You are on page 1of 11

J Nephrol

DOI 10.1007/s40620-017-0392-z

REVIEW

Cisplatin nephrotoxicity: a review of the literature


Sandhya Manohar1 · Nelson Leung1,2   

Received: 31 January 2017 / Accepted: 11 March 2017


© Italian Society of Nephrology 2017

Abstract  Cisplatin is a platinum containing drug first Introduction


approved as an antineoplastic agent in 1978. It remains an
important and effective therapy in many forms of cancer The discovery of cisplatin is truly a journey of serendipity.
today. Cisplatin mediates its tumorcidal effects via a num- In 1965, Rosenberg and his colleagues noticed that when
ber of different cytotoxic mechanisms. Although it is best the bacteria Escherichia coli was incubated in an ammo-
known for DNA damage, cisplatin also causes cytoplas- nium chloride growth medium inside a chamber with plati-
mic organelle dysfunction particularly with the endoplas- num electrodes, they exhibited an unusual growth limiting
mic reticulum and mitochondria. It also activates apoptotic and filamentation behavior [1]. The chemical compound
pathways and inflicts cellular damage via oxidative stress was isolated and studied for its potential tumor growth
and inflammation. One of its dose limiting toxicities is its suppression properties. Cis-diamminedichloridoplatinum
effects on the kidney. This includes acute kidney injury [2] or cisplatin was identified and eventually approved for
as well as tubular injury resulting in  electrolyte wasting. use in ovarian and testicular cancer by US Food and Drug
Extensive research has found that cisplatin entry into a cell Administration in 1978. Interestingly, cisplatin was already
is facilitated by a number of cellular transporters including known to the world as Peyronie’s chloride which was first
human copper transport protein 1 (Ctr1) and the organic synthesized in 1845 by Michele Peyrone [1].
cation transporter 2 (OCT2) which are expressed on renal Despite tremendous advances in the field of oncology,
tubular cells. The interactions between the mechanisms of cisplatin continues to be a much sought after chemothera-
cytotoxicity and cellular transport play an important role in peutic agent. Currently, it is used in the treatment of the fol-
the nephrotoxicity. Better understanding of these interac- lowing cancers: testicular, ovarian, bladder, head and neck,
tions could one day help devise better renoprotection that esophageal, small and non-small cell lung, breast, cervical,
would not reduce its anti-tumor effects. stomach, prostate cancers, Hodgkin’s and non-Hodgkin’s
lymphomas, neuroblastoma, sarcomas, multiple myeloma,
Keywords  Cisplatin · Nephrotoxicity · Cytotoxicity · melanoma and mesothelioma. Cisplatin has been on the
Chemotherapy · Renal protection market for 38 years, and yet we are still struggling with its
major side effect of nephrotoxicity. Many nephro-protective
measures have been studied. In this review, we discuss the
current knowledge available regarding cisplatin, its effects
on the kidney and consider the possibilities for future direc-
tions of research.
* Nelson Leung
Leung.nelson@mayo.edu
1
Division of Nephrology and Hypertension, Department Mechanism of action of cisplatin
of Internal Medicine, Mayo Clinic, Rochester, MN, USA
2
Division of Hematology, Department of Internal Medicine, Metals are essential for many of the biological processes
Mayo Clinic, Rochester, MN, USA in the human cell, and cisplatin is a metallic-platinum

13
Vol.:(0123456789)
J Nephrol

containing drug. It is believed to affect many signal trans- cytoprotective effect of Amifostine is not consistently dem-
duction pathways that eventually cause an induction of onstrated, with multiple reports showing evidence of tox-
apoptosis [3]. Many mechanisms have been found to play icity despite administration at optimal doses with ototoxic-
a role. The best-known mechanism is its effect on DNA ity being more commonly reported [13, 14]. Procainamide
synthesis and repairs; which results in cell cycle arrest. is also another drug that is believed to work similarly to
Research has also shown that cisplatin, like most metals, reduce the formation of the nephrotoxin without compro-
is involved in generation of reactive oxygen species (ROS), mising the anti-tumor effect. In fact, it is thought to com-
affects the tumor-suppressor protein p53, induces apopto- plex with the platinum bound to DNA and prevents disso-
sis via the death receptor-tumor necrosis factor interaction ciation and enhances the anti-tumor effects. Esposito et al.
and intrinsic caspases, causes mitochondrial dysfunction as noted in their mice study that when they used a combina-
well as affects the calcium signaling in the cell via stress tion of an otherwise lethal dose of cisplatin with procaina-
on the endoplasmic reticulum (ER) [3]. These are but some mide, the leukemic mice had a 100% survival at day 14 as
of the mechanisms by which it exerts its anti-neoplastic compared to zero in those receiving cisplatin alone. They
effects. Just as its anti-neoplastic effects, cisplatin has dif- also noted a decrease in the tubular degenerative changes in
ferent nephrotoxic effects on different parts of the nephron. those treated with procainamide [15, 16].
It is known to cause tubular damage, inflammatory damage Once cisplatin enters the tubular cell, it has been noted
in the interstitium as well as vascular injury [4]. to exert many effects which culminate in apoptosis/necrosis
of the cell. The commonly known target is the nuclear DNA
damage but only about 1 percent of the cytosolic cisplatin
Intracellular events is noted in the nucleus [17]. In the last four decades, mul-
tiple cellular targets have been identified. For the ease of
After cisplatin enters the tubular cell, it appears to undergo understanding, these have been consolidated under 5 sec-
a complex series of reactions that results in its activation to tions: (1) DNA damage (2) Cytoplasmic organelle dysfunc-
a more potent toxin. It is unclear from the current data if all tion: with endoplasmic reticulum stress and mitochondrial
the cytotoxic effects are mediated via the activated cisplatin dysfunction (3) Apoptotic pathways both caspase-depend-
metabolites. Townsend et  al. had initially looked into this ent and death receptor-mediated (4) Oxidative Stress: with
with in vivo studies by blocking the enzymatic pathways of formation of reactive oxygen species and (5) Inflammation:
g-glutamyl transpeptidase (GGT) and cysteine-S-conjugate mediated via tumor necrosis factor and other chemokines.
b-lyase and found reduced nephrotoxicity from cisplatin.
Further studies showed that the two enzymes along with
glutathione-cisplatin conjugate resulted in the formation of DNA damage
a reactive thiol [5, 6] which is believed to be a more potent
nephrotoxin. It is well known that cisplatin mediates its antitumor effects
To mitigate the effects of the cisplatin’s toxic metabo- from DNA damage and a similar mechanism appears to
lites, many potential targets have been evaluated. High be at play in the kidney cells. Once the cisplatin enters the
doses of glutathione are believed to inhibit GGT competi- cell, its complex interactions with the cellular environment
tively, and some studies have shown renoprotective action converting it into a positively charged electrophile that has
against cisplatin, though the doses required to achieve this a high affinity to DNA [18]. This results in the formation of
were large [7, 8]. Interestingly even though the conjuga- DNA cross links that prevents DNA synthesis and replica-
tion of cisplatin with glutathione results in a more potent tion causing the cell to be in cell-cycle arrest mode. Inter-
nephrotoxin; this conjugation may actually decrease cispl- estingly, the nuclear DNA may not be the only site at which
atin’s anti-tumor effects [9]. High intracellular glutathione cisplatin exerts its cytotoxic effect. Dissociation enhanced
has been noted to be associated with cisplatin-resistant lanthanide fluoroimmunoassay studies have revealed a four
tumors, and a similar effect was observed in the tumor cells to six-fold higher proportion of cisplatin adducts to mito-
with a high expression of GGT [10, 11]. chondrial DNA as compared to nuclear DNA [19]. This is
Thiol agents are used in the prevention of cisplatin- due to higher initial binding as well as the lack of cisplatin
related kidney injury. It is believed that the thiol agent may adducts removal by the cell.
be forming a complex with cisplatin thus preventing the
formation of a glutathione-cisplatin-conjugate [8]. Ami-
fostine is one such drug. It is also the only FDA-approved Cytoplasmic organelle dysfunction
drug to prevent cisplatin-induced nephrotoxicity in patients
with non-small cell lung cancer and advanced ovarian can- Evidence from recent studies reveals that the endoplasmic
cer [12]. Unfortunately despite being FDA approved, the reticulum (ER) and mitochondria may play a critical role

