Next Article in Journal
The Preparation and Structure Analysis Methods of Natural Polysaccharides of Plants and Fungi: A Review of Recent Development
Next Article in Special Issue
Heterocycles in Medicinal Chemistry
Previous Article in Journal
The Effect of Carbonic Maceration during Winemaking on the Color, Aroma and Sensory Properties of ‘Muscat Hamburg’ Wine
Previous Article in Special Issue
Fluorimetric Properties of 3-Aminoflavone Biomolecule (3-AF). X-ray Crystal Structure of New Polymorph of 3-AF
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Analysis of the Mechanisms of Action of Naphthoquinone-Based Anti-Acute Myeloid Leukemia Chemotherapeutics

1
Department of Medicine Division of Hematology/Oncology University of Maryland School of Medicine, Baltimore, MD 21201, USA
2
Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 South Greene Street, Baltimore, MD 21201, USA
3
Department of Chemistry, McDaniel College, 2 College Hill, Westminster, MD 21157, USA
*
Author to whom correspondence should be addressed.
Molecules 2019, 24(17), 3121; https://doi.org/10.3390/molecules24173121
Submission received: 3 August 2019 / Revised: 24 August 2019 / Accepted: 25 August 2019 / Published: 28 August 2019
(This article belongs to the Special Issue Heterocycles in Medicinal Chemistry)

Abstract

:
Acute myeloid leukemia (AML) is a neoplastic disorder resulting from clonal proliferation of poorly differentiated immature myeloid cells. Distinct genetic and epigenetic aberrations are key features of AML that account for its variable response to standard therapy. Irrespective of their oncogenic mutations, AML cells produce elevated levels of reactive oxygen species (ROS). They also alter expression and activity of antioxidant enzymes to promote cell proliferation and survival. Subsequently, selective targeting of redox homeostasis in a molecularly heterogeneous disease, such as AML, has been an appealing approach in the development of novel anti-leukemic chemotherapeutics. Naphthoquinones are able to undergo redox cycling and generate ROS in cancer cells, which have made them excellent candidates for testing against AML cells. In addition to inducing oxidative imbalance in AML cells, depending on their structure, naphthoquinones negatively affect other cellular apparatus causing neoplastic cell death. Here we provide an overview of the anti-AML activities of naphthoquinone derivatives, as well as analysis of their mechanism of action, including induction of reduction-oxidation imbalance, alteration in mitochondrial transmembrane potential, Bcl-2 modulation, initiation of DNA damage, and modulation of MAPK and STAT3 activity, alterations in the unfolded protein response and translocation of FOX-related transcription factors to the nucleus.

1. Introduction

1.1. Chemistry of Naphthoquinones

Naphthoquinones (C10H6O2) are oxidized naphthalenes (C10H8), and similar to other quinones, such as benzoquinone and anthraquinone, as they possess a conjugated electron system that can participate in chemical reactions transporting electrons to other molecules [1]. These reactions can result in the generation of free radicals, including highly active oxygen radicals called reactive oxygen species (ROS) [1]. Production of ROS occurs when quinones are reduced to semiquinones and subsequently to hydroquinones by one- or two-electron reductions via different enzymes [2,3,4]. While ROS, at low and highly regulated intracellular levels, contribute to normal cellular signaling function; excess ROS plays an important role in damaging cellular components, including proteins, lipids and nucleic acids [5]. Agents with similar structures to naphthoquinones have been approved for use by the FDA. For example, Mitomycin, a benzoquinone, is used in the clinic for intravesical therapy after transurethral resection of bladder tumor for non-invasive (stage 0) or minimally invasive (stage I) bladder cancers, and in combination with 5-fluorouracil (5-FU) and radiation for treatment of stage I-III anal cancer [6,7]. Anthraquinones, such as daunorubicin, idarubicin, doxorubicin, epirubicin and mitoxantrone are commonly used for the treatment of many hematologic and solid neoplasms [8,9].
Natural and synthetic, oligomeric substituted and heterocyclic naphthoquinone derivatives have shown promising anti-neoplastic activities against epithelial, mesenchymal and hematopoietic malignancies [2,10,11,12]. Notwithstanding fifty years of basic, translational, and clinical studies, the mechanism by which naphthoquinone-containing agents induce cancer cell death remains somewhat unclear. The apparent redox-cycling and electrophilic properties of naphthoquinones culminating in ROS generation and induction of apoptosis have subjugated the field of mechanistic investigations of these compounds [13,14]. Nevertheless, several other mechanisms, including covalent alkylation of DNA, DNA intercalation, inhibition of topoisomerase II, epigenetic modulation, to name a few, might be significantly contributing to the cause of death in different cancer cells.

1.2. Acute Myeloid Leukemia

Acute myeloid leukemia (AML) is a neoplastic disorder characterized by rapidly proliferating myeloblasts in bone marrow, blood and occasionally other organs resulting in a very poor clinical outcome, particularly in elderly and medically unfit patients. The diverse cytogenetics and molecular mutations of myeloblasts among patients and even within one patient makes AML an extremely heterogeneous blood cancer [15,16]. Irrespective of their genetic characteristics, AML cells generate excess ROS [17]. In turn, these cells have prominent antioxidant machinery to mitigate the effects of additional ROS and maintain cellular oxidative states compatible with cell survival [17,18]. With this in mind, it was hypothesized that naphthoquinone-induced ROS augmentation in myeloblasts could cause an imbalance to the already stressed redox balance, overwhelming the ROS buffering capacity of the AML cells and cause cell death [13]. However, depending on the chemical structures of the naphthoquinone, certain derivatives might negatively affect other cellular machinery involved in the initiation and propagation of neoplastic phenomenon seen in AML. In this article, we aimed to analyze the entire publically available mechanism of actions of naphthoquinone-based anti-AML chemotherapeutics. We believe this will help chemists to synthesize new generations of anti-cancer naphthoquinones and also assist biologists and clinicians to design naphthoquinone-based combination chemotherapies rationally.

2. Results

2.1. Anti-Proliferative Activity

Table 1 shows the naphthoquinone derivatives that have been reported to have anti-AML activity. Mono- or di-substituted monomeric naphthoquinones, including menadione [19], juglone [20], lawson [21], glycinyl-1,4-naphthoquinone [22], plumbagin [23,24,25,26,27], lapachol (and nor-lapachol) [21,28,29], atovaquone [30,31], ramentaceone [32], cordiaquinone J [33], and TW-92 [34] showed anti-AML activity in different AML cell lines and primary cells from AML patients with wide IC50s ranging from 0.6 to 100 micromolar (μM), Table 1. Atovaquone was found to have an additive effect when combined with standard induction chemotherapy (cytarabine and daunorubicin) in AML cell lines [31]. Multi-substituted naphthoquinones, including shikonin [35,36,37,38,39] and its derivative SH-7 [40], and other dihydroxy or dimethoxy 1,4-naphthoquinones [41,42], compared with mono- or di-substituted naphthoquinones, demonstrated superior in vitro activity against AML cells with IC50s ranging 0.1–4 μM. Heterocyclic monomeric naphthoquinones included furanonaphthoquinones FNQ3 [43] and FN6-one [44], β-lapachone [45,46,47,48,49] and nor-β-lapachone [45,46,50,51], dunnione [47], and pterocarpanquinone LQB-118 [28,52]. FNQ3 was significantly more effective than low dose cytarabine in reducing cell viability (p < 0.001) and combining the two drugs led to an even greater reduction in cell viability in NB4 and U937 cells (p < 0.01) [43].
By inhibiting HIV integrase, the hydroxylated dimeric naphthoquinones were originally synthesized as anti-HIV agents [53,54]. Redox modulating ability of these compounds resulted in studies to determine their potency against AML cells, as well as their therapeutic indices in relation to normal hematopoietic stem cells [4,55]. The IC50 values (μM) of 3-bromo-3′-hydroxyl-dimeric 1,4-naphthoquinone (BiQ1) against AML cell lines MOLM-14, THP1, and one primary AML cells from patients, as well as normal bone marrow cells were 5.5 ± 0.8, 4.2 ± 1.9, 0.4 and 14.5, respectively [55]. Bis-aziridinyl dimeric naphthoquinone containing two nitrogen mustard alkylating groups was synthesized to improve the potency and bioavailability of dimeric naphthoquinones [56]. This compound showed a potent anti-leukemic activity (IC50 range 0.18–2 μM) against three AML cell lines and four primary AML cells from patients [56]. Of note, the patient-derived AML cells had a heterogeneous cytogenetic and molecular mutation profile. In addition to inducing a decrease in cell survival and viability, exposure to the bis-aziridinyl dimeric naphthoquinone at concentrations relative to the respective IC50 values for 24 h resulted in a marked reduction in clonogenic activity—an in vitro assay to assess the capability of a single AML cell in the population to generate a colony—the quintessential anti-cancer in vitro assay [56]. Dimeric naphthoquinones had favorable therapeutic indices as demonstrated by their more potent IC50 values in AML cells compared to normal hematopoietic cells.

