Overview
Primary bone cancers are extremely rare neoplasms accounting for ∼0.2% of all cancers, although the true incidence is difficult to determine secondary to the rarity of these tumors.1 In 2025 an estimated 3,770 people will be diagnosed in the United States and 2,190 people will die of these diseases.2
Primary bone cancers demonstrate wide clinical heterogeneity and may be curable with proper treatment. In adults, chondrosarcoma is the most common primary bone cancer, accounting for more than 40% of newly diagnosed bone cancers, followed by osteosarcoma (∼25%), chordoma (10%), and Ewing sarcoma (8%).3 Other less common primary bone tumors include giant cell tumor of bone (GCTB) and undifferentiated pleomorphic sarcoma. In children and adolescents, osteosarcoma and Ewing sarcoma are far more common than chondrosarcoma and chordoma.3,4 High-grade undifferentiated pleomorphic sarcoma of bone and GCTB are relatively rare tumors, with each constituting<5% of primary bone tumors.5 GCTB has both benign and exceptionally rare malignant forms. Various types of bone cancers are named based on their histologic line of differentiation. Chondrosarcomas differentiate toward cartilage, osteosarcomas toward bone, angiosarcoma and epithelioid hemangioendothelioma show endothelial (vascular) differentiation, and chordoma shows notochordal differentiation. Several primary bone cancers, including Ewing sarcoma, are of unknown lineage. Chondrosarcoma is more prevalent in individuals>50 years of age.6 Osteosarcoma and Ewing sarcoma develop mainly in children and young adults. Chordoma is more common in males, with the peak incidence in the fifth to sixth decade of life.7–9
The pathogenesis and etiology of most bone cancers remain unclear. Gene rearrangements between the FET (usually EWSR1) and ETS (usually FLI1) family of genes have been implicated in the pathogenesis of Ewing sarcoma.10–14 Specific germline mutations have been linked to the development of osteosarcoma.15 Li-Fraumeni syndrome, characterized by a germline mutation in the TP53 gene, is associated with a high risk of developing cancer, including osteosarcoma.16–18 Osteosarcoma is the most common second primary malignancy in patients with a history of retinoblastoma, characterized by a germline mutation in the retinoblastoma gene RB1.19–21 Increased incidences of osteosarcoma have also been associated with other genetic mutations and inherited genetic predisposition syndromes.21 Osteosarcoma is also the most common radiation-induced bone sarcoma.22,23
The development of multiagent chemotherapy regimens for neoadjuvant and adjuvant treatment has markedly improved the prognosis for patients with osteosarcoma and Ewing sarcoma.24,25 With current multimodality treatment, approximately 60%–70% of patients diagnosed with osteosarcoma (without clinically evident metastases) are cured, and ∼90% of patients diagnosed with osteosarcoma can be treated with limb-sparing approaches.26–28 Five-year relative survival rates have improved to 82% in patients with localized Ewing sarcoma.29 In patients with Ewing sarcoma and osteosarcoma, a cure is still achievable in selected patients diagnosed with metastatic disease at presentation.30,31 The 5-year survival across all types of primary bone cancers is 68%.1
The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Bone Cancer focus on chordoma, chondrosarcoma, Ewing sarcoma, and osteosarcoma. The guidelines also provide recommendations for treating GCTB. Although typically benign, GCTB is locally aggressive and can lead to significant bone destruction. Malignant transformation is also possible.
Staging
The eighth edition of the AJCC staging classification (2018) is based on the assessment of histologic grade (G), tumor size (T), and presence of regional (N) and/or distant metastases (M).32
The NCCN Bone Cancer Panel would like to clarify that although some studies interpret imaging before chemotherapy treatment based on the extent of tumor invasion relative to the periosteum (eg, extraperiosteal, intraperiosteal) for prognostic purposes, these terms do not specifically occur in any validated staging systems and the significance is unknown.
Principles of Bone Cancer Management
Multidisciplinary Team Involvement
Primary bone tumors and selected metastatic tumors should be evaluated and treated by a multidisciplinary team of physicians with demonstrated expertise in the management of these tumors. The core team should include, at minimum, the following: orthopedic oncologist, bone pathologist, medical/pediatric oncologist, radiation oncologist, and musculoskeletal radiologist. Other specialists who may be critical in certain cases include the following: thoracic surgeon, plastic surgeon, interventional radiologist, physiatrist, vascular/general surgeon, neurosurgeon/orthopedic spine surgeon, and palliative care physician. Additional surgical subspecialties may be included as clinically indicated.
Long-term surveillance and follow-up are necessary when considering the risk of recurrence and comorbidities associated with systemic therapy and radiation therapy (RT). Life-long follow-up is recommended for surveillance and treatment of late effects of surgery, RT, and systemic therapy in long-term survivors. Patients should be given a survivorship prescription to schedule follow-up with a multidisciplinary team. Fertility issues should be discussed with appropriate patients.33 For information on disease- and survivorship-related issues for adolescent and young adult (AYA) patients, please refer to the NCCN Guidelines for Adolescent and Young Adult (AYA) Oncology (available at NCCN.org) as clinically appropriate. Finally, select patients with a family history of genetic predisposition to bone sarcomas may benefit from genetic consultation and testing.
Diagnostic Workup
Suspicion of a malignant bone tumor in a patient with a symptomatic lesion often begins when a poorly marginated lesion is seen on a plain radiograph.
In patients <40 years, an aggressive, symptomatic bone lesion has a significant risk of being a malignant primary bone tumor. Referral to an orthopedic oncologist should be considered before further workup.
In patients ≥40 years of age, workup for potential bone metastasis is recommended as indicated. This would include history and physical, bone scan, chest x-ray, and CT of the chest/abdomen/pelvis with intravenous contrast. Additional testing, such as serum protein electrophoresis, complete blood count (CBC), comprehensive metabolic panel, prostate-specific antigen test, or mammogram may also be included.34 Other imaging studies such as FDG-PET/CT (category 2B) may be performed as clinically indicated.34–36
Prior to biopsy, all patients with suspected bone sarcoma should undergo complete staging and consultation with an oncologist regarding prebiopsy imaging.
Depending on the suspected type of primary bone cancer, the standard staging workup can include history and physical, x-ray, cross-sectional imaging of the primary site (eg, contrast-enhanced MRI of the entire bone, and/or CT scan if MRI contraindicated), screening MRI of spinal axis, and CT chest/abdomen/pelvis with contrast.37 FDG-PET/CT and/or bone scan can be considered if clinically indicated. Fertility consultation should be offered to individuals of child-bearing potential if cytotoxic chemotherapy will be given.
Laboratory studies, such as CBC, comprehensive metabolic panel with calcium to assess for hypercalcemia, lactate dehydrogenase (LDH), and alkaline phosphatase (ALP) should be done before definitive treatment and periodically during treatment and surveillance. Molecular studies can be considered to delineate potential therapeutic options.
Biopsy
Percutaneous (core needle) or incisional (open) biopsy are the 2 techniques historically used in the diagnosis of musculoskeletal lesions.38,39
Open biopsy is the most accurate method because of larger sample size, which is useful for performing additional studies such as immunohistochemistry or cytogenetics.40 However, open biopsy requires general or regional anesthesia and operating room facilities; moreover, it can lead to contamination of surrounding tissues. Core biopsy can be performed under moderate sedation. Core needle biopsy is also highly diagnostic, with accuracy rates ranging from 88% to 96% when adequate samples are obtained.41–44 Core biopsy is associated with a low complication rate, and cost savings may be realized when needle biopsy is employed in selected patients.41,44,45 Advances in imaging techniques have contributed to the increasing use of image-guided percutaneous biopsy for the diagnosis of primary and secondary bone tumors.46 Furthermore, rates of complications, particularly altered treatment and outcomes, are considerably higher with open biopsy.39,47 Although no randomized controlled trials have compared core needle biopsy with open biopsy, the higher complication rate and cost of open biopsy have resulted in a shift to the use of primarily core biopsy for diagnosis.
The guidelines recommend core-needle or open biopsy to confirm the diagnosis of primary bone tumor before any surgical procedure or fixation of primary site. Biopsy should be performed at a center that will provide definitive treatment for patients with a suspected primary malignant bone tumor. At the time of biopsy, careful consideration should be given to appropriate measures to protect against impending pathologic fracture. The placement of biopsy is critical to the planning of limb-sparing surgery. A multidisciplinary approach including the radiologist and orthopedic oncologic surgeon, should be taken to follow appropriate biopsy procedures and decrease risk of adverse patient outcomes.48,49 In a multicenter review of 597 patients with musculoskeletal tumors, alteration of the treatment plan (complex resection or the use of adjunctive treatment) was encountered in 19% of patients and unnecessary amputation was performed in 18 patients.47
Both core needle and open biopsy techniques are associated with risk of local tumor recurrence, either by tumor spillage or tumor seeding along the biopsy tract, if the scar is not removed en bloc during the tumor resection. The risk of tumor seeding is less with core needle biopsy.50–52 Nevertheless, the same principles should be applied for core needle and open biopsy. Appropriate communication between the surgeon, musculoskeletal or interventional radiologist, and bone pathologist is critical in planning the biopsy route. In the case of children, consultation with a pediatric oncologist is recommended. It is essential to select the biopsy route in collaboration with the surgeon to ensure that the biopsy tract lies within the planned resection bed so that it can be resected with the same wide margins as the primary tumor during surgery. Although the risk of tumor seeding is not significant with FNA biopsy, it is not suitable for the diagnosis of primary lesions since the diagnostic accuracy of FNA is less than that of core needle biopsy.53
Surgery
Surgical margins should be negative for most sarcomas, wide enough to minimize potential local recurrence, and narrow enough to maximize function. Wide excision implies histologically negative surgical margins which is necessary to optimize local control. Local control may be achieved either by limb-sparing surgery or amputation. In selected cases, amputation may be the most appropriate option to achieve this goal. However, limb-sparing surgery is preferred if reasonable functional outcomes can be achieved. Final pathologic evaluation should include assessment of surgical margins and size/dimensions of tumor. Fresh tissue may be needed for molecular studies and tissue banking. The response to preoperative therapy should be evaluated utilizing pathologic mapping. Consultation with a physiatrist may assist with mobility training and prescribing an appropriate rehabilitation program.
Radiation Therapy
RT is used either as an adjuvant to surgery for patients with resectable tumors or as definitive therapy in patients with tumors not amenable to surgery. Specialized techniques such as intensity-modulated RT; particle beam RT with protons, carbon ions, or other heavy ions; or stereotactic radiosurgery/stereotactic RT should be considered as clinically indicated to deliver high radiation doses while maximizing normal tissue sparing.54,55 RT should be administered at the same specialized center that is providing surgical and systemic interventions. See “Principles of Radiation Therapy” in these guidelines at NCCN.org for treatment volumes and radiation doses specific to each subtype.
Ewing Sarcoma
Ewing sarcoma is characterized by the fusion of the EWS gene (EWSR1) on chromosome 22q12 with various members of the ETS gene family (FLI1, ERG, ETV1, ETV4, and FEV).11,12 The EWSR1::FLI1 fusion transcript resulting from the fusion of EWSR1 and FLI1 on chromosome 11 and the corresponding chromosomal translocation, t(11;22)(q24;q12), is identified in about 85% of patients with Ewing sarcoma.11,14 In 5% to 10% of cases, EWSR1 is fused with other members of the ETS gene family. In rare cases, FUS can substitute for EWSR1, resulting in fusion transcripts with no EWSR1 rearrangement [FUS::ERG fusion transcript resulting from the translocation t(16;21)(p11;q24) or FUS::FEV fusion transcript resulting from the translocation t(2;16)(q35;p11)].56,57 Overall, 90% of Ewing sarcomas will have 1 of 4 cytogenetic translocations. Ewing sarcoma is also characterized by the strong expression of cell surface glycoprotein MIC2 (CD99).58,59 The expression of CD99 may be useful in the differential diagnosis of Ewing sarcoma from other small round-cell neoplasms, although it is not exclusively specific to these tumors.60 Similar in morphology with various molecular signatures, other primary round cell sarcoma of bone, previously referred to as Ewing-like sarcomas, are a heterogenous group of tumors that primarily affect the pediatric and adolescent population.61 These tumors include CIC-rearranged sarcomas (eg, CIC::DUX4), BCOR-rearranged sarcomas, and round cell sarcomas with EWSR1 fusions with non-ETS genes.62
Typically, Ewing sarcoma occurs in AYAs, with an incidence of 1 case per 1.5 million.63 The most common primary sites are the pelvic bones, femur, and the bones of the chest wall, although any bone may be affected.24 However, in approximately 20% of cases, these tumors emerge from extraosseous tissue.63 When arising in a long bone, the diaphysis is the most frequently affected site.24 On imaging, the bone appears mottled. Periosteal reaction is classic, and it is referred to as “onion skin” by radiologists.
Patients with Ewing sarcoma, as with most patients with bone sarcomas, seek attention because of localized pain or swelling. Unlike other bone sarcomas, constitutional symptoms such as fever, weight loss, and fatigue are occasionally noted at presentation. Abnormal laboratory studies may include elevated serum LDH and leukocytosis.
Prognostic Factors
The important indicators of favorable prognosis include a distal/peripheral site of primary disease, tumor volume <100 mL, normal LDH level at presentation, and the absence of metastatic disease at presentation.64–71 Ewing sarcoma in the spine and sacrum is associated with significantly worse outcome and prognosis than primary Ewing sarcoma in other sites.72 In a systematic review, Bosma et al also reported tumor size (diameter >8 cm) and histologic response to initial systemic treatment (≥90% necrosis) to be important prognostic variables.73 Nevertheless, metastatic disease at presentation is the most significant adverse prognostic factor observed in those with Ewing sarcoma, as it is for other bone sarcomas.30,68,74 The lungs, bone, and bone marrow are the most common sites of metastasis. In a retrospective analysis of 975 patients from the EICESS Study Group, 5-year relapse-free survival (RFS) was 22% for patients with metastatic disease at diagnosis compared with 55% for patients without metastases at diagnosis.30 Among patients with metastases, there was a trend for better survival for those with lung metastases compared with those with bone metastases or a combination of lung and bone metastases.30,75 Metastases to uncommon sites (ie, brain, liver, spleen) were associated with a worse prognosis in a retrospective study of 30 patients.76 Poor histologic/radiologic response to chemotherapy has also been identified as an adverse prognostic factor in patients with localized nonmetastatic disease,67,71,77,78 even when chemotherapy was followed by complete (R0) resection.79
The results of the Intergroup Ewing’s Sarcoma Study (IESS) analyzing the clinicopathologic features of 303 cases of Ewing sarcoma showed that patients with primary tumors in pelvic bones have lower survival rates compared with patients with lesions in distal bones of the extremities.80 In an analysis of 53 patients (24 adult and 29 pediatric) with Ewing sarcoma treated with chemotherapy, Gupta et al identified pelvic disease and time to local therapy as significant prognostic factors associated with event-free survival (EFS) in a multivariate analysis.81 In another retrospective analysis of patients with Ewing sarcoma from a large population-based cancer registry, Lee et al determined that adult age, Hispanic ancestry, metastatic disease, large tumor size, and low socioeconomic status are poor prognostic factors for overall survival (OS).82
Workup
If Ewing sarcoma is suspected as a diagnosis, the patient should undergo complete staging before biopsy (Figure 1). This should include CT of the chest with or without contrast as clinically indicated (noncontrast CT is recommended for restaging); contrast-enhanced; MRI with or without CT of the primary site; whole body FDG-PET/CT (preferred) and/or bone scan; and possibly bone marrow biopsy and/or screening MRI with or without contrast of the spine and pelvis. In a systematic review and meta-analysis, Treglia et al reported that the combination of FDG-PET/CT with conventional imaging is a valuable tool for the staging and restaging of Ewing sarcoma, with 96% sensitivity and 92% specificity.83 Another systematic review indicated that FDG-PET without bone marrow biopsy may be considered for staging in newly diagnosed Ewing sarcoma patients, as FDG-PET demonstrated 100% sensitivity and 96% specificity based on pooled patient data from four studies.84 Results from ACRIN 6660, a multicenter prospective cohort study conducted by the American College of Radiology Imaging Network comparing whole-body MRI and conventional imaging in pediatric patients with common malignant tumors (including Ewing sarcoma), found that the noninferior accuracy for diagnosis of distant metastasis was not established for the use of whole-body MRI compared with conventional imaging.85 However, the accuracy of whole-body MRI was higher for patients with solid tumors compared with those with lymphomas (P=.006).
EW-1. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Bone Cancer, Version 2.2025.
Citation: Journal of the National Comprehensive Cancer Network 23, 4; 10.6004/jnccn.2025.0017
Cytogenetic and/or molecular studies of the biopsy specimen should be performed to evaluate t(11;22), or other less common translocations (Figure 1). Comprehensive genomic profiling has also been used to identify potentially actionable translocations in patients with sarcoma.86,87 In the case that pathologic workup of targeted PCR, fluorescence in situ hybridization, or cytogenetics is negtive, comprehensive genomic profiling or other fusion panels for Ewing sarcoma should be considered to identify translocations, including atypical translocations.
Preliminary reports suggest that EWSR1::FLI1 translocation is associated with a better prognosis than other variants.88–90 However, reports from the EURO-EWING 99 study and the Children’s Oncology Group study suggest that with currently available effective therapies, patients with Ewing sarcoma have similar outcomes, regardless of fusion subtype in contrast to previous reports.91,92 In addition to EWSR1, FUS should be considered as a fusion gene partner in the molecular diagnosis to identify the rare cases of Ewing sarcoma with FUS::ERG or FUS::FEV fusion transcripts.56,57 Because serum LDH has been shown to have prognostic value as a tumor marker, the guidelines have included this test as part of initial evaluation (Figure 1).
Fertility consultation should also be considered as appropriate. The American Society for Reproductive Medicine recommends that conversations concerning fertility be undertaken by an interdisciplinary medical team comprised of oncologists, reproductive endocrinologists and urologists, and reproductive surgeons trained in fertility preservation methods and that fertility preservation programs be affiliated with an experienced assisted reproductive technology program.93,94 For further details and recommendations, refer to the NCCN Guidelines for Adolescent and Young Adult (AYA) Oncology (available at NCCN.org).
Treatment
Chemotherapy
Multiagent chemotherapy regimens including VDC (vincristine, doxorubicin, and cyclophosphamide) alternating with ifosfamide and etoposide (IE) have been shown to be effective in patients with localized Ewing sarcoma in single- as well as multi-institution collaborative trials in the United States and Europe. Multiagent chemotherapy for at least 9 weeks is recommended prior to surgery to downstage the tumor and increase the probability of achieving a complete resection with microscopically negative margins (R0) (Figure 1). Data supports the use of adjuvant chemotherapy following surgical resection, resulting in improved RFS and OS.95–99 Surgical resection with or without RT is used for local control following chemotherapy (Figure 2).
EW-2. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Bone Cancer, Version 2.2025.
Citation: Journal of the National Comprehensive Cancer Network 23, 4; 10.6004/jnccn.2025.0017
The addition of ifosfamide, alone or in combination with etoposide to standard chemotherapy, was evaluated in patients with newly diagnosed, nonmetastatic Ewing sarcoma.97,100–104 In the Pediatric Oncology Group-Children’s Cancer Group study (INT-0091), 398 patients with nonmetastatic Ewing sarcoma were randomized to receive chemotherapy with VACD (vincristine, dactinomycin, cyclophosphamide, and doxorubicin) alone or VACD alternating with ifosfamide and etoposide (VACD-IE) for a total of 17 cycles.97 The 5-year EFS rate was significantly higher in the VACD-IE group than in the VACD alone group (69% and 54%, respectively; P=.005). The 5-year OS rate was also significantly better among patients in the VACD-IE group (72% and 61%, respectively; P=.01). VACD-IE also was associated with lower incidences of local failure (11%) compared with VACD (30%) irrespective of the type of local control therapy; 5-year cumulative incidences of local failure were 30% in the VACD arm compared with 11% in the VACD-IE arm.105
While dose escalation of alkylating agents in the VDC-IE regimen did not improve the outcome for patients with localized disease,106 chemotherapy intensification through interval compression improved outcome in patients with localized disease.107 In a randomized trial for patients <50 years with localized Ewing sarcoma (n=568), Womer et al reported that VDC-IE given on an every-2-week schedule was found to be more effective than VDC-IE given on an every-3-week schedule, with no increase in toxicity; median 5-year EFS was 73% and 65%, respectively.107
The EURO EWING 2012 was a randomized analysis conducted to compare the induction and consolidation regimens for newly diagnosed Ewing sarcoma in both the United States and Europe.108 Six hundred forty patients between the ages of 2 and 49 years were randomized to 2 treatment arms. Treatment arm A received the European regimen: VIDE (vincristine, ifosfamide, doxorubicin, and etoposide) induction therapy followed by vincristine, actinomycin D, with ifosfamide or cyclophosphamide, or busulfan and melphalan consolidation. Treatment arm B received the United States regimen: compressed VDC/IE induction followed by vincristine and cyclophosphamide (VC), or vincristine, actinomycin D, and ifosfamide, with busulfan and melphalan consolidation. The primary endpoint of the study was EFS and the secondary endpoints were OS and toxicity. EFS at 3 years was 61% and 67% for treatment arm A and B, respectively (adjusted hazard ratio [HR], 0·71 [95% credible interval, 0·55–0·92 in favor of treatment arm A). Grade 3–5 febrile neutropenia occurred in 74% of participants in treatment arm A and in 58% of participants in treatment arm B. More participants in treatment arm A required at least one platelet transfusion; however, more participants in treatment arm B required more blood transfusions. The VDC/IE regimen proved superior to the European regimen in terms of EFS and OS with similar toxicity profiles.
IESS-I and IESS-II showed that RT plus adjuvant chemotherapy with VACD was superior to VAC in patients with localized nonmetastatic disease.96 The 5-year RFS rates were 60% and 24% for VACD and VAC, respectively (P<.001). The corresponding OS rates were 65% and 28% (P<.001).
In the INT-0091 study, which included 120 patients with metastatic disease, there was no significant difference in the EFS and OS rates between the VACD-IE and VACD regimens.97 The 5-year EFS rate was 22% for both regimens and the 5-year OS rate was 34% and 35% for the VACD-IE and VACD groups, respectively. In a study of 68 patients (44 patients with locoregional disease and 24 patients with distant metastases), Kolb et al reported 4-year EFS and OS rates of 82% and 89%, respectively, for patients with locoregional disease treated with intensive chemotherapy (doxorubicin and vincristine with or without high-dose cyclophosphamide) followed by ifosfamide and etoposide.102 In patients with distant metastases, the corresponding survival rates were 12% and 18%, respectively. Miser et al also reported similar findings in patients with Ewing sarcoma with metastases at diagnosis.109
The EICESS-92 study investigated whether cyclophosphamide has efficacy similar to ifosfamide in patients with standard-risk Ewing sarcoma (small localized tumors) and whether the addition of etoposide to a regimen already containing ifosfamide improves survival in patients with high-risk disease (large tumors or metastatic disease at diagnosis).110 Patients with standard-risk disease were randomly assigned to VAID (vincristine, dactinomycin, ifosfamide, and doxorubicin; n=76) followed by either VAID or VACD (n=79).110 The 3-year EFS rates were 73% and 74%, respectively, for VACD and VAID, suggesting that cyclophosphamide has the same efficacy as ifosfamide in this group of patients. Patients with high-risk disease were randomly assigned to VAID or VAID plus etoposide (EVAID). The 3-year EFS rate was not significantly different between the 2 treatment groups (52% and 47%, respectively, for EVAID and VAID).110
As a follow-up to the EICESS-92 study, the Euro-EWING99-R1 trial evaluated cyclophosphamide as a replacement for ifosfamide as part of consolidation therapy that also included vincristine and dactinomycin (VAC vs VAI) after VIDE induction chemotherapy in 856 patients with standard-risk Ewing sarcoma. VAC was statistically not inferior to VAI but was associated with a slight increase in events (−2.8% decrease in 3-year EFS). The proportion of patients experiencing severe hematologic toxicity was slightly higher in the VAC arm, but renal tubular function impairment was more significant for patients receiving VAI.111
High-Dose Therapy Followed by Hematopoietic Cell Transplant
High-dose therapy followed by hematopoietic cell transplant (HDT/HCT) has been evaluated in patients with localized as well as metastatic disease. HDT/HCT has been associated with potential survival benefit in patients with nonmetastatic disease.112,113 However, studies that have evaluated HDT/HCT in patients with primary metastatic disease have shown conflicting results.114–120
The EURO-EWING 99 study was the first large randomized trial designed to evaluate the efficacy and safety of multiagent induction chemotherapy with 6 courses of VIDE, local treatment (surgery and/or RT), and HDT/HCT in 281 patients with Ewing sarcoma with primary disseminated disease.115 After a median follow-up of 3.8 years, the EFS and OS rates at 3 years for the entire study cohort were 27% and 34%, respectively.119 The EFS rates were 57% and 25%, respectively, for patients with complete and partial response after HDT/HCT. Patients’ age, tumor volume, and extent of metastatic spread were identified as relevant risk factors. The outcome of patients with and without HDT/HCT was not performed because of the bias introduced early in the nontransplant group (82% of patients without HDT/HCT died after a median time of 1 year).
The EURO-EWING 99 and Ewing-2008 randomized trial asked whether consolidation high-dose chemotherapy improved survival in patients with localized Ewing sarcoma.120 Two hundred forty patients at high risk were randomly assigned to receive 7 VAI courses (n=118) or one course of busulfan and melphalan (BuMel) HDT with autologous HCT (n=122), after a VIDE 6-course induction plus one VAI consolidation course. Patients were followed up for 15 years; median follow-up time was 7.8 years. Patients treated with BuMel had greater improvement in 3-year EFS (69.0% vs 56.7%) and 8-year EFS (60.7% vs 47.1%) compared with VAI-treated patients. Three treatment-related deaths occurred: two due to BuMel toxicity and one due to VAI toxicity. More patients experienced severe acute toxicities related to the BuMel versus the VAI course.
Local Control Therapy
Surgery and RT are the local control treatment modalities used for patients with localized disease, but no randomized trials have compared these approaches head-to-head (Figure 2).
In patients with localized Ewing sarcoma treated in cooperative intergroup studies, no significant effect of local control modality (surgery, RT, or surgery plus RT) was seen on OS or EFS rates.105,121 In the CESS 86 trial, although radical surgery and resection plus RT resulted in better local control rates (100% and 95%, respectively) than definitive RT (86%), no improvement was seen in RFS or OS because of higher frequency of metastases after surgery.121 In the INT-0091 study, the incidences of local failure were similar for patients treated with surgery or RT alone (25%), but surgery plus RT resulted in lower incidences of local failure (10.5%).105 The 5-year EFS rate was also not significantly different between the groups (42%, 52%, and 47% for patients treated with surgery, RT, and surgery plus RT, respectively).
Data from other retrospective analyses suggest that surgery (with or without postoperative RT) affords better local control than RT alone in patients with localized disease.122,123 The combined analysis of 1,058 patients treated in the CESS 81, CESS 86, and EICESS 92 trials showed that the rate of local failure was significantly lower after surgery (with or without postoperative RT) than after definitive RT (7.5% vs 26.3%, respectively; P=.001), whereas the local control rate with preoperative RT was comparable to that of the surgery group (5.3%).122 A retrospective analysis of sequential studies (INT-0091, INT-0154, or AEWS0031) performed by the Children’s Oncology Group also demonstrated that definitive RT was associated with a higher risk of local failure than surgery plus RT at 25.0% and 6.3%, respectively, but there was no effect on distant failure.124
Definitive RT could be an effective treatment option for patients with tumors in anatomic locations not amenable to achieve surgery with wider resection margins.125,126 In a retrospective analysis of patients with Ewing sarcoma of vertebrae treated in the CESS 81/86 and EICESS 92 studies, definitive RT resulted in a local control rate of 22.6%, which was comparable to those of other tumor sites treated with definitive RT; EFS and OS at 5 years were 47% and 58%, respectively.125 Tumor size and RT dose have been shown to be predictive of local control rates in patients with nonmetastatic Ewing sarcoma treated with chemotherapy and definitive RT.127,128 Local control therapy has also been associated with improved outcomes in patients with primary metastatic disease.129–131 In the EURO-EWING 99 trial, the 3-year EFS was significantly lower in patients with primary metastatic disease who did not receive any local control therapy compared with those treated with local therapy for primary tumor.129 Retrospective analysis of 198 patients from EURO-EWING 99 showed no improvement of 5-year EFS associated with adjuvant RT in the setting of completely resected disease of the chest wall.132
NCCN Recommendations
All patients with Ewing sarcoma should be treated with the following protocol: primary treatment followed by local control therapy and adjuvant treatment. Primary treatment consists of multiagent chemotherapy along with appropriate growth factor support for at least 9 weeks (category 1) (Figure 1). Longer duration could be considered for patients with metastatic disease based on response. VDC/IE is the preferred regimen for patients with localized disease and is a category 1 recommendation. See “Bone Cancer Systemic Therapy Agents” in the full NCCN Guidelines (available at NCCN.org) for a list of other chemotherapy regimens that are recommended for patients with localized and metastatic disease. Of note, patients with other primary round cell sarcoma of bone can be treated like Ewing sarcoma.
Disease should be restaged with imaging following primary treatment (Figure 1). Chest imaging (specifically noncontrast CT) is recommended for restaging. Additionally, primary site imaging should include contrast-enhanced MRI with or without CT and plain radiography. Head-to-toe FDG-PET/CT or bone scan can be used for restaging depending on the imaging technique that was used in the initial workup.
Patients with stable or improved disease after primary treatment should be treated with local control therapy (Figure 2). Local control options include wide excision, definitive RT with chemotherapy, or amputation in selected cases.122,125,127,129 The choice of local control therapy should be individualized and depends on tumor location, size, response to chemotherapy, patient age, anticipated morbidity, and patient preference.105
Adjuvant chemotherapy following wide excision or amputation is recommended for all patients regardless of surgical margins. (EW-2) The panel strongly recommends that the duration of chemotherapy after wide excision or amputation should be between 28 and 49 weeks depending on the type of regimen and the dosing schedule (category 1).95–97 The addition of postoperative RT to chemotherapy is recommended for patients with positive or very close surgical margins.122 Denbo et al reported that in patients with smaller tumor size (<8 cm) and negative margins, postoperative RT can be omitted without any decrement in OS.133 The 15-year estimated OS for patients who received adjuvant RT was 80% compared with 100% for those who did not. The guidelines have included adjuvant chemotherapy alone for patients treated with wide excision and negative margins.
In the setting of widely metastatic disease, palliative therapies may be considered (Figure 3). For metastatic disease that may be amenable to local therapy, local control modalities, in the form of wide excision or definitive RT with adjuvant chemotherapy, are recommended. Regardless of postoperative margin status, chemotherapy for at least 28 to 49 weeks is to be administered (category 1). RT, in addition to chemotherapy, may be considered for positive surgical margins. After adjuvant treatment, metastases may be managed according to the location. In the case of oligometastatic disease, excision or RT is recommended. Stereotactic radiosurgery/stereotactic body RT (SBRT) can be considered, especially for oligometastases.134–136 For pulmonary metastases, dependent on the response, resection and/or whole lung irradiation (WLI) may be considered.137 Based on the EORTC-SIOP phase III study published in 1988, which concluded there to be no survival benefit of WLI over adjuvant chemotherapy for patients with osteosarcoma, a systematic review of both prophylactic as well as curative WLI in patients with osteosarcoma and Ewing sarcoma was conducted.138,139 Only 2 studies compared the results of chemotherapy alone and chemotherapy and curative WLI in patients with metastatic Ewing sarcoma. In both trials, patients experienced some benefit with WLI and chemotherapy when compared with chemotherapy alone. For instance, in the EICESS-92 trial, patients who also received WLI showed a 12% improvement in 5-year OS.139,140 Ultimately, it was concluded that the decision to use WLI should be based on the patient’s risk of pulmonary metastases and any coexisting respiratory diseases.139 Progressive disease after primary treatment is best managed with RT and/or surgery to primary site followed by chemotherapy or best supportive care.
EW-3. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Bone Cancer, Version 2.2025.
Citation: Journal of the National Comprehensive Cancer Network 23, 4; 10.6004/jnccn.2025.0017
Surveillance
Surveillance of patients with Ewing sarcoma should include a physical examination, CBC and other laboratory studies (as indicated), contrast-enhanced MRI with or without CT, and plain radiographs of the primary site (Figure 2). Chest imaging (x-ray or CT) is recommended every 3 months. Head-to-toe FDG-PET/CT or bone scan can be considered as appropriate. Surveillance intervals should be increased after 2 years. Long-term surveillance should be performed annually after 5 years, and as clinically indicated (indefinitely) (category 2B).141
Relapsed or Refractory Disease
Approximately 30%–40% of patients with localized Ewing sarcoma at presentation and 60%–80% with metastatic disease at presentation experience recurrence (local and/or distant) and have a very poor prognosis.142 Patients with a longer time to first recurrence have a better chance of survival after recurrence. Late relapse (≥2 years from the time of original diagnosis), lung-only metastases, local recurrence that can be treated with radical surgery, and intensive chemotherapy are the most favorable prognostic factors, whereas early relapse (<2 years from the time of original diagnosis) with metastases in lungs and/or other sites, recurrence at local and distant sites, elevated LDH at initial diagnosis, and initial recurrence are considered adverse prognostic factors.143–146 In a retrospective analysis, site of first relapse and time to first relapse were significant prognostic factors for adult patients with localized Ewing sarcoma.147 The probability of 5-year postrelapse survival was 55% and 22%, respectively, for patients with local and distant relapse. The probability of 5-year postrelapse survival was also significantly higher for patients with late relapse than for those with early relapse.30,142,147 Overall, it is reported that close to 70% of relapses are early relapses, of which two-thirds occur at distant sites (in the lungs and/or bones). Patients who initially presented with widespread disease are more likely to relapse at distant sites, whereas those individuals who presented with localized disease are more likely to develop local relapse.148
Topoisomerase I inhibitors (topotecan and irinotecan) in combination with cyclophosphamide and temozolomide have been associated with favorable response rates in patients with relapsed or refractory bone sarcomas.149–155 In a series of 54 patients with relapsed or refractory Ewing sarcoma, cyclophosphamide and topotecan induced responses in 44% of patients (35% of patients had a complete response and 9% had a partial response).150 After a median follow-up of 23 months, 26% of patients were in continuous remission. In a retrospective analysis of patients with recurrent or progressive Ewing sarcoma, irinotecan and temozolomide resulted in an overall objective response rate of 63%. The median time to progression (TTP) for all the evaluable patients (n=20) was 8.3 months (16.2 months for the subset of patients with recurrent disease).153 Patients who were in a 2-year first remission and those with primary localized disease had better median TTP compared with those who relapsed within 2 years from diagnosis and patients with metastatic disease at diagnosis.
Combination therapy with vincristine, irinotecan, and temozolomide also appears to be active and well-tolerated in patients with relapsed or refractory Ewing sarcoma, with an overall response rate (ORR) of 68%.156 A review of 107 patients with relapsed or refractory Ewing sarcoma examined the combination of etoposide with a platinum agent (ie, cisplatin or carboplatin), suggesting that further study of etoposide and carboplatin may be warranted.157 HDT/HCT has been associated with improved long-term survival in patients with relapsed or progressive Ewing sarcoma in small, single-institution studies.158–160 The role of this approach is yet to be determined in prospective randomized studies.
The CABONE trial, a multicenter, single-arm phase II trial, evaluated the activity of cabozantinib in patients with advanced Ewing sarcoma and osteosarcoma.161 Currently approved for renal carcinoma, hepatocellular carcinoma, and medullary thyroid cancer, cabozantinib is a vascular endothelial growth factor receptor-2 (VEGFR2) tyrosine kinase inhibitor with inhibitory activity against the MET receptor. For Ewing sarcoma, the primary endpoint in this study was a 6-month objective response, while the secondary endpoints included safety, 6-month nonprogression, best overall response, 1-year and 2-year PFS and OS, and metabolic response (evaluated by FDG-PET/CT 28 days after the first dose). The primary endpoint was achieved with a 6-month objective response of 26% (95% CI, 13–42) among 39 patients with Ewing sarcoma. Additionally, the median OS was reported to be 10.2 months with a median PFS of 4.4 months. OS was noted to be 84% at 6 months, 48% at 12 months, and finally 14% at 24 months. Forty two percent (95% CI, 25–61) of patients exhibited a metabolic tumor response. Cabozantinib was well-tolerated among patients with the most common grade 3 or 4 adverse effects being hypophosphatemia, elevated aspartate aminotransferase, palmar-plantar syndrome, pneumothorax, and neutropenia.161
The efficacy of regorafenib has been evaluated in the phase II SARC024 trial in 30 patients with metastatic Ewing sarcoma whose disease progressed following at least 1 line of therapy. The 8-week progression-free rate was 63% and the median OS was 53 weeks.162
The phase III rEECur trial compared the efficacy of high-dose ifosfamide (IFOS) (n=73) against topotecan and cyclophosphamide (TC) (n=73) in patients with relapsed or refractory Ewing sarcoma.163 The median EFS was 3.7 months and 5.7 months for TC and IFOS, respectively, and the median OS was 10.4 months and 16.8 months for TC and IFOS, respectively. Greater survival for EFS and OS was also seen in individuals aged <14 years.
A retrospective study evaluated docetaxel in combination with gemcitabine in 22 children and young adults with refractory bone sarcomas.164 The median duration of response was 4.8 months and the objective response rate was 29%.
The combination of ifosfamide, carboplatin, and etoposide has also been evaluated in three Children’s Cancer Group phase I/II trials in 97 patients with relapsed or refractory bone sarcomas.165 The ORR was 51%.
A single-arm phase II study evaluated the efficacy and safety of lurbinectedin in a cohort of 28 adult patients with relapsed Ewing sarcoma.166 Treatment with lurbinectedin resulted in an ORR of 14.3% and a median duration of response of 4.2 months.
In a study evaluating 26 patients treated for local recurrence of Ewing sarcoma, surgical treatment was associated with better survival (P<.001).167 In addition to chemotherapy, surgery may be a treatment option for some patients who experience relapsed disease.
NCCN Recommendations
Treatment options for patients with relapsed or refractory disease include participation in a clinical trial and chemotherapy (with or without RT or with or without surgery) (Figure 2). See “Bone Cancer Systemic Therapy Agents” in these guidelines (available at NCCN.org) for a list of other systemic therapy regimens recommended for patients with relapsed or refractory Ewing sarcoma.
All patients with recurrent and metastatic disease should be considered for clinical trials investigating new treatment approaches.
Osteosarcoma
Osteosarcoma is the most common primary malignant bone tumor in children and young adults. With bimodal age distribution, incidences first peak among patients aged 10 to 14 years, and again in patients >65 years.168 In adults >65 years, osteosarcoma may develop as a secondary malignancy related to Paget disease of the bone.19 Osteosarcoma is broadly classified into 3 histologic subtypes (intramedullary, surface, and extraskeletal).169
High-grade intramedullary osteosarcoma is the classic or conventional form, comprising nearly 80% of osteosarcomas.169 It is a spindle cell or pleomorphic tumor that produces osteoid or immature bone. The most frequent sites include the metaphysis of the distal femur or proximal tibia, which correspond to the physes of maximum growth in normal skeletal development. Low-grade intramedullary osteosarcoma comprise <2% of all osteosarcomas; the most common sites are similar to that of conventional osteosarcoma.170
Parosteal and periosteal osteosarcomas are juxtacortical or surface variants. Parosteal osteosarcomas are low-grade lesions accounting for up to 5% of all osteosarcomas.170 The most common site is the posterior distal femur. This variant is associated with very low metastatic potential. Transformation of low-grade parosteal osteosarcoma into high-grade osteosarcoma has been documented in 24%–43% of cases.171,172 Periosteal osteosarcomas are intermediate-grade lesions, showing partial cartilaginous differentiation, and most often involve the femur, followed by the tibia.170 High-grade surface osteosarcomas are very rare, accounting for 10% of all juxtacortical osteosarcomas.173,174
Pain and swelling are the most frequent early symptoms. Pain is often intermittent in the beginning, and a thorough workup sometimes is delayed because symptoms may be confused with growing pains or confounded by musculoskeletal injury. Osteosarcoma spreads hematogenously, with the lung being the most common metastatic site, though “skip lesions” in the same bone can also occur.
For treating extraskeletal osteosarcomas, please see the NCCN Guidelines for Soft Tissue Sarcoma (available at NCCN.org).
Prognostic Factors
Tumor site and size, patient age, presence and location of metastases, histologic response to chemotherapy, and type of surgery and surgical margins are significant prognostic factors for patients with osteosarcoma of the extremities and trunk.175–183 A meta-analysis and systematic review of 18,126 participants showed that patient sex, tumor site (extremity versus nonextremity), tumor size, response to chemotherapy, and surgery (amputation versus salvage) were identified as prognostic factors in osteosarcoma.184 In patients with extremity osteosarcomas, in addition to these variables, size and location within the limb at diagnosis also had significant influence on outcome.184 All factors except age were significant in multivariate testing, with surgical remission and histologic response to chemotherapy emerging as the key prognostic factors. In a meta-analysis of data from prospective neoadjuvant chemotherapy trials in 4,838 patients with osteosarcoma, female sex was associated with increased chemotherapy-induced tumor necrosis and greater OS, and children had better outcomes than adolescents and adults.185 In a report of the combined analysis of 3 European Osteosarcoma Intergroup randomized controlled trials, Whelan et al reported that good histologic response to preoperative chemotherapy, distal location (other than proximal humerus/femur), and female gender were associated with improved survival.180 However, high body mass index in patients with osteosarcoma was associated with lower OS compared with patients with normal body mass index.186
In patients with proven primary metastatic osteosarcoma, the number of metastases at diagnosis and the completeness of surgical resection of all clinically detected tumor sites are of independent prognostic value.31 Patients with one or a few resectable pulmonary metastases have a survival rate that approaches that of patients without metastatic disease.187,188
Elevated serum ALP and LDH levels have also been identified as prognostic indicators in patients with osteosarcoma.176,178,179,189,190 In a cohort of 1,421 patients with osteosarcoma of the extremity, Bacci et al reported significantly higher serum LDH levels in patients with metastatic disease at presentation than in patients with localized disease (36.6% vs 18.8%; P<.0001).178 The 5-year disease-free survival (DFS) correlated with serum LDH levels (39.5% for patients with high LDH levels and 60% for those with normal values). In another retrospective analysis of 789 patients with osteosarcoma of the extremity, it was reported that serum ALP level was a significant prognostic factor of EFS in patients with osteosarcoma of the extremity; the 5-year EFS rate was 24% for patients with a serum ALP value of more than four times higher than the normal value and 46% for patients with high values below this limit (P<.001).179 However, in multivariate analyses, these markers did not retain their prognostic significance when compared with tumor volume, age, and histologic response to chemotherapy.176,178
Workup
Osteosarcomas present both a local control problem and a concern for distant metastasis. Initial workup should include imaging of the primary site (contrast-enhanced MRI with or without CT), chest imaging including chest CT, and head-to-toe FDG-PET/CT and/or bone scan (Figure 4). More detailed imaging (MRI with and without contrast or CT) of skeletal metastatic sites to identify abnormalities identified on primary imaging is required for suspected metastatic disease.
OSTEO-1. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Bone Cancer, Version 2.2025.
Citation: Journal of the National Comprehensive Cancer Network 23, 4; 10.6004/jnccn.2025.0017
Plain x-rays of osteosarcomas show cortical destruction and irregular reactive bone formation. Bone scan, while uniformly abnormal at the lesion, may be useful to identify additional synchronous lesions. MRI provides excellent soft tissue contrast and may be essential for operative planning. MRI is the best imaging modality to define the extent of the lesion within the bone as well as within the soft tissues, to detect “skip” metastases and to evaluate anatomic relationships with the surrounding structures. In addition, ALP and LDH are frequently elevated in patients with osteosarcoma. Serum LDH was significantly higher in patients with metastatic disease at presentation than in patients with localized disease.178
Given that osteosarcoma is most common among children and AYAs, the effect of cancer and its treatment on fertility must be discussed with patients (Figure 4). Fertility preservation methods and alternatives should be discussed with patients as appropriate. For further details and recommendations, refer to the NCCN Guidelines for Adolescent and Young Adult (AYA) Oncology (available at NCCN.org).
Finally, a number of genetic aberrations may underly osteosarcoma.191 For instance, it is reported that nearly 70% of patients with osteosarcoma may exhibit mutations in the tumor suppressor retinoblastoma gene, RB1.191 Genetic cancer syndromes that exhibit a predisposition for osteosarcoma include Li-Fraumeni syndrome, hereditary retinoblastoma, Rothmund-Thomson syndrome type 2, Bloom syndrome, Werner syndrome, RAPADILINO syndrome, and Diamond-Blackfan anemia. Thus, the panel recommends that genetic consultation and testing be considered for patients diagnosed with chondrosarcoma or osteosarcoma who possess a family or personal history of bone sarcomas (Figure 4).21
Treatment
Surgery
Surgery (limb-sparing surgery or amputation) remains an essential part of comprehensive care for patients with osteosarcoma (Figure 4, Figure 5, and Figure 6).192 Studies that have compared limb-sparing surgery and amputation in patients with high-grade, nonmetastatic osteosarcoma have not shown any significant difference in survival and local recurrence rates between these procedures.193–195 However, limb-sparing surgery is associated with better functional outcomes.196 In patients with high-grade osteosarcomas with good histologic response to neoadjuvant chemotherapy, limb-sparing surgery is considered the preferred surgical modality if wide surgical margins can be achieved (Figure 5).193,197 Amputation is generally reserved for patients with tumors in unfavorable anatomic locations not amenable to limb-sparing surgery with adequate surgical margins and for management of local relapse when limb-sparing procedures are not practical.192,197
OSTEO-2. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Bone Cancer, Version 2.2025.
Citation: Journal of the National Comprehensive Cancer Network 23, 4; 10.6004/jnccn.2025.0017
OSTEO-3. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Bone Cancer, Version 2.2025.
Citation: Journal of the National Comprehensive Cancer Network 23, 4; 10.6004/jnccn.2025.0017
Chemotherapy
The addition of adjuvant and neoadjuvant chemotherapy regimens to surgery has revolutionized outcomes in patients with localized osteosarcoma (Figure 4, Figure 5).198 Early trials used chemotherapy regimens including at least 3 or more of the following drugs: doxorubicin, cisplatin, bleomycin, cyclophosphamide or ifosfamide, dactinomycin, and high-dose methotrexate.199–204 Subsequent clinical trials have demonstrated that short, intensive chemotherapy regimens including cisplatin and doxorubicin with or without high-dose methotrexate and ifosfamide produce excellent long-term results, similar to those achieved with standard-dose multiagent chemotherapy.205–214 Cisplatin and doxorubicin and high-dose methotrexate, cisplatin, and doxorubicin (MAP) are included as category 1 recommended regimens for first-line therapy. MAP is preferred in patients <40 years with excellent performance status. In the event a patient receiving high-dose methotrexate experiences delayed elimination due to renal impairment, glucarpidase is strongly recommended.
In a randomized trial conducted by the European Osteosarcoma Group, the combination of doxorubicin and cisplatin was better tolerated compared with a multidrug regimen with no difference in survival between the groups in patients with operable, nonmetastatic osteosarcoma.206 The 3-year and 5-year OS rates were 65% and 55%, respectively, in both groups. The 5-year PFS rate was 44% in both groups. In the INT-0133 study, which compared the 3-drug regimen (cisplatin, doxorubicin, and methotrexate) with the 4-drug regimen (cisplatin, doxorubicin, methotrexate, and ifosfamide) for the treatment of patients with nonmetastatic resectable osteosarcoma, there was no difference in the 6-year EFS (63% and 64%, respectively) and OS (74% and 70%, respectively) between the two groups.212
Chemotherapy regimens without doxorubicin or cisplatin have also been evaluated in patients with localized osteosarcoma with the aim of minimizing long-term cardiotoxicity and ototoxicity.215,216 In a randomized multicenter trial (SFOP-OS94), the combination of ifosfamide and etoposide resulted in a higher histologic response rate than the regimen containing high-dose methotrexate and doxorubicin (56% and 39%, respectively). However, the 5-year OS was similar in both arms and there was no significant difference in 5-year EFS rates.216
Good histopathologic response (>90% necrosis) to neoadjuvant chemotherapy is associated with improved survival regardless of the type of chemotherapy administered after surgery.141,217,218 In an analysis of 881 patients with nonmetastatic osteosarcoma of the extremities treated with neoadjuvant chemotherapy and surgery at the Rizzoli Orthopaedic Institute, Bacci et al showed that the 5-year DFS and OS correlated significantly with histologic response to chemotherapy.219 The 5-year DFS and OS in patients with good and poor response to chemotherapy were 67.9% versus 51.3% (P<.0001) and 78.4% versus 63.7% (P<.0001), respectively. A report from the Children’s Oncology Group also confirmed these findings; the 8-year postoperative EFS and OS rates were 81% and 87%, respectively, in patients with good response.217 The corresponding survival rates were 46% and 52%, respectively, in patients with poorly responding disease. For individuals with poorly responding disease, completion of chemotherapy is still warranted, as outcomes are still better than those who do not complete chemotherapy.
NCCN Recommendations
Localized Disease
The guidelines recommend wide excision as the primary treatment of patients with low-grade (intramedullary and surface) osteosarcomas and periosteal lesions (Figure 4). If pathologic high-grade disease is discovered after wide excision, adjuvant chemotherapy is a category 1 recommendation (Figure 4). Long-term results (>25 years of follow-up) from patients with high-grade, localized osteosarcoma reveal significant benefits of adjuvant chemotherapy on DFS and OS.218 Chemotherapy prior to wide excision could be considered for patients with periosteal lesions (Figure 4). Although chemotherapy (neoadjuvant or adjuvant) has been used in the treatment of patients with periosteal osteosarcoma, there are no data to support that the addition of chemotherapy to wide excision improves outcome in patients with periosteal osteosarcoma.220–222 In a review of 119 patients with periosteal sarcoma published by the European Musculo-Skeletal Oncology Society, the use of neoadjuvant chemotherapy was not a prognostic factor, although it was used in the majority of the patients.221 Cesari et al also reported similar findings; the 10-year OS rates were 86% and 83%, respectively, for patients who received adjuvant chemotherapy with surgery and for those who underwent surgery alone (P=.73).220 A systematic review of 13 studies, including 291 patients with periosteal osteosarcoma, found no statistically significant improvement in mortality rates among those who received surgery and (neo-) adjuvant chemotherapy and those who received surgery alone.222
Neoadjuvant chemotherapy prior to wide excision is preferred for those with high-grade osteosarcoma (category 1; Figure 5).187,205–207,210–212,215,216,223 Repeat imaging using pretreatment imaging modalities should be used to reassess the tumor for resectability. Selected older patients may benefit from immediate surgery.
After wide excision, patients whose disease has a good histologic response (amount of viable tumor is <10% of the tumor area) should continue to receive several more cycles of the same chemotherapy. Surgical re-excision with or without RT can be considered in cases with positive surgical margins. In a study of 119 patients with osteosarcoma of the head and neck, combined modality treatment with surgery and RT (vs surgery alone) improved local control and OS for patients with positive or uncertain surgical margins.224 Combined photon/proton or proton beam RT has been shown to be effective for local control in some patients with unresectable or incompletely resected osteosarcoma.225,226
Patients whose disease has a poor response (viable tumor is ≥10% of the tumor area) can continue with the preoperative regimen or be considered for chemotherapy with a different regimen (category 3; Figure 5). However, attempts to improve the outcome of those with poor response by modifying the adjuvant chemotherapy remain unsuccessful.213,227–230
In individuals whose sarcoma remains unresectable after preoperative chemotherapy, RT or adjuvant chemotherapy is recommended (Figure 5). A randomized phase III trial of the European and American Osteosarcoma Study (EURAMOS) Group evaluated treatment strategies for resectable osteosarcoma based on histologic response to preoperative chemotherapy.227,231 The EURAMOS-1 trial included cohorts that received maintenance therapy with MAP; MAP with IFN-α-2b therapy; or MAP with ifosfamide and etoposide (MAPIE). The addition of maintenance IFN-α-2b therapy to MAP in the adjuvant setting did not improve outcomes for individuals whose disease had a good response to preoperative chemotherapy.231 However, the authors note that a significant portion of patients assigned to the IFN arm (n=357) did not receive the intended dose of IFN-α-2b due to patient declination to initiate therapy (n=86; 24%) or premature termination of therapy (n=105; 39%). Additionally, adding ifosfamide and etoposide to MAP (ie, MAPIE) did not improve outcomes for individuals whose disease responded poorly to preoperative chemotherapy.227 Of the 307 EFS events reported, data showed restricted mean survival time was 43.3 months (95% CI, 40.1–46.4) for individuals who received MAP (n=153) and 44.1 months (41.1–47.1) in individuals assigned to the MAPIE (n=154) cohort. No difference in EFS between cohorts was reported.
Chemotherapy should include appropriate growth factor support. See the NCCN Guidelines for Hematopoietic Growth Factors (available at NCCN.org) for growth factor support. See “Bone Cancer Systemic Therapy Agents” in these guidelines (available at NCCN.org) for a list of specific chemotherapy regimens. Additional treatment options for patients whose disease has positive margins and poor response may include additional local therapy such as re-excision with or without RT (Figure 5).
Metastatic Disease at Presentation
Approximately 10%–20% of patients present with metastatic disease at diagnosis.31,232,233 The number of metastases at diagnosis and complete surgical resection of all clinically detected tumor sites are of independent prognostic value in patients with primary metastatic disease at presentation.31 Unilateral metastases and lower number of lung nodules were associated with improved outcomes with chemotherapy in patients with synchronous lung metastases.187,188 The 2-year DFS rate was significantly higher for patients with only 1 or 2 metastatic lesions than for patients with 3 or more lesions (78% and 28%, respectively).187
Although chemotherapy is associated with improved outcomes in patients with nonmetastatic, high-grade, localized osteosarcoma, the results were significantly poorer in patients with metastatic disease at presentation.232,234–236 In a study of 57 patients with metastatic disease at presentation treated with cisplatin, doxorubicin, and a high dose of methotrexate and ifosfamide, the 2-year EFS and OS rates were 21% and 55%, respectively, compared with 75% and 94% in patients with nonmetastatic disease at presentation, treated with the same chemotherapy protocol.235 High-dose ifosfamide plus etoposide was examined in a phase II/III trial of 43 patients with newly diagnosed metastatic osteosarcoma, revealing an ORR of 59% ± 8%, but considerable toxicity.237
Among patients with primary metastases treated in cooperative osteosarcoma trials, long-term survival rates were higher for patients whose metastases were excised following chemotherapy and surgery of the primary tumor compared with those patients whose metastases could not be removed (48% and 5%, respectively).238 The combination of aggressive chemotherapy with simultaneous resection of primary and metastatic lesions has also resulted in improved outcomes in patients with osteosarcoma of the extremity with lung metastases at presentation.239
NCCN Recommendations
For patients with resectable metastases (pulmonary, visceral, or skeletal) at presentation, the guidelines recommend preoperative chemotherapy followed by wide excision of the primary tumor (Figure 6). Chemotherapy, metastasectomy, and stereotactic RT are included as options for the management of resectable metastatic disease. In the event that pulmonary metastasectomy is not feasible, ablation procedures may be considered. A retrospective review of 83 patients with high-grade osteosarcoma and widespread metastases who underwent complete resection of the primary tumor and surgical removal of all detectable metastases followed by chemotherapy analyzed tumor characteristics, treatment regimens, and their impact on survival.240 Nine (10.8%) patients had an initial complete remission, of which one patient remained 12.7 years later. Sixty-three of the remaining 74 patients died secondary to their disease, and 2 died of other causes. Actuarial EFS and OS after 5 years was 1.4 ± 1.4%, and 8.7 ± 3.3%, respectively. Data showed that complete microscopic resection of the primary tumor and macroscopic complete resection of metastases (and local treatment), in addition to good response to first-line chemotherapy, resulted in better outcomes when compared with individuals whose disease progressed during first-line treatment. Patients who presented with pleural effusion and metastasis at distant bones also had poorer outcomes.
In a study conducted by UCLA, 16 patients (who had received either prior chemotherapy or surgery) with lung metastases from high-grade sarcomas were treated with SBRT.135 In total, 25 lesions were identified and treated with a median SBRT dose of 54 Gy (range 36–54 Gy) in 3 to 4 fractions.135 Overall survival at 4 years was reported to be 78%.135 In another study, 30 patients with sarcoma with pulmonary metastases received SBRT at a median dose of 50 Gy in 4 to 5 fractions.134 Patients had received prior chemotherapy, surgery, or thoracic RT. Local control at 12 and 24 months was reported to be 94% and 86%, respectively, while OS was 76% and 43%.134 These reports suggest that SBRT may prove to be a promising alternative to surgery for oligometastatic disease. Unresectable metastatic disease should be managed with chemotherapy and/or RT followed by reassessment of the primary site for local control (Figure 6).
Surveillance
Once treatment is completed, surveillance should occur every 3 months for years 1 and 2, then every 4 months for year 3, then every 6 months for years 4 and 5, and annually thereafter, as clinically indicated. (OSTEO-4) Surveillance should include a complete physical exam and imaging of the primary site and chest as performed during initial disease workup. Head-to-toe FDG-PET/CT and/or bone scan (category 2B) may also be considered. Functional reassessment should be performed at every visit. CBC and other laboratory studies can be performed as clinically indicated.
Relapsed or Refractory Disease
Approximately 30%–40% of patients with localized disease and 80% of patients presenting with metastatic disease will relapse.241,242 The presence of solitary metastases, time to first relapse, and complete resectability of the disease at first recurrence have been reported to be the most important prognostic indicators for improved survival, whereas patients not amenable to surgery and those with a second or third recurrence have a poor prognosis.241–246 In patients with primary nonmetastatic osteosarcoma, a longer relapse-free interval to pulmonary metastases was significantly associated with better survival.245 The prognostic significance of surgical clearance among patients with second and subsequent recurrences was also confirmed in a report of survival estimates derived from large cohorts of unselected patients treated at the COSS group trials.247
The combination of etoposide with cyclophosphamide or ifosfamide has been evaluated in clinical trials.248–250 In a phase II trial of the French Society of Pediatric Oncology, high-dose ifosfamide and etoposide resulted in a response rate of 48% in patients with relapsed or refractory osteosarcoma.249 In another phase II trial, cyclophosphamide and etoposide resulted in a 19% response rate and 35% rate of stable disease in patients with relapsed high-risk osteosarcoma.248 PFS at 4 months was 42%.
In a noncomparative, double-blind, placebo-controlled, phase II trial (REGOBONE), the efficacy and safety of regorafenib, a multikinase inhibitor, was evaluated among patients with progressive metastatic osteosarcoma (who underwent 1–2 previous lines of chemotherapy and had a performance status of ECOG 0–1).251 It was found that 65% of patients in the regorafenib arm exhibited nonprogressive disease at 8 weeks compared with no patients in the placebo arm. In view of confirmed disease progression, 10 patients in the placebo arm were permitted to cross over to the regorafenib arm to receive treatment. The most commonly noted adverse effects associated with regorafenib included hypertension and hand-foot skin reaction. It was concluded that regorafenib displayed antitumor activity in progressive metastatic osteosarcoma, delaying disease progression. Similarly, in another randomized, double-blind phase II study (SARC024), the activity of regorafenib was again evaluated in patients with progressive metastatic osteosarcoma.252 The study met its primary endpoint with a median PFS of 3.6 months in the regorafenib arm versus 1.7 months in the placebo arm (CI, 0.21–0.85; P=.017; HR, 0.42). The NCCN panel has included regorafenib under second-line therapy for osteosarcoma (relapsed, refractory, or metastatic disease) with a category 1 recommendation.
Similar to its activity in patients with advanced Ewing sarcoma, cabozantinib, as aforementioned, also exhibited activity in patients with advanced osteosarcoma. In the CABONE trial, the primary endpoint for patients with osteosarcoma included 6-month objective response as well as 6-month nonprogression.161 Secondary endpoints included safety, best overall response, 1-year and 2-year PFS and OS, and metabolic response (evaluated by FDG-PET/CT 28 days after the first dose). Similar to that of Ewing sarcoma, the primary endpoints for patients with osteosarcoma were reached as 12% of patients showed an objective response and 33% were progression-free at 6 months. Seventeen percent of patients exhibited partial response and 62% of patients showed stable disease as their best overall response. Of those with stable disease, 33% of individuals displayed tumor shrinkage. Cabozantinib has thus been included in the guideline as a second-line treatment option for patients with relapsed, refractory, or metastatic osteosarcoma.
Single-agent gemcitabine and combination regimens such as docetaxel and gemcitabine; cyclophosphamide and topotecan; high-dose methotrexate with or without etoposide and ifosfamide; or ifosfamide, carboplatin, and etoposide have also been effective in the treatment of patients with relapsed or refractory bone sarcomas.152,164,165,216,253–257
Radium 223 dichloride is an alpha-emitting, bone-targeting radiopharmaceutical and was studied in a phase I trial in 18 patients with recurrent or metastatic osteosarcoma.258 A metabolic response was observed in 1 patient, while mixed responses were observed in 4 patients. A response in brain metastases was observed in 1 patient. The median OS time was 25 weeks.
Samarium-153 ethylene diamine tetramethylene phosphonate (Sm-153-EDTMP) is a beta-particle–emitting, bone-seeking radiopharmaceutical that was previously recommended as a second-line treatment option in the guidelines based on data in patients with locally recurrent or metastatic osteosarcoma or skeletal metastases.259,260 However, NCCN no longer recommends Sm-153-EDTMP in the guidelines as it is no longer available in the United States.
Targeted inhibition of a variety of molecular pathways such as mTOR, SRC family of kinases, and VEGFRs are being evaluated in clinical trials to improve outcomes in patients with relapsed or refractory osteosarcoma. In a phase II trial of the Italian Sarcoma Group (n=30), sorafenib (VEGFR inhibitor) demonstrated activity in patients with relapsed and unresectable high-grade osteosarcoma after progression of disease on standard multimodal therapy.261 The PFS at 4 months (primary endpoint) was 46%. Median PFS and OS were 4 months and 7 months, respectively. The CBR (defined as no progression at 6 months) was 29%. Partial response and stable disease were seen in 8% and 34% of patients, respectively, and were durable for≥6 months in 17% of patients. Sorafenib is a preferred second-line therapy option for osteosarcoma in the guidelines.
To extend the duration of activity, a study examined sorafenib combined with everolimus for patients with unresectable or relapsed high-grade osteosarcoma (n=38).262 Data suggested that this regimen is active in the second-line setting, but toxicity required dose reductions and/or treatment interruptions in 66% of patients. Therefore, under second-line options for patients with osteosarcoma, sorafenib in combination with everolimus is categorized under “Other Recommended Regimens” (category 2B recommendation).
The safety and efficacy of HDT/HCT in patients with locally advanced, metastatic, or relapsed osteosarcoma have also been evaluated.263,264 In the Italian Sarcoma Group study, treatment with carboplatin and etoposide was followed by stem cell rescue, combined with surgery-induced complete response in chemosensitive disease.264 Transplant-related mortality was 3.1%. The 3-year OS and DFS rates were 20% and 12%, respectively. The efficacy of this approach in patients with high-risk disease is yet to be determined in prospective randomized studies.
The optimal treatment strategy for patients with relapsed or refractory disease has yet to be defined. If relapse occurs, the patient should receive second-line chemotherapy and/or surgical resection when feasible, followed by imaging to assess treatment response (Figure 7). See “Bone Cancer Systemic Therapy Agents” in the complete guidelines (available at NCCN.org) for a complete list of second-line chemotherapy regimens. Surveillance is recommended for patients with disease that responds to second-line therapy.
OSTEO-4. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Bone Cancer, Version 2.2025.
Citation: Journal of the National Comprehensive Cancer Network 23, 4; 10.6004/jnccn.2025.0017
Patients with disease progression or relapse after second-line therapy could be treated with resection, RT265 (that may include radiopharmaceuticals, including radium-223), or best supportive care (Figure 7). Participation in a clinical trial is strongly encouraged.
References
- 1.↑
National Cancer Institute. SEER cancer statistics fact sheets: bone and joint cancer. Accessed July 10, 2024. Available at: https://seer.cancer.gov/statfacts/html/bones.html
- 2.↑
Siegel RL, Kratzer TB, Giaquinto AN, et al. Cancer statistics, 2025. CA Cancer J Clin 2025;75:10–45.
- 3.↑
MD Anderson Cancer Center. Bone cancer. Accessed January 16, 2025. Available at: https://www.mdanderson.org/cancer-types/bone-cancer.html#:∼:text=In%20the%20United%20States%2C%20more,all% 20cancers%20in%20this%20country
- 4.↑
National Cancer Institute. Primary bone cancer. Accessed January 16, 2025. Available at: https://www.cancer.gov/types/bone/bone-fact-sheet
- 5.↑
WHO Classification of Tumours Editorial Board. WHO Classification of Tumours: Soft Tissue and Bone Tumours, 5th ed. IARC Press; 2020.
- 6.↑
Limaiem F, Davis DD, Sticco KL. Chondrosarcoma. Accessed January 16, 2025. Available at: https://www.ncbi.nlm.nih.gov/books/NBK538132/
- 7.↑
McMaster ML, Goldstein AM, Bromley CM, et al. Chordoma: incidence and survival patterns in the United States, 1973–1995. Cancer Causes Control 2001;12:1–11.
- 8.↑
Walcott BP, Nahed BV, Mohyeldin A, et al. Chordoma: current concepts, management, and future directions. Lancet Oncol 2012;13:e69–76.
- 9.↑
Wedekind MF, Widemann BC, Cote G. Chordoma: Current status, problems, and future directions. Curr Probl Cancer 2021;45:100771.
- 10.↑
de Alava E, Gerald WL. Molecular biology of the Ewing’s sarcoma/ primitive neuroectodermal tumor family. J Clin Oncol 2000;18:204–213.
- 11.↑
Delattre O, Zucman J, Melot T, et al. The Ewing family of tumors–a subgroup of small-round-cell tumors defined by specific chimeric transcripts. N Engl J Med 1994;331:294–299.
- 13.↑
Burchill SA. Molecular abnormalities in Ewing’s sarcoma. Expert Rev Anticancer Ther 2008;8:1675–1687.
- 14.↑
Apfelbaum AA, Wrenn ED, Lawlor ER. The importance of fusion protein activity in Ewing sarcoma and the cell intrinsic and extrinsic factors that regulate it: a review. Front Oncol 2022;12:1044707.
- 15.↑
Mirabello L, Zhu B, Koster R, et al. Frequency of pathogenic germline variants in cancer-susceptibility genes in patients with osteosarcoma. JAMA Oncol 2020;6:724–734.
- 16.↑
Malkin D, Li FP, Strong LC, et al. Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science 1990;250:1233–1238.
- 17.↑
McIntyre JF, Smith-Sorensen B, Friend SH, et al. Germline mutations of the p53 tumor suppressor gene in children with osteosarcoma. J Clin Oncol 1994;12:925–930.
- 18.↑
Ognjanovic S, Olivier M, Bergemann TL, Hainaut P. Sarcomas in TP53 germline mutation carriers: a review of the IARC TP53 database. Cancer 2012;118:1387–1396.
- 20.↑
MacCarthy A, Bayne AM, Draper GJ, et al. Non-ocular tumours following retinoblastoma in Great Britain 1951 to 2004. Br J Ophthalmol 2009;93:1159–1162.
- 21.↑
Calvert GT, Randall RL, Jones KB, et al. At-risk populations for osteosarcoma: the syndromes and beyond. Sarcoma 2012;2012:152382.
- 22.↑
Kalra S, Grimer RJ, Spooner D, et al. Radiation-induced sarcomas of bone: factors that affect outcome. J Bone Joint Surg Br 2007;89:808–813.
- 23.↑
Mavrogenis AF, Pala E, Guerra G, Ruggieri P. Post-radiation sarcomas. Clinical outcome of 52 Patients. J Surg Oncol 2012;105:570–576.
- 24.↑
Bernstein M, Kovar H, Paulussen M, et al. Ewing’s sarcoma family of tumors: current management. Oncologist 2006;11:503–519.
- 25.↑
Subbiah V, Anderson P, Lazar AJ, et al. Ewing’s sarcoma: standard and experimental treatment options. Curr Treat Options Oncol 2009;10:126–140.
- 26.↑
Rathore R, Van Tine BA. Pathogenesis and current treatment of osteosarcoma: perspectives for future therapies. J Clin Med 2021;10:1182.
- 27.↑
Federman N, Bernthal N, Eilber FC, Tap WD. The multidisciplinary management of osteosarcoma. Curr Treat Options Oncol 2009;10:82–93.
- 28.↑
Xu M, Wang Z, Yu XC, et al. Guideline for limb-salvage treatment of osteosarcoma. Orthop Surg 2020;12:1021–1029.
- 29.↑
American Cancer Society. Early detection, diagnosis, and staging of Ewing tumors. Accessed July 18, 2024. Available at: https://www.cancer.org/content/dam/CRC/PDF/Public/8621.00.pdf
- 30.↑
Cotterill SJ, Ahrens S, Paulussen M, et al. Prognostic factors in Ewing’s tumor of bone: analysis of 975 patients from the European Intergroup Cooperative Ewing’s Sarcoma Study Group. J Clin Oncol 2000;18:3108–3114.
- 31.↑
Kager L, Zoubek A, Potschger U, et al. Primary metastatic osteosarcoma: presentation and outcome of patients treated on neoadjuvant Cooperative Osteosarcoma Study Group protocols. J Clin Oncol 2003;21:2011–2018.
- 32.↑
Amin MB, Edge SB, Greene FL, et al. AJCC Cancer Staging Manual, 8th ed. New York, NY: Springer; 2017.
- 33.↑
Lee SJ, Schover LR, Partridge AH, et al. American Society of Clinical Oncology recommendations on fertility preservation in cancer patients. J Clin Oncol 2006;24:2917–2931.
- 34.↑
Rougraff BT, Kneisl JS, Simon MA. Skeletal metastases of unknown origin. A prospective study of a diagnostic strategy. J Bone Joint Surg Am 1993;75:1276–1281.
- 35.↑
Schuetze SM. Utility of positron emission tomography in sarcomas. Curr Opin Oncol 2006;18:369–373.
- 36.↑
Volker T, Denecke T, Steffen I, et al. Positron emission tomography for staging of pediatric sarcoma patients: results of a prospective multicenter trial. J Clin Oncol 2007;25:5435–5441.
- 37.↑
Heck RK, Peabody TD, Simon MA. Staging of primary malignancies of bone. CA Cancer J Clin 2006;56:366–375.
- 38.↑
Chambers M, O’Hern K, Kerr DA. Fine-needle aspiration biopsy for the diagnosis of bone and soft tissue lesions: a systematic review and meta-analysis. J Am Soc Cytopathol 2020;9:429–441.
- 39.↑
Dirks M, Ewerbeck NK, Ballhause TM, et al. The diagnostic accuracy of 332 incisional biopsies in patients with malignant tumors in the musculoskeletal system. World J Surg Oncol 2023;21:4.
- 40.↑
Ashford RU, McCarthy SW, Scolyer RA, et al. Surgical biopsy with intra-operative frozen section. An accurate and cost-effective method for diagnosis of musculoskeletal sarcomas. J Bone Joint Surg Br 2006;88:1207–1211.
- 41.↑
Skrzynski MC, Biermann JS, Montag A, Simon MA. Diagnostic accuracy and charge-savings of outpatient core needle biopsy compared with open biopsy of musculoskeletal tumors. J Bone Joint Surg Am 1996;78:644–649.
- 42.↑
Welker JA, Henshaw RM, Jelinek J, et al. The percutaneous needle biopsy is safe and recommended in the diagnosis of musculoskeletal masses. Cancer 2000;89:2677–2686.
- 43.↑
Mitsuyoshi G, Naito N, Kawai A, et al. Accurate diagnosis of musculoskeletal lesions by core needle biopsy. J Surg Oncol 2006;94:21–27.
- 44.↑
Adams SC, Potter BK, Pitcher DJ, Temple HT. Office-based core needle biopsy of bone and soft tissue malignancies: an accurate alternative to open biopsy with infrequent complications. Clin Orthop Relat Res 2010;468:2774–2780.
- 45.↑
Rimondi E, Rossi G, Bartalena T, et al. Percutaneous CT-guided biopsy of the musculoskeletal system: results of 2027 cases. Eur J Radiol 2011;77:34–42.
- 46.↑
Gogna A, Peh WC, Munk PL. Image-guided musculoskeletal biopsy. Radiol Clin North Am 2008;46:455–473.
- 47.↑
Mankin HJ, Mankin CJ, Simon MA. The hazards of the biopsy, revisited. Members of the Musculoskeletal Tumor Society. J Bone Joint Surg Am 1996;78:656–663.
- 48.↑
Liu PT, Valadez SD, Chivers FS, et al. Anatomically based guidelines for core needle biopsy of bone tumors: implications for limb-sparing surgery. Radiographics 2007;27:189–205.
- 49.↑
Huang AJ, Kattapuram SV. Musculoskeletal neoplasms: biopsy and intervention. Radiol Clin North Am 2011;49:1287–1305.
- 50.↑
Davies NM, Livesley PJ, Cannon SR. Recurrence of an osteosarcoma in a needle biopsy track. J Bone Joint Surg Br 1993;75:977–978.
- 51.↑
Saghieh S, Masrouha KZ, Musallam KM, et al. The risk of local recurrence along the core-needle biopsy tract in patients with bone sarcomas. Iowa Orthop J 2010;30:80–83.
- 52.↑
Seeger LL. Revisiting tract seeding and compartmental anatomy for percutaneous image-guided musculoskeletal biopsies. Skeletal Radiol 2019;48:499–501.
- 53.↑
Yang YJ, Damron TA. Comparison of needle core biopsy and fine- needle aspiration for diagnostic accuracy in musculoskeletal lesions. Arch Pathol Lab Med 2004;128:759–764.
- 54.↑
Patel S, DeLaney TF. Advanced-technology radiation therapy for bone sarcomas. Cancer Control 2008;15:21–37.
- 55.↑
McGovern SL, Mahajan A. Progress in radiotherapy for pediatric sarcomas. Curr Oncol Rep 2012;14:320–326.
- 56.↑
Shing DC, McMullan DJ, Roberts P, et al. FUS/ERG gene fusions in Ewing’s tumors. Cancer Res 2003;63:4568–4576.
- 57.↑
Ng TL, O’Sullivan MJ, Pallen CJ, et al. Ewing sarcoma with novel translocation t(2;16) producing an in-frame fusion of FUS and FEV. J Mol Diagn 2007;9:459–463.
- 58.↑
Ambros IM, Ambros PF, Strehl S, et al. MIC2 is a specific marker for Ewing’s sarcoma and peripheral primitive neuroectodermal tumors. Evidence for a common histogenesis of Ewing’s sarcoma and peripheral primitive neuroectodermal tumors from MIC2 expression and specific chromosome aberration. Cancer 1991;67:1886–1893.
- 59.↑
Perlman EJ, Dickman PS, Askin FB, et al. Ewing’s sarcoma–routine diagnostic utilization of MIC2 analysis: a Pediatric Oncology Group/Children’s Cancer Group Intergroup Study. Hum Pathol 1994;25:304–307.
- 60.↑
Olsen SH, Thomas DG, Lucas DR. Cluster analysis of immunohistochemical profiles in synovial sarcoma, malignant peripheral nerve sheath tumor, and Ewing sarcoma. Mod Pathol 2006;19:659–668.
- 61.↑
Machado I, Navarro S, Llombart-Bosch A. Ewing sarcoma and the new emerging Ewing-like sarcomas: (CIC and BCOR-rearranged-sarcomas). A systematic review. Histol Histopathol 2016;31:1169–1181.
- 62.↑
Sbaraglia M, Righi A, Gambarotti M, Dei Tos AP. Ewing sarcoma and Ewing-like tumors. Virchows Arch 2020;476:109–119.
- 64.↑
Glaubiger DL, Makuch R, Schwarz J, et al. Determination of prognostic factors and their influence on therapeutic results in patients with Ewing’s sarcoma. Cancer 1980;45:2213–2219.
- 65.↑
Gobel V, Jurgens H, Etspuler G, et al. Prognostic significance of tumor volume in localized Ewing’s sarcoma of bone in children and adolescents. J Cancer Res Clin Oncol 1987;113:187–191.
- 66.↑
Ahrens S, Hoffmann C, Jabar S, et al. Evaluation of prognostic factors in a tumor volume-adapted treatment strategy for localized Ewing sarcoma of bone: the CESS 86 experience. Cooperative Ewing Sarcoma Study. Med Pediatr Oncol 1999;32:186–195.
- 67.↑
Bacci G, Longhi A, Ferrari S, et al. Prognostic factors in non-metastatic Ewing’s sarcoma tumor of bone: an analysis of 579 patients treated at a single institution with adjuvant or neoadjuvant chemotherapy between 1972 and 1998. Acta Oncol 2006;45:469–475.
- 68.↑
Rodríguez-Galindo C, Liu T, Krasin MJ, et al. Analysis of prognostic factors in ewing sarcoma family of tumors: review of St. Jude Children’s Research Hospital studies. Cancer 2007;110:375–384.
- 69.↑
Serlo J, Helenius I, Vettenranta K, et al. Surgically treated patients with axial and peripheral Ewing’s sarcoma family of tumours: a population based study in Finland during 1990–2009. Eur J Surg Oncol 2015;41:893–898.
- 70.↑
Grevener K, Haveman LM, Ranft A, et al. Management and outcome of Ewing sarcoma of the head and neck. Pediatr Blood Cancer 2016;63:604–610.
- 71.↑
American Cancer Society. Survival rates for Ewing tumors. Accessed September 12, 2024. Available at: https://www.cancer.org/cancer/types/ewing-tumor/detection-diagnosis-staging/survival-rates.html
- 72.↑
Bacci G, Boriani S, Balladelli A, et al. Treatment of nonmetastatic Ewing’s sarcoma family tumors of the spine and sacrum: the experience from a single institution. Eur Spine J 2009;18:1091–1095.
- 73.↑
Bosma SE, Ayu O, Fiocco M, et al. Prognostic factors for survival in Ewing sarcoma: a systematic review. Surg Oncol 2018;27:603–610.
- 74.↑
Cangir A, Vietti TJ, Gehan EA, et al. Ewing’s sarcoma metastatic at diagnosis. Results and comparisons of two intergroup Ewing’s sarcoma studies. Cancer 1990;66:887–893.
- 75.↑
Dang A, Feng X, Hamm J, et al. Survival outcomes in metastatic Ewing sarcoma treated with whole-lung radiation. Cureus 2022;14:e26750.
- 76.↑
Paulino AC, Mai WY, Teh BS. Radiotherapy in metastatic ewing sarcoma. Am J Clin Oncol 2013;36:283–286.
- 77.↑
Oberlin O, Deley MC, Bui BN, et al. Prognostic factors in localized Ewing’s tumours and peripheral neuroectodermal tumours: the third study of the French Society of Paediatric Oncology (EW88 study). Br J Cancer 2001;85:1646–1654.
- 78.↑
Paulussen M, Ahrens S, Dunst J, et al. Localized Ewing tumor of bone: final results of the cooperative Ewing’s Sarcoma Study CESS 86. J Clin Oncol 2001;19:1818–1829.
- 79.↑
Pan HY, Morani A, Wang WL, et al. Prognostic factors and patterns of relapse in ewing sarcoma patients treated with chemotherapy and r0 resection. Int J Radiat Oncol Biol Phys 2015;92:349–357.
- 80.↑
Kissane JM, Askin FB, Foulkes M, et al. Ewing’s sarcoma of bone: clinicopathologic aspects of 303 cases from the Intergroup Ewing’s Sarcoma Study. Hum Pathol 1983;14:773–779.
- 81.↑
Gupta AA, Pappo A, Saunders N, et al. Clinical outcome of children and adults with localized Ewing sarcoma: impact of chemotherapy dose and timing of local therapy. Cancer 2010;116:3189–3194.
- 82.↑
Lee J, Hoang BH, Ziogas A, Zell JA. Analysis of prognostic factors in Ewing sarcoma using a population-based cancer registry. Cancer 2010;116:1964–1973.
- 83.↑
Treglia G, Salsano M, Stefanelli A, et al. Diagnostic accuracy of (1)(8)F-FDG-PET and PET/CT in patients with Ewing sarcoma family tumours: a systematic review and a meta-analysis. Skeletal Radiol 2012;41:249–256.
- 84.↑
Campbell KM, Shulman DS, Grier HE, DuBois SG. Role of bone marrow biopsy for staging new patients with Ewing sarcoma: a systematic review. Pediatr Blood Cancer 2021;68:e28807.
- 85.↑
Siegel MJ, Acharyya S, Hoffer FA, et al. Whole-body MR imaging for staging of malignant tumors in pediatric patients: results of the American College of Radiology Imaging Network 6660 Trial. Radiology 2013;266:599–609.
- 86.↑
Boddu S, Walko CM, Bienasz S, et al. Clinical utility of genomic profiling in the treatment of advanced sarcomas: a single-center experience. JCO Precis Oncol 2018;2:1–8.
- 87.↑
Laetsch TW, Roy A, Xu L, et al. Undifferentiated sarcomas in children harbor clinically relevant oncogenic fusions and gene copy-number alterations: a report from the Children’s Oncology Group. Clin Cancer Res 2018;24:3888–3897.
- 88.↑
Avigad S, Cohen IJ, Zilberstein J, et al. The predictive potential of molecular detection in the nonmetastatic Ewing family of tumors. Cancer 2004;100:1053–1058.
- 89.↑
de Alava E, Kawai A, Healey JH, et al. EWS-FLI1 fusion transcript structure is an independent determinant of prognosis in Ewing’s sarcoma. J Clin Oncol 1998;16:1248–1255.
- 90.↑
Zoubek A, Dockhorn-Dworniczak B, Delattre O, et al. Does expression of different EWS chimeric transcripts define clinically distinct risk groups of Ewing tumor patients? J Clin Oncol 1996;14:1245–1251.
- 91.↑
Le Deley MC, Delattre O, Schaefer KL, et al. Impact of EWS-ETS fusion type on disease progression in Ewing’s sarcoma/peripheral primitive neuroectodermal tumor: prospective results from the cooperative Euro-E.W.I.N.G. 99 trial. J Clin Oncol 2010;28:1982–1988.
- 92.↑
van Doorninck JA, Ji L, Schaub B, et al. Current treatment protocols have eliminated the prognostic advantage of type 1 fusions in Ewing sarcoma: a report from the Children’s Oncology Group. J Clin Oncol 2010;28:1989–1994.
- 93.↑
Lambertini M, Del Mastro L, Pescio MC, et al. Cancer and fertility preservation: international recommendations from an expert meeting. BMC Med 2016;14:1.
- 94.↑
Practice Committee of the American Society for Reproductive Medicine. Electronic address aao. Fertility preservation in patients undergoing gonadotoxic therapy or gonadectomy: a committee opinion. Fertil Steril 2019;112:1022–1033.
- 95.↑
Burgert EO, Nesbit ME, Garnsey LA, et al. Multimodal therapy for the management of nonpelvic, localized Ewing’s sarcoma of bone: intergroup study IESS-II. J Clin Oncol 1990;8:1514–1524.
- 96.↑
Nesbit ME, Gehan EA, Burgert EO, et al. Multimodal therapy for the management of primary, nonmetastatic Ewing’s sarcoma of bone: a long-term follow-up of the First Intergroup study. J Clin Oncol 1990;8:1664–1674.
- 97.↑
Grier HE, Krailo MD, Tarbell NJ, et al. Addition of ifosfamide and etoposide to standard chemotherapy for Ewing’s sarcoma and primitive neuroectodermal tumor of bone. N Engl J Med 2003;348:694–701.
- 98.↑
Shamberger RC, LaQuaglia MP, Gebhardt MC, et al. Ewing sarcoma/primitive neuroectodermal tumor of the chest wall: impact of initial versus delayed resection on tumor margins, survival, and use of radiation therapy. Ann Surg 2003;238:563–567.
- 99.↑
Krasin MJ, Davidoff AM, Rodriguez-Galindo C, et al. Definitive surgery and multiagent systemic therapy for patients with localized Ewing sarcoma family of tumors: local outcome and prognostic factors. Cancer 2005;104:367–373.
- 100.↑
Wexler LH, DeLaney TF, Tsokos M, et al. Ifosfamide and etoposide plus vincristine, doxorubicin, and cyclophosphamide for newly diagnosed Ewing’s sarcoma family of tumors. Cancer 1996;78:901–911.
- 101.↑
Rosito P, Mancini AF, Rondelli R, et al. Italian Cooperative Study for the treatment of children and young adults with localized Ewing sarcoma of bone: a preliminary report of 6 years of experience. Cancer 1999;86:421–428.
- 102.↑
Kolb EA, Kushner BH, Gorlick R, et al. Long-term event-free survival after intensive chemotherapy for Ewing’s family of tumors in children and young adults. J Clin Oncol 2003;21:3423–3430.
- 103.↑
Oberlin O, Habrand JL, Zucker JM, et al. No benefit of ifosfamide in Ewing’s sarcoma: a nonrandomized study of the French Society of Pediatric Oncology. J Clin Oncol 1992;10:1407–1412.
- 104.↑
Bacci G, Picci P, Ferrari S, et al. Neoadjuvant chemotherapy for Ewing’s sarcoma of bone: no benefit observed after adding ifosfamide and etoposide to vincristine, actinomycin, cyclophosphamide, and doxorubicin in the maintenance phase–results of two sequential studies. Cancer 1998;82:1174–1183.
- 105.↑
Yock TI, Krailo M, Fryer CJ, et al. Local control in pelvic Ewing sarcoma: analysis from INT-0091–a report from the Children’s Oncology Group. J Clin Oncol 2006;24:3838–3843.
- 106.↑
Granowetter L, Womer R, Devidas M, et al. Dose-intensified compared with standard chemotherapy for nonmetastatic Ewing sarcoma family of tumors: a Children’s Oncology Group Study. J Clin Oncol 2009;27:2536–2541.
- 107.↑
Womer RB, West DC, Krailo MD, et al. Randomized controlled trial of interval-compressed chemotherapy for the treatment of localized Ewing sarcoma: a report from the Children’s Oncology Group. J Clin Oncol 2012;30:4148–4154.
- 108.↑
Brennan B, Kirton L, Marec-Berard P, et al. Comparison of two chemotherapy regimens in patients with newly diagnosed Ewing sarcoma (EE2012): an open-label, randomised, phase 3 trial. Lancet 2022;400:1513–1521.
- 109.↑
Miser JS, Krailo MD, Tarbell NJ, et al. Treatment of metastatic Ewing’s sarcoma or primitive neuroectodermal tumor of bone: evaluation of combination ifosfamide and etoposide–a Children’s Cancer Group and Pediatric Oncology Group study. J Clin Oncol 2004;22:2873–2876.
- 110.↑
Paulussen M, Craft AW, Lewis I, et al. Results of the EICESS-92 Study: two randomized trials of Ewing’s sarcoma treatment–cyclophosphamide compared with ifosfamide in standard-risk patients and assessment of benefit of etoposide added to standard treatment in high-risk patients. J Clin Oncol 2008;26:4385–4393.
- 111.↑
Le Deley MC, Paulussen M, Lewis I, et al. Cyclophosphamide compared with ifosfamide in consolidation treatment of standard-risk Ewing sarcoma: results of the randomized noninferiority Euro-EWING99-R1 trial. J Clin Oncol 2014;32:2440–2448.
- 112.↑
Ferrari S, Sundby Hall K, Luksch R, et al. Nonmetastatic Ewing family tumors: high-dose chemotherapy with stem cell rescue in poor responder patients. Results of the Italian Sarcoma Group/Scandinavian Sarcoma Group III protocol. Ann Oncol 2011;22:1221–1227.
- 113.↑
Gaspar N, Rey A, Berard PM, et al. Risk adapted chemotherapy for localised Ewing’s sarcoma of bone: the French EW93 study. Eur J Cancer 2012;48:1376–1385.
- 114.↑
Kushner BH, Meyers PA. How effective is dose-intensive/myeloablative therapy against Ewing’s sarcoma/primitive neuroectodermal tumor metastatic to bone or bone marrow? The Memorial Sloan-Kettering experience and a literature review. J Clin Oncol 2001;19:870–880.
- 115.↑
Juergens C, Weston C, Lewis I, et al. Safety assessment of intensive induction with vincristine, ifosfamide, doxorubicin, and etoposide (VIDE) in the treatment of Ewing tumors in the EURO-E.W.I.N.G. 99 clinical trial. Pediatr Blood Cancer 2006;47:22–29.
- 116.↑
Oberlin O, Rey A, Desfachelles AS, et al. Impact of high-dose busulfan plus melphalan as consolidation in metastatic Ewing tumors: a study by the Societe Francaise des Cancers de l’Enfant. J Clin Oncol 2006;24:3997–4002.
- 117.↑
Rosenthal J, Bolotin E, Shakhnovits M, et al. High-dose therapy with hematopoietic stem cell rescue in patients with poor prognosis Ewing family tumors. Bone Marrow Transplant 2008;42:311–318.
- 118.↑
Burdach S, Thiel U, Schoniger M, et al. Total body MRI-governed involved compartment irradiation combined with high-dose chemotherapy and stem cell rescue improves long-term survival in Ewing tumor patients with multiple primary bone metastases. Bone Marrow Transplant 2010;45:483–489.
- 119.↑
Ladenstein R, Potschger U, Le Deley MC, et al. Primary disseminated multifocal Ewing sarcoma: results of the Euro-EWING 99 trial. J Clin Oncol 2010;28:3284–3291.
- 120.↑
Whelan J, Le Deley MC, Dirksen U, et al. High-dose chemotherapy and blood autologous stem-cell rescue compared with standard chemotherapy in localized high-risk Ewing sarcoma: results of Euro-E.W.I.N.G.99 and Ewing-2008. J Clin Oncol 2018;36:JCO2018782516.
- 121.↑
Dunst J, Jurgens H, Sauer R, et al. Radiation therapy in Ewing’s sarcoma: an update of the CESS 86 trial. Int J Radiat Oncol Biol Phys 1995;32:919–930.
- 122.↑
Schuck A, Ahrens S, Paulussen M, et al. Local therapy in localized Ewing tumors: results of 1058 patients treated in the CESS 81, CESS 86, and EICESS 92 trials. Int J Radiat Oncol Biol Phys 2003;55:168–177.
- 123.↑
DuBois SG, Krailo MD, Gebhardt MC, et al. Comparative evaluation of local control strategies in localized Ewing sarcoma of bone: a report from the Children’s Oncology Group. Cancer 2015;121:467–475.
- 124.↑
Ahmed SK, Witten BG, Harmsen WS, et al. Analysis of local control outcomes and clinical prognostic factors in localized pelvic Ewing sarcoma patients treated with radiation therapy: a report from the Children’s Oncology Group. Int J Radiat Oncol Biol Phys 2023;115:337–346.
- 125.↑
Schuck A, Ahrens S, von Schorlemer I, et al. Radiotherapy in Ewing tumors of the vertebrae: treatment results and local relapse analysis of the CESS 81/86 and EICESS 92 trials. Int J Radiat Oncol Biol Phys 2005;63:1562–1567.
- 126.↑
Indelicato DJ, Keole SR, Shahlaee AH, et al. Definitive radiotherapy for ewing tumors of extremities and pelvis: long-term disease control, limb function, and treatment toxicity. Int J Radiat Oncol Biol Phys 2008;72:871–877.
- 127.↑
Krasin MJ, Rodriguez-Galindo C, Billups CA, et al. Definitive irradiation in multidisciplinary management of localized Ewing sarcoma family of tumors in pediatric patients: outcome and prognostic factors. Int J Radiat Oncol Biol Phys 2004;60:830–838.
- 128.↑
Paulino AC, Nguyen TX, Mai WY, et al. Dose response and local control using radiotherapy in non-metastatic Ewing sarcoma. Pediatr Blood Cancer 2007;49:145–148.
- 129.↑
Haeusler J, Ranft A, Boelling T, et al. The value of local treatment in patients with primary, disseminated, multifocal Ewing sarcoma (PDMES). Cancer 2010;116:443–450.
- 130.↑
Paulussen M, Ahrens S, Burdach S, et al. Primary metastatic (stage IV) Ewing tumor: survival analysis of 171 patients from the EICESS studies. European Intergroup Cooperative Ewing Sarcoma Studies. Ann Oncol 1998;9:275–281.
- 131.↑
Casey DL, Wexler LH, Meyers PA, et al. Radiation for bone metastases in Ewing sarcoma and rhabdomyosarcoma. Pediatr Blood Cancer 2015;62:445–449.
- 132.↑
Bedetti B, Wiebe K, Ranft A, et al. Local control in Ewing sarcoma of the chest wall: results of the EURO-EWING 99 trial. Ann Surg Oncol 2015;22:2853–2859.
- 133.↑
Denbo JW, Shannon Orr W, Wu Y, et al. Timing of surgery and the role of adjuvant radiotherapy in ewing sarcoma of the chest wall: a single- institution experience. Ann Surg Oncol 2012;19:3809–3815.
- 134.↑
Baumann BC, Nagda SN, Kolker JD, et al. Efficacy and safety of stereotactic body radiation therapy for the treatment of pulmonary metastases from sarcoma: a potential alternative to resection. J Surg Oncol 2016;114:65–69.
- 135.↑
Mehta N, Selch M, Wang PC, et al. Safety and efficacy of stereotactic body radiation therapy in the treatment of pulmonary metastases from high grade sarcoma. Sarcoma 2013;2013:360214.
- 136.↑
Brown LC, Lester RA, Grams MP, et al. Stereotactic body radiotherapy for metastatic and recurrent ewing sarcoma and osteosarcoma. Sarcoma 2014;2014:418270.
- 137.↑
Tanguturi SK, George S, Marcus KJ, et al. Whole lung irradiation in adults with metastatic Ewing sarcoma: practice patterns and implications for treatment. Sarcoma 2015;2015:591698.
- 138.↑
Burgers JM, van Glabbeke M, Busson A, et al. Osteosarcoma of the limbs. Report of the EORTC-SIOP 03 trial 20781 investigating the value of adjuvant treatment with chemotherapy and/or prophylactic lung irradiation. Cancer 1988;61:1024–1031.
- 139.↑
Ronchi L, Buwenge M, Cortesi A, et al. Whole lung irradiation in patients with osteosarcoma and Ewing sarcoma. Anticancer Res 2018;38:4977–4985.
- 140.↑
Bolling T, Schuck A, Paulussen M, et al. Whole lung irradiation in patients with exclusively pulmonary metastases of Ewing tumors. Toxicity analysis and treatment results of the EICESS-92 trial. Strahlenther Onkol 2008;184:193–197.
- 141.↑
Bacci G, Forni C, Longhi A, et al. Long-term outcome for patients with non-metastatic Ewing’s sarcoma treated with adjuvant and neoadjuvant chemotherapies. 402 patients treated at Rizzoli between 1972 and 1992. Eur J Cancer 2004;40:73–83.
- 142.↑
Stahl M, Ranft A, Paulussen M, et al. Risk of recurrence and survival after relapse in patients with Ewing sarcoma. Pediatr Blood Cancer 2011;57:549–553.
- 143.↑
Rodriguez-Galindo C, Billups CA, Kun LE, et al. Survival after recurrence of Ewing tumors: the St Jude Children’s Research Hospital experience, 1979-1999. Cancer 2002;94:561–569.
- 144.↑
Bacci G, Ferrari S, Longhi A, et al. Therapy and survival after recurrence of Ewing’s tumors: the Rizzoli experience in 195 patients treated with adjuvant and neoadjuvant chemotherapy from 1979 to 1997. Ann Oncol 2003;14:1654–1659.
- 145.↑
Leavey PJ, Mascarenhas L, Marina N, et al. Prognostic factors for patients with Ewing sarcoma (EWS) at first recurrence following multi-modality therapy: a report from the Children’s Oncology Group. Pediatr Blood Cancer 2008;51:334–338.
- 146.↑
Ferrari S, Luksch R, Hall KS, et al. Post-relapse survival in patients with Ewing sarcoma. Pediatr Blood Cancer 2015;62:994–999.
- 147.↑
Robinson SI, Ahmed SK, Okuno SH, et al. Clinical outcomes of adult patients with relapsed Ewing sarcoma: a 30-year single-institution experience. Am J Clin Oncol 2014;37:585–591.
- 148.↑
Van Mater D, Wagner L. Management of recurrent Ewing sarcoma: challenges and approaches. Onco Targets Ther 2019;12:2279–2288.
- 149.↑
Bernstein ML, Devidas M, Lafreniere D, et al. Intensive therapy with growth factor support for patients with Ewing tumor metastatic at diagnosis: Pediatric Oncology Group/Children’s Cancer Group phase II study 9457–a report from the Children’s Oncology Group. J Clin Oncol 2006;24:152–159.
- 150.↑
Hunold A, Weddeling N, Paulussen M, et al. Topotecan and cyclophosphamide in patients with refractory or relapsed Ewing tumors. Pediatr Blood Cancer 2006;47:795–800.
- 151.↑
Kushner BH, Kramer K, Meyers PA, et al. Pilot study of topotecan and high-dose cyclophosphamide for resistant pediatric solid tumors. Med Pediatr Oncol 2000;35:468–474.
- 152.↑
Saylors RL, Stine KC, Sullivan J, et al. Cyclophosphamide plus topotecan in children with recurrent or refractory solid tumors: a Pediatric Oncology Group phase II study. J Clin Oncol 2001;19:3463–3469.
- 153.↑
Casey DA, Wexler LH, Merchant MS, et al. Irinotecan and temozolomide for Ewing sarcoma: the Memorial Sloan-Kettering experience. Pediatr Blood Cancer 2009;53:1029–1034.
- 154.↑
Wagner LM, Crews KR, Iacono LC, et al. Phase I trial of temozolomide and protracted irinotecan in pediatric patients with refractory solid tumors. Clin Cancer Res 2004;10:840–848.
- 155.↑
Wagner LM, McAllister N, Goldsby RE, et al. Temozolomide and intravenous irinotecan for treatment of advanced Ewing sarcoma. Pediatr Blood Cancer 2007;48:132–139.
- 156.↑
Raciborska A, Bilska K, Drabko K, et al. Vincristine, irinotecan, and temozolomide in patients with relapsed and refractory Ewing sarcoma. Pediatr Blood Cancer 2013;60:1621–1625.
- 157.↑
van Maldegem AM, Benson C, Rutkowski P, et al. Etoposide and carbo-or cisplatin combination therapy in refractory or relapsed Ewing sarcoma: a large retrospective study. Pediatr Blood Cancer 2015;62:40–44.
- 158.↑
Burdach S, Jurgens H, Peters C, et al. Myeloablative radiochemotherapy and hematopoietic stem-cell rescue in poor-prognosis Ewing’s sarcoma. J Clin Oncol 1993;11:1482–1488.
- 159.↑
Barker LM, Pendergrass TW, Sanders JE, Hawkins DS. Survival after recurrence of Ewing’s sarcoma family of tumors. J Clin Oncol 2005;23:4354–4362.
- 160.↑
McTiernan A, Driver D, Michelagnoli MP, et al. High dose chemotherapy with bone marrow or peripheral stem cell rescue is an effective treatment option for patients with relapsed or progressive Ewing’s sarcoma family of tumours. Ann Oncol 2006;17:1301–1305.
- 161.↑
Italiano A, Mir O, Mathoulin-Pelissier S, et al. Cabozantinib in patients with advanced Ewing sarcoma or osteosarcoma (CABONE): a multicentre, single-arm, phase 2 trial. Lancet Oncol 2020;21:446–455.
- 162.↑
Attia S, Bolejack V, Ganjoo KN, et al. A phase II trial of regorafenib in patients with advanced Ewing sarcoma and related tumors of soft tissue and bone: SARC024 trial results. Cancer Med 2023;12:1532–1539.
- 163.↑
McCabe M, Kirton L, Khan M, et al. Phase III assessment of topotecan and cyclophosphamide and high-dose ifosfamide in rEECur: an international randomized controlled trial of chemotherapy for the treatment of recurrent and primary refractory Ewing sarcoma (RR-ES). J Clin Oncol 2022;40(Suppl):Abstract LBA2.
- 164.↑
Navid F, Willert JR, McCarville MB, et al. Combination of gemcitabine and docetaxel in the treatment of children and young adults with refractory bone sarcoma. Cancer 2008;113:419–425.
- 165.↑
Van Winkle P, Angiolillo A, Krailo M, et al. Ifosfamide, carboplatin, and etoposide (ICE) reinduction chemotherapy in a large cohort of children and adolescents with recurrent/refractory sarcoma: the Children’s Cancer Group (CCG) experience. Pediatr Blood Cancer 2005;44:338–347.
- 166.↑
Subbiah V, Brana I, Longhi A, et al. Antitumor activity of lurbinectedin, a selective inhibitor of oncogene transcription, in patients with relapsed Ewing sarcoma: results of a basket phase II study. Clin Cancer Res 2022;28:2762–2770.
- 167.↑
Xue R, Lewis VO, Moon BS, Lin PP. Local recurrence of Ewing sarcoma: is wide excision an acceptable treatment? J Surg Oncol 2019;120: 746–752.
- 168.↑
Kim C, Davis LE, Albert CM, et al. Osteosarcoma in pediatric and adult populations: are adults just big kids? Cancers (Basel) 2023;15:5044.
- 169.↑
Klein MJ, Siegal GP. Osteosarcoma: anatomic and histologic variants. Am J Clin Pathol 2006;125:555–581.
- 170.↑
Antonescu CR, Huvos AG. Low-grade osteogenic sarcoma arising in medullary and surface osseous locations. Am J Clin Pathol 2000;114(Suppl):S90–103.
- 171.↑
Sheth DS, Yasko AW, Raymond AK, et al. Conventional and dedifferentiated parosteal osteosarcoma. Diagnosis, treatment, and outcome. Cancer 1996;78:2136–2145.
- 172.↑
Bertoni F, Bacchini P, Staals EL, Davidovitz P. Dedifferentiated parosteal osteosarcoma: the experience of the Rizzoli Institute. Cancer 2005;103:2373–2382.
- 173.↑
Okada K, Unni KK, Swee RG, Sim FH. High grade surface osteosarcoma: a clinicopathologic study of 46 cases. Cancer 1999;85:1044–1054.
- 174.↑
Staals EL, Bacchini P, Bertoni F. High-grade surface osteosarcoma: a review of 25 cases from the Rizzoli Institute. Cancer 2008;112:1592–1599.
- 175.↑
Davis AM, Bell RS, Goodwin PJ. Prognostic factors in osteosarcoma: a critical review. J Clin Oncol 1994;12:423–431.
- 176.↑
Ferrari S, Bertoni F, Mercuri M, et al. Predictive factors of disease-free survival for non-metastatic osteosarcoma of the extremity: an analysis of 300 patients treated at the Rizzoli Institute. Ann Oncol 2001;12:1145–1150.
- 177.↑
Bielack SS, Kempf-Bielack B, Delling G, et al. Prognostic factors in high-grade osteosarcoma of the extremities or trunk: an analysis of 1,702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols. J Clin Oncol 2002;20:776–790.
- 178.↑
Bacci G, Longhi A, Ferrari S, et al. Prognostic significance of serum lactate dehydrogenase in osteosarcoma of the extremity: experience at Rizzoli on 1421 patients treated over the last 30 years. Tumori 2004;90:478–484.
- 179.↑
Bacci G, Longhi A, Versari M, et al. Prognostic factors for osteosarcoma of the extremity treated with neoadjuvant chemotherapy: 15-year experience in 789 patients treated at a single institution. Cancer 2006;106:1154–1161.
- 180.↑
Whelan JS, Jinks RC, McTiernan A, et al. Survival from high-grade localised extremity osteosarcoma: combined results and prognostic factors from three European Osteosarcoma Intergroup randomised controlled trials. Ann Oncol 2012;23:1607–1616.
- 181.↑
Ogura K, Fujiwara T, Yasunaga H, et al. Development and external validation of nomograms predicting distant metastases and overall survival after neoadjuvant chemotherapy and surgery for patients with nonmetastatic osteosarcoma: a multi-institutional study. Cancer 2015;121:3844–3852.
- 182.↑
Bertrand TE, Cruz A, Binitie O, et al. Do surgical margins affect local recurrence and survival in extremity, nonmetastatic, high-grade osteosarcoma? Clin Orthop Relat Res 2016;474:677–683.
- 183.↑
Lee RJ, Arshi A, Schwartz HC, Christensen RE. Characteristics and prognostic factors of osteosarcoma of the jaws: a retrospective cohort study. JAMA Otolaryngol Head Neck Surg 2015;141:470–477.
- 184.↑
Xin S, Wei G. Prognostic factors in osteosarcoma: a study level meta-analysis and systematic review of current practice. J Bone Oncol 2020;21:100281.
- 185.↑
Collins M, Wilhelm M, Conyers R, et al. Benefits and adverse events in younger versus older patients receiving neoadjuvant chemotherapy for osteosarcoma: findings from a meta-analysis. J Clin Oncol 2013;31:2303–2312.
- 186.↑
Altaf S, Enders F, Jeavons E, et al. High-BMI at diagnosis is associated with inferior survival in patients with osteosarcoma: a report from the Children’s Oncology Group. Pediatr Blood Cancer 2013;60:2042–2046.
- 187.↑
Bacci G, Briccoli A, Ferrari S, et al. Neoadjuvant chemotherapy for osteosarcoma of the extremities with synchronous lung metastases: treatment with cisplatin, adriamycin and high dose of methotrexate and ifosfamide. Oncol Rep 2000;7:339–346.
- 188.↑
Daw NC, Billups CA, Rodriguez-Galindo C, et al. Metastatic osteosarcoma. Cancer 2006;106:403–412.
- 189.↑
Fu Y, Lan T, Cai H, et al. Meta-analysis of serum lactate dehydrogenase and prognosis for osteosarcoma. Medicine (Baltimore) 2018;97:e0741.
- 190.↑
Gu R, Sun Y. Does serum alkaline phosphatase level really indicate the prognosis in patients with osteosarcoma? A meta-analysis. J Cancer Res Ther 2018;14:S468–472.
- 191.↑
Sun J, Xu H, Qi M, et al. Identification of key genes in osteosarcoma by meta-analysis of gene expression microarray. Mol Med Rep 2019;20:3075–3084.
- 192.↑
Marulanda GA, Henderson ER, Johnson DA, et al. Orthopedic surgery options for the treatment of primary osteosarcoma. Cancer Control 2008;15:13–20.
- 193.↑
Bacci G, Ferrari S, Lari S, et al. Osteosarcoma of the limb. Amputation or limb salvage in patients treated by neoadjuvant chemotherapy. J Bone Joint Surg Br 2002;84:88–92.
- 194.↑
Mavrogenis AF, Abati CN, Romagnoli C, Ruggieri P. Similar survival but better function for patients after limb salvage versus amputation for distal tibia osteosarcoma. Clin Orthop Relat Res 2012;470:1735–1748.
- 195.↑
Simon MA, Aschliman MA, Thomas N, Mankin HJ. Limb-salvage treatment versus amputation for osteosarcoma of the distal end of the femur. J Bone Joint Surg Am 2005;87:2822.
- 196.↑
Aksnes LH, Bauer HCF, Jebsen NL, et al. Limb-sparing surgery preserves more function than amputation: a Scandinavian sarcoma group study of 118 patients. J Bone Joint Surg Br 2008;90:786–794.
- 197.↑
Nagarajan R, Neglia JP, Clohisy DR, Robison LL. Limb salvage and amputation in survivors of pediatric lower-extremity bone tumors: what are the long-term implications? J Clin Oncol 2002;20:4493–4501.
- 198.↑
Carrle D, Bielack SS. Current strategies of chemotherapy in osteosarcoma. Int Orthop 2006;30:445–451.
- 199.↑
Winkler K, Beron G, Kotz R, et al. Neoadjuvant chemotherapy for osteogenic sarcoma: results of a Cooperative German/Austrian study. J Clin Oncol 1984;2:617–624.
- 200.↑
Link MP, Goorin AM, Miser AW, et al. The effect of adjuvant chemotherapy on relapse-free survival in patients with osteosarcoma of the extremity. N Engl J Med 1986;314:1600–1606.
- 201.↑
Eilber F, Giuliano A, Eckardt J, et al. Adjuvant chemotherapy for osteosarcoma: a randomized prospective trial. J Clin Oncol 1987;5:21–26.
- 202.↑
Link MP, Goorin AM, Horowitz M, et al. Adjuvant chemotherapy of high-grade osteosarcoma of the extremity. Clin Orthop Relat Res 1991:8–14.
- 203.↑
Meyers PA, Heller G, Healey J, et al. Chemotherapy for nonmetastatic osteogenic sarcoma: the Memorial Sloan-Kettering experience. J Clin Oncol 1992;10:5–15.
- 204.↑
Goorin AM, Schwartzentruber DJ, Devidas M, et al. Presurgical chemotherapy compared with immediate surgery and adjuvant chemotherapy for nonmetastatic osteosarcoma: Pediatric Oncology Group Study POG-8651. J Clin Oncol 2003;21:1574–1580.
- 205.↑
Bramwell VH, Burgers M, Sneath R, et al. A comparison of two short intensive adjuvant chemotherapy regimens in operable osteosarcoma of limbs in children and young adults: the first study of the European Osteosarcoma Intergroup. J Clin Oncol 1992;10:1579–1591.
- 206.↑
Souhami RL, Craft AW, Van der Eijken JW, et al. Randomised trial of two regimens of chemotherapy in operable osteosarcoma: a study of the European Osteosarcoma Intergroup. Lancet 1997;350:911–917.
- 207.↑
Fuchs N, Bielack SS, Epler D, et al. Long-term results of the co-operative German-Austrian-Swiss osteosarcoma study group’s protocol COSS-86 of intensive multidrug chemotherapy and surgery for osteosarcoma of the limbs. Ann Oncol 1998;9:893–899.
- 208.↑
Bacci G, Ferrari S, Bertoni F, et al. Long-term outcome for patients with nonmetastatic osteosarcoma of the extremity treated at the istituto ortopedico rizzoli according to the istituto ortopedico rizzoli/osteosarcoma-2 protocol: an updated report. J Clin Oncol 2000;18:4016–4027.
- 209.↑
Bacci G, Briccoli A, Ferrari S, et al. Neoadjuvant chemotherapy for osteosarcoma of the extremity: long-term results of the Rizzoli’s 4th protocol. Eur J Cancer 2001;37:2030–2039.
- 210.↑
Ferrari S, Smeland S, Mercuri M, et al. Neoadjuvant chemotherapy with high-dose Ifosfamide, high-dose methotrexate, cisplatin, and doxorubicin for patients with localized osteosarcoma of the extremity: a joint study by the Italian and Scandinavian Sarcoma Groups. J Clin Oncol 2005;23:8845–8852.
- 211.↑
Lewis IJ, Nooij MA, Whelan J, et al. Improvement in histologic response but not survival in osteosarcoma patients treated with intensified chemotherapy: a randomized phase III trial of the European Osteosarcoma Intergroup. J Natl Cancer Inst 2007;99:112–128.
- 212.↑
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 Clin Oncol 2008;26:633–638.
- 213.↑
Winkler K, Beron G, Delling G, et al. Neoadjuvant chemotherapy of osteosarcoma: results of a randomized cooperative trial (COSS-82) with salvage chemotherapy based on histological tumor response. J Clin Oncol 1988;6:329–337.
- 214.↑
Li Z, Ma X, Wang Z, et al. A meta-analysis of the efficacy and safety of first-line chemotherapeutic agents for osteosarcoma. Adv Clin Exp Med 2024;33:445–454.
- 215.↑
Basaran M, Bavbek ES, Saglam S, et al. A phase II study of cisplatin, ifosfamide and epirubicin combination chemotherapy in adults with nonmetastatic and extremity osteosarcomas. Oncology 2007;72:255–260.
- 216.↑
Le Deley MC, Guinebretiere JM, Gentet JC, et al. SFOP OS94: a randomised trial comparing preoperative high-dose methotrexate plus doxorubicin to high-dose methotrexate plus etoposide and ifosfamide in osteosarcoma patients. Eur J Cancer 2007;43:752–761.
- 217.↑
Provisor AJ, Ettinger LJ, Nachman JB, et al. Treatment of nonmetastatic osteosarcoma of the extremity with preoperative and postoperative chemotherapy: a report from the Children’s Cancer Group. J Clin Oncol 1997;15:76–84.
- 218.↑
Bernthal NM, Federman N, Eilber FR, et al. Long-term results (>25 years) of a randomized, prospective clinical trial evaluating chemotherapy in patients with high-grade, operable osteosarcoma. Cancer 2012;118:5888–5893.
- 219.↑
Bacci G, Mercuri M, Longhi A, et al. Grade of chemotherapy-induced necrosis as a predictor of local and systemic control in 881 patients with non-metastatic osteosarcoma of the extremities treated with neoadjuvant chemotherapy in a single institution. Eur J Cancer 2005;41:2079–2085.
- 220.↑
Cesari M, Alberghini M, Vanel D, et al. Periosteal osteosarcoma: a single-institution experience. Cancer 2011;117:1731–1735.
- 221.↑
Grimer RJ, Bielack S, Flege S, et al. Periosteal osteosarcoma–a European review of outcome. Eur J Cancer 2005;41:2806–2811.
- 222.↑
Tsukamoto S, Righi A, Kido A, et al. Effect of adjuvant chemotherapy on periosteal osteosarcoma: a systematic review. Jpn J Clin Oncol 2022;52:896–904.
- 223.↑
Bacci G, Ferrari S, Tienghi A, et al. A comparison of methods of loco- regional chemotherapy combined with systemic chemotherapy as neo-adjuvant treatment of osteosarcoma of the extremity. Eur J Surg Oncol 2001;27:98–104.
- 224.↑
Guadagnolo BA, Zagars GK, Raymond AK, et al. Osteosarcoma of the jaw/craniofacial region: outcomes after multimodality treatment. Cancer 2009;115:3262–3270.
- 225.↑
Ciernik IF, Niemierko A, Harmon DC, et al. Proton-based radiotherapy for unresectable or incompletely resected osteosarcoma. Cancer 2011;117:4522–4530.
- 226.↑
DeLaney TF, Park L, Goldberg SI, et al. Radiotherapy for local control of osteosarcoma. Int J Radiat Oncol Biol Phys 2005;61:492–498.
- 227.↑
Marina NM, Smeland S, Bielack SS, et al. Comparison of MAPIE versus MAP in patients with a poor response to preoperative chemotherapy for newly diagnosed high-grade osteosarcoma (EURAMOS-1): an open- label, international, randomised controlled trial. Lancet Oncol 2016;17:1396–1408.
- 228.↑
Smeland S, Muller C, Alvegard TA, et al. Scandinavian Sarcoma Group Osteosarcoma Study SSG VIII: prognostic factors for outcome and the role of replacement salvage chemotherapy for poor histological responders. Eur J Cancer 2003;39:488–494.
- 229.↑
Smeland S, Bruland OS, Hjorth L, et al. Results of the Scandinavian Sarcoma Group XIV protocol for classical osteosarcoma: 63 patients with a minimum follow-up of 4 years. Acta Orthop 2011;82:211–216.
- 230.↑
Ferrari S, Ruggieri P, Cefalo G, et al. Neoadjuvant chemotherapy with methotrexate, cisplatin, and doxorubicin with or without ifosfamide in nonmetastatic osteosarcoma of the extremity: an Italian Sarcoma Group trial ISG/OS-1. J Clin Oncol 2012;30:2112–2118.
- 231.↑
Bielack SS, Smeland S, Whelan JS, et al. Methotrexate, doxorubicin, and cisplatin (MAP) plus maintenance pegylated interferon alfa-2b versus MAP alone in patients with resectable high-grade osteosarcoma and good histologic response to preoperative MAP: first results of the EURAMOS-1 good response randomized controlled trial. J Clin Oncol 2015;33:2279–2287.
- 232.↑
Meyers PA, Heller G, Healey JH, et al. Osteogenic sarcoma with clinically detectable metastasis at initial presentation. J Clin Oncol 1993;11:449–453.
- 234.↑
Bacci G, Briccoli A, Mercuri M, et al. Osteosarcoma of the extremities with synchronous lung metastases: long-term results in 44 patients treated with neoadjuvant chemotherapy. J Chemother 1998;10:69–76.
- 235.↑
Bacci G, Briccoli A, Rocca M, et al. Neoadjuvant chemotherapy for osteosarcoma of the extremities with metastases at presentation: recent experience at the Rizzoli Institute in 57 patients treated with cisplatin, doxorubicin, and a high dose of methotrexate and ifosfamide. Ann Oncol 2003;14:1126–1134.
- 236.↑
Smeland S, Bielack SS, Whelan J, et al. Survival and prognosis with osteosarcoma: outcomes in more than 2000 patients in the EURAMOS-1 (European and American Osteosarcoma Study) cohort. Eur J Cancer 2019;109:36–50.
- 237.↑
Goorin AM, Harris MB, Bernstein M, et al. Phase II/III trial of etoposide and high-dose ifosfamide in newly diagnosed metastatic osteosarcoma: a pediatric oncology group trial. J Clin Oncol 2002;20:426–433.
- 238.↑
Winkler K, Torggler S, Beron G, et al. [Results of treatment in primary disseminated osteosarcoma. Analysis of the follow-up of patients in the cooperative osteosarcoma studies COSS-80 and COSS-82]. Onkologie 1989;12:92–96.
- 239.↑
Bacci G, Mercuri M, Briccoli A, et al. Osteogenic sarcoma of the extremity with detectable lung metastases at presentation. Results of treatment of 23 patients with chemotherapy followed by simultaneous resection of primary and metastatic lesions. Cancer 1997;79:245–254.
- 240.↑
Mettmann VL, Blattmann C, Friedel G, et al. Primary multi-systemic metastases in osteosarcoma: presentation, treatment, and survival of 83 patients of the Cooperative Osteosarcoma Study Group. Cancers (Basel) 2024;16:275.
- 241.↑
Daw NC, Chou AJ, Jaffe N, et al. Recurrent osteosarcoma with a single pulmonary metastasis: a multi-institutional review. Br J Cancer 2015;112:278–282.
- 242.↑
Tabone MD, Kalifa C, Rodary C, et al. Osteosarcoma recurrences in pediatric patients previously treated with intensive chemotherapy. J Clin Oncol 1994;12:2614–2620.
- 243.↑
Saeter G, Hoie J, Stenwig AE, et al. Systemic relapse of patients with osteogenic sarcoma. Prognostic factors for long term survival. Cancer 1995;75:1084–1093.
- 244.↑
Ferrari S, Briccoli A, Mercuri M, et al. Postrelapse survival in osteosarcoma of the extremities: prognostic factors for long-term survival. J Clin Oncol 2003;21:710–715.
- 245.↑
Buddingh EP, Anninga JK, Versteegh MI, et al. Prognostic factors in pulmonary metastasized high-grade osteosarcoma. Pediatr Blood Cancer 2010;54:216–221.
- 246.↑
Briccoli A, Rocca M, Salone M, et al. High grade osteosarcoma of the extremities metastatic to the lung: long-term results in 323 patients treated combining surgery and chemotherapy, 1985–2005. Surg Oncol 2010;19:193–199.
- 247.↑
Bielack SS, Kempf-Bielack B, Branscheid D, et al. Second and subsequent recurrences of osteosarcoma: presentation, treatment, and outcomes of 249 consecutive cooperative osteosarcoma study group patients. J Clin Oncol 2009;27:557–565.
- 248.↑
Berger M, Grignani G, Ferrari S, et al. Phase 2 trial of two courses of cyclophosphamide and etoposide for relapsed high-risk osteosarcoma patients. Cancer 2009;115:2980–2987.
- 249.↑
Gentet JC, Brunat-Mentigny M, Demaille MC, et al. Ifosfamide and etoposide in childhood osteosarcoma. A phase II study of the French Society of Paediatric Oncology. Eur J Cancer 1997;33:232–237.
- 250.↑
Miser JS, Kinsella TJ, Triche TJ, et al. Ifosfamide with mesna uroprotection and etoposide: an effective regimen in the treatment of recurrent sarcomas and other tumors of children and young adults. J Clin Oncol 1987;5:1191–1198.
- 251.↑
Duffaud F, Mir O, Boudou-Rouquette P, et al. Efficacy and safety of regorafenib in adult patients with metastatic osteosarcoma: a non-comparative, randomised, double-blind, placebo-controlled, phase 2 study. Lancet Oncol 2019;20:120–133.
- 252.↑
Davis LE, Bolejack V, Ryan CW, et al. Randomized double-blind phase II study of regorafenib in patients with metastatic osteosarcoma. J Clin Oncol 2019;37:1424–1431.
- 253.↑
Merimsky O, Meller I, Flusser G, et al. Gemcitabine in soft tissue or bone sarcoma resistant to standard chemotherapy: a phase II study. Cancer Chemother Pharmacol 2000;45:177–181.
- 254.↑
Palmerini E, Jones RL, Marchesi E, et al. Gemcitabine and docetaxel in relapsed and unresectable high-grade osteosarcoma and spindle cell sarcoma of bone. BMC Cancer 2016;16:280.
- 255.↑
Magnan H, Goodbody CM, Riedel E, et al. Ifosfamide dose-intensification for patients with metastatic Ewing sarcoma. Pediatr Blood Cancer 2015;62:594–597.
- 256.↑
Maki RG, Wathen JK, Patel SR, et al. Randomized phase II study of gemcitabine and docetaxel compared with gemcitabine alone in patients with metastatic soft tissue sarcomas: results of sarcoma alliance for research through collaboration study 002 [corrected]. J Clin Oncol 2007;25:2755–2763.
- 257.↑
Low K, Foulkes P, Hills F, et al. The efficacy of gemcitabine and docetaxel chemotherapy for the treatment of relapsed and refractory osteosarcoma: a systematic review and pre-clinical study. Cancer Med 2024;13:e70248.
- 258.↑
Subbiah V, Anderson PM, Kairemo K, et al. Alpha particle radium 223 dichloride in high-risk osteosarcoma: a phase I dose escalation trial. Clin Cancer Res 2019;25:3802–3810.
- 259.↑
Anderson PM, Wiseman GA, Dispenzieri A, et al. High-dose samarium-153 ethylene diamine tetramethylene phosphonate: low toxicity of skeletal irradiation in patients with osteosarcoma and bone metastases. J Clin Oncol 2002;20:189–196.
- 260.↑
Loeb DM, Garrett-Mayer E, Hobbs RF, et al. Dose-finding study of 153Sm-EDTMP in patients with poor-prognosis osteosarcoma. Cancer 2009;115:2514–2522.
- 261.↑
Grignani G, Palmerini E, Dileo P, et al. A phase II trial of sorafenib in relapsed and unresectable high-grade osteosarcoma after failure of standard multimodal therapy: an Italian Sarcoma Group study. Ann Oncol 2012;23:508–516.
- 262.↑
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. Lancet Oncol 2015;16:98–107.
- 263.↑
Lashkari A, Chow WA, Valdes F, et al. Tandem high-dose chemotherapy followed by autologous transplantation in patients with locally advanced or metastatic sarcoma. Anticancer Res 2009;29:3281–3288.
- 264.↑
Fagioli F, Aglietta M, Tienghi A, et al. High-dose chemotherapy in the treatment of relapsed osteosarcoma: an Italian sarcoma group study. J Clin Oncol 2002;20:2150–2156.
- 265.↑
Elledge CR, Krasin MJ, Ladra MM, et al. A multi-institutional phase 2 trial of stereotactic body radiotherapy in the treatment of bone metastases in pediatric and young adult patients with sarcoma. Cancer 2021;127:739–747.