Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleClinical Studies

Impact of Zoledronic Acid and Denosumab Treatment on Growth Factor Concentration in Platelet Rich Fibrin of Patients With Osteolytic Bone Metastases

DANIEL STELLER, RONJA SIMON, ROBERT VON BIALY, RALPH PRIES and SAMER G. HAKIM
Anticancer Research August 2021, 41 (8) 3917-3923; DOI: https://doi.org/10.21873/anticanres.15187
DANIEL STELLER
1Departments of Maxillofacial Surgery, Head and Neck Surgery, University Hospital of Lübeck, Lübeck, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: Daniel.steller{at}uksh.de
RONJA SIMON
1Departments of Maxillofacial Surgery, Head and Neck Surgery, University Hospital of Lübeck, Lübeck, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ROBERT VON BIALY
1Departments of Maxillofacial Surgery, Head and Neck Surgery, University Hospital of Lübeck, Lübeck, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
RALPH PRIES
2Departments of Otolaryngology, Head and Neck Surgery, University Hospital of Lübeck, Lübeck, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SAMER G. HAKIM
1Departments of Maxillofacial Surgery, Head and Neck Surgery, University Hospital of Lübeck, Lübeck, Germany
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Side effects of zolendronic acid (ZA) and RANKL inhibitors (RANKL-I) include impaired wound healing and osteonecrosis of the jaw. Platelet rich fibrin (PRF) enhances wound healing and bone remodelling in vivo and in vitro. However, the topical use PRF in the surgical treatment of patients with medicament-related osteonecrosis of the jaw is relatively new and not thoroughly investigated. Furthermore, the potential attenuation of the PRF effect following antiresorptive treatment remains unclear. Therefore, we investigated the concentration of growth factors within the PRF in healthy volunteers and in patients with antiresorptive treatment. Patients and Methods: Blood samples from healthy volunteers and patients were used to produce PRF. The levels of EGF, VEGF, PDGF-BB, TGF-β1, BMP-2, and CD31 in the PRF was investigated by ELISA. Results: ZA treatment induced a significant decrease in EGF and TGF-β1 levels, whereas RANKL-I caused lower TGF-β1 levels. Conclusion: Reduced EGF levels in PRF after ZA treatment may explain the delayed wound healing and question the positive effect of PRF in these patients. PRF use in patients undergoing RANKL-I treatment seems to be more justified.

Key Words:
  • Platelet rich fibrin
  • zoledronic acid
  • denosumab
  • growth factor
  • bisphosphonates
  • denosumab

Bone metabolism disorders such as osteoporosis as well as bone metastases are usually treated by bisphosphonates or RANKL-inhibitors (1-4). These drugs influence the interaction, differentiation and function of osteoclasts and osteoblasts via different mechanisms (5). A main side effect of these substances is medicament-related osteonecrosis of the jaw (MRONJ) that mostly occurs after dentoalveolar intervention and is associated with prolonged wound healing (6). Several studies have shown that autologous platelet rich fibrin (PRF) supports wound healing due to high concentrations of growth factors such as PDGF-BB, TGF-β1, VEGF, EGF, CD31, and Bmp2 in non-MRONJ patients (7-12). PDGF is a powerful chemotactic stimulus, TGF-β1 is a regulatory protein involved in bone remodelling and fracture healing whereas VEGF supports bone healing by promoting angiogenesis (13-15). EGF also promotes bone formation and shows a positive effect on epithelial wound closure (16-18), whereas CD31 functions as a negative regulator of osteoclastogenesis, and BMP-2 plays a key role in bone remodelling, especially in fracture healing (19-21).

While patients with MRONJ are thought to benefit from local PRF treatment, related studies assessed disease stage-dependent response to this treatment in patients undergoing anti-resorptive therapy (10, 22, 23).

To evaluate the value of PRF treatment in MRONJ patients, it is mandatory to investigate the effect of bisphosphonates and RANKL-Inhibitors on the quality of PRF in these patients in comparison to healthy population. The aim of this study was to investigate growth factor expression as a surrogate parameter for the wound and bone healing potential of PRF under the influence of zoledronic acid (ZA) or Denosumab therapy compared to PRF of healthy volunteers.

Patients and Methods

Patients. Blood samples were obtained from 10 patients treated with Denosumab and from further 10 patients treated with zoledronic acid. Patients with anticoagulative therapy or dialysis were not included due to potential impact on thrombocyte number and/or function. Ten further healthy volunteers, without any medication, provided control blood samples. Detailed patient characteristics are given in Table I.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

Characteristics of patients included in the study.

