Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Anticancer Agent Shikonin Is an Incompetent Inducer of Cancer Drug Resistance

  • Hao Wu,

    Affiliation Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China

  • Jiansheng Xie,

    Affiliation Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China

  • Qiangrong Pan,

    Affiliation Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China

  • Beibei Wang,

    Affiliation Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China

  • Danqing Hu,

    Affiliation Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China

  • Xun Hu

    huxun@zju.edu.cn

    Affiliation Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China

Abstract

Purpose

Cancer drug resistance is a major obstacle for the success of chemotherapy. Since most clinical anticancer drugs could induce drug resistance, it is desired to develop candidate drugs that are highly efficacious but incompetent to induce drug resistance. Numerous previous studies have proven that shikonin and its analogs not only are highly tumoricidal but also can bypass drug-transporter and apoptotic defect mediated drug resistance. The purpose of this study is to investigate if or not shikonin is a weak inducer of cancer drug resistance.

Experimental Design

Different cell lines (K562, MCF-7, and a MDR cell line K562/Adr), after repeatedly treated with shikonin for 18 months, were assayed for drug resistance and gene expression profiling.

Results

After 18-month treatment, cells only developed a mere 2-fold resistance to shikonin and a marginal resistance to cisplatin and paclitaxel, without cross resistance to shikonin analogs and other anticancer agents. Gene expression profiles demonstrated that cancer cells did strongly respond to shikonin treatment but failed to effectively mobilize drug resistant machineries. Shikonin-induced weak resistance was associated with the up-regulation of βII-tubulin, which physically interacted with shikonin.

Conclusion

Taken together, apart from potent anticancer activity, shikonin and its analogs are weak inducers of cancer drug resistance and can circumvent cancer drug resistance. These merits make shikonin and its analogs potential candidates for cancer therapy with advantages of avoiding induction of drug resistance and bypassing existing drug resistance.

Introduction

Cancer drug resistance is one of the major obstacles significantly interfering with the efficacy of cancer chemotherapy. Cellular factors that contribute to drug resistance include: (1) drug transporter-mediated increased efflux and decreased influx of anticancer drugs, (2) activation of DNA repair, (3) activation of detoxification system, and (4) blocked apoptosis [1], [2], [3], [4], [5], [6]. All these problems arise as a result of cancer cells' adapting to chemotherapeutic agents, i.e., the former have the capacity to attenuate the stimulation from the latter. Thus, in order to solve the problem, anticancer drugs that are toxic toward cancer cells but incompetent to induce drug resistance are desired.

Shikonin is a naturally occurring naphthoquinone compound, and the main component of red pigment extracts from Lithospermiun erythrorhizon Sieb et Zucc of East Asia. Shikonin and its analogues are potential pharmaceutical agents with anticancer activities well documented. Shikonin and its analogues could kill cancer cells via inhibiting topoisomerase-I [7], [8], [9], polo-like kinase 1 (PLK1) and protein tyrosine kinase (PTK) [10], [11], regulating the activities of phosphorylated extracellular regulated protein kinase (pERK), c-Jun N-terminal kinase (JNK), and protein kinase C-a (PKC-a) [12], suppressing the expression of tumor necrosis factor receptor-associated protein 1 (TRAP1) [13], activating caspase activities[14], [15], inhibiting proteasome [16], among others. Sankawa et al. found that shikonin and a range of simple derivatives completely inhibited tumor growth in mice at a dose of 5–10 mg/kg/day [17], [18]. The LD50 of shikonin and some derivatives to mice by intraperitonal administration ranged from 20 mg/kg to 48 mg/kg [19], [20]. Notably, a clinical study indicated that shikonin mixture was effective in treatment of 19 patients with later-stage lung cancer who were not suitable for surgery, radiotherapy, and chemotherapy [21]. We reported that naturally-occurring shikonin and its analogues (Fig. 1) could circumvent cancer drug resistance mediated by drug transporters P-glycoprotein (P-gp), multidrug resistance-associated protein 1 (MRP1), and brest cancer resistance protein (BCRP1), and by antiapoptotic proteins Bcl-2 and Bcl-xL, by induction of necroptosis [22], [23], [24], [25], [26], a cell death recently defined and studied in depth by Degterev A and Yuan J [27], [28], [29]. Recently, we identified shikonin and its analogues were potent inhibitors to pyruvate kinase isozyme M2 (PKM2) or tumor M2-PK [30], which almost ubiquitously expresses in tumor cells [31] and plays important roles in cancer cell metabolism and growth [32], [33], [34]. Taken together, all these lines of evidence support that shikonin and its analogues are strong anticancer agents. However, the evidence does not mean that shikonin and its analogues are incapable of inducing drug resistance. In this study, we show that shikonin is a weak inducer of cancer drug resistance.

Materials and Methods

Reagents

Shikonin was purchased from National Institute for the Control of Pharmaceutical and Biological Products (Beijing, China) with purity of >99%. Doxorubicin, paclitaxel, vincristine, Methotrexate, cisplatin, dicumarol, and MTT (3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) were purchased from Sigma-Aldrich. Shikonin analogues (Fig. 1) were purchased from the Tokyo Chemical Industry (TCI, Tokyo).

Cell lines

All cell lines were obtained from and characterized by The Cell Bank of Type Culture Collection of Chinese Academy of Sciences according to the cell line authentification testing (vitality, species confirmation and interspecies contamination, DNA fingerprinting and mycoplasma contamination). MCF-7 cell was maintained in DMEM containing 10% fetal bovine serum (FBS). MCF-7/Adr cell was grown in RPMI 1640 containing 10% FBS and 1 µg/mL doxorubicin. K562 was maintained in RPMI 1640 supplemented with 10% FBS. K562/Adr cell was grown in RPMI 1640 supplemented with 10% FBS and 500 ng/ml doxorubicin.

Antibodies

The α-tubulin Rabbit polyclonal antibody was purchased from Cell Signaling Technology, Inc. (Boston, MA). Mouse monoclonal antibodies for α-tubulin, β-tubulin II and β-tubulin were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA). The β-tubulin I, III and IV Mouse monoclonal antibodies were purchased from Sigma-Aldrich. Mouse anti-actin IgG was purchased from Biomedical Technologies Inc (Stoughton, MA). The secondary antibodies used are HRP-conjugated anti-mouse IgG and HRP-conjugated anti-rabbit IgG (Santa Cruz Biotechnology, CA).

Generation of MCF-7/Shk, MCF-7/Shk-dox, K562/Shk, K562/Shk-dox and K562/Adr/Shk cell lines

MCF-7/Shk was derived by repeated exposure of MCF-7 to 4–6 µM shikonin. Briefly, MCF-7 cells (1×106) were incubated with shikonin (4 µM) for 4 hours, then shikonin was removed and cells were incubated in complete DMEM medium. Cells were treated again immediately after cell growth was recovered. The total cycles of treatment were 25 with the time period of 18 months. Similarly, K562/Shk and K562/Adr/Shk were derived by repeated exposure of K562 and MDR cell line K562/Adr to shikonin.

MCF-7/Shk-dox was generated by alternative treatment of shikonin (4 µM) and doxorubicin (250 ng/mL). Briefly, cells were firstly treated with shikonin. After cell growth was recovered, cells were treated with doxorubicin. When cells resumed growth, again cells were treated with shikonin. The time period is 1.5 years with 13 cycles of treatment. Similarly, K562/Shk-dox was developed by alternative treatment of shikonin and doxorubicin.

Cell growth inhibition assay

The drug-related effect of shikonin analogues against drug-sensitive and -resistant cells was determined by MTT assay as described [23]. Cells were seeded into 96-well plates (cultured overnight for adherent cells) and treated with shikonin analogues at serial concentrations. After a 72-hour incubation, 20 µl MTT (5 mg/ml) was added into each well for another 4-hour incubation. After that, the supernatant was removed and 150 µl dimethyl-sulphoxide (DMSO, Sigma) was added into each well in order to solubilize the blue-purple crystals of formazan. The absorbance was then measured using a model ELX800 Micro Plate Reader (Bio-Tek Instruments, Inc.) at 570 nm. The inhibition rate was calculated according to the following formula: Inhibition rate = (Absorbance of control−absorbance of treatment)/Absorbance of control×100% [35].

Western blotting assay

Western Blotting was performed as described by us previously [22], [25]. The protein was applied to a 12% SDS polyacrylamide gel, transferred to a PVDF membrane, and then detected by the proper primary and secondary antibodies before visualization by EZ-ECL (Biological Industries, Israel). Film was scanned and densitometry was determined with Quantity One software (Bio-Rad Laboratories, CA).

Identification of interaction between shikonin and βII-tubulin using solid-phase shikonin extraction combined with Western Blot

Solid phase shikonin was prepared according to our previous reported method [36]. The procedure to determine the interaction between βII-tubulin and solid-phase shikonin is as follows. MCF-7 cells were lysed at 4°C for 0.5 hour with M-PER Mammalian Protein Extraction Reagent (Pierce biotechnology, Rockford, IL) plus Halt Protease Inhibitor Cocktail (Pierce biotechnology, Rockford, IL), followed by centrifugation at 13,000 rpm at 4°C for 15 minutes. The clear supernatant was collected and the pellet was discarded. The total protein concentration in the supernatant was determined using a BCA protein quantification kit (Pierce biotechnology, Rockford, IL). 4 mg protein was mixed with 100 µl of shikonin-conjugated Epoxy-activated Sepharose-6B (GE Healthcare, Sweden) to a total volume of 300 µl. The mixture was incubated for 6 h at 4°C with constant and gentle stirring. The mixture was spun down at 5000 r.p.m. for 5 min at 4°C. The supernatant was discarded and the precipitate was washed by chilled washing buffer (50 mM Tris–HCl (pH 7.0), 500 mM NaCl, 10% glycerol, 1 mM DTT, 1 mM phenylmethylsulfonyl fluoride) with or without 1 mM Shikonin by centrifugation (5000 r.p.m. for 5 min at 4°C) for five times. The precipitate was mixed with SDS–PAGE sample buffer (Pierce biotechnology, Rockford, IL) and boiled for 10 min. The samples were subjected to immunoblotting analysis for βII-tubulin.

Preparation of RNA for gene arrays

Cells were trypsinized, and total RNA was extracted using RNeasy kit (Qiagen, Germany). The quality of the total RNA was verified using a BioAnalyzer (Agilent, Santa Clara, CA). Sample preparation, labeling and hybridization to Human Genome U133 Plus 2.0 Arrays (Affymetrix, Santa Clara, CA) were performed according to manufacturer's protocols at ShanghaiBio Corporation (Shanghai, China). Raw and processed data have been deposited in Gene Expression Omnibus (GSE34298).

Microarray data analysis

We used the Affymetrix Human U133 Plus 2.0 microarray, which interrogates over 47,000 transcripts, with an average of 27 probes per gene. Affymetrix raw data files [cell intensity (CEL) files] were first analyzed with Robust multi-array Average (RMA) normalization as implemented in the Affymetrix Expression Console Software (version 1.1) to remove between-array effects and to standardize the low-level data [37]. In order to detect differentially expressed genes, significance analysis of microarrays (SAM) algorithm [38] was used to calculate the q-values (false discovery rate) for genes in the indicated time points. SAM was performed with the software tool of The Institute for Genomic Research (TIGR) MeV (http://www.tigr.org/software/tm4/mev.html) [39]. We had three replicates for each cell lines. The list of differentially expressed genes of each indicated time point were obtained by SAM with the fold change ≥1.75, and q-values≤0.0015 compared to control.

Gene Ontology (GO) category enrichment analysis was performed on differentially expressed genes using the publicly available software DAVID (Database for Annotation, Visualization and Integrated Discovery, http://david.abcc.ncifcrf.gov/, Bethesda, MD) with default parameters [40]. The goal of this analysis was to search for GO terms in molecular function, biological process, and cellular component that were significantly enriched in the gene lists obtained above.

Statistical analysis

Unless otherwise stated, data were expressed as the mean ± SD, and analyzed by two-tailed unpaired Student's t test.

Results

1. Time frame for developing cell sublines

Repeated exposure of cancer cells to anticancer drugs would lead to development of cancer drug resistance eventually. Previously, numerous reports demonstrated that the time needed for induction of drug resistance by different drugs usually took days to months (Table S1). In this study, cancer cells were repeatedly treated with shikonin for a time period of 18 months, which was empirically sufficiently long for inducing drug resistance.

2. Shikonin induces a marginal resistance to shikonin, cisplatin, and paclitaxel

In general, the resistance induced by shikonin is weak. After 18-months of treatment with shikonin, MCF-7/Shk and K562/Shk showed a 2-fold resistance to shikonin but no cross-resistance to its analogues. MCF-7/Shk showed a marginal resistance to paclitaxel and cisplatin but without obvious cross-resistance to other conventional anticancer drugs (Table 1 and 2).

thumbnail
Table 1. IC50 profile of all the parental cells and the drug-resistant cells to shikonin analogues.

https://doi.org/10.1371/journal.pone.0052706.t001

thumbnail
Table 2. IC50 profile of all the parental cells and the drug-resistant cells to conventional anticancer drugs.

https://doi.org/10.1371/journal.pone.0052706.t002

Similarly, K562/Shk only exhibited a marginal resistance to shikonin and cisplatin, without cross-resistance to other shikonin analogues and other conventional anticancer drugs (Table 1 and 2).

Overall, these results demonstrated a weak potency of shikonin in inducing cancer drug resistance.

3. Shikonin induces a global change of gene expression profile

We thought that the failure to induce drug resistance by shikonin could be due to the insufficient interaction between cancer cells and the compound. In order to rule out this possibility, we compared the gene expression profiles of shikonin-treated cells and control cells, which showed a global change of gene expression in shikonin-treated cells (Table S2), which encompasses all of the cellular processes as categorized by GO terms including cell cycle, cell death/survival, metabolism, organelle organization, cell motion, among others (Table S3 and S4). The results demonstrated that cancer cells did respond to shikonin but failed to effectively mobilize drug resistant machineries.

4. Shikonin-induced resistance is not associated with DT diaphroase

Shikonin is structurally similar to vitamin K3. Cancer cells can develop resistance to vitamine K3 via upregulation of DT diaphorase, which can be reversed by dicumarol, an inhibitor to the enzyme [41]. We assayed if DT diaphorase was involved in shikonin-induced drug resistance. The results demonstrated that in the presence or absence of dicumarol, there was no significant change of the sensitivity of MCF-7/Shk toward shikonin, indicating DT diaphorase did not play a major role in shikonin resistance (Fig. 2).

thumbnail
Figure 2. DT diaphorase is not involved in shikonin induced drug resistance.

MCF-7 and MCF-7/Shk cells were treated with shikonin in the presence or absence of 20 µM dicumarol.

https://doi.org/10.1371/journal.pone.0052706.g002

5. Shikonin-induced resistance is associated with βII-tubulin

Since MCF-7/Shk showed a weak cross-resistance to paclitaxel and cisplatin, and since resistance to these 2 agents could be associated with altered expression of tubulin [42], we examined tubulin expression and found that MCF-7/Shk had an increased expression of βII-tubulin (Fig. 3A&B).

thumbnail
Figure 3. Shikonin induced resistance is associated with βII-tubulin.

(A) Determination of cellular tubulins by Western blot. (B) The densitometry of βII-tubulin band in Western blot (Data are mean ± SD, n = 3). (C) Specific binding of βII-tubulin with solid-phase shikonin. MCF-7 and K562 cell lysate was incubated with Sepharose 6B or shikonin-conjugated Sepharose 6B in the absence or presence of free shikonin, followed by thorough wash. The resin is then mixed with loading buffer, boiled, and subjected for Western Blot (for details, see Materials and Methods).

https://doi.org/10.1371/journal.pone.0052706.g003

Then why K562/Shk was not cross-resistant to paclitaxel, since it also showed an upregulated βII-tubulin (Fig. 3A&B). Perhaps, cells from different origins could respond to drugs differentially, e.g., MCF-7 is a breast cancer cell line whose growth depends on attachment, so that the role of tubulin is apparently more important for MCF-7 than for K562, a leukemia cell line whose growth does not depend on attachment.

The results suggested that βII-tubulin was the target by shikonin. To validate it, we used solid-phase shikonin affinity extraction of cellular protein combined with Western Blot. The results showed that βII-tubulin in MCF-7 and K562 cell lysate bound with shikonin-conjugated resin and proved the physical interaction between shikonin and βII-tubulin (Fig. 3C).

The results explained the mechanism of resistance to shikonin, but not the non-cross-resistance to its analogues. Perhaps, shikonin analogues with different R group (Fig. 1) may have lower affinity to βII-tubulin than shikonin, which can make the marginal resistance insignificant.

6. MDR cell line K562/Adr repeatedly exposed to shikonin develops weak resistance to this agent

K562/Adr is a cell line developed by repeated exposure of K562 to doxorubicin [43]. The cell line is characterized with overexpression of P-gp and cross resistance to anthracycline antibiotics, Vinca alkaloids, taxanes, and epipodophyllotoxins [43], [44], [45], [46], [47]. Although P-gp plays a major role in this cell line's drug resistance, given that doxorubicin kills cancer cells via producing free radicals through quinone redox cycling, inhibiting DNA Topoisomerase, intercalating into DNA helix, etc. [48], this cell line should be presumably equipped with multiple defense lines in addition to P-gp against anticancer drug. Since K562/Adr is much more adapted to drug environment than its parental cell K562, we presumed that K562/Adr could develop stronger and quicker resistance to shikonin and its analogues, if such resistance could be induced. However, after an 18-month treatment, K562/Adr/Shk, like K562/Shk, showed only a 2-fold resistance to shikonin and no cross-resistance to its analogues (Table 1), although K562/Adr/Shk kept a cross-resistance to paclitaxel, doxorubicin, and vincristine, similar to its parental cell (Table 2). These results further strengthen the notion that shikonin is an incompetent inducer of drug resistance.

7. Cells alternatively treated with shikonin and doxorubicin develop resistance to doxorubicin, vincristine, and paclitaxel, but not shikonin and its analogues

There are 2 possible consequences of drug-resistance induction by combined treatment of cells with shikonin and doxorubicin. First, since shikonin is incompetent to induce drug resistance, adding doxorubicin (a strong inducer of drug resistance) into drug resistance-inducing procedure would possibly help cells to acquire stronger resistance to shikonin. Conversely, since shikonin and doxorubicin kill cancer cells with distinct mechanism, the combined treatment with these 2 drugs may reciprocally reduce the magnitude of resistance.

For an 18-month alternative exposure to shikonin and doxorubicin, MCF-7/Shk-dox and K562/Shk-dox showed a cross resistance to paclitaxel, doxorubicin, and vincristine, but not to shikonin and its analogues.

The resistance to paclitaxel, doxorubicin, and vincristine was apparently induced by doxorubicin, because shikonin alone did not induce resistance to these drugs (Table 2), whereas doxorubicin had been confirmed to be a strong inducer of drug resistance with multiple mechanisms [48].

Obviously, the magnitude of resistance of MCF-7/Shk-dox and K562/Shk-dox in comparison to MCF-7/Adr and K562/Adr (2 cell lines derived by exposure of cells to doxorubicin only) was orders lower (Table 2). Cells either pulsed with high concentration or maintained in low concentration of doxorubicin alone developed fast and strong resistance [49], [50], [51], [52], [53], which could be mediated by P-gp, MRP1, DNA Topoisomerase II, among others [48], [49], [50], [51], [52], [53]. The results thus suggest that alternative treatment with shikonin and doxorubicin would reciprocally reduce the magnitude of drug resistance.

Discussion

In summary, shikonin and likely its analogues are incompetent inducers of drug resistance. Different cell lines (K562, MCF-7, and MDR cell K562/Adr), treated with shikonin alone only developed about a 2-fold resistance to shikonin, cisplatin, and paclitaxel, without cross resistance to other shikonin analogues and anticancer drugs. Shikonin-induced resistance was associated with the up-regulation of βII-tubulin, which was a target by shikonin. Combined with our previous reports that shikonin and its analogues could circumvent drug transport and apoptosis associated cancer drug resistance [23], [24], [25], [26], [54], this class of compound deserves further attention.

The drug resistance induced by shikonin is insignificant, if known anticancer agents are used as references. In general, there are two major criteria to evaluate the capacity of a drug to induce cancer drug resistance, the time needed for the occurrence of resistance after exposure to the drug and the magnitude of resistance to the drug and cross resistance to other structurally and functionally similar or irrelevant drugs. This is routinely evaluated by exposure of cancer cell lines to tested drugs. It has been well documented that strong drug resistance can be induced by anticancer agents covering categories of platinum, Vinca alkaloids, anthracyclines, taxanes, antimetabolic agents, alkylating agents, topoisomerase inhibitors, tyrosine kinase inhibitors, and retinoids (Table S1).

One critical point is why shikonin and its analogues are incompetent inducer of cancer drug resistance. The ‘nasty’ nature of shikonin – targeting multiple important molecules – is probably a key to this solution. It has been shown that shikonin and its analogues targeted a plethora of important molecules including topoisomerase-I [7], [8], [9], PLK1, PTK [10], [11], pERK, JNK, and PKC-a [12], TRAP1 [13], caspases [14], [15], proteasome [16], PKM2 [30], among others. The broad-spectrum activity of shikonin may ‘confuse’ cancer cells to properly respond to the stimulation. Transcript profiling study provided indirect evidence to this proposed notion (Tables S2, S3, S4). Shikonin treatment did cause a global change of gene expression, but the change obviously did not contribute to the resistance, indicating that cancer cells were possibly ‘confused’ to make a decision how to properly mobilize defense machineries to the stimulation.

In summary, shikonin and its analogues have 3 merits, i.e., they are strong anticancer agents, weak inducer of cancer drug resistance, and can circumvent drug transporter and apoptotic defect mediated cancer drug resistance. The merits suggest the potential candidacy of this class of chemicals for cancer treatment.

Supporting Information

Table S1.

Anticancer agents induce strong drug resistance.

https://doi.org/10.1371/journal.pone.0052706.s001

(PDF)

Table S2.

The list of significantly regulated genes in K562/Shk cell. Cells were treated with shikonin for 18 months, control cells were treated with vehicle as described in Materials and Methods.

https://doi.org/10.1371/journal.pone.0052706.s002

(PDF)

Table S3.

The common gene ontology terms of up regulated genes in K562/Shk cells. Cells were treated with shikonin for 18 months, control cells were treated with vehicle as described in Materials and Methods.

https://doi.org/10.1371/journal.pone.0052706.s003

(PDF)

Table S4.

The common gene ontology terms of down regulated genes in K562/Shk cells. Cells were treated with shikonin for 18 months, control cells were treated with vehicle as described in Materials and Methods.

https://doi.org/10.1371/journal.pone.0052706.s004

(PDF)

Author Contributions

Conceived and designed the experiments: XH HW. Performed the experiments: HW JX QP BW DH. Analyzed the data: HW. Wrote the paper: XH HW.

References

  1. 1. Gottesman MM (2002) Mechanisms of cancer drug resistance. Annu Rev Med 53: 615–627.
  2. 2. Gottesman MM, Fojo T, Bates SE (2002) Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer 2: 48–58.
  3. 3. Relling MV, Dervieux T (2001) Pharmacogenetics and cancer therapy. Nat Rev Cancer 1: 99–108.
  4. 4. Helleday T, Petermann E, Lundin C, Hodgson B, Sharma RA (2008) DNA repair pathways as targets for cancer therapy. Nat Rev Cancer 8: 193–204.
  5. 5. Cotter TG (2009) Apoptosis and cancer: the genesis of a research field. Nat Rev Cancer 9: 501–507.
  6. 6. Hoskins JM, Carey LA, McLeod HL (2009) CYP2D6 and tamoxifen: DNA matters in breast cancer. Nat Rev Cancer 9: 576–586.
  7. 7. Ahn BZ, Baik KU, Kweon GR, Lim K, Hwang BD (1995) Acylshikonin analogues: synthesis and inhibition of DNA topoisomerase-I. J Med Chem 38: 1044–1047.
  8. 8. Bailly C (2000) Topoisomerase I poisons and suppressors as anticancer drugs. Curr Med Chem 7: 39–58.
  9. 9. Plyta ZF, Li T, Papageorgiou VP, Mellidis AS, Assimopoulou AN, et al. (1998) Inhibition of topoisomerase I by naphthoquinone derivatives. Bioorg Med Chem Lett 8: 3385–3390.
  10. 10. Masuda Y, Nishida A, Hori K, Hirabayashi T, Kajimoto S, et al. (2003) Beta-hydroxyisovalerylshikonin induces apoptosis in human leukemia cells by inhibiting the activity of a polo-like kinase 1 (PLK1). Oncogene 22: 1012–1023.
  11. 11. Nakaya K, Miyasaka T (2003) A shikonin derivative, beta-hydroxyisovalerylshikonin, is an ATP-non-competitive inhibitor of protein tyrosine kinases. Anticancer Drugs 14: 683–693.
  12. 12. Kim SH, Kang IC, Yoon TJ, Park YM, Kang KS, et al. (2001) Antitumor activities of a newly synthesized shikonin derivative, 2-hyim-DMNQ-S-33. Cancer Lett 172: 171–175.
  13. 13. Masuda Y, Shima G, Aiuchi T, Horie M, Hori K, et al. (2004) Involvement of tumor necrosis factor receptor-associated protein 1 (TRAP1) in apoptosis induced by beta-hydroxyisovalerylshikonin. J Biol Chem 279: 42503–42515.
  14. 14. Hsu PC, Huang YT, Tsai ML, Wang YJ, Lin JK, et al. (2004) 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 52: 6330–6337.
  15. 15. Yoon Y, Kim YO, Lim NY, Jeon WK, Sung HJ (1999) Shikonin, an ingredient of Lithospermum erythrorhizon induced apoptosis in HL60 human premyelocytic leukemia cell line. Planta Med 65: 532–535.
  16. 16. Yang H, Zhou P, Huang H, Chen D, Ma N, et al. (2009) Shikonin exerts antitumor activity via proteasome inhibition and cell death induction in vitro and in vivo. Int J Cancer 124: 2450–2459.
  17. 17. Sankawa U, Ebizuka Y, Miyazaki T, Isomura Y, Otsuka H (1977) Antitumor activity of shikonin and its derivatives. Chem Pharm Bull (Tokyo) 25: 2392–2395.
  18. 18. Sankawa U, Otsuka H, Kataoka Y, Iitaka Y, Hoshi A, et al. (1981) Antitumor activity of shikonin, alkannin and their derivatives. II. X-ray analysis of cyclo-alkannin leucoacetate, tautomerism of alkannin and cyclo-alkannin and antitumor activity of alkannin derivatives. Chem Pharm Bull (Tokyo) 29: 116–122.
  19. 19. Hayashi M (1977) [Pharmacological studies of Shikon and Tooki. (2) Pharmacological effects of the pigment components, Shikonin and acetylshikonin]. Nihon Yakurigaku Zasshi 73: 193–203.
  20. 20. Papageorgiou VP, Assimopoulou AN, Couladouros EA, Hepworth D, Nicolaou KC (1999) The Chemistry and Biology of Alkannin, Shikonin, and Related Naphthazarin Natural Products. Angewandte Chemie International Edition 38: 270–301.
  21. 21. Guo XP, Zhang XY, Zhang SD (1991) [Clinical trial on the effects of shikonin mixture on later stage lung cancer]. Zhong Xi Yi Jie He Za Zhi 11: 598–580, 598-599, 580.
  22. 22. Li L, Han W, Gu Y, Qiu S, Lu Q, et al. (2007) Honokiol induces a necrotic cell death through the mitochondrial permeability transition pore. Cancer Res 67: 4894–4903.
  23. 23. Xuan Y, Hu X (2009) Naturally-occurring shikonin analogues–a class of necroptotic inducers that circumvent cancer drug resistance. Cancer Lett 274: 233–242.
  24. 24. Hu X, Han W, Li L (2007) Targeting the weak point of cancer by induction of necroptosis. Autophagy 3: 490–492.
  25. 25. Han W, Li L, Qiu S, Lu Q, Pan Q, et al. (2007) Shikonin circumvents cancer drug resistance by induction of a necroptotic death. Mol Cancer Ther 6: 1641–1649.
  26. 26. Hu X, Xuan Y (2008) Bypassing cancer drug resistance by activating multiple death pathways–a proposal from the study of circumventing cancer drug resistance by induction of necroptosis. Cancer Lett 259: 127–137.
  27. 27. Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, et al. (2005) Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol 1: 112–119.
  28. 28. Degterev A, Hitomi J, Germscheid M, Ch'en IL, Korkina O, et al. (2008) Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat Chem Biol 4: 313–321.
  29. 29. Degterev A, Yuan J (2008) Expansion and evolution of cell death programmes. Nat Rev Mol Cell Biol 9: 378–390.
  30. 30. Chen J, Xie J, Jiang Z, Wang B, Wang Y, et al. (2011) Shikonin and its analogs inhibit cancer cell glycolysis by targeting tumor pyruvate kinase-M2. Oncogene
  31. 31. Altenberg B, Greulich KO (2004) Genes of glycolysis are ubiquitously overexpressed in 24 cancer classes. Genomics 84: 1014–1020.
  32. 32. Spoden GA, Rostek U, Lechner S, Mitterberger M, Mazurek S, et al. (2009) Pyruvate kinase isoenzyme M2 is a glycolytic sensor differentially regulating cell proliferation, cell size and apoptotic cell death dependent on glucose supply. Exp Cell Res 315: 2765–2774.
  33. 33. Spoden GA, Mazurek S, Morandell D, Bacher N, Ausserlechner MJ, et al. (2008) Isotype-specific inhibitors of the glycolytic key regulator pyruvate kinase subtype M2 moderately decelerate tumor cell proliferation. Int J Cancer 123: 312–321.
  34. 34. Christofk HR, Vander Heiden MG, Harris MH, Ramanathan A, Gerszten RE, et al. (2008) The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature 452: 230–233.
  35. 35. Ling CQ, Li B, Zhang C, Zhu DZ, Huang XQ, et al. (2005) Inhibitory effect of recombinant adenovirus carrying melittin gene on hepatocellular carcinoma. Ann Oncol 16: 109–115.
  36. 36. Chen J, Xie J, Jiang Z, Wang B, Wang Y, et al. (2011) Shikonin and its analogs inhibit cancer cell glycolysis by targeting tumor pyruvate kinase-M2. Oncogene 30: 4297–4306.
  37. 37. Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, et al. (2003) Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4: 249–264.
  38. 38. Tusher VG, Tibshirani R, Chu G (2001) Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A 98: 5116–5121.
  39. 39. Saeed AI, Sharov V, White J, Li J, Liang W, et al. (2003) TM4: a free, open-source system for microarray data management and analysis. Biotechniques 34: 374–378.
  40. 40. Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, et al. (2003) DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol 4: P3.
  41. 41. Floreani M, Napoli E, Palatini P (2000) Protective action of cardiac DT-diaphorase against menadione toxicity in guinea pig isolated atria. Biochem Pharmacol 60: 601–605.
  42. 42. Seve P, Dumontet C (2008) Is class III beta-tubulin a predictive factor in patients receiving tubulin-binding agents? Lancet Oncol 9: 168–175.
  43. 43. Hu X, Yang H, Pan QR, Zheng S (1995) Does P-glycoprotein play a pivotal role in the drug resistance of an MDR variant, K562/Dox? Chemotherapy 41: 296–305.
  44. 44. Qiangrong P, Wang T, Lu Q, Hu X (2005) Schisandrin B–a novel inhibitor of P-glycoprotein. Biochem Biophys Res Commun 335: 406–411.
  45. 45. Pan Q, Lu Q, Zhang K, Hu X (2006) Dibenzocyclooctadiene lingnans: a class of novel inhibitors of P-glycoprotein. Cancer Chemother Pharmacol 58: 99–106.
  46. 46. Xu D, Lu Q, Hu X (2006) Down-regulation of P-glycoprotein expression in MDR breast cancer cell MCF-7/ADR by honokiol. Cancer Lett 243: 274–280.
  47. 47. Li L, Pan Q, Sun M, Lu Q, Hu X (2007) Dibenzocyclooctadiene lignans: a class of novel inhibitors of multidrug resistance-associated protein 1. Life Sci 80: 741–748.
  48. 48. Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L (2004) Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev 56: 185–229.
  49. 49. Abolhoda A, Wilson AE, Ross H, Danenberg PV, Burt M, et al. (1999) Rapid activation of MDR1 gene expression in human metastatic sarcoma after in vivo exposure to doxorubicin. Clin Cancer Res 5: 3352–3356.
  50. 50. Allen JD, Brinkhuis RF, Wijnholds J, Schinkel AH (1999) The mouse Bcrp1/Mxr/Abcp gene: amplification and overexpression in cell lines selected for resistance to topotecan, mitoxantrone, or doxorubicin. Cancer Res 59: 4237–4241.
  51. 51. Beketic-Oreskovic L, Duran GE, Chen G, Dumontet C, Sikic BI (1995) Decreased mutation rate for cellular resistance to doxorubicin and suppression of mdr1 gene activation by the cyclosporin PSC 833. J Natl Cancer Inst 87: 1593–1602.
  52. 52. Lau DH, Lewis AD, Ehsan MN, Sikic BI (1991) Multifactorial mechanisms associated with broad cross-resistance of ovarian carcinoma cells selected by cyanomorpholino doxorubicin. Cancer Res 51: 5181–5187.
  53. 53. Yeh PY, Chuang SE, Yeh KH, Song YC, Cheng AL (2003) Involvement of nuclear transcription factor-kappa B in low-dose doxorubicin-induced drug resistance of cervical carcinoma cells. Biochem Pharmacol 66: 25–33.
  54. 54. Han W, Xie J, Li L, Liu Z, Hu X (2009) Necrostatin-1 reverts shikonin-induced necroptosis to apoptosis. Apoptosis 14: 674–686.