Chronic myeloid leukemia (CML) was considered for a long time one of the most hostile leukemia that was incurable for most of the patients, predominantly due to the extreme resistance to chemotherapy. Part of the resistance to cell death (apoptosis) is the result of increased levels of anti-apoptotic and decreased levels of pro-apoptotic member of the BCL-2 family induced by the BCR-ABL1 oncoprotein. BCR-ABL1 is a constitutively active tyrosine kinase responsible for initiating multiple and oncogenic signaling pathways. With the development of specific BCR-ABL1 tyrosine kinase inhibitors (TKIs) CML became a much more tractable disease. Nevertheless, TKIs do not cure CML patients and a substantial number of them develop intolerance or become resistant to the treatment. Therefore, novel anti-cancer strategies must be developed to treat CML patients independently or in combination with TKIs. Here, we will discuss the mechanisms of BCR-ABL1-dependent and -independent resistance to TKIs and the use of BH3-mimetics as a potential tool to fight CML.

Chronic Myeloid Leukemia (CML) is a Philadelphia chromosome (Ph1)-derived myeloproliferative disease [1] characterized by the expansion of leukemic cells extremely resistant to chemotherapy [2–8]. The reciprocal translocation between chromosomes 9 and 22 — t(9;22)(q34;q11) that originates the Ph1 chromosome also generates the chimeric BCR-ABL1 tyrosine-kinase. BCR-ABL1 constitutive enzymatic activity is responsible for a complex signaling transduction cascade [9] that accounts for the malignant phenotype observed in CML, including the strong resistance to chemotherapy-induced cell death (largely apoptosis).

BCR-ABL1 inhibits apoptosis induced by a variety of stimuli, including chemotherapeutic drugs, oxidative stress, UV or γ-irradiation, growth factor withdrawal and death receptor ligands [5,6,8]. To a great extent, BCR-ABL1-mediated resistance to apoptosis involves up-regulation of anti-apoptotic and down-regulation of pro-apoptotic BCL-2 (B-cell lymphoma/leukemia-2 gene) family members [5,6,10] thereby preventing the translocation of cytochrome c from the mitochondria to the cytosol and the consequent activation of effector caspases [5,6].

Over the past three decades, the BCL-2 family of intracellular proteins have emerged as central regulators of apoptosis signaling, dictating cellular fate by inhibiting or promoting cell death. Apoptosis can occur via two distinct cellular pathways — the intrinsic (aka mitochondrial, stress-induced, BCL-2-regulated) and the extrinsic (aka death receptor-induced) pathways [11,12]. The intrinsic or BCL-2-regulated apoptotic pathway can be initiated in conceivably all cell types in response to a plethora of intrinsic stress stimuli. These stimuli include DNA damage, oxidative stress, endoplasmic reticulum (ER) stress, aberrant calcium flux and nutrient deprivation. Once activated, the intrinsic apoptotic pathway leads to mitochondrial outer membrane permeabilization (MOMP). This is mediated by the pro-apoptotic multi BH (BCL-2 homology) domain BCL-2 family members BAX and BAK [13]. MOMP causes release of apoptogenic proteins from the mitochondrial intermembrane space into the cytoplasm. This includes cytochrome c as well as or SMAC/DIABLO, which both drive activation of the cascade of aspartate-specific cysteine proteases (caspases) that bring about the ordered demolition of the dying cells.

The members of the BCL-2 family proteins contain up to four (relatively loosely) conserved BCL-2 homology (BH) domains, BH1–BH4. Based on structure and function, the BCL-2 proteins can be classified into three groups: (a) the pro-survival proteins (BCL-2, BCL-XL, BCL-W, MCL-1, BFL-1/A1); (b) the pro-apoptotic multi-BH domain containing effectors of apoptosis (BAK, BAX, BOK), and (c) the pro-apoptotic BH3-only proteins (BIM, PUMA, BID, NOXA, BMF, BIK, BAD, HRK) [13]. Apoptosis signaling is controlled by complex interactions between the pro-survival BCL-2 family members, the pro-apoptotic BH3-only proteins and the effectors of apoptosis, BAX and BAK. The pro-survival BCL-2 proteins can either directly inhibit the effector proteins BAX and BAK by binding to them or by binding to the BH3-only proteins, thereby preventing them from activating the effector proteins.

The BH3-only proteins are essential for the initiation of apoptosis signaling. Their levels are increased in response to diverse stresses through a broad range of transcriptional, post-transcriptional and post-translational processes [13–15]. The BH3-only proteins such as BIM, PUMA and t-BID (the proteolytically activated form of BID), can bind to and inhibit the pro-survival BCL-2 proteins [16,17], thereby releasing BAX and BAK from their restraint, unleashing MOMP and the downstream events in apoptosis. BAD selectively binds to BCL-2, BCL-XL and BCL-W, whereas NOXA selectively binds to MCL-1 and A1 [16]. Importantly, certain BH3-only proteins, including PUMA and BIM, can also directly activate BAX and BAK. BAX and BAK also differ in their ability to bind to the pro-survival BCL-2 proteins. BAX associates with all the pro-survival proteins whereas BAK associates specifically with MCL-1 and BCL-XL but surprisingly not with BCL-2, BCL-W, or A1 [18]. Interestingly, the effector protein BOK is not regulated by any of the pro-survival or the BH3-only proteins.

Therefore, the balance between the different pro-survival and pro-apoptotic BCL-2 family members and the interactions between them dictate cellular outcome. Once activated, the effector proteins BAX and BAK oligomerize to form pores in the outer mitochondrial membrane leading to MOMP [19], which is considered the point of no return in apoptosis signaling [20,21]. While BAK is already located at the mitochondrial outer membrane in unstressed cells, BAX translocates from the cytosol to the mitochondrial membrane in response to stresses that induce apoptosis [22,23]. Following MOMP, the formation of the apoptosome, comprising cytochrome c, APAF-1 and dATP, leads to the recruitment and activation of the initiator caspase, caspase-9, which then unleashes the downstream executioner caspases, caspase-3, -6, -7, that dismantle the cell in an orderly manner.

As mentioned above, the extreme resistance to apoptosis conferred by the expression of BCR-ABL1 is largely due to its multi-layered modulation of BCL-2 family members [2,4–8]. BCR-ABL1 was shown to up-regulate the expression of the anti-apoptotic members BCL-2, BCL-XL, MCL-1 and A1 [6,10,24,25]. BCL-XL and MCL-1 up-regulation occurs downstream of the activation of STAT5 [10,26,27], whereas BCL-2 appears to rely on Ras pathway [28] and A1 is associated with NFκB activation [29]. Although overexpression of BCL-2 or BCL-XL was shown to confer resistance to imatinib [30], not a single anti-apoptotic member of the BCL-2 family seems to account for the extraordinary resistant phenotype observed in CML cells [8].

BCR-ABL1 also modulates pro-apoptotic BH3-only members of the Bcl-2 family, particularly BIM, BAD and BMF [31–34]. BCR-ABL1 was shown to interfere with the expression of BIM and BMF and post-translationally regulate BIM and BAD [32,33]. Whereas BMF seems to play a minor role on the BCR-ABL1-mediated resistance to apoptosis, BIM and BAD appears to cooperate and fully account for the anti-apoptotic phenotype, at least against TKIs, since imatinib does not induced apoptosis in BCR-ABL1-transformed bim−/−bad−/− cell lines [33]. Whether these double-knockout cells are also resistant to the full spectrum of chemotherapeutic drugs remains elusive. Interestingly enough, it was recently shown that BCR-ABL1 mediates the formation of a HSP70/BIM complex in a kinase-independent way [35,36]. This complex apparently contributes to BCR-ABL1-driven leukemogenesis via the stabilization and consequence up-regulation of AKT and eIF4E protein levels and inactivation of BAK [36]. Finally, BIM was shown to be epigenetically downmodulated in a set of patients that showed decreased cytogenetic and molecular responses to imatinib [37]. This observation indicates that combinations of TKIs and demethylating agents could improve the response of certain BIM-deficient patients.

The constitutive activity of the BCR-ABL1 oncoprotein is the trigger to the signaling cascades responsible for the extreme resistance to apoptosis found in CML cells. This understanding guided scientists to the design of specific TKIs that could stop this primary oncogenic signal and be used in clinic to treat CML patients. In short, TKIs are small molecules capable of interacting with BCR-ABL1 tyrosine kinase at the adenosine triphosphate (ATP) pocket, thereby obstructing its enzymatic activity [38,39].

Imatinib mesylate (IM) was the first TKI approved by the Food and Drug Administration (FDA) to treat CML patients in the chronic phase (CML-CP) [38]. In IRIS study (International Randomized Study of Interferon and STI571), IM induced complete cytogenetic remission in ∼74% of patients with CML-CP and complete hematologic response in 11% of patients with CML in blastic phase (CML-BP) [40,41]. Patients from the IRIS trial who achieved more than 3 log reduction in BCR-ABL1 transcripts levels had low risk for disease progression [42]. Despite the high rates of cytogenetic and molecular responses to IM, the emerging resistance to this drug has been recognized as a major problem in the treatment of Ph-positive leukemia, particularly for CML patients in accelerated and blastic phases [43,44]. Almost 17% of CML patients from the IRIS study developed IM resistance after five years of follow-up.

The second and third generation TKI (dasatinib, nilotinib, bosutinib, and ponatinib) have been developed for CML therapy as an attempt to overcome IM resistance and intolerance [45–49]. Although these TKIs provide a better rate of molecular remission, none of them can cure CML patients, prevent disease progression or overcome the patients’ resistance to therapy.

Before introduction of TKIs, CML patients diagnosed at chronic phase progressed to accelerated or blastic phases after three to five years [39]. This scenario changed significantly after introduction of TKIs as first-line therapy. However, still several patients develop resistance to TKIs leading to relapse of the disease and death of these patients. Therefore, a better knowledge about the mechanisms underlying CML resistance to TKIs is essential to design novel BCR-ABL1-dependent or -independent therapeutic strategies that can serve as more broad and effective therapy approaches.

The resistance to TKI treatment is defined as primary resistance — the absence of response to therapy at the very beginning of treatment; or acquired or secondary resistance — when patients’ leukemic cells develop molecular alterations that enable them to escape from ongoing therapy [50,51]. Importantly, the mechanisms involved in CML resistance to TKI therapy is divided into BCR-ABL1-dependent and -independent categories, as discussed below (Figure 1) [52,53].

BCR-ABL1-independent and -dependent mechanisms of resistance in chronic myeloid leukemia.

Figure 1.
BCR-ABL1-independent and -dependent mechanisms of resistance in chronic myeloid leukemia.

BCR-ABL1-dependent mechanisms are BCR-ABL1 duplication and overexpression, microRNAs deregulated expression, and Kinase-domain mutations. The BCR-ABL1-independent mechanisms are the chronic myeloid leukemic stem cell (CML-LSC) persistence and high stemness, the overexpression of ATP binding cassette transporters such as P-glycoprotein and ABCG2 efflux pumps, epigenetic deregulation (microRNAs, histonemarking systems and the polycomb repressive complex 1 and 2), oncoinflammatory bone marrow microenvironment, increased drug metabolism, and non-compliance to therapy (low drug adherence).

Figure 1.
BCR-ABL1-independent and -dependent mechanisms of resistance in chronic myeloid leukemia.

BCR-ABL1-dependent mechanisms are BCR-ABL1 duplication and overexpression, microRNAs deregulated expression, and Kinase-domain mutations. The BCR-ABL1-independent mechanisms are the chronic myeloid leukemic stem cell (CML-LSC) persistence and high stemness, the overexpression of ATP binding cassette transporters such as P-glycoprotein and ABCG2 efflux pumps, epigenetic deregulation (microRNAs, histonemarking systems and the polycomb repressive complex 1 and 2), oncoinflammatory bone marrow microenvironment, increased drug metabolism, and non-compliance to therapy (low drug adherence).

Close modal

BCR-ABL1-dependent mechanisms of resistance to TKIs

It is well-known that some BCR-ABL1-positive cells persist even in CML patients who respond to TKI therapy, indicating that these drugs do not eliminate all leukemic cells, particularly a BCR-ABL1-positive quiescent leukemic stem cells (LSC) population. Consequently, CML patients need to undergo continuous treatment with TKIs to keep the disease in check and to avoid progression to blastic phase. Unfortunately, it has been proved that the prolonged treatment with TKIs frequently leads to acquired resistance [54].

CML patients treated with IM may develop point mutations in the ABL1 kinase domain (KD) of BCR-ABL1 protein that reduces IM binding. Mutation in KD domain alters the protein structure and impairs the TKI and BCR-ABL1 interaction or stabilizes the kinase structure and reactivates the tyrosine kinase enzymatic activity [53,55]. These KD mutations are the most frequent mechanism of TKI resistance in CML patients (Figure 1), accounting for 9 to 48% of primary resistance and 10 to 68% of secondary resistance to IM [56–58]. Notably, CML patients at accelerated and blastic phases acquire KD mutations more frequently than CML-CP patients [59]. The KD mutations can be categorized into three types: (1) A-loop: mutation located at activation loop region that regulates the interaction with catalytic site; (2) P-loop: the ATP-binding loop, and (3) ATP-binding site [53,55].

Although Dasatinib, nilotinib, bosutinib, and ponatinib therapy have been successful for CML patients with the major KD mutations, these TKIs lack efficacy in a minimal number of different leukemic clones and all of them but ponatinib lack efficacy against the T315I mutation. The most frequent KD mutations associated with CML resistance to imatinib, nilotinib, dasatinib, bosutinib, and ponatinib according to their potency and region of BCR-ABL1 that each of these mutations target are shown at Figure 2 [52].

The most frequent KD mutations associated with CML resistance to imatinib, nilotinib, dasatinib, bosutinib, and ponatinib according to their potency and region of BCR-ABL1 that each of these mutations target.

Figure 2.
The most frequent KD mutations associated with CML resistance to imatinib, nilotinib, dasatinib, bosutinib, and ponatinib according to their potency and region of BCR-ABL1 that each of these mutations target.

Green: BCR-ABL1 KD mutation sensitive to TKI therapy; Yellow: BCR-ABL1 KD mutation resistant to TKI therapy; Orange: BCR-ABL1 KD mutation moderately resistant to TKI therapy; Red: BCR-ABL1 KD mutation highly resistant to TKI therapy (adapted from [52]).

Figure 2.
The most frequent KD mutations associated with CML resistance to imatinib, nilotinib, dasatinib, bosutinib, and ponatinib according to their potency and region of BCR-ABL1 that each of these mutations target.

Green: BCR-ABL1 KD mutation sensitive to TKI therapy; Yellow: BCR-ABL1 KD mutation resistant to TKI therapy; Orange: BCR-ABL1 KD mutation moderately resistant to TKI therapy; Red: BCR-ABL1 KD mutation highly resistant to TKI therapy (adapted from [52]).

Close modal

Additional BCR-ABL1-dependent mechanisms of resistance have been described, including BCR-ABL1 overexpression due to BCR-ABL1 gene amplification, Ph chromosome duplication, and differential regulation of oncogene transcription (Figure 1). BCR-ABL1 gene overexpression enables constitutive activation of tyrosine kinase despite the presence of TKI, which leads to leukemic cell expansion until a KD mutation is acquired and confers overt resistance [60]. As mentioned above, to overcome KD mutation, physicians should detect the type of mutation linked to a particular CML patient and select an adequate and effective new generation TKIs.

BCR-ABL1-independent mechanisms of resistance to TKIs

The BCR-ABL1-independent mechanisms of TKI resistance (Figures 1 and 3) in CML are particularly associated with persistence of LSC (Figure 3) [52,61,62], which may be related to their interactions with bone marrow niche microenvironment [61] as well as deregulated drug influx/efflux pumps [63,64], other chromosomal abnormalities, altered transcriptional regulatory and signaling networks or mitochondrial/metabolic changes [65], epigenetic alterations, and patient nonadherence to medication (Figures 1 and 3). In this regard, persistence of CML-LSC in the bone marrow have been described as the cause of the minimal residual disease during TKI treatment, which can also lead to TKI resistance over time and relapse after therapy discontinuation [66]. CML-LSC have high stemness potential, are resistant to TKI-induced-apoptosis, interact with bone marrow stromal cells, and express high levels of BCR-ABL1 and drug influx/efflux proteins.

Tyrosine kinase inhibitor-resistant leukemia stem cell.

Figure 3.
Tyrosine kinase inhibitor-resistant leukemia stem cell.

The chronic myeloid leukemia stem cell (CML-LSC) is resistant to TKI due to oncoinflammatory bone marrow niche, PI3-kinase and JAK/STAT signaling pathways; overexpression of anti-apoptotic-related genes (A); the low activity of the organic-cation transporter-1 (OCT-1) and overexpression of ATP binding cassette transporters such as P-glycoprotein and ABCG2 efflux pumps (B); BCR-ABL1 overexpression and aberrant activation Beta-catenin (C). Taken together all these alterations lead to CML-LSC exacerbated stemness, inhibition of apoptosis, genetic instability and TKI resistance.

Figure 3.
Tyrosine kinase inhibitor-resistant leukemia stem cell.

The chronic myeloid leukemia stem cell (CML-LSC) is resistant to TKI due to oncoinflammatory bone marrow niche, PI3-kinase and JAK/STAT signaling pathways; overexpression of anti-apoptotic-related genes (A); the low activity of the organic-cation transporter-1 (OCT-1) and overexpression of ATP binding cassette transporters such as P-glycoprotein and ABCG2 efflux pumps (B); BCR-ABL1 overexpression and aberrant activation Beta-catenin (C). Taken together all these alterations lead to CML-LSC exacerbated stemness, inhibition of apoptosis, genetic instability and TKI resistance.

Close modal

Deregulated drug influx/efflux pumps and drug sequestration are also related to TKI resistance because they interfere with intracellular drug availability. The low activity of the organic-cation transporter-1 (OCT-1), a cellular influx pump for IM, impairs the drug influx into leukemic cells. In contrast, elevated OCT-1 activity is associated with molecular response and high overall survival of IM-treated CML patients [63,64]. The high expression of the ATP-binding cassette (ABC) efflux transporters P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) reduces intracellular TKI accumulation and results in CML resistance to therapy [67,68]. The high expression of the influx transporter SLC22A1 is associated with a successful IM therapy in CML patients [67]. In addition, genetic polymorphisms of membrane transporters also contribute to CML resistance to IM. The SLCO1B3 699GG and 344TT genotypes are associated with lack of response to IM, while the ABCA3 4548-91 CC/CA genotypes are linked to poor prognosis in CML patients treated with IM standard dose [69]. Importantly, increase in TKI doses overcomes the low intracellular drug availability caused by alterations in drug influx/efflux pumps and sequestration processes.

The signals from the bone marrow (BM) microenvironment (BMM), especially from oncoinflammatory signals (pro-inflammatory cytokines and chemokines) and mesenchymal stromal cells also support LSC persistence. The alterations in CML-BMM, including the presence of high levels of inflammatory cytokines and chemokines (IL-1α, IL-1β, IL-6, G-CSF, TNF-α, CCL3, and CCL4), contribute to CML oncoinflammation, CML-LSC genetic instability, and to TKI resistance [70]. In a mice model, the reduction in Cxcl12, a chemokine secreted by mesenchymal stromal cells (MSC) leads to normal HSC numbers but promotes LSC expansion by increasing self-renewing cell divisions. On the other hand, the absence of endothelial cell-specific Cxcl12 reduces LSC proliferation, suggesting niche-specific effects in CML [71]. Activation of alternative pathways such as PI3K/AKT, WNT/β catenin and STAT provides several survival signals to CML-LSC in the bone marrow niche [53,72]. The up-regulation of PI3K/AKT signaling pathway is important for CML LSCs maintenance in bone marrow niche and seems to correlates with P210BCRABL1 transcripts levels [73]. Airiau et al. [74] also reported that PI3K/mTOR inhibitors is capable of sensitize CML LSC to nilotinib, a potent TKI. The STAT3 pathway seems to favor leukemic cells survival via intrinsic signaling and via bone marrow stromal cells modulation [53,75,76]. In CML LSC many survival genes (MCL-1, BCL-2 and BCL-XL) are up-regulated because of STAT5 increased activation [77] leading to apoptosis resistance.The WNT/β-catenin is relevant for CML LSC stemness and quiescence. CML animal model deficient in β-catenin present an impairment of CML LSC self-renewal potential [78]. In addition, the β-catenin overexpression is related to CML progression to advanced phase and TKI resistance [79] (Figure 3).

In another avenue, there is limited knowledge about the relationship between resistance to TKI and the metabolome of CML patients. Recently, the analysis of lipid metabolism (sphingolipids and eicosanoids) in CML showed that patients who are resistant to TKI exhibited higher ceramide and lower sphingomyelin levels compared with CML patients responsive to TKI therapy. The enzyme sirtuin 1 (SIRT1) deacetylase is overexpressed in CML CD34+ cell regulating their metabolism and leukemogenic potential. SIRT1 regulates many cellular pathways, including energy metabolism and stress response to tumorigenesis. Recently, this enzyme overexpression was associated with CML-LSC survival as well as for acquisition of genetic mutations that promote TKI resistance [80,81]. In addition, SIRT1 overexpression contributes to genetic mutation occurrence in CML leading to TKI resistance [81]. It was also reported that the SIRT1 substrate, the PGC-1α, contributed to oxidative phosphorylation and TKI resistance in CML by interfering in LSC mitochondrial respiration [81].

Furthermore, the deregulation of epigenetic intrinsic cell-regulatory mechanisms has been notably associated with TKI resistance and CML-LSC persistence after TKI treatment. The alteration in microRNAs, histonemarking systems and the polycomb repressive complex 1 and 2 is frequent in hematological malignancies.

Altered expression of microRNAs (miRs) is implicated in the acquisition of TKI resistance in CML patients. The miRs can not only regulate BCR-ABL1 expression but also modulate the expression of genes involved in apoptosis regulation (apoptomiRs), drug transport or activation of essential signaling pathways. Several authors have described the association between TKI resistance and miRs. For instance, the apoptomiRs miR-26a, miR-29c, miR-130b and miR-146a expression are down-regulated in IM-resistant CML patients while their predicted target genes C-IAP-1 and MCL-1 are overexpressed. Deregulation of these miRs and their predicted targets can be a mechanism of IM resistance, since these genes are anti-apoptotic and inhibit the TKI-induced BCR-ABL1-positive cell death [82].

The deregulation of epigenetic intrinsic cell-regulatory mechanisms has been also notably associated with the CML-LSC persistence after TKI treatment. For instance, the histone methyltransferase enhancer of zeste homolog 2 (EZH2) is overexpressed in CML-LSC and is associated with LSC protection from apoptosis and TKI-resistance [83]. Importantly, treatment with an EZH2-specific inhibitor resulted in increased sensitivity to dasatinib that is associated with up-regulated levels of p53 and the p53 targets BAX, BIM, PUMA and NOXA [83]. These results suggest that increased levels of EZH2, observed in CML-LSC, prevent p53 mediated intrinsic apoptosis. In addition, our group has previously shown that EZH2 also contributes to the resistance to TRAIL-mediated extrinsic apoptosis in CML cell lines [84,85]. Down-regulation of EZH2 by shRNA up-regulates TRAIL and sensitizes CML cell lines to imatinib (IM) [84]. Importantly, blockage of TRAIL signaling by treatment with TRAILR2-Fc prevented the combined effect of knocking-down of EZH2 and imatinib. Inhibition of TRAIL by EZH2 requires the expression of the tumor antigen preferentially expressed antigen of melanoma (PRAME) [84]. EZH2 forms a complex with PRAME to mediate epigenetic silencing of retinoic acid (RA)-target genes [86]. Indeed, we found that EZH2 is enriched at the retinoic acid responsive element (RARE) located at the TRAIL promoter in K562 cells and the down-regulation of PRAME resulted in significant loss of EZH2 from the TRAIL promoter [84]. Moreover, increased expression of PRAME, but not EZH2, was associated with the progression of disease in CML patients and knocking down of PRAME also sensitizes CML cell lines to chemotherapy, including IM [84,85].

Additionally, the overexpression of BMI1 a polycomb repressive complex 1 (PRC1) member is associated with CML progression, poor patient outcome and low response to TKI. In contrast, the increased expression of polycomb repressive complex 2 (PRC2) members, the PRC1-like and chromobox homolog 6 and 7 is linked to a better CML response to TKI [87–89].

Finally, drug adherence is one of most important steps for CML patients’ response to TKI therapy. Oral TKI therapy is a prolonged and a non-curative treatment for CML. Recent studies have reported that 20 to 30% of CML patients fail to adhere to TKI treatment due to lack of medication, forgetfulness, disbelief in the drug potential, drug side effects, and size and number of pills to take. The patients’ low compliance leads to low TKI plasma levels and suboptimal or no response to therapy. Pharmaceutical intervention showing the effectiveness of medication for disease control could improve the patients’ compliance. Therefore, description of the processes involved in BCR-ABL1-independent mechanisms of TKI resistance in CML may contribute to design new therapeutic strategies capable of circumventing TKI resistance and eliminating CML-LSC [39].

The discovery that anti-apoptotic BCL-2 family of proteins are implicated in the pathogenesis of several human cancers, including hematological malignancies such as CML [6,24,90], multiple myeloma [91,92]; and AML [93–95], let to the development of synthetic drugs that could inhibit prosurvival BCL-2 proteins. These drugs, so called ‘BH3-mimetics', mimic BH3-only proteins by antagonizing the pro-survival function of anti-apoptotic proteins, unleashing the mitochondrial apoptosis machinery to eliminate cancer cells [96]. After 30 years of numerous scientific efforts several BH3-mimetic drugs have reached clinical trials for hematopoietic malignancies. Of those, five are BCL-2 targeting drugs, including the Pan-BCL-2 inhibitors GX15-070 (obatoclax) [97] and AT-101 (Gossypol) [98], the BCL-2 specific drugs BCL201 [99] and ABT-199 (venetoclax) [100] and the BCL-2/BCL-XL/BCL-w inhibitor ABT-263 (navitoclax) [101].

The pharmaceutical companies AbbVie and Idun developed the first BH3-mimetic ABT-737 [102]. With high affinity for BCL-2, BCL-XL and BCL-W, this inhibitor was shown to kill primary cells from patients with B-cell malignancies [103] and, in preclinical studies, presented efficacy on eliminating small cell lung cancer cells (SCLC) and other blood cancers [104]. Unfortunately, ABT-737 was not bioavailable after oral administration, leading to the development of an orally available drug ABT-263 or Navitoclax [101]. In preclinical studies ABT-263 showed efficacy as both a single and combined treatment for Chronic Lymphocytic Leukemia (CLL) [105], SCLC [106,107], triple negative breast cancer [108], and several other cancers. However, in vivo preclinical studies revealed that inhibition of BCL-XL by ABT-737 and ABT-263 promoted cell death of platelets and immature B and T cells [109,110]. This cytotoxicity was confirmed in Phase I clinical trials where CLL and SCLC patients treated with ABT-263 developed thrombocytopenia [105]. These results limited the progression of navitoclax for the treatment of cancer. Importantly, the subsequent development of a highly specific BCL-2 inhibitor ABT-199 or Venetoclax, allows the elimination of cancer cells with less induced thrombocytopenia [100]. In clinical trials, Venetoclax was well-tolerated in chemotherapy refractory CLL patients [111]. These results led to the FDA approval of Venetoclax/ABT-199 in 2016, for the treatment of CLL and Small Lymphocytic Lymphoma (SLL).

More recently, in combination with other anti-cancer drugs or non-chemotherapies, ABT-199 has been approved for untreated CLL patients and newly diagnosed AML patients who are unable to receive intensive induction chemotherapy [112]. Venetoclax combined therapy in other cancers, including CML, has been explored and will be further discussed in this review. Despite the clinical success of ABT-199, recent studies show an intrinsic resistance to this drug in leukemia patients. Although Venetoclax resistance has been strongly linked to molecular deregulation of BCL-2 family of proteins including, increased expression of prosurvival proteins BCL-xL and MCL-1, acquired mutations and amplifications in BCL-2 protein and decrease in one or more pro-apoptotic proteins [113–115], recent studies have also shown that loss of TP53 and overactivation of signaling pathways such as FLT3 or Ras may also be important markers for venetoclax treatment response [116].

The evidence that MCL-1 is involved in disease relapse and that it has an important role in the growth and survival of malignant cells from several types of cancer [10,117–121], led to the generation of potent MCL-1 inhibitors. The MCL1 inhibitor S63845, from the pharmaceutical company Servier, was one of the first anti-MCL-1 drugs to provide insights into the efficacy of MCL-1 targeting for the elimination of cancer cells [122,123]. Anti-tumor activity of S63845 was reported in leukemia, multiple myeloma, lymphoma, SCLC, prostate and breast cancer [122,124–126]. To date, six MCL-1 inhibitors have entered phase 1 clinical trials S64315/MIK665 [127] (NCT02992483, NCT02979366, NCT03672695), AZD5991 [128] (NCT03218683, NCT03013998), ABBV-467 (NCT04178902), PRT1419 [129] (NCT05107856, NCT04837677, NCT04543305) and AMGEN MCL-1 inhibitors AMG-176 [130] (NCT 03797261, NCT05209152, NCT02675452) and AMG-397 [131] (NCT03465540). Despite the AMGEN compounds, all the other MCL-1 inhibitors are currently being evaluated for their safety, tolerability in Phase I clinical trials in recurrent hematologic malignancies including AML, multiple myeloma and lymphoma (NCT02992483; NCT02979366; NCT04178902 and NCT04543305). The MCL-1 inhibitors AMG-176 and AMG-397 were also in clinical trials for relapse or refractory MM or AML. However due to cardiotoxicity effects the trials have been suspended. Finally, preclinical and clinical studies combining two BH3-mimetics such as Venetoclax and MCL-1 inhibitors (NCT03672695; NCT03218683; NCT02675452) [132,133] may also provide further evidence on the most effective treatment for recurrent malignancies.

The discovery that CML cell survival and resistance to TKIs is associated with deregulation of pro and anti-apoptotic BCL-2 family of proteins such as BIM, BAD BCL-2, BCL-XL and MCL-1 [5,6,26,37,134,135], led to the investigation of BH3-mimetics for the treatment of CML.

The observation that overexpression of BCR-ABL in leukemia cell lines results in increased expression of BCL-XL and resistance to mitochondrial-mediated apoptosis [5,6], whereas inhibition of BCR-ABL kinase leads to down-regulation of BCL-XL and sensitization of CML chronic phase cells [26], suggest a strong involvement of BCL-XL in disease progression and a potential target for the treatment of CML. The BH3-mimetic ABT-737, was one of the first of its kind to be tested in BCR-ABL positive leukemias. The combination of ABT-737 with INNO-46, an BCR-ABL tyrosine kinase inhibitor (TKI), increased the expression of pro-apoptotic BH3-only proteins (Bim, Bad, Bmf and Bik) and sensitized Philadelphia-positive (Ph(+)) leukemias to intrinsic apoptosis [34]. Studies using ABT-263, another BCL-XL antagonist, have also shown that inhibition of BCL-XL combined with activation of BAD increased apoptosis in CML blast crisis cell lines [136].

Although BCL-XL appears to be the anti-apoptotic BCL-2 protein mostly affected by BCR-ABL, deregulation of anti-apoptotic protein BCL-2 have also been associated with CML TKI resistance [134,137,138]. BCR-ABL TKIs such as Imatinib and Dasatinib were shown to be efficient in inducing cell death of newly CML cells, they are inactive against quiescent CML stem cells, leading to disease relapse is a common feature once treatment has stopped [139,140]. When compared with normal hematopoietic cells, CML-LSC and blast crisis CML cells show increased expression of anti-apoptotic BCL-2 [134]. Inhibition of BCL-2 by sabutoclax, a pan-BCL-2 antagonist, sensitized CML-LSC to low doses of TKI, suggesting that the combination treatment of TKI plus BCL-2 inhibitors may eliminate CML-LSC, reduce disease relapse and prolong patients survival [134]. Furthermore, in vivo studies have shown that co-treatment of TKIs and the clinical BCL-2 inhibitor Venetoclax, reduced the engraftment capacity of CML-LSC and prolonged survival in a mice CML model [138]. Importantly, recent studies have also shown that CD26-target, immunoliposome loaded with Venetoclax, selective induced apoptosis of CML-LSCs sparing normal HSCs [141].

Whilst BCL-2/BCL-XL inhibitors provided promising results in inducing apoptosis in BCR-ABL(+) CML cell lines and primary cells and prolonged the survival of CML mouse models, their effectivity appear to be enhanced limited in leukemic cells harboring ABL kinase mutations such as T3151 [142]. This resistance could be overcome by addition of an MCL-1 inhibitor, suggesting that multi-targeting of BCL-2 proteins may be required for eradication of the disease. Indeed, a recent study shows that dual-inhibition of BCL-2 anti-apoptotic proteins dramatically increases the killing of CML cell lines and blast-phase CML primary cells [143]. The investigation of pan-BH3-mimetic in CML has also been extended to combined therapy with alternative chemotherapeutic agents such as Paclitaxel, used to overcome TKI primary and secondary resistance in CML [144]. While TKI resistance has been strongly linked to overexpression of BCL-2 and BCL-XL, studies using CML-derived cell line K562 show that Paclitaxel resistance is associated with overexpression of anti-apoptotic protein MCL-1. Treatment of TKI/Paclitaxel resistant CML cell lines with the pan-BH3-mimetic S1, a BCL-2/BCL-XL/MCL-1 inhibitor, could induce apoptosis in leukemic cells. These results once again suggest that the combination of TKI and inhibitors of BCL-2/BCL-XL/MCL-1 may provide a more effective therapy strategy for CML patients.

The strong evidence that BCL-2 family of proteins are associated with cell death resistance and poor response to TKI treatment, led to clinical trials combining BH3-mimetics and TKI inhibitors clinical trials. Four phase II trials combining Venetoclax with different TKI are in progress (Table 1). The comparison between Dasatinib single therapy and combination treatment Dasatinib/Venetoclax (NCT02689440) is being investigated for the treatment of BCR-ABL1-positive early chronic phase CML patients. The efficacy of venetoclax triple therapy Decitabine/ Venetoclax/ Ponatininb (NCT04188405) Azacitidine/Venetoclax/Flumatinib (NCT05433532) and Venetoclax/Ponatinib/Dexamethasone (NCT03576547) are being investigated for the treatment of Ph+ AML, accelerated phase or blast CML patients, and BCR-ABL1-positive relapse or refractory ALL and CML, respectively.

Table 1
Clinical trials involving BH3 mimetics for BCR-ABL1-positive leukemia
Clinical trialStudyStart dateTreatmentStatus
Phase 1 NCT00438178 Obatoclax mesylate, safety and efficacy in hematological Malignancies (AML, MDS, CML in myeloid blast phase, myelofibrosis, CLL and Aplastic Anemia) October 2005 Obatoclax Completed 
Phase 1 NCT00933985 Study of side effects and best dose of obatoclax in combination with chemotherapies for young patients with relapse/refractory solid tumors and leukemias, including blastic Phase and childhood CML June 2009 Obatoclax + Vincristine, Doxorubicin, Dexrazoxane Terminated 
Phase 2 NCT02115295 Cladribine plus Idarubicin plus cytarabine plus venetoclax in patients with blastic phase chronic myeloid leukemia (BCR-ABL1 positive) or AML or MDS. May 2014 Cladribine, Cytarabine, Gilteritinib, Idarubicin, Midostaurin, Venetoclax Recruiting 
Phase 2 NCT02689440 Dasatinib and Venetoclax in Treating Patients with Philadelphia Chromosome Positive or BCR-ABL1 Positive Early CML February 2016 Dasatinib, Venetoclax Recruiting 
Phase 1/2 NCT03576547 Venetoclax, Ponatinib, and Dexamethasone in Participants with Philadelphia Chromosome or BCR-ABL Positive Relapsed or Refractory ALL or CML July 2018 Dexamethasone, Ponatinib Hydrochloride, Venetoclax Active, not recruiting 
Phase 2 NCT04188405 Decitabine, Venetoclax, and Ponatinib for the Treatment of Philadelphia Chromosome-Positive AML or Myeloid Blast Phase or CML Accelerated Phase December 2019 Decitabine, Venetoclax, Ponatinib Recruiting 
Phase 2 NCT05433532 Azacitidine, Venetoclax and Flumatinib in newly diagnosed Ph-positive Acute Leukemia and CML accelerated phase or blast phase June 2022 Azacitidine, Venetoclax, Flumatinib Recruiting 
Clinical trialStudyStart dateTreatmentStatus
Phase 1 NCT00438178 Obatoclax mesylate, safety and efficacy in hematological Malignancies (AML, MDS, CML in myeloid blast phase, myelofibrosis, CLL and Aplastic Anemia) October 2005 Obatoclax Completed 
Phase 1 NCT00933985 Study of side effects and best dose of obatoclax in combination with chemotherapies for young patients with relapse/refractory solid tumors and leukemias, including blastic Phase and childhood CML June 2009 Obatoclax + Vincristine, Doxorubicin, Dexrazoxane Terminated 
Phase 2 NCT02115295 Cladribine plus Idarubicin plus cytarabine plus venetoclax in patients with blastic phase chronic myeloid leukemia (BCR-ABL1 positive) or AML or MDS. May 2014 Cladribine, Cytarabine, Gilteritinib, Idarubicin, Midostaurin, Venetoclax Recruiting 
Phase 2 NCT02689440 Dasatinib and Venetoclax in Treating Patients with Philadelphia Chromosome Positive or BCR-ABL1 Positive Early CML February 2016 Dasatinib, Venetoclax Recruiting 
Phase 1/2 NCT03576547 Venetoclax, Ponatinib, and Dexamethasone in Participants with Philadelphia Chromosome or BCR-ABL Positive Relapsed or Refractory ALL or CML July 2018 Dexamethasone, Ponatinib Hydrochloride, Venetoclax Active, not recruiting 
Phase 2 NCT04188405 Decitabine, Venetoclax, and Ponatinib for the Treatment of Philadelphia Chromosome-Positive AML or Myeloid Blast Phase or CML Accelerated Phase December 2019 Decitabine, Venetoclax, Ponatinib Recruiting 
Phase 2 NCT05433532 Azacitidine, Venetoclax and Flumatinib in newly diagnosed Ph-positive Acute Leukemia and CML accelerated phase or blast phase June 2022 Azacitidine, Venetoclax, Flumatinib Recruiting 

CML, chronic myeloid leukemia; CLL, chronic lymphoid leukemia; AML, acute myeloid leukemia; MDS, myelodysplastic syndromes; ALL, Acute Lymphoblastic leukemia.

Indubitably, the results obtained from these clinical studies will provide further and critical information about the efficacy of BH3-mimetics in CML. Notwithstanding, if the promising preclinical results are correct, is predicted that BH3-mimetic combination therapies will overcome TKI resistance, improve elimination of CML stem cells and prolong survival of CML patients (Figure 4).

Graphic overview of BCR-ABL1-induced resistance to cell death and the proposed effect of BH3 mimetics and TKI combined therapy for Chronic Myeloid Leukemia.

Figure 4.
Graphic overview of BCR-ABL1-induced resistance to cell death and the proposed effect of BH3 mimetics and TKI combined therapy for Chronic Myeloid Leukemia.

Expression of BCR-ABL1 induces a complex signaling cascade that confers resistance to apoptosis, including down-regulation of pro-apoptotic and up-regulation of anti-apoptotic Bcl-2 family members, which control the mitochondrial/intrinsic pathway of apoptosis. TKIs may stop signals emanating from BCR-ABL1 kinase but do not interfere with BCR-ABL1-independent resistance to cell death that emerges during the advanced phases of the disease. Combination of TKI and BH3 mimetics provides synergistic death-inducing signal that can be used to eliminate leukemic cells in CML patients.

Figure 4.
Graphic overview of BCR-ABL1-induced resistance to cell death and the proposed effect of BH3 mimetics and TKI combined therapy for Chronic Myeloid Leukemia.

Expression of BCR-ABL1 induces a complex signaling cascade that confers resistance to apoptosis, including down-regulation of pro-apoptotic and up-regulation of anti-apoptotic Bcl-2 family members, which control the mitochondrial/intrinsic pathway of apoptosis. TKIs may stop signals emanating from BCR-ABL1 kinase but do not interfere with BCR-ABL1-independent resistance to cell death that emerges during the advanced phases of the disease. Combination of TKI and BH3 mimetics provides synergistic death-inducing signal that can be used to eliminate leukemic cells in CML patients.

Close modal

The authors declare that there are no competing interests associated with the manuscript.

G.B. is supported by Cancer Australia and Leukaemia Foundation Australia Priority Grant PdCCRS 1162023). D.K. is supported by Walter and Eliza Hall Johnson PhD Scholarship and Melbourne Research Scholarship (University of Melbourne). F.A.C. is supported by the Brazilian Research Council (CNPq, Brazil). GPA-M is supported by grants from Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP, Brazil - 2015/09568-5; 2018/25395-1) and from CNPq (408909/2018-8, 303810/2018-1, 308927/2019-2). This work was also made possible through the Australian Cancer Research Foundation and Victorian State Government Operational Infrastructure Support and Australian Government NHMRC IRIISS (9000433).

Gustavo P. Amarante-Mendes: Conceptualization, Resources, Funding acquisition, Writing — original draft, Writing — review and editing. Gabriela Brumatti: Conceptualization, Resources, Funding acquisition, Writing — original draft, Writing — review and editing. Deeksha Kaloni: Writing — original draft, Writing — review and editing. Fabíola Attié Castro: Conceptualization, Resources, Funding acquisition, Writing — original draft, Writing — review and editing.

The authors are thankful to Felipe Campos Almeida and Gabriel Dessotti Barretto, from Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto for references organization and figure preparation, respectively.

BH

BCL-2 homology

BM

bone marrow

BMM

bone marrow microenvironment

CLL

chronic lymphocytic leukemia

CML

chronic myeloid leukemia

EZH2

enhancer of zeste homolog 2

FDA

Food and Drug Administration

IM

Imatinib mesylate

KD

kinase domain

LSC

leukemic stem cells

MOMP

mitochondrial outer membrane permeabilization

PRAME

preferentially expressed antigen of melanoma

PRC

polycomb repressive complex

SCLC

small cell lung cancer cells

SIRT1

sirtuin 1

TKIs

tyrosine kinase inhibitors

1
Nowell
,
P.C.
and
Hungerford
,
D.A.
(
1960
)
Chromosome studies on normal and leukemic human leukocytes
.
J. Natl Cancer Inst.
25
,
85
109
2
Bedi
,
A.
,
Zehnbauer
,
B.A.
,
Barber
,
J.P.
,
Sharkis
,
S.J.
and
Jones
,
R.J.
(
1994
)
Inhibition of apoptosis by BCR-ABL in chronic myeloid leukemia
.
Blood
83
,
2038
2044
3
Cortez
,
D.
,
Kadlec
,
L.
and
Pendergast
,
A.M.
(
1995
)
Structural and signaling requirements for BCR-ABL-mediated transformation and inhibition of apoptosis
.
Mol. Cell. Biol.
15
,
5531
5541
4
McGahon
,
A.J.
,
Brown
,
D.G.
,
Martin
,
S.J.
,
Amarante-Mendes
,
G.P.
,
Cotter
,
T.G.
,
Cohen
,
G.M.
et al (
1997
)
Downregulation of Bcr-Abl in K562 cells restores susceptibility to apoptosis: characterization of the apoptotic death
.
Cell Death Differ.
4
,
95
104
5
Amarante-Mendes
,
G.P.
,
Naekyung Kim
,
C.
,
Liu
,
L.
,
Huang
,
Y.
,
Perkins
,
C.L.
,
Green
,
D.R.
et al (
1998
)
Bcr-Abl exerts its antiapoptotic effect against diverse apoptotic stimuli through blockage of mitochondrial release of cytochrome C and activation of caspase-3
.
Blood
91
,
1700
1705
6
Amarante-Mendes
,
G.P.
,
McGahon
,
A.J.
,
Nishioka
,
W.K.
,
Afar
,
D.E.
,
Witte
,
O.N.
and
Green
,
D.R.
(
1998
)
Bcl-2-independent Bcr-Abl-mediated resistance to apoptosis: protection is correlated with up regulation of Bcl-xL
.
Oncogene
16
,
1383
1390
7
Fernandez-Luna
,
J.L.
(
2000
)
Bcr-Abl and inhibition of apoptosis in chronic myelogenous leukemia cells
.
Apoptosis
5
,
315
318
8
Brumatti
,
G.
,
Weinlich
,
R.
,
Chehab
,
C.F.
,
Yon
,
M.
and
Amarante-Mendes
,
G.P.
(
2003
)
Comparison of the anti-apoptotic effects of Bcr-Abl, Bcl-2 and Bcl-x(L) following diverse apoptogenic stimuli
.
FEBS Lett.
541
,
57
63
9
Amarante-Mendes
,
G.P.
,
Rana
,
A.
,
Datoguia
,
T.S.
,
Hamerschlak
,
N.
and
Brumatti
,
G.
(
2022
)
BCR-ABL1 tyrosine kinase complex signaling transduction: challenges to overcome resistance in chronic myeloid leukemia
.
Pharmaceutics
14
,
215
10
Aichberger
,
K.J.
,
Mayerhofer
,
M.
,
Krauth
,
M.-T.
,
Skvara
,
H.
,
Florian
,
S.
,
Sonneck
,
K.
et al (
2005
)
Identification of mcl-1 as a BCR/ABL-dependent target in chronic myeloid leukemia (CML): evidence for cooperative antileukemic effects of imatinib and mcl-1 antisense oligonucleotides
.
Blood
105
,
3303
3311
11
Amarante-Mendes
,
G.P.
and
Green
,
D.R.
(
1999
)
The regulation of apoptotic cell death
.
Braz. J. Med. Biol. Res.
32
,
1053
1061
12
Pereira
,
W.O.
and
Amarante-Mendes
,
G.P.
(
2011
)
Apoptosis: a programme of cell death or cell disposal?
Scand. J. Immunol.
73
,
401
407
13
Czabotar
,
P.E.
,
Lessene
,
G.
,
Strasser
,
A.
and
Adams
,
J.M.
(
2014
)
Control of apoptosis by the BCL-2 protein family: implications for physiology and therapy
.
Nat. Rev. Mol. Cell Biol.
15
,
49
63
14
Huang
,
D.C.
and
Strasser
,
A.
(
2000
)
BH3-Only proteins-essential initiators of apoptotic cell death
.
Cell
103
,
839
842
15
Puthalakath
,
H.
and
Strasser
,
A.
(
2002
)
Keeping killers on a tight leash: transcriptional and post-translational control of the pro-apoptotic activity of BH3-only proteins
.
Cell Death Differ.
9
,
505
512
16
Chen
,
L.
,
Willis
,
S.N.
,
Wei
,
A.
,
Smith
,
B.J.
,
Fletcher
,
J.I.
,
Hinds
,
M.G.
et al (
2005
)
Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function
.
Mol. Cell
17
,
393
403
17
Kuwana
,
T.
,
Bouchier-Hayes
,
L.
,
Chipuk
,
J.E.
,
Bonzon
,
C.
,
Sullivan
,
B.A.
,
Green
,
D.R.
et al (
2005
)
BH3 domains of BH3-only proteins differentially regulate Bax-mediated mitochondrial membrane permeabilization both directly and indirectly
.
Mol. Cell
17
,
525
535
18
Willis
,
S.N.
,
Chen
,
L.
,
Dewson
,
G.
,
Wei
,
A.
,
Naik
,
E.
,
Fletcher
,
J.I.
et al (
2005
)
Proapoptotic Bak is sequestered by Mcl-1 and Bcl-xL, but not Bcl-2, until displaced by BH3-only proteins
.
Genes Dev.
19
,
1294
1305
19
Cosentino
,
K.
and
García-Sáez
,
A.J.
(
2017
)
Bax and Bak pores: are we closing the circle?
Trends Cell Biol.
27
,
266
275
20
Green
,
D.R.
and
Amarante-Mendes
,
G.P.
(
1998
)
The point of no return: mitochondria, caspases, and the commitment to cell death
.
Results Probl. Cell Differ.
24
,
45
61
21
Amarante-Mendes
,
G.P.
,
Finucane
,
D.M.
,
Martin
,
S.J.
,
Cotter
,
T.G.
,
Salvesen
,
G.S.
and
Green
,
D.R.
(
1998
)
Anti-apoptotic oncogenes prevent caspase-dependent and independent commitment for cell death
.
Cell Death Differ.
5
,
298
306
22
Todt
,
F.
,
Cakir
,
Z.
,
Reichenbach
,
F.
,
Emschermann
,
F.
,
Lauterwasser
,
J.
,
Kaiser
,
A.
et al (
2015
)
Differential retrotranslocation of mitochondrial Bax and Bak
.
EMBO J.
34
,
67
80
23
Wolter
,
K.G.
,
Hsu
,
Y.T.
,
Smith
,
C.L.
,
Nechushtan
,
A.
,
Xi
,
X.G.
and
Youle
,
R.J.
(
1997
)
Movement of Bax from the cytosol to mitochondria during apoptosis
.
J. Cell Biol.
139
,
1281
1292
24
Sánchez-García
,
I.
and
Grütz
,
G.
(
1995
)
Tumorigenic activity of the BCR-ABL oncogenes is mediated by BCL2
.
Proc. Natl Acad. Sci. U.S.A.
92
,
5287
5291
25
Nieborowska-Skorska
,
M.
,
Hoser
,
G.
,
Kossev
,
P.
,
Wasik
,
M.A.
and
Skorski
,
T.
(
2002
)
Complementary functions of the antiapoptotic protein A1 and serine/threonine kinase pim-1 in the BCR/ABL-mediated leukemogenesis
.
Blood
99
,
4531
4539
26
Horita
,
M.
,
Andreu
,
E.J.
,
Benito
,
A.
,
Arbona
,
C.
,
Sanz
,
C.
,
Benet
,
I.
et al (
2000
)
Blockade of the Bcr-Abl kinase activity induces apoptosis of chronic myelogenous leukemia cells by suppressing signal transducer and activator of transcription 5-dependent expression of Bcl-xL
.
J. Exp. Med.
191
,
977
984
27
Gesbert
,
F.
and
Griffin
,
J.D.
(
2000
)
Bcr/Abl activates transcription of the Bcl-X gene through STAT5
.
Blood
96
,
2269
2276
28
Sánchez-García
,
I.
and
Martín-Zanca
,
D.
(
1997
)
Regulation of Bcl-2 gene expression by BCR-ABL is mediated by Ras
.
J. Mol. Biol.
267
,
225
228
29
Zong
,
W.X.
,
Edelstein
,
L.C.
,
Chen
,
C.
,
Bash
,
J.
and
Gélinas
,
C.
(
1999
)
The prosurvival Bcl-2 homolog Bfl-1/A1 is a direct transcriptional target of NF-kappaB that blocks TNFalpha-induced apoptosis
.
Genes Dev.
13
,
382
387
30
Ferrao
,
P.T.
,
Frost
,
M.J.
,
Siah
,
S.-P.
and
Ashman
,
L.K.
(
2003
)
Overexpression of P-glycoprotein in K562 cells does not confer resistance to the growth inhibitory effects of imatinib (STI571) in vitro
.
Blood
102
,
4499
4503
31
Neshat
,
M.S.
,
Raitano
,
A.B.
,
Wang
,
H.G.
,
Reed
,
J.C.
and
Sawyers
,
C.L.
(
2000
)
The survival function of the Bcr-Abl oncogene is mediated by Bad-dependent and -independent pathways: roles for phosphatidylinositol 3-kinase and Raf
.
Mol. Cell. Biol.
20
,
1179
1186
32
Kuribara
,
R.
,
Honda
,
H.
,
Matsui
,
H.
,
Shinjyo
,
T.
,
Inukai
,
T.
,
Sugita
,
K.
et al (
2004
)
Roles of Bim in apoptosis of normal and Bcr-Abl-expressing hematopoietic progenitors
.
Mol. Cell. Biol.
24
,
6172
6183
33
Kuroda
,
J.
,
Puthalakath
,
H.
,
Cragg
,
M.S.
,
Kelly
,
P.N.
,
Bouillet
,
P.
,
Huang
,
D.C.S.
et al (
2006
)
Bim and Bad mediate imatinib-induced killing of Bcr/Abl+ leukemic cells, and resistance due to their loss is overcome by a BH3 mimetic
.
Proc. Natl Acad. Sci. U.S.A.
103
,
14907
14912
34
Kuroda
,
J.
,
Kimura
,
S.
,
Strasser
,
A.
,
Andreeff
,
M.
,
O'Reilly
,
L.A.
,
Ashihara
,
E.
et al (
2007
)
Apoptosis-based dual molecular targeting by INNO-406, a second-generation Bcr-Abl inhibitor, and ABT-737, an inhibitor of antiapoptotic Bcl-2 proteins, against Bcr-Abl-positive leukemia
.
Cell Death Differ.
14
,
1667
1677
35
Guo
,
Z.
,
Song
,
T.
,
Wang
,
Z.
,
Lin
,
D.
,
Cao
,
K.
,
Liu
,
P.
et al (
2020
)
The chaperone Hsp70 is a BH3 receptor activated by the pro-apoptotic Bim to stabilize anti-apoptotic clients
.
J. Biol. Chem.
295
,
12900
9
36
Zhang
,
H.
,
Song
,
T.
,
Wang
,
Z.
,
Laura Bonnette
,
U.
,
Guo
,
Y.
,
Wang
,
H.
et al (
2022
)
Bcr-Abl drives the formation of Hsp70/Bim PPI to stabilize oncogenic clients and prevent cells from undergoing apoptosis
.
Biochem. Pharmacol.
198
,
114964
37
San José-Eneriz
,
E.
,
Agirre
,
X.
,
Jiménez-Velasco
,
A.
,
Cordeu
,
L.
,
Martín
,
V.
,
Arqueros
,
V.
et al (
2009
)
Epigenetic down-regulation of BIM expression is associated with reduced optimal responses to imatinib treatment in chronic myeloid leukaemia
.
Eur. J. Cancer
45
,
1877
1889
38
Druker
,
B.J.
and
Lydon
,
N.B.
(
2000
)
Lessons learned from the development of an abl tyrosine kinase inhibitor for chronic myelogenous leukemia
.
J. Clin. Invest.
105
,
3
7
39
Jabbour
,
E.
and
Kantarjian
,
H.
(
2018
)
Chronic myeloid leukemia: 2018 update on diagnosis, therapy and monitoring
.
Am. J. Hematol.
93
,
442
459
40
Kantarjian
,
H.
,
Sawyers
,
C.
,
Hochhaus
,
A.
,
Guilhot
,
F.
,
Schiffer
,
C.
,
Gambacorti-Passerini
,
C.
et al (
2002
)
Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia
.
N. Engl. J. Med.
346
,
645
652
41
Druker
,
B.J.
,
Talpaz
,
M.
,
Resta
,
D.J.
,
Peng
,
B.
,
Buchdunger
,
E.
,
Ford
,
J.M.
et al (
2001
)
Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia
.
N. Engl. J. Med.
344
,
1031
1037
42
Hughes
,
T.P.
,
Kaeda
,
J.
,
Branford
,
S.
,
Rudzki
,
Z.
,
Hochhaus
,
A.
,
Hensley
,
M.L.
et al (
2003
)
Frequency of major molecular responses to imatinib or interferon alfa plus cytarabine in newly diagnosed chronic myeloid leukemia
.
N. Engl. J. Med.
349
,
1423
1432
43
Tauchi
,
T.
and
Ohyashiki
,
K.
(
2004
)
Molecular mechanisms of resistance of leukemia to imatinib mesylate
.
Leuk. Res.
28
,
S39
S45
44
Yoshida
,
C.
and
Melo J
,
V.
(
2004
)
Biology of chronic myeloid leukemia and possible therapeutic approaches to imatinib-resistant disease
.
Int. J. Hematol.
79
,
420
433
45
Kantarjian
,
H.
,
Shah
,
N.P.
,
Hochhaus
,
A.
,
Cortes
,
J.
,
Shah
,
S.
,
Ayala
,
M.
et al (
2010
)
Dasatinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia
.
N. Engl. J. Med.
362
,
2260
2270
46
Kantarjian
,
H.M.
,
Hughes
,
T.P.
,
Larson
,
R.A.
,
Kim
,
D.-W.
,
Issaragrisil
,
S.
,
le Coutre
,
P.
et al (
2021
)
Long-term outcomes with frontline nilotinib versus imatinib in newly diagnosed chronic myeloid leukemia in chronic phase: ENESTnd 10-year analysis
.
Leukemia
35
,
440
453
47
Cortes
,
J.E.
,
Kantarjian
,
H.
,
Shah
,
N.P.
,
Bixby
,
D.
,
Mauro
,
M.J.
,
Flinn
,
I.
et al (
2012
)
Ponatinib in refractory Philadelphia chromosome-positive leukemias
.
N. Engl. J. Med.
367
,
2075
2088
48
Cortes
,
J.E.
,
Gambacorti-Passerini
,
C.
,
Deininger
,
M.W.
,
Mauro
,
M.J.
,
Chuah
,
C.
,
Kim
,
D.-W.
et al (
2018
)
Bosutinib versus imatinib for newly diagnosed chronic myeloid leukemia: results from the randomized BFORE trial
.
J. Clin. Oncol.
36
,
231
237
49
Yun
,
S.
,
Vincelette
,
N.D.
,
Segar
,
J.M.
,
Dong
,
Y.
,
Shen
,
Y.
,
Kim
,
D.-W.
et al (
2016
)
Comparative effectiveness of newer tyrosine kinase inhibitors versus imatinib in the first-line treatment of chronic-phase chronic myeloid leukemia across risk groups: a systematic review and meta-analysis of eight randomized trials
.
Clin. Lymphoma Myeloma Leuk.
16
,
e85
e94
50
Lovly
,
C.M.
and
Shaw
,
A.T.
(
2014
)
Molecular pathways: resistance to kinase inhibitors and implications for therapeutic strategies
.
Clin. Cancer Res.
20
,
2249
2256
51
Hochhaus
,
A.
(
2006
)
Chronic myelogenous leukemia (CML): resistance to tyrosine kinase inhibitors
.
Ann. Oncol.
17
,
x274
x279
52
Talati
,
C.
and
Pinilla-Ibarz
,
J.
(
2018
)
Resistance in chronic myeloid leukemia: definitions and novel therapeutic agents
.
Curr. Opin. Hematol.
25
,
154
161
53
Patel
,
A.B.
,
O'Hare
,
T.
and
Deininger
,
M.W.
(
2017
)
Mechanisms of resistance to ABL kinase inhibition in chronic myeloid leukemia and the development of next generation ABL kinase inhibitors
.
Hematol. Oncol. Clin. North Am.
31
,
589
612
54
O'Hare
,
T.
,
Zabriskie
,
M.S.
,
Eiring
,
A.M.
and
Deininger
,
M.W.
(
2012
)
Pushing the limits of targeted therapy in chronic myeloid leukaemia
.
Nat. Rev. Cancer
12
,
513
526
55
Shah
,
N.P.
,
Nicoll
,
J.M.
,
Nagar
,
B.
,
Gorre
,
M.E.
,
Paquette
,
R.L.
,
Kuriyan
,
J.
et al (
2002
)
Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia
.
Cancer Cell
2
,
117
125
56
Soverini
,
S.
,
Colarossi
,
S.
,
Gnani
,
A.
,
Rosti
,
G.
,
Castagnetti
,
F.
,
Poerio
,
A.
et al (
2006
)
Contribution of ABL kinase domain mutations to imatinib resistance in different subsets of Philadelphia-positive patients: by the GIMEMA Working Party on Chronic Myeloid Leukemia
.
Clin. Cancer Res.
12
,
7374
7379
57
Soverini
,
S.
,
Hochhaus
,
A.
,
Nicolini
,
F.E.
,
Gruber
,
F.
,
Lange
,
T.
,
Saglio
,
G.
et al (
2011
)
BCR-ABL kinase domain mutation analysis in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors: recommendations from an expert panel on behalf of European LeukemiaNet
.
Blood
118
,
1208
1215
58
Soverini
,
S.
,
Branford
,
S.
,
Nicolini
,
F.E.
,
Talpaz
,
M.
,
Deininger
,
M.W.N.
,
Martinelli
,
G.
et al (
2014
)
Implications of BCR-ABL1 kinase domain-mediated resistance in chronic myeloid leukemia
.
Leuk. Res
38
,
10
20
59
Branford
,
S.
,
Rudzki
,
Z.
,
Walsh
,
S.
,
Parkinson
,
I.
,
Grigg
,
A.
,
Szer
,
J.
et al (
2003
)
Detection of BCR-ABL mutations in patients with CML treated with imatinib is virtually always accompanied by clinical resistance, and mutations in the ATP phosphate-binding loop (P-loop) are associated with a poor prognosis
.
Blood
102
,
276
283
60
Barnes
,
D.J.
,
Palaiologou
,
D.
,
Panousopoulou
,
E.
,
Schultheis
,
B.
,
Yong
,
A.S.M.
,
Wong
,
A.
et al (
2005
)
Bcr-Abl expression levels determine the rate of development of resistance to imatinib mesylate in chronic myeloid leukemia
.
Cancer Res.
65
,
8912
8919
61
Shah
,
M.
and
Bhatia
,
R.
(
2018
)
Preservation of quiescent chronic myelogenous leukemia stem cells by the bone marrow microenvironment
.
Adv. Exp. Med. Biol.
1100
,
97
110
62
Neviani
,
P.
(
2014
)
Genetic events other than BCR-ABL1
.
Curr. Hematol. Malig. Rep.
9
,
24
32
63
White
,
D.L.
,
Saunders
,
V.A.
,
Dang
,
P.
,
Engler
,
J.
,
Venables
,
A.
,
Zrim
,
S.
et al (
2007
)
Most CML patients who have a suboptimal response to imatinib have low OCT-1 activity: higher doses of imatinib may overcome the negative impact of low OCT-1 activity
.
Blood
110
,
4064
4072
64
White
,
D.L.
,
Dang
,
P.
,
Engler
,
J.
,
Frede
,
A.
,
Zrim
,
S.
,
Osborn
,
M.
et al (
2010
)
Functional activity of the OCT-1 protein is predictive of long-term outcome in patients with chronic-phase chronic myeloid leukemia treated with imatinib
.
J. Clin. Oncol.
28
,
2761
2767
65
de Almeida
,
F.C.
,
Berzoti-Coelho
,
M.G.
,
Toro
,
D.M.
,
da Costa Cacemiro
,
M.
,
Bassan
,
V.L.
,
Barretto
,
G.D.
et al (
2022
)
Bioactive lipids as chronic myeloid leukemia's potential biomarkers for disease progression and response to tyrosine kinase inhibitors
.
Front. Immunol.
13
,
840173
66
Loscocco
,
F.
,
Visani
,
G.
,
Galimberti
,
S.
,
Curti
,
A.
and
Isidori
,
A.
(
2019
)
BCR-ABL independent mechanisms of resistance in chronic myeloid leukemia
.
Front. Oncol.
9
,
939
67
de Lima
,
L.T.
,
Vivona
,
D.
,
Bueno
,
C.T.
,
Hirata
,
R.D.C.
,
Hirata
,
M.H.
,
Luchessi
,
A.D.
et al (
2014
)
Reduced ABCG2 and increased SLC22A1 mRNA expression are associated with imatinib response in chronic myeloid leukemia
.
Med. Oncol.
31
,
851
68
Dohse
,
M.
,
Scharenberg
,
C.
,
Shukla
,
S.
,
Robey
,
R.W.
,
Volkmann
,
T.
,
Deeken
,
J.F.
et al (
2010
)
Comparison of ATP-binding cassette transporter interactions with the tyrosine kinase inhibitors imatinib, nilotinib, and dasatinib
.
Drug Metab. Dispos.
38
,
1371
1380
69
de Lima
,
L.T.
,
Bueno
,
C.T.
,
Vivona
,
D.
,
Hirata
,
R.D.C.
,
Hirata
,
M.H.
,
de Moraes Hungria
,
V.T.
et al (
2015
)
Relationship between SLCO1B3 and ABCA3 polymorphisms and imatinib response in chronic myeloid leukemia patients
.
Hematology
20
,
137
142
70
Zhang
,
B.
,
Ho
,
Y.W.
,
Huang
,
Q.
,
Maeda
,
T.
,
Lin
,
A.
,
Lee
,
S.-U.
et al (
2012
)
Altered microenvironmental regulation of leukemic and normal stem cells in chronic myelogenous leukemia
.
Cancer Cell
21
,
577
592
71
Agarwal
,
P.
,
Isringhausen
,
S.
,
Li
,
H.
,
Paterson
,
A.J.
,
He
,
J.
,
Gomariz
,
Á.
et al (
2019
)
Mesenchymal niche-specific expression of Cxcl12 controls quiescence of treatment-resistant leukemia stem cells
.
Cell Stem Cell
24
,
769
784.e6
72
Eiring
,
A.M.
,
Page
,
B.D.G.
,
Kraft
,
I.L.
,
Mason
,
C.C.
,
Vellore
,
N.A.
,
Resetca
,
D.
et al (
2015
)
Combined STAT3 and BCR-ABL1 inhibition induces synthetic lethality in therapy-resistant chronic myeloid leukemia
.
Leukemia
29
,
586
597
73
Diaz-Blanco
,
E.
,
Bruns
,
I.
,
Neumann
,
F.
,
Fischer
,
J.C.
,
Graef
,
T.
,
Rosskopf
,
M.
et al (
2007
)
Molecular signature of CD34(+) hematopoietic stem and progenitor cells of patients with CML in chronic phase
.
Leukemia
21
,
494
504
74
Airiau
,
K.
,
Mahon
,
F.-X.
,
Josselin
,
M.
,
Jeanneteau
,
M.
and
Belloc
,
F.
(
2013
)
PI3K/mTOR pathway inhibitors sensitize chronic myeloid leukemia stem cells to nilotinib and restore the response of progenitors to nilotinib in the presence of stem cell factor
.
Cell Death Dis.
4
,
e827
75
Bewry
,
N.N.
,
Nair
,
R.R.
,
Emmons
,
M.F.
,
Boulware
,
D.
,
Pinilla-Ibarz
,
J.
and
Hazlehurst
,
L.A.
(
2008
)
Stat3 contributes to resistance toward BCR-ABL inhibitors in a bone marrow microenvironment model of drug resistance
.
Mol. Cancer Ther.
7
,
3169
3175
76
Traer
,
E.
,
MacKenzie
,
R.
,
Snead
,
J.
,
Agarwal
,
A.
,
Eiring
,
A.M.
,
O'Hare
,
T.
et al (
2012
)
Blockade of JAK2-mediated extrinsic survival signals restores sensitivity of CML cells to ABL inhibitors
.
Leukemia
26
,
1140
1143
77
Bibi
,
S.
,
Arslanhan
,
M.D.
,
Langenfeld
,
F.
,
Jeanningros
,
S.
,
Cerny-Reiterer
,
S.
,
Hadzijusufovic
,
E.
et al (
2014
)
Co-operating STAT5 and AKT signaling pathways in chronic myeloid leukemia and mastocytosis: possible new targets of therapy
.
Haematologica
99
,
417
429
78
Zhao
,
C.
,
Blum
,
J.
,
Chen
,
A.
,
Kwon
,
H.Y.
,
Jung
,
S.H.
,
Cook
,
J.M.
et al (
2007
)
Loss of beta-catenin impairs the renewal of normal and CML stem cells in vivo
.
Cancer Cell
12
,
528
541
79
Jamieson
,
C.H.M.
,
Ailles
,
L.E.
,
Dylla
,
S.J.
,
Muijtjens
,
M.
,
Jones
,
C.
,
Zehnder
,
J.L.
et al (
2004
)
Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML
.
N. Engl. J. Med.
351
,
657
667
80
Li
,
L.
,
Wang
,
L.
,
Li
,
L.
,
Wang
,
Z.
,
Ho
,
Y.
,
McDonald
,
T.
et al (
2012
)
Activation of p53 by SIRT1 inhibition enhances elimination of CML leukemia stem cells in combination with imatinib
.
Cancer Cell
21
,
266
281
81
Wang
,
Z.
,
Yuan
,
H.
,
Roth
,
M.
,
Stark
,
J.M.
,
Bhatia
,
R.
and
Chen
,
W.Y.
(
2013
)
SIRT1 deacetylase promotes acquisition of genetic mutations for drug resistance in CML cells
.
Oncogene
32
,
589
598
82
Ferreira
,
A.F.
,
Moura
,
L.G.
,
Tojal
,
I.
,
Ambrósio
,
L.
,
Pinto-Simões
,
B.
,
Hamerschlak
,
N.
et al (
2014
)
Apoptomirs expression modulated by BCR-ABL is linked to CML progression and imatinib resistance
.
Blood Cells Mol. Dis.
53
,
47
55
83
Scott
,
M.T.
,
Korfi
,
K.
,
Saffrey
,
P.
,
Hopcroft
,
L.E.M.
,
Kinstrie
,
R.
,
Pellicano
,
F.
et al (
2016
)
Epigenetic reprogramming sensitizes CML stem cells to combined EZH2 and tyrosine kinase inhibition
.
Cancer Discov.
6
,
1248
1257
84
De Carvalho
,
D.D.
,
Binato
,
R.
,
Pereira
,
W.O.
,
Leroy
,
J.M.G.
,
Colassanti
,
M.D.
,
Proto-Siqueira
,
R.
et al (
2011
)
BCR-ABL-mediated upregulation of PRAME is responsible for knocking down TRAIL in CML patients
.
Oncogene
30
,
223
233
85
De Carvalho
,
D.D.
,
Mello
,
B.P.
,
Pereira
,
W.O.
and
Amarante-Mendes
,
G.P.
(
2013
)
PRAME/EZH2-mediated regulation of TRAIL: a new target for cancer therapy
.
Curr. Mol. Med.
13
,
296
304
86
Epping
,
M.T.
,
Wang
,
L.
,
Edel
,
M.J.
,
Carlée
,
L.
,
Hernandez
,
M.
and
Bernards
,
R.
(
2005
)
The human tumor antigen PRAME is a dominant repressor of retinoic acid receptor signaling
.
Cell
122
,
835
847
87
Mohty
,
M.
,
Yong
,
A.S.M.
,
Szydlo
,
R.M.
,
Apperley
,
J.F.
and
Melo J
,
V.
(
2007
)
The polycomb group BMI1 gene is a molecular marker for predicting prognosis of chronic myeloid leukemia
.
Blood
110
,
380
383
88
Crea
,
F.
,
Di Paolo
,
A.
,
Liu
,
H.H.
,
Polillo
,
M.
,
Clermont
,
P.-L.
,
Guerrini
,
F.
et al (
2015
)
Polycomb genes are associated with response to imatinib in chronic myeloid leukemia
.
Epigenomics
7
,
757
765
89
Galimberti
,
S.
,
Grassi
,
S.
,
Baratè
,
C.
,
Guerrini
,
F.
,
Ciabatti
,
E.
,
Perutelli
,
F.
et al (
2018
)
The polycomb BMI1 protein is co-expressed with CD26+ in leukemic stem cells of chronic myeloid leukemia
.
Front. Oncol.
8
,
555
90
Jaiswal
,
S.
,
Traver
,
D.
,
Miyamoto
,
T.
,
Akashi
,
K.
,
Lagasse
,
E.
and
Weissman
,
I.L.
(
2003
)
Expression of BCR/ABL and BCL-2 in myeloid progenitors leads to myeloid leukemias
.
Proc. Natl Acad. Sci. U.S.A.
100
,
10002
7
91
Morales
,
A.A.
,
Kurtoglu
,
M.
,
Matulis
,
S.M.
,
Liu
,
J.
,
Siefker
,
D.
,
Gutman
,
D.M.
et al (
2011
)
Distribution of Bim determines Mcl-1 dependence or codependence with Bcl-xL/Bcl-2 in Mcl-1-expressing myeloma cells
.
Blood
118
,
1329
1339
92
Touzeau
,
C.
,
Ryan
,
J.
,
Guerriero
,
J.
,
Moreau
,
P.
,
Chonghaile
,
T.N.
,
Le Gouill
,
S.
et al (
2016
)
BH3 profiling identifies heterogeneous dependency on Bcl-2 family members in multiple myeloma and predicts sensitivity to BH3 mimetics
.
Leukemia
30
,
761
764
93
Campos
,
L.
,
Rouault
,
J.P.
,
Sabido
,
O.
,
Oriol
,
P.
,
Roubi
,
N.
,
Vasselon
,
C.
et al (
1993
)
High expression of bcl-2 protein in acute myeloid leukemia cells is associated with poor response to chemotherapy
.
Blood
81
,
3091
3096
94
Kornblau
,
S.M.
,
Thall
,
P.F.
,
Estrov
,
Z.
,
Walterscheid
,
M.
,
Patel
,
S.
,
Theriault
,
A.
et al (
1999
)
The prognostic impact of BCL2 protein expression in acute myelogenous leukemia varies with cytogenetics
.
Clin. Cancer Res.
5
,
1758
1766
95
Brumatti
,
G.
,
Salmanidis
,
M.
,
Kok
,
C.H.
,
Bilardi
,
R.A.
,
Sandow
,
J.J.
,
Silke
,
N.
et al (
2013
)
Hoxa9 regulated Bcl-2 expression mediates survival of myeloid progenitors and the severity of HoxA9-dependent leukemia
.
Oncotarget
4
,
1933
1947
96
Lessene
,
G.
,
Czabotar
,
P.E.
and
Colman
,
P.M.
(
2008
)
BCL-2 family antagonists for cancer therapy
.
Nat. Rev. Drug Discov.
7
,
989
1000
97
Shore
,
G.C.
and
Viallet
,
J.
(
2005
)
Modulating the bcl-2 family of apoptosis suppressors for potential therapeutic benefit in cancer
.
Hematology Am. Soc. Hematol. Educ. Program
,
2005
,
226
230
98
Kitada
,
S.
,
Leone
,
M.
,
Sareth
,
S.
,
Zhai
,
D.
,
Reed
,
J.C.
and
Pellecchia
,
M.
(
2003
)
Discovery, characterization, and structure-activity relationships studies of proapoptotic polyphenols targeting B-cell lymphocyte/leukemia-2 proteins
.
J. Med. Chem.
46
,
4259
4264
99
Casara
,
P.
,
Davidson
,
J.
,
Claperon
,
A.
,
Le Toumelin-Braizat
,
G.
,
Vogler
,
M.
,
Bruno
,
A.
et al (
2018
)
S55746 is a novel orally active BCL-2 selective and potent inhibitor that impairs hematological tumor growth
.
Oncotarget
9
,
20075
20088
100
Souers
,
A.J.
,
Leverson
,
J.D.
,
Boghaert
,
E.R.
,
Ackler
,
S.L.
,
Catron
,
N.D.
,
Chen
,
J.
et al (
2013
)
ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets
.
Nat. Med.
19
,
202
208
101
Tse
,
C.
,
Shoemaker
,
A.R.
,
Adickes
,
J.
,
Anderson
,
M.G.
,
Chen
,
J.
,
Jin
,
S.
et al (
2008
)
ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor
.
Cancer Res.
68
,
3421
3428
102
Oltersdorf
,
T.
,
Elmore
,
S.W.
,
Shoemaker
,
A.R.
,
Armstrong
,
R.C.
,
Augeri
,
D.J.
,
Belli
,
B.A.
et al (
2005
)
An inhibitor of Bcl-2 family proteins induces regression of solid tumours
.
Nature
435
,
677
681
103
Vogler
,
M.
,
Dinsdale
,
D.
,
Sun
,
X.-M.
,
Young
,
K.W.
,
Butterworth
,
M.
,
Nicotera
,
P.
et al (
2008
)
A novel paradigm for rapid ABT-737-induced apoptosis involving outer mitochondrial membrane rupture in primary leukemia and lymphoma cells
.
Cell Death Differ.
15
,
820
830
104
Hann
,
C.L.
,
Daniel
,
V.C.
,
Sugar
,
E.A.
,
Dobromilskaya
,
I.
,
Murphy
,
S.C.
,
Cope
,
L.
et al (
2008
)
Therapeutic efficacy of ABT-737, a selective inhibitor of BCL-2, in small cell lung cancer
.
Cancer Res.
68
,
2321
2328
105
Roberts
,
A.W.
,
Seymour
,
J.F.
,
Brown
,
J.R.
,
Wierda
,
W.G.
,
Kipps
,
T.J.
,
Khaw
,
S.L.
et al (
2012
)
Substantial susceptibility of chronic lymphocytic leukemia to BCL2 inhibition: results of a phase I study of navitoclax in patients with relapsed or refractory disease
.
J. Clin. Oncol.
30
,
488
496
106
Faber
,
A.C.
,
Farago
,
A.F.
,
Costa
,
C.
,
Dastur
,
A.
,
Gomez-Caraballo
,
M.
,
Robbins
,
R.
et al (
2015
)
Assessment of ABT-263 activity across a cancer cell line collection leads to a potent combination therapy for small-cell lung cancer
.
Proc. Natl Acad. Sci. U.S.A.
112
,
E1288
E1296
107
Shoemaker
,
A.R.
,
Mitten
,
M.J.
,
Adickes
,
J.
,
Ackler
,
S.
,
Refici
,
M.
,
Ferguson
,
D.
et al (
2008
)
Activity of the Bcl-2 family inhibitor ABT-263 in a panel of small cell lung cancer xenograft models
.
Clin. Cancer Res.
14
,
3268
3277
108
Zoeller
,
J.J.
,
Vagodny
,
A.
,
Daniels
,
V.W.
,
Taneja
,
K.
,
Tan
,
B.Y.
,
DeRose
,
Y.S.
et al (
2020
)
Navitoclax enhances the effectiveness of EGFR-targeted antibody-drug conjugates in PDX models of EGFR-expressing triple-negative breast cancer
.
Breast Cancer Res.
22
,
132
109
Mérino
,
D.
,
Khaw
,
S.L.
,
Glaser
,
S.P.
,
Anderson
,
D.J.
,
Belmont
,
L.D.
,
Wong
,
C.
et al (
2012
)
Bcl-2, Bcl-x(L), and Bcl-w are not equivalent targets of ABT-737 and navitoclax (ABT-263) in lymphoid and leukemic cells
.
Blood
119
,
5807
5816
110
Mason
,
K.D.
,
Carpinelli
,
M.R.
,
Fletcher
,
J.I.
,
Collinge
,
J.E.
,
Hilton
,
A.A.
,
Ellis
,
S.
et al (
2007
)
Programmed anuclear cell death delimits platelet life span
.
Cell
128
,
1173
1186
111
Roberts
,
A.W.
,
Davids
,
M.S.
,
Pagel
,
J.M.
,
Kahl
,
B.S.
,
Puvvada
,
S.D.
,
Gerecitano
,
J.F.
et al (
2016
)
Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia
.
N. Engl. J. Med.
374
,
311
322
112
Parry
,
N.
,
Wheadon
,
H.
and
Copland
,
M.
(
2021
)
The application of BH3 mimetics in myeloid leukemias
.
Cell Death Dis.
12
,
222
113
Tahir
,
S.K.
,
Smith
,
M.L.
,
Hessler
,
P.
,
Rapp
,
L.R.
,
Idler
,
K.B.
,
Park
,
C.H.
et al (
2017
)
Potential mechanisms of resistance to venetoclax and strategies to circumvent it
.
BMC Cancer
17
,
399
114
Lin
,
K.H.
,
Winter
,
P.S.
,
Xie
,
A.
,
Roth
,
C.
,
Martz
,
C.A.
,
Stein
,
E.M.
et al (
2016
)
Targeting MCL-1/BCL-XL forestalls the acquisition of resistance to ABT-199 in acute myeloid leukemia
.
Sci. Rep.
6
,
27696
115
Chen
,
X.
,
Glytsou
,
C.
,
Zhou
,
H.
,
Narang
,
S.
,
Reyna
,
D.E.
,
Lopez
,
A.
et al (
2019
)
Targeting mitochondrial structure sensitizes acute myeloid leukemia to venetoclax treatment
.
Cancer Discov.
9
,
890
909
116
DiNardo
,
C.D.
,
Tiong
,
I.S.
,
Quaglieri
,
A.
,
MacRaild
,
S.
,
Loghavi
,
S.
,
Brown
,
F.C.
et al (
2020
)
Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML
.
Blood
135
,
791
803
117
Sieghart
,
W.
,
Losert
,
D.
,
Strommer
,
S.
,
Cejka
,
D.
,
Schmid
,
K.
,
Rasoul-Rockenschaub
,
S.
et al (
2006
)
Mcl-1 overexpression in hepatocellular carcinoma: a potential target for antisense therapy
.
J. Hepatol.
44
,
151
157
118
Li
,
Q.-F.
,
Huang
,
W.-R.
,
Duan
,
H.-F.
,
Wang
,
H.
,
Wu
,
C.-T.
and
Wang
,
L.-S.
(
2007
)
Sphingosine kinase-1 mediates BCR/ABL-induced upregulation of Mcl-1 in chronic myeloid leukemia cells
.
Oncogene
26
,
7904
7908
119
Glaser
,
S.P.
,
Lee
,
E.F.
,
Trounson
,
E.
,
Bouillet
,
P.
,
Wei
,
A.
,
Fairlie
,
W.D.
et al (
2012
)
Anti-apoptotic Mcl-1 is essential for the development and sustained growth of acute myeloid leukemia
.
Genes Dev.
26
,
120
125
120
Kelly
,
G.L.
,
Grabow
,
S.
,
Glaser
,
S.P.
,
Fitzsimmons
,
L.
,
Aubrey
,
B.J.
,
Okamoto
,
T.
et al (
2014
)
Targeting of MCL-1 kills MYC-driven mouse and human lymphomas even when they bear mutations in p53
.
Genes Dev.
28
,
58
70
121
Campbell
,
K.J.
,
Dhayade
,
S.
,
Ferrari
,
N.
,
Sims
,
A.H.
,
Johnson
,
E.
,
Mason
,
S.M.
et al (
2018
)
MCL-1 is a prognostic indicator and drug target in breast cancer
.
Cell Death Dis.
9
,
19
122
Kotschy
,
A.
,
Szlavik
,
Z.
,
Murray
,
J.
,
Davidson
,
J.
,
Maragno
,
A.L.
,
Le Toumelin-Braizat
,
G.
et al (
2016
)
The MCL1 inhibitor S63845 is tolerable and effective in diverse cancer models
.
Nature
538
,
477
482
123
Szlavik
,
Z.
,
Csekei
,
M.
,
Paczal
,
A.
,
Szabo
,
Z.B.
,
Sipos
,
S.
,
Radics
,
G.
et al (
2020
)
Discovery of S64315, a potent and selective Mcl-1 inhibitor
.
J. Med. Chem.
63
,
13762
13795
124
Merino
,
D.
,
Whittle
,
J.R.
,
Vaillant
,
F.
,
Serrano
,
A.
,
Gong
,
J.-N.
,
Giner
,
G.
et al (
2017
)
Synergistic action of the MCL-1 inhibitor S63845 with current therapies in preclinical models of triple-negative and HER2-amplified breast cancer
.
Sci. Transl. Med.
9
,
eaam7049
125
Arai
,
S.
,
Jonas
,
O.
,
Whitman
,
M.A.
,
Corey
,
E.
,
Balk
,
S.P.
and
Chen
,
S.
(
2018
)
Tyrosine kinase inhibitors increase MCL1 degradation and in combination with BCLXL/BCL2 inhibitors drive prostate cancer apoptosis
.
Clin. Cancer Res.
24
,
5458
5470
126
Yasuda
,
Y.
,
Ozasa
,
H.
,
Kim
,
Y.H.
,
Yamazoe
,
M.
,
Ajimizu
,
H.
,
Yamamoto Funazo
,
T.
et al (
2020
)
MCL1 inhibition is effective against a subset of small-cell lung cancer with high MCL1 and low BCL-X(L) expression
.
Cell Death Dis.
11
,
177
127
Mukherjee
,
N.
,
Skees
,
J.
,
Todd
,
K.J.
,
West
,
D.A.
,
Lambert
,
K.A.
,
Robinson
,
W.A.
et al (
2020
)
MCL1 inhibitors S63845/MIK665 plus navitoclax synergistically kill difficult-to-treat melanoma cells
.
Cell Death Dis.
11
,
443
128
Tron
,
A.E.
,
Belmonte
,
M.A.
,
Adam
,
A.
,
Aquila
,
B.M.
,
Boise
,
L.H.
,
Chiarparin
,
E.
et al (
2018
)
Discovery of Mcl-1-specific inhibitor AZD5991 and preclinical activity in multiple myeloma and acute myeloid leukemia
.
Nat. Commun.
9
,
5341
129
Bhagwat
,
N.
,
Grego
,
A.
,
Gowen-MacDonald
,
W.
,
Wang
,
M.
,
Cowart
,
M.
,
Wu
,
X.
et al (
2021
)
Abstract 983: preclinical characterization of PRT1419, a potent, selective and orally available inhibitor of MCL1
.
Cancer Res.
81
,
983
130
Caenepeel
,
S.
,
Brown
,
S.P.
,
Belmontes
,
B.
,
Moody
,
G.
,
Keegan
,
K.S.
,
Chui
,
D.
et al (
2018
)
AMG 176, a selective MCL1 inhibitor, is effective in hematologic cancer models alone and in combination with established therapies
.
Cancer Discov.
8
,
1582
1597
131
Caenepeel
,
S.
,
Karen
,
R.
,
Belmontes
,
B.
,
Verlinsky
,
A.
,
Tan
,
H.
,
Yang
,
Y.
et al (
2020
)
Abstract 6218: discovery and preclinical evaluation of AMG 397, a potent, selective and orally bioavailable MCL1 inhibitor
.
Cancer Res.
80
,
6218
132
Ramsey
,
H.E.
,
Fischer
,
M.A.
,
Lee
,
T.
,
Gorska
,
A.E.
,
Arrate
,
M.P.
,
Fuller
,
L.
et al (
2018
)
A novel MCL1 inhibitor combined with venetoclax rescues venetoclax-resistant acute myelogenous leukemia
.
Cancer Discov.
8
,
1566
1581
133
Hormi
,
M.
,
Birsen
,
R.
,
Belhadj
,
M.
,
Huynh
,
T.
,
Cantero Aguilar
,
L.
,
Grignano
,
E.
et al (
2020
)
Pairing MCL-1 inhibition with venetoclax improves therapeutic efficiency of BH3-mimetics in AML
.
Eur. J. Haematol.
105
,
588
596
134
Goff
,
D.J.
,
Court Recart
,
A.
,
Sadarangani
,
A.
,
Chun
,
H.-J.
,
Barrett
,
C.L.
,
Krajewska
,
M.
et al (
2013
)
A Pan-BCL2 inhibitor renders bone-marrow-resident human leukemia stem cells sensitive to tyrosine kinase inhibition
.
Cell Stem Cell
12
,
316
328
135
Ng
,
K.P.
,
Hillmer
,
A.M.
,
Chuah
,
C.T.H.
,
Juan
,
W.C.
,
Ko
,
T.K.
,
Teo
,
A.S.M.
et al (
2012
)
A common BIM deletion polymorphism mediates intrinsic resistance and inferior responses to tyrosine kinase inhibitors in cancer
.
Nat. Med
18
,
521
528
136
Harb
,
J.G.
,
Neviani
,
P.
,
Chyla
,
B.J.
,
Ellis
,
J.J.
,
Ferenchak
,
G.J.
,
Oaks
,
J.J.
et al (
2013
)
Bcl-xL anti-apoptotic network is dispensable for development and maintenance of CML but is required for disease progression where it represents a new therapeutic target
.
Leukemia
27
,
1996
2005
137
Korfi
,
K.
,
Mandal
,
A.
,
Furney
,
S.J.
,
Wiseman
,
D.
,
Somervaille
,
T.C.P.
and
Marais
,
R.
(
2015
)
A personalised medicine approach for ponatinib-resistant chronic myeloid leukaemia
.
Ann. Oncol.
26
,
1180
1187
138
Carter
,
B.Z.
,
Mak
,
P.Y.
,
Mu
,
H.
,
Zhou
,
H.
,
Mak
,
D.H.
,
Schober
,
W.
et al (
2016
)
Combined targeting of BCL-2 and BCR-ABL tyrosine kinase eradicates chronic myeloid leukemia stem cells
.
Sci. Transl. Med.
8
,
355ra117
139
Graham
,
S.M.
,
Jørgensen
,
H.G.
,
Allan
,
E.
,
Pearson
,
C.
,
Alcorn
,
M.J.
,
Richmond
,
L.
et al (
2002
)
Primitive, quiescent, Philadelphia-positive stem cells from patients with chronic myeloid leukemia are insensitive to STI571 in vitro
.
Blood
99
,
319
325
140
Copland
,
M.
,
Hamilton
,
A.
,
Elrick
,
L.J.
,
Baird
,
J.W.
,
Allan
,
E.K.
,
Jordanides
,
N.
et al (
2006
)
Dasatinib (BMS-354825) targets an earlier progenitor population than imatinib in primary CML but does not eliminate the quiescent fraction
.
Blood
107
,
4532
4539
141
Houshmand
,
M.
,
Garello
,
F.
,
Stefania
,
R.
,
Gaidano
,
V.
,
Cignetti
,
A.
,
Spinelli
,
M.
et al (
2021
)
Targeting chronic myeloid leukemia stem/progenitor cells using venetoclax-loaded immunoliposome
.
Cancers (Basel)
13
,
1311
142
Kuroda
,
J.
,
Kimura
,
S.
,
Andreeff
,
M.
,
Ashihara
,
E.
,
Kamitsuji
,
Y.
,
Yokota
,
A.
et al (
2008
)
ABT-737 is a useful component of combinatory chemotherapies for chronic myeloid leukaemias with diverse drug-resistance mechanisms
.
Br. J. Haematol.
140
,
181
190
143
Parry
,
N.
,
Busch
,
C.
,
Aßmann
,
V.
,
Cassels
,
J.
,
Hair
,
A.
,
Helgason
,
G.V.
et al (
2022
)
BH3 mimetics in combination with nilotinib or ponatinib represent a promising therapeutic strategy in blast phase chronic myeloid leukemia
.
Cell Death Discov.
8
,
457
144
Song
,
T.
,
Chai
,
G.
,
Liu
,
Y.
,
Xie
,
M.
,
Chen
,
Q.
,
Yu
,
X.
et al (
2015
)
Mechanism of synergy of BH3 mimetics and paclitaxel in chronic myeloid leukemia cells: Mcl-1 inhibition
.
Eur. J. Pharm. Sci.
70
,
64
71
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY-NC-ND).