Skip to main content
Erschienen in: BMC Musculoskeletal Disorders 1/2016

Open Access 01.12.2016 | Research article

Clinical trial and in-vitro study comparing the efficacy of treating bony lesions with allografts versus synthetic or highly-processed xenogeneic bone grafts

verfasst von: Eva Johanna Kubosch, Anke Bernstein, Laura Wolf, Tobias Fretwurst, Katja Nelson, Hagen Schmal

Erschienen in: BMC Musculoskeletal Disorders | Ausgabe 1/2016

Abstract

Background

Our study aim was to compare allogeneic cancellous bone (ACB) and synthetic or highly-processed xenogeneic bone substitutes (SBS) in the treatment of skeletal defects in orthopedic surgery.

Methods

232 patients treated for bony lesions with ACB (n = 116) or SBS (n = 116) within a 10-year time period were included in this case–control study. Furthermore, both materials were seeded with human osteoblasts (hOB, n = 10) and analyzed by histology, for viability (AlamarBlue®) and protein expression activity (Luminex®).

Results

The complication rate was 14.2 %, proportion of defects without bony healing 3.6 %; neither outcome parameter differed comparing the intervention groups. Failed consolidation correlated with an increase in complications (p < 0.03). The rate of complications was further highly significant in association with the location of use (p < 0.001), but did not depend on age, ASA risk classification, BMI, smoking behavior or type of insurance. However, those factors did significantly influence the bony healing rate (p < 0.02). Complication and consolidation rates were independent of gender and the filling substances employed within the different locations. Histological examination revealed similar bone structures, whereas cell remnants were apparent only in the allografts. Both materials were biocompatible in-vitro, and seeded with human osteoblasts. The cells remained vital over the 3-week culture period and produced microscopically typical bone matrix. We observed initially increased expression of osteocalcin, osteopontin, and osteoprotegerin as well as leptin and adiponectin secretion declining after 1 week, especially in the ACB group.

Conclusion

Although both investigated materials appeared to be similarly suitable for the treatment of skeletal lesions in-vivo and in-vitro, outcome was decisively influenced by other factors such as the site of use or epidemiological parameters.
Hinweise
An erratum to this article can be found at http://​dx.​doi.​org/​10.​1186/​s12891-016-0976-0.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

EJK was involved in the conception and the design of the study, the analysis and interpretation of the data, writing of the article, and the final approval of the submitted article. AB supported the lab work, supervised the sample analysis, was responsible for sample storage, revised the article draft critically, and approved the final version of the article. LW was responsible for the collection, assembly and management of data, performed the experiments, contributed to the article draft, calculated the scores and the descriptive statistics, and approved the final version of the article. TF has done the basic histology of the in-vitro-analyzed bone substitutes, contributed to the article draft, and approved the final article. KN was involved in the conception of the study, supervised histology examinations, critically revised the article draft, and approved the final version. HS was responsible for the conception and the design of the study, for obtaining of funding, the analysis and interpretation of the data, tutorial of Laura Wolf, drafting of the article, and the final approval of the submitted article. All authors take responsibility for the integrity of the work as a whole, from inception to finished article.

Background

The treatment of large bone defects caused by trauma, degenerative or congenital diseases and tumor lesions is one of the greatest challenges in current orthopedic research, making the development of effective bone regeneration therapies a major topic. Although the amount of available autogenous cancellous bone is naturally limited, and graft harvesting from the iliac crest leads to significant donor-site morbidity [1], autogenous bone grafts combining osteogenic, osteoconductive and osteoinductive properties must be considered the gold standard for bone replacement for now [2]. The osseous regeneration capacity is limited in older patients especially, and donor site morbidity increases [3]. The known disadvantages of autologous bone grafting have prompted the search for alternatives. The long-term clinical goal is to regenerate an adequate amount of bony tissue with anatomical, physiological, three-dimensional morphology. In addition to bone applications, current bone regeneration strategies include cell-based or stem cell-based treatments, the application of bioactive factors such as BMP-2 and BMP-7, different biologic or artificial scaffolds and various combinations [2]. However, current and emerging therapies are still significantly restricted. Several artificial bone materials have recently been tested. Artificial bone grafts, most based on tricalcium phosphate (TCP) or hydroxyapatite (components of natural bone) usually only possess osteoconductive properties enabling adjacent osteoblasts to migrate [4]. Synthetic bone grafts are an alternative to autologous and allogeneic graft options, given their wide availability, comparatively low cost, slow biodegradation, superior strength in compression and the absence of risks such as donor-site morbidity and viral transmission. However, because they are strictly osteoconductive, their biologic role is limited, especially in fracture healing [5].
In the early 2000s more than 500,000 bone grafting procedures per year were carried out in the United States and 2.2 million worldwide in order to augment bone defects in orthopedics or other fields as dentistry [6]. Allogenous bone grafts are harvested from humans or human cadavers, undergo sterile processing and are transplanted to a recipient. Depending on the preparation process, allografts exhibit osteoconductive and sometimes osteoinductive potential. Allogeneic transplants can be infectious [7, 8], may trigger immunological reactions [911], and the sterilization process prior to transplantation leads to the loss of osteoinductive properties [1214]. Recent studies by Ghanaati et al. [11] and Fretwurst et al. [15] revealed the presence of organic/cellular remnants in allografts and xenogeneic bone blocks commercially available for dental and orthopedic surgery that might induce an immune response. As freezing alone is insufficient in safety terms [16], terminal sterilization, usually irradiation, is carried out as well. However, the incorporation of allografts is reduced to approximately 40 % after irradiation [17, 18], compared to 80–100 % in non-irradiated grafts [1921]. The freezing process also delays the vascularization of allografts after implantation [22], which might be detrimental, as vascularity has been identified as a central component influencing bone healing and necessary to effective graft repair [2]. The limited osteoinductive capacity of allografts presumably plays a fundamental role in allograft failure due to fracture or nonunion [14].
In contrast to adult hyaline cartilage, which does not restore its native structure once damaged, the human skeleton has a remarkable ability to regenerate after injury. Nevertheless, the conditions for spontaneous bone healing are not always ideal. Substantial bone loss in case of trauma or tumor lesions often requires bone-graft augmentation. Evidence of the effectiveness of graft incorporation and fracture healing remains elusive [23, 24]. Most of the literature addresses medium and long-term outcomes after revision arthroplasty [25]. In terms of bone grafting in fracture therapy, the latest data merely focus on certain anatomical locations [24, 26].
The aim of this study was to compare and determine the clinical effectiveness of allogeneic cancellous bone grafts (ACB) and synthetic or highly processed xenogeneic bone substitutes (SBS) for treating bony defects in different sites considering epidemiological and custom-designed parameters. Our rationale for comparing these two groups was the substantial differences described above, including the presumed presence of cell remnants in ACB in contrast to SBS. Nevertheless, we hypothesized that there would be no difference between ACB and SBS in their effectiveness in bone regeneration or consolidation. To clarify possible differences regarding biocompatibility and osteoconductivity, in-vitro testing was additionally carried out. Our hypothesis regarding the in-vitro experiments was that it is possible to predict in-vivo differences.

Methods

The study is divided into a clinical and an experimental part.

Patients

Within one decade, from 2001 to 2011, we identified 232 patients treated with ACB or SBS (117 female, 115 male; mean age 59 ± 18 years) based on the documented OPS code. The following SBS were used: BioOss® (Geistlich, Wolhusen, Switzerland) in 41.4 %, and at clearly lower percentages, respectively: Norian SRS® (Synthes) (8.6 %), Chronos® (Synthes) (1.7 %), Atlantik® (Argomedical) (2.6 %), Alaska® (Argomedical) (0.9 %), Endobone® (Biomet) (4.3 %), Pyrost® (Stryker) (0.9 %), Nanostim® (Medtronic) (37.1 %), Actifuse® (Baxter) (0.9 %), Tutobone® (Novomedics) (0.9 %) and PerOssal® (Botiss) (0.9 %). The SBS group thus contained various xenogenic or synthetic, commercially available bone substitutes, which, according to the manufacturers’ information, had undergone multiple purification processes and were supposed to be free of organic residues. Commercially-available human allografts, freeze-dried cancellous bone from femoral heads and freeze-dried cancellous cubes or blocks (DIZG, German Institute for Cell and Tissue Replacement, Berlin, Germany) were used in the ACB group. The study’s end points were defined as consolidation or treatment failure. We also determined the occurrence of complications. Similar to trials following the German Pharmaceuticals Act regulations, all complications were documented as adverse events. We investigated the relationship between a given complication and the use of a particular bone-graft material in both groups. When in doubt, we presumed a correlation between the complication and the use of bone graft, which was then documented as an adverse event. Basically, complications were defined as the need for surgical revision/re-operation or a patient’s death. Documented complications were: death, re-operation, infection/wound infection, seroma, pseudarthrosis, re-fracture, plate or screw fracture or loosening, secondary dislocation, loss of reduction, necrosis of the humeral head, cup/implant loosening, nerve palsy and cyst recurrence. We ultimately enrolled 116 patients treated with allografts and 116 patients treated with synthetic or highly-processed bone grafts in this study.
To ensure better comparability of the various bone-graft materials used, we defined influencing factors: besides gender and age, clinical outcome was assessed in regard to defect location, defect size, reason and type of application (fracture or bone defect for other reasons (e.g., bone cysts, osteochondritis dissecans, tumor); ACB or SBS), time to consolidation, duration of hospital stay, kind of insurance, BMI (body mass index), ASA (American Society of Anesthesiologists) risk classification, and smoking habits.
The study was approved by the Ethics Committee of the University of Freiburg (registration number 10010/15). All included individuals have declared their informed consent for the retrospective analysis of their treatment data.

Experimental setup

To test their biocompatibility, the most frequently used representatives of ACB and SBS were seeded with human osteoblasts and cell viability and metabolic activity were analyzed. The cells’ preparation protocols were approved by the Ethics Committee of the University of Freiburg as part of the ‘Tissue bank for research in the field of tissue engineering’ project (GTE-2002) and the biobank ‘Osteo’ (AN-EK-FRBRG-135/14). Cancellous bone (DIZG, Berlin, Germany) and BioOss®-Granula (Geistlich, Wolhusen, Switzerland) in size of 3–4 mm were therefore used. BioOss® (Geistlich, Wolhusen, Switzerland) is a natural, nanocrystalline, carbonated hydroxyapatite of bovine origin. A patented multi-stage purification process prior to gamma-sterilization should remove proteins and de-activate viruses and other pathogens. The inorganic bone matrix with its macro- and microporous structure provides osteoconductive properties similar to human cancellous bone. We used BioOss® in further in-vitro experiments in the SBS group because of its frequent in-vivo application. ACB and BioOss® were examined for any cells or cellular remnants prior to colonization with hOB by histology.

