Whole-transcriptome analysis in acute lymphoblastic leukemia: a report from the DFCI ALL Consortium Protocol 16-001

Thai Hoa Tran, Sylvie Langlois, Caroline Meloche, Maxime Caron, Pascal Saint-Onge, Alexandre Rouette, Alain R Bataille, Camille Jimenez-Cortes, Thomas Sontag, Henrique Bittencourt, Caroline Laverdière, Vincent-Philippe Lavallée, Jean-Marie Leclerc, Peter D Cole, Lisa M Gennarini, Justine M Kahn, Kara M Kelly, Bruno Michon, Raoul Santiago, Kristen E Stevenson, Jennifer J G Welch, Kaitlin M Schroeder, Victoria Koch, Sonia Cellot, Lewis B Silverman, Daniel Sinnett, Thai Hoa Tran, Sylvie Langlois, Caroline Meloche, Maxime Caron, Pascal Saint-Onge, Alexandre Rouette, Alain R Bataille, Camille Jimenez-Cortes, Thomas Sontag, Henrique Bittencourt, Caroline Laverdière, Vincent-Philippe Lavallée, Jean-Marie Leclerc, Peter D Cole, Lisa M Gennarini, Justine M Kahn, Kara M Kelly, Bruno Michon, Raoul Santiago, Kristen E Stevenson, Jennifer J G Welch, Kaitlin M Schroeder, Victoria Koch, Sonia Cellot, Lewis B Silverman, Daniel Sinnett

Abstract

The molecular hallmark of childhood acute lymphoblastic leukemia (ALL) is characterized by recurrent, prognostic genetic alterations, many of which are cryptic by conventional cytogenetics. RNA sequencing (RNA-seq) is a powerful next-generation sequencing technology that can simultaneously identify cryptic gene rearrangements, sequence mutations and gene expression profiles in a single assay. We examined the feasibility and utility of incorporating RNA-seq into a prospective multicenter phase 3 clinical trial for children with newly diagnosed ALL. The Dana-Farber Cancer Institute ALL Consortium Protocol 16-001 enrolled 173 patients with ALL who consented to optional studies and had samples available for RNA-seq. RNA-seq identified at least 1 alteration in 157 patients (91%). Fusion detection was 100% concordant with results obtained from conventional cytogenetic analyses. An additional 56 gene fusions were identified by RNA-seq, many of which confer prognostic or therapeutic significance. Gene expression profiling enabled further molecular classification into the following B-cell ALL (B-ALL) subgroups: high hyperdiploid (n = 36), ETV6-RUNX1/-like (n = 31), TCF3-PBX1 (n = 7), KMT2A-rearranged (KMT2A-R; n = 5), intrachromosomal amplification of chromosome 21 (iAMP21) (n = 1), hypodiploid (n = 1), Philadelphia chromosome (Ph)-positive/Ph-like (n = 16), DUX4-R (n = 11), PAX5 alterations (PAX5 alt; n = 11), PAX5 P80R (n = 1), ZNF384-R (n = 4), NUTM1-R (n = 1), MEF2D-R (n = 1), and others (n = 10). RNA-seq identified 141 nonsynonymous mutations in 93 patients (54%); the most frequent were RAS-MAPK pathway mutations. Among 79 patients with both low-density array and RNA-seq data for the Philadelphia chromosome-like gene signature prediction, results were concordant in 74 patients (94%). In conclusion, RNA-seq identified several clinically relevant genetic alterations not detected by conventional methods, which supports the integration of this technology into front-line pediatric ALL trials. This trial was registered at www.clinicaltrials.gov as #NCT03020030.

© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Comprehensive heatmap of molecular profiling findings by RNA-seq of 173 patients with ALL enrolled on DFCI 16-001. RNA-seq data from 173 patients with ALL are summarized in this heatmap. Patients were classified on the basis of their respective clustering analysis subtype, gene fusions, and somatic mutations. Specific gene fusions and mutational categories are indicated by colored squares and classified by associated gene signaling pathways. Mutation types included single nucleotide variants, indels, and intragenic IKZF1 deletion (IK6). iAMP21, intrachromosomal amplification of chromosome 21; NA, not applicable; TCR-R, T cell receptor rearrangement; VUS, variant of unknown significance.
Figure 2.
Figure 2.
Timeline of the clinical implementation of RNA-seq from sample receipt to report delivery and factors contributing to timeline variation at each step.
Figure 3.
Figure 3.
Novel ZBTB44-JAK2 fusion detected in a patient with Ph-like ALL. Exon 2 of ZBTB44 fused in-frame to exon 19 of JAK2, conserving an intact kinase domain. The fusion was validated by reverse transcriptase polymerase chain reaction and Sanger sequencing. The figure was adapted from Arriba output.
Figure 4.
Figure 4.
Detection of intragenic IKZF1 deletions by RNA-seq. Intragenic deletions of exons 4 to 7 of IKZF1 visualized using the GVIZ R package. Green line corresponds to normal IKZF1 transcript (IK1), and the red line corresponds to IK6 transcript missing exons 4 to 7 (example with patient #22).
Figure 5.
Figure 5.
Molecular subtype clustering of patients with ALL based on gene expression signatures. (A) Hierarchical clustering and (B) experimental t-distributed stochastic neighbor embedding (tSNE) performed using the top 500 variable genes from 1134 ALL samples from both in-house and public RNA-seq data sets. (C) The neural network probability score for each of the subtypes listed for panels A-C. Results for patient #124 are shown by the arrow in A and the red dot in B.
Figure 6.
Figure 6.
Estimation of blood cell populations using a deconvolution tool. Proportion of primary hematopoietic cell types in 173 patients with ALL using 13 primary cell-type expression profiles. CLP, common lymphoid progenitor; CMP, common myeloid progenitor; GMP, granulocyte-monocyte progenitor; HSC, hematopoietic stem cell; LMPP, lymphoid-primed multipotential progenitor; MEP, megakaryocyte-erythrocyte progenitor; MPP, multipotent progenitor; NK cell, natural killer cell.
Figure 7.
Figure 7.
Proposed tiered algorithm for a time- and cost-effective clinical implementation of RNA-seq. Proposed algorithm incorporating FISH and RNA-seq with an estimated turnaround time of 4 weeks from the time of sample receipt to final report delivery based on our experience. BA FISH, break-apart fluorescence in situ hybridization.