2.2. In Vivo Tolerability and Efficacy Studies

A few studies tested the tolerability and safety of naphthoquinone derivatives in vivo as wells as their anti-AML activity [4,24,35,40,56]. Plumbagin dosed 2 mg/kg via intraperitoneal injection, IP) daily for three weeks significantly reduced tumor volume in NB4 tumor xenograft in NOD/SCID mice; 153 mm3 in plumbagin versus 194 mm3 in control after one week of treatment and approximately 65% tumor volume reduction at the completion of the study [24]. In the same study using the same xenograft model, tumors of mice treated with doxorubicin (1 mg/kg thrice weekly) demonstrated greater regression in tumor volume than those treated with plumbagin (p < 0.05). However, doxorubicin at the doses tested was toxic to heart and liver (pathologic findings), whereas plumbagin treated mice did not show any obvious tissue changes.
Our group tested tolerability of a different naphthoquinone containing a compound called BiQ1 in mice dosed via IP and subcutaneous (SC) injections [4]. Mice tolerated eight days of IP injections dosed at 10 mg/kg with no overt weight loss or physical complications. However, mice were unable to ambulate, eat or drink with just two doses of 25 mg/kg IP injections. Interestingly, mice were able to tolerate SC doses of 25 mg/kg with the only overt side effect being irritation at the injection site. The poorer tolerability of IP versus SC at higher dose might be related to the acidity of BiQ1 injected in intra-abdominal cavity of mice causing peritonitis.
Carter-Cooper et al. reported that 3,3′-bis-aziridinyl dimeric naphthoquinone was well tolerated by NOD SCID gamma (NSG) mice after five consecutive days of 5, 10, and 15 mg/kg (IP) without significant weight loss [56]. Finally, Nestal de Moraes et al. reported that naphthoquinone derivatives may be safe against normal bone marrow-derived cells by testing compound LQB-118 IP in mice with normal bone marrow cells [52].
Atovaquone is an FDA-approved, anti-microbial drug that has been well tolerated, even in immunosuppressed patients with organ dysfunction. In a retrospective study of adult AML patients who underwent allogeneic hematopoietic stem cell transplant (HSCT), patients who received atovaquone for longer duration had lower rates of AML relapse over three years compared to those with lower atovaquone exposure (13% versus 23%, p = 0.037) [30]. In vivo, female mice treated with atovaquone 200 mg/kg daily demonstrated decreased disease burden compared to the control mice [31]. Furthermore, prolonged atovaquone survival of male xenografted mice compared to the control mice (median survival of 63 days in control mice versus only one mouse succumbing to disease by day 70 in the atovaquone group, p = 0.0048).

2.3. Mechanisms of Anti-AML Action

2.3.1. Induction of Apoptosis

The primary mechanism by which naphthoquinones reduce AML cell survival is via apoptosis. A dose- and time-dependent increase in apoptosis was observed with several naphthoquinone-based compounds. Induction of apoptosis rather than necrosis as the cause of cell death was evident by changes observed in cell morphology, cell cycle progression, and caspase activation. Interestingly, 5,8-dihydroxy-2-(1-hydroxy-2-nitroethyl)naphthalene-1,4-dione [41] and LQB-118 [52] induced apoptosis without significant changes observed in cell cycle. The anti-AML activity of these naphthoquinones appeared to be mediated through a combination of ROS enhancement and topoisomerase inhibition that ultimately led to DNA damage and cell death.

2.3.2. Alteration in Mitochondrial Membrane Potential

Naphthoquinone derivatives reduce mitochondrial membrane potential (MMP) by the opening of the mitochondrial permeability transition pore resulting in cytochrome C release into the cytosol and activating caspase-3; key features of the intrinsic pathway of apoptosis [57,58]. Hallak et al. reported that TW-74, a chloronaphthoquinone with a methyl group at the meta position, effectively killed U937 myeloid leukemia cells via early reduction of MMP, cytochrome C release and caspase activation [59]. We reported that hydroxylated dimeric naphthoquinone BiQ1 effectively inhibited induced depolarization of the mitochondrial transmembrane potential (ΔΨm) as measured by flow cytometry with MitoPotential Red [4].

2.3.3. Reduction-Oxidation Imbalance

ROS are derived from the metabolism of oxygen, and they normally exist in balance with antioxidants in all cells. Oxidative stress occurs when this balance is disrupted, due to excess production of ROS and/or antioxidant diminution. Generation of ROS and the consequent increase in oxidative stress has proven to be fundamental in the cytotoxic activity of naphthoquinone derived compounds. Given the high proliferative index, most AML cells, compared with non-malignant cells, have a higher level of endogenous ROS, lower activities in respiratory chain complex and lower spare reserve capacity [60,61]. This phenomenon, as well as lower compensatory reserves that handle additional oxygen radicals [13,60], make AML cells particularly sensitive to oxidative stress [62]. Several studies have demonstrated that the presence of antioxidants significantly attenuates mitochondrial-dependent apoptosis and cell cycle arrest in naphthoquinone-treated tumor cells [24,34,63]. While antioxidants, such as N-acetyl cysteine (NAC) may serve to lessen naphthoquinone cytotoxicity by neutralizing ROS through increased glutathione production, they also could exert their actions by chemically interacting directly with naphthoquinones or their derivatives to reduce their cytotoxicity [1,64,65].
Much evidence exists in the literature linking redox imbalance and anti-AML activity. Treatment of leukemic cell lines with bis-aziridinyl dimeric naphthoquinone resulted in a dose-dependent increase in intracellular production of ROS within two hours of exposure [56]. Plumbagin similarly demonstrated increased intracellular ROS in NB4 cells that peaked after 1.5 h of exposure with a decline after two hours [66]. Xu and colleagues further demonstrated that ROS production was significantly increased in NB4 cells treated with plumbagin compared to healthy control cells, indicative of selective toxicity against neoplastic cells [24]. The chloro-amino-phenyl naphthoquinone TW-92 induced apoptosis in U937 cells, which was preceded by accumulation of intracellular hydrogen peroxide that was abolished by an inhibitor of NADPH oxidase [34]. U937 cells treated with TW-92 versus untreated cells showed a rapid decline in endogenous glutathione levels (65%) [34]. This depletion of glutathione may have contributed to the intracellular accumulation of hydrogen peroxide. Chau et al. compared intracellular H2O2 concentration in HL-60 cells treated with β-lapachone (a naphthoquinone) and camptothecin (a non-quinone topoisomerase inhibitor) [67]. The study found a seven-fold increase in H2O2 production in HL-60 cells treated with β-lapachone compared to a less than two-fold increase in cells treated with camptothecin. This increase in H2O2 was markedly reduced in the presence of NAC, which corresponded with reduced β-lapachone-induced apoptosis as evidenced by reduced DNA fragmentation and sub-G1 hypodiploid cells. These results suggest that intracellular increase in H2O2 may be one mechanism by which naphthoquinones induce cell death.

2.3.4. DNA Damage

Quinones are able to induce DNA damage, as shown by the increase in histone H2Ax phosphorylation, a step which always follows double-stranded DNA breaks to activate apoptosis [68]. Quinones have also been shown to exert antitumor effects by inhibiting nuclear enzyme topoisomerases I and II [69]. Topoisomerase mediated DNA damage of leukemic cells was observed in the activity of a synthetic naphthoquinone derivative of alkannin and shikonin against HL-60 cells [41]. β-Lapachone (3,4-dihydro-2,2-dimethyl-2H-naphtho[1,2-b]pyran-5,6-dione) is also a topoisomerase I inhibitor [63], by the proposed mechanism of locking the enzyme onto the DNA and blocking replication fork movement [48].
Shikonin has been shown to inhibit both topoisomerases I and II in different cancer cells [70,71,72]. To overcome the poor solubility of Shikonin, its derivative SH-7 was synthesized [40]. SH-7 displayed significant cytotoxicity against HL-60 cells with IC50 of 2 μM and showed significant inhibition of topoisomerase II via stabilization of the topoisomerase II-DNA cleavable complex [40]. Bey et al. suggested that the anti-tumor activity of topoisomerases inhibitors may be attributed to the overexpression of the enzymes and associated deficiency of protective mechanism against DNA damage in cancer cells or to the increase in topoisomerase bioactivation by the NAD(P)H:quinone oxidoreductase (NQO1) [73].

2.3.5. Bcl-2 Modulation

Bcl-2 family of proteins are integral to the mitochondria-dependent pathway of apoptosis [74]. Cell death is regulated by a complex interaction between the pro-apoptotic and pro-survival members of the Bcl-2 family. Pro-apoptotic members of the Bcl-2 family, such as Bax or Bak translocate from the cytosol to the mitochondria, leading to the release of cytochrome C into the cytosol [75]. Anti-apoptotic proteins, Bcl-2 [76] and Mcl-1 [77] have been studied extensively, given their ability to induce chemo-resistance in different cancers, including AML. Venetoclax, a Bcl-2 inhibitor, in combination with azacitidine or decitabine or low-dose cytarabine is approved (conditionally) by the US Food and Drug Administration (FDA) for treatment of adult patients with newly-diagnosed AML who are age 75 years or older, or who have comorbidities that preclude use of intensive induction chemotherapy [78].
The antitumor activities of naphthoquinones may be mediated by their ability to up- or down-regulate the proteins in the Bcl-2 family [4,79,80]. Shikonin down-regulated antiapoptotic proteins, such as Bcl-2 and Bcl-XL in AML cells and induced apoptosis [79]. Plumbagin-treated NB4 cells had increased expression of Bax and Bak with a decrease in Bcl-XL within 8 h of treatment [24]. TW-92 decreased expression of Mcl-1, increased Bax expression without change in expression of Bcl-2 and induced apoptosis in U937 cells [34]. Chau and colleagues reported ectopic overexpression of Bcl-2 in HL-60 cells acquired resistant phenotype to β-lapachone with decreased intracellular H2O2 [67].

2.3.6. MAPK Pathway

Mitogen-activated protein kinase (MAPK) pathways are involved in the regulation of cellular stress response by transducing extracellular signals to the cell nucleus to impact transcription factors important for cell proliferation, differentiation, survival, and apoptosis [81]. Constitutive activation of the MAPK cascades drives the oncogenic transformation of normal fibroblasts and is commonly detected in cancers. Inappropriate activation of these pathways may play a role in leukemic transformation of myeloid cells and their ability to proliferate and escape programmed cell death [82]. Several naphthoquinones have shown the ability to impact the MAPK pathways to induce apoptosis in leukemic cells [34,37], which may be a promising therapeutic strategy for AML. In the shikonin and shikonin derivative study [37], these agent’s influence on MAPK and AKT signaling cascades led to apoptosis of AML cells in vitro by direct interaction, and down-regulation of c-MYC, a transcription factor integral to cell cycle regulation and proliferation [37]. More investigations are warranted on the role of MAPK cascades on naphthoquinone-induced apoptosis as it has been established that MAPK cascades converge to the mitochondria and promote mitochondrial-induced apoptosis by complex interplay with Bcl-2 family [83].

2.3.7. STAT3 Inhibition

Signal transducer and activator of transcription 3 (STAT3) is an oncogenic transcription factor that is often dysregulated in AML and can be used as a valid target for AML treatment [84]. Atovaquone is an anti-microbial drug that has recently been discovered to have antileukemic efficacy in vitro and in vivo [31]. Xiang and colleagues found that atovaquone diminishes the expression of gp130, a key protein involved in STAT3 activation which in turn potently inhibited STAT3 signaling from interleukin-6 (IL-6), a critical survival factor in AML [30]. The reduction of STAT3 activation consequently led to the reduction of STAT3 target genes mediating cell survival and proliferation culminating in apoptosis.

2.3.8. Unfolded Protein Response Modulation

Unfolded protein responses (UPR) is a mechanism by which cells are able to combat adverse effects of protein accumulation in the endoplasmic reticulum (ER) following ER stress by inducing activation of this pro-apoptotic pathway [85]. UPR plays an important role in chemo-resistance in cancer cells, including leukemia cells [86,87]. ERP57, a key protein situated on the ER, is involved in the proper folding of newly synthesized polypeptides, and is overexpressed in AML cells compared to healthy cells [36]. Overexpression of ERP57 significantly reduced Shikonin-induced apoptosis, though its expression is downregulated by Shikonin itself [36].

2.3.9. Forkhead Box (Fox) Proteins

Fox proteins are a family of transcription factors that regulate the expression of genes involved in cell proliferation, growth, and differentiation [88]. They recognize DNA breaks and initiate repair or excision by binding to the Forkhead response element region (FHRE). Members of Fox class M and O have been identified as essential components of oncogenic and tumor suppressive pathways and are often deregulated in leukemia.
FOXO3A is a tumor suppressor transcription factor that is a member of the FoxO subfamily [89]. High levels of phosphorylated FOXO3A was identified as an independent, poor prognostic factor in AML and was associated with increased leukemic cell proliferation, drug resistance, and shorter duration of remission [90]. Phosphorylation leads to cytoplasmic localization and inactivation of FOXO3A, which leads to the proliferation of leukemic cells in AML. Treatment of HL-60 cells with naphthoquinone LQB-118 resulted in FOXO3A nuclear translocation with upregulation of Bim, a pro-apoptotic member of the Bcl-2 family [52]. In contrast, U937 cells treated with LQB-118 had FOXO3A nuclear exclusion and Bim downregulation [52] suggesting that LQB-118 promotes differential modulation of FOXO3A localization in different AML subtypes.
Fox protein M1 (FOXM1) is a regulator of cell cycle progression and was found to be abnormally expressed in AML blasts [91]. LQB-118-treated U937 cells had progressively decreasing FOXM1 expression, with concurrent downregulation of Survivin [52]. Of note, Survivin is an anti-apoptotic protein that is correlated with poor prognosis in AML and is known to be regulated by FOXM1 [92].

3. Methods and Materials

3.1. Search Strategy and Inclusion Criteria

An electronic search was conducted on the MEDLINE database for articles published up to 7 August 2018, using the combined search terms acute, leukemia, and naphthoquinone. Seventy-eight articles were preliminarily identified. Studies that investigated the activity of naphthoquinone derivatives against AML were selectively reviewed. Twenty-one publications reported that anti-AML efficacy, in addition to at least one proposed mechanism of action of the studied naphthoquinone(s) were included for this analysis. The reference lists of these selected articles were also examined to include any relevant publications not captured in the initial search. In this report, we will not compare or comment on the study designs of the included articles. Additionally, in studies that synthesized and analyzed multiple naphthoquinone derivatives with the same core structure, only the compound with the strongest activity against AML cells, as determined by their IC50 (i.e., concentration that decreases viable cell numbers by 50%) values, were included for discussion.

3.2. Cell Cultures

In the included studies, in vitro activities of the naphthoquinone derivatives were examined in AML cell lines, and myeloblasts isolated from peripheral blood (PB) and bone marrow (BM) of AML patients. Often, the cytotoxicity of the compounds was studied in normal human cell lines and primary cells from healthy donors as comparators. Just a few in vivo studies have been conducted to examine the safety and tolerability of naphthoquinone-based agents.

3.3. Cell Proliferation and Cell Survival Assays

Inhibition of cell growth was assessed by measuring cell metabolic activity by MTT-like assays, including 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) (XTT), and water-soluble tetrazolium salts (WST) assays. Cell viability was determined using trypan blue exclusion.

3.4. Analysis of Apoptosis

Apoptosis was measured using the Annexin V flow cytometric kits, or acridine orange/ethidium bromide stains. Means of cell death were further evaluated with cell cycle progression (propidium iodide), mitochondrial membrane potential, Western Blot analyses, nuclei staining, or caspase activation.

4. Conclusions

The ability of naphthoquinone analogs to affect multiple steps in the apoptotic pathway and cellular signaling pathways play a role in their anti-AML activity. The simple mono- and di-substituted naphthoquinones have the ability to inhibit cell growth of various AML cell lines at low micromolar concentrations with the most potent derivatives being the 5-hydroxy-substituted derivative plumbagin (0.6–1.4 μM). Consequently, plumbagin also increases intracellular ROS and decreases expression of Bcl-2, indicating that the increased potency may be due to multiple mechanisms of action. From the multi-substituted naphthoquinones, this same pattern is observed with the shikonin derivatives demonstrating low micromolar (shikonin and SH-7) and in one case, (5,8-dihydroxy-2-(1-hydroxy-2-nitroethyl)-1,4-naphthoquinone, 0.14 μM) sub-micromolar potency against AML cell lines. Most likely multiple mechanisms of action for these derivatives provides the robust activity, including inhibition of topoisomerase, down-regulation of Bcl-2/BclxL, and down regulation of c-Myc and maybe the unfolded protein response. Thus, depending on the oxidoreductases or other bases for altered electron donation, present in AML cells and a few other neoplastic cells, the electron-accepting potential of quinones could in principle be “tuned” to yield selective cytotoxicity for cells with a particular “redox” environment, allowing initiation of a cascade of electron transport only in malignant cells, with dysregulated redox state, and not in normal cells, with the consequent increase in ROS producing selective cell killing. The dimeric naphthoquinones represent some of the most potent compounds against AML cell lines, and they have the ability to increase intracellular ROS like plumbagin.

Author Contributions

Conceptualization, A.E.; methodology, M.H.L., R.G.L.; validation, D.F., A.E.; formal analysis, M.H.L., A.E.; writing—original draft preparation, M.H.L.; writing—review and editing, D.F., R.G.L., A.E.; visualization, D.F.; supervision, R.G.L., A.E.; funding acquisition, A.E.

Funding

The manuscript was partially funded by National Cancer Institute (NCI), grant number: P30CA134274.

Conflicts of Interest

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

References

  1. Emadi, A.; Ross, A.E.; Cowan, K.M.; Fortenberry, Y.M.; Vuica-Ross, M. A chemical genetic screen for modulators of asymmetrical 2,2′-dimeric naphthoquinones cytotoxicity in yeast. PLoS ONE 2010, 5, e10846. [Google Scholar] [CrossRef] [PubMed]
  2. Verma, R.P. Anti-cancer activities of 1, 4-naphthoquinones: A QSAR study. Anti Cancer Agents Med. Chem. 2006, 6, 489–499. [Google Scholar] [CrossRef]
  3. Pidugu, L.S.; Mbimba, J.C.; Ahmad, M.; Pozharski, E.; Sausville, E.A.; Emadi, A.; Toth, E.A. A direct interaction between NQO1 and a chemotherapeutic dimeric naphthoquinone. BMC Struct. Biol. 2016, 16, 1. [Google Scholar] [CrossRef] [PubMed]
  4. Lapidus, R.G.; Carter-Cooper, B.A.; Sadowska, M.; Choi, E.Y.; Wonodi, O.; Muvarak, N.; Natarajan, K.; Pidugu, L.S.; Jaiswal, A.; Toth, E.A.; et al. Hydroxylated Dimeric Naphthoquinones Increase the Generation of Reactive Oxygen Species, Induce Apoptosis of Acute Myeloid Leukemia Cells and Are Not Substrates of the Multidrug Resistance Proteins ABCB1 and ABCG2. Pharmaceuticals 2016, 9, 4. [Google Scholar] [CrossRef] [PubMed]
  5. Bardaweel, S.K.; Gul, M.; Alzweiri, M.; Ishaqat, A.; HA, A.L.; Bashatwah, R.M. Reactive Oxygen Species: The Dual Role in Physiological and Pathological Conditions of the Human Body. Eurasian J. Med. 2018, 50, 193–201. [Google Scholar] [CrossRef] [PubMed]
  6. Milla, P.; Fiorito, C.; Soria, F.; Arpicco, S.; Cattel, L.; Gontero, P. Intravesical thermo-chemotherapy based on conductive heat: A first pharmacokinetic study with mitomycin C in superficial transitional cell carcinoma patients. Cancer Chemother. Pharmacol. 2014, 73, 503–509. [Google Scholar] [CrossRef] [PubMed]
  7. Vinayan, A.; Glynne-Jones, R. Anal cancer—What is the optimum chemoradiotherapy? Best Pract. Res. Clin. Gastroenterol. 2016, 30, 641–653. [Google Scholar] [CrossRef]
  8. Hortobagyi, G.N. Anthracyclines in the treatment of cancer. An overview. Drugs 1997, 54, 1–7. [Google Scholar] [CrossRef]
  9. Emadi, A.; Karp, J.E. The clinically relevant pharmacogenomic changes in acute myelogenous leukemia. Pharmacogenomics 2012, 13, 1257–1269. [Google Scholar] [CrossRef] [Green Version]
  10. Driscoll, J.S. Quinone structure-antitumor activity relationships. Cancer Chemother. Rep. Part 2 1974, 4, 3–4. [Google Scholar]
  11. Ross, A.E.; Emadi, A.; Marchionni, L.; Hurley, P.J.; Simons, B.W.; Schaeffer, E.M.; Vuica-Ross, M. Dimeric naphthoquinones, a novel class of compounds with prostate cancer cytotoxicity. BJU Int. 2011, 108, 447–454. [Google Scholar] [CrossRef] [PubMed]
  12. Emadi, A.; Le, A.; Harwood, C.J.; Stagliano, K.W.; Kamangar, F.; Ross, A.E.; Cooper, C.R.; Dang, C.V.; Karp, J.E.; Vuica-Ross, M. Metabolic and electrochemical mechanisms of dimeric naphthoquinones cytotoxicity in breast cancer cells. Bioorg. Med. Chem. 2011, 19, 7057–7062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Irwin, M.E.; Rivera-Del Valle, N.; Chandra, J. Redox control of leukemia: From molecular mechanisms to therapeutic opportunities. Antioxid. Redox Signal. 2013, 18, 1349–1383. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, H.; Fang, H.; Wang, K. Reactive oxygen species in eradicating acute myeloid leukemic stem cells. Stem Cell Investig. 2014, 1, 1–10. [Google Scholar]
  15. Dohner, H.; Estey, E.; Grimwade, D.; Amadori, S.; Appelbaum, F.R.; Buchner, T.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Larson, R.A.; et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017, 129, 424–447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Shih, A.H.; Jiang, Y.; Meydan, C.; Shank, K.; Pandey, S.; Barreyro, L.; Antony-Debre, I.; Viale, A.; Socci, N.; Sun, Y.; et al. Mutational cooperativity linked to combinatorial epigenetic gain of function in acute myeloid leukemia. Cancer Cell 2015, 27, 502–515. [Google Scholar] [CrossRef] [PubMed]
  17. Guzman, M.L.; Rossi, R.M.; Karnischky, L.; Li, X.; Peterson, D.R.; Howard, D.S.; Jordan, C.T. The sesquiterpene lactone parthenolide induces apoptosis of human acute myelogenous leukemia stem and progenitor cells. Blood 2005, 105, 4163–4169. [Google Scholar] [CrossRef]
  18. Emadi, A.; Sadowska, M.; Carter-Cooper, B.; Bhatnagar, V.; van der Merwe, I.; Levis, M.J.; Sausville, E.A.; Lapidus, R.G. Perturbation of cellular oxidative state induced by dichloroacetate and arsenic trioxide for treatment of acute myeloid leukemia. Leuk. Res. 2015, 39, 719–729. [Google Scholar] [CrossRef]
  19. Yeo, H.S.; Shehzad, A.; Lee, Y.S. Prostaglandin E2 blocks menadione-induced apoptosis through the Ras/Raf/Erk signaling pathway in promonocytic leukemia cell lines. Mol. Cells 2012, 33, 371–378. [Google Scholar] [CrossRef] [Green Version]
  20. Xu, H.L.; Yu, X.F.; Qu, S.C.; Zhang, R.; Qu, X.R.; Chen, Y.P.; Ma, X.Y.; Sui, D.Y. Anti-proliferative effect of Juglone from Juglans mandshurica Maxim on human leukemia cell HL-60 by inducing apoptosis through the mitochondria-dependent pathway. Eur. J. Pharmacol. 2010, 645, 14–22. [Google Scholar] [CrossRef]
  21. Esteves-Souza, A.; Lucio, K.A.; Da Cunha, A.S.; Da Cunha Pinto, A.; Da Silva Lima, E.L.; Camara, C.A.; Vargas, M.D.; Gattass, C.R. Antitumoral activity of new polyamine-naphthoquinone conjugates. Oncol. Rep. 2008, 20, 225–231. [Google Scholar] [CrossRef] [PubMed]
  22. de Moraes, T.A.; Filha, M.J.; Camara, C.A.; Silva, T.M.; Soares, B.M.; Bomfim, I.S.; Pessoa, C.; Ximenes, G.C.; Silva Junior, V.A. Synthesis and cytotoxic evaluation of a series of 2-amino-naphthoquinones against human cancer cells. Molecules 2014, 19, 13188–13199. [Google Scholar] [CrossRef] [PubMed]
  23. Kawiak, A.; Piosik, J.; Stasilojc, G.; Gwizdek-Wisniewska, A.; Marczak, L.; Stobiecki, M.; Bigda, J.; Lojkowska, E. Induction of apoptosis by plumbagin through reactive oxygen species-mediated inhibition of topoisomerase II. Toxicol. Appl. Pharmacol. 2007, 223, 267–276. [Google Scholar] [CrossRef] [PubMed]
  24. Xu, K.H.; Lu, D.P. Plumbagin induces ROS-mediated apoptosis in human promyelocytic leukemia cells in vivo. Leuk. Res. 2010, 34, 658–665. [Google Scholar] [CrossRef] [PubMed]
  25. Gaascht, F.; Teiten, M.H.; Cerella, C.; Dicato, M.; Bagrel, D.; Diederich, M. Plumbagin modulates leukemia cell redox status. Molecules 2014, 19, 10011–10032. [Google Scholar] [CrossRef] [PubMed]
  26. Kong, X.; Luo, J.; Xu, T.; Zhou, Y.; Pan, Z.; Xie, Y.; Zhao, L.; Lu, Y.; Han, X.; Li, Z.; et al. Plumbagin enhances TRAIL-induced apoptosis of human leukemic Kasumi1 cells through upregulation of TRAIL death receptor expression, activation of caspase-8 and inhibition of cFLIP. Oncol. Rep. 2017, 37, 3423–3432. [Google Scholar] [CrossRef]
  27. Zhang, J.; Peng, S.; Li, X.; Liu, R.; Han, X.; Fang, J. Targeting thioredoxin reductase by plumbagin contributes to inducing apoptosis of HL-60 cells. Arch. Biochem. Biophys. 2017, 619, 16–26. [Google Scholar] [CrossRef]
  28. de Souza Reis, F.R.; de Faria, F.C.; Castro, C.P.; de Souza, P.S.; da Cunha Vasconcelos, F.; Bello, R.D.; da Silva, A.J.; Costa, P.R.; Maia, R.C. The therapeutical potential of a novel pterocarpanquinone LQB-118 to target inhibitor of apoptosis proteins in acute myeloid leukemia cells. Anti Cancer Agents Med. Chem. 2013, 13, 341–351. [Google Scholar] [CrossRef]
  29. Araujo, A.J.; de Souza, A.A.; da Silva Junior, E.N.; Marinho-Filho, J.D.; de Moura, M.A.; Rocha, D.D.; Vasconcellos, M.C.; Costa, C.O.; Pessoa, C.; de Moraes, M.O.; et al. Growth inhibitory effects of 3’-nitro-3-phenylamino nor-beta-lapachone against HL-60: A redox-dependent mechanism. Toxicol. In Vitro 2012, 26, 585–594. [Google Scholar] [CrossRef]
  30. Xiang, M.; Kim, H.; Ho, V.T.; Walker, S.R.; Bar-Natan, M.; Anahtar, M.; Liu, S.; Toniolo, P.A.; Kroll, Y.; Jones, N.; et al. Gene expression-based discovery of atovaquone as a STAT3 inhibitor and anticancer agent. Blood 2016, 128, 1845–1853. [Google Scholar] [CrossRef]
  31. Stevens, A.M.; Munoz, J.O.; Krueger, M.; Horton, T.M.; Redell, M. Atovaquone Demonstrates Single Agent Activity and an Additive Effect in Combination with Cytotoxic Chemotherapy (Cytarabine and Daunorubicin) in AML. Blood 2017, 130, 3899. [Google Scholar]
  32. Kawiak, A.; Zawacka-Pankau, J.; Wasilewska, A.; Stasilojc, G.; Bigda, J.; Lojkowska, E. Induction of apoptosis in HL-60 cells through the ROS-mediated mitochondrial pathway by ramentaceone from Drosera aliciae. J. Nat. Prod. 2012, 75, 9–14. [Google Scholar] [CrossRef] [PubMed]
  33. Marinho-Filho, J.D.; Bezerra, D.P.; Araujo, A.J.; Montenegro, R.C.; Pessoa, C.; Diniz, J.C.; Viana, F.A.; Pessoa, O.D.; Silveira, E.R.; de Moraes, M.O.; et al. Oxidative stress induction by (+)-cordiaquinone J triggers both mitochondria-dependent apoptosis and necrosis in leukemia cells. Chem. Biol. Interact. 2010, 183, 369–379. [Google Scholar] [CrossRef] [PubMed]
  34. Hallak, M.; Win, T.; Shpilberg, O.; Bittner, S.; Granot, Y.; Levy, I.; Nathan, I. The anti-leukaemic activity of novel synthetic naphthoquinones against acute myeloid leukaemia: Induction of cell death via the triggering of multiple signalling pathways. Br. J. Haematol. 2009, 147, 459–470. [Google Scholar] [CrossRef] [PubMed]
  35. Yang, H.; Zhou, P.; Huang, H.; Chen, D.; Ma, N.; Cui, Q.C.; Shen, S.; Dong, W.; Zhang, X.; Lian, W.; et al. Shikonin exerts antitumor activity via proteasome inhibition and cell death induction in vitro and in vivo. Int. J. Cancer 2009, 124, 2450–2459. [Google Scholar] [CrossRef] [PubMed]
  36. Trivedi, R.; Muller, G.A.; Rathore, M.S.; Mishra, D.P.; Dihazi, H. Anti-Leukemic Activity of Shikonin: Role of ERP57 in Shikonin Induced Apoptosis in Acute Myeloid Leukemia. Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 2016, 39, 604–616. [Google Scholar] [CrossRef] [PubMed]
  37. Zhao, Q.; Assimopoulou, A.N.; Klauck, S.M.; Damianakos, H.; Chinou, I.; Kretschmer, N.; Rios, J.L.; Papageorgiou, V.P.; Bauer, R.; Efferth, T. Inhibition of c-MYC with involvement of ERK/JNK/MAPK and AKT pathways as a novel mechanism for shikonin and its derivatives in killing leukemia cells. Oncotarget 2015, 6, 38934–38951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Zhao, B.; Yang, Y.; Wang, X.; Chong, Z.; Yin, R.; Song, S.H.; Zhao, C.; Li, C.; Huang, H.; Sun, B.F.; et al. Redox-active quinones induces genome-wide DNA methylation changes by an iron-mediated and Tet-dependent mechanism. Nucleic Acids Res. 2014, 42, 1593–1605. [Google Scholar] [CrossRef]
  39. Zhang, B.; Chen, N.; Chen, H.; Wang, Z.; Zheng, Q. The critical role of redox homeostasis in shikonin-induced HL-60 cell differentiation via unique modulation of the Nrf2/ARE pathway. Oxidative Med. Cell. Longev. 2012, 2012, 781516. [Google Scholar] [CrossRef]
  40. Yang, F.; Chen, Y.; Duan, W.; Zhang, C.; Zhu, H.; Ding, J. SH-7, a new synthesized shikonin derivative, exerting its potent antitumor activities as a topoisomerase inhibitor. Int. J. Cancer 2006, 119, 1184–1193. [Google Scholar] [CrossRef]
  41. Beretta, G.L.; Ribaudo, G.; Menegazzo, I.; Supino, R.; Capranico, G.; Zunino, F.; Zagotto, G. Synthesis and Evaluation of New Naphthalene and Naphthoquinone Derivatives as Anticancer Agents. Arch. Der Pharm. 2017, 350, e1600286. [Google Scholar] [CrossRef] [PubMed]
  42. Li, K.; Wang, B.; Zheng, L.; Yang, K.; Li, Y.; Hu, M.; He, D. Target ROS to induce apoptosis and cell cycle arrest by 5, 7-dimethoxy-1, 4-naphthoquinone derivative. Bioorganic Med. Chem. Lett. 2017, 28, 273–277. [Google Scholar] [CrossRef] [PubMed]
  43. Desmond, J.C.; Kawabata, H.; Mueller-Tidow, C.; Simamura, E.; Heber, D.; Hirai, K.; Koeffler, H.P. The synthetic furanonaphthoquinone induces growth arrest, apoptosis and differentiation in a variety of leukaemias and multiple myeloma cells. Br. J. Haematol. 2005, 131, 520–529. [Google Scholar] [CrossRef] [PubMed]
  44. Inagaki, R.; Ninomiya, M.; Tanaka, K.; Watanabe, K.; Koketsu, M. Synthesis and cytotoxicity on human leukemia cells of furonaphthoquinones isolated from tabebuia plants. Chem. Pharm. Bull. 2013, 61, 670–673. [Google Scholar] [CrossRef] [PubMed]
  45. da Silva Junior, E.N.; de Souza, M.C.; Pinto, A.V.; Pinto Mdo, C.; Goulart, M.O.; Barros, F.W.; Pessoa, C.; Costa-Lotufo, L.V.; Montenegro, R.C.; de Moraes, M.O.; et al. Synthesis and potent antitumor activity of new arylamino derivatives of nor-beta-lapachone and nor-alpha-lapachone. Bioorg. Med. Chem. 2007, 15, 7035–7041. [Google Scholar] [CrossRef] [PubMed]
  46. da Silva, E.N., Jr.; de Deus, C.F.; Cavalcanti, B.C.; Pessoa, C.; Costa-Lotufo, L.V.; Montenegro, R.C.; de Moraes, M.O.; Pinto Mdo, C.; de Simone, C.A.; Ferreira, V.F.; et al. 3-arylamino and 3-alkoxy-nor-beta-lapachone derivatives: Synthesis and cytotoxicity against cancer cell lines. J. Med. Chem. 2010, 53, 504–508. [Google Scholar] [CrossRef] [PubMed]
  47. Inagaki, R.; Ninomiya, M.; Tanaka, K.; Koketsu, M. Synthesis, Characterization, and Antileukemic Properties of Naphthoquinone Derivatives of Lawsone. ChemMedChem 2015, 10, 1413–1423. [Google Scholar] [CrossRef] [PubMed]
  48. Planchon, S.M.; Wuerzberger, S.; Frydman, B.; Witiak, D.T.; Hutson, P.; Church, D.R.; Wilding, G.; Boothman, D.A. Beta-lapachone-mediated apoptosis in human promyelocytic leukemia (HL-60) and human prostate cancer cells: A p53-independent response. Cancer Res. 1995, 55, 3706–3711. [Google Scholar] [PubMed]
  49. Planchon, S.M.; Wuerzberger-Davis, S.M.; Pink, J.J.; Robertson, K.A.; Bornmann, W.G.; Boothman, D.A. Bcl-2 protects against beta-lapachone-mediated caspase 3 activation and apoptosis in human myeloid leukemia (HL-60) cells. Oncol. Rep. 1999, 6, 485–492. [Google Scholar] [CrossRef]
  50. Cavalcanti, B.C.; Barros, F.W.; Cabral, I.O.; Ferreira, J.R.; Magalhaes, H.I.; Junior, H.V.; da Silva Junior, E.N.; de Abreu, F.C.; Costa, C.O.; Goulart, M.O.; et al. Preclinical genotoxicology of nor-beta-lapachone in human cultured lymphocytes and Chinese hamster lung fibroblasts. Chem. Res. Toxicol. 2011, 24, 1560–1574. [Google Scholar] [CrossRef] [PubMed]
  51. Cardoso, M.F.; Rodrigues, P.C.; Oliveira, M.E.; Gama, I.L.; da Silva, I.M.; Santos, I.O.; Rocha, D.R.; Pinho, R.T.; Ferreira, V.F.; de Souza, M.C.; et al. Synthesis and evaluation of the cytotoxic activity of 1, 2-furanonaphthoquinones tethered to 1, 2, 3-1H-triazoles in myeloid and lymphoid leukemia cell lines. Eur. J. Med. Chem. 2014, 84, 708–717. [Google Scholar] [CrossRef] [PubMed]
  52. Nestal de Moraes, G.; Castro, C.P.; Salustiano, E.J.; Dumas, M.L.; Costas, F.; Lam, E.W.; Costa, P.R.; Maia, R.C. The pterocarpanquinone LQB-118 induces apoptosis in acute myeloid leukemia cells of distinct molecular subtypes and targets FoxO3a and FoxM1 transcription factors. Int. J. Oncol. 2014, 45, 1949–1958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Emadi, A.; Harwood, J.S.; Kohanim, S.; Stagliano, K.W. Regiocontrolled synthesis of the trimeric quinone framework of conocurvone. Org. Lett. 2002, 4, 521–524. [Google Scholar] [CrossRef] [PubMed]
  54. Stagliano, K.W.; Emadi, A.; Lu, Z.; Malinakova, H.C.; Twenter, B.; Yu, M.; Holland, L.E.; Rom, A.M.; Harwood, J.S.; Amin, R.; et al. Regiocontrolled synthesis and HIV inhibitory activity of unsymmetrical binaphthoquinone and trimeric naphthoquinone derivatives of conocurvone. Bioorg. Med. Chem. 2006, 14, 5651–5665. [Google Scholar] [CrossRef] [PubMed]
  55. Emadi, A.; Sadowska, M.; Carter-Cooper, B.; Wonodi, O.; Muvarak, N.; Rassool, F.; Jaiswal, A.; Baer, M.R.; Lapidus, R.G.; Sausville, E.A. Dimeric Naphthoquinones: Novel Anti-Leukemic Agents Modulating Cellular Redox Status. Blood 2013, 122, 1290. [Google Scholar]
  56. Carter-Cooper, B.A.; Fletcher, S.; Ferraris, D.; Choi, E.Y.; Kronfli, D.; Dash, S.; Truong, P.; Sausville, E.A.; Lapidus, R.G.; Emadi, A. Synthesis, characterization and antineoplastic activity of bis-aziridinyl dimeric naphthoquinone—A novel class of compounds with potent activity against acute myeloid leukemia cells. Bioorg. Med. Chem. Lett. 2017, 27, 6–10. [Google Scholar] [CrossRef] [PubMed]
  57. Green, D.R.; Reed, J.C. Mitochondria and apoptosis. Science 1998, 281, 1309–1312. [Google Scholar] [CrossRef] [PubMed]
  58. Gerasimenko, J.V.; Gerasimenko, O.V.; Palejwala, A.; Tepikin, A.V.; Petersen, O.H.; Watson, A.J. Menadione-induced apoptosis: Roles of cytosolic Ca(2+) elevations and the mitochondrial permeability transition pore. J. Cell Sci. 2002, 115, 485–497. [Google Scholar] [PubMed]
  59. Hallak, M.; Thakur, B.K.; Winn, T.; Shpilberg, O.; Bittner, S.; Granot, Y.; Levy, I.; Nathan, I. Induction of death of leukemia cells by TW-74, a novel derivative of chloro-naphthoquinone. Anticancer Res. 2013, 33, 183–190. [Google Scholar]
  60. Sriskanthadevan, S.; Jeyaraju, D.V.; Chung, T.E.; Prabha, S.; Xu, W.; Skrtic, M.; Jhas, B.; Hurren, R.; Gronda, M.; Wang, X.; et al. AML cells have low spare reserve capacity in their respiratory chain that renders them susceptible to oxidative metabolic stress. Blood 2015, 125, 2120–2130. [Google Scholar] [CrossRef] [Green Version]
  61. Gourdin, T.S.; Zou, Y.; Ning, Y.; Emadi, A.; Duong, V.H.; Tidwell, M.L.; Chen, C.; Rassool, F.V.; Baer, M.R. High frequency of rare structural chromosome abnormalities at relapse of cytogenetically normal acute myeloid leukemia with FLT3 internal tandem duplication. Cancer Genet. 2014, 207, 467–473. [Google Scholar] [CrossRef] [PubMed]
  62. Mantymaa, P.; Guttorm, T.; Siitonen, T.; Saily, M.; Savolainen, E.R.; Levonen, A.L.; Kinnula, V.; Koistinen, P. Cellular redox state and its relationship to the inhibition of clonal cell growth and the induction of apoptosis during all-trans retinoic acid exposure in acute myeloblastic leukemia cells. Haematologica 2000, 85, 238–245. [Google Scholar] [PubMed]
  63. Li, J.Z.; Ke, Y.; Misra, H.P.; Trush, M.A.; Li, Y.R.; Zhu, H.; Jia, Z. Mechanistic studies of cancer cell mitochondria- and NQO1-mediated redox activation of beta-lapachone, a potentially novel anticancer agent. Toxicol. Appl. Pharmacol. 2014, 281, 285–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Bittner, S. When quinones meet amino acids: Chemical, physical and biological consequences. Amino Acids 2006, 30, 205–224. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, X.; Thomas, B.; Sachdeva, R.; Arterburn, L.; Frye, L.; Hatcher, P.G.; Cornwell, D.G.; Ma, J. Mechanism of arylating quinone toxicity involving Michael adduct formation and induction of endoplasmic reticulum stress. Proc. Natl. Acad. Sci. USA 2006, 103, 3604–3609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Zhao, Y.L.; Lu, D.P. Effects of plumbagin on the human acute promyelocytic leukemia cells in vitro. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2006, 14, 208–211. [Google Scholar]
  67. Chau, Y.P.; Shiah, S.G.; Don, M.J.; Kuo, M.L. Involvement of hydrogen peroxide in topoisomerase inhibitor beta-lapachone-induced apoptosis and differentiation in human leukemia cells. Free Radic. Biol. Med. 1998, 24, 660–670. [Google Scholar] [CrossRef]
  68. Kuo, L.J.; Yang, L.X. Gamma-H2AX—A novel biomarker for DNA double-strand breaks. In Vivo 2008, 22, 305–309. [Google Scholar]
  69. Boothman, D.A.; Trask, D.K.; Pardee, A.B. Inhibition of potentially lethal DNA damage repair in human tumor cells by beta-lapachone, an activator of topoisomerase I. Cancer Res. 1989, 49, 605–612. [Google Scholar]
  70. Ahn, B.Z.; Baik, K.U.; Kweon, G.R.; Lim, K.; Hwang, B.D. Acylshikonin analogues: Synthesis and inhibition of DNA topoisomerase-I. J. Med. Chem. 1995, 38, 1044–1047. [Google Scholar] [CrossRef]
  71. Fujii, N.; Yamashita, Y.; Arima, Y.; Nagashima, M.; Nakano, H. Induction of topoisomerase II-mediated DNA cleavage by the plant naphthoquinones plumbagin and shikonin. Antimicrob. Agents Chemother. 1992, 36, 2589–2594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Plyta, Z.F.; Li, T.; Papageorgiou, V.P.; Mellidis, A.S.; Assimopoulou, A.N.; Pitsinos, E.N.; Couladouros, E.A. Inhibition of topoisomerase I by naphthoquinone derivatives. Bioorg. Med. Chem. Lett. 1998, 8, 3385–3390. [Google Scholar] [CrossRef]
  73. Bey, E.A.; Bentle, M.S.; Reinicke, K.E.; Dong, Y.; Yang, C.R.; Girard, L.; Minna, J.D.; Bornmann, W.G.; Gao, J.; Boothman, D.A. An NQO1- and PARP-1-mediated cell death pathway induced in non-small-cell lung cancer cells by beta-lapachone. Proc. Natl. Acad. Sci. USA 2007, 104, 11832–11837. [Google Scholar] [CrossRef] [PubMed]
  74. Gross, A.; McDonnell, J.M.; Korsmeyer, S.J. BCL-2 family members and the mitochondria in apoptosis. Genes Dev. 1999, 13, 1899–1911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Liu, X.; Kim, C.N.; Yang, J.; Jemmerson, R.; Wang, X. Induction of apoptotic program in cell-free extracts: Requirement for dATP and cytochrome c. Cell 1996, 86, 147–157. [Google Scholar] [CrossRef]
  76. O’Gorman, D.M.; Cotter, T.G. Molecular signals in anti-apoptotic survival pathways. Leukemia 2001, 15, 21–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Kitada, S.; Zapata, J.M.; Andreeff, M.; Reed, J.C. Protein kinase inhibitors flavopiridol and 7-hydroxy-staurosporine down-regulate antiapoptosis proteins in B-cell chronic lymphocytic leukemia. Blood 2000, 96, 393–397. [Google Scholar]
  78. Food and Drug Administration (FDA). Venetoclax (VENCLEXTA); FDA: Baltimore, MD, USA, 2018.
  79. Hsu, P.C.; Huang, Y.T.; Tsai, M.L.; Wang, Y.J.; Lin, J.K.; Pan, M.H. Induction of apoptosis by shikonin through coordinative modulation of the Bcl-2 family, p27, and p53, release of cytochrome c, and sequential activation of caspases in human colorectal carcinoma cells. J. Agric. Food Chem. 2004, 52, 6330–6337. [Google Scholar] [CrossRef]
  80. Wang, X.; Wang, Y. Ginsenoside Rh2 Mitigates Pediatric Leukemia Through Suppression of Bcl-2 in Leukemia Cells. Cell. Physiol. Biochem. 2015, 37, 641–650. [Google Scholar] [CrossRef]
  81. Zhang, W.; Liu, H.T. MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Res. 2002, 12, 9–18. [Google Scholar] [CrossRef]
  82. Dash, A.; Gilliland, D.G. Molecular genetics of acute myeloid leukaemia. Best Pract. Res. Clin. Haematol. 2001, 14, 49–64. [Google Scholar] [CrossRef] [PubMed]
  83. Miyoshi, N.; Uchida, K.; Osawa, T.; Nakamura, Y. A link between benzyl isothiocyanate-induced cell cycle arrest and apoptosis: Involvement of mitogen-activated protein kinases in the Bcl-2 phosphorylation. Cancer Res. 2004, 64, 2134–2142. [Google Scholar] [CrossRef] [PubMed]
  84. Bruserud, O.; Nepstad, I.; Hauge, M.; Hatfield, K.J.; Reikvam, H. STAT3 as a possible therapeutic target in human malignancies: Lessons from acute myeloid leukemia. Expert Rev. Hematol. 2015, 8, 29–41. [Google Scholar] [CrossRef] [PubMed]
  85. Sovolyova, N.; Healy, S.; Samali, A.; Logue, S.E. Stressed to death—Mechanisms of ER stress-induced cell death. Biol. Chem. 2014, 395, 1–13. [Google Scholar] [CrossRef]
  86. Epple, L.M.; Dodd, R.D.; Merz, A.L.; Dechkovskaia, A.M.; Herring, M.; Winston, B.A.; Lencioni, A.M.; Russell, R.L.; Madsen, H.; Nega, M.; et al. Induction of the unfolded protein response drives enhanced metabolism and chemoresistance in glioma cells. PLoS ONE 2013, 8, e73267. [Google Scholar] [CrossRef]
  87. Pyrko, P.; Schonthal, A.H.; Hofman, F.M.; Chen, T.C.; Lee, A.S. The unfolded protein response regulator GRP78/BiP as a novel target for increasing chemosensitivity in malignant gliomas. Cancer Res. 2007, 67, 9809–9816. [Google Scholar] [CrossRef]
  88. Myatt, S.S.; Lam, E.W. The emerging roles of forkhead box (Fox) proteins in cancer. Nat. Rev. Cancer 2007, 7, 847–859. [Google Scholar] [CrossRef]
  89. Liu, Y.; Ao, X.; Ding, W.; Ponnusamy, M.; Wu, W.; Hao, X.; Yu, W.; Wang, Y.; Li, P.; Wang, J. Critical role of FOXO3a in carcinogenesis. Mol. Cancer 2018, 17, 104. [Google Scholar] [CrossRef]
  90. Kornblau, S.M.; Singh, N.; Qiu, Y.; Chen, W.; Zhang, N.; Coombes, K.R. Highly phosphorylated FOXO3A is an adverse prognostic factor in acute myeloid leukemia. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2010, 16, 1865–1874. [Google Scholar] [CrossRef]
  91. Nakamura, S.; Hirano, I.; Okinaka, K.; Takemura, T.; Yokota, D.; Ono, T.; Shigeno, K.; Shibata, K.; Fujisawa, S.; Ohnishi, K. The FOXM1 transcriptional factor promotes the proliferation of leukemia cells through modulation of cell cycle progression in acute myeloid leukemia. Carcinogenesis 2010, 31, 2012–2021. [Google Scholar] [CrossRef] [Green Version]
  92. Adida, C.; Recher, C.; Raffoux, E.; Daniel, M.T.; Taksin, A.L.; Rousselot, P.; Sigaux, F.; Degos, L.; Altieri, D.C.; Dombret, H. Expression and prognostic significance of survivin in de novo acute myeloid leukaemia. Br. J. Haematol. 2000, 111, 196–203. [Google Scholar] [CrossRef] [PubMed]
Sample Availability: Samples of the compounds are not available from the authors.
Table 1. Anti-acute myeloid leukemia (AML) naphthoquinones.
Table 1. Anti-acute myeloid leukemia (AML) naphthoquinones.
StructureAML Cell LinesIC50 (μM)In VivoReferences
Mono- or di-substituted monomeric naphthoquinones
Molecules 24 03121 i001
Menadione
2-methyl-1,4-naphthoquinone
HL-60, U937Not reported directlyNot testedYeo HS, et al. 2012
Molecules 24 03121 i002
Juglone
5-hydroxyl-1,4-naphthoquinone
HL-608Not testedXu HL, et al., 2010
Molecules 24 03121 i003
Lawsone
2-hydroxyl-1,4-naphthoquinone
HL-60>50Not testedEsteves-Souza A, et al., 2008
Molecules 24 03121 i004
2-glycinyl-1,4-naphthoquinone
HL-603Not testedde Moraes TA, et al., 2014
Molecules 24 03121 i005
Plumbagin
5-hydroxy-2-methyl-1,4-naphthoquinone
Kasumi-1, HL-60, U9370.6–1.4Activity: Tumor volume reduction
Tolerability: No significant weight loss, tissue damage or behavior change (Xu KH, et al.)
Kawiak A, et al., 2007
Xu KH, et al., 2010
Gaascht F, et al., 2014
Kong X, et al., 2017
Zhang J, et al., 2017
Molecules 24 03121 i006
Lapachol
2-hydroxy-3-(3-methyl-2-butenyl)-1,4-naphthoquinone
Kasumi-1
Primary cells from patients
HL-60
Not reported directly
>50
Not testedde Souza Reis FR, et al., 2013
Esteves-Souza A, et al., 2008
Molecules 24 03121 i007
Atovaquone
trans-2-[4-(4-chlorophenyl)cyclohexyl]-3-hydroxy-1,4-naphthalenedione
MOLM13, MV4-11, THP-1, NB4, Kasumi-1, HL-60, KG1, HEL13.7–98.9Decreased diseased burden and prolonged survival in the treatment group compared to the control group (Stevens AM, et al.)Xiang M, et al., 2016
Stevens AM, et al., 2017
Molecules 24 03121 i008
2-((4-((3-aminopropyl)(benzyl)amino)butyl) amino)naphthalene-1,4-dione
HL-6010.5Not testedEsteves-Souza A, et al., 2008
Molecules 24 03121 i009
Ramentaceone
7-Methyljuglone
HL-608.8Not testedKawiak A, et al., 2012
Molecules 24 03121 i010
Cordiaquinone J
HL-602.7Not tested Marinho-Filho JD., et al., 2010
Molecules 24 03121 i011
TW-92
2-chloro-3-(3-amino-4-methyl-phenylamino)-1,4-naphthoquinone
U937, peripheral blood mononuclear cells from AML patients3.2Not testedHallak M, et al., 2009
Multi-substituted monomeric naphthoquinones
Molecules 24 03121 i012
Shikonin
5,8-dihydroxy-2[(1R)-hydroxy-4-methylpent-3-en-1-yl]-3-methyl-1,4-naphthoquinone
HL-60, U937,
primary AML cells
3.8Prolonged survival observed in the treatment group compared to the control group (Yang H, et al.)Yang H, et al., 2009
Zhang B, et al. 2012
Zhao Q, et al., 2015
Trivedi, et al., 2016
Molecules 24 03121 i013
SH-7
1-(1,4-dihydro-5,8-dihydroxy-1,4-dioxonaphthalen-2-yl)-4-methylpent-3-enylfuran-2-caroxylate
HL-602Not testedYang F, et al., 2006
Molecules 24 03121 i014
5,8-dihydroxy-2-(1-hydroxy-2-nitroethyl)-1,4-naphthoquinone
HL-600.14Not testedBeretta GL, et al., 2017
Molecules 24 03121 i015
3-butyl-2-chloro-5,7-dimethoxy-1,4-naphthoquinone
HL-603.8Not testedLi K, et al., 2018
Heterocyclic monomeric naphthoquinones
Molecules 24 03121 i016
FNQ3 (Furanonaphthquinone)
2-Methylnaphtho[2–b]furan-4,9-dione
HL60, NB-4, U937, THP1, primary AML cells5.9–8.2 Not testedDesmond JC, et al., 2005
Molecules 24 03121 i017
FN6-one (Furonaphthoquinone)
HL-60, U9370.87–3.0Not testedInagaki R, et al., 2013
Molecules 24 03121 i018
Nor-β-lapachone
HL-60, KG1, K5620.4–17.2Not testedda Silva EN Jr, et al. 2007, 2010
Cavalcanti, et al., 2011
Araújo AJ, et al., 2012
Cardoso, et al., 2014
Molecules 24 03121 i019
β-lapachone
3,4-dihydro-2,2-dimethyl-2H-naphtho[1–b]pyran-5,6-dione
HL-607.1Not testedPlanchon SM, et al., 1995, 1999
da Silva EN Jr, et al. 2007, 2010
Inagaki, et al., 2015
Molecules 24 03121 i020
Dunnione
HL-600.9 Not testedInagaki, et al., 2015
Molecules 24 03121 i021
LQB-118 (pterocarpanquinone)
HL60, U937,
Kasumi-1
6–9Non-cytotoxic to normal BM cells in healthy control mice.de Souza Reis, et al., 2013, Nestal De Moraes, et al., 2014
Dimeric naphthoquinones
Molecules 24 03121 i022
3-bromo-3′-hydroxyl-dimeric 1,4-naphthoquinone
MOLM-14, THP-1, primary AML cells0.36–8.5Tested for tolerability and side effects with IP and SC injections.Lapidus, et al., 2016
Molecules 24 03121 i023
3,3′-bis-aziridinyl-dimeric 1,4-naphthoquinone
MOLM-14, THP-1, primary AML cells 0.18–2Tested for tolerability with IP injections.Carter-Cooper, et al., 2017
BM = bone marrow, IP = intraperitoneally, SC = subcutaneously.

Share and Cite

MDPI and ACS Style

Lee, M.H.; Lapidus, R.G.; Ferraris, D.; Emadi, A. Analysis of the Mechanisms of Action of Naphthoquinone-Based Anti-Acute Myeloid Leukemia Chemotherapeutics. Molecules 2019, 24, 3121. https://doi.org/10.3390/molecules24173121

AMA Style

Lee MH, Lapidus RG, Ferraris D, Emadi A. Analysis of the Mechanisms of Action of Naphthoquinone-Based Anti-Acute Myeloid Leukemia Chemotherapeutics. Molecules. 2019; 24(17):3121. https://doi.org/10.3390/molecules24173121

Chicago/Turabian Style

Lee, Michelle H., Rena G. Lapidus, Dana Ferraris, and Ashkan Emadi. 2019. "Analysis of the Mechanisms of Action of Naphthoquinone-Based Anti-Acute Myeloid Leukemia Chemotherapeutics" Molecules 24, no. 17: 3121. https://doi.org/10.3390/molecules24173121

Article Metrics

Back to TopTop