Abstract
The Children’s Oncology Group (COG) Bone Tumor Committee is responsible for clinical trials and biologic research on localized, metastatic and recurrent osteosarcoma and Ewing sarcoma (EWS). Results of clinical trials in localized disease completed and published in the past 10 years have led to international standard of care chemotherapy for osteosarcoma and EWS. A recent focus on identifying disease subgroups has led to the identification of biologic features associated with poor outcome including the presence of circulating tumor DNA (ctDNA) at diagnosis, and specific genomic alterations -- MYC amplification for osteosarcoma and STAG2 and TP53 mutation for EWS. Studies validating these potential biomarkers are underway. Clinical trials evaluating the addition of multi-targeted kinase inhibitors, which are active in relapsed bone sarcomas, to standard chemotherapy are underway in osteosarcoma and planned in EWS. In addition, the committee has data analyses and a clinical trial underway to evaluate approaches to local management of the primary tumor and metastatic sites. Given the rarity of bone sarcomas, we have prioritized international interactions and are in the process of forming an international data sharing consortium to facilitate refinement of risk stratification and study of rare disease subtypes.
Keywords: research priorities, bone sarcoma, Ewing sarcoma, osteosarcoma
INTRODUCTION
For osteosarcoma and Ewing sarcoma (EWS), treatment relies on intensive chemotherapy and local control with surgery and/or radiation for both primary and metastatic disease. The COG Bone Tumor Committee (referred to as the Committee hereafter) has conducted pivotal trials establishing current international standards of care. As with other sarcomas, multi-targeted kinase inhibitors (mTKIs) demonstrate activity in relapsed and refractory disease warranting further study. The Committee has clinical trials combining mTKIs and chemotherapy in newly diagnosed osteosarcoma and planned for EWS. Genomic characterization of bone sarcomas has not yet identified opportunities for existing molecularly targeted therapies. Similarly, immunotherapy has yet to make a significant impact in bone sarcomas.
Several outstanding questions significantly impact clinical trial conduct and patient care. Essentially all well-studied cancers have biologic subtypes whose identification explain resistance to upfront therapy, predict recurrence, and identify new potential therapies. The Committee is utilizing available clinical trial data and generating genomic data with the goal to more precisely risk-stratify bone sarcomas beyond the current system whereby the presence or absence of metastatic disease and, for osteosarcoma, pathologic response are used. Drugging the known molecular basis of these diseases – the canonical translocation for EWS and chromosomal instability for osteosarcoma – is an area of active investigation with some promising approaches emerging. To facilitate the investigation of new drugs, the Committee has utilized historical clinical trial data to inform clinical trial design in relapsed/refractory patients facilitating the efficient conduct of phase 2 trials with novel therapies in these populations.
STATE OF THE DISEASE – CLINICAL
Ewing Sarcoma
EWS is a classic translocation-associated sarcoma of bone (approximately 80% of patients) and soft tissues (approximately 20% of patients), with a peak incidence in adolescence and young adults (AYA). With <1,000 new cases per year annually in the United States1, collaborative research is essential to improve outcomes. As a result of sequential COG clinical trials, outcomes for patients with localized disease were improved (5-year event-free survival (EFS) is 87%) through chemotherapy intensification with every 2 week vincristine/doxorubicin/cyclophosphamide and ifosfamide/etoposide (VDC/IE) chemotherapy(Figure 1).2 In contrast, patients with newly diagnosed metastatic or recurrent disease have not seen improvements in outcomes for several decades. For example, on the most recently completed COG trial AEWS1221, the 3-year EFS rate was 38.3%3. Importantly, a range of rational strategies to intensify therapy for these patients have not achieved improved outcomes, highlighting a degree of intrinsic chemotherapy resistance for these patients. Likewise, most patients with recurrent EWS will not survive. COG analyses have provided important outcome data for this population, including a median overall survival (OS) rate of 9 months after first relapse4 and a 12.7% EFS rate at 6 months for patients treated on phase 2 trials mainly for multiply recurrent disease5. A critical focus for the field therefore is to develop novel strategies for these patients with unsatisfactory survival.
Figure 1.

Improved outcomes in localized EWS through COG trials of chemotherapy intensification.
EFS=Event-free survival; EWS=Ewing sarcoma; IE=ifosfamide and etoposide; standard timing = every 3 weeks; intensive timing = every 2 weeks
Osteosarcoma
High-grade osteosarcoma has a slightly higher incidence than EWS and is the most common primary bone malignancy with a peak presentation during the pubertal growth spurt6. There is a later “second” peak incidence in adults with similar outcomes to AYA patients when treated with cytotoxic chemotherapy treatment7. Osteosarcoma can occur after radiation therapy, and it has long been recognized as a malignancy associated with hereditary retinoblastoma and Li Fraumeni syndrome, caused by RB1 and TP53 germline mutations, respectively. A recent study of germline whole exome sequencing of over 1,000 osteosarcoma cases, 40% contributed by COG patients, revealed pathogenic or likely pathogenic germline mutations present in almost 30% of patients8.
Outcomes in localized osteosarcoma have plateaued since the early 1980s when the survival benefit of adjuvant systemic therapy in addition to complete surgical resection was demonstrated. The 5-year EFS is 60% (95% CI: 57–62%) and 5-year OS is 76% (95% CI: 74–78%)6. In localized osteosarcoma, poor tumor necrosis (≤90%) after neoadjuvant chemotherapy, an axial skeleton tumor site, age >18, and having a large primary tumor (≥1/3 of the involved bone) portend worse outcomes9. Prognosis is worse for patients with metastatic disease, with 5-year EFS of 28% (95% CI: 23–33%) and 5-year OS of 45% (95% CI: 39–50%)9. The most frequent sites of metastases are the lungs or bone sites distant from the primary tumor10. The most recent phase 3 trial in newly diagnosed osteosarcoma, an international trial into which COG enrolled the majority of patients, evaluated chemotherapy intensification and immunotherapy but failed to demonstrate improved outcomes with either approach11,12. Further, there is a significant burden of early and late side effects of treatment. Hence, the major focus of clinical trials in osteosarcoma is on novel therapies. A Committee publication regarding poor outcomes in recurrent disease is frequently utilized to inform clinical trial design13.
