Next Article in Journal
Risk Factors and Outcome of C. difficile Infection after Hematopoietic Stem Cell Transplantation
Next Article in Special Issue
Molecular Testing in CML between Old and New Methods: Are We at a Turning Point?
Previous Article in Journal
Glycation Increases the Risk of Microbial Traversal through an Endothelial Model of the Human Blood-Brain Barrier after Use of Anesthetics
Previous Article in Special Issue
Health-Related Quality of Life in Patients with Chronic Myeloid Leukemia Treated with First- Versus Second-Generation Tyrosine Kinase Inhibitors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The New ELN Recommendations for Treating CML

by
Rüdiger Hehlmann
ELN-Foundation, Weinheim and Medical Faculty Mannheim of Heidelberg University, 69126 Mannheim, Germany
J. Clin. Med. 2020, 9(11), 3671; https://doi.org/10.3390/jcm9113671
Submission received: 23 October 2020 / Revised: 10 November 2020 / Accepted: 11 November 2020 / Published: 16 November 2020
(This article belongs to the Special Issue Prognosis and Therapy of Chronic Myeloid Leukemia)

Abstract

:
After normal survival has been achieved in most patients with chronic myeloid leukemia (CML), a new goal for treating CML is survival at good quality of life, with treatment discontinuation in sustained deep molecular response (DMR; MR4 or deeper) and treatment-free remission (TFR). Four tyrosine kinase inhibitors (TKIs) have been approved for first-line therapy: imatinib, dasatinib, nilotinib, bosutinib. Unexpectedly, the outcome of long-term randomized trials has shown that faster response as achieved by higher doses of imatinib, imatinib in combination, or second-generation (2G)-TKIs, does not translate into a survival advantage. Serious and frequent, and in part cumulative long-term toxicities, have led to a reevaluation of the role of 2G-TKIs in first-line therapy. Generic imatinib is the current most cost-effective first-line therapy in the chronic phase. A change of treatment is recommended when intolerance cannot be ameliorated or molecular milestones are not reached. Patient comorbidities and contraindications of all TKIs must be considered. Risk profile at diagnosis should be assessed with the EUTOS score for long-term survival (ELTS). Monitoring of response is by polymerase chain reaction (PCR). Cytogenetics is still required in the case of atypical translocations, atypical transcripts, and additional chromosomal aberrations. TKIs are contraindicated during pregnancy. Since the majority of patients are at risk of lifelong exposure to TKIs, amelioration of chronic low-grade side effects is important.

1. Introduction

Over the last two decades, clinical trials and population-based registries have shown that the vast majority of chronic myeloid leukemia (CML) patients treated with the tyrosine kinase inhibitor (TKI) imatinib have achieved normal life expectancy [1,2,3,4,5]. The European LeukemiaNet (ELN) has accounted for this development with management recommendations in 2006, 2009, and 2013 [6,7,8].
However, expectations of a general cure for CML remain unmet. Most patients have residual molecular disease, are not yet cured, and require life-long therapy. CML did not become the expected model disease for targeting other leukemias or cancers with TKIs, but the elucidation of pathogenesis for successfully treating cancer has been convincingly demonstrated in CML.
Some patients, who stop treatment in sustained deep molecular remission (DMR), achieve durable treatment-free remissions (TFRs) and may be cured. Treatment discontinuation and TFRs have become an important new treatment goal of CML. Duration of DMR (MR4 or deeper) may be the best predictor of success. A review of CML, spanning from its first recognition to its current progress, has recently been published [9].
To reflect this new situation, the ELN has revised and updated its recommendations for treating CML [10]. This article will review, and in some instances update, these recommendations.

2. Initial Diagnostic Workup

At baseline, a complete blood cell count with differential should be obtained and a physical examination should be conducted, paying special attention to spleen and liver size. A bone marrow aspirate is recommended, as is a cytogenetic analysis for the verification of the Philadelphia (Ph)-chromosome. A qualitative polymerase chain reaction (PCR) is required to identify BCR-ABL1 transcripts and transcript types. An electrocardiogram, standard clinical chemistry, and a hepatitis serology are recommended for the recognition of comorbidities [10].
Risk score at diagnosis: In the TKI era, evaluation of risk at diagnosis should be done with the EUTOS score for long-term survival (ELTS), since it predicts death by CML more accurately than the other scores [11,12]. The ELTS score uses the same variables as the Sokal score, but with different weights (Table 1). Age has become a much less important factor, since in the TKI era death by CML is less dependent on age. The numbers in red highlight the most relevant differences between Sokal and ELTS scores.

3. Treatment Monitoring

Monitoring of treatment is now almost exclusively done molecularly by quantitative PCR, according to the International Scale (IS) [13,14]. Molecular monitoring of treatment with imatinib was systematically undertaken by CML study IV since its beginning in 2002 [2,15,16] and resulted in the recognition of the impact of early response on outcome and of benchmark times for deep molecular remission (DMR). PCR results of the IRIS study served to define response milestones based on the IS [7,8,17]. BCR-ABL1 ≤1% was determined to be equivalent to complete cytogenetic remission [18]. Results of quantitative PCR depend on the ability of laboratories to measure absolute numbers of control-gene transcripts (ABL1, GUS), and to achieve the PCR sensitivity required for BCR-ABL1 detection. The updated milestone proposal by the ELN is shown in Table 2.
Response milestones are the same for first- and second-line therapy. Cytogenetics is no longer recommended for routine monitoring. A cytogenetic analysis with bone marrow puncture is indicated if molecular monitoring is not possible, as in the case of atypical translocations, atypical transcripts, or additional chromosomal aberrations (ACAs) [10].
For patients aiming at TFR, BCR-ABL1 < 0.01% (<MR4) at any time is considered to be the optimal response.
The ELN recommendations accept a change of treatment if major molecular remission (MMR) is not reached by 36–48 months [10].

4. First-Line Treatment

First-line treatment continues to be a TKI. Meanwhile, 4 TKIs are approved for first-line use: imatinib (Glivec, Novartis), dasatinib (Sprycel, BMS), nilotinib (Tasigna, Novartis), and now also bosutinib (Bosulif, Pfizer). Radotinib (Supect, Dae Wong Pharma) is approved in South Korea only and is not discussed further in this review.
Imatinib: The first-generation TKI imatinib was compared to conventional IFN-based therapy in the IRIS study and showed superior cytogenetic and molecular response rates and a survival advantage [17]. Tolerability was better with imatinib than with IFN. The most frequent side effects were fluid retention, gastro-intestinal symptoms, and fatigue [17,19,20]. No serious toxicity has surfaced in more than 20 years of use. Patients with low cardiac ejection fractions and reduced glomerular filtration rates need to be observed more carefully [20].
Five- and ten-year data of randomized trials with imatinib combined with IFN or low-dose cytarabine, with imatinib in a higher dosage, or with the second-generation TKIs (2G-TKIs) dasatinib and nilotinib, have shown that faster responses do not translate into better survival rates than treatment with imatinib alone at the standard dose of 400 mg daily [2,21,22].
Generic imatinib, now available worldwide, has been determined to be a cost-effective initial treatment in chronic phase (CP) CML in several independent evaluations [23,24,25].
Dasatinib: This 2G-TKI inhibits the imatinib-resistant BCR-ABL1 kinase domain (KD)-mutations Y253H, E255V/K, and F359V/I/C, and induces responses faster than imatinib. Regarding the pleuro-pulmonary toxicity of dasatinib, a history of pleura and lung diseases represents a contraindication for dasatinib [20,21]. The approved dose is 100 mg/day in CPs and 140 mg/day in advanced phases. A more tolerable dose of 50 mg/day also seems to be effective in CPs [26].
Nilotinib: Nilotinib is another 2G-TKI that inhibits the imatinib-resistant BCR-ABL1 KD mutations F317L/VLI/C, T315A, and V299L, and induces faster responses than imatinib. Regarding the cardio-vascular toxicity of nilotinib, a history of coronary heart disease, cerebral-vascular events, or peripheral arterial obstructive disease represent contraindications for nilotinib. Hypertension, diabetes mellitus, hypercholesterolemia, and a history of pancreatitis are relative contraindications [20,22,27]. As a first-line treatment, the approved dose is 300 mg twice daily.
The lack of a survival advantage, and the serious and in part cumulative long-term toxicity, have led to the re-evaluation of 2G-TKIs for use in first-line therapy. Comorbidities of patients [28] and long-term toxicities of TKIs need to be carefully considered [19,20].
Bosutinib: Bosutinib is a third 2G-TKI which recognizes imatinib-resistant BCR-ABL1 KD mutations, and also induces faster responses than imatinib. No relevant comorbidities have been detected as contraindications for bosutinib [29]. However, no long-term data are available yet. Frequently, annoying but usually self-limited diarrhea occurs. If needed, treatment with loperamide is recommended. The approved dose is 400 mg/day in first-line treatments.

