Expression of high mobility group box 1 protein predicts a poorer prognosis for patients with osteosarcoma

  • Authors:
    • Jiliang He
    • Peng Zhang
    • Qinghu Li
    • Dongsheng Zhou
    • Ping Liu
  • View Affiliations

  • Published online on: November 10, 2015     https://doi.org/10.3892/ol.2015.3907
  • Pages: 293-298
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The high mobility group box 1 (HMGB1) protein functions as an extracellular signaling molecule that is critical in inflammation and carcinogenesis. The HMGB1 protein is actively secreted by natural killer cells, monocytes and macrophages, and acts as an inflammatory cytokine. The present study enrolled 174 patients that underwent a tumorectomy between 2006 and 2013 in Shandong Provincial Hospital. The age of the patients ranged between 13 and 74 years, with a median age of 27 years. The tumors of the patients were staged according to the Union for International Cancer Control 2009 tumor‑node‑metastasis tumor staging system. Nuclear grading was based on the Fuhrman grading system. In the osteosarcoma tissue samples, HMGB1 expression was detected in 84 samples (48.3%) with a low immunoreactivity and in 90 samples (51.7%) with a high immunoreactivity. The association between clinicopathological characteristics and tumor cell HMGB1 expression (low vs. high) was summarized. The association between HMGB1 expression and tumor size, tumor stage and nuclear grade was statistically significant (P=0.034, 0.008 and 0.019, respectively). There was no significant association between HMGB1 expression and the age of the patients (P=0.335; Table I). The current study demonstrated that patients with a high HMGB1 expression (>50% cells expressing HMGB1) had poorer survival rates, and therefore a poorer prognosis, compared with patients with low HMGB1 immunostaining (10‑50% cells expressing HMGB1). The results of the present study suggest that higher expression levels of HMGB1 are significantly associated with a poorer prognosis and may act as a marker for prognosis in osteosarcoma, particularly osteosarcoma recurrence. Additional studies investigating the biological features of HMGB1 may confirm the potential role of HMGB1 as a novel target for anticancer therapy in osteosarcoma.

Introduction

Osteosarcoma accounts for ~20% of pediatric, solid and malignant tumors, and it is the most common malignant bone tumor in adolescents and young adults (1). In total, ~40% of osteosarcoma tumors metastasize and patients possess a poor overall prognosis (24). The 5-year survival rate is reported to be 37% in osteosarcoma patients with tumor metastasis and 19% for patients with >5 metastatic lung lesions (5). Uncontrolled cell proliferation and robust tumor angiogenesis are characteristics that contribute to the poor prognosis of osteosarcoma patients (6). Aggressive osteosarcoma cells demonstrate increased angiogenesis and vasculogenic mimicry (7). Several studies have revealed that tumor cells may directly form tumor blood vessels through vasculogenic mimicry (813). This is closely associated with tumor metastasis and the poor prognosis of various cancers, including osteosarcoma (11,12). Consequently, highly proliferative and vasculogenic osteosarcoma cells are a clear target for novel anti-osteosarcoma drug identification.

The high mobility group box 1 (HMGB1) protein was originally described as a non-histone DNA binding protein that is highly conserved, with a high acidic and basic amino acid content (14). The HMGB1 protein binds to minor grooves in DNA and promotes the assembly of site-specific transcriptional proteins (15). In addition to its well-established nuclear functions, the HMGB1 protein acts as an extracellular signaling molecule that is critical in inflammation and carcinogenesis (16,17). The HMGB1 protein is actively secreted by natural killer cells, monocytes and macrophages, and functions as an inflammatory cytokine (18,19). Necrotic cells, particularly those derived from cancer tissues, passively release the HMGB1 protein, which mediates local inflammation and the development of cancer (16,20). Extracellular HMGB1 protein interacts with several receptors, including the receptor for advanced glycation end products (RAGE), Toll-like receptors and cluster of differentiation (CD)24 (21). It has been reported that HMGB1 and RAGE are overexpressed in clear cell renal cell carcinoma (ccRCC). In addition, HMGB1 promotes the development and progression of ccRCC via extracellular-signal-regulated kinase 1/2 activation, which is partially mediated by RAGE (22).

Although the expression of HMGB1 has been associated with the prognosis of urinary tumors, including bladder urothelial carcinoma (23), no studies have investigated the prognostic role of HMGB1 in osteosarcoma, such as the association between HMGB1 expression and the prognosis of osteosarcoma. In the present study, the expression of HMGB1 in clinical osteosarcoma samples, prognostic role of HMGB1, and the association between HMGB1 expression and clinicopathological features were investigated, with the aim of identifying HMGB1 as a prognostic marker in patients with osteosarcoma.

Materials and methods

Patients and tissue specimens

The present study assessed 174 patients who underwent tumorectomies between 2006 and 2013 at Shandong Provincial Hospital (Jinan, Shandong, China). The age of patients ranged between 13 and 74 years, with a median age of 27 years. The tumors of the patients were staged according to the Union for International Cancer Control 2009 tumor-node-metastasis (TNM) tumor staging system (24), and nuclear grading was based on the Fuhrman grading system (25). The follow-up time was calculated from the date of surgery to the date of the patient's last follow-up or mortality. Patients were excluded if they had incomplete medical records or inadequate follow-up. Tissue samples were obtained from the surgical samples of patients with osteosarcoma. The specimens were fixed in 4% buffered formalin and embedded in paraffin for analysis. The present study was approved by the ethical committees of Shandong Provincial Hospital.

Immunohistochemistry (IHC)

Sections of formalin-fixed, paraffin-embedded osteosarcoma tissue samples were cut to 4 µm thick slides in the Department of Pathology of Shandong Provincial Hospital. The slides were deparaffinized in an oven at 60°C for 2 h and maintained in xylene (Jining Huakai Resin Co., Ltd., Jining, China) for 20 min. The slides were then rehydrated in graded ethanol (100% for 5 min, 95% for 5 min and 75% for 5 min; (Jining Huakai Resin Co., Ltd.) and washed with 0.01 M phosphate-buffered saline (PBS; pH 7; Boster Biologics, Pleasanton, CA, USA) three times for 5 min each. Antigen retrieval was performed using high-pressure heating of the samples in a 0.01 M sodium citrate buffer at 95°C for 10 min. The slides were washed three times with PBS, and 3% H2O2 (Jiangmen Hengjian Pharmaceutical Co., Ltd., Jiangmen, China) was added for 30 min at 37°C to quench endogenous peroxidase activity. Subsequently, the slides were blocked with 10% goat serum (Beijing Jorferin Biotechnology Co., Ltd., Beijing, China) for 30 min at 37°C, followed by incubation with the rabbit anti-human polyclonal primary antibody targeting HMGB1 (cat no. ab18256; Abcam, Cambridge, UK), which was diluted to 1:100 with PBS, at 4°C overnight. Subsequent to being washed three times with PBS, the slides were incubated with biotinylated goat anti-rabbit secondary antibody (1:200 dilution; cat no. ZDR-5118; Histostain-Plus kit; ZSGB-Bio, Beijing, China) and horseradish peroxidase (Roche, Basel, Switzerland) for 30 min. The slides were then stained with 5% diaminobenzidine (Beijing Jorferin Biotechnology Co., Ltd.) for 1 min. The slides were counterstained with hematoxylin (Shanghai Bogoo Biotechnology Co., Ltd., Shanghai, China) for 2 min and washed with distilled water for 5 min. Subsequently, the slides were differentiated in 1% acid alcohol for ~10 sec, dehydrated, and cleared in graded alcohol and xylene (75% alcohol for 5 min, 95% alcohol for 5 min, 100% alcohol for 5 min and xylene for 20 min). Incubation of a tissue sample with PBS and without the primary antibody formed a negative control. The slides were routinely stained with hematoxylin and eosin (Shanghai Bogoo Biotechnology Co., Ltd.) to observe the nucleus and the cytoplasm of the cells.

Scoring of immunohistochemical staining

For the evaluation of the immunoreactivity of HMGB1 staining, the images of HMGB1 staining were independently analyzed by 2 pathologists (Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University), who were blinded to the clinicopathological data and prognosis of the patients. The fractions of HMGB1 staining reactivity were assessed by positive cell proportion analysis; ≥50 tumor cells were counted in 4 randomly selected regions of each section of the tissue and the mean percentage of stained cells was evaluated. The staining intensity of HMGB1 expression in tumor cells was estimated per section and classified as low reactivity (10–50% cells expressing HMGB1) and high reactivity (>50% cells expressing HMGB1).

Statistical analysis

Statistical data was analyzed using SPSS software, version 17.0 (SPSS, Inc., Chicago, IL, USA). The association between HMGB1 expression and clinicopathological parameters was explored with Pearson's χ2 test or Fisher's exact test. The Kaplan-Meier method was applied to calculate survival curves for the recurrence-free survival rate, and the log-rank test assessed the significance of the observed differences. The patients that succumbed during the follow-up period were censored. For multivariate analysis, Cox's proportional hazards regression model was performed to assess the risk factors for tumor recurrence. P<0.05 was regarded as statistically significant. Two-tailed tests were applied to all the analyses.

Results

Immunohistochemical expression and the association with clinicopathological features

Osteosarcoma cell membrane and cytoplasm staining is exhibited in Fig. 1A and B. In osteosarcoma tissue samples, HMGB1 expression was detected in 84 samples (48.3%) with low immunoreactivity and in 90 samples (51.7%) with high immunoreactivity. The association between clinicopathological characteristics and tumor cell HMGB1 expression (low vs. high) was summarized in Table I. The association between HMGB1 expression and tumor size, stage and nuclear grade was statistically significant (P=0.034, 0.008 and 0.019, respectively). There was no significant association between HMGB1 expression and the age of the patients (P=0.335; Table I). The current study concluded that patients with high HMGB1 expression (>50% cells expressing HMGB1) had poorer survival rates, and therefore a poorer prognosis, compared with patients with low HMGB1 immunostaining (10–50% cells expressing HMGB1).

Survival analysis

In total, 174 patients were analyzed in the current study. The follow-up time for the patients ranged between 5 and 80 months (median, 32.5 months). The Kaplan-Meier survival curves demonstrated that high HMGB1 expression was associated with a significantly lower recurrence-free survival rate compared with low HMGB1 expression (P=0.003; Fig. 2; Table II). In addition, the recurrence-free survival rate was significantly associated with tumor size (P<0.001), clinical stage (P<0.001) and tumor grade (P<0.001). Furthermore, Cox's proportional hazard model revealed that tumor stage, tumor grade and HMGB1 expression were independent predictors of a poorer prognosis in patients with osteosarcoma (P<0.001, 0.002 and 0.033, respectively; Table III), while the age of patients and tumor size were excluded as independent prognostic predictors (P=0.146 and 0.083, respectively; Table III).

Table II.

Univariate analyses of the recurrence-free survival in 174 osteosarcoma cases.

Table II.

Univariate analyses of the recurrence-free survival in 174 osteosarcoma cases.

Univariate analysisP-value
Age   0.750
  <30 vs. ≥30 years
TNM stage   <0.001*
  I vs. II vs. III vs. IV
Nuclear grade   <0.001*
  1 vs. 2 vs. 3–4
Tumor size <0.001*
HMGB1 reactivity   0.002*
  Low vs. high

{ label (or @symbol) needed for fn[@id='tfn2-ol-0-0-3907'] } Univariate analyses performed using Kaplan-Meier method and log-rank test.

* P<0.05. HMGB1, high mobility group box 1; TNM, tumor-node-metastasis.

Table III.

Multivariate analysis for predictors of survival.

Table III.

Multivariate analysis for predictors of survival.

Covariate analysisP-value
Age   0.246
TNM stage <0.001*
Nuclear grade    0.001*
Tumor size   0.083
HMGB1 reactivity    0.025*

{ label (or @symbol) needed for fn[@id='tfn4-ol-0-0-3907'] } Mulivariate analyses performed using Cox's proportional hazards regression model.

* P<0.05. HMGB1, high mobility group box 1; TNM, tumor-node-metastasis.

Discussion

Osteosarcoma accounts for ~20% of pediatric, solid and malignant tumors, and is the most common malignant bone tumor in adolescents and young adults (26). In total, ~40% of osteosarcomas metastasize and patients possess a poor overall prognosis (27,28). The 5-year survival rate is reported to be 37% for osteosarcoma patients with tumor metastasis and 19% for patients with >5 metastatic lung lesions (5). Uncontrolled cell proliferation and robust tumor angiogenesis are prominent characteristics that contribute to the poor prognosis of osteosarcoma patients (6). Although there has been considerable progress in the diagnosis, therapeutic strategies and understanding of the biological behaviors of osteosarcoma, certain aspects of prognosis remain unclear, particularly concerning the prognostic markers for tumor recurrence and the patient survival rate. The most concerning problems in osteosarcoma are the inability to identify patients that will or will not respond to standard therapy, and the lack of a marker that may predict the prognosis of an osteosarcoma patient (29). Consequently, significant attention has been directed to the elucidation of novel prognostic markers that may be associated with tumor recurrence and the progression of osteosarcoma, in order to predict the prognosis of patients with osteosarcoma (3032).

