Vol 53, No 3 (2022)
Review article
Published online: 2022-04-05

open access

Page views 4466
Article views/downloads 455
Get Citation

Connect on Social Media

Connect on Social Media

Chronic myeloid leukemia: where do we stand, where can we go?

Krzysztof Lewandowski1, Jakub Lewandowski2
Acta Haematol Pol 2022;53(3):153-165.

Abstract

The introduction of BCR-ABL tyrosine kinase inhibitors to the treatment of chronic myeloid leukemia (CML) has significantly changed the long term therapy results.

After an initial 12 months of therapy with tyrosine kinase inhibitor (TKI), a 3-log reduction of the BCR-ABL copies number on an international scale is possible in 22–46% of patients, depending on the TKI used. In TKI-responsive patients, long-term TKI treatment results are even better, with the BCR-ABL transcript level decreasing over time, even to the point of becoming undetectable. Therefore, an operational cure can be diagnosed in CML patients with an optimal response to 1st-line TKI treatment, atherapy duration of longer than 5–8 years, and BCR-ABL transcript level below MR4.0–MR4.5 for a period of more than two years. The latter has been the basis of multiple concepts of permanent or periodic discontinuation of TKI treatment [treatment-free remission (TFR)]. Initial TKI discontinuation clinical trials resulted in satisfactory results, with a disease recurrence rate of c.40–60% after 2–3 years. The mechanism of disease recurrence was then studied , with detailed characterization of the CML stem cells (CML SCs) immunophenotype and the mechanisms of survival and self-renewal under TKI selective pressure. A better understanding of the biology of CML allowed the formulation of new therapy concepts of CML SCs eradication, and new criteria for successful TFR qualification.

REVIEW ARTICLE

Acta Haematologica Polonica 2022

Number 3, Volume 53, pages 153–165

DOI: 10.5603/AHP.a2022.0012

ISSN 0001–5814

e-ISSN 2300–7117

Chronic myeloid leukemia: where do we stand, where can we go?

Krzysztof Lewandowski●iD1*Jakub Lewandowski●iD2
1Department of Hematology and Bone Marrow Transplantation, Poznań University of Medical Sciences, Poznań, Poland
2Faculty of Chemistry, Adam Mickiewicz University, Poznań, Poland

*Address for correspondence: Krzysztof Lewandowski, Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, Szamarzewskiego 84, 60–569 Poznań, Poland, phone +48 61 854 93 83,
fax +48 61 854 93 56, e-mail: krzysztof.lewandowski@skpp.edu.pl

Copyright © 2022

The Polish Society of Haematologists and Transfusiologists,
Insitute of Haematology and Transfusion Medicine.

All rights reserved.

Received: 03.02.2022 Accepted: 11.02.2022

Abstract
The introduction of BCR-ABL tyrosine kinase inhibitors to the treatment of chronic myeloid leukemia (CML) has significantly changed the long term therapy results.
After an initial 12 months of therapy with tyrosine kinase inhibitor (TKI), a 3-log reduction of the BCR-ABL copies number on an international scale is possible in 22–46% of patients, depending on the TKI used. In TKI-responsive patients, long-term TKI treatment results are even better, with the BCR-ABL transcript level decreasing over time, even to the point of becoming undetectable. Therefore, an operational cure can be diagnosed in CML patients with an optimal response to 1st-line TKI treatment, a therapy duration of longer than 5–8 years, and BCR-ABL transcript level below MR4.0–MR4.5 for a period of more than two years. The latter has been the basis of multiple concepts of permanent or periodic discontinuation of TKI treatment [treatment-free remission (TFR)]. Initial TKI discontinuation clinical trials resulted in satisfactory results, with a disease recurrence rate of c.40–60% after 2–3 years. The mechanism of disease recurrence was then studied, with detailed characterization of the CML stem cells (CML SCs) immunophenotype and the mechanisms of survival and self-renewal under TKI selective pressure. A better understanding of the biology of CML allowed the formulation of new therapy concepts of CML SCs eradication, and new criteria for successful TFR qualification.
Key words: chronic myeloid leukemia, tyrosine kinase inhibitors, chronic myeloid leukemia stem cells, immune system escape, treatment-free remission, new treatment concepts
Acta Haematologica Polonica 2022; 53, 3: 153–165

Introduction

The introduction of imatinib (IM) to the treatment of chronic myeloid leukemia (CML) significantly changed the long term therapy results, with 5-year overall survival (OS) of 91.7% and progression-free survival (PFS) of 94.7% [1]. Recently, the second (2G-TKI, nilotinib, dasatinib) and third generation (3G-TKI, ponatinib) of BCR-ABL tyrosine kinase inhibitors (TKIs) have become widely used in CML patients intolerant/resistant to first line treatment with TKIs [2, 3]. Moreover, the fourth-generation allosteric BCR-ABL1 tyrosine kinase inhibitor [4G-TKI, asciminib (ABL001)] has been approved by the US FDA in CML patients resistant to first-, second- and third-generation TKIs. Its high efficacy has been proven, both in clinical trials [4] and in real-life conditions [5]. The detailed characteristics of currently used TKIs in CML patients are set out in Table I [4–31].

Table I. Characteristics of currently used tyrosine kinase inhibitors (TKIs) in chronic myeloid leukemia patients

TKI/drug properties

Imatinib

Nilotinib

Dasatinib

Bosutinib

Ponatinib

Asciminib

Chemical structure

Lewandowski_tabela_01_kol1.jpg

Lewandowski_tabela_01_kol2.jpg

Lewandowski_tabela_01_kol3.jpg

Lewandowski_tabela_01_kol4_NEW.jpg

Lewandowski_tabela_01_kol5.jpg

Lewandowski_tabela_01_kol6.jpg

International Union of Pure and Applied Chemistry (IUAPC) name

4-[(4-methylpiperazin-1-yl)methyl]-N-[4-methyl-3-[(4-pyridin-3-ylpyrimidin-2-yl)amino]phenyl]benzamide

4-methyl-N-[3-(4--methylimidazol-1-yl)-5-(trifluoromethyl)phenyl]--3-[(4-pyridin-3-ylpyrimidin--2-yl)amino]benzamide

N-(2-chloro-6-methylphenyl)--2-[[6-[4-(2-hydroxyethyl)-
piperazin-1-yl]-2-methyl-pyrimidin-4-yl]amino]-1,3-thiazole-5-carboxamide

4-(2,4-dichloro-5-methoxyanilino)-6-methoxy-7-[3-(4-methyl-piperazin-1-yl)propoxy]-
quinoline-3-carbonitrile

3-(2-imidazo[1,2-b]-
pyridazin-3-ylethynyl)-4-methyl-N-[4-[(4-methyl-piperazin-1-yl)methyl]-3--(trifluoromethyl)phenyl]-
benzamide

N-[4-[chloro(difluoro)-
methoxy]phenyl]-6-[(3R)-3-hydroxypyrrolidin-1-yl]-5-(1H-pyrazol-5-yl)pyridine-3-carboxamide

Molecular formula

C29H31N7O

C28H22F3N7O

C22H26ClN7O2S

C26H29Cl2N5O3

C29H27F3N6O

C20H18ClF2N5O3

Spectrum of inhibitory activity

ABL1

ARG

BCR-ABL

KIT

PDGFR

DDR1

NQO2

ABL1

ARG

BCR-ABL

KIT

PDGFR

DDR1

NQO2

ABL1, ARG, BCR-ABL, KIT, PDGFR, SRC,YES,FYN, LYN, HCK, LCK, FGR, BLK, FRK, CSK,BTK, TEC, BMX, TXK, DDR1, DDR2, ACK, ACTR2B, ACVR2, BRAF, EGFR/ERBB1-5, EPHA8,EPHB1-2, EPHB4, EPHB6, ERBB2, ERBB4,FAK, GAK, GCK, HH498/TNNI3K, ILK, LIMK1-2, MAP2K5, MAP3K1-4, MAP4K1, MAP4K5/KHS1, MAPK11/

/p38 beta, MAPK14/p38 alpha, MYT1,NLK, PTK6/

/Brk, QIK, QSK, RAF1, RET, RIPK2, SLK, STK36/ULK, SYK, TAO3, TESK2, TYK2, ZAK [6]

BCR-ABL1, ABL1, SRC, LYN, HCK

ABL1, KIT, PDGFR, SRC family, VEGFR, EGFR, HER2, FLT3, FGFR, and JAK2 [7]

BCR-ABL1 [8]

BCR-ABL tyrosine kinase inhibitory mode of action

Works by binding close to the

ATP binding site, locking it in a closed or self-inhibited conformation, therefore inhibiting the enzyme activity of the protein semicompetitively [9, 10] CGP57148B, a 2-phenylaminopyrimidine derivative, has been shown to selectively inhibit the tyrosine kinase of ABL and BCR-ABL. We report here that this compound selectively suppresses the growth of colony- forming unit-granulocyte/macrophage (CFU-GM

Binds to and stabilizes inactive conformation of kinase domain of Abl protein [11]

Binds to ATP-binding site, but extends in opposite direction from imatinib. Binds inactive and active conformation of ABL kinase domain, requires fewer contact points with ABL, and has a greater affinity to ABL kinase domain compared to IM [12, 13]

ATP-competitive inhibitor of Src and Abl tyrosine kinases [14]

Acts as a multikinase inhibitor. Introduction of a triple bond ethynyl linker allowed spanning of bulky T315I isoleucine residue side chain in ATP-binding site, and overcame resistance to prior generation TKIs [15, 16]

Acts as an allosteric inhibitor and engages a vacant pocket at site of kinase domain normally occupied by myristoylated N-terminal of ABL1 — a motif that serves as an allosteric negative regulatory element lost on fusion of ABL1 to BCR

BCR, myristoylated N-terminal is lost and ABL1 kinase is activated. By allosterically binding, to myristoyl site, asciminib mimics myristate and restores inhibition of BCR-ABL1 kinase activity [4]

BCR-ABL tyrosine kinase binding conformation

Inactive

Inactive

Active

Both

Inactive

Specifically targeting ABL myristoyl pocket

Half life time (T1/2)

~20 hours

~17 hours

3–5 hours

32.4–41.2 hours enabling daily dose [17]

24 hours

5.5 hours (40 mg/d)

9 hours (200 mg bid)

Resistant BCR-ABL KD mutants**
[8, 15, 18–23]

Y253

E255

T315

M244

L248

G250

Q252

F317

M351

M355

F359

H396

T315

L248

Y253

E255

F359

T315

V299

F317

T315

V299

L248

G250

E255

F317

E250*

Y253*

E255*

F311

A337

W464

P465

V468

I502

Oral dose per day

CP 400 mg/d

AP 600 mg/d

BP 800 mg/d

CP 2 × 300 mg
(2nd-line)

2 × 400 mg
(1st-line)

CP 100 mg/d

AP/BP 140 mg/d

CP 500 mg/d

CP 15–45 mg/d

CP 80 mg/d
or 40 mg bid

Main off-target effect

Hematologic:

  • anemia
  • neutropenia
  • thrombocytopenia

Non-hematologic:

  • edema (periorbital and peripheral)
  • muscle cramps
  • musculoskeletal pain
  • diarrhea [24]

Hematologic:

  • thrombocytopenia
  • granulocytopenia
  • anemia

Non-hematologic:

  • pruritus
  • asthenia

Cardiovascular:

  • cardiovascular
  • ischemic adverse events [25, 26]

Hematologic:

  • thrombocytopenia
  • anemia
  • neutropenia

Non-hematologic:

  • endocrine disorders (gynecomastia, irregular menses, hypoglycemia, hyperglycemia, increased triglyceride and cholesterol levels)
  • fluid retention
  • nausea, vomiting, diarrhea

Cardiovascular:

  • pericardial effusion
  • pulmonary artery hypertension [27, 28]

Hematologic:

  • thrombocytopenia
  • neutropenia

Non-hematologic:

  • rash
  • nausea
  • diarrhea
  • vomiting
  • elevated serum aminotransferases [29]

Hematologic:

  • anemia
  • thrombocytopenia
  • neutropenia

Non-hematologic:

  • rash
  • elevated serum lipase
  • pancreatitis

Cardiovascular:

  • hypertension
  • chest pain [30]

Hematologic:

  • thrombocytopenia and/or neutropenia

Non-hematologic:

  • hepatic impairment
  • asymptomatic amylase and/or lipase elevations

Cardiovascular:

Current results of CML treatment

Data originating from clinical trials and from real life studies has confirmed the high efficacy of TKIs in CML patients in the chronic phase in terms of the 3-log reduction of BCR-ABL1 copies number (major molecular remission, MMR) with a well standardized real-time PCR technique in the blood after 12 months of treatment. MMR response rates differed depending on the type and dose of TKI used, and amounted to 22–36.9% for IM 400 mg once a day, 44% for nilotinib 300 mg twice a day, 43% for nilotinib 400 mg twice a day, 46% for dasatinib 100mg once a day, and 47.2% for bosutinib 500 mg once a day [1, 32–34].

The long-term TKI treatment results show that in the TKI-responsive patients the BCR-ABL transcript level continuously decreases over time, to the point of becoming undetectable. The overall cumulative incidence of the confirmed MR4.5, and stable MR4.5 (4.5log reduction in BCR-ABL1 copies number in international scale, IS) after eight years of IM therapy is 51.7% and 36.5%, respectively [35]. Real life data is in agreement with the computer simulation results, showing treatment time to MR4.5 to be 10.7 and 9.1 years in IM-treated patients participating in the IRIS trial (training set) and the CML IV trial (validation set), respectively [36]. This data forms the basis of the concept of an operational cure and the permanent or temporary discontinuation of TKI treatment [treatment-free remission (TFR)] [37].

Data concerning the frequency of deep molecular responses (DMR, defined as the reduction of the transcript level below MR4 or MR4.5) on TKI treatment has been accumulated subsequently. Its analysis allowed the formulation of minimal criteria which should be fulfilled for a TFR attempt in CML patients, including a low or intermediate Sokal score, a typical BCR-ABL1 transcript type at diagnosis, a chronic phase of CML in the past history, an optimal response to 1st-line TKI treatment, a TKI therapy duration of longer than eight years, a DMR at the time of qualification, and a duration of DMR monitored in a standardized laboratory of longer than two years. Initial study results showed that only 10–12% of patients on IM appeared to be eligible for the discontinuation of a TKI [37, 38]. Subsequent data has shown that only a minority of CML patients reaching the sustained DMR on TKI therapy were candidates for the discontinuation of treatment without the risk of a molecular disease recurrence. Until now, many possible solutions have been proposed for optimizing the process of CML patient qualification for a TFR attempt. Initially, only those patients with an MR5.0 or an undetectable BCR-ABL transcript were qualified for the TFR studies (STIM pilot, STIM1, STRIM2, ASTIM and TWISTER) [39–42]. In all the aforementioned trials, a molecular CML recurrence was defined as BCR-ABL1 positivity in two consecutive assessments, or the loss of a MMR (or ≥1 log increase of the transcript level in STIM1 and STIM 2 trails). The TFR rate after the median follow-up of 12 months was 61% (STIM2), 50% after 18 months (STIM pilot), 61% after 31 months (A-STIM), 47% after 42 months (TWISTER), and 38% after 77 months (STIM2). The EURO-SKI trial results confirmed that there was no difference in the TFR rates between patients with >MR4.5 and MR4 [43, 44]. Afterwards, patients with a stable MR4.0 were also enrolled in TFR clinical trials. Unfortunately, to date there is no consensus regarding the criteria which should be used for the qualification of CML patients for an TFR attempt to minimize the probability of MMR loss. According to the different criteria for TFR proposed by Hughes et al., Rea et al., Hochhaus et al., and Radich et al., the probability of CML patient recruitment for TFR varies from 9.5% to 55% [45–49]. Molecular recurrence-free survival after TKI cessation, according to the eligibility criteria proposed by Hughes et al., Rea et al., Radich et al., and Hochhaus et al., varies from 35% to 60% after a follow-up of 45–100 months [45–49]. Therefore, considerable efforts have been made to further optimize the criteria for TFR attempt qualification. The incorporation of genomic data into future model(s) of unfavorable risk assessment will likely change the currently used algorithms for TFR qualification [50–53].

