NQO1highCATlow provides a therapeutic window for utilizing of β-Lap in EC.
In our previous study, it had been confirmed that nMIL-100(Fe) catalyzed exogenous H2O2 through Fenton-like reaction to produce ·OH, which significantly inhibited the survival of KLE type II EC cells [29]. Lower expression of CAT (an enzymatic H2O2 scavenger) in KLE cells may be associated with high cell lethality, which forced cells being subjected to excessive ROS attack. In addition, since b-Lap was a NQO1-dependent bioactivatable drug, high expression of NQO1 was a prerequisite for b-Lap based antitumor therapy. Whether there was a NQO1highCATlow therapeutic window for b-Lap application to EC therapy. To probe this aspect, both NQO1 and CAT transcript levels were initially analyzed from TCGA database (https://tcga-data.nci.nih.gov/tcga/). Compared with normal tissues, NQO1 expression was obviously elevated accompanied by a decrease in CAT levels in tumor tissues, regardless of cancer individual stages and histological subtypes (Fig. 1a-c). Remarkably, low expression levels of CAT were strongly associated with the advanced clinical stages (Stage 4) and the histological subtype (Serous, type II EC) with poorer prognosis (p=5.76×10-9 and p=1.65×10-12, respectively). From theses, we speculated that in this type of EC, there may be a vulnerability in the endogenous H2O2 scavenging mechanism dependent on CAT. In parallel, we also observed in Fig. 1d the expression CAT was positively correlated with both ER and PR levels (Spearman R CAT/ER=0.47, p=6.2×10-11; Spearman R CAT/PR=0.39, p=9.6×10-8, respectively). The correlation between the expression of CAT and ER and / or PR further confirmed the above speculation by the fact that type II EC was mostly negative for ER and PR. The expression of NQO1 and CAT did not have direct carcinogenic or anti-cancer activity and were not significantly associated with overall survival in EC patients (Fig. 1e). Instead, as bioactive drug targets, they are involved in the anti-cancer strategy of regulating ROS: enhancing drug induction of ROS production (NQO1high) and inhibiting the antioxidant defense (CAT low) of tumor cells. Subsequently, to verify the existence of our TCGA database-based findings, we first assessed the expression status of NQO1 and CAT in tissue samples, and found that compared with normal endometrial tissues, there was such a treatment window for NQO1high CAT low in EC regardless of clinical stages (Fig. 1f). On top of evaluation in tissues, the levels of NQO1 and CAT were further validated in human EC cell lines including type I EC cells: well-differentiated Ishikawa (ER+/PR+), intermediately-differentiated HEC-1A (ER±/PR-) and HEC-1B (ER±/PR±); poorly-differentiated AN3CA (ER-/PR-) and type II EC cells: KLE (ER-/PR-). Human endometrial epithelial cells (EEC) were served as a normal cell control (Fig. 1g). The results showed such a high NQO1/CAT ratio was more prominent, especially in advanced EC tissue (II, III stages) and in KLE and AN3CA EC cells. Taken together, a NQO1 high CAT low therapeutic window for the use of b-Lap had been found in EC, especially in the advanced type I and type II EC. These data provided a rationale for developing therapeutic strategies for β-Lap induced tumor-specific ROS production in these EC.
β-Lap suppressed EC cell lines growth in an NQO1-dependent manner.
Currently, b-Lap is undergoing clinical trials of monotherapy or combination therapy in patients with advanced NQO1+ solid tumors (linicalTrials.gov identifiers NCT02514031 and NCT01502800). However, there has been little discussion about the use of b-Lap in EC. Thus, we first validated EC cell lines with high expression of NQO1 (Ishikawa, AN3CA, HEC-1B and KLE) were more sensitive to β-Lap therapy (Fig. 2a). That may be due to β-Lap-induced cell cycle arrest at the G1 phase, as confirmed by the concentration-dependent down-regulated Cyclin D1, c-Myc and upregulated P27 Kip1 levels (Fig. 2i). At concentrations of 8 mM or higher, b-Lap significantly inhibited cell proliferation, even lead to cell lethality. While NQO1-absent normal human endometrial epithelial cells EEC and NQO1-low expressing 2 cervical cancer cell lines (HeLa and ME-180) (Additional file 1: Fig. S1), were resistant to β-Lap (Fig. 2a). Dicoumarol (DIC, an inhibitor of NQO1) spared NQO1high EC cells from b-Lap-induced lethality (Fig. 2b), partially recovered the colony-forming ability of KLE cells (Fig. 2c and Additional file 1: Fig. S2). One possible reason could be that b-Lap was not only an excellent substrate for NQO1, but also a well-characterized substrate for one-electron oxidoreductases (e.g. NADH: cytochrome b5 reductase, b5R), which simultaneously induced other one-electron redox reactions to further oxidize the biological macromolecules, such as DNA, RNA or proteins [21]. On top of that, the b-Lap enhanced-intracellular ROS levels restored to the observed baseline after DIC exposure (Fig. 2d). SiRNA-mediated knockdown of NQO1 deprived KLE cells sensitivity to β-lap exposure (Fig. 2e). While overexpressing NQO1 in HeLa cells endowed the susceptibility to β-Lap, and DIC reversed the effect of β-Lap on NQO1-overexpressed HeLa cells (Fig. 2f). Likewise, β-Lap significantly upregulated the ROS levels in NQO1-overexpressed HeLa cells and specific inhibition of NQO1 activity by DIC addition spared the above effect. Moreover, β-Lap induced KLE cells apoptosis in time- and concentration-dependent manner (Fig. 2h). Apoptosis marker protein cleaved Caspase-3 (cCaspase-3) and its cleaved product cleaved PARP (cPARP) were progressively accumulated 48 h after various concentrations β-Lap treatment (Fig. 2i). However, a lethal dose of β-Lap had been shown to induce “programmed necrosis” independent of caspase activation [30]. Thus, it is necessary to carry out another independent study to further reveal the correlation between the β-Lap lethality and caspase activation in EC. Then, H2O2 (500 mM, 15min) served as a positive stimulus to induce DNA damage. Following different treatments annotated with 1-6 in Fig. 2j for 48 h, the levels of phospho-histone H2A.X (γ-H2A.X) (Ser 139), an indicator of DNA double-strand breaks, were assayed by immunoblots and immunofluorescence, respectively. Similar to H2O2 exposure, γ-H2A.X expression was upregulated after β-Lap stimulation (group 2 vs group 5) compared with control or DIC treatment alone (group 1 and group 3). However, the salient difference was that β-Lap induced enhancement of γ-H2A.X expression could be eliminated by the NQO1 inhibitor-DIC, while the effect caused by H2O2 was not influenced by DIC (group 4 vs group 6). Given that NQO1 catalyzed β-Lap to incur a futile redox cycle, resulting in excessive H2O2 production and 2-fold consumption of NAD(P)H [26]. As displayed in Fig. 2k, β-Lap promoted the concentration-dependent release of H2O2 both in KLE and AN3CA cells only in short-term treatment, accompanied by a remarkable increase in intracellular NAD+/NADH ratio (Fig. 2l). Collectively, these data demonstrated that in vitro β-Lap induced H2O2 production and depleted intracellular NADH in an NQO1-dependent manner in EC cells. The elevated H2O2 caused extensive DNA double-strand breaks that induced a broad spectrum of DNA damage, which further stimulated hyperactivation of PARP to prevent massive DNA damage from repairing, leading to severe NAD+/ATP pool losses [[31], [32]]. These cumulative oxidative damage in turn triggered EC cell death referred to as “programmed necrosis” [33] or “necroptosis” [34], exclusive to the NQO1 bioactivatable drugs [35]. Could β-Lap be used as a monotherapy for patients with NQO1+ EC, such as monoclonal antibodies and small molecules against typical targets (e.g. epidermal growth factor receptor: EGFR, vascular endothelial growth factor receptor: VEGFR)? Unfortunately, hemolytic anemia appeared as the primary dose-limiting toxicity (DLT) of prodrug of β-Lap during clinical trials, which was suspected to occur in almost all patients receiving high doses. Therefore, the maximum tolerated dose (MTD) of ARQ 501 and ARQ 761 had to be designed to 390 mg/m2 to alleviate the side effects. However, ARQ761 monotherapy was only moderately effective, leading to mild radiographic response in about 20% of patients, but no partial response [36]. Although the combination of β-Lap analogs and other chemodrugs such as PARP inhibitors [26], glutamine metabolism inhibitors [37], alkylating agents [38] and nicotinamide phosphoribosyl transferase inhibitors [39], exhibited superior synergistic antitumor effects in vitro and in vivo, the potential overlapping toxicity should be noted.
Selective targeting in EC cells of Fenton-like catalyst RGD-nMIL-100(Fe)
As mentioned above, due to dramatic NAD+ depletion and ATP pool losses, NQO1-driven futile redox cycle may fail to function, resulting in an interruption of H2O2 output to the EC tumor site. Known from ROS electrochemistry, the most oxidizing one-electron oxidants (radical species, such as O2·−and ·OH) was ·OH, which had a reduction potential E 0'(·OH, H+/H2O) = 2310 mV, obviously higher than E 0'(H2O2, H+/H2O, ·OH) = 320 mV [[7], [40]]. In addition, the reactivity of two-electron oxidants (nonradical species, such as H2O2) was determined by kinetic considerations. Due to the requirements for higher reaction activation energy and relatively slower reaction rate, H2O2 was actually only moderately reactive. Moreover, in clinical trials, β-Lap at therapeutic doses was associated with varying degrees of adverse effects, including anemia (79%), fatigue (45%), hypoxia (33%), nausea (17%), and vomiting (17%) [36]. Encouragingly, β-Lap prodrugs prestored inside engineered micelles [41] and polymeric implants [42] enabled efficient intra-tumoral delivery and increased its tolerability. How could we reduce the dose of β-Lap without weakening oxidative stress it mediates? Therefore, the introduction of Fenton-like reagents into NQO1+ tumor sites could promote the reductive cleavage of excessive H2O2 induced by subtherapeutic dose β-Lap to highly reactive ·OH, which would further up-regulate intracellular redox state to expand the oxidative damage. MOF(Fe) with high porosity, high specific surface area and abundant iron active centers were selected as Fenton-like nanocatalyst, which had been confirmed in our previous work [29]. RGD (arginine-glycine-aspartic) peptides offered high affinity for integrin αVβ3 receptor, which overexpressed on the surface of neovascularization and a broad variety of tumor cells [[43], [44], [45], [46]]. To increase cellular internalization, nanoscale MIL-100(Fe) (abbreviated as nMIL-100(Fe) below) was modified with FITC-labeled cyclic RGD motif to obtain cRGD-functionalized nMIL-100(Fe)s (RGD-nMIL-100(Fe)) with tumor target ability (Fig 3a). Not only the morphologies (size and shape) of nMIL-100(Fe) particles were not significantly altered before and after conjugation with cRGD (Fig 3b), but also the catalytic activities (Fig 3c-g). Fenton nanozyme effects of RGD-nMIL-100(Fe) particles were investigated using 2,2′-azino-bis(3-ethylbenzthiazdine-6)-sulfonic acid (ABTS) and 3,3’,5,5’ - tetramethylbenzedine (TMB) as substrates. Compared with the particles or H2O2 alone, RGD-nMIL-100(Fe) could oxidize the two substrates separately into oxidized ABTS (oxABTS) and TMB (oxTMB) in the presence of H2O2, the maximum absorption peaks of which were at 500-900nm and 652 nm respectively (Fig. 3c, d). This data indirectly reflected H2O2 was converted into highly oxidative ·OH catalyzed by RGD-nMIL-100(Fe), as noted with nMIL-100(Fe) [29]. Furthermore, our nanocatalysts exerted RGD-nMIL-100(Fe) and H2O2 concentration-dependent catalytical activities (Fig. 3e, f). Besides, the generating of ·OH radicals with a shorter half-life (10-9 s)was assessed by electron spin resonance (ESR) by adding 5,5-dimethyl-1-pyrroline-N-oxide (DMPO) as the radical trapping agent [47]. As presented in Figure 3g, the typical 1:2:2:1 spectra of DMPO/·OH adducts was only detected in the RGD-nMIL-100(Fe)+H2O2+DMPO group, while no signal could be captured in RGD-nMIL-100(Fe)+DMPO, H2O2+DMPO and the DMPO groups, proving the high catalytic capability of RGD-nMIL-100(Fe) for ·OH production. Then, in order to determine the optimal target cells for RGD-nMIL-100(Fe) nanoparticles, the obtained EC cells were analysed for integrin αV and β3 level by immunoblots, using EEC as a negative control (Fig. 3h). The results revealed that both integrin αV and β3 were significantly overexpressed in KLE, Ishikawa, AN3CA and HEC-1A cells, which were regarded as RGD-positive target cells. While RL95-2 and HEC-1B (expressing integrin αV only) were treated as RGD-negative target cells. Next, cellular internalization efficiency of RGD-nMIL-100(Fe) nanoparticles was measured by immunofluorescence. Pearson’s correlation coefficient (Rr) and Manders’ Overlap coefficient (M1) at the white solid line were calculated in the colocalization channel, by Image J software plug-in, Colocalization-Finder (scatter plots) and Plot Profile (line graphs). After incubation for 6 h, the FITC-labeled nanoparticles (green spots) were found inside the KLE cells. This also reflected the successful modification of nanoparticles by free FITC or FITC-labeled RGD motif, because they didn't contain any fluorophore themselves, as shown in the control group (Fig. 3i). Most of these were scattered in the cytoplasm and almost completely overlapped with cytoskeletal protein-actin (red fluorescence, M1=0.95 vs 0.98, respectively), while a few entered the nuclei (blue fluorescence). Compared to nMIL-100(Fe), RGD-functionalized nMIL-100(Fe) nanoparticles achieved a high uptake rate of KLE cells (Rr = 0.50 vs 0.12, respectively). However, this discrepancy in internalization efficacy attributed to RGD modification was not as pronounced in RGD-negative target-HEC-1B cells as that observed in KLE cells, as shown in the bar graphs (Additional file 1: Fig. S3). Collectively, these data demonstrated that the successful modification of RGD peptides endowed Fenton nanocatalyst nMIL-100(Fe) with EC-targeting ability without compromising its catalytic performance, which set the stage for efficient cellular uptake of nanoparticles and their subsequent chemical catalysis.
Amplified ROS signal combined with Mdivi-1 enhanced the anticancer effect of NQO1high EC cells
Although the concentration of H2O2 in tumor sites (∼50-100 μmol/L) was higher than that in normal tissues, it was not sufficient for effective chemodynamic reactions [20]. Also, the aforementioned data had verified that β-Lap was a potent H2O2-generating agent in NQO1high EC. Hence, it is reasonable to speculate that massive NQO1 initiated-H2O2 could be converted into highly oxidizing ·OH by the internalized RGD-MOF(Fe) mediated-Fenton reaction, resulting in step-wise amplified oxidative damage to NQO1high EC. Thus, RGD-positive target cells KLE (type II EC) and AN3CA (metastatic type I EC) of NQO1highCATlow were selected as model cells to investigate the oxidative stress caused by β-Lap-combined with nMIL-100(Fe) nanoparticles. As shown in Fig 2a and 2i, 2.0 mM β-Lap had little effect on the growth of NQO1high EC cells, while 25mg ml-1 nMIL-100(Fe) had the ability to catalyze H2O2 to generate ·OH (Fig 3e). To verify the “1+1 > 2” combined effect, the dosage of nMIL-100(Fe) and β-Lap was reduced to 25 mg ml-1 and 2.0 mM, respectively. We first evaluated the mean fluorescence intensities (MFI) of DCFH-DA probe for intracellular ROS accumulation in response to the established exposures (Fig 4a). As expected, compared with MOF(Fe) nanoparticles or β-Lap treated alone groups and control groups, DCFH-DA fluorescence was brighter in the combined treatment group, especially in the RGD-nMIL-100(Fe) addition group. This indicated that in NQO1high EC cells, the co-treatment of MOF(Fe) nanoparticles and β-Lap could catalyze Fenton like-reaction to increase the production of intracellular ROS. The consistent results were further verified in cell viability assessment of both KLE and AN3CA cells (Fig 4b). Interestingly, the same dose of RGD-nMIL-100(Fe) alone appeared to achieve or even exceed the effect of nMIL-100(Fe) combined with subtherapeutic dose of β-Lap, while 50 mg ml-1 RGD-nMIL-100(Fe) combined group had the lowest proliferation inhibition rate. The possible reason was that due to higher internalization, the increased intracellular content and prolonged retention of RGD-nMIL-100(Fe) nanozyme in turn reduced the threshold of Fenton reaction in NQO1high EC cells. This further elevated the efficiency of intracellular H2O2 conversion into ·OH, whether H2O2 was endogenous or newly generated from NQO1-dependent futile redox cycles. Given that autophagy was a stumbling block in chemodynamic antitumor therapy involving MOF(Fe) [29]. Additionally, microtubule-associated protein 1 light chain 3B (LC3B)-II/LC3B-I ratio was significantly upregulated in β-Lap-treated KLE cells (Additional file 1: Fig. S4), suggesting β-Lap induced intracellular autophagy activation [48]. Coinciding with Mdivi-1 (a mitophagy inhibitor) would probably ensure better anticancer efficacy. Mdivi-1 addition (10 mM, 1 h) first increased the percentage of both KLE and AN3CA cell proliferation repression in almost all treated groups. Albeit cell viability was slightly suppressed by Mdivi-1 pretreatment combined with non-lethal dose β-Lap (1.0 mM). There was no overt difference between this combination arm and Mdivi-1 alone group. This indicated that the inhibitory effect mainly attributed to autophagy block instead of β-Lap mediated oxidative stress. Additionally, both cell growth rates were the lowest when Mdivi-1, β-Lap and MOF(Fe) were present together, especially in the group containing RGD-nMIL-100(Fe) nanoparticles (Fig 4c). In line with the CCK-8 assays, the aforementioned combination of “Mdivi-1+RGD-nMIL-100(Fe)+β-Lap” resulted in significantly more apoptosis in KLE cells, especially after 48h of treatment (Fig 4d). Similarly, the Calcein AM probe staining indicated that compared with exposure to either solitary or any two treatments, RGD-nMIL-100(Fe) in combination with even 1.0 mM β-Lap significantly increased the relative proportion of cells with PI (red) staining for dead and apoptotic cells, aided by Mdivi-1 (Fig 4e). Besides, the expression of proliferation-related c-Myc and cyclinD1 were accordingly down-regulated; however, although caspase-3 remained activated, the substrate PARP, which was intended to repair broken DNA, was not reactively upregulated (Fig 4f). This was characteristic of “programmed necrosis” independent of caspase activation. As mentioned above, the anticancer strategy regulating intracellular ROS by "enhancing offense and weakening defense" was worth promoting in the treatment of type II EC.
Investigation of synergistic antitumor effect in vivo
Inspired by the excellent synergistic antitumor effects in vitro, we next assessed the anti-tumor efficacy and systemic toxicity of different treatments in vivo (Fig. 5a). The nude mice bearing KLE tumor were randomly divided into 5 groups: PBS, RGD-nMIL-100(Fe) (RM), β-Lap, RM+β-Lap and Mdivi-1+RM+β-Lap (n=5/group). Compared to the PBS-treated mice, similar tumor growth was observed in the RM treated group (Fig. 5b), indicating that RM exposure alone had a negligible effect on tumor growth. Tumor growth was reduced in mice treated with β-Lap alone or RM+β-Lap, with tumor growth inhibition (TGI) percentages of 41.19% and 46.25%, respectively at endpoint. These two groups had suboptimal anti-tumor effects, which might be due to the low effective concentration of β-Lap at the tumor site and fast clearance in the blood stream. On the other hand, the NQO1-dependent ·OH mediated by RGD-nMIL-100(Fe) combining with β-Lap had a short half-life and also compensatively activated several antioxidant defense mechanisms. Excitingly, the TGI of the “Mdivi-1+RM+β-Lap” group was 85.92%, suggesting that this group-mediated therapy was more efficient than free RM or β-Lap or RM+β-Lap, which could be explained by prior blockade of self-antioxidant defense modulation-mitophagy and subsequent stepwise increasing oxidation intensity arising from the increased cellular internalization capability and highly efficient Fenton-like reaction of RM nanoparticles. Meanwhile, no differences in body weight were observed among each group (Fig. 5c). Tumor weight of all the treatment groups were measured at the experiment endpoint (day 17) from euthanized mice, agreeing well with the aforementioned tumor volume results. The minimal tumor weight was formed in the “Mdivi-1+RM+β-Lap” -treated group, demonstrating again that the highest tumor inhibition ratio in this group (Fig. 5d and Additional file 1: Fig. S5). The above results firmly investigated that the anti-tumor efficacy of combined therapy with RM+β-Lap-mediated CDT and mitophagy inhibition by Mdivi-1 was remarkable. As illustrated in Fig. 5e, immunohistochemical (IHC) staining of KLE tumor sections from the mice received monotherapy (RM: RGD-nMIL-100(Fe) and β-Lap) or combination therapy (RM+β-Lap) displayed that Parkin (brown), a key protein in activated PINK1/Parkin mitophagy pathway, was variably upregulated in comparison to PBS-treated group. These enhanced CDT could trigger cellular protective mitophagy effect to varying extent, as noted in our previous study [29]. Pretreatment with Mdivi-1 (10mg/kg) markedly suppressed the expression of Parkin, which might be responsible for the higher anti-tumor efficacy of the Mdivi-1+RM+β-Lap administrations. Additionally, the Ki-67 IHC and staining transferase-mediated dUTP nick end-labeling (TUNEL) assays were used to further detect the proliferation and apoptosis of KLE xenograft tumors, respectively. The lowest proliferating cells (brown) and the most apoptotic cells (green) were observed in the Mdivi-1+RM+β-Lap group. This revealed that anticancer efficacy was due to reduced cell proliferation and aggravated cell apoptosis. Notwithstanding, haematoxylin and eosin (H&E) staining showed no obvious signs of morphological changes or tissue damages in the heart, liver, spleen, lung and kidney of mice after 17 days of different treatments (Fig. 5f). As well, the corresponding blood biochemical indexes were in the normal ranges (Additional file 1: Fig. S6). The above results suggested that NQO1-dependent Fenton nano-catalyst initiated-CDT combined with Mdivi-1 mediated mitophagy blockade exerted convincing synergistic antitumor effects without organ toxicity in type II EC.
In summary, these insights offered additional therapeutic avenues for EC patients, especially for those with refractory EC (e.g. type II EC, advanced, recurrent, or persistent EC) and with demands for fertility-sparing. We reported for the first time there was a NQO1highCATlow therapeutic window in EC for the use of β-Lap, the clinical forms of which (ARQ 501 or ARQ 761) were being tested in clinical trials in other NQO1+ tumors (ClinicalTrials.gov identifiers NCT02514031 and NCT01502800). In vitro β-Lap exerted selectively antineoplastic activity by NQO1 mediated-EC cell lethality, but no killing effect on NQO1- normal cells. Taking into account the prominent dose-limiting toxicity (DLT) of β-Lap analogues in clinical trials, we found a shortcut to reduce the amount of β-Lap without weakening its mediated oxidative stress as follows: RGD-functionalized Fenton nanocatalyst-nMIL-100(Fe) was introduced to catalyze the decomposition of intracellular accumulated H2O2 (including intrinsic and the newly produced by NQO1-catalyzed nonlethal dose of β-Lap) into higher oxidative activity ·OH. Such a process from an increase in the abundance of ROS (H2O2 levels) to escalation in oxidation intensity (·OH generation) stepwise amplified the oxidation signals released by NQO1+ cells. In turn, the enhanced intracellular “oxidative attack” broke the original redox equilibrium within the TME to exert greater cytotoxic effects on both NQO1+ and neighboring NQO1- EC cells via a bystander effect. Lastly, based on our previous findings, the addition of Mdivi-1 spared mitophagy-initiated “antioxidant defense”. Taken together, our current data highlights the combination of nonlethal dose of β-Lap plus nMIL-100(Fe) with Mdivi-1 pretreatment represented an encouraging synergistic anticancer effect of “1 + 1 + 1 > 3”.
Nevertheless, the present work did have potential limitations. As a preliminary exploration of EC therapy, the blood circulation of RGD-nMIL-100(Fe) nanoparticles hadn't been taken into account. Thus, only local intra-tumoral injection was performed for different formulations in vivo. Besides, although RGD-modified nMIL-100(Fe) had a satisfactory targeting effect at the cellular level, the ability of its active targeting tumor tissue still needed to be further warranted in vivo. Thirdly, there was a lack of more precise quantitative analysis methods such as Chou-Talalay to evaluate the synergistic effect of drug combination, so as to provide a more accurate reference for guiding rational drug use in the follow-up. Despite its exploratory nature, this study offered some valuable insights into the establishment of effective treatment strategies for refractory or special types of EC.