All procedures were conducted according to the ethical standards of the Declaration of Helsinki and were approved by the institutional ethical committee of the University of Lübeck, Germany.

Preparation of PRF. A standard vein puncture (median basilica vein, median cubital vein, and median cephalic vein) was performed to prepare PRF according to a standard protocol as previously described (24). Ten ml of blood was drawn into a tube and immediately centrifuged at 145 g for 8 min (DUO Quattro; A-PRF Mectron, Carasco, Italy) (8, 25, 26). After centrifugation, the PRF clot was removed and transferred into a new tube and frozen at −80˚C until further investigations were performed.

Growth factor quantification by ELISA test. Before evaluation of the different growth factors (GF) of PRF, samples were thawed and centrifugated at 408 g for 20 min at 18˚C as previously described (24). The levels of the growth factors TGF-β1, PDGF-BB, CD31/PECAM-1, VEGF, EGF, and BMP-2 were determined in the supernatant. The expression of growth factors was assessed using enzyme-linked immunosorbent assay tests (ELISA; R&D Systems, Minneapolis, MN, USA) and conducted according to the manufacturers’ manual. Measurements were performed with benchmark plus microplate spectrophotometer set to 540 nm (Clariostar, BMG-Labtech, Ortenberg, Germany). Each ELISA sample was run in duplicate.

Statistical evaluation. Statistical analysis was performed using the statistical package IBM SPSS Statistics version 26 (IBM, Armonk, NY, USA). Values were expressed as mean±standard deviation. Differences were assessed using a t-test for unpaired samples as well as the Pearson correlation test. Results were considered significant when p≤0.05.

Results

Generally, the patients of the Denosumab group showed a significant lower number of platelets compared to the control (plateletscontrol: 258×109/l; plateletsdenosumab 176×109/l; p=0.02) but no difference in the number of leucocytes (leucocytescontrol: 6.7×109/l; leucocytesdenosumab 6.1×109/l) was observed. On the contrary, the mean values of platelets and leucocytes of patients treated with zoledronic acid showed no significant differences among the groups (plateletscontrol: 258×109/l; plateletszoledronic acid 254×109/l; leucocytes 6.7×109/l in both).

Impact of platelet, leucocyte number, and Denosumab and ZA treatment period on growth factor expression in PRF. Zoledronic acid treatment resulted in a significant decrease of EGF concentration in PRF in comparison to control and denosumab treatment [ZA: 391±236 pg/ml control: 658±301 pg/ml (p=0.02); denosumab: 729±427 pg/ml (p=0.03)] (Figure 1C), while TGF-beta1 concentration was significantly lower in both zoledronic acid and denosumab groups compared to the control [control: 715±168 pg/ml ZA: 608±117 pg/ml (p=0.05); denosumab: 490±119 pg/ml (p<0.01)] (Figure 1E). All other growth factor concentrations remained unchanged within the different groups (Figure 1A-F).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Results of the ELISA assays showing the concentration of BMP2 (A), CD31 (B), EGF (C), PDGF (D), TGF (E), and VEGF (F) in platelet rich fibrin (PRF) of healthy volunteers in comparison to patients treated with zoledronic acid and denosumab.

Interestingly, we could not observe any correlation between the number of platelets or leucocytes with the concentration of growth factors in PRF in the different treatment groups and the control (Figure 2A-F). Similarly, the treatment time had no significant impact on growth factor concentration in PRF in both treatment groups (Figure 2H-I).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Correlation of the number of platelets, leucocytes as well as of treatment duration of the different groups with the concentration of BMP2, CD31, EGF, PDGF, TGF, and VEGF in their platelet rich fibrin (PRF). No significant correlation was found.

Discussion

Drugs affecting bone metabolism such as zoledronic acid and RANKL-Inhibitors can lead to osteonecrosis of the jaw since they influence bone formation and turn-over, resulting in bone exposure with wound dehiscence, fistulas and chronic inflammation (27). Due to its high concentration of growth factors, PRF supports wound and bone healing (7-12). The use of PRF is conceivable for the treatment of MRONJ, but its effectiveness as local therapy in these patients cannot be definitively proven unless the crucial levels of growth factors in PRF under antiresorptive treatment have been assessed (28-30). Therefore, the aim of this study was to investigate PRF quality in patients undergoing ZA and RANKL-I treatment compared to control population.

Patients of the ZA group had a comparable platelet and leucocyte number to those in the control group, a finding that justifies the comparison of growth factor concentration in the PRF of both groups. On the contrast, patients in the RANKL-I group displayed significantly lower number of platelets whereas the number of leucocytes was not affected This result is in accordance with Weibrich et al. (31) who reported a low correlation between the baseline platelet number from whole blood and related growth factor levels. However, it is known that reduction of the centrifugal force during preparation of PRF results in higher cell number and increased growth factor release (32).

When the concentration of individual growth factors was assessed, a significant decrease of EGF in the ZA group compared to the RANKL-I and control group was found. Given the known impact of EGF on epithelial migration and thus on wound healing, the toxic effect of ZA on the oral mucosa and the disturbance in the subsequent wound healing can be understood. This effect has been also shown in vitro using human oral keratinocytes; ZA treatment resulted in reduced viability, impaired migration ability, and increased apoptosis rate (33-35). Reversible effects were also demonstrated by EGF stimulation (35, 36).

Hypothetically, the observed reduction of EGF in ZA patients may reflect the known EGFR-based anti-neoplastic effect of ZA, which inhibits cancer cell proliferation directly and modulates stroma cells in the tumor microenvironment (37, 38). In other words, the decreased concentration of EGF in PRF during ZA treatment is a sign of its anti-neoplastic effect.

TGFβ-1 concentration was reduced in both ZA and RANKL-I groups compared to the control. Recent studies demonstrated that ZA intervenes with the TGFβ-signalling pathway and - especially in low drug concentration - inhibits TGF expression and related wound closure, possibly through suppression of Smad2/3 signalling (39).

Similar to EGF, the pro-migratory transforming growth factor TGF-β1 is considered a key factor contributing to tumor progression (40) since the blockade of TGF-β signalling has been shown to effectively prevent osteolytic bone metastasis (41). This emphasizes the antineoplastic effect of ZA and indicates that patients undergoing this therapy may carry the humoral risk of disturbed wound healing (42-44).

There is a crucial lack of information on the impact of ZA and RANKL-I therapy on wound healing in MRONJ patients (45). However, along with the results presented in the current study, a reasonable difference in the composition and certainly in the potential regenerative effect of PRF from ZA and RANKL-I patients should be postulated.

Future studies should investigate this feature in a therapeutic context and should consider application of immunologically neutral allogeneic PRF in the surgical treatment of MRONJ patients.

Conclusion

While the concentration of PDGF, CD31, VEGF, and BMP-2 showed no significant differences in PRF from ZA and RANKL-I patients, EGF and TGF in PRF from ZA-treated patients as well as TGF from RANKL-I-treated patients were significantly reduced, indicating possible negative effects on the regenerative quality of their PRF.

Footnotes

  • Authors’ Contributions

    D. Steller and S.G. Hakim designed the study and wrote the main manuscript. R. Simon performed the measurements and described the results. R. von Bialy designed the Figures. R. Simon and D. Steller equally contributed to the study.

  • Conflicts of Interest

    The Authors declare that they have no competing interests in relation to this study.

  • Received April 28, 2021.
  • Revision received July 2, 2021.
  • Accepted July 5, 2021.
  • Copyright © 2021 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.

References

  1. ↵
    1. Almubarak H,
    2. Jones A,
    3. Chaisuparat R,
    4. Zhang M,
    5. Meiller TF and
    6. Scheper MA
    : Zoledronic acid directly suppresses cell proliferation and induces apoptosis in highly tumorigenic prostate and breast cancers. J Carcinog 10: 2, 2011. PMID: 21297922. DOI: 10.4103/1477-3163.75723
    OpenUrlCrossRefPubMed
    1. Coleman RE,
    2. Winter MC,
    3. Cameron D,
    4. Bell R,
    5. Dodwell D,
    6. Keane MM,
    7. Gil M,
    8. Ritchie D,
    9. Passos-Coelho JL,
    10. Wheatley D,
    11. Burkinshaw R,
    12. Marshall SJ,
    13. Thorpe H and AZURE (BIG01/04) Investigators
    : The effects of adding zoledronic acid to neoadjuvant chemotherapy on tumour response: exploratory evidence for direct anti-tumour activity in breast cancer. Br J Cancer 102(7): 1099-1105, 2010. PMID: 20234364. DOI: 10.1038/sj.bjc.6605604
    OpenUrlCrossRefPubMed
    1. Cummings SR,
    2. San Martin J,
    3. McClung MR,
    4. Siris ES,
    5. Eastell R,
    6. Reid IR,
    7. Delmas P,
    8. Zoog HB,
    9. Austin M,
    10. Wang A,
    11. Kutilek S,
    12. Adami S,
    13. Zanchetta J,
    14. Libanati C,
    15. Siddhanti S,
    16. Christiansen C and FREEDOM Trial
    : Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med 361(8): 756-765, 2009. PMID: 19671655. DOI: 10.1056/NEJMoa0809493
    OpenUrlCrossRefPubMed
  2. ↵
    1. Lipton A,
    2. Steger GG,
    3. Figueroa J,
    4. Alvarado C,
    5. Solal-Celigny P,
    6. Body JJ,
    7. de Boer R,
    8. Berardi R,
    9. Gascon P,
    10. Tonkin KS,
    11. Coleman RE,
    12. Paterson AH,
    13. Gao GM,
    14. Kinsey AC,
    15. Peterson MC and
    16. Jun S
    : Extended efficacy and safety of denosumab in breast cancer patients with bone metastases not receiving prior bisphosphonate therapy. Clin Cancer Res 14(20): 6690-6696, 2008. PMID: 18927312. DOI: 10.1158/1078-0432.CCR-07-5234
    OpenUrlAbstract/FREE Full Text
  3. ↵
    1. Baron R,
    2. Ferrari S and
    3. Russell RG
    : Denosumab and bisphosphonates: different mechanisms of action and effects. Bone 48(4): 677-692, 2011. PMID: 21145999. DOI: 10.1016/j.bone.2010.11.020
    OpenUrlCrossRefPubMed
  4. ↵
    1. Filleul O,
    2. Crompot E and
    3. Saussez S
    : Bisphosphonate-induced osteonecrosis of the jaw: a review of 2,400 patient cases. J Cancer Res Clin Oncol 136(8): 1117-1124, 2010. PMID: 20508948. DOI: 10.1007/s00432-010-0907-7
    OpenUrlCrossRefPubMed
  5. ↵
    1. Anitua E,
    2. Tejero R,
    3. Alkhraisat MH and
    4. Orive G
    : Platelet-rich plasma to improve the bio-functionality of biomaterials. BioDrugs 27(2): 97-111, 2013. PMID: 23329397. DOI: 10.1007/s40259-012-0004-3
    OpenUrlCrossRefPubMed
  6. ↵
    1. Fujioka-Kobayashi M,
    2. Miron RJ,
    3. Hernandez M,
    4. Kandalam U,
    5. Zhang Y and
    6. Choukroun J
    : Optimized platelet-rich fibrin with the low-speed concept: Growth factor release, biocompatibility, and cellular response. J Periodontol 88(1): 112-121, 2017. PMID: 27587367. DOI: 10.1902/jop.2016.160443
    OpenUrlCrossRefPubMed
    1. Herford AS,
    2. Miller M and
    3. Signorino F
    : Maxillofacial defects and the use of growth factors. Oral Maxillofac Surg Clin North Am 29(1): 75-88, 2017. PMID: 27890229. DOI: 10.1016/j.coms.2016.08.006
    OpenUrlCrossRefPubMed
  7. ↵
    1. Miron RJ,
    2. Fujioka-Kobayashi M,
    3. Bishara M,
    4. Zhang Y,
    5. Hernandez M and
    6. Choukroun J
    : Platelet-rich fibrin and soft tissue wound healing: A systematic review. Tissue Eng Part B Rev 23(1): 83-99, 2017. PMID: 27672729. DOI: 10.1089/ten.TEB.2016.0233
    OpenUrlCrossRefPubMed
    1. Zimmermann R,
    2. Jakubietz R,
    3. Jakubietz M,
    4. Strasser E,
    5. Schlegel A,
    6. Wiltfang J and
    7. Eckstein R
    : Different preparation methods to obtain platelet components as a source of growth factors for local application. Transfusion 41(10): 1217-1224, 2001. PMID: 11606819. DOI: 10.1046/j.1537-2995.2001.41101217.x
    OpenUrlCrossRefPubMed
  8. ↵
    1. Agren MS,
    2. Rasmussen K,
    3. Pakkenberg B and
    4. Jørgensen B
    : Growth factor and proteinase profile of Vivostat® platelet-rich fibrin linked to tissue repair. Vox Sang 107(1): 37-43, 2014. PMID: 24320875. DOI: 10.1111/vox.12120
    OpenUrlCrossRefPubMed
  9. ↵
    1. Fiedler J,
    2. Etzel N and
    3. Brenner RE
    : To go or not to go: Migration of human mesenchymal progenitor cells stimulated by isoforms of PDGF. J Cell Biochem 93(5): 990-998, 2004. PMID: 15389881. DOI: 10.1002/jcb.20219
    OpenUrlCrossRefPubMed
    1. Joyce ME,
    2. Jingushi S and
    3. Bolander ME
    : Transforming growth factor-beta in the regulation of fracture repair. Orthop Clin North Am 21(1): 199-209, 1990. PMID: 2296458.
    OpenUrlPubMed
  10. ↵
    1. Street J,
    2. Bao M,
    3. deGuzman L,
    4. Bunting S,
    5. Peale FV Jr.,
    6. Ferrara N,
    7. Steinmetz H,
    8. Hoeffel J,
    9. Cleland JL,
    10. Daugherty A,
    11. van Bruggen N,
    12. Redmond HP,
    13. Carano RA and
    14. Filvaroff EH
    : Vascular endothelial growth factor stimulates bone repair by promoting angiogenesis and bone turnover. Proc Natl Acad Sci USA 99(15): 9656-9661, 2002. PMID: 12118119. DOI: 10.1073/pnas.152324099
    OpenUrlAbstract/FREE Full Text
  11. ↵
    1. Chim SM,
    2. Tickner J,
    3. Chow ST,
    4. Kuek V,
    5. Guo B,
    6. Zhang G,
    7. Rosen V,
    8. Erber W and
    9. Xu J
    : Angiogenic factors in bone local environment. Cytokine Growth Factor Rev 24(3): 297-310, 2013. PMID: 23611723. DOI: 10.1016/j.cytogfr.2013.03.008
    OpenUrlCrossRefPubMed
    1. Falanga V,
    2. Eaglstein WH,
    3. Bucalo B,
    4. Katz MH,
    5. Harris B and
    6. Carson P
    : Topical use of human recombinant epidermal growth factor (h-EGF) in venous ulcers. J Dermatol Surg Oncol 18(7): 604-606, 1992. PMID: 1624634. DOI: 10.1111/j.1524-4725.1992.tb03514.x
    OpenUrlCrossRefPubMed
  12. ↵
    1. Zhang X,
    2. Tamasi J,
    3. Lu X,
    4. Zhu J,
    5. Chen H,
    6. Tian X,
    7. Lee TC,
    8. Threadgill DW,
    9. Kream BE,
    10. Kang Y,
    11. Partridge NC and
    12. Qin L
    : Epidermal growth factor receptor plays an anabolic role in bone metabolism in vivo. J Bone Miner Res 26(5): 1022-1034, 2011. PMID: 21542005. DOI: 10.1002/jbmr.295
    OpenUrlCrossRefPubMed
  13. ↵
    1. Onishi T,
    2. Ishidou Y,
    3. Nagamine T,
    4. Yone K,
    5. Imamura T,
    6. Kato M,
    7. Sampath TK,
    8. ten Dijke P and
    9. Sakou T
    : Distinct and overlapping patterns of localization of bone morphogenetic protein (BMP) family members and a BMP type II receptor during fracture healing in rats. Bone 22(6): 605-612, 1998. PMID: 9626398. DOI: 10.1016/s8756-3282(98)00056-8
    OpenUrlCrossRefPubMed
    1. Tsuji K,
    2. Bandyopadhyay A,
    3. Harfe BD,
    4. Cox K,
    5. Kakar S,
    6. Gerstenfeld L,
    7. Einhorn T,
    8. Tabin CJ and
    9. Rosen V
    : BMP2 activity, although dispensable for bone formation, is required for the initiation of fracture healing. Nat Genet 38(12): 1424-1429, 2006. PMID: 17099713. DOI: 10.1038/ng1916
    OpenUrlCrossRefPubMed
  14. ↵
    1. Wu Y and
    2. Madri J
    : Insights into monocyte-driven osteoclastogenesis and its link with hematopoiesis: regulatory roles of PECAM-1 (CD31) and SHP-1. Crit Rev Immunol 30(5): 423-433, 2010. PMID: 21083524. DOI: 10.1615/critrevimmunol.v30.i5.20
    OpenUrlCrossRefPubMed
  15. ↵
    1. Del Fabbro M,
    2. Gallesio G and
    3. Mozzati M
    : Autologous platelet concentrates for bisphosphonate-related osteonecrosis of the jaw treatment and prevention. A systematic review of the literature. Eur J Cancer 51(1): 62-74, 2015. PMID: 25466505. DOI: 10.1016/j.ejca.2014.10.015
    OpenUrlCrossRefPubMed
  16. ↵
    1. Giudice A,
    2. Barone S,
    3. Giudice C,
    4. Bennardo F and
    5. Fortunato L
    : Can platelet-rich fibrin improve healing after surgical treatment of medication-related osteonecrosis of the jaw? A pilot study. Oral Surg Oral Med Oral Pathol Oral Radiol 126(5): 390-403, 2018. PMID: 30108028. DOI: 10.1016/j.oooo.2018.06.007
    OpenUrlCrossRefPubMed
  17. ↵
    1. Steller D,
    2. Herbst N,
    3. Pries R,
    4. Juhl D and
    5. Hakim SG
    : Impact of incubation method on the release of growth factors in non-Ca2+-activated PRP, Ca2+-activated PRP, PRF and A-PRF. J Craniomaxillofac Surg 47(2): 365-372, 2019. PMID: 30578012. DOI: 10.1016/j.jcms.2018.10.017