STATE OF THE DISEASE – BIOLOGICAL
Ewing Sarcoma
EWS is defined by the canonical FET::ETS fusion, most commonly EWSR1::FLI114,15. This fusion protein most often joins exon 7 of EWSR1 to either exon 6 (60%) or exon 5 (25%) of FLI1 15. Other rare variants involve alternative exons of EWSR1 or FLI1, or alternative fusion partners such as ERG, ETV1, ETV4 or FEV16–20. Studies by the COG and others showed an absence of prognostic significance of exon 6 vs. exon 5 fusions in spite of a higher proliferative rate in the lab for exon 6 fusions21,22. The prognostic significance of rarer variant fusions is incompletely investigated.
Like other translocation-positive sarcomas, the genetic background of EWS is characterized by few other recurrent mutations. Copy number alterations (CNAs) are the most common feature with gains of chromosome 8 (50% of cases), chromosome 2 (25% of cases), 1q (25% of cases) and loss of 16q (30%) being the most common23–26. Mutations in STAG2 (15–20%), TP53 (5–10%), and CDKN2A (10–12%)24,26 are rare but studies utilizing samples from children on COG trials and a recent international consensus report suggest that they may be prognostically significant23,27. An additional feature associated with poor outcome, also identified through analysis of samples from Ewing patients enrolled on COG studies, is the detection of circulating tumor DNA (ctDNA) at the time of diagnosis28. If validated, these findings may provide opportunitites to biologically stratify patients based on the combination of clinical and molecular factors. Validation is underway using tissue from patients enrolled on prospective COG clinical trials. Meanwhile, basic biology work continues to better understand how these mutations serve as disease modifiers in the background of a clear oncogenic driver, EWSR1::FLI1 29,30.
The dependence of EWS cells on the canonical FET::ETS fusion is quite clear and has been known for more than 25 years, but transcription factors are challenging drug targets particularly for proteins characterized by low complexity domains as found in the 5’ EWSR1 portion of EWSR1::FLI131,32. While the initial clinical evaluation of cytarabine, mithramycin, TK216, trabectedin, and seclidemstat (LSD1 inhibitor) have been disappointing, new second generation inhibitors with improved toxicity profiles, the mithramycin analog AIT-102 and lurbinectedin show promise33–40. These initial negative clinical studies may reflect the need for alternative schedules of administration for transcription factor drug targets, as has been suggested preclinically41. Emerging data suggest an alternative schedule of trabectedin in combination with low-dose irinotecan may be effective with confirmed partial responses reported 42 Finally, immunotherapy has not yet demonstrated robust activity in EWS with active ongoing efforts exploring this potential therapeutic approach43,44.
Osteosarcoma
The osteosarcoma genome is characterized by copy number gains and losses and mostly non-recurrent fusions suggesting it develops due to chromosomal instability45–48. TP53 loss is nearly universal as are alterations in cell cycle genes46. Recurrent RB1 loss and amplification of CDK4, CCNE1 and 6p21 containing CCND3 have been noted by multiple groups45–47. Other frequent genomic alterations observed in osteosarcoma include amplification of PDGFRB often with adjacent genes KDR and KIT and loss of ATRX through deletion of inactivating mutations. Emerging approaches to target chromosomal instability through targeting kinases involved in DNA damage repair and exploring strategies related to BRCA1 functional deficiency such as PARP inhibitors49 may have relevance in osteosarcoma given the presence of mutational signature 3 in 30% of cases. Data regarding whether the DNA damage leading to the complex osteosarcoma genome is an ongoing process after diagnosis50 are emerging as are data regarding epigenetic and metabolic acclimations of metastasis and progression51,52. Immune therapies have been and are being actively investigated with single agent immune checkpoint inhibitors lacking a clinical signal alone53. A potentially promising approach is antibody-drug conjugates targeting membrane proteins selectively expressed on osteosarcoma cells54.
MAJOR RECENT FINDINGS
Ewing Sarcoma
The Committee has four recently reported major findings. First, we completed long-term follow-up of AEWS0031, a phase 3 trial comparing standard timing with vincristine/doxorubicin/cyclophosphamide alternating every three weeks with ifosfamide/etoposide (VDC/IE regimen) to interval compressed VDC/IE given every two weeks55. These 10-year follow-up data demonstrate the EFS advantage of interval-compressed chemotherapy is maintained, show an OS advantage with interval compression, and report similar rates of second malignancy with either schedule. AEWS0031 results established a North American standard of care which was extended internationally with the recent EuroEwing2012 trial results demonstrating superiority of interval-compressed VDC/IE regimen compared to a different European standard regimen56.
Our successor trial for patients with localized EWS, AEWS1031, evaluated the role of vincristine/topotecan/cyclophosphamide (VTC) in this population2. Patients were randomized to interval-compressed VDC/IE regimen or interval-compressed chemotherapy with VDC/IE/VTC. The addition of VTC did not improve 5-year EFS (79%) compared to interval-compressed VDC/IE (78%). The outcomes of patients on AEWS1031 which included adult patients are the best reported by any cooperative group trial for this population further confirming the role of interval-compressed chemotherapy and extending it as a standard beyond pediatrics.
In the newly-diagnosed metastatic setting, we completed the AEWS1221 trial evaluating the role of IGF-1R inhibition3, based upon a large number of phase 1 and 2 trials demonstrating activity of that approach in patients with relapsed EWS. Patients on AEWS1221 were randomized at study entry to interval-compressed VDC/IE vs. that same therapy plus the IGF-1R monoclonal antibody ganitumab. The trial enrolled 298 eligible patients over 4.25 years, demonstrating the ability of the COG to complete a randomized trial in a feasible timeline in this rare subset of a rare disease. Ganitumab did not improve outcomes (3-year EFS 39.1%) compared to interval-compressed chemotherapy (3-year EFS 37.4%) and may have increased toxicity, including pneumonitis. Importantly, AEWS1221 included the option for stereotactic body radiotherapy (SBRT) to sites of bone metastasis and was the first COG trial to evaluate SBRT.
From a biology perspective, we have leveraged our rich biospecimen archive to provide new biological insights in this disease. In the past decade, specimens from patients enrolled on COG tumor banking protocols have been distributed for 23 projects. Analyses of COG specimens have revealed that the presence of ctDNA at the time of diagnosis and loss of STAG2 protein by immunohistochemistry27,28 are each associated with poorer prognosis. In addition, we determined that translocations are incompletely characterized by local testing 57.
Osteosarcoma
COG sites contributed the majority of the 2,260 patients who enrolled on the international collaborative phase 3 EURAMOS158 trial, also known as AOST0331, for newly diagnosed osteosarcoma. Results, published in 2015 and 201611,12,58 showed no statistically significant difference in EFS or OS for patients with >90% necrosis to preoperative chemotherapy who were randomized to the addition of IFN-α−2b maintenance following the chemotherapy regimen methotrexate, adriamycin and cisplatin (MAP). Patients with poor response (≤90% necrosis after preoperative chemotherapy) randomization to MAP plus Ifosfamide/Etoposide (IE) compared to those randomized to MAP alone also did not have improved EFS or OS. There was increased toxicity seen in patients receiving MAP/IE. This trial therefore confirmed that ifosfamide/etoposide are not recommended to be added to MAP in upfront treatment of osteosarcoma patients based on necrosis and established MAP as an international standard of care for AYA patients.
The Committee focused its efforts on identifying drugs with signals of activity in the relapsed setting which could be combined with chemotherapy in trials in newly diagnosed patients aiming to improve OS. The approach utilizes a historical benchmark for EFS based on analysis of COG legacy clinical trial data13. Osteosarcoma-specific endpoints are required because the tumor has a calcified bone matrix that does not shrink in response to active therapy59 and because 25% of the recurrent patient population undergoes, as a standard of care treatment approach, resection of measurable disease. Utilizing this approach, we were able to study four new agents in recurrent osteosarcoma though none of them demonstrated sufficient efficacy for further investigation(Table 1) 60–63. These studies had rapid patient accrual, including AYA patients, revealing an unmet need for trials in this patient population. In addition, the osteosarcoma-specific endpoints we proposed have been widely adopted, with similar endpoints being adopted for non-COG trials in this patient population and over 120 citations of our original publication. Several industry, collaborative group, and academic trials of multi-targeted kinase inhibitors in recurrent osteosarcoma have been conducted. These have demonstrated consistent class efficacy as measured by improvements in progression-free survival durations and rates compared to the historical benchmark and randomized controls (Table 1)64–68.
Table 1.
Phase 2 trials of new agents in osteosarcoma conducted by COG and select phase 2 trials of mTKI conducted by other groups
| Phase 2 Trial | Drug and mechanism | Relapsed Disease States Included | Target change in % Progression Free | % PF | Accrued (N) | Accrual Duration (mos) |
|---|---|---|---|---|---|---|
| AOST 1322 61 | Eribulin (microtubule inhibitor)* | Measurable | 20%➔40% @ 4 mos | 0% | 16 | 4 |
| AOST 1521 63 | Glembatumumab (GPNMB-targeting ADC)* | Measurable | 20%➔40% @ 4 mos | 13.6% (3/22) | 22 | 6 |
| AOST 1321 60 | Denosumab* (RANKL Ab) |
Measurable NED |
20%➔40% @ 4 mos 30%➔50% @ 12 mos |
6.7% (1/15) 26.3% (10/38) |
16 38 |
6 22 |
| AOST 1421 62 | Dinutuximab + GM-CSF (GD2 Ab) | NED | 30%➔50% @ 12 mos | 28.2% (11/39) | 39 | 24 |
| PEPN1924 72 | Trastuzumab deruxtecan (ADC) | Measurable | PFS @ 6 mos | 11% (1/9) | 9 | 7 |
| Cabozantinib67 | mTKI (VEGFR2/MET/AXL) | Measurable | PF @ 4 mos | 71% | 45 OS | 39 |
| Regorafenib68 | mTKI (VEGF/PDGFR/FGFR/KIT/RET/RAF) | Measurable | PF @ 4 mos | 44.4% Rego vs 0% placebo | 43 (38 evaluable) | 30 |
PF = progression free; NED=no evidence of disease; Ab=Antibody; ADC=Antibody-drug conjugate; mTKI=multi-targeted tyrosine kinase inhibitors; mos=months; GPNMB=glycoprotein non-metastatic protein B; RANKL=Receptor activator of nuclear factor kappa-Β ligand; GM-CSF=granulocyte-macrophage colony-stimulating factor; PF=Progression-free; OS=osteosarcoma
First clinical trial in children
To address some of the high-priority biology questions, emphasis has been placed on supporting basic science investigation in osteosarcoma. There were 939 distinct patients with specimens banked through protocol AOST06B1 and stored at the Biopathology Center. With the opening of Project:EveryChild (APEC14B1) in October 2015, AOST06B1 closed to enrollment in December 2017. In the last decade, biospecimens have been distributed to 26 projects. Results of these studies have provided proof of principle for biologic subtypes associated with treatment response and resistance such MYC amplification and the presence of ctDNA at diagnosis with poor outcome28,69.
STRATEGIC APPROACH
The primary focus of the Committee continues to be to design and conduct clinical trials leading to improved patient outcomes and decreased toxicity. In patients with relapsed/refractory disease, the Committee prioritizes clinical trials focusing on determining whether new agents with pre-clinical activity have clinical activity. In newly diagnosed patients, we focus on assessing the impact of adding to chemotherapy novel agents which, when given to relapsed patients, show a clinical signal of activity. Clinical trials to be pursued are discussed further below.
Moreover, we employ a multidisciplinary therapeutic approach to trial design as well investigating novel local control approaches to disease management at primary and metastatic sites with surgery and radiation. In osteosarcoma, the COG Surgery Committee identified the approach to resection of lung metastasis in osteosarcoma as the highest priority surgical local control question leading to the design of AOST2031 discussed below. We are similarly evaluating local control outcomes on our most recent trials for patients with EWS in order to determine risk factors for local recurrence which can then be addressed with improvement in local control approaches in subsequent trials.
Additional strategic priorities are those that will prepare the Committee to design the next generation of clinical trials in EWS and osteosarcoma. Our approach has been to organize and harmonize clinical data and investigate associated biospecimens to further develop and validate clinical-biological subtyping of bone sarcomas. For example, preliminary data suggest that osteosarcoma may be divided into unique biologic subtypes with distinct prognostic implications45,70 based on the presence of MYC amplification. The Committee is a third of the way done with a case-control study using COG banked samples to validate this observation71. In an analysis of samples from patients enrolled on COG banking studies, the detection of ctDNA at the time of diagnosis has been associated with poor outcome in both EWS and osteosarcoma.28 COG is contributing samples to a prospective study validation of these findings regarding ctDNA and outcome which will also assess changes in ctDNA over time. Other examples of studies underway within the committee utilizing banked samples include AEWS18B1, aimed at validating the reported association between STAG2 and TP53 outcome in a 350-patient cohort who received treatment with interval compressed VDC/IE on COG trials.
The Committee has embarked on an international data-sharing effort to facilitate refinement of risk prediction and study rare subtypes. The Harmonization International Bone Sarcoma Consortium (HIBiSCus), memorandum of understanding was signed in April 2021 and data contributor agreements are in process. The Data for the Common Good (D4CG) will host the aggregated data, making it available for use.
The Committee further facilitates biologic discovery by supporting scientists requesting samples. We provide detailed sample acquisition guidance in AOST2031 and have embedded a patient-derived xenograft study into this protocol. We continue efforts to improve the utility of banked specimens by incentivizing submission of frozen samples and samples from metastatic sites with ongoing pathology review and nucleic acid extraction of specimens {Sonja Chen, 2022 #57}.
KEY TRIALS TO BE PURSUED
Ewing Sarcoma
The committee has plans to pursue trials in all three clinical groups: newly diagnosed localized disease; newly diagnosed metastatic disease; and recurrent disease (Table 2). In the newly diagnosed localized population, we are validating clinical and molecular factors to inform a future risk-stratified trial. Guiding principles will be therapy reduction (e.g., fewer cycles of VDC/IE) for patients with defined low risk localized tumors and addition of targeted therapies for patients with defined high-risk localized tumors. Candidate biomarkers include STAG2 mutation and protein expression status, TP53 mutation status, baseline ctDNA burden, and response to therapy.
Table 2.
Ongoing and Planned Clinical Trials in Ewing Sarcoma and Osteosarcoma
| Trial Number | Patient population | Phase | Intervention | Primary outcome measure | Status |
|---|---|---|---|---|---|
| Ewing Sarcoma | |||||
| AEWS2231 | Newly dx, metastatic | 3 | VDC and either VIR or IE | EFS | In development |
| ADVL1921 | Recurrent | 2 | irinotecan/temozolomide alone or with palbociclib | EFS | Active |
| Pending assigned study number; Newly Diagnosed Task Force | Newly dx, localized | Unk | Risk-based therapy | Unk | In development |
| Osteosarcoma | |||||
| AOST2032 | Newly dx | Feasibility, 2/3 | Cabozantinib + MAP | EFS | Active |
| AOST2031 | Lung only metastatic (newly diagnosed and recurrent) | 3 | Randomized: thoracotomy or thoracoscopy | Pulmonary EFS | Active |
| AOST2121 | Recurrent, fully resected | 2 | OST31–164 Her2-targeting Listeria | EFS | Active |
| PEPN1924 | Recurrent | 2 | Trastuzumab Deruxtecan | EFS | Active |
| AOST2221 | Recurrent, fully resected | 2 | Regorafenib | EFS | In development |
VDC=vincristine/doxorubicin/cyclophosphamide; VIR=vincristine/irinotecan/regorafenib; EFS=event-free survival; MAP=methotrexate/adriamycin/cisplatin; Unk=Unknown; HER2=human erythroblastic oncogene B
In the newly-diagnosed metastatic population, we are pursuing the combination of vincristine/irinotecan/regorafenib (VIR) added to VDC in a randomized phase 3 trial compared to VDC/IE. Patients on the VIR arm will also receive maintenance regorafenib. This proposed trial will be the first in COG to include central molecular confirmation of EWS translocation status and the first to include patients with molecularly-defined other round cell sarcomas in a parallel cohort.
In the relapse population, we are contributing to an ongoing international randomized evaluation of irinotecan/temozolomide with or without palbociclib (NCT03709680). In addition, we work closely with COG’s Developmental Therapeutics Committee to monitor for signals of activity emerging from PEP-CTN studies that might inform a phase 2 trial for patients with relapsed EWS. Drugs currently being evaluated in EWS include the Beta-catenin pathway inhibitor tegavivint and the ATR inhibitor BAY1895344.
Osteosarcoma
As stated above, the class effect of angiogenesis-inhibiting mTKIs have clear signal of activity in relapsed osteosarcoma patients. Thus, our approach in the frontline is a feasibility (to optimize dose and schedule and evaluate safety of this novel combination) followed by a phase 2/3 randomized controlled clinical trial investigating to MAP with or without the addition of cabozantinib in both the localized and metastatic populations (NCT05691478). This opened to enrollment in 2023 and includes six months of cabozantinib as maintenance for those randomized to receive it (Table 2).
While chemotherapy and surgery often address the primary site of osteosarcoma, pulmonary recurrence is the most likely route of failure for initially localized disease and the most common site of metastases at presentation. The optimal approach to the surgical management of pulmonary metastases is not clear. The Surgery and Bone Tumor Committees developed AOST2031 to address this critical surgical question by randomizing the surgical approach in a phase 3 study (Figure 2, NCT05235165). This trial is designed to determine if open thoracotomy is superior to thoracoscopic resection for thoracic EFS in patients with oligometastatic pulmonary osteosarcoma. Secondary aims include quality of life assessments utilizing patient reported outcomes. Biology and pathology correlative aims will leverage resected specimens to generate multi-omic data and well-characterized, clinically annotated models of metastatic osteosarcoma.
Figure 2.

AOST2031
In the relapse setting, we continue to pursue clinical trials to assess new drugs for osteosarcoma. An industry-partnered phase 2 trial, AOST2121 launched in 2021 is examining OST31–164, a live-attenuated Listeria bacterial vector and plasmid system coupled with internal and external epitopes of the Her2 receptor, as adjuvant therapy after surgical remission in pulmonary-only relapse. The trial is ongoing with swift accrual. A study of adjuvant regorafenib after resection of pulmonary metastatic disease is in development.
