?REL=0&showinfo=0&autohide=1
top of page
Search

A Step Forward in Realizing the Promise of Genomic Medicine for Childhood Rhabdomyosarcoma

Rhabdomyosarcoma (RMS) is a rare mesenchymal malignancy that is primarily a disease of children and adolescents with approximately 50% of patients being diagnosed in the first decade of life.1 Clinicians recognized early the value of histologic classification dividing RMS broadly into embryonal rhabdomyosarcoma (ERMS) and alveolar rhabdomyosarcoma (ARMS) subtypes with the former having a more favorable prognosis.2 The earliest known molecular finding in RMS was the loss of heterozygosity at chromosome 11p15.3 Soon after, almost 30 years ago, the discovery of the recurring translocations between chromosome 2 or 1 and chromosome 13 resulting in the fusion of the PAX3 or PAX7 gene with the FOXO1(FKHR) gene became pathognomonic of ARMS as it was seen in the majority of cases.4-6 In addition to loss of heterozygosity at 11p15 that was more frequently noted in ERMS, loss of imprinting at the same locus was noted in ARMS.7Subsequently, a smaller subset of alveolar histology that did not harbor the FOXO1 fusion was found to have a gene expression profile and a more favorable prognosis similar to ERMS.8,9 However, there was significant hesitancy for many years to classify RMS molecularly into two broad categories of FOXO1 fusion–positive (FP) or FOXO1 fusion–negative (FN) subgroups.10 It is relatively recent that sufficient data have been generated to broadly use this molecular classification together with Clinical Stage and Group.11,12 Other molecular findings described in RMS include copy number variations including gene amplifications, and point mutations culminating in a landmark report of the genomic landscape of childhood RMS13-15 and have been recently summarized.16 As such, ERMS may be considered a malignancy featured by point mutations and aneuploidy, whereas ARMS is a malignancy of gene fusions and amplifications. Pleomorphic rhabdomyosarcoma represents a third genomic form of RMS, with greater kinship to undifferentiated pleomorphic sarcoma than ERMS or ARMS.17,18 Pleomorphic rhabdomyosarcoma is found nearly exclusively in adults and is not studied in pediatric clinical trials. Over the past five decades, cooperative group trials in RMS using multiagent chemotherapy, surgery, and/or radiation therapy initially resulted in significant improvements in survival outcomes for patients with low-risk and intermediate-risk disease, whereas those with distant metastases continued to fare poorly; however, these outcomes have since plateaued.19 Given the young age of most patients with RMS and the use of radiation therapy for local tumor control, the burden of late effects of treatment in patients with RMS is immense.20,21 Molecular targeted agents have only recently been used in clinical trials in combination with classic cytotoxic chemotherapy for poor prognosis patients,22-24 and patient-specific strategies are yet to be incorporated. Traditional clinical and pathologic risk assignment results in a large majority of patients being lumped together in the Children's Oncology Group (COG) intermediate-risk group with some patient having a prognosis closer to low-risk patients and others closer to those with disseminated disease.25 Recent analysis has helped further delineate risk by incorporation of fusion status together with clinicopathologic features.26,27 In the article that accompanies this editorial, Shern et al28 report on genomic findings from 641 clinically annotated RMS patient tumor samples collected on COG, MMT, and RMS2005 trials spanning a 22-year period and analyze survival outcomes to identify additional genetic risk factors beyond FOXO1 fusion status that can potentially further refine risk stratification. Approximately 20% of samples had no known driver mutation. TP53 mutations were found at a higher incidence than previously reported and portended a poorer prognosis in both FP and FN tumors. MYOD1 mutations were restricted to FN tumors, were not limited to spindle or sclerosing histology, and were associated with a dismal prognosis. The magnitude of effect on prognosis appears to be greater for the presence of a MYOD1 rather than a TP53 mutation in FN RMS. As expected, gene amplification events involving CDK4 and MYC were observed in FP tumors, whereas RAS pathway mutations were noted in more than 50% of FN tumors. 37% of FN tumors had > 1 mutation and had a poorer prognosis compared with those with one or no candidate gene mutation in the COG cohort. The authors also propose a risk stratification incorporating MYOD1 and TP53 mutation status where any MYOD1-mutant tumor and TP53-mutant tumor irrespective of clinical features are classified as very or ultra-high risk (expected long-term failure-free survival [FFS] < 20%); stage 4, clinical group IV FN tumor with > 1 metastatic site, any stage 4, clinical group IV FP TP53 wild-type tumor, or any TP53-mutant tumor that is not low risk or very high risk as high risk (expected FFS 20 to < 40%); any stage, clinical group III nonorbit primary, FN stage 3, clinical group I or II, FN stage 4, clinical group IV with one metastatic site, FN low-risk TP53-mutant FN tumors and FP stage 1-3, clinical group I-III tumors that are TP53 wild-type as intermediate risk (FFS 60%-75%); and stage 1-2, clinical group I-II and stage 1, clinical group III orbit primary FN tumors as low risk (FFS > 85%). This collaborative effort between the COG and researchers in the United Kingdom is to be applauded as it allows for further refinement of risk stratification for RMS, hopefully leading to more precision in determining risk groups allowing for potential opportunities to affect outcomes by improving survival, decreasing morbidity, and considering targeted therapies that are more patient specific. These data have several limitations. They were generated from 470 (14.7%) of 3,184 unique patients treated on seven therapeutic trials and enhanced by 171 samples from patients enrolled on D9902, a biology study where limited therapeutic information was available. This data set should therefore be considered a convenience cohort, the deficiencies of which have been eloquently discussed previously.29 This is likely why mutations reported previously in RMS such as MTOR, ALK, SOS2, PDGFRA, BRAF, AKT, and others were not detected in this study. It may also be the reason why > 1 mutation was not associated with inferior outcomes in the United Kingdom cohort. Therapy delivered is a critical variable in determining outcome. During the time covered by the clinical trials that provided the clinically annotated tumor samples, similar risk groups had significantly different FFS particularly in ERMS/FN RMS with the caveat that the comparisons were with historical controls.30-32 ERMS and FN RMS appear to benefit from alkylator intensification by dose or duration,33-35 whereas therapy seems to be a less important variable in FP RMS.27 Paired germline testing was not performed, and therefore, the contribution of germline mutations in TP53 and other known cancer predisposition genes to outcome could not be assessed. As such, these data need to be validated prospectively and is planned for the next COG low-risk RMS trial. Nevertheless, this report opens many opportunities in addition to more precise risk stratification to be considered in improving the outcomes of patients with RMS. Given that only approximately 350 patients are diagnosed with RMS each year in the United States (US) and a similar number of patients in Western Europe, it is going to be almost impossible to conduct traditional randomized phase III clinical trials with cytotoxic chemotherapy given the number of patients needed and the duration to accrue them. Furthermore, there have been only four positive randomized clinical trials to date conducted in RMS.24,36-38 The recently formed International Soft-Tissue Sarcoma Database Consortium (INSTRuCT), a collaboration of the COG Soft-Tissue Sarcoma Committee, the European Pediatric Soft-Tissue Sarcoma Study Group, and the Cooperative Weichteilsarkom Studiengruppe ,will help immensely in the harmonization of data collection and its prospective implementation across cooperative groups for RMS clinical trials. This will result in more robust data sets to analyze for outcomes across clinical trials including providing opportunities to investigate RMS gene signatures,39,40 allowing for meaningful tools to risk stratify or assign a specific treatment. Compelling questions to be addressed in RMS include the possibility of minimizing late effects of treatment, targeting the FOXO1 fusion, targeting other genetic and epigenetic aberrations, and addressing germline mutations and tumor heterogeneity, all this while at least maintaining outcomes achieved by surgery and/or radiation and multiagent chemotherapy. There are several examples in medical oncology where molecular targeted therapy has been shown to be superior to chemotherapy including targeting epidermal growth factor receptor with gefitinib in pulmonary adenocarcinoma41 and ALK with crizotinib in anaplastic lymphoma kinase–positive lung cancer,42 encorafenib, binimetinib, and cetuximab in BRAF V600E–mutated colorectal cancer,43 and alpelisib in PIK3CA-mutated, hormone receptor–positive advanced breast cancer.44 This was achievable in part given the low progression-free survival on chemotherapy alone in these cancers. It is unclear whether simple intensification of chemotherapy may improve outcomes for patients with MYOD1- or TP53-mutated RMS. It is highly unlikely that a single or dual molecular targeted agent will prove superior to standard chemotherapy and radiation in RMS. Significant response rates and improved progression-free survival have been reported in cancers that are driven by fusions, notably tropomyosin receptor kinase fusions in both adults and children,45-47 and rearranged-during-transfection alterations in adults.48,49 However, targeting the FOXO1 fusion in RMS has been elusive thus far. A recent publication describing germline cancer predisposition variants in 615 patients with pediatric RMS found 7.3% to harbor a molecular finding consistent with a cancer predisposition including TP53, NF-1, and HRAS mutations, which were also reported as somatic mutations in RMS. The use of poly ADP-ribose polymerase inhibition has been shown to be beneficial in metastatic human epidermal growth factor receptor 2–negative breast cancer patients with BRCA mutations,50 raising the possibility of harnessing germline mutations to inform treatment of RMS patients with cancer predisposition. Targeting the BCR-ABL gene fusion in childhood Philadelphia chromosome–positive acute lymphoblastic anemia with imatinib together with standard cytotoxic chemotherapy significantly improved the survival outcomes of these poor prognosis patients.51 A similar approach targeting a genetic alteration in RMS together with standard treatment may lead to improved outcomes. Another avenue to pursue could be to target > 1 mutation when indicated. For example, combination therapy with trametinib and alpelisib may be considered in a patient whose RMS harbors an RAS and PIK3CA mutation. Finally, limiting the morbidity of late effects in patients with RMS is still worthwhile. Observation following resection of stage 1 testicular germ cell tumors is standard of care in pediatrics with a 100% salvage rate with standard chemotherapy, thus eliminating the risk of ototoxicity, pulmonary toxicity, and second malignancy in most of the stage 1 patients. Although chemotherapy is universally recommended for all patients with RMS at this time, a future consideration within the context of a clinical trial may be observation for a selected group of patients with RMS age < 10 years with stage 1, clinical group I small primary FN RMS tumors that do not harbor a deleterious genetic mutation. Given the limited number of patients diagnosed each year with RMS, the report by Shern et al28 paves the way for a possible international collaborative umbrella clinical trial in newly diagnosed patients with RMS, seeking to improve outcomes by tailoring therapy according to a genomic classification and targeting molecular alterations as feasible to improve the overall survival—a definite step forward in realizing the promise of genomic medicine for childhood RMS. © 2021 by American Society of Clinical OncologySee accompanying article doi:10.1200/JCO.20.03060 DISCLAIMERThe author is a member of the Soft Tissue Sarcoma Committee of the Children's Oncology Group (COG). This editorial is not meant to reflect the views of the COG Soft Tissue Sarcoma Committee. SUPPORTSupported by NIH/NCI 2U10CA180886-06; Associates Sarcoma Program Chair. AUTHOR'S DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST A Step Forward in Realizing the Promise of Genomic Medicine for Childhood Rhabdomyosarcoma The following represents disclosure information provided by the author of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO’s conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/jco/authors/author-center. Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments). Leo Mascarenhas Consulting or Advisory Role: Bayer Speakers' Bureau: Bayer Research Funding: AstraZeneca/MedImmune, Lilly, Bayer, Loxo, Salarius Pharmaceuticals, Turning Point Therapeutics, Pfizer, Incyte, Amgen Travel, Accommodations, Expenses: Bayer, Lilly, Thermo Fisher Scientific, Salarius Pharmaceuticals Uncompensated Relationships: Children's Oncology Group Foundation, The Pablove Foundation, American Society of Pediatric Hematology/Oncology No other potential conflicts of interest were reported. REFERENCES ChooseTop of pageREFERENCES << 1.Gurney JG, Young JL, Roffers SD, et al: Soft Tissue Sarcomas. Bethesda, MD, NIH, 1999Google Scholar2.Newton WA Jr, Gehan EA, Webber BL, et al: Classification of rhabdomyosarcomas and related sarcomas. Pathologic aspects and proposal for a new classification—An Intergroup Rhabdomyosarcoma Study. Cancer 76:1073-1085, 1995 Crossref, Medline, Google Scholar3.Loh WE Jr, Scrable HJ, Livanos E, et al: Human chromosome 11 contains two different growth suppressor genes for embryonal rhabdomyosarcoma. Proc Natl Acad Sci USA 89:1755-1759, 1992 Crossref, Medline, Google Scholar4.Barr FG, Galili N, Holick J, et al: Rearrangement of the PAX3 paired box gene in the paediatric solid tumour alveolar rhabdomyosarcoma. Nat Genet 3:113-117, 1993 Crossref, Medline, Google Scholar5.Davis RJ, D'Cruz CM, Lovell MA, et al: Fusion of PAX7 to FKHR by the variant t(1;13)(p36;q14) translocation in alveolar rhabdomyosarcoma. Cancer Res 54:2869-2872, 1994Medline, Google Scholar6.Biegel JA, Nycum LM, Valentine V, et al: Detection of the t(2;13)(q35;q14) and PAX3-FKHR fusion in alveolar rhabdomyosarcoma by fluorescence in situ hybridization. Genes Chromosomes Cancer 12:186-192, 1995 Crossref, Medline, Google Scholar7.Anderson J, Gordon A, McManus A, et al: Disruption of imprinted genes at chromosome region 11p15.5 in paediatric rhabdomyosarcoma. Neoplasia 1:340-348, 1999 Crossref, Medline, Google Scholar8.Davicioni E, Anderson MJ, Finckenstein FG, et al: Molecular classification of rhabdomyosarcoma—Genotypic and phenotypic determinants of diagnosis: A report from the Children's Oncology Group. Am J Pathol 174:550-564, 2009 Crossref, Medline, Google Scholar9.Williamson D, Missiaglia E, de Reynies A, et al: Fusion gene-negative alveolar rhabdomyosarcoma is clinically and molecularly indistinguishable from embryonal rhabdomyosarcoma. J Clin Oncol 28:2151-2158, 2010 Link, Google Scholar10.Anderson JR, Barr FG, Hawkins DS, et al: Fusion-negative alveolar rhabdomyosarcoma: Modification of risk stratification is premature. J Clin Oncol 28:e587-e588, 2010; author reply e589-e590 Link, Google Scholar11.Missiaglia E, Williamson D, Chisholm J, et al: PAX3/FOXO1 fusion gene status is the key prognostic molecular marker in rhabdomyosarcoma and significantly improves current risk stratification. J Clin Oncol 30:1670-1677, 2012 Link, Google Scholar12.Skapek SX, Anderson J, Barr FG, et al: PAX-FOXO1 fusion status drives unfavorable outcome for children with rhabdomyosarcoma: A Children's Oncology Group report. Pediatr Blood Cancer 60:1411-1417, 2013 Crossref, Medline, Google Scholar13.Weber-Hall S, Anderson J, McManus A, et al: Gains, losses, and amplification of genomic material in rhabdomyosarcoma analyzed by comparative genomic hybridization. Cancer Res 56:3220-3224, 1996 Medline, Google Scholar14.Bridge JA, Liu J, Qualman SJ, et al: Genomic gains and losses are similar in genetic and histologic subsets of rhabdomyosarcoma, whereas amplification predominates in embryonal with anaplasia and alveolar subtypes. Genes Chromosomes Cancer 33:310-321, 2002 Crossref, Medline, Google Scholar15.Shern JF, Chen L, Chmielecki J, et al: Comprehensive genomic analysis of rhabdomyosarcoma reveals a landscape of alterations affecting a common genetic axis in fusion-positive and fusion-negative tumors. Cancer Discov 4:216-231, 2014 Crossref, Medline, Google Scholar16.Skapek SX, Ferrari A, Gupta AA, et al: Rhabdomyosarcoma. Nat Rev Dis Primers 5:1, 2019Crossref, Medline, Google Scholar17.Leiner J, Le Loarer F: The current landscape of rhabdomyosarcomas: An update. Virchows Arch 476:97-108, 2020 Crossref, Medline, Google Scholar18.Bompas E, Campion L, Italiano A, et al: Outcome of 449 adult patients with rhabdomyosarcoma: An observational ambispective nationwide study. Cancer Med 7:4023-4035, 2018 Crossref, Medline, Google Scholar19.Arndt CAS, Bisogno G, Koscielniak E: Fifty years of rhabdomyosarcoma studies on both sides of the pond and lessons learned. Cancer Treat Rev 68:94-101, 2018 Crossref, Medline, Google Scholar20.Oeffinger KC, Mertens AC, Sklar CA, et al: Chronic health conditions in adult survivors of childhood cancer. N Engl J Med 355:1572-1582, 2006 Crossref, Medline, Google Scholar21.Stevens MC: Treatment for childhood rhabdomyosarcoma: The cost of cure. Lancet Oncol 6:77-84, 2005 Crossref, Medline, Google Scholar22.Chisholm JC, Merks JHM, Casanova M, et al: Open-label, multicentre, randomised, phase II study of the EpSSG and the ITCC evaluating the addition of bevacizumab to chemotherapy in childhood and adolescent patients with metastatic soft tissue sarcoma (the BERNIE study). Eur J Cancer 83:177-184, 2017 Crossref, Medline, Google Scholar23.Malempati S, Weigel BJ, Chi YY, et al: The addition of cixutumumab or temozolomide to intensive multiagent chemotherapy is feasible but does not improve outcome for patients with metastatic rhabdomyosarcoma: A report from the Children's Oncology Group. Cancer 125:290-297, 2019 Crossref, Medline, Google Scholar24.Mascarenhas L, Chi YY, Hingorani P, et al: Randomized phase II trial of bevacizumab or temsirolimus in combination with chemotherapy for first relapse rhabdomyosarcoma: A report from the Children's Oncology Group. J Clin Oncol 37:2866-2874, 2019 Link, Google Scholar25.Meza JL, Anderson J, Pappo AS, et al: Analysis of prognostic factors in patients with nonmetastatic rhabdomyosarcoma treated on intergroup rhabdomyosarcoma studies III and IV: The Children's Oncology Group. J Clin Oncol 24:3844-3851, 2006 Link, Google Scholar26.Hibbitts E, Chi YY, Hawkins DS, et al: Refinement of risk stratification for childhood rhabdomyosarcoma using FOXO1 fusion status in addition to established clinical outcome predictors: A report from the Children's Oncology Group. Cancer Med 8:6437-6448, 2019Crossref, Medline, Google Scholar27.Heske CM, Chi YY, Venkatramani R, et al: Survival outcomes of patients with localized FOXO1 fusion-positive rhabdomyosarcoma treated on recent clinical trials: A report from the soft tissue sarcoma Committee of the Children's Oncology Group. Cancer 127:946-956, 2021 Crossref, Medline, Google Scholar28.Shern JF, Selfe J, Izquierdo E, et al: Genomic classification and clinical outcome in rhabdomyosarcoma: A report from an International Consortium. J Clin Oncol doi:10.1200/JCO.20.03060 [epub ahead of print on June 24, 2021] Abstract, Google Scholar29.Rosenberg AR, Skapek SX, Hawkins DS: The inconvenience of convenience cohorts: Rhabdomyosarcoma and the PAX-FOXO1 biomarker. Cancer Epidemiol Biomarkers Prev 21:1012-1018, 2012 Crossref, Medline, Google Scholar30.Walterhouse DO, Pappo AS, Meza JL, et al: Reduction of cyclophosphamide dose for patients with subset 2 low-risk rhabdomyosarcoma is associated with an increased risk of recurrence: A report from the Soft Tissue Sarcoma Committee of the Children's Oncology Group. Cancer 123:2368-2375, 2017 Crossref, Medline, Google Scholar31.Hawkins DS, Chi YY, Anderson JR, et al: Addition of vincristine and irinotecan to vincristine, dactinomycin, and cyclophosphamide does not improve outcome for intermediate-risk rhabdomyosarcoma: A report from the Children's Oncology Group. J Clin Oncol 36:2770-2777, 2018 Link, Google Scholar32.Casey DL, Chi YY, Donaldson SS, et al: Increased local failure for patients with intermediate-risk rhabdomyosarcoma on ARST0531: A report from the Children's Oncology Group. Cancer 125:3242-3248, 2019 Crossref, Medline, Google Scholar33.Weigel BJ, Lyden E, Anderson JR, et al: Intensive multiagent therapy, including dose-compressed cycles of ifosfamide/etoposide and vincristine/doxorubicin/cyclophosphamide, irinotecan, and radiation, in patients with high-risk rhabdomyosarcoma: A report from the Children's Oncology Group. J Clin Oncol 34:117-122, 2016 Link, Google Scholar34.Arndt CA, Stoner JA, Hawkins DS, et al: Vincristine, actinomycin, and cyclophosphamide compared with vincristine, actinomycin, and cyclophosphamide alternating with vincristine, topotecan, and cyclophosphamide for intermediate-risk rhabdomyosarcoma: Children's Oncology Group study D9803. J Clin Oncol 27:5182-5188, 2009 Link, Google Scholar35.Gallego S, Chi YY, De Salvo GL, et al: Alveolar rhabdomyosarcoma with regional nodal involvement: Results of a combined analysis from two cooperative groups. Pediatr Blood Cancer 68:e28832, 2021 Crossref, Medline, Google Scholar36.Heyn RM, Holland R, Newton WA Jr, et al: The role of combined chemotherapy in the treatment of rhabdomyosarcoma in children. Cancer 34:2128-2142, 1974 Crossref, Medline, Google Scholar37.Bisogno G, De Salvo GL, Bergeron C, et al: Vinorelbine and continuous low-dose cyclophosphamide as maintenance chemotherapy in patients with high-risk rhabdomyosarcoma (RMS 2005): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 20:1566-1575, 2019 Crossref, Medline, Google Scholar38.Defachelles AS, Bogart E, Casanova M, et al: Randomized phase 2 trial of the combination of vincristine and irinotecan with or without temozolomide, in children and adults with refractory or relapsed rhabdomyosarcoma (RMS). J Clin Oncol 37, 2019 (suppl; abstr 10000) Link, Google Scholar39.Hingorani P, Missiaglia E, Shipley J, et al: Clinical application of prognostic gene expression signature in fusion gene-negative rhabdomyosarcoma: A report from the Children's Oncology Group. Clin Cancer Res 21:4733-4739, 2015 Crossref, Medline, Google Scholar40.Xu L, Zheng Y, Liu J, et al: Integrative Bayesian analysis identifies rhabdomyosarcoma disease genes. Cell Rep 24:238-251, 2018 Crossref, Medline, Google Scholar41.Mok TS, Wu YL, Thongprasert S, et al: Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 361:947-957, 2009 Crossref, Medline, Google Scholar42.Shaw AT, Kim DW, Nakagawa K, et al: Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med 368:2385-2394, 2013 Crossref, Medline, Google Scholar43.Kopetz S, Grothey A, Yaeger R, et al: Encorafenib, binimetinib, and cetuximab in BRAF V600e-mutated colorectal cancer. N Engl J Med 381:1632-1643, 2019 Crossref, Medline, Google Scholar44.Andre F, Ciruelos E, Rubovszky G, et al: Alpelisib for PIK3CA-mutated, hormone receptor-positive advanced breast cancer. N Engl J Med 380:1929-1940, 2019 Crossref, Medline, Google Scholar45.Drilon A, Laetsch TW, Kummar S, et al: Efficacy of larotrectinib in TRK fusion-positive cancers in adults and children. N Engl J Med 378:731-739, 2018 Crossref, Medline, Google Scholar46.Laetsch TW, DuBois SG, Mascarenhas L, et al: Larotrectinib for paediatric solid tumours harbouring NTRK gene fusions: Phase 1 results from a multicentre, open-label, phase 1/2 study. Lancet Oncol 19:705-714, 2018 Crossref, Medline, Google Scholar47.Hong DS, DuBois SG, Kummar S, et al: Larotrectinib in patients with TRK fusion-positive solid tumours: A pooled analysis of three phase 1/2 clinical trials. Lancet Oncol 21:531-540, 2020 Crossref, Medline, Google Scholar48.Drilon A, Oxnard GR, Tan DSW, et al: Efficacy of selpercatinib in RET fusion-positive non-small-cell lung cancer. N Engl J Med 383:813-824, 2020 Crossref, Medline, Google Scholar49.Wirth LJ, Sherman E, Robinson B, et al: Efficacy of selpercatinib in RET-altered thyroid cancers. N Engl J Med 383:825-835, 2020 Crossref, Medline, Google Scholar50.Robson M, Im SA, Senkus E, et al: Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med 377:523-533, 2017 Crossref, Medline, Google Scholar51.Schultz KR, Bowman WP, Aledo A, et al: Improved early event-free survival with imatinib in Philadelphia chromosome-positive acute lymphoblastic leukemia: A Children's Oncology Group study. J Clin Oncol 27:5175-5181, 2009 Link, Google Scholar OPTIONS & TOOLS Export CitationTrack CitationAdd To Favorites Purchase

  • Rights & Permissions

COMPANION ARTICLES

ARTICLE CITATION DOI: 10.1200/JCO.21.01296 Journal of Clinical Oncology Published online June 28, 2021. PMID: 34181486 WE RECOMMEND

  1. Reply to S. Stegmaier et alEdoardo Missiaglia et al., J Clin Oncol, 2012

  2. Rhabdomyosarcoma, Ewing Sarcoma, and Other Round Cell SarcomasAlberto S. Pappo et al., J Clin Oncol, 2017

  3. Fusion-Negative Alveolar Rhabdomyosarcoma: Modification of Risk Stratification Is PrematureJames R. Anderson et al., J Clin Oncol, 2010

  4. Diagnosing Alveolar Rhabdomyosarcoma: Morphology Must Be Coupled With Fusion ConfirmationLeonard H. Wexler et al., J Clin Oncol, 2010

  5. Reply to J.R. Anderson et alDaniel Williamson et al., J Clin Oncol, 2010


  1. An epigenetic tactic to treat RMSLeslie K. Ferrarelli, Sci Signal, 2017

  2. Results of RS-99 protocol for childhood solid tumorsJiao-Yang Cai et al., World Journal of Pediatrics, 2010

  3. Treatment of pediatric average-risk medulloblastoma using craniospinal irradiation less than 2500 cGy and chemotherapy: single center experience in KoreaJong Hyung Yoon et al., World Journal of Pediatrics, 2017

  4. The HDAC3–SMARCA4–miR-27a axis promotes expression of the PAX3:FOXO1 fusion oncogene in rhabdomyosarcomaNarendra Bharathy et al., Sci Signal, 2018

  5. People in the NewsGenomeWeb, 2010






56 views0 comments

Recent Posts

See All
bottom of page