Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Apr 15.
Published in final edited form as: Adv Cancer Res. 2022 Apr 26;155:77–129. doi: 10.1016/bs.acr.2022.03.005

Nephrotoxicity in Cancer Treatment: An Update

Chaoling Chen 1, Dengpiao Xie 1, David A Gewirtz 1, Ningjun Li 1
PMCID: PMC11017963  NIHMSID: NIHMS1980859  PMID: 35779877

Abstract

It has been estimated that nearly 80% of anticancer drug-treated patients receive potentially nephrotoxic drugs, while the kidneys play a central role in the excretion of anticancer drugs. Nephrotoxicity has long been a serious complication that hampers the effectiveness of cancer treatment and continues to influence both mortality and length of hospitalization among cancer patients exposed to either conventional cytotoxic agents or targeted therapies. Kidney injury arising from anticancer drugs tends to be associated with preexisting comorbidities, advanced cancer stage, and the use of concomitant non-chemotherapeutic nephrotoxic drugs. Despite the prevalence and impact of kidney injury on therapeutic outcomes, the field is sorely lacking in an understanding of the mechanisms driving cancer drug-induced renal pathophysiology, resulting in quite limited and largely ineffective management of anticancer drug-induced nephrotoxicity. Consequently, there is a clear imperative for understanding the basis for nephrotoxic manifestations of anticancer agents for the successful management of kidney injury by these drugs. This article provides an overview of current preclinical research on the nephrotoxicity of cancer treatments and highlights prospective approaches to mitigate cancer therapy-related renal toxicity.

Keywords: Chemotherapy, targeted cancer therapy, kidney toxicity, risk factor, immune checkpoint, non-coding RNAs, sphingolipids

Introduction

The kidneys represent one of the major sites for the elimination of anticancer drugs. Approximately 80 percent of cancer patients are administered anticancer drugs that potentially have nephrotoxic effects (Launay-Vacher et al., 2007). The nephrotoxicity of anticancer drugs remains a serious complication of cancer therapy, leading to delayed treatment, the lengthening of hospital stays, as well as increased mortality rates for cancer patients (A. Kitchlu et al., 2019; Lameire, Flombaum, Moreau, & Ronco, 2005). Therefore, anticancer drug-induced renal toxicity is increasingly regarded as a principal factor that limits the efficacy of cancer treatments.

Nephrotoxicity is defined as the loss of the kidneys’ ability to filtrate, detoxify and/or excrete harmful metabolites due to anticancer drugs-induced injuries to renal structure and/or function. Drug-induced renal toxicity can affect different cell types in the kidneys including glomerular, tubular, interstitial or vascular cells. Both conventional chemotherapy and targeted agents can cause a diversity of renal complications such as proteinuria, electrolyte imbalance, glomerulopathy, hypertension, interstitial nephritis, and tubulointerstitial damage. Consequently, in many cases, anticancer drugs require dose modification or discontinuation in the setting of renal impairment (Naughton, 2008).

It is also of critical importance to identify risk factors and associated pathogenic mechanisms for nonchemotherapeutic nephrotoxic therapies that could potentially contribute to the deterioration of renal function in cancer patients. In addition to radiation therapy-induced kidney injury (Klaus, Niyazi, & Lange-Sperandio, 2021), concomitant use of analgesics, bisphosphonates, or other chronic disease mediations are common factors leading to renal dysfunction, which need to be taken into consideration (Abhijat Kitchlu et al., 2018; Lameire et al., 2005). Moreover, cancer patients with underlying compromised renal function are prone to develop acute kidney injury (AKI) and subsequent long-term chronic kidney disease (CKD) during anticancer therapy (A. Kitchlu et al., 2019). Unfortunately, the biochemical, physiological and molecular mechanisms underlying anticancer drug-induced renal toxicities remain poorly understood. As nephrological management for most anticancer drugs largely depend on dosage adjustment, discontinuation of treatment and/or symptom management (Chiruvella, Annamaraju, & Guddati, 2020), a comprehensive understanding of nephrotoxic mechanisms of anticancer drugs is necessary to more effectively mitigate nephrotoxicity in cancer patients. This chapter presents an overview of fundamental risk factors associated with nephrotoxicity in cancer patients, findings during the last decade related to some novel molecular pathways putatively involved in the nephrotoxicity as well as efforts to minimize cancer therapeutics-induced nephrotoxicity.

2. Risk factors for renal toxicities in cancer therapy

2.1. General risk factors for nephrotoxicity of anticancer drugs and treatment

A variety of risk factors can exacerbate the course of renal dysfunction and are likely to enhance the nephrotoxicity of cancer therapeutic agents. Renal toxicities can be initiated by the antitumor drugs themselves, elevated doses or prolonged treatment, formation of toxic crystals within intratubular lumens or concomitant use of other nephrotoxic drugs (including aminoglycosides, nonsteroidal anti-inflammatory drugs, radiographic ionic contrast media or other anticancer drugs). In addition, gene variants in hepatic and renal cytochrome P450 (CYP) enzymes or renal transporters that result in decreased metabolism or reduced drug excretion can increase intracellular anticancer drug concentrations, thereby elevating nephrotoxic risk (Miteva-Marcheva, Ivanov, Dimitrov, & Stoyanova, 2020). Fluid deficits are also one of the most common factors that potentiate nephrotoxic effects due to vomiting or diarrhea caused by antineoplastic drugs (Hassan Izzedine & Mark A. Perazella, 2017; Mark A. Perazella, 2009; Perazella, 2012; Perazella & Moeckel, 2010). A spectrum of risk factors for renal toxicity in cancer therapeutics are listed in Table 1.

Table 1.

Risk factors for renal toxicities in cancer therapy

Patient-specific factors
Older age
Female sex
Preexisting AKI or CKD
Hepatic failure
Other comorbidities (multiple myeloma, lymphoma, leukemia, renal cancer, diabetes, sepsis, and acid-base disturbances)
Kidney-specific factors
High rate of blood delivery (20–25% of cardiac output)
Proximal tubular uptake of toxins apical tubular uptake by endocytosis/pinocytosis basolateral tubular transport through OAT and OCT pathways
Relatively hypoxic renal environment high metabolic rate of tubular cells in the loop of Henle Increased drug/toxin concentration in renal medulla and interstitium
Biotransformation of substances to reactive oxygen species causing oxidative stress
Sex difference/ species difference
Tumor cell-induced alternation in expressions of renal transporters
Drug-specific factors
Prolonged dosing periods, rapid infusion rates of drugs
Potent direct nephrotoxic effects of the drug or compound
Concomitant use of nephrotoxic drugs
Competition between endogenous and exogenous toxins for transporters, increasing toxin accumulation within the tubular cell
Insoluble parent compound and metabolite with intratubular crystal precipitation (urine pH/drug pKa)

AKI: acute kidney injury; CKD: chronic kidney disease; OAT: organic anion transporters; OCT: organic cation transporters. Modified from (Mark A. Perazella, 2009).

2.2. Patient-specific factors

Several patient-specific risk factors contribute to increased risk of anticancer agent-induced nephrotoxicity, including underlying disease, advanced age and (female) gender. Those factors can be associated with decreased estimated glomerular filtration rates (eGFR), reduced body water or lower serum albumin levels, resulting in increases in drug concentration. However, there is still no consensus as to sex difference in acute kidney injury (AKI) (Schiffl, 2020). Additionally, preexisting AKI and chronic kidney disease (CKD) may raise the risk for nephrotoxicity. For instance, AKI resulting from intravascular volume depletion results in increased drug exposure to renal cells. Nephrotic syndrome with hypoalbuminemia can lead to decreased drug binding and increased active unbound drug concentrations (Perazella, 2018). Patients with advanced hepatic failure are especially at high risk for drug-induced nephrotoxicity, given that advanced hepatic failure exhibits hypoalbuminemia, intravascular volume depletion, and underlying AKI or CKD (Yeung, Yong, & Wong, 2004). In addition to these factors, comorbidities in cancer patients such as multiple myeloma, lymphoma, leukemia, renal cancer, diabetes, sepsis and acid-base disturbances can also enhance the risk for nephrotoxicity (H. Izzedine & M. A. Perazella, 2017).

2.3. Kidney-specific factors

The kidney and its segments are vulnerable to drugs-induced nephrotoxicity owing to the high renal blood flow rate (25% of cardiac output). The nephrotoxic potential of drugs is highly related to the renal microenvironment. High concentration of drugs or drug metabolites can present in the glomerular ultrafiltrate. Furthermore, the renal cortex is likely to be exposed to high concentrations of toxic metabolites as this portion of the kidney receives approximately 80% of total renal blood flow; consequently, many anticancer drugs can accumulate in proximal tubular cells through both apical uptake and basolateral transport (Drozdzik, Drozdzik, & Oswald, 2021; Enomoto & Endou, 2005; Hucke & Ciarimboli, 2016). In addition to the proximal tubules, epithelial membranes of distal and collecting tubules express several carriers and transporters that facilitate bidirectional movement of substrate molecules. In contrast to the cortex region, the loop of Henle and medullary collecting duct cells in the renal medulla require high metabolic rates for energy-consuming active transport of many solutes; these excess cellular workloads generate hypoxic environments, thereby increasing susceptibility to the nephrotoxicity of anticancer drug therapies (Hassan Izzedine & Mark A. Perazella, 2017; Perazella, 2010).

Renal biotransformation of drugs also plays a critical role in drug-related nephrotoxicity. Renal biotransformation involves several enzyme systems, where the generation of oxidative stress and the formation of injurious reactive oxygen species are likely to damage the kidney. For instance, Glutathione S-transferases (GSTs) are well-known to catalyze the conjugation of glutathione (GSH) to xenobiotics electrophilic compounds, a central step in drug detoxication and biotransformation (Townsend & Tew, 2003). On the other hand, this mechanism could also produce strong mutagens or carcinogens due to GST polymorphisms.

GST-α is primarily expressed in the convoluted proximal tubule with some existing in the thin loops of Henle (Bauchet, Masson, Guffroy, & Slaoui, 2011). GST-π is mainly localized in podocytes, parietal cells of the Bowman’s capsule, distal convoluted tubules and collecting ducts, with significantly lower levels in proximal tubules (Harrison, Kharbanda, Cunningham, McLellan, & Hayes, 1989). Glutathione (GSH) has been found to protect against cisplatin-induced renal and systemic toxicity without affecting its antitumor activity (Y.-Y. Xu, Jiang, Liu, Qu, & Wang, 2012; Zunino et al., 1989); however, biotransformation of trichloroethylene (TCE) conjugated GSH can result in nephrotoxic metabolites such as S-(1,2-dichlorovinyl)-L-cysteine (DCVC) through γ-glutamyltransferase and subsequent β-elimination reaction as well as the metabolite trichloroethylene-cysteine S-conjugate sulfoxide (DCVCS , a highly reactive Michael acceptor) via flavin-containing monooxygenase 3. N-acetylcysteine (NAC, or acetylcysteine) is a widely available nutritional supplement and a medication that can rescue the GSH depletion. NAC has been extensively studied for its antioxidant effects against both cancers and kidney injuries (Kwon, 2021; Mlejnek, Dolezel, Kriegova, & Pastvova, 2021; D. Y. Zhang et al., 2021). NAC has been shown to prevent radiocontrast, cisplatin- and radiation-induced nephrotoxicity in preclinical studies (Güntürk et al., 2019; S. Huang et al., 2019; Mercantepe et al., 2019; Richter & Crannage, 2015). Moreover, a relatively recent study demonstrated the development of tumor-selective GSH-dependent Michael acceptor prodrugs of 6-Mercaptopurine (6-MP) and 6-thioguanine (6-TG) which can have antitumor effects with reduced toxicity in mice (X. Y. Zhang & Elfarra, 2018).

P450 isoenzymes and UDP-glucuronosyltransferase (UGT) also play significant roles in drug biotransformation and detoxification in the kidneys. Of note, some of these enzymes are particularly expressed in the kidneys and specific renal segments (Bauchet et al., 2011; Knights, Rowland, & Miners, 2013). Consequently, alternations in transporter activity such as loss of function mutations and/or competition for carrier proteins/enzymes can hinder parent drug/metabolite excretion and induce nephrotoxicity. Alternation in the activity of these enzymes can be attributed to various factors, such as age, ethnicity, disease states and genetic polymorphisms.

2.3.1. Genetic polymorphisms, sex and other kidney-related factors in renal enzymes

The activity of metabolizing enzymes in the renal tubules can differ between individuals and sexes (Bozina, Bradamante, & Lovrić, 2009). Genetic polymorphisms may affect enzyme expression. Multiple single nucleotide polymorphisms (SNPs) for the organic cation transporter (OCT) genes (SLC22A1, SLC22A2 and SLC22A3) are found to alter their transport functions, where a gain-in-function mutation results in drug accumulation in renal proximal tubule cells (K. M. Huang et al., 2020; Yee et al., 2018; Zazuli et al., 2020). However, Fujita et al showed that a polymorphism in renal ATP-binding cassette transporters (ABC transporters) or renal solute carrier (SLC) transporter did not directly affect cisplatin-induced nephrotoxicity in patients with esophageal cancer (Fujita K, 2016). A study of AKI outcome based on serum creatinine (SCr) showed associations between genetic variants of ERCC1, ERCC2, SLC22A2 and cisplatin-induced nephrotoxicity in adult testicular cancer, suggesting that genetic variations are involved in the inter-individual susceptibility to cisplatin-induced nephrotoxicity (Trendowski et al., 2019).

Interestingly, the expression levels of transporters in renal proximal tubules also differ across species and sexes. A recent study showed that the sex differences in protein abundance of 12 transporters were only significant in rodents and dogs but not in humans (Basit, Radi, Vaidya, Karasu, & Prasad, 2019), indicating that cross-species and sex-dependent protein abundance data should be taken into consideration when evaluating interspecies scaling in drug clearance and assessing kidney toxicity for new therapeutic candidates under development.

2.4. Drug-specific factors

Nephrotoxicity can be augmented and prolonged with repeated drug dosing. Hartmann et al showed that patients who received 2 cycles of a single 50-mg/m2 dose of cisplatin had a 24% decrease in GFR after the second cycle (Hartmann, Kollmannsberger, Kanz, & Bokemeyer, 1999). Patients with higher doses and prolonged durations of treatment with aminoglycosides or amphotericin showed increase nephrotoxicity (M. A. Perazella, 2009). In addition, the cumulative dose, rate of administration, and increased frequency of anticancer drug administration are associated with an increased risk for nephrotoxicity (Caglar et al., 2002; Kobayashi et al., 2016).

Combinations of nephrotoxic drugs with anticancer drugs also contribute to enhanced nephrotoxicity. Many drugs, including angiotensin-converting enzyme (ACE) inhibitors, angiotensin receptor blockers (ARBs), immunosuppressants such as cyclosporins, tacrolimus and nonsteroidal anti-inflammatory drugs (NSAIDs), can influence intraglomerular hemodynamics and decrease glomerular filtration rates (Ghane Shahrbaf & Assadi, 2015; Patel JB, 2021; Perazella, 2005). Furthermore, drugs such as aminoglycosides, amphotericin B, adefovir, foscarnet and cisplatin can potentiate tubular cell toxicity via the formation of free radicals, mitochondrial dysfunction, and transport systems damage (Perazella, 2018). Importantly, the use of drugs that cause drug-related acute interstitial nephritis (AIN) should not be ignored; these include the antituberculosis drug, rifampin, antibiotics, proton pump inhibitors (PPI), NSAIDs and many types of anticancer drugs (Martínez-Valenzuela et al., 2021; Mérida & Praga, 2019; Moledina & Perazella, 2016; Nagata, Ohji, & Iwata, 2019; Perazella & Markowitz, 2010). In addition, chronic interstitial nephritis can arise from medications, including analgesics, lithium, anticancer drugs, and calcineurin inhibitors (Patel JB, 2021). Of note, despite the high frequency of NSAIDs-induced AIN, the occurrence of nephrotic syndrome does not appear to be similarly increased (González et al., 2008). This suggests that patients exposed to NSAIDs-related AIN rarely reach the nephrotic range; instead, they are prone to present increased proteinuria rather than other types of drug-induced AIN (Markowitz & Perazella, 2005). Therefore, monitoring of renal injury and function is crucial for patients receiving combinations of anticancer drugs and other nephrotoxic drugs.

Drugs or metabolites that are insoluble in the urine may cause acute crystalline nephropathy upon precipitation in distal tubular lumens and collecting ducts. This process can occur with or without an interstitial reaction and can result from the reduction of urinary flow rates, change of urine pH/drug pKa, excessive doses and rapid drug infusion rates (Markowitz & Perazella, 2005; Perazella, 2018). Agents such as the antiviral, acyclovir, and the antibiotic, ampicillin, have demonstrated common etiologies of insoluble crystal formation within renal tissue (Chávez-Iñiguez et al., 2018; Garnier et al., 2020; Yarlagadda & Perazella, 2008).

Certain drugs that interfere with renal transporters may also enhance the nephrotoxicity of anticancer agents. For instance, cimetidine, trimethoprim, pyrimethamine and salicylates block tubular secretion of creatinine with or without affecting their glomerular filtration (Andreev, Koopman, & Arisz, 1999). In addition, a variety of drugs can act as substrates competing for a renal transporter and further increase drug deposition in kidneys (Drozdzik et al., 2021; S. Zhou, Zeng, & Shu, 2021). Moreover, Hong et al. demonstrated that repeated contrast-enhanced computed tomography (CECT) within 72 h can predispose cancer patients to contrast-induced nephropathy (CIN) (Hong et al., 2016). Therefore, concomitant use of radiocontrast and NSAIDs should be avoided with nephrotoxic anticancer drugs (Ozkok & Ozkok, 2017; Mark A. Perazella, 2009). The involvement of novel anticancer drugs in AIN and other kidney injuries has been discussed in previous review articles (Hassan Izzedine & Mark A. Perazella, 2017; Martínez-Valenzuela et al., 2021). Renal manifestations associated with commonly used anticancer drugs and recent novel cancer therapies are listed in table 2.

Table 2.

Renal manifestation associated with commonly used anticancer drugs and recent novel cancer therapies

Renal manifestation
Tubular injury Glomerular injury TMA Fanconi syndrome Proteinuria AKI Tubulointerstitial disease
Anticancer agents
Ifofamide MitomycinC Ipilimumab Ifosfamide Bevacizumab Doxorubicin Ipilimumab
Cisplatin Doxorubicin Bevacizumab Lenalidomide Carfilzomib Nivobumab Nivobumab
Doxorubicin Actinomycin Carfilzomib Everolimus Pembrolizumab Pembrolizumab
Cetuximab Bevacizumab Bortezomib Sirolimus Cisplatin Sorafenib
Panitumumab Carfilzomib Clofarabine Carfilzomib Sunitinib
Methotrexate IFN-α Vemurafenib Sirolimus Vemurafenib
Vemurafenib Lenalidomide Clofarabine
Cirzotinib Dabrafenib
Everolimus Vemurafenib

TMA: thrombotic microangiopathy; AKI: acute kidney injury; IFN-α: interferon alpha

3. Nephrotoxicity of chemotherapeutic agents and management

As basic information describing anticancer drug-induced nephrotoxicity has been extensively reviewed and summarized elsewhere in the literature, this review will focus on the most recent findings related to the kidney damage and promising management for the nephrotoxicity of anticancer drugs. A summary of current and potential strategies for the management of cancer therapeutics-induced nephrotoxicity is provided in Table 3.

Table 3.