Nowadays, the NCCN and ELN guidelines recommend the re-initiation of TKI therapy at the time of molecular recurrence, defined as a loss of MMR after the first TFR attempt [48, 54].

The 2G-TKI discontinuation studies results are limited. They include 327 patients with 1st-line TKI failure (IM, interferon/IM), and 190 patients treated upfront with nilotinib. Unfortunately, different criteria were applied in different studies for the TFR attempt qualification before the 2G-TKI discontinuation (MR4–MR4.5), and different definitions of molecular disease relapse (loss of MMR to loss of MR4.5) were applied. For these reasons, the interpretation of TFR rates at 24 months (ranging from 44 to 62.8 months) is difficult [50, 55–59].

According to recent data, the re-initiation of TKI therapy results in secondary molecular remission in a significant proportion of CML patients. Therefore, the idea of a second TFR attempt has been tested. In 2017, Legros et al. presented data concerning 70 patients successfully treated with IM who attempted a first TFR (IM = 60, nilotinib = 5, dasatinib = 5) and, after disease relapse, underwent a second TFR attempt. The TFR probability at 6, 12, 24, and 36 months after the second attempt to discontinue TKI was established at 66%, 48%, 42%, and 35%, respectively [60]. However, it should be mentioned that a second TFR is not yet considered standard practice.

It was previously documented that the majority of patients in DMR still harbor leukemic cells [61] capable of initiating disease relapse upon the withdrawal of TKI treatment [41]. The presence of CML leukemic stem cells in IM-treated patients in DMR with undetectable levels of mRNA was confirmed in 32.8% of samples by Pagani et al. with the help of a genomic DNA Q-PCR assay [62]. The mechanism of the persistence of CML SCs in TKI-responsive CML patients in DMR is not fully understood. Recently, it was postulated that CML SCs are protected from MHC class I-dependent CD8+ cytotoxic T lymphocytes (CTLs) elimination in the bone marrow by regulatory T cells (Tregs) expressing tumor factor 4 receptor (Tnfrsf4). This hypothesis is based on the observation that Tregs are preferentially localized in the CML bone marrow close to CD8+ CTLs, and that TNFRSF4 mRNA levels correlate with the expression of Treg-restricted transcription factor FOXP3 [63]. Moreover, it has been shown that human CML stem cells are insensitive to IM, despite the inhibition of BCR-ABL activity [64]. This observation forms the basis of a hypothesis that primitive CML cells are not oncogene-addicted [65, 66].

CML, TKI treatment and immune system function

It has been postulated that TKI-induced changes in immune system functioning may affect the risk of a CML relapse after TKI discontinuation (Table II)

Table II. Off-target effect of BCR-ABL tyrosine kinase inhibitor’s administration on immune system in patients with chronic myeloid leukemia

Off target effect on immune system

Imatinib

Nilotinib

Dasatinib

Bosutinib

Ponatinib

Hypogammaglobulinemia
(via inhibition of Burton kinase) [72–74]

+

Decrease in memory-B-cell count
(via inhibition of Burton kinase) [72, 75]

+

+

B-cell immune responses impairment [74, 76]

+

+

+

Treg and effector T cells function impairment
(via inhibition of Src kinase) [77–79]

+

+

+

Decreased cytotoxicity and reactivity
of NK cells [79]

+

+

Abrogation NK cells cytokine production [79]

+

Decreased proliferation and function
of CD8+ T lymphocytes [80]

+

LGLs expansion [81, 82]

+

Increased proliferation of cytotoxic (CD3+CD8+) T cells and/or NK cells

(CD3–CD16+/56+) [83]

+

Decrease of NK cells count [75]

+

+

+

Decrease of MDSCs level [84]

?

?

?

?

?

in a different way. In CML patients in the chronic phase, suppression of the immune system is present at the time of diagnosis. This is mainly caused by the promotion of the expansion of myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg) by cytokines/chemokines released by proliferating CML progenitor cells. MDSC originate from malignant BCR-ABL1 clone mediate immunosuppressive activity via a number of mechanisms, including the increased production of reactive oxygen, nitrogen species, arginase-1 (molecule inhibiting T cells), and TGF-β1 [67].

Moreover, MDSC can recruit Treg and inhibit cytotoxic T cells [68]. The immune escape of malignant cells is also promoted due to increased expression of the programmed death-1 (PD-1) inhibitory molecule on the CD4+/CD8+ T cells [69] and PD-L1 upregulation on CML cells [70, 71]. Quantitative and functional defects of the innate effector natural killer (NK) cells and the cytotoxic T-lymphocyte responses to leukemia-associated antigens (CTL-LAA) broaden the spectrum of immune system defects in CML patients at the time of diagnosis [69].

The significance of the immune system for a successful TFR was confirmed by the Immunostim study, which documented an association between elevated peripheral blood natural killer (NK) cells and a positive clinical outcome following IM discontinuation [85]. Other immunomodulatory effects of TKI administration leading to immune system re-activation and restoration of effector-mediated immune surveillance were recently documented. TKI treatment resulted in the restoration of NK cell receptor repertoire and an enhanced NK cell function, a decrease of immune suppressors (MDSC, Treg and T lymphocytes PD-1+), the restoration of LLA-CTL responses including PD-1 downregulation to normal levels, and an increase in DC number and antigen presenting cell function [67]. The restoration of immune system function in CML patients on TKI seems to be optimal after reaching MR4.5 (BCR-ABL1 ≤0.0032%), a time when the increased effector NK cell number and function and T cell immune responses, and reduced numbers of PD-1+ CD4+/CD8+ T cells and monocytic MDSC are maximal [69].

Another question concerns the role of immune surveillance by natural killer and T cells in maintaining a successful TFR and disease activity control [66]. In 2017, Jacomet et al. postulated that the deficiency of iNKT/InnateCD8+ T cells axis is present in CML patients [86]. Their hypothesis was confirmed by the observation of CML patients in TFR ≥2 years carried out by Cayssials et al. documenting the increase of functionally active innate CD8(+) T-cells [NK-like KIR/NKG2A(+)] and their number [87]. The presence of specific CTLs directed against CXorf48 (cancer testis antigen) expressed in LSC is also correlated with the relapse rate in CML patients who discontinued imatinib after maintaining complete molecular remission for more than two years [88]. The success of the TFR attempt likely depends on the ‘quite normal’ efficiency of the immune system. This is possible only if MR4.5 response to TKI treatment is reached, and when NK cells number and effector T-cell cytolytic function is increased, and when PD-1 expression on the T-cell and numbers of monocytic MDSCs is reduced [69].

Chronic myeloid leukemia stem cells

CML SCs are not fully defined yet in terms of immune and functional characteristics [89]. CML SCs likely share an immunophenotypic profile with normal hematopoietic stem cells (HSCs) and reside in the CD34+/CD38–/Lyn– cell fraction [90]. Also, CD25 and CD44 are expressed in both CML SCs and healthy HSCs [91, 92]. On the other hand, interleukin-1-receptor accessory protein (IL-1RAP) and CD26 [dipeptidyl peptidase-4 (DPP4)] are aberrantly expressed on the CML SCs’ surface, but not in normal CD34+ cells [93–95]. CML SCs can self-renew and generate large numbers of leukemic progenitor cells (CD34+CD38+) with the capacity to differentiate or enter a dormant state. In 2012, BCR-ABL1-independent CML SCs was postulated by Hamilton et al. [96]. They documented that the process of survival and self-renewal of CML LSCs was associated with activation of the cellular signalling process, including cell-intrinsic and cell-extrinsic survival pathways.

