Interactions of Analgesics with Cisplatin: Comparison
Please note this is a comparison between Version 2 by Beatrix Zheng and Version 1 by Azza Ali Kamel El-Sheikh.

Cisplatin (CDDP), one of the most eminent cancer chemotherapeutic agents, has been successfully used to treat more than half of all known cancers worldwide. Despite its effectiveness, CDDP might cause severe toxic adverse effects on multiple body organs during cancer chemotherapy, including the kidneys, heart, liver, gastrointestinal tract, and auditory system, as well as peripheral nerves causing severely painful neuropathy. The latter, among other pains patients feel during chemotherapy, is an indication for the use of analgesics during treatment with CDDP. Different types of analgesics, such as acetaminophen, non-steroidal anti-inflammatory drugs (NSAIDS), and narcotic analgesics, could be used according to the severity of pain. Administered analgesics might modulate CDDP’s efficacy as an anticancer drug. NSAIDS, on one hand, might have cytotoxic effects on their own and few of them can potentiate CDDP’s anticancer effects via inhibiting the CDDP-induced cyclooxygenase (COX) enzyme, or through COX-independent mechanisms. On the other hand, some narcotic analgesics might ameliorate CDDP’s anti-neoplastic effects, causing chemotherapy to fail. Concerning safety, some analgesics share the same adverse effects on normal tissues as CDDP, augmenting its potentially hazardous effects on organ impairment.

  • cisplatin
  • analgesics
  • acetaminophen
  • non-steroidal anti-inflammatory drugs
  • morphine
  • cytotoxicity

1. Interactions of NSAIDs with CDDP

The major mechanism of action of NSAIDs in treating pain is through the inhibition of the cyclooxygenase (COX) enzyme that catalyzes the formation of eicosanoids that mediate inflammation and pain, such as thromboxanes, prostaglandins, and prostacyclins, from membrane phospholipid arachidonic acid [24][1]. Since inflammation offers a suitable microenvironment for malignancies to develop, it is conceivable that NSAIDs possessing anti-inflammatory properties may help in the management of cancer. Interestingly, CDDP can induce COX-2 that causes the secretion of large amounts of prostaglandins, resulting in a decrease in CDDP chemotherapeutic efficacy [25,26][2][3]. It is, thus, logical that NSAIDs, especially selective COX-2 inhibitors, might act as chemosensitizers to resistant cancers, making them more susceptible to treatment by CDDP [27][4]. Interestingly, several non-selective NSAIDs, such as ketoprofen and naproxen, were assumed to have cytotoxic, anti-proliferative effects on their own, which was independent from the COX pathway, but seemed to be, at least partially, due to the induction of the NSAID-activated gene; NAG-1 [28][5]. NSAIDs that hold some potential to improve CDDP anticancer effects are summarized in Figure 21.
Figure 21. Effect of different analgesics on cisplatin’s anticancer efficacy. Analgesic names in green letters are non-steroidal anti-inflammatory drugs and those in red letters are narcotic analgesics.

1.1. Interactions of Salicylates with CDDP

Salicylate, the prototype of NSAIDs, has shown promising anticancer effects [36,37,38][6][7][8]. Several studies indicated that salicylate can, by different mechanisms, increase the cytotoxic efficacy of CDDP. In one study, salicylate was reported to improve the anti-tumor effects of CDDP against T cell lymphoma via changing the tumor microenvironment pH, altering the expression of the cell cycle’s regulatory/apoptotic factors, such as p53, bcl-2, bcl-xL, cyclin B1, and D, as well as cytokines IFN-γ, VEGF, IL-4, and -10 [39][9]. Other studies showed that salicylate also increased the anti-tumor effect of CDDP against osteosarcoma, through modulating the NF-κB pathway [40][10], and against non-small cell lung carcinoma stem-like cells by repressing migration through acting on the mTOR-Akt axis [41][11]. In addition, salicylate improved CDDP toxicity against colon cancer cells through preventing NF-κB binding to a COX-2 promoter [42][12] against lung cancer cells, by abrogating cancer cell stemness [43][13], against epithelial ovarian cancer cells by increasing p53 acetylation and promoting apoptosis [44][14], and against oesophageal squamous cell carcinoma through epigenetic modulation of chromatin by altering histone acetylation levels [45][15]. Due to these beneficial effects, asplatin or prodrug platin-A, which are CDDP-based Pt(IV) prodrugs complexed with salicylate, were developed to improve cytotoxicity against resistant cancer cells [46,47][16][17]. Despite its obvious potentiating cytotoxic effects on tumor cells, salicylate might have a protective effect on normal cells against CDDP-induced nephrotoxicity, ototoxicity, and neurotoxicity [48,49,50][18][19][20].

1.2. Interaction of Propionic Acid-Derived NSAIDs (Profens) with CDDP

Ibuprofen, one of the propionic acid-derived NSAIDs, showed cytotoxic effects when administered alone to human promyelocytic leukemia and colon carcinoma cells [51][21]. Some studies succeeded in synthesizing lipid encapsulated ibuprofen metallodrug nanoparticles to overcome CDDP chemoresistance in glioblastoma cancer cells [52][22]. It was also reported that ibuprofen increased CDDP anticancer efficacy against lung cancer cells through depletion of heat shock protein 70, thus enhancing tumor cell apoptosis [53][23]. In addition, combining ibuprofen with CDDP caused a higher cytotoxic effect on thyroid and pancreatic cancer cells in vitro [54][24]. Furthermore, an ibuprofen/CDDP combination reversed CDDP resistance in non-small-cell lung cancer through a COX-independent mechanism [55][25]. In addition to increasing CDDP’s cytotoxic efficacy, ibuprofen was reported to inhibit human ovarian cancer cell metastasis into several organs, such as the liver, lungs, bone marrow, and spleen in mice [56][26]. Unfortunately, through stimulating oxidative stress, ibuprofen might cause toxicity similar to CDDP on the kidneys and liver [57,58][27][28].
Ketoprofen, another propionic acid-derived NSAID, was conjugated with CDDP-based Pt(IV) prodrug to form ketoplatin that could delay breast cancer cells’ tumor growth and had less systemic toxic effects compared to CDDP alone in vitro and in vivo [59][29]. Interestingly, ketoprofen was suggested to protect against CDDP-induced nephrotoxicity [60][30], which is in line with more recent findings that ketoprofen has no nephrotoxic effects [61][31]. Several trials were also performed to assess the anti-tumor effects of combining CDDP with a third propionic acid-derived NSAID, naproxen [62[32][33],63], where the combination showed higher cytoxicity than CDDP alone on human cancer cells of the lungs and ovaries, with less toxicity on normal human liver cells [64][34]. Similar results were shown for a naproxen/CDDP combination on triple-negative breast cancer [65][35], as well as on ovarian endometrioid adenocarcinoma, lung adenocarcinoma, malignant pleural mesothelioma, and colon carcinoma cells [28][5]. Carprofen alone was able to ameliorate canine osteosarcoma in vitro [66][36]. Novel NSAIDs were created, such as derivatives of naproxen, flurbiprofen, and ibuprofen, that showed promising anticancer effect against cultured human glioblastoma cells [67][37], as well as human liver, breast, and colon carcinoma cells [68][38]. Whether the anticancer effects of these NSAIDS would be additive to that of CDDP or not still needs further investigation.

1.3. Interaction of Acetic Acid-Derived NSAIDS with CDDP

One of the acetic acid-derived NSAIDs, indomethacin, attenuated the growth of human oesophageal squamous carcinoma cells [69][39]. Sulindac could also ameliorate the growth rate of oral tumor cells and help their elimination by natural killer cells [70][40]. In addition, sulindac could prevent the progression of colorectal cancer clinically, by up-regulating cyclin G2 which resulted in delaying tumor cell cycle progression [71][41]. Interestingly, sulindac showed comparable cytotoxic effects to those of CDDP when tested on HEK293 cells [72][42]. Given together with CDDP, ketorolac succeeded in reversing CDDP chemo-resistance in a patient-derived cell xenograft model [73][43]. Diclofenac also showed improved CDDP anticancer effects against human lung adenocarcinoma CDDP-resistant cells [74,75][44][45]. To the contrary to what is expected from non-selective COX inhibitors, diclofenac did not deteriorate CDDP-induced nephrotoxicity [74][44]. Nevertheless, diclofenac, as with CDDP, had the hazard of causing hepatotoxicity as an adverse effect [76][46].

1.4. Interaction of Enolic Acid Derivatives of NSAIDs (Oxicams) with CDDP

Meloxicam, an enolic acid derivative of NSAIDs with relative preferential selectivity to inhibit COX-2, had a synergistic effect on CDDP cytotoxicity in human osteosarcoma cells [77][47]. Interestingly, meloxicam protected the kidney from CDDP-induced renal lesions in mice [78][48]. Oxicams have been suggested as chemosensitizers of CDDP, and some trials attempted to develop CDDP–oxicam complexes as anticancer drugs, using meloxicam and isoxicam, where the results showed promising cytotoxic effects on different cell lines in vitro [79][49]. Piroxicam, another enolic acid derivative of NSAIDs, when given as an adjuvant to CDDP-loaded nanoparticles, increased apoptosis in mesothelioma cells [80][50]. Unfortunately, unlike meloxicam, piroxicam was shown to worsen CDDP-induced nephrotoxicity in rats [81][51]. Tenoxicam alone seemed tolerable in patients with renal impairment [82][52], but was reported to have an injurious effect on the liver [83][53].

1.5. Interaction of Anthranilic Acid and Naphthylalanine Derivatives of NSAIDs (Fenamates) with CDDP

The anthranilic acid derivatives, flufenamic and mefenamic acids, were reported to augment CDDP’s anticancer effect in vitro through inhibiting aldo–keto reductase 1C enzyme [84,85][54][55]. Similarly, tolfenamic acid was coupled with CDDP to form a nanoprodrug that had tumor apoptotic and anti-metastatic effects on breast cancer in vitro and in vivo [26][3]. On their own, neither meclofenamic nor niflumic acid showed promising anticancer effects against uterine cervical cancer and breast adenocarcinoma cells, respectively [86,87][56][57]. Concerning safety, meclofenamic acid could aggravate CDDP-induced renal damage [88][58]. However, meclofenamic acid seemed to have the potential to protect against CDDP-induced ototoxicity via improving the viability of ear hair cells [89][59]. Nabumetone, a naphthylalanine derivative, had an antiproliferative effect on MCF-7 and MDA-MB-231 breast carcinoma cells [90][60], with low toxic effects on gastric mucosa cells [91][61].

1.6. Interaction of COX-II Selective NSAIDS (Coxibs) with CDDP

Selective COX-2 inhibitors, frequently referred to as “coxibs”, were reported to have, on their own, promising potential for preventing and treating malignancies [92,93][62][63]. Administered with CDDP, rofecoxib was reported to enhance cytotoxic effects on gastric cancer cells by down regulating multidrug resistance protein 1 expression [94][64]. Nevertheless, combining CDDP with celecoxib did not improve the anticancer activity of CDDP against human esophageal squamous cell carcinoma xenograft model in vivo [95][65]. Despite their safety with regards to gastric ulceration, selective COX-2 inhibitors were reported to mediate cardiotoxicity [32][66]. Indeed, several members of this group, such as valdecoxib and rofecoxib, were removed from the market due to their potential cardiovascular hazards [96,97][67][68]. Despite its reported hazard on cardiomyocytes [98][69], celecoxib only received a box warning on its pack, but is still sold in the market. Interestingly, parecoxib was reported to have a protective effect on ischemia-reperfusion injury of the heart in rats [99][70]. Celecoxib showed protective effect against CDDP-induced nephrotoxicity [100][71]. Another coxib, still present on the market, etoricoxib, was tested for possible nephroprotective effects against CDDP-induced renal toxicity in rats, but, unfortunately, the results were not conclusive [101][72].

References

  1. Ghlichloo, I.; Gerriets, V. Nonsteroidal Anti-Inflammatory Drugs (NSAIDs); StatPearls Publishing: Treasure Island, FL, USA, 2021; pp. 1–11.
  2. Ibrahim, M.A.; Albahlol, I.A.; Wani, F.A.; Tammam, A.A.-E.; Kelleni, M.T.; Sayeed, M.U.; El-Fadeal, N.M.A.; Mohamed, A.A. Resveratrol protects against cisplatin-induced ovarian and uterine toxicity in female rats by attenuating oxidative stress, inflammation and apoptosis. Chem. Interact. 2021, 338, 109402.
  3. Xing, L.; Yang, C.-X.; Zhao, D.; Shen, L.-J.; Zhou, T.-J.; Bi, Y.-Y.; Huang, Z.-J.; Wei, Q.; Li, L.; Li, F.; et al. A carrier-free anti-inflammatory platinum (II) self-delivered nanoprodrug for enhanced breast cancer therapy. J. Control. Release 2021, 331, 460–471.
  4. Dehkordi, N.G.; Mirzaei, S.A.; Elahian, F. Pharmacodynamic mechanisms of anti-inflammatory drugs on the chemosensitization of multidrug-resistant cancers and the pharmacogenetics effectiveness. Inflammopharmacology 2021, 29, 49–74.
  5. Ravera, M.; Zanellato, I.; Gabano, E.; Perin, E.; Rangone, B.; Coppola, M.; Osella, D. Antiproliferative Activity of Pt(IV) Conjugates Containing the Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) Ketoprofen and Naproxen. Int. J. Mol. Sci. 2019, 20, 3074.
  6. O’Brien, A.J.; Villani, L.A.; Broadfield, L.A.; Houde, V.P.; Galic, S.; Blandino, G.; Kemp, B.E.; Tsakiridis, T.; Muti, P.; Steinberg, G.R. Salicylate activates AMPK and synergizes with metformin to reduce the survival of prostate and lung cancer cells ex vivo through inhibition of de novo lipogenesis. Biochem. J. 2015, 469, 177–187.
  7. Broadfield, L.A.; Marcinko, K.; Tsakiridis, E.; Zacharidis, P.G.; Villani, L.; Lally, J.S.V.; Menjolian, G.; Maharaj, D.; Mathurin, T.; Smoke, M.; et al. Salicylate enhances the response of prostate cancer to radiotherapy. Prostate 2019, 79, 489–497.
  8. Karalis, T.T.; Chatzopoulos, A.; Kondyli, A.; Aletras, A.J.; Karamanos, N.K.; Heldin, P.; Skandalis, S.S. Salicylate suppresses the oncogenic hyaluronan network in metastatic breast cancer cells. Matrix Biol. Plus 2020, 6, 100031.
  9. Kumar, A.; Singh, S.M. Priming effect of aspirin for tumor cells to augment cytotoxic action of cisplatin against tumor cells: Implication of altered constitution of tumor microenvironment, expression of cell cycle, apoptosis, and survival regulatory molecules. Mol. Cell. Biochem. 2012, 371, 43–54.
  10. Liao, D.; Zhong, L.; Duan, T.; Zhang, R.-H.; Wang, X.; Wang, G.; Hu, K.; Lv, X.; Kang, T. Aspirin Suppresses the Growth and Metastasis of Osteosarcoma through the NF-κB Pathway. Clin. Cancer Res. 2015, 21, 5349–5359.
  11. Khan, P.; Bhattacharya, A.; Sengupta, D.; Banerjee, S.; Adhikary, A.; Das, T. Aspirin enhances cisplatin sensitivity of resistant non-small cell lung carcinoma stem-like cells by targeting mTOR-Akt axis to repress migration. Sci. Rep. 2019, 9, 16913.
  12. Jiang, W.; Yan, Y.; Chen, M.; Luo, G.; Hao, J.; Pan, J.; Hu, S.; Guo, P.; Li, W.; Wang, R.; et al. Aspirin enhances the sensitivity of colon cancer cells to cisplatin by abrogating the binding of NF-κB to the COX-2 promoter. Aging 2020, 12, 611–627.
  13. Zhao, M.; Wang, T.; Hui, Z. Aspirin overcomes cisplatin resistance in lung cancer by inhibiting cancer cell stemness. Thorac. Cancer 2020, 11, 3117–3125.
  14. Guo, J.; Zhu, Y.; Yu, L.; Li, Y.; Guo, J.; Cai, J.; Liu, L.; Wang, Z. Aspirin inhibits tumor progression and enhances cisplatin sensitivity in epithelial ovarian cancer. PeerJ 2021, 9, e11591.
  15. Zou, Z.; Zheng, W.; Fan, H.; Deng, G.; Lu, S.-H.; Jiang, W.; Yu, X. Aspirin enhances the therapeutic efficacy of cisplatin in oesophageal squamous cell carcinoma by inhibition of putative cancer stem cells. Br. J. Cancer 2021, 125, 1–13.
  16. Cheng, Q.; Shi, H.; Wang, H.; Min, Y.; Wang, J.; Liu, Y. The ligation of aspirin to cisplatin demonstrates significant synergistic effects on tumor cells. Chem. Commun. 2014, 50, 7427–7430.
  17. Pathak, R.K.; Marrache, S.; Choi, J.H.; Berding, T.B.; Dhar, S. The Prodrug Platin-A: Simultaneous Release of Cisplatin and Aspirin. Angew. Chem. Int. Ed. 2014, 53, 1963–1967.
  18. Ulubaş, B.; Cimen, M.Y.B.; Apa, D.D.; Saritaş, E.; Muslu, N.; Cimen, Ö.B. The Protective Effects of Acetylsalicylic Acid on Free Radical Production in Cisplatin Induced Nephrotoxicity: An Experimental Rat Model. Drug Chem. Toxicol. 2003, 26, 259–270.
  19. Yıldırım, M.; Inançlı, H.M.; Samancı, B.; Oktay, M.F.; Enöz, M.; Topçu, I. Preventing cisplatin induced ototoxicity by N-acetylcysteine and salicylate. Kulak Burun Bogaz Ihtis Derg 2010, 20, 173–183.
  20. Cetin, D.; Hacımuftuoglu, A.; Tatar, A.; Turkez, H.; Togar, B. The in vitro protective effect of salicylic acid against paclitaxel and cisplatin-induced neurotoxicity. Cytotechnology 2016, 68, 1361–1367.
  21. Kłobucki, M.; Urbaniak, A.; Grudniewska, A.; Kocbach, B.; Maciejewska, G.; Kiełbowicz, G.; Ugorski, M.; Wawrzeńczyk, C. Syntheses and cytotoxicity of phosphatidylcholines containing ibuprofen or naproxen moieties. Sci. Rep. 2019, 9, 220.
  22. Alves, S.R.; Colquhoun, A.; Wu, X.Y.; Silva, D.D.O. Synthesis of terpolymer-lipid encapsulated diruthenium(II, III)-anti-inflammatory metallodrug nanoparticles to enhance activity against glioblastoma cancer cells. J. Inorg. Biochem. 2020, 205, 110984.
  23. Endo, H.; Yano, M.; Okumura, Y.; Kido, H. Ibuprofen enhances the anticancer activity of cisplatin in lung cancer cells by inhibiting the heat shock protein 70. Cell Death Dis. 2014, 5, e1027.
  24. Petruzzella, E.; Sirota, R.; Solazzo, I.; Gandin, V.; Gibson, D. Triple action Pt(iv) derivatives of cisplatin: A new class of potent anticancer agents that overcome resistance. Chem. Sci. 2018, 9, 4299–4307.
  25. Fan, C.-C.; Tsai, S.-T.; Lin, C.-Y.; Chang, L.-C.; Yang, J.-C.; Chen, G.; Sher, Y.-P.; Wang, S.-C.; Hsiao, M.; Chang, W. EFHD2 contributes to non-small cell lung cancer cisplatin resistance by the activation of NOX4-ROS-ABCC1 axis. Redox Biol. 2020, 34, 101571.
  26. Gunjal, P.M.; Schneider, G.; Ismail, A.A.; Kakar, S.S.; Kucia, M.; Ratajczak, M.Z. Evidence for induction of a tumor metastasis-receptive microenvironment for ovarian cancer cells in bone marrow and other organs as an unwanted and underestimated side effect of chemotherapy/radiotherapy. J. Ovarian Res. 2015, 8, 1–11.
  27. Awad, D.S.; Ali, R.M.; Mhaidat, N.M.; Shotar, A.M. Zizyphus jujuba protects against ibuprofen-induced nephrotoxicity in rats. Pharm. Biol. 2013, 52, 182–186.
  28. Kim, M.; Lee, E.J.; Lim, K.-M. Ibuprofen Increases the Hepatotoxicity of Ethanol through Potentiating Oxidative Stress. Biomol. Ther. 2021, 29, 205–210.
  29. Ma, Z.-Y.; Song, X.-Q.; Hu, J.-J.; Wang, D.-B.; Ding, X.-J.; Liu, R.-P.; Dai, M.-L.; Meng, F.-Y.; Xu, J.-Y. Ketoplatin in triple-negative breast cancer cells MDA-MB-231: High efficacy and low toxicity, and positive impact on inflammatory microenvironment. Biochem. Pharmacol. 2021, 188, 114523.
  30. Yasuyuki, S.; Yoshihiko, S.; Yoshio, T.; Sadao, H. Protection against cisplatin-induced nephrotoxicity in the rat by inducers and an inhibitor of glutathione S-transferase. Biochem. Pharmacol. 1994, 48, 453–459.
  31. Fazzio, L.; Raggio, S.; Romero, J.; Membrebe, J.; Minervino, A. Safety Study on Ketoprofen in Pigs: Evaluating the Effects of Different Dosing and Treatment Scheme on Hematological, Hepatic, and Renal Parameters. Vet. Sci. 2021, 8, 30.
  32. Chen, Y.; Wang, Q.; Li, Z.; Liu, Z.; Zhao, Y.; Zhang, J.; Liu, M.; Wang, Z.; Li, D.; Han, J. Naproxen platinum(iv) hybrids inhibiting cycloxygenases and matrix metalloproteinases and causing DNA damage: Synthesis and biological evaluation as antitumor agents in vitro and in vivo. Dalton Trans. 2020, 49, 5192–5204.
  33. Wang, Q.; Hou, X.; Gao, J.; Ren, C.; Guo, Q.; Fan, H.; Liu, J.; Zhang, W.; Liu, J. A coassembled peptide hydrogel boosts the radiosensitization of cisplatin. Chem. Commun. 2020, 56, 13017–13020.
  34. Li, L.; Chen, Y.; Wang, Q.; Li, Z.; Liu, Z.; Hua, X.; Han, J.; Chang, C.; Wang, Z.; Li, D. Albumin-encapsulated Nanoparticles of Naproxen Platinum(IV) Complexes with Inflammation Inhibitory Competence Displaying Effective Antitumor Activities in vitro and in vivo. Int. J. Nanomed. 2021, 16, 5513–5529.
  35. Jin, S.; Muhammad, N.; Sun, Y.; Tan, Y.; Yuan, H.; Song, D.; Guo, Z.; Wang, X. Multispecific Platinum(IV) Complex Deters Breast Cancer via Interposing Inflammation and Immunosuppression as an Inhibitor of COX-2 and PD-L1. Angew. Chem. Int. Ed. 2020, 59, 23313–23321.
  36. Poradowski, D.; Obmińska-Mrukowicz, B. Effect of selected nonsteroidal anti-inflammatory drugs on the viability of canine osteosarcoma cells of the D-17 line: In vitro studies. J. Vet. Res. 2019, 63, 399–403.
  37. Özdemir, Ö.; Marinelli, L.; Cacciatore, I.; Ciulla, M.; Emsen, B.; Di Stefano, A.; Mardinoglu, A.; Turkez, H. Anticancer effects of novel NSAIDs derivatives on cultured human glioblastoma cells. Zeitschrift für Naturforschung C 2021, 76, 329–335.
  38. Aytaç, P.; Sahin, I.D.; Atalay, R.Ç.; Tozkoparan, B. Design, Synthesis, and Biological Evaluation of Novel Triazolothiadiazoles Derived from NSAIDs as Anticancer Agents. Anti-Cancer Agents Med. Chem. 2021, 21, 1.
  39. Liu, F.; Wu, Q.; Han, W.; Laster, K.; Hu, Y.; Ma, F.; Chen, H.; Tian, X.; Qiao, Y.; Liu, H.; et al. Targeting integrin αvβ3 with indomethacin inhibits patient-derived xenograft tumour growth and recurrence in oesophageal squamous cell carcinoma. Clin. Transl. Med. 2021, 11, e548.
  40. Kozlowska, A.K.; Topchyan, P.; Kaur, K.; Tseng, H.-C.; Teruel, A.; Hiraga, T.; Jewett, A. Differentiation by NK cells is a prerequisite for effective targeting of cancer stem cells/poorly differentiated tumors by chemopreventive and chemotherapeutic drugs. J. Cancer 2017, 8, 537–554.
  41. Zhao, H.; Yi, B.; Liang, Z.; Phillips, C.; Lin, H.-Y.; Riker, A.I.; Xi, Y. Cyclin G2, a novel target of sulindac to inhibit cell cycle progression in colorectal cancer. Genes Dis. 2021, 8, 320–330.
  42. Machkalyan, G.; Hèbert, T.E.; Miller, G.J. PPIP5K1 Suppresses Etoposide-triggered Apoptosis. J. Mol. Signal. 2016, 11, 4.
  43. Shriwas, O.; Priyadarshini, M.; Samal, S.K.; Rath, R.; Panda, S.; Das Majumdar, S.K.; Muduly, D.K.; Botlagunta, M.; Dash, R. DDX3 modulates cisplatin resistance in OSCC through ALKBH5-mediated m6A-demethylation of FOXM1 and NANOG. Apoptosis 2020, 25, 233–246.
  44. Okamoto, K.; Ueda, H.; Saito, Y.; Narumi, K.; Furugen, A.; Kobayashi, M. Diclofenac potentiates the antitumor effect of cisplatin in a xenograft mouse model transplanted with cisplatin-resistant cells without enhancing cisplatin-induced nephrotoxicity. Drug Metab. Pharmacokinet. 2021, 41, 100417.
  45. Okamoto, K.; Saito, Y.; Narumi, K.; Furugen, A.; Iseki, K.; Kobayashi, M. Anticancer effects of non-steroidal anti-inflammatory drugs against cancer cells and cancer stem cells. Toxicol. In Vitro 2021, 74, 105155.
  46. Heidarian, E.; Nouri, A. Hepatoprotective effects of silymarin against diclofenac-induced liver toxicity in male rats based on biochemical parameters and histological study. Arch. Physiol. Biochem. 2021, 127, 112–118.
  47. Naruse, T.; Nishida, Y.; Ishiguro, N. Synergistic effects of meloxicam and conventional cytotoxic drugs in human MG-63 osteosarcoma cells. Biomed. Pharmacother. 2007, 61, 338–346.
  48. Honma, S.; Takahashi, N.; Shinohara, M.; Nakamura, K.; Mitazaki, S.; Abe, S.; Yoshida, M. Amelioration of cisplatin-induced mouse renal lesions by a cyclooxygenase (COX)-2 selective inhibitor. Eur. J. Pharmacol. 2013, 715, 181–188.
  49. Tamasi, G.; Casolaro, M.; Magnani, A.; Sega, A.; Chiasserini, L.; Messori, L.; Gabbiani, C.; Valiahdi, S.M.; Jakupec, M.A.; Keppler, B.; et al. New platinum–oxicam complexes as anti-cancer drugs. Synthesis, characterization, release studies from smart hydrogels, evaluation of reactivity with selected proteins and cytotoxic activity in vitro. J. Inorg. Biochem. 2010, 104, 799–814.
  50. Menale, C.; Piccolo, M.T.; Favicchia, I.; Aruta, M.G.; Baldi, A.; Nicolucci, C.; Barba, V.; Mita, D.G.; Crispi, S.; Diano, N. Efficacy of Piroxicam Plus Cisplatin-Loaded PLGA Nanoparticles in Inducing Apoptosis in Mesothelioma Cells. Pharm. Res. 2014, 32, 362–374.
  51. Greene, S.N.; Ramos-Vara, J.A.; Craig, B.A.; Hooser, S.B.; Anderson, C.; Fourez, L.M.; Johnson, B.M.; Stewart, J.C.; Knapp, D.W. Effects of cyclooxygenase inhibitor treatment on the renal toxicity of cisplatin in rats. Cancer Chemother. Pharmacol. 2010, 65, 549–556.
  52. Nilsen, O.G.; Aasarod, K.; Wideroe, T.-E.; Guentert, T.W. Single- and multiple-dose pharmacokinetics, kidney tolerability and plasma protein binding of tenoxicam in renally impaired patients and healthy volunteers. Pharmacol. Toxicol. 2001, 89, 265–272.
  53. Karatopuk, D.U.; Gokcimen, A. Effect of tenoxicam on rat liver tissue. Turk. J. Gastroenterol. 2010, 21, 146–152.
  54. Matsumoto, R.; Tsuda, M.; Yoshida, K.; Tanino, M.; Kimura, T.; Nishihara, H.; Abe, T.; Shinohara, N.; Nonomura, K.; Tanaka, S. Aldo-keto reductase 1C1 induced by interleukin-1β mediates the invasive potential and drug resistance of metastatic bladder cancer cells. Sci. Rep. 2016, 6, 34625.
  55. Shiiba, M.; Yamagami, H.; Yamamoto, A.; Minakawa, Y.; Okamoto, A.; Kasamatsu, A.; Sakamoto, Y.; Uzawa, K.; Takiguchi, Y.; Tanzawa, H. Mefenamic acid enhances anticancer drug sensitivity via inhibition of aldo-keto reductase 1C enzyme activity. Oncol. Rep. 2017, 37, 2025–2032.
  56. Soriano-Hernandez, A.D.; Madrigal-Pérez, D.; Galvan-Salazar, H.R.; Martinez-Fierro, M.L.; Valdez, L.; Gomez, F.E.; Vazquez-Vuelvas, O.F.; Olmedo-Buenrostro, B.A.; Guzman-Esquivel, J.; Rodriguez-Sanchez, I.P.; et al. Anti-inflammatory drugs and uterine cervical cancer cells: Antineoplastic effect of meclofenamic acid. Oncol. Lett. 2015, 10, 2574–2578.
  57. Caglar, S.; Altay, A.; Kuzucu, M.; Caglar, B. In Vitro Anticancer Activity of Novel Co(II) and Ni(II) Complexes of Non-steroidal Anti-inflammatory Drug Niflumic Acid Against Human Breast Adenocarcinoma MCF-7 Cells. Cell Biophys. 2021, 79, 729–746.
  58. Zhou, P.; Wu, M.; Ye, C.; Xu, Q.; Wang, L. Meclofenamic acid promotes cisplatin-induced acute kidney injury by inhibiting fat mass and obesity-associated protein-mediated m6A abrogation in RNA. J. Biol. Chem. 2019, 294, 16908–16917.
  59. Li, H.; Song, Y.; He, Z.; Chen, X.; Wu, X.; Li, X.; Bai, X.; Liu, W.; Li, B.; Wang, S.; et al. Meclofenamic Acid Reduces Reactive Oxygen Species Accumulation and Apoptosis, Inhibits Excessive Autophagy, and Protects Hair Cell-Like HEI-OC1 Cells from Cisplatin-Induced Damage. Front. Cell. Neurosci. 2018, 12, 139.
  60. Grande, F.; Giordano, F.; Occhiuzzi, M.; Rocca, C.; Ioele, G.; De Luca, M.; Ragno, G.; Panno, M.; Rizzuti, B.; Garofalo, A. Toward Multitasking Pharmacological COX-Targeting Agents: Non-Steroidal Anti-Inflammatory Prodrugs with Antiproliferative Effects. Molecules 2021, 26, 3940.
  61. Arai, Y.; Tanaka, K.-I.; Ushijima, H.; Tomisato, W.; Tsutsumi, S.; Aburaya, M.; Hoshino, T.; Yokomizo, K.; Suzuki, K.; Katsu, T.; et al. Low Direct Cytotoxicity of Nabumetone on Gastric Mucosal Cells. Dig. Dis. Sci. 2005, 50, 1641–1646.
  62. Frejborg, E.; Salo, T.; Salem, A. Role of Cyclooxygenase-2 in Head and Neck Tumorigenesis. Int. J. Mol. Sci. 2020, 21, 9246.
  63. Li, W.; Zhang, Z.; Wang, B.; Liang, N.; Zhou, Q.; Long, S. MicroRNA and cyclooxygenase-2 in breast cancer. Clin. Chim. Acta 2021, 522, 36–44.
  64. Zhu, F.S.; Chen, X.M.; Huang, Z.G.; Wang, Z.R.; Zhang, D.W.; Zhang, X. Rofecoxib augments anticancer effects by reversing intrinsic multidrug resistance gene expression in BGC-823 gastric cancer cells. J. Dig. Dis. 2010, 11, 34–42.
  65. Yu, L.; Chen, M.; Li, Z.; Wen, J.; Fu, J.; Guo, D.; Jiang, Y.; Wu, S.; Cho, C.-H.; Liu, S. Celecoxib Antagonizes the Cytotoxicity of Cisplatin in Human Esophageal Squamous Cell Carcinoma Cells by Reducing Intracellular Cisplatin Accumulation. Mol. Pharmacol. 2010, 79, 608–617.
  66. Arora, M.; Choudhary, S.; Singh, P.K.; Sapra, B.; Silakari, O. Structural investigation on the selective COX-2 inhibitors mediated cardiotoxicity: A review. Life Sci. 2020, 251, 117631.
  67. Sun, S.X.; Lee, K.Y.; Bertram, C.T.; Goldstein, J.L. Withdrawal of COX-2 selective inhibitors rofecoxib and valdecoxib: Impact on NSAID and gastroprotective drug prescribing and utilization. Curr. Med Res. Opin. 2007, 23, 1859–1866.
  68. Brenner, G.B.; Makkos, A.; Nagy, C.T.; Onódi, Z.; Sayour, N.V.; Gergely, T.G.; Kiss, B.; Görbe, A.; Sághy, É.; Zádori, Z.S.; et al. Hidden Cardiotoxicity of Rofecoxib Can be Revealed in Experimental Models of Ischemia/Reperfusion. Cells 2020, 9, 551.
  69. Atashbar, S.; Jamali, Z.; Khezri, S.; Salimi, A. Celecoxib decreases mitochondrial complex IV activity and induces oxidative stress in isolated rat heart mitochondria: An analysis for its cardiotoxic adverse effect. J. Biochem. Mol. Toxicol. 2021, 2021, e22934.
  70. Wu, F.; Wang, W.; Duan, Y.; Guo, J.; Li, G.; Ma, T. Effect of Parecoxib Sodium on Myocardial Ischemia-Reperfusion Injury Rats. Med. Sci. Monit. 2020, 27, e928205-1.
  71. Okamoto, K.; Saito, Y.; Narumi, K.; Furugen, A.; Iseki, K.; Kobayashi, M. Comparison of the nephroprotective effects of non-steroidal anti-inflammatory drugs on cisplatin-induced nephrotoxicity in vitro and in vivo. Eur. J. Pharmacol. 2020, 884, 173339.
  72. El-Kader, M.A.; Taha, R.I. Comparative nephroprotective effects of curcumin and etoricoxib against cisplatin-induced acute kidney injury in rats. Acta Histochem. 2020, 122, 151534.
More
Video Production Service