References

    1. Howlader N, Noone AM, Krapcho M, et al., eds. SEER Cancer Statistics Review, 1975-2010. National Cancer Institute Bethesda, MD, based on November 2012 SEER data submission, posted to the SEER web site, April 2013.
    1. Hunger SP, Mullighan CG. Acute lymphoblastic leukemia in children. N Engl J Med. 2015;373(16):1541-1552.
    1. Mullighan CG. Genomic characterization of childhood acute lymphoblastic leukemia. Semin Hematol. 2013;50(4):314-324.
    1. Mullighan CG. How advanced are we in targeting novel subtypes of ALL? Best Pract Res Clin Haematol. 2019;32(4):101095.
    1. Tran TH, Hunger SP. The genomic landscape of pediatric acute lymphoblastic leukemia and precision medicine opportunities [published online ahead of print 13 November 2020]. Semin Cancer Biol. doi: 10.1016/j.semcancer.2020.10.013.
    1. Khater F, Vairy S, Langlois S, et al. . Molecular profiling of hard-to-treat childhood and adolescent cancers. JAMA Netw Open. 2019;2(4):e192906.
    1. Wong M, Mayoh C, Lau LMS, et al. . Whole genome, transcriptome and methylome profiling enhances actionable target discovery in high-risk pediatric cancer. Nat Med. 2020;26(11):1742-1753.
    1. Inaba H, Azzato EM, Mullighan CG. Integration of next-generation sequencing to treat acute lymphoblastic leukemia with targetable lesions: The St. Jude Children’s Research Hospital approach. Front Pediatr. 2017;5:258.
    1. Chang F, Lin F, Cao K, et al. . Development and clinical validation of a large fusion gene panel for pediatric cancers. J Mol Diagn. 2019;21(5):873-883.
    1. Ching T, Duncan ME, Newman-Eerkes T, et al. . Analytical evaluation of the clonoSEQ Assay for establishing measurable (minimal) residual disease in acute lymphoblastic leukemia, chronic lymphocytic leukemia, and multiple myeloma. BMC Cancer. 2020;20(1):612.
    1. Dobin A, Davis CA, Schlesinger F, et al. . STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15-21.
    1. 1000 Genomes Project Consortium, Auton A, Brooks LD, et al. . A global reference for human genetic variation. Nature. 2015;526(7571):68-74.
    1. Tate JG, Bamford S, Jubb HC, et al. . COSMIC: the Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res. 2019;47(D1):D941-D947.
    1. Vaser R, Adusumalli S, Leng SN, Sikic M, Ng PC. SIFT missense predictions for genomes. Nat Protoc. 2016;11(1):1-9.
    1. Adzhubei IA, Schmidt S, Peshkin L, et al. . A method and server for predicting damaging missense mutations. Nat Methods. 2010;7(4):248-249.
    1. Nicorici D, Şatalan M, Edgren H, et al. . FusionCatcher – a tool for finding somatic fusion genes in paired-end RNA-sequencing data. bioRxiv 2014.
    1. Haas BJ, Dobin A, Stransky N, et al. . STAR-fusion: fast and accurate fusion transcript detection from RNA-Seq. bioRxiv 2017.
    1. Uhrig S, Ellermann J, Walther T, et al. . Accurate and efficient detection of gene fusions from RNA sequencing data. Genome Res. 2021;31(3):448-460.
    1. Corces MR, Buenrostro JD, Wu B, et al. . Lineage-specific and single-cell chromatin accessibility charts human hematopoiesis and leukemia evolution. Nat Genet. 2016;48(10):1193-1203.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550.
    1. Lilljebjörn H, Henningsson R, Hyrenius-Wittsten A, et al. . Identification of ETV6-RUNX1-like and DUX4-rearranged subtypes in paediatric B-cell precursor acute lymphoblastic leukaemia. Nat Commun. 2016;7(1):11790.
    1. National Cancer Institute, Office of Cancer Genomics. The Therapeutically Applicable Research to Generate Effective Treatments (TARGET) initiative. . Accessed 30 April 2021.
    1. Hahne F, Ivanek R. Visualizing genomic data using Gviz and Bioconductor. Methods Mol Biol. 2016;1418:335-351.
    1. Gong T, Szustakowski JD. DeconRNASeq: a statistical framework for deconvolution of heterogeneous tissue samples based on mRNA-Seq data. Bioinformatics. 2013;29(8):1083-1085.
    1. Iacobucci I, Mullighan CG. Genetic basis of acute lymphoblastic leukemia. J Clin Oncol. 2017;35(9):975-983.
    1. Gu Z, Churchman ML, Roberts KG, et al. . PAX5-driven subtypes of B-progenitor acute lymphoblastic leukemia. Nat Genet. 2019;51(2):296-307.
    1. Roberts KG, Li Y, Payne-Turner D, et al. . Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia. N Engl J Med. 2014;371(11):1005-1015.
    1. Stanulla M, Cavé H, Moorman AV. IKZF1 deletions in pediatric acute lymphoblastic leukemia: still a poor prognostic marker? Blood. 2020;135(4):252-260.
    1. Vairy S, Tran TH. IKZF1 alterations in acute lymphoblastic leukemia: the good, the bad and the ugly. Blood Rev. 2020;44:100677.
    1. Simonin M, Lhermitte L, Dourthe ME, et al. . IKZF1 alterations predict poor prognosis in adult and pediatric T-ALL. Blood. 2021;137(12):1690-1694.
    1. Mullighan CG, Su X, Zhang J, et al. ; Children’s Oncology Group . Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N Engl J Med. 2009;360(5):470-480.
    1. Kuiper RP, Waanders E, van der Velden VH, et al. . IKZF1 deletions predict relapse in uniformly treated pediatric precursor B-ALL. Leukemia. 2010;24(7):1258-1264.
    1. Khan M, Siddiqi R, Tran TH. Philadelphia chromosome-like acute lymphoblastic leukemia: a review of the genetic basis, clinical features, and therapeutic options. Semin Hematol. 2018;55(4):235-241.
    1. Reshmi SC, Harvey RC, Roberts KG, et al. . Targetable kinase gene fusions in high-risk B-ALL: a study from the Children’s Oncology Group. Blood. 2017;129(25):3352-3361.
    1. Liu Y, Easton J, Shao Y, et al. . The genomic landscape of pediatric and young adult T-lineage acute lymphoblastic leukemia. Nat Genet. 2017; 49(8):1211-1218.
    1. Yeoh AEJ, Lu Y, Chin WHN, et al. . Intensifying treatment of childhood B-lymphoblastic leukemia with IKZF1 deletion reduces relapse and improves overall survival: results of Malaysia-Singapore ALL 2010 Study. J Clin Oncol. 2018;36(26):2726-2735.
    1. Schieck M, Lentes J, Thomay K, et al. . Implementation of RNA sequencing and array CGH in the diagnostic workflow of the AIEOP-BFM ALL 2017 trial on acute lymphoblastic leukemia. Ann Hematol. 2020;99(4):809-818.
    1. Maeda T. Regulation of hematopoietic development by ZBTB transcription factors. Int J Hematol. 2016;104(3):310-323.
    1. Zhou X, Yang C, Zou Z, et al. . ZBTB44-FLT3 fusion in a patient with a myeloproliferative neoplasm. Br J Haematol. 2020;191(2):297-301.
    1. Peterson JF, Blackburn PR, Webley MR, et al. . Identification of a novel ZBTB20-JAK2 fusion by mate-pair sequencing in a young adult with B-lymphoblastic leukemia/lymphoma. Mayo Clin Proc. 2019;94(7):1381-1384.
    1. Tasian SK, Loh ML, Hunger SP. Philadelphia chromosome-like acute lymphoblastic leukemia. Blood. 2017;130(19):2064-2072.
    1. Gotti G, Puligandla M, Stevenson KE, et al. . Frequency and prognostic significance of recurrent gene mutations in pediatric B-ALL: Report from the DFCI ALL Consortium. Blood. 2020;136(suppl 1):15-16.
    1. Burns MA, Place AE, Stevenson KE, et al. . Identification of prognostic factors in childhood T-cell acute lymphoblastic leukemia: Results from DFCI ALL Consortium Protocols 05-001 and 11-001. Pediatr Blood Cancer. 2021;68(1):e28719.
    1. Paganin M, Grillo MF, Silvestri D, et al. . The presence of mutated and deleted PTEN is associated with an increased risk of relapse in childhood T cell acute lymphoblastic leukaemia treated with AIEOP-BFM ALL protocols. Br J Haematol. 2018;182(5):705-711.
    1. Petit A, Trinquand A, Chevret S, et al. ; French Acute Lymphoblastic Leukemia Study Group (FRALLE) . Oncogenetic mutations combined with MRD improve outcome prediction in pediatric T-cell acute lymphoblastic leukemia. Blood. 2018;131(3):289-300.
    1. Maude SL, Dolai S, Delgado-Martin C, et al. . Efficacy of JAK/STAT pathway inhibition in murine xenograft models of early T-cell precursor (ETP) acute lymphoblastic leukemia. Blood. 2015;125(11):1759-1767.
    1. Li Y, Buijs-Gladdines JG, Canté-Barrett K, et al. . IL-7 receptor mutations and steroid resistance in pediatric T cell acute lymphoblastic leukemia: a genome sequencing study. PLoS Med. 2016;13(12):e1002200.
    1. Lonetti A, Cappellini A, Spartà AM, et al. . PI3K pan-inhibition impairs more efficiently proliferation and survival of T-cell acute lymphoblastic leukemia cell lines when compared to isoform-selective PI3K inhibitors. Oncotarget. 2015;6(12):10399-10414.
    1. Lonetti A, Antunes IL, Chiarini F, et al. . Activity of the pan-class I phosphoinositide 3-kinase inhibitor NVP-BKM120 in T-cell acute lymphoblastic leukemia. Leukemia. 2014;28(6):1196-1206.
    1. Habets RA, de Bock CE, Serneels L, et al. . Safe targeting of T cell acute lymphoblastic leukemia by pathology-specific NOTCH inhibition. Sci Transl Med. 2019;11(494):eaau6246.
    1. Agnusdei V, Minuzzo S, Frasson C, et al. . Therapeutic antibody targeting of Notch1 in T-acute lymphoblastic leukemia xenografts. Leukemia. 2014;28(2):278-288.
    1. Brown LM, Lonsdale A, Zhu A, et al. . The application of RNA sequencing for the diagnosis and genomic classification of pediatric acute lymphoblastic leukemia. Blood Adv. 2020;4(5):930-942.
    1. Kluk MJ, Lindsley RC, Aster JC, et al. . Validation and implementation of a custom next-generation sequencing clinical assay for hematologic malignancies. J Mol Diagn. 2016;18(4):507-515.

Source: PubMed

Подписаться