Skip to main content

Radiotherapy in bone sarcoma: the quest for better treatment option

Abstract

Bone sarcomas are rare tumors representing 0.2% of all cancers. While osteosarcoma and Ewing sarcoma mainly affect children and young adults, chondrosarcoma and chordoma have a preferential incidence in people over the age of 40. Despite this range in populations affected, all bone sarcoma patients require complex transdisciplinary management and share some similarities. The cornerstone of all bone sarcoma treatment is monobloc resection of the tumor with adequate margins in healthy surrounding tissues. Adjuvant chemo- and/or radiotherapy are often included depending on the location of the tumor, quality of resection or presence of metastases. High dose radiotherapy is largely applied to allow better local control in case of incomplete primary tumor resection or for unresectable tumors. With the development of advanced techniques such as proton, carbon ion therapy, radiotherapy is gaining popularity for the treatment of bone sarcomas, enabling the delivery of higher doses of radiation, while sparing surrounding healthy tissues. Nevertheless, bone sarcomas are radioresistant tumors, and some mechanisms involved in this radioresistance have been reported. Hypoxia for instance, can potentially be targeted to improve tumor response to radiotherapy and decrease radiation-induced cellular toxicity. In this review, the benefits and drawbacks of radiotherapy in bone sarcoma will be addressed. Finally, new strategies combining a radiosensitizing agent and radiotherapy and their applicability in bone sarcoma will be presented.

Peer Review reports

Introduction

Bone sarcoma are rare tumors accounting for 0.2% of all tumors with an incidence in North America and Europe of 0.75 / 100 000 [1]. Bone sarcoma can be classified according to the age of tumor onset. On the one hand, osteosarcoma (OS) and bone Ewing sarcoma (EWS) mostly affect children and young adults, and on the other hand chondrosarcoma (CHS) and chordoma (CD) occur after the age of 40 [1]. The survival rate of adults with bone sarcoma is low, around 50–60% at 5 years and 30% at 10 years, principally because of the indolent nature of these tumors [2,3,4,5]. For localized pediatric bone sarcomas, the 5-year survival rate is around 70% [2,3,4,5] and drops to 30% for pediatric bone sarcoma presenting metastases at diagnosis, which occurs in 20–25% of pediatric bone sarcoma [1, 3].

Notwithstanding the age of tumor onset or histological type of sarcoma, the management of all bone sarcoma patients is based on a transdisciplinary approach where surgery, with complete resection of the primary tumor, remains the cornerstone. Indeed, the quality of resection is an essential prognostic factor for all bone sarcomas. Depending on the location of the tumor and the tumoral invasion of peripheral tissues, surgery can be challenging and is not feasible in all cases. Radiotherapy is frequently used to ensure better local control [5,6,7,8]. In the case of Ewing sarcoma surgical resection and radiotherapy are both standard options for local control. Conversely, radiations are not applied as first-line treatment for resectable osteosarcoma, chordoma and chondrosarcoma, albeit high doses of radiotherapy are used as adjuvant treatment in case of marginal or incomplete resection, and as definitive local treatment for unresectable tumors [9,10,11]. Treatment strategy must be adapted according to tumor location, ease of resection and treatment-associated morbidity, as unnecessary high doses of radiation can trigger serious side effects such as neuropathies and fractures [12,13,14]. Challenging bone sarcoma of the axial skeleton are frequently treated with Intensity Modulated photon Radiation Therapy (IMRT) because of the higher dose applied to the tumor and the sparing of healthy tissues [15]. The development of advanced radiotherapy techniques like carbon-ion, or proton therapy has drastically improved patient care by reducing the exposure of nearby critical organs to radiations and increasing the dose of radiations delivered specifically to the tumor [12,13,14]. Combined proton and photon radiotherapy is also increasingly used for the treatment of sarcoma of the spine and sacrum and seems to improve local control [12, 16]. Excellent clinical results have been observed for sarcomas of the skull and cervical spine treated with proton therapy [13, 17]. Interesting results have also been reported with heavier particles, such as carbon ion. Access to these advanced RT techniques is increasing in developed countries. Hence, radiotherapy is an important component in bone sarcoma management, and in this review, we will discuss the benefits of radiotherapy for bone sarcoma, the mechanisms involved in tumor radioresistance, and the innovative ways to improve radiotherapy efficacy in these tumors.

Radiotherapy for bone sarcomas

The place of radiotherapy in the treatment of bone sarcoma has evolved with the development of new types of radiations and new ways to deliver these radiations (Table 1). Nevertheless, this evolution raises the question of choosing the best radiotherapeutic approach for the right tumor depending on patient age, tumor locations, histological subtype, tumor grade, previous treatment. The following paragraphs include an overview of the efficacy of conventional radiotherapy and non-conventional radiotherapy in bone sarcoma.

Table 1 Bone sarcomas

Role of radiotherapy in CD and CHS treatment

Over the past few years, more information has become available on the effects of radiotherapy in bone sarcoma patients with unresectable or residual tumors. In this part, we summarize treatment guidelines and present the latest clinical studies evaluating the efficacy of radiotherapy in bone sarcoma (Table 2).

Table 2 Radiotherapy in chordoma and chondrosarcoma

In chondrosarcoma, radiotherapy can be considered for unresectable disease (primary or recurrent), after incomplete surgery and for symptom palliation. High-dose RT is currently recommended for patients with skull base chondrosarcoma and for inoperable, locally advanced, and metastatic high-grade chondrosarcomas with a poor prognosis. For chordoma, en bloc R0 resection is the recommended treatment for primary localized disease when feasible and sequelae are accepted by the patient. If these conditions are not met, RT alone without debulking is an alternative. For skull base and upper cervical tract chordoma, resection with negative margins can rarely be done, and microscopically-positive margins should be the goal of surgery. Adjuvant RT should be considered for skull base and cervical spine chordomas, and for sacral and mobile spine chordomas with R1 resection margins.

A few historical retrospective studies have been conducted to determine whether chordoma and chondrosarcoma patients could benefit from peri-operative radiotherapy. Two major retrospective studies have evaluated the role of radiotherapy in chordoma, comparing surgery alone vs surgery and conventional radiotherapy in 1478 and 5427 chordoma patients, respectively (level of evidence 2b) [9, 19]. Both studies concluded that radiotherapy peri-operatively improves patient local control when surgery with positive margins are performed. High-dose RT is also associated with better outcome [9, 19]. The same observation has be made in a retrospective study of 743 high-grade chondrosarcoma defining radiotherapy as an independent protective factor (level of evidence 2b) [21].

Different advanced radiotherapeutic techniques have been developed in the last few decades (Table 3, see Table 2 [9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35]). First, the use of proton therapy is associated with better outcome than conventional radiotherapy in both chordoma and chondrosarcoma [9, 19, 21, 23]. The administration of proton and photon therapy post-operatively tend to be more efficient with a 5-year local control rate of 85.4% in CD, while it does not exceed 74% when combining surgery and photon radiotherapy alone [22]. When radiotherapy is administered as a single treatment (e.g. in unresectable tumors), proton therapy is a better option than conventional radiotherapy for both CHS and CD, resulting in a 5-year overall survival of 75% for CHS and 100% for CD, whereas the 5-year overall survival is only 19.1% for CHS and 34.1% for CD for conventional radiotherapy [23]. In skull base chordoma and chondrosarcoma, which are particularly difficult to handle surgically due to their proximity to vital structures, carbon ion radiotherapy administered peri-operatively has shown promising results with a 5-year local control of 80% and 89% in CD and CHS, respectively [20]. Stereotactic Radiation Therapy (SRT) has also been used in both chordoma and chondrosarcoma, and retrospective studies reported different results, with local control rates varying between 57% at 10 months and 90% at 28 months [28, 29].

Table 3 Radiotherapy principles

Chondrosarcoma and chordoma have a very low incidence, thus international clinical trials uniting bone sarcoma centers worldwide are ongoing to determine the best therapeutic option depending on the type of the tumor, its localization (NCT05033288, NCT01182779) and its resectability (NCT02986516).

Role of radiotherapy in the treatment of Ewing sarcoma and osteosarcoma

Radiotherapy may be considered in osteosarcoma patients with unresectable tumors, primary tumors where surgery would be unacceptably morbid, or as adjuvant treatment of tumors at high risk of local recurrence and with limited option for further surgery. For patients with bone Ewing sarcoma, RT with definitive intent alone should be used instead of surgery if complete surgical excision is not possible and in cases with challenging local sites such as axial or spinal tumors, where surgery will be unacceptably morbid. Adjuvant RT (45–60 Gy) significantly reduces Local Recurrence in patients with large tumors (> 200 ml), poor histological response or inadequate surgical margins and should be recommended in these circumstances [IV, B].

In addition, adjuvant RT should be considered in patients with non-sacral pelvic Ewing Sarcoma regardless of surgical margins, tumor volume or histological response, as this was shown to provide superior local control and survival outcome compared with surgery alone.

Several studies aimed at determining the best use of radiotherapy for EWS patients comparing radiotherapy alone with i) surgery alone, ii) post-operative RT, or iii) polychemotherapy (see Table 4). In a retrospective study (INT0091, INT0154, AEWS0031), radiotherapy alone increased the rate of local relapse compared to surgery alone in EWS patients with localized tumors [31].However, no difference was observed in the overall survival and overall disease control between those two treatments [30]. For patients with extremity and pelvic tumors, surgery clearly improved local control compared to definitive radiotherapy (local relapse rates 3.7% and 3.9% vs 14.8 and 22.4%, respectively) [30]. For other tumor locations, no difference was detected between the different treatment groups. Of note, in this study, patients treated with surgery had favorable prognostic factors such as a younger age or tumors of the extremities, and most of the patients were treated with older techniques of radiotherapy. Another study compared the same treatment options (surgery vs radiotherapy vs combined treatment) in metastatic EWS. The combination of surgery and radiotherapy improved the local control of metastatic tumors compared to surgery or radiotherapy alone (EFS at 3 years: RT: 0.35, surgery: 0.35, combination: 0.56) [31].

Table 4 Radiotherapy in Ewing sarcoma and Osteosarcoma

Stereotactic Body Radiation Therapy (SBRT) (Table 1) uses several radiation beams of various intensities targeting the tumor from different angles and is considered an effective strategy for metastatic EWS and OS [33]. SBRT used to control pulmonary metastases was reported to lead to a 2-year local control of 60% in 13 metastatic patients (IV) [33]. In osteosarcoma, the local control at 5 years was shown to range between 68 and 72% with conventionally fractionated proton RT doses of 68-70 Gy (1.8-2 Gy per day) in a retrospective study including 41 OS unresected or incompletely resected [36]. Carbon ion radiotherapy was effectively used in the treatment of unresectable pediatric osteosarcoma, with a local tumor control of 62% at 5 years in a retrospective study [34]. More recently, Combined Ion Beam Radiotherapy with protons and carbon ions in a multimodal treatment strategy of inoperable osteosarcoma was evaluated. Results showed an overall survival and a progression-free survival of 68% and 45%, respectively (2b). These results are particularly promising in craniofacial osteosarcoma [35]. Recently, a randomized controlled phase III study evaluated the efficacy of carbon ions, photon, and proton therapy in chordoma and chondrosarcoma (except skull-based tumors). This study will be extremely valuable in determining the benefits of using carbon ion radiotherapy as it is a prospective study and it compares the effects to a reference treatment [37].

Even though chemotherapy is a preferred treatment choice, RT plays a primordial role in the treatment of bone sarcomas. The development of new techniques makes RT an approach of interest for the treatment of incompletely or unresectable tumors, for tumors localized near critical structures, and for metastases. These new radiotherapies can lead to a better management of sarcoma patients who have an unfavorable prognosis and limited treatment options. With great advances in the development of targeted therapies, moving on to personalized combination approaches able to enhance the efficacy of radiotherapy, may be a promising strategy. To achieve this goal, a better understanding of radiotherapy mechanisms of action is necessary.

Potential target for combination with radiotherapy in bone sarcomas

Radiotherapy is currently focused on the precise delivery of high doses of radiation within the tumor bulk, sparing surrounding healthy tissues. However, the development of targeted therapy arguably has the potential to enhance radiotherapy efficacy. The possibility to molecularly profile tumors at diagnosis, together with improvements in radiotherapy could potentially pave the way for a more personalized approach to bone sarcoma treatment. Several key molecular pathways could theoretically enhance the therapeutic effect of radiation. In addition, it is important to determine the timing for combining molecular targeted therapy with radiation, as it could greatly affect the outcome depending on which pathway is being inhibited.

To determine which potential pathway could be a promising target in bone sarcomas, it is first necessary to review the radiation process and its consequences at the cellular and molecular levels. This paragraph summarizes, in chronological order, the principal steps and actors involved in the cellular response to radiotherapy (Fig. 1).

Fig. 1
figure 1

Schematic representation of the major known actors involved in radioresistance of bone sarcomas- when a damage occurs in DNA, ATM and ATR kinases are recruited and activate checkpoint kinases 1 and 2, leading to cell cycle arrest and to the recruitment of diverse effectors of DNA repair, such as the complex MRN PARP, RAD51, NBS1, RAD50. Diverse alterations in cell cycle proteins including p16 and CRIF, and in DNA repair proteins enhance bone sarcoma radioresistance. The accumulation of DNA damage is generally leads to cell death. However, bone sarcoma cells present defects in this pathway too, leading to cell survival after radiotherapy. Created using BioRender.com

Irrespective of the type of radiations used (e.g. X-rays, Proton, carbon ion), ionizing radiation affects all cellular compartments and their main target is DNA. Under ionizing radiations, micro-deposits of energy are generated in the nucleus near DNA. This accumulation of energy destabilizes and causes damage to the DNA structure. Moreover, by ionizing water molecules, a phenomenon known as water radiolysis, radiation triggers the formation of Reactive Oxygen Species (ROS) that lead to further DNA damage. DNA damage caused directly or indirectly by radiations, includes DNA oxidation, loss of a base, single-strand break and double-strand breaks [38, 39]. Double-Strand Breaks (DSB) are considered the most lethal type of lesions and are induced at a higher level by proton rather than photon therapy [40]. Each type of damage is recognized and corrected by specific repair mechanisms, each acting with a different degree of precision and speed (Table 5).

Table 5 Biological differences between photons, protons, and carbon ions

The response of a cancer cell to an ionizing radiation can be divided into several steps, from the recognition of the damage to the induction of cell death. At each step, bone sarcoma cells can have properties allowing them to counteract radiation-induced cell death, representing potential targets for combination therapy (Tables 6 and 7). Most of the studies on the biological effects of radiotherapy in bone sarcoma focus on X-rays or γ-rays, which will be presented in the next paragraph, and since very few studies (only 2 studies) deal with protons or carbon ions these will be presented when necessary.

Table 6 TP53 mutations in bone sarcomas
Table 7 Combination of radiotherapy and pharmacological inhibition of targets in bone sarcoma

DNA damage recognition

DNA damage is first recognized by 2 enzymes: Ataxia Telengiectasie Mutated (ATM) and Ataxia Telengiectasie RAD3-related (ATR). ATM recognizes double-strand breaks, while ATR can detect single-strand breaks and replication fork alterations. After the recognition of a DSB, ATM phosphorylates the histone H2AX (yH2AX), involved in stabilizing DNA extremities and in the recruitment of DNA repair complexes. ATM and ATR also phosphorylate the checkpoint kinases 1 and 2 (CHK1 and CHK2), leading to cell cycle arrest. ATR can phosphorylate many other substrates including Replication Fork components: MCM (MiniChromosome Maintenance) proteins, Rpa (Replication Protein A), polymerase, PCNA (Proliferating Cell Nuclear Antigen), and Claspin (Mrc1) [66, 67]. Cancer cells can resist radiation by increasing their efficiency in DNA repair through the increased expression of proteins involved in DNA damage recognition and repair, including ATM and ATR. A correlation was shown between radioresistance levels and the expression of 7 proteins involved in the DSB DNA repair machinery in 5 sarcoma cell lines, including one OS cell line. ATM, ATR and NBS (Nijmegen breakage syndrome protein 1), proteins involved in DNA damage recognition presented the strongest correlation [68]. In CD, an increased expression of ATM, ATR and yH2AX was observed in 26 patient samples in comparison with surrounding healthy tissue. However, this observation has not been directly correlated to the level of radioresistance of CD [69, 70]. Drugs targeting both ATR and ATM are already approved by the FDA and in clinical trials in other cancers (Bay1895344, NCT03188965; AZD1390, NCT03423628).

Once activated, ATM, CHK1 and CHK2 phosphorylate p53, the most studied tumor suppressing protein. P53 is the protein the most often mutated in all cancers and plays major roles in genomic stability, cell cycle regulation, cell death induction and in radioresistance.

P53 activation

P53 is a transcription factor that is stabilized following radiation and induces transcription of genes associated with cell cycle arrest, apoptosis, and metabolism, thereby functioning as a tumor suppressor [71]. Mutations affecting the normal functions of p53 are found in 80% of OS, 20% of CHS, and 10% of EWS (Table 6) [72]. Typically, the majority of TP53 mutations are missense mutations in its DNA binding domain, preventing TP53 from inducing transcription of its target genes and thus causing the loss of its tumor suppressive function [71]. In OS and CHS TP53 functions can also be altered indirectly through the amplification of Murine Double Minute 2 (MDM2) that results in P53 degradation. Recent results have demonstrated that TP53 mutations are associated with a radioresistant phenotype and poor survival in EWS patients [73]. TP53 is rarely mutated in CD; a whole genome sequencing study conducted on 63 CD samples revealed that only one sample carried a p53 mutation [74]. However, an increased expression of p53 was observed in 9/10 patients presenting relapsed tumors compared to patients with a stabilized disease. Thus, in chordoma overexpression of TP53 is correlated with tumor relapse and is a poor prognostic factor [75, 76]. Other studies are needed to understand the role of p53 in CD radioresistance.

If TP53 involvement in radioresistance is quite clear, further molecular studies are needed to precisely determine the underlying mechanisms of p53-driven radioresistance in bone sarcomas in terms of effectors and functions. In addition, although multiple p53 reactivators have been developed, only two drugs have entered clinical trials, APR-246 and COTI-2, currently making p53 hardly targetable.

Cell cycle arrest

Cell cycle regulation is a critical biological function involved in response to radiation. Arresting cell cycle progression is an essential step to enabling the recruitment of DNA repair machinery when DNA damage is caused by radiations. Several major actors of cell cycle regulation are involved in bone sarcoma radioresistance (Fig. 1). The gene Cyclin Dependent Kinase Inhibitor 2A (CDKN2A) encodes the P16 protein that inhibits Cyclin Dependent Kinases 4 and 6 (CDK4/6), inducing cell cycle arrest in G1 phase [77]. CDK4/6 usually bind to cyclin D1 and phosphorylate the tumor suppressor protein Rb1. The phosphorylation of Rb1 prevents its binding to the protein E2F, which in turn activates the transcription of genes allowing entry into the S phase [78].

The CDKN2A locus, is frequently deleted in bone sarcomas [74, 79,80,81]. The absence of p16 allows CDK4/6 activation and entry into the S phase of the cell cycle and could represent an advantage for cancer cells in response to radiation. These alterations could explain their low sensitivity to radiation. Pre-clinical studies refer to the synergistic effect of CDK4/6 inhibitors-radiotherapy combination. For instance, different clinical studies are ongoing in other cancers to determine the efficacy of combining radiation therapy and Palbociclib in breast cancer patients (NCT03691493, NCT03870919) and in locally advanced squamous cell carcinoma (NCT03024489). Further studies need to be done to determine the therapeutic potential of CDK4/6 inhibition in combination with radiotherapy.

Another protein involved in sarcoma radioresistance is CRIF, a protein regulating cell cycle. This protein phosphorylates CDK2, inducing cell cycle arrest and promoting DNA repair [82], a strong expression of CRIF has been detected in OS patient samples. CRIF inhibition by siRNA in both OS cell lines and OS xenografts was shown to increase sensitivity to irradiation, delay DNA damage repair, inactivate G1/S checkpoint, induce mitochondrial dysfunction and tumor regression in vivo [82]. Other strategies aimed at inhibiting cell cycle to reinduce radiosensitivity. In OS, the inhibition of PLK1 [5, 54], WEE1 [83], or PI3K [55] combined to radiotherapy generated cell growth arrest and cell death through mitotic catastrophe. Other studies are urgently needed to decipher the therapeutic potential of cell cycle gene alterations.

Once DNA damage is recognized and the cell cycle is arrested, the next step in cellular response to radiation is DNA repair.

DNA damage repair (DDR)

DNA repair involves a complex machinery and is orchestrated by numerous actors. Here, we will present the major DNA repair actors involved in the response of bone sarcoma to radiation-induced DSBs. For DSB DNA repair, two major pathways are activated: homologous recombination (HR) and non-homologous end joining (NHEJ).

NHEJ occurs during the G1 phase; it binds broken DNA extremities together leading to an increased number of errors. NHEJ initially recognizes DNA damage through a heterodimer Ku70-Ku80. This complex block 5’ DNA extremity and maintains DNA extremities close to each other to allow their binding. This complex also activates the protein 53BP1, which protects DNA extremities from more damage. γH2AX phosphorylation by ATM is also involved in stabilizing DNA extremities. The final steps following assembly of the repair machinery involve binding of DNA extremities by ligases (LIG4, XRCC4, and XLF) [84].

Homologous recombination (HR) only takes place in late S and G2 phases of the cell cycle, as this DNA repair mode is based on the use of the sister chromatid to synthesize an identical DNA strand. This reparation system is more precise than NHEJ. Here, The DNA DSB is recognized by the MRN complex composed of 3 proteins (MRE11, RAD50, NBS1), which initiate resection of DNA extremities in collaboration with CTIP. A loop with the sister chromatid is then formed and a DNA polymerase replicates DNA and ligases bind DNA to the strand break [84, 85]. Certain strategies aim at inhibiting DNA repair to induce cell death such as RAD51 inhibition, a recombinase involved in the DDR machinery. In OS and CD, the inhibition of RAD51, combined with radiations lead to a decreased cell proliferation and an increased apoptosis [86, 87]. In CHS and EWS, the PARP1 inhibitor Olaparib in combination with radiations was reported to decrease cell survival and clonogenic capacities [57, 60].

In this system, PARP-1 is rapidly recruited and activated by DNA DSBs. Upon activation, PARP-1 synthesizes a structurally complex polymer composed of ADP-ribose units that facilitate local chromatin relaxation and the recruitment of DNA repair factors [57]. In both CHS and EWS, PARP-1 seems to play a role in radioresistance. In 2 EWS cell lines, the combination of the PARP-1 inhibitor Olaparib and radiation therapy was more effective than radiotherapy or Olaparib alone. This combination induced a 4-fold increase in apoptosis in comparison with both treatments alone and lead to increased and sustained DNA damage in EWS cell lines. Moreover, in in vivo xenografts models of EWS, the combination of Olaparib and radiation therapy stopped tumor progression [57]. In the CHS cell line CH2879, Olaparib enhanced the efficacy of radiation by 1.3-fold for X rays, 1.8-fold for protons and 1.5-fold for carbon ions [60]. In a study of 11 advanced CD, a mutational signature associated with HR deficiency was found in 72.7% of samples, co-occurring with genomic instability. The use of Olaparib led to prolonged survival in a patient with refractory advanced CD [70]. Olaparib is currently being dose escalated in combination with radical (chemo-)radiotherapy regimens for non-small cell lung cancer (NSCLC), breast cancer and head and neck squamous cell carcinoma (HNSCC) in three parallel single institution phase 1 trials (Study protocols of three parallel phase 1 trials combining radical radiotherapy with the PARP inhibitor Olaparib).

After exposure to radiation, cells normally accumulate DNA damage that cannot be repaired fast enough and with enough precision for the cell to reenter the cell cycle. Proteins involved in genomic stability such as p53 then trigger cell death. However, sarcoma cells often lack the proteins supposed to control genomic integrity and present defects in cell death pathways [88].

Cell death

In response to DNA damage, apoptosis can be induced by different ways: i) activation of p53 or ii) accumulation of ROS. TP53 can directly promote cell death after DNA damage or after incomplete repair of DNA damage [89]. This is mediated through the activation of pro-apoptotic proteins, such as Tumor Necrosis factor Receptor superfamily (TNFR), triggering the extrinsic apoptosis pathway [90].

ROS accumulation can also induce cell death through the loss of mitochondrial membrane potential, leading to the release of cytochrome c. Moreover, ROS cause lipid damage, which activates sphyngomyelinase and induces the production and release of ceramide that in turn can activate caspases 1, 3 and 6, leading to cell death [91, 92].

An incomplete DNA repair can also induce a mitotic catastrophe, during which an abnormal chromosomal condensation occurs and the cell enters in mitosis before the end of S and G2 phases of the cell cycle [93].

Few studies have focused on the involvement of cell death defects in the response of bone sarcoma to radiotherapy. In CHS, the anti-apoptotic proteins Bcl-2, Bcl-xL and XIAP were found to be overexpressed in 2 CHS cell lines in comparison with 2 normal chondrocytes cell lines. When the expression of these anti-apoptotic proteins was inhibited by siRNA, a 10-fold increase in radiosensitivity was observed in CHS cell lines [94]. In EWS cell lines, an exposure to 2 to 10 Gy X-rays was reported to increase the expression of the anti-apoptotic protein survivin in a dose-dependent manner. Survivin inhibition by siRNA doubles apoptotic cell death [95, 96]. Several BH3 mimetics are currently used in the clinic, for example Venetoclax is approved for routine clinical practice in chronic lymphocytic leukemia (CLL) and acute myeloid leukemia (AML). To our knowledge, BH3-mimetics have not yet been combined to radiotherapy in patients.

Bone sarcomas arise in a particular environment (i.e. the bone or cartilage) and one of the characteristics of this environment is its hypoxic content that plays a role in resistance to conventional radiotherapy. Other factors of the tumor microenvironment, like the presence of immune cells or the extracellular matrix are likely involved in bone sarcoma radioresistance but studies regarding these are lacking.

Hypoxia

Hypoxia is a common feature of solid tumors, resulting from the imbalance between oxygen availability and consumption, and is defined as one of the most important causes of radiotherapy failure [97]. In bone sarcoma, the presence of hypoxic zones is correlated with tumor relapse, metastases and resistance to treatments [98,99,100,101,102]. These hypoxic zones are also predictive of poor tumor response to conventional radiotherapy. Different mechanisms have been suggested to explain the link between bone sarcoma, radioresistance and hypoxia. Evidence suggests that hypoxia inhibits the indirect effects of radiotherapy driven by the accumulation of ROS, creating more damage in cells which finally undergo cell death. The first mechanism proposed for hypoxia-induced radioresistance is the acceleration of ROS clearance. In a study including 35 OS and 20 EWS samples, it was shown that radiotherapy does not affect oxidative stress levels. However, it is known that radiotherapy induces ROS production which should increase oxidative stress. Hence, if oxidative stress levels remain constant, this implies that ROS clearance in the tumor cells is accelerated. The activation of autophagy and increased antioxidant metabolism are two hypotheses which can explain how sarcoma cells can accelerate ROS clearance. Indeed, it was demonstrated in OS that hypoxia confers cells resistance to radiation through activated autophagy to accelerate the clearance of cellular ROS products [103]. The increased antioxidant metabolism, mediated by the increase in two antioxidant enzymes, namely Aldehyde dehydrogenase (ALDH) 1 and 3, was shown in CD in an in vitro study. In this study, the pharmacological inhibition of the ALDH1 and 3 restored radiosensitivity to CD spheroid models in vitro [65]. Hypoxia-induced conventional radioresistance can potentially be counteracted by the addition of proton therapy, which has a higher efficacy in hypoxic zones (NCT02802969).

Other potential therapeutic targets with pre-clinical efficacy

Inhibition of histone deacetylases or demethylating agents has proven to be effective in combination with radiation, particularly in OS. Indeed, Histone DeACetylase (HDAC) inhibitors in combination with radiation was reported to increase cell death and DNA damages in several OS cell lines [46,47,48,49,50,51]. In CD and CHS, strategies targeting cancer-initiating cells (CIC) have been tested. One study highlighted that the use of disulfiram, an FDA-approved anti-alcoholism drug, complexed with Cu can radiosensitize CHS CIC. Indeed, the addition of DSF/Cu to a CHS cell line and a CD cell line decreased the clonogenicity of cells, while increasing apoptosis. Moreover, in an orthotopic model of CHS, the combination of DSF/Cu and radiation induced a strong decrease in tumor growth [61]. Similar results were obtained in CD, where the inhibition of ALDH1 and 3, proteins overactivated in CIC, radiosensitized 3D culture of CD cell lines [65]. Efforts need to be made to evaluate the potential of other radiosensitizing strategies. To do this, genetic inhibition of targets in combination with radiotherapy have been tested (Table 8).

Table 8 Combination of radiotherapy and genetic inhibition of targets in bone sarcoma

Future directions could also lead to the combination of immunomodulators and radiotherapy. It is now widely accepted that RT can trigger a systemic immune response supporting a strong rationale for the combination of RT and immunotherapy [110]. Radiations induce a series of biological effects including enhancing tumor antigen release and presentation, promoting priming and activation of immune cells, increasing density of tumor-infiltrating lymphocytes, facilitating recognition of tumor cells by T cells [110]. Combination of immunotherapy and radiotherapy has been evaluated in different solid tumors including melanoma, Non-Small Cell Lung Cancer and other solid tumors. The efficiency of Immune Checkpoint Inhibitors as single agents in bone sarcoma patients has been limited [111, 112]. Given the strong systemic anti-tumor immune effect induced by radiotherapy, an interesting rationale could be the combination of radiotherapy and immune checkpoint inhibitors. To our knowledge, no study has been reported in bone sarcoma concerning radiotherapy-induced anti-tumor immunity, or proof of concept of the combination of radiotherapy and immunotherapy so it would be crucial to investigate further pre-clinically the rationale and to determine efficient and precise biomarkers to predict and evaluate response to this kind of treatment.

Combination of radiotherapy and pharmacological/genetic inhibition of targets in bone sarcoma in clinical trials

Ongoing clinical trials combining drugs with radiotherapy are summarized in Table 9. In CD, 2 clinical trials show promising combinations. These trials evaluated the efficacy of a combination of an anti-brachyury vaccine with radiotherapy. Brachyury is involved in CD tumorigenesis, progression and poor prognosis, and the vaccine targeting brachyury as monotherapy is in phase I. The results of the phase I clinical trial of brachyury vaccine as monotherapy have demonstrated that brachyury vaccine induces a specific immune response. As radiotherapy can induce immunogenic cell death triggering a strong immune response, the combination of brachyury vaccine and radiotherapy could have a synergistic effect. Other studies combining different chemotherapy regimens with radiotherapy are being tested in OS and EWS.

Table 9 Clinical trials combining radiotherapy and FDA approved drugs in bone sarcoma

Other studies are necessary to test the efficacy of specific targeted therapy that could theoretically play a role in the response to radiotherapy. With the development of new radiotherapeutic approaches and their improved efficacy, specific studies deciphering the mechanistic action of these approaches in bone sarcoma would be not only interesting, but welcome to gain further insight into personalized medicine.

Toxicity & limitations

The improved efficacy of new radiotherapy techniques, such as proton beam or carbon ion therapy, offers new therapeutic perspectives in bone sarcoma. However, radiotherapy is still associated with short- and long-term toxicity, as described in Tables 2 and 4. Toxicity depends on the location of the tumor, and children are often particularly vulnerable to radiation-induced late toxicity and to secondary malignancies due to their immature tissue. In a cohort of 222 patients (151 skull-base CD and 71 CHS) treated post-operatively with proton therapy, long-term high grade (> 3) toxicity-free survival was 87%. High-grade late toxicity was characterized by optic neuropathy, temporal lobe necrosis with cerebellum brain parenchyma Grade 3 necrosis, spinal cord necrosis and unilateral hearing loss [113]. In spinal tumors, spinal cord toxicity and insufficiency fractures are the most common radiotherapy-associated side-effects observed [114]. In children pelvic Ewing sarcoma, radiation can cause pelvic pain, premature ovarian deficiency, unequal limb length due to slow bone growth [115]. Aside from radiotherapy toxicity, one major drawback in cancer patient treatment by radiotherapy is the cost and lack of accessibility with only 30 proton therapy centers in Europe.

Conclusion

Bone sarcomas are a group of rare and heterogenous tumors, affecting people of all ages. Surgery is still the mainstay of bone sarcoma patients’ treatment. However, due to the localization of the tumor and the co-morbidity associated with surgery, complete resection is often difficult. Radiotherapy is used in case of incomplete resection or for unresectable tumors.

In the last decades, there has been an improvement in radiotherapy, both in terms of methods of delivery and types of radiation used, leading to more important doses delivered to tumors and less toxicity for surrounding healthy tissue. Currently, retrospective cohorts, case–control studies and systematic reviews are the main studies evaluating the efficacy of radiotherapy in bone sarcoma. Thus high-quality, multicentric randomized controlled trials are desperately needed to precisely determine the benefits of radiotherapy in bone sarcoma. Efforts are ongoing to standardize the treatment in these rare diseases, regroup patients into adapted clinical trials, and improve patient management. A better understanding of the cellular and molecular mechanisms induced by radiotherapy could offer new therapeutic perspectives.

In vitro and in vivo pre-clinical data combining drugs and radiotherapy have shown promising results in bone sarcomas. However, it is important to remember that during the last decade, very few new drugs have been approved for concurrent radiotherapy administration in other cancers where pre-clinical data were also promising. Out of hundreds of clinical trials, only 2 compounds were finally approved for concurrent radiotherapy: the alkylating agent temozolomide and the anti-EGFR antibody cetuximab [116]. This highlights clear gaps between experimental models and the clinical reality that need to be addressed in bone sarcoma research. Efforts need to be made to improve translational research through in vitro and in vivo models to match radiotherapy specificities and challenges, but also through experimental design revision to unveil synergistic combinations. This need is particularly illustrated by the most recent studies showing the strong efficiency of immunotherapy combined to radiotherapy, even in immune desert tumors [117]. The tumor microenvironment plays a primordial role in tumor initiation and progression and a way to improve tumor modeling could be to reproduce the TME, both in vitro and in vivo. This could be of particular interest in CHS and CD, which are considered immune desert tumors, and where radiotherapy could reverse tumor immune desertification. Finally, strategies focusing on the delivery of targeted therapies and radiotherapy may also offer improved approaches in the treatment of bone sarcoma.

Availability of data and materials

Non applicable.

Abbreviations

OS:

Osteosarcoma

EWS:

Ewing Sarcoma

CHS:

Chondrosarcoma

CD:

Chordoma

TP53:

Tumor protein 53

FLI1:

Friend Leukemia Integration 1

IDH:

Isocitrate DeHydrogenase

RT:

Radiotherapy

IMRT:

Intensity Modulated RadioTherapy

SBRT:

Stereotactic Body Radiation Therapy

ROS:

Reactive Oxygen Species

ATM:

Ataxia Telengiectasie Mutated

ATR:

Ataxia Telengiectasie RAD3-related

H2AX:

H2A.X variant histone

CHK:

Checkpoint kinases

MCM:

MiniChromosome Maintenance

RPA:

Replication Protein A

PCNA:

Proliferating Cell Nuclear Antigen

MDM2:

Murine Double Minute 2

CDKN2A:

Cyclin Dependant Kinase Inhibitor 2A

CDK4/6:

Cyclin Dependant Kinase 4 and 6

Rb1:

Retinoblastoma protein 1

E2F:

E2 factor

RAD:

Recombinase

NBS1:

Nijmegen Breakage Syndrome 1

CRIF:

CR6 Interacting Factor

53BP1:

P53 binding protein 1

PRKDC:

Protein Kinase DNA-activated, Catalytic sub unit

LIG4:

DNA ligase 4

XRCC4:

X-Ray Repair Cross Complementing 4

XLF:

XRCC4 Like Factor

CTIP:

CtBP Interacting Protein

PARP-1:

Poly ADP ribose polymerase

Bcl proteins:

B cell lymphoma proteins

XIAP:

X-linked inhibitor of apoptosis

TNFSFR:

Tumor Necrosis Factor Superfamily Receptor

PLK1:

Polo Like Kinase 1

WEE1:

G2 checkpoint kinase

PI3K:

Phosphoinositide 3 phosphate

HDAC:

Histone DeACetylase

iNOS:

Nitric Oxide Synthase

UBE2T:

Ubiquitin-conjugating enzyme E2 T

Akt:

Protein kinase B

IGF2R:

Insulin Growth Factor Receptor

References

  1. Soft Tissue and Bone Tumours. WHO Classification of Tumours, Volume 3. 5th ed. WHO Classification of Tumours Editorial Board; 2020.

  2. Luetke A, Meyers PA, Lewis I, Juergens H. Osteosarcoma treatment - where do we stand? A state of the art review. Cancer Treat Rev. 2014;40:523–32. https://doi.org/10.1016/j.ctrv.2013.11.006.

    Article  PubMed  Google Scholar 

  3. Strauss SJ, Frezza AM, Abecassis N, Bajpai J, Bauer S, Biagini R, Bielack S, Blay JY, Bolle S, Bonvalot S, et al. Bone sarcomas: ESMO-EURACAN-GENTURIS-ERN PaedCan clinical practice guideline for diagnosis, treatment and follow-Up. Ann Oncol. 2021;32:1520–36. https://doi.org/10.1016/j.annonc.2021.08.1995.

    Article  PubMed  CAS  Google Scholar 

  4. Riedel RF, Larrier N, Dodd L, Kirsch D, Martinez S, Brigman BE. The clinical management of chondrosarcoma. Curr Treat Options Oncol. 2009;10:94–106. https://doi.org/10.1007/s11864-009-0088-2.

    Article  PubMed  Google Scholar 

  5. Heery CR. Chordoma: the quest for better treatment options. Oncol Ther. 2016;4:35–51. https://doi.org/10.1007/s40487-016-0016-0.

    Article  PubMed  Google Scholar 

  6. Radaelli S, Fossati P, Stacchiotti S, Akiyama T, Asencio JM, Bandiera S, Boglione A, Boland P, Bolle S, Bruland Ø, et al. The sacral chordoma margin. Eur J Surg Oncol. 2020. https://doi.org/10.1016/j.ejso.2020.04.028.

    Article  PubMed  Google Scholar 

  7. Stacchiotti S, Sommer J. Chordoma global consensus group building a global consensus approach to chordoma: a position paper from the medical and patient community. Lancet Oncol. 2015;16:e71–83. https://doi.org/10.1016/S1470-2045(14)71190-8.

    Article  PubMed  Google Scholar 

  8. Stacchiotti S, Gronchi A, Fossati P, Akiyama T, Alapetite C, Baumann M, Blay JY, Bolle S, Boriani S, Bruzzi P, et al. Best practices for the management of local-regional recurrent chordoma: a position paper by the chordoma global consensus group. Ann Oncol. 2017;28:1230–42. https://doi.org/10.1093/annonc/mdx054.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Dial BL, Kerr DL, Lazarides AL, Catanzano AA, Green CL, Risoli T Jr, Blazer DG, Goodwin CR, Brigman BE, Eward WC, et al. The role of radiotherapy for chordoma patients managed with surgery: analysis of the national cancer database. Spine (Phila Pa 1976). 2020;45:E742–51. https://doi.org/10.1097/BRS.0000000000003406.

    Article  PubMed  Google Scholar 

  10. Krochak R, Harwood AR, Cummings BJ, Quirt IC. Results of radical radiation for chondrosarcoma of bone. Radiother Oncol. 1983;1:109–15. https://doi.org/10.1016/s0167-8140(83)80014-0.

    Article  PubMed  CAS  Google Scholar 

  11. McNaney D, Lindberg RD, Ayala AG, Barkley HT, Hussey DH. Fifteen year radiotherapy experience with chondrosarcoma of bone. Int J Radiat Oncol Biol Phys. 1982;8:187–90. https://doi.org/10.1016/0360-3016(82)90512-0.

    Article  PubMed  CAS  Google Scholar 

  12. Fujiwara T, Tsuda Y, Stevenson J, Parry M, Jeys L. Sacral chordoma: do the width of surgical margin and the use of photon/proton radiotherapy affect local disease control? Int Orthop. 2020;44:381–9. https://doi.org/10.1007/s00264-019-04460-5.

    Article  PubMed  Google Scholar 

  13. Ares C, Hug EB, Lomax AJ, Bolsi A, Timmermann B, Rutz HP, Schuller JC, Pedroni E, Goitein G. Effectiveness and safety of spot scanning proton radiation therapy for chordomas and chondrosarcomas of the skull base: first long-term report. Int J Radiat Oncol Biol Phys. 2009;75:1111–8. https://doi.org/10.1016/j.ijrobp.2008.12.055.

    Article  PubMed  Google Scholar 

  14. Ciernik IF, Niemierko A, Harmon DC, Kobayashi W, Chen Y-L, Yock TI, Ebb DH, Choy E, Raskin KA, Liebsch N, et al. Proton-based radiotherapy for unresectable or incompletely resected osteosarcoma. Cancer. 2011;117:4522–30. https://doi.org/10.1002/cncr.26037.

    Article  PubMed  Google Scholar 

  15. Zabel-du Bois A, Nikoghosyan A, Schwahofer A, Huber P, Schlegel W, Debus J, Milker-Zabel S. Intensity modulated radiotherapy in the management of sacral chordoma in primary versus recurrent disease. Radiother Oncol. 2010;97:408–12. https://doi.org/10.1016/j.radonc.2010.10.008.

    Article  PubMed  Google Scholar 

  16. Chen Y-L, Liebsch N, Kobayashi W, Goldberg S, Kirsch D, Calkins G, Childs S, Schwab J, Hornicek F, DeLaney T. Definitive high-dose photon/proton radiotherapy for unresected mobile spine and sacral chordomas. Spine (Phila Pa 1976). 2013;38:E930–936. https://doi.org/10.1097/BRS.0b013e318296e7d7.

    Article  PubMed  Google Scholar 

  17. Alahmari M, Temel Y. Skull base chordoma treated with proton therapy: a systematic review. Surg Neurol Int. 2019;10:96. https://doi.org/10.25259/SNI-213-2019.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Frezza AM, Botta L, Trama A, Dei Tos AP, Stacchiotti S. Chordoma: update on disease, epidemiology, biology and medical therapies. Curr Opin Oncol. 2019;31:114–20. https://doi.org/10.1097/CCO.0000000000000502.

    Article  PubMed  Google Scholar 

  19. Catanzano AA, Kerr DL, Lazarides AL, Dial BL, Lane WO, Blazer DG, Larrier NA, Kirsch DG, Brigman BE, Eward WC. Revisiting the role of radiation therapy in chondrosarcoma: a national cancer database study. Sarcoma. 2019;2019:4878512. https://doi.org/10.1155/2019/4878512.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Zhou J, Yang B, Wang X, Jing Z. Comparison of the effectiveness of radiotherapy with photons and particles for chordoma after surgery: a meta-analysis. World Neurosurg. 2018;117:46–53. https://doi.org/10.1016/j.wneu.2018.05.209.

    Article  PubMed  Google Scholar 

  21. Gao Z, Lu T, Song H, Gao Z, Ren F, Ouyang P, Wang Y, Zhu J, Zhou S, He X. Prognostic factors and treatment options for patients with high-grade chondrosarcoma. Med Sci Monit. 2019;25:8952–67. https://doi.org/10.12659/MSM.917959.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Kabolizadeh P, Chen Y-L, Liebsch N, Hornicek FJ, Schwab JH, Choy E, Rosenthal DI, Niemierko A, DeLaney TF. Updated outcome and analysis of tumor response in mobile spine and sacral chordoma treated with definitive high-dose photon/proton radiation therapy. Int J Radiat Oncol Biol Phys. 2017;97:254–62. https://doi.org/10.1016/j.ijrobp.2016.10.006.

    Article  PubMed  Google Scholar 

  23. Palm RF, Oliver DE, Yang GQ, Abuodeh Y, Naghavi AO, Johnstone PAS. The role of dose escalation and proton therapy in perioperative or definitive treatment of chondrosarcoma and chordoma: an analysis of the national cancer data base. Cancer. 2019;125:642–51. https://doi.org/10.1002/cncr.31958.

    Article  PubMed  CAS  Google Scholar 

  24. Lu VM, O’Connor KP, Mahajan A, Carlson ML, Van Gompel JJ. Carbon ion radiotherapy for skull base chordomas and chondrosarcomas: a systematic review and meta-analysis of local control, survival, and toxicity outcomes. J Neurooncol. 2020;147:503–13. https://doi.org/10.1007/s11060-020-03464-1.

    Article  PubMed  CAS  Google Scholar 

  25. Imai R, Kamada T, Araki N, WORKING GROUP FOR BONE and SOFT-TISSUE SARCOMAS. Clinical efficacy of carbon ion radiotherapy for unresectable chondrosarcomas. Anticancer Res. 2017;37:6959–64. https://doi.org/10.21873/anticanres.12162.

    Article  PubMed  CAS  Google Scholar 

  26. Wu H, Yu J, Kong D, Xu Y, Zhang Z, Shui J, Li Z, Luo H, Wang K. Population and single-cell transcriptome analyses reveal diverse transcriptional changes associated with radioresistance in esophageal squamous cell carcinoma. Int J Oncol. 2019;55:1237–48. https://doi.org/10.3892/ijo.2019.4897.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Lockney DT, Shub T, Hopkins B, Lockney NA, Moussazadeh N, Lis E, Yamada Y, Schmitt AM, Higginson DS, Laufer I, et al. Spinal stereotactic body radiotherapy following intralesional curettage with separation surgery for initial or salvage chordoma treatment. Neurosurg Focus. 2017;42:E4. https://doi.org/10.3171/2016.9.FOCUS16373.

    Article  PubMed  Google Scholar 

  28. Nakamura R, Sugawara J, Yamaguchi S, Kakuhara H, Kikuchi K, Ariga H. Stereotactic body radiotherapy with a single isocentre for multiple pulmonary metastases. BJR Case Rep. 2020;6:20190121. https://doi.org/10.1259/bjrcr.20190121.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Vasudevan HN, Raleigh DR, Johnson J, Garsa AA, Theodosopoulos PV, Aghi MK, Ames C, McDermott MW, Barani IJ, Braunstein SE. Management of chordoma and chondrosarcoma with fractionated stereotactic radiotherapy. Front Surg. 2017;4:35. https://doi.org/10.3389/fsurg.2017.00035.

    Article  PubMed  PubMed Central  Google Scholar 

  30. DuBois SG, Krailo MD, Gebhardt MC, Donaldson SS, Marcus KJ, Dormans J, Shamberger RC, Sailer S, Nicholas RW, Healey JH, et al. Comparative evaluation of local control strategies in localized ewing sarcoma of bone: a report from the children’s oncology group. Cancer. 2015;121:467–75. https://doi.org/10.1002/cncr.29065.

    Article  PubMed  Google Scholar 

  31. Haeusler J, Ranft A, Boelling T, Gosheger G, Braun-Munzinger G, Vieth V, Burdach S, van den Berg H, Juergens H, Dirksen U. The value of local treatment in patients with Primary, Disseminated, Multifocal Ewing Sarcoma (PDMES). Cancer. 2010;116:443–50. https://doi.org/10.1002/cncr.24740.

    Article  PubMed  Google Scholar 

  32. DeLaney TF, Liebsch NJ, Pedlow FX, Adams J, Weyman EA, Yeap BY, Depauw N, Nielsen GP, Harmon DC, Yoon SS, et al. Long-term results of phase II study of high dose photon/proton radiotherapy in the management of spine chordomas, chondrosarcomas, and other sarcomas. J Surg Oncol. 2014;110:115–22. https://doi.org/10.1002/jso.23617.

    Article  PubMed  Google Scholar 

  33. Brown LC, Lester RA, Grams MP, Haddock MG, Olivier KR, Arndt CAS, Rose PS, Laack NN. Stereotactic body radiotherapy for metastatic and recurrent Ewing sarcoma and osteosarcoma. Sarcoma. 2014;2014:418270. https://doi.org/10.1155/2014/418270.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Mohamad O, Imai R, Kamada T, Nitta Y, Araki N. Carbon ion radiotherapy for inoperable pediatric osteosarcoma. Oncotarget. 2018;9:22976–85. https://doi.org/10.18632/oncotarget.25165.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Seidensaal K, Mattke M, Haufe S, Rathke H, Haberkorn U, Bougatf N, Kudak A, Blattmann C, Oertel S, Kirchner M, et al. The Role of Combined Ion-Beam Radiotherapy (CIBRT) with Protons and Carbon Ions in a Multimodal Treatment Strategy of Inoperable Osteosarcoma. Radiother Oncol. 2021;159:8–16. https://doi.org/10.1016/j.radonc.2021.01.029.

    Article  PubMed  CAS  Google Scholar 

  36. DeLaney TF, Park L, Goldberg SI, Hug EB, Liebsch NJ, Munzenrider JE, Suit HD. Radiotherapy for local control of osteosarcoma. Int J Radiat Oncol Biol Phys. 2005;61:492–8. https://doi.org/10.1016/j.ijrobp.2004.05.051.

    Article  PubMed  Google Scholar 

  37. Balosso J, Febvey-Combes O, Iung A, Lozano H, Alloh AS, Cornu C, Hervé M, Akkal Z, Lièvre M, Plattner V, et al. A randomized controlled phase III study comparing hadrontherapy with carbon ions versus conventional radiotherapy - including photon and proton therapy - for the treatment of radioresistant tumors: the ETOILE trial. BMC Cancer. 2022;22:575. https://doi.org/10.1186/s12885-022-09564-7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Emami B, Lyman J, Brown A, Coia L, Goitein M, Munzenrider JE, Shank B, Solin LJ, Wesson M. Tolerance of normal tissue to therapeutic irradiation. Int J Radiat Oncol Biol Phys. 1991;21:109–22. https://doi.org/10.1016/0360-3016(91)90171-y.

    Article  PubMed  CAS  Google Scholar 

  39. Maier P, Wenz F, Herskind C. Radioprotection of normal tissue cells. Strahlenther Onkol. 2014;190:745–52. https://doi.org/10.1007/s00066-014-0637-x.

    Article  PubMed  Google Scholar 

  40. Vitti ET, Parsons JL. The radiobiological effects of proton beam therapy: Impact on DNA Damage and Repair. Cancers (Basel). 2019;11:946. https://doi.org/10.3390/cancers11070946.

    Article  PubMed  CAS  Google Scholar 

  41. Kim EH, Kim M-S, Lee K-H, Koh J-S, Jung W-G, Kong C-B. Zoledronic acid is an effective radiosensitizer in the treatment of osteosarcoma. Oncotarget. 2016;7:70869–80. https://doi.org/10.18632/oncotarget.12281.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Sawai Y, Murata H, Horii M, Koto K, Matsui T, Horie N, Tsuji Y, Ashihara E, Maekawa T, Kubo T, et al. Effectiveness of sulforaphane as a radiosensitizer for murine osteosarcoma cells. Oncol Rep. 2013;29:941–5. https://doi.org/10.3892/or.2012.2195.

    Article  PubMed  CAS  Google Scholar 

  43. Zhang X-Y, Sun K, Zhu Q, Song T, Liu Y. Ginseng polysaccharide serves as a potential radiosensitizer through inducing apoptosis and autophagy in the treatment of osteosarcoma. Kaohsiung J Med Sci. 2017;33:535–42. https://doi.org/10.1016/j.kjms.2017.07.001.

    Article  PubMed  CAS  Google Scholar 

  44. Bogado RFE, Pezuk JA, de Oliveira HF, Tone LG, Brassesco MS. BI 6727 and GSK461364 suppress growth and radiosensitize osteosarcoma cells, but show limited cytotoxic effects when combined with conventional treatments. Anticancer Drugs. 2015;26:56–63. https://doi.org/10.1097/CAD.0000000000000157.

    Article  PubMed  CAS  Google Scholar 

  45. Mamo T, Mladek AC, Shogren KL, Gustafson C, Gupta SK, Riester SM, Maran A, Galindo M, van Wijnen AJ, Sarkaria JN, et al. Inhibiting DNA-PKCS radiosensitizes human osteosarcoma cells. Biochem Biophys Res Commun. 2017;486:307–13. https://doi.org/10.1016/j.bbrc.2017.03.033.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Blattmann C, Thiemann M, Stenzinger A, Christmann A, Roth E, Ehemann V, Debus J, Kulozik AE, Weichert W, Huber PE, et al. Radiosensitization by histone deacetylase inhibition in an osteosarcoma mouse model. Strahlenther Onkol. 2013;189:957–66. https://doi.org/10.1007/s00066-013-0372-8.

    Article  PubMed  CAS  Google Scholar 

  47. Ogawa Y, Takahashi T, Kobayashi T, Kariya S, Nishioka A, Ohnishi T, Saibara T, Hamasato S, Tani T, Seguchi H, et al. Apoptotic-resistance of the human osteosarcoma cell line HS-Os-1 to irradiation is converted to apoptotic-susceptibility by hydrogen peroxide: a potent role of hydrogen peroxide as a new radiosensitizer. Int J Mol Med. 2003;12:845–50.

    PubMed  CAS  Google Scholar 

  48. Liu G, Wang H, Zhang F, Tian Y, Tian Z, Cai Z, Lim D, Feng Z. The effect of VPA on increasing radiosensitivity in osteosarcoma cells and primary-culture cells from chemical carcinogen-induced breast cancer in rats. Int J Mol Sci. 2017;18:E1027. https://doi.org/10.3390/ijms18051027.

    Article  CAS  Google Scholar 

  49. Johnson AM, Bennett PV, Sanidad KZ, Hoang A, Jardine JH, Keszenman DJ, Wilson PF. Evaluation of histone deacetylase inhibitors as radiosensitizers for proton and light ion radiotherapy. Front Oncol. 2021;11:735940. https://doi.org/10.3389/fonc.2021.735940.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Blattmann C, Oertel S, Ehemann V, Thiemann M, Huber PE, Bischof M, Witt O, Deubzer HE, Kulozik AE, Debus J, et al. Enhancement of radiation response in osteosarcoma and rhabdomyosarcoma cell lines by histone deacetylase inhibition. Int J Radiat Oncol Biol Phys. 2010;78:237–45. https://doi.org/10.1016/j.ijrobp.2010.03.010.

    Article  PubMed  CAS  Google Scholar 

  51. Li Y, Geng P, Jiang W, Wang Y, Yao J, Lin X, Liu J, Huang L, Su B, Chen H. Enhancement of radiosensitivity by 5-Aza-CdR through activation of G2/M checkpoint response and apoptosis in osteosarcoma cells. Tumour Biol. 2014;35:4831–9. https://doi.org/10.1007/s13277-014-1634-5.

    Article  PubMed  CAS  Google Scholar 

  52. Berberine Enhances the Radiosensitivity of Osteosarcoma by Targeting Rad51 and Epithelial-Mesenchymal Transition - PubMed Available online: https://pubmed.ncbi.nlm.nih.gov/32474504/ (Accessed on 11 Oct 2021).

  53. Brassesco MS, Pezuk JA, de Oliveira JC, Valera ET, de Oliveira HF, Scrideli CA, Umezawa K, Tone LG. Activator protein-1 inhibition by 3-[(Dodecylthiocarbonyl)Methyl]-glutamaride impairs invasion and radiosensitizes osteosarcoma cells in vitro. Cancer Biother Radiopharm. 2013;28:351–8. https://doi.org/10.1089/cbr.2012.1305.

    Article  PubMed  CAS  Google Scholar 

  54. Lund-Andersen C, Patzke S, Nähse-Kumpf V, Syljuåsen RG. PLK1-inhibition can cause radiosensitization or radioresistance dependent on the treatment schedule. Radiother Oncol. 2014;110:355–61. https://doi.org/10.1016/j.radonc.2013.12.014.

    Article  PubMed  CAS  Google Scholar 

  55. Kubota N, Ozawa F, Okada S, Inada T, Komatsu K, Okayasu R. The phosphatidylinositol 3-kinase inhibitor Wortmannin sensitizes quiescent but not proliferating MG-63 human osteosarcoma cells to radiation. Cancer Lett. 1998;133:161–7. https://doi.org/10.1016/s0304-3835(98)00221-3.

    Article  PubMed  CAS  Google Scholar 

  56. Lin MY, Damron TA, Oest ME, Horton JA. Mithramycin a radiosensitizes EWS:Fli1+ Ewing sarcoma cells by inhibiting double strand break repair. Int J Radiat Oncol Biol Phys. 2021;109:1454–71. https://doi.org/10.1016/j.ijrobp.2020.12.010.

    Article  PubMed  Google Scholar 

  57. Lee H-J, Yoon C, Schmidt B, Park DJ, Zhang AY, Erkizan HV, Toretsky JA, Kirsch DG, Yoon SS. Combining PARP-1 inhibition and radiation in Ewing sarcoma results in lethal DNA damage. Mol Cancer Ther. 2013;12:2591–600. https://doi.org/10.1158/1535-7163.MCT-13-0338.

    Article  PubMed  CAS  Google Scholar 

  58. Veeraraghavan J, Natarajan M, Herman TS, Aravindan N. Curcumin-Altered P53-response genes regulate radiosensitivity in P53-Mutant Ewing’s sarcoma cells. Anticancer Res. 2010;30:4007–15.

    PubMed  CAS  Google Scholar 

  59. Attawia MA, Borden MD, Herbert KM, Katti DS, Asrari F, Uhrich KE, Laurencin CT. Regional drug delivery with radiation for the treatment of Ewing’s sarcoma. In vitro development of a Taxol release system. J Control Release. 2001;71:193–202. https://doi.org/10.1016/s0168-3659(01)00217-6.

    Article  PubMed  CAS  Google Scholar 

  60. Césaire M, Ghosh U, Austry J-B, Muller E, Cammarata FP, Guillamin M, Caruso M, Castéra L, Petringa G, Cirrone GAP, et al. Sensitization of chondrosarcoma cells with PARP inhibitor and high-LET radiation. J Bone Oncol. 2019;17:100246. https://doi.org/10.1016/j.jbo.2019.100246.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Wang K, Michelakos T, Wang B, Shang Z, DeLeo AB, Duan Z, Hornicek FJ, Schwab JH, Wang X. Targeting cancer stem cells by disulfiram and copper sensitizes radioresistant chondrosarcoma to radiation. Cancer Lett. 2021;505:37–48. https://doi.org/10.1016/j.canlet.2021.02.002.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Saeed A, Singh P, Malyapa RS, Mahmood J, Vujaskovic Z. Hyperthermia induces radiosensitization and leads to reduced brachyury levels in chordoma cells. Int J Radiat Oncol Biol Phys. 2020;108:e518. https://doi.org/10.1016/j.ijrobp.2020.07.1626.

    Article  Google Scholar 

  63. Huq S, Kedda J, Zhao T, Serra R, Ding A, Morales M, Ehresman J, Brem H, Gallia GL, Sciubba DM et al. Repositioning the antiviral drug ribavirin as a radiosensitizing agent in chordoma. Neurosurgery. 2020;67, https://doi.org/10.1093/neuros/nyaa447_887.

  64. Hao S, Song H, Zhang W, Seldomridge A, Jung J, Giles AJ, Hutchinson M-K, Cao X, Colwell N, Lita A, et al. Protein phosphatase 2A inhibition enhances radiation sensitivity and reduces tumor growth in chordoma. Neuro Oncol. 2018;20:799–809. https://doi.org/10.1093/neuonc/nox241.

    Article  PubMed  CAS  Google Scholar 

  65. Locquet M-A, Dechaume A-L, Berchard P, Abbes L, Pissaloux D, Tirode F, Ramos I, Bedoucha J, Valantin J, Karanian M, et al. Aldehyde dehydrogenase, a therapeutic target in chordoma: analysis in 3D cellular models. Cells. 2021;10:399. https://doi.org/10.3390/cells10020399.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Shiloh Y. ATM and related protein kinases: safeguarding genome integrity. Nat Rev Cancer. 2003;3:155–68. https://doi.org/10.1038/nrc1011.

    Article  PubMed  CAS  Google Scholar 

  67. Abraham RT. Cell cycle checkpoint signaling through the ATM and ATR Kinases. Genes Dev. 2001;15:2177–96. https://doi.org/10.1101/gad.914401.

    Article  PubMed  CAS  Google Scholar 

  68. Ernst A, Anders H, Kapfhammer H, Orth M, Hennel R, Seidl K, Winssinger N, Belka C, Unkel S, Lauber K. HSP90 inhibition as a means of radiosensitizing resistant, aggressive soft tissue sarcomas. Cancer Lett. 2015;365:211–22. https://doi.org/10.1016/j.canlet.2015.05.024.

    Article  PubMed  CAS  Google Scholar 

  69. Zhang C, Wang B, Li L, Li Y, Li P, Lv G. Radioresistance of chordoma cells is associated with the ATM/ATR pathway, in which RAD51 serves as an important downstream effector. Exp Ther Med. 2017;14:2171–9. https://doi.org/10.3892/etm.2017.4736.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Gröschel S, Hübschmann D, Raimondi F, Horak P, Warsow G, Fröhlich M, Klink B, Gieldon L, Hutter B, Kleinheinz K, et al. Defective homologous recombination DNA repair as therapeutic target in advanced chordoma. Nat Commun. 2019;10:1–9. https://doi.org/10.1038/s41467-019-09633-9.

    Article  CAS  Google Scholar 

  71. Thoenen E, Curl A, Iwakuma T. TP53 in bone and soft tissue sarcomas. Pharmacol Ther. 2019;202:149–64. https://doi.org/10.1016/j.pharmthera.2019.06.010.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Casey DL, Pitter KL, Wexler L, Slotkin E, Gupta GP, Wolden SL. P53 pathway mutations increase radioresistance in rhabdomyosarcoma and Ewing sarcoma. Int J Radiat Oncol Biol Phys. 2020;108:S179. https://doi.org/10.1016/j.ijrobp.2020.07.963.

    Article  Google Scholar 

  73. Casey DL, Pitter KL, Wexler LH, Slotkin EK, Gupta GP, Wolden SL. TP53 mutations increase radioresistance in rhabdomyosarcoma and Ewing sarcoma. Br J Cancer. 2021;125:576–81. https://doi.org/10.1038/s41416-021-01438-2.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Tarpey PS, Behjati S, Young MD, Martincorena I, Alexandrov LB, Farndon SJ, Guzzo C, Hardy C, Latimer C, Butler AP, et al. The driver landscape of sporadic chordoma. Nat Commun. 2017;8:890. https://doi.org/10.1038/s41467-017-01026-0.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Matsuno A, Sasaki T, Nagashima T, Matsuura R, Tanaka H, Hirakawa M, Murakami M, Kirino T. Immunohistochemical examination of proliferative potentials and the expression of cell cycle-related proteins of intracranial chordomas. Hum Pathol. 1997;28:714–9. https://doi.org/10.1016/s0046-8177(97)90181-7.

    Article  PubMed  CAS  Google Scholar 

  76. Sakai K, Hongo K, Tanaka Y, Nakayama J. Analysis of immunohistochemical expression of P53 and the proliferation marker Ki-67 antigen in skull base chordomas: relationships between their expression and prognosis. Brain Tumor Pathol. 2007;24:57–62. https://doi.org/10.1007/s10014-007-0222-4.

    Article  PubMed  CAS  Google Scholar 

  77. Weinberg WC, Denning MF. P21Waf1 control of epithelial cell cycle and cell fate. Crit Rev Oral Biol Med. 2002;13:453–64. https://doi.org/10.1177/154411130201300603.

    Article  PubMed  Google Scholar 

  78. Jackson SP, Bartek J. The DNA-damage response in human biology and disease. Nature. 2009;461:1071–8. https://doi.org/10.1038/nature08467.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Lerman DM, Monument MJ, McIlvaine E, Liu X, Huang D, Monovich L, Beeler N, Gorlick RG, Marina NM, Womer RB, et al. Tumoral TP53 and/or CDKN2A alterations are not reliable prognostic biomarkers in patients with localized Ewing sarcoma: a report from the children’s oncology group. Pediatr Blood Cancer. 2015;62:759–65. https://doi.org/10.1002/pbc.25340.

    Article  PubMed  CAS  Google Scholar 

  80. Mohseny AB, Szuhai K, Romeo S, Buddingh EP, Briaire-de Bruijn I, de Jong D, van Pel M, Cleton-Jansen AM, Hogendoorn PCW. Osteosarcoma originates from mesenchymal stem cells in consequence of aneuploidization and genomic loss of Cdkn2. J Pathol. 2009;219:294–305. https://doi.org/10.1002/path.2603.

    Article  PubMed  CAS  Google Scholar 

  81. de Jong Y, Ingola M, Briaire-de Bruijn IH, Kruisselbrink AB, Venneker S, Palubeckaite I, Heijs BP, Cleton-Jansen AM, Haas RL, Bovée JV. Radiotherapy resistance in chondrosarcoma cells; a possible correlation with alterations in cell cycle related genes. Clin Sarcoma Res. 2019;9:9. https://doi.org/10.1186/s13569-019-0119-0.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Ran Q, Jin F, Xiang Y, Xiang L, Wang Q, Li F, Chen L, Zhang Y, Wu C, Zhou L, et al. CRIF1 as a potential target to improve the radiosensitivity of osteosarcoma. Proc Natl Acad Sci U S A. 2019;116:20511–6. https://doi.org/10.1073/pnas.1906578116.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Caretti V, Hiddingh L, Lagerweij T, Schellen P, Koken PW, Hulleman E, van Vuurden DG, Vandertop WP, Kaspers GJL, Noske DP, et al. WEE1 Kinase inhibition enhances the radiation response of diffuse intrinsic pontine gliomas. Mol Cancer Ther. 2013;12:141–50. https://doi.org/10.1158/1535-7163.MCT-12-0735.

    Article  PubMed  CAS  Google Scholar 

  84. Chang HHY, Pannunzio NR, Adachi N, Lieber MR. Non-homologous DNA end joining and alternative pathways to double-strand break repair. Nat Rev Mol Cell Biol. 2017;18:495–506. https://doi.org/10.1038/nrm.2017.48.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. Branzei D, Foiani M. Regulation of DNA repair throughout the cell cycle. Nat Rev Mol Cell Biol. 2008;9:297–308. https://doi.org/10.1038/nrm2351.

    Article  PubMed  CAS  Google Scholar 

  86. Du L, Bai J, Liu Q, Wang Y, Zhao P, Chen F, Wang H, Fan F. Correlation of RAD51 and radiosensitization of methotrexate. Chin J Radiol Med Protect. 2012:44–6.

  87. Zhang H-T, Yang J, Liang G-H, Gao X-J, Sang Y, Gui T, Liang Z-J, Tam M-S, Zha Z-G. Andrographolide induces cell cycle arrest and apoptosis of chondrosarcoma by targeting TCF-1/SOX9 axis. J Cell Biochem. 2017. https://doi.org/10.1002/jcb.26122.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Rello-Varona S, Herrero-Martín D, Lagares-Tena L, López-Alemany R, Mulet-Margalef N, Huertas-Martínez J, Garcia-Monclús S, García del Muro X, Muñoz-Pinedo C, Tirado OM. The importance of being dead: cell death mechanisms assessment in anti-sarcoma therapy. Front Oncol. 2015;5:82. https://doi.org/10.3389/fonc.2015.00082.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Vousden KH. P53: death star. Cell. 2000;103:691–4. https://doi.org/10.1016/s0092-8674(00)00171-9.

    Article  PubMed  CAS  Google Scholar 

  90. Harms K, Nozell S, Chen X. The common and distinct target genes of the P53 family transcription factors. Cell Mol Life Sci. 2004;61:822–42. https://doi.org/10.1007/s00018-003-3304-4.

    Article  PubMed  CAS  Google Scholar 

  91. Popadiuk S, Renke J, Woźniak M, Korzon M. Does chemotherapy and radiotherapy influence the level of oxidative stress in children with malignant bone tumours? Med Wieku Rozwoj. 2006;10:855–9.

    PubMed  Google Scholar 

  92. Kolesnick R. The therapeutic potential of modulating the ceramide/sphingomyelin pathway. J Clin Invest. 2002;110:3–8. https://doi.org/10.1172/JCI16127.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Castedo M, Perfettini J-L, Roumier T, Andreau K, Medema R, Kroemer G. Cell death by mitotic catastrophe: a molecular definition. Oncogene. 2004;23:2825–37. https://doi.org/10.1038/sj.onc.1207528.

    Article  PubMed  CAS  Google Scholar 

  94. Kim DW, Seo SW, Cho SK, Chang SS, Lee HW, Lee SE, Block JA, Hei TK, Lee FY. Targeting of cell survival genes using Small Interfering RNAs (SiRNAs) enhances radiosensitivity of grade II chondrosarcoma cells. J Orthop Res. 2007;25:820–8. https://doi.org/10.1002/jor.20377.

    Article  PubMed  CAS  Google Scholar 

  95. Greve B, Sheikh-Mounessi F, Kemper B, Ernst I, Götte M, Eich HT. Survivin, a target to modulate the radiosensitivity of Ewing’s sarcoma. Strahlenther Onkol. 2012;188:1038–47. https://doi.org/10.1007/s00066-012-0223-z.

    Article  PubMed  CAS  Google Scholar 

  96. De Jong Y, Van Oosterwijk JG, Kruisselbrink AB, Briaire-de Bruijn IH, Agrogiannis G, Baranski Z, Cleven AH, Cleton-Jansen AM, Van De Water B, Danen EH, et al. Targeting Survivin as a Potential New Treatment for Chondrosarcoma of Bone. Oncogenesis 2016;5:e222 https://doi.org/10.1038/oncsis.2016.33.

  97. Brown JM, Wilson WR. Exploiting tumour hypoxia in cancer treatment. Nat Rev Cancer. 2004;4:437–47. https://doi.org/10.1038/nrc1367.

    Article  PubMed  CAS  Google Scholar 

  98. Harrison DJ, Parisi MT, Shulkin BL. The role of 18F-FDG-PET/CT in pediatric sarcoma. Semin Nucl Med. 2017;47:229–41. https://doi.org/10.1053/j.semnuclmed.2016.12.004.

    Article  PubMed  Google Scholar 

  99. Cheney MD, Chen Y-L, Lim R, Winrich BK, Grosu AL, Trofimov AV, Depauw N, Shih HA, Schwab JH, Hornicek FJ, et al. 18F-FMISO PET/CT visualization of tumor hypoxia in patients with chordoma of the mobile and sacrococcygeal spine. Int J Radiat Oncol Biol Phys. 2014;90:1030–6. https://doi.org/10.1016/j.ijrobp.2014.08.016.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Brenner W, Conrad EU, Eary JF. FDG PET imaging for grading and prediction of outcome in chondrosarcoma patients. Eur J Nucl Med Mol Imaging. 2004;31:189–95. https://doi.org/10.1007/s00259-003-1353-4.

    Article  PubMed  Google Scholar 

  101. Rajendran JG, Wilson DC, Conrad EU, Peterson LM, Bruckner JD, Rasey JS, Chin LK, Hofstrand PD, Grierson JR, Eary JF, et al. [(18)F]FMISO and [(18)F]FDG PET imaging in soft tissue sarcomas: correlation of hypoxia, metabolism and VEGF expression. Eur J Nucl Med Mol Imaging. 2003;30:695–704. https://doi.org/10.1007/s00259-002-1096-7.

    Article  PubMed  CAS  Google Scholar 

  102. Brizel DM, Scully SP, Harrelson JM, Layfield LJ, Bean JM, Prosnitz LR, Dewhirst MW. Tumor oxygenation predicts for the likelihood of distant metastases in human soft tissue sarcoma. Cancer Res. 1996;56:941–3.

    PubMed  CAS  Google Scholar 

  103. Feng H, Wang J, Chen W, Shan B, Guo Y, Xu J, Wang L, Guo P, Zhang Y. Hypoxia-Induced autophagy as an additional mechanism in human osteosarcoma radioresistance. J Bone Oncol. 2016;5:67–73. https://doi.org/10.1016/j.jbo.2016.03.001.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Dai N, Qing Y, Cun Y, Zhong Z, Li C, Zhang S, Shan J, Yang X, Dai X, Cheng Y, et al. MiR-513a-5p regulates radiosensitivity of osteosarcoma by targeting human apurinic/apyrimidinic endonuclease. Oncotarget. 2018;9:25414–26. https://doi.org/10.18632/oncotarget.11003.

    Article  PubMed  Google Scholar 

  105. Yang Z, Wa Q-D, Lu C, Pan W, Lu Z-Μ, Ao J. MiR-328-3p enhances the radiosensitivity of osteosarcoma and regulates apoptosis and cell viability via H2AX. Oncol Rep. 2018;39:545–53. https://doi.org/10.3892/or.2017.6112.

    Article  PubMed  CAS  Google Scholar 

  106. Turek M, Padilla M, Argyle DJ. Evaluation of the gene for inducible nitric oxide synthase as a radiosensitizer under hypoxic and oxic conditions. Vet Comp Oncol. 2007;5:250–5. https://doi.org/10.1111/j.1476-5829.2007.00138.x.

    Article  PubMed  CAS  Google Scholar 

  107. Shen L, Zhao K, Li H, Ning B, Wang W, Liu R, Zhang Y, Zhang A. Downregulation of UBE2T can enhance the radiosensitivity of osteosarcoma in vitro and in vivo. Epigenomics. 2019;11:1283–305. https://doi.org/10.2217/epi-2019-0125.

    Article  PubMed  CAS  Google Scholar 

  108. Liu Y, Zhu S-T, Wang X, Deng J, Li W-H, Zhang P, Liu B-S. MiR-200c regulates tumor growth and chemosensitivity to cisplatin in osteosarcoma by targeting AKT2. Sci Rep. 2017;7:13598. https://doi.org/10.1038/s41598-017-14088-3.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  109. Wang Y-H, Wang Z-X, Qiu Y, Xiong J, Chen Y-X, Miao D-S, De W. Lentivirus-mediated RNAi knockdown of insulin-like growth factor-1 receptor inhibits growth, reduces invasion, and enhances radiosensitivity in human osteosarcoma cells. Mol Cell Biochem. 2009;327:257–66. https://doi.org/10.1007/s11010-009-0064-y.

    Article  PubMed  CAS  Google Scholar 

  110. Zhang Z, Liu X, Chen D, Yu J. Radiotherapy combined with immunotherapy: the dawn of cancer treatment. Signal Transduct Target Ther. 2022;7:258. https://doi.org/10.1038/s41392-022-01102-y.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Heymann M-F, Schiavone K, Heymann D. Bone sarcomas in the immunotherapy Era. Br J Pharmacol. 2021;178:1955–72. https://doi.org/10.1111/bph.14999.

    Article  PubMed  CAS  Google Scholar 

  112. Siozopoulou V, Domen A, Zwaenepoel K, Van Beeck A, Smits E, Pauwels P, Marcq E. Immune checkpoint inhibitory therapy in sarcomas: is there light at the end of the tunnel? Cancers (Basel). 2021;13:360. https://doi.org/10.3390/cancers13020360.

    Article  PubMed  CAS  Google Scholar 

  113. Weber DC, Malyapa R, Albertini F, Bolsi A, Kliebsch U, Walser M, Pica A, Combescure C, Lomax AJ, Schneider R. Long term outcomes of patients with skull-base low-grade chondrosarcoma and chordoma patients treated with pencil beam scanning proton therapy. Radiother Oncol. 2016;120:169–74. https://doi.org/10.1016/j.radonc.2016.05.011.

    Article  PubMed  Google Scholar 

  114. Snider JW, Schneider RA, Poelma-Tap D, Stieb S, Murray FR, Placidi L, Albertini F, Lomax A, Bolsi A, Kliebsch U, et al. Long-term outcomes and prognostic factors after pencil-beam scanning proton radiation therapy for spinal chordomas: a large, single-institution cohort. Int J Radiat Oncol Biol Phys. 2018;101:226–33. https://doi.org/10.1016/j.ijrobp.2018.01.060.

    Article  PubMed  Google Scholar 

  115. Uezono H, Indelicato DJ, Rotondo RL, Mailhot Vega RB, Bradfield SM, Morris CG, Bradley JA. Treatment outcomes after proton therapy for ewing sarcoma of the pelvis. Int J Radiat Oncol Biol Phys. 2020;107:974–81. https://doi.org/10.1016/j.ijrobp.2020.04.043.

    Article  PubMed  Google Scholar 

  116. Chargari C, Levy A, Paoletti X, Soria J-C, Massard C, Weichselbaum RR, Deutsch E. Methodological development of combination drug and radiotherapy in basic and clinical research. Clin Cancer Res. 2020;26:4723–36. https://doi.org/10.1158/1078-0432.CCR-19-4155.

    Article  PubMed  CAS  Google Scholar 

  117. Herrera FG, Ronet C, Ochoa de Olza M, Barras D, Crespo I, Andreatta M, Corria-Osorio J, Spill A, Benedetti F, Genolet R, et al. Low-dose radiotherapy reverses tumor immune desertification and resistance to immunotherapy. Cancer Discov. 2022;12:108–33. https://doi.org/10.1158/2159-8290.CD-21-0003.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

The authors want to thank Brigitte Manship for editing the manuscript and correcting the English.

Funding

Non applicable.

Author information

Authors and Affiliations

Authors

Contributions

MA.L. wrote the main manuscript text and prepared figures; A.D wrote and edited the manuscript; JY.B and M.B. read and edited the manuscript. All authors reviewed the manuscript.

Corresponding author

Correspondence to Aurélie Dutour.

Ethics declarations

Ethics approval and consent to participate

Non applicable.

Consent for publication

Non applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Locquet, MA., Brahmi, M., Blay, JY. et al. Radiotherapy in bone sarcoma: the quest for better treatment option. BMC Cancer 23, 742 (2023). https://doi.org/10.1186/s12885-023-11232-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-023-11232-3

Keywords