Molecular pathogenesis and therapeutic strategies of human osteosarcoma

2016-02-11 17:30SahityaDenduluriZhongliangWangZhengjianYanJingWangQiangWeiMaryamMohammedRexHaydonHueLuuTongChuanHe
THE JOURNAL OF BIOMEDICAL RESEARCH 2016年1期

Sahitya K Denduluri,Zhongliang Wang,2,Zhengjian Yan,2,Jing Wang,2,Qiang Wei,2, Maryam K Mohammed,Rex C Haydon,Hue H Luu,Tong-Chuan He,✉

1The University of Chicago Pritzker School of Medicine,and Molecular Oncology Laboratory,Department of Orthopaedic Surgery and Rehabilitation Medicine,The University of Chicago Medical Center,Chicago,IL 60637,USA;

2Ministry of Education Key Laboratory of Diagnostic Medicine,and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016,China.

Molecular pathogenesis and therapeutic strategies of human osteosarcoma

Sahitya K Denduluri1,Zhongliang Wang1,2,Zhengjian Yan1,2,Jing Wang1,2,Qiang Wei1,2, Maryam K Mohammed1,Rex C Haydon1,Hue H Luu1,Tong-Chuan He1,✉

1The University of Chicago Pritzker School of Medicine,and Molecular Oncology Laboratory,Department of Orthopaedic Surgery and Rehabilitation Medicine,The University of Chicago Medical Center,Chicago,IL 60637,USA;

2Ministry of Education Key Laboratory of Diagnostic Medicine,and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016,China.

Osteosarcoma(OS)is a devastating illness with rapid rates of dissemination and a poor overall prognosis,despite aggressive standard-of-care surgical techniques and combination chemotherapy regimens.Identifying the molecular mechanisms involved in disease pathogenesis and progression may offer insight into new therapeutic targets. Defects in mesenchymal stem cell differentiation,abnormal expression of oncogenes and tumor suppressors,and dysregulation within various important signaling pathways have all been implicated in development of various disease phenotypes.As such,a variety of basic science and translational studies have shown promise in identifying novel markers and modulators of these disease-specific aberrancies.Born out of these and similar investigations, a variety of emerging therapies are now undergoing various phases of OS clinical testing.They broadly include angiogenesis inhibitors,drugs that act on the bone microenvironment,receptor tyrosine kinase inhibitors,immune system modulators,and other radio-or chemo-sensitizing agents.As new forms of drug delivery are being developed simultaneously,the possibility of targeting tumors locally while minimizing systemic toxicityis is seemingly more achievable now than ever.In this review,we not only summarize our current understanding of OS disease processes,but also shed light on the multitude of potential therapeutic strategies the scientific community can use to make long-term improvements in patient prognosis.

osteosarcoma,soft tissue tumors,bone tumors,cancer therapy,osteogenic tumors,osteogenic differentiation

Introduction

Osteogenic sarcoma(osteosarcoma,OS)is the most common primary malignancy of bone in children,with a distinct correlation between periods of rapid bone growth and development of disease[1-2].More commonly affecting males,primary tumors often arise in the metaphyses of long bones such as the femur or tibia[1,3-4].OS disseminates rapidly throughout the body, with 20%of patients noted to have secondary involvement at the time of diagnosis;90%of such metastases are found in the lungs[5-7].Though treatment approaches can vary considerably,the standard of care generally involves wide surgical resection with either neoadjuvantor adjuvant chemotherapy regimen[8].With one of the lowest survival rates among pediatric cancers,OS imparts a 5-year survivorship of 70%in patients treated for localized disease,but only 30%when metastases are present[6,9].Therefore,there is a critical need to better understand the underlying mechanisms of disease development and progression.This review seeks to provide a concise and useful synthesis of findings from recent, promising research efforts into these topics so as to encourage future investigations aimed at improving OS therapy and patient outcomes.

Molecular basis of OS and potential targets for therapy

Defects in osteogenic differentiation leading to OS development

OS commonly develops during the pubertal growth spurt,when bone turnover is high and defects in differentiation and signaling have the potential to be amplified[10-11].Osteoblasts arise from mesenchymal stem cells(MSCs),undifferentiated bone marrow stromal cells with the potential to self-renew and proliferate into bone,muscle,tendon,and fat[12-13].Milieus of endogenous and exogenous factors are involved in driving the osteogenic pathway from MSC to osteoblast.Dysregulation of these markers,or new exposure to non-native stimuli(e.g.pro-tumor inflammatory cytokines),causes an imbalance between cellular differentiation and proliferation,ultimately contributing to a malignant phenotype[14-16].

There are thought to be various similarities between early osteoprogenitors and OS cells,including a highly proliferative nature,resistance to apoptosis,and similar expression profiles of genes such as alkaline phosphatase (ALP)and connective tissue growth factor(CTGF)[17-18]. Furthermore,it is widely held that the earlier these defects occur in the osteoblastic lineage,the more undifferentiated or aggressive the cancer cells[15,19-20]. Accordingly,more invasive OS cells are noted to have minimal expression of osteocalcin(OCN)and osteopontin(OPN),both of which are observed at higher levels in mature osteoblasts[21-23].Another notable difference between late osteoprogenitors and OS tumor cells is the ability of the latter to evade senescence through an alternative lengthening of telomere(ALT)pathway[24]. Unlike more terminally differentiated osteoblasts with shortened telomeres resulting from many replication cycles,ALT allows OS cells to remain in a stem cell-like state and responsive to exogenous stimuli[25].

Bone morphogenetic proteins(BMPs)represent one such group of factors involved in OS stimulation[15]. Normally involved in carrying MSCs along an osteogenic lineage,BMPs are not only unable to induce differentiation of OS cells but may actually promote a more aggressive phenotype[26-27].This is due to an intrinsic underexpression of Runx2,a transcription factor which usually serves as a master regulator of BMP activity by causing exit from the cell cycle and promoting terminal differentiation[15,28].However, RUNX2 overexpression is also correlated with poor prognosis of OS tumors,indicating that its expression is likely tightly controlled in normal osteogenesis[29].

Select BMPs additionally exert effects through the Wnt glycoprotein pathway,a signaling network that has been extensively implicated in suppressing osteoblastic differentiation[30-33].Aberrant signaling by Wnt can also result in increased cell proliferation and migration through both the canonical β-catenin and noncanonical pathways[34-35].Accordingly,many research efforts have shown promise for OS therapy through inhibition of Wnt and downstream proto-oncogenes[36-38].However,similar to Runx2,it appears that normal Wnt signaling is also finely tuned,with other studies demonstrating a correlation between decreased pathway activity and hypoxic chemoresistance in OS[39].

Beyond understanding the roles of BMPs,Runx2, and Wnt,researchers have identified additional proteins of interest that may promote differentiation of OS cells,thereby inhibiting proliferation and increasing susceptibility to apoptosis.These include super proteins of the nuclear receptor family,such as PPARγ,retinoids,and estrogens[20,40-44].1,25-dihydroxyvitamin D3[1,25(OH)2D3],another nuclear receptor agonist,has shown promise by increasing expression levels of p21,a pro-apoptotic cell cycle regulator which drives osteogenic differentiation and senescence[45-48].Finally,parathyroid hormone-related peptide appears to promote differentiation of OS cells, observed through upregulation of osteoprogenitor markers ALP and collagen type I[49].Ultimately,it appears that loss of differentiation plays a critical role in osteo sarcoma genesis,but numerous molecular targets involved in the osteoblastic lineage may offer significant promise in developing new treatments.

Abnormal expression of oncogenes and tumor suppressors

As observed in most cancers,abnormal activity of oncogenes and tumor suppressors is a key molecular underpinning of osteosarcoma[50].c-Myc,perhaps one of the most researched and well-understood oncogenes in OS pathogenesis,is overexpressed in over 10% of tumors and is correlated with increased tumorrecurrence.Specifically,it increases invasiveness of cells through activation of the MEK-ERK pathway and decreases apoptotic potential[51-52].Recent studies have shown that inhibition of c-Myc activity results in decreased proliferation,invasion,and viability of OS cells,demonstrating its considerable value as a therapeutic target[53-56].Similar to c-Myc,c-Fos is another oncogene that correlates with a higher rate of metastasis when upregulated in a primary tumor[57]. Sorafenib,a kinase inhibitor commonly used in treatment of hepatocellular and renal cell carcinomas, causes a favorable response in OS cells by downregulating c-Fos and S100A4,another oncogene implicated in regulating the cell cycle,decreasing apoptosis,and inhibiting osteogenic differentiation of OS cells[58-59].

MDM2,a protein that marks the tumor suppressor p53(see below)for degradation,is amplified in at least 1 out of 10 patients[60,61].Furthermore,higher co-expression levels of MDM2 and CDK4,which promotes cell cycle progression,can be used reliably to distinguish low-grade OS from benign masses and correlates with further dedifferentiation into high-grade lesions[62-64]. The transcription factor MEF2D is overexpressed in clinical specimens from OS patients with poor prognoses,and silencing the protein using a miRNA suppresses cell proliferation by triggering G2-M cell cycle arrest[65].AURKA(coding for Aurora-A kinase) is an oncogene and an important regulator of mitosis that has undergone much recent investigation[66]. AURKA silencers and inhibitors of Aurora-A kinase have shown promising results in OS by not only causing hyperploidy and apoptosis,but also by working synergistically with traditional chemotherapeutics in cell lines that have become resistant to single-drug treatment[67-68].Indicating the true complexity of OS molecular biology,additional oncogenes have also been recently attributed to disease progression,including those that code for p21-activated kinase 7(PAK7), E2F transcription factor 2(E2F2),special AT-rich sequence-binding protein-1(SATB1),and several microRNAs such as 301a[69-72].These proteins are of high interest to researchers as potential targets for therapy in the future.

