Sunteți pe pagina 1din 9

Onco-Nephrology: Renal Toxicities of Chemotherapeutic

Agents
Mark A. Perazella

Summary
Despite dramatic improvements in patient survival and drug tolerability, nephrotoxicity remains an important
complication of chemotherapy. Adverse renal effects occur because of innate drug toxicity and a number of
patient- and drug-related factors. To provide cutting edge care for these patients, nephrologists and oncologists Section of
Nephrology,
must be familiar with the nephrotoxicity of these drugs, particularly their associated clinical and laboratory Department of
manifestations. Rapid diagnosis, targeted treatment, and supportive care are critical to improving care for these Medicine, Yale
patients. Unfortunately, some patients who develop nephrotoxicity will be left with long-term complications such University, New
as chronic tubulopathies and CKD. Onco-Nephrology is a new area that is rapidly expanding and requires a Haven, Connecticut
close working relationship between oncologists and nephrologists.
Correspondence: Dr.
Clin J Am Soc Nephrol 7: 1713–1721, 2012. doi: 10.2215/CJN.02780312
Mark A. Perazella,
Section of
Nephrology,
Introduction with exposure to a potential nephrotoxin. In fact, Department of
Treatment of cancer has undergone many significant Medicine, Yale
nearly 60% of patients with cancer have some form
University, 330 Cedar
advances in recent times. As such, the oncology land- of renal disease (1,2). Direct malignant effects include Street, New Haven,
scape has changed and improved dramatically for myeloma-related kidney injury, infiltration of the re- CT 06520-8029.
many patients. Those patients previously considered nal parenchyma as seen with leukemias and lympho- Email: mark.
therapy failures are now deriving significant benefit mas, urinary tract obstruction from various cancers, perazella@yale.edu
with decreased tumor progression and increased sur- and secondary glomerulopathies (2). Indirect effects in-
vival, often with less severe adverse systemic drug clude true or effective volume depletion from nausea/
effects. Despite this positive advancement in chemo- vomiting, diarrhea, overdiuresis, malignant ascites or
therapeutics for various malignancies, drug nephro- pleural effusions, sepsis, and cardiac involvement,
toxicity remains a complication and sometimes limits which sensitizes the kidney to nephrotoxins by
life-saving therapy (1–3). inducing a prerenal state (2). Also, susceptibility to
As a number of effective but potentially nephrotoxic drug toxicity occurs with metabolic disturbances such
chemotherapeutic regimens are released into clinical as hyperuricemia and hypercalcemia.
practice, oncologists and consulting nephrologists Undoubtedly, the toxicity of the chemotherapeutic
should be familiar with their adverse renal effects. These agent used importantly determines both the develop-
effects include the clinical and histopathologic manifes- ment and type of kidney injury sustained. High doses
tations of renal toxicity (1–6). Clinicians should also be and prolonged therapy increase the chance of renal
well versed in preventive measures available for the injury developing, regardless of the absence of other
various chemotherapy regimens, as well as effective risk factors (2–6). Furthermore, combined exposure of
treatment options for nephrotoxic consequences (1–6). chemotherapeutic agents with other nephrotoxins
The relationship of nephrologists and oncologists in the will raise the risk for kidney injury (7–9).
care of these patients has given rise to the nascent but There are a number of patient-specific risk factors
growing area of Onco-Nephrology. that must be considered with chemotherapy-associated
nephrotoxicity. Many patients are elderly—possessing
reduced total body water and an unrecognized de-
Renal Susceptibility to Chemotherapeutic pressed GFR, higher rates of renal oxidative stress,
Agents and excessive levels of angiotensin-II/endothelin, all
Not all patients exposed to nephrotoxic chemother- of which increase drug nephrotoxicity (10,11). Another
apeutic agents develop kidney injury, suggesting the nonmodifiable risk factor is the host’s underlying ge-
presence of several factors that enhance patient risk for netic makeup, which is likely a powerful explanation
nephrotoxicity. In addition to innate drug toxicity, certain for the heterogeneous response to chemotherapeutic
host characteristics and renal handling of the drug in- agents (12–14). Gene polymorphisms in the renal cy-
crease renal injury. In general, one or more of these factors tochrome P450 enzyme system, which favor reduced
combine to increase risk for kidney injury (Table 1). metabolism and renal excretion, enhance nephrotoxic
Many cancers involve the kidneys either directly or risk. Other examples are loss of function mutations in
indirectly—heightening the risk for kidney injury apical secretory transporters and mutations in kinases

www.cjasn.org Vol 7 October, 2012 Copyright © 2012 by the American Society of Nephrology 1713
1714 Clinical Journal of the American Society of Nephrology