Histologic processing

Histologic processing was done as described [15]. The specimen was dehydrated in 100 % ethanol and subsequently infiltrated, embedded and polymerized in Technovit 9100 (Heraeus Kulzer, Wehrheim, Germany) according to the manufacturer’s instructions. After the polymerization process, samples were cut into 500 μm sections using a rotary diamond saw Secotom- 50 (Stuers, Ballerup, Denmark). The sections were mounted onto opac acrylic-slides (Maertin, Freiburg, Germany) and ground to a final thickness of approximately 60 μm on a rotating grinding plate (Stuers, Ballerup, Denmark). All specimens were stained with azure II and pararosaniline (Merck, Darmstadt, Germany).
Slides were imaged with an Axio Imager M1 microscope equipped with a digital AxioCam HRc (Carl Zeiss, Göttingen, Germany). The histologic sections were analyzed via analySIS FIVE – software (Soft Imaging System, Münster, Germany).

Isolation of osteoblasts

Femoral heads were obtained during hip arthroplasty operations following femoral neck fractures. The material used was taken following informed consent from patients in accordance with the Ethics Committee of the University of Freiburg as part of the ‘Tissue bank for research in the field of tissue engineering’ project (GTE-2002) and the biobank ‘Osteo’ (AN-EK-FRBRG-135/14). The degree of osteoarthritis was evaluated on X-rays using Croft’s modification of the Kellgren and Lawrence grading system. Cells from patients with a Kellgren and Lawrence Score ≤2 were used in our experiments. Post-surgery human primary osteoblasts (hOB) were isolated within 8 h from cancellous bone by cell outgrowth from small pieces, as described [27]. Human OBs were cultured and expanded in medium 199 (Gibco, Carlsbad, CA), supplemented with 10 % FCS and 1 % penicillin-streptomycin at 37 °C and 5 % CO2. Human OBs from passage 1 or 2 were used in all further experiments.

Cell seeding

Prior to application, we assessed cell viability, cytotoxicity and apoptosis events as well as specific osteogenic characteristics using ApoTox-Glo™ Triplex and AttoPhos® Assays (Promega, Madison, WI, USA) showing vital (>90 %) and typical osteoblasts. Similar amounts of BioOss® and allogeneic cancellous bone (20–25 mg) were weighed into 50 ml falcons and moistened with medium 199, respectively. Medium was discarded; bone grafts were seeded per falcon at a density of 1 × 106 cells in 3 ml medium 199 and centrifuged at 1,200 rpm for 5 min. Cells were cultivated for 72 h at 37 °C and 5 % CO2 and then used in the experiments.

AlamarBlue® - assay

The AlamarBlue®-Assay (AbD Serotec, Oxford, UK) is a quantitative test to measure the cell proliferation and viability of various human and animal cell lines, bacteria, plant and fungi. When cells are alive, they maintain a reducing environment within the cell’s cytosol. Resazurin, the active ingredient in AlamarBlue® reagent, is a non-toxic, cell permeable compound blue in color and highly fluorescent. Upon entering cells, resazurin is reduced to resorufin, a red compound that is highly fluorescent. Viable cells continuously convert resazurin to resorufin, increasing the overall fluorescence and color of the media surrounding cells [28].
Cells were cultivated for 1 week after isolation, followed by incubation with medium 199 containing 10 % AlamarBlue® for another 24 h. Cells were centrifuged for 5 min at 1,200 rpm, and supernatant collected to assess the resulting fluorescence. Cell-seeded constructs were washed with PBS, centrifuged and cultivated further in fresh medium. Supernatants were aliquoted into microtiter plates and the absorbance signal at 570 and 600 nm was detected on an ELISA plate reader. The same assay was repeated after 3 weeks’ cultivation.

Milliplex® Human Bone Panel 1B

Milliplex® Human Bone Panel 1B Kit (Millipore AG, Zug, Switzerland) is a magnetic bead-based antibody microarray founded upon the sandwich immunoassay principle. The assay aims to quantify biomarkers that play a role in bone metabolism. We investigated the following analytes: osteocalcin (OC), osteopontin (OPN), osteoprotegerin (OPG), leptin and adiponectin. The assay was performed according to the manufacturers’ guidelines. Briefly, color-code microspheres are coated with a specific capture antibody. After a test sample’s analyte is captured by the bead, a biotinylated detection antibody is introduced and the reaction mixture is then incubated with streptavidin-PE conjugate, the reporter molecule, to complete the reaction on each microsphere’s surface. The Luminex®-100 reader is a system based on flow-cytometry principles. The reader identifies the bead’s color-code as well as the bead surface’s PE-fluorescence [29]. Analysis was performed with the following adjustments: 50 μl probe, 50 beads per set, gate 8,000–15,000, RP1 calibrator low PMT, time out 60 s. Protein was analyzed with the analytical software provided by the manufacturers.

Scanning electron microscopy (SEM)

Scanning electron microscopy was performed after 1 and after 3 weeks of cultivation to observe the cell loading and growth patterns of hOBs on ACB and BioOss®. The cell-seeded grafts were centrifuged, supernatant discarded, and samples washed twice with PBS-buffer and fixed with fixation buffer overnight. Specimens were then dehydrated by rinsing them in an ascending ethanol-water mixture (30, 50, 70, 80, 90, 99 %; 2 × 5 min at RT for each step) and then rinsed in an ethanol-hexamethyldisalazin mixture (EtOH:HMDS 2:1, 1:1, 1:2, HMDS; 2x5 min at RT for each step). Finally, the samples were dried, sputtered with a thin layer of gold (SC7620 Mini Sputter Coater, Gala Instrumente, Bad Schwalbach, Germany), and examined under a Phenom scanning electron microscope (PHENOM Pure Desktop SEM, Phenom-World BV, Eindhoven, Netherlands).

Statistical analysis

All values are expressed as mean ± standard deviation. Normality was tested by the Kolmogorov-Smirnov test. The paired t-Test was then used to assess differences among normally-distributed mean values. In case of heterogeneous variances, the Aspin-Welch Test was applied. Individual group mean scores were compared with the Mann–Whitney U test. The statistical comparison of incidences was done using the chi-square test. p < 0.05 was considered significant in all tests. Statistical analysis was carried out by the Statistical Package for the Social Sciences (SPSS) software version 21 (SPSS Inc., Chicago, USA).

Results

Characterization of patients treated with allografts

116 patients were treated with ACB. The gender ratio was female/male (f/m) 62/54. The defect sites were distributed as: humerus 10 patients, forearm 4 patients, hip/pelvis 48 patients, femur 27 patients, tibia 10 patients, foot 10 patients, spine 1 patient (Fig. 1). 45 patients were treated for a fracture involving the loss of bone substance, 72 patients had a bony defect caused by other reasons such as tumor or cyst lesions requiring surgery. We observed bony consolidation of the defect in 104 patients, treatment failure in 6 patients, 6 patients were dropouts. The average time to consolidation was 3.93 ± 2.45 months; defect size averaged 20.21 ± 21.00 cm2. Mean age in this group was 62.6 ± 19.6 at the time of surgery, mean BMI was 26.01 ± 5.15 kg/m2. 23 patients were current smokers, the mean ASA risk classification was 2.31 ± 0.72 (Table 1). Complications occurred in 17 cases (14.7 %) (Table 2), 5 patients passed away during the follow-up period. Duration of hospital stay averaged 17.85 days. 90 patients had compulsory health insurance (C), 21 private health insurance (P) and 6 patients were covered by an employer’s mutual insurance association (E).
Table 1
Epidemiological and characterizing parameters
 
Allogenic cancellous bone (ACB)
N
Synthetic bone substitutes (SBS)
N
P
Age
62.6 ± 19.6
116
55.8 ± 16.3
116
0.004a
Gender (f/m)
62/54
116
55/61
116
n.s.b
ASA
2.31 ± 0.72
116
1.95 ± 0.67
116
<0.0001c
Defect size
20.21 ± 21.00
114
5.75 ± 8.33
115
<0.0001a
BMI
26.01 ± 5.15
116
26.99 ± 5.60
116
n.s.a
Consolidation time (months)
3.93 ± 2.45
104
3.16 ± 3.31
107
n.s.d
Complication rate
17 (14.6 %)
116
16 (13.8 %)
116
n.s.b
Healing rate
104 (89.7 %)
116
107 (92.2 %)
116
n.s.b
Overview about the epidemiological and characterizing parameters comparing both intervention groups, f female, m male, ASA physical status according to the American Society of Anesthesiologists, BMI body mass index, n.s. – not significant, a t-Test, b χ 2-Test, c U-Test (Mann und Whitney), dAspin-Welch-Test
Table 2
Influence on complications
 
Complication
N
No complication
N
 
Age
63.52 ± 18.44
33
58.47 ± 18.24
199
n.s.a
Gender (f/m)
20/13
33
97/102
199
n.s.b
ASA
2.24 ± 0.66
33
2.11 ± 0.72
199
n.s.c
Defect size
10.98 ± 14.48
33
13.28 ± 17.95
196
n.s.a
BMI
26.79 ± 5.41
33
26.45 ± 5.39
199
n.s.a
Cancellous/Synthetic bone
17/16
33
100/99
199
n.s.b
Smoker/Non-Smoker
10/23
33
49/150
199
n.s.b
Insurance status W/C/P
23.5 %/13.4 %/12.2 %
32
76.5 %/86.6 %/87.8 %
198
n.s.b
Healing rate
7 (21.2 %)
33
1 (0.5 %)
186
<0.0001b
Consolidation time (months)
4.79 ± 4.39
26
3.36 ± 2.64
185
0.02a
Overview about the comparing parameters in patients with or without a complication, ffemale, m male, ASA physical status according to the American Society of Anesthesiologists, BMI body mass index, n.s. not significant. W employers mutual insurance association, C compulsory health insurance fund, P private health insurance fund. a t-Test, b χ 2-Test, c U-Test (Mann und Whitney)

Characterization of patients treated with synthetic and highly processed bone grafts

116 patients were treated with SBS. The f/m gender ratio was 55/61. 41.4 % of the patients (n = 48) were treated with BioOss® (Geistlich, Wolhusen, Switzerland). The defect sites were distributed as: humerus 9 patients, forearm 10 patients, pelvis 3 patients, femur 3 patients, tibia 32 patients, foot 8 patients, spine 44 patients (Fig. 1). Bony consolidation was observed in 107 patients, treatment failure in 2 patients and the healing state in 7 patients remained unknown. The average time to consolidation was 3.16 ± 3.31 months; defect size averaged 5.75 ± 8.33 cm2. 52 patients were treated for a fracture, 64 patients were suffering from bony defects for other reasons. This group’s mean age at the time of surgery in was 55.8 ± 16.3 years, mean BMI was 26.99 ± 5.60 kg/m2. 36 patients were current smokers, the mean ASA risk classification was 1.95 ± 0.67 (Table 1). Complications occurred in 16 (13.8 %) cases (Table 2), 1 patient passed away during follow-up. Mean duration of hospital stay was 14.22 days. 82 patients had compulsory health insurance (C), 19 private health insurance (P) and 11 patients were covered by an employer’s mutual insurance association (E). To summarize: both groups (4.1. and 4.2.) differed substantially in age, co-morbidities and defect size, as indicated in Table 1.

