Effect of mitoxantrone on outcome of children with first relapse of acute lymphoblastic leukaemia (ALL R3): an open-label randomised trial

Catriona Parker, Rachel Waters, Carly Leighton, Jeremy Hancock, Rosemary Sutton, Anthony V Moorman, Philip Ancliff, Mary Morgan, Ashish Masurekar, Nicholas Goulden, Nina Green, Tamas Révész, Philip Darbyshire, Sharon Love, Vaskar Saha, Catriona Parker, Rachel Waters, Carly Leighton, Jeremy Hancock, Rosemary Sutton, Anthony V Moorman, Philip Ancliff, Mary Morgan, Ashish Masurekar, Nicholas Goulden, Nina Green, Tamas Révész, Philip Darbyshire, Sharon Love, Vaskar Saha

Abstract

Background: Although survival of children with acute lymphoblastic leukaemia has improved greatly in the past two decades, the outcome of those who relapse has remained static. We investigated the outcome of children with acute lymphoblastic leukaemia who relapsed on present therapeutic regimens.

Methods: This open-label randomised trial was undertaken in 22 centres in the UK and Ireland and nine in Australia and New Zealand. Patients aged 1-18 years with first relapse of acute lymphoblastic leukaemia were stratified into high-risk, intermediate-risk, and standard-risk groups on the basis of duration of first complete remission, site of relapse, and immunophenotype. All patients were allocated to receive either idarubicin or mitoxantrone in induction by stratified concealed randomisation. Neither patients nor those giving interventions were masked. After three blocks of therapy, all high-risk group patients and those from the intermediate group with postinduction high minimal residual disease (≥10(-4) cells) received an allogenic stem-cell transplant. Standard-risk and intermediate-risk patients with postinduction low minimal residual disease (<10(-4) cells) continued chemotherapy. The primary outcome was progression-free survival and the method of analysis was intention-to-treat. Randomisation was stopped in December, 2007 because of differences in progression-free and overall survival between the two groups. This trial is registered, reference number ISCRTN45724312.

Findings: Of 239 registered patients, 216 were randomly assigned to either idarubicin (109 analysed) or mitoxantrone (103 analysed). Estimated 3-year progression-free survival was 35·9% (95% CI 25·9-45·9) in the idarubicin group versus 64·6% (54·2-73·2) in the mitoxantrone group (p=0·0004), and 3-year overall survival was 45·2% (34·5-55·3) versus 69·0% (58·5-77·3; p=0·004). Differences in progression-free survival between groups were mainly related to a decrease in disease events (progression, second relapse, disease-related deaths; HR 0·56, 0·34-0·92, p=0·007) rather than an increase in adverse treatment effects (treatment death, second malignancy; HR 0·52, 0·24-1·11, p=0·11).

Interpretation: As compared with idarubicin, mitoxantrone conferred a significant benefit in progression-free and overall survival in children with relapsed acute lymphobastic leukaemia, a potentially useful clinical finding that warrants further investigation.

Funding: Cancer Research UK, Leukaemia and Lymphoma Research, Cancer Council NSW, and Sporting Chance Cancer Foundation.

Copyright © 2010 Elsevier Ltd. All rights reserved.

Figures

Figure 1
Figure 1
Risk stratification (A) and trial design (B) (A) Stratification according to immunophenotype, site of relapse, and time to relapse. Risk groups: very early refers to less than 18 months from first diagnosis; early refers to 18 months or more after first diagnosis and less than 6 months from stopping therapy; and late refers to 6 months or more after stopping therapy. (B) MRD sampling TPs are marked. At TP1, standard-risk and intermediate-risk patients with MRD lower than 10−4 cells were ineligible, and high-risk and intermediate-risk patients with MRD of 10−4 cells or more were eligible for allo-SCT. Localised radiotherapy was given to those with extramedullary disease and not proceeding to allo-SCT. When MRD assessment was not possible in intermediate-risk patients, allo-SCT was allowed provided relapse occurred within 24 months of stopping therapy. Details of the phases are provided in table 1. MRD=minimal residual disease. TP=timepoint. Allo-SCT=allogenic stem-cell transplant.
Figure 2
Figure 2
Trial profile All patients were scheduled to receive two doses of anthracycline. Data for having received the drug were not available for 12 patients, six in each group.
Figure 3
Figure 3
Kaplan Meier estimates of progression-free (A) and overall (B) survival by randomised treatment 3-year estimated survival percentages (95% CI) are presented.
Figure 4
Figure 4
Randomised drug effect on progression-free survival by patient characteristics, from Cox models with interactions p values for interaction test the hypothesis that the randomised treatment effect varies between the different subgroups. Hazard ratios indicate that all subgroups show a treatment effect favouring mitoxantrone. HR=hazard ratio. MRD=minimal residual disease. Cyto=cytogenetic subgroups. *Poor refers to poor outcome after relapse (webappendix p 2).

References

    1. Schrappe M, Nachman J, Hunger S. Educational symposium on long-term results of large prospective clinical trials for childhood acute lymphoblastic leukemia (1985–2000) Leukemia. 2010;24:253–254.
    1. Harned TM, Gaynon P. Relapsed acute lymphoblastic leukemia: current status and future opportunities. Curr Oncol Rep. 2008;10:453–458.
    1. Lawson SE, Harrison G, Richards S. The UK experience in treating relapsed childhood acute lymphoblastic leukaemia: a report on the medical research council UKALLR1 study. Br J Haematol. 2000;108:531–543.
    1. Roy A, Cargill A, Love S. Outcome after first relapse in childhood acute lymphoblastic leukaemia—lessons from the United Kingdom R2 trial. Br J Haematol. 2005;130:67–75.
    1. Tallen G, Ratei R, Mann G. Long-term outcome in children with relapsed acute lymphoblastic leukemia after time-point and site-of-relapse stratification and intensified short-course multidrug chemotherapy: results of trial ALL-REZ BFM 90. J Clin Oncol. 2010;28:2339–2347.
    1. Malempati S, Gaynon PS, Sather H, La MK, Stork LC. Outcome after relapse among children with standard-risk acute lymphoblastic leukemia: Children's Oncology Group study CCG-1952. J Clin Oncol. 2007;25:5800–5807.
    1. Krishnan S, Wade R, Moorman AV. Temporal changes in the incidence and pattern of central nervous system relapses in children with acute lymphoblastic leukaemia treated on four consecutive Medical Research Council trials, 1985–2001. Leukemia. 2010;24:450–459.
    1. Raetz EA, Borowitz MJ, Devidas M. Reinduction platform for children with first marrow relapse of acute lymphoblastic leukemia: a Children's Oncology Group Study. J Clin Oncol. 2008;26:3971–3978.
    1. Stam RW, den Boer ML, Meijerink JP. Differential mRNA expression of Ara-C-metabolizing enzymes explains Ara-C sensitivity in MLL gene-rearranged infant acute lymphoblastic leukemia. Blood. 2003;101:1270–1276.
    1. von Stackelberg A, Hartmann R, Buhrer C. High-dose compared with intermediate-dose methotrexate in children with a first relapse of acute lymphoblastic leukemia. Blood. 2008;111:2573–2580.
    1. Wells RJ, Feusner J, Devney R. Sequential high-dose cytosine arabinoside-asparaginase treatment in advanced childhood leukemia. J Clin Oncol. 1985;3:998–1004.
    1. Testi AM, Del Giudice I, Arcese W. A single high dose of idarubicin combined with high-dose ARA-C for treatment of first relapse in childhood ‘high-risk’ acute lymphoblastic leukaemia: a study of the AIEOP group. Br J Haematol. 2002;118:741–747.
    1. Reid JM, Pendergrass TW, Krailo MD, Hammond GD, Ames MM. Plasma pharmacokinetics and cerebrospinal fluid concentrations of idarubicin and idarubicinol in pediatric leukemia patients: a Childrens Cancer Study Group report. Cancer Res. 1990;50:6525–6528.
    1. Hongo T, Fujii Y. In vitro chemosensitivity of lymphoblasts at relapse in childhood leukemia using the MTT assay. Int J Hematol. 1991;54:219–230.
    1. Chan A, Weilbach FX, Toyka KV, Gold R. Mitoxantrone induces cell death in peripheral blood leucocytes of multiple sclerosis patients. Clin Exp Immunol. 2005;139:152–158.
    1. Errington F, Willmore E, Leontiou C, Tilby MJ, Austin CA. Differences in the longevity of topo IIalpha and topo IIbeta drug-stabilized cleavable complexes and the relationship to drug sensitivity. Cancer Chemother Pharmacol. 2004;53:155–162.
    1. van der Velden VH, Hochhaus A, Cazzaniga G, Szczepanski T, Gabert J, van Dongen JJ. Detection of minimal residual disease in hematologic malignancies by real-time quantitative PCR: principles, approaches, and laboratory aspects. Leukemia. 2003;17:1013–1034.
    1. Trotti A, Colevas AD, Setser A. CTCAE v3.0: development of a comprehensive grading system for the adverse effects of cancer treatment. Semin Radiat Oncol. 2003;13:176–181.
    1. Clinical trial authorisations: Safety reporting—SUSARS and ASRs. 2010. (accessed June 1, 2010).
    1. McCullagh P, Nelder JA. Generalised Linear Models. Chapman and Hall; London: 1983.
    1. Fine JP, Gray RJ. A proportional hazards model for the subdistribution of a competing risk. J Am Stat Assoc. 1999;94:496–509.
    1. Gray RJ. A class of K-sample tests for comparing the cumulative incidence of a competing risk. Ann Stat. 1988;16:1140–1154.
    1. Henze G, Fengler R, Hartmann R. Six-year experience with a comprehensive approach to the treatment of recurrent childhood acute lymphoblastic leukemia (ALL-REZ BFM 85). A relapse study of the BFM group. Blood. 1991;78:1166–1172.
    1. Einsiedel HG, von Stackelberg A, Hartmann R. Long-term outcome in children with relapsed ALL by risk-stratified salvage therapy: results of trial acute lymphoblastic leukemia-relapse study of the Berlin-Frankfurt-Munster Group 87. J Clin Oncol. 2005;23:7942–7950.
    1. Testi AM, Moleti ML, Giona F. A single high dose of idarubicin combined with high-dose ARA-C (MSKCC ALL-3 protocol) in adult and pediatric patients with acute lymphoblastic leukemia. Experience at the University “La Sapienza” of Rome. Haematologica. 1997;82:664–667.
    1. Ko RH, Ji L, Barnette P. Outcome of patients treated for relapsed or refractory acute lymphoblastic leukemia: a Therapeutic Advances in Childhood Leukemia Consortium study. J Clin Oncol. 2010;28:648–654.
    1. Nachman JB, Sather HN, Sensel MG. Augmented post-induction therapy for children with high-risk acute lymphoblastic leukemia and a slow response to initial therapy. N Engl J Med. 1998;338:1663–1671.
    1. Seibel NL, Steinherz PG, Sather HN. Early postinduction intensification therapy improves survival for children and adolescents with high-risk acute lymphoblastic leukemia: a report from the Children's Oncology Group. Blood. 2008;111:2548–2555.
    1. Fleming TR. Surrogate endpoints and FDA's accelerated approval process. Health Aff (Millwood) 2005;24:67–78.
    1. Susman E. Accelerated approval seen as triumph and roadblock for cancer drugs. J Natl Cancer Inst. 2004;96:1495–1496.
    1. Parker BS, Buley T, Evison BJ. A molecular understanding of mitoxantrone-DNA adduct formation: effect of cytosine methylation and flanking sequences. J Biol Chem. 2004;279:18814–18823.
    1. Karl S, Pritschow Y, Volcic M. Identification of a novel pro-apopotic function of NF-kappaB in the DNA damage response. J Cell Mol Med. 2009;13:4239–4256.
    1. Cao C, Han Y, Ren Y, Wang Y. Mitoxantrone-mediated apoptotic B16-F1 cells induce specific anti-tumor immune response. Cell Mol Immunol. 2009;6:469–475.
    1. Gibson BE, Wheatley K, Hann IM. Treatment strategy and long-term results in paediatric patients treated in consecutive UK AML trials. Leukemia. 2005;19:2130–2138.

Source: PubMed

3
S'abonner