5. Second- and Higher-Line Treatment

Second-line therapy, after intolerance or resistance to the first-line TKI, is usually also a TKI. If resistance is suspected, compliance should be assessed first. If resistance is confirmed, a mutational analysis should be initiated, and treatment changed on clinical grounds without waiting for the mutation results. Resistant BCR-ABL1 KD-mutations account for about a third of cases with resistance in CPs, and are relatively rare, occurring in 4.6% in 1536 CP patients over 10 years [2]. Resistant mutations are more frequent in advanced phases. A definite choice of the second-line TKI is made according to the mutational analysis and the patient’s comorbidities. An allogeneic transplantation must be considered, and a donor search initiated.
BCR-ABL1 KD mutations are detectable with sensitivities of about 20% by Sanger sequencing, and of about 3% with next-generation sequencing (NGS) [30,31]. NGS is the recommended technology for the detection of resistant BCR-ABL1 mutations.
Ponatinib: Ponatinib, a third-generation TKI, is still the only approved TKI with efficacy against the resistant T315I mutation. After resistance to two TKIs, treatment should be changed to another 2G-TKI or to ponatinib [32]. Because of the cardio-vascular toxicity of ponatinib, a dose reduction from 45 to 15 mg/day after a response is achieved may be appropriate. Evidence for this reduction is provided by preliminary results from the randomized dose-optimization study Optic, comparing efficacy and tolerability of ponatinib at 45, 30, and 15 mg/day [33]. In the case of a T315I mutation, or of compound mutations or progression, ponatinib at a dose of 45 mg/day should be given immediately, with reduction to 15 mg/day after response.
All treatment decisions should be made by shared decision-making [34], after full information is given to the patient on the efficacy and side effects of the respective treatments, including transplantation, the option of treatment discontinuation, and TFR.
Allogeneic stem cell transplantation: Transplantation has retained an important place in CML treatment. Early transplantation in the CP is associated with better outcomes than late transplant [35]. Transplant-related mortality is low in CPs, but graft-versus-host disease (GvHD) remains a problem. In the TKI era, transplantation has shifted from CP to blast crisis (BC). In a report from the German CML Study Group, 6-year survival of 28 imatinib-pretreated patients in advanced phase, and 25 in blast crisis, was 49% [36,37]. Similar data were reported from China [38] and by another German group [39]. Most long-time survivors after BC had undergone transplantation (Figure 1), making transplantation a preferred treatment modality in BC. Early transplantation also seems to be more successful when high-risk additional chromosomal aberrations (ACAs) are present [40].

6. CML End-Phase and Blast Crisis

The outcome of patients in BC has not much improved. Survival is generally less than one year, and new approaches are needed. Earlier recognition of BC by genetic assessment (ACAs, mutations) might improve outcomes [40,41,42,43,44,45]. End-phase CML comprises early progression with emerging high-risk ACAs, and late progression with failing hematopoiesis and blast increase. Figure 2 illustrates the current understanding of CML progression and of ABL1 activation by radical oxygen species (ROS) [46], and/or polycomb repressive complex (PRC) [47] driven epigenetic reprogramming [48].
Chromosomal aberrations, in addition to the Ph-chromosome [40,41,42,43,44,45,49,50] and BCR-ABL1 KD mutations [51], are observed in up to 90% and 80% of BC patients, respectively. Additionally, somatic mutations are reported in BC and are associated with poor-risk disease [44,45]. Mutated genes include RUNX1, ASXL1, and IKZF1. High-risk ACAs comprise the most frequently observed ACAs in BC, that is, +8, +Ph, i(17q), +19, +21, +17 (major route ACA), some minor route ACAs (-7/7q-, 3q26.2, and 11q23 rearrangements), and complex aberrant karyotypes [49,50]. High-risk ACAs at low blast counts herald death by CML [40]. The emergence of high-risk ACAs, rather than blast increase, may signal the appropriate time for a change of therapy.
Accelerated phase (AP) should be treated as a high-risk disease. Table 3 shows updates of the current options for managing BC [10,52].
Some form of remission or a return to a second CP (CP2) is recommended prior to transplantation. Transplantation in BC is not advised.

7. Treatment Discontinuation and Treatment-Free Remission

Treatment discontinuation in stable DMR, for achieving TFR and possibly cure, is a new goal of CML treatment. DMR is defined as a molecular response of MR4 or deeper. In DMR, progression is extremely rare [16]. Benchmark times, when DMR can be expected, have been determined in long-term clinical TKI trials and are summarized in Table 4 [2,16,21,22,27].
Since the initial reports by Rousselot in 2007 [56] and Mahon in 2010 [57], numerous reports on treatment discontinuation have followed. Table 5 lists a selection of 21 studies that include close to 3000 patients. Relapse-free remission at two years ranges around 50% (33–72% at 0.5–10 years).
Durations of DMR and of TKI treatment have been determined as the most important predictors of successful TFR. Low-risk disease and treatment with IFN may also be beneficial [57,80]. EURO-SKI, which with 755 patients is the largest study, reports a TFR rate of 49% at two years [58]. Longer durations of treatment and of DMR correlate with higher TFR rates. The DESTINY Study from the UK indicates that TFR may be more successful if TKI dosage is first reduced before being completely stopped [77]. It is noteworthy that treatment discontinuation in routine care shows similar, if not higher, TFR rates [78], Richter, ESH 2020. This has been associated with longer treatment duration outside clinical trials and more frequent use of 2G-TKIs. However, due to their frequent and serious long-term toxicity, 2G-TKIs are not generally recommended by the ELN for the faster achievement of DMR. Exemptions might be younger patients with low- or intermediate-risk disease, patients for whom TFR is a high priority, and women who wish to become pregnant.
Loss of MMR indicates failure after TFR [61]. After the restart of treatment, about 95% of patients regain pre-discontinuation levels of response. Sources of relapse are thought to be residual BCR-ABL transcripts and the persistence of BCR-ABL1 in the genome [60,81]. In a genomic analysis of 42 patients in DMR, those with a negative PCR in both RNA and DNA had a stable TFR rate of 80–100%, compared to 57–67% in those with a negative PCR in RNA and a positive PCR in DNA, and 20% in those with a positive PCR in both RNA and DNA [81].
Most relapses occur early within the first 6 months, but late relapses have been reported in 15% of cases up to 6 years after discontinuation [78].
After relapse, a second treatment discontinuation can be tried, but the success rate is lower [79].
Mandatory, minimal, and optimal requirements for successful treatment discontinuation have been proposed by the European LeukemiaNet [10] (Table 6).