Clinical results

The complication rate reached overall 14.2 %; Table 2 compares the examined parameters in the groups with or without complication. The proportion of defects without bony healing was 3.6 %; Table 3 compares the examined parameters in the groups with or without healing. Failed consolidation correlated with an increase in complications (p < 0.03). The complication rate correlated very closely with the site of use (p < 0.0002) (Fig. 2), but not with age, gender, ASA risk classification, BMI, smoking behavior or type of insurance (Table 2). However, these factors significantly influenced the bony healing rate (p < 0.02) (Table 3). Complication and consolidation rates were independent of gender and the filling substances applied within the different sites. We detected no significant differences in the effectiveness of treating bony lesions with allografts versus synthetic or highly-processed xenogeneic bone grafts in various sites (Table 4). Higher complication rates were documented in association with the treatment of bony lesions in the humerus, tibia and the foot (Fig. 2). In the vicinity of the humerus we documented screw perforation, secondary loss of reduction, wound infection and osteonecrosis of the humeral head. Complications in the treatment of bony lesions of the proximal tibia were mainly secondary loss of fracture reduction, deep infections and wound infections. Wound infections were the main complication that occurred in the foot area. Considering the SBS group’s heterogeneity, further stratification was analyzed comparing complication and consolidation rates by discriminating between synthetic and highly-processed xenogeneic materials within the SBS-group in addition to the allografts (ACB) 116/61/55). Again, we found no statistically significant differences (p for chi square test >0.05 for all comparisons) in any endpoint (complications: ACB 17/99, synthetic 7/54, xenogeneic 9/46, consolidation failure: ACB 6/105, synthetic 0/58, xenogeneic 2/48, missing values 13).
Table 3
Influences on bone healing
 
Bony healing
N
No healing
N
P
Age
57.89 ± 18.01
211
72.51 ± 13.45
8
0.0025a
Gender (f/m)
102/109
211
6/2
8
n.s.b
ASA
2.09 ± 0.71
211
2.75 ± 0.46
8
0.014c
Defect size
12.90 ± 17.62
211
18.51 ± 20.54
8
n.s.a
BMI
26.42 ± 5.38
211
26.94 ± 4.19
8
n.s.a
Cancellous/Synthetic bone
104/107
211
6/2
8
n.s.b
Smoker/Non-Smoker
51/160
211
5/3
8
0.014b
Insurance status W/C/P
13/159/38
210
3/3/1
7
0.0012b
Overview about the comparing parameters in patients with or without bony consolidation, ffemale, m male, ASA physical status according to the American Society of Anesthesiologists, BMI body mass index, n.s. not significant, W employers mutual insurance association, C compulsory health insurance fund, P private health insurance fund. a t-Test, b χ 2-Test, c U-Test (Mann und Whitney)
Table 4
Bone healing and complications within different locations
 
Consolidation yes/no
N
P
Complication yes/no
N
P
 
Cancellous
Synthetic
Cancellous
Synthetic
Humerus
10/0
8/1
19
n.s.a
5/7
5/5
22
n.s.a
Forearm
4/0
10/0
14
n.s.a
0/4
1/10
15
n.s.a
Femur
24/3
3/0
30
n.s.a
5/22
0/3
30
n.s.a
Tibia
9/1
31/1
42
n.s.a
3/7
5/29
44
n.s.a
Spine
0/1
44/0
45
n.s.a
1/0
2/45
48
n.s.a
Pelvis
47/1
3/0
51
n.s.a
2/50
0/3
55
n.s.a
Foot
10/0
8/0
18
n.s.a
1/9
3/5
18
n.s.a
Number of complications and consolidation rates dependent on the group of applied bone substitutes, n.s. not significant, a χ 2-Test

In-vitro results

Histologic findings

Osteocytes, adipocytes and fibrocytes were detected in ACB (Fig. 3a). No cells or cellular remnants were detectable on BioOss® prior to cell seeding with hOB. Osteocyte lacunae were empty (Fig. 3b, arrows). In contrast to ACB, no soft tissue was detected.

Alamar Blue®- assay

Results were analyzed by plotting absorbance versus compound concentration, and we calculated the absolute reduction. This value correlates with cell number and metabolic activity. Human osteoblasts cultivated on BioOss® revealed a reduction in resazurin after 1 week in the AlamarBlue®-assay (53.1 ± 25.1 %); 41.8 ± 29.45 % after 3 weeks. Osteoblasts cultivated on cancellous bone (ACB) reduced resazurin by 35.2 ± 32.1 % after 1 week, and 13.3 ± 9.8 % after 3 weeks of total resazurin applied. Although the absolute resazurin reduction (cell number and metabolic activity) decreased over time especially in the allograft group (ACB), we detected no statistically significant differences between the two intervention groups and the two time points of cultivation (Fig. 4).

Milliplex Human Bone Panel 1B

We identified and quantified bone-specific biomarkers in the supernatant after 1 and 3 weeks of cultivation. Osteocalcin (OC), osteopontin (OPN), osteoprotegerin (OPG), leptin and adiponectin were detected and analyzed, revealing maintenance of the cells’ stable osteogenic phenotype independent of the biomaterial employed.
Osteocalcin, a marker of bone anabolism, was detected in all probes. Mean OC concentrations in cell culture supernatants of human osteoblasts (hOB) cultivated on BioOss® for 1 week were 160.2 ± 85.2 pg/ml; 62.9 ± 4.6 pg/ml in the group of cancellous bone. Concentrations fell after 3 weeks in both groups to 43.7 ± 6.1 pg/ml (BioOss®) and 34.5 ± 3.7 pg/ml (ACB), respectively. The ACB group’s concentrations decreased significantly from 1 to 3 weeks (p = 0.002) (Fig. 5a, but there was no statistically significant difference between the groups analyzing the respective time points.
Mean concentrations of osteopontin were 1,514.2 ± 883.9 pg/ml after 1 week in supernatants of osteoblasts cultivated on BioOss®. Concentrations decreased after 3 weeks to 187.5 ± 70.1 pg/ml. Osteoblasts cultivated on cancellous bone (ACB) showed mean concentrations of 185.2 ± 32.1 pg/ml after 1 week and 51.6 ± 14.8 pg/ml after 3 weeks of cultivation. The ACB group’s OPN concentrations dropped significantly from 1 week to 3 weeks (p = 0.0045). OPN concentrations were lower on cancellous bone after 1 (p = 0.050, n.s.) and after 3 weeks compared to concentrations on BioOss® (p = 0.052, n.s.) without reaching statistical significance (Fig. 5b).
Osteoprotegerin was secreted by human osteoblasts seeded onto BioOss®; concentrations averaged 61.7 ± 19.4 pg/ml after 1 week and 73.3 ± 32.5 pg/ml after 3 weeks. Osteoblasts seeded onto cancellous bone displayed concentrations of 29.7 ± 9.0 pg/ml after 1 week and of 10.6 ± 2.8 pg/ml after 3 weeks. OPG was expressed less in the supernatant of osteoblasts seeded onto cancellous bone when compared to BioOss® after 1 (p = 0.081, n.s.) and after 3 weeks (p = 0.039) (Fig. 5c).
We detected leptin after 1 week in the BioOss® group at concentrations of 10.7 ± 2.9 pg/ml, after 3 weeks of 8.9 ± 1.99 pg/ml; concentrations of 11.9 ± 0.9 pg/ml and 4.1 ± 1.5 pg/ml were measured in the ACB group, respectively. The ACB group’s decrease was significant from 1 to 3 weeks (p = 0.002) (Fig. 5d), but there was no statistically significant difference between the groups analyzing the respective time points.
Mean adiponectin concentration in the BioOss® group after 1 week reached 130.3 ± 50.9 pg/ml, and 103.1 ± 21.9 pg/ml after 3 weeks. Mean concentration in the allograft group after 1 week was 153.0 ± 25.3 pg/ml, and after 3 weeks 76.9 ± 19.2 pg/ml. The ACB group’s decrease from 1 to 3 weeks was again significant (p = 0.04) (Fig. 5e), but there was no statistically significant difference between the groups analyzing the respective time points.
Whichever graft material was used, human osteoblasts seeded onto ABC or SBS maintained their biochemical characteristics. In short: the expression of osteogenic biomarkers of human OB cultured on different graft materials is similar and reveals a time-dependent regulation pattern in-vitro.

Scanning electron microscopy (SEM)

Electron microscopy showed a homogeneous distribution pattern of osteoblasts seeded onto different bone graft materials, good adhesion to the material and the pericellular deposition of extracellular matrix. We also noted that that after just one week, osseous matrix adhered to the implants in both groups (Fig. 6a/b). After 3 weeks, intimate contact with osteoblasts embedded in a mineralized, fibril-rich extracellular matrix was present on both ACB and SBS (BioOss®). Although there were no significant differences between the two groups in cell viability or proliferation, semiquantative impression indicated slightly more extensive osteoblast growth on the BioOss® surface (Fig. 6c/d). We observed no interpositioned interfacial layer or foreign-body reactions (not expected) in any of the samples.

Discussion

Aim of this study was to compare the effectiveness of treating bony lesions with allografts (ACB) versus synthetic or highly-processed xenogeneic bone grafts (SBS) in terms of patient-dependent factors and the various sites of the bone defects. The key finding we made in this study is the similar efficacy of ACB and SBS in the treatment of skeletal lesions in-vivo and in-vitro, confirming our hypothesis. Other factors such as the location of use and epidemiological aspects have a greater impact on the clinical outcome than does the type of bone graft.
We detected no significant differences between the ACB and SBS groups either in their clinical findings or in-vitro biocompatibility. The consolidation rate was influenced significantly by epidemiological and patient-dependent factors such as age, ASA risk classification, smoking habits and type of insurance, but not by the bone-graft material used. Interestingly, although the defect size differed significantly between the groups, the consolidation rate was not influenced by it. The complication rate in our study correlated very significantly with defect location. Complication and consolidation rates did not display significant differences between ACB and SBS within the same location, however. Furthermore, the complication rate was also not influenced significantly by the biomaterial group used, but the bony-healing rate did correlate significantly with the type of insurance, revealing worse outcomes in patients covered by an employer’s or compulsory health insurance in comparison to those with private coverage. While patients cannot influence the injury site and/or fracture or defect characteristics, bony healing may be influenced by epidemiological and patient-dependent factors, such as a healthy life style. Well-educated patients are more likely to have private health insurance and show potentially better compliance, factors that would explain these clinical findings.
Current evidence-based treatment paradigms for bone grafting in bony defects or fracture healing remain still elusive and are frequently controversial [23, 24]. Most of the available literature addresses medium- and long-term outcomes after revision arthroplasty [25]. The latest data on bone grafting in fracture care merely focuses on certain anatomical locations [24, 26]. There is very little data on bone substitutes and a paucity of randomized controlled studies in particular, an observation also supported by a Cochrane review from Board et al. concerning the comparison of processed versus fresh-frozen bone for impaction bone grafting in revision hip arthroplasty. No randomized controlled trial comparing the clinical use of processed versus fresh-frozen bone in revision hip surgery has been published. Surgeons’ choices of bone graft nowadays are more apt to be based on personal preference and the graft material’s availability than on evidence [23]. Our data do not contradict to this approach, because we could show that complication rates after bone grafting of the hip is associated with a high success rate independent on the used material. In contrast, several randomized trials on bone-graft alternatives for spinal fusion procedures have been published. Approximately 1,400 products for use as bone void fillers are available on the international market [30]. Most of the trials compare the clinical effectiveness of allografts, synthetic/processed bone grafts or ceramics with autologous bone grafting. Thalgott et al. conducted a prospective, blind, single-site study to evaluate the outcomes and fusion rates of anterior lumbar interbody fusions with fresh-frozen or freeze-dried femoral ring allograft as part of a circumferential fusion in 40 patients. Radiography revealed fusion in 71.4 % of the 40 fusion levels [31]. Correlating with our data, smoking was in this study strongly associated with pseudarthrosis development. 58.3 % of the smokers who received freeze-dried allografts and 16.7 % of the smokers who underwent fresh-frozen allografts required revision for non-unions. In contrast, only one non-smoker (7.1 %) required revision for pseudarthrosis after a freeze-dried allograft and another non-smoker after a fresh-frozen allograft. Those results support our study findings and confirm the influence of smoking on the consolidation rate. Suchomel et al. conducted a prospective semi-randomized study comparing allograft to iliac crest bone graft in 79 patients undergoing one- or two-level anterior cervical discectomy and fusion, detecting no difference in graft migration or collapse. They noted a significantly slower fusion rate at 3 and 6 months in the allograft group, which was no longer present at 1 and 2 years [32]. A possible explanation are the immunological consequences of the histologically shown cell remnants that were found during our investigation. Several other studies confirm good consolidation rates in the field of spine fusion independent of the graft material [30, 33], which would be in line with our finding that this anatomical location provides a good biological environment for graft integration. In their prospective, randomized study of 29 patients, McConnell et al. observed 100 % fusion rates in anterior cervical discectomy and fusion with either coralline hydroxyapatite or iliac crest bone grafting. However, the hydroxyapatite group displayed a significantly higher rate of graft fragmentation and settling [30, 34]. Fischer et al. recommend the use of ceramics in common spinal fusions as osteoconductors, but not without employing additional osteoinductive material [30]. In fact, a combination of autologous graft material and SBS was used in 44 out of 45 cases of bone grafting in the spinal area in our study. Overall, good consolidation rates in the field of spine fusion may result from the combination of the osteoconductive properties of SBS and osteoinductive capacities of autologous graft material. Furthermore, a usually ample blood supply may support consolidation in spine fusion independent of different osteoconductive graft materials. Nevertheless, problems arise with the use of different graft materials when consolidation is radiologically assessed. A scintigraphic study of 12 patients comparing hydroxyapatite grafts and iliac crest bone graft showed the same rate of uptake until fusion. Therefore, the authors concluded that the radiolucent line is not always a sign of pseudarthrosis in patients treated with hydroxyapatite [35]. As long as the fusion confirmation is based on X-ray or CT-scan, it is hard to differentiate between the bone graft material itself and a lack of bone ingrowth. Multiple studies on clinical outcomes after posterolateral lumbar fusion using ceramics as bone graft extender have reported good results. Hence, Fischer et al. hypothesize that the radiographic findings represent new bone formation [30].
The challenges of post-interventional radiologic assessment lead to the subject of bone grafting in fracture care. Unlike autograft or allograft, the total radiographic disappearance of hydroxyapatite is not to be expected, since its incorporation is associated with osseous ingrowth and slight marginal resorption. Hydroxyapatite possesses osteoconductive properties and serves as a scaffold, in contrast to the complete remodeling that occurs with auto- and allografts. Concerning bone grafting in fracture care, current data basically focuses on certain anatomical locations [24, 26]. Herrera et al. reported good clinical results in 17 patients after external fixation and cancellous allografting with freeze-dried, irradiated cancellous bone in unstable distal radius fractures. In all cases, bone-graft incorporation was evident, non-union did not occur. Incorporation was defined as the loss of allograft radiodensity and trabeculae crossing the fracture line [26]. A study by Arora et al. evaluated complications following internal fixation using a palmar 2.4 mm-locking compression plate in unstable distal radius fractures, with most complications involving tendon irritation. Delayed fracture union occurred in 3 patients, and 1 patient experienced intra-operative intra-articular screw displacement [36]. Correlating with the described excellent bone healing capacity of the forearm, we detected no non-union after bone grafting in the forearm in our study, and only one complication. The risk of non-union is minimal in distal radius fractures. Consequently, bone-graft substitutes are primarily used to provide structural stability and fill larger bony defects [37]. Furthermore, advances in plate design and technology such as locking plates [38] reduce the field of application for bone grafts in distal radius fractures. There is still a lack of robust evidence concerning bone grafting in distal radius fractures. A Cochrane Database analysis of randomized clinical trials concluded that there was insufficient evidence regarding functional outcomes and safety in the use of bone grafts and substitutes to treat distal radius fractures [39]. Bone quality, the size of the bony defect, blood flow to the fracture site, and the method of fixation/immobilization, as well as patient-dependent epidemiological factors all affect the healing process and maintenance of fracture reduction. As shown by our data, different anatomic locations entail varying levels of bone-forming activity and stability. Therefore, a single study validating the use of a bone-graft material in one location may not predict its performance in another anatomic site. Good clinical results concerning bone grafting in spine surgery, hip-joint arthroplasty or its application in distal radius fractures [25, 30, 39, 40] are in line with our results from both groups of bone-graft materials in those locations.
With tibial plateau fractures, restoration of the plateau surface is a key treatment factor. Residual depression zones in the weight-bearing area on the tibial plateau bear the risk of axis deviation and posttraumatic osteoarthritis [41]. However, restoring the tibial-plateau anatomy often leaves larger cancellous bone defects, especially when bone fragments have become impacted within the soft metaphyseal bone. Various bone-graft materials are available and have been used to fill metaphyseal cancellous bone defects. Various challenges exist, such as inferior mechanical properties, donor-site morbidity in autografting, toxic and exothermic reactions when using traditional polymethyl-methacrylate cement, graft-material dislocation and custom-designed challenges such as graft preshaping or the inappropriate application form. Lobenhoffer et al. conducted a prospective study of 26 patients suffering from type B2, B3 and C3 fractures of the tibial plateau using an injectable calcium phosphate bone cement. 2 patients presented partial secondary loss of reduction, one of whom could not comply with partial weight bearing [42]. Veitch et al. presented a study of 6 patients treated with compaction morselized fresh-frozen bone allograft in tibial-plateau fractures. Despite their small cohort, the authors recommend the treatment of tibial-plateau fractures with fresh-frozen allograft because of its good graft incorporation, good remodeling and lower costs [24]. Several single-site studies addressing the use of different bone-graft materials in tibial-plateau fractures are available, but there is little data from controlled, randomized studies of bone grafting in tibia-plateau fractures. A current study protocol of Nusselt et al. is comparing the treatment of fracture defects in tibial-plateau fractures with a bioresorbable hydroxyapatite/calcium sulphate cement (Cerament™ Bone Void Filler) and autologous bone grafting [43]. Correlating with the comparable experiences of different material classes, we observed no significant group differences in consolidation or complication rates in tibial-plateau fractures in our study. 76 % of our study patients with a tibial-plafond fracture underwent fracture treatment with SBS to prevent the donor-site morbidity associated with autografting and potential immune reactions after allografting for relatively smaller bone defects. Only 2 failures of consolidation were documented, whereas an 18 % complication rate in tibial-plateau fracture therapy in both intervention groups was recorded. The complications we observed are in line with the current literature concerning tibial-plateau fractures, as they tend to entail secondary loss of reduction and wound infections [24, 44].
There is a paucity of evidence-based data on bone grafting in humeral bone defects. Daugaard et al. conducted a study in 20 canine proximal humerus, examining the combined effect of parathyroid hormone and bone graft on implant fixation [45]. Taehoon et al. evaluated the effect of β-tricalcium phosphate and poly-L-lactide-co-glycolide-co-epsiolon-caprolactone membrane in canine-humerus defects, obtaining the expected results from the osteoconductive material, leading to the original morphology’s restoration. However, the new cortical bone was thinner and less well organized than the adjacent intact cortex, and the amount of new cancellous bone was also sparse [46]. Studies in humans mainly focus on fracture care, the anatomical characteristics of different fracture locations, and frequent complications [4750]. Südkamp et al. conducted a prospective, multicenter, observation study investigating the open reduction and internal fixation of proximal humeral fractures employing a proximal humerus locking plate; 34 % complications were registered in 155 patients. 40 % of the complications were attributable to problems of surgical technique, mostly the intra-operative perforation of the humeral head by a screw. Further complications included plate fracture (1.9 %), impingement (2.6 %), pseudarthrosis (2.6 %), wound infection (3.9 %), loss of reduction (7.1 %), and necrosis of the humeral head (3.9 %) [51]. Although the general complication rate in our study cohort amounted to only 14.2 %, the location-specific complication rate was 30.3 % for the humerus. This higher proportion is in line with the findings of Südkamp et al. and seen after fracture care of the proximal humerus with a locking plate. A failure of consolidation in the humerus only occurred in 1 of 19 patients (SBS), whereas 10 complications occurred (5 in each group). Good consolidation rates in both groups indicate that treatment efficacy depends on other influencing factors (such as epidemiological aspects, as well as local factors such as blood supply, bone quality, anatomical characteristics and mechanical demands) rather than on the bone-graft material, at least when it merely provides osteoconductive properties. Other bone-graft techniques in humeral fractures aim to prevent known difficulties. Khmelnitskaya et al. described a D.G. Lorich technique to treat 4-part proximal humerus fractures using an intramedullary fibula strut graft as a reduction aid and as a structural augmentation for screw purchase in patients with poor bone quality. This technique seems to avoid frequent problems in humeral-fracture care such as screw penetration or hardware cut-out [47], but it does not improve the already good consolidation rate.
BioOss® was most frequently used in the SBS group, thus we decided to compare the in-vitro characteristics of ACB and BioOss®. Allografts and highly-processed bovine hydroxyapatite showed similar in-vitro osteoconductive properties and similar biocompatibility. Human osteoblasts on both materials maintained their stable osteogenic phenotype, resulting in the expression of similar amounts of osseous marker proteins. SEM imaging demonstrated good adherence of the osteoblasts on both materials. After a 3-week culture, osteoblasts were organized as nodular aggregations, and the amount of deposited extracellular matrix was greater than that after 1 week’s cultivation. SEM imaging demonstrated dense collagen nanofibers deposited on the scaffold spread homogenously and covering the entire surface. Those findings concur with the results of Sachar et al., who investigated osteoblasts gained from mice calvaria on three-dimensional nanofibrous gelatin scaffolds [52]. However, they noted an increase in protein synthesis and greater cell density in 5-day cultures compared to cultures lasting 14 days. We hypothesized that the decrease in protein and hormone concentrations in both our study groups may be caused by a limited amount of medium substrates in the in-vitro cultures. Another reason for the decreasing proliferation and protein synthesis in-vitro in our study might be the use of human osteoblasts isolated from elderly patients who had undergone hip-arthroplasty surgery following femoral neck fractures, whereas many other studies employed murine osteoblasts [52, 53].
We examined the protein and hormone synthesis of OPN, OC, OPG, leptin and adiponectin in osteoblasts on ACB and SBS. Osteopontin, a glycoprotein also known as bone sialoprotein I, is a linking protein that binds hydroxyapatite, and its cell-adhesion properties are required for osteoclastogenesis. OPN is expressed during the early stages of differentiation of osteoclast and osteoblast progenitors. OPN and OC are considered the most sensitive markers for bone-specific tissue formation [54, 55]. After 1 week of cultivation on BioOss®, we detected high levels of OPN and OC, whereas the early marker OPN decreased especially after 3 weeks. OC levels on ACB fell significantly after 3 weeks, indicating a lack of substrates or a lower proportion of matrix-embedded cells on ACB. Furthermore, diminished protein synthesis in osteoblasts isolated from elderly patients is conceivable. OPG and RANKL are known to be essential effectors for osteoclastogenesis [5658]. OPG, a cytokine receptor and member of the tumor necrosis factor receptor superfamily secreted by osteoblasts, inhibits differentiation of the osteoclast precursor into mature osteoclasts by attaching to RANKL and inhibiting osteoclastogenesis [59]. An increasing level of OPG on BioOss® from 1 to 3 weeks indicates an anabolic condition, whereas OPG on cancellous bone was expressed significantly less after 3 weeks. Luo et al. described the interrelation of adiponectin and the RANKL/OPG axis on bone metabolism. Adiponectin induces RANKL and inhibits OPG expression in human osteoblasts through the AdipoR1/p38 MAPK pathway, inducing osteoclast formation [60]. The different dynamics in protein levels of adiponectin and OPG on BioOss®, together with a rise in OPG after 3 weeks and a falling level of adiponectin therefore reveal anabolic bone metabolism on BioOss® after 3 weeks. The adiponectin level on ACB rises, whereas OPG decreases, indicating a catabolic condition, which may be supported by overall lower protein levels (OC, OPN) on ACB in comparison to BioOss®. The role of leptin, a pluripotent hormone linked to human body fat regulation in bone metabolism is controversial [6163]. Reseland et al. demonstrated leptin secretion of human osteoblasts, which promotes osteoblastic cell growth and bone mineralization via an autocrine and endocrine mechanism [64]. However, the expression of leptin seems to be restricted to the mineralization and/or osteocyte transition period during hOB differentiation [65]. The nearly-constant leptin level of hOB cultured on BioOss® from 1 to 3 weeks indicates the ongoing mineralization phase, which is supported by SEM findings showing ample matrix deposition, whereas a significant decrease was observed in the leptin level on ACB from 1 to 3 weeks. Significant differences between the protein and hormone synthesis on ACB and SBS were only detected in conjunction with particular parameters in a temporal pattern. Nevertheless, hOB cultivated on BioOss® tend to demonstrate more beneficial levels of bone-specific markers. Sachar et al. emphasize the importance of three-dimensional cell embedding of hOB concerning the expression of cell-adhesion proteins and of other osteogenic marker genes [52]. Allografts obtained from older donors may be inferior to SBS in terms of their osteoconductive properties because of lower bone-mineral density. The SEM analysis revealed a greater amount of osteoblasts on SBS and a homogenously-distributed matrix deposition enabling three-dimensional embedding of the cells. Another explanation for the slightly inferior protein synthesis on ACB is the freeze-drying process and/or subsequent irradiation that might cause alterations in the anorganic structure. Organic or cellular remnants in the allograft might trigger immune reactions or impaired growth and secretion behavior of hOB despite the purging processes. Our hypothesis that in-vitro studies may predict in-vivo data could be confirmed; ACB and SBS had similar biocompatibility with osteoblasts and clinically equal applicability. However, considering the importance of clinically influencing factors, the informative and especially predictive value of the in-vitro studies seems to be limited.
Although we identified no significant group differences in the Alamar Blue® assay, a tendency towards a decrease of cell numbers and metabolic activity, especially in the ACB group, was seen. Despite, this demonstrates similar in-vitro biocompatibility of ACB and SBS. The decreasing cell number over time may be explained by the absent blood supply, which more efficiently transports nutrients and growth hormones compared to cell-culture-typical diffusion, a phenomenon described by e.g., Kneser et al. [66]. This would also explain the decline in osteogenic marker production that was especially seen in the ACB group after the first week.
The aim of this study was to compare and determine the clinical effectiveness of allogeneic cancellous bone grafts (ACB) and synthetic or highly-processed xenogeneic bone substitutes (SBS) to treat bony defects in different locations while considering epidemiological and custom-designed parameters. We hypothesized that there would be no difference between ACB and SBS in their effectiveness to regenerate and consolidate bone, a hypothesis we confirmed. Our study’s complication rate correlated highly significantly with the site of the bone defect, but not with the bone-graft material used.
We are aware of our study’s limitations. The SBS group contains a heterogenous amount of different xenogenic and synthetic bone-graft materials reflecting the clinical routine and occasional personal preferences. Nevertheless, the most important finding we made is the similar efficacy of ACB and SBS in the treatment of skeletal lesions independent of the graft material used. Other key factors such as the location of use and epidemiological aspects are much more likely to influence clinical outcomes than is the type of bone graft.
The main bone-grafting sites in our study were the humerus, tibia and foot in both intervention groups, in line with the usual complication rates in fracture care and implying that the occurrence of complications is not closely related to bone grafting. The consolidation rate, our other study endpoint, is primarily and significantly influenced by epidemiological factors, again not by the biomaterial applied. The in-vitro analysis comparing the different classes of bone substitutes is limited to biocompatibility assays in combination with measurements of factors defining an osteogenic environment. Although SEM supplemented this, the examination lacks other cell types as osteoclasts, osteocytes or chondrocytes that are necessary in-vivo for the natural bone turnover.
In allogeneic bone transplantation, the risk of microbiological contamination, transmission of viruses, delayed incorporation, and cellular and humoral immune reactions must be taken into consideration [67]. There have been many satisfactory clinical results reported following the application of allo- and xenografts [68, 69]. Nevertheless, Reikeras et al. describe complications after using deep-frozen allogeneic bone in 25 to 35 % of patients [70]. Sterilization and disinfection can reduce the immune response and the risk of infection, but they have major effects on the grafts’ mechanical and biological properties. Shegarfi et al. suspect that major histocompatibility complex peptide proteins from donor cells might survive the freezing process, making them subject to targeting by antigen-presenting cells after transplantation, thus triggering a long-term immune response [69]. Ghanaati et al. recently published data about organic and cellular remnants in allogenic and xenogenic bone blocks subjected to patented processing techniques that may cause immune reactions [11].
Due to the sterilization process prior to transplantation, allografts usually only provide osteoconductive properties, having lost their osteogenic capacity by purging from organic matrix and cell components. Nevertheless, our histologic findings confirm the existence of soft tissue as well as osteocytes, adipocytes and fibrocytes in the ACB graft. ACB’s high osteogenic capacity after the sterilization process seems doubtful. Lower levels of protein and hormone secretion on ACB in comparison to BioOss® may even reflect immune-reactions processes or compromised cell growth and cell metabolism due to effects on the mechanical and biological properties of ACB.
Individual differences such as age and the co-morbidities of our study’s osteoblast donors could have caused differences in growth characteristics, protein secretion levels and matrix deposition without attaining significance. Defects involving greater diameters and costs will probably present limitations regarding the use of SBS. The risks associated with microbiological contamination, virus transmission, and cellular or humoral immune reactions can only be reduced through continued research, in particular of xenogenic material.

Conclusion

Both the materials we investigated (ACB vs. SBS) displayed similar in-vivo efficacy in the treatment of skeletal lesions. Other factors influencing the clinical outcome are the location of use and epidemiological parameters. Both materials were biocompatible in-vitro. We detected only marginal differences in the levels of protein and hormone secretion of hOBs cultivated on BioOss® or ACB.

Availability of data and materials section

The datasets supporting the conclusions of this article are included within the article and its additional files.

Acknowledgments

The study was funded by the AO Foundation Germany and the Department of Education and Research Germany (01EC1001D). BioOss®-Granula were kindly provided by Geistlich, Wolhusen, Switzerland. We thank Birgit Schäfer for her scientific support and advice.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

EJK was involved in the conception and the design of the study, the analysis and interpretation of the data, writing of the article, and the final approval of the submitted article. AB supported the lab work, supervised the sample analysis, was responsible for sample storage, revised the article draft critically, and approved the final version of the article. LW was responsible for the collection, assembly and management of data, performed the experiments, contributed to the article draft, calculated the scores and the descriptive statistics, and approved the final version of the article. TF has done the basic histology of the in-vitro-analyzed bone substitutes, contributed to the article draft, and approved the final article. KN was involved in the conception of the study, supervised histology examinations, critically revised the article draft, and approved the final version. HS was responsible for the conception and the design of the study, for obtaining of funding, the analysis and interpretation of the data, tutorial of Laura Wolf, drafting of the article, and the final approval of the submitted article. All authors take responsibility for the integrity of the work as a whole, from inception to finished article.
Literatur
1.
Zurück zum Zitat Banwart JC, Asher MA, Hassanein RS. Iliac crest bone graft harvest donor site morbidity. A statistical evaluation. Spine (Phila Pa 1976). 1995;20(9):1055–60.CrossRef Banwart JC, Asher MA, Hassanein RS. Iliac crest bone graft harvest donor site morbidity. A statistical evaluation. Spine (Phila Pa 1976). 1995;20(9):1055–60.CrossRef
2.
Zurück zum Zitat Soucacos PN, Johnson EO, Babis G. An update on recent advances in bone regeneration. Injury. 2008;39 Suppl 2:S1–4.CrossRef Soucacos PN, Johnson EO, Babis G. An update on recent advances in bone regeneration. Injury. 2008;39 Suppl 2:S1–4.CrossRef
3.
Zurück zum Zitat Jager M, Hernigou P, Zilkens C, Herten M, Fischer J, Krauspe R. [Cell therapy in bone-healing disorders]. Orthopade. 2010;39(4):449–62. quiz 463.CrossRefPubMed Jager M, Hernigou P, Zilkens C, Herten M, Fischer J, Krauspe R. [Cell therapy in bone-healing disorders]. Orthopade. 2010;39(4):449–62. quiz 463.CrossRefPubMed
4.
Zurück zum Zitat Bauer TW, Muschler GF. Bone graft materials. An overview of the basic science. Clin Orthop Relat Res. 2000;371:10–27.CrossRefPubMed Bauer TW, Muschler GF. Bone graft materials. An overview of the basic science. Clin Orthop Relat Res. 2000;371:10–27.CrossRefPubMed
5.
Zurück zum Zitat Roberts TT, Rosenbaum AJ. Bone grafts, bone substitutes and orthobiologics: the bridge between basic science and clinical advancements in fracture healing. Organogenesis. 2012;8(4):114–24.CrossRefPubMedPubMedCentral Roberts TT, Rosenbaum AJ. Bone grafts, bone substitutes and orthobiologics: the bridge between basic science and clinical advancements in fracture healing. Organogenesis. 2012;8(4):114–24.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Giannoudis PV, Dinopoulos H, Tsiridis E. Bone substitutes: an update. Injury. 2005;36 Suppl 3:S20–27.CrossRefPubMed Giannoudis PV, Dinopoulos H, Tsiridis E. Bone substitutes: an update. Injury. 2005;36 Suppl 3:S20–27.CrossRefPubMed
7.
Zurück zum Zitat Conrad EU, Gretch DR, Obermeyer KR, Moogk MS, Sayers M, Wilson JJ, et al. Transmission of the hepatitis-C virus by tissue transplantation. J Bone Joint Surg Am. 1995;77(2):214–24.PubMed Conrad EU, Gretch DR, Obermeyer KR, Moogk MS, Sayers M, Wilson JJ, et al. Transmission of the hepatitis-C virus by tissue transplantation. J Bone Joint Surg Am. 1995;77(2):214–24.PubMed
8.
Zurück zum Zitat Li CM, Ho YR, Liu YC. Transmission of human immunodeficiency virus through bone transplantation: a case report. J Formos Med Assoc. 2001;100(5):350–1.PubMed Li CM, Ho YR, Liu YC. Transmission of human immunodeficiency virus through bone transplantation: a case report. J Formos Med Assoc. 2001;100(5):350–1.PubMed
9.
Zurück zum Zitat Graham SM, Leonidou A, Aslam-Pervez N, Hamza A, Panteliadis P, Heliotis M, et al. Biological therapy of bone defects: the immunology of bone allo-transplantation. Expert Opin Biol Ther. 2010;10(6):885–901.CrossRefPubMed Graham SM, Leonidou A, Aslam-Pervez N, Hamza A, Panteliadis P, Heliotis M, et al. Biological therapy of bone defects: the immunology of bone allo-transplantation. Expert Opin Biol Ther. 2010;10(6):885–901.CrossRefPubMed
10.
Zurück zum Zitat Rodrigo JJ, Heiden E, Hegyes M, Sharkey NA. Immune response inhibition by irrigating subchondral bone with cytotoxic agents. Clin Orthop Relat Res. 1996;326:96–106.CrossRefPubMed Rodrigo JJ, Heiden E, Hegyes M, Sharkey NA. Immune response inhibition by irrigating subchondral bone with cytotoxic agents. Clin Orthop Relat Res. 1996;326:96–106.CrossRefPubMed
11.
Zurück zum Zitat Ghanaati S, Barbeck M, Booms P, Lorenz J, Kirkpatrick CJ, Sader RA. Potential lack of “standardized” processing techniques for production of allogeneic and xenogeneic bone blocks for application in humans. Acta Biomater. 2014;10(8):3557–62.CrossRefPubMed Ghanaati S, Barbeck M, Booms P, Lorenz J, Kirkpatrick CJ, Sader RA. Potential lack of “standardized” processing techniques for production of allogeneic and xenogeneic bone blocks for application in humans. Acta Biomater. 2014;10(8):3557–62.CrossRefPubMed
12.
Zurück zum Zitat Hallfeldt KK, Stutzle H, Puhlmann M, Kessler S, Schweiberer L. Sterilization of partially demineralized bone matrix: the effects of different sterilization techniques on osteogenetic properties. J Surg Res. 1995;59(5):614–20.CrossRefPubMed Hallfeldt KK, Stutzle H, Puhlmann M, Kessler S, Schweiberer L. Sterilization of partially demineralized bone matrix: the effects of different sterilization techniques on osteogenetic properties. J Surg Res. 1995;59(5):614–20.CrossRefPubMed
13.
Zurück zum Zitat Knaepler H, Haas H, Puschel HU. [Biomechanical properties of heat and irradiation treated spongiosa]. Unfallchirurgie. 1991;17(4):194–9.CrossRefPubMed Knaepler H, Haas H, Puschel HU. [Biomechanical properties of heat and irradiation treated spongiosa]. Unfallchirurgie. 1991;17(4):194–9.CrossRefPubMed
14.
Zurück zum Zitat Blokhuis TJ, Lindner T. Allograft and bone morphogenetic proteins: an overview. Injury. 2008;39 Suppl 2:S33–36.CrossRefPubMed Blokhuis TJ, Lindner T. Allograft and bone morphogenetic proteins: an overview. Injury. 2008;39 Suppl 2:S33–36.CrossRefPubMed
15.
Zurück zum Zitat Fretwurst T, Spanou A, Nelson K, Wein M, Steinberg T, Stricker A. Comparison of four different allogeneic bone grafts for alveolar ridge reconstruction: a preliminary histologic and biochemical analysis. Oral Surg Oral Med Oral Pathol Oral Radiol. 2014;118:424–31.CrossRefPubMed Fretwurst T, Spanou A, Nelson K, Wein M, Steinberg T, Stricker A. Comparison of four different allogeneic bone grafts for alveolar ridge reconstruction: a preliminary histologic and biochemical analysis. Oral Surg Oral Med Oral Pathol Oral Radiol. 2014;118:424–31.CrossRefPubMed
16.
Zurück zum Zitat Heyligers IC, Klein-Nulend J. Detection of living cells in non-processed but deep-frozen bone allografts. Cell Tissue Bank. 2005;6(1):25–31.CrossRefPubMed Heyligers IC, Klein-Nulend J. Detection of living cells in non-processed but deep-frozen bone allografts. Cell Tissue Bank. 2005;6(1):25–31.CrossRefPubMed
17.
Zurück zum Zitat Deakin DE, Bannister GC. Graft incorporation after acetabular and femoral impaction grafting with washed irradiated allograft and autologous marrow. J Arthroplasty. 2007;22(1):89–94.CrossRefPubMed Deakin DE, Bannister GC. Graft incorporation after acetabular and femoral impaction grafting with washed irradiated allograft and autologous marrow. J Arthroplasty. 2007;22(1):89–94.CrossRefPubMed
18.
Zurück zum Zitat Robinson DE, Lee MB, Smith EJ, Learmonth ID. Femoral impaction grafting in revision hip arthroplasty with irradiated bone. J Arthroplasty. 2002;17(7):834–40.CrossRefPubMed Robinson DE, Lee MB, Smith EJ, Learmonth ID. Femoral impaction grafting in revision hip arthroplasty with irradiated bone. J Arthroplasty. 2002;17(7):834–40.CrossRefPubMed
19.
Zurück zum Zitat Elting JJ, Mikhail WE, Zicat BA, Hubbell JC, Lane LE, House B. Preliminary report of impaction grafting for exchange femoral arthroplasty. Clin Orthop Relat Res. 1995;319:159–67.PubMed Elting JJ, Mikhail WE, Zicat BA, Hubbell JC, Lane LE, House B. Preliminary report of impaction grafting for exchange femoral arthroplasty. Clin Orthop Relat Res. 1995;319:159–67.PubMed
20.
Zurück zum Zitat Gie GA, Linder L, Ling RS, Simon JP, Slooff TJ, Timperley AJ. Impacted cancellous allografts and cement for revision total hip arthroplasty. J Bone Joint Surg. 1993;75(1):14–21. Gie GA, Linder L, Ling RS, Simon JP, Slooff TJ, Timperley AJ. Impacted cancellous allografts and cement for revision total hip arthroplasty. J Bone Joint Surg. 1993;75(1):14–21.
21.
Zurück zum Zitat Schreurs BW, Bolder SB, Gardeniers JW, Verdonschot N, Slooff TJ, Veth RP. Acetabular revision with impacted morsellised cancellous bone grafting and a cemented cup. A 15- to 20-year follow-up. J Bone Joint Surg. 2004;86(4):492–7. Schreurs BW, Bolder SB, Gardeniers JW, Verdonschot N, Slooff TJ, Veth RP. Acetabular revision with impacted morsellised cancellous bone grafting and a cemented cup. A 15- to 20-year follow-up. J Bone Joint Surg. 2004;86(4):492–7.
22.
Zurück zum Zitat Stevenson S, Emery SE, Goldberg VM. Factors affecting bone graft incorporation. Clin Orthop Relat Res. 1996;324:66–74.CrossRefPubMed Stevenson S, Emery SE, Goldberg VM. Factors affecting bone graft incorporation. Clin Orthop Relat Res. 1996;324:66–74.CrossRefPubMed
23.
Zurück zum Zitat Board TN, Brunskill S, Doree C, Hyde C, Kay PR, Meek RD, Webster R, Galea G. Processed versus fresh frozen bone for impaction bone grafting in revision hip arthroplasty. Cochrane Database Syst Rev. 2009;4:CD006351. Board TN, Brunskill S, Doree C, Hyde C, Kay PR, Meek RD, Webster R, Galea G. Processed versus fresh frozen bone for impaction bone grafting in revision hip arthroplasty. Cochrane Database Syst Rev. 2009;4:CD006351.
24.
Zurück zum Zitat Veitch SW, Stroud RM, Toms AD. Compaction bone grafting in tibial plateau fracture fixation. J Trauma. 2010;68(4):980–3.CrossRefPubMed Veitch SW, Stroud RM, Toms AD. Compaction bone grafting in tibial plateau fracture fixation. J Trauma. 2010;68(4):980–3.CrossRefPubMed
25.
Zurück zum Zitat Mehendale S, Learmonth ID, Smith EJ, Nedungayil S, Maheshwari R, Hassaballa MA. Use of irradiated bone graft for impaction grafting in acetabular revision surgery: a review of fifty consecutive cases. Hip Int. 2009;19(2):114–9.PubMed Mehendale S, Learmonth ID, Smith EJ, Nedungayil S, Maheshwari R, Hassaballa MA. Use of irradiated bone graft for impaction grafting in acetabular revision surgery: a review of fifty consecutive cases. Hip Int. 2009;19(2):114–9.PubMed
26.
Zurück zum Zitat Herrera M, Chapman CB, Roh M, Strauch RJ, Rosenwasser MP. Treatment of unstable distal radius fractures with cancellous allograft and external fixation. J Hand Surg Am. 1999;24(6):1269–78.CrossRefPubMed Herrera M, Chapman CB, Roh M, Strauch RJ, Rosenwasser MP. Treatment of unstable distal radius fractures with cancellous allograft and external fixation. J Hand Surg Am. 1999;24(6):1269–78.CrossRefPubMed
27.
Zurück zum Zitat Finkenzeller G, Mehlhorn AT, Schmal H, Stark GB. Post-transcriptional regulation of osteoblastic platelet-derived growth factor receptor-alpha expression by co-cultured primary endothelial cells. Cells Tissues Organs. 2010;192(1):28–38.CrossRefPubMed Finkenzeller G, Mehlhorn AT, Schmal H, Stark GB. Post-transcriptional regulation of osteoblastic platelet-derived growth factor receptor-alpha expression by co-cultured primary endothelial cells. Cells Tissues Organs. 2010;192(1):28–38.CrossRefPubMed
28.
Zurück zum Zitat Mashige F, Imai K, Osuga T. A simple and sensitive assay of total serum bile acids. Clin Chim Acta. 1976;70(1):79–86.CrossRefPubMed Mashige F, Imai K, Osuga T. A simple and sensitive assay of total serum bile acids. Clin Chim Acta. 1976;70(1):79–86.CrossRefPubMed
29.
Zurück zum Zitat Schäfer B. MilliplexTM Human Bone Panel - Standard Operation Protocol. Geislich Biomaterials: Wolhusen; 2009. Schäfer B. MilliplexTM Human Bone Panel - Standard Operation Protocol. Geislich Biomaterials: Wolhusen; 2009.
30.
Zurück zum Zitat Fischer CR, Cassilly R, Cantor W, Edusei E, Hammouri Q, Errico T. A systematic review of comparative studies on bone graft alternatives for common spine fusion procedures. Eur Spine J. 2013;22(6):1423–35.CrossRefPubMedPubMedCentral Fischer CR, Cassilly R, Cantor W, Edusei E, Hammouri Q, Errico T. A systematic review of comparative studies on bone graft alternatives for common spine fusion procedures. Eur Spine J. 2013;22(6):1423–35.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Thalgott JS, Fogarty ME, Giuffre JM, Christenson SD, Epstein AK, Aprill C. A prospective, randomized, blinded, single-site study to evaluate the clinical and radiographic differences between frozen and freeze-dried allograft when used as part of a circumferential anterior lumbar interbody fusion procedure. Spine (Phila Pa 1976). 2009;34(12):1251–6.CrossRef Thalgott JS, Fogarty ME, Giuffre JM, Christenson SD, Epstein AK, Aprill C. A prospective, randomized, blinded, single-site study to evaluate the clinical and radiographic differences between frozen and freeze-dried allograft when used as part of a circumferential anterior lumbar interbody fusion procedure. Spine (Phila Pa 1976). 2009;34(12):1251–6.CrossRef
32.
Zurück zum Zitat Suchomel P, Barsa P, Buchvald P, Svobodnik A, Vanickova E. Autologous versus allogenic bone grafts in instrumented anterior cervical discectomy and fusion: a prospective study with respect to bone union pattern. Eur Spine J. 2004;13(6):510–5.CrossRefPubMedPubMedCentral Suchomel P, Barsa P, Buchvald P, Svobodnik A, Vanickova E. Autologous versus allogenic bone grafts in instrumented anterior cervical discectomy and fusion: a prospective study with respect to bone union pattern. Eur Spine J. 2004;13(6):510–5.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Cho DY, Lee WY, Sheu PC, Chen CC. Cage containing a biphasic calcium phosphate ceramic (Triosite) for the treatment of cervical spondylosis. Surg Neurol. 2005;63(6):497–503. discussion 503–494.CrossRefPubMed Cho DY, Lee WY, Sheu PC, Chen CC. Cage containing a biphasic calcium phosphate ceramic (Triosite) for the treatment of cervical spondylosis. Surg Neurol. 2005;63(6):497–503. discussion 503–494.CrossRefPubMed
34.
Zurück zum Zitat McConnell JR, Freeman BJ, Debnath UK, Grevitt MP, Prince HG, Webb JK. A prospective randomized comparison of coralline hydroxyapatite with autograft in cervical interbody fusion. Spine (Phila Pa 1976). 2003;28(4):317–23. McConnell JR, Freeman BJ, Debnath UK, Grevitt MP, Prince HG, Webb JK. A prospective randomized comparison of coralline hydroxyapatite with autograft in cervical interbody fusion. Spine (Phila Pa 1976). 2003;28(4):317–23.
35.
Zurück zum Zitat Iseda T, Nakano S, Suzuki Y, Miyahara D, Uchinokura S, Moriyama T, Sameshima T, Goya T, Wakisaka S. Radiographic and scintigraphic courses of union in cervical interbody fusion: hydroxyapatite grafts versus iliac bone autografts. J Nucl Med. 2000;41(10):1642–5. Iseda T, Nakano S, Suzuki Y, Miyahara D, Uchinokura S, Moriyama T, Sameshima T, Goya T, Wakisaka S. Radiographic and scintigraphic courses of union in cervical interbody fusion: hydroxyapatite grafts versus iliac bone autografts. J Nucl Med. 2000;41(10):1642–5.
36.
Zurück zum Zitat Arora R, Gabl M, Erhart S, Schmidle G, Dallapozza C, Lutz M. Aspects of current management of distal radius fractures in the elderly individuals. Geriatr Orthop Surg Rehabil. 2011;2(5–6):187–94.CrossRefPubMedPubMedCentral Arora R, Gabl M, Erhart S, Schmidle G, Dallapozza C, Lutz M. Aspects of current management of distal radius fractures in the elderly individuals. Geriatr Orthop Surg Rehabil. 2011;2(5–6):187–94.CrossRefPubMedPubMedCentral
37.
38.
Zurück zum Zitat Schmal H, Strohm PC, Jaeger M, Sudkamp NP. Flexible fixation and fracture healing: do locked plating ‘internal fixators’ resemble external fixators? J Orthop Trauma. 2011;25 Suppl 1:S15–20.CrossRefPubMed Schmal H, Strohm PC, Jaeger M, Sudkamp NP. Flexible fixation and fracture healing: do locked plating ‘internal fixators’ resemble external fixators? J Orthop Trauma. 2011;25 Suppl 1:S15–20.CrossRefPubMed
39.
Zurück zum Zitat Handoll HH, Watts AC. Bone grafts and bone substitutes for treating distal radial fractures in adults. Cochrane Database Syst Rev. 2008;2:CD006836.PubMed Handoll HH, Watts AC. Bone grafts and bone substitutes for treating distal radial fractures in adults. Cochrane Database Syst Rev. 2008;2:CD006836.PubMed
40.
Zurück zum Zitat Oakes DA, Cabanela ME. Impaction bone grafting for revision hip arthroplasty: biology and clinical applications. J Am Acad Orthop Surg. 2006;14(11):620–8.PubMed Oakes DA, Cabanela ME. Impaction bone grafting for revision hip arthroplasty: biology and clinical applications. J Am Acad Orthop Surg. 2006;14(11):620–8.PubMed
41.
Zurück zum Zitat Moore TM, Patzakis MJ, Harvey JP. Tibial plateau fractures: definition, demographics, treatment rationale, and long-term results of closed traction management or operative reduction. J Orthop Trauma. 1987;1(2):97–119.CrossRefPubMed Moore TM, Patzakis MJ, Harvey JP. Tibial plateau fractures: definition, demographics, treatment rationale, and long-term results of closed traction management or operative reduction. J Orthop Trauma. 1987;1(2):97–119.CrossRefPubMed
42.
Zurück zum Zitat Lobenhoffer P, Gerich T, Witte F, Tscherne H. Use of an injectable calcium phosphate bone cement in the treatment of tibial plateau fractures: a prospective study of twenty-six cases with twenty-month mean follow-up. J Orthop Trauma. 2002;16(3):143–9.CrossRefPubMed Lobenhoffer P, Gerich T, Witte F, Tscherne H. Use of an injectable calcium phosphate bone cement in the treatment of tibial plateau fractures: a prospective study of twenty-six cases with twenty-month mean follow-up. J Orthop Trauma. 2002;16(3):143–9.CrossRefPubMed
43.
Zurück zum Zitat Nusselt T, Hofmann A, Wachtlin D, Gorbulev S, Rommens PM. CERAMENT treatment of fracture defects (CERTiFy): protocol for a prospective, multicenter, randomized study investigating the use of CERAMENT BONE VOID FILLER in tibial plateau fractures. Trials. 2014;15:75.CrossRefPubMedPubMedCentral Nusselt T, Hofmann A, Wachtlin D, Gorbulev S, Rommens PM. CERAMENT treatment of fracture defects (CERTiFy): protocol for a prospective, multicenter, randomized study investigating the use of CERAMENT BONE VOID FILLER in tibial plateau fractures. Trials. 2014;15:75.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Choo KJ, Morshed S. Postoperative complications after repair of tibial plateau fractures. J Knee Surg. 2014;27(1):11–9.CrossRefPubMed Choo KJ, Morshed S. Postoperative complications after repair of tibial plateau fractures. J Knee Surg. 2014;27(1):11–9.CrossRefPubMed
45.
Zurück zum Zitat Daugaard H, Elmengaard B, Andreassen TT, Baas J, Bechtold JE, Soballe K. The combined effect of parathyroid hormone and bone graft on implant fixation. J Bone Joint Surg. 2011;93(1):131–9.CrossRef Daugaard H, Elmengaard B, Andreassen TT, Baas J, Bechtold JE, Soballe K. The combined effect of parathyroid hormone and bone graft on implant fixation. J Bone Joint Surg. 2011;93(1):131–9.CrossRef
46.
Zurück zum Zitat Oh T, Rahman MM, Lim JH, Park MS, Kim DY, Yoon JH, et al. Guided bone regeneration with beta-tricalcium phosphate and poly L-lactide-co-glycolide-co-epsilon-caprolactone membrane in partial defects of canine humerus. J Vet Sci. 2006;7(1):73–7.CrossRefPubMedPubMedCentral Oh T, Rahman MM, Lim JH, Park MS, Kim DY, Yoon JH, et al. Guided bone regeneration with beta-tricalcium phosphate and poly L-lactide-co-glycolide-co-epsilon-caprolactone membrane in partial defects of canine humerus. J Vet Sci. 2006;7(1):73–7.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Khmelnitskaya E, Lamont LE, Taylor SA, Lorich DG, Dines DM, Dines JS. Evaluation and management of proximal humerus fractures. Adv Orthop. 2012;2012:861598.CrossRefPubMedPubMedCentral Khmelnitskaya E, Lamont LE, Taylor SA, Lorich DG, Dines DM, Dines JS. Evaluation and management of proximal humerus fractures. Adv Orthop. 2012;2012:861598.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Strohm PC, Kubosch DC, Hubner EJ, Sudkamp NP, Jaeger M, Reising K. [Therapy of humeral shaft fractures]. Chirurg. 2011;82(10):947–54. quiz 955.CrossRefPubMed Strohm PC, Kubosch DC, Hubner EJ, Sudkamp NP, Jaeger M, Reising K. [Therapy of humeral shaft fractures]. Chirurg. 2011;82(10):947–54. quiz 955.CrossRefPubMed
49.
Zurück zum Zitat Zhang W, Zeng L, Liu Y, Pan Y, Zhang W, Zhang C, et al. The mechanical benefit of medial support screws in locking plating of proximal humerus fractures. PLoS One. 2014;9(8):e103297.CrossRefPubMedPubMedCentral Zhang W, Zeng L, Liu Y, Pan Y, Zhang W, Zhang C, et al. The mechanical benefit of medial support screws in locking plating of proximal humerus fractures. PLoS One. 2014;9(8):e103297.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Maier D, Jaeger M, Izadpanah K, Strohm PC, Suedkamp NP. Proximal humeral fracture treatment in adults. J Bone Joint Surg Am. 2014;96(3):251–61.CrossRefPubMed Maier D, Jaeger M, Izadpanah K, Strohm PC, Suedkamp NP. Proximal humeral fracture treatment in adults. J Bone Joint Surg Am. 2014;96(3):251–61.CrossRefPubMed
51.
Zurück zum Zitat Sudkamp N, Bayer J, Hepp P, Voigt C, Oestern H, Kaab M, Luo C, Plecko M, Wendt K, Kostler W. Open reduction and internal fixation of proximal humeral fractures with use of the locking proximal humerus plate. Results of a prospective, multicenter, observational study. J Bone Joint Surg Am. 2009;91(6):1320–8. Sudkamp N, Bayer J, Hepp P, Voigt C, Oestern H, Kaab M, Luo C, Plecko M, Wendt K, Kostler W. Open reduction and internal fixation of proximal humeral fractures with use of the locking proximal humerus plate. Results of a prospective, multicenter, observational study. J Bone Joint Surg Am. 2009;91(6):1320–8.
52.
Zurück zum Zitat Sachar A, Strom TA, Serrano MJ, Benson MD, Opperman LA, Svoboda KK, et al. Osteoblasts responses to three-dimensional nanofibrous gelatin scaffolds. J Biomed Mater Res A. 2012;100(11):3029–41.CrossRefPubMedPubMedCentral Sachar A, Strom TA, Serrano MJ, Benson MD, Opperman LA, Svoboda KK, et al. Osteoblasts responses to three-dimensional nanofibrous gelatin scaffolds. J Biomed Mater Res A. 2012;100(11):3029–41.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Bilousova G, du Jun H, King KB, De Langhe S, Chick WS, Torchia EC, Chow KS, Klemm DJ, Roop DR, Majka SM. Osteoblasts derived from induced pluripotent stem cells form calcified structures in scaffolds both in vitro and in vivo. Stem Cells. 2011;29(2):206–16. Bilousova G, du Jun H, King KB, De Langhe S, Chick WS, Torchia EC, Chow KS, Klemm DJ, Roop DR, Majka SM. Osteoblasts derived from induced pluripotent stem cells form calcified structures in scaffolds both in vitro and in vivo. Stem Cells. 2011;29(2):206–16.
54.
Zurück zum Zitat Bianco P, Riminucci M, Bonucci E, Termine JD, Robey PG. Bone sialoprotein (BSP) secretion and osteoblast differentiation: relationship to bromodeoxyuridine incorporation, alkaline phosphatase, and matrix deposition. J Histochem Cytochem. 1993;41(2):183–91.CrossRefPubMed Bianco P, Riminucci M, Bonucci E, Termine JD, Robey PG. Bone sialoprotein (BSP) secretion and osteoblast differentiation: relationship to bromodeoxyuridine incorporation, alkaline phosphatase, and matrix deposition. J Histochem Cytochem. 1993;41(2):183–91.CrossRefPubMed
55.
Zurück zum Zitat Machwate M, Jullienne A, Moukhtar M, Marie PJ. Temporal variation of c-Fos proto-oncogene expression during osteoblast differentiation and osteogenesis in developing rat bone. J Cell Biochem. 1995;57(1):62–70.CrossRefPubMed Machwate M, Jullienne A, Moukhtar M, Marie PJ. Temporal variation of c-Fos proto-oncogene expression during osteoblast differentiation and osteogenesis in developing rat bone. J Cell Biochem. 1995;57(1):62–70.CrossRefPubMed
56.
Zurück zum Zitat Aubin JE, Bonnelye E. Osteoprotegerin and its ligand: a new paradigm for regulation of osteoclastogenesis and bone resorption. Osteoporos Int. 2000;11(11):905–13.CrossRefPubMed Aubin JE, Bonnelye E. Osteoprotegerin and its ligand: a new paradigm for regulation of osteoclastogenesis and bone resorption. Osteoporos Int. 2000;11(11):905–13.CrossRefPubMed
57.
58.
Zurück zum Zitat Takahashi N, Udagawa N, Suda T. A new member of tumor necrosis factor ligand family, ODF/OPGL/TRANCE/RANKL, regulates osteoclast differentiation and function. Biochem Biophys Res Commun. 1999;256(3):449–55.CrossRefPubMed Takahashi N, Udagawa N, Suda T. A new member of tumor necrosis factor ligand family, ODF/OPGL/TRANCE/RANKL, regulates osteoclast differentiation and function. Biochem Biophys Res Commun. 1999;256(3):449–55.CrossRefPubMed
59.
Zurück zum Zitat Kobayashi Y, Udagawa N, Takahashi N. Action of RANKL and OPG for osteoclastogenesis. Crit Rev Eukaryot Gene Expr. 2009;19(1):61–72.CrossRefPubMed Kobayashi Y, Udagawa N, Takahashi N. Action of RANKL and OPG for osteoclastogenesis. Crit Rev Eukaryot Gene Expr. 2009;19(1):61–72.CrossRefPubMed
60.
Zurück zum Zitat Luo XH, Guo LJ, Xie H, Yuan LQ, Wu XP, Zhou HD, et al. Adiponectin stimulates RANKL and inhibits OPG expression in human osteoblasts through the MAPK signaling pathway. J Bone Miner Res. 2006;21(10):1648–56.CrossRefPubMed Luo XH, Guo LJ, Xie H, Yuan LQ, Wu XP, Zhou HD, et al. Adiponectin stimulates RANKL and inhibits OPG expression in human osteoblasts through the MAPK signaling pathway. J Bone Miner Res. 2006;21(10):1648–56.CrossRefPubMed
61.
Zurück zum Zitat Cornish J, Callon KE, Bava U, Lin C, Naot D, Hill BL, et al. Leptin directly regulates bone cell function in vitro and reduces bone fragility in vivo. J Endocrinol. 2002;175(2):405–15.CrossRefPubMed Cornish J, Callon KE, Bava U, Lin C, Naot D, Hill BL, et al. Leptin directly regulates bone cell function in vitro and reduces bone fragility in vivo. J Endocrinol. 2002;175(2):405–15.CrossRefPubMed
62.
Zurück zum Zitat Thomas T, Gori F, Khosla S, Jensen MD, Burguera B, Riggs BL. Leptin acts on human marrow stromal cells to enhance differentiation to osteoblasts and to inhibit differentiation to adipocytes. Endocrinology. 1999;140(4):1630–8.PubMed Thomas T, Gori F, Khosla S, Jensen MD, Burguera B, Riggs BL. Leptin acts on human marrow stromal cells to enhance differentiation to osteoblasts and to inhibit differentiation to adipocytes. Endocrinology. 1999;140(4):1630–8.PubMed
63.
Zurück zum Zitat Ducy P, Amling M, Takeda S, Priemel M, Schilling AF, Beil FT, et al. Leptin inhibits bone formation through a hypothalamic relay: a central control of bone mass. Cell. 2000;100(2):197–207.CrossRefPubMed Ducy P, Amling M, Takeda S, Priemel M, Schilling AF, Beil FT, et al. Leptin inhibits bone formation through a hypothalamic relay: a central control of bone mass. Cell. 2000;100(2):197–207.CrossRefPubMed
64.
Zurück zum Zitat Reseland JE, Syversen U, Bakke I, Qvigstad G, Eide LG, Hjertner O, Gordeladze JO, Drevon CA. Leptin is expressed in and secreted from primary cultures of human osteoblasts and promotes bone mineralization. J Bone Miner Res. 2001;16(8):1426–33. Reseland JE, Syversen U, Bakke I, Qvigstad G, Eide LG, Hjertner O, Gordeladze JO, Drevon CA. Leptin is expressed in and secreted from primary cultures of human osteoblasts and promotes bone mineralization. J Bone Miner Res. 2001;16(8):1426–33.
65.
Zurück zum Zitat Stein GS, Lian JB, Stein JL, Van Wijnen AJ, Montecino M. Transcriptional control of osteoblast growth and differentiation. Physiol Rev. 1996;76(2):593–629.PubMed Stein GS, Lian JB, Stein JL, Van Wijnen AJ, Montecino M. Transcriptional control of osteoblast growth and differentiation. Physiol Rev. 1996;76(2):593–629.PubMed
66.
Zurück zum Zitat Kneser U, Stangenberg L, Ohnolz J, Buettner O, Stern-Straeter J, Mobest D, et al. Evaluation of processed bovine cancellous bone matrix seeded with syngenic osteoblasts in a critical size calvarial defect rat model. J Cell Mol Med. 2006;10(3):695–707.CrossRefPubMedPubMedCentral Kneser U, Stangenberg L, Ohnolz J, Buettner O, Stern-Straeter J, Mobest D, et al. Evaluation of processed bovine cancellous bone matrix seeded with syngenic osteoblasts in a critical size calvarial defect rat model. J Cell Mol Med. 2006;10(3):695–707.CrossRefPubMedPubMedCentral
67.
Zurück zum Zitat McAllister DR, Joyce MJ, Mann BJ, Vangsness Jr CT. Allograft update: the current status of tissue regulation, procurement, processing, and sterilization. Am J Sports Med. 2007;35(12):2148–58.CrossRefPubMed McAllister DR, Joyce MJ, Mann BJ, Vangsness Jr CT. Allograft update: the current status of tissue regulation, procurement, processing, and sterilization. Am J Sports Med. 2007;35(12):2148–58.CrossRefPubMed
68.
Zurück zum Zitat Houser BE, Mellonig JT, Brunsvold MA, Cochran DL, Meffert RM, Alder ME. Clinical evaluation of anorganic bovine bone xenograft with a bioabsorbable collagen barrier in the treatment of molar furcation defects. Int J Periodontics Restorative Dent. 2001;21(2):161–9.PubMed Houser BE, Mellonig JT, Brunsvold MA, Cochran DL, Meffert RM, Alder ME. Clinical evaluation of anorganic bovine bone xenograft with a bioabsorbable collagen barrier in the treatment of molar furcation defects. Int J Periodontics Restorative Dent. 2001;21(2):161–9.PubMed
69.
Zurück zum Zitat Shegarfi H, Reikeras O. Review article: bone transplantation and immune response. J Orthopaed Surg. 2009;17(2):206–11. Shegarfi H, Reikeras O. Review article: bone transplantation and immune response. J Orthopaed Surg. 2009;17(2):206–11.
70.
Zurück zum Zitat Reikeras O, Shegarfi H, Naper C, Reinholt FP, Rolstad B. Impact of MHC mismatch and freezing on bone graft incorporation: an experimental study in rats. J Orthop Res. 2008;26(7):925–31.CrossRefPubMed Reikeras O, Shegarfi H, Naper C, Reinholt FP, Rolstad B. Impact of MHC mismatch and freezing on bone graft incorporation: an experimental study in rats. J Orthop Res. 2008;26(7):925–31.CrossRefPubMed
Metadaten
Titel
Clinical trial and in-vitro study comparing the efficacy of treating bony lesions with allografts versus synthetic or highly-processed xenogeneic bone grafts
verfasst von
Eva Johanna Kubosch
Anke Bernstein
Laura Wolf
Tobias Fretwurst
Katja Nelson
Hagen Schmal
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
BMC Musculoskeletal Disorders / Ausgabe 1/2016
Elektronische ISSN: 1471-2474
DOI
https://doi.org/10.1186/s12891-016-0930-1

Weitere Artikel der Ausgabe 1/2016

BMC Musculoskeletal Disorders 1/2016 Zur Ausgabe

Arthropedia

Grundlagenwissen der Arthroskopie und Gelenkchirurgie. Erweitert durch Fallbeispiele, Videos und Abbildungen. 
» Jetzt entdecken

Update Orthopädie und Unfallchirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.