    OpenUrlCrossRefPubMed
  18. ↵
    1. Ghanaati S,
    2. Booms P,
    3. Orlowska A,
    4. Kubesch A,
    5. Lorenz J,
    6. Rutkowski J,
    7. Landes C,
    8. Sader R,
    9. Kirkpatrick C and
    10. Choukroun J
    : Advanced platelet-rich fibrin: a new concept for cell-based tissue engineering by means of inflammatory cells. J Oral Implantol 40(6): 679-689, 2014. PMID: 24945603. DOI: 10.1563/aaid-joi-D-14-00138
    OpenUrlCrossRefPubMed
  19. ↵
    1. Choukroun J and
    2. Ghanaati S
    : Reduction of relative centrifugation force within injectable platelet-rich-fibrin (PRF) concentrates advances patients’ own inflammatory cells, platelets and growth factors: the first introduction to the low speed centrifugation concept. Eur J Trauma Emerg Surg 44(1): 87-95, 2018. PMID: 28283682. DOI: 10.1007/s00068-017-0767-9
    OpenUrlCrossRefPubMed
  20. ↵
    1. Migliorati CA,
    2. Siegel MA and
    3. Elting LS
    : Bisphosphonate-associated osteonecrosis: a long-term complication of bisphosphonate treatment. Lancet Oncol 7(6): 508-514, 2006. PMID: 16750501. DOI: 10.1016/S1470-2045(06)70726-4
    OpenUrlCrossRefPubMed
  21. ↵
    1. Kim JW,
    2. Kim SJ and
    3. Kim MR
    : Leucocyte-rich and platelet-rich fibrin for the treatment of bisphosphonate-related osteonecrosis of the jaw: a prospective feasibility study. Br J Oral Maxillofac Surg 52(9): 854-859, 2014. PMID: 25138613. DOI: 10.1016/j.bjoms.2014.07.256
    OpenUrlCrossRefPubMed
    1. Nørholt SE and
    2. Hartlev J
    : Surgical treatment of osteonecrosis of the jaw with the use of platelet-rich fibrin: a prospective study of 15 patients. Int J Oral Maxillofac Surg 45(10): 1256-1260, 2016. PMID: 27179556. DOI: 10.1016/j.ijom.2016.04.010
    OpenUrlCrossRefPubMed
  22. ↵
    1. Fortunato L,
    2. Bennardo F,
    3. Buffone C and
    4. Giudice A
    : Is the application of platelet concentrates effective in the prevention and treatment of medication-related osteonecrosis of the jaw? A systematic review. J Craniomaxillofac Surg 48(3): 268-285, 2020. PMID: 32063481. DOI: 10.1016/j.jcms.2020.01.014
    OpenUrlCrossRefPubMed
  23. ↵
    1. Weibrich G,
    2. Kleis WK,
    3. Hafner G and
    4. Hitzler WE
    : Growth factor levels in platelet-rich plasma and correlations with donor age, sex, and platelet count. J Craniomaxillofac Surg 30(2): 97-102, 2002. PMID: 12069512. DOI: 10.1054/jcms.2002.0285
    OpenUrlCrossRefPubMed
  24. ↵
    1. Wend S,
    2. Kubesch A,
    3. Orlowska A,
    4. Al-Maawi S,
    5. Zender N,
    6. Dias A,
    7. Miron RJ,
    8. Sader R,
    9. Booms P,
    10. Kirkpatrick CJ,
    11. Choukroun J and
    12. Ghanaati S
    : Reduction of the relative centrifugal force influences cell number and growth factor release within injectable PRF-based matrices. J Mater Sci Mater Med 28(12): 188, 2017. PMID: 29071440. DOI: 10.1007/s10856-017-5992-6
    OpenUrlCrossRefPubMed
  25. ↵
    1. Steller D,
    2. Herbst N,
    3. Pries R,
    4. Juhl D and
    5. Hakim SG
    : Positive impact of Platelet-rich plasma and Platelet-rich fibrin on viability, migration and proliferation of osteoblasts and fibroblasts treated with zoledronic acid. Sci Rep 9(1): 8310, 2019. PMID: 31165745. DOI: 10.1038/s41598-019-43798-z
    OpenUrlCrossRefPubMed
    1. Kim EH,
    2. Kim MS,
    3. Lee KH,
    4. Koh JS,
    5. Jung WG and
    6. Kong CB
    : Zoledronic acid is an effective radiosensitizer in the treatment of osteosarcoma. Oncotarget 7(43): 70869-70880, 2016. PMID: 27765919. DOI: 10.18632/oncotarget.12281
    OpenUrlCrossRefPubMed
  26. ↵
    1. Wang Q,
    2. Liu J,
    3. Guo T,
    4. Liu D and
    5. Pan J
    : Epidermal growth factor reverses the inhibitory effects of the bisphosphonate, zoledronic acid, on human oral keratinocytes and human vascular endothelial cells in vitro via the epidermal growth factor receptor (EGFR)/Akt/phosphoinositide 3-kinase (PI3K) signaling pathway. Med Sci Monit 25: 700-710, 2019. PMID: 30675875. DOI: 10.12659/MSM.911579
    OpenUrlCrossRefPubMed
  27. ↵
    1. Pabst AM,
    2. Krüger M,
    3. Sagheb K,
    4. Ziebart T,