13
J Nephrol

in cisplatin cellular toxicity [20, 21]. Mandic et al. were the activation of p53 which can trigger apoptosis. ROS, which
first to show this by using enucleated tumor cells to demon- forms either from DNA damage, mitochondrial dysfunc-
strate other triggers of apoptotic pathways via the ER [21]. tion or inflammatory triggers, can also trigger apoptosis.
They showed an upregulation in the caspase-12 activity in The many mechanisms through which cisplatin can exert
these cells. Caspase-12 is localized to the ER and is acti- its influence on the cell and cause cell death is precisely
vated by any changes to the calcium homeostasis, and ER the reason that makes it an indispensable anti-cancer drug
is an essential regulator of calcium homeostasis in the cyto- and a challenge to protect the healthy cells from innocent
sol [21]. bystander death. A simplified summary of this has been
The other important cytosolic target of cisplatin is depicted in Fig. 1.
believed to be the mitochondria. From an electrical charge The role of autophagy has also been explored, and its
perspective, the positively charged cisplatin electrophile protective role has been demonstrated by Periyasamy-
would preferentially accumulate in the negatively charged Thandavan et  al. in cultured proximal tubular cells [27].
mitochondria. Hirama et  al. demonstrated that cisplatin- They noted autophagosomes develop within hours of cis-
resistant ovarian tumor cells had a lower mitochondrial platin administration with p53 possibly playing a role in
membrane potential as compared to the cisplatin sensi- the signaling process. When autophagy was pharmaco-
tive cells [22] thus limiting the entry of cisplatin into the logically blocked, there was accelerated apoptotic process
mitochondria. The cisplatin-resistant cells also had less noted [27]. Anti-apoptotic measures have been evaluated
mitochondrial DNA damage. Mitochondrial DNA lack effi- for renoprotective strategy. P53 inhibition in  vitro studies
cient DNA repair mechanisms and are more susceptible to like siRNA targeted at p53 have shown a reduction in kid-
damage. ney injury but considering the vast array of cell protective
Mitochondrial dysfunction results in widespread effects. functions of p53, extensive in vivo studies will be needed
A drop in ATP synthesis forces the stressed cell to function before human use. A major theoretical concern as Fraser
in a starvation mode. It causes an activation of caspase- et al. demonstrated is the possibility of affecting cisplatin’s
mediated apoptosis by the release of caspase-9 mediators. anti-tumor activity by the p53 inhibition [28].
Abnormal lipid peroxidation, hypoxic injury from cispl-
atin effect on the local vasculature and the disruption of
the ATP synthesis process results in the production of free Oxidative stress
radicals or reactive oxygen species [23]. Mouse kidney cell
studies by Portilla have demonstrated a role of cisplatin in Oxidative stress is the imbalance between the free radi-
inhibiting fatty acid oxidation via peroxisome proliferator cal production and consumption. Three mechanisms have
activated receptor α (PPARα) transcription downregulation been proposed that result in the production of ROS [29].
thus affecting energy synthesis [24]. The role of a PPARα First, its interaction with glutathione during its activation
ligand was explored and was found to reduce the incidence to a more potent nephrotoxin depletes the cell of a potent
of acute kidney injury in mice [25]. antioxidant molecule like glutathione. Second, it affects the
Qian et al. showed an interesting association between the mitochondrial respiratory chain resulting in the production
cell sensitivity to cisplatin-induced cell death and the mito- of ROS and the third is via cytochrome p450 effect in the
chondrial density of that cell. They showed this in intestinal microsomes [29]. The downstream target of the ROS is not
cells where the density of mitochondria decreases as one clear. It appears to act via multiple mechanisms as would
moves from the duodenum to the ileum and they were able be expected based on their highly reactive nature. Admin-
to demonstrate a difference in the cell-specific sensitivity istration of an antioxidant dimethylthiourea (DMTU) abol-
to cisplatin with greater cell death occurring in the duode- ished p38 activation in one study [30] and p53 activation in
num as compared to ileum [26]. This is interesting as the another [31].
proximal tubule cells are known to have the highest density Supplementation with antioxidants has hence been con-
of mitochondria as compared to the rest of the tubules, and sidered for nephroprotection as well. Both natural and syn-
this is where cisplatin-induced damage is commonly noted. thetic formulations have been evaluated. Vitamin C, vita-
min E, selenium, alpha lipoic acid, ubiquinone and other
enzymatic antioxidants like DMTU are just to name a few.
Apoptosis Most of the beneficial effects were noted in the in  vitro
and in vivo mice studies [29]. An attempt to replicate this
Cisplatin activates not just the intrinsic apoptotic path- in humans by Weijl et  al. with vitamin C, vitamin E, and
ways (via the ER or mitochondrial dysfunction) but also selenium supplementation though showed no benefit from
the extrinsic death receptor pathway by tumor necrosis fac- the antioxidant micronutrient supplementation [32]. Mag-
tor alpha (TNFα) production. DNA damage can result in nesium is another supplement that has been shown to be

13
J Nephrol

Cisplan
Ctr1
OCT2

Entry into the cell

GGT
cysteine-S-conjugate b-lyase

Acvaon in the cell


Death receptor
pathway
Endoplasmic
Reculum
DAMPs DNA Damage pathway
P53 acvaon
Acvaon Mitochondrial
Of TLR4 ROS pathway
NF-KB
pathway Caspase 8
Cytochrome c AIF Caspase 12 acvaon
Cytokines/Chemokines
acvaon
TNFα
Caspase 9 Caspase
acvaon independent

Inflammaon Apoptosis

Fig. 1  Summary of the major pathways of Cisplatin induced tubu- molecular pattern molecules, TLR toll-like receptor, TNFα tumor
lar cell injury. Ctr1 copper transporter 1, OCT2 organic cation trans- necrosis factor alpha, ROS reactive oxygen species, AIF apoptosis
porter 2, GGT g-glutamyl transpeptidase, DAMPs damage-associated inducing factor

helpful in reducing cisplatin related toxicity. Solanki et al. play a vital role in the immune system. Via multiple
have noted in their mice studies that a deficiency of magne- pathways, TLRs in turn cause the release of chemokines
sium amplifies markers of renal damage which is promptly and other cytokines like TNFα to be released attracting
reversed by supplementation with magnesium. It is a cofac- the inflammatory cells [34, 35]. This was demonstrated
tor for many of the signaling pathways in the cell and its by Zhang et al. using TLR 4 knockout (KO) mice. They
role in regulation of oxidative stress, apoptosis and cispl- showed that the levels of cytokines like TNFα were sig-
atin efflux is well demonstrated in their studies without loss nificantly higher in the cisplatin-treated wild-type mice
of anti-tumor activity [33]. as compared to the cisplatin-treated TLR4 KO mice. The
former also had more renal damage as compared to the
latter suggesting that DAMPS-TLR4 TNFα pathway may
Inflammation be contributing to the renal injury [36] (Fig.  1). IL-10
may have a protective role in decreasing the inflammation
Cisplatin has also been found to have an inflamma- [37].
tory effect on the kidneys. Cisplatin is believed to cause In order to mitigate these effects of inflammation cas-
intracellularly injury resulting in the release of damage- cade, intervention at different stages has been evaluated.
associated molecular pattern molecules (DAMPS) also Salicylates have a well know anti-inflammatory action.
called “alarmins”. DAMPs are endogenous molecules Studies found cisplatin-exposed mice treated with salicy-
that are produced during tissue injury. DAMPs are known late had a significant reduction of TNFα protein levels in
to act on Toll-like receptors (TLRs). TLRS are a family their serum [38]. Considering higher doses are needed
of receptors that have a pattern recognition feature and for the anti-inflammatory effect and its usage is limited in

13
J Nephrol

patients with renal dysfunction, the use of salicylate may be urine via the organic anion transporter 1 (OAT1) [45, 46].
a challenge. This led to a phase I trial was performed by Jacobs et al. by
The production of TNFα after cisplatin administration infusing probenecid before and for 24 h after the adminis-
seems to be dependent on a nuclear factor-kappa B (NF- tration of cisplatin [47]. The authors found no significant
κB) pathway and TNFα, in turn, needs to act on endothe- nephrotoxicity in their probenecid treated patients despite
lial adhesion molecules (ICAM-1) to attract inflammatory dose escalation of cisplatin. Unfortunately, patients devel-
cells. Alpha lipoic acid has an anti-inflammatory effect oped ototoxicity followed by neurotoxicity at higher doses.
by affecting the expression of ICAM-1 and suppressing The OCT2 belongs to the SLC22 family of transporters.
the NF-κB signaling pathway. This was demonstrated by It is expressed in the intestine, kidney, liver and brain. In the
Kang et al. [39] in their study in 2009. Similarly, the role kidney, it also has a high expression on the basolateral side
of TNFα antagonism, TLR4 antagonism, and p38 inhibi- of the proximal convoluted tubule [48]. These transporters
tors has been explored [30, 36, 40]. A major concern is that are poly-specific uniporters that can operate in either direc-
by controlling the inflammation, there would be mitigation tion, but they preferentially mediate the transport of the cat-
of the long-term consequences of fibrosis. This is yet to be ions into the cell at normal membrane potential [49]. OCT2
explored. KO mice studies by Filipsi et  al. demonstrated protection
from cisplatin-mediated renal tubular damage [50]. Similar
studies were performed by Ciarimboli et al. using a double
Cellular uptake KO mice model, where both OCT 1 and 2 receptors were
knocked out [51, 52]. These KO mice had no signs of oto-
Cisplatin is a drug that is not only freely filtered but also toxicity and only mild nephrotoxicity. There was a substan-
actively secreted into the urine. Movement of cisplatin tial reduction (about four-fold) in the quantity of cisplatin
through the renal tubular cells is from a basolateral to api- that accumulated in the kidney [51]. Interestingly, the less
cal direction. Two primary transporters are now known nephrotoxic platinum compound carboplatin does not seem
to be involved in transporting cisplatin into the tubular to be transported via OCT2 [52]. Oxaliplatin is transported
cells, the human copper transport protein 1 (Ctr1) and the by OCT2 but it is rapidly transported out of the renal tubu-
organic cation transporter 2 (OCT2). Ctr1 is a protein that lar cell by multidrug and toxin extrusion (MATE/SLC47A)
is expressed in multiple organ sites in the body and its pri- thus reducing its nephrotoxicity [53]. Finally, procainamide
mary role, as the name implies, is in copper transport but is transported by OCT2 and has been shown to interfere
only in the last decade have we have found evidence for its with the transport of other organic cations. However, pro-
role in the transport of cisplatin [41]. In animal models, cainamide is mainly transported by OCT2A and its interfer-
Pabla et al. found high expression of Ctr1 in both proximal ence with cisplatin transport remains uninvestigated [54].
and distal tubules mostly localized on the basolateral side Many in vitro and in vivo studies have been done look-
[42]. The KO Ctr1 mice also showed a decrease in cisplatin ing into the role of cimetidine in blocking the renal effects
uptake resulting in a decrease in tubular cell apoptosis at of cisplatin [51, 52]. Sleijfer et  al. had conducted a small
low dose exposure as well as cell necrosis in high dose cis- case-control study wherein verapamil and cimetidine were
platin exposure. administered to a group of patients [55]. Verapamil was
The presence of excess copper degrades and down- given mainly for its vasodilatory effects in the kidney. They
regulates the Ctr1 expression. A similar effect is induced noted that the patients that received the two-drug combina-
by cisplatin [41]. This has led to the discovery that certain tion did not have a significant drop in the glomerular filtra-
cisplatin-resistant tumors have low expressions of Ctr1 tion rate (GFR) as compared to those that received cisplatin
[43]. It has also been noted that certain resistant tumors alone.
have high expression of copper effluxors on the cancer cell
membrane. Recently, it has been pointed out that the use
of copper chelator may help improve the sensitivity of the Clinical presentation of cisplatin nephrotoxicity
previously cisplatin-resistant tumors [44]. So consider-
ing the crucial role Ctr1 plays in the entry of cisplatin into Nephrotoxicity with cisplatin does not refer to renal failure
the tumor cell, concern has been raised that targeting this alone. In fact, acute kidney injury is seen in just approxi-
transporter for a renoprotective strategy may affect the anti- mately 20–30% of the patients [56–58]. Before the era of
tumor effects of the drug. hydration and diuresis, the incidence was noted in almost
Work has also been done looking at the role of secre- 100% of the patients [56, 58]. The more common kidney
tion of cisplatin from the tubules into the lumen and its role manifestation is hypomagnesemia, the prevalence is esti-
in causing tubular injury. In rats, probenecid was found to mated to be between 40 and 100% [59–61]. Other mani-
be capable of blocking cisplatin tubular secretion into the festations Fanconi-like syndrome [62–64], distal renal

