Next Article in Journal
Targeted Therapy and Immunotherapy in Early-Stage Non-Small Cell Lung Cancer: Current Evidence and Ongoing Trials
Next Article in Special Issue
Diclofenac Sensitizes Signet Ring Cell Gastric Carcinoma Cells to Cisplatin by Activating Autophagy and Inhibition of Survival Signal Pathways
Previous Article in Journal
The Influence of Proteins on Fate and Biological Role of Circulating DNA
Previous Article in Special Issue
Nimodipine Treatment Protects Auditory Hair Cells from Cisplatin-Induced Cell Death Accompanied by Upregulation of LMO4
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pro-Inflammatory Signalling PRRopels Cisplatin-Induced Toxicity

Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(13), 7227; https://doi.org/10.3390/ijms23137227
Submission received: 31 May 2022 / Revised: 26 June 2022 / Accepted: 27 June 2022 / Published: 29 June 2022
(This article belongs to the Special Issue The Discovery and Development of Cisplatin)

Abstract

:
Cisplatin is a platinum-based chemotherapeutic that has long since been effective against a variety of solid-cancers, substantially improving the five-year survival rates for cancer patients. Its use has also historically been limited by its adverse drug reactions, or cisplatin-induced toxicities (CITs). Of these reactions, cisplatin-induced nephrotoxicity (CIN), cisplatin-induced peripheral neuropathy (CIPN), and cisplatin-induced ototoxicity (CIO) are the three most common of several CITs recognised thus far. While the anti-cancer activity of cisplatin is well understood, the mechanisms driving its toxicities have only begun to be defined. Most of the literature pertains to damage caused by oxidative stress that occurs downstream of cisplatin treatment, but recent evidence suggests that the instigator of CIT development is inflammation. Cisplatin has been shown to induce pro-inflammatory signalling in CIN, CIPN, and CIO, all of which are associated with persisting markers of inflammation, particularly from the innate immune system. This review covered the hallmarks of inflammation common and distinct between different CITs, the role of innate immune components in development of CITs, as well as current treatments targeting pro-inflammatory signalling pathways to conserve the use of cisplatin in chemotherapy and improve long-term health outcomes of cancer patients.

1. Introduction

Cis-diaminedichloroplatinum (II), or cisplatin, is a powerful chemotherapeutic agent that has been in use for decades to treat a multitude of cancers alone or in combination therapies. Its ability to inhibit cellular division was first discovered by Dr. Burnett Rosenburg in 1965—120 years after it was first synthesised—it was licensed for medical use in chemotherapy shortly after, in 1970 [1,2,3]. As the first platinum-based antineoplastic drug approved by the FDA, it has since then pioneered research and development into countless other transition-metal based chemotherapies [4]. Specifically, the anti-tumoural properties of cisplatin were linked to its ability to restrict cell division through its intercalation into the DNA of reproducing cells [5,6,7,8]. Cisplatin is aquated when it enters the cell and attacks the purine bases in the tumoural DNA, with particular affinity for the N7 of guanine. The formation of intra-strand adducts by covalently binding adjacent guanine or adenosine bases was found to be strongly associated with its cytotoxic effects, though inter-strand adducts are also formed with less frequency, as shown in Figure 1. This distorts the DNA structure and recruits mismatch repair machinery to the nucleus, but ultimately an inability for cells to repair their DNA induces oxidative stress, cell cycle arrest, and initiation of pro-apoptotic pathways [5,6,7]. This mechanism of action allows it to target tumour cells with particular preference due to the accessibility of replicating DNA and high basal levels of reactive oxygen species. The simplicity and efficacy of its anti-cancer activity has allowed cisplatin to become an indispensable drug in cancer therapy, contributing to improved survival rates in solid-state cancers. Since then, more and more attention has shifted to the adverse drug reactions caused by cisplatin chemotherapy. Despite its efficacy in treating solid-state cancers ranging from the head-and-neck [9,10], to ovarian [11,12], and testicular [13,14], and several others [15,16], especially childhood-related cancers [17,18,19,20,21,22,23], the toxicity of cisplatin has put serious limitations on its clinical use as well as the long-term health outcomes and quality of life of cancer survivors. The adverse drug reactions caused by cisplatin and its toxicity profile have led to the development of several cisplatin-induced toxicities (CITs) [24,25,26], which appear to target specific parts of the body such as the kidneys, liver, neurons, and inner ear. This is due to the preferential accumulation of cisplatin in these regions following repeated intravenous administration during treatment [15,27,28]. As such, some of the most commonly reported CITs include cisplatin-induced nephrotoxicity (CIN) [27,29,30], cisplatin-induced hepatotoxicity (CIH) [31,32], cisplatin-induced peripheral-neurotoxicity (CIPN) [33,34,35], and cisplatin-induced ototoxicity (CIO) [36,37,38].
Over the years, several methods have been developed to alleviate the effects of cisplatin, though none have proven to be effective as a generic countermeasure against all forms of CIT due to the diverse nature of its activity and differences in responses to cisplatin at distinct target sites.
This has led to two primary branches of investigation: one which seeks to develop drugs related to cisplatin which are intrinsically less toxic (such as carboplatin and oxaliplatin), and one which seeks to identify the underlying mechanisms driving the development of CITs so that they may be specifically targeted and inhibited, ideally without affecting treatment efficacy; see Figure 2 for a summary of specific signalling pathways activated by cisplatin. The mechanism-based studies seek to preserve the use of cisplatin in chemotherapy and potentiate its use in clinic, but most research seeking to prevent CITs has been focused on the nature and impact of reactive oxygen and nitrogen species, which are known to be the downstream damage-inducing agents in CITs; however, this branch of research does not focus on identifying the actual instigators of cisplatin-induced damage and subsequent oxidative stress. Instead, there has been rising interest in another phenomenon that has appeared to be common amongst nearly all forms of CIT: inflammation. Hallmarks of inflammation have been used to gauge the severity of CITs since they were first characterised, and the role of cisplatin-induced inflammation as a direct initiator of the damage associated with CIT development has grown to be an especially hot topic of research; see Figure 3 for a summary of the inflammatory mechanisms contributing to each major CIT. Increasingly, natural and synthetic anti-inflammatory compounds with the potential to significantly ameliorate CITs have become more prominent and promising.
In this review, we highlighted the immunological traits of cisplatin-induced toxicities and covered the importance of pro-inflammatory signalling systems in their development, especially through the innate immune system. We also discussed the prospects of anti-inflammatory agents as protective combinatorial therapies centred on immunomodulation to curtail CITs and preserve and potentiate the use of cisplatin in cancer therapy.

Inflammation and Pro-Inflammatory Signalling

Immune responses can be classified as either ‘innate’ or ‘adaptive’. Adaptive immune responses are activated in response to persistent infection and/or damage; they are tailored to facilitate responses to specific forms of antigens and are carried out by adaptive immune cells, such as helper and cytotoxic T-cells, and antibody-producing B-cells. Innate immune responses are, conversely, immediate, non-antigen-specific, responses that can be triggered by the vast majority of cells and mediated by myeloid phagocytic cells. These include, but are not limited to, neutrophils, macrophages, and monocytes [39,40].
The process of inflammation is predominantly handled by the systems associated with the innate immune system which rely on ‘input signals’ generated by pattern-recognition receptors (PRRs), as well as cytokines and chemokines and their respective unique receptors.
PRRs bind common pathogen-associated molecular patterns (PAMPs) and initiate downstream signalling cascades that culminate in the expression of genes required to recruit dedicated immune cells and kickstart inflammation. Often, these PRRs also double as damage-recognition receptors, in that they can also bind and trigger responses to host cellular damage-associated molecular patterns (DAMPs), such as extracellular genomic DNA and mitochondrial DNA (mtDNA), and heat shock proteins. PRRs can be classified into four main categories, or families: (1) Toll-Like Receptors (TLRs), (2) Nucleotide-Binding Oligomerisation Domain (NOD)-Like Receptors (NLRs), (3) Retinoic-Acid-Inducible-Gene (RIG)-Like Receptors (RLRs), and C-Type Lectin Receptors (CLRs) [41].
TLRs localise to the cell surface or intracellular compartments and are thus prime to detect extracellular or vesicular signs of pathogens, such as bacterial lipopolysaccharides (LPS) [42]. They are also responsible for the detection of most DAMPs when paired with specific accessory proteins or coreceptors [43]. Certain TLRs have also been implicated in the development of unique hypersensitivity/allergic reactions [44,45,46,47]. NLRs, conversely, are responsible for the recognition of intracellular signifiers of infection, such as bacterial peptidoglycan components (iE-DAP), bacterial toxins, and viral nucleic acid structures [48,49]. Upon exposure to any of these potential agonists, NLRs trigger downstream signals that reflect that of TLRs but also facilitate the creation of complexes known as inflammasomes that can additionally modulate/control cell survival/death systems [50,51,52]. Like NLRs, RLRs are also responsible for detecting intracellular pathogens, but are equipped to specifically bind common genetic elements of both viral and intracellular bacterial pathogens [53,54,55]. CLRs, on the other hand, are more akin to TLRs, binding pathogen-associated carbohydrates from the extracellular space to cause pro-inflammatory gene expression changes identical to that of most other PRRs. Unlike TLRs, however, CLRs have a more direct role in arbitrating the transition from innate to adaptive immune responses by using captured antigens for presentation and provision to dendritic cells that can explicitly activate adaptive immune cells [56,57].
The Toll-like Receptors each bind a specific array of PAMPs and DAMPs and, through several unique and shared downstream signalling systems, activate three key transcription factors, nuclear factor kappa B (NF-κB), activator protein-1 (AP-1), and interferon regulatory factor 3, (IRF3), responsible for enabling the expression and secretion of soluble pro-inflammatory signalling molecules such as cytokines and chemokines.
Most, if not all forms of PRR, conclude in the activation of at least one of three key gene transcription factors: (1) AP1, (2) NF-κB, and (3) IRFs. It is through these three transcription factors that the state of inflammation can be modulated [58]. AP1 consists of homo- or hetero-dimeric complexes which consist of four types of DNA-binding proteins: Jun-family proteins, Fos-family proteins, activating transcription factors/cAMP response element binding protein (ATF/CREB)-family proteins, and musculoaponeurotic fibrosarcoma (MAF) family proteins. Different combinations of these subunits, in the context of different cells and conditions, can lead to distinct gene expression profiles, including the upregulation of pro-inflammatory cytokines and chemokines [59,60,61]. NF-κB can similarly mediate the upregulation of pro-inflammatory cytokines and chemokines, and can be considered the primary regulator of inflammation given that it can play a pivotal role in micromanaging both innate and adaptive immune responses. That said, NF-κB is also the ‘master regulator’ for countless other homeostatic genes, allowing it to also alter cell cycle progression in conjunction with inflammasomes and other inflammatory factors, activate dedicated immune cells to manipulate their maturation and differentiation processes, and influence the expression of tertiary features with additional roles in inflammation, such as adhesion molecules [62,63,64]. Juxtaposed to the prior two transcription factors, IRF3 contributes to inflammation by promoting the secretion of a unique class of cytokines, called Type I Interferons. Unlike AP1 and NF-κB, which are targets shared between almost all PRR downstream signalling mechanisms, IRF3 is under the purview of only a specific subset of PRRs—namely, TLR members, TLR3 and TLR4, as well as RLRs. Its activity is also under the control of a limited set of non-PRR, DAMP-specific, pro-inflammatory complexes such as cGAS-STING [65,66].
Cytokines and chemokines, unlike PRRs that centre on ‘paracrine’ interactions, can operate as autocrine and endocrine means of cellular communication, and can direct the development of inflammation through self-reinforcing signals and long-distance signalling. Both cytokines and chemokines are small proteins that can vary wildly in function and can either be pro-inflammatory or anti-inflammatory. Typically, cytokines incite their own set of downstream signalling events to alter gene expression while chemokines mainly operate as chemoattractants—recruiting different subsets of immune cells.
Pro-inflammatory cytokines and chemokines in particular are produced and released as a result of PRR signalling cascades—each PRR family is responsible for the production and release of specific cytokines and chemokines. The most common pro-inflammatory cytokines are the interleukins (IL), IL-1β and IL-6 especially, as well as Tumour Necrosis Factor-α (TNF-α). IL-1β has been identified as a critical factor in the development of fevers and the pain responses associated with inflammation. IL-6 has been shown to activate acute phase responses (APRs)—a set of changes to serum protein concentrations—intended to render adjacent areas inhospitable to any pathogens present. TNF-α, on the other hand, helps protect against intracellular threats by making host cells inhospitable environments and contributing to pro-apoptotic signalling systems.
The most common chemokines in comparison tend to be CXC-motif chemokine 2/keratinocyte-derived chemokine (CXCL2/KC) and CXC-motif chemokine 8/interleukin-8 (CXCL8/IL-8), which control the recruitment of neutrophils as T-cells to sites of infection/inflammation [67,68]. ‘Positive’ acute-phase proteins are proteins that are increased to promote inflammation, and include, but are not limited to complement factors, degradative enzymes, and iron chelators, all of which can also hurt the host with prolonged exposure [69,70].

2. Pattern Recognition Receptors (PRRs) in Cisplatin-Induced Toxicities

2.1. Toll-Like Receptors (TLRs)

Toll-Like Receptors (TLRs) have been one of several features of immunology heavily studied in association with cisplatin-induced toxicities. Of the ten different members of the TLR family, two appear to have a major influence on the progression of CITs and the damage they can ultimately do: TLR4 and TLR2 (Figure 2).
TLR4 presents on the cell membrane as a homo-dimer and is special in that it can transduce signalling through two intracellular pathways associated with TLRs: Myeloid differentiation primary response 88 (MyD88) dependent and Myd88 independent pathways [71]. MyD88 is an adaptor molecule that allows for signal convergence following TLR stimulation and while other TLRs utilise one or the other pathway, TLR4 can use both, allowing for significant crossover and potentiation of signalling. It is well known for being stimulated by a bacterial PAMP called lipopolysaccharide, a component of gram-negative bacterial cell membranes, as well as some DAMPs with the help of various co-receptors. This allows it to detect bacterial infection as well as surrounding cell damage, and is necessary for stimulating a localised immune response and maturation of adaptive immune cells. It has also been shown to mediate allergic hypersensitivity reactions to transition metals, such as nickel, cobalt, and palladium [44,45,46,47]. Importantly, TLR4 is associated with co-receptors, myeloid differentiation factor 2 (MD2), and cluster of differentiation 14 (CD14), which are necessary for signal transduction in response to lipopolysaccharide [42]. In the context of CIO, TLR4 appears to play an important role in exacerbating inflammation in the inner ear. Inhibition of TLR4 chemically via small molecule inhibitors or through gene silencing has been shown to confer protection in the context of the three most prevalent CITs (CIN, CIPN and CIO), as well as rarer cases, such as cisplatin-induced hepatotoxicity (CIH) [72].
Though the exact mechanism(s) driving the relationship between CITs and TLR4 have yet to be completely elucidated, several aspects have been typified, and a number of theories proposed. For example, the activation of TLR4 mitogen-activated protein kinase (MAPK) pathway-associated proteins, c-Jun N-terminal kinase (JNK) and p38, appear to correspond with toxicity in CIN and inversely correlates with increased cell viability in TLR4-deficient model organisms [72]. In studies, bone marrow chimeric mice have been used to showcase the importance of localised cell TLR4 proteins compared to responding immune cell TLR4s [72]. Transforming growth factor-β-activated kinase 1 (TAK1), a key component of the TLR4 MyD88-dependent signalling pathway, has also shown promise as a potentially valuable target for protecting against CIN. The inhibition of TAK1 leads to reduced activation of the extracellular signal-regulated kinase (ERK) and p38 MAPK signalling pathways through TLR4—leading to a reduction in tubular damage in the kidneys [73,74].
In cases of CIO, compounds that block MAPK activation, akin to the calcium channel antagonist Flunarizine, have been shown to prevent the progression of downstream TLR4 functions as well [75]. Directed inhibition of ERK caused a significant reduction to typical levels of NF-κB activation and pro-inflammatory cytokine secretion—sufficient to increase cell viability [75]. It has also been demonstrated that LPS-mediated TLR4 activation has a synergistic effect on CIO and that cisplatin induces the upregulation of TLR4, increasing the extent of toxicity and subsequent hearing loss [76]. This implies that TLR4-mediated induction of inflammation may be a separately looping but nevertheless contributing factor to CITs. This same principle has been found to apply to CIN and endotoxin insult and septic shock [77].
On the other hand, numerous studies exist indicating that cisplatin-induced pro-inflammatory responses can develop even in presumptively sterile conditions and in an MD2-independent manner. In 2021, Babolmorad et al. demonstrated that cisplatin-induced pro-inflammatory cytokine secretion could be induced in human embryonic kidney cells that express TLR4 but do not express MD2 [78]. The inhibition of TLR4 activation through commercially available chemical inhibitors like TAK242, and the prevention of TLR4 expression through siRNA and clustered regularly interspaced short palindromic repeats (CRISPR), all proved sufficient to reduce CIT-associated proinflammatory responses in the same system [78]. Moreover, it was also demonstrated that TLR4 may specifically interact with the platinum component of cisplatin to mediate inflammation as it has been shown to do so for metal allergens, suggesting that potentially direct interactions between cisplatin and TLR4 could be stimulating CIO; however further evidence is required to confirm this. In 2015, TLR4-deficient mice and MyD88/TRIF-deficient mice obtained significant to near-absolute protection from CIPN signified by mechanical allodynia (pain sensitivity) [79,80]. Transcriptomic analyses have identified seven genes that appear to dictate CIPN severity, almost all of which are linked to immunity, including TLR4 [81]. Ibudilast and isomers of the opioid antagonists, Naltrexone and Naloxone, have been shown to inhibit TLR4 activity and have all been shown to reduce chemotherapy-induced peripheral neuropathies similar to CIPN as well [82,83,84]. The opioid antagonists are believed to operate by co-opting MD2 to bind and block TLR4 [85]. Recent data suggests that Ibudilast can directly interact with and inhibit interleukin-1 receptor-associated kinase 1 (IRAK1) to inhibit the MyD88-dependent signalling pathway of TLR4—among several of its other indirectly anti-inflammatory modes of action as a phosphodiesterase inhibitor [86].
In contrast to TLR4, TLR2 has a protective role against CITs. TLR2 exists primarily on cell surface membranes such as TLR4, but is expressed exclusively and pre-emptively as part of heterodimers with either TLR1 or TLR6. TLR2 facilitates the recognition of and innate immune responses to a variety of multi-acylated lipopeptides as opposed to lipopolysaccharides. Multi-acylated lipopeptides, specifically di- and tri-acylated lipopeptides, can be found in bacteria, viruses, fungi, and even parasites, making TLR2 an extremely versatile PRR. TLR2 can nevertheless also mediate responses to typical DAMPs, such as heat shock proteins and high mobility group box 1 protein (HMGB1).
Like other TLRs, TLR2 depends on particular coreceptors to bolster the recognition of particular PAMPs and DAMPs. Along with CD14, CD36 also assists with TLR2 activation in the vast majority of responses. Most TLRs are also known to have one downstream signalling pathway in common—the MyD88-dependent downstream signalling pathway, and TLR2 is no exception. Upon binding a ligand, TLR2 initiates the MyD88-dependent downstream signalling cascade but differentiates itself from TLR4 in that it must undergo internalisation first to do so, and does not signal through the MyD88-independent signalling pathway [42,87]. At least two independent studies have shown that TLR2 activation can provide a considerable degree of protection from CITs. Removal of TLR2-enhanced CIN specifically, coinciding with a reduction in the markers of autophagy, and a shift to immunosuppressive cytokine regimens and adaptive immune responses by resident dendritic cells [88,89,90]. Renal stem cell recovery systems, which can limit cisplatin-induced acute kidney injuries, appear to be at least partly dependent on TLR2 activation as well [91]. Some studies have shown that depletion of TLR2 is associated with protection and subsequent reductions in pro-inflammatory IL-17A in CIN, and especially when there is simultaneous inhibition of TLR4 expression or activity. However, there are also studies that connote to the opposite [92,93]. Reports on its role in CITs are limited though and it poses an intriguing area for further investigation, particularly since it is not clear which heterodimer is more involved in this process—TLR2/TLR1 or TLR2/TLR6.
Little more is known about the connection between TLRs and CITs beyond that. The TLR4 and TLR2 agonists present or released following cisplatin treatment and responsible for dictating the course of cisplatin-induced toxicities have remained unknown. TLR9 has also recently drawn attention as new reports indicate that it too may play a role in CITs, albeit differently. TLR9, another TLR PRR, is canonically geared towards the recognition of DNA structures associated with pathogens (unmethylated CpG-DNA motifs), but it has also been shown to recognise DNA-related DAMPs and bacterial by-products and components [42,94]. As such, TLR9 typically exist in distinct types of endosomes that dictate the weighting of their signalling cascades towards either pro-inflammatory IRF-dependent interferons or the typical NF-κB-related repertoire of cytokines and chemokines [42]. In CIN, it surprisingly limits neutrophil invasion and mediates the recruitment of protective adaptive immune cells, T-Regulatory-Cells (TRegs) [95]. Cases of chemotherapeutic peripheral neuropathy not necessarily limited to just cisplatin alternatively suggest that TLR9 may instead exacerbate CIPN through the recruitment and activation of pro-inflammatory macrophages [96].
Regardless, while the link between specific TLRs and CITs appears to be robust, it is not absolute. Neither the inhibition of TLR4, nor the expression of TLR2, can guarantee complete protection from cisplatin-induced toxicities, suggesting that other factors are also at play, some of which may be immunological in nature, but not entirely dependent on TLRs alone.