Deficient tumor suppressor activity appears to play an equally important role as dysregulation of oncogenes in OS pathogenesis.Rb,a regulator of the G1/ S cell cycle transition,is found to be insufficient in about 70%of all sporadic cases of OS,not to mention the nearly 1000-fold increased risk for developing OS in individuals who inherit an inactivated copy of the gene[73-75].Similarly,mutations in tumor suppressor p53 are commonly found in OS cells and contribute to disease progression by permitting cells with damaged DNA repair mechanisms to evade checkpoints and apoptosis[74,76-77].In fact,patients with type 2 neurofibromatosis actually have a higher incidence of OS due to increased activity of MDM2 and destabilization of p53[78].Finally,p16INK4Ais another tumor suppressor that normally inactivates CDK4 and has undergone much recent investigation as a biomarker that is positively correlated with patient survival in OS[79].

Signaling pathway dysregulation

Aberrant cell signaling is an equally important piece in the molecular biology puzzle underlying osteosar coma development and progression.Several ubiquitous pathways have been implicated in the disease,providing numerous potential therapeutic targets for researchers moving forward.The insulin-like growth factor (IGF)signaling axis is one that ties in closely with the development of disease during periods of significant bone growth,such as in adolescence.The IGF-1 receptor(IGF-1R)is a member of the tyrosine kinase family and is most commonly activated by the IGF-1 ligand,ultimately stimulating proliferation,protein synthesis,and glucose metabolism while inhibiting apoptosis[80].Normal functioning of this pathway is integral to both tissue homeostasis and growth,but loss of regulation has been extensively implicated in tumorigenesis and spread of disease[81-82].Specifically, increased expression levels of IGF-1and IGF-1R are associated with worse prognosis in patients with OS[83-84].Furthermore,IGF-2 mediates chemoresistance through a state of autophagic dormancy that preserves cell survival[85].A downstream mediator involved in both IGF and insulin signaling,insulin receptor substrate 1(IRS-1)is critically important for MSC differentiation.Its deregulation appears to be involved in malignant transformation of OS cells[86].The IGF binding proteins(IGFBPs),which modulate signaling through both IGF-dependent and IGF-independent mechanisms,have recently been implicated in OS[87]. Notably,IGFBP-5 expression is significantly downregulated in various cell lines,and exogenous administration of the protein has been shown to suppress tumor growth and metastasis by multiple mechanisms[88-89].

Downstream of IGF-1R,signaling is propagated through the PI3K/AKT and Ras/MAPK/ERK pathways[80].Upregulation of the former has been significantly implicated in OS pathogenesis,resulting in increased proliferation,increased invasion,and decreased apoptosis of tumor cells[90].Researchers have shown that various molecules cause this activation, including the long noncoding RNA metastasis-associatedlung adenocarcinoma transcript 1(MALAT1),tumor necrosis factor receptor-associated factor 4(TRAF 4), and autophagy related protein 6(Beclin-1)[91-93]. Furthermore,suppressing such activation may not only be involved in decreasing the aggressiveness of tumors, but may also be involved in overcoming chemoresistance,further demonstrating the importance of PI3K/ AKT signaling as a therapeutic target[91,94-97].Discussed earlier,Aurora-A(and-B)kinase inhibitors appear to also suppress this pathway as a means of halting tumor progression,representing a class of potential drugs seeking to match the complexity of the underlying pathophysiology[98-99].

Finally,inflammation and cytokine signaling have been heavily implicated in the tumorigenesis of OS[100].For example,transforming growth factor β (TGF-β)is linked to the dedifferentiation of osteosarcoma cells into cancer stem cells,a dynamic population associated with tumor invasion,radio-and chemoresistance,and poor prognosis[101].Often found to be involved in autocrine signaling by cancer cells, TGF-β increases the migration potential of OS cells through MAPK activation[102].Similarly,tumor necrosis factor α(TNF-α)is strongly correlated with disease spread,though researchers have recently shown that infliximab,a monoclonal antibody(mAb)to TNF-α, can decrease OS cell motility and pulmonary metastases in a mouse model[103].

Interleukins represent another important class of cytokines with similar roles in disease progression. A pro-inflammatory cytokine,interleukin 32 has a dose dependent effect on promoting invasion and migration of OS cells via activation of the AKT pathway and upregulation of matrix metalloproteinase 13[104]. Regulated by TNF-α and IL-1β,interleukin 34 is expressed by OS cells and similarly promotes tumor spread through neo-angiogenesis and recruitment of tumor-associated M2 macrophages,which further produce TGF-β and promote tumor growth[105-106]. Finally,interleukin 11 receptor α,a marker of poor long-term prognosis in various cancers,has been found to be overexpressed in OS and can actually serve in the development of improved noninvasive imaging and targeted therapy[107].

Inflammation maintains tumors in an aggressive state due to the milieu of molecules released by macrophages, many of which further recruit other inflammatory cells. Monocyte chemoattractant protein 1(MCP-1,or CCL2) is an example of this type of chemokine,involved in the critical migration of monocytes across the vascular endothelium and into tissues[108].Additionally in OS, MCP-1 expression is significantly upregulated and activates AKT signaling,with knockdown inhibiting both the proliferation and invasion of tumor cells[109]. Downstream of cytokines such as interleukins,the JAK2/STAT3 pathway also represents a notable target for potential therapeutics[110].Recent studies involving the use JAK2/STAT3 inhibitors delayed OS growth in vitro and in vivo,with similar results seen through short hairpin RNA knockdown of STAT3[111-112].Overall,it has become increasingly clear that modulating several notable signaling pathways and quelling the inflammatory response to tumors may lead to profound therapeutic response in OS.

Emerging therapies and clinical trials

Angiogenesis inhibitors

As in most types of cancer,the ability of an OS tumor to acquire a robust blood supply has significant implications for growth,metastasis,and ultimately prognosis[113-115].Therefore,drugs aimed at limiting angiogenesis have become increasingly studied in the treatment of various malignancies,including OS[116-117]. Vascular endothelial growth factor(VEGF)is one of the key regulators in angiogenesis and a well-studied marker associated with decreased disease-free survival in OS[118-121].VEGF inhibitors have demonstrated considerable success in basic science and translational studies by reducing growth and metastatic potential of OS tumors,with the potential to sensitize cells to chemotherapy[122-126].There are also several early clinical trials that are investigating bevacizumab,a mAb to VEGF,in patients with OS.However,there is concern that this drug may cause adverse events,particularlyin the pediatric population,including lymphopenia,pneumothorax,and increased wound dehiscence[127-131]. Finally,sorafenib is a tyrosine kinase inhibitor of the VEGF receptor family that has also undergone clinical investigation[132].In a phase II trial that studied 35 patients with unresectable OS previously unresponsive to standard therapy,sorafenib resulted in a 46% progression-free survival at 4 months and a reduction in tumor density in those with stable disease(34%of all patients)[133].Therefore,such targeted therapy warrants further investigation.Results from other welldesigned clinical trials are needed to shed light on outcomes,feasibility of combination therapy,and appropriate dosing of VEGF inhibitors in the treatment of OS.

Part of the PI3K/AKT pathway,mammalian target of rapamycin(mTOR)is another signaling molecule involved in angiogenesis,and mTOR inhibitors are under clinical investigation for use in mesenchymal tumors,including OS[134-136].A phase II clinical study published in 2015 reported that combination treatmentwith sorafenib and everolimus,an mTOR mAb, resulted in progression-free survival of unresectable OS at 6 months in 17 of 38 patients who had previously failed standard therapy[134].However,another phase II trial involving an IGF-1R inhibitor,cituxumumab,and mTOR mAb temsirolimus found no objective effect in 11 pediatric patients with OS[137].Since both of these studies involved the use of combination therapy, the role of mTOR inhibition alone in OS still remains unclear in the clinical setting,warranting the need for future studies addressing this issue.

Bisphosphonates

Bisphosphonates are used in the treatment of osteoporosis and can reduce skeletal-related events in adult cancers,but have also shown promise in specifically treating OS[138-139].Pre-clinical studies demonstrated that these agents inhibit the proliferation and metastasis of OS through activating apoptosis,suppressing tumor induced angiogenesis,augmenting T-cell-mediated cytotoxicity,and sensitizing to chemotherapy[140-147]. Due to the potential for adverse craniofacial effectssuch as osteonecrosis of the jaw,there have been clinical trials seeking to understand the feasibility and dosing of bisphosphonates in OS treatment[139,148].In 2011,Meyers et al.demonstratedthat pamidronate could be added to chemotherapy regimens without any increase in toxicity,resulting in 5-year event-free survival(EFS)rates of 72%and 45%for patients with localized and metastatic disease,respectively.Though the authors did not incorporate a no-treatment arm with regard to bisphosphonate,they commented that the agent might improve durability of limb reconstruction[149].Now that bisphosphonates have been deemed potentially safe,there are ongoing phase II/III clinical trials that may shed light on the role of the more-potent zoledronic acid in treating high-grade osteosarcoma, both alone and in conjunction with combination chemotherapy(NCT00691236,NCT00470223).