of drugs by several enzyme systems present in the kidney


Table 1. Risk factors for chemotherapy-induced favors toxic metabolite and reactive oxygen species formation.
nephrotoxicity Renal injury may occur, because drug byproducts cause harm
through lipid peroxidation, protein damage, nucleic acid
Tumor-related kidney effects alkylation or oxidation, and DNA strand breaks (18–20).
direct renal involvement
myeloma-related kidney injury
renal infiltration (lymphoma and leukemia)
urinary obstruction Classification of Chemotherapy-Associated Renal
neoplasia-associated glomerulopathies Lesions
indirect renal involvement There are a number of ways that one can approach
true volume depletion (N/V, diarrhea, and classifying the kidney lesions caused by the various chemo-
overdiuresis) therapeutic agents. For example, one could categorize the
effective volume depletion (cardiomyopathy, chemotherapy-related kidney lesions based on the nephron
malignant ascites, and pleural effusions) sites primarily affected by the drug. Agents that injure the
metabolic effects (hyperuricemia and hypercalcemia) renal vasculature, glomerulus, proximal and distal tubular
Innate drug toxicity
segments, and collecting ducts are described (Table 2), recog-
high-dose drug exposure and prolonged course of
therapy nizing that all nephrotoxic drugs cannot possibly be covered.
insoluble drug or metabolite form crystals within
intratubular lumens
potent direct nephrotoxic effects of the drug or toxin Renal Vasculature: Thrombotic Microangiopathy
drug combinations enhance nephrotoxicity Chemotherapeutic agents, such as bevacizumab and
NSAIDs, aminoglycosides, and radiocontrast gemcitabine, can injure the renal vasculature and cause
Patient factors thrombotic microangiopathy (TMA). TMA presents clinically
older age as microangiopathic hemolytic anemia, thrombocytopenia,
underlying AKI or CKD hypertension, and AKI with hematuria and proteinuria,
immune response genes although renal-limited TMA does occur.
increased allergic reactions to drugs
pharmacogenetics favoring drug/toxin toxicity
gene mutations in hepatic and renal CYP450 Bevacizumab
enzyme systems Recognition that tumor growth was highly dependent on
gene mutations in transport proteins and renal pathologic angiogenesis induced by local production of
transporters vascular endothelial growth factor (VEGF) paved the way
Renal drug handling for the development of drugs targeting this pathway (21).
high blood (and drug) delivery rate to the kidneys This pathway was a logical point of attack to supplement
proximal tubular uptake of toxins other tumor-directed therapies—and turned out to be a ben-
apical tubular uptake by endocytosis
eficial addition to the therapeutic armamentarium. Although
or another pathway
basolateral tubular transport through OAT there are numerous drugs that target VEGF effects, the anti-
and OCT pathways VEGF antibody bevacizumab will be the focus of discussion,
relatively hypoxic renal environment recognizing that there are differences among the agents.
high metabolic rate of tubular cells in the loop of Henle VEGF importantly regulates vasculogenesis and angio-
increased drug/toxin concentration in renal medulla genesis during development and in disease through reg-
and interstitium ulation of vascular permeability, endothelial cell migration,
biotransformation of substances to ROS causing proliferation, and survival (21). This regulation raises the
oxidative stress possibility that these drugs may be associated with ad-
verse effects. In fact, this finding is the case, because a
N/V, nausea/vomiting; NSAIDs, nonsteroidal anti- number of adverse systemic end-organ effects have been
inflammatory drugs; OAT, organic anion transporter; OCT, described, including kidney injury. This finding is not sur-
organic cation transporters; ROS, reactive oxygen species.
prising, because VEGF is produced by renal visceral epi-
thelial cells and binds to VEGF receptors located on
glomerular endothelium and mesangium, as well as peri-
that regulate drug carrier proteins, which can impair drug tubular capillaries (21). Local VEGF production maintains
excretion and induce nephrotoxicity by increasing intracel- normal functioning of all of these cells, including injury
lular drug concentrations (13,15). repair and cell turnover. Importantly, there is crosstalk
Last, the renal handling of drugs is another risk factor for between the glomerular endothelium and epithelium,
the development of nephrotoxicity. The kidney is exposed maintaining the integrity of the filtration barrier.
to considerable drug concentrations based on the high renal The most important renal effects described with anti-
blood flow rate—approximately 25% of cardiac output. Sig- angiogenesis therapy are new or worsened hypertension
nificant drug uptake occurs in the proximal tubular cells and kidney-specific injury, including proteinuria and AKI.
through both apical uptake and basolateral transport (16,17). Importantly, the development of hypertension in patients
Trafficking of these agents through tubular cells explains, in predicts a better tumor response to therapy (22), and it
part, their nephrotoxicity. The high metabolic rate and hyp- should prompt clinicians to continue therapy and control
oxic environment of loop of Henle and medullary collecting BP with antihypertensive agents rather than discontinuing
duct cells impart nephrotoxic drug risk (18,19). Metabolism antiangiogenic therapy. Animal experiments documented a
Clin J Am Soc Nephrol 7: 1713–1721, October, 2012 Nephrotoxicity of Chemotherapy, Perazella 1715