The first group includes abnormal signaling via the Janus kinase–signal transducer and activator of transcription 3/5 (JAK–STAT3/5), WNT/β-catenin, sonic Hedgehog (Hh) or PIK3/AKT pathways, abnormal function of protein phosphatase-2 (PP2A), promyelocytic leukemia (PML) protein, dual specificity tyrosine phosphorylation regulated kinase 2 (DYRK2), repression of autophagy process, deregulated expression of microRNAs (i.e. the upregulation of miR-29a-3p, miR660-5p, has-mir183), disturbed epigenetic regulation of genes expression by enhancer of Zeste Homolog 2 [a member of the polycomb repressive complex 2 (PRC2)], deregulation of fatty acid cellular metabolism due to arachidonate 5-lipoxygenase (ALOX5)-associated abnormalities of arachidonic acid conversion to leukotrienes required for malignant cells self-renewal, and BCR-ABL1-related autocrine production of cytokines resulting in growth factors-independent STAT5 activation [i.e. interleukin (IL) 3, granulocyte colony-stimulating factor (G-CSF)]. Other key regulators influencing the apoptosis, self-renewal, cell fate and senescence process of CML SCs include abnormal transforming growth factor-β (TGF-β)/Forkhead box O (FOXO) interaction and Musashi 2 (Msi2)/Numb of NOTCH signaling [97–105].

Bone marrow microenvironment and CML SCs

CML SCs reside in the same bone marrow microenvironment (BMM) as normal stem cells. The cross-talk between CML SCs and BMM is mediated by soluble factors like cytokines [IL-3, IL-1α/β, granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-6, and interferon gamma (IFN-γ), myostatin propeptide (MSTNpp), sCD14, IL-21 and IL-13v, and CCL-28], chemokines [i.e. C-X-C motif chemokine ligand 12 (CXCL12)] secreted by mesenchymal stromal cells, and osteoblastic cells), growth factors via autocrine and/or paracrine mechanisms, and cell-to-cell (mesenchymal stromal cells, osteoblastic cells endothelial cells, neurons) direct interactions via surface adhesive molecules (i.e. β1-integrin). The aforementioned interaction may have resulted in an enhanced proliferation, quiescence, and drug resistance of CML SCs [106–109]. Lastly, the role of miR-126 secreted by the endothelial cells in the process of CML SC quiescence and self-renewal control has been postulated [110]. Similarly, the role of miR-300, expressing dual anti-proliferative and PP2A-acivating properties, in the process of CML SCs quiescence and persistence has recently also been confirmed [111].

Due to the high diversity of CML SCs (subclonal character, different metabolic characteristics, and molecular and immunophenotypic fingerprints) and high inter- and intra-patient heterogeneity, the possibility of a common, unified strategy for CML SC eradication has been neglected. What is more, CML SCs eradication is now irrelevant due to the significant improvement of TKI long term treatment results and encouraging results of TFR attempts [112].

The current concept of chronic myeloid leukemia treatment with tyrosine kinase inhibitors based on the reduction of measurable residual disease and the recovery of immune system function is set out in Figure 1.

154536.png
Figure 1. Current concept of chronic myeloid leukemia (CML) treatment with tyrosine kinase inhibitors based on reduction of measurable residual disease and immune effector recovery; CTL — cytotoxic T lymphocytes; LAA — leukemia-associated antigens; MDSCs — myeloid derived suppressor cells; MR — molecular response in log scale; PD-L1 — programmed death-ligand 1

New therapeutic approaches concerning CML treatment

A better understanding of the biology of CML has allowed the formulation of a number of new therapy concepts for the eradication of leukemic cells with the help of immunotherapy or chimeric antigen receptor–engineered T cells (CAR-T) directed against CXorf48 (cancer testis antigen) or IL1RAP (IL-1 receptor accessory protein) [88, 113]. Recently, quiescent primitive SCs insensitive to IM subpopulation of CML were identified in a CD36+ cell subpopulation with the help of an RNA-seq study [114]. This data forms the basis of the concept of antibody-based therapeutic targeting of CML SCs [112, 114, 115].

Moreover, an innovative strategy based on a liposome loaded with the BCL2 inhibitor venetoclax exploiting begelomab (an anti-CD26 antibody) has been proposed to target positive CML SCs CD26+ [116] more selectively.

In our opinion, its use, in combination with TKI and other drugs targeting alternative CML LCs survival pathways, should be the future of combinatorial therapy for the eradication of CML stem cells.

CML and the future

Integrative genomic analysis reveals cancer-associated mutations at the diagnosis of CML in patients. WES, RNA-seq and gene expression profiling studies have identified a number of molecular aberrations in addition to BCR-ABL1, among others affecting epigenetic regulators such as ASXL1, DNMT3A, TET2, SETD1B and transcription factors (IKZF1, RUNX1) [51–53, 117]. Its presence could also potentially influence the process of making therapeutic decisions in future [53, 118].

Conflict of interest

None.

Financial support

None.

Ethics

The work described in this article has been carried out in accordance with The Code of Ethics of the World Medical Association (Declaration of Helsinki) for experiments involving humans; EU Directive 2010/63/EU for animal experiments; Uniform requirements for manuscripts submitted to biomedical journals.

References

  1. 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; 30(5): 1044–1054, doi: 10.1038/leu.2016.5, indexed in Pubmed: 26837842.
  2. Pajiep M, Conte C, Huguet F, et al. Patterns of tyrosine kinase inhibitor utilization in newly treated patients with chronic myeloid leukemia: an exhaustive population-based study in France. Front Oncol. 2021; 11: 675609, doi: 10.3389/fonc.2021.675609, indexed in Pubmed: 34660261.
  3. Gambacorti-Passerini C, Chen C, Davis C, et al. Treatment patterns and clinical outcomes of tyrosine kinase inhibitors in chronic-phase CML in clinical practice: 3-year European SIMPLICITY data. Eur J Hae­matol. 2021; 106(1): 82–89, doi: 10.1111/ejh.13524, indexed in Pubmed: 32989776.
  4. Hughes TP, Mauro MJ, Cortes JE, et al. Asciminib in chronic myeloid leukemia after ABL kinase inhibitor failure. N Engl J Med. 2019; 381(24): 2315–2326, doi: 10.1056/NEJMoa1902328, indexed in Pubmed: 31826340.
  5. Garcia-Gutiérrez V, Luna A, Alonso-Dominguez JM, et al. Safety and efficacy of asciminib treatment in chronic myeloid leukemia patients in real-life clinical practice. Blood Cancer J. 2021; 11(2): 16, doi: 10.1038/s41408-021-00420-8, indexed in Pubmed: 33563899.
  6. Greuber EK, Smith-Pearson P, Wang J, et al. Role of ABL family kinases in cancer: from leukaemia to solid tumours. Nat Rev Cancer. 2013; 13(8): 559–571, doi: 10.1038/nrc3563, indexed in Pubmed: 23842646.
  7. Lee H, Basso IN, Kim DD. Target spectrum of the BCR-ABL tyrosine kinase inhibitors in chronic myeloid leukemia. Int J Hematol. 2021; 113(5): 632–641, doi: 10.1007/s12185-021-03126-6, indexed in Pubmed: 33772728.
  8. Wylie AA, Schoepfer J, Jahnke W, et al. The allosteric inhibitor ABL001 enables dual targeting of BCR-ABL1. Nature. 2017; 543(7647): 733–737, doi: 10.1038/nature21702, indexed in Pubmed: 28329763.
  9. Buchdunger E, Zimmermann J, Mett H, et al. Inhibition of the Abl protein-tyrosine kinase in vitro and in vivo by a 2-phenylaminopyrimidine derivative. Cancer Res. 1996; 56(1): 100–104, indexed in Pubmed: 8548747.
  10. O’Hare T, Pollock R, Stoffregen EP, et al. The tyrosine kinase inhibitor CGP57148B selectively inhibits the growth of BCR-ABL-positive cells. Blood. 1997; 90(9): 3691–3698, indexed in Pubmed: 9345054.
  11. Weisberg E, Manley PW, Breitenstein W, et al. Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell. 2005; 7(2): 129–141, doi: 10.1016/j.ccr.2005.01.007, indexed in Pubmed: 15710326.
  12. Lombardo LJ, Lee FY, Chen P, et al. Discovery of N-(2-chloro-6-methyl- phenyl)-2-(6-(4-(2-hydroxyethyl)- piperazin-1-yl)-2-methylpyrimidin-4- -ylamino)thiazole-5-carboxamide (BMS-354825), a dual Src/Abl kinase inhibitor with potent antitumor activity in preclinical assays. J Med Chem. 2004; 47(27): 6658–6661, doi: 10.1021/jm049486a, in­dexed in Pubmed: 15615512.
  13. Shah NP, Tran C, Lee FY, et al. Overriding imatinib resistance with a novel ABL kinase inhibitor. Science. 2004; 305(5682): 399–401, doi: 10.1126/science.1099480, indexed in Pubmed: 15256671.
  14. Konig H, Holyoake TL, Bhatia R. Effective and selective inhibition of chronic myeloid leukemia primitive hematopoietic progenitors by the dual Src/Abl kinase inhibitor SKI-606. Blood. 2008; 111(4): 2329–2338, doi: 10.1182/blood-2007-05-092056, indexed in Pubmed: 18056843.
  15. O’Hare T, Shakespeare WC, Zhu X, et al. AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell. 2009; 16(5): 401–412, doi: 10.1016/j.ccr.2009.09.028, indexed in Pubmed: 19878872.
  16. Zhou T, Commodore L, Huang WS, et al. Structural mechanism of the Pan-BCR-ABL inhibitor ponatinib (AP24534): lessons for overcoming kinase inhibitor resistance. Chem Biol Drug Des. 2011; 77(1): 1–11, doi: 10.1111/j.1747-0285.2010.01054.x, indexed in Pubmed: 21118377.
  17. Abbas R, Hug BA, Leister C, et al. A phase I ascending single-dose study of the safety, tolerability, and pharmacokinetics of bosutinib (SKI-606) in healthy adult subjects. Cancer Chemother Pharmacol. 2012; 69(1): 221–227, doi: 10.1007/s00280-011-1688-7, indexed in Pubmed: 21691746.
  18. Cowan-Jacob SW, Fendrich G, Floersheimer A, et al. Structural biology contributions to the discovery of drugs to treat chronic myelogenous leukaemia. Acta Crystallogr D Biol Crystallogr. 2007; 63(Pt 1): 80–93, doi: 10.1107/S0907444906047287, indexed in Pubmed: 17164530.
  19. Levinson NM, Boxer SG. Structural and spectroscopic analysis of the kinase inhibitor bosutinib and an isomer of bosutinib binding to the Abl tyrosine kinase domain. PLoS One. 2012; 7(4): e29828, doi: 10.1371/journal.pone.0029828, indexed in Pubmed: 22493660.
  20. Tokarski JS, Newitt JA, Chang CY, et al. The structure of dasatinib (BMS-354825) bound to activated ABL kinase domain elucidates its inhibitory activity against imatinib-resistant ABL mutants. Cancer Res. 2006; 66(11): 5790–5797, doi: 10.1158/0008-5472.CAN-05-4187, indexed in Pubmed: 16740718.
  21. Lu S, Qiu Y, Ni D, et al. Emergence of allosteric drug-resistance mutations: new challenges for allosteric drug discovery. Drug Discov Today. 2020; 25(1): 177–184, doi: 10.1016/j.drudis.2019.10.006, indexed in Pubmed: 31634592.
  22. Qiang W, Antelope O, Zabriskie MS, et al. Mechanisms of resistance to the BCR-ABL1 allosteric inhibitor asciminib. Leukemia. 2017; 31(12): 2844–2847, doi: 10.1038/leu.2017.264, indexed in Pubmed: 28819281.
  23. Braun TP, Eide CA, Druker BJ. Response and resistance to BCR-ABL1-targeted therapies. Cancer Cell. 2020; 37(4): 530–542, doi: 10.1016/j.ccell.2020.03.006, indexed in Pubmed: 32289275.
  24. Mauro MJ. Lifelong TKI therapy: how to manage cardiovascular and other risks. Hematology Am Soc Hematol Educ Program. 2021; 2021(1): 113–121, doi: 10.1182/hematology.2021000239, indexed in Pubmed: 34889360.
  25. Huguet F, Cayuela JM, Cambier N, et al. AdheRMC Investigators. Nilotinib efficacy, safety, adherence and impact on quality of life in newly diagnosed patients with chronic myeloid leukaemia in chronic phase: a prospective observational study in daily clinical practice. Br J Haematol. 2019; 187(5): 615–626, doi: 10.1111/bjh.16145, indexed in Pubmed: 31394591.
  26. Fachi MM, Tonin FS, Leonart LP, et al. Haematological adverse events associated with tyrosine kinase inhibitors in chronic myeloid leukaemia: a network meta-analysis. Br J Clin Pharmacol. 2019; 85(10): 2280–2291, doi: 10.1111/bcp.13933, indexed in Pubmed: 30907446.
  27. Shah NP, Rousselot P, Schiffer C, et al. Dasatinib in imatinib-resistant or -intolerant chronic-phase, chronic myeloid leukemia patients: 7-year follow-up of study CA180-034. Am J Hematol. 2016; 91(9): 869–874, doi: 10.1002/ajh.24423, indexed in Pubmed: 27192969.
  28. Nekoukar Z, Moghimi M, Salehifar E. A narrative review on adverse effects of dasatinib with a focus on pharmacotherapy of dasatinib-induced pulmonary toxicities. Blood Res. 2021; 56(4): 229–242, doi: 10.5045/br.2021.2021117, indexed in Pubmed: 34776414.
  29. Khoury HJ, Gambacorti-Passerini C, Brümmendorf TH. Practical management of toxicities associated with bosutinib in patients with Philadelphia chromosome-positive chronic myeloid leukemia. Ann Oncol. 2018; 29(3): 578–587, doi: 10.1093/annonc/mdy019, indexed in Pubmed: 29385394.
  30. Jain P, Kantarjian H, Jabbour E, et al. Ponatinib as first-line treatment for patients with chronic myeloid leukaemia in chronic phase: a phase 2 study. Lancet Haematol. 2015; 2(9): e376–e383, doi: 10.1016/S2352-3026(15)00127-1, indexed in Pubmed: 26436130.
  31. Réa D, Mauro MJ, Boquimpani C, et al. A phase 3, open-label, randomized study of asciminib, a STAMP inhibitor, vs bosutinib in CML after 2 or more prior TKIs. Blood. 2021; 138(21): 2031–2041, doi: 10.1182/blood.2020009984, indexed in Pubmed: 34407542.
  32. 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(20): 2333–2340, doi: 10.1200/JCO.2015.64.8899, indexed in Pubmed: 27217448.
  33. Brümmendorf TH, Cortes JE, de Souza CA, 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(1): 69–81, doi: 10.1111/bjh.13108, indexed in Pubmed: 25196702.
  34. Cortes JE, Gambacorti-Passerini C, Deininger MW, et al. Bosutinib versus imatinib for newly diagnosed chronic myeloid leukemia: results from the randomized BFORE trial. J Clin Oncol. 2018; 36(3): 231–237, doi: 10.1200/JCO.2017.74.7162, indexed in Pubmed: 29091516.
  35. Branford S, Yeung DT, Ross DM, et al. Early molecular response and female sex strongly predict stable undetectable BCR-ABL1, the criteria for imatinib discontinuation in patients with CML. Blood. 2013; 121(19): 3818–3824, doi: 10.1182/blood-2012-10-462291, indexed in Pubmed: 23515925.
  36. Horn M, Glauche I, Müller MC, et al. Model-based decision rules reduce the risk of molecular relapse after cessation of tyrosine kinase inhibitor therapy in chronic myeloid leukemia. Blood. 2013; 121(2): 378–384, doi: 10.1182/blood-2012-07-441956, indexed in Pubmed: 23175686.
  37. Ross DM, Branford S, Seymour JF, 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(4): 515–522, doi: 10.1182/blood-2013-02-483750, indexed in Pubmed: 23704092.
  38. Cortes J, Rea D, Lipton JH. Treatment-free remission with first- and second-generation tyrosine kinase inhibitors. Am J Hematol. 2019; 94(3): 346–357, doi: 10.1002/ajh.25342, indexed in Pubmed: 30394563.
  39. Hughes TP, Lipton JH, Spector N, et al. Deep molecular responses achieved in patients with CML-CP who are switched to nilotinib after long-term imatinib. Blood. 2014; 124(5): 729–736, doi: 10.1182/blood-2013-12-544015, indexed in Pubmed: 24948656.
  40. Verma D, Kantarjian H, Jain N, et al. Sustained complete molecular response after imatinib discontinuation in a patient with chronic myeloid leukemia not previously exposed to interferon alpha. Leuk Lymphoma. 2008; 49(7): 1399–1402, doi: 10.1080/10428190802043903, indexed in Pubmed: 18452066.
  41. Mahon FX, Réa D, Guilhot J, et al. Intergroupe Français des Leucémies Myéloïdes Chroniques. 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(11): 1029–1035, doi: 10.1016/S1470-2045(10)70233-3, indexed in Pubmed: 20965785.
  42. Etienne G, Guilhot J, Rea D, et al. Long-term follow-up of the French Stop Imatinib (STIM1) study in patients with chronic myeloid leukemia. J Clin Oncol. 2017; 35(3): 298–305, doi: 10.1200/JCO.2016.68.2914, indexed in Pubmed: 28095277.
  43. Saussele S, Richter J, Guilhot J, et al. EURO-SKI investigators. Discontinuation of tyrosine kinase inhibitor therapy in chronic myeloid leukaemia (EURO-SKI): a prespecified interim analysis of a prospective, multicentre, non-randomised, trial. Lancet Oncol. 2018; 19(6): 747–757, doi: 10.1016/S1470-2045(18)30192-X, indexed in Pubmed: 29735299.
  44. Atallah E, Schiffer CA, Radich JP, et al. Assessment of outcomes after stopping tyrosine kinase inhibitors among patients with chronic myeloid leukemia: a nonrandomized clinical trial. JAMA Oncol. 2021; 7(1): 42–50, doi: 10.1001/jamaoncol.2020.5774, indexed in Pubmed: 33180106.
  45. Hughes TP, Ross DM. Moving treatment-free remission into mainstream clinical practice in CML. Blood. 2016; 128(1): 17–23, doi: 10.1182/blood-2016-01-694265, indexed in Pubmed: 27013442.
  46. Baxter E, Scott L, Campbell P, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005; 365(9464): 1054–1061, doi: 10.1016/S0140-6736(05)71142-9, indexed in Pubmed: 15781101.
  47. Rea D, Ame S, Berger M, et al. French Chronic Myeloid Leukemia Study Group. Discontinuation of tyrosine kinase inhibitors in chronic myeloid leukemia: Recommendations for clinical practice from the French Chronic Myeloid Leukemia Study Group. Cancer. 2018; 124(14): 2956–2963, doi: 10.1002/cncr.31411, indexed in Pubmed: 29723417.
  48. Hochhaus A, Baccarani M, Silver RT, et al. European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia. 2020; 34(4): 966–984, doi: 10.1038/s41375-020-0776-2, indexed in Pubmed: 32127639.
  49. Radich JP, Deininger M, Abboud CN, et al. Chronic Myeloid Leukemia, Version 1.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2018; 16(9): 1108–1135, doi: 10.6004/jnccn. 2018.0071, indexed in Pubmed: 30181422.
  50. Rea D, Nicolini FE, Tulliez M, et al. France Intergroupe des Leucémies Myéloïdes Chroniques. Discontinuation of dasatinib or nilotinib in chronic myeloid leukemia: interim analysis of the STOP 2G-TKI study. Blood. 2017; 129(7): 846–854, doi: 10.1182/blood-2016-09-742205, indexed in Pubmed: 27932374.
  51. Adnan Awad S, Kankainen M, Ojala T, et al. Mutation accumulation in cancer genes relates to nonoptimal outcome in chronic myeloid leukemia. Blood Adv. 2020; 4(3): 546–559, doi: 10.1182/bloodadvances.2019000943, indexed in Pubmed: 32045476.
  52. Branford S, Wang P, Yeung DT, et al. Integrative genomic analysis reveals cancer-associated mutations at diagnosis of CML in patients with high-risk disease. Blood. 2018; 132(9): 948–961, doi: 10.1182/blood-2018-02-832253, indexed in Pubmed: 29967129.
  53. Branford S, Kim DD, Apperley JF, et al. International CML Foundation Genomics Alliance. Laying the foundation for genomically-based risk assessment in chronic myeloid leukemia. Leukemia. 2019; 33(8): 1835–1850, doi: 10.1038/s41375-019-0512-y, indexed in Pubmed: 31209280.
  54. NCCN Guidelines Version 3.2022 Chronic Myeloid Leukemia Continue NCCN Guidelines Panel Disclosures. https://www.thd.org.tr/thdData/userfiles/file/NCCN-CML-2022-v2.pdf (February 1, 2022).
  55. Mahon FX, Boquimpani C, Kim DW, et al. Treatment-free remission after second-line nilotinib treatment in patients with chronic myeloid leukemia in chronic phase: results from a single-group, phase 2, open-label study. Ann Intern Med. 2018; 168(7): 461–470, doi: 10.7326/M17-1094, indexed in Pubmed: 29459949.
  56. Okada M, Imagawa J, Tanaka H, et al. DADI Trial Group, Japan. Final 3-year results of the dasatinib discontinuation trial in patients with chronic myeloid leukemia who received dasatinib as a second-line treatment. Clin Lymphoma Myeloma Leuk. 2018; 18(5): 353–360.e1, doi: 10.1016/j.clml.2018.03.004, indexed in Pubmed: 29610029.
  57. Ross DM, Masszi T, Gómez Casares MT, et al. Durable treatment-free remission in patients with chronic myeloid leukemia in chronic phase following frontline nilotinib: 96-week update of the ENESTfreedom study. J Cancer Res Clin Oncol. 2018; 144(5): 945–954, doi: 10.1007/s00432-018-2604-x, indexed in Pubmed: 29468438.
  58. Takahashi N, Nishiwaki K, Nakaseko C, et al. STAT study group. Treatment-free remission after two-year consolidation therapy with nilotinib in patients with chronic myeloid leukemia: STAT2 trial in Japan. Haematologica. 2018; 103(11): 1835–1842, doi: 10.3324/haematol.2018.194894, indexed in Pubmed: 29976734.
  59. Di Q, Deng H, Zhao Y, et al. Second-generation tyrosine kinase inhibitor discontinuation in chronic myeloid leukemia patients with stable deep molecular response: a systematic review and a meta-analysis. Comput Math Methods Med. 2021; 2021: 3110622, doi: 10.1155/2021/3110622, indexed in Pubmed: 34956393.
