Bone Cancer

Authors:
J. Sybil Biermann
Search for other papers by J. Sybil Biermann in
Current site
Google Scholar
PubMed
Close
 MD
,
Douglas R. Adkins
Search for other papers by Douglas R. Adkins in
Current site
Google Scholar
PubMed
Close
 MD
,
Mark Agulnik
Search for other papers by Mark Agulnik in
Current site
Google Scholar
PubMed
Close
 MD
,
Robert S. Benjamin
Search for other papers by Robert S. Benjamin in
Current site
Google Scholar
PubMed
Close
 MD
,
Brian Brigman
Search for other papers by Brian Brigman in
Current site
Google Scholar
PubMed
Close
 MD, PhD
,
James E. Butrynski
Search for other papers by James E. Butrynski in
Current site
Google Scholar
PubMed
Close
 MD
,
David Cheong
Search for other papers by David Cheong in
Current site
Google Scholar
PubMed
Close
 MD
,
Warren Chow
Search for other papers by Warren Chow in
Current site
Google Scholar
PubMed
Close
 MD
,
William T. Curry
Search for other papers by William T. Curry in
Current site
Google Scholar
PubMed
Close
 MD
,
Deborah A. Frassica
Search for other papers by Deborah A. Frassica in
Current site
Google Scholar
PubMed
Close
 MD
,
Frank J. Frassica
Search for other papers by Frank J. Frassica in
Current site
Google Scholar
PubMed
Close
 MD
,
Kenneth R. Hande
Search for other papers by Kenneth R. Hande in
Current site
Google Scholar
PubMed
Close
 MD
,
Francis J. Hornicek
Search for other papers by Francis J. Hornicek in
Current site
Google Scholar
PubMed
Close
 MD, PhD
,
Robin L. Jones
Search for other papers by Robin L. Jones in
Current site
Google Scholar
PubMed
Close
 MD, BS, MRCP
,
Joel Mayerson
Search for other papers by Joel Mayerson in
Current site
Google Scholar
PubMed
Close
 MD
,
Sean V. McGarry
Search for other papers by Sean V. McGarry in
Current site
Google Scholar
PubMed
Close
 MD
,
Brian McGrath
Search for other papers by Brian McGrath in
Current site
Google Scholar
PubMed
Close
 MD
,
Carol D. Morris
Search for other papers by Carol D. Morris in
Current site
Google Scholar
PubMed
Close
 MD, MS
,
Richard J. O'Donnell
Search for other papers by Richard J. O'Donnell in
Current site
Google Scholar
PubMed
Close
 MD
,
R. Lor Randall
Search for other papers by R. Lor Randall in
Current site
Google Scholar
PubMed
Close
 MD
,
Victor M. Santana
Search for other papers by Victor M. Santana in
Current site
Google Scholar
PubMed
Close
 MD
,
Robert L. Satcher
Search for other papers by Robert L. Satcher in
Current site
Google Scholar
PubMed
Close
 MD, PhD
,
Herrick J. Siegel
Search for other papers by Herrick J. Siegel in
Current site
Google Scholar
PubMed
Close
 MD
,
Margaret von Mehren
Search for other papers by Margaret von Mehren in
Current site
Google Scholar
PubMed
Close
 MD
,
Mary Anne Bergman
Search for other papers by Mary Anne Bergman in
Current site
Google Scholar
PubMed
Close
, and
Hema Sundar
Search for other papers by Hema Sundar in
Current site
Google Scholar
PubMed
Close
 PhD
View More View Less
Full access

Primary bone cancers are extremely rare neoplasms, accounting for fewer than 0.2% of all cancers. The evaluation and treatment of patients with bone cancers requires a multidisciplinary team of physicians, including musculoskeletal, medical, and radiation oncologists, and surgeons and radiologists with demonstrated expertise in the management of these tumors. Long-term surveillance and follow-up are necessary for the management of treatment late effects related to surgery, radiation therapy, and chemotherapy. These guidelines discuss the management of chordoma, giant cell tumor of the bone, and osteosarcoma.

NCCN Categories of Evidence and Consensus

Category 1: Based upon high-level evidence, there is uniform NCCN consensus that the intervention is appropriate.

Category 2A: Based upon lower-level evidence, there is uniform NCCN consensus that the intervention is appropriate.

Category 2B: Based upon lower-level evidence, there is NCCN consensus that the intervention is appropriate. Category 3: Based upon any level of evidence, there is major NCCN disagreement that the intervention is appropriate.

All recommendations are category 2A unless otherwise noted.

Clinical trials: NCCN believes that the best management for any cancer patient is in a clinical trial. Participation in clinical trials is especially encouraged.

Overview

Primary bone cancers are extremely rare neoplasms accounting for fewer than 0.2% of all cancers, although the true incidence is difficult to determine because of the rarity of these tumors.1,2 In 2013, an estimated 3010 new cases will be diagnosed in the United States and 1440 people will die of the disease.3 Primary bone cancers demonstrate wide clinical heterogeneity and are often curable with proper treatment. Osteosarcoma (35%), chondrosarcoma (30%), and Ewing sarcoma (16%) are the most common forms of bone cancer. Malignant fibrous histiocytoma, fibrosarcoma, chordoma, and giant cell tumor of the bone (GCTB) are rare tumors, constituting up to 1% to 5% of all primary malignant bone tumors.4 GCTB has both benign and malignant forms, with the benign form the most common subtype.

Various types of bone cancers are named based on their histologic origin: chondrosarcomas arise from cartilage, osteosarcomas arise from bone, and fibrogenic tissue is the origin of fibrosarcoma of bone, whereas vascular tissue gives rise to hemangioendothelioma and hemangiopericytoma. Notochordal tissue gives rise to chordoma. Several primary bone cancers, including Ewing sarcoma family of tumors (ESFT), are of unknown histologic origin. Chondrosarcoma is usually found in middle-aged and older adults. Osteosarcoma and Ewing sarcoma develop mainly in children and young adults. Chordoma is more common in men, with peak incidence in the fifth to sixth decades of life.5,6

The pathogenesis and origin of most bone cancers remains unclear. Gene rearrangements between the EWS and ETS family of genes have been implicated in the pathogenesis of ESFT.7-10 Specific germ-line mutations have also been implicated in the pathogenesis of osteosarcoma.11,12 The Li-Fraumeni syndrome characterized by a germ-line mutation in the TP53 gene is associated with a high risk of developing osteosarcoma.13-15 Osteosarcoma is the most common second primary malignancy in patients with a history of retinoblastoma, characterized by a mutation in the retinoblastoma gene RB1.11,16,17 Increased incidences of osteosarcoma have also been associated with other inherited genetic predisposition syndromes characterized by mutations in the DNA helicase genes.11,12 Osteosarcoma

F1

NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013 CHORDOMA

Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 11, 6; 10.6004/jnccn.2013.0088

F2

NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013 GIANT CELL TUMOR OF THE BONE

Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 11, 6; 10.6004/jnccn.2013.0088

F3

NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013 GIANT CELL TUMOR OF THE BONE

Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 11, 6; 10.6004/jnccn.2013.0088

F4

NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013 OSTEOSARCOMA

Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 11, 6; 10.6004/jnccn.2013.0088

F5

NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013 OSTEOSARCOMA

Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 11, 6; 10.6004/jnccn.2013.0088

F6

NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013

Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 11, 6; 10.6004/jnccn.2013.0088

F7

NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013

Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 11, 6; 10.6004/jnccn.2013.0088

F8

NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013

Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

Citation: Journal of the National Comprehensive Cancer Network J Natl Compr Canc Netw 11, 6; 10.6004/jnccn.2013.0088

is also the most common radiation-induced bone sarcoma.18,19

The development of multiagent chemotherapy regimens for neoadjuvant and adjuvant treatment has considerably improved the prognosis for patients with osteosarcoma and ESFT.20,21 With current multimodality treatment, approximately three-quarters of all patients diagnosed with osteosarcoma are cured and 90% to 95% of adult patients diagnosed with osteosarcoma can be successfully treated with limb-sparing approaches rather than amputation.22 Survival rates have improved to almost 70% in patients with localized ESFT.21 A cure is still achievable in selected patients diagnosed with metastatic ESFT and osteosarcoma at presentation.23,24

These guidelines focus on chordoma, chondrosarcoma, ESFT, GCTB, and osteosarcoma. This manuscript highlights only a portion of the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Bone Cancer. Please refer to NCCN.org for the complete version of these guidelines.

Staging

The 2010 AJCC staging classification is shown in Table 1 (available online, in these guidelines, at NCCN. org [ST-1]). This system is based on the assessment of histologic grade (G), tumor size (T), and presence of regional (N) and/or distant metastases (M). The Surgical Staging System (SSS) is another staging system for bone and soft tissue sarcomas developed by the Musculoskeletal Tumor Society (Table 2, available at NCCN.org [ST-1]).25 (available at NCCN.org [ST-1]).25 This system stratifies both bone and soft tissue sarcomas by the assessment of the surgical grade (G), local extent (T), and presence or absence of regional or distant metastases. It may be used in addition to the AJCC staging system.

Principles of Bone Cancer Management

Multidisciplinary Team Involvement

Primary bone tumors and selected metastatic tumors should be evaluated and treated by a multidisciplinary team of physicians with demonstrated expertise in the management of these tumors. Long-term surveillance and follow-up are necessary when considering the risk of recurrence and comorbidities associated with chemotherapy and radiation therapy (RT). Life-long follow-up is recommended for surveillance and treatment of late effects of surgery, RT, and chemotherapy in long-term survivors. Patients should be given a survivorship prescription to schedule follow-up with a multidisciplinary team. Fertility issues should be discussed with appropriate patients before initiation of treatment.26

Diagnostic Workup

Suspicion of a malignant bone tumor in a patient with a painful lesion often begins when a poorly marginated lesion is seen on a plain radiograph. In patients younger than 40 years, an aggressive, painful bone lesion has a significant risk of being a malignant primary bone tumor, and referral to an orthopedic oncologist should be considered before further workup. In patients 40 years of age and older, CT scan of the chest, abdomen, and pelvis; bone scan; mammogram; and other imaging studies as clinically indicated should be performed if plain radiographs do not suggest a specific diagnosis.27

All patients with suspected bone sarcoma should undergo complete staging before biopsy. The standard staging workup for a suspected primary bone cancer should include chest imaging (chest radiograph or chest CT to detect pulmonary metastases), appropriate imaging of the primary site (plain radiographs, MRI for local staging, and/or CT scan), and bone scan.28 Whole-body MRI is a sensitive imaging technique for detecting skeletal metastases in patients with small cell neoplasms, ESFT, and osteosarcoma.29,30 Imaging of painless bone lesions should be evaluated by a musculoskeletal radiologist followed by appropriate referral to a multidisciplinary treatment team if necessary. Laboratory studies, such as CBC, lactate dehydrogenase (LDH), and alkaline phosphatase (ALP), should be performed before initiation of treatment.

PET or PET/CT is an alternative imaging technique that has been used in the pretreatment staging of soft tissue and bone sarcomas.31,32 Recent reports in the literature have shown the utility of PET scans in the evaluation of response to chemotherapy in patients with osteosarcoma, ESFT, and advanced chordoma.33-36 PET or PET/CT with the investigational radioactive substance 18F-fluoromisonidazole (FMISO) has been shown to identify the hypoxic component in residual chordomas before RT.37 This approach is being evaluated in clinical trials and would be helpful in identifying tumors with low oxygen levels that are more resistant to RT.

Biopsy

Incisional (open) and percutaneous biopsy (core needle or fine-needle aspiration) are the techniques used historically in the diagnosis of musculoskeletal lesions.38,39 Open biopsy is the most accurate method because of the larger sample size, which is useful for performing additional studies such as immunohistochemistry or cytogenetics.40 However, open biopsy requires general or regional anesthesia and an operating room, whereas core biopsy can be performed under local anesthesia, with or without sedation. Core needle biopsy has also been used as an alternative to open biopsy for diagnosing musculoskeletal lesions, with accuracy rates ranging from 88% to 96% when adequate samples are obtained.41-44 Cost savings may be realized when needle biopsy is used in selected patients.41 Recent advances in imaging techniques have contributed to the increasing use of image-guided percutaneous biopsy for the diagnosis of primary and secondary bone tumors.45 The preferred method for biopsy remains controversial because no randomized controlled trials have compared core needle biopsy with open biopsy.

The guidelines recommend core needle or open biopsy to confirm the diagnosis of primary bone tumor before any surgical procedure or fixation of primary site. Biopsy should be performed at the center that will provide definitive treatment for patients with a suspected primary malignant bone tumor. At biopsy, careful consideration should be given to appropriate stabilization of the bone and/or to measures to protect against impending pathologic fracture. The placement of biopsy is critical to the planning of limb-sparing surgery, and failure to follow appropriate biopsy procedures may lead to adverse patient outcomes.38,39 In a multicenter review of 597 patients with musculoskeletal tumors, alteration of the treatment plan (complex resection or the use of adjunctive treatment) was encountered in 19%, and unnecessary amputation was performed in 18 patients.46

Both open and core needle biopsy techniques are associated with risk of local tumor recurrence, either through spillage of tumor or tumor seeding along the biopsy tract if the scar is not removed en bloc during tumor resection. The risk of tumor seeding is less with core needle biopsy.47,48 Nevertheless, the same principles should be applied for core needle and open biopsy. Appropriate communication between the surgeon, musculoskeletal oncologist, and bone pathologist is critical in planning the biopsy route. It is essential to select the biopsy route in collaboration with the surgeon to ensure that the biopsy tract lies within the planned resection bed so that it can be resected with the same wide margins as the primary tumor during surgery. Although fine-needle aspiration is not associated with a significant risk of tumor seeding, it is not suitable for the diagnosis of primary lesions, because its diagnostic accuracy is less than that of core needle biopsy.49

Surgery

Surgical margins should be negative, wide enough to minimize potential local recurrence, and narrow enough to maximize function. Wide excision implies histologically negative surgical margins, and is necessary to optimize local control. Local tumor control may be achieved either through limb-sparing resection or limb amputation. In selected cases, amputation may be the most appropriate option to achieve this goal. However, limb-sparing resection is preferred if reasonable functional outcomes can be achieved. Final pathologic evaluation should include assessment of surgical margins and size/dimensions of tumor. The response to the preoperative regimen should be evaluated using pathologic mapping. Consultation with a physiatrist is recommended to evaluate for mobility training and to prescribe an appropriate rehabilitation program.

Radiation Therapy

RT is used either as an adjuvant to surgery for patients with resectable tumors or as definitive therapy in patients with tumors not amenable to surgery. Specialized techniques such as intensity-modulated radiation therapy (IMRT); particle beam RT with protons, carbon ions, or other heavy ions; stereotactic radiosurgery (SRS); or fractionated stereotactic radiotherapy (FSRT) should be considered as clinically indicated to deliver high radiation doses while maximizing normal tissue-sparing. RT should be administered at the same specialized center that is providing surgical and systemic interventions. See “Principles of Radiation Therapy” in the full version of these guidelines for treatment volumes and radiation doses specific to each subtype (to view the most recent version of these guidelines, visit NCCN.org).

Chordoma

Chordomas arise from the embryonic remnants of the notochord and are more common in older adults. Chordomas predominantly arise in the axial skeleton, with the sacrum (50%-60%), skull base (25%-35%), and spine (15%) the most common primary sites.6,50 Chordomas are classified into 3 histologic variants: conventional, chondroid, and dedifferentiated. Conventional chordomas are the most common histologic subtype, characterized by the absence of cartilaginous or mesenchymal components. Chondroid chordomas present with histologic features of chordoma and cartilage elements, accounting for 5% to 15% of all chordomas. Dedifferentiated chordomas constitute approximately 2% to 8% of all chordomas and have features of high-grade spindle cell sarcoma and an aggressive clinical course.50

Chordomas of the spine and sacrum present with localized deep pain or radiculopathies, whereas cervical chordomas can cause airway obstruction or dysphagia and might present as an oropharyngeal mass. Neurologic deficit is more often associated with chordomas of the skull base and mobile spine than with chordomas of the sacrococcygeal region.6

Workup

Initial workup should include a history and physical examination with adequate imaging (radiograph, CT, and MRI) of the primary site, screening MRI of spinal axis, and CT scan of the chest, abdomen, and pelvis. PET scan or bone scan (if PET scan is negative) can be considered for unusual cases. Benign notochordal cell tumors (BNCTs) are considered precursors to chordomas and do not require surgical management.51,52 CT scan and MRI may be useful in distinguishing BNCTs from chordomas.53,54

For skull base chordomas, CT is useful to delineate bone destruction and the presence of calcifications, whereas MRI is the preferred modality for defining the tumor margin from brain, characterizing the position and extension of tumors into the adjacent soft tissue structures, and visualizing blood vessels.55,56 For sacrococcygeal chordomas, CT and MRI are useful for assessing soft tissue involvement, calcifications, and epidural extension.57-59 MRI provides more precise and superior contrast with surrounding soft tissues compared with CT, and is helpful to assess recurrent or metastatic lesions.57,58 CT is also of particular importance to assess bony involvement, calcifications, and soft tissue and epidural extension of spinal chordomas, whereas MRI is the best imaging modality to detect tumor extension, cord compression, local recurrence, and residual tumor in the surgical scar tissue after surgical resection.60,61 CT scan is also useful in planning the reconstruction of the resistant osseous defect in tumors of the proximal sacrum.

Biopsy to confirm histologic subtype should be performed after imaging studies and may vary depending on the anatomic location of the tumor. Needle biopsy is not recommended for skull base tumors. Suspected sacral chordomas should be biopsied dorsally rather than transrectally.

Treatment

Surgery: Wide excision with adequate margins is the preferred primary treatment for patients with chordoma.62,63 A recent retrospective analysis of 962 patients with chordoma identified in the SEER database demonstrated that surgery significantly improves the overall survival (OS).63 Several other reports have confirmed the prognostic significance of wide surgical margins, in terms of relapse-free survival and OS, in patients with chordomas of the sacrum,64-66 skull base67-71 and spine.66,72

Among patients with chordoma of the mobile spine, Boriani et al72 reported that only margin-free en bloc resection was associated with continuous disease-free survival (DFS) with a follow-up of longer than 5 years; 12 of 18 patients were continuously disease-free at an average of 8 years after en bloc resection, whereas all patients who were treated with intralesional excision experienced recurrences in less than 2 years. In patients with chordomas of the sacrum and spine, Ruggieri et al65 reported a local recurrence rate of only 17% after wide surgical margins compared with 81% after intralesional excision or marginal surgery. Tzortzidis et al68 reported that aggressive microsurgical resection is associated with long-term, tumor-free survival with good functional outcome in patients with cranial base chordomas; gross total removal was achieved in 72% of patients, resulting in local control rates of 50%. In a recent 10-year meta-analysis that included 802 patients with skull base chordomas, Di Maio et al71 reported that those with incomplete resection were 3.83 times more likely to experience a recurrence at 5 years than those with complete resection.

Radiation Therapy: RT (preoperative, postoperative, or intraoperative) is used in combination with surgery to improve local control and DFS for patients with resectable sacral and skull base chordomas.67,73-78 In a retrospective series involving 24 patients with sacral and spine chordomas, combination of short-course preoperative RT, resection, and reduced-field high-dose postoperative RT resulted in 5-year DFS and local control rates of 54% and 72%, respectively.75 In a more recent retrospective series of 15 patients with sacrococcygeal chordoma who underwent surgical treatment, intralesional resection with postoperative RT was associated with lower local recurrence rates (20%) than extralesional resection without RT (100%, with a mean time to recurrence of 2 years); the time to recurrence was also significantly longer in patients who received RT after surgery.77 RT in combination with surgery is also associated with improved local recurrence rates in patients with conventional or chondroid chordomas of the skull base.74,76

Particle beam RT (either alone or in combination with photon beam RT) with high-energy protons79-87 or carbon ions88-92 has resulted in local control rates ranging from 62% to 81% in patients with skull base and extracranial chordomas involving the spine and sacrum. In patients with sacral chordomas tumors treated with a combination of high-energy proton and photon beam RT, local control rates were higher in patients with primary tumors compared with those with recurrent tumors.85 Carbon ion RT also resulted in preservation of urinary and anorectal function compared with surgery in patients with sacral chordomas.92

Specialized techniques such as IMRT, SRS, and FSRT have also been associated with good local control rates in cranial and extracranial chordomas.93-97

Systemic Therapy: Chordomas are not sensitive to chemotherapy except for the potentially dedifferentiated portion of high-grade dedifferentiated chordomas.98 Several signal transduction pathways, including platelet-derived growth factor receptor, epidermal growth factor receptor (EGFR), and mammalian target of rapamycin (mTOR), have been implicated in the pathogenesis of chordomas, leading to the development of targeted therapies.99,100

In a phase II trial of 56 patients with advanced chordoma treated with imatinib, 70% of patients had stable disease. The clinical benefit rate as determined based on RECIST criteria (complete response + partial response and stable disease ≥6 months) was 64%, and the median progression-free survival (PFS) in the intention-to-treat population was 9 months.36 Imatinib in combination with cisplatin or sirolimus has also been effective in small series of patients with advanced chordoma resistant to prior imatinib therapy.101,102 The efficacy of EGFR inhibitors such as gefitinib, erlotinib, and cetuximab in patients with advanced chordoma resistant to imatinib has also been demonstrated in case reports.103-105

NCCN Recommendations: Tumor location is the most important variable determining the choice of primary treatment for patients with conventional or chondroid chordomas. Dedifferentiated chordomas are usually managed as described in the NCCN Guidelines for Soft Tissue Sarcoma (to view the most recent version of these guidelines, visit NCCN.org).

Wide excision, with or without RT, is the primary treatment option for patients with resectable conventional or chondroid chordomas of the sacrum and mobile spine.62,63 Intralesional excision with or without RT (followed by MRI to assess the adequacy of resection) is the preferred treatment for patients with resectable skull base tumors of conventional or chondroid histology. Maximal safe resection is recommended when appropriate.70 Adjuvant treatment with RT can be considered for large extracompartmental tumors or for positive surgical margins after resection. Postoperative RT has been associated with improved local control and DFS after surgery with macroscopic surgical margins or intralesional excision.73,76,77,106,107 Re-resection, if necessary, can be considered for skull base tumors with positive surgical margins.

RT is the primary treatment option for patients with unresectable chordomas, irrespective of the tumor location.

Surveillance

Surveillance consists of a physical examination, imaging (radiograph, MRI with or without CT) of the surgical site as clinically indicated, chest imaging (every 6 months for 5 years and annually thereafter), and annual cross-sectional abdominal imaging.

Relapsed Disease

Chordomas are characterized by a high rate of local recurrence, and distant metastases to lungs, bone, soft tissue, lymph nodes, liver, and skin have been reported in up to 40% of patients with local recurrence.64,79,108,109 Among patients with recurrent chordomas of skull base and spine, Fagundes et al79 reported a higher 2-year actuarial OS rate for patients treated with subtotal resection than those who received supportive care only (63% and 21%, respectively; P=.001). However, some studies have reported that surgery and RT are associated with lower local control rates for recurrent tumors compared with primary tumors in patients with sacral chordomas.85,95

Patients with recurrent disease can be managed with surgery and/or RT and/or systemic therapy. The guidelines have included imatinib with or without cisplatin or sirolimus and erlotinib with or without cetuximab as systemic therapy options for patients with recurrent tumors.

Giant Cell Tumor of the Bone

GCTB is a rare benign primary tumor of the bone, accounting for approximately 3% to 5% of all primary bone tumors, with a strong tendency for local recurrence, which may metastasize to the lungs.110,111 GCTB usually occurs between 20 and 40 years of age. The distal femur and proximal tibia are the most common primary sites. Malignant transformation to high-grade osteosarcoma has been observed in rare cases and is associated with a poor prognosis.112,113

Workup

Initial workup should include a history and physical examination with adequate imaging (radiograph, CT, and MRI) of the primary site. CT is useful to define the extent of cortical destruction, whereas MRI is the preferred imaging modality to assess the extension of tumors into the adjacent soft tissue and neurovascular structures.114,115 Chest imaging is essential to identify the presence of metastatic disease. Bone scan can be considered for unusual cases. Biopsy is essential to confirm the diagnosis. Brown tumor of hyperparathyroidism should be considered as a differential diagnosis, although routine evolution of serum calcium, phosphate, and parathyroid hormone levels can help exclude this diagnosis.116

Treatment

Surgery: Wide excision and intralesional curettage are the 2 surgical treatment options for patients with resectable tumors.117-123 Wide excision is associated with a lower risk of local recurrence than intralesional curettage, with the local recurrence rates ranging from 0% to 12% for wide excision and 12% to 65% for intralesional curettage. In some studies, the extent of intralesional excision and the tumor stage have been identified as prognostic indicators for risk of recurrence.124-126 Blackley et al125 reported a local recurrence rate of 12% in 59 patients who were treated with curettage with high-speed burr and bone grafting, which was similar to that observed with the use of adjuvants; most of the patients had grade II or III tumors. In another retrospective analysis of 137 patients, Prosser et al126 reported local recurrences in 19% patients after curettage as a primary treatment; the local recurrence rate was only 7% for patients with grade I and II tumors confined to the bone, compared with 29% for those with grade III tumors with extraosseous extension.

Surgical adjuvants have been used in conjunction with intralesional curettage to improve local control rates. However, the findings from studies that have evaluated intralesional curettage with and without adjuvant in the same cohort of patients with primary or recurrent GCTB are inconsistent, with some reporting decreased local recurrence rates with the use of adjuvants.121,123,127-129 Others, however, have reported no significant difference in local recurrence rates with and without adjuvants.130-132

Wide excision is also associated with poor functional outcome and more surgical complications.133-137 Therefore, intralesional curettage is considered the preferred treatment in most patients with stage I or II tumors. Wide excision is usually reserved for more aggressive stage III tumors with extraosseous extension or for otherwise unresectable tumors.126,138-141

Radiation Therapy: RT has been used either as a primary treatment or in combination with surgery to improve local control and DFS for patients with marginally resected, unresectable, progressive, or recurrent disease.142-152 In a recent retrospective analysis of 58 patients with GCTB (45 with primary tumor and 13 with recurrent tumor) treated with RT, the 5-year local control and OS rates were 85% and 94%, respectively.151 Median follow-up was 8 years. In this analysis, patient age was the only prognostic factor associated with rates of local control (96% for younger patients vs 73% for the older group), OS (100% vs 87%), and DFS (96% vs 65%). Other studies have identified tumor size greater than 4 cm, recurrent tumors, and RT doses of 40 Gy or less as negative prognostic factors for local control.147-149

Specialized techniques such as 3-dimensional conformal RT and IMRT have also been associated with good local control rates in patients with GCTB in locations that are not amenable to complete surgical resection.153,154

NCCN Recommendations Localized Disease: Intralesional excision with or without an effective adjuvant is an adequate primary treatment for resectable tumors.130-132

Serial arterial embolizations have been shown to be effective in the management of patients with giant cell tumors of the extremities, especially for tumors with large cortical defects or joint involvement, and those with large giant cell tumors of the sacrum.155-158 A few case reports have reported the efficacy of interferon and pegylated interferon in the management of GCTB.159-162 More recently, results of a phase II study of denosumab (a fully humanized monoclonal antibody against the RANK ligand) showed significant activity in patients with unresectable or recurrent GCTB, resulting in tumor response (defined as the elimination of at least 90% of giant cells or no radiologic progression of the target lesion for up to 25 weeks) in 86% (30 of 35) of evaluable patients.163,164

For patients with lesions that are resectable with unacceptable morbidity or unresectable axial lesions, the guidelines have included serial embolizations, denosumab, interferon, or pegylated interferon as primary treatment options. RT has been associated with an increased risk of malignant transformation and should be used in patients with tumors that are not amenable to embolization, denosumab, or interferons.

After primary treatment, patients with stable/improved disease can be observed. For patients with stable/improved disease with incomplete healing after primary treatment, intralesional excision is recommended if the lesion has become resectable. Patients with unresectable disease should be treated with denosumab. The guidelines recommend continuation of denosumab until disease progression in patients experiencing response.

Metastatic Disease: For patients presenting with resectable metastases, the guidelines recommend that the primary tumor be managed as described for localized disease.110,111,165,166 Intralesional excision is recommended for resectable metastatic sites. Denosumab, interferon, or pegylated interferon; observation; or RT are included as options for patients with unresectable metastases.

Surveillance

Surveillance should include a physical examination, imaging (radiograph, MRI with or without CT) of the surgical site as clinically indicated, and chest imaging every 6 months for 2 years and then annually thereafter.

Recurrent disease (local or metastatic) should be managed as per primary treatment for localized disease or metastatic disease at presentation.

Osteosarcoma

Osteosarcoma is the most common primary malignant bone tumor in children and young adults.2 The median age for all patients with osteosarcoma is 20 years. In adults older than 65 years, osteosarcoma develops as a secondary malignancy related to Paget disease of the bone.16

Osteosarcoma constitutes a family of lesions with a variety of histologic features and natural histories. Osteosarcomas are broadly classified into intramedullary, surface, and extraskeletal.167 High-grade intramedullary osteosarcoma is the classic or conventional form, constituting nearly 80% of osteosarcomas.167 It is a spindle cell tumor that produces osteoid or immature bone. The most frequent sites are the metaphyseal areas of the distal femur or proximal tibia, which are the sites of maximum growth. Low-grade intramedullary osteosarcoma constitutes fewer than 2% of all osteosarcomas, and the most common sites are similar to those of conventional osteosarcoma.168

Parosteal and periosteal osteosarcomas are juxtacortical or surface variants. Parosteal osteosarcomas are low-grade lesions accounting for up to 5% of all osteosarcomas.168 The most common site is the posterior distal femur. This variant tends to metastasize later than the conventional form. Transformation of low-grade parosteal osteosarcoma into high-grade sarcoma has been documented in 24% to 43% of cases.169,170 Periosteal osteosarcomas are intermediate-grade lesions most often involving the femur followed by the tibia.168 High-grade surface osteosarcomas are rare, accounting for 10% of all juxtacortical osteosarcomas.171,172 Pain and swelling are the most frequent early symptoms. Pain is often intermittent in the beginning and a thorough workup sometimes is delayed because symptoms may be confused with growing pains. Osteosarcoma spreads hematogenously, with the lung being the most common metastatic site.

Prognostic Factors

Tumor site and size; patient age; presence and location of metastases; histologic response to chemotherapy; type of surgery; and surgical margins are significant prognostic factors for patients with osteosarcoma of the extremities and trunk.173-178 In an analysis of 1702 patients with osteosarcoma of the trunk or extremities treated in the Cooperative Osteosarcoma Study Group (COSS) protocols, patient age at diagnosis, tumor site, and primary metastases were identified as predictors of survival.175 In patients with extremity osteosarcomas, size and location of the tumor within the limb at diagnosis also had a significant influence on outcome.175 All factors except age were significant in multivariate testing, with surgical remission and histologic response to chemotherapy emerging as the key prognostic factors. In a recent report of the combined analysis of 3 European Osteosarcoma Intergroup randomized controlled trials, Whelan et al178 reported that good histologic response to preoperative chemotherapy, distal location (other than proximal humerus/femur), and female sex were associated with improved survival.

In patients with proven primary metastatic osteosarcoma, the number of metastases at diagnosis and the completeness of surgical resection of all clinically detected tumor sites are of independent prognostic value.24 Patients with one or a few resectable pulmonary metastases have a survival rate that approaches that of patients with no metastatic disease.179,180

Elevated serum ALP and LDH levels have also been identified as prognostic indicators in patients with osteosarcoma.174,176,177 In a cohort of 1421 patients with osteosarcoma of the extremity, Bacci et al176 reported significantly higher serum LDH levels in patients with metastatic disease at presentation than in those with localized disease (36.6% vs 18.8%; P<.0001). The 5-year DFS correlated with serum LDH level (39.5% for patients with high LDH levels and 60% for those with normal values). In another retrospective analysis of 789 patients with osteosarcoma of the extremity, Bacci et al177 reported that the serum ALP level was a significant prognostic factor of event-free survival (EFS) in patients with osteosarcoma of extremity; the 5-year EFS rate was 24% for patients with a serum ALP value more than 4 times higher than the normal value, and 46% for patients with high values below this limit (P<.001). However, in multivariate analysis, these markers did not retain their prognostic significance when compared with tumor volume, age, and histologic response to chemotherapy.174,176

Workup

Osteosarcomas present both a local problem and a concern for distant metastasis. Initial workup should include imaging of the primary site (MRI, with or without CT), chest imaging, PET scan, and/or bone scan. More detailed imaging (CT or MRI) of abnormalities identified on primary imaging is required for suspected metastatic disease.

Plain radiographs of osteosarcomas show cortical destruction and irregular reactive bone formation. Bone scan, although uniformly abnormal at the lesion, may be useful to identify additional synchronous lesions. MRI provides excellent soft tissue contrast and may be essential for operative planning. MRI is the best imaging modality to define the extent of the lesion within the bone and soft tissues, detect “skip” metastases, and evaluate anatomic relationships with the surrounding structures. In addition, ALP and LDH are frequently elevated in patients with osteosarcoma. Serum LDH was significantly higher in patients with metastatic disease at presentation than in patients with localized disease.176

Treatment

Surgery: Surgery (limb-sparing surgery or amputation) remains an essential part of management of patients with osteosarcoma.181 Studies that have compared limb-sparing surgery and amputation in patients with high-grade nonmetastatic osteosarcoma have not shown any significant difference in survival and local recurrence rates.182-184 However, limb-sparing surgery is associated with better functional outcomes.185 In patients with high-grade osteosarcomas with good histologic response to neoadjuvant chemotherapy, limb-sparing surgery is considered the preferred surgical modality if wide surgical margins could be achieved.182,186 Amputation is generally reserved for patients with tumors in unfavorable anatomic locations not amenable to limb-sparing surgery with adequate surgical margins.181,186

Chemotherapy: The addition of adjuvant and neoadjuvant chemotherapy regimens to surgery has improved outcomes in patients with localized osteosarcoma. Early trials used chemotherapy regimens including at least 3 or more of the following drugs: doxorubicin, cisplatin, bleomycin, cyclophosphamide or ifosfamide, dactinomycin, and high-dose methotrexate.187-192 Subsequent clinical trials have demonstrated that short intensive chemotherapy regimens including cisplatin and doxorubicin with or without high-methotrexate and ifosfamide produce excellent long-term results, similar to those that have been achieved with multiagent chemotherapy.193-200

In a randomized trial conducted by the European Osteosarcoma Group, the combination of doxorubicin and cisplatin was better tolerated compared with a multidrug regimen, with no difference in survival between the groups in patients with operable, nonmetastatic osteosarcoma.194 In both groups, the 3- and 5-year OS rates were 65% and 55%, respectively, and the 5-year PFS rate was 44%. In the INT-0133 study, which compared the 3-drug regimen (cisplatin, doxorubicin, and methotrexate) with the 4-drug regimen (cisplatin, doxorubicin, methotrexate, and ifosfamide) for the treatment of patients with nonmetastatic resectable osteosarcoma, no difference was seen in 6-year EFS rates (63% and 64%, respectively) and OS rates (74% and 70%, respectively) between the groups.200

Chemotherapy regimens without doxorubicin or cisplatin have also been evaluated in patients with localized osteosarcoma with the goal of minimizing long-term cardiotoxicity and ototoxicity.201,202 In a phase II study, the combination of cisplatin, ifosfamide, and epirubicin was active and reasonably well tolerated in patients with nonmetastatic osteosarcoma of the extremity.201 With a median follow-up of 64 months, the 5-year DFS and OS rates were 41.9% and 48.2%, respectively. In another randomized multicenter trial (SFOP-OS94), the combination of ifosfamide and etoposide resulted in a higher histologic response rate than the regimen containing high-dose methotrexate and doxorubicin (56% and 39%, respectively). However, the 5-year OS was similar in both arms and no significant difference was seen in 5-year EFS rates.202 Good histopathologic response (>90% necrosis) to neoadjuvant chemotherapy has been shown to be predictive of survival regardless of the type of chemotherapy administered after surgery.176,203 In an analysis of 881 patients with nonmetastatic osteosarcoma of the extremities treated with neoadjuvant chemotherapy and surgery at the Rizzoli Institute, Bacci et al204 showed that the 5-year DFS and OS correlated significantly with histologic response to chemotherapy. The 5-year DFS and OS rates in good and poor responders were 67.9% versus 51.3% (P<.0001) and 78.4% versus 63.7% (P<.0001), respectively. A report from the Children's Oncology Group also confirmed these findings; the 8-year postoperative EFS and OS rates were 81% and 87%, respectively, in good responders.203 The corresponding survival rates were 46% and 52%, respectively, in poor responders.

The addition of muramyl tripeptide phosphatidyl-ethanolamine (MTP-PE) to chemotherapy has also been evaluated in patients with osteosarcoma.200,205 The addition of MTP-PE to chemotherapy was associated with a statistically significant improvement in 6-year OS (from 70% to 78%) and a trend toward better EFS in patients with nonmetastatic resectable osteosarcoma.200 However, the improvement was not statistically different in patients with metastatic disease.205 MTP-PE is not approved by the FDA for the treatment of patients with osteosarcoma.

Localized Disease

The guidelines recommend wide excision as the primary treatment for patients with low-grade (intramedullary and surface) osteosarcomas and periosteal lesions. Chemotherapy before wide excision could be considered for patients with periosteal lesions. Although chemotherapy (neoadjuvant or adjuvant) has been used in the treatment of patients with periosteal osteosarcoma, no data support that the addition of chemotherapy to wide excision improves outcome in patients with periosteal osteosarcoma.206,207 In a review of 119 patients with periosteal sarcoma published by the European Musculoskeletal Oncology Society, the use of neoadjuvant chemotherapy was not a prognostic factor, although it was used in most of the patients.207 More recently, Cesari et al206 also reported similar findings; the 10-year OS rate was 86% and 83% for patients who received adjuvant chemotherapy with surgery and those who underwent surgery alone, respectively (P=.73). After wide excision (of resectable lesions), the guidelines have included postoperative chemotherapy with a category 2B recommendation for patients with low-grade (intramedullary and surface) or periosteal sarcomas with pathologic findings of high-grade disease.

Preoperative chemotherapy before wide excision is preferred for patients with high-grade osteosarcoma (category 1).193-202 Selected elderly patients may benefit from immediate surgery. After wide excision, patients with a good histologic response (amount of viable tumor <10% of the tumor area) should continue to receive several more cycles of the same chemotherapy. Patients with a poor response (viable tumor ≥10% of the tumor area) could be considered for chemotherapy with a different regimen. However, attempts to improve the outcome of poor responders through modifying the adjuvant chemotherapy remain unsuccessful.208-211 Surgical re-resection with or without RT can be considered for positive surgical margins. An ongoing randomized trial of the European and American Osteosarcoma Study Group is evaluating treatment strategies for resectable osteosarcoma based on histologic response to preoperative chemotherapy

RT or adjuvant chemotherapy is recommended if the sarcoma remains unresectable after preoperative chemotherapy. Proton beam RT has been shown to be effective for local control in some patients with unresectable or incompletely resected osteosarcoma.212

Chemotherapy should include appropriate growth factor support (see the NCCN Guidelines for Myeloid Growth Factors for growth factor support; to view the most recent version of these guidelines, visit NCCN.org). A list of specific chemotherapy regimens is available in “Bone Cancer Systemic Therapy Agents” on pages 702-703.