13
J Nephrol

tubular acidosis [65], renal concentrating defect [66, 67] Table 2  Risk factors for Cisplatin induced nephrotoxicity
and thrombotic microangiopathy [68] are among a few of
 Patient factors
the well-known complications (Table 1).
 Older age
 Low functional status
 Malnourished
Risk factors for cisplatin nephrotoxicity
 Dehydration
 Chronic comorbid illness —diabetes mellitus, Liver dysfunction
It is well known that cisplatin kidney injury is dose-dura-
 Preexisting chronic kidney disease
tion-frequency dependent [58, 75]. Higher peak plasma
 Concurrent nephrotoxic drug use (iodinated contrast, NSAIDS,
concentrations from higher doses per treatment result in gemcitabine)
greater injury [76, 77]. A higher cumulative dose has also  Electrolyte disturbances (low magnesium)
been shown to increase risk for future kidney injury [78].  Alcohol ingestion
There are also certain patient-related factors that make Cisplatin drug related factors
them more susceptible to kidney damage. Ignard–Bagnis  High dose per treatment (greater 50 mg/m2)
et al. demonstrated that older patients, patients with signifi-  Frequency of administration
cant comorbid illness and baseline poor functional status  Cumulative dose
were at a higher risk [23]. Additionally, patients with lower  Hydration with cisplatin administration
albumin are also presumed to have a higher unbound frac-
tion of cisplatin resulting in greater peak plasma concen- NSAIDS non-steroidal anti-inflammatory drugs
trations [75]. Patients with preexisting kidney disease and
concurrent use of nephrotoxic medications (like non-steroi- management. Based on the pathophysiology, most of the
dal anti-inflammatory drugs NSAIDS, iodinated contrast) damage is expected to be in the tubulo-interstitial com-
are also at a greater risk [23, 75] (Table 2). partment with minimal glomerular changes which has
been confirmed in both rat and human autopsy studies
[79, 80]. There appeared to be a gradual involvement of
Pathological changes in cisplatin nephrotoxicity the tubules with a predominant proximal tubular compo-
nent. It is not known to cause interstitial nephritis [80].
Patients with cisplatin-mediated kidney injury do not In patients who have had chronic use of cisplatin, it has
need a routine kidney biopsy unless the clinical situa- been noted to cause significant tubular atrophy and inter-
tion demands a clarification that can alter the patient’s stitial fibrosis with relative glomerular sparing [81].

Table 1  Some common Clinical manifestations of Cisplatin induced Nephrotoxicity


Clinical manifestation Mechanism References

Acute kidney injury Dose and frequency dependent [58, 64]


Hypomagnesaemia Possibly related to distal tubular injury affecting magnesium [69, 70]
reabsorption
Magnesium deficiency may also exacerbate the renal toxicity of
cisplatin
Fanconi-like syndrome From proximal tubular injury [71]
Distal renal tubular acidosis From tubular injury [65]
Renal concentrating defect Affects proximal tubular sodium reabsorption [72, 73]
Occurs in 2 phases Decreases aquaporin expression in the distal tubule
 a. Early phase (within 24–48 h) and resolve spontaneously. Presumed also due to damage to loop of Henle especially Na-K-
Presumed prostaglandin mediated. Prevented by vasopressin 2Cl transporter and affecting countercurrent regulatory system
and aspirin
 b. Second phase (72–96 h) with a decrease in GFR. Associated
with decrease in medullary tonicity
Thrombotic microangiopathy Unclear but has been seen in conjunction with bleomycin or [68]
gemcitabine use
cisplatin-induced glucose intolerance Rat studies show a relative insulin deficiency state as well as an [74]
impaired glucagon response

GFR glomerular filtration rate; Na-K-2Cl sodium–potassium-2 chloride

13
Table 3  Strategies for preventing or reducing nephrotoxicity from Cisplatin that have been explored
Level of action Drug Mechanism of action Comments
J Nephrol

Glomerular filtration Micellar/liposomal cisplatin Incorporating cisplatin into polymeric micelles which can Mice models showed a decrease in nephrotoxicity but
possibly reduce its filtration into urine caused hepatic dysfunction
Cell entry OCT2 Inhibitors: Cimetidine, metformin Reduces the entry of cisplatin into the tubular cells as a Cimetidine appears to be promising but paucity of human
competitive substrate for OCT2 receptor studies
Ctr1 inhibitors: copper Reduces the entry of cisplatin into the tubular cells as a Concern for affecting the anti-tumor effect of cisplatin has
competitive substrate for Ctr1 receptor been raised
Cell activation GGT inhibitors: acivicin Reduces activation of cisplatin-Glutathione conjugate into Acivicin is the drug used for GGT inhibition and has poten-
a more reactive and nephrotoxic thiol compound tial anti-neoplastic activity as well that is being explored
Glutathione transferase inhibitors: ketoprofen Reduces activation of cisplatin-glutathione conjugate into The mice model used ketoprofen as the inhibitor which is
a more reactive and nephrotoxic thiol compound a non-steroidal anti-inflammatory drug that can cause
kidney injury on its own
High dose glutathione Competitively inhibits GGT and so preventing the metabo- High doses of glutathione are needed
lism of glutathione-cisplatin-conjugate into a more
potent nephrotoxin
Thiol agents: amifostine, procainamide Complexes with cisplatin and preventing the formation of Amifostine is the only FDA approved drug for reducing
a glutathione-cisplatin-conjugate and its further metabo- nephrotoxicity in ovarian cancer and non-small cell lung
lism into a more potent nephrotoxin cancer
Inflammatory response TNFα antagonists Prevent the release of cytokine/chemokine that propagate –
TLR4 antagonists the inflammatory process
p38 inhibitors
JNK inhibitors Cisplatin causes reactive oxygen species which stimulates The selective inhibitor reduced not only renal inflammation
the JNK pathway causing the release of pro-inflamma- but also apoptotic cell death
tory cytokines
Salicylates Reduces the production of TNF-alpha High doses are needed for anti-inflammatory action which
may be the limiting factor in these that have a propensity
for renal dysfunction
PPAR-α ligands [25, 84] Reduced the expression of cytokine/chemokines by cispl- Fibrates are PPAR α ligands used in dyslipidemia and one
atin injured cell likely via inhibition of NF-κB pathway could consider its role in patients that also have concur-
Also has a role in mitochondrial fatty acid oxidation and rent lipid disorder
energy synthesis which is affected by cisplatin
Alpha lipoic acid Reduces the expression of ICAM-1 on endothelial cells Currently used as an adjunct antioxidant effect in patients
and suppresses the NF-κ B signaling pathway for TNFα with painful neuropathy
production
IL-10 [85] Known anti-inflammatory cytokine IL-10 was found to be protective from cisplatin associated
injury

13

Table 3  (continued)
Level of action Drug Mechanism of action Comments

13
Oxidative stress Natural antioxidants [29] In vitro and in vivo studies have shown a reduction in the Human studies have not shown any benefit
 Selenium ROS generation as well as reduction in the renal injury
 Magnesium
 Vitamin E
 Vitamin C
 Naringenin (citrus fruits)
 Lycopene (tomatoes)
 Spirulin (algae)
 Garlic
Among others
Enzymatic anti-oxidants [29]
 Superoxide dismutase
 N-acetyl cysteine
 Dimethylthiourea (DMTU)
Among others
PARP inhibitors Inhibition of PARP activation was shown to downstream Enalapril is an inhibitor of PARP activation and was studied
effects of reduction in oxidative stress pathways of with reduction in renal ­injurya
MAPKs/TNFα/NF-κB mediated inflammation and
apoptosis
Anti-apoptosis P53 inhibitors P53 is a major mediator in cisplatin induced apoptosis P53 suppression was shown by Fraser et al. to promote
and the trigger is believed to be DNA damage oxidative cisplatin resistance in ovarian cancer cell lines
stress
P21 agonist/CDK2 inhibitors P21 is a cell cycle inhibitor which upregulates on exposure
to cisplatin and by upregulating it tubule cell ­protectionb

OCT2 organic cation transporter 2, Ctr1 copper transporter 1, GGT g-glutamyl transpeptidase, TNFα Tumor necrosis factor- alpha, TLR = Toll-like receptor, JNK c-jun N-terminal kinase,
PPAR-α peroxisome proliferator activated receptor alpha, NF-κB nuclear factor κB, IL interleukin, PARP poly ADP ribose polymerase, MAPK mitogen-activated protein kinases
a
 Rani et al. [86]
b
 Price et al [87]
J Nephrol
J Nephrol

Long‑term kidney consequences in cisplatin Conflict of interest  We do not have any financial or non-financial
nephrotoxicity potential conflicts of interest.

Ethical approval  This article does not contain any studies with
In the last few decades, significant effects of chronic kid- human participants performed by any of the authors.
ney disease (CKD) on the overall morbidity and mortality
of these patients have been noted. To add to this, more
patients are surviving longer after cancer treatment and
in their older age are dealing with the consequences of References
the chronic kidney disease that they acquired from their
intense therapy earlier in their life. Dekker et  al. looked 1. Prestayko AW, Crooke ST, Carter SK, University of Alabama
in Birmingham (1980) Comprehensive Cancer Center., Bristol
at over 800 childhood cancer survivors with a median Laboratories.: Cisplatin, current status and new developments.
follow-up of 18 years noted that one of the risk factors for Academic Press, New York
development of CKD was high dose cisplatin use [82]. 2. Liira J, Verbeek JH, Costa G, Driscoll TR, Sallinen M, Isotalo
Interestingly a Cochrane Systematic review showed a LK, Ruotsalainen JH (2014) Pharmacological interventions for
sleepiness and sleep disturbances caused by shift work. Cochrane
trend toward cisplatin-based chemotherapy and chronic Database Syst Rev 2014(8):CD009776
kidney disease but did not reach statistical significance. 3. Florea AM, Busselberg D (2011) Cisplatin as an anti-tumor
They did note a significant association with proteinuria. drug: cellular mechanisms of activity, drug resistance and
Significant heterogeneity of the data with limited follow- induced side effects. Cancers (Basel) 3(1):1351–1371
4. Pabla N, Dong Z (2008) Cisplatin nephrotoxicity: mechanisms
up made it difficult to come to a definite conclusion [83]. and renoprotective strategies. Kidney Int 73(9):994–1007
5. Townsend DM, Hanigan MH (2002) Inhibition of gamma-glu-
tamyl transpeptidase or cysteine S-conjugate beta-lyase activity
blocks the nephrotoxicity of cisplatin in mice. J Pharmacol Exp
Ther 300(1):142–148
Experimental strategies to prevent cisplatin 6. Townsend DM, Deng M, Zhang L, Lapus MG, Hanigan MH
nephrotoxicity (2003) Metabolism of Cisplatin to a nephrotoxin in proximal
tubule cells. J Am Soc Nephrol 14(1):1–10
Based on the pathophysiology discussion earlier, there 7. Cozzaglio L, Doci R, Colella G, Zunino F, Casciarri G, Gennari
L (1990) A feasibility study of high-dose cisplatin and 5-fluoro-
are many potential options for intervention and preven- uracil with glutathione protection in the treatment of advanced
tion of cisplatin-induced kidney injury. As previously colorectal cancer. Tumori 76(6):590–594
mentioned, many have been tried. The biggest challenge 8. Zunino F, Pratesi G, Micheloni A, Cavalletti E, Sala F, Tofanetti
is to protect the kidney while not reducing its tumor- O (1989) Protective effect of reduced glutathione against cispl-
atin-induced renal and systemic toxicity and its influence on the
cidal effects. The best-known and most commonly used therapeutic activity of the antitumor drug. Chem Biol Interact
strategy is that of hydration and forced diuresis with diu- 70(1–2):89–101
retics or mannitol which will not be further discussed 9. Gosland M, Lum B, Schimmelpfennig J, Baker J, Doukas M
here. Table  3 lists a summary of some of the strategies (1996) Insights into mechanisms of cisplatin resistance and
potential for its clinical reversal. Pharmacotherapy 16(1):16–39
that have been explored and have a potential for future 10. Godwin AK, Meister A, O’Dwyer PJ, Huang CS, Hamilton TC,
research. Anderson ME (1992) High resistance to cisplatin in human ovar-
ian cancer cell lines is associated with marked increase of glu-
tathione synthesis. Proc Natl Acad Sci USA 89(7):3070–3074
11. Hanigan MH, Gallagher BC, Townsend DM, Gabarra V (1999)
Gamma-glutamyl transpeptidase accelerates tumor growth and
Future direction increases the resistance of tumors to cisplatin in vivo. Carcino-
genesis 20(4):553–559
In summary, the last few years has given us new insights 12. Kemp G, Rose P, Lurain J, Berman M, Manetta A, Roullet B,
Homesley H, Belpomme D, Glick J (1996) Amifostine pre-
regarding the various mechanisms at play in causing treatment for protection against cyclophosphamide-induced
cisplatin-induced nephrotoxicity. The complexity of cis- and cisplatin-induced toxicities: results of a randomized con-
platin’s cytotoxic actions explains why nephrotoxicity trol trial in patients with advanced ovarian cancer. J Clin Oncol
continues to remains a major challenge. Uncovering the 14(7):2101–2112
13. Sastry J, Kellie SJ (2005) Severe neurotoxicity, ototoxicity and
pathways will trigger research looking at focused reno- nephrotoxicity following high-dose cisplatin and amifostine.
protective strategies, and hopefully, human clinical trials Pediatr Hematol Oncol 22(5):441–445
will soon follow. 14. Ekborn A, Hansson J, Ehrsson H, Eksborg S, Wallin I, Wagenius
G, Laurell G (2004) High-dose Cisplatin with amifostine: oto-
Compliance with ethical standards  toxicity and pharmacokinetics. Laryngoscope 114(9):1660–1667
15. Esposito M, Viale M, Vannozzi MO, Zicca A, Cadoni A,

Merlo F, Gogioso L (1996) Effect of the antiarrhythmic
Funding None.
drug procainamide on the toxicity and antitumor activity of

13
J Nephrol

cis-diamminedichloroplatinum(II). Toxicol Appl Pharmacol 34. Gluba A, Banach M, Hannam S, Mikhailidis DP, Sakowicz A,
140(2):370–377 Rysz J (2010) The role of Toll-like receptors in renal diseases.
16. Viale M, Vannozzi MO, Pastrone I, Mariggio MA, Zicca A, Nat Rev Nephrol 6(4):224–235
Cadoni A, Cafaggi S, Tolino G, Lunardi G, Civalleri D et  al 35. Kono H, Rock KL (2008) How dying cells alert the immune sys-
(2000) Reduction of cisplatin nephrotoxicity by procainamide: tem to danger. Nat Rev Immunol 8(4):279–289
does the formation of a cisplatin-procainamide complex play a 36. Zhang B, Ramesh G, Uematsu S, Akira S, Reeves WB (2008)
role? J Pharmacol Exp Ther 293(3):829–836 TLR4 signaling mediates inflammation and tissue injury in
17. Miller RP, Tadagavadi RK, Ramesh G, Reeves WB (2010)
nephrotoxicity. J Am Soc Nephrol 19(5):923–932
Mechanisms of Cisplatin nephrotoxicity. Toxins (Basel) 37. Tadagavadi RK, Reeves WB (2010) Endogenous IL-10 attenu-
2(11):2490–2518 ates cisplatin nephrotoxicity: role of dendritic cells. J Immunol
18. Wang D, Lippard SJ (2005) Cellular processing of platinum anti- 185(8):4904–4911
cancer drugs. Nat Rev Drug Discov 4(4):307–320 38. Ramesh G, Reeves WB (2004) Salicylate reduces cisplatin

19. Jamieson ER, Lippard SJ (1999) Structure, recognition, and pro- nephrotoxicity by inhibition of tumor necrosis factor-alpha. Kid-
cessing of cisplatin-DNA adducts. Chem Rev 99(9):2467–2498 ney Int 65(2):490–499
20. Cullen KJ, Yang Z, Schumaker L, Guo Z (2007) Mitochondria 39. Kang KP, Kim DH, Jung YJ, Lee AS, Lee S, Lee SY, Jang KY,
as a critical target of the chemotheraputic agent cisplatin in head Sung MJ, Park SK, Kim W (2009) Alpha-lipoic acid attenuates
and neck cancer. J Bioenerg Biomembr 39(1):43–50 cisplatin-induced acute kidney injury in mice by suppressing
21. Mandic A, Hansson J, Linder S, Shoshan MC (2003) Cisplatin renal inflammation. Nephrol Dial Transplant 24(10):3012–3020
induces endoplasmic reticulum stress and nucleus-independent 40. Ramesh G, Reeves WB (2002) TNF-alpha mediates chemokine
apoptotic signaling. J Biol Chem 278(11):9100–9106 and cytokine expression and renal injury in cisplatin nephrotox-
22. Hirama M, Isonishi S, Yasuda M, Ishikawa H (2006) Characteri- icity. J Clin Investig 110(6):835–842
zation of mitochondria in cisplatin-resistant human ovarian car- 41. Ishida S, Lee J, Thiele DJ, Herskowitz I (2002) Uptake of

cinoma cells. Oncol Rep 16(5):997–1002 the anticancer drug cisplatin mediated by the copper trans-
23. Isnard-Bagnis C, Moulin B, Launay-Vacher V, Izzedine H, Tos- porter Ctr1 in yeast and mammals. Proc Natl Acad Sci USA
tivint I, Deray G (2005) [Anticancer drug-induced nephrotoxic- 99(22):14298–14302
ity]. Nephrol Ther 1(2):101–114 42. Pabla N, Murphy RF, Liu K, Dong Z (2009) The copper trans-
24. Portilla D, Dai G, McClure T, Bates L, Kurten R, Megyesi J, porter Ctr1 contributes to cisplatin uptake by renal tubular cells
Price P, Li S (2002) Alterations of PPARalpha and its coactiva- during cisplatin nephrotoxicity. Am J Physiol Renal Physiol
tor PGC-1 in cisplatin-induced acute renal failure. Kidney Int 296(3):F505–511
62(4):1208–1218 43. Hall MD, Okabe M, Shen DW, Liang XJ, Gottesman MM (2008)
25. Li S, Wu P, Yarlagadda P, Vadjunec NM, Proia AD, Harris RA, The role of cellular accumulation in determining sensitivity to
Portilla D (2004) PPAR alpha ligand protects during cisplatin- platinum-based chemotherapy. Annu Rev Pharmacol Toxicol
induced acute renal failure by preventing inhibition of renal FAO 48:495–535
and PDC activity. Am J Physiol Renal Physiol 286(3):F572-580. 44. Ishida S, McCormick F, Smith-McCune K, Hanahan D (2010)
26. Qian W, Nishikawa M, Haque AM, Hirose M, Mashimo M, Sato Enhancing tumor-specific uptake of the anticancer drug cisplatin
E, Inoue M (2005) Mitochondrial density determines the cellu- with a copper chelator. Cancer Cell 17(6):574–583
lar sensitivity to cisplatin-induced cell death. Am J Physiol Cell 45. Jacobs C, Coleman CN, Rich L, Hirst K, Weiner MW (1984)
Physiol 289(6):C1466–C1475 Inhibition of cis-diamminedichloroplatinum secretion by the
27. Periyasamy-Thandavan S, Jiang M, Wei Q, Smith R, Yin XM, human kidney with probenecid. Cancer Res 44(8):3632–3635
Dong Z (2008) Autophagy is cytoprotective during cisplatin 46. Klein J, Bentur Y, Cheung D, Moselhy G, Koren G (1991) Renal
injury of renal proximal tubular cells. Kidney Int 74(5):631–640 handling of cisplatin: interactions with organic anions and cati-
28. Fraser M, Bai T, Tsang BK (2008) Akt promotes cisplatin
ons in the dog. Clin Investig Med 14(5):388–394
resistance in human ovarian cancer cells through inhibition 47. Jacobs C, Kaubisch S, Halsey J, Lum BL, Gosland M, Coleman
of p53 phosphorylation and nuclear function. Int J Cancer CN, Sikic BI (1991) The use of probenecid as a chemoprotector
122(3):534–546 against cisplatin nephrotoxicity. Cancer 67(6):1518–1524
29. Chirino YI, Pedraza-Chaverri J (2009) Role of oxidative and 48. Koepsell H (1998) Organic cation transporters in intestine, kid-
nitrosative stress in cisplatin-induced nephrotoxicity. Exp Toxi- ney, liver, and brain. Annu Rev Physiol 60:243–266
col Pathol 61(3):223–242 49. Koepsell H, Schmitt BM, Gorboulev V (2003) Organic cation
30. Ramesh G, Reeves WB (2005) p38 MAP kinase inhibition ame- transporters. Rev Physiol Biochem Pharmacol 150:36–90
liorates cisplatin nephrotoxicity in mice. Am J Physiol Renal 50. Filipski KK, Mathijssen RH, Mikkelsen TS, Schinkel AH, Spar-
Physiol 289(1):F166–174 reboom A (2009) Contribution of organic cation transporter 2
31. Jiang M, Wei Q, Pabla N, Dong G, Wang CY, Yang T, Smith SB, (OCT2) to cisplatin-induced nephrotoxicity. Clin Pharmacol
Dong Z (2007) Effects of hydroxyl radical scavenging on cispl- Ther 86(4):396–402
atin-induced p53 activation, tubular cell apoptosis and nephro- 51. Ciarimboli G, Deuster D, Knief A, Sperling M, Holtkamp M,
toxicity. Biochem Pharmacol 73(9):1499–1510 Edemir B, Pavenstadt H, Lanvers-Kaminsky C, am Zehnhoff-
32. Weijl NI, Elsendoorn TJ, Lentjes EG, Hopman GD, Wipkink- Dinnesen A, Schinkel AH et al (2010) Organic cation transporter
Bakker A, Zwinderman AH, Cleton FJ, Osanto S (2004) Sup- 2 mediates cisplatin-induced oto- and nephrotoxicity and is a tar-
plementation with antioxidant micronutrients and chemotherapy- get for protective interventions. Am J Pathol 176(3):1169–1180
induced toxicity in cancer patients treated with cisplatin-based 52. Ciarimboli G, Ludwig T, Lang D, Pavenstadt H, Koepsell H,
chemotherapy: a randomised, double-blind, placebo-controlled Piechota HJ, Haier J, Jaehde U, Zisowsky J, Schlatter E (2005)
study. Eur J Cancer 40(11):1713–1723 Cisplatin nephrotoxicity is critically mediated via the human
33. Solanki MH, Chatterjee PK, Xue X, Gupta M, Rosales I, Yeboah organic cation transporter 2. Am J Pathol 167(6):1477–1484
MM, Kohn N, Metz CN: Magnesium protects against cisplatin- 53. Yonezawa A, Inui K (2011) Organic cation transporter OCT/
induced acute kidney injury without compromising cisplatin- SLC22A and H(+)/organic cation antiporter MATE/SLC47A are
mediated killing of an ovarian tumor xenograft in mice. Am J key molecules for nephrotoxicity of platinum agents. Biochem
Physiol Renal Physiol 2015, 309(1):F35–47 Pharmacol 81(5):563–568

13
J Nephrol

54. Urakami Y, Akazawa M, Saito H, Okuda M, Inui K (2002) 72. Kim SW, Lee JU, Nah MY, Kang DG, Ahn KY, Lee HS, Choi
cDNA cloning, functional characterization, and tissue distribu- KC (2001) Cisplatin decreases the abundance of aquaporin water
tion of an alternatively spliced variant of organic cation trans- channels in rat kidney. J Am Soc Nephrol 12(5):875–882
porter hOCT2 predominantly expressed in the human kidney. J 73. Yao X, Panichpisal K, Kurtzman N, Nugent K (2007) Cisplatin
Am Soc Nephrol 13(7):1703–1710 nephrotoxicity: a review. Am J Med Sci 334(2):115–124
55. Sleijfer DT, Offerman JJ, Mulder NH, Verweij M, van der Hem 74. Goldstein RS, Mayor GH, Gingerich RL, Hook JB, Rosenbaum
GK, Schraffordt Koops HS, Meijer S (1987) The protective RW, Bond JT (1983) The effects of cisplatin and other divalent
potential of the combination of verapamil and cimetidine on cis- platinum compounds on glucose metabolism and pancreatic
platin-induced nephrotoxicity in man. Cancer 60(11):2823–2828 endocrine function. Toxicol Appl Pharmacol 69(3):432–441
56. Goldstein RS, Mayor GH (1983) Minireview. The nephrotoxicity 75. Stewart DJ, Dulberg CS, Mikhael NZ, Redmond MD, Montpetit
of cisplatin. Life Sci 32(7):685–690 VA, Goel R (1997) Association of cisplatin nephrotoxicity with
57. Hartmann JT, Kollmannsberger C, Kanz L, Bokemeyer C (1999) patient characteristics and cisplatin administration methods.
Platinum organ toxicity and possible prevention in patients with Cancer Chemother Pharmacol 40(4):293–308
testicular cancer. Int J Cancer 83(6):866–869 76. Reece PA, Stafford I, Russell J, Khan M, Gill PG (1987) Cre-
58. Madias NE, Harrington JT (1978) Platinum nephrotoxicity. Am J atinine clearance as a predictor of ultrafilterable platinum dis-
Med 65(2):307–314 position in cancer patients treated with cisplatin: relationship
59. Maxwell MH, Kleeman CR, Narins RG (1994) Maxwell & Klee- between peak ultrafilterable platinum plasma levels and nephro-
man’s clinical disorders of fluid and electrolyte metabolism, toxicity. J Clin Oncol 5(2):304–309
5th edn. McGraw-Hill, Health Professions Division, New York 77. Skinner R, Pearson AD, English MW, Price L, Wyllie RA, Coul-
60. Schilsky RL, Anderson T (1979) Hypomagnesemia and renal thard MG, Craft AW (1998) Cisplatin dose rate as a risk factor
magnesium wasting in patients receiving cisplatin. Ann Intern for nephrotoxicity in children. Br J Cancer 77(10):1677–1682
Med 90(6):929–931 78. Caglar K, Kinalp C, Arpaci F, Turan M, Saglam K, Ozturk B,
61. Sutton RA, Walker VR, Halabe A, Swenerton K, Coppin CM Komurcu S, Yavuz I, Yenicesu M, Ozet A et al (2002) Cumula-
(1991) Chronic hypomagnesemia caused by cisplatin: effect of tive prior dose of cisplatin as a cause of the nephrotoxicity of
calcitriol. J Lab Clin Med 117(1):40–43 high-dose chemotherapy followed by autologous stem-cell trans-
62. Goldstein RS, Mayor GH, Rosenbaum RW, Hook JB, Santiago plantation. Nephrol Dial Transplant 17(11):1931–1935
JV, Bond JT (1982) Glucose intolerance following cis-platinum 79. Tanaka H, Ishikawa E, Teshima S, Shimizu E (1986) Histopatho-
treatment in rats. Toxicology 24(3–4):273–280 logical study of human cisplatin nephropathy. Toxicol Pathol
63. Kim YK, Byun HS, Kim YH, Woo JS, Lee SH (1995) Effect of 14(2):247–257
cisplatin on renal function in rabbits: mechanism of reduced glu- 80. Vickers AE, Rose K, Fisher R, Saulnier M, Sahota P, Bentley P
cose reabsorption. Toxicol Appl Pharmacol 130(1):19–26 (2004) Kidney slices of human and rat to characterize cisplatin-
64. Wangila GW, Nagothu KK, Steward R, 3rd, Bhatt R, Iyere PA, induced injury on cellular pathways and morphology. Toxicol
Willingham WM, Sorenson JR, Shah SV, Portilla D (2006) Pre- Pathol 32(5):577–590
vention of cisplatin-induced kidney epithelial cell apoptosis with 81. Cornelison TL, Reed E (1993) Nephrotoxicity and hydration

a Cu superoxide dismutase-mimetic [copper2II(3,5-ditertiarybut management for cisplatin, carboplatin, and ormaplatin. Gynecol
ylsalicylate)4(ethanol)4]. Toxicol In Vitro 20(8):1300–1312 Oncol 50(2):147–158
65. Swainson CP, Colls BM, Fitzharris BM (1985) Cis-platinum and 82. Dekkers IA, Blijdorp K, Cransberg K, Pluijm SM, Pieters R,
distal renal tubule toxicity. NZ Med J 98(779):375–378 Neggers SJ, van den Heuvel-Eibrink MM (2013) Long-term
66. Safirstein R, Miller P, Dikman S, Lyman N, Shapiro C (1981) nephrotoxicity in adult survivors of childhood cancer. Clin J Am
Cisplatin nephrotoxicity in rats: defect in papillary hypertonicity. Soc Nephrol CJASN 8(6):922–929
Am J Physiol 241(2):F175–185 83. Knijnenburg SL, Mulder RL, Schouten-Van Meeteren AY,

67. Seguro AC, Shimizu MH, Kudo LH, dos Santos Rocha A (1989) Bokenkamp A, Blufpand H, van Dulmen-den Broeder E, Veen-
Renal concentration defect induced by cisplatin. The role of ing MA, Kremer LC, Jaspers MW: Early and late renal adverse
thick ascending limb and papillary collecting duct. Am J Neph- effects after potentially nephrotoxic treatment for childhood can-
rol 9(1):59–65 cer. Cochrane Database Syst Rev 2013(10):CD008944
68. Jackson AM, Rose BD, Graff LG, Jacobs JB, Schwartz JH,
84. Li S, Gokden N, Okusa MD, Bhatt R, Portilla D: Anti-inflamma-
Strauss GM, Yang JP, Rudnick MR, Elfenbein IB, Narins RG tory effect of fibrate protects from cisplatin-induced ARF. Am J
(1984) Thrombotic microangiopathy and renal failure associated Physiol Renal Physiol 2005, 289(2):F469–480
with antineoplastic chemotherapy. Ann Intern Med 101(1):41–44 85. Deng J, Kohda Y, Chiao H, Wang Y, Hu X, Hewitt SM, Miyaji
69. Lam M, Adelstein DJ (1986) Hypomagnesemia and renal mag- T, McLeroy P, Nibhanupudy B, Li S et al (2001) Interleukin-10
nesium wasting in patients treated with cisplatin. Am J Kidney inhibits ischemic and cisplatin-induced acute renal injury. Kid-
Dis 8(3):164–169 ney Int 60(6):2118–2128
70. Lajer H, Kristensen M, Hansen HH, Nielsen S, Frokiaer J, Oster- 86. Rani N, Bharti S, Tomar A, Dinda AK, Arya DS, Bhatia J (2016)
gaard LF, Christensen S, Daugaard G, Jonassen TE (2005) Mag- Inhibition of PARP activation by enalapril is crucial for its reno-
nesium depletion enhances cisplatin-induced nephrotoxicity. protective effect in cisplatin-induced nephrotoxicity in rats. Free
Cancer Chemother Pharmacol 56(5):535–542 Radic Res 50(11):1226–1236
71. Portilla D, Li S, Nagothu KK, Megyesi J, Kaissling B, Schnack- 87. Price PM, Safirstein RL, Megyesi J: Protection of renal cells
enberg L, Safirstein RL, Beger RD (2006) Metabolomic study of from cisplatin toxicity by cell cycle inhibitors. Am J Physiol
cisplatin-induced nephrotoxicity. Kidney Int 69(12):2194–2204 Renal Physiol 2004, 286(2):F378–384

13

You might also like