Sie sind auf Seite 1von 9

Bone 70 (2015) 93–101

Contents lists available at ScienceDirect

Bone
journal homepage: www.elsevier.com/locate/bone

Review

Bone fracture healing: Cell therapy in delayed unions and nonunions


Enrique Gómez-Barrena a,⁎, Philippe Rosset b,c, Daniel Lozano d, Julien Stanovici b,c,
Christian Ermthaller e, Florian Gerbhard e
a
Dept. of Orthopaedic Surgery and Traumatology, Hospital La Paz-IdiPAZ, Universidad Autónoma de Madrid, Madrid, Spain
b
Service of Orthopaedic Surgery and Traumatology, CHU Tours, Université François-Rabelais de Tours, PRES Centre-Val de Loire Université, Tours, France
c
Inserm U957, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives (LPRO), Faculté de Médecine, Université de Nantes, France
d
Metabolic Bone Research Unit, Instituto de Investigación Sanitaria FJD, Madrid, Spain
e
Klinik für Unfallchirurgie-, Hand-, Plastische und Wiederherstellungschirurgie Zentrum für Chirurgie Universitätsklinikum Ulm, Ulm, Germany

a r t i c l e i n f o a b s t r a c t

Article history: Bone fracture healing impairment related to mechanical problems has been largely corrected by advances in
Received 13 March 2014 fracture management. Better protocols, more strict controls of time and function, and hardware and surgical
Revised 26 July 2014 technique evolution have contributed to better prognosis, even in complex fractures. However, atrophic non-
Accepted 28 July 2014
union persists in clinical cases where, for different reasons, the osteogenic capability is impaired. When this is
Available online 2 August 2014
the case, a better understanding of the basic mechanisms under bone repair and augmentation techniques
Keywords:
may put in perspective the current possibilities and future opportunities. Among those, cell therapy particu-
Bone healing larly aims to correct this insufficient osteogenesis. However, the launching of safe and efficacious cell thera-
Nonunion pies still requires substantial amount of research, especially clinical trials. This review will envisage the
Cell therapy current clinical trials on bone healing augmentation based on cell therapy, with the experience provided by
Clinical trials the REBORNE Project, and the insight from investigator-driven clinical trials on advanced therapies towards
MSCs the future. This article is part of a Special Issue entitled Stem Cells and Bone.
© 2014 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/3.0/).

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93
Clinical bone healing impairment: from hypertrophic to atrophic nonunions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 94
Clinical and imaging diagnoses . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
Cells and molecules in bone healing after fractures and pharmacological new therapies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 95
Bone healing augmentation: grafts and biomaterials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 96
Advanced therapy (AMTP) proposed solutions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
Safety and preclinical rationale to launch clinical trials on cell therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 98
On-going cell therapy clinical trials to treat long bone non unions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 98
Future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100
Conflict of interest disclosure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100

Introduction

Bone fracture clinical management is oriented to obtain bone


⁎ Corresponding author at: Cirugía Ortopédica y Traumatología, Hospital La Paz,
Universidad Autónoma de Madrid, P° Castellana 261, 28046 Madrid, Spain. Fax: +34
healing in the shortest time frame, with the best possible functional re-
914269774. covery, and with less complications. However, an overall rate of 5 to 10%
E-mail address: enrique.gomezbarrena@uam.es (E. Gómez-Barrena). delayed union or nonunion is widely accepted as a perceived proportion

http://dx.doi.org/10.1016/j.bone.2014.07.033
8756-3282/© 2014 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/3.0/).
94 E. Gómez-Barrena et al. / Bone 70 (2015) 93–101

for bone healing problems, although this figure is not homogenous. Alternatively proposed strategies include those relying on the
Rather, different nonunion rates are found in different types of fracture, osteoconductive or osteoinductive capabilities of an implanted tissue
somewhat ranging from up to 18.5% in the tibia diaphysis [1] to 1.7% in (such as allograft or demineralized bone matrix) or a synthetic mate-
the femoral shaft after reamed nailing [2]. rial (such as bioceramics in different forms and compositions). Also,
The definition of delayed union and nonunion or pseudarthrosis different strategies have been defined to supplement potential mo-
certainly deserves more discussion. Those cases that correspond to a dif- lecular deficiency in the stimulation of local cell differentiation in
ferent healing rate than expected (slow healing rate) should be clearly the osteoprogenitor line (such as BMP or other growth factor local
separated from those in which the bone healing is no longer expected deliveries). These strategies rely on the surrounding or available cells
without treatment. A better understanding of fracture healing biology that might eventually produce the required local bone regeneration.
would help in fostering preclinical studies and clinical proposals in The expected fracture healing is seriously constrained in cases where
both of these directions: accelerating bone fracture healing in case previous efforts to heal the fracture have failed. Particularly in those
of slow healing rate, based on biological stimulation, and promoting cases with a supposed cell insufficiency, cell-based alternatives devel-
bone fracture healing in case of no healing expectations, based on oped over mesenchymal stem cells (MSCs) [4] have been proposed,
redeveloping the bone regeneration capability, whether fully lost or at and are currently under investigation and evaluation.
least under the required threshold to healing. In this context, this review progresses from clinical concepts of bone
Major limb injuries related to traffic accidents and multiple trauma healing impairment to advanced therapies under trial [5]. In this jour-
are a major health issue in developed countries, resulting in long treat- ney, cellular and molecular bases of bone regeneration in fracture
ments with substantial socioeconomic effects. But these injuries are also healing will be considered as the foundations of so-called therapy plat-
severely impacting less developed countries, where secondary compli- forms [6], state of the art and recent contributions to bone induction and
cations frequently generate major disabilities [3]. Long bone fractures augmentation will be appraised, and particular emphasis will be placed
are difficult and slow to heal and may require months until consolida- on cell therapy proposals and current cell therapy based orthopedic
tion is completed. Long treatments not only associate significant loss clinical trials.
of working days with economic effects on the patient and the society,
but also carry the risk of nonunion and permanent disabilities related Clinical bone healing impairment: from hypertrophic to
to malunion, joint stiffness, muscular atrophy, or reflex sympathetic atrophic nonunions
dystrophy.
The ability of fractured bone to regenerate and undergo repair may In a normal biological environment, many skeletal fractures heal un-
be compromised when insufficient osteogenic reaction is observed eventfully in the first 6 to 8 weeks. In case of an impaired bone healing
in the fracture callus, up to developing an atrophic nonunion. Those process due to a disturbed biological or mechanical environment, or
cases cannot be solved through a mechanical approach, as occurs with in cases where thick cortices are involved such as in femoral and tibial
hypertrophic nonunions. Treatment of these atrophic nonunions re- diaphysis, fractures may take a longer time to heal [7]. Per conventional
quires some form of bone healing augmentation, providing that vascu- definition, if a fracture is not healed after 4 months, it can be considered
larization is sufficient and confirming that infection is absent. a delayed union. If no bony healing is obtained in 6 months after
Conventional, standard treatment to augment bone healing is based the fracture, it can be clinically considered as nonunion, although the
on bone autograft, today's most accepted gold standard. The application diagnosis requires specific radiological features showing bone ending
of autologous cancellous and corticocancellous grafts, or larger, even changes.
vascularized, segmental bone grafts (frequently constructed out of the There are two distinct variants of nonunions with opposed under-
fibula) when the defect exceeds some centimeters, may permit the lying pathomechanisms, namely hypertrophic and atrophic nonunions.
most appraised personalized management to this problem. Yet this clas- A hypertrophic nonunion presents with a large, vital callus, although
sical orthopedic approach may be not appropriate. And this happens inefficient to regenerate bony union. On conventional radiographs,
when the autograft strategy has already failed, when the osteogenic po- the hypertrophic nonunion displays a large, broaden callus towards
tential of the available donor site is altered (due to cell scarcity, fibrous the fracture gap, with a radiolucent area instead of bone bridging. Due
tissue abundance due to previous harvesting, or other impairments), or to its radiological features (Fig. 1), the hypertrophic nonunion is also
when the risk/benefit evaluation of the autologous bone graft obtention called elephant foot nonunion [8]. Its basic problem is the mechanical
is unbalanced or refused by the patient. disturbance of the chosen fixation technique. The most recognized

Fig. 1. Radiological AP and lateral views of a tibial midshaft hypertrophic nonunion.


E. Gómez-Barrena et al. / Bone 70 (2015) 93–101 95

etiology underlying hypertrophic nonunions is the inefficient and un- intramedullary nail can influence the further course of healing and re-
stable fixation of the fracture allowing for multidirectional motion of orient a delayed union or even some nonunions towards adequate
fracture fragments. bone consolidation. Patients with a manifest nonunion usually complain
Whereas limited axial compressive movements can increase callus about pain in the fracture area with, and sometimes even without,
formation and accelerate fracture healing [9], shear displacement has weight bearing. The affected bone is usually sensitive upon pressure
demonstrated to hinder callus formation [10]. Up to a critical value, an and patients are not able to bear full weight [17].
increasing interfragmentary motion leads to an increase in callus forma- In cases of suspected infectious genesis of nonunion with possible
tion. Above a critical threshold, especially in combination with larger additional symptoms like reddening, hyperthermia and elevated body
gap sizes, interfragmentary motion leads to hypertrophic nonunions temperature, laboratory analysis should be obtained for infectious pa-
[9,11,12]. Most frequently, the treatment of hypertrophic nonunions rameters such as white blood cell count and inflammation parame-
is surgically oriented. Exchange of the fixation technique towards ters [19].
a more stable osteosynthesis aims to restrict the fracture gap with a After clinical and laboratory evaluation, conventional radiographs in
limited amount of compressive forces [13,14]. Secondarily, additional two orthogonal planes represent the basic diagnostic imaging tool,
treatment by ultrasound or external shock wave therapy has also been where the radiolucent gap between bone endings is associated to
proposed, although definite evidence is still lacking and significant con- closure of intramedullary canals of diaphyseal bone endings. Besides,
troversy remains about this issue [15,16]. the basic characteristics of the nonunion (status of consolidation,
The pathomechanisms leading to atrophic bone nonunions are hypertrophic/atrophic nonunion, segmental bone defects) can be evalu-
completely different. Claimed underlying causes usually incorporate ated for a more precise diagnosis.
biological impairment, sometimes in combination with mechanical fac- If the amount of consolidation or the radiological signs of nonunion
tors. In most cases, atrophic nonunions are the expression of impaired do not become obvious in conventional radiographic evaluation, a com-
biological support for bone healing, as for damaged vascular supply, puter tomography (CT) of the affected region is mandatory. Three-
and destruction of the periosteum and endosteum. This impairment is dimensional reconstructions and exact illustration of the fractured
frequently associated to cofactors such as polytrauma or soft tissue region, with the amount and location of possible callus bridges, can be
damage, with detraction of surrounding tissues [17]. Consecutively, evaluated through CT imaging (Fig. 3).
fracture healing is impaired because of the deficiency of important me- In some cases, especially with doubtful aseptic pathogenesis of the
diators, blood supply or other indispensable biological parameters. non-union, additional diagnostic evaluation should be performed.
Mechanical reasons can also be involved in the development of atro- While bone scintigraphy no longer represents the state of the art diag-
phic nonunions. Excessively rigid fixation, insufficient compressive nostic imaging tool, fluorodeoxyglucose positron emission computer
forces, and a fracture gap too wide to allow bony bridging of the frag- tomography (FDG-PET-CT) has become more and more relevant in
ments can also contribute. In radiological images, the atrophic nonunion clinical daily routine. This imaging tool combines the exact imaging
demonstrates the absence of callus tissue, the narrowing of bone ends, from the CT with additional information about the metabolism of the
and a large radiolucent zone in the fracture gap (Figs. 2 and 3). The examined area with a high diagnostic sensitivity for a chronic osteitis.
treatment of atrophic bone nonunion requires a surgical intervention. FDG-PET-CT has been shown to be of good diagnostic accuracy in
The aims of this intervention are the elimination of disturbing mechan- bone pathology discrimination [20] and chronic osteomyelitis [21].
ical factors through modification of osteosynthesis and reduction of the The combination of clinical examination, laboratory analysis and ra-
fracture gap, the increase of compressive motion between fracture frag- diological imaging by conventional radiographs, CT and possibly PET-CT
ments, and the biological improvement by introduction of new cells, should be sufficient for a clear diagnosis. Underlying cellular and molec-
vascular supply and healthy bone tissue into the fracture gap [18]. ular mechanisms are key to develop specific treatments and its review is
mandatory to further progress in clinical research strategies.
Clinical and imaging diagnoses
Cells and molecules in bone healing after fractures and
In the regular follow-up of patients after bone fracture, the course of pharmacological new therapies
fracture consolidation is reviewed by conventional, two orthogonal pro-
jection radiographs. Therefore, the development of a nonunion can be Bone healing of fractures and small bone defects is a unique and very
monitored clinically and through imaging. In case of insufficient fracture effective process involving complex and well-orchestrated interactions
healing, early modification of osteosynthesis like dynamization of an between cells, cytokines, osteo-conductive matrix and a mechanically

Fig. 2. Radiological AP and lateral views of a femoral upper diaphysis atrophic nonunion.
96 E. Gómez-Barrena et al. / Bone 70 (2015) 93–101

