26. 180. Reddi AH. Cartilage morphogenetic proteins: role in joint 198. Juergensen K, Aeschlimann D, Cavin V, Genge M, development, homoeostasis, and regeneration. Ann Rheum Hunziker EB. A new biological glue for cartilage-cartilage Dis 2003;62(Suppl 2):ii73e78. interfaces: tissue transglutaminase. J Bone Joint Surg Am 181. Glowacki J. In vitro engineering of cartilage. J Rehabil Res Dev 1995;79A:185e93. 2000;37:171e7. 199. Hunziker EB, Kapfinger E. Removal of proteoglycans from the 182. van der Kraan PM, Buma P, van Kuppevelt T, van den surface of defects in articular cartilage transiently enhances Berg WB. Interaction of chondrocytes, extracellular matrix coverage by repair cells. J Bone Joint Surg Br 1998;80: and growth factors: relevance for articular cartilage tissue 144e50. engineering. Osteoarthritis Cartilage 2002;10:631e7. 200. Visna P, Pasa L, Cizmar I, Hart R, Hoch J. Treatment of deep 183. Freyria AM, Mallein-Gerin F. Chondrocytes or adult stem cells cartilage defects of the knee using autologous chondrograft for cartilage repair: the indisputable role of growth factors. transplantation and by abrasive techniquesea randomized Injury 2012;43:259e65. controlled study. Acta Chir Belg 2004;104:709e14. 184. Andrades JA, Han B, Becerra J, Sorgente N, Hall FL, Nimni ME. 201. Hunziker EB. Methods and Compositions for the Treatment A recombinant human TGF-beta1 fusion protein with and Repair of Defects or Lesions in Cartilage. U.S. Patent 5 206 collagen-binding domain promotes migration, growth, and 023 1991. 350 E.B. Hunziker et al. / Osteoarthritis and Cartilage 23 (2015) 334e350

202. Hunziker EB, Tyler JA. Articular cartilage repair. In: Brandt K, of cartilage degeneration? Osteoarthritis Cartilage 2012;20: Doherty M, Lohmander S, Eds. Osteoarthritis 2003:93e101. 223e32. 203. Lee CH, Cook JL, Mendelson A, Moioli EK, Yao H, Mao JJ. 212. Sun MM, Beier F. Chondrocyte hypertrophy in skeletal Regeneration of the articular surface of the rabbit synovial development, growth, and disease. Birth Defects Res C Em- joint by cell homing: a proof of concept study. Lancet bryo Today 2014;102:74e82. 2010;376:440e8. 213. Mason JM, Grande DA, Barcia M, Grant R, Pergolizzi RG, 204. Murphy SV, Atala A. 3D bioprinting of tissues and organs. Nat Breitbart AS. Expression of human bone morphogenic protein Biotechnol 2014;32:773e85. 7 in primary rabbit periosteal cells: potential utility in gene 205. Hunziker EB, Kapfinger E, Geiss J. The structural architecture of therapy for osteochondral repair. Gene Ther 1998;5: adult mammalian articular cartilage evolves by a synchronized 1098e104. process of tissue resorption and neoformation during post- 214. Pascher A, Palmer GD, Steinert A, Oligino T, Gouze E, natal development. Osteoarthritis Cartilage 2007;15:403e13. Gouze JN, et al. Gene delivery to cartilage defects using 206. Wang G, Woods A, Sabari S, Pagnotta L, Stanton LA, Beier F. coagulated bone marrow aspirate. Gene Ther 2004;11: RhoA/ROCK signaling suppresses hypertrophic chondrocyte 133e41. differentiation. J Biol Chem 2004;279:13205e14. 215. Mankin HJ, Dorfman H, Lippiello L, Zarins A. Biochemical and 207. Saito T, Ikeda T, Nakamura K, Chung UI, Kawaguchi H. S100A1 metabolic abnormalities in articular cartilage from osteo- and S100B, transcriptional targets of SOX trio, inhibit terminal arthritic human hips. II. Correlation of morphology with differentiation of chondrocytes. EMBO Rep 2007;8:504e9. biochemical and metabolic data. J Bone Joint Surg Am 208. Gelse K, Klinger P, Koch M, Surmann-Schmitt C, von der 1971;53:523e37. Mark K, Swoboda B, et al. Thrombospondin-1 prevents 216. O'Driscoll SW, Keeley FW, Salter RB. The chondrogenic po- excessive ossification in cartilage repair tissue induced by tential of free autogenous periosteal grafts for biological osteogenic protein-1. Tissue Eng Part A 2011;17:2101e12. resurfacing of major full-thickness defects in joint surfaces 209. Klinger P, Surmann-Schmitt C, Brem M, Swoboda B, under the influence of continuous passive motion. An Distler JH, Carl HD, et al. Chondromodulin 1 stabilizes the experimental investigation in the rabbit. J Bone Joint Surg Am chondrocyte phenotype and inhibits endochondral ossifica- 1986;68:1017e35. tion of porcine cartilage repair tissue. Arthritis Rheum 217. Mainil-Varlet P, Aigner T, Brittberg M, Bullough P, 2011;63:2721e31. Hollander A, Hunziker E, et al. Histological assessment of 210. Takayama Y, Mizumachi K. Inhibitory effect of lactoferrin on cartilage repair: a report by the Histology Endpoint Com- hypertrophic differentiation of ATDC5 mouse chon- mittee of the International Cartilage Repair Society (ICRS). droprogenitor cells. Biometals 2010;23:477e84. J Bone Joint Surg Am 2003;85-A(Suppl 2):45e57. 211. van der Kraan PM, van den Berg WB. Chondrocyte hyper- 218. Hunziker E. Cartilage histomorphometry. Methods Mol Med trophy and osteoarthritis: role in initiation and progression 2006;135:147e66.