    5. Jacobs C,
    6. Blatt S,
    7. Goetze E and
    8. Walter C
    : The influence of geranylgeraniol on microvessel sprouting after bisphosphonate substitution in an in vitro 3D-angiogenesis assay. Clin Oral Investig 21(3): 771-778, 2017. PMID: 27170294. DOI: 10.1007/s00784-016-1842-z
    OpenUrlCrossRefPubMed
  28. ↵
    1. Irwin ME,
    2. Mueller KL,
    3. Bohin N,
    4. Ge Y and
    5. Boerner JL
    : Lipid raft localization of EGFR alters the response of cancer cells to the EGFR tyrosine kinase inhibitor gefitinib. J Cell Physiol 226(9): 2316-2328, 2011. PMID: 21660955. DOI: 10.1002/jcp.22570
    OpenUrlCrossRefPubMed
  29. ↵
    1. Lu X and
    2. Kang Y
    : Epidermal growth factor signalling and bone metastasis. Br J Cancer 102(3): 457-461, 2010. PMID: 20010942. DOI: 10.1038/sj.bjc.6605490
    OpenUrlCrossRefPubMed
  30. ↵
    1. Zhao Z,
    2. Shen W,
    3. Zhu H,
    4. Lin L,
    5. Jiang G,
    6. Zhu Y,
    7. Song H and
    8. Wu L
    : Zoledronate inhibits fibroblasts’ proliferation and activation via targeting TGF-β signaling pathway. Drug Des Devel Ther 12: 3021-3031, 2018. PMID: 30271117. DOI: 10.2147/DDDT.S168897
    OpenUrlCrossRefPubMed
  31. ↵
    1. Alsina-Sanchís E,
    2. Figueras A,
    3. Lahiguera A,
    4. Gil-Martín M,
    5. Pardo B,
    6. Piulats JM,
    7. Martí L,
    8. Ponce J,
    9. Matias-Guiu X,
    10. Vidal A,
    11. Villanueva A and
    12. Viñals F
    : TGFβ controls ovarian cancer cell proliferation. Int J Mol Sci 18(8): 1658, 2017. PMID: 28758950. DOI: 10.3390/ijms18081658
    OpenUrlCrossRefPubMed
  32. ↵
    1. Juárez P,
    2. Fournier PGJ,
    3. Mohammad KS,
    4. McKenna RC,
    5. Davis HW,
    6. Peng XH,
    7. Niewolna M,
    8. Mauviel A,
    9. Chirgwin JM and
    10. Guise TA
    : Halofuginone inhibits TGF-β/BMP signaling and in combination with zoledronic acid enhances inhibition of breast cancer bone metastasis. Oncotarget 8(49): 86447-86462, 2017. PMID: 29156807. DOI: 10.18632/oncotarget.21200
    OpenUrlCrossRefPubMed
  33. ↵
    1. Hu L,
    2. Wen Y,
    3. Xu J,
    4. Wu T,
    5. Zhang C,
    6. Wang J,
    7. Du J and
    8. Wang S
    : Pretreatment with bisphosphonate enhances osteogenesis of bone marrow mesenchymal stem cells. Stem Cells Dev 26(2): 123-132, 2017. PMID: 27736364. DOI: 10.1089/scd.2016.0173
    OpenUrlCrossRefPubMed
    1. Manzano-Moreno FJ,
    2. Ramos-Torrecillas J,
    3. Melguizo-Rodríguez L,
    4. Illescas-Montes R,
    5. Ruiz C and
    6. García-Martínez O
    : Bisphosphonate modulation of the gene expression of different markers involved in osteoblast physiology: Possible implications in bisphosphonate-related osteonecrosis of the jaw. Int J Med Sci 15(4): 359-367, 2018. PMID: 29511371. DOI: 10.7150/ijms.22627
    OpenUrlCrossRefPubMed
  34. ↵
    1. Naidu A,
    2. Dechow PC,
    3. Spears R,
    4. Wright JM,
    5. Kessler HP and
    6. Opperman LA
    : The effects of bisphosphonates on osteoblasts in vitro. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 106(1): 5-13, 2008. PMID: 18504149. DOI: 10.1016/j.tripleo.2008.03.036
    OpenUrlCrossRefPubMed
  35. ↵
    1. Komatsu Y,
    2. Ibi M,
    3. Chosa N,
    4. Kyakumoto S,
    5. Kamo M,
    6. Shibata T,
    7. Sugiyama Y and
    8. Ishisaki A
    : Zoledronic acid suppresses transforming growth factor-β-induced fibrogenesis by human gingival fibroblasts. Int J Mol Med 38(1): 139-147, 2016. PMID: 27176567. DOI: 10.3892/ijmm.2016.2582
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Anticancer Research
Vol. 41, Issue 8
August 2021
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Impact of Zoledronic Acid and Denosumab Treatment on Growth Factor Concentration in Platelet Rich Fibrin of Patients With Osteolytic Bone Metastases
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
15 + 1 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Impact of Zoledronic Acid and Denosumab Treatment on Growth Factor Concentration in Platelet Rich Fibrin of Patients With Osteolytic Bone Metastases
DANIEL STELLER, RONJA SIMON, ROBERT VON BIALY, RALPH PRIES, SAMER G. HAKIM
Anticancer Research Aug 2021, 41 (8) 3917-3923; DOI: 10.21873/anticanres.15187

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Impact of Zoledronic Acid and Denosumab Treatment on Growth Factor Concentration in Platelet Rich Fibrin of Patients With Osteolytic Bone Metastases
DANIEL STELLER, RONJA SIMON, ROBERT VON BIALY, RALPH PRIES, SAMER G. HAKIM
Anticancer Research Aug 2021, 41 (8) 3917-3923; DOI: 10.21873/anticanres.15187
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Patients and Methods
    • Results
    • Discussion
    • Conclusion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Efficacy and Safety of Monthly Minodronate Therapy in Postmenopausal Breast Cancer Patients Receiving Aromatase Inhibitors
  • Google Scholar

More in this TOC Section

  • The Posterior First Approach in Robot-assisted Radical Prostatectomy for Prostate Cancer Reduces Positive Surgical Margins on the Bladder Neck Side
  • Gamma Knife Radiotherapy of Brain Metastasis Resection Cavities: Outcome Analysis of a Single-center Cohort
  • Efficacy and Safety of Chemoimmunotherapy in Patients With Advanced Non-small Cell Lung Cancer With Pre-existing Interstitial Pneumonia and Low PD-L1 Expression
Show more Clinical Studies

Similar Articles

Keywords

  • Platelet rich fibrin
  • zoledronic acid
  • denosumab
  • growth factor
  • bisphosphonates
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire