Next Article in Journal
SonoVue® vs. Sonazoid™ vs. Optison™: Which Bubble Is Best for Low-Intensity Sonoporation of Pancreatic Ductal Adenocarcinoma?
Next Article in Special Issue
Impact of circ-0000221 in the Pathogenesis of Hepatocellular via Modulation of miR-661–PTPN11 mRNA Axis
Previous Article in Journal
Purification Process of a Recombinant Human Follicle Stimulating Hormone Biosimilar (Primapur®) to Yield a Pharmaceutical Product with High Batch-to-Batch Consistency
Previous Article in Special Issue
CpG Oligodeoxynucleotides for Anticancer Monotherapy from Preclinical Stages to Clinical Trials
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Making Sense of Antisense Oligonucleotide Therapeutics Targeting Bcl-2

Bio-Path Holdings, Inc., Bellaire, TX 77401, USA
*
Author to whom correspondence should be addressed.
Pharmaceutics 2022, 14(1), 97; https://doi.org/10.3390/pharmaceutics14010097
Submission received: 19 November 2021 / Revised: 17 December 2021 / Accepted: 28 December 2021 / Published: 1 January 2022

Abstract

:
The B-cell lymphoma 2 (Bcl-2) family, comprised of pro- and anti-apoptotic proteins, regulates the delicate balance between programmed cell death and cell survival. The Bcl-2 family is essential in the maintenance of tissue homeostasis, but also a key culprit in tumorigenesis. Anti-apoptotic Bcl-2, the founding member of this family, was discovered due to its dysregulated expression in non-Hodgkin’s lymphoma. Bcl-2 is a central protagonist in a wide range of human cancers, promoting cell survival, angiogenesis and chemotherapy resistance; this has prompted the development of Bcl-2-targeting drugs. Antisense oligonucleotides (ASO) are highly specific nucleic acid polymers used to modulate target gene expression. Over the past 25 years several Bcl-2 ASO have been developed in preclinical studies and explored in clinical trials. This review will describe the history and development of Bcl-2-targeted ASO; from initial attempts, optimizations, clinical trials undertaken and the promising candidates at hand.

1. Introduction

The Bcl-2 family of proteins regulates apoptosis, a form of programmed cell death. The family members are highly conserved and have important roles during development and adult tissue homeostasis [1,2,3]. Members of this family, related only by their conserved Bcl-2 homology (BH) domains, are divided into two opposing groups: pro-apoptotic vs. anti-apoptotic [4].
Bcl-2 is the founding member of this family and has anti-apoptotic function. Like other anti-apoptotic members (Bcl-xL, Mcl-1, Bcl-W, Bfl-1, Bcl-b), Bcl-2 has 4 BH domains (BH1 to BH4) (Figure 1A). These domains form a hydrophobic cleft that acts as the binding site for the BH3 domains of their pro-apoptotic counterparts, an interaction which impedes the latter’s function. Pro-apoptotic members are classified as either effectors or initiators of apoptosis. Effectors, Bax and Bak, use their BH domains (BH1 to BH3) to oligomerize and form pores on the outer mitochondrial membrane [5]. Initiators, also known as ‘BH3-only’ proteins, fall into two categories: activators (tBID, Bim, Puma) directly bind to and activate Bax and Bak, whilst sensitizers (Bad, Bik, Bmf, Noxa) bind to anti-apoptotic proteins to disrupt their sequestration of BH3 activators and interactions with Bax and Bak [4,5] (Figure 1A).
The intrinsic apoptotic pathway involves the permeabilization of the outer mitochondrial membrane, and release of cytochrome C and caspase activating proteins via the pores formed by Bax and Bak. Apoptotic stimuli, such as oxidative stress, calcium flux, ER stress and DNA damage, disrupt the steady-state interactions between pro- and anti-apoptotic proteins that are normally kept in fine equilibrium [6]. It is the relative changes in protein abundance and affinities that ultimately determine cell fate [7] (Figure 1B).
Cancer cells advantageously manipulate the apoptotic machinery to resist cell death [8]. Bcl-2 itself was discovered due to its overexpression in B cell lymphomas as a result of chromosomal translocation t(14;18), in which the bcl-2 gene on chromosome 18 fuses to the immunoglobulin heavy-chain (IgH) gene on chromosome 14 and is expressed under the control of the IgH enhancer [9,10,11]. Over the past 25 years in addition to hematological malignancies, elevated Bcl-2 expression and function has been observed in various tumors, including melanoma [12], breast cancer [13,14], colorectal cancer [15] and lung cancer [16]. Mechanisms of overexpression include gene amplification [17], somatic mutations at the bcl-2 promoter [18], transcriptional upregulation [19] and loss of microRNA (miRNA) that negatively regulate Bcl-2 post-transcriptionally [20].
Beyond evading cell death, the BH4 domain of Bcl-2 gives it the ability to interact with non-apoptotic proteins and take part in other cellular processes [21]. Angiogenesis is an important part of tumor growth and metastasis. Solid tumors rely on the formation of blood vessels for continued supply of nutrients, oxygen and as a route in the metastatic pathway [22]. Vascular endothelial growth factor (VEGF) is a key mediator of angiogenesis that promotes the differentiation, proliferation and migration of endothelial cells [23,24,25]. VEGF and Bcl-2 have a synergistic relationship [24,26]. Both in vitro and in vivo studies have shown that elevated Bcl-2 level in cancer cells under hypoxia condition induces VEGF expression, enhances its mRNA stability, promotes its secretion and its transcriptional activity, resulting in increased vascularization that is independent of cell survival [24,27]. Overexpression of Bcl-2 lacking the BH4 domain has no effect on VEGF expression/activity, making the BH4 domain the key region for Bcl-2’s crosstalk with angiogenesis [27,28]. The BH4 domain has also been shown to play a part in the upregulation of NF-κB transcriptional activity through Raf-1/MEKK1-mediated activation of IKKβ [14,29,30]. NF-κB is an important regulator of epithelial-mesenchymal genes (N-cadherin, vimentin) and matrix metalloproteinase (mmp-9, mmp-2), all required for cancer metastasis [29,31,32]. Similarly, under hypoxia conditions Bcl-2 acts as a cofactor that facilitates nuclear transport of transcription factor twist1; once again encouraging epithelial-mesenchymal transition, angiogenesis and tumor development [25].
Given its multifunctional and dynamic roles in tumorigenesis, it is not surprising that Bcl-2 has been a popular target in cancer drug design. Although Bcl-2 has crucial functions in non-cancerous cells, therapies targeting Bcl-2 have a predominant effect on tumor cells. This could be due to the fact that tumor cells are ‘primed’ for apoptosis as a result of DNA damage, hyper proliferation and aberrant oncogenic signaling [33,34]. Normal cells are not primed for apoptosis so a decrease in Bcl-2 expression does not necessarily result in cell death. Indeed, many adult tissues, such as the heart, brain, kidneys and liver are refractory to apoptosis [35]. Hematopoietic cells are not refractory to apoptosis and express varying levels of Bcl-2 depending on their population, developmental stage and activation [36]. Yet, studies had shown that inhibition of Bcl-2 using ABT-737, a BH3 mimetic, significantly reduced the viability of multiple myeloma cells with no effect on normal peripheral blood mononuclear cells [37]. It is possible that the ratio between pro- and anti-apoptotic proteins and the specific dependency on Bcl-2 for cancer cell survival permits the establishment of a therapeutic index and safe targeting of Bcl-2.
Oligonucleotide therapeutics, specifically antisense oligonucleotides (ASO) therapeutics, are an eminent drug platform with advantages in terms of specificity, production, and delivery [38]. These short (18–25 bases in length), chemically modified nucleic acid polymers rely on Watson-Crick base pair binding to target mRNA, resulting in either gene silencing, steric hindrance, or alternative splicing [39]. There are currently six FDA approved ASO drugs [40]. These approved drugs fall into three generations of modified ASO, which display the optimization strategies used. First generation ASOs have a phosphorothioate (PS) rather than phosphodiester (PO) backbone, to increase cellular uptake and nuclease resistance [41]. Second generation ASOs have PS modified central regions with methyl or methoxyethyl modifications at the 2′ position of the sugar moieties at the 3′ and 5′ ends (2′-O-methyl (2′-OMe) or 2′-O-methoxyethyl (2′-MOE)). These gapmer structures further increase hybridization and nuclease resistance [42,43,44]. Similar to second generation, third generation ASOs also have a gapmer configuration but with more complex modification at the 3′ and 5′ ends. These include: (i) locked nucleic acids (LNA) in which a methylene bridge connects the 2′ oxygen and 4′ carbon of the riboses; (ii) phosphorodiamidate morpholino oligomers (PMOs) in which the ribose is replaced by morpholine moiety and nucleotides are connected via methylene phosphoramidate bonds; (iii) peptide nucleic acids (PNAs) in which the deoxyribose phosphate backbone is replaced by a pseudopeptide polymer [45,46].
In this review the history of ASO targeting Bcl-2 will be described. By recognizing their challenges and appreciating their optimization it becomes clear why ASO remain a promising Bcl-2 therapeutic.

2. The Beginning of Bcl-2 ASO Therapies

Identified as an oncoprotein in the mid-1980s [47] and as a mediator of chemotherapy resistance in the mid-1990s [48,49,50], Bcl-2 has high potential as a cancer therapeutic target. Since Bcl-2 is an intracellular protein with no enzymatic activity, Bcl-2 was not amenable to the conventional therapeutic approaches at that time, which consisted of antibodies targeting cell surface proteins and small molecules inhibiting enzyme activities [51]. This led to utilizing ASO technology to inhibit Bcl-2 expression. The evolution of ASO technology has been accompanied by increased efficacy in Bcl-2 regulation with changes in target sequence selection, polymer backbone modifications and delivery strategies.

2.1. Initial Bcl-2 ASOs In Vitro Studies

Initial studies exploring the ability of Bcl-2 ASO to inhibit leukemia cell survival used a 20-base pair oligonucleotide complementary to the transcription initiation site of Bcl-2 mRNA [52]. A comparison between oligonucleotides with PO vs. PS backbones, in the absence of delivery technology, revealed the latter to be more resistant to nucleases and 5–10 times more potent at inhibiting 697 cell (human pre-B-cell acute lymphocytic leukemia) proliferation [52]. Although both oligonucleotides initially suppressed cell proliferation via non-cytotoxic mechanisms, with inhibition of cell replication preceding cell death, their rate of cellular uptake and kinetics differed. In serum-free conditions, PO-ASO achieved maximum intracellular concentrations within two days after initial treatments and maximum cell inhibition within two to three days, whereas PS-ASO required four to five days to achieve comparable intracellular levels and four to seven days to achieve maximum inhibition [52]. Similar results were obtained in human leukemic myeloid cell lines: Daudi, KG1a and HL-60 [53]. Treatment of acute myeloid leukemia (AML) patient cells in the presence of serum with PS-ASO showed increased sensitivity to cytostatic drugs: daunorubicin or l-P-D-arabinofura-nosyl-cytosine (ara-C) [53], thereby establishing Bcl-2 PS-ASO as more clinically relevant.
The dependency of ASO’s effectiveness on which Bcl-2 region is targeted was investigated using 13 different ASO directed against Bcl-2 coding regions, 5′ or 3′ untranslated regions and translation initiation or termination sites. Amongst these, the ASO targeting codon 141–147 of Bcl-2 mRNA (ASO-2009) proved most cytotoxic in a human small cell lung cancer (SCLC) cell line, SW2 [54]. Contrary to the PS-ASO described above, ASO 2009’s inhibitory activity was apparent after only two days. This could be due to its facilitated intracellular delivery with cationic lipids [54]. Combination treatments of ASO-2009 with etoposide, doxorubicin or cisplatin in SCLC cell lines displayed synergistic cytotoxicity, making them a novel treatment strategy [55].

2.2. Bcl-2/Bcl-xL-Bispecific ASO

In addition to Bcl-2, upregulation of anti-apoptotic Bcl-xL is also common in various cancers and is associated with resistance to therapeutics [56,57,58,59]. While being functionally redundant and structurally similar to Bcl-2, Bcl-xL may protect cells from different apoptotic stimuli [60]. Cancer cells can take advantage of this and use Bcl-xL as an alternative to, or in conjunction with, Bcl-2 to survive [61,62]. Upon identifying a region of high sequence homology in the Bcl-2 and Bcl-xL mRNAs (nucleotides 605–624 and 687–706, respectively, which differs by only three nucleotides), three bi-specific ASOs targeting Bcl-2 and Bcl-xL were designed to simultaneously inhibit both survival factors. These 20-base pair ASO are classified as second generation with 2′-MOE modification at their ends and hold a maximum of three mismatching bases [63]. Analysis in SCLC and non-small cell lung cancer (NSCLC) cell lines showed that ASO-4625, which has no mismatching nucleotides to Bcl-2 but three mismatching nucleotides to Bcl-xL, had the strongest bispecific activity, inhibiting the expression of both proteins and inducing cell death [63]. Such activity was also confirmed in breast, colorectal, prostate and malignant melanoma cells [58,59]. In vivo studies proved ASO-4625’s therapeutic potential, with a 51% reduction of breast cancer (MDA-MB-231) xenografts and 59% reduction in colorectal cancer (Colo-320) xenografts after a 3-week treatment period [64]. In addition to restoring apoptosis, treatment of melanoma cells with ASO-4625 led to a 50–60% reduction in VEGF secretion and a 10–12 fold decrease in the vascularization of matrigel plugs subcutaneously injected into mice; reiterating Bcl-2’s relevance in angiogenesis and implicating Bcl-xL’s role in it [65]. ASO-4625 also markedly increased the sensitivity of prostate cancer cells to mitoxantrone, docetaxel and paclitaxel, suggesting its use in combination treatments [66,67].
To optimize the binding of ASO-4625 to Bcl-xL and overcome the potential destabilization due to mismatches, an isosequential analog with LNA, ASO-5005, was developed, [68]. ASO-5005 simultaneously downregulated Bcl-2 and Bcl-xL expression and reduced cell viability more effectively than ASO-4625 [68]. Combination treatments of ASO-5005 with doxorubicin or paclitaxel in MDA-MB-231 cells and cisplatin, paclitaxel, or gemcitabine in H125 cells (NSCLC) were stronger than those of chemotherapies alone or in combination with ASO-4625 [68]. Unfortunately, the investigators for ASO-4625 and ASO-5005 were not able to identify suitable pharmaceutical/business partners to help develop these drug candidates. Despite these encouraging results the above ASO did not progress beyond in vitro and in vivo preclinical studies.

2.3. Oblimersen

Oblimersen (also known as: G3139, Genasense, Oblimersen sodium; from Genta Inc., Berkeley Heights, NJ, USA) was the first Bcl-2 ASO to enter clinical trials [69,70]. It obtained orphan drug status in both the US and EU in 2001 for chronic lymphocytic leukemia (CLL) and multiple myeloma [71]. This 18-base pair PS-ASO (5′-TCT CCC AGC GTG CGC CAT-3′) binds to the first six codons of the Bcl-2 mRNA [72]. Preclinical evidence indicated that Oblimersen enhanced the in vivo efficacy of chemotherapeutic agents against hematological malignancies and solid tumors. In a lymphoma xenograft model, Klasa et al. showed that the median survival of SCID mice inoculated with 5 million DoHH2 lymphoma cells was 44 days, with 0% of mice surviving beyond 90 days [73]. In this model, the median survival of mice administered with 5 mg/kg of Oblimersen or 35 mg/kg cyclophosphamide were 79 days and 47 days, respectively; percentages of mice administered with Oblimersen or cyclophosphamide that survived >90 days were 48% and 0%, respectively. The median survival of mice treated with the Oblimersen and cyclophosphamide combination was not obtained as 61% of mice survived >90 days [73]. Similarly, Oblimersen (5 mg/kg) was reported to enhance docetaxel (20 mg/kg) efficacy in a preclinical H-460 lung cancer model [74]. Tumor growth delay by Oblimersen, docetaxel, and combination were 5 days, 13 days, and 21 days, respectively. Four of seven mice achieved complete responses with the combination treatment, whereas no complete response was observed in mice treated with Oblimersen or docetaxel alone [74].
Following its successful preclinical antitumor activity, Oblimersen has since been in 45 clinical trials (https://clinicaltrials.gov/ last accessed 30 October 2021). Since minimal clinical activity was observed in Oblimersen single agent phase I/II studies [75,76,77,78], subsequent phase III clinical trials for patients with hematological malignancies or melanoma, involve Oblimersen in combination with other chemotherapeutic drugs such as dacarbazine [79], dexamethasone [80], cyclophosphamide [80] and fludarabine [81]. An earlier phase I study reported that addition of Oblimersen to standard cytarabine/daunorubicin chemotherapy led to complete remission (CR) in 14 of 29 untreated AML patients over the age of 60 [82]. Among 13 patients assessable for both clinical response and Bcl-2 protein levels, approximately a 20% decrease in Bcl-2 protein levels was observed in patients with CR (n = 7) but no changes in Bcl-2 protein level in non-responders (n = 6). Although the number of samples was small, patients’ response appeared to correlate with baseline Bcl-2 levels; among 22 patients assessable for both clinical response and Bcl-2 mRNA levels, median number of Bcl-2 mRNA copies were higher in the CR patients (n = 12) than those in the non-responders (n = 10). These results led to the phase III Cancer and Leukemia Group B (CALGB) 10201 study in which the effect of adding Oblimersen to standard cytarabine/daunorubicin chemotherapy in previously untreated AML patients over the age of 60 was assessed and evaluated based on the rates of CR, disease free survival (DFS) and overall survival (OS). Of the 506 patients enrolled those that received combination treatment displayed no statistically significant improvement in CR, DFS or OS [83]. A subset analysis of patients with secondary AML, however, did display improved DFS with the addition of Oblimersen to their treatments [83]. It is not clear why Oblimersen combination treatment failed in the CALGB 10201 study. This might be due to the fact that baseline Bcl-2 levels were not used to stratify enrolled patients, target Bcl-2 inhibition was not sufficient, and/or other disease mechanisms that precluded Oblimersen from inducing a response. Retrospective subset analysis of a randomized phase III study showed that combining Oblimersen with dacarbazine significantly improved OS and progression-free survival (PFS) in chemotherapy-naïve patients with advanced melanoma and normal baseline serum lactate dehydrogenase (LDH) levels [79]. But a prospective, double-blind, placebo-controlled study did not confirm that addition of Oblimersen to dacarbazine could significantly improve OS or PFS in chemotherapy-naïve patients with advanced melanoma and low-normal LDH levels [84]. The authors noted that advanced melanoma patients with low-normal LDH levels might represent a population with a better chance of benefitting from systemic therapy, thus obscuring the earlier analysis and revealing the weakness of retrospective subgroup analysis of clinical studies. The inability of these combination treatments to affect OS, which is the primary outcome measure in these studies, puts the efficacy of Oblimersen in question and denies its FDA approval [85] (Figure 2).
The overall failure of Oblimersen in phase III trials may be due to instability, limited cellular uptake or intracellular compartmentalization. This has encouraged the optimization of Bcl-2 ASO structure and delivery.

3. Optimizations of Bcl-2 ASOs

3.1. SPC2996

SPC2996, developed by Santaris Pharma, is a 16 PS-base pair oligonucleotide with two LNA moieties at each end [86] (Figure 3A). It differs in sequence from Oblimersen by only 3 nucleotides [86]. Delivered to cells gynomically (i.e., without conjugates or transfectants), it was efficiently internalized by a range of cell lines (melanoma, lymphoma and fibrosarcoma) and was more potent than Oblimersen at reducing Bcl-2 expression [86]. CLL patients in a SPC2996 phase I/II clinical trial did not achieve significant clinical remission despite having a dose-dependent reduction in circulating CLL cells [87]. Treatment caused dose-dependent inflammatory reactions in 39% of patients, a side effect also seen in Oblimersen trials [87]. Transcriptomic profiling of peripheral blood samples revealed upregulation of 466 genes, many of which are involved in the activation and regulation of immune responses. Toll like receptor (TLR) early response genes such as IL-1β, MIP-1α and TNF-α were particularly overexpressed. These findings support the hypothesis that TLR9 is activated by the CpG motifs in the SPC2996 and Oblimersen sequences, explaining their paralleled and concomitant inflammatory effects [88,89].

3.2. PNT2258

Developed by ProNai therapeutics, now known as Serra Oncology, PNT2258 optimized the silencing of Bcl-2 in two ways: (i) the target sequence selection, and (ii) the delivery mechanism. Rather than hybridizing to Bcl-2 mRNA, PNT2258 adopted the DNA interference (DNAi) approach in which a non-coding regulatory region upstream of the transcription start site is targeted [90]. The regulation of gene expression by distal elements is complex and includes the activity of enhancer, modulators of chromatin structure, insulators, locus control regions and promoters [91,92]. Indeed, in t(14;18) lymphoma cells, aberrant expression of bcl-2 is a result of IgH 3′ enhancers interacting with bcl-2’s promoter regions [93]. The 24-base phosphodiester oligonucleotide used in PNT2258, PNT100, binds to a sequence in bcl-2’s P1 promotor region which is recognized by transcription factor Sp1 [94]. Obstructing the interaction of the promotor with the transcriptional machinery abrogates bcl-2 expression. This unmodified oligonucleotide is delivered to cells in an amphoteric liposome (SMARTIClES®, [95]) composed of 1-Palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine, cholesterol hemisuccinate, and cholesteryl-4-([2-(4-morpholinyl) ethyl]amino)-4-oxoburanoate [96] (Figure 3B). These pH-sensitive liposomes are anionic at neutral pH and cationic under acidic conditions. Their anionic state averts the aggregation of serum components thereby promoting serum stability and reducing toxicities. Their cationic state mediates efficient encapsulation of oligonucleotides during production and fusion with endosomes, thereby enabling endosomal escape within the cells [97].
Preclinical studies in non-Hodgkin’s lymphoma (NHL) cell lines (WSU-FSCCL, WSU-DLCL2 and WSU-WM) showed that PNT2258 treatments decreased cell viability in a dose-dependent manner [94]. This effect was more significant in cell lines with the t(14;18) translocation (WSU-FSCCL, WSU-DLCL2), supporting the notion that since high Bcl-2 levels in these cells was dependent on increased transcription and its distal regulation, PNT100’s activity had a greater impact [94]. Apoptotic markers such as cell shrinkage and nuclear chromatin condensation were seen 72 h after treatments with concentrations as low as 2.5 µM. DNA fragmentation, evident in the late stages of apoptosis, increased from 20% after 48 h to 40% after 72 h [94]. In addition to apoptosis, PNT2259 also induced cell cycle arrest in a non-Bcl-2, rather CDK4-dependent mechanism [98]. The exposure of cells to 2.5 µM PNT22598 increased the proportion of cells in the sub-G0 and G0/G1 phases and decreased those in G2/M [94]. The efficacy of the liposomal carrier was evidenced by the stability of PNT2285 in whole blood for at least 24 h and the absence of PNT100 in plasma post intravenous administration of PNT2258 in mice [96]. The antitumor activity of PNT2258 was tested in WSU-DLCL2, Daudi-Burkitt’s (lymphoma), PC3 (prostate) and A375 (melanoma) tumor-bearing mice. In the WSU-DLCL2 model PNT2258 demonstrated effective activity as a single agent and even more so in combination with Rituximab. Rituximab combination treatments also had a dramatic impact on tumor growth in Daudi xenografts as did Docetaxel combination treatments in A375 xenografts; showing PNT2258′s potential as an addition to standard treatments in non t(14;18) and highly resistant tumors [96].
A phase I clinical trial in patients with advanced solid tumors (NCT01191775) confirmed the high tolerability and low toxicity of PNT2258 at doses up to 150 mg/m2 [99]. Delivery was systemic and 27% of the patients receiving PNT2258 benefited with disease stabilization [99]. In a phase II clinical trial (NCT01733238) 13 patients with relapsed/refractory B-cell malignancies received PNT2258 monotherapy and had overall response rate of 53.8% and duration of response of 23.4 months [100]. Unfortunately, such results were not replicated in a larger phase II trial (Wolverine, NCT02226965). After a response rate of only 8.1% in patients with relapsed/refractory diffuse large B cell lymphoma, ProNai halted PNT2258’s development [101].

3.3. BP1002

BP1002 is a novel liposomal 20-base pair ASO which targets Bcl-2 mRNA at the translation start site. Its P-ethoxy backbone, a hydrophobic analog of phosphodiesters, makes it nuclease-resistant and lipophilic. The liposomes are composed of dioleoylphosphatidylcholine (DOPC) phospholipids and Tween 20 surfactant. DOPC is non-immunogenic and with BP1002’s overall neutral charge, potential toxicities due to interactions with cellular proteins of plasma proteins are reduced. Pharmacokinetic and tissue distribution studies of L-Bcl-2 (pilot name for BP1002) in mice caused no adverse effects and had primary accumulation in the liver and spleen [102]. As these are the primary organs in which leukemias and lymphomas manifest, preclinical studies of L-Bcl-2 have focused on such disease models.
Bcl-2 expression in HL-60 (AML) cells decreased significantly with L-Bcl-2 treatments, as evidenced by western blot and flow cytometry. The latter showed a 39% decrease in Bcl-2 expression after three days of treatment and a 60% decrease by day five [103]. Cell viability also decreased significantly and was mirrored by an increase in apoptotic sub G1 cells [103]. Similarly, L-Bcl-2 decreased proliferation of lymphoma cells lines [104]. Combination treatments of L-Bcl-2 with ara-C also proficiently induced cytotoxicity in doxorubicin-resistant HL-60 cells, which express elevated levels of both Bcl-2 and Bcl-xL [105,106]. In primary AML patient samples, 57.9% showed a decrease in viability and increase in apoptosis [105]. However, only in patient samples with low levels of Bcl-2 was L-Bcl-2 able to increase the sensitivity to ara-C [105]. Given that low Bcl-2 levels had been associated with shorter remission in AML patients with unfavorable cytogenetics, it is encouraging to think that L-Bcl-2 treatments may serve to increase their sensitivity to different chemotherapies [107]. BP1002 toxicology studies involved the treatment of mice with 7.5, 15 or 30 mg BP1002/kg and the treatment of rabbits with 3.75, 7.5, or 10 mg BP1002/kg biweekly for four weeks. In both animal models no changes in animal weight, hematological parameters (platelet count and coagulation profile) or hepatic and renal functions were observed. Thrombocytopenia is a commonly reported dose-limiting toxicity in patients administered with ASOs in clinical trials and has led to drug withdrawals [108,109]. Although the exact mechanism by which ASOs cause a reduction in platelets is still under investigation [108], the high tolerability of BP1002 in preclinical studies is promising and may be due to its P-ethoxy structure [110]. Mice harboring CJ cell xenografts (B-cell lymphoma cells) that were injected with 20 mg/kg of L-Bcl-2 biweekly for five weeks displayed delayed disease progression and had an 80% survival rate. Untreated or empty liposomes treated mice had 20% and 0% survival, respectively, by week five [104]. Establishing BP1002’s preclinical active dose to be 10 to 20 mg/kg in mice, it is estimated to be between 30 to 60 mg/m2 in humans [111].
A phase I clinical trial is currently enrolling patients with advanced lymphoid malignancies to evaluate the safety, pharmacokinetics and efficacy of BP1002 monotherapy (NCT0407258). Escalating doses of BP1002, starting at 20 mg/m2, will be intravenously administered biweekly for four weeks. BP1002 is currently the only Bcl-2 specific ASO in clinical trial (Figure 2).

4. Outlook on Bcl-2 ASO and Alternative Oligonucleotide Therapies

4.1. Overcoming Venetoclax Resistance

After years of intensive research, small molecules that can target the Bcl-2 family were developed. Venetoclax (ABT199) is the only FDA approved Bcl-2 inhibitor. This BH3 mimetic obstructs Bcl-2’s BH3 binding groove, making Bcl-2 unable to sequester and neutralize pro-apoptotic proteins [112]. Although Venetoclax has provided a much-needed step forward in cancer treatments, with a successful impact on patients with CLL, small lymphocytic leukemia (SLL) and AML, limitations such as toxicities and drug resistance remain prominent [113,114]. A detailed description of drug resistance mechanisms are provided in several recent reviews [115,116,117,118]; they include: (i) the compensatory upregulation of anti-apoptotic proteins Bcl-xL and Mcl-1 [119,120,121], (ii) super enhancer associated transcriptional reprogramming [116], (iii) enhanced mitochondrial architecture and translation [122,123] and (iv) Bcl-2 point mutations that inhibit Venetoclax binding (G101V, D103Y) [115,124]. A common approach to improve or restore the efficacy of Venetoclax is combination treatments, summarized by Yue et.al [117]. These treatments involve more than 2 different drugs and carry elevated risks of hematological and gastrointestinal toxicities [117]. Recently, the ability of BP1002 to overcome venetoclax resistance in leukemia and lymphoma cells (MV-4-11, SU-DHL-2, SU-DHL-6) in combination with decitabine has been assessed. Preliminary in vitro results show that the BP1002 + decitabine combination is more effective at decreasing cell viability in Venetoclax-resistant cells than the Venetoclax + decitabine combination [125]. Although BP1002 combination treatment studies in vivo and a phase I clinical trial are currently underway, it is encouraging to know that in a phase 1 clinical trial of BP1001 (a sister drug targeting Grb2, differing only by the ASO sequence) patients did not experience any significant dose limiting toxicities, making the maximum tolerated dose yet undetermined [126]. BP1002 may therefore be a more tolerable and efficient alternative to Venetoclax in combination treatments for Venetoclax-resistant malignancies. Nonetheless, the infancy of BP1002 makes it difficult to directly compare it to Venetoclax. While BP1002’s low toxicity is certainly advantageous and holds great promise as an adjuvant therapy, BP1002’s ability to overcome Venetoclax resistance will depend on the resistance mechanism(s) employed. For example, ASO mediated inhibition of Bcl-2 might be of little use in cells that are refractory to Venetoclax due to elevated Mcl-1 expression or reduced Bax expression. On the other hand, BP1002 might overcome Venetoclax resistance if the mechanism is due to acquired mutations in Bcl-2’s BH3 binding groove. This is due to the fact that BP1002 and Venetoclax target Bcl-2 differently; BP1002 targets the translational initiation region of the Bcl-2 mRNA while Venetoclax targets the BH3 domain of the Bcl-2 protein. Recently, Venetoclax has been shown to have off-target metabolic effects that are independent of its Bcl-2 targeting activity. Venetoclax treatment can alter mitochondrial morphology and reduce mitochondrial respiration even in cells with CRISPR/Cas9 mediated Bcl-2 deletion [127]. These changes are dependent on the transcription factor ATF4 and may promote a cell’s sensitivity to Venetoclax [127]. Off-target effects of BP1002 have not yet been observed and will be investigated.

4.2. Alternative Oligonucleotide Therapies

The regulation of Bcl-2 holds such therapeutic potential that in addition to ASOs, RNA based therapies are being developed to target it.

4.2.1. MRX34

MiRNA are endogenous 20–25 nucleotide non-coding RNAs that act as mRNA regulators. When bound to the 3′UTR of their target mRNAs, miRNA suppress target translation and promote target mRNA degradation [128,129]. Discovered in the early 1990’s in C. elegans [130], they were quickly identified across species and are now known to regulate >60% of human protein coding genes [131]. With such an important and delicate role in gene expression it is not surprising that miRNA dysregulation is seen in tumorigenesis and drug resistance [132,133,134]. MiRNA-34a is a tumor suppressor miRNA of the miRNA-34 family. Regulated by transcription factor p53, which mediates anti-proliferative stress responses, miRNA-34a suppresses the expression of more than 200 targets that have regulatory roles in cell cycle, metastasis, epithelial-mesenchymal transition, stemness, invasion and cell survival [135,136,137]. Bcl-2 is a miRNA-34a target, thus the reduced expression of miRNA-34a in various solid and hematological malignancies is often accompanied by an increase in Bcl-2 expression [138,139,140,141]. Mirna Therapeutics, Inc generated a miRNA-34a mimetic which was incorporated into a liposome; the drug product was named MRX34 [142]. Preclinical in vivo studies reported systemic delivery, low toxicity and sufficient suppression of tumor growth in mouse models with lymphoma, liver and lung cancer [142,143,144]. A phase I clinical trial (NCT02862145) however, had to be withdrawn and MRX34 development halted after severe immune mediated adverse effects resulted in the death of four patients [145].

4.2.2. TargomiR

MiRNA-15/16 is another family of tumor suppressing miRNA that regulates Bcl-2 levels and has reduced expression in various cancer [146,147]. Malignant pleural mesothelioma (MPM) patients samples revealed a 22-fold decrease of miRNA-16 in tumor tissues compared to normal tissues, a 4-fold decrease of miRNA-15a, and a 10-fold decrease of miRNA-15b [148]. Transfection of MPM cells with a miRNA-16 mimic most effectively reduced cell growth and colony formation, led to the accumulation of cells in G0/G1 and increased cell sensitivity to chemotherapeutic drugs [148]. Treatment of tumor xenografts with mimic miRNA-16 resulted in dose-dependent inhibition of tumor growth. MiRNA-16 is delivered to the tumor cells in a nano vehicle named TargomiRs. These are nonliving bacterial minicells, which have intrinsic endosomal escape mechanisms, coated with EGFR antibodies to enable targeting [148] (Figure 3C). In a dose escalation phase I clinical trial (NCT02369198) TargomiR was well tolerated in patients with MPM and NSCLC, with no adverse immune effects and early signs of antitumor activity [149]. These results encourage further trials in combination with chemotherapy or immune checkpoint inhibitors (Figure 2).

4.2.3. siBCL-2 NKExos

As shown in the examples above the development of oligonucleotide therapeutics is accompanied by innovative delivery technologies: from neutral liposomes to pH sensitive liposomes, from bacterial minicells to endogenous nanovesicles. Exosomes are vesicles released from cells to mediate long distance cell-cell communication [44]. They are emerging delivery vehicles with high potential given their nanosize (50 to 150 nm), biocompatibility, extended blood half-life and low immunogenicity [150] (Figure 3D). The proteins and lipids that exosomes are composed of depends on their cell of origin and can have different therapeutic implications. Exosomes from natural killer (NK) cells contain granzymes, which activate caspases in target cells, and carry a variety of receptors that specifically direct them to ligands expressed on stressed/tumor cells [151,152,153]. These intrinsic cytotoxic, anti-tumor traits have made NK-exosomes (NKExos) ideal therapeutic drug carriers. A recent study demonstrated that NKExos holding Bcl-2 small interfering RNA (siRNA), generated from lentivirally modified NK cells, enhanced apoptosis in estrogen receptor-positive breast cancer cells [154]. The advantages that NKExos provide in terms of tumor specificity, cell uptake and siRNA release make them a promising delivery option for Bcl-2 targeting.
A novel cell-penetrating-peptide equipped exosome has recently been engineered for the optimization of drug delivery via the endocytic pathway. Using Oblimersen as a model, these HepG2 cell derived exosomes, hold a polyarginine peptide (R9) at their surface, EXO-R9-G3139 [155]. R9 is a cell penetrating peptide that serves to enhance exosome cell binding and internalization [155]. HepG2 cells treated with EXO-R9-G3139 for 48 h experienced a 73% decrease in Bcl-2 mRNA expression, and a 70% decrease in Bcl-2 protein expression [155]. Cells treated with EXO-G3139, lacking the R9 peptide, had a 44% and 52% decrease in Bcl-2 mRNA and protein, respectively [155]. Cells treated with G3139 or R9-G3139, on the other hand, showed little difference to untreated cells [155]. This work suggests that exosomes or exosome-like vesicles can be engineered to enhance their drug delivery capacities and may be the next innovative step in oligonucleotide therapies.

5. Conclusions

Once identified as a proto-oncogene the targeting of Bcl-2 with ASO therapies was set in motion. Throughout the years much has been learnt about target sequence selection, chemical modifications, stability, delivery and toxicity. Although the progression of several Bcl-2-ASO candidates through clinical trials has ended in failure, due to adverse effects and limited efficacy, the potential of this technology in cancer therapies has not been dispelled. BP1002 remains a promising Bcl-2-ASO in clinical trials. BP1002 builds on the lessons learnt from previous candidates. Its unique P-ethoxy backbone limits the adverse effects of thrombocytopenia and complement activation. Its incorporation into a liposomal delivery vehicle further prolongs its half-life and cellular uptake. Its ability to efficiently suppress Bcl-2 without causing adverse effects nominates it as a valuable contributor in combination therapies and may become a more tolerable alternative to Venetoclax.
The need to safely transport ASO to target cells and allow their intracellular release continues to launch novel delivery technologies. Targeted delivery to cancer cells is a desired goal in the field. Ligand targeted liposomes, which have antibodies or peptides attached to their outer surface, offer site and cell specific delivery. The technology mainly relies on overexpression of specific proteins on tumor cell surfaces [156], however toxicity to normal tissues may still arise as expression of these proteins are found in normal tissues albeit at lower levels. Manufacturing of pharmaceutical grade ligand targeted, ASO-incorporated liposomes might also be complex and expensive. Endogenous and engineered exosomal vesicles also hold great promise [157]. Their biocompatibility and inherent targeted transport makes them exciting ASO carriers with low toxicity and efficient cellular uptake [158]. Nonetheless, large scale purification of exosomal vesicles to meet pharmaceutical quality could be challenging. ASO technology targeting Bcl-2 has come a long way and continues to develop. Although optimizations are still being tested, the potential impact they will have on cancer therapeutics is certainly worth the wait.

Author Contributions

Conceptualization, M.G. and A.T.A.; writing—original draft preparation, M.G.; writing—review and editing, M.G. and A.T.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

M.G. and A.T.A. are employees of Bio-Path Holdings, Inc. (Bellaire, TX, USA).

References

  1. Arya, R.; White, K. Cell death in development: Signaling pathways and core mechanisms. Semin. Cell Dev. Biol. 2015, 39, 12–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Akhtar, R.S.; Ness, J.M.; Roth, K.A. Bcl-2 family regulation of neuronal development and neurodegeneration. Biochim. Biophys. Acta Mol. Cell Res. 2004, 1644, 189–203. [Google Scholar] [CrossRef] [PubMed]
  3. Lu, Q.L.; Abel, P.; Foster, C.S.; Lalani, E.N. Bcl-2: Role in epithelial differentiation and oncogenesis. Hum. Pathol. 1996, 27, 102–110. [Google Scholar] [CrossRef]
  4. Kelekar, A.; Thompson, C.B. Bcl-2-family proteins: The role of the BH3 domain in apoptosis. Trends Cell Biol. 1998, 8, 324–330. [Google Scholar] [CrossRef]
  5. Westphal, D.; Kluck, R.M.; Dewson, G. Building blocks of the apoptotic pore: How bax and bak are activated and oligomerize during apoptosis. Cell Death Differ. 2013, 21, 196–205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Pihán, P.; Carreras-Sureda, A.; Hetz, C. BCL-2 Family: Integrating stress responses at the ER to control cell demise. Cell Death Differ. 2017, 24, 1478–1487. [Google Scholar] [CrossRef] [Green Version]
  7. Kønig, S.M.; Rissler, V.; Terkelsen, T.; Lambrughi, M.; Papaleo, E. Alterations of the interactome of Bcl-2 proteins in breast cancer at the transcriptional, mutational and structural level. PLoS Comput. Biol. 2019, 15, e1007485. [Google Scholar] [CrossRef]
  8. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  9. Tsujimoto, Y.; Gorham, J.; Cossman, J.; Jaffe, E.; Croce, C.M. The t(14;18) chromosome translocations involved in B-cell neoplasms result from mistakes in VDJ joining. Science 1985, 229, 1390–1393. [Google Scholar] [CrossRef]
  10. Tsujimoto, Y.; Finger, L.R.; Yunis, J.; Nowell, P.C.; Croce, C.M. Cloning of the Chromosome Breakpoint of Neoplastic B Cells with the t(14;18) Chromosome Translocation. Science 1984, 226, 1097–1099. [Google Scholar] [CrossRef]
  11. Tsujimoto, Y.; Cossman, J.; Jaffe, E.; Croce, C.M. Involvement of the Bcl-2 gene in human follicular lymphoma. Science 1985, 228, 1440–1443. [Google Scholar] [CrossRef]
  12. Eberle, J.; Hossini, A. Expression and function of Bcl-2 proteins in melanoma. Curr. Genom. 2008, 9, 409–419. [Google Scholar] [CrossRef] [Green Version]
  13. Merino, D.; Lok, S.W.; Visvader, J.E.; Lindeman, G.J. Targeting BCL-2 to enhance vulnerability to therapy in estrogen receptor-positive breast cancer. Oncogene 2016, 35, 1877–1887. [Google Scholar] [CrossRef] [PubMed]
  14. Du, C.; Zhang, X.; Yao, M.; Lv, K.; Wang, J.; Chen, L.; Chen, Y.; Wang, S.; Fu, P. Bcl-2 promotes metastasis through the epithelial-to-mesenchymal transition in the BCap37 medullary breast cancer cell line. Oncol. Lett. 2018, 15, 8991–8998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Zhao, D.-P.; Ding, X.-W.; Peng, J.-P.; Zheng, Y.-X.; Zhang, S.-Z. Prognostic significance of Bcl-2 and P53 expression in colorectal carcinoma. J. Zhejiang Univ. Sci. B 2005, 6, 1163–1169. [Google Scholar] [CrossRef] [Green Version]
  16. Anagnostou, V.K.; Lowery, F.J.; Zolota, V.; Tzelepi, V.; Gopinath, A.; Liceaga, C.; Panagopoulos, N.; Frangia, K.; Tanoue, L.; Boffa, D.; et al. High expression of BCL-2 predicts favorable outcome in non-small cell lung cancer patients with non squamous histology. BMC Cancer 2010, 10, 186. [Google Scholar] [CrossRef] [Green Version]
  17. Monni, O.; Joensuu, H.; Franssila, K.; Klefstrom, J.; Alitalo, K.; Knuutila, S. Bcl-2 overexpression associated with chromosomal amplification in diffuse large B-cell lymphoma. Blood 1997, 90, 1168–1174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Saito, M.; Novak, U.; Piovan, E.; Basso, K.; Sumazin, P.; Schneider, C.; Crespo, M.; Shen, Q.; Bhagat, G.; Califano, A.; et al. Bcl-6 suppression of Bcl-2 via Miz1 and its disruption in diffuse large B cell lymphoma. Proc. Natl. Acad. Sci. USA 2009, 106, 11294–11299. [Google Scholar] [CrossRef] [Green Version]
  19. Catz, S.D.; Johnson, J.L. Transcriptional regulation of Bcl-2 by nuclear factor ΚB and its significance in prostate cancer. Oncogene 2001, 20, 7342–7351. [Google Scholar] [CrossRef] [Green Version]
  20. Cimmino, A.; Calin, G.A.; Babbri, M.; Iorio, M.V.; Ferracin, M.; Shimizu, M.; Wojcik, S.E.; Aqeilan, R.I.; Zupo, S.; Dono, M.; et al. MiR-15 and MiR-16 induce apoptosis by targeting BCL2. Proc. Natl. Acad. Sci. USA 2005, 39, 13944–13949. [Google Scholar] [CrossRef] [Green Version]
  21. Gabellini, C.; Trisciuoglio, D.; Del Bufalo, D. Non-canonical roles of Bcl-2 and Bcl-XL proteins: Relevance of BH4 domain. Carcinogenesis 2017, 38, 579–587. [Google Scholar] [CrossRef]
  22. Folkman, J. Role of angiogenesis in tumor growth and metastasis. Semin. Oncol. 2002, 29, 15–18. [Google Scholar] [CrossRef] [PubMed]
  23. Karl, E.; Warner, K.; Zeitlin, B.; Kaneko, T.; Wurtzel, L.; Jin, T.; Chang, J.; Wang, S.; Wang, C.-Y.; Strieter, R.M.; et al. Bcl-2 acts in a proangiogenic signaling pathway through nuclear factor-ΚB and CXC chemokines. Cancer Res. 2005, 65, 5063–5069. [Google Scholar] [CrossRef] [Green Version]
  24. Iervolino, A.; Trisciuoglio, D.; Ribatti, D.; Candiloro, A.; Biroccio, A.; Zupi, G.; Del Bufalo, D. Bcl-2 overexpression in human melanoma cells increases angiogenesis through VEGF MRNA stabilization and HIF-1-mediated transcriptional activity. FASEB J. 2002, 16, 1453–1455. [Google Scholar] [CrossRef]
  25. Sun, T.; Sun, B.C.; Zhao, X.L.; Zhao, N.; Dong, X.Y.; Che, N.; Yao, Z.; Ma, Y.M.; Gu, Q.; Zong, W.K.; et al. Promotion of tumor cell metastasis and vasculogenic mimicry by way of transcription coactivation by Bcl-2 and Twist1: A study of hepatocellular carcinoma. Hepatology 2011, 54, 1690–1706. [Google Scholar] [CrossRef] [PubMed]
  26. Nör, J.E.; Christensen, J.; Mooney, D.J.; Polverini, P.J. Vascular Endothelial Growth Factor (VEGF)-mediated angiogenesis is associated with enhanced endothelial cell survival and induction of Bcl-2 expression. Am. J. Pathol. 1999, 154, 375–384. [Google Scholar] [CrossRef] [Green Version]
  27. Trisciuoglio, D.; Gabellini, C.; Desideri, M.; Ragazzoni, Y.; De Luca, T.; Ziparo, E.; Del Bufalo, D. Involvement of BH4 domain of Bcl-2 in the regulation of HIF-1-mediated VEGF expression in hypoxic tumor cells. Cell Death Differ. 2011, 18, 1024–1035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Gabellini, C.; De Luca, T.; Trisciuoglio, D.; Desideri, M.; Di Martile, M.; Passeri, D.; Candiloro, A.; Biffoni, M.; Rizzo, M.G.; Orlandi, A.; et al. BH4 domain of Bcl-2 protein is required for its proangiogenic function under hypoxic condition. Carcinogenesis 2013, 34, 2558–2567. [Google Scholar] [CrossRef] [Green Version]
  29. Ricca, A.; Biroccio, A.; Del Bufalo, D.; Mackay, A.R.; Santoni, A.; Cippitelli, M. Bcl-2 over-expression enhances NF-ΚB activity and induces Mmp-9 transcription in human MCF7(ADR) breast-cancer cells. Int. J. Cancer 2000, 86, 188–196. [Google Scholar] [CrossRef]
  30. Regula, K.M.; Ens, K.; Kirshenbaum, L.A. IKKβ is required for Bcl-2-mediated NF-ΚB activation in ventricular myocytes. J. Biol. Chem. 2002, 277, 38676–38682. [Google Scholar] [CrossRef] [Green Version]
  31. Pires, B.R.B.; Mencalha, A.L.; Ferreira, G.M.; De Souza, W.F.; Morgado-Díaz, J.A.; Maia, A.M.; Corrêa, S.; Abdelhay, E.S.F.W. NF-KappaB is involved in the regulation of EMT genes in breast cancer cells. PLoS ONE 2017, 12, e0169622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Choi, J.; Choi, K.; Benveniste, E.N.; Hong, Y.-S.; Lee, J.-H.; Kim, J.; Park, K. Bcl-2 promotes invasion and lung metastasis by inducing matrix metalloproteinase-2. Cancer Res. 2005, 65, 5554–5560. [Google Scholar] [CrossRef] [Green Version]
  33. Montero, J.; Letai, A. Why do BCL-2 inhibitorswork and where should we use them in the clinic? Cell Death Differ. 2018, 25, 56–64. [Google Scholar] [CrossRef]
  34. Certo, M.; Moore, V.D.G.; Nishino, M.; Wei, G.; Korsmeyer, S.; Armstrong, S.A.; Letai, A. Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2 family members. Cancer Cell 2006, 9, 351–365. [Google Scholar] [CrossRef] [Green Version]
  35. Sarosiek, K.A.; Fraser, C.; Muthalagu, N.; Bhola, P.D.; Chang, W.; McBrayer, S.K.; Cantlon, A.; Fisch, S.; Golomb-Mello, G.; Ryan, J.A.; et al. Developmental regulation of mitochondrial apoptosis by C-myc governs age- and tissue-specific sensitivity to cancer therapeutics. Cancer Cell 2017, 31, 142–156. [Google Scholar] [CrossRef] [Green Version]
  36. Perini, G.F.; Ribeiro, G.N.; Pinto Neto, J.V.; Campos, L.T.; Hamerschlak, N. BCL-2 as therapeutic target for hematological malignancies. J. Hematol. Oncol. 2018, 11, 65. [Google Scholar] [CrossRef] [PubMed]
  37. Chauhan, D.; Velankar, M.; Brahmandam, M.; Hideshima, T.; Podar, K.; Richardson, P.; Schlossman, R.; Ghobrial, I.; Raje, N.; Munshi, N.; et al. A novel Bcl-2/Bcl-XL/Bcl-w inhibitor ABT-737 as therapy in multiple myeloma. Oncogene 2007, 26, 2374–2380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Gagliardi, M.; Ashizawa, A.T. The challenges and strategies of antisense oligonucleotide drug delivery. Biomedicines 2021, 9, 433. [Google Scholar] [CrossRef] [PubMed]
  39. Dias, N.; Stein, C.A. Antisense oligonucleotides: Basic concepts and mechanisms. Mol. Cancer Ther. 2002, 1, 347–355. [Google Scholar]
  40. Dhuri, K.; Bechtold, C.; Quijano, E.; Pham, H.; Gupta, A.; Vikram, A.; Bahal, R. Antisense oligonucleotides: An emerging area in drug discovery and development. J. Clin. Med. 2020, 9, 2004. [Google Scholar] [CrossRef]
  41. Eckstein, F. Phosphorothioates, essential components of therapeutic oligonucleotides. Nucleic Acid Ther. 2014, 24, 374–387. [Google Scholar] [CrossRef]
  42. Baker, B.; Lot, S.; Condon, T.; Cheng-Flournoy, S.; Lesnik, E.; Sasmor, H.; Bennett, C. 2’-O-(2-Methoxy)Ethyl-modified anti-intercellular adhesion molecule 1 (ICAM-1) oligonucleotides selectively increase the ICAM-1 MRNA level and inhibit formation of the ICAM-1 translation initiation complex in human umbilical vein endothelial cells. J. Biol. Chem. 1997, 272, 11994–12000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Shen, W.; De Hoyos, C.L.; Sun, H.; Vickers, T.A.; Liang, X.H.; Crooke, S.T. Acute hepatotoxicity of 2 fluoro-modified 5–10–5 gapmer phosphorothioate oligonucleotides in mice correlates with intracellular protein binding and the loss of DBHS proteins. Nucleic Acids Res. 2018, 46, 2204–2217. [Google Scholar] [CrossRef] [Green Version]
  44. Luan, X.; Sansanaphongpricha, K.; Myers, I.; Chen, H.; Yuan, H.; Sun, D. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacol. Sin. 2017, 38, 754–763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Vasquez, G.; Freestone, G.C.; Wan, W.B.; Low, A.; De Hoyos, C.L.; Yu, J.; Prakash, T.P.; Ostergaard, M.E.; Liang, X.H.; Crooke, S.T.; et al. Site-Specific Incorporation of 5′-methyl DNA enhances the therapeutic profile of gapmer ASOs. Nucleic Acids Res. 2021, 49, 1828–1839. [Google Scholar] [CrossRef]
  46. Khvorova, A.; Watts, J. The chemical evolution of oligonucleotide therapies of clinical utility. Physiol. Behav. 2017, 35, 238–248. [Google Scholar] [CrossRef] [PubMed]
  47. 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] [PubMed]
  48. Reed, J.C.; Kitada, S.; Takayama, S.; Miyashita, T. Regulation of chemoresistance by the Bcl-2 oncoprotein in non-Hodgkin’s lymphoma and lymphocytic leukemia cell lines. Ann. Oncol. 1994, 5, 61–65. [Google Scholar] [CrossRef]
  49. Maung, Z.T.; MacLean, F.R.; Reid, M.M.; Pearson, A.D.J.; Proctor, S.J.; Hamilton, P.J.; Hall, A.G. The relationship between Bcl-2 expression and response to chemotherapy in acute leukaemia. Br. J. Haematol. 1994, 88, 105–109. [Google Scholar] [CrossRef]
  50. Kitada, S.; Takayama, S.; de Riel, K.; Tanaka, S.; Reed, J.C. Reversal of chemoresistance of lymphoma cells by antisense-mediated reduction of Bcl-2 gene expression. Antisense Res. Dev. 1994, 4, 71–79. [Google Scholar] [CrossRef]
  51. Dang, C.V.; Reddy, E.P.; Shokat, K.M.; Soucek, L. Drugging the “undruggable” cancer targets. Nat. Rev. Cancer 2017, 17, 502–508. [Google Scholar] [CrossRef]
  52. Reed, J.C.; Yum, S.; Stein, C.; Subasinghe, C.; Cohen, J.; Haldar, S.; Croce, C.M. Antisense-mediated inhibition of BCL2 protooncogene expression and leukemic cell growth and survival: Comparisons of phosphodiester and phosphorothioate oligodeoxynucleotides. Cancer Res. 1990, 50, 6565–6570. [Google Scholar] [PubMed]
  53. Campos, L.; Sabido, O.; Rouault, J.; Guyotat, D. Effects of BCL-2 Antisense oligodeoxynucleotides on in vitro proliferation and survival of normal marrow progenitors and leukemic cells. Blood 1994, 84, 595–600. [Google Scholar] [CrossRef] [Green Version]
  54. Ziegler, A.; Luedke, G.H.; Fabbro, D.; Altmann, K.H.; Stahel, R.A.; Zangemeister-Wittke, U. Induction of Apoptosis in small-cell lung cancer cells by an antisense oligodeoxynucleotide targeting the Bcl-2 coding sequence. J. Natl. Cancer Inst. 1997, 89, 1027–1036. [Google Scholar] [CrossRef]
  55. Zangemeister-Wittke, U.; Schenker, T.; Luedke, G.H.; Stahel, R.A. Synergistic cytotoxicity of Bcl-2 antisense oligodeoxynucleotides and etoposide, doxorubicin and cisplatin on small-cell lung cancer cell lines. Br. J. Cancer 1998, 78, 1035–1042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Fanfone, D.; Idbaih, A.; Mammi, J.; Gabut, M.; Ichim, G. Profiling anti-apoptotic Bcl-Xl protein expression in glioblastoma tumorspheres. Cancers 2020, 12, 2853. [Google Scholar] [CrossRef]
  57. Bessou, M.; Lopez, J.; Gadet, R.; Deygas, M.; Popgeorgiev, N.; Poncet, D.; Nougarède, A.; Billard, P.; Mikaelian, I.; Gonzalo, P.; et al. The apoptosis inhibitor Bcl-XL controls breast cancer cell migration through mitochondria-dependent reactive oxygen species production. Oncogene 2020, 39, 3056–3074. [Google Scholar] [CrossRef] [PubMed]
  58. Trisciuoglio, D.; Tupone, M.G.; Desideri, M.; Di Martile, M.; Gabellini, C.; Buglioni, S.; Pallocca, M.; Alessandrini, G.; D’Aguanno, S.; Del Bufalo, D. BCL-XL overexpression promotes tumor progression-associated properties. Cell Death Dis. 2017, 8, 1–15. [Google Scholar] [CrossRef] [Green Version]
  59. Lebedeva, I.; Rando, R.; Ojwang, J.; Cossum, P.; Stein, C.A. Bcl-XL in prostate cancer cells: Effects of overexpression and down-regulation on chemosensitivity. Cancer Res. 2000, 60, 6052–6060. [Google Scholar] [PubMed]
  60. Simonian, P.L.; Grillot, D.A.M.; Nuñez, G. Bcl-2 and Bcl-XL can differentially block chemotherapy-induced cell death. Blood 1997, 90, 1208–1216. [Google Scholar] [CrossRef] [PubMed]
  61. Han, Z.; Chatterjee, D.; Early, J.; Pantazis, P.; Hendrickson, E.A.; Wyche, J.H. Isolation and characterization of an apoptosis-resistant variant of human leukemia HL-60 cells that has switched expression from Bcl-2 to Bcl-XL 1. Cancer Res. 1996, 56, 1621–1628. [Google Scholar] [PubMed]
  62. Fiebig, A.A.; Zhu, W.; Hollerbach, C.; Leber, B.; Andrews, D.W. Bcl-XL is qualitatively different from and ten times more effective than Bcl-2 when expressed in a breast cancer cell line. BMC Cancer 2006, 6, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Zangemeister-Wittke, U.; Leech, S.H.; Olie, R.A.; Simões-Wüst, A.P.; Gautschi, O.; Luedke, G.H.; Natt, F.; Häner, R.; Martin, P.; Hall, J.; et al. A novel bispecific antisense oligonucleotide inhibiting both Bcl-2 and Bcl-XL expression efficiently induces apoptosis in tumor cells. Clin. Cancer Res. 2000, 6, 2547–2555. [Google Scholar]
  64. Gautschi, O.; Tschopp, S.; Olie, R.A.; Leech, S.H.; Simões-Wüst, A.P.; Ziegler, A.; Baumann, B.; Odermatt, B.; Hall, J.; Stahel, R.A.; et al. Activity of a novel Bcl-2/Bcl-XL-bispecific antisense oligonucleotide against tumors of diverse histologic origins. J. Natl. Cancer Inst. 2001, 93, 463–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Del Bufalo, D.; Trisciuoglio, D.; Scarsella, M.; Zangemeister-Wittke, U.; Zupi, G. Treatment of melanoma cells with a Bcl-2/Bcl-XL antisense oligonucleotide induces antiangiogenic activity. Oncogene 2003, 22, 8441–8447. [Google Scholar] [CrossRef] [Green Version]
  66. Yamanaka, K.; Rocchi, P.; Miyake, H.; Fazli, L.; So, A.; Zangemeister-Wittke, U.; Gleave, M.E. Induction of apoptosis and enhancement of chemosensitivity in human prostate cancer LNCaP cells using bispecific antisense oligonucleotide targeting Bcl-2 and Bcl-XL genes. BJU Int. 2006, 97, 1300–1308. [Google Scholar] [CrossRef]
  67. Yamanaka, K.; Rocchi, P.; Miyake, H.; Fazli, L.; Vessella, B.; Zangemeister-Wittke, U.; Gleave, M.E. A novel antisense oligonucleotide inhibiting several antiapoptotic Bcl-2 Family members induces apoptosis and enhances chemosensitivity in androgen-independent human prostate cancer PC3 cells. Mol. Cancer Ther. 2005, 4, 1689–1698. [Google Scholar] [CrossRef] [Green Version]
  68. Simões-Wüst, A.P.; Hopkins-Donaldson, S.; Sigrist, B.; Belyanskaya, L.; Stahel, R.A.; Zangemeister-Wittke, U. A Functionally improved locked nucleic acid antisense oligonucleotide inhibits Bcl-2 and Bcl-XL expression and facilitates tumor cell apoptosis. Oligonucleotides 2004, 14, 199–209. [Google Scholar] [CrossRef]
  69. Schlagbauer-Wadl, H.; Klosner, G.; Heere-Ress, E.; Waltering, S.; Moll, I.; Wolff, K.; Pehamberger, H.; Jansen, B. Bcl-2 Antisense oligonucleotides (G3139) inhibit merkel cell carcinoma growth in SCID mice. J. Investig. Dermatol. 2000, 114, 725–730. [Google Scholar] [CrossRef] [Green Version]
  70. Webb, A.; Cunningham, D.; Cotter, F.; Clarke, P.A.; Di Stefano, F.; Ross, P.; Corbo, M.; Dziewanowska, Z. BCL-2 Antisense therapy in patients with non-hodgkin lymphoma. Lancet 1997, 349, 1137–1141. [Google Scholar] [CrossRef]
  71. Oblimersen: Augmerosen, BCL-2 Antisense Oligonucleotide—Genta, G 3139, GC 3139, oblimersen sodium. Drugs R D 2007, 8, 321–334. [CrossRef]
  72. Kitada, S.; Miyashita, T.; Tanaka, S.; Reed, J.C. Investigations of antisense oligonucleotides targeted against Bcl-2 RNAs. Antisense Res. Dev. 1993, 3, 157–169. [Google Scholar] [CrossRef] [PubMed]
  73. Klasa, R.J.; Bally, M.B.; Ng, R.; Goldie, J.H.; Gascoyne, R.D.; Wong, F.M.P. Eradication of human non-hodgkin’s lymphoma in SCID mice by BCL-2 antisense oligonucleotides combined with low-dose cyclophosphamide. Clin. Cancer Res. 2000, 6, 2492–2500. [Google Scholar] [PubMed]
  74. Herbst, R.S.; Frankel, S.R. Oblimersen sodium (Genasense Bcl-2 antisense oligonucleotide): A rational therapeutic to enhance apoptosis in therapy of lung cancer. Clin. Cancer Res. 2004, 10, 4245s–4248s. [Google Scholar] [CrossRef]
  75. Leleu, X.; Gay, J.; Roccaro, A.M.; Moreau, A.S.; Paulain, S.; Dulery, R.; Champs, B.; Robu, D.; Ghobrial, I.M. Update on therapeutic options in waldenström macroglobulinemia. Eur. J. Haematol. 2011, 82, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Gertz, M.A.; Geyer, S.M.; Badros, A.; Kahl, B.S.; Erlichman, C. Early results of a phase I trial of oblimersen sodium for relapsed or refractory Waldenström’s macroglobulinemia. Clin. Lymphoma 2005, 5, 282–284. [Google Scholar] [CrossRef]
  77. Morris, M.J.; Tong, W.P.; Cordon-Cardo, C.; Drobnjak, M.; Kelly, W.K.; Slovin, S.F.; Terry, K.L.; Siedlecki, K.; Swanson, P.; Rafi, M.; et al. Phase I trial of BCL-2 antisense oligonucleotide (G3139) administered by continuous intravenous infusion in patients with advanced cancer. Clin. Cancer Res. 2002, 8, 679–683. [Google Scholar]
  78. O’Brien, S.M.; Cunningham, C.C.; Golenkov, A.K.; Turkina, A.G.; Novick, S.C.; Rai, K.R. Phase I to II multicenter study of oblimersen sodium, a Bcl-2 Antisense oligonucleotide, in patients with advanced chronic lymphocytic leukemia. J. Clin. Oncol. 2005, 23, 7697–7702. [Google Scholar] [CrossRef]
  79. Bedikian, A.Y.; Millward, M.; Pehamberger, H.; Conry, R.; Gore, M.; Trefzer, U.; Pavlick, A.C.; DeConti, R.; Hersh, E.M.; Hersey, P.; et al. Bcl-2 antisense (Oblimersen Sodium) plus dacarbazine in patients with advanced melanoma: The oblimersen melanoma study group. J. Clin. Oncol. 2006, 24, 4738–4745. [Google Scholar] [CrossRef]
  80. Chanan-Khan, A.; Niesvizky, R.; Hohl, R.J.; Zimmerman, T.M.; Christiansen, N.P.; Schiller, G.J.; Callander, N.; Lister, J.; Oken, M.; Jagannath, S. Phase III randomised study of dexamethasone with or without oblimersen sodium for patients with advanced multiple myeloma. Leuk. Lymphoma 2009, 50, 559–565. [Google Scholar] [CrossRef]
  81. O’Brien, S.; Moore, J.O.; Boyd, T.E.; Larratt, L.M.; Skotnicki, A.; Koziner, B.; Chanan-Khan, A.A.; Seymour, J.F.; Bociek, R.G.; Pavletic, S.; et al. Randomized phase III trial of fludarabine plus cyclophosphamide with or without oblimersen sodium (Bcl-2 Antisense) in patients with relapsed or refractory chronic lymphocytic leukemia. J. Clin. Oncol. 2007, 25, 1114–1120. [Google Scholar] [CrossRef]
  82. Marcucci, G.; Stock, W.; Dai, G.; Klisovic, R.B.; Liu, S.; Klisovic, M.I.; Blum, W.; Kefauver, C.; Sher, D.A.; Green, M.; et al. Phase I study of oblimersen sodium, an antisense to Bcl-2, in untreated older patients with acute myeloid leukemia: Pharmacokinetics, pharmacodynamics, and clinical activity. J. Clin. Oncol. 2005, 23, 3404–3411. [Google Scholar] [CrossRef]
  83. Walker, A.R.; Marcucci, G.; Yin, J.; Blum, W.; Stock, W.; Kohlschmidt, J.; Mrózek, K.; Carroll, A.J.; Eisfeld, A.K.; Wang, E.S.; et al. Phase 3 randomized trial of chemotherapy with or without oblimersen in older AML patients: CALGB 10201 (Alliance). Blood Adv. 2021, 5, 2775–2787. [Google Scholar] [CrossRef] [PubMed]
  84. Bedikian, A.Y.; Garbe, C.; Conry, R.; Lebbe, C.; Grob, J.J. Dacarbazine with or without Oblimersen (a Bcl-2 Antisense Oligonucleotide) in chemotherapy-naive patients with advanced melanoma and low-normal serum lactate dehydrogenase: “The AGENDA trial”. Melanoma Res. 2014, 24, 237–243. [Google Scholar] [CrossRef] [PubMed]
  85. Frantz, S. Lessons learnt from Genasense’s failure. Nat. Rev. Drug Discov. 2004, 3, 542–543. [Google Scholar] [CrossRef] [PubMed]
  86. Stein, C.A.; Hansen, J.B.; Lai, J.; Wu, S.; Voskresenskiy, A.; Høg, A.; Worm, J.; Hedtjärn, M.; Souleimanian, N.; Miller, P.; et al. Efficient gene silencing by delivery of locked nucleic acid antisense oligonucleotides, unassisted by transfection reagents. Nucleic Acids Res. 2010, 38, e3. [Google Scholar] [CrossRef] [Green Version]
  87. Dürig, J.; Dührsen, U.; Klein-Hitpass, L.; Worm, J.; Hansen, J.B.R.; Orum, H.; Wissenbach, M. The novel antisense Bcl-2 inhibitor SPC2996 causes rapid leukemic cell clearance and immune activation in chronic lymphocytic leukemia. Leukemia 2011, 25, 638–647. [Google Scholar] [CrossRef]
  88. Lamphier, M.S.; Sirois, C.M.; Verma, A.; Golenbock, D.T.; Latz, E. TLR9 and the recognition of self and non-self nucleic acids. Ann. N. Y. Acad. Sci. 2006, 1082, 31–43. [Google Scholar] [CrossRef]
  89. Dampmann, M.; Görgens, A.; Möllmann, M.; Murke, F.; Dührsen, U.; Giebel, B.; Dürig, J. CpG Stimulation of chronic lymphocytic leukemia cells induces a polarized cell shape and promotes migration in vitro and in vivo. PLoS ONE 2020, 15, e0228674. [Google Scholar] [CrossRef]
  90. ProNAi Announces Dna Interference (DNAI(R)) Drug Development Strategy For 2007 | DNA RNA and Cells | News Channels. Available online: https://pipelinereview.com/index.php/200701099082/DNA-RNA-and-Cells/ProNAi-Announces-Dna-Interference-DNAIR-Drug-Development-Strategy-For-2007.html (accessed on 16 October 2021).
  91. Maston, G.A.; Evans, S.K.; Green, M.R. Transcriptional regulatory elements in the human genome. Annu. Rev. Genom. Hum. Genet. 2006, 7, 29–59. [Google Scholar] [CrossRef] [Green Version]
  92. Gaffney, D.J. Mapping and predicting gene–Enhancer interactions. Nat. Genet. 2019, 51, 1662–1663. [Google Scholar] [CrossRef]
  93. Duan, H.; Xiang, H.; Ma, L.; Boxer, L.M. Functional long-range interactions of the IgH 3′ enhancers with the Bcl-2 promoter region in t(14;18) lymphoma cells. Oncogene 2008, 27, 6720–6728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Ebrahim, A.S.; Kandouz, M.; Liddane, A.; Sabbagh, H.; Hou, Y.; Li, C.; Al-Katib, A. PNT2258, a novel deoxyribonucleic acid inhibitor, induces cell cycle arrest and apoptosis via a distinct mechanism of action: A new class of drug for non-Hodgkin’s lymphoma. Oncotarget 2016, 7, 42374–42384. [Google Scholar] [CrossRef] [Green Version]
  95. Adami, R.C.; Seth, S.; Harvie, P.; Johns, R.; Fam, R.; Fosnaugh, K.; Zhu, T.; Farber, K.; McCutcheon, M.; Goodman, T.T.; et al. An Amino Acid-Based Amphoteric Liposomal Delivery System for Systemic Administration of SiRNA. Mol. Ther. 2011, 19, 1141–1151. [Google Scholar] [CrossRef] [PubMed]
  96. Rodrigueza, W.V.; Woolliscroft, M.J.; Ebrahim, A.S.; Forgey, R.; McGovren, P.J.; Endert, G.; Wagner, A.; Holewa, D.; Aboukameel, A.; Gill, R.D.; et al. Development and antitumor activity of a BCL-2 targeted single-stranded DNA oligonucleotide. Cancer Chemother. Pharmacol. 2014, 74, 151–166. [Google Scholar] [CrossRef] [Green Version]
  97. Siepi, E.; Lutz, S.; Meyer, S.; Panzner, S. An ion switch regulates fusion of charged membranes. Biophys. J. 2011, 100, 2412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Ebrahim, A.S.; Kandouz, M.; Emara, N.; Sugalski, A.B.; Lipovich, L.; Al-Katib, A.M. Unintended target effect of anti-BCL-2 DNAi. Cancer Manag. Res. 2017, 9, 427. [Google Scholar] [CrossRef] [Green Version]
  99. Tolcher, A.W.; Rodrigueza, W.V.; Rasco, D.W.; Patnaik, A.; Papadopoulos, K.P.; Amaya, A.; Moore, T.D.; Gaylor, S.K.; Bisgaier, C.L.; Sooch, M.P.; et al. A phase 1 study of the BCL2-targeted deoxyribonucleic acid inhibitor (DNAi) PNT2258 in patients with advanced solid tumors. Cancer Chemother. Pharmacol. 2014, 73, 363–371. [Google Scholar] [CrossRef] [Green Version]
  100. Harb, W.; Lakhani, N.J.; Messmann, R.; Klencke, B.; Al-Katib, A.M. A phase 2 study of PNT2258 for treatment of relapsed or refractory B-cell malignancies. Clin. Lymphoma Myeloma Leuk. 2021, 21, 823–830. [Google Scholar] [CrossRef]
  101. ProNAi Reports Interim Data from Wolverine Phase 2 Trial of PNT2258 in DLBCL. Available online: https://www.prnewswire.com/news-releases/pronai-reports-interim-data-from-wolverine-phase-2-trial-of-pnt2258-in-dlbcl-581947101.html (accessed on 17 October 2021).
  102. Gutiérrez-Puente, Y.; Tari, A.M.; Stephens, C.; Rosenblum, M.; Guerra, R.T.; Lopez-Berestein, G. Safety, Pharmacokinetics, and tissue distribution of liposomal P-ethoxy antisense oligonucleotides targeted to Bcl-2. J. Pharmacol. Exp. Ther. 1999, 291, 865–869. [Google Scholar]
  103. Fresquet, V.; Rieger, M.; Carolis, C.; García-Barchino, M.J.; Martinez-Climent, J.A. Acquired mutations in BCL2 family proteins conferring resistance to the BH3 mimetic ABT-199 in lymphoma. Blood 2014, 123, 4111–4119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Tari-Ashizawa, A.; Guiterrez-Puente, Y.; Ford, R.J.; Lopez-Berestein, G. Activity of Bcl-2 antisense therapeutic in aggressive non-Hodgkin’ s lymphoma. In Proceedings of the AACR Annual Meeting 2017, Washington, DC, USA, 1–5 April 2017. [Google Scholar]
  105. Konopleva, M.; Tari, A.M.; Estrov, Z.; Harris, D.; Xie, Z.; Zhao, S.; López-Berestein, G.; Andreeff, M. Liposomal Bcl-2 antisense oligonucleotides enhance proliferation, sensitize acute myeloid leukemia to cytosine-arabinoside, and induce apoptosis independent of other antiapoptotic proteins. Blood 2000, 95, 3929–3938. [Google Scholar] [CrossRef]
  106. Campone, M.; Vavasseur, F.; Le Cabellec, M.T.; Meflah, K.; Vallette, F.M.; Oliver, L. Induction of chemoresistance in HL-60 cells concomitantly causes a resistance to apoptosis and the synthesis of P-glycoprotein. Leukemia 2001, 15, 1377–1387. [Google Scholar] [CrossRef] [Green Version]
  107. Kornblau, S.M.; Thall, P.F.; Estrov, Z.; Walterscheid, M.; Patel, S.; Theriault, A.; Keating, M.J.; Kantarjian, H.; Estey, E.; Andreeff, M. The prognostic impact of BCL2 protein expression in acute myelogenous leukemia varies with cytogenetics. Clin. Cancer Res. 1999, 5, 1758–1766. [Google Scholar]
  108. Frazier, K.S. Antisense oligonucleotide therapies: The promise and the challenges from a toxicologic pathologist’s perspective. Toxicol. Pathol. 2015, 43, 78–89. [Google Scholar] [CrossRef]
  109. Waters, J.S.; Webb, A.; Cunningham, D.; Clarke, P.A.; Raynaud, F.; Di Stefano, F.; Cotter, F.E. Phase I clinical and pharmacokinetic study of Bcl-2 antisense oligonucleotide therapy in patients with non-Hodgkin’s lymphoma. J. Clin. Oncol. 2000, 18, 1812–1823. [Google Scholar] [CrossRef] [PubMed]
  110. Sewing, S.; Roth, A.B.; Winter, M.; Dieckmann, A.; Bertinetti-Lapatki, C.; Tessier, Y.; McGinnis, C.; Huber, S.; Koller, E.; Ploix, C.; et al. Assessing single-stranded oligonucleotide drug-induced effects in vitro reveals key risk factors for thrombocytopenia. PLoS ONE 2017, 12, e0187574. [Google Scholar] [CrossRef] [Green Version]
  111. Nair, A.; Jacob, S. A simple practice guide for dose conversion between animals and human. J. Basic Clin. Pharm. 2016, 7, 27. [Google Scholar] [CrossRef] [Green Version]
  112. 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]
  113. Konopleva, M.; Pollyea, D.A.; Potluri, J.; Chyla, B.; Hogdal, L.; Busman, T.; McKeegan, E.; Salem, A.H.; Zhu, M.; Ricker, J.L.; et al. Efficacy and biological correlates of response in a phase II study of venetoclax monotherapy in patients with acute myelogenous leukemia. Cancer Discov. 2016, 6, 1106–1117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Fischer, K.; Al-Sawaf, O.; Hallek, M. Preventing and monitoring for tumor lysis syndrome and other toxicities of venetoclax during treatment of chronic lymphocytic leukemia. Hematol. Am. Soc. Hematol. Educ. Progr. 2020, 2020, 357. [Google Scholar] [CrossRef]
  115. Tausch, E.; Close, W.; Dolnik, A.; Bloehdorn, J.; Chyla, B.; Bullinger, L.; Döhner, H.; Mertens, D.; Stilgenbauer, S. Venetoclax Resistance and acquired BCL2 mutations in chronic lymphocytic leukemia. Haematologica 2019, 104, E434–E437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Zhao, X.; Ren, Y.; Lawlor, M.; Shah, B.D.; Park, P.M.C.; Lwin, T.; Wang, X.; Liu, K.; Wang, M.; Gao, J.; et al. BCL2 amplicon loss and transcriptional remodeling drives ABT-199 resistance in B cell lymphoma models. Cancer Cell 2019, 35, 752–766.e9. [Google Scholar] [CrossRef] [PubMed]
  117. Yue, X.Y.; Chen, Q.; He, J.S. Combination strategies to overcome resistance to the BCL2 inhibitor venetoclax in hematologic malignancies. Cancer Cell Int. 2020, 20, 1–14. [Google Scholar] [CrossRef] [PubMed]
  118. Salah, H.T.; Dinardo, C.D.; Konopleva, M.; Khoury, J.D. Potential biomarkers for treatment response to the Bcl-2 inhibitor venetoclax: State of the art and future directions. Cancers 2021, 13, 2974. [Google Scholar] [CrossRef]
  119. Choudhary, G.S.; Al-Harbi, S.; Mazumder, S.; Hill, B.T.; Smith, M.R.; Bodo, J.; Hsi, E.D.; Almasan, A. 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. [Google Scholar] [CrossRef]
  120. Lin, K.H.; Winter, P.S.; Xie, A.; Roth, C.; Martz, C.A.; Stein, E.M.; Anderson, G.R.; Tingley, J.P.; Wood, K.C. Targeting MCL-1/BCL-XL forestalls the acquisition of resistance to ABT-199 in acute myeloid leukemia. Sci. Rep. 2016, 6, 1–10. [Google Scholar] [CrossRef] [Green Version]
  121. Li, Z.; He, S.; Look, A.T. The MCL1-specific inhibitor S63845 acts synergistically with venetoclax/ABT-199 to induce apoptosis in T-cell acute lymphoblastic leukemia cells. Leukemia 2019, 33, 262–266. [Google Scholar] [CrossRef]
  122. Chen, X.; Glytsou, C.; Zhou, H.; Narang, S.; Reyna, D.E.; Lopez, A.; Sakellaropoulos, T.; Gong, Y.; Kloetgen, A.; Yap, Y.S.; et al. Targeting mitochondrial structure sensitizes acute myeloid leukemia to venetoclax treatment. Cancer Discov. 2019, 9, 890–909. [Google Scholar] [CrossRef]
  123. Sharon, D.; Cathelin, S.; Mirali, S.; Di Trani, J.M.; Yanofsky, D.J.; Keon, K.A.; Rubinstein, J.L.; Schimmer, A.D.; Ketela, T.; Chan, S.M. Inhibition of mitochondrial translation overcomes venetoclax resistance in AML through activation of the integrated stress response. Sci. Transl. Med. 2019, 11. [Google Scholar] [CrossRef] [PubMed]
  124. Blombery, P.; Anderson, M.A.; Gong, J.N.; Thijssen, R.; Birkinshaw, R.W.; Thompson, E.R.; Teh, C.E.; Nguyen, T.; Xu, Z.; Flensburg, C.; 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, 342–353. [Google Scholar] [CrossRef] [Green Version]
  125. Gagliardi, M.; Tari Ashizawa, A. The combination of liposomal Bcl-2 antisense oligonucleotide (BP1002) with decitabine is efficacious in venetoclax-resistant cells. Cancer Res. 2021, 81, 939. [Google Scholar]
  126. Ohanian, M.; Tari Ashizawa, A.; Garcia-Manero, G.; Pemmaraju, N.; Kadia, T.; Jabbour, E.; Ravandi, F.; Borthakur, G.; Andreeff, M.; Konopleva, M.; et al. Liposomal Grb2 antisense oligodeoxynucleotide (BP1001) in patients with refractory or relapsed haematological malignancies: A single-centre, open-label, dose-escalation, phase 1/1b trial. Lancet Haematol. 2018, 5, e136–e146. [Google Scholar] [CrossRef]
  127. Roca-Portoles, A.; Rodriguez-Blanco, G.; Sumpton, D.; Cloix, C.; Mullin, M.; Mackay, G.M.; O’Neill, K.; Lemgruber, L.; Luo, X.; Tait, S.W.G. Venetoclax causes metabolic reprogramming independent of BCL-2 inhibition. Cell Death Dis. 2020, 11, 616. [Google Scholar] [CrossRef]
  128. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [Green Version]
  129. Cannell, I.G.; Kong, Y.W.; Bushell, M. How do MicroRNAs regulate gene expression? Biochem. Soc. Trans. 2008, 36, 1224–1231. [Google Scholar] [CrossRef] [Green Version]
  130. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. Elegans heterochronic gene Lin-4 encodes small RNAs with antisense complementarity to Lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef]
  131. Friedman, R.C.; Farh, K.K.H.; Burge, C.B.; Bartel, D.P. Most mammalian MRNAs are conserved targets of MicroRNAs. Genome Res. 2009, 19, 92–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Peng, Y.; Croce, C.M. The role of MicroRNAs in human cancer. Signal Transduct. Target. Ther. 2016, 1, 1–9. [Google Scholar] [CrossRef] [Green Version]
  133. Lionetti, M.; Musto, P.; Di Martino, M.T.; Fabris, S.; Agnelli, L.; Todoerti, K.; Tuana, G.; Mosca, L.; Cantafio, M.E.G.; Grieco, V.; et al. Biological and clinical relevance of MiRNA expression signatures in primary plasma cell leukemia. Clin. Cancer Res. 2013, 19, 3130–3142. [Google Scholar] [CrossRef] [Green Version]
  134. Si, W.; Shen, J.; Zheng, H.; Fan, W. The role and mechanisms of action of MicroRNAs in cancer drug resistance. Clin. Epigenet. 2019, 11, 1–24. [Google Scholar] [CrossRef]
  135. Rokavec, M.; Li, H.; Jiang, L.; Hermeking, H. The P53/MiR-34 axis in development and disease. J. Mol. Cell Biol. 2014, 6, 214–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Li, X.J.; Ren, R.J.; Tang, J.H. MicroRNA-34a: A potential therapeutic target in human cancer. Cell Death Dis. 2014, 5, e1327. [Google Scholar] [CrossRef] [PubMed]
  137. Bader, A.G.; Brown, D.; Stoudemire, J.; Lammers, P. Developing therapeutic MicroRNAs for cancer. Gene Ther. 2011, 18, 1121–1126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Cole, K.A.; Attiyeh, E.F.; Mosse, Y.P.; Laquaglia, M.J.; Diskin, S.J.; Brodeur, G.M.; Maris, J.M. A functional screen identifies MiR-34a as a candidate neuroblastoma tumor suppressor gene. Mol. Cancer Res. 2008, 6, 735–742. [Google Scholar] [CrossRef] [Green Version]
  139. Yang, F.; Li, Q.J.; Gong, Z.B.; Zhou, L.; You, N.; Wang, S.; Li, X.L.; Li, J.J.; An, J.Z.; Wang, D.S.; et al. MicroRNA-34a targets Bcl-2 and sensitizes human hepatocellular carcinoma cells to sorafenib treatment. Technol. Cancer Res. Treat. 2014, 13, 77–86. [Google Scholar] [CrossRef]
  140. Lin, X.; Guan, H.; Huang, Z.; Liu, J.; Li, H.; Wei, G.; Cao, X.; Li, Y. Downregulation of Bcl-2 expression by Mir-34a mediates palmitate-induced Min6 cells apoptosis. J. Diabetes Res. 2014, 2014, 1–7. [Google Scholar] [CrossRef] [Green Version]
  141. Zhao, Y.; Wang, X. MiR-34a targets BCL-2 to suppress the migration and invasion of sinonasal squamous cell carcinoma. Oncol. Lett. 2018, 16, 6566–6572. [Google Scholar] [CrossRef]
  142. Daige, C.L.; Wiggins, J.F.; Priddy, L.; Nelligan-Davis, T.; Zhao, J.; Brown, D. Systemic delivery of a MiR34a mimic as a potential therapeutic for liver cancer. Mol. Cancer Ther. 2014, 13, 2352–2360. [Google Scholar] [CrossRef] [Green Version]
  143. Craig, V.J.; Tzankov, A.; Flori, M.; Schmid, C.A.; Bader, A.G.; Müller, A. Systemic MicroRNA-34a delivery induces apoptosis and abrogates growth of diffuse large B-cell lymphoma in vivo. Leukemia 2012, 26, 2421–2424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Wiggins, J.F.; Ruffino, L.; Kelnar, K.; Omotola, M.; Patrawala, L.; Brown, D.; Bader, A.G. Development of a lung cancer therapeutic based on the tumor suppressor MicroRNA-34. Cancer Res. 2010, 70, 5923–5930. [Google Scholar] [CrossRef] [Green Version]
  145. Hong, D.S.; Kang, Y.; Borad, M.; Sachdev, J.; Ejadi, S.; Lim, H.Y.; Brenner, A.J.; Park, K.; Lee, J.; Kim, T.; et al. Phase 1 study of MRX34, a liposomal MiR-34a mimic, in patients with advanced solid tumours. Br. J. Cancer 2020, 122, 1630–1637. [Google Scholar] [CrossRef]
  146. Lovat, F.; Fassan, M.; Sacchi, D.; Ranganathan, P.; Palamarchuk, A.; Bill, M.; Karunasiri, M.; Gasparini, P.; Nigita, G.; Distefano, R.; et al. Knockout of both MiR-15/16 loci induces acute myeloid leukemia. Proc. Natl. Acad. Sci. USA 2018, 115, 13069–13074. [Google Scholar] [CrossRef] [Green Version]
  147. Calin, G.A.; Dumitru, C.D.; Shimizu, M.; Bichi, R.; Zupo, S.; Noch, E.; Aldler, H.; Rattan, S.; Keating, M.; Rai, K.; et al. Frequent deletions and down-regulation of Micro-RNA genes MiR15 and MiR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. USA 2002, 99, 15524–15529. [Google Scholar] [CrossRef] [Green Version]
  148. Reid, G.; Pel, M.E.; Kirschner, M.B.; Cheng, Y.Y.; Mugridge, N.; Weiss, J.; Williams, M.; Wright, C.; Edelman, J.J.B.; Vallely, M.P.; et al. Restoring expression of MiR-16: A novel approach to therapy for malignant pleural mesothelioma. Ann. Oncol. 2013, 24, 3128–3135. [Google Scholar] [CrossRef] [PubMed]
  149. Van Zandwijk, N.; Pavlakis, N.; Kao, S.C.; Linton, A.; Boyer, M.J.; Clarke, S.; Huynh, Y.; Chrzanowska, A.; Fulham, M.J.; Bailey, D.L.; et al. Safety and activity of MicroRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: A first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017, 18, 1386–1396. [Google Scholar] [CrossRef]
  150. Arrighetti, N.; Corbo, C.; Evangelopoulos, M.; Tasciotti, E. Exosome-like nanovectors for drug delivery in cancer. Curr. Med. Chem. 2020, 26, 6132–6148. [Google Scholar] [CrossRef]
  151. Wu, C.H.; Li, J.; Li, L.; Sun, J.; Fabbri, M.; Wayne, A.S.; Seeger, R.C.; Jong, A.Y. Extracellular vesicles derived from natural killer cells use multiple cytotoxic proteins and killing mechanisms to target cancer cells. J. Extracell. Vesicles 2019, 8, 1588538. [Google Scholar] [CrossRef] [Green Version]
  152. Di Pace, A.L.; Tumino, N.; Besi, F.; Alicata, C.; Conti, L.A.; Munari, E.; Maggi, E.; Vacca, P.; Moretta, L. Characterization of human NK cell-derived exosomes: Role of DNAM1 receptor in exosome-mediated cytotoxicity against tumor. Cancers 2020, 12, 661. [Google Scholar] [CrossRef] [Green Version]
  153. Jong, A.Y.; Wu, C.H.; Li, J.; Sun, J.; Fabbri, M.; Wayne, A.S.; Seeger, R.C. Large-scale isolation and cytotoxicity of extracellular vesicles derived from activated human natural killer cells. J. Extracell. Vesicles 2017, 6, 1294368. [Google Scholar] [CrossRef] [Green Version]
  154. Kaban, K.; Hinterleitner, C.; Zhou, Y.; Salva, E.; Kantarci, A.G.; Salih, H.R.; Märklin, M. Therapeutic silencing of Bcl-2 using Nk cell-derived exosomes as a novel therapeutic approach in breast cancer. Cancers 2021, 13, 2397. [Google Scholar] [CrossRef] [PubMed]
  155. Xu, H.; Liao, C.; Liang, S.; Ye, B.-C. A novel peptide-equipped exosomes platform for delivery of antisense oligonucleotides. ACS Appl. Mater. Interfaces 2021, 13, 10760–10767. [Google Scholar] [CrossRef] [PubMed]
  156. Sercombe, L.; Veerati, T.; Moheimani, F.; Wu, S.Y.; Sood, A.K.; Hua, S. Advances and challenges of liposome assisted drug delivery. Front. Pharmacol. 2015, 6, 286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Briolay, T.; Petithomme, T.; Fouet, M.; Nguyen-Pham, N.; Blanquart, C.; Boisgerault, N. Delivery of cancer therapies by synthetic and bio-inspired nanovectors. Mol. Cancer 2021, 20, 55. [Google Scholar] [CrossRef] [PubMed]
  158. Zhao, Y.; Liu, P.; Tan, H.; Chen, X.; Wang, Q.; Chen, T. Exosomes as smart nanoplatforms for diagnosis and therapy of cancer. Front. Oncol. 2021, 11. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Bcl-2 family proteins and apoptosis overview. (A) Bcl-2 family proteins are composed of anti-apoptotic proteins (multiple BH domains) and pro-apoptotic proteins (multi-BH domain effectors and BH3-only initiators). (B) Under normal conditions anti-apoptotic proteins inhibit the activity of pro-apoptotic effectors, impeding mitochondrial outer membrane permeabilization (MOMP) and cytochrome C release. Cellular stress activates pro-apoptotic initiators which bind to and inhibit anti-apoptotic proteins, allowing pro-apoptotic effectors to proceed.
Figure 1. Bcl-2 family proteins and apoptosis overview. (A) Bcl-2 family proteins are composed of anti-apoptotic proteins (multiple BH domains) and pro-apoptotic proteins (multi-BH domain effectors and BH3-only initiators). (B) Under normal conditions anti-apoptotic proteins inhibit the activity of pro-apoptotic effectors, impeding mitochondrial outer membrane permeabilization (MOMP) and cytochrome C release. Cellular stress activates pro-apoptotic initiators which bind to and inhibit anti-apoptotic proteins, allowing pro-apoptotic effectors to proceed.
Pharmaceutics 14 00097 g001
Figure 2. Clinical trial timeline of Bcl-2 ASO and miRNA. Clinical trial start and end dates are marked for each drug. Arrows indicate ongoing clinical trials; blunted arrows represent trials that have been terminated.
Figure 2. Clinical trial timeline of Bcl-2 ASO and miRNA. Clinical trial start and end dates are marked for each drug. Arrows indicate ongoing clinical trials; blunted arrows represent trials that have been terminated.
Pharmaceutics 14 00097 g002
Figure 3. Bcl-2 oligonucleotide delivery. (A) Gapmer ASO have nuclease resistant, second, and third generation modifications at the 5′ and 3′ ends. (B) Liposomes are composed of phospholipid bilayers (one or multi bilayers; 20–1000 nm in diameter). Depending on the lipids used, liposomes can be cationic, anionic, neutral or pH-sensitive. Hydrophobic cargo is encapsulated within the lipid bilayers, and hydrophilic cargo within the aqueous core or tethered to liposome surface. Additional surface modifications include polyethylene glycol to prolong blood circulation and antibodies or targeting proteins to enhance cancer cell specific delivery. (C) Minicells are anucleated, non-dividing, and metabolically active cells (100–300 nm) that bud from abnormally dividing bacteria. Cancer cell targeting can be achieved by the addition of specific peptides or ligands to the minicell surface. (D) Exosomes are extracellular vesicles released from cells (50–150 nm). They carry a range of proteins, lipids, and nucleotides, some of which are specific to their cell of origin. Common markers found in exosomes include major histocompatibility complex (MHC), heat shock proteins, tetraspanins, phosphatidylserine, actin, and cholesterol.
Figure 3. Bcl-2 oligonucleotide delivery. (A) Gapmer ASO have nuclease resistant, second, and third generation modifications at the 5′ and 3′ ends. (B) Liposomes are composed of phospholipid bilayers (one or multi bilayers; 20–1000 nm in diameter). Depending on the lipids used, liposomes can be cationic, anionic, neutral or pH-sensitive. Hydrophobic cargo is encapsulated within the lipid bilayers, and hydrophilic cargo within the aqueous core or tethered to liposome surface. Additional surface modifications include polyethylene glycol to prolong blood circulation and antibodies or targeting proteins to enhance cancer cell specific delivery. (C) Minicells are anucleated, non-dividing, and metabolically active cells (100–300 nm) that bud from abnormally dividing bacteria. Cancer cell targeting can be achieved by the addition of specific peptides or ligands to the minicell surface. (D) Exosomes are extracellular vesicles released from cells (50–150 nm). They carry a range of proteins, lipids, and nucleotides, some of which are specific to their cell of origin. Common markers found in exosomes include major histocompatibility complex (MHC), heat shock proteins, tetraspanins, phosphatidylserine, actin, and cholesterol.
Pharmaceutics 14 00097 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gagliardi, M.; Ashizawa, A.T. Making Sense of Antisense Oligonucleotide Therapeutics Targeting Bcl-2. Pharmaceutics 2022, 14, 97. https://doi.org/10.3390/pharmaceutics14010097

AMA Style

Gagliardi M, Ashizawa AT. Making Sense of Antisense Oligonucleotide Therapeutics Targeting Bcl-2. Pharmaceutics. 2022; 14(1):97. https://doi.org/10.3390/pharmaceutics14010097

Chicago/Turabian Style

Gagliardi, Maria, and Ana Tari Ashizawa. 2022. "Making Sense of Antisense Oligonucleotide Therapeutics Targeting Bcl-2" Pharmaceutics 14, no. 1: 97. https://doi.org/10.3390/pharmaceutics14010097

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