Clinical Implications of Minimal Residual Disease Detection in Infants With KMT2A-Rearranged Acute Lymphoblastic Leukemia Treated on the Interfant-06 Protocol

Janine Stutterheim, Inge M van der Sluis, Paola de Lorenzo, Julia Alten, Philip Ancliffe, Andishe Attarbaschi, Benoit Brethon, Andrea Biondi, Myriam Campbell, Giovanni Cazzaniga, Gabriele Escherich, Alina Ferster, Rishi S Kotecha, Birgitte Lausen, Chi Kong Li, Luca Lo Nigro, Franco Locatelli, Rolf Marschalek, Claus Meyer, Martin Schrappe, Jan Stary, Ajay Vora, Jan Zuna, Vincent H J van der Velden, Tomasz Szczepanski, Maria Grazia Valsecchi, Rob Pieters, Janine Stutterheim, Inge M van der Sluis, Paola de Lorenzo, Julia Alten, Philip Ancliffe, Andishe Attarbaschi, Benoit Brethon, Andrea Biondi, Myriam Campbell, Giovanni Cazzaniga, Gabriele Escherich, Alina Ferster, Rishi S Kotecha, Birgitte Lausen, Chi Kong Li, Luca Lo Nigro, Franco Locatelli, Rolf Marschalek, Claus Meyer, Martin Schrappe, Jan Stary, Ajay Vora, Jan Zuna, Vincent H J van der Velden, Tomasz Szczepanski, Maria Grazia Valsecchi, Rob Pieters

Abstract

Purpose: Infant acute lymphoblastic leukemia (ALL) is characterized by a high incidence of KMT2A gene rearrangements and poor outcome. We evaluated the value of minimal residual disease (MRD) in infants with KMT2A-rearranged ALL treated within the Interfant-06 protocol, which compared lymphoid-style consolidation (protocol IB) versus myeloid-style consolidation (araC, daunorubicin, etoposide/mitoxantrone, araC, etoposide).

Materials and methods: MRD was measured in 249 infants by DNA-based polymerase chain reaction of rearranged KMT2A, immunoglobulin, and/or T-cell receptor genes, at the end of induction (EOI) and end of consolidation (EOC). MRD results were classified as negative, intermediate (< 5 × 10-4), and high (≥ 5 × 10-4).

Results: EOI MRD levels predicted outcome with 6-year disease-free survival (DFS) of 60.2% (95% CI, 43.2 to 73.6), 45.0% (95% CI, 28.3 to 53.1), and 33.8% (95% CI, 23.8 to 44.1) for infants with negative, intermediate, and high EOI MRD levels, respectively (P = .0039). EOC MRD levels were also predictive of outcome, with 6-year DFS of 68.2% (95% CI, 55.2 to 78.1), 40.1% (95% CI, 28.1 to 51.9), and 11.9% (95% CI, 2.6 to 29.1) for infants with negative, intermediate, and high EOC MRD levels, respectively (P < .0001). Analysis of EOI MRD according to the type of consolidation treatment showed that infants treated with lymphoid-style consolidation had 6-year DFS of 78.2% (95% CI, 51.4 to 91.3), 47.2% (95% CI, 33.0 to 60.1), and 23.2% (95% CI, 12.1 to 36.4) for negative, intermediate, and high MRD levels, respectively (P < .0001), while for myeloid-style-treated patients the corresponding figures were 45.0% (95% CI, 23.9 to 64.1), 41.3% (95% CI, 23.2 to 58.5), and 45.9% (95% CI, 29.4 to 60.9).

Conclusion: This study provides support for the idea that induction therapy selects patients for subsequent therapy; infants with high EOI MRD may benefit from AML-like consolidation (DFS 45.9% v 23.2%), whereas patients with low EOI MRD may benefit from ALL-like consolidation (DFS 78.2% v 45.0%). Patients with positive EOC MRD had dismal outcomes. These findings will be used for treatment interventions in the next Interfant protocol.

Trial registration: ClinicalTrials.gov NCT00550992.

Figures

FIG 1.
FIG 1.
Prognostic impact of minimal residual disease (MRD) levels at the end of induction (EOI), as shown by Kaplan-Meier estimates of disease-free survival (DFS), for all patients (A), patients treated with protocol IB (B), and patients treated with araC, daunorubicin, etoposide (ADE)/mitoxantrone, araC, etoposide (MAE) (C). Outcome by treatment given of patients according to MRD at EOI, negative (D), intermediate (E), and high (F). Neg, MRD-negative; Interm, MRD-intermediate (−4); High, MRD-high (≥ 5 × 10−4). CR1, first complete remission.
FIG 2.
FIG 2.
Prognostic impact of minimal residual disease (MRD) levels at the end of consolidation (EOC) (A-D) and after MARMA (E), as shown by Kaplan-Meier estimates of disease-free survival (DFS): EOC MRD for all patients (A), patients treated with protocol IB (B), patients treated with araC, daunorubicin, etoposide (ADE)/mitoxantrone, araC, etoposide (MAE) (C), and for all patients with MRD at EOI and EOC (D) and MRD after MARMA for all patients (E). Neg, MRD-negative; Interm, MRD-intermediate (−4); High, MRD-high (≥ 5 × 10−4). (D): Neg, MRD-negative at EOI and EOC; New Neg, MRD-positive at EOI and MRD-negative at EOC. For (A-D), One MRD-negative patient treated with ADE/MAE was excluded from these analyses due to missing data on first complete remission (CR1). For (E), One MRD-intermediate patient was excluded from this analysis due to missing data on CR1.
FIG 3.
FIG 3.
CONSORT diagram. ADE, araC, daunorubicin, etoposide; CR1, first complete remission; KMT2Ar, KMT2A-rearranged; MAE, mitoxantrone, araC, etoposide; MRD, minimal residual disease.

References

    1. Pieters R Schrappe M De Lorenzo P, et al. : A treatment protocol for infants younger than 1 year with acute lymphoblastic leukaemia (Interfant-99): An observational study and a multicentre randomised trial. Lancet 370:240-250, 2007
    1. Pieters R De Lorenzo P Ancliffe P, et al. : Outcome of infants younger than 1 year with acute lymphoblastic leukemia treated with the interfant-06 protocol: Results from an international phase III randomized study. J Clin Oncol 37:2246-2256, 2019
    1. Van Dongen JJ Seriu T Panzer-Grümayer ER, et al. : Prognostic value of minimal residual disease in acute lymphoblastic leukaemia in childhood. Lancet 352:1731-1738, 1998
    1. Faderl S Kantarjian HM Talpaz M, et al. : Clinical significance of minimal residual disease in leukemia. Int J Oncol 17:1277-1287, 2000
    1. Pieters R de Groot-Kruseman H Van der Velden V, et al. : Successful therapy reduction and intensification for childhood acute lymphoblastic leukemia based on minimal residual disease monitoring: Study ALL10 from the Dutch Childhood Oncology Group. J Clin Oncol 34:2591-2601, 2016
    1. Conter V Bartram CR Valsecchi MG, et al. : Molecular response to treatment redefines all prognostic factors in children and adolescents with B-cell precursor acute lymphoblastic leukemia: Results in 3184 patients of the AIEOP-BFM ALL 2000 study. Blood 115:3206-3214, 2010
    1. Schrappe M Bleckmann K Zimmermann M, et al. : Reduced-intensity delayed intensification in standard-risk pediatric acute lymphoblastic leukemia defined by undetectable minimal residual disease: Results of an international randomized trial (AIEOP-BFM ALL 2000). J Clin Oncol 36:244-253, 2018
    1. Schrappe M Valsecchi MG Bartram CR, et al. : Late MRD response determines relapse risk overall and in subsets of childhood T-cell ALL: Results of the AIEOP-BFM-ALL 2000 study. Blood 118:2077-2084, 2011
    1. Li A Goldwasser MA Zhou J, et al. : Distinctive IGH gene segment usage and minimal residual disease detection in infant acute lymphoblastic leukaemias. Br J Haematol 131:185-192, 2005
    1. Van der Velden VHJ Corral L Valsecchi MG, et al. : Prognostic significance of minimal residual disease in infants with acute lymphoblastic leukemia treated within the Interfant-99 protocol. Leukemia 23:1073-1079, 2009
    1. Popov A Buldini B De Lorenzo P, et al. : Prognostic value of minimal residual disease measured by flow-cytometry in two cohorts of infants with acute lymphoblastic leukemia treated according to either MLL-baby or interfant protocols. Leukemia 34:3042-3046, 2020
    1. Burmeister T Marschalek R Schneider B, et al. : Monitoring minimal residual disease by quantification of genomic chromosomal breakpoint sequences in acute leukemias with MLL aberrations. Leukemia 20:451-457, 2006
    1. Meyer C Schneider B Reichel M, et al. : Diagnostic tool for the identification of MLL rearrangements including unknown partner genes. Proc Natl Acad Sci U S A 102:449-454, 2005
    1. van der Velden VHJ Cazzaniga G Schrauder A, et al. : Analysis of minimal residual disease by Ig/TCR gene rearrangements: Guidelines for interpretation of real-time quantitative PCR data. Leukemia 21:604-611, 2007
    1. van der Velden VHJ, van Dongen JJM: MRD detection in acute lymphoblastic leukemia patients using IG/TR gene rearrangements as targets for real-time quantitative PCR. Methods Mol Biol 538, 115-150, 2009
    1. Cazzaniga G De Lorenzo P Alten J, et al. : Predictive value of minimal residual disease in Philadelphia-chromosome-positive acute lymphoblastic leukemia treated with imatinib in the European intergroup study of post-induction treatment of Philadelphia-chromosome-positive acute lymphoblastic leukemia, based on immunoglobulin/T-cell receptor and BCR/ABL1 methodologies. Haematologica 103:107-115, 2018
    1. Dworzak MN Buldini B Gaipa G, et al. : AIEOP-BFM consensus guidelines 2016 for flow cytometric immunophenotyping of pediatric acute lymphoblastic leukemia. Cytometry 94:82-93, 2018
    1. Fine JP, Gray RJ: A proportional hazards model for the subdistribution of a competing risk. J Am Stat Assoc 94:496-509, 1999
    1. Bland JM; Altman D: Statistical methods for assessing agreement between two methods of clinical measurement. Lancet 1:307-310, 1986
    1. van der Velden VHJ, Wijkhuijs JM, van Dongen JJM: Non-specific amplification of patient-specific Ig/TCR gene rearrangements depends on the time point during therapy: Implications for minimal residual disease monitoring. Leukemia 22:641-644, 2008
    1. Kotrova M van der Velden VHJ van Dongen JJM, et al. : Next-generation sequencing indicates false-positive MRD results and better predicts prognosis after SCT in patients with childhood ALL. Bone Marrow Transplant 52:962-968, 2017
    1. Enshaei A O’Connor D Bartram J, et al. : A validated novel continuous prognostic index to deliver stratified medicine in pediatric acute lymphoblastic leukemia. Blood 135:1438-1446, 2020
    1. Maloney KW Devidas M Wang C, et al. : Outcome in children with standard-risk B-cell acute lymphoblastic leukemia: Results of Children's Oncology Group Trial AALL0331. J Clin Oncol 38:602-612, 2020
    1. Burke MJ Salzer WL Devidas M, et al. : Replacing cyclophosphamide/cytarabine/mercaptopurine with cyclophosphamide/etoposide during consolidation/delayed intensification does not improve outcome for pediatric B-cell acute lymphoblastic leukemia: A report from the COG. Haematologica 104:986-992, 2019

Source: PubMed

3
Abonneren