CONCLUSIONS
Over the past 10 years, the Committee’s clinical trials have contributed significantly to harmonization of standard of care chemotherapy for localized disease across the young adult age spectrum and internationally. While we lack an agreed upon risk stratification in localized disease, the Committee activities dedicated to clinical data harmonization efforts and analysis of associated biospecimens hold the promise for identifying subgroups groups in bone sarcomas. Metastatic bone sarcomas continue to be challenging without survival improvements, with ongoing Committee efforts focused on an important local control question in osteosarcoma and addition of mTKIs to chemotherapy in both EWS and osteosarcoma. New strategies including biologic insights and novel agents will be needed to improve outcomes in relapse bone sarcoma patients. We have a clearly delineated strategy for the study of new agents with agents targeting disease specific surface antigens prioritized for study in osteosarcoma and agents affecting the characteristic translocation such as alternative dose trabectidin prioritized for EWS.
Funding support:
Grant support from the National Institute of Health, U10CA180886, U10CA180899, U10CA098543, U10CA098413, U24CA196173, U24CA114766, U10CA180884.
Abbreviation Key:
- ATR
Ataxia telangiectasia and Rad3 related
- ATRX
alpha-thalassemia/mental retardation, X-linked
- AYA(s)
Adolescents and young adult(s)
- BRCA1
Breast cancer type 1 susceptibility
- CCND3
Cyclin D3
- CCNE1
Cyclin E1
- CDK4
Cyclin-dependent kinase 4
- CDKN2A
Cyclin-dependent kinase inhibitor 2A
- CI
Confidence Interval
- CNA
Copy Number Alterations
- COG
Children’s Oncology Group
- ctDNA
Circulating tumor DNA
- D4CG
Data for the Common Good
- DFS
Disease-free survival
- DNA
Deoxyribonucleic acid
- EFS
Event-free survival
- ERG
ETS-related gene
- ETS
erythroblast transformation-specific
- ETV
E twenty-six
- EURAMOS
European and American Osteosarcoma Study group
- EWS
Ewing sarcoma
- EWSR1
Ewing Sarcoma ribonucleic acid binding protein 1
- FDA
Food and Drug Administration
- FET
Female Expressed Transcript
- FEV
Fifth Ewing Variant
- FLI/FLI1
Friend Leukemia Integration 1
- HIBiSCus
Harmonization International Bone Sarcoma Consortium
- JCO
Journal of Clinical Oncology
- KDR
Kinase Insert Domain Receptor
- KIT
receptor tyrosine kinase that responds to stem cell factor
- LSD1
Lysine-specific demethylase 1
- MAP
Methotrexate, Adriamycin (Doxorubicin) and Cisplatin
- mTKIs
multi-targeted kinase inhibitors
- MYC
myelocytomatosis
- NCI
National Cancer Institute
- NIH
National Institute of Health
- PARP
Poly (ADP-ribose) polymerase
- PEP-CTN
Pediatric Early Phase Clinical Trials Network
- PGDFRB
Platelet derived growth factor receptor beta
- OS
Overall survival
- RB1
Retinoblastoma 1
- SBRT
Stereotactic Body Radiation Therapy
- SES
Socioeconomic status
- STAG2
Stromal Antigen 2
- TP53
Tumor protein P53 or transformation-related protein 53
- VDC/IE
Vincristine, Doxorubicin, Cyclophosphamide alternating with Ifosfamide and Etoposide
- VIR
Vincristine, Irinotecan and Regorafenib
- VTC
Vincristine, Topotecan and Cyclophosphamide
Footnotes
CONFLICT OF INTEREST
KAJ consulting for Bayer, Ipsen and Illumina.
DRR advisory role (DSMC) for Eisai and Springworks
SGD travel expenses from Loxo Oncology, Roche, and Salarius and consulting fees from Amgen, Bayer, and Jazz.
OB is a consultant for Onkos
REFERENCES
- 1.Jawad MU, Cheung MC, Min ES, Schneiderbauer MM, Koniaris LG, Scully SP. Ewing sarcoma demonstrates racial disparities in incidence-related and sex-related differences in outcome: an analysis of 1631 cases from the SEER database, 1973–2005. Cancer Aug 1 2009;115(15):3526–36. doi: 10.1002/cncr.24388 [DOI] [PubMed] [Google Scholar]
- 2.Leavey PJ, Laack NN, Krailo MD, et al. Phase III Trial Adding Vincristine-Topotecan-Cyclophosphamide to the Initial Treatment of Patients With Nonmetastatic Ewing Sarcoma: A Children’s Oncology Group Report. J Clin Oncol Dec 20 2021;39(36):4029–4038. doi: 10.1200/JCO.21.00358 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.DuBois SG, Krailo MD, Glade-Bender J, et al. Randomized Phase III Trial of Ganitumab With Interval-Compressed Chemotherapy for Patients With Newly Diagnosed Metastatic Ewing Sarcoma: A Report From the Children’s Oncology Group. J Clin Oncol Apr 10 2023;41(11):2098–2107. doi: 10.1200/JCO.22.01815 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Leavey PJ, Mascarenhas L, Marina N, et al. Prognostic factors for patients with Ewing sarcoma (EWS) at first recurrence following multi-modality therapy: A report from the Children’s Oncology Group. Pediatr Blood Cancer Sep 2008;51(3):334–8. doi: 10.1002/pbc.21618 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Collier AB, 3rd, Krailo MD, Dang HM, et al. Outcome of patients with relapsed or progressive Ewing sarcoma enrolled on cooperative group phase 2 clinical trials: A report from the Children’s Oncology Group. Pediatr Blood Cancer Dec 2021;68(12):e29333. doi: 10.1002/pbc.29333 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Cole S, Gianferante DM, Zhu B, Mirabello L. Osteosarcoma: A Surveillance, Epidemiology, and End Results program-based analysis from 1975 to 2017. Cancer Jun 1 2022;128(11):2107–2118. doi: 10.1002/cncr.34163 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ferrari S, Bielack SS, Smeland S, et al. EURO-B.O.S.S.: A European study on chemotherapy in bone-sarcoma patients aged over 40: Outcome in primary high-grade osteosarcoma. Tumori Jan-Feb 2018;104(1):30–36. doi: 10.5301/tj.5000696 [DOI] [PubMed] [Google Scholar]
