1,615
Views
1
CrossRef citations to date
0
Altmetric
Editorial

Will tyrosine kinase inhibitors be part of the treatment armamentarium for CML in the future?

&
Pages 79-81 | Received 04 Sep 2017, Accepted 21 Dec 2017, Published online: 07 Jan 2018

1. Introduction

The history of chronic myeloid leukemia (CML) therapy over the last 6 decades is one of the rapid changes and is fascinating. It started with supportive care, hydrea, busulfan, and radiation back in the 1960s [Citation1]. These therapies mainly controlled the manifestations of the disease with little, if any, effect on the disease itself. With those therapies, the median survival was 3 years, and almost all patients died by 7 years. Following that, interferon therapy was introduced in the 1980s. With interferon, a small percentage of patients achieved cytogenetic and molecular remission, but with significant toxicity and a short duration of response [Citation2,Citation3]. Allogeneic hematopoietic stem cell transplantation (HCT) for CML also was introduced in the 1980s [Citation4]. HCT demonstrated a survival benefit when compared to supportive care only and became the standard of care for eligible patients with CML. However, HCT is associated with significant morbidity, mortality, and reduced quality of life. Fast forward to 2001, when the first tyrosine kinase inhibitor (TKI), imatinib (STI571), was introduced in the clinic [Citation5]. Imatinib not only led to high response rates, but also markedly improved survival and quality of life when compared to historical controls. Since then, several TKIs, including dasatinib [Citation6], nilotinib [Citation7], bosutinib [Citation8], and ponatinib [Citation9], have been developed and approved by the FDA for the therapy of patients with CML. With TKIs, a recent study demonstrated that the survival of patients with CML is almost similar to the general population [Citation10Citation12]; however, treatment with these drugs is associated with significant impact on quality of life when compared to the general population [Citation13]. In a study by Efficace et al., patients consistently reported increased fatigue and depression, difficulties with sleep, and high symptom burden particularly nausea, diarrhea, pain, fluid retention (puffy face), and skin problems [Citation13], especially as compared to peers without cancer [Citation14]. With this marked improvement in survival, research has now shifted to mainly improving patients’ quality of life. The best way to improve quality of life is by discontinuing TKI therapy. This led several groups to investigate stopping TKIs in patients with a sustained deep molecular response (SDMR). Of all newly diagnosed patients with CML, 30–50% will achieve an SDMR after 5 years of therapy with nilotinib [Citation15], dasatinib [Citation16], or imatinib [Citation15,Citation16], thereby becoming eligible for a treatment-free remission (TFR) trial [Citation15,Citation16]. Of those patients, only 50% will successfully achieve TFR. These data suggest that only 20% of newly diagnosed CML patients will be able to achieve a TFR with current therapies and 70–80% will need to continue lifelong TKI therapy [Citation17].

For those patients who need to continue on lifelong therapy, the cost of TKIs cannot be overstated. Currently, the cost of TKIs in the USA ranges from $90,000 to $250,000 annually [Citation18]. The cost of lifelong TKI therapy is so prohibitive in some developing countries that HCT remains the standard of care [Citation19]. However, the cost should be decreasing soon with the availability of generic TKIs.

2. Expert opinion

Since the development of TKIs, little research has been ongoing in CML outside of TKI therapy. Some researchers and physicians think that no further research is needed in CML, given that survival is similar to the general population. However, most patients would disagree with that. Having to take a medication for the rest of their life with even low-grade side effects and possible long-term toxicities places significant psychological, physical, and financial burden on them. In recent years, most research has focused on optimizing TKI therapy by comparing the TKIs, reducing the dose of TKIs [Citation20], or attempting to stop TKIs and achieving a TFR. Three large randomized trials have demonstrated that second-generation TKIs (dasatinib [Citation6], nilotinib [Citation7], and bosutinib [Citation21]) lead to faster and deeper responses, but with no difference in overall survival. Now that the concept of TFR is finally catching on (and included in the National Comprehensive Cancer Network [NCCN] guidelines) more patients and physicians will consider TFR as the ultimate goal. To achieve that, current TKI therapy alone will not suffice. Several other therapies, either alone or in combination with TKIs, have demonstrated efficacy in the therapy of CML (e.g. proteosome inhibition, bcl-2 inhibition, immunotherapy, JAK2 inhibition, inecalcitol, interferon, arsenic trioxide, ABL001, and pioglitazone) [Citation22]. Incorporating other therapies in combination with TKIs will be necessary for more patients to achieve a TFR. The biggest challenge in further CML research is the toxicity of those potential combinations. Many patients and physicians will be hesitant to enroll on studies given the excellent overall survival with TKIs alone. In addition, the bar will be set very high for adverse events, and there will be a low tolerance for side effects in this patient population. The goal of those studies would be to eradicate the leukemia stem cell or maintain residual disease in a quiescent stage without therapy [Citation23].

Research in the coming years will focus on several areas. The first is better identification of patients who can achieve TFR. TKI discontinuation studies have demonstrated that certain criteria such as age, duration of TKI therapy, duration of deep molecular response, Sokal risk, proportion of mature Natural Killer (NK) cells, and digital PCR can predict successful achievement of TFR [Citation24Citation27]. However, these have not been consistent across studies. Recently, studies have demonstrated that patients with higher percentage of NK cells in the peripheral blood are more likely to achieve TFR. These new data suggest that a particular patients’ immune profile may predict relapse [Citation28]. Second, with the introduction of NCCN guidelines on discontinuation [Citation29], the importance of proper education of patients and providers on eligibility for TFR, of appropriate monitoring after stopping TKIs, and of the minimal triggers to restart therapy is prudent. The worst thing that could happen is that patients’ survival would be affected because of improper selection for TFR or improper monitoring. Another focus of research will be the addition of new therapies to improve the likelihood of TKI-treated patients to achieve a deep molecular response in order to attempt drug discontinuation. Ongoing studies include adding ruxolitinib or interferon [Citation30] to TKIs and attempting TFR for patients who achieve an SDMR with the addition of those therapies. Another ongoing study by Rousselot et al. will randomize patients to either TKI alone or TKI + pioglitazone. The study is designed to add experimental arms in the future in order to find the best combination for patients to achieve a TFR. Planned experimental arms include interferon, arsenic trioxide, anti-PDL1, and omacetaxine [Citation30]. In the USA, several CML experts are currently collaborating in a CML consortium named after the late Jean Khoury, MD (Jean Khoury Cure CML consortium), with the goal of developing trials to cure CML. Cure defined as off-therapy with no evidence of disease.

In conclusion, there has been rapid progression through the last century of effective treatment for CML with TKIs. While TKI therapy has been a major advance, for most patients it is a lifelong treatment with accompanied long-term side effects and financial toxicity. In our opinion, with the ongoing reignited interest in CML research, TKIs will remain part of the treatment armamentarium for CML therapy in the future, but will not be the only therapy and will not be lifelong.

Declaration of interest

A Atallah has participated in a consulting or advisory role for Novartis, Bristol-Myers Squibb and Pfizer. E Ritchie participated in a consulting or advisory role for Incyte, Pfizer and Novartis, received speakers' Bureau from Novartis and has received research funding via his institute from Bristol-Myers Squibb, Novartis and Pfizer. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. A reviewer on this manuscript has disclosed advisory board for Novartis, Pfizer, and Ariad/Incyte as well as consultant for Bristol–Myers Squibb.

Additional information

Funding

The authors are supported by National Cancer Institute grant 1R01CA184798-01A1.

References

  • Koeffler HP, Golde DW. Chronic myelogenous leukemia – new concepts (second of two parts). N Engl J Med. 1981 May;304:1269–1274.
  • Latagliata R, Romano A, Mancini M, et al. Discontinuation of alpha-interferon treatment in patients with chronic myeloid leukemia in long-lasting complete molecular response. Leuk Lymphoma. 2016;57:99–102.
  • Talpaz M, Mercer J, Hehlmann R. The interferon-alpha revival in CML. Ann Hematol. 2015 Apr;94(Suppl 2):S195–S207.
  • Champlin R, Ho W, Arenson E, et al. Allogeneic bone marrow transplantation for chronic myelogenous leukemia in chronic or accelerated phase. Blood. 1982 Oct;60:1038–1041.
  • Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001 Apr;344:1031–1037.
  • Kantarjian H, Shah NP, Hochhaus A, et al. Dasatinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med. 2010 Jun;17(362):2260–2270.
  • Saglio G, Kim DW, Issaragrisil S, et al. Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. N Engl J Med. 2010 Jun;17(362):2251–2259.
  • Cortes JE, Kim DW, Kantarjian HM, et al. Bosutinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia: results from the BELA trial. J Clin Oncol. 2012 Oct;1(30):3486–3492.
  • Cortes JE, Kantarjian H, Shah NP, et al. Ponatinib in refractory Philadelphia chromosome-positive leukemias. N Engl J Med. 2012 Nov;29(367):2075–2088.
  • Bower H, Bjorkholm M, Dickman PW, et al. Life expectancy of patients with chronic myeloid leukemia approaches the life expectancy of the general population. J Clin Oncol. 2016 Aug;20(34):2851–2857.
  • Gambacorti-Passerini C, Antolini L, Mahon FX, et al. Multicenter independent assessment of outcomes in chronic myeloid leukemia patients treated with imatinib. J Natl Cancer Inst. 2011 Apr;06(103):553–561.
  • Vigano I, Di Giacomo N, Bozzani S, et al. First-line treatment of 102 chronic myeloid leukemia patients with imatinib: a long-term single institution analysis. Am J Hematol. 2014 Oct;89:E184–7.
  • Efficace F, Cardoni A, Cottone F, et al. Tyrosine-kinase inhibitors and patient-reported outcomes in chronic myeloid leukemia: a systematic review. Leuk Res. 2013 Feb;37:206–213.
  • Phillips KM, Pinilla-Ibarz J, Sotomayor E, et al. Quality of life outcomes in patients with chronic myeloid leukemia treated with tyrosine kinase inhibitors: a controlled comparison. Supportive Care Cancer. 2013;21:1097–1103.
  • Hochhaus A, Saglio G, Hughes TP, 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 05//print;30:1044–1054.
  • Cortes JE, Saglio G, Kantarjian HM, 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.
  • Khoury HJ, Williams LA, Atallah E, et al. chronic myeloid leukemia: what every practitioner needs to know in 2017. Am Soc Clin Oncol Educ Book. 2017;37:468–479.
  • Experts in Chronic Myeloid L. The price of drugs for chronic myeloid leukemia (CML) is a reflection of the unsustainable prices of cancer drugs: from the perspective of a large group of CML experts. Blood. 2013 May;30(121):4439–4442.
  • Pasquini MC. Hematopoietic cell transplantation for chronic myeloid leukemia in developing countries: perspectives from Latin America in the post-tyrosine kinase inhibitor era. Hematology. 2012 Apr;17(Suppl 1):S79–82.
  • Naqvi K, Cortes JE, Skinner JA, et al. Early results of lower-dose dasatinib, 50 mg daily, in newly diagnosed Philadelphia chromosome-positive chronic myeloid leukemia in chronic phase. J Clin Oncol. 2017;35:e18551–e.
  • Cortes JE, Gambacorti-Passerini C, Deininger MWN, et al. Bosutinib (BOS) versus imatinib (IM) for newly diagnosed chronic myeloid leukemia (CML): initial results from the BFORE trial. J Clin Oncol. 2017;35:7002.
  • Ahmed W, Van Etten RA. Alternative approaches to eradicating the malignant clone in chronic myeloid leukemia: tyrosine-kinase inhibitor combinations and beyond. Hematol Am Soc Hematol Educ Program. 2013;2013:189–200.
  • Tantravahi SK, Guthula RS, O’Hare T, et al. Minimal residual disease eradication in CML: does it really matter? Curr Hematol Malig Rep. 2017 Oct 1;12(5):495–505.
  • Mori S, Vagge E, le Coutre P, et al. Age and dPCR can predict relapse in CML patients who discontinued imatinib: the ISAV study. Am J Hematol. 2015;90:910–914.
  • Kim DDH, Bence-Bruckler I, Forrest DL, et al. Treatment-free remission accomplished by dasatinib (TRAD): preliminary results of the Pan-Canadian tyrosine kinase inhibitor discontinuation trial. Blood. 2016;128:1922.
  • Lee SE, Choi SY, Song HY, 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.
  • Mahon F-X, Richter J, Guilhot J, et al. Cessation of tyrosine kinase inhibitors treatment in chronic myeloid leukemia patients with deep molecular response: results of the Euro-Ski trial. Blood. 2016;128:787.
  • Ilander M, Olsson-Stromberg U, Schlums H, et al. Increased proportion of mature NK cells is associated with successful imatinib discontinuation in chronic myeloid leukemia. Leukemia. 2017 May;31:1108–1116.
  • NCCN guidelines clinical practice guidelines in oncology. Version I. 2018-Jul 26;2017 [cited Aug 25].
  • Clinicaltrials.gov. 2017. https://clinicaltrials.gov/

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.