Receptor tyrosine kinase inhibitors

As previously mentioned,signaling pathway aberrancies are heavily involved in the aggressiveness of OS. Receptor tyrosine kinases(RTK)represent a class of molecules involved in propagating extracellular signals from a variety of sources,often growth factors,resulting in increased gene transcription,protein synthesis, and cell proliferation[150].VEGF receptors,blocked by sorafenib as discussed above,are members of the RTK family[151].Another important RTK is the human epidermal growth factor receptor 2(HER2/neu),extensively studied in the pathogenesis and treatment of breast cancer,but also of considerable interest in OS research[152-153].Pre-clinical trials have shown that both direct and indirect HER2 inhibition can have significant effects on decreasing OS proliferation,inhibiting migration,and promoting apoptosis[154-155].A recent phase II clinical trial found that trastuzumab,a mAb to HER2,can be safely dosed in conjunction with a chemotherapy regimen,but doesnot offer any improvement in outcome.However,the study did not randomize patients into treatment groups,so the trastu-zumab-specific effects still remain to be identified.Of note,two recently completed clinical trials may offer some insight into the use of trastuzumab,but are yet to be published.A group from Memorial Sloan Kettering is studying the drug as a single neoadjuvant agent before surgery in recurrent OS,whereas a study from the National Cancer Institute is comparing patients receiving standard-of-care chemotherapy with and without trastuzumab(NCT00005033 and NCT00023998).

Discussed earlier,the IGF-1 receptor is a tyrosine kinase and an important therapeutic target in OS,especially for adolescent patients with increased serum levels of growth factors seen during the pubertal growth spurt[156].IGF-1R inhibitors,including antibodies,have shown promising results in vitro and in vivo using animal models[157-159].Some clinical studies have addressed the use of an IGF-1R mAb in patients with various soft tissue and bone tumors,reporting that it is well tolerated but may have limited or no response in terms of outcomes[137,160-163].Another large,multi-center trial is evaluating the combination of cixutumumab, an IGF-1R mAb,and a VEGF-R inhibitor in patients with bone and soft tissue sarcoma.Of 54 patients with IGF-1R-positive bone sarcoma(18 with OS),19 were progression-free at 12 weeks.Of the 54 patients with IGF-1R-negative soft tissue and bone tumors(6 with OS)who were also followed,12 were progression-free at 12 weeks.Furthermore,based on histology,13%of the patients with OS showed a partial response to therapy.As this trial is ongoing,the median overall survival for OS has not yet been reached[164].Though promising,this study is evaluating two different drugs on a variety of tumors as classified by IGF-1R expression,only a small subset of which is OS.Therefore, there is a need to design clinical trials intended to understand the effects of IGF-1R inhibition on osteosarcoma specifically.

Immunotherapy

The immune system can be a valuable tool for targeting and destroying tumor cells,a topic which hasreceived considerable attention in the popular press[165-166]. Recent in vitro and in vivo work has shown the ability of a variety of immune cells,including natural killer cells,genetically modified T-cells,and viruses,to effectively kill OS tumor cells,making immunotherapy an intriguing prospect[167-170].Mifamurtide,or liposomal muramyl tripeptide phosphatidylethanolamine(LMTP-PE,Mepact),is an agent that causes recruitment of inflammatory macrophages and has been under clinical investigation with promising results[171,172].The drug is of particular interest to researchers because of its favorable safety profile and ability to target metastases,such as in the lung[173-174].

In Europe,mifamurtide has already been approved for use in children,adolescents,and young adults for non-metastatic osteosarcoma after tumor excision[175]. This was in response to the results of a large multi-center,randomized phase III trial,known as the Intergroup Study 0133,demonstrating the drug’s ability to improve overall survival in conjunction with 3-or 4-agent combination chemotherapy in newly diagnosed,high-grade,non-metastatic,resectable osteosarcoma[9].Though a similar trend was seen in a smaller cohort of patients presenting with metastatic disease, the result was not of statistical significance[176]. A recent Markov process model using data from the Intergroup 0133 trial found that mifamurtide improved the lifetime effectiveness of chemotherapy in both metastatic and nonmetastatic disease[177].Currently,in the United States,mifamurtide remains an orphaned drug,but additional well-designed,prospective,randomized controlled trials may offer a path for this promising drug to be re-introduced into the market as adjuvant therapy.

Granulocyte macrophage-colony stimulating factor (GM-CSF)exhibits similar macrophage-stimulating properties,but may also extend its reach to CD4 T-cells,natural killer cells,and dendritic cells[178].In laboratory studies,GM-CSF has shown an ability to induce osteoblastic differentiation and apoptosis in OS cells[179-180].An inhaled form of this factor was studied in a phase I trial evaluating patients with first isolated pulmonary recurrence of OS,with results showing that it had low toxicity but no discernible effects on immunostimulation or outcomes[181].The same group recently completed a phase II study looking at inhaled recombinant GM-CSF,sargramostim, on a similar group of patients with first pulmonary recurrence of the disease,but the results are yet to be published(NCT00066365).Overall,immune system modulators represent an important class of anti-neoplastic agents that may undergo considerable growth and development in the near future.

Chemo-and radio-sensitizing agents

As tumor resistance remains one of the most significant barriers to improving patient prognosis,numerous research efforts have recently been directed towards increasing OS response to existing chemotherapy and radiotherapy regimens[80,182-188].In addition to studies looking at various anti-neoplastic agents discussed above in combination with chemotherapy,a few researchers have also aimed to repurpose existing non-cancer drugs for use in OS[189].The most notable example is metformin,an insulin-sensitizer considered to be the first-line treatment for type II diabetes mellitus[190].An in vitro study found that metformin not only inhibits tumor cell growth but also sensitizes three different cisplatin-resistant cell lines to the drug,demonstrating a synergistic effect[191].This effect might be mediated through crosstalk that exists between insulinand IGF-signaling[192].Similarly,proton pump inhibitors(PPIs)normally used for dyspepsia have shown promise in OS.A recent translational study studied two cell lines in culture as well as a murine xenograft model,finding that in both settings pre-treatment with esomeprazole sensitized tumor cells to cisplatin.The same study then evaluated 98 patients aged 40 years or younger with resectable nonmetastatic OS of the extremities who received esomeprazole in the two days before each round of neoadjuvant chemotherapy. When compared to a historical study that used the same chemotherapy regimen,the authors found that PPI pre-treatment increased local cytotoxicity of the drugs as evidenced by histologic tumor necrosis[193]. Though the study did not randomize patients or look at survival rates as an outcome,this represents an exciting starting point for researchers to design future clinical studies involving the potential chemosensitizing effects of PPIs.

Radiotherapy plays a considerable role in treating incompletely-or un-resectable primary tumors and chemoresisant metastases to the lung and axial skeleton, though not a standard OS treatment modality due to resistance[8,194-195].Therefore,finding agents capable of sensitizing tumors to radiation may improve outcomes in patients who may need it as an end-of-the-line treatment option.Though most of the work to date has taken place in a laboratory setting,the findings do appear promising.A small molecule inhibitor of WEE1 kinase,found in many OS tissue samples,has been shown to bypass the G2 cell cycle checkpoint following radiation exposure,resulting in mitotic catastrophe[196].Similarly,radiation combined with parthenolide,a naturally occurring molecule that interferes with NF-κB cell survival signaling,has asynergistic effect on previously radio-resistant OS tumor cells,including cancer stem cells[197]. Previously discussed,the ALT pathway allows cancer cells to evade senescence and remain in a stem cell-like state,but also appears to play a role in resistance to irradiation.Suppression of Ku80,a protein involved in DNA repair via nonhomologous end joining,radio sensitizes an ALT OS line in vitro,with affected cells demonstrating shortened telomeres[198].Results such as this should spur more investigations to overcome resistance to radiation,a treatment modality that has been underutilized in OS.

Novel drug delivery mechanisms

Though the most effective treatment modality to date in OS,chemotherapy also carries with it toxic effects,several of which can promote the development of a secondary malignancy or cause significant morbidity[199-201].In response,as has been the approach in other cancers,researchers have studied the use of vectors for delivering OS therapy to a localized area,hoping to minimize unintended consequences.Stem cells represent such a promising option moving forward.Using bone-marrow-derived MSCs expressing the cytosine deaminase/5-fluorocytosine prodrug,a recent study found that the MSCs were able to migrate toward OS cells in vitro,resulting in cytotoxicity,and also inhibit subcutaneous tumor growth when injected locally into mice[202].Furthermore,another study showed that RFP-labeled human MSCs could be injected into the tail vein of athymic nude mice and effectively localize to OS tumors.The MSCs,which were carrying the osteo protegerin(OPG)gene,caused expression of the protein at the tumor site and resulted in decreased tumor growth and bone destruction[203].

Using nanocarriers may also be effective for delivering drugs to specific locations with sustained release[204-205].A recent study demonstrated that nanoparticles loaded with paclitaxel and etoposide demonstrated increased cytotoxic effects on OS cell lines when compared to a combination of the drugs in native form[206].Similarly,nanoparticles carrying the antibiotic salinomycin were designed to target CD133+osteosarcoma cancer stem cells and caused pronounced in vitro and in vivo cell death.A new technology involves nanotubes made from halloysite, which can be mined from natural deposits,and has been shown to inhibit OS cell proliferation when delivering methotrexate[207].Finally,organic molecules, such as liposomes and micelles,loaded with existing and experimental anti-neoplastic agents have been used with promising results in suppressing OS tumor cells in culture and in a xenograft model[208-210].This approach may also be useful for potential drugs,such as curcumin,that previously could not be properly formulated due to their chemical properties(e.g.water insoluble)[211,212].

Conclusions and future directions

In conclusion,a considerable body of recent,cutting edge research provides an optimistic view of osteosar coma treatment and patient prognosis in the future. With continued investigation into the molecular underpinnings of this aggressive disease,we can hope to better understand the interplay between various signaling and differentiation pathways,identifying the most critical molecular targets for therapy.An effort to design meaningful translational studies can then allow for a bench-to-bedside approach involving potential therapeutics.Finally,there is a pivotal need to implement more high-quality randomized clinical trials,focusing on just patients with osteosarcoma,as it has become increasingly clear that the unique properties of this malignancy make it difficult to predict drug response in comparison to other bone or soft tissue tumors. Ultimately,there may exist in some combination of surgery,chemotherapy,and localized molecular therapy that can significantly improve outcomes and quality of life for those suffering from osteosarcoma.

Acknowledgments

The authors’laboratories were supported in part by research grants from the National Institutes of Health (AT004418,AR50142,AR054381 to TCH,RCH and HHL),and the 973 Program of Ministry of Science and Technology(MOST)of China(#2011CB707900 to TCH).SKD was a recipient of the Pritzker Fellowship and AOA Carolyn L.Kuckein Fellowship.MKM was a recipient of Howard Hughes Medical Institute Medical Research Fellowship.

References

[1]Cotterill SJ,Wright CM,Pearce MS,et al.Stature of young people with malignant bone tumors[J].Pediatr Blood Cancer,2004,42(1):59-63.

[2]Longhi A,Errani C,De Paolis M,et al.Primary bone osteosarcoma in the pediatric age:state of the art[J]. Cancer Treat Rev,2006,32(6):423-436.

[3]Cho WH,Lee SY,Song WS,et al.Osteosarcoma in preadolescent patients[J].J Int Med Res,2006,34(6):676-681.

[4]Wittig JC,Bickels J,Priebat D,et al.Osteosarcoma:a multidisciplinary approach to diagnosis and treatment[J]. Am Fam Physician,2002,65(6):1123-1132.

[5]Bielack SS,Kempf-Bielack B,Delling G,et al.Prognostic factors in high-grade osteosarcoma of the extremities ortrunk:an analysis of 1,702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols[J].J Clin Oncol,2002,20(3):776-790.

[6]Chou AJ,Kleinerman ES,Krailo MD,et al.Addition of muramyl tripeptide to chemotherapy for patients with newly diagnosed metastatic osteosarcoma:a report from the Children’s Oncology Group[J].Cancer,2009,115(22):5339-5348.

[7]Wesolowski R,Budd GT.Use of chemotherapy for patients with bone and soft-tissue sarcomas[J].Cleve Clin J Med,2010,77 Suppl 1S23-26.

[8]Lamplot Joseph D SDJQ,Ruidong Li,Xing Liu,Hongyu Zhang,Xiang Chen,Ning Wang,Abdullah Pratt,Wei Shui,Xiaoji Luo,Guoxin Nan,Zhong-Liang Deng, Jinyong Luo,Rex C Haydon,Tong-Chuan He,and Hue H. Luu.The Current and Future Therapies for Human Osteosarcoma[J].Current Cancer Therapy Reviews,2013(9):55-77.

[9]Meyers PA,Schwartz CL,Krailo MD,et al.Osteosarcoma:the addition of muramyl tripeptide to chemotherapy improves overall survival-a report from the Children’s Oncology Group[J].J Clin Oncol,2008,26(4):633-638. [10]Marina N,Gebhardt M,Teot L,et al.Biology and therapeutic advances for pediatric osteosarcoma[J].Oncologist, 2004,9(4):422-441.

[11]Ottaviani G,Jaffe N.The epidemiology of osteosarcoma[J]. Cancer Treat Res,2009,1523-13.

[12]Reya T,Morrison SJ,Clarke MF,et al.Stem cells,cancer, and cancer stem cells[J].Nature,2001,414(6859):105-111.

[13]Deng ZL,Sharff KA,Tang N,et al.Regulation of osteogenic differentiation during skeletal development[J]. Front Biosci,2008,132001-2021.

[14]Tu B,Peng ZX,Fan QM,et al.Osteosarcoma cells promote the production of pro-tumor cytokines in mesenchymal stem cells by inhibiting their osteogenic differentiation through the TGF-beta/Smad2/3 pathway[J].Exp Cell Res,2014, 320(1):164-173.

[15]Wagner ER,Luther G,Zhu G,et al.Defective Osteogenic Differentiation in the Development of Osteosarcoma[J]. Sarcoma,2011,201112.

[16]Mori T,Sato Y,Miyamoto K,et al.TNFalpha promotes osteosarcoma progression by maintaining tumor cells in an undifferentiated state[J].Oncogene,2014,33(33):4236-4241.

[17]Harris SA,Enger RJ,Riggs BL,et al.Development and characterization of a conditionally immortalized human fetal osteoblastic cell line[J].J Bone Miner Res,1995, 10(2):178-186.

[18]Luo Q,Kang Q,Si W,et al.Connective tissue growth factor(CTGF)is regulated by Wnt and bone morphogenetic proteins signaling in osteoblast differentiation of mesenchymal stem cells[J].J Biol Chem,2004,279(53):55958-55968.

[19]TangN,SongWX,LuoJ,etal.Osteosarcoma Development and Stem Cell Differentiation[J].Clin Orthop Relat Res, 2008,466(9):2114-2130.

[20]He BC,Chen L,Zuo GW,et al.Synergistic antitumor effect of the activated PPAR gamma and retinoid receptors on human osteosarcoma[J].Clin Cancer Res,2010,16(8):2235-2245.

[21]Luo J,Sun MH,Kang Q,et al.Gene therapy for bone regeneration[J].Curr Gene Ther,2005,5(2):167-179.

[22]Gamblin AL,Renaud A,Charrier C,et al.Osteoblastic and osteoclastic differentiation of human mesenchymal stem cells and monocytes in a miniaturized three-dimensional culture with mineral granules[J].Acta Biomater, 2014,10(12):5139-5147.

[23]Li YS,Deng ZH,Zeng C,et al.Role of osteopontin in osteosarcoma[J].Med Oncol,2015,32(1):449.

[24]Wang LL.Biology of osteogenic sarcoma[J].Cancer J, 2005,11(4):294-305.

[25]Martin JW,Squire JA,Zielenska M.The genetics of osteosarcoma[J].Sarcoma,2012,2012627254.

[26]Cheng H,Jiang W,Phillips FM,et al.Osteogenic activity of the fourteen types of human bone morphogenetic proteins(BMPs)[J].J Bone Joint Surg Am,2003,85-a(8):1544-1552.

[27]Luo X,Chen J,Song WX,et al.Osteogenic BMPs promote tumor growth of human osteosarcomas that harbor differentiation defects[J].Lab Invest,2008,88(12):1264-1277.

[28]Thomas DM,Johnson SA,Sims NA,et al.Terminal osteoblast differentiation,mediated by runx2 and p27KIP1,is disrupted in osteosarcoma[J].The Journal of Cell Biology,2004,167(5):925-934.

[29]Martin JW,Zielenska M,Stein GS,et al.The Role of RUNX2 in Osteosarcoma Oncogenesis[J].Sarcoma, 2011,2011282745.

[30]Lin CH,Ji T,Chen CF,et al.Wnt signaling in osteosarcoma[J].Adv Exp Med Biol,2014,80433-45.

[31]Lv Z,Wang C,Yuan T,et al.Bone morphogenetic protein 9 regulates tumor growth of osteosarcoma cells through the Wnt/beta-catenin pathway[J].Oncol Rep,2014,31(2):989-994.

[32]Cai Y,Cai T,Chen Y.Wnt pathway in osteosarcoma, from oncogenic to therapeutic[J].J Cell Biochem,2014, 115(4):625-631.

[33]de Boer J,Siddappa R,Gaspar C,et al.Wnt signaling inhibits osteogenic differentiation of human mesenchymal stem cells[J].Bone,2004,34(5):818-826.

[34]Zhang A,He S,Sun X,et al.Wnt5a promotes migration of human osteosarcoma cells by triggering a phosphatidylinositol-3 kinase/Akt signals[J].Cancer Cell Int,2014, 1415.

[35]Li C,Shi X,Zhou G,et al.The canonical Wnt-betacatenin pathway in development and chemotherapy of osteosarcoma[J].Front Biosci(Landmark Ed),2013, 181384-1391.

[36]McQueen P,Ghaffar S,Guo Y,et al.The Wnt signaling pathway:implications for therapy in osteosarcoma[J]. Expert Rev Anticancer Ther,2011,11(8):1223-1232.

[37]Zhao S,Kurenbekova L,Gao Y,et al.NKD2,a negative regulator of Wnt signaling,suppresses tumor growth and metastasis in osteosarcoma[J].Oncogene,2015.

[38]Wang DZ,Gao JF,Jing SF,et al.Antitumor Effect of Docetaxel in Osteosarcoma by the Inhibition of Wnt Signal Channel[J].Drug Res(Stuttg),2014.

[39]Scholten DJ,2nd,Timmer CM,Peacock JD,et al.Down regulation of Wnt signaling mitigates hypoxia-induced chemoresistance in human osteosarcoma cells[J].PLoS One,2014,9(10):e111431.

[40]Wagner ER,He BC,Chen L,et al.Therapeutic Implications of PPARgamma in Human Osteosarcoma[J].PPAR Res, 2010,2010956427.

[41]Fang D,Yang H,Lin J,et al.17beta-estradiol regulates cell proliferation,colony formation,migration,invasion and promotes apoptosis by upregulating miR-9 and thus degrades MALAT-1 in osteosarcoma cell MG-63 in an estrogen receptor-independent manner[J].Biochem Biophys Res Commun,2015,457(4):500-506.

[42]Kallio A,Guo T,Lamminen E,et al.Estrogen and the selective estrogen receptor modulator(SERM)protection against cell death in estrogen receptor alpha and beta expressing U2OS cells[J].Mol Cell Endocrinol,2008, 289(1-2):38-48.

[43]Haydon RC,Zhou L,Feng T,et al.Nuclear receptor agonists as potential differentiation therapy agents for human osteosarcoma[J].Clin Cancer Res,2002,8(5):1288-1294.

[44]Haydon RC,Luu HH,He TC.Osteosarcoma and osteoblastic differentiation:a new perspective on oncogenesis[J]. Clin Orthop Relat Res,2007,454237-246.

[45]Thompson L,Wang S,Tawfik O,et al.Effect of 25-hydroxyvitamin D3 and 1 alpha,25 dihydroxyvitamin D3 on differentiation and apoptosis of human osteosarcoma cell lines[J].J Orthop Res,2012,30(5):831-844.

[46]Kanemaru M,Maehara N,Chijiiwa K.Antiproliferative effect of 1alpha,25-dihydroxyvitamin D3 involves upregulation of cyclin-dependent kinase inhibitor p21 in human pancreatic cancer cells[J].Hepatogastroenterology, 2013,60(125):1199-1205.

[47]Fei C,Zhao Y,Guo J,et al.Senescence of bone marrow mesenchymal stromal cells is accompanied by activation of p53/p21 pathway in myelodysplastic syndromes[J]. Eur J Haematol,2014,93(6):476-486.

[48]Nozaki K,Kadosawa T,Nishimura R,et al.1,25-Dihydroxyvitamin D3,recombinant human transforming growth factor-beta 1,and recombinant human bone morphogenetic protein-2 induce in vitro differentiation of canine osteosarcoma cells[J].J Vet Med Sci,1999, 61(6):649-656.

[49]Carpio L,Gladu J,Goltzman D,et al.Induction of osteoblast differentiation indexes by PTHrP in MG-63 cells involves multiple signaling pathways[J].Am J Physiol Endocrinol Metab,2001,281(3):E489-499.

[50]Broadhead ML,Clark JCM,Myers DE,et al.The Molecular Pathogenesis of Osteosarcoma:A Review[J]. Sarcoma,2011,2011.

[51]Han G,Wang Y,Bi W.C-Myc overexpression promotes osteosarcoma cell invasion via activation of MEK-ERK pathway[J].Oncol Res,2012,20(4):149-156.

[52]Wu X,Cai Z-d,Lou L-m,et al.Expressions of p53,c-MYC,BCL-2 and apoptotic index in human osteosarcoma and their correlations with prognosis of patients[J]. Cancer Epidemiology,2012,36(2):212-216.

[53]He N,Zhang Z.Baicalein suppresses the viability of MG-63 osteosarcoma cells through inhibiting c-MYC expression via Wnt signaling pathway[J].Mol Cell Biochem, 2015.

[54]Li Z,Liu H,Li B,et al.Saurolactam Inhibits Proliferation, Migration,and Invasion of Human Osteosarcoma Cells[J].Cell Biochem Biophys,2015.

[55]Xu N,Li Z,Yu Z,et al.MicroRNA-33b suppresses migration and invasion by targeting c-Myc in osteosarcoma cells[J].PLoS One,2014,9(12):e115300.

[56]Liu Z,Zhang G,Li J,et al.The tumor-suppressive microRNA-135b targets c-myc in osteoscarcoma[J]. PLoS One,2014,9(7):e102621.

[57]Gamberi G,Benassi MS,Bohling T,et al.C-myc and cfos in human osteosarcoma:prognostic value of mRNA and protein expression[J].Oncology,1998,55(6):556-563.

[58]Chen X,Luther G,Zhang W,et al.The E-F hand calciumbinding protein S100A4 regulates the proliferation,survival and differentiation potential of human osteosarcoma cells[J].Cell Physiol Biochem,2013,32(4):1083-1096.

[59]Walter I,Wolfesberger B,Miller I,et al.Human osteosarcoma cells respond to sorafenib chemotherapy by downregulation of the tumor progression factors S100A4, CXCR4 and the oncogene FOS[J].Oncol Rep,2014, 31(3):1147-1156.

[60]Lonardo F,Ueda T,Huvos AG,et al.p53 and MDM2 alterations in osteosarcomas:correlation with clinico pathologic features and proliferative rate[J].Cancer, 1997,79(8):1541-1547.

[61]Miller CW,Aslo A,Won A,et al.Alterations of the p53, Rb and MDM2 genes in osteosarcoma[J].J Cancer Res Clin Oncol,1996,122(9):559-565.

[62]Yoshida A,Ushiku T,Motoi T,et al.Immunohisto -chemical analysis of MDM2 and CDK4 distinguishes low-grade osteosarcoma from benign mimics[J].Mod Pathol,2010,23(9):1279-1288.

[63]Yoshida A,Ushiku T,Motoi T,et al.MDM2 and CDK4 immunohistochemical coexpression in high-grade osteosarcoma:correlation with a dedifferentiated subtype[J]. Am J Surg Pathol,2012,36(3):423-431.

[64]Jeon DG,Koh JS,Cho WH,et al.Clinical outcome of low-grade central osteosarcoma and role of CDK4 and MDM2 immunohistochemistry as a diagnostic adjunct[J]. J Orthop Sci,2015.

[65]Yu H,Sun H,Bai Y,et al.MEF2D overexpression contributes to the progression of osteosarcoma[J].Gene,2015, 563(2):130-135.

[66]Zhu X,Mei J,Wang Z.Aurora-A kinase:potential tumor marker of osteosarcoma[J].J Cancer Res Ther,2014,10 SupplC102-107.

[67]Jiang Z,Jiang J,Yang H,et al.Silencing of Aurora kinase A by RNA interference inhibits tumor growth in human osteosarcoma cells by inducing apoptosis and G2/M cell cycle arrest[J].Oncol Rep,2014,31(3):1249-1254.

[68]Tavanti E,Sero V,Vella S,et al.Preclinical validation of Aurora kinases-targeting drugs in osteosarcoma[J].Br J Cancer,2013,109(10):2607-2618.

[69]Zhang Y,Duan G,Feng S.MicroRNA-301a modulates doxorubicin resistance in osteosarcoma cells by targeting AMP-activated protein kinase alpha 1[J].Biochem Biophys Res Commun,2015,459(3):367-373.

[70]Iwasaki T,Tanaka K,Kawano M,et al.Tumor-suppressive microRNA-let-7a inhibits cell proliferation via targeting of E2F2 in osteosarcoma cells[J].Int J Oncol,2015, 46(4):1543-1550.

[71]Han K,Zhou Y,Gan ZH,et al.p21-activated kinase 7 is an oncogene in human osteosarcoma[J].Cell Biol Int, 2014,38(12):1394-1402.

[72]Wang G,Li B,Fu Y,et al.miR-23a suppresses proliferation of osteosarcoma cells by targeting SATB1[J]. Tumour Biol,2015.

[73]Capodano A.Bone:Osteosarcoma[J].Atlas Genet Cytogenet Oncol Haematol,2003,7(1):44-47.

[74]Berman SD,Calo E,Landman AS,et al.Metastatic osteosarcoma induced by inactivation of Rb and p53 in the osteoblast lineage[J].Proc Natl Acad Sci U S A,2008, 105(33):11851-11856.

[75]Feugeas O,Guriec N,Babin-Boilletot A,et al.Loss of heterozygosity of the RB gene is a poor prognostic factor in patients with osteosarcoma[J].J Clin Oncol,1996, 14(2):467-472.

[76]Overholtzer M,Rao PH,Favis R,et al.The presence of p53 mutations in human osteosarcomas correlates with high levels of genomic instability[J].Proceedings of the National Academy of Sciences,2003,100(20):11547-11552.

[77]Guo W,Wang X,Feng C.P53 gene abnormalities in osteosarcoma[J].Chin Med J(Engl),1996,109(10):752-755.

[78]Stemmer-Rachamimov AO,Nielsen GP,Rosenberg AE, et al.The NF2 gene and merlin protein in human osteosarcomas[J].Neurogenetics,1998,2(1):73-74.

[79]Bu J,Li H,Liu LH,et al.P16(INK4a)overexpression and survival in osteosarcoma patients:a meta analysis[J].Int J Clin Exp Pathol,2014,7(9):6091-6096.

[80]Denduluri SK,Idowu O,Wang Z,et al.Insulin-like growth factor(IGF)signaling in tumorigenesis and the development of cancer drug resistance[J].Genes&Diseases, 2015,2(1):13-25.

[81]MacEwen EG,Pastor J,Kutzke J,et al.IGF-1 receptor contributes to the malignant phenotype in human and canine osteosarcoma[J].J Cell Biochem,2004,92(1):77-91.

[82]LeRoith D,Roberts Jr CT.The insulin-like growth factor system and cancer[J].Cancer Letters,2003,195(2):127-137.

[83]Jentzsch T,Robl B,Husmann M,et al.Worse prognosis of osteosarcoma patients expressing IGF-1 on a tissue microarray[J].Anticancer Res,2014,34(8):3881-3889.

[84]Wang YH,Han XD,Qiu Y,et al.Increased expression of insulin-like growth factor-1 receptor is correlated with tumor metastasis and prognosis in patients with osteosarcoma[J].J Surg Oncol,2012,105(3):235-243.

[85]Shimizu T,Sugihara E,Yamaguchi-Iwai S,et al.IGF2 preserves osteosarcoma cell survival by creating an autophagic state of dormancy that protects cells against chemotherapeutic stress[J].Cancer Res,2014,74(22):6531-6541.

[86]Contaldo C,Myers TJ,Zucchini C,et al.Expression levels of insulin receptor substrate-1 modulate the osteoblastic differentiation of mesenchymal stem cells and osteosarcoma cells[J].Growth Factors,2014,32(1):41-52.

[87]Vivian H,Youngman O,Ron GR.The Insulin-Like Growth Factor-Binding Protein(IGFBP)Superfamily[J]. Endocrine Reviews,1999,20(6):761-787.

[88]Luther GA,Lamplot J,Chen X,et al.IGFBP5 domains exert distinct inhibitory effects on the tumorigenicity and metastasis of human osteosarcoma[J].Cancer Lett, 2013,336(1):222-230.

[89]Su Y,Wagner ER,Luo Q,et al.Insulin-like growth factor binding protein 5 suppresses tumor growth and metastasis of human osteosarcoma[J].Oncogene,2011,30(37):3907-3917.

[90]Zhang J,Yu X-H,Yan Y-G,et al.PI3K/Akt signaling in osteosarcoma[J].Clinica Chimica Acta,2015,444(0):182-192.

[91]Zhang W,Li Q,Song C,et al.Knockdown of autophagyrelated protein 6,Beclin-1,decreases cell growth,invasion, and metastasis and has a positive effect on chemotherapyinduced cytotoxicity in osteosarcoma cells[J].Tumour Biol,2015,36(4):2531-2539.

[92]Dong Y,Liang G,Yuan B,et al.MALAT1 promotes the proliferation and metastasis of osteosarcoma cells by activating the PI3K/Akt pathway[J].Tumour Biol,2015, 36(3):1477-1486.

[93]Yao W,Wang X,Cai Q,et al.TRAF4 enhances osteosarcoma cell proliferation and invasion by Akt signaling pathway[J].Oncol Res,2014,22(1):21-28.

[94]Zhou Y,Zhu LB,Peng AF,et al.LY294002 inhibits the malignant phenotype of osteosarcoma cells by modulating the phosphatidylinositol 3kinase/Akt/fatty acid synthase signaling pathway in vitro[J].Mol Med Rep,2015, 11(2):1352-1357.

[95]Gupte A,Baker E,Wan SS,et al.Systematic screening identifies dual PI3K and mTOR inhibition as a conserved therapeutic vulnerability in osteosarcoma[J].Clin Cancer Res,2015.

[96]Yang L,Shu T,Liang Y,et al.GDC-0152 attenuates the malignant progression of osteosarcoma promoted by ANGPTL2 via PI3K/AKT but not p38MAPK signaling pathway[J].Int J Oncol,2015,46(4):1651-1658.

[97]Perry JA,Kiezun A,Tonzi P,et al.Complementary genomic approaches highlight the PI3K/mTOR pathway as a common vulnerability in osteosarcoma[J].Proc Natl Acad Sci U S A,2014,111(51):E5564-5573.

[98]Niu NK,Wang ZL,Pan ST,et al.Pro-apoptotic and proautophagic effects of the Aurora kinase A inhibitor alisertib(MLN8237)on human osteosarcoma U-2 OS and MG-63 cells through the activation of mitochondriamediated pathway and inhibition of p38 MAPK/PI3K/ Akt/mTOR signaling pathway[J].Drug Des Devel Ther, 2015,91555-1584.

[99]Zhu LB,Jiang J,Zhu XP,et al.Knockdown of Aurora-B inhibits osteosarcoma cell invasion and migration via modulating PI3K/Akt/NF-kappaB signaling pathway[J]. Int J Clin Exp Pathol,2014,7(7):3984-3991.

[100]Bianchi E,Artico M,Di Cristofano C,et al.Growth factors,their receptor expression and markers for proliferation of endothelial and neoplastic cells in human osteosarcoma[J].Int J Immunopathol Pharmacol,2013, 26(3):621-632.

[101]Zhang H,Wu H,Zheng J,et al.Transforming growth factor beta1 signal is crucial for dedifferentiation of cancer cells to cancer stem cells in osteosarcoma[J].Stem Cells,2013,31(3):433-446.

[102]Chen J,Song Y,Yang J,et al.The up-regulation of cysteine-rich protein 61 induced by transforming growth factor beta enhances osteosarcoma cell migration[J].Mol Cell Biochem,2013,384(1-2):269-277.

[103]Kato H,Wakabayashi H,Naito Y,et al.Anti-tumor necrosis factor therapy inhibits lung metastasis in an osteosarcoma cell line[J].Oncology,2015,88(3):139-146.

[104]Zhou Y,Hu Z,Li N,et al.Interleukin-32 stimulates osteosarcoma cell invasion and motility via AKT pathwaymediated MMP-13 expression[J].Int J Mol Med,2015.

[105]Segaliny AI,Mohamadi A,Dizier B,et al.Interleukin-34 promotes tumor progression and metastatic process in osteosarcoma through induction of angiogenesis and macrophage recruitment[J].Int J Cancer,2015,137(1):73-85.

[106]Martinez FO,Gordon S.The M1 and M2 paradigm of macrophage activation:time for reassessment[J]. F1000Prime Rep,2014,6.

[107]Liu T,Ma Q,Zhang Y,et al.Interleukin-11 receptor alpha is overexpressed in human osteosarcoma,and near-infrared-labeled IL-11Ralpha imaging agent could detect osteosarcoma in mouse tumor xenografts[J].Tumour Biol, 2015,36(4):2369-2375.

[108]Deshmane SL,Kremlev S,Amini S,et al.Monocyte chemoattractant protein-1(MCP-1):an overview[J]. J Interferon Cytokine Res,2009,29(6):313-326.

[109]Chen Q,Sun W,Liao Y,et al.Monocyte chemotactic protein-1 promotes the proliferation and invasion of osteosarcoma cells and upregulates the expression of AKT[J].Mol Med Rep,2015,12(1):219-225.

[110]Kamran MZ,Patil P,Gude RP.Role of STAT3 in Cancer Metastasis and Translational Advances[J].Biomed Res Int,2013,2013.

[111]Yan J,Wang Q,Zou K,et al.Inhibition of the JAK2/ STAT3 signaling pathway exerts a therapeutic effect on osteosarcoma[J].Mol Med Rep,2015,12(1):498-502.

[112]Liu Y,Wang L,Wu Y,et al.Pterostilbene exerts antitumor activity against human osteosarcoma cells by inhibiting the JAK2/STAT3 signaling pathway[J].Toxicology, 2013,304120-131.

[113]Huang Z,Bao SD.Roles of main pro-and anti-angiogenic factors in tumor angiogenesis[J].World J Gastroenterol, 2004,10(4):463-470.

[114]Kreuter M,Bieker R,Bielack SS,et al.Prognostic relevance of increased angiogenesis in osteosarcoma[J]. Clin Cancer Res,2004,10(24):8531-8537.

[115]Uehara F,Tome Y,Miwa S,et al.Osteosarcoma cells enhance angiogenesis visualized by color-coded imaging in the in vivo Gelfoam(R)assay[J].J Cell Biochem, 2014,115(9):1490-1494.

[116]Cook KM,Figg WD.Angiogenesis inhibitors:current strategies and future prospects[J].CA Cancer J Clin, 2010,60(4):222-243.

[117]El-Kenawi AE,El-Remessy AB.Angiogenesis inhibitors in cancer therapy:mechanistic perspective on classification and treatment rationales[J].Br J Pharmacol,2013, 170(4):712-729.

[118]Hoeben A,Landuyt B,Highley MS,et al.Vascular endothelial growth factor and angiogenesis[J].Pharmacol Rev,2004,56(4):549-580.

[119]Bajpai J,Sharma M,Sreenivas V,et al.VEGF expression as a prognostic marker in osteosarcoma[J].Pediatr Blood Cancer,2009,53(6):1035-1039.

[120]Charity RM,Foukas AF,Deshmukh NS,et al.Vascular endothelial growth factor expression in osteosarcoma[J]. Clin Orthop Relat Res,2006,448193-198.

[121]Dong-Ju Z,Ai-Ju X,Yun-Jiao T,et al.Polymorphisms of vascular endothelial growth factor on prognosis in osteosarcoma patients[J].Pak J Med Sci,2014,30(5):1072-1076.

[122]Scharf VF,Farese JP,Coomer AR,et al.Effect of bevacizumab on angiogenesis and growth of canine osteosarcoma cells xenografted in athymic mice[J].Am J Vet Res,2013,74(5):771-778.

[123]Zhao D,Jia P,Wang W,et al.VEGF-mediated suppression of cell proliferation and invasion by miR-410 in osteosarcoma[J].Mol Cell Biochem,2015,400(1-2):87-95.

[124]Zhang K,Zhang C,Liu L,et al.A key role of microRNA-29b in suppression of osteosarcoma cell proliferation and migration via modulation of VEGF[J].Int J Clin Exp Pathol,2014,7(9):5701-5708.

[125]Gao YS,Mei J,Tong TL,et al.Inhibitory effects of VEGF-siRNA mediated by adenovirus on osteosarcomabearing nude mice[J].Cancer Biother Radiopharm, 2009,24(2):243-247.

[126]Yang Q,Zhang S,Kang M,et al.Synergistic growth inhibition by sorafenib and cisplatin in human osteosarcoma cells[J].Oncol Rep,2015,33(5):2537-2544.

[127]Glade-Bender J,Kandel JJ,Yamashiro DJ.VEGF blocking therapy in the treatment of cancer[J].Expert Opin Biol Ther,2003,3(2):263-276.

[128]Turner DC,Navid F,Daw NC,et al.Population Pharmacokinetics of Bevacizumab in Children with Osteosarcoma:Implications for Dosing[J].Clinical Cancer Research,2014,20(10):2783-2792.

[129]Howdieshell TR,Callaway D,Webb WL,et al.Antibody Neutralization of Vascular Endothelial Growth Factor Inhibits Wound Granulation Tissue Formation[J]. Journal of Surgical Research,2001,96(2):173-182.

[130]Wagner L,Turpin B,Nagarajan R,et al.Pilot study of vincristine,oral irinotecan,and temozolomide(VOIT regimen)combined with bevacizumab in pediatric patients with recurrent solid tumors or brain tumors[J]. Pediatr Blood Cancer,2013,60(9):1447-1451.

[131]de Pasquale MD,Castellano A,de Sio L,et al.Bevacizumab in pediatric patients:how safe is it?[J]Anticancer Res,2011,31(11):3953-3957.

[132]Adnane L,Trail PA,Taylor I,et al.Sorafenib(BAY 43-9006,Nexavar),a dual-action inhibitor that targets RAF/ MEK/ERK pathway in tumor cells and tyrosine kinases VEGFR/PDGFR in tumor vasculature[J].Methods Enzymol, 2006,407597-612.

[133]Grignani G,Palmerini E,Dileo P,et al.A phase II trial of sorafenib in relapsed and unresectable high-grade osteo -sarcoma after failure of standard multi modal therapy:an Italian Sarcoma Group study[J].Ann Oncol,2012,23(2):508-516.

[134]Grignani G,Palmerini E,Ferraresi V,et al.Sorafenib and everolimus for patients with unresectable high-grade osteosarcoma progressing after standard treatment:a non-randomised phase 2 clinical trial[J].Lancet Oncol, 2015,16(1):98-107.

[135]Chawla SP,Staddon AP,Baker LH,et al.Phase II study of the mammalian target of rapamycin inhibitor ridaforolimus in patients with advanced bone and soft tissue sarcomas[J].J Clin Oncol,2012,30(1):78-84.

[136]Karar J,Maity A.PI3K/AKT/mTOR Pathway in Angiogenesis[J].Front Mol Neurosci,2011,451.

[137]Wagner LM,Fouladi M,Ahmed A,et al.Phase II study of cixutumumab in combination with temsirolimus in pediatric patients and young adults with recurrent or refractory sarcoma:a report from the Children’s Oncology Group[J]. Pediatr Blood Cancer,2015,62(3):440-444.

[138]Drake MT,Clarke BL,Khosla S.Bisphosphonates:Mechanism of Action and Role in Clinical Practice[J]. Mayo Clin Proc,2008,83(9):1032-1045.

[139]Goldsby RE,Fan TM,Villaluna D,et al.Feasibility and dose discovery analysis of zoledronic acid with concurrent chemotherapy in the treatment of newly diagnosed metastatic osteosarcoma:A report from the Children’s Oncology Group[J].European Journal of Cancer,2013, 49(10):2384-2391.

[140]Yang XN,Zeng JC,Song YC,et al.Targeted antiosteosarcoma methotrexate-bisphosphonate conjugate induces apoptosis of osteosarcoma cells in vitro[J].Eur Rev Med Pharmacol Sci,2014,18(15):2116-2123.

[141]Ohba T,Cates JM,Cole HA,et al.Pleiotropic effects of bisphosphonates on osteosarcoma[J].Bone,2014, 63110-120.

[142]Lamoureux F,Baud’huin M,Ory B,et al.Clusterin inhibition using OGX-011 synergistically enhances zoledronic acid activity in osteosarcoma[J].Oncotarget,2014, 5(17):7805-7819.

[143]Chang J,Wang W,Zhang H,et al.Bisphosphonates regulate cell proliferation,apoptosis and pro-osteoclastic expression in MG-63 human osteosarcoma cells[J]. Oncol Lett,2012,4(2):299-304.

[144]Li Z,Peng H,Xu Q,et al.Sensitization of human osteosarcoma cells to Vgamma9Vdelta2 T-cell-mediated cytotoxicity by zoledronate[J].J Orthop Res,2012,30(5):824-830. [145]Ohba T,Cole HA,Cates JM,et al.Bisphosphonates inhibit osteosarcoma-mediated osteolysis via attenuation of tumor expression of MCP-1 and RANKL[J].J Bone Miner Res,2014,29(6):1431-1445.

[146]Li Z,Tang J,Sun L,et al.[Effect of zoledronate on the cytotoxicity of gammadelta T cells from PBMCs of osteosarcoma patients against osteosarcoma][J].Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi,2013,29(1):6-9.

[147]Moriceau G,Ory B,Mitrofan L,et al.Zoledronic acid potentiates mTOR inhibition and abolishes the resistance of osteosarcoma cells to RAD001(Everolimus):pivotal role of the prenylation process[J].Cancer Res,2010, 70(24):10329-10339.

[148]Lezot F,Chesneau J,Battaglia S,et al.Preclinical evidence of potential craniofacial adverse effect of zoledronic acid in pediatric patients with bone malignancies[J]. Bone,2014,68146-152.

[149]Meyers PA,Healey JH,Chou AJ,et al.Addition of pamidronate to chemotherapy for the treatment of osteosarcoma[J]. Cancer,2011,117(8):1736-1744.

[150]Lemmon MA,Schlessinger J.Cell signaling by receptor tyrosine kinases[J].Cell,2010,141(7):1117-1134.

[151]Olsson A-K,Dimberg A,Kreuger J,et al.VEGF receptor signalling?in control of vascular function[J].Nat Rev Mol Cell Biol,2006,7(5):359-371.

[152]Gill J,Geller D,Gorlick R.HER-2 involvement in osteosarcoma[J].Adv Exp Med Biol,2014,804161-177. [153]Brown-Glaberman U,Dayao Z,Royce M.HER2-targeted therapy for early-stage breast cancer:a comprehensive review[J].Oncology(Williston Park),2014,28(4):281-289.

[154]Wang TF,Wang H,Peng AF,et al.Inhibition of fatty acid synthase suppresses U-2 OS cell invasion and migration via downregulating the activity of HER2/PI3K/AKT signaling pathway in vitro[J].Biochem Biophys Res Commun, 2013,440(2):229-234.

[155]Long XH,Zhang GM,Peng AF,et al.Lapatinib alters the malignant phenotype of osteosarcoma cells via downregulation of the activity of the HER2-PI3K/AKT-FASN axis in vitro[J].Oncol Rep,2014,31(1):328-334.

[156]Juul A,Dalgaard P,Blum WF,et al.Serum levels of insulin-like growth factor(IGF)-binding protein-3(IGFBP-3) in healthy infants,children,and adolescents:the relation to IGF-I,IGF-II,IGFBP-1,IGFBP-2,age,sex,body mass index,and pubertal maturation[J].J Clin Endocrinol Metab,1995,80(8):2534-2542.

[157]Beltran PJ,Chung YA,Moody G,et al.Efficacy of ganitumab(AMG 479),alone and in combination with rapamycin,in Ewing’s and osteogenic sarcoma models[J]. J Pharmacol Exp Ther,2011,337(3):644-654.

[158]Kuijjer ML,Peterse EF,van den Akker BE,et al.IR/ IGF1R signaling as potential target for treatment of high-grade osteosarcoma[J].BMC Cancer,2013,13245. [159]Duan Z,Choy E,Harmon D,et al.Insulin-like growth factor-I receptor tyrosine kinase inhibitor cyclolignan picropodophyllin inhibits proliferation and induces apoptosis in multidrug resistant osteosarcoma cell lines[J].Mol Cancer Ther,2009,8(8):2122-2130.

[160]Malempati S,Weigel B,Ingle AM,et al.Phase I/II trial and pharmacokinetic study of cixutumumab in pediatric patients with refractory solid tumors and Ewing sarcoma:a report from the Children’s Oncology Group[J].J Clin Oncol,2012,30(3):256-262.

[161]Weigel B,Malempati S,Reid JM,et al.Phase 2 trial of cixutumumab in children,adolescents,and young adults with refractory solid tumors:a report from the Children’s Oncology Group[J].Pediatr Blood Cancer,2014,61(3):452-456.

[162]Pappo AS,Vassal G,Crowley JJ,et al.A phase 2 trial of R1507,a monoclonal antibody to the insulin-like growth factor-1 receptor(IGF-1R),in patients with recurrent or refractory rhabdomyosarcoma,osteosarcoma,synovial sarcoma,and other soft tissue sarcomas:results of a Sarcoma Alliance for Research Through Collaboration study[J].Cancer,2014,120(16):2448-2456.

[163]Tap WD,Demetri G,Barnette P,et al.Phase II study of ganitumab,a fully human anti-type-1 insulin-like growth factor receptor antibody,in patients with metastatic Ewing family tumors or desmoplastic small round cell tumors[J].J Clin Oncol,2012,30(15):1849-1856.

[164]Schwartz GK,Tap WD,Qin LX,et al.Cixutumumab and temsirolimus for patients with bone and soft-tissue sarcoma:a multicentre,open-label,phase 2 trial[J].Lancet Oncol,2013,14(4):371-382.

[165]Alderton GK,Bordon Y.Tumour immunotherapy-leukocytes take up the fight[J].Nat Rev Immunol,2012, 12(4):237-237.

[166]Botter SM,Neri D,Fuchs B.Recent advances in osteosarcoma[J].Curr Opin Pharmacol,2014,1615-23. [167]Tarek N,Lee DA.Natural killer cells for osteosarcoma[J]. Adv Exp Med Biol,2014,804341-353.

[168]DeRenzo C,Gottschalk S.Genetically modified T-cell therapy for osteosarcoma[J].Adv Exp Med Biol,2014, 804323-340.

[169]Hingorani P,Sampson V,Lettieri C,et al.Oncolytic viruses for potential osteosarcoma therapy[J].Adv Exp Med Biol,2014,804259-283.

[170]Li B,Zhu X,Sun L,et al.Induction of a specific CD8+T-cell response to cancer/testis antigens by demethylating pre-treatment against osteosarcoma[J].Oncotarget,2014, 5(21):10791-10802.