- 8.Mirabello L, Zhu B, Koster R, et al. Frequency of Pathogenic Germline Variants in Cancer-Susceptibility Genes in Patients With Osteosarcoma. JAMA Oncol May 1 2020;6(5):724–734. doi: 10.1001/jamaoncol.2020.0197 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Smeland S, Bielack SS, Whelan J, et al. Survival and prognosis with osteosarcoma: outcomes in more than 2000 patients in the EURAMOS-1 (European and American Osteosarcoma Study) cohort. Eur J Cancer Mar 2019;109:36–50. doi: 10.1016/j.ejca.2018.11.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.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. Nov 15 2009;115(22):5339–48. doi: 10.1002/cncr.24566 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Bielack SS, Smeland S, Whelan JS, et al. Methotrexate, Doxorubicin, and Cisplatin (MAP) Plus Maintenance Pegylated Interferon Alfa-2b Versus MAP Alone in Patients With Resectable High-Grade Osteosarcoma and Good Histologic Response to Preoperative MAP: First Results of the EURAMOS-1 Good Response Randomized Controlled Trial. J Clin Oncol Jul 10 2015;33(20):2279–87. doi: 10.1200/JCO.2014.60.0734 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Marina NM, Smeland S, Bielack SS, et al. Comparison of MAPIE versus MAP in patients with a poor response to preoperative chemotherapy for newly diagnosed high-grade osteosarcoma (EURAMOS-1): an open-label, international, randomised controlled trial. Lancet Oncol Oct 2016;17(10):1396–1408. doi: 10.1016/S1470-2045(16)30214-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Lagmay JP, Krailo MD, Dang H, et al. Outcome of Patients With Recurrent Osteosarcoma Enrolled in Seven Phase II Trials Through Children’s Cancer Group, Pediatric Oncology Group, and Children’s Oncology Group: Learning From the Past to Move Forward. J Clin Oncol Sep 1 2016;34(25):3031–8. doi: 10.1200/JCO.2015.65.5381 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Delattre O, Zucman J, Plougastel B, et al. Gene fusion with an ETS DNA-binding domain caused by chromosome translocation in human tumours. Nature Sep 10 1992;359(6391):162–5. doi: 10.1038/359162a0 [DOI] [PubMed] [Google Scholar]
- 15.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 Aug 4 1994;331(5):294–9. [DOI] [PubMed] [Google Scholar]
- 16.Sorensen PH, Lessnick SL, Lopez-Terrada D, Liu XF, Triche TJ, Denny CT. A second Ewing’s sarcoma translocation, t(21;22), fuses the EWS gene to another ETS-family transcription factor, ERG. Nat Genet Feb 1994;6(2):146–51. doi: 10.1038/ng0294-146 [DOI] [PubMed] [Google Scholar]
- 17.Jeon IS, Davis JN, Braun BS, et al. A variant Ewing’s sarcoma translocation (7;22) fuses the EWS gene to the ETS gene ETV1. Oncogene Mar 16 1995;10(6):1229–34. [PubMed] [Google Scholar]
- 18.Ishida S, Yoshida K, Kaneko Y, et al. The genomic breakpoint and chimeric transcripts in the EWSR1-ETV4/E1AF gene fusion in Ewing sarcoma. Cytogenet Cell Genet 1998;82(3–4):278–83. doi: 10.1159/000015119 [DOI] [PubMed] [Google Scholar]
- 19.Peter M, Couturier J, Pacquement H, et al. A new member of the ETS family fused to EWS in Ewing tumors. Oncogene Mar 13 1997;14(10):1159–64. doi: 10.1038/sj.onc.1200933 [DOI] [PubMed] [Google Scholar]
- 20.Kinnaman MD, Zhu C, Weiser DA, et al. Survey of Paediatric Oncologists and Pathologists regarding Their Views and Experiences with Variant Translocations in Ewing and Ewing-Like Sarcoma: A Report of the Children’s Oncology Group. Sarcoma 2020;2020:3498549. doi: 10.1155/2020/3498549 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.van Doorninck JA, Ji L, Schaub B, et al. Current treatment protocols have eliminated the prognostic advantage of type 1 fusions in Ewing sarcoma: a report from the Children’s Oncology Group. J Clin Oncol Apr 20 2010;28(12):1989–94. doi: 10.1200/JCO.2009.24.5845 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Le Deley MC, Delattre O, Schaefer KL, et al. Impact of EWS-ETS fusion type on disease progression in Ewing’s sarcoma/peripheral primitive neuroectodermal tumor: prospective results from the cooperative Euro-E.W.I.N.G. 99 trial. J Clin Oncol Apr 20 2010;28(12):1982–8. doi: 10.1200/JCO.2009.23.3585 [DOI] [PubMed] [Google Scholar]
- 23.Shulman DS, Whittle SB, Surdez D, et al. An international working group consensus report for the prioritization of molecular biomarkers for Ewing sarcoma. NPJ Precis Oncol Sep 17 2022;6(1):65. doi: 10.1038/s41698-022-00307-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Brohl AS, Solomon DA, Chang W, et al. The genomic landscape of the Ewing Sarcoma family of tumors reveals recurrent STAG2 mutation. PLoS Genet Jul 2014;10(7):e1004475. doi: 10.1371/journal.pgen.1004475 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Crompton BD, Stewart C, Taylor-Weiner A, et al. The genomic landscape of pediatric Ewing sarcoma. Cancer Discov Nov 2014;4(11):1326–41. doi: 10.1158/2159-8290.CD-13-1037 [DOI] [PubMed] [Google Scholar]
- 26.Tirode F, Surdez D, Ma X, et al. Genomic landscape of Ewing sarcoma defines an aggressive subtype with co-association of STAG2 and TP53 mutations. Cancer Discov Nov 2014;4(11):1342–53. doi: 10.1158/2159-8290.cd-14-0622 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Shulman DS, Chen S, Hall D, et al. Adverse prognostic impact of the loss of STAG2 protein expression in patients with newly diagnosed localised Ewing sarcoma: A report from the Children’s Oncology Group. Br J Cancer Dec 2022;127(12):2220–2226. doi: 10.1038/s41416-022-01977-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Shulman DS, Klega K, Imamovic-Tuco A, et al. Detection of circulating tumour DNA is associated with inferior outcomes in Ewing sarcoma and osteosarcoma: a report from the Children’s Oncology Group. Br J Cancer Aug 2018;119(5):615–621. doi: 10.1038/s41416-018-0212-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.El Beaino M, Liu J, Wasylishen AR, et al. Loss of Stag2 cooperates with EWS-FLI1 to transform murine Mesenchymal stem cells. BMC Cancer Jan 2 2020;20(1):3. doi: 10.1186/s12885-019-6465-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Adane B, Alexe G, Seong BKA, et al. STAG2 loss rewires oncogenic and developmental programs to promote metastasis in Ewing sarcoma. Cancer Cell Jun 14 2021;39(6):827–844 e10. doi: 10.1016/j.ccell.2021.05.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kovar H, Aryee DN, Jug G, et al. EWS/FLI-1 antagonists induce growth inhibition of Ewing tumor cells in vitro. Cell Growth Differ Apr 1996;7(4):429–37. [PubMed] [Google Scholar]
- 32.Boulay G, Sandoval GJ, Riggi N, et al. Cancer-Specific Retargeting of BAF Complexes by a Prion-like Domain. Cell Sep 21 2017;171(1):163–178 e19. doi: 10.1016/j.cell.2017.07.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Subbiah V, Brana I, Longhi A, et al. Antitumor Activity of Lurbinectedin, a Selective Inhibitor of Oncogene Transcription, in Patients with Relapsed Ewing Sarcoma: Results of a Basket Phase II Study. Clin Cancer Res Jul 1 2022;28(13):2762–2770. doi: 10.1158/1078-0432.CCR-22-0696 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Stegmaier K, Wong JS, Ross KN, et al. Signature-based small molecule screening identifies cytosine arabinoside as an EWS/FLI modulator in Ewing sarcoma. Research Support, N.I.H., Extramural Research Support, Non-U.S. Gov’t. PLoS Med Apr 2007;4(4):e122. doi: 10.1371/journal.pmed.0040122 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Erkizan HV, Kong Y, Merchant M, et al. A small molecule blocking oncogenic protein EWS-FLI1 interaction with RNA helicase A inhibits growth of Ewing’s sarcoma. Research Support, N.I.H., Extramural Research Support, Non-U.S. Gov’t. Nat Med Jul 2009;15(7):750–6. doi: 10.1038/nm.1983 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sankar S, Theisen ER, Bearss J, et al. Reversible LSD1 inhibition interferes with global EWS/ETS transcriptional activity and impedes Ewing sarcoma tumor growth. Clin Cancer Res Sep 1 2014;20(17):4584–97. doi: 10.1158/1078-0432.CCR-14-0072 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Grohar PJ, Woldemichael GM, Griffin LB, et al. Identification of an Inhibitor of the EWS-FLI1 Oncogenic Transcription Factor by High-Throughput Screening. J Natl Cancer Inst Jun 22 2011;103(12):962–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Grohar PJ, Griffin LB, Yeung C, et al. Ecteinascidin 743 interferes with the activity of EWS-FLI1 in Ewing sarcoma cells. Neoplasia Feb 2011;13(2):145–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Osgood CL, Maloney N, Kidd CG, et al. Identification of Mithramycin Analogues with Improved Targeting of the EWS-FLI1 Transcription Factor. Clin Cancer Res Aug 15 2016;22(16):4105–18. doi: 10.1158/1078-0432.CCR-15-2624 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Harlow ML, Maloney N, Roland J, et al. Lurbinectedin Inactivates the Ewing Sarcoma Oncoprotein EWS-FLI1 by Redistributing It within the Nucleus. Cancer Res Nov 15 2016;76(22):6657–6668. doi: 10.1158/0008-5472.CAN-16-0568 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Harlow ML, Chasse MH, Boguslawski EA, et al. Trabectedin Inhibits EWS-FLI1 and Evicts SWI/SNF from Chromatin in a Schedule-dependent Manner. Clin Cancer Res Jun 1 2019;25(11):3417–3429. doi: 10.1158/1078-0432.CCR-18-3511 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Grohar P, Ballman K, Heise R, et al. ARC037: Results of phase I study of trabectedin given as a 1-hour (h) infusion in combination with low dose irinotecan in relapsed/refractory Ewing sarcoma (ES). J Clin Oncol 2023;41(suppl 16):abstr 11519. [Google Scholar]
- 43.Brohl AS, Sindiri S, Wei JS, et al. Immuno-transcriptomic profiling of extracranial pediatric solid malignancies. Cell Rep Nov 23 2021;37(8):110047. doi: 10.1016/j.celrep.2021.110047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Morales E, Olson M, Iglesias F, Luetkens T, Atanackovic D. Targeting the tumor microenvironment of Ewing sarcoma. Immunotherapy Dec 2021;13(17):1439–1451. doi: 10.2217/imt-2020-0341 [DOI] [PubMed] [Google Scholar]
- 45.Sayles LC, Breese MR, Koehne AL, et al. Genome-Informed Targeted Therapy for Osteosarcoma. Cancer Discov Jan 2019;9(1):46–63. doi: 10.1158/2159-8290.CD-17-1152 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Chen X, Bahrami A, Pappo A, et al. Recurrent somatic structural variations contribute to tumorigenesis in pediatric osteosarcoma. Cell Rep Apr 10 2014;7(1):104–12. doi: 10.1016/j.celrep.2014.03.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Perry JA, Kiezun A, Tonzi P, et al. Complementary genomic approaches highlight the PI3K/mTOR pathway as a common vulnerability in osteosarcoma. Proc Natl Acad Sci U S A Dec 23 2014;111(51):E5564–73. doi: 10.1073/pnas.1419260111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Picco G, Chen ED, Alonso LG, et al. Functional linkage of gene fusions to cancer cell fitness assessed by pharmacological and CRISPR-Cas9 screening. Nat Commun May 16 2019;10(1):2198. doi: 10.1038/s41467-019-09940-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kovac M, Blattmann C, Ribi S, et al. Exome sequencing of osteosarcoma reveals mutation signatures reminiscent of BRCA deficiency. Nat Commun Dec 3 2015;6:8940. doi: 10.1038/ncomms9940 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Rajan S, Zaccaria S, Cannon MV, et al. Structurally Complex Osteosarcoma Genomes Exhibit Limited Heterogeneity within Individual Tumors and across Evolutionary Time. Cancer Res Commun Apr 2023;3(4):564–575. doi: 10.1158/2767-9764.CRC-22-0348 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Rajan S, Franz EM, McAloney CA, et al. Osteosarcoma tumors maintain intra-tumoral transcriptional heterogeneity during bone and lung colonization. BMC Biol Apr 27 2023;21(1):98. doi: 10.1186/s12915-023-01593-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Morrow JJ, Bayles I, Funnell APW, et al. Positively selected enhancer elements endow osteosarcoma cells with metastatic competence. Nat Med Feb 2018;24(2):176–185. doi: 10.1038/nm.4475 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wu CC, Livingston JA. Genomics and the Immune Landscape of Osteosarcoma. Adv Exp Med Biol 2020;1258:21–36. doi: 10.1007/978-3-030-43085-6_2 [DOI] [PubMed] [Google Scholar]
