Next Article in Journal
Very Early Onset of Therapy-Related Acute Myeloid Leukemia with 11q23 Rearrangement Presenting with Unusual PET Findings after R-DA-EPOCH for Primary Mediastinal Large B-Cell Lymphoma
Previous Article in Journal
Long-Term Outcomes of Chronic Cough Reduction after Laparoscopic Nissen Fundoplication—A Single-Center Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Interactions of Analgesics with Cisplatin: Modulation of Anticancer Efficacy and Potential Organ Toxicity

1
Basic Health Sciences Department, Faculty of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh 11671, Saudi Arabia
2
Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt
3
Department of Surgery, Faculty of Medicine, Jazan University, Jazan 45142, Saudi Arabia
*
Author to whom correspondence should be addressed.
Medicina 2022, 58(1), 46; https://doi.org/10.3390/medicina58010046
Submission received: 1 December 2021 / Revised: 19 December 2021 / Accepted: 20 December 2021 / Published: 28 December 2021
(This article belongs to the Section Pharmacology)

Abstract

:
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. This article offers an overview of the reported literature on the interactions between analgesics and CDDP, paying special attention to possible mechanisms that modulate CDDP’s cytotoxic efficacy and potential adverse reactions.

1. Introduction

Cisplatin (CDDP) is a platinum-based agent that has long been used in the treatment of various types of malignancies [1]. Unfortunately, CDDP may cause toxic side effects on normal human tissues that might lead to multiple organ damage [2,3]. During chemotherapy, several patients may suffer from pain and are likely to take medications to relief it. According to the level of the pain, these medications may range from acetaminophen or non-steroidal anti-inflammatory drugs (NSAIDs), in the case of mild to moderate pain, reaching up to stronger pain killers such as narcotic analgesics in the case of severe pain. It is possible that administering these medications concomitantly with CDDP might augment CDDP-induced organ toxicity or alter its anticancer efficacy. To date, the interactions of analgesics with CDDP have not been fully reviewed. Here, data were collected from the literature to formulate an updated review of the molecular mechanisms that might be involved in the interactions of different types of analgesics with CDDP, and the implications of such interactions on rational use of analgesics for the treatment of pain during CDDP cancer chemotherapy in humans.

2. CDDP Efficacy and Toxicity

CDDP was originally created by M. Peyrone in 1844, and in 1893 its chemical composition (Figure 1) was first revealed by Alfred Werner [4]. This was followed by the accidental discovery of CDDP’s cytotoxic actions by Dr. Rosenborg in 1965 [5] and the approval of CDDP for medical use in 1978 [6]. Since then, this anticancer drug has been used to treat numerous neoplasms, including those of the testes, ovaries, uterus, breasts, bladder, gastrointestinal tract, lung, bone, and brain [4,6,7]. CDDP performs its anticancer actions through forming covalent intra-strand DNA adducts between its CDDP platinum complexes and the neoplastic cell DNA, which causes subsequent DNA damage and obstruction of efficient DNA repair, resulting in restriction of DNA synthesis and inhibition of tumor cell growth [8,9]. CDDP induces free-radical formation, especially reactive oxygen species (ROS) that can be the initial trigger of cancer-programmed cellular death: apoptosis. This is due to the induction of pro-apoptotic factors, such as Bax and Bid, and the dysregulation of anti-apoptotic factors, such as Bcl-2, as well as the activation of caspases, which result in an apoptotic cascade [10]. Unfortunately, CDDP, by the same mechanisms, may also affect normal tissues, resulting in morbid, and sometimes fatal, side effects. Nearly a quarter of patients treated with CDDP develop nephrotoxicity as a side effect, through epigenetic DNA methylation, histone modification, oxidative stress, inflammation, and apoptosis [11]. Similar mechanisms are involved in CDDP-induced hepatotoxicity [12,13], cardiotoxicity [14], gastrointestinal toxicity [15], and ototoxicity [16]. CDDP was also found to be neurotoxic [17], affecting mainly sensory nerves, inducing painful neuropathy as a side effect [18], which may be a strong indication for the use of analgesics concomitantly with CDDP to relief such pain.

3. Interactions of Acetaminophen with CDDP

Acetaminophen, also acknowledged as paracetamol, is a para-aminophenol derivative that may be used for the management of mild to moderate pain during CDDP anticancer treatment, as well as for treatment of CDDP-chemotherapy-related fever [19]. Since it lacks anti-inflammatory properties, acetaminophen is usually not considered as one of the NSAIDs. It was reported that acetaminophen may act as a chemo-enhancer that promotes the cytotoxic effect of CDDP on hepatocarcinoma and hepatoblastoma cells, by decreasing GSH and the induction of oxidative stress [20]. The same mechanism was seen when an acetaminophen/CDDP combination was administered to resistant atypical teratoid rhabdoid pediatric tumor cells [21] and human ovarian carcinoma [22]. Unfortunately, both acetaminophen and CDDP are considered hepato- and nephrotoxic [23], thus may be cautiously used concomitantly if the patient has kidney or liver function impairment.

4. 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]. 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]. 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]. 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]. NSAIDs that hold some potential to improve CDDP anticancer effects are summarized in Figure 2.
The NSAIDs can be subdivided into salicylates, propionic acids, acetic acids, enolic acids, anthranilic acids, naphthylalanine, and selective COX-2 inhibitors [24]. Due to their chemical diversity, NSAIDs show different levels of selectivity on inhibiting COX-1 and COX-2 enzymes [29]. In general, most non-selective NSAIDs are known to induce gastric ulceration [30], as well as having renal side effects including tubulointerstitial nephritis, nephrotic syndrome, acute kidney injury, and chronic kidney disease [31], whereas COX-2 selective NSAIDs may cause cardiovascular side effects [32]. Still, there are several exceptions. For example, the non-selective NSAID loxoprofen, might not harm the gastric mucosa as much as its peer NSAIDs [33]. Its derivative fluoro-loxoprofen, might even have gastroprotective effects [34]. Indomethacin, on the other hand, was reported to have the highest gastrotoxic potential [35].

4.1. Interactions of Salicylates with CDDP

Salicylate, the prototype of NSAIDs, has shown promising anticancer effects [36,37,38]. 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]. Other studies showed that salicylate also increased the anti-tumor effect of CDDP against osteosarcoma, through modulating the NF-κB pathway [40], and against non-small cell lung carcinoma stem-like cells by repressing migration through acting on the mTOR-Akt axis [41]. In addition, salicylate improved CDDP toxicity against colon cancer cells through preventing NF-κB binding to a COX-2 promoter [42] against lung cancer cells, by abrogating cancer cell stemness [43], against epithelial ovarian cancer cells by increasing p53 acetylation and promoting apoptosis [44], and against oesophageal squamous cell carcinoma through epigenetic modulation of chromatin by altering histone acetylation levels [45]. 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]. 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].

4.2. 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]. Some studies succeeded in synthesizing lipid encapsulated ibuprofen metallodrug nanoparticles to overcome CDDP chemoresistance in glioblastoma cancer cells [52]. 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]. In addition, combining ibuprofen with CDDP caused a higher cytotoxic effect on thyroid and pancreatic cancer cells in vitro [54]. Furthermore, an ibuprofen/CDDP combination reversed CDDP resistance in non-small-cell lung cancer through a COX-independent mechanism [55]. 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]. Unfortunately, through stimulating oxidative stress, ibuprofen might cause toxicity similar to CDDP on the kidneys and liver [57,58].
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]. Interestingly, ketoprofen was suggested to protect against CDDP-induced nephrotoxicity [60], which is in line with more recent findings that ketoprofen has no nephrotoxic effects [61]. Several trials were also performed to assess the anti-tumor effects of combining CDDP with a third propionic acid-derived NSAID, naproxen [62,63], 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]. Similar results were shown for a naproxen/CDDP combination on triple-negative breast cancer [65], as well as on ovarian endometrioid adenocarcinoma, lung adenocarcinoma, malignant pleural mesothelioma, and colon carcinoma cells [28]. Carprofen alone was able to ameliorate canine osteosarcoma in vitro [66]. Novel NSAIDs were created, such as derivatives of naproxen, flurbiprofen, and ibuprofen, that showed promising anticancer effect against cultured human glioblastoma cells [67], as well as human liver, breast, and colon carcinoma cells [68]. Whether the anticancer effects of these NSAIDS would be additive to that of CDDP or not still needs further investigation.

4.3. 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]. Sulindac could also ameliorate the growth rate of oral tumor cells and help their elimination by natural killer cells [70]. 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]. Interestingly, sulindac showed comparable cytotoxic effects to those of CDDP when tested on HEK293 cells [72]. Given together with CDDP, ketorolac succeeded in reversing CDDP chemo-resistance in a patient-derived cell xenograft model [73]. Diclofenac also showed improved CDDP anticancer effects against human lung adenocarcinoma CDDP-resistant cells [74,75]. To the contrary to what is expected from non-selective COX inhibitors, diclofenac did not deteriorate CDDP-induced nephrotoxicity [74]. Nevertheless, diclofenac, as with CDDP, had the hazard of causing hepatotoxicity as an adverse effect [76].

4.4. 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]. Interestingly, meloxicam protected the kidney from CDDP-induced renal lesions in mice [78]. 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]. Piroxicam, another enolic acid derivative of NSAIDs, when given as an adjuvant to CDDP-loaded nanoparticles, increased apoptosis in mesothelioma cells [80]. Unfortunately, unlike meloxicam, piroxicam was shown to worsen CDDP-induced nephrotoxicity in rats [81]. Tenoxicam alone seemed tolerable in patients with renal impairment [82], but was reported to have an injurious effect on the liver [83].

4.5. 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]. 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]. On their own, neither meclofenamic nor niflumic acid showed promising anticancer effects against uterine cervical cancer and breast adenocarcinoma cells, respectively [86,87]. Concerning safety, meclofenamic acid could aggravate CDDP-induced renal damage [88]. However, meclofenamic acid seemed to have the potential to protect against CDDP-induced ototoxicity via improving the viability of ear hair cells [89]. Nabumetone, a naphthylalanine derivative, had an antiproliferative effect on MCF-7 and MDA-MB-231 breast carcinoma cells [90], with low toxic effects on gastric mucosa cells [91].

4.6. 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]. Administered with CDDP, rofecoxib was reported to enhance cytotoxic effects on gastric cancer cells by down regulating multidrug resistance protein 1 expression [94]. Nevertheless, combining CDDP with celecoxib did not improve the anticancer activity of CDDP against human esophageal squamous cell carcinoma xenograft model in vivo [95]. Despite their safety with regards to gastric ulceration, selective COX-2 inhibitors were reported to mediate cardiotoxicity [32]. Indeed, several members of this group, such as valdecoxib and rofecoxib, were removed from the market due to their potential cardiovascular hazards [96,97]. Despite its reported hazard on cardiomyocytes [98], 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]. Celecoxib showed protective effect against CDDP-induced nephrotoxicity [100]. 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].

5. Interaction of Narcotic Analgesics with CDDP

Opioids have different impacts on cancer viability. Both morphine and fentanyl might promote cancer, while buprenorphine had no effect on cancer, and tramadol might ameliorate cancer by modulating the activity of natural killer cells [102]. Tramadol initiated apoptotic effects in colon cancer stem cells [103]. Still, tramadol might interfere with CDDP cytotoxicity via a different mechanism, as it suppresses gap junction activity [104]. It seems that opioids, especially μ- and κ-receptor agonists, suppressed natural killer cells cytotoxicity, promoting viability of cancer cells [105]. Indeed, fentanyl decreased the sensitivity of lung cancer cells to CDDP [106]. We have shown that morphine, the prototype agonist of opioid μ-receptor, also reduced the anticancer efficacy of CDDP on breast cancer cells [107]. An exception to this is methadone, another opioid μ-receptor agonist, that might enhance CDDP anticancer effects against bladder cancer [108], as well as head and neck cancer cells [109]. Regarding toxicity, we have previously reported the hazardous effects of morphine on CDDP-induced cardiotoxicity and hepatotoxicity [13,107]. Tapentadol was also reported to cause lung, heart, and neuronal toxicity [110], as well as hepatorenal toxic effects [111]. Further studies are needed to validate if tapentadol’s side effects would be cumulative to that of CDDP if taken together. Table 1 summarizes the effect of different analgesics on CDDP-induced toxicities.

6. Conclusions

Despite the absolute need for analgesics for the treatment of pain during cancer chemotherapy with CDDP, physicians should bear in mind the consequences of the combination of different analgesics on CDDP efficacy and toxicity. Rational evidence-based combinatorial therapy with CDDP and analgesics can provide enormous benefits in providing higher selectivity in targeting cancer cells and avoiding augmentation of the hazards of CDDP’s side effects. Still, it should be noted that the majority of available data concerning the interaction between CDDP and analgesics on the level of efficacy and toxicity were generally interpreted from in vitro or in vivo animal models. Future clinical studies are needed to verify the impact of the CDDP/analgesic interaction during actual patient chemotherapeutic settings.

Author Contributions

Conceptualization, A.E.-S. and Z.K.; data curation, A.E.-S.; writing—original draft preparation, A.E.-S.; writing—review and editing, A.E.-S.; supervision, Z.K.; project administration, A.E.-S.; funding acquisition, A.E.-S. and Z.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Deanship of Scientific Research at Princess Nourah bint Abdulrahman University, grant number 240/S/39.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ghosh, S. Cisplatin: The first metal based anticancer drug. Bioorg. Chem. 2019, 88, 102925. [Google Scholar] [CrossRef]
  2. Oun, R.; Moussa, Y.E.; Wheate, N.J. The side effects of platinum-based chemotherapy drugs: A review for chemists. Dalton Trans. 2018, 47, 6645–6653. [Google Scholar] [CrossRef]
  3. Qi, L.; Luo, Q.; Zhang, Y.; Jia, F.; Zhao, Y.; Wang, F. Advances in Toxicological Research of the Anticancer Drug Cisplatin. Chem. Res. Toxicol. 2019, 32, 1469–1486. [Google Scholar] [CrossRef]
  4. Dasari, S.; Tchounwou, P.B. Cisplatin in cancer therapy: Molecular mechanisms of action. Eur. J. Pharmacol. 2014, 740, 364–378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Rosenberg, B.; Van Camp, L.; Krigas, T. Inhibition of Cell Division in Escherichia coli by Electrolysis Products from a Platinum Electrode. Nature 1965, 205, 698–699. [Google Scholar] [CrossRef] [PubMed]
  6. McSweeney, K.; Gadanec, L.; Qaradakhi, T.; Ali, B.; Zulli, A.; Apostolopoulos, V. Mechanisms of Cisplatin-Induced Acute Kidney Injury: Pathological Mechanisms, Pharmacological Interventions, and Genetic Mitigations. Cancers 2021, 13, 1572. [Google Scholar] [CrossRef] [PubMed]
  7. Mu, Q.; Lv, Y.; Luo, C.; Liu, X.; Huang, C.; Xiu, Y.; Tang, L. Research Progress on the Functions and Mechanism of circRNA in Cisplatin Resistance in Tumors. Front. Pharmacol. 2021, 12, 709324. [Google Scholar] [CrossRef] [PubMed]
  8. Makovec, T. Cisplatin and beyond: Molecular mechanisms of action and drug resistance development in cancer chemotherapy. Radiol. Oncol. 2019, 53, 148–158. [Google Scholar] [CrossRef] [Green Version]
  9. Tchounwou, P.B.; Dasari, S.; Noubissi, F.K.; Ray, P.; Kumar, S. Advances in Our Understanding of the Molecular Mechanisms of Action of Cisplatin in Cancer Therapy. J. Exp. Pharmacol. 2021, 13, 303–328. [Google Scholar] [CrossRef] [PubMed]
  10. Mirzaei, S.; Hushmandi, K.; Zabolian, A.; Saleki, H.; Torabi, S.; Ranjbar, A.; SeyedSaleh, S.; Sharifzadeh, S.; Khan, H.; Ashrafizadeh, M.; et al. Elucidating Role of Reactive Oxygen Species (ROS) in Cisplatin Chemotherapy: A Focus on Molecular Pathways and Possible Therapeutic Strategies. Molecules 2021, 26, 2382. [Google Scholar] [CrossRef]
  11. Loren, P.; Saavedra, N.; Saavedra, K.; Zambrano, T.; Moriel, P.; Salazar, L. Epigenetic Mechanisms Involved in Cisplatin-Induced Nephrotoxicity: An Update. Pharmaceuticals 2021, 14, 491. [Google Scholar] [CrossRef] [PubMed]
  12. Rashid, N.A.; Halim, S.A.S.A.; Teoh, S.L.; Budin, S.B.; Hussan, F.; Ridzuan, N.R.A.; Jalil, N.A.A. The role of natural antioxidants in cisplatin-induced hepatotoxicity. Biomed. Pharmacother. 2021, 144, 112328. [Google Scholar] [CrossRef] [PubMed]
  13. El-Sheikh, A.A. P-Glycoprotein/ABCB1 Might Contribute to Morphine/Cisplatin-Induced Hepatotoxicity in Rats. Sci. Pharm. 2020, 88, 14. [Google Scholar] [CrossRef] [Green Version]
  14. Dugbartey, G.J.; Peppone, L.J.; de Graaf, I.A. An integrative view of cisplatin-induced renal and cardiac toxicities: Molecular mechanisms, current treatment challenges and potential protective measures. Toxicology 2016, 371, 58–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Shahid, F.; Farooqui, Z.; Khan, F. Cisplatin-induced gastrointestinal toxicity: An update on possible mechanisms and on available gastroprotective strategies. Eur. J. Pharmacol. 2018, 827, 49–57. [Google Scholar] [CrossRef]
  16. Gentilin, E.; Simoni, E.; Candito, M.; Cazzador, D.; Astolfi, L. Cisplatin-Induced Ototoxicity: Updates on Molecular Targets. Trends Mol. Med. 2019, 25, 1123–1132. [Google Scholar] [CrossRef] [PubMed]
  17. Stankovic, J.S.K.; Selakovic, D.; Mihailovic, V.; Rosic, G. Antioxidant Supplementation in the Treatment of Neurotoxicity Induced by Platinum-Based Chemotherapeutics—A Review. Int. J. Mol. Sci. 2020, 21, 7753. [Google Scholar] [CrossRef]
  18. Hopkins, H.L.; Duggett, N.; Flatters, S.J. Chemotherapy-induced painful neuropathy. Curr. Opin. Support. Palliat. Care 2016, 10, 119–128. [Google Scholar] [CrossRef] [Green Version]
  19. Eroglu, N.; Erduran, E.; Reis, G.P.; Bahadır, A. Chemotherapy-related fever or infection fever? Support. Care Cancer 2021, 29, 1859–1862. [Google Scholar] [CrossRef] [PubMed]
  20. Neuwelt, A.J.; Wu, Y.J.; Knap, N.; Losin, M.; Neuwelt, E.A.; Pagel, M.A.; Warmann, S.; Fuchs, J.; Czauderna, P.; Woźniak, M. Using Acetaminophen’s Toxicity Mechanism to Enhance Cisplatin Efficacy in Hepatocarcinoma and Hepatoblastoma Cell Lines. Neoplasia 2009, 11, 1003–1011. [Google Scholar] [CrossRef] [Green Version]
  21. Neuwelt, A.J.; Nguyen, T.; Wu, Y.J.; Donson, A.M.; Vibhakar, R.; Venkatamaran, S.; Amani, V.; Neuwelt, E.A.; Rapkin, L.B.; Foreman, N.K. Preclinical high-dose acetaminophen with N -acetylcysteine rescue enhances the efficacy of cisplatin chemotherapy in atypical teratoid rhabdoid tumors. Pediatr. Blood Cancer 2014, 61, 120–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Wu, Y.-J.J.; Neuwelt, A.J.; Muldoon, L.L.; Neuwelt, E.A. Acetaminophen enhances cisplatin- and paclitaxel-mediated cytotoxicity to SKOV3 human ovarian carcinoma. Anticancer Res. 2013, 33, 2391–2400. [Google Scholar] [PubMed]
  23. Sohail, N.; Hira, K.; Tariq, A.; Sultana, V.; Ehteshamul-Haque, S. Marine macro-algae attenuates nephrotoxicity and hepatotoxicity induced by cisplatin and acetaminophen in rats. Environ. Sci. Pollut. Res. 2019, 26, 25301–25311. [Google Scholar] [CrossRef]
  24. Ghlichloo, I.; Gerriets, V. Nonsteroidal Anti-Inflammatory Drugs (NSAIDs); StatPearls Publishing: Treasure Island, FL, USA, 2021; pp. 1–11. [Google Scholar]
  25. 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. [Google Scholar] [CrossRef] [PubMed]
  26. 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. [Google Scholar] [CrossRef]
  27. 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. [Google Scholar] [CrossRef]
  28. 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. [Google Scholar] [CrossRef] [Green Version]
  29. Walker, C. Are All Oral COX-2 Selective Inhibitors the Same? A Consideration of Celecoxib, Etoricoxib, and Diclofenac. Int. J. Rheumatol. 2018, 2018, 1–12. [Google Scholar] [CrossRef]
  30. Morsy, M.; El-Sheikh, A. Prevention of Gastric Ulcers. In Peptic Ulcer Disease; IntechOpen: London, UK, 2011; pp. 437–460. [Google Scholar]
  31. Drożdżal, S.; Lechowicz, K.; Szostak, B.; Rosik, J.; Kotfis, K.; Machoy-Mokrzyńska, A.; Białecka, M.; Ciechanowski, K.; Gawrońska-Szklarz, B. Kidney damage from nonsteroidal anti-inflammatory drugs—Myth or truth? Review of selected literature. Pharmacol. Res. Perspect. 2021, 9, e00817. [Google Scholar] [CrossRef]
  32. 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. [Google Scholar] [CrossRef]
  33. Yamakawa, N.; Suemasu, S.; Kimoto, A.; Arai, Y.; Ishihara, T.; Yokomizo, K.; Okamoto, Y.; Otsuka, M.; Tanaka, K.-I.; Mizushima, T. Low Direct Cytotoxicity of Loxoprofen on Gastric Mucosal Cells. Biol. Pharm. Bull. 2010, 33, 398–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Suemasu, S.; Yamakawa, N.; Ishihara, T.; Asano, T.; Tahara, K.; Tanaka, K.-I.; Matsui, H.; Okamoto, Y.; Otsuka, M.; Takeuchi, K.; et al. Identification of a unique nsaid, fluoro-loxoprofen with gastroprotective activity. Biochem. Pharmacol. 2012, 84, 1470–1481. [Google Scholar] [CrossRef] [PubMed]
  35. Hall, A.; Tripp, M.; Howell, T.; Darland, G.; Bland, J.; Babish, J. Gastric mucosal cell model for estimating relative gastrointestinal toxicity of non-steroidal anti-inflammatory drugs. Prostaglandins Leukot. Essent. Fat. Acids 2006, 75, 9–17. [Google Scholar] [CrossRef] [PubMed]
  36. 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. [Google Scholar] [CrossRef] [Green Version]
  37. 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. [Google Scholar] [CrossRef] [PubMed]
  38. 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. [Google Scholar] [CrossRef] [PubMed]
  39. 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. [Google Scholar] [CrossRef] [PubMed]
  40. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. 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. [Google Scholar] [CrossRef]
  42. 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. [Google Scholar] [CrossRef]
  43. Zhao, M.; Wang, T.; Hui, Z. Aspirin overcomes cisplatin resistance in lung cancer by inhibiting cancer cell stemness. Thorac. Cancer 2020, 11, 3117–3125. [Google Scholar] [CrossRef] [PubMed]
  44. 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. [Google Scholar] [CrossRef] [PubMed]
  45. 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. [Google Scholar] [CrossRef]
  46. 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. [Google Scholar] [CrossRef]
  47. 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. [Google Scholar] [CrossRef]
  48. 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. [Google Scholar] [CrossRef]
  49. 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. [Google Scholar]
  50. 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. [Google Scholar] [CrossRef] [PubMed]
  51. 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. [Google Scholar] [CrossRef]
  52. 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. [Google Scholar] [CrossRef] [PubMed]
  53. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. 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. [Google Scholar] [CrossRef]
  56. 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. [Google Scholar] [CrossRef] [Green Version]
  57. 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. [Google Scholar] [CrossRef] [PubMed]
  58. Kim, M.; Lee, E.J.; Lim, K.-M. Ibuprofen Increases the Hepatotoxicity of Ethanol through Potentiating Oxidative Stress. Biomol. Ther. 2021, 29, 205–210. [Google Scholar] [CrossRef] [PubMed]
  59. 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. [Google Scholar] [CrossRef]
  60. 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. [Google Scholar] [CrossRef]
  61. 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. [Google Scholar] [CrossRef] [PubMed]
  62. 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. [Google Scholar] [CrossRef]
  63. 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. [Google Scholar] [CrossRef]
  64. 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. [Google Scholar] [CrossRef]
  65. 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. [Google Scholar] [CrossRef] [PubMed]
  66. 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. [Google Scholar] [CrossRef] [Green Version]
  67. 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. [Google Scholar] [CrossRef]
  68. 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. [Google Scholar] [CrossRef] [PubMed]
  69. 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. [Google Scholar] [CrossRef]
  70. 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. [Google Scholar] [CrossRef]
  71. 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. [Google Scholar] [CrossRef]
  72. Machkalyan, G.; Hèbert, T.E.; Miller, G.J. PPIP5K1 Suppresses Etoposide-triggered Apoptosis. J. Mol. Signal. 2016, 11, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. 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. [Google Scholar] [CrossRef] [PubMed]
  74. 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. [Google Scholar] [CrossRef]
  75. 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. [Google Scholar] [CrossRef]
  76. 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. [Google Scholar] [CrossRef]
  77. 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. [Google Scholar] [CrossRef]
  78. 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. [Google Scholar] [CrossRef]
  79. 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. [Google Scholar] [CrossRef] [PubMed]
  80. 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. [Google Scholar] [CrossRef] [PubMed]
  81. 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. [Google Scholar] [CrossRef] [PubMed]
  82. 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. [Google Scholar] [CrossRef] [PubMed]
  83. Karatopuk, D.U.; Gokcimen, A. Effect of tenoxicam on rat liver tissue. Turk. J. Gastroenterol. 2010, 21, 146–152. [Google Scholar] [CrossRef] [PubMed]
  84. 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. [Google Scholar] [CrossRef] [Green Version]
  85. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. 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. [Google Scholar] [CrossRef] [Green Version]
  87. 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. [Google Scholar] [CrossRef] [PubMed]
  88. 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. [Google Scholar] [CrossRef] [PubMed]
  89. 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. [Google Scholar] [CrossRef]
  90. 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. [Google Scholar] [CrossRef]
  91. 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. [Google Scholar] [CrossRef]
  92. Frejborg, E.; Salo, T.; Salem, A. Role of Cyclooxygenase-2 in Head and Neck Tumorigenesis. Int. J. Mol. Sci. 2020, 21, 9246. [Google Scholar] [CrossRef]
  93. 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. [Google Scholar] [CrossRef] [PubMed]
  94. 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. [Google Scholar] [CrossRef]
  95. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. 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. [Google Scholar] [CrossRef] [PubMed]
  97. 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. [Google Scholar] [CrossRef] [Green Version]
  98. 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. [Google Scholar] [CrossRef]
  99. 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. [Google Scholar] [CrossRef]
  100. 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. [Google Scholar] [CrossRef] [PubMed]
  101. 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. [Google Scholar] [CrossRef] [PubMed]
  102. Boland, J.W.; Pockley, A. Influence of opioids on immune function in patients with cancer pain: From bench to bedside. Br. J. Pharmacol. 2017, 175, 2726–2736. [Google Scholar] [CrossRef]
  103. Özgürbüz, U.; Gencür, S.; Kurt, F.Ö.; Özkalkanlı, M.; Vatansever, H.S. The effects of tramadol on cancer stem cells and metabolic changes in colon carcinoma cells lines. Gene 2019, 718, 144030. [Google Scholar] [CrossRef] [PubMed]
  104. He, B.; Tong, X.; Wang, L.; Wang, Q.; Ye, H.; Liu, B.; Hong, X.; Tao, L.; Harris, A. Tramadol and Flurbiprofen Depress the Cytotoxicity of Cisplatin via Their Effects on Gap Junctions. Clin. Cancer Res. 2009, 15, 5803–5810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Maher, D.P.; Walia, D.; Heller, N.M. Suppression of Human Natural Killer Cells by Different Classes of Opioids. Anesthesia Analg. 2019, 128, 1013–1021. [Google Scholar] [CrossRef] [PubMed]
  106. Yao, J.; Ma, C.; Gao, W.; Liang, J.; Liu, C.; Yang, H.; Yan, Q.; Wen, Q. Fentanyl induces autophagy via activation of the ROS/MAPK pathway and reduces the sensitivity of cisplatin in lung cancer cells. Oncol. Rep. 2016, 36, 3363–3370. [Google Scholar] [CrossRef]
  107. El-Sheikh, A.A.K.; Khired, Z. Morphine Deteriorates Cisplatin-Induced Cardiotoxicity in Rats and Induces Dose-Dependent Cisplatin Chemoresistance in MCF-7 Human Breast Cancer Cells. Cardiovasc. Toxicol. 2021, 21, 553–562. [Google Scholar] [CrossRef]
  108. Michalska, M.; Schultze-Seemann, S.; Kuckuck, I.; Katzenwadel, A.; Wolf, P. Impact of Methadone on Cisplatin Treatment of Bladder Cancer Cells. Anticancer Res. 2018, 38, 1369–1375. [Google Scholar] [CrossRef]
  109. Landgraf, V.; Griessmann, M.; Roller, J.; Polednik, C.; Schmidt, M. DL-Methadone as an Enhancer of Chemotherapeutic Drugs in Head and Neck Cancer Cell Lines. Anticancer Res. 2019, 39, 3633–3639. [Google Scholar] [CrossRef]
  110. Barbosa, J.; Faria, J.; Garcez, F.; Leal, S.; Afonso, L.; Nascimento, A.; Moreira, R.; Pereira, F.; Queirós, O.; Carvalho, F.; et al. Repeated Administration of Clinically Relevant Doses of the Prescription Opioids Tramadol and Tapentadol Causes Lung, Cardiac, and Brain Toxicity in Wistar Rats. Pharmaceuticals 2021, 14, 97. [Google Scholar] [CrossRef]
  111. Barbosa, J.; Faria, J.; Garcez, F.; Leal, S.; Afonso, L.; Nascimento, A.; Moreira, R.; Queirós, O.; Carvalho, F.; Dinis-Oliveira, R. Repeated Administration of Clinical Doses of Tramadol and Tapentadol Causes Hepato- and Nephrotoxic Effects in Wistar Rats. Pharmaceuticals 2020, 13, 149. [Google Scholar] [CrossRef]
Figure 1. Chemical structure of cisplatin. Two neutral ammonia (NH3) ligands and two chloride (Cl) anions are coordinated to the central platinum (Pt) ion.
Figure 1. Chemical structure of cisplatin. Two neutral ammonia (NH3) ligands and two chloride (Cl) anions are coordinated to the central platinum (Pt) ion.
Medicina 58 00046 g001
Figure 2. 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.
Figure 2. 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.
Medicina 58 00046 g002
Table 1. Effect of analgesics on organ toxicity that may deteriorate or protect against cisplatin-induced organ/tissue damage.
Table 1. Effect of analgesics on organ toxicity that may deteriorate or protect against cisplatin-induced organ/tissue damage.
Name of NSAIDOrgan/TissueEffectType of ExperimentRef.
AcetaminophenKidneyNephrotoxicityAnimal study (rat)[23]
LiverHepatotoxicity
NSAIDs 1KidneyNephro-protectiveAnimal study (rat)[48]
1. SalicylateAuditory systemProtect against ototoxicityHuman study[49]
NeuronsNeuro-protectiveIn vitro[50]
2. Propionic acid-derived NSAIDs
Fluoro-loxoprofenStomachGastroprotectiveAnimal study (rats)[34]
IbuprofenKidneyNephrotoxicityAnimal study (rat)[57]
Liver cellsHepatotoxicityIn vitro[58]
KetoprofenKidneyNephro-protectiveAnimal studies (rat and pig)[60,61]
3. Acetic acid-derived NSAIDS
IndomethacinStomach cellsGastric ulcerationIn vitro[35]
DiclofenacKidneyNephrotoxicityHuman (review)[31]
LiverHepatotoxicityAnimal study (rat)[76]
4. Enolic acid-derived NSAIDs
MeloxicamKidneyNephroprotectiveAnimal study (mouse)[78]
PiroxicamStomachGastric ulcerationHuman (review)[30]
KidneyNephrotoxicityAnimal study (rat)[81]
TenoxicamLiverHepatotoxicityAnimal study (rat)[83]
5. Anthranilic acid-derived NSAIDs
Meclofenamic acid KidneyNephrotoxicityAnimal study (mouse) and in vitro[88]
Cochlear hair cellProtect against ototoxicityIn vitro[89]
6. COX-II 2 selective NSAIDS
ValdecoxibHeartCardiotoxicityHuman (review)[96]
RofecoxibHeartCardiotoxicityAnimal study (rat)[97]
CelecoxibCardiomyocytesCardiotoxicityIn vitro[98]
KidneyNephroprotectiveAnimal study (rat) and in vitro[100]
ParecoxibHeartCardio-protectiveAnimal study (rat)[99]
Narcotic analgesics
MorphineHeartCardiotoxicityAnimal study (rat)[13]
LiverHepatotoxicityAnimal study (rat)[107]
TapentadolLung, heart, and neuronsLung, heart, and neuronal toxicitiesAnimal study (rat)[110]
Liver, KidneyHepato- and nephrotoxicityAnimal study (rat)[111]
1 NSAIDs; non-steroidal anti-inflammatory drugs, 2 COX-II; cyclooxygenase-II.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

El-Sheikh, A.; Khired, Z. Interactions of Analgesics with Cisplatin: Modulation of Anticancer Efficacy and Potential Organ Toxicity. Medicina 2022, 58, 46. https://doi.org/10.3390/medicina58010046

AMA Style

El-Sheikh A, Khired Z. Interactions of Analgesics with Cisplatin: Modulation of Anticancer Efficacy and Potential Organ Toxicity. Medicina. 2022; 58(1):46. https://doi.org/10.3390/medicina58010046

Chicago/Turabian Style

El-Sheikh, Azza, and Zenat Khired. 2022. "Interactions of Analgesics with Cisplatin: Modulation of Anticancer Efficacy and Potential Organ Toxicity" Medicina 58, no. 1: 46. https://doi.org/10.3390/medicina58010046

Article Metrics

Back to TopTop