Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Molecular profiling identifies targeted therapy opportunities in pediatric solid cancer

Abstract

To evaluate the clinical impact of molecular tumor profiling (MTP) with targeted sequencing panel tests, pediatric patients with extracranial solid tumors were enrolled in a prospective observational cohort study at 12 institutions. In the 345-patient analytical population, median age at diagnosis was 12 years (range 0–27.5); 298 patients (86%) had 1 or more alterations with potential for impact on care. Genomic alterations with diagnostic, prognostic or therapeutic significance were present in 61, 16 and 65% of patients, respectively. After return of the results, impact on care included 17 patients with a clarified diagnostic classification and 240 patients with an MTP result that could be used to select molecularly targeted therapy matched to identified alterations (MTT). Of the 29 patients who received MTT, 24% had an objective response or experienced durable clinical benefit; all but 1 of these patients received targeted therapy matched to a gene fusion. Of the diagnostic variants identified in 209 patients, 77% were gene fusions. MTP with targeted panel tests that includes fusion detection has a substantial clinical impact for young patients with solid tumors.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Relationship between genes containing actionable variants and the drug class of the iCat recommendation.
Fig. 2: Summary infographic of the outcome for the 345 patients in the analytical cohort after return of genomic results with diagnostic or therapeutic significance.
Fig. 3: Swimmer plot of treatment response for 29 patients who received MTT.
Fig. 4

Similar content being viewed by others

Data availability

A Supplementary Data file contains all SNVs, CNVs and fusions identified and TMB and mismatch repair status determined by the DNA or RNA panel tests. The Supplementary Data file also includes clinical interpretation of variants determined to have prognostic or diagnostic significance and the associated AMP/CAP/ASCO tier. All iCat recommendations are provided in the Supplementary Data including the iCat tier at the time of the clinical interpretation report and at the time of re-tiering.

Code availability

There is no relevant code provided for this study.

References

  1. Brandão, M., Caparica, R., Eiger, D. & de Azambuja, E. Biomarkers of response and resistance to PI3K inhibitors in estrogen receptor-positive breast cancer patients and combination therapies involving PI3K inhibitors. Ann. Oncol. 30, x27–x42 (2019).

    Article  PubMed  Google Scholar 

  2. Drilon, A. et al. Efficacy of larotrectinib in TRK fusion-positive cancers in adults and children. N. Engl. J. Med. 378, 731–739 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. Havel, J. J., Chowell, D. & Chan, T. A. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat. Rev. Cancer 19, 133–150 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Le, D. T. et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357, 409–413 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Lindeman, N. I. et al. Updated molecular testing guideline for the selection of lung cancer patients for treatment with targeted tyrosine kinase inhibitors: guideline from the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. J. Thorac. Oncol. 13, 323–358 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. FDA Fact Sheet: CDRH’S Approach to Tumor Profiling Next Generation Sequencing Tests (US Food & Drug Administration, accessed May 23, 2022); https://www.fda.gov/media/109050/download

  7. Guidelines Version 4.2021 Non-Small Cell Lung Cancer. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) Vol. 2021 (National Comprehensive Cancer Network, 2021).

  8. Next Generation Sequencing (NGS) for Medicare Beneficiaries with Advanced Cancer (CAG-00450N) (CMS.gov, 2018).

  9. Laetsch, T. W. & Hawkins, D. S. Larotrectinib for the treatment of TRK fusion solid tumors. Expert Rev. Anticancer Ther. 19, 1–10 (2019).

    Article  CAS  PubMed  Google Scholar 

  10. Mosele, F. et al. Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group. Ann. Oncol. 31, 1491–1505 (2020).

    Article  CAS  PubMed  Google Scholar 

  11. Li, W. et al. Trends in molecular testing of lung cancer in mainland People’s Republic of China over the decade 2010 to 2019. JTO Clin. Res. Rep. 2, 100163 (2021).

    PubMed Central  PubMed  Google Scholar 

  12. AACR Project GENIE: Data (American Association for Cancer Research, 2022).

  13. Chmielecki, J. et al. Genomic profiling of a large set of diverse pediatric cancers identifies known and novel mutations across tumor spectra. Cancer Res. 77, 509–519 (2017).

    Article  CAS  PubMed  Google Scholar 

  14. Parsons, D. W. et al. Actionable tumor alterations and treatment protocol enrollment of pediatric and young adult patients with refractory cancers in the National Cancer Institute-Children’s Oncology Group Pediatric MATCH Trial. J. Clin. Oncol. https://doi.org/10.1200/JCO.21.02838 (2022).

  15. Data Sharing Opportunities in Childhood, Adolescent and Young Adult (AYA) Cancer Research for the National Cancer Institute: Report of the Board of Scientific Advisors on the Childhood Cancer Data Initiative (CCDI) (National Institutes of Health, National Cancer Institute Board of Scientific Advisors, 2020).

  16. van Tilburg, C. M. et al. The pediatric precision oncology INFORM registry: clinical outcome and benefit for patients with very high-evidence targets. Cancer Discov. 11, 2764–2779 (2021).

    Article  PubMed  Google Scholar 

  17. Wong, M. et al. Whole genome, transcriptome and methylome profiling enhances actionable target discovery in high-risk pediatric cancer. Nat. Med. 26, 1742–1753 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Harris, M. H. et al. Multicenter feasibility study of tumor molecular profiling to inform therapeutic decisions in advanced pediatric solid tumors: the Individualized Cancer Therapy (iCat) study. JAMA Oncol. 2, 608–615 (2016).

    Article  PubMed  Google Scholar 

  19. Leijen, S. et al. Phase I study evaluating WEE1 Inhibitor AZD1775 as monotherapy and in combination with gemcitabine, cisplatin, or carboplatin in patients with advanced solid tumors. J. Clin. Oncol. 34, 4371–4380 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Allen, C. E. et al. Selumetinib in patients with tumors with MAPK pathway alterations: results from Arm E of the NCI-COG pediatric MATCH trial. J. Clin. Oncol. 39, 10008 (2021).

    Article  Google Scholar 

  21. Eisenhauer, E. A. et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer 45, 228–247 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Ortiz, M. V. et al. Activity of the highly specific RET inhibitor selpercatinib (LOXO-292) in pediatric patients with tumors harboring RET gene alterations. JCO Precis. Oncol. 4, PO.19.00401 (2020).

    PubMed Central  PubMed  Google Scholar 

  23. Hillier, K. et al. A novel ALK fusion in pediatric medullary thyroid carcinoma. Thyroid 29, 1704–1707 (2019).

    Article  CAS  PubMed  Google Scholar 

  24. Li, M. M. et al. Standards and guidelines for the interpretation and reporting of sequence variants in cancer: a joint consensus recommendation of the Association for Molecular Pathology, American Society of Clinical Oncology, and College of American Pathologists. J. Mol. Diagn. 19, 4–23 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Oliveira, A. M. & Chou, M. M. USP6-induced neoplasms: the biologic spectrum of aneurysmal bone cyst and nodular fasciitis. Hum. Pathol. 45, 1–11 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Chen, X. et al. Recurrent somatic structural variations contribute to tumorigenesis in pediatric osteosarcoma. Cell Rep. 7, 104–112 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. Thyroid Carcinoma, Version 3.2021. NCCN Practice Guidelines in Oncology (National Comprehensive Cancer Network, 2021).

  28. Neuroendocrine and Adrenal Tumors, Version 4.2021. NCCN Clinical Practice Guidelines in Oncology (National Comprehensive Cancer Network, 2021).

  29. Bone Cancer, Version 2.2022. NCCN Practice Guidelines in Oncology (National Comprehensive Cancer Network, 2022).

  30. Hepatobiliary Cancers, Version 5.2021. NCCN Practice Guidelines in Oncology (National Comprehensive Cancer Network, 2021).

  31. Soft Tissue Sarcoma, Version 3.2021. NCCN Practice Guidelines in Oncology (National Comprehensive Cancer Network, 2022).

  32. Oberg, J. A. et al. Implementation of next generation sequencing into pediatric hematology-oncology practice: moving beyond actionable alterations. Genome Med. 8, 133 (2016).

    Article  PubMed  Google Scholar 

  33. Inaba, H. & Mullighan, C. G. Pediatric acute lymphoblastic leukemia. Haematologica 105, 2524–2539 (2020).

    Article  CAS  PubMed  Google Scholar 

  34. Ramaswamy, V. et al. Risk stratification of childhood medulloblastoma in the molecular era: the current consensus. Acta Neuropathol. 131, 821–831 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Khater, F. et al. Molecular profiling of hard-to-treat childhood and adolescent cancers. JAMA Netw. Open 2, e192906 (2019).

    Article  PubMed  Google Scholar 

  36. Schienda, J. et al. Germline sequencing improves tumor-only sequencing interpretation in a precision genomic study of patients with pediatric solid tumor. JCO Precis. Oncol. 5, PO.21.00281 (2021).

    PubMed Central  PubMed  Google Scholar 

  37. Parsons, D. W. et al. Diagnostic yield of clinical tumor and germline whole-exome sequencing for children with solid tumors. JAMA Oncol. 2, 616–624 (2016).

    Article  PubMed  Google Scholar 

  38. Fiala, E. M. et al. Prospective pan-cancer germline testing using MSK-IMPACT informs clinical translation in 751 patients with pediatric solid tumors. Nat. Cancer 2, 357–365 (2021).

    Article  CAS  PubMed  Google Scholar 

  39. Zhang, J. et al. Germline mutations in predisposition genes in pediatric cancer. N. Engl. J. Med. 373, 2336–2346 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. NCCR*Explorer: An Interactive Website for NCCR Cancer Statistics (National Cancer Institute, 2021).

  41. Parsons, D. W. et al. Identification of targetable molecular alterations in the NCI-COG Pediatric MATCH trial. J. Clin. Oncol. 37, 10011 (2019).

    Article  Google Scholar 

  42. FDA Reauthorization Act of 2017 (FDARA) (US Food & Drug Administration, 2017).

  43. Janes, M. R. et al. Targeting KRAS mutant cancers with a covalent G12C-specific inhibitor. Cell 172, 578–589.e17 (2018).

    Article  CAS  PubMed  Google Scholar 

  44. Chen, Y.-N. et al. Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases. Nature 535, 148–152 (2016).

    Article  CAS  PubMed  Google Scholar 

  45. International Classification of Diseases for Oncology (ICD-O) (World Health Organization, 2020).

  46. Abo, R. P. et al. BreaKmer: detection of structural variation in targeted massively parallel sequencing data using kmers. Nucleic Acids Res. 43, e19 (2015).

    Article  PubMed  Google Scholar 

  47. Garcia, E. P. et al. Validation of OncoPanel: a targeted next-generation sequencing assay for the detection of somatic variants in cancer. Arch. Pathol. Lab. Med. 141, 751–758 (2017).

    Article  CAS  PubMed  Google Scholar 

  48. Nowak, J. A. et al. Detection of mismatch repair deficiency and microsatellite instability in colorectal adenocarcinoma by targeted next-generation sequencing. J. Mol. Diagn. 19, 84–91 (2017).

    Article  CAS  PubMed  Google Scholar 

  49. Sholl, L. M. et al. Institutional implementation of clinical tumor profiling on an unselected cancer population. JCI Insight 1, e87062 (2016).

    Article  PubMed  Google Scholar 

  50. Paulson, V. A. et al. Recurrent and novel USP6 fusions in cranial fasciitis identified by targeted RNA sequencing. Mod. Pathol. 33, 775–780 (2020).

    Article  CAS  PubMed  Google Scholar 

  51. Haas, B. J. et al. Accuracy assessment of fusion transcript detection via read-mapping and de novo fusion transcript assembly-based methods. Genome Biol. 20, 213 (2019).

    Article  PubMed  Google Scholar 

  52. Rodríguez-Martín, B. et al. ChimPipe: accurate detection of fusion genes and transcription-induced chimeras from RNA-seq data. BMC Genomics 18, 7 (2017).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Funding for this study was provided by the Precision For Kids Pan Mass Challenge Team, the 4C’s Fund, Lamb Family Fund, C&S Wholesale Grocers and C&S Charities and Alexandra Simpson Pediatric Research Fund. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. We also thank the study participants and their families and the research coordinators at each of the study sites.

Author information

Authors and Affiliations

Authors

Contributions

A.J.C., N.P., L.M., T.W.L., A.K., S.I.C., M.E.M., M.A.A., R.B., A.J.S., D.A.W., M.H.H., J.L.G-B., C.M.C., B.D.C., J.K., L.E.M., S.L.V., N.I.L., E.V.A., S.G.D., W.B.L. and K.A.J. contributed to study design. A.J.C., L.B.C., P-C.K., A.I-T., D.R., D.D., W.K., N.P., L.M., T.W.L., A.K., S.I.C., M.E.M., M.A.A., R.B., A.J.S., D.A.W., J.L.G-B., A.C.H., J.H., H.H., D.M., A.A., G.R.S., L.A.L., R.S.P., L.L., M.V.H., N.J.L., S.C., H.C., M.H.H., S.J.F., C.M.C., B.D.C., J.K., L.E.M., N.I.L., E.V.A., S.G.D. and K.A.J. obtained regulatory approval and performed study conduct, patient recruitment and consent, sequencing, clinical interpretation and clinical data collection. A.J.C., L.B.C., P-C.K., A.I-T., D.D., W.K., H.H., L.L., Y.L., H.G., A.D.C., Y-C.L., H.C., S.J.F., L.E.M., N.I.L., E.V.A., W.B.L. and K.A.J. contributed to data analysis. All authors contributed to manuscript preparation and reviewed and approved the final manuscript.

Corresponding author

Correspondence to Alanna J. Church.

Ethics declarations

Competing interests

A.J.C. sits on an advisory board for Bayer. L.B.C. is an employee at Sema4 and is a consultant for X-Chem and Biomatics. L.M. is a consultant for Jazz Pharmaceuticals. T.W.L. is an advisory board member for Bayer and has received honoraria from Bayer, Cellectis, Novartis, Deciphera, Jumo Health and Y-mAbs and has received research funding from Pfizer and Bayer. M.A.A. is an advisory board member for Fennec Pharmaceuticals. Y-C.L. is an advisory board member for Takeda Pharmaceutical Company. A.D.C. has received research support from Bayer and his spouse is employed by Labcorp. J.K.’s spouse has received consulting fees from ROME Therapeutics, Foundation Medicine, NanoString, Merck Millipore and Pfizer that are not related to this work. J.K.’s spouse is a founder and has equity in ROME Therapeutics, PanTher Therapeutics and TellBio, which is not related to this work. J.K.’s spouse receives research support from ACD Bio-Techne, PureTech Health and Ribon Therapeutics, which was not used in this work. S.L.V. is a consultant for CVS Accordant. E.V.A. has provided advisory/consulting work for Tango Therapeutics, Genome Medical, Invitae, Enara Bio, Janssen, Manifold Bio and Monte Rosa Therapeutics, has received research support from Novartis and Bristol Myers Squibb, holds equity in Tango Therapeutics, Genome Medical, Syapse, Enara Bio, Manifold Bio, Microsoft and Monte Rosa Therapeutics, has received travel reimbursement from Roche/Genentech and holds institutional patents filed on chromatin mutations and immunotherapy response and methods for clinical interpretation. S.G.D. has consulted for Bayer and received travel expenses from Loxo Oncology, Roche and Salarius. W.B.L. has served on the data safety monitoring boards for Merck Millipore and Jubilant Draximage. K.A.J. has consulted for Ipsen and Bayer, and has received honoraria from Foundation Medicine and Takeda Pharmaceutical Company. The other authors declare no competing interests.

Peer review

Peer review information

Nature Medicine thanks Elaine Mardis and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling editor: Anna Maria Ranzoni, in collaboration with the Nature Medicine team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 CONSORT diagram.

CONSORT diagram of 389 patients enrolled in the GAIN/iCat2 study between 11/2015 and 12/2018 identifying the analytic cohort. Percentages are based on the analytic population (n = 345).

Extended Data Fig. 2 iCat Recommendations.

Number of genes with iCat recommendations per patient at the time of initial report (February, 2016 to June, 2019) and with updated evidence reviewed between May and June, 2020 (a). Highest tier of iCat therapeutic recommendation for each patient at the time of initial report (February, 2016 to June, 2019) and with updated evidence reviewed between May and June, 2020 (b).

Extended Data Fig. 3 Changes in iCat Recommendations.

Changes in tiering of individual iCat recommendations from the time of initial report (February, 2016 to June, 2019) to re-tiering with updated evidence reviewed between May and June, 2020 upon reevaluation of evidence for expected response to matched targeted therapy.

Extended Data Fig. 4 Genes with iCat Recommendations.

iCat tiers reported according to alteration type (a). Top genes with strong evidence for therapeutic impact (iCat tiers 1-2) at the time of report (February 2016 to June 2019) (b) and with updated evidence reviewed between May and June, 2020 (c).

Extended Data Fig. 5 Responders to Matched Targeted Therapy.

Details of 5 responders to matched targeted therapy. GAIN patient 317 with an ALK fusion in a medullary thyroid carcinoma also responded, with images available in the primary report (Hillier et al., 2019).

Extended Data Fig. 6 Oncoprint for Patients Receiving MTT.

Tumor mutational burden (TMB) and additional alterations in 29 patients who received MTT. Additional alterations are shown if they occurred in >1 case or were potentially targetable. (For the case with high TMB all potentially actionable variants are not shown).

Extended Data Fig. 7 Top Genes with Diagnostic and Prognostic Significance.

All genes or chromosomes arms with tier 1 or 2 AMP/CAP/ASCO guideline evidence for prognostic impact (top; yellow). Top alterations with tier 1 or 2 AMP/CAP/ASCO guideline evidence for diagnostic impact, representing 181 of 227 diagnostically significant alterations (bottom; blue).

Extended Data Fig. 8 Diagnostic Impact Case.

Details of an illustrative case (GAIN318) of diagnostic impact: MRI (sagittal short inversion time inversion recovery (STIR) sequence) of a distal tibia tumor at diagnosis (a) and one year later at recurrence (b). The diagnosis rendered in the pathology report at initial diagnosis was aneurysmal bone cyst (ABC) while biopsy of the recurrence demonstrated osteogenic cells with pleomorphism and atypical mitotic figures (c, H&E stain, single experiment, not repeated). p53 IHC (d) shows loss of p53 expression in tumor but not normal cells (single experiment, not repeated). GAIN sequencing identified a novel TP53::USP6 fusion connecting TP53 intron 1 to USP6 intron 7, supporting a diagnosis of osteosarcoma in which TP53 rearrangements are common (e, created with Biorender.com). RNA analysis shows high expression of USP6, as measured by the number of unique RNA reads across 4 USP6 target regions [chr17:5031701, chr17:5033235, chr17:5033666, chr17:5033937], shown in the context of 12 cases with USP6 fusions (left, average read count 1552 reads) compared to 20 control cases with no USP6 fusions (right, average read count 8.0). This represents a significant difference in expression (unpaired two-tail t-test, p = 6.3e-10). Box plots represent maximum and minimum values (whiskers), first and third quartiles (bounds of box) and median (center line) (f).

Extended Data Fig. 9 Overview of the iCat2/GAIN Study.

Overview of the iCat2/GAIN study (a, created with Biorender.com). Targeted DNA NGS is performed on one or more tumor samples from each patient. Selected patients also have tumors subjected to RNA sequencing. Test results are returned to the treating oncologist and follow-up treatment and response data are collected. Details of clinical interpretation of test reports including molecular tumor board are shown in extended data Fig. 3. Testing strategy (b) to select patients for additional sequencing with either whole transcriptome sequencing or targeted RNA fusion panel testing (RNASeq). RNASeq was not performed if it was unlikely to contribute to research findings or clinical care. In this study, transcriptome sequencing was analyzed only for structural variants (SVs) and OncoPanel detects rearrangements in 60 genes (c). The testing triage is based on several assumptions: 1) False positives for SV detection OncoPanel are uncommon; 2) If oncogenic fusions have not been described in a particular solid tumor in previous studies and typical oncogenic events for that diagnosis are present then novel oncogenic fusions are unlikely; and 3) very rare pediatric solid malignancies might harbor previously undescribed fusions because they are understudied.

Extended Data Fig. 10 Details of Clinical Interpretation.

Details of clinical interpretation of test reports. A knowledgebase and report generation tool, iCatalog, was developed specifically for this study. iCatalog contains pediatric cancer specific knowledge on the gene and variant level including associated references (stored with PMID) and clinical trials (stored by NCT number). iCatalog knowledge is maintained by a staff scientist and research coordinator both at several scheduled times and when interpreting cases. iCatalog uses API to annotate variants. Resources available to the iCatalog user are shown below. Cases with Tier 5 iCat recommendations, previously undiscussed evidence or conflicting evidence are discussed at the molecular tumor board. Clinical interpretation reports are returned to the lead site investigator or enrolling oncologist.

Supplementary information

Supplementary Information

Study Protocol

Reporting Summary

Supplementary Data

Description of each tab of the supplementary data file: genes targeted for exonic sequence variants by the DNA sequencing panel; genes targeted for rearrangements by the DNA sequencing panel; gene list for the LaMPP solid and brain tumor fusion panel; table of single nucleotide variants (SNVs) identified by OncoPanel sequencing; table of copy number variants (CNVs) identified by OncoPanel sequencing; table of structural variants (SVs) identified by OncoPanel sequencing; table of tumor mutational burden (TMB) and mismatch repair (MMR) status identified by OncoPanel sequencing; table of diagnostic, prognostic and therapeutic alterations; list of iCat recommendations for therapeutically actionable alterations; detection of Genome4Kids solid tumor (ST) patient actionable variants by GAIN sequencing; Genomes4Kids solid tumor (ST) patient actionable variants annotated with detected or not detected by GAIN sequencing; Genomes4Kids solid tumor (ST) patient actionable variants annotated not detected by GAIN sequencing.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Church, A.J., Corson, L.B., Kao, PC. et al. Molecular profiling identifies targeted therapy opportunities in pediatric solid cancer. Nat Med 28, 1581–1589 (2022). https://doi.org/10.1038/s41591-022-01856-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-022-01856-6

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer