ArticlePDF AvailableLiterature Review

Abstract and Figures

Osteoporosis is a worldwide disease with a very high prevalence in humans older than 50. The main clinical consequence is bone fractures, which often lead to patient disability or even death. Currently, there are a number of commercial biomaterials used to treat osteoporotic bone fractures, but most of them have not been specifically designed for that purpose. Simultaneously, many drug- or cell-loaded biomaterials have been proposed in research laboratories, but hardly any has received approval for commercial use. In order to analyze this scenario and propose alternatives to overcome this scenario, the Spanish and European Network of Excellence for the Prevention and Treatment of Osteoporotic Fractures "Ageing" was created. This network integrates three communities, e. g. clinicians, materials scientists and industrial advisors, tackling the same problem from three different points of view. Keeping in mind the premise "living longer, living better", this commentary is the result of the thoughts, proposals and conclusions obtained after one year working in the framework of this network.
Content may be subject to copyright.
Opinion Paper
The relevance of biomaterials to the prevention and treatment of
osteoporosis
D. Arcos
a,b,c
, A.R. Boccaccini
a,d
, M. Bohner
a,e
, A. Díez-Pérez
a,f
, M. Epple
a,g
, E. Gómez-Barrena
a,h
,
A. Herrera
a,i,j
, J.A. Planell
a,b,k
, L. Rodríguez-Mañas
a,l
, M. Vallet-Regí
a,b,c,
a
Envejecimiento: red de excelencia española y europea para la prevención y tratamiento local de fracturas osteoporóticas, MINECO, Spain
b
Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
c
Dpto. Química Inorgánica y Bioinorgánica, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital, 12 de Octubre i+12, Madrid, Spain
d
University of Erlangen-Nuremberg, Institute of Biomaterials, 91058 Erlangen, Germany
e
RMS Fundation, CH-2544 Bettlach, Switzerland
f
Hospital del Mar-IMIM, Department Internal Medicine, Universidad Autónoma de Barcelona, Barcelona, Spain
g
University of Duisburg-Essen, Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), D-45117 Essen, Germany
h
Servicio Cirugia Ortopedica & Traumatologia, Hospital La Paz, IdiPAZ, Universidad Autonoma Madrid, Madrid 28046, Spain
i
University of Zaragoza, Department of Surgery, E-50009 Zaragoza, Spain
j
Miguel Servet University Hospital, Department of Orthopaedic Surgery & Traumatology, Zaragoza 50009, Spain
k
Institute of Bioengineering of Catalonia IBEC, BaldiriReixac 15-20, Barcelona 08028, Spain
l
University Hospital Getafe, Division Geriatric Medicine, Madrid, Spain
article info
Article history:
Received 6 November 2013
Received in revised form 24 December 2013
Accepted 3 January 2014
Available online xxxx
Keywords:
Osteoporosis
Biomaterials
Ageing
Bone
abstract
Osteoporosis is a worldwide disease with a very high prevalence in humans older than 50. The main clin-
ical consequences are bone fractures, which often lead to patient disability or even death. A number of
commercial biomaterials are currently used to treat osteoporotic bone fractures, but most of these have
not been specifically designed for that purpose. Many drug- or cell-loaded biomaterials have been pro-
posed in research laboratories, but very few have received approval for commercial use. In order to ana-
lyze this scenario and propose alternatives to overcome it, the Spanish and European Network of
Excellence for the Prevention and Treatment of Osteoporotic Fractures, ‘‘Ageing’’, was created. This net-
work integrates three communities, e.g. clinicians, materials scientists and industrial advisors, tackling
the same problem from three different points of view. Keeping in mind the premise ‘‘living longer, living
better’’, this commentary is the result of the thoughts, proposals and conclusions obtained after one year
working in the framework of this network.
Ó2014 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
1. Introduction and context
Ageing of the musculoskeletal system is a rapidly growing issue
due to the demographics associated with ageing populations
throughout the world. Its consequences on health are linked,
among others, to osteoporosis, degenerative osteoarthritis and
muscle deterioration or sarcopenia. These three elements interact
to produce a picture of frailty, which often leads to bone fracture
when a fall occurs, typically in people of advanced age. Fracture
prevention, patient recovery, and avoidance of subsequent frac-
tures constitute a challenge that has not yet been resolved [1].
No satisfactory solution exists to the problem of bone weaken-
ing due to osteoporosis. Surgical procedures to implant a device in
weakened bone often lead to a clinical result that is worse than if
such an intervention were to be performed on a young and strong
bone. The risk of fracture increases exponentially with age, and the
recovery process from a fracture is often slow, difficult and may
lead to a disability or even to the death of the patient. Up to 25%
of patients who suffer a femur fracture will die within a year. Of
those who survive, approximately half are totally or partially
dependent [2]. Chronic pain, functional limitation, social depen-
dence, psychological disorders, reduced mental health score and
social isolation combine to cause a serious deterioration in the
quality of life.
Various treatments are currently available to reduce the impact
of bone fragility, but there is a lack of alternatives to restore bone
strength. Moreover, there are no comprehensive treatments
http://dx.doi.org/10.1016/j.actbio.2014.01.004
1742-7061/Ó2014 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
Corresponding author at: Dpto. Química Inorgánica y Bioinorgánica, Univers-
idad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital, 12 de
Octubre i+12, Madrid, Spain. Tel.: +34 913941861.
E-mail address: vallet@farm.ucm.es (M. Vallet-Regí).
Acta Biomaterialia xxx (2014) xxx–xxx
Contents lists available at ScienceDirect
Acta Biomaterialia
journal homepage: www.elsevier.com/locate/actabiomat
Please cite this article in press as: Arcos D et al. The relevance of biomaterials to the prevention and treatment of osteoporosis. Acta Biomater (2014),
http://dx.doi.org/10.1016/j.actbio.2014.01.004
available for the whole damaged system, i.e. able to address the
three factors of musculoskeletal fragility: bone, cartilage and mus-
cle. These three factors are both the cause and consequence of
osteoporotic fracture [3].
The economic aspects that surround this social and health issue
are also of paramount importance [4], hence any action aimed at
cost reduction need to be seriously considered. It is worth men-
tioning that a surgical intervention for a hip prosthesis implanta-
tion has a health service cost of around 20,000, including direct,
indirect and intangible costs [5]. The total current cost of hip frac-
ture treatments of osteoporotic origin in the US is US$20.3 billion.
This cost, far from diminishing, has experienced continuous
growth throughout the 20th century and the first decade of the
21st century [6–8]. In fact, the total number of osteoporotic frac-
tures in 1950 was 1.47 million and the projection for 2050 is
around 6.3 million [9]. In the European Union, direct costs related
to osteoporotic fractures in 2000 were estimated at 31.7 billion
[10]. Moreover, increasing life expectancy suggests even more seri-
ous difficulties for the future.
In the context of a socio-health problem such as osteoporosis,
we may highlight the three main agents involved:
The patient. All publications and websites devoted to osteopo-
rosis show that there is currently no satisfactory solution and
that it remains one of the major challenges for public health.
This claim is based on significant mortality and disability (per-
sonal costs) caused by osteoporosis and the economic costs of
the management, including treatment, of patients (social costs).
The patient, as a key player in the issue, is therefore still waiting
for a satisfactory solution.
The therapeutic team. Once the fracture (hip, spine, wrist, etc.)
has occurred, the surgeon has very few surgical solutions avail-
able. In addition, other issues concerning general health (frailty,
malnutrition, cognitive disorders, co-morbid conditions, poly-
pharmacy, etc.) may make both the pre- and post-surgical man-
agement of patients difficult. The reports issued by many of
these professionals support the idea that enormous patient ben-
efits could stem from preventive solutions.
The national health system. In the European Union, most
national health systems are the purchasers of prosthetic prod-
ucts and the defrayers of the intervention and hospitalization
costs. Undoubtedly, the gradual ageing of society will entail
an increased number of osteoporotic patients, and this scenario
could challenge the sustainability of some national health sys-
tems in the next few years.
Research into biomaterials over the last 30 years has produced
many variants of technologies, materials and physical forms in-
tended to supplement or replace osteoporotic bone. However, very
few of these have achieved broad application in either prevention
or treatment of osteoporotic bone and its lesions. For those who
are not directly involved with the commercialization of biomateri-
als it may be surprising that so little biomaterials research output
has reached the operating. However, there are many obstacles that
biomaterials and their creators must overcome to prove safety and
efficacy to the level required by the regulatory bodies in each coun-
try. Even where these are overcome it is far from certain that a bio-
material will be commercially exploited. Thus, the position today is
that there are more limitations than opportunities for transferring
biomaterials into clinical use, particularly in the management of
poor bone quality. This opinion article will discuss some of the is-
sues that account for the paucity of biomaterials in clinical use and
also suggest what more might be done to encourage more viable
clinical applications.
To begin with, there are some mismatches between the aspira-
tions of biomaterials researchers and (unmet) clinical needs. The
goals of industry and academia are broadly similar to those of
the surgical community in wanting to improve the clinical out-
comes of a given treatment. However, biomaterials are often devel-
oped by identifying unmet clinical needs (note the terms used here
such as ‘‘user needs’’ are those required in design control and qual-
ity systems for medical device design). Even where the unmet
needs are properly researched, their effective translation from user
needs through to formal clinical outcome studies is quite uncom-
mon. Even rarer is that a proper post-market surveillance is under-
taken to ensure that any adverse outcomes are recognized as part
of a quality-control system.
Considering this scenario, an interdisciplinary group of experts
comprising clinicians, materials scientists and industrial advisors
have initiated the Spanish and European Network of Excellence
for the Prevention and Treatment of Osteoporotic Fractures, ‘‘Age-
ing’’ (www.agening.net). This network shares and puts together
information coming from different specialties and proposes possi-
ble solutions for the treatment of ageing diseases in bone, such as:
identifying the most prevalent clinical problems and the most
relevant clinical solutions;
analyzing and discussing the role of drugs, implants, biomateri-
als and surgical techniques in such treatments; and
thinking about possible innovative treatments and even
prevention.
This article is the result of one year of discussions and thoughts,
proposals and conclusions obtained via the ‘‘Ageing’’ framework.
2. Osteoporosis—a clinician’s point of view
2.1. Significance of osteoporosis to frailty and disability
Over the last hundred years a profound demographic change
has taken place, the main consequence of which has been a signif-
icant increase in the percentage of people older than 65, who now
constitute around 17% of the population. In addition to this demo-
graphic change, a second transition occurred—an epidemiological
transition that produced a shift from the predominance of acute,
single and communicable diseases to a predominance of chronic,
multiple and non-communicable diseases. As a consequence of
these two complementary transitions, the spectrum of diseases
and the mode of illness has dramatically changed. If we finally take
into account that life expectancy will continue to increase slowly
(EUROPOP forecasts support a modest increase in life expectancy
of around 5–7 years in EU countries over the next 50 years), the fo-
cus of health interventions should change from prolonging life to
improving the quality of life.
The most important surrogate of this quality of life is function.
As functional status deteriorates, the quality of life gets worse.
During the last century, as we prolonged life expectancy we were
opening the door to the increased risk of disability that comes with
age. With aging, the loss of functional reserve capacity puts people
at high risk of developing disability. This is the explanation for the
increase in the rates of disability that accompanied the extension
of life expectancy during the last century (Fig. 1). During the last
25 years, however, this tendency has changed in many countries.
Although we do not know the causes explaining this drop in dis-
ability rates, this trend clearly shows that disability is an avoidable
consequence of the ageing process and that if its main causes were
known we could intervene in the hope of decreasing the preva-
lence of disability [11].
The fight against disability has several options, but probably the
least attractive is to passively wait for the development of a dis-
ability. As previously stated, the main cause of disability is the loss
2D. Arcos et al./ Acta Biomaterialia xxx (2014) xxx–xxx
Please cite this article in press as: Arcos D et al. The relevance of biomaterials to the prevention and treatment of osteoporosis. Acta Biomater (2014),
http://dx.doi.org/10.1016/j.actbio.2014.01.004
of functional reserve capacity. However, to successfully recover
from disability a high level of that capacity is needed. In fact, only
a small percentage (30%) of older people with disability are able
to improve functionally during the first year [12]. This is why the
main approach to improving the functional status of the popula-
tion should be the prevention of disability, instead of waiting to
treat it. With this purpose in mind, the concept of frailty has
emerged as a relevant tool to detect people with the highest risk,
both short and long term, of developing a disability (Fig. 2). Frailty
is a state of increased vulnerability to stressors due to a decreased
physiological reserve in multiple systems and a limited capacity to
maintain homeostasis [13]. Its prevalence is around 10%, ranging
from 1% to 3% for 65 year old people to more than 30% in people
over the age of 80. It predicts the risk of suffering from multiple ad-
verse outcomes, including death, hospitalization, disability and
falls [14]. Detecting frailty is of clinical importance [15] as it has
been shown that some therapeutic approaches, mainly based on
physical exercise, are able to bring patients out of a state of frailty,
or at least delay the progression of frailty to full disability [16].
Within this framework, the approach to osteoporosis, as is also
the case for many other chronic conditions prevalent in older peo-
ple, should be focused on the prevention of the clinical problems
associated with frailty and disability. The most frequent of these
conditions is hip fracture. Taking into account that the most impor-
tant risk factor for hip fracture is falling, the right way to manage
these patients is based on a comprehensive assessment and man-
agement of falling patients. For this purpose the introduction of
specialized units to deal with falls and fractures is becoming the
standard approach for managing these patients, as recommended
by a number of different organizations, e.g. World Health Organi-
zation, National Institute for Health and Care Excellence, British
Orthopaedic Association and the US Preventive Services Task Force.
In this regard, the focus in these units is to reinforce the idea of
a continuum of care and a comprehensive approach to avoiding or
preventing disability in these patients, taking advantage of all
available opportunities. One of these opportunities stems from ad-
vances in the biomaterials used in the surgical materials used in
those patients with hip fracture. These materials have a twofold
aim in the prevention of frailty and disability: firstly, to allow an
early mobilization of the patient after surgery; and secondly, to
provide a quick and efficient restoration of bone and, as far as
possible, of the musculoskeletal unit.
2.2. Clinical aspects, prophylaxis and treatment of osteoporosis
Osteoporosis is a common disease the incidence of which is rap-
idly increasing as the population ages [17,18]. The loss of bone and
deterioration in bone quality [19] induce decreased bone strength
with the associated increase in fracture risk [20]. The main clinical
consequences of the condition, therefore, are fractures, which are
associated with significant morbidity [21] and mortality [22].
Several factors contribute to an increased risk of osteoporosis
and fracture. These risk factors have recently been structured in
a decision algorithm, the FRAXÓtool, that allows the clinician to
calculate the absolute risk of fracture in ten years for an individual
patient [23]. This formula has been validated in a number of coun-
tries [24] and is available in several languages and for a large num-
ber of countries. Moreover, and also partially integrated into FRAX,
an important number of comorbidities have been identified that
also influence fracture risk either by deteriorating bone or by
increasing the propensity of patients to fall [25].
The clinical diagnosis of osteoporosis relies mainly in the mea-
surement of bone mineral density by dual-energy X-ray absorpti-
ometry (DEXA) in several skeletal regions [26]. Other methods
also calculate the mineral density by using ultrasound or quanti-
fied computerized tomography (QCT) scanners but their use in
the general population is limited. These methods have in common
the limitation of measuring only one of the determinants of bone
strength, the amount of mineral or bone mass, but do not capture
the quality of the material. For this to be done adequately, labora-
tory testing of ex vivo specimens is required to measure a broad
spectrum of mechanical properties (brittleness, toughness, work
Fig. 1. Three trajectories of ageing, with a differential risk for disability.
Fig. 2. The path from robustness towards frailty and disability: factors, biomarkers and modulators.
D. Arcos et al. / Acta Biomaterialia xxx (2014) xxx–xxx 3
Please cite this article in press as: Arcos D et al. The relevance of biomaterials to the prevention and treatment of osteoporosis. Acta Biomater (2014),
http://dx.doi.org/10.1016/j.actbio.2014.01.004
to failure, etc.) [27]. Recently new techniques have been developed
for the direct measurement of bone tissue strength [28,29].
Biochemical markers of bone turnover, reflecting the rate of cell
activity in the bone remodeling cycle, have been also extensively
developed and can predict the future fracture risk [30]. They also
provide the clinician with information about how the patient is
responding to treatment [31]. Last but not least, the simple detec-
tion in a routine radiograph or after a trauma permits the identifi-
cation of fractures, the cornerstone of the disease and the event
responsible for the morbidity and mortality associated with
osteoporosis. Suffering a low-energy traumatic fracture (fragility
fracture) is the ultimate demonstration of osteoporosis and as such
constitutes a diagnostic marker.
Osteoporosis prevention has two phases, primary prevention
and secondary prevention. Primary prevention starts during intra-
uterine life, childhood and adolescence, given that this determines
the development of a healthy strong skeleton in the adulthood.
Even though genetic factors determine to a great extent the nature
of our bones, the promotion of physical activity, adequate nutrition
and the avoidance of negative factors for normal bone develop-
ment are extremely important.
Secondary prevention starts once bone loss or bone fracture has
occurred. Suffering a fracture is the most potent predictor of new
fractures, in a progression of risk referred to as a ‘fracture cascade’.
Two main groups of drugs are currently available for the manage-
ment of the patients with osteoporosis: anticatabolics and anabo-
lics [32]. The anticatabolic or antiresorptive agents suppress or
attenuate the activity of the bone-resorbing cells, the osteoclasts,
hence stopping bone loss and increasing bone strength. On the
other hand, anabolic agents are capable of inducing bone forma-
tion, and therefore can reverse in part the deterioration induced
by the osteoporosis progression. The advances in this therapeutic
field have been very significant over the last two decades since
there are now several classes of drugs, including both chemical
and biological entities, that decrease the risk of vertebral fracture
and in some cases also of non-vertebral fractures (e.g. hip). The
main development in treatment has been in postmenopausal
osteoporosis in women although treatments also exist for gluco-
corticoid-induced osteoporosis [33,34] and osteoporosis in men
[35]. In spite of the substantial body of clinical evidence, the man-
agement of this disease is still highly variable across different
countries [36] and a considerable proportion of cases does not re-
spond to the treatment [37,38]. Whilst antiosteoporosis drugs have
their side effects [39], these bone treatments have shown other
health benefits such as cancer reduction in some instances [40]
and also a decrease of the overall mortality [41]. In summary, there
are now some effective tools that enable detection, diagnosis and
treatment of osteoporosis to combat the progression of this meta-
bolic disease, resulting from the ageing of societies worldwide.
2.3. Surgical treatment of osteoporotic fractures
Osteoporotic bone has special morphological and biological
characteristics. Fracture healing depends mainly on the mechanical
stability at the fracture site and the biological process of bone repair.
Osteoporosis entails a decreased bone mass and an altered bone
structure, leading to a lowered mechanical strength. Therefore, oste-
oporotic fractures are often severely comminuted, especially in tra-
becular bone areas. Fracture comminution and trabecular collapse
not only result in bone defects with impaired fracture stability,
but also make anatomical reduction and surgical reconstruction dif-
ficult. Biologically, the mesenchymal stem cells (MSCs) of osteopo-
rotic bone have less capacity to differentiate into osteoblasts than
those of healthy bone, possibly due to impaired osteoinductive sig-
nals and/or lower expression of bone morphogenetic protein-2
(BMP-2). A decreased angiogenetic capacity at the fracture site is
also common in osteoporotic bone [42].
Fracture fixation planning is determined by the special mechan-
ical properties of osteoporotic bone. It has to be noted that even in
normal everyday activity, the loads supported by bones and joints
are both large and dynamic, changing in both magnitude and direc-
tion with every step the patient makes. While implant failure is
potentially a risk, less rigid fixation devices, such as intramedullary
nails, bridge plates and tension band constructs, are preferred for
minimizing bone–implant interface stress and the concomitant
risk of bone failure. In compromised bone, improved screw-holding
power is essential to prevent screw pullout and/or migration and
to minimize implant loading. Moreover, comminution makes frac-
ture reduction more difficult, requiring the use of autologous bone
grafts or biomaterials to fill bone defects and to augment bone
fragments. It is therefore possible by combining implants and bio-
materials to achieve sufficient overall assembly stability and frac-
ture healing. Less load-resistant materials, even with bioactive
capacity, may be used in the upper limb fractures. However, spine
and lower limb fractures require the use of inert biomaterials with
greater load-bearing capacity, although calcium phosphate ce-
ments may occasionally be used.
The most common osteoporotic fractures involve the spine, the
hip and the distal radius. Surgical treatment of these fractures has
changed in recent years. The distal radius fracture is known as
‘‘sentinel fracture’’ because it is the first warning sign of osteoporo-
sis. We now know that surgical treatment of distal radial fractures,
particularly by plates through palmar or dorsal approaches, does
better than conservative treatment [43]. Severe comminution
and bone fragment collapse are often present, requiring the use
of biomaterials to fill bone defects, achieve fracture stability and
promote bone union.
Spine fracture is the most frequent osteoporotic fracture. Early
diagnosis and medical treatment with anabolic drugs are essential
to increase bone strength and prevent the so-called ‘‘fracture cas-
cade’’. Vertebroplasty and kyphoplastyas minimally invasive tech-
niques show excellent results in terms of quality of life, pain relief
and functional recovery, both short and long term [44]. More
importantly, Edidin et al. [45] showed that the mortality of
patients suffering from a vertebral bone fracture was significantly
reduced when their fractures were treated by vertebroplasty or
kyphoplasty.
Treatment of hip fractures varies according to their anatomical
location and classification. These are divided according to anatom-
ically defined regions: intracapsular neck, extracapsular neck, per-
trochanteric, intertrochanteric and subtrochanteric. Intracapsular
fractures (see Fig. 3) present biological problems due to the loss
of blood supply to the proximal fragment. Osteosynthesis is only
indicated for undisplaced Garden I and II type intracapsular frac-
tures in patients younger than 70 years. Total or partial hip arthro-
plasty, depending on the patient age, is the preferred technique for
displaced grade (Garden III and IV) fractures [46]. The dilemma
over the choice of fixation method of prosthesis to bone (cemented
or uncemented) is solved in favor of cemented prostheses: taking
into account the mean age of our patients (>80 years) more of
95% of arthroplasties are fixed to bone with cement, because loss
of bone mass in osteoporotic bone prevents good primary stability
by press-fitting of uncemented prosthesis and hinders perfect bony
integration of components; on the other hand, cemented prosthe-
ses have lower revision rates than uncemented due to aseptic
loosening and offer excellent clinical results. Uncemented arthro-
plasties are indicated only in younger patients (<70 years) with
good bone quality and long life expectancy [47–51].
The sliding hip screw, a device used in the treatment of trochan-
teric fractures, has been to some extent replaced over the years by
intramedullary nails with a sliding cephalic screw. These are often
4D. Arcos et al. / Acta Biomaterialia xxx (2014) xxx–xxx
Please cite this article in press as: Arcos D et al. The relevance of biomaterials to the prevention and treatment of osteoporosis. Acta Biomater (2014),
http://dx.doi.org/10.1016/j.actbio.2014.01.004
made from titanium alloy and may feature reduced diameter and
length to enable implantation by minimally invasive surgery
[52]. The key determinants for good outcome are the correct place-
ment of the cephalic screw and good anatomical fracture reduc-
tion. If a large posteromedial comminution is present, the bone
defect can be augmented with a bone substitute such as a calcium
phosphate cement. In cases of severe osteoporosis, the bone struc-
ture of the femoral head can be strengthened by injecting poly-
methylmethacrylate (PMMA) cement through the cephalic screw,
a technique known as augmentation, which prevents the cut-out.
The challenge for the future will be the development of bioac-
tive biomaterials that combine load bearing, interconnected poros-
ity and the ability to be loaded with biological factors that promote
fracture healing.
3. Osteoporosis. A materials scientist’s point of view
In an osteoporotic scenario, the paucity of bone and the de-
creased osteoblastic function result in an impaired response to im-
plants compared with healthy bones. As mentioned in previous
sections, this evidence is often observed by orthopedic surgeons
in their daily practice of fracture reduction in osteoporotic patients.
The experience of clinicians, outlined in Sec. 2, teaches us that the
primary issue with these patients is that they suffer from fractures
that require some form of fixation, and the fixation devices such as
plates, screws, nails, etc., are difficult to fix in low-quality bone. In
addition, there are scenarios other than the fractures, which also
affect biomaterials performance in osteoporotic patients. For in-
stance, the response of osteoporotic bone to endosseous implants,
such as stems of total joint prostheses or endosseous dental im-
plants, is also strongly impaired. In these cases, the implant failure
is due to poor biological fixation, which is a consequence of insuf-
ficient osteogenesis around the implant [53]. Despite this evidence,
there are no clinically approved biomaterials specifically tailored
for application in osteoporotic bones. Certainly, there are some
examples of medical devices for osteosynthesis with special de-
signs, but they are made of the same biomaterials as the conven-
tional devices, such as titanium alloys, cobalt alloys or stainless
steel.
Attempts to reduce osteoporotic fractures usually involve two
classes of biomaterials: metallic implants and cements. Their func-
tion is slightly different: whereas metallic implants are used as
primary fixation devices, cements are mainly used as reinforce-
ment of the metallic hardware. However, cements are also used
as stand-alone devices, e.g. for bone augmentation procedures
(injection of the cement into osteoporotic bone) [54]. Indeed there
is increasing research in the field of injectable calcium phosphate
cements with recent efforts focusing on incorporating different
additives including inorganic bioactive elements, e.g. bioactive
glass [55], radiopacifiers, e.g. tantalum oxide or barium sulfate
[56], biodegradable polymers to improve the injectability [57]
and modifications to incorporate antibiotic releasing capability
[58]. Another difference between metallic implants and cements
is the way they are adapted for osteoporosis-related indications.
As mentioned above, metallic biomaterials are the same as those
used in non-osteoporotic patients, but the implant shape is modi-
fied to accommodate osteoporosis-specific requirements. Some-
times, even new implants are created. For cements, the accent is
on changing the composition to obtain specific properties such as
constant viscosity or high radiopacity. In addition, bioactive and/
or resorbable ceramics such as calcium phosphates or bioactive
glasses can be used to fill voids, thus avoiding the harvest of autog-
enous bone from the iliac crest.
Besides the osteoporotic fracture reduction, biomaterials sci-
ence also offers a possible approach to dealing with the challenge
of impaired osteointegration of permanent endosseous implants.
Osteointegration in these cases is seriously affected, mainly due
to the decreased osteoblast activity. An osteoporotic environment
strongly affects the primary (short-term) stability of the implant,
because the quality of the host bone is significantly decreased.
Moreover, biological stability (early and long-term) is also im-
paired, as it requires deposition of newly formed bone in intimate
contact with the implant [59]. Since this process involves the bal-
anced action of osteogenic and bone-resorbing cells, osteoporosis
often has a poor prognosis and delayed healing and osteointegra-
tion with endosseous implants. However, similarly to devices for
fracture fixation purposes, research efforts have so far not involved
the preparation of new metal alloys specifically intended to fabri-
cate endosseous implants for osteoporotic patients [60].
On the contrary, research on bioceramics (even playing a minor
role compared with metals in the treatment of osteoporosis)
envisions this scenario in a different way and often deals with
the specific case of osteoporotic bone. In this sense, calcium phos-
phatebioceramics [61,62] and SiO
2
-based mesoporous materials
have been widely proposed as potential local antiosteoporotic drug
delivery systems [63], when used for void fillers in fracture fixa-
tion, bone grafting or augmentation.
Combinations of biomaterials with cellular therapy and local
drug delivery are of enormous interest because of the great oppor-
tunities that they offer to this problem [64–66]. However, the tech-
nical and biological problems associated with the cells or materials
to be used are important. These include (i) the limited cell viability;
(ii) the transient mechanical properties of the materials once im-
planted until they are substituted by regenerated bone; and (iii)
the biological integration at the specific bone site where they are
implanted. To solve these problems, in vitro and in vivo tests
should be performed in experimental conditions that mimic as
much as possible the most prevalent situations associated with
bone pathologies: estrogen depletion, diabetes mellitus, ageing
and treatment with glucocorticoids. To make progress in this area,
standard procedures to collect, manipulate and store mesenchymal
osteoprogenitors (such as the bone marrow) should be defined.
Thereafter, it would be possible to start considering the fabrication
of a medical device following criteria accepted by national and
international regulatory agencies for on-demand administration.
3.1. Metallic implants
Since osteoporotic bone is much more fragile than healthy bone,
metallic implants used to treat osteoporotic bone fractures have to
be designed differently. One strategy consists in increasing the
contact surface area between bone and implant. This can be done
by increasing the diameter of osteosynthesis screws [67]. Another
approach is to use locked osteosynthesis plates [68]. In the latter
case, plate loosening is only possible if all screws become loose
simultaneously. This is in large contrast with unlocked plates
whose fixation onto the bone relies on the compressive action of
screws. A third approach consists in designing completely new
Fig. 3. Displaced intracapsular fracture (left) and trochanteric fracture (right).
D. Arcos et al. / Acta Biomaterialia xxx (2014) xxx–xxx 5
Please cite this article in press as: Arcos D et al. The relevance of biomaterials to the prevention and treatment of osteoporosis. Acta Biomater (2014),
http://dx.doi.org/10.1016/j.actbio.2014.01.004
implants, such as expandable spacers for vertebral height restora-
tion (e.g. VBS (DePuy Synthes), Spinejack Vexim (Vexim), Kiva
(Benvenue Medical)) or cannulated screws (e.g. Matrix Spine Sys-
tem (Depuy Synthes)) to permit cement injection through the
screw [69]. Cement injection through the screws (Figs. 4 and 5) re-
sults in an increase in the resistance of the fractured vertebral body
and avoids the pull-out of screws in osteoporotic spine [70,71].
Another interesting topic is the role of surface roughness and
wettability in implant osteointegration [72]. This strategy has
achieved some degree of success especially in metallic implants
for periodontal surgery. For instance, modifications of the microto-
pography of titanium implants have demonstrated enhanced
osteointegration [73]. Prospective studies on implants with rough
surfaces evidence very promising clinical results compared with
those with smoother surfaces [74]. In addition, the recent develop-
ment of nanotechnology in the biomaterials field also allows the
incorporation of nanofeatures on the implant surface. In this sense
there are some studies that evidence the significance of nanoto-
pography in the success of peri-implant bone formation [75,76].
In addition, the surface wettability is closely related to the surface
micro/nanoroughness and also influences the osteoblast behavior.
In principle, hydrophilic surfaces enhance osteoblast maturation
[77], thus leading to better clinical results [78].
The coating of the surface of metallic implants has been suc-
cessfully applied for decades to improve their bone-binding prop-
erties, with the cementless hip endoprosthesis and dental implants
being the best-known examples. In particular, calcium phosphate
coatings have been applied by various techniques, e.g. plasma
spraying, sputtering techniques and sol–gel coating [79–82].
Although an increased bone-binding ability has been found, a re-
cent review points out that long-term clinical studies indicate con-
tradictory results [83]. However, it may be envisioned that in
osteoporotic bone, a surface modification of metallic implants, be
it by calcium phosphate or by drug-releasing coatings, will help
to improve the clinical outcome, at least in the short-term perfor-
mance when the primary stability is needed.
3.2. Cements
PMMA cement is the material of choice for the reinforcement of
metallic implant fixation or osteoporotic bone due to its high
mechanical properties and low cost. However, PMMA cement has
very important drawbacks such as monomer toxicity [84], risk of
bone necrosis due to a highly exothermic setting reaction [85], ab-
sence of biodegradation that may lead to fatigue failure [86], or too
high material stiffness that may increase the fracture risk of verte-
bra adjacent to PMMA-reinforced vertebra [87]. As a result, various
cements have been proposed to replace PMMA cement, but their
success remains very limited due to toxicity, regulatory issues,
price or mechanical properties. For example, a few years ago the
company Orthovita proposed a dual-paste cement called Cortoss.
This cement was inspired from the composition of dental cement,
i.e. it consists of a matrix of bis-GMA (2,2-bis[4-(2-hydroxymeth-
acryloxypropyl)phenyl] propane), bis-EMA (2,2-bis[4-(2-methac-
ryloxyethoxy)]phenyl propane) and TEGDMA (triethylene glycol
dimethacrylate) reinforced with bioactive glass particles [88]. Cor-
toss presents better handling, higher mechanical properties and
lower toxicity than PMMA cements. Unfortunately, it has had lim-
ited success, possibly on the grounds of price (higher production
costs than PMMA cements). More recently, a silicone cement called
VK100 was proposed by the company BonWRX. This dual-paste ce-
ment contains dimethyl methylvinyl siloxanes (87%), barium sul-
fate powder (14%) and a platinum catalyst (15 ppm as metal) in
the first component, and dimethyl methylvinyl siloxanes (78%),
barium sulfate powder (15%) and a methylhydrogensiloxane
cross-linker (7%) in the second component. Unfortunately, preli-
minary results for bone augmentation applications (‘‘elastoplasty’’)
are very poor with more than 60% leakage (cement flowing outside
the targeted location, e.g. into the spinal canal) and pulmonary
embolism [89]. The very slow setting reaction was also mentioned.
Considering the poor biological properties of polymer cements,
quite a few ceramic cements have been proposed for bone aug-
mentation procedures. Interesting candidates have included cal-
cium phosphate cements (CPCs), but the results have been rather
disappointing [90–93]. One main issue with CPCs is their poor
mechanical (shear stress) properties. Thus, CPCs can at most be
used in load-sharing sites. Also, several deaths have been reported
after the use of Norian XR CPC [94,95], suggesting some biocom-
patibility issues of CPCs in spinal applications. Higher mechanical
properties were achieved with a calcium aluminate cement (Xera-
spine from Doxa AB), but the results were not very good either
[96]. The last ceramic cement that should be mentioned here is
Plaster of Paris (calcium sulfate hemihydrate). This material readily
dissolves in vivo due to its comparatively high solubility (i.e. with-
out the help of osteoclasts), but nevertheless has been proposed for
bone augmentation [97,98] and bone void filling applications, e.g.
for the filling of the Kiva device. Besides bone augmentation,
ceramic cements have also been used for screw augmentation
[99–102]. Some results are very promising [103,104], but more
data are needed to assess the long-term success of this approach.
3.3. Bioceramics for bone tissue regeneration
Altogether, calcium phosphate ceramics and related com-
pounds, i.e. calcium phosphate cements, bioglasses and calcium
sulfate cements, represent the most important class of biomateri-
als for bone regeneration. Different types of calcium phosphate
ceramics, glass–ceramics and glasses are currently being used
and further developed for bone reconstruction and repair. In the
present section the most prominent inorganic systems and, where
appropriate, their composites in combination with polymers are
described, highlighting the effects of ion release to induce osteo-
genesis and angiogenesis, i.e. the two functions required for
effective bone tissue regeneration. In this sense, the design and
Fig. 4. Osteoporotic vertebral fractures in T12 and L4. Vertebroplasty in T12 and reduction and fixation with cannulated screws and cement in L4.
6D. Arcos et al. / Acta Biomaterialia xxx (2014) xxx–xxx
Please cite this article in press as: Arcos D et al. The relevance of biomaterials to the prevention and treatment of osteoporosis. Acta Biomater (2014),
http://dx.doi.org/10.1016/j.actbio.2014.01.004
development of porous ceramics has attracted much attention in
the last years. Not only pore size, but also pore distribution can
play a fundamental role in bone regeneration, angiogenesis and
implant degradation [105]. The incorporation of free-form prepara-
tion methods such as 3-D printing to the biomaterials field has al-
lowed the design of hierarchical pore structures to facilitate these
processes [106]. An interconnected macropore structure of 150–
1000
l
m allows cell colonization and enhances the diffusion rates
to and from the center of a scaffold, as well as angiogenesis and
bone in growth [107,108]. Small pores allow phagocytic cells to ad-
here and resorb the scaffolds, whereas larger pores encourage the
invasion of new vessels and in growth of bone tissue [109].
3.3.1. Calcium phosphates
Calcium phosphate constitutes the inorganic mineral phase in
mammalian bone and teeth. Therefore it is well recognized by
the body and biocompatible according to all current standards
[110–112]. The calcium phosphate mineral in bone consists of
nanocrystalline platelets of biological apatite, which chemically is
a hydroxyapatite with ionic substitutions, mainly carbonate
[113]. A number of synthetic calcium phosphate ceramics are on
the market as bone substitution material, with hydroxyapatite,
Ca
5
(PO
4
)
3
OH (HAP), and b-tricalcium phosphate, b-Ca
3
(PO
4
)
2
(b-
TCP) and combinations of them (biphasic calcium phosphate(BCP))
being the most prominent. They are available in different morphol-
ogies (typically as solid or porous blocks or as granules with differ-
ent particle sizes) and with different origins (fully synthetic or
derived from biological sources such as animal bone or chemically
transformed calcareous algae) [114]. In general, they are well ac-
cepted by the body, but as ceramics they are brittle by nature
and therefore not able to withstand the mechanical challenge in
a larger defect. With time, newly formed bone grows onto and into
calcium phosphate ceramics and finally leads to stable osteointe-
gration [115]. The resorption of calcium phosphate ceramics typi-
cally involves acidic dissolution by osteoclasts [116,117].
3.3.2. Bioactive glasses
Of all bioceramics, glasses have a special position due to their
ability to rapidly release different ions, but also to strongly bind
to bone through the formation of an apatite-like phase in the
bone–implant interface (‘‘bioactivity’’). Depending on their chemi-
cal composition, bioactive glasses can be resorbed and their degra-
dation byproducts can stimulate the osteogenic pathways in MSCs
present at the fracture location. Indeed, the tailored effect of disso-
lution products from bioactive glasses on cellular responses, e.g. to
upregulate the expression of genes controlling osteogenesis and to
enhance vascular endothelial growth factor (VEGF) secretion
in vitro to induce vascularization, are attractive qualities of bioac-
tive glasses (and their composites) in the context of bone regener-
ation strategies [118]. For instance, the effect of silicate ions was
recently investigated in relation to proliferation, osteogenic differ-
entiation and cell signaling pathways of bone marrow stromal cells
[119]. It has also been shown that cells where the calcium sensing
receptor (CaSR) is present, such as MSCs and endothelial cells, re-
spond to specific calcium concentrations in the environment by
migrating, proliferating and differentiating, expressing alkaline
phosphatase and collagen 1, and mineralizing and forming tubules,
respectively [120]. Therefore, it is hypothesized that any biomate-
rial with the appropriate calcium-releasing capacity would be a
good candidate for bone regeneration where angiogenesis is
necessary.
Typical silicate-based compositions such as 45S5 (45 wt.% SiO
2
,
24.5 wt.% Na
2
O, 24.5 wt.% CaO, 6 wt.% P
2
O
5
) are characterized by a
high surface bioactivity enabling strong bonding to bone tissue
[121,122] leading also to stimulating effects on osteogenesis
[123] and angiogenesis [124]. Starting with the classic 45S5 Bio-
glass
Ò
composition [121], a great number of silicate systems incor-
porating specific ions into the silicate network is continuously
developed. The typical ions that are under investigation are mag-
nesium, strontium, silver, iron, copper, boron, potassium, lithium,
cobalt, fluoride and zinc [121]. Absorbable calcium phosphate
glasses are able to solubilize completely with degradation times
ranging from days to years, depending on their chemical composi-
tion. The vitreous network of [PO
4
] tetrahedra is easily hydrolyzed.
The chemical stability of these glasses can only be modified by
including different metallic oxides, such as Al
2
O
3
, ZnO, Fe
2
O
3
and
TiO
2
, into the 3-D vitreous network. TiO
2
has proved to be very effi-
cient, given its four-valence state, linking to four phosphate tetra-
hedra [125]. The capacity of calcium phosphate glasses to promote
cell adhesion [126] and to induce vessel formation at the site of
implantation [127] can be interpreted in terms of ion release.
Being of great relevance in the context of osteoporosis-combat-
ing materials, the effect of specific ions on bone-resorbing osteo-
clast cells must be considered. For example, some researchers
have investigated the addition of strontium ions into silicate glasses
as an effective approach for developing improved bioactive glasses
[128], considering the positive results achieved with strontium
ranelate (SrR) applied as a drug to treat and prevent osteoporosis
especially in post-menopausal women. Dedicated in vivo studies
to assess potential bone healing enhancement in osteoporotic bone
by grafting with bioactive glasses are still scarce [129], indicating a
need for future research to realistically consider bioactive glasses as
osteoporosis combating substances. It is also important to note that
specific morphologies of silicate bioactive glasses (and silica), e.g.
with a mesoporous structure [130], are attractive systems which
enable the incorporation of a drug delivery function to enhance
the intrinsic bioactive character of the inorganic silicate carrier.
3.4. Associations of biomaterials with biological entities: gene and
cellular therapies
3.4.1. Perspectives of gene therapies for bone regeneration purposes
As there is clearly a need for rapid bone regeneration after a
fracture in osteoporotic bone, people have wondered for decades
Fig. 5. Osteoporotic vertebral fractures treatment. Fixation with cannulated screws and cement.
D. Arcos et al. / Acta Biomaterialia xxx (2014) xxx–xxx 7
Please cite this article in press as: Arcos D et al. The relevance of biomaterials to the prevention and treatment of osteoporosis. Acta Biomater (2014),
http://dx.doi.org/10.1016/j.actbio.2014.01.004
how bone growth can be stimulated by adding osteogenic
compounds to biomaterials. This has led to systems providing
local drug delivery, e.g. of bone morphogenetic proteins (BMPs)
[131–133] or of angiogenic proteins such as VEGF [134,135], e.g.
from polymers or ceramics. The release typically consists of a burst
in the first days or weeks. The preparation and incorporation of
proteins into biomaterials are usually costly. Another approach is
a local gene therapy, provided by suitable biomaterials in direct
bone contact. Gene therapy involves the delivery of DNA which
can induce the production of the encoded protein after uptake by
cells (so-called ‘‘transfection’’). To accomplish this goal, suitable
carriers are needed as nucleic acids alone cannot penetrate the
cell wall. Furthermore, they are subject to rapid biodegradation
by nucleases in the body.
Two types of carriers for nucleic acids are currently discussed:
viruses and nanoparticles. These can be taken up by cells, together
with their cargo of DNA. Viral transporter systems have the advan-
tage of a very high transfection efficiency, but concerns remain
about possible side-effects [136–138]. Nanoparticles can be organ-
ic (e.g. liposomes or polymeric nanoparticles) or inorganic in nat-
ure [139,140]. Their efficiency is typically lower than that of
viruses, but they have the advantage that they can be more easily
controlled due to their non-biological nature.
The advantage of such local gene delivery is the comparative
ease of producing DNA on a milligram scale and the long-lasting
action. In principle, all kinds of cells around such a DNA-releasing
implant can take up nanoparticles and start to produce DNA. In this
way, proteins such as BMPs or VEGF can be produced and delivered
in vivo to induce bone growth and vascularization [141–145].It
was recently shown that it is possible to induce the production
of BMP-7 and VEGF-A from a paste of DNA-functionalized calcium
phosphate nanoparticles. Thereby, osteoconductivity (by calcium
phosphate) and osteoinduction (by production of the proteins
around the implantation site) were combined [146].Fig. 6 shows
a scanning electron microscopy image of DNA-carrying calcium
phosphate nanoparticles. Note that in general, nanoparticles have
not only been discussed as delivery agents, but they can also be
used to increase the mechanical strength after embedding into a
polymeric matrix (e.g. [147]). This follows the concept of a biomi-
metic hierarchically structured material, mimicking bone itself
[148–150].
Another option within the framework of gene therapy is the
silencing of selected genes by administration of small-interfering
RNA (siRNA). As such, the production of proteins which, for exam-
ple, inhibit bone growth or vascularization, can be down-regulated.
This is called gene silencing, another highly promising method
within the framework of gene therapy [151]. Again, suitable carri-
ers such as nanoparticles are necessary [152–154]. They have been
successfully tested, for example, to down-regulate inflammatory
genes [155] or osteopontin and osteocalcin in osteoblasts [156].
Transfection by nanoparticles is always temporary, i.e. after a
few weeks or months (depending on the release kinetics from
the scaffold) it ceases, ideally after bone healing in complete. This
increases confidence in this approach after application in a bone
defect.
Subjecting osteoporotic bone to gene therapy to improve its
strength is likely to involve a local delivery of a nanoparticle-based
system which carries suitable DNA. It is conceivable that this might
work to prevent later fractures.
3.4.2. Perspectives in the use of cell therapy in the reconstruction of
osteoporotic bone
Recent developments of biomaterials reviewed in this paper
converge on the requirement for biomaterials that foster
osteoinduction and osteogenesis to support and augment bone
healing after osteoporotic fractures. Indeed, the reconstruction of
osteoporotic bone faces significant difficulties with the solutions
currently available. Osteoporotic bone, currently defined by its de-
creased quality (not quantity) leading to a mechanically incompe-
tent biological material [157], manifests itself by the occurrence of
so-called osteoporosis-related fractures. The immediate conse-
quences are bone collapse in metaphyseal compression fractures
with bone defects and joint malfunction, comminution and de-
layed union or non-union after diaphyseal low-energy fractures
with thin cortices, or periprosthetic complex fractures on sclerotic
bone surrounding implants. These problematic fractures seldom
heal, restricting patient mobility and eventually leading to suffer-
ers becoming bed-ridden or even dying. Therefore, therapeutic tar-
gets can be defined for which advances in biomaterials are badly
needed. Basically, it has become clear that a biological problem is
underlying the occurrence of this brittle, non-resistant bone. Fre-
quently underestimated, biological insufficiency leads to signifi-
cant bone-healing problems.
While some osteoinductivity is observed with various biomate-
rials, osteogenesis is required to obtain satisfactory bone repair
and directly relies on osteoprogenitors and derived osseous cell
lines [158]. However, the number of osteoprogenitors available in
the vicinity of a fracture is unclear and unpredictable. Furthermore,
the number of available progenitors decreases drastically with age,
and estimates of stromal cells in the bone marrow drop to
one-eighth from young adulthood to old age. Consequently, elderly
patients with osteoporosis who are more prone to fractures are
associated with limited biological capabilities to heal bone. In that
context, it is interesting to use a cell-based therapy. However, de-
spite significant advances in this field, efforts are hampered by var-
ious constraints: (i) the large number of in vivo and in vitro studies
that are required in a pre-clinical stage; (ii) the safety and efficacy
issues; and finally (iii) the regulatory and legal constraints.
Three main cellular therapy strategies have been developed and
translated into bone regenerative clinical solutions [159]. Mesen-
chymal stem cells (MSCs) from fresh, concentrated autologous
bone marrow have been widely used to enhance bone healing at
non-unions [160], usually in adults at an early age. Only a slight
improvement in biological regenerative potential can be expected
when 1000–1400 MSCs are obtained per 2 ml bone marrow aspi-
rate in young patients; more than 55,000 MSCs per injury are re-
quired. This autologous treatment allows for augmentation of
surgical treatment without the need to introduce cell-based med-
ication into the patient, avoiding significant legal barriers when the
whole process occurs during surgery.
To further increase the biological regenerative potential, grow-
ing bone marrow MSCs from patients may produce millions of
MSCs within a few weeks. Although this manipulation transforms
the cell product into an advanced therapy medicinal product
Fig. 6. DNA-loaded calcium phosphate nanorods which are able to induce the
formation of BMP and VEGF.
8D. Arcos et al. / Acta Biomaterialia xxx (2014) xxx–xxx
Please cite this article in press as: Arcos D et al. The relevance of biomaterials to the prevention and treatment of osteoporosis. Acta Biomater (2014),
http://dx.doi.org/10.1016/j.actbio.2014.01.004
(ATMP) that requires fabrication in certified GMP (good manufac-
turing practices) facilities, proposals are currently being developed
through clinical trials. A major barrier to the use of this solution
with elderly and osteoporotic patients is the limited number of
stromal cells in the bone marrow progenitor pool. Other sources
of MSCs face similar problems in elderly patients and osteogenic
line proliferation and differentiation may be further limited. Allo-
geneic expanded cells would be an ideal solution but their safety
and efficacy remains unproven and the significant problems they
present do not appear to have a clear solution.
A third strategy under development involves MSC growth on
biomaterials. If an appropriate combination of biomaterial, cell
dosage and stimulating molecules could be found, the structural
and biological potential of this approach would facilitate an ade-
quate bone substitute through tissue engineering to create a mate-
rial similar to real bone. Only a few publications have addressed
clinical cases treated with this strategy [161], and in terms of
autologous design, its application in osteoporotic patients is a long
way off.
Major issues remain to be solved before these advances can be
safely applied in clinical trials in elderly patients with osteoporotic
fractures or complications. Many questions remain about the
adherence and osteogenic differentiation of osteoprogenitors on a
variety of biomaterials. Furthermore, it remains unproven whether
osteoprogenitors grown on biomaterials maintain their adherence
and thus their location; or if the functional capabilities, in particu-
lar the osteogenic potential, of these cells are retained after surgi-
cal implantation.
However, even if serious barriers still need to be overcome, re-
search efforts are in place and the confirmed requirements for
these particular targets will probably transform in the coming
years the way we understand the clinical potential of bone tissue
engineering based on advanced biomaterials and cell therapy
solutions.
To conclude, the biomaterials used for osteoporosis-related
clinical indications are fairly traditional, including metals, poly-
mers and ceramics. Their design (shape, composition) is generally
adapted to better accommodate osteoporosis-related require-
ments. Part of the gap between clinical biomaterials and academic
research can be explained by increasingly stringent safety regula-
tions, as well as cost pressures. This will be discussed in more de-
tails in the next sections.
4. Biomaterials and osteoporosis. The industrial view point
The previous sections have shown that there is a great need to
improve the treatment of osteoporotic patients before and after the
occurrence of a bone fracture. Various routes for therapeutic
progress have been highlighted, including tissue engineering,
drug-loaded bone graft substitutes and gene therapy. Despite these
efforts, little progress has been seen clinically. In fact, the launch of
BMP-loaded products a decade ago was the last important innova-
tion. This impression is confirmed by the evolution of pre-market
approvals (PMAs) issued by the FDA (Fig. 7). A PMA is compulsory
for any device that does not have any equivalent on the market. In
other words, the most innovative products have to go through a
PMA review process. Over the last 10 years, the number of PMAs
accepted by the FDA has decreased dramatically. Specifically, 23
PMAs were accepted from 2003 to 2007. This number dropped to
8 from 2008 to 2012. Simultaneously, there was a 50% increase
in 510 k notifications, which are issued for products equivalent
to existing products that already have FDA approval. These two
trends mean that companies in the US are shifting their efforts
from innovation to incremental improvements of existing technol-
ogies. The situation in Europe is similar.
Many factors have contributed to this decrease in the number of
innovations. The most obvious one is related to the laws regulating
medical devices. Over the past decades, the European authorities
have strengthened their directives, not only by asking for more
data per product, but also by transferring certain products into
higher product classes. This is the case for joint prostheses that
were class II products until March 2010, and which are now class
III products. In addition, new ISO standards are continuously ap-
proved, which means that more tests have to be done to apply
for a CE marking. It is clear that ISO standards are not compulsory,
but it is often easier to perform the study than to explain why it
has not been performed. This is particularly disturbing for tests
that have been shown to present important weaknesses, such as
ISO 10993-5 (Biological evaluation of medical devices—Part 5:
Tests for in vitro cytotoxicity), or the so-called ‘‘bioactivity test’’
(ISO 23317; Implants for surgery—In vitro evaluation for apatite-
forming ability of implant materials). In both cases, false positives
and false negatives can be found (for ISO 23317, see Refs.
[162,163]). The trend towards stricter regulations is not expected
to stop soon. In fact, much stricter directives have been proposed
following the recall of PIP breast implants and DePuy metal-on-
metal hip prostheses. These directives are currently awaiting
approval.
In 1985, the European Parliament decided to harmonize the
laws regulating medical devices within Europe to facilitate the
transfer of goods. Directives were defined and implemented. Even
though CE-marked products can access the European market, other
hurdles are still in place. For example, the French authorities ask
companies to register their products prior to any reimbursement.
In the US, the government decided a few years ago to stop reim-
bursing vertebral bone augmentation procedures following the
publication of two articles demonstrating an absence of significant
effect between the treatment group and a ‘‘placebo’’ group
[164,165]. The fact that numerous publications have shown the
limitations of these two studies has not changed the situation. In
fact, medical device companies are increasingly asked to show
the effectiveness of their product before reimbursement is issued.
Since artificial joints may only prove their efficacy after 10 years,
this requirement is particularly questionable and cost-intensive.
In addition, proving the efficacy of an implant may require multi-
ple in vivo or clinical studies to support broad claims. For example,
whereas it was possible in the past to perform only one in vivo
study for a ‘‘bone void filler, the authorities now tend to require
more than one in vivo study. This is often very costly and may take
years. Once a product is accepted for sale, governments may decide
on its value. According to the French ‘‘Liste des Produits et Presta-
tions’’ (LPP), a resorbable interference screw for ligament fixation
is worth 234.16.
Currently, there is a trend towards the alignment of pharma and
orthopedic product approvals. However, there is a major
Fig. 7. FDA pre-market approvals (PMAs) and pre-market notifications (510 k) over
the past 10 years. Sources: http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/
cfPMA/pma.cfm and http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfPMN/
pmn.cfm.
D. Arcos et al. / Acta Biomaterialia xxx (2014) xxx–xxx 9
Please cite this article in press as: Arcos D et al. The relevance of biomaterials to the prevention and treatment of osteoporosis. Acta Biomater (2014),
http://dx.doi.org/10.1016/j.actbio.2014.01.004
difference: whereas pharma products have generally a systemic
action, orthopedic products have a local action (narrow range of
indication). In other words, osteosynthesis plates or orthopedic im-
plants are bone/joint specific, so each plate/implant requires a sep-
arate registration. The limited market size and increasingly large
regulatory burdens are obviously important aspects of, and brakes
in, the decision-making processes occurring during product devel-
opment. In fact, many R&D departments are nowadays focused on
maintaining the product portfolio and reducing costs, rather than
developing new products.
Governments are facing a dilemma: if they tighten the rules to
obtain the CE mark for a new medical product, they restrict inno-
vation, hence reducing the chances of seeing new therapies; if they
do not tighten the rules after the recent medical device scandals
(PIP breast implants, DePuy metal-on-metal prosthesis), their elec-
tors might punish them at the next elections or even sue them as
seen with the contaminated blood scandal. Currently, the former
strategy is pursued worldwide, to the detriment of all clinical
needs and research efforts [166].
5. Conclusions
Osteoporosis is a disease that has become a worldwide chal-
lenge and comprises clinical, social and economic issues. This is
mainly due to the increase in life expectancy, and therefore society,
health systems and industry should be aware of this problem, as an
ageing population will be more prone to osteoporosis.
The main clinical consequences of osteoporosis are fractures.
The success of biomaterials for fracture fixation in osteoporotic pa-
tients, or simply for bone augmentation treatments, is impaired by
the poor quality of bone and the decreased osteoblastic activity. In
this sense, although there are a number of biomaterials to treat
problems with bone on the market, they are not necessarily appro-
priate to address osteoporotic bone. Unsuccessful implantation can
result in crossing the line between frailty and disability in osteopo-
rotic patients.
Clinicians, biomaterials scientists and industrial advisors are
making important efforts to improve current implants and their
applications, as well as provide new alternatives. Compounds able
to stimulate the bone regeneration such as calcium phosphates
(both ceramics and cements), calcium sulfates or bioglasses are
being widely considered, especially associated with local drug
and/or gene delivery as well as with cell therapy.
However, all these efforts only will be fruitful if these new
biomaterials can be successfully developed and commercialized.
Currently, the level of commercial innovation remains well below
expectation and the situation is only expected to worsen due to
more stringent certification requirements and higher cost pres-
sures. Thus, those biomaterials that will play a major role in the
prevention and treatment of osteoporotic conditions will most
likely feature the following points:
– A well-developed definition of unmet needs.
– It will most likely be indication-specific.
– It will enable quantifiable clinical benefits to be proven in level
11 clinical studies.
– It will be profitable.
– A collaborative petition to agencies such as FDA could be
helpful.
Consequently, more research is necessary, driven by clinical de-
mand, to solve this problem. The fact that the current situation is
difficult to manage should not prevent us from seeking new solu-
tions. For this purpose, a close cooperation between fundamental
research, industrial research, clinical research and regulatory
bodies is required for the future. This will not be available for free
(i.e. without financial costs), but will pay off in the long run for
everybody.
In addition, the presence of new patients with new characteris-
tics and new needs, mainly (but not exclusively) frail older people,
should change not only the way to provide their management and
treatment, but also the aims of the care and the way to assess the
technological improvements. Regarding this last issue it should
concern both the changes on how to organize the delivery of the
care provided to these patients, the characteristics of the devices
and its outcomes as well. This new model to assess the efficacy
and effectiveness of the new technologies in the new patients
should prompt a change in the rules of the regulatory agencies in
order to adapt their procedures to the current needs of both the pa-
tients, the health systems and the industry, thus contributing to
the well-being of the patients, the sustainability of the health sys-
tems and the competitiveness of the industry.
Acknowledgements
The Spanish and European Network of Excellence for the Pre-
vention and Treatment of Osteoporotic Fractures ‘‘Ageing’’ is finan-
cially supported by the Ministerio de Economía y Competitividad
through the project CSO2010-11384-E.
Appendix A. Figures with essential color discrimination
Certain figures in this article, particularly Figs. 1, 2, 5 and 7, are
difficult to interpret in black and white. The full color images can
be found in the on-line version, at http://dx.doi.org/10.1016/
j.actbio.2014.01.004
References
[1] Planell JA, Navarro M. Challenges of bone repair. In: Planell, Best, Lacroix,
Merolli, editors. Bone repair biomaterials. Cambridge: CRC Press; 2009.
[2] Streubel PN, Ricci WM, Wong A, Gardner MJ. Mortality after distal femur
fractures in elderly patients. Clin Orthop Relat Res 2011;469:1188–96.
[3] National Osteoporosis Foundation. http://www.nof.org.
[4] Hoerger TJ, Downs KE, Lakshmanan MC, Lindrooth RC, Plouffe Jr L, Wendling
B, et al. Healthcare use among U.S. women aged 45 and older: total costs and
costs for selected postmenopausal health risks. J Womens Health Gend Based
Med 2007;8:1077–89.
[5] Rissanen P, Aro S, Sintonen H, Asikainen K, Slätis P, Paavolainen P. Costs and
cost-effectiveness in hip and knee replacements: a prospective study. Int J
Technol Assess Health Care 1997;13:574–88.
[6] Day JC. Population projections of the United States by age, sex, race and
Hispanic origin: 1995 to 2050. Washington, DC: US Government Printing
Office; 1996.
[7] Ray NF, Chan JK, Thamer M, Melton III LJ. Medical expenditures for the
treatment of osteoporotic fractures in the United States in 1995: report from
the National Osteoporosis Foundation. J Bone Miner Res 1997;12:24–35.
[8] Chrischilles E, Shireman T, Wallace R. Costs and health effects of osteoporotic
fractures. Bone 1994;15:377–86.
[9] Melton LJ. Hip fractures: a worldwide problem today and tomorrow. Bone
1993;14:1–8.
[10] Kanis JA, Johnell O. Requirements for DXA for the management of
osteoporosis in Europe. Osteoporos Int 2005;16:229–38.
[11] Manton KG, Gu X. Changes in the prevalence of chronic disability in the
United States black and nonblack population above age 65 from 1982 to
1999. Proc Natl Acad Sci USA 2001;98:6354–9.
[12] Boyd CM, Landefeld CS, Counsell SR, Palmer RM, Fortinsky RH, Kresevic D,
et al. Recovery of activities of daily living in older adults after hospitalization
for acute medical illness. J Am Geriatr Soc. 2008;56:2171–9.
[13] Clegg A, Young J, Iliffe S, Rikkert MO, Rockwood K. Frailty in elderly people.
Lancet 2013;381:752–62.
[14] Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, et al.
Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci
2001;56:M146–56.
[15] Rodríguez-Mañas L, Féart C, Mann G, Viña J, Chatterji S, Chodzko-Zajko W,
et al. Searching for an operational definition of frailty: a Delphi method based
consensus statement: the frailty operative definition-consensus conference
project. J Gerontol A Biol Sci Med Sci 2013;68:62–7.
[16] Espeland MA, Gill TM, Guralnik J, Miller ME, Fielding R, Newman AB, et al.
Designing clinical trials of interventions for mobility disability: results from
10 D. Arcos et al. / Acta Biomaterialia xxx (2014) xxx–xxx
Please cite this article in press as: Arcos D et al. The relevance of biomaterials to the prevention and treatment of osteoporosis. Acta Biomater (2014),
http://dx.doi.org/10.1016/j.actbio.2014.01.004
the lifestyle interventions and independence for elders pilot (LIFE-P) trial. J
Gerontol A Biol Sci Med Sci 2007;62:1237–43.
[17] Felsenberg D, Silman AJ, Lunt M, Armbrecht G, Ismail AA, Finn JD, et al.
Incidence of vertebral fracture in europe: results from the European
Prospective Osteoporosis Study (EPOS). J Bone Miner Res 2002;17:716–24.
[18] Gauthier A, Kanis JA, Jiang Y, Martin M, Compston JE, Borgstrom F, et al.
Epidemiological burden of postmenopausal osteoporosis in the UK from 2010
to 2021: estimations from a disease model. Arch Osteoporos 2011;6:179–88.
[19] Seeman E, Delmas PD. Bone quality—the material and structural basis of bone
strength and fragility. N Engl J Med 2006;354:2250–61.
[20] Consensus conference. Osteoporosis. JAMA 1984;252:799–802.
[21] Ismail AA, Cooper C, Felsenberg D, Varlow J, Kanis JA, Silman AJ, et al. Number
and type of vertebral deformities: epidemiological characteristics and
relation to back pain and height loss. European Vertebral Osteoporosis
Study Group. Osteoporos Int 1999;9:206–13.
[22] Ettinger B, Black DM, Mitlak BH, Knickerbocker RK, Nickelsen T, Genant HK,
et al. Reduction of vertebral fracture risk in postmenopausal women with
osteoporosis treated with raloxifene: results from a 3-year randomized
clinical trial. Multiple Outcomes of Raloxifene Evaluation (MORE)
Investigators. J Am Med Assoc 1999;282:637–45.
[23] Lewiecki EM, Compston JE, Miller PD, Adachi JD, Adams JE, Leslie WD, et al.
Official positions for FRAX(R) bone mineral density and FRAX(R)
simplification from Joint Official Positions Development Conference of the
International Society for Clinical Densitometry and International
Osteoporosis Foundation on FRAX(R). J Clin Densitom 2011;14:226–36.
[24] Gonzalez-Macias J, Marin F, Vila J, Diez-Perez A. Probability of fractures
predicted by FRAX(R) and observed incidence in the Spanish ECOSAP Study
cohort. Bone 2012;50:373–7.
[25] Dennison EM, Compston JE, Flahive J, Siris ES, Gehlbach SH, Adachi JD, et al.
Effect of co-morbidities on fracture risk: findings from the Global
Longitudinal Study of Osteoporosis in Women (GLOW). Bone
2012;50:1288–93.
[26] Hans DB, Kanis JA, Baim S, Bilezikian JP, Binkley N, Cauley JA, et al. Joint
official positions of the International Society for Clinical Densitometry and
International Osteoporosis Foundation on FRAX
Ò
. Executive summary of the
2010 Position Development Conference on Interpretation and use of FRAX
Ò
in
clinical practice. J Clin Densitom 2011;14:171–80.
[27] Zioupos P, Hansen U, Currey JD. Microcracking damage and the fracture
process in relation to strain rate in human cortical bone tensile failure. J
Biomech 2008;41:2932–9.
[28] Diez-Perez A, Guerri R, Nogues X, Caceres E, Pena MJ, Mellibovsky L, et al.
Microindentation for in vivo measurement of bone tissue mechanical
properties in humans. J Bone Miner Res 2010;25:1877–85.
[29] Guerri-Fernandez RC, Nogues X, Quesada Gomez JM, Torres Del Pliego E, Puig
L, Garcia-Giralt N, et al. Microindentation for in vivo measurement of bone
tissue material properties in atypical femoral fracture patients and controls. J
Bone Miner Res 2013;28:162–8.
[30] Garnero P, Sornay-Rendu E, Claustrat B, Delmas PD. Biochemical markers of
bone turnover, endogenous hormones and the risk of fractures in
postmenopausal women: the OFELY study. J Bone Miner Res
2000;15:1526–36.
[31] Vasikaran S, Eastell R, Bruyere O, Foldes AJ, Garnero P, Griesmacher A, et al.
Markers of bone turnover for the prediction of fracture risk and monitoring of
osteoporosis treatment: a need for international reference standards.
Osteoporos Int 2011;22:391–420.
[32] Kanis JA, McCloskey EV, Johansson H, Cooper C, Rizzoli R, Reginster JY.
European guidance for the diagnosis and management of osteoporosis in
postmenopausal women. Osteoporos Int 2013;24:23–57.
[33] Lekamwasam S, Adachi JD, Agnusdei D, Bilezikian J, Boonen S, Borgstrom F,
et al. A framework for the development of guidelines for the management of
glucocorticoid-induced osteoporosis. Osteoporos Int 2012;23:2257–76.
[34] Rizzoli R, Adachi JD, Cooper C, Dere W, Devogelaer JP, Diez-Perez A, et al.
Management of glucocorticoid-induced osteoporosis. Calcif Tiss Int
2012;91:225–43.
[35] Kaufman JM, Reginster JY, Boonen S, Brandi ML, Cooper C, Dere W, et al.
Treatment of osteoporosis in men. Bone 2013;53:134–44.
[36] Diez-Perez A, Hooven FH, Adachi JD, Adami S, Anderson FA, Boonen S, et al.
Regional differences in treatment for osteoporosis. The Global Longitudinal
Study of Osteoporosis in Women (GLOW). Bone 2011;49:493–8.
[37] Diez-Perez A, Olmos JM, Nogues X, Sosa M, Diaz-Curiel M, Perez-Castrillon JL,
et al. Risk factors for prediction of inadequate response to antiresorptives. J
Bone Miner Res 2012;27:817–24.
[38] Diez-Perez A, Adachi JD, Agnusdei D, Bilezikian JP, Compston JE, Cummings
SR, et al. Treatment failure in osteoporosis. Osteoporos Int 2012;23:2769–74.
[39] Rizzoli R, Reginster JY, Boonen S, Breart G, Diez-Perez A, Felsenberg D, et al.
Adverse reactions and drug–drug interactions in the management of women
with postmenopausal osteoporosis. Calcif Tiss Int 2011;89:91–104.
[40] Pazianas M, Abrahamsen B, Eiken PA, Eastell R, Russell RG. Reduced colon
cancer incidence and mortality in postmenopausal women treated with an
oral bisphosphonate—Danish National Register Based Cohort Study.
Osteoporos Int 2012;23:2693–701.
[41] Lyles KW, Colon-Emeric CS, Magaziner JS, Adachi JD, Pieper CF, Mautalen C,
et al. Zoledronic acid and clinical fractures and mortality after hip fracture. N
Eng J Med 2007;357:1799–809.
[42] Kwong FN, Harris MB. Recent developments in the biology of fracture repair. J
Am Acad Orthop Surg 2008;16:619–25.
[43] Diaz-Garcia RJ, Oda T, Shauver MJ, Chung KC. A systematic review of
outcomes and complications of treating unstable distal radius fractures in the
elderly. J Hand Surg Am 2011 May;36(5):824–35.
[44] Papanastassiou ID, Phillips FM, Van Meirhaeghe J, Berenson JR, Andersson GB,
Chung G, et al. Comparing effects of kyphoplasty, vertebroplasty, and non-
surgical management in a systematic review of randomized and non-
randomized controlled studies. Eur Spine J 2012;21:1826–43.
[45] Edidin AA, Ong KL, Lau E, Kurtz SM. Mortality risk for operated and
nonoperated vertebral fracture patients in the Medicare population. J Bone
Miner Res 2011;26:1617–26.
[46] Gao H, Liu Z, Xing D, Gong M. Which is the best alternative for displaced
femoral neck fractures in the elderly? A meta-analysis. Clin Orthop Relat Res
2012;470:1782–91.
[47] Keating JF, Grant A, Masson M, Scott NW, Forbes JF. Randomized comparison
of reduction and fixation, bipolar hemiarthroplasty, and total hip
arthroplasty. Treatment of displaced intracapsular hip fractures in healthy
older patients. J Bone Joint Surg Am 2006;88:249–60.
[48] Morshed S, Bozic KJ, Ries MD, Malchau H, Colford Jr JM. Comparison of
cemented and uncemented fixation in total hip replacement: a meta-analysis.
Acta Orthop 2007;78:315–26.
[49] Yamada H, Yoshihara Y, Henmi O, Morita M, Shiromoto Y, Kawano T, et al.
Cementless total hip replacement: past, present, and future. J Orthop Sci
2009;14:228–41.
[50] Parker MJ, Gurusamy KS, Azegami S. Arthroplasties (with and without bone
cement) for proximal femoral fractures in adults. Cochrane Database Syst
Rev. 2010;16:CD001706.
[51] Abdulkarim A, Ellanti P, Motterlini N, Fahey T, O’Byrne JM. Cemented versus
uncemented fixation in total hip replacement: a systematic review and meta-
analysis of randomized controlled trials. Orthop Rev (Pavia) 2013;5:e8.
[52] Lorich DG, Geller DS, Nielson JH. Osteoporotic pertrochanteric hip fractures.
Management and current controversies. J Bone Joint Surg Am
2004;86:398–410.
[53] Fini M, Giavaresi G, Torricelli P, Krajewski A, Ravaglioli A, Mattioli Belmonte
M, et al. Biocompatibility and osteointergration in osteoporotic bone. J Bone
Joint Surg Br 2001;83:139–43.
[54] Heini PF, Berlemann U, Kaufman M, Lippuner K, Fankhauser C, Van Landuyt P.
Augmentation of mechanical properties in osteoporotic vertebral bones – a
biomechanical investigation of vertebroplasty efficacy with different bone
cements. Eur Spine J 2001;10:164–71.
[55] Yu L, Li Y, Zhao K, Tang Y, Cheng Z, Chen J, et al. A novel injectable calcium
phosphate cement-bioactive glass composite for boneregeneration. PLOS One
2013. http://dx.doi.org/10.1371/journal.pone.0062570.
[56] Hoekstra JWM, van den Beucken JJJP, Leeuwenburgh SCG, Bronkhorst EM,
Meijer GJ, Jansen JA. Tantalum oxide and barium sulfate as radiopacifiers in
injectable calcium phosphate-poly(lactic-co-glycolic acid) cements for
monitoring in vivo degradation. J Biomed Mater Res 2014;A102:141–9.
[57] Chen W, Zhou H, Weir MD, Tang M, Bao C, Xu HHK. Human embryonic stem
cell-derived mesenchymal stem cell seeding on calcium phosphate cement-
chitosan-RGD scaffold for bone repair. Tiss Eng A 2013;19:915–27.
[58] Vorndran E, Geffers M, Ewald A, Lemm M, Nies B, Gbureck U. Ready-to-use
injectable calcium phosphate bone cement paste as drug carrier. Acta
Biomater 2013;9:9558–67.
[59] Franchi M, Fini M, Giavaresi G, Ottani V. Peri-implant osteogenesis in health
and osteoporosis. Micron 2005;36:630–44.
[60] Fini M, Giavaresis G, Torricelli P, Borsari V, Giardino R, Nicolini A, et al.
Osteoporosis and biomaterial osteointegration. Biomed Pharmacother
2004;58:487–93.
[61] Verron E, Gauthier O, Janvier P, Pilet P, Lesouer J, Bujoli B, et al. In vivo bone
augmentation in an osteoporotic environment using bisphosphonate-loaded
calcium deficient apatite. Biomaterials 2010;31:7776–84.
[62] Manzano M, Lozano D, Arcos D, Portal-Nuñez S, López C, Esbrit P, et al.
Comparison of the osteoblastic activity conferred to si-doped hydroxyapatite
scaffolds by different osteostatin coating. Acta Biomater 2011;7:3555–62.
[63] Arcos D, Vallet-Regi M. Bioceramics for drug delivery. Acta Mater
2013;61:890–911.
[64] Verron E, Bouler JM, Guicheux J. Controlling the biological function of calcium
phosphate bone substitutes with drugs. Acta Biomater 2012;8:3541–51.
[65] Trejo CG, Lozano D, Manzano M, Doadrio JC, Salinas AJ, Dapia S, et al. The
osteoinductive properties of mesoporous silicate coated with osteostatin in a
rabbit femur cavity defect model. Biomaterials 2010;31:8564–73.
[66] Manzano M, Vallet-Regí M. Revisiting bioceramics: bone regenerative and
local drug delivery systems. Prog. Solid State Ch 2012;40:17–30.
[67] Chapman JR, Harrington RM, Lee KM, Anderson PA, Tencer AF, Kowalski D.
Factors affecting the pullout strength of cancellous bone screws. J Biomech
Eng 1996;118:391–8.
[68] Frigg R, Frenk A, Wagner M. Biomechanics of plate osteosynthesis. Tech
Orthop 2007;22:203–8.
[69] McKoy BE, An YH. An injectable cementing screw for fixation in osteoporotic
bone. J Biomed Mater Res 2000;53:216–20.
[70] Hu MH, Wu HT, Chang MC, Yu WK, Wang ST, Liu CL. Polymethylmethacrylate
augmentation of the pedicle screw: the cement distribution in the vertebral
body. Eur Spine J 2011;20:1281–8.
[71] Choma TJ, Pfeiffer FM, Swope RW, Hirner JP. Pedicle screw design and cement
augmentation in osteoporotic vertebrae: effects of fenestrations and cement
viscosity on fixation and extraction. Spine (Phila Pa 1976)
2012;37:E1628–32.
D. Arcos et al. / Acta Biomaterialia xxx (2014) xxx–xxx 11
Please cite this article in press as: Arcos D et al. The relevance of biomaterials to the prevention and treatment of osteoporosis. Acta Biomater (2014),
http://dx.doi.org/10.1016/j.actbio.2014.01.004
[72] Gittens RA, Olivares-Navarrete R, Cheng A, Anderson DM, McLachlan T,
Stephan I, et al. The roles of titanium surface micro/nanotopography and
wettability on the differential response of human osteoblast lineage cells.
Acta Biomater 2013;9:6268–77.
[73] Buser D, Schenk RK, Steinemann S, Fiorellini JP, Fox CH, Stich H. Influence of
surface characteristics on bone integration of titanium implants – a
histomorphometric study in miniature pigs. J Biomed Mater Res
1991;25:889–902.
[74] Cocharan DL, Jackson JM, Bernard JP, ten Bruggenkate CM, Buser D, Taylor TD,
et al. A 5-year prospective multicenter study of early loaded titanium
implants with a sandblasted and acid-etched surface. Int Oral Maxillofac
Implants 2011;26:1324–32.
[75] Orsini G, Piattelli M, Scarano A, Petrone G, Kenealy J, Piattelli A, et al.
Randomized, controlled histologic and histomorphometric evaluation of
implants with nanometer-scale calcium phosphate added to the dual acid-
etched surace in the human posterior maxilla. J Periodontol 2007;78:209–18.
[76] Collaert B, Wijnen L, De Bruyn H. A 2-year prospective study on immediate
loading with fluoride-modified implants in the edentulous mandible. Clin
Oral Implants Res 2011;22:1111–6.
[77] Park JH, Wasilewski CE, Almodovar N, Olivares-Navarrete R, Boyan BD,
Tannenbaum R, et al. The responses to surface wettability gradientes induced
by chitosan nanofilms on microtextured titanium mediated by specific
integrin receptors. Biomaterials 2012;33:7386–93.
[78] Bornstein MM, Wittneben JG, Bragger U, Buser D. Early loading at 21 days of
non-submerged titanium implants with a chemically modified sandblasted
and acid-etched surface. 3-year results of a prospective study in the posterior
mandible. J Periodontol 2010;81:809–18.
[79] Nguyen HQ, Deporter DA, Pilliar RM, Valiquette N, Yakubovich R. The effect of
sol–gel-formed calcium phosphate coatings on bone in growth and
osteoconductivity of porous-surfaced Ti alloy implants. Biomaterials
2004;25:865–76.
[80] Yang Y, Kim KH, Ong JL. A review on calcium phosphate coatings produced
using a sputtering process—an alternative to plasma spraying. Biomaterials
2005;26:327–37.
[81] Surmenev RA. A review of plasma-assisted methods for calcium phosphate-
based coatings fabrication. Surf Coat Technol 2012;206:2035–56.
[82] Heimann RB. Thermal spraying of biomaterials. Surface Coatings Technol
2006;201:2012–9.
[83] Surmenev RA, Surmeneva MA, Ivanova AA. Significance of calcium phosphate
coatings for the enhancement of new bone osteogenesis – A review. Acta
Biomater 2014;10:557–79.
[84] Bright DS, Clark HG, McCollum DE. Serum analysis and toxic effects of
methylmethacrylate. Surg Forum 1972;23:455–7.
[85] Leeson MC, Lippitt SB. Thermal aspects of the use of polymethylmethacrylate
in large metaphyseal defects in bone: A clinical review and laboratory study.
Clin Orthop Relat Res 1993:239–45.
[86] Topoleski LDT, Ducheyne P, Cuckler JM. A fractographic analysis of in vivo
poly(methyl methalcrylate) bone cement failure mechanisms. J Biomed
Mater Res 1990;24:135–54.
[87] Berlemann U, Ferguson SJ, Nolte LP, Hein PF. Adjacent vertebral failure after
vertebroplasty—a biomechanical investigation. J Bone Joint Surg Br
2002;84B:748–52.
[88] Lewis G. Injectable bone cements for use in vertebroplasty and kyphoplasty:
state-of-the-art review. J Biomed Mater Res B Appl Biomater
2006;76:456–68.
[89] Urlings TAJ, Van Der Linden E. Elastoplasty: first experience in 12 patients.
Cardiovasc Intervent Radiol 2013;36:479–83.
[90] Blattert TR, Jestaedt L, Weckbach A. Suitability of a calcium phosphate cement
in osteoporotic vertebral body fracture augmentation: a controlled,
randomized, clinical trial of balloon kyphoplasty comparing calcium
phosphate versus polymethylmethacrylate. Spine 2009;34:108–14.
[91] Piazzolla A, De Giorgi G, Solarino G. Vertebral body recollapse without trauma
after kyphoplasty with calcium phosphate cement. Musculoskelet Surg
2011;95:141–5.
[92] Boszczyk B. Prospective study of standalone balloon kyphoplasty with
calcium phosphate cement augmentation in traumatic fractures (G.
Maestretti et al.). Eur Spine J 2007;16:611.
[93] Maestretti G, Cremer C, Otten P, Jakob RP. Prospective study of standalone
balloon kyphoplasty with calcium phosphate cement augmentation in
traumatic fractures. Eur Spine J 2007;16:601–10.
[94] Bernards CM, Chapman JR, Mirza SK. Lethality of embolizednorian bone
cement varies with the time between mixing and embolization. In: 50th
Annual Meeting of the Orthopaedic Research Society, San Fransisco; 2004. p.
0254.
[95] Krebs J, Aebli N, Goss BG, Sugiyama S, Bardyn T, Boecken I, et al.
Cardiovascular changes after pulmonary embolism from injecting calcium
phosphate cement. J Biomed Mater Res B Appl Biomater 2007;82:526–32.
[96] Dressel S, Jarvers JSG, Josten C, Blattert TR. Percutaneous balloon kyphoplasty
in osteoporotic vertebral body fracture with a calcium aluminate ceramic
(XeraspineÓ). Eur Musculoskelet Rev 2012;7:209–12.
[97] Rauschmann M, Vogl T, Verheyden A, Pflugmacher R, Werba T, Schmidt S,
et al. Bioceramic vertebral augmentation with a calcium sulphate/
hydroxyapatite composite (Cerament™ SpineSupport) in vertebral
compression fractures due to osteoporosis. Eur Spine J 2010;19:887–92.
[98] Masala S, Nano G, Marcia S, Muto M, Fucci FPM, Simonetti G. Osteoporotic
vertebral compression fractures augmentation by injectable partly resorbable
ceramic bone substitute (Cerament™|SPINE SUPPORT): A prospective
nonrandomized study. Neuroradiology 2012;54:589–96.
[99] Bai B, Kummer FJ, Spivak J. Augmentation of anterior vertebral body screw
fixation by an injectable, biodegradable calcium phosphate bone substitute.
Spine 2001;26:2679–83.
[100] Bohner M, Lemaitre J, Cordey J, Gogolewski S, Ring TA, Perren SM. Potential
use of biodegradable bone cement in bone surgery: holding strength of
screws in reinforced osteoporotic bone. Orthop Trans 1992;16:401–2.
[101] Mermelstein LE, Chow LC, Friedman C. Crisco Iii JJ. The reinforcement of
cancellous bone screws with calcium phosphate cement. J Orthop Trauma
1996;10:15–20.
[102] Stankewich CJ, Swiontkowski MF, Tencer AF, Yetkinler DN, Poser RD.
Augmentation of femoral neck fracture fixation with an injectable calcium-
phosphate bone mineral cement. J Orthop Res 1996;14:786–93.
[103] Larsson S, Stadelmann VA, Arnoldi J, Behrens M, Hess B, Procter P, et al.
Injectable calcium phosphate cement for augmentation around cancellous
bone screws. In vivo biomechanical studies. J Biomech 2012;45:1156–60.
[104] Stadelmann VA, Bretton E, Terrier A, Procter P, Pioletti DP. Calcium phosphate
cement augmentation of cancellous bone screws can compensate for the
absence of cortical fixation. J Biomech 2010;43:2869–74.
[105] Vallet-Regí M, Colilla M, Izquierdo-Barba I. Bioactive mesoporoussilicas as
controlled delivery systems: Application in bonetissue regeneration. J
Biomed Nanotechnol 2008;4:1–15.
[106] Hutmacher DW. Scaffolds in tissue engineering bone and cartilage.
Biomaterials 2000:2529–43.
[107] Stevens MM, George J. Exploring and engineering the cell surface interface.
Science 2005;310:1135–8.
[108] Schieker M, Seitz H, Drosse I, Seitz S, Mutschler W. Biomaterials as scaffolds
for bone tissue engineering. Eur J Trauma 2006;32:114–24.
[109] Kruyt MC, de Bruijn JD, Wilson CE, et al. Viable osteogenic cells are obligatory
for tissue-engineered ectopic bone formation in goats. Tissue Eng
2003;9:327–36.
[110] Dorozhkin SV, Epple M. Biological and medical significance of calcium
phosphates. Angew Chem Int Ed 2002;41:3130–46.
[111] Vallet-Regí M, González-Calbet JM. Calcium phosphates as substitution of
bone tissues. Progr Solid State Chem 2004;32:1–31.
[112] Bohner M. Resorbable biomaterials as bone graft substitutes. Mater Today
2010;13:24–30.
[113] Weiner S, Wagner HD. The material bone: structure–mechanical function
relations. Annu Rev Mater Sci 1998;28:271–9.
[114] Tadic D, Epple M. A thorough physicochemical characterisation of 14 calcium
phosphate-based bone substitution materials in comparison to natural bone.
Biomaterials 2004;25:987–94.
[115] Weiss P, Obadia L, Magne D, Bourges X, Rau C, Weitkamp T, et al. Synchrotron
X-ray microtomography (on a micron scale) provides three-dimensional
imaging representation of bone ingrowth in calcium phosphate biomaterials.
Biomaterials 2003;24:4591–601.
[116] Schilling AF, Linhart W, Filke S, Gebauer M, Schinke T, Rueger JM, et al.
Resorbability of bone substitute biomaterials by human osteoclasts.
Biomaterials 2004;25:3963–72.
[117] Detsch R, Hagmeyer D, Neumann M, Schaefer S, Vortkamp A, Wuelling M,
et al. The resorption of nanocrystalline calcium phosphates by osteoclast-like
cells. Acta Biomater 2010;6:3223–33.
[118] Xynos ID, Hukkanen MVJ, Batten JJ, Buttery LD, Hench LL, Polak JM. Bioglass
(R) 45S5 stimulates osteoblast turnover and enhances bone formation
in vitro: implications and applications for bone tissue engineering. Calcif
Tiss Int 2000;67:321–9.
[119] Han P, Wu C, Xiao Y. The effect of silicate ions on proliferation, osteogenic
differentiation and cell signaling pathways (WNT and SHH) of bone marrow
stromal cells. Biomater Sci 2013;1:379–92.
[120] Aguirre A, González A, Planell JA, Engel E. Extracellular calcium modulates
in vitro bone marrow-derived Flk-1+ CD34+ progenitor cell chemotaxis and
differentiation through a calcium-sensing receptor. Biochem Biophys Res
Commun 2010;393:156–61.
[121] Hench LL. Genetic design of bioactive glass. J. Eur Ceram Soc
2009;29:1257–65.
[122] Rahaman MN, Day DE, Bal BS, Fu Q, Jung SB, Bonewald LF, et al. Bioactive
glass in tissue engineering. Acta Biomater 2011;7:2355–73.
[123] Xynos ID, Edgar AJ, Buttery LDK, Hench LL, Polak JM. Gene-expression
profiling of human osteoblasts following treatment with the ionic products of
Bioglass
Ò
45S5 dissolution. J Biomed Mater Res 2001;55:151–7.
[124] Gorustovich AA, Roether JA, Boccaccini AR. Effect of bioactive glasses on
angiogenesis: in-vitro and in-vivo evidence. A review. Tiss Eng B
2010;16:199–207.
[125] Navarro M, Ginebra MP, Planell JA. Cellular response to calcium
phosphate glasses with controlled solubility. J Biomed Mater Res
2003;67A:1009–15.
[126] Charles-Harris M, Koch M, Navarro M, Lacroix D, Engel E, Planell JA. A PLA/
calcium phosphate degradable composite material for bone tissue
engineering: an in vitro study. J Mater Sci Mater Med 2008;19:1503–13.
[127] Navarro M, Sanzana ES, Planell JA, Ginebra MP, Torres PA. In vivo behavior of
calcium phosphate glasses with controlled solubility. Key Eng Mater
2005;284–286:893–6.
[128] Gentleman E, Fredholm YC, Jell G, Lotfibakhshaiesh N, O’Donnell MD, Hill RG,
et al. The effects of strontium-substituted bioactive glasses on osteoblasts
and osteoclasts in vitro. Biomaterials 2010;31:3949–56.
12 D. Arcos et al. / Acta Biomaterialia xxx (2014) xxx–xxx
Please cite this article in press as: Arcos D et al. The relevance of biomaterials to the prevention and treatment of osteoporosis. Acta Biomater (2014),
http://dx.doi.org/10.1016/j.actbio.2014.01.004
[129] Teofilo JM, Brentegani LG, Lamano-Carvalho TL. Bone healing in osteoporotic
female rats following intra-alveolar grafting of bioactive glass. Arch Oral Biol
2004;49:755–62.
[130] Vallet-Regí M, Balas F, Arcos D. Mesoporous materials for drug delivery.
Angew Chem Int Ed 2007;46:7548–58.
[131] Schliephake H, Weich HA, Dullin C, Gruber R, Frahse S. Mandibular bone
repair by implantation of rhBMP-2 in a slow release carrier or polylactic
acid—an experimental study in rats. Biomaterials 2008;29:103–10.
[132] Devescovi V, Leonardi E, Ciapetti G, Cenni E. Growth factors in bone repair.
Chir Organi Mov 2008;92:161–8.
[133] Lissenberg-Thunnissen SN, de Gorter DJJ, Sier CFM, Schipper IB. Use and
efficacy of bone morphogenetic proteins in fracture healing. Int Orthop
2011;35:1271–80.
[134] Wernike E, Montjovent MO, Liu Y, Wismeijer D, Hunziker EB, Siebenrock KA,
et al. VEGF incorporated into calcium phosphate ceramics promotes
vascularisation and bone formation in vivo. Eur Cells Mater 2010;19:30–40.
[135] Lode A, Wolf-Brandstetter C, Reinstorf A, Bernhardt A, Koenig U, Pompe W,
et al. Calcium phosphate bone cements, functionalized with VEGF: release
kinetics and biological activity. J Biomed Mater Res 2007;81A:474–83.
[136] Evans CH. Gene therapy for bone healing. Exp Rev Mol Med 2010;23(12):e18.
[137] Bushman FD. Retroviral integration and human gene therapy. J Clin Invest
2007;117:2083–6.
[138] Yi Y, Hahm SH, Lee KH. Retroviral gene therapy: safety issues and possible
solutions. Curr Gene Ther 2005;5:25–35.
[139] Sokolova V, Epple M. Inorganic nanoparticles as carriers of nucleic acids into
cells. Angew Chem Int Ed 2008;47:1382–95.
[140] Guo X, Huang L. Recent advances in nonviral vectors for gene delivery. Acc
Chem Res 2012;45:971–9.
[141] Wegman F, Bijenhof A, Schuijff L, Oner FC, Dhert WJA, Alblas J. Osteogenic
differentiation as a result of BMP-2 plasmid DNA based gene therapy in vitro
and in vivo. Eur Cell Mater 2011;21:230–42.
[142] Zhang C, Wang KZ, Qiang H, Tang YL, Li QA, Li MA, et al. Angiopoiesis and
bone regeneration via co-expression of the hVEGF and hBMP genes from an
adeno-associated viral vector in vitro and in vivo. Acta Pharmacol Sin
2010;31:821–30.
[143] Krebs MD, Salter E, Chen E, Sutter KA, Alsberg E. Calcium phosphate-DNA
nanoparticle gene delivery from alginate hydrogels induces in vivo
osteogenesis. J Biomed Mater Res A 2010;92A:1131–8.
[144] Keeney M, van den Beucken JJJP, van der Kraan PM, Jansen JA, Pandit A. The
ability of a collagen/calcium phosphate scaffold to act as its own vector for
gene delivery and to promote bone formation via transfection with VEGF165.
Biomaterials 2010;31:2893–902.
[145] Cheang TY, Wang SM, Hu ZJ, Xing ZH, Chang GQ, Yao C, et al. Calcium
carbonate/CaIP6 nanocomposite particles as gene delivery vehicles for
human vascular smooth muscle cells. J Mater Chem 2010;20:8050–5.
[146] Chernousova S, Klesing J, Soklakova N, Epple M. A genetically active nano-
calcium phosphate paste for bone substitution, encoding the formation of
BMP-7 and VEGF-A. RSC Adv 2013;3:11155–61.
[147] Liu H, Webster TJ. Mechanical properties of dispersed ceramic nanoparticles
in polymer composites for orthopedic applications. Int J Nanomed
2010;5:299–313.
[148] Dunlop JWC, Fratzl P. Biological composites. Ann Rev Mater Res
2010;40:1–24.
[149] Weiner S, Wagner HD. The material bone: structure-mechanical function
relations. Annu Rev Mater Sci 1998;28:271–98.
[150] Nikolov S, Petrov M, Lymperakis L, Friak M, Sachs C, Fabritius HO, et al.
Revealing the design principles of high-performance biological composites
using ab initio and multiscale simulations: The example of lobster cuticle.
Adv Mater 2010;22:519.
[151] Kurreck J. RNA Interference: from basic research to therapeutic applications.
Angew Chem Int Ed 2009;48:1378–98.
[152] Kesharwani P, Gajbhiye V, Jain NK. A review of nanocarriers for the delivery
of small interfering RNA. Biomaterials 2012;33:7138–50.
[153] Reischl D, Zimmer A. Drug delivery of siRNA therapeutics: potentials and
limits of nanosystems. Nanomedicine 2009;5:8–20.
[154] Ghildiyal M, Zamore PD. Small silencing RNAs: an expanding universe. Nat
Rev Genet 2009;10(2):94–108.
[155] Laroui H, Theiss AL, Yan Y, Dalmasso G, Nguyen HTT, Sitaraman SV, et al.
Functional TNF-a gene silencing mediated by polyethyleneimine/TNF-a
siRNAnanocomplexes in inflamed colon. Biomaterials 2011;32:1218–28.
[156] Zhang X, Kovtun A, Mendoza-Palomares C, Oulad-Abdelghani M, Facca S,
Fioretti F, et al. SiRNA-loaded multi-shell nanoparticles incorporated into a
multilayered film as a reservoir for gene silencing. Biomaterials
2010;31:6013–8.
[157] NIH Consensus Development Panel on Osteoporosis Prevention, Diagnosis,
and Therapy. Osteoporosis prevention, diagnosis, and therapy. JAMA
2001;14(285):785–95.
[158] Arvidson K, Abdallah BM, Applegate LA, Nicola Baldini N, Cenni E, Gomez-
Barrena E, et al. Bone regeneration and stem cells. J Cell Mol Med
2011;15:718–46.
[159] Gómez-Barrena E, Rosset P, Muller I, Giordano R, Bunu C, Laroylle P, et al.
Bone regeneration: stem cell therapies and clinical studies in orthopaedics
and traumatology. J Cell Mol Med 2011;15:1266–86.
[160] Hernigou P, Poignard A, Beaujean F, Rouard H. Percutaneous autologous
bone-marrow grafting for nonunions. Influence of the number and
concentration of progenitor cells. J Bone Joint Surg Am 2005;87:1430–7.
[161] Jakob F, Ebert R, Ignatius A, Matsushita T, Watanabe Y, Groll J, et al. Bone
tissue engineering in osteoporosis. Maturitas 2013;75:118–24.
[162] Bohner M, Lemaitre J. Can bioactivity be tested in vitro with SBF solution?
Biomaterials 2009;30:2175–9.
[163] Pan H, Zhao X, Darvell BW, Lu WW. Apatite-formation ability – predictor of
‘‘bioactivity’’? Acta Biomater 2010;6:4181–8.
[164] Kallmes DF, Comstock BA, Heagerty PJ, Turner JA, Wilson DJ, Diamond TH,
et al. A randomized controlled trial of vertebroplasty for osteoporotic spine
fractures. N Engl J Med 2009;361:69–579.
[165] Buchbinder R, Osborne RH, Ebeling PR, Wark JD, Mitchell P, Wriedt C, et al. A
randomized trial of vertebroplasty for painful osteoporotic vertebral
fractures. N Engl J Med 2009;361:557–68.
[166] http://en.wikipedia.org/wiki/Contaminated_haemophilia_blood_products.
D. Arcos et al. / Acta Biomaterialia xxx (2014) xxx–xxx 13
Please cite this article in press as: Arcos D et al. The relevance of biomaterials to the prevention and treatment of osteoporosis. Acta Biomater (2014),
http://dx.doi.org/10.1016/j.actbio.2014.01.004
... Regrettably, there are significant limitations to existing osteoporosis therapies, including inadequacies and long-term safety concerns [3] . There is currently no adequate treatment for osteoporosis-related bone thinning [4] . The only effective treatments for osteoporosis are antiresorptive medications, which slow down excessive bone resorption (the main cause of primary osteoporosis), and anabolic agents, which successfully rebuild the lost bone mass caused by excessive resorption [5] . ...
... to stimulate osteoblasts, make up the majority of therapeutic approaches to prevent fracture and restrict bone loss [11] . Drugs used to treat anti-resorptive disorders primarily work by decreasing osteoclast activity, maintaining bone mass, and boosting bone strength [4] . On the other hand, anabolic medicines have the ability to stimulate the development of new bone and can stop the bone degeneration brought on by the progression of osteoporosis [12] . ...
Article
Full-text available
Osteoporosis, characterized by low bone density and increased risk of fractures, represents a major healthcare challenge. Antiresorptive and anabolic medications are now used to treat osteoporosis in an effort to reduce bone loss and increase bone mass. Innovative methods are required since current therapies have drawbacks. Promising options for improving bone health and medicine delivery are provided by nanotechnology. Bisphosphonates with tetracyclines and oligopeptides, among other compounds that target the bone, make it easier to provide a particular medication to bone tissue. Additionally, nanocarriers are essential for the administration of both organic and inorganic nanoparticles in the treatment of osteoporosis. Drug encapsulation and controlled release may be done in a variety of ways using organic nanoparticles. Inorganic nanoparticles have special qualities that help in medication transport and bone repair. This review explores the potential of nanoparticle-based strategies in the treatment of osteoporosis.
... Biopolymers and polyesters, on the other hand, decompose into more easily digestible compounds like sugar or lactic acid. Mesoporous silica nanoparticles are another kind of multifunctional porous nanoparticle used for medication delivery [58,59]. ...
Article
Full-text available
Calcium phosphate (CaP) is a kind of eco-friendly biodegradable material chemically similar to human hard tissue like bone and teeth and is highly biocompatible. CaPs have excellent biological quality, are cheap and easy to produce, are safe, and may be approved for clinical applications quite fast. CaP materials have proven themselves, yet their future is still bright. Today’s major global public health issue is the prevalence of injury and illness brought on by bone fractures. There are a number of problems with the current treatments that call for research into better methods of dealing with bone disorders. Gene therapy has recently gained attention as a promising strategy for efficient bone repair and regeneration via the use of RNA interference (RNAi) systems to modulate gene expression in the bone microenvironment. CaP nanoparticles have been shown to be efficient transporters for biomolecules that cannot enter cells to exert their biological impacts, such as nucleic acids, proteins, peptides, antibodies, and medicines. Surface functionalization and incorporation of cargo molecules make these nanoparticles distinct from their solid counterparts. This protects against nucleases and allows for selective cellular targeting. In this overview, we looked at the fundamentals of nanoparticles of calcium phosphates, including their production techniques, physicochemical features, and applications in transfection, gene silencing, drug administration, the environment, electricity, tissue engineering, drug delivery, and biomedicine. This study is predicted to be extremely valuable and advantageous to the researcher in the future progress of the research work in therapeutic applications since it comprises very comprehensive thoughts about calcium phosphate nanoparticles. Graphical Abstract Calcium phosphate is known to be a “super material” with improved biological qualities such as bioactivity, biodegradability, cellular function, and osteoconduction. The synthesis and fabrication of calcium phosphate by different crucial techniques in enhancing the physical and chemical properties allow it to be potential in various bioengineering fields such as drug delivery, bone regeneration, gene silencing, and electrical as well as environmental applications.
... For all prepared hybrid systems, as well as for ColChCS mod 30_20, the wettability studies were performed by the means of contact angle measurements ( Figure 3C). The hydrophilicity of materials prepared for bone tissue engineering influences their applicability and ability to successfully adhere cells, as well as their proliferation [45,46]. For example, the increased wettability in the Ti surface allowed for improved osseointegration in the presence of osteoporosis [47], and enhanced bone formation by regulating angiogenesis, bone mineralization, and bone remodeling [48,49]. ...
Article
Full-text available
Alendronate (ALN) is the most commonly prescribed oral nitrogen-containing bisphosphonate for osteoporosis therapy. However, its administration is associated with serious side effects. Therefore, the drug delivery systems (DDS) enabling local administration and localized action of that drug are still of great importance. Herein, a novel multifunctional DDS system based on the hydroxyapatite-decorated mesoporous silica particles (MSP-NH2-HAp-ALN) embedded into collagen/chitosan/chondroitin sulfate hydrogel for simultaneous osteoporosis treatment and bone regeneration is proposed. In such a system, the hydrogel serves as a carrier for the controlled delivery of ALN at the site of implantation, thus limiting potential adverse effects. The involvement of MSP-NH2-HAp-ALN in the crosslinking process was established, as well as the ability of hybrids to be used as injectable systems. We have shown that the attachment of MSP-NH2-HAp-ALN to the polymeric matrix provides a prolonged ALN release (up to 20 days) and minimizes the initial burst effect. It was revealed that obtained composites are effective osteoconductive materials capable of supporting the osteoblast-like cell (MG-63) functions and inhibiting osteoclast-like cell (J7741.A) proliferation in vitro. The purposely selected biomimetic composition of these materials (biopolymer hydrogel enriched with the mineral phase) allows their biointegration (in vitro study in the simulated body fluid) and delivers the desired physicochemical features (mechanical, wettability, swellability). Furthermore, the antibacterial activity of the composites in in vitro experiments was also demonstrated.
... Several treatments for damaged bone tissue, including autografts, allografts, alloplastic materials, and implants have been developed (Li et al., 2017;Zhang et al., 2018). However, due to low bioactivity and failure to support bone formation in the host tissue, peripheral infection, and immunologic response after the surgery many implants will fail (Campoccia et al., 2006;Tomsia et al., 2011;Arcos et al., 2014). To overcome these drawbacks, tissue-engineered implants have been proposed, which accelerate the healing process by providing an environment like extracellular matrix (ECM) for the cells to adhere, differentiate, propagate, and produce neo-tissue. ...
Article
Fabricating a multifunctional orthopedic implant which prevents post-surgery infection is highly desirable in advanced materials applications. However, designing an antimicrobial implant, which simultaneously promotes a sustained drug release and satisfactory cell proliferation, remains a challenge. The current study presents a drug-loaded surface-modified titanium nanotube (TNT) implant with different surface chemistry which was developed to investigate the effect of surface coating on drug release, antimicrobial activity, and cell proliferation. Accordingly, sodium alginate and chitosan were coated on the surface of TNT implants with different coating orders through layer-by-layer assembly. The coatings' swelling ratio and degradation rate were around and , respectively. The drug release results showed that surface-coatings prolonged the releasing profile for about 4 weeks. Chitosan coated TNTs demonstrated greater inhibition zone at compared with the other samples where no inhibition zone was observed. However, chitosan and alginate coated TNTs exhibited smaller inhibition zones at and , respectively, compared to bare TNT, which can be attributed to the coatings preventing the antibiotic burst release. Higher viability of cultured osteoblast cells was observed for chitosan-coated TNT as the top layer compared to the bare TNT at , indicating improved bioactivity of TNT implants when the chitosan has the most contact with cells. Coupled with the cell viability assay, molecular dynamics (MD) simulations were carried out by placing collagen and fibronectin near the considered substrates. In agreement with cell viability results, MD simulations also indicated that chitosan had the highest adsorption energy approximately . In summary, the proposed bilayer chitosan-coated drug-loaded TNT implant with chitosan and sodium alginate coating as the top and the bottom layers, respectively, can be a potential candidate for orthopedic applications in the light of its bacterial biofilm prevention, better osteoconductivity, and providing an adequate drug release profile.
... Bone is an inorganic-bioorganic composite material consisting mainly of collagen and calcium phosphate [1,2]. Calcium phosphate nanoparticles that occur as hydroxyapatite constitute the inorganic basis of this biomineralized hard tissue, i.e., mammalian bone [3,4]. As they are inherently biocompatible and biodegradable [5], they have been extensively applied in the therapeutic delivery of drugs and biomolecules [1,[6][7][8][9]. ...
Article
Full-text available
Calcium phosphate nanoparticles are highly biocompatible and biodegradable in bone regeneration. On the other hand, strontium and magnesium enhance the formation of bone. The substitution of calcium by strontium and magnesium is an efficient way to improve the biological properties of calcium phosphate-based biomaterials. Strontium-doped calcium phosphate nanoparticles and magnesium-doped calcium phosphate nanoparticles with degrees of cation substitution of 5, 10, 15, and 20 mol% with respect to calcium were prepared by precipitation, followed by surface functionalization with polyethyleneimine (PEI, cationic) or carboxymethylcellulose (CMC, anionic). The nanoparticles were characterized by dynamic light scattering (DLS), zeta potential measurement, scanning electron microscopy (SEM), atomic absorption spectrometry (AAS), energy dispersive X-ray analysis (EDX), and X-ray powder diffraction (XRD). The particles were approximately spherical (diameter 40–70 nm). The addition of magnesium and strontium considerably decreased the internal crystallinity, i.e., the doped particles were almost X-ray amorphous. The cell-biological effects were assessed on three different cell lines, i.e., HeLa cells, MG63 cells, and MC3T3 cells. Cell viability tests (MTT) showed a low cytotoxicity, the alkaline phosphatase (ALP) activity was strongly increased, and the nanoparticles were taken up well by the three cell lines.
Preprint
Full-text available
Due to bioactive properties, introducing spongin-like collagen (SPG) into the biosilica (BS) extracted from marine sponges, would present an enhanced biological material for improving osteoporotic fracture healing by increasing bone formation rate. Our aim was to characterize the morphology of the BS/SPG scaffolds by scanning electron microscopy (SEM), the chemical bonds of the material by Fourier transform infrared spectroscopy (FTIR) and evaluate the orthotopic in vivo response of BS/SPG scaffolds in tibial defects of osteoporotic fractures in rats (histology, histomorphometry and immunohistochemistry) in 2 experimental periods (15 and 30 days). SEM showed that scaffolds were porous, showing the spicules of BS and fibrous aspect of SPG. FTIR showed characteristic peaks of BS and SPG. For the in vivo studies, after 30 days BS and BS/SPG showed a higher amount of newly formed bone compared to the first experimental period, observed both in the periphery and in the central region of the bone defect. For histomorphometry, BS/SPG presented higher %BV/TV compared to the other experimental groups. After 15 days, BS presented higher volumes of collagen type I. After 30 days, all groups demonstrated higher volumes of collagen type III compared to volumes at 15 days. After 30 days, BS/SPG presented higher immunostaining of osteoprotegerin compared to the other experimental groups at the same experimental period. The results showed that BS and BS/SPG scaffolds were able of improving bone healing. Future researches should focus on the effects of BS/SPG on longer periods in vivo studies.
Article
Full-text available
A major worldwide health problem is osteoporosis, particularly in older people. There are restrictions on current therapies, such as anti-resorptive medications. Promising techniques including stem cell treatment, gene therapy, and growth factors are available in regenerative medicine. Preclinical and clinical research indicate promise for mesenchymal stem cells derived from bone marrow, adipose tissue, and umbilical cord blood. Enhancing bone regeneration with gene therapy that targets osteogenic factors such as BMPs is a promising approach. In order to mend bones, growth factors, cytokines, and scaffold materials are essential. Realising the benefits of regenerative medicine in treating osteoporosis requires cooperation and ongoing research, despite obstacles such limited clinical acceptability and regulatory barriers.
Article
Full-text available
Snail ( Achatina fulica ) is one of the sources of duck fodder commonly used by local breeders to meet protein needs of poultry farms. However, its shells as a by-product are rarely used and just become waste. To overcome this problem, we try to utilize the waste to be used as a source of bioceramic. Snail shells contain a lot of calcium which can be used as a calcium precursor to make bioceramics. One of the bioceramics that has the potential are tricalcium phosphate (TCP) and calcium pyrophosphate (CPP). They are mostly used as material for bone regeneration. It has advantages in terms of biocompatibility and osteoinductivity. In this study, we synthesized and characterized calcium phosphate from snails by coprecipitation method. It was then analyzed using X-Ray fluorescence (XRF), X-Ray diffraction (XRD), fourier transform infrared (FTIR), energy dispersive X-Ray (EDX), and scanning electron microscope (SEM) analysis to see its characteristic. Based on XRF analysis, snail shell (raw material) contains 81.83% of calcium and calcium phosphate mixed bioceramics (TCP and CPP). Average crystal size is 17.00 nm. The surface shape of the formed particles was irregular, and also flux, sphere, and fracture shaped. Nevertheless, interfering compounds were still found, hence a further purification process needs to be conducted.
Article
Inflammation-induced by biomaterials is a critical event to determine the success and efficiency of tissue repair. Macrophages are a major population that participates the biomaterial induced inflammation. The response of macrophages depends on the characteristics of biomaterials, thus causing a cascade reaction in subsequent biological processes. In this study, porous biphase calcium phosphate (BCP) ceramics with the different surface structures were constructed to compare the effect of surface structure on bone generation potential, and further reveal the inflammation-involved mechanism. Our results demonstrated that macrophages on three ceramics showed distinct morphologies and spreading areas. The nanoscale whisker structure did induce more bone generation in the mice thigh muscle. The in vitro result revealed that nanoscale whisker structure could drive macrophage polarization towards M1-like phenotype, indicated by a higher expression of pro-inflammatory specific markers (iNOS and CCR7), and mass secretion of TNF-α. Further research indicated that additional TNF-α could promote the osteogenic differentiation of mesenchymal stem cells (MSCs). However, excess addition of TNF-α showed an opposite effect on the osteogenic differentiation of MSCs by initiating the NF-κB signaling pathway, which suppresses the osteogenesis process.
Article
The effect of the ionic products of Bioglass® 45S5 dissolution on the gene‐expression profile of human osteoblasts was investigated by cDNA microarray analysis of 1,176 genes. Treatment with the ionic products of Bioglass® 45S5 dissolution increased the levels of 60 transcripts twofold or more and reduced the levels of five transcripts to one‐half or less than in control. Markedly up‐regulated genes included RCL, a c‐myc responsive growth related gene, cell cycle regulators such as G1/S specific cyclin D1, and apoptosis regulators including calpain and defender against cell death (DAD1). Other significantly up‐regulated genes included the cell surface receptors CD44 and integrin β1, and various extracellular matrix regulators including metalloproteinases‐2 and ‐4 and their inhibitors TIMP‐1 and TIMP‐2. The identification of differentially expressed genes by cDNA microarray analysis has offered new insights into the mode of action of bioactive glasses and has proven to be an effective tool in evaluating their osteoproductive properties. © 2001 John Wiley & Sons, Inc. J Biomed Mater Res, 55: 151–157, 2001
Article
Vertebroplasty, which is the percutaneous injection of bone cement into vertebral bodies has recently been used to treat painful osteoporotic compression fractures. Early clinical results have been encouraging, but very little is known about the consequences of augmentation with cement for the adjacent, non-augmented level. We therefore measured the overall failure, strength and structural stiffness of paired osteoporotic two-vertebra functional spine units (FSUs). One FSU of each pair was augmented with polymethyl-methacrylate bone cement in the caudal vertebra, while the other served as an untreated control. Compared with the controls, the ultimate failure load for FSUs treated by injection of cement was lower. The geometric mean treated/untreated ratio of failure load was 0.81, with 95% confidence limits from 0.70 to 0.92, (p < 0.01). There was no significant difference in overall FSU stiffness. For treated FSUs, there was a trend towards lower failure loads with increased filling with cement (r ² = 0.262, p = 0.13). The current practice of maximum filling with cement to restore the stiffness and strength of a vertebral body may provoke fractures in adjacent, non-augmented vertebrae. Further investigation is required to determine an optimal protocol for augmentation.
Article
The pharmacological management of disease should involve consideration of the balance between the beneficial effects of treatment on outcome and the probability of adverse effects. The aim of this review is to explore the risk of adverse drug reactions and drug–drug interactions with treatments for postmenopausal osteoporosis. We reviewed evidence for adverse reactions from regulatory documents, randomized controlled trials, pharmacovigilance surveys, and case series. Bisphosphonates are associated with gastrointestinal effects, musculoskeletal pain, and acute-phase reactions, as well as, very rarely, atrial fibrillation, atypical fracture, delayed fracture healing, osteonecrosis of the jaw, hypersensitivity reactions, and renal impairment. Cutaneous effects and osteonecrosis of the jaw are of concern for denosumab (both very rare), though there are no pharmacovigilance data for this agent yet. The selective estrogen receptor modulators are associated with hot flushes, leg cramps, and, very rarely, venous thromboembolism and stroke. Strontium ranelate has been linked to hypersensitivity reactions and venous thromboembolism (both very rare) and teriparatide with headache, nausea, dizziness, and limb pain. The solidity of the evidence base depends on the frequency of the reaction, and causality is not always easy to establish for the very rare adverse reactions. Drug–drug interactions are rare. Osteoporosis treatments are generally safe and well tolerated, though they are associated with a few very rare serious adverse reactions. While these are a cause for concern, the risk should be weighed against the benefits of treatment itself, i.e., the prevention of osteoporotic fracture.