Next Article in Journal
A Chiral-LC-MS Method for the Simultaneous Quantification of Short-Chain Fatty Acids and D/L-Lactate in the Ruminal Fluid of Dairy Cows
Next Article in Special Issue
Antimicrobial Activity and Phytochemical Characterization of Baccharis concava Pers., a Native Plant of the Central Chilean Coast
Previous Article in Journal
Preparation and Characterization of Pullulan-Based Packaging Paper for Fruit Preservation
Previous Article in Special Issue
Anti-Psoriatic Activity of Black, Green and White Tea Extracts from Southeastern China
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Anticancer Potential and Safety Profile of β-Lapachone In Vitro

by
Karina Motta Melo Lima
1,2,*,
Luana França Calandrini de Azevedo
1,
Jorge Dores Rissino
1,
Valdicley Vieira Vale
3,
Erica Vanessa Souza Costa
3,
Maria Fani Dolabela
3,
Cleusa Yoshiko Nagamachi
1 and
Julio Cesar Pieczarka
1
1
Center for Advanced Studies of the Biodiversity, Guamá Science and Technology Park, Federal University of Pará, Belém 66075-750, PA, Brazil
2
Campus Tomé Açu, Federal Rural University of the Amazon, Tomé Açu 68680-000, PA, Brazil
3
Postgraduate Program in Pharmaceutical Innovation, Federal University of Pará, Belém 66075-110, PA, Brazil
*
Author to whom correspondence should be addressed.
Molecules 2024, 29(6), 1395; https://doi.org/10.3390/molecules29061395
Submission received: 12 December 2023 / Revised: 13 March 2024 / Accepted: 15 March 2024 / Published: 21 March 2024
(This article belongs to the Special Issue Advances in Natural Products and Their Biological Activities)

Abstract

:
Ipê is a plant of the Bignoniaceae family. Among the compounds extracted from this tree, lapachol is notable because its structural modification allows the production of β-lapachone, which has anticancer properties. The objective of this work was to test this hypothesis at a cellular level in vitro and assess its potential safety for use. The following tests were performed: MTT cell viability assay, apoptotic index determination, comet assay, and micronucleus test. The results showed that β-lapachone had a high cytotoxic capacity for all cell lines tested: ACP02 (gastric adenocarcinoma cells), MCF7 (breast carcinoma cells), HCT116 (colon cancer cells) and HEPG2 (hepatocellular carcinoma cells). Regarding genotoxicity, the exposure of cells to sublethal doses of β-lapachone induced DNA damage (assessed by the comet assay) and nuclear abnormalities, such as nucleoplasmic bridges and nuclear buds (assessed by the micronucleus test). All tested cell lines responded similarly to β-lapachone, except for ACP02 cells, which were relatively resistant to the cytotoxic effects of the compound in the MTT test. Our results collectively indicate that although β-lapachone showed antiproliferative activity against cancer cell lines, it also caused harmful effects in these cells, suggesting that the use of β-lapachone in treating cancer should be carried out with caution.

Graphical Abstract

1. Introduction

Plant-derived agents are important sources of drug constituents and have notably contributed to modern medicine. The bioactive compounds of plants comprise various phytochemicals arising from secondary metabolism; they show therapeutic actions in humans and can be refined to produce drugs [1,2,3].
Ipê is a plant of the Bignoniaceae family. It is native to tropical forests, and infusions obtained from the species have been used medicinally for centuries. The active components of Ipê extract include large quantities of lapachol, which can be structurally altered to the desirable compound, naphthoquinone (β-lapachone) [4,5].
β-Lapachone has been widely studied for its pharmacological effects. Studies have found that it exhibits cytotoxic potential in different types of cancer, including breast cancer [6], prostate cancer [7], multiple myeloma [8,9], hepatocellular carcinoma [10], pancreatic cancer [11,12], human leukemia [13], and others. When studying compounds with potential anticancer activity and evaluating new medicines for use in treating illness, it is important to determine whether they are safe to use. The genotoxic and mutagenic potential of a candidate therapeutic should be examined, since changes in genetic material can cause effects on somatic and/or germ cells and lead to the formation of cancer and/or malformation during embryonic development [14].
The present work aims to determine and compare the antiproliferative activity of β-lapachone in various neoplastic cell lines and evaluate its safety in the context of genotoxicity and mutagenicity.

2. Results

2.1. MTT Test

As shown in Figure 1, the MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazoliumbromide) assay revealed that β-lapachone dose-dependently decreased survival among four neoplastic cell lines. In the ACP02 and MCF7 cell lines, the decrease was significant from 4 µg/mL in relation to the other concentrations, while in the HCT116 and HEPG2 lines, the decrease was significant from 2 µg/mL in relation to the other concentrations. A statistical comparison of the IC50 values for each cell line (Figure 2) showed that ACP02 cells were more resistant than the other cell lines, which had IC50 values similar to one another for β-lapachone.

2.2. Apoptotic Index

When we tested the apoptotic index in the four neoplastic cell lines under treatment with β-lapachone, we obtained consistent results in all four cell lines. Compared to the negative control, apoptosis was significantly induced by the highest concentration tested (1.5 µg/mL) and by doxorubicin (DOX, positive control), but not by the other tested concentrations of β-lapachone (Figure 3A). As shown in Figure 3B, we compared the percentage of apoptotic cells seen at the highest concentration tested (1.5 µg/mL) and found no significant difference among the four cell lines.

2.3. Comet Assay

The comet assay was used to evaluate DNA damage. As shown in Figure 4, treatment with the highest dose of β-lapachone (1.5 µg/mL) and the positive control significantly increased the DNA damage index relative to the negative control.

2.4. Micronucleus Test

The micronucleus test revealed that the cytokinesis block proliferation index (CBPI) values were significantly lower among cells treated with the highest concentration of β-lapachone (1.5 µg/mL), compared to the control group (Figure 5A). Regarding the MN frequency, only the positive control showed significantly higher values compared to the other groups (Figure 5B). Other nuclear abnormalities (NAs, described further below) were significantly increased in cells treated only with the lowest concentration (0.5 µg/mL) of β-lapachone and the positive control, relative to the other treatments (Figure 5C).
Regarding the nuclear abnormalities found in treatments with β-lapachone, the presence of nuclear sprouts (Figure 6C) and nucleoplasmic bridges (Figure 6D) stood out. In the control group, the occurrence of binucleated cells without micronuclei was observed (Figure 6A). Only the positive control showed the occurrence of micronuclei in binucleated cells (Figure 6B).

3. Discussion

Our results are consistent with previous reports that β-lapachone exerts antiproliferative activity and is a promising candidate for treating cancer. The low IC50 found in our results shows that a very small amount of β-lapachone could cause cell death in the tested cancer cell lines. When we compared the sensitivity of the tested cell lines, we observed that ACP02 cells were relatively resistant to β-lapachone. This cell line is derived from stomach cancer; this disease is difficult to prognose and exhibits low response rates to chemotherapy or radiochemotherapy [15], and these aspects may be related to our present observations. Different cancer cell lines often vary in their sensitivity to a given agent, at least partly reflecting tumor heterogeneity and the complexity of many types of cancer [16]. However, the IC50 of β-lapachone toward ACP02 cells was around 3 µg/mL. This still represents a fairly high cytotoxic potential; previous studies reporting that β-lapachone and its derivatives had promising cytotoxicity in several human cancer cell lines based this conclusion on observing IC50 values less than 4 μg/mL [16,17].
Numerous published studies have identified various action mechanisms of β-lapachone. These include its ability to induce the formation of reactive oxygen species (ROS) via catalysis by NAD(P)H:quinone oxidoreductase 1(NQO1); this may benefit potential antitumor activity, since NQO1 is usually expressed more significantly in tumors than in normal cells [12]. Other action mechanisms include the inhibition of topoisomerase action and modification of p53 expression levels [4,18].
Regarding the involved cellular death pathway(s), different types of cancer have been associated with different cell death modes induced by β-lapachone, including apoptosis [19,20,21], autophagy [22], cytostasis [23], and necroptosis [24,25]. In this work, we observed significant β-lapachone-induced apoptosis in all four cell lines. Pro-apoptotic activity is considered advantageous in anticancer therapy, given that a cell undergoing apoptosis is phagocytized by neighboring cells or macrophages before it releases any cellular content. Necrosis, in contrast, involves the release of toxins that can spread to healthy cells in tissues near the primary tumor and thereby cause local inflammation [26,27,28,29].
Regarding the induction of apoptosis, we observed a discrepancy in the cytotoxicity values observed by the MTT test, since the apoptosis index already reveals high occurrence at doses below the IC50 detected by the MTT. These data corroborate a previous report that the MTT test has lower sensitivity for detection [30]. The authors suggested that the fluorescent dye used to detect apoptosis increases the sensitivity of the test, as it allows visualization of morphological changes characteristic of early-stage apoptosis, prior to the decrease in mitochondrial respiration that is detected by the MTT test. Regardless, our results obtained with both assays indicated that β-lapachone has high cytotoxic potential in the four cancer cell lines tested herein.
Despite the previous and present findings, many challenges remain unresolved regarding the potential use of β-lapachone in anticancer therapy, including issues related to its solubility and bioavailability in the body [4,18]. Various strategies have been used in an attempt to overcome these issues. For example, β-lapachone-containing nanotherapeutics have been developed and shown to exhibit greater antiproliferative effects than free β-lapachone, along with the ability to target the cancer site and/or cancer cells [31,32,33]. Other efforts have sought to identify β-lapachone derivatives with improved biological potency, solubility, and/or bioavailability [18,34,35].
Further work is still needed to fully assess the safety profile of β-lapachone, including any potential secondary effects on healthy cells and/or at sublethal concentrations. The present work is one of the few studies that has evaluated the potential for β-lapachone to cause genotoxicity and mutagenicity. The nuclear abnormalities observed in the micronucleus test may support a previous suggestion that the formation of nucleoplasmic bridges and that of nuclear buds are connected via induction of the break–fusion–bridge cycle [36]. In this model, a genotoxic substance causes breaks at the ends of chromosomes, leading them to attach to their sister chromatids. The pairs thus remain together during the anaphase and when chromosomes seek to migrate to opposite poles, forming the nucleoplasmic bridge. An attached chromosome will tend to break unevenly, generating gene amplifications in some cells. In this model, the amplifications may tend to localize to the periphery of nuclei, where they form nuclear buds.
Notably, we observed more frequent nuclear abnormalities at the lower concentrations tested. We speculate that the high level of DNA damage caused by the highest concentrations of β-lapachone (detected by the comet assay) could trigger DNA-damage-related cell death, meaning that nuclear abnormalities were seen less frequently. This fact could be confirmed by the increase in apoptosis at the highest concentration, a factor that may have been induced by the occurrence of excessive DNA damage [18,37,38]. In this sense, the β-lapachone-induced increase in DNA damage detected in the comet assay appears to be directly related to the ability of this agent to induce NQO1-catalyzed ROS generation [18]. Another factor that may be involved in the ability of β-lapachone to induce nuclear abnormalities only at lower concentrations is the possible cytostatic effect of higher concentrations, as evidenced by our CBPI results. In this case, the changes would not be visualized through the micronucleus test because the cells do not progress through the cell cycle to complete mitosis. A cytostatic effect of β-lapachone has also been reported by other authors [4,16,39].
In sum, the nuclear changes herein detected in cancer cells treated with relatively low concentrations of β-lapachone should be considered further as potential side effects relevant to the potential use of β-lapachone in anticancer therapy.

4. Materials and Methods

4.1. Sample Collection and Extraction and Characterization of β-Lapachone

Sample collection and extraction and characterization of β-lapachone were carried out by the research group at the Laboratory of Pharmacology and Neglected Diseases at the Federal University of Pará (UFPA). All β-lapachone used in this work was extracted from sawdust of Ipê wood, which was collected from a sawmill located in the state of Pará, Brazil.
For extraction of β-lapachone, 1.859 g of sawdust was mixed with 10 L of 1% NaHCO3 solution. The mixture was left to rest for 45 min and then filtered. The pH of the extract was adjusted to pH 3 with 6M HCl until the red solution changed color, forming a yellow precipitate. The material was filtered and was retained and placed in a desiccator to remove moisture. To obtain lapachol, the residue was scraped off the filter paper and 5.540 g of this material was fractionated on a chromatographic column (125 g of MERK silica gel 70–230 mm; height, 38 cm; and width, 4 cm), using dichloromethane as the mobile phase. Thin layer chromatography (TLC) was performed using dichloromethane–hexane (8:2) as the eluent. The fraction richest in lapachol was fractionated again and the fraction with the highest pure lapachol content was chosen for use.
The structural determination of lapachol was carried out using nuclear magnetic resonance spectroscopy (NMR) of hydrogen (1H NMR) and carbon 13 (13C NMR). The 1H NMR and 13C NMR spectra were obtained by a Varian Unity Plus 300 apparatus using deuterated chloroform (CDCl3) and tetramethylsilane (TMS) solution as internal references. The chemical shift values were measured in parts per million (ppm) in relation to the TMS and the coupling constants (J) in Hertz (Hz).
To synthesize β-lapachone, 500 mg of lapachol was mixed with concentrated sulfuric acid (1.5 mL) in an ice bath for approximately 10 min and then ice-cold distilled water (50 mL) was added. This solution was filtered and recrystallized with ethanol and the obtained orange solid was solubilized in deuterated chloroform. Nuclear magnetic resonance analysis (Bruker Advance DPX 200 NMR Spectrometer, Billerica, MA, USA) was used to confirm the structure of β-lapachone (Supplementary Materials; Figures S1 and S2, Table S1).

4.2. Cell Lines and Growing Conditions

For in vitro tests, the following human cells of different neoplastic tissue origins were used: ACP02 (gastric adenocarcinoma cells), MCF7 (breast carcinoma cells), HCT116 (colon cancer cells) and HEPG2 (hepatocellular carcinoma cells). Some cells were purchased from the Rio de Janeiro Cell Bank and others were kindly provided by the Human Cytogenetics Laboratory at UFPA.
All cell lines were cultivated in DMEM (Dulbecco’s Modified Minimum Essential Media) supplemented with 10% Fetal Bovine Serum (FBS) and amphotericin (2.5 μg/mL). The cells were kept in plastic bottles and incubated in a sterile incubator in an atmosphere of 5% CO2 at 37 °C until the tests were carried out.

4.3. Cell Viability

4.3.1. MTT Test

Cells were seeded in a 96-well plate at 5 × 104 per well and the plates were incubated in a CO2 incubator at 37 °C. After 24 h, the wells were loaded with seven concentrations of β-lapachone (0.25–16 µg/mL) that were chosen based on our preliminary tests and previous reports in the literature. DMEM was used as a negative control and doxorubicin (DOX, 100 µg/mL) was used as a positive control. After 24 h of exposure, the cells were exposed to 100 μL of MTT (5 mg/mL) for 3 h. The MTT was removed and 100 μL of DMSO was added to each well. After 1 h, the absorbance was read on a spectrophotometer (Epoch Biotek, Winooski, USA) at a wavelength of 570 nm using the Gen5 program (2003) version 2.03.1. Cell viability (%S) was determined using the following formula: %S = 100 × [(Atested − Ablank)/(Acontrol − Ablank)], where A represents the absorbance of each well and blank represents the results from an empty well. The IC50 (inhibitory concentration of 50% of cells) was calculated using the Prisma GraphPad version 5 program.

4.3.2. Apoptotic Index

Cells were seeded in a 12-well plate at 8 × 104 cells/well and incubated in a CO2 incubator at 37 °C containing 95% air and 5% CO2. After 24 h, the cells were exposed to 0.5 µg/mL, 1 µg/mL, and 1.5 µg/mL of β-lapachone (values corresponding to sublethal doses: approximately 25, 50, and 75% of the IC50). DMEM was used as the negative control and DOX (100 µg/mL) was used as the positive control. After 24 h of exposure, the cells were trypsinized and removed from the plate. Cells were centrifuged in a Falcon tube; 50 μL of cells were mixed and removed and 2 μL of acridine orange solution (100 µg/mL) was used to prepare the slides. Analysis was carried out using a Nikon fluorescence microscope (HS 50S) with a 40× objective and FITC filter. Three hundred cells were counted per treatment, based on previous reports [40,41], and the analytic parameters were as previously described [42]. Cells were defined as follows: viable cells were stained green and had organized chromatin and apoptotic cells were stained green and had highly fragmented chromatin (presence of apoptotic bodies). The percentage of apoptotic cells was calculated as follows: % of apoptotic cells = 100 × (number of cells in apoptosis/total cells).

4.4. Genotoxicity Assays

For the genotoxicity assays, we used HepG2 cells. This cell line is commonly recommended for such work because its hepatic origin and capacity to metabolize compounds improve its ability to reflect possible correlations with in vivo effects [43]. HepG2 cells were seeded in 12-well plates at 8 × 104 cells/well, maintained for 24 h in a sterile incubator at an atmosphere of 5% CO2 and 37 °C, and then exposed to 0.5 µg/mL, 1 µg/mL, and 1.5 µg/mL of β-lapachone (approximately 25, 50, and 75% of the IC50 determined by the MTT test, respectively). DMEM was used as the negative control and 5 µg/mL of DOX and 0.02 µg/mL were used as positive controls for the comet assay and micronucleus test, respectively. Treated cells were trypsinized and the comet assay and micronucleus test were performed.

4.4.1. Comet Assay

The comet assay was carried out according to a modified version of the published methodology [44]. After 3 h of exposure to β-lapachone, the cells were collected and resuspended in DMEM, and 20 µL of cell suspension was mixed with 0.5% low-melting-temperature agarose. The mixture was placed on slides that had been pre-coated with normal agarose (1.5%) and covered with a coverslip. The slides were incubated for 15 min at 4 °C and then placed in a lysis solution (2.5 M NaCl, 100 mM EDTA, and 10 mM Tris; pH 10.0–10.5) containing 1% Triton X-100 and 10% DMSO. After 24 h, the slides were transferred to a horizontal electrophoresis vat and covered with alkaline buffer (300 mM NaOH and 1 mM EDTA, pH > 13) for 30 min and then subjected to electrophoresis for another 30 min at 0.8 V/cm. Finally, the slides were washed three times for 5 min each with deionized water and then fixed for 5 min in P.A. ethyl alcohol. The slides were dried and stored in a refrigerator until analysis. The slides were stained with ethidium bromide (20 μg/mL) and analyzed using a Nikon H550S fluorescence microscope (510–560 nm filter, 590 nm filter barrier, and 40× magnification). Comets were categorized based on tail size [45] and 100 cells were analyzed per treatment. The damage index (DI) was calculated as a percentage using the following formula: DI = 100 × [(1 × n1) + (2 × n2) + (3 × n3) + (4 × n4)/n], where n represents the total number of cells analyzed and n1 to n4 represent the number of cells with damage levels from 0 to 4, respectively.

4.4.2. Micronucleus Test

The micronucleus test was carried out according to the standard protocol [46], with Cytochalasin B (6 µg/mL) added 24 h before the cells were removed. After 24 h of exposure to β-lapachone, the cells were trypsinized, transferred to a Falcon tube, treated with 2.5 mL of hypotonic solution (KCl) for 3 min, and then treated with 2 mL of Carnoy’s fixative (methanol–acetic acid, 3:1). The cells were centrifuged, the supernatant was removed, another 1 mL of Carnoy’s fixative was added, and the sample was stored at 4 °C. For analysis, the cells were dripped onto slides and analyzed under a Zeiss optical microscope with a 40× objective. For each slide, 500 cells were analyzed and quantified. The cytokinesis block proliferation index (CBPI) was calculated using the following formula: CBPI = [M1 + 2(M2) + 3(M3) + 4(M4)]/N, where M1, M2, M3, and M4 represent the number of cells with 1, 2, 3, and 4 nuclei, respectively, and N is the total number of viable cells. To calculate MN and other nuclear abnormalities (NAs), 1000 binucleated cells per slide were assessed for micronuclei or other NAs, and their frequencies were calculated as follows: fMN or NA = nºMN or NA/1000.

4.5. Statistical Analysis

Statistical analyses were performed using an ANOVA or Kruskal–Wallis test, which were chosen according to the normality standard of the data defined using the Kolmogorov–Smirnov test. All statistical analyses were carried out using the Bioestat 5.0 program (p ≤ 0.05).

5. Conclusions

Our present results and the previous reports clearly support the idea that β-lapachone may be a promising candidate as a cancer chemotherapeutic. However, it will be important to further evaluate its safety for use, especially at sublethal concentrations. Here, we have shown that β-lapachone induces genotoxic effects in vitro at relatively low exposure concentrations. Therefore, further studies are needed to establish threshold exposure concentrations. Moreover, additional studies should be carried out using technologies and/or β-lapachone analogs for targeted delivery to the relevant organ(s), thereby improving the therapeutic benefits and minimizing adverse effects.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/molecules29061395/s1: Figure S1: Representation of the hydrogen (H1) nuclear magnetic resonance spectrum of β-lapachone (CDCl3, 200 MHz); Table S1: Data from the attributions of nuclear magnetic resonance signals from β-lapachone (1H 200 MHz,13C 50 MHz, CD3OD); Figure S2. Representation of the carbon 13 (C13) nuclear magnetic resonance spectrum of β-lapachone (CDCl3, 200 MHz).

Author Contributions

Conceptualization, investigation, and writing—original draft preparation: K.M.M.L.; methodology: K.M.M.L., L.F.C.d.A., J.D.R., V.V.V. and E.V.S.C.; validation and formal analysis: M.F.D., C.Y.N., and J.C.P.; resources, supervision, and funding acquisition: J.C.P.; writing—review and editing and visualization: L.F.C.d.A., M.F.D., C.Y.N. and J.C.P. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the Coordination for the Improvement of Higher Education Personnel (CAPES; Pro-Amazônia Biodiversity and Sustainability Grant 047/2012) and the National Bank for Economic and Social Development—BNDES (Operation 2.318.697.0001).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are available from the corresponding author upon request.

Acknowledgments

We thank the National Council for Scientific and Technological Development (CNPQ) for Research Productivity Grants to Cleusa Yoshiko Nagamachi (3171770/2021-7) and Julio Cesar Pieczarka (307154/2021-1). We also thank the Postgraduate Program in Biotechnology at the Federal University of Pará; this work formed part of Karina Motta Melo Lima’s post-doctoral studies carried out under the aforementioned program (scholarship from the National Post-Doctoral Program of the Coordination for the Improvement of Higher Education Personnel—PNPD/CAPES).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Pandey, M.; Debnath, M.; Gupta, S.; Chikara, S.K. Phytomedicine: An ancient approach turning into future potential source of therapeutics. J. Pharmacogn. Phytother. 2011, 3, 27–37. [Google Scholar]
  2. Stankovic, M.S.; Radic, Z.S.; Blanco-Salas, J.; Vázquez-Pardo, F.M.; Ruiz-Téllez, T. Screening of selected species from Spanish flora as a source of bioactive substances. Ind. Crops Prod. 2017, 95, 493–501. [Google Scholar] [CrossRef]
  3. Aleebrahim-Dehkordy, E.; Rafieian-Kopaei, M.; Amini-Khoei, H.; Abbasi, S. In Vitro Evaluation of Antioxidant Activity and Antibacterial Effects and Measurement of Total Phenolic and Flavonoid Contents of Quercus brantii L. Fruit Extract. J. Diet. Suppl. 2019, 16, 408–416. [Google Scholar] [CrossRef]
  4. Da Costa, D.C.F.; Rangel, L.P.; Martins-Dinis, M.M.D.C.; Ferretti, G.D.S.; Ferreira, V.F.; Silva, J.L. Anticancer Potential of Resveratrol, β-Lapachone and Their Analogues. Molecules 2020, 25, 2–22. [Google Scholar]
  5. Pradeep, K.S.; Mallya, P.; Jain, V. Naphthoquinones in the Treatment of Cancer. Pharm. J. Pharm. Sci. Res. 2020, 12, 587–590. [Google Scholar]
  6. Wuerzberger, S.M.; Pink, J.J.; Planchon, S.M.; Byers, K.L.; Bornmann, W.G.; Boothman, D.A. Induction of apoptosis in MCF-7: WS8 breast cancer cells by β-Lapachone. Cancer Res. 1998, 58, 1876–1885. [Google Scholar]
  7. Dong, Y.; Chin, S.F.; Blanco, E.; Bey, E.A.; Kabbani, W.; Xie, X.J.; Bornmann, W.G.; Boothman, D.A.; Gao, J. Intratumoral delivery of β-lapachone via polymer implants for prostate cancer therapy. Clin. Cancer Res. 2009, 15, 131–139. [Google Scholar] [CrossRef]
  8. Li, Y.; Li, C.J.; Yu, D.; Pardee, A.B. Potent Induction of Apoptosis by β-Lapachone in Human Multiple Myeloma Cell Lines and Patient Cells. Mol. Med. 2000, 6, 1008–1015. [Google Scholar] [CrossRef]
  9. Gupta, D.; Podar, K.; Tai, Y.T.; Lin, B.; Hideshima, T.; Akiyama, M.; LeBlanc, R.; Catley, L.; Mitsiades, N.; Mitsiades, C.; et al. β-lapachone, a novel plant product, overcomes drug resistance in human multiple myeloma cells. Exp. Hematol. 2002, 30, 711–720. [Google Scholar] [CrossRef]
  10. Park, E.J.; Min, K.J.; Lee, T.J.; Yoo, Y.H.; Kim, Y.S.; Kwon, T.K. β-Lapachone induces programmed necrosis through the RIP1-PARP-AIF-dependent pathway in human hepatocellular carcinoma SK-Hep1 cells. Cell Death Dis. 2014, 5, e1230. [Google Scholar] [CrossRef]
  11. Ough, M.; Lewis, A.; Bey, E.A.; Gao, J.; Ritchie, J.M.; Bornmann, W.; Boothman, D.A.; Oberley, L.W.; Cullen, J.J. Efficacy of beta-lapachone in pancreatic câncer treatment: Exploiting the novel, therapeutic target NQO1. Cancer Biol. Ther. 2005, 4, 95–102. [Google Scholar] [CrossRef]
  12. Silvers, M.A.; Deja, S.; Singh, N.; Egnatchik, R.A.; Sudderth, J.; Luo, X.; Beg, M.S.; Burgess, S.C.; DeBerardinis, R.J.; Boothman, D.A.; et al. The NQO1 bioactivatable drug, β-lapachone, alters the redox state of NQO1 pancreaztic cancer cells, causing perturbation in central carbon metabolism. J. Biol. Chem. 2017, 292, 18203–18216. [Google Scholar] [CrossRef]
  13. Chau, Y.P.; Shiah, S.G.; Don, M.J.; Kuo, M.L. Involvement of hydrogen peroxide in topoisomerase inhibitor β-lapachone-induced apoptosis and differentiation in human leukemia cells. Free Radic. Biol. Med. 1998, 24, 660–670. [Google Scholar] [CrossRef]
  14. Da Silva, J.; Erdtmann, B.; Henriques, J.A.P. Genética Toxicológica, 1st ed.; Alcancer: Porto Alegre, Brasil, 2003; 424p. [Google Scholar]
  15. Schauer, M.; Peiper, M.; Theisen, J.; Knoefel, W. Prognostic factors in patients with diffuse type gastric cancer (linitis plastica) after operative treatment. Eur. J. Med. Res. 2011, 16, 29–33. [Google Scholar] [CrossRef]
  16. Dias, R.B.; De Araújo, T.B.S.; De Freitas, R.D.; Rodrigues, A.C.B.C.; Sousa, L.P.; Salesb, C.B.S.; Valverdea, L.F.; Soaresa, M.B.P.; Dos Reisa, M.G.; Coletta, R.D.; et al. β-Lapachone and its iodine derivatives cause cell cycle arrest at G2/M phase and reactive oxygen species-mediated apoptosis in human oral squamous cell carcinoma cells. Free Radic. Biol. Med. 2018, 126, 87–100. [Google Scholar] [CrossRef]
  17. Boik, J. Natural Compounds in Cancer Therapy; Oregon Medical Press: Princeton, MI, USA, 2001. [Google Scholar]
  18. Gong, Q.; Hu, J.; Wang, P.; Li, X.; Zhang, X. A comprehensive review on β-lapachone: Mechanisms, structural modifications, and therapeutic potentials. Eur. J. Med. Chem. 2021, 210, 112962. [Google Scholar] [CrossRef] [PubMed]
  19. Choi, B.T.; Cheong, J.; Choi, Y.H. β-Lapachone-induced apoptosis is associated with activation of caspase-3 and inactivation of NF-jB in human colon cancer HCT-116 cells. Anti-Cancer Drugs 2003, 14, 845–850. [Google Scholar] [CrossRef] [PubMed]
  20. Lien, Y.C.; Kung, H.N.; Lu, K.S.; Jeng, C.J.; Chau, Y.P. Involvement of endoplasmic reticulum stress and activation of MAP kinases in ß-lapachone-induced human prostate cancer cell apoptosis. Histol. Histopathol. 2008, 23, 1299–1308. [Google Scholar] [PubMed]
  21. Yu, H.Y.; Kim, S.O.; Jin, C.Y.; Kim, G.Y.; Kim, W.J.; Yoo, Y.H.; Choi, Y.H. β-lapachone-Induced Apoptosis of Human Gastric Carcinoma AGS Cells Is Caspase-Dependent and Regulated by the PI3K/ Akt Pathway. Biomol. Ther. 2014, 22, 184–192. [Google Scholar] [CrossRef] [PubMed]
  22. Park, E.J.; Choi, K.S.; Kwon, T.K. β-Lapachone-induced reactive oxygen species (ROS) generation mediates autophagic cell death in glioma U87 MG cells. Chem. Biol. Interact. 2011, 189, 37–44. [Google Scholar] [CrossRef] [PubMed]
  23. Huang, L.; Pardee, A.B. β-lapachone induces cell cycle arrest and apoptosis in human colon cancer cells. Mol. Med. 1999, 5, 711–720. [Google Scholar] [CrossRef] [PubMed]
  24. 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 β-lapachone. Proc. Natl. Acad. Sci. USA 2007, 104, 11832–11837. [Google Scholar] [CrossRef] [PubMed]
  25. Bey, E.A.; Reinicke, K.E.; Srougi, M.C.; Varnes, M.; Anderson, V.E.; Pink, J.J.; Li, L.S.; Patel, M.; Cao, L.; Moore, Z.; et al. Catalase abrogates β-lapachone-induced PARP1 hyperactivation-directed programmed necrosis in NQO1-positive breast cancers. Mol. Cancer Ther. 2013, 12, 2110–2120. [Google Scholar] [CrossRef] [PubMed]
  26. Kerr, J.F.; Wyllie, A.H.; Currie, A.R. Apoptosis: A basic biological phenomenon with wide ranging implications in tissue kinetics. Br. J. Cancer 1972, 26, 239–257. [Google Scholar] [CrossRef] [PubMed]
  27. Zhang, J.; Xu, M. Apoptotic DNA fragmentation and tissue homeostasis. Trends Cell Biol. 2002, 12, 84–89. [Google Scholar] [CrossRef]
  28. Alberts, B.; Johnson, A.; Lewis, J.; Raff, M.; Roberts, K.; Walter, P. Molecular Biology of the Cell, 5th ed.; Garland Science: New York, NY, USA, 2008; 1616p. [Google Scholar]
  29. Fulda, S. Targeting apoptosis for anticancer therapy. Semin. Cancer Biol. 2015, 31, 84–88. [Google Scholar] [CrossRef]
  30. Porfírio-Dias, C.L.; Melo, K.M.; Bastos, C.E.M.C.; Ferreira, T.A.A.; Azevedo, L.F.C.; Salgado, H.L.; Santos, A.S.; Rissino, J.D.; Nagamachi, C.Y.; Pieczarka, J.C. Andiroba oil (Carapa guianensis Aubl) shows cytotoxicity but no mutagenicity in the ACP02 gastric cancer cell line. J. Appl. Toxicol. 2020, 40, 1060–1066. [Google Scholar] [CrossRef]
  31. Jeong, S.Y.; Park, S.J.; Yoon, S.M.; Jung, J.; Woo, H.N.; Yi, S.L.; Song, S.Y.; Park, H.J.; Kim, C.; Lee, J.S.; et al. Systemic delivery and preclinical evaluation of Au nanoparticle containing β-lapachone for radiosensitization. J. Control Release 2009, 139, 239–245. [Google Scholar] [CrossRef]
  32. Park, C.; Youn, H.; Kim, H.; Noh, T.; Kook, Y.H.; Oh, E.T.; Park, H.J.; Kim, C. Cyclodextrin-covered gold nanoparticles for targeted delivery of an anti-cancer drug. J. Mater. Chem. 2009, 19, 2310–2315. [Google Scholar] [CrossRef]
  33. Blanco, E.; Bey, E.A.; Khemtong, C.; Yang, S.G.; Setti-Guthi, J.; Chen, H.; Kessinger, C.W.; Carnevale, K.A.; Bornmann, W.G.; Boothman, D.A.; et al. β-Lapachone Micellar Nanotherapeutics for Non–Small Cell Lung Cancer Therapy. Cancer Res. 2010, 70, 3896–3904. [Google Scholar] [CrossRef]
  34. Kim, S.; Lee, S.; Cho, J.Y.; Yoon, S.H.; Jang, I.; Yu, K. Pharmacokinetics and tolerability of MB12066, a beta-lapachone derivative targeting NA D(P)H: Quinone oxidoreductase 1: Two independent, double-blind, placebo-controlled, combined single and multiple ascending dose first-in-human clinical trials. Drug Des. Devel Ther. 2017, 11, 3187–3195. [Google Scholar] [CrossRef] [PubMed]
  35. Gerber, D.E.; Beg, M.S.; Fattah, F.; Frankel, A.E.; Fatunde, O.; Arriaga, Y.; Dowell, J.E.; Bisen, A.; Leff, R.D.; Meek, C.C.; et al. Phase 1 study of ARQ 761, a β-lapachone analogue that promotes NQO1-mediated programmed cancer cell necrosis. Br. J. Cancer 2018, 119, 928–936. [Google Scholar] [CrossRef] [PubMed]
  36. Fenech, M.; Kirsch-Volders, M.; Natarajan, A.T.; Surralles, J.; Crott, J.W.; Parry, J.; Norppa, H.; Eastmond, D.A.; Tucker, J.D.; Thomas, P. Molecular mechanisms of micronucleus, nucleoplasmic bridge and nuclear bud formation in mammalian and human cells. Mutagenesis 2011, 26, 125–132. [Google Scholar] [CrossRef] [PubMed]
  37. Aitken, R.J.; Koppers, A.J. Apoptosis and DNA damage in human spermatozoa. Asian J. Androl. 2011, 13, 36–42. [Google Scholar] [CrossRef]
  38. Ross, W.P. DNA damage and the balance between survival and death in cancer biology. Nat. Rev. Cancer. 2016, 16, 20–33. [Google Scholar] [CrossRef] [PubMed]
  39. Villamil, S.F.; Stoppani, A.O.M.; Dubin, M. Redox Cycling of β-Lapachone and Structural Analogues in Microsomal and Cytosol Liver Preparations. Meth. Enzymol. 2004, 378, 67–87. [Google Scholar]
  40. Alcântara, D.D.; Ribeiro, H.F.; Cardoso, P.C.; Araújo, T.M.; Burbano, R.R.; Guimarães, A.C.; Khayat, A.S.; De Oliveira Bahia, M. In vitro evaluation of the cytotoxic and genotoxic effects of artemether, an antimalarial drug, in a gastric cancer cell line (PG100). J. Appl. Toxicol. 2013, 33, 151–156. [Google Scholar] [CrossRef]
  41. Calandrini de Azevedo, L.F.; Ferreira, T.A.A.; Melo, K.M.; Porfírio Dias, C.L.; Bastos, C.E.M.C.; Santos, S.F.; Santos, A.S.; Nagamachi, C.Y.; Pieczarka, J.C. Aqueous ethanol extract of Libidibia ferrea (Mart. Ex Tul) L.P. Queiroz (juca) exhibits antioxidant and migration-inhibiting activity in human gastric adenocarcinoma (ACP02) cells. PLoS ONE 2020, 15, e0257134. [Google Scholar] [CrossRef]
  42. Melo, K.M.; Grisolia, C.K.; Pieczarka, J.C.; De Souza, L.R.; De Souza Filho, J.; Nagamachi, C.Y. FISH in micronucleus test demonstrates aneugenic action of rotenone in a common freshwater fish species, Nile tilapia (Oreochromis niloticus). Mutagenesis 2014, 29, 215–219. [Google Scholar] [CrossRef]
  43. Ribeiro, L.R.; Salvadori, D.M.F.; Marques, E.K. Mutagenese Ambiental, 1st ed.; Editora Ulbra: Canoas, Brasil, 2003; 355p. [Google Scholar]
  44. Singh, N.P.; McCoy, M.T.; Tice, R.R.; Schneider, E.L. A simple technique for quantitation of low levels of DNA damage in individual cells. Exp. Cell Res. 1998, 175, 184–191. [Google Scholar] [CrossRef]
  45. Collins, A.R.; Ma, A.G.; Duthie, S.J. The kinetics of repair and oxidative DNA damage (Strand breaks and oxidized pyrimidines) in human cells. Mutat. Res. 1995, 336, 69–77. [Google Scholar] [CrossRef] [PubMed]
  46. Fenech, M. The cytokinesis-block micronucleus technique: A detailed description of the method and its application to genotoxicity studies in human populations. Mutat. Res. Fundam. Mol. Mech. Mutagen. 1993, 285, 35–44. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The survival of neoplastic cell lines under β-lapachone treatment was evaluated using the MTT test. The tested cell lines were (A) ACP02, (B) MCF7, (C) HCT116, and (D) HEPG2. Statistical analysis was performed using ANOVA parametric test (multiple comparisons—Tukey); * indicates a significant difference relative to the other treatments.
Figure 1. The survival of neoplastic cell lines under β-lapachone treatment was evaluated using the MTT test. The tested cell lines were (A) ACP02, (B) MCF7, (C) HCT116, and (D) HEPG2. Statistical analysis was performed using ANOVA parametric test (multiple comparisons—Tukey); * indicates a significant difference relative to the other treatments.
Molecules 29 01395 g001
Figure 2. IC50 values of β-lapachone in four neoplastic cell lines. Statistical analysis was performed using ANOVA parametric test (multiple comparisons—Tukey); * indicates a significant difference relative to the other cell lines.
Figure 2. IC50 values of β-lapachone in four neoplastic cell lines. Statistical analysis was performed using ANOVA parametric test (multiple comparisons—Tukey); * indicates a significant difference relative to the other cell lines.
Molecules 29 01395 g002
Figure 3. Apoptosis in neoplastic cells treated with β-lapachone for 24 h. (A) The cell lines were exposed to different concentrations of β-lapachone and the apoptotic index was determined. (B) The percentage of apoptotic cells seen at the highest concentration tested (1.5 µg/mL) was compared across the cell lines. NC: negative control; PC: positive control. Statistical analysis was performed using ANOVA parametric test (multiple comparisons—Tukey); * indicates a significant difference with respect to the NC, 0.5 µg/mL, and 1 µg/mL groups.
Figure 3. Apoptosis in neoplastic cells treated with β-lapachone for 24 h. (A) The cell lines were exposed to different concentrations of β-lapachone and the apoptotic index was determined. (B) The percentage of apoptotic cells seen at the highest concentration tested (1.5 µg/mL) was compared across the cell lines. NC: negative control; PC: positive control. Statistical analysis was performed using ANOVA parametric test (multiple comparisons—Tukey); * indicates a significant difference with respect to the NC, 0.5 µg/mL, and 1 µg/mL groups.
Molecules 29 01395 g003
Figure 4. DNA damage index in HepG2 cells treated for 3 h with various doses of β-lapachone. NC: negative control; PC: positive control. Statistical analysis was performed using ANOVA parametric test (multiple comparisons—Tukey); * indicates a significant difference relative to the NC.
Figure 4. DNA damage index in HepG2 cells treated for 3 h with various doses of β-lapachone. NC: negative control; PC: positive control. Statistical analysis was performed using ANOVA parametric test (multiple comparisons—Tukey); * indicates a significant difference relative to the NC.
Molecules 29 01395 g004
Figure 5. Micronucleus test results for cells exposed for 24 h to different concentrations of β-lapachone and control treatments. (A) Cytokinesis block proliferation index (CBPI). (B) Frequency of micronuclei (MN). (C) Frequency of nuclear abnormalities (NAs). NC: negative control; PC: positive control. Statistical analysis was performed using ANOVA parametric test (multiple comparisons—Tukey); a indicates a significant difference relative to the NC group; b indicates a significant difference relative to all other treatments.
Figure 5. Micronucleus test results for cells exposed for 24 h to different concentrations of β-lapachone and control treatments. (A) Cytokinesis block proliferation index (CBPI). (B) Frequency of micronuclei (MN). (C) Frequency of nuclear abnormalities (NAs). NC: negative control; PC: positive control. Statistical analysis was performed using ANOVA parametric test (multiple comparisons—Tukey); a indicates a significant difference relative to the NC group; b indicates a significant difference relative to all other treatments.
Molecules 29 01395 g005
Figure 6. Parameters analyzed in the micronucleus test of cells exposed for 24 h to β-lapachone. (A) Binucleated cell without any change (negative control cells). (B) Binucleated cell with micronucleus (arrow) from the positive control group. (C) Cell treated with 0.5 µg/mL of β-lapachone, exhibiting a nuclear bud (arrow). (D) Cell treated with 1 µg/mL of β-lapachone, exhibiting a nucleoplasmic bridge (arrow).
Figure 6. Parameters analyzed in the micronucleus test of cells exposed for 24 h to β-lapachone. (A) Binucleated cell without any change (negative control cells). (B) Binucleated cell with micronucleus (arrow) from the positive control group. (C) Cell treated with 0.5 µg/mL of β-lapachone, exhibiting a nuclear bud (arrow). (D) Cell treated with 1 µg/mL of β-lapachone, exhibiting a nucleoplasmic bridge (arrow).
Molecules 29 01395 g006
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lima, K.M.M.; Calandrini de Azevedo, L.F.; Rissino, J.D.; Vale, V.V.; Costa, E.V.S.; Dolabela, M.F.; Nagamachi, C.Y.; Pieczarka, J.C. Anticancer Potential and Safety Profile of β-Lapachone In Vitro. Molecules 2024, 29, 1395. https://doi.org/10.3390/molecules29061395

AMA Style

Lima KMM, Calandrini de Azevedo LF, Rissino JD, Vale VV, Costa EVS, Dolabela MF, Nagamachi CY, Pieczarka JC. Anticancer Potential and Safety Profile of β-Lapachone In Vitro. Molecules. 2024; 29(6):1395. https://doi.org/10.3390/molecules29061395

Chicago/Turabian Style

Lima, Karina Motta Melo, Luana França Calandrini de Azevedo, Jorge Dores Rissino, Valdicley Vieira Vale, Erica Vanessa Souza Costa, Maria Fani Dolabela, Cleusa Yoshiko Nagamachi, and Julio Cesar Pieczarka. 2024. "Anticancer Potential and Safety Profile of β-Lapachone In Vitro" Molecules 29, no. 6: 1395. https://doi.org/10.3390/molecules29061395

Article Metrics

Back to TopTop