Learn about the medical, dental, pharmacy, behavioral, and voluntary benefits your employer may offer.
Dramatic improvements in survival have been achieved for children and adolescents with cancer.[
Studies using immunohistochemical markers,[
The World Health Organization (WHO) classification of tumors of soft tissue and bone was modified in 2020 to introduce a new chapter on undifferentiated small round cell sarcomas of bone and soft tissue. This chapter consists of Ewing sarcoma and three main categories, including round cell sarcomas with EWSR1::non-ETS fusions, CIC-rearranged sarcoma, and sarcomas with BCOR genetic alterations.[
Before the widespread availability of genomic testing, Ewing sarcoma was identified by the appearance of small round blue cells on light microscopic examination, along with positive staining for CD99 by immunohistochemistry. The identification of the recurring t(11;22) translocation in most Ewing sarcoma tumors led to the discovery that most tumors classified as Ewing sarcoma had a translocation that juxtaposed a portion of the EWSR1 gene to a portion of an ETS gene family member, resulting in a transforming transcript. Not all undifferentiated small round cell sarcomas of bone and soft tissue have such a translocation. Further research identified additional genetic changes, including tumors with translocations of the CIC gene or the BCOR gene. These groups of tumors occur much less frequently than Ewing sarcoma, and definitive clinical outcomes for these patients are based on smaller sample sizes and less homogeneous treatment; therefore, patient outcomes are harder to quantitate with precision. Most of these tumors have been treated with regimens designed for Ewing sarcoma, and there is consensus that they were often included in clinical trials for the treatment of Ewing sarcoma, sometimes as translocation-negative Ewing sarcoma. There is agreement that these tumors are sufficiently different from Ewing sarcoma. These tumors should be stratified and analyzed separately from Ewing sarcoma with the common translocation, even if they are treated with similar therapy. In this summary, they are described separately. For more information about these smaller groups of tumors, see the following sections:
Incidence
The incidence of Ewing sarcoma has remained unchanged for 30 years.[
The relative paucity of Ewing sarcoma in people of African or Asian descent may be explained, in part, by a specific polymorphism in the EGR2 gene.[
The median age of patients with Ewing sarcoma is 15 years, and more than 50% of patients are adolescents. Well-characterized cases of Ewing sarcoma in neonates and infants have been described.[
Genetic Predisposition to Ewing Sarcoma
Conventional understanding of translocation-driven sarcoma such as Ewing sarcoma suggests that these patients do not have a genetic predisposition.[
Clinical Presentation
Primary sites of bone disease include the following:
For extraosseous primary tumors, the most common primary sites of disease include the following:[
The time from the first symptom to diagnosis of Ewing sarcoma is often long, with a median interval reported from 2 to 5 months. Longer times are associated with older age and pelvic primary sites. Time from the first symptom to diagnosis has not been associated with metastasis, surgical outcome, or survival.[
The Surveillance, Epidemiology, and End Results (SEER) Program database was used to compare patients younger than 40 years with Ewing sarcoma who presented with skeletal and extraosseous primary sites (see Table 1).[
Characteristic | Extraosseous Ewing Sarcoma | Skeletal Ewing Sarcoma | P Value |
---|---|---|---|
a Adapted from Applebaum et al.[ |
|||
Mean age (range), years | 20 (0–39) | 16 (0–39) | <.001 |
Male | 53% | 63% | <.001 |
White race | 85% | 93% | <.001 |
Axial primary sites | 73% | 54% | <.001 |
Pelvic primary sites | 20% | 27% | .001 |
Diagnostic Evaluation
The following tests and procedures may be used to diagnose or stage Ewing sarcoma:
For primary appendicular bone tumors, imaging of the entire involved bone is standardly performed to evaluate for skip metastasis. In one retrospective study, skip metastasis was seen in 15.8% of patients. The presence of skip metastasis was associated with an increased risk of distant metastatic disease.[
A systematic review of Ewing sarcoma studies was performed to assess the incidence of bone marrow metastasis and the role of fluorine F 18-fludeoxyglucose (18F-FDG) PET imaging to detect bone marrow metastasis.[
Prognostic Factors
The two major types of prognostic factors for patients with Ewing sarcoma are grouped as follows:
Pretreatment factors
Patients with metastatic disease confined to the lung have a better prognosis than do patients with extrapulmonary metastatic sites.[
Patients with metastasis to only bone seem to have a better outcome than do patients with metastases to both bone and lung.[
Based on an analysis from the SEER database, regional lymph node involvement in patients is associated with an inferior overall outcome when compared with patients without regional lymph node involvement.[
In a study of 299 patients with Ewing sarcoma, 41 patients (14%) had STAG2 variants and 16 patients (5%) had TP53 variants.[
The COG analyzed STAG2 expression by immunohistochemistry in children with Ewing sarcoma who participated in frontline treatment trials.[
The following are not considered to be adverse prognostic factors for Ewing sarcoma:
Response to initial therapy factors
Multiple studies have shown that patients with minimal or no residual viable tumor after presurgical chemotherapy have a significantly better EFS than do patients with larger amounts of viable tumor.[
Patients with poor response to presurgical chemotherapy have an increased risk of local recurrence.[
A retrospective analysis of risk factors for recurrence was performed in patients who received initial chemotherapy and underwent surgical resection of the primary tumor.[
Detection of Ewing sarcoma in the peripheral blood
Several techniques to evaluate the presence of Ewing sarcoma in the peripheral blood have been proposed. Flow cytometry for cells that express the CD99 antigen was not sufficiently sensitive to serve as a reliable biomarker.[
A more sensitive technique that used patient-specific primers designed after identification of the specific translocation breakpoint in combination with droplet digital PCR reported a sensitivity of 0.009% to 0.018%.[
A hybrid capture sequencing assay employing the introns at which EWSR1 and FLI1 fusions occur has also been developed to detect evidence of the EWSR1::FLI1 translocation in circulating cell-free DNA.[
References:
Ewing sarcoma belongs to the group of neoplasms commonly referred to as small round blue cell tumors of childhood. The individual cells of Ewing sarcoma contain round-to-oval nuclei, with fine dispersed chromatin without nucleoli. Occasionally, cells with smaller, more hyperchromatic, and probably degenerative nuclei are present, giving a light cell/dark cell pattern. The cytoplasm varies in amount, but in the classic case, it is clear and contains glycogen, which can be highlighted with a periodic acid-Schiff stain. The tumor cells are tightly packed and grow in a diffuse pattern without evidence of structural organization. Tumors with the requisite translocation that show neuronal differentiation are not considered a separate entity, but rather, part of a continuum of differentiation.
CD99 is a surface membrane protein that is expressed in most cases of Ewing sarcoma and is useful in diagnosing these tumors when the results are interpreted in the context of clinical and pathological parameters.[
For more information about the cellular classification of other undifferentiated small round cell sarcomas, see the Undifferentiated Small Round Cell (Ewing-Like) Sarcomas section.
References:
Molecular Features of Ewing Sarcoma
The detection of a translocation involving the EWSR1 gene on chromosome 22 band q12 and any one of a number of partner chromosomes is the key feature in the diagnosis of Ewing sarcoma (see Table 2).[
Besides these consistent aberrations involving the EWSR1 gene at 22q12, additional numerical and structural aberrations have been observed in Ewing sarcoma, including gains of chromosomes 2, 5, 8, 9, 12, and 15; the nonreciprocal translocation t(1;16)(q12;q11.2); and deletions on the short arm of chromosome 6. Trisomy 20 may be associated with a more aggressive subset of Ewing sarcoma.[
Three papers have described the genomic landscape of Ewing sarcoma and all show that these tumors have a relatively silent genome, with a paucity of variants in pathways that might be amenable to treatment with novel targeted therapies.[
A discovery cohort (n = 99) highlighted the frequency of chromosome 8 gain, the co-occurrence of chromosome 1q gain and chromosome 16q loss, the mutual exclusivity of CDKN2A deletion and STAG2 variant, and the relative paucity of recurrent single nucleotide variants for Ewing sarcoma.[
Ewing sarcoma translocations can all be found with standard cytogenetic analysis. A more rapid analysis looking for a break apart of the EWSR1 gene is now frequently done to confirm the diagnosis of Ewing sarcoma molecularly.[
Genome-wide association studies have identified susceptibility loci for Ewing sarcoma at 1p36.22, 10q21, and 15q15.[
TET Family Partner | Fusion With ETS-Like Oncogene Partner | Translocation | Comment |
---|---|---|---|
a These partners are not members of the ETS family of oncogenes. | |||
EWSR1 | EWSR1::FLI1 | t(11;22)(q24;q12) | Most common; approximately 85% to 90% of cases |
EWSR1::ERG | t(21;22)(q22;q12) | Second most common; approximately 10% of cases | |
EWSR1::ETV1 | t(7;22)(p22;q12) | Rare | |
EWSR1::ETV4 | t(17;22)(q12;q12) | Rare | |
EWSR1::FEV | t(2;22)(q35;q12) | Rare | |
EWSR1::NFATC2a | t(20;22)(q13;q12) | Rare | |
EWSR1::POU5F1a | t(6;22)(p21;q12) | ||
EWSR1::SMARCA5a | t(4;22)(q31;q12) | Rare | |
EWSR1::PATZ1a | t(6;22)(p21;q12) | ||
EWSR1::SP3a | t(2;22)(q31;q12) | Rare | |
FUS | FUS::ERG | t(16;21)(p11;q22) | Rare |
FUS::FEV | t(2;16)(q35;p11) | Rare |
References:
Pretreatment staging studies for Ewing sarcoma may include the following:
For patients with confirmed Ewing sarcoma, pretreatment staging studies include MRI and/or CT scan, depending on the primary site. Despite the fact that CT and MRI are both equivalent in terms of staging, use of both imaging modalities may help radiation therapy planning.[
Although 18F-FDG PET or 18F-FDG PET-CT are optional staging modalities, they have demonstrated high sensitivity and specificity in Ewing sarcoma and may provide additional information that alters therapy planning. In one institutional study, 18F-FDG PET had a very high correlation with bone scan; the investigators suggested that it could replace bone scan for the initial extent of disease evaluation.[
Bone marrow aspiration and biopsy have been considered the standard of care for Ewing sarcoma. However, two retrospective studies showed that for patients (N = 141) who were evaluated by bone scan and/or PET scan and lung CT without evidence of metastases, bone marrow aspirates and biopsies were negative in every case.[
For Ewing sarcoma, the tumor is defined as localized when, by clinical and imaging techniques, there is no spread beyond the primary site or regional lymph node involvement. Continuous extension into adjacent soft tissue may occur. If there is a question of regional lymph node involvement, pathological confirmation is indicated.
References:
It is important that patients be evaluated by specialists from the appropriate disciplines (e.g., radiologists, medical oncologists, pathologists, surgical or orthopedic oncologists, and radiation oncologists) as early as possible.
Appropriate imaging studies of the site are obtained before biopsy. To ensure that the incision is placed in an acceptable location, the surgical or orthopedic oncologist who will perform the definitive surgery is involved in the decision regarding biopsy-incision placement. This is especially important if it is thought that the lesion can subsequently be totally excised after initial systemic therapy or if a limb salvage procedure may be attempted. It is almost never appropriate to attempt a primary resection of Ewing sarcoma. With rare exceptions, Ewing sarcoma is sensitive to chemotherapy and will respond to initial systemic therapy. This therapy reduces the risk of tumor spread to surrounding tissues and makes ultimate surgery easier and safer. Biopsy should be from soft tissue as often as possible to avoid increasing the risk of fracture.[
Table 3 describes the treatment options for localized, metastatic, and recurrent Ewing sarcoma.