Evidence supporting the role of HMGB1 in cancer progression, angiogenesis, invasion and metastasis development has been steadily accumulating (33). Existing studies suggest that HMGB1 may demonstrate an important role in tumor progression beyond cancer development, including the association between HMGB1 overexpression and the presence of lymph node metastasis and advanced-stage hepatocellular, head and neck, esophageal squamous cell, cervical and ovarian carcinoma (3438). The present study demonstrates that HMGB1 overexpression, as determined by immunohistochemistry, is an important prognostic factor in osteosarcoma. To the best of our knowledge, the present study is the first to demonstrate that HMGB1 expression is associated with clinical prognosis in osteosarcoma. This observation provides the opportunity to consider potential clinical applications of HMGB1 as a prognostic marker. IHC staining for HMGB1 expression revealed that the immunoreactivity was localized to the tubular epithelium and osteosarcoma cytoplasm, and there was a significantly higher staining intensity associated with a more advanced tumor differentiation grade.

HMGB1 was initially defined as a chromatin-associated protein with high acidic and basic amino acid content (14). HMGB1 is a nuclear protein that acts as a chromatin-binding factor and exists in the nuclei of cancerous and normal cells (17). HMGB1 modifies the interaction of DNA with transcription factors, including p53 steroid hormone receptors, by non-specifically binding to the minor groove of DNA, thereby playing a role in DNA repair, transcription, differentiation, extracellular signalization, and somatic recombination (39).

HMGB1 demonstrates affinity for various DNA structures, including supercoiled and single-stranded DNA, B- and Z-DNA, DNA mini-circles, 4-way junctions, looped structures, hemicatenated DNA, and triplex DNA (39). Native HMGB1 released from tumor cells inhibits DNA replication. However, this effect decreases following acetylation, and in addition, recombinant HMGB1 is phosphorylated by the in vitro protein kinase C (40); therefore, HMGB1 cannot inhibit the replication of DNA (23).

Rapid tumor growth causes a decrease in the intensity of chronic hypoxia, and the formation of microvessels and necrotic foci (41). Antigenic factors are released from hypoxic and necrotic regions and inflammatory cells, such as macrophages, which are stimulated to release angiogenic cytokines and growth factors, migrate to necrotic foci (41). HMGB1 activation results in NF-κB activation, and this stimulates the release of leucocyte adhesion molecules and pro-inflammatory cytokines, leading to the enhancement of inflammation and angiogenesis (41). HMGB1 also stimulates angiogenesis by activating factors, including vascular endothelial growth factor (VEGF) (42). Wang et al investigated the association between HMGB1 expression and angiogenesis in bladder cancer samples. The authors reported that HMGB1 is associated with CD34 and VEGF, which are angiogenesis indicators (43). HMGB1 is associated with the pathological stage of tumors, as quantitative polymerase chain reaction has revealed an increase in HMGB1 mRNA expression as the tumor stage progresses (4345).

However, there is little data concerning the prognostic role of HMGB1 in osteosarcoma. The results of the current study demonstrated that HMGB1 expression is negatively associated with the clinical prognosis of patients with osteosarcoma. The survival curves demonstrated that, in patients with osteosarcoma, increased HMGB1 expression is associated with a poorer recurrence-free survival rate. The present study confirmed that the TNM stage and tumor size were predictive prognostic markers for the recurrence-free survival rate in patients with osteosarcoma, which was consistent with the findings of previous studies (6,46,47). Cox's multivariate regression revealed that independent prognostic factors for osteosarcoma consisted of tumor stage, tumor grade and HMGB1 reactivity, while the age and tumor size were not prognostic factors.

In summary, the results of the present study revealed that increased expression levels of HMGB1 were significantly associated with a poorer clinical prognosis, and therefore may act as a marker for prognosis, particularly in osteosarcoma recurrence. Additional studies concerning the biological features of HMGB1 are required to confirm the potential role of HMGB1 as a novel target for anticancer therapy in osteosarcoma.

Acknowledgements

The present study was supported by grants from the National Natural Science Foundation of China (grant no. 81300964), China Postdoctoral Science Foundation (grant nos. 2013M531611 and 2014T70648) and Natural Science Foundation of Shandong Province (grant no. ZR2011HM054).

References

1 

Broadhead ML, Dass CR and Choong PF: Systemically administered PEDF against primary and secondary tumours in a clinically relevant osteosarcoma model. Br J Cancer. 105:1503–1511. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Mirabello L, Troisi RJ and Savage SA: International osteosarcoma incidence patterns in children and adolescents, middle ages and elderly persons. Int J Cancer. 125:229–234. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Cong Y, Li CJ, Zhao JN, Liu XZ and Shi X: Associations of polymorphisms in the bone morphogenetic protein-2 gene with risk and prognosis of osteosarcoma in a Chinese population. Tumour Biol. 36:2059–2064. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Zhang F, Huang W, Sheng M and Liu T: MiR-451 inhibits cell growth and invasion by targeting CXCL16 and is associated with prognosis of osteosarcoma patients. Tumour Biol. 36:2041–2048. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Briccoli A, Rocca M, Salone M, Guzzardella GA, Balladelli A and Bacci G: High grade osteosarcoma of the extremities metastatic to the lung: Long-term results in 323 patients treated combining surgery and chemotherapy, 1985–2005. Surg Oncol. 19:193–199. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Bielack SS, Kempf-Bielack B, Delling G, Exner GU, Flege S, Helmke K, Kotz R, Salzer-Kuntschik M, Werner M, Winkelmann W, et al: Prognostic factors in high-grade osteosarcoma of the extremities or trunk: An analysis of 1,702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols. J Clin Oncol. 20:776–790. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Ren K, Yao N, Wang G, Tian L, Ma J, Shi X, Zhang L, Zhang J, Zhou X, Zhou G, et al: Vasculogenic mimicry: A new prognostic sign of human osteosarcoma. Hum Pathol. 45:2120–2129. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Seftor RE, Hess AR, Seftor EA, Kirschmann DA, Hardy KM, Margaryan NV and Hendrix MJ: Tumor cell vasculogenic mimicry: From controversy to therapeutic promise. Am J Pathol. 181:1115–1125. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Schmidt T and Carmeliet P: Angiogenesis: A target in solid tumors, also in leukemia? Hematology Am Soc Hematol Educ Program. 2011:1–8. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Fukumura D, Duda DG, Munn LL and Jain RK: Tumor microvasculature and microenvironment: Novel insights through intravital imaging in pre-clinical models. Microcirculation. 17:206–225. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Cao Z, Bao M, Miele L, Sarkar FH, Wang Z and Zhou Q: Tumour vasculogenic mimicry is associated with poor prognosis of human cancer patients: A systemic review and meta-analysis. Eur J Cancer. 49:3914–3923. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Cao Z, Shang B, Zhang G, Miele L, Sarkar FH, Wang Z and Zhou Q: Tumor cell-mediated neovascularization and lymphangiogenesis contrive tumor progression and cancer metastasis. Biochim Biophys Acta. 1836:273–286. 2013.PubMed/NCBI

13 

Liu YR, Sun B, Zhao XL, Gu Q, Liu ZY, Dong XY, Che N and Mo J: Basal caspase-3 activity promotes migration, invasion, and vasculogenic mimicry formation of melanoma cells. Melanoma Res. 23:243–253. 2013.PubMed/NCBI

14 

Alexandrova EA and Beltchev BG: Acetylated HMG1 protein interacts specifically with homologous DNA polymerase alpha in vitro. Biochem Biophys Res Commun. 154:918–927. 1988. View Article : Google Scholar : PubMed/NCBI

15 

Bonaldi T, Längst G, Strohner R, Becker PB and Bianchi ME: The DNA chaperone HMGB1 facilitates ACF/CHRAC-dependent nucleosome sliding. EMBO J. 21:6865–6873. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Scaffidi P, Misteli T and Bianchi ME: Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature. 418:191–195. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Ellerman JE, Brown CK, de Vera M, Zeh HJ, Billiar T, Rubartelli A and Lotze MT: Masquerader: High mobility group box-1 and cancer. Clin Cancer Res. 13:2836–2848. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Wang H, Vishnubhakat JM, Bloom O, Zhang M, Ombrellino M, Sama A and Tracey KJ: Proinflammatory cytokines (tumor necrosis factor and interleukin 1) stimulate release of high mobility group protein-1 by pituicytes. Surgery. 126:389–392. 1999. View Article : Google Scholar : PubMed/NCBI

19 

Tang D, Kang R, Van Houten B, Zeh HJ, Billiar TR and Lotze MT: High mobility group box 1 (HMGB1) phenotypic role revealed with stress. Mol Med. 20:359–362. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Luo Y, Chihara Y, Fujimoto K, Sasahira T, Kuwada M, Fujiwara R, Fujii K, Ohmori H and Kuniyasu H: High mobility group box 1 released from necrotic cells enhances regrowth and metastasis of cancer cells that have survived chemotherapy. Eur J Cancer. 49:741–751. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Tian J, Avalos AM, Mao SY, Chen B, Senthil K, Wu H, Parroche P, Drabic S, Golenbock D, Sirois C, et al: Toll-like receptor 9-dependent activation by DNA-containing immune complexes is mediated by HMGB1 and RAGE. Nat Immunol. 8:487–496. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Lin T, Sammy F, Yang H, Thundivalappil S, Hellman J, Tracey KJ and Warren HS: Identification of hemopexin as an anti-inflammatory factor that inhibits synergy of hemoglobin with HMGB1 in sterile and infectious inflammation. J Immunol. 189:2017–2022. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Zhang G, Chen F, Cao Y, Amos JV, Shah G and See WA: HMGB1 release by urothelial carcinoma cells in response to Bacillus Calmette-Guérin functions as a paracrine factor to potentiate the direct cellular effects of Bacillus Calmette-Guérin. J Urol. 190:1076–1082. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Balch CM, Gershenwald JE, Soong SJ, Thompson JF, Atkins MB, Byrd DR, Buzaid AC, Cochran AJ, Coit DG, Ding S, et al: Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol. 27:61992009. View Article : Google Scholar : PubMed/NCBI

25 

Fuhrman SA, Lasky LC and Limas C: Prognostic significance of morphologic parameters in renal cell carcinoma. Am J Surg Pathol. 6:655–663. 1982. View Article : Google Scholar : PubMed/NCBI

26 

Broadhead ML, Clark JC, Myers DE, Dass CR and Choong PF: The molecular pathogenesis of osteosarcoma: A review. Sarcoma. 2011:9592482011. View Article : Google Scholar : PubMed/NCBI

27 

Aragon-Ching JB and Maki RG: Treatment of adult soft tissue sarcoma: Old concepts, new insights, and potential for drug discovery. Cancer Invest. 30:300–308. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Mirabello L, Troisi RJ and Savage SA: Osteosarcoma incidence and survival rates from 1973 to 2004: Data from the Surveillance, Epidemiology, and End Results Program. Cancer. 115:1531–1543. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Kong C and Hansen MF: Biomarkers in osteosarcoma. Expert Opin Med Diagn. 3:13–23. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Laverdiere C, Hoang BH, Yang R, Sowers R, Qin J, Meyers PA, Huvos AG, Healey JH and Gorlick R: Messenger RNA expression levels of CXCR4 correlate with metastatic behavior and outcome in patients with osteosarcoma. Clin Cancer Res. 11:2561–2567. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Somers GR, Ho M, Zielenska M, Squire JA and Thorner PS: HER2 amplification and overexpression is not present in pediatric osteosarcoma: A tissue microarray study. Pediatr Dev Pathol. 8:525–532. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Osaka E, Suzuki T, Osaka S, Yoshida Y, Sugita H, Asami S, Tabata K, Hemmi A, Sugitani M, Nemoto N and Ryu J: Survivin as a prognostic factor for osteosarcoma patients. Acta Histochem Cytochem. 39:95–100. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Shen X, Hong L, Sun H, Shi M and Song Y: The expression of high-mobility group protein box 1 correlates with the progression of non-small cell lung cancer. Oncol Rep. 22:535–539. 2009.PubMed/NCBI

34 

Liu F, Zhang Y, Peng Z, Gao H, Xu L and Chen M: High expression of high mobility group box 1 (hmgb1) predicts poor prognosis for hepatocellular carcinoma after curative hepatectomy. J Transl Med. 10:1352012. View Article : Google Scholar : PubMed/NCBI

35 

Chuangui C, Peng T and Zhentao Y: The expression of high mobility group box 1 is associated with lymph node metastasis and poor prognosis in esophageal squamous cell carcinoma. Pathol Oncol Res. 18:1021–1027. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Chen J, Xi B, Zhao Y, Yu Y, Zhang J and Wang C: High-mobility group protein B1 (HMGB1) is a novel biomarker for human ovarian cancer. Gynecol Oncol. 126:109–117. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Liu Y, Xie C, Zhang X, Huang D, Zhou X, Tan P, Qi L, Hu G, Tian Y and Qiu Y: Elevated expression of HMGB1 in squamous-cell carcinoma of the head and neck and its clinical significance. Eur J Cancer. 46:3007–3015. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Hao Q, Du XQ, Fu X and Tian J: Expression and clinical significance of HMGB1 and RAGE in cervical squamous cell carcinoma. Zhonghua Zhong Liu Za Zhi. 30:292–295. 2008.(In Chinese). PubMed/NCBI

39 

Kang HJ, Lee H, Choi HJ, Youn JH, Shin JS, Ahn YH, Yoo JS, Paik YK and Kim H: Non-histone nuclear factor HMGB1 is phosphorylated and secreted in colon cancers. Lab Invest. 89:948–959. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Topalova D, Ugrinova I, Pashev IG and Pasheva EA: HMGB1 protein inhibits DNA replication in vitro: A role of the acetylation and the acidic tail. Int J Biochem Cell Biol. 40:1536–1542. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Schlueter C, Weber H, Meyer B, Rogalla P, Röser K, Hauke S and Bullerdiek J: Angiogenetic signaling through hypoxia: HMGB1: an angiogenetic switch molecule. Am J Pathol. 166:1259–1263. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Sitohy B, Nagy JA and Dvorak HF: Anti-VEGF/VEGFR therapy for cancer: Reassessing the target. Cancer Res. 72:1909–1914. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Wang W, Jiang H, Zhu H, Zhang H, Gong J, Zhang L and Ding Q: Overexpression of high mobility group box 1 and 2 is associated with the progression and angiogenesis of human bladder carcinoma. Oncol Lett. 5:884–888. 2013.PubMed/NCBI

44 

Tang D, Kang R, Zeh HJ III and Lotze MT: High-mobility group box 1 and cancer. Biochim Biophys Acta. 1799:131–140. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Xiao J, Ding Y, Huang J, Li Q, Liu Y, Ni W, Zhang Y, Zhu Y, Chen L and Chen B: The association of HMGB1 gene with the prognosis of HCC. PLoS One. 9:e890972014. View Article : Google Scholar : PubMed/NCBI

46 

Yonemoto T, Tatezaki S, Ishii T, Satoh T, Kimura H and Iwai N: Prognosis of osteosarcoma with pulmonary metastases at initial presentation is not dismal. Clin Orthop Relat Res. 349:194–199. 1998. View Article : Google Scholar : PubMed/NCBI

47 

Daw NC, Billups CA, Rodriguez-Galindo C, McCarville MB, Rao BN, Cain AM, Jenkins JJ, Neel MD and Meyer WH: Metastatic osteosarcoma. Cancer. 106:403–412. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2016
Volume 11 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
He J, Zhang P, Li Q, Zhou D and Liu P: Expression of high mobility group box 1 protein predicts a poorer prognosis for patients with osteosarcoma. Oncol Lett 11: 293-298, 2016
APA
He, J., Zhang, P., Li, Q., Zhou, D., & Liu, P. (2016). Expression of high mobility group box 1 protein predicts a poorer prognosis for patients with osteosarcoma. Oncology Letters, 11, 293-298. https://doi.org/10.3892/ol.2015.3907
MLA
He, J., Zhang, P., Li, Q., Zhou, D., Liu, P."Expression of high mobility group box 1 protein predicts a poorer prognosis for patients with osteosarcoma". Oncology Letters 11.1 (2016): 293-298.
Chicago
He, J., Zhang, P., Li, Q., Zhou, D., Liu, P."Expression of high mobility group box 1 protein predicts a poorer prognosis for patients with osteosarcoma". Oncology Letters 11, no. 1 (2016): 293-298. https://doi.org/10.3892/ol.2015.3907