8. Pregnancy

All TKIs are potentially teratogenic and should not be given during pregnancy [82,83]. Since TKIs may be secreted in breast milk, TKI treatment during breast feeding is not recommended [84]. Sperm quality has been found to be unchanged after treatment with TKIs [85].

9. Conclusions

Since currently only 15–20% of patients reach stable TFRs, and most patients are at risk of life-long TKI treatment, the amelioration of chronic low-grade side effects is important. The projection for the next 5 years is that more patients will reach TFR through longer treatment duration or treatment with newer more efficacious drugs at a good quality of life, that less patients progress to BC, and that we can address a cure of CML more definitely in a cost-effective manner.

Funding

This research received no external funding.

Acknowledgments

The IT support by J. Hehlmann, MA, is acknowledged.

Conflicts of Interest

The author declares no conflict of interest

References

  1. Sasaki, K.; Strom, S.S.; O’Brien, S.; Jabbour, E.; Ravandi, F.; Konopleva, M.; Borthakur, G.; Pemmaraju, N.; Daver, N.; Jain, P.; et al. Relative survival in patients with chronic-phase chronic myeloid leukaemia in the tyrosine-kinase inhibitor era: Analysis of patient data from six prospective clinical trials. Lancet Haematol. 2015, 2, e186–e193. [Google Scholar] [CrossRef] [Green Version]
  2. Hehlmann, R.; Lauseker, M.; Saußele, S.; Pfirrmann, M.; Krause, S.; Kolb, H.J.; Neubauer, A.; Hossfeld, D.K.; Nerl, C.; Gratwohl, A.; 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, 2398–2406. [Google Scholar] [CrossRef] [PubMed]
  3. Thielen, N.; Visser, O.; Ossenkoppele, G.J.; Janssen, J. Chronic myeloid leukemia in the Netherlands: A population-based study on incidence, treatment, and survival in 3585 patients from 1989 to 2012. Eur. J. Haematol. 2015, 97, 145–154. [Google Scholar] [CrossRef] [PubMed]
  4. Bower, H.; Björkholm, M.; Dickman, P.W.; Höglund, M.; Lambert, P.C.; Andersson, T.M.-L. Life expectancy of patients with chronic myeloid leukemia approaches the life expectancy of the general population. J. Clin. Oncol. 2016, 34, 2851–2857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Welch, H.G.; Kramer, B.S.; Black, W.C. Epidemiologic signatures in cancer. N. Engl. J. Med. 2019, 381, 1378–1386. [Google Scholar] [CrossRef] [PubMed]
  6. Baccarani, M.; Saglio, G.; Goldman, J.; Hochhaus, A.; Simonsson, B.; Appelbaum, F.; Apperley, J.; Cervantes, F.; Cortes, J.; Deininger, M.; et al. Evolving concepts in the management of chronic myeloid leukemia: Recommendations from an expert panel on behalf of the European LeukemiaNet. Blood 2006, 108, 1809–1820. [Google Scholar] [CrossRef] [Green Version]
  7. Baccarani, M.; Cortes, J.; Pane, F.; Niederwieser, D.; Saglio, G.; Apperley, J.; Cervantes, F.; Deininger, M.; Gratwohl, A.; Guilhot, F.; et al. Chronic myeloid leukemia: An update of concepts and management recommendations of European LeukemiaNet. J. Clin. Oncol. 2009, 27, 6041–6051. [Google Scholar] [CrossRef] [Green Version]
  8. Baccarani, M.; Deininger, M.W.; Rosti, G.; Hochhaus, A.; Soverini, S.; Apperley, J.F.; Cervantes, F.; Clark, R.E.; Cortes, J.E.; Guilhot, F.; et al. European LeukemiaNet recommendations for the management of chronic myeloid leukemia: 2013. Blood 2013, 122, 872–884. [Google Scholar] [CrossRef]
  9. Hehlmann, R. Chronic myeloid leukemia in 2020. HemaSphere 2020, 4, e468. [Google Scholar] [CrossRef]
  10. Hochhaus, A.; Baccarani, M.; Silver, R.T.; Schiffer, C.; Apperley, J.F.; Cervantes, F.; Clark, R.E.; Cortes, J.E.; Deininger, M.W.; Guilhot, F.; et al. European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia 2020, 34, 966–984. [Google Scholar] [CrossRef] [Green Version]
  11. Pfirrmann, M.; Baccarani, M.; Saussele, S.; Guilhot, J.; Cervantes, F.; Ossenkoppele, G.; Hoffmann, V.S.; Castagnetti, F.; Hasford, J.; Hehlmann, R.; et al. Prognosis of long-term survival considering disease-specific death in patients with chronic myeloid leukemia. Leukemia 2016, 30, 48–56. [Google Scholar] [CrossRef] [PubMed]
  12. Pfirrmann, M.; Clark, R.E.; Prejzner, W.; Lauseker, M.; Baccarani, M.; Saussele, S.; Guilhot, F.; Heibl, S.; Hehlmann, R.; Faber, E.; et al. The EUTOS long-term survival (ELTS) score is superior to the Sokal score for predicting survival in chronic myeloid leukemia. Leukemia 2020, 34, 2138–2149. [Google Scholar] [CrossRef] [PubMed]
  13. Cross, N.C.P.; White, H.E.; Muller, M.C.; Saglio, G.; Hochhaus, A. Standardized definitions of molecular response in chronic myeloid leukemia. Leukemia 2012, 26, 2172–2175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Cross, N.C.P.; White, H.E.; Colomer, D.; Ehrencrona, H.; Foroni, L.; Gottardi, E.; Lange, T.; Lion, T.; Polakova, K.M.; Dulucq, S.; et al. Laboratory recommendations for scoring deep molecular responses following treatment for chronic myeloid leukemia. Leukemia 2015, 29, 999–1003. [Google Scholar] [CrossRef] [Green Version]
  15. Hehlmann, R.; Lauseker, M.; Jung-Munkwitz, S.; Leitner, A.; Müller, M.C.; Pletsch, N.; Proetel, U.; Haferlach, C.; Schlegelberger, B.; Balleisen, L.; et al. Tolerability-adapted imatinib 800 mg/d versus 400 mg/d versus 400 mg/d plus interferon-α in newly diagnosed chronic myeloid leukemia. J. Clin. Oncol. 2011, 29, 1634–1642. [Google Scholar] [CrossRef]
  16. Hehlmann, R.; Müller, M.C.; Lauseker, M.; Hanfstein, B.; Fabarius, A.; Schreiber, A.; Proetel, U.; Pletsch, N.; Pfirrmann, M.; Haferlach, C.; 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, 415–423. [Google Scholar] [CrossRef]
  17. Hochhaus, A.; Larson, R.A.; Guilhot, F.; Radich, J.P.; Branford, S.; Hughes, T.P.; Baccarani, M.; Deininger, M.W.; Cervantes, F.; Fujihara, S.; et al. Long-term outcomes of imatinib treatment for CML. N. Engl. J. Med. 2017, 376, 917–927. [Google Scholar] [CrossRef]
  18. Lauseker, M.; Hanfstein, B.; Haferlach, C.; Schnittger, S.; Pfirrmann, M.; Fabarius, A. 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, 1965–1969. [Google Scholar] [CrossRef]