Treatment Group | Standard Treatment Options | |
---|---|---|
Localized Ewing sarcoma | Chemotherapy | |
Local-control measures: | ||
Surgery | ||
Radiation therapy | ||
High-dose chemotherapy with autologous stem cell rescue | ||
Metastatic Ewing sarcoma | Chemotherapy | |
Surgery | ||
Radiation therapy | ||
Recurrent Ewing sarcoma | Chemotherapy(not considered standard treatment) | |
Surgery(not considered standard treatment) | ||
Radiation therapy(not considered standard treatment) | ||
High-dose chemotherapy with stem cell support(not considered standard treatment) | ||
Other therapies(not considered standard treatment) |
The successful treatment of patients with Ewing sarcoma requires systemic chemotherapy [
Chemotherapy for Ewing Sarcoma
Multidrug chemotherapy for Ewing sarcoma always includes vincristine, doxorubicin, ifosfamide, and etoposide. Most protocols also use cyclophosphamide and some incorporate dactinomycin. The mode of administration and dose intensity of cyclophosphamide within courses differs markedly between protocols. A European Intergroup Cooperative Ewing Sarcoma Study (EICESS) trial suggested that 1.2 g of cyclophosphamide produced a similar event-free survival (EFS) compared with 6 g of ifosfamide in patients with lower-risk disease, and identified a trend toward better EFS for patients with localized Ewing sarcoma and higher-risk disease when treatment included etoposide (GER-GPOH-EICESS-92 [NCT00002516]).[
Protocols in the United States generally alternate courses of vincristine, cyclophosphamide, and doxorubicin (VDC) with courses of ifosfamide and etoposide (IE),[
Evidence (chemotherapy):
Local Control (Surgery and Radiation Therapy) for Ewing Sarcoma
Treatment approaches for Ewing sarcoma and therapeutic aggressiveness must be adjusted to maximize local control while also minimizing morbidity.
Surgery is the most commonly used form of local control.[
Timing of local control may impact outcome. A retrospective review from the National Cancer Database identified 1,318 patients with Ewing sarcoma.[
For patients with metastatic Ewing sarcoma, any benefit of combined surgery and radiation therapy compared with either therapy alone for local control is relatively less substantial because the overall prognosis of these patients is much worse than the prognosis of patients who have localized disease.
Randomized trials that directly compare surgery and radiation therapy do not exist, and their relative roles remain controversial. Although retrospective institutional series suggest superior local control and survival with surgery than with radiation therapy, most of these studies are compromised by selection bias. An analysis using propensity scoring to adjust for clinical features that may influence the preference for surgery only, radiation only, or combined surgery and radiation demonstrated that similar EFS is achieved with each mode of local therapy.[
The EURO-EWING-INTERGROUP-EE99 (NCT00020566) trial prospectively treated 180 patients with pelvic primary tumors without clinically detectable metastatic disease.[
For patients who undergo gross-total resection with microscopic residual disease, a radiation therapy dose of 50.4 Gy is indicated; for patients treated with primary radiation therapy, the radiation dose is 55.8 Gy (45 Gy to the initial tumor volume and an additional 10.8 Gy to the postchemotherapy volume).[
Evidence (postoperative radiation therapy):
Thoracic primary tumors
Evidence (surgery):
In summary, surgery is chosen as definitive local therapy for suitable patients, but radiation therapy is appropriate for patients with unresectable disease or those who would experience functional or cosmetic compromise by definitive surgery. The possibility of impaired function or cosmesis needs to be measured against the possibility of second tumors in the radiation field. Adjuvant radiation therapy should be considered for patients with residual microscopic disease or inadequate margins.
When preoperative assessment has suggested a high probability that surgical margins will be close or positive, preoperative radiation therapy has achieved tumor shrinkage and allowed surgical resection with clear margins.[
High-Dose Chemotherapy With Stem Cell Support for Ewing Sarcoma
For patients with a high risk of relapse with conventional treatments, certain investigators have used high-dose chemotherapy with hematopoietic stem cell transplant (HSCT) as consolidation treatment, in an effort to improve outcome.[
Evidence (high-dose therapy with stem cell support):
Both study arms were compromised by the potential for selection bias for patients who were eligible for and accepted randomization, which may limit the generalizability of the results. Only 40% of eligible patients were randomized.
The induction regimen employed in the EURO-EWING-INTERGROUP-EE99 trial was VIDE. This regimen is less dose intensive than the regimen employed in COG studies. This can be inferred from the intended dose intensity of the agents employed for the 21-week period that preceded randomization in the EURO-EWING-INTERGROUP-EE99 study (see Table 4). The lower dose intensity can also be inferred from the outcome of the EURO-EWING-INTERGROUP-EE99 study for patients in the localized disease stratum. Results from this study include the following:
The observation that high-dose therapy with autologous stem cell rescue improved outcomes for patients with a poor response to initial therapy in the EURO-EWING-INTERGROUP-EE99 study must be interpreted in this context. The advantage of high-dose therapy as consolidation for patients with a poor response to initial treatment with a less intensive regimen cannot be extrapolated to a population of patients who received a more intensive treatment regimen as initial therapy.
Chemotherapy Agent | Prescribed Dose Intensity (mg/week) | |
---|---|---|
| EURO-EWING-INTERGROUP-EE99 Trial[ |
COG Interval Dose Compression[ |
COG = Children's Oncology Group. | ||
Vincristine | 0.5 mg/m2 | 0.43 mg/m2 |
Doxorubicin | 17.1 mg/m2 | 21.4 mg/m2 |
Ifosfamide | 3,000 mg/m2 | 2,150 mg/m2 |
Cyclophosphamide | 0 | 343 mg/m2 |
Cyclophosphamide equivalent dose (= cyclophosphamide dose + ifosfamide dose × 0.244) | 732 mg/m2 | 868 mg/m2 |
Extraosseous Ewing Sarcoma
Multiple analyses have evaluated diagnostic findings, treatment, and outcome of patients with bone lesions at the following anatomical primary sites:
Extraosseous Ewing sarcoma is biologically similar to Ewing sarcoma arising in bone. Historically, most children and young adults with extraosseous Ewing sarcoma were treated on protocols designed for the treatment of rhabdomyosarcoma. This is important because many of the treatment regimens for rhabdomyosarcoma do not include an anthracycline, which is a critical component of current treatment regimens for Ewing sarcoma. Currently, patients with extraosseous Ewing sarcoma are eligible for studies that include Ewing sarcoma of bone.
Evidence (treatment of extraosseous Ewing sarcoma):
Cutaneous Ewing sarcoma is a soft tissue tumor in the skin or subcutaneous tissue that seems to behave as a less-aggressive tumor than primary bone or soft tissue Ewing sarcoma. Tumors can form throughout the body, although the extremity is the most common site, and they are almost always localized.
Evidence (treatment of cutaneous Ewing sarcoma):
References:
Cancer in children and adolescents is rare, although the overall incidence has been slowly increasing since 1975.[
For specific information about supportive care for children and adolescents with cancer, see the summaries on
The American Academy of Pediatrics has outlined guidelines for pediatric cancer centers and their role in the treatment of pediatric patients with cancer.[
Childhood and adolescent cancer survivors require close monitoring because side effects of cancer therapy may persist or develop months or years after treatment. For specific information about the incidence, type, and monitoring of late effects in childhood and adolescent cancer survivors, see Late Effects of Treatment for Childhood Cancer.
References:
Standard treatment options for localized Ewing sarcoma include the following:
Because most patients with apparently localized disease at diagnosis have occult metastatic disease, multidrug chemotherapy and local disease control with surgery and/or radiation therapy is indicated in the treatment of all patients.[
Chemotherapy
Current standard chemotherapy in the United States includes vincristine, doxorubicin, and cyclophosphamide (VDC), alternating with ifosfamide and etoposide (IE) or VDC/IE.[
Evidence (chemotherapy):
Local-Control Measures
Local control can be achieved by surgery and/or radiation therapy. Decisions regarding the optimal modality for local control for an individual patient involve consideration of the following:
An analysis using propensity scoring (a method that adjusts for the inherent selection bias of the location and size of the tumor) to adjust for clinical features that may influence the preference for surgery only, radiation only, or combined surgery and radiation demonstrated that similar EFS rates are achieved with each mode of local therapy after propensity adjustment.[
Surgery
Surgery is generally the preferred approach if the lesion is resectable.[
A single-institution retrospective analysis of 78 patients with Ewing sarcoma suggested that pathological fracture at initial presentation was associated with inferior EFS and OS.[
Radiation therapy
Radiation therapy is usually employed in the following cases:
Radiation therapy is delivered in a setting in which stringent planning techniques are applied by those experienced in the treatment of Ewing sarcoma. Such an approach will result in local control of the tumor with acceptable morbidity in most patients.[
The radiation dose may be adjusted depending on the extent of residual disease after the initial surgical procedure. When no surgical resection is performed, radiation therapy is generally administered in fractionated doses totaling approximately 55.8 Gy to the prechemotherapy tumor volume. A randomized study of 40 patients with Ewing sarcoma using 55.8 Gy to the prechemotherapy tumor extent with a 2-cm margin compared with the same total-tumor dose after 39.6 Gy to the entire bone showed no difference in local control or EFS.[
Preoperative radiation therapy is an approach that can be used when surgical resection is deemed possible but with the likelihood of microscopic residual disease. A panel of international expert clinicians used a three-stage modified Delphi technique to develop consensus statements about local treatment. The panel reached a strong consensus that preoperative radiation therapy may be given when an inadequate (marginal) margin at resection is foreseen on imaging.[
For patients with residual disease after an attempt at surgical resection, the Intergroup Ewing Sarcoma Study (INT-0091) recommended 45 Gy to the original disease site plus a 10.8 Gy boost for patients with gross residual disease and 45 Gy plus a 5.4 Gy boost for patients with microscopic residual disease. No radiation therapy was recommended for those who had no evidence of microscopic residual disease after surgical resection.[
For patients who are deemed to have unresectable disease after induction chemotherapy, radiation therapy is given, using the same doses as those administered for patients with partially resected disease.[
In a single-institution nonrandomized study, patients who had primary tumors 8 cm or larger were treated with higher-dose radiation therapy (median dose, 64.8 Gy). The 5-year cumulative incidence of local failure rate was 6.6%, which compares favorably to other published local failure rates in this group of patients.[
Comparison of proton-beam radiation therapy and intensity-modulated radiation therapy (IMRT) treatment plans has shown that proton-beam radiation therapy can spare more normal tissue adjacent to Ewing sarcoma primary tumors than IMRT.[
Higher rates of local failure are seen in patients older than 14 years who have tumors larger than 8 cm in length.[
Radiation therapy is associated with the development of subsequent neoplasms. A retrospective study noted that patients who received 60 Gy or more had an incidence of second malignancy of 20%. Patients who received 48 Gy to 60 Gy had an incidence of 5%, and those who received less than 48 Gy did not develop a second malignancy.[
High-Dose Chemotherapy With Autologous Stem Cell Rescue
Evidence (high-dose chemotherapy with autologous stem cell rescue):
Both study arms were compromised by the potential for selection bias for patients who were eligible for and accepted randomization, which may limit the generalizability of the results. Only 40% of eligible patients were randomized.
