Skip to main content
Intended for healthcare professionals
Free access
Research article
First published online July 10, 2015

Animal Models for Evaluation of Bone Implants and Devices: Comparative Bone Structure and Common Model Uses

Abstract

Bone implants and devices are a rapidly growing field within biomedical research, and implants have the potential to significantly improve human and animal health. Animal models play a key role in initial product development and are important components of nonclinical data included in applications for regulatory approval. Pathologists are increasingly being asked to evaluate these models at the initial developmental and nonclinical biocompatibility testing stages, and it is important to understand the relative merits and deficiencies of various species when evaluating a new material or device. This article summarizes characteristics of the most commonly used species in studies of bone implant materials, including detailed information about the relevance of a particular model to human bone physiology and pathology. Species reviewed include mice, rats, rabbits, guinea pigs, dogs, sheep, goats, and nonhuman primates. Ultimately, a comprehensive understanding of the benefits and limitations of different model species will aid in rigorously evaluating a novel bone implant material or device.
Bone and joint implants and devices are a rapidly growing portion of the health care industry, with an estimated $9 billion in Medicare expenditures spent on implantable orthopedic devices in 2009.17 Considerable research continues toward developing novel devices to improve bone and joint function and health. The implant and device field contains diverse classes, including tissue-engineered constructs, combination products (ie, antimicrobial coatings for devices), and biomaterials (synthetic or natural nondrug substance(s) used as a whole or as a part of a system to treat, augment, or replace any tissue, organ, or function11). For the purpose of this review, the term implant will be used to refer to this wide variety of products.
Animal models are a key component of many nonclinical implant studies, and the ideal species varies by the specific implant and its intended use. This review outlines how bone and joint implants are evaluated in models of human diseases and the advantages and limitations of species commonly used in implant studies.

Evaluating Implants In Vivo: Biological Relevance, Biofunctionality, Biocompatibility/Safety, and Clinical Relevance/Efficacy

Bone and joint implants can have a variety of effects on the host, from providing basic mechanical support to initiating local and systemic host responses. To evaluate both intended and unintended effects, several stages are used to evaluate implants, including biological relevance, biofunctionality, biocompatibility/safety, and clinical relevance/efficacy.

Biological Relevance

Initial laboratory testing is used to demonstrate fundamental biological relevance of an implant’s structure and material composition53 (biological relevance defined here as effects likely to be relevant and meaningful for humans and/or animals). This stage uses a combination of in vitro, ex vivo (ie, tissue explants), and in vivo models. Ex vivo tissue and in vivo animal models provide initial insights into how an implant may alter the physiological, biomechanical, and hormonal functions of bone. This stage of study often uses smaller animal models (rabbits, rodents), including animals that mimic specific human conditions (ie, transgenic or knockout mice, guinea pigs with spontaneous degenerative joint disease).

Biofunctionality

A related testing stage that can be separate from or combined with biological relevance is biofunctionality (defined here as the ability of an implant to alter tissue function in a desired way). Unique bone and joint properties often measured include the following:53,78
Chondrogenesis: the ability of an implant to induce or support regrowth of cartilage
Osteointegration: growth of tissue into or along the surface of implanted material (Figs. 1–3)
Osteoconductivity: ability of a scaffold to promote/support osteogenic cells within a defined space
Osteoinduction: soluble or matrix-bound signals that interact with and drive local uncommitted host stem cells/progenitors toward an osteoblastic phenotype; osteoinductive implants can induce bone formation at heterotopic sites (ie, muscle; Fig. 4)9
Figure 1. Local cortical tissue response to implant, femur cortical bone, rabbit. Local tissue response to implantation of high-density plastic (*) at 26 weeks prior, showing no significant osteointegration or immune response. New bone has been deposited (osteointegration) along the marrow surface (black arrow) and the endosteal surface (white arrows). Exakt processed fully mineralized section. Hematoxylin and eosin (HE). Figure 2. Local periosteal reaction without significant osteointegration in response to stainless steel implant, femur cortical bone, rabbit. Fibrous connective tissue directly lines the implant surface (*) with minimal giant cell response along the implant surface (arrow). Woven and plexiform bone has formed adjacent to the fibrous connective tissue. Exakt processed fully mineralized section. HE. Figure 3. (a) Woven bone response to alveolar bone resection, oral cavity, dog. Proliferation of woven bone in response to resection of alveolar bone to depth of a reference notch created in the tooth at the time of surgery (vertical black line). Red staining predominates on the surface (newer bone, thin arrow) adjacent to deeper blue-stained bone (older bone, thick arrow). Implanted bone void filler (asterisk) is surrounded by macrophages and giant cells (shown in Fig. 3b) and shows no osteointegration. Decalcified, modified Masson’s trichrome. (b) Immune response to bone void filler, oral cavity, dog. A serial section of the region indicated by the asterisk in Figure 3a at higher magnification, showing the presence of giant cells (thick arrow) and macrophages (thin arrow) adjacent to bone void filler (*). Decalcified, HE.
Figure 4. Osteoinduction within xenograft, skeletal muscle, athymic mouse. Demineralized human bone implanted within skeletal muscle for 35 days facilitated production of hyaline cartilage within preexisting osteonal canals (thin black arrow) and appositional woven bone deposition (thick black arrows indicating osteocytes) along the surface of the decellularized implant (white arrows indicating empty lacunae). Decalcified, hematoxylin and eosin.
As with evaluating biological relevance, biofunctionality is often tested in rodents and rabbits if the implant can be scaled to a small size and placed in a bone with similar physiology to that of the intended human site. If small animal models are not appropriate, larger species are used (dogs, pigs, small ruminants).

Biocompatibility/Safety

While biofunctionality tests if the implant has the desired effects (ie, osteoinduction when implanted in muscle), biocompatibility/safety studies can identify undesirable local or systemic effects (ie, immune reaction to osteoinductive material).30 Biocompatible implants (and any breakdown or leachable derivatives) should be nontoxic, nonthrombogenic, noncarcinogenic, nonantigenic, and nonmutagenic.30 Safety studies often use rodents and rabbits to detect local tissue damage (often including histiocytic and multinucleated cell infiltrates; Fig. 3b)47,80 and systemic toxicities (often from breakdown or leachable derivatives). While tissue reactions to specific materials are not discussed here, recent reviews30,47 provide detailed information about host reactions to different materials.

Clinical Relevance/Efficacy

Finally, while functionality and safety can be established in smaller species, it may be necessary to demonstrate efficacy in larger animals (dogs, small ruminants, pigs, nonhuman primates [NHPs]), where local blood flow and mechanical loading can be critical to implant success.53,78

Diseases/Conditions Commonly Modeled

Enhancing Osteoinductive and Chondrogenic Potential After Injury

Implants that initiate or enhance bone or cartilage growth are highly desirable for injured tissues, especially those that have limited vascular or marrow supply to support intrinsic repair (ie, membranous craniofacial bone79). Osteoinductive and/or chondrogenic potential is often measured in athymic mice and rats.65,79 Chondrogenic implants are used in an attempt to repair synovial joints16,65 and intervertebral disks. Osteoinductive implants are used as interventions to repair vertebral fractures,10,79 during dental and craniofacial bone surgery,18,79 and in segmental defects/nonunions (discussed in detail below).

Large Bone Defects and Nonunions

Large bone defects occur secondary to a pathologic event (ie, trauma, inflammation, surgical resection64) that causes extensive bone loss. In human clinical settings, critical-size defects are defined as those that do not heal within ∼6 months postinjury in otherwise healthy adults.12 Affected sites (ie, anteromedial tibia20) often have limited adjacent muscle that could provide physical coverage and an auxiliary blood supply. Critical-size defects in animal models are defined as “the smallest size intraosseous wound in a particular bone and species of animal that will not heal spontaneously during the lifetime of the animal”27,67 or a defect with <10% bone regeneration over the animal’s lifetime.27 Surgically created critical defects are a gap in length >2.25 times the diameter of the affected bone26,46 or large cylindrical defects in femoral condyles or tibial metaphysis. Rats, rabbits, and increasingly small ruminants are used in defect models.59 One advantage of small ruminants is that their repair rate is more comparable to humans than rodents (a large tibial defect can fully repair within 12 weeks in rats but may take 32–52 weeks in goats or sheep32).

Implant-Related Complications

Finally, while implants have an overall positive impact on human health, individuals may have implant-related complications (usually modeled in rodents and rabbits). Negative responses to implants can include reactions to wear debris, osteolytic bone resorption, anatomic malalignment, implant instability/breakage, heterotopic ossification, pain, and immunologic reactions/hypersensitivity.80 Over 100 000 cases of implant-associated infection and osteomyelitis are reported annually in the United States.55 On average, 2% to 5% of internal fixation devices become infected19,74 and may require surgical removal to eliminate the infection. Up to 70% of organisms isolated in prosthetic joint infections are Gram-positive agents, including Staphylococcus epidermidis, Staphylococcus aureus, and β-hemolytic streptococci.55,80

Human Bone: Terminology, Organization, and Development

Terminology

Understanding how human bone is organized and develops is important when selecting an appropriate model species. Since there are multiple anatomic and histologic terms for some bone structures, in this review, a single term is used for each feature, with alternate terms listed in parentheses. Microscopically, bone is generally organized into woven (type I collagen fibers arranged haphazardly) or lamellar (collagen in parallel layers or concentric rings) patterns. Additionally, plexiform (or laminar or fibrolamellar) bone is produced within the periosteum when significant bone strength and rapid deposition are required. Plexiform bone is lamellar bone deposited on brick-like cores of nonlamellar bone (Fig. 2).60 Plexiform, woven, primary lamellar, and primary osteons are forms of primary bone (formed during initial bone modeling on a substrate of bone or cartilage without resorption of preexisting bone). Secondary bone includes secondary osteons (Haversian systems) that replace primary bone through remodeling (coordinated deposition of new bone with resorption of preexisting bone). Macroscopically, bone is generally organized into cortical (or compact, referring to filling in spaces in plexiform bone) and cancellous bone (or trabecular or spongy or medullary). Structural differences between the 2 forms include bone density (cortical > cancellous), vascular access (cancellous > cortical), and presence of full osteons (cortical) or hemiosteons (cancellous).

Organization

All normal adult human cortical bone is lamellar with extensive secondary osteonal remodeling.28,36 Plexiform bone is rarely present in humans, and in adults, woven bone is only formed during rapid bone formation (ie, fracture callus, hyperparathyroidism).45,64 Macroscopically, approximately 80% of the adult human skeleton comprises cortical bone (by weight), and the remaining 20% is trabecular.24,34 Load-bearing long bones are 80% to 100% cortical bone, while vertebrae are 80% cancellous and 20% cortical.69 Subchondral plate bone adjacent to articular cartilage is generally thin36 with minimal regenerative capabilities.

Development

Ossification in humans begins early in the neonatal period and continues throughout early adulthood, with fusion of the proximal and distal femoral epiphyses completed by 19 to 20 years in males and 2 to 3 years earlier in females.15,53,84 Human remodeling rates range from 10% to 55% (average whole-body trabecular bone turnover per year64), with age-associated cortical bone loss occurring primarily at the endocortical endosteum.10 Bone adjacent to red marrow generally has higher rates of remodeling (eg, 10%–15% per year in ilium trabecular bone) compared with sites adjacent to yellow (fatty) marrow.63

Factors to Consider When Selecting a Model

Certain characteristics are desirable when selecting any animal model, including low intra- and interanimal variation, the availability of sensitive methods to assess a biological parameter, and the use of treatment conditions that most closely match human clinical disease.53 Additional factors should be considered in bone and joint models (Table 1). First, since implants may be intended for use in either cortical or cancellous bone, it is important to identify which sites and at what age a model species’ macro- and microstructure best matches human bone. Second, blood supplies that support bone healing can differ markedly between species84 and by macroscopic structure (cancellous > cortical). Third, control groups or sites (ie, sham-operated, autologous cancellous bone graft78) can aid in differentiating implant effects on bone repair from intrinsic bone-healing abilities (which can be significant in rodents and rabbits). Next, repair responses in skeletally immature animals can be more rapid than in mature adults.65 For example, femoral fractures have been shown to heal within 4 weeks in 6-week-old rats but may take up to 26 weeks in 52-week-old rats.51 These age-related changes can be further complicated by sexual dimorphism, which has been well documented in rodents and is influenced by sex hormones.40 Finally, implants that cannot be scaled down (ie, joint replacement) require a large animal model.53
Table 1. Species Factors to Consider When Selecting a Bone/Joint Implant Model.
Factor Implications for Model
Implant site Cancellous vs cortical vs specialized (ie, alveolar bone in oral cavity)
  Microstructure at implant site
  Bone large enough to accept human-sized implant
  Vascular supply to bone
  Adjacent soft tissue that will be disturbed during implantation/provide vascular support
Control groups and materials Autologous graft availability (gold standard for bone repair material)
  Sham-operated sites
Nutrition Reduced healing under food restriction or reduced ability to eat postimplant28,76
Age Greater bone and cartilage healing ability in younger animals15,57
Genetic background Strain variability, inbred vs outbred34,43
The following sections outline common model uses and comparative bone pathology for the most common preclinical species (summarized in Table 2).
Table 2. Summary of Human and Animal Bone Properties.
Species Secondary Centers of Ossification Present at Birth28 Macroscopic Similarity to Human Bone37,5253,60 Microscopic Structure45,60,62,64,81 Osteonal Remodeling10,28,48,56,66 Age of Skeletal Maturity3,11,15,52 Average Cartilage Thickness (Femur, mm)16
Human Yes, limited to distal femur and proximal tibia NA Circumferential lamellar bone with extensive remodeling Yes (in utero) 16–20 y15,84 2.2–2.523
NHP No Very similar Similar to human Yes (age varies by species) 5 y (baboon), 7 y (rhesus) 0.57–0.72 (cynomolgus)7,13
Rodent No Not similar Primary bone predominates over life span Minimal, advanced age ∼26 wk, with potential for continuous growth68 0.030–0.1 (mouse), 0.17 (rat)73
Rabbit No Minimally similar Primary bone predominates, secondary osteons develop with age Minimal, advanced age 8–11 mo 0.2–0.44
Dog No Moderately similar Plexiform bone, some remodeling to small secondary osteons Yes (<1 y) 10–18 mo83 0.6–1.3
Pig Yes Moderately similar Plexiform with transition to dense secondary osteonal Yes (>6 m) 18–24 mo (mini),75 2–4 y (conventional) 1.5
Small ruminant Yes Very similar Plexiform until 7–9 y (sheep), heterogeneous secondary osteonal with age (goat) Yes (>1 y) 15–18 mo (sheep), 2–3 y (goat) 0.4–0.5 (sheep), 0.6–1.7 (goat)57
NA, not applicable; NHP, nonhuman primate.