- 54.Wang Y, Tian X, Zhang W, et al. Comprehensive Surfaceome Profiling to Identify and Validate Novel Cell-Surface Targets in Osteosarcoma. Mol Cancer Ther Jun 1 2022;21(6):903–913. doi: 10.1158/1535-7163.MCT-21-0836 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Womer RB, West DC, Krailo MD, et al. Randomized controlled trial of interval-compressed chemotherapy for the treatment of localized Ewing sarcoma: a report from the Children’s Oncology Group. J Clin Oncol Nov 20 2012;30(33):4148–54. doi: 10.1200/JCO.2011.41.5703 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Brennan B, Kirton L, Marec-Berard P, et al. Comparison of two chemotherapy regimens in patients with newly diagnosed Ewing sarcoma (EE2012): an open-label, randomised, phase 3 trial. Lancet Oct 29 2022;400(10362):1513–1521. doi: 10.1016/S0140-6736(22)01790-1 [DOI] [PubMed] [Google Scholar]
- 57.DuBois SG, Krailo MD, Buxton A, et al. Patterns of Translocation Testing in Patients Enrolling in a Cooperative Group Trial for Newly Diagnosed Metastatic Ewing Sarcoma. Arch Pathol Lab Med Dec 1 2021;145(12):1564–1568. doi: 10.5858/arpa.2020-0671-OA [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Whelan JS, Bielack SS, Marina N, et al. EURAMOS-1, an international randomised study for osteosarcoma: results from pre-randomisation treatment. Ann Oncol Feb 2015;26(2):407–14. doi: 10.1093/annonc/mdu526 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Guenther LM, Rowe RG, Acharya PT, et al. Response Evaluation Criteria in Solid Tumors (RECIST) following neoadjuvant chemotherapy in osteosarcoma. Pediatr Blood Cancer Apr 2018;65(4)doi: 10.1002/pbc.26896 [DOI] [PubMed]
- 60.Janeway K AJ C, Hingorani P, et al. AOST1321, a phase 2 trial of RANKL antibody, denosumab, in 2 cohorts of patients with recurrent or refractory osteosarcoma: A report from the Children’s Oncology Group. CTOS Annual Meeting 2019;
- 61.Isakoff MS, Goldsby R, Villaluna D, et al. A phase II study of eribulin in recurrent or refractory osteosarcoma: A report from the Children’s Oncology Group. Pediatr Blood Cancer Feb 2019;66(2):e27524. doi: 10.1002/pbc.27524 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hingorani P, Krailo M, Buxton A, et al. Phase 2 study of anti-disialoganglioside antibody, dinutuximab, in combination with GM-CSF in patients with recurrent osteosarcoma: A report from the Children’s Oncology Group. Eur J Cancer Sep 2022;172:264–275. doi: 10.1016/j.ejca.2022.05.035 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kopp LM, Malempati S, Krailo M, et al. Phase II trial of the glycoprotein non-metastatic B-targeted antibody-drug conjugate, glembatumumab vedotin (CDX-011), in recurrent osteosarcoma AOST1521: A report from the Children’s Oncology Group. Eur J Cancer Nov 2019;121:177–183. doi: 10.1016/j.ejca.2019.08.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.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 Feb 2012;23(2):508–16. doi: 10.1093/annonc/mdr151 [DOI] [PubMed] [Google Scholar]
- 65.Gaspar N, Campbell-Hewson Q, Gallego Melcon S, et al. Phase I/II study of single-agent lenvatinib in children and adolescents with refractory or relapsed solid malignancies and young adults with osteosarcoma (ITCC-050)(☆). ESMO Open Oct 2021;6(5):100250. doi: 10.1016/j.esmoop.2021.100250 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Duffaud F, Mir O, Boudou-Rouquette P, et al. Efficacy and safety of regorafenib in adult patients with metastatic osteosarcoma: a non-comparative, randomised, double-blind, placebo-controlled, phase 2 study. Lancet Oncol Jan 2019;20(1):120–133. doi: 10.1016/S1470-2045(18)30742-3 [DOI] [PubMed] [Google Scholar]
- 67.Italiano A, Mir O, Mathoulin-Pelissier S, et al. Cabozantinib in patients with advanced Ewing sarcoma or osteosarcoma (CABONE): a multicentre, single-arm, phase 2 trial. Lancet Oncol Mar 2020;21(3):446–455. doi: 10.1016/S1470-2045(19)30825-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Davis LE, Bolejack V, Ryan CW, et al. Randomized Double-Blind Phase II Study of Regorafenib in Patients With Metastatic Osteosarcoma. J Clin Oncol Jun 1 2019;37(16):1424–1431. doi: 10.1200/JCO.18.02374 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.De Noon S, Ijaz J, Coorens TH, et al. MYC amplifications are common events in childhood osteosarcoma. J Pathol Clin Res Sep 2021;7(5):425–431. doi: 10.1002/cjp2.219 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Jiang Y, Wang J, Sun M, et al. Multi-omics analysis identifies osteosarcoma subtypes with distinct prognosis indicating stratified treatment. Nat Commun Nov 23 2022;13(1):7207. doi: 10.1038/s41467-022-34689-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Marinoff AE, Spurr LF, Fong C, et al. Clinical Targeted Next-Generation Panel Sequencing Reveals MYC Amplification Is a Poor Prognostic Factor in Osteosarcoma. JCO Precis Oncol Mar 2023;7:e2200334. doi: 10.1200/PO.22.00334 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Reed D, Janeway K, Minard C, et al. PEPN1924, a phase 2 study of trastuzumab deruxtecan (DS-8201a, T-DXd) in adolescents and young adults with recurrent HER2+ osteosarcoma: A Children’s Oncology Group Pediatric Early-Phase Clinical Trial Network study. J Clin Oncol 2023;41(suppl 16):abstr 11527. [Google Scholar]