The induction regimen employed in the EURO-EWING-INTERGROUP-EE99 trial included VIDE. This regimen is less dose intensive than the regimen employed in COG studies. This can be inferred from the intended dose intensity of the agents employed for the 21-week period that preceded randomization in the EURO-EWING-INTERGROUP-EE99 study (see Table 4). The lower dose intensity can also be inferred from the outcome of the EURO-EWING-INTERGROUP-EE99 study for patients in the localized disease stratum. Results from this study include the following:
The observation that high-dose therapy with autologous stem cell rescue improved outcomes for patients with a poor response to initial therapy in the EURO-EWING-INTERGROUP-EE99 study must be interpreted in this context. The advantage of high-dose therapy as consolidation for patients with a poor response to initial treatment with a less intensive regimen cannot be extrapolated to a population of patients who received a more intensive treatment regimen as initial therapy.
Current Clinical Trials
Use our
References:
Approximately 25% of patients with Ewing sarcoma have metastases at diagnosis.[
The following factors independently predict a poor outcome in patients presenting with metastatic disease:[
Standard treatment options for metastatic Ewing sarcoma include the following:
Chemotherapy
For patients with metastatic Ewing sarcoma, standard treatment that uses alternating cycles of vincristine/doxorubicin/cyclophosphamide and ifosfamide/etoposide (VDC/IE) combined with adequate local-control measures applied to both primary and metastatic sites often results in complete or partial responses; however, the overall cure rate is 20%.[
The following chemotherapy regimens have not shown benefit:
Surgery and Radiation Therapy
Systematic use of surgery and radiation therapy for metastatic sites may improve overall outcome in patients with extrapulmonary metastases.
Evidence (surgery and radiation therapy):
These results must be interpreted with caution. The patients who received local-control therapy to all known sites of metastatic disease were selected by the treating investigator, not randomly assigned. Patients with so many metastases that radiation to all sites would result in bone marrow failure were not selected to receive radiation to all sites of metastatic disease. Patients who did not achieve control of the primary tumor did not go on to have local control of all sites of metastatic disease. There was a selection bias such that while all patients in these reports had multiple sites of metastatic disease, the patients who had surgery and/or radiation therapy to all sites of clinically detectable metastatic disease had better responses to systemic therapy and fewer sites of metastasis than did patients who did not undergo similar therapy of metastatic sites.
Radiation therapy, delivered in a setting in which stringent planning techniques are applied by those experienced in the treatment of Ewing sarcoma, should be considered. Such an approach will result in local control of the tumor with acceptable morbidity in most patients.[
The radiation dose depends on the metastatic site of disease:
Other Therapies
More intensive therapies, many of which incorporate high-dose chemotherapy with or without total-body irradiation in conjunction with stem cell support, have not shown improvement in EFS rates for patients with bone and/or bone marrow metastases.[
Current Clinical Trials
Use our
References:
Recurrence of Ewing sarcoma is most common within 2 years of initial diagnosis (approximately 80%).[
The overall prognosis for patients with recurrent Ewing sarcoma is poor; the 5-year survival rate after recurrence is approximately 10% to 15%.[
Prognostic factors include the following:
The selection of treatment for patients with recurrent disease depends on many factors, including the following:
There is no standardized second-line treatment for patients with relapsed or refractory Ewing sarcoma. Most patients in first relapse are treated with conventional systemic chemotherapy. Patients who demonstrate a response to therapy may undergo local control to sites of recurrence.
Treatment options for recurrent Ewing sarcoma include the following:
Chemotherapy
Combinations of chemotherapy, such as cyclophosphamide and topotecan or irinotecan and temozolomide with or without vincristine, are active in recurrent Ewing sarcoma and can be considered for these patients.[
Study Reference | Trial Phase (Total No. of Patients) | Median Age (Range) (y) | CR/PR | RR | Cyclophosphamide (mg/m2)/Topotecan (mg/m2) × d | Other Agents |
---|---|---|---|---|---|---|
II = phase II trial; CR = complete response; PR = partial response; R = retrospective; RR = objective response rate; VCR = vincristine. | ||||||
Saylors et al.[ |
II (17) | 13.8 (1–21) | 1/3 | 29% | 250 × 5/0.75 × 5 | None |
Hunold et al.[ |
R (54) | 17.4 (3–49) | 0/16 | 30% | 250 × 5/0.75 × 5 | None |
Farhat et al.[ |
R (14) | 11 (2–19) | 0/3 | 21% | 250 × 5/0.75 × 5 | None |
Kebudi et al.[ |
R (14) | 13 (3–16) | 2/5 | 50% | 250 × 5/0.75 × 5 | VCR |
These studies were retrospective, not prospective; prospective trials with clearly defined eligibility cohorts and intent-to-treat analyses are lacking. When combined, these studies accrued 99 patients and observed 3 complete remissions and 27 partial remissions. The objective response rate was 30%.
Study Reference | Trial Phase (Total No. of Patients) | Median Age (Range) (y) | CR/PR | RR | Temozolomide (mg/m2)/Irinotecan (mg/m2) × d × wk | Other Agents |
---|---|---|---|---|---|---|
I = phase I trial; II = phase II trial; Acta Onc = Acta Oncologica; An Ped = Annals of Pediatrics; Clin Cancer Res = Clinical Cancer Research; Exp Opin = Expert Opinion Investigational Drugs; Clin Transl Oncol = Clinical and Translational Oncology; BEV = bevacizumab; CR = complete response; IV = intravenous; N/A = not applicable; PBC = Pediatric Blood and Cancer; Ped Hem Onc = Pediatric Hematology and Oncology; PO = oral; PR = partial response; R = retrospective trial; RR = objective response rate; TMS = temsirolimus; UK = unknown; VCR = vincristine. | ||||||
Wagner et al. (PBC, 2007)[ |
R (16) | 18 (7–33) | 1/3 | 29% | 100 × 5/IV 10–20 × 5 × 2 | None |
Casey et al. (PBC, 2009)[ |
R (19) | 19.5 (2–40) | 5/7 | 63% | 100 × 5/IV 20 × 5 × 2 | None |
Hernandez-Marques et al. (An Ped, 2013)[ |
R (8) | 13 (6–18) | 0/3 | 37% | 80–100 × 5/IV 10–20 × 5 × 2 | None |
Raciborska et al. (PBC, 2013)[ |
R (22) | 14.3 | 5/7 | 54% | 125 × 5/IV 50 × 5 | VCR |
McKnall-Knapp et al. (PBC, 2010)[ |
I (1) | N/A | 0/1 | 100% | 100 × 5/IV 20 × 5 × 2 | VCR |
Wagner et al. (PBC, 2010)[ |
I (5) | (<21) | 1/1 | 40% | 100–150 × 5/PO 35–90 × 5 | VCR |
Wagner et al. (PBC, 2013)[ |
I (2) | 20, 22 | 1/1 | 100% | 150 × 5/PO 90 × 5 | VCR, BEV |
Bagatell et al. (PBC, 2014)[ |
I (7) | (<21) | 0/1 | 14% | 100–150 × 5/PO 50–90 × 5 | TMS |
Kurucu et al. (Ped Hem Onc, 2015)[ |
R (20) | 14 (1–18) | UK | 55% | 100 × 5/IV 20 × 5 × 2 | None |
Anderson et al. (Exp Opin, 2008)[ |
R (25) | 15 | 7/9 | 64% | 100 × 5/IV 10 × 5 × 2 | None |
Palmerini et al. (Acta Onc, 2018)[ |
R (51) | 21 (3–65) | 5/12 | 34% | 100 × 5/IV 40 × 5 | None |
Salah et al. (Clin Transl Oncol, 2021)[ |
R (53) | 20 (5–45) | 1/11 | 28% | 100 × 5/IV 40 × 5 in 21 patients; IV 50 × 5 in 24 patients; IV 20 × 5 × 2 in 6 patients | None |
Xu et al. (Clin Cancer Res, 2023)[ |
II (24) | 16.5 ± 7.9 | 1 CR/4 PR | 20.8% | 100 × 5/50 × 5 | VCR 1.4 mg/m2 day 1 |
Xu et al. (Clin Cancer Res, 2023)[ |
II (22) | 15.2 ± 6.3 | 1 CR/11 PR | 54.5% | 100 × 5/20 × 5 × 2 | VCR 1.4 mg/m2 days 1 and 8 |
Most of these studies were retrospective, not prospective; there are only four prospective trials with well-defined eligibility cohorts and report by intent to treat. In addition, there is significant variability among the reports in doses and dose schedules of irinotecan and temozolomide and the use of additional agents. When combined, these studies accrued 275 patients and observed 21 complete remissions and 82 partial remissions. The objective response rate was 37.5%.
Evidence (chemotherapy):
Local Therapy for Relapsed Disease
Surgery
Treatment with aggressive surgery (such as amputation or hemipelvectomy) may be considered for patients with nonmetastatic locally recurrent disease, even if the prognosis is limited.[
The role of pulmonary metastasectomy in patients with relapsed disease and isolated lung metastases is controversial.[
Radiation therapy
In the relapsed setting, radiation therapy may be used (similar to first-line strategies) for patients who relapsed after the beginning of front-line therapy and/or who present only with pulmonary metastases.[
Palliation of painful lesions in children with recurrent or progressive disease can be achieved using a short course (10 or fewer fractions) of radiation therapy. In a retrospective study of 213 children with various malignancies, who were treated with such short course radiation therapy, 85% of patients had complete or partial pain relief, with low levels of toxicity.[
High-Dose Chemotherapy With Stem Cell Support
Aggressive attempts to control the disease, including myeloablative regimens, have been used, but there is no evidence at this time to conclude that myeloablative therapy is superior to standard chemotherapy.[
Most published reports about the use of high-dose therapy and stem cell support for patients with high-risk Ewing sarcoma have significant flaws in methodology. The most common error is the comparison of this high-risk group with an inappropriate control group. Patients with Ewing sarcoma at high risk of treatment failure who received high-dose therapy are compared with patients who did not receive high-dose therapy. Patients who undergo high-dose therapy must respond to systemic therapy, remain alive and respond to treatment long enough to reach the time at which stem cell therapy can be applied, be free of comorbid toxicity that precludes high-dose therapy, and have an adequate stem cell collection. Patients who undergo high-dose therapy and stem cell support are a highly selected group; comparing this patient group with all patients with high-risk Ewing sarcoma is inappropriate and leads to the erroneous conclusion that this strategy improves outcome.
Surveys of patients who underwent allogeneic hematopoietic stem cell transplant (HSCT) for recurrent Ewing sarcoma did not show improved EFS when compared with patients who underwent autologous HSCT, and allogeneic HSCT was associated with a higher complication rate.[
Other Therapies
Other therapies that have been studied in the treatment of recurrent Ewing sarcoma include the following:
A prospective, randomized, double-blind trial compared regorafenib with placebo in patients with recurrent Ewing sarcoma.[
In addition to this single-agent experience, regorafenib was shown to be tolerable on a sequential schedule with vincristine and irinotecan in 21 pediatric patients. Five patients with relapsed Ewing sarcoma were included as part of this phase I trial. Three of five patients had partial responses with this regimen.[
Sequencing of recurrent and refractory Ewing sarcoma tumors from pediatric (n = 79) and young adult patients (n = 25) enrolled in the National Cancer Institute (NCI)-COG Pediatric Molecular Analysis for Therapeutic Choice (MATCH) trial revealed genomic alterations that were considered actionable for treatment on MATCH study arms in 8 of 104 tumors (7.7%), including EZH2 variants in 2 of 104 tumors (1.9%).[
Treatment Options Under Clinical Evaluation for Recurrent Ewing Sarcoma
Information about NCI-supported clinical trials can be found on the
Current Clinical Trials
Use our
References:
There are undifferentiated small round cell sarcomas of bone and soft tissue that do not have the EWSR1::ETS gene family member translocations and appear to be biologically distinctive from Ewing sarcoma with EWSR1::ETS gene family member translocations. This includes tumors with translocations of the CIC gene or the BCOR gene, as well as tumors with EWSR1 translocations involving non-ETS gene family members. These groups occur much less frequently than Ewing sarcoma, and descriptions of clinical outcomes for these patients are based on smaller sample sizes and less homogeneous treatment; therefore, patient outcomes are hard to quantitate with precision. Most of these tumors have been treated with regimens designed for Ewing sarcoma, and there is consensus that they were often included in past clinical trials for the treatment of Ewing sarcoma, sometimes called translocation-negative Ewing sarcoma. There is agreement that these tumors are sufficiently different from Ewing sarcoma; they should be stratified and analyzed separately from Ewing sarcoma with the common translocations, even if they are treated with similar therapy. The summary of these entities are presented below and follows the categorization of the 2020 World Health Organization (WHO) Classification of Tumours: Soft Tissue and Bone Tumours (5th edition).[
Undifferentiated Small Round Cell Sarcomas WithBCORGenetic Alterations
Clinical presentation
Undifferentiated round cell sarcomas with BCOR::CCNB3 rearrangements account for about 5% of all EWSR1-negative rearranged sarcomas and more commonly affects males. More than 70% of cases occur in patients younger than 18 years (median age at diagnosis, 13–15 years).[
Genomic characteristics
The most common types of undifferentiated small round cell sarcoma with BCOR rearrangements are those with the BCOR::CCNB3 rearrangement.[
BCOR internal tandem duplications (ITD) involving exon 15 are observed in infantile undifferentiated round cell sarcomas and primitive myxoid mesenchymal tumors of infancy (PMMTI).[
BCOR ITD have been reported in 90% of cases of clear cell sarcoma of the kidney, with a smaller subset harboring YWHAE::NUTM2B/E or BCOR::CCNB3 gene fusions.[
The transcriptional profiles induced by BCOR gene fusions, BCOR ITD, and YWHAE::NUTM2B/E fusions appear to be similar to each other and distinctive from that of Ewing sarcoma.[
Treatment of undifferentiated round cell sarcomas withBCORgenetic alterations
When treated with Ewing sarcoma–like therapies, 75% of patients show significant treatment-associated pathological responses. In one series of 36 cases, the 3-year and 5-year survival rates were 93% and 72%, respectively.[
A multi-institution retrospective analysis of patients aged 0 to 24 years identified 29 patients with sarcomas and CIC gene fusions and 25 patients with BCOR-associated sarcomas (18 with BCOR::CCNB3 gene fusions and 7 with BCOR-ITD).[
Undifferentiated Small Round Cell Sarcomas WithCICGenetic Alterations
Clinical presentation
Undifferentiated small round cell sarcomas with CIC::DUX4 rearrangements most commonly affect young adults, with 50% of cases occurring between the ages of 21 and 40 years. In a series of 115 cases, the median age at diagnosis was 32 years, and 22% of cases occurred in patients younger than 18 years.[
Genomic characteristics
CIC-rearranged sarcomas most commonly have a CIC gene fusion with DUX4, resulting from either a t(4;19)(q35;q13) or a t(10;19)(q26;q13) translocation.[
Treatment of undifferentiated small round cell sarcomas withCICgenetic alterations
In a series of 115 cases of CIC-rearranged small round cell sarcomas, 57 patients had adequate follow-up information.[
An international retrospective cohort study further highlighted the poor outcomes for patients with CIC-rearranged sarcomas. The 3-year OS rate was only 39.6%, which was significantly worse than outcomes for patients with other undifferentiated round cell sarcomas.[
In another series of 79 patients with CIC-rearranged round cell sarcomas, outcomes were likewise poor, with a median OS of 18 months.[
A multi-institution retrospective analysis of patients aged 0 to 24 years identified 29 patients with sarcomas and CIC gene fusions and 25 patients with BCOR-associated sarcomas (18 with BCOR::CCNB3 gene fusions and 7 with BCOR-ITD).[
Undifferentiated small round cell sarcomas withCIC::NUTM1rearrangements
Undifferentiated small round cell sarcomas with CIC::NUTM1 rearrangements have been described and occur much less frequently than undifferentiated round cell sarcomas with CIC::DUX4 rearrangements.[
Undifferentiated small round cell sarcomas withATXN1::NUTM2AorATXN1L::NUTM2Afusions
In one report, three children had tumors with ATXN1::NUTM2A or ATXN1L::NUTM2A fusions.[
Undifferentiated Small Round Cell Sarcomas WithEWSR1::non-ETSFusions
Sarcomas withEWSR1::NFATC2andFUS::NFATC2fusions
Sarcomas with EWSR1::NFATC2 and FUS::NFATC2 fusions typically arise in long bones, show a strong male predominance, and are more common in adults than in children.[
EWSR1::NFATC2 and FUS::NFATC2 rearrangements are also observed in a substantial proportion of solitary bone cysts (also known as simple bone cysts), a benign condition that typically presents in the metadiaphyses of the long bones of skeletally immature individuals.[
Sarcomas withEWSR1::PATZ1fusions
Sarcomas with the EWSR1::PATZ1 fusion are very uncommon. In the small number of cases described, there appears to be gender balance, a propensity for presentation at truncal primary sites (particularly the chest), and a median age of presentation of between 40 to 50 years, with cases rarely occurring in the pediatric age range.[
The EWSR1::PATZ1 fusion has been described more commonly in brain tumors. It has been suggested that this fusion may define a novel form of glioblastoma.[
References:
The PDQ cancer information summaries are reviewed regularly and updated as new information becomes available. This section describes the latest changes made to this summary as of the date above.
General Information About Ewing Sarcoma and Undifferentiated Small Round Cell Sarcomas of Bone and Soft Tissue
Added text about the results of one study that retrospectively analyzed a single-institution's experience with visceral Ewing sarcoma. The study focused on surgical management and compared the outcomes of patients with visceral Ewing sarcoma with those of patients with osseous and soft tissue Ewing sarcoma (cited Wallace et al. as reference 30).
Treatment of Localized Ewing Sarcoma
Added text about the results of the European Ewing 2008R1 trial that evaluated 284 patients with standard-risk Ewing sarcoma who received chemotherapy. During the sixth cycle of consolidation, patients were randomly assigned to therapy with either the addition of zoledronic acid for nine 28-day cycles or no maintenance therapy (cited Koch et al. as reference 16).
Added text to state that in a single-institution nonrandomized study, patients who had primary tumors 8 cm or larger were treated with higher-dose radiation therapy. The 5-year cumulative incidence of local failure rate was 6.6%, which compares favorably to other published local failure rates in this group of patients (cited Kacar et al. as reference 27).
Treatment of Recurrent Ewing Sarcoma
Added text about the results of a prospective, randomized, double-blind trial that compared regorafenib with placebo in patients with recurrent Ewing sarcoma (cited Duffaud et al. as reference 60).
Added text to state that in addition to single-agent experience, regorafenib was shown to be tolerable on a sequential schedule with vincristine and irinotecan in 21 pediatric patients. Five patients with relapsed Ewing sarcoma were included as part of this phase I trial. Three of five patients had partial responses with this regimen (cited Casanova et al. as reference 61).
Added cabozantinib as a multitargeted kinase inhibitor that has been studied in the treatment of recurrent Ewing sarcoma. Also added text to state that a real-world evidence analysis included 16 patients with relapsed or refractory Ewing sarcoma who were treated with cabozantinib. Four patients had objective responses (cited Peretz Soroka et al. as reference 63).
This summary is written and maintained by the
Purpose of This Summary
This PDQ cancer information summary for health professionals provides comprehensive, peer-reviewed, evidence-based information about the treatment of childhood Ewing sarcoma and undifferentiated small round cell sarcomas of bone and soft tissue. It is intended as a resource to inform and assist clinicians in the care of their patients. It does not provide formal guidelines or recommendations for making health care decisions.
Reviewers and Updates
This summary is reviewed regularly and updated as necessary by the
Board members review recently published articles each month to determine whether an article should:
Changes to the summaries are made through a consensus process in which Board members evaluate the strength of the evidence in the published articles and determine how the article should be included in the summary.
The lead reviewers for Ewing Sarcoma and Undifferentiated Small Round Cell Sarcomas of Bone and Soft Tissue Treatment are:
Any comments or questions about the summary content should be submitted to Cancer.gov through the NCI website's
Levels of Evidence
Some of the reference citations in this summary are accompanied by a level-of-evidence designation. These designations are intended to help readers assess the strength of the evidence supporting the use of specific interventions or approaches. The PDQ Pediatric Treatment Editorial Board uses a formal evidence ranking system in developing its level-of-evidence designations.
Permission to Use This Summary
PDQ is a registered trademark. Although the content of PDQ documents can be used freely as text, it cannot be identified as an NCI PDQ cancer information summary unless it is presented in its entirety and is regularly updated. However, an author would be permitted to write a sentence such as "NCI's PDQ cancer information summary about breast cancer prevention states the risks succinctly: [include excerpt from the summary]."
The preferred citation for this PDQ summary is:
PDQ® Pediatric Treatment Editorial Board. PDQ Ewing Sarcoma and Undifferentiated Small Round Cell Sarcomas of Bone and Soft Tissue Treatment. Bethesda, MD: National Cancer Institute. Updated <MM/DD/YYYY>. Available at:
Images in this summary are used with permission of the author(s), artist, and/or publisher for use within the PDQ summaries only. Permission to use images outside the context of PDQ information must be obtained from the owner(s) and cannot be granted by the National Cancer Institute. Information about using the illustrations in this summary, along with many other cancer-related images, is available in
Disclaimer
Based on the strength of the available evidence, treatment options may be described as either "standard" or "under clinical evaluation." These classifications should not be used as a basis for insurance reimbursement determinations. More information on insurance coverage is available on Cancer.gov on the
Contact Us
More information about contacting us or receiving help with the Cancer.gov website can be found on our
Last Revised: 2024-04-19
This information does not replace the advice of a doctor. Ignite Healthwise, LLC, disclaims any warranty or liability for your use of this information. Your use of this information means that you agree to the
Healthwise, Healthwise for every health decision, and the Healthwise logo are trademarks of Ignite Healthwise, LLC.
Individual and family medical and dental insurance plans are insured by Cigna Health and Life Insurance Company (CHLIC), Cigna HealthCare of Arizona, Inc., Cigna HealthCare of Illinois, Inc., Cigna HealthCare of Georgia, Inc., Cigna HealthCare of North Carolina, Inc., Cigna HealthCare of South Carolina, Inc., and Cigna HealthCare of Texas, Inc. Group health insurance and health benefit plans are insured or administered by CHLIC, Connecticut General Life Insurance Company (CGLIC), or their affiliates (see
All insurance policies and group benefit plans contain exclusions and limitations. For availability, costs and complete details of coverage, contact a licensed agent or Cigna sales representative. This website is not intended for residents of New Mexico.