Frequency and Prognostic Impact of ALK Amplifications and Mutations in the European Neuroblastoma Study Group (SIOPEN) High-Risk Neuroblastoma Trial (HR-NBL1)

Angela Bellini, Ulrike Pötschger, Virginie Bernard, Eve Lapouble, Sylvain Baulande, Peter F Ambros, Nathalie Auger, Klaus Beiske, Marie Bernkopf, David R Betts, Jaydutt Bhalshankar, Nick Bown, Katleen de Preter, Nathalie Clément, Valérie Combaret, Jaime Font de Mora, Sally L George, Irene Jiménez, Marta Jeison, Barbara Marques, Tommy Martinsson, Katia Mazzocco, Martina Morini, Annick Mühlethaler-Mottet, Rosa Noguera, Gaelle Pierron, Maria Rossing, Sabine Taschner-Mandl, Nadine Van Roy, Ales Vicha, Louis Chesler, Walentyna Balwierz, Victoria Castel, Martin Elliott, Per Kogner, Geneviève Laureys, Roberto Luksch, Josef Malis, Maja Popovic-Beck, Shifra Ash, Olivier Delattre, Dominique Valteau-Couanet, Deborah A Tweddle, Ruth Ladenstein, Gudrun Schleiermacher, Angela Bellini, Ulrike Pötschger, Virginie Bernard, Eve Lapouble, Sylvain Baulande, Peter F Ambros, Nathalie Auger, Klaus Beiske, Marie Bernkopf, David R Betts, Jaydutt Bhalshankar, Nick Bown, Katleen de Preter, Nathalie Clément, Valérie Combaret, Jaime Font de Mora, Sally L George, Irene Jiménez, Marta Jeison, Barbara Marques, Tommy Martinsson, Katia Mazzocco, Martina Morini, Annick Mühlethaler-Mottet, Rosa Noguera, Gaelle Pierron, Maria Rossing, Sabine Taschner-Mandl, Nadine Van Roy, Ales Vicha, Louis Chesler, Walentyna Balwierz, Victoria Castel, Martin Elliott, Per Kogner, Geneviève Laureys, Roberto Luksch, Josef Malis, Maja Popovic-Beck, Shifra Ash, Olivier Delattre, Dominique Valteau-Couanet, Deborah A Tweddle, Ruth Ladenstein, Gudrun Schleiermacher

Abstract

Purpose: In neuroblastoma (NB), the ALK receptor tyrosine kinase can be constitutively activated through activating point mutations or genomic amplification. We studied ALK genetic alterations in high-risk (HR) patients on the HR-NBL1/SIOPEN trial to determine their frequency, correlation with clinical parameters, and prognostic impact.

Materials and methods: Diagnostic tumor samples were available from 1,092 HR-NBL1/SIOPEN patients to determine ALK amplification status (n = 330), ALK mutational profile (n = 191), or both (n = 571).

Results: Genomic ALK amplification (ALKa) was detected in 4.5% of cases (41 out of 901), all except one with MYCN amplification (MNA). ALKa was associated with a significantly poorer overall survival (OS) (5-year OS: ALKa [n = 41] 28% [95% CI, 15 to 42]; no-ALKa [n = 860] 51% [95% CI, 47 to 54], [P < .001]), particularly in cases with metastatic disease. ALK mutations (ALKm) were detected at a clonal level (> 20% mutated allele fraction) in 10% of cases (76 out of 762) and at a subclonal level (mutated allele fraction 0.1%-20%) in 3.9% of patients (30 out of 762), with a strong correlation between the presence of ALKm and MNA (P < .001). Among 571 cases with known ALKa and ALKm status, a statistically significant difference in OS was observed between cases with ALKa or clonal ALKm versus subclonal ALKm or no ALK alterations (5-year OS: ALKa [n = 19], 26% [95% CI, 10 to 47], clonal ALKm [n = 65] 33% [95% CI, 21 to 44], subclonal ALKm (n = 22) 48% [95% CI, 26 to 67], and no alteration [n = 465], 51% [95% CI, 46 to 55], respectively; P = .001). Importantly, in a multivariate model, involvement of more than one metastatic compartment (hazard ratio [HR], 2.87; P < .001), ALKa (HR, 2.38; P = .004), and clonal ALKm (HR, 1.77; P = .001) were independent predictors of poor outcome.

Conclusion: Genetic alterations of ALK (clonal mutations and amplifications) in HR-NB are independent predictors of poorer survival. These data provide a rationale for integration of ALK inhibitors in upfront treatment of HR-NB with ALK alterations.

Trial registration: ClinicalTrials.gov NCT01704716.

Conflict of interest statement

Walentyna BalwierzHonoraria: Shire, Gilead Sciences, Novartis, AmgenConsulting or Advisory Role: Amgen, Novartis, Roche, TakedaTravel, Accommodations, Expenses: Jazz Pharmaceuticals, Shire, Roche, Servier Martin ElliottConsulting or Advisory Role: Bayer Dominique Valteau-CouanetConsulting or Advisory Role: EUSA PharmaResearch Funding: Orphelia PharmaPatents, Royalties, Other Intellectual Property: Royalties from Apeiron to SIOPENTravel, Accommodations, Expenses: EUSA Pharma, Jazz Pharmaceuticals Deborah A. TweddleHonoraria: Eusa PharmaTravel, Accommodations, Expenses: EUSA Pharma Ruth LadensteinHonoraria: Apeiron Biologics, Boehringer Ingelheim, EUSA PharmaConsulting or Advisory Role: Apeiron Biologics, Boehringer Ingelheim, EUSA PharmaResearch Funding: Apeiron Biologics, EUSA PharmaPatents, Royalties, Other Intellectual Property: Apeiron Biologics, EUSA PharmaExpert Testimony: Apeiron Biologics, EUSA PharmaTravel, Accommodations, Expenses: Apeiron Biologics, EUSA Pharma Gudrun SchleiermacherHonoraria: BMSResearch Funding: Bristol Myers Squibb, Pfizer, MSDavenir, RocheTravel, Accommodations, Expenses: RocheNo other potential conflicts of interest were reported.