  60. Legros L, Nicolini FE, Etienne G, et al. French Intergroup for Chronic Myeloid Leukemias. Second tyrosine kinase inhibitor discontinuation attempt in patients with chronic myeloid leukemia. Cancer. 2017; 123(22): 4403–4410, doi: 10.1002/cncr.30885, indexed in Pubmed: 28743166.
  61. Cross NCP, White HE, Müller MC, et al. Standardized definitions of molecular response in chronic myeloid leukemia. Leukemia. 2012; 26(10): 2172–2175, doi: 10.1038/leu.2012.104, indexed in Pubmed: 22504141.
  62. Pagani IS, Spinelli O, Mattarucchi E, et al. Genomic quantitative real-time PCR proves residual disease positivity in more than 30% samples with negative mRNA-based qRT-PCR in Chronic Myeloid Leukemia. Oncoscience. 2014; 1(7): 510–521, doi: 10.18632/oncoscience.65, indexed in Pubmed: 25594053.
  63. Hinterbrandner M, Rubino V, Stoll C, et al. Tnfrsf4-expressing regulatory T cells promote immune escape of chronic myeloid leukemia stem cells. JCI Insight. 2021; 6(23), doi: 10.1172/jci.insight.151797, indexed in Pubmed: 34727093.
  64. Chomel JC, Bonnet ML, Sorel N, et al. Leukemic stem cell persistence in chronic myeloid leukemia patients in deep molecular response induced by tyrosine kinase inhibitors and the impact of therapy discontinuation. Oncotarget. 2016; 7(23): 35293–35301, doi: 10.18632/oncotarget.9182, indexed in Pubmed: 27167108.
  65. Corbin AS, Agarwal A, Loriaux M, et al. Human chronic myeloid leukemia stem cells are insensitive to imatinib despite inhibition of BCR-ABL activity. J Clin Invest. 2011; 121(1): 396–409, doi: 10.1172/JCI35721, indexed in Pubmed: 21157039.
  66. Patel AB, O’Hare T, Deininger MW. Mechanisms of resistance to ABL kinase inhibition in chronic myeloid leukemia and the development of next generation ABL kinase inhibitors. Hematol Oncol Clin North Am. 2017; 31(4): 589–612, doi: 10.1016/j.hoc.2017.04.007, indexed in Pubmed: 28673390.
  67. Hughes A, Yong ASM. Immune effector recovery in chronic myeloid leukemia and treatment-free remission. Front Immunol. 2017; 8: 469, doi: 10.3389/fimmu.2017.00469, indexed in Pubmed: 28484463.
  68. Cheng JN, Yuan YX, Zhu Bo, et al. Myeloid-derived suppressor cells: a multifaceted accomplice in tumor progression. Front Cell Dev Biol. 2021; 9: 740827, doi: 10.3389/fcell.2021.740827, indexed in Pub­med: 35004667.
  69. Hughes A, Clarson J, Tang C, et al. CML patients with deep molecular responses to TKI have restored immune effectors and decreased PD-1 and immune suppressors. Blood. 2017; 129(9): 1166–1176, doi: 10.1182/blood-2016-10-745992, indexed in Pubmed: 28049640.
  70. Mumprecht S, Schürch C, Schwaller J, et al. Programmed death 1 signaling on chronic myeloid leukemia-specific T cells results in T-cell exhaustion and disease progression. Blood. 2009; 114(8): 1528–1536, doi: 10.1182/blood-2008-09-179697, indexed in Pubmed: 19420358.
  71. Christiansson L, Söderlund S, Svensson E, et al. Increased level of myeloid-derived suppressor cells, programmed death receptor ligand 1/programmed death receptor 1, and soluble CD25 in Sokal high risk chronic myeloid leukemia. PLoS One. 2013; 8(1): e55818, doi: 10.1371/journal.pone.0055818, indexed in Pubmed: 23383287.
  72. Steegmann JL, Moreno G, Aláez C, et al. Chronic myeloid leukemia patients resistant to or intolerant of interferon alpha and subsequently treated with imatinib show reduced immunoglobulin levels and hypogammaglobulinemia. Haematologica. 2003; 88(7): 762–768, indexed in Pubmed: 12857554.
  73. Cwynarski K, Laylor R, Macchiarulo E, et al. Imatinib inhibits the activation and proliferation of normal T lymphocytes in vitro. Leukemia. 2004; 18(8): 1332–1339, doi: 10.1038/sj.leu.2403401, indexed in Pubmed: 15190258.
  74. Rajala HLM, Missiry MEl, Ruusila A, et al. Tyrosine kinase inhibitor therapy-induced changes in humoral immunity in patients with chronic myeloid leukemia. J Cancer Res Clin Oncol. 2017; 143(8): 1543–1554, doi: 10.1007/s00432-017-2378-6, indexed in Pubmed: 28337541.
  75. Marinelli Busilacchi E, Costantini A, Viola N, et al. Immunomodulatory effects of tyrosine kinase inhibitor in vitro and in vivo study. Biol Blood Marrow Transplant. 2018; 24(2): 267–275, doi: 10.1016/j.bbmt.2017.10.039, indexed in Pubmed: 29128554.
  76. de Lavallade H, Khoder A, Hart M, et al. Tyrosine kinase inhibitors impair B-cell immune responses in CML through off-target inhibition of kinases important for cell signaling. Blood. 2013; 122(2): 227–238, doi: 10.1182/blood-2012-11-465039, indexed in Pubmed: 23719297.
  77. Hantschel O, Rix U, Superti-Furga G. Target spectrum of the BCR-ABL inhibitors imatinib, nilotinib and dasatinib. Leuk Lymphoma. 2008; 49(4): 615–619, doi: 10.1080/10428190801896103, indexed in Pubmed: 18398720.
  78. Remsing Rix LL, Rix U, Colinge J, et al. Global target profile of the kinase inhibitor bosutinib in primary chronic myeloid leukemia cells. Leukemia. 2009; 23(3): 477–485, doi: 10.1038/leu.2008.334, indexed in Pubmed: 19039322.
  79. Salih J, Hilpert J, Placke T, et al. The BCR/ABL-inhibitors imatinib, nilotinib and dasatinib differentially affect NK cell reactivity. Int J Can­cer. 2010; 127(9): 2119–2128, doi: 10.1002/ijc.25233, indexed in Pubmed: 20143399.
  80. Chen J, Schmitt A, Chen B, et al. Nilotinib hampers the proliferation and function of CD8+ T lymphocytes through inhibition of T cell receptor signalling. J Cell Mol Med. 2008; 12(5B): 2107–2118, doi: 10.1111/j.1582-4934.2008.00234.x, indexed in Pubmed: 18194453.
  81. Powers JJ, Dubovsky JA, Epling-Burnette PK, et al. A molecular and functional analysis of large granular lymphocyte expansions in patients with chronic myelogenous leukemia treated with tyrosine kinase inhibitors. Leuk Lymphoma. 2011; 52(4): 668–679, doi: 10.3109/10428194.2010.550074, indexed in Pubmed: 21271862.
  82. Mustjoki S, Ekblom M, Arstila TP, et al. Clonal expansion of T/NK-cells during tyrosine kinase inhibitor dasatinib therapy. Leukemia. 2009; 23(8): 1398–1405, doi: 10.1038/leu.2009.46, indexed in Pubmed: 19295545.
  83. Bantscheff M, Eberhard D, Abraham Y, et al. Quantitative chemical proteomics reveals mechanisms of action of clinical ABL kinase inhibitors. Nat Biotechnol. 2007; 25(9): 1035–1044, doi: 10.1038/nbt1328, indexed in Pubmed: 17721511.
  84. Giallongo C, Parrinello N, Tibullo D, et al. Myeloid derived suppressor cells (MDSCs) are increased and exert immunosuppressive activity together with polymorphonuclear leukocytes (PMNs) in chronic myeloid leukemia patients. PLoS One. 2014; 9(7): e101848, doi: 10.1371/journal.pone.0101848, indexed in Pubmed: 25014230.
  85. Rea D, Henry G, Khaznadar Z, et al. Natural killer-cell counts are associated with molecular relapse-free survival after imatinib dis­continuation in chronic myeloid leukemia: the IMMUNOSTIM study. Haematologica. 2017; 102(8): 1368–1377, doi: 10.3324/haematol.2017.165001.
  86. Jacomet F, Cayssials E, Barbarin A, et al. The hypothesis of the human iNKT/innate CD8(+) T-cell axis applied to cancer: evidence for a deficiency in chronic myeloid leukemia. Front Immunol. 2016; 7: 688, doi: 10.3389/fimmu.2016.00688, indexed in Pubmed: 28138330.
  87. Cayssials E, Jacomet F, Piccirilli N, et al. Sustained treatment-free remission in chronic myeloid leukaemia is associated with an increased frequency of innate CD8(+) T-cells. Br J Haematol. 2019; 186(1): 54–59, doi: 10.1111/bjh.15858, indexed in Pubmed: 30864168.
  88. Matsushita M, Ozawa K, Suzuki T, et al. CXorf48 is a potential therapeutic target for achieving treatment-free remission in CML patients. Blood Cancer J. 2017; 7(9): e601, doi: 10.1038/bcj.2017.84, indexed in Pubmed: 28862699.
  89. Ito K, Ito K. Leukemia stem cells as a potential target to achieve therapy-free remission in chronic myeloid leukemia. Cancers (Basel). 2021; 13(22), doi: 10.3390/cancers13225822, indexed in Pubmed: 34830976.
  90. Eisterer W, Jiang X, Christ O, et al. Different subsets of primary chronic myeloid leukemia stem cells engraft immunodeficient mice and produce a model of the human disease. Leukemia. 2005; 19(3): 435–441, doi: 10.1038/sj.leu.2403649, indexed in Pubmed: 15674418.
  91. Sadovnik I, Hoelbl-Kovacic A, Herrmann H, et al. Identification of CD25 as STAT5-dependent growth regulator of leukemic stem cells in Ph+ CML. Clin Cancer Res. 2016; 22(8): 2051–2061, doi: 10.1158/1078-0432.CCR-15-0767, indexed in Pubmed: 26607600.
  92. Florian S, Sonneck K, Hauswirth AW, et al. Detection of molecular targets on the surface of CD34+/CD38- — stem cells in various myeloid malignancies. Leuk Lymphoma. 2006; 47(2): 207–222, doi: 10.1080/10428190500272507, indexed in Pubmed: 16321850.
  93. Zhang B, Chu Su, Agarwal P, et al. Inhibition of interleukin-1 signaling enhances elimination of tyrosine kinase inhibitor-treated CML stem cells. Blood. 2016; 128(23): 2671–2682, doi: 10.1182/blood-2015-11-679928, indexed in Pubmed: 27621307.
  94. Landberg N, Hansen N, Askmyr M, et al. IL1RAP expression as a measure of leukemic stem cell burden at diagnosis of chronic myeloid leukemia predicts therapy outcome. Leukemia. 2016; 30(1): 253–257, doi: 10.1038/leu.2015.135, indexed in Pubmed: 26067823.
  95. Bocchia M, Sicuranza A, Abruzzese E, et al. Residual peripheral blood CD26 leukemic stem cells in chronic myeloid leukemia patients during TKI therapy and during treatment-free remission. Front Oncol. 2018; 8: 194, doi: 10.3389/fonc.2018.00194, indexed in Pubmed: 29900128.
  96. Hamilton A, Helgason GV, Schemionek M, et al. Chronic myeloid leukemia stem cells are not dependent on Bcr-Abl kinase activity for their survival. Blood. 2012; 119(6): 1501–1510, doi: 10.1182/blood-2010-12-326843, indexed in Pubmed: 22184410.
  97. Welner RS, Amabile G, Bararia D, et al. Treatment of chronic myelogenous leukemia by blocking cytokine alterations found in normal stem and progenitor cells. Cancer Cell. 2015; 27(5): 671–681, doi: 10.1016/j.ccell.2015.04.004, indexed in Pubmed: 25965572.
  98. Zhao C, Blum J, Chen A, et al. Loss of beta-catenin impairs the renewal of normal and CML stem cells in vivo. Cancer Cell. 2007; 12(6): 528–541, doi: 10.1016/j.ccr.2007.11.003, indexed in Pubmed: 18068630.
  99. Zhao C, Chen A, Jamieson CH, et al. Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia. Nature. 2009; 458(7239): 776–779, doi: 10.1038/nature07737, indexed in Pubmed: 19169242.
  100. Aljedai A, Buckle AM, Hiwarkar P, et al. Potential role of Notch signalling in CD34+ chronic myeloid leukaemia cells: cross-talk between Notch and BCR-ABL. PLoS One. 2015; 10(4): e0123016, doi: 10.1371/journal.pone.0123016, indexed in Pubmed: 25849484.
  101. Naka K, Hoshii T, Muraguchi T, et al. TGF-beta-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia. Nature. 2010; 463(7281): 676–680, doi: 10.1038/nature08734, indexed in Pubmed: 20130650.
  102. Xie X, Feng M, Wang Q, et al. Cellular and molecular state of myeloid leukemia stem cells. Adv Exp Med Biol. 2019; 1143: 41–57, doi: 10.1007/978-981-13-7342-8_2, indexed in Pubmed: 31338814.
  103. Mojtahedi H, Yazdanpanah N, Rezaei N. Chronic myeloid leukemia stem cells: targeting therapeutic implications. Stem Cell Res Ther. 2021; 12(1): 603, doi: 10.1186/s13287-021-02659-1, indexed in Pubmed: 34922630.
  104. Nishimoto Y, Okano H. New insight into cancer therapeutics: induction of differentiation by regulating the Musashi/Numb/Notch pathway. Cell Res. 2010; 20(10): 1083–1085, doi: 10.1038/cr.2010.122, indexed in Pubmed: 20805843.
  105. Ito T, Kwon HY, Zimdahl B, et al. Regulation of myeloid leukaemia by the cell-fate determinant Musashi. Nature. 2010; 466(7307): 765––768, doi: 10.1038/nature09171, indexed in Pubmed: 20639863.
  106. Lundell BI, McCarthy JB, Kovach NL, et al. Activation of beta1 integrins on CML progenitors reveals cooperation between beta1 integrins and CD44 in the regulation of adhesion and proliferation. Leukemia. 1997; 11(6): 822–829, doi: 10.1038/sj.leu.2400653, indexed in Pubmed: 9177435.
  107. von Palffy S, Landberg N, Sandén C, et al. A high-content cytokine screen identifies myostatin propeptide as a positive regulator of primitive chronic myeloid leukemia cells. Haematologica. 2020; 105(8): 2095–2104, doi: 10.3324/haematol.2019.220434, indexed in Pubmed: 31582541.
  108. Baba T, Naka K, Morishita S, et al. MIP-1α/CCL3-mediated maintenance of leukemia-initiating cells in the initiation process of chronic myeloid leukemia. J Exp Med. 2013; 210(12): 2661–2673, doi: 10.1084/jem.20130112, indexed in Pubmed: 24166712.
  109. Agarwal P, Isringhausen S, Li H, et al. Mesenchymal niche-specific expression of Cxcl12 controls quiescence of treatment-resistant leukemia stem cells. Cell Stem Cell. 2019; 24(5): 769–784.e6, doi: 10.1016/j.stem.2019.02.018, indexed in Pubmed: 30905620.
  110. Zhang B, Nguyen LeX, Li L, et al. Bone marrow niche trafficking of miR-126 controls the self-renewal of leukemia stem cells in chronic myelogenous leukemia. Nat Med. 2018; 24(4): 450–462, doi: 10.1038/nm.4499, indexed in Pubmed: 29505034.
  111. Silvestri G, Trotta R, Stramucci L, et al. Persistence of drug-resistant leukemic stem cells and impaired NK cell immunity in CML patients depend on antiproliferative and PP2A-activating functions. Blood Cancer Discov. 2020; 1(1): 48–67, doi: 10.1158/0008-5472.BCD-19-0039, indexed in Pubmed: 32974613.
  112. Soverini S, De Santis S, Monaldi C, et al. Targeting leukemic stem cells in chronic myeloid leukemia: is it worth the effort? Int J Mol Sci. 2021; 22(13), doi: 10.3390/ijms22137093, indexed in Pubmed: 34209376.
  113. Warda W, Larosa F, Neto Da Rocha M, et al. CML hematopoietic stem cells expressing IL1RAP can be targeted by chimeric antigen receptor-engineered T cells. Cancer Res. 2019; 79(3): 663–675, doi: 10.1158/0008-5472.CAN-18-1078, indexed in Pubmed: 30514753.
  114. Landberg N, von Palffy S, Askmyr M, et al. CD36 defines primitive chronic myeloid leukemia cells less responsive to imatinib but vulnerable to antibody-based therapeutic targeting. Haematologica. 2018; 103(3): 447–455, doi: 10.3324/haematol.2017.169946, indexed in Pubmed: 29284680.
  115. Järås M, Johnels P, Hansen N, et al. Isolation and killing of candidate chronic myeloid leukemia stem cells by antibody targeting of IL-1 receptor accessory protein. Proc Natl Acad Sci USA. 2010; 107(37): 16280–16285, doi: 10.1073/pnas.1004408107, indexed in Pubmed: 20805474.
  116. Houshmand M, Garello F, Stefania R, et al. Targeting chronic myeloid leukemia stem/progenitor cells using venetoclax-loaded immunoliposome. Cancers (Basel). 2021; 13(6), doi: 10.3390/cancers13061311, indexed in Pubmed: 33804056.
  117. Krishnan V, Kim DD, Hughes TP, et al. Integrating genetic and epigenetic factors in chronic myeloid leukemia risk assessment: toward gene expression-based biomarkers. Haematologica. 2022; 107(2): 358–370, doi: 10.3324/haematol.2021.279317, indexed in Pubmed: 34615339.
  118. Rinke J, Hochhaus A, Ernst T. CML — not only BCR-ABL1 matters. Best Pract Res Clin Haematol. 2020; 33(3): 101194, doi: 10.1016/j.beha.2020.101194, indexed in Pubmed: 33038988.