factor b1 (TGFb1) is a potent chemotactic stimulator of mesenchymal


stem cells that enhances osteoblast precursors and chondrocyte prolif-
eration, and may participate in recruitment of bone cells in the trauma
area [31]. In addition, TGFb1 induces the production of extracellular
bone matrix proteins such as collagen, osteopontin, and alkaline phos-
phatase [7] and regulates different cell types implicated in bone turn-
over and fracture healing [31]. Sox9, Runx2, and Osterix are three
essential transcription factors that have important roles in the cell-fate
decision process by which mesenchymal cells become chondrocytes
and osteoblasts, through activation of cell type-specific genes [32].
Macrophages can also produce and secrete these factors including
bone morphogenetic proteins (BMPs) and vascular endothelial growth
factor (VEGF), which induce angiogenesis and bone formation [33,34].
Neovascularization of damaged tissue is crucial to successful bone
healing, providing oxygen and delivering progenitor cells [35]. The
vascular endothelium lost integrity produces hypoxic conditions that
induce chondrogenesis, as occurs in the central avascular area of the
callus [36]. In this regard, VEGF is a key osteogenic and angiogenic factor
that is expressed under the control of hypoxia-inducible factor (HIF)-1α
in low oxygen tension [35]. Overexpression of HIF1α in mature osteo-
blasts, in mice with distraction osteogenesis, stimulates bone regenera-
tion indicating an angiogenic response related to new bone formation.
BMPs, parathyroid hormone (PTH)-related protein (PTHrP) and other
osteogenic factors stimulate the expression of VEGF in osteoblastic
cells [37,38]. In this reparative phase, neoangiogenesis and chondrogen-
esis predominate to bridge the gap in the fracture and complete bone
healing, but this soft callus is then replaced with a hard callus connecting
bone fragments with new bone. Osteoblasts can form woven bone rap-
idly, but it is randomly arranged and mechanically weak [28], requiring
bone remodeling by which newly formed woven bone is replaced by la-
Fig. 3. CT confirmation of the gap in an atrophic nonunion.
mellae through the activity of osteoclasts and osteoblasts [39].
This cellular and molecular background justifies different strategies
stable environment with a good blood supply, according to the to promote bone regeneration based on molecular osteoinduction. The
“diamond concept” [22] to generate new bone instead of a fibrous use of agents that increase vascularization and osteoblastic maturation
scar, as occurs in other connective tissues. could contribute to early callus formation. In this context, PTH exerts
This complex dynamic process requires the precise orchestration anabolic actions throughout cAMP–PKA pathway activation, implicating
of various events during overlapping stages [23] with distinctive histo- on bone formation in vitro and in vivo [40] and interacting with impor-
logical characteristics, from the initial inflammatory response, the for- tant bone local factors such as PTHrP, BMPs, Wnt-β-catenin, EGF, and
mation of a cartilaginous soft callus, the formation of a bone hard FGF [40]. The possibility of using Wnt pathway molecules as anabolic
callus, and finally the bone union followed by remodeling. As is widely agents in bone repair is complex because their effects depend on
accepted, this bone repair in adults recapitulates the normal develop- the cell differentiation state [41]. In addition, this pathway is implicated
ment of the skeleton during embryogenesis [24]. Moreover, the current in tumoral processes. Studies with Wnt pathway antagonists such
paradigm of bone tissue engineering also relies on biomimetics to re- as DKK-1, SFRP and sclerostin are in progress. Several studies demon-
produce bone formation from development biology [25,26]. Prenatal strated that the use of these factors can promote bone formation in
bone formation starts with mesenchymal cell condensation and sub- rodent models associated with a decreased BMD and higher bone turn-
sequent differentiation to chondrocytes (through endochondral ossifi- over [42,43]. Sost (sclerostin-encoding gene) is a key modulator of
cation) or, in precise cases, straight forward to osteoblasts (through bone remodeling and its expression was rapidly reduced in the callus,
intramembranous ossification) [27]. Both processes are implicated in indicating that this would allow osteoblasts to escape from its inhibitory
the callus formation after fracture [24]. However, callus formation effect to promote bone repair [44].
in adult bone is highly influenced by factors such as inflammation, However, translation into clinical trials is limited at this point. PTH
presence of pluripotent and osteoprogenitor cells, gap distance between trials in fractures [45,46] aimed at accelerating fracture repair, particu-
bone fracture endings, and mechanical stabilization and loading. The larly in fractures that are seldom prone to nonunion. BMPs have been
endochondral ossification mechanism predominates in the majority of frequently used in clinical trials with significant heterogeneity. A sys-
fracture healing cases, advancing through several phases that involve tematic review and metaanalysis on 11 trials observed comparable
multiple cellular and molecular events [28] in the so-called “bone times to fracture healing between BMP and controls and confirmed
healing cascade” [29] from hematoma and inflammation to angiogene- some evidence of increased healing rates with BMP without a secondary
sis and chondrogenesis, to finally complete osteogenesis followed by procedure compared with usual care control in acute tibial fractures
bone remodeling. [47], but observed that achieving union for nonunited fractures was
The interruption of vascular endothelium integrity is the first step similar to bone graft substitutes. Other strategies such as local applica-
following trauma, accompanied by a disruption of the blood supply tion of FGF2 were found to accelerate tibial shaft fractures [48], although
and hematoma formation, associating the presence of necrotic material. no data are available in nonunions.
This facilitates a potent inflammatory response related to the pro-
duction of pro-inflammatory cytokines from aggregated platelets, as Bone healing augmentation: grafts and biomaterials
interleukin-1 (IL-1), IL-6 or tumor necrosis factor-α, which have che-
motactic activity towards endothelial cells, fibroblasts, lymphocytes Bone grafting is widely used in hospitals to repair injured, aged or
and monocytes–macrophages [30]. Specifically, transforming growth diseased skeletal tissue. In Europe, about one million patients encounter
E. Gómez-Barrena et al. / Bone 70 (2015) 93–101 97

surgical bone reconstruction annually and the numbers are increasing preclinical features when coating Si-based ceramics both in vitro and
due to our aging population. Bone grafting intends to facilitate bone in vivo [61,62].
healing through osteogenesis (i.e. bone generation) at the site of damage, But besides bone grafts, substitutes and their augmentation with
but this is only attained when augmentation includes cells capable growth factors and anabolic strategies, cell therapies have been pro-
of forming bone. Other options to augment this bone repair include posed to evolve towards new osteoinductive and osteogenic solutions
osteoinductive (i.e. bone inducers) and osteoconductive (i.e. bone that could safely and efficaciously compete with currently available
guides) capabilities of the supplied coadjuvants to the surgical treatment. standards.
Bone autograft is the safest and most effective grafting procedure,
since it contains a patient's own bone growing cells (to enhance osteo- Advanced therapy (AMTP) proposed solutions
genesis) and proteins (to enhance osteoinduction), while providing a
framework for the new bone to grow into (osteoconduction). However, In view of these limitations and the increasing number of bone
bone autograft is limited in quantity (about 20 cm3) and its harvesting grafting procedures, surgeons are looking for alternatives with added
(e.g. from the iliac crest) represents an additional surgical intervention, value compared to osteoconductive substitutes, such as cell therapy
with frequent consequences of pain and complications [49]. and tissue engineering [63].
The next solution is allograft bone directly coming from tissue According to the above mentioned diamond concept [22], MSCs play
banks (fresh-frozen) or prepared to be conserved (dried or lyophi- a crucial role in bone repair, and thus cell therapy can serve as an alter-
lized). This solution does not contain living cells and some matrix pro- native to autologous bone grafting. A large number of osteoprogenitor
teins are destroyed by virus-inactivation treatments and the freezing cells may be implanted at the injury site, either alone or combined
process, thus it only guarantees osteoconductive properties. Moreover, with a matrix. Autologous bone marrow (BM) is rich in growth factors
allograft bone may transfer disease or lead to immunological rejections and osteoprogenitors as MSCs are present in the mononuclear cellular
[50]. An interesting alternative is to combine allograft with MSCs fraction of the bone marrow. Bone marrow MSCs are currently the
from concentrated bone marrow, as has been proposed in bone defects most appropriate cells for inducing bone repair, as they have a strong
after revision hip surgery [51] but also preliminarily explored in long osteogenic potential and are easily obtained by culturing iliac crest aspi-
bone pseudarthrosis [52]. Yet the number of cells may be an issue, as rates. Several MSC-based cell therapy modalities have been developed,
the available evidence in preclinical models recommends a high num- i.e., with and without cell culturing, and with or without a matrix.
ber of cells [53] and many ongoing clinical trials are thus based on The mononuclear cell fraction of the bone marrow, which contains
high number of MSCs that require cell expansion, as will be discussed the MSCs, can be used directly by percutaneous injection of aspirated
later. BM into the injury site. To increase the number of injected mononucle-
Since both autograft and allograft have drawbacks, scientists have ar cells and consequently of MSCs, it is possible to separate the mono-
long searched for biocompatible materials that could be used in place nuclear cells by centrifugation and concentrate them 3-fold to 6-fold
of the transplanted bone [50,54]. Several biomaterials can be chosen [64] with good results in pseudarthrosis [65]. The healing rate in-
depending on the goal (mechanical strength or filling) and approach creased in proportion to the injected MSC concentration. Patients
(percutaneous or surgical). The most widely used biomaterials are whose fractures did not heal received fewer than 1000 MSCs per mL
calcium-phosphate ceramics, which usually combine hydroxyapatite and fewer than 30,000 MSCs in total, whereas those whose fractures
and tricalcium phosphate as granules or, more rarely, sticks, and exhibit healed received significantly higher MSC concentrations and counts,
interconnected pores each measuring 100–400 μm. These biomaterials with a mean of 1500 MSCs per mL and 54,000 MSCs in total, in a volume
promote the adhesion, proliferation, and osteoblastic differentiation of of 20 mL.
MSCs, as well as the production of the collagen matrix that subsequently Concentrated or unconcentrated mononuclear cells can be mixed in
undergoes mineralization. Collagen sponges and biodegradable poly- the operating room with a synthetic or natural osteo-conducting matrix
mers can also be used. The biomaterials must be absorbable, at a vari- (e.g., allogeneic bone graft or coral) before implantation. Few published
able rate depending on their anticipated biomechanical role, and must studies assessed the combined use of concentrated or unconcentrated
allow the ingrowth of newly formed blood vessels from the neighboring BM with a biomaterial [66–68]. This method is a valid option for every-
tissues. Good quality vascularization of the tissue in contact with the im- day practice, provided that CE-marked (that is, approved for clinical use
plant is crucial. in Europe) biomaterials are used and concentration (if used) is achieved
Although most of the available synthetic bone substitutes possess via an approved procedure.
some of the positive properties of autograft (particularly, osteoconductive Mononuclear cells may also be cultured in vitro to allow selection
capabilities and occasionally, osteoinductive properties), none has all and expansion of an adherent fraction corresponding to MSC. This
the benefits of one's own bone yet (osteogenic properties). Basically increases the number of MSC to millions of cells. Expanded MSCs can
and besides bone autografting, which is the only truly osteogenic mate- be extemporaneously mixed with scaffolds during surgery (Fig. 4) so
rial, orthobiological solutions today available to surgeons include that this composite material is used in the same way as bone grafts.
osteoconductive and osteoinductive products, such as different prep- Quarto et al. [69] first reported the use of cultured BM MSCs combined
arations of bone allograft (fresh-frozen or dried by lyophilization, intra-operatively with hydroxyapatite blocks to fill large bone defects
warranting osteoconduction), different synthetic substitutes (with (4–7 cm). They successfully treated 3 patients, with defects in the
variable properties but particularly osteoconductive), and synthetic tibia, humerus, and ulna, respectively. A subsequent study confirmed
pharmaceuticals with osteoinductive properties (such as bone morpho- healing of the defects after 6–7 years [70]. The expanded MSCs may
genetic proteins, BMPs). Available evidence confirms the outcome of also be injected alone percutaneously in the site of fracture or osteotomy
fractures and non-unions treated by surgical techniques augmented with interesting results in two studies [71,72].
by autograft [55] and by BMPs [47]; thus this information may be com- Tissue engineering combines bone marrow cells or mesenchymal
pared to efficacy studies about other solutions. stem cells, synthetic scaffolds and molecular signals (growth or differ-
An alternative strategy to accelerate bone healing includes the use entiating factors) in order to form hybrid constructs. In a classical ap-
of degradable biomaterials in combination with osteogenic factors. proach, bone tissue engineering consists of harvesting bone marrow
Besides the already mentioned growth factors, emerging anabolic oste- from a patient, isolating MSCs by their adherence to tissue culture plas-
ogenic factors are under scrutiny. This applies not only to PTH but also to tic, expanding and differentiating those cells in culture to a sufficient
PTHrP whose C-terminal 107–111 domain (also known as osteostatin) number, and then seeding them onto a suitable synthetic scaffold to
exhibits osteogenic features in vitro, and stimulates bone formation be expanded in vitro on this scaffold during several days/weeks. This al-
in vivo [56–60]. PTHrP also conferred both osteogenic and angiogenic lows for scaffold colonization and for cell differentiation, before grafting
98 E. Gómez-Barrena et al. / Bone 70 (2015) 93–101

in many publications, which compounds the difficulties to confirm


these products in well-designed clinical trials.

Safety and preclinical rationale to launch clinical trials on


cell therapy

Not only are complex design and management of clinical trial regu-
lation and subsequent approval applicable to cell therapy, but specific
ethical and regulatory issues are also present. Therefore, a substantial
amount of efforts are required to support clinical trial proposals on pre-
clinical strong arguments and data. In this context, cell therapy is con-
sidered an advanced therapy (AT) by the European legislation [77],
where cells or tissues are considered ‘engineered’ if they have been
subjected to substantial manipulation and are not intended to be used
for the same essential function or functions in the recipient as in the
donor. Principles applying to advanced therapies include marketing
authorization (pre-market approval), demonstration of quality, safety
and efficacy, and post-authorization vigilance. Manufacturing of these
Fig. 4. Cell and bioceramic intraoperative preparation before nonunion augmentation. products requires authorization by the competent authority of the
member state ensuring national traceability and pharmacovigilance re-
quirements as well as specific quality standards. The regulatory require-
of this processed composite material at the affected site, prior to im-
ments, currently derived from the field of pharmaceutical medications,
plantation into the same patient [73].
will have to evolve in accordance with the specific characteristics of cell
For bone reconstruction purposes, human MSCs have been seeded
therapy trials in surgery.
and cultured on porous calcium phosphate ceramics in osteogenic
At present, only autologous MSCs are used for bone repair cell thera-
media (dexamethasone, ascorbic acid, β-glycerophosphate). Early pro-
py. Intra-operative BM concentration in the operating room using small
posals lead to clinical studies with low numbers of patients using this
centrifuges and CE-marked kits does not require authorization and is per-
approach, but the outcomes were inconsistent showing low efficacy
formed under the responsibility of the surgeon. Safety of this procedure
in bone regeneration. From these, it is clear that the strategy requires
has been confirmed by Hernigou on 1873 patients [78]. For the cultured
significant tuning [74,75].
MSCs, Tarte et al. [79] found no evidence of deleterious changes or malig-
The reasons of the limited clinical success may be due to several
nant transformation of cultured MSCs used in two national multicentric
bottlenecks in the multidisciplinary field of bone tissue engineering,
immune-hematology trials. However, the immunomodulating effects of
particularly about biomaterials and cell limitations. Biomaterials used
MSCs and their stromal properties (ability to maintain the survival and
as bone void fillers are inspired by the bone extracellular matrix
growth of associated cells) warrant caution in patients treated for neo-
(hydroxyapatite, collagen I) but need to be colonized by cells and
plastic diseases, most notably bone malignancies.
vascularized in order to promote bone tissue formation and healing.
Preclinical rationale requires solid indices of feasibility and efficacy.
The regenerative capabilities of current biomaterials are still limited
In this field, preclinical studies only orient towards the real feasibility
to small bone defects. Regarding cell limitations, barriers are found in
and efficacy, whose definite proof requires clinical trials. Bone research
the autologous approach, the cell selection, the association of cells and
scientists scaling their hypothesis to clinical trials on bone cell therapy
materials, and the osteogenic differentiation of implanted cells. The au-
should be aware of the underlying strategy that should orient their pre-
tologous approach for isolation and osteogenic differentiation of MSCs is
clinical research to maximize the probabilities of satisfying regulators
highly demanding in terms of logistics, production and safety of culture
and obtaining the required approvals to launch trials. Understandably,
conditions leading to a costly therapeutic procedure. The selection of a
this strategy will be modified as upcoming evidence may make some
restricted population of cells from different donors with age and genetic
requirements unnecessary, while other new data may recommend dif-
diversities remains a challenge for regenerative medicine at this early
ferent preclinical approaches prior to clinical trials.
stage of research due to patient variability. The association of biomate-
In this context, the REBORNE European Union FP7th large inte-
rials and osteoprogenitor cells raises technical challenges (i.e. cell
grating project (www.reborne.org) has fostered our consortium to or-
sources, types, doses, timing) and regulatory issues (devices with me-
ganize the current preclinical requirements to request approval from
dicinal drugs) to implement clinical trials. Moreover, bone formation re-
multinational European competent authorities. Both in vitro and animal
quires different cell populations that cooperate to set up complex 3D
studies have been launched to preclinically support the derived clinical
tissue under the guidance of biomechanical cues while vascularization
trials. Particularly, a clinical multicentric phase I/IIa trial (EudraCT 2011-
plays a major role in tissue healing. Finally, osteogenic differentiation in-
005441-13, NCT01842477) aiming at safety and efficacy of cellular
duced in vitro is not fully supported by the in vivo release of osteogenic
therapy was started in May 2013 to assess the use of cultured, expanded
factors from the graft itself.
autologous BM cells intra-operatively loaded onto biphasic calcium-
An alternative to the previous strategies is to implant the composite
phosphate granules as an alternative to autologous cancellous bone
material (cell + scaffold) into a heterotopic site, e.g. in a richly
grafting in patients with long bone nonunion or delayed union.
vascularized muscle, to promote angiogenesis and blood vessel growth
into the construct for some weeks and then to transfer it to the affected
site with vascular anastomoses for the transferred muscle flap con- On-going cell therapy clinical trials to treat long bone non unions
taining the implant [76], but only anecdotical experience is available.
Most crucial in the lack of evidence on satisfactory AMTP solutions The review of international clinical trial databases is the only
that could be acceptable in current clinical practice is the insufficient updated source of on-going clinical trials. Search can be performed ini-
number of clinical trials with reasonable, standardized, and preclinically tially through the WHO International Clinical Trials Registry Platform —
well-supported cell products. Scientific preclinical proofs of efficacy are ICTRP [80]. This platform incorporates weekly updates of the European
frequently weak, and the proposed cell products are also difficult to re- Clinical Trials Database — EudraCT [81], the ClinicalTrials.gov database
produce in a standardized manner, based on the provided information [82], the International Standard Randomised Controlled Trial Number
Table 1
Clinical trials related with long bone fracture and mesenchymal cell therapy, cited in trial registries and completed or recruiting patients, with sufficient information about intervention.

Title Identifier Sponsor Principal Study phase Indication Intervention Status


investigator

BMAC injected
Treatment of non-union of long bone fractures by NCT01206179 Royan Institute, Mohssen Emadedin 1 Long bone nonunion BMAC injected percutaneously Completed 2011
autologous mesenchymal stem cell Tehran, Iran
Percutaneous autologous bone-marrow grafting NCT00512434 University Hospital, Philippe Rosset Open tibial fractures BMAC injected percutaneously Completed 2013
for open tibial shaft fracture (IMOCA) Tours, France
The efficacy of mesenchymal stem cells for NCT01788059 Emdadi Kamyab Mohammad 2 Nonunion femur BMAC injected percutaneously Completed 2013
stimulate the union in treatment of non-united Hospital, Khorasan, Iran Taghi Peivandi and tibia
tibial and femoral fractures

BMAC with bone substitute or DBM


Autologous implantation of mesenchymal stem NCT00250302 Hadassah Medical Meir Liebergall Distal tibia fracture BMAC + carrier Completed 2011
cells for the treatment of distal tibial fractures Organization,
Jerusalem, Israel

E. Gómez-Barrena et al. / Bone 70 (2015) 93–101


Clinical trial based on the use of mononuclear NCT01813188 FFISRM, Murcia, Spain Luis Meseguer 2 randomized Tibial pseudarthrosis BMAC + TCP + DBM Recruiting
cells from autologous bone marrow in patients vs autologous participants
with pseudoarthrosis bone graft
Evaluation the treatment of nonunion of long bone NCT01958502 Emdadi Kamyab Mohammad 2 Nonunion femur BMAC + collagenic 3-D Recruiting
fracture of lower extremities (femur and tibia) Hospital, Khorasan, Iran Taghi Peivandi and tibia scaffold with BMP-2 participants
using mononuclear stem cells from the iliac wing
within a 3-D tissue engineered scaffold
Use of adult bone marrow mononuclear cells in NCT01581892 HUCA, Oviedo, Spain Jesus Otero 1–2 Nonunion BMAC + osteogenic matrix Recruiting
patients with long bone nonunion participants

Expanded MSC alone


Treatment of atrophic nonunion by preosteoblast NCT00916981 University Hospital of Jean Philippe Hauzeur 1–2 Long bone nonunion Expanded MSC injected Completed 2012
cells Liege, Belgium percutaneously
Pivotal phase 2b/3 study on autologous osteoblastic EUCTR2011-005584-24-NL, Bone Therapeutics S.A., Stefan Desmyter 2b-3 randomized Long bone nonunion Expanded MSC injected Recruiting
cells implantation in hypotrophic non-union NCT01756326 Belgium vs autologous percutaneously (PREOB®) participants
fractures bone graft
Phase 1/2a study on allogeneic osteoblastic cells NCT02020590* Bone Therapeutics S.A., Enrico Bastianelli 1–2a Long bone nonunion Allogeneic osteoblastic cells Recruiting
implantation in delayed-union fractures Belgium injected percutaneously participants
(ALLOB®)

Expanded MSC + bone substitute


Feasibility study of Aastrom tissue repair cells to NCT00424567 Aastrom Biosciences, USA Matthew Jiménez 1-2 Long bone nonunion Cultured bone marrow Completed 2007
treat non-union fractures. tissue + bone matrix
Mesenchymal stem cells; donor and role in NCT01626625 Indonesia University Phedy Phe 1 compared Nonunion Expanded MSC + HA Recruiting
management and reconstruction of nonunion to iliac bone participants
fracture autograft
Evaluation of efficacy and safety of autologous MSCs EUCTR2011-005441-13, INSERM, France, Enrique Gómez Barrena 1–2a Long bone nonunion Expanded MSC + HA + TCP Recruiting
combined to biomaterials to enhance bone NCT01842477 (FP7 European participants
healing (OrthoCT1) Project REBORNE)

NCT: ClinicalTrials.gov; EUCTR: EudraCT; BMAC = bone marrow aspirate concentrate; DBM = demineralized bone matrix; HA = HYDROXYAPatite; TCP = tricalcium phosphate; MSC = mesenchymal stem cell.
All mentioned studies are on autologous cells except if indicated by an *.

99
100 E. Gómez-Barrena et al. / Bone 70 (2015) 93–101

Register — ISRCTN, and the Australian New Zealand Clinical Trials Regis- way. And last but not least, a future step that may help to further define
try, as well as monthly updates of national clinical trial registries. and spread these therapies is a careful cost–benefit assessment and
A particular distinction of European clinical trials on advanced ther- a broad economic evaluation to clarify the best indications of bone
apies is the large proportion of sponsors from academic and charitable repair cell therapy as a standard procedure, if confirmation of safety
organizations, as seen in a recent review of 318 trials from 2004 to and efficacy is clearly derived from current trials.
2010 on 250 therapies [83]. This aspect is reinforced by the fostering
of investigator-driven clinical trials from institutions and organizations Conflict of interest disclosure
across Europe [84], spreading the opportunities for more available clin-
ical information about the myriad possibilities that can be considered in The authors do not have anything to disclose.
the cell therapy field.
Yet, many declared clinical trials in any of the available international Acknowledgments
and national trial registries, both from academic and industrial spon-
sors, do not offer results or just provide initial information about the This work was supported in part by the European Commission,
research effort, and then the development of the trial and the final out- Seventh Framework Programme (FP7), through the REBORNE Project,
comes are difficult to trace. This is equally confirmed in the long bone grant agreement no. 241879.
nonunion cell therapy trials. To further illustrate the current situation, EGB also acknowledges support from the Spanish General Direc-
the available on-going trials on the topic of this review are summarized torate on Scientific and Technological Research, Ministry of Economy
in Table 1. and Competitiveness (grant no. SAF2012-40149-C02-01).
Excluding trials with unknown status or not yet recruiting, 13 trials
related with long bone fracture or nonunion and mesenchymal cell References
therapy were identified as they have been cited in clinical trial registries
as completed (6 of them) or recruiting patients. They may be classified [1] Fong K, Truong V, Foote CJ, Petrisor B, Williams D, Ristevski B, et al. Predictors of
nonunion and reoperation in patients with fractures of the tibia: an observational
into four groups to allow for comparative analysis. The first group, with study. BMC Musculoskelet Disord 2013;14:103.
3 trials all completed, includes the simplest technique, percutaneous [2] Nonunion following intramedullary nailing of the femur with and without reaming.
injection of bone marrow aspirate concentrate (BMAC); none of these Results of a multicenter randomized clinical trial. J Bone Joint Surg Am 2003;85-A:
2093–6.
trials have been published yet. In the second group (4 trials), BMAC
[3] WHO. The global burden of disease: 2004 update. Geneva: World Health Organisation;
is associated with bone substitutes or demineralized bone matrix 2008.
(DBM); results have been published about one single trial only [85], [4] Caplan AI. Mesenchymal stem cells. J Orthop Res 1991;9:641–50.
[5] Gomez-Barrena E, Rosset P, Muller I, Giordano R, Bunu C, Layrolle P, et al. Bone regen-
observing a shorter time to bone union with cells than in the controls.
eration: stem cell therapies and clinical studies in orthopaedics and traumatology.
In the third group, 3 trials intend to test percutaneous injection of ex- J Cell Mol Med 2011;15:1266–86.
panded MSCs, but the only completed trial is not yet published. In the [6] Arvidson K, Abdallah BM, Applegate LA, Baldini N, Cenni E, Gomez-Barrena E, et al.
fourth group, 3 trials address the association of expanded MSC and Bone regeneration and stem cells. J Cell Mol Med 2011;15:718–46.
[7] Einhorn TA. The cell and molecular biology of fracture healing. Clin Orthop Relat Res
bone matrix or substitute, but the only completed trial has not been 1998:S7–S21.
published yet. Needless to say that follow-up of these and other trials [8] Babhulkar S, Pande K, Babhulkar S. Nonunion of the diaphysis of long bones. Clin
on the topic will enlighten the future of the field. Orthop Relat Res 2005:50–6.
[9] Claes LE, Heigele CA, Neidlinger-Wilke C, Kaspar D, Seidl W, Margevicius KJ, et al.
A major criticism on the available trials are the underreported re- Effects of mechanical factors on the fracture healing process. Clin Orthop Relat Res
sults, which may reflect lack of protocol adherence, patient heterogene- 1998;355(Suppl.: S):132–47.
ity in small unicentric trials, confounding efficacy results in part due to [10] Augat P, Burger J, Schorlemmer S, Henke T, Peraus M, Claes L. Shear movement at the
fracture site delays healing in a diaphyseal fracture model. J Orthop Res 2003;21:
patient or to protocol variability, or others. Many of these trials do not 1011–7.
offer sufficient information about the cell product to correlate with [11] Carter DR, Blenman PR, Beaupre GS. Correlations between mechanical stress history
the results in other trials and many are also impossible to reproduce and tissue differentiation in initial fracture healing. J Orthop Res 1988;6:736–48.
[12] Claes LE, Heigele CA. Magnitudes of local stress and strain along bony surfaces pre-
in other centers due to lack of transparency. However, reliability is par-
dict the course and type of fracture healing. J Biomech 1999;32:255–66.
ticularly challenged by the size and design of the currently available [13] Augat P, Margevicius K, Simon J, Wolf S, Suger G, Claes L. Local tissue properties in
trials. Unless large, comparative trials with well-defined cell products bone healing: influence of size and stability of the osteotomy gap. J Orthop Res
1998;16:475–81.
are published, evidence on this therapy will remain controversial or
[14] Hak DJ. Management of aseptic tibial nonunion. J Am Acad Orthop Surg 2011;19:
even negative. 563–73.
[15] Romano CL, Romano D, Logoluso N. Low-intensity pulsed ultrasound for the treatment
Future directions of bone delayed union or nonunion: a review. Ultrasound Med Biol 2009;35:529–36.
[16] Bashardoust Tajali S, Houghton P, MacDermid JC, Grewal R. Effects of low-intensity
pulsed ultrasound therapy on fracture healing: a systematic review and meta-
A strong need of clinical results is required to further progress in cell analysis. Am J Phys Med Rehabil 2012;91:349–67.
therapy. Launched trials will hopefully provide this information in the [17] Bishop JA, Palanca AA, Bellino MJ, Lowenberg DW. Assessment of compromised frac-
ture healing. J Am Acad Orthop Surg 2012;20:273–82.
near future. If clinical results are positive, far greater challenges may [18] Hobby B, Lee MA. Managing atrophic nonunion in the geriatric population: inci-
be raised by the development of more complex tissue engineering tech- dence, distribution, and causes. Orthop Clin N Am 2013;44:251–6.
niques, and this may allow the treatment of large bone defects and [19] Struijs PA, Poolman RW, Bhandari M. Infected nonunion of the long bones. J Orthop
Trauma 2007;21:507–11.
unsolved situations [86] after appropriate in vivo models confirm the [20] Bischoff M, Bischoff G, Buck A, von Baer A, Pauls S, Scheffold F, et al. Integrated
specific solution to submit to trials. A multidisciplinary approach will FDG-PET–CT: its role in the assessment of bone and soft tissue tumors. Arch Orthop
be required to improve implanted cell survival and to ensure prompt Trauma Surg 2010;130:819–27.
[21] Stecker FF, Schierz JH, Opfermann T, Driesch D, Hofmann GO, Winkens T, et al. Early
vessel ingrowth into the biomaterial via careful selection of structure dynamic 18 F-FDG PET/CT to diagnose chronic osteomyelitis following lower
and shape, together with addition of cytokines and growth factors. extremity fractures. A pilot study. Nuklearmedizin 2013;52.
The development of new materials and cell combinations (hydrogel- [22] Giannoudis PV, Einhorn TA, Marsh D. Fracture healing: the diamond concept. Injury
2007;38(Suppl. 4):S3–6.
based, bioceramic-based, or other) that could eventually craft solutions
[23] Schindeler A, McDonald MM, Bokko P, Little DG. Bone remodeling during fracture
for supplying cells and biomaterials percutaneously is expected in the repair: the cellular picture. Semin Cell Dev Biol 2008;19:459–66.
near future. The immunosuppressive properties of MSCs may allow [24] Deschaseaux F, Sensebe L, Heymann D. Mechanisms of bone repair and regenera-
the transplantation of allogeneic MSCs in various orthopedic conditions, tion. Trends Mol Med 2009;15:417–29.
[25] Lenas P, Moos M, Luyten FP. Developmental engineering: a new paradigm for the de-
with the establishment of cell banks for regenerative medicine. Early sign and manufacturing of cell-based products. Part I: from three-dimensional cell
trials evaluating allogeneic MSCs in delayed unions are already under growth to biomimetics of in vivo development. Tissue Eng Part B Rev 2009;15:381–94.
E. Gómez-Barrena et al. / Bone 70 (2015) 93–101 101

[26] Lenas P, Moos M, Luyten FP. Developmental engineering: a new paradigm for the [57] Chai YC, Roberts SJ, Van Bael S, Chen Y, Luyten FP, Schrooten J. Multi-level factorial
design and manufacturing of cell-based products. Part II: from genes to networks: analysis of Ca2+/Pi supplementation as bio-instructive media for in vitro biomimetic
tissue engineering from the viewpoint of systems biology and network science. engineering of three-dimensional osteogenic hybrids. Tissue Eng Part C Methods
Tissue Eng Part B Rev 2009;15:395–422. 2012;18:90–103.
[27] Fang J, Hall BK. Chondrogenic cell differentiation from membrane bone periostea. [58] Ryan G, Pandit A, Apatsidis DP. Fabrication methods of porous metals for use in
Anat Embryol (Berl) 1997;196:349–62. orthopaedic applications. Biomaterials 2006;27:2651–70.
[28] Marsh DR, Li G. The biology of fracture healing: optimising outcome. Br Med Bull [59] Wysolmerski JJ. Parathyroid hormone-related protein: an update. J Clin Endocrinol
1999;55:856–69. Metab 2012;97:2947–56.
[29] Phillips AM. Overview of the fracture healing cascade. Injury 2005;36(Suppl. 3):S5–7. [60] Valin A, Guillen C, Esbrit P. C-terminal parathyroid hormone-related protein (PTHrP)
[30] Kon T, Cho TJ, Aizawa T, Yamazaki M, Nooh N, Graves D, et al. Expression of osteo- (107–139) stimulates intracellular Ca(2+) through a receptor different from the type
protegerin, receptor activator of NF-kappaB ligand (osteoprotegerin ligand) and re- 1 PTH/PTHrP receptor in osteoblastic osteosarcoma UMR 106 cells. Endocrinology
lated proinflammatory cytokines during fracture healing. J Bone Miner Res 2001;16: 2001;142:2752–9.
1004–14. [61] Takemoto M, Fujibayashi S, Neo M, Suzuki J, Kokubo T, Nakamura T. Mechanical
[31] Vo TN, Kasper FK, Mikos AG. Strategies for controlled delivery of growth factors and properties and osteoconductivity of porous bioactive titanium. Biomaterials 2005;
cells for bone regeneration. Adv Drug Deliv Rev 2012;64:1292–309. 26:6014–23.
[32] Sinha KM, Zhou X. Genetic and molecular control of osterix in skeletal formation. [62] Stewart AF, Cain RL, Burr DB, Jacob D, Turner CH, Hock JM. Six-month daily admin-
J Cell Biochem 2013;114:975–84. istration of parathyroid hormone and parathyroid hormone-related protein peptides
[33] Champagne CM, Takebe J, Offenbacher S, Cooper LF. Macrophage cell lines produce to adult ovariectomized rats markedly enhances bone mass and biomechanical
osteoinductive signals that include bone morphogenetic protein-2. Bone 2002;30:26–31. properties: a comparison of human parathyroid hormone 1–34, parathyroid
[34] Lehmann W, Edgar CM, Wang K, Cho TJ, Barnes GL, Kakar S, et al. Tumor necrosis hormone-related protein 1–36, and SDZ-parathyroid hormone 893. J Bone Miner
factor alpha (TNF-alpha) coordinately regulates the expression of specific matrix Res 2000;15:1517–25.
metalloproteinases (MMPS) and angiogenic factors during fracture healing. Bone [63] Langer R, Vacanti JP. Tissue engineering. Science 1993;260:920–6.
2005;36:300–10. [64] Hernigou P, Mathieu G, Poignard A, Manicom O, Beaujean F, Rouard H. Percutaneous
[35] Salinas AJ, Esbrit P, Vallet-Regi M. A tissue engineering approach based on the use of autologous bone-marrow grafting for nonunions. Surgical technique. J Bone Joint
bioceramics for bone repair. Biomater Sci 2013;1:40–51. Surg Am 2006;88(Suppl. 1):322–7 Pt 2:.
[36] Boerckel JD, Uhrig BA, Willett NJ, Huebsch N, Guldberg RE. Mechanical regulation of [65] Hernigou P, Poignard A, Beaujean F, Rouard H. Percutaneous autologous bone-
vascular growth and tissue regeneration in vivo. Proc Natl Acad Sci U S A 2011;108: marrow grafting for nonunions. Influence of the number and concentration of pro-
E674–80. genitor cells. J Bone Joint Surg Am 2005;87:1430–7.
[37] Akeel S, El-Awady A, Hussein K, El-Refaey M, Elsalanty M, Sharawy M, et al. Recom- [66] Ateschrang A, Ochs BG, Ludemann M, Weise K, Albrecht D. Fibula and tibia fusion
binant bone morphogenetic protein-2 induces up-regulation of vascular endothelial with cancellous allograft vitalised with autologous bone marrow: first results for
growth factor and interleukin 6 in human pre-osteoblasts: role of reactive oxygen infected tibial non-union. Arch Orthop Trauma Surg 2009;129:97–104.
species. Arch Oral Biol 2012;57:445–52. [67] Dallari D, Savarino L, Stagni C, Cenni E, Cenacchi A, Fornasari PM, et al. Enhanced
[38] Esbrit P, Alvarez-Arroyo MV, De Miguel F, Martin O, Martinez ME, Caramelo C. tibial osteotomy healing with use of bone grafts supplemented with platelet gel or
C-terminal parathyroid hormone-related protein increases vascular endothelial platelet gel and bone marrow stromal cells. J Bone Joint Surg Am 2007;89:2413–20.
growth factor in human osteoblastic cells. J Am Soc Nephrol 2000;11:1085–92. [68] Ochs BG, Schmid U, Rieth J, Ateschrang A, Weise K, Ochs U. Acetabular bone recon-
[39] Zhang L, Richardson M, Mendis P. Role of chemical and mechanical stimuli in struction in revision arthroplasty: a comparison of freeze-dried, irradiated and
mediating bone fracture healing. Clin Exp Pharmacol Physiol 2012;39:706–10. chemically-treated allograft vitalised with autologous marrow versus frozen non-
[40] Siddappa R, Martens A, Doorn J, Leusink A, Olivo C, Licht R, et al. cAMP/PKA pathway irradiated allograft. J Bone Joint Surg (Br) 2008;90:1164–71.
activation in human mesenchymal stem cells in vitro results in robust bone forma- [69] Quarto R, Mastrogiacomo M, Cancedda R, Kutepov SM, Mukhachev V, Lavroukov A,
tion in vivo. Proc Natl Acad Sci U S A 2008;105:7281–6. et al. Repair of large bone defects with the use of autologous bone marrow stromal
[41] Chen Y, Alman BA. Wnt pathway, an essential role in bone regeneration. J Cell cells. N Engl J Med 2001;344:385–6.
Biochem 2009;106:353–62. [70] Marcacci M, Kon E, Moukhachev V, Lavroukov A, Kutepov S, Quarto R, et al. Stem
[42] Kulkarni NH, Wei T, Kumar A, Dow ER, Stewart TR, Shou J, et al. Changes in osteo- cells associated with macroporous bioceramics for long bone repair: 6- to 7-year
blast, chondrocyte, and adipocyte lineages mediate the bone anabolic actions of outcome of a pilot clinical study. Tissue Eng 2007;13:947–55.
PTH and small molecule GSK-3 inhibitor. J Cell Biochem 2007;102:1504–18. [71] Kitoh H, Kawasumi M, Kaneko H, Ishiguro N. Differential effects of culture-expanded
[43] Heath DJ, Chantry AD, Buckle CH, Coulton L, Shaughnessy Jr JD, Evans HR, et al. bone marrow cells on the regeneration of bone between the femoral and the tibial
Inhibiting Dickkopf-1 (Dkk1) removes suppression of bone formation and prevents lengthenings. J Pediatr Orthop 2009;29:643–9.
the development of osteolytic bone disease in multiple myeloma. J Bone Miner Res [72] Kim SJ, Shin YW, Yang KH, Kim SB, Yoo MJ, Han SK, et al. A multi-center, randomized,
2009;24:425–36. clinical study to compare the effect and safety of autologous cultured
[44] Caetano-Lopes J, Lopes A, Rodrigues A, Fernandes D, Perpetuo IP, Monjardino T, et al. osteoblast(Ossron) injection to treat fractures. BMC Musculoskelet Disord 2009;10:20.
Upregulation of inflammatory genes and downregulation of sclerostin gene expres- [73] Petite H, Viateau V, Bensaid W, Meunier A, de Pollak C, Bourguignon M, et al. Tissue-
sion are key elements in the early phase of fragility fracture healing. PLoS One 2011; engineered bone regeneration. Nat Biotechnol 2000;18:959–63.
6:e16947. [74] Derubeis AR, Cancedda R. Bone marrow stromal cells (BMSCs) in bone engineering:
[45] Aspenberg P, Genant HK, Johansson T, Nino AJ, See K, Krohn K, et al. Teriparatide for limitations and recent advances. Ann Biomed Eng 2004;32:160–5.
acceleration of fracture repair in humans: a prospective, randomized, double-blind [75] Khan Y, Yaszemski MJ, Mikos AG, Laurencin CT. Tissue engineering of bone: material
study of 102 postmenopausal women with distal radial fractures. J Bone Miner Res and matrix considerations. J Bone Joint Surg Am 2008;90(Suppl. 1):36–42.
2010;25:404–14. [76] Warnke PH, Wiltfang J, Springer I, Acil Y, Bolte H, Kosmahl M, et al. Man as living
[46] Peichl P, Holzer LA, Maier R, Holzer G. Parathyroid hormone 1–84 accelerates bioreactor: fate of an exogenously prepared customized tissue-engineered mandible.
fracture-healing in pubic bones of elderly osteoporotic women. J Bone Joint Surg Biomaterials 2006;27:3163–7.
Am 2011;93:1583–7. [77] Regulation (EC) No 1394/2007 of the European Parliament and of the Council of 13
[47] Garrison KR, Shemilt I, Donell S, Ryder JJ, Mugford M, Harvey I, et al. Bone morpho- November 2007 on advanced therapy medicinal products and amending Directive
genetic protein (BMP) for fracture healing in adults. Cochrane Database Syst Rev 2001/83/EC and Regulation (EC) No 726/2004. Retrieved 12/03/2014, from http://
2010:CD006950. ec.europa.eu/health/documents/eudralex/vol-1/index_en.htm#reg.
[48] Kawaguchi H, Oka H, Jingushi S, Izumi T, Fukunaga M, Sato K, et al. A local applica- [78] Hernigou P, Homma Y, Flouzat-Lachaniette CH, Poignard A, Chevallier N, Rouard H.
tion of recombinant human fibroblast growth factor 2 for tibial shaft fractures: a ran- Cancer risk is not increased in patients treated for orthopaedic diseases with autol-
domized, placebo-controlled trial. J Bone Miner Res 2010;25:2735–43. ogous bone marrow cell concentrate. J Bone Joint Surg Am 2013;95:2215–21.
[49] Goulet JA, Senunas LE, DeSilva GL, Greenfield ML. Autogenous iliac crest bone [79] Tarte K, Gaillard J, Lataillade JJ, Fouillard L, Becker M, Mossafa H, et al. Clinical-grade
graft. Complications and functional assessment. Clin Orthop Relat Res 1997:76–81. production of human mesenchymal stromal cells: occurrence of aneuploidy without
[50] Burchardt H. Biology of bone transplantation. Orthop Clin N Am 1987;18:187–96. transformation. Blood 2010;115:1549–53.
[51] Hernigou P, Pariat J, Queinnec S, Homma Y, Lachaniette CH, Chevallier N, et al. [80] WHO International Clinical Trials Registry Platform — ICTRP. Retrieved 12.03.2014,
Supercharging irradiated allografts with mesenchymal stem cells improves acetabu- from http://www.who.int/ictrp/en/.
lar bone grafting in revision arthroplasty. Int Orthop 2014 [Epub ahead of print]. [81] European Clinical Trials Database—EudraCT. Retrieved 12.03.2014, from https://
[52] Fernandez-Bances I, Perez-Basterrechea M, Perez-Lopez S, Nunez Batalla D, www.clinicaltrialsregister.eu.
Fernandez Rodriguez MA, Alvarez-Viejo M, et al. Repair of long-bone pseudoarthrosis [82] ClinicalTrials.gov. Retrieved 12.03.2014, from www.clinicaltrials.gov.
with autologous bone marrow mononuclear cells combined with allogenic bone [83] Maciulaitis R, D'Apote L, Buchanan A, Pioppo L, Schneider CK. Clinical development
graft. Cytotherapy 2013;15:571–7. of advanced therapy medicinal products in Europe: evidence that regulators must
[53] Liao Y, Zhang XL, Li L, Shen FM, Zhong MK. Stem cell therapy for bone repair: a sys- be proactive. Mol Ther 2012;20:479–82.
tematic review and meta-analysis of preclinical studies with large animal model. Br J [84] ESF. Forward look on investigator-driven clinical trials. Strasbourg: European
Clin Pharmacol 2014. http://dx.doi.org/10.1111/bcp.12382 [Epub ahead of print]. Science Foundation; 2009.
[54] Laurencin C, Khan Y, El-Amin SF. Bone graft substitutes. Expert Rev Med Devices [85] Liebergall M, Schroeder J, Mosheiff R, Gazit Z, Yoram Z, Rasooly L, et al. Stem cell-
2006;3:49–57. based therapy for prevention of delayed fracture union: a randomized and prospec-
[55] Sen MK, Miclau T. Autologous iliac crest bone graft: should it still be the gold stan- tive preliminary study. Mol Ther 2013;21:1631–8.
dard for treating nonunions? Injury 2007;38(Suppl. 1):S75–80. [86] Petite H, Vandamme K, Monfoulet L, Logeart-Avramoglou D. Strategies for improving
[56] Lozano D, Feito MJ, Portal-Nunez S, Lozano RM, Matesanz MC, Serrano MC, et al. the efficacy of bioengineered bone constructs: a perspective. Osteoporos Int 2011;22:
Osteostatin improves the osteogenic activity of fibroblast growth factor-2 immobilized 2017–21.
in Si-doped hydroxyapatite in osteoblastic cells. Acta Biomater 2012;8:2770–7.

Das könnte Ihnen auch gefallen