Figures

FIG 1.
FIG 1.
Flow diagram of patient inclusion. A total of 3,334 patients with HR-NB were enrolled in the HR-NBL1 trial from 188 centers. Among these, 2,350 patients were not included in this study, either because no contributive tumor material was available, or because these was no FU data, or both. Thus, 1,092 patients from 132 centers were included in this study. aClonal level: > 20% MAF. bSubclonal level: MAF 0.1%-20%. FU, follow-up; HR-NB, high-risk neuroblastoma; MAF, mutated allele fraction.
FIG 2.
FIG 2.
Genetic alterations of ALK in patients with HR-NB. (A) Copy-number profile of case 536. Genomic coamplification of MYCN and ALK is observed on chromosome 2, encompassing the regions between position 15,440,477 and 16,822,999 and between 29,113,790 and 30,309,749 bp (human genome assembly hg19; UCSC Genome Browser Home). (B) Frequency distribution of mutated ALK alleles at the studied chromosome regions, encompassing the AA positions F1174, L1190, L1196, R1245, D1270, G1272, M1273, A1274, R1275, and Y1278 detected, in 762 samples. ALK mutations involved the common mutational hotspots (F1174, F1245, and R1275) in 12.5% (97 out of 772) of cases, at a clonal level (MAF 20%-93%) in 72 cases, and at a subclonal level (MAF < 20%) in 25 cases. At the F1174 hotspot (chr2: 29,443,695-29,443,697), alterations were observed in 44 cases: 42 cases harbored a mutation leading to the AA change F1174L, one case with F1174I, and one case with F1174S, with MAFs ranging from 0.12% to 78%. At the R1275 hotspot (chr2: 29,432,849-29,430,139), mutations were detected in 43 cases: 38 cases harbored a mutation leading to the AA change R1275Q and five cases with R1275L, with the MAFs ranging from 0.2% to 93%. Ten cases showed ALK mutations at the F1245 hotspot (chr2: 29,436,858-29,436,860) within exon 24. Three samples showed the F1245L mutation, three cases carried the F1245C mutation, three showed the F1245I mutation, and one showed mutation F1245V mutation (Fig 1 and Appendix Table A1). Other ALK mutations were detected at residues I1170, L1190 (two cases), L1196, D1270, G1272, M1273, A1274, and Y1278 within the explored regions, leading to a nonsynonymous AA change with a predicted functional impact. All these mutations were clonal (MAF > 20%) except for M1273I (MAF 0.2%) and I1170 (MAF 2.8%). AA, amino acid; aCGH, array comparative genomic hybridization; bp, base pair; HR-NB, high-risk neuroblastoma; MAF, mutated allele fraction; UCSC, University of California, Santa Cruz.
FIG 3.
FIG 3.
Survival in the ALK analysis cohort. (A) OS according to ALK amplification status in 901 patients: presence of ALK amplification (n = 41), 5-year OS 28% (95% CI, 15 to 42) versus absence of ALK amplification (n = 860), 5-year OS 51% (95% CI, 47 to 54); P < .0001. (B) OS according to ALK mutation status in 762 patients: presence of an ALK mutation (n = 106), 5-year OS 41% (95% CI, 31 to 51) versus absence of an ALK mutation (n = 656), 5-year OS 49% (95% CI, 45 to 53); P = NS. (C) OS according to ALK clonal or subclonal mutation status in 762 patients: no mutation (n = 656), 5-year OS 49% (95% CI, 45 to 53); clonal mutations (n = 76), 5-year OS 34% (95% CI, 23 to 45); and subclonal mutations (n = 30), 5-year OS 59% (95% CI, 39 to 74), respectively; P = .018. (D) OS according to the presence of any ALK alterations in 611 patients with known ALK amplification and ALK mutation status: presence of an ALK alteration (n = 146), 5-year OS 37% (95% CI, 29 to 45); versus absence of ALK alterations (n = 465), 5-year OS 51% (95% CI, 46 to 55); P = .005. (E) OS according to the type of ALK alteration in the cohort of 571 patients with known ALK amplification and ALK mutation status: no alteration (n = 465), 5-year OS 51% (95% CI, 46 to 55); clonal mutations (n = 65), 5-year OS 33% (95% CI, 21 to 44); subclonal mutations (n = 12), 5-year OS 48% (95% CI, 26 to 67); and ALK amplification (n = 19), 5-year OS 26% (95% CI, 10 to 47), respectively; P = .001. (F) OS according to ALK alterations (ALK amplification or clonal ALK mutation) in patients who received immunotherapy (n = 141): To evaluate the impact of ALK alterations (ALK amplification or clonal ALK mutation) in patients who received dinutuximab beta, OS was calculated from the start of dinutuximab beta treatment and evaluated using the same approaches as described in the Materials and Methods section. ALK alteration (ALK amplification or clonal ALK mutation, n = 29, 5-year OS 48% [95% CI, 28 to 65]) versus no ALK alteration (n = 112) 67% (95% CI, 56 to 75); P = .034. Patient details: Appendix Table A3. HR, hazard ratio; NS, not significant; OS, overall survival; ref, reference.
FIG A1.
FIG A1.