  19. Kalmanti, L.; Saussele, S.; Lauseker, M.; Müller, M.C.; Dietz, C.T.; Heinrich, L. Safety and efficacy of imatinib in CML over a period of 10 years: Data from the randomized CML-study IV. Leukemia 2015, 29, 1123–1132. [Google Scholar] [CrossRef]
  20. Steegmann, J.L.; Baccarani, M.; Breccia, M.; Casado, L.F.; García-Gutiérrez, V.; Hochhaus, A.; Kim, D.-W.; Kim, T.D.; Khoury, H.J.; Le Coutre, P.; et al. European LeukemiaNet recommendations for the management and avoidance of adverse events of treatment in chronic myeloid leukaemia. Leukemia 2016, 30, 1648–1671. [Google Scholar] [CrossRef] [Green Version]
  21. Cortes, J.E.; Saglio, G.; Kantarjian, H.M.; Baccarani, M.; Mayer, J.; Boqué, C.; Shah, N.P.; Chuah, C.; Casanova, L.; Bradley-Garelik, B.; et al. Final 5-year study results of DASISION: The dasatinib versus imatinib study in treatment-naïve chronic myeloid leukemia patients trial. J. Clin. Oncol. 2016, 34, 2333–2340. [Google Scholar] [CrossRef] [PubMed]
  22. Hochhaus, A.; Saglio, G.; Hughes, T.P.; Larson, A.R.; Kim, D.-W.; Issaragrisil, S.; Le Coutre, P.D.; Etienne, G.; Dorlhiac-Llacer, E.P.; Clark, E.R.; et al. Long-term benefits and risks of frontline nilotinib vs. imatinib for chronic myeloid leukemia in chronic phase: 5-year update of the randomized ENESTnd trial. Leukemia 2016, 30, 1044–1054. [Google Scholar] [CrossRef] [PubMed]
  23. Padula, W.V.; Larson, R.A.; Dusetzina, S.B.; Apperley, J.F.; Hehlmann, R.; Baccarani, M.; Eigendorff, E.; Guilhot, J.; Guilhot, F.; Mahon, F.-X.; et al. Cost-effectiveness of tyrosine kinase inhibitor treatment strategies for chronic myeloid leukemia in chronic phase after generic entry of imatinib in the united states. J. Natl. Cancer Inst. 2016, 108, 003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Shih, Y.-C.T.; Cortes, J.E.; Kantarjian, H.M. Treatment value of second-generation BCR-ABL1 tyrosine kinase inhibitors compared with imatinib to achieve treatment-free remission in patients with chronic myeloid leukaemia: A modelling study. Lancet Haematol. 2019, 6, e398–e408. [Google Scholar] [CrossRef]
  25. Yamamoto, C.; Nakashima, H.; Ikeda, T.; Kawaguchi, S.-I.; Toda, Y.; Ito, S.; Mashima, K.; Nagayama, T.; Umino, K.; Minakata, D.; et al. Analysis of the cost-effectiveness of treatment strategies for CML with incorporation of treatment discontinuation. Blood Adv. 2019, 3, 3266–3277. [Google Scholar] [CrossRef] [Green Version]
  26. Naqvi, K.; Jabbour, E.; Skinner, J.; Bs, K.A.; Bs, S.D.; Yilmaz, M.; Ferrajoli, A.; Bose, P.; Thompson, P.; Alvarado, Y.; et al. Long-term follow-up of lower dose dasatinib (50 mg daily) as frontline therapy in newly diagnosed chronic-phase chronic myeloid leukemia. Cancer 2020, 126, 67–75. [Google Scholar] [CrossRef] [Green Version]
  27. Hughes, T.P.; Saglio, G.; Larson, R.A.; Kantarjian, H.M.; Kim, D.N.-W.; Issaragrisil, S.; Le Coutre, P.; Etienne, G.; Boquimpani, C.; Clark, R.E.; et al. Long-term outcomes in patients with chronic myeloid leukemia in chronic phase receiving frontline nilotinib versus imatinib: Enestnd 10-year analysis. Blood 2019, 134, 2924. [Google Scholar] [CrossRef]
  28. Saußele, S.; Krauß, M.-P.; Hehlmann, R.; Lauseker, M.; Proetel, U.; Kalmanti, L.; Hanfstein, B.; Fabarius, A.; Kraemer, D.; Berdel, W.E.; 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. [Google Scholar] [CrossRef] [Green Version]
  29. Cortes, J.E.; Gambacorti-Passerini, C.; Deininger, M.W.; Mauro, M.J.; Chuah, C.; Kim, D.-W.; Dyagil, I.; Glushko, N.; Milojkovic, D.; Le Coutre, P.; et al. Bosutinib versus imatinib for newly diagnosed chronic myeloid leukemia: Results from the randomized BFORE trial. J. Clin. Oncol. 2018, 36, 231–237. [Google Scholar] [CrossRef]
  30. Soverini, S.; Bavaro, L.; De Benedittis, C.; Martelli, M.; Iurlo, A.; Orofino, N.; Sica, S.; Sora, F.; Lunghi, F.; Ciceri, F.; et al. Prospective assessment of NGS-detectable mutations in CML patients with non-optimal response: The NEXT-in-CML study. Blood J. Am. Soc. Hematol. 2020, 135, 534–541. [Google Scholar]
  31. Kizilors, A.; Crisà, E.; Lea, N.; Passera, R.; Mian, S.; Anwar, J.; Best, S.; Nicolini, F.E.; Ireland, R.; Aldouri, M.; et al. Effect of low-level BCR-ABL1 kinase domain mutations identified by next-generation sequencing in patients with chronic myeloid leukaemia: A population-based study. Lancet Haematol. 2019, 6, e276–e284. [Google Scholar] [CrossRef]
  32. Cortes, J.E.; Kim, D.-W.; Pinilla-Ibarz, J.L.; Le Coutre, P.; Paquette, R.; Chuah, C.; Nicolini, F.; Apperley, J.; Khoury, H.; Talpaz, M.; et al. A phase 2 trial of ponatinib in philadelphia chromosome–positive leukemias. N. Engl. J. Med. 2013, 369, 1783–1796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Cortes, J.; Lomaia, E.; Turkina, A.; Moiraghi, B.; Sutton, U.M.; Pavlovsky, C.; Kim, D.W. Interim analysis from the OPTIC trial-A dose-ranging study of 3 starting doses of ponatinib. Clin. Lymphoma Myeloma Leuk. 2020, 20, S234. [Google Scholar]
  34. Lichtin, A.E.; Woolf, S.H.; Silver, R.T.; Hehlmann, R. Chronic myeloid leukemia—ASH practice guideline and beyond. Hematol. Am. Soc. Hematol. Educ. Prog. 1998, 435–453. [Google Scholar]
  35. Gratwohl, A.; Pfirrmann, M.; Zander, A.; Kröger, N.; Beelen, D.; Novotny, J.; Nerl, C.; Scheid, C.; Spiekermann, K.; Mayer, J. Long-term outcome of patients with newly diagnosed chronic myeloid leukemia: A randomized comparison of stem cell transplantation with drug treatment. Leukemia 2016, 30, 562–569. [Google Scholar] [CrossRef] [Green Version]
  36. Saussele, S.; Lauseker, M.; Gratwohl, A.; Beelen, D.W.; Bunjes, D.; Schwerdtfeger, R.; Kolb, H.-J.; Ho, A.D.; Falge, C.; Holler, E.; et al. Allogeneic hematopoietic stem cell transplantation (allo SCT) for chronic myeloid leukemia in the imatinib era: Evaluation of its impact within a subgroup of the randomized German CML Study IV. Blood 2010, 115, 1880–1885. [Google Scholar] [CrossRef]
  37. Saussele, S.; Lauseker, M.; Müller, M.C.; Gratwohl, A.; Beelen, D.; Bunjes, D.W.; Schwerdtfeger, R.; Kolb, H.-J.; Ho, A.D.; Falge, C.; et al. Allogeneic hematopoietic stem cell transplantation (hsct) in the imatinib-era: Update on the survival outcome following allogeneic HSCT after imatinib failure; results of the german CML study IV. Blood 2014, 124, 2567. [Google Scholar] [CrossRef]
  38. Jiang, H.; Xu, L.-P.; Liu, D.-H.; Liu, K.-Y.; Chen, S.-S.; Jiang, B.; Jiang, Q.; Chen, H.; Chen, Y.-H.; Han, W.; et al. Allogeneic hematopoietic SCT in combination with tyrosine kinase inhibitor treatment compared with TKI treatment alone in CML blast crisis. Bone Marrow Transplant. 2014, 49, 1146–1154. [Google Scholar] [CrossRef] [Green Version]
  39. Oyekunle, A.; Zander, A.R.; Binder, M.; Ayuk, F.; Zabelina, T.; Christopeit, M.; Stübig, T.; Alchalby, H.; Schafhausen, P.; Lellek, H.; et al. Outcome of allogeneic SCT in patients with chronic myeloid leukemia in the era of tyrosine kinase inhibitor therapy. Ann. Hematol. 2013, 92, 487–496. [Google Scholar] [CrossRef]
  40. Hehlmann, R.; Voskanyan, A.; Lauseker, M.; Pfirrmann, M.; Kalmanti, L.; Rinaldetti, S.; Kohlbrenner, K.; Haferlach, C.; Schlegelberger, B.; Fabarius, A.; et al. High-risk additional chromosomal abnormalities at low blast counts herald death by CML. Leukemia 2020, 34, 2074–2086. [Google Scholar] [CrossRef]
  41. Fabarius, A.; Kalmanti, L.; Dietz, C.T.; Lauseker, M.; Rinaldetti, S.; Haferlach, C.; Göhring, G.; Schlegelberger, B.; Jotterand, M.; Hanfstein, B.; et al. Impact of unbalanced minor route versus major route karyotypes at diagnosis on prognosis of CML. Ann. Hematol. 2015, 94, 2015–2024. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, W.; Cortes, J.E.; Tang, G.; Khoury, J.D.; Wang, S.; Bueso-Ramos, C.E.; DiGiuseppe, J.A.; Chen, Z.; Kantarjian, H.M.; Medeiros, L.J.; et al. Risk stratification of chromosomal abnormalities in chronic myelogenous leukemia in the era of tyrosine kinase inhibitor therapy. Blood 2016, 127, 2742–2750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Gong, Z.; Medeiros, L.J.; Cortes, J.E.; Chen, Z.; Zheng, L.; Li, Y.; Bai, S.; Lin, P.; Miranda, R.N.; Jorgensen, J.L.; et al. Cytogenetics-based risk prediction of blastic transformation of chronic myeloid leukemia in the era of TKI therapy. Blood Adv. 2017, 1, 2541–2552. [Google Scholar] [CrossRef] [PubMed]
  44. Grossmann, V.; Kohlmann, A.; Zenger, M.; Schindela, S.; Eder, C.; Weissmann, S.; Schnittger, S.; Kern, W.; Muller, M.C.; Hochhaus, A.; et al. A deep-sequencing study of chronic myeloid leukemia patients in blast crisis (BC-CML) detects mutations in 76.9% of cases. Leukemia 2011, 25, 557–560. [Google Scholar] [CrossRef]
  45. Branford, S.; Wang, P.; Yeung, D.T.; Thomson, D.; Purins, A.; Wadham, C.; Shahrin, N.H.; Marum, J.E.; Nataren, N.; Parker, W.T.; et al. Integrative genomic analysis reveals cancer-associated mutations at diagnosis of CML in patients with high-risk disease. Blood 2018, 132, 948–961. [Google Scholar] [CrossRef]
  46. Koptyra, M.; Falinski, R.; Nowicki, M.O.; Stoklosa, T.; Majsterek, I.; Nieborowska-Skorska, M.; Blasiak, J.; Skorski, T. BCR/ABL kinase induces self-mutagenesis via reactive oxygen species to encode imatinib resistance. Blood 2006, 108, 319–327. [Google Scholar] [CrossRef] [Green Version]
  47. Mohty, M.; Yong, A.S.M.; Szydlo, R.; Apperley, J.F.; Melo, J.V. The polycomb group BMI1 gene is a molecular marker for predicting prognosis of chronic myeloid leukemia. Blood 2007, 110, 380–383. [Google Scholar] [CrossRef] [Green Version]
  48. Ko, T.K.; Javed, A.; Lee, K.L.; Pathiraja, T.N.; Liu, X.; Malik, S.; Soh, S.X.; Heng, X.T.; Takahashi, N.; Tan, J.H.J.; et al. An integrative model of pathway convergence in genetically heterogeneous blast crisis chronic myeloid leukemia. Blood 2020, 135, 2337–2353. [Google Scholar] [CrossRef]
  49. Johansson, B.; Fioretos, T.; Mitelman, F. Cytogenetic and molecular genetic evolution of chronic myeloid leukemia. Acta Haematol. 2002, 107, 76–94. [Google Scholar] [CrossRef]
  50. Mitelman, F.; LeVan, G.; Nilsson, P.G.; Brandt, L. Non-random karyotypic evolution in chronic myeloid leukemia. Int. J. Cancer 1976, 18, 24–30. [Google Scholar] [CrossRef]
  51. Soverini, S.; Hochhaus, A.; Nicolini, F.E.; Gruber, F.; Lange, T.; Saglio, G.; Pane, F.; Müller, M.C.; Ernst, T.; Rosti, G.; et al. BCR-ABL kinase domain mutation analysis in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors: Recommendations from an expert panel on behalf of European LeukemiaNet. Blood 2011, 118, 1208–1215. [Google Scholar] [CrossRef] [PubMed]
  52. Hehlmann, R. How I treat CML blast crisis. Blood 2012, 120, 737–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Milojkovic, D.; Ibrahim, A.; Reid, A.; Foroni, L.; Apperley, J.; Marin, D. Efficacy of combining dasatinib and FLAG-IDA for patients with chronic myeloid leukemia in blastic transformation. Haematologica 2011, 97, 473–474. [Google Scholar] [CrossRef] [Green Version]
  54. Copland, M.; Slade, D.; Byrne, J.; Brock, K.; De Lacallade, H.; Craddock, C.; Clark, R.; Smith, M.; Bishiop, R.; Milojkovic, D.; et al. FLAG-IDA and ponatinib in patients with blast phase chronic myeloid leukemia: Results from the phase I/II UK trials acceleration program matchpoint trial. Blood 2017, 134, 497. [Google Scholar] [CrossRef]
  55. Strati, P.; Kantarjian, H.; Thomas, D.; O’Brien, S.; Konoplev, S.; Jorgensen, J.L.; Luthra, R.; Abruzzo, L.; Jabbour, E.; Quintas-Cardama, A.; et al. HCVAD plus imatinib or dasatinib in lymphoid blastic phase chronic myeloid leukemia. Cancer 2014, 120, 373–380. [Google Scholar] [CrossRef] [PubMed]
  56. Rousselot, P.; Huguet, F.; Rea, D.; Legros, L.; Cayuela, J.-M.; Maarek, O.; Blanchet, O.; Marit, G.; Gluckman, E.; Reiffers, J.; et al. Imatinib mesylate discontinuation in patients with chronic myelogenous leukemia in complete molecular remission for more than 2 years. Blood 2007, 109, 58–60. [Google Scholar] [CrossRef]
  57. Mahon, F.X.; Réa, D.; Guilhot, J.; Guilhot, F.; Huguet, F.; Nicolini, F.; Lgros, L.; Charbonnier, A.; Guerci, A.; Varet, B.; et al. Discontinuation of imatinib in patients with chronic myeloid leukemia who have maintained complete molecular remission for at least 2 years: The prospective, multicenter Stop Imatinib (STIM) trial. Lancet. Oncol. 2010, 11, 1029–1035. [Google Scholar] [CrossRef]
  58. Saussele, S.; Richter, J.; Guilhot, J.; Gruber, F.X.; Hjorth-Hansen, H.; Almeida, A.; Janssen, J.J.W.M.; Mayer, P.J.; Koskenvesa, P.; Panayiotidis, P.; et al. Discontinuation of tyrosine kinase inhibitor therapy in chronic myeloid leukemia (EURO-SKI): A prespecified interim analysis of a prospective, multicenter, non-randomized, trial. Lancet Oncol. 2018, 19, 747–757. [Google Scholar] [CrossRef] [Green Version]
  59. Etienne, G.; Guilhot, J.; Rea, D.; Rigal-Huguet, F.; Nicolini, F.; Charbonnier, A.; Guerci-Bresler, A.; Legros, L.; Varet, B.; Gardembas, M.; et al. Long-term follow-up of the french stop imatinib (STIM1) study in patients with chronic myeloid leukemia. J. Clin. Oncol. 2017, 35, 298–305. [Google Scholar] [CrossRef] [Green Version]
  60. Ross, D.M.; Branford, S.; Seymour, J.F.; Schwarer, A.P.; Arthur, C.; Yeung, D.T.; Dang, P.; Goyne, J.M.; Slader, C.; Filshie, R.J.; 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, 515–522. [Google Scholar] [CrossRef]
  61. Rousselot, P.; Charbonnier, A.; Cony-Makhoul, P.; Agape, P.; Nicolini, F.E.; Varet, B.; Gardembas, M.; Etienne, G.; Réa, D.; Roy, L.; et al. Loss of major molecular response as a trigger for restarting tyrosine kinase inhibitor therapy in patients with chronic-phase chronic myelogenous leukemia who have stopped imatinib after durable undetectable disease. J. Clin. Oncol. 2014, 32, 424–430. [Google Scholar] [CrossRef] [PubMed]
  62. Lee, S.-E.; Choi, S.Y.; Song, H.-Y.; Kim, S.-H.; Choi, M.-Y.; Park, J.S.; Kim, H.-J.; Zang, D.Y.; Oh, S.; Do, Y.R.; et al. Imatinib withdrawal syndrome and longer duration of imatinib have a close association with a lower molecular relapse after treatment discontinuation: The KID study. Haematologica 2016, 101, 717–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Zang, D.Y.; Lee, W.S.; Mun, Y.-C.; Do, Y.R.; Oh, S.; Lee, S.-E.; Choi, S.Y.; Kim, D.-W. Long-term follow-up after treatment discontinuation in patients with chronic myeloid leukemia: The korean imatinib discontinuation (KID) study. Blood 2018, 132, 4252. [Google Scholar] [CrossRef]
  64. Nicolini, F.E.; Dulucq, S.; Guilhot, J.; Etienne, G.; Mahon, F.-X. The evaluation of residual disease by digital PCR, and TKI duration are critical predictive factors for molecular recurrence after for stopping imatinib first-line in chronic phase CML patients: Results of the STIM2 study. Blood 2018, 132, 462. [Google Scholar] [CrossRef]
  65. Mori, S.; Vagge, E.; Le Coutre, P.; Abruzzese, E.; Martino, B.; Pungolino, E.; Elena, C.; Pierri, I.; Assouline, S.; D’Emilio, A.; et al. Age and dPCR can predict relapse in CML patients who discontinued imatinib: The ISAV study. Am. J. Hematol. 2015, 90, 910–914. [Google Scholar] [CrossRef]
  66. Mori, S.; Le Coutre, P.; Abruzzese, E.; Martino, B.; Pungolino, E.; Elena, C.; Bergamaschi, M.; Assouline, S.; Di Bona, E.; Gozzini, A.; et al. Imatinib suspension and validation (ISAV) study: Final results at 79 months. Blood 2018, 132, 461. [Google Scholar] [CrossRef]
  67. Rea, D.; Nicolini, F.E.; Tulliez, M.; Guilhot, F.; Guilhot, J.; Guerci-Bresler, A.; Gardembas, M.; Coiteux, V.; Guillerm, G.; Legros, L.; et al. Discontinuation of dasatinib or nilotinib in chronic myeloid leukemia: Interim analysis of the STOP 2G-TKI study. Blood 2017, 129, 846–854. [Google Scholar] [CrossRef] [Green Version]
  68. Imagawa, J.; Tanaka, H.; Okada, M.; Nakamae, H.; Hino, P.M.; Murai, K.; Ishida, P.Y.; Kumagai, T.; Sato, S.; Ohashi, K.; et al. Discontinuation of dasatinib in patients with chronic myeloid leukemia who have maintained deep molecular response for longer than 1 year (DADI trial): A multicenter phase 2 trial. Lancet Haematol. 2015, 2, e528–e535. [Google Scholar] [CrossRef]
  69. Kadowaki, N.; Kawaguchi, T.; Kuroda, J.; Nakamae, H.; Matsumura, I.; Miyamoto, T.; Ishikawa, J.; Nagafuji, K.; Imamura, Y.; Yamazaki, H.; et al. Discontinuation of nilotinib in patients with chronic myeloid leukemia who have maintained deep molecular responses for at least 2 years: A multicenter phase 2 stop nilotinib (nilst) trial. Blood 2016, 128, 790. [Google Scholar] [CrossRef]
  70. Kim, D.D.H.; Bence-Bruckler, F.I.; Forrest, D.L.; Savoie, M.L.; Couban, S.; Busque, P.F.L.; Delage, M.R.; Laneuville, F.P.; Liew, E.; Xenocostas, A.; et al. Treatment-free remission accomplished by dasatinib (TRAD): Preliminary results of the pan-canadian tyrosine kinase inhibitor discontinuation trial. Blood 2016, 128, 1922. [Google Scholar] [CrossRef]
  71. Shah, N.; García-Gutiérrez, J.V.; Jiménez-Velasco, A.; Larson, S.; Saussele, S.; Rea, D.; Mahon, F.-X.; Levy, M.; Gómez-Casares, M.; Luciano, L.; et al. PF408 dasfree 2-year update: Dasatinib discontinuation in patients (PTS) with chronic myeloid leukemia in chronic phase (CML-CP) and deep molecular response (DMR). HemaSphere 2019, 3, 156. [Google Scholar] [CrossRef]
  72. Hughes, F.T.P.; Boquimpani, C.M.; Takahashi, N.; Benyamini, N.; Clementino, N.C.D.; Shuvaev, V.; Ailawadhi, S.; Lipton, J.H.; Turkina, A.G.; Moiraghi, E.B.; et al. Treatment-free remission in patients with chronic myeloid leukemia in chronic phase according to reasons for switching from imatinib to nilotinib: Subgroup analysis from ENESTop. Blood 2016, 128, 792. [Google Scholar] [CrossRef]
  73. Takahashi, N.; Nishiwaki, K.; Nakaseko, C.; Aotsuka, N.; Sano, K.; Ohwada, C.; Kuroki, J.; Kimura, H.; Tokuhira, M.; Mitani, K.; et al. Treatment-free remission after two-year consolidation therapy with nilotinib in patients with chronic myeloid leukemia: STAT2 trial in Japan. Haematologica 2018, 103, 1835–1842. [Google Scholar] [CrossRef] [PubMed]
  74. Hochhaus, A.; Masszi, T.; Giles, F.J.; Radich, J.P.; Ross, D.M.; Casares, M.T.G.; Hellmann, A.; Stentoft, J.; Conneally, E.; García-Gutiérrez, V.; et al. Treatment-free remission following frontline nilotinib in patients with chronic myeloid leukemia in chronic phase: Results from the ENESTfreedom study. Leukemia 2017, 31, 1525–1531. [Google Scholar] [CrossRef] [PubMed]
  75. Kumagai, T.; Nakaseko, C.; Nishiwaki, K.; Yoshida, C.; Ohashi, K.; Takezako, N.; Takano, H.; Kouzai, Y.; Murase, T.; Matsue, K.; et al. Discontinuation of dasatinib after deep molecular response for over 2 years in patients with chronic myelogenous leukemia and the unique profiles of lymphocyte subsets for successful discontinuation: A prospective, multicenter Japanese trial (D-STOP Trial). Blood 2016, 128, 791. [Google Scholar] [CrossRef]
  76. Hernández-Boluda, J.C.; Pereira, A.; Pastor-Galán, I.; Alvarez-Larrán, A.; Savchuk, A.; Puerta, J.M.; Sánchez-Pina, J.M.; Collado, R.; Díaz-González, A.; Angona, A.; et al. Feasibility of treatment discontinuation in chronic myeloid leukemia in clinical practice: Results from a nationwide series of 236 patients. Blood Cancer J. 2018, 8, 1–8. [Google Scholar] [CrossRef] [Green Version]
  77. Clark, R.E.; Polydoros, F.; Apperley, J.F.; Milojkovic, D.; Rothwell, K.; Pocock, C. Initial reduction of therapy prior to complete treatment discontinuation in chronic myeloid leukaemia: Final results of the British DESTINY Study. Lancet Haematol. 2019, 6, e375–e383. [Google Scholar] [CrossRef] [Green Version]
  78. Rousselot, P.; Loiseau, C.; Delord, M.; Cayuela, J.M.; Spentchian, M. Late molecular recurrences in patients with chronic myeloid leukemia experiencing treatment-free remission. Blood Adv. 2020, 4, 3034–3040. [Google Scholar] [CrossRef]
  79. Legros, L.; Nicolini, F.E.; Etienne, G.; Rousselot, P.; Rea, D.; Giraudier, S.; Guerci-Bresler, A.; Huguet, F.; Gardembas, M.; Escoffre, M.; et al. Second tyrosine kinase inhibitor discontinuation attempt in patients with chronic myeloid leukemia. Cancer 2017, 123, 4403–4410. [Google Scholar] [CrossRef] [Green Version]
  80. Talpaz, M.; Hehlmann, R.; Quintás-Cardama, A.; Mercer, J.; Cortes, J. Re-emergence of interferon-α in the treatment of chronic myeloid leukemia. Leukemia 2012, 27, 803–812. [Google Scholar] [CrossRef] [Green Version]
  81. Machova-Polakova, K.; Zizkova, H.; Zuna, J.; Motlova, E.; Hovorkova, L.; Gottschalk, A.; Glauche, I.; Koblihova, J.; Pecherkova, P.; Klamova, H.; et al. Analysis of chronic myeloid leukaemia during deep molecular response by genomic PCR: A traffic light stratification model with impact on treatment-free remission. Leukemia 2020. [Google Scholar] [CrossRef] [PubMed]
  82. Abruzzese, E.; Trawinska, M.M.; De Fabritiis, P.; Perrotti, A.P. Tyrosine kinase inhibitors and pregnancy. Mediterr. J. Hematol. Infect. Dis. 2014, 6, e2014028. [Google Scholar] [CrossRef] [PubMed]
  83. Cortes, J.E.; Abruzzese, E.; Chelysheva, E.; Guha, M.; Wallis, N.; Apperley, J.F. The impact of dasatinib on pregnancy outcomes. Am. J. Hematol. 2015, 90, 1111–1115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Chelysheva, E.; Aleshin, S.; Polushkina, E.; Shmakov, R.; Shokhin, I.; Chilov, G.; Turkina, A. Breastfeeding in patients with chronic myeloid leukemia: Case series with measurements of drug concentrations in maternal milk and literature review. Mediterr. J. Hematol. Infect. Dis. 2018, 10, e2018027. [Google Scholar] [CrossRef] [PubMed]
  85. Nicolini, F.-E.; Alcazer, V.; Huguet, F.; Cony-Makhoul, P.; Heiblig, M.; Fort, M.-P.; Morisset, S.; Guerci-Bresler, A.; Soula, V.; Sobh, M.; et al. CML patients show sperm alterations at diagnosis that are not improved with imatinib treatment. Leuk. Res. 2016, 48, 80–83. [Google Scholar] [CrossRef]
Figure 1. Survival after blast crisis (BC) with conventional (blue) versus tyrosine kinase inhibitor (TKI) therapy (red). German CML Study Group experience, updated 2020 (M.Lauseker). Ten-year survival was 19% (68 of 87 died) after TKI, and 3% after conventional therapy (678 of 699 died); 74% of living TKI-treated BC patients (14 of 19) and 71% of living chemotherapy-treated BC patients (15 of 21) had been transplanted.
Figure 1. Survival after blast crisis (BC) with conventional (blue) versus tyrosine kinase inhibitor (TKI) therapy (red). German CML Study Group experience, updated 2020 (M.Lauseker). Ten-year survival was 19% (68 of 87 died) after TKI, and 3% after conventional therapy (678 of 699 died); 74% of living TKI-treated BC patients (14 of 19) and 71% of living chemotherapy-treated BC patients (15 of 21) had been transplanted.
Jcm 09 03671 g001
Figure 2. Model of progression of CML from chronic phase (CP) to end-phase and BC. Suggested modulators are reactive oxygen species (ROS) [46] and the polycomb repressive complex (PRC) [48]. Elimination of BCR-ABL1 should stop, or reduce, progression to BC.
Figure 2. Model of progression of CML from chronic phase (CP) to end-phase and BC. Suggested modulators are reactive oxygen species (ROS) [46] and the polycomb repressive complex (PRC) [48]. Elimination of BCR-ABL1 should stop, or reduce, progression to BC.
Jcm 09 03671 g002
Table 1. Prognostic score at baseline and comparison of outcome with Sokal and ELTS scores [11,12].
Table 1. Prognostic score at baseline and comparison of outcome with Sokal and ELTS scores [11,12].
(A) ELTS score calculation.
CalculationDefinition of Risk Groups
0.0025 × (age/10)3
+ 0.0615 × spleen size
+ 0.1052 × peripheral blood blasts
+ 0.4104 × (platelet count/1000)−0.5
Low risk: < 1.5680
Intermediate risk: 1.5680–2.2185
High risk: > 2.2185
(B) Risk strata proportions and outcome.
Low RiskIntermediate RiskHigh Risk
n= 5154SokalELTSSokalELTSSokalELTS
%385538282313
10-year OS89%88%81%79%75%68%
6-year LRD3%2%4%5%8%12%
ELTS: EUTOS score for long-term survival, considering leukemia-related death (LRD) and overall survival (OS). Age is given in years; spleen size in cm below the costal margin and measured by palpation (maximum distance); blasts in percent of peripheral blood differential; platelet count as 109/L. All values are pre-treatment. To calculate Sokal and ELTS scores, go to http://www.leukemia-net.org/content/leukemias/cml/elts_score/index_eng.html.
Table 2. Milestones for treating chronic myeloid leukemia (CML) expressed as % BCR-ABL1 on the International Scale.
Table 2. Milestones for treating chronic myeloid leukemia (CML) expressed as % BCR-ABL1 on the International Scale.
OptimalWarningFailure
BaselineNAHigh-risk ACA, high-risk ELTS scoreNA
3 months≤10%>10%>10% if confirmed within 1–3 months
6 months≤1%>1–10%>10%
12 months≤0.1%>0.1–1%>1%
Any time≤0.1%>0.1%
loss of ≤ 0.1% (MMR) *
>1%, resistance mutations high-risk ACA
* Loss of MMR (BCR-ABL1 > 0.1%) indicates failure after TFR. NA: not applicable; ELTS: EUTOS score for long-term survival; ACAs: additional chromosomal aberrations; MMR: major molecular remission.
Table 3. BC management.
Table 3. BC management.
Prevention of BCEffective treatment, elimination of BCR-ABL1
High-risk ACAObserve closely, intensify treatment
APTreat as high-risk CML
Primary BCStart with imatinib
Assess for allo-SCT, initiate donor search
Resistance to 2G-TKIPonatinib
Failure to ponatinibEarly allo-SCT recommended
Progress to BCAttempt at return to CP2 *
For myeloid BC: AML regimens + TKI [53,54]
For lymphoid BC: ALL regimens + TKI [55]
After CP2: allo-SCT without delay
* CP2 = second chronic phase; AP = accelerated phase; SCT = stem cell transplantation; 2G-TKI = 2nd generation TKI; AML = acute myeloid leukemia; ALL = acute lymphoblastic leukemia.
Table 4. Benchmark times for DMR (MR4 and MR4.5) after 5 and 10 years of treatment with imatinib, nilotinib, and dasatinib.
Table 4. Benchmark times for DMR (MR4 and MR4.5) after 5 and 10 years of treatment with imatinib, nilotinib, and dasatinib.
Study 5 Years (%)10 Years (%)
CML study IV *Imatinib MR46881
Imatinib MR4.55372
ENESTnd **Nilotinib MR46673
Nilotinib MR4.55464
Imatinib MR44256
Imatinib MR4.53545
Dasision ***Dasatinib MR4.542NA
Imatinib MR4.533NA
* imatinib (n = 1442), ** nilotinib 300 mg twice daily (n = 282), imatinib 400 mg daily (n = 283), *** dasatinib 100 mg once daily (n = 259), imatinib 400 mg daily (n = 260). DMR rates of these trials cannot be directly compared, owing to different methods of trial evaluation. The larger difference between responses to imatinib and 2G-TKI in ENESTnd compared to Dasision correlates with a higher drop-out rate of imatinib-treated patients (50%) compared to nilotinib-treated patients (40%). NA: not available.
Table 5. Selected TKI-discontinuation studies.
Table 5. Selected TKI-discontinuation studies.
StudyTKIMin. Treatment Duration (Years)nDepth of MRMin. Duration of MR (Years)RFS with at Least MMRReferences
Euro-SKIIM3755MR4149% at 2 yearsSaußele et al., 2018 [58]
STIMIM2100MR5237% at 10 yearsEtienne et al., 2017 [59] Update at ESH 2019
TWISTERIM340MR4.5245% at 42 monthsRoss et al., 2013 [60]
A-STIMIM380UMRD264% at 23 monthsRousselot et al., 2014 [61]
KID studyIM3126MR4.5258% at 2 yearsLee et al., 2016 [62] Update Zang 2018 ASH a. 4252 [63]
STIM2IM2200MR4.5246% at 2 yearsNicolini et al., 2018 [64] ASH a. 462
ISAVIM2112UMRD1.552% at 22 monthsMori et al., 2015 [65] Update at ASH 2018 a. 461 [66]
STOP 2G-TKIDasa/Nilo260MR4.52ca. 55% at 4 yearsRea et al., 2017 [67]
DADIDasa 2nd lineND63MR4149% at 6 monthsImagawa et al., 2015 [68]
NILSTNilo287MR4.5259% at 1 yearKadowaki et al., 2016 ASH a. 790 [69]
TRADIM/Dasa375MR4.5258% at 6 monthsKim et al., 2016 ASH a. 1922 [70]
DasfreeDasa284MR4.5146% at 2 yearsShah et al., 2019 [71]
Update at ESH 2019
ENESTopNilo 2nd line3126MR4.5158% at 4 yearsHughes et al., 2016 ASH a. 792 [72]
STAT2IM/Nilo296MR4.5268% at 1 yearTakahashi et al., 2018 [73]
ENESTfreedomNilo2190MR4.5152% at 4 yearsHochhaus et al., 2017 [74]
D-STOPIM/DasaND54MR4263% at 1 yearKumagai et al., 2016 ASH a. 791 [75]
Spanish studyIM/Nilo/Dasa3236MR4.5264% at 4 yearsBoluda et al., 2018 ASH a. 47 [76]
DESTINYIM/Nilo/Dasa6.9 (median)125MR4372% at 3 yearsClark et al., 2019 [77]
Routine CareTKI7.1128MR4467% at 2.9 yearsRousselot et al., 2020 [78]
Swedish CML-RegistryTKI (53% IM)7.7131DMR2.9 (median)61% outside, 35% inside a study at 2 yearsRichter, ESH 2020
RE-STIM(2nd stop)3.1 (median)106MR4.51.7 (median)33% at 4 yearsLegros et al., 2017 [79] Update at EHA 2019
Total: 21 2974 33–72% at 0.5–10 years
Updated from Hehlmann 2020 [9].
Table 6. Requirements for tyrosine kinase inhibitor discontinuation.
Table 6. Requirements for tyrosine kinase inhibitor discontinuation.
MandatoryCML in first CP only (data are lacking outside this setting)
Motivated patient with structured communication
Access to high-quality quantitative PCR using the International Scale (IS), with rapid turn-around of PCR test results
Patient’s agreement to more frequent monitoring after stopping treatment.
This means monthly for the first 6 months, every 2 months for months 6–12, and every 3 months thereafter
Minimal (stop allowed)First-line therapy or second-line, if intolerance was the only reason for changing TKI
Typical e13a2 or e14a2 BCR-ABL1 transcripts
Duration of TKI therapy > 5 years (>4 years for 2G-TKI)
Duration of DMR (MR4 or better) > 2 years
No prior treatment failure
Optimal (stop recommended for consideration)Duration of TKI therapy > 5 years
Duration of DMR > 3 years if MR4
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hehlmann, R. The New ELN Recommendations for Treating CML. J. Clin. Med. 2020, 9, 3671. https://doi.org/10.3390/jcm9113671

AMA Style

Hehlmann R. The New ELN Recommendations for Treating CML. Journal of Clinical Medicine. 2020; 9(11):3671. https://doi.org/10.3390/jcm9113671

Chicago/Turabian Style

Hehlmann, Rüdiger. 2020. "The New ELN Recommendations for Treating CML" Journal of Clinical Medicine 9, no. 11: 3671. https://doi.org/10.3390/jcm9113671

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop