The effect of non-steroidal anti-inflammatory drugs on ......The effect of non-steroidal...

10
2681 Abstract. OBJECTIVE: Osteosarcoma (OS), an aggressive malignancy, is the most common primary bone tumor in children. Nonsteroidal an- ti-inflammatory drugs (NSAIDs) are widely used to reduce pain and inflammation. NSAIDs have shown to be toxic to certain malignancies such as colorectal, breast, and pancreatic cancers, but are not well-studied in OS. The purpose of this study is to assess whether ketorolac induces apoptosis in OS cells, compare this to indometh- acin, which has been shown to inhibit OS prolif- eration, and explore the underlying mechanism. MATERIALS AND METHODS: A rat OS cell line (UMR-108) was exposed to various concen- trations of ketorolac and indomethacin. Cell vi- ability, cytotoxicity, apoptosis induction, DNA fragmentation and the expression of apopto- sis-related markers were examined by MTT as- say, colony formation assay, flow cytometry, agarose gel electrophoresis, and Western blot respectively. RESULTS: The results indicated that ketoro- lac and indomethacin could induce apoptosis of rat OS cells in a dose- and time-dependent man- ner. Apoptosis was confirmed by cell morphology and annexin positivity. The molecular data showed that NSAIDs affected expression of Bcl-2, survivin, and Poly (ADP-ribose) polymerase-1 (PARP). CONCLUSIONS: These findings demonstrat- ed that NSAIDs induced apoptosis in rat OS cells in vitro. Further research focusing on the poten- tial cytotoxicity of NSAIDs in vivo is needed. Key Words: Osteosarcoma, Non-Steroidal Anti-Inflammatory Drugs (NSAIDs), Ketorolac, Indomethacin, Apoptosis. Introduction Osteosarcoma (OS) is the most common pri- mary malignant bone tumor and typically affects the metaphyses of the distal femur, proximal tibia, or humerus in children and adolescents 1,2 . Stan- dard treatment for OS includes chemotherapy and surgery. Even though there has been a significant improvement in mortality, metastatic disease still occurs, which is the major cause of mortality in patients with OS 3,4 . Currently, the prognosis of patients with OS is 75% to 77% when no disea- se is detected. Without chemotherapy, the 5-year survival rate is no more than 20% 5-9 . Unfortu- nately, survival rates have remained unchanged over the past several decades as no new effective drug regimen has been developed. Non-steroidal anti-inflammatory drugs (NSAIDs) are frequent- ly used to reduce postoperative pain and chronic inflammation in orthopaedic patients worldwi- de 10 . NSAIDs block the synthesis of prostaglan- dins, particularly prostaglandin E2 (PGE2) in the cyclooxygenase (COX) pathway 11 . PGE2 has been shown to promote tumor cell growth and invasive- ness and to contribute to tumor angiogenesis 12,13 . NSAIDs can cause antitumor activity in different cell populations by inhibiting their growth capa- city and reducing cell migration, which may re- sult in a potential benefit of these drugs in cancer treatments 13-15 . Multiple types of sarcomas, inclu- ding OS, Ewing’s sarcoma and chondrosarcoma, express cyclooxygenase-2 (COX-2) 16-18 . Inhibition of COX-2 with NSAIDs such as meloxicam and celecoxib, have been shown to be toxic to OS cel- ls in vitro 19-21 . Indomethacin blocks the activity of both cyclooxygenase-1 (COX-1) and COX-2, although its greatest effect is on COX-1. Indo- methacin has been shown to have an effect on the osteoblastic proliferation of the human MG-63 OS cell line 10 . In addition to this, preclinical stu- dies 22-25 have established that indomethacin has significant efficacy against colon, breast and lung European Review for Medical and Pharmacological Sciences 2019; 23: 2681-2690 L.M. ZUCKERMAN 1 , W.L. FRAMES 1 , J.G. ELSISSY 1 , T.G. SHIELDS 1 , R. DE NECOCHEA-CAMPION 3 , H.R. MIRSHAHIDI 2 , N.L. WILLIAMS 1 , S. MIRSHAHIDI 3 1 Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA 2 Division of Hematology/Oncology, Loma Linda University School of Medicine, Loma Linda, CA, USA 3 Loma Linda Cancer Center Biospecimen Laboratory, Loma Linda University Medical Center, Loma Linda, CA, USA Corresponding Author: William L. Frames, MD; email: [email protected] The effect of non-steroidal anti-inflammatory drugs on osteosarcoma cells

Transcript of The effect of non-steroidal anti-inflammatory drugs on ......The effect of non-steroidal...

  • 2681

    Abstract. – OBJECTIVE: Osteosarcoma (OS), an aggressive malignancy, is the most common primary bone tumor in children. Nonsteroidal an-ti-inflammatory drugs (NSAIDs) are widely used to reduce pain and inflammation. NSAIDs have shown to be toxic to certain malignancies such as colorectal, breast, and pancreatic cancers, but are not well-studied in OS. The purpose of this study is to assess whether ketorolac induces apoptosis in OS cells, compare this to indometh-acin, which has been shown to inhibit OS prolif-eration, and explore the underlying mechanism.

    MATERIALS AND METHODS: A rat OS cell line (UMR-108) was exposed to various concen-trations of ketorolac and indomethacin. Cell vi-ability, cytotoxicity, apoptosis induction, DNA fragmentation and the expression of apopto-sis-related markers were examined by MTT as-say, colony formation assay, flow cytometry, agarose gel electrophoresis, and Western blot respectively.

    RESULTS: The results indicated that ketoro-lac and indomethacin could induce apoptosis of rat OS cells in a dose- and time-dependent man-ner. Apoptosis was confirmed by cell morphology and annexin positivity. The molecular data showed that NSAIDs affected expression of Bcl-2, survivin, and Poly (ADP-ribose) polymerase-1 (PARP).

    CONCLUSIONS: These findings demonstrat-ed that NSAIDs induced apoptosis in rat OS cells in vitro. Further research focusing on the poten-tial cytotoxicity of NSAIDs in vivo is needed.

    Key Words:Osteosarcoma, Non-Steroidal Anti-Inflammatory Drugs

    (NSAIDs), Ketorolac, Indomethacin, Apoptosis.

    Introduction

    Osteosarcoma (OS) is the most common pri-mary malignant bone tumor and typically affects

    the metaphyses of the distal femur, proximal tibia, or humerus in children and adolescents1,2. Stan-dard treatment for OS includes chemotherapy and surgery. Even though there has been a significant improvement in mortality, metastatic disease still occurs, which is the major cause of mortality in patients with OS3,4. Currently, the prognosis of patients with OS is 75% to 77% when no disea-se is detected. Without chemotherapy, the 5-year survival rate is no more than 20%5-9. Unfortu-nately, survival rates have remained unchanged over the past several decades as no new effective drug regimen has been developed. Non-steroidal anti-inflammatory drugs (NSAIDs) are frequent-ly used to reduce postoperative pain and chronic inflammation in orthopaedic patients worldwi-de10. NSAIDs block the synthesis of prostaglan-dins, particularly prostaglandin E2 (PGE2) in the cyclooxygenase (COX) pathway11. PGE2 has been shown to promote tumor cell growth and invasive-ness and to contribute to tumor angiogenesis12,13. NSAIDs can cause antitumor activity in different cell populations by inhibiting their growth capa-city and reducing cell migration, which may re-sult in a potential benefit of these drugs in cancer treatments13-15. Multiple types of sarcomas, inclu-ding OS, Ewing’s sarcoma and chondrosarcoma, express cyclooxygenase-2 (COX-2)16-18. Inhibition of COX-2 with NSAIDs such as meloxicam and celecoxib, have been shown to be toxic to OS cel-ls in vitro19-21. Indomethacin blocks the activity of both cyclooxygenase-1 (COX-1) and COX-2, although its greatest effect is on COX-1. Indo-methacin has been shown to have an effect on the osteoblastic proliferation of the human MG-63 OS cell line10. In addition to this, preclinical stu-dies22-25 have established that indomethacin has significant efficacy against colon, breast and lung

    European Review for Medical and Pharmacological Sciences 2019; 23: 2681-2690

    L.M. ZUCKERMAN1, W.L. FRAMES1, J.G. ELSISSY1, T.G. SHIELDS1, R. DE NECOCHEA-CAMPION3, H.R. MIRSHAHIDI2, N.L. WILLIAMS1, S. MIRSHAHIDI3

    1Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA2Division of Hematology/Oncology, Loma Linda University School of Medicine, Loma Linda, CA, USA3Loma Linda Cancer Center Biospecimen Laboratory, Loma Linda University Medical Center, Loma Linda, CA, USA

    Corresponding Author: William L. Frames, MD; email: [email protected]

    The effect of non-steroidal anti-inflammatory drugs on osteosarcoma cells

  • L.M. Zuckerman, W.L. Frames, J.G. Elsissy, T.G. Shields, R. de Necochea-Campion, et al.

    2682

    cancers. Ketorolac is a non-selective COX inhi-bitor of COX-1 and COX-2, has potent analgesic affects, and is given perioperatively, intraopera-tively and postoperatively to reduce moderate to severe surgical and cancer related pain26. Despite a lack of inhibition of serotonin and opioid recep-tors, ketorolac treatment confers opioid level pain relief more potent than many opioids27. Ketorolac is non-habit forming and patients do not develop tolerance. Ketorolac’s cost effectiveness relative to opioids or steroids, makes it a popular choice for alternative short term pain relief 28. It has been suggested that there is an association between the use of ketorolac and better outcomes after ma-stectomy for breast cancer and with ovarian and lung cancer29-31. However, the effect of ketorolac has not been studied in OS. This study investi-gated the effects of ketorolac and indomethacin and the underlying antitumor mechanisms in rat OS cells.

    Materials and Methods

    Cell Culture and ReagentsA rat OS cell line UMR-108 (ATCC CRL-1663)

    was obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA) for this study. Cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM), supplemented with 10% fetal bovine serum (FBS), 100 U/mL penicil-lin G, 100 mg/mL streptomycin and 1% nonessen-tial amino acids. Cell cultures were maintained in a humidified incubator at 37°C with 5% CO2. Ke-torolac (30 mg/ml) and indomethacin (2 mg/ml) were purchased from McKesson Medical-Surgi-cal Inc. (Chino, CA, USA).

    Cell Viability AssayThe UMR-108 cell line was seeded in triplicate

    at a density of 4000 cells/well in 96-well plates. After cell confluence reached 80%, the cells were treated at various time points between 24 and 74 hours at 37°C with various concentrations of keto-rolac and indomethacin. The untreated group was placed in phosphate-buffered saline (PBS) with a pH of 5.5 and 100% ethanol in order to control for the pH and microenvironment of the treated cells. Cell viability was assessed with 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT reagent, Roche Diagnostics, Indianapolis, IN, USA) according to the manufacturer’s pro-tocol. The plates were read, and the absorbance at 570 nm was measured on a microplate reader

    (Bio-Rad Model 3550, Hercules, CA, USA). The background death of the untreated group was subtracted from the treated groups. The results were expressed as a percentage of the untreated control (% of control). Each experiment was done in triplicate.

    Microscopic Observation of Cell Morphology

    Morphologic changes in tumor cells were exa-mined by phase-contrast photomicrographs at 24, 48 and 72 hours after exposure to various concen-trations of ketorolac (0.93, 1.87, 3.75 mg) and in-domethacin (0.125, 0.250, 0.500, 1 mg). Apoptotic cells were characterized by cell shrinkage and de-tachment from the plates.

    Clonogenic Survival AssayUMR-108 cells were exposed to various con-

    centrations of ketorolac (0.93, 1.87, 3.75 mg) and indomethacin (0.125, 0.250, 0.500, 1 mg) for 24 and 48 hours. Then, 1000 cells from each time point were seeded in 6-well plates. After 12-14 days the colonies were stained with crystal vio-let, and the number of colonies was counted. Each experiment was done in triplicate.

    Analysis of Apoptosis and Cell CycleApoptotic cells were evaluated by an An-

    nexin-V (Ann) and propidium iodide (PI) apop-tosis detection kit (BD Biosciences, Franklin La-kes, NJ, USA). 1x105 cells were seeded in 6-well plates, and when confluence reached 80%, they were exposed to medications at various concen-trations. After 24 and 72 hours, both floating and attached cells were harvested and washed. Flow cytometry with FITC-conjugated Ann/PI double staining was used to assess the number of apopto-tic cells. Samples were analyzed by flow cytome-try (MACSQuant, Miltenyi Biotech, Auburn, CA, USA). The results were displayed as 4 quadrants, in which the lower right quadrant represented the apoptosis rate during the early stages, the upper right quadrant indicated advanced apoptosis rate, the upper left quadrant represented dead cells, and the lower left quadrant represented living cells. The apoptotic rate was calculated as early apop-tosis (Ann+/PI-) and late apoptosis (Ann+/PI+) using the FlowJo software (TreeStar, Ashland, OR, USA). This experiment was repeated 3 times.

    DNA Fragmentation on Agarose GelsUMR-108 cells were plated in 12-well plates,

    and after 24 hours cells were exposed to keto-

  • The effect of non-steroidal anti-inflammatory drugs on osteosarcoma cells

    2683

    rolac (0.93, 1.87, 3.75 mg) and indomethacin (0.125, 0.250, 0.500, 1 mg) for 48 hours. All of the cells, including floating cells, were harvested by trypsinization and washed with Dulbeccò s Phosphate Buffered Saline. The genomic DNA was extracted with a DNA extraction kit (Qui-ck-g DNA miniprep kit, Zymo Research, Irvine, CA, USA) as described by the manufacturer. The DNA from each group was measured by Nano-Vue (GE Healthcare, Chicago, IL, USA) and 0.4 μg was then applied to 1.5% agarose gels contai-ning μg/ml ethidium bromide. A DNA molecular marker (1 kb DNA step ladder, Promega, Ma-dison, WI, USA) with the DNA from apoptotic UMR-108 cells induced by H2O2 (0.4 mM) was used as a positive control. The DNA fragmenta-tion pattern was examined in photographs taken under UV illumination.

    Western Blot AnalysisUMR-108 cells were exposed to ketorolac (0.93,

    1.87, 3.75 mg) and indomethacin (0.125, 0.250, 0.500, 1 mg) for 48 and 72 hours. Treated cells were harvested, washed and resuspended in NP-40 lysis buffer. Whole cell lysates were homoge-nized and the protein concentrations were deter-mined by using the Pierce BCA Protein Assay Kit (Thermo Fisher Scientific, Waltham, MA, USA). Equal amounts of protein (20 µg) from each sam-ple were loaded and separated through 10-12% sodium dodecyl sulfate polyacrylamide (SDS-PA-GE) gels and then transferred to polyvinylidene difluoride (PVDF) membranes (Millipore, Bille-rica, MA, USA). The membranes were blocked with 5% BSA in PBST buffer for 1 hour at room temperature and incubated with primary antibo-dies overnight at 4°C. The following antibodies were used; Bcl-2, Poly(ADP-ribose) polymera-se-1 (PARP), caspase-3, β-Actin (Cell Signaling Technologies, Danvers, MA, USA) and survivin (Novus, Littleton, CO, USA). After washing with PBST, membranes were incubated with horsera-dish peroxidase (HRP) conjugated anti-rabbit IgG antibody (Cell Signaling Technologies, Beverly, MA, USA) for 1 hour at room temperature. The bands were visualized by an enhanced chemilu-minescence kit (Thermo Fisher Scientific, Wal-tham, MA, USA) according to the manufacturer’s instructions. The data was normalized to corre-sponding values of β-actin densitometry.

    Statistical AnalysisEach assay was performed at least three times

    as independent experiments. Statistical analyses

    were performed with ANOVA and done with Pri-sm 5.01 software (GraphPad Software, La Jolla, CA, USA). A p-value of

  • L.M. Zuckerman, W.L. Frames, J.G. Elsissy, T.G. Shields, R. de Necochea-Campion, et al.

    2684

    NSAIDs Induced Apoptosis in Tumor Cells Quantitative analysis showed that total apop-

    totic cells (Ann+) increased significantly when cells were exposed to the different doses of both ketorolac and indomethacin (Figure 4). The total apoptotic cells significantly increased from 10% to 32% after 24 hours and from 27% to 87% after 72 hours of exposure to ketorolac. At 24 hours, the apoptotic cells increased from 7% to 48% and from 16% to 79% after 72 hours of exposure to indomethacin. This was again found to occur in a time- and dose-dependent manner when compa-red to untreated tumor cells.

    NSAIDs Induced Apoptosis is Characterized by the Degradation of DNA

    One of the hallmarks of apoptosis is the degra-dation of nuclear DNA into nucleosomal units, which is demonstrated as a ‘DNA ladder’ by gel 32. We further ascertained the drug-induced apop-tosis by DNA gel electrophoresis at 48 hours. As shown in Figure 5, both drugs significantly incre-ased the intensity of the fluorescence of ethidium bromide. A significant increase in the DNA frag-mentation occurred in a dose-dependent manner.

    NSAIDs Modulate the Expression of Apoptosis Related Proteins

    To further elucidate the mechanisms of ke-torolac- and indomethacin-induced apoptosis in OS cells, we evaluated the changes in expression of proteins involved in apoptosis such as Bcl-2, survivin, caspase-3 and PARP by Western blot analysis. UMR-108 cells were exposed to various

    doses of ketorolac and indomethacin. After 48 and 72 hours, total protein was isolated. The densito-metry analysis demonstrated that treatment with ketorolac and indomethacin significantly reduced the level of Bcl-2 in UMR-108 cells in a dose-de-pendent manner (Figure 6A). The time-dependent effect was more pronounced at 72 hours when compared to 48 hours (***p

  • The effect of non-steroidal anti-inflammatory drugs on osteosarcoma cells

    2685

    reducing cell migration, which inhibits metasta-sis33. It has also been shown that NSAIDs are ca-spase inhibitors34. The results of this study showed that ketorolac and indomethacin affect caspase-3 in OS. Caspase-3 is involved in both the intrinsic and extrinsic apoptotic pathway35. When caspa-se-3 is activated, PARP cleavage occurs. The cle-avage of PARP causes the enzyme to lose much of its DNA binding capacity36,37. This degradation of PARP also affects DNA repair38. These results demonstrate that the effect of NSAID’s on OS is likely in the apoptotic pathway. These findings are furthered by the effect of NSAID’s on survivin in this study. Survivin inhibits both the intrinsic and extrinsic apoptotic pathways39. It has been shown

    that DNA damage results in down-regulation of survivin and apoptosis40. Survivin has also been found to target caspase-3 which further disrupts the apoptotic pathway41. Bcl-2 was also affected by both NSAIDs in this study. Bcl-2 has a diffe-rent role in apoptosis compared to survivin. Whe-reas survivin inhibits caspases, which complete apoptosis, Bcl-2 prevents the initiation of apop-tosis42. Bcl-2 protects cell membrane integrity by interacting with mitochondrial proteins to prevent the opening of the mitochondrial permeability transition pore and the release of apoptogenic fac-tors43. Scholars10,17 have shown that COX inhibi-tion in certain malignancies results in impaired tumor cell proliferation, migration, and metasta-

    Figure 2. NSAIDs decrease colony formation capacity of OS tumor cells. UMR-108 cells were exposed to ketorolac and indo-methacin at various concentrations for A) 24 hours and B) 48 hours. Colonies were stained with crystal violet after 12-14 days, and the number of colonies was counted. Error bars indicate the standard deviation of the mean of three independent experiments performed in triplicate (***p

  • L.M. Zuckerman, W.L. Frames, J.G. Elsissy, T.G. Shields, R. de Necochea-Campion, et al.

    2686

    Figure 3. NSAIDs induce abnormal morphological changes in OS tumor cells. UMR-108 cells were treated with ketoroloc and indomethacin at various concentrations. Morphologic changes in tumor cells were examined by phase-contrast photomicrograph after 24, 48 and 72 hours. Cells exhibited abnormal morphology characterized by cellular shrinkage, turning round, floating and eventually death when compared to untreated tumor cells.

    Figure 4. Flow cytometric analysis of NSAIDs on OS tumor cells. UMR-108 cells were exposed to ketorolac and indometha-cin at various for 24 and 72 hours. Both floating and attached cells were harvested and then washed. Flow cytometry with FITC conjugated Annexin-V/propidium iodide double staining was used to assess the number of apoptotic cells and compared to the untreated group using the FlowJo software. Error bars indicate the standard deviation of the mean of three independent experi-ments performed in triplicate (***p

  • The effect of non-steroidal anti-inflammatory drugs on osteosarcoma cells

    2687

    Figure 5. Effect of NSAIDs on the DNA fragmentation in OS tumor cells. UMR-108 cells were exposed to ketorolac and indo-methacin at various concentrations for 48 hours. The genomic DNA was applied to 1.5% agarose gels containing mg/ml ethidium bromide. DNA step ladder (1 kb) and the DNA from apoptotic UMR-108 cells induced by H2O2 (0.4 mM) were used as a positive control. The DNA fragmentation pattern was examined in photographs taken under UV illumination. Error bars indicate the stan-dard deviation of the mean of three independent experiments performed in triplicate (***p

  • L.M. Zuckerman, W.L. Frames, J.G. Elsissy, T.G. Shields, R. de Necochea-Campion, et al.

    2688

    tic potential. Our data supports and adds to these findings. The NSAIDs were found to act in both a time- and dose-dependent fashion to inhibit co-lony formation, induce abnormal cell morphology and induce apoptosis. This was observed direct-ly by the analysis of the cell shape, as well as by DNA fragmentation assay and cell viability stu-dies. The demonstration that the NSAIDs affect proliferation, migration, and induce apoptosis can carry clinical relevance and potentially help to direct future studies. A weakness of the study includes the fact that this is an in vitro study. The behavior of medications in the in vivo setting can-not be assumed based on in vitro studies. Further in vivo studies should be performed in order to evaluate the effect as well as proper dosing of the medications in order to achieve a similar effect. The use of a cell line is both an advantage and disadvantage. The cell line allows for consistency and reproducibility of the findings. The problem with a cell line, in regards to OS, is that OS has multiple subtypes44. These subtypes have varying responses to chemotherapy and differing survival rates45. This heterogeneity and varying response to treatment between patients make it difficult to fully extrapolate the results to a clinical setting. NSAIDs are well studied, inexpensive and readily available medications with known side effect pro-files10. Due to this, translational research can be performed without having to develop new medi-cations or testing for toxicity. Targeted therapies could be developed based on the mechanism of action found and be utilized as well. The medica-tions also help decrease inflammation and pain, which can assist the cancer-related pain that OS patients endure.

    Conclusions

    The findings of this study support the role for evaluating NSAIDs in the clinical setting. If a translatable effect is demonstrated, the medica-tions could be considered as both a neoadjuvant and adjuvant treatment.

    Conflict of InterestThe Authors declare that they have no conflict of interest.

    DisclosuresLee M. Zuckerman - consultant to NuVasive Specialized Orthopedics.

    References

    1) Picci P. Osteosarcoma (osteogenic sarcoma). Or-phanet J Rare Dis 2007; 2: 6.

    2) Wang W, Li X, Meng FB, Wang ZX, Zhao RT, Yang cY. Effects of the long non-coding RNA HOST2 on the proliferation, migration, invasion and apoptosis of human osteosarcoma cells. Cell Physiol Biochem 2017; 43: 320-330.

    3) aRndT ca, Rose Ps, FoLPe aL, Laack nn. Common musculoskeletal tumors of childhood and adole-scence. Mayo Clin Proc 2012; 87: 475-487.

    4) Maki Rg. Ifosfamide in the neoadjuvant treatment of osteogenic sarcoma. J Clin Oncol 2012; 30: 2033-2035.

    5) FeRRaRi s, sMeLand s, MeRcuRi M, BeRToni F, Longhi a, RuggieRi P. Neoadjuvant chemotherapy with high-dose ifosfamide, high-dose methotrexate, cisplatin, and doxorubicin for patients with lo-calized osteosarcoma of the extremity: a joint study by the Italian and Scandinavian Sarcoma Groups. J Clin Oncol 2005; 23: 8845-8852.

    6) dahLin dc, covenTRY MB. Osteogenic sarcoma. A study of six hundred cases. J Bone Joint Surg Am 1967; 49: 101-110.

    7) Bacci, g, RuggieRi P, BeRToni F, FeRRaRi s, Longhi a, Biagini R, ZavaTTa M, veRsaRi M, FoRni c. Local and systemic control for osteosarcoma of the extre-mity treated with neoadjuvant chemotherapy and limb salvage surgery: the Rizzoli experience. Oncol Rep 2000; 7: 1129-33.

    8) JaFFe n, FRei e. Osteogenic sarcoma: advances in treatment. CA Cancer J Clin 1976; 26: 351-359.

    9) MiRaBeLLo L, TRoisi RJ, savage sa. Osteosarcoma incidence and survival rates from 1973 to 2004: data from the surveillance, epidemiology, and end results program. Cancer 2009; 115: 1531-1543.

    10) diaZ-RodRigueZ L, gaRcía-MaRTíneZ o, MoRaLes Ma, RodRígueZ-PéReZ L, RuBio-RuiZ B, RuiZ c. Effects of indomethacin, nimesulide, and diclofenac on human MG-63 osteosarcoma cell line. Biol Res Nurs 2012; 14: 98-107.

    11) vane JR, BoTTing RM. Anti-inflammatory drugs and their mechanism of action. Inflamm Res 1998; 47: 78-87.

    12) Bai XM, Zhang W, Liu nB, Jiang h, Lou kX, Peng T, Ma J, Zhang L, Zhang h, Leng J. Focal adhesion kinase: important to prostaglandin E2-mediated adhesion, migration and invasion in hepatocel-lular carcinoma cells. Oncol Rep 2009; 21: 129-136.

    13) guRPinaR e, gRiZZLe We, PiaZZa ga. NSAIDs inhibit tumorigenesis, but how? Clin Cancer Res 2014; 20: 1104-1113.

    14) soBoLeWski c, ceReLLa c, dicaTo M, ghiBeLLi L, diede-Rich M. The role of cyclooxygenase-2 in cell pro-liferation and cell death in human malignancies. Int J Cell Biol 2010; 2010: 215158.

  • The effect of non-steroidal anti-inflammatory drugs on osteosarcoma cells

    2689

    15) huLL Ma, gaRdneR sh, haWcRoFT g. Activity of the non-steroidal anti-inflammatory drug indometha-cin against colorectal cancer. Cancer Treat Rev 2003; 29: 309-320.

    16) suTTon kM, WRighT M, FondRen g, ToWLe ca, Man-kin hJ. Cyclooxygenase-2 expression in chon-drosarcoma. Oncology 2004; 66: 275-280.

    17) Lee eJ, choi eM, kiM sR, PaRk Jh, kiM h, ha ks, kiM YM, kiM ss, choe M, kiM Ji, han Ja. Cyclooxyge-nase-2 promotes cell proliferation, migration and invasion in U2OS human osteosarcoma cells. Exp Mol Med 2007; 39: 469-476.

    18) Xu Z, choudhaRY s, voZnesenskY o, MehRoTRa M, WoodaRd M, hansen M, heRschMan h, PiLBeaM c. Overexpression of COX-2 in human osteosarco-ma cells decreases proliferation and increases apoptosis. Cancer Res 2006; 66: 6657-6664.

    19) dickens ds, koZieLski R, khan J, FoRus a, cRiPe TP. Cyclooxygenase-2 expression in pediatric sar-comas. Pediatr Dev Pathol 2002; 5: 356-364.

    20) Liu B, shi ZL, Feng J, Tao hM. Celecoxib, a cyclo-oxygenase-2 inhibitor, induces apoptosis in hu-man osteosarcoma cell line MG-63 via down-re-gulation of PI3K/Akt. Cell Biol Int 2008; 32: 494-501.

    21) naRuse T, nishida Y, hosono k, ishiguRo n. Meloxi-cam inhibits osteosarcoma growth, invasiveness and metastasis by COX-2-dependent and inde-pendent routes. Carcinogenesis 2006; 27: 584-592.

    22) LundhoLM k, geLin J, hYLTandeR a, LönnRoTh c, sandsTRöM R, svaningeR g, köRneR u, güLich M, käRReFoRs i, noRLi B, haFsTRoM o, keWenTeR J, oLBe L, LundeLL L. Anti-inflammatory treatment may prolong survival in undernourished patients with metastatic solid tumors. Cancer Res 1994; 54: 5602-5606.

    23) Zhou d, PaPaYannis i, MackenZie gg, aLsTon n, ouYang n, huang L, nie T, Wong cc, Rigas B. The anticancer effect of phospho-tyrosol-indometha-cin (MPI-621), a novel phosphoderivative of indo-methacin: in vitro and in vivo studies. Carcinoge-nesis 2013; 34: 943-951.

    24) ackeRsTaFF e, giMi B, aRTeMov d, BhuJWaLLa ZM. An-ti-inflammatory agent indomethacin reduces in-vasion and alters metabolism in a human breast cancer cell line. Neoplasia 2007; 9: 222-235.

    25) kaTo T, FuJino h, oYaMa s, kaWashiMa T, MuRaYaMa T. Indomethacin induces cellular morphological change and migration via epithelial-mesenchy-mal transition in A549 human lung cancer cells: a novel cyclooxygenase-inhibition-independent effect. Biochem Pharmacol 2011; 82: 1781-1791.

    26) vacha Me, huang W, Mando-vandRick J. The role of subcutaneous ketorolac for pain management. Hosp Pharm 2015; 50: 108-112.

    27) vanegas h, vaZqueZ e, ToRToRici v. NSAIDs, opio-ids, cannabinoids and the control of pain by the central nervous system. Pharmaceuticals (Ba-sel) 2010; 3: 1335-1347.

    28) JeLinek ga. Ketorolac versus morphine for severe pain. Ketorolac is more effective, cheaper, and has fewer side effects. BMJ 2000; 321: 1236-1237.

    29) FoRgeT P, BenTin c, MachieLs JP, BeRLieRe M, couLie Pg, de kock M. Intraoperative use of ketorolac or diclofenac is associated with improved dise-ase-free survival and overall survival in conser-vative breast cancer surgery. Br J Anaesth 2014; 113: 82-87.

    30) FoRgeT P, vandenhende J, BeRLieRe M, MachieLs JP, nussBauM B, LegRand c, de kock M. Do intraopera-tive analgesics influence breast cancer recurren-ce after mastectomy? A retrospective analysis. Anesth Analg 2010; 110: 1630-1635.

    31) guo Y, kenneY sR, cook L, adaMs sF, RuTLedge T, RoMeRo e, oPRea Ti, skLaR La, BedRick e, Wiggins cL, kang h, LoMo L, MuLLeR cY, WandingeR-ness a, hudson Lg. A novel pharmacologic activity of ke-torolac for therapeutic benefit in ovarian cancer patients. Clin Cancer Res 2015; 21: 5064-5072.

    32) RahBaR saadaT Y, saeidi n, Zununi vahed s, BaRZe-gaRi a, BaRaR J. An update to DNA ladder assay for apoptosis detection. Bioimpacts 2015; 5: 25-28.

    33) aXeLsson h, LönnRoTh c, andeRsson M, LundhoLM k. Mechanisms behind COX-1 and COX-2 inhibi-tion of tumor growth in vivo. Int J Oncol 2010; 37: 1143-1152.

    34) sMiTh ce, soTi s, Jones Ta, nakagaWa a, Xue d, Yin h. Non-steroidal anti-inflammatory drugs are caspase inhibitors. Cell Chem Biol 2017; 24: 281-292.

    35) MciLWain dR, BeRgeR T, Mak TW. Caspase fun-ctions in cell death and disease. Cold Spring Harb Perspect Biol 2013; 5: a008656.

    36) eLMoRe s. Apoptosis: a review of programmed cell death. Toxicol Pathol 2007; 35: 495-516.

    37) chaiTanYa gv, sTeven aJ, BaBu PP. PARP-1 clea-vage fragments: signatures of cell-death protea-ses in neurodegeneration. Cell Commun Signal 2010; 8: 31.

    38) oniZuka s, TaMuRa R, Yonaha T, oda n, kaWasa-ki Y, shiRasaka T, shiRaishi s, TsuneYoshi i. Clinical dose of lidocaine destroys the cell membrane and induces both necrosis and apoptosis in an identified Lymnaea neuron. J Anesth 2012; 26: 54-61.

    39) aLTieRi dc. Validating survivin as a cancer thera-peutic target. Nat Rev Cancer 2003; 3: 46-54.

    40) Zhou M, gu L, Li F, Zhu Y, Woods Wg, FindLeY hW. DNA damage induces a novel p53-survi-vin signaling pathway regulating cell cycle and apoptosis in acute lymphoblastic leukemia cel-ls. J Pharmacol Exp Ther 2002; 303: 124-131.

    41) Li Yh, Wang c, Meng k, chen LB, Zhou XJ. Influen-ce of survivin and caspase-3 on cell apoptosis and prognosis in gastric carcinoma. World J Ga-stroenterol 2004; 10: 1984-1988.

  • L.M. Zuckerman, W.L. Frames, J.G. Elsissy, T.G. Shields, R. de Necochea-Campion, et al.

    2690

    42) chen X, Zhou Y, Wang J, Wang J, Yang J, Zhai Y, Li B. Dual silencing of Bcl-2 and survivin by HSV-1 vector shows better antitumor efficacy in higher PKR phosphorylation tumor cells in vitro and in vivo. Cancer Gene Ther 2015; 22: 380-386.

    43) indRan iR, TuFo g, PeRvaiZ s, BRenneR c. Recent advances in apoptosis, mitochondria and drug resistance in cancer cells. Biochim Biophys Acta 2011; 1807: 735-745.

    44) FoX Mg, TRoTTa BM. Osteosarcoma: review of the various types with emphasis on recent advance-ments in imaging. Semin Musculoskelet Radiol 2013; 17: 123-136.

    45) oBiedaT h, aLRaBadi n, suLTan e, aL shaTTi M, ZihLiF M. The effect of ERCC1 and ERCC2 gene poly-morphysims on response to cisplatin based the-rapy in osteosarcoma patients. BMC Med Genet 2018; 19: 112.