Summary of current and potential strategies for the management of cancer therapeutics- induced nephrotoxicity

Agent Nephrotoxicity Management Potential Strategies
Platinum agents (Cisplatin) AKI; CKD; Fanconi-like syndrome (hypomagnesemia, fanconi-like syndrome, hypocalcemia, proteinuria, hyperuricemia) Hydration; Mannitol; Furosemide; Magnesium supplement N-acetylcysteine; Inhibition of renal uptake transporters; Reducing inflammation response and cellular death; Use of DPP-4 inhibitors and pioglitazone; Appropriate dose of ACEi, ARB, CCB, aprepitant, thrombin inhibitor or PPI; Targeting GGT enzyme and GGT-associated inflammatory pathway; Activation of Pregnane X receptor; Use of sodium thiosulfate or probiotics; Tempol (an antidoxant)
Alkylating agents (Ifosfamide, cyclophosphamide) Tubular damage (glucosuria, aminoaciduria; Polyuria, and proteinuria); Interstitial inflammation and renal fibrosis Control total dose usage; Monitor and supplementation of electrolyte N-acetylcysteine; Targeting oxidative stress, mitochondrial dysfunction, apoptosis, DPP-4 or vasopressin V2 receptor and use of acetylcysteine
Antitumor antibiotics (Mitomycin C, Doxorubicin, Actinomycin) AKI; Nephrotic syndrome; Endothelial cell and podocyte injury Control total dose usage Combination of cisplatin with mitomycin C, targeting oxidative stress, inflammatory, apoptosis. Inhibition of renal podocyte detachment (angiopoietin-like-3).
Antimetabolites cancer drugs (Methotrexate, Clofarabine) Decreased GFR; Tubular injury; AKI; Proteinuria Hydration; Alkalinization; Hemodialysis Use of Glucarpidase; Targeting oxidative stress and inflammation, apoptosis; Inhibiting miRNA-145–5p-mediated oxidative stress; Targeting ER stress signaling; Restoring OAT3 expression; Use of Rebamipide or HA-230 adsorber; Targeting miR-181a; Use of camel milk, hesperidin and curcumin
EGFR inhibitors (Etuximab, Panitumumab Tubular injury; Glomerupathy; Nephritic syndrome; Electrolyte disorder Glucocorticoid; Discontinuation; Monitor and supplementation of electrolyte Sodium restriction and diuretics treatment
VEGF inhibitors (Bevacizumab, Sorafenib, Sunitinib) Proteinuria; TMA; Hypertension; Glomerulopathy; Electrolyte disorder; Interstitial nephritis ACEi or ARBs Use of PDE5 inhibitor or selective ETA receptor antagonists; Targeting downstream of ET-1 signaling
Immune checkpoint inhibitors (Ipilimumab, Nivolumab, Pembrolizumab) AKI; Acute tubulointerstitial nephritis; Electrolyte disturbance and TMA Glucocorticoids; Discontinuation Use of weak checkpoint blockade; TNF-α blocker
Proteasome inhibitors (Carfilzomib, Bortezomib) TMA; Proteinuria; AKI; Podocyte injury Anti-hypertension; Intravenous hydration; Reduction of dose; ACEI or ARBs Use of acetylcysteine or glucocorticoid; Targeting oxidative stress and inflammation
mTOR protein kinase inhibitors (Sirolimus, Everolimus) AKI; Proteinuria ACEI or ARBs; Discontinuation NA
Biologic agents (IL-2, IFN-α) Hypotension; Edema; Oliguria; MCD; FSGF; TMA Fluid management; Discontinuation Urine output monitoring Low and intermediate-dose of vasopressors (dopamine); Targeting vascular endothelial protein tyrosine phosphatase (VE-PTP)
BRAF inhibitors (Dabrafenib, Vemurafenib) Hyperkalemia; Fanconi syndrome; MCD; AKI; Crystal nephropathy Routine monitoring of electrolytes and serum creatinine Combined therapy of MEK inhibitor, cobimetinib and vemurafenib
ALK inhibitor (Crizotinib) Electrolyte disorder; Renal cyst; Acute tubular necrosis; Glomerular mesangiolysis; Discontinuation Monitoring of renal function and examination of kidney biopsy

AKI: acute kidney injury; TMA: thrombotic microangiopathy; CKD: chronic kidney disease; ACEi: angiotensin-converting enzyme inhibitors; ARBs: angiotensin receptor blockers; DPP-4: dipeptidyl peptidase-4; CCB: calcium channel blocker; GGT: gamma-glutamyl transpeptidase; PPI: proton pump inhibitors; PDE5: phosphodiesterase type 5; MCD: minimal change disease; FSGF: focal segmental glomerulosclerosis; GGT: gamma-glutamyl transpeptidase; ET: endothelin, NA: not available; OAT: organic anion transporters

3.1. Platinum agents

Cisplatin has been used as an effective platinum-containing chemotherapeutic drug for several decades in a wide range of cancers (Brown, Kumar, & Tchounwou, 2019). Cisplatin elicits its antitumor effects through binding to DNA, generation of DNA cross links and damage, a blockade to cell division and, in many cases, the promotion of apoptosis (Tchounwou, Dasari, Noubissi, Ray, & Kumar, 2021). Nephrotoxicity has been recognized as the most critical side effect that limits the clinical use of cisplatin (Sandhya Manohar & Leung, 2018). Current measures to reduce cisplatin-induced nephrotoxicity are limited to hydration and magnesium preloading (McKibbin et al., 2016; Workeneh, Uppal, Jhaveri, & Rondon-Berrios, 2021; Yoshida et al., 2014). Unfortunately, the effectiveness of these approaches is modest, at best. The pathways that contribute to cisplatin-induced kidney damage are complex and include apoptosis, necrosis, necroptosis, oxidative stress, endoplasmic reticulum (ER) stress, mitochondrial dysfunction, autophagy, inflammatory responses, and cell cycle dysregulation (Duan, Cai, Li, & Chen, 2020; Sharp & Siskind, 2017; Y. Xu et al., 2015; J. Zhang, Ye, Tew, & Townsend, 2021). In the course of the last decade, numerous studies have explored potential therapeutic strategies involving inhibition of drug uptake transporters such as organic cation transporter 2 (OCT2) and copper transporter 1 (CTR1), and reducing the inflammation response (McSweeney et al., 2021).

Nonetheless, clinical applications by targeting the above mechanistic pathways in cisplatin-induced kidney toxicity are challenging as many of these molecular pathways are at the same time crucially involved in the cytotoxic activity of cisplatin against tumor cells; consequently, alterations in their functions could possibly interfere with cisplatin-induced anti-tumor effects (Volarevic et al., 2019). Notable efforts have also been made in the potential application of over-the counter (OTC) drugs and prescription drugs against cisplatin-induced nephrotoxicity. The most recent findings on the potential applications of clinically used drugs in cisplatin-induced nephrotoxicity are discussed below.

3.1.1. Prospective use of diabetes drugs

A retrospective study revealed that the dipeptidyl peptidase-4 (DPP-4) inhibitors reduced cisplatin-induced AKI in cancer patients with diabetes mellitus by suppressing inflammation and promoting tubular regeneration. (Takamasa Iwakura et al., 2020; T. Iwakura et al., 2019). Another DPP-4 inhibitor, gemigliptin, was shown to protect against cisplatin-induced nephrotoxicity through the inhibition of apoptosis and inflammatory responses by increasing heme oxygenase-1 (HO-1) and NAD(P)H:quinone oxidoreductase 1 (NQO1) expression (Choi, Leem, & Lee, 2017). The outcome of a clinical trial evaluating the effect of gemigliptin on cisplatin-induced nephrotoxicity is not yet known (ClinicalTrials.gov Identifier: NCT02250872). Cisplatin-induced nephrotoxicity was also found to be prevented by the thiazolidinedione-type diabetes drugs, pioglitazone, via inhibition of the p53-mediated mitochondrial apoptotic pathway via SIRT1 activation (J. Zhang et al., 2020). These findings suggest the potential benefits of these drugs in cancer patient with diabetes, and possibly, non-diabetic cancer patients. However, more clinical data are needed for their use as a renoprotective strategy against cisplatin-induced nephrotoxicity.

3.1.2. Prospective use of antihypertensive agents

Several types of antihypertensive agents appear to represent potential strategies for mitigation of cisplatin nephrotoxicity. One study demonstrated protective effects of enalaprilat, an angiotensin-converting enzyme inhibitor (ACEi), for cisplatin-induced renal toxicity in mice not only by reducing the formation of angiotensin II, but also by reversing cisplatin-induced upregulation of the kinin B1 receptor and decreased aminopeptidase P activity, thereby preventing tubular cell apoptosis and inflammation. (Estrela et al., 2020). In contrast, a recent single-center observational study revealed that patients exposed to target or above target dosage of ACEi or angiotensin II receptor blockers (ARBs) had a higher risk of AKI in emergency medical admissions (Feidakis et al., 2021). Hence, dosage adjustment of ACEi for cisplatin-treated patients should not be overlooked for future potential application in cisplatin-induced nephrotoxicity.

Amlodipine, a calcium channel blocker (CCB), was shown to inhibit the gamma-glutamyl transpeptidase (GGT) enzyme, which metabolizes platinum-glutathione (Pt-GSH)-conjugates to a reactive toxic thiol that causes tubular cell death. Amlodipine can also interfere with the GGT-associated inflammatory pathway (Azouz, Abdel-Nassir Abdel-Razek, & Abo-Youssef, 2020). Although the β-adrenoceptor blockers carvedilol and high-dose propranolol exhibited renoprotection against cisplatin renal dysfunction, they failed to improve cisplatin-induced electrolyte imbalance (Esmaeeli, Keshavarz, Dehdar, Assadi, & Seyedabadi, 2020). As a result, we should carefully interpret the effects of β-adrenoceptor blockers on electrolyte levels in cisplatin-treated patients. Overall, these findings indicate potential uses of these antihypertensive agents in cisplatin-induced nephrotoxicity.

3.1.3. Prospective use of other clinically available drugs

Some other types of clinically available drugs are also found to mitigate cisplatin nephrotoxicity. A thrombin inhibitor, dabigatran (Pradaxa), may offer beneficial effects against cisplatin-mediated nephrotoxicity through inhibiting the thrombin-associated apoptotic and oxidative effects (Ewees et al., 2021). In addition, proton pump inhibitors (PPIs), omeprazole and pantoprazole, were demonstrated to provide protection against cisplatin-induced kidney injury in both in vivo animal studies and randomized controlled clinical trial (ClinicalTrials.gov Identifier: NCT04217512) (Gao et al., 2020; Ghonaim, El-Haggar, & Gohar, 2021; Ismail, El-Awady, & Hassan, 2020). The underlying mechanism for the renoprotection of PPIs could be by suppressing the release and production of inflammatory cytokines stimulated by cisplatin. On the other hand, data from the US FDA adverse event reporting system showed a high correlation between PPIs use and the AKI and CKD events. Hence, monitoring of renal function and dosage modification of PPIs would be necessary in patients receiving cisplatin treatment. Finally, aprepitant is an antiemetic drug provided for cisplatin-treated patients. Interestingly, a novel role for aprepitant was revealed as an effective option for cisplatin-induced nephrotoxicity through anti-oxidative and anti-inflammatory effects (Un et al., 2020).

3.1.4. Prospective targets and other compounds

Several novel mechanisms underlying protective effects against cisplatin-induced renal injury were discovered recently. Pregnane X receptor (PXR), a master transcription factor of xenobiotic detoxification, was shown to play a protective role in cisplatin-AKI mediated by activating the PI3K/AKT pathway (Luan et al., 2021). The hydrogen sulfide metabolite, sodium thiosulfate (STS), was identified as a promising candidate molecule that could protect against renal toxicity following hyperthermic intraperitoneal chemotherapy (HIPEC) with cisplatin; this protection was possibly mediated by the binding of STS to free platinum (Laplace et al., 2020; M. Y. Zhang, Dugbartey, Juriasingani, & Sener, 2021). In addition, supplementation with the probiotics Lactobacillus reuteri and Clostridium butyricum was demonstrated to alleviate cisplatin nephrotoxicity by restoring gut microbiome dysbiosis, thus further reducing uremic toxin production in rats (Hsiao et al., 2021). Other compounds, such as the epoxyeicosatrienoic acid analog (EET-F01) by kidney-targeted delivery, and Pevonedistat, a NEDD8-activating enzyme inhibitor, were recently found to downregulate the expression of inflammatory mediators to relieve cisplatin-induced nephrotoxicity (El-Far & El-Mesery, 2021; Imig et al., 2021).Tempol is a nitroxide that has an antioxidant property and is shown to reverse cisplatin AKI in mice through decreasing kidney injury markers and restoration of aquaporins (AQP2) (Afjal et al., 2020). Currently, Tempol is undergoing clinical trial in head and neck cancer patients with cisplatin and radiation treatment (ClinicalTrials.gov Identifier: NCT03480971).

Overall, the exploration of new targets and mechanism of mitigation by clinically available medications should provide a pathway for the discovery of novel and readily translatable treatments for kidney protection in cancer therapy. Also, targeting the prospective signaling pathways, such as those discussed above, the transcription factor of xenobiotic detoxification, microbiome, supplementation of sodium thiosulfate and kidney-targeted drug delivery, are promising strategies to attenuate kidney injury caused by cisplatin.

3.2. Alkylating agents

Ifosfamide and cyclophosphamide are two of the most widely used alkylating chemotherapeutic drugs that are also currently undergoing clinical trials in combination treatment with different novel cancer therapies (Fox et al., 2021; S. Zhou et al., 2021; Zsiros et al., 2021). This class of drugs is quite frequently associated with kidney dysfunction (Bhat, Kalthur, Padmashali, & Monappa, 2018; Ensergueix et al., 2020). Either the prodrug forms, their metabolites or intermediates may mediate nephrotoxic effects to renal tubular cells (Dobrek, Nalik-Iwaniak, Fic, & Arent, 2020). A retrospective study revealed that in patients who received ifosfamide treatment, kidney biopsies showed tubular necrosis, vacuolation and nuclear atypias in renal epithelial cells, as well as interstitial inflammation and renal fibrosis (Ensergueix et al., 2020). Management of ifosfamide-induced nephrotoxicity has generally been limited to supportive care. However, a growing body of reports have identified several new targets mediating alkylating agents-induced kidney injury through e.g. increased oxidative stress, mitochondrial dysfunction, apoptosis or activation of the arginine vasopressin V2 receptor that leads to anticancer drug-induced nephrogenic syndrome of inappropriate antidiuresis (NSIAD) (S. Kim, Jo, & Kim, 2021).

A recent study found that the antioxidant, Carnosine, alleviated ifosfamide-induced oxidative stress as well as mitochondrial impairment, and consequently attenuated renal injury and electrolyte imbalance (Ommati et al., 2020). Similarly, Annona species ethanolic extracts reduced the renal toxicity of ifosfamide by suppressing oxidative stress, inflammation and apoptosis in a rat kidney model (Abd-Elrazek, Shapana, Shukry, & Galilah, 2021). Mesna is the clinically approved drug to prevent ifosfamide- and cyclophosphamide-induced hemorrhagic cystitis; however, its preventive action towards tubular toxicity of ifosfamide and cyclophosphamide remains undetermined (Reddy V, 2021). In the rat model of cyclophosphamide- and ifosfamide-induced cystitis, the antioxidant, acetylcysteine, which acts as a donor of -SH groups similar to the action of mesna, was reported to produce both uro- and nephron-protective effect against cyclophosphamide and ifosfamide treatment (Dobrek et al., 2020). Alogliptin, a selective inhibitor of the enzyme dipeptidyl peptidase-4 (DPP-4), is an FDA-approved drug for type 2 diabetes. Interestingly, Alogliptin was recently shown to produce beneficial effects in cyclophosphamide-induced nephrotoxicity as indicated by inhibiting the MAP3K/JNK/SMAD3 signaling cascade, which can initiate oxidative stress and production of inflammatory and fibrotic mediators (Salama, Nasr, Abdelhakeem, Roshdy, & ElGamal, 2020). The dual beneficial effects of Alogliptin could be a novel therapeutic approach for cancer patients with diabetes.

A pentadecapeptide derived from Cyclina sinensis was shown to mitigate cyclophosphamide-induced kidney injury by activating the antioxidative enzymes superoxide dismutase, glutathione peroxidase and catalase (Jiang et al., 2020). A more recent study reported that cyclophosphamide-induced inflammation and oxidative stress could be ameliorated by pretreatment with gallic acid, a nature phenolic compound, in mice (Baharmi et al., 2021). Cyclophosphamide is known to be one of the major causes of the syndrome of inappropriate antidiuretic hormone secretion (SIADH) in cancer patients (M. I. Khan, Waguespack, & Ahmed, 2019; Abhijat Kitchlu & Rosner, 2019). Tolvaptan, a selective vasopressin V2 receptor antagonist, is used for the treatment of hyponatremia in patients with heart failure or SIADH in cancer patients. The protective effect of tolvaptan against cyclophosphamide-induced nephrotoxicity was recently demonstrated through the attenuation of apoptosis markers in rat models (El-Shabrawy et al., 2020). While this growing body of evidence does identify new potential strategies for mitigation of alkylating agents-induce nephrotoxicity, it should be noted that those findings need to be confirmed in clinical trials.

3.3. Antitumor antibiotics

Patients receiving antitumor antibiotics such as mitomycin C, actinomycin and doxorubicin can develop nephrotoxicity (Groff, Kozak, Boehmer, Demko, & Diamond, 1997; Y. B. Sun et al., 2013). While mitomycin C was described to be involved in end-stage renal disease, a retrospective report showed that the combination of cisplatin and mitomycin C in laparoscopic hyperthermic intraperitoneal chemotherapy (HIPEC) did not, in fact, increase the incidence of AKI in gastric cancer (Kapoor et al., 2019).

Doxorubicin (adriamycin) has been reported to cause AKI and nephrotic syndrome (Carron, Padilla, & Maurizi Balzan, 2014; Yemm, Alwan, Malik, & Salazar, 2019). More recent studies identified several new compounds/agents to protect against doxorubicin-induced nephropathy, such as omega-3 fatty acids, flavonoids, Acacia hydaspica tannin-rich ethyl acetate fraction and naringenin, through mechanisms of reducing oxidative stress, inflammation, apoptosis as well as renal podocyte detachment (Afsar, Razak, Almajwal, & Al-Disi, 2020; T. H. Khan et al., 2020; Navarro-Hortal et al., 2020; Saleh et al., 2020). A novel compound, YH0618, was reported to reduce renal toxicity of adriamycin through a FOXO4-mediated Bcl-2 Bax/Bcl-2 mechanism (You et al., 2019). Curcumin demonstrated improvement of doxorubicin-induced toxicity in renal podocytes through the activation of Nrf2, inhibition of NF-κB activity, and the upregulation of podocin (Fan et al., 2020). Loss of podocytes is one of the mechanisms involved in adriamycin-induced glomerulosclerosis; however, the mechanistic pathway is unclear. Deficiency of angiopoietin-like-3 (ANGPTL3) was found to protect against adriamycin-induced glomerulosclerosis and podocyte loss in mice (Dai et al., 2019). Though inhibition of ANGPTL3 seems a promising approach against adriamycin nephrotoxicity, it might block the beneficial effect of ANGPTL3 in renal cell carcinoma metastasis (Y.-j. Zhang, Zhang, Feng, & Cao, 2021; Zhao et al., 2019). Therefore, the prospective treatment using the ANGPTL3 inhibitor, evinacumab, which is currently tested in patients with homozygous familial hypercholesterolemia (Raal et al., 2020), in adriamycin-induced nephropathy should be carefully evaluated in patients with renal cell carcinoma metastases (Wilson et al., 2021).

3.4. Antimetabolites cancer drugs

Antimetabolites including methotrexate, 5-fluorouracil, clofarabine, and gemcitabine have been shown to cause renal insufficiency and tubular injury. Methotrexate (MTX) is a classical antifolate, which interferes with folate metabolism. MTX is used to treat a broad range of cancers such as acute lymphoblastic leukemia, head and neck cancer, and breast cancer, among others (Koźmiński, Halik, Chesori, & Gniazdowska, 2020). In addition to plasma MTX concentration monitoring, hydration, and alkalinization to prevent/ameliorate the MTX-induced nephrotoxicity (Howard, McCormick, Pui, Buddington, & Harvey, 2016), recent consensus guidelines and case reports have suggested that glucarpidase (Carboxypeptidase G2) can provide nonrenal elimination of MTX and should be used with folic acid rescue to attenuate MTX-induced nephrotoxicity (Misra, Santagostino, Dine, & Bonhomme Faivre, 2019; Ramsey et al., 2018; Young et al., 2019).

A number of studies have identified natural compounds and other potential approaches to mitigate nephrotoxicity of MTX. Natural compounds such as Rosmarinic Acid and Apigenin that exhibit antioxidant and anti-inflammatory properties have been reported to attenuate MTX nephrotoxicity in rodent models (Jafaripour et al., 2021; Sahindokuyucu-Kocasari, Akyol, Ozmen, Erdemli-Kose, & Garli, 2021). Another natural product, Dioscin, can relieve MTX-induced kidney damage via inhibiting miRNA-145–5p-mediated oxidative stress (Y. Li et al., 2021). A MTX metabolite, 7-OH MTX, has been considered as a primary toxic metabolite responsible for nephrotoxicity due to its lower water solubility than MTX (Holmboe, Andersen, Mørkrid, Slørdal, & Hall, 2012). A study found that Nobiletin, a flavonoid isolated from Citrus aurantium L, could reduce 7-OH MTX nephrotoxicity via endoplasmic reticulum stress-dependent PERK/CHOP signaling and protect tubular cell survival (Song et al., 2021). A compound modified from Paeoniflorin, paeoniflorin-6′-O-benzene sulfonate (CP-25), showed protection against MTX-renal toxicity by preventing tubular cells apoptosis and facilitating MTX excretion through recovering OAT3 expression (Wei et al., 2021).

Rebamipide is a gastroprotective drug for the treatment of gastric ulcers and gastritis. It was found that Rebamipide could potentially mitigate nephrotoxicity of MTX through activation of NRF-2/SIRT-1/FOXO-3 and mTOR/PI3K/AKT signaling while inhibiting NF-κB-p65/TLR-4 (Elmansy, Seleem, Mahmoud, Hassanein, & Ali, 2021). Moreover, using HA-230 adsorber in hemadsorption procedure was found to be a new therapeutic approach to reduce MTX toxicity in pediatric patients with acute lymphocytic leukemia who had delayed MTX clearance after high-dose MTX treatment (Sazonov et al., 2021).

5-fluorouracil (5-FU), another antimetabolite cancer drug, has multiple mechanisms of action through inhibition of DNA synthesis and misincorporation into DNA. Although patients with renal impairment do not require dose adjustment of 5-FU (Lexicomp, 2022), there is increasing in vivo and in vitro evidence that 5-FU can cause renal dysfunction as indicated by the promotion of apoptosis, induction of oxidative stress, and tubular injury. Recent studies have demonstrated potential approaches to mitigate 5-FU nephrotoxicity. Inhibition of miR-181a has been found to attenuate 5-FU-induced mesangial cell apoptosis, inflammation and kidney injury (X.-Y. Liu et al., 2018). Camel milk is shown to have renoprotection against 5-FU, which could be mediated by suppressing MAPKs, NF-kappaB and PI3K/Akt/eNOS (Arab, Salama, & Maghrabi, 2018). In addition, hesperidin and curcumin play beneficial roles in 5-FU-induced nephrotoxicity by inhibiting oxidative stress, lipid peroxidation, apoptosis, and renal dysfunction (Gelen et al., 2021). Herein, we have summarized some recently discovered targets and pathways, such as nonrenal metabolizing enzyme, transporter, the utilization of HA-230 adsorber and natural compounds, which could be used as potential therapeutic strategies in MTX and 5-FU nephrotoxicity in the future.

3.5. Epidermal growth factor receptor pathway inhibitors (EGFR inhibitors)

EGFR inhibitors have been used in the treatment of various malignancies including colon, head, neck, and non-small cell lung cancers. Monoclonal antibodies such as cetuximab and panitumumab are EGFR inhibitors associated with tubular toxicity, glomerulopathies and electrolyte disorders. (H. Izzedine, Boostandoost, & Mathian, 2017; Jhaveri, Wanchoo, Sakhiya, Ross, & Fishbane, 2017). Panitumumab-induced immune complex glomerulonephritis could be mitigated by drug discontinuation and glucocorticoids treatment (H. Izzedine et al., 2017). Hypomagnesaemia of EGFR inhibitors is the most common side effect that could lead to renal toxicity (Jhaveri et al., 2017). Therefore, electrolyte disturbances should be carefully monitored and managed with subsequent fluid and sodium restriction, diuretics treatment, and magnesium supplementation or discontinuation of EGFR inhibitors.

3.6. Vascular endothelial growth factor pathway inhibitors (VEGF inhibitors)

The vascular endothelial growth factor (VEGF) plays an essential role in angiogenesis, which provides oxygen and nutrients to support tumor growth and metastasis. The VEGF inhibitors bevacizumab, sorafenib and sunitinib are clinically used for metastatic colon cancer, rectal cancer, non-small cell lung cancer, and breast cancer, among others (Qin et al., 2019). Common renal toxicities of VEGF inhibitors include proteinuria, glomerular disease and thrombotic microangiopathy (TMA) (Hanna et al., 2019; Shye et al., 2020). Currently, there are no molecule-specific targeted therapies for VEGF inhibitor-induced renal adverse effects, except for supportive care, dose reduction, medication discontinuation or treatment with ACEi or ARBs for proteinuria. It should be noted that ACEi and ARBs also contribute to an increased risk of AKI under certain conditions. Hence, it would be appropriate to monitor renal function and individualize treatment in patients with VEGF inhibitor if using ACEi or ARBs for renal side effects (Porta et al., 2020).

Results from a rat experimental model demonstrated that the phosphodiesterase type 5 (PDE5) inhibitor, sildenafil, reduced sunitinib-induced proteinuria (S. Lankhorst et al., 2014). Unfortunately, the clinical application of sildenafil in sunitinib-induced nephrotoxicity has not yet been verified. A better understanding of the underlying mechanism(s) of VEGF inhibitor-induced nephrotoxicity could lead to the development of a selective compound or adjuvant drug to minimize the off-target effects of VEGF inhibitors. The side effects of VEGF inhibitors are partially attributed to the off-target effect from VEGF inhibitors-induced circulating endothelin-1 levels (Stephanie Lankhorst et al., 2015). A recent study showed that sunitinib-induced albuminuria can be improved by a selective endothelin (ETA) receptor antagonist (sitaxentan) but not ETA/B receptor antagonist (macitentan) in rats. Sitaxentan and ambrisentan are FDA-approved selective ETA receptor antagonists for pulmonary hypertension treatment. Despite the withdrawal of Sitaxentan, ambrisentan seems a promising therapeutic approach against VEGF inhibitor-induced nephrotoxicity. Moreover, it is possible that targeting downstream of the ET-1 signaling pathway could also provide the renoprotection against VEGF inhibitors (Mirabito Colafella et al., 2020). Therefore, it is clear that additional research is needed to develop new strategies for mitigation of VEGF inhibitor-induced nephrotoxicity.

3.7. Immune checkpoint inhibitors

Immune checkpoint inhibitors (CPIs) directly target inhibitory receptors on immune and tumor cells, allowing for the activation of T cells and antitumor action. These receptors include programmed cell death 1 ligand (PD-L1) and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4). CPIs can reactivate the immune cells to attack the cancer cells (Robert, 2020; Weber, 2010). Immune checkpoint inhibitors such as CTLA-4 antagonists (ipilimumab) and PD-L1 antagonists (nivolumab and pembrolizumab) are approved by the FDA for treatment of renal cell cancer, non-small lung cancer, and melanoma (Wanchoo, Karam, et al., 2017). Extensive evidence has shown that CPIs are often associated with the incidence of AKI and acute interstitial nephritis (AIN) (Belliere et al., 2016; Cortazar et al., 2020; Gupta et al., 2021; Oleas et al., 2020; Patel et al., 2020). Current management for CPIs-induced AIN involves treatment with glucocorticoids (GCs) or discontinuation of the medications (S. Manohar et al., 2019; Oleas et al., 2020; Qu et al., 2021). A recent multicenter study recommends that patient can return to CPIs treatment after renal injury has been relieved (Cortazar et al., 2020; Koks et al., 2021); nonetheless, fewer than 50% of patients receiving GCs can fully recover from kidney injury. A retrospective study reported that infliximab, an TNF-α (tumor necrosis factor) blocker, would be another treatment option for relapsed CPI-AIN in patients who do not respond to or tolerate the side effects of GCs (J. S. Lin et al., 2021). However, a larger sample size and standardization in the timing of the infliximab treatment in CPI-AIN are required for future clinical studies.

A study suggested that the checkpoint inhibition was not a major mechanism for efficacy of anti-CTLA-4 antibodies. The investigators identified a weak checkpoint blockade by the anti-CTLA-4 antibody, GIGA-564, which had reduced kidney damage compared with the CTLA-4 antagonist, ipilimumab, in murine models without affecting antitumor effects. This finding suggests a novel strategy for optimizing anti-CTLA-4 drugs based on regulatory T cells instead of checkpoint inhibition (Stone et al., 2021). Considering the very limited strategies available for mitigation of CPIs-induced kidney adverse effects, further research should emphasize the structure or affinity-guided design of antagonist/inhibitors for immune cells or tumor cells.

3.8. Proteasome inhibitors

Carfilzomib is a proteasome inhibitor used to treat patients with relapsed and refractory myeloma. Carfilzomib-induced renal manifestations include thrombotic microangiopathy (TMA), proteinuria and AKI (Fotiou, Roussou, Gakiopoulou, Psimenou, & Gavriatopoulou, 2020). The pathophysiology behind carfilzomib-induced nephrotoxicity is associated with tumor lysis syndrome, endothelial injury, and podocyte injury (Fotiou et al., 2020). Current management for acute renal function impairment is achieved by hydration and drug discontinuation (Bringhen et al., 2019). A case report showed that acetylcysteine could prevent carfilzomib-induced vasoconstriction-related renal injury (Wanchoo, Khan, Kolitz, & Jhaveri, 2015). Rutin, a bioflavonoid, ameliorates carfilzomib-induced oxidative stress and inflammation in nephrotoxicity through the inhibition of the iNOS-mediated NF-κB signaling pathway (Al-Harbi et al., 2019).

Another proteasome inhibitor, Bortezomib, is effective in multiple myeloma and mantle cell lymphoma. Common renal side effects of bortezomib include TMA and acute interstitial nephritis (AIN) (Chiruvella et al., 2020). A case reported showed that the bortezomib renal impairment was improved after glucocorticoid therapy and discontinuation, but AKI reoccurred following reinitiating bortezomib (Cheungpasitporn et al., 2015). Therefore, the utilization of glucocorticoid therapy for bortezomib-induced AIN remains undetermined.

3.9. mTOR protein kinase inhibitors

The mammalian target of rapamycin (mTOR) is a serine/threonine kinase, which regulates many biological processes, including cell survival, proliferation, autophagy, and metabolism (Hua et al., 2019). Inhibitors of mTOR, sirolimus and everolimus, are used for the treatment of breast cancer, renal cell carcinoma and pancreatic neuroendocrine tumors (Roskoski, 2019). It has been shown that both everolimus and sirolimus induce proteinuria and AKI. Although the chances of mTOR inhibitors leading to life-threatening AKI is minimal, they may cause development of CKD and other long-term complications (Chandra, Rao, Malhotra, Rastogi, & Khurana, 2017; Paluri et al., 2019; Wanchoo, Abudayyeh, et al., 2017). Novel therapeutics and strategies are not currently available. Current strategies to mitigate mTOR inhibitors-induced nephrotoxicity depend primarily on monitoring of renal injury and proteinuria, early use of ACEi and ARBs and drug discontinuation.

3.10. Biologic agents

Biologic agents such as interleukin-2 (IL-2) and interferon alpha (IFN-α) have utility in cancer therapy. High-dose interleukin-2 (IL-2) is effective in metastatic renal cancer, and metastatic melanoma (Marabondo & Kaufman, 2017; Perazella & Shirali, 2018). However, high-dose IL-2 causes severe hypotension and cytokine-mediated inflammation, leading to AKI (Guleria et al., 1994; Marabondo & Kaufman, 2017). Current managements for IL-2 nephrotoxicity are primarily reliant on urine output monitoring and fluid management, which can improve mild oliguria or increased serum creatinine caused by IL-2 (Marabondo & Kaufman, 2017). Low and intermediate-dose of vasopressors such as dopamine can also be used in the treatment of hypotension and oliguria (Perazella & Shirali, 2018). However, advanced therapeutic approaches are limited.

A novel mechanism underlying IL-2-induced vascular leak syndrome is associated with increased circulating angiopoietin-2 levels. Endothelial damage is one of the major causes of AKI. A study revealed that diabetes mellitus can increase angiopoietin-2. Angiopoietin-2 is the competitive antagonist for angiopoietin-1 and a partial agonist/antagonist of the receptor tyrosine kinase TIE2 in endothelial cells (Yuan, Khankin, Karumanchi, & Parikh, 2009). Increased circulating angiopoietin-2 leads to vascular wall destabilization and promotes neovascularization (Fiedler & Augustin, 2006). Vascular endothelial protein tyrosine phosphatase (VE-PTP) is involved in balancing TIE2 signaling to stabilize the vasculature. Inhibition of VE-PTP by AKB-9778 can reset TIE2 signaling and therefore decrease serum angiopoietin-2 level and rescue vascular stability (G. Li, Sachdev, Peters, Liang, & Lotze, 2019). In addition, targeting VE-PTP phosphatase was demonstrated to protect against diabetic kidney injury (Carota et al., 2019).

Therefore, VE-PTP inhibitor AKB-9778 could be a promising candidate to attenuate IL-2-induced vascular leak and endothelial dysfunction which could lead to AKI.

3.11. BRAF inhibitors

BRAF is a member of rapidly accelerated fibrosarcoma kinase family proteins, and also a proto-oncogene. BRAF inhibitors (dabrafenib and vemurafenib) are approved for the treatment of BRAF V600E mutation-positive melanoma (Holderfield, Deuker, McCormick, & McMahon, 2014). BRAF inhibitors can cause tubular interstitial nephritis, acute tubular necrosis, increases of serum creatinine, proteinuria, and electrolyte disorders, including hypophosphatemia, hyponatremia and hypokalemia (Wanchoo, Jhaveri, Deray, & Launay-Vacher, 2016). Overall, compared with vemurafenib, dabrafenib is associated with a lower incidence of kidney disease (Wanchoo et al., 2016). A vemurafenib-induced increase in serum creatinine is usually immediate and reversible (Hurabielle et al., 2016). Routine monitoring of electrolytes and serum creatinine are recommended during treatment with BRAF inhibitors (Hurabielle et al., 2016). A recent study showed that combined therapy of MEK inhibitor, cobimetinib, and BRAF inhibitor, vemurafenib, in the treatment of BRAF V600-mutated metastatic melanoma exhibited a 60% reduction of AKI compared with BRAF inhibitor monotherapy (Teuma et al., 2017). Therefore, it is important to note that patients with different combined therapies of BRAF and MEK inhibitors could result in different responses in renal disorders (Meirson, Asher, Bomze, & Markel, 2020). In this regard, future investigations should also highlight the optimal timing of drug switching and the best combination therapy to reduce renal toxicities.

3.12. Anaplastic lymphoma kinase inhibitor

Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase, which regulate diverse cellular process, including cellular proliferation, survival, and proliferation. Mutation of ALK is associated with the development of neuroblastoma, diffuse large B-cell lymphoma, anaplastic large-cell lymphoma, myofibroblastic tumor, esophageal and squamous cell carcinoma, as well as other malignancies (Webb et al., 2009). Crizotinib is an ALK inhibitor for the treatment of non-small cell lung cancer with the ALK mutation. To this point in time, only a few cases of crizotinib-associated nephrotoxicity have been described. Electrolyte disorder and renal cyst are common adverse events in crizotinib treatment, which can be reversed after crizotinib discontinuation (H. Izzedine, El-Fekih, & Perazella, 2016; Y. T. Lin et al., 2014). Crizotinib was reported to increase serum creatinine and reduce the eGFR, which could be recovered after cessation of therapy (Brosnan et al., 2014; Camidge, Brosnan, DeSilva, Koo, & Chonchol, 2014; Martín Martorell, Huerta Alvaro, Solís Salguero, & Insa Molla, 2014). Recently, kidney biopsy data on cirzotinib-induced renal injury showed that cirzotinib induced acute tubular necrosis and glomerular mesangiolysis after drug rechallenge (Gastaud et al., 2013). Another case study described arteriolar myocyte vacuolization following crizotinib treatment (Hassan Izzedine, Brocheriou, Amoura, & Mathian, 2021). However, the mechanism of crizotinib-induced kidney damage remains unknown. In a rodent study, it was demonstrated that long-term crizotinib treatment could induce fibrosis and renal dysfunction via activating the TNF-α/NF-κB signaling pathway (Yasuma et al., 2018). It is also speculated that the crizotinib-induced kidney damage can be mediated by the inhibition of the mesenchymal epithelial transition growth factor (c-Met) pathway (Hassan Izzedine et al., 2021). Due to limited data and unclear pathophysiological explanations for the kidney damage induced by crizotinib, careful monitoring of renal function and examination of kidney biopsies during crizotinib therapy are recommended.

4. Renoprotective effects of some novel anticancer therapeutics

Because emerging preclinical and clinical studies are devoted to developing and testing promising novel anticancer drugs, efforts should be made to prevent/reduce the potential nephrotoxicity. Developing and discovering an anticancer drug that exhibits dual action of anti-tumor and renoprotection would be very beneficial for cancer patients. Therefore, identifying the common targets and mechanisms shared by both carcinogenesis and nephrotoxicity could serve to develop a novel and effective drug that simultaneously provides anticancer as well as renoprotective activities. Herein, we discuss some strategies associated with novel anticancer therapies in mitigating renal toxicities in the context of the immune system, sphingolipid signaling, DNA repair, histone modifications, and non-coding RNAs.

4.1. The immune system

The innate immune system contributes to the destruction of tumor cells that present tumor antigens through various pattern recognition receptors (PRRs), including toll-like receptors (TLRs) and macrophage-inducible C-type lectin (Mincle). TLRs can regulate immune responses both positively and negatively (Bai et al., 2020). A specific TLR2 agonist designed by Feng et al can generate macrophages that have strong anti-tumor properties in mice (Feng et al., 2019). Furthermore, the combination of TLR9 agonists and immune checkpoint inhibitors can maximize TLR9-incduced T-cell activity and provoke the antitumor immune response (Buss & Bhatia, 2020; Chuang et al., 2020). TLRs have been recognized to play multifaceted roles in AKI (Habib, 2021). TLR-2 and TLR-9 were shown to protect against cisplatin-caused nephrotoxicity in mice (Alikhan et al., 2016; Andrade-Silva et al., 2018). However, recent studies showed that depletion of TLR9 reduced renal ischemia-reperfusion injury and that nanoparticle-mediated selective targeting of renal tubular TLR9 decreased renal tubular inflammation, apoptosis and necrosis after ischemia reperfusion (Han et al., 2020; Zheng et al., 2021). These apparent discrepancies could be due to disease/cells-dependent effects of TLR9. In addition to the anticancer effects, TLR agonists may at the same time exert renal protective effects. However, care should be taken in the interpretation of these preclinical data because either overaction of TLRs by agonist or deletion/blockade of TLRs can dysregulate tissue repair during different phases of AKI in human.

Mincle (Macrophage-inducible C-type lectin), a pattern recognition receptor, was recently demonstrated as a novel target for cancer treatment (C. Li et al., 2020). Furthermore, it was found to be associated with M1 macrophage activation during cisplatin-induced AKI. Interestingly, adoptive transfer of Mincle-knockdown macrophages reduced the nephrotoxicity caused by cisplatin (Inoue, 2017). Additionally, the natural anti-inflammatory compound curcumin elicits inhibitory effect on the Mincle-maintained M1 macrophage phenotype, providing a promising therapeutic strategy for cisplatin-induced nephrotoxicity (Tan et al., 2019).

Cytokines released from innate immune cells are responsible for regulating the host innate immune response toward tumor cells and promoting their apoptosis. Cytokine therapy has drawn attention to treat cancers in recent years (Xue, Hsu, Fu, & Peng, 2021). Interestingly, an IL-2 and IL-33 hybrid cytokine, IL233, was shown to protect mice from cisplatin- and doxorubicin-induced nephrotoxic injury in mice (Sabapathy, Cheru, Corey, Mohammad, & Sharma, 2019; Stremska et al., 2017). Thus, modulation of the immune system could be a promising strategy for mitigation of nephrotoxicity in cancer treatment.

4.2. Sphingolipid signaling

Sphingolipids, including ceramide, sphingosine and sphingosine-1-phosphate (S1P) have been proposed to participate in important cellular functions, such as cell growth, cell senescence, differentiation, cellular inflammation and cell cycle regulation (Hannun & Obeid, 2018). It has been increasingly recognized that inflammatory mediators, growth factors, and cellular stress could disrupt the balance between ceramide-induced cell death and S1P-induced cell growth, which are responsible for the cell fate. Sphingosine kinases (SPHKs) represent two isoforms, SPHK1 and SPHK2, that catalyze the conversion of sphingosine to sphingosine-1-phosphate (S1P). SPHKs are shown to be involved in tumorigenesis in many types of cancers (Pitman, Oehler, & Pitson, 2021; Pyne & Pyne, 2020). Recently, SPHK1 and SPHK2 inhibitors have been under development to treat breast cancer, ovarian cancer and cholangiocarcinoma (Alshaker, Thrower, & Pchejetski, 2020; Ding et al., 2016; F. I. Khan, Lai, Anwer, Azim, & Khan, 2020). ABC294640, a specific SPHK2 inhibitor, enhanced the antitumor effects of TNF-related apoptosis-inducing ligand (TRAIL) by inducing apoptosis in non-small-cell lung cancer (Yang et al., 2015). Moreover, ABC294640 (Yeliva ®) is currently being tested in a phase IIa clinical trial with hydroxychloroquine sulfate in treatment of patients with advanced cholangiocarcinoma (ClinicalTrials.gov Identifier: NCT03377179). It has also been reported that the antitumor action of ABC294640 can be enhanced by sorafenib, a multiple tyrosine kinase inhibitor in human cholangiocarcinoma cells (Ding et al., 2016; Evangelisti et al., 2016).

Despite the role of sphingolipids in cancers, they are increasingly being viewed as bioactive factors in the regulation of renal physiology. It has been shown that the activation of the sphingosine kinase/S1P/S1P receptor (SphK/S1P/S1PR) pathway contributes to different kidney diseases (Lyu, Wang, Ji, Ritter, & Li, 2020; Yokota, Bhunu, Toba, & Intapad, 2021; Xiwen Zhang, Ritter, & Li, 2018; X. Zhang, Wang, Ji, Ritter, & Li, 2019). A more recent study demonstrated that oral administration of ABC294640 could attenuate cisplatin-induced nephrotoxicity in mice (Xie et al., 2020). Therefore, sphingolipid signaling is a novel potential target for anticancer agent-induced nephrotoxicity in combination with the antitumor effect.

4.3. DNA repair pathway

Many antitumor therapies, including antimetabolites (5-fluorouracil, Methotrexate), DNA cross linking and alkylating agents (cisplatin, temozolomide, ifosfamide), topoisomerase II inhibitors (etoposide, doxorubicin), and radiotherapy, target proliferating cells. Several inhibitors of DNA damage repair have been tested in clinical trials; these include poly (ADP-ribose) polymerase (PARP) inhibitors, ataxia-telangiectasia mutated (ATM) inhibitors, checkpoint kinases (CHK1/2) inhibitor and DNA methyltransferases (DNMT1) inhibitor (Chan, Tan, & Cornelissen, 2021; Isono, Okubo, Asano, & Sato, 2021; Smith, Southgate, Tweddle, & Curtin, 2020; Wong, 2020; Yap et al., 2021). Generally, these agents could impair DNA repair ability or cause excessive DNA damage resulting in cell death following DNA replication (R. Huang & Zhou, 2021). Herein, we discuss the current inhibitors targeting DNA damage repair and their role in kidney injury.

Poly (ADP-ribose) polymerase (PARP), PARP-1:

Poly (ADP-ribose) polymerase (PARP) is responsible for single-stranded DNA break (SSB) or double-strand DNA break (DSB) repair. The occurrence of SSB or DSB will increase the activity of PARP-1, enhancing poly (ADP-ribose) (PAR) activity, which in turn helps synthesize long branched PAR chains in order to recruit base excision repair (BER) enzymes to the damage site (Cerrato, Morra, & Celetti, 2016; Y. Huang et al., 2018). Several PARP inhibitors have been approved for the treatment of breast and ovarian cancers. However, three PARP inhibitors (olaparib, rucaparib, and velaparib) also increase serum creatinine by inhibiting proximal tubular transporter channels such as multidrug and toxin extruder 1 and 2 (MATE1 and MATE 2) and the organic cationic transporters 1and 2 (OCT 1 and OCT 2) (LaFargue, Dal Molin, Sood, & Coleman, 2019). A more recent study showed that creatinine-derived eGFR may not actually reflect the renal function affected by olaparib. An alternative renal marker, cystatin C should be considered to more accurately measure eGFR in patients taking olaparib (Bruin et al., 2021), which would be more helpful in the evaluation of kidney damage by this class of drugs. A clinical trial is active to test whether cystatin C could be an early renal function marker for children with nephrotoxic chemotherapy (ClinicalTrials.gov Identifier: NCT02822404).

Recently, niraparib (ZEJULA), a PARP inhibitor, was approved in 2020 for patients with advanced ovarian cancer following front-line platinum treatment (González-Martín et al., 2019). Based on previous studies, it is not necessary for the renal adjustment of niraparib dosage in patients with mild or moderate declines in kidney function (Deshpande, Perazella, & Jhaveri, 2021; Zibetti Dal Molin et al., 2020). However, it remains undetermined to what extent dosing modification for PARP inhibitors might be indicated in cancer patients with advanced CKD and end stage kidney disease (ESKD). It is still unclear about the long-term effects of PARP inhibitors on kidney function and proteinuria (Deshpande et al., 2021). Therefore, it should be noted that patients could develop nephrotoxicity due to preexistent changes in intrarenal homeostasis (Lazareth et al., 2020). While PARP-1 inhibition was demonstrated to attenuate ischemic AKI (Jang et al., 2020), one study revealed that PARP-1 deficiency promoted an alkylating agent methyl methanesulfonate (MMS)-induced nephrotoxicity in alkyladenine DNA glycosylase-transgenic mice, which was sex dependent, i.e. MMS-induced nephrotoxicity was observed in male, but not female mice (Calvo et al., 2016). This discrepancy of PARP inhibition impact in kidney damage could be due to different phases of AKI caused by different insults. Therefore, more studies are required to determine the role of PARP on anticancer agents-induced nephrotoxicity.

4.4. Histone modifications

4.4.1. Histone acetyltransferases (HATs)

Histone acetyltransferases (HATs) are the enzymes responsible for transferring an acetyl group to the lysine residue of cellular proteins, including transcription factors and histones, to promote gene expression. p300, a member of HATs, plays a definitive role in cell proliferation, differentiation, and apoptosis. p300 has been implicated in tumorigenesis and epithelial-mesenchymal transition in non-small cell lung cancer cells (Hou et al., 2018; Iyer, Özdag, & Caldas, 2004). Cisplatin was reported to induce kidney injury through activating one member of HATs, p300 activity, thereby increasing acetylation of histone H3 and further enhancing oxidative stress, inflammation, and apoptosis. It was shown that a potent p300 inhibitor, Garcinol, reversed cisplatin-induced kidney injury (J.-Y. Kim, Jo, Leem, & Park, 2020). It is speculated that targeting HAT could be a potential strategy to treat anticancer drug-induced kidney damage. Interestingly, p300 inhibition has demonstrated anticancer effects (Liu et al., 2020; Y. M. Wang et al., 2017). Targeting p300 may provide dual action of protection against renal chemotoxicity and an enhanced antitumor effect.

4.4.2. Histone deacetylases (HDACs)

Histone deacetylases (HDACs) are enzymes involved in the removal of acetyl group from lysine residue of histones or non-histones proteins. HDACs are grouped into four classes, which have distinct functions. HDACs participate in tumorigenesis in several cancers (Pant, Peixoto, Richard, & Gradilone, 2020; P. Wang, Wang, & Liu, 2020). HDAC inhibitors have shown synergistic antitumor effects in combination with other cancer drugs in preclinical and clinical tests (Hontecillas-Prieto et al., 2020; Jenke, Reßing, Hansen, Aigner, & Büch, 2021). Emerging studies suggest that HDAC inhibitors are a potential strategy to decrease the AKI induced by cisplatin. For example, HDAC inhibitors such as trichostatin A (TSA) and suberoylanilide hydroxamic acid (SAHA) are effective in reducing cisplatin-induced AKI through increasing autophagy in renal tubular cells (J. Liu et al., 2018; Tang et al., 2018). More recently, a highly selective HDAC6 inhibitor, 2-Methylquinazoline derivative 23BB, was demonstrated to improve cisplatin-induced AKI (Hao et al., 2020). Another selective HDAC6 inhibitor, Ricolinostat (ACY-1215), elicited suppressive effects on TGF-β and EGFR signaling pathways to mitigate kidney damage in obstructive nephropathy (Chen et al., 2020). Given their antitumor potential, HDAC inhibitors could represent an additional candidate to protect against renal chemotoxicity and simultaneously enhance the antitumor action of cisplatin.

4.4.3. Sirtuins (SIRT)

Sirtuins (SIRT) are NAD+-dependent histone deacetylases belonging to class III HDACs. Unlike other HDACs, the SIRT are not affected by the inhibitors of HDACs. Different SIRTs have distinct roles in cancers as some SIRTs act as oncoproteins or as tumor suppressors. Accumulating evidence have indicated diverse roles of SIRT in kidney injuries (Peasley, Chiba, Goetzman, & Sims-Lucas, 2021). Activation of SIRT1, SIRT3 or SIRT6 has demonstrated attenuation of cisplatin-induced kidney damage through various mechanisms such as repressing apoptosis, inflammation and oxidative stress (J. Y. Kim et al., 2019; Z. Li et al., 2018; Yoon & Kim, 2016). Additionally, SIRT5 mediates the balance between mitochondrial and peroxisomal fatty acid oxidation in proximal tubular epithelial cells, which protect against injury in AKI (Chiba et al., 2019). Downregulation of SIRT3 by cisplatin can be restored through PARP-1 inhibition, which mediates protection against cisplatin-induced oxidative stress in tubular cells (Yoon & Kim, 2016). Consistent with this finding, deletion of SIRT3 exacerbates cisplatin nephrotoxicity via increasing apoptosis and inflammatory response (D. Kim et al., 2018). Moreover, upregulation of SIRT6 inhibits ERK1/2 expression and thereby mitigates renal dysfunction, inflammation and apoptosis caused by cisplatin (Z. Li et al., 2018). Recently, SIRT2 overexpression was show to reverse cisplatin-downregulated mitogen-activated protein kinase phosphatase-1 (MKP-1) and further ameliorate renal injury (Jung, Park, Kang, & Kim, 2020). Hence, manipulation of SIRT could be a potential approach to simultaneously mitigate cancer and anticancer drugs-induced nephrotoxicity by targeting various pathways. However, it should be noted that either overactivation or deactivation of HDAC or SIRT could lead to opposing effect in cancers and kidneys. Further testing in animal studies and human are needed due to distinct functions of HDACs and SIRTs.

4.5. Non-Coding RNAs

Non-Coding RNAs, such as miRNA, lncRNAs, and circRNAs, are believed to account for a variety of physiological and pathological functions at both transcriptional and posttranscriptional regulations. In addition to numerous studies in cancers, they have gained increasing attentions from researchers in the kidney area in the recent decade. Their roles in cisplatin-induced renal injuries have also been extensively studied (Du et al., 2017; Guo et al., 2018; Loren et al., 2021; Pavkovic et al., 2016). A natural steroid saponin, Dioscin, exerts antioxidant effect against cisplatin-induced nephrotoxicity through a mechanism of increased SIRT1 expression regulated by microRNA-34a (Y. Zhang et al., 2017). Urolithin A (UA) is a gut metabolite of dietary polymeric polyphenols ellagitannins. Oral gavage of biocompatible nanoparticle urolithin A attenuates the reduction of miRNA (miR-192–5p and miR-140–5p) by cisplatin and reduces renal oxidative stress. Liposomes carrying microRNA-500a-3P elicit inhibitory effect on necroptosis-related protein expression and inflammatory responses by cisplatin (S. Zhang, Sun, Kong, & Zhang, 2020). Several potential agents such as scutellarin, puerarin, curcumin and pentoxifylline also show renoprotective effect involving miRNAs regulation (El Magdoub, Schaalan, Rahmo, Farag, & Khedr, 2020; S. J. Huang et al., 2020; C. Y. Sun et al., 2019; Wu, Li, Li, Li, & Lu, 2020).

Ginkgo Biloba extract mediates renoprotection against methotrexate-induced renal injury by interrupting the PI3K/Akt/mTOR signaling and the expression of long non-coding RNA (lncRNA), metastasis-associated lung adenocarcinoma transcript-1 (MALAT1) (Sherif, Al-Shaalan, & Sabry, 2019). Overexpression of lncRNA XLOC_032768 is revealed to repress the gene expression of TNF-α thus attenuating cisplatin-induced apoptosis and inflammatory effects of renal tubular cells (X. Zhou et al., 2020). Interestingly, cisplatin-induced IL-1β upregulates lncRNA9884 expression via the NF-κB pathway; lncRNA9884 subsequently activates transcription of macrophage migration inhibitory factor (MIF) gene via binding to MIF promoter region, which in turn promotes IL-1β/NF-κB signaling and triggers a cytokine storm (Y. Zhang et al., 2020). Furthermore, lncRNA PRNCR1 overexpression is shown to reduce cisplatin-induced renal epithelial cell apoptosis through interference with the miR-182–5p/EZH1 axis (J. Li, Fan, Wang, Gong, & Guo, 2021).

Besides microRNAs and lncRNAs, circular RNAs (circRNAs) have recently attracted increasing attention as promising targets in both cancers and kidney diseases (van Zonneveld, Kölling, Bijkerk, & Lorenzen, 2021). One study uncovered that 368 circRNAs were expressed differentially in cisplatin treatment mice (C.-M. Li et al., 2019). Another study demonstrated that the elevated circRNA, circ-0114427, was found in the early stage of a cisplatin-AKI model, which was implicated in the binding of miR-494 and the increase of ATF3 expression; as a result, circ-0114427 led to a decrease in the production of inflammatory cytokines. Therefore, circ-0114427 could be a novel target for early treatment strategies for cisplatin-induced AKI (Cao et al., 2020). Overall, studies have demonstrated that the non-coding RNAs, including miRNA, lncRNAs, and circRNAs, are involved in both cancer therapy and kidney chemotoxicity, indicating that non-coding RNAs could be promising targets for developing novel drugs in the management of anticancer agent-induced nephrotoxicity, and intriguingly, bear potential dual action of renoprotection and anticancer.

5. Conclusions

There are currently no effective drugs targeting specific biochemical or molecular pathways associated with anticancer therapy-induced nephrotoxicity in clinical use or clinical trials, although several clinically used drugs, such as gemigliptin (DPP-4 inhibitor), pantoprazole (proton pump inhibitor) and Tempol, are under clinical trials for renal chemotoxicity. This chapter reviews and summarizes the kidney- and drug-associated risk factors of anticancer drug-induced nephrotoxicity that will allow clinicians and researchers to carefully assess the risk and benefits of treatments. This review also proposes prospective strategies to diagnose/mitigate nephrotoxicity arising from cancer therapeutics. In addition to the assessment of route of drug administration, timing of drug treatments, and pharmacodynamics, pharmacokinetics and pharmacogenetics among patients with cancer, future studies should also focus on identifying novel mechanisms and targets that may lead to the development of new approaches in the management of chemotherapy-induced nephrotoxicity.

Abbreviations

ABC transporters

ATP-binding cassette transporters

ACEi

angiotensin-converting enzyme inhibitors

AIN

acute interstitial nephritis

AKI

acute kidney injury

ARBs

angiotensin receptor blockers

CCB

calcium channel blocker

CECT

contrast-enhanced computed tomography

CIN

contrast-induced nephropathy

CKD

chronic kidney disease

CPIs

immune checkpoint inhibitors

CYP450

cytochrome P450

DPP-4

dipeptidyl peptidase-4

eGFR

estimated glomerular filtration rate

FSGF

focal segmental glomerulosclerosis

5-FU

5-fluorouracil

GCs

glucocorticoids

GGT

gamma-glutamyl transpeptidase

GSTs

glutathione S-transferases

HDACs

histone deacetylases

IL-2

interleukin-2

MCD

minimal change disease

mTOR

mammalian target of rapamycin

MTX

methotrexate

NAC

N-acetylcysteine or acetylcysteine

NSAIDs

nonsteroidal anti-inflammatory drugs

OAT

organic anion transporters

OCT

organic cation transporters

PDE5

phosphodiesterase type 5

PPI

proton pump inhibitors

SLC

renal solute carrier transporter

TLR

toll-like receptor

TMA

thrombotic microangiopathy

TNF-α

tumor necrosis factor-α

References

  1. Abd-Elrazek A, Shapana H, Shukry W, & Galilah D (2021). Comparison between Annona squamosa, Annona cherimolia and Annona atemoya ethanolic extracts extenuative impact against oxidative stress, inflammation and apoptosis in rat kidney induced by Ifosfamid. Toxicol Res (Camb), 10(4), 947–958. doi: 10.1093/toxres/tfab078 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Afjal MA, Goswami P, Ahmad S, Dabeer S, Akhter J, Salman M, … Raisuddin S (2020). 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, 1–9. doi: 10.1080/01480545.2020.1831011 [DOI] [PubMed] [Google Scholar]
  3. Afsar T, Razak S, Almajwal A, & Al-Disi D (2020). Doxorubicin-induced alterations in kidney functioning, oxidative stress, DNA damage, and renal tissue morphology; Improvement by Acacia hydaspica tannin-rich ethyl acetate fraction. Saudi J Biol Sci, 27(9), 2251–2260. doi: 10.1016/j.sjbs.2020.07.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Al-Harbi NO, Imam F, Al-Harbi MM, Al-Shabanah OA, Alotaibi MR, As Sobeai HM, … Al Rikabi AC (2019). Rutin inhibits carfilzomib-induced oxidative stress and inflammation via the NOS-mediated NF-κB signaling pathway. Inflammopharmacology, 27(4), 817–827. doi: 10.1007/s10787-018-0550-5 [DOI] [PubMed] [Google Scholar]
  5. Alikhan MA, Summers SA, Gan PY, Chan AJ, Khouri MB, Ooi JD, … Holdsworth SR (2016). Endogenous Toll-Like Receptor 9 Regulates AKI by Promoting Regulatory T Cell Recruitment. Journal of the American Society of Nephrology, 27(3), 706–714. doi: 10.1681/asn.2014090927 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Alshaker H, Thrower H, & Pchejetski D (2020). Sphingosine Kinase 1 in Breast Cancer—A New Molecular Marker and a Therapy Target. Frontiers in Oncology, 10(289). doi: 10.3389/fonc.2020.00289 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Andrade-Silva M, Cenedeze Marcos A., Perandini Luiz A., Felizardo R. José F., Watanabe I. Kazue M., Agudelo J. Sebastian H., … de Almeida Danilo C. (2018). TLR2 and TLR4 play opposite role in autophagy associated with cisplatin-induced acute kidney injury. Clinical Science, 132(16), 1725–1739. doi: 10.1042/cs20170262 [DOI] [PubMed] [Google Scholar]
  8. Andreev E, Koopman M, & Arisz L (1999). A rise in plasma creatinine that is not a sign of renal failure: which drugs can be responsible? J Intern Med, 246(3), 247–252. doi: 10.1046/j.1365-2796.1999.00515.x [DOI] [PubMed] [Google Scholar]
  9. Arab HH, Salama SA, & Maghrabi IA (2018). Camel Milk Ameliorates 5-Fluorouracil-Induced Renal Injury in Rats: Targeting MAPKs, NF-κB and PI3K/Akt/eNOS Pathways. Cellular Physiology and Biochemistry, 46(4), 1628–1642. doi: 10.1159/000489210 [DOI] [PubMed] [Google Scholar]
  10. Azouz AA, Abdel-Nassir Abdel-Razek E, & Abo-Youssef AM (2020). Amlodipine alleviates cisplatin-induced nephrotoxicity in rats through gamma-glutamyl transpeptidase (GGT) enzyme inhibition, associated with regulation of Nrf2/HO-1, MAPK/NF-κB, and Bax/Bcl-2 signaling. Saudi Pharm J, 28(11), 1317–1325. doi: 10.1016/j.jsps.2020.08.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Baharmi S, Kalantari H, Kalantar M, Goudarzi M, Mansouri E, & Kalantar H (2021). Pretreatment with Gallic Acid Mitigates Cyclophosphamide Induced Inflammation and Oxidative Stress in Mice. Curr Mol Pharmacol. doi: 10.2174/1874467214666210531162741 [DOI] [PubMed] [Google Scholar]
  12. Bai L, Li W, Zheng W, Xu D, Chen N, & Cui J (2020). Promising targets based on pattern recognition receptors for cancer immunotherapy. Pharmacological Research, 159, 105017. doi: 10.1016/j.phrs.2020.105017 [DOI] [PubMed] [Google Scholar]
  13. Basit A, Radi Z, Vaidya VS, Karasu M, & Prasad B (2019). Kidney cortical transporter expression across species using quantitative proteomics. Drug Metabolism and Disposition, dmd.119.086579. doi: 10.1124/dmd.119.086579 [DOI] [PubMed] [Google Scholar]
  14. Bauchet A-L, Masson R, Guffroy M, & Slaoui M (2011). Immunohistochemical Identification of Kidney Nephron Segments in the Dog, Rat, Mouse, and Cynomolgus Monkey. Toxicologic Pathology, 39(7), 1115–1128. doi: 10.1177/0192623311425060 [DOI] [PubMed] [Google Scholar]
  15. Belliere J, Meyer N, Mazieres J, Ollier S, Boulinguez S, Delas A, … Faguer S (2016). Acute interstitial nephritis related to immune checkpoint inhibitors. Br J Cancer, 115(12), 1457–1461. doi: 10.1038/bjc.2016.358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Bhat N, Kalthur SG, Padmashali S, & Monappa V (2018). Toxic Effects of Different Doses of Cyclophosphamide on Liver and Kidney Tissue in Swiss Albino Mice: A Histopathological Study. Ethiop J Health Sci, 28(6), 711–716. doi: 10.4314/ejhs.v28i6.5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bozina N, Bradamante V, & Lovrić M (2009). Genetic polymorphism of metabolic enzymes P450 (CYP) as a susceptibility factor for drug response, toxicity, and cancer risk. Arh Hig Rada Toksikol, 60(2), 217–242. doi: 10.2478/10004-1254-60-2009-1885 [DOI] [PubMed] [Google Scholar]
  18. Bringhen S, Milan A, D’Agostino M, Ferri C, Wäsch R, Gay F, … Engelhardt M (2019). Prevention, monitoring and treatment of cardiovascular adverse events in myeloma patients receiving carfilzomib A consensus paper by the European Myeloma Network and the Italian Society of Arterial Hypertension. Journal of Internal Medicine, 286(1), 63–74. doi: 10.1111/joim.12882 [DOI] [PubMed] [Google Scholar]
  19. Brosnan EM, Weickhardt AJ, Lu X, Maxon DA, Barón AE, Chonchol M, & Camidge DR (2014). Drug-induced reduction in estimated glomerular filtration rate in patients with ALK-positive non-small cell lung cancer treated with the ALK inhibitor crizotinib. Cancer, 120(5), 664–674. doi: 10.1002/cncr.28478 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Brown A, Kumar S, & Tchounwou PB (2019). Cisplatin-Based Chemotherapy of Human Cancers. J Cancer Sci Ther, 11(4). [PMC free article] [PubMed] [Google Scholar]
  21. Bruin MAC, Korse CM, van Wijnen B, de Jong VMT, Linn SC, van Triest B, … Huitema ADR (2021). A real or apparent decrease in glomerular filtration rate in patients using olaparib? European Journal of Clinical Pharmacology, 77(2), 179–188. doi: 10.1007/s00228-020-03070-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Buss CG, & Bhatia SN (2020). Nanoparticle delivery of immunostimulatory oligonucleotides enhances response to checkpoint inhibitor therapeutics. Proc Natl Acad Sci U S A, 117(24), 13428–13436. doi: 10.1073/pnas.2001569117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Caglar K, Kinalp C, Arpaci F, Turan M, Saglam K, Ozturk B, … Vural A (2002). Cumulative prior dose of cisplatin as a cause of the nephrotoxicity of high-dose chemotherapy followed by autologous stem-cell transplantation. Nephrology Dialysis Transplantation, 17(11), 1931–1935. doi: 10.1093/ndt/17.11.1931 [DOI] [PubMed] [Google Scholar]
  24. Calvo JA, Allocca M, Fake KR, Muthupalani S, Corrigan JJ, Bronson RT, & Samson LD (2016). Parp1 protects against Aag-dependent alkylation-induced nephrotoxicity in a sex-dependent manner. Oncotarget, 7(29). Retrieved from https://www.oncotarget.com/article/10440/text/ [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Camidge DR, Brosnan EM, DeSilva C, Koo PJ, & Chonchol M (2014). Crizotinib effects on creatinine and non-creatinine-based measures of glomerular filtration rate. J Thorac Oncol, 9(11), 1634–1637. doi: 10.1097/jto.0000000000000321 [DOI] [PubMed] [Google Scholar]
  26. Cao Y, Mi X, Zhang D, Wang Z, Zuo Y, & Tang W (2020). Transcriptome sequencing of circular RNA reveals a novel circular RNA-has_circ_0114427 in the regulation of inflammation in acute kidney injury. Clinical Science, 134(2), 139–154. doi: 10.1042/cs20190990 [DOI] [PubMed] [Google Scholar]
  27. Carota IA, Kenig-Kozlovsky Y, Onay T, Scott R, Thomson BR, Souma T, … Quaggin SE (2019). Targeting VE-PTP phosphatase protects the kidney from diabetic injury. Journal of Experimental Medicine, 216(4), 936–949. doi: 10.1084/jem.20180009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Carron PL, Padilla M, & Maurizi Balzan J (2014). Nephrotic syndrome and acute renal failure during pegylated liposomal doxorubicin treatment. Hemodial Int, 18(4), 846–847. doi: 10.1111/hdi.12196 [DOI] [PubMed] [Google Scholar]
  29. Cerrato A, Morra F, & Celetti A (2016). Use of poly ADP-ribose polymerase [PARP] inhibitors in cancer cells bearing DDR defects: the rationale for their inclusion in the clinic. J Exp Clin Cancer Res, 35(1), 179. doi: 10.1186/s13046-016-0456-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Chan CY, Tan KV, & Cornelissen B (2021). PARP Inhibitors in Cancer Diagnosis and Therapy. Clinical Cancer Research, 27(6), 1585–1594. doi: 10.1158/1078-0432.Ccr-20-2766 [DOI] [PubMed] [Google Scholar]
  31. Chandra A, Rao NS, Malhotra KP, Rastogi M, & Khurana R (2017). Everolimus-associated Acute Kidney Injury in Patients with Metastatic Breast Cancer. Indian J Nephrol, 27(5), 406–409. doi: 10.4103/ijn.IJN_304_16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Chávez-Iñiguez JS, Medina-Gonzalez R, Aguilar-Parra L, Torres-Vázquez EJ, Maggiani-Aguilera P, Cervantes-Pérez E, & García-García G (2018). Oral acyclovir induced hypokalemia and acute tubular necrosis a case report. BMC Nephrology, 19(1), 324. doi: 10.1186/s12882-018-1121-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Chen X, Yu C, Hou X, Li J, Li T, Qiu A, … Zhuang S (2020). Histone deacetylase 6 inhibition mitigates renal fibrosis by suppressing TGF-β and EGFR signaling pathways in obstructive nephropathy. American Journal of Physiology-Renal Physiology, 319(6), F1003–F1014. doi: 10.1152/ajprenal.00261.2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Cheungpasitporn W, Leung N, Rajkumar SV, Cornell LD, Sethi S, Angioi A, & Fervenza FC (2015). Bortezomib-induced acute interstitial nephritis. Nephrology Dialysis Transplantation, 30(7), 1225–1229. doi: 10.1093/ndt/gfv222 [DOI] [PubMed] [Google Scholar]
  35. Chiba T, Peasley KD, Cargill KR, Maringer KV, Bharathi SS, Mukherjee E, … Sims-Lucas S (2019). Sirtuin 5 Regulates Proximal Tubule Fatty Acid Oxidation to Protect against AKI. Journal of the American Society of Nephrology, 30(12), 2384–2398. doi: 10.1681/asn.2019020163 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Chiruvella V, Annamaraju P, & Guddati AK (2020). Management of nephrotoxicity of chemotherapy and targeted agents: 2020. Am J Cancer Res, 10(12), 4151–4164. [PMC free article] [PubMed] [Google Scholar]
  37. Choi SH, Leem J, & Lee IK (2017). Protective Effects of Gemigliptin, a Dipeptidyl Peptidase-4 Inhibitor, against Cisplatin-Induced Nephrotoxicity in Mice. Mediators Inflamm, 2017, 4139439. doi: 10.1155/2017/4139439 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Chuang YC, Tseng JC, Huang LR, Huang CM, Huang CF, & Chuang TH (2020). Adjuvant Effect of Toll-Like Receptor 9 Activation on Cancer Immunotherapy Using Checkpoint Blockade. Front Immunol, 11, 1075. doi: 10.3389/fimmu.2020.01075 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Cortazar FB, Kibbelaar ZA, Glezerman IG, Abudayyeh A, Mamlouk O, Motwani SS, … Leaf DE (2020). Clinical Features and Outcomes of Immune Checkpoint Inhibitor–Associated AKI: A Multicenter Study. Journal of the American Society of Nephrology, 31(2), 435–446. doi: 10.1681/asn.2019070676 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Dai R, Liu H, Han X, Liu J, Zhai Y, Rao J, … Xu H (2019). Angiopoietin-like-3 knockout protects against glomerulosclerosis in murine adriamycin-induced nephropathy by attenuating podocyte loss. BMC Nephrology, 20(1), 185. doi: 10.1186/s12882-019-1383-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Deshpande PP, Perazella MA, & Jhaveri KD (2021). PARP inhibitors and the Kidney. Journal of Onco-Nephrology, 5(1), 42–47. doi: 10.1177/2399369320987090 [DOI] [Google Scholar]
  42. Ding X, Chaiteerakij R, Moser CD, Shaleh H, Boakye J, Chen G, … Roberts LR (2016). Antitumor effect of the novel sphingosine kinase 2 inhibitor ABC294640 is enhanced by inhibition of autophagy and by sorafenib in human cholangiocarcinoma cells. Oncotarget, 7(15). Retrieved from https://www.oncotarget.com/article/7914/text/ [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Dobrek L, Nalik-Iwaniak K, Fic K, & Arent Z (2020). The Effect of Acetylcysteine on Renal Function in Experimental Models of Cyclophosphamide-and Ifosfamide-Induced Cystitis. Current Urology, 14(3), 150–162. doi: 10.1159/000499245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Drozdzik M, Drozdzik M, & Oswald S (2021). Membrane Carriers and Transporters in Kidney Physiology and Disease. Biomedicines, 9(4). doi: 10.3390/biomedicines9040426 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Du B, Dai X. m., Li S, Qi G. l., Cao G. x., Zhong Y, … Yang X. s. (2017). MiR-30c regulates cisplatin-induced apoptosis of renal tubular epithelial cells by targeting Bnip3L and Hspa5. Cell Death & Disease, 8(8), e2987–e2987. doi: 10.1038/cddis.2017.377 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Duan Z, Cai G, Li J, & Chen X (2020). Cisplatin-induced renal toxicity in elderly people. Ther Adv Med Oncol, 12, 1758835920923430. doi: 10.1177/1758835920923430 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. El-Far YM, & El-Mesery M (2021). Pevonedistat attenuates cisplatin-induced nephrotoxicity in mice by downregulating the release of inflammatory mediators. J Biochem Mol Toxicol, e22908. doi: 10.1002/jbt.22908 [DOI] [PubMed] [Google Scholar]
  48. El-Shabrawy M, Mishriki A, Attia H, Emad Aboulhoda B, Emam M, & Wanas H (2020). Protective effect of tolvaptan against cyclophosphamide-induced nephrotoxicity in rat models. Pharmacology Research & Perspectives, 8(5), e00659. doi: 10.1002/prp2.659 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. El Magdoub HM, Schaalan MF, Rahmo RM, Farag DB, & Khedr LH (2020). Implications of miRNAs on TGF-β/TAK1/mTOR pathway in mediating the renoprotective effects of pentoxifylline against cisplatin-induced nephrotoxicity in rats. Toxicol Appl Pharmacol, 404, 115184. doi: 10.1016/j.taap.2020.115184 [DOI] [PubMed] [Google Scholar]
  50. Elmansy RA, Seleem HS, Mahmoud AR, Hassanein EHM, & Ali FEM (2021). Rebamipide potentially mitigates methotrexate-induced nephrotoxicity via inhibition of oxidative stress and inflammation: A molecular and histochemical study. Anat Rec (Hoboken), 304(3), 647–661. doi: 10.1002/ar.24482 [DOI] [PubMed] [Google Scholar]
  51. Enomoto A, & Endou H (2005). Roles of organic anion transporters (OATs) and a urate transporter (URAT1) in the pathophysiology of human disease. Clin Exp Nephrol, 9(3), 195–205. doi: 10.1007/s10157-005-0368-5 [DOI] [PubMed] [Google Scholar]
  52. Ensergueix G, Pallet N, Joly D, Levi C, Chauvet S, Trivin C, & Augusto JF (2020). Ifosfamide nephrotoxicity in adult patients. 13(4), 660–665. doi: 10.1093/ckj/sfz183 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Esmaeeli A, Keshavarz Z, Dehdar F, Assadi M, & Seyedabadi M (2020). The effects of carvedilol, metoprolol and propranolol on cisplatin-induced kidney injury. Drug Chem Toxicol, 1–7. doi: 10.1080/01480545.2020.1846551 [DOI] [PubMed] [Google Scholar]
  54. Estrela GR, Wasinski F, Gregnani MF, Freitas-Lima LC, Arruda AC, Morais RL, … Araújo RC (2020). Angiotensin-Converting Enzyme Inhibitor Protects Against Cisplatin Nephrotoxicity by Modulating Kinin B1 Receptor Expression and Aminopeptidase P Activity in Mice. Frontiers in Molecular Biosciences, 7(96). doi: 10.3389/fmolb.2020.00096 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Evangelisti C, Evangelisti C, Buontempo F, Lonetti A, Orsini E, Chiarini F, … Martelli AM (2016). Therapeutic potential of targeting sphingosine kinases and sphingosine 1-phosphate in hematological malignancies. Leukemia, 30(11), 2142–2151. doi: 10.1038/leu.2016.208 [DOI] [PubMed] [Google Scholar]
  56. Ewees MGE, Abdel-Bakky MS, Bayoumi AMA, Abo-Saif AA, Altowayan WM, Alharbi KS, & Messiha BAS (2021). Dabigatran mitigates cisplatin-mediated nephrotoxicity through down regulation of thrombin pathway. J Adv Res, 31, 127–136. doi: 10.1016/j.jare.2020.12.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Fan HY, Wang XK, Li X, Ji K, Du SH, Liu Y, … Qi D (2020). Curcumin, as a pleiotropic agent, improves doxorubicin-induced nephrotic syndrome in rats. J Ethnopharmacol, 250, 112502. doi: 10.1016/j.jep.2019.112502 [DOI] [PubMed] [Google Scholar]
  58. Feidakis A, Panagiotou M-R, Tsoukakis E, Bacharaki D, Gounari P, Nikolopoulos P, … Vlahakos D (2021). Impact of Angiotensin-Converting Enzyme Inhibitors or Angiotensin Receptor Blockers on Acute Kidney Injury in Emergency Medical Admissions. Journal of Clinical Medicine, 10(3), 412. Retrieved from https://www.mdpi.com/2077-0383/10/3/412 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Feng Y, Mu R, Wang Z, Xing P, Zhang J, Dong L, & Wang C (2019). A toll-like receptor agonist mimicking microbial signal to generate tumor-suppressive macrophages. Nature Communications, 10(1), 2272. doi: 10.1038/s41467-019-10354-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Fiedler U, & Augustin HG (2006). Angiopoietins: a link between angiogenesis and inflammation. Trends Immunol, 27(12), 552–558. doi: 10.1016/j.it.2006.10.004 [DOI] [PubMed] [Google Scholar]
  61. Fotiou D, Roussou M, Gakiopoulou C, Psimenou E, & Gavriatopoulou M (2020). Carfilzomib-associated renal toxicity is common and unpredictable: a comprehensive analysis of 114 multiple myeloma patients. 10(11), 109. doi: 10.1038/s41408-020-00381-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Fox CP, Ali AS, McIlroy GW, Thust SC, Martinez-Calle N, Jackson AE, … Cwynarski K (2021). A phase I/II study of thiotepa-based immunochemotherapy in relapsed/refractory primary CNS lymphoma: the TIER trial. Blood Adv. doi: 10.1182/bloodadvances.2021004779 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Fujita K, N. T, Motoyama S, Miura M (2016). Influence of Renal ABC and SLC Transporter Polymorphisms on Cisplatin-induced Nephrotoxicity in Patients with Esophageal Cancer. Clin Pharmacol Biopharm, 5(158). doi:doi: 10.4172/2167-065X.1000158 [DOI] [Google Scholar]
  64. Gao H, Wang X, Qu X, Zhai J, Tao L, Zhang Y, … Zhang W (2020). Omeprazole attenuates cisplatin-induced kidney injury through suppression of the TLR4/NF-κB/NLRP3 signaling pathway. Toxicology, 440, 152487. doi: 10.1016/j.tox.2020.152487 [DOI] [PubMed] [Google Scholar]
  65. Garnier AS, Dellamaggiore J, Brilland B, Lagarce L, Abgueguen P, Furber A, … Augusto JF (2020). High Incidence of Amoxicillin-Induced Crystal Nephropathy in Patients Receiving High Dose of Intravenous Amoxicillin. J Clin Med, 9(7). doi: 10.3390/jcm9072022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Gastaud L, Ambrosetti D, Otto J, Marquette CH, Coutts M, Hofman P, … Favre G (2013). Acute kidney injury following crizotinib administration for non-small-cell lung carcinoma. Lung Cancer, 82(2), 362–364. doi: 10.1016/j.lungcan.2013.08.007 [DOI] [PubMed] [Google Scholar]
  67. Gelen V, Şengül E, Yıldırım S, Senturk E, Tekin S, & Kükürt A (2021). The protective effects of hesperidin and curcumin on 5-fluorouracil-induced nephrotoxicity in mice. Environ Sci Pollut Res Int, 28(34), 47046–47055. doi: 10.1007/s11356-021-13969-5 [DOI] [PubMed] [Google Scholar]
  68. Ghane Shahrbaf F, & Assadi F (2015). Drug-induced renal disorders. J Renal Inj Prev, 4(3), 57–60. doi: 10.12861/jrip.2015.12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Ghonaim E, El-Haggar S, & Gohar S (2021). Possible protective effect of pantoprazole against cisplatin-induced nephrotoxicity in head and neck cancer patients: a randomized controlled trial. Med Oncol, 38(9), 108. doi: 10.1007/s12032-021-01558-y [DOI] [PubMed] [Google Scholar]
  70. González-Martín A, Pothuri B, Vergote I, DePont Christensen R, Graybill W, Mirza MR, … Monk BJ (2019). Niraparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. New England Journal of Medicine, 381(25), 2391–2402. doi: 10.1056/NEJMoa1910962 [DOI] [PubMed] [Google Scholar]
  71. González E, Gutiérrez E, Galeano C, Chevia C, de Sequera P, Bernis C, … Praga M (2008). Early steroid treatment improves the recovery of renal function in patients with drug-induced acute interstitial nephritis. Kidney International, 73(8), 940–946. doi: 10.1038/sj.ki.5002776 [DOI] [PubMed] [Google Scholar]
  72. Groff JA, Kozak M, Boehmer JP, Demko TM, & Diamond JR (1997). Endotheliopathy: A continuum of hemolytic uremic syndrome due to mitomycin therapy. American Journal of Kidney Diseases, 29(2), 280–284. doi: 10.1016/S0272-6386(97)90042-1 [DOI] [PubMed] [Google Scholar]
  73. Guleria AS, Yang JC, Topalian SL, Weber JS, Parkinson DR, MacFarlane MP, … et al. (1994). Renal dysfunction associated with the administration of high-dose interleukin-2 in 199 consecutive patients with metastatic melanoma or renal carcinoma. J Clin Oncol, 12(12), 2714–2722. doi: 10.1200/jco.1994.12.12.2714 [DOI] [PubMed] [Google Scholar]
  74. Güntürk I, Yazici C, Köse SK, Dağli F, Yücel B, & Yay AH (2019). The effect of N-acetylcysteine on inflammation and oxidative stress in cisplatin-induced nephrotoxicity: a rat model. Turk J Med Sci, 49(6), 1789–1799. doi: 10.3906/sag-1903-225 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Guo Y, Ni J, Chen S, Bai M, Lin J, Ding G, … Zhang A (2018). MicroRNA-709 Mediates Acute Tubular Injury through Effects on Mitochondrial Function. J Am Soc Nephrol, 29(2), 449–461. doi: 10.1681/asn.2017040381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Gupta S, Short SAP, Sise ME, Prosek JM, Madhavan SM, Soler MJ, … Leaf DE (2021). Acute kidney injury in patients treated with immune checkpoint inhibitors. J Immunother Cancer, 9(10). doi: 10.1136/jitc-2021-003467 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Habib R (2021). Multifaceted roles of Toll-like receptors in acute kidney injury. Heliyon, 7(3), e06441. doi: 10.1016/j.heliyon.2021.e06441 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Han SJ, Williams RM, D’Agati V, Jaimes EA, Heller DA, & Lee HT (2020). Selective nanoparticle-mediated targeting of renal tubular Toll-like receptor 9 attenuates ischemic acute kidney injury. Kidney International, 98(1), 76–87. doi: 10.1016/j.kint.2020.01.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Hanna RM, Barsoum M, Arman F, Selamet U, Hasnain H, & Kurtz I (2019). Nephrotoxicity induced by intravitreal vascular endothelial growth factor inhibitors: emerging evidence. Kidney Int, 96(3), 572–580. doi: 10.1016/j.kint.2019.02.042 [DOI] [PubMed] [Google Scholar]
  80. Hannun YA, & Obeid LM (2018). Sphingolipids and their metabolism in physiology and disease. Nature Reviews Molecular Cell Biology, 19(3), 175–191. doi: 10.1038/nrm.2017.107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Hao Y, Guo F, Huang Z, Feng Y, Xia Z, Liu J, … Fu P (2020). 2-Methylquinazoline derivative 23BB as a highly selective histone deacetylase 6 inhibitor alleviated cisplatin-induced acute kidney injury. Biosci Rep, 40(1). doi: 10.1042/bsr20191538 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Harrison DJ, Kharbanda R, Cunningham DS, McLellan LI, & Hayes JD (1989). Distribution of glutathione S-transferase isoenzymes in human kidney: basis for possible markers of renal injury. J Clin Pathol, 42(6), 624–628. doi: 10.1136/jcp.42.6.624 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Hartmann JT, Kollmannsberger C, Kanz L, & Bokemeyer C (1999). Platinum organ toxicity and possible prevention in patients with testicular cancer. Int J Cancer, 83(6), 866–869. doi: [DOI] [PubMed] [Google Scholar]
  84. Holderfield M, Deuker MM, McCormick F, & McMahon M (2014). Targeting RAF kinases for cancer therapy: BRAF-mutated melanoma and beyond. Nat Rev Cancer, 14(7), 455–467. doi: 10.1038/nrc3760 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Holmboe L, Andersen AM, Mørkrid L, Slørdal L, & Hall KS (2012). High dose methotrexate chemotherapy: pharmacokinetics, folate and toxicity in osteosarcoma patients. Br J Clin Pharmacol, 73(1), 106–114. doi: 10.1111/j.1365-2125.2011.04054.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Hong SI, Ahn S, Lee YS, Kim WY, Lim KS, Lee JH, & Lee JL (2016). Contrast-induced nephropathy in patients with active cancer undergoing contrast-enhanced computed tomography. Support Care Cancer, 24(3), 1011–1017. doi: 10.1007/s00520-015-2875-6 [DOI] [PubMed] [Google Scholar]
  87. Hontecillas-Prieto L, Flores-Campos R, Silver A, de Álava E, Hajji N, & García-Domínguez DJ (2020). Synergistic Enhancement of Cancer Therapy Using HDAC Inhibitors: Opportunity for Clinical Trials. Frontiers in Genetics, 11(1113). doi: 10.3389/fgene.2020.578011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Hou X, Gong R, Zhan J, Zhou T, Ma Y, Zhao Y, … Zhang L (2018). p300 promotes proliferation, migration, and invasion via inducing epithelial-mesenchymal transition in non-small cell lung cancer cells. BMC Cancer, 18(1), 641. doi: 10.1186/s12885-018-4559-3 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  89. Howard SC, McCormick J, Pui CH, Buddington RK, & Harvey RD (2016). Preventing and Managing Toxicities of High-Dose Methotrexate. Oncologist, 21(12), 1471–1482. doi: 10.1634/theoncologist.2015-0164 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Hsiao YP, Chen HL, Tsai JN, Lin MY, Liao JW, Wei MS, … Ou CC (2021). Administration of Lactobacillus reuteri Combined with Clostridium butyricum Attenuates Cisplatin-Induced Renal Damage by Gut Microbiota Reconstitution, Increasing Butyric Acid Production, and Suppressing Renal Inflammation. Nutrients, 13(8). doi: 10.3390/nu13082792 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Hua H, Kong Q, Zhang H, Wang J, Luo T, & Jiang Y (2019). Targeting mTOR for cancer therapy. J Hematol Oncol, 12(1), 71. doi: 10.1186/s13045-019-0754-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Huang KM, Uddin ME, DiGiacomo D, Lustberg MB, Hu S, & Sparreboom A (2020). Role of SLC transporters in toxicity induced by anticancer drugs. Expert Opin Drug Metab Toxicol, 16(6), 493–506. doi: 10.1080/17425255.2020.1755253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Huang R, & Zhou P-K (2021). DNA damage repair: historical perspectives, mechanistic pathways and clinical translation for targeted cancer therapy. Signal Transduction and Targeted Therapy, 6(1), 254. doi: 10.1038/s41392-021-00648-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Huang S, You J, Wang K, Li Y, Zhang Y, Wei H, … Liu Y (2019). N-Acetylcysteine Attenuates Cisplatin-Induced Acute Kidney Injury by Inhibiting the C5a Receptor. Biomed Res Int, 2019, 4805853. doi: 10.1155/2019/4805853 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Huang SJ, Huang J, Yan YB, Qiu J, Tan RQ, Liu Y, … Gong Q (2020). The renoprotective effect of curcumin against cisplatin-induced acute kidney injury in mice: involvement of miR-181a/PTEN axis. Ren Fail, 42(1), 350–357. doi: 10.1080/0886022x.2020.1751658 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Huang Y, Shao Q, Luo X, Yang D, Zeng B, Xiang T, … Cheng Q (2018). Poly(ADP-ribose) polymerase-1 promotes recruitment of meiotic recombination-11 to chromatin and DNA double-strand break repair in Ku70-deficient breast cancer cells. Faseb j, fj201800092R. doi: 10.1096/fj.201800092R [DOI] [PubMed] [Google Scholar]
  97. Hucke A, & Ciarimboli G (2016). The Role of Transporters in the Toxicity of Chemotherapeutic Drugs: Focus on Transporters for Organic Cations. The Journal of Clinical Pharmacology, 56(S7), S157–S172. doi: 10.1002/jcph.706 [DOI] [PubMed] [Google Scholar]
  98. Hurabielle C, Pillebout E, Stehlé T, Pagès C, Roux J, Schneider P, … Flamant M (2016). Mechanisms Underpinning Increased Plasma Creatinine Levels in Patients Receiving Vemurafenib for Advanced Melanoma. PLoS One, 11(3), e0149873. doi: 10.1371/journal.pone.0149873 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Imig JD, Hye Khan MA, Burkhan A, Chen G, Adebesin AM, & Falck JR (2021). Kidney-Targeted Epoxyeicosatrienoic Acid Analog, EET-F01, Reduces Inflammation, Oxidative Stress, and Cisplatin-Induced Nephrotoxicity. Int J Mol Sci, 22(6). doi: 10.3390/ijms22062793 [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Inoue T (2017). M1 macrophage triggered by Mincle leads to a deterioration of acute kidney injury. Kidney International, 91(3), 526–529. doi: 10.1016/j.kint.2016.11.026 [DOI] [PubMed] [Google Scholar]
  101. Ismail RS, El-Awady MS, & Hassan MH (2020). Pantoprazole abrogated cisplatin-induced nephrotoxicity in mice via suppression of inflammation, apoptosis, and oxidative stress. Naunyn-Schmiedeberg’s Archives of Pharmacology, 393(7), 1161–1171. doi: 10.1007/s00210-020-01823-3 [DOI] [PubMed] [Google Scholar]
  102. Isono M, Okubo K, Asano T, & Sato A (2021). Inhibition of checkpoint kinase 1 potentiates anticancer activity of gemcitabine in bladder cancer cells. Scientific Reports, 11(1), 10181. doi: 10.1038/s41598-021-89684-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Iwakura T, Fukasawa H, Kitamura A, Ishibuchi K, Yasuda H, & Furuya R (2020). Effect of dipeptidyl peptidase-4 inhibitors on cisplatin-induced acute nephrotoxicity in cancer patients with diabetes mellitus: A retrospective study. PLOS ONE, 15(2), e0229377. doi: 10.1371/journal.pone.0229377 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Iwakura T, Zhao Z, Marschner JA, Devarapu SK, Yasuda H, & Anders HJ (2019). Dipeptidyl peptidase-4 inhibitor teneligliptin accelerates recovery from cisplatin-induced acute kidney injury by attenuating inflammation and promoting tubular regeneration. Nephrol Dial Transplant, 34(10), 1669–1680. doi: 10.1093/ndt/gfy397 [DOI] [PubMed] [Google Scholar]
  105. Iyer NG, Özdag H, & Caldas C (2004). p300/CBP and cancer. Oncogene, 23(24), 4225–4231. doi: 10.1038/sj.onc.1207118 [DOI] [PubMed] [Google Scholar]
  106. Izzedine H, Boostandoost H, & Mathian A (2017). Panitumumab-Induced Immune Complex Glomerulonephritis. Am J Kidney Dis, 69(2), 320–321. doi: 10.1053/j.ajkd.2016.09.011 [DOI] [PubMed] [Google Scholar]
  107. Izzedine H, Brocheriou I, Amoura Z, & Mathian A (2021). Acute Tubular Injury and Renal Arterial Myocyte Vacuolization Following Crizotinib Administration. Kidney International Reports, 6(2), 526–528. doi: 10.1016/j.ekir.2020.11.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Izzedine H, El-Fekih RK, & Perazella MA (2016). The renal effects of ALK inhibitors. Invest New Drugs, 34(5), 643–649. doi: 10.1007/s10637-016-0379-y [DOI] [PubMed] [Google Scholar]
  109. Izzedine H, & Perazella MA (2017). Anticancer Drug-Induced Acute Kidney Injury. Kidney Int Rep, 2(4), 504–514. doi: 10.1016/j.ekir.2017.02.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Jafaripour L, Naserzadeh R, Alizamani E, Javad Mashhadi SM, Moghadam ER, Nouryazdan N, & Ahmadvand H (2021). Effects of Rosmarinic Acid on Methotrexate-induced Nephrotoxicity and Hepatotoxicity in Wistar Rats. Indian J Nephrol, 31(3), 218–224. doi: 10.4103/ijn.IJN_14_20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Jang HR, Lee K, Jeon J, Kim J-R, Lee JE, Kwon GY, … Huh W (2020). Poly (ADP-Ribose) Polymerase Inhibitor Treatment as a Novel Therapy Attenuating Renal Ischemia-Reperfusion Injury. Frontiers in Immunology, 11(2574). doi: 10.3389/fimmu.2020.564288 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Jenke R, Reßing N, Hansen FK, Aigner A, & Büch T (2021). Anticancer Therapy with HDAC Inhibitors: Mechanism-Based Combination Strategies and Future Perspectives. Cancers (Basel), 13(4). doi: 10.3390/cancers13040634 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Jhaveri KD, Wanchoo R, Sakhiya V, Ross DW, & Fishbane S (2017). Adverse Renal Effects of Novel Molecular Oncologic Targeted Therapies: A Narrative Review. Kidney International Reports, 2(1), 108–123. doi: 10.1016/j.ekir.2016.09.055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Jiang X, Ren Z, Zhao B, Zhou S, Ying X, & Tang Y (2020). Ameliorating Effect of Pentadecapeptide Derived from Cyclina sinensis on Cyclophosphamide-Induced Nephrotoxicity. Mar Drugs, 18(9). doi: 10.3390/md18090462 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Jung YJ, Park W, Kang KP, & Kim W (2020). SIRT2 is involved in cisplatin-induced acute kidney injury through regulation of mitogen-activated protein kinase phosphatase-1. Nephrology Dialysis Transplantation, 35(7), 1145–1156. doi: 10.1093/ndt/gfaa042 [DOI] [PubMed] [Google Scholar]
  116. Kapoor R, Robinson KA, Cata JP, Owusu-Agyemang P, Soliz JM, Hernandez M, … Badgwell B (2019). Assessment of nephrotoxicity associated with combined cisplatin and mitomycin C usage in laparoscopic hyperthermic intraperitoneal chemotherapy. International Journal of Hyperthermia, 36(1), 492–497. doi: 10.1080/02656736.2019.1597175 [DOI] [PubMed] [Google Scholar]
  117. Khan FI, Lai D, Anwer R, Azim I, & Khan MKA (2020). Identifying novel sphingosine kinase 1 inhibitors as therapeutics against breast cancer. Journal of Enzyme Inhibition and Medicinal Chemistry, 35(1), 172–186. doi: 10.1080/14756366.2019.1692828 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Khan MI, Waguespack SG, & Ahmed I (2019). Recent advances in the management of hyponatremia in cancer patients. Journal of Cancer Metastasis and Treatment, 5, 71. doi: 10.20517/2394-4722.2019.017 [DOI] [Google Scholar]
  119. Khan TH, Ganaie MA, Alharthy KM, Madkhali H, Jan BL, & Sheikh IA (2020). Naringenin prevents doxorubicin-induced toxicity in kidney tissues by regulating the oxidative and inflammatory insult in Wistar rats. Arch Physiol Biochem, 126(4), 300–307. doi: 10.1080/13813455.2018.1529799 [DOI] [PubMed] [Google Scholar]
  120. Kim D, Park W, Lee S, Kim W, Park SK, & Kang KP (2018). Absence of Sirt3 aggravates cisplatin nephrotoxicity via enhanced renal tubular apoptosis and inflammation. Mol Med Rep, 18(4), 3665–3672. doi: 10.3892/mmr.2018.9350 [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Kim J-Y, Jo J, Leem J, & Park K-K (2020). Inhibition of p300 by Garcinol Protects against Cisplatin-Induced Acute Kidney Injury through Suppression of Oxidative Stress, Inflammation, and Tubular Cell Death in Mice. Antioxidants, 9(12), 1271. Retrieved from https://www.mdpi.com/2076-3921/9/12/1271 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Kim JY, Jo J, Kim K, An HJ, Gwon MG, Gu H, … Park KK (2019). Pharmacological Activation of Sirt1 Ameliorates Cisplatin-Induced Acute Kidney Injury by Suppressing Apoptosis, Oxidative Stress, and Inflammation in Mice. Antioxidants (Basel), 8(8). doi: 10.3390/antiox8080322 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Kim S, Jo CH, & Kim G-H (2021). The Role of Vasopressin V2 Receptor in Drug-Induced Hyponatremia. Frontiers in Physiology, 12(2272). doi: 10.3389/fphys.2021.797039 [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Kitchlu A, McArthur E, Amir E, Booth CM, Sutradhar R, Majeed H, … Wald R (2018). Acute Kidney Injury in Patients Receiving Systemic Treatment for Cancer: A Population-Based Cohort Study. JNCI: Journal of the National Cancer Institute, 111(7), 727–736. doi: 10.1093/jnci/djy167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Kitchlu A, McArthur E, Amir E, Booth CM, Sutradhar R, Majeed H, … Wald R (2019). Acute Kidney Injury in Patients Receiving Systemic Treatment for Cancer: A Population-Based Cohort Study. J Natl Cancer Inst, 111(7), 727–736. doi: 10.1093/jnci/djy167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Kitchlu A, & Rosner MH (2019). Hyponatremia in patients with cancer. Current Opinion in Nephrology and Hypertension, 28(5), 433–440. doi: 10.1097/mnh.0000000000000525 [DOI] [PubMed] [Google Scholar]
  127. Klaus R, Niyazi M, & Lange-Sperandio B (2021). Radiation-induced kidney toxicity: molecular and cellular pathogenesis. Radiation Oncology, 16(1), 43. doi: 10.1186/s13014-021-01764-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Knights KM, Rowland A, & Miners JO (2013). Renal drug metabolism in humans: the potential for drug–endobiotic interactions involving cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT). British Journal of Clinical Pharmacology, 76(4), 587–602. doi: 10.1111/bcp.12086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Kobayashi R, Suzuki A, Matsuura K, Yamada N, Nakano M, Deguchi T, … Itoh Y (2016). Risk analysis for cisplatin-induced nephrotoxicity during first cycle of chemotherapy.
  130. Koks MS, Ocak G, Suelmann BBM, Hulsbergen-Veelken CAR, Haitjema S, Vianen ME, … Khairoun M (2021). Immune checkpoint inhibitor-associated acute kidney injury and mortality: An observational study. PLOS ONE, 16(6), e0252978. doi: 10.1371/journal.pone.0252978 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Koźmiński P, Halik PK, Chesori R, & Gniazdowska E (2020). Overview of Dual-Acting Drug Methotrexate in Different Neurological Diseases, Autoimmune Pathologies and Cancers. Int J Mol Sci, 21(10). doi: 10.3390/ijms21103483 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Kwon Y (2021). Possible Beneficial Effects of N-Acetylcysteine for Treatment of Triple-Negative Breast Cancer. Antioxidants (Basel), 10(2). doi: 10.3390/antiox10020169 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. LaFargue CJ, Dal Molin GZ, Sood AK, & Coleman RL (2019). Exploring and comparing adverse events between PARP inhibitors. The Lancet Oncology, 20(1), e15–e28. doi: 10.1016/S1470-2045(18)30786-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Lameire NH, Flombaum CD, Moreau D, & Ronco C (2005). Acute renal failure in cancer patients. Ann Med, 37(1), 13–25. doi: 10.1080/07853890510007205 [DOI] [PubMed] [Google Scholar]
  135. Lankhorst S, Baelde HJ, Kappers MHW, Smedts FMM, Hansen A, Groningen M. C. C. v., … Meiracker A. H. v. d. (2015). Greater Sensitivity of Blood Pressure Than Renal Toxicity to Tyrosine Kinase Receptor Inhibition With Sunitinib. Hypertension, 66(3), 543–549. doi:doi: 10.1161/HYPERTENSIONAHA.115.05435 [DOI] [PubMed] [Google Scholar]
  136. Lankhorst S, Kappers MH, van Esch JH, Smedts FM, Sleijfer S, Mathijssen RH, … van den Meiracker AH (2014). Treatment of hypertension and renal injury induced by the angiogenesis inhibitor sunitinib: preclinical study. Hypertension, 64(6), 1282–1289. doi: 10.1161/hypertensionaha.114.04187 [DOI] [PubMed] [Google Scholar]
  137. Laplace N, Kepenekian V, Friggeri A, Vassal O, Ranchon F, Rioufol C, … Bakrin N (2020). Sodium thiosulfate protects from renal impairement following hyperthermic intraperitoneal chemotherapy (HIPEC) with Cisplatin. International Journal of Hyperthermia, 37(1), 897–902. doi: 10.1080/02656736.2020.1795277 [DOI] [PubMed] [Google Scholar]
  138. Launay-Vacher V, Oudard S, Janus N, Gligorov J, Pourrat X, Rixe O, … Deray G (2007). Prevalence of Renal Insufficiency in cancer patients and implications for anticancer drug management: the renal insufficiency and anticancer medications (IRMA) study. Cancer, 110(6), 1376–1384. doi: 10.1002/cncr.22904 [DOI] [PubMed] [Google Scholar]
  139. Lazareth H, Delanoy N, Cohen R, Boissier E, Ayari H, Combe P, … Pallet N (2020). Nephrotoxicity Associated With Niraparib. American Journal of Kidney Diseases, 76(6), 898–900. doi: 10.1053/j.ajkd.2020.05.021 [DOI] [PubMed] [Google Scholar]
  140. Lexicomp. (2022). Fluorouracil (systemic): Drug information. Retrieved from https://www-uptodate-com.proxy.library.vcu.edu/contents/fluorouracil-systemic-drug-information?search=Fluorouracil&source=panel_search_result&selectedTitle=1~147&usage_type=panel&display_rank=1&showDrugLabel=true. https://www-uptodate-com.proxy.library.vcu.edu/contents/fluorouracil-systemic-drug-information?search=Fluorouracil&source=panel_search_result&selectedTitle=1~147&usage_type=panel&display_rank=1&showDrugLabel=true
  141. Li C-M, Li M, Ye Z-C, Huang J-Y, Li Y, Yao Z-Y, … Lou T-Q (2019). Circular RNA expression profiles in cisplatin-induced acute kidney injury in mice. Epigenomics, 11(10), 1191–1207. doi: 10.2217/epi-2018-0167 [DOI] [PubMed] [Google Scholar]
  142. Li C, Xue VW, Wang Q-M, Lian G-Y, Huang X-R, Lee T-L, … Lan H-Y (2020). The Mincle/Syk/NF-κB Signaling Circuit Is Essential for Maintaining the Protumoral Activities of Tumor-Associated Macrophages. Cancer Immunology Research, 8(8), 1004–1017. doi: 10.1158/2326-6066.Cir-19-0782 [DOI] [PubMed] [Google Scholar]
  143. Li G, Sachdev U, Peters K, Liang X, & Lotze MT (2019). The VE-PTP Inhibitor AKB-9778 Improves Antitumor Activity and Diminishes the Toxicity of Interleukin 2 (IL-2) Administration. Journal of Immunotherapy, 42(7), 237–243. doi: 10.1097/cji.0000000000000290 [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Li J, Fan X, Wang Q, Gong Y, & Guo L (2021). Long Noncoding RNA PRNCR1 Reduces Renal Epithelial Cell Apoptosis in Cisplatin-Induced AKI by Regulating miR-182–5p/EZH1. Kidney and Blood Pressure Research, 46(2), 162–172. doi: 10.1159/000510157 [DOI] [PubMed] [Google Scholar]
  145. Li Y, Gao M, Yin LH, Xu LN, Qi Y, Sun P, & Peng JY (2021). Dioscin ameliorates methotrexate-induced liver and kidney damages via adjusting miRNA-145–5p-mediated oxidative stress. Free Radic Biol Med, 169, 99–109. doi: 10.1016/j.freeradbiomed.2021.03.035 [DOI] [PubMed] [Google Scholar]
  146. Li Z, Xu K, Zhang N, Amador G, Wang Y, Zhao S, … Wang Z (2018). Overexpressed SIRT6 attenuates cisplatin-induced acute kidney injury by inhibiting ERK1/2 signaling. Kidney Int, 93(4), 881–892. doi: 10.1016/j.kint.2017.10.021 [DOI] [PubMed] [Google Scholar]
  147. Lin JS, Mamlouk O, Selamet U, Tchakarov A, Glass WF, Sheth RA, … Abudayyeh A (2021). Infliximab for the treatment of patients with checkpoint inhibitor associated acute tubular interstitial nephritis. OncoImmunology, 10(1), 1877415. doi: 10.1080/2162402X.2021.1877415 [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Lin YT, Wang YF, Yang JC, Yu CJ, Wu SG, Shih JY, & Yang PC (2014). Development of renal cysts after crizotinib treatment in advanced ALK-positive non-small-cell lung cancer. J Thorac Oncol, 9(11), 1720–1725. doi: 10.1097/jto.0000000000000326 [DOI] [PubMed] [Google Scholar]
  149. Liu J, Livingston MJ, Dong G, Tang C, Su Y, Wu G, … Dong Z (2018). Histone deacetylase inhibitors protect against cisplatin-induced acute kidney injury by activating autophagy in proximal tubular cells. Cell Death Dis, 9(3), 322. doi: 10.1038/s41419-018-0374-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Liu X-Y, Zhang F-R, Shang J-Y, Liu Y-Y, Lv X-F, Yuan J-N, … Liang S-J (2018). Renal inhibition of miR-181a ameliorates 5-fluorouracil-induced mesangial cell apoptosis and nephrotoxicity. Cell Death & Disease, 9(6), 610. doi: 10.1038/s41419-018-0677-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Liu Y, Yang EJ, Shi C, Mou PK, Zhang B, Wu C, … Shim JS (2020). Histone Acetyltransferase (HAT) P300/CBP Inhibitors Induce Synthetic Lethality in PTEN-Deficient Colorectal Cancer Cells through Destabilizing AKT. International journal of biological sciences, 16(11), 1774–1784. doi: 10.7150/ijbs.42197 [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Loren P, Saavedra N, Saavedra K, Zambrano T, Moriel P, & Salazar LA (2021). Epigenetic Mechanisms Involved in Cisplatin-Induced Nephrotoxicity: An Update. Pharmaceuticals (Basel), 14(6). doi: 10.3390/ph14060491 [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Luan Z, Wei Y, Huo X, Sun X, Zhang C, Ming W, … Zhang X (2021). Pregnane X receptor (PXR) protects against cisplatin-induced acute kidney injury in mice. Biochim Biophys Acta Mol Basis Dis, 1867(3), 165996. doi: 10.1016/j.bbadis.2020.165996 [DOI] [PubMed] [Google Scholar]
  154. Lyu B, Wang W, Ji XY, Ritter JK, & Li N (2020). Detrimental role of sphingosine kinase 1 in kidney damage in DOCA-salt hypertensive model: evidence from knockout mice. BMC Nephrol, 21(1), 173. doi: 10.1186/s12882-020-01815-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Manohar S, Kompotiatis P, Thongprayoon C, Cheungpasitporn W, Herrmann J, & Herrmann SM (2019). Programmed cell death protein 1 inhibitor treatment is associated with acute kidney injury and hypocalcemia: meta-analysis. Nephrol Dial Transplant, 34(1), 108–117. doi: 10.1093/ndt/gfy105 [DOI] [PubMed] [Google Scholar]
  156. Manohar S, & Leung N (2018). Cisplatin nephrotoxicity: a review of the literature. Journal of Nephrology, 31(1), 15–25. doi: 10.1007/s40620-017-0392-z [DOI] [PubMed] [Google Scholar]
  157. Marabondo S, & Kaufman HL (2017). High-dose interleukin-2 (IL-2) for the treatment of melanoma: safety considerations and future directions. Expert Opin Drug Saf, 16(12), 1347–1357. doi: 10.1080/14740338.2017.1382472 [DOI] [PubMed] [Google Scholar]
  158. Markowitz GS, & Perazella MA (2005). Drug-induced renal failure: a focus on tubulointerstitial disease. Clin Chim Acta, 351(1–2), 31–47. doi: 10.1016/j.cccn.2004.09.005 [DOI] [PubMed] [Google Scholar]
  159. Martín Martorell P, Huerta Alvaro M, Solís Salguero MA, & Insa Molla A (2014). Crizotinib and renal insufficiency: A case report and review of the literature. Lung Cancer, 84(3), 310–313. doi: 10.1016/j.lungcan.2014.03.001 [DOI] [PubMed] [Google Scholar]
  160. Martínez-Valenzuela L, Draibe J, Fulladosa X, Gomà M, Gómez F, Antón P, … Torras J (2021). Acute Tubulointerstitial Nephritis in Clinical Oncology: A Comprehensive Review. International Journal of Molecular Sciences, 22(5), 2326. Retrieved from https://www.mdpi.com/1422-0067/22/5/2326 [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. McKibbin T, Cheng LL, Kim S, Steuer CE, Owonikoko TK, Khuri FR, … Saba NF (2016). Mannitol to prevent cisplatin-induced nephrotoxicity in patients with squamous cell cancer of the head and neck (SCCHN) receiving concurrent therapy. Support Care Cancer, 24(4), 1789–1793. doi: 10.1007/s00520-015-2978-0 [DOI] [PubMed] [Google Scholar]
  162. McSweeney KR, Gadanec LK, Qaradakhi T, Ali BA, Zulli A, & Apostolopoulos V (2021). Mechanisms of Cisplatin-Induced Acute Kidney Injury: Pathological Mechanisms, Pharmacological Interventions, and Genetic Mitigations. Cancers (Basel), 13(7). doi: 10.3390/cancers13071572 [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Meirson T, Asher N, Bomze D, & Markel G (2020). Safety of BRAF+MEK Inhibitor Combinations: Severe Adverse Event Evaluation. Cancers, 12(6), 1650. Retrieved from https://www.mdpi.com/2072-6694/12/6/1650 [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Mercantepe T, Topcu A, Rakici S, Tumkaya L, Yilmaz A, & Mercantepe F (2019). The radioprotective effect of N-acetylcysteine against x-radiation-induced renal injury in rats. Environ Sci Pollut Res Int, 26(28), 29085–29094. doi: 10.1007/s11356-019-06110-0 [DOI] [PubMed] [Google Scholar]
  165. Mérida E, & Praga M (2019). NSAIDs and Nephrotic Syndrome. Clinical Journal of the American Society of Nephrology, 14(9), 1280–1282. doi: 10.2215/cjn.08090719 [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Mirabito Colafella KM, Neves KB, Montezano AC, Garrelds IM, van Veghel R, de Vries R, … Versmissen J (2020). Selective ETA vs. dual ETA/B receptor blockade for the prevention of sunitinib-induced hypertension and albuminuria in WKY rats. Cardiovasc Res, 116(10), 1779–1790. doi: 10.1093/cvr/cvz260 [DOI] [PubMed] [Google Scholar]
  167. Misra SC, Santagostino A, Dine G, & Bonhomme Faivre L (2019). Acute Kidney Injury Following High-Dose Methotrexate Administration in a Day Care Hospital. Drug Safety - Case Reports, 6(1), 12. doi: 10.1007/s40800-019-0106-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Miteva-Marcheva NN, Ivanov HY, Dimitrov DK, & Stoyanova VK (2020). Application of pharmacogenetics in oncology. Biomarker Research, 8(1), 32. doi: 10.1186/s40364-020-00213-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Mlejnek P, Dolezel P, Kriegova E, & Pastvova N (2021). N-acetylcysteine Can Induce Massive Oxidative Stress, Resulting in Cell Death with Apoptotic Features in Human Leukemia Cells. Int J Mol Sci, 22(23). doi: 10.3390/ijms222312635 [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Moledina DG, & Perazella MA (2016). Proton Pump Inhibitors and CKD. Journal of the American Society of Nephrology, 27(10), 2926–2928. doi: 10.1681/asn.2016020192 [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Nagata M, Ohji G, & Iwata K (2019). Acute tubulointerstitial nephritis caused by rifampicin: An increasing and often overlooked side effect in elderly patients Int J Clin Pharmacol Ther, 57(5), 264–269. doi: 10.5414/cp203287 [DOI] [PubMed] [Google Scholar]
  172. Naughton CA (2008). Drug-induced nephrotoxicity. Am Fam Physician, 78(6), 743–750. [PubMed] [Google Scholar]
  173. Navarro-Hortal MD, Varela-López A, Romero-Márquez JM, Rivas-García L, Speranza L, Battino M, & Quiles JL (2020). Role of flavonoids against adriamycin toxicity. Food Chem Toxicol, 146, 111820. doi: 10.1016/j.fct.2020.111820 [DOI] [PubMed] [Google Scholar]
  174. Oleas D, Bolufer M, Agraz I, Felip E, Muñoz E, Gabaldón A, … Soler MJ (2020). Acute interstitial nephritis associated with immune checkpoint inhibitors: a single-centre experience. Clinical Kidney Journal, 14(5), 1364–1370. doi: 10.1093/ckj/sfaa008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Ommati MM, Farshad O, Ghanbarinejad V, Mohammadi HR, Khadijeh M, Negar A, … Heidari R (2020). The Nephroprotective Role of Carnosine Against Ifosfamide-Induced Renal Injury and Electrolytes Imbalance is Mediated Via the Regulation of Mitochondrial Function and Alleviation of Oxidative Stress. Drug Res (Stuttg), 70(1), 49–56. doi: 10.1055/a-1017-5085 [DOI] [PubMed] [Google Scholar]
  176. Ozkok S, & Ozkok A (2017). Contrast-induced acute kidney injury: A review of practical points. World J Nephrol, 6(3), 86–99. doi: 10.5527/wjn.v6.i3.86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Paluri RK, Sonpavde G, Morgan C, Rojymon J, Mar AH, & Gangaraju R (2019). Renal toxicity with mammalian target of rapamycin inhibitors: A meta-analysis of randomized clinical trials. Oncol Rev, 13(2), 455. doi: 10.4081/oncol.2019.455 [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Pant K, Peixoto E, Richard S, & Gradilone SA (2020). Role of Histone Deacetylases in Carcinogenesis: Potential Role in Cholangiocarcinoma. Cells, 9(3). doi: 10.3390/cells9030780 [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Patel JB, S. A (2021, [Updated 2021 Jun 5]). Nephrotoxic Medications. Retrieved from https://www.ncbi.nlm.nih.gov/books/NBK553144/ [PubMed]
  180. Patel V, Elias R, Formella J, Schwartzman W, Christie A, Cai Q, … Brugarolas J (2020). Acute interstitial nephritis, a potential predictor of response to immune checkpoint inhibitors in renal cell carcinoma. J Immunother Cancer, 8(2). doi: 10.1136/jitc-2020-001198 [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Pavkovic M, Robinson-Cohen C, Chua AS, Nicoara O, Cárdenas-González M, Bijol V, … Vaidya VS (2016). Detection of Drug-Induced Acute Kidney Injury in Humans Using Urinary KIM-1, miR-21, −200c, and −423. Toxicol Sci, 152(1), 205–213. doi: 10.1093/toxsci/kfw077 [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Peasley K, Chiba T, Goetzman E, & Sims-Lucas S (2021). Sirtuins play critical and diverse roles in acute kidney injury. Pediatric Nephrology. doi: 10.1007/s00467-020-04866-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Perazella MA (2005). Drug-induced nephropathy: an update. Expert Opin Drug Saf, 4(4), 689–706. doi: 10.1517/14740338.4.4.689 [DOI] [PubMed] [Google Scholar]
  184. Perazella MA (2009). Renal Vulnerability to Drug Toxicity. Clinical Journal of the American Society of Nephrology, 4(7), 1275–1283. doi: 10.2215/cjn.02050309 [DOI] [PubMed] [Google Scholar]
  185. Perazella MA (2010). Toxic Nephropathies: Core Curriculum 2010. American Journal of Kidney Diseases, 55(2), 399–409. doi: 10.1053/j.ajkd.2009.10.046 [DOI] [PubMed] [Google Scholar]
  186. Perazella MA (2012). Onco-Nephrology: Renal Toxicities of Chemotherapeutic Agents. Clinical Journal of the American Society of Nephrology, 7(10), 1713–1721. doi: 10.2215/cjn.02780312 [DOI] [PubMed] [Google Scholar]
  187. Perazella MA (2018). Pharmacology behind Common Drug Nephrotoxicities. Clinical Journal of the American Society of Nephrology, 13(12), 1897–1908. doi: 10.2215/cjn.00150118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Perazella MA, & Markowitz GS (2010). Drug-induced acute interstitial nephritis. Nature Reviews Nephrology, 6(8), 461–470. doi: 10.1038/nrneph.2010.71 [DOI] [PubMed] [Google Scholar]
  189. Perazella MA, & Moeckel GW (2010). Nephrotoxicity From Chemotherapeutic Agents: Clinical Manifestations, Pathobiology, and Prevention/Therapy. Seminars in Nephrology, 30(6), 570–581. doi: 10.1016/j.semnephrol.2010.09.005 [DOI] [PubMed] [Google Scholar]
  190. Perazella MA, & Shirali AC (2018). Nephrotoxicity of Cancer Immunotherapies: Past, Present and Future. J Am Soc Nephrol, 29(8), 2039–2052. doi: 10.1681/asn.2018050488 [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Pitman M, Oehler MK, & Pitson SM (2021). Sphingolipids as multifaceted mediators in ovarian cancer. Cellular Signalling, 81, 109949. doi: 10.1016/j.cellsig.2021.109949 [DOI] [PubMed] [Google Scholar]
  192. Porta C, Bamias A, Danesh FR, Dębska-Ślizień A, Gallieni M, Gertz MA, … Zakharova E (2020). KDIGO Controversies Conference on onco-nephrology: understanding kidney impairment and solid-organ malignancies, and managing kidney cancer. Kidney International, 98(5), 1108-1119. doi: 10.1016/j.kint.2020.06.046 [DOI] [PubMed] [Google Scholar]
  193. Pyne NJ, & Pyne S (2020). Recent advances in the role of sphingosine 1-phosphate in cancer. FEBS Letters, 594(22), 3583–3601. doi: 10.1002/1873-3468.13933 [DOI] [PubMed] [Google Scholar]
  194. Qin S, Li A, Yi M, Yu S, Zhang M, & Wu K (2019). Recent advances on anti-angiogenesis receptor tyrosine kinase inhibitors in cancer therapy. Journal of Hematology & Oncology, 12(1), 27. doi: 10.1186/s13045-019-0718-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Qu J, Ding Y, Jiang K, Hao J, Li Y, Zhang A, … Li Z (2021). Nephrotoxicity of Immune Checkpoint Inhibitors: A Disproportionality Analysis from 2013 to 2020. The Tohoku Journal of Experimental Medicine, 254(4), 275–282. doi: 10.1620/tjem.254.275 [DOI] [PubMed] [Google Scholar]
  196. Raal FJ, Rosenson RS, Reeskamp LF, Hovingh GK, Kastelein JJP, Rubba P, … Gaudet D (2020). Evinacumab for Homozygous Familial Hypercholesterolemia. N Engl J Med, 383(8), 711–720. doi: 10.1056/NEJMoa2004215 [DOI] [PubMed] [Google Scholar]
  197. Ramsey LB, Balis FM, O’Brien MM, Schmiegelow K, Pauley JL, Bleyer A, … Heldrup J (2018). Consensus Guideline for Use of Glucarpidase in Patients with High-Dose Methotrexate Induced Acute Kidney Injury and Delayed Methotrexate Clearance. Oncologist, 23(1), 52–61. doi: 10.1634/theoncologist.2017-0243 [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Reddy V, W. N (2021, 2020 Nov 17). Retrieved from https://www.ncbi.nlm.nih.gov/books/NBK556021/
  199. Richter SK, & Crannage AJ (2015). Evaluation of N-acetylcysteine for the prevention of contrast-induced nephropathy. J Community Hosp Intern Med Perspect, 5(3), 27297. doi: 10.3402/jchimp.v5.27297 [DOI] [PMC free article] [PubMed] [Google Scholar]
  200. Robert C (2020). A decade of immune-checkpoint inhibitors in cancer therapy. Nature Communications, 11(1), 3801. doi: 10.1038/s41467-020-17670-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Roskoski R Jr. (2019). Properties of FDA-approved small molecule protein kinase inhibitors. Pharmacol Res, 144, 19–50. doi: 10.1016/j.phrs.2019.03.006 [DOI] [PubMed] [Google Scholar]
  202. Sabapathy V, Cheru NT, Corey R, Mohammad S, & Sharma R (2019). A Novel Hybrid Cytokine IL233 Mediates regeneration following Doxorubicin-Induced Nephrotoxic Injury. Scientific Reports, 9(1), 3215. doi: 10.1038/s41598-019-39886-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Sahindokuyucu-Kocasari F, Akyol Y, Ozmen O, Erdemli-Kose SB, & Garli S (2021). Apigenin alleviates methotrexate-induced liver and kidney injury in mice. Hum Exp Toxicol, 9603271211009964. doi: 10.1177/09603271211009964 [DOI] [PubMed] [Google Scholar]
  204. Salama RM, Nasr MM, Abdelhakeem JI, Roshdy OK, & ElGamal MA (2020). Alogliptin attenuates cyclophosphamide-induced nephrotoxicity: a novel therapeutic approach through modulating MAP3K/JNK/SMAD3 signaling cascade. Drug and Chemical Toxicology, 1–10. doi: 10.1080/01480545.2020.1814319 [DOI] [PubMed] [Google Scholar]
  205. Saleh D, Abdelbaset M, Hassan A, Sharaf O, Mahmoud S, & Hegazy R (2020). Omega-3 fatty acids ameliorate doxorubicin-induced cardiorenal toxicity: In-vivo regulation of oxidative stress, apoptosis and renal Nox4, and in-vitro preservation of the cytotoxic efficacy. PLOS ONE, 15(11), e0242175. doi: 10.1371/journal.pone.0242175 [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Sazonov V, Tobylbayeva Z, Saparov A, Jubaniyazov B, Issakov S, & Gaipov A (2021). New Therapeutic Approach to Reduce Methotrexate Toxicity after High-Dose Chemotherapy in a Child with Acute Lymphocytic Leukemia: Efficacy and Safety of Hemoadsorption with HA-230 Adsorber. Blood Purif, 1–5. doi: 10.1159/000514135 [DOI] [PubMed] [Google Scholar]
  207. Schiffl H (2020). Gender differences in the susceptibility of hospital-acquired acute kidney injury: more questions than answers. International Urology and Nephrology, 52(10), 1911–1914. doi: 10.1007/s11255-020-02526-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Sharp CN, & Siskind LJ (2017). Developing better mouse models to study cisplatin-induced kidney injury. American Journal of Physiology-Renal Physiology, 313(4), F835–F841. doi: 10.1152/ajprenal.00285.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Sherif IO, Al-Shaalan NH, & Sabry D (2019). Ginkgo Biloba Extract Alleviates Methotrexate-Induced Renal Injury: New Impact on PI3K/Akt/mTOR Signaling and MALAT1 Expression. Biomolecules, 9(11), 691. Retrieved from https://www.mdpi.com/2218-273X/9/11/691 [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Shye M, Hanna RM, Patel SS, Tram-Tran N, Hou J, McCannel C, … Kurtz I (2020). Worsening proteinuria and renal function after intravitreal vascular endothelial growth factor blockade for diabetic proliferative retinopathy. Clin Kidney J, 13(6), 969–980. doi: 10.1093/ckj/sfaa049 [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Smith HL, Southgate H, Tweddle DA, & Curtin NJ (2020). DNA damage checkpoint kinases in cancer. Expert Rev Mol Med, 22, e2. doi: 10.1017/erm.2020.3 [DOI] [PubMed] [Google Scholar]
  212. Song Y, Liu L, Liu B, Liu R, Chen Y, Li C, … Liu Y (2021). Interaction of nobiletin with methotrexate ameliorates 7-OH methotrexate-induced nephrotoxicity through endoplasmic reticulum stress-dependent PERK/CHOP signaling pathway. Pharmacol Res, 165, 105371. doi: 10.1016/j.phrs.2020.105371 [DOI] [PubMed] [Google Scholar]
  213. Stone EL, Carter KP, Wagner EK, Asensio MA, Benzie E, Chiang YY, … Johnson DS (2021). Lack of blocking activity in anti-CTLA-4 antibodies reduces toxicity, but not anti-tumor efficacy. bioRxiv, 2021.2007.2012.452090. doi: 10.1101/2021.07.12.452090 [DOI] [Google Scholar]
  214. Stremska ME, Jose S, Sabapathy V, Huang L, Bajwa A, Kinsey GR, … Sharma R (2017). IL233, A Novel IL-2 and IL-33 Hybrid Cytokine, Ameliorates Renal Injury. Journal of the American Society of Nephrology, 28(9), 2681–2693. doi: 10.1681/asn.2016121272 [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Sun CY, Nie J, Zheng ZL, Zhao J, Wu LM, Zhu Y, … Feng B (2019). Renoprotective effect of scutellarin on cisplatin-induced renal injury in mice: Impact on inflammation, apoptosis, and autophagy. Biomed Pharmacother, 112, 108647. doi: 10.1016/j.biopha.2019.108647 [DOI] [PubMed] [Google Scholar]
  216. Sun YB, Qu X, Zhang X, Caruana G, Bertram JF, & Li J (2013). Glomerular endothelial cell injury and damage precedes that of podocytes in adriamycin-induced nephropathy. PLoS One, 8(1), e55027. doi: 10.1371/journal.pone.0055027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Tan RZ, Liu J, Zhang YY, Wang HL, Li JC, Liu YH, … Wang L (2019). Curcumin relieved cisplatin-induced kidney inflammation through inhibiting Mincle-maintained M1 macrophage phenotype. Phytomedicine, 52, 284–294. doi: 10.1016/j.phymed.2018.09.210 [DOI] [PubMed] [Google Scholar]
  218. Tang J, Shi Y, Liu N, Xu L, Zang X, Li P, … Zhuang S (2018). Blockade of histone deacetylase 6 protects against cisplatin-induced acute kidney injury. Clin Sci (Lond), 132(3), 339–359. doi: 10.1042/cs20171417 [DOI] [PubMed] [Google Scholar]
  219. Tchounwou PB, Dasari S, Noubissi FK, Ray P, & Kumar S (2021). Advances in Our Understanding of the Molecular Mechanisms of Action of Cisplatin in Cancer Therapy. J Exp Pharmacol, 13, 303–328. doi: 10.2147/jep.S267383 [DOI] [PMC free article] [PubMed] [Google Scholar]
  220. Teuma C, Pelletier S, Amini-Adl M, Perier-Muzet M, Maucort-Boulch D, Thomas L, … Dalle S (2017). Adjunction of a MEK inhibitor to Vemurafenib in the treatment of metastatic melanoma results in a 60% reduction of acute kidney injury. Cancer Chemother Pharmacol, 79(5), 1043–1049. doi: 10.1007/s00280-017-3300-2 [DOI] [PubMed] [Google Scholar]
  221. Townsend DM, & Tew KD (2003). The role of glutathione-S-transferase in anti-cancer drug resistance. Oncogene, 22(47), 7369–7375. doi: 10.1038/sj.onc.1206940 [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Trendowski MR, El-Charif O, Ratain MJ, Monahan P, Mu Z, Wheeler HE, … Dolan ME (2019). Clinical and Genome-Wide Analysis of Serum Platinum Levels after Cisplatin-Based Chemotherapy. Clinical Cancer Research, 25(19), 5913–5924. doi: 10.1158/1078-0432.Ccr-19-0113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Un H, Ugan RA, Kose D, Bayir Y, Cadirci E, Selli J, & Halici Z (2020). A novel effect of Aprepitant: Protection for cisplatin-induced nephrotoxicity and hepatotoxicity. Eur J Pharmacol, 880, 173168. doi: 10.1016/j.ejphar.2020.173168 [DOI] [PubMed] [Google Scholar]
  224. van Zonneveld AJ, Kölling M, Bijkerk R, & Lorenzen JM (2021). Circular RNAs in kidney disease and cancer. Nature Reviews Nephrology. doi: 10.1038/s41581-021-00465-9 [DOI] [PubMed] [Google Scholar]
  225. Volarevic V, Djokovic B, Jankovic MG, Harrell CR, Fellabaum C, Djonov V, & Arsenijevic N (2019). Molecular mechanisms of cisplatin-induced nephrotoxicity: a balance on the knife edge between renoprotection and tumor toxicity. J Biomed Sci, 26(1), 25. doi: 10.1186/s12929-019-0518-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Wanchoo R, Abudayyeh A, Doshi M, Edeani A, Glezerman IG, Monga D, … Jhaveri KD (2017). Renal Toxicities of Novel Agents Used for Treatment of Multiple Myeloma. Clin J Am Soc Nephrol, 12(1), 176–189. doi: 10.2215/cjn.06100616 [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Wanchoo R, Jhaveri KD, Deray G, & Launay-Vacher V (2016). Renal effects of BRAF inhibitors: a systematic review by the Cancer and the Kidney International Network. Clin Kidney J, 9(2), 245–251. doi: 10.1093/ckj/sfv149 [DOI] [PMC free article] [PubMed] [Google Scholar]
  228. Wanchoo R, Karam S, Uppal NN, Barta VS, Deray G, Devoe C, … Jhaveri KD (2017). Adverse Renal Effects of Immune Checkpoint Inhibitors: A Narrative Review. Am J Nephrol, 45(2), 160–169. doi: 10.1159/000455014 [DOI] [PubMed] [Google Scholar]
  229. Wanchoo R, Khan S, Kolitz JE, & Jhaveri KD (2015). Carfilzomib-related acute kidney injury may be prevented by N-acetyl-L-cysteine. J Oncol Pharm Pract, 21(4), 313–316. doi: 10.1177/1078155214531804 [DOI] [PubMed] [Google Scholar]
  230. Wang P, Wang Z, & Liu J (2020). Role of HDACs in normal and malignant hematopoiesis. Molecular Cancer, 19(1), 5. doi: 10.1186/s12943-019-1127-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  231. Wang YM, Gu ML, Meng FS, Jiao WR, Zhou XX, Yao HP, & Ji F (2017). Histone acetyltransferase p300/CBP inhibitor C646 blocks the survival and invasion pathways of gastric cancer cell lines. Int J Oncol, 51(6), 1860–1868. doi: 10.3892/ijo.2017.4176 [DOI] [PubMed] [Google Scholar]
  232. Webb TR, Slavish J, George RE, Look AT, Xue L, Jiang Q, … Morris SW (2009). Anaplastic lymphoma kinase: role in cancer pathogenesis and small-molecule inhibitor development for therapy. Expert Rev Anticancer Ther, 9(3), 331–356. doi: 10.1586/14737140.9.3.331 [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Weber J (2010). Immune checkpoint proteins: a new therapeutic paradigm for cancer--preclinical background: CTLA-4 and PD-1 blockade. Semin Oncol, 37(5), 430–439. doi: 10.1053/j.seminoncol.2010.09.005 [DOI] [PubMed] [Google Scholar]
  234. Wei X, Wu Y, Tang H, Wang B, Wang Y, Sun W, … Wang C (2021). CP-25 ameliorates methotrexate induced nephrotoxicity via improving renal apoptosis and methotrexate excretion. J Pharmacol Sci, 146(1), 21–28. doi: 10.1016/j.jphs.2021.02.007 [DOI] [PubMed] [Google Scholar]
  235. Wilson N, Wiele AJ, Surasi DS, Rao P, Sircar K, Shah AY, … Msaouel P (2021). Efficacy of gemcitabine plus doxorubicin (Gem + Dox) in patients with renal medullary carcinoma (RMC). Journal of Clinical Oncology, 39(6_suppl), 324–324. doi: 10.1200/JCO.2021.39.6_suppl.324 [DOI] [Google Scholar]
  236. Wong KK (2020). DNMT1 as a therapeutic target in pancreatic cancer: mechanisms and clinical implications. Cellular Oncology, 43(5), 779–792. doi: 10.1007/s13402-020-00526-4 [DOI] [PubMed] [Google Scholar]
  237. Workeneh BT, Uppal NN, Jhaveri KD, & Rondon-Berrios H (2021). Hypomagnesemia in the Cancer Patient. Kidney360, 2(1), 154–166. doi: 10.34067/kid.0005622020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  238. Wu Z, Li C, Li Q, Li J, & Lu X (2020). Puerarin alleviates cisplatin-induced acute renal damage and upregulates microRNA-31-related signaling. Exp Ther Med, 20(4), 3122–3129. doi: 10.3892/etm.2020.9081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Xie DL, Hu G, Wang W, Li P-L, Chen C, & Li N (2020). Inhibition of Sphingosine Kinase 2 Attenuated Cisplatin-induced Nephrotoxicity in Mice. The FASEB Journal, 34(S1), 1–1. doi: 10.1096/fasebj.2020.34.s1.06411 [DOI] [Google Scholar]
  240. Xu Y-Y, Jiang N, Liu T-S, Qu H-Q, & Wang T (2012). Evaluation of the effect of glutathione on cisplatin antitumor activity and kidney injury at different administration times. Mol Med Rep, 6(5), 1075–1080. doi: 10.3892/mmr.2012.1033 [DOI] [PubMed] [Google Scholar]
  241. Xu Y, Ma H, Shao J, Wu J, Zhou L, Zhang Z, … Han J (2015). A Role for Tubular Necroptosis in Cisplatin-Induced AKI. J Am Soc Nephrol, 26(11), 2647–2658. doi: 10.1681/asn.2014080741 [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Xue D, Hsu E, Fu Y-X, & Peng H (2021). Next-generation cytokines for cancer immunotherapy. Antibody Therapeutics, 4(2), 123–133. doi: 10.1093/abt/tbab014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Yap TA, Tan DSP, Terbuch A, Caldwell R, Guo C, Goh BC, … de Bono JS (2021). First-in-Human Trial of the Oral Ataxia Telangiectasia and RAD3-Related (ATR) Inhibitor BAY 1895344 in Patients with Advanced Solid Tumors. Cancer Discovery, 11(1), 80–91. doi: 10.1158/2159-8290.Cd-20-0868 [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Yarlagadda SG, & Perazella MA (2008). Drug-induced crystal nephropathy: an update. Expert Opinion on Drug Safety, 7(2), 147–158. doi: 10.1517/14740338.7.2.147 [DOI] [PubMed] [Google Scholar]
  245. Yasuma T, Kobayashi T, D’Alessandro-Gabazza CN, Fujimoto H, Ito K, Nishii Y, … Hataji O (2018). Renal Injury during Long-Term Crizotinib Therapy. Int J Mol Sci, 19(10). doi: 10.3390/ijms19102902 [DOI] [PMC free article] [PubMed] [Google Scholar]
  246. Yee SW, Brackman DJ, Ennis EA, Sugiyama Y, Kamdem LK, Blanchard R, … Giacomini KM (2018). Influence of Transporter Polymorphisms on Drug Disposition and Response: A Perspective From the International Transporter Consortium. Clinical Pharmacology & Therapeutics, 104(5), 803–817. doi: 10.1002/cpt.1098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Yemm KE, Alwan LM, Malik AB, & Salazar LG (2019). Renal toxicity with liposomal doxorubicin in metastatic breast cancer. J Oncol Pharm Pract, 25(7), 1738–1742. doi: 10.1177/1078155218798157 [DOI] [PubMed] [Google Scholar]
  248. Yeung E, Yong E, & Wong F (2004). Renal dysfunction in cirrhosis: diagnosis, treatment and prevention. MedGenMed, 6(4), 9. [PMC free article] [PubMed] [Google Scholar]
  249. Yokota R, Bhunu B, Toba H, & Intapad S (2021). Sphingolipids and Kidney Disease: Possible Role of Preeclampsia and Intrauterine Growth Restriction (IUGR). Kidney360, 2(3), 534–541. doi: 10.34067/kid.0006322020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Yoon SP, & Kim J (2016). Poly(ADP-ribose) polymerase 1 contributes to oxidative stress through downregulation of sirtuin 3 during cisplatin nephrotoxicity. Anat Cell Biol, 49(3), 165–176. doi: 10.5115/acb.2016.49.3.165 [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Yoshida T, Niho S, Toda M, Goto K, Yoh K, Umemura S, … Ohe Y (2014). Protective effect of magnesium preloading on cisplatin-induced nephrotoxicity: a retrospective study. Jpn J Clin Oncol, 44(4), 346–354. doi: 10.1093/jjco/hyu004 [DOI] [PubMed] [Google Scholar]
  252. You J, Gao F, Tang H, Peng F, Jia L, Huang K, … Chen J (2019). A medicinal and edible formula YH0618 ameliorates the toxicity induced by Doxorubicin via regulating the expression of Bax/Bcl-2 and FOXO4. Journal of Cancer, 10, 3665–3677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  253. Young A, Beriault D, Jung B, Nikonova A, Abosh D, Lee S, … Perl J (2019). DAMPAned Methotrexate: A Case Report and Review of the Management of Acute Methotrexate Toxicity. Can J Kidney Health Dis, 6, 2054358119895078. doi: 10.1177/2054358119895078 [DOI] [PMC free article] [PubMed] [Google Scholar]
  254. Yuan HT, Khankin EV, Karumanchi SA, & Parikh SM (2009). Angiopoietin 2 is a partial agonist/antagonist of Tie2 signaling in the endothelium. Mol Cell Biol, 29(8), 2011–2022. doi: 10.1128/mcb.01472-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
  255. Zazuli Z, Duin N, Jansen K, Vijverberg SJH, Maitland-van der Zee AH, & Masereeuw R (2020). The Impact of Genetic Polymorphisms in Organic Cation Transporters on Renal Drug Disposition. Int J Mol Sci, 21(18). doi: 10.3390/ijms21186627 [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Zhang DY, Tu T, Younis MR, Zhu KS, Liu H, Lei S, … Huang P (2021). Clinically translatable gold nanozymes with broad spectrum antioxidant and anti-inflammatory activity for alleviating acute kidney injury. Theranostics, 11(20), 9904–9917. doi: 10.7150/thno.66518 [DOI] [PMC free article] [PubMed] [Google Scholar]
  257. Zhang J, Ye Z. w., Tew KD, & Townsend DM (2021). Chapter Eight - Cisplatin chemotherapy and renal function. In Tew KD & Fisher PB (Eds.), Advances in Cancer Research (Vol. 152, pp. 305–327): Academic Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  258. Zhang J, Zou Y, Cheng-Jing Y, Xiang-Heng L, Wang XP, Yu XJ, … Wang J (2020). Pioglitazone alleviates cisplatin nephrotoxicity by suppressing mitochondria-mediated apoptosis via SIRT1/p53 signalling. J Cell Mol Med, 24(20), 11718–11728. doi: 10.1111/jcmm.15782 [DOI] [PMC free article] [PubMed] [Google Scholar]
  259. Zhang MY, Dugbartey GJ, Juriasingani S, & Sener A (2021). Hydrogen Sulfide Metabolite, Sodium Thiosulfate: Clinical Applications and Underlying Molecular Mechanisms. Int J Mol Sci, 22(12). doi: 10.3390/ijms22126452 [DOI] [PMC free article] [PubMed] [Google Scholar]
  260. Zhang S, Sun H, Kong W, & Zhang B (2020). Functional role of microRNA-500a-3P-loaded liposomes in the treatment of cisplatin-induced AKI. IET Nanobiotechnology, 14(6), 465–469. doi: 10.1049/iet-nbt.2019.0247 [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Zhang X, Ritter JK, & Li N (2018). Sphingosine-1-phosphate pathway in renal fibrosis. American Journal of Physiology-Renal Physiology, 315(4), F752–F756. doi: 10.1152/ajprenal.00596.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Zhang X, Wang W, Ji XY, Ritter JK, & Li N (2019). Knockout of Sphingosine Kinase 1 Attenuates Renal Fibrosis in Unilateral Ureteral Obstruction Model. Am J Nephrol, 50(3), 196–203. doi: 10.1159/000502448 [DOI] [PMC free article] [PubMed] [Google Scholar]
  263. Zhang XY, & Elfarra AA (2018). Toxicity mechanism-based prodrugs: glutathione-dependent bioactivation as a strategy for anticancer prodrug design. Expert Opin Drug Discov, 13(9), 815–824. doi: 10.1080/17460441.2018.1508207 [DOI] [PMC free article] [PubMed] [Google Scholar]
  264. Zhang Y. j., Zhang L, Feng F, & Cao Q. f. (2021). ANGPTL3 Overexpression Suppresses the Development of Oncogenic Properties in Renal Cell Carcinoma via the Wnt/β-Catenin Signaling Pathway and Predicts Good Prognosis. Disease Markers, 2021, 2863856. doi: 10.1155/2021/2863856 [DOI] [PMC free article] [PubMed] [Google Scholar]
  265. Zhang Y, Tang PM-K, Niu Y, García Córdoba CA, Huang X-R, Yu C, & Lan H-Y (2020). Long Non-coding RNA LRNA9884 Promotes Acute Kidney Injury via Regulating NF-kB-Mediated Transcriptional Activation of MIF. Frontiers in Physiology, 11(1399). doi: 10.3389/fphys.2020.590027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  266. Zhang Y, Tao X, Yin L, Xu L, Xu Y, Qi Y, … Peng J (2017). Protective effects of dioscin against cisplatin-induced nephrotoxicity via the microRNA-34a/sirtuin 1 signalling pathway. Br J Pharmacol, 174(15), 2512–2527. doi: 10.1111/bph.13862 [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Zhao T, Liang X, Chen J, Bao Y, Wang A, Gan X, … Wang L (2019). ANGPTL3 inhibits renal cell carcinoma metastasis by inhibiting VASP phosphorylation. Biochemical and Biophysical Research Communications, 516(3), 880–887. doi: 10.1016/j.bbrc.2019.06.120 [DOI] [PubMed] [Google Scholar]
  268. Zheng H, Zhang Y, Li L, Zhang R, Luo Z, Yang Z, … Sun Q (2021). Depletion of Toll-Like Receptor-9 Attenuates Renal Tubulointerstitial Fibrosis After Ischemia-Reperfusion Injury. Frontiers in Cell and Developmental Biology, 9(236). doi: 10.3389/fcell.2021.641527 [DOI] [PMC free article] [PubMed] [Google Scholar]
  269. Zhou S, Zeng S, & Shu Y (2021). Drug-Drug Interactions at Organic Cation Transporter 1. Front Pharmacol, 12, 628705. doi: 10.3389/fphar.2021.628705 [DOI] [PMC free article] [PubMed] [Google Scholar]
  270. Zhou X, Jiang K, Luo H, Wu C, Yu W, & Cheng F (2020). Novel lncRNA XLOC_032768 alleviates cisplatin-induced apoptosis and inflammatory response of renal tubular epithelial cells through TNF-α. Int Immunopharmacol, 83, 106472. doi: 10.1016/j.intimp.2020.106472 [DOI] [PubMed] [Google Scholar]
  271. Zibetti Dal Molin G, Westin SN, Msaouel P, Gomes LM, Dickens A, & Coleman RL (2020). Discrepancy in calculated and measured glomerular filtration rates in patients treated with PARP inhibitors. International Journal of Gynecologic Cancer, 30(1), 89–93. doi: 10.1136/ijgc-2019-000714 [DOI] [PubMed] [Google Scholar]
  272. Zsiros E, Lynam S, Attwood KM, Wang C, Chilakapati S, Gomez EC, … Odunsi K (2021). Efficacy and Safety of Pembrolizumab in Combination With Bevacizumab and Oral Metronomic Cyclophosphamide in the Treatment of Recurrent Ovarian Cancer: A Phase 2 Nonrandomized Clinical Trial. JAMA Oncology, 7(1), 78–85. doi: 10.1001/jamaoncol.2020.5945 [DOI] [PMC free article] [PubMed] [Google Scholar]
  273. Zunino F, Pratesi G, Micheloni A, Cavalletti E, Sala F, & Tofanetti O (1989). Protective effect of reduced glutathione against cisplatin-induced renal and systemic toxicity and its influence on the therapeutic activity of the antitumor drug. Chemico-Biological Interactions, 70(1), 89–101. doi: 10.1016/0009-2797(89)90065-3 [DOI] [PubMed] [Google Scholar]

RESOURCES