Rodents

Model Uses

Knockout and transgenic rodent models (ie, senescence accelerated mice) are a key resource in the biological relevance stage of implant studies.12,48 Athymic nude mice and rats are also commonly used to test osteoinductive and cartilage regenerative potential of allogenic and xenogeneic materials.16 Rats are used to model bone infection55 and extraoral surgical approaches.18 While rodents have limited cortical remodeling or secondary osteon formation, they can model cancellous bone responses in lumbar vertebral bodies and the distal femoral and proximal tibial metaphyses.4,28,63

Relevance to Human Bone and Joint Conditions

There are significant limitations to consider when extrapolating implant findings from rodents to humans, including differences in bone micro- and macrostructure, rates of growth, and size. Unlike the secondary bone present in the adult human long bone cortices, rodent long bone cortices are predominantly primary bone and have minimal cancellous bone available to harvest for autografts.53 Bone growth in rats continues much longer after sexual maturity than in humans.39 Bone-healing capacity is higher in rodents than in humans at sites such as craniofacial bones,71 and the retention of open physes in adults may enhance intrinsic articular cartilage healing potential.16 Finally, some implants cannot be scaled down to a rodent size to test clinical equivalency.12

Guinea Pig

While guinea pigs are not a common model for bone implants, strains that develop spontaneous degenerative joint disease, like the Dunkin-Hartley, are used to evaluate cartilage regeneration and joint support implants. Dunkin-Hartley male guinea pigs develop histologic lesions by 3 months of age, which progress within 6 months to lesions that closely resemble human joint changes.38,76

Rabbit

Model Uses

Rabbit hindlimbs are commonly used for cortical and cancellous bone implant studies. The tibia is easily accessible due to minimal associated soft tissue, and the femur has sufficient medullary space for many internal fixation studies.14 Mature rabbit condyles can support 3- to 4-mm defects to test biocompatibility and osteoinduction in cancellous bone.16 Rabbits are also used to model vertebral fracture repair, methods to control common implant-associated pathogens (ie, Staphylococcus epidermidis14,22), effects of wear debris particles,12 and extraoral surgical approaches.18

Relevance to Human Bone and Joint Conditions

Rabbits offer advantages over large animal models by reaching skeletal maturity at a relatively early age and advantages over rodents by undergoing more secondary osteonal remodeling.53 Despite these positive characteristics, there are significant differences between rabbit and human bone. The rate of cortical bone remodeling is rapid in the young rabbits often used in studies, which can be less representative of the healing response in adult humans.78 Rabbits also have relatively fatty bone marrow, which is not ideal for autogenous bone grafts.53 Rabbit femoral condyle cartilage is relatively thin,5,65 and articular cartilage in young, 3- to 9-month-old rabbits has rapid healing potential that can be a confounding factor in long-term studies.16,82

Dog

Model Uses

Dogs are commonly used in dental studies60 and are a valuable model of peri-implantitis (inflammation of tissue adjacent to dental implants), due to their susceptibility to biofilm accumulation and spontaneous periodontitis.18 Large-breed dogs can often support human-sized implants,60 and their large synovial joints, such as the stifle, can be monitored via arthroscopy.16 Additionally, sufficient material for autogenous bone grafts can be harvested from the proximal humerus.53 Finally, dogs are also amenable to studies that evaluate how rehabilitation activities affect implant function.16

Relevance to Human Bone and Joint Conditions

Canine and human bone have many similarities, including formation of secondary osteons, epiphyseal fusion after maturity, comparable intracortical remodeling activity,28 and age-associated bone loss.10 The 2 species also have similar bone weight, density, and composition of bone organic, inorganic, and water fractions.1,64 Humans and dogs also both lack substantial intrinsic healing ability for cartilage defects.16 Key differences include the retention of plexiform bone on canine cortical periosteal and endosteal surfaces45,81 and thinner articular cartilage in dogs.16 Dogs have higher annual total-body trabecular bone turnover rates (averaging 94%–101% in male and female canine trabecular bone compared with an estimated 10%–55% in adult humans41,60,64), which may partially explain the high rate of solid bony fusion and low rates of nonunion in canine models.64 One important limitation of using dogs is that remodeling and apposition rates can vary significantly both within and between dogs.25 For example, individual ribs in a single-purpose-bred Beagle may have statistically different bone remodeling parameters.6

Pig (Conventional and Minipig)

Model Uses

Conventional pigs and minipigs are used to evaluate bone osteogenic material in craniomaxillary facial surgery models,12 extra- and intraoral surgical approaches for dental implants,18 interventions for femoral head osteonecrosis, and repair of cartilage and bone fractures.60 Sufficient material for autogenous cancellous bone grafts can be harvested from the proximal humerus.53 Skeletally mature conventional pig articular cartilage is also thick enough (∼1.5 mm) to create both full- and partial-thickness cartilage defects that can be monitored via arthroscopy.16

Relevance to Human Bone and Joint Conditions

Bone mineral density, anatomy, morphology, remodeling rate, and healing of pigs are similar to humans.64 Pigs have both growing and adult skeletal phases5 with well-developed Haversian systems in mature bone.10 Swine intra-articular cartilage and associated ligaments in synovial joints are similar to the conformation in human joints.75 Disadvantages include high growth rates, excessive body weight, and difficulty in being handled compared with other species.64 Pig tibial and femoral length is also relatively short, which can prevent direct testing of human-sized implants64 or creation of stable, critical-size defects.53

Sheep and Goats

Model Uses

Use of small ruminant orthopedic models is increasing, with reports of sheep and goat models growing from ∼5% of bone papers in the 1980s to 11% to 15% in the 2000s.50,59 Small ruminants are used to model defect repair in long bones, bone filler materials in cranial defects, fracture repair devices, tissue response to wear debris, extraoral surgical approaches, and intervertebral disk replacements.12,18 The relatively vertical alignment of the cervical vertebrae in small ruminants undergoes some of the same axial compression and rotation forces as the human spine.53

Relevance to Human Bone and Joint Conditions

Small ruminants and humans have a comparable body weight, bone size, and bone-healing potential, as well as tibial blood supply (young sheep) and bone composition and remodeling rates (goats).64 Additionally, the ratio of cartilage to subchondral bone, subchondral bone consistency, joint size, accessibility for arthroscopy, and limited intrinsic healing capacity in goats are similar to human joints.2,16,33 Autogenous cancellous grafts can also be performed in small ruminants.53 Despite these similarities, there are several important differences between small ruminant and human bone. Sheep cortical bone remains predominantly plexiform through 7 to 9 years of age.64,66 Sheep undergo seasonal bone loss, and if a study occurs during winter (when bone formation is depressed), control animals may spontaneously lose and regain bone.10 While in mature sheep, trabecular bone is more dense and stronger than human bone, immature sheep have weaker, low-density trabecular bone that is highly flexible (due to higher collagen content54).

NHPs

NHPs and humans share significant genetic homology, have comparable osteonal remodeling in skeletally mature individuals, and develop similar diseases, including osteoarthritis and age-associated bone loss.37 Comparable bone biopsy sites are also available in both species, such as vertebral bodies, the cortex of the seventh rib, proximal femur, and ilium.4,63 NHPs are generally used in bone implant studies when other large animal models do not adequately represent human bone biomechanics and loading, including dental implants and vertebral repair products. The tooth and jaw structure of NHPs provide relevant information about stress responses to mechanical loading of dental implants.18 Baboons are also used to evaluate specialized intervertebral artificial disks.12 While NHPs have key advantages as a model for these specialized implant sites, implants for other sites such as long bones are sometimes better suited for other large animal models. NHP long bones can be too small to support adult human implants. For example, a mature male rhesus has a ∼16.5-cm-long femur with a 1.25-cm mid-shaft diameter while mature hounds can have a 20-cm-long femur with a 2.5- to 3-cm mid-shaft diameter.53

Study Design, Nomenclature, and Scoring Criteria for Bone/Joint Implants

While many factors used to design effective toxicology studies of small and large molecules are also applicable to bone implant studies, there are several additional criteria to consider. Reactions to implants are generally time, not dose, dependent.72 Implants designed to promote bone growth may require studies lasting longer than 6 months to allow sufficient time for bone regeneration, remodeling, and detection of implant absorption or degradation.64 Also, implant placement almost always incites a significant nonspecific localized tissue reaction, secondary to surgical/implantation trauma.72 It is therefore important to include sham surgery and/or control implants to differentiate reactions that are implant induced from those that are primarily implantation induced.
Histologic processing of bone and joint implant samples can be uniquely challenging, and variations in implants are too numerous to be addressed here. Readers interested in processing specific implant materials and common bone-staining methods are referred to recent publications.4,70 Additional analytic techniques (macroscopic evaluation, radiographs, computed tomography [CT], micro-CT8) can also provide key information on 3D structure and device surface-tissue interactions, factors that can be difficult to appreciate with histology alone.
Excellent resources are available for both the design and interpretation of in vivo studies, including guidances that are the basis for implant regulations in several countries (Table 3). Clinical and basic bone researchers have developed standardized histomorphometry terms and evaluation methods to provide consistency across studies.21,63 Cartilage scoring criteria are recommended by the International Cartilage Repair Society (ICRS, preferred for implant studies31,49) and the Osteoarthritis Research Society International Histopathology Initiative (OARSI, preferred for drug-related effects42,43,61). Finally, standard nomenclature and image resources for spontaneous bone and joint lesions are available in the forthcoming bone, cartilage, and tooth INHAND manuscript (http://goreni.org/index.php) and National Toxicology Program Non-neoplastic Lesion Atlas (http://ntp.niehs.nih.gov/nnl/guide/index.htm).
Table 3. International Standards for Biological Evaluation of Bone and Joint Implants.
Issuing Organization Primary Category Standard No. Standard Name
International Organization for Standardization (ISO) Biocompatibility 10993-6 Tests for local effects after implantation
  10993-11 Tests for systemic toxicity
ASTM (formerly American Society for Testing and Materials) Biocompatibility F748—06 (2010) Standard Practice for Selecting Generic Biological Test Methods for Materials and Devices
F763—04 (2010) Standard Practice for Short-Term Screening of Implant Materials
F981—04 (2010) Standard Practice for Assessment of Compatibility of Biomaterials for Surgical Implants with Respect to Effect of Materials on Muscle and Bone
F1027—86 (2012) Standard Practice for Assessment of Tissue and Cell Compatibility of Orofacial Prosthetic Materials and Devices
F1408—97 (2013) Standard Practice for Subcutaneous Screening Test for Implant Materials
F1439—03 (2013) Standard Guide for Performance of Lifetime Bioassay for the Tumorigenic Potential of Implant Materials
F1904—14 Standard Practice for Testing the Biological Responses to Particles In Vivo
  F1983—99 (2008) Standard Practice for Assessment of Compatibility of Absorbable/Resorbable Biomaterials for Implant Applications
Biofunctionality F2721-09 Standard Guide for Preclinical In Vivo Evaluation in Critical Sized Segmental Bone Defects
F2451—05 (2010) Standard Guide for In Vivo Assessment of Implantable Devices Intended to Repair or Regenerate Articular Cartilage
F2529—13 Standard Guide for In Vivo Evaluation of Osteoinductive Potential for Materials Containing Demineralized Bone (DBM)
F2721—09 Standard Guide for Preclinical In Vivo Evaluation in Critical Size Segmental Bone Defects
F2884—12 Standard Guide for Preclinical In Vivo Evaluation of Spinal Fusion

Concluding Thoughts

As implant development and clinical use continues to expand, it is important for pathologists evaluating bone and joint implant studies to understand the basics of bone structure and the advantages and disadvantages of different animal models. While no single animal model completely recapitulates human bone and joint development and repair, various species can provide key support when developing implants.

Acknowledgements

Dr David H. Johnson provided assistance with reference formatting and figure preparation.

Declaration of Conflicting Interests

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding

The author(s) received no financial support for the research, authorship, and/or publication of this article.

References

1. Aerssens J, Boonen S, Lowet G, et al. Interspecies differences in bone composition, density, and quality: potential implications for in vivo bone research. Endocrinology. 1998;139(2):663–670.
2. Ahern BJ, Parvizi J, Boston R, et al. Preclinical animal models in single site cartilage defect testing: a systematic review. Osteoarthritis Cartilage. 2009;17(6):705–713.
3. Aigner T, Cook JL, Gerwin N, et al. Histopathology atlas of animal model systems—overview of guiding principles. Osteoarthritis Cartilage. 2010;18(suppl 3):S2–S6.
4. Allen MR, Burr DB. Techniques in histomorphometry. In: Burr DB, Allen MR eds. Basic and Applied Bone Biology. Amsterdam, the Netherlands: Academic Press; 2014:131–148.
5. An YH, Freidman RJ. Animal selections in orthopaedic research. In: An YH, Freidman RJ, eds. Animal Models in Orthopaedic Research. Boca Raton, FL: CRC Press; 1999:39–57.
6. Anderson C, Danylchuk KD. Bone-remodeling rates of the Beagle: a comparison between different sites on the same rib. Am J Vet Res. 1978;39(11):1763–1765.
7. Athanasiou KA, Rosenwasser MP, Buckwalter JA, et al. Interspecies comparisons of in situ intrinsic mechanical properties of distal femoral cartilage. J Orthop Res. 1991;9(3):330–340.
8. Bagi CM, Berryman E, Moalli MR. Comparative bone anatomy of commonly used laboratory animals: implications for drug discovery. Comp Med. 2011;61(1):76–85.
9. Barradas AM, Yuan H, van Blitterswijk CA, et al. Osteoinductive biomaterials: current knowledge of properties, experimental models and biological mechanisms. Eur Cell Mater. 2011;21:407–429.
10. Bonucci E, Ballanti P. Osteoporosis—bone remodeling and animal models. Toxicol Pathol. 2014;42(6):957–969.
11. Boretos JW, Eden M. Contemporary Biomaterials, Material and Host Response, Clinical Applications, New Technology and Legal Aspects. Park Ridge, NJ: Noyes Publications; 1984.
12. Boutrand J. Methods and interpretation of performance studies for bone implants. In: Boutrand J, ed. Biocompatibility and Performance of Medical Devices. Oxford, UK: Woodhead Publishing Limited; 2012:272–279.
13. Buckwalter JA, Martin JA, Olmstead M, et al. Osteochondral repair of primate knee femoral and patellar articular surfaces: implications for preventing post-traumatic osteoarthritis. Iowa Orthop J. 2003;23:66–74.
14. Burkholder TH, Linton G, Hoyt RF Jr, et al. The rabbit as an experimental model. In: Suckow MA, Stevens KA, Wilson RP, eds. The Laboratory Rabbit, Guinea Pig, Hamster, and Other Rodents. Amsterdam, the Netherlands: Academic Press; 2012:548–550.
15. Cardoso HF. Epiphyseal union at the innominate and lower limb in a modern Portuguese skeletal sample, and age estimation in adolescent and young adult male and female skeletons. Am J Phys Anthropol. 2008;135(2):161–170.
16. Chu CR, Szczodry M, Bruno S. Animal models for cartilage regeneration and repair. Tissue Eng Part B Rev. 2010;16(1):105–115.
17. Cosgrove J. MEDICARE: Lack of Price Transparency May Hamper Hospitals’ Ability to Be Prudent Purchasers of Implantable Medical Devices. 2012;GAO-12-126 Implantable Medical Devices, St. Louis.
18. Dard M. Methods and interpretation of performance studies for dental implants. In: Boutrand J, ed. Biocompatibility and Performance of Medical Devices. Oxford, UK: Woodhead Publishing Limited; 2012:308–344.
19. Darouiche RO. Treatment of infections associated with surgical implants. N Engl J Med. 2004;350(14):1422–1429.
20. DeCoster TA, Gehlert RJ, Mikola EA, et al. Management of posttraumatic segmental bone defects. J Am Acad Orthop Surg. 2004;12(1):28–38.
21. Dempster DW, Compston JE, Drezner MK, et al. Standardized nomenclature, symbols, and units for bone histomorphometry: a 2012 update of the report of the ASBMR Histomorphometry Nomenclature Committee. J Bone Miner Res. 2013;28(1):2–17.
22. Donlan RM. Biofilm formation: a clinically relevant microbiological process. Clin Infect Dis. 2001;33(8):1387–1392.
23. Frisbie DD, Cross MW, McIlwraith CW. A comparative study of articular cartilage thickness in the stifle of animal species used in human pre-clinical studies compared to articular cartilage thickness in the human knee. Vet Comp Orthop Traumatol. 2006;19(3):142–146.
24. Frost HM. Introduction to a New Skeletal Physiology. 2 vols. Pueblo, CO: Pajaro Group; 1995.
25. Greaves P. Histopathology of Preclinical Toxicity Studies: Interpretation and Relevance in Drug Safety Evaluation. Amsterdam, the Netherlands: Academic Press; 2012.
26. Gugala Z, Lindsey RW, Gogolewski S. New approaches in the treatment of critical-size segmental defects in long bones. Macromol Symp. 2007;253:147–161.
27. Gugala Z, Gogolewski S. Regeneration of segmental diaphyseal defects in sheep tibiae using resorbable polymeric membranes: a preliminary study. J Orthop Trauma. 1999;13(3):187–195.
28. Haschek WM, Rousseaux CG, Wallig MA. Bones and joints. In: Haschek WM, Rousseaux CG, Wallig MA eds. Fundamentals of Toxicologic Pathology. 2nd ed. Amsterdam, the Netherlands: Academic Press; 2010:416–417.
29. Heinrichs C, Colli M, Yanovski JA, et al. Effects of fasting on the growth plate: systemic and local mechanisms. Endocrinology. 1997;138(12):5359–5365.
30. Helmus MN, Gibbons DF, Cebon D. Biocompatibility: meeting a key functional requirement of next-generation medical devices. Toxicol Pathol. 2008;36(1):70–80.
31. Hoemann C, Kandel R, Roberts S, et al. International Cartilage Repair Society (ICRS) recommended guidelines for histological endpoints for cartilage repair studies in animal models and clinical trials. Cartilage. 2011;2(2):153–172.
32. Horner EA, Kirkham J, Wood D, et al. Long bone defect models for tissue engineering applications: criteria for choice. Tissue Eng Part B Rev. 2010;16(2):263–271.
33. Jackson DW, Lalor PA, Aberman HM, et al. Spontaneous repair of full-thickness defects of articular cartilage in a goat model: a preliminary study. J Bone Joint Surg Am. 2001;83(1):53–64.
34. Jee WSS. Structure and function of bone tissue. In: Bronner F, Worrell RV, eds. Orthopaedics: Principles of Basic and Clinical Science. Boca Raton, FL: CRS Press; 1999:5–25.
35. Jepsen KJ, Price C, Silkman LJ, et al. Genetic variation in the patterns of skeletal progenitor cell differentiation and progression during endochondral bone formation affects the rate of fracture healing. J Bone Miner Res. 2008;23(8):1204–1216.
36. Jerome C, Hoch B. Skeletal system. In: Treuting PM, Dintzis SM, eds. Comparative Anatomy and Histology: A Mouse and Human Atlas. Amsterdam, the Netherlands: Academic Press; 2012:53–64.
37. Jerome CP, Peterson PE. Nonhuman primate models in skeletal research. Bone. 2001;29(1):1–6.
38. Jimenez PA, Glasson SS, Trubetskoy OV, et al. Spontaneous osteoarthritis in Dunkin Hartley guinea pigs: histologic, radiologic, and biochemical changes. Lab Anim Sci. 1997;47(6):598–601.
39. Kilborn SH, Trudel G, Uhthoff H. Review of growth plate closure compared with age at sexual maturity and lifespan in laboratory animals. Contemp Top Lab Anim Sci. 2002;41(5):21–26.
40. Kim BT, Mosekilde L, Duan Y, et al. The structural and hormonal basis of sex differences in peak appendicular bone strength in rats. J Bone Miner Res. 2003;18(1):150–155.
41. Kimmel DB, Jee WS. A quantitative histologic study of bone turnover in young adult beagles. Anat Rec. 1982;203(1):31–45.
42. Kraus VB, Huebner JL, DeGroot J, et al. The OARSI histopathology initiative—recommendations for histological assessments of osteoarthritis in the guinea pig. Osteoarthritis Cartilage. 2010;18(suppl 3):S35–S52.
43. Laverty S, Girard CA, Williams JM, et al. The OARSI histopathology initiative—recommendations for histological assessments of osteoarthritis in the rabbit. Osteoarthritis Cartilage. 2010;18(suppl 3):S53–S65.
44. Li X, Gu W, Masinde G, et al. Genetic variation in bone-regenerative capacity among inbred strains of mice. Bone. 2001;29(2):134–140.
45. Liebschner MA. Biomechanical considerations of animal models used in tissue engineering of bone. Biomaterials. 2004;25(9):1697–1714.
46. Lindsey RW, Gugala Z, Milne E, et al. The efficacy of cylindrical titanium mesh cage for the reconstruction of a critical-size canine segmental femoral diaphyseal defect. J Orthop Res. 2006;24(7):1438–1453.
47. Long PH. Medical devices in orthopedic applications. Toxicol Pathol. 2008;36(1):85–91.
48. Long PH, Leininger JR. Bones, joints, and synovia. In: Maronpot RR, Boorman GA, Gaul BW, eds. Pathology of the Mouse. St Louis, MO: Cache River Press; 1999:645–652.
49. Mainil-Varlet P, Van Damme B, Nesic D, et al. A new histology scoring system for the assessment of the quality of human cartilage repair: ICRS II. Am J Sports Med. 2010;38(5):880–890.
50. Martini L, Fini M, Giavaresi G, et al. Sheep model in orthopedic research: a literature review. Comp Med. 2001;51(4):292–299.
51. Meyer RA Jr, Tsahakis PJ, Martin DF, et al. Age and ovariectomy impair both the normalization of mechanical properties and the accretion of mineral by the fracture callus in rats. J Orthop Res. 2001;19(3):428–435.
52. Mosekilde L, Weisbrode SE, Safron JA, et al. Calcium-restricted ovariectomized Sinclair S-1 minipigs: an animal model of osteopenia and trabecular plate perforation. Bone. 1993;14(3):379–382.
53. Muschler GF, Raut VP, Patterson TE, et al. The design and use of animal models for translational research in bone tissue engineering and regenerative medicine. Tissue Eng Part B Rev. 2010;16(1):123–145.
54. Nafei A, Danielsen CC, Linde F, et al. Properties of growing trabecular ovine bone, part I: mechanical and physical properties. J Bone Joint Surg Br. 2000;82(6):910–920.
55. Nair MB, Kretlow JD, Mikos AG, et al. Infection and tissue engineering in segmental bone defects—a mini review. Curr Opin Biotechnol. 2011;22(5):721–725.
56. Newman E, Turner AS, Wark JD. The potential of sheep for the study of osteopenia: current status and comparison with other animal models. Bone. 1995;16(4)(suppl):277S–284S.
57. Niederauer GG, Niederauer GM, Cullen LC Jr, et al. Correlation of cartilage stiffness to thickness and level of degeneration using a handheld indentation probe. Ann Biomed Eng. 2004;32(3):352–359.
58. O’Driscoll SW, Saris DB, Ito Y, et al. The chondrogenic potential of periosteum decreases with age. J Orthop Res. 2001;19(1):95–103.
59. O’Loughlin PF, Morr S, Bogunovic L, et al. Selection and development of preclinical models in fracture-healing research. J Bone Joint Surg Am. 2008;90(suppl 1):79–84.
60. Pearce AI, Richards RG, Milz S, et al. Animal models for implant biomaterial research in bone: a review. Eur Cell Mater. 2007;13:1–10.
61. Pritzker KP, Gay S, Jimenez SA, et al. Osteoarthritis cartilage histopathology: grading and staging. Osteoarthritis Cartilage. 2006;14(1):13–29.
62. Qin L, Mak AT, Cheng CW, et al. Histomorphological study on pattern of fluid movement in cortical bone in goats. Anat Rec. 1999;255(4):380–387.
63. Recker RR, Kimmel DB, Dempster D, et al. Issues in modern bone histomorphometry. Bone. 2011;49(5):955–964.
64. Reichert JC, Saifzadeh S, Wullschleger ME, et al. The challenge of establishing preclinical models for segmental bone defect research. Biomaterials. 2009;30(12):2149–2163.
65. Reinholz GG, Lu L, Saris DB, et al. Animal models for cartilage reconstruction. Biomaterials. 2004;25(9):1511–1521.
66. Reinwald S, Burr D. Review of nonprimate, large animal models for osteoporosis research. J Bone Miner Res. 2008;23(9):1353–1368.
67. Rimondini L, Nicoli-Aldini N, Fini M, et al. In vivo experimental study on bone regeneration in critical bone defects using an injectable biodegradable PLA/PGA copolymer. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2005;99(2):148–154.
68. Roach HI, Mehta G, Oreffo RO, et al. Temporal analysis of rat growth plates: cessation of growth with age despite presence of a physis. J Histochem Cytochem. 2003;51(3):373–383.
69. Rosenberg AE, Roth SI. Bone. In: Mills SE, ed. Histology for Pathologists. 4th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2012:85–106.
70. Rousselle S, Wicks J. Preparation of medical devices for evaluation. Toxicol Pathol. 2008;36(1):81–84.
71. Schmitz JP, Hollinger JO. The critical size defect as an experimental model for craniomandibulofacial nonunions. Clin Orthop Relat Res. 1986;205:299–308.
72. Schuh JC. Medical device regulations and testing for toxicologic pathologists. Toxicol Pathol. 2008;36(1):63–69.
73. Simon WH. Scale effects in animal joints, I: articular cartilage thickness and compressive stress. Arthritis Rheum. 1970;13(3):244–256.
74. Stinner DJ, Keeney JA, Hsu JR, et al. Outcomes of internal fixation in a combat environment. J Surg Orthop Adv. 2010;19(1):49–53.
75. Swindle MM, Makin A, Herron AJ, et al. Swine as models in biomedical research and toxicology testing. Vet Pathol. 2012;49(2):344–356.
76. Taylor DK, Lee VK. Guinea pigs as experimental models. In: Suckow MA, Stevens KA, Wilson RP, eds. The Laboratory Rabbit, Guinea Pig, Hamster, and Other Rodents. Amsterdam, the Netherlands: Academic Press; 2012:709–710.
77. van Leeuwen BL, Hartel RM, Jansen HW, et al. The effect of chemotherapy on the morphology of the growth plate and metaphysis of the growing skeleton. Eur J Surg Oncol. 2003;29(1):49–58.
78. Viateau V Guillemin G. Experimental animal models for tissue-engineered bone regeneration. In: Petite H, Quarto R, eds. Engineered Bone. Georgetown, TX: Eurekah.com/Landes Bioscience; 2005:89–91.
79. Viateau V, Guillemin G. Experimental animal models for tissue-engineered bone regeneration In: Petite H, Quarto R, eds. Engineered Bone. Georgetown, TX: Eurekah.com/Landes Bioscience; 2005:94.
80. Vigorita VJ, Ghelman B, Mintz D. Orthopaedic Pathology. Philadelphia, PA: Lippincott Williams &Wilkins; 2008.
81. Wang X, Mabrey JD, Agrawal CM. An interspecies comparison of bone fracture properties. Biomed Mater Eng. 1998;8(1):1–9.
82. Wei X, Gao J, Messner K. Maturation-dependent repair of untreated osteochondral defects in the rabbit knee joint. J Biomed Mater Res. 1997;34(1):63–72.
83. Yamasaki K. Histologic study of the femoral growth plate in beagle dogs. Toxicol Pathol. 1995;23(5):612–616.
84. Zoetis T, Tassinari MS, Bagi C, et al. Species comparison of postnatal bone growth and development. Birth Defects Res B Dev Reprod Toxicol. 2003;68(2):86–110.

Cite article

Cite article

Cite article

OR

Download to reference manager

If you have citation software installed, you can download article citation data to the citation manager of your choice

Share options

Share

Share this article

Share with email
EMAIL ARTICLE LINK
Share on social media

Share access to this article

Sharing links are not relevant where the article is open access and not available if you do not have a subscription.

For more information view the Sage Journals article sharing page.

Information, rights and permissions

Information

Published In

Article first published online: July 10, 2015
Issue published: September 2015

Keywords

  1. animal models
  2. biocompatibility
  3. bone implants
  4. bone remodeling
  5. dental implants
  6. medical device
  7. peri-implantitis
  8. safety assessment

Rights and permissions

© The Author(s) 2015.
Request permissions for this article.
PubMed: 26163303

Authors

Affiliations

L. M. Wancket
Biocompatibility, NAMSA, Northwood, OH, USA

Notes

L. M. Wancket, Biocompatibility, NAMSA, 6750 Wales Road, Northwood, OH 43619, USA. Email: [email protected]

Metrics and citations

Metrics

Journals metrics

This article was published in Veterinary Pathology.

VIEW ALL JOURNAL METRICS

Article usage*

Total views and downloads: 15947

*Article usage tracking started in December 2016


Altmetric

See the impact this article is making through the number of times it’s been read, and the Altmetric Score.
Learn more about the Altmetric Scores



Articles citing this one

Receive email alerts when this article is cited

Web of Science: 133 view articles Opens in new tab

Crossref: 142

  1. Effects of immediate loading directionality on the mechanical sensing ...
    Go to citation Crossref Google Scholar
  2. Calcium hydroxide nanoparticles synthesized with medicinal plants extr...
    Go to citation Crossref Google Scholar
  3. Comparative Evaluation of Osseodensification vs Conventional Osteotomy...
    Go to citation Crossref Google Scholar
  4. Regeneration of alveolar bone defects in the experimental pig model: A...
    Go to citation Crossref Google Scholar
  5. Treatment with PB125® Increases Femoral Long Bone Strength in 15-Month...
    Go to citation Crossref Google Scholar
  6. Histomorphometric and CBCT comparison of osseointegration around ortho...
    Go to citation Crossref Google Scholar
  7. Gathering Evidence to Leverage Musculoskeletal Magnetic Stimulation To...
    Go to citation Crossref Google Scholar
  8. Exploring rabbit as a nonrodent species for general toxicology studies
    Go to citation Crossref Google Scholar
  9. Artificial Intelligence-Based 3D Printing Strategies for Bone Scaffold...
    Go to citation Crossref Google Scholar
  10. Mechanistic insights into bone remodelling dysregulation by human vira...
    Go to citation Crossref Google Scholar
  11. Sintered fluorapatite scaffolds as an autograft‐like engineered bone g...
    Go to citation Crossref Google Scholar
  12. Swine models in translational research and medicine
    Go to citation Crossref Google Scholar
  13. Nanoscaled Titanium Oxide Layer Provokes Quick Osseointegration on 3D-...
    Go to citation Crossref Google Scholar
  14. Polymer nanocomposites in biomedical implants
    Go to citation Crossref Google Scholar
  15. Contrast enhanced computed tomography of small ruminants: Caprine and ...
    Go to citation Crossref Google Scholar
  16. Effect of systemic antiresorptive medication on the histopathological ...
    Go to citation Crossref Google Scholar
  17. Intramedullary bone tissue reaction of ion-releasing resin-modified gl...
    Go to citation Crossref Google Scholar
  18. Osteogenic differentiation and reconstruction of mandible defects usin...
    Go to citation Crossref Google Scholar
  19. The Suitability of Trabecular Patterns in the Assessment of Dental Imp...
    Go to citation Crossref Google Scholar
  20. Investigation of Bone Regeneration Efficacy of New Bovine Bone Mineral...
    Go to citation Crossref Google Scholar
  21. Three-Dimensional Printing of Poly(3-hydroxybutyrate-co-3-hydroxyvaler...
    Go to citation Crossref Google Scholar
  22. Degradation and Bone-Contact Biocompatibility of Two Drillable Magnesi...
    Go to citation Crossref Google Scholar
  23. Osseointegration of Tantalum Trabecular Metal in Titanium Dental Impla...
    Go to citation Crossref Google Scholar
  24. Trivalent rare earth elements substitution in 58S bioactive glass matr...
    Go to citation Crossref Google Scholar
  25. Effect of Splinting on Orthodontic Mini-Implant Tipping and Bone Histo...
    Go to citation Crossref Google Scholar
  26. Preclinical Evaluation of Bioactive Scaffolds for the Treatment of Man...
    Go to citation Crossref Google Scholar
  27. In‐vivo evaluations of bone regenerative potential of two novel bioact...
    Go to citation Crossref Google Scholar
  28. Regenerative Potential of Hydroxyapatite-Based Ceramic Biomaterial on ...
    Go to citation Crossref Google Scholar
  29. Local Antibiotic Delivery Ceramic Bone Substitutes for the Treatment o...
    Go to citation Crossref Google Scholar
  30. The Influence of Storage in Saline or Irradiation by Ultraviolet on Su...
    Go to citation Crossref Google Scholar
  31. Preclinical model for lumbar interbody fusion in small ruminants: Rati...
    Go to citation Crossref Google Scholar
  32. Advances in experimental models of rheumatoid arthritis
    Go to citation Crossref Google Scholar
  33. Preclinical Studies Design and Place of Rabbits
    Go to citation Crossref Google Scholar
  34. Preclinical Studies Design and Place of Ovine and Caprine
    Go to citation Crossref Google Scholar
  35. Preclinical Studies Design and Place of Rodents
    Go to citation Crossref Google Scholar
  36. Translational Aspects in Living Mammalian Organisms
    Go to citation Crossref Google Scholar
  37. Biomechanical duality of fracture healing captured using virtual mecha...
    Go to citation Crossref Google Scholar
  38. 3D-printed porous Ti6Al4V scaffolds for long bone repair in animal mod...
    Go to citation Crossref Google Scholar
  39. Therapeutic effect of intra-articular injected 3′-sialyllactose on a m...
    Go to citation Crossref Google Scholar
  40. Antimicrobial Biomaterials for the Healing of Infected Bone Tissue: A ...
    Go to citation Crossref Google Scholar
  41. Biocompatibility and degradation comparisons of four biodegradable cop...
    Go to citation Crossref Google Scholar
  42. Black phosphorus biomaterials for photo-controlled bone tissue enginee...
    Go to citation Crossref Google Scholar
  43. The impact of medication on osseointegration and implant anchorage in ...
    Go to citation Crossref Google Scholar
  44. Effectiveness of biomolecule-based bioactive surfaces, on os-seointegr...
    Go to citation Crossref Google Scholar
  45. Characterization of bone remodeling of two different porous types of β...
    Go to citation Crossref Google Scholar
  46. A review of bioceramics scaffolds for bone defects in different types ...
    Go to citation Crossref Google Scholar
  47. Biodegradable interbody cages for lumbar spine fusion: Current concept...
    Go to citation Crossref Google Scholar
  48. Zirconia versus Titanium Implants: 8-Year Follow-Up in a Patient Cohor...
    Go to citation Crossref Google Scholar
  49. NiTi laser textured implants with improved in vivo osseointegration: A...
    Go to citation Crossref Google Scholar
  50. Osseointegration Assessment of Multi‐Material Ti6Al4V‐β TCP Implants: ...
    Go to citation Crossref Google Scholar
  51. Selection of animal bone surrogate samples for orthopaedic screw testi...
    Go to citation Crossref Google Scholar
  52. Bone Regeneration Exploiting Corticoperiosteal Tissue Transfer for Sca...
    Go to citation Crossref Google Scholar
  53. Axillary ultrasound-guided approach for the brachial plexus in pig cad...
    Go to citation Crossref Google ScholarPub Med
  54. 3D-Printed Hydroxyapatite and Tricalcium Phosphates-Based Scaffolds fo...
    Go to citation Crossref Google Scholar
  55. A Biofabrication Strategy for a Custom-Shaped, Non-Synthetic Bone Graf...
    Go to citation Crossref Google Scholar
  56. Bone regeneration capacity of newly developed spherical magnesium phos...
    Go to citation Crossref Google Scholar
  57. Multi-walled carbon nanotube/hydroxyapatite nanocomposite with leukocy...
    Go to citation Crossref Google Scholar
  58. Tapping basement membrane motifs: Oral junctional epithelium for surfa...
    Go to citation Crossref Google Scholar
  59. Bilateral double site (calvarial and mandibular) critical-size bone de...
    Go to citation Crossref Google Scholar
  60. A Review of In Vivo and Clinical Studies Applying Scaffolds and Cell S...
    Go to citation Crossref Google Scholar
  61. A systematic review and meta‐analyses on animal models used in bone ad...
    Go to citation Crossref Google Scholar
  62. A Large Animal Model for Orthopedic Foot and Ankle Research
    Go to citation Crossref Google Scholar
  63. Clinically relevant preclinical animal models for testing novel cranio...
    Go to citation Crossref Google Scholar
  64. The Effect of Bone Marrow Stimulation for Cartilage Repair on the Subc...
    Go to citation Crossref Google ScholarPub Med
  65. In Vitro and In Vivo Models to Understand Biofilm Implant Infections
    Go to citation Crossref Google Scholar
  66. Animal Models in Toxicologic Research: Rabbit
    Go to citation Crossref Google Scholar
  67. Biomechanical and histomorphometric analysis of endosteal implants pla...
    Go to citation Crossref Google Scholar
  68. Host responses to implants revealed by intravital microscopy
    Go to citation Crossref Google Scholar
  69. Osteocytes Influence on Bone Matrix Integrity Affects Biomechanical Co...
    Go to citation Crossref Google Scholar
  70. Recent Advances in the Evaluation of Antimicrobial Materials for Resol...
    Go to citation Crossref Google Scholar
  71. The osseointegration and stability of dental implants with different s...
    Go to citation Crossref Google Scholar
  72. Evaluation of biofilm colonization on multi-part dental implants in a ...
    Go to citation Crossref Google Scholar
  73. A novel surgical model for the preclinical assessment of the osseointe...
    Go to citation Crossref Google Scholar
  74. A guide to reducing adverse outcomes in rabbit models of sciatic nerve...
    Go to citation Crossref Google Scholar
  75. Novel resin tissue array system reduces sample preparation time, labou...
    Go to citation Crossref Google Scholar
  76. Biodegradable metals for bone defect repair: A systematic review and m...
    Go to citation Crossref Google Scholar
  77. Pre-Clinical Models in Implant Dentistry: Past, Present, Future
    Go to citation Crossref Google Scholar
  78. Biomechanical Performance of BoneHelix® Compared with Elastic Stable I...
    Go to citation Crossref Google Scholar
  79. RNA ‐ seq Analysis of ...
    Go to citation Crossref Google Scholar
  80. 3D nanoscale analysis of bone healing around degrading Mg implants eva...
    Go to citation Crossref Google Scholar
  81. Nano-Hydroxyapatite as a Delivery System for Promoting Bone Regenerati...
    Go to citation Crossref Google Scholar
  82. In vivo evaluation of deer antler trabecular bone as a reconstruction ...
    Go to citation Crossref Google Scholar
  83. From tissue retrieval to electron tomography: nanoscale characterizati...
    Go to citation Crossref Google Scholar
  84. Comparative Evaluation of Two Glass Polyalkenoate Cements: An In Vivo ...
    Go to citation Crossref Google Scholar
  85. Bioactive coatings for dental implants: A review of alternative strate...
    Go to citation Crossref Google Scholar
  86. Animal models of vertical bone augmentation (Review)
    Go to citation Crossref Google Scholar
  87. Effects of low‐dose rhBMP‐2 on peri‐implant ridge augmentation in a ca...
    Go to citation Crossref Google Scholar
  88. Temporal Changes in Reverse Torque of Locking-Head Screws Used in the ...
    Go to citation Crossref Google Scholar
  89. Implant‐associated osteomyelitis: Development, characterisation, and a...
    Go to citation Crossref Google Scholar
  90. Systematic Review of the Preclinical Technology Readiness of Orthopedi...
    Go to citation Crossref Google Scholar
  91. Additive manufacturing pertaining to bone: Hopes, reality and future c...
    Go to citation Crossref Google Scholar
  92. Enhanced Osseointegration by the Hierarchical Micro-Nano Topography on...
    Go to citation Crossref Google Scholar
  93. Essentials of Swine Housing
    Go to citation Crossref Google Scholar
  94. Biological Assessment of Bioceramics: In Vitro and In Vivo Tests
    Go to citation Crossref Google Scholar
  95. Biocompatibility and cytotoxicity evaluation of isophorone diisocyanat...
    Go to citation Crossref Google Scholar
  96. GDF-5 variant loading on composite scaffolds promotes spinal fusion th...
    Go to citation Crossref Google Scholar
  97. International Harmonization of Nomenclature and Diagnostic Criteria (I...
    Go to citation Crossref Google Scholar
  98. Análisis biomecánico de dos constructos de osteosíntesis en fracturas ...
    Go to citation Crossref Google Scholar
  99. A novel cryo-embedding method for in-depth analysis of craniofacial mi...
    Go to citation Crossref Google Scholar
  100. A radiological study of bone remodeling with two different types of po...
    Go to citation Crossref Google Scholar
  101. Is maxillary diastema an appropriate site for implantation in rats?
    Go to citation Crossref Google Scholar
  102. Interspecies comparison of alveolar bone biology: Tooth extraction soc...
    Go to citation Crossref Google Scholar
  103. Biological and mechanical performance and degradation characteristics ...
    Go to citation Crossref Google Scholar
  104. Effects of the local administration of antibiotics on bone formation o...
    Go to citation Crossref Google Scholar
  105. Bone Healing Evaluation Following Different Osteotomic Techniques in A...
    Go to citation Crossref Google Scholar
  106. Articular Repair/Regeneration in Healthy and Inflammatory Conditions: ...
    Go to citation Crossref Google Scholar
  107. Biological responses to physicochemical properties of biomaterial surf...
    Go to citation Crossref Google Scholar
  108. Technologies to Enhance Spinal Fusion: Bench to Bedside
    Go to citation Crossref Google ScholarPub Med
  109. Systematic scoping review of mandibular bone tissue engineering
    Go to citation Crossref Google Scholar
  110. Chicken proximal interphalangeal joint as an animal model for human fi...
    Go to citation Crossref Google Scholar
  111. MagnetOs, Vitoss, and Novabone in a Multi-endpoint Study of Posterolat...
    Go to citation Crossref Google Scholar
  112. Significant Loss of ACL Graft Force With Tibial-Sided Soft Tissue Inte...
    Go to citation Crossref Google ScholarPub Med
  113. An In Vivo Study in Rat Femurs of Bioactive Silicate Coatings on Titan...
    Go to citation Crossref Google Scholar
  114. Calcium Phosphate Carrying Simvastatin Enhances Bone Regeneration: A S...
    Go to citation Crossref Google Scholar
  115. The Impact of Bioceramic Scaffolds on Bone Regeneration in Preclinical...
    Go to citation Crossref Google Scholar
  116. Biodegradable Magnesium‐Based Implants in Orthopedics—A General Review...
    Go to citation Crossref Google Scholar
  117. A preclinical large-animal model for the assessment of critical-size l...
    Go to citation Crossref Google Scholar
  118. Postoperative Administration of Alpha-tocopherol Enhances Osseointegra...
    Go to citation Crossref Google Scholar
  119. Animal Surgery and Care of Animals
    Go to citation Crossref Google Scholar
  120. Overview of Tissue Engineering Concepts and Applications
    Go to citation Crossref Google Scholar
  121. Advances in Exotic Animal Osteosynthesis
    Go to citation Crossref Google Scholar
  122. Bioactive Glass and Silicate-Based Ceramic Coatings on Metallic Implan...
    Go to citation Crossref Google Scholar
  123. Development and Bone Regeneration Capacity of Premixed Magnesium Phosp...
    Go to citation Crossref Google Scholar
  124. Gross osteology and radiology of the pelvic limb of the adult small Ea...
    Go to citation Crossref Google Scholar
  125. An Overview on the Considerations for the Planning of Nonclinical Necr...
    Go to citation Crossref Google ScholarPub Med
  126. Animal models of orthopaedic infections. A review of rabbit models use...
    Go to citation Crossref Google Scholar
  127. Relative effects of age on implant integration in a rat model: A longi...
    Go to citation Crossref Google Scholar
  128. The Role of Strontium Enriched Hydroxyapatite and Tricalcium Phosphate...
    Go to citation Crossref Google Scholar
  129. Investigating the Mechanical Characteristics of Bone-Metal Implant Int...
    Go to citation Crossref Google Scholar
  130. Investigating the Osteoinductive Potential of a Decellularized Xenogra...
    Go to citation Crossref Google Scholar
  131. Bone regeneration strategies: Engineered scaffolds, bioactive molecule...
    Go to citation Crossref Google Scholar
  132. Finite Element Study of a Threaded Fastening: The Case of Surgical Scr...
    Go to citation Crossref Google Scholar
  133. Blood Prefabrication Subcutaneous Small Animal Model for the Evaluatio...
    Go to citation Crossref Google Scholar
  134. Comparison of oscillometric, Doppler and invasive blood pressure measu...
    Go to citation Crossref Google Scholar
  135. New bone formation and trabecular bone microarchitecture of highly por...
    Go to citation Crossref Google Scholar
  136. A single intraperitoneal injection of bovine fetuin-A attenuates bone ...
    Go to citation Crossref Google Scholar
  137. Bone and Joint Medical Devices: Methods, Models, and Regulations
    Go to citation Crossref Google ScholarPub Med
  138. Periodontoloji Alaninda Hayvan Çalışmaları: Deneysel Periodontal ve Pe...
    Go to citation Crossref Google Scholar
  139. Platelet-rich plasma for regeneration of neural feedback pathways arou...
    Go to citation Crossref Google Scholar
  140. Establishment of a preclinical ovine screening model for the investiga...
    Go to citation Crossref Google Scholar
  141. No Bones About It: The Challenges and Rewards of Osteopathology
    Go to citation Crossref Google ScholarPub Med

Figures and tables

Figures & Media

Tables

View Options

View options

PDF/ePub

View PDF/ePub

Get access

Access options

If you have access to journal content via a personal subscription, university, library, employer or society, select from the options below:


Alternatively, view purchase options below:

Purchase 24 hour online access to view and download content.

Access journal content via a DeepDyve subscription or find out more about this option.