Metastatic Disease at Presentation

Approximately 10% to 20% of patients present with metastatic disease at diagnosis.24,213 The number of metastases at diagnosis and complete surgical resection of all clinically detected tumor sites are of independent prognostic value in patients with primary metastatic disease at presentation.24 Unilateral metastases and lower number of lung nodules were associated with improved outcomes with chemotherapy in patients with synchronous lung metastases.179,180 The 2-year DFS rate was significantly higher for patients with only 1 or 2 metastatic lesions than for those with 3 or more lesions (78% and 28%, respectively).179

Although chemotherapy is associated with improved outcomes in those with nonmetastatic high-grade localized osteosarcoma, the results were significantly poorer in those with metastatic disease at presentation.213-215 In a study of 57 patients with metastatic disease at presentation treated with cisplatin, doxorubicin, and a high dose of methotrexate and ifosfamide, the 2-year EFS and OS rates were 21% and 55%, respectively, compared with 75% and 94% in patients with nonmetastatic disease at presentation, treated with the same chemotherapy protocol.215

Among patients with primary metastases treated in cooperative osteosarcoma trials, long-term survival rates were higher for those whose metastases were excised after chemotherapy and surgery of the primary tumor compared with those whose metastases could not be removed (48% and 5%, respectively).216 The combination of aggressive chemotherapy with simultaneous resection of primary and metastatic lesions has also resulted in improved outcomes in patients with osteosarcoma of the extremity with lung metastases at presentation.217

For patients with resectable metastases (pulmonary, visceral, or skeletal) at presentation, the guidelines recommend preoperative chemotherapy followed by wide excision of the primary tumor. Chemotherapy and metastasectomy are included as options for the management of metastatic disease. Unresectable metastatic disease should be managed with chemotherapy and/or RT followed by reassessment of the primary site for local control.

Surveillance

Once treatment is completed, surveillance should occur every 3 months for 2 years, then every 4 months for year 3, every 6 months for years 4 and 5, and annually thereafter. Surveillance should include a complete physical, chest imaging, and imaging of the primary site. PET scan and/or bone scan (category 2B) may also be considered. Functional reassessment should be performed at every visit.

Relapsed or Refractory Disease

Approximately 30% of patients with localized disease and 80% of patients presenting with metastatic disease will experience relapse. The presence of solitary metastases, longer time to first relapse, and complete resectability of the disease at first recurrence have been reported to be the most important prognostic indicators for improved survival, whereas patients not amenable to surgery and those with a second or a third recurrence have a poor prognosis.218-222 In patients with primary nonmetastatic osteosarcoma, a longer relapse-free interval to pulmonary metastases was significantly associated with better survival.221 The prognostic significance of surgical clearance among patients with second and subsequent recurrences was also confirmed in a recent report of survival estimates derived from large cohorts of unselected patients treated in the COSS group trials.223

The combination of etoposide with cyclophosphamide or ifosfamide has been evaluated in clinical trials.224,225 In a phase II trial of French Society of Pediatric Oncology, ifosfamide and etoposide resulted in a response rate of 48% in patients with relapsed or refractory osteosarcoma.225 In another phase II trial, cyclophosphamide and etoposide resulted in a 19% response rate and a 35% rate of stable disease in patients with relapsed high-risk osteosarcoma.224 PFS at 4 months was 42%. Single-agent gemcitabine and combination regimens such as docetaxel and gemcitabine; cyclophosphamide and topotecan; and ifosfamide, carboplatin, and etoposide have also been effective in the treatment of patients with relapsed or refractory bone sarcomas.226-229

Samarium-153 ethylene diamine tetramethylene phosphonate (153Sm-EDTMP), a bone seeking radiopharmaceutical, has been evaluated in patients with locally recurrent or metastatic osteosarcoma or skeletal metastases.230,231 Anderson et al230 reported that 153Sm-EDTMP with peripheral blood progenitor cell support had low nonhematologic toxicity and provided pain palliation for patients with osteosarcoma local recurrences or osteoblastic bone metastases.230 Results of a recent dose-finding study also showed that 153Sm-EDTMP can be effective in the treatment of patients with high-risk osteosarcoma.231

Targeted inhibition of a variety of molecular pathways, such as insulin-like growth factor 1 receptor (IGF-1R), mTOR, SRC family of kinases, and vascular endothelial growth factor receptors (VEGFRs), is being evaluated in clinical trials to improve outcomes in patients with relapsed or refractory osteosarcoma. In a recent phase II trial from the Italian Sarcoma Group (n=30), sorafenib (a VEGFR inhibitor) showed activity in patients with relapsed and unresectable high-grade osteosarcoma after failure of standard multimodal therapy.232 The PFS at 4 months (primary end point) was 46%. Median PFS and OS were 4 and 7 months, respectively. The clinical benefit rate (defined as no progression at 6 months) was 29%. Partial response and stable disease were seen in 8% and 34% of patients, respectively, and were durable for 6 months or more in 17% of patients.

The safety and efficacy of high-dose chemotherapy/autologous stem cell transplantation has also been evaluated in patients with locally advanced, metastatic, or relapsed sarcoma.233,234 In the Italian Sarcoma Group study, treatment with carboplatin and etoposide followed by stem cell rescue, combined with surgery, induced complete response in chemosensitive patients.234 Transplant-related mortality was 3.1%. The 3-year OS and DFS rates were 20% and 12%, respectively. The efficacy of this approach in high-risk patients has yet to be determined in prospective randomized studies.

The optimal treatment strategy for patients with relapsed or refractory disease has not been defined. If relapse occurs, patients should undergo second-line chemotherapy and/or surgical resection. Based on the results of the recent phase II trial, the guidelines have included sorafenib as a systemic therapy option for patients with relapsed disease.232 A list of other second-line chemotherapy regimens is provided in “Bone Cancer Systemic Therapy Agents” on pages 702-703. Surveillance is recommended for patients responding to second-line therapy.

Patients experiencing disease progression or relapse after second-line therapy could be managed with resection, palliative RT, or best supportive care. The guidelines have also included 153Sm-EDTMP as an option. Participation in a clinical trial should be strongly encouraged.

Individual Disclosures for the NCCN Bone Cancer Panel

T1

References

  • 1

    Unni KK. Dahlin's Bone Tumors: General Aspects and Data on 11,087 Cases. 5th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 1996.

    • Search Google Scholar
    • Export Citation
  • 2

    Chow W, Haglund K, Randall RL. Bone sarcomas. In: Haller DG, Wagman LD, Camphausen C, Hoskins WJ, eds. Cancer Management: A Multidisciplinary Approach Medical, Surgical, & Radiation Oncology. 14th ed. Norwalk, CT: UBM Medica LLC; 2011.

    • Search Google Scholar
    • Export Citation
  • 3

    Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin 2013;63:11-30.

  • 4

    Fletcher D, Unni K, Mertens F. World Health Organization Classification of Tumours: Pathology and Genetics of Tumours of Soft Tissue and Bone. Lyon: IARC Press; 2002.

    • Search Google Scholar
    • Export Citation
  • 5

    McMaster ML, Goldstein AM, Bromley CM et al.. Chordoma: incidence and survival patterns in the United States, 1973-1995. Cancer Causes Control 2001;12:1-11.

    • Search Google Scholar
    • Export Citation
  • 6

    Walcott BP, Nahed BV, Mohyeldin A et al.. Chordoma: current concepts, management, and future directions. Lancet Oncol 2012;13:e69-76.

  • 7

    de Alava E, Gerald WL. Molecular biology of the Ewing's sarcoma/primitive neuroectodermal tumor family. J Clin Oncol 2000;18:204-213.

  • 8

    Delattre O, Zucman J, Melot T et al.. The Ewing family of tumors—a subgroup of small-round-cell tumors defined by specific chimeric transcripts. N Engl J Med 1994;331:294-299.

    • Search Google Scholar
    • Export Citation
  • 9

    Denny CT. Gene rearrangements in Ewing's sarcoma. Cancer Invest 1996;14:83-88.

  • 10

    Burchill SA. Molecular abnormalities in Ewing's sarcoma. Expert Rev Anticancer Ther 2008;8:1675-1687.

  • 11

    Calvert GT, Randall RL, Jones KB et al.. At-risk populations for osteosarcoma: the syndromes and beyond. Sarcoma 2012;2012:152382.

  • 12

    Martin JW, Squire JA, Zielenska M. The genetics of osteosarcoma. Sarcoma 2012;2012:627254.

  • 13

    Malkin D, Li FP, Strong LC et al.. Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science 1990;250:1233-1238.

    • Search Google Scholar
    • Export Citation
  • 14

    McIntyre JF, Smith-Sorensen B, Friend SH et al.. Germline mutations of the p53 tumor suppressor gene in children with osteosarcoma. J Clin Oncol 1994;12:925-930.

    • Search Google Scholar
    • Export Citation
  • 15

    Ognjanovic S, Olivier M, Bergemann TL, Hainaut P. Sarcomas in TP53 germline mutation carriers: a review of the IARC TP53 database. Cancer 2012;118:1387-1396.

    • Search Google Scholar
    • Export Citation
  • 16

    Ottaviani G, Jaffe N. The etiology of osteosarcoma. Cancer Treat Res 2009;152:15-32.

  • 17

    MacCarthy A, Bayne AM, Draper GJ et al.. Non-ocular tumours following retinoblastoma in Great Britain 1951 to 2004. Br J Ophthalmol 2009;93:1159-1162.

    • Search Google Scholar
    • Export Citation
  • 18

    Kalra S, Grimer RJ, Spooner D et al.. Radiation-induced sarcomas of bone: factors that affect outcome. J Bone Joint Surg Br 2007;89:808-813.

  • 19

    Mavrogenis AF, Pala E, Guerra G, Ruggieri P. Post-radiation sarcomas. Clinical outcome of 52 Patients. J Surg Oncol 2012;105:570-576.

  • 20

    Bernstein M, Kovar H, Paulussen M et al.. Ewing's sarcoma family of tumors: current management. Oncologist 2006;11:503-519.

  • 21

    Subbiah V, Anderson P, Lazar AJ et al.. Ewing's sarcoma: standard and experimental treatment options. Curr Treat Options Oncol 2009;10:126-140.

  • 22

    Federman N, Bernthal N, Eilber FC, Tap WD. The multidisciplinary management of osteosarcoma. Curr Treat Options Oncol 2009;10:82-93.

  • 23

    Cotterill SJ, Ahrens S, Paulussen M et al.. Prognostic factors in Ewing's tumor of bone: analysis of 975 patients from the European Intergroup Cooperative Ewing's Sarcoma Study Group. J Clin Oncol 2000;18:3108-3114.

    • Search Google Scholar
    • Export Citation
  • 24

    Kager L, Zoubek A, Potschger U et al.. Primary metastatic osteosarcoma: presentation and outcome of patients treated on neoadjuvant Cooperative Osteosarcoma Study Group protocols. J Clin Oncol 2003;21:2011-2018.

    • Search Google Scholar
    • Export Citation
  • 25

    Enneking WF, Spanier SS, Goodman MA. A system for the surgical staging of musculoskeletal sarcoma. Clin Orthop Relat Res 1980:106-120.

  • 26

    Lee SJ, Schover LR, Partridge AH et al.. American Society of Clinical Oncology recommendations on fertility preservation in cancer patients. J Clin Oncol 2006;24:2917-2931.

    • Search Google Scholar
    • Export Citation
  • 27

    Rougraff BT, Kneisl JS, Simon MA. Skeletal metastases of unknown origin. A prospective study of a diagnostic strategy. J Bone Joint Surg Am 1993;75:1276-1281.

    • Search Google Scholar
    • Export Citation
  • 28

    Heck RK, Peabody TD, Simon MA. Staging of primary malignancies of bone. CA Cancer J Clin 2006;56:366-375.

  • 29

    Kumar J, Seith A, Kumar A et al.. Whole-body MR imaging with the use of parallel imaging for detection of skeletal metastases in pediatric patients with small-cell neoplasms: comparison with skeletal scintigraphy and FDG PET/CT. Pediatr Radiol 2008;38:953-962.

    • Search Google Scholar
    • Export Citation
  • 30

    Daldrup-Link HE, Franzius C, Link TM et al.. Whole-body MR imaging for detection of bone metastases in children and young adults: comparison with skeletal scintigraphy and FDG PET. AJR Am J Roentgenol 2001;177:229-236.

    • Search Google Scholar
    • Export Citation
  • 31

    Schuetze SM. Utility of positron emission tomography in sarcomas. Curr Opin Oncol 2006;18:369-373.

  • 32

    Volker T, Denecke T, Steffen I et al.. Positron emission tomography for staging of pediatric sarcoma patients: results of a prospective multicenter trial. J Clin Oncol 2007;25:5435-5441.

    • Search Google Scholar
    • Export Citation
  • 33

    Hawkins DS, Rajendran JG, Conrad EU et al.. Evaluation of chemotherapy response in pediatric bone sarcomas by [F-18]-fluorodeoxy-D-glucose positron emission tomography. Cancer 2002;94:3277-3284.

    • Search Google Scholar
    • Export Citation
  • 34

    Hawkins DS, Schuetze SM, Butrynski JE et al.. [18F] Fluorodeoxyglucose positron emission tomography predicts outcome for Ewing sarcoma family of tumors. J Clin Oncol 2005;23:8828-8834.

    • Search Google Scholar
    • Export Citation
  • 35

    Hawkins DS, Conrad EU 3rd, Butrynski JE et al.. [F-18]-fluorodeoxy-D-glucose-positron emission tomography response is associated with outcome for extremity osteosarcoma in children and young adults. Cancer 2009;115:3519-3525.

    • Search Google Scholar
    • Export Citation
  • 36

    Stacchiotti S, Longhi A, Ferraresi V et al.. Phase II study of imatinib in advanced chordoma. J Clin Oncol 2012;30:914-920.

  • 37

    Mammar H, Kerrou K, Nataf V et al.. Positron emission tomography/computed tomography imaging of residual skull base chordoma before radiotherapy using fluoromisonidazole and fluorodeoxyglucose: potential consequences for dose painting. Int J Radiat Oncol Biol Phys 2012;84:681-687.

    • Search Google Scholar
    • Export Citation
  • 38

    Liu PT, Valadez SD, Chivers FS et al.. Anatomically based guidelines for core needle biopsy of bone tumors: implications for limb-sparing surgery. Radiographics 2007;27:189-205; discussion 206.

    • Search Google Scholar
    • Export Citation
  • 39

    Huang AJ, Kattapuram SV. Musculoskeletal neoplasms: biopsy and intervention. Radiol Clin North Am 2011;49:1287-1305.

  • 40

    Ashford RU, McCarthy SW, Scolyer RA et al.. Surgical biopsy with intra-operative frozen section. An accurate and cost-effective method for diagnosis of musculoskeletal sarcomas. J Bone Joint Surg Br 2006;88:1207-1211.

    • Search Google Scholar
    • Export Citation
  • 41

    Skrzynski MC, Biermann JS, Montag A, Simon MA. Diagnostic accuracy and charge-savings of outpatient core needle biopsy compared with open biopsy of musculoskeletal tumors. J Bone Joint Surg Am 1996;78:644-649.

    • Search Google Scholar
    • Export Citation
  • 42

    Welker JA, Henshaw RM, Jelinek J et al.. The percutaneous needle biopsy is safe and recommended in the diagnosis of musculoskeletal masses. Cancer 2000;89:2677-2686.

    • Search Google Scholar
    • Export Citation
  • 43

    Mitsuyoshi G, Naito N, Kawai A et al.. Accurate diagnosis of musculoskeletal lesions by core needle biopsy. J Surg Oncol 2006;94:21-27.

  • 44

    Adams SC, Potter BK, Pitcher DJ, Temple HT. Office-based core needle biopsy of bone and soft tissue malignancies: an accurate alternative to open biopsy with infrequent complications. Clin Orthop Relat Res 2010;468:2774-2780.

    • Search Google Scholar
    • Export Citation
  • 45

    Gogna A, Peh WC, Munk PL. Image-guided musculoskeletal biopsy. Radiol Clin North Am 2008;46:455-473, v.

  • 46

    Mankin HJ, Mankin CJ, Simon MA. The hazards of the biopsy, revisited. Members of the Musculoskeletal Tumor Society. J Bone Joint Surg Am 1996;78:656-663.

    • Search Google Scholar
    • Export Citation
  • 47

    Davies NM, Livesley PJ, Cannon SR. Recurrence of an osteosarcoma in a needle biopsy track. J Bone Joint Surg Br 1993;75:977-978.

  • 48

    Saghieh S, Masrouha KZ, Musallam KM et al.. The risk of local recurrence along the core-needle biopsy tract in patients with bone sarcomas. Iowa Orthop J 2010;30:80-83.

    • Search Google Scholar
    • Export Citation
  • 49

    Yang YJ, Damron TA. Comparison of needle core biopsy and fine-needle aspiration for diagnostic accuracy in musculoskeletal lesions. Arch Pathol Lab Med 2004;128:759-764.

    • Search Google Scholar
    • Export Citation
  • 50

    Chugh R, Tawbi H, Lucas DR et al.. Chordoma: the nonsarcoma primary bone tumor. Oncologist 2007;12:1344-1350.

  • 51

    Yamaguchi T, Suzuki S, Ishiiwa H, Ueda Y. Intraosseous benign notochordal cell tumours: overlooked precursors of classic chordomas? Histopathology 2004;44:597-602.

    • Search Google Scholar
    • Export Citation
  • 52

    Deshpande V, Nielsen GP, Rosenthal DI, Rosenberg AE. Intraosseous benign notochord cell tumors (BNCT): further evidence supporting a relationship to chordoma. Am J Surg Pathol 2007;31:1573-1577.

    • Search Google Scholar
    • Export Citation
  • 53

    Yamaguchi T, Iwata J, Sugihara S et al.. Distinguishing benign notochordal cell tumors from vertebral chordoma. Skeletal Radiol 2008;37:291-299.

  • 54

    Nishiguchi T, Mochizuki K, Ohsawa M et al.. Differentiating benign notochordal cell tumors from chordomas: radiographic features on MRI, CT, and tomography. AJR Am J Roentgenol 2011;196:644-650.

    • Search Google Scholar
    • Export Citation
  • 55

    Meyers SP, Hirsch WL Jr, Curtin HD et al.. Chordomas of the skull base: MR features. AJNR Am J Neuroradiol 1992;13:1627-1636.

  • 56

    Weber AL, Liebsch NJ, Sanchez R, Sweriduk ST Jr. Chordomas of the skull base. Radiologic and clinical evaluation. Neuroimaging Clin N Am 1994;4:515-527.

    • Search Google Scholar
    • Export Citation
  • 57

    Rosenthal DI, Scott JA, Mankin HJ et al.. Sacrococcygeal chordoma: magnetic resonance imaging and computed tomography. AJR Am J Roentgenol 1985;145:143-147.

    • Search Google Scholar
    • Export Citation
  • 58

    Plathow C, Weber MA, Debus J, Kauczor HU. [Imaging of sacral chordoma: comparison between MRI and CT]. Radiologe 2005;45:63-68.

  • 59

    Sung MS, Lee GK, Kang HS et al.. Sacrococcygeal chordoma: MR imaging in 30 patients. Skeletal Radiol 2005;34:87-94.

  • 60

    Meyer JE, Lepke RA, Lindfors KK et al.. Chordomas: their CT appearance in the cervical, thoracic and lumbar spine. Radiology 1984;153:693-696.

  • 61

    Ducou le Pointe H, Brugieres P, Chevalier X et al.. Imaging of chordomas of the mobile spine. J Neuroradiol 1991;18:267-276.

  • 62

    Ferraresi V, Nuzzo C, Zoccali C et al.. Chordoma: clinical characteristics, management and prognosis of a case series of 25 patients. BMC Cancer 2010;10:22.

    • Search Google Scholar
    • Export Citation
  • 63

    Jawad MU, Scully SP. Surgery significantly improves survival in patients with chordoma. Spine (Phila Pa 1976) 2010;35:117-123.

  • 64

    Fuchs B, Dickey ID, Yaszemski MJ et al.. Operative management of sacral chordoma. J Bone Joint Surg Am 2005;87:2211-2216.

  • 65

    Ruggieri P, Angelini A, Ussia G et al.. Surgical margins and local control in resection of sacral chordomas. Clin Orthop Relat Res 2010;468:2939-2947.

    • Search Google Scholar
    • Export Citation
  • 66

    Stacchiotti S, Casali PG, Lo Vullo S et al.. Chordoma of the mobile spine and sacrum: a retrospective analysis of a series of patients surgically treated at two referral centers. Ann Surg Oncol 2010;17:211-219.

    • Search Google Scholar
    • Export Citation
  • 67

    Forsyth PA, Cascino TL, Shaw EG et al.. Intracranial chordomas: a clinicopathological and prognostic study of 51 cases. J Neurosurg 1993;78:741-747.

    • Search Google Scholar
    • Export Citation
  • 68

    Tzortzidis F, Elahi F, Wright D et al.. Patient outcome at long-term follow-up after aggressive microsurgical resection of cranial base chordomas. Neurosurgery 2006;59:230-237.

    • Search Google Scholar
    • Export Citation
  • 69

    Sen C, Triana AI, Berglind N et al.. Clival chordomas: clinical management, results, and complications in 71 patients. J Neurosurg 2010;113:1059-1071.

    • Search Google Scholar
    • Export Citation
  • 70

    Wu Z, Zhang J, Zhang L et al.. Prognostic factors for long-term outcome of patients with surgical resection of skull base chordomas-106 cases review in one institution. Neurosurg Rev 2010;33:451-456.

    • Search Google Scholar
    • Export Citation
  • 71

    Di Maio S, Temkin N, Ramanathan D, Sekhar LN. Current comprehensive management of cranial base chordomas: 10-year meta-analysis of observational studies. J Neurosurg 2011;115:1094-1105.

    • Search Google Scholar
    • Export Citation
  • 72

    Boriani S, Bandiera S, Biagini R et al.. Chordoma of the mobile spine: fifty years of experience. Spine (Phila Pa 1976) 2006;31:493-503.

  • 73

    Catton C, O'Sullivan B, Bell R et al.. Chordoma: long-term follow-up after radical photon irradiation. Radiother Oncol 1996;41:67-72.

  • 74

    Foweraker KL, Burton KE, Maynard SE et al.. High-dose radiotherapy in the management of chordoma and chondrosarcoma of the skull base and cervical spine: Part 1—clinical outcomes. Clin Oncol (R Coll Radiol) 2007;19:509-516.

    • Search Google Scholar
    • Export Citation
  • 75

    Wagner TD, Kobayashi W, Dean S et al.. Combination short-course preoperative irradiation, surgical resection, and reduced-field high-dose postoperative irradiation in the treatment of tumors involving the bone. Int J Radiat Oncol Biol Phys 2009;73:259-266.

    • Search Google Scholar
    • Export Citation
  • 76

    Jian BJ, Bloch OG, Yang I et al.. Adjuvant radiation therapy and chondroid chordoma subtype are associated with a lower tumor recurrence rate of cranial chordoma. J Neurooncol 2010;98:101-108.

    • Search Google Scholar
    • Export Citation
  • 77

    Moojen WA, Vleggeert-Lankamp CL, Krol AD, Dijkstra SP. Long-term results: adjuvant radiotherapy in en bloc resection of sacrococcygeal chordoma is advisable. Spine (Phila Pa 1976) 2011;36:E656-661.

    • Search Google Scholar
    • Export Citation
  • 78

    Potluri S, Jefferies SJ, Jena R et al.. Residual postoperative tumour volume predicts outcome after high-dose radiotherapy for chordoma and chondrosarcoma of the skull base and spine. Clin Oncol (R Coll Radiol) 2011;23:199-208.

    • Search Google Scholar
    • Export Citation
  • 79

    Fagundes MA, Hug EB, Liebsch NJ et al.. Radiation therapy for chordomas of the base of skull and cervical spine: patterns of failure and outcome after relapse. Int J Radiat Oncol Biol Phys 1995;33:579-584.

    • Search Google Scholar
    • Export Citation
  • 80

    Hug EB, Loredo LN, Slater JD et al.. Proton radiation therapy for chordomas and chondrosarcomas of the skull base. J Neurosurg 1999;91:432-439.

  • 81

    Munzenrider JE, Liebsch NJ. Proton therapy for tumors of the skull base. Strahlenther Onkol 1999;175(Suppl 2):57-63.

  • 82

    Noel G, Habrand JL, Mammar H et al.. Combination of photon and proton radiation therapy for chordomas and chondrosarcomas of the skull base: the Centre de Protontherapie D'Orsay experience. Int J Radiat Oncol Biol Phys 2001;51:392-398.

    • Search Google Scholar
    • Export Citation
  • 83

    Noel G, Feuvret L, Ferrand R et al.. Radiotherapeutic factors in the management of cervical-basal chordomas and chondrosarcomas. Neurosurgery 2004;55:1252-1260.

    • Search Google Scholar
    • Export Citation
  • 84

    Noel G, Feuvret L, Calugaru V et al.. Chordomas of the base of the skull and upper cervical spine. One hundred patients irradiated by a 3D conformal technique combining photon and proton beams. Acta Oncol 2005;44:700-708.

    • Search Google Scholar
    • Export Citation
  • 85

    Park L, Delaney TF, Liebsch NJ et al.. Sacral chordomas: impact of high-dose proton/photon-beam radiation therapy combined with or without surgery for primary versus recurrent tumor. Int J Radiat Oncol Biol Phys 2006;65:1514-1521.

    • Search Google Scholar
    • Export Citation
  • 86

    Staab A, Rutz HP, Ares C et al.. Spot-scanning-based proton therapy for extracranial chordoma. Int J Radiat Oncol Biol Phys 2011;81:489-496.

  • 87

    Yasuda M, Bresson D, Chibbaro S et al.. Chordomas of the skull base and cervical spine: clinical outcomes associated with a multimodal surgical resection combined with proton-beam radiation in 40 patients. Neurosurg Rev 2012;35:171-182.

    • Search Google Scholar
    • Export Citation
  • 88

    Schulz-Ertner D, Nikoghosyan A, Thilmann C et al.. Results of carbon ion radiotherapy in 152 patients. Int J Radiat Oncol Biol Phys 2004;58:631-640.

    • Search Google Scholar
    • Export Citation
  • 89

    Schulz-Ertner D, Karger CP, Feuerhake A et al.. Effectiveness of carbon ion radiotherapy in the treatment of skull-base chordomas. Int J Radiat Oncol Biol Phys 2007;68:449-457.

    • Search Google Scholar
    • Export Citation
  • 90

    Takahashi S, Kawase T, Yoshida K et al.. Skull base chordomas: efficacy of surgery followed by carbon ion radiotherapy. Acta Neurochir (Wien) 2009;151:759-769.

    • Search Google Scholar
    • Export Citation
  • 91

    Imai R, Kamada T, Sugahara S et al.. Carbon ion radiotherapy for sacral chordoma. Br J Radiol 2011;84 Spec No 1:48-54.

  • 92

    Nishida Y, Kamada T, Imai R et al.. Clinical outcome of sacral chordoma with carbon ion radiotherapy compared with surgery. Int J Radiat Oncol Biol Phys 2011;79:110-116.

    • Search Google Scholar
    • Export Citation
  • 93

    Gabriele P, Macias V, Stasi M et al.. Feasibility of intensity-modulated radiation therapy in the treatment of advanced cervical chordoma. Tumori 2003;89:298-304.

    • Search Google Scholar
    • Export Citation
  • 94

    Yoneoka Y, Tsumanuma I, Fukuda M et al.. Cranial base chordoma—long term outcome and review of the literature. Acta Neurochir (Wien) 2008;150:773-778.

    • Search Google Scholar
    • Export Citation
  • 95

    Zabel-du Bois A, Nikoghosyan A, Schwahofer A et al.. Intensity modulated radiotherapy in the management of sacral chordoma in primary versus recurrent disease. Radiother Oncol 2010;97:408-412.

    • Search Google Scholar
    • Export Citation
  • 96

    Kano H, Iqbal FO, Sheehan J et al.. Stereotactic radiosurgery for chordoma: a report from the North American Gamma Knife Consortium. Neurosurgery 2011;68:379-389.

    • Search Google Scholar
    • Export Citation
  • 97

    Bugoci DM, Girvigian MR, Chen JCT et al.. Photon-based Fractionated Stereotactic Radiotherapy for Postoperative Treatment of Skull Base Chordomas. Am J Clin Oncol 2012.

    • Search Google Scholar
    • Export Citation
  • 98

    Fleming GF, Heimann PS, Stephens JK et al.. Dedifferentiated chordoma. Response to aggressive chemotherapy in two cases. Cancer 1993;72:714-718.

  • 99

    Tamborini E, Miselli F, Negri T et al.. Molecular and biochemical analyses of platelet-derived growth factor receptor (PDGFR) B, PDGFRA, and KIT receptors in chordomas. Clin Cancer Res 2006;12:6920-6928.

    • Search Google Scholar
    • Export Citation
  • 100

    Tamborini E, Virdis E, Negri T et al.. Analysis of receptor tyrosine kinases (RTKs) and downstream pathways in chordomas. Neuro Oncol 2010;12:776-789.

    • Search Google Scholar
    • Export Citation
  • 101

    Casali PG, Stacchiotti S, Grosso F et al.. Adding cisplatin (CDDP) to imatinib (IM) re-establishes tumor response following secondary resistance to IM in advanced chordoma [abstract]. J Clin Oncol 2007;25:Abstract 10038.

    • Search Google Scholar
    • Export Citation
  • 102

    Stacchiotti S, Marrari A, Tamborini E et al.. Response to imatinib plus sirolimus in advanced chordoma. Ann Oncol 2009;20:1886-1894.

  • 103

    Singhal N, Kotasek D, Parnis FX. Response to erlotinib in a patient with treatment refractory chordoma. Anticancer Drugs 2009;20:953-955.

  • 104

    Launay SG, Chetaille B, Medina F et al.. Efficacy of epidermal growth factor receptor targeting in advanced chordoma: case report and literature review. BMC Cancer 2011;11:423.

    • Search Google Scholar
    • Export Citation
  • 105

    Linden O, Ehinger M, Kinhult S et al.. Cetuximab and EGFR inhibitors in patients with neurological symptoms due to chordoma (abstract). J Clin Oncol 2012;30(Suppl):Abstract 10024.

    • Search Google Scholar
    • Export Citation
  • 106

    Thieblemont C, Biron P, Rocher F et al.. Prognostic factors in chordoma: role of postoperative radiotherapy. Eur J Cancer 1995;31A:2255-2259.

  • 107

    Boriani S, Saravanja D, Yamada Y et al.. Challenges of local recurrence and cure in low grade malignant tumors of the spine. Spine (Phila Pa 1976) 2009;34:S48-57.

    • Search Google Scholar
    • Export Citation
  • 108

    Chambers PW, Schwinn CP. Chordoma. A clinicopathologic study of metastasis. Am J Clin Pathol 1979;72:765-776.

  • 109

    McPherson CM, Suki D, McCutcheon IE et al.. Metastatic disease from spinal chordoma: a 10-year experience. J Neurosurg Spine 2006;5:277-280.

  • 110

    Dominkus M, Ruggieri P, Bertoni F et al.. Histologically verified lung metastases in benign giant cell tumours—14 cases from a single institution. Int Orthop 2006;30:499-504.

    • Search Google Scholar
    • Export Citation
  • 111

    Viswanathan S, Jambhekar NA. Metastatic giant cell tumor of bone: are there associated factors and best treatment modalities? Clin Orthop Relat Res 2010;468:827-833.

    • Search Google Scholar
    • Export Citation
  • 112

    Anract P, De Pinieux G, Cottias P et al.. Malignant giant-cell tumours of bone. Clinico-pathological types and prognosis: a review of 29 cases. Int Orthop 1998;22:19-26.

    • Search Google Scholar
    • Export Citation
  • 113

    Bertoni F, Bacchini P, Staals EL. Malignancy in giant cell tumor of bone. Cancer 2003;97:2520-2529.

  • 114

    Purohit S, Pardiwala DN. Imaging of giant cell tumor of bone. Indian J Orthop 2007;41:91-96.

  • 115

    Thomas DM, Skubitz KM. Giant cell tumour of bone. Curr Opin Oncol 2009;21:338-344.

  • 116

    Werner M. Giant cell tumour of bone: morphological, biological and histogenetical aspects. Int Orthop 2006;30:484-489.

  • 117

    McDonald DJ, Sim FH, McLeod RA, Dahlin DC. Giant-cell tumor of bone. J Bone Joint Surg Am 1986;68:235-242.

  • 118

    Campanacci M, Baldini N, Boriani S, Sudanese A. Giant-cell tumor of bone. J Bone Joint Surg Am 1987;69:106-114.

  • 119

    Saiz P, Virkus W, Piasecki P et al.. Results of giant cell tumor of bone treated with intralesional excision. Clin Orthop Relat Res 2004:221-226.

  • 120

    Malek F, Krueger P, Hatmi ZN et al.. Local control of long bone giant cell tumour using curettage, burring and bone grafting without adjuvant therapy. Int Orthop 2006;30:495-498.

    • Search Google Scholar
    • Export Citation
  • 121

    Kivioja AH, Blomqvist C, Hietaniemi K et al.. Cement is recommended in intralesional surgery of giant cell tumors: a Scandinavian Sarcoma Group study of 294 patients followed for a median time of 5 years. Acta Orthop 2008;79:86-93.

    • Search Google Scholar
    • Export Citation
  • 122

    Errani C, Ruggieri P, Asenzio MA et al.. Giant cell tumor of the extremity: a review of 349 cases from a single institution. Cancer Treat Rev 2010;36:1-7.

    • Search Google Scholar
    • Export Citation
  • 123

    Klenke FM, Wenger DE, Inwards CY et al.. Giant cell tumor of bone: risk factors for recurrence. Clin Orthop Relat Res 2011;469:591-599.

  • 124

    O'Donnell RJ, Springfield DS, Motwani HK et al.. Recurrence of giant-cell tumors of the long bones after curettage and packing with cement. J Bone Joint Surg Am 1994;76:1827-1833.

    • Search Google Scholar
    • Export Citation
  • 125

    Blackley HR, Wunder JS, Davis AM et al.. Treatment of giant-cell tumors of long bones with curettage and bone-grafting. J Bone Joint Surg Am 1999;81:811-820.

    • Search Google Scholar
    • Export Citation
  • 126

    Prosser GH, Baloch KG, Tillman RM et al.. Does curettage without adjuvant therapy provide low recurrence rates in giant-cell tumors of bone? Clin Orthop Relat Res 2005:211-218.

    • Search Google Scholar
    • Export Citation
  • 127

    Becker WT, Dohle J, Bernd L et al.. Local recurrence of giant cell tumor of bone after intralesional treatment with and without adjuvant therapy. J Bone Joint Surg Am 2008;90:1060-1067.

    • Search Google Scholar
    • Export Citation
  • 128

    Balke M, Schremper L, Gebert C et al.. Giant cell tumor of bone: treatment and outcome of 214 cases. J Cancer Res Clin Oncol 2008;134:969-978.

  • 129

    Pietschmann MF, Dietz RA, Utzschneider S et al.. The influence of adjuvants on local recurrence rate in giant cell tumour of the bone. Acta Chir Belg 2010;110:584-589.

    • Search Google Scholar
    • Export Citation
  • 130

    Trieb K, Bitzan P, Lang S et al.. Recurrence of curetted and bone-grafted giant-cell tumours with and without adjuvant phenol therapy. Eur J Surg Oncol 2001;27:200-202.

    • Search Google Scholar
    • Export Citation
  • 131

    Turcotte RE, Wunder JS, Isler MH et al.. Giant cell tumor of long bone: a Canadian Sarcoma Group study. Clin Orthop Relat Res 2002:248-258.

  • 132

    Ruggieri P, Mavrogenis AF, Ussia G et al.. Recurrence after and complications associated with adjuvant treatments for sacral giant cell tumor. Clin Orthop Relat Res 2010;468:2954-2961.

    • Search Google Scholar
    • Export Citation
  • 133

    Oda Y, Miura H, Tsuneyoshi M, Iwamoto Y. Giant cell tumor of bone: oncological and functional results of long-term follow-up. Jpn J Clin Oncol 1998;28:323-328.

    • Search Google Scholar
    • Export Citation
  • 134

    Boons HW, Keijser LC, Schreuder HW et al.. Oncologic and functional results after treatment of giant cell tumors of bone. Arch Orthop Trauma Surg 2002;122:17-23.

    • Search Google Scholar
    • Export Citation
  • 135

    Su YP, Chen WM, Chen TH. Giant-cell tumors of bone: an analysis of 87 cases. Int Orthop 2004;28:239-243.

  • 136

    Rastogi S, Prashanth I, Khan SA et al.. Giant cell tumor of bone: is curettage the answer? Indian J Orthop 2007;41:109-114.

  • 137

    Guo W, Sun X, Zang J, Qu H. Intralesional excision versus wide resection for giant cell tumor involving the acetabulum: which is better? Clin Orthop Relat Res 2012;470:1213-1220.

    • Search Google Scholar
    • Export Citation
  • 138

    Labs K, Perka C, Schmidt RG. Treatment of stages 2 and 3 giant-cell tumor. Arch Orthop Trauma Surg 2001;121:83-86.

  • 139

    Mankin HJ, Hornicek FJ. Treatment of giant cell tumors with allograft transplants: a 30-year study. Clin Orthop Relat Res 2005;439:144-150.

    • Search Google Scholar
    • Export Citation
  • 140

    Mendenhall WM, Zlotecki RA, Scarborough MT et al.. Giant cell tumor of bone. Am J Clin Oncol 2006;29:96-99.

  • 141

    Saini R, Bali K, Bachhal V et al.. En bloc excision and autogenous fibular reconstruction for aggressive giant cell tumor of distal radius: a report of 12 cases and review of literature. J Orthop Surg Res 2011;6:14.

    • Search Google Scholar
    • Export Citation
  • 142

    Dahlin DC. Caldwell Lecture. Giant cell tumor of bone: highlights of 407 cases. AJR Am J Roentgenol 1985;144:955-960.

  • 143

    Schwartz LH, Okunieff PG, Rosenberg A, Suit HD. Radiation therapy in the treatment of difficult giant cell tumors. Int J Radiat Oncol Biol Phys 1989;17:1085-1088.

    • Search Google Scholar
    • Export Citation
  • 144

    Bennett CJ, Marcus RB, Million RR, Enneking WF. Radiation therapy for giant cell tumor of bone. Int J Radiat Oncol Biol Phys 1993;26:299-304.

  • 145

    Chakravarti A, Spiro IJ, Hug EB et al.. Megavoltage radiation therapy for axial and inoperable giant-cell tumor of bone. J Bone Joint Surg Am 1999;81:1566-1573.

    • Search Google Scholar
    • Export Citation
  • 146

    Nair MK, Jyothirmayi R. Radiation therapy in the treatment of giant cell tumor of bone. Int J Radiat Oncol Biol Phys 1999;43:1065-1069.

  • 147

    Miszczyk L, Wydmanski J, Spindel J. Efficacy of radiotherapy for giant cell tumor of bone: given either postoperatively or as sole treatment. Int J Radiat Oncol Biol Phys 2001;49:1239-1242.

    • Search Google Scholar
    • Export Citation
  • 148

    Caudell JJ, Ballo MT, Zagars GK et al.. Radiotherapy in the management of giant cell tumor of bone. Int J Radiat Oncol Biol Phys 2003;57:158-165.

  • 149

    Feigenberg SJ, Marcus RB Jr, Zlotecki RA et al.. Radiation therapy for giant cell tumors of bone. Clin Orthop Relat Res 2003:207-216.

  • 150

    Ruka W, Rutkowski P, Morysinski T et al.. The megavoltage radiation therapy in treatment of patients with advanced or difficult giant cell tumors of bone. Int J Radiat Oncol Biol Phys 2010;78:494-498.

    • Search Google Scholar
    • Export Citation
  • 151

    Bhatia S, Miszczyk L, Roelandts M et al.. Radiotherapy for marginally resected, unresectable or recurrent giant cell tumor of the bone: a rare cancer network study. Rare Tumors 2011;3.

    • Search Google Scholar
    • Export Citation
  • 152

    Kriz J, Eich HT, Mucke R et al.. Radiotherapy for giant cell tumors of the bone: a safe and effective treatment modality. Anticancer Res 2012;32:2069-2073.

    • Search Google Scholar
    • Export Citation
  • 153

    Hug EB, Muenter MW, Adams JA et al.. 3-D-conformal radiation therapy for pediatric giant cell tumors of the skull base. Strahlenther Onkol 2002;178:239-244.

    • Search Google Scholar
    • Export Citation
  • 154

    Roeder F, Timke C, Zwicker F et al.. Intensity modulated radiotherapy (IMRT) in benign giant cell tumors—a single institution case series and a short review of the literature. Radiat Oncol 2010;5:18.

    • Search Google Scholar
    • Export Citation
  • 155

    Lin PP, Guzel VB, Moura MF et al.. Long-term follow-up of patients with giant cell tumor of the sacrum treated with selective arterial embolization. Cancer 2002;95:1317-1325.

    • Search Google Scholar
    • Export Citation
  • 156

    Hosalkar HS, Jones KJ, King JJ, Lackman RD. Serial arterial embolization for large sacral giant-cell tumors: mid- to long-term results. Spine (Phila Pa 1976) 2007;32:1107-1115.

    • Search Google Scholar
    • Export Citation
  • 157

    Onishi H, Kaya M, Wada T et al.. Giant cell tumor of the sacrum treated with selective arterial embolization. Int J Clin Oncol 2010;15:416-419.

  • 158

    Emori M, Kaya M, Sasaki M et al.. Pre-operative selective arterial embolization as a neoadjuvant therapy for proximal humerus giant cell tumor of bone: radiological and histological evaluation. Jpn J Clin Oncol 2012;42:851-855.

    • Search Google Scholar
    • Export Citation
  • 159

    Kaiser U, Neumann K, Havemann K. Generalised giant-cell tumour of bone: successful treatment of pulmonary metastases with interferon alpha, a case report. J Cancer Res Clin Oncol 1993;119:301-303.

    • Search Google Scholar
    • Export Citation
  • 160

    Kaban LB, Troulis MJ, Ebb D et al.. Antiangiogenic therapy with interferon alpha for giant cell lesions of the jaws. J Oral Maxillofac Surg 2002;60:1103-1111.

    • Search Google Scholar
    • Export Citation
  • 161

    Yasko AW. Interferon therapy for giant cell tumor of bone. Current Opinion in Orthopaedics 2006;17:568-572.

  • 162

    Wei F, Liu X, Liu Z et al.. Interferon alfa-2b for recurrent and metastatic giant cell tumor of the spine: report of two cases. Spine (Phila Pa 1976) 2010;35:E1418-1422.

    • Search Google Scholar
    • Export Citation
  • 163

    Thomas D, Henshaw R, Skubitz K et al.. Denosumab in patients with giant-cell tumour of bone: an open-label, phase 2 study. Lancet Oncol 2010;11:275-280.

    • Search Google Scholar
    • Export Citation
  • 164

    Branstetter DG, Nelson SD, Manivel JC et al.. Denosumab induces tumor reduction and bone formation in patients with giant-cell tumor of bone. Clin Cancer Res 2012;18:4415-4424.

    • Search Google Scholar
    • Export Citation
  • 165

    Cheng JC, Johnston JO. Giant cell tumor of bone. Prognosis and treatment of pulmonary metastases. Clin Orthop Relat Res 1997:205-214.

  • 166

    Siebenrock KA, Unni KK, Rock MG. Giant-cell tumour of bone metastasising to the lungs. A long-term follow-up. J Bone Joint Surg Br 1998;80:43-47.

    • Search Google Scholar
    • Export Citation
  • 167

    Klein MJ, Siegal GP. Osteosarcoma: anatomic and histologic variants. Am J Clin Pathol 2006;125:555-581.

  • 168

    Antonescu CR, Huvos AG. Low-grade osteogenic sarcoma arising in medullary and surface osseous locations. Am J Clin Pathol 2000;114 Suppl:S90-103.

    • Search Google Scholar
    • Export Citation
  • 169

    Sheth DS, Yasko AW, Raymond AK et al.. Conventional and dedifferentiated parosteal osteosarcoma. Diagnosis, treatment, and outcome. Cancer 1996;78:2136-2145.

    • Search Google Scholar
    • Export Citation
  • 170

    Bertoni F, Bacchini P, Staals EL, Davidovitz P. Dedifferentiated parosteal osteosarcoma: the experience of the Rizzoli Institute. Cancer 2005;103:2373-2382.

    • Search Google Scholar
    • Export Citation
  • 171

    Okada K, Unni KK, Swee RG, Sim FH. High grade surface osteosarcoma: a clinicopathologic study of 46 cases. Cancer 1999;85:1044-1054.

  • 172

    Staals EL, Bacchini P, Bertoni F. High-grade surface osteosarcoma: a review of 25 cases from the Rizzoli Institute. Cancer 2008;112:1592-1599.

    • Search Google Scholar
    • Export Citation
  • 173

    Davis AM, Bell RS, Goodwin PJ. Prognostic factors in osteosarcoma: a critical review. J Clin Oncol 1994;12:423-431.

  • 174

    Ferrari S, Bertoni F, Mercuri M et al.. Predictive factors of disease-free survival for non-metastatic osteosarcoma of the extremity: an analysis of 300 patients treated at the Rizzoli Institute. Ann Oncol 2001;12:1145-1150.

    • Search Google Scholar
    • Export Citation
  • 175

    Bielack SS, Kempf-Bielack B, Delling G et al.. Prognostic factors in high-grade osteosarcoma of the extremities or trunk: an analysis of 1,702 patients treated on neoadjuvant Cooperative Osteosarcoma Study Group protocols. J Clin Oncol 2002;20:776-790.

    • Search Google Scholar
    • Export Citation
  • 176

    Bacci G, Longhi A, Ferrari S et al.. Prognostic significance of serum lactate dehydrogenase in osteosarcoma of the extremity: experience at Rizzoli on 1421 patients treated over the last 30 years. Tumori 2004;90:478-484.

    • Search Google Scholar
    • Export Citation
  • 177

    Bacci G, Longhi A, Versari M et al.. Prognostic factors for osteosarcoma of the extremity treated with neoadjuvant chemotherapy: 15-year experience in 789 patients treated at a single institution. Cancer 2006;106:1154-1161.

    • Search Google Scholar
    • Export Citation
  • 178

    Whelan JS, Jinks RC, McTiernan A et al.. Survival from high-grade localised extremity osteosarcoma: combined results and prognostic factors from three European Osteosarcoma Intergroup randomised controlled trials. Ann Oncol 2012;23:1607-1616.

    • Search Google Scholar
    • Export Citation
  • 179

    Bacci G, Briccoli A, Ferrari S et al.. Neoadjuvant chemotherapy for osteosarcoma of the extremities with synchronous lung metastases: treatment with cisplatin, adriamycin and high dose of methotrexate and ifosfamide. Oncol Rep 2000;7:339-346.

    • Search Google Scholar
    • Export Citation
  • 180

    Daw NC, Billups CA, Rodriguez-Galindo C et al.. Metastatic osteosarcoma. Cancer 2006;106:403-412.

  • 181

    Marulanda GA, Henderson ER, Johnson DA et al.. Orthopedic surgery options for the treatment of primary osteosarcoma. Cancer Control 2008;15:13-20.

  • 182

    Bacci G, Ferrari S, Lari S et al.. Osteosarcoma of the limb. Amputation or limb salvage in patients treated by neoadjuvant chemotherapy. J Bone Joint Surg Br 2002;84:88-92.

    • Search Google Scholar
    • Export Citation
  • 183

    Mavrogenis AF, Abati CN, Romagnoli C, Ruggieri P. Similar survival but better function for patients after limb salvage versus amputation for distal tibia osteosarcoma. Clin Orthop Relat Res 2012;470:1735-1748.

    • Search Google Scholar
    • Export Citation
  • 184

    Simon MA, Aschliman MA, Thomas N, Mankin HJ. Limb-salvage treatment versus amputation for osteosarcoma of the distal end of the femur. 1986. J Bone Joint Surg Am 2005;87:2822.

    • Search Google Scholar
    • Export Citation
  • 185

    Aksnes LH, Bauer HC, Jebsen NL et al.. Limb-sparing surgery preserves more function than amputation: a Scandinavian sarcoma group study of 118 patients. J Bone Joint Surg Br 2008;90:786-794.

    • Search Google Scholar
    • Export Citation
  • 186

    Nagarajan R, Neglia JP, Clohisy DR, Robison LL. Limb salvage and amputation in survivors of pediatric lower-extremity bone tumors: what are the long-term implications? J Clin Oncol 2002;20:4493-4501.

    • Search Google Scholar
    • Export Citation
  • 187

    Winkler K, Beron G, Kotz R et al.. Neoadjuvant chemotherapy for osteogenic sarcoma: results of a Cooperative German/Austrian study. J Clin Oncol 1984;2:617-624.

    • Search Google Scholar
    • Export Citation
  • 188

    Link MP, Goorin AM, Miser AW et al.. The effect of adjuvant chemotherapy on relapse-free survival in patients with osteosarcoma of the extremity. N Engl J Med 1986;314:1600-1606.

    • Search Google Scholar
    • Export Citation
  • 189

    Eilber F, Giuliano A, Eckardt J et al.. Adjuvant chemotherapy for osteosarcoma: a randomized prospective trial. J Clin Oncol 1987;5:21-26.

  • 190

    Link MP, Goorin AM, Horowitz M et al.. Adjuvant chemotherapy of high-grade osteosarcoma of the extremity. Updated results of the Multi-Institutional Osteosarcoma Study. Clin Orthop Relat Res 1991:8-14.

    • Search Google Scholar
    • Export Citation
  • 191

    Meyers PA, Heller G, Healey J et al.. Chemotherapy for nonmetastatic osteogenic sarcoma: the Memorial Sloan-Kettering experience. J Clin Oncol 1992;10:5-15.

    • Search Google Scholar
    • Export Citation
  • 192

    Goorin AM, Schwartzentruber DJ, Devidas M et al.. Presurgical chemotherapy compared with immediate surgery and adjuvant chemotherapy for nonmetastatic osteosarcoma: Pediatric Oncology Group Study POG-8651. J Clin Oncol 2003;21:1574-1580.

    • Search Google Scholar
    • Export Citation
  • 193

    Bramwell V, Burgers M, Sneath R et al.. A comparison of two short intensive adjuvant chemotherapy regimens in operable osteosarcoma of limbs in children and young adults: the first study of the European Osteosarcoma Intergroup. J Clin Oncol 1992;10:1579-1591.

    • Search Google Scholar
    • Export Citation
  • 194

    Souhami RL, Craft AW, Van der Eijken JW et al.. Randomised trial of two regimens of chemotherapy in operable osteosarcoma: a study of the European Osteosarcoma Intergroup. Lancet 1997;350:911-917.

    • Search Google Scholar
    • Export Citation
  • 195

    Fuchs N, Bielack SS, Epler D et al.. Long-term results of the cooperative German-Austrian-Swiss osteosarcoma study group's protocol COSS-86 of intensive multidrug chemotherapy and surgery for osteosarcoma of the limbs. Ann Oncol 1998;9:893-899.

    • Search Google Scholar
    • Export Citation
  • 196

    Bacci G, Ferrari S, Bertoni F et al.. Long-term outcome for patients with nonmetastatic osteosarcoma of the extremity treated at the istituto ortopedico rizzoli according to the istituto ortopedico rizzoli/osteosarcoma-2 protocol: an updated report. J Clin Oncol 2000;18:4016-4027.

    • Search Google Scholar
    • Export Citation
  • 197

    Bacci G, Briccoli A, Ferrari S et al.. Neoadjuvant chemotherapy for osteosarcoma of the extremity: long-term results of the Rizzoli's 4th protocol. Eur J Cancer 2001;37:2030-2039.

    • Search Google Scholar
    • Export Citation
  • 198

    Ferrari S, Smeland S, Mercuri M et al.. Neoadjuvant chemotherapy with high-dose Ifosfamide, high-dose methotrexate, cisplatin, and doxorubicin for patients with localized osteosarcoma of the extremity: a joint study by the Italian and Scandinavian Sarcoma Groups. J Clin Oncol 2005;23:8845-8852.

    • Search Google Scholar
    • Export Citation
  • 199

    Lewis IJ, Nooij MA, Whelan J et al.. Improvement in histologic response but not survival in osteosarcoma patients treated with intensified chemotherapy: a randomized phase III trial of the European Osteosarcoma Intergroup. J Natl Cancer Inst 2007;99:112-128.

    • Search Google Scholar
    • Export Citation
  • 200

    Meyers PA, Schwartz CL, Krailo MD et al.. Osteosarcoma: the addition of muramyl tripeptide to chemotherapy improves overall survival—a report from the Children's Oncology Group. J Clin Oncol 2008;26:633-638.

    • Search Google Scholar
    • Export Citation
  • 201

    Basaran M, Bavbek ES, Saglam S et al.. A phase II study of cisplatin, ifosfamide and epirubicin combination chemotherapy in adults with nonmetastatic and extremity osteosarcomas. Oncology 2007;72:255-260.

    • Search Google Scholar
    • Export Citation
  • 202

    Le Deley MC, Guinebretiere JM, Gentet JC et al.. SFOP OS94: a randomised trial comparing preoperative high-dose methotrexate plus doxorubicin to high-dose methotrexate plus etoposide and ifosfamide in osteosarcoma patients. Eur J Cancer 2007;43:752-761.

    • Search Google Scholar
    • Export Citation
  • 203

    Provisor AJ, Ettinger LJ, Nachman JB et al.. Treatment of nonmetastatic osteosarcoma of the extremity with preoperative and postoperative chemotherapy: a report from the Children's Cancer Group. J Clin Oncol 1997;15:76-84.

    • Search Google Scholar
    • Export Citation
  • 204

    Bacci G, Mercuri M, Longhi A et al.. Grade of chemotherapy-induced necrosis as a predictor of local and systemic control in 881 patients with non-metastatic osteosarcoma of the extremities treated with neoadjuvant chemotherapy in a single institution. Eur J Cancer 2005;41:2079-2085.

    • Search Google Scholar
    • Export Citation
  • 205

    Chou AJ, Kleinerman ES, Krailo MD et al.. Addition of muramyl tripeptide to chemotherapy for patients with newly diagnosed metastatic osteosarcoma: a report from the Children's Oncology Group. Cancer 2009;115:5339-5348.

    • Search Google Scholar
    • Export Citation
  • 206

    Cesari M, Alberghini M, Vanel D et al.. Periosteal osteosarcoma: a single-institution experience. Cancer 2011;117:1731-1735.

  • 207

    Grimer RJ, Bielack S, Flege S et al.. Periosteal osteosarcoma—a European review of outcome. Eur J Cancer 2005;41:2806-2811.

  • 208

    Winkler K, Beron G, Delling G et al.. Neoadjuvant chemotherapy of osteosarcoma: results of a randomized cooperative trial (COSS-82) with salvage chemotherapy based on histological tumor response. J Clin Oncol 1988;6:329-337.

    • Search Google Scholar
    • Export Citation
  • 209

    Smeland S, Muller C, Alvegard TA et al.. Scandinavian Sarcoma Group Osteosarcoma Study SSG VIII: prognostic factors for outcome and the role of replacement salvage chemotherapy for poor histological responders. Eur J Cancer 2003;39:488-494.

    • Search Google Scholar
    • Export Citation
  • 210

    Smeland S, Bruland OS, Hjorth L et al.. Results of the Scandinavian Sarcoma Group XIV protocol for classical osteosarcoma: 63 patients with a minimum follow-up of 4 years. Acta Orthop 2011;82:211-216.

    • Search Google Scholar
    • Export Citation
  • 211

    Ferrari S, Ruggieri P, Cefalo G et al.. Neoadjuvant chemotherapy with methotrexate, cisplatin, and doxorubicin with or without ifosfamide in nonmetastatic osteosarcoma of the extremity: An Italian Sarcoma Group trial ISG/OS-1. J Clin Oncol 2012;30:2112-2118.

    • Search Google Scholar
    • Export Citation
  • 212

    Ciernik IF, Niemierko A, Harmon DC et al.. Proton-based radiotherapy for unresectable or incompletely resected osteosarcoma. Cancer 2011;117:4522-4530.

    • Search Google Scholar
    • Export Citation
  • 213

    Meyers PA, Heller G, Healey JH et al.. Osteogenic sarcoma with clinically detectable metastasis at initial presentation. J Clin Oncol 1993;11:449-453.

    • Search Google Scholar
    • Export Citation
  • 214

    Bacci G, Briccoli A, Mercuri M et al.. Osteosarcoma of the extremities with synchronous lung metastases: long-term results in 44 patients treated with neoadjuvant chemotherapy. J Chemother 1998;10:69-76.

    • Search Google Scholar
    • Export Citation
  • 215

    Bacci G, Briccoli A, Rocca M et al.. Neoadjuvant chemotherapy for osteosarcoma of the extremities with metastases at presentation: recent experience at the Rizzoli Institute in 57 patients treated with cisplatin, doxorubicin, and a high dose of methotrexate and ifosfamide. Ann Oncol 2003;14:1126-1134.

    • Search Google Scholar
    • Export Citation
  • 216

    Winkler K, Torggler S, Beron G et al.. [Results of treatment in primary disseminated osteosarcoma. Analysis of the follow-up of patients in the cooperative osteosarcoma studies COSS-80 and COSS-82]. Onkologie 1989;12:92-96.

    • Search Google Scholar
    • Export Citation
  • 217

    Bacci G, Mercuri M, Briccoli A et al.. Osteogenic sarcoma of the extremity with detectable lung metastases at presentation. Results of treatment of 23 patients with chemotherapy followed by simultaneous resection of primary and metastatic lesions. Cancer 1997;79:245-254.

    • Search Google Scholar
    • Export Citation
  • 218

    Tabone MD, Kalifa C, Rodary C et al.. Osteosarcoma recurrences in pediatric patients previously treated with intensive chemotherapy. J Clin Oncol 1994;12:2614-2620.

    • Search Google Scholar
    • Export Citation
  • 219

    Saeter G, Hoie J, Stenwig AE et al.. Systemic relapse of patients with osteogenic sarcoma. Prognostic factors for long term survival. Cancer 1995;75:1084-1093.

    • Search Google Scholar
    • Export Citation
  • 220

    Ferrari S, Briccoli A, Mercuri M et al.. Postrelapse survival in osteosarcoma of the extremities: prognostic factors for long-term survival. J Clin Oncol 2003;21:710-715.

    • Search Google Scholar
    • Export Citation
  • 221

    Buddingh EP, Anninga JK, Versteegh MI et al.. Prognostic factors in pulmonary metastasized high-grade osteosarcoma. Pediatr Blood Cancer 2010;54:216-221.

    • Search Google Scholar
    • Export Citation
  • 222

    Briccoli A, Rocca M, Salone M et al.. High grade osteosarcoma of the extremities metastatic to the lung: long-term results in 323 patients treated combining surgery and chemotherapy, 1985-2005. Surg Oncol 2010;19:193-199.

    • Search Google Scholar
    • Export Citation
  • 223

    Bielack SS, Kempf-Bielack B, Branscheid D et al.. Second and subsequent recurrences of osteosarcoma: presentation, treatment, and outcomes of 249 consecutive cooperative osteosarcoma study group patients. J Clin Oncol 2009;27:557-565.

    • Search Google Scholar
    • Export Citation
  • 224

    Berger M, Grignani G, Ferrari S et al.. Phase 2 trial of two courses of cyclophosphamide and etoposide for relapsed high-risk osteosarcoma patients. Cancer 2009;115:2980-2987.

    • Search Google Scholar
    • Export Citation
  • 225

    Gentet JC, Brunat-Mentigny M, Demaille MC et al.. Ifosfamide and etoposide in childhood osteosarcoma. A phase II study of the French Society of Paediatric Oncology. Eur J Cancer 1997;33:232-237.

    • Search Google Scholar
    • Export Citation
  • 226

    Merimsky O, Meller I, Flusser G et al.. Gemcitabine in soft tissue or bone sarcoma resistant to standard chemotherapy: a phase II study. Cancer Chemother Pharmacol 2000;45:177-181.

    • Search Google Scholar
    • Export Citation
  • 227

    Navid F, Willert JR, McCarville MB et al.. Combination of gemcitabine and docetaxel in the treatment of children and young adults with refractory bone sarcoma. Cancer 2008;113:419-425.

    • Search Google Scholar
    • Export Citation
  • 228

    Saylors RL, Stine KC, Sullivan J et al.. Cyclophosphamide plus topotecan in children with recurrent or refractory solid tumors: a Pediatric Oncology Group phase II study. J Clin Oncol 2001;19:3463-3469.

    • Search Google Scholar
    • Export Citation
  • 229

    Van Winkle P, Angiolillo A, Krailo M et al.. Ifosfamide, carboplatin, and etoposide (ICE) reinduction chemotherapy in a large cohort of children and adolescents with recurrent/refractory sarcoma: the Children's Cancer Group (CCG) experience. Pediatr Blood Cancer 2005;44:338-347.

    • Search Google Scholar
    • Export Citation
  • 230

    Anderson PM, Wiseman GA, Dispenzieri A et al.. High-dose samarium-153 ethylene diamine tetramethylene phosphonate: low toxicity of skeletal irradiation in patients with osteosarcoma and bone metastases. J Clin Oncol 2002;20:189-196.

    • Search Google Scholar
    • Export Citation
  • 231

    Loeb DM, Garrett-Mayer E, Hobbs RF et al.. Dose-finding study of 153Sm-EDTMP in patients with poor-prognosis osteosarcoma. Cancer 2009;115:2514-2522.

    • Search Google Scholar
    • Export Citation
  • 232

    Grignani G, Palmerini E, Dileo P et al.. A phase II trial of sorafenib in relapsed and unresectable high-grade osteosarcoma after failure of standard multimodal therapy: an Italian Sarcoma Group study. Ann Oncol 2012;23:508-516.

    • Search Google Scholar
    • Export Citation
  • 233

    Lashkari A, Chow WA, Valdes F et al.. Tandem high-dose chemotherapy followed by autologous transplantation in patients with locally advanced or metastatic sarcoma. Anticancer Res 2009;29:3281-3288.

    • Search Google Scholar
    • Export Citation
  • 234

    Fagioli F, Aglietta M, Tienghi A et al.. High-dose chemotherapy in the treatment of relapsed osteosarcoma: an Italian Sarcoma Group study. J Clin Oncol 2002;20:2150-2156.

    • Search Google Scholar
    • Export Citation
  • Collapse
  • Expand
  • View in gallery

    NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013 CHORDOMA

    Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

  • View in gallery

    NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013 GIANT CELL TUMOR OF THE BONE

    Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

  • View in gallery

    NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013 GIANT CELL TUMOR OF THE BONE

    Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

  • View in gallery

    NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013 OSTEOSARCOMA

    Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

  • View in gallery

    NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013 OSTEOSARCOMA

    Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

  • View in gallery

    NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013

    Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

  • View in gallery

    NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013

    Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

  • View in gallery

    NCCN Clinical Practice Guidelines in Oncology: Bone Cancer, Version 2.2013

    Version 2.2013, 01-31-13 ©2013 National Comprehensive Cancer Network, Inc. All rights reserved. The NCCN Guidelines® and this illustration may not be reproduced in any form without the express written permission of NCCN®.

  • 1

    Unni KK. Dahlin's Bone Tumors: General Aspects and Data on 11,087 Cases. 5th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 1996.

    • Search Google Scholar
    • Export Citation
  • 2

    Chow W, Haglund K, Randall RL. Bone sarcomas. In: Haller DG, Wagman LD, Camphausen C, Hoskins WJ, eds. Cancer Management: A Multidisciplinary Approach Medical, Surgical, & Radiation Oncology. 14th ed. Norwalk, CT: UBM Medica LLC; 2011.

    • Search Google Scholar
    • Export Citation
  • 3

    Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin 2013;63:11-30.

  • 4

    Fletcher D, Unni K, Mertens F. World Health Organization Classification of Tumours: Pathology and Genetics of Tumours of Soft Tissue and Bone. Lyon: IARC Press; 2002.

    • Search Google Scholar
    • Export Citation
  • 5

    McMaster ML, Goldstein AM, Bromley CM et al.. Chordoma: incidence and survival patterns in the United States, 1973-1995. Cancer Causes Control 2001;12:1-11.

    • Search Google Scholar
    • Export Citation
  • 6

    Walcott BP, Nahed BV, Mohyeldin A et al.. Chordoma: current concepts, management, and future directions. Lancet Oncol 2012;13:e69-76.

  • 7

    de Alava E, Gerald WL. Molecular biology of the Ewing's sarcoma/primitive neuroectodermal tumor family. J Clin Oncol 2000;18:204-213.

  • 8

    Delattre O, Zucman J, Melot T et al.. The Ewing family of tumors—a subgroup of small-round-cell tumors defined by specific chimeric transcripts. N Engl J Med 1994;331:294-299.

    • Search Google Scholar
    • Export Citation
  • 9

    Denny CT