Skip to main content

Progress in understanding the mechanisms of resistance to BCL-2 inhibitors

Abstract

Venetoclax is a new type of BH3 mimetic compound that can target the binding site in the BCL-2 protein and induce apoptosis in cancer cells by stimulating the mitochondrial apoptotic pathway. Venetoclax is especially used to treat haematological malignancies. However, with the recent expansion in the applications of venetoclax, some cases of venetoclax resistance have appeared, posing a major problem in clinical treatment. In this article, we explored several common mechanisms of venetoclax resistance. Increased expression of the antiapoptotic proteins MCL-1 and BCL-XL plays a key role in conferring cellular resistance to venetoclax. These proteins can bind to the released BIM in the context of venetoclax binding to BCL-2 and thus continue to inhibit mitochondrial apoptosis. Structural mutations in BCL-2 family proteins caused by genetic instability lead to decreased affinity for venetoclax and inhibit the intrinsic apoptosis pathway. Mutation or deletion of the BAX gene renders the BAX protein unable to anchor to the outer mitochondrial membrane to form pores. In addition to changes in BCL-2 family genes, mutations in other oncogenes can also confer resistance to apoptosis induced by venetoclax. TP53 mutations and the expansion of FLT3-ITD promote the expression of antiapoptotic proteins MCL-1 and BCL-XL through multiple signalling pathways, and interfere with venetoclax-mediated apoptosis processes depending on their affinity for BH3-only proteins. Finally, the level of mitochondrial oxidative phosphorylation in venetoclax-resistant leukaemia stem cells is highly abnormal. Not only the metabolic pathways but also the levels of important metabolic components are changed, and all of these alterations antagonize the venetoclax-mediated inhibition of energy metabolism and promote the survival and proliferation of leukaemia stem cells. In addition, venetoclax can change mitochondrial morphology independent of the BCL-2 protein family, leading to mitochondrial dysfunction. However, mitochondria resistant to venetoclax antagonize this effect, forming tighter mitochondrial cristae, which provide more energy for cell survival.

Introduction

The phenomenon of apoptosis resistance is an important indicator of the occurrence and development of haematological malignancies. Blocking apoptosis causes cancer cells to proliferate uncontrollably [1, 2]. BCL-2 family proteins play an important role in the mitochondria-mediated intrinsic apoptosis pathway. These proteins are a class of proteins with similar domains and are divided into 3 main categories: antiapoptotic proteins, proapoptotic proteins and regulatory proteins. The regulatory proteins contain only the BH3 domain and are thus called BH3-only proteins. If a BH3-only protein binds to the antiapoptotic protein BCL-2, BCL-2 can no longer bind to the proapoptotic proteins BAX/BAK, inhibiting their recruitment and thereby blocking the proapoptotic pathway. However, binding of a BH3-only protein to BAX/BAK can promote the recruitment and oligomerization of BAX/BAK, thereby resulting in the formation of pores on the outer mitochondrial membrane, which release cytochrome C, leading to proteolysis and apoptosis [3, 4]. Therefore, apoptosis and survival are balanced by regulating antiapoptotic proteins and proapoptotic proteins. Abnormal expression of BCL-2 family proteins is a common finding in haematological malignancies, the most important of which is overexpression of BCL-2. High levels of BCL-2 can be observed in patients with follicular lymphoma (FL), chronic lymphocytic leukaemia (CLL), mantle cell lymphoma (MCL) and Waldenström's macroglobulinaemia [5, 6]. Therefore, in BCL-2-dependent haematological malignancies, BCL-2 inhibitors can exert a targeted therapeutic effect and relieve apoptosis inhibition. Venetoclax is a new type of BH3 mimetic compound that can target BCL-2 and replace BIM or other regulatory proteins in binding to BCL-2. In this way, venetoclax promotes the release of these BH3-only proteins to activate BAX and BAK, promote their oligomerization and mediate apoptosis [7, 8]. A large number of clinical trials have shown that haematological malignancies are more sensitive to venetoclax than to conventional drugs and that venetoclax has a better therapeutic effect. Currently, the Food and Drug Administration (FDA) has approved venetoclax as monotherapy for the treatment of patients with CLL and small lymphocytic lymphoma (SLL) and in combination with other drugs for the treatment of patients with acute myeloid leukaemia (AML). Although venetoclax has great clinical application value as a new therapeutic drug, studies have shown that many patients still experience relapse in several months to several years after remission [9,10,11]. Therefore, further research on the specific resistance mechanisms of venetoclax is necessary [12]. In this article, we reviewed and summarized some of the known main mechanisms of resistance to venetoclax to provide a definitive theoretical basis for overcoming clinical venetoclax resistance and further combining it with other drugs (Fig. 1).

Fig. 1
figure 1

The mechanisms of resistance to BCL-2 inhibitors

Increased expression of other antiapoptotic proteins

MCL-1 and BCL-XL are antiapoptotic proteins in the BCL-2 family that play an important role in the survival of cancer cells. They are overexpressed in AML, non-Hodgkin's lymphoma (NHL), multiple myeloma (MM) and other cancers [13, 14]. Their mode of action is similar to that of the BCL-2 protein. They bind to BIM to prevent it from binding to BAX/BAK, inhibit the recruitment of BAX/BAK, and ultimately prevent the activation of the apoptotic pathway [8, 15]. ABT-737 is the first-in-class BH3 mimetic compound, and it promotes tumour cell apoptosis through interaction with BCL-2 family proteins. In tumours where the MCL-1 level is low and MCL-1 is absent or inactivated, ABT-737 has a significant effect as monotherapy, but ABT-737 monotherapy is not effective in tumours with high MCL-1 expression. After using RNA interference to downregulate MCL-1 in tumour cells, the sensitivity of the cells to ABT-737 was increased.[16,17,18,19] The structure of venetoclax is similar to that of ABT-737; thus, venetoclax resistance may also be related to the level of MCL-1 in tumour cells. Pei et al. retrospectively reviewed 100 newly diagnosed, previously untreated AML patients who received venetoclax combination treatment and found that primary AML cells with a monocyte phenotype were more likely to develop drug resistance than less differentiated AML cells [20]. Analysis of the expression of apoptotic genes showed that the expression rate of BCL-2 in primary AML cells with a monocyte phenotype was decreased, while that of MCL-1 was increased [21]. Moreover, Romain et al. found that the MCL-1 protein content in the venetoclax-resistant (VEN-R) OCI-Ly1 cell line was higher than that in venetoclax-sensitive OCI-Ly1 cells [22]. In addition, MCL-1 amplification and overexpression were observed in the tissues of patients treated with venetoclax [5]. The use of MCL-1 antagonists increases the sensitivity of cells to venetoclax, proving that MCL-1 plays an important role in venetoclax resistance. A variety of MCL-1 inhibitors have been developed and most of them have entered clinical trials, among which S63845, S64315, VU661013, AZD5991, AMG-176, AMG-397, A-1210477 target the MCL-1 protein for binding to dissociate BIM–MCL-1 complexes. After use in combination with venetoclax, not only do they convert leukaemia cells from resistant to sensitive to venetoclax, they also have a synergistic effect in T-cell acute lymphoblastic leukemia (ALL) and diffuse large B-cell lymphoma(DLBCL), recurrent MCL [23,24,25,26]. In addition to MCL-1 expression, BCL-XL expression is also increased in VEN-R cells [27,28,29]. The BCL-XL antagonist A-1155463 can also reverse resistance to venetoclax [30]. Additional studies have shown that the affinity of BIM and other antiapoptotic proteins and thus their importance in venetoclax resistance in CLL follow the order BCL-2 > BCL-XL > MCL-1 > BFL-1. Therefore, BCL-XL plays a greater role than MCL-1 in the drug resistance of tumour cells [27, 31,32,33]. Kallesh et al. found that for naïve Riva cells, the level of BCL-XL in Riva cells with acquired resistance to venetoclax was increased. However, the expression of MCL-1 did not differ greatly in these two cell lines [34].

The antiapoptotic proteins MCL-1 and BCL-XL confer resistance to venetoclax, and their expression is regulated by related signalling pathways in the cell. The PI3K/AKT/mTOR signalling pathway is an important signalling pathway that regulates cell proliferation, apoptosis and autophagy [35, 36]. AKT can promote the transcription, translation and posttranslational regulation of BCL-2 family proteins by regulating mTOR, GSK3, FOXO and NF-κB [28]. Indeed, the activity of AKT in the VEN-R OCI-Ly1 cell line and the VEN-R SU-DHL-6 cell line was increased to a certain extent compared to that in the corresponding control group. Cells receive signals by interacting with microenvironmental components. Agonists in the microenvironment, such as interleukin-10, CD40L and unmethylated DNA, can stimulate cells through the PI3K/AKT/mTOR signalling pathway to activate NF-κB signalling and promote the expression of MCL-1 and BCL-XL [7, 31, 37]. The effects of inhibitors targeting different components in the pathway also indirectly support this viewpoint. For example, the NF-κB signalling inhibitor BMS345541 can block the expression of MCL-1 and BCL-XL. After treatment with the PI3K, AKT or mTOR pathway inhibitors NVP-BEZ235 and GS-1101, cells resistant to venetoclax become sensitive. The PI3Kδ inhibitor GS-1101 can antagonize the activity of AKT, reduce the expression of antiapoptotic proteins and sensitize VEN-R cells [28, 31].

Genomic instability

BCL-2 mutations

Mutations in drug binding sites are a common mechanism by which tumour cells resist treatment. Many types of gene mutations have indeed been found in the laboratory and case studies of drug resistance to venetoclax; these mutations can inhibit apoptosis by reducing the affinity of the drug and its binding site or affecting the function of proapoptotic proteins [38]. Genetic testing was performed on 15 CLL patients who received venetoclax treatment and progressed. A new BCL-2 mutation, Gly101Val, was found in 7 patients [39]. This new type of mutation decreases the affinity of venetoclax for BCL-2 180-fold [40]. Further studies based on the molecular structure showed that the Gly101Val mutation is located in the BCL-2 α2 helix stacking against the α5 helix and is in the BCL-2 BH3 motif. In this mutant, the valine substituted for glycine is the only amino acid with a prominent large methyl group near the helical backbone of BCL-2, which can provide a stable extension of the helical structure and allow the helix to shift [40]. Therefore, the mutation in this region causes BCL-2 to retain the ability to bind to the BH3 motifs in the regulatory protein, affects the binding of venetoclax to its target site in BCL-2, and significantly reduces the affinity between the two, resulting in resistance to venetoclax. This mutation also prevents the regulatory protein from being released and binding to BAX/BAK, leading to inhibition of apoptosis [41].

With a mechanism similar to that of the BCL-2 Gly101Val mutation, the BCL-2 Phe104Ile mutation was found in a relapsed/refractory FL patient with decreased sensitivity to venetoclax. In this mutation, the phenylalanine at amino acid position 104 in the BCL-2 protein is replaced by isoleucine, which can decrease the affinity of venetoclax to BCL-2 to decrease approximately 40-fold [42]. In addition, two missense mutations F101C and F101L in the same codon in the BH3 domain of BCL-2 were detected in a murine human-like MCL cell line resistant to venetoclax. Both of these mutations prevent venetoclax from binding to the BH3 domain and inhibit mitochondrial apoptosis. In addition, the Phe104Cys and Phe104Leu mutations in BCL-2 also reduce the sensitivity of cells to venetoclax through a similar mechanism [39]. By analysing the genotypes of multiple VEN-R CLL patients, Blombery et al. observed recurrent mutations in the Asp103 codon, resulting in the presence of tyrosine, glutamic acid, and valine residues. However, the Asp103 residue in the P4 pocket is of great importance for the hydrogen binding of the azaindole moiety of venetoclax to BCL-2 protein [43]. Tausch et al. also observed the BCL-2 Asp103Tyr mutation and attached great importance to it in CLL patients with venetoclax resistance. Because this mutation, located in the pocket of BH3, it not only reduces the affinity for venetoclax but also greatly impacts the binding of other proteins. Compared with the abovementioned mutations, the BCL-2 Asp103Glu mutation can reduce the sensitivity of cells to venetoclax, but it does not change the sensitivity of cells to navitoclax, another BH3 mimetic compound. This is because this mutation changes the BCL-2 protein to make it more similar to the BCL-XL protein but does not affect the ability of BCL-2 to bind and release regulatory proteins [40]. Patients who are resistant to venetoclax can harbour a single BCL-2 mutation, but in most patients, two or more mutations are present. For example, patients with recurrent CLL acquire the BCL-2 Gly101Val and BCL-2 Asp103Glu mutations during venetoclax treatment, which seriously affects their prognosis [43].

BAX loss

The BAX and BAK genes are the executive genes of the cellular mitochondrial apoptosis pathway. The BAX and BAK proteins encoded by these genes are indispensable for the formation of mitochondrial membrane pores during apoptosis. The function of any BH3 mimic compound requires its downstream effectors BAX and BAK [16, 44]. Therefore, venetoclax-mediated apoptosis of cancer cells greatly depends on the mitochondrial membrane pores and the increased permeability of the outer mitochondrial membrane formed by these two proteins. In AML cells lacking the BAX gene, venetoclax largely fails to induce apoptosis, but venetoclax can trigger apoptosis in cells lacking BAK, indicating that BAX is an important mediator of venetoclax-induced apoptosis [45, 46]. Further studies showed that MV4-11 cells lacking BAX are resistant to venetoclax-mediated mitochondrial depolarization, mitochondrial outer membrane pore formation, cytochrome C release, caspase activation and apoptosis. Knockout of the BAX gene can block the venetoclax-dependent mitochondrial apoptosis pathway; thus, cells with BAX knockout also show resistance to venetoclax. Not only the loss of the BAX gene, but also the mutations of the BAX gene affect the sensitivity of cells to venetoclax [47]. 20 different BAX mutations have been detected in bone marrow or blood samples of VEN-R CLL patients, and more than one BAX mutation, including missense and frameshift/nonsense mutations, was detected in multiple patient samples. The location of the mutation is more important than the type of mutation. Mutations in the hydrophobic part of the C-terminal transmembrane domain of BAX affect the key α9-helix that targets BAX to the outer mitochondrial membrane. These mutations not only lead to a change in the number of α9 helices but also remove the critical terminal amino acids [48, 49]. As a result, BAX cannot anchor to mitochondria, thereby blocking venetoclax-induced apoptosis in vivo and in vitro [50, 51].

Changes in other cancer-related genes

TP53 is a highly conserved tumour suppressor gene located in the telomeric region of the short arm of chromosome 17. The encoded protein plays an important role in regulating apoptosis, ageing, DNA repair, autophagy and metabolism [52]. In addition, the mutations in TP53 are also related to disease progression, poor drug treatment effects, and poor prognosis in patients with MCL [53, 54], as well as to venetoclax resistance. Zhao et al. explored the genomic profile of MCL patients who progressed on venetoclax. They found that acquisition of BCL-2 mutations was infrequent; instead, acquisition of TP53 alterations played a role in disease progression on venetoclax. The frequency of TP53 alteration was increased > 2-fold in MCL patients before and after treatment with venetoclax [55]. In addition to MCL patients, TP53 mutations occur in AML patients during the acquisition of resistance to venetoclax [45, 56, 57]. A study by Tamilla et al. showed that compared with 282 wild-type AML patient samples, 16 patient samples with deleterious TP53 mutations showed reduced sensitivity to venetoclax. As TP53 is a gene that controls the expression of proapoptotic proteins, its inactivation or mutation decreases the expression of BCL-2 in AML cell lines. The lower the expression of TP53, the lower is the expression of BCL-2 [45]. Because venetoclax exerts an apoptosis-activating effect by binding to BCL-2, when the level of BCL-2 decreases, the sensitivity of cells to venetoclax decreases [45]. In addition to promoting the expression of the BCL-2 protein, TP53 can also change the level of MCL-1. Studies have shown that the total level of mitogen-activated protein kinase (MAPK) in TP53 mutant cells is increased. The MAPK signalling pathway is involved in the regulation of cell proliferation and apoptosis and can increase the expression of MCL-1 protein, thereby increasing the competitive binding of MCL-1 and BCL-2 to venetoclax and thus affecting venetoclax-mediated cell apoptosis [45, 58,59,60,61]. Further studies have shown that the expression level of BCL-XL was increased and the levels of PMAIP1 (NOXA), PUMA and BAK were decreased in a TP53 knockout cell line with reduced sensitivity to venetoclax, which caused inhibition of apoptosis. These cells were resistant to venetoclax-mediated apoptosis. Based on the results of a study indicating that one-third of patients with primary refractory AML resistant to venetoclax carry TP53 mutations, some researchers noted that the integrity of the TP53 gene could be checked to determine whether acquisition of venetoclax resistance was possible in patients with initial or remitted AML to facilitate the selection of a better treatment [62].

A study of the genome of CLL before treatment with venetoclax and after acquisition of venetoclax resistance revealed changes in cancer-related genes other than TP53, such as BRAF, CD274, NOTCH1, RB1, and SF3B1. In addition, mutations in BTG1 and homozygous deletions in CDKN2A/B were detected. These changes may be a major cause of resistance to venetoclax in CLL patients [63]. In AML patients, reconstructed existing mutations, such as expansion of FLT3-ITD, are main reasons for the poor therapeutic effect of venetoclax [40, 64, 65]. Zhang et al. even believed that compared to BCL-2 mutations, expansion of FLT3-ITD was the main contributor to venetoclax resistance in AML [64]. DiNardo et al. conducted a study with 81 AML patients who received venetoclax-based therapy and tracked the corresponding dynamic molecular changes. To identify dynamic molecular changes indicative of adaptive resistance to venetoclax, they compared the variant allele frequency of individual mutations at diagnosis, during remission, and at relapse to identify clones expanded at relapse. Their analysis revealed that relapse associated with progressive clonal expansion of FLT3-ITD was a feature in their study cohort. Expansion of FLT3-ITD can induce ligand-independent autophosphorylation and activation of receptors. Through the PI3K/AKT/mTOR and MEK/ERK pathways and various STAT5 downstream signalling events, the MCL-1 protein level is increased while the expression of BAD and BIM is suppressed to inhibit AML cell apoptosis [62, 66]. FLT3 inhibition by HQP1351 synergizes with BCL-2 inhibitor treatment to potentiate cellular apoptosis in FLT3-ITD mutant AML. HQP1351 inhibits the phosphorylation of FLT3 and its downstream signalling molecules, such as ERK1/2, AKT and STAT5, thus downregulating the antiapoptotic proteins MCL-1 and BCL-XL. In these AML cells, the increased cleavage of caspase 3 and poly(ADP-ribose) polymerase (PARP) prove the enhanced apoptosis. This finding also indirectly proves that expansion of FLT3-ITD is indeed involved in the resistance of cells to venetoclax [67].

Abnormal oxidative phosphorylation (OXPHOS)

Amino acid metabolism plays an important role in a variety of malignant tumours—for example, leukaemia stem cells (LSCs) cannot upregulate glycolysis and their survival is thus dependent on mitochondrial amino acid metabolism [68,69,70]. These cells rely on amino acid metabolism to provide energy for their survival. The combination of venetoclax and the hypomethylation agent azacitidine kills LSCs by decreasing amino acid uptake in these cells, resulting in decreased amino acid catabolism, inhibition of the mitochondrial electron transport chain and consequent inhibition of energy metabolism in LSCs. However, Stevens et al. found that LSCs used fatty acid metabolism to obviate the need for amino acid metabolism, which led to a significant reduction in their sensitivity to venetoclax. Analysis of bulk primary AML specimens revealed the link between mutations in RAS family genes and fatty acid metabolism. Carnitine synthesis, fatty acid metabolism, fatty acid extension in mitochondria, and β-oxidation of long-chain fatty acids in cells with RAS mutations were greatly increased, providing energy for the survival and growth of LSCs treated with venetoclax combinations [71,72,73]. In addition to the shift from amino acid metabolism to fatty acid metabolism, the components of metabolism were altered in drug-resistant AML LSCs compared with the initial AML LSCs; for example, the level of nicotinamide was increased. The increased level of nicotinamide resulted in increased production of nicotinamide adenine dinucleotide (NAD +), an essential coenzyme that is used in various enzymatic reactions and plays indispensable roles in energy metabolism. LSCs rely on increased NAD + to sustain OXPHOS by promoting the flux of amino acids and fatty acids into the tricarboxylic acid (TCA) cycle. Therefore, the inhibition of energy metabolism induced by venetoclax combination therapy is antagonized [74, 75]. Further studies on VEN-R OCI-Ly1 cells and cases of VEN-R CLL showed amplified chromosomal regions, which contained genes encoding the regulatory subunits of AMP-activated protein kinase (AMPK). As a classic energy sensor, AMPK can not only promote cellular respiration to produce ATP but also limit other physiological processes that consume ATP, maintaining the cell in a high-OXPHOS condition for a long time [76]. Activated AMPK can activate the PI3K/AKT signalling pathway [77,78,79]. Activation of the PI3K/AKT signalling pathway results in the phosphorylation of specific serine residues of the BAD protein, which binds to the 14–3-3 protein and is sequestered in the cytoplasm, preventing its transfer to mitochondria; in addition, the antiapoptotic proteins BCL-XL, BCL-2 and BCL-W dimerize, thereby blocking apoptosis [80,81,82]. In addition to the overexpression of AMPK itself, the expression level of its downstream target acetyl-CoA carboxylase is increased. After treatment with the AMPK activator A-769662, the cell line exhibited reduced sensitivity to venetoclax, and after treatment with the AMPK inhibitor dorsomorphin, it exhibited increased sensitivity to venetoclax. The abnormol intracellular OXPHOS caused by activation of the AMPK signalling pathway plays an important role in the decreased sensitivity of cells to venetoclax [83]. The increased MCL-1 in VEN-R AML cells also regulates pathways involved in bioenergetics and carbohydrate metabolism, including the TCA cycle, glycolysis and the pentose phosphate pathway, to change the OXPHOS level in the internal environment.[20, 84] Whether through amino acid metabolism or fatty acid metabolism, primary human LSCs rely on OXPHOS for energy, and in this cell population, OXPHOS at least partially depends on BCL-2 family proteins. Therefore, venetoclax, a highly selective BCL-2 inhibitor, displaces regulatory proteins from BCL-2, promoting oligomerization of BAX or BAK at the mitochondrial outer membrane to initiate apoptosis. In addition, studies have shown that venetoclax can affect cell respiration independently of the BCL-2 family. It can change the level of intracellular OXPHOS by changing the morphology of mitochondria and extensively inhibiting the function of the electron transport chain (complexes I, II, IV) [85, 86]. When venetoclax induces apoptosis in AML cells, hydrolysis of the mitochondrial protein optic atrophy 1 (OPA1) in AML cells increases. This protein plays an important role in maintaining mitochondrial cristae. A reduction in its expression leads to a decrease in the number and detrimental alterations in the morphology of mitochondrial cristae, collapse of the mitochondrial membrane potential and, ultimately, mitochondrial dysfunction. However, AML cells resistant to venetoclax can induce the expression of the OPA1 protein, making the mitochondrial cristae tighter and enriched in the metabolism of amino acids, coenzymes, and ATP [87]. More importantly, these metabolic effects of venetoclax have recently been shown to reciprocally regulate its efficacy [88]. Studies have noted that increased levels of OXPHOS and reactive oxygen species (ROS) confer chemotherapeutic resistance on cancer stem cells [89]. Compared with nonresistant cells, a tumour cell line resistant to venetoclax showed a higher maximum oxygen consumption rate and a higher steady-state level of ROS, and the activity of OXPHOS was indeed significantly increased [22]. Sharon et al. used CRISPR–Cas9 to screen VEN-R cell lines and identified genes associated with venetoclax resistance. These genes encode components of the mitochondrial translational machinery, such as mitochondrial ribosomal protein L54 (MRPL54), mitochondrial ribosomal protein L17 (MRPL17) and ribosome binding factor A (RBFA), which can regulate mitochondrial protein synthesis and alter mitochondrial metabolism. Their abnormal expression results in AML cells becoming resistant to venetoclax. On this basis, they found that the combination of venetoclax and tedizolid, a second-generation oxazolidinone-class antibiotic that blocks mitochondrial protein synthesis, can synergistically activate a heightened integrated stress response (ISR) without altering the expression of BCL-2 family proteins. ISR activation blocks glycolysis, inhibits OXPHOS activity, depletes ATP at the metabolic level, and leads to morphological effects of mitochondrial swelling and vacuolization. The efficacy of the combination regimen of venetoclax and tedizolid indirectly proves that mitochondrial ribosomal proteins play a role in the resistance of cells to venetoclax [90].

Treatment after venetoclax resistance

The imbalance between antiapoptotic proteins and proapoptotic proteins affects the survival of cells, and one of major mechanisms of resistance to venetoclax is the increase antiapoptotic proteins. We have mentioned that the combination of MCL-1 inhibitors, BCL-XL inhibitors and venetoclax is the most direct method to increase the sensitivity of cells to venetoclax. Based on this principle, BTK inhibitor (BTKi) is also a good choice [91, 92]. It can decrease the expression of MCL-1 protein and increase the expression of BIM protein, while it has no effect on BCL-2 protein level. Therefore, venetoclax and BTKis have a synergistic effect, enhancing the dependence of apoptosis on BCL-2 and increasing the affinity for venetoclax. Victor et al. evaluated the prognosis of 23 CLL patients who received a BTKi after stopping venetoclax due to disease progression [93]. Median progression-free survival and median overall survival were 34 and 42 months in these patients, they considered BTKi therapy to be an effective treatment option for VEN-R CLL patients. An international, multicenter, retrospective cohort study also showed that for VEN-R CLL patients, BTKi resulted in high response rates and durable remissions [94]. Ibrutinib plus venetoclax demonstrat promising efficacy and reliable safety in patients with relapsed/refractory MCL [95, 96]. In addition, DLBCL, FL and AML cells are highly sensitive to ibrutinib combined with venetoclax, suggesting that the combination of these two drugs could be an effective treatment modality for these patients [97, 98]. Besides,arsenic trioxide can lead to the phosphorylation of MCL-1 at Ser159 through PI3K/AKT pathway and attenuate MCL-1 phosphorylation at Thr163 through MEK/ERK pathway, triggering MCL-1 destabilisation and degradation. This causes the release of BIM bound to MCL-1, leading to apoptosis in VEN-R cells [99]. The anti-CD20 monoclonal antibodies (rituximab and obinutuzumab) also alter the expression of BCL-2 family proteins through a similar mechanism, reversing tumor cells resistance to venetoclax-induced apoptosis,making patients benefit from the venetoclax and anti-CD20 monoclonal antibodies combination [100,101,102].

In addition to affecting the balance between antiapoptotic and proapoptotic proteins, some drugs improve prognosis in drug-resistant patients by altering the level of mitochondrial OXPHOS. Patients undergoing treatment with venetoclax + azacitidine showed not only restricted uptake of amino acid, but also disruption of the TCA cycle and inhibition of electron transport chain complex II. [103, 104] This is a very promising treatment option and AML patients receiving this regimen have achieved high overall response rates in clinical trials [105, 106]. What’ s more,venetoclax combined with azacitidine or chemotherapy regimens are effective and safe in patients with refractory or relapsed acute leukemias of ambiguous lineage, not otherwise specified [107]. LSCs not only rely on amino acid metabolism for energy supply, but fatty acid metabolism plays an important role in these VEN-R cells. 8-chloro-adenosine (8-Cl-Ado), a novel nucleoside analog can interfere with fatty acid metabolism by downregulating important proteins involved in fatty acid oxidation, significantly reducing OXPHOS level in the internal environment, in this way to counteract cellular resistance to venetoclax [108]. In addition, acylchlorohydroquinone and tigecycline can also increase the sensitivity of cells to venetoclax by inhibiting mitochondrial OXPHOS or affecting mitochondrial function [109, 110]. Given the crucial role of mitochondria in cell death and metabolism, many mitochondria-targeted drugs are being studied in order to synergize with venetoclax in the treatment of haematological malignancies.

Conclusion

With the extensive application of venetoclax in haematological malignancies, cases of drug resistance to venetoclax have emerged in current clinical practice. The mechanisms of resistance to venetoclax include increased expression of other antiapoptotic proteins, genetic instability, and abnormal OXPHOS. The antiapoptotic proteins MCL-1 and BCL-XL, as regulatory factors, play a pivotal role in conferring resistance to venetoclax. Although venetoclax binds to BCL-2 and releases BIM, MCL-1 and BCL-XL can directly bind to BIM, which hinders its interaction with BAX/BAK and blocks apoptosis. Genetic instability is the most important reason for decreased sensitivity in patients treated with venetoclax, and mutation of BCL-2 has the most direct effect on this resistance. Although BCL-2 mutations are diverse, their effects are similar. They can reduce the affinity of the binding site for venetoclax and promote the tight binding of BCL-2 to regulatory proteins, making them unable to trigger the apoptotic response. Loss or mutation of the BAX gene also affects the venetoclax-mediated apoptosis process, especially mutations in the hydrophobic part of the C-terminal transmembrane domain, which directly prevents BAX from targeting the outer mitochondrial membrane to form pores. This effect ultimately leads to inhibition of the mitochondrial apoptotic pathway. In addition, mutations in other oncogenes play a regulatory role. In patients with MCL or AML, TP53 mutations induce cellular apoptosis in leukaemia by regulating the expression levels of BCL-2 family proteins through the MAPK signalling pathway. Moreover, in AML patients, reconstructed existing mutations, such as expansion of FLT3-ITD, promote the expression of the antiapoptotic protein MCL-1 and reduce the expression of proapoptotic proteins to interfere with intrinsic apoptosis pathways through the PI3K/AKT signalling pathway. Intrinsic apoptosis depends mainly on mitochondria (Fig. 2). In drug-resistant patients, mitochondria shift from relying on amino acid metabolism to relying on fatty acid metabolism. The level of the important metabolic component nicotinamide increases. These changes antagonize venetoclax-mediated inhibition of energy metabolism, resulting in increased cellular tolerance to venetoclax. Correspondingly, the internal high-OXPHOS environment changes the activity of BCL-2 family proteins and hinders the interactions between venetoclax and these proteins. In addition, venetoclax changes mitochondrial morphology and function alone, forming an internal environment with high metabolism and high OXPHOS levels, which blocks cancer cell apoptosis. Different haematological malignancies have different mechanisms of venetoclax resistance, but all usually involve an interaction of the multiple abovementioned mechanisms. NHL patients not only exhibit expression of antiapoptotic proteins but also show clonal heterogeneity. In addition, abnormalities in mitochondrial OXPHOS are more widespread in AML and CLL patients. We believe that a comprehensive understanding of the venetoclax resistance mechanisms of haematological malignancies will help patients to select appropriate follow-up therapy in clinic and maximize the prognosis of patients. Based on these resistance mechanisms, various clinical trials have been conducted in recent years to explore solutions. We summarize some promising regimens and we consider venetoclax-based combinations to be important therapeutic options for the treatment of haematological malignancies that can be dynamically and individually modified to achieve durable disease remission.

Fig. 2
figure 2

Abnormal oxidative phosphorylation in the mitochondria of VEN-R cells

Availability of data and materials

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

Abbreviations

FL:

Follicular lymphoma

CLL:

Chronic lymphocytic leukaemia

MCL:

Mantle cell lymphoma

FDA:

The Food and Drug Administration

SLL:

Small lymphocytic lymphoma

AML:

Acute myeloid leukaemia

NHL:

Non-Hodgkin's lymphoma

MM:

Multiple myeloma

VEN-R:

Venetoclax-resistant

ALL:

Acute lymphoblastic leukaemia

DLBCL:

Diffuse large B-cell lymphoma

MAPK:

Mitogen-activated protein kinase

PARP:

Poly(ADP-ribose) polymerase

OXPHOS:

Oxidative phosphorylation

LSCs:

Leukaemia stem cells

NAD + :

Nicotinamide adenine dinucleotide

TCA:

Tricarboxylic acid

AMPK:

AMP-activated protein kinase

OPA1:

Optic atrophy 1

ROS:

Reactive oxygen species

MRPL54:

Mitochondrial ribosomal protein L54,

MRPL17:

Mitochondrial ribosomal protein L17

RBFA:

Ribosome binding factor A

ISR:

Integrated stress response

BTKi:

BTK inhibitor

8-Cl-Ado:

8-Chloro-adenosine

References

  1. Monks NR, Biswas DK, Pardee AB. Blocking anti-apoptosis as a strategy for cancer chemotherapy: NF-kappaB as a target. J Cell Biochem. 2004;92(4):646–50.

    Article  CAS  PubMed  Google Scholar 

  2. Luo Y, Fu X, Ru R, Han B, Zhang F, Yuan L, et al. CpG Oligodeoxynucleotides Induces Apoptosis of Human Bladder Cancer Cells via Caspase-3-Bax/Bcl-2-p53 Axis. Arch Med Res. 2020;51(3):233–44.

    Article  CAS  PubMed  Google Scholar 

  3. Guerra VA, DiNardo C, Konopleva M. Venetoclax-based therapies for acute myeloid leukemia. Best Pract Res Clin Haematol. 2019;32(2):145–53.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Kale J, Osterlund EJ, Andrews DW. BCL-2 family proteins: changing partners in the dance towards death. Cell Death Differ. 2018;25(1):65–80.

    Article  CAS  PubMed  Google Scholar 

  5. Thijssen R, Roberts AW. Venetoclax in lymphoid malignancies: new insights more to learn. Cancer Cell. 2019;36(4):341–3.

    Article  CAS  PubMed  Google Scholar 

  6. Ruefli-Brasse A, Reed JC. Therapeutics targeting Bcl-2 in hematological malignancies. Biochem J. 2017;474(21):3643–57.

    Article  CAS  PubMed  Google Scholar 

  7. Ryan CE, Davids MS. BCL-2 inhibitors, present and future. Cancer J (Sudbury, Mass). 2019;25(6):401–9.

    Article  CAS  Google Scholar 

  8. Bose P, Gandhi V, Konopleva M. Pathways and mechanisms of venetoclax resistance. Leuk Lymphoma. 2017;58(9):1–17.

    Article  PubMed  CAS  Google Scholar 

  9. Lasica M, Anderson MA. Review of venetoclax in CLL, AML and multiple myeloma. J Pers Med. 2021;11(6):463.

    Article  PubMed  PubMed Central  Google Scholar 

  10. DiNardo CD, Jonas BA, Pullarkat V, Thirman MJ, Garcia JS, Wei AH, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2020;383(7):617–29.

    Article  CAS  PubMed  Google Scholar 

  11. Saliba AN, John AJ, Kaufmann SH. Resistance to venetoclax and hypomethylating agents in acute myeloid leukemia. Cancer Drug Resist (Alhambra, Calif). 2021;4:125–42.

    CAS  Google Scholar 

  12. Roberts AW, Ma S, Kipps TJ, Coutre SE, Davids MS, Eichhorst B, et al. Efficacy of venetoclax in relapsed chronic lymphocytic leukemia is influenced by disease and response variables. Blood. 2019;134(2):111–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ashkenazi A, Fairbrother WJ, Leverson JD, Souers AJ. From basic apoptosis discoveries to advanced selective BCL-2 family inhibitors. Nat Rev Drug Discov. 2017;16(4):273–84.

    Article  CAS  PubMed  Google Scholar 

  14. Agarwal R, Chan YC, Tam CS, Hunter T, Vassiliadis D, Teh CE, et al. Dynamic molecular monitoring reveals that SWI-SNF mutations mediate resistance to ibrutinib plus venetoclax in mantle cell lymphoma. Nat Med. 2019;25(1):119–29.

    Article  CAS  PubMed  Google Scholar 

  15. Cheng EH, Wei MC, Weiler S, Flavell RA, Mak TW, Lindsten T, et al. BCL-2, BCL-X(L) sequester BH3 domain-only molecules preventing BAX- and BAK-mediated mitochondrial apoptosis. Mol Cell. 2001;8(3):705–11.

    Article  CAS  PubMed  Google Scholar 

  16. van Delft MF, Wei AH, Mason KD, Vandenberg CJ, Chen L, Czabotar PE, et al. The BH3 mimetic ABT-737 targets selective Bcl-2 proteins and efficiently induces apoptosis via Bak/Bax if Mcl-1 is neutralized. Cancer Cell. 2006;10(5):389–99.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Konopleva M, Contractor R, Tsao T, Samudio I, Ruvolo PP, Kitada S, et al. Mechanisms of apoptosis sensitivity and resistance to the BH3 mimetic ABT-737 in acute myeloid leukemia. Cancer Cell. 2006;10(5):375–88.

    Article  CAS  PubMed  Google Scholar 

  18. Yecies D, Carlson NE, Deng J, Letai A. Acquired resistance to ABT-737 in lymphoma cells that up-regulate MCL-1 and BFL-1. Blood. 2010;115(16):3304–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Mazumder S, Choudhary GS, Al-Harbi S, Almasan A. Mcl-1 Phosphorylation defines ABT-737 resistance that can be overcome by increased NOXA expression in leukemic B cells. Cancer Res. 2012;72(12):3069–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Pei S, Pollyea DA, Gustafson A, Stevens BM, Minhajuddin M, Fu R, et al. Monocytic subclones confer resistance to venetoclax-based therapy in patients with acute myeloid leukemia. Cancer Discov. 2020;10(4):536–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kuusanmäki H, Leppä AM, Pölönen P, Kontro M, Dufva O, Deb D, et al. Phenotype-based drug screening reveals association between venetoclax response and differentiation stage in acute myeloid leukemia. Haematologica. 2020;105(3):708–20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Guieze R, Liu VM, Rosebrock D, Jourdain AA, Hernandez-Sanchez M, Martinez Zurita A, et al. Mitochondrial reprogramming underlies resistance to BCL-2 inhibition in lymphoid malignancies. Cancer Cell. 2019;36(4):369-84.e13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Li Z, He S, Look AT. The MCL1-specific inhibitor S63845 acts synergistically with venetoclax/ABT-199 to induce apoptosis in T-cell acute lymphoblastic leukemia cells. Leukemia. 2019;33(1):262–6.

    Article  PubMed  Google Scholar 

  24. Prukova D, Andera L, Nahacka Z, Karolova J, Svaton M, Klanova M, et al. Cotargeting of BCL2 with venetoclax and MCL1 with S63845 Is synthetically lethal in vivo in relapsed mantle cell lymphoma. Clin Cancer Res. 2019;25(14):4455–65.

    Article  CAS  PubMed  Google Scholar 

  25. Tron AE, Belmonte MA, Adam A, Aquila BM, Boise LH, Chiarparin E, et al. Discovery of Mcl-1-specific inhibitor AZD5991 and preclinical activity in multiple myeloma and acute myeloid leukemia. Nat Commun. 2018;9(1):5341.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang H, Guo M, Wei H, Chen Y. Targeting MCL-1 in cancer: current status and perspectives. J Hematol Oncol. 2021;14(1):67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Haselager MV, Kielbassa K, Ter Burg J, Bax DJC, Fernandes SM, Borst J, et al. Changes in Bcl-2 members after ibrutinib or venetoclax uncover functional hierarchy in determining resistance to venetoclax in CLL. Blood. 2020;136(25):2918–26.

    Article  PubMed  Google Scholar 

  28. Choudhary GS, Al-Harbi S, Mazumder S, Hill BT, Smith MR, Bodo J, et al. MCL-1 and BCL-xL-dependent resistance to the BCL-2 inhibitor ABT-199 can be overcome by preventing PI3K/AKT/mTOR activation in lymphoid malignancies. Cell Death Dis. 2015;6: e1593.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Chiron D, Dousset C, Brosseau C, Touzeau C, Maïga S, Moreau P, et al. Biological rational for sequential targeting of Bruton tyrosine kinase and Bcl-2 to overcome CD40-induced ABT-199 resistance in mantle cell lymphoma. Oncotarget. 2015;6(11):8750–9.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Tahir SK, Smith ML, Hessler P, Rapp LR, Idler KB, Park CH, et al. Potential mechanisms of resistance to venetoclax and strategies to circumvent it. BMC Cancer. 2017;17(1):399.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Jayappa KD, Portell CA, Gordon VL, Capaldo BJ, Bekiranov S, Axelrod MJ, et al. Microenvironmental agonists generate de novo phenotypic resistance to combined ibrutinib plus venetoclax in CLL and MCL. Blood Adv. 2017;1(14):933–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Punnoose EA, Leverson JD, Peale F, Boghaert ER, Belmont LD, Tan N, et al. Expression profile of BCL-2, BCL-XL, and MCL-1 predicts pharmacological response to the BCL-2 selective antagonist venetoclax in multiple myeloma models. Mol Cancer Ther. 2016;15(5):1132–44.

    Article  CAS  PubMed  Google Scholar 

  33. Wang Q, Wan J, Zhang W, Hao S. MCL-1 or BCL-xL-dependent resistance to the BCL-2 antagonist (ABT-199) can be overcome by specific inhibitor as single agents and in combination with ABT-199 in acute myeloid leukemia cells. Leuk Lymphoma. 2019;60(9):2170–80.

    Article  CAS  PubMed  Google Scholar 

  34. Kerkhofs M, Vervloessem T, Stopa KB, Smith VM, Vogler M, Bultynck G. DLBCL cells with acquired resistance to venetoclax are not sensitized to BIRD-2 but can be resensitized to venetoclax through Bcl-XL inhibition. Biomolecules. 2020;10(7):1081.

    Article  CAS  PubMed Central  Google Scholar 

  35. Klanova M, Andera L, Brazina J, Svadlenka J, Benesova S, Soukup J, et al. Targeting of BCL2 family proteins with ABT-199 and homoharringtonine reveals BCL2- and MCL1-dependent subgroups of diffuse large B-Cell lymphoma. Clin Cancer Res. 2016;22(5):1138–49.

    Article  CAS  PubMed  Google Scholar 

  36. Wang R, Song F, Li S, Wu B, Gu Y, Yuan Y. Salvianolic acid A attenuates CCl4-induced liver fibrosis by regulating the PI3K/AKT/mTOR, Bcl-2/Bax and caspase-3/cleaved caspase-3 signaling pathways. Drug Des Devel Ther. 2019;13:1889–900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Tromp JM, Tonino SH, Elias JA, Jaspers A, Luijks DM, Kater AP, et al. Dichotomy in NF-kappaB signaling and chemoresistance in immunoglobulin variable heavy-chain-mutated versus unmutated CLL cells upon CD40/TLR9 triggering. Oncogene. 2010;29(36):5071–82.

    Article  CAS  PubMed  Google Scholar 

  38. Shah NP, Nicoll JM, Nagar B, Gorre ME, Paquette RL, Kuriyan J, et al. 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. 2002;2(2):117–25.

    Article  CAS  PubMed  Google Scholar 

  39. Blombery P, Anderson MA, Gong JN, Thijssen R, Birkinshaw RW, Thompson ER, et al. Acquisition of the recurrent Gly101Val mutation in BCL2 confers resistance to venetoclax in patients with progressive chronic lymphocytic leukemia. Cancer Discov. 2019;9(3):342–53.

    Article  CAS  PubMed  Google Scholar 

  40. Tausch E, Close W, Dolnik A, Bloehdorn J, Chyla B, Bullinger L, et al. Venetoclax resistance and acquired BCL2 mutations in chronic lymphocytic leukemia. Haematologica. 2019;104(9):e434–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Birkinshaw RW, Gong JN, Luo CS, Lio D, White CA, Anderson MA, et al. Structures of BCL-2 in complex with venetoclax reveal the molecular basis of resistance mutations. Nat Commun. 2019;10(1):2385.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Blombery P, Birkinshaw RW, Nguyen T, Gong JN, Thompson ER, Xu Z, et al. Characterization of a novel venetoclax resistance mutation (BCL2 Phe104Ile) observed in follicular lymphoma. Br J Haematol. 2019;186(6):e188–91.

    Article  PubMed  Google Scholar 

  43. Blombery P, Thompson ER, Nguyen T, Birkinshaw RW, Gong JN, Chen X, et al. Multiple BCL2 mutations cooccurring with Gly101Val emerge in chronic lymphocytic leukemia progression on venetoclax. Blood. 2020;135(10):773–7.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Lindsten T, Ross AJ, King A, Zong WX, Rathmell JC, Shiels HA, et al. The combined functions of proapoptotic Bcl-2 family members bak and bax are essential for normal development of multiple tissues. Mol Cell. 2000;6(6):1389–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Nechiporuk T, Kurtz SE, Nikolova O, Liu T, Jones CL, D’Alessandro A, et al. The TP53 apoptotic network is a primary mediator of resistance to BCL2 inhibition in AML cells. Cancer Discov. 2019;9(7):910–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Rahmani M, Nkwocha J, Hawkins E, Pei X, Parker RE, Kmieciak M, et al. Cotargeting BCL-2 and PI3K Induces BAX-dependent mitochondrial apoptosis in AML cells. Cancer Res. 2018;78(11):3075–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Izzo F, Landau DA. A BAX door to venetoclax resistance. Blood. 2022;139(8):1124–6.

    Article  CAS  PubMed  Google Scholar 

  48. Wolter KG, Hsu YT, Smith CL, Nechushtan A, Xi XG, Youle RJ. Movement of Bax from the cytosol to mitochondria during apoptosis. J Cell Biol. 1997;139(5):1281–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zhou H, Hou Q, Hansen JL, Hsu YT. Complete activation of Bax by a single site mutation. Oncogene. 2007;26(50):7092–102.

    Article  CAS  PubMed  Google Scholar 

  50. Fresquet V, Rieger M, Carolis C, Garcia-Barchino MJ, Martinez-Climent JA. Acquired mutations in BCL2 family proteins conferring resistance to the BH3 mimetic ABT-199 in lymphoma. Blood. 2014;123(26):4111–9.

    Article  CAS  PubMed  Google Scholar 

  51. Blombery P, Lew TE, Dengler MA, Thompson ER, Lin VS, Chen X, et al. Clonal hematopoiesis, myeloid disorders and BAX-mutated myelopoiesis in patients receiving venetoclax for CLL. Blood. 2021. https://doi.org/10.1182/blood.2021012775.

    Article  PubMed  Google Scholar 

  52. Morabito F, Gentile M, Monti P, Recchia AG, Menichini P, Skafi M, et al. TP53 dysfunction in chronic lymphocytic leukemia: clinical relevance in the era of B-cell receptors and BCL-2 inhibitors. Expert Opin Investig Drugs. 2020;29(8):869–80.

    Article  CAS  PubMed  Google Scholar 

  53. Jain P, Wang M. Mantle cell lymphoma: 2019 update on the diagnosis, pathogenesis, prognostication, and management. Am J Hematol. 2019;94(6):710–25.

    Article  PubMed  Google Scholar 

  54. Ferrero S, Rossi D, Rinaldi A, Bruscaggin A, Spina V, Eskelund CW, et al. KMT2D mutations and TP53 disruptions are poor prognostic biomarkers in mantle cell lymphoma receiving high-dose therapy: a FIL study. Haematologica. 2020;105(6):1604–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zhao S, Kanagal-Shamanna R, Navsaria L, Ok CY, Zhang S, Nomie K, et al. Efficacy of venetoclax in high risk relapsed mantle cell lymphoma (MCL) - outcomes and mutation profile from venetoclax resistant MCL patients. Am J Hematol. 2020;95(6):623–9.

    Article  CAS  PubMed  Google Scholar 

  56. Kim K, Maiti A, Loghavi S, Pourebrahim R, Kadia TM, Rausch CR, et al. Outcomes of TP53-mutant acute myeloid leukemia with decitabine and venetoclax. Cancer. 2021. https://doi.org/10.1002/cncr.33689.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Venugopal S, Shoukier M, Konopleva M, Dinardo CD, Ravandi F, Short NJ, et al. Outcomes in patients with newly diagnosed TP53-mutated acute myeloid leukemia with or without venetoclax-based therapy. Cancer. 2021;127(19):3541–51.

    Article  CAS  PubMed  Google Scholar 

  58. Konopleva M, Milella M, Ruvolo P, Watts JC, Ricciardi MR, Korchin B, et al. MEK inhibition enhances ABT-737-induced leukemia cell apoptosis via prevention of ERK-activated MCL-1 induction and modulation of MCL-1/BIM complex. Leukemia. 2012;26(4):778–87.

    Article  CAS  PubMed  Google Scholar 

  59. Choi JH, Bogenberger JM, Tibes R. Targeting apoptosis in acute myeloid leukemia: current status and future directions of BCL-2 inhibition with venetoclax and beyond. Target Oncol. 2020;15(2):147–62.

    Article  PubMed  Google Scholar 

  60. Kawiak A, Domachowska A, Krolicka A, Smolarska M, Lojkowska E. 3-chloroplumbagin induces cell death in breast cancer cells through MAPK-Mediated Mcl-1 inhibition. Front Pharmacol. 2019;10:784.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Zhang Q, Riley-Gillis B, Han L, Jia Y, Lodi A, Zhang H, et al. Activation of RAS/MAPK pathway confers MCL-1 mediated acquired resistance to BCL-2 inhibitor venetoclax in acute myeloid leukemia. Signal Transduct Target Ther. 2022;7(1):51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. DiNardo CD, Tiong IS, Quaglieri A, MacRaild S, Loghavi S, Brown FC, et al. Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML. Blood. 2020;135(11):791–803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Herling CD, Abedpour N, Weiss J, Schmitt A, Jachimowicz RD, Merkel O, et al. Clonal dynamics towards the development of venetoclax resistance in chronic lymphocytic leukemia. Nat Commun. 2018;9(1):727.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Zhang X, Qian J, Wang H, Wang Y, Zhang Y, Qian P, et al. Not BCL2 mutation but dominant mutation conversation contributed to acquired venetoclax resistance in acute myeloid leukemia. Biomark Res. 2021;9(1):30.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Konopleva MY. Mechanisms for resistance in AML insights into molecular pathways mediating resistance to venetoclax. Best Pract Res Clin Haematol. 2021;34(1): 101251.

    Article  CAS  PubMed  Google Scholar 

  66. Watanabe D, Nogami A, Okada K, Akiyama H, Umezawa Y, Miura O. FLT3-ITD activates RSK1 to enhance proliferation and survival of AML cells by activating mTORC1 and eIF4B cooperatively with PIM or PI3K and by inhibiting Bad and BIM. Cancers (Basel). 2019;11(12):1827.

    Article  CAS  Google Scholar 

  67. Fang DD, Zhu H, Tang Q, Wang G, Min P, Wang Q, et al. FLT3 inhibition by olverembatinib (HQP1351) downregulates MCL-1 and synergizes with BCL-2 inhibitor lisaftoclax (APG-2575) in preclinical models of FLT3-ITD mutant acute myeloid leukemia. Transl Oncol. 2022;15(1): 101244.

    Article  PubMed  Google Scholar 

  68. Cramer SL, Saha A, Liu J, Tadi S, Tiziani S, Yan W, et al. Systemic depletion of L-cyst(e)ine with cyst(e)inase increases reactive oxygen species and suppresses tumor growth. Nat Med. 2017;23(1):120–7.

    Article  CAS  PubMed  Google Scholar 

  69. Raffel S, Falcone M, Kneisel N, Hansson J, Wang W, Lutz C, et al. BCAT1 restricts αKG levels in AML stem cells leading to IDHmut-like DNA hypermethylation. Nature. 2017;551(7680):384–8.

    Article  CAS  PubMed  Google Scholar 

  70. El Sayed R, Haibe Y, Amhaz G, Bouferraa Y, Shamseddine A. Metabolic factors affecting tumor immunogenicity: what is happening at the cellular level? Int J Mol Sci. 2021. https://doi.org/10.3390/ijms22042142.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Stevens BM, Jones CL, Pollyea DA, Culp-Hill R, D’Alessandro A, Winters A, et al. Fatty acid metabolism underlies venetoclax resistance in acute myeloid leukemia stem cells. Nat Cancer. 2020;1(12):1176–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Konoplev S, Wang X, Tang G, Li S, Wang W, Xu J, et al. Comprehensive immunophenotypic study of acute myeloid leukemia with KMT2A (MLL) rearrangement in adults: a single-institution experience. Cytometry B Clin Cytom. 2021. https://doi.org/10.1002/cyto.b.22051.

    Article  PubMed  Google Scholar 

  73. Gouw AM, Eberlin LS, Margulis K, Sullivan DK, Toal GG, Tong L, et al. Oncogene KRAS activates fatty acid synthase, resulting in specific ERK and lipid signatures associated with lung adenocarcinoma. Proc Natl Acad Sci USA. 2017;114(17):4300–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Jones CL, Stevens BM, Pollyea DA, Culp-Hill R, Reisz JA, Nemkov T, et al. Nicotinamide metabolism mediates resistance to venetoclax in relapsed acute myeloid leukemia stem cells. Cell Stem Cell. 2020;27(5):748-64.e4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Chiarugi A, Dölle C, Felici R, Ziegler M. The NAD metabolome–a key determinant of cancer cell biology. Nat Rev Cancer. 2012;12(11):741–52.

    Article  CAS  PubMed  Google Scholar 

  76. Cai Z, Li CF, Han F, Liu C, Zhang A, Hsu CC, et al. Phosphorylation of PDHA by AMPK drives TCA cycle to promote cancer metastasis. Mol Cell. 2020;80(2):263-78.e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Zheng T, Yang X, Wu D, Xing S, Bian F, Li W, et al. Salidroside ameliorates insulin resistance through activation of a mitochondria-associated AMPK/PI3K/Akt/GSK3beta pathway. Br J Pharmacol. 2015;172(13):3284–301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Tzatsos A, Kandror KV. Nutrients suppress phosphatidylinositol 3-kinase/Akt signaling via raptor-dependent mTOR-mediated insulin receptor substrate 1 phosphorylation. Mol Cell Biol. 2006;26(1):63–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Chopra I, Li HF, Wang H, Webster KA. Phosphorylation of the insulin receptor by AMP-activated protein kinase (AMPK) promotes ligand-independent activation of the insulin signalling pathway in rodent muscle. Diabetologia. 2012;55(3):783–94.

    Article  CAS  PubMed  Google Scholar 

  80. She QB, Solit DB, Ye Q, O’Reilly KE, Lobo J, Rosen N. The BAD protein integrates survival signaling by EGFR/MAPK and PI3K/Akt kinase pathways in PTEN-deficient tumor cells. Cancer Cell. 2005;8(4):287–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Yang E, Zha J, Jockel J, Boise LH, Thompson CB, Korsmeyer SJ. Bad, a heterodimeric partner for Bcl-XL and Bcl-2, displaces Bax and promotes cell death. Cell. 1995;80(2):285–91.

    Article  CAS  PubMed  Google Scholar 

  82. Lindsay J, Esposti MD, Gilmore AP. Bcl-2 proteins and mitochondria–specificity in membrane targeting for death. Biochim Biophys Acta. 2011;1813(4):532–9.

    Article  CAS  PubMed  Google Scholar 

  83. Alkhatabi HA, Zohny SF, Shait Mohammed MR, Choudhry H, Rehan M, Ahmad A, et al. Venetoclax-resistant MV4-11 leukemic cells activate PI3K/AKT pathway for metabolic reprogramming and redox adaptation for survival. Antioxidants. 2022. https://doi.org/10.3390/antiox11030461.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Carter BZ, Mak PY, Tao W, Warmoes M, Lorenzi PL, Mak D, et al. Targeting MCL-1 dysregulates cell metabolism and leukemia-stroma interactions and resensitizes acute myeloid leukemia to BCL-2 inhibition. Haematologica. 2020. https://doi.org/10.3324/haematol.2020.260331.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Roca-Portoles A, Rodriguez-Blanco G, Sumpton D, Cloix C, Mullin M, Mackay GM, et al. Venetoclax causes metabolic reprogramming independent of BCL-2 inhibition. Cell Death Dis. 2020;11(8):616.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Lee JB, Khan DH, Hurren R, Xu M, Na Y, Kang H, et al. Venetoclax enhances T cell-mediated antileukemic activity by increasing ROS production. Blood. 2021;138(3):234–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Chen X, Glytsou C, Zhou H, Narang S, Reyna DE, Lopez A, et al. Targeting mitochondrial structure sensitizes acute myeloid leukemia to venetoclax treatment. Cancer Discov. 2019;9(7):890–909.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Lucantoni F, Düssmann H, Llorente-Folch I, Prehn JHM. BCL2 and BCL(X)L selective inhibitors decrease mitochondrial ATP production in breast cancer cells and are synthetically lethal when combined with 2-deoxy-D-glucose. Oncotarget. 2018;9(40):26046–63.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Lee KM, Giltnane JM, Balko JM, Schwarz LJ, Guerrero-Zotano AL, Hutchinson KE, et al. MYC and MCL1 cooperatively promote chemotherapy-resistant breast cancer stem cells via regulation of mitochondrial oxidative phosphorylation. Cell Metab. 2017;26(4):633-47.e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Sharon D, Cathelin S, Mirali S, Di Trani JM, Yanofsky DJ, Keon KA, et al. Inhibition of mitochondrial translation overcomes venetoclax resistance in AML through activation of the integrated stress response. Sci Transl Med. 2019. https://doi.org/10.1126/scitranslmed.aax2863.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Deng J, Isik E, Fernandes SM, Brown JR, Letai A, Davids MS. Bruton’s tyrosine kinase inhibition increases BCL-2 dependence and enhances sensitivity to venetoclax in chronic lymphocytic leukemia. Leukemia. 2017;31(10):2075–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Cervantes-Gomez F, Lamothe B, Woyach JA, Wierda WG, Keating MJ, Balakrishnan K, et al. Pharmacological and protein profiling suggests venetoclax (ABT-199) as optimal partner with ibrutinib in chronic lymphocytic leukemia. Clin Cancer Res. 2015;21(16):3705–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Lin VS, Lew TE, Handunnetti SM, Blombery P, Nguyen T, Westerman DA, et al. BTK inhibitor therapy is effective in patients with CLL resistant to venetoclax. Blood. 2020;135(25):2266–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Mato AR, Roeker LE, Jacobs R, Hill BT, Lamanna N, Brander D, et al. Assessment of the efficacy of therapies following venetoclax discontinuation in CLL Reveals BTK inhibition as an effective strategy. Clin Cancer Res. 2020;26(14):3589–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Wang M, Ramchandren R, Chen R, Karlin L, Chong G, Jurczak W, et al. Concurrent ibrutinib plus venetoclax in relapsed/refractory mantle cell lymphoma: the safety run-in of the phase 3 SYMPATICO study. J Hematol Oncol. 2021;14(1):179.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Zhang J, Lu X, Li J, Miao Y. Combining BTK inhibitors with BCL2 inhibitors for treating chronic lymphocytic leukemia and mantle cell lymphoma. Biomark Res. 2022;10(1):17.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Eide CA, Kurtz SE, Kaempf A, Long N, Agarwal A, Tognon CE, et al. Simultaneous kinase inhibition with ibrutinib and BCL2 inhibition with venetoclax offers a therapeutic strategy for acute myeloid leukemia. Leukemia. 2020;34(9):2342–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Kuo HP, Ezell SA, Schweighofer KJ, Cheung LWK, Hsieh S, Apatira M, et al. Combination of ibrutinib and ABT-199 in diffuse large B-cell lymphoma and follicular lymphoma. Mol Cancer Ther. 2017;16(7):1246–56.

    Article  CAS  PubMed  Google Scholar 

  99. Cho H, Jang JE, Eom JI, Jeung HK, Chung H, Kim JS, et al. Arsenic trioxide synergistically promotes the antileukaemic activity of venetoclax by downregulating Mcl-1 in acute myeloid leukaemia cells. Exp Hematol Oncol. 2021;10(1):28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Byrd JC, Kitada S, Flinn IW, Aron JL, Pearson M, Lucas D, et al. The mechanism of tumor cell clearance by rituximab in vivo in patients with B-cell chronic lymphocytic leukemia: evidence of caspase activation and apoptosis induction. Blood. 2002;99(3):1038–43.

    Article  CAS  PubMed  Google Scholar 

  101. Seymour JF, Kipps TJ, Eichhorst B, Hillmen P, D’Rozario J, Assouline S, et al. Venetoclax-rituximab in relapsed or refractory chronic lymphocytic leukemia. N Engl J Med. 2018;378(12):1107–20.

    Article  CAS  PubMed  Google Scholar 

  102. Flinn IW, Gribben JG, Dyer MJS, Wierda W, Maris MB, Furman RR, et al. Phase 1b study of venetoclax-obinutuzumab in previously untreated and relapsed/refractory chronic lymphocytic leukemia. Blood. 2019;133(26):2765–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Jones CL, Stevens BM, D’Alessandro A, Reisz JA, Culp-Hill R, Nemkov T, et al. Inhibition of amino acid metabolism selectively targets human leukemia stem cells. Cancer Cell. 2018;34(5):724-40.e4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Pollyea DA, Stevens BM, Jones CL, Winters A, Pei S, Minhajuddin M, et al. Venetoclax with azacitidine disrupts energy metabolism and targets leukemia stem cells in patients with acute myeloid leukemia. Nat Med. 2018;24(12):1859–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. DiNardo CD, Pratz KW, Letai A, Jonas BA, Wei AH, Thirman M, et al. Safety and preliminary efficacy of venetoclax with decitabine or azacitidine in elderly patients with previously untreated acute myeloid leukaemia: a non-randomised, open-label, phase 1b study. Lancet Oncol. 2018;19(2):216–28.

    Article  CAS  PubMed  Google Scholar 

  106. DiNardo CD, Pratz K, Pullarkat V, Jonas BA, Arellano M, Becker PS, et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood. 2019;133(1):7–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Liu K, Li Y, Qiu S, Zhou C, Wei S, Lin D, et al. Efficacy of combination of venetoclax with azacitidine or chemotherapy in refractory/relapse acute leukemias of ambiguous lineage, not otherwise specified. Exp Hematol Oncol. 2021;10(1):46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Buettner R, Nguyen LXT, Morales C, Chen MH, Wu X, Chen LS, et al. Targeting the metabolic vulnerability of acute myeloid leukemia blasts with a combination of venetoclax and 8-chloro-adenosine. J Hematol Oncol. 2021;14(1):70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Donoso-Bustamante V, Borrego EA, Schiaffino-Bustamante Y, Gutiérrez DA, Millas-Vargas JP, Fuentes-Retamal S, et al. An acylhydroquinone derivative produces OXPHOS uncoupling and sensitization to BH3 mimetic ABT-199 (Venetoclax) in human promyelocytic leukemia cells. Bioorg Chem. 2020;100: 103935.

    Article  CAS  PubMed  Google Scholar 

  110. Dong Z, Abbas MN, Kausar S, Yang J, Li L, Tan L, et al. Biological functions and molecular mechanisms of antibiotic tigecycline in the treatment of cancers. Int J Mol Sci. 2019;20(14):3577.

    Article  CAS  PubMed Central  Google Scholar 

Download references

Acknowledgements

Thanks are due to American Journal Experts (AJE) for assistance with professional language editing.

Funding

This work was supported by the Zhejiang Provincial Natural Science Foundation of China (LY20H080002), and the Medical Health Science and Technology Project of Zhejiang Provincial Health Commission (2019KY452).

Author information

Authors and Affiliations

Authors

Contributions

YX designed this review, collected the related references and drafted the manuscript. HY performed the selection of literature, prepared the figures and revised the manuscript. All authors contributed to this manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Haige Ye.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, Y., Ye, H. Progress in understanding the mechanisms of resistance to BCL-2 inhibitors. Exp Hematol Oncol 11, 31 (2022). https://doi.org/10.1186/s40164-022-00283-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40164-022-00283-0

Keywords