Table 2. Kidney injury associated with chemotherapeutic


agents

Renal vasculature
hemodynamic AKI (capillary leak syndrome)
IL-2, denileukin diftitox
thrombotic microangiopathy
antiangiogenesis drugs (bevacizumab and tyrosine
kinase inhibitors)
gemcitabine and cisplatin
mitomycin C and IFN
Glomeruli
minimal change disease
IFN
pamidronate
focal segmental glomerulosclerosis
IFN
Figure 1. | A glomerulus exhibits mesangiolysis, endothelial de-
pamidronate
nudation, red blood cell congestion, and glomerular basement mem-
zoledronate (rare)
brane duplication in this example of thrombotic microangiopathy.
Tubulointerstitium
(Jones methenamine silver stain; original magnification, 3600.) Cour-
acute tubular necrosis
tesy of Glen S. Markowitz.
platinums, zoledronate, ifosfamide, and mithramycin
pentostatin, imatinib, diaziquone, and pemetrexed
tubulopathies
Fanconi syndrome Gemcitabine
cisplatin, ifosfamide, and azacitadine, Gemcitabine is a cell cycle–specific pyrimidine antago-
diaziquone, imatinib, and pemetrexed nist that is an effective therapy for certain malignancies,
salt wasting primarily carcinomas of the lung, pancreas, bladder, and
cisplatin and azacitadine breast. Unfortunately, as with other chemotherapeutic
magnesium wasting agents, it is complicated by kidney injury. Numerous
cisplatin, cetuximab, and panitumumab case reports and case series have documented AKI from
nephrogenic diabetes insipidus gemcitabine, primarily from TMA. In addition, hyperten-
cisplatin, ifosfamide, and pemetrexed sion, microangiopathic hemolytic anemia, and ischemic
syndrome of inappropriate antidiuresis
skin lesion may be present. A recent case series of 29 pa-
cyclophosphamide and vincristine
acute interstitial nephritis tients treated with gemcitabine described the various clin-
sorafenib and sunitinib ical renal manifestations (26). All patients developed AKI;
crystal nephropathy TMA was seen in four patients who underwent kidney
methotrexate biopsy. New or worsening hypertension occurred in 26
of 29 patients, whereas edema (21/29) and congestive
heart failure (7/29) also complicated gemcitabine therapy.
Classic systemic TMA occurred in all patients and was
two- to threefold increase in proteinuria in mice injected manifested by anemia, thrombocytopenia, and increased
with a single dose of anti-VEGF antibody (23). Renal his- lactate dehydrogenase levels. Suppressed haptoglobin lev-
topathology revealed glomerular endothelial cell swelling, els (23/26) and schistocytes on peripheral smear (21/24)
vacuolization, and detachment, as well as disruption of ep- were also noted. Urinalysis revealed hematuria/proteinuria
ithelial cell slit diaphragms. Immunohistochemistry also (27/29) and red blood cell casts (n58).
showed downregulation of nephrin, which was partially Gemcitabine-associated TMA is relatively rare, with the
restored with administration of recombinant VEGF. The major risk factors being previous therapy with mitomycin-C
renal effects of bevacizumab therapy in six patients with and total drug dose. Often, it is impossible to predict who will
various malignancies were described in detail (24). Renal develop this complication, although new or worsened hy-
findings developed within 3–17 months of drug exposure: pertension may precede other clinical manifestations of TMA.
proteinuria occurred in all patients, with five patients hav- Unfortunately, therapy is generally limited and mainly sup-
ing at least 1 g/d and two patients having nephrotic-level portive, consisting of drug discontinuation, antihypertensive
proteinuria. Hypertension and AKI developed in 50% of medications, and dialysis when indicated. Plasmapheresis has
patients, and all had TMA on kidney histology (Figure 1). been used with minimal or no success. Renal outcomes are
Importantly, proteinuria, hypertension, and AKI generally highly variable. In the largest cases series (26), 19 patients had
improved on withdrawal of bevacizumab. full or partial renal recovery, whereas 3 patients developed
Although a number of renal lesions have been described CKD and 7 patients had dialysis-requiring ESRD.
with the antiangiogenesis drugs, the predominant histo-
pathology is TMA. Other lesions described on kidney
biopsy include focal segmental glomerulosclerosis (FSGS), Glomerulus: Podocytopathy
mebranoproliferative GN, glomerular endotheliosis, IFN
cryoglobulinemic GN, nonspecific immune complex GN, IFN is a glycoprotein synthesized and released by
and acute interstitial nephritis (21,24,25). leukocytes, fibroblasts, T cells, and natural killer cells in
1716 Clinical Journal of the American Society of Nephrology

response to pathogens, such as viruses, parasites, and bac- present in 12 of 13 patients, with proteinuria ranging
teria, as well as tumor cells. It is a protective defense that from 1.9 to 27 g/d. AKI occurred in 11 patients. Urine
allows communication between cells to eradicate infection sediment was bland in nearly one-half of the patients,
or malignant cells. In general, IFN-a and -b reduce viral whereas red and white blood cells were seen in five and
replication and protein synthesis in neighboring cells, one patients, respectively. Remission was inconsistent and
whereas IFN-g activates macrophage and MHC expression incomplete in most patients after IFN discontinuation.
(27,28). Based on these characteristics, exogenous IFN has a With follow-up in 10 patients, which ranged from 2 to
number of therapeutic uses. The most commonly used 54 months, complete remission was noted in 1 patient
agent is IFN-a, which is used to treat hepatitis C and B and partial remission was noted in 3 patients, whereas
viruses and various malignancies. IFN-b is used to treat some improvement in proteinuria and kidney function
multiple sclerosis, whereas IFN-g was studied as a treatment was described in 5 patients. Of these 10 patients, 8 patients
for chronic granulomatous disease but has been abandoned. received steroids, and 1 patient received cyclophospha-
Chronic IFN therapy is associated with clinical renal disease, mide. Thus, although IFN discontinuation sometimes
which seems to be associated primarily with podocyte injury helps, it is not always associated with remission. Steroids
(27,28). Based on published cases, minimal change disease seem unhelpful, especially in FSGS.
has been described with both IFN-a (n56) and -b (n52). The The mechanism of podocyte injury with IFN is incom-
lesion developed anywhere from 5 days to 22 months after pletely understood. A number of putative mechanisms
therapy (27). Nephrotic syndrome with urinary protein lev- have been put forward (27). A direct IFN effect on the
els of 2.3–28 g/d occurred in all patients, whereas AKI com- podocyte is possible through receptor binding and activa-
plicated the course of two patients. In follow-up ranging tion that promotes two potential injurious effects: (1) al-
from 53 days to 12 months, complete remission was noted tered cellular proliferation and metabolism of the
in all patients, with discontinuation of IFN and steroid ther- pododyte and (2) increased podocyte oxidative capacity
apy in three patients (27,28). and increased MHC class II antigen expression. Indirect
FSGS constitutes another form of podocytopathy that can IFN effects on the podocyte may also contribute to the
complicate IFN therapy. The lesion FSGS-not otherwise development of FSGS. IFNs activate adaptive immune
specified has been described in 10 patients treated with IFN mechanisms that increase macrophage activation—an ex-
therapy (IFN-a in 9 patients and -g in 1 patient) (27,28). In ample is hemophagocytic syndrome, which is associated
these cases, IFN therapy ranged from 19 days to 20 months with collapsing FSGS. Viral diseases such as HIV and par-
and was associated with nephrotic syndrome in all 10 patients, vovirus B19, which increase IFN production, are also as-
with proteinuria of 6.3–42 g/d and AKI in 8 patients. With sociated with collapsing FSGS. Finally, IFN may enhance
follow-up ranging from 1 to 16 months in 9 of 10 patients, synthesis of pathogenic cytokines such as IL-6 and -13,
complete or partial remission was noted in 4 of 9 patients which are permeability factors in FSGS and minimal
with discontinuation of IFN. Six patients received steroids, change disease.
of which only two patients benefited with remission.
Collapsing FSGS (Figure 2) also complicates IFN ther-
apy. In 14 patients with this lesion, IFN-a was the causa- Tubules: Acute Tubular Injury
tive agent in 9 patients, whereas IFN-b and -g therapy was Cisplatin
used in 3 and 2 patients, respectively (27,28). IFN exposure Cisplatin is a platinum compound that is an effective
ranged from 1 to 48 months; nephrotic syndrome was therapy for many carcinomas and sarcomas, as well as
lymphomas. Its major adverse effect is nephrotoxicity,
although ototoxicity also occurs. Both are dose-related
toxicities, causing apoptosis and necrosis of cells (29–32).
Cisplatin injures multiple renal compartments, including
blood vessels, glomeruli, and most commonly, the tubules
(29). Nephrotoxicity is generally reversible, but it can be
permanent. Tubular injury as manifested by AKI and tu-
bular dysfunction syndromes will be described.
Cisplatin’s mechanism of nephrotoxicity is related to its
drug characteristics, its renal handling, and the kidney re-
sponse to the cisplatin molecule (29–32). Chloride at the
cis-position of the molecule is one such factor that pro-
motes kidney injury, whereas the pathway of excretion
through the cell (in through organic anion transporter 1
and out through efflux transporters) potentially increases
intracellular concentrations (Figure 3). After it is inside the
tubular cell, a number of intracellular injury pathways oc-
cur. These pathways include caspase activation, cyclin-
Figure 2. | A glomerulus exhibits global wrinkling and retraction of
the glomerular basement membrane and diffuse swelling and hy- dependent kinases, mitogen-activated protein kinase
perplasia of overlying visceral epithelial cells in this example of activation, and p53 signaling. In addition, cellular injury
collapsing focal segmental glomerulosclerosis. (Jones methenamine also develops from inflammation and oxidative stress,
silver stain; original magnification, 3600.) Courtesy of Glen S. whereas vascular injury and decreased GFR with ischemic
Markowitz. injury also occur (29–32). These pathways of injury result
Clin J Am Soc Nephrol 7: 1713–1721, October, 2012 Nephrotoxicity of Chemotherapy, Perazella 1717

Figure 3. | Cisplatin (Cis) nephrotoxicity is, in part, related to its uptake by proximal tubular cells. Cis enters cells through organic cation
transporters (OCTs), and when it accumulates within cells, it causes cell injury through multiple mechanisms. Apoptosis and necrosis of tubular
cells result and cause clinical AKI and tubulopathy. CDKs, cyclin-dependent kinases; MAPK, mitogen-activated protein kinase; MRP, multidrug-
resistant protein; NaDC, sodium dicarboxylate; OAT, organic anion transporter; P53, protein 53; Pgp, P glycoprotein; ROS, reactive oxygen
species.

in renal tubular cell apoptosis and necrosis—resulting in nucleophilic sulfur thiols, neurotrophins, phosphonic acid,
clinical AKI and/or a tubulopathy. melanocortins, and free oxygen radical scavengers, but their
Tubular injury without AKI also complicates cisplatin utility is unclear. In high-risk patients, other platinums such
therapy and manifests as a number of tubulopathies, which as carboplatin and oxalaplatin are used based on their less
include isolated proximal tubulopathy (proteinuria and nephrotoxic profile compared with cisplatin. Two potential
phosphate wasting) or full-blown Fanconi syndrome (2,29). explanations for reduced nephrotoxicity exist. Neither of
Sodium wasting is another consequence of cisplatin therapy, these molecules is transported by organic cation transporter
which can be associated with hypovolemia, orthostasis, 2 (OCT-2), thereby reducing proximal tubular intracellular
and prerenal AKI. In the distal nephron, cisplatin can im- concentrations (29–32). In addition, the chloride at cis-position
pair reabsorption of magnesium, causing refractory hypo- in cisplatin is replaced by carboxylate and cyclobutane in car-
magnesemia. Finally, water absorption in the collecting boplatin and oxalaplatin, respectively, which may further re-
duct can be disturbed, resulting in a form of nephrogenic duce toxicity (29–32).
diabetes insipidus. Treatment of toxic renal manifestations is primarily
AKI is a dose-related complication of cisplatin—it can be a supportive (2,28). There are no effective therapies to reverse
functional decline in GFR or caused by TMA, but most AKI or tubular dysfunction. Dialysis is reserved for ad-
commonly, it is a result of acute tubular injury/necrosis. vanced AKI as manifested by uremia, metabolic disturban-
Although AKI can recover, renal outcomes such as pro- ces, and hypervolemia. There is no role for dialytic removal
gressive CKD from chronic tubulointerstitial fibrosis and of cisplatin. Maneuvers to correct hypovolemia from salt
irreversible chronic tubulopathies may result (2,29). wasting (intravenous normal saline or oral sodium chloride)
A focus of care for patients receiving cisplatin is pre- and address symptomatic hypomagnesemia with intrave-
vention of kidney injury (2,29). Forced diuresis with intra- nous and/or oral magnesium are required. Fanconi syn-
venous normal saline or hypertonic saline is used to drome is notoriously difficult to treat.
counteract the toxic effect of chloride on the cis-position
of the molecule. The addition of mannitol to induce a Ifosfamide
forced diuresis is sometimes used, but evidence of benefit Ifosfamide is an alkylating agent used for certain cancers,
is lacking; in some cases, this approach may cause wors- including sarcomas, testicular cancer, and some forms of
ened kidney function (33). Amifostine is a glutathione an- lymphoma. Ifosfamide’s major adverse effect is kidney in-
alog taken up by normal cells that blunts cisplatin-induced jury. Nephrotoxic manifestations include tubulopathies
cellular injury. It is, however, complicated by nausea and such as proximal tubular injury or Fanconi syndrome
vomiting. A number of other agents have been used to and nephrogenic diabetes insipidus; additionally, AKI is often
reduce nephrotoxicity, such as sodium thiosulfate, reversible but can be permanent (2,34,35). Histopathology
1718 Clinical Journal of the American Society of Nephrology

reveals features of tubular cell injury/necrosis with swollen, mesothelioma and non-small cell lung cancer. This agent is
dysmorphic mitochondria. excreted unchanged by the kidneys (70%–90% in 24 hours),
The difference in adverse effects for ifosfamide (neph- with a half-life of 3.5 hours (36). Its renal handling may
rotoxicity) and cyclophosphamide (hemorrhagic cystitis), explain some of the drug’s nephrotoxicity. It is postulated
which are related compounds, is caused by the major toxic that pemetrexed enters proximal tubular cells through two
metabolite that they produce. Acrolein produced by cyclo- separate pathways. One cell entry site is the basolateral
phosphamide is non-nephrotoxic, whereas chloracetaldehyde membrane, where pemetrexed is transported through re-
produced by ifosfamide injures kidney tissue. At equivalent duced folate carrier, whereas the other is apical drug uptake
doses, ifosfamide produces 40 times more chloroacetaldehyde through the folate receptor-a transport pathway (Figure 4).
than cyclophosphamide (2,34,35). Furthermore, ifosfamide After inside the cell, pemetrexed is polyglutamylated with
enters proximal tubular cells through OCT2, whereas cy- two resulting effects. First, with polyglutamylation, the
clophosphamide does not (2,35). Risk factors for adverse drug can no longer be transported out of the cell, increasing
renal effects include previous cisplatin exposure, cumulative intracellular concentrations. Second, the higher drug con-
dose .90 g/m2, and underlying CKD. centrations more fully inhibit folate metabolism enzymes
Preventive measures are limited for ifosfamide. Mesna, and impair cellular RNA/DNA synthesis.
which is effective for hemorrhagic cystitis, is of limited Although its major adverse effects are myelosuppression
value for ifosfamide-induced kidney injury. Dose reduction and neutropenia, reversible AKI has been noted with high-
helps but also limits the efficacy of tumor killing. Because dose therapy (600 mg/m2). Pemetrexed nephrotoxicity has
this agent is transported into cells through OCT2, compet- been described in several case reports: acute tubular necrosis
itive inhibition of this pathway with cimetidine is being (n55), acute interstitial nephritis (n52), nephrogenic diabe-
evaluated (35). Treatment, as with many of these agents, is tes insipidus/renal tubular acidosis (n51), and nephrogenic
supportive. Attention to supplementing electrolyte defi- diabetes insipidus (n51) (36). A decline in CrCl from 88 to
ciencies, monitoring for progressive CKD, and dialysis as 77 ml/min was reported after four cycles of pemetrexed in a
indicated are important. In addition to CKD and ESRD, mesothelioma trial (36). Most patients present with AKI and
long-term complications include a permanent proximal tu- minimal proteinuria, which stabilizes with drug discontinu-
bulopathy (1%) and isolated renal phosphaturia in up to ation but can lead to permanent CKD. Renal histology re-
20%. This latter complication may cause osteomalacia or veals primarily chronic tubulointerstitial fibrosis and tubular
growth problems in children and exacerbate osteoporosis atrophy, consistent with a toxic tubular injury (36).
in the elderly (2,34,35).

Pemetrexed Tubules: Magnesium Wasting


Pemetrexed is an antifolate agent that inhibits enzymes Cetuximab
involved in purine/pyrimidine metabolism, thereby im- Cetuximab is a chimeric monoclonal antibody against EGF
pairing RNA/DNA synthesis in tumors such as malignant receptor (EGFR) used to treat various epithelial malignancies,

Figure 4. | Pemetrexed (Pmx) nephrotoxicity may result from proximal tubular cell uptake of drug through apical (folate receptor-a [FR-a])
and basolateral (reduced folate carrier [RFC]) transporters. After it is inside the cell, Pmx is polyglutamylated, which inhibits drug transport
out of the cell and enhances drug inhibition of folate metabolism. Impaired synthesis of RNA and DNA by cells causes clinical AKI and
tubulopathy. Pmx-glt, polyglutamylated pemetrexed.
Clin J Am Soc Nephrol 7: 1713–1721, October, 2012 Nephrotoxicity of Chemotherapy, Perazella 1719

including colorectal, head/neck, breast, and lung cancers. Treatment of hypomagnesemia requires intravenous
EGF overexpression present in these malignancies reduces repletion, because oral magnesium is ineffective and often
apoptosis and enhances tumor cell growth. Cetuximab has complicated by diarrhea. Along with magnesium sup-
a 10-fold greater affinity for EGFR than natural ligand, making plementation, calcium and potassium repletion are also
it an effective targeted therapy in these cancers. However, one commonly required. In general, renal magnesium wasting
of the drug’s adverse effects is hypomagnesemia (37–41). improves and eventually resolves approximately 4–6 weeks
Cetuximab-induced hypomagnesemia results from a renal after discontinuation of cetuximab.
leak of magnesium. Magnesium reabsorption in the distal
convoluted tubule is, in part, dependent on EGF binding its
receptor on the basolateral membrane (37,38,41). Activation Tubules: Crystal Nephropathy
of the EGFR sets in motion intracellular signals that stimu- Methotrexate
late the movement of the cation channel transient receptor The dihydrofolate reductase inhibitor methotrexate is
potential M6 into the apical membrane, which facilitates the widely used, especially in high dose, to treat malignancies
reabsorption of magnesium from the urinary space into the such as high-grade lymphomas. Nephrotoxicity is a known
cell (37,38,41). Cetuximab competitively inhibits EGF bind- complication of high-dose therapy (1–12 g/m2) but rarely
ing to its receptor (Figure 5), thereby blunting the placement occurs with long-term conventional dosing (1,2). The in-
of transient receptor potential M6 into the apical membrane cidence is highly variable depending on the patient’s risk
and causing renal magnesium wasting (37,38,41). factors and the appropriate employment of preventive
The incidence of hypomagnesemia with cetuximab in measures, such as intravenous fluids (1,2,43).
initial colorectal cancer trials was only 1.8%–5.8% (37,38). Methotrexate and its major metabolite, 7-OH metho-
However, a higher incidence was noted when magnesium trexate, are filtered by the glomerulus and secreted into
levels were measured more rigorously. More than one-half the urinary space by proximal tubules. AKI is primarily
of patients develop hypomagnesemia with cetuximab, and the result of acute tubular injury from precipitation of
nearly 100% of patients have some decline in serum mag- methotrexate/7-OH methotrexate in distal tubular lumens
nesium concentrations (37,38,41). In fact, a meta-analysis (2,43). However, tubular apoptosis/necrosis may also de-
of randomized controlled trials of cetuximab versus other velop from oxygen radicals associated with decreased
therapies showed an odds ratio of 4.7 for all-grade hypo- adenosine deaminase activity (44). AKI incidence, when
magnesemia and 5.3 for grade 3/4 hypomagnesemia (42). defined as grade .2 nephrotoxicity, ranges from 1.8% to
In addition, hypokalemia and hypocalcemia occur with 12% (2,43).
severe hypomagnesemia. Risk factors for hypomagnesemia Risk factors for nephrotoxicity include intravascular
include duration of cetuximab therapy, older age, and base- volume depletion with sluggish urinary flow, acid urine
line magnesium concentration. Panitumumab is also com- pH, and underlying kidney disease (GFR,60 ml/min)
plicated by hypomagnesemia (36%) but of less severe (2,42). Based on these factors, prevention is focused on
grade, because only 3% developed grade 3/4 hypomag- volume repletion before/during drug infusion, appropri-
nesemia (1,2). ate drug dosing, and alkalinization of the urine (pH.7.1).

Figure 5. | Cetuximab (C) is an EGF receptor (EGFR) antibody that causes renal magnesium wasting by competing with EGF for its receptor.
Normally, EGF binds its receptor (EGFR) and stimulates magnesium reabsorption in the distal convoluted cell. EGFR activation is associated
with magnesium absorption through transient receptor potential M6 (TRPM6) in the apical membrane. NCC, sodium chloride cotransporter.
1720 Clinical Journal of the American Society of Nephrology

Treatment is comprised of leucovorin rescue at 24–36 15. Lang F: Regulating renal drug elimination? J Am Soc Nephrol 16:
hours of methotrexate therapy to reduce nonmalignant 1535–1536, 2005
16. Enomoto A, Endou H: Roles of organic anion transporters (OATs)
cell injury (2,43,45). Glucarbidase cleaves methotrexate to and a urate transporter (URAT1) in the pathophysiology of human
noncytotoxic metabolites. It is reserved for use when meth- disease. Clin Exp Nephrol 9: 195–205, 2005
otrexate levels are toxic, and there is significant risk for 17. Ciarimboli G, Ludwig T, Lang D, Pavenstädt H, Koepsell H,
systemic toxicity (45). High-flux hemodialysis clears the Piechota HJ, Haier J, Jaehde U, Zisowsky J, Schlatter E: Cisplatin
plasma of methotrexate fairly well (76%) but is associated nephrotoxicity is critically mediated via the human organic
cation transporter 2. Am J Pathol 167: 1477–1484, 2005
with immediate postdialysis plasma rebound (1,2). It may 18. Cummings BS, Schnellmann RG: Pathophysiology of nephro-
have a role when severe AKI is present, but it may become toxic cell injury. In: Diseases of the Kidney and Urogenital Tract,
unnecessary with the availability of glucarbidase. edited by Schrier RW, Philadelphia, PA, Lippincott Williams &
Wilkinson, 2001, pp 1071–1136
19. Kaloyanides GJ, Bosmans J-L, DeBroe ME: Antibiotic and
immunosuppression-related renal failure. In: Diseases of the
Conclusion Kidney and Urogenital Tract, edited by Schrier RW, Philadelphia,
Chemotherapeutic agents have improved cancer patient PA, Lippincott Williams & Wilkinson, 2001, pp 1137–1174
survival; however, nephrotoxicity remains an important 20. Aleksa K, Matsell D, Krausz K, Gelboin H, Ito S, Koren G: Cyto-
complication. A number of patient- and drug-related fac- chrome P450 3A and 2B6 in the developing kidney: Implications
for ifosfamide nephrotoxicity. Pediatr Nephrol 20: 872–885, 2005
tors increase risk for adverse renal events; some events are 21. Gurevich F, Perazella MA: Renal effects of anti-angiogenesis
modifiable, and others are not. Clinicians must be familiar therapy: Update for the internist. Am J Med 122: 322–328,
with the nephrotoxicity of these drugs, particularly the as- 2009
sociated clinical and laboratory manifestations. Preventive 22. De Stefano A, Carlomagno C, Pepe S, Bianco R, De Placido S:
measures should be used when possible, as well as support- Bevacizumab-related arterial hypertension as a predictive
marker in metastatic colorectal cancer patients. Cancer
ive care and available therapies. Some patients will be left Chemother Pharmacol 68: 1207–1213, 2011
with long-term complications such as chronic tubulopathies 23. Sugimoto H, Hamano Y, Charytan D, Cosgrove D, Kieran M,
and CKD. Onco-Nephrology is a rapidly expanding area that Sudhakar A, Kalluri R: Neutralization of circulating vascular
requires a close working relationship between oncologists endothelial growth factor (VEGF) by anti-VEGF antibodies and
and nephrologists. soluble VEGF receptor 1 (sFlt-1) induces proteinuria. J Biol Chem
278: 12605–12608, 2003
24. Eremina V, Jefferson JA, Kowalewska J, Hochster H, Haas M,
Disclosures Weisstuch J, Richardson C, Kopp JB, Kabir MG, Backx PH,
None. Gerber HP, Ferrara N, Barisoni L, Alpers CE, Quaggin SE: VEGF
inhibition and renal thrombotic microangiopathy. N Engl J Med
358: 1129–1136, 2008
References 25. Izzedine H, Rixe O, Billemont B, Baumelou A, Deray G: An-
1. Sahni V, Choudhury D, Ahmed Z: Chemotherapy-associated re- giogenesis inhibitor therapies: Focus on kidney toxicity and hy-
nal dysfunction. Nat Rev Nephrol 5: 450–462, 2009 pertension. Am J Kidney Dis 50: 203–218, 2007
2. Perazella MA, Moeckel GW: Nephrotoxicity from chemothera- 26. Glezerman IG, Kris MG, Miller V, Seshan S, Flombaum CD:
peutic agents: Clinical manifestations, pathobiology, and Gemcitabine nephrotoxicity and hemolytic uremic syndrome:
prevention/therapy. Semin Nephrol 30: 570–581, 2010 Report of 29 cases from a single institution. Clin Nephrol 71:
3. Finkel KW, Foringer JR: Renal disease in patients with cancer. Nat 130–139, 2009
Clin Pract Nephrol 3: 669–678, 2007 27. Markowitz GS, Nasr SH, Stokes MB, D’Agati VD: Treatment with
4. Lameire NH, Flombaum CD, Moreau D, Ronco C: Acute renal IFN-alpha, -beta, or -gamma is associated with collapsing focal
failure in cancer patients. Ann Med 37: 13–25, 2005 segmental glomerulosclerosis. Clin J Am Soc Nephrol 5: 607–
5. de Jonge MJA, Verweij J: Renal toxicities of chemotherapy. Semin 615, 2010
Oncol 33: 68–73, 2006 28. Colovic M, Jurisic V, Jankovic G, Jovanovic D, Nikolic LJ,
6. Humphreys BD, Soiffer RJ, Magee CC: Renal failure associated Dimitrijevic J: Interferon alpha sensitisation induced fatal renal
with cancer and its treatment: An update. J Am Soc Nephrol insufficiency in a patient with chronic myeloid leukaemia: Case
16: 151–161, 2005 report and review of literature. J Clin Pathol 59: 879–881, 2006
7. Evenepoel P: Acute toxic renal failure. Best Pract Res Clin An- 29. Pabla N, Dong Z: Cisplatin nephrotoxicity: Mechanisms and
aesthesiol 18: 37–52, 2004 renoprotective strategies. Kidney Int 73: 994–1007, 2008
8. Singh NP, Ganguli A, Prakash A: Drug-induced kidney diseases. 30. Kawai Y, Nakao T, Kunimura N, Kohda Y, Gemba M: Relationship
J Assoc Physicians India 51: 970–979, 2003 of intracellular calcium and oxygen radicals to Cisplatin-related
9. Guo X, Nzerue C: How to prevent, recognize, and treat drug- renal cell injury. J Pharmacol Sci 100: 65–72, 2006
induced nephrotoxicity. Cleve Clin J Med 69: 289–297, 2002 31. Faubel S, Ljubanovic D, Reznikov L, Somerset H, Dinarello CA,
10. Jerki
c M, Vojvodic S, López-Novoa JM: The mechanism of in- Edelstein CL: Caspase-1-deficient mice are protected against
creased renal susceptibility to toxic substances in the elderly. Part I. cisplatin-induced apoptosis and acute tubular necrosis. Kidney
The role of increased vasoconstriction. Int Urol Nephrol 32: 539– Int 66: 2202–2213, 2004
547, 2001 32. Ramesh G, Reeves WB: TNFR2-mediated apoptosis and necrosis
11. Perazella MA: Renal vulnerability to drug toxicity. Clin J Am Soc in cisplatin-induced acute renal failure. Am J Physiol Renal
Nephrol 4: 1275–1283, 2009 Physiol 285: F610–F618, 2003
12. Harty L, Johnson K, Power A: Race and ethnicity in the era of 33. Morgan KP, Buie LW, Savage SW: The role of mannitol as a
emerging pharmacogenomics. J Clin Pharmacol 46: 405–407, nephroprotectant in patients receiving cisplatin therapy. Ann
2006 Pharmacother 46: 276–281, 2012
13. Ciarimboli G, Koepsell H, Iordanova M, Gorboulev V, Dürner B, 34. Zamlauski-Tucker MJ, Morris ME, Springate JE: Ifosfamide metab-
Lang D, Edemir B, Schröter R, Van Le T, Schlatter E: Individual olite chloroacetaldehyde causes Fanconi syndrome in the perfused
PKC-phosphorylation sites in organic cation transporter 1 de- rat kidney. Toxicol Appl Pharmacol 129: 170–175, 1994
termine substrate selectivity and transport regulation. J Am Soc 35. Ciarimboli G, Holle SK, Vollenbröcker B, Hagos Y, Reuter S,
Nephrol 16: 1562–1570, 2005 Burckhardt G, Bierer S, Herrmann E, Pavenstädt H, Rossi R, Kleta
14. Ulrich CM, Bigler J, Potter JD: Non-steroidal anti-inflammatory R, Schlatter E: New clues for nephrotoxicity induced by ifosfamide:
drugs for cancer prevention: Promise, perils and pharmacogenetics. Preferential renal uptake via the human organic cation trans-
Nat Rev Cancer 6: 130–140, 2006 porter 2. Mol Pharm 8: 270–279, 2011
Clin J Am Soc Nephrol 7: 1713–1721, October, 2012 Nephrotoxicity of Chemotherapy, Perazella 1721

36. Glezerman IG, Pietanza MC, Miller V, Seshan SV: Kidney tubular 42. Cao Y, Liao C, Tan A, Liu L, Gao F: Meta-analysis of incidence and
toxicity of maintenance pemetrexed therapy. Am J Kidney Dis 58: risk of hypomagnesemia with cetuximab for advanced cancer.
817–820, 2011 Chemotherapy 56: 459–465, 2010
37. Glaudemans B, Knoers NV, Hoenderop JG, Bindels RJ: New 43. Perazella MA: Crystal-induced acute renal failure. Am J Med 106:
molecular players facilitating Mg(21) reabsorption in the distal 459–465, 1999
convoluted tubule. Kidney Int 77: 17–22, 2010 44. Pinheiro FV, Pimentel VC, De Bona KS, Scola G, Salvador M,
38. Schrag D, Chung KY, Flombaum C, Saltz L: Cetuximab therapy Funchal C, Moretto MB: Decrease of adenosine deaminase ac-
and symptomatic hypomagnesemia. J Natl Cancer Inst 97: 1221– tivity and increase of the lipid peroxidation after acute metho-
1224, 2005 trexate treatment in young rats: protective effects of grape seed
39. Fakih MG, Wilding G, Lombardo J: Cetuximab-induced hypo- extract. Cell Biochem Funct 28: 89–94, 2010
magnesemia in patients with colorectal cancer. Clin Colorectal 45. Patterson DM, Lee SM: Glucarbidase following high-dose
Cancer. 6: 152–156, 2006 methotrexate: Update on development. Expert Opin Biol Ther
40. Saif MW: Management of hypomagnesemia in cancer patients 10: 105–111, 2010
receiving chemotherapy. J Support Oncol 6: 243–248, 2008
41. Dietrich A, Chubanov V, Gudermann T: Renal TRPathies. J Am Published online ahead of print. Publication date available at www.
Soc Nephrol 21: 736–744, 2010 cjasn.org.

S-ar putea să vă placă și