Defining therapy goals for major molecular remission in chronic myeloid leukemia: results of the randomized CML Study IV

Susanne Saussele, Rüdiger Hehlmann, Alice Fabarius, Sabine Jeromin, Ulrike Proetel, Sebastien Rinaldetti, Katharina Kohlbrenner, Hermann Einsele, Christiane Falge, Lothar Kanz, Andreas Neubauer, Michael Kneba, Frank Stegelmann, Michael Pfreundschuh, Cornelius F Waller, Elisabeth Oppliger Leibundgut, Dominik Heim, Stefan W Krause, Wolf-Karsten Hofmann, Joerg Hasford, Markus Pfirrmann, Martin C Müller, Andreas Hochhaus, Michael Lauseker, Susanne Saussele, Rüdiger Hehlmann, Alice Fabarius, Sabine Jeromin, Ulrike Proetel, Sebastien Rinaldetti, Katharina Kohlbrenner, Hermann Einsele, Christiane Falge, Lothar Kanz, Andreas Neubauer, Michael Kneba, Frank Stegelmann, Michael Pfreundschuh, Cornelius F Waller, Elisabeth Oppliger Leibundgut, Dominik Heim, Stefan W Krause, Wolf-Karsten Hofmann, Joerg Hasford, Markus Pfirrmann, Martin C Müller, Andreas Hochhaus, Michael Lauseker

Abstract

Major molecular remission (MMR) is an important therapy goal in chronic myeloid leukemia (CML). So far, MMR is not a failure criterion according to ELN management recommendation leading to uncertainties when to change therapy in CML patients not reaching MMR after 12 months. At monthly landmarks, for different molecular remission status Hazard ratios (HR) were estimated for patients registered to CML study IV who were divided in a learning and a validation sample. The minimum HR for MMR was found at 2.5 years with 0.28 (compared to patients without remission). In the validation sample, a significant advantage for progression-free survival (PFS) for patients in MMR could be detected (p-value 0.007). The optimal time to predict PFS in patients with MMR could be validated in an independent sample at 2.5 years. With our model we provide a suggestion when to define lack of MMR as therapy failure and thus treatment change should be considered. The optimal response time for 1% BCR-ABL at about 12-15 months was confirmed and for deep molecular remission no specific time point was detected. Nevertheless, it was demonstrated that the earlier the MMR is achieved the higher is the chance to attain deep molecular response later.

Trial registration: ClinicalTrials.gov NCT00055874.

Conflict of interest statement

The authors declare that they have no conflict of interest. S.S.: Novartis: Honoraria, Research Funding, Travel Other; B.-M.S.: Honoraria, Research Funding, Travel, Travel Other; Pfizer: Honoraria, Travel, Travel Other. R.H.: B.-M.S.: Research Funding; Novartis: Research Funding. S.J.: MLL Munich Leukemia Laboratory: Equity Ownership. A.N.: MedUpdate: Honoraria, Speakers Bureau. M.K.: Novartis: Consultancy, Equity Ownership, Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding. M.Pfi.: Novartis: Consultancy; B.-M.S.: Honoraria. A.H.: Novartis: Consultancy, Honoraria, Research Funding; B.-M.S.: Consultancy, Honoraria; ARIAD: Honoraria, Research Funding; Pfizer: Consultancy, Research Funding. M.C.M.: Novartis: Honoraria, Research Funding; B.-M.S.: Honoraria, Research Funding; ARIAD: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding.

Figures

Fig. 1
Fig. 1
Consort diagram
Fig. 2
Fig. 2
Distribution of patients according to remission status achieved at different landmarks from 6 months to 5 years (a) and respective percentages of these states (b)
Fig. 3
Fig. 3
Median hazard ratio functions for the comparison of patients who had achieved a MMR resp. MR2 to those who did not have any remission at different landmarks with respect to PFS from 6 months to 5 years together with the 95% confidence intervals. On the y axis, the hazard ratio for PFS is plotted on a logarithmic scale. Note that on the x axis the landmark time is plotted instead of the event time. A hazard ratio of, e.g., 0.5 at landmark 6 months indicates that patients with MMR have only half the risk of patients with no MMR before or at 6 months
Fig. 4
Fig. 4
Median hazard ratio function for the comparison of patients who had achieved an MMR to those who did not have any remission at different landmarks with respect to MR4.5 from 6 months to 5 years together with the 95% confidence intervals. On the y axis, the hazard ratio for MR4.5 is plotted on a logarithmic scale. Note that on the x axis the landmark time is plotted instead of the event time

References

    1. Bower H, Bjorkholm M, Dickman PW, Hoglund M, Lambert PC, Andersson TM. Life expectancy of patients with chronic myeloid leukemia approaches the life expectancy of the general population. J Clin Oncol. 2016;34:2851–7. doi: 10.1200/JCO.2015.66.2866.
    1. Saussele S, Krauss MP, Hehlmann R, Lauseker M, Proetel U, Kalmanti L, et al. Impact of comorbidities on overall survival in patients with chronic myeloid leukemia: results of the randomized CML Study IV. Blood. 2015;126:42–49. doi: 10.1182/blood-2015-01-617993.
    1. Pfirrmann M, Baccarani M, Saussele S, Guilhot J, Cervantes F, Ossenkoppele G, et al. Prognosis of long-term survival considering disease-specific death in patients with chronic myeloid leukemia. Leukemia. 2016;30:48–56. doi: 10.1038/leu.2015.261.
    1. Hasford J, Baccarani M, Hoffmann V, Guilhot J, Saussele S, Rosti G, et al. Predicting complete cytogenetic response and subsequent progression-free survival in 2060 patients with CML on imatinib treatment: the EUTOS score. Blood. 2011;118:686–92. doi: 10.1182/blood-2010-12-319038.
    1. Baccarani M, Deininger MW, Rosti G, Hochhaus A, Soverini S, Apperley JF, et al. European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood. 2013;122:872–84. doi: 10.1182/blood-2013-05-501569.
    1. Hehlmann R, Lauseker M, Jung-Munkwitz S, Leitner A, Mueller MC, Pletsch N, et al. Tolerability-adapted imatinib 800 mg/d versus 400 mg/d versus 400 mg/d plus interferon-alpha in newly diagnosed chronic myeloid leukemia. J Clin Oncol. 2011;29:1634–42. doi: 10.1200/JCO.2010.32.0598.
    1. Hehlmann R, Lauseker M, Saußele S, Pfirrmann M, Krause SW, Kolb HJ, et al. Assessment of imatinib as first-line treatment of chronic myeloid leukemia: 10–year survival results of the randomized CML study IV and impact of non-CML determinants. Leukemia. 2017;31:2398–406. doi: 10.1038/leu.2017.253.
    1. Hughes TP, Hochhaus A, Branford S, Müller MC, Kaeda JS, Foroni L, et al. Long-term prognostic significance of early molecular response to imatinib in newly diagnosed chronic myeloid leukemia: an analysis from the International Randomized Study of Interferon and STI571 (IRIS) Blood. 2010;116:3758–65. doi: 10.1182/blood-2010-03-273979.
    1. Falchi L, Kantarjian HM, Wang X, Verma D, Quintas-Cardama A, O’Brien S, et al. Significance of deeper molecular responses in patients with chronic myeloid leukemia in early chronic phase treated with tyrosine kinase inhibitors. Am J Hematol. 2013;88:1024–9. doi: 10.1002/ajh.23560.
    1. Kalmanti L, Saussele S, Lauseker M, Muller MC, Dietz CT, Heinrich L, et al. Safety and efficacy of imatinib in CML over a period of 10 years: data from the randomized CML-study IV. Leukemia. 2015;29:1123–32. doi: 10.1038/leu.2015.36.
    1. Druker BJ, Guilhot F, O’Brien SG, Gathmann I, Kantarjian H, Gattermann N, et al. Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med. 2006;355:2408–17. doi: 10.1056/NEJMoa062867.
    1. Hehlmann R, Müller MC, Lauseker M, Hanfstein B, Fabarius A, Schreiber A, et al. Deep molecular response is reached by the majority of patients treated with imatinib, predicts survival, and is achieved more quickly by optimized high-dose imatinib: results from the randomized CML-study IV. J Clin Oncol. 2014;32:415–23. doi: 10.1200/JCO.2013.49.9020.
    1. Cross NCP, White HE, Müller MC, Saglio G, Hochhaus A. Standardized definitions of molecular response in chronic myeloid leukemia. Leukemia. 2012;26:2172–5. doi: 10.1038/leu.2012.104.
    1. Müller MC, Cross NCP, Erben P, Schenk T, Hanfstein B, Ernst T, et al. Harmonization of molecular monitoring of CML therapy in Europe. Leukemia. 2009;23:1957–63. doi: 10.1038/leu.2009.168.
    1. Lauseker M, Hanfstein B, Haferlach C, Schnittger S, Pfirrmann M, Fabarius A, et al. Equivalence of BCR-ABL transcript levels with complete cytogenetic remission in patients with chronic myeloid leukemia in chronic phase. J Cancer Res Clin Oncol. 2014;140:1965–9. doi: 10.1007/s00432-014-1746-8.
    1. Hughes T, Deininger M, Hochhaus A, Branford S, Radich J, Kaeda J, et al. Monitoring CML patients responding to treatment with tyrosine kinase inhibitors: review and recommendations for harmonizing current methodology for detecting BCR-ABL transcripts and kinase domain mutations and for expressing results. Blood. 2006;108:28–37. doi: 10.1182/blood-2006-01-0092.
    1. Van Houwelingen HC. Dynamic prediction by landmarking in event history analysis. Scand J Stat. 2007;34:70–85. doi: 10.1111/j.1467-9469.2006.00529.x.
    1. Van Houwelingen HC, Putter H. Dynamic predicting by landmarking as an alternative for multi-state modeling: an application to acute lymphoid leukemia data. Lifetime Data Anal. 2008;14:447–63. doi: 10.1007/s10985-008-9099-8.
    1. Hoffmann VS, Baccarani M, Lindoerfer D, Castagnetti F, Turkina A, Zaritsky A, et al. The EUTOS prognostic score: review and validation in 1288 patients with CML treated frontline with imatinib. Leukemia. 2013;27:2016–22. doi: 10.1038/leu.2013.171.
    1. Mahon FX, Rea D, Guilhot J, Guilhot F, Huguet F, Nicolini F, et al. Discontinuation of imatinib in patients with chronic myeloid leukaemia who have maintained complete molecular remission for at least 2 years: the prospective, multicentre Stop Imatinib (STIM) trial. Lancet Oncol. 2010;11:1029–35. doi: 10.1016/S1470-2045(10)70233-3.
    1. Saussele S, Richter J, Hochhaus A, Mahon FX. The concept of treatment-free remission in chronic myeloid leukemia. Leukemia. 2016;30:1638–47. doi: 10.1038/leu.2016.115.
    1. Ross DM, Branford S, Seymour JF, Schwarer AP, Arthur C, Yeung DT, et al. Safety and efficacy of imatinib cessation for CML patients with stable undetectable minimal residual disease: results from the TWISTER study. Blood. 2013;122:515–22. doi: 10.1182/blood-2013-02-483750.
    1. Hughes TP, Saglio G, Kantarjian HM, Guilhot F, Niederwieser D, Rosti G, et al. Early molecular response predicts outcomes in patients with chronic myeloid leukemia in chronic phase treated with frontline nilotinib or imatinib. Blood. 2013;123:1353–60. doi: 10.1182/blood-2013-06-510396.
    1. Hochhaus A, Rosti G, Cross NC, Steegmann JL, le Coutre P, Ossenkoppele G, et al. Frontline nilotinib in patients with chronic myeloid leukemia in chronic phase: results from the European ENEST1st study. Leukemia. 2015;30:57–64. doi: 10.1038/leu.2015.270.
    1. Firwana B, Sonbol MB, Diab M, Raza S, Hasan R, Yousef I, et al. Tyrosine kinase inhibitors as a first-line treatment in patients with newly diagnosed chronic myeloid leukemia in chronic phase: A mixed-treatment comparison. Int J Cancer. 2016;138:1545–53. doi: 10.1002/ijc.29889.
    1. Hjorth-Hansen H, Stenke L, Soderlund S, Dreimane A, Ehrencrona H, Gedde-Dahl T, et al. Dasatinib induces fast and deep responses in newly diagnosed chronic myeloid leukaemia patients in chronic phase: clinical results from a randomised phase-2 study (NordCML006) Eur J Haematol. 2015;94:243–50. doi: 10.1111/ejh.12423.
    1. Jabbour E, Kantarjian HM, Saglio G, Steegmann JL, Shah NP, Boque C, et al. Early response with dasatinib or imatinib in chronic myeloid leukemia: 3-year follow-up from a randomized phase 3 trial (DASISION) Blood. 2014;123:494–500. doi: 10.1182/blood-2013-06-511592.
    1. Brummendorf TH, Cortes JE, de Souza CA, Guilhot F, Duvillie L, Pavlov D, et al. Bosutinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukaemia: results from the 24-month follow-up of the BELA trial. Br J Haematol. 2015;168:69–81. doi: 10.1111/bjh.13108.

Source: PubMed

3
Abonner