2.2. NOD-Like Receptors (NLRs) and Inflammasomes

Similar to TLRs, nucleotide-binding and oligomerisation domain (NOD)-like receptors (NLRs) are also a type of pattern-recognition receptor family. Unlike TLRs, they operate exclusively in the cytosol and detect various pathogenic and damage signals as well as homeostatic disruptions in the cell to stimulate a signalling cascade and facilitate activation of the innate and adaptive immune systems [97,98]. The NLR family is further divided into sub-families, each of which are defined by their distinct N-terminal domains, leading to various overlapping signalling pathways that either activate transcriptional activators or lead to the assembly of inflammasomes, which are large protein complexes in the cytosol that catalyse apoptotic effector functions. In this review, we do not detail the different kinds of NLRs and their signal transduction but instead focus on NLRs that appear to be most closely associated with CIT development (Figure 2). For an extensive discussion on NLRs and their involvement in human disease, please see the excellent review by Zhong et al. (2013) [97].
Of the various types of NLRs in mammalian cells, the one that appears to be the most involved in CIT is the inflammasome-forming NLRP3. While many PRRs are specific to a certain subset of activating signals, NLRP3 signalling is induced by a wide array of PAMPs and DAMPs [99]. The activation of the NLRP3 inflammasome is complex and involves many mediators that lead to increased cell stress; however, in a simplified model, the sensing of cell stress by NLRP3 is typically preceded by a two-step process [97]. First, there must be an upregulation of NLRP3 and other inflammasome factors such as caspase 1, which carries out the effector functions of the NLRP3 inflammasome, and pro-IL-1β, which is the inactive form of a pro-inflammatory cytokine that is cleaved by caspase 1. This first step is called the priming step and occurs through activation of other PRRs and receptors in response to their respective PAMPs and DAMPs. Interestingly, some of these priming responses overlap with inflammatory signalling pathways we have seen to be involved in CIT development, including TLR4-LPS interactions or TNFα and IL-1β stimulation [97,100]. The priming leads to NFκB-mediated transcription of inflammasome components so that an inactive form of NLRP3 in the cytosol is prepared to respond to cell stress. The second step is the activation step of the NLRP3 inflammasome, and is mediated by various stimuli that disturb cell homeostasis such as changes in K+ and Cl efflux or lysosomal disruption, to name a few. The DAMPs or PAMPs involved in stimulating these cellular disturbances are quite diverse and are not known to interact directly with NLRP3 itself; rather, NLRP3 senses these changes through unknown mechanisms and becomes “activated”, leading to its oligomerization and initiation of inflammasome formation. The pyrin domain at its N-terminus allows for interaction with apoptosis-associated speck-like protein containing a CARD (caspase activation and recruitment domain) (ASC), an adaptor that can then interact with caspase 1 and complete the assembly of the inflammasome [101,102]. Ultimately, the inflammasome allows for caspase 1-mediated cleavage of the inactive form of IL-1β and IL-18 to active forms, as well as cleavage and release of the pore-forming gasdermin D, which leads to localised cytokine-mediated inflammation or cell death through pyroptosis, respectively.
The role of NLRP3 in CIT is most commonly reported in the kidneys, specifically owing to the similarity between the inflammatory profile of NLRP3-mediated diseases, ischemic (non-chemically induced) acute-kidney injury (AKI) and cisplatin-induced AKI [102,103,104]. This fostered interest in the role of NLRP3 in cisplatin-induced nephrotoxicity.
Cisplatin-induced renal injury is generally associated with an increase in NLRP3 inflammasome components, but there are conflicting reports about its significance. In a study investigating cisplatin-induced renal dysfunction, younger male C57BL/6 mice were found to have increased IL-1β, IL-18, ASC, caspase 1, as well as NLRP3 in the kidneys 3 days following treatment [105]. This study specifically looked at the role of a receptor, purinergic receptor P2X7 (P2X7R), in exacerbating CIN through NLRP3 activation. This receptor has been reported to induce NLRP3 inflammasome activation by responding to extracellular ATP, a DAMP, and inducing pore-formation and K+ efflux, acting as a component of the second activation step of NLRP3 stimulation [106,107]. Chemical inhibition of this receptor protected mice from renal dysfunction and injury, reduced levels of inflammasome components and pro-inflammatory cytokines, and protected them from oxidative stress and apoptosis. Thus, reduction of inflammasome components following cisplatin treatment was associated with protection against CIN. Similarly, several other reports found an increase in NLRP3 inflammasome activity following cisplatin treatment that was associated with kidney injury [108,109,110,111,112,113]. Specifically, they found increases in inflammasome components, pro-inflammatory cytokines like IL-1β, IL-18, TNF-α, or even increased pyroptotic activity. Stimulation of pyroptosis in CIN occurred through increased levels of Gasdermin D, a protein involved in mediating pyroptosis that is also a substrate of the NLRP3 inflammasome [111]. Cisplatin-induced NF-κB activity increased NLRP3 inflammasome components as well as pyroptotic activity in mouse kidneys, all of which could be ameliorated by vitamin D-induced downregulation of NF-κB. Many of these studies assess processes that occur upstream of NLRP3 activation such as cisplatin-induced mitochondrial dysfunction or NF-κB upregulation, which speaks to its role as a downstream effector of cisplatin-induced inflammation, but not necessarily a causative agent of CIN. The most direct evidence for NLRP3 involvement in CIN comes from NLRP3-specific inhibitor experiments that showed protection against CIN. Cisplatin-induced kidney injury was mitigated by chemical inhibition of NLRP3 with MCC950, demonstrating the role of the NLRP3 inflammasome in exacerbating CIN directly [114].
In contrast to this, one study found that NLRP3 knockout mice were not protected from cisplatin-induced AKI and there was little to no change in the pro-inflammatory cytokine profile in cisplatin-treated wild type versus knockout mice [115]. While they did find that caspase 1 knockout mice were protected against cisplatin-induced apoptosis and renal failure [116], follow up studies provided weak evidence that inhibition of NLRP3 activity protected against cisplatin-induced AKI [115]. Kim et al. (2013) also found an increase in ASC and caspase 1 in older male C57BL/6 mice after cisplatin treatment, indicating increased NLRP3 inflammasome activity; however, there was no significant increase in the NLRP3 protein itself in the mouse kidney [115].
Similarly, Kim et al. (2013) found no increase in IL-1β in-vivo and mouse macrophages did not show a cisplatin-induced increase in NLRP3 in-vitro, thereby discounting the idea that NLRP3 mediated damage could be coming from invading innate immune cells [115]. While the authors did find that the NLRP3 inflammasome appeared to be involved in ischemic AKI, they concluded that it has no role in cisplatin-induced AKI. Interestingly, NLRP1 (a different NLRP-inflammasome) and its inflammasome effector caspase, caspase 5, were shown to be increased in mouse kidneys following cisplatin treatment. This suggests a role for NLRP1 in cisplatin-induced AKI. Even more intriguing was their finding that NLRP3 knockout decreased NLRP1 levels following cisplatin treatment, implicating a dependence of NLRP1 on NLRP3 in CIN. Moreover, NLRP3 inflammasome activity is associated with various kinds of kidney injury [117,118,119]. While cisplatin may induce its activity, the role of the NLRP3 inflammasome in CIN may work in concert with the injury cisplatin causes in the kidney through other mechanisms.
Due to its reliance on several exogenous signals for activation, the NLRP3 inflammasome is in a position where its involvement in CITs is closely tied to other inflammatory pathways, including other PRRs. Namely, TLR4 stimulation leads to an NF-κB-mediated increase in NLRP3 in response to cisplatin treatment in kidneys [108]. Cisplatin induces upregulation of all of these factors, and stimulation of TLR4 allows priming of NLRP3 components that are later activated by cisplatin-induced stress. A proton-pump inhibitor called omeprazole, which should only affect the NLRP3 inflammasome activation portion of this signalling axis, was effective in ameliorating CIN and decreasing TLR4/NF-κB/NLRP3 levels. In this way, signal convergence from other PRRs like TLR4 may facilitate NLRP3-mediated CIN.
Along with CIN, cisplatin-induced liver toxicity is also associated with NLRP3 inflammasome activity, though this is far less studied. Cisplatin-treated rat liver has increased NLRP3 protein, IL-1β, and caspase 1 that is correlated with increased oxidative stress, inflammation, and liver injury [112,120]. These effects were reversed by compounds that are suggested to inhibit factors such as NF-κB and MAPK, both of which work upstream of NLRP3 and, in the case of NF-κB, are involved in priming of NLRP3 inflammasome components. While this demonstrates its involvement, the direct role of NLRP3 in exacerbating inflammation in CIH remains to be seen with specific inhibition of NLRP3 in hepatocytes or gene silencing.
While the abundance of evidence indicating its significance in CIN and CIH imply NLRP3 to be a fascinating target for protective therapy, suppression of NLRP3 has also been associated with increased tumour resistance to cisplatin [121]. Cisplatin-resistant tumour cells in non-small lung cancers had downregulated NLRP3 and upon upregulation of NLRP3, tumour cells were once again sensitised to cisplatin treatment. This means that, while it may very well be involved in exacerbation of CITs, it may not be an ideal target for protection during cancer therapy as it potentially compromises the efficacy of cisplatin’s anti-tumoural activity.

3. Pro-Inflammatory Messengers (Cytokines and Chemokines)

Resulting from the downstream signalling of PRRs and the consequent activation of gene transcription factors such as NF-κB are the pro-inflammatory cytokines and chemokines. Pro-inflammatory cytokines and chemokines, such as IL-1β, IL-6, IL-8, and TNF-α, essentially serve as easily quantifiable indicators of cisplatin-induced toxicities since they are expressed directly proportional to the severity of conditions (Figure 2). In all of the prior publications mentioned, increases in toxicity (such as with TLR2 inhibition) corresponded with increases in IL-6, IL-1β, and TNF-α by at least 20–50%, while protection from toxicity corresponded with reductions in secretion by up to 50–80%. This relationship is also directly related to the dose of cisplatin used and the exposure time.
All in all, the relationship between cytokine secretion and cisplatin toxicity does not appear to be linear either; pro-inflammatory cytokines appear to actively contribute to the pathology of prolonged cisplatin exposure in a positive-feedback loop. So et al. (2007) demonstrated that the provision of recombinant exogenous pro-inflammatory cytokines associated with CITs could elicit cell viability loss capable of accounting for up to 20% of the cell death associated with CIO [122]. More importantly, the group also showed that the targeted depletion of pro-inflammatory cytokines through blocking antibodies could sufficiently mitigate the death caused by cisplatin treatment, the most effective of which being anti-TNF-α [122]. In 2007, Zhang et al. used chimeric mouse models to demonstrate that the expression of TNF-α by immune cells residing in the kidney was critical for CIN development as well [123]. That said, the impact of IL-6 expression remains contentious in CIN, as Faubel et al. indicated that IL-6 deficiencies, alone, provide little to no respite from CIN [124]. Corroborating evidence came from Kim et al. (2011) who discovered that STAT6−/− mice produced far less of the characteristic three pro-inflammatory cytokines and were accordingly protected from CIO [125]. Like their predecessors, they reported that anti-IL-6 and anti-TNF-α provided the greatest resistance to cisplatin cytotoxicity. It may also be important to note that pro-inflammatory cytokines, like TNF-α, have the capacity to trigger conditions associated with CIPN—not just CIO—as well. Both the natural upregulation of pro-inflammatory cytokines by injured Schwann cells, and the injection of exogenous pro-inflammatory cytokines like TNF-α, have the potential to provoke mechanical allodynia and thermal hyperalgesia—common manifestations of CIPN [126,127]. In-vitro, the provision of TNF-α also confers a permanent degree of hyper-responsiveness that would presumably lead to pain, and hypersensitivity to subsequent immune factors [126,127]. This is likely due to the pleiotropic effector functions of both the IL-6 and TNF-α and their receptors. IL-6 receptors (IL6R/CD126/gp80) can exist as either membrane-bound or cytosolic receptors, though they typically exist in their soluble “trans-signalling” form in non-immune cells [128,129]. Soluble IL6R is primarily responsible for pro-inflammatory induction through the activation of Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) and Src-homology region 2 (SH2)-containing protein tyrosine phosphatase 2 (SHP2)/MAPK pathways which exert control over monocytic differentiation, vasculature, immune cell infiltration, and indirectly promote deleterious ROS production [129,130,131].
TNF-α receptors (TNFR) also come in two forms, TNFR1 and TNFR2, both of which drastically affect cisplatin toxicities. TNFR1 is considered one the principal drivers of pro-apoptotic signalling as a dedicated ‘Death Receptor’ equipped with a Death Domain that grants access to caspase activation cascades, while TNFR2 is associated with the propagation of inflammation through dedicated immune cell activity [132]. Inhibition of at least one TNFR is enough to confer significant protection from CIN, though the exact importance of each to toxicity remains contentious. Tsuruya et al. (2003) highlighted protection attributed to TNFR1 deficiency, but Ramesh and Reeves (2003) contrastingly reported that TNFR2 deficiency provided a greater degree of resistance to toxicity despite the upregulation of both receptors in murine models of CIN [133,134,135]. Therapeutics designed to mimic TNF receptors, such as etanercept, operate by sequestering available TNF-α from functional membrane forms, and have proven to be sufficient in reducing aspects of CIO in-vivo [136].
Note that IL-6, IL-1β, and TNF-α may be the most commonly used indicators of CIT onset but they are historically not the only ones. For example, CIO development in HEI-OC1s has also correlated with the expression of IL-4 and IL-13, which appear to trigger the signalling cascade that leads to the phosphorylation and activation of STAT6. CIO also correlated with an upregulation in IL-5, but it was oddly found irrelevant to pathology [125]. Cisplatin toxicity has also been measured through the proportional upregulation or downregulation of IL-8 in the context of both HEK293 and HeLa cells [78].
Attempts to ascertain the entire scope of cytokine and chemokine profile changes associated with CITs have revealed the relevance of certain effector regulation, including IL-1/IL-1Β, regulated upon activation, normal T-cell expressed, and secreted (RANTES) (CCL5), MCP-1 (CCL2), MIP2 (CXCL2), Macrophage Migration Inhibitory Factor (MIF), IP-10 (CXCL10), KC (murine IL-8), IL-17A, IL-18, IFN-γ, and IL-10 [124,125,134,137,138,139,140,141,142,143,144,145,146,147,148]. However, their role in CITs has only grown in complexity. Cytokines can be extremely multifaceted; the same cytokine can elicit both pro-inflammatory and anti-inflammatory responses depending on circumstance, and this is something that must be contended with in the pursuit of novel therapies. For example, while IL-6 appears to adhere to its canonical designation as a pro-inflammatory cytokine, at least in the context of cisplatin-induced toxicities specifically, IL-4 exerts its influence variably. IL-4 expression is directly related to the severity of CIO, as mentioned previously, but it is inversely related to the severity of CIN and CIPN and is a hallmark of protection paired with IL10 [125,143]. IFN-γ similarly manifests as a pro-inflammatory and toxic effector in CIO (when detectable), but conversely presents far more phenotypically ‘complex’ in models in CIN [125,145,149,150,151]. Ultimately, despite the headway made thus far in the analysis of cytokine and chemokine involvement in CITs, there is clear room for growth in our understanding of the actual underlying mechanisms involved.

4. Oxidative Stress and Immunologic Regulation

Cisplatin is well characterised to execute cell damage and death via reactive oxygen species (ROS) and reactive nitrogen species (RNS) [152,153,154]. As such, there was previously an emphasis on employing ROS scavengers and anti-oxidative species such as N-acetyl cysteine, glutathione, or sodium thiosulfate in treatments for CITs, particularly in CIO [155]. As opposed to immune-mediated CIT, the release of ROS occurs downstream in the signalling pathway of cisplatin-induced damage in healthy cells. Because the significance of ROS as the damage-causing agent has been established in CIT, it is only touched on briefly in this review—specifically in the context of cisplatin-induced inflammation. It is becoming more and more apparent that immunologic signalling is the initiator of damage effects under cisplatin treatment, and targeting components upstream of ROS work to block the actual cisplatin interface of this signalling pathway. Many reports highlight the close ties between innate immune stimulation and oxidative stress, with each positively regulating the activity of the other [75,154,156]. Incidentally, immunogenic signalling and oxidative stress appears to be important in potentiating cisplatin’s anti-cancer efficacy as well [157,158]. As such, general targeting of ROS and DAMP signalling in all cells may interfere with cisplatin functionality. While oxidative stress is a widespread effect that plays a role in both CITs as well as cisplatin’s anti-tumour effects, immune receptors appear to be predominantly involved in CIT development. In this way, targeting oxidative stress is becoming a less favorable target to anti-inflammation-based therapy, which could allow selectivity for non-tumour cell protection while not interfering with cisplatin’s anti-cancer activity.
CIO is associated with a depletion of endogenous anti-oxidant factors and enzymes in the cochlea, as well as an increase in oxidative enzymes such as the NADPH oxidase [154,158], all leading to an increase in oxidative stress under cisplatin treatment. This elevated ROS profile following cochlear injury mirrors effects that are seen following noise-induced injury as well [159]. Consequently, antioxidants protect against both cisplatin-induced and noise-induced hair cell death in the cochlea of the inner ear [154,160]. In keeping with the tightly regulated relationship between ROS generation and immune signalling, increased inflammatory signalling has been found to precede ROS generation in CIO [121], but upregulation of transcription factors involved in anti-oxidation effects like nuclear factor erythroid 2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) can attenuate pro-inflammatory cytokine secretion and resulting CIO [74,161], suggesting a positive feedback mechanism between oxidative stress and inflammation in CITs. Similarly, NADPH oxidases enhance TLR4-mediated inflammation in models of sepsis, where a lack of Nrf2 exacerbates inflammation and ROS generation [156]. It has been suggested that this positive feedback effect in CIO may occur via regulators like STAT1, which enhance both pro-inflammatory cytokine production like TNF-α as well as oxidative species like iNOS [135]. Of course, the role of ROS is not limited to CIO but is also involved in CIN and CIH, where oxidative stress is stimulated by inflammatory mediators, or by cisplatin-induced mitochondrial, endoplasmic reticular, or homeostatic dysfunction [104,112,162,163,164].
Cisplatin-induced cell stress and ROS generation facilitate the release of DAMPs which exacerbate inflammation and activation of cell death pathways. For more depth on the role of ROS in CITs, please see a more comprehensive review [153]. For the purposes of this review, our focus is on the role of inflammation as an instigator of CITs as a result of the growing interest in identifying specific mechanisms and targets upstream of cisplatin-induced damage.

5. Anti-Inflammatory Remedies

The prevention and treatment of cisplatin-induced toxicities thus far have been limited. The most common course of action to prevent CIT onset has been to reduce dosage, risking reduced efficacy of chemotherapy. Certain conditions, such as cisplatin-induced nephrotoxicity, do have established standard of care procedures. These often only alleviate symptoms rather than prevent or reverse the damage accumulated throughout the chemotherapeutic process. Some strategies may even pose direct negative effects on the efficacy of cancer treatment. There is thus considerable interest in the use of natural products and the development of pharmaceuticals capable of preventing or ameliorating cisplatin-induced toxicities with extreme specificity. Of the numerous compounds that have been investigated, the most extensively studied have had anti-inflammatory properties as either their primary or secondary mode of action. Other reviews have covered the entire gamut of potential remedies; in the following, there will be a particular focus on the most well-established, and most wide-acting, antioxidant and anti-inflammatory options.
For preclinical, all-natural remedies of CITs, the options are abundant (Table 1). At the same time, there is a strong trend in preclinical research towards repurposing natural compounds for scientific and medical purposes as de novo drug discovery and drug synthesis is both time-consuming and expensive. Notably, there is a discrepancy in trends between preclinical natural remedies (Table 1) and current preclinical pharmacological ‘repurposed’ options (Table 2). Moreover, there is likely a great desire to determine whether ancient forms of medicine hold up to academic scrutiny. Of those chosen and listed, the vast majority do show promise as no-cost or low-cost dietary supplements, derived from naturally occurring, edible plants and/or fauna. Whether their aptitude for rescue in-vitro and in-vivo will translate to success in medical practice has yet to be seen; at the moment, only two colloquially-considered natural products have cleared at least Phase I of clinical testing for CIT therapy: Ginko Biloba Extract, for CIO, and Silymarin, for CIN (Table 3), neither of which predominantly operate through specifically anti-inflammatory mechanisms. While there is a substantial number of naturopathic treatments that test a variety of explicitly anti-inflammatory mechanisms, of which this is not a comprehensive list, there is a distinct lack of an anti-inflammatory focus in the pool of anti-CIT pharmaceuticals. Based on current and extensive reviews on the most promising therapeutics and therapeutic targets, there are really quite few pharmacological intervention options available that are explicitly anti-inflammatory by design. Most of the preclinical and clinical pharmaceuticals under investigation indicate a trend towards a focus on antioxidation, even if several of them do boast potent, secondary, anti-inflammatory qualities.
This is not necessarily unusual, or unexpected. The use of anti-inflammatory drugs can be complicated—especially within the framework of cancer and chemotherapy. The use of anti-inflammatory prescription medication is extremely regulated. Regimens must be well optimised, if not outright personalised, to avoid dangerous levels of immunosuppression and risk of complications. Patients undergoing chemotherapy are often rendered immunosuppressed to some extent already, so the introduction of additional anti-inflammatory agents may certainly prove to be problematic. There is, for instance, quite a complicated relationship between non-steroidal anti-inflammatory drugs (NSAIDs) and cisplatin efficacy already. Most NSAIDs operate by inhibiting the actions of the pro-inflammatory factor, COX2—as many of the preclinical natural and pharmaceutical anti-inflammatory options listed. Unlike those therapeutics, however, meta-analyses have identified cases wherein NSAIDs have, unexpectedly, resulted in either the inhibition of cisplatin treatment or the promotion of CITs—CINs and CICs specifically [311]. Despite this, research into NSAID use during chemotherapy has continued, and several NSAIDs appear capable of improving the outcomes of cisplatin chemotherapy in-vitro and in-vivo [312]. Etoricoxib, as described above, qualifies as a typical NSAID and is representative, in truth, of quite a number of NSAIDs, such as the salicylates, with demonstrable capacity to alleviate CITs, such as CIO and CIN, with little to negative effects on chemotherapy. There are reports of salicylates selectively boosting the cytotoxicity of cisplatin against tumours as chemosensitisers as well [313,314,315,316]. Indeed, there are numerous studies that suggest that anti-inflammatory approaches, even beyond the use of NSAIDs, may actually improve the outcomes of chemotherapy [312,317,318,319,320,321]. More importantly, literature detailing the potential of aforementioned compounds (Table 1 and Table 3) as chemosensitisers for chemotherapy—cisplatin included—is growing likely extensively enough to warrant its own review [322,323,324,325,326,327].
As such, there is still a substantial area of research open in CIT therapeutics for targeted, anti-inflammatory therapies that would be designed to interfere with the initiation of the inflammatory signalling cascade, a prospect that is not available for therapies that rely on scavenging of ROS. Given the current landscape of this research and its shift from anti-oxidative to anti-inflammatory therapies, we are closer than ever before to understanding the mechanism of CIT and intercepting the pathways to inflammation, and subsequent ROS generation, to prevent toxicities before they are able to occur. Further insights into these pathways and development of combinatorial therapies with cisplatin and inflammation-based protectants may play an important role in improving long-term health outcomes for cancer patients.

Author Contributions

Conceptualization, all authors; writing—original draft preparation, I.K.D. and A.L.; writing—review and editing, all authors; visualization, I.K.D. and A.L.; supervision, A.P.B.; funding acquisition, A.P.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Canadian Institutes of Health Research grant number PJT-178327. This research was also funded from the Li Ka Shing Institute of Virology (LKSIoV), the Cancer Research Institute of Northern Alberta (CRINA) and the generous support of the Stollery Children’s Hospital Foundation through the Women and Children’s Health Research Institute (WCHRI). APB holds a Canada Research Chair (Tier 2) and this research was undertaken, in part, thanks to funding from the Canada Research Chairs Program (231622).

Acknowledgments

Figures were created with BioRender.com and used under license.

Conflicts of Interest

The authors have no conflict of interest to declare.

References

  1. Rosenberg, B.; VanCamp, L. The Successful Regression of Large Solid Sarcoma 180 Tumors by Platinum Compounds. Cancer Res. 1970, 30, 1799–1802. [Google Scholar] [PubMed]
  2. Rosenberg, B.; VanCamp, 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]
  3. Peyrone, M. Ueber die Einwirkung des Ammoniaks auf Platinchlorur [On the Action of Ammonia on Platinum Chloride]. Ann. Chemi Pharm. 1845, 51, 1–29. [Google Scholar]
  4. Kelland, L. The resurgence of platinum-based cancer chemotherapy. Nat. Rev. Cancer 2007, 7, 573–584. [Google Scholar] [CrossRef] [PubMed]
  5. Roberts, J.J.; Thompson, A.J.; Cohen, W. The Mechanism of Action of Antitumor Platinum Compounds. Prog. Nucleic Acid Res. Mol. Biol. 1979, 22, 71–133. [Google Scholar] [PubMed]
  6. Pinto, A.L.; Lippard, S.J. Binding of the anti-tumor drug cisdiammine dichloroplatinum(ii) (cisplatin) to DNA. Biochim. Biophys. Acta 1985, 780, 167–180. [Google Scholar] [PubMed]
  7. Eastman, A. The formation, isolation and characterization of DNA adducts produced by anticancer platinum complexes. Pharmacol. Ther. 1987, 34, 155–166. [Google Scholar] [CrossRef]
  8. Kelland, L.R. New Platinum Antitumor Complexes. Crit. Rev. Oncol. /Hematol. 1993, 5, 91–93. [Google Scholar] [CrossRef]
  9. Nagasawa, S.; Takahashi, J.; Suzuki, G.; Hideya, Y.; Yamada, K. Why Concurrent CDDP and Radiotherapy Has Synergistic Antitumor Effects: A Review of In Vitro Experimental and Clinical-Based Studies. Int. J. Mol. Sci. 2021, 22, 3140. [Google Scholar] [CrossRef]
  10. Muzaffar, J.; Bari, S.; Kirtane, K.; Chung, C.H. Recent advances and future directions in clinical management of head and neck squamous cell carcinoma. Cancers 2021, 13, 1–16. [Google Scholar]
  11. Low, J.J.H.; Ilancheran, A.; Ng, J.S. Malignant Ovarian Germ-Cell Tumours. Best Pract. Res. Clin. Obstet. Gynaecol. 2012, 26, 347–355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Bookman, M.A. Optimal primary therapy of ovarian cancer. Ann. Oncol. 2016, 27, i58–i62. [Google Scholar] [CrossRef] [PubMed]
  13. Einhorn, L.H. Curing Metastatic Testicular Cancer. Proc. Natl. Acad. Sci. USA 2002, 99, 4592–4595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. de Vries, G.; Rosas-Plaza, X.; van Vugt, M.A.T.M.; Gietema, J.A.; de Jong, S. Testicular Cancer: Determinants of Cisplatin Sensitivity and Novel Therapeutic Opportunities. Cancer Treat. Rev. 2020, 88, 102054. [Google Scholar] [CrossRef]
  15. Dasari, S.; Tchounwou, P.B. Cisplatin in cancer therapy: Molecular mechanisms of action. Eur. J. Pharmacol. 2014, 740, 364–378. [Google Scholar] [CrossRef] [Green Version]
  16. Brown, A.; Kumar, S.; Tchounwou, P.B. Cisplatin-Based Chemotherapy of Human Cancers. J. Cancer Sci. Ther. 2019, 11, 97. [Google Scholar]
  17. Kamalakar, P.; Freeman, A.I.; Higby, D.J.; Wallace, H.J.; Sinks, L.F. Clinical response and toxicity with cisdichlorodiammineplatinum(II) in children. Cancer Treat. Rep. 1977, 61, 835–839. [Google Scholar]
  18. Meyers, P.A.; Schwartz, C.L.; Krailo, M.; Kleinerman, E.S.; Betcher, D.; Bernstein, M.L.; Conrad, E.; Ferguson, W.; Gebhardt, M.; Goorin, A.M.; et al. Osteosarcoma: A Randomized, Prospective Trial of the Addition of Ifosfamide and/or Muramyl Tripeptide to Cisplatin, Doxorubicin, and High-Dose Methotrexate. J. Clin. Oncol. 2005, 23, 2004–2011. [Google Scholar] [CrossRef]
  19. Schmid, I.; von Schweinitz, D. Pediatric hepato-cellular carcinoma: Challenges and solutions. J. Hepatocell. Carcinoma 2017, 4, 15–21. [Google Scholar] [CrossRef] [Green Version]
  20. Cushing, B.; Giller, R.; Cullen, J.W.; Marina, N.M.; Lauer, S.J.; Olson, T.A.; Castleberry, R.P. Randomized comparison of combination chemotherapy with etoposide, bleomycin, and either high-dose or standard-dose cis-platin in children and adolescents with high-risk malignant germ cell tumors: A pediatric intergroup study-Pediatric Oncology Group 9049 and Children’s Cancer Group 8882. J. Clin. Oncol. 2004, 22, 2691–2700. [Google Scholar]
  21. Perilongo, G.; Maibach, R.; Shafford, E.; Brugieres, L.; Brock, P.; Morland, B.; de Camargo, B.; Zsiros, J.; Roebuck, D.; Zimmermann, A.; et al. Cisplatin vs. Cisplatin-Doxorubicin in Hepatoblastoma. N. Engl. J. Med. 2009, 361, 1662–1670. [Google Scholar] [CrossRef] [Green Version]
  22. Tajbakhsh, M.; Houghton, P.J.; Morton, C.L.; Kolb, E.A.; Gorlick, R.; Maris, J.M.; Keir, S.T.; Wu, J.; Reynolds, C.P.; Smith, M.A.; et al. Initial testing of cisplatin by the pediatric preclinical testing program. Pediatr. Blood Cancer 2008, 50, 992–1000. [Google Scholar] [CrossRef] [PubMed]
  23. Kilday, J.-P.; Bouffet, E. Chemotherapy in Childhood Brain Tumors. Curr. Pediatr. Rep. 2013, 2, 38–49. [Google Scholar] [CrossRef] [Green Version]
  24. Shahid, F.; Farooqui, Z.; Khan, F. Cisplatin-induced gastrointestinal toxicity: An update on possible mechanisms and on available gastroprotec-tive strategies. Eur. J. Pharmacol. 2018, 827, 49–57. [Google Scholar] [CrossRef] [PubMed]
  25. El-Awady, E.-S.E.; Moustafa, Y.M.; Abo-Elmatty, D.M.; Radwan, A. Cisplatin-induced cardiotoxicity: Mechanisms and cardioprotective strategies. Eur. J. Pharmacol. 2011, 650, 335–341. [Google Scholar] [CrossRef] [PubMed]
  26. Tsang, R.Y.; Al-Fayea, T.; Au, H.-J. Cisplatin Overdose Toxicities and Management. Drug Saf. 2009, 32, 1109–1122. [Google Scholar] [CrossRef]
  27. Volarevic, V.; Djokovic, B.; Jankovic, M.G.; Harrell, C.R.; Fellabaum, C.; Djonov, V.; Arsenijevic, N. Molecular mechanisms of cis-platin-induced nephrotoxicity: A balance on the knife edge between renoprotection and tumor toxicity. J. Biomed. Sci. 2019, 26, 1–14. [Google Scholar] [CrossRef] [Green Version]
  28. Breglio, A.M.; Rusheen, A.E.; Shide, E.D.; Fernandez, K.A.; Spielbauer, K.K.; McLachlin, K.M.; Hall, M.D.; Amable, L.; Cunningham, L.L. Cisplatin is retained in the cochlea indefinitely following chemotherapy. Nat. Commun. 2017, 8, 1654. [Google Scholar] [CrossRef] [Green Version]
  29. Ruggiero, A.; Ferrara, P.; Attinà, G.; Rizzo, D.; Riccardi, R. Renal toxicity and chemotherapy in children with cancer. Br. J. Clin. Pharmacol. 2017, 83, 2605–2614. [Google Scholar] [CrossRef]
  30. Manohar, S.; Leung, N. Cisplatin nephrotoxici-ty: A review of the literature. J. Nephrol. 2018, 31, 15–25. [Google Scholar] [CrossRef]
  31. Vincenzi, B.; Armento, G.; Ceruso, M.S.; Catania, G.; Leakos, M.; Santini, D.; Minotti, G.; Tonini, G. Drug-induced hepatotoxicity in cancer patients-implication for treatment. Expert Opin. Drug Saf. 2016, 15, 1219–1238. [Google Scholar] [CrossRef] [PubMed]
  32. Yaegashi, A.; Yoshida, K.; Suzuki, N.; Shimada, I.; Tani, Y.; Saijo, Y.; Toyama, A. A case of severe hepatotoxicity induced by cisplatin and 5-fluorouracil. Int. Cancer Conf. J. 2019, 9, 24–27. [Google Scholar] [CrossRef] [PubMed]
  33. Chiorazzi, A.; Semperboni, S.; Marmiroli, P. Current View in Platinum Drug Mechanisms of Peripheral Neurotoxicity. Toxics 2015, 3, 304–321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Staff, N.P.; Cavaletti, G.; Islam, B.; Lustberg, M.; Psimaras, D.; Tamburin, S. Platinum-induced peripheral neurotoxicity: From pathogenesis to treatment. J. Peripher. Nerv. Syst. 2019, 24, S26–S39. [Google Scholar] [CrossRef]
  35. Albers, J.W.; Chaudhry, V.; Cavaletti, G.; Donehower, R.C. Interventions for preventing neuropathy caused by cisplatin and related com-pounds. Cochrane Database Syst. Rev. 2014, 3, 1–79. [Google Scholar]
  36. Romano, A.; Capozza, M.A.; Mastrangelo, S.; Maurizi, P.; Triarico, S.; Rolesi, R.; Attinà, G.; Fetoni, A.R.; Ruggiero, A. Assessment and Management of Platinum-Related Ototoxicity in Children Treated for Cancer. Cancers 2020, 12, 1266. [Google Scholar] [CrossRef]
  37. van As, J.W.; van den Berg, H.; van Dalen, E.C. Platinum-induced hearing loss after treatment for childhood cancer. Cochrane Database Syst. Rev. 2016, 8, 1–72. [Google Scholar] [CrossRef]
  38. Callejo, A.; Sedó-Cabezón, L.; Juan, I.D.; Llorens, J. Cisplatin-Induced Ototoxicity: Effects, Mechanisms and Protection Strategies. Toxics 2015, 3, 268–293. [Google Scholar] [CrossRef] [Green Version]
  39. Marshall, J.S.; Warrington, R.; Watson, W.; Kim, H.L. An introduction to immunology and immunopathology. Allergy Asthma Clin. Immunol. 2018, 14, 1–10. [Google Scholar] [CrossRef] [Green Version]
  40. Chaplin, D.D. Overview of the immune response. J. Allergy Clin. Immunol. 2010, 125, S3–S23. [Google Scholar] [CrossRef]
  41. Takeuchi, O.; Akira, S. Pattern Recognition Receptors and Inflammation. Cell 2010, 140, 805–820. [Google Scholar] [CrossRef] [Green Version]
  42. Kawasaki, T.; Kawai, T. Toll-like receptor signaling pathways. Front. Immunol. 2014, 5, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Piccinini, A.M.; Midwood, K.S. DAMPening Inflammation by Modulating TLR Signalling. Mediat. Inflamm. 2010, 2010, 1–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Schmidt, M.; Raghavan, B.; Müller, V.; Vogl, T.; Fejer, G.; Tchaptchet, S.; Keck, S.; Kalis, C.; Nielsen, P.J.; Galanos, C.; et al. Crucial role for human Toll-like receptor 4 in the development of contact allergy to nickel. Nat. Immunol. 2010, 11, 814–819. [Google Scholar] [CrossRef] [PubMed]
  45. Raghavan, B.; Martin, S.F.; Esser, P.; Goebeler, M.; Schmidt, M. Metal allergens nickel and cobalt facilitate TLR4 homodimerization independently of MD2. EMBO Rep. 2012, 13, 1109–1115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Oblak, A.; Pohar, J.; Jerala, R. MD-2 determinants of nickel and cobalt-mediated activation of human TLR4. PLoS ONE 2015, 10, e0120583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Rachmawati, D.; Bontkes, H.J.; Verstege, M.I.; Muris, J.; von Blomberg, B.M.E.; Scheper, R.J.; van Hoogstraten, I.M.W. Transition metal sensing by Toll-like receptor-4: Next to nickel, cobalt and palladium are potent human dendritic cell stimulators. Contact Dermat. 2013, 68, 331–338. [Google Scholar] [CrossRef]
  48. Brodsky, I.E.; Monack, D. NLR-mediated control of inflammasome assembly in the host response against bacterial pathogens. Semin. Immunol. 2009, 21, 199–207. [Google Scholar] [CrossRef]
  49. Lupfer, C.; Kanneganti, T.-D. The expanding role of NLRs in antiviral immunity. Immunol. Rev. 2013, 255, 13–24. [Google Scholar] [CrossRef] [Green Version]
  50. Platnich, J.M.; Muruve, D.A. NOD-like receptors and inflammasomes: A review of their canonical and non-canonical signaling pathways. Arch. Biochem. Biophys. 2019, 670, 4–14. [Google Scholar] [CrossRef] [PubMed]
  51. Broz, P.; Dixit, V.M. Inflammasomes: Mechanism of assembly, regulation and signalling. Nat. Rev. Immunol. 2016, 16, 407–420. [Google Scholar] [CrossRef] [PubMed]
  52. Zheng, D.; Liwinski, T.; Elinav, E. Inflammasome activation and regulation: Toward a better understanding of complex mechanisms. Cell Discov. 2020, 36, 1–22. [Google Scholar] [CrossRef] [PubMed]
  53. Chiu, Y.H.; MacMillan, J.B.; Chen, Z.J. RNA Polymerase III Detects Cytosolic DNA and Induces Type I Interferons through the RIG-I Pathway. Cell 2009, 138, 576–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Rehwinkel, J.; Gack, M.U. RIG-I-like receptors: Their regulation and roles in RNA sensing. Nat. Rev. Immunol. 2020, 20, 537–551. [Google Scholar] [CrossRef]
  55. Onomoto, K.; Onoguchi, K.; Yoneyama, M. Regulation of RIG-I-like receptor-mediated signaling: Interaction between host and viral factors. Cell. Mol. Immunol. 2021, 18, 539–555. [Google Scholar] [CrossRef]
  56. Drouin, M.; Saenz, J.; Chiffoleau, E. C-Type Lectin-Like Receptors: Head or Tail in Cell Death Immunity. Front. Immunol. 2020, 11, 251. [Google Scholar] [CrossRef] [Green Version]
  57. Brown, G.D.; Willment, J.; Whitehead, L. C-type lectins in immunity and homeostasis. Nat. Rev. Immunol. 2018, 18, 374–389. [Google Scholar] [CrossRef]
  58. Mogensen, T.H. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin. Microbiol. Rev. 2009, 22, 240–273. [Google Scholar] [CrossRef] [Green Version]
  59. Shaulian, E.; Karin, M. AP-1 as a regulator of cell life and death. Nat. Cell Biol. 2002, 4, E131–E136. [Google Scholar] [CrossRef]
  60. Wagner, E.F.; Eferl, R. Fos/AP-1 proteins in bone and the immune system. Immunol. Rev. 2005, 208, 126–140. [Google Scholar] [CrossRef]
  61. Trop-Steinberg, S.; Azar, Y. AP-1 Expression and its Clinical Relevance in Immune Disorders and Cancer. Am. J. Med. Sci. 2017, 353, 474–483. [Google Scholar] [CrossRef] [PubMed]
  62. Kucharczak, J.; Simmons, M.J.; Fan, Y.; Gélinas, C. To be, or not to be: NF-κB is the answer-Role of Rel/NF-κB in the regulation of apoptosis. Oncogene 2003, 22, 8961–8982. [Google Scholar] [CrossRef] [Green Version]
  63. Sun, S.C. The non-canonical NF-κB pathway in immunity and inflammation. Nat. Rev. Immunol. 2017, 17, 545–558. [Google Scholar] [CrossRef] [PubMed]
  64. Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, e17023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Tamura, T.; Yanai, H.; Savitsky, D.; Taniguchi, T. The IRF Family Transcription Factors in Immunity and Oncogenesis. Annu. Rev. Immunol. 2008, 26, 535–584. [Google Scholar] [CrossRef]
  66. Jefferies, C.A. Regulating IRFs in IFN driven dis-ease. Front. Immunol. 2019, 10, 325. [Google Scholar] [CrossRef] [Green Version]
  67. Germolec, D.R.; Shipkowski, K.A.; Frawley, R.P.; Evans, E. Markers of Inflammation. In Immunotoxicity Testing—Methods in Molecular Biology; Springer: Berlin/Heidelberg, Germany, 2018; Volume 1803, pp. 57–79. [Google Scholar]
  68. Turner, M.D.; Nedjai, B.; Hurst, T.; Pennington, D.J. Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease. Biochim. Biophys. Acta 2014, 1843, 2563–2582. [Google Scholar] [CrossRef] [Green Version]
  69. Cray, C. Acute phase proteins in animals. Prog. Mol. Biol. Transl. Sci. 2012, 105, 113–150. [Google Scholar]
  70. Schrödl, W.; Büchler, R.; Wendler, S.; Reinhold, P.; Muckova, P.; Reindl, J.; Rhode, H. Acute phase proteins as promising biomarkers: Perspectives and limitations for human and veterinary medicine. Proteom.-Clin. Appl. 2016, 10, 1077–1092. [Google Scholar] [CrossRef]
  71. Kawai, T.; Akira, S. TLR signaling. Cell Death Differ. 2006, 13, 816–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Zhang, B.; Ramesh, G.; Uematsu, S.; Akira, S.; Reeves, W.B. TLR4 Signaling Mediates Inflammation and Tissue Injury in Nephrotoxicity. J. Am. Soc. Nephrol. 2008, 19, 923–932. [Google Scholar] [CrossRef] [PubMed]
  73. Zhou, J.; Fan, Y.; Zhong, J.; Huang, Z.; Huang, T.; Lin, S.; Chen, H. TAK1 mediates excessive autophagy via p38 and ERK in cisplatin-induced acute kidney injury. J. Cell. Mol. Med. 2018, 22, 2908–2921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zhou, J.; An, C.; Jin, X.; Hu, Z.; Safirstein, R.L.; Wang, Y. TAK1 deficiency attenuates cisplatin-induced acute kidney injury. Am. J. Physiol. -Ren. Physiol. 2020, 318, F209–F215. [Google Scholar] [CrossRef] [PubMed]
  75. So, H.; Kim, H.; Kim, Y.; Kim, E.; Pae, H.O.; Chung, H.T.; Park, R. Evidence that cisplatin-induced auditory damage is attenuated by downregulation of pro-inflammatory cytokines via Nrf2/HO-1. JARO-J. Assoc. Res. Otolaryngol. 2008, 9, 290–306. [Google Scholar] [CrossRef] [Green Version]
  76. Oh, G.-S.; Kim, H.-J.; Choi, J.-H.; Shen, A.; Kim, C.-H.; Kim, S.-J.; Shin, S.-R.; Hong, S.-H.; Kim, Y.; Park, C.; et al. Activation of Lipopolysaccharide–TLR4 Signaling Accelerates the Ototoxic Potential of Cisplatin in Mice. J. Immunol. 2010, 186, 1140–1150. [Google Scholar] [CrossRef]
  77. Ramesh, G.; Zhang, B.; Uematsu, S.; Akira, S.; Reeves, W.B. Endotoxin and cisplatin synergistically induce renal dysfunction and cytokine production in mice. Am. J. Physiol.-Ren. Physiol. 2007, 293, 325–332. [Google Scholar] [CrossRef]
  78. Babolmorad, G.; Latif, A.; Domingo, I.K.; Pollock, N.M.; Delyea, C.; Rieger, A.M.; Bhavsar, A.P. Toll-like receptor 4 is activated by platinum and contributes to cisplatin-induced ototoxicity. EMBO Rep. 2021, 22, e51280. [Google Scholar] [CrossRef]
  79. Woller, S.A.; Corr, M.; Yaksh, T.L. Differences in cisplatin-induced mechanical allodynia in male and female mice. Eur. J. Pain 2015, 19, 1476–1485. [Google Scholar] [CrossRef] [Green Version]
  80. Park, H.J.; Stokes, J.A.; Corr, M.; Yaksh, T.L. Toll-like receptor signaling regulates cisplatin-induced mechanical allodynia in mice. Cancer Chemother. Pharmacol. 2013, 73, 25–34. [Google Scholar] [CrossRef] [Green Version]
  81. Hutchinson, M.R.; Zhang, Y.; Brown, K.; Coats, B.D.; Shridhar, M.; Sholar, P.W.; Watkins, L.R. Co-expression gene modules involved in cisplatin-induced peripheral neuropathy according to sensitivity, status, and severity. J. Peripher. Nerv. Syst. 2020, 25, 366–376. [Google Scholar]
  82. Fumagalli, G.; Monza, L.; Cavaletti, G.; Rigolio, R.; Meregalli, C. Neuroinflammatory Process Involved in Different Preclinical Models of Chemotherapy-Induced Peripheral Neuropathy. Front. Immunol. 2021, 11, 626687. [Google Scholar] [CrossRef] [PubMed]
  83. Hutchinson, M.R.; Zhang, Y.; Brown, K.; Coats, B.D.; Shridhar, M.; Sholar, P.W.; Patel, S.J.; Crysdale, N.Y.; Harrison, J.A.; Maier, S.F.; et al. Non-stereoselective reversal of neuropathic pain by naloxone and naltrexone: Involvement of toll-like receptor 4 (TLR4). Eur. J. Neurosci. 2008, 28, 20–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Johnston, I.N.; Tan, M.; Cao, J.; Matsos, A.; Forrest, D.R.; Si, E.; Fardell, J.E.; Hutchinson, M. Ibudilast reduces oxaliplatin-induced tactile allodynia and cognitive impairments in rats. Behav. Brain Res. 2017, 334, 109–118. [Google Scholar] [CrossRef]
  85. Hutchinson, M.R.; Northcutt, A.L.; Hiranita, T.; Wang, X.; Lewis, S.S.; Thomas, J.; Van Steeg, K.; Kopajtic, T.A.; Loram, L.C.; Sfregola, C.; et al. Opioid Activation of Toll-Like Receptor 4 Contributes to Drug Reinforcement. J. Neurosci. 2012, 32, 11187–11200. [Google Scholar] [CrossRef] [Green Version]
  86. Oliveros, G.; Wallace, C.H.; Chaudry, O.; Liu, Q.; Qiu, Y.; Xie, L.; Serrano, P. A Repurposing ibudilast to mitigate Alzheimer’s disease by 1 targeting inflammation. Brain 2022. [Google Scholar] [CrossRef]
  87. Oliviera Nascimento, L.; Massari, P.; Wetzler, L.M. The Role of TLR2 in Infection and Immunity. Front. Immunol. 2012, 3, 79. [Google Scholar] [CrossRef] [Green Version]
  88. Andrade-Silva, M.; Cenedeze, M.A.; Perandini, L.A.; Felizardo, R.J.F.; Watanabe, I.K.M.; Agudelo, J.S.H.; de Almeida, D.C. TLR2 and TLR4 play opposite role in autophagy associated with cisplatin-induced acute kidney injury. Clin. Sci. 2018, 132, 1725–1739. [Google Scholar] [CrossRef]
  89. Shen, Q.; Zhang, X.; Li, Q.; Zhang, J.; Lai, H.; Gan, H.; Du, X.; Li, M. TLR2 protects cisplatin-induced acute kidney injury associated with autophagy via PI3K/Akt signaling pathway. J. Cell. Biochem. 2018, 120, 4366–4374. [Google Scholar] [CrossRef]
  90. Volarevic, V.; Markovic, B.S.; Jankovic, M.G.; Djokovic, B.; Jovicic, N.; Harrell, C.R.; Lukic, M.L. Galectin 3 protects from cispla-tin-induced acute kidney injury by promoting TLR-2-dependent activation of IDO1/Kynurenine pathway in renal DCs. Theranostics 2019, 9, 5976–6001. [Google Scholar] [CrossRef]
  91. Sallustio, F.; Costantino, V.; Cox, S.N.; Loverre, A.; Divella, C.; Rizzi, M.; Schena, F.P. Human renal stem/progenitor cells repair tubular epithelial cell injury through TLR2-driven inhibin-A and microvesicle-shuttled decorin. Kidney Int. 2013, 83, 392–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Chan, A.J.; Alikhan, M.A.; Odobasic, D.; Gan, P.Y.; Khouri, M.B.; Steinmetz, O.M.; Summers, S.A. Innate IL-17A-producing leuko-cytes promote acute kidney injury via inflammasome and toll-like receptor activation. Am. J. Pathol. 2014, 184, 1411–1418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Awadalla, A.; Mahdi, M.R.; Zahran, M.H.; Abdelbaset-Ismail, A.; El-Dosoky, M.; Negm, A. Baicalein and Alpha-Tocopherol Inhibit Toll-like Receptor Pathways in Cisplatin-Induced Nephrotoxicity. Molecules 2022, 27, 2179. [Google Scholar] [CrossRef]
  94. Zhang, Q.; Raoof, M.; Chen, Y.; Sumi, Y.; Sursal, T.; Junger, W.; Brohi, K.; Itagaki, K.; Hauser, C.J. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature 2010, 464, 104–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Alikhan, M.A.; Summers, S.A.; Gan, P.Y.; Chan, A.J.; Khouri, M.B.; Ooi, J.D.; Holdsworth, S.R. Endogenous Toll-like Recep-tor 9 Regulates AKI by Promoting Regulatory T-cell Recruitment. J. Am. Soc. Nephrol. 2016, 27, 706–714. [Google Scholar] [CrossRef]
  96. Luo, X.; Huh, Y.; Bang, S.; He, Q.; Zhang, L.; Matsuda, M.; Ji, R.-R. Macrophage Toll-like Receptor 9 Contributes to Chemotherapy-Induced Neuropathic Pain in Male Mice. J. Neurosci. 2019, 39, 6848–6864. [Google Scholar] [CrossRef]
  97. Ezhong, Y.; Ekinio, A.; Esaleh, M. Functions of NOD-Like Receptors in Human Diseases. Front. Immunol. 2013, 4, 333. [Google Scholar] [CrossRef] [Green Version]
  98. Akira, S.; Uematsu, S.; Takeuchi, O. Pathogen recognition and innate immunity. Cell 2006, 124, 783–801. [Google Scholar] [CrossRef] [Green Version]
  99. Swanson, K.V.; Deng, M.; Ting, J.P.-Y. The NLRP3 inflammasome: Molecular activation and regulation to therapeutics. Nat. Rev. Immunol. 2019, 19, 477–489. [Google Scholar] [CrossRef]
  100. Bauernfeind, F.G.; Horvath, G.; Stutz, A.; Alnemri, E.S.; MacDonald, K.; Speert, D.; Fernandes-Alnemri, T.; Wu, J.; Monks, B.G.; Fitzgerald, K.A.; et al. Cutting Edge: NF-κB Activating Pattern Recognition and Cytokine Receptors License NLRP3 Inflammasome Activation by Regulating NLRP3 Expression. J. Immunol. 2009, 183, 787–791. [Google Scholar] [CrossRef]
  101. Vajjhala, P.R.; Mirams, R.E.; Hill, J.M. Multiple binding sites on the pyrin domain of ASC protein allow self-association and interaction with NLRP3 protein. J. Biol. Chem. 2012, 287, 41732–41743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Stutz, A.; Kolbe, C.-C.; Stahl, R.; Horvath, G.L.; Franklin, B.S.; Van Ray, O.; Brinkschulte, R.; Geyer, M.; Meissner, F.; Latz, E. NLRP3 inflammasome assembly is regulated by phosphorylation of the pyrin domain. J. Exp. Med. 2017, 214, 1725–1736. [Google Scholar] [CrossRef] [PubMed]
  103. Ramesh, G.; Reeves, W.B. Inflammatory cytokines in acute renal failure. Kidney Int. 2004, 66 (Suppl. 91), S56–S61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Devarajan, P. Update on Mechanisms of Ischemic Acute Kidney Injury. J. Am. Soc. Nephrol. 2006, 17, 1503–1520. [Google Scholar] [CrossRef] [Green Version]
  105. Zhang, Y.; Yuan, F.; Cao, X.; Zhai, Z.; Huang, G.; Du, X.; Wang, Y.; Zhang, J.; Huang, Y.; Zhao, J.; et al. P2X7 receptor blockade protects against cisplatin-induced nephrotoxicity in mice by decreasing the activities of inflammasome com-ponents, oxidative stress and caspase-3. Toxicol. Appl. Pharmacol. 2014, 281, 1–10. [Google Scholar] [CrossRef]
  106. Muñoz-Planillo, R.; Kuffa, P.; Martínez-Colón, G.; Smith, B.L.; Rajendiran, T.M.; Núñez, G. K+ Efflux Is the Common Trigger of NLRP3 Inflammasome Activation by Bacterial Toxins and Particulate Matter. Immunity 2013, 38, 1142–1153. [Google Scholar] [CrossRef] [Green Version]
  107. Suprenant, A.; Rassendren, F.; Kawashima, E.; North, R.; Buell, G. The Cytolytic P2z Receptor for Extracellular ATP Identified as a P2x Receptor (P2x7). Science 1996, 272, 735–738. [Google Scholar] [CrossRef]
  108. Yang, S.-K.; Han, Y.-C.; He, J.-R.; Yang, M.; Zhang, W.; Zhan, M.; Li, A.-M.; Li, L.; Song, N.; Liu, Y.-T.; et al. Mitochondria targeted peptide SS-31 prevent on cisplatin-induced acute kidney injury via regulating mitochondrial ROS-NLRP3 pathway. Biomed. Pharmacother. 2020, 130, 110521. [Google Scholar] [CrossRef]
  109. Gao, H.; Wang, X.; Qu, X.; Zhai, J.; Tao, L.; Zhang, Y.; Song, Y.; Zhang, W. Omeprazole attenuates cisplatin-induced kidney injury through suppression of the TLR4/NF-κB/NLRP3 signaling pathway. Toxicology 2020, 440, 152487. [Google Scholar] [CrossRef]
  110. Lee, D.W.; Faubel, S.; Edelstein, C.L. A pan caspase inhibitor decreases caspase-1, IL-1αand IL-1β, and protects against necrosis of cisplatin-treated freshly isolated proximal tubules. Ren. Fail. 2014, 37, 144–150. [Google Scholar] [CrossRef]
  111. Jiang, S.; Zhang, H.; Li, X.; Yi, B.; Huang, L.; Hu, L.; Li, A.; Du, J.; Li, Y.; Zhang, W. Vitamin D/VDR attenuate cisplatin-induced AKI by down-regulating NLRP3/Caspase-1/GSDMD pyroptosis pathway. J. Steroid Biochem. Mol. Biol. 2021, 206, 105789. [Google Scholar] [CrossRef] [PubMed]
  112. Zhang, Q.; Sun, Q.; Tong, Y.; Bi, X.; Chen, L.; Lu, J.; Ding, W. Leonurine attenuates cisplatin nephrotoxicity by suppressing the NLRP3 inflammasome, mitochondrial dysfunction, and endoplasmic reticulum stress. Int. Urol. Nephrol. 2022, 1–10. [Google Scholar] [CrossRef] [PubMed]
  113. Qu, X.; Gao, H.; Tao, L.; Zhang, Y.; Zhai, J.; Sun, J.; Song, Y.; Zhang, S. Astragaloside IV protects against cis-platin-induced liver and kidney injury via autophagy-mediated inhibition of NLRP3 in rats. J. Toxicol. Sci. 2019, 44, 167–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Li, S.; Lin, Q.; Shao, X.; Mou, S.; Gu, L.; Wang, L.; Zhang, Z.; Shen, J.; Zhou, Y.; Qi, C.; et al. NLRP3 inflammasome inhibition attenuates cisplatin-induced renal fibrosis by decreasing oxidative stress and inflammation. Exp. Cell Res. 2019, 383, 111488. [Google Scholar] [CrossRef]
  115. Kim, H.-J.; Lee, D.W.; Ravichandran, K.; Keys, D.O.; Akcay, A.; Nguyen, Q.; He, Z.; Jani, A.; Ljubanovic, D.; Edelstein, C.L. NLRP3 Inflammasome Knockout Mice Are Protected against Ischemic but Not Cisplatin-Induced Acute Kidney Injury. J. Pharmacol. Exp. Ther. 2013, 346, 465–472. [Google Scholar] [CrossRef] [Green Version]
  116. Faubel, S.; Ljubanovic, D.; Reznikov, L.; Somerset, H.; Dinarello, C.A.; Edelstein, C.L. Caspase-1-deficient mice are protected against cisplatin-induced apoptosis and acute tubular necrosis. Kidney Int. 2004, 66, 2202–2213. [Google Scholar] [CrossRef] [Green Version]
  117. Vilaysane, A.; Chun, J.; Seamone, M.E.; Wang, W.; Chin, R.; Hirota, S.; Li, Y.; Clark, S.A.; Tschopp, J.; Trpkov, K.; et al. The NLRP3 Inflammasome Promotes Renal Inflammation and Contributes to CKD. J. Am. Soc. Nephrol. 2010, 21, 1732–1744. [Google Scholar] [CrossRef] [Green Version]
  118. Mulay, S.R.; Kulkarni, O.P.; Rupanagudi, K.V.; Migliorini, A.; Darisipudi, M.N.; Vilaysane, A.; Muruve, D.; Shi, Y.; Munro, F.; Liapis, H.; et al. Calcium oxalate crystals induce renal inflammation by NLRP3-mediated IL-1β secretion. J. Clin. Investig. 2012, 123, 236–246. [Google Scholar] [CrossRef]
  119. Xiong, J.; Wang, Y.; Shao, N.; Gao, P.; Tang, H.; Su, H.; Zhang, C.; Meng, X.F. The Expression and Significance of NLRP3 Inflammasome in Patients with Primary Glomerular Diseases. Kidney Blood Press. Res. 2015, 40, 344–354. [Google Scholar] [CrossRef]
  120. Eisa, N.H.; El-Sherbiny, M.; El-Magd, N.F.A. Betulin alleviates cisplatin-induced hepatic injury in rats: Targeting apoptosis and Nek7-independent NLRP3 inflammasome pathways. Int. Immunopharmacol. 2021, 99, 107925. [Google Scholar] [CrossRef]
  121. Shi, F.; Zhang, L.; Liu, X.; Wang, Y. Knock-down of microRNA miR-556-5p increases cisplatin-sensitivity in non-small cell lung cancer (NSCLC) via activating NLR family pyrin domain containing 3 (NLRP3)-mediated pyroptotic cell death. Bioengineered 2021, 12, 6332–6342. [Google Scholar] [CrossRef] [PubMed]
  122. So, H.; Kim, H.; Lee, J.H.; Park, C.; Kim, Y.; Kim, E.; Kim, J.K.; Yun, K.J.; Lee, K.M.; Lee, H.Y.; et al. Cisplatin cytotoxicity of auditory cells requires secretions of proinflammatory cytokines via activation of ERK and NF-κB. JARO-J. Assoc. Res. Otolaryngol. 2007, 8, 338–355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Zhang, B.; Ramesh, G.; Norbury, C.; Reeves, W. Cisplatin-induced nephrotoxicity is mediated by tumor necrosis factor-α produced by renal parenchymal cells. Kidney Int. 2007, 72, 37–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Faubel, S.; Lewis, E.C.; Reznikov, L.; Ljubanovic, D.; Hoke, T.S.; Somerset, H.; Oh, D.; Lu, L.; Klein, C.L.; Dinarello, C.A.; et al. Cisplatin-induced acute renal failure is associated with an increase in the cytokines interleukin (IL)-1β, IL-18, IL-6, and neutrophil in-filtration in the kidney. J. Pharmacol. Exp. Ther. 2007, 322, 8–15. [Google Scholar] [CrossRef] [PubMed]
  125. Kim, H.J.; Oh, G.S.; Lee, J.H.; Lyu, A.R.; Ji, H.M.; Lee, S.H.; Song, J.; Park, S.J.; You, Y.O.; Sul, J.D.; et al. Cisplatin ototoxicity involves cy-tokines and STAT6 signaling network. Cell Res. 2011, 21, 944–956. [Google Scholar] [CrossRef] [Green Version]
  126. Sommer, C.; Kress, M. Recent findings on how proinflammatory cytokines cause pain: Peripheral mechanisms in inflammatory and neuropathic hyperalgesia. Neurosci. Lett. 2004, 361, 184–187. [Google Scholar] [CrossRef]
  127. Wang, X.-M.; Lehky, T.J.; Brell, J.M.; Dorsey, S.G. Discovering cytokines as targets for chemotherapy-induced painful peripheral neuropathy. Cytokine 2012, 59, 3–9. [Google Scholar] [CrossRef] [Green Version]
  128. Rose-John, S.; Waetzig, G.H.; Cheller, J.; Grötzinger, J.; Seegert, D. The IL-6/sIL-6R complex as a novel target for therapeutic approaches. Expert Opin. Ther. Targets 2007, 11, 613–624. [Google Scholar] [CrossRef]
  129. Jones, S.A.; Jenkins, B.J. Recent insights into targeting the IL-6 cytokine family in inflammatory diseases and cancer. Nat. Rev. Immunol. 2018, 18, 773–789. [Google Scholar] [CrossRef]
  130. Didion, S.P. Cellular and Oxidative Mechanisms Associated with Interleukin-6 Signaling in the Vasculature. Int. J. Mol. Sci. 2017, 18, 2563. [Google Scholar] [CrossRef] [Green Version]
  131. Wassmann, S.; Stumpf, M.; Strehlow, K.; Schmid, A.; Schiefffer, B.; Böhm, M.; Nickenig, G. Interleukin-6 Induces Oxidative Stress and Endothelial Dysfunction by Overexpression of the Angiotensin II Type 1 Receptor. Circ. Res. 2004, 94, 534–541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Zelová, H.; Hošek, J. TNF-α signalling and in-flammation: Interactions between old acquaintances. Inflamm. Res. 2013, 62, 641–651. [Google Scholar] [CrossRef]
  133. Tsuruya, K.; Ninomiya, T.; Tokumoto, M.; Hirakawa, M.; Masutani, K.; Taniguchi, M.; Fukuda, K.; Kanai, H.; Kishihara, K.; Hirakata, H.; et al. Direct involvement of the receptor-mediated apoptotic pathways in cisplatin-induced renal tubular cell death. Kidney Int. 2003, 63, 72–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Ramesh, G.; Reeves, W. TNFR2-mediated apoptosis and necrosis in cisplatin-induced acute renal failure. Am. J. Physiol. Physiol. 2003, 285, F610–F618. [Google Scholar] [CrossRef] [PubMed]
  135. Ramesh, G.; Brian Reeves, W. TNF-α mediates chemokine and cytokine expression and renal injury in cisplatin nephrotoxicity. J. Clin. Investig. 2002, 110, 835–842. [Google Scholar] [CrossRef]
  136. Kaur, T.; Mukherjea, D.; Sheehan, K.; Jajoo, S.; Rybak, L.P.; Ramkumar, V. Short interfering RNA against STAT1 attenuates cisplatin-induced ototoxicity in the rat by suppressing inflammation. Cell Death Dis. 2011, 2, e180. [Google Scholar] [CrossRef] [Green Version]
  137. Digby, J.L.M.; Vanichapol, T.; Przepiorski, A.; Davidson, A.J.; Sander, V. Evaluation of cisplatin-induced injury in human kidney organoids. Am. J. Physiol. Physiol. 2020, 318, F971–F978. [Google Scholar] [CrossRef]
  138. Li, J.; Tang, Y.; Tang, P.M.K.; Lv, J.; Huang, X.R.; Carlsson-Skwirut, C.; Da Costa, L.; Aspesi, A.; Frohlich, S.; Szcześniak, P.; et al. Blocking Macrophage Migration Inhibitory Factor Protects Against Cisplatin-Induced Acute Kidney Injury in Mice. Mol. Ther. 2018, 26, 2523–2532. [Google Scholar] [CrossRef] [Green Version]
  139. Liu, P.; Li, X.; Lv, W.; Xu, Z. Inhibition of CXCL1-CXCR2 axis ameliorates cisplatin-induced acute kidney injury by mediating inflammatory response. Biomed. Pharmacother. 2019, 122, 109693. [Google Scholar] [CrossRef]
  140. Dupre, T.; Doll, M.A.; Shah, P.P.; Sharp, C.N.; Siow, D.; Megyesi, J.; Shayman, J.; Bielawska, A.; Bielawski, J.; Beverly, L.J.; et al. Inhibiting glucosylceramide synthase exacerbates cisplatin-induced acute kidney injury. J. Lipid Res. 2017, 58, 1439–1452. [Google Scholar] [CrossRef] [Green Version]
  141. Ozkok, A.; Ravichandran, K.; Wang, Q.; Ljuba-novic, D.; Edelstein, C.L. NF-κB transcriptional inhibition ameliorates cisplatin-induced acute kidney injury (AKI). Toxicol. Lett. 2016, 240, 105–113. [Google Scholar] [CrossRef] [PubMed]
  142. Liu, P.; Feng, Y.; Chen, X.; Wang, G.; Nawaz, I.; Hu, L.; Zhao, L. Paracrine action of human placental trophoblast cells attenuates cisplatin-induced acute kidney injury. Life Sci. 2019, 230, 45–54. [Google Scholar] [CrossRef]
  143. Brandolini, L.; D’Angelo, M.; Antonosante, A.; Cimini, A.; Allegretti, M. Chemokine Signaling in Chemotherapy-Induced Neuropathic Pain. Int. J. Mol. Sci. 2019, 20, 2904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Kawai, H.; Sato, W.; Yuzawa, Y.; Kosugi, T.; Matsuo, S.; Takei, Y.; Kadomatsu, K.; Muramatsu, T. Lack of the Growth Factor Midkine Enhances Survival against Cisplatin-Induced Renal Damage. Am. J. Pathol. 2004, 165, 1603–1612. [Google Scholar] [CrossRef] [Green Version]
  145. Schmitt, N.C.; Rubel, E.W.; Nathanson, N.M. Cisplatin-Induced Hair Cell Death Requires STAT1 and Is Attenuated by Epigallocatechin Gallate. J. Neurosci. 2009, 29, 3843–3851. [Google Scholar] [CrossRef]
  146. Wei, J.; Chen, X.; Li, Q.; Chen, J.; Khan, N.; Wang, B.; Cheng, J.W.; Gordon, J.R.; Li, F. ELR-CXC chemokine antagonism and cisplatin co-treatment additively reduce H22 hepatoma tumor progression and ameliorate cis-platin-induced nephrotoxicity. Oncol. Rep. 2014, 31, 1599–1604. [Google Scholar] [CrossRef]
  147. Stevanović, M.; Lazic, A.; Popović, J.; Paunesku, T.; Woloschak, G.E. Insights into platinum-induced peripheral neuropathy–current perspective. Neural Regen. Res. 2020, 15, 1623. [Google Scholar] [CrossRef]
  148. Deng, J.; Kohda, Y.; Chiao, H.; Wang, Y.; Hu, X.; Hewitt, S.M.; Miyaji, T.; Mcleroy, P.; Nibhanupudy, B.; Li, S.; et al. Interleukin-10 inhibits ischemic and cisplatin-induced acute renal injury. Kidney Int. 2001, 60, 2118–2128. [Google Scholar] [CrossRef] [Green Version]
  149. Moon, S.K.; Woo, J.I.; Lim, D.J. Involvement of TNF-α and IFN-γ in Inflammation-Mediated Cochlear Injury. Ann. Otol. Rhinol. Laryngol. 2019, 128, 8S–15S. [Google Scholar] [CrossRef]
  150. Tadagavadi, R.K.; Reeves, W.B. Endogenous IL-10 Attenuates Cisplatin Nephrotoxicity: Role of Dendritic Cells. J. Immunol. 2010, 185, 4904–4911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Kimura, A.; Ishida, Y.; Inagaki, M.; Nakamura, Y.; Sanke, T.; Mukaida, N.; Kondo, T. Interferon-γ is protective in cis-platin-induced renal injury by enhancing autophagic flux. Kidney Int. 2012, 82, 1093–1104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Lee, J.E.; Nakagawa, T.; Kita, T.; Kim, T.S.; Iguchi, F.; Endo, T.; Shiga, A.; Lee, S.H.; Ito, J. Mechanisms of Apoptosis Induced by Cisplatin in Marginal Cells in Mouse Stria Vascularis. ORL 2004, 66, 111–118. [Google Scholar] [CrossRef] [PubMed]
  153. Rybak, L.P.; Mukherjea, D.; Jajoo, S.; Ramkumar, V. Cisplatin Ototoxicity and Protection: Clinical and Experimental Studies. Tohoku J. Exp. Med. 2009, 219, 177–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. 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]
  155. Rybak, L.P.; Whitworth, C.; Somani, S. Application of Antioxidants and Other Agents to Prevent Cisplatin Ototoxicity from the Departments of Surgery. Laryngoscope 1999, 109, 1740–1744. [Google Scholar] [CrossRef]
  156. Kong, X.; Thimmulappa, R.; Kombairaju, P.; Biswal, S. NADPH Oxidase-Dependent Reactive Oxygen Species Mediate Amplified TLR4 Signaling and Sepsis-Induced Mortality in Nrf2-Deficient Mice. J. Immunol. 2010, 185, 569–577. [Google Scholar] [CrossRef] [Green Version]
  157. Hato, S.V.; Khong, A.; de Vries, I.J.M.; Lesterhuis, W.J. Molecular Pathways: The Immunogenic Effects of Platinum-Based Chemotherapeutics. Clin. Cancer Res. 2014, 20, 2831–2837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Xue, D.-F.; Pan, S.-T.; Huang, G.; Qiu, J.-X. ROS enhances the cytotoxicity of cisplatin by inducing apoptosis and autophagy in tongue squamous cell carcinoma cells. Int. J. Biochem. Cell Biol. 2020, 122, 105732. [Google Scholar] [CrossRef]
  159. Kim, H.J.; Lee, J.H.; Kim, S.J.; Oh, G.S.; Moon, H.D.; Kwon, K.B.; Park, C.; Park, B.H.; Lee, H.K.; Chung, S.Y.; et al. Roles of NADPH oxidases in cis-platin-induced reactive oxygen species generation and ototoxicity. J. Neurosci. 2010, 30, 3933–3946. [Google Scholar] [CrossRef] [Green Version]
  160. Ohlemiller, K.K.; Wright, J.S.; Dugan, L.L. Early Elevation of Cochlear Reactive Oxygen Species following Noise Exposure. Audiol. Neurotol. 1999, 4, 229–236. [Google Scholar] [CrossRef]
  161. Henderson, D.; McFadden, S.L.; Liu, C.C.; Hight, N.; Zheng, X.Y. The role of antioxidants in protection from impulse noise. Ann. N. Y. Acad. Sci. 1999, 884, 368–380. [Google Scholar] [CrossRef]
  162. Kim, S.-J.; Park, C.; Han, A.L.; Youn, M.-J.; Lee, J.-H.; Kim, Y.; Kim, E.-S.; Kim, H.-J.; Kim, J.-K.; Lee, H.-K.; et al. Ebselen attenuates cisplatin-induced ROS generation through Nrf2 activation in auditory cells. Heart Res. 2009, 251, 70–82. [Google Scholar] [CrossRef]
  163. Zou, J.; Wang, S.-P.; Wang, Y.-T.; Wan, J.-B. Regulation of the NLRP3 inflammasome with natural products against chemical-induced liver injury. Pharmacol. Res. 2020, 164, 105388. [Google Scholar] [CrossRef]
  164. Choi, Y.-M.; Kim, H.; Shim, W.; Anwar, M.A.; Kwon, J.-W.; Kwon, H.-K.; Kim, H.J.; Jeong, H.; Kim, H.M.; Hwang, D.; et al. Mechanism of Cisplatin-Induced Cytotoxicity Is Correlated to Impaired Metabolism Due to Mitochondrial ROS Generation. PLoS ONE 2015, 10, e0135083. [Google Scholar] [CrossRef] [Green Version]
  165. De Sá Coutinho, D.; Pacheco, M.T.; Frozza, R.L.; Bernardi, A. Anti-Inflammatory Effects of Resveratrol: Mechanistic Insights. Int. J. Mol. Sci. 2018, 19, 1812. [Google Scholar] [CrossRef] [Green Version]
  166. Salehi, B.; Mishra, A.P.; Nigam, M.; Sener, B.; Kilic, M.; Sharifi-Rad, M.; Fokou, P.V.T.; Martins, N.; Sharifi-Rad, J. Resveratrol: A Double-Edged Sword in Health Benefits. Biomedicines 2018, 6, 91. [Google Scholar] [CrossRef] [Green Version]
  167. Meng, T.; Xiao, D.; Muhammed, A.; Deng, J.; Chen, L.; He, J. Anti-Inflammatory Action and Mechanisms of Resveratrol. Molecules 2021, 26, 229. [Google Scholar] [CrossRef]
  168. Do Amaral, C.L.; Francescato, H.D.C.; Coimbra, T.M.; Costa, R.S.; Darin, J.D.C.; Antunes, L.M.G.; Bianchi, M.D.L.P. Resveratrol attenuates cis-platin-induced nephrotoxicity in rats. Arch. Toxicol. 2008, 82, 363–370. [Google Scholar] [CrossRef]
  169. Olgun, Y.; Kırkım, G.; Kolatan, E.; Kıray, M.; Bagrıyanık, A.; Olgun, A.; Kızmazoglu, D.C.; Ellıdokuz, H.; Serbetcıoglu, B.; Altun, Z.; et al. Friend or foe? Effect of oral resveratrol on cisplatin ototoxicity. Laryngoscope 2013, 124, 760–766. [Google Scholar] [CrossRef]
  170. 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: Resveratrol protects against cisplatin toxicity in rats. Chem. Interact. 2021, 338, 109402. [Google Scholar] [CrossRef]
  171. Lee, C.H.; Kim, K.W.; Lee, S.M.; Kim, S.Y. Dose-Dependent Effects of Resveratrol on Cisplatin-Induced Hearing Loss. Int. J. Mol. Sci. 2020, 22, 113. [Google Scholar] [CrossRef]
  172. Darwish, M.A.; Abo-Youssef, A.M.; Khalaf, M.M.; Abo-Saif, A.A.; Saleh, I.G.; Abdelghany, T.M. Resveratrol influences platinum pharmacokinetics: A novel mechanism in protection against cisplatin-induced nephrotoxicity. Toxicol. Lett. 2018, 290, 73–82. [Google Scholar] [CrossRef]
  173. Kim, D.H.; Jung, Y.J.; Lee, J.E.; Lee, A.E.; Kang, K.P.; Lee, S.; Park, S.K.; Han, M.K.; Lee, S.Y.; Ramkumar, K.M.; et al. SIRT1 activation by resveratrol ameliorates cisplatin-induced renal injury through deacetylation of p53. Am. J. Physiol.-Ren. Physiol. 2011, 301, 427–435. [Google Scholar] [CrossRef] [Green Version]
  174. Liu, Y.; Wu, H.; Zhang, F.; Yang, J.; He, J. Resveratrol upregulates miR-455-5p to antagonize cisplatin ototoxicity via modulating the PTEN–PI3K–AKT axis. Biochem. Cell Biol. 2021, 99, 385–395. [Google Scholar] [CrossRef]
  175. Hao, Q.; Xiao, X.; Zhen, J.; Feng, J.; Song, C.; Jiang, B.; Hu, Z. Resveratrol attenuates acute kidney injury by inhibiting death receptor-mediated apoptotic pathways in a cisplatin-induced rat model. Mol. Med. Rep. 2016, 14, 3683–3689. [Google Scholar] [CrossRef] [Green Version]
  176. Said, R.S.; Mantawy, E.M.; El-Demerdash, E. Mechanistic perspective of protective effects of resveratrol against cisplatin-induced ovarian injury in rats: Emphasis on anti-inflammatory and anti-apoptotic effects. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2019, 392, 1225–1238. [Google Scholar] [CrossRef]
  177. Hamad, R.; Jayakumar, C.; Ranganathan, P.; Mohamed, R.; El-Hamamy, M.M.I.; Dessouki, A.A.; Ibrahim, A.; Ramesh, G.; El-Hamamy, M.M.I. Honey feeding protects kidney against cisplatin nephrotoxicity through suppression of inflammation. Clin. Exp. Pharmacol. Physiol. 2015, 42, 843–848. [Google Scholar] [CrossRef] [Green Version]
  178. Choi, J.; Kim, S.H.; Rah, Y.C.; Chae, S.W.; Lee, J.D.; Lee, B.D.; Park, M.K. Effects of caffeic acid on cisplatin-induced hair cell damage in HEI-OC1 auditory cells. Int. J. Pediatr. Otorhinolaryngol. 2014, 78, 2198–2204. [Google Scholar] [CrossRef]
  179. Huang, H.; Shen, Z.; Geng, Q.; Wu, Z.; Shi, P.; Miao, X. Protective effect of Schisandra chinensis bee pollen extract on liver and kidney injury induced by cisplatin in rats. Biomed. Pharmacother. 2017, 95, 1765–1776. [Google Scholar] [CrossRef]
  180. Tender, T.; Rahangdale, R.R.; Balireddy, S.; Nampoothiri, M.; Sharma, K.K.; Chandrashekar, H.R. Melittin, a honeybee venom de-rived peptide for the treatment of chemotherapy-induced peripheral neuropathy. Med. Oncol. 2021, 38, 52. [Google Scholar] [CrossRef]
  181. Kim, H.; Hong, J.Y.; Jeon, W.-J.; Baek, S.H.; Ha, I.-H. Bee Venom Melittin Protects against Cisplatin-Induced Acute Kidney Injury in Mice via the Regulation of M2 Macrophage Activation. Toxins 2020, 12, 574. [Google Scholar] [CrossRef]
  182. Kim, H.; Lee, G.; Park, S.; Chung, H.-S.; Lee, H.; Kim, J.-Y.; Nam, S.; Kim, S.K.; Bae, H. Bee Venom Mitigates Cisplatin-Induced Nephrotoxicity by Regulating CD4+CD25+Foxp3+Regulatory T Cells in Mice. Evid.-Based Complement. Altern. Med. 2013, 2013, 879845. [Google Scholar] [CrossRef] [Green Version]
  183. Rezaee, R.; Momtazi, A.A.; Monemi, A.; Sahebkar, A. Curcumin: A potentially powerful tool to reverse cisplatin-induced toxicity. Pharmacol. Res. 2017, 117, 218–227. [Google Scholar] [CrossRef]
  184. Mercantepe, F.; Mercantepe, T.; Topcu, A.; Yılmaz, A.; Tumkaya, L. Protective effects of amifostine, curcumin, and melatonin against cispla-tin-induced acute kidney injury. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2018, 391, 915–931. [Google Scholar] [CrossRef]
  185. El-Gizawy, M.M.; Hosny, E.N.; Mourad, H.H.; Abd-El Razik, A.N. Curcumin nanoparticles ameliorate hepatotoxicity and nephrotoxicity in-duced by cisplatin in rats. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2020, 393, 1941–1953. [Google Scholar] [CrossRef]
  186. Khadrawy, Y.A.; El-Gizawy, M.M.; Sorour, S.M.; Sawie, H.G.; Hosny, E.N. Effect of curcumin nanoparticles on the cisplatin-induced neurotoxicity in rat. Drug Chem. Toxicol. 2018, 42, 194–202. [Google Scholar] [CrossRef]
  187. Ueki, M.; Ueno, M.; Morishita, J.; Maekawa, N. Curcumin ameliorates cisplatin-induced nephrotoxicity by inhibiting renal inflammation in mice. J. Biosci. Bioeng. 2012, 115, 547–551. [Google Scholar] [CrossRef]
  188. Topcu-Tarladacalisir, Y.; Sapmaz-Metin, M.; Karaca, T. Curcumin counteracts cisplatin-induced nephrotoxicity by preventing renal tubular cell apoptosis. Ren. Fail. 2016, 38, 1741–1748. [Google Scholar] [CrossRef] [Green Version]
  189. Ali, B.H.; Abdelrahman, A.; Al Suleimani, Y.; Manoj, P.; Ali, H.; Nemmar, A.; Al Za’Abi, M. Effect of concomitant treatment of curcumin and melatonin on cisplatin-induced nephrotoxicity in rats. Biomed. Pharmacother. 2020, 131, 110761. [Google Scholar] [CrossRef]
  190. 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]
  191. al Fayi, M.; Otifi, H.; Alshyarba, M.; Dera, A.A.; Rajagopalan, P. Thymoquinone and curcumin combination protects cisplatin-induced kidney inju-ry, nephrotoxicity by attenuating NFκB, KIM-1 and ameliorating Nrf2/HO-1 signalling. J. Drug Target. 2020, 28, 913–922. [Google Scholar] [CrossRef]
  192. Fetoni, A.R.; Paciello, F.; Mezzogori, D.; Rolesi, R.; Eramo, S.L.M.; Paludetti, G.; Troiani, D. Molecular targets for anti-cancer redox chemotherapy and cisplatin-induced ototoxicity: The role of curcumin on pSTAT3 and Nrf-2 signalling. Br. J. Cancer 2015, 113, 1434–1444. [Google Scholar] [CrossRef] [Green Version]
  193. Kumar, P.; Sulakhiya, K.; Barua, C.C.; Mundhe, N. TNF-α, IL-6 and IL-10 expressions, responsible for disparity in action of curcumin against cispla-tin-induced nephrotoxicity in rats. Mol. Cell. Biochem. 2017, 431, 113–122. [Google Scholar] [CrossRef]
  194. Borse, V.; Aameri, R.F.H.A.; Sheehan, K.; Sheth, S.; Kaur, T.; Mukherjea, D.; Tupal, S.; Lowy, M.; Ghosh, S.; Dhukwa, A.; et al. Epigallocatechin-3-gallate, a proto-typic chemopreventative agent for protection against cisplatin-based ototoxicity. Cell Death Dis. 2017, 8, e2921. [Google Scholar] [CrossRef]
  195. Yang, D.; Cao, G.; Ba, X.; Jiang, H. Epigallocat-echin-3-O-gallate promotes extracellular matrix and inhibits inflammation in IL-1β stimulated chon-drocytes by the PTEN/miRNA-29b pathway. Pharm. Biol. 2022, 60, 589–599. [Google Scholar] [CrossRef]
  196. Fatima, S.; Suhail, N.; Alrashed, M.; Wasi, S.; Aljaser, F.S.; AlSubki, R.A.; Alsharidah, A.S.; Banu, N. Epigallocatechin gallate and coen-zyme Q10 attenuate cisplatin-induced hepatotoxicity in rats via targeting mitochondrial stress and apoptosis. J. Biochem. Mol. Toxicol. 2021, 35, e22701. [Google Scholar] [CrossRef]
  197. Arafa, M.H.; Atteia, H.H. Protective Role of Epigallocatechin Gallate in a Rat Model of Cisplatin-Induced Cerebral Inflammation and Oxidative Damage: Impact of Modulating NF-κB and Nrf2. Neurotox. Res. 2020, 37, 380–396. [Google Scholar] [CrossRef]
  198. Bae, K.H.; Tan, S.; Yamashita, A.; Ang, W.X.; Gao, S.J.; Wang, S.; Chung, J.E.; Kurisawa, M. Hyaluronic acid-green tea cate-chin micellar nanocomplexes: Fail-safe cisplatin nanomedicine for the treatment of ovarian cancer without off-target toxicity. Biomaterials 2017, 148, 41–53. [Google Scholar] [CrossRef]
  199. Malik, S.; Suchal, K.; Bhatia, J.; Gamad, N.; Dinda, A.K.; Gupta, Y.K.; Arya, D.S. Molecular mechanisms underlying attenuation of cisplatin-induced acute kidney injury by epicatechin gallate. Lab. Investig. 2016, 96, 853–861. [Google Scholar] [CrossRef] [Green Version]
  200. Gündoğdu, R.; Erkan, M.; Kökoğlu, K. Assessment of the effectiveness of quercetin on cisplatin-induced ototoxicity in rats. J. Int. Adv. Otol. 2019, 15, 229–236. [Google Scholar] [CrossRef]
  201. Lee, S.K.; Oh, K.H.; Chung, A.Y.; Park, H.C.; Lee, S.H.; Kwon, S.Y.; Choi, J. Protective role of quercetin against cisplatin-induced hair cell damage in zebrafish embryos. Hum. Exp. Toxicol. 2015, 34, 1043–1052. [Google Scholar] [CrossRef] [PubMed]
  202. Sánchez-González, P.D.; López-Hernández, F.J.; Dueñas, M.; Prieto, M.; Sanchez-Lopez, E.; Thomale, J.; Ruiz-Ortega, M.; López-Novoa, J.M.; Morales, A.I. Differential effect of quercetin on cisplatin-induced toxicity in kidney and tumor tissues. Food Chem. Toxicol. 2017, 107, 226–236. [Google Scholar] [CrossRef] [PubMed]
  203. Muñoz-Reyes, D.; Casanova, A.G.; González-Paramás, A.M.; Martín, Á.; Santos-Buelga, C.; Morales, A.I.; López-Hernández, F.J.; Prieto, M. Protective Effect of Quercetin 3-O-Glucuronide against Cisplatin Cytotoxicity in Renal Tubular Cells. Molecules 2022, 27, 1319. [Google Scholar] [CrossRef] [PubMed]
  204. Azevedo, M.I.; Pereira, A.F.; Nogueira, R.B.; Rolim, F.E.; Brito, G.A.; Wong, D.V.T.; Lima-Júnior, R.C.; Ribeiro, R.D.A.; Vale, M.L. The Antioxidant Effects of the Flavonoids Rutin and Quercetin Inhibit Oxaliplatin-Induced Chronic Painful Peripheral Neuropathy. Mol. Pain 2013, 9, 53. [Google Scholar] [CrossRef] [Green Version]
  205. Warren, C.A.; Paulhill, K.J.; Davidson, L.A.; Lupton, J.R.; Taddeo, S.S.; Hong, M.Y.; Carroll, R.J.; Chapkin, R.S.; Turner, N.D. Quercetin May Suppress Rat Aberrant Crypt Foci Formation by Suppressing Inflammatory Mediators That Influence Proliferation and Apoptosis. J. Nutr. 2008, 139, 101–105. [Google Scholar] [CrossRef] [Green Version]
  206. Sahu, B.D.; Kumar, J.M.; Sistla, R. Baicalein, a bioflavonoid, prevents cisplatin- Induced acute kidney injury by up- regulating antioxidant defenses and down- Regulating the MAPKs and NF-κB Pathways. PLoS ONE 2015, 10, e0134139. [Google Scholar] [CrossRef] [Green Version]
  207. Huynh, D.L.; Ngau, T.H.; Nguyen, N.H.; Tran, G.B.; Nguyen, C.T. Potential therapeutic and pharmacological effects of Wogonin: An updated re-view. Mol. Biol. Rep. 2020, 47, 9779–9789. [Google Scholar] [CrossRef]
  208. Meng, X.M.; Li, H.D.; Wu, W.F.; Tang, P.M.K.; Ren, G.L.; Li, X.F.; Yang, Y.; Xu, T.; Ma, T.T. Wogonin protects against cis-platin-induced acute kidney injury by targeting RIPK1-mediated necroptosis. Lab. Investig. 2018, 98, 79–94. [Google Scholar] [CrossRef] [Green Version]
  209. Chi, Y.S.; Cheon, B.S.; Kim, H.P. Effect of wogonin, a plant flavone from Scutellaria radix, on the suppression of cyclooxygenase-2 and the in-duction of inducible nitric oxide synthase in lipopolysaccharide-treated RAW 264.7 cell. Biochem. Pharmacol. 2001, 61, 1195–1203. [Google Scholar]
  210. Man, Q.; Deng, Y.; Li, P.; Ma, J.; Yang, Z.; Yang, X.; Zhou, Y.; Yan, X. Licorice Ameliorates Cisplatin-Induced Hepatotoxicity Through Antiapoptosis, Antioxidative Stress, Anti-Inflammation, and Acceleration of Metabolism. Front. Pharmacol. 2020, 11, 563750. [Google Scholar] [CrossRef]
  211. Richard, S.A. Exploring the Pivotal Immunomodulatory and Anti-Inflammatory Potentials of Glycyrrhizic and Glycyrrhetinic Acids. Mediat. Inflamm. 2021, 2021, 6699560. [Google Scholar] [CrossRef] [PubMed]
  212. Wu, C.-H.; Chen, A.-Z.; Yen, G.-C. Protective Effects of Glycyrrhizic Acid and 18β-Glycyrrhetinic Acid against Cisplatin-Induced Nephrotoxicity in BALB/c Mice. J. Agric. Food Chem. 2015, 63, 1200–1209. [Google Scholar] [CrossRef] [PubMed]
  213. Chen, X.; Wei, W.; Li, Y.; Huang, J.; Ci, X. Hesperetin relieves cisplatin-induced acute kidney injury by mitigating oxidative stress, inflammation and apoptosis. Chem.-Biol. Interact. 2019, 308, 269–278. [Google Scholar] [CrossRef]
  214. Vasaikar, N.; Mahajan, U.; Patil, K.R.; Suchal, K.; Patil, C.R.; Ojha, S.; Goyal, S.N. D-pinitol attenuates cisplatin-induced nephrotoxicity in rats: Impact on pro-inflammatory cytokines. Chem. Interact. 2018, 290, 6–11. [Google Scholar] [CrossRef] [PubMed]
  215. Wang, M.; Chen, Z.; Yang, L.; Ding, L. Sappanone A Protects Against Inflammation, Oxidative Stress and Apoptosis in Cerebral Ischemia-Reperfusion Injury by Alleviating Endoplasmic Reticulum Stress. Inflammation 2021, 44, 934–945. [Google Scholar] [CrossRef]
  216. Kang, L.; Zhao, H.; Chen, C.; Zhang, X.; Xu, M.; Duan, H. Sappanone A protects mice against cisplatin-induced kidney injury. Int. Immunopharmacol. 2016, 38, 246–251. [Google Scholar] [CrossRef]
  217. Lee, S.; Choi, S.Y.; Choo, Y.Y.; Kim, O.; Tran, P.T.; Dao, C.T.; Min, B.S.; Lee, J.H. Sappanone A exhibits anti-inflammatory effects via modulation of Nrf2 and NF-κB. Int. Immunopharmacol. 2015, 28, 328–336. [Google Scholar] [CrossRef]
  218. Li, F.; Yao, Y.; Huang, H.; Hao, H.; Ying, M. Xanthohumol attenuates cisplatin-induced nephrotoxicity through inhibiting NF-κB and activating Nrf2 signaling pathways. Int. Immunopharmacol. 2018, 61, 277–282. [Google Scholar] [CrossRef]
  219. Ma, X.; Yan, L.; Zhu, Q.; Shao, F. Puerarin attenuates cisplatin-induced rat nephrotoxicity: The involvement of TLR4/NF-KB signaling pathway. PLoS ONE 2017, 12, e0171612. [Google Scholar] [CrossRef] [Green Version]
  220. Xu, B.; Li, J.; Chen, X.; Kou, M. Puerarin atten-uates cisplatin-induced apoptosis of hair cells through the mitochondrial apoptotic pathway. Biochim. Biophys. Acta-Mol. Cell Res. 1869, 4, 119208. [Google Scholar]
  221. Zhou, Y.-D.; Hou, J.-G.; Yang, G.; Jiang, S.; Chen, C.; Wang, Z.; Liu, Y.-Y.; Ren, S.; Li, W. Icariin ameliorates cisplatin-induced cytotoxicity in human embryonic kidney 293 cells by suppressing ROS-mediated PI3K/Akt pathway. Biomed. Pharmacother. 2018, 109, 2309–2317. [Google Scholar] [CrossRef] [PubMed]
  222. Ma, P.; Zhang, S.; Su, X.; Qiu, G.; Wu, Z. Protective effects of icariin on cisplatin-induced acute renal injury in mice. Am. J. Transl. Res. 2015, 7, 2105–2114. [Google Scholar] [PubMed]
  223. Sung, M.J.; Kim, D.H.; Jung, Y.J.; Kang, K.P.; Lee, A.S.; Lee, S.; Kim, W.; Davaatseren, M.; Hwang, J.-T.; Kim, H.-J.; et al. Genistein protects the kidney from cisplatin-induced injury. Kidney Int. 2008, 74, 1538–1547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Tan, M.; Toplu, Y.; Varan, E.; Sapmaz, E.; Özhan, O.; Parlakpınar, H.; Polat, A. The effect of genistein on cisplatin induced ototoxicity and oxidative stress. Braz. J. Otorhinolaryngol. 2021, 88, 105–111. [Google Scholar] [CrossRef]
  225. Huang, Y.-C.; Tsai, M.-S.; Hsieh, P.-C.; Shih, J.-H.; Wang, T.-S.; Wang, Y.-C.; Lin, T.-H.; Wang, S.-H. Galangin ameliorates cisplatin-induced nephrotoxicity by attenuating oxidative stress, inflammation and cell death in mice through inhibition of ERK and NF-kappaB signaling. Toxicol. Appl. Pharmacol. 2017, 329, 128–139. [Google Scholar] [CrossRef]
  226. Tomar, A.; Vasisth, S.; Khan, S.I.; Malik, S.; Nag, T.C.; Arya, D.S.; Bhatia, J. Galangin ameliorates cisplatin induced nephrotoxicity in vivo by modulation of oxidative stress, apoptosis and inflammation through interplay of MAPK signaling cascade. Phytomedicine 2017, 34, 154–161. [Google Scholar] [CrossRef]
  227. Domitrović, R.; Cvijanović, O.; Pugel, E.P.; Zagorac, G.B.; Mahmutefendić, H.; Škoda, M. Luteolin ameliorates cisplatin-induced nephrotoxicity in mice through inhibition of platinum accumulation, inflammation and apoptosis in the kidney. Toxicology 2013, 310, 115–123. [Google Scholar] [CrossRef]
  228. Li, W.; Yan, M.H.; Liu, Y.; Liu, Z.; Wang, Z.; Chen, C.; Zhang, J.; Sun, Y.S. Ginsenoside Rg5 ameliorates cisplatin-induced nephrotoxicity in mice through inhibition of inflammation, oxidative stress, and apoptosis. Nutrients 2016, 8, 566. [Google Scholar] [CrossRef] [Green Version]
  229. Zhang, W.; Hou, J.; Yan, X.; Leng, J.; Li, R.; Zhang, J.; Xing, J.; Chen, C.; Wang, Z.; Li, W. Platycodon grandiflorum saponins ameliorate cisplatin-induced acute nephrotoxicity through the NF-κB-mediated inflammation and PI3K/Akt/apoptosis signaling pathways. Nutrients 2018, 10, 1328. [Google Scholar] [CrossRef] [Green Version]
  230. Wan, Y.; Wang, J.; Xu, J.F.; Tang, F.; Chen, L.; Tan, Y.-Z.; Rao, C.L.; Ao, H.; Peng, C. Panax ginseng and its ginsenosides: Potential candidates for the prevention and treatment of chemotherapy-induced side effects. J. Ginseng Res. 2021, 45, 617–630. [Google Scholar] [CrossRef]
  231. Olgun, Y.; Kirkim, G.; Altun, Z.; Aktaş, S.; Kolatan, E.; Kiray, M.; Bağriyanik, A.; Olgun, A.; Kizmazoğlu, D.C.; Özoğul, C.; et al. Protective effect of Korean Red Ginseng on cisplatin ototoxicity: Is it effective enough? J. Int. Adv. Otol. 2016, 12, 177–183. [Google Scholar] [CrossRef] [PubMed]
  232. Kim, S.J.; Kwak, H.J.; Kim, D.S.; Choi, H.M.; Sim, J.E.; Kim, S.H.; Um, J.Y.; Hong, S.H. Protective mechanism of Korean Red Ginseng in cisplatin-induced ototoxicity through attenuation of nuclear factor-κB and caspase-1 activation. Mol. Med. Rep. 2015, 12, 315–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Ma, X.; Dang, C.; Kang, H.; Dai, Z.; Lin, S.; Guan, H.; Liu, X.; Wang, X.; Hui, W. Saikosaponin-D reduces cisplatin-induced nephrotoxicity by repressing ROS-mediated activation of MAPK and NF-κB signalling pathways. Int. Immunopharmacol. 2015, 28, 399–408. [Google Scholar] [CrossRef]
  234. Meng, J.; Qiu, S.; Zhang, L.; You, M.; Xing, H.; Zhu, J. Berberine Alleviate Cisplatin-Induced Peripheral Neuropathy by Modulating Inflammation Signal via TRPV1. Front. Pharmacol. 2022, 12, 774795. [Google Scholar] [CrossRef] [PubMed]
  235. Kim, J.-H.; Baek, J.-I.; Lee, I.-K.; Kim, U.-K.; Kim, Y.-R.; Lee, K.-Y. Protective effect of berberine chloride against cisplatin-induced ototoxicity. Genes Genom. 2021, 44, 1–7. [Google Scholar] [CrossRef]
  236. Hagar, H.; El Medany, A.; Salam, R.; El Medany, G.; Nayal, O.A. Betaine supplementation mitigates cisplatin-induced nephrotoxicity by abrogation of oxidative/nitrosative stress and suppression of inflammation and apoptosis in rats. Exp. Toxicol. Pathol. 2015, 67, 133–141. [Google Scholar] [CrossRef]
  237. al Za’abi, M.; Ali, H.; al Sabahi, M.; Ali, B.H. The salutary action of melatonin and betaine, given singly or concomitantly, on cisplatin-induced nephrotoxicity in mice. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2021, 394, 1693–1701. [Google Scholar] [CrossRef]
  238. Michel, H.E.; Menze, E.T. Tetramethylpyra-zine guards against cisplatin-induced nephrotoxicity in rats through inhibiting HMGB1/TLR4/NF-κB and activating Nrf2 and PPAR-γ signaling pathways. Eur. J. Pharmacol. 2019, 857, 172422. [Google Scholar] [CrossRef]
  239. Bayram, A.; Kaya, A.; Akay, E.; Hıra, İ.; Özcan, İ. The protective role of tetramethylpyrazine against cisplatin-induced ototoxicity. Int. J. Pediatric Otorhinolaryngol. 2017, 94, 1–7. [Google Scholar] [CrossRef]
  240. Yan, W.; Xu, Y.; Yuan, Y.; Tian, L.; Wang, Q.; Xie, Y.; Shao, X.; Zhang, M.; Ni, Z.; Mou, S. Renoprotective mechanisms of Astragaloside IV in cisplatin-induced acute kidney injury. Free Radic. Res. 2017, 51, 669–683. [Google Scholar] [CrossRef]
  241. Singh, T.G.; Singh, H.P.; Singh, R. Sinapic acid attenuates cisplatin-induced nephrotoxicity through peroxisome proliferator-activated receptor gamma agonism in rats. J. Pharm. Bioallied Sci. 2020, 12, 146–154. [Google Scholar] [CrossRef] [PubMed]
  242. Ansari, M.A. Sinapic acid modulates Nrf2/HO-1 signaling pathway in cisplatin-induced nephrotoxicity in rats. Biomed. Pharmacother. 2017, 93, 646–653. [Google Scholar] [CrossRef] [PubMed]
  243. Fouad, A.A.; Al-Melhim, W.N. Vanillin mitigates the adverse impact of cisplatin and methotrexate on rat kidneys. Hum. Exp. Toxicol. 2018, 37, 937–943. [Google Scholar] [CrossRef] [PubMed]
  244. Younis, N.N.; Elsherbiny, N.M.; Shaheen, M.A.; Elseweidy, M.M. Modulation of NADPH oxidase and Nrf2/HO-1 pathway by vanillin in cisplatin-induced nephrotoxicity in rats. J. Pharm. Pharmacol. 2020, 72, 1546–1555. [Google Scholar] [CrossRef]
  245. Potočnjak, I.; Marinić, J.; Batičić, L.; Šimić, L.; Broznić, D.; Domitrović, R. Aucubin administered by either oral or parenteral route protects against cisplatin-induced acute kidney injury in mice. Food Chem. Toxicol. 2020, 142, 111472. [Google Scholar] [CrossRef]
  246. Kadir, A.; Siddiqui, R.A.; Mirza, T. Nephroprotective role of eugenol against cisplatin-induced acute kidney injury in mice Dectection of Xeno-tropic murine leukemia virus related virus (XMRV) in Prostate Adenocarcinoma Biopsy Specimens View project Protective effects of different compounds on acetaminophen-induced hepatic damage in mice and rats. View Proj. Pak. J. Pharm. Sci. 2020, 33, 1281–1287. [Google Scholar] [CrossRef]
  247. Yogalakshmi, B.; Viswanathan, P.; Anuradha, C.V. Investigation of antioxidant, anti-inflammatory and DNA-protective properties of eugenol in thi-oacetamide-induced liver injury in rats. Toxicology 2010, 268, 204–212. [Google Scholar] [CrossRef]
  248. Sakat, M.S.; Kilic, K.; Akdemir, F.N.E.; Yildirim, S.; Eser, G.; Kiziltunc, A. The effectiveness of eugenol against cisplatin-induced ototoxicity. Braz. J. Otorhinolaryngol. 2018, 85, 766–773. [Google Scholar] [CrossRef]
  249. Akdemir, F.N.E.; Yildirim, S.; Kandemir, F.M.; Aksu, E.H.; Guler, M.C.; Ozmen, H.K.; Kucukler, S.; Eser, G. The antiapoptotic and antioxidant effects of eugenol against cisplatin-induced testicular damage in the experimental model. Andrologia 2019, 51, e13353. [Google Scholar] [CrossRef]
  250. Zhang, Y.; Chen, Y.; Li, B.; Ding, P.; Jin, D.; Hou, S.; Cai, X.; Sheng, X. The effect of monotropein on al-leviating cisplatin-induced acute kidney injury by inhibiting oxidative damage, inflammation and apoptosis. Biomed. Pharmacother. 2020, 129, 110408. [Google Scholar] [CrossRef]
  251. Shin, J.-S.; Yun, K.-J.; Chung, K.-S.; Seo, K.-H.; Park, H.-J.; Cho, Y.-W.; Baek, N.-I.; Jang, D.; Lee, K.-T. Monotropein isolated from the roots of Morinda officinalis ameliorates proinflammatory mediators in RAW 264.7 macrophages and dextran sulfate sodium (DSS)-induced colitis via NF-κB inactivation. Food Chem. Toxicol. 2013, 53, 263–271. [Google Scholar] [CrossRef] [PubMed]
  252. Zhu, F.B.; Wang, J.Y.; Zhang, Y.L.; Hu, Y.G.; Yue, Z.S.; Zeng, L.R.; Zheng, W.J.; Hou, Q.; Yan, S.G.; Quan, R.F. Mechanisms underlying the antiapoptotic and anti-inflammatory effects of monotropein in hydrogen peroxide-treated osteo-blasts. Mol. Med. Rep. 2016, 14, 5377–5384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Fan, X.; Wei, W.; Huang, J.; Liu, X.; Ci, X. Isoorientin Attenuates Cisplatin-Induced Nephrotoxicity Through the Inhibition of Oxidative Stress and Apoptosis via Activating the SIRT1/SIRT6/Nrf-2 Pathway. Front. Pharmacol. 2020, 11, 264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Muthuraman, A.; Sood, S.; Singla, S.K.; Singh, A.; Singh, A.; Singh, J. Ameliorative effect of flunarizine in cisplatin-induced acute renal failure via mitochondrial permeability transition pore in-activation in rats. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2011, 383, 57–64. [Google Scholar] [CrossRef] [PubMed]
  255. Muthuraman, A.; Singla, S.K.; Peters, A. Exploring the potential of flunarizine for cisplatin-induced painful uremic neuropathy in rats. Int. Neurourol. J. 2011, 15, 127–134. [Google Scholar] [CrossRef] [Green Version]
  256. So, H.-S.; Kim, H.-J.; Lee, J.-H.; Park, S.-Y.; Park, C.; Kim, Y.-H.; Kim, J.-K.; Lee, K.-M.; Kim, K.-S.; Chung, S.-Y.; et al. Flunarizine induces Nrf2-mediated transcriptional activation of heme oxygenase-1 in protection of auditory cells from cisplatin. Cell Death Differ. 2006, 13, 1763–1775. [Google Scholar] [CrossRef] [Green Version]
  257. Kaur, T.; Borse, V.; Sheth, S.; Sheehan, K.; Ghosh, S.; Tupal, S.; Jajoo, S.; Mukherjea, D.; Rybak, L.P.; Ramkumar, V. Adenosine A1 receptor protects against cisplatin ototoxicity by suppressing the NOX3/STAT1 inflammatory pathway in the cochlea. J. Neurosci. 2016, 36, 3962–3977. [Google Scholar] [CrossRef] [Green Version]
  258. Nishida, T.; Tsubota, M.; Kawaishi, Y.; Yamanishi, H.; Kamitani, N.; Sekiguchi, F.; Ishikura, H.; Liu, K.; Nishibori, M.; Kawabata, A. Involvement of high mobility group box 1 in the development and maintenance of chemotherapy-induced peripheral neuropathy in rats. Toxicology 2016, 365, 48–58. [Google Scholar] [CrossRef]
  259. Tsubota, M.; Fukuda, R.; Hayashi, Y.; Miyazaki, T.; Ueda, S.; Yamashita, R.; Koike, N.; Sekiguchi, F.; Wake, H.; Wakatsuki, S.; et al. Role of non-macrophage cell-derived HMGB1 in oxaliplatin-induced peripheral neuropathy and its prevention by the throm-bin/thrombomodulin system in rodents: Negative impact of anticoagulants. J. Neuroinflamm. 2019, 16, 199. [Google Scholar] [CrossRef]
  260. Sekiguchi, F.; Kawabata, A. Role of HMGB1 in Chemotherapy-Induced Peripheral Neuropathy. Int. J. Mol. Sci. 2020, 22, 367. [Google Scholar] [CrossRef]
  261. Miyagi, A.; Kawashiri, T.; Shimizu, S.; Shigematsu, N.; Kobayashi, D.; Shimazoe, T. Dimethyl Fumarate Attenuates Oxaliplatin-Induced Peripheral Neuropathy without Affecting the Anti-tumor Activity of Oxaliplatin in Rodents. Biol. Pharm. Bull. 2019, 42, 638–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  262. Sasaki, A.; Koike, N.; Murakami, T.; Suzuki, K. Dimethyl fumarate ameliorates cisplatin-induced renal tubulointerstitial lesions. J. Toxicol. Pathol. 2019, 32, 79–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  263. US National Library of Medicine. Cisplatin-Induced Ototoxicity. Clinical Trials. 2022. Available online: https://www.clinicaltrials.gov/ct2/results?cond=Cisplatin-in-duced+ototoxicity&age_v=&gndr=&type=&rslt=&phase=0&phase=1&phase=2&phase=3&Search=Apply (accessed on 24 May 2022).
  264. US National Library of Medicine. Cisplatin-Induced Nephrotoxicity. Clinical Trials. 2022. Available online: https://www.clinicaltrials.gov/ct2/results?cond=cisplatin-in-duced+nephrotoxicity&age_v=&gndr=&type=&rslt=&phase=0&phase=1&phase=2&phase=3&Search=Apply (accessed on 24 May 2022).
  265. Kawashiri, T.; Miyagi, A.; Shimizu, S.; Shigematsu, N.; Kobayashi, D.; Shimazoe, T. Dimethyl fumarate ameliorates chemotherapy agent-induced neurotoxicity in vitro. J. Pharmacol. Sci. 2018, 137, 202–211. [Google Scholar] [CrossRef] [PubMed]
  266. Tenório, M.C.d.S.; Graciliano, N.G.; Moura, F.; de Oliveira, A.C.M.; Goulart, M.O.F. N-Acetylcysteine (NAC): Impacts on Human Health. Antioxidants 2021, 10, 967. [Google Scholar] [CrossRef]
  267. Wang, W.; Chen, E.; Ding, X.; Lu, P.; Chen, J.; Ma, P.; Lu, L. N-acetylcysteine protect inner hair cells from cisplatin by alleviated celluar oxidative stress and apoptosis. Toxicol. Vitr. 2022, 81, 105354. [Google Scholar] [CrossRef]
  268. Riga, M.G.; Chelis, L.; Kakolyris, S.; Papadopoulos, S.; Stathakidou, S.; Chamalidou, E.; Xenidis, N.; Amarantidis, K.; Dimopoulos, P.; Danielides, V. Transtympanic injections of N-acetylcysteine for the prevention of cisplatin-induced ototoxicity: A feasible method with promising efficacy. Am. J. Clin. Oncol. Cancer Clin. Trials 2013, 36, 1–6. [Google Scholar] [CrossRef]
  269. Abdel-Wahab, W.M.; Moussa, F.I.; Saad, N.A. Synergistic protective effect of N-acetylcysteine and taurine against cisplatin-induced nephrotoxicity in rats. Drug Des. Dev. Ther. 2017, 11, 901–908. [Google Scholar] [CrossRef] [Green Version]
  270. Chen, C.-H.; Huang, C.-Y.; Lin, H.-Y.H.; Wang, M.-C.; Chang, C.-Y.; Cheng, Y.-F. Association of Sodium Thiosulfate with Risk of Ototoxic Effects From Platinum-Based Chemotherapy. JAMA Netw. Open 2021, 4, e2118895. [Google Scholar] [CrossRef]
  271. Zhang, M.Y.; Dugbartey, G.J.; Juriasingani, S.; Sener, A. Hydrogen sulfide metabolite, sodium thiosulfate: Clinical applications and underlying molecular mechanisms. Int. J. Mol. Sci. 2021, 22, 6452. [Google Scholar] [CrossRef]
  272. Diamond, B.J.; Shiflett, S.C.; Feiwel, N.; Matheis, R.J.; Noskin, O.; Richards, J.A.; Schoenberger, N.E. Ginkgo biloba extract: Mechanisms and clinical indications. Arch. Phys. Med. Rehabil. 2000, 81, 668–678. [Google Scholar] [CrossRef]
  273. Huang, X.; Whitworth, C.A.; Rybak, L.P. Ginkgo Biloba Extract (EGb 761) Protects Against Cisplatin-Induced Ototoxicity in Rats. Otol. Neurotol. 2007, 28, 828–833. [Google Scholar] [CrossRef] [PubMed]
  274. Cakil, B.; Basar, F.S.; Atmaca, S.; Cengel, S.K.; Tekat, A.; Tanyeri, Y. The protective effect of Ginkgo biloba extract against experimental cisplatin ototoxicity: Animal research using distortion product otoacoustic emissions. J. Laryngol. Otol. 2012, 126, 1097–1101. [Google Scholar] [CrossRef] [PubMed]
  275. Esen, E.; Özdoğan, F.; Gürgen, S.G.; Özel, H.E.; Baser, S.; Genҫ, S.; Selҫuk, A. Ginkgo biloba and lycopene are effective on cisplatin induced ototoxicity? J. Int. Adv. Otol. 2018, 14, 23–27. [Google Scholar] [CrossRef] [PubMed]
  276. Grosser, N.; Erdmann, K.; Hemmerle, A.; Berndt, G.; Hinkelmann, U.; Smith, G.; Schröder, H. Rosuvastatin upregulates the antioxidant defense protein heme oxygenase-1. Biochem. Biophys. Res. Commun. 2004, 325, 871–876. [Google Scholar] [CrossRef] [PubMed]
  277. Wang, P.; Luo, L.; Shen, Q.; Shi, G.; Mohammed, A.; Ni, S.; Wu, X. Rosuvastatin improves myocardial hypertrophy after hemodynamic pressure overload via regulating the crosstalk of Nrf2/ARE and TGF-β/smads pathways in rat heart. Eur. J. Pharmacol. 2018, 820, 173–182. [Google Scholar] [CrossRef]
  278. Mostafa, R.E.; Saleh, D.O.; Mansour, D.F. Cisplatin-induced nephrotoxicity in rats: Modulatory role of simvastatin and rosuvastatin against apoptosis and inflammation. J. Appl. Pharm. Sci. 2018, 8, 43–50. [Google Scholar]
  279. Kavalipati, N.; Shah, J.; Ramakrishan, A.; Vasnawala, H. Pleiotropic effects of statins. Indian J. Endocrinol. Metab. 2015, 19, 554–562. [Google Scholar]
  280. Bedi, O.; Dhawan, V.; Sharma, P.L.; Kumar, P. Pleiotropic effects of statins: New therapeutic targets in drug design. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2016, 389, 695–712. [Google Scholar] [CrossRef]
  281. Maheshwari, R.A.; Sailor, G.U.; Patel, L.; Balaraman, R. Amelioration of cisplatin-induced nephrotoxicity by statins. Indian J. Pharmacol. 2013, 45, 354–358. [Google Scholar] [CrossRef]
  282. Kim, S.Y.; Lee, C.H.; Min, C.; Yoo, D.M.; Choi, H.G. Association between statin medication and hearing impairment in a national health screening cohort. Sci. Rep. 2021, 11, 14388. [Google Scholar] [CrossRef]
  283. Marshak, T.; Steiner, M.; Kaminer, M.; Levy, L.; Shupak, A. Prevention of cisplatin-induced hearing loss by intratympanic dexamethasone: A ran-domized controlled study. Otolaryngol.-Head Neck Surg. 2014, 150, 983–990. [Google Scholar] [CrossRef] [PubMed]
  284. Waissbluth, S.; Salehi, P.; He, X.; Daniel, S.J. Systemic dexamethasone for the prevention of cisplatin-induced ototoxicity. Eur. Arch. Oto-Rhino-Laryngol. 2012, 270, 1597–1605. [Google Scholar] [CrossRef] [PubMed]
  285. Simsek, G.; Taş, B.M.; Muluk, N.B.; Azman, M.; Kılıç, R. Comparison of the protective efficacy between intratympanic dexamethasone and resveratrol treatments against cisplatin-induced ototoxicity: An experimental study. Eur. Arch. Oto-Rhino-Laryngol. 2019, 276, 3287–3293. [Google Scholar] [CrossRef] [PubMed]
  286. Özel, H.E.; Özdoğan, F.; Gürgen, S.G.; Esen, E.; Genҫ, S.; Selҫuk, A. Comparison of the protective effects of intratympanic dexamethasone and methylprednisolone against cisplatin-induced ototoxicity. J. Laryngol. Otol. 2016, 130, 225–234. [Google Scholar] [CrossRef] [PubMed]
  287. Haake, S.M.; Dinh, C.T.; Chen, S.; Eshraghi, A.A.; van de Water, T.R. Dexamethasone protects auditory hair cells against TNFα-initiated apopto-sis via activation of PI3K/Akt and NFκB signaling. Hear. Res. 2009, 255, 22–32. [Google Scholar] [CrossRef] [PubMed]
  288. Dinh, C.T.; Chen, S.; Bas, E.; Dinh, J.; Goncalves, S.; Telischi, F.; Angeli, S.; Eshraghi, A.A.; Van De Water, T. Dexamethasone Protects Against Apoptotic Cell Death of Cisplatin-exposed Auditory Hair Cells In Vitro. Otol. Neurotol. 2015, 36, 1566–1571. [Google Scholar] [CrossRef]
  289. Fernandez, R.; Harrop-Jones, A.; Wang, X.; Dellamary, L.; LeBel, C.; Piu, F. The sustained-exposure dexamethasone formulation OTO-104 offers effective protection against cisplatin-induced hearing loss. Audiol. Neurotol. 2016, 21, 22–29. [Google Scholar] [CrossRef]
  290. Ahmed, L.A.; Shehata, N.I.; Abdelkader, N.F.; Khattab, M.M. Tempol, a superoxide dismutase mimetic agent, ameliorates cisplatin-induced ne-phrotoxicity through alleviation of mitochondrial dysfunction in mice. PLoS ONE 2014, 9, e115983. [Google Scholar] [CrossRef] [Green Version]
  291. Youn, C.K.; Kim, J.; Jo, E.-R.; Oh, J.; Do, N.Y.; Cho, S.I. Protective Effect of Tempol against Cisplatin-Induced Ototoxicity. Int. J. Mol. Sci. 2016, 17, 1931. [Google Scholar] [CrossRef] [Green Version]
  292. Ewees, M.G.; Messiha, B.A.S.; Abdel-Bakky, M.S.; Bayoumi, A.M.A.; Abo-Saif, A.A. Tempol, a superoxide dismutase mimetic agent, reduces cisplatin-induced nephrotoxicity in rats. Drug Chem. Toxicol. 2018, 42, 657–664. [Google Scholar] [CrossRef]
  293. Afjal, M.A.; Goswami, P.; Ahmad, S.; Dabeer, S.; Akhter, J.; Salman, M.; Mangla, A.; Raisuddin, S. Tempol (4-hydroxy tempo) protects mice from cisplatin-induced acute kidney injury via modulation of expression of aquaporins and kidney injury molecule-1. Drug Chem. Toxicol. 2022, 45, 1355–1363. [Google Scholar] [CrossRef] [PubMed]
  294. Surai, P.F. Silymarin as a Natural Antioxidant: An Overview of the Current Evidence and Perspectives. Antioxidants 2015, 4, 204–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  295. Fanoudi, S.; Alavi, M.S.; Karimi, G.; Hosseinzadeh, H. Milk thistle (Silybum Marianum) as an antidote or a protective agent against natural or chemical toxicities: A review. Drug Chem. Toxicol. 2018, 43, 240–254. [Google Scholar] [CrossRef] [PubMed]
  296. Esmaeil, N.; Anaraki, S.B.; Gharagozloo, M.; Moayedi, B. Silymarin impacts on immune system as an immunomodulator: One key for many locks. Int. Immunopharmacol. 2017, 50, 194–201. [Google Scholar] [CrossRef]
  297. Hayes, D.M.; Cvitkovic, E.; Golbey, R.B.; Scheiner, E.; Helson, L.; Krakoff, I.H. High Dose Cis-Platinum Di-ammine Dichloride Amelioration of Renal Toxicity by Mannitol Diuresis. Cancer 1977, 39, 1372–1381. [Google Scholar] [CrossRef]
  298. Williams, R.P.; Ferlas, B.W.; Morales, P.C.; Kurtzweil, A.J. Mannitol for the prevention of cisplatin-induced nephrotoxicity: A retrospective comparison of hydration plus mannitol versus hydration alone in inpatient and outpatient regimens at a large academic medical center. J. Oncol. Pharm. Pract. 2016, 23, 422–428. [Google Scholar] [CrossRef]
  299. Ruggiero, A.; Ariano, A.; Triarico, S.; Capozza, M.A.; Romano, A.; Maurizi, P.; Mastrangelo, S.; Attiná, G. Cisplatin-induced nephrotoxi-city in children: What is the best protective strategy? J. Oncol. Pharm. Pract. 2021, 27, 180–186. [Google Scholar] [CrossRef]
  300. Sainamthip, P.; Saichaemchan, S.; Satirapoj, B.; Prasongsook, N. The Effect of Intravenous Mannitol Combined with Normal Saline in Preventing Cisplatin-Induced Nephrotoxicity: A Randomized, Double-Blind, Placebo-Controlled Trial. JCO Glob. Oncol. 2022, 8, 275. [Google Scholar] [CrossRef]
  301. Solanki, M.H.; Chatterjee, P.K.; Xue, X.; Gupta, M.; Rosales, I.; Yeboah, M.M.; Kohn, N.; Metz, C.N. Magnesium protects against cisplatin-induced acute kidney injury without compromising cisplatin-mediated killing of an ovarian tumor xenograft in mice. Am. J. Physiol. Renal Physiol. 2015, 309, F35–F47. [Google Scholar] [CrossRef] [Green Version]
  302. Solanki, M.H.; Chatterjee, P.K.; Gupta, M.; Xue, X.; Plagov, A.; Metz, M.H.; Mintz, R.; Singhal, P.; Metz, C.N. Magnesium protects against cisplatin-induced acute kidney injury by regulating platinum accumulation. Am. J. Physiol. Physiol. 2014, 307, F369–F384. [Google Scholar] [CrossRef]
  303. Saito, Y.; Okamoto, K.; Kobayashi, M.; Narumi, K.; Yamada, T.; Iseki, K. Magnesium attenuates cisplatin-induced nephrotoxicity by regulating the expression of renal transporters. Eur. J. Pharmacol. 2017, 811, 191–198. [Google Scholar] [CrossRef] [PubMed]
  304. Kubo, Y.; Miyata, H.; Sugimura, K.; Shinno, N.; Ushigome, H.; Yanagimoto, Y.; Takahashi, Y.; Yamamoto, K.; Nishimura, J.; Wada, H.; et al. Prophylactic Effect of Premedication with Intravenous Magnesium on Renal Dysfunction in Preoperative Cisplatin-Based Chemotherapy for Esophageal Cancer. Oncology 2019, 97, 319–326. [Google Scholar] [CrossRef] [PubMed]
  305. Hamroun, A.; Lenain, R.; Bigna, J.J.; Speyer, E.; Bui, L.; Chamley, P.; Pottier, N.; Cauffiez, C.; Dewaeles, E.; Dhalluin, X.; et al. Prevention of Cisplatin-Induced Acute Kidney Injury: A Systematic Review and Meta-Analysis. Drugs 2019, 79, 1567–1582. [Google Scholar] [CrossRef]
  306. Danwilai, K.; Lohitnavy, O.; Sakunrag, I.; Dilokthornsakul, P. The effect of magnesium supplementation on cisplatin induced nephrotoxicity: A sys-tematic review and meta-analysis. Pharm. Sci. Asia 2021, 48, 25–36. [Google Scholar] [CrossRef]
  307. Osman, N.M.; Copley, M.P.; Litterst, C.L. Amelioration of cisplatin-induced nephrotoxicity by the diuretic acetazolamide in F344 rats. Cancer Treat. Rep. 1984, 68, 999–1004. [Google Scholar]
  308. el Hamamsy, M.; Kamal, N.; Bazan, N.S.; el Haddad, M. Evaluation of the effect of acetazolamide versus mannitol on cisplatin-induced ne-phrotoxicity, a pilot study. Int. J. Clin. Pharm. 2018, 40, 1539–1547. [Google Scholar] [CrossRef] [PubMed]
  309. Ismail, R.S.; El-Awady, M.S.; Hassan, M.H. Pantoprazole abrogated cisplatin-induced nephrotoxicity in mice via suppression of inflammation, apoptosis, and oxidative stress. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2020, 393, 1161–1171. [Google Scholar] [CrossRef]
  310. Ghonaim, E.; El-Haggar, S.; Gohar, S. Possible protective effect of pantoprazole against cisplatin-induced nephrotoxicity in head and neck cancer patients: A randomized controlled trial. Med. Oncol. 2021, 38, 1–12. [Google Scholar] [CrossRef]
  311. Okamoto, K.; Saito, Y.; Narumi, K.; Furugen, A.; Iseki, K.; Kobayashi, M. Non-steroidal anti-inflammatory drugs are a risk factor for cisplatin-induced nephrotoxicity: A meta-analysis of retrospective studies. Anticancer. Res. 2020, 40, 1747–1751. [Google Scholar] [CrossRef]
  312. Ramos-Inza, S.; Ruberte, A.C.; Sanmartín, C.; Sharma, A.K.; Plano, D. NSAIDs: Old Acquaintance in the Pipeline for Cancer Treatment and Pre-vention-Structural Modulation, Mechanisms of Action, and Bright Future. J. Med. Chem. 2021, 64, 16380–16421. [Google Scholar] [CrossRef]
  313. Sidhu, H.; Capalash, N. Synergistic anti-cancer action of salicylic acid and cisplatin on HeLa cells elucidated by network pharmacology and in vitro analysis. Life Sci. 2021, 282, 119802. [Google Scholar] [CrossRef] [PubMed]
  314. 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] [PubMed]
  315. 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]
  316. 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] [PubMed]
  317. 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] [Green Version]
  318. Neumann, W.; Crews, B.C.; Sárosi, M.B.; Daniel, C.M.; Ghebreselasie, K.; Scholz, M.S.; Marnett, L.J.; Hey-Hawkins, E. Conjugation of cisplatin analogues and cyclooxygenase inhibitors to overcome cisplatin resistance. ChemMedChem 2015, 10, 183–192. [Google Scholar] [CrossRef] [Green Version]
  319. Neumann, W.; Crews, B.C.; Marnett, L.J.; Hey-Hawkins, E. Conjugates of Cisplatin and Cyclooxygenase Inhibitors as Potent Antitumor Agents Overcoming Cisplatin Resistance. ChemMedChem 2014, 9, 1150–1153. [Google Scholar] [CrossRef] [Green Version]
  320. Intini, F.P.; Zajac, J.; Novohradsky, V.; Saltarella, T.; Pacifico, C.; Brabec, V.; Natile, G.; Kasparkova, J. Novel Antitumor Platinum(II) Conjugates Containing the Nonsteroidal Anti-inflammatory Agent Diclofenac: Synthesis and Dual Mechanisms of Antiproliferative Effects. Inorg. Chem. 2017, 56, 1483–1497. [Google Scholar] [CrossRef]
  321. Hou, J.; Karin, M.; Sun, B. Targeting cancer-promoting inflammation—Have anti-inflammatory therapies come of age? Nat. Rev. Clin. Oncol. 2021, 18, 261–279. [Google Scholar] [CrossRef]
  322. Paramanantham, A.; Kim, M.J.; Jung, E.J.; Kim, H.J.; Chang, S.H.; Jung, J.M.; Hong, S.C.; Shin, S.C.; Kim, G.S.; Lee, W.S. Anthocyanins isolated from vitis coignetiae pulliat enhances cisplatin sensitivity in MCF-7 human breast cancer cells through inhibition of Akt and NF-κB activation. Molecules 2020, 25, 3623. [Google Scholar] [CrossRef]
  323. Sun, C.-Y.; Zhang, Q.-Y.; Zheng, G.-J.; Feng, B. Phytochemicals: Current strategy to sensitize cancer cells to cisplatin. Biomed. Pharmacother. 2018, 110, 518–527. [Google Scholar] [CrossRef] [PubMed]
  324. Cheuk, I.W.; Chen, J.; Siu, M.; Ho, J.C.; Lam, S.S.; Shin, V.Y.; Kwong, A. Resveratrol enhanced chemo-sensitivity by reversing macrophage polarization in breast cancer. Clin. Transl. Oncol. 2022, 24, 854–863. [Google Scholar] [CrossRef] [PubMed]
  325. Chang, W.S.; Tsai, C.W.; Yang, J.S.; Hsu, Y.M.; Shih, L.C.; Chiu, H.Y.; Bau, D.T.; Tsai, F.J. Resveratrol inhibited the metastatic behaviors of cisplatin-resistant human oral cancer cells via phosphorylation of ERK/p-38 and suppression of MMP-2/9. J. Food Biochem. 2021, 45, e13666. [Google Scholar] [CrossRef] [PubMed]
  326. Gajski, G.; Čimbora-Zovko, T.; Rak, S.; Rožman, M.; Osmak, M.; Garaj-Vrhovac, V. Combined antitumor effects of bee venom and cisplatin on human cervical and laryngeal carcinoma cells and their drug resistant sublines. J. Appl. Toxicol. 2013, 34, 1332–1341. [Google Scholar] [CrossRef] [Green Version]
  327. Paciello, F.; Fetoni, A.R.; Mezzogori, D.; Rolesi, R.; Di Pino, A.; Paludetti, G.; Grassi, C.; Troiani, D. The dual role of curcumin and ferulic acid in counteracting chemoresistance and cisplatin-induced ototoxicity. Sci. Rep. 2020, 10, 1063. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Mechanism of action of cisplatin in tumour cells. Cisplatin enters the cell either passively or through a transporter, where two water molecules replace the chloride groups. As an electrophile, cisplatin is attracted to the nitrogen on purine bases in the DNA, where it forms inter or intra-strand crosslinks, interrupting DNA repair and replication processes. Downstream this leads to oxidative stress and activation of apoptotic signalling pathways.
Figure 1. Mechanism of action of cisplatin in tumour cells. Cisplatin enters the cell either passively or through a transporter, where two water molecules replace the chloride groups. As an electrophile, cisplatin is attracted to the nitrogen on purine bases in the DNA, where it forms inter or intra-strand crosslinks, interrupting DNA repair and replication processes. Downstream this leads to oxidative stress and activation of apoptotic signalling pathways.
Ijms 23 07227 g001
Figure 2. Pattern recognition receptors and pro-inflammatory signalling pathways involved in cisplatin-induced toxicities. Pattern Recognition Receptors interact with a specific array of PAMPs and DAMPs to mediate signals that induce pro-inflammatory signalling. Most of these pathways share downstream signalling that converge on NF-κB, AP-1, or IRF3, which regulate expression of pro-inflammatory signalling molecules such as cytokines and chemokines to influence inflammation in surrounding cells. These molecules bind receptors like TNF-R and IL6-R in other cells that mediate similar signalling pathways. This localised inflammation is exacerbated by cisplatin treatment and is involved in various CITs through mechanisms that remain to be elucidated.
Figure 2. Pattern recognition receptors and pro-inflammatory signalling pathways involved in cisplatin-induced toxicities. Pattern Recognition Receptors interact with a specific array of PAMPs and DAMPs to mediate signals that induce pro-inflammatory signalling. Most of these pathways share downstream signalling that converge on NF-κB, AP-1, or IRF3, which regulate expression of pro-inflammatory signalling molecules such as cytokines and chemokines to influence inflammation in surrounding cells. These molecules bind receptors like TNF-R and IL6-R in other cells that mediate similar signalling pathways. This localised inflammation is exacerbated by cisplatin treatment and is involved in various CITs through mechanisms that remain to be elucidated.
Ijms 23 07227 g002
Figure 3. Summary of reported innate immune receptors involved in cisplatin-induced ototoxicity, peripheral neurotoxicity, nephrotoxicity, or hepatotoxicity. The depicted innate immune receptors are involved in either protecting (green arrow) or exacerbating (red arrow) inflammation in response to systemically delivered cisplatin, and a few key receptors have been reported in both cases.
Figure 3. Summary of reported innate immune receptors involved in cisplatin-induced ototoxicity, peripheral neurotoxicity, nephrotoxicity, or hepatotoxicity. The depicted innate immune receptors are involved in either protecting (green arrow) or exacerbating (red arrow) inflammation in response to systemically delivered cisplatin, and a few key receptors have been reported in both cases.
Ijms 23 07227 g003
Table 1. Examples of Prospective Natural Anti-Inflammatory CIT Remedies.
Table 1. Examples of Prospective Natural Anti-Inflammatory CIT Remedies.
ClassificationNameAnti-Inflammatory
Mechanism(s) of Action
Targeted CITsReference(s)
Non-
Flavonoid
Polyphenol
Phytoalexin
ResveratrolInhibits TLR4 Signalling
Inhibits IL-6, TNF-α,
Inhibits NF-κB Signalling
Radical Scavenging
Metal Chelation
CIO
CIOV (Ovarian)
CIU (Uterine)
CIN
[165,166,167,168,169,170,171,172,173,174,175,176]
Bee ProductsHoneyInhibits IL6, TNF-α, MCP-1, ICAM1CIN[177]
Caffeic AcidRadical ScavengingCIO[178]
Bee Pollen ExtractInhibits IL6, IL1β
Inhibits NF-κB Signalling
CIN
CIH
[179]
Mellit
(Bee Venom)
Inhibits IL-6, IL-1β, TNF-α
Upregulates IL-10
Alters/Inhibits Mθ Invasion
Enhanced T-Reg Activity
Inhibits COX2
CIN
CIPN (Potential)
[180,181,182]
FlavonoidCurcuminInhibits IL-6, IL-1β, TNF-α, MCP-1
Upregulates/maintains IL-10
Free Radical Scavenging
Inhibits COX
Inhibits JAK/STAT Signalling
Inhibits NF-κB Signalling
CIN
CIO
CIPN
CIH
[183,184,185,186,187,188,189,190,191,192,193]
Epigallocatechin Gallate
(EGCG)
Inhibits MAPK Signalling
Dis-regulates STAT1/STAT3
Signalling
Inhibits IL-6, IL-1β, TNF-α
Inhibits NF-κB Signalling
Upregulates Nrf2
CIN
CIH
CICN
(Cerebral
Neurotoxicity)
[194,195,196,197,198,199]
QuercetinInhibits NF-κB Signalling
Potential COX Inhibition
Potential iNOS Inhibition
CIO
CIN
[200,201,202,203,204,205]
BaicaleinInhibits IL-6, TNF-α
Inhibits NF-κB
Inhibits TLR-2, TLR-4 Signalling
Upregulates Nrf2
Upregulates HO-1
CIN[93,206]
WogoninInhibits IL-6, IL-8, IL-1β, MCP-1, TNF-α
Inhibits NF-κB Signalling
Inhibit COX2
CIN[207,208,209]
Glycyrrhizic Acid (Licorice)Inhibits IL-1β, TNF-α
Inhibits NF-κB Signalling
Reduces DAMP Release
CIN
CIH
[210,211,212]
HesperetinInhibits MAPK Signalling
Upregulates TNF-α Signal Inhibitor
Upregulates Nrf2
Upregulates HO-1
CIN[213]
D-PinitolInhibits IL-6, IL-1β, TNF-αCIN[214]
Sappanone AInhibits IL-6, IL-1β, TNF-α
Inhibits NF-κB Signalling
Inhibits COX2
Upregulates Nrf2
CIN[215,216,217]
XanthohumolInhibits IL-6, IL-1β, TNF-α
Inhibits TLR4 Expression
Inhibits NF-κB Signalling
CIN[218]
PuerarinInhibits IL-6, TNF-α
Inhibits TLR4 Signalling
Inhibits ΝF-κB
CIN
CIO
[219,220]
IcariinInhibits IL-1β, TNF-α
Inhibits NF-κΒ Signalling
Inhibits iNOS
CIN[221,222]
GenisteinInhibits ICAM-1, MCP-1
Inhibits Mθ Infiltration
Inhibits NF-κB Signalling
CIN
CIO
[223,224]
GalanginInhibits IL-6, IL-1β, TNF-α
Inhibits MAPK Signalling
Inhibits NF-κΒ Signalling
CIN[225,226]
LuteolinInhibits TNF-α
Inhibits NF-κB Signalling
Inhibits COX2
CIN[227]
SaponinsGinsenosides
(Several)
Inhibit IL-1β, TNF-α
Inhibit NF-κB Signalling
Inhibit COX2
Inhibit iNOS
CIN
CIO
[228,229,230,231,232,233]
AlkaloidsBerberineInhibits IL-6, IL-1β, TNF-α
Inhibits MAPK Signalling
Inhibits NF-κB Signalling
Upregulates IL-10 s
Antioxidant Effect (Reduced ROS
Generation)
CIPN
CIO
[234,235]
BetaineInhibits IL-6, IL-1β, TNF-α
Inhibits NF-κB Signalling
CIN[236,237]
TetramethylpyrazineInhibits IL-1β, TNF-α
Inhibits TLR4 Signalling
Reduces DAMP Release
Upregulates Nrf2s
CIN
CIO (Potential)
[238,239]
Other
Natural
Compounds
AstragalosideInhibits IL-1β, TNF-α
Inhibits Caspase-1 & NLRP3
Inhibits NF-κB Signalling
Upregulates Nrf2
CIN[113,240]
Sinapic AcidInhibits IL-6, IL-1β, TNF-α
Inhibits NF-κB Signalling
Upregulates Nrf2
Upregulates HO-1
CIN[241,242]
VanillinInhibits IL-18, TNF-α
Inhibits NF-κB Signalling
Inhibits iNOS
Upregulates IL-10
Upregulates Nrf2
CIN[243,244]
Table 2. Examples of preclinical anti-inflammatory therapeutics for cisplatin-induced toxicities.
Table 2. Examples of preclinical anti-inflammatory therapeutics for cisplatin-induced toxicities.
ClassificationNameAnti-Inflammatory
Mechanism(s) of Action
Targeted CITsReference(s)
Anti-Inflammatory
(Iridoid Glycoside)
AucubinInhibits TNF-α
Inhibits STAT3/MAPK Signalling
Inhibits NF-κB
Signalling
Upregulates HO-1
CIN[245]
Anti-Inflammatory
(COX2 Inhibitor)
EtoricoxibInhibits TNF-α
Inhibits iNOS
Inhibits COX2
CIN[173]
Anti-Inflammatory
(Clove Oil)
EugenolInhibits IL-6, TNF-α
Inhibits COX2
CIO
CIN
CITT
(Testicular)
[246,247,248,249]
Anti-Inflammatory
(Iridoid Monoterpinoid)
MonotropeinUpregulated HO-1
Upregulated Nrf2
CIN[250,251,252]
Anti-Inflammatory
(Synthetic Luteolin)
IsoorientinInhibits MAPK
Signalling
Reduces DAMP Release
Inhibits NF-κB
Signalling
Upregulated HO-1
Upregulates Nrf2
CIN[253]
Anti-Inflammatory
(Calcium Antagonist)
FlunarizineInhibits IL-6, IL-1β, TNF-α
Inhibits MAPK
Signalling
Inhibits NF-κB
Signalling
Upregulates Nrf2
Upregulates HO-1
CIO
CIUN
(Uremic Neuropathy)
[75,254,255,256]
Anti-InflammatoryR-Phenylisopropyladenosine
(R-PIA)
Inhibits TNF-α
Inhibits MAPK
Signalling
Inhibits STAT1
Signalling
Inhibits iNOS
Inhibits COX2
CIO[257]
Solubilised Membrane ProteinThrombomodulin AlfaThrombin-Mediated DAMP Degradation Chemotherapy-IPN[258,259,260]
Anti-Inflammatory
(Nrf2 Activator)
Dimethyl FumarateInhibits Immune Cell
Infiltrations
Upregulation of Nrf2s
Inhibits IL-6, IL4, IL-10, TNF-α
Chemotherapy-IPN
CIPNs
CIN
[261,262,263]
Table 3. Therapeutics for CITs in clinical trials [264,265].
Table 3. Therapeutics for CITs in clinical trials [264,265].
Clinical Trial StatusIntervention
(Name)
Mechanism(s) of ActionListed Target Condition (CIT)Additional
Reference(s)
RecruitingN-Acetylcysteine AntioxidantCIO
(CIN)
[266,267,268,269]
TerminatedSodium Thiosulfate
(Trans-Tympanic Gel)
AntioxidantCIO[270,271]
CompletedGinkgo Biloba Extract
(GBE761)
Antioxidant
Anti-Inflammatory
CIO[272,273,274,275]
CompletedSodium ThiosulfateAntioxidantCIO[270,271]
Not RecruitingSodium Thiosulfate
(+Mannitol)
AntioxidantCIO[270,271]
RecruitingRosuvastatin Cholesterol Reduction
(HMG-CoA Inhibition)
Antioxidant
Anti-Inflammatory
CIO
CIN
[276,277,278,279,280,281,282]
CompletedDexamethasone
(Synthetic Corticosteroid)
Anti-Inflammatory
Antioxidant
CIO[283,284,285,286,287,288]
TerminatedOTO-104
(Dexamethasone Hydrogel)
Anti-Inflammatory
Antioxidant
CIO[289]
RecruitingTempol
(SOD and Catalase Mimetic)
AntioxidantCIN
CIO
[290,291,292,293]
CompletedSilymarinAntioxidant
Anti-Inflammatory
CIN[294,295,296]
CompletedMannitol
(Intravenous)
Osmotic Diuretic CIN[297,298,299,300]
CompletedPreloaded Magnesium Homeostatic
Cisplatin Efflux Regulation
(Downregulated Transporters)
CIN[301,302,303,304,305,306]
CompletedAcetazolamide
+ Mannitol,
(+N-Acetylcysteine)
Alkaline Diuretic
(Carbonic Anhydrase Inhibitor)
CIN[307,308]
RecruitingPantoprazoleCisplatin Influx Regulation
(OCT2 Inhibitor)
Anti-Inflammatory
CIN[309,310]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Domingo, I.K.; Latif, A.; Bhavsar, A.P. Pro-Inflammatory Signalling PRRopels Cisplatin-Induced Toxicity. Int. J. Mol. Sci. 2022, 23, 7227. https://doi.org/10.3390/ijms23137227

AMA Style

Domingo IK, Latif A, Bhavsar AP. Pro-Inflammatory Signalling PRRopels Cisplatin-Induced Toxicity. International Journal of Molecular Sciences. 2022; 23(13):7227. https://doi.org/10.3390/ijms23137227

Chicago/Turabian Style

Domingo, Ivan K., Asna Latif, and Amit P. Bhavsar. 2022. "Pro-Inflammatory Signalling PRRopels Cisplatin-Induced Toxicity" International Journal of Molecular Sciences 23, no. 13: 7227. https://doi.org/10.3390/ijms23137227

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop