Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Orthop. Sep 18, 2016; 7(9): 527-538
Published online Sep 18, 2016. doi: 10.5312/wjo.v7.i9.527
Management of Ewing sarcoma family of tumors: Current scenario and unmet need
Bivas Biswas, Sameer Bakhshi, Department of Medical Oncology, Dr.B.R.A.I.R.C.H, All India Institute of Medical Sciences, New Delhi 110029, India
Author contributions: Biswas B performed the literature search, critical analysis, wrote the paper recorded the audio core tip; Bakhshi S reviewed and edited the paper and done the critical analysis.
Conflict-of-interest statement: No potential conflicts of interest to declare for each author.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Dr. Bivas Biswas, Assistant Professor, Department of Medical Oncology, Dr.B.R.A.I.R.C.H, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India. bivasbiswas@gmail.com
Telephone: +91-98-30922005 Fax: +91-11-26588663
Received: April 27, 2016
Peer-review started: April 28, 2016
First decision: June 16, 2016
Revised: June 21, 2016
Accepted: July 11, 2016
Article in press: July 13, 2016
Published online: September 18, 2016

Abstract

Ewing sarcoma family tumors (ESFT) are heterogeneous, aggressive group of disease with peak incidence in adolescent and young adults. The outcome has been improved dramatically from 10% with surgery and radiotherapy alone to 65%-70% now, in localized disease, with the introduction of chemotherapy. Chemotherapy regimen evolved from single agent to multiagent with effort of many cooperative clinical trials over decades. The usual treatment protocol include introduction of multi-agent chemotherapy in neoadjuvant setting to eradicate systemic disease with timely incorporation of surgery and/or radiotherapy as local treatment modality and further adjuvant chemotherapy to prevent recurrence. Risk adapted chemotherapy in neoadjuvant and adjuvant setting along with radiotherapy has been used in many international collaborative trials and has resulted in improved outcome, more so in patients with localized disease. The role of high dose chemotherapy with stem cell rescue is still debatable. The outcome of patients with metastatic disease is dismal with long term outcome ranges from 20%-40% depending on the sites of metastasis and intensity of treatment. There is a huge unmet need to improve outcome further, more so in metastatic setting. Novel therapy targeting the molecular pathways and pathogenesis of ESFT is very much required. Here we have discussed the current standard of management in patients with ESFT, investigational targeted or novel therapies along with future promises.

Key Words: Outcome, Review, Chemotherapy, Ewing sarcoma, Targeted therapy, Radiotherapy, Prognostic factors

Core tip: Ewing sarcoma family tumors are a heterogeneous and aggressive group of disease of bone and soft tissue in childhood. The outcome has improved with introduction of chemotherapy and multimodality management. But, the prognosis of patients with metastatic disease is dismal. Novel targeted therapies are investigational and may offer some hope in future, especially in metastatic setting. In this review we have discussed current treatment modality, prognostic factors, ongoing trials and novel investigational therapies.



INTRODUCTION

Ewing sarcoma families of tumors (ESFT) are a heterogeneous and aggressive group of disease of bone and soft tissue that includes classical Ewing sarcoma, peripheral primitive neuroectodermal tumor and Askin tumor. It is the 2nd most common primary malignant bone tumor (34%) after osteosarcoma[1] with peak in 2nd decade of life, though approximately 20% to 30% of all cases occur in 1st decade. Over the last four decades, the survival has been improved from 10% with radiotherapy alone to near 70%, in localized disease, with the introduction of chemotherapy and multimodality approach. This improvement in outcome has been achieved after many international collaborative clinical trials and close liaison between orthopedic surgeons, adult and pediatric oncologists, radiation oncologist, pediatric surgeons, biomedical engineers, pathologists and radiologists as well as invention of better diagnostic imaging, radiotherapy techniques and prosthesis.

With the improvement in outcome and more number of long-term survivors, the focus is now on to minimize toxicities, such as chemotherapy related, radiotherapy related and surgery related long-term complication without compromising the oncological outcomes. The multidisciplinary approach with risk adapted chemotherapy and local treatment (surgery and/or radiotherapy) has been used in recent times in many collaborative trials to minimize the overtreatment and thus treatment related side effects with maintaining high cure rates. This approach is the current standard of care in maximum institutions all over the globe.

Even with current armamentarium the outcome of ESFT patients with metastatic disease is dismal with cure rate varying between 20%-40%[2,3] and even less in those with recurrent/refractory diseases. The current emphasis is to improve the survival outcome in ESFT patients with metastatic disease and also in the recurrent setting. There is a vacuum in novel and targeted therapies as compared to adult solid tumors, and there lies a huge unmet need to improve the outcome of poor risk ESFTs.

Researchers across the world have tried to understand the pathogenesis of ESFT along with the molecular downstream pathways enhancing the survival of ESFT tumor cells. The primary focus was on EWS-FLI1 fusion oncogene, and other similar fusions that were thought to drive the oncogenic pathway in ESFT, but targeted therapy by blocking its product has not resulted into any meaningful clinical outcome[4,5]. In this review we have discussed the current standard of treatment-chemotherapy, local treatment modalities, role of high dose chemotherapy, and salvage treatment along with novel targeted therapies under investigation and potential future promises.

WHAT ARE THE CLINICALLY RELEVANT PROGNOSTIC FACTORS?

Many international cooperative trials has been performed over last four decades to improve the outcome of ESFTs and further analysis of those studies revealed many prognostic factors that predicted differential outcomes and successively helped in designing tailored clinical trials to optimize the treatment strategies depending on the risk group and to decrease over treatment and treatment related side effects. Further refinements and validation of those prognostic factors (clinic-pathological and treatment related factors) has been done in further studies and in routine clinical practices. Amongst all the clinic-pathological and treatment related factors, presence of metastasis at baseline is the strongest prognostic factor and has been proven in all clinical studies and routine clinical practices. The prognosis also depends on burden of metastasis and site of metastasis. Spectrum of outcome varies from worst with bone marrow metastasis (3-year EFS of < 10%) with non-pulmonary metastasis in the middle and single pulmonary metastasis having the best outcome (3-year EFS of 40%-50%)[6-10]. No research group or study had prospectively evaluated the prognostic significance of burden of metastasis until recently. Study from our center in ESFT patients with metastatic disease found hypoalbuminemia (< 3.5 g/dL) as a novel and independent poor prognostic factor to affect outcome[2]. The recent EuroEwing 99 (EE99) systematically risk stratified the metastatic group and the 3rd randomized arm (disseminated multifocal ESFT) identified novel prognostic factors, such as - age at diagnosis > 14 years, presence and number of bone metastasis, number of pulmonary metastasis and bone marrow involvement and also developed a prognostic score to predict differential outcome ranging from 8%-40%[10].

Systemic symptom (fever and weight loss) is a poor prognostic factor along with high lactate dehydrogenase level that denotes tumor burden. These two prognostic factors has been used to risk stratify ESFT patients to tailor therapy. Tumor size and tumor volume is well established prognostic across the clinical studies with tumor size > 8 cm[11] and tumor volume > 200 mL[12,13] is poor prognostic and has been used in all clinical trials for risk adapted therapies. Site of primary tumor is also of prognostic significance with axial primary especially pelvic location is poor prognostic. Both tumor size and pelvic primary has been proven as poor prognostic in our institutional experience[2,14]. But recently histological response to neoadjuvant chemotherapy (poor response has been defined as > 10% viable tumor cells as per Salzer-Kuntschik grading system[13]) has been emerged as the strongest prognostic factor overriding tumor size, tumor volume or tumor location. The recent EE99 study risk stratified ESFT patients in respect to histological response to chemotherapy to tailor therapy[15]. WBC count has been emerged as independent prognostic factor in our experience of ESFT[14,16-18] treated with uniform chemotherapy protocol with high WBC (> 11000/μL) having poor outcome. It may signify micrometastatic disease and inflammatory nature of the disease and will require further validation in a prospective study.

Collaborative efforts have been made to identify potential biomarker in this rare aggressive tumor. Early retrospective studies[19,20] reported prognostic significance of different transcripts (EWS-ETS fusion types) but failed to do so in prospective studies[21]. High throughput methods has revealed gene copy number variations[22], such as - 1q-, 18q-, 20-, 16q+ along with mutation in TP53, CDKN2A and STAT2 and concurrent presence of STAT2 and TP53 reported as poor prognostic[23]. Detection of disseminated tumor cells in blood and bone marrow[24] in localized patients (up to 20%) found to poor prognostic and its detection after completion of therapy (minimal residual disease) by flow cytometry or other sensitive method can tailor therapy, detect early recurrence and can be of future prognostic significance[20].

WHAT IS THE ROLE OF BIO-IMAGING?

Histological response to neoadjuvant chemotherapy is the strongest prognostic factor in localized ESFT and metastatic disease is the strongest one in whole cohort of ESFT. Efforts have been made to predict metastatic potential of ESFT in view of its aggressive nature and to escalate therapy in case of poor responder before initiation of local therapy. 18F-FDG PET is a relatively non-invasive test that can escape invasive diagnostic metastatic work-up like bone marrow aspiration/biopsy and tissue diagnosis in case of doubtful lung metastasis if it predicts site of metastases, and also post chemotherapy response assessment before the histological response is assessed. Retrospective studies have shown it can predict response to neoadjuvant chemotherapy and have a high concordance rate with the histological response by measuring reduction in standardized uptake value[25,26] and also reduction in metabolic tumor volume[27]. But no prospective study has been done in regards to its predicting power of detecting metastasis and to predict response to chemotherapy before its utilization in routine clinical practice.

LOCALIZED DISEASE
What chemotherapy, what intensity, and how long?

Historically, in early 1970s the outcome of ESFT with radiotherapy and surgery alone was dismal with only < 10% patients surviving and all invariably experienced relapse within 2 years. With the introduction of chemotherapy the outcome have improved drastically to 70% cure rate in localized disease. The evolution of chemotherapy started from single agent vincristine to multiagent chemotherapy (VAC - vincristine, actinomycin D and cyclophosphamide) and from adjuvant to neoadjuvant setting (Table 1). Then comes the role of anthracyclines and addition of doxorubicin resulted in improved survival in 342 localized ESFT along with additional benefit of whole lung irradiation (WLI) in metastasis prevention in Intergroup Ewing’s Sarcoma Study 1 (IESS-1)[28]. The value of doxorubicin further potentiated in IESS-II which showed that high dose intermittent doxorubicin is better than low dose continuous therapy in 214 non-pelvic localized ESFT[29]. The landmark United States intergroup study (INT0091) by Grier et al[6] showed additional benefit of ifosfamide and etoposide (IE) in addition to vincristine, doxorubicin, cyclophosphamide (VDC) in localized patients with ESFT after the beneficial effect of IE has been demonstrated in recurrent setting[30], but the trial failed to demonstrate any benefit of IE in a relatively smaller numbers of patients with metastatic disease. Replacement of ifosfamide with cyclophosphamide has showed conflicting results[31] and EICESS-92 trial showed similar result with four drug chemotherapy regimen in a relatively underpowered study of 155 localized ESFT along with non-significant advantage of addition of etoposide in metastatic disease[7]. In a different strategy to improve outcome by intensifying the alkylator dose and thus reducing the chemotherapy duration to 30 wk as compared to standard 48 wk with VDC-IE failed to improve outcomes in a United States intergroup trial (INT 154)[32]. But subsequent Children Oncology Group (COG) study used dose compressed study of 3-weekly vs 2-weekly VDC-IE with use of filgrastim, thus maintaining the dose intensity, and demonstrated superior outcome in 2-weekly arm (5-year EFS of 73% vs 65%, P = 0.048)[33] without any increasing toxicity in the experimental arm. The latest and largest trial in ESFT is the ongoing EE99 trial that compared cyclophosphamide with ifosfamide in standard risk ESFT and the early result showed equivalent outcome in both arm with pending long-term toxicity results[15] (Table 2). The role of high dose chemotherapy with autologus stem cell rescue not well studied in localized disease as consolidation in comparison to continuation or maintenance chemotherapy. Two cooperative trials[34,35] used BuMel conditioning regimen with stem cell rescue as consolidation therapy in a non-randomized manner in high risk localized disease (defined as poor histologic response to chemotherapy) and showed improved survival as compared to historical control and similar to that of standard risk patients. Following that result the recent ongoing EE99 trial randomized (arm 2) BuMel based high dose chemotherapy vs continuation of standard chemotherapy after VIDE induction chemotherapy in high risk localized patients and the result is still pending (Table 2).

Table 1 Randomized studies of chemotherapy in upfront treatment of Ewing sarcoma family tumors.
StudyPatients (n)Intervention1OutcomePComments
IESS I[78]Localized (342)(1) VAC5-yr RFS 24%Beneficial of doxorubicin and benefit of lung RT
(2) VACD60%--
(3) VAC + Lung RT44%
IESS II[29]Non-pelvic, localized (214)VACD5-yr RFS0.04Intermittent, high dose better
(1) Intermittent, high dose (3 weekly)73%
(2) Continuous, moderate dose (weekly)56%
POG-CCSG INT-0091[6]Localized (398)(1) VDC5-yr RFS 54%0.005IE is beneficial in addition to VDC in localized but not in metastatic disease
(2) VDC + IE69%
Metastatic (120)(1) VDC22%NS
(2) VDC + IE22%
POG-CCSGLocalized (478)5-yr EFSNSDose intensification not effective
INT-154[32](1) VDC + IE (standard)70%
(2) VDC + IE (intensified)72%
COG AEWS0031[33]Localized (568)(1) VDC + IE (3 weekly)5-yr EFS 65%0.053-weekly better than 2-weekly with no increase in toxicity
(2) VDC + IE (2 weekly)73%
EICESS92[7]n = 155SR (localized and < 100 mL) 4#VAIA → 8# VAIA vs 8#VACD3-yr EFS 73% vs 74%NSCyclophosphamide and ifosfamide is similar in efficacy in SR patients
n = 492HR (metastatic, > 100 mL)47% vs 52%0.12No benefit of etoposide in HR patients
14# VAIA vs 14#EVAIA
Euro-Ewing 99[49]Detailed in Table 2
Table 2 Euro-Ewing 99 trial design and details.
Randomized armPatientsnRandomization3-yr EFS
Arm 1SR, Localized (good histologic response, < 200 mL + RT)8566#VIDE + 1#VAI f/b 7#VAI vs 7#VAC78% vs 75%
Arm 2HR, Localized (poor histologic response, ≥ 200 mL and RT alone)---6#VIDE + 1#VAI f/b 7# VAI vs BuMel (n = 281)45% (BuMel)
Lung metastasis only---6#VIDE + 1#VAI f/b 7# VAI + WLI vs BuMel---
Arm 3Extrapulmonary metastasis---6#VIDE + 1#VAI f/b BuMel/TreoMel vs clinical trial---
What local therapy and when?

The outcome of ESFT with surgery or radiotherapy alone was dismal and with introduction of polychemotherapy regimen improved the outcome dramatically by eradicating systemic micrometastsis in localized disease. But, chemotherapy alone can’t eradicate ESF tumor cells and timely incorporation of local therapy either surgery and/or radiotherapy is crucial for optimum management and to produce high cure rate. The approach of local therapy evolved over time with better understanding of the disease biology, better radiation technique, invention of newer engineered prosthesis, better imaging modalities and more information of therapy related complications. The choice of local treatment influenced by multiple factors, such as age of the patients, site and size of the tumor, local extent of the tumor, clinic-radiological response to chemotherapy, expertise and experience of the treating institution and surgeon and patient’s choice, etc. The different modalities of local treatment include - surgery (amputation, limb salvage or organ sparing surgery) with or without adjuvant radiotherapy, radical radiotherapy, pre-operative radiotherapy, extracorporeal radiotherapy. No prospective formal comparison done between surgery and radiotherapy as local treatment. Across the clinical trials and institutional experience, surgery done better in terms of long term outcome (both local and systemic control), and thus a formal comparison between this two seems not feasible in future.

ESFT is a radiosensitive tumor, but the long term outcome was < 10% with high incidence of local[36] and systemic recurrence. Surgery slowly replaced radiotherapy in view of better local control rate, lesser long-term complication compared to radiotherapy and with invention of better bone replacement materials (endoprosthesis, bone cement, allograft, vascularized autograft) the rate of limb sparing surgery has increased as a norm now-a-days[36,37]. But, the surgery is also associated with long-term complications, such as-post-op infection, limb-length discrepancy, fractures, etc. Maintaining limb length is difficult in growing children and expandable endoprosthesis comes handy in this scenario with its increasing uses.

The surgical resection principle depends on the respectability, size and sites the tumor and its operability after chemotherapy. A functional limb or organ is the norm after any local treatment modality. Amputation is rarely indicated and limb salvage or organ sparing surgery should be tried whenever feasible. Surgical resection should be tried whenever a marginal or wide resection is feasible as the outcome seems to be superior to radical radiotherapy as local control[11,32,36,38-42]. Intralesional or debulking surgery should be avoided as the outcome is not superior over radiotherapy alone[41].

Definitive or radical radiotherapy as local treatment modality used where non-mutilating, wide local excision is not feasible with a functional organ, more so in axial primary, such as - head and area, spine, pelvic primary and in very large lesion not amenable to curative surgery even after neoadjuvant chemotherapy, and in case of a metastatic disease, etc. The recommended dose varies from 55 to 60 Gy in standard fractionation with 2 cm margin that should include original biopsy scar[39]. Care should be taken to avoid toxicity to adjacent normal organ and newer techniques, such as - intensity modulated radiotherapy, image guided planning or proton therapy, etc. should be used in more cases[40]. Data on use of post-operative radiotherapy is mostly debated in view of conflicting results from observational studies[41-43]. The only clear cut indication is that of intralesional surgery[32] where further resection with remaining functional organ is not feasible. Many European institutions use adjuvant radiotherapy in patients with poor histologic response to chemotherapy (> 10% viable tumor cells) and in a soft tissue primary. Recent EE99 study also showed beneficial effect of adjuvant radiotherapy even in good responder and thus broadens its future use, though the risk-benefit ratio to be calculated stringently with long-term radiotherapy related complication. Pre-operative radiation therapy is a good viable future alternative where a complete resection looks not feasible after chemotherapy and thus can sterilize the compartment before reconsideration of surgery after radiotherapy, like in pelvic or spinal primary[40].

What are the current and future studies?

Many collaborative studies are ongoing in ESFT to find out the most appropriate risk-stratified approach for improving outcome with incorporation of high dose chemotherapy (Ewing 2008 and Italian ISG/AIEOP EW-1 study), introduction of metronomic chemotherapy as maintenance (COG AEWS1031 study), role of zoledronic acid (Ewing 2008 and Euro-Ewing 2012 study), optimum use of post-operative radiotherapy along with dose intensified approach (Euro-Ewing 2012 study), and comparison of vincristine, ifosfamide, doxorubicin and etoposide (VIDE) standard therapy in Europe vs VAC-IE (Euro-Ewing 2012 study).

METASTATIC DISEASE
Is site of metastasis prognostic?

ESFTs are an aggressive group of disease with high incidence of metastasis at presentation ranging from 20%-40% across different clinical trials and observation studies[2,3,44-47]. Outcome of patients with lung metastasis was better as compared to those with bone metastasis or combined or to those with bone marrow involvement and single metastasis done better as compared to multiple metastases[7]. The recent EE99 trial also revealed the presence and number of bone metastasis, presence and number of lung metastasis and bone marrow involvement as prognostic factors[10]. More prospective studies are needed to define the prognostic nature of site and number of metastasis in a more stringent manner to tailor and intensify therapies.

Is the treatment same as localized disease?

The treatment protocol is similar like in those with localized disease with curative intent - neoadjuvant chemotherapy followed by institution of local therapy (surgery and/or radiotherapy) and further maintenance therapy or consolidation with high dose chemotherapy or an investigation novel agents/targeted therapy. Many agents in combination or with total body irradiation have been used as consolidation in metastatic disease. Definitive radiotherapy used more as compared to surgery in view of high residual disease at metastatic disease after neoadjuvant chemotherapy and a more conservative approach especially in those with disseminated metastases or with bone marrow involvement.

LOCAL TREATMENT: WHAT AND WHEN?

Local treatment usually incorporated after initial 5-6 cycles of chemotherapy and if there is good response to chemotherapy in both local site and metastatic site(s). WLI has been tried in clinical studies in patients with lung only metastasis[48] with 5-year EFS up to 50% along with conventional chemotherapy compared to similar results with high dose chemotherapy without WLI[8]. The EE99 study randomized (arm 2) patients with lung only metastasis VIDE chemotherapy followed by VAI as maintenance plus WLI vs BuMel based high dose chemotherapy as consolidation and the result is pending[49]. Local excision or radiation therapy has been tried in patients with bone metastases in retrospective studies with favorable outcome[50]. Resection of pulmonary metastasis failed to show efficacy in two retrospective studies[51,52] but require validation in a prospective manner especially in those with single lung metastasis.

WHAT IS THE ROLE OF HIGH DOSE CHEMOTHERAPY?

Dose intensity[33] and high dose chemotherapy[34,35] found to be effective and improved outcome in patients with localized disease especially in high risk disease. With the principle of dose intensity and dose density high dose chemotherapy with stem cell rescue has been tried in many randomized and non-randomized study to improve outcome in metastatic disease with mixed results. Single agent high dose melphalan failed to improve outcome[53]. In a single arm study, BuMel conditioning in metastatic patients showed 5-year EFS of 52% in patients with lung metastasis and 36% in those with bone metastases[8]. Subsequently many clinical trials have tried high dose chemotherapy in metastatic patients and showed mixed outcome as compared to conventional chemotherapy only (Table 3). Three large studies using high dose chemotherapy - the EE99 study randomized ling only metastatic group in to BuMel based high dose therapy vs WLI along with conventional chemotherapy after initial VIDE chemotherapy and the mature result is pending[49]. The third arm randomized patients with extrapulmonary metastasis in BuMel or TreoMel based high dose chemotherapy vs investigational agent after standard VIDE based induction chemotherapy regimen and the early results showed 3-year overall survival[10]. In study by Italian and Scandinavian sarcoma study group used BuMel conditioning with WLI in patients with lung only metastasis or single bone metastasis with 5-year EFS of 43%[9]. The Ewing 2008 trial randomized patients with extrapulmonary metastasis after VAC chemotherapy to TreoMel based high dose chemotherapy vs continuation of VAC and the result is pending. On the contrary a study by Children’s Cancer Group failed to show any improvement in outcome of patients with extrapulmonary metastases after VAC-IE based chemotherapy followed by high dose chemotherapy with melphalan, etoposide and total body irradiation[54].

Table 3 Selected studies of high dose chemotherapy with stem cell rescue in Ewing sarcoma family tumors.
Study (type)Disease settingnConditioningConclusion
CESS (restrospective)[78]Recurrent or progressive disease (HDC after CR or PR)73BuMel (15) TreoMel (38) Other (20)Early relapse - poor prognostic
Société Française des Cancers de l’Enfant (prospective)[8]Metastatic at diagnosis75BuMelBeneficial for lung only or bone metastases
Italian Sarcoma Group/Scandinavian Sarcoma Group IV Protocol (phase II)[9]Metastatic at diagnosis (lung or single bone metastasis)79BuMel ± TBIHDC with WLI is effective
Italian Sarcoma Group/Scandinavian Sarcoma Group III Protocol (prospective)[34]High risk, localized126BuMelEffective and feasible in patients with PR after chemotherapy
Euro-Ewing 99 (prospective)[10]Metastatic at diagnosis169BuMel (123) Mel (15) Others (20)Effective in Bone and Bone marrow metastases
EBMT registry (retrospective)[79]Metastatic and HR, localized (n = 2411) Recurrent (n = 719)3695Heterogeneous regimensPrognostic factors: Age, response to treatment, BuMel regimen
RELAPSED AND RECURRENT DISEASE

The progress of improved outcome in ESFT is attenuated by high incidence of recurrence (local and/or systemic) and remains the main challenge in multidisciplinary management of this aggressive malignancy, especially in those with metastatic disease. The incidence of local or distant relapse is approximately 20%-25% in the published literature[45,55] and can reach up to 40% in metastatic setting[2,47]. There is no standard established salvage therapy exist in recurrent disease with dismal outcome of 20% in case of localized relapse[56]. Few chemotherapy agents showed activity in recurrent disease with moderate but short lasting response rate - topotecan and cyclophosphamide[57], ifosfamide in combination with carboplatin and etoposide[58], irinotecan and temozolamide[59]. Gemcitabine and docetaxel[60] combination failed to show any clinically meaningful activity in recurrent ESFT. The mostly studied chemotherapeutic regimen in recurrent ESFT is of irinotecan and temozolamide from a phase 1 trial and from few institutional experiences[61-65], like in other pediatric solid tumors. This combination used protracted course of irinotecan with synergistic activity of temozolamide and produced overall response rate of 25%-60% (Table 4). No prospective study to evaluate role of high dose chemotherapy has been done so far in recurrent ESFT and retrospective institutional data[66,67] is available with modest activity. A well selected prospective trial in needed in this regards. The Euro-Ewing consortium started a randomized phase II/III four arm study (cyclophosphamide-topotecan vs gemcitabine-docetaxel vs high dose ifosfamide vs irinotecan-temozolamide) in recurrent ESFT and the trial will complete recruitment in 2019. A huge vacuum exist in effective salvage therapy of recurrent/refractory ESFT and novel targeted therapy is very much need to fill that unmet need. Future therapeutic trials will eye on combination of chemotherapy with targeted therapy in recurrent/refractory as well as metastatic disease.

Table 4 Data on Irinotecan-temozolamide salvage regimen in recurrent/refractory Ewing sarcoma family tumors.
Ref.nIrinotecan scheduleORR (n)Toxicity (grade 3 and 4)
Kurucu et al[65]2020 mg/m2 (D1-5 and D8-D12)55% (11)Diarrhea - 9.2%
Neutropenia - 11.3%
McNall-Knapp et al[64]2515 mg/m2vs 20 mg/m2 (D1-5 and D8-D12)120% (5)Diarrhea - 5%
Raciborska et al[63]2250 mg/m2 (D1-D5)150% (12)Diarrhea - 15%
Hematological - 10%
Wagner et al[62]1610-20 mg/m2 (D1-5 and D8-D12) - 3 weekly vs 4 weekly25% (4)Diarrhea - 11%
Casey et al[61]2020 mg/m2 (D1-5 and D8-D12)63%Diarrhea - 4.5%
Hematological - 22%
NEW TARGETS AND TARGETED THERAPIES

With the plateau of survival with the current conventional chemotherapy and stem cell transplantation in metastatic and recurrent setting of ESFT the urgent need for novel targeted therapy should match the ongoing research in understanding the biology of ESFT with revelation of more and more oncologic pathways and targets. Various drugs has been discovered and tested in preclinical and clinical studies in ESFT by targeting EWS-FLI 1 fusion protein, the hall mark of ESFT-CD99, angiogenic pathways (VEGF and its receptor), mammalian target of rapamycin (mTOR) and insulin-like growth factor-1 (IGF1) pathways, the osteoclastic-osteoblastic homeostasis and bone microenvironment, enzymatic pathways (poly ADP-ribose polymerase 1 - PARP1), and GD2 ganglioside pathways.

The mostly studied targeted therapy used in ESFT is by inhibiting EWS-FLI1 transcriptional complex. Many agents have been discovered that directly or indirectly inhibit EWS-FLI1 pathways - small-molecule YK-4-279 is in pre-clinical phase that directly inhibit interaction of EWS-FLI1 and RNA helicase A[68], certain chemotherapy agents (doxorubicin, etoposide and cytarabine), midostaurin (broad spectrum protein kinase inhibitor)[69], mithramycin (antibiotic inhibiting RNA synthesis)[70], and the early clinical studies failed to show any clinical benefit of inhibiting EWS-FLI1 pathway in spite of being the driver in ESFT carcinogenesis.

Anti-angiogenic approach has been used in recurrent tumors and thus inhibiting the tumor growth and its metastatic potential. Bevacizumab (monoclonal antibody against VEGFR) has been tested in phase II study by COG in combination with chemotherapy (vincristine, topotecan and cyclophosphamide) in recurrent setting with pending results (NCT00516295). Pazopanib (a small molecule multi-kinase inhibitor including VEGFR) has been tested in a phase I trial[71] in refractory pediatric solid tumor and now being tested in a phase II study by COG that include ESFT and other sarcomas (NCT01956669). Regorafenib (a small molecule multi-kinase inhibitor like pazopanib) also being tested in refractory sarcomas including ESFT (NCT02048371).

ESFT is characterized by osteolytic bone lesion with extensive soft tissue component which is marked by osteoclastic activity and interaction between RANK and its ligand - RANKL[72]. RANKL facilitates osteoclastic activity, with bone resorption and destruction, and tumor growth. Zoledronic acid, a bisphosphonate inhibit osteoclastic activity and its migration along with inhibition of RANK, showed anti-tumor activity in in-vivo model of ESFT and the effect was accentuated by addition of ifosfamide[73]. Ewing 2008 and Euro-Ewing 2012 trial is evaluating the benefit of zoledronic after combining with chemotherapy in localized ESFT.

IGF1 receptor (IGF1R) plays an important role downstream to EWS-FLI1 for cell survival, angiogenesis and metastasis. But, disappointing results with anti-IGF1R monoclonal antibody let to stoppage of further study of this novel agent due to dramatic but very short lasting response in refractory ESFTs. The cause of this early resistance is not fully understood though up-regulation of IGF1R or mTOR has been postulated[74]. The COG future trial has planned combination of VAC-IE with anti-IGF1R antibody in metastatic disease to overcome this resistance.

The other potential targeted therapies include - PARP1 inhibitor olaparib in combination with temozolamide showed in-vivo and in-vitro activity[75], anti-GD2 ganglioside (a neuroendocrine marker present in ESFT cells) chimeric antigen[76], and anti-CD99 monoclonal antibody[77] are in preclinical study periods.

FERTILITY ISSUE: A IMPORTANT BUT OFTEN IGNORED ISSUE

ESFT is a disease of young adolescent age group along with many patients in reproductive age group. All three treatment modality in ESFT affects gonadal and reproductive function in these patients. Multi-agent chemotherapy, especially with alkylators and anthracyclines, and radiotherapy are the two major culprits in this scenario due to their goandotoxic effects. Many strategies have been taken to minimize or counter the gonadotoxic effects of cancer treatment especially with chemotherapy and radiotherapy.

Sperm cryopreservation is the standard and effective modality of fertility preservation in case of male patients, whenever indicated. Toxicity to ovarian follicle and embryo is a special scenario especially with pelvic radiotherapy. In a COG study[78], 8.3% of female cancer survivors experienced acute ovarian failure, defined as loss of ovarian function with 5-years of cancer diagnosis. Radiotherapy toxicity to ovary depends on age of the patients, concurrent ovariotoxic chemotherapy, dose and fractionation of radiotherapy and volume of radiation field. Cost, experience, expertise, infrastructure and low success rate are the main logistic issue in female fertility preservation.

Embryo crypreservation is an option for fertility preservation in female patients with a male partner, whereas cryopreservation of mature or immature oocytes is a viable option for those refuse to opt for a sperm donor. Cumulative pregnancy rate up to 40% has been reported with the former technique[79] but with a modest success with the later technique[80]. Cryopreservation of ovarian tissue is the only measures of fertility preservation in very young girls, and recent reports of successful pregnancy have been described in literature[81,82].

Ovarian transposition or ovariopexy is the method to preserve ovarian function in patients receiving pelvic radiotherapy but high rate of permanent cessation of ovarian function has been reported in earlier series[83]. Intensity modulated radiotherapy or 3-D conformal CT planning in radiotherapy can minimize the gonadal toxicity in case of very small pelvic tumor or a tumor distant to ovary.

Protective role of concurrent GnRH-a has been described in literature during combination gonadotoxic chemotherapy. In study of lymphoma patients aged 15-40 years, GnRH-a group resumed menstruation in 93.7% cases within 3-8 mo of chemotherapy as compared to 39% in historical controls of same disease group who didn’t received GnRH-a[84]. In important issue remains in fertility preservation where the ovary itself is the primary site of disease in ESFT and rarely ESFT can metastasize to the ovary[85-87].

CONCLUSION

ESFTs are a rare aggressive tumor with high rate of metastasis at presentation and high incidence of recurrence. The outcome of those with localized improved to 70% after multimodality approach mainly by better understanding of disease biology, risk adapted chemotherapeutic approach, timely incorporation of local therapy, and improvement in technology. But, the outcome of those with metastatic and recurrent disease is dismal and no significant advancement has been made in these patients to improve outcome in last four decades. The overall improvement in outcome of ESFT has been made through the tremendous efforts of researcher, clinicians all over the world, better liaison between all the stakeholders of treating team, and collaborative international research in a huge number of cases. The main challenge now remains in preventing recurrence, preventing drug resistance, reducing therapy related long-term toxicities and improving outcome in those with metastatic and relapsed/recurrent disease. No potential biomarker has been identified so far to predict therapeutic efficacy of chemotherapeutic agents and predicting recurrences. The future hope lies in finding useful biomarker, better understanding of disease biology and chemotherapy resistance of ESFT cells, proper designing and execution of targeted therapies currently going under clinical trials, better use of high throughput method to detect novel driver mutations/pathways and potential targets. Better selection of risk group and designing of trials combining chemotherapeutic agents with targeted therapies to bypass drug resistance along with judicious use of high dose chemotherapies, selection of more non-toxic agents with high efficacy and broad therapeutic windows will help to improve future outcomes in expense of decreased treatment related long-term toxicities and good quality of life in survivors.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Orthopedics

Country of origin: India

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): B, B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): E

P- Reviewer: Blumenfeld Z, Mocellin S, Romani A, Shimoyama S S- Editor: Ji FF L- Editor: A E- Editor: Li D

References
1.  Damron TA, Ward WG, Stewart A. Osteosarcoma, chondrosarcoma, and Ewing’s sarcoma: National Cancer Data Base Report. Clin Orthop Relat Res. 2007;459:40-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 332]  [Cited by in F6Publishing: 345]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
2.  Biswas B, Rastogi S, Khan SA, Shukla NK, Deo SV, Agarwala S, Sharma DN, Thulkar S, Vishnubhatla S, Pathania S. Hypoalbuminaemia is an independent predictor of poor outcome in metastatic Ewing’s sarcoma family of tumours: a single institutional experience of 150 cases treated with uniform chemotherapy protocol. Clin Oncol (R Coll Radiol). 2014;26:722-729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 27]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
3.  Cotterill SJ, Ahrens S, Paulussen M, Jürgens HF, Voûte PA, Gadner H, Craft AW. Prognostic factors in Ewing’s tumor of bone: analysis of 975 patients from the European Intergroup Cooperative Ewing’s Sarcoma Study Group. J Clin Oncol. 2000;18:3108-3114.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Delattre O, Zucman J, Plougastel B, Desmaze C, Melot T, Peter M, Kovar H, Joubert I, de Jong P, Rouleau G. Gene fusion with an ETS DNA-binding domain caused by chromosome translocation in human tumours. Nature. 1992;359:162-165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1352]  [Cited by in F6Publishing: 1310]  [Article Influence: 40.9]  [Reference Citation Analysis (0)]
5.  Gaspar N, Di Giannatale A, Geoerger B, Redini F, Corradini N, Enz-Werle N, Tirode F, Marec-Berard P, Gentet JC, Laurence V. Bone sarcomas: from biology to targeted therapies. Sarcoma. 2012;2012:301975.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
6.  Grier HE, Krailo MD, Tarbell NJ, Link MP, Fryer CJ, Pritchard DJ, Gebhardt MC, Dickman PS, Perlman EJ, Meyers PA. Addition of ifosfamide and etoposide to standard chemotherapy for Ewing’s sarcoma and primitive neuroectodermal tumor of bone. N Engl J Med. 2003;348:694-701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 942]  [Cited by in F6Publishing: 860]  [Article Influence: 41.0]  [Reference Citation Analysis (0)]
7.  Paulussen M, Craft AW, Lewis I, Hackshaw A, Douglas C, Dunst J, Schuck A, Winkelmann W, Köhler G, Poremba C. Results of the EICESS-92 Study: two randomized trials of Ewing’s sarcoma treatment--cyclophosphamide compared with ifosfamide in standard-risk patients and assessment of benefit of etoposide added to standard treatment in high-risk patients. J Clin Oncol. 2008;26:4385-4393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 196]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
8.  Oberlin O, Rey A, Desfachelles AS, Philip T, Plantaz D, Schmitt C, Plouvier E, Lejars O, Rubie H, Terrier P. Impact of high-dose busulfan plus melphalan as consolidation in metastatic Ewing tumors: a study by the Société Française des Cancers de l’Enfant. J Clin Oncol. 2006;24:3997-4002.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 99]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
9.  Luksch R, Tienghi A, Hall KS, Fagioli F, Picci P, Barbieri E, Gandola L, Eriksson M, Ruggieri P, Daolio P. Primary metastatic Ewing’s family tumors: results of the Italian Sarcoma Group and Scandinavian Sarcoma Group ISG/SSG IV Study including myeloablative chemotherapy and total-lung irradiation. Ann Oncol. 2012;23:2970-2976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 63]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
10.  Ladenstein R, Pötschger U, Le Deley MC, Whelan J, Paulussen M, Oberlin O, van den Berg H, Dirksen U, Hjorth L, Michon J. Primary disseminated multifocal Ewing sarcoma: results of the Euro-EWING 99 trial. J Clin Oncol. 2010;28:3284-3291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 319]  [Cited by in F6Publishing: 323]  [Article Influence: 23.1]  [Reference Citation Analysis (0)]
11.  Krasin MJ, Rodriguez-Galindo C, Davidoff AM, Billups CA, Fuller CE, Neel MD, Kun LE, Merchant TE. Efficacy of combined surgery and irradiation for localized Ewings sarcoma family of tumors. Pediatr Blood Cancer. 2004;43:229-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 50]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
12.  Göbel V, Jürgens H, Etspüler G, Kemperdick H, Jungblut RM, Stienen U, Göbel U. Prognostic significance of tumor volume in localized Ewing’s sarcoma of bone in children and adolescents. J Cancer Res Clin Oncol. 1987;113:187-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 180]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
13.  Oberlin O, Deley MC, Bui BN, Gentet JC, Philip T, Terrier P, Carrie C, Mechinaud F, Schmitt C, Babin-Boillettot A. Prognostic factors in localized Ewing’s tumours and peripheral neuroectodermal tumours: the third study of the French Society of Paediatric Oncology (EW88 study). Br J Cancer. 2001;85:1646-1654.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 136]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
14.  Biswas B, Rastogi S, Khan SA, Shukla NK, Deo SV, Agarwala S, Mohanti BK, Sharma MC, Vishnubhatla S, Bakhshi S. Developing a prognostic model for localized Ewing sarcoma family of tumors: A single institutional experience of 224 cases treated with uniform chemotherapy protocol. J Surg Oncol. 2015;111:683-689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 18]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
15.  Le Deley MC, Paulussen M, Lewis I, Brennan B, Ranft A, Whelan J, Le Teuff G, Michon J, Ladenstein R, Marec-Bérard P. Cyclophosphamide compared with ifosfamide in consolidation treatment of standard-risk Ewing sarcoma: results of the randomized noninferiority Euro-EWING99-R1 trial. J Clin Oncol. 2014;32:2440-2448.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 108]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
16.  Biswas B, Thakar A, Mohanti BK, Vishnubhatla S, Bakhshi S. Prognostic factors in head and neck Ewing sarcoma family of tumors. Laryngoscope. 2015;125:E112-E117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 24]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
17.  Biswas B, Shukla NK, Deo SV, Agarwala S, Sharma DN, Vishnubhatla S, Bakhshi S. Evaluation of outcome and prognostic factors in extraosseous Ewing sarcoma. Pediatr Blood Cancer. 2014;61:1925-1931.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
18.  Biswas B, Rastogi S, Khan SA, Mohanti BK, Sharma DN, Sharma MC, Mridha AR, Bakhshi S. Outcomes and prognostic factors for Ewing-family tumors of the extremities. J Bone Joint Surg Am. 2014;96:841-849.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
19.  Zoubek A, Dockhorn-Dworniczak B, Delattre O, Christiansen H, Niggli F, Gatterer-Menz I, Smith TL, Jürgens H, Gadner H, Kovar H. Does expression of different EWS chimeric transcripts define clinically distinct risk groups of Ewing tumor patients? J Clin Oncol. 1996;14:1245-1251.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  de Alava E, Lozano MD, Patiño A, Sierrasesúmaga L, Pardo-Mindán FJ. Ewing family tumors: potential prognostic value of reverse-transcriptase polymerase chain reaction detection of minimal residual disease in peripheral blood samples. Diagn Mol Pathol. 1998;7:152-157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 53]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
21.  van Doorninck JA, Ji L, Schaub B, Shimada H, Wing MR, Krailo MD, Lessnick SL, Marina N, Triche TJ, Sposto R. Current treatment protocols have eliminated the prognostic advantage of type 1 fusions in Ewing sarcoma: a report from the Children’s Oncology Group. J Clin Oncol. 2010;28:1989-1994.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 96]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
22.  Kovar H, Alonso J, Aman P, Aryee DN, Ban J, Burchill SA, Burdach S, De Alava E, Delattre O, Dirksen U. The first European interdisciplinary ewing sarcoma research summit. Front Oncol. 2012;2:54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
23.  Tirode F, Surdez D, Ma X, Parker M, Le Deley MC, Bahrami A, Zhang Z, Lapouble E, Grossetête-Lalami S, Rusch M. Genomic landscape of Ewing sarcoma defines an aggressive subtype with co-association of STAG2 and TP53 mutations. Cancer Discov. 2014;4:1342-1353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 334]  [Cited by in F6Publishing: 349]  [Article Influence: 34.9]  [Reference Citation Analysis (0)]
24.  Schleiermacher G, Delattre O. [Detection of micrometastases and circulating tumour cells using molecular biology technics in solid tumours]. Bull Cancer. 2001;88:561-570.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Gupta K, Pawaskar A, Basu S, Rajan MG, Asopa RV, Arora B, Nair N, Banavali S. Potential role of FDG PET imaging in predicting metastatic potential and assessment of therapeutic response to neoadjuvant chemotherapy in Ewing sarcoma family of tumors. Clin Nucl Med. 2011;36:973-977.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 43]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
26.  Hawkins DS, Schuetze SM, Butrynski JE, Rajendran JG, Vernon CB, Conrad EU, Eary JF. [18F]Fluorodeoxyglucose positron emission tomography predicts outcome for Ewing sarcoma family of tumors. J Clin Oncol. 2005;23:8828-8834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 245]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
27.  Gaston LL, Di Bella C, Slavin J, Hicks RJ, Choong PF. 18F-FDG PET response to neoadjuvant chemotherapy for Ewing sarcoma and osteosarcoma are different. Skeletal Radiol. 2011;40:1007-1015.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 52]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
28.  Nesbit ME, Perez CA, Tefft M, Burgert EO, Vietti TJ, Kissane J, Pritchard DJ, Gehan EA. Multimodal therapy for the management of primary, nonmetastatic Ewing’s sarcoma of bone: an Intergroup Study. Natl Cancer Inst Monogr. 1981;255-262.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Burgert EO, Nesbit ME, Garnsey LA, Gehan EA, Herrmann J, Vietti TJ, Cangir A, Tefft M, Evans R, Thomas P. Multimodal therapy for the management of nonpelvic, localized Ewing’s sarcoma of bone: intergroup study IESS-II. J Clin Oncol. 1990;8:1514-1524.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Carli M, Passone E, Perilongo G, Bisogno G. Ifosfamide in pediatric solid tumors. Oncology. 2003;65 Suppl 2:99-104.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Oberlin O, Habrand JL, Zucker JM, Brunat-Mentigny M, Terrier-Lacombe MJ, Dubousset J, Gentet JC, Schmitt C, Ponvert D, Carrié C. No benefit of ifosfamide in Ewing’s sarcoma: a nonrandomized study of the French Society of Pediatric Oncology. J Clin Oncol. 1992;10:1407-1412.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Granowetter L, Womer R, Devidas M, Krailo M, Wang C, Bernstein M, Marina N, Leavey P, Gebhardt M, Healey J. Dose-intensified compared with standard chemotherapy for nonmetastatic Ewing sarcoma family of tumors: a Children’s Oncology Group Study. J Clin Oncol. 2009;27:2536-2541.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 235]  [Cited by in F6Publishing: 245]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
33.  Womer RB, West DC, Krailo MD, Dickman PS, Pawel BR, Grier HE, Marcus K, Sailer S, Healey JH, Dormans JP. Randomized controlled trial of interval-compressed chemotherapy for the treatment of localized Ewing sarcoma: a report from the Children’s Oncology Group. J Clin Oncol. 2012;30:4148-4154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 419]  [Cited by in F6Publishing: 464]  [Article Influence: 38.7]  [Reference Citation Analysis (0)]
34.  Ferrari S, Sundby Hall K, Luksch R, Tienghi A, Wiebe T, Fagioli F, Alvegard TA, Brach Del Prever A, Tamburini A, Alberghini M. Nonmetastatic Ewing family tumors: high-dose chemotherapy with stem cell rescue in poor responder patients. Results of the Italian Sarcoma Group/Scandinavian Sarcoma Group III protocol. Ann Oncol. 2011;22:1221-1227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 90]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
35.  Gaspar N, Rey A, Bérard PM, Michon J, Gentet JC, Tabone MD, Roché H, Defachelles AS, Lejars O, Plouvier E. Risk adapted chemotherapy for localised Ewing’s sarcoma of bone: the French EW93 study. Eur J Cancer. 2012;48:1376-1385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 44]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
36.  DuBois SG, Krailo MD, Gebhardt MC, Donaldson SS, Marcus KJ, Dormans J, Shamberger RC, Sailer S, Nicholas RW, Healey JH. Comparative evaluation of local control strategies in localized Ewing sarcoma of bone: a report from the Children’s Oncology Group. Cancer. 2015;121:467-475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 102]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
37.  Hardes J, von Eiff C, Streitbuerger A, Balke M, Budny T, Henrichs MP, Hauschild G, Ahrens H. Reduction of periprosthetic infection with silver-coated megaprostheses in patients with bone sarcoma. J Surg Oncol. 2010;101:389-395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 229]  [Cited by in F6Publishing: 227]  [Article Influence: 16.2]  [Reference Citation Analysis (1)]
38.  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;33:1061-1069.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Donaldson SS, Torrey M, Link MP, Glicksman A, Gilula L, Laurie F, Manning J, Neff J, Reinus W, Thompson E. A multidisciplinary study investigating radiotherapy in Ewing’s sarcoma: end results of POG #8346. Pediatric Oncology Group. Int J Radiat Oncol Biol Phys. 1998;42:125-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 173]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
40.  Schuck A, Rübe C, Könemann S, Rübe CE, Ahrens S, Paulussen M, Dunst J, Jürgens H, Willich N. Postoperative radiotherapy in the treatment of Ewing tumors: influence of the interval between surgery and radiotherapy. Strahlenther Onkol. 2002;178:25-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
41.  Schuck A, Ahrens S, Paulussen M, Kuhlen M, Könemann S, Rübe C, Winkelmann W, Kotz R, Dunst J, Willich N. Local therapy in localized Ewing tumors: results of 1058 patients treated in the CESS 81, CESS 86, and EICESS 92 trials. Int J Radiat Oncol Biol Phys. 2003;55:168-177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 253]  [Cited by in F6Publishing: 253]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
42.  Bacci G, Forni C, Longhi A, Ferrari S, Donati D, De Paolis M, Barbieri E, Pignotti E, Rosito P, Versari M. Long-term outcome for patients with non-metastatic Ewing’s sarcoma treated with adjuvant and neoadjuvant chemotherapies. 402 patients treated at Rizzoli between 1972 and 1992. Eur J Cancer. 2004;40:73-83.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Shankar AG, Pinkerton CR, Atra A, Ashley S, Lewis I, Spooner D, Cannon S, Grimer R, Cotterill SJ, Craft AW. 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;35:1698-1704.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Obata H, Ueda T, Kawai A, Ishii T, Ozaki T, Abe S, Tanaka K, Tsuchiya H, Matsumine A, Yabe H. Clinical outcome of patients with Ewing sarcoma family of tumors of bone in Japan: the Japanese Musculoskeletal Oncology Group cooperative study. Cancer. 2007;109:767-775.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
45.  Rodríguez-Galindo C, Navid F, Liu T, Billups CA, Rao BN, Krasin MJ. Prognostic factors for local and distant control in Ewing sarcoma family of tumors. Ann Oncol. 2008;19:814-820.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 107]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
46.  Lee J, Hoang BH, Ziogas A, Zell JA. Analysis of prognostic factors in Ewing sarcoma using a population-based cancer registry. Cancer. 2010;116:1964-1973.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 122]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
47.  Biswas B, Agarwala S, Rastogi S, Khan SA, Mohanti BK, Sharma DN, Pathy S, Bakhshi S. High burden of metastases and poor outcome in pelvic PNET. Pediatr Blood Cancer. 2013;60:E97-E99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
48.  Paulussen M, Ahrens S, Craft AW, Dunst J, Fröhlich B, Jabar S, Rübe C, Winkelmann W, Wissing S, Zoubek A. Ewing’s tumors with primary lung metastases: survival analysis of 114 (European Intergroup) Cooperative Ewing’s Sarcoma Studies patients. J Clin Oncol. 1998;16:3044-3052.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Juergens C, Weston C, Lewis I, Whelan J, Paulussen M, Oberlin O, Michon J, Zoubek A, Juergens H, Craft A. Safety assessment of intensive induction with vincristine, ifosfamide, doxorubicin, and etoposide (VIDE) in the treatment of Ewing tumors in the EURO-E.W.I.N.G. 99 clinical trial. Pediatr Blood Cancer. 2006;47:22-29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 195]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
50.  Casey DL, Wexler LH, Meyers PA, Magnan H, Chou AJ, Wolden SL. Radiation for bone metastases in Ewing sarcoma and rhabdomyosarcoma. Pediatr Blood Cancer. 2015;62:445-449.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
51.  Heij HA, Vos A, de Kraker J, Voûte PA. Prognostic factors in surgery for pulmonary metastases in children. Surgery. 1994;115:687-693.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  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:999-1001.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Ladenstein R, Hartmann O, Pinkerton CR. The role of megatherapy with autologous bone marrow rescue in solid tumours of childhood. Ann Oncol. 1993;4 Suppl 1:45-58.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 32]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
54.  Meyers PA, Krailo MD, Ladanyi M, Chan KW, Sailer SL, Dickman PS, Baker DL, Davis JH, Gerbing RB, Grovas A. High-dose melphalan, etoposide, total-body irradiation, and autologous stem-cell reconstitution as consolidation therapy for high-risk Ewing’s sarcoma does not improve prognosis. J Clin Oncol. 2001;19:2812-2820.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Yock TI, Krailo M, Fryer CJ, Donaldson SS, Miser JS, Chen Z, Bernstein M, Laurie F, Gebhardt MC, Grier HE. Local control in pelvic Ewing sarcoma: analysis from INT-0091--a report from the Children’s Oncology Group. J Clin Oncol. 2006;24:3838-3843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 117]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
56.  Rodriguez-Galindo C, Billups CA, Kun LE, Rao BN, Pratt CB, Merchant TE, Santana VM, Pappo AS. Survival after recurrence of Ewing tumors: the St Jude Children’s Research Hospital experience, 1979-1999. Cancer. 2002;94:561-569.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 130]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
57.  Hunold A, Weddeling N, Paulussen M, Ranft A, Liebscher C, Jürgens H. Topotecan and cyclophosphamide in patients with refractory or relapsed Ewing tumors. Pediatr Blood Cancer. 2006;47:795-800.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 124]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
58.  Van Winkle P, Angiolillo A, Krailo M, Cheung YK, Anderson B, Davenport V, Reaman G, Cairo MS. Ifosfamide, carboplatin, and etoposide (ICE) reinduction chemotherapy in a large cohort of children and adolescents with recurrent/refractory sarcoma: the Children’s Cancer Group (CCG) experience. Pediatr Blood Cancer. 2005;44:338-347.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 92]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
59.  Wagner LM, Crews KR, Iacono LC, Houghton PJ, Fuller CE, McCarville MB, Goldsby RE, Albritton K, Stewart CF, Santana VM. Phase I trial of temozolomide and protracted irinotecan in pediatric patients with refractory solid tumors. Clin Cancer Res. 2004;10:840-848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 94]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
60.  Fox E, Patel S, Wathen JK, Schuetze S, Chawla S, Harmon D, Reinke D, Chugh R, Benjamin RS, Helman LJ. Phase II study of sequential gemcitabine followed by docetaxel for recurrent Ewing sarcoma, osteosarcoma, or unresectable or locally recurrent chondrosarcoma: results of Sarcoma Alliance for Research Through Collaboration Study 003. Oncologist. 2012;17:321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 90]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
61.  Casey DA, Wexler LH, Merchant MS, Chou AJ, Merola PR, Price AP, Meyers PA. Irinotecan and temozolomide for Ewing sarcoma: the Memorial Sloan-Kettering experience. Pediatr Blood Cancer. 2009;53:1029-1034.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 132]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
62.  Wagner LM, McAllister N, Goldsby RE, Rausen AR, McNall-Knapp RY, McCarville MB, Albritton K. Temozolomide and intravenous irinotecan for treatment of advanced Ewing sarcoma. Pediatr Blood Cancer. 2007;48:132-139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 156]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
63.  Raciborska A, Bilska K, Drabko K, Chaber R, Pogorzala M, Wyrobek E, Polczyńska K, Rogowska E, Rodriguez-Galindo C, Wozniak W. Vincristine, irinotecan, and temozolomide in patients with relapsed and refractory Ewing sarcoma. Pediatr Blood Cancer. 2013;60:1621-1625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 69]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
64.  McNall-Knapp RY, Williams CN, Reeves EN, Heideman RL, Meyer WH. Extended phase I evaluation of vincristine, irinotecan, temozolomide, and antibiotic in children with refractory solid tumors. Pediatr Blood Cancer. 2010;54:909-915.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
65.  Kurucu N, Sari N, Ilhan IE. Irinotecan and temozolamide treatment for relapsed Ewing sarcoma: a single-center experience and review of the literature. Pediatr Hematol Oncol. 2015;32:50-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
66.  Gardner SL, Carreras J, Boudreau C, Camitta BM, Adams RH, Chen AR, Davies SM, Edwards JR, Grovas AC, Hale GA. Myeloablative therapy with autologous stem cell rescue for patients with Ewing sarcoma. Bone Marrow Transplant. 2008;41:867-872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
67.  McTiernan A, Driver D, Michelagnoli MP, Kilby AM, Whelan JS. High dose chemotherapy with bone marrow or peripheral stem cell rescue is an effective treatment option for patients with relapsed or progressive Ewing’s sarcoma family of tumours. Ann Oncol. 2006;17:1301-1305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
68.  Erkizan HV, Scher LJ, Gamble SE, Barber-Rotenberg JS, Sajwan KP, Üren A, Toretsky JA. Novel peptide binds EWS-FLI1 and reduces the oncogenic potential in Ewing tumors. Cell Cycle. 2011;10:3397-3408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 25]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
69.  Boro A, Prêtre K, Rechfeld F, Thalhammer V, Oesch S, Wachtel M, Schäfer BW, Niggli FK. Small-molecule screen identifies modulators of EWS/FLI1 target gene expression and cell survival in Ewing’s sarcoma. Int J Cancer. 2012;131:2153-2164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 52]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
70.  Grohar PJ, Griffin LB, Yeung C, Chen QR, Pommier Y, Khanna C, Khan J, Helman LJ. Ecteinascidin 743 interferes with the activity of EWS-FLI1 in Ewing sarcoma cells. Neoplasia. 2011;13:145-153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 93]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
71.  Glade Bender JL, Lee A, Reid JM, Baruchel S, Roberts T, Voss SD, Wu B, Ahern CH, Ingle AM, Harris P. Phase I pharmacokinetic and pharmacodynamic study of pazopanib in children with soft tissue sarcoma and other refractory solid tumors: a children’s oncology group phase I consortium report. J Clin Oncol. 2013;31:3034-3043.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 112]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
72.  Wittrant Y, Théoleyre S, Chipoy C, Padrines M, Blanchard F, Heymann D, Rédini F. RANKL/RANK/OPG: new therapeutic targets in bone tumours and associated osteolysis. Biochim Biophys Acta. 2004;1704:49-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 89]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
73.  Odri GA, Dumoucel S, Picarda G, Battaglia S, Lamoureux F, Corradini N, Rousseau J, Tirode F, Laud K, Delattre O. Zoledronic acid as a new adjuvant therapeutic strategy for Ewing’s sarcoma patients. Cancer Res. 2010;70:7610-7619.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 62]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
74.  Garofalo C, Mancarella C, Grilli A, Manara MC, Astolfi A, Marino MT, Conte A, Sigismund S, Carè A, Belfiore A. Identification of common and distinctive mechanisms of resistance to different anti-IGF-IR agents in Ewing’s sarcoma. Mol Endocrinol. 2012;26:1603-1616.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
75.  Brenner JC, Feng FY, Han S, Patel S, Goyal SV, Bou-Maroun LM, Liu M, Lonigro R, Prensner JR, Tomlins SA. PARP-1 inhibition as a targeted strategy to treat Ewing’s sarcoma. Cancer Res. 2012;72:1608-1613.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 199]  [Article Influence: 16.6]  [Reference Citation Analysis (0)]
76.  Kailayangiri S, Altvater B, Meltzer J, Pscherer S, Luecke A, Dierkes C, Titze U, Leuchte K, Landmeier S, Hotfilder M. The ganglioside antigen G(D2) is surface-expressed in Ewing sarcoma and allows for MHC-independent immune targeting. Br J Cancer. 2012;106:1123-1133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 94]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
77.  Scotlandi K, Perdichizzi S, Bernard G, Nicoletti G, Nanni P, Lollini PL, Curti A, Manara MC, Benini S, Bernard A. Targeting CD99 in association with doxorubicin: an effective combined treatment for Ewing’s sarcoma. Eur J Cancer. 2006;42:91-96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 51]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
78.  Chemaitilly W, Mertens AC, Mitby P, Whitton J, Stovall M, Yasui Y, Robison LL, Sklar CA. Acute ovarian failure in the childhood cancer survivor study. J Clin Endocrinol Metab. 2006;91:1723-1728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 333]  [Cited by in F6Publishing: 281]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
79.  Frederick JL, Ord T, Kettel LM, Stone SC, Balmaceda JP, Asch RH. Successful pregnancy outcome after cryopreservation of all fresh embryos with subsequent transfer into an unstimulated cycle. Fertil Steril. 1995;64:987-990.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
80.  Sonmezer M, Oktay K. Fertility preservation in female patients. Hum Reprod Update. 2004;10:251-266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 344]  [Cited by in F6Publishing: 359]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
81.  Meirow D, Levron J, Eldar-Geva T, Hardan I, Fridman E, Zalel Y, Schiff E, Dor J. Pregnancy after transplantation of cryopreserved ovarian tissue in a patient with ovarian failure after chemotherapy. N Engl J Med. 2005;353:318-321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 676]  [Cited by in F6Publishing: 566]  [Article Influence: 29.8]  [Reference Citation Analysis (0)]
82.  Silber SJ, Lenahan KM, Levine DJ, Pineda JA, Gorman KS, Friez MJ, Crawford EC, Gosden RG. Ovarian transplantation between monozygotic twins discordant for premature ovarian failure. N Engl J Med. 2005;353:58-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 155]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
83.  Husseinzadeh N, Nahhas WA, Velkley DE, Whitney CW, Mortel R. The preservation of ovarian function in young women undergoing pelvic radiation therapy. Gynecol Oncol. 1984;18:373-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 69]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
84.  Blumenfeld Z, Avivi I, Linn S, Epelbaum R, Ben-Shahar M, Haim N. Prevention of irreversible chemotherapy-induced ovarian damage in young women with lymphoma by a gonadotrophin-releasing hormone agonist in parallel to chemotherapy. Hum Reprod. 1996;11:1620-1626.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 216]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
85.  Abir R, Feinmesser M, Yaniv I, Fisch B, Cohen IJ, Ben-Haroush A, Meirow D, Felz C, Avigad S. Occasional involvement of the ovary in Ewing sarcoma. Hum Reprod. 2010;25:1708-1712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 75]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
86.  Sullivan HC, Shulman SC, Olson T, Ricketts R, Oskouei S, Shehata BM. Unusual presentation of metastatic Ewing sarcoma to the ovary in a 13 year-old: a case report and review. Fetal Pediatr Pathol. 2012;31:159-163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
87.  Young RH, Scully RE. Sarcomas metastatic to the ovary: a report of 21 cases. Int J Gynecol Pathol. 1990;9:231-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 96]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]