Treatment flowchart of the HR-NBL1 Protocol (ClinicalTrials.gov: NCT01704716, EudraCT: 2006-001489-17) over the whole period. aInfants and children with a body weight below 12 kg will be dosed at 0.67 mg/kg/d. In infants weighing ≤ 5 kg, a further 1/3 dose reduction is advised. AUC, area under the curve; BUMEL, busulfan and melphalan; CAV, cyclophosphamide plus doxorubicin or vincristine; CEM, carboplatin, etoposide, and melphalan; CH14.18/CHO, human-mouse chimeric monoclonal anti-disialoganglioside GD2 antibody ch14.18 produced in Chinese hamster ovary (CHO) cells; COJEC, chemotherapy schedule COJEC defined below; GFR, glomerular filtration rate; IL-2, interleukin-2; IV, intravenous; P or E, cisplatin or etoposide; R1, randomization 1; R2, randomization 2; R3, randomization 3; R4, randomization 4; RT, radiotherapy; SCR, stringent complete response; TP, time period; TVD, topotecan-vincristine-doxorubicin.
FIG A2.
FIG A2.
Comparison of patients in the ALK analysis cohort and patients not in the ALK analysis cohort. (A and B) EFS and OS of the ALK analysis cohort and patients not in the ALK cohort. (A) No statistically significant difference in EFS and (B) OS was observed between patients included in the ALK analysis cohort (n = 1,092, from 132 centers; red line), patients not included in this study from the same centers (n = 1,665, blue line) and patients not included in this study from centers not participating in this study (n = 577, green line) (5-year EFS: 40% [95% CI, 37 to 43] v 37% [95% CI, 35 to 40] v 33% [95% CI, 29 to 37]; 5-year OS: 49% [95% CI, 46 to 53] v 48% [95% CI, 46 to 51] v 44% [95% CI, 40 to 59]; P = NS). (C) Recruitment, by year (x-axis), in the ALK analysis cohort (% of patients: y-axis; absolute numbers: in the blue bars). The % and number of patients not included in the ALK analysis cohort from centers participating, and from nonparticipating centers, are indicated in orange and gray, respectively. EFS, event-free survival; NS, not significant; OS, overall survival.
FIG A3.
FIG A3.
Double event of ALK amplification and ALK mutation detected in one case (case 15). The SNP array shows an amplified region in chromosome 2 encompassing the ALK gene. Sanger sequencing profile shows R1275Q mutation (MAF = 93.3%) in the same case. HD, high definition; MAF, mutated allele fraction; SNP, single-nucleotide polymorphism.
FIG A4.
FIG A4.
MAF of subclonal ALK mutations detected by TDS and confirmed by a second independent TDS experiment. Red spots representing the MAF for each ALK mutation are plotted on the x-axis (first TDS experiment) and y-axis (second TDS experiment), with a strong correlation between the two independent experiments (r2 = 0.9924, P < .0001). Blue spots represent subclonal ALK mutations with a very low MAF (< 0.1%) not confirmed in an independent experiment and not retained in the analysis (n = 6). MAF, mutated allele fraction; TDS, targeted deep sequencing.
FIG A5.
FIG A5.
Survival in the ALK analysis cohort (n = 1,092 patients) according to known prognostic factors. (A) EFS and OS in the ALK analysis cohort population (n = 1,092 patients). Five-year EFS (blue line) 40% (95% CI, 37 to 43); 5-year OS (red line) 49% (95% CI, 46 to 53). (B) OS according to age. Five-year OS in patients < 1 year of age at diagnosis (red line) 50% (95% CI, 37 to 61); in patients 1-1.5 years of age at diagnosis (blue line) 58% (95% CI, 49 to 66); in patients 1.5-5 years of age at diagnosis (green line) 50% (95% CI, 46 to 53); and in patients > 5 years of age at diagnosis (purple line) 43% (95% CI, 35 to 50); P = NS (pseudo-value regression). (C) OS according to number of involved MCs. Five-year OS in patients with localized disease (red line) 67% (95% CI, 58 to 75), in patients with involvement of one MC (blue line) 65% (95% CI, 55 to 73), two MCs (green line) 52% (95% CI, 46 to 58), or over two MCs (purple line) 41% (95% CI, 36 to 46); P < .001. (D) OS according to stage. Five-year OS in patients with localized disease (red line) 67% (95% CI, 58 to 75), in patients with stage 4 disease (blue line) 47% (95% CI, 44 to 50), or stage 4s disease (green line) 54% (95% CI, 25 to 76); P < .001. (E) OS according to MYCN amplification in stage 4 disease. Five-year OS in patients with MNA (blue line) 46% (95% CI, 41 to 51), in patients without MNA (red line) 48% (95% CI, 44 to 53), NS (pseudo-value regression). (F) OS according to treatment period, before (< March 2010) or after (> March 2010) the definition of HDC by BUMEL and immunotherapy maintenance as standard treatment. A significant improvement survival because of BUMEL and GD2 standard therapy is observed. Five-year OS in patients having been treated before March 2010 (red line) 46% (95% CI, 41 to 51) versus after March 2010 (blue line) 51% (95% CI, 47 to 56); P = .039.- BUMEL, busulfan and melphalan; cHR, crude hazard ratio; EFS, event-free survival; HDC, high-dose chemotherapy; HR, hazard ratio; MC, metastatic compartment; MNA, MYCN-amplified; NS, not significant; OS, overall survival; ref, reference.

References

    1. Matthay KK, Maris JM, Schleiermacher G, et al. : Neuroblastoma. Nat Rev Dis Primers 2:16078, 2016
    1. Holmes K, Potschger U, Pearson ADJ, et al. : Influence of surgical excision on the survival of patients with stage 4 high-risk neuroblastoma: A report from the HR-NBL1/SIOPEN study. J Clin Oncol 38:2902-2915, 2020
    1. Ladenstein R, Potschger U, Pearson ADJ, et al. : Busulfan and melphalan versus carboplatin, etoposide, and melphalan as high-dose chemotherapy for high-risk neuroblastoma (HR-NBL1/SIOPEN): An international, randomised, multi-arm, open-label, phase 3 trial. Lancet Oncol 18:500-514, 2017
    1. Ladenstein R, Potschger U, Valteau-Couanet D, et al. : Investigation of the role of dinutuximab beta-based immunotherapy in the SIOPEN high-risk neuroblastoma 1 trial (HR-NBL1). Cancers (Basel) 12:309, 2020
    1. Ladenstein R, Potschger U, Valteau-Couanet D, et al. : Interleukin 2 with anti-GD2 antibody ch14.18/CHO (dinutuximab beta) in patients with high-risk neuroblastoma (HR-NBL1/SIOPEN): A multicentre, randomised, phase 3 trial. Lancet Oncol 19:1617-1629, 2018
    1. Ozkaynak MF, Gilman AL, London WB, et al. : A comprehensive safety trial of chimeric antibody 14.18 with GM-CSF, IL-2, and isotretinoin in high-risk neuroblastoma patients following myeloablative therapy: Children's Oncology Group study ANBL0931. Front Immunol 9:1355, 2018
    1. Park JR, Kreissman SG, London WB, et al. : Effect of tandem autologous stem cell transplant vs single transplant on event-free survival in patients with high-risk neuroblastoma: A randomized clinical trial. JAMA 322:746-755, 2019
    1. Pinto N, Naranjo A, Hibbitts E, et al. : Predictors of differential response to induction therapy in high-risk neuroblastoma: A report from the Children's Oncology Group (COG). Eur J Cancer 112:66-79, 2019
    1. Seeger RC, Brodeur GM, Sather H, et al. : Association of multiple copies of the N-myc oncogene with rapid progression of neuroblastomas. N Engl J Med 313:1111-1116, 1985
    1. Janoueix-Lerosey I, Schleiermacher G, Michels E, et al. : Overall genomic pattern is a predictor of outcome in neuroblastoma. J Clin Oncol 27:1026-1033, 2009
    1. Peifer M, Hertwig F, Roels F, et al. : Telomerase activation by genomic rearrangements in high-risk neuroblastoma. Nature 526:700-704, 2015
    1. Molenaar JJ, Koster J, Zwijnenburg DA, et al. : Sequencing of neuroblastoma identifies chromothripsis and defects in neuritogenesis genes. Nature 483:589-593, 2012
    1. Pugh TJ, Morozova O, Attiyeh EF, et al. : The genetic landscape of high-risk neuroblastoma. Nat Genet 45:279-284, 2013
    1. Sausen M, Leary RJ, Jones S, et al. : Integrated genomic analyses identify ARID1A and ARID1B alterations in the childhood cancer neuroblastoma. Nat Genet 45:12-17, 2012
    1. Chen Y, Takita J, Choi YL, et al. : Oncogenic mutations of ALK kinase in neuroblastoma. Nature 455:971-974, 2008
    1. George RE, Sanda T, Hanna M, et al. : Activating mutations in ALK provide a therapeutic target in neuroblastoma. Nature 455:975-978, 2008
    1. Janoueix-Lerosey I, Lequin D, Brugieres L, et al. : Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma. Nature 455:967-970, 2008
    1. Mosse YP, Laudenslager M, Longo L, et al. : Identification of ALK as a major familial neuroblastoma predisposition gene. Nature 455:930-935, 2008
    1. Bellini A, Bessoltane-Bentahar N, Bhalshankar J, et al. : Study of chromatin remodeling genes implicates SMARCA4 as a putative player in oncogenesis in neuroblastoma. Int J Cancer 145:2781-2791, 2019