[171]Frampton JE.Mifamurtide:a review of its use in the treatment of osteosarcoma[J].Paediatr Drugs,2010,12(3):141-153.

[172]Meyers PA,Chou AJ.Muramyl tripeptide-phosphatidyl ethanolamine encapsulated in liposomes(L-MTP-PE)in the treatment of osteosarcoma[J].Adv Exp Med Biol, 2014,804307-321.

[173]Asano T,Kleinerman ES.Liposome-encapsulated MTPPE:a novel biologic agent for cancer therapy[J].J Immunother Emphasis Tumor Immunol,1993,14(4):286-292.

[174]Anderson PM,Meyers P,Kleinerman E,et al.Mifamurtide in metastatic and recurrent osteosarcoma:a patient access study with pharmacokinetic,pharmacodynamic,and safety assessments[J].Pediatr Blood Cancer,2014,61(2):238-244.

[175]Bielack S,Carrle D,Casali PG.Osteosarcoma:ESMO clinical recommendations for diagnosis,treatment and follow-up[J].Ann Oncol,2009,20 Suppl 4137-139.

[176]Chou AJ,Kleinerman ES,Krailo MD,et al.Addition of muramyl tripeptide to chemotherapy for patients with newly diagnosed metastatic osteosarcoma:a report from the Children’s Oncology Group[J].Cancer,2009, 115(22):5339-5348.

[177]Song HJ,Lee JA,Han E,et al.Lifetime effectiveness of mifamurtide addition to chemotherapy in nonmetastatic and metastatic osteosarcoma:a Markov process model analysis[J].Tumour Biol,2015.

[178]Shi Y,Liu CH,Roberts AI,et al.Granulocyte -macrophage colony-stimulating factor(GM-CSF)and T-cell responses:what we do and don’t know[J].Cell Res, 2006,16(2):126-133.

[179]Postiglione L,Di Domenico G,Giordano -Lanza G,et al. Effect of human granulocyte macrophage-colony stimulating factor on differentiation and apoptosis of the human osteosarcoma cell line SaOS-2[J].Eur J Histochem, 2003,47(4):309-316.

[180]Postiglione L,Domenico GD,Montagnani S,et al. Granulocyte -macrophage colony-stimulating factor(GMCSF)induces the osteoblastic differentiation of the human osteosarcoma cell line SaOS-2[J].Calcif Tissue Int,2003, 72(1):85-97.

[181]Arndt CA,Koshkina NV,Inwards CY,et al.Inhaled granulocyte -macrophage colony stimulating factor for first pulmonary recurrence of osteosarcoma:effects on diseasefree survival and immuno modulation.a report from the Children’s Oncology Group[J].Clin Cancer Res,2010, 16(15):4024-4030.

[182]Chou AJ,Gorlick R.Chemotherapy resistance in osteosarcoma:current challenges and future directions[J].Expert Rev Anticancer Ther,2006,6(7):1075-1085.

[183]PosthumaDeBoer J,van Royen BJ,Helder MN. Mechanisms of therapy resistance in osteosarcoma:a review[J].Oncology Discovery,2013,1(1).

[184]Zhang C,Hong CS,Hu X,et al.Inhibition of protein phosphatase 2A with the small molecule LB100 overcomes cell cycle arrest in osteosarcoma after cisplatin treatment[J].Cell Cycle,20150.

[185]Ren XF,Mu LP,Jiang YS,et al.LY2109761 inhibits metastasis and enhances chemosensitivity in osteosarcoma MG-63 cells[J].Eur Rev Med Pharmacol Sci, 2015,19(7):1182-1190.

[186]Xia YZ,Ni K,Guo C,et al.Alopecurone B reverses doxorubicin -resistant human osteosarcoma cell line by inhibiting P-glycoprotein and NF-kappa B signaling[J]. Phytomedicine,2015,22(3):344-351.

[187]Wang Z,Yang L,Xia Y,et al.Icariin enhances cytotoxicity of doxorubicin in human multidrug-resistant osteosarcoma cells by inhibition of ABCB1 and down-regulation of the PI3K/Akt pathway[J].Biol Pharm Bull,2015, 38(2):277-284.

[188]Li S,Sun W,Wang H,et al.Research progress on the multidrug resistance mechanisms of osteosarcoma chemotherapy and reversal[J].Tumour Biol,2015,36(3):1329-1338.

[189]Kansara M,Teng MW,Smyth MJ,et al.Translational biology of osteosarcoma[J].Nat Rev Cancer,2014, 14(11):722-735.

[190]Holman R.Metformin as first choice in oral diabetes treatment:the UKPDS experience[J].Journ Annu Diabetol Hotel Dieu,200713-20.

[191]Quattrini I,Conti A,Pazzaglia L,et al.Metformin inhibits growth and sensitizes osteosarcoma cell lines to cisplatin through cell cycle modulation[J].Oncol Rep,2014, 31(1):370-375.

[192]Garofalo C,Capristo M,Manara MC,et al.Metformin as an adjuvant drug against pediatric sarcomas:hypoxia limits therapeutic effects of the drug[J].PLoS One,2013, 8(12):e83832.

[193]Ferrari S,Perut F,Fagioli F,et al.Proton pump inhibitor chemosensitization in human osteosarcoma:from the bench to the patients’bed[J].J Transl Med,2013,11268. [194]Schwarz R,Bruland O,Cassoni A,et al.The role of radiotherapy in oseosarcoma[J].Cancer Treat Res,2009, 152147-164.

[195]Stea B,Cavazzana A,Kinsella TJ.Small-cell osteosarcoma:correlation of in vitro and clinical radiation response[J].Int J Radiat Oncol Biol Phys,1988,15(5):1233-1238.

[196]PosthumaDeBoer J,Wurdinger T,Graat HC,et al.WEE1 inhibition sensitizes osteosarcoma to radiotherapy[J]. BMC Cancer,2011,11156.

[197]Zuch D,Giang AH,Shapovalov Y,et al.Targeting radioresistant osteosarcoma cells with parthenolide[J].J Cell Biochem,2012,113(4):1282-1291.

[198]Hu L,Wu QQ,Wang WB,et al.Suppression of Ku80 correlates with radiosensitivity and telomere shortening in the U2OS telomerase-negative osteosarcoma cell line[J]. Asian Pac J Cancer Prev,2013,14(2):795-799.

[199]Plenderleith IH.Treating the Treatment:Toxicity of Cancer Chemotherapy[J].Can Fam Physician,1990, 361827-1830.

[200]Khorana AA,Francis CW,Culakova E,et al.Thromboembolism is a leading cause of death in cancer patients receiving outpatient chemotherapy[J].Journal of Thrombosis and Haemostasis,2007,5(3):632-634.

[201]Ng AK,Travis LB.Subsequent malignant neoplasms in cancer survivors[J].Cancer J,2008,14(6):429-434.

[202]NguyenThai QA,Sharma N,Luong DH,et al.Targeted inhibition of osteosarcoma tumor growth by bone marrow-derived mesenchymal stem cells expressing cytosine deaminase/5-fluorocytosine in tumor-bearing mice[J]. J Gene Med,2015.

[203]Qiao B,Shui W,Cai L,et al.Human mesenchymal stem cells as delivery of osteoprotegerin gene:homing and therapeutic effect for osteosarcoma[J].Drug Des Devel Ther,2015,9969-976.

[204]Peer D,Karp JM,Hong S,etal.Nanocarriers as an emerging platform for cancer therapy[J].Nat Nano,2007,2(12):751-760.

[205]Alexis F,Pridgen EM,Langer R,et al.Nanoparticle technologies for cancer therapy[J].Handb Exp Pharmacol, 2010(197):55-86.

[206]Wang B,Yu XC,Xu SF,et al.Paclitaxel and etoposide coloaded polymeric nanoparticles for the effective combination therapy against human osteosarcoma[J].J Nanobiotechnology,2015,13(1):22.

[207]Sun L,Mills DK.Halloysite nanotube -based drug delivery system for treating osteosarcoma[J].Conf Proc IEEE Eng Med Biol Soc,2014,20142920-2923.

[208]Sun L,Zhou DS,Zhang P,et al.Gemcitabine and gamma cyclodextrin/docetaxel inclusion complex-loaded liposome for highly effective combinational therapy of osteosarcoma[J].Int J Pharm,2014,478(1):308-317.

[209]Low SA,Yang J,Kopecek J.Bone-targeted acid-sensitive doxorubicin conjugate micelles as potential osteosarcoma therapeutics[J].Bioconjug Chem,2014,25(11):2012-2020. [210]Federman N,Chan J,Nagy JO,et al.Enhanced Growth Inhibition of Osteosarcoma by Cytotoxic Polymerized Liposomal Nanoparticles Targeting the Alcam Cell Surface Receptor[J].Sarcoma,2012,201211.

[211]Dhule SS,Penfornis P,He J,et al.The combined effect of encapsulating curcumin and C6 ceramide in liposomal nanoparticles against osteosarcoma[J].Mol Pharm, 2014,11(2):417-427.

[212]Kimura R,Rokkaku T,Takeda S,et al.Cytotoxic Effects of Fucoidan Nanoparticles against Osteosarcoma[J].Mar Drugs,2013,11(11):4267-4278.

✉Tong-Chuan He,MD,PhD,Molecular Oncology Laboratory,The University of Chicago Medical Center, 5841 South Maryland Avenue,MC 3079 Chicago,IL 60637,USA,Tel/ Fax:(773)702-7169/(773)834-4598,E-mail:tche@uchicago.edu.

16 May 2015,Accepted 16 June 2015,Epub 30 August 2015

R738.1,Document code:A

The authors reported no conflict of interests.