Journal of Leukemia

Journal of Leukemia
Open Access

ISSN: 2329-6917

+44 1300 500008

Research Article - (2014) Volume 2, Issue 4

Combinatorial Efficacy of Nanoliposomal Ceramide and the Antioxidant 7,8- Benzoflavone for Acute Myeloid Leukemia

Brian M. Barth1,2,3*, Timothy J. Brown1,2, Matthew T. Adams1,2, Aileen M. Garcia3,4, Lindsey N. Fisher1,2, Jennifer5, Adam J. Beck5, Colin M. McGill6, Mark Kester2,3,7, Melissa A. Tran5 and David F. Claxton1,2
1Division of Hematology and Oncology, Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
2Penn State Hershey Cancer Institute, Penn State College of Medicine, Hershey, PA, USA
3Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
4Department of Biomedical Science, The Inter American University of Puerto Rico, Ponce, PR, USA
5Drug Discovery, Development, and Delivery Core, Penn State College of Medicine, Hershey, PA, USA
6Department of Chemistry, University of Alaska-Anchorage, Anchorage, AK, USA
7University of Virginia Cancer Center, Charlottesville, VA, USA
*Corresponding Author: Brian M. Barth, Division of Hematology and Oncology, Department of Medicine, Penn State Hershey Cancer Institute, Penn State College of Medicine, 500 University Drive, PO Box 850, CH46 Hershey, PA 17033, USA, Tel: 717-531-0003 (289457) Email:

Abstract

Ceramide-based therapeutics have gained recent attention as anti-neoplastic therapeutics. These include standard of care therapeutics that in part exert efficacy through the generation of ceramide, as well as new therapeutics that seek to specifically deliver or augment ceramide levels in malignant cells. Ceramide is a bioactive sphingolipid involved in apoptotic and stress cellular signaling pathways. It has also been shown to regulate oxidative stress, which may negate its otherwise anti-neoplastic effects by promoting the proliferation of leukemia cells. Metabolism of ceramide to neutral or pro-oncogenic metabolites can serve as a further pathway of therapeutic resistance. In this study, the antioxidant 7,8-benzoflavone (BF) was identified through a natural products chemical library screening process as a compound that can augment the efficacy of nanoliposomal C6-ceramide (Lip-C6) in cellular models of Acute Myeloid Leukemia (AML). This study demonstrates that BF exerts an antioxidant effect in AML, which likely refines the bioactivity of ceramide as an anti-leukemic agent. Intriguingly, BF has been shown to block drug efflux pumps, such as P-glycoprotein, allowing BF to also impede P-glycoprotein-mediated ceramide glycosylation. In this study, BF was further formulated into nanoliposomes for in vivo studies using two murine models of AML. Treatment of C3H/HeJ mice engrafted with a FLT3-ITD driven AML with a combinatorial nanoliposomal formulation of BF and Lip-C6 significantly augmented the survival of mice beyond that of nanoliposomal formulations containing either agent alone. This was in contrast to the modest extension of survival of C57BL/6J mice engrafted with C1498 AML cells utilizing either single agent or combinatorial nanoliposomal formulations. Altogether this study demonstrates that the anti-AML efficacy of Lip-C6 as a ceramide-based therapeutic can be augmented for particular types of AML, such as that driven by FLT3-ITD, by combinatorial treatment with the antioxidant BF.

Keywords: Acute myeloid leukemia; Ceramide; 7,8-benzoflavone; Antioxidant; Nanoliposome

Introduction

Flavonoids are a natural plant product ubiquitous in nature whose potential benefits in medicine have been known and extensively studied [1]. 7,8-Benzoflavone (BF) is a synthetic flavonoid that has previously been implicated in aromatase inhibition, breast cancer resistant protein inhibition, and aryl hydrocarbon receptor signaling [1-4]. Flavones have been investigated as chemopreventive agents due to their ability to scavenge reactive oxygen species, either produced de novo or by carcinogens, and have been shown to be antiproliferative in vitro [5,6].

Over-production of Reactive Oxygen Species (ROS) can alter the redox environment of the cell and have consequences on growth regulation. In particular, oxidation of a cysteine residue in the catalytic center of protein tyrosine phosphatase prevents removal of phosphate groups on receptor tyrosine kinase (RTK) target molecules [7,8]. This, in turn, removes an important regulatory point and constitutive activation of these molecules, promoting a pro-mitogenic cellular environment [7,8]. Theoretically, alteration of the redox state of cells could prevent unregulated growth of cells. Of particular interest to this study is the balance between ROS accumulation and the propagation and survival of Acute Myeloid Leukemia (AML) cells in vitro through modification of tyrosine phosphatases. Several studies have linked increases in ROS to both hematopoietic stem cell proliferation and the proliferation of AML cells [9-12]. AML is a cancer of myeloid precursor cells that results in proliferation of immature myeloblasts and can often follow a rapid clinical course [13-16]. Several cytogenetic abnormalities are responsible for the development of AML and many have been clinically categorized by prognosis [15,16]. While knowledge of the molecular basis of AML has grown substantially in the past 30 years, development and implementation of new therapeutics has been somewhat stagnant [15,16].

Ceramide is a natural sphingolipid that can be produced de novo in cells or formed from a variety of metabolites and is known to be extensively involved with apoptosis [17-22]. Furthermore, altered ceramide metabolism and efflux has been linked to cellular resistance to apoptosis and is frequently found in cancer cells as a mechanism clearly associated with the development of drug resistance [17,19,21,22]. Ceramide and its metabolite, sphingosine-1-phosphate, strike a delicate intracellular balance between ceramide – induced apoptosis and sphingosine-1-phosphate-influenced cell survival and proliferation [18-20]. Another ceramide metabolite, glucosylceramide, has been linked to drug-resistant breast cancer [19-21]. Interestingly, the glycosylation of ceramide species occurs at the Golgi membrane by the coordinated activities of glucosylceramide synthase and P-glycoprotein [19,21]. Importantly, BF may behave as an inhibitor of P-glycoprotein, a drug efflux pump implicated in conferring drug resistance in multiple tumor types [23]. The use of BF has been shown to re-sensitize tumors to chemotherapies [23]. Liposomal formulations of ceramide and ceramide metabolites have been extensively studied in our lab [19,20,24,25]. Due to its apoptosis-inducing properties, cellular ceramide accumulation is an ideal pharmacologic target. The liposomal formulation of ceramide allows for stabilization of the otherwise insoluble ceramide as well as enhanced ceramide delivery to the cells [19]. Recently we demonstrated that blocking ceramide metabolism could induce apoptosis or autophagy in leukemia cells [20].

In the present study, we used a chemical library screen to identify BF as a compound that augments the anti-AML efficacy of nanoliposomal C6-ceramide (Lip-C6). We further showed that BF and Lip-C6 act synergistically to decrease the viability of multiple AML cell lines and that BF exerts an antioxidant effect independent of, and complementary to, the effect of ceramide. Finally, this study demonstrated that the combination of Lip-C6 with BF can significantly increase the survival of mice engrafted with AML driven by FLT3-ITD.

Materials and Methods

Cell culture

Human HL-60, HL-60/VCR, 32D-FLT3-ITD, and murine C1498 cells, were maintained at 37°C, and 5% CO2, in RPMI-1640 supplemented with 10% Fetal Bovine Serum (FBS) and 1% penicillin/streptomycin.

Nanoliposome formulation

Nanoliposomes were prepared by the Penn State College of Medicine Drug Discovery Core following previously established methods with minor changes for the nanoliposomal BF (Lip-BF) formulation. All lipids were obtained from Avanti Polar Lipids (Alabaster, AL, USA). Ghost nanoliposomes (Lip-Ghost) and Lip-C6 were prepared as previously described [20,24,25]. Briefly, lipids dissolved in chloroform, or other organic solvents, were combined in specific molar ratios. For Lip-BF, aliquots of DSPC (1,2-distearoyl-sn-glycero-3-phosphocholine), DOPE (1,2-dioleoyl-sn-glycero-3-phosphoethanolamine), DSPE-PEG2000 (1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000]), and BF, were made in a 5.66:2.87:1.47:0.05 molar ratio. For Lip-C6 containing BF (Lip-C6/BF) the same molar equivalent of BF in Lip-BF was added to the Lip-C6 formulation. Solutions were dried to a film under a stream of nitrogen, and then hydrated by addition of 0.9% NaCl. Solutions were sealed, heated at 60°C (60 min), and subjected to vortex mixing and sonicated until light no longer diffracted through the suspension. The lipid vesicle-containing solution was quickly extruded at 60°C by passing the solution 10 times through 100 nm polycarbonate filters in an Avanti Mini-Extruder. Nanoliposomal size and integrity was determined using a Malvern Zetasizer Nano ZS at 25°C. Nanoliposome formulaitons were stored at room temperature until use.

Cellular viability assays

Human HL-60, HL-60/VCR, 32D-FLT3-ITD, and murine C1498 cells, were plated at 2.5x104 cells per well in 96-well tissue culture plates and treated for 48 h. Following treatment, cellular viability was assessed using a Cell Titer 96 AQueous Non-Radioactive Cell Proliferation Assay according to the manufacturer’s instructions (Promega, Madison, WI, USA). Viability was determined by measuring absorbance at 490 nm using a microplate reader and normalizing to the viability observed under control conditions. CalcuSyn Software (Biosoft, Cambridge, UK) was used to determine combinatorial effects of treatments [24]. Cellular viability data was used for this analysis, and a Combination Index (CI) less than or equal to 0.9 was considered synergistic. CI values greater than or equal to 1.1 were considered antagonistic, whereas CI values between 0.9 and 1.1 were considered additive.

TimTec natural products chemical library screen

Cellular viability assays evaluating HL-60/VCR cells were used to screen the Tim Tec natural products chemical library (TimTec, Newark, DE, USA) for compounds that augmented the efficacy of Lip-C6. A screen of 480 different compounds was performed (10 µM per compound) in combination with Lip-C6 (10 µM) and compared to the Lip-C6 alone. Hits were determined as those compounds that augmented the anti-AML efficacy of Lip-C6 beyond three standard deviations from the mean according to Z-score analysis (3/480 = 0.625% hit rate).

ROS assay

HL-60/VCR cells were treated for 24 h prior to addition of 2 µM of the redox-sensitive indicator H2-dichlorofluorescein diacetate, which was added directly to the culture media 30 min prior to analysis. Upon oxidation, dichlorofuorescein (DCF) fluorescence was indicative of the generation of ROS, and was evaluated at the Penn State College of Medicine Flow Cytometry Core using a LSR II flow cytometer and BD FACS Diva software.

In Vivo studies

All procedures were approved by the Penn State College of Medicine Institutional Animal Care and Use Committee. C57BL/6J mice were engrafted by retro-orbital injection with 1x106 C1498 cells and C3H/HeJ mice were engrafted by retro-orbital injection with 2.5x106 32D-FLT3-ITD cells. Mice were treated with Lip-Ghost, Lip-C6, Lip-BF, or Lip-C6/BF (0.1 mL i.p. injections of 25 mg/mL liposomal formulations) three times per week for four weeks or until they became moribund and were euthanized.

Results and Discussion

We screened the TimTec natural products chemical library (480 molecules) to identify compounds that enhance the anti-AML efficacy of Lip-C6. Using the HL-60/VCR human AML cell line three flavone compounds were identified including glabranine, 6,7-dimethoxyflavone, and BF (Figure 1). Overall, there was a positive hit rate of 0.625% with a positive result identified as being three standard deviations beyond the mean in a Z-score analysis. In particular, we observed BF exerting its effects due in part to its ability to act as an antioxidant. Current literature proposes that antioxidants such as BF prevent oxidation of cysteine residues in the catalytic center on tyrosine phosphatases that can lead to unregulated proliferation by constitutive activation of RTK second messengers [7,8]. Additionally, BF may behave as an inhibitor of drug efflux pumps such as P-glycoprotein [1,23]. In addition to traditional roles eliminating cytotoxic agents from cells, drug efflux pumps such as P-glycoprotein have also been shown to participate in the metabolism of ceramide to glucosylceramide by glycosylation at the Golgi membrane [19-21]. Therefore, in addition to an antioxidant role for BF, alteration of the activity of P-glycoprotein may serve to alter the metabolism of ceramide to favor its accumulation. For these reasons, we chose to further evaluate the combination of BF and ceramide as an anti-AML therapeutic strategy.

leukemia-natural-products

Figure 1: Screen of natural products chemical library evaluating those that augment the anti-AML efficacy of nanoliposomal C6- ceramide. Z-score analysis was used to identify hits as those augmenting therapeutic efficacy at least 3 standard deviations from the mean. 3/480 hits were identified (0.625%).

Following the identification of BF from screening the Tim Tec natural products library as a compound that can augment the anti-AML efficacy of Lip-C6, a more thorough analysis of the combinatorial effects was evaluated using a variety of AML cell lines and cellular viability assays (Figure 2). Profound combinatorial efficacy of BF and Lip-C6 was observed in both 32D-FLT3-ITD and HL-60/VCR cell lines, with synergistic efficacy confirmed for the later (CI=0.177). Parental HL-60 cells (not selected by vincristine resistance) and C1498 cells were sensitive to either agent alone but only combinatorial effects were modest and/or not dose dependent. For the more substantial combinatorial effects observed with 32D-FLT3-ITD and HL-60/VCR cells the combination promotes an anti-AML effect at lower doses than either treatment alone. It is possible that C1498 cells do not show this same combinatorial effect due to differential expression of drug efflux pumps [20], and therefore less sensitivity to BF antagonism of ceramide glycosylation, or because they do not generate and benefit from a profound pro-oxidant state. In further support of the notion that BF exerts its combinatorial anti-AML efficacy with Lip-C6 by acting as an antioxidant, by using an ROS assay we observed that HL-60/VCR cells existed in a substantial pro-oxidant environment that was specifically down regulated by BF (Figure 3). This was noted both with and without the addition of Lip-C6. By reducing the redox state of the cells, BF may prevent cysteine oxidation on protein tyrosine phosphatases which otherwise may lead to unregulated growth and proliferation [7,8]. It is known that ceramide induces oxidative stress within the cell. Therefore, the addition of BF may relieve this pro-oxidant effect which would refine the effect of ceramide to be more specifically directed towards inducing apoptosis of the malignant cells. More so, BF may be a better candidate to pair with Lip-C6 than other antioxidants, such as vitamin E, given BF’s potential role in blocking ceramide glycosylation by impeding P-glycoprotein [19-21].

leukemia-cell-lines

Figure 2: AML cell lines were evaluated for therapeutic sensitivity to nanoliposomal C6-ceramide (Lip-C6) and 7,8-benzoflavone (BF) using cellular viability assays. (A) BF anti-AML efficacy was evaluated using murine C1498 and 32D-FLT3-ITD cells as well as human HL-60 and HL-60/VCR cells. (B) Lip-C6 anti-AML efficacy was evaluated using murine C1498 and 32D-FLT3-ITD cells as well as human HL-60 and HL-60/VCR cells. (C) The combination of Lip-C6 and BF was evaluated using HL-60/VCR cells by evaluating a range of Lip-C6 concentrations while holding BF at constant concentration. (D) Isobologram depicting the synergistic combinatorial index (CI) observed for Lip-C6 and BF using HL-60/VCR cells. The combination of Lip-C6 and BF was evaluated using C1498 (E), 32D-FLT3-ITD (F), HL-60 (G), and HL-60/VCR (H) cells by evaluating a range of BF concentrations while holding Lip-C6 at constant concentration.

leukemia-antioxidant-effect

Figure 3: 7,8,-Benzoflavone (BF) exerted an antioxidant effect in HL-60/VCR cells, an effect apparent where nanoliposomal C6-ceramide (Lip-C6) and BF exert synergistic efficacy. Following treatment, cells were loaded with a redox-sensitive indicator and analyzed by flow cytometry. Untreated control (A), DMSO vehicle control (B), empty “ghost” nanoliposomal (Lip-Ghost) control (C), BF treatment (D), Lip- C6 treatment (E), and treatment with the combination of Lip-C6 and BF (F).

A murine study further reinforced the in vitro findings. First, to improve its in vivo delivery BF was formulated into nanoliposomes with or without C6-ceramide (Figure 4a). The anti-AML efficacy of Lip-BF compared with free BF using the 32D-FLT3-ITD cell lines, and was found to have nearly equivalent efficacy (Figure 4b). C57BL/6J mice engrafted with C1498 cells (Figure 4d), and C3H/HeJ mice engrafted with 32D-FLT3-ITD cells (Figure 4e), were then treated with Lip-Ghost, Lip-C6, Lip-BF, or the combinatorial Lip-C6/BF. Survival curves from mice engrafted with 32D-FLT3-ITD cells demonstrated enhanced survival with the combinatorial liposomal formulation than compared to control and either treatment alone. However, the mice engrafted with C1498 cells showed no enhanced survival by the combinatorial formulation compared to single agent treatment. These in vivo studies confirmed in vitro observations, suggesting that the combination of BF with Lip-C6 may only work in the scenarios where the leukemia has a profound pro-oxidant state by offsetting the additional and counterproductive pro-oxidant actions of Lip-C6. This was evidenced in our study by the substantial increase in survival of C3H/HeJ mice engrafted with 32D-FLT3-ITD cells, with 60% of the mice surviving long term when treated with the combinatorial Lip-C6/BF formulation. Overall, this study identified the antioxidant BF as a compound that can effectively be combined with Lip-C6 for the treatment of certain AMLs with enhanced oxidative states such as those harboring mutations of FLT3 [12]. The further development of liposomal formulations of BF, and other antioxidants, may hold a promising future for the treatment of aggressive AML.

leukemia-Nanoliposomal-formulation

Figure 4: Nanoliposomal formulation of 7,8-benzoflavone (BF) and in vivo evaluation of anti-AML efficacy with nanoliposomal ceramide (Lip-C6). (A) Light scatter analysis confirmed nanoliposomal BF (Lip-BF) nanosize and stability. (B) Cellular viability assays confirmed that Lip-BF and free/unencapsulated BF exerted similar therapeutic efficacy using 32D-FLT3-ITD cells. (C) Light scatter analysis confirmed combinatorial nanoliposomal C6- ceramide and BF (Lip-C6/BF) nanosize and stability. (D-E) Survival was evaluated using two murine models treated with liposomal formulations. Combinatorial extension in survival was only observed in 32D-FLT3-ITD-engrafted C3H/HeJ mice, but not C1498-engrafted C57BL/6J mice. This confirmed in vitro studies which showed that 32D-FLT3-ITD cells were synergistically sensitive to Lip-C6 and BF, but C1498 cells were not.

Acknowledgements

This study was funded in part by the Penn State University Kiesendahl Family Endowed Leukemia Research Fund, the Kenneth Noel Memorial Fund, NIH grant CA171983 (B.M.B., M.K., and D.F.C.), and PA Tobacco Settlement funds. The Penn State Research Foundation has licensed ceramide nanoliposomes, and other nonliposomal technology, to Keystone Nano, Inc. (State College, PA). M.K. is Chief Medical Officer of Keystone Nano, Inc.

References

  1. Zhang S, Yang X, Coburn RA, Morris ME (2005) Structure activity relationships and quantitative structure activity relationships for the flavonoid-mediated inhibition of breast cancer resistance protein. BiochemPharmacol 70: 627-639.
  2. Kellis JT Jr, Vickery LE (1984) Inhibition of human estrogensynthetase (aromatase) by flavones. Science 225: 1032-1034.
  3. Fukazawa H, Suzuki T, Wakita T, Murakami Y (2012) A cell-based, microplate colorimetric screen identifies 7,8-benzoflavone and green tea gallatecatechins as inhibitors of the hepatitis C virus. Biol Pharm Bull 35: 1320-1327.
  4. Fischer SM, Reiners JJ Jr (1986) 7,8-Benzoflavone: an inhibitor of prostaglandin synthesis and ornithine decarboxylase in murine epidermal cultures. Carcinogenesis 7: 933-935.
  5. Kuntz S, Wenzel U, Daniel H (1999) Comparative analysis of the effects of flavonoids on proliferation, cytotoxicity, and apoptosis in human colon cancer cell lines. Eur J Nutr 38: 133-142.
  6. Zhang Q, Zhao XH, Wang ZJ (2008) Flavones and flavonols exert cytotoxic effects on a human oesophageal adenocarcinoma cell line (OE33) by causing G2/M arrest and inducing apoptosis. Food ChemToxicol 46: 2042-2053.
  7. Wall SB, Oh JY, Diers AR, Landar A (2012) Oxidative modification of proteins: an emerging mechanism of cell signaling. Front Physiol 3: 369.
  8. Truong TH, Carroll KS (2012) Redox regulation of epidermal growth factor receptor signaling through cysteine oxidation. Biochemistry 51: 9954-9965.
  9. Sardina JL, López-Ruano G, Sánchez-Sánchez B, Llanillo M, Hernández-Hernández A (2012) Reactive oxygen species: are they important for haematopoiesis? Crit Rev OncolHematol 81: 257-274.
  10. Toyokuni S, Okamoto K, Yodoi J, Hiai H (1995) Persistent oxidative stress in cancer. FEBS Lett 358: 1-3.
  11. Sallmyr A, Fan J, Rassool FV (2008) Genomic instability in myeloid malignancies: increased reactive oxygen species (ROS), DNA double strand breaks (DSBs) and error-prone repair. Cancer Lett 270: 1-9.
  12. Sallmyr A, Fan J, Datta K, Kim KT, Grosu D, et al. (2008) Internal tandem duplication of FLT3 (FLT3/ITD) induces increased ROS production, DNA damage, and misrepair: implications for poor prognosis in AML. Blood 111: 3173-3182.
  13. Bar-Natan M, Nelson EA, Xiang M, Frank DA (2012) STAT signaling in the pathogenesis and treatment of myeloid malignancies. JAKSTAT 1: 55-64.
  14. Levine RL (2012) JAK-mutant myeloproliferative neoplasms. Curr Top MicrobiolImmunol 355: 119-133.
  15. Patel JP, Gönen M, Figueroa ME, Fernandez H, Sun Z, et al. (2012) Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med 366: 1079-1089.
  16. Rai KR, Holland JF, Glidewell OJ, Weinberg V, Brunner K, et al. (1981) Treatment of acute myelocyticleukemia: a study by cancer and leukemia group B. Blood 58: 1203-1212.
  17. Senchenkov A, Litvak DA, Cabot MC (2001) Targeting ceramide metabolism--a strategy for overcoming drug resistance. J Natl Cancer Inst 93: 347-357.
  18. Van Brocklyn JR, Williams JB (2012) The control of the balance between ceramide and sphingosine-1-phosphate by sphingosinekinase: oxidative stress and the seesaw of cell survival and death. Comp BiochemPhysiol B Biochem Mol Biol 163: 26-36.
  19. Barth BM, Cabot MC, Kester M (2011) Ceramide-based therapeutics for the treatment of cancer. Anticancer Agents Med Chem 11: 911-919.
  20. Brown TJ, Garcia AM, Kissinger LN (2013) Therapeutic Combination of NanoliposomalSafingol and NanoliposomalCeramide for Acute Myeloid Leukemia: Journal of Leukemia1: 110.
  21. Gouazé V, Yu JY, Bleicher RJ, Han TY, Liu YY, et al. (2004) Overexpression of glucosylceramide synthase and P-glycoprotein in cancer cells selected for resistance to natural product chemotherapy. Mol Cancer Ther 3: 633-639.
  22. Kester M, Kolesnick R (2003) Sphingolipids as therapeutics. Pharmacol Res 47: 365-371.
  23. Morris ME, Zhang S (2006) Flavonoid-drug interactions: effects of flavonoids on ABC transporters. Life Sci 78: 2116-2130.
  24. Jiang Y, DiVittore NA, Kaiser JM, Shanmugavelandy SS, Fritz JL, et al. (2011) Combinatorial therapies improve the therapeutic efficacy of nanoliposomalceramide for pancreatic cancer. Cancer BiolTher 12: 574-585.
  25. Morad SA, Levin JC, Shanmugavelandy SS (2012) Ceramide--antiestrogennanoliposomal combinations--novel impact of hormonal therapy in hormone-insensitive breast cancer. Mol Cancer Ther 11: 2352-2361.
Citation: Barth BM, Keasey NR, Wang X, Shanmugavelandy SS, Rampal R, et al. (2015) Combinatorial Efficacy of Nanoliposomal Ceramide and the Antioxidant 7,8-Benzoflavone for Acute Myeloid Leukemia. J Leuk (Los Angel) 2:152.

Copyright: © 2014 Barth BM et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top