    1. Javanmardi N, Fransson S, Djos A, et al. : Low frequency ALK hotspots mutations in neuroblastoma tumours detected by ultra-deep sequencing: Implications for ALK inhibitor treatment. Sci Rep 9:2199, 2019
    1. Combaret V, Iacono I, Bellini A, et al. : Detection of tumor ALK status in neuroblastoma patients using peripheral blood. Cancer Med 4:540-550, 2015
    1. Bellini A, Bernard V, Leroy Q, et al. : Deep sequencing reveals occurrence of subclonal ALK mutations in neuroblastoma at diagnosis. Clin Cancer Res 21:4913-4921, 2015
    1. Eleveld TF, Oldridge DA, Bernard V, et al. : Relapsed neuroblastomas show frequent RAS-MAPK pathway mutations. Nat Genet 47:864-871, 2015
    1. Bresler SC, Weiser DA, Huwe PJ, et al. : ALK mutations confer differential oncogenic activation and sensitivity to ALK inhibition therapy in neuroblastoma. Cancer Cell 26:682-694, 2014
    1. Fransson S, Hansson M, Ruuth K, et al. : Intragenic anaplastic lymphoma kinase (ALK) rearrangements: Translocations as a novel mechanism of ALK activation in neuroblastoma tumors. Genes Chromosomes Cancer 54:99-109, 2015
    1. De Brouwer S, De Preter K, Kumps C, et al. : Meta-analysis of neuroblastomas reveals a skewed ALK mutation spectrum in tumors with MYCN amplification. Clin Cancer Res 16:4353-4362, 2010
    1. Friboulet L, Li N, Katayama R, et al. : The ALK inhibitor ceritinib overcomes crizotinib resistance in non-small cell lung cancer. Cancer Discov 4:662-673, 2014
    1. Guan J, Tucker ER, Wan H, et al. : The ALK inhibitor PF-06463922 is effective as a single agent in neuroblastoma driven by expression of ALK and MYCN. Dis Model Mech 9:941-952, 2016
    1. Solomon BJ, Besse B, Bauer TM, et al. : Lorlatinib in patients with ALK-positive non-small-cell lung cancer: Results from a global phase 2 study. Lancet Oncol 19:1654-1667, 2018
    1. Ladenstein R, Potschger U, Siabalis D, et al. : Dose finding study for the use of subcutaneous recombinant interleukin-2 to augment natural killer cell numbers in an outpatient setting for stage 4 neuroblastoma after megatherapy and autologous stem-cell reinfusion. J Clin Oncol 29:441-448, 2010
    1. Ladenstein R, Valteau-Couanet D, Brock P, et al. : Randomized trial of prophylactic granulocyte colony-stimulating factor during rapid COJEC induction in pediatric patients with high-risk neuroblastoma: The European HR-NBL1/SIOPEN study. J Clin Oncol 28:3516-3524, 2010
    1. Garaventa A, Poetschger U, Valteau-Couanet D, et al. : Randomized trial of two induction therapy regimens for high-risk neuroblastoma: HR-NBL1.5 International Society of Pediatric Oncology European Neuroblastoma Group study. J Clin Oncol 39:2552-2563, 2021
    1. Ambros PF, Ambros IM, Brodeur GM, et al. : International consensus for neuroblastoma molecular diagnostics: Report from the International Neuroblastoma Risk Group (INRG) Biology Committee. Br J Cancer 100:1471-1482, 2009
    1. Ambros IM, Brunner B, Aigner G, et al. : A multilocus technique for risk evaluation of patients with neuroblastoma. Clin Cancer Res 17:792-804, 2011
    1. Schleiermacher G, Michon J, Ribeiro A, et al. : Segmental chromosomal alterations lead to a higher risk of relapse in infants with MYCN-non-amplified localised unresectable/disseminated neuroblastoma (a SIOPEN collaborative study). Br J Cancer 105:1940-1948, 2011
    1. Schleiermacher G, Mosseri V, London WB, et al. : Segmental chromosomal alterations have prognostic impact in neuroblastoma: A report from the INRG project. Br J Cancer 107:1418-1422, 2012
    1. Ambros IM, Brunner C, Abbasi R, et al. : Ultra-high density SNParray in neuroblastoma molecular diagnostics. Front Oncol 4:202, 2014
    1. UCSC Genome Browser Home :
    1. Kaplan E, Meier P: Nonparametric estimation from incomplete observations. J Am Stat Assoc 53:457-481, 1958
    1. Peto R, Pike MC, Armitage P, et al. : Design and analysis of randomized clinical trials requiring prolonged observation of each patient. II. Analysis and examples. Br J Cancer 35:1-39, 1977
    1. Andersen PK, Perme MP: Pseudo-observations in survival analysis. Stat Methods Med Res 19:71-99, 2011
    1. Morgenstern DA, Potschger U, Moreno L, et al. : Risk stratification of high-risk metastatic neuroblastoma: A report from the HR-NBL-1/SIOPEN study. Pediatr Blood Cancer 65:e27363, 2018
    1. Depuydt P, Boeva V, Hocking TD, et al. : Genomic amplifications and distal 6q loss: Novel markers for poor survival in high-risk neuroblastoma patients. J Natl Cancer Inst 110:1084-1093, 2018
    1. Mosse YP, Lim MS, Voss SD, et al. : Safety and activity of crizotinib for paediatric patients with refractory solid tumours or anaplastic large-cell lymphoma: A Children's Oncology Group phase 1 consortium study. Lancet Oncol 14:472-480, 2013
    1. Guan J, Fransson S, Siaw JT, et al. : Clinical response of the novel activating ALK-I1171T mutation in neuroblastoma to the ALK inhibitor ceritinib. Cold Spring Harb Mol Case Stud 4:a002550, 2018
    1. Krytska K, Ryles HT, Sano R, et al. : Crizotinib synergizes with chemotherapy in preclinical models of neuroblastoma. Clin Cancer Res 22:948-960, 2016
    1. Bogen D, Brunner C, Walder D, et al. : The genetic tumor background is an important determinant for heterogeneous MYCN-amplified neuroblastoma. Int J Cancer 139:153-163, 2016
    1. Marrano P, Irwin MS, Thorner PS: Heterogeneity of MYCN amplification in neuroblastoma at diagnosis, treatment, relapse, and metastasis. Genes Chromosomes Cancer 56:28-41, 2017
    1. Berbegall AP, Bogen D, Potschger U, et al. : Heterogeneous MYCN amplification in neuroblastoma: A SIOP Europe Neuroblastoma study. Br J Cancer 118:1502-1512, 2018
    1. Turajlic S, Sottoriva A, Graham T, et al. : Resolving genetic heterogeneity in cancer. Nat Rev Genet 20:404-416, 2019
    1. Williams JB, Li S, Higgs EF, et al. : Tumor heterogeneity and clonal cooperation influence the immune selection of IFN-gamma-signaling mutant cancer cells. Nat Commun 11:602, 2020
    1. Padovan-Merhar OM, Raman P, Ostrovnaya I, et al. : Enrichment of targetable mutations in the relapsed neuroblastoma genome. PLoS Genet 12:e1006501, 2017
    1. Schleiermacher G, Javanmardi N, Bernard V, et al. : Emergence of new ALK mutations at relapse of neuroblastoma. J Clin Oncol 32:2727-2734, 2014
    1. Chicard M, Colmet-Daage L, Clement N, et al. : Whole-exome sequencing of cell-free DNA reveals temporo-spatial heterogeneity and identifies treatment-resistant clones in neuroblastoma. Clin Cancer Res 24:939-949, 2018
    1. Ackermann S, Cartolano M, Hero B, et al. : A mechanistic classification of clinical phenotypes in neuroblastoma. Science 362:1165-1170, 2018
    1. Koneru B, Lopez G, Farooqi A, et al. : Telomere maintenance mechanisms define clinical outcome in high-risk neuroblastoma. Cancer Res 80:2663-2675, 2020

Source: PubMed

3
Předplatit