ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : -8 مورد

Treatment of Ewing sarcoma

Treatment of Ewing sarcoma
Authors:
Mark C Gebhardt, MD
Steven G DuBois, MD, MS
Elizabeth H Baldini, MD, MPH, FASTRO
Section Editors:
Alberto S Pappo, MD
Robert G Maki, MD, PhD
Raphael E Pollock, MD
Deputy Editor:
Melinda Yushak, MD, MPH
Literature review current through: Apr 2025. | This topic last updated: Mar 17, 2025.

INTRODUCTION — 

Ewing sarcoma (ES) is a rare primary bone malignancy and most often develops in the long bones of the extremities or the flat bones of the pelvis. Less commonly, it arises in soft tissue (extraosseous ES). Multimodality therapy is the standard of care for most patients as this approach has markedly improved long-term survival. (See "Clinical presentation, staging, and prognostic factors of Ewing sarcoma", section on 'Prognostic factors'.)

This topic will discuss the management of ES. The pathology, molecular genetics, clinical presentation, diagnosis, and prognosis are discussed separately.

(See "Epidemiology, pathology, and molecular genetics of Ewing sarcoma".)

(See "Clinical presentation, staging, and prognostic factors of Ewing sarcoma".)

PRETREATMENT EVALUATION — 

The initial management of patients depends on an accurate evaluation and staging to determine if the disease is localized or there are any sites of metastases. This is discussed in more detail separately. (See "Clinical presentation, staging, and prognostic factors of Ewing sarcoma", section on 'Staging evaluation'.)

DEFINITION OF LOCALIZED DISEASE — 

Localized ES refers to disease confined to the site of origin and without evidence of distant metastatic disease.

Metastatic disease refers to disease anywhere other than the primary site and can include involvement of lung, bone, bone marrow, distant lymph nodes, lesions discontinuous from the primary tumor (ie, skip metastases), or other distant sites.

Patients with regional lymph node involvement have generally been included in trials for patients with localized ES.

MULTIMODALITY THERAPY FOR LOCALIZED DISEASE — 

Patients with localized ES are treated with multimodality therapy (ie, chemotherapy plus surgery and/or radiation therapy (algorithm 1)).

Although <25 percent of patients have overt metastases at the time of diagnosis, ES is considered a systemic disease. Patients undergoing local therapy alone have a high relapse rate (80 to 90 percent) suggesting that most patients have subclinical metastatic disease at the time of diagnosis, even in the absence of overt metastases. Chemotherapy can successfully eradicate these deposits, and modern treatment plans all include chemotherapy, usually administered prior to (neoadjuvant) and following (adjuvant) local treatment.

For patients with localized disease, the addition of intensive multiagent chemotherapy to local therapy has had a dramatic impact on survival, and reported five-year survival rates are approximately 70 percent [1-10].

While they constitute a minority of patients, adults and those with extraosseous ES are treated in a similar fashion to other patients with ES except as specifically described below:

Adult patients – Treatment of adults with ES should generally be guided by the same general principles as those used for younger individuals, with therapy modification based upon comorbidities and treatment tolerance. However, few clinical trials address treatment in adults since most studies have excluded older individuals. (See 'Patients 18 years or older' below.)

Less than 5 percent of cases of ES arise in adults over the age of 40. Some studies have defined "older" patients as those over the age of 15 at initial diagnosis, while others have used a cutoff of 18 years. Although some studies suggest that older age is an adverse prognostic factor in ES, it is not clear if the worse prognosis in older individuals is due to biologic differences or differences in treatment regimens (ie, regimens used in pediatric versus those used in adult medical oncology). (See "Clinical presentation, staging, and prognostic factors of Ewing sarcoma", section on 'Age'.)

Extraosseous Ewing sarcoma – Patients with extraosseous ES should be treated with the same protocols used for osseous ES as a similar response to multimodality therapy has been demonstrated [7,11-14].

Neoadjuvant chemotherapy — Neoadjuvant chemotherapy is a standard part of ES treatment, given to reduce local tumor volume, facilitate resection, and treat micrometastatic disease. Reduction in tumor volume may enable limb-sparing procedures for extremity lesions, but it may also be important for rib, chest wall, and vertebral primaries [15-17].

Clinical evidence that chemotherapy is effectively treating the tumor include relief of tumor-related pain, decrease in tumor size, fall in lactate dehydrogenase level, radiologic improvement, and evidence of necrosis in the resected specimen.

Patients <18 years

VDC/IE preferred — For patients under 18 years with localized ES, we recommend interval-compressed neoadjuvant chemotherapy with alternating cycles of vincristine/doxorubicin/cyclophosphamide and ifosfamide/etoposide (VDC/IE) given every 14 days with hematopoietic growth factor support (table 1). Patients are treated for a total of six cycles (12 weeks) prior to local therapy. VDC/IE is preferred over vincristine, ifosfamide, doxorubicin, and etoposide (VIDE) because it improves overall survival (OS), has a shorter duration of therapy, and decreased toxicity [18].

The superiority of VDC/IE over VIDE was established in an open-label phase III trial (EE2012) conducted in Europe [18]. In this study, 640 patients with ES were randomly assigned to initial treatment with VDC/IE administered on an interval-compressed schedule (every 14 days with hematopoietic growth factor support) or the VIDE regimen. At a median follow-up of 47 months, VDC/IE improved event-free survival (EFS) and OS compared with VIDE (three-year EFS 67 versus 61 percent; hazard ratio [HR] 0.71, 95% CI 0.55-0.92; three-year OS 82 versus 74 percent; HR 0.62, 95% CI 0.46-0.85). While grade ≥3 toxicity rates were similar between the two treatment arms (90 versus 91 percent), the risk of grade ≥3 febrile neutropenia was lower for VDC/IE compared with VIDE (58 versus 74 percent). The duration of therapy for VDC/IE was also shorter by approximately two months. Based upon this finding, interval-compressed VDC/IE is a standard regimen.

Combining additional chemotherapy agents with VDC/IE does not give any additional clinical benefit in patients with localized disease. In a randomized phase III Children's Oncology Group (COG) trial (AEWS1031) conducted in 629 patients with treatment-naïve localized ES, the addition of vincristine/topotecan/cyclophosphamide to the interval-compressed VDC/IE regimen did not improve EFS or OS [19].

Interval-compressed VDC/IE — In patients <18 years with localized ES, interval-compressed therapy with VDC/IE given every 14 days with hematopoietic growth factor support (table 1) is preferred to administration every 21 days because it improves EFS and OS without increased toxicity or higher risk of second (subsequent) malignant neoplasms.

A randomized phase III trial (AEWS0031) from the COG conducted in North America established interval-compressed therapy with alternating cycles of VDC/IE as the preferred initial chemotherapy regimen for children with localized ES (table 1) [20-22]. In this study, 587 patients with localized ES were randomly assigned to receive 14 alternating cycles of VDC/IE every 21 or 14 days. Most patients enrolled were age <18 years (88 percent). Interval-compressed VDC/IE given every 14 days improved 10-year EFS (70 versus 61 percent) and 10-year OS (76 versus 69 percent) compared with VDC/IE given at 21-day intervals.

In subgroup analyses, compared with chemotherapy every 21 days, interval-compressed VDC/IE every 14 days also improved 10-year EFS in patients <18 years of age (73 versus 64 percent; HR 0.71, 95% CI 0.52-0.99), and in patients with:

Pelvic primary tumors (67 versus 43 percent)

Tumor volume ≥200 mL (74 versus 46 percent)

Viable tumor at the time of surgery (75 versus 47 percent)

The toxicity of both regimens were similar [21]. There were similar rates in the 10-year cumulative incidence of second (subsequent) malignant neoplasms between treatment every 14 days (3.2 percent) and treatment every 21 days (4.2 percent).

Although interval compression is recommended, there is no role for dose escalation. In a randomized phase III trial conducted by the COG, dose escalation of VDC/IE without hematopoietic cell support did not improve outcomes in patients with newly diagnosed localized disease [23]. Furthermore, concerns for an increased risk of secondary malignancies in patients receiving dose-intense therapy have tempered enthusiasm for this approach.

Limited role for high-dose chemotherapy with HCT — The role of consolidation with high-dose chemotherapy with autologous hematopoietic cell transplantation (HCT; rescue) is uncertain and should be reserved for patients enrolled on clinical trials.

Although this approach improved outcomes for children with high-risk localized disease who were treated with the VIDE induction regimen [23], high-dose chemotherapy with autologous HCT has not been used with interval-compressed VDC/IE chemotherapy [24]. Additionally, in a phase III trial (EE2012), interval-compressed VDC/IE improved OS over VIDE among the subgroup of patients with high-risk localized disease (tumor volume ≥200 mL at diagnosis) [18]. It is not known whether high-dose chemotherapy with autologous HCT would lead to further improvements in this population. (See 'Interval-compressed VDC/IE' above.)

The role of consolidative high-dose chemotherapy followed by autologous HCT for localized high-risk disease was evaluated in the European Ewing Tumour Working Initiative of National Groups (Euro-EWING) 99/EWING 2008 trial [23]. High-risk disease was defined as a poor histologic response after receiving six cycles of VIDE (76 percent of the cohort) or tumor volume at diagnosis ≥200 mL if unresected, initially resected, or resected after radiation therapy (RT). In this study, 240 patients with high-risk localized ES were treated with six courses of VIDE plus one course of consolidation vincristine, dactinomycin, and ifosfamide (VAI). Patients were then randomly assigned to either one course of busulfan plus melphalan followed by autologous HCT or seven courses of standard chemotherapy with VAI. At a median follow-up of 7.8 years, high-dose chemotherapy followed by HCT improved both EFS (eight-year EFS 61 versus 47 percent) and OS (eight-year OS 65 versus 56 percent).

Severe acute toxicities were more common in the high-dose chemotherapy group, and three patients died in this group, two of treatment-related toxicity; the third patient did not receive dose-intense chemotherapy or high-dose chemotherapy because of kidney impairment. The risk of secondary malignancies in long-term survivors was not reported.

Patients 18 years or older — For patients ≥18 years with localized ES, we recommend neoadjuvant chemotherapy with alternating cycles of VDC/IE rather than VIDE.

Support for the use of VDC/IE in this population is largely based on extrapolation of data from trials conducted primarily in patients <18 years and described separately (see 'Patients <18 years' above). Compared with VIDE, VDC/IE improves OS, has a shorter duration of therapy, and decreased toxicity. Many of these trials included adults, but adults accounted for a minority of the population.

In patients ≥18 years with localized ES, VDC/IE can be administered as either:

VDC/IE every 14 days with hematopoietic growth factor support for a total of six cycles (12 weeks of therapy prior to local control), with adjuvant therapy after local control.

VDC/IE every 21 days with hematopoietic growth factor support for a total of four cycles (12 weeks of therapy prior to local control), with adjuvant therapy after local control.

As discussed above, every-14-day therapy is preferred over every-21-day therapy in younger patients based on the AEWS0031 trial that demonstrated improved EFS and OS in localized ES [20-22]. However, the benefit of every-14-day therapy is less clear in older patients and may be more challenging to administer. Even in the pediatric cohort, the mean cycle duration given was 17 days instead of the goal of 14 days [21]. In addition, while EFS was numerically higher with every-14-day in 31 patients >18 years (10-year EFS of 53 versus 37 percent; HR 0.75, 95% CI 0.38-1.47), it did not reach statistical significance [22]. Similarly, in the EE2012 trial, the effect of interval-compressed VDC/IE was less pronounced in those ≥14 years [18]. While a 14-day cycle may be a laudable goal for treatment of ES in an adult population, the choice of cycle length is made based on patient specific factors including patient age, comorbidities, and the practicalities of the tolerance of treatment.

Local treatment — Following neoadjuvant systemic therapy, patients receive local treatment with surgery, RT, or both. Modern treatment protocols emphasize surgery for optimal local control. However, the choice among these options should be made by a multidisciplinary team and is based on patient characteristics, location of disease, expected toxicity, functional outcomes, potential complications of local treatment, and patient preference.

For most patients, chemotherapy is combined with local therapy that uses one modality (RT or surgery); the combination of all three modalities (RT, chemotherapy, and surgery) is not routinely recommended. However, in a review of local treatment modalities used on the Ewing 2008 study, patients with a poor response to chemotherapy appeared to benefit from the combination of surgery and radiation over surgery alone (EFS HR 0.49, 95% CI 0.27-0.89) [25]. By contrast, the overall group had no difference in EFS, OS, or local relapse amongst the surgery group compared with the surgery and radiation group.

Because no randomized trial has directly compared surgery versus RT, only a relative comparison can be made from retrospective reports and the prospective trials that have mainly tested different multiagent chemotherapy regimens. In many retrospective series, rates of local control and survival are superior after surgery compared with RT alone [2,5,12,26-40]. However, selection bias likely accounts for at least some of these results. Smaller, more favorably situated peripheral tumors are more often resected while larger, axial lesions (which have a higher rate of local failure in most series and a poorer prognosis overall [2,5,26,41]) are more likely to be referred for RT. By contrast, some series suggest local control rates with RT and surgery are similar when controlling for age and primary tumor site [42-44]. (See "Clinical presentation, staging, and prognostic factors of Ewing sarcoma", section on 'Tumor site and size'.)

Timing of local control — Local treatment (surgery, RT, or both) should be initiated immediately after completion of 12 weeks of induction chemotherapy. An analysis of data derived from the National Cancer Database noted that patients initiating local therapy by week 15 versus week ≥16 had improved five-year OS (78.7 versus 70.4 percent) and 10-year OS (70.3 versus 57.1 percent) [45]. On subgroup analysis, the difference in OS according to time to local therapy was particularly large in patients receiving RT alone (five-year OS 80.0 versus 58.9 percent). (See 'Multimodality therapy for localized disease' above.)

Resectable disease

Surgery preferred — Surgery is the preferred modality for tumors that can be resected with adequate margins, and acceptable morbidity and functional outcome. The determination about resectability of a lesion is often made by a multidisciplinary team. Tumor location, size, and impact of adjacent structures all influence the decision about whether a tumor should be resected. Tumors arising in dispensable bones (eg, fibula, rib, small lesions of the hands or feet) are generally able to be resected with minimal morbidity, whereas tumors of the long bones of the leg, distal humerus, or ulna are typically resected using intercalary techniques (allografts, autografts, or metallic prostheses) or joint replacement. In addition, ES is a chemotherapy-sensitive disease and lesions that may initially appear unresectable may be amenable for resection after neoadjuvant chemotherapy. (See "Bone sarcomas: Preoperative evaluation, histologic classification, and principles of surgical management".)

Modern surgical techniques offer the opportunity for limb reconstruction with endoprostheses that can preserve function and minimize disability. Patient selection is evolving as surgical morbidity has decreased with advances in reconstruction and prosthesis options (eg, 3D printed, custom prostheses) [46-48]. For larger, bulky tumors that are resectable or borderline resectable, sometimes preoperative RT is offered to help facilitate the resection. The surgical principles that apply to resection of the primary tumor and reconstruction are similar to those in patients with osteosarcoma and are discussed elsewhere. (See "Bone sarcomas: Preoperative evaluation, histologic classification, and principles of surgical management".)

Support for surgery as the preferred modality is based on the following:

It avoids the risk of secondary radiation-associated sarcomas.

It allows for analysis of tumor necrosis in the excised tumor to refine prognosis estimates.

It avoids the risk of impacting bone growth and resulting deformity in the skeletally immature child.

The COG reported improved rates of local control in patients treated with surgery. This study analyzed local control outcomes in patients treated with surgery, radiation, or both on three consecutive protocols using standard-dose, five-drug chemotherapy every 21 days [38,49]. Local failure rates were lower among patients treated with surgery alone or surgery plus RT when compared with those treated with RT alone (3.9 and 6.6 versus 15.3 percent, respectively). On multivariate analysis, compared with surgery, use of RT was associated with a higher risk for local failure (HR 2.41, 95% CI 1.24-4.68), although there were no significant differences detected in EFS (HR 1.42, 95% CI 0.94-2.14), OS (HR 1.37, 95% CI 0.83-2.26), or distant failure (HR 1.13, 95% CI 0.70-1.84). As to be expected, patients who underwent surgery were younger and had more appendicular primary tumors. The local failure incidence was higher for pelvic tumors (13.2 percent) than for other sites (9.1 percent for extraskeletal tumors, 5.4 percent for extremity tumors, 5.3 percent for axial non-spine tumors, and 3.6 percent for spine tumors).

Similarly, a secondary analysis of the Ewing 2008 trial found that the adjusted hazards for any event (a composite endpoint of EFS, OS, and local recurrence) was increased for those treated with RT alone compared with surgery (HR 1.53, 95% CI 1.02-2.31) [25]. The analysis included 863 patients who received surgery alone, surgery and radiation, or radiation alone while receiving protocol-specified chemotherapy. When compared with other locations, pelvic and spine primaries were more frequently treated with RT as the primary local treatment modality (40 and 31 percent). This likely reflected the inability to resect tumors in these locations.

Although a benefit for intraoperative RT (IORT) has been suggested in retrospective series involving a small number of patients [50,51], this approach is not commonly used. Peripheral nerves are dose-limiting structures for IORT, so the decision to use IORT must take into account the risk of severe neuropathy and soft tissue necrosis.

Bulky tumors in difficult sites — For bulky tumors in difficult sites such as the pelvis, decisions about resectability and using RT are made by a multidisciplinary team.

For lesions that are felt to have a high likelihood of positive margins, definitive RT is often pursued. However, combined surgery and RT might allow for a more limited surgical procedure, better functional outcome, and enhanced local control as compared with single-modality therapy [26,52-54]. Preoperative RT is considered when it is likely that margins will be positive. This may shrink the tumor and allow for resection. However, it can also adversely impact the soft tissues making dissection more challenging and cause a greater likelihood of wound complications. Postoperative radiation is also an option, but can be challenging to identify where to direct RT if there is a large field and uncertainty exactly where the positive margin was located.

Unresectable disease — RT is preferred over surgery when the tumor is unresectable or if a resection will result in significant morbidity. The following are examples of locations that are typically treated with RT instead of surgery:

Primary tumors of the spine [55].

Tumors of the skull or facial bones because of the difficulty in achieving negative margins without substantial functional deficit [56].

Tumors in locations such as the periacetabular region of the pelvis or the sacroiliac joint because of a loss of critical function from surgical procedures that involve resecting the acetabulum or resecting sacral nerve roots. However, the treatment of pelvic tumors is controversial. Lesions of the iliac wing, ischium, or pubis can usually be resected after a good response to chemotherapy without substantial functional morbidity [28,52,57-59]. (See "Bone sarcomas: Preoperative evaluation, histologic classification, and principles of surgical management".)

Support for the use of RT comes from case series or secondary analyses of trials that were prospectively evaluating the use of chemotherapy. This approach has limitations as many patients who were referred for RT had lesions that were difficult to resect. The results are summarized as below:

No difference in local failure or EFS rates were shown in a subgroup of 75 patients with nonmetastatic pelvic ES treated with surgery, RT, or both [60]. The Intergroup study 0091, a randomized comparison of vincristine, doxorubicin, cyclophosphamide, and dactinomycin with or without alternating IE (ifosfamide plus etoposide), allowed treating clinicians to choose the local control modality. Five-year EFS was 42 percent in patients who received surgery, 52 percent for those receiving RT, and 47 percent in patients who received both treatment modalities.

However, the outcomes for surgery versus RT may depend on site of disease. In a COG analysis of patients on trials comparing chemotherapy protocols who were treated with surgery or RT, local failure rates with surgery were numerically lower but did not reach statistical significance for the subgroup of patients with axial nonspine (2.5 versus 10.6 percent), spine (0 versus 5.6 percent), and extraskeletal tumors (7.7 versus 10.9 percent) [49]. However, this review, which included 956 patients treated with ifosfamide- and etoposide-based chemotherapy on three consecutive protocols, did show lower rates of local failure in patients with extremity and pelvic tumors treated with surgery compared with those treated with RT. Local failure rates did not differ based on location for surgically treated patients, but did for patients treated with RT.

Adults with ES who are treated with modern chemotherapy protocols and RT techniques have outcomes that are similar to those of younger patients, with anticipated local control rates of approximately 80 percent after definitive RT [61-63]. With a median follow-up of 41 months, disease-free survival and OS rates for patients with nonmetastatic disease at presentation were 70 and 86 percent, respectively. [61] There were nine local recurrences, five of which developed in patients who presented with overt metastases. The three-year actuarial local control rates for the entire group and those undergoing definitive RT alone were 77 and 78 percent, respectively. The presence of overt metastases emerged as the only significant adverse prognostic factor for local control, a finding that has been confirmed by others [26,64]. Long-term treatment-related complications were minimal, and there were no secondary malignancies.

In patients with pelvic primaries treated on the Euro-EWING99 trial, an analysis of local control methods found no association with OS at five years in patients treated with definitive radiation versus surgery and radiation (73 versus 78 percent) [65].

Suboptimal RT delivery techniques with inadequate tumor coverage may have contributed to poorer outcomes in early series. As an example, in the Cooperative Ewing Sarcoma Study (CESS)-81 trial, relapse-free survival (RFS) for patients undergoing irradiation of the primary site in the initial phase of the trial was only 55 percent, significantly lower than for surgically treated patients [30]. On review, the high rate of local failure after RT alone (50 percent) was attributed to geographic "miss." After a quality assurance program was initiated, local control rates improved substantially and RFS increased to 80 percent. In a subsequent study, CESS-86, which included central quality assurance, the five-year RFS rates following chemotherapy plus either RT or surgery were 67 and 65 percent, respectively [66].

Treatment volume — Modern treatment protocols for ES include tailored RT fields that target the site of the primary lesion with a margin rather than the whole bone [67]. While whole bone RT had been used in past, a randomized trial of whole bone versus tailored-field RT after 12 weeks of induction chemotherapy was stopped early for benefit after demonstrating that a tailored field was as effective as whole bone RT [6]. At three years, the rates of local control and EFS were 76 and 54 percent, respectively. Additional studies have shown the benefit of reducing the irradiated field and targeting higher doses to the site of the initial primary tumor [30,64].Tailored RT is administered to an initial clinical target volume that includes the original bone and soft tissue tumor extent with a 1 cm margin. For patients with chest wall tumors and malignant effusions, the field includes the entire ipsilateral hemithorax to a dose of 15 Gy prior to cone-down to the chest wall tumor. For patients where the soft tissue tumor displaces but is not thought to violate an anatomic compartment (ie, pelvic ES displacing bowel or bladder), the initial target volume includes the post-chemotherapy soft tissue extent of disease abutting the anatomic compartment, rather than the original prechemotherapy extent of disease.

The initial clinical target volume is treated to 45 Gy in 25 fractions followed by a field reduction to encompass the postchemotherapy gross soft tissue tumor as well as all of the originally involved bone with a margin of 1 cm; this volume is treated to another 10.8 Gy in six fractions in patients undergoing definitive RT and to 5.4 Gy in three fractions for patients undergoing adjuvant RT for microscopic residual disease after surgery (with boost up to 10.8 Gy for any sites of gross disease after surgery). Additional margin expansions on these volumes are used for the planning target volume expansion to account for any variability in daily setup; this margin should be individualized by the treating clinician based on the reproducibility of immobilization and the kind of image guidance used but is generally a minimum of 5 mm and usually in the range of 0.5 to 1 cm.

Attention to potential RT effects on normal tissue is critical in radiation planning to minimize late effects, particularly in children:

Treatment of uninvolved epiphyseal growth plates is avoided to minimize treatment-induced limb shortening [68].

Circumferential irradiation of a limb is avoided to reduce the risk of limb edema and fibrosis.

Gonadal avoidance or additional shielding (for the testes) is important to retain fertility.

Excellent results can be achieved from RT of lesions of the hand or foot [67]. However, walking places repetitive stress on the soft tissues of the sole of the feet, and dose sparing for the sole of the foot can improve the functional outcome of treatment.

Irradiation of the Achilles tendon is usually avoided. Lesions of the calcaneus can be treated; although some atrophy of the heel pad has been reported, the functional impairment is usually minor [69].

Nail beds should be excluded from the radiated field when possible.

For pelvic tumors, distention of the bladder prior to each day's treatment can reduce the amount of small bowel in the radiated field, but care must be taken to avoid radiation to significant portions of the bladder in patients receiving cyclophosphamide or ifosfamide to reduce the risk of hemorrhagic cystitis.

Radiation modality — RT for ES can be delivered using three-dimensional conformal radiation therapy (3D-CRT), intensity modulated radiation therapy (IMRT), or proton therapy. The choice of which modality to use is determined by location of the lesion, potential long-term impact of treatment, and availability of different modalities. It is anticipated that the improvements in RT techniques will reduce the risk of long-term complications while maximizing local control rates.

IMRT versus 3D-CRT – IMRT is typically preferred over 3D-CRT. IMRT utilizes variable, computer-controlled intensities within each RT beam, in contrast to the uniform doses within each 3D-CRT beam. Accordingly, compared with 3D-CRT, IMRT typically achieves better dose conformity to the planned target and better sparing of normal tissue, thereby providing high rates of local control and a lower incidence of some late complications. [53]. IMRT is especially useful when target volumes have complex shapes or concave regions. (See "Radiation therapy techniques in cancer treatment", section on 'Intensity-modulated radiation therapy'.)

The tradeoff of improved dose conformity with IMRT is the delivery of low-moderate doses of radiation to a larger volume of surrounding tissue than with 3D-CRT techniques. In addition, patients receiving IMRT may also receive a larger total-body dose because of leakage of radiation [70]. Because of this, there is a theoretical concern for a higher potential risk of second malignancy with IMRT, but clinical studies have not shown this to be true. One observational study of adult and pediatric patients treated for nine tumor types reported that second malignancy rates for IMRT were comparable with those with 3D conformal photons but were reduced with protons (relative risk 0.31, 95% CI 0.26-0.36) [71]. These questions will be further clarified by additional follow-up of patients treated with IMRT.

Proton beam therapy — Compared with photon beam irradiation, proton beam therapy permits the delivery of high doses of RT to the target volume while reducing the radiation dose received by normal tissues distal to the target. Depending on the tumor target configuration and the portal selection, this can result in up to 60 percent reduction in the dose received by uninvolved normal tissue compared with other radiation methods [71]. The ability of proton beam irradiation to minimize exit dose, renders it useful for tumors located close to sensitive tissues. For example, it may be particularly beneficial for ES arising in the bones of the paranasal sinus (image 1 and image 2), the pelvis, and the spine and for those requiring higher doses. (See "Radiation therapy techniques in cancer treatment", section on 'Particle therapy'.)

Proton beam therapy has demonstrated better sparing of the intestine, rectum, bladder, pelvic bone marrow, and femoral head as compared with conventional photon irradiation for pelvic tumors [72]. In a retrospective review of 30 children treated with proton therapy for ES with a median follow-up of 38 months, three-year actuarial rates of EFS, local control, and OS were 60, 86, and 89 percent, respectively [73]. Proton beam therapy was well tolerated with only mild to moderate skin reactions. There were four cases of late hematologic malignancies, but these are known risks of chemotherapy used for ES.

Since proton beam therapy has a lower volume of normal tissue in the irradiated field, it may have a lower risk of secondary in field malignancies [74,75]. A review from the National Cancer Database of pediatric and adult patients treated for nine tumor types suggested a reduction in radiation-associated second cancers with protons compared with photons (relative risk of 0.31, 95% CI 0.26-0.36) [71]. While there is still concern about late radiation effects including neutron scatter radiation, modern treatment protocols have decreased this risk [70,76,77]. Further study is needed to confirm these results [78-80].

Dose — For patients treated with chemotherapy and RT, cooperative studies in the United States have employed 45 Gy in 25 fractions to the initial clinical target volume as defined above, followed by a 10.8 Gy boost in six fractions to the site of original bony disease and any residual soft tissue disease that remains following chemotherapy. Because of the proximity of the spinal cord, the dose to vertebral body primaries has been often limited to 45 Gy, although techniques such as proton beam therapy, IMRT, and stereotactic body RT can permit the safe delivery of higher doses [81].

RT dose is an important factor in local control, particularly for large tumors [64,82]. While 45 Gy is typically used, lower doses (eg, 30 to 36 Gy) have also been studied [82,83]. In one early report of patients undergoing definitive RT, local control rates were 90 percent using RT doses of 35 Gy for smaller lesions (≤8 cm) with a favorable response to chemotherapy, but they were inferior (52 percent) for larger lesions [82].

Dose-escalation RT to limited fields may be beneficial for high-risk tumors and may reduce local recurrence rates, although this approach is considered investigational. A single-institutional phase II trial employed focal limited-margin RT using conformal or intensity-modulated techniques [84]. Unresected tumors <8 cm at diagnosis received a standard dose of 55.8 Gy, and tumors >8 cm received an escalated dose to 64.8 Gy. Patients with microscopic residual disease after resection received adjuvant RT to 50.4 Gy. Adjuvant brachytherapy was permitted in selected patients. Forty-five patients were enrolled, 26 with localized and 19 with metastatic disease. Seventeen patients received adjuvant RT, 16 received standard-dose, and 12 received escalated-dose RT. Failures included 1 local, 10 distant, and 1 combined local/distant. The estimated 10-year cumulative incidence of local failure was 4.4 ± 3.1 percent, with no statistical difference seen between RT treatment groups; there were no local failures in the escalated-dose RT treatment group.

By contrast, a randomized phase III trial of 95 patients with localized ES demonstrated improved five-year local control in the dose-escalation arm compared with standard dose RT (76 versus 49 percent) [85]. However, the OS did not achieve statistical significance at five years despite a numerically higher value (59 versus 45 percent). There was a higher incidence of skin toxicity in the dose-escalation arm compared with standard RT (10 versus 2 percent). Similarly, a retrospective study of 32 patients with newly diagnosed Ewing sarcoma ≥8 cm treated with dose escalation (59 to 69 Gy) was associated with high rates of local control (five-year local failure rate of 6.6 percent) [86].

This approach, if validated, has particular appeal because of the reduction in radiation target volume, which should reduce both acute and late toxicity, as well as the selective use of dose escalation for high-risk lesions >8 cm, reported in multiple series to be at higher risk for local failure with conventional RT doses. Another report also suggests that patients treated with definitive radiation doses ≥56 Gy had a lower incidence of local recurrence (17 versus 28 percent) [87].

Radiation schedule — Conventional RT schedules consist of once daily RT doses of 1.8 Gy per fraction and this approach is the standard of care for ES. Accelerated fractionation RT does not seem to improve rates of local control or survival for ES [29,30,53,66,88].

A case series of 75 patients with ES suggested limited field sizes with hyperfractionated RT (1.2 Gy twice daily with a six hour interfraction interval) could minimize long-term complications and provide superior functional outcomes [29,89]. However, treatment with a twice daily approach is not standard and should only be considered as part of a clinical trial.

Adjuvant therapy — Since most treatment failures are attributable to systemic metastatic disease, local therapy considerations should be planned to avoid complications that might compromise the administration of systemic therapy. After patients have healed from surgery, we restart systemic therapy as soon as possible. For patients treated with radiation rather than surgery, chemotherapy is given concurrently with radiotherapy, with the exception of doxorubicin which is held during radiotherapy.

Adjuvant chemotherapy — For patients receiving VDC/IE, we typically administer a total of 14 to 17 cycles which includes those cycles received as neoadjuvant therapy. A total of either 14 or 17 cycles is acceptable as both regimens have been used in large trials and both have shown favorable outcomes [19,21]. However, they have not been directly compared. A decision about the total number of cycles must take into account patient specific factors such as tolerability of treatment and ability to complete the total number of cycles (algorithm 1).

The routine administration of intensive multiagent chemotherapy, which can eradicate metastatic deposits, has had a dramatic impact on outcomes. For ES, from 1975 to 2010, the five-year survival rate increased from 59 to 78 percent for children younger than 15 years and from 20 to 60 percent for adolescents aged 15 to 19 years [90,91].

When to use adjuvant radiation therapy — For patients with residual disease after surgery defined as either positive margins or gross residual disease, we suggest adjuvant RT to improve local control. Adjuvant RT reduces the risk of local failure, although effective chemotherapy can also reduce this risk [3,9]. By contrast, adjuvant RT is not usually offered to patients without residual disease (ie, resected with negative margins) to avoid exposing them to the risk of a radiation-induced malignancy.

Indications for adjuvant RT include:

Residual microscopic or gross disease after surgery, or inadequate surgical margins (ie, a marginal or intralesional resection (table 2)).

Adjuvant hemithorax irradiation is indicated in patients with high-risk chest wall primary tumors (eg, with cytology-positive initial pleural effusion, pleural infiltration, and intraoperative contamination of the pleural space).

Positive or inadequate margins or gross residual disease — For patients with positive margins or gross residual disease following surgery, we suggest adjuvant RT. Patients who are left with macroscopic amounts of viable tumor in the resected specimen following neoadjuvant chemotherapy have worse survival than those with minimal or no residual tumor [2,27,42,92-96]. In addition, inadequate resection margins (ie, a marginal or intralesional resection (table 2)) are associated with a worse outcome as compared with radical or wide resection [34,97].

For patients undergoing adjuvant postoperative RT, the dose depends on the amount of residual tumor with doses of 50.4 Gy administered for microscopic disease (positive margins), and 55.8 Gy for gross residual disease. Although some studies suggest that doses as low as 30 Gy may be sufficient in the adjuvant setting [83], further studies are needed before lower doses of RT can be accepted as standard practice.

The Euro-EWING group reported a large observational study of 599 patients who underwent surgery after chemotherapy for standard-risk localized disease [98]. Postoperative RT was recommended for patients with inadequate surgical margins, and it was also common practice for patients with a vertebral tumor or with a pleural effusion contiguous to a rib or thoracic wall primary tumor. After controlling for possible confounders, there was a reduction in local recurrence in patients treated with surgery plus postoperative RT compared with surgery alone (HR 0.43, 95% CI 0.21-0.88). They also observed a nonsignificant trend for benefit associated with postoperative RT for disease-free survival, EFS, and OS. Among the 132 patients for whom RT would have been recommended according to the policies of the Euro-EWING group, 50 did not receive it. Among these patients, local recurrence rates were 30 percent at eight years compared with 11 percent when RT was performed.

On the other hand, a lack of benefit for adjuvant RT was suggested in a single-institutional retrospective series of 512 patients, in which the addition of moderate-dose adjuvant RT (45 Gy) to patients with inadequate margins did not seem to improve local control or overall disease-free survival [99]. The RT dose used in this series is on the low end of the range currently used for microscopic residual disease, and lower than that used for macroscopic residual disease and may have contributed to the lack of difference seen.

Prophylactic bilateral whole-lung irradiation — The role of adjuvant bilateral whole-lung irradiation is unsettled and we do not recommend it as prophylactic treatment in the setting of localized disease. The IESS-I trial showed that bilateral whole-lung irradiation was an effective adjuvant treatment for patients with localized ES, but prolonged follow-up favored four-drug multiagent chemotherapy [4]. Prophylactic whole-lung irradiation has not been studied in any subsequent trial of localized disease, and no studies have used it in addition to modern combination chemotherapy regimens [99], except in patients with lung metastases at diagnosis.

TREATMENT FOR METASTATIC DISEASE — 

Patients with overt metastatic disease at presentation have a worse outcome than those with localized disease. However, aggressive multimodality therapy can relieve pain and delay progression.

In a review of 13 different series in which patients with metastatic ES were predominantly treated with chemotherapy, five-year event-free survival (EFS) and overall survival (OS) rates averaged 25 (range 9 to 55) and 33 (range 14 to 61) percent, respectively [1,5,100-110]. The small numbers of patients in each series and the heterogeneity in location and extent of metastatic disease probably account for these wide variations in outcome. (See "Clinical presentation, staging, and prognostic factors of Ewing sarcoma", section on 'Disease extent'.)

A subset of patients with advanced disease may be cured by multimodality therapy, although the long-term survival rates are clearly lower than for patients with localized disease [111]. Because it can be difficult to predict which patients with metastatic disease will be long-term relapse-free survivors [101], treatment for select patients with potentially favorable metastatic disease should be administered with curative intent. Clinicians experienced in the treatment of ES must direct multimodality therapy and coordination between specialists [5,30,82]. The site(s) of metastatic disease is an important variable. For children with isolated lung and pleural metastases, five-year EFS rates up to 50 percent are reported with multimodality therapy; for metastases involving bone or bone marrow, EFS rates fall to 10 to 20 percent; and, for combined sites, to less than 15 percent [108].

Initial selection of chemotherapy

VDC/IE for metastatic disease — Patients with disseminated disease at diagnosis often respond well to the same type of systemic chemotherapy as is used for localized disease. Randomized trials are limited in this population as it accounts for only 25 to 30 percent of patients with ES. Whenever possible, patients with newly diagnosed metastatic ES should be offered enrollment in clinical trials evaluating novel approaches. In the absence of a clinical trial, regimens similar to those used for the treatment of patients with newly diagnosed localized disease are commonly used. (See 'Neoadjuvant chemotherapy' above.)

For most patients with metastatic ES, we suggest alternating cycles of vincristine/doxorubicin/cyclophosphamide and ifosfamide/etoposide (VDC/IE) based on extrapolation of data from trials in localized disease. In contrast to the experience for patients with localized disease, studies in patients with metastatic disease have not demonstrated a specific benefit for the addition of I/E to the VDC backbone at the time of diagnosis [103,104,112-115]. As such, some centers utilize VDC for patients with unequivocal evidence of metastatic disease. Given robust improvement in survival with IE in the localized setting, patients with equivocal findings should be treated with VDC/IE.

Several trials have included interval-compressed VDC/IE administered every 14 days in the context of metastatic ES [18,116]. As an example, the COG AEWS1221 trial evaluated the addition of ganitumab prescribed interval-compressed VDC/IE for all patients, with outcomes similar to the historic experience [116]. Similarly, the EE2012 trial described above included patients with localized or metastatic disease and showed a benefit for interval-compressed VDC/IE compared with vincristine, ifosfamide, doxorubicin, and etoposide (VIDE) chemotherapy [18]. The interval-compressed VDC/IE regimen is frequently used in this context as it does not appear to be more toxic and patients complete therapy more quickly. However, given the lack of randomized data comparing every-14-day versus every-21-day chemotherapy in this population, an acceptable alternative is for administration of chemotherapy every 21 days. (See 'Interval-compressed VDC/IE' above.)

By contrast, data from a nonrandomized trial of dose intensification of VDC/IE with augmented alkylator doses did not suggest a benefit in patients with newly diagnosed metastatic ES [117].

Limited role for high-dose chemotherapy in metastatic disease — High-dose chemotherapy with hematopoietic cell transplantation (HCT; rescue) does not have a significant role in the treatment of patients who present with metastatic disease, and we do not offer this approach. In randomized studies, the addition of high-dose chemotherapy with HCT did not significantly improve EFS or OS and was associated with significant toxicity [118-120].

High-dose chemotherapy with HCT was evaluated in a randomized clinical trial (Euro-EWING 99 and EWING 2008) [118]. In this trial, 287 patients with isolated pulmonary (lung or pleural) metastatic disease received six cycles of VIDE and one cycle of vincristine, dactinomycin, and ifosfamide (VAI). Subsequently, patients were randomly assigned to receive either one course of busulfan plus melphalan high-dose chemotherapy followed by autologous HCT, or seven cycles of conventional chemotherapy with VAI followed by whole-lung irradiation. At a median follow-up of approximately eight years, there was no statistically significant difference in EFS between the two groups (eight-year EFS 53 versus 43 percent; hazard ratio [HR] 0.79, 95% CI 0.56-1.1), and OS results were comparable (eight-year OS 55 versus 54 percent; HR 1, 95% CI 0.7-1.44). Additionally, rates of infection as well as gastrointestinal and liver toxicities were higher in the high-dose chemotherapy group and included four deaths versus no deaths in the conventional chemotherapy group.

Similar results were demonstrated in a separate open-label phase III trial (Ewing 2008R3) in which 109 patients with metastatic ES (excluding those with pulmonary metastases only) were treated with six cycles of VIDE and consolidation therapy with eight cycles of vincristine, actinomycin D, and cyclophosphamide [120]. Patients were then randomly assigned to either treosulfan plus melphalan high-dose chemotherapy followed by HCT or no further treatment. At median follow-up of 3.3 years, the addition of high-dose chemotherapy with HCT did not improve three-year EFS (21 versus 19 percent; HR 0.85, 95% CI 0.55-1.32). However, in a posthoc analysis of the subset of 41 patients less than 14 years of age, a potential EFS benefit for this approach was demonstrated (three-year EFS 39 versus 9 percent; HR 0.4, 95% CI 0.19-0.87). Given the limited sample size and lack of preplanned analysis for this subgroup, these results should be interpreted with caution.

Studies evaluating HCT in patients with localized disease are presented separately. (See 'Limited role for high-dose chemotherapy with HCT' above.)

Local control in patients with metastatic disease — Outcomes are best when chemotherapy is combined with optimal local therapy, including radiation and sometimes resection of sites of gross metastatic disease [109,121-123].

Local control of the primary tumor — Most patients benefit from surgery and/or radiation therapy for local control of the primary tumor. Management of the primary site in the setting of metastatic disease requires multidisciplinary input. Local treatment of the primary lesion is important to prevent complications such as fracture, pain, or wound development. In addition, combining local treatment with chemotherapy can increase overall response rates, which will hopefully translate into improved long-term outcomes. However, long-term relapse-free survival is still only achieved in the minority of patients [101].

The choice between surgery versus radiation therapy (RT) for local control is patient specific. As an example, we typically offer surgical resection of the primary to patients who have experienced a significant volume reduction in the primary site after chemotherapy and only have small-volume metastatic disease that is amenable to either resection or RT. By contrast, the presence of diffuse metastatic disease can make it difficult to justify a large resection, which would necessitate a lengthy period off systemic chemotherapy [52,101]. As such, RT is typically used to treat the primary tumor in this setting and usually provides adequate local control with acceptable morbidity [5,30,66,82]. However, if substantial amounts of bone marrow will need to be included in the radiation treatment volume and/or the patient's performance status is poor such that the potential risks of RT outweigh the potential benefits, RT to the primary site may be deferred.

Isolated pulmonary metastases — For patients with pulmonary metastases, we offer a multimodality approach that includes chemotherapy and supplemental low-dose whole-lung irradiation. Whole-lung radiation is typically administered after completion of planned systemic therapy. Surgical resection is reserved for lung metastases that do not resolve with chemotherapy [124]. Patients with lung metastases have a better prognosis than patients with metastatic disease at other sites (ie, bone or bone marrow). Although complete resection may be possible, it is not sufficient therapy on its own; chemotherapy is a necessary component of therapy, and five-year survival between 20 and 40 percent can be achieved with multimodality therapy [108,125,126].

Retrospective reports from large cooperative groups and single institution series suggest that low-dose bilateral whole-lung irradiation benefits patients with the ES family of tumors presenting with pulmonary metastases, even if all lesions are resected or completely respond to chemotherapy [3,30,104,108,109,122,126-129]. In the Cooperative Ewing Sarcoma Study (CESS) and European Intergroup Cooperative Ewing Sarcoma Study (EICESS) trials, the addition of lung RT reduced the rate of pulmonary relapse and improved EFS (from 19 to 40 percent in EICESS) [104,126]. However, most of the available data supporting benefit from RT in this setting are from nonrandomized series, and patient selection factors confound interpretation of the data. There are no randomized trials that confirm the benefit of this approach.

Despite the lack of controlled trials, low-dose bilateral lung irradiation (15 to 18 Gy, in daily 1.5 Gy fractions) with a focal boost dose to a total of 40 to 50 Gy to large deposits is typically administered for patients with pulmonary metastases who have had a good response to chemotherapy. Recommended doses are 15 Gy in 10 fractions for patients <14 years old and 18 Gy in 12 fractions for those >14 years old [18].In the rare event of a child <7 years, we would give 12 Gy in 8 fractions. Long-term toxicity appears to be acceptable. This was shown in a retrospective analysis of data from the prospectively performed EICESS, in which pulmonary function tests were available for 37 patients who were treated with whole-lung irradiation (with or without a boost) [130]. At a median follow-up of 25 months, an assessment of pulmonary complications in patients treated with whole-lung irradiation without further boost were classified as none (43 percent), mild (29 percent), moderate (21 percent), and severe (7 percent). Slightly higher complication rates were reported in patients who had an additional radiation boost or surgery to the thorax in addition to RT. The addition of consolidation whole-lung RT appears to be well tolerated in adult patients with lung metastases, with minimal reported significant acute or late toxicity and a three-year rate of freedom from pulmonary relapse of 45 percent [129]. Boost using stereotactic body RT is also an attractive option. It is an area of active investigation in patients with ES and oligometastatic sites of disease [131].

Bone and soft tissue metastases — For young, fit patients with solitary or circumscribed bone or soft tissue metastases, we suggest aggressive multimodality treatment. Approximately 10 percent of such patients will achieve long-term survival with this approach. RT can be delivered to metastatic lesions (doses equivalent to 40 to 50 Gy) in addition to irradiation of the primary tumor. One small series of 13 patients with metastatic ES and rhabdomyosarcoma reported only one local failure at irradiated sites with minimal associated toxicity [131]. Stereotactic body RT is an area of active investigation in patients with oligometastatic sites of disease [132].

RECURRENT DISEASE — 

Most relapses occur within two years of initial diagnosis, but late relapse is not uncommon [133-135]. In a report from the Childhood Cancer Survivor Study, the 20-year cumulative incidence of a late recurrence among five-year survivors of ES was 13 percent [135]. For this reason, patients should be followed indefinitely for the potential of late relapse. (See 'Posttreatment surveillance' below.)

In general, survival after an early relapse is poor, with few survivors among those who relapse within two years of therapy [136,137]. By contrast, up to 15 to 20 percent of those who relapse later may survive long-term. Other prognostic factors associated with an increased risk of death in patients with recurrent ES include recurrence at combined local and distant sites, and an elevated lactate dehydrogenase at initial diagnosis. Some patients with a relapse may be candidates for local therapy in addition to systemic therapy. (See 'Local control in patients with metastatic disease' above.)

Initial recurrence — For patients with an initial recurrence of ES, we suggest either high-dose ifosfamide or the combination of irinotecan plus temozolomide (IT) with or without vincristine, rather than topotecan plus cyclophosphamide (TC) [138-140]. For patients who decline or are ineligible for either of these regimens due to potential toxicities, TC is an appropriate alternative. Since treatment intent is generally palliative and each chemotherapy regimen carries a different toxicity profile, clinicians should discuss the risks and benefits of each treatment with their patients. Some patients may be candidates for local therapy. (See 'Local control in patients with metastatic disease' above.)

Although the prognosis for patients with recurrent disease is poor, some patients can be successfully salvaged, particularly patients with late relapses [141,142]. Sites of recurrence, prior treatment, and relapse-free interval affect remaining treatment choices. Most patients with recurrent disease will receive systemic therapy prior to attempts at additional local control measures. Patients relapsing after a lengthy disease-free interval off chemotherapy may respond again to the same agents used as part of initial therapy.

An international phase III trial (rEECur) of 439 patients with recurrent or primary refractory ES randomly assigned treatment to one of four different chemotherapy regimens: high-dose ifosfamide, TC, IT, and gemcitabine plus docetaxel (GD) [143]. During the first two interim analyses, IT and GD were dropped from further study as these regimens were predicted to have a lower probability of superiority compared with the remaining arms.

The phase III portion of rEECur compared high-dose ifosfamide versus TC in 146 patients. At a median follow-up of 40 months, presented in abstract form, high-dose ifosfamide demonstrated [143]:

Numerically higher event-free survival (EFS; median 5.7 versus 3.5 months; six-month EFS 47 versus 37 percent; hazard ratio [HR] 0.73, 95% CI 0.51-1.05) and overall survival (OS; median 15.4 versus 10.5 months; one-year OS 55 versus 45 percent; HR 0.73, 95% CI 0.50-1.08); this difference, while potentially clinically significant, did not reach statistically significance. Objective response rates were also higher for high-dose ifosfamide compared with TC (30 versus 21 percent). In a subgroup analysis, high-dose ifosfamide conferred a higher EFS benefit versus TC for children (age <14 years) compared with adolescents and adults (age ≥14 years).

Higher rates of grade ≥3 neurotoxicity (7 versus 0 percent), nephrotoxicity (8 versus 0 percent), and infection (14 versus 8 percent), leading to higher rates of treatment discontinuation. Other grade ≥3 toxicity rates were similar between the two treatment arms including febrile neutropenia (25 versus 26 percent), vomiting (1 percent each), nausea (3 versus 0 percent), and diarrhea (1 percent each).

Due to the trial design of rEECur, it is difficult to compare the efficacy of high-dose ifosfamide versus IT. These two regimens were not directly compared in the randomized phase III portion of the study, and the dropping of the IT treatment arm implies that its efficacy was inferior to high-dose ifosfamide. However, in preliminary results of an interim analysis of rEECur, at median follow-up of nine months, among the 127 patients treated with IT arm, median EFS and OS were 4.7 and 13.9 months, respectively, which were only slightly lower than that seen with high-dose ifosfamide (5.7 and 15.7 months, respectively) but higher than that seen with TC (3.5 and 10.5 months, respectively) [144]. Grade ≥3 toxicity for IT were diarrhea (17 percent), nausea, vomiting (6 percent each), fatigue, and febrile neutropenia (3 percent each). Since data on patient-reported outcomes are limited, toxicity profiles are different for each regimens, and treatment is generally palliative in this situation, both IT and TC remain reasonable options in patients with recurrent ES. [125,145].

For patients treated with IT, a randomized single-center phase II trial reported a higher response rate using a 10-day irinotecan schedule rather than the 5-day schedule studied in rEECur [138]. This regimen also included vincristine and the extent to which vincristine contributed to these outcomes remains unclear. These findings have not been validated in larger studies and choice of the 5-day versus 10-day schedule remains individualized based upon goals of care and feasibility of frequent infusion visits.

Multiply relapsed disease — Patients with multiply relapsed ES are treated with palliative intent. Options include:

Multitargeted tyrosine kinase inhibitors – The multitargeted tyrosine kinase inhibitors cabozantinib and regorafenib are alternative options for subsequent-line therapy in patients with relapsed/refractory or metastatic disease. Cabozantinib has demonstrated activity in patients with advanced ES, with an objective response rate of 26 percent and median progression-free survival of five months in one phase II trial (CABONE) [146]. Likewise, a non-randomized phase II trial of regorafenib reported a 10 percent response rate in patients with recurrent ES [147]. In addition, a randomized placebo-controlled phase II trial of regorafenib reported nominally longer PFS in patients randomized to regorafenib compared with placebo [148].

Investigational agents – Patients with recurrent or advanced ES should be encouraged to participate in clinical trials, where available. A full listing of clinical trials in this patient population is available at clinicaltrials.gov. Future therapies will likely emerge as the fundamental biology of the fusion oncoproteins driving this disease is better understood [111,149]. (See "Epidemiology, pathology, and molecular genetics of Ewing sarcoma" and "Clinical presentation, staging, and prognostic factors of Ewing sarcoma", section on 'Prognostic factors'.)

In patients with recurrent or refractory ES, initial studies suggested activity for antibodies that target insulin-life growth factor-1 receptor, such as ganitumab [150-154]. However, in a randomized phase III trial of patients with treatment-naïve metastatic ES, the addition of ganitumab to interval-compressed chemotherapy with vincristine/doxorubicin/cyclophosphamide and ifosfamide/etoposide did not improve EFS and increased toxicity [116].

Local therapies — Management of a local recurrence usually includes surgery (and possibly an amputation if the local recurrence involves an irradiated extremity), radiation therapy (RT), or both. RT to bone lesions usually provides pain relief, while surgery and/or RT can eradicate disease in patients with isolated lung metastases [125]. As an example, one study of 26 patients with local recurrence showed that the survival at five years post local recurrence was 28 percent. Better survival outcomes were seen in those who did not have metastases at diagnosis of the recurrence, had a surgical treatment for the recurrence, and had complete eradication of all disease [145]. For patients with pulmonary recurrence who have not previously had whole-lung radiation, use of whole-lung radiation as a component of relapse therapy may prolong progression-free survival [155].

POSTTREATMENT SURVEILLANCE

Surveillance — There are no prospective data that address the appropriate schedule and type of surveillance for patients with ES after initial treatment. For patients with localized disease, we obtain a physical examination, complete blood count, chest imaging (radiographs or computed tomography [CT]), and surveillance imaging of the primary site every three months for two years, every six months for years 3 to 5, and annually thereafter. This is a similar approach to consensus-based guidelines from the National Comprehensive Cancer Network and from the Children's Oncology Group (table 3) [156-159]. For patients who presented with metastatic disease the frequency of imaging will depend on their treatment status and time since diagnosis. Patients who are on active treatment will need regular interval imaging at least every three months.

Local imaging of the primary site depends upon the specific site and prior local therapy. For example:

Patients with a metal endoprosthesis are usually imaged with plain radiographs. The metal limits the utility of magnetic resonance imaging (MRI) in this setting, although metal subtraction techniques have improved the ability to assess the surrounding soft tissues in some instances. In addition, not all extendible endoprostheses are compatible with MRI due to the magnetic mechanism that allows for extension.

Ultrasound may be useful to assess for soft tissue masses around an endoprosthesis or allograft with metal plate fixation.

For patients treated with definitive radiation therapy (RT), MRI may be appropriate.

For patients with chest wall primary tumors, a chest CT scan or chest radiograph may be used, depending upon level of concern for risk of recurrence.

Patients with symptoms or abnormal imaging or those who are planning surgical interventions may also be offered fluorodeoxyglucose (FDG) positron emission tomography (PET)-CT. A meta-analysis study showed that FDG-PET and PET-CT have a high accuracy in detecting distant metastases and postoperative recurrences in patients with ES [160].

We and others suggest that patients be followed indefinitely for the development of treatment-related toxicities and relapse [161]. Although most recurrences are observed within 10 years, patients with ES are at risk for late relapse and late development of treatment-related complications such as second neoplasms.

Clinicians performing posttreatment surveillance must be aware of concerns for radiation exposure and the risk for secondary malignancies, particularly in younger individuals. (See 'Complications in long-term survivors' below and "Radiation-related risks of imaging".)

Patients on surveillance who develop symptoms — Recurrent disease may be suspected in patients with symptoms at the primary site or elsewhere. Evaluation of suspected recurrence should include an assessment for disease at the primary site and for the presence of metastatic disease to guide therapy. Most patients with a local recurrence have either gross or microscopic metastatic disease. The evaluation for metastatic disease is similar to that done at the time of initial staging, as described separately. (See "Clinical presentation, staging, and prognostic factors of Ewing sarcoma", section on 'Staging evaluation'.)

The documentation of a local recurrence can be challenging, especially in patients with metallic implants given distortion on imaging. In patients with metallic endoprostheses, MRI and CT evaluation can be distorted by metal artifact, although this is improved with contemporary MRI and CT imaging technology. In patients who are treated with certain extendable "growing" prostheses, MRI may be contraindicated. The interpretation of irradiated areas on imaging studies can be challenging because of the changes in bone caused by the radiation. Soft tissue masses may represent residual fibrosis rather than recurrent tumor. The evaluation of intraosseous sites is even more difficult since the response to prior therapy and the variability in remodeling may complicate the interpretation of radiographic studies. Progressive cortical destruction or increasing radiolucent areas suggest local recurrence, as do bone scans that demonstrate increased radiotracer uptake.

In patients with suspicion for recurrence we use the following guidance:

PET scans may be useful to assess the likelihood of recurrence at a site that is suspicious on cross-sectional radiographic imaging.

Ultrasound can also be useful to assess for soft tissue masses around an endoprosthesis.

The decision to biopsy a site of suspected recurrent local or metastatic disease depends upon patient history and the level of evidence from imaging studies. Core needle biopsies can usually distinguish between recurrence versus reactive tissue/fibrosis and is the preferred initial biopsy technique. If this is nondiagnostic, then an open bone biopsy should be performed. However, open biopsies can be associated with local morbidity (ie, wound complications and bone fracture).

COMPLICATIONS IN LONG-TERM SURVIVORS — 

Long-term survivors of ES may have considerable burden of the late effects of their therapy and need to be followed lifelong for development of toxicities related to treatment [10,42,161-165]. Complications of chemotherapy include subsequent primary cancers, reduced fertility, kidney impairment, and cardiomyopathy. Complications of radiation therapy (RT) include subsequent primary cancers, pathologic fractures, wound complications, pulmonary fibrosis, neuropathy, limb leg discrepancy, and femoral head necrosis. The frequency and severity of these complications with modern treatment approaches are difficult to predict given the evolution of therapy and lag in data on long-term follow-up. Long-term follow-up guidelines after treatment of childhood malignancy are available from the Children's Oncology Group.

Secondary myelodysplasia (MDS) and leukemia are particular concerns for this population [166-170]. This was illustrated in a report from the Children's Oncology Group of 578 children with ES who were treated with three different regimens over a six-year period [168]. Overall, 11 children developed secondary MDS/acute myeloid leukemia, and the cumulative risk was higher among children treated with a regimen incorporating higher doses of doxorubicin, cyclophosphamide, and ifosfamide as compared with those receiving standard-dose vincristine, doxorubicin, cyclophosphamide, and dactinomycin with or without ifosfamide plus etoposide (11, 0.9, and 0.4 percent at five years, respectively).

The health status of long-term (≥5 years) survivors was addressed in a cohort study of 568 individuals who were diagnosed with ES before age 21 from 1970 to 1986, including a subset of 403 patients who were participating in the Childhood Cancer Survivor Study [164]. Cumulative mortality among all survivors was 25 percent at 25 years after diagnosis, and the cumulative incidence of secondary malignancy was 9 percent. Disease progression/recurrence accounted for 60 percent of all deaths, while other causes included secondary neoplasms, cardiac disease, pulmonary disease, and other medical causes. The cumulative mortality attributed to subsequent malignant neoplasms and cardiopulmonary disease potentially attributed to treatment was 8.3 percent at 25 years. In addition, compared with their siblings, survivors had higher rates of severe, disabling, or chronic health conditions; lower fertility rates; and higher rates of self-reported moderate to extreme adverse health status.

It is anticipated that the risk of late effects will decrease as a result of improvements in RT technique over the last 20 years (ie, the adoption of tailored radiation ports as opposed to whole-bone ports used in the period 1960 to 1980; three-dimensional conformal radiation therapy, intensity-modulated radiation therapy, and proton therapy; and use of lower and risk-adapted RT doses) [82,171]. In addition, trends in chemotherapy intensification may alter the pattern of secondary malignancies [163]. Nevertheless, the relatively high complication rate seen with earlier treatment approaches and the delayed nature of many of these complications underscore the need for long-term posttreatment follow-up.

SOCIETY GUIDELINE LINKS — 

Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Soft tissue sarcoma" and "Society guideline links: Bone sarcomas".)

INFORMATION FOR PATIENTS — 

UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topics (see "Patient education: Ewing sarcoma (The Basics)" and "Patient education: Bone cancer (The Basics)")

SUMMARY AND RECOMMENDATIONS

Definition of localized disease – The treatment of Ewing sarcoma (ES) differs depending upon whether the patient has localized or metastatic disease. Localized ES is confined to the site where it originated (see 'Definition of localized disease' above). Metastatic disease refers to disease anywhere other than the primary site and can include involvement of lung, bone, bone marrow, distant lymph nodes, lesions discontinuous from the primary tumor (ie, skip metastases), or other distant sites.

Multimodality therapy for localized disease – Patients with localized ES should be treated with multimodality therapy which includes neoadjuvant chemotherapy; local treatment such as surgery or radiation therapy (RT); and adjuvant chemotherapy (algorithm 1).

Neoadjuvant chemotherapy – For patients with localized ES, we recommend initial chemotherapy with alternating cycles of vincristine/doxorubicin/cyclophosphamide and ifosfamide/etoposide (VDC/IE) rather than vincristine, ifosfamide, doxorubicin, and etoposide (Grade 1B), as this regimen improves overall survival (OS) and is shorter in duration with less toxicity. Neoadjuvant chemotherapy is given to reduce local tumor volume, facilitate resection, and treat micrometastatic disease. Patients receive 12 weeks of neoadjuvant therapy prior to local therapy. (See 'VDC/IE preferred' above.)

-Patients age <18 years – For patients age <18 years with localized ES, we recommend interval-compressed VDC/IE given every 14 days with hematopoietic growth factor support (table 1), rather than every 21 days (Grade 1B), as this approach improves event-free survival and OS without increased toxicity or higher risk of second (subsequent) malignant neoplasms. (See 'Interval-compressed VDC/IE' above.)

-Patients age ≥18 years – For patients age ≥18 years, we offer VDC/IE either every 14 or 21 days with hematopoietic growth factor support. (See 'Patients 18 years or older' above.)

Local treatment – Either surgery or RT can provide effective local control. Tumor location, size, and impact of adjacent structures all influence choice of therapy. For tumors that can be resected with adequate margins and acceptable function and morbidity, we suggest surgery rather than RT (Grade 2C). RT is an acceptable alternative and preferred when the tumor is unresectable or if a resection will result in significant morbidity. (See 'Local treatment' above.)

When RT is delivered as local therapy, chemotherapy should not be interrupted. It should be delivered concurrently with RT (with omission of doxorubicin).

Adjuvant Therapy

-Adjuvant chemotherapy – Adjuvant chemotherapy should be commenced as soon as possible after surgery and given concurrently with radiation for patients who do not have surgery. For patients receiving VDC/IE, we typically administer a total of 14 to 17 cycles which includes those cycles received as neoadjuvant therapy. (See 'Adjuvant chemotherapy' above.)

-Adjuvant radiation therapy – For patients with positive margins, gross residual disease, and/or high risk chest wall tumors, we suggest adjuvant RT (Grade 2C). Adjuvant RT is not usually offered to other populations. (See 'When to use adjuvant radiation therapy' above.)

Metastatic disease

Chemotherapy - Patients with metastatic ES at diagnosis receive multimodality treatment given with potentially curative intent as a subset of these patients may be cured. For most patients with metastatic ES, we suggest alternating cycles of VDC/IE (Grade 2C), based on extrapolation of data from trials in localized disease. However, since VDC/IE has not demonstrated a specific benefit for the addition of I/E in this population, some clinicians may opt for VDC for patients with unequivocal evidence of metastatic disease. (See 'Initial selection of chemotherapy' above.)

Local therapy – In those with metastatic ES, local treatment of the primary tumor is performed as for patients with localized disease, though more of these patients are selected for definitive radiotherapy rather than surgery. In addition, local therapy of metastatic sites is offered to patients with isolated pulmonary metastases or solitary or circumscribed bone or soft tissue metastases. (See 'Local control in patients with metastatic disease' above.)

Recurrent disease – Some patients with recurrent ES can achieve long-term disease control. The sites of recurrence, prior treatment, and relapse-free interval affect the treatment choices. Most patients with recurrent ES will receive systemic therapy prior to attempts at additional local control measures. (See 'Recurrent disease' above.)

Posttreatment monitoring – Most relapses occur within two years of initial diagnosis, but later relapses have been observed. To monitor for relapse and late effects of therapy, patients are followed indefinitely with physical examination, bloodwork, and imaging (table 3). (See 'Surveillance' above.)

Lifelong follow-up is needed because disease relapse, treatment-related complications, and second malignancies can all occur beyond five years after treatment. (See 'Complications in long-term survivors' above.)

ACKNOWLEDGMENT — 

The UpToDate editorial staff acknowledges David C Harmon, MD, who contributed to earlier versions of this topic review.

  1. Craft A, Cotterill S, Malcolm A, et al. Ifosfamide-containing chemotherapy in Ewing's sarcoma: The Second United Kingdom Children's Cancer Study Group and the Medical Research Council Ewing's Tumor Study. J Clin Oncol 1998; 16:3628.
  2. 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.
  3. Nesbit ME Jr, Gehan EA, Burgert EO Jr, 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.
  4. Obata H, Ueda T, Kawai A, et al. Clinical outcome of patients with Ewing sarcoma family of tumors of bone in Japan: the Japanese Musculoskeletal Oncology Group cooperative study. Cancer 2007; 109:767.
  5. Donaldson SS, Torrey M, Link MP, et al. A multidisciplinary study investigating radiotherapy in Ewing's sarcoma: end results of POG #8346. Pediatric Oncology Group. Int J Radiat Oncol Biol Phys 1998; 42:125.
  6. Nilbert M, Saeter G, Elomaa I, et al. Ewing's sarcoma treatment in Scandinavia 1984-1990--ten-year results of the Scandinavian Sarcoma Group Protocol SSGIV. Acta Oncol 1998; 37:375.
  7. Raney RB, Asmar L, Newton WA Jr, et al. Ewing's sarcoma of soft tissues in childhood: a report from the Intergroup Rhabdomyosarcoma Study, 1972 to 1991. J Clin Oncol 1997; 15:574.
  8. Ferrari S, Mercuri M, Rosito P, et al. Ifosfamide and actinomycin-D, added in the induction phase to vincristine, cyclophosphamide and doxorubicin, improve histologic response and prognosis in patients with non metastatic Ewing's sarcoma of the extremity. J Chemother 1998; 10:484.
  9. Kinsella TJ, Miser JS, Waller B, et al. Long-term follow-up of Ewing's sarcoma of bone treated with combined modality therapy. Int J Radiat Oncol Biol Phys 1991; 20:389.
  10. 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.
  11. Jürgens H, Bier V, Harms D, et al. Malignant peripheral neuroectodermal tumors. A retrospective analysis of 42 patients. Cancer 1988; 61:349.
  12. Ahmad R, Mayol BR, Davis M, Rougraff BT. Extraskeletal Ewing's sarcoma. Cancer 1999; 85:725.
  13. Gururangan S, Marina NM, Luo X, et al. Treatment of children with peripheral primitive neuroectodermal tumor or extraosseous Ewing's tumor with Ewing's-directed therapy. J Pediatr Hematol Oncol 1998; 20:55.
  14. Castex MP, Rubie H, Stevens MC, et al. Extraosseous localized ewing tumors: improved outcome with anthracyclines--the French society of pediatric oncology and international society of pediatric oncology. J Clin Oncol 2007; 25:1176.
  15. Shamberger RC, Laquaglia MP, Krailo MD, et al. Ewing sarcoma of the rib: results of an intergroup study with analysis of outcome by timing of resection. J Thorac Cardiovasc Surg 2000; 119:1154.
  16. 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.
  17. Mirzaei L, Kaal SE, Schreuder HW, Bartels RH. The Neurological Compromised Spine Due to Ewing Sarcoma. What First: Surgery or Chemotherapy? Therapy, Survival, and Neurological Outcome of 15 Cases With Primary Ewing Sarcoma of the Vertebral Column. Neurosurgery 2015; 77:718.
  18. Brennan B, Kirton L, Marec-Bérard 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.
  19. Leavey PJ, Laack NN, Krailo MD, et al. Phase III Trial Adding Vincristine-Topotecan-Cyclophosphamide to the Initial Treatment of Patients With Nonmetastatic Ewing Sarcoma: A Children's Oncology Group Report. J Clin Oncol 2021; 39:4029.
  20. Cash T, Krailo MD, Buxton A, et al. Long-term outcomes in patients with localized Ewing sarcoma treated with interval-compressed chemotherapy: A long-term follow-up report from Children’s Oncology Group study AEWS0031. J Clin Oncol 2022; 40;16S.
  21. 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.
  22. Cash T, Krailo MD, Buxton AB, et al. Long-Term Outcomes in Patients With Localized Ewing Sarcoma Treated With Interval-Compressed Chemotherapy on Children's Oncology Group Study AEWS0031. J Clin Oncol 2023; 41:4724.
  23. 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.
  24. 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.
  25. Heesen P, Ranft A, Bhadri V, et al. Association between local treatment modalities and event-free survival, overall survival, and local recurrence in patients with localised Ewing Sarcoma. Report from the Ewing 2008 trial. Eur J Cancer 2023; 192:113260.
  26. 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.
  27. 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.
  28. Carrie C, Mascard E, Gomez F, et al. Nonmetastatic pelvic Ewing sarcoma: report of the French society of pediatric oncology. Med Pediatr Oncol 1999; 33:444.
  29. 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.
  30. Dunst J, Sauer R, Burgers JM, et al. Radiation therapy as local treatment in Ewing's sarcoma. Results of the Cooperative Ewing's Sarcoma Studies CESS 81 and CESS 86. Cancer 1991; 67:2818.
  31. Wilkins RM, Pritchard DJ, Burgert EO Jr, Unni KK. Ewing's sarcoma of bone. Experience with 140 patients. Cancer 1986; 58:2551.
  32. Givens SS, Woo SY, Huang LY, et al. Non-metastatic Ewing's sarcoma: twenty years of experience suggests that surgery is a prime factor for successful multimodality therapy. Int J Oncol 1999; 14:1039.
  33. Indelicato DJ, Keole SR, Shahlaee AH, et al. Long-term clinical and functional outcomes after treatment for localized Ewing's tumor of the lower extremity. Int J Radiat Oncol Biol Phys 2008; 70:501.
  34. Ozaki T, Hillmann A, Hoffmann C, et al. Significance of surgical margin on the prognosis of patients with Ewing's sarcoma. A report from the Cooperative Ewing's Sarcoma Study. Cancer 1996; 78:892.
  35. Marcove RC, Rosen G. Radical en bloc excision of Ewing's sarcoma. Clin Orthop Relat Res 1980; :86.
  36. Sailer SL, Harmon DC, Mankin HJ, et al. Ewing's sarcoma: surgical resection as a prognostic factor. Int J Radiat Oncol Biol Phys 1988; 15:43.
  37. Aparicio J, Munárriz B, Pastor M, et al. Long-term follow-up and prognostic factors in Ewing's sarcoma. A multivariate analysis of 116 patients from a single institution. Oncology 1998; 55:20.
  38. 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.
  39. Shankar AG, Pinkerton CR, Atra A, et al. Local therapy and other factors influencing site of relapse in patients with localised Ewing's sarcoma. United Kingdom Children's Cancer Study Group (UKCCSG). Eur J Cancer 1999; 35:1698.
  40. Werier J, Yao X, Caudrelier JM, et al. A systematic review of optimal treatment strategies for localized Ewing's sarcoma of bone after neo-adjuvant chemotherapy. Surg Oncol 2016; 25:16.
  41. Bacci G, Ferrari S, Mercuri M, et al. Multimodal therapy for the treatment of nonmetastatic Ewing sarcoma of pelvis. J Pediatr Hematol Oncol 2003; 25:118.
  42. 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.
  43. Bacci G, Ferrari S, Bertoni F, et al. Prognostic factors in nonmetastatic Ewing's sarcoma of bone treated with adjuvant chemotherapy: analysis of 359 patients at the Istituto Ortopedico Rizzoli. J Clin Oncol 2000; 18:4.
  44. Daw NC, Laack NN, McIlvaine EJ, et al. Local Control Modality and Outcome for Ewing Sarcoma of the Femur: A Report From the Children's Oncology Group. Ann Surg Oncol 2016; 23:3541.
  45. Lin TA, Ludmir EB, Liao KP, et al. Timing of Local Therapy Affects Survival in Ewing Sarcoma. Int J Radiat Oncol Biol Phys 2019; 104:127.
  46. Guo W, Li D, Tang X, et al. Reconstruction with modular hemipelvic prostheses for periacetabular tumor. Clin Orthop Relat Res 2007; 461:180.
  47. Fan H, Fu J, Li X, et al. Implantation of customized 3-D printed titanium prosthesis in limb salvage surgery: a case series and review of the literature. World J Surg Oncol 2015; 13:308.
  48. Woo SH, Sung MJ, Park KS, Yoon TR. Three-dimensional-printing Technology in Hip and Pelvic Surgery: Current Landscape. Hip Pelvis 2020; 32:1.
  49. Ahmed SK, Randall RL, DuBois SG, et al. Identification of Patients With Localized Ewing Sarcoma at Higher Risk for Local Failure: A Report From the Children's Oncology Group. Int J Radiat Oncol Biol Phys 2017; 99:1286.
  50. Stea B, Kinsella TJ, Triche TJ, et al. Treatment of pelvic sarcomas in adolescents and young adults with intensive combined modality therapy. Int J Radiat Oncol Biol Phys 1987; 13:1797.
  51. Calvo FA, Ortiz de Urbina D, Sierrasesúmaga L, et al. Intraoperative radiotherapy in the multidisciplinary treatment of bone sarcomas in children and adolescents. Med Pediatr Oncol 1991; 19:478.
  52. Hoffmann C, Ahrens S, Dunst J, et al. Pelvic Ewing sarcoma: a retrospective analysis of 241 cases. Cancer 1999; 85:869.
  53. La TH, Meyers PA, Wexler LH, et al. Radiation therapy for Ewing's sarcoma: results from Memorial Sloan-Kettering in the modern era. Int J Radiat Oncol Biol Phys 2006; 64:544.
  54. Rödl RW, Hoffmann C, Gosheger G, et al. Ewing's sarcoma of the pelvis: combined surgery and radiotherapy treatment. J Surg Oncol 2003; 83:154.
  55. Vogin G, Helfre S, Glorion C, et al. Local control and sequelae in localised Ewing tumours of the spine: a French retrospective study. Eur J Cancer 2013; 49:1314.
  56. 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.
  57. Donati D, Yin J, Di Bella C, et al. Local and distant control in non-metastatic pelvic Ewing's sarcoma patients. J Surg Oncol 2007; 96:19.
  58. Sucato DJ, Rougraff B, McGrath BE, et al. Ewing's sarcoma of the pelvis. Long-term survival and functional outcome. Clin Orthop Relat Res 2000; :193.
  59. Indelicato DJ, Keole SR, Shahlaee AH, et al. Impact of local management on long-term outcomes in Ewing tumors of the pelvis and sacral bones: the University of Florida experience. Int J Radiat Oncol Biol Phys 2008; 72:41.
  60. 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.
  61. Casey DL, Meyers PA, Alektiar KM, et al. Ewing sarcoma in adults treated with modern radiotherapy techniques. Radiother Oncol 2014; 113:248.
  62. Pretz JL, Barysauskas CM, George S, et al. Localized Adult Ewing Sarcoma: Favorable Outcomes with Alternating Vincristine, Doxorubicin, Cyclophosphamide, and Ifosfamide, Etoposide (VDC/IE)-Based Multimodality Therapy. Oncologist 2017; 22:1265.
  63. Wagner MJ, Gopalakrishnan V, Ravi V, et al. Vincristine, Ifosfamide, and Doxorubicin for Initial Treatment of Ewing Sarcoma in Adults. Oncologist 2017; 22:1271.
  64. 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.
  65. Andreou D, Ranft A, Gosheger G, et al. Which Factors Are Associated with Local Control and Survival of Patients with Localized Pelvic Ewing's Sarcoma? A Retrospective Analysis of Data from the Euro-EWING99 Trial. Clin Orthop Relat Res 2020; 478:290.
  66. Dunst J, Jürgens 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.
  67. Johnstone PA, Wexler LH, Venzon DJ, et al. Sarcomas of the hand and foot: analysis of local control and functional result with combined modality therapy in extremity preservation. Int J Radiat Oncol Biol Phys 1994; 29:735.
  68. Gonzalez DG, Breur K. Clinical data from irradiated growing long bones in children. Int J Radiat Oncol Biol Phys 1983; 9:841.
  69. Kinsella TJ, Loeffler JS, Fraass BA, Tepper J. Extremity preservation by combined modality therapy in sarcomas of the hand and foot: an analysis of local control, disease free survival and functional result. Int J Radiat Oncol Biol Phys 1983; 9:1115.
  70. Hall EJ. Intensity-modulated radiation therapy, protons, and the risk of second cancers. Int J Radiat Oncol Biol Phys 2006; 65:1.
  71. Xiang M, Chang DT, Pollom EL. Second cancer risk after primary cancer treatment with three-dimensional conformal, intensity-modulated, or proton beam radiation therapy. Cancer 2020; 126:3560.
  72. Miralbell R, Lomax A, Cella L, Schneider U. Potential reduction of the incidence of radiation-induced second cancers by using proton beams in the treatment of pediatric tumors. Int J Radiat Oncol Biol Phys 2002; 54:824.
  73. Rombi B, DeLaney TF, MacDonald SM, et al. Proton radiotherapy for pediatric Ewing's sarcoma: initial clinical outcomes. Int J Radiat Oncol Biol Phys 2012; 82:1142.
  74. Chung CS, Keating N, Yock T, Tarbell N. Comparative analysis of second malignancy risk in patients treated with proton therapy versus conventional photon therapy. Int J Radiat Oncol Biol Phys 2008; 72:S8.
  75. Paganetti H, Athar BS, Moteabbed M, et al. Assessment of radiation-induced second cancer risks in proton therapy and IMRT for organs inside the primary radiation field. Phys Med Biol 2012; 57:6047.
  76. Polf JC, Newhauser WD, Titt U. Patient neutron dose equivalent exposures outside of the proton therapy treatment field. Radiat Prot Dosimetry 2005; 115:154.
  77. Schneider U, Agosteo S, Pedroni E, Besserer J. Secondary neutron dose during proton therapy using spot scanning. Int J Radiat Oncol Biol Phys 2002; 53:244.
  78. Uezono H, Indelicato DJ, Rotondo RL, et al. Treatment Outcomes After Proton Therapy for Ewing Sarcoma of the Pelvis. Int J Radiat Oncol Biol Phys 2020; 107:974.
  79. Kharod SM, Indelicato DJ, Rotondo RL, et al. Outcomes following proton therapy for Ewing sarcoma of the cranium and skull base. Pediatr Blood Cancer 2020; 67:e28080.
  80. Lawell MP, Indelicato DJ, Paulino AC, et al. An open invitation to join the Pediatric Proton/Photon Consortium Registry to standardize data collection in pediatric radiation oncology. Br J Radiol 2020; 93:20190673.
  81. Vogin G, Biston MC, Marchesi V, et al. [Localized Ewing sarcoma of the spine: a preliminary dose-escalation study comparing innovative radiation techniques in a single patient]. Cancer Radiother 2013; 17:26.
  82. Arai Y, Kun LE, Brooks MT, et al. Ewing's sarcoma: local tumor control and patterns of failure following limited-volume radiation therapy. Int J Radiat Oncol Biol Phys 1991; 21:1501.
  83. Merchant TE, Kushner BH, Sheldon JM, et al. Effect of low-dose radiation therapy when combined with surgical resection for Ewing sarcoma. Med Pediatr Oncol 1999; 33:65.
  84. Talleur AC, Navid F, Spunt SL, et al. Limited Margin Radiation Therapy for Children and Young Adults With Ewing Sarcoma Achieves High Rates of Local Tumor Control. Int J Radiat Oncol Biol Phys 2016; 96:119.
  85. Laskar S, Sinha S, Chatterjee A, et al. Radiation Therapy Dose Escalation in Unresectable Ewing Sarcoma: Final Results of a Phase 3 Randomized Controlled Trial. Int J Radiat Oncol Biol Phys 2022; 113:996.
  86. Kacar M, Nagel MB, Liang J, et al. Radiation therapy dose escalation achieves high rates of local control with tolerable toxicity profile in pediatric and young adult patients with Ewing sarcoma. Cancer 2024; 130:1836.
  87. Ahmed SK, Robinson SI, Arndt CA, et al. Pelvis Ewing sarcoma: Local control and survival in the modern era. Pediatr Blood Cancer 2017.
  88. Bolek TW, Marcus RB Jr, Mendenhall NP, et al. Local control and functional results after twice-daily radiotherapy for Ewing's sarcoma of the extremities. Int J Radiat Oncol Biol Phys 1996; 35:687.
  89. Davis AM, Wright JG, Williams JI, et al. Development of a measure of physical function for patients with bone and soft tissue sarcoma. Qual Life Res 1996; 5:508.
  90. Smith MA, Altekruse SF, Adamson PC, et al. Declining childhood and adolescent cancer mortality. Cancer 2014; 120:2497.
  91. Smith MA, Gurney JG, Ries LA. Cancer among adolescents 15 to 19 years old. In: Cancer Incidence and Survival Among Children and Adolescents: United States SEER Program 1975-1995, Ries LA, Smith MAS, Gurney JG, et al (Eds). SEER Program, National Cancer Institute, Bethesda, MD 1999. (Pub #99-4649). https://seer.cancer.gov/archive/publications/childhood/childhood-monograph.pdf.
  92. 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.
  93. 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.
  94. Wunder JS, Paulian G, Huvos AG, et al. The histological response to chemotherapy as a predictor of the oncological outcome of operative treatment of Ewing sarcoma. J Bone Joint Surg Am 1998; 80:1020.
  95. 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.
  96. Picci P, Rougraff BT, Bacci G, et al. Prognostic significance of histopathologic response to chemotherapy in nonmetastatic Ewing's sarcoma of the extremities. J Clin Oncol 1993; 11:1763.
  97. Sluga M, Windhager R, Lang S, et al. The role of surgery and resection margins in the treatment of Ewing's sarcoma. Clin Orthop Relat Res 2001; :394.
  98. Foulon S, Brennan B, Gaspar N, et al. Can postoperative radiotherapy be omitted in localised standard-risk Ewing sarcoma? An observational study of the Euro-E.W.I.N.G group. Eur J Cancer 2016; 61:128.
  99. Bacci G, Longhi A, Briccoli A, et al. The role of surgical margins in treatment of Ewing's sarcoma family tumors: experience of a single institution with 512 patients treated with adjuvant and neoadjuvant chemotherapy. Int J Radiat Oncol Biol Phys 2006; 65:766.
  100. Kushner BH, Meyers PA, Gerald WL, et al. Very-high-dose short-term chemotherapy for poor-risk peripheral primitive neuroectodermal tumors, including Ewing's sarcoma, in children and young adults. J Clin Oncol 1995; 13:2796.
  101. Pinkerton CR, Bataillard A, Guillo S, et al. Treatment strategies for metastatic Ewing's sarcoma. Eur J Cancer 2001; 37:1338.
  102. Hayes FA, Thompson EI, Parvey L, et al. Metastatic Ewing's sarcoma: remission induction and survival. J Clin Oncol 1987; 5:1199.
  103. Sandoval C, Meyer WH, Parham DM, et al. Outcome in 43 children presenting with metastatic Ewing sarcoma: the St. Jude Children's Research Hospital experience, 1962 to 1992. Med Pediatr Oncol 1996; 26:180.
  104. 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.
  105. Craft AW, Cotterill SJ, Bullimore JA, Pearson D. Long-term results from the first UKCCSG Ewing's Tumour Study (ET-1). United Kingdom Children's Cancer Study Group (UKCCSG) and the Medical Research Council Bone Sarcoma Working Party. Eur J Cancer 1997; 33:1061.
  106. Deméocq F, Oberlin O, Benz-Lemoine E, et al. Initial chemotherapy including ifosfamide in the management of Ewing's sarcoma: preliminary results. A protocol of the French Pediatric Oncology Society (SFOP). Cancer Chemother Pharmacol 1989; 24 Suppl 1:S45.
  107. Marina NM, Pappo AS, Parham DM, et al. Chemotherapy dose-intensification for pediatric patients with Ewing's family of tumors and desmoplastic small round-cell tumors: a feasibility study at St. Jude Children's Research Hospital. J Clin Oncol 1999; 17:180.
  108. Paulussen M, Ahrens S, Craft AW, et al. Ewing's tumors with primary lung metastases: survival analysis of 114 (European Intergroup) Cooperative Ewing's Sarcoma Studies patients. J Clin Oncol 1998; 16:3044.
  109. 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.
  110. Brunetto AL, Castillo LA, Petrilli AS, et al. Carboplatin in the treatment of Ewing sarcoma: Results of the first Brazilian collaborative study group for Ewing sarcoma family tumors-EWING1. Pediatr Blood Cancer 2015; 62:1747.
  111. Balamuth NJ, Womer RB. Ewing's sarcoma. Lancet Oncol 2010; 11:184.
  112. 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.
  113. 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.
  114. 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.
  115. 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.
  116. DuBois SG, Krailo MD, Glade-Bender J, et al. Randomized Phase III Trial of Ganitumab With Interval-Compressed Chemotherapy for Patients With Newly Diagnosed Metastatic Ewing Sarcoma: A Report From the Children's Oncology Group. J Clin Oncol 2023; 41:2098.
  117. Goldsby RE, Ablin AR. Surviving childhood cancer; now what? Controversies regarding long-term follow-up. Pediatr Blood Cancer 2004; 43:211.
  118. Dirksen U, Brennan B, Le Deley MC, et al. High-Dose Chemotherapy Compared With Standard Chemotherapy and Lung Radiation in Ewing Sarcoma With Pulmonary Metastases: Results of the European Ewing Tumour Working Initiative of National Groups, 99 Trial and EWING 2008. J Clin Oncol 2019; 37:3192.
  119. Haveman LM, van Ewijk R, van Dalen EC, et al. High-dose chemotherapy followed by autologous haematopoietic cell transplantation for children, adolescents, and young adults with primary metastatic Ewing sarcoma. Cochrane Database Syst Rev 2021; 9:CD011405.
  120. Koch R, Gelderblom H, Haveman L, et al. High-Dose Treosulfan and Melphalan as Consolidation Therapy Versus Standard Therapy for High-Risk (Metastatic) Ewing Sarcoma. J Clin Oncol 2022; 40:2307.
  121. Miser JS, Kinsella TJ, Triche TJ, et al. Preliminary results of treatment of Ewing's sarcoma of bone in children and young adults: six months of intensive combined modality therapy without maintenance. J Clin Oncol 1988; 6:484.
  122. Pilepich MV, Vietti TJ, Nesbit ME, et al. Radiotherapy and combination chemotherapy in advanced Ewing's Sarcoma-Intergroup study. Cancer 1981; 47:1930.
  123. 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.
  124. Raciborska A, Bilska K, Rychłowska-Pruszyńska M, et al. Management and follow-up of Ewing sarcoma patients with isolated lung metastases. J Pediatr Surg 2016; 51:1067.
  125. Bacci G, Briccoli A, Picci P, Ferrari S. Metachronous pulmonary metastases resection in patients with Ewing's sarcoma initially treated with adjuvant or neoadjuvant chemotherapy. Eur J Cancer 1995; 31A:999.
  126. Dunst J, Paulussen M, Jürgens H. Lung irradiation for Ewing's sarcoma with pulmonary metastases at diagnosis: results of the CESS-studies. Strahlenther Onkol 1993; 169:621.
  127. Whelan JS, Burcombe RJ, Janinis J, et al. A systematic review of the role of pulmonary irradiation in the management of primary bone tumours. Ann Oncol 2002; 13:23.
  128. Spunt SL, McCarville MB, Kun LE, et al. Selective use of whole-lung irradiation for patients with Ewing sarcoma family tumors and pulmonary metastases at the time of diagnosis. J Pediatr Hematol Oncol 2001; 23:93.
  129. Casey DL, Alektiar KM, Gerber NK, Wolden SL. Whole lung irradiation for adults with pulmonary metastases from Ewing sarcoma. Int J Radiat Oncol Biol Phys 2014; 89:1069.
  130. Bölling 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.
  131. Liu AK, Stinauer M, Albano E, et al. Local control of metastatic sites with radiation therapy in metastatic Ewing sarcoma and rhabdomyosarcoma. Pediatr Blood Cancer 2011; 57:169.
  132. Brown LC, Lester RA, Grams MP, et al. Stereotactic body radiotherapy for metastatic and recurrent ewing sarcoma and osteosarcoma. Sarcoma 2014; 2014:418270.
  133. Weston CL, Douglas C, Craft AW, et al. Establishing long-term survival and cure in young patients with Ewing's sarcoma. Br J Cancer 2004; 91:225.
  134. Bacci G, Balladelli A, Forni C, et al. Adjuvant and neo-adjuvant chemotherapy for Ewing's sarcoma family tumors and osteosarcoma of the extremity: further outcome for patients event-free survivors 5 years from the beginning of treatment. Ann Oncol 2007; 18:2037.
  135. Wasilewski-Masker K, Liu Q, Yasui Y, et al. Late recurrence in pediatric cancer: a report from the Childhood Cancer Survivor Study. J Natl Cancer Inst 2009; 101:1709.
  136. 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.
  137. 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.
  138. Xu J, Xie L, Sun X, et al. Longer versus Shorter Schedules of Vincristine, Irinotecan, and Temozolomide (VIT) for Relapsed or Refractory Ewing Sarcoma: A Randomized Controlled Phase 2 Trial. Clin Cancer Res 2023; 29:1040.
  139. Dogan I, Iribas A, Ahmed MA, Basaran M. Efficacy of the VIT (vincristine, irinotecan and temozolomide) regimen in adults with metastatic Ewing sarcoma. J Chemother 2023; 35:343.
  140. 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.
  141. 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.
  142. Barker LM, Pendergrass TW, Sanders JE, Hawkins DS. Survival after recurrence of Ewing's sarcoma family of tumors. J Clin Oncol 2005; 23:4354.
  143. 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; 17S.
  144. McCabe MB, Kirston L, Khan M, et al. Results of the second interim assessment of rEECur, an international randomized controlled trial of chemotherapy for the treatment of recurrent and primary refractory Ewing sarcoma (RR-ES). J Clin Oncol 2020; 38;15S.
  145. 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.
  146. 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.
  147. 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.
  148. Duffaud F, Blay JY, Le Cesne A, et al. Regorafenib in patients with advanced Ewing sarcoma: results of a non-comparative, randomised, double-blind, placebo-controlled, multicentre Phase II study. Br J Cancer 2023; 129:1940.
  149. de Alava E, Gerald WL. Molecular biology of the Ewing's sarcoma/primitive neuroectodermal tumor family. J Clin Oncol 2000; 18:204.
  150. Juergens H, Daw NC, Geoerger B, et al. Preliminary efficacy of the anti-insulin-like growth factor type 1 receptor antibody figitumumab in patients with refractory Ewing sarcoma. J Clin Oncol 2011; 29:4534.
  151. Pappo AS, Patel SR, Crowley J, et al. R1507, a monoclonal antibody to the insulin-like growth factor 1 receptor, in patients with recurrent or refractory Ewing sarcoma family of tumors: results of a phase II Sarcoma Alliance for Research through Collaboration study. J Clin Oncol 2011; 29:4541.
  152. Tap WD, Demetri G, Barnette P, et al. Phase II study of ganitumab, a fully human anti-type-1 insulin-like growth factor receptor antibody, in patients with metastatic Ewing family tumors or desmoplastic small round cell tumors. J Clin Oncol 2012; 30:1849.
  153. Schwartz GK, Tap WD, Qin LX, et al. Cixutumumab and temsirolimus for patients with bone and soft-tissue sarcoma: a multicentre, open-label, phase 2 trial. Lancet Oncol 2013; 14:371.
  154. Naing A, LoRusso P, Fu S, et al. Insulin growth factor-receptor (IGF-1R) antibody cixutumumab combined with the mTOR inhibitor temsirolimus in patients with refractory Ewing's sarcoma family tumors. Clin Cancer Res 2012; 18:2625.
  155. Scobioala S, Ranft A, Wolters H, et al. Impact of Whole Lung Irradiation on Survival Outcome in Patients With Lung Relapsed Ewing Sarcoma. Int J Radiat Oncol Biol Phys 2018; 102:584.
  156. National Comprehensive Cancer Network (NCCN). NCCN clinical practice guidelines in oncology. Available at: https://www.nccn.org/ (Accessed on April 18, 2025).
  157. Children's Oncology Group: Long-term follow-up guidelines for survivors of childhood, adolescent, and young adult cancer. http://www.survivorshipguidelines.org/ (Accessed on May 15, 2019).
  158. Meyer JS, Nadel HR, Marina N, et al. Imaging guidelines for children with Ewing sarcoma and osteosarcoma: a report from the Children's Oncology Group Bone Tumor Committee. Pediatr Blood Cancer 2008; 51:163.
  159. Cederberg KB, Iyer RS, Chaturvedi A, et al. Imaging of pediatric bone tumors: A COG Diagnostic Imaging Committee/SPR Oncology Committee White Paper. Pediatr Blood Cancer 2023; 70 Suppl 4:e30000.
  160. Huang T, Li F, Yan Z, et al. Effectiveness of 18F-FDG PET/CT in the diagnosis, staging and recurrence monitoring of Ewing sarcoma family of tumors: A meta-analysis of 23 studies. Medicine (Baltimore) 2018; 97:e13457.
  161. Marina NM, Liu Q, Donaldson SS, et al. Longitudinal follow-up of adult survivors of Ewing sarcoma: A report from the Childhood Cancer Survivor Study. Cancer 2017; 123:2551.
  162. Fuchs B, Valenzuela RG, Inwards C, et al. Complications in long-term survivors of Ewing sarcoma. Cancer 2003; 98:2687.
  163. Navid F, Billups C, Liu T, et al. Second cancers in patients with the Ewing sarcoma family of tumours. Eur J Cancer 2008; 44:983.
  164. Ginsberg JP, Goodman P, Leisenring W, et al. Long-term survivors of childhood Ewing sarcoma: report from the childhood cancer survivor study. J Natl Cancer Inst 2010; 102:1272.
  165. Hamilton SN, Carlson R, Hasan H, et al. Long-term Outcomes and Complications in Pediatric Ewing Sarcoma. Am J Clin Oncol 2017; 40:423.
  166. Burdach S, van Kaick B, Laws HJ, et al. Allogeneic and autologous stem-cell transplantation in advanced Ewing tumors. An update after long-term follow-up from two centers of the European Intergroup study EICESS. Stem-Cell Transplant Programs at Düsseldorf University Medical Center, Germany and St. Anna Kinderspital, Vienna, Austria. Ann Oncol 2000; 11:1451.
  167. 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.
  168. Bhatia S, Krailo MD, Chen Z, et al. Therapy-related myelodysplasia and acute myeloid leukemia after Ewing sarcoma and primitive neuroectodermal tumor of bone: A report from the Children's Oncology Group. Blood 2007; 109:46.
  169. Rodriguez-Galindo C, Poquette CA, Marina NM, et al. Hematologic abnormalities and acute myeloid leukemia in children and adolescents administered intensified chemotherapy for the Ewing sarcoma family of tumors. J Pediatr Hematol Oncol 2000; 22:321.
  170. Caruso J, Shulman DS, DuBois SG. Second malignancies in patients treated for Ewing sarcoma: A systematic review. Pediatr Blood Cancer 2019; 66:e27938.
  171. Kuttesch JF Jr, Wexler LH, Marcus RB, et al. Second malignancies after Ewing's sarcoma: radiation dose-dependency of secondary sarcomas. J Clin Oncol 1996; 14:2818.
Topic 7740 Version 58.0

References