Next Article in Journal
YAP, ΔNp63, and β-Catenin Signaling Pathways Are Involved in the Modulation of Corneal Epithelial Stem Cell Phenotype Induced by Substrate Stiffness
Next Article in Special Issue
Epilepsy in Tubulinopathy: Personal Series and Literature Review
Previous Article in Journal / Special Issue
Regulation of Microtubule Nucleation in Mouse Bone Marrow-Derived Mast Cells by Protein Tyrosine Phosphatase SHP-1
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Regulating the BCL2 Family to Improve Sensitivity to Microtubule Targeting Agents

by
Robert H. Whitaker
and
William J. Placzek
*
Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
*
Author to whom correspondence should be addressed.
Cells 2019, 8(4), 346; https://doi.org/10.3390/cells8040346
Submission received: 28 February 2019 / Revised: 28 March 2019 / Accepted: 9 April 2019 / Published: 12 April 2019
(This article belongs to the Special Issue Tubulin: Structure, Functions and Roles in Disease)

Abstract

:
Chemotherapeutic targeting of microtubules has been the standard of care in treating a variety of malignancies for decades. During mitosis, increased microtubule dynamics are necessary for mitotic spindle formation and successful chromosomal segregation. Microtubule targeting agents (MTAs) disrupt the dynamics necessary for successful spindle assembly and trigger programmed cell death (apoptosis). As the critical regulators of apoptosis, anti-apoptotic BCL2 family members are often amplified during carcinogenesis that can result in MTA resistance. This review outlines how BCL2 family regulation is positioned within the context of MTA treatment and explores the potential of combination therapy of MTAs with emerging BCL2 family inhibitors.

1. Introduction

Chemotherapeutics aim to exploit unique metabolic differences between cancer and normal cells in an effort to kill cancer cells more rapidly than the surrounding healthy tissue. For this reason, microtubule targeting agents (MTAs, also called microtubule poisons) have been employed in treating a variety of malignancies, including hematologic and solid tumors for decades [1]. MTAs are effective, as they target the persistent growth phenotype exhibited by cancer. This heightened growth potential is one of the key differences between malignant and healthy cells that allows MTAs to be a standard of care in the treatment of multiple human cancers. Despite the prevalent historical and current application of MTAs, an inherent issue of the persistent use of broad chemotherapeutics is the emergence of drug resistant cellular populations. Multiple mechanisms for resistance to chemotherapeutics have been observed in human cancers, including altered drug metabolism, altered clearance, as well as modulation of cell death [2]. For MTAs, a primary determinant of response and resistance occurs through modulation of the stress-induced BCL2 protein family of programmed cell death (apoptosis) regulators. However, microtubules and the BCL2 family are connected through more than just chemoresistance, as both are regulated differentially and sequentially throughout the cell cycle. Indeed, MTAs are often termed “anti-mitotic” due to their significant effect during mitosis, even though microtubule dynamics are involved in processes throughout the cell cycle [3]. The purpose of this review is to outline how BCL2 family regulation is positioned within the context of MTA treatment and the potential that the application of combining MTAs with emerging BCL2 family inhibitors has on improving anti-cancer therapy.

2. Microtubules and Tubulins

Microtubules are polymeric protein assemblies that are involved in many diverse cellular structures and processes. They have key functions outside the cell cycle, including cytoskeleton formation, cell movement, and intra-cellular trafficking. During cell division, microtubules have increasingly critical functions that ensure proper chromosome segregation beginning in interphase and culminating in mitosis. The various functions of microtubules, including both cellular structure and movement, are possible due to their dynamic, hollow, and cylindrical polymeric structure. As polymers, canonical microtubules are formed from 13 protofilament chains composed of both alpha and beta tubulin dimers. This is an evolutionarily conserved structural feature present in all eukaryotic supergroups with the 13 protofilaments providing a linear lattice that runs parallel to the length of the polymer and may allow for direct transport along the microtubule [4]. The sequential addition of tubulin heterodimer (αβ/αβ/αβ/αβ…) creates a polarity within both the protofilament and entire microtubule. The termini of the microtubule are therefore referred to as either the minus end or plus end with alpha tubulin always capping the minus end and the beta tubulin capping the plus end. (Figure 1). The minus and plus termini do not function equally, as the plus termini lengthens and shortens at a faster rate than the minus termini [1,5]. This dynamic instability of the microtubule poles, the constant lengthening and shortening, is called microtubule dynamics. Microtubule dynamics allows the overall tubular structure of the microtubule to be utilized by the cell in of diverse set of functions, including mitotic chromosomal segregation and intracellular trafficking. Although variable, when viewed across the cell cycle, a regular pattern of microtubule dynamics emerges with a maximum occurring during mitosis.
The essential unit of both the protofilament and microtubule is the protein tubulin. There are six different eukaryotic families (Alpha, Beta, Gamma, Delta, Epsilon, and Zeta) that comprise the tubulin superfamily. The alpha and beta tubulin families are the major constituents and serve as the primary components that form the microtubule. The gamma tubulin family forms gamma tubulin ring complexes (gamma-TuRC) that serve as templates for microtubule nucleation [6]. The other three tubulin families have specialized functions and are typically associated with the centriole [7]. Due to their central role, both alpha and beta tubulin have multiple isotypes. The alpha tubulin family consists of eight isotypes, while the beta tubulin family has nine isotypes. Members of the beta family share immense sequence and structural homology with primary sequence variation of only 4-16%, which is clustered at the C terminus [8,9,10]. This C-terminal variability is the basis for beta tubulin isotope identification and naming convention [9]. Beta tubulin isotype expression can be either constitutive or highly tissue specific. For example, beta tubulin isotope IVa typically has high expression in neurons, while isotype I is found in almost all cell types [8]. However, the diversity of both alpha and beta tubulin isotypes does not limit their incorporation within the same microtubule. Due to this observation, microtubules have been called “mosaics” that are formed from the variety of tubulin isotype populations that exist within the cellular milieu [11]. Characterization of the effect that tubulin isotype diversity has on its organization or patterning within the microtubule and how this impacts both microtubule function and MTA treatment is ongoing [11,12]. Importantly, while both alpha and beta tubulins have similar masses and three dimensional structures, all MTAs bind exclusively to members of the beta tubulin family.
Alpha and beta tubulin exist in an equilibrium between a soluble pool of tubulin heterodimers and polymerized tubulin that is assembled into microtubules [11]. This equilibrium changes throughout the cell cycle, where the microtubule half time varies from minutes to hours during interphase but only 10–30 s in mitosis [13]. This peak of microtubule dynamics that occurs during mitosis is necessary for mitotic spindle formation. Correct formation of the mitotic spindle is tightly regulated and oversight of its proper formation is termed the spindle assembly checkpoint (SAC). Regulated spindle assemble and SAC progression are needed for successful chromosomal segregation [14]. The purpose of the SAC is to prevent improper chromosomal segregation through inhibition of the anaphase promoting complex (APC) [15,16]. The APC is an E3 ubiquitin ligase that targets cyclin B1 for degradation and serves as a primary signal for mitotic exit [15]. Cellular treatment with MTAs disrupts necessary microtubule dynamics during mitosis which results in an increase in SAC inhibition of the APC and ultimately SAC dependent mitotic arrest [15]. Physiologically, this type of arrest is necessary to allow all kinetochores located at chromosomal centromeres time to organize and attach to the polar microtubules that will guide sister chromatids to opposite poles of mitotic cells [14,17]. The physiologic increase of microtubule dynamics necessary for mitotic spindle formation serves as a viable chemotherapeutic target due to the robust growth potential of cancer that increases the overall percentage of time that a cell spends in mitosis. MTAs interfere with this process and significantly prolong mitotic arrest inducing initiation of the desired apoptotic cascade.

3. Microtubule Targeting Agents (MTAs)

3.1. Background

MTAs were first identified in the middle of the 20th century through natural products research searching for anti-cancer agents [18]. Since their identification, both vinca alkaloids and taxanes have been used extensively as broad anti-cancer chemotherapeutics [19]. Initially, MTAs were discovered through their anti-mitotic ability, even before their effects on microtubule dynamics were understood. MTAs have been found to associate with one of four possible binding sites on beta tubulins [3]. Importantly, the site that a MTA binds to determines which of two possible outcomes the MTA induces on the microtubule. Association with either the colchicine or vinca alkaloid site results in microtubule depolymerization, while association with the laulimalide site or taxane/epithilone site stabilizes the microtubule [20]. Despite the advent of “second generation” anti-mitotic drugs developed to target specific proteins involved in chromosomal segregation, spindle formation, or mitosis [21], the initially identified microtubule targeting agents remain the chemotherapeutic gold standard, as many of these new compounds fail to retain sufficient cell killing potential [22]. For the purpose of this review, we will focus on the compounds that target either the vinca alkaloid (depolymerizing) or taxane (stabilization) sites due to their clinical relevance in cancer. For the vinca alkaloids, these include vinblastine (Velban) and vincristine (Oncovin), while for the taxanes, these include both paclitaxel (Abraxane) and docetaxel (Taxotere).
MTAs are a diverse set of compounds, while the primary function of taxanes and vinca alkaloids as either stabilizing or destabilizing divides them into two categories [1]. Interestingly, the biochemical outcome of either category of drug is shared at low concentrations, as they both cause an overall inhibition in microtubule dynamics [1]. This negative effect on microtubule dynamics has a direct effect on spindle efficacy that results in extended mitotic arrest [23]. This induces the cell to undergo programmed cell death (apoptosis), the clinically desired outcome of MTAs.

3.2. Vinca Alkaloids

Both of the vinca alkaloids that ultimately entered the clinic were discovered in the 1950s through analysis of extracts of the subtropical flower Vinca rosea (Catharanthus roseus) [24]. Vincristine was the first of these to be approved by the FDA in 1963, with vinblastine shortly following in 1965 [25]. Three other vinca alkaloids, vinorelbine, vindesine, and vinflunine, are semisynthetic derivatives of vinblastine, though they have not progressed to the clinic [3]. Although vincristine has neurotoxic effects, it is indicated for use in leukemias and lymphomas as well as lung and testicular solid cancers. Vinblastine, while myelosuppressive, is tolerated at higher doses than vincristine, and indicated for Hodgkin’s lymphoma, bladder, breast, and brain cancer [26]. Vinca alkaloids associate with both free beta tubulin and with exposed polymerized beta tubulin at the plus end of the microtubules. Mechanistically, vinca alkaloids function by binding to and sequestering free beta tubulin which limits its supply for microtubule extension. Likewise, vinca alkaloids can bind to polymerized beta tubulin, blocking further microtubule extension [27]. In each case, the compounds stop microtubule growth which switches the dynamics to favor loss in microtubule length and thus destabilization.

3.3. Taxanes

Like vinca alkaloids, paclitaxel was discovered in the early 1960s in extracts of Taxus brevifolia (Pacific Yew) bark during a National Cancer Institute screen for natural products with anti-cancer activity [18,28]. Subsequent characterization of paclitaxel’s chemical structure and its function on microtubules was completed over the next two decades [19,29,30]. Paclitaxel was initially only available as a natural product and with a limited supply of Pacific Yew a new source or synthetic scheme to make paclitaxel was sought. Docetaxel is a semi-synthetic derivative of a taxane, 10-deacetylbaccatin III, identified in Taxus baccata (English Yew) that was discovered as a result of the immense demand for paclitaxel [18]. A natural product from the English Yew, 10-deacetylbaccatin III, was a viable candidate to serve as an alternative starting point for the synthesis of paclitaxel. However, during the organic synthesis, an esterification reaction did not initially work which serendipitously directed the reaction to the end product of docetaxel [18]. Conveniently, docetaxel is more soluble than paclitaxel and exhibits less neurotoxicity [18,31]. Paclitaxel is indicated for multiple solid tumors including ovarian, breast, and lung, while docetaxel is indicated for breast, gastric, head and neck, and prostate cancers [24].
Unlike vinca alkaloids, paclitaxel and other taxanes predominantly bind polymerized beta tubulin at a separate site within the lumen of the microtubule nestled between parallel protofilaments [32]. Instead of destabilizing, this association stabilizes the protofilaments as well as the microtubule as a whole, inhibiting disassembly of the microtubule (also called microtubule catastrophe). As hydrophobic cytotoxic chemicals, MTAs are not without their problems, which include off-target toxicity and poor drug solubility. To this end, both vinca alkaloid and paclitaxel have served as the chemical foundation for larger chemical families that are derived from their chemical scaffolds. While these retain their specific regional interactions on beta tubulin (Figure 1) and their functional inhibition of microtubule dynamics, the majority of these novel chemicals do not retain the potency observed their parental compounds [31].

3.4. MTA Chemoresistance

Cancer cells have evolved to evade normal regulation of growth and differentiation. Many chemotherapeutics have been identified due to their ability to exploit such changes in cancer cell growth and metabolism. Yet, the same alterations that enable cancer to emerge leads to the proliferation of chemotherapeutic resistant populations that develops either the ability to evade chemotherapy induced death or the ability to remove the chemotherapeutic agents from the cellular environment. As taxanes and vinca alkaloids closely mimic one another’s cellular function, likewise, many of the same mechanisms are employed to gain resistance to both vinca alkaloids and taxanes. These include the induction of multi-drug resistance efflux transporters, e.g., P-glycoprotein, that pump hydrophobic natural products, including vinca alkaloids and taxanes, out of the cell [28]. Interestingly, as low concentrations of both of these two classes of MTAs were shown to perturb microtubule dynamics, it was proposed that this mechanism might not be as detrimental to MTA activity as once thought [1]. This may explain why the co-treatment of MTAs with transport inhibitors has seen limited success [33,34]. Another mechanism of MTA resistance is the diversity of tubulin isotypes. There are nine different isotypes of beta tubulin located on separate genes and with different primary amino acid sequences. It was shown that some beta isotypes are more resistant to MTA treatment. Specifically, the beta tubulin III isotype is particularly troublesome for both paclitaxel and vinca alkaloids [8,33,35]. Typically, beta tubulin III has been shown to increase microtubule dynamics and is physiologically observed in neurons where it plays a role in cellular projections or neurites [36]. When elevated in cancer cells, this modulation of microtubule dynamics appears to destabilize the microtubule and mitigate MTA efficacy [8,36]. A third mechanism of MTA chemotherapeutic resistance is suppression of the apoptotic response. A primary function of MTA treatment is the induction of apoptosis or programmed cell death. It was shown that MTA treatment increases post-translational modification of the anti-apoptotic BCL2 family through kinase activation (JNK, CDK1, and many others [37,38,39]. Although phosphorylation of these anti-apoptotic proteins can either facilitate or inhibit their function, as a whole, this type of phosphorylation event is primarily inhibitory and, thus, pro-apoptotic [3,40]. Yet, efficacy of MTA compounds can be attenuated through presence or upregulation of anti-apoptotic proteins [41]. Indeed, this was shown to occur through the altered expression of proteins, such as p53 and BCL2 [28,42]. Importantly, it was shown that the MTA drug concentrations needed to directly promote apoptosis are not met in vivo [43]. This necessitates combination therapy of MTAs with other chemotherapeutic or targeted approaches. Functional MTAs canonically induce cell death during mitosis, with intrinsic apoptosis serving as the major mechanism regulating mitotic cell death [44,45]. In human cell, the intrinsic apoptosis response is primarily regulated by the BCL2 family. Thus, MTA efficacy and BCL2 functionality are intrinsically linked and this link has been strengthened through targeted studies over the past two decades [46]. Likewise, MTA resistance was shown to be strongly impacted by BCL2 family expression and function [47,48]. These links between the BCL2 family and both MTA efficacy and resistance uniquely position the BCL2 family to be targeted alongside MTA therapy.

4. BCL2 Family of Proteins

To understand the impact of the BCL2 family on MTA efficacy, it is first necessary to review how the BCL2 family regulates apoptosis. BCL2, the eponym of the BCL2 family, was initially observed in the late 1970s through its upregulation due to chromosome 14 translocations in lymphomas [49,50]. Within the next decade, human BCL2 was cloned and identified as an anti-apoptotic oncogene with the potency to protect against multiple forms of cellular stress [50,51,52]. Subsequent sequence analysis has identified a number of proteins that share from one to four homologous sequence motifs, referred to as BCL2 homology (BH) motifs with BCL2, thus making them part of the BCL2 family (outlined in Reference [53]). Based on the composition of these four motifs (BH1, BH2, BH3, and BH4), the BCL2 family can be divided into three main categories: Anti-apoptotic, pro-apoptotic effectors, and pro-apoptotic activators. As a group, the BCL2 family is a collection of both pro- and anti-apoptotic proteins that are evolutionarily conserved and found in all animals [54]. The function of the BCL2 family is diverse, extending beyond apoptosis with important roles in both development [55] and homeostasis [56]. The dysregulation of the BCL2 family, either through the upregulation of anti-apoptotic members or downregulation of pro-apoptotic members, has been implicated throughout tumorigenesis and tumor progression [57]. The interplay between the pro-survival and pro-apoptotic BCL2 proteins provides the mechanism or biochemical balance that cellular signaling is filtered through in order to determine cell fate. In the event of increased pro-apoptotic signaling, the BCL2 family can initiate a cascade that concludes with caspase activation, cellular degradation, and phagocytosis in a manner that does not stress the neighboring tissue [58,59].
The focal point of BCL2 family regulation centers on the pro-apoptotic effectors, BAK and BAX, which can be induced through upregulation or activation to form homo-oligomers in the mitochondrial outer membrane. These oligomers form pores in the mitochondrial outer membrane, inducing mitochondrial outer membrane permeabilization (MOMP), a necessary event for cytochrome c release and subsequent apoptosis. In a non-stressed cell with basal expression of BAK and BAX, these proteins reside in the cytosol or mitochondrial outer membrane, respectively, in a monomeric and inactive state. Stress-induced upregulation of either BAK or BAX or one of the BH3-only activators (BID, BIM, or PUMA), can induce BAK or BAX oligomerization. In either case, oligomerization of BAK and BAX can be held in check by the presence of anti-apoptotic BCL2 family members (BCL2, BCLxL, BCLW, MCL1, and BFL1/A1) that contain all four BH motifs structurally folded in such a way to form a hydrophobic groove or pocket. This pocket binds to amphipathic alpha helical BH3 motifs that are present in both classes of pro-apoptotic BCL2 family members. The affinity of the BH3 helix and the BH3 binding pocket is the hinge between the two poles, pro-apoptotic and pro-survival, within the BCL2 family and the basis for BCL2 regulation of cell fate. The canonical BH3 helix is a structural motif whose primary sequence is capped in the C-terminal direction with aspartic acid [53]. Moving in the N-terminal direction, the next residues are often glycine, followed by isoleucine. The rest of the BH3 primary sequence is interspersed with both charged and polar residues, such that when the BH3 helix is folded, the hydrophobic residues reside throughout one half of the long axis of the helix, thus creating its amphipathic nature. The four hydrophobic residues of the BH3 bind into specific pockets (p1-p4) [60,61,62] within the BH3 binding groove (Figure 2). This hydrophobic side of the BH3 helix is largely the driver of binding to the hydrophobic BH3 pocket found on the anti-apoptotic BCL2 family members.
Association of anti-apoptotic BCL2 family proteins with either the BAK or BAX BH3 helix inhibits their oligomerization. The third BCL2 subfamily, the BH3 only proteins, retain only the BH3 motif. Two prevailing models (direct and indirect) were proposed to describe the impact that BH3-only proteins utilize to regulate the BCL2 family [53,63]. In the direct model, BH3 only proteins bind directly to the pro-apoptotic effectors, BAK and BAK, stimulating oligomerization [64,65,66]. In the indirect, BH3 only protein act as apoptotic sensitizers, inhibiting the anti-apoptotic BCL2 family members and thereby allowing BAK and BAX to oligomerize [63,67,68]. The BH3-only subfamily contains over 20 identified members in humans, and different members were shown to favor one or the other of these models. Further, it was demonstrated that in the absence of BH3 only proteins, BAK and BAX, are able to homo-oligomerize and permeabilize the mitochondrial outer membrane [69]. Thus, both the direct and indirect activation of pro-apoptotic effectors are not mutually exclusive and a unified theory that combines both was proposed [63]. Not unlike tubulin heterodimers cycling between polymerized tubulin within the microtubule and solubilized tubulin populations, BAK and BAX are also able to translocate between the cytosol, endoplasmic reticulum, and the mitochondrial outer membrane [70,71]. Regulation of this process mimics the general regulation of BAK and BAX oligomerization with BAK and BAX retro-translocation away from the mitochondria being carried out by the anti-apoptotic BCL2 family members [72], while the BH3 only protein, BIM, is involved in BCL2 translocation back to the mitochondria [66]. Thus, the BCL2 family regulates not only BAK and BAX oligomerization, but also their localization into the mitochondrial outer membrane in their regulation of stress-induced apoptosis.

4.1. Anti-Apoptotic BCL2 Family and MTAs

The BCL2 family serve as the gatekeepers of an irreversible intrinsic apoptotic cascade. As critical regulators of cellular viability, the BCL2 family effectively determines cell fate through the interpretation and status of cellular stress. For this reason, many of the functions and capabilities of the BCL2 family have been elucidated through their effects on chemotherapy [41]. The treatment of cancer cells goes beyond the natural stress that tumorigenesis can often induce (e.g., chromosomal abnormalities, metabolic stress), as broad-spectrum chemotherapeutics intentionally induce acute cellular stress. MTAs targeting of microtubule dynamics introduces enormous stress on the cell to which the anti-apoptotic BCL2 family members are obligated to respond to enable the cell to survive. Repeatedly, studies have highlighted the importance of the BCL2 family in mitigating the effects of MTAs, both taxanes [21] and vinca alkaloids [3]. Interestingly, both of these drug classes have been shown to collectively initiate downstream effects on all five anti-apoptotic BCL2 family proteins. Given the importance of the anti-apoptotic BCL2 family members in enabling cancer cell survival, a number of small molecule inhibitors of this family were identified and are being tested for their anti-cancer activity. With the first of these compounds entering the clinic [73], one interesting question to be answered is how the combination of MTAs with different and individually specific anti-apoptotic BCL2 inhibitors can overcome MTA resistance or improve response. Thus far, a number of studies investigated cell lines and xenograft models to determine the impact that anti-apoptotic protein expression or suppression has on MTA activity. These studies are reviewed in the following sections and summarized in Table 1.

4.2. BCL2

As the namesake of the BCL2 family and the template anti-apoptotic family member, the BCL2 protein plays an unexpected role in MTA sensitivity. As an anti-apoptotic protein, it might be expected that elevated levels of BCL2 protein expression would lead to MTA resistance. Interestingly, the opposite is observed in both lung and breast cancer lines where elevated BCL2 protein expression has been shown to increase paclitaxel sensitivity [37]. This is supported by the observation in prostate cancer models that loss of BCL2, such as in the BCL2 null cell line (DU145), corresponds with resistance to paclitaxel, while a BCL2 expressing cell line (PC3) is sensitive to paclitaxel [39]. One possible explanation for this effect is that elevated BCL2 expression coincides with increased expression of the pro-apoptotic, BH3 only protein, BIM [37]. Thus, when the cell overexpresses BCL2 to try and evade death, the corresponding upregulation of BIM balances the scales to ultimately provide no protective effect. In addition, a number of studies suggest that paclitaxel can be sequestered directly by BCL2 [85,86,87] and that this binding inhibits the pro-apoptotic effect of paclitaxel. As it was shown that the BCL2 protein is known to associate with microtubules [88], this may increase the stochastic possibility of BCL2 binding to and sequestering paclitaxel. In further agreement, loss of BCL2 was also associated with resistance to the vinca alkaloid, vinflunine, in ovarian cancer cells [89]. These data suggest that in solid tumors, BCL2 expression actually sensitizes cancer cells to the apoptotic inducing effect of MTAs. Intriguingly, the opposite effect is seen in the 697 leukemia cell line where overexpression of BCL2 protects these from paclitaxel-induced apoptosis [74]. This alternative effect similarly observed in another leukemia cell line (HL-60) wherein extended treatment with both paclitaxel and vincristine have been shown to decrease BCL2 mRNA expression [75]. These studies demonstrate that cell type, particularly the difference between solid and hematopoietic tumors, can impact both the presence and role of both BCL2 protein in its regulation of MTA treatment sensitivity.

4.3. BCLxL

Although it shares many functional similarities with its anti-apoptotic homolog, BCL2, the anti-apoptotic BCL2 family member, BCLxL, does not have as complicated a relationship with MTAs as BCL2. Exogenous expression of BCLxL was shown to protect both leukemia and solid tumor cells from paclitaxel-induced cell death [76,90]. Further, while targeted inhibition of BCLxL using a BH3 mimetic alone had minimal effect on cell viability, combination of this compound with paclitaxel led to a synergistic response [90]. The ability for BCLxL inhibitor to work in combination with taxanes was further demonstrated in a study to evaluate how selective and targeted inhibition of BCLxL impacts docetaxel treatment of an array of mouse xenografts [79]. These studies highlight the potential of using anti-apoptotic BCL2 family inhibitors alongside MTAs to improve therapeutic response.

4.4. BCLW and BFL1/A1

Both of the anti-apoptotic BCL2 family members BCLW and BFL1/A1 function as much as BCLxL with regard to their impact on MTA sensitivity. Analysis of siRNA knockdown or CRISPR-Cas9 deletion of BCLW resulted in an increase in the rate of paclitaxel-induced cell death [77], while overexpression of BCLW prolonged cell viability in the presence of paclitaxel [77]. Further, exogenous upregulation of BFL1/A1 in leukemia cells decreased apoptotic induction by paclitaxel [78]. This impact on MTA activity was also observed in vinca alkaloid combinations in a murine lymphoma model. In these studies, cellular resistance to the vinca alkaloid, vinfuline, was characterized by elevated BFL1/A1 protein levels [84]. These studies highlight the necessity of targeting the specific anti-apoptotic BCL2 proteins that seem to be upregulated in a compensatory manner upon MTA treatment.

4.5. MCL1

The final anti-apoptotic BCL2 family member, MCL1, has been closely linked with MTA resistance. This involvement in MTA chemoresistance assisted in identifying MCL1′s key role in acting as a mitotic clock and serving as the key anti-apoptotic BCL2 protein during mitosis [44]. During extended mitotic arrest, the cell is presented with two options: Death or mitotic slippage. Throughout this cellular decision, the abundance of MCL1 protein was identified to be a critical factor that acts as a mitotic timer [48,91,92]. When MCL1 protein levels are low or absent, the arrested cell is tipped into the apoptotic cascade. But, when MCL1 is in high abundance, cellular viability is maintained and the cell may either correct itself and divide with proper chromosomal separation or experience mitotic slippage. When the cell escapes mitotic arrest through mitotic slippage, the cell bypasses the final stages of mitosis, which often results in aberrant cytokinesis, including incomplete or asymmetric chromosomal segregation. This ultimately leads to the presence of abnormal chromosome copy numbers in daughter cells. Clinically, mitotic slippage caused by prolonged MTA-induced mitotic arrest is undesirable. Since MCL1 was recognized as regulator of mitotic escape, a number of studies continue to highlight how modulation of MCL1 effects MTA treatment [80,81,93].
Initial studies to assess the impact of MCL1 on MTA sensitivity highlighted the impact that MCL1 levels have on both sensitivity and resistance to taxanes and vinca alkaloids [48]. Likewise, knockdown of PIAS1, an E3 SUMO ligase that is linked with decreased MCL1 protein levels, was shown to sensitize a docetaxel-resistance PC3 sub-cell line to docetaxel [80]. Regulation of MCL1 expression can similarly impact MTA activity as it has been shown that expression of the RNA binding protein, PTBP1 (HUR1) regulates sensitivity to both vincristine and paclitaxel through its regulation of MCL1 [81]. Interestingly, the vinca alkaloid, vinblastine, is able to mediate a decrease in MCL1 protein through phosphorylation events leading to its ubiquitination [82,83]. Multiple drugs have been developed to inhibit the BCL2 family, while it is notable that cancer sensitivity to combination therapies that include MTAs can be adversely affected by drug regulation of the BCL2 family. For instance, therapeutic combination of proteasome inhibitors, such as Bortezomib, and MTAs are employed in the clinic to treat multiple cancers [94]. Yet, proteasome inhibition was shown to increase MCL1 protein levels (as it blocks MCL1 proteolytic degradation). This increase was shown to inhibit taxane-induced mitotic death [91].

5. Combination of BCL2 Family Inhibitors and MTAs

The tension between the pro- and anti- apoptotic BCL2 proteins dictated by both stoichiometry and binding affinity results in a homeostatic balance within the BCL2 family (Figure 2). Cancers that are addicted to the anti-apoptotic side of the family [41] are tipped toward the pro-survival direction. One method to therapeutically correct this scale is to introduce BH3 mimetics, small molecules that bind into the hydrophobic BH3 pocket of the anti-apoptotic members, and displace the pro-apoptotic activators and effectors. These inhibitors were first developed to bind to BCLxL and BCL2 and utilized the p3 and p4 hydrophobic pockets, as well as mimicking the conserved aspartate salt bridge (Figure 2). Thus far, a specific inhibitor for BCL2 (ABT-199, Venetoclax [95]) obtained approval for treatment of chronic lymphocytic leukemia (CLL). However, leading up to the FDA approval of ABT-199, many other small molecules inhibitors of the BCL2 family were developed, and these have been invaluable for pre-clinical assessment of anti-apoptotic BCL2 targeting. These include, ABT-737 [96], ABT-263 (Navitoclax) [97], Sabutoclax [98], A1210477 [79], and S63845 [41,99]. The utility of these individual small molecule inhibitors is that while they are all BH3 mimetics, they do not bind equally to all of the anti-apoptotic BCL2 family members. The first of these inhibitors to be developed, ABT-737, specifically targets BCL2, BCLW, and BCLxL as is the clinical candidate that was developed based on its initial preclinical success, ABT-263. Sabutoclax was the first pan-active BCL2 inhibitor capable of targeting all 5 anti-apoptotic proteins. A1210477 and S63845 followed up on the promise that MCL1 inhibition seemed to offer as MCL1 specific inhibitors. This has allowed direct studies to determine the importance of individual BCL2 proteins both as single agents and in combination with other chemotherapeutic agents.
The identification that anti-apoptotic BCL2 family members can regulate cellular sensitivity to MTAs led to a series of studies evaluating how these various inhibitors combine with both taxanes and vinca alkaloids. Thus far, each of these compounds been shown to increase the efficacy of MTAs (Table 2). Figure 3 demonstrates how BH3 mimetics feed into BCL2 signaling resulting in cell death. The first developed and tested BH3 mimetic, ABT-737, was shown to sensitized multiple different cancer cell types (breast [100], prostate [101], melanoma [102], and hepatoblastoma [103]) to taxanes. MCL1 inhibition further provided synergistic activity with docetaxel in both prostate cancer cell lines and xenograft studies [101]. It is perhaps no accident that ABT-199, specific for BCL2, was approved for treatment of CLL based on the differences observed in MTA regulation of BCL2 between solid and liquid tumors. This effect is recapitulated by small molecule inhibition of BCL2, where in both prostate and breast cell models ABT-199 and BCL2 inhibition do not synergize with paclitaxel treatment but both ABT-737 [104] and ABT-263 [105] do synergize, likely due to their inhibition of BCLxL. In prostate cells, inhibition of BCLxL has been shown to be beneficial, though not equally, for both paclitaxel sensitive and resistant cells [105]. Teasing apart the effects of BH3 mimetics that target more than one BCL2 protein has led to the continued development of inhibitors for individual anti-apoptotic BCL2 members. Inhibitors of both MCL1 and BCLxL are in clinical trials [73,106,107]. Unlike BCL2 inhibition, the MCL1 specific inhibitor, S63845, has shown promise in targeting and sensitizing triple negative breast cancer to docetaxel [108]. From these early studies, BH3 mimetics may enable significant reduction in MTA dosing while retaining clinical effect. As more potent and specific anti-apoptotic BCL2 family inhibitors emerge, continued assessment of their impact on MTA sensitivity will be important not only to identify cancers that can be best treated through combination of BH3 mimetics and MTAs, but also as BH3 mimetics may resensitize cancers to MTA treatment.

6. Conclusions

Since their identification, MTAs have become the standard of care in treating many cancer malignancies. However, MTAs are not without their problems, namely toxicity toward healthy tissues, including the brain. The BCL2 family, as the key regulators of programmed cell death and crucial mediators for cellular death during mitosis, play a key role in MTA effectiveness. The recent development of small molecules that can specifically target the BCL2 family opened a new avenue for improving cancer therapy and, specifically, MTA effectiveness. The clinical potential of combining MTAs with BCL2 inhibition is only beginning to be explored, yet this limited work already hints at promising new combinations that exploit the ability of BCL2-inhibitors and MTAs to work in synergy to improve targeted cancer cell death, allow for reduced MTA dosage, and result in decreased toxicity. However, knowing when and where to target the BCL2 family individually or as a whole will lead to better combinatorial therapies with existing MTA therapies.

Funding

This work was supported, in part, by funding from the National Institutes of Health Grants R01GM117391 (to W.J.P.) and T32-NS048039 (to R.H.W.).

Acknowledgments

We would like to thank Hayley Widden and Kirsten Dotson for critical reading.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Mukhtar, E.; Adhami, V.M.; Mukhtar, H. Targeting microtubules by natural agents for cancer therapy. Mol. Cancer Ther. 2014, 13, 275–284. [Google Scholar] [CrossRef] [PubMed]
  2. Pan, S.T.; Li, Z.L.; He, Z.X.; Qiu, J.X.; Zhou, S.F. Molecular mechanisms for tumour resistance to chemotherapy. Clin. Exp. Pharmacol. Physiol. 2016, 43, 723–737. [Google Scholar] [CrossRef]
  3. Bates, D.; Eastman, A. Microtubule destabilising agents: Far more than just antimitotic anticancer drugs. Br. J. Clin. Pharmacol. 2017, 83, 255–268. [Google Scholar] [CrossRef]
  4. Chaaban, S.; Brouhard, G.J. A microtubule bestiary: Structural diversity in tubulin polymers. Mol. Biol. Cell 2017, 28, 2924–2931. [Google Scholar] [CrossRef] [PubMed]
  5. Jordan, M.A.; Wilson, L. Microtubules as a target for anticancer drugs. Nat. Rev. Cancer 2004, 4, 253–265. [Google Scholar] [CrossRef]
  6. Wiese, C.; Zheng, Y. Microtubule nucleation: Gamma-tubulin and beyond. J. Cell Sci. 2006, 119 Pt 20, 4143–4153. [Google Scholar] [CrossRef]
  7. McKean, P.G.; Vaughan, S.; Gull, K. The extended tubulin superfamily. J. Cell Sci. 2001, 114 Pt 15, 2723–2733. [Google Scholar]
  8. Burkhart, C.A.; Kavallaris, M.; Horwitz, S.B. The role of beta-tubulin isotypes in resistance to antimitotic drugs. Biochim. Biophys. Acta 2001, 2, O1–O9. [Google Scholar]
  9. Sullivan, K.F.; Cleveland, D.W. Identification of conserved isotype-defining variable region sequences for four vertebrate beta tubulin polypeptide classes. Proc. Natl. Acad. Sci. USA 1986, 83, 4327–4331. [Google Scholar] [CrossRef]
  10. Sullivan, K.F. Structure and utilization of tubulin isotypes. Annu. Rev. Cell Biol. 1988, 4, 687–716. [Google Scholar] [CrossRef]
  11. Roll-Mecak, A. How cells exploit tubulin diversity to build functional cellular microtubule mosaics. Curr. Opin. Cell Biol. 2019, 56, 102–108. [Google Scholar] [CrossRef]
  12. Cross, R.A. Microtubule lattice plasticity. Curr. Opin. Cell Biol. 2019, 56, 88–93. [Google Scholar] [CrossRef] [PubMed]
  13. Saxton, W.M.; Stemple, D.L.; Leslie, R.J.; Salmon, E.D.; Zavortink, M.; McIntosh, J.R. Tubulin dynamics in cultured mammalian cells. J. Cell Biol. 1984, 99, 2175–2186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Mitchison, T.J. Microtubule dynamics and kinetochore function in mitosis. Annu. Rev. Cell Biol. 1988, 4, 527–549. [Google Scholar] [CrossRef] [PubMed]
  15. Musacchio, A.; Salmon, E.D. The spindle-assembly checkpoint in space and time. Nat. Rev. Mol. Cell Biol. 2007, 8, 379–393. [Google Scholar] [CrossRef]
  16. Izawa, D.; Pines, J. How APC/C-Cdc20 changes its substrate specificity in mitosis. Nat. Cell Biol. 2011, 13, 223–233. [Google Scholar] [CrossRef]
  17. Maciejowski, J.; Drechsler, H.; Grundner-Culemann, K.; Ballister, E.R.; Rodriguez-Rodriguez, J.A.; Rodriguez-Bravo, V.; Jones, M.J.K.; Foley, E.; Lampson, M.A.; Daub, H.; et al. Mps1 Regulates Kinetochore-Microtubule Attachment Stability via the Ska Complex to Ensure Error-Free Chromosome Segregation. Dev. Cell 2017, 41, 143–156. [Google Scholar] [CrossRef]
  18. Le Roux, M.A.G. Francoise, From the Pacific Yew (Taxus brevifolia) to the English Yew (Taxus baccata): Steps Towards the Discovery of Docetaxel (Taxotere®). In Navelbine® and Taxotere®; Elsevier: Oxford, UK, 2017; pp. 151–212. [Google Scholar]
  19. Huizing, M.T.; Misser, V.H.; Pieters, R.C.; ten Bokkel Huinink, W.W.; Veenhof, C.H.; Vermorken, J.B.; Pinedo, H.M.; Beijnen, J.H. Taxanes: A new class of antitumor agents. Cancer Investig. 1995, 13, 381–404. [Google Scholar] [CrossRef]
  20. Akhmanova, A.; Steinmetz, M.O. Control of microtubule organization and dynamics: Two ends in the limelight. Nat. Rev. Mol. Cell Biol. 2015, 16, 711–726. [Google Scholar] [CrossRef]
  21. Haschka, M.; Karbon, G.; Fava, L.L.; Villunger, A. Perturbing mitosis for anti-cancer therapy: Is cell death the only answer? EMBO Rep. 2018, 19, e45440. [Google Scholar] [CrossRef] [PubMed]
  22. Otto, T.; Sicinski, P. Cell cycle proteins as promising targets in cancer therapy. Nat. Rev. Cancer 2017, 17, 93–115. [Google Scholar] [CrossRef] [Green Version]
  23. Seve, P.; Dumontet, C. Class III beta tubulin expression in nonsmall cell lung cancer. Rev. Mal. Respir. 2010, 27, 383–386. [Google Scholar] [CrossRef]
  24. Noble, R.L.; Beer, C.T.; Cutts, J.H. Role of chance observations in chemotherapy: Vinca rosea. Ann. N. Y. Acad. Sci. 1958, 76, 882–894. [Google Scholar] [CrossRef]
  25. DeVita, V.T., Jr.; Chu, E. A history of cancer chemotherapy. Cancer Res. 2008, 68, 8643–8653. [Google Scholar] [CrossRef]
  26. Xie, S.; Zhou, J. Harnessing Plant Biodiversity for the Discovery of Novel Anticancer Drugs Targeting Microtubules. Front. Plant Sci. 2017, 8, 720. [Google Scholar] [CrossRef]
  27. Dumontet, C.; Jordan, M.A. Microtubule-binding agents: A dynamic field of cancer therapeutics. Nat. Rev. Drug Discov. 2010, 9, 790–803. [Google Scholar] [CrossRef]
  28. Barbuti, A.M.; Chen, Z.S. Paclitaxel Through the Ages of Anticancer Therapy: Exploring Its Role in Chemoresistance and Radiation Therapy. Cancers 2015, 7, 2360–2371. [Google Scholar] [CrossRef] [Green Version]
  29. Wani, M.C.; Taylor, H.L.; Wall, M.E.; Coggon, P.; McPhail, A.T. Plant antitumor agents. VI. The isolation and structure of taxol, a novel antileukemic and antitumor agent from Taxus brevifolia. J. Am. Chem. Soc. 1971, 93, 2325–2327. [Google Scholar] [CrossRef]
  30. Schiff, P.B.; Fant, J.; Horwitz, S.B. Promotion of microtubule assembly in vitro by taxol. Nature 1979, 277, 665–667. [Google Scholar] [CrossRef]
  31. Yared, J.A.; Tkaczuk, K.H. Update on taxane development: New analogs and new formulations. Drug Des. Dev. Ther. 2012, 6, 371–384. [Google Scholar]
  32. Steinmetz, M.O.; Prota, A.E. Microtubule-Targeting Agents: Strategies to Hijack the Cytoskeleton. Trends Cell Biol. 2018, 28, 776–792. [Google Scholar] [CrossRef]
  33. Zhang, Y.; Yang, S.H.; Guo, X.L. New insights into Vinca alkaloids resistance mechanism and circumvention in lung cancer. Biomed. Pharmacother. 2017, 96, 659–666. [Google Scholar] [CrossRef]
  34. Thomas, H.; Coley, H.M. Overcoming multidrug resistance in cancer: An update on the clinical strategy of inhibiting p-glycoprotein. Cancer Control 2003, 10, 159–165. [Google Scholar] [CrossRef]
  35. Parker, A.L.; Kavallaris, M.; McCarroll, J.A. Microtubules and their role in cellular stress in cancer. Front. Oncol. 2014, 4, 153. [Google Scholar] [CrossRef]
  36. Lebok, P.; Ozturk, M.; Heilenkotter, U.; Jaenicke, F.; Muller, V.; Paluchowski, P.; Geist, S.; Wilke, C.; Burandt, E.; Lebeau, A.; et al. High levels of class III beta-tubulin expression are associated with aggressive tumor features in breast cancer. Oncol. Lett. 2016, 11, 1987–1994. [Google Scholar] [CrossRef]
  37. Savry, A.; Carre, M.; Berges, R.; Rovini, A.; Pobel, I.; Chacon, C.; Braguer, D.; Bourgarel-Rey, V. Bcl-2-enhanced efficacy of microtubule-targeting chemotherapy through Bim overexpression: Implications for cancer treatment. Neoplasia 2013, 15, 49–60. [Google Scholar] [CrossRef]
  38. Blagosklonny, M.V.; Giannakakou, P.; el-Deiry, W.S.; Kingston, D.G.; Higgs, P.I.; Neckers, L.; Fojo, T. Raf-1/bcl-2 phosphorylation: A step from microtubule damage to cell death. Cancer Res. 1997, 57, 130–135. [Google Scholar]
  39. Haldar, S.; Chintapalli, J.; Croce, C.M. Taxol induces bcl-2 phosphorylation and death of prostate cancer cells. Cancer Res. 1996, 56, 1253–1255. [Google Scholar]
  40. Pathan, N.; Aime-Sempe, C.; Kitada, S.; Haldar, S.; Reed, J.C. Microtubule-targeting drugs induce Bcl-2 phosphorylation and association with Pin1. Neoplasia 2001, 3, 70–79. [Google Scholar] [CrossRef]
  41. Montero, J.; Letai, A. Why do BCL-2 inhibitors work and where should we use them in the clinic? Cell Death Differ. 2018, 25, 56–64. [Google Scholar] [CrossRef]
  42. Huang, Y.; Ibrado, A.M.; Reed, J.C.; Bullock, G.; Ray, S.; Tang, C.; Bhalla, K. Co-expression of several molecular mechanisms of multidrug resistance and their significance for paclitaxel cytotoxicity in human AML HL-60 cells. Leukemia 1997, 11, 253–257. [Google Scholar] [CrossRef] [Green Version]
  43. Zasadil, L.M.; Andersen, K.A.; Yeum, D.; Rocque, G.B.; Wilke, L.G.; Tevaarwerk, A.J.; Raines, R.T.; Burkard, M.E.; Weaver, B.A. Cytotoxicity of paclitaxel in breast cancer is due to chromosome missegregation on multipolar spindles. Sci. Transl. Med. 2014, 6, 229ra43. [Google Scholar] [CrossRef]
  44. Haschka, M.D.; Soratroi, C.; Kirschnek, S.; Hacker, G.; Hilbe, R.; Geley, S.; Villunger, A.; Fava, L.L. The NOXA-MCL1-BIM axis defines lifespan on extended mitotic arrest. Nat. Commun. 2015, 6, 6891. [Google Scholar] [CrossRef]
  45. Blagosklonny, M.V. Mitotic arrest and cell fate: Why and how mitotic inhibition of transcription drives mutually exclusive events. Cell Cycle 2007, 6, 70–74. [Google Scholar] [CrossRef]
  46. Haldar, S.; Basu, A.; Croce, C.M. Bcl2 is the guardian of microtubule integrity. Cancer Res. 1997, 57, 229–233. [Google Scholar]
  47. Mollinedo, F.; Gajate, C. Microtubules, microtubule-interfering agents and apoptosis. Apoptosis 2003, 8, 413–450. [Google Scholar] [CrossRef] [Green Version]
  48. Wertz, I.E.; Kusam, S.; Lam, C.; Okamoto, T.; Sandoval, W.; Anderson, D.J.; Helgason, E.; Ernst, J.A.; Eby, M.; Liu, J.; et al. Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7. Nature 2011, 471, 110–114. [Google Scholar] [CrossRef]
  49. Fukuhara, S. Significance of 14q translocations in non-Hodgkin lymphomas. Virchows Arch. B 1978, 29, 99–106. [Google Scholar]
  50. Delbridge, A.R.; Grabow, S.; Strasser, A.; Vaux, D.L. Thirty years of BCL-2: Translating cell death discoveries into novel cancer therapies. Nat. Rev. Cancer 2016, 16, 99–109. [Google Scholar] [CrossRef]
  51. Reed, J.C.; Cuddy, M.; Slabiak, T.; Croce, C.M.; Nowell, P.C. Oncogenic potential of bcl-2 demonstrated by gene transfer. Nature 1988, 336, 259–261. [Google Scholar] [CrossRef]
  52. Tsujimoto, Y. Stress-resistance conferred by high level of bcl-2 alpha protein in human B lymphoblastoid cell. Oncogene 1989, 4, 1331–1336. [Google Scholar]
  53. Letai, A.; Bassik, M.C.; Walensky, L.D.; Sorcinelli, M.D.; Weiler, S.; Korsmeyer, S.J. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell 2002, 2, 183–192. [Google Scholar] [CrossRef] [Green Version]
  54. Lanave, C.; Santamaria, M.; Saccone, C. Comparative genomics: The evolutionary history of the Bcl-2 family. Gene 2004, 333, 71–79. [Google Scholar] [CrossRef]
  55. Ke, F.F.S.; Vanyai, H.K.; Cowan, A.D.; Delbridge, A.R.D.; Whitehead, L.; Grabow, S.; Czabotar, P.E.; Voss, A.K.; Strasser, A. Embryogenesis and Adult Life in the Absence of Intrinsic Apoptosis Effectors BAX, BAK, and BOK. Cell 2018, 173, 1217–1230. [Google Scholar] [CrossRef]
  56. Knudson, C.M.; Tung, K.S.; Tourtellotte, W.G.; Brown, G.A.; Korsmeyer, S.J. Bax-deficient mice with lymphoid hyperplasia and male germ cell death. Science 1995, 270, 96–99. [Google Scholar] [CrossRef]
  57. Luke, J.J.; van de Wetering, C.I.; Knudson, C.M. Lymphoma development in Bax transgenic mice is inhibited by Bcl-2 and associated with chromosomal instability. Cell Death Differ. 2003, 10, 740–748. [Google Scholar] [CrossRef] [Green Version]
  58. Los, M.; Van de Craen, M.; Penning, L.C.; Schenk, H.; Westendorp, M.; Baeuerle, P.A.; Droge, W.; Krammer, P.H.; Fiers, W.; Schulze-Osthoff, K. Requirement of an ICE/CED-3 protease for Fas/APO-1-mediated apoptosis. Nature 1995, 375, 81–83. [Google Scholar] [CrossRef] [Green Version]
  59. Sabbatini, P.; Han, J.; Chiou, S.K.; Nicholson, D.W.; White, E. Interleukin 1 beta converting enzyme-like proteases are essential for p53-mediated transcriptionally dependent apoptosis. Cell Growth Differ. 1997, 8, 643–653. [Google Scholar]
  60. Sattler, M.; Liang, H.; Nettesheim, D.; Meadows, R.P.; Harlan, J.E.; Eberstadt, M.; Yoon, H.S.; Shuker, S.B.; Chang, B.S.; Minn, A.J.; et al. Structure of Bcl-xL-Bak peptide complex: Recognition between regulators of apoptosis. Science 1997, 275, 983–986. [Google Scholar] [CrossRef]
  61. Liu, X.; Dai, S.; Zhu, Y.; Marrack, P.; Kappler, J.W. The structure of a Bcl-xL/Bim fragment complex: Implications for Bim function. Immunity 2003, 19, 341–352. [Google Scholar] [CrossRef]
  62. Feng, W.; Huang, S.; Wu, H.; Zhang, M. Molecular basis of Bcl-xL’s target recognition versatility revealed by the structure of Bcl-xL in complex with the BH3 domain of Beclin-1. J. Mol. Biol. 2007, 372, 223–235. [Google Scholar] [CrossRef]
  63. Czabotar, P.E.; Lessene, G.; Strasser, A.; Adams, J.M. Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat. Rev. Mol. Cell Biol. 2014, 15, 49–63. [Google Scholar] [CrossRef]
  64. Du, H.; Wolf, J.; Schafer, B.; Moldoveanu, T.; Chipuk, J.E.; Kuwana, T. BH3 domains other than Bim and Bid can directly activate Bax/Bak. J. Biol. Chem. 2011, 286, 491–501. [Google Scholar] [CrossRef]
  65. Hockings, C.; Anwari, K.; Ninnis, R.L.; Brouwer, J.; O’Hely, M.; Evangelista, M.; Hinds, M.G.; Czabotar, P.E.; Lee, E.F.; Fairlie, W.D.; et al. Bid chimeras indicate that most BH3-only proteins can directly activate Bak and Bax, and show no preference for Bak versus Bax. Cell Death Dis. 2015, 6, e1735. [Google Scholar] [CrossRef]
  66. Vela, L.; Gonzalo, O.; Naval, J.; Marzo, I. Direct interaction of Bax and Bak proteins with Bcl-2 homology domain 3 (BH3)-only proteins in living cells revealed by fluorescence complementation. J. Biol. Chem. 2013, 288, 4935–4946. [Google Scholar] [CrossRef]
  67. Willis, S.N.; Chen, L.; Dewson, G.; Wei, A.; Naik, E.; Fletcher, J.I.; Adams, J.M.; Huang, D.C. Proapoptotic Bak is sequestered by Mcl-1 and Bcl-xL, but not Bcl-2, until displaced by BH3-only proteins. Genes Dev. 2005, 19, 1294–1305. [Google Scholar] [CrossRef] [Green Version]
  68. Willis, S.N.; Fletcher, J.I.; Kaufmann, T.; van Delft, M.F.; Chen, L.; Czabotar, P.E.; Ierino, H.; Lee, E.F.; Fairlie, W.D.; Bouillet, P.; et al. Apoptosis initiated when BH3 ligands engage multiple Bcl-2 homologs, not Bax or Bak. Science 2007, 315, 856–859. [Google Scholar] [CrossRef]
  69. O’Neill, K.L.; Huang, K.; Zhang, J.; Chen, Y.; Luo, X. Inactivation of prosurvival Bcl-2 proteins activates Bax/Bak through the outer mitochondrial membrane. Genes Dev. 2016, 30, 973–988. [Google Scholar] [CrossRef] [Green Version]
  70. Zong, W.X.; Li, C.; Hatzivassiliou, G.; Lindsten, T.; Yu, Q.C.; Yuan, J.; Thompson, C.B. Bax and Bak can localize to the endoplasmic reticulum to initiate apoptosis. J. Cell Biol. 2003, 162, 59–69. [Google Scholar] [CrossRef] [Green Version]
  71. Thomenius, M.J.; Distelhorst, C.W. Bcl-2 on the endoplasmic reticulum: Protecting the mitochondria from a distance. J. Cell Sci. 2003, 116 Pt 22, 4493–4499. [Google Scholar] [CrossRef]
  72. Edlich, F.; Banerjee, S.; Suzuki, M.; Cleland, M.M.; Arnoult, D.; Wang, C.; Neutzner, A.; Tjandra, N.; Youle, R.J. Bcl-x(L) retrotranslocates Bax from the mitochondria into the cytosol. Cell 2011, 145, 104–116. [Google Scholar] [CrossRef]
  73. Ruefli-Brasse, A.; Reed, J.C. Therapeutics targeting Bcl-2 in hematological malignancies. Biochem. J. 2017, 474, 3643–3657. [Google Scholar] [CrossRef]
  74. Tang, C.; Willingham, M.C.; Reed, J.C.; Miyashita, T.; Ray, S.; Ponnathpur, V.; Huang, Y.; Mahoney, M.E.; Bullock, G.; Bhalla, K. High levels of p26BCL-2 oncoprotein retard taxol-induced apoptosis in human pre-B leukemia cells. Leukemia 1994, 8, 1960–1969. [Google Scholar]
  75. Thomadaki, H.; Floros, K.V.; Scorilas, A. Molecular response of HL-60 cells to mitotic inhibitors vincristine and taxol visualized with apoptosis-related gene expressions, including the new member BCL2L12. Ann. N. Y. Acad. Sci. 2009, 1171, 276–283. [Google Scholar] [CrossRef]
  76. Ibrado, A.M.; Liu, L.; Bhalla, K. Bcl-xL overexpression inhibits progression of molecular events leading to paclitaxel-induced apoptosis of human acute myeloid leukemia HL-60 cells. Cancer Res. 1997, 57, 1109–1115. [Google Scholar]
  77. Huang, S.; Tang, R.; Poon, R.Y. BCL-W is a regulator of microtubule inhibitor-induced mitotic cell death. Oncotarget 2016, 7, 38718–38730. [Google Scholar] [CrossRef]
  78. Xia, L.; Wurmbach, E.; Waxman, S.; Jing, Y. Upregulation of Bfl-1/A1 in leukemia cells undergoing differentiation by all-trans retinoic acid treatment attenuates chemotherapeutic agent-induced apoptosis. Leukemia 2006, 20, 1009–1016. [Google Scholar] [CrossRef] [Green Version]
  79. Leverson, J.D.; Zhang, H.; Chen, J.; Tahir, S.K.; Phillips, D.C.; Xue, J.; Nimmer, P.; Jin, S.; Smith, M.; Xiao, Y.; et al. Potent and selective small-molecule MCL-1 inhibitors demonstrate on-target cancer cell killing activity as single agents and in combination with ABT-263 (navitoclax). Cell Death Dis. 2015, 6, e1590. [Google Scholar] [CrossRef]
  80. Puhr, M.; Hoefer, J.; Neuwirt, H.; Eder, I.E.; Kern, J.; Schafer, G.; Geley, S.; Heidegger, I.; Klocker, H.; Culig, Z. PIAS1 is a crucial factor for prostate cancer cell survival and a valid target in docetaxel resistant cells. Oncotarget 2014, 5, 12043–12056. [Google Scholar] [CrossRef] [Green Version]
  81. Cui, J.; Placzek, W.J. PTBP1 modulation of MCL1 expression regulates cellular apoptosis induced by antitubulin chemotherapeutics. Cell Death Differ. 2016, 23, 1681–1690. [Google Scholar] [CrossRef] [Green Version]
  82. Chu, R.; Alford, S.E.; Hart, K.; Kothari, A.; Mackintosh, S.G.; Kovak, M.R.; Chambers, T.C. Mitotic arrest-induced phosphorylation of Mcl-1 revisited using two-dimensional gel electrophoresis and phosphoproteomics: Nine phosphorylation sites identified. Oncotarget 2016, 7, 78958–78970. [Google Scholar] [CrossRef]
  83. Eichhorn, J.M.; Sakurikar, N.; Alford, S.E.; Chu, R.; Chambers, T.C. Critical role of anti-apoptotic Bcl-2 protein phosphorylation in mitotic death. Cell Death Dis. 2013, 4, e834. [Google Scholar] [CrossRef] [PubMed]
  84. Kruczynski, A.; Etievant, C.; Perrin, D.; Chansard, N.; Duflos, A.; Hill, B.T. Characterization of cell death induced by vinflunine, the most recent Vinca alkaloid in clinical development. Br. J. Cancer 2002, 86, 143–150. [Google Scholar] [CrossRef] [Green Version]
  85. Rodi, D.J.; Janes, R.W.; Sanganee, H.J.; Holton, R.A.; Wallace, B.A.; Makowski, L. Screening of a library of phage-displayed peptides identifies human bcl-2 as a taxol-binding protein. J. Mol. Biol. 1999, 285, 197–203. [Google Scholar] [CrossRef]
  86. Rodi, D.J.; Makowski, L. Similarity between the sequences of taxol-selected peptides and the disordered loop of the anti-apoptotic protein, Bcl-2. Pac. Symp. Biocomput. 1999, 4, 532–541. [Google Scholar]
  87. Ferlini, C.; Cicchillitti, L.; Raspaglio, G.; Bartollino, S.; Cimitan, S.; Bertucci, C.; Mozzetti, S.; Gallo, D.; Persico, M.; Fattorusso, C.; et al. Paclitaxel directly binds to Bcl-2 and functionally mimics activity of Nur77. Cancer Res. 2009, 69, 6906–6914. [Google Scholar] [CrossRef] [PubMed]
  88. Knipling, L.; Wolff, J. Direct interaction of Bcl-2 proteins with tubulin. Biochem. Biophys. Res. Commun. 2006, 341, 433–439. [Google Scholar] [CrossRef] [PubMed]
  89. Esteve, M.A.; Carre, M.; Bourgarel-Rey, V.; Kruczynski, A.; Raspaglio, G.; Ferlini, C.; Braguer, D. Bcl-2 down-regulation and tubulin subtype composition are involved in resistance of ovarian cancer cells to vinflunine. Mol. Cancer Ther. 2006, 5, 2824–2833. [Google Scholar] [CrossRef] [Green Version]
  90. Bennett, A.; Sloss, O.; Topham, C.; Nelson, L.; Tighe, A.; Taylor, S.S. Inhibition of Bcl-xL sensitizes cells to mitotic blockers, but not mitotic drivers. Open Biol. 2016, 6, 160134. [Google Scholar] [CrossRef] [Green Version]
  91. Sloss, O.; Topham, C.; Diez, M.; Taylor, S. Mcl-1 dynamics influence mitotic slippage and death in mitosis. Oncotarget 2016, 7, 5176–5192. [Google Scholar] [CrossRef] [Green Version]
  92. Millman, S.E.; Pagano, M. MCL1 meets its end during mitotic arrest. EMBO Rep. 2011, 12, 384–385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Jackson II, R.S.; Placzek, W.; Fernandez, A.; Ziaee, S.; Chu, C.Y.; Wei, J.; Stebbins, J.; Kitada, S.; Fritz, G.; Reed, J.C.; et al. Sabutoclax, a Mcl-1 antagonist, inhibits tumorigenesis in transgenic mouse and human xenograft models of prostate cancer. Neoplasia 2012, 14, 656–665. [Google Scholar] [CrossRef]
  94. Rapino, F.; Naumann, I.; Fulda, S. Bortezomib antagonizes microtubule-interfering drug-induced apoptosis by inhibiting G2/M transition and MCL-1 degradation. Cell Death Dis. 2013, 4, e925. [Google Scholar] [CrossRef]
  95. Souers, A.J.; Leverson, J.D.; Boghaert, E.R.; Ackler, S.L.; Catron, N.D.; Chen, J.; Dayton, B.D.; Ding, H.; Enschede, S.H.; Fairbrother, W.J.; et al. ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat. Med. 2013, 19, 202–208. [Google Scholar] [CrossRef]
  96. Oltersdorf, T.; Elmore, S.W.; Shoemaker, A.R.; Armstrong, R.C.; Augeri, D.J.; Belli, B.A.; Bruncko, M.; Deckwerth, T.L.; Dinges, J.; Hajduk, P.J.; et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature 2005, 435, 677–681. [Google Scholar] [CrossRef] [PubMed]
  97. Tse, C.; Shoemaker, A.R.; Adickes, J.; Anderson, M.G.; Chen, J.; Jin, S.; Johnson, E.F.; Marsh, K.C.; Mitten, M.J.; Nimmer, P.; et al. ABT-263: A potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 2008, 68, 3421–3428. [Google Scholar] [CrossRef] [PubMed]
  98. Wei, J.; Stebbins, J.L.; Kitada, S.; Dash, R.; Placzek, W.; Rega, M.F.; Wu, B.; Cellitti, J.; Zhai, D.; Yang, L.; et al. BI-97C1, an optically pure Apogossypol derivative as pan-active inhibitor of antiapoptotic B-cell lymphoma/leukemia-2 (Bcl-2) family proteins. J. Med. Chem. 2010, 53, 4166–4176. [Google Scholar] [CrossRef]
  99. Billard, C. BH3 mimetics: Status of the field and new developments. Mol. Cancer Ther. 2013, 12, 1691–1700. [Google Scholar] [CrossRef]
  100. Kutuk, O.; Letai, A. Alteration of the mitochondrial apoptotic pathway is key to acquired paclitaxel resistance and can be reversed by ABT-737. Cancer Res. 2008, 68, 7985–7994. [Google Scholar] [CrossRef] [Green Version]
  101. Parrondo, R.; de Las Pozas, A.; Reiner, T.; Perez-Stable, C. ABT-737, a small molecule Bcl-2/Bcl-xL antagonist, increases antimitotic-mediated apoptosis in human prostate cancer cells. PeerJ 2013, 1, e144. [Google Scholar] [CrossRef]
  102. Watanabe, M.; Umezawa, K.; Higashihara, M.; Horie, R. Combined inhibition of NF-kappaB and Bcl-2 triggers synergistic reduction of viability and induces apoptosis in melanoma cells. Oncol. Res. 2013, 21, 173–180. [Google Scholar] [CrossRef]
  103. Lieber, J.; Eicher, C.; Wenz, J.; Kirchner, B.; Warmann, S.W.; Fuchs, J.; Armeanu-Ebinger, S. The BH3 mimetic ABT-737 increases treatment efficiency of paclitaxel against hepatoblastoma. BMC Cancer 2011, 11, 362. [Google Scholar] [CrossRef]
  104. Bah, N.; Maillet, L.; Ryan, J.; Dubreil, S.; Gautier, F.; Letai, A.; Juin, P.; Barille-Nion, S. Bcl-xL controls a switch between cell death modes during mitotic arrest. Cell Death Dis. 2014, 5, e1291. [Google Scholar] [CrossRef]
  105. Wang, C.; Huang, S.B.; Yang, M.C.; Lin, Y.T.; Chu, I.H.; Shen, Y.N.; Chiu, Y.H.; Hung, S.H.; Kang, L.; Hong, Y.R.; et al. Combining paclitaxel with ABT-263 has a synergistic effect on paclitaxel resistant prostate cancer cells. PLoS ONE 2015, 10, e0120913. [Google Scholar] [CrossRef]
  106. Vogler, M.; Walter, H.S.; Dyer, M.J.S. Targeting anti-apoptotic BCL2 family proteins in haematological malignancies - from pathogenesis to treatment. Br. J. Haematol. 2017, 178, 364–379. [Google Scholar] [CrossRef]
  107. Kotschy, A.; Szlavik, Z.; Murray, J.; Davidson, J.; Maragno, A.L.; Le Toumelin-Braizat, G.; Chanrion, M.; Kelly, G.L.; Gong, J.N.; Moujalled, D.M.; et al. The MCL1 inhibitor S63845 is tolerable and effective in diverse cancer models. Nature 2016, 538, 477–482. [Google Scholar] [CrossRef]
  108. Merino, D.; Whittle, J.R.; Vaillant, F.; Serrano, A.; Gong, J.N.; Giner, G.; Maragno, A.L.; Chanrion, M.; Schneider, E.; Pal, B.; et al. 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. 2017, 9, 401. [Google Scholar] [CrossRef]
  109. Ackler, S.; Mitten, M.J.; Foster, K.; Oleksijew, A.; Refici, M.; Tahir, S.K.; Xiao, Y.; Tse, C.; Frost, D.J.; Fesik, S.W.; et al. The Bcl-2 inhibitor ABT-263 enhances the response of multiple chemotherapeutic regimens in hematologic tumors in vivo. Cancer Chemother. Pharmacol. 2010, 66, 869–880. [Google Scholar] [CrossRef]
  110. Dai, H.; Ding, H.; Meng, X.W.; Lee, S.H.; Schneider, P.A.; Kaufmann, S.H. Contribution of Bcl-2 phosphorylation to Bak binding and drug resistance. Cancer Res. 2013, 73, 6998–7008. [Google Scholar] [CrossRef]
  111. Castro, J.E.; Loria, O.J.; Aguillon, R.A.; James, D.; Llanos, C.A.; Rassenti, L.; Wood, B.A.; Homlund, J.T.; Kipps, T.J. A phase II, open label study of AT-101 in combination with rituximab in patients with relapsed or refractory chronic lymphocytic leukemia. Evaluation of two dose regimens. Blood 2007, 110, 3119. [Google Scholar]
  112. Cakar, B.; Gursoy, P.; Atmaca, H.; Kisim, A.; Bozhurt, E.; Uzunoglu, S.; Sezgin, C.; Sanli, U.A.; Karabulut, B.; Uslu, R.; et al. Paclitaxel in combination with AT-101 induces apoptosis via supressing Bcl-2, bcl-XL, mcl-1 proteins in human breast cancer cells. J. Clin. Oncol. 2017, 31 (Suppl. 15), e13578. [Google Scholar]
  113. Caenepeel, S.; Brown, S.P.; Belmontes, B.; Moody, G.; Keegan, K.S.; Chui, D.; Whittington, D.A.; Huang, X.; Poppe, L.; Cheng, A.C.; et al. AMG 176, a selective MCL1 inhibitor, is effective in hematologic cancer models alone and in combiantion with established therapies. Cancer Discov. 2018, 8, 1582–1597. [Google Scholar]
  114. Tron, A.E.; Belmonte, M.A.; Adam, A.; Aquila, B.M.; Boise, L.H.; Chiarparin, E.; Cidado, J.; Embrey, K.J.; Gangl, E.; Gibbons, F.D.; et al. Discovery of Mcl-1-specific inhibitor AZD5991 and preclinical activity in multiple myeloma and acute myeloid leukemia. Nat. Commun. 2018, 9, 5341. [Google Scholar] [CrossRef]
Figure 1. Microtubule composition, structure, and microtubule targeting agents (MTA) binding sites. (A) Cartoon of microtubule consisting of 13 protofilaments formed from alpha (blue) and beta (brown) tubulin heterodimers. Vinca alkaloids (yellow) bind to the + end of the microtubule and taxanes (pink) bind inside the lumen (lower left). (B) Space Filling model an of alpha (Blue) beta (Brown) heterodimer with vinca alkaloid binding site (yellow) and taxane binding site (pink) located on beta tubulin [PDB: 3J8Y].
Figure 1. Microtubule composition, structure, and microtubule targeting agents (MTA) binding sites. (A) Cartoon of microtubule consisting of 13 protofilaments formed from alpha (blue) and beta (brown) tubulin heterodimers. Vinca alkaloids (yellow) bind to the + end of the microtubule and taxanes (pink) bind inside the lumen (lower left). (B) Space Filling model an of alpha (Blue) beta (Brown) heterodimer with vinca alkaloid binding site (yellow) and taxane binding site (pink) located on beta tubulin [PDB: 3J8Y].
Cells 08 00346 g001
Figure 2. BCL2 Family Structure, Function, and Equilibrium. (A) Surface structure of MCL1 (grey) with BH3 binding pocket, representative of the anti-apoptotic BCL2 family, bound to BIM BH3 Only protein (Black) with highlighted residues: Hydrophobic (Pink), conserved Glycine (red), and invariant Aspartic Acid (Yellow). [PDB: 2NL9] (B) BH3 Sequence homology between BCL2 effectors and selected BH3 Only proteins. (C) BCL2 Family Interactions Balance Cell Stress Signaling, where BCL2 family stoichiometry and affinity dictate cell fate.
Figure 2. BCL2 Family Structure, Function, and Equilibrium. (A) Surface structure of MCL1 (grey) with BH3 binding pocket, representative of the anti-apoptotic BCL2 family, bound to BIM BH3 Only protein (Black) with highlighted residues: Hydrophobic (Pink), conserved Glycine (red), and invariant Aspartic Acid (Yellow). [PDB: 2NL9] (B) BH3 Sequence homology between BCL2 effectors and selected BH3 Only proteins. (C) BCL2 Family Interactions Balance Cell Stress Signaling, where BCL2 family stoichiometry and affinity dictate cell fate.
Cells 08 00346 g002
Figure 3. Mechanism of MTAs and BH3 Mimetics on BCL2 Family Signaling. MTAs induce cellular stress that leads to upregulation of pro-apoptotic BCL2 family proteins and ultimately apoptosis. Cancer cells can overcome this through upregulation of anti-apoptotic BCL2 family proteins. BH3 mimetics directly target the anti-apoptotic BCL2 family members to inhibit their suppression of BH3-only activators and/or of BAK/BAX oligomerization.
Figure 3. Mechanism of MTAs and BH3 Mimetics on BCL2 Family Signaling. MTAs induce cellular stress that leads to upregulation of pro-apoptotic BCL2 family proteins and ultimately apoptosis. Cancer cells can overcome this through upregulation of anti-apoptotic BCL2 family proteins. BH3 mimetics directly target the anti-apoptotic BCL2 family members to inhibit their suppression of BH3-only activators and/or of BAK/BAX oligomerization.
Cells 08 00346 g003
Table 1. MTA Efficacy and sensitivity within the context of the anti-apoptotic BCL2 proteins.
Table 1. MTA Efficacy and sensitivity within the context of the anti-apoptotic BCL2 proteins.
MTAProteinCell Type/ModelEffectReference
PaclitaxelBCL2Breast, Lung, Prostate* Presence sensitizes [37,39]
BCL2LeukemiaPresence induces resistance; paclitaxel decreases BCL2 mRNA expression[74,75]
BCLxLLeukemia, ColonUpregulation induces resistance, inhibition sensitizes [76]
BCLWLeukemiaKnockdown/out sensitizes [77]
BFL1/A1LeukemiaUpregulation induces resistance[78]
DocetaxelBCLxLLung, MyelomaInhibition sensitizes[79]
MCL1Prostate* Inhibition or downregulation sensitizes[80,81]
VincristineBCL2LeukemiaTreatment decreases BCL2 mRNA expression[75]
VinblastineMCL1HeLaTreatment decreases MCL1 protein levels[82,83]
VinflunineBFL1/A1LymphomaIncrease linked with resistance[84]
* Denotes studies that were performed in both cell lines and mouse xenograft models.
Table 2. BCL2 family inhibitors and their effect in combination with MTAs.
Table 2. BCL2 family inhibitors and their effect in combination with MTAs.
BH3 MimeticMCL1BFL1/A1BCLWBCLxLBCL2Cell Type/ModelEffect Reference
ABT-737 XXXMelanoma, Breast, Prostate, LiverSensitizes cells to paclitaxel or docetaxel[100,102,103]
ABT-263 XXXProstateAdditive with vincristine; synergy with paclitaxel[104,109]
ABT-199 XLeukemia, LymphomaApproved for CLL; sensitizes cells to paclitaxel[95,110]
WEHI-539 X ColonSensitizes to Paclitaxel[90]
SabutoclaxXXXXXProstateSensitizes to docetaxel[81,93]
AT-101X XXXBreastSynergizes with paclitaxel[111,112]
S63845X BreastSynergizes with docetaxel[107,108]
A1210477X [79]
AMG-176X [113]
AZD-5991X [114]

Share and Cite

MDPI and ACS Style

Whitaker, R.H.; Placzek, W.J. Regulating the BCL2 Family to Improve Sensitivity to Microtubule Targeting Agents. Cells 2019, 8, 346. https://doi.org/10.3390/cells8040346

AMA Style

Whitaker RH, Placzek WJ. Regulating the BCL2 Family to Improve Sensitivity to Microtubule Targeting Agents. Cells. 2019; 8(4):346. https://doi.org/10.3390/cells8040346

Chicago/Turabian Style

Whitaker, Robert H., and William J. Placzek. 2019. "Regulating the BCL2 Family to Improve Sensitivity to Microtubule Targeting Agents" Cells 8, no. 4: 346. https://doi.org/10.3390/cells8040346

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop