Next Article in Journal
Therapeutic Potential of Magnetic Nanoparticle-Based Human Adipose-Derived Stem Cells in a Mouse Model of Parkinson’s Disease
Next Article in Special Issue
Sex-Related Differences in Chronic Myeloid Neoplasms: From the Clinical Observation to the Underlying Biology
Previous Article in Journal
QTL Mapping and Candidate Gene Identification of Swollen Root Formation in Turnip
Previous Article in Special Issue
NPM1-Mutated Myeloid Neoplasms with <20% Blasts: A Really Distinct Clinico-Pathologic Entity?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Abnormal Hematopoietic Stem Cells in Chronic Myeloid Leukemia and Philadelphia Chromosome-Negative Classical Myeloproliferative Neoplasms

Division of Haematology, Medical Oncology and Haemopoietic Stem Cell Transplantation, Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(2), 659; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22020659
Submission received: 9 December 2020 / Revised: 5 January 2021 / Accepted: 6 January 2021 / Published: 11 January 2021
(This article belongs to the Special Issue Stem Cell Biology of Myeloid Neoplasms)

Abstract

:
Myeloproliferative neoplasms (MPNs) are unique hematopoietic stem cell disorders sharing mutations that constitutively activate the signal-transduction pathways involved in haematopoiesis. They are characterized by stem cell-derived clonal myeloproliferation. The key MPNs comprise chronic myeloid leukemia (CML), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). CML is defined by the presence of the Philadelphia (Ph) chromosome and BCR-ABL1 fusion gene. Despite effective cytoreductive agents and targeted therapy, complete CML/MPN stem cell eradication is rarely achieved. In this review article, we discuss the novel agents and combination therapy that can potentially abnormal hematopoietic stem cells in CML and MPNs and the CML/MPN stem cell-sustaining bone marrow microenvironment.

Graphical Abstract

1. Introduction

Myeloproliferative neoplasms (MPN) are a collection of clonal hematopoietic stem cell disorders characterized by the proliferation of one of more of the hematopoietic lineages [1,2,3,4]. The major clinicopathologic entities comprise chronic myeloid leukemia (CML), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF) [5,6]. CML is defined by the presence of the Philadelphia (Ph) chromosome that results from t(9;22) (q34.1;q11.2) and formation of the constitutively expressed oncoprotein BCR-ABL1 [7,8]. Philadelphia chromosome-negative myeloproliferative neoplasms (Ph-negative MPNs) arise from a single clonal hematopoietic stem cell (HSC) leading to proliferation of more than one cell lineage, with transitional forms from one entity to another. Over 95% of PV, ET, and PMF are associated with mutually exclusive somatic driver mutations JAK2V617F, calreticulin (CALR), and myeloproliferative leukemia protein (MPL) [9,10,11,12,13]. Acquisition of somatic driver mutations leads to the development of the MPN stem cells. In addition, disease initiation and progression of Ph-negative MPNs involve the interplay between cell-intrinsic and cell-extrinsic activities. This is characterized by survival advantage of MPN stem cells over normal HSCs that is sustained by a dysregulated bone marrow niche via a positive feedback mechanism [9,14] (Figure 1). Further acquisition of non-driver mutations then plays a pivotal role in determining disease phenotype and promoting leukemic progression [9,10,13] (Figure 2). At diagnosis, all CML patients harbor abnormal HSCs [8,15,16]. They are characterized by an unlimited potential and unrestricted ability to self-regenerate, remain quiescent, mediate BCR-ABL1-independent tyrosine kinase inhibitor (TKI)-resistance and evade the host immunity, and allowing disease initiation, development, maintenance and progression [8,15,17,18,19,20,21,22]. The abnormal HSCs in CML are able to survive and thrive through various mechanisms such as modulation of downstream signaling pathways (e.g., JAK/STAT, PI3K/AKT/mTOR, Wnt/β-catenin, Hedgehog signalling), induction of autophagy, selective advantage in homing and engraftment in the bone marrow microenvironment (BMM), and alterations in cellular metabolism [8,15,23,24,25,26,27,28,29]. While leukemia stem cells (LCSs) are generally CD34+/CD38, the abnormal HSC populations in CML have extremely heterogeneous and unstable cell surface antigens expression, and vary greatly in terms of their leukemogenic capacity [8,17,21,30,31]. Although TKIs show some efficacy in targeting abnormal HSCs in CML, they may not be adequate for disease eradication.
The persistence of abnormal HSCs in CML and MPN has led to the development of novel therapies targeting CML or MPN stem cells. In this review, we discuss the current and emerging therapeutic options that may target CML and MPN stem cells with the aim of disease modification and eradication. The various pathways involved in the biology of CML and Ph-negative MPNs are depicted in Supplementary Materials Files S2–S7, highlighting the rationale of their therapeutic targeting.

2. Current Therapeutic Options in CML and Their Effects on CML Stem Cells

TKIs competitively bind to the ATP-binding site of the BCR-ABL1 to reduce abnormal phosphorylation of the dysregulated tyrosine kinase and inhibit downstream pathways and leukaemogenesis [7,32,33]. TKIs (Table 1) have modest effects on CML stem cells as single agents may be rendered ineffective in targeting CML stem cells as a result of BCR-ABL1-independent mechanisms. They may arise from mutations in epigenetic regulators (e.g., DNMT3A, EZH2, IDH1/2) [7,34] or the loss of tumour suppressor genes (e.g., TP53, PTEN, TET1/2) [17,29] and genes that code for anti-oxidant systems (e.g., FoxO, EPAS1) [17]. Quiescence of CML stem cells is another major challenge, and may contribute to TKI resistance and relapse in CML. Despite the fact that 50% of CML patients achieve treatment-free remissions without relapse after achieving deep molecular response, most harbour residual CML stem cells [15,17,35].

2.1. First and Second Generation TKIs

Imatinib (IM), a first generation TKI, is highly effective in inducing apoptosis in BCR-ABL1-positive cells. CML stem cells have dysregulated intracellular calcium signalling, uncontrolled pro-inflammatory cytokines such as interleukin-6 (IL-6), IL-8 and nuclear factor kappa beta (NF-κB), and overproduction of activator protein 1 (AP-1), which are hallmarks for CML stem cell survival [29,39,40,41]. In-vivo and in vitro studies showed that these could be reversed by IM antagonizing inositol-triphosphate (IP3)-mediated calcium mobilization (p < 0.05) and oxidative stress via IP3 receptor inhibition (IP3R) on the endoplasmic reticulum (ER) [39,40,42]. Herrmann et al. demonstrated a decrease in CD26+ stem cells after in vitro IM therapy [16], yet Willmann et al. showed otherwise [36]. Moreover, nilotinib induced CML stem cell apoptosis [36], and nilotinib and dasatinib showed higher potency in IP3R inhibition [42]. IM may also downregulate overexpressed EZH2 in CML stem cells, with minimal effects in normal HSCs [17,43]. Also, in-vitro studies showed that post-dasatinib or -IM therapy, programmed death receptor 1 (PD-1, immune marker for immune-evasion) expression was found to be reduced on CD8+ T cells and monocytic myeloid-derived suppressor cells (MDSCs), leading to increased cytotoxic T-lymphocyte- (CTL) and Natural Killer (NK) cell-mediated cytotoxicity [18,44,45,46]. However, contradicting evidence was presented in another in vitro study, which showed that IM enhanced mRNA and protein expression of autophagy-related 4B cysteine peptidase (Atg4B), resulting in TKI-induced autophagy and selective survival in CD34+ CML cells (p ≤ 0.05) [39].

2.2. Ponatinib

Ponatinib, a third generation TKI, is indicated in CML with BCR-ABL1T315I mutations or refractoriness to ≥2 TKIs. Steric hindrance is produced due to replacement of threonine by isoleucine at the ATP-binding site [47,48]. Presence of a carbon-carbon triple bond in ponatinib allows it to be 500-fold more potent than IM in overcoming TKI-resistance [22,47,48,49]. Other pathways targeted by ponatinib include VEGFR, KIT, SRC, FGFR, PDGFR, FLT3, and KIT [47]. In-vivo murine models with CML stem cells that are linSca-1+c-kit+ showed that ponatinib was effective in CML stem cell eradication and spleen size reduction. Thirty-percent residual BCR-ABL1 chimerism at 28 days was achieved compared to >50% in dasatinib and IM [22,47,50].

2.3. Asciminib

Asciminib (ABL001), a recent, FDA-approved, fourth generation TKI, is an allosteric inhibitor that binds to the BCR-ABL1 myristoyl-pocket (STAMP) [8,33,49,51,52]. It is effective against BCR-ABL1 KD-dependent and -independent mutations as monotherapy or in combination with other TKIs to restore TKI-sensitivity in resistant cell lines and produce drug synergism in reducing CRK-like protein (CRKL) phosphorylation for CML stem cells [49,53]. Initial results in a phase I trial (ClinicalTrials.gov number, NCT03595917) demonstrated that 82% of patients with TKI-resistance achieved major cytogenetic response (MCyR) by 3 months and 30% of patients reached CCyR at 5 months [51]. In the phase III ASCEBEL trial, asciminib showed superiority over bosutinib in achieving MMR at 24 weeks [53]. Ongoing trials using asciminib as monotherapy, in combination with other TKIs and/or corticosteroids are underway (ClinicalTrials.gov identifiers: NCT04216563, NCT03906292, NCT04360005, NCT03106779, NCT03595917, NCT03578367 and NCT02081378).

2.4. Interferon-α

IFNα was used as first-line treatment before the emergence of TKIs. It induces apoptosis of LSCs via Fas-receptors upregulation, FADD/caspase-8 pathway activation, and cytochrome-c release, leading to mitochondrial disruption and cellular apoptosis independent of anti-apoptotic B-cell lymphoma 2 (Bcl-2), cell-cycle arrest and tumour-suppressor p53 [54,55,56]. IFNα also restores normal function of the dysregulated BMM through β1-integrin for cellular differentiation and elimination of the protective barrier established for LSC quiescence [54,57,58]. IFN-α-mediated increase in expression of major histocompatibility complex (MHC) class I molecules and tumour-associated antigens cause reactivation of CTL and prompt CTL-mediated cytotoxicity against LSCs [54,55]. The 5-year survival rate of IFNα was 57% as shown in a meta-analysis of 7 data sets of randomized trials consisting of 1,554 patients [54,59]. In another study using IFNα monotherapy, the 10-year survival rate was 72%, where 46% remained in CCyR [55,60]. These highlight the potential re-emergence of IFNα for LSC elimination, where clinical trials using IFNα alone or in combination with other TKIs showed promising results for TFR (ClinicalTrials.gov identifiers: NCT02001818, NCT01657604, NCT03117816, NCT03831776, NCT04126681, NCT01316250, NCT02381379, and NCT00452023).

3. Current Therapeutic Options in MPN and Their Effects on MPN Stem Cells

3.1. IFNα

A major significance of Peg-IFNα-2a is its ability to target MPN stem cells and reduce mutant allele burden in MPN [61,62,63,64,65,66,67,68]. Sustained molecular, haematological response and regression of BM fibrosis were seen in some patients after discontinuation of Peg-IFNα-2a, indicating the eradication of MPN stem cells [65,69] (Table 2). Interestingly, the effect of Peg-IFNα-2a on JAK2V617F+ stem cells was greater than that on CALR-mutated stem cells, with no difference in hematological response [70,71]. This is due to the phosphorylation and activation of JAK1-STAT1 pathway in JAK2V617F cells, but not in CALR-mutated cells, resulting in JAK2V617F-positive cells priming towards Peg-IFNα-2a [70]. There is a paucity of data suggesting that Peg-IFNα-2a targeting MPL-mutated clones could be due to the low frequency of MPL mutations in MPN.

3.2. JAK Inhibitors

Treatment with ruxolitinib showed some reduction in JAK2V617F mutant allele burden in PV patients [74,75], although its effect on reduction of mutant allele burden in MF is mild [76]. Additionally, several second generation JAK inhibitors were developed to improve efficacy and reduce side effects of ruxolitinib [77,78,79,80,81]. However, none of them show significance in eradicating LSCs [77,79,80,82,83]. Some studies have shown that fedratinib, a newly FDA approved JAK2/FLT3 inhibitor in 2019, reduced JAK2V617F variant allele frequency (VAF) [79,84]. Yet, the results were not consistent with other trials [79,83,85]. Therefore, combinations of various novel therapies with ruxolitinib emerge in the hope of eliminating MPN LSCs [86].

3.3. Allogeneic HSCT

Allo-HSCT is able to overcome high molecular risks (HMR) mutations, and its recipients usually harbor additional molecular mutations. These additional molecular mutations, including those conferring a poor prognosis in MPN (ASXL1, EZH2, SRSF2, IDH1/2, TP53) generally do not affect relapse-free survival (RFS) and OS in patients receiving allo-HSCT [87]. However, discrepancies are seen in terms of relapse risks. The ASXL1 mutation, which accounts for >90% in intermediate-2 and high-risk MF patients, is found to be associated with higher relapse risks [88]. In a study assessing the outcome of allo-HSCT in MPL-mutated PMF and secondary myelofibrosis (SMF), the only relapsed patient harbored ASXL1 and EZH2 mutation [89]. Meanwhile, some post-transplant ASXL1-mutated patients may die without relapsing [87].

4. Novel Therapy Targeting Signaling Pathways in CML Stem Cells

4.1. Novel Tyrosine Kinase Inhibitors

PF-114 is an orally bioavailable fourth generation TKI that is selectively active against BCR-ABL1-dependent and -independent mutations, as well as non-mutational TKI-resistant cell lines [8,90,91,92]. It acts as a BCR-ABL1 KD antagonist and/or STAMP inhibitor, suppressing the constitutively active PI3K/AKT/ERK1/2 and JAK/STAT3/5 signalling, elevating p27 levels, leading to G1 cell cycle arrest [91,93]. It induced apoptosis in patient-derived K562 and KCL-22 cell lines [91]. In K562 nude mouse xenograft, it caused complete eradication of the tumour bulk (p < 0.001) without recurrence [90]. It also showed excellent toxicity profile as it spared VEGFR, FLT3, EPHRIN, FGFR and B-RAF, implying less cardiac, pulmonary and vascular complications [90], hence making it a promising agent for patients refractory and/or resistant to frontline therapies. Phase I/II trials showed that 55% of heavily pretreated patients receiving PF-114 300mg daily achieved MCyR and 36% achieved MMR [92].

4.2. Targetting microRNAs

MicroRNAs (miR) are short pleiotropic non-coding RNA sequences that cleave or repress transcription, hence controlling at least 10–40% of human mRNA expression [94,95]. Malfunctioning miR is key to leukemogenesis and TKI-resistance, maintenance and self-regeneration [17,94,95,96]. Overt expression of miR-29a leads to the depletion of tumor suppressor TET2 and antioxidant-coding EPAS1, with upregulation of anti-apoptotic genes Bcl-2 and Mcl-1 apoptosis regulator (Mcl-1) [94,97,98,99]. Downregulation of Bcl-2 inhibitor miR-153-3p is mediated by uncontrolled c-Myc/miR-150 expression in LSC and Ph+ cells, impairing myeloid differentiation and promoting TKI-resistance [100,101]. Prolonged exposure of TKIs triggers drug resistance in LSCs and K562 cells, and is associated with low miR-217 and high DNA methyltransferases (DNMTs) [94,102,103]. miR-424 acts as a direct inhibitor of BCR-ABL1, showing markedly low expression in CD34+/CD38 and BCR-ABL1+ cells alongside high levels of oncoprotein Cobll1 [94,97,104,105]. In vitro studies suggested that miR-217 overexpression might allow restoration of tumour suppressor effects [94,97,104]. Reduced tumor suppressor miR-142 levels in LSCs and TKI-resistant cells were found to be associated with excessive oncoproteins Mcl-1, cKIT, and SRI, precipitating unimpeded PI3K/AKT, JAK/STAT, and RAS/RAF/MEK/ERK downstream signalling with anti-apoptotic, pro-survival and therapy-resistant effects [94,97,106,107,108,109]. Correction of dysregulated miR levels in preclinical studies exhibited propitious results in reduction of tumour bulk. This may not only yield a new field of clinical investigation for targeted therapy, but also as a tool in aiding diagnosis, a prognostic indicator and a predictor of treatment response [110].

4.3. Targeting BCR-ABL1/Gab2/Grb2 Axis

Dysregulated growth factor receptor-bound protein 2 (Grb2) expression permits direct binding to the Src homology (SH2) domain of BCR-ABL1, forming Grb2-SOS complexes and leading to downstream hyperactivation of RAS/MAPK pathway [111,112,113,114]. BP1001, a liposome-incorporated antisense oligodeoxynucleotide targeted against the Grb2 mRNA, inhibits subsequent protein expression and the RAS/MEK/ERK pathway. Preclinical studies showed that Grb2 depletion induced LSC knockdown in CD34+ cells, without affecting normal HSCs and STAT inhibition [111,114]. Combination with TKIs overcomes resistance and induces drug synergism [113]. A phase I clinical trial showed that BP1001 enhanced the effects of dasatinib by 2–6-fold and reduced phosphorylation of ERK1/2 and Grb2 levels by >25% in 52% and 49% patients respectively [113]. Trametinib, a MEK inhibitor in combination with imatinib inhibited the MEK/ERK and NF-κB-mediated LSC survival, restoring TKI sensitivity in-vitro and in-vivo [18,115,116].

4.4. Targeting MAPK/MNK1/2 Pathway

The MAPK/MNK1/2 pathway is amplified in CML stem cells but not in normal HSCs. The constitutive phosphorylation of eukaryotic initiation factor 4E (eIF4E, an oncoprotein essential for LSC proliferation) induces nuclear activation and translocation of β-catenin, contributing to leukemogenesis and TKI-resistance [117]. Preclinical studies showed that ETC-1907206, a selective MNK1/2 inhibitor, suppressed eIF4E phosphorylation and β-catenin signalling [117].

4.5. Targeting mTOR Pathway

mTOR, is a catalytic kinase for the protein complexes mTORC1 and mTORC2 in the PI3K/AKT/mTORC1 pathway [118,119]. It inhibits mRNA translation through initiation factor 4E binding protein (4E-BP1) and p70 ribosomal S6 kinase (p70S6K, S6K) [118,119,120]. Liver kinase B1 (LKB1, tumour suppressor), an upstream kinase of AMP-activated protein kinase (AMPK) induces AMPK phosphorylation of tuberous sclerosis complex 2 (TSC2) to suppress Ras homolog enriched in brain (Rheb) and inhibit mTOR. In LSCs, dysregulated PI3K/AKT/mTOR signalling increases reactive oxygen species (ROS) production, leading to the loss of negative regulation by LKB1 and AMPK, promoting survival, proliferation, drug resistance and stemness [118,121]. Preclinical studies showed that metformin, an AMPK activator inhibited aberrant PI3K/AKT/mTOR hyperactivation to reduce oxidative phosphorylation of glucose and fatty acids, leading to LSC apoptosis [122,123]. However, this led to the compensatory upregulation of glucose and glycolysis which could be be overcome by the addition of 2 deoxy-glucose (2-DG), a hexokinase inhibitor that mimics d-glucose and inhibits glycolysis to induce cell death [123,124]. Combination with TKIs boosts effects and induces TKI-mediated apoptosis [123,124]. Resveratrol, a natural antioxidant found in grapes is also found to stimulate AMPK activation, hence upregulating p38 expression and JNK-mediated phosphorylation of H2AX, downregulating Bcl-2 and triggering caspase-3-mediated LSC apoptosis and cell-cycle arrest [120,125,126]. 5-aminoimidazole-4-carboxamide riboside (AICAR), undergoes phosphorylation and binds to an allosteric site of AMPK to activate it and inhibits mTOR regardless of TKI sensitivity [127].

4.6. Bcl-2 Targeting

Bcl-2, a key anti-apoptotic gene that regulates mitochondrial-mediated apoptosis through the JAK/STAT and PI3K/AKT pathways [8,18,128,129], is overwhelmingly expressed in LSCs. In addition, BCR-ABL1 induces upregulation of Bcl-2 anti-apoptotic proteins, including Mcl-1 and B-cell lymphoma-extra large (Bcl-xL) [8,18,128]. Venetoclax, a Bcl-2 inhibitor, binds to the hydrophobic groove of the Bcl-2 homology 3 (BH3) domain of Bcl-2, releasing its inhibition on Bcl-2-associated X protein (BAX) to drive p53/BAX-mediated programmed cell death [18,128]. Preclinical studies showed synergism between venetoclax and TKI in targeting mitochondrial oxidative phosphorylation to eliminate CD34+/CD38+ and CD34+ cells [18,128]. A retrospective study using venetoclax in combination with TKIs showed 60% complete remission (CR), 75% ORR, median survival of 10.9 months and median RSF of 3.9 months [130].

4.7. JAK2 Inhibition

JAK2 mediates cytokine-mediated signaling in CML cells. It leads to uncontrolled STAT3/5 phosphorylation by directly binding to the SH2 domain of BCR-ABL1, which is stabilized by Abelson helper integration site 1 (AHI-1), an oncogenic adaptor for LSC survival. [18,30,131,132]. In LSCs, induced expression of MPL enhances JAK/STAT signaling to trigger ROS formation and subsequent clonal evolution, contributing to stemness and TKI-resistance [8,18,30,131]. However, LSCs remain sensitive to JAK2 inhibitors such as ruxolitinib. Preclinical studies showed that combining ruxolitinib with the CML-specific TKIs eliminated CD34+/CD38 stem cells with no effects on normal HSCs in-vitro, and reduced CD34+ cell engraftment to the BM in-vivo [131]. Sweet et al. showed that 33% of patients had ≥1 log reduction in BCR-ABL1 transcripts and 44% achieved MR4.5 when co-treated with nilotinib in a phase I trial [133]. Another phase I trial with nilotinib demonstrated that 40% of patients had molecularly undetectable BCR-ABL1 transcripts over 6 months [134]. A phase II trial using ruxolitinib alone showed 60% ORR, where 33% observed clinical benefit in one or more categories: platelet count improvement, hemoglobin improvement, ≥50% reduction in spleen size and ≥50% reduction in symptoms [135]. A phase I/II trial in combining ruxolitinib with CML-specific TKIs showed achievement of CCyR in 87.5% and MMR in 37.5% of patients [136].

4.8. Targeting PPARγ/STAT5/HIF2α Axis

STAT5 activation leads to the induction of hypoxia inducible factor-2α (HIF-2α)/CITED pathway for adaptation in low oxygen levels of the BMM to maintain LSC dormancy and self-renewal potential [137,138,139,140]. PPARγ, a negative regulator of the STAT5/HIF-2α/CITED pathway inhibits adhesion of LSCs to the extracellular matrix and drives apoptosis [8,137,140,141]. Preclinical studies demonstrated that thiazolidinediones (PPARγ agonists) upregulate matrix metalloproteinase-9 (MMP-9) and MMP-2 to inhibit LSC invasion and adhesion to the BMM. They also activate caspase-3 for LSC apoptosis [8,137,140,141]. Other findings include upregulation of PPARα ligands e.g., clofibrate and enhanced expression of human organic cation transporter 1 (hOCT1) via WY-12643, which increase cellular uptake of TKIs to promote TKI-mediated apoptosis [139,141]. Preliminary clinical studies in 3 CML patients showed that combined use of pioglitazone and IM accomplished sustained complete molecular remission for up to 4.7 years in all patients, even after pioglitazone withdrawal [141]. Phase II ACTIM trial showed that IM and pioglitazone had no drug interactions, yet their combination achieved MR4.5 at 12 months in 56% of patients [138]. Novel STAT3 inhibitor BP-5087, derived from SF-1-066, demonstrated 10-fold greater potency in reducing STAT3 phosphorylation and translocation, inhibiting survival of TKI-resistant CML cells and LSCs in preclinical studies [18,142]. Combination with TKIs showed dramatic increase in effects, whereas monotherapy of either was evidently inferior [142]. However, STAT3/5 inhibition may be less effective than JAK inhibition as other STATs may compensate for STAT3/5 loss [143].

4.9. Prostaglandin E (PGE) 1 Analogue

PGE2 is a pro-inflammatory prostaglandin upregulated by BCR-ABL1 [17,144]. It promotes β-catenin nuclear accumulation, stabilization and localization to promote the β-catenin/Wnt signalling, conferring to LSC stemness, TKI-resistance and disease progression [17,144]. On the contrary, PGE1 exhibits protective functions against LSCs [17,144]. Preclinical studies showed that misoprostol, a PGE1 analogue acts via EP4 receptor to inhibit Tcf1/Lef1 and Fos/FosB, hence reducing LSCs by >10-fold [17,144]. Exhibiting negligible effects on normal HSCs, the activation of PGE1 poses as a promising target for CML stem cell eradication.

4.10. Activation of Promyelocytic Leukaemia—Nuclear Bodies (PML-NB)

Promyelocytic leukaemia (PML) forms PML-NBs to repair DNA double-strand breaks (DSBs), maintain telomere homeostasis and maintain normal HSC asymmetric division through the PML/PPAR/FAO pathway [17,145,146]. Preclinical studies showed that PML upregulation in mesenchymal stromal cells upregulated inflammatory cytokines (IL-6/IL-6R and CXCL1/CXCR2), which are crucial for the maintenance in the BMM and TKI-resistance of CML stem cells [146]. Arsenic trioxide (ATO) was used as a first-line treatment for CML before the development of TKIs, but preclinical studies showed limited effectiveness in targeting CML stem cells as a single agent [147,148,149]. However, combination with TKIs showed LSC targeting, downregulation of VEGFR and angiogenesis, upregulation of NKG2D ligands to induce NK-cell mediated cytotoxicity, growth arrest, inhibition of RAS/MAPK and PI3K/AKT pathways, and apoptosis via extrinsic pathways (caspase-8/-10, TNFR1) and intrinsic (BAX) pathways and the induction of ER stress [147,148,149]. ATO/IFNα combination therapy demonstrated superior in-vivo and in vitro results compared to ATO/TKIs, where it induced cell-cycling of dormant LSCs and inhibited the Hh pathway, hence, leading to autophagy-induced cell death [145]. The established ability of ATO/IFNα to overcome TKI-resistance and abolish CML stem cells in preclinical studies [145,150] has led to phase I clinical trials [151]. In a cohort of eight patients, decrease in BCR-ABL1 fusion transcript was seen in 100% and 87.5% patients after trial and 12 months after trial respectively. MR4.5 or above was achieved in 87.5% and 55.6% patients immediately after study and 12 months later, respectively [151].

5. Targeting the CML Stem Cell Microenviroment, Survival and Self-Renewal

Normal HSCs interact with endothelial cells, neural cells, osteoclasts, mesenchymal stromal cells and osteoblasts in the BMM [17,25,152,153]. Selectins, integrins, and CD44 expressions are required for HSC engraftment and adhesion between fibronectin on the extracellular matrix and CD106 (VCAM-I) on the BM endothelium [23,25,152,153,154]. HSC rolling and homing is mediated by interaction between constitutively expressed E- and P- selectins and VLA4, where SDF1 and its receptor CXCR4 acts as a chemo-attractant through β1/2− integrins and SDF1 for stable engraftment [23,25,152,153,154,155,156]. BCR-ABL1 impairs the SDF1/CXCR4 axis in normal HSCs but upregulates it in CML stem cells, conferring selective homing and survival in the BM niche [17,23,152,153,157,158]. In addition, CML stem cells have defective β1-integrin levels (VLA4 or VLA5), allowing redistribution and mobilization into the PB and other organs, e.g., spleen with the potential of uncontrolled extramedullary myeloproliferation and local LSC reservoirs [152,154,157]. CML stem cells alter extrinsic factors and upregulate expression of CD44+ and E-selectin to promote prominent BMM changes such as marrow fibrosis for exclusive stem cell engraftment and dormancy, offering protection from drug-targeting [25,152,154,158,159,160,161,162].

5.1. Dipeptidyl-Peptidase (DPP-4) Inhibition

DPP-4 (CD26) is an overtly expressed protease on LSC surface, where it cleaves the SDF1/CXCR4 axis to facilitate LSC mobilization into the PB independent of niche regulations [16,17]. TKIs decrease CD26+ LSCs but levels dramatically increase following resistance or relapse [16,17,137]. CD26 is not expressed on normal HSCs, suggesting that it may be a marker for LSCs as concentrations correlate with white blood cell (WBC) count [16]. DPP-4 inhibitors (gliptins) normalize the dysregulated SDF1/CXCR4 axis to restore and promote homing of LSCs [16,36]. Interestingly, Willmann et al. demonstrated that single agent nilotinib could inhibit engraftment and induce apoptosis of LSCs, while neither vildagliptin nor imatinib addition exhibited these effects [36]. Combination of nilotinib with vildagliptin also did not produce cooperative results, suggesting insignificant effects of co-administration [36]. However, vildagliptin alone reduced disease expansion through limiting LSC mobilization [16,36]. In samples of two nilotinib-pretreated CML patients with diabetes mellitus using gliptins for diabetic control, BCR-ABL1 transcripts were near undetectable or undetectable [16].

5.2. E-Selectin Antagonist

Uproleselan (GMI-1271) is an E-selectin inhibitor which dislocates homed LSC from the BM niche into PB for cellular differentiation [154,163]. Promising phase III study results in acute myeloid leukaemia (AML) for LSC eradication [163] has led to preclinical studies in CML. In vitro studies demonstrated cell cycle progression via upregulated CDK6 (cell cycle promotor) and downregulated p16 (cell cycle inhibitor), leading to an increase in cells in G-phase and increase G2/S/M phase when used as monotherapy or in combination with IM [152,154]. Reduced CD44+ expression via the Scl/Tal1 pathway, increased CML stem cell cycling, and restoration of TKI-sensitivity were also noted [152,154]. Murine models showed depletion of LSC and BCR-ABL1+ cells, spleen size reduction, impaired LSC engraftment to the BM and spleen, and improved OS [152,154].

5.3. Targeting SDF1/CXCR4/CXCR7 Axis

Preclinical studies showed that disruption of the SDF1/CXCR4/CXCR7 axis of mesenchymal stromal cells reduced EZH2 expression [164], increased self-renewal capacity in LSCs and the ability to override TKI-resistance with no effect on osteoprogenitor cells, mesenchymal stromal cells and normal HSCs [164,165,166]. NOX-A12, a pegylated Spiegelmer, inhibits SDF1 and antagonizes the SDF1-CXCR4 or -CXCR7 interactions to inhibit LSC homing and causes TKI-sensitization [167]. In-vitro studies showed enhanced abolishment of SDF1-mediated migration in BCR-ABL1+ cells and induction of apoptosis when combination with imatinib was used (p < 0.00005) [168]. In-vivo studies showed eradication of FLT3-ITD+ cells and inhibition of SDF1-mediated migration of FLT3-ITD+ cells [168]. Plerixafor, an allosteric CXCR7 agonist and CXCR4 antagonist/partial agonist, is clinically used for stem cell mobilization in HSCT in multiple myeloma and non-Hodgkin lymphoma [165,168,169]. Its use in in vitro studies with K562 and KU812 cell lines showed reduction of drug-resistance, cellular migration and adhesion to BMM and sensitization to TKI [165]. Plerixafor in in-vivo murine models mobilized LSCs to the PB, potentiating TKI-induced tumor bulk eradication [165]. However, Agarwal et al. presented contradicting in-vivo results, which demonstrated that TKI plus plerixafor led to stem cell infiltration of the central nervous system (CNS) and subsequent development of neurological deficits [170].

5.4. Hypoxia-Inducible Factor (HIF) Targeting

HIFs interact with HIF-responsive elements (HRE) for gene regulation, depending on the oxygen concentration of the microenvironment [17,28,171,172,173]. The hypoxic BM niche contains high ROS levels which causes upregulation of HIF-1α and HIF-2α, suppressing BCR-ABL1 oncoprotein yet permitting CML stem cell survival, quiescence, immune-evasion, TKI-resistance and potential transformation of normal HSC into induced pluripotent stem cells capable of becoming LSCs [17,28,172,173]. This is mediated by glucose transporter 1 (GLUT1) and tumour M2-pyruvate kinase (PKM2), which lead to increased glycolytic flux [28], p21 upregulation for cellular proliferation [172], suppression of p53 [28,172,173], increased transcription of antioxidant enzymes (FoxO and Nrf2) [28,173], overt Oct4 and c-Myc for transformation of other haematopoietic cells into LSCs [173], and evasion of cellular immunity through B7H1/programmed death ligand 1 (PD-L1) expression, and soluble factors such as nitric oxide [28]. LSCs have high enough ROS levels for clonal evolution yet low enough levels to maintain stemness [8,17]. Acriflavine, a HIF-1 inhibitor prevents dimerization of the HIF complex and reduces LSC formation, maintenance, survival and stemness through three mechanisms [17,173,174]: depleting c-Myc at mRNA and protein levels, promoting expression of tumour suppressors (e.g., p57, p19Arf and p16Ink4a) and inhibiting genes that favour LSC stemness (e.g., NANOG, Oct4, Sox9). In vitro studies using K562, KCL22, and LAMA-84 CML cell lines and in-vivo studies demonstrated cytotoxicity against BCR-ABL1+ cells and LSCs, where adverse effects on normal HSCs were significantly less-severe in-vivo [174].

5.5. Targeting Hh Pathway

Hh homologues bind to the Patched (PTCH) receptor, activating Smoothened (Smo) and Gli family of transcription factors to mediate downstream signalling (e.g., Myc, cyclin-D1, Bcl-2, SOX2) for cellular regeneration and homeostasis [8,18,29,175,176]. Low Shh levels in mesenchymal stromal cells along with hyperactivation of Shh and Smo in CD34+ and c-kit+ (p < 0.05) LSCs stimulate cyclin-D1-mediated LSC quiescence, maintenance and uncontrolled expansion through the Wnt/β-integrin pathway [175]. Hh overactivation is seen in 50% chronic phase (CP)-CML, 70% accelerated phase (AP)-CML and >80% blast-phase (BP)-CML patients [175]. In vitro and vivo studies showed that LDE225 (sonidegib), a highly selective and potent Smo inhibitor, was effective alone and in combination with TKIs in eradicating Hh-mediated self-renewal capacity of CD34+ and BCR-ABL1+ CML cells [18,177]. In a phase I trial CA180323, another Smo inhibitor, BMS-833923, was investigated in combination with dasatinib, showing no drug interactions and undesirable toxicity profiles and minimal reduction of BCR-ABL1 progenitors [176]. Although preclinical and preliminary clinical studies showed conflicting results, the Hh pathway remains a significant target worth investigating.

5.6. Targeting Wnt/β-Catenin Signalling

Porcupine (PORCN)-dependent acetylation of Wnt ligands is essential in Wnt/β-catenin signalling for maintenance of cellular functions [17,178,179]. BCR-ABL1 drives constitutive secretion of Wnt-ligands and overexpression of frizzled-4 (FZD4) receptors to promote nuclear transduction and stabilization of β-catenin, mediating TKI-resistance [178,179,180]. Riether et al. proposed that it might be induced by prolonged TKI exposure as TKI-therapy depleted miR29 and amplified CD70 expression, leading to CD27-mediated Wnt activation for LSC quiescence and therapy resistance [181]. In an in-vivo study using transgenic murine models with CD34+ and c-kit cells, potent PORCN inhibitor WNT974 in combination with nilotinib was efficacious in reducing neutrophils, white blood cells and myeloid cells in PB, with eradication of CML stem cells and other progenitors in the BM and spleen [18,178,179]. Mice treated with nilotinib monotherapy died after 30 days while nilotinib plus WNT974-treated mice had prolonged survival with prominent suppression of c-Myc, cyclin-D1 and Axin-2 expression [18,178]. C82, a β-catenin inhibitor, downregulated CD44, c-Myc, STAT5, survivin, and CRKL in T315I and E255V mutant cell lines, eliminating LSCs in-vitro and in-vivo [27].

5.7. Targeting Protein Phosphatase 2A (PP2A)

PP2A is a serine-threonine phosphatase that acts as a tumour suppressor, contributing to >90% intracellular phosphatase activity alongside PP1 [29,182,183,184,185]. PP2A dephosphorylates Myc, disrupting Myc/MAX interaction and inhibits gene expression for mitochondrial biogenesis. BCR-ABL1 oncoprotein amplifies endogenous expression of potent PP2A inhibitors protein SET (SET), cancerous inhibitor of PP2A (CIP2A) and PP2A-Aα that inactivate phosphatase activity, hence, resulting in high levels of Myc and uncontrolled DNA synthesis for LSCs survival and maintenance [29,182,183,186,187]. The Myc/MAX complex can directly bind BCR-ABL1 to upregulate its mRNA and protein content, establishing a positive feedback loop for LSCs [182,185]. Myc inhibitor 10058-F4 suppressed CIP2A in 80% of CD34+ cells (p = 0.04) and 85% of K562 cells (p = 0.01), preventing Myc/MAX interaction and restoring tumour-suppressor functions in vitro K562 and CD34+ cell lines [29,184]. However, Myc-targeting remains a therapeutic challenge due to the lack of a clear ligand binding domain [182]. OP449, a SET antagonist reactivated PP2A and significantly reduced JAK/STAT5 and PI3K/AKT pathways in vitro CD34+ and K562 CML cells, as well as alleviated tumour burden in vivo xenografted mice with human CML cells [182,186]. FTY720, a SET antagonist activates extrinsic and intrinsic apoptotic pathways in a PP2A-dependent manner [29,184,186,188] or via the activation of caspase-3/-8/-9 and pro-apoptotic BH3-only proteins (BIM and BID) in-vitro K562, MYL, KBM5 and KCL22 cell lines [184]. It can also overcome BIM-deletion-mediated, Gal-3-mediated BCR-ABL1 kinase-domain-mediated TKI resistance, with synergistic activity in combination with imatinib [184]. However, Bcl-2 expression partially hinders FTY720-mediated apoptosis [184]. Combination of FTY720 or OP449 with TKIs showed drug synergism [182,184,186,188].

6. Targeting CML Stem Cells via Epigenetic, Ribosomal and Transcriptional Regulation

Epigenetic modifications are reversible and heritable changes that regulate DNA expression while maintaining the same nucleotide sequence [189,190,191]. High ROS levels and hypoxic conditions of the BMM lead to DNA damage and ineffective repair, making LSCs prime candidates to undergo genetic evolution. Pre-leukaemic lesions in epigenetic regulators (e.g., DNMT3A, IDH1/2, TET1/2, TP53) result in clonal hematopoiesis of intermediate potential (CHIP), a predisposing factor for haematological malignancies [8,192,193,194,195]. Despite having a peak incidence of 15–20% in the general healthy population after the age of 70 [8,196,197,198,199], CHIP is not a cause for CML [8,21,193]. However, the concurrent presence of CHIP and leukaemia drives LSC transformation and survival, and is associated with an inferior prognosis [8,17,189,195,196,197,198,199].

6.1. Bromodomain and Extra-Terminal (BET) Inhibitor

BET proteins are epigenetic regulators of transcription, inflammatory processes, and cell-cycle regulation [18,200,201,202]. Bromodomain-containing protein 4 (BRD4) is the only ubiquitously expressed member of the family directly bound to P-TEFb to maintain chromatin stability and G2/M phase transition in the cell cycle. BCR-ABL1 and LSCs can acquire secretory-associated senescent phenotype (SASP) to drive BRD4 activity and upregulation of Myc, leading to overt release of pro-inflammatory cytokines (e.g., IL-1, IL-6, IL-8, IL-17, IL-23, BMP2, TNFα, CCL9, NF-κB) to favour LSC senescence. Aberrant BRD4 activity also induces active transcription and expression of PD-L1 on leukaemic cells, myeloid dendritic cells and macrophages for immune-evasion. BRD4 inhibitor JQ-1, as well as BRD4 degraders dBET1 and dBET6 have shown to be promising and potent inhibitors that downregulate Myc in targeting LSCs and overcoming TKI-resistance [200,202,203,204,205]. In vitro studies showed that JQ-1 increased IL-12β to reduce VEGFR-mediated angiogenesis while decreasing PD-L1 expression to promote CTL-cytotoxicity [200,201,202,204] and IL-6-mediated Jagged1/Notch1 cellular invasion and migration [201]. In vitro studies using K562 and KU812 cell lines showed superior potency of dBET6 and dBET1 over JQ-1 in suppressing BRD4 and Myc expression, where dBET6 and dBET1 could eliminate BCR-ABL1+ cells and CD34+/CD38 LSCs while JQ-1 failed [202]. In vivo studies demonstrated that dBET6 could override niche cell-induced TKI-resistance in CML LSCs, while JQ-1 was only able to restore TKI effects completely in KU812 cells and partially in K562 cells [202]. All three BRD4 inhibitors inhibited IFNγ-induced PD-L1 expression in LSCs [202,205]. Promising preclinical studies have led to the development of novel BRD4 inhibitor CPI-0610, which is currently in phase I clinical trials (ClinicalTrials.gov identifier: NCT02158858).

6.2. EZH2 Inhibition

EZH2 is a histone methyltransferase and a component of the polycomb repressive complex 2 (PRC2) for histone H3 methylation (H3K27me3) and transcription inactivation [17,206,207,208]. EZH2 hyperactivity blocks myeloid differentiation to promote LSC expansion, survival and TKI-resistance [17,43,207,208,209,210,211]. In vitro studies showed that EZH2 inhibitor EPZ-6438 upregulated tumour suppressor p16 to deplete leukaemic cells in K562, HEL, Kasumi-1, ME-1, Mv4-11 and MOLM13 cell lines [207,210]. Inactivation of EZH2 showed significant reduction in WBC count and LSCs in the PB, prolonged survival, and absence of splenomegaly and pulmonary hemorrhage compared to the control arm [210]. Another in vitro study showed 20–40% reduction in CD34+ cells and 60–80% reduction in progenitor granulocyte/erythroid/megakaryocyte/macrophage (GEMM) and total colony forming cell (CFC) [43]. Combination with TKI increased activation of H3K27me3 targets (e.g., CDKN2A) and upregulated pro-apoptotic targets of p53 (e.g., NOXA, p53 upregulated modulator of apoptosis (PUMA), BAX, CDKN2A, TNFRS10B), showing >70% reduction in CD34+/CD38/CD45+ cells, near complete eradication of CD34+/CD45+ cells and restoration of TKI-sensitivity [43,211]. Murine xenograft models with CD34+ cells showed similar findings [43].

6.3. Histone Deacetylase (HDAC) Inhibitor

HDAC packs histones tightly to inhibit transcription. Aberrant activity in CML LSC halts myeloid differentiation to allow LSC survival [18,212,213,214]. Panobinostat (LBH589), a HDAC inhibitor combined with TKIs in preclinical studies showed impairment of LSC quiescence and engraftment, promoting TKI-mediated apoptosis [18,212,213,214] in vivo CD34+ mice [212,213] and in vitro cell lines CD34+/CD38, K562, K562/IM-R1, Ba/F3 and Fa/F3/T315I [18,212,213]. A phase I clinical trial exhibited no dose-limiting toxicities and 44% patients achieving >1 log reduction in BCR-ABL1/ABL1 transcripts, but was discontinued due to slow accrual [18]. A phase II study also showed no MCyR nor molecular response. However, in light of encouraging preclinical results, a phase Ib trial is currently underway (ClinicalTrials.gov identifier: NCT03878524). Chidamide, an orally bioavailable HDAC inhibitor in vitro increased acetylation of Histone H3, activated caspase-3/-9 and decreased levels of β-catenin, surviving and Myc in CD34+ cells, inducing apoptosis [215]. It also exhibited limited toxicity to normal HSCs, and produced drug-synergism in combination with TKIs in overriding BCR-ABL1-dependent and -independent mutations [215]. Another novel pan-HDAC inhibitor MAKV-8 reduced c-Myc expression, activated caspase-3/-9-mediated apoptosis and triggered ER stress for LSC eradication using K562 and MEG-01 cell lines in vitro [214]. When used in combination with TKIs, in vivo K562 cells in zebra-fish were completely abolished [214].

6.4. Protein Arginine Methyltransferase (PRMT5) Inhibitor

PRMT5 catalyzes histone methylation for RNA metabolism, transcriptional regulation, ribosome biogenesis and cell-cycle regulation [216]. Aberrantly expressed PRMT5 binds to BCR-ABL1 to form a positive feedback loop, which is associated with worse progression-free survival in CML patients [216,217,218]. Preclinical studies showed that PJ-68, a PRMT5 inhibitor suppressed the Wnt/β-catenin pathway and induced negative regulators of LSC cellular renewal P15INK4B and p27KIP1 for CD34+CD38 apoptosis [18,218].

7. Targeting CML Stem Cells via P53 Modulation

P53 is crucial in tumour suppression and apoptotic control [29,219,220]. It is dysfunctional in CML as it directly binds to IκBα and BCR-ABL1, leading to dysregulated p53 and Myc levels [29,219,220]. Preclinical studies showed that depletion of E3 ligase FBWX7 (SCF) led to Myc-induced p53 upregulation, promoting p53-mediated cell-cycle regulation and apoptosis in CD34+ and HeLa cell lines [219]. This spared normal HSCs and achieved near complete elimination of BCR-ABL1+ cells and LSCs [219]. Dasatinib showed some efficacy in STAT5 inhibition and mutant TP53 reduction. However, it was insufficient to eliminate LSCs [219,221]. RITA (NSC652287) binds and prevents p53 degradation. In vitro studies showed degradation of IκBα and downregulation of NF-κB-regulated proliferative (c-Myc) and anti-apoptotic (Bcl-2, XIAP, cIAP1) genes, inhibition of PI3K/AKT and JAK/STAT5 signalling pathways, increased p53-mediated apoptosis and decreased c-Myc levels leading to subsequent CD34+ and K562 cellular knockdown [219,222,223]. In-vivo mouse CML models using RITA in combination with CPI-203 (BET family inhibitor) showed decreased levels of CD11b, CD19, CD33, CD34 and CD45, suggesting reduced LSC engraftment [219,222]. P53-mediated apoptosis requires phosphorylation at Serine-46 (Ser46), where deficiency leads to drug resistance. In vitro studies showed that, despite the ability of RITA to override HDM2 inhibition, it remained ineffective against p53-mutant cells lacking phosphorylated Ser46 [224,225].

7.1. Sirtuin 1 (SIRT1) Inhibition

SIRT1, a NAD+-dependent deacetylase, is a potent suppressor of tumour suppressor p53 found to be overly expressed in CML stem cells [18,221,226]. It activates PGC-1α to promote mitochondrial DNA replication to maintain the bioenergetic demands of LSCs [226]. SIRT1 deletion in vitro and in vivo demonstrated downregulation of mitochondrial genes and upregulation of p53 acetylation in LSCs and progenitor cells [221,226]. While TKI treatment did not affect mitochondrial respiration [226], combination with SIRT inhibitors restored sensitivity to TKIs and subsequent TKI-mediated apoptosis [221,226].

7.2. Human Double Minute 2 Protein (HDM2) Inhibition

HDM2, another p53 negative regulator, inhibits TP53 transcription via binding to its transactivation domain [18,227,228]. Hyperactivity of CML LSCs leads to p53 proteasomal degradation and evasion of apoptosis. DS-5272, an HDM2 antagonist, restored TKI sensitivity via p53 reactivation and induction of NOXA, leading to silencing of anti-apoptotic Mcl-1 [229]. Combination with TKIs or BET inhibitors suppressed downstream Myc-related pathways and upregulated p53, NOXA and BAXA, reducing the threshold for TKI-mediated apoptosis [229]. In vitro and in vivo results demonstrated high selectivity and near complete eradication of LSCs [229]. MI-219 directly stabilized and reactivated p53, reduced CD44+ for LSC homing and engraftment, and depleted important genes for LSC self-renewal (e.g., JARID2, PRDM16) both in vitro and in vivo [18,227]. MI-219 had limited effects on normal HSCs, and it upregulated IFNAR1 to drive LSCs into the cell cycle and exhaust them [227].

8. Targeting Autophagy in CML Stem Cells

Autophagy is the stress-induced formation of autophagosome for recycling and degradation of damaged and/or aged cytoplasmic components to sustain bioenergetic and nutritional demands [24,29,230,231,232]. A metabolic shift in LSCs results in increased glucose influx, pyruvate shuttling, glycolysis, anaplerosis, oxidative phosphorylation, and ROS overload (Warburg effect) for survival and maintenance of stemness [8,29,171,232]. Moreover, the upregulation of Beclin-1 is essential in autophagic flux [29,230,231], acting as a protective mechanism to avoid oxidative stress and apoptosis for the maintenance of stemness [24,29,230,231,232].

8.1. Tigecycline

Tigecycline, a third generation tetracycline, is active against LSCs via three mechanisms with negligible effects on normal HSCs [8,203,232,233,234,235]: Atg7 knockdown depleting glucose levels in LSCs, downregulation of signalling pathways (e.g., Wnt/β-catenin, PI3K/AKT/mTORC1, p21CIP1/Warf1, hypoxia-inducible factors (HIF), c-Myc) for autophagosome formation, and binding to 28S subunit of ribosome (homologous to 30S in bacteria) to activate cytochrome-c/caspase-9/caspase-3 causing defective mitochondrial translation, oxidative phosphorylation, electron transport chain and oxygen consumption. In vitro studies using CD34+/CD38 cells and in vivo murine models demonstrated superior efficacy in the reduction of tumour load in combination with IM than either agent alone [233].

8.2. Chloroquine (CQ)

CQ becomes protonated and trapped in lysosomes to alkalize acidic hydrolases, prevents fusion with autophagosomes, increases cellular stress and drives apoptosis [236]. In vitro studies showed that CQ eliminated BCR-ABL1+ cells and sensitized CD34+/CD38 to TKI-mediated apoptosis [24,237]. These lead to CHOICES (Chloroquine and Imatinib Combination to Eliminate Stem cells), a randomized phase II trial in comparing IM alone versus IM plus hydroxychloroquine (HCQ) [24,238]. MMR was 92% (IM/HCQ) vs. 80% (IM) at 12 months, and the qPCR level at 24 months with ≥0.5 log reduction was 75% (IM/HCQ) versus 67% (IM) [238].

9. Immunotherapeutic Targeting of CML Stem Cell

Targeting PD-1/PD-L1 Axis

The PD-1/PD-L1 axis is responsible for self-tolerance [44,239]. CML induces IFNγ-mediated PD-L1 expression for LSCs to aid evasion of CTL-cytotoxicity and recruitment of MDSCs and regulatory T cells for immune-evasion. Preclinical studies showed that T-cell immunotherapy with PD-1 inhibition eliminated LSCs [239]. Nivolumab, a monoclonal IgG4 antibody (Ab) against PD-1, was used on an 82-year-old man, with the ability to achieve undetectable BCR-ABL1 transcripts as a single agent [240]. Results of phase I clinical trials with dasatinib are pending (ClinicalTrials.gov identifier: NCT02011945). Avelumab [241], a monoclonal IgG1 Ab against PD-L1, is currently in clinical trials with various TKIs (ClinicalTrials.gov identifier: NCT02767063).

10. Novel Therapies Targeting MPN Stem Cells via Signaling, Apoptotic and Cell Cycle Pathways

10.1. Telomerase Inhibition

Telomerase is a ribonuclear protein complex comprised of human telomerase reverse transcriptase (hTERT), an RNA template (hTR), and specialized proteins (e.g., dyskerin), that extend the length of telomere [86,242,243,244,245]. It maintains replicative potential and is actively expressed in HSPC [86,242,243,244,245]. In MPN, telomerase is overexpressed and upregulated [86,242,243].
Imetelstat (GRN163L) is a 13-mer oligonucleotide that inhibits hTR in telomerase [86,242,243,244,245,246], resulting in selective apoptosis of MPN stem cells [242,243,245]. Preclinical studies demonstrated preferential apoptosis of MF CD34+ cells irrespective of driver mutations [243], and reduced polyploidization and maturation of CD41+/CD42b+ megakaryocytes [242]. Decreased malignant megakaryocytes led to reduced growth factors (e.g., platelet-derived growth factor (PDGF), fibroblast growth factor 2 (FGF-2)) and inflammatory cytokines, hence reduction in BM fibrosis in MPN cultures [242] A pilot study suggested that additional spliceosome mutations (e.g., U2AF1, SF3B1) might lead to suboptimal telomerase upregulation and increase patients’ sensitivity to telomerase inhibition [244] (Table 3).

10.2. BET Inhibition

BET proteins are chromatin-reader proteins [262]. The hydrophobic N-terminal bromodomain (BRD) of BET binds to acetylated lysine on histone tails and recruits chromatin factors to regulate transcription of c-Myc, Bcl-2, and NF-κB [86,262,263,264]. NF-κB is crucial in mediating chronic inflammation in MPN [86]. BET inhibitors displace BET proteins to attenuate NF-κB signalling [262]. In-vivo studies showed BET inhibitor JQ-1 diminished NF-κB activation and pro-inflammatory cytokine production [265]. In MPN, both JAK/STAT activation and BET activation promote NF-κB pathway. Interplay between NF-κB-activated cytokines and JAK/STAT3 further contributes to disease progression [264,265]. Preclinical studies showed that combination of JQ-1 with ruxolitinib produced synergistic effects in reducing fibrosis, extramedullary haematopoiesis (EMH), leukocytosis, and targeting of primary CD34+ progenitor cells in MF patients [86,264,265]. In MF, 10% patients display loss of EZH2, which promotes acetylation of histone H3 at lysine 27 (H3K27ac). This increases the sensitivity of BET inhibitor JQ-1 to JAK2V617F/EZH2Δ/Δ cells [264,266] (Table 3).

10.3. Mouse Double Minute 2 Homolog (MDM2) Inhibition

MDM2, coupled with MDM4, ubiquitylates and degrades p53 to maintain p53, a tumour suppressor gene that plays a critical role in apoptosis and DNA repair [247,267,268,269], to maintain it at a low level [247]. In MPN, MDM2 is overexpressed in PV and MF CD34+ cells [247,267,268,269]. Gain of 1q and 12q chromosomal rearrangements are associated with increased expression of MDM4 and MDM2 respectively [268,269]. Gain of 1q is seen in 43% MPN patents with leukaemic transformation. The presence of +1q contributes to MPN disease progression and is also found to be specifically associated with JAK2V617F mutation, while 12q gain is associated with MF [269]. MDM2 inhibitors, nutlins (Nutlin-3, RG7112, RG7388 (idasanutlin), HDM201, and KRT232), act by hindering MDM2-p53 interactions, increasing p53 levels, resulting in MPN CD34+ cell apoptosis [268]. IFNα can also act through p53 to mediate PV haematopoietic progenitor cell (HPC) apoptosis by activating the p38 MAPK pathway [63,247,267]. Preclinical study showed that low dose RG7112 and Peg-IFNα-2a increased p53 downstream pro-apoptotic proteins (e.g., PUMA, BAX), leading to MPN CD34+ cell apoptosis. The elimination of MPN stem cells was demonstrated by the significant reduction of JAK2V617F allele burden in PV, MF CD34+ cells in BM as shown in the study [267]. Clinical trials of combination therapy with ruxolitinib in PV and MF are also underway, but results are yet to be published [270,271,272].

10.4. Heat Shock Protein (HSP) Inhibition

HSP is a family of ATP-dependent, cytoprotective, stress-response protein chaperones that bind and stabilize client proteins in their functional active form, thus maintaining survival advantage of MPN cells. [248,272,273,274,275]. HSP90 chaperones JAK2 while HSP27 modulates STAT5 phosphorylation, making them therapeutic targets in MPN [273]. HSP90 inhibitor disrupts association between HSP90 and JAK2, leading to JAK2 misfolding and degradation by 26S proteasome [272,275]. In vitro and in vivo studies have demonstrated that HSP90 inhibitor (PU-H71) inhibited, and even degraded JAK2, resulting in abrogation of downstream signaling pathway (e.g., STAT3, STAT5, MAPK). A reduction of MPLW515L allele burden, EMH, and normalization of blood counts were seen in PU-H71-treated murine models [276]. However, preclinical study using another HSP90 inhibitor AUY922 showed a rapid elevation of JAK2V617F level upon AUY922 termination [275]. A phase 2 study also showed that AUY922 failed to demonstrate consistent reduction of JAK2V617F mutant allele burden. This suggests that HSP90-mediated JAK/STAT pathway inhibition may be short-lived [248] (Table 3). An important finding is that the combination of JAK inhibitor (TG101209) with AUY922 acted synergistically to induce apoptosis in primary MF CD34+ cells [273,274,275]. The pro-apoptotic effect was also seen in JAK1/2 inhibitor-resistant cells [275], suggesting a possible solution to overcome ruxolitinib resistance. An emerging role of HSP27 inhibitor (KNK437) is also displayed by its synergistic action with ruxolitinib in JAK2V617F cell lines and PV patient cells. In murine models, HSP27 inhibitor (OGX-427, Apartosen) reduced splenomegaly, BM fibrosis and normalized counts, reflecting the therapeutic potential of HSP27 inhibitors [273].

10.5. Poly-ADP-Ribose Polymerase (PARP) Inhibition

PARP is a protein involved in DNA repair. It maintains MPN cell survival alongside two other DNA repair mechanisms: BRCA1/2-mediated homologous recombination repair (HRR) and DNA-dependent protein kinase, catalytic subunit-mediated non-homologous end-joining (D-NHEJ) [277]. ROS levels are found to be elevated in MPN LSCs, predisposing cells to toxic DNA DSBs [277,278], which activate DNA repair by PARP via the recruitment of repair proteins [279]. The significance of PARP inhibitors is revealed when used in combination with ruxolitinib [277]. Ruxolitinib downregulates important molecules in HRR and D-NHEJ in cell lines of all three driver mutations and sensitizes both proliferating and quiescent MPN stem cells to PARP inhibitors. Preclinical studies demonstrated promising synergistic effects of PARP inhibitors (Olaparib and BMN673) with ruxolitinib in eradicating MPN stem cells by apoptosis [277].

10.6. CD123 Targeting

Interleukin-3 receptor (IL-3R) consists of an alpha chain (CD123) and a common beta chain (CD131) [86,280]. IL-3, a cytokine released by activated T-lymphocytes, binds to CD123 which dimerizes with CD131 to trigger downstream JAK2 signalling pathway [280,281]. CD123 is overexpressed in CD34+/CD38 LSCs in AML, but not in normal HSCs [282]. Overexpression of CD123 is also found in some MF patients, especially in patients with monocytosis, which confers a poor prognosis [249,281]. Tagraxofusp is a recombinant protein genetically engineered from the fusion of IL-3 to the catalytic and translocation domains of diphtheria toxin [86,281]. Upon binding to CD123 and internalization into MPN LSCs, the catalytic domain of diphtheria toxin is cleaved. This inactivates elongation factor 2 (EF2), which is responsible for protein synthesis. Thus, the apoptosis of LSC is driven [280]. Early phase clinical trials performed to evaluate the efficacy of tagraxofusp in advanced MF patients showed promising results [249,283] (Table 3).

10.7. Proviral Integration Site for Moloney Murine Leukemia Virus (PIM) Kinase Inhibition

PIM kinase is a family of anti-apoptotic serine/threonine proto-oncogenes that are transcriptionally activated by JAK/STAT signalling [272,284,285,286]. In MPN, continuous activation of JAK/STAT pathway is observed, making PIM kinase a potential therapeutic target [272,286]. Although the efficacy of PIM kinase inhibitor monotherapy is limited [285], it acts synergistically with ruxolitinib to suppress JAK2 signalling effectively [285,286]. In MPN, pro-apoptotic BAD protein is phosphorylated and inhibited by PIM kinase, prolonging MPN cell survival. Therefore, PIM kinase inhibitor (AZD1208) liberates BAD protein to induce JAK2V617F cellular apoptosis with ruxolitinib [285]. Enhanced cleavage of DNA repair enzyme PARP was also demonstrated to promote apoptosis in a preclinical study [286]. Importantly, preclinical studies showed that PIM kinase inhibitor resensitized ruxolitinib-resistant cell lines to apoptosis [285], while combination treatment prevented disease progression of myeloproliferation and splenomegaly in murine models [286].

10.8. PI3K/AKT/mTOR Inhibition

The PI3K/AKT/mTOR cascade is a key downstream pathway of JAK/STAT pathway for cellular survival and proliferation [86,272,287,288]. One of the limitations of standard therapy ruxolitinib is the incomplete suppression of STAT5 phosphorylation in JAK2V617F cell lines [287]. Hence, the synergistic effect of PI3K/mTOR inhibitor and ruxolitinib is postulated. Ruxolitinib inhibits phosphorylation of STAT5a while PI3K/mTOR inhibitor dephosphorylates STAT5b via CIP2A/PP2A axis [287]. Fiskus et al. showed that synergism of dual PI3K/mTOR inhibitor (BEZ235) and JAK2 inhibitor induced preferential apoptosis in MF JAK2V617F CD34+ cells, while sparing normal HSCs [288]. Hence, clinical trials are being carried out [261,289].

10.9. Bcl-xL Inhibition

Bcl-2 family includes anti-apoptotic proteins (e.g., Bcl-xL, Bcl-2) and pro-apoptotic proteins (e.g., BAX, Bcl-2 homologous antagonist killer (BAK)). Bcl-xL heterodimerizes with BAX and BAK to exert anti-apoptotic effect [290]. In MPN, Bcl-xL is overexpressed with highest level displayed in MF, followed by PV then ET regardless of JAK2V617F mutation status [290]. Significance of Bcl-xL inhibitor is seen in the cotreatment with ruxolitinib. In-vitro studies showed ABT-737, a BH3 mimetic inhibitor that inhibits both Bcl-xL and Bcl2, acted synergistically with ruxolitinib in driving cellular apoptosis [290]. It is also important to note that Bcl-xL inhibitor is a possible treatment to overcome ruxolitinib resistance. The activation of RAS and its downstream pathways inhibit pro-apoptotic Bcl-2-antagonist of cell death (BAD). Hence, BAD cannot bind and inhibit Bcl-xL, contributing to JAK2 inhibitor resistance [291]. Resensitization in JAK2 inhibitor-resistant cells was manifested by co-treatment of JAK2 inhibitor and ABT-737 in preclinical study [292]. Phase 2 clinical study has demonstrated promising results with combination treatment [293].

11. Targeting MPN Stem Cells via Epigenetic Regulation

11.1. Lysine Specific Demethylase-1 (LSD-1) Inhibition

LSD-1 is an epigenetic enzyme which demethylates lysine residue on histone H3 to sustain MPN LSC self-renewal [272,294]. In MPN, LSD1 is overexpressed and accounts for 58% of MF patients [272]. An LSD-1 inhibitor, bomedemstat (IMG-7289), is shown to markedly decrease mutant allele burden by TP53 activation and cell cycle arrest [294]. IMG-7289 restores TP53 methylation, leading to an increase in PUMA to induce apoptosis. Meanwhile, anti-apoptotic Bcl-xL is suppressed by TP53, further facilitating apoptosis and reducing mutant allele burden in murine models. Other promising effects include the reduction of BM fibrosis, EMH-mediated splenomegaly and inflammation, as well as the normalization of blood counts. Furthermore, ruxolitinib plus IMG-7289 were shown to eliminate MPN stem cells in murine models, encouraging further investigations [294]. Clinical trials of IMG-7289 are currently underway (Table 3).

11.2. PRMT5 Inhibition

PRMT5 functions by methylating histones and cellular proteins such as p53 [295]. In MPN, particularly PV, PRMT5 is phosphorylated by JAK2V617F [295,296]. The phosphorylation activity inhibits PRMT5 methylation activity, and promotes clonal myeloproliferation [295]. PRMT5 inhibitor binds and impairs phosphorylation of PRMT5 by JAK2V617F, impairs E2F transcription factor 1 (E2F1) methylation, and results in downstream epigenetic dysregulation [295,296]. In addition, a preclinical study demonstrated that PRMT5 inhibitor (CTx034) abrogated erythropoiesis in JAK2V617F cells without suppressing normal haematopoiesis [297]. Erythroid progenitors were significantly reduced by another PRMT5 inhibitor (C220) [296]. Decrease in hepatosplenomegaly, marrow fibrosis, and proinflammatory cytokines were also displayed in JAK2V617F and MPLW515L murine models. These effects were further enhanced by the combination with JAK inhibitor [296]. Phase 1 clinical trial is currently underway [255] (Table 3).

11.3. HDAC Inhibition

HDAC is a family of proteins that diminish acetylation of histones and non-histone proteins (e.g., p53, NF-κB, HSP90, STAT3, Ku70 (enzyme for DNA repair)) [272,298]. In MPN, particularly MF, HDAC is overexpressed, inducing a number of downstream pathways [298], leading to myeloproliferation. Similar to HSP90 inhibitors, HDAC inhibitors (givinostat and panobinostat) acetylate HSP90, prevent JAK2V617F-HSP90 interactions, and degrade JAK2V617F [74,298]. A preclinical study showed that givinostat acetylated and downregulated transcriptional factor nuclear factor, erythroid 2 (NFE2), hence, reducing erythroid differentiation in CD34+ cells [299]. The aforementioned pathway is independent of JAK2/STAT5, which is targeted by JAK2 inhibitors [299]. Panobinostat was shown to act synergistically with JAK2 inhibitor (TG101209) to drive apoptosis in primary CD34+ MF cells [300]. Efficacy is further demonstrated in a phase Ib clinical trial [301]. Although HDAC inhibitors exert multiple effects, poor tolerability remains a major issue [254,272] with the exception of treatment with givinostat in PV patients, which showed positive results in a few clinical trials [74,253,284] (Table 3).

12. Targeting the MPN Stem Cell Niche and Marrow Microenvironment

The BM niche is essential for sustaining self-renewal of MPN stem cells, hence, providing them with survival advantages over normal HSCs via generation of ROS pro-inflammatory cytokines [14,302]. In view of the positive feedback loop between BM niche and MPN stem cells, various novel agents are developed.

12.1. β-3 Sympathomimetic Agonists

Nestin+ mesenchymal stem cell (MSC) is one of the three major types of perivascular stromal cells that secrete high levels of SDF1 for HSC quiescence and migration [302,303]. This SDF1 release negatively regulates the proliferation of JAK2V617F HSCs [303], and its expression is inhibited by the action of noradrenaline, which is released by sympathetic nerve fibres, on the β-adrenergic receptors of nestin+ MSCs [304,305]. In MPN, LSCs secretes IL-1β, leading to Schwann cell death and subsequent sympathetic neuropathy [256,303,305]. This results in apoptosis of nestin+ MSCs [256]. β-3 sympathomimetic agonist restores sympathetic regulation of nestin+ MSCs. In JAK2V617F murine models, β-3 sympathomimetic agonist (BRL37344) rescued nestin+ MSC, reduced BM fibrosis and diminished MPN HPCs [256,303,305]. Mirabegron, another β-3 sympathomimetic agonist, showed some encouraging results in a phase II clinical trial [256] (Table 3).

12.2. Targeting Transforming Growth Factor-β (TGF-β)

BM fibrosis is a notable feature in MPN which involves increased matrix synthesis and decreased matrix degradation [306]. In MPN, TGF-β, coupled with FGF and PDGF, are secreted by neoplastic megakaryocytes. This results in collagen production and tenascin, fibronectin and proteoglycan deposition, increasing matrix synthesis [306,307]. TGF-β also downregulates matrix metalloproteinase (MMP) and upregulates tissue inhibitors of metalloproteinase (TIMP) to inhibit matrix degradation [306]. TGF-β receptor I kinase (ALK5) inhibitors, galunisertib, and SB431542 antagonize TGF-β via ALK5/mothers against decapentaplegic homolog 3 (Smad3) pathway [308,309]. Preclinical studies have shown reduced collagen deposition, BM fibrosis and splenomegaly upon galunisertib treatment [308]. Activin receptor IIA ligand trap (ActRIIA) (e.g., luspatercept, sotatercept) is another novel agent that sequester TGF-β superfamily ligands (e.g., GDF11, Activin A) to restore terminal erythroid differentiation and improve anemia. Clinical trials are reported [257,258] (Table 3).

12.3. Aurora Kinase A (AURKA) Inhibition

Malignant, atypical megakaryocytes in MPN suppress the expression of GATA binding protein 1 (GATA1), which is responsible for megakaryocyte differentiation and maturation [259,260,310]. MLN8237, an AURKA inhibitor, showed efficacy in enhancing GATA1 expression, promoting megakaryocyte differentiation and polyploidization, as well as reducing BM fibrosis in animal models [310]. Additionally, MLN8237 and ruxolitinib produced synergistic effects to eliminate BM fibrosis and reduced burden of immature megakaryocytes [310]. Clinical investigations of alisertib, another AURKA inhibitor, demonstrated promising results [259,260] (Table 3).

12.4. Antifibrotic Therapy

The pentraxin family consists of C-reactive protein (CRP/PTX1), pentraxin-2 (serum amyloid P, SAP), and pentraxin-3. SAP is a 125-kD protein synthesized by hepatocytes to inhibit differentiation from monocytes to fibrocytes and further fibrocyte proliferation [311]. Besides the release of growth factors from atypical megakaryocytes, neoplastic fibrocytes also play a vital role in inducing BM fibrosis. Analysis via quantitative allele-specific PCR showed the presence of JAK2V617F and CALR mutations in fibrocytes, but not MSCs, suggesting that these fibrocytes were derived from a malignant clone [312]. PRM-151 is a recombinant SAP which inhibits PMF fibrocyte differentiation in BM and spleen to slow down development of fibrosis [312]. Due to the unique activity of PRM-151, it is being tested in clinical trials in combination with ruxolitinib [313,314].

13. Immunotherapeutic Targetting of MPN Stem Cells

13.1. Targeting PD-1/PD-L1 Pathway

An elevated expression of PD-1/PD-L1 has been observed in all 3 types of classical Ph-negative MPNs [273,315,316]. In MPN, JAK2V617F increases phosphorylation of STAT3 and STAT5 to promote PD-L1 expression mainly on the surfaces of monocytes, MDSCs, megakaryocytes and platelets [317]. In preclinical studies, a positive feedback was shown in MDSCs, in which it interacted with T cells and resulted in IL-10 secretion by activated T cells, hence resulting in the phosphorylation of STAT3 and induction of PD-L1 expression in MDSCs [273,315,318]. Furthermore, PV and ET patients have increased toll-like receptor 2 (TLR2) levels. This activates the MEK/ERK and STAT pathway, enhancing expression of PD-L1 [273,315]. All these contribute to the oncogene-mediated immune escape via JAK2/STAT3/STAT5/PD-L1 axis. The enhanced PD-L1 expressed in MPN binds to PD-1 on T cells to suppress their cysteine metabolism. This led to anergy, decreased cell cycle activity and exhaustion of T cells [317]. Several anti-PD-1 and anti-PD-L1 antibodies are developed with ongoing clinical trials. Their results, however, were not very encouraging [319,320,321] (Table 3).

13.2. Peptide Vaccination in CALR Exon 9

Mutant CALR possesses a new and large C-terminal peptide sequence which is completely distinct from wild-type CALR. This warrants interests in using CALR as an immunotherapeutic target [322,323]. In MPN, mutant CALR is overexpressed. This reduces MHC-I assembly and loading on cell surface, and impairs CD8+ T-lymphocyte targeting [323]. Interestingly, it was observed that some healthy donors harboured CD4+ memory T-cells towards CALR epitopes [323]. Based on the above observation, a CALR-mutated CD4+ T-lymphocyte clone has been designed to induce cytotoxic effects against CALR-mutated cells [323]. A phase I clinical trial is underway, but no result has been released yet [324] (Table 3).

14. Other Potential Therapeutic Strategies Targeting MPN Stem Cells

Several new approaches have been observed to potentially target MPN stem cells, either as single agents or in combination with Peg-IFNα-2a or ruxolitinib (Table 4 and Table 5).

15. Conclusions

Understanding the biology of CML and Ph-negative MPNs is an important area of scientific research. With considerable data providing insights into the biology and therapeutic targeting abnormal HSCs in CML, efforts towards identifying and quantifying abnormal HSCs in CML may facilitate efforts in achieving treatment-free remissions. In Ph-negative MPNs, this understanding may shift the treatment paradigm from cytoreduction and symptom control to effective disease modification.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/1422-0067/22/2/659/s1, File S1: Prognostic models for myelofibrosis. File S2: Molecular pathways in chronic myeloid leukemia and their therapeutic targeting. File S3: The bone marrow microenvironment in chronic myeloid leukemia and its therapeutic targets. File S4: Immunologic pathways and their therapeutic targeting in chronic myeloid leukemia. File S5: Molecular pathways in Philadelphia chromosome-negative myeloproliferative neoplasms and their therapeutic targeting. File S6: The bone marrow microenvironment in Philadelphia chromosome-negative myeloproliferative neoplasms and its therapeutic targeting. File S7: Immunologic pathways and their therapeutic targeting in Philadelphia chromosome-negative myeloproliferative neoplasms. File S8: Legends and abbreviations for Files S2–S7.

Author Contributions

Conceptualization, H.G.; writing—original draft preparation, Y.Y., E.L., H.-T.C., P.-K.Y., and H.G.; writing—review and editing, H.G; supervision, H.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The project was conducted under the Enrichment Year (EY) of the Bachelor of Medicine and Bachelor of Surgery (MBBS) curriculum, LKS Faculty of Medicine, the University of Hong Kong.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tefferi, A. Myeloproliferative neoplasms: A decade of discoveries and treatment advances. Am. J. Hematol. 2016, 91, 50–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Shallis, R.M.; Wang, R.; Davidoff, A.; Ma, X.; Podoltsev, N.A.; Zeidan, A.M. Epidemiology of the classical myeloproliferative neoplasms: The four corners of an expansive and complex map. Blood Rev. 2020, 42, 100706. [Google Scholar] [CrossRef] [PubMed]
  3. Hultcrantz, M.; Ravn Landtblom, A.; Andréasson, B.; Samuelsson, J.; Dickman, P.W.; Kristinsson, S.Y.; Björkholm, M.; Andersson, T.M. Incidence of myeloproliferative neoplasms—Trends by subgroup and age in a population-based study in Sweden. J. Intern. Med. 2020, 287, 448–454. [Google Scholar] [CrossRef] [PubMed]
  4. Pellicano, F.; Park, L.; Hopcroft, L.E.M.; Shah, M.M.; Jackson, L.; Scott, M.T.; Clarke, C.J.; Sinclair, A.; Abraham, S.A.; Hair, A.; et al. hsa-mir183/EGR1-mediated regulation of E2F1 is required for CML stem/progenitor cell survival. Blood 2018, 131, 1532–1544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Kvasnicka, H.M. The differential diagnosis of classical myeloproliferative neoplasms (MPN): The updated WHO criteria. Rinsho Ketsueki 2019, 60, 1166–1175. [Google Scholar]
  6. Passamonti, F.; Maffioli, M. Update from the latest WHO classification of MPNs: A user’s manual. Hematology 2016, 2016, 534–542. [Google Scholar] [CrossRef] [Green Version]
  7. Pophali, P.A.; Patnaik, M.M. The Role of New Tyrosine Kinase Inhibitors in Chronic Myeloid Leukemia. Cancer J. 2016, 22, 40–50. [Google Scholar] [CrossRef] [Green Version]
  8. Vetrie, D.; Helgason, G.V.; Copland, M. The leukaemia stem cell: Similarities, differences and clinical prospects in CML and AML. Nat. Rev. Cancer 2020, 20, 158–173. [Google Scholar] [CrossRef]
  9. Mead, A.J.; Mullally, A. Myeloproliferative neoplasm stem cells. Blood 2017, 129, 1607–1616. [Google Scholar] [CrossRef] [Green Version]
  10. Nangalia, J.; Green, A.R. Myeloproliferative neoplasms: From origins to outcomes. Blood 2017, 130, 2475–2483. [Google Scholar] [CrossRef]
  11. Grabek, J.; Straube, J.; Bywater, M.; Lane, S.W. MPN: The Molecular Drivers of Disease Initiation, Progression and Transformation and their Effect on Treatment. Cells 2020, 9, 1901. [Google Scholar] [CrossRef] [PubMed]
  12. Vainchenker, W.; Kralovics, R. Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms. Blood 2017, 129, 667–679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Jang, M.-A.; Choi, C.W. Recent insights regarding the molecular basis of myeloproliferative neoplasms. Korean J. Intern. Med. 2020, 35, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Allegra, A.; Pioggia, G.; Tonacci, A.; Casciaro, M.; Musolino, C.; Gangemi, S. Synergic Crosstalk between Inflammation, Oxidative Stress, and Genomic Alterations in BCR–ABL-Negative Myeloproliferative Neoplasm. Antioxidants 2020, 9, 1037. [Google Scholar] [CrossRef] [PubMed]
  15. Zhou, H.; Xu, R. Leukemia stem cells: The root of chronic myeloid leukemia. Protein Cell 2015, 6, 403–412. [Google Scholar] [CrossRef] [Green Version]
  16. Herrmann, H.; Sadovnik, I.; Cerny-Reiterer, S.; Rülicke, T.; Stefanzl, G.; Willmann, M.; Hoermann, G.; Bilban, M.; Blatt, K.; Herndlhofer, S.; et al. Dipeptidylpeptidase IV (CD26) defines leukemic stem cells (LSC) in chronic myeloid leukemia. Blood 2014, 123, 3951–3962. [Google Scholar] [CrossRef] [Green Version]
  17. Houshmand, M.; Simonetti, G.; Circosta, P.; Gaidano, V.; Cignetti, A.; Martinelli, G.; Saglio, G.; Gale, R.P. Chronic myeloid leukemia stem cells. Leukemia 2019, 33, 1543–1556. [Google Scholar] [CrossRef] [Green Version]
  18. Bhatia, R. Novel approaches to therapy in CML. Hematol. Am. Soc. Hematol. Educ. Program 2017, 2017, 115–120. [Google Scholar] [CrossRef] [Green Version]
  19. Hehlmann, R. Chronic Myeloid Leukemia in 2020. Hemasphere 2020, 4, e468. [Google Scholar] [CrossRef]
  20. Ye, H.; Adane, B.; Khan, N.; Sullivan, T.; Minhajuddin, M.; Gasparetto, M.; Stevens, B.; Pei, S.; Balys, M.; Ashton, J.M.; et al. Leukemic Stem Cells Evade Chemotherapy by Metabolic Adaptation to an Adipose Tissue Niche. Cell Stem Cell 2016, 19, 23–37. [Google Scholar] [CrossRef] [Green Version]
  21. Valent, P.; Sadovnik, I.; Eisenwort, G.; Bauer, K.; Herrmann, H.; Gleixner, K.V.; Schulenburg, A.; Rabitsch, W.; Sperr, W.R.; Wolf, D. Immunotherapy-Based Targeting and Elimination of Leukemic Stem Cells in AML and CML. Int. J. Mol. Sci. 2019, 20, 4233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Tanaka, Y.; Fukushima, T.; Mikami, K.; Adachi, K.; Fukuyama, T.; Goyama, S.; Kitamura, T. Efficacy of tyrosine kinase inhibitors on a mouse chronic myeloid leukemia model and chronic myeloid leukemia stem cells. Exp. Hematol. 2020, 90, 46–51.e2. [Google Scholar] [CrossRef] [PubMed]
  23. Houshmand, M.; Blanco, T.M.; Circosta, P.; Yazdi, N.; Kazemi, A.; Saglio, G.; Zarif, M.N. Bone marrow microenvironment: The guardian of leukemia stem cells. World J. Stem Cells 2019, 11, 476–490. [Google Scholar] [CrossRef] [PubMed]
  24. Helgason, G.V.; Mukhopadhyay, A.; Karvela, M.; Salomoni, P.; Calabretta, B.; Holyoake, T.L. Autophagy in chronic myeloid leukaemia: Stem cell survival and implication in therapy. Curr. Cancer Drug Targets 2013, 13, 724–734. [Google Scholar] [CrossRef] [PubMed]
  25. Arrigoni, E.; Del Re, M.; Galimberti, S.; Restante, G.; Rofi, E.; Crucitta, S.; Baratè, C.; Petrini, M.; Danesi, R.; Di Paolo, A. Concise Review: Chronic Myeloid Leukemia: Stem Cell Niche and Response to Pharmacologic Treatment. Stem Cells Transl. Med. 2018, 7, 305–314. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Baquero, P.; Dawson, A.; Mukhopadhyay, A.; Kuntz, E.M.; Mitchell, R.; Olivares, O.; Ianniciello, A.; Scott, M.T.; Dunn, K.; Nicastri, M.C.; et al. Targeting quiescent leukemic stem cells using second generation autophagy inhibitors. Leukemia 2019, 33, 981–994. [Google Scholar] [CrossRef] [Green Version]
  27. Zhou, H.; Mak, P.Y.; Mu, H.; Mak, D.H.; Zeng, Z.; Cortes, J.; Liu, Q.; Andreeff, M.; Carter, B.Z. Combined inhibition of β-catenin and Bcr-Abl synergistically targets tyrosine kinase inhibitor-resistant blast crisis chronic myeloid leukemia blasts and progenitors in vitro and in vivo. Leukemia 2017, 31, 2065–2074. [Google Scholar] [CrossRef]
  28. Zhang, H.; Li, H.; Xi, H.S.; Li, S. HIF1α is required for survival maintenance of chronic myeloid leukemia stem cells. Blood 2012, 119, 2595–2607. [Google Scholar] [CrossRef] [Green Version]
  29. Carrà, G.; Cartellà, A.; Maffeo, B.; Morotti, A. Strategies For Targeting Chronic Myeloid Leukaemia Stem Cells. Blood Lymphat. Cancer 2019, 9, 45–52. [Google Scholar] [CrossRef] [Green Version]
  30. Zhang, B.; Li, L.; Ho, Y.; Li, M.; Marcucci, G.; Tong, W.; Bhatia, R. Heterogeneity of leukemia-initiating capacity of chronic myelogenous leukemia stem cells. J. Clin. Invest. 2016, 126, 975–991. [Google Scholar] [CrossRef] [Green Version]
  31. Herrmann, H.; Cerny-Reiterer, S.; Gleixner, K.V.; Blatt, K.; Herndlhofer, S.; Rabitsch, W.; Jäger, E.; Mitterbauer-Hohendanner, G.; Streubel, B.; Selzer, E.; et al. CD34(+)/CD38(-) stem cells in chronic myeloid leukemia express Siglec-3 (CD33) and are responsive to the CD33-targeting drug gemtuzumab/ozogamicin. Haematologica 2012, 97, 219–226. [Google Scholar] [CrossRef] [PubMed]
  32. Chaar, M.; Kamta, J.; Ait-Oudhia, S. Mechanisms, monitoring, and management of tyrosine kinase inhibitors-associated cardiovascular toxicities. Onco. Targets Ther. 2018, 11, 6227–6237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Jiao, Q.; Bi, L.; Ren, Y.; Song, S.; Wang, Q.; Wang, Y.-s. Advances in studies of tyrosine kinase inhibitors and their acquired resistance. Mol. Cancer 2018, 17, 36. [Google Scholar] [CrossRef] [PubMed]
  34. Jiang, L.; Wang, H.; Zhu, X.; Liu, W.; Zhou, S.; Geng, Z.; Xiao, Y.; Zou, P.; You, Y.; Li, Q.; et al. The Impact of Tyrosine Kinase Inhibitors on Chronic Myeloid Leukemia Stem Cells and the Implication in Discontinuation. Stem Cells Dev. 2019, 28, 1480–1485. [Google Scholar] [CrossRef] [PubMed]
  35. Hamad, A.; Sahli, Z.; El Sabban, M.; Mouteirik, M.; Nasr, R. Emerging Therapeutic Strategies for Targeting Chronic Myeloid Leukemia Stem Cells. Stem Cells Int. 2013, 2013, 1–12. [Google Scholar] [CrossRef] [Green Version]
  36. Willmann, M.; Sadovnik, I.; Eisenwort, G.; Entner, M.; Bernthaler, T.; Stefanzl, G.; Hadzijusufovic, E.; Berger, D.; Herrmann, H.; Hoermann, G.; et al. Evaluation of cooperative antileukemic effects of nilotinib and vildagliptin in Ph + chronic myeloid leukemia. Exp. Hematol. 2018, 57, 50–59.e6. [Google Scholar] [CrossRef]
  37. Pungolino, E.; Rossi, G.; De Canal, G.; Trojani, A.; D’Adda, M.; Perego, A.; Orlandi, E.M.; Lunghi, F.; Turrini, M.; Borin, L.; et al. Nilotinib induced bone marrow CD34+/lin-Ph+ cells early clearance in newly diagnosed CP-chronic myeloid leukemia. Am. J. Hematol. 2018, 93, E162–E164. [Google Scholar] [CrossRef]
  38. Konig, H.; Holyoake, T.L.; 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, 2329–2338. [Google Scholar] [CrossRef]
  39. Ciarcia, R.; Vitiello, M.T.; Galdiero, M.; Pacilio, C.; Iovane, V.; d’Angelo, D.; Pagnini, D.; Caparrotti, G.; Conti, D.; Tomei, V.; et al. Imatinib treatment inhibit IL-6, IL-8, NF-KB and AP-1 production and modulate intracellular calcium in CML patients. J. Cell Physiol. 2012, 227, 2798–2803. [Google Scholar] [CrossRef]
  40. Corrado, C.; Saieva, L.; Raimondo, S.; Santoro, A.; De Leo, G.; Alessandro, R. Chronic myelogenous leukaemia exosomes modulate bone marrow microenvironment through activation of epidermal growth factor receptor. J. Cell Mol. Med. 2016, 20, 1829–1839. [Google Scholar] [CrossRef] [Green Version]
  41. Masamoto, Y.; Kurokawa, M. Targeting chronic myeloid leukemia stem cells: Can transcriptional program be a druggable target for cancers? Stem Cell Investig. 2018, 5, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Ciarcia, R.; Damiano, S.; Puzio, M.V.; Montagnaro, S.; Pagnini, F.; Pacilio, C.; Caparrotti, G.; Bellan, C.; Garofano, T.; Polito, M.S.; et al. Comparison of Dasatinib, Nilotinib, and Imatinib in the Treatment of Chronic Myeloid Leukemia. J. Cell Physiol. 2016, 231, 680–687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Scott, M.T.; Korfi, K.; Saffrey, P.; Hopcroft, L.E.; Kinstrie, R.; Pellicano, F.; Guenther, C.; Gallipoli, P.; Cruz, M.; Dunn, K.; et al. Epigenetic Reprogramming Sensitizes CML Stem Cells to Combined EZH2 and Tyrosine Kinase Inhibition. Cancer Discov. 2016, 6, 1248–1257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Lee, M.Y.; Park, C.-j.; Cho, Y.-U.; You, E.; Jang, S.; Seol, C.A.; Seo, E.-J.; Lee, J.-H. Expression Levels of PD-1 on CD8+ T Cells in Chronic Myeloid Leukemia with and without BCR-ABL1 kinase Mutation. Blood 2017, 130 (Suppl. 1), 4178. [Google Scholar]
  45. Lee, M.Y.; Park, C.-J.; Cho, Y.-U.; You, E.; Jang, S.; Seol, C.A.; Seo, E.-J.; Choi, E.-J.; Lee, J.-H. Differences in PD-1 expression on CD8+ T-cells in chronic myeloid leukemia patients according to disease phase and TKI medication. Cancer Immunol. Immunother. 2020, 69, 2223–2232. [Google Scholar] [CrossRef] [PubMed]
  46. Hughes, A.; Clarson, J.; Tang, C.; Vidovic, L.; White, D.L.; Hughes, T.P.; Yong, A.S. CML patients with deep molecular responses to TKI have restored immune effectors and decreased PD-1 and immune suppressors. Blood 2017, 129, 1166–1176. [Google Scholar] [CrossRef]
  47. Molica, M.; Scalzulli, E.; Colafigli, G.; Foà, R.; Breccia, M. Insights into the optimal use of ponatinib in patients with chronic phase chronic myeloid leukaemia. Ther. Adv. Hematol. 2019, 10, 2040620719826444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Jabbour, E.; Kantarjian, H.; Cortes, J. Use of second- and third-generation tyrosine kinase inhibitors in the treatment of chronic myeloid leukemia: An evolving treatment paradigm. Clin. Lymphoma Myeloma Leuk. 2015, 15, 323–334. [Google Scholar] [CrossRef] [Green Version]
  49. Eide, C.A.; Zabriskie, M.S.; Savage Stevens, S.L.; Antelope, O.; Vellore, N.A.; Than, H.; Schultz, A.R.; Clair, P.; Bowler, A.D.; Pomicter, A.D.; et al. Combining the Allosteric Inhibitor Asciminib with Ponatinib Suppresses Emergence of and Restores Efficacy against Highly Resistant BCR-ABL1 Mutants. Cancer Cell 2019, 36, 431–443.e5. [Google Scholar] [CrossRef]
  50. Jabbour, E.; Kantarjian, H. Chronic myeloid leukemia: 2020 update on diagnosis, therapy and monitoring. Am. J. Hematol. 2020, 95, 691–709. [Google Scholar] [CrossRef]
  51. Hughes, T.P.; Mauro, M.J.; Cortes, J.E.; Minami, H.; Rea, D.; DeAngelo, D.J.; Breccia, M.; Goh, Y.-T.; Talpaz, M.; Hochhaus, A.; et al. Asciminib in Chronic Myeloid Leukemia after ABL Kinase Inhibitor Failure. N. Engl. J. Med. 2019, 381, 2315–2326. [Google Scholar] [CrossRef] [PubMed]
  52. Kavanagh, S.; Nee, A.; Lipton, J.H. Emerging alternatives to tyrosine kinase inhibitors for treating chronic myeloid leukemia. Expert Opin. Emerg. Drugs 2018, 23, 51–62. [Google Scholar] [CrossRef] [PubMed]
  53. Özgür Yurttaş, N.; Eşkazan, A.E. Novel therapeutic approaches in chronic myeloid leukemia. Leuk. Res. 2020, 91, 106337. [Google Scholar] [CrossRef] [PubMed]
  54. Talpaz, M.; Mercer, J.; Hehlmann, R. The interferon-alpha revival in CML. Ann. Hematol. 2015, 94 (Suppl. 2), S195–S207. [Google Scholar] [CrossRef] [PubMed]
  55. Talpaz, M.; Hehlmann, R.; Quintás-Cardama, A.; Mercer, J.; Cortes, J. Re-emergence of interferon-α in the treatment of chronic myeloid leukemia. Leukemia 2013, 27, 803–812. [Google Scholar] [CrossRef] [Green Version]
  56. Chawla-Sarkar, M.; Lindner, D.J.; Liu, Y.F.; Williams, B.R.; Sen, G.C.; Silverman, R.H.; Borden, E.C. Apoptosis and interferons: Role of interferon-stimulated genes as mediators of apoptosis. Apoptosis 2003, 8, 237–249. [Google Scholar] [CrossRef]
  57. Bhatia, R.; McCarthy, J.B.; Verfaillie, C.M. Interferon-alpha restores normal beta 1 integrin-mediated inhibition of hematopoietic progenitor proliferation by the marrow microenvironment in chronic myelogenous leukemia. Blood 1996, 87, 3883–3891. [Google Scholar] [CrossRef] [Green Version]
  58. Kunisaki, Y.; Bruns, I.; Scheiermann, C.; Ahmed, J.; Pinho, S.; Zhang, D.; Mizoguchi, T.; Wei, Q.; Lucas, D.; Ito, K.; et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature 2013, 502, 637–643. [Google Scholar] [CrossRef] [Green Version]
  59. Chronic Myeloid Leukemia Trialists’ Collaborative Group. Interferon Alfa Versus Chemotherapy for Chronic Myeloid Leukemia: A Meta-analysis of Seven Randomized Trials. JNCI J. Natl. Cancer Inst. 1997, 89, 1616–1620. [Google Scholar] [CrossRef] [Green Version]
  60. Bonifazi, F.; Vivo, A.; Rosti, G.; Guilhot, F.; Guilhot, J.; Trabacchi, E.; Hehlmann, R.; Hochhaus, A.; Shepherd, P.; Steegmann, J.; et al. Chronic myeloid leukemia and interferon-alpha: A study of complete cytogenetic responders. Blood 2001, 98, 3074–3081. [Google Scholar] [CrossRef]
  61. Tefferi, A.; Barbui, T. Polycythemia vera and essential thrombocythemia: 2019 update on diagnosis, risk-stratification and management. Am. J. Hematol. 2019, 94, 133–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. How, J.; Hobbs, G. Use of Interferon Alfa in the Treatment of Myeloproliferative Neoplasms: Perspectives and Review of the Literature. Cancers 2020, 12, 1954. [Google Scholar] [CrossRef] [PubMed]
  63. Lu, M.; Zhang, W.; Li, Y.; Berenzon, D.; Wang, X.; Wang, J.; Mascarenhas, J.; Xu, M.; Hoffman, R. Interferon-α targets JAK2V617F-positive hematopoietic progenitor cells and acts through the p38 MAPK pathway. Exp. Hematol. 2010, 38, 472–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Kiladjian, J.-J.; Mesa, R.A.; Hoffman, R. The renaissance of interferon therapy for the treatment of myeloid malignancies. Blood 2011, 117, 4706–4715. [Google Scholar] [CrossRef] [Green Version]
  65. Utke Rank, C.; Weis Bjerrum, O.; Larsen, T.S.; Kjær, L.; De Stricker, K.; Riley, C.H.; Hasselbalch, H.C. Minimal residual disease after long-term interferon-alpha2 treatment: A report on hematological, molecular and histomorphological response patterns in 10 patients with essential thrombocythemia and polycythemia vera. Leuk. Lymphoma 2016, 57, 348–354. [Google Scholar] [CrossRef]
  66. Skov, V. Next Generation Sequencing in MPNs. Lessons from the Past and Prospects for Use as Predictors of Prognosis and Treatment Responses. Cancers 2020, 12, 2194. [Google Scholar] [CrossRef]
  67. Hasselbalch, H.C.; Holmström, M.O. Perspectives on interferon-alpha in the treatment of polycythemia vera and related myeloproliferative neoplasms: Minimal residual disease and cure? Semin. Immunopathol. 2019, 41, 5–19. [Google Scholar] [CrossRef] [Green Version]
  68. Austin, R.J.; Straube, J.; Bruedigam, C.; Pali, G.; Jacquelin, S.; Vu, T.; Green, J.; Gräsel, J.; Lansink, L.; Cooper, L.; et al. Distinct effects of ruxolitinib and interferon-alpha on murine JAK2V617F myeloproliferative neoplasm hematopoietic stem cell populations. Leukemia 2020, 34, 1075–1089. [Google Scholar] [CrossRef]
  69. Verger, E.; Soret-Dulphy, J.; Maslah, N.; Roy, L.; Rey, J.; Ghrieb, Z.; Kralovics, R.; Gisslinger, H.; Grohmann-Izay, B.; Klade, C.; et al. Ropeginterferon alpha-2b targets JAK2V617F-positive polycythemia vera cells in vitro and in vivo. Blood Cancer J. 2018, 8, 94. [Google Scholar] [CrossRef] [Green Version]
  70. Czech, J.; Cordua, S.; Weinbergerova, B.; Baumeister, J.; Crepcia, A.; Han, L.; Maié, T.; Costa, I.G.; Denecke, B.; Maurer, A.; et al. JAK2V617F but not CALR mutations confer increased molecular responses to interferon-α via JAK1/STAT1 activation. Leukemia 2019, 33, 995–1010. [Google Scholar] [CrossRef]
  71. Mosca, M.; Lamrani, L.; Marzac, C.; Tisserand, A.; Edmond, V.; Dagher, T.; El-Khoury, M.; Marty, C.; Campario, H.; Casadevall, N.; et al. Differential Impact of Interferon Alpha on JAK2V617F and Calr Mutated Hematopoietic Stem and Progenitor Cells in Classical MPN. Blood 2018, 132 (Suppl. 1), 4333. [Google Scholar] [CrossRef]
  72. Kjær, L.; Cordua, S.; Holmström, M.O.; Thomassen, M.; Kruse, T.A.; Pallisgaard, N.; Larsen, T.S.; De Stricker, K.; Skov, V.; Hasselbalch, H.C. Differential Dynamics of CALR Mutant Allele Burden in Myeloproliferative Neoplasms during Interferon Alfa Treatment. PLoS ONE 2016, 11, e0165336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Verger, E.; Cassinat, B.; Chauveau, A.; Dosquet, C.; Giraudier, S.; Schlageter, M.-H.; Ianotto, J.-C.; Yassin, M.A.; Al-Dewik, N.; Carillo, S.; et al. Clinical and molecular response to interferon-α therapy in essential thrombocythemia patients with CALR mutations. Blood 2015, 126, 2585–2591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Chifotides, H.T.; Bose, P.; Verstovsek, S. Givinostat: An emerging treatment for polycythemia vera. Expert Opin. Investig. Drugs 2020, 29, 525–536. [Google Scholar] [CrossRef] [PubMed]
  75. Passamonti, F.; Griesshammer, M.; Palandri, F.; Egyed, M.; Benevolo, G.; Devos, T.; Callum, J.; Vannucchi, A.M.; Sivgin, S.; Bensasson, C.; et al. Ruxolitinib for the treatment of inadequately controlled polycythaemia vera without splenomegaly (RESPONSE-2): A randomised, open-label, phase 3b study. Lancet Oncol. 2017, 18, 88–99. [Google Scholar] [CrossRef]
  76. Verstovsek, S.; Gotlib, J.; Mesa, R.A.; Vannucchi, A.M.; Kiladjian, J.-J.; Cervantes, F.; Harrison, C.N.; Paquette, R.; Sun, W.; Naim, A.; et al. Long-term survival in patients treated with ruxolitinib for myelofibrosis: COMFORT-I and -II pooled analyses. J. Hematol. Oncol. 2017, 10, 156. [Google Scholar] [CrossRef] [Green Version]
  77. Oh, S.T.; Talpaz, M.; Gerds, A.T.; Gupta, V.; Verstovsek, S.; Mesa, R.; Miller, C.B.; Rivera, C.E.; Fleischman, A.G.; Goel, S.; et al. ACVR1/JAK1/JAK2 inhibitor momelotinib reverses transfusion dependency and suppresses hepcidin in myelofibrosis phase 2 trial. Blood Adv. 2020, 4, 4282–4291. [Google Scholar] [CrossRef]
  78. Mesa, R.A.; Kiladjian, J.-J.; Catalano, J.V.; Devos, T.; Egyed, M.; Hellmann, A.; Mclornan, D.; Shimoda, K.; Winton, E.F.; Deng, W.; et al. SIMPLIFY-1: A Phase III Randomized Trial of Momelotinib Versus Ruxolitinib in Janus Kinase Inhibitor–Naïve Patients With Myelofibrosis. J. Clin. Oncol. 2017, 35, 3844–3850. [Google Scholar] [CrossRef]
  79. Talpaz, M.; Kiladjian, J.-J. Fedratinib, a newly approved treatment for patients with myeloproliferative neoplasm-associated myelofibrosis. Leukemia 2020. [Google Scholar] [CrossRef]
  80. Bose, P.; Verstovsek, S. JAK Inhibition for the Treatment of Myelofibrosis: Limitations and Future Perspectives. HemaSphere 2020, 4, e424. [Google Scholar] [CrossRef]
  81. Mascarenhas, J.; Hoffman, R.; Talpaz, M.; Gerds, A.T.; Stein, B.; Gupta, V.; Szoke, A.; Drummond, M.; Pristupa, A.; Granston, T.; et al. Pacritinib vs Best Available Therapy, Including Ruxolitinib, in Patients With Myelofibrosis. JAMA Oncol. 2018, 4, 652. [Google Scholar] [CrossRef] [PubMed]
  82. Harrison, C.N.; Schaap, N.; Mesa, R.A. Management of myelofibrosis after ruxolitinib failure. Ann. Hematol. 2020, 99, 1177–1191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Pardanani, A.; Tefferi, A.; Jamieson, C.; Gabrail, N.Y.; Lebedinsky, C.; Gao, G.; Liu, F.; Xu, C.; Cao, H.; Talpaz, M. A phase 2 randomized dose-ranging study of the JAK2-selective inhibitor fedratinib (SAR302503) in patients with myelofibrosis. Blood Cancer J. 2015, 5, e335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Pardanani, A.; Gotlib, J.R.; Jamieson, C.; Cortes, J.E.; Talpaz, M.; Stone, R.M.; Silverman, M.H.; Gilliland, D.G.; Shorr, J.; Tefferi, A. Safety and Efficacy of TG101348, a Selective JAK2 Inhibitor, in Myelofibrosis. J. Clin. Oncol. 2011, 29, 789–796. [Google Scholar] [CrossRef] [PubMed]
  85. Pardanani, A.; Harrison, C.; Cortes, J.E.; Cervantes, F.; Mesa, R.A.; Milligan, D.; Masszi, T.; Mishchenko, E.; Jourdan, E.; Vannucchi, A.M.; et al. Safety and Efficacy of Fedratinib in Patients With Primary or Secondary Myelofibrosis. JAMA Oncol. 2015, 1, 643. [Google Scholar] [CrossRef]
  86. Bankar, A.; Gupta, V. Investigational non-JAK inhibitors for chronic phase myelofibrosis. Expert Opin. Investig. Drugs 2020, 29, 461–474. [Google Scholar] [CrossRef]
  87. Tamari, R.; Rapaport, F.; Zhang, N.; Mcnamara, C.; Kuykendall, A.; Sallman, D.A.; Komrokji, R.; Arruda, A.; Najfeld, V.; Sandy, L.; et al. Impact of High-Molecular-Risk Mutations on Transplantation Outcomes in Patients with Myelofibrosis. Biol. Blood Marrow Transplant. 2019, 25, 1142–1151. [Google Scholar] [CrossRef]
  88. Kröger, N.; Panagiota, V.; Badbaran, A.; Zabelina, T.; Triviai, I.; Araujo Cruz, M.M.; Shahswar, R.; Ayuk, F.; Gehlhaar, M.; Wolschke, C.; et al. Impact of Molecular Genetics on Outcome in Myelofibrosis Patients after Allogeneic Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2017, 23, 1095–1101. [Google Scholar] [CrossRef] [Green Version]
  89. Mannina, D.; Gagelmann, N.; Badbaran, A.; Ditschkowski, M.; Bogdanov, R.; Robin, M.; Cassinat, B.; Heuser, M.; Shahswar, R.; Thol, F.; et al. Allogeneic stem cell transplantation in patients with myelofibrosis harboring the MPL mutation. Eur. J. Haematol. 2019, 103, 552–557. [Google Scholar] [CrossRef]
  90. Mian, A.A.; Rafiei, A.; Haberbosch, I.; Zeifman, A.; Titov, I.; Stroylov, V.; Metodieva, A.; Stroganov, O.; Novikov, F.; Brill, B.; et al. PF-114, a potent and selective inhibitor of native and mutated BCR/ABL is active against Philadelphia chromosome-positive (Ph+) leukemias harboring the T315I mutation. Leukemia 2015, 29, 1104–1114. [Google Scholar] [CrossRef]
  91. Ivanova, E.S.; Tatarskiy, V.V.; Yastrebova, M.A.; Khamidullina, A.I.; Shunaev, A.V.; Kalinina, A.A.; Zeifman, A.A.; Novikov, F.N.; Dutikova, Y.V.; Chilov, G.G.; et al. PF-114, a novel selective inhibitor of BCR-ABL tyrosine kinase, is a potent inducer of apoptosis in chronic myelogenous leukemia cells. Int. J. Oncol. 2019, 55, 289–297. [Google Scholar] [CrossRef] [PubMed]
  92. Turkina, A.G.; Vinogradova, O.; Lomaia, E.; Shatokhina, E.; Shukhov, O.A.; Chelysheva, E.Y.; Shikhbabaeva, D.; Nemchenko, I.; Petrova, A.; Bykova, A.; et al. PF-114: A 4th Generation Tyrosine Kinase-Inhibitor for Chronic Phase Chronic Myeloid Leukaemia Including BCRABL1T315I. Blood 2019, 134 (Suppl. 1), 1638. [Google Scholar] [CrossRef]
  93. Rossari, F.; Minutolo, F.; Orciuolo, E. Past, present, and future of Bcr-Abl inhibitors: From chemical development to clinical efficacy. J. Hematol. Oncol. 2018, 11, 84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Di Stefano, C.; Mirone, G.; Perna, S.; Marfe, G. The roles of microRNAs in the pathogenesis and drug resistance of chronic myelogenous leukemia (Review). Oncol. Rep. 2016, 35, 614–624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Litwińska, Z.; Machaliński, B. miRNAs in chronic myeloid leukemia: Small molecules, essential function. Leuk. Lymphoma 2017, 58, 1297–1305. [Google Scholar] [CrossRef] [PubMed]
  96. Agatheeswaran, S.; Pattnayak, N.C.; Chakraborty, S. Identification and functional characterization of the miRNA-gene regulatory network in chronic myeloid leukemia lineage negative cells. Sci. Rep. 2016, 6, 32493. [Google Scholar] [CrossRef] [Green Version]
  97. Klümper, T.; Bruckmueller, H.; Diewock, T.; Kaehler, M.; Haenisch, S.; Pott, C.; Bruhn, O.; Cascorbi, I. Expression differences of miR-142-5p between treatment-naïve chronic myeloid leukemia patients responding and non-responding to imatinib therapy suggest a link to oncogenic ABL2, SRI, cKIT and MCL1 signaling pathways critical for development of therapy resistance. Exp. Hematol. Oncol. 2020, 9, 26. [Google Scholar] [PubMed]
  98. Salati, S.; Salvestrini, V.; Carretta, C.; Genovese, E.; Rontauroli, S.; Zini, R.; Rossi, C.; Ruberti, S.; Bianchi, E.; Barbieri, G.; et al. Deregulated expression of miR-29a-3p, miR-494-3p and miR-660-5p affects sensitivity to tyrosine kinase inhibitors in CML leukemic stem cells. Oncotarget 2017, 8, 49451–49469. [Google Scholar] [CrossRef] [Green Version]
  99. Fiserova, B.; Kubiczkova, L.; Sedlarikova, L.; Hajek, R.; Sevcikova, S. The miR-29 family in hematological malignancies. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc. Czech Repub. 2015, 159, 184–191. [Google Scholar] [CrossRef] [Green Version]
  100. Srutova, K.; Curik, N.; Burda, P.; Savvulidi, F.; Silvestri, G.; Trotta, R.; Klamova, H.; Pecherkova, P.; Sovova, Z.; Koblihova, J.; et al. BCR-ABL1 mediated miR-150 downregulation through MYC contributed to myeloid differentiation block and drug resistance in chronic myeloid leukemia. Haematologica 2018, 103, 2016–2025. [Google Scholar] [CrossRef]
  101. Li, Y.-L.; Tang, J.-M.; Chen, X.-Y.; Luo, B.; Liang, G.-H.; Qu, Q.; Lu, Z.-Y. MicroRNA-153-3p enhances the sensitivity of chronic myeloid leukemia cells to imatinib by inhibiting B-cell lymphoma-2-mediated autophagy. Hum. Cell 2020, 33, 610–618. [Google Scholar] [CrossRef] [PubMed]
  102. Pan, B.; Yang, J.; Wang, X.; Xu, K.; Ikezoe, T. miR-217 sensitizes chronic myelogenous leukemia cells to tyrosine kinase inhibitors by targeting pro-oncogenic anterior gradient 2. Exp. Hematol. 2018, 68, 80–88.e2. [Google Scholar] [CrossRef] [PubMed]
  103. Nishioka, C.; Ikezoe, T.; Yang, J.; Nobumoto, A.; Tsuda, M.; Yokoyama, A. Downregulation of miR-217 correlates with resistance of Ph(+) leukemia cells to ABL tyrosine kinase inhibitors. Cancer Sci. 2014, 105, 297–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Hershkovitz-Rokah, O.; Modai, S.; Pasmanik-Chor, M.; Toren, A.; Shomron, N.; Raanani, P.; Shpilberg, O.; Granot, G. Restoration of miR-424 suppresses BCR-ABL activity and sensitizes CML cells to imatinib treatment. Cancer Lett. 2015, 360, 245–256. [Google Scholar] [CrossRef]
  105. Han, B.; Zhao, Y.; Lin, Y.; Fu, S.; Wang, L.; Zhang, M.; Tie, R.; Wang, B.; Luo, Y.; Liu, L.; et al. Hydroxychloroquine sensitizes chronic myeloid leukemia cells to Vγ9Vδ2 T cell-mediated lysis independent of autophagy. Int. J. Oncol. 2017, 50, 1810–1820. [Google Scholar] [CrossRef] [Green Version]
  106. Lv, M.; Zhang, X.; Jia, H.; Li, D.; Zhang, B.; Zhang, H.; Hong, M.; Jiang, T.; Jiang, Q.; Lu, J.; et al. An oncogenic role of miR-142-3p in human T-cell acute lymphoblastic leukemia (T-ALL) by targeting glucocorticoid receptor-α and cAMP/PKA pathways. Leukemia 2012, 26, 769–777. [Google Scholar] [CrossRef] [Green Version]
  107. Shen, J.-Z.; Zhang, Y.-Y.; Fu, H.-Y.; Wu, D.-S.; Zhou, H.-R. Overexpression of microRNA-143 inhibits growth and induces apoptosis in human leukemia cells. Oncol. Rep. 2014, 31, 2035–2042. [Google Scholar] [CrossRef] [Green Version]
  108. Yap, E.; Norziha, Z.A.; Simbun, A.; Tumian, N.R.; Cheong, S.K.; Leong, C.F.; Wong, C.L. Downregulation of Mir-146a-5p, Mir-99b-5p, Mir-143-3p, Mir-10a-5p and Mir-151a-3p Associated with PI3K/AKT, p53, NF-Kb, and Fanconi Anemia/BRCA Signaling Pathways Are Observed in Imatinib-Resistant Chronic Myeloid Leukemia Patients without Detectable BCR-ABL kinase Domain Mutations. Blood 2016, 128, 3060. [Google Scholar]
  109. Flamant, S.; Ritchie, W.; Guilhot, J.; Holst, J.; Bonnet, M.L.; Chomel, J.C.; Guilhot, F.; Turhan, A.G.; Rasko, J.E. Micro-RNA response to imatinib mesylate in patients with chronic myeloid leukemia. Haematologica 2010, 95, 1325–1333. [Google Scholar] [CrossRef] [Green Version]
  110. Jurkovicova, D.; Lukackova, R.; Magyerkova, M.; Kulcsar, L.; Krivjanska, M.; Krivjansky, V.; Chovanec, M. microRNA expression profiling as supportive diagnostic and therapy prediction tool in chronic myeloid leukemia. Neoplasma 2015, 62, 949–958. [Google Scholar] [CrossRef]
  111. Modi, H.; Li, L.; Chu, S.; Rossi, J.; Yee, J.K.; Bhatia, R. Inhibition of Grb2 expression demonstrates an important role in BCR-ABL-mediated MAPK activation and transformation of primary human hematopoietic cells. Leukemia 2011, 25, 305–312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Chen, M.; Turhan, A.G.; Ding, H.; Lin, Q.; Meng, K.; Jiang, X. Targeting BCR-ABL+ stem/progenitor cells and BCR-ABL-T315I mutant cells by effective inhibition of the BCR-ABL-Tyr177-GRB2 complex. Oncotarget 2017, 8, 43662–43677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Tari Ashizawa, A.; Ohanian, M.; Cortes, J.E. BP1001, a Novel Therapeutic for Chronic Myelogenous Leukemia. Blood 2016, 128, 4239. [Google Scholar] [CrossRef]
  114. Peng, Z.; Luo, H.W.; Yuan, Y.; Shi, J.; Huang, S.F.; Li, C.L.; Cao, W.X.; Huang, Z.G.; Feng, W.L. Growth of chronic myeloid leukemia cells is inhibited by infection with Ad-SH2-HA adenovirus that disrupts Grb2-Bcr-Abl complexes. Oncol. Rep. 2011, 25, 1381–1388. [Google Scholar] [PubMed] [Green Version]
  115. Ma, L.; Shan, Y.; Bai, R.; Xue, L.; Eide, C.A.; Ou, J.; Zhu, L.J.; Hutchinson, L.; Cerny, J.; Khoury, H.J.; et al. A therapeutically targetable mechanism of BCR-ABL-independent imatinib resistance in chronic myeloid leukemia. Sci. Transl. Med. 2014, 6, 252ra121. [Google Scholar] [CrossRef] [Green Version]
  116. Chorzalska, A.; Ahsan, N.; Rao, R.S.P.; Roder, K.; Yu, X.; Morgan, J.; Tepper, A.; Hines, S.; Zhang, P.; Treaba, D.O.; et al. Overexpression of Tpl2 is linked to imatinib resistance and activation of MEK-ERK and NF-κB pathways in a model of chronic myeloid leukemia. Mol. Oncol. 2018, 12, 630–647. [Google Scholar] [CrossRef] [Green Version]
  117. Lim, S.; Saw, T.Y.; Zhang, M.; Janes, M.R.; Nacro, K.; Hill, J.; Lim, A.Q.; Chang, C.T.; Fruman, D.A.; Rizzieri, D.A.; et al. Targeting of the MNK-eIF4E axis in blast crisis chronic myeloid leukemia inhibits leukemia stem cell function. Proc. Natl. Acad. Sci. USA 2013, 110, E2298–E2307. [Google Scholar] [CrossRef] [Green Version]
  118. Jacquel, A.; Luciano, F.; Robert, G.; Auberger, P. Implication and Regulation of AMPK during Physiological and Pathological Myeloid Differentiation. Int. J. Mol. Sci. 2018, 19, 2991. [Google Scholar] [CrossRef] [Green Version]
  119. Dinner, S.; Platanias, L.C. Targeting the mTOR Pathway in Leukemia. J. Cell. Biochem. 2016, 117, 1745–1752. [Google Scholar] [CrossRef]
  120. Frazzi, R.; Guardi, M. Cellular and Molecular Targets of Resveratrol on Lymphoma and Leukemia Cells. Molecules 2017, 22, 885. [Google Scholar] [CrossRef] [Green Version]
  121. Vakana, E.; Platanias, L.C. AMPK in BCR-ABL expressing leukemias. Regulatory effects and therapeutic implications. Oncotarget 2011, 2, 1322–1328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Shafiei-Irannejad, V.; Samadi, N.; Salehi, R.; Yousefi, B.; Zarghami, N. New insights into antidiabetic drugs: Possible applications in cancer treatment. Chem. Biol. Drug Des. 2017, 90, 1056–1066. [Google Scholar] [CrossRef] [PubMed]
  123. Biondani, G.; Peyron, J.F. Metformin, an Anti-diabetic Drug to Target Leukemia. Front. Endocrinol 2018, 9, 446. [Google Scholar] [CrossRef] [Green Version]
  124. Shi, R.; Lin, J.; Gong, Y.; Yan, T.; Shi, F.; Yang, X.; Liu, X.; Naren, D. The antileukemia effect of metformin in the Philadelphia chromosome-positive leukemia cell line and patient primary leukemia cell. Anticancer Drugs 2015, 26, 913–922. [Google Scholar] [CrossRef]
  125. Wu, X.P.; Xiong, M.; Xu, C.S.; Duan, L.N.; Dong, Y.Q.; Luo, Y.; Niu, T.H.; Lu, C.R. Resveratrol induces apoptosis of human chronic myelogenous leukemia cells in vitro through p38 and JNK-regulated H2AX phosphorylation. Acta Pharmacol. Sin. 2015, 36, 353–361. [Google Scholar] [CrossRef]
  126. Puissant, A.; Robert, G.; Fenouille, N.; Luciano, F.; Cassuto, J.-P.; Raynaud, S.; Auberger, P. Resveratrol Promotes Autophagic Cell Death in Chronic Myelogenous Leukemia Cells via JNK-Mediated p62/SQSTM1 Expression and AMPK Activation. Cancer Res. 2010, 70, 1042–1052. [Google Scholar] [CrossRef] [Green Version]
  127. Kirchner, J.; Brüne, B.; Namgaladze, D. AICAR inhibits NFκB DNA binding independently of AMPK to attenuate LPS-triggered inflammatory responses in human macrophages. Sci. Rep. 2018, 8, 7801. [Google Scholar] [CrossRef] [Green Version]
  128. Carter, B.Z.; Mak, P.Y.; Mu, H.; Zhou, H.; Mak, D.H.; Schober, W.; Leverson, J.D.; Zhang, B.; Bhatia, R.; Huang, X.; et al. Combined targeting of BCL-2 and BCR-ABL tyrosine kinase eradicates chronic myeloid leukemia stem cells. Sci. Transl. Med. 2016, 8, 355ra117. [Google Scholar] [CrossRef] [Green Version]
  129. Mattes, K.; Gerritsen, M.; Folkerts, H.; Geugien, M.; van den Heuvel, F.A.; Svendsen, A.F.; Yi, G.; Martens, J.H.A.; Vellenga, E. CD34(+) acute myeloid leukemia cells with low levels of reactive oxygen species show increased expression of stemness genes and can be targeted by the BCL2 inhibitor venetoclax. Haematologica 2020, 105, e399–e403. [Google Scholar] [CrossRef]
  130. Maiti, A.; Franquiz, M.J.; Ravandi, F.; Cortes, J.E.; Jabbour, E.J.; Sasaki, K.; Marx, K.; Daver, N.G.; Kadia, T.M.; Konopleva, M.Y.; et al. Venetoclax and BCR-ABL Tyrosine Kinase Inhibitor Combinations: Outcome in Patients with Philadelphia Chromosome-Positive Advanced Myeloid Leukemias. Acta Haematol. 2020, 143, 567–573. [Google Scholar] [CrossRef]
  131. Gallipoli, P.; Cook, A.; Rhodes, S.; Hopcroft, L.; Wheadon, H.; Whetton, A.D.; Jørgensen, H.G.; Bhatia, R.; Holyoake, T.L. JAK2/STAT5 inhibition by nilotinib with ruxolitinib contributes to the elimination of CML CD34+ cells in vitro and in vivo. Blood 2014, 124, 1492–1501. [Google Scholar] [CrossRef] [PubMed]
  132. Chen, M.; Gallipoli, P.; Degeer, D.; Sloma, I.; Forrest, D.L.; Chan, M.; Lai, D.; Jorgensen, H.; Ringrose, A.; Wang, H.M.; et al. Targeting Primitive Chronic Myeloid Leukemia Cells by Effective Inhibition of a New AHI-1-BCR-ABL-JAK2 Complex. J. Natl. Cancer Inst. 2013, 105, 405–423. [Google Scholar] [CrossRef] [PubMed]
  133. Sweet, K.; Hazlehurst, L.; Sahakian, E.; Powers, J.; Nodzon, L.; Kayali, F.; Hyland, K.; Nelson, A.; Pinilla-Ibarz, J. A Phase I Study of Ruxolitinib Plus Nilotinib in Chronic Phase CML Patients with Molecular Evidence of Disease. Blood 2016, 128, 1892. [Google Scholar] [CrossRef]
  134. Sweet, K.; Hazlehurst, L.; Sahakian, E.; Powers, J.; Nodzon, L.; Kayali, F.; Hyland, K.; Nelson, A.; Pinilla-Ibarz, J. A phase I clinical trial of ruxolitinib in combination with nilotinib in chronic myeloid leukemia patients with molecular evidence of disease. Leuk. Res. 2018, 74, 89–96. [Google Scholar] [CrossRef] [PubMed]
  135. Dao, K.-H.; Collins, R.H.; Cortes, J.E.; Deininger, M.W.; Druker, B.J.; Gotlib, J.R.; Macey, T.A.; Oh, S.T.; Tyner, J.W.; Winton, E.F. Phase 2 Study of Ruxolitinib in Patients with Chronic Neutrophilic Leukemia or Atypical Chronic Myeloid Leukemia. Blood 2018, 132 (Suppl. 1), 350. [Google Scholar] [CrossRef]
  136. Guerra, V.A.; Kantarjian, H.M.; Borthakur, G.M.; Verstovsek, S.; Pike, A.; Ravandi, F.; Jabbour, E.; Cortes, J.E. A Phase I-II Study of Ruxolitinib (INCB18424) for Patients with Chronic Myeloid Leukemia with Minimal Residual Disease While on Therapy with Imatinib. Blood 2019, 134 (Suppl. 1), 5906. [Google Scholar] [CrossRef]
  137. Westerweel, P.E.; te Boekhorst, P.A.W.; Levin, M.-D.; Cornelissen, J.J. New Approaches and Treatment Combinations for the Management of Chronic Myeloid Leukemia. Front. Oncol. 2019, 9, 665. [Google Scholar] [CrossRef]
  138. Rousselot, P.; Prost, S.; Guilhot, J.; Roy, L.; Etienne, G.; Legros, L.; Charbonnier, A.; Coiteux, V.; Cony-Makhoul, P.; Huguet, F.; et al. Pioglitazone together with imatinib in chronic myeloid leukemia: A proof of concept study. Cancer 2017, 123, 1791–1799. [Google Scholar] [CrossRef]
  139. Glodkowska-Mrowka, E.; Manda-Handzlik, A.; Stelmaszczyk-Emmel, A.; SeferzDska, I.; Stoklosa, T.; Przybylski, J.; Mrowka, P. PPAU≥ ligands increase antileukemic activity of second- and third-generation tyrosine kinase inhibitors in chronic myeloid leukemia cells. Blood Cancer J. 2016, 6, e377. [Google Scholar] [CrossRef] [Green Version]
  140. Yousefi, B.; Samadi, N.; Baradaran, B.; Shafiei-Irannejad, V.; Zarghami, N. Peroxisome Proliferator-Activated Receptor Ligands and Their Role in Chronic Myeloid Leukemia: Therapeutic Strategies. Chem. Biol. Drug Des. 2016, 88, 17–25. [Google Scholar] [CrossRef]
  141. Prost, S.; Relouzat, F.; Spentchian, M.; Ouzegdouh, Y.; Saliba, J.; Massonnet, G.; Beressi, J.P.; Verhoeyen, E.; Raggueneau, V.; Maneglier, B.; et al. Erosion of the chronic myeloid leukaemia stem cell pool by PPARγ agonists. Nature 2015, 525, 380–383. [Google Scholar] [CrossRef] [PubMed]
  142. Eiring, A.M.; Page, B.D.G.; Kraft, I.L.; Mason, C.C.; Vellore, N.A.; Resetca, D.; Zabriskie, M.S.; Zhang, T.Y.; Khorashad, J.S.; Engar, A.J.; et al. Combined STAT3 and BCR-ABL1 inhibition induces synthetic lethality in therapy-resistant chronic myeloid leukemia. Leukemia 2017, 31, 1253–1254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Brachet-Botineau, M.; Polomski, M.; Neubauer, H.A.; Juen, L.; Hédou, D.; Viaud-Massuard, M.-C.; Prié, G.; Gouilleux, F. Pharmacological Inhibition of Oncogenic STAT3 and STAT5 Signaling in Hematopoietic Cancers. Cancers 2020, 12, 240. [Google Scholar] [CrossRef] [Green Version]
  144. Li, F.; He, B.; Ma, X.; Yu, S.; Bhave, R.R.; Lentz, S.R.; Tan, K.; Guzman, M.L.; Zhao, C.; Xue, H.H. Prostaglandin E1 and Its Analog Misoprostol Inhibit Human CML Stem Cell Self-Renewal via EP4 Receptor Activation and Repression of AP-1. Cell Stem Cell 2017, 21, 359–373.e5. [Google Scholar] [CrossRef] [Green Version]
  145. El Eit, R.; Itani, A.R.; Nassar, F.; Rasbieh, N.; Jabbour, M.; Santina, A.; Zaatari, G.; Mahon, F.-X.; Bazarbachi, A.; Nasr, R. Antitumor efficacy of arsenic/interferon in preclinical models of chronic myeloid leukemia resistant to tyrosine kinase inhibitors. Cancer 2019, 125, 2818–2828. [Google Scholar] [CrossRef]
  146. Guarnerio, J.; Mendez, L.M.; Asada, N.; Menon, A.V.; Fung, J.; Berry, K.; Frenette, P.S.; Ito, K.; Pandolfi, P.P. A non-cell-autonomous role for Pml in the maintenance of leukemia from the niche. Nat. Commun. 2018, 9, 66. [Google Scholar] [CrossRef] [Green Version]
  147. Zhou, L.; Hou, J.; Chan, G.; Sze, D. Arsenic Trioxide for Non Acute Promyelocytic Leukemia Hematological Malignancies: A New Frontier. J. Blood Disord. 2014, 1, 1018. [Google Scholar]
  148. Du, Y.; Wang, K.; Fang, H.; Li, J.; Xiao, D.; Zheng, P.; Chen, Y.; Fan, H.; Pan, X.; Zhao, C.; et al. Coordination of intrinsic, extrinsic, and endoplasmic reticulum-mediated apoptosis by imatinib mesylate combined with arsenic trioxide in chronic myeloid leukemia. Blood 2006, 107, 1582–1590. [Google Scholar] [CrossRef]
  149. Wang, W.; Lv, F.-f.; Du, Y.; Li, N.; Chen, Y.; Chen, L. The effect of nilotinib plus arsenic trioxide on the proliferation and differentiation of primary leukemic cells from patients with chronic myoloid leukemia in blast crisis. Cancer Cell Int. 2015, 15, 10. [Google Scholar] [CrossRef] [Green Version]
  150. El Eit, R.M.; Iskandarani, A.N.; Saliba, J.L.; Jabbour, M.N.; Mahfouz, R.A.; Bitar, N.M.; Ayoubi, H.R.; Zaatari, G.S.; Mahon, F.X.; De Thé, H.B.; et al. Effective targeting of chronic myeloid leukemia initiating activity with the combination of arsenic trioxide and interferon alpha. Int. J. Cancer 2014, 134, 988–996. [Google Scholar] [CrossRef]
  151. Heibl, S.; Buxhofer-Ausch, V.; Schmidt, S.; Webersinke, G.; Lion, T.; Piringer, G.; Kuehr, T.; Wolf, D.; Melchardt, T.; Greil, R.; et al. A phase 1 study to evaluate the feasibility and efficacy of the addition of ropeginterferon alpha-2b to imatinib treatment in patients with chronic phase chronic myeloid leukemia (CML) not achieving a deep molecular response (molecular remission 4.5)-AGMT_CML 1. Hematol. Oncol. 2020, 28, 792–798. [Google Scholar]
  152. Mitchell, R.; Copland, M. Defining niche interactions to target chronic myeloid leukemia stem cells. Haematologica 2020, 105, 2–4. [Google Scholar] [CrossRef] [PubMed]
  153. Murakami, J.L.; Xu, B.; Franco, C.B.; Hu, X.; Galli, S.J.; Weissman, I.L.; Chen, C.C. Evidence that β7 Integrin Regulates Hematopoietic Stem Cell Homing and Engraftment Through Interaction with MAdCAM-1. Stem Cells Dev. 2016, 25, 18–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Godavarthy, P.S.; Kumar, R.; Herkt, S.C.; Pereira, R.S.; Hayduk, N.; Weissenberger, E.S.; Aggoune, D.; Manavski, Y.; Lucas, T.; Pan, K.T.; et al. The vascular bone marrow niche influences outcome in chronic myeloid leukemia via the E-selectin—SCL/TAL1—CD44 axis. Haematologica 2020, 105, 136–147. [Google Scholar] [CrossRef] [Green Version]
  155. Windisch, R.; Pirschtat, N.; Kellner, C.; Chen-Wichmann, L.; Lausen, J.; Humpe, A.; Krause, D.; Wichmann, C. Oncogenic Deregulation of Cell Adhesion Molecules in Leukemia. Cancers 2019, 11, 311. [Google Scholar] [CrossRef] [Green Version]
  156. Klamer, S.; Voermans, C. The role of novel and known extracellular matrix and adhesion molecules in the homeostatic and regenerative bone marrow microenvironment. Cell Adhes. Migr. 2014, 8, 563–577. [Google Scholar] [CrossRef] [Green Version]
  157. Villatoro, A.; Konieczny, J.; Cuminetti, V.; Arranz, L. Leukemia Stem Cell Release From the Stem Cell Niche to Treat Acute Myeloid Leukemia. Front. Cell Dev. Biol. 2020, 8, 607. [Google Scholar] [CrossRef]
  158. Zhou, H.-S.; Carter, B.Z.; Andreeff, M. Bone marrow niche-mediated survival of leukemia stem cells in acute myeloid leukemia: Yin and Yang. Cancer Biol. Med. 2016, 13, 248–259. [Google Scholar] [CrossRef] [Green Version]
  159. Schepers, K.; Pietras, E.M.; Reynaud, D.; Flach, J.; Binnewies, M.; Garg, T.; Wagers, A.J.; Hsiao, E.C.; Passegué, E. Myeloproliferative neoplasia remodels the endosteal bone marrow niche into a self-reinforcing leukemic niche. Cell Stem Cell 2013, 13, 285–299. [Google Scholar] [CrossRef] [Green Version]
  160. Schepers, K.; Campbell, T.B.; Passegué, E. Normal and leukemic stem cell niches: Insights and therapeutic opportunities. Cell Stem Cell 2015, 16, 254–267. [Google Scholar] [CrossRef] [Green Version]
  161. Stramucci, L.; Perrotti, D. Twisting IL-1 signaling to kill CML stem cells. Blood 2016, 128, 2592–2593. [Google Scholar] [CrossRef] [PubMed]
  162. Krause, D.S.; Lazarides, K.; Lewis, J.B.; von Andrian, U.H.; Van Etten, R.A. Selectins and their ligands are required for homing and engraftment of BCR-ABL1+ leukemic stem cells in the bone marrow niche. Blood 2014, 123, 1361–1371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. DeAngelo, D.J.; Erba, H.P.; Jonas, B.A.; O’Dwyer, M.; Marlton, P.; Huls, G.A.; Liesveld, J.; Cooper, B.W.; Bhatnagar, B.; Armstrong, M.; et al. A phase III trial to evaluate the efficacy of uproleselan (GMI-1271) with chemotherapy in patients with relapsed/refractory acute myeloid leukemia. J. Clin. Oncol. 2019, 37 (Suppl. 15), TPS7066. [Google Scholar] [CrossRef]
  164. Agarwal, P.; Isringhausen, S.; Li, H.; Paterson, A.J.; He, J.; Gomariz, Á.; Nagasawa, T.; Nombela-Arrieta, C.; Bhatia, R. Mesenchymal Niche-Specific Expression of Cxcl12 Controls Quiescence of Treatment-Resistant Leukemia Stem Cells. Cell Stem Cell 2019, 24, 769–784.e6. [Google Scholar] [CrossRef]
  165. Weisberg, E.; Azab, A.K.; Manley, P.W.; Kung, A.L.; Christie, A.L.; Bronson, R.; Ghobrial, I.M.; Griffin, J.D. Inhibition of CXCR4 in CML cells disrupts their interaction with the bone marrow microenvironment and sensitizes them to nilotinib. Leukemia 2012, 26, 985–990. [Google Scholar] [CrossRef] [Green Version]
  166. Melo, R.; Ferro, K.; Saad, S. CXCR7 participates in CXCL12-mediated migration and homing of leukemic and normal hematopoietic cells. Stem Cell Res. Ther. 2018, 9, 34. [Google Scholar] [CrossRef]
  167. Kaur, H.; Bruno, J.; Kumar, A.; Sharma, T. Aptamers in the Therapeutics and Diagnostics Pipelines. Theranostics 2018, 8, 4016–4032. [Google Scholar] [CrossRef]
  168. Weisberg, E.L.; Sattler, M.; Azab, A.K.; Eulberg, D.; Kruschinski, A.; Manley, P.W.; Stone, R.; Griffin, J.D. Inhibition of SDF-1-induced migration of oncogene-driven myeloid leukemia by the L-RNA aptamer (Spiegelmer), NOX-A12, and potentiation of tyrosine kinase inhibition. Oncotarget 2017, 8, 109973–109984. [Google Scholar] [CrossRef] [Green Version]
  169. Fruehauf, S. Current Clinical Indications for Plerixafor. Transfus. Med. Hemother. 2013, 40, 246–250. [Google Scholar] [CrossRef] [Green Version]
  170. Agarwal, A.; Fleischman, A.G.; Petersen, C.L.; Mackenzie, R.; Luty, S.; Loriaux, M.; Druker, B.J.; Woltjer, R.L.; Deininger, M.W. Effects of plerixafor in combination with BCR-ABL kinase inhibition in a murine model of CML. Blood 2012, 120, 2658–2668. [Google Scholar] [CrossRef] [Green Version]
  171. Peng, G.; Liu, Y. Hypoxia-inducible factors in cancer stem cells and inflammation. Trends Pharmacol. Sci. 2015, 36, 374–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Chen, H.; Shen, Y.; Gong, F.; Jiang, Y.; Zhang, R. HIF-α Promotes Chronic Myelogenous Leukemia Cell Proliferation by Upregulating p21 Expression. Cell Biochem. Biophys. 2015, 72, 179–183. [Google Scholar] [CrossRef] [PubMed]
  173. Saito, S.; Lin, Y.-C.; Tsai, M.-H.; Lin, C.-S.; Murayama, Y.; Sato, R.; Yokoyama, K.K. Emerging roles of hypoxia-inducible factors and reactive oxygen species in cancer and pluripotent stem cells. Kaohsiung J. Med. Sci. 2015, 31, 279–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Cheloni, G.; Tanturli, M.; Tusa, I.; Ho DeSouza, N.; Shan, Y.; Gozzini, A.; Mazurier, F.; Rovida, E.; Li, S.; Dello Sbarba, P. Targeting chronic myeloid leukemia stem cells with the hypoxia-inducible factor inhibitor acriflavine. Blood 2017, 130, 655–665. [Google Scholar] [CrossRef] [Green Version]
  175. Su, W.; Meng, F.; Huang, L.; Zheng, M.; Liu, W.; Sun, H. Sonic hedgehog maintains survival and growth of chronic myeloid leukemia progenitor cells through β-catenin signaling. Exp. Hematol. 2012, 40, 418–427. [Google Scholar] [CrossRef]
  176. Shah, N.P.; Cortes, J.E.; Martinelli, G.; Smith, B.D.; Clarke, E.; Copland, M.; Strauss, L.; Talpaz, M. Dasatinib Plus Smoothened (SMO) Inhibitor BMS-833923 in Chronic Myeloid Leukemia (CML) with Resistance or Suboptimal Response to a Prior Tyrosine Kinase Inhibitor (TKI): Phase I Study CA180323. Blood 2014, 124, 4539. [Google Scholar] [CrossRef]
  177. Irvine, D.A.; Zhang, B.; Kinstrie, R.; Tarafdar, A.; Morrison, H.; Campbell, V.L.; Moka, H.A.; Ho, Y.; Nixon, C.; Manley, P.W.; et al. Deregulated hedgehog pathway signaling is inhibited by the smoothened antagonist LDE225 (Sonidegib) in chronic phase chronic myeloid leukaemia. Sci. Rep. 2016, 6, 25476. [Google Scholar] [CrossRef] [Green Version]
  178. Agarwal, P.; Zhang, B.; Ho, Y.; Cook, A.; Li, L.; Mikhail, F.M.; Wang, Y.; McLaughlin, M.E.; Bhatia, R. Enhanced targeting of CML stem and progenitor cells by inhibition of porcupine acyltransferase in combination with TKI. Blood 2017, 129, 1008–1020. [Google Scholar] [CrossRef] [Green Version]
  179. Agarwal, P.; Zhang, B.; Ho, Y.; Cook, A.; Li, L.; Wang, Y.; Mclaughlin, M.E.; Bhatia, R. Inhibition of CML Stem Cell Renewal By the Porcupine Inhibitor WNT974. Blood 2015, 126, 54. [Google Scholar] [CrossRef]
  180. Grassi, S.; Palumbo, S.; Mariotti, V.; Liberati, D.; Guerrini, F.; Ciabatti, E.; Salehzadeh, S.; Baratè, C.; Balducci, S.; Ricci, F.; et al. The WNT Pathway Is Relevant for the BCR-ABL1-Independent Resistance in Chronic Myeloid Leukemia. Front. Oncol. 2019, 9, 532. [Google Scholar] [CrossRef] [Green Version]
  181. Riether, C.; Schürch, C.M.; Flury, C.; Hinterbrandner, M.; Drück, L.; Huguenin, A.L.; Baerlocher, G.M.; Radpour, R.; Ochsenbein, A.F. Tyrosine kinase inhibitor-induced CD70 expression mediates drug resistance in leukemia stem cells by activating Wnt signaling. Sci. Transl. Med. 2015, 7, 298ra119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Pippa, R.; Odero, M.D. The Role of MYC and PP2A in the Initiation and Progression of Myeloid Leukemias. Cells 2020, 9, 544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Lucas, C.M.; Harris, R.J.; Giannoudis, A.; Clark, R.E. c-Myc inhibition decreases CIP2A and reduces BCR-ABL1 tyrosine kinase activity in chronic myeloid leukemia. Haematologica 2015, 100, e179–e182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Kiyota, M.; Kuroda, J.; Yamamoto-Sugitani, M.; Shimura, Y.; Nakayama, R.; Nagoshi, H.; Mizutani, S.; Chinen, Y.; Sasaki, N.; Sakamoto, N.; et al. FTY720 induces apoptosis of chronic myelogenous leukemia cells via dual activation of BIM and BID and overcomes various types of resistance to tyrosine kinase inhibitors. Apoptosis 2013, 18, 1437–1446. [Google Scholar] [CrossRef]
  185. Sharma, N.; Magistroni, V.; Piazza, R.; Citterio, S.; Mezzatesta, C.; Khandelwal, P.; Pirola, A.; Gambacorti-Passerini, C. BCR/ABL1 and BCR are under the transcriptional control of the MYC oncogene. Mol. Cancer 2015, 14, 132. [Google Scholar] [CrossRef] [Green Version]
  186. Agarwal, A.; Mackenzie, R.; Pippa, R.; Eide, C.; Oddo, J.; Tyner, J.; Sears, R.; Vitek, M.; Christensen, D.; Druker, B. Antagonism of SET Using OP449 Enhances the Efficacy of Tyrosine Kinase Inhibitors and Overcomes Drug Resistance in Myeloid Leukemia. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2014, 20, 2092–2103. [Google Scholar] [CrossRef] [Green Version]
  187. Albajar, M.; Gomez-Casares, M.T.; Llorca, J.; Mauleon, I.; Vaque, J.P.; Acosta, J.C.; Bermudez, A.; Donato, N.; Delgado, M.D.; Leon, J. MYC in Chronic Myeloid Leukemia: Induction of Aberrant DNA Synthesis and Association with Poor Response to Imatinib. Mol. Cancer Res. 2011, 9, 564–576. [Google Scholar] [CrossRef] [Green Version]
  188. Neviani, P.; Harb, J.G.; Oaks, J.J.; Santhanam, R.; Walker, C.J.; Ellis, J.J.; Ferenchak, G.; Dorrance, A.M.; Paisie, C.A.; Eiring, A.M.; et al. PP2A-activating drugs selectively eradicate TKI-resistant chronic myeloid leukemic stem cells. J. Clin. Invest. 2013, 123, 4144–4157. [Google Scholar] [CrossRef]
  189. Keramatinia, A.; Ahadi, A.; Akbari, M.E.; Mohseny, M.; Mosavi Jarahi, A.; Bahadori-Monfared, A.; Hashemi, M.; Moradi, A.; Mehrvar, N.; Kazemi, E.; et al. The roles of DNA epigenetics and clinical significance in Chronic Myeloid Leukemia: A review. Cell Mol. Biol. 2018, 64, 58–63. [Google Scholar] [CrossRef]
  190. Goldman, J.M.; Gordon, M.; Bazeos, A.; Marin, D. Biology of CML stem cells: The basis for clinical heterogeneity? Leuk. Suppl. 2012, 1, S43–S45. [Google Scholar] [CrossRef]
  191. Biray Avci, C.; Goker Bagca, B.; Tetik Vardarli, A.; Saydam, G.; Gunduz, C. Epigenetic modifications in chronic myeloid leukemia cells through ruxolitinib treatment. J. Cell Biochem. 2019, 120, 4555–4563. [Google Scholar] [CrossRef] [PubMed]
  192. Olszewski, A.J.; Chorzalska, A.D.; Kim, A.S.; Quesenberry, P.J.; Lopresti, M.L.; Fenton, M.A.; Reagan, J.L.; Butera, J.N.; Sahin, I.; Hamel, C.; et al. Clonal haematopoiesis of indeterminate potential among cancer survivors exposed to myelotoxic chemotherapy. Br. J. Haematol. 2019, 186, e31–e35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Steensma, D.P. Myelodysplastic syndromes current treatment algorithm 2018. Blood Cancer J. 2018, 8, 47. [Google Scholar] [CrossRef] [PubMed]
  194. Busque, L.; Buscarlet, M.; Mollica, L.; Levine, R.L. Concise Review: Age-Related Clonal Hematopoiesis: Stem Cells Tempting the Devil. Stem Cells 2018, 36, 1287–1294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Valent, P.; Kern, W.; Hoermann, G.; Milosevic Feenstra, J.; Sotlar, K.; Pfeilstöcker, M.; Germing, U.; Sperr, W.; Reiter, A.; Wolf, D.; et al. Clonal Hematopoiesis with Oncogenic Potential (CHOP): Separation from CHIP and Roads to AML. Int. J. Mol. Sci. 2019, 20, 789. [Google Scholar] [CrossRef] [Green Version]
  196. Park, S.J.; Bejar, R. Clonal Hematopoiesis in Aging. Curr. Stem Cell Rep. 2018, 4, 209–219. [Google Scholar] [CrossRef]
  197. Abelson, S.; Wang, J.C.Y. Age-related clonal hematopoiesis: Implications for hematopoietic stem cell transplantation. Curr. Opin. Hematol. 2018, 25, 441–445. [Google Scholar] [CrossRef]
  198. Jaiswal, S.; Ebert, B.L. Clonal hematopoiesis in human aging and disease. Science 2019, 366, eaan4673. [Google Scholar] [CrossRef]
  199. Genovese, G.; Kähler, A.K.; Handsaker, R.E.; Lindberg, J.; Rose, S.A.; Bakhoum, S.F.; Chambert, K.; Mick, E.; Neale, B.M.; Fromer, M.; et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 2014, 371, 2477–2487. [Google Scholar] [CrossRef] [Green Version]
  200. Doroshow, D.B.; Eder, J.P.; LoRusso, P.M. BET inhibitors: A novel epigenetic approach. Ann. Oncol. 2017, 28, 1776–1787. [Google Scholar] [CrossRef]
  201. White, M.E.; Fenger, J.M.; Carson, W.E. 3rd, Emerging roles of and therapeutic strategies targeting BRD4 in cancer. Cell Immunol. 2019, 337, 48–53. [Google Scholar] [CrossRef] [PubMed]
  202. Peter, B.; Eisenwort, G.; Keller, A.; Bauer, K.; Berger, D.; Sadovnik, I.; Stefanzl, G.; Hoermann, G.; Wolf, D.; Racil, Z.; et al. BRD4 Degradation Is a Potent Approach to Block MYC Expression and to Overcome Multiple Forms of Stem Cell Resistance in Ph+ CML. Blood 2018, 132 (Suppl. 1), 1722. [Google Scholar] [CrossRef]
  203. White, E. The role for autophagy in cancer. J. Clin. Investig. 2015, 125, 42–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Zhu, H.; Bengsch, F.; Svoronos, N.; Rutkowski, M.R.; Bitler, B.G.; Allegrezza, M.J.; Yokoyama, Y.; Kossenkov, A.V.; Bradner, J.E.; Conejo-Garcia, J.R.; et al. BET Bromodomain Inhibition Promotes Anti-tumor Immunity by Suppressing PD-L1 Expression. Cell Rep. 2016, 16, 2829–2837. [Google Scholar] [CrossRef] [Green Version]
  205. Hogg, S.J.; Vervoort, S.J.; Deswal, S.; Ott, C.J.; Li, J.; Cluse, L.A.; Beavis, P.A.; Darcy, P.K.; Martin, B.P.; Spencer, A.; et al. BET-Bromodomain Inhibitors Engage the Host Immune System and Regulate Expression of the Immune Checkpoint Ligand PD-L1. Cell Rep. 2017, 18, 2162–2174. [Google Scholar] [CrossRef] [Green Version]
  206. Jones, B.A.; Varambally, S.; Arend, R.C. Histone Methyltransferase EZH2: A Therapeutic Target for Ovarian Cancer. Mol. Cancer Ther. 2018, 17, 591–602. [Google Scholar] [CrossRef] [Green Version]
  207. Ueda, K.; Yoshimi, A.; Kagoya, Y.; Nishikawa, S.; Marquez, V.E.; Nakagawa, M.; Kurokawa, M. Inhibition of histone methyltransferase EZH2 depletes leukemia stem cell of mixed lineage leukemia fusion leukemia through upregulation of p16. Cancer Sci. 2014, 105, 512–519. [Google Scholar] [CrossRef]
  208. Rinke, J.; Chase, A.; Cross, N.C.P.; Hochhaus, A.; Ernst, T. EZH2 in Myeloid Malignancies. Cells 2020, 9, 1639. [Google Scholar] [CrossRef]
  209. Wen, Y.; Cai, J.; Hou, Y.; Huang, Z.; Wang, Z. Role of EZH2 in cancer stem cells: From biological insight to a therapeutic target. Oncotarget 2017, 8, 37974–37990. [Google Scholar] [CrossRef] [Green Version]
  210. Xie, H.; Peng, C.; Huang, J.; Li, B.E.; Kim, W.; Smith, E.C.; Fujiwara, Y.; Qi, J.; Cheloni, G.; Das, P.P.; et al. Chronic Myelogenous Leukemia- Initiating Cells Require Polycomb Group Protein EZH2. Cancer Discov. 2016, 6, 1237–1247. [Google Scholar] [CrossRef] [Green Version]
  211. Bavaro, L.; Martelli, M.; Cavo, M.; Soverini, S. Mechanisms of Disease Progression and Resistance to Tyrosine Kinase Inhibitor Therapy in Chronic Myeloid Leukemia: An Update. Int. J. Mol. Sci. 2019, 20, 6141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Matsuda, Y.; Yamauchi, T.; Hosono, N.; Uzui, K.; Negoro, E.; Morinaga, K.; Nishi, R.; Yoshida, A.; Kimura, S.; Maekawa, T.; et al. Combination of panobinostat with ponatinib synergistically overcomes imatinib-resistant CML cells. Cancer Sci. 2016, 107, 1029–1038. [Google Scholar] [CrossRef] [PubMed]
  213. Zhang, B.; Strauss, A.C.; Chu, S.; Li, M.; Ho, Y.; Shiang, K.D.; Snyder, D.S.; Huettner, C.S.; Shultz, L.; Holyoake, T.; et al. Effective targeting of quiescent chronic myelogenous leukemia stem cells by histone deacetylase inhibitors in combination with imatinib mesylate. Cancer Cell 2010, 17, 427–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Lernoux, M.; Schnekenburger, M.; Losson, H.; Vermeulen, K.; Hahn, H.; Gérard, D.; Lee, J.-Y.; Mazumder, A.; Ahamed, M.; Christov, C.; et al. Novel HDAC inhibitor MAKV-8 and imatinib synergistically kill chronic myeloid leukemia cells via inhibition of BCR-ABL/MYC-signaling: Effect on imatinib resistance and stem cells. Clin. Epigenet. 2020, 12, 69. [Google Scholar] [CrossRef]
  215. He, B.; Wang, Q.; Xu, N.; Lu, Z.; Pan, C.; Sun, J.; Zhou, H.; Xiaoli, L. Chidamide, a Novel Histone Deacetylase Inhibitor, Combined with Imatinib Induces Synthetic Lethality in Drug-Resistant Chronic Myeloid Leukemia. Blood 2017, 130 (Suppl. 1), 4990. [Google Scholar]
  216. Zhu, F.; Rui, L. PRMT5 in gene regulation and hematologic malignancies. Genes Dis. 2019, 6, 247–257. [Google Scholar] [CrossRef]
  217. Xiao, W.; Chen, X.; Liu, L.; Shu, Y.; Zhang, M.; Zhong, Y. Role of protein arginine methyltransferase 5 in human cancers. Biomed. Pharmacother. 2019, 114, 108790. [Google Scholar] [CrossRef]
  218. Jin, Y.; Zhou, J.; Xu, F.; Jin, B.; Cui, L.; Wang, Y.; Du, X.; Li, J.; Li, P.; Ren, R.; et al. Targeting methyltransferase PRMT5 eliminates leukemia stem cells in chronic myelogenous leukemia. J. Clin. Invest. 2016, 126, 3961–3980. [Google Scholar] [CrossRef] [Green Version]
  219. Abraham, S.A.; Hopcroft, L.E.; Carrick, E.; Drotar, M.E.; Dunn, K.; Williamson, A.J.; Korfi, K.; Baquero, P.; Park, L.E.; Scott, M.T.; et al. Dual targeting of p53 and c-MYC selectively eliminates leukaemic stem cells. Nature 2016, 534, 341–346. [Google Scholar] [CrossRef]
  220. Crivellaro, S.; Panuzzo, C.; Carrà, G.; Volpengo, A.; Crasto, F.; Gottardi, E.; Familiari, U.; Papotti, M.; Torti, D.; Piazza, R.; et al. Non genomic loss of function of tumor suppressors in CML: BCR-ABL promotes IκBα mediated p53 nuclear exclusion. Oncotarget 2015, 6, 25217–25225. [Google Scholar] [CrossRef]
  221. Li, L.; Wang, L.; Li, L.; Wang, Z.; Ho, Y.; McDonald, T.; Holyoake, T.L.; Chen, W.; Bhatia, R. Activation of p53 by SIRT1 inhibition enhances elimination of CML leukemia stem cells in combination with imatinib. Cancer Cell 2012, 21, 266–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Mobaraki, R.N.; Karimi, M.; Alikarami, F.; Farhadi, E.; Amini, A.; Bashash, D.; Paridar, M.; Kokhaei, P.; Rezvani, M.R.; Kazemi, A.; et al. RITA induces apoptosis in p53-null K562 leukemia cells by inhibiting STAT5, Akt, and NF-κB signaling pathways. Anticancer Drugs 2018, 29, 847–853. [Google Scholar] [CrossRef] [PubMed]
  223. Issaeva, N.; Bozko, P.; Enge, M.; Protopopova, M.; Verhoef, L.G.; Masucci, M.; Pramanik, A.; Selivanova, G. Small molecule RITA binds to p53, blocks p53-HDM-2 interaction and activates p53 function in tumors. Nat. Med. 2004, 10, 1321–1328. [Google Scholar] [CrossRef] [PubMed]
  224. Ma, T.; Yamada, S.; Ichwan, S.J.; Iseki, S.; Ohtani, K.; Otsu, M.; Ikeda, M.A. Inability of p53-reactivating compounds Nutlin-3 and RITA to overcome p53 resistance in tumor cells deficient in p53Ser46 phosphorylation. Biochem. Biophys. Res. Commun. 2012, 417, 931–937. [Google Scholar] [CrossRef]
  225. Hientz, K.; Mohr, A.; Bhakta-Guha, D.; Efferth, T. The role of p53 in cancer drug resistance and targeted chemotherapy. Oncotarget 2017, 8, 8921–8946. [Google Scholar] [CrossRef] [Green Version]
  226. Abraham, A.; Qiu, S.; Chacko, B.K.; Li, H.; Paterson, A.J.; He, J.; Agarwal, P.; Shah, M.; Welner, R.S.; Darley-Usmar, V.M.; et al. SIRT1 Mediates Enhanced Mitochondrial Oxidative Phosphorylation in Chronic Myelogenous Leukemia Stem Cells. Blood 2018, 132 (Suppl. 1), 932. [Google Scholar] [CrossRef]
  227. Peterson, L.F.; Lo, M.-C.; Liu, Y.; Giannola, D.; Mitrikeska, E.; Donato, N.J.; Johnson, C.N.; Wang, S.; Mercer, J.; Talpaz, M. Induction of p53 suppresses chronic myeloid leukemia. Leuk. Lymphoma 2017, 58, 2165–2175. [Google Scholar] [CrossRef]
  228. Sanz, G.; Singh, M.; Peuget, S.; Selivanova, G. Inhibition of p53 inhibitors: Progress, challenges and perspectives. J. Mol. Cell Biol. 2019, 11, 586–599. [Google Scholar] [CrossRef] [Green Version]
  229. Grant, S. Recruiting TP53 to target chronic myeloid leukemia stem cells. Haematologica 2020, 105, 1172–1174. [Google Scholar] [CrossRef]
  230. Yun, C.W.; Lee, S.H. The Roles of Autophagy in Cancer. Int. J. Mol. Sci. 2018, 19, 3466. [Google Scholar] [CrossRef] [Green Version]
  231. Huang, X.; Li, Y.; Shou, L.; Li, L.; Chen, Z.; Ye, X.; Qian, W. The molecular mechanisms underlying BCR/ABL degradation in chronic myeloid leukemia cells promoted by Beclin1-mediated autophagy. Cancer Manag. Res. 2019, 11, 5197–5208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Lu, Z.; Xu, N.; He, B.; Pan, C.; Lan, Y.; Zhou, H.; Liu, X. Inhibition of autophagy enhances the selective anti-cancer activity of tigecycline to overcome drug resistance in the treatment of chronic myeloid leukemia. J. Exp. Clin. Cancer Res. 2017, 36, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Kuntz, E.M.; Baquero, P.; Michie, A.M.; Dunn, K.; Tardito, S.; Holyoake, T.L.; Helgason, G.V.; Gottlieb, E. Targeting mitochondrial oxidative phosphorylation eradicates therapy-resistant chronic myeloid leukemia stem cells. Nat. Med. 2017, 23, 1234–1240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Greer, N.D. Tigecycline (Tygacil): The First in the Glycylcycline Class of Antibiotics. Bayl. Univ. Med. Cent. Proc. 2006, 19, 155–161. [Google Scholar] [CrossRef]
  235. Santana-Codina, N.; Mancias, J.D.; Kimmelman, A.C. The Role of Autophagy in Cancer. Annu. Rev. Cancer Biol. 2017, 1, 19–39. [Google Scholar] [CrossRef]
  236. Mauthe, M.; Orhon, I.; Rocchi, C.; Zhou, X.; Luhr, M.; Hijlkema, K.-J.; Coppes, R.P.; Engedal, N.; Mari, M.; Reggiori, F. Chloroquine inhibits autophagic flux by decreasing autophagosome-lysosome fusion. Autophagy 2018, 14, 1435–1455. [Google Scholar] [CrossRef]
  237. Calabretta, B.; Salomoni, P. Inhibition of autophagy: A new strategy to enhance sensitivity of chronic myeloid leukemia stem cells to tyrosine kinase inhibitors. Leuk. Lymphoma 2011, 52 (Suppl. 1), 54–59. [Google Scholar] [CrossRef]
  238. Horne, G.A.; Stobo, J.; Kelly, C.; Mukhopadhyay, A.; Latif, A.L.; Dixon-Hughes, J.; McMahon, L.; Cony-Makhoul, P.; Byrne, J.; Smith, G.; et al. A randomised phase II trial of hydroxychloroquine and imatinib versus imatinib alone for patients with chronic myeloid leukaemia in major cytogenetic response with residual disease. Leukemia 2020, 34, 1775–1786. [Google Scholar] [CrossRef]
  239. Riether, C.; Gschwend, T.; Huguenin, A.L.; Schürch, C.M.; Ochsenbein, A.F. Blocking programmed cell death 1 in combination with adoptive cytotoxic T-cell transfer eradicates chronic myelogenous leukemia stem cells. Leukemia 2015, 29, 1781–1785. [Google Scholar] [CrossRef] [Green Version]
  240. Rousselot, P.; Renard, P.; de Buyer, A.; Finet, A.; Spentchian, M.; Saiag, P. Nivolumab to control molecular response in chronic myeloid leukemia. Leuk. Res. 2018, 72, 5–6. [Google Scholar] [CrossRef]
  241. Collins, J.M.; Gulley, J.L. Product review: Avelumab, an anti-PD-L1 antibody. Hum. Vaccin. Immunother. 2019, 15, 891–908. [Google Scholar] [CrossRef] [PubMed]
  242. Mosoyan, G.; Kraus, T.; Ye, F.; Eng, K.; Crispino, J.D.; Hoffman, R.; Iancu-Rubin, C. Imetelstat, a telomerase inhibitor, differentially affects normal and malignant megakaryopoiesis. Leukemia 2017, 31, 2458–2467. [Google Scholar] [CrossRef] [PubMed]
  243. Wang, X.; Hu, C.S.; Petersen, B.; Qiu, J.; Ye, F.; Houldsworth, J.; Eng, K.; Huang, F.; Hoffman, R. Imetelstat, a telomerase inhibitor, is capable of depleting myelofibrosis stem and progenitor cells. Blood Adv. 2018, 2, 2378–2388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Tefferi, A.; Lasho, T.L.; Begna, K.H.; Patnaik, M.M.; Zblewski, D.L.; Finke, C.M.; Laborde, R.R.; Wassie, E.; Schimek, L.; Hanson, C.A.; et al. A Pilot Study of the Telomerase Inhibitor Imetelstat for Myelofibrosis. N. Engl. J. Med. 2015, 373, 908–919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Mascarenhas, J.; Komrokji, R.S.; Cavo, M.; Martino, B.; Niederwieser, D.; Reiter, A.; Scott, B.L.; Baer, M.R.; Hoffman, R.; Odenike, O.; et al. Imetelstat Is Effective Treatment for Patients with Intermediate-2 or High-Risk Myelofibrosis Who Have Relapsed on or Are Refractory to Janus Kinase Inhibitor Therapy: Results of a Phase 2 Randomized Study of Two Dose Levels. Blood 2018, 132 (Suppl. 1), 685. [Google Scholar] [CrossRef]
  246. Baerlocher, G.M.; Oppliger Leibundgut, E.; Ottmann, O.G.; Spitzer, G.; Odenike, O.; Mcdevitt, M.A.; Röth, A.; Daskalakis, M.; Burington, B.; Stuart, M.; et al. Telomerase Inhibitor Imetelstat in Patients with Essential Thrombocythemia. N. Engl. J. Med. 2015, 373, 920–928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Mascarenhas, J.; Lu, M.; Kosiorek, H.; Virtgaym, E.; Xia, L.; Sandy, L.; Mesa, R.; Petersen, B.; Farnoud, N.; Najfeld, V.; et al. Oral idasanutlin in patients with polycythemia vera. Blood 2019, 134, 525–533. [Google Scholar] [CrossRef] [Green Version]
  248. Hobbs, G.S.; Hanasoge Somasundara, A.V.; Kleppe, M.; Litvin, R.; Arcila, M.; Ahn, J.; Mckenney, A.S.; Knapp, K.; Ptashkin, R.; Weinstein, H.; et al. Hsp90 inhibition disrupts JAK-STAT signaling and leads to reductions in splenomegaly in patients with myeloproliferative neoplasms. Haematologica 2018, 103, e5–e9. [Google Scholar] [CrossRef] [Green Version]
  249. Pemmaraju, N.; Gupta, V.; Ali, H.; Yacoub, A.; Wang, E.S.; Lee, S.; Schiller, G.J.; Sardone, M.; Wysowskyj, H.; Chen, J.; et al. Results from a Phase 1/2 Clinical Trial of Tagraxofusp (SL-401) in Patients with Intermediate, or High Risk, Relapsed/Refractory Myelofibrosis. Blood 2019, 134 (Suppl. 1), 558. [Google Scholar] [CrossRef]
  250. Pettit, K.; Gerds, A.T.; Yacoub, A.; Watts, J.M.; Tartaczuch, M.; Bradley, T.J.; Shortt, J.; Stevenson, W.S.; Curtin, N.J.; Rossetti, J.M.; et al. A Phase 2a Study of the LSD1 Inhibitor Img-7289 (bomedemstat) for the Treatment of Myelofibrosis. Blood 2019, 134 (Suppl. 1), 556. [Google Scholar] [CrossRef]
  251. Hergert, J. First-in-Class LSD1 Inhibitor Elicits Clinical Activity in Advanced Myelofibrosis. Available online: onclive.com/view/first-in-class-lsd1-inhibitor-elicits-clinical-activity-in-advanced-myelofibrosis (accessed on 30 November 2019).
  252. Hugh Rienhoff, M. IMG-7289 in Patients With Essential Thrombocythemia, 2020-02-05 ed.; U.S. National Library of Medicine: Bethesda, MD, USA, 2020.
  253. Rambaldi, A.; Iurlo, A.; Vannucchi, A.M.; Noble, R.; Von Bubnoff, N.; Guarini, A.; Martino, B.; Pezzutto, A.; Carli, G.; De Muro, M.; et al. Safety and efficacy of the maximum tolerated dose of givinostat in polycythemia vera: A two-part Phase Ib/II study. Leukemia 2020, 34, 2234–2237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Deangelo, D.J.; Mesa, R.A.; Fiskus, W.; Tefferi, A.; Paley, C.; Wadleigh, M.; Ritchie, E.K.; Snyder, D.S.; Begna, K.; Ganguly, S.; et al. Phase II trial of panobinostat, an oral pan-deacetylase inhibitor in patients with primary myelofibrosis, post-essential thrombocythaemia, and post-polycythaemia vera myelofibrosis. Br. J. Haematol. 2013, 162, 326–335. [Google Scholar] [CrossRef] [PubMed]
  255. A Study of PRT543 in Participants With Advanced Solid Tumors and Hematologic Malignancies, 2019-03-22 ed.; U.S. National Library of Medicine: Bethesda, MD, USA, 2019.
  256. Drexler, B.; Passweg, J.R.; Tzankov, A.; Bigler, M.; Theocharides, A.P.; Cantoni, N.; Keller, P.; Stussi, G.; Ruefer, A.; Benz, R.; et al. The sympathomimetic agonist mirabegron did not lower JAK2-V617F allele burden, but restored nestin-positive cells and reduced reticulin fibrosis in patients with myeloproliferative neoplasms: Results of phase II study SAKK 33/14. Haematologica 2019, 104, 710–716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Gerds, A.T.; Vannucchi, A.M.; Passamonti, F.; Kremyanskaya, M.; Gotlib, J.R.; Palmer, J.M.; McCaul, K.; Ribrag, V.; Mead, A.J.; Harrison, C.N.; et al. A Phase 2 Study of Luspatercept in Patients with Myelofibrosis-Associated Anemia. Blood 2019, 134 (Suppl. 1), 557. [Google Scholar] [CrossRef]
  258. Bose, P.; Pemmaraju, N.; Masarova, L.; Bledsoe, S.D.; Daver, N.; Jabbour, E.; Kadia, T.M.; Estrov, Z.E.; Kornblau, S.M.; Andreeff, M.; et al. Sotatercept (ACE-011) for Anemia of Myelofibrosis: A Phase 2 Study. Blood 2020, 136 (Suppl. 1), 10–11. [Google Scholar] [CrossRef]
  259. Piszczatowski, R.T.; Steidl, U. Aurora Kinase A Inhibition: A Mega-Hit for Myelofibrosis Therapy? Clin. Cancer Res. 2019, 25, 4868–4870. [Google Scholar] [CrossRef]
  260. Gangat, N.; Marinaccio, C.; Swords, R.; Watts, J.M.; Gurbuxani, S.; Rademaker, A.; Fought, A.J.; Frankfurt, O.; Altman, J.K.; Wen, Q.J.; et al. Aurora Kinase A Inhibition Provides Clinical Benefit, Normalizes Megakaryocytes, and Reduces Bone Marrow Fibrosis in Patients with Myelofibrosis: A Phase I Trial. Clin. Cancer Res. 2019, 25, 4898–4906. [Google Scholar] [CrossRef] [Green Version]
  261. Durrant, S.T.; Nagler, A.; Guglielmelli, P.; Lavie, D.; Le Coutre, P.; Gisslinger, H.; Chuah, C.; Maffioli, M.; Bharathy, S.; Dong, T.; et al. Results from HARMONY: An open-label, multicenter, 2-arm, phase 1b, dose-finding study assessing the safety and efficacy of the oral combination of ruxolitinib and buparlisib in patients with myelofibrosis. Haematologica 2019, 104, e551–e554. [Google Scholar] [CrossRef] [Green Version]
  262. Saenz, D.T.; Fiskus, W.; Manshouri, T.; Rajapakshe, K.; Krieger, S.; Sun, B.; Mill, C.P.; Dinardo, C.; Pemmaraju, N.; Kadia, T.; et al. BET protein bromodomain inhibitor-based combinations are highly active against post-myeloproliferative neoplasm secondary AML cells. Leukemia 2017, 31, 678–687. [Google Scholar] [CrossRef] [Green Version]
  263. Pérez-Salvia, M.; Esteller, M. Bromodomain inhibitors and cancer therapy: From structures to applications. Epigenetics 2017, 12, 323–339. [Google Scholar] [CrossRef]
  264. Jiang, Q.; Jamieson, C. BET’ing on Dual JAK/BET Inhibition as a Therapeutic Strategy for Myeloproliferative Neoplasms. Cancer Cell 2018, 33, 3–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Kleppe, M.; Koche, R.; Zou, L.; Van Galen, P.; Hill, C.E.; Dong, L.; De Groote, S.; Papalexi, E.; Hanasoge Somasundara, A.V.; Cordner, K.; et al. Dual Targeting of Oncogenic Activation and Inflammatory Signaling Increases Therapeutic Efficacy in Myeloproliferative Neoplasms. Cancer Cell 2018, 33, 29–43.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Sashida, G.; Wang, C.; Tomioka, T.; Oshima, M.; Aoyama, K.; Kanai, A.; Mochizuki-Kashio, M.; Harada, H.; Shimoda, K.; Iwama, A. The loss of Ezh2 drives the pathogenesis of myelofibrosis and sensitizes tumor-initiating cells to bromodomain inhibition. J. Exp. Med. 2016, 213, 1459–1477. [Google Scholar] [CrossRef] [PubMed]
  267. Lu, M.; Xia, L.; Li, Y.; Wang, X.; Hoffman, R. The orally bioavailable MDM2 antagonist RG7112 and pegylated interferon α 2a target JAK2V617F-positive progenitor and stem cells. Blood 2014, 124, 771–779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  268. Eran, Z.; Zingariello, M.; Bochicchio, M.T.; Bardelli, C.; Migliaccio, A.R. Novel strategies for the treatment of myelofibrosis driven by recent advances in understanding the role of the microenvironment in its etiology. F1000Research 2019, 8, 1662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  269. Marcellino, B.K.; Hoffman, R.; Tripodi, J.; Lu, M.; Kosiorek, H.; Mascarenhas, J.; Rampal, R.K.; Dueck, A.; Najfeld, V. Advanced forms of MPNs are accompanied by chromosomal abnormalities that lead to dysregulation of TP53. Blood Adv. 2018, 2, 3581–3589. [Google Scholar] [CrossRef] [Green Version]
  270. Garcia-Delgado, R.; McLornan, D.P.; Rejtő, L.; Jourdan, E.; Al-Ali, H.K.; Pluta, A.; Hus, M.; Ewing, J.; Khan, I.; Hebart, H.; et al. An Open-Label, Phase 2 Study of KRT-232, a First-in-Class, Oral Small Molecule Inhibitor of MDM2, for the Treatment of Patients with Myelofibrosis (MF) Who Have Previously Received Treatment with a JAK Inhibitor. Blood 2019, 134 (Suppl. 1), 2945. [Google Scholar] [CrossRef]
  271. Gotlib, J.; Gabrail, N.; O’Connell, C.L.; Garcia-Delgado, R.; Sbardellati, T.; Rothbaum, W.M.; McGreivy, J.; Harrison, C.N.; Kiladjian, J.-J. A Randomized, Open-Label, Multicenter, Phase 2 Study to Evaluate the Efficacy, Safety, and Pharmacokinetics of KRT-232 Compared with Ruxolitinib in Patients with Phlebotomy-Dependent Polycythemia Vera. Blood 2019, 134 (Suppl. 1), 4168. [Google Scholar] [CrossRef]
  272. Kuykendall, A.T.; Horvat, N.P.; Pandey, G.; Komrokji, R.; Reuther, G.W. Finding a Jill for JAK: Assessing Past, Present, and Future JAK Inhibitor Combination Approaches in Myelofibrosis. Cancers 2020, 12, 2278. [Google Scholar] [CrossRef]
  273. De Almeida, S.; Regimbeau, M.; Jego, G.; Garrido, C.; Girodon, F.; Hermetet, F. Heat Shock Proteins and PD-1/PD-L1 as Potential Therapeutic Targets in Myeloproliferative Neoplasms. Cancers 2020, 12, 2592. [Google Scholar] [CrossRef]
  274. Gilani, J.A.; Ashfaq, M.A.; Mansoor, A.-E.-R.; Abdul Jabbar, A.; Siddiqui, T.; Khan, M. Overview of the Mutational Landscape in Primary Myelofibrosis and Advances in Novel Therapeutics. Asian Pac. J. Cancer Prev. 2019, 20, 1691–1699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Fiskus, W.; Verstovsek, S.; Manshouri, T.; Rao, R.; Balusu, R.; Venkannagari, S.; Rao, N.N.; Ha, K.; Smith, J.E.; Hembruff, S.L.; et al. Heat Shock Protein 90 Inhibitor Is Synergistic with JAK2 Inhibitor and Overcomes Resistance to JAK2-TKI in Human Myeloproliferative Neoplasm Cells. Clin. Cancer Res. 2011, 17, 7347–7358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  276. Marubayashi, S.; Koppikar, P.; Taldone, T.; Abdel-Wahab, O.; West, N.; Bhagwat, N.; Caldas-Lopes, E.; Ross, K.N.; Gönen, M.; Gozman, A.; et al. HSP90 is a therapeutic target in JAK2-dependent myeloproliferative neoplasms in mice and humans. J. Clin. Investig. 2010, 120, 3578–3593. [Google Scholar] [CrossRef] [PubMed]
  277. Nieborowska-Skorska, M.; Maifrede, S.; Dasgupta, Y.; Sullivan, K.; Flis, S.; Le, B.V.; Solecka, M.; Belyaeva, E.A.; Kubovcakova, L.; Nawrocki, M.; et al. Ruxolitinib-induced defects in DNA repair cause sensitivity to PARP inhibitors in myeloproliferative neoplasms. Blood 2017, 130, 2848–2859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  278. Tyner, J.W. DNA distress creates lethal opportunity in MPN. Blood 2017, 130, 2814–2816. [Google Scholar] [CrossRef] [PubMed]
  279. Rose, M.; Burgess, J.T.; O’Byrne, K.; Richard, D.J.; Bolderson, E. PARP Inhibitors: Clinical Relevance, Mechanisms of Action and Tumor Resistance. Front. Cell Dev. Biol. 2020, 8, 564601. [Google Scholar] [CrossRef]
  280. Braun, L.M.; Zeiser, R. Immunotherapy in Myeloproliferative Diseases. Cells 2020, 9, 1559. [Google Scholar] [CrossRef]
  281. Alkharabsheh, O.; Frankel, A.E. Clinical Activity and Tolerability of SL-401 (Tagraxofusp): Recombinant Diphtheria Toxin and Interleukin-3 in Hematologic Malignancies. Biomedicines 2019, 7, 6. [Google Scholar] [CrossRef] [Green Version]
  282. Testa; Pelosi; Castelli, CD123 as a Therapeutic Target in the Treatment of Hematological Malignancies. Cancers 2019, 11, 1358. [CrossRef] [Green Version]
  283. Krishnan, A.; Pagane, M.; Roshal, M.; McGovern, E.; Stone-Molloy, Z.; Chen, J.; Brooks, C.; Levine, R.L.; Rampal, R.K. Evaluation of Tagraxofusp (SL-401) Alone and in Combination with Ruxolitinib for the Treatment of Myeloproliferative Neoplasms. Blood 2019, 134 (Suppl. 1), 2967. [Google Scholar] [CrossRef]
  284. Bose, P.; Masarova, L.; Verstovsek, S. Novel Concepts of Treatment for Patients with Myelofibrosis and Related Neoplasms. Cancers 2020, 12, 2891. [Google Scholar] [CrossRef] [PubMed]
  285. Mazzacurati, L.; Lambert, Q.T.; Pradhan, A.; Griner, L.N.; Huszar, D.; Reuther, G.W. The PIM inhibitor AZD1208 synergizes with ruxolitinib to induce apoptosis of ruxolitinib sensitive and resistant JAK2-V617F-driven cells and inhibit colony formation of primary MPN cells. Oncotarget 2015, 6, 40141–40157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  286. Mazzacurati, L.; Collins, R.J.; Pandey, G.; Lambert-Showers, Q.T.; Amin, N.E.; Zhang, L.; Stubbs, M.C.; Epling-Burnette, P.K.; Koblish, H.K.; Reuther, G.W. The pan-PIM inhibitor INCB053914 displays potent synergy in combination with ruxolitinib in models of MPN. Blood Adv. 2019, 3, 3503–3514. [Google Scholar] [CrossRef] [PubMed]
  287. Bartalucci, N.; Calabresi, L.; Balliu, M.; Martinelli, S.; Rossi, M.C.; Villeval, J.L.; Annunziato, F.; Guglielmelli, P.; Vannucchi, A.M. Inhibitors of the PI3K/mTOR pathway prevent STAT5 phosphorylation in JAK2V617F mutated cells through PP2A/CIP2A axis. Oncotarget 2017, 8, 96710–96724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  288. Fiskus, W.; Verstovsek, S.; Manshouri, T.; Smith, J.E.; Peth, K.; Abhyankar, S.; McGuirk, J.; Bhalla, K.N. Dual PI3K/AKT/mTOR Inhibitor BEZ235 Synergistically Enhances the Activity of JAK2 Inhibitor against Cultured and Primary Human Myeloproliferative Neoplasm Cells. Mol. Cancer Ther. 2013, 12, 577–588. [Google Scholar] [CrossRef] [Green Version]
  289. Moyo, T.K.; Sochacki, A.; Ayers, G.D.; Byrne, M.T.; Strickland, S.A.; Mohan, S.R.; Harrison, J.; Berry, L.D.; Dudley, C.V.; Severs, R.; et al. Preliminary Results from a Phase I Dose Escalation Trial of Ruxolitinib and the PI3Kδ Inhibitor TGR-1202 in Myelofibrosis. Blood 2016, 128, 1125. [Google Scholar] [CrossRef]
  290. Petiti, J.; Lo Iacono, M.; Rosso, V.; Andreani, G.; Jovanovski, A.; Podestà, M.; Lame, D.; Gobbi, M.D.; Fava, C.; Saglio, G.; et al. Bcl-xL represents a therapeutic target in Philadelphia negative myeloproliferative neoplasms. J. Cell. Mol. Med. 2020, 24, 10978–10986. [Google Scholar] [CrossRef]
  291. Winter, P.S.; Sarosiek, K.A.; Lin, K.H.; Meggendorfer, M.; Schnittger, S.; Letai, A.; Wood, K.C. RAS signaling promotes resistance to JAK inhibitors by suppressing BAD-mediated apoptosis. Sci. Signal. 2014, 7, ra122. [Google Scholar] [CrossRef] [Green Version]
  292. Waibel, M.; Solomon, V.S.; Knight, D.A.; Ralli, R.A.; Kim, S.K.; Banks, K.M.; Vidacs, E.; Virely, C.; Sia, K.C.; Bracken, L.S.; et al. Combined Targeting of JAK2 and Bcl-2/Bcl-xL to Cure Mutant JAK2-Driven Malignancies and Overcome Acquired Resistance to JAK2 Inhibitors. Cell Rep. 2013, 5, 1047–1059. [Google Scholar] [CrossRef] [Green Version]
  293. Harrison, C.N.; Garcia, J.S.; Mesa, R.A.; Somervaille, T.C.; Komrokji, R.S.; Pemmaraju, N.; Jamieson, C.; Papadantonakis, N.; Foran, J.M.; O’Connell, C.L.; et al. Results from a Phase 2 Study of Navitoclax in Combination with Ruxolitinib in Patients with Primary or Secondary Myelofibrosis. Blood 2019, 134 (Suppl. 1), 671. [Google Scholar] [CrossRef]
  294. Jutzi, J.S.; Kleppe, M.; Dias, J.; Staehle, H.F.; Shank, K.; Teruya-Feldstein, J.; Gambheer, S.M.M.; Dierks, C.; Rienhoff, H.Y.J.; Levine, R.L.; et al. LSD1 Inhibition Prolongs Survival in Mouse Models of MPN by Selectively Targeting the Disease Clone. HemaSphere 2018, 2, e54. [Google Scholar] [CrossRef] [PubMed]
  295. Liu, F.; Zhao, X.; Perna, F.; Wang, L.; Koppikar, P.; Abdel-Wahab, O.; Harr, M.W.; Levine, R.L.; Xu, H.; Tefferi, A.; et al. JAK2V617F-Mediated Phosphorylation of PRMT5 Downregulates Its Methyltransferase Activity and Promotes Myeloproliferation. Cancer Cell 2011, 19, 283–294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  296. Pastore, F.; Bhagwat, N.; Krishnan, A.; Pastore, A.; Li, B.; Bowman, R.L.; Xiao, W.; Viny, A.D.; Karzai, A.; Zouak, A.; et al. PRMT5 Inhibition Modulates E2F1 Methylation and Gene Regulatory Networks Leading to Therapeutic Efficacy in JAK2VF Mutant MPN. Blood 2019, 134 (Suppl. 1), 473. [Google Scholar] [CrossRef]
  297. Sonderegger, S.; Cerruti, L.; Tremblay, C.; Toulmin, E.; Saw, J.; Nebl, T.; Hannan, K.M.; Lane, S.W.; Falk, H.; Unnikrishnan, A.; et al. Small-Molecule Inhibition of PRMT5 Induces Translational Stress and p53 in JAK2V617F Mutant Myeloproliferative Neoplasms. Blood 2018, 132 (Suppl. 1), 53. [Google Scholar] [CrossRef]
  298. Bose, P.; Verstovsek, S. Investigational histone deacetylase inhibitors (HDACi) in myeloproliferative neoplasms. Expert Opin. Investig. Drugs 2016, 25, 1393–1403. [Google Scholar] [CrossRef] [PubMed]
  299. Amaru Calzada, A.; Todoerti, K.; Donadoni, L.; Pellicioli, A.; Tuana, G.; Gatta, R.; Neri, A.; Finazzi, G.; Mantovani, R.; Rambaldi, A.; et al. The HDAC inhibitor Givinostat modulates the hematopoietic transcription factors NFE2 and C-MYB in JAK2V617F myeloproliferative neoplasm cells. Exp. Hematol. 2012, 40, 634–645.e10. [Google Scholar] [CrossRef]
  300. Wang, Y.; Fiskus, W.; Chong, D.G.; Buckley, K.M.; Natarajan, K.; Rao, R.; Joshi, A.; Balusu, R.; Koul, S.; Chen, J.; et al. Cotreatment with panobinostat and JAK2 inhibitor TG101209 attenuates JAK2V617F levels and signaling and exerts synergistic cytotoxic effects against human myeloproliferative neoplastic cells. Blood 2009, 114, 5024–5033. [Google Scholar] [CrossRef]
  301. Harrison, C.N.; Kiladjian, J.-J.; Heidel, F.H.; Vannucchi, A.M.; Passamonti, F.; Hayat, A.; Conneally, E.; Martino, B.; Kindler, T.; Lipka, D.B.; et al. Efficacy, Safety, and Confirmation of the Recommended Phase 2 Starting Dose of the Combination of Ruxolitinib (RUX) and Panobinostat (PAN) in Patients (Pts) with Myelofibrosis (MF). Blood 2015, 126, 4060. [Google Scholar] [CrossRef]
  302. Gleitz, H.F.; Kramann, R.; Schneider, R.K. Understanding deregulated cellular and molecular dynamics in the haematopoietic stem cell niche to develop novel therapeutics for bone marrow fibrosis. J. Pathol. 2018, 245, 138–146. [Google Scholar] [CrossRef] [Green Version]
  303. Arranz, L.; Sánchez-Aguilera, A.; Martín-Pérez, D.; Isern, J.; Langa, X.; Tzankov, A.; Lundberg, P.; Muntión, S.; Tzeng, Y.-S.; Lai, D.-M.; et al. Neuropathy of haematopoietic stem cell niche is essential for myeloproliferative neoplasms. Nature 2014, 512, 78–81. [Google Scholar] [CrossRef]
  304. Fielding, C.; Méndez-Ferrer, S. Neuronal regulation of bone marrow stem cell niches. F1000Research 2020, 9, 614. [Google Scholar] [CrossRef] [PubMed]
  305. Herlihy, N.; Harrison, C.N.; McLornan, D.P. Exploitation of the neural-hematopoietic stem cell niche axis to treat myeloproliferative neoplasms. Haematologica 2019, 104, 639–641. [Google Scholar] [CrossRef] [PubMed]
  306. Archana, A.; Morrone, K.; Bartenstein, M.; Zhao, Z.J.; Verma, A.; Goel, S. Bone marrow fibrosis in primary myelofibrosis: Pathogenic mechanisms and the role of TGF-β. Stem Cell Investig. 2016, 3, 5. [Google Scholar]
  307. Teodorescu, P.; Pasca, S.; Jurj, A.; Gafencu, G.; Joelsson, J.P.; Selicean, S.; Moldovan, C.; Munteanu, R.; Onaciu, A.; Tigu, A.B.; et al. Transforming growth factor β-mediated micromechanics modulates disease progression in primary myelofibrosis. J. Cell. Mol. Med. 2020, 24, 11100. [Google Scholar] [CrossRef]
  308. Yue, L.; Bartenstein, M.; Zhao, W.; Ho, W.T.; Han, Y.; Murdun, C.; Mailloux, A.W.; Zhang, L.; Wang, X.; Budhathoki, A.; et al. Efficacy of ALK5 inhibition in myelofibrosis. JCI Insight 2017, 2, e90932. [Google Scholar] [CrossRef] [Green Version]
  309. Ceglia, I.; Dueck, A.C.; Masiello, F.; Martelli, F.; He, W.; Federici, G.; Petricoin, E.F.; Zeuner, A.; Iancu-Rubin, C.; Weinberg, R.; et al. Preclinical rationale for TGF-β inhibition as a therapeutic target for the treatment of myelofibrosis. Exp. Hematol. 2016, 44, 1138–1155.e4. [Google Scholar] [CrossRef] [Green Version]
  310. Jeremy Wen, Q.; Yang, Q.; Goldenson, B.; Malinge, S.; Lasho, T.; Schneider, R.K.; Breyfogle, L.J.; Schultz, R.; Gilles, L.; Koppikar, P.; et al. Targeting megakaryocytic-induced fibrosis in myeloproliferative neoplasms by AURKA inhibition. Nat. Med. 2015, 21, 1473–1480. [Google Scholar] [CrossRef]
  311. Pilling, D.; Gomer, R.H. The Development of Serum Amyloid P as a Possible Therapeutic. Front. Immunol. 2018, 9, 2328. [Google Scholar] [CrossRef] [Green Version]
  312. Verstovsek, S.; Manshouri, T.; Pilling, D.; Bueso-Ramos, C.E.; Newberry, K.J.; Prijic, S.; Knez, L.; Bozinovic, K.; Harris, D.M.; Spaeth, E.L.; et al. Role of neoplastic monocyte-derived fibrocytes in primary myelofibrosis. J. Exp. Med. 2016, 213, 1723–1740. [Google Scholar] [CrossRef]
  313. Verstovsek, S.; Hasserjian, R.P.; Pozdnyakova, O.; Veletic, I.; Mesa, R.A.; Foltz, L.; Mascarenhas, J.; Ritchie, E.K.; Palmer, J.; Silver, R.T.; et al. PRM-151 in Myelofibrosis: Efficacy and Safety in an Open Label Extension Study. Blood 2018, 132 (Suppl. 1), 686. [Google Scholar] [CrossRef]
  314. Verstovsek, S.; Mesa, R.A.; Foltz, L.M.; Gupta, V.; Mascarenhas, J.O.; Ritchie, E.K.; Hoffman, R.; Silver, R.T.; Kremyanskaya, M.; Pozdnyakova, O.; et al. PRM-151 in Myelofibrosis: Durable Efficacy and Safety at 72 Weeks. Blood 2015, 126, 56. [Google Scholar] [CrossRef]
  315. Wang, J.-C.; Chen, C.; Kundra, A.; Kodali, S.; Pandey, A.; Wong, C.; Cheung, T.; Gotlieb, V.; Joseph, G.; Tribie, S. Programmed Cell Death Receptor (PD-1) Ligand (PD-L1) expression in Philadelphia chromosome-negative myeloproliferative neoplasms. Leuk. Res. 2019, 79, 52–59. [Google Scholar] [CrossRef] [PubMed]
  316. Holmström, M.O.; Riley, C.H.; Skov, V.; Svane, I.M.; Hasselbalch, H.C.; Andersen, M.H. Spontaneous T-cell responses against the immune check point programmed-death-ligand 1 (PD-L1) in patients with chronic myeloproliferative neoplasms correlate with disease stage and clinical response. OncoImmunology 2018, 7, e1433521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  317. Prestipino, A.; Emhardt, A.J.; Aumann, K.; O’Sullivan, D.; Gorantla, S.P.; Duquesne, S.; Melchinger, W.; Braun, L.; Vuckovic, S.; Boerries, M.; et al. Oncogenic JAK2V617Fcauses PD-L1 expression, mediating immune escape in myeloproliferative neoplasms. Sci. Transl. Med. 2018, 10, eaam7729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  318. Pinton, L.; Solito, S.; Damuzzo, V.; Francescato, S.; Pozzuoli, A.; Berizzi, A.; Mocellin, S.; Rossi, C.R.; Bronte, V.; Mandruzzato, S. Activated T cells sustain myeloid-derived suppressor cell-mediated immune suppression. Oncotarget 2016, 7, 1168–1184. [Google Scholar] [CrossRef] [Green Version]
  319. John Mascarenhas, M.; Gabriela Hobbs, M.D. PD-1 Inhibition in Advanced Myeloproliferative Neoplasms, 2017-02-27 ed.; U.S. National Library of Medicine: Bethesda, MD, USA, 2017.
  320. Verstovsek, S. Nivolumab in Treating Patients With Primary Myelofibrosis, Post-Essential Thrombocythemia Myelofibrosis, or Post-Polycythemia Vera Myelofibrosis, 2019-05-15 ed.; U.S. National Library of Medicine: Bethesda, MD, USA, 2019.
  321. Brady Stein, M. MHS, Durvalumab in Treating Patients With Primary, Post-Polycythemia Vera, or Post-Essential Thrombocythemia Myelofibrosis, 2019-08-20 ed.; U.S. National Library of Medicine: Bethesda, MD, USA, 2016.
  322. Holmström, M.O.; Martinenaite, E.; Ahmad, S.M.; Met, Ö.; Friese, C.; Kjær, L.; Riley, C.H.; Thor Straten, P.; Svane, I.M.; Hasselbalch, H.C.; et al. The calreticulin (CALR) exon 9 mutations are promising targets for cancer immune therapy. Leukemia 2018, 32, 429–437. [Google Scholar] [CrossRef]
  323. How, J.; Hobbs, G.S.; Mullally, A. Mutant calreticulin in myeloproliferative neoplasms. Blood 2019, 134, 2242–2248. [Google Scholar] [CrossRef] [Green Version]
  324. Jacob, H.; Grauslund, M. CALR Exon 9 Mutant Peptide Vaccine to Patients With CALR-mutant Myeloproliferative Neoplasms, 2018-06-25 ed.; U.S. National Library of Medicine: Bethesda, MD, USA, 2018.
  325. Sørensen, A.L.; Mikkelsen, S.U.; Knudsen, T.A.; Bjørn, M.E.; Andersen, C.L.; Bjerrum, O.W.; Brochmann, N.; Patel, D.A.; Gjerdrum, L.M.R.; El Fassi, D.; et al. Ruxolitinib and interferon-α2 combination therapy for patients with polycythemia vera or myelofibrosis: A phase II study. Haematologica 2020, 105, 2262–2272. [Google Scholar] [CrossRef] [Green Version]
  326. Mascarenhas, J.; Kremyanskaya, M.; Hoffman, R.; Bose, P.; Talpaz, M.; Harrison, C.N.; Gupta, V.; Leber, B.; Sirhan, S.; Kabir, S.; et al. MANIFEST, a Phase 2 Study of CPI-0610, a Bromodomain and Extraterminal Domain Inhibitor (BETi), As Monotherapy or "Add-on" to Ruxolitinib, in Patients with Refractory or Intolerant Advanced Myelofibrosis. Blood 2019, 134 (Suppl. 1), 670. [Google Scholar] [CrossRef]
  327. Study of Oral Navitoclax Tablet in Combination with Oral Ruxolitinib Tablet when Compared with Oral Ruxolitinib Tablet to Assess Change in Spleen Volume in Adult Participants with Myelofibrosis (TRANSFORM-1), 2020-07-15 ed.; U.S. National Library of Medicine: Bethesda, MD, USA, 2020.
  328. Safety, Efficacy, and Assessment of Change in Spleen Volume Study of Oral Navitoclax Tablet in Combination with Oral Ruxolitinib Tablet in Adult Participants with Relapsed/Refractory Myelofibrosis (TRANSFORM-2), 2020-07-13 ed.; U.S. National Library of Medicine: Bethesda, MD, USA, 2020.
  329. Dagher, T.; Maslah, N.; Edmond, V.; Cassinat, B.; Vainchenker, W.; Giraudier, S.; Pasquier, F.; Verger, E.; Niwa-Kawakita, M.; Lallemand-Breitenbach, V.; et al. JAK2V617F myeloproliferative neoplasm eradication by a novel interferon/arsenic therapy involves PML. J. Exp. Med. 2021, 218, e20201268. [Google Scholar] [CrossRef]
  330. Maslah1, N.; Dagher, T.; Edmond, V.; Cassinat, B.; Lallemand-Breitenbach, V.; Verger, E.; Plo, I.; Kiladjian, J.-J.; Villeval, J.-L.; de Thé, H. Specific and Synergistic Targeting of JAK2V617F Cells By Interferon Alpha and Arsenic. Blood 2018, 132 (Suppl. 1), 52. [Google Scholar] [CrossRef]
  331. Ichiro Kawashima, K.K. Metformin inhibits JAK2V617F activity in MPN cells by activating AMPK and PP2A complexes containing the B56α subunit. Exp. Hematol. 2016, 44, 1156–1165. [Google Scholar] [CrossRef] [PubMed]
  332. Machado-Neto, J.A.; Fenerich, B.A.; Scopim-Ribeiro, R.; Eide, C.A.; Coelho-Silva, J.L.; Dechandt, C.R.P.; Fernandes, J.C.; Rodrigues Alves, A.P.N.; Scheucher, P.S.; Simões, B.P.; et al. Metformin exerts multitarget antileukemia activity in JAK2V617F-positive myeloproliferative neoplasms. Cell Death Dis. 2018, 9, 311. [Google Scholar] [CrossRef] [PubMed]
  333. Campos, P.D.M.; Pagnano, K.B.; Mancuso, R.; Teófilo, F.B.S.; Coelho-Silva, J.L.; Della Via, F.I.; Freitas, L.L.L.; Carvalho, H.F.; Costa, F.F.; Traina, F.; et al. Analysis of Metformin Effects on Bone Marrow Fibrosis and Disease Progression in Primary Myelofibrosis Patients: Preliminary Results of an Open Label Phase II Trial (FIBROMET). Blood 2019, 134 (Suppl. 1), 554. [Google Scholar] [CrossRef]
  334. Azizian, N.G.; Li, Y. XPO1-dependent nuclear export as a target for cancer therapy. J. Hematol. Oncol. 2020, 13. [Google Scholar] [CrossRef] [PubMed]
  335. Yan, D.; Pomicter, A.D.; Tantravahi, S.; Mason, C.C.; Senina, A.V.; Ahmann, J.M.; Wang, Q.; Than, H.; Patel, A.B.; Heaton, W.L.; et al. Nuclear–Cytoplasmic Transport Is a Therapeutic Target in Myelofibrosis. Clin. Cancer Res. 2019, 25, 2323–2335. [Google Scholar] [CrossRef] [PubMed]
  336. Srinivas Tantravahi, M. Selinexor in Myelofibrosis Refractory or Intolerant to JAK1/2 Inhibitors (ESSENTIAL), 2018-08-13 ed.; U.S. National Library of Medicine: Bethesda, MD, USA, 2018.
  337. Uras, I.Z.; Maurer, B.; Nivarthi, H.; Jodl, P.; Kollmann, K.; Prchal-Murphy, M.; Milosevic Feenstra, J.D.; Zojer, M.; Lagger, S.; Grausenburger, R.; et al. CDK6 coordinates JAK2V617F mutant MPN via NF-κB and apoptotic networks. Blood 2019, 133, 1677–1690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  338. Uras, I.Z.; Sexl, V.; Kollmann, K. CDK6 Inhibition: A Novel Approach in AML Management. Int. J. Mol. Sci. 2020, 21, 2528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  339. Mohi, G.; Dutta, A.; Nath, D.; Yang, Y. The CDK6 Inhibitor Palbociclib in Combination with Ruxolitinib Remarkably Improves Myelofibrosis in Murine Models. Blood 2019, 134 (Suppl. 1), 4200. [Google Scholar] [CrossRef]
  340. Lu, M.; Xia, L.; Salama, M.E.; Hoffman, R. Splenic Micro Environmental Cells from Patients with Myelofibrosis Elaborate a Cascade of Cytokines and Serve As a Niche for Malignant Hematopoiesis. Blood 2016, 128, 953. [Google Scholar] [CrossRef]
Figure 1. Interplay between cell-intrinsic and cell-extrinsic activities in the disease initiation of MPN. MPN: myeloproliferative neoplasm; MF: myelofibrosis.
Figure 1. Interplay between cell-intrinsic and cell-extrinsic activities in the disease initiation of MPN. MPN: myeloproliferative neoplasm; MF: myelofibrosis.
Ijms 22 00659 g001
Figure 2. Disease initiation and progression in Ph-negative MPNs. (A) shows the acquisition of somatic driver mutations (JAK2V617F, CALR, MPL) in a haematopoietic stem cell (HSC), leading to myeloproliferation. (B) shows the acquisition of somatic driver mutation, JAK2V617F, in HSC without progression to MPN. One of the reasons is the insufficient JAK2V617F allele burden to give rise to a MPN phenotype. (C) shows the acquisition of non-driver mutations without progression to MPN, indicating the presence of clonal hematopoiesis of indeterminate potential (CHIP). (D) The presence of CHIP increases the chance of HSC to acquire driver mutations, leading to myeloproliferation. (D,E) Acquisition of additional non-driver mutations in MPN stem cells could lead to disease progression or leukemic transformation to secondary acute myeloid leukemia (sAML).
Figure 2. Disease initiation and progression in Ph-negative MPNs. (A) shows the acquisition of somatic driver mutations (JAK2V617F, CALR, MPL) in a haematopoietic stem cell (HSC), leading to myeloproliferation. (B) shows the acquisition of somatic driver mutation, JAK2V617F, in HSC without progression to MPN. One of the reasons is the insufficient JAK2V617F allele burden to give rise to a MPN phenotype. (C) shows the acquisition of non-driver mutations without progression to MPN, indicating the presence of clonal hematopoiesis of indeterminate potential (CHIP). (D) The presence of CHIP increases the chance of HSC to acquire driver mutations, leading to myeloproliferation. (D,E) Acquisition of additional non-driver mutations in MPN stem cells could lead to disease progression or leukemic transformation to secondary acute myeloid leukemia (sAML).
Ijms 22 00659 g002
Table 1. Tyrosine kinase inhibitors and their effects on CML stem cells.
Table 1. Tyrosine kinase inhibitors and their effects on CML stem cells.
TKIObservationsReferences
Imatinib
  • Reduction in CD26, a specific CML stem cell marker, in circulation after imatinib treatment
  • In-vitro enhanced mitochondrial oxidative phosphorylation upon imatinib discontinuation, leading to CML stem cell proliferation
[16,34]
Dasatinib
  • Lower CD34+ cell proliferation in dasatinib-treated cells compared to imatinib-treated group upon TKI discontinuation
[34]
Nilotinib
  • Induction of apoptosis in CML CD34+/CD38 stem cells
  • Inhibition of CML stem cell engraftment in murine models
  • More rapid and deeper CD34+/linPh+ cell clearance may might increase treatment-free remission
[36,37]
Bosutinib
  • Higher potency in inhibiting Src phosphorylation, hence inhibiting CML primitive progenitor cells compared to imatinib
  • Growth inhibition of CD34+/CD38 CML stem cells in combination with gemtuzumab ozogamicin
[31,38]
Ponatinib
  • Higher efficacy in eradicating CML-associated LSK cells than imatinib and dasatinib in murine models
[22]
TKI: Tyrosine kinase inhibitor; CML: Chronic myeloid leukaemia: LSK cells: lin−Sca-1+c-Kit+ cells.
Table 2. Targeting of JAK2V617F and CALR-mutated MPN by IFNα preparations.
Table 2. Targeting of JAK2V617F and CALR-mutated MPN by IFNα preparations.
IFNα PreparationMPN SubtypeObservationsReferences
IFNαPV, ET, MF
  • Preferential targeting JAK2V617F HSPC, especially in CD90+/CD34+/CD38 HSC-enriched progenitors, compared to mature blood cells (p < 0.05).
  • Faster response in homozygous JAK2V617F clones than that of heterozygous clones.
  • Faster response in targeting JAK2V617F HSPC than CALR-mutated HSPC.
[71]
CALR-mutated ET, MF
  • Sixty-two percent showed decrease in CALR-mutant allele burden (median decline: 29% from baseline).
  • Nineteen percent (4 out of 21) showed MR with >50% mutant allele burden reduction.
[62,72]
Peg-IFNα-2aCALR-mutated ET
  • Reduction of median CALR-mutant allele burden from 41% to 26%.
  • Seven percent (2 out of 31) showed complete MR.
  • Variation in responses in patients with additional molecular mutations
[73]
Ropeginterferon α-2bPV
  • Preferential inhibition and reduction of JAK2V617F-mutated primary hematopoietic progenitors with sparing of JAK2-wild type cells.
[69]
2.
Increase in proportion of wild-type to JAK2V617F-mutated colonies after 12 months of treatment compared with hydroxyurea, reflecting reduction of malignant progenitors in the BM.
PV: polycythemia vera; ET: essential thrombocythemia; MF: myelofibrosis; HSPC: hematopoietic stem and progenitor cells; HSC: hematopoietic stem cell; MR: molecular response; Peg-IFNα-2a: pegylated interferon-alpha-2a; IFNα: interferon-alpha; BM: bone marrow.
Table 3. Clinical trials of novel therapies as single agents for targeting Ph-negative MPNs.
Table 3. Clinical trials of novel therapies as single agents for targeting Ph-negative MPNs.
Novel TherapyMPN Subtype ObservationsReferences
Telomerase inhibitor (imetelstat)DIPSS-plus Int-2/high risk MF
  • CR and PR: 21% (7 out of 33)
  • (Median duration of CR: 18 months; median duration of PR: 10 months)
  • Reversal of marrow fibrosis in all 4 CR patients. Three of them demonstrated MR.
  • Thirty-eight percent SF3B1/U2AF1 patients showed complete response, which was higher than that of patients who did not harbour SF3B1/U2AF1 mutations (4%) (p = 0.04)
[244]
DIPSS Int-2/high risk MF
  • ≥35% SVR at week 24: 10.2% in 9.4mg/kg arm; 0% in 4.7mg/kg arm
  • Median OS: NR in 9.4mg/kg arm; 19.9 months in 4.7mg/kg arm
  • No significant difference of OS in all 3 driver mutations (JAK2V617F, CALR, MPL)
  • Higher median OS in triple-negative patients in 9.4mg/kg arm
[245]
ET
  • HR: 100%
  • MR in 88% JAK2V617F patients
  • Reduction by 15–66% in CALR and MPL mutant allele burden
[246]
MDM2 inhibitor (idasanutlin)PV/ET (only 1 ET patient was enrolled)
  • ORR after 6 cycles: 58% in idasanutlin monotherapy; 50% when combined with Peg-IFNα-2a
  • (Median duration of response: 16.8 months)
  • Median reduction of JAK2V617F VAF: 43%
[247]
HSP90 inhibitor (AUY922)PV, ET, MF (IPSS score: ≥2)
  • Reduction in splenomegaly in all patients
  • Effect to JAK2V617F mutant allele burden: modest
[248]
CD123 (tagraxofusp)DIPSS-plus int-1, int-2, high risk MF
  • Spleen size reduction: 53%
  • Four out of five patients who had baseline splenomegaly and monocytosis showed spleen size reduction
  • ≥50% TSS score reduction: 45%
  • Effect of mutant allele burden: NR
[249]
LSD-1 inhibitor (IMG-7289)IPSS Int-2, high risk MF
  • SVR after cycle 1: 50%
  • ≥50% TSS score reduction after cycle 1: 21%
  • Reduced BM fibrosis at day 84: 15.4%
[250,251]
ETOngoing[252]
HDAC inhibitor (givinostat)PV
  • ORR: 80.6%
  • ≥35% SVR: 19.4%
  • Reduction of JAK2V617F mutant allele burden: moderate
  • Complete response after a median of 4 years: 11%
  • Partial response: 89%
  • Normalization of spleen size: 56%
  • Reduction of JAK2V617F mutant allele burden: 22%
[74,253]
HDAC inhibitor (panobinostat)PMF, SMF
  • Poorly tolerated. Only 45.7% complete ≥2 cycles of treatment
  • Reduction of JAK2V617F mutant allele burden: 36.82% (pre-treated) to 0.86% (cycle 4)
  • Downregulation of pro-inflammatory cytokines
[254]
PRMT5 inhibitor (PRT-543)R/R MFOngoing[255]
β-3 sympathomimetic (mirabegron)39 JAK2 V617F MPN patients (7 ET, 21 PV, 5 PMF, 3 post-PV MF, 3 post-ET MF)
  • ≥50% reduction in JAK2V617F mutant allele burden at week 24 (primary endpoint): 0%;
  • Twenty-five percent reduction of JAK2V617F mutant allele burden is seen in 1 patient
  • Increased levels of nestin+ MSCs
  • Reduction of reticulin fibre content from grade 1 to 0.5 at week 24; the effect is restricted in patients not treated with hydroxyurea. The mechanism is unknown
[256]
ActRIIA (luspatercept) DIPSS int-1, int-2, high risk MF
  • Improvement in MF-associated anemia as shown by RBC-TI and Hb increase
[257]
ActRIIA (sotatercept) DIPSS int-1, int-2, high risk MF
  • Response rate: 29%
[258]
AURKA inhibitor (alisertib)DIPSS int-1, int-2, high risk MF
  • Reduction of splenomegaly: 29%
  • Decrease in symptom burden: 32%
  • Inhibition of marrow fibrosis
  • Normalization of megakaryocytes
[259,260]
Anti-PD-1 (nivolumab)PMF, post-PV, ET MF, hepatomegaly, splenomegaly
  • Primary outcome: efficacy and clinical activity in MF
  • Terminated due to serious adverse effects in 75% patients
[261]
DIPSS: Dynamic International Prognostic Scoring System; IPSS: International Prognostic Scoring System; PV: polycythaemia vera; ET: essential thrombocythaemia; MF: myelofibrosis; PMF: primary myelofibrosis; SMF: secondary myelofibrosis; Int-2: Intermediate-2; CR: complete remission; PR: partial remission; MR: molecular response; HR: haematological response; SVR: spleen volume reduction; NR: not reached; ORR: overall response rate; DLT: dose-limiting toxicity; VAF: variant allele frequency; TSS: Total Symptom Score; MDM2: Mouse double minute 2 homolog; Peg-IFNα-2a: pegylated interferon-alpha-2a; HSP: heat shock protein; LSD-1: lysine specific demethylase-1; HDAC: histone deacetylase; PRMT5: protein arginine methyltransferase 5; R/R: relapsed/refractory; MTD: maximally tolerated dose; MSC: mesenchymal stem cell; ActRIIA: Activin receptor IIA ligand trap; RBC-TI: red blood cell transfusion independence; TRAE: treatment-related adverse effects; AURKA: aurora kinase A. Refer to Supplementary Materials File S1 on the prognostic models of MF.
Table 4. Novel therapies in combination with standard treatment (interferon-alpha/ruxolitinb) in targeting Ph-negative MPNs.
Table 4. Novel therapies in combination with standard treatment (interferon-alpha/ruxolitinb) in targeting Ph-negative MPNs.
CombinationMPN SubtypeStudyObservationsReferences
Combination with IFNα
MDM2 inhibitor (RG-7112) ± Peg-IFNα-2aPV, PMFPreclinical
  • PV: Low dose (200 nM) of RG7112 and 200ng/mL Peg-IFNα-2a reduced JAK2V617F heterozygous colonies and increased JAK2 WT colonies
  • PV and PMF: RG7112 and Peg-IFNα-2a activated p53 pathway, resulting MPN CD34+ cell apoptosis
  • PV and PMF: RG7112 and Peg-IFNα-2a reduced repopulation of MPN CD34+ cells in BM and spleens of NSG mice
  • Reduction of JAK2V617F mutant allele burden: PV: 75%; PMF: 80%
[267]
IFNαPV, MFPhase II
  • PV: 31% remission (CR: 9%; PR: 22%)
  • MF: 44% remission (CR: 28%; PR: 17%)
  • Median JAK2V617F allele burden reduced from 47% from baseline to 12% after 2 years (CMR: 2%; PMR: 39%)
[325]
Combinations with Ruxolitinib
BET inhibitor (CPI-0610) ± ruxolitinibMF Phase II
  • SVR: 94%
  • TSS improvement: 93%
  • ≥50% TSS improvement: 39%
  • ≥1 grade marrow fibrosis and/or reticulin improvement: 58%
  • Increase in Hb by 1.5 mg/dL post-baseline (CPI-0610 monotherapy: 50%; CPI-0610 + ruxolitinib: 16%)
[326]
CD123 (tagraxofusp) ± ruxolitinibMF-AP, BPPreclinical
  • Combination of tagraxofusp and ruxolitinib had a lower IC50 JAK2V617F mutant cell line compared to tagraxofusp monotherapy
  • Reduction of colony formation regardless of HMR mutation (TP53, ASXL1) in both single agent and combination
[283]
Bcl-2 family protein inhibitor (navitoclax) + ruxolitinibPMF, SMFPhase II
  • >5% reduction in mutant allele burden: 42%
  • ≥35% SVR at week 24: 29%
  • ≥1 grade improvement in BM fibrosis: 25%
[293]
MFPhase IIIOngoing[327]
R/R MFPhase IIIOngoing[328]
HDAC inhibitor (panobinostat) + ruxolitinibIPSS int-1, int-2, high risk PMF, SMFPhase Ib
  • ≥35% SVR at week 24: 57%
  • ≥35% SVR at week 48: 39%
  • ≥20% reduction in mutant allele burden at week 48: 29%
[301]
PI3K-delta inhibitor (umbralisib; TGR-1202) + ruxolitinibMFPhase I
  • Improvement in haematological parameters: 77.8% (7 out of 9)
  • Median reduction of TSS: 33%
[289]
Pan-PI3K inhibitor (buparlisib; BKM120) + ruxolitinibIPSS int, high riskPhase Ib
  • ≥ 35% SVR: 55.6% (5 out of 9) in JAK naive arm; 42.9% (3 out of 7) in prior JAK inhibitor arm
[261]
SAP (PRM-151) ± ruxolitinibDIPSS int-1, int-2 PMF, SMFPhase II
  • BM morphological response at week 72: 54%
  • ≥25% reduction of palpable spleen size: 50%
  • 100% reduction of TSS between week 24 to 72: 38%
[314]
PMF, SMFPhase II
  • Median reduction of spleen size at week 24: 26.1%
[313]
CR: complete remission; PR: partial remission; CMR: complete molecular remission; PMR: partial molecular remission; Mouse double minute 2 homolog; Peg-IFNα-2a: pegylated interferon-alpha-2a; IFNα: interferon-alpha; BM: bone marrow; BET: Bromodomain and extra-terminal; SVR: spleen volume reduction; TSS: Total Symptom Score; Hb: haemoglobin; AP: accelerated phase; BP: blast phase; IC50: half maximal inhibitory concentration; HMR: high molecular risks; BM: bone marrow; R/R: relapsed/refractory; HDAC: histone deacetylase; PI3K-delta: Phosphatidylinositol 3-kinase-delta; Int: intermediate; SAP: serum amyloid P.
Table 5. Other potential therapeutic targets in MPN in preclinical phase.
Table 5. Other potential therapeutic targets in MPN in preclinical phase.
Novel TherapyCombinationMechanismsObservationsReferences
Arsenic trioxidePeg-IFNα-2aIn MPN, expression of JAK2V617F increases PML expression
ATO and IFNα enhanced the formation of PML-NB, amplifying activation of p53 and the eradication of JAK2V617F MPN stem cells
  • Combination treatment significantly reduced JAK2V617F-mutated erythroid colonies and BFU-E progenitors, while sparing JAK2-WT progenitors
  • ATO enhanced clearance of JAK2V617F-mutated LSCs by IFNα, leading to long-term disease clearance in vivo
  • Combination treatment significantly increased PML-NB formation
  • Marked reduction in JAK2V617F mutant allele burden in granulocytes, platelets and erythrocytes in vivo
[329,330]
MetforminN/AActivation of AMPK pathway suppresses mTOR and reduces synthesis of anti-apoptotic Mcl-1Apoptosis of JAK2V617F cell lines[331,332]
Attenuation of JAK2V617F downstream signaling by activating B56α subunit of PP2AReduced JAK2 and STAT5 phosphorylation in MPN cells[331]
RuxolitinibDelayed transition from G1 to S phase by downregulation of cyclin D1 and upregulation of p27 Combination treatment showed significant downregulation of cyclin D1 and upregulation of p27 in PCR and qPCR[332]
N/AFIBROMET (phase II clinical trial): metformin is safe and well-tolerated. Reduction in BM fibrosis was seen but the results were not statistically significant due to small sample size[333]
XPO1 inhibitor/SINE compounds N/AXPO1 exports tumour suppressor proteins (e.g., p53, NPM, p27) from the nucleus to cytosol
XPO1 inhibitor (KPT-330, KPT-8602) decreases NCT
Nuclear retention of p53 in MF CD34+ cells, leading to apoptosis and inhibition of colony formation in MF CD34+ cells[334,335]
RuxolitinibCo-treatment showed greater diminution of JAK2V617F-expressing cells in blood and spleen in mouse model
N/AESSENTIAL (phase II clinical trial): ongoing; Assessment of safety and efficacy of selinexor in MF patients who are refractory and intolerant to JAK1/2 inhibitors[336]
CDK6 inhibitorN/AOverexpressed CDK6 in MPN increases chronic inflammation
-
CDK6 in MPN binds to NF-κB p65 subunit to increase NF-κB pathway and IL-8 expression
-
CDK6 reduces the transcription of genes that encode IkB, leading to NF-κB activation to drive chronic inflammation
Inhibition of CDK6 reduces inflammation
Absence of CDK6 decreased spleen size and induced symptom relief in murine modelsReduced pro-inflammatory cytokines in plasma of JAK2V617F/CDK6−/− murine modelsPalbociclib is a CDK4/6 inhibitor that decreased splenomegaly in MPL mouse models, synergistic effect was seen when co-treated with ruxolitinib[337,338,339]
Promotion of quiescence and deactivation of JAK2V617F MPN stem cellsJAK2V617F/CDK6−/− murine models had elevated short term LT-HSCs
ApoptosisPalbociclib drove apoptosis in JAK2V617F haematopoietic cells in combination with ruxolitinib
ReparixinN/AReparixin inhibits IL-8 receptor and reduces proliferation of splenic endothelial cells, leading to a disrupted niche that cannot sustain MF HSC growthMF spleen showed elevated LCN2 levels that increased MF haematopoiesis
Reparixin led to the reduction of MF CD34+ cells by 35%
[14,268,340]
N/A: not applicable (used as single agent); ATO: arsenic trioxide; IFNα: interferon-alpha; PML: promyelocytic leukemia protein; NB: nuclear bodies; LSC: leukemic stem cells; BFU-E: burst-forming unit-erythroid; WT: wild type; AMPK: AMP-activated protein kinase; mTOR: mammalian target of rapamycin; Mcl-1: myeloid cell leukemia 1; SHP2: SH2 containing protein tyrosine phosphatase-2; ROS: reactive oxygen species; PP2A: protein phosphatase 2; PCR: polymerase chain reaction; BM: bone marrow; XPO1: exportin 1; SINE: selective inhibitors of nuclear export; NCT: nuclear cytoplasmic transport; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; IL-8: interleukin-8; IkB: inhibitor of NF-κB protein; LT-HSC: long term hematopoietic stem cell; LCN2: lipocalin-2.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yung, Y.; Lee, E.; Chu, H.-T.; Yip, P.-K.; Gill, H. Targeting Abnormal Hematopoietic Stem Cells in Chronic Myeloid Leukemia and Philadelphia Chromosome-Negative Classical Myeloproliferative Neoplasms. Int. J. Mol. Sci. 2021, 22, 659. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22020659

AMA Style

Yung Y, Lee E, Chu H-T, Yip P-K, Gill H. Targeting Abnormal Hematopoietic Stem Cells in Chronic Myeloid Leukemia and Philadelphia Chromosome-Negative Classical Myeloproliferative Neoplasms. International Journal of Molecular Sciences. 2021; 22(2):659. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22020659

Chicago/Turabian Style

Yung, Yammy, Emily Lee, Hiu-Tung Chu, Pui-Kwan Yip, and Harinder Gill. 2021. "Targeting Abnormal Hematopoietic Stem Cells in Chronic Myeloid Leukemia and Philadelphia Chromosome-Negative Classical Myeloproliferative Neoplasms" International Journal of Molecular Sciences 22, no. 2: 659. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22020659

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