You are on page 1of 8

Tubulointerstitial Injury Associated With

Chemotherapeutic Agents
Anushree C. Shirali and Mark A. Perazella
Chemotherapy holds tremendous potential in improving the survival of patients with cancer. However, the side effects of these
drugs, including those that affect the kidney, can adversely affect patient outcomes. Prompt recognition of these adverse kid-
ney effects allows early intervention that can minimize or prevent patient morbidity. In this review, we examine the nephrotoxic
potential of chemotherapy drugs. In concentrating on tubulointerstitial injury, we will review common agents that result in
acute kidney injury due to acute tubular necrosis, tubulopathies, crystal nephropathy, acute interstitial nephritis, and chronic
interstitial nephritis, and we will present preventive and management strategies.
Q 2014 by the National Kidney Foundation, Inc. All rights reserved.
Key Words: Chemotherapy, Acute kidney injury, Chronic kidney disease, Fanconi syndrome

Introduction dling marks it as a particular target for toxic drug effects.


Because it receives a significant percentage of cardiac out-
Anticancer therapies provide oncologists with opportuni-
put, drug delivery to the kidney is highly efficient. This is
ties to treat their patients and favorably affect disease
particularly true for renal tubular epithelial cells
progression and overall survival. Unfortunately, drug-
(RTECs), which receive drug at their basolateral surface
associated nephrotoxicity occurs commonly with some
via peritubular capillary blood flow. These agents then
of these agents. Clinically, patients may experience acute
enter tubular cells via uptake through organic anionic
kidney injury (AKI) and some of its complications,
transporters and organic cationic transporters (OCTs).1
including excessive chemotherapy drug levels due to
Excretion into urine occurs when drugs are secreted
kidney underexcretion, increased hospitalization rates
into the tubular lumen via apical efflux transporters.
and longer lengths of stay, and increased morbidity and
Other drugs are filtered at the glomerulus and then
mortality. Recognizing the potential of adverse kidney
gain apical entry into tubular cells via endocytosis or pi-
events is important for all clinicians, but in particular
nocytosis.2 Alteration in tubular drug transport is 1 ave-
for the nephrologist who is asked to evaluate and treat
nue by which drug toxicity may occur in certain
new onset of kidney disease in the oncology patient.
individuals. For example, genetic polymorphisms that
This article will review the common clinical manifesta-
modulate the drug trafficking activity of these trans-
tions of chemotherapy-associated nephrotoxicity, focus-
porters may explain patient variability in cisplatin neph-
ing on those agents that result in tubulointerstitial injury.
rotoxicity.3 Once drugs are effluxed into tubular lumens
to join the glomerular filtrate, they become concentrated
Risk Factors for Nephrotoxicity of as water is reabsorbed in the descending loop of Henle.
Chemotherapeutics This allows a critical amount of drug to accumulate and
increases the potential for tubular cell injury. Tubular
Drug Factors and Kidney Handling of Drugs cells in the loop of Henle and medullary collecting duct
The propensity of chemotherapy to induce kidney dis- are particularly sensitive to this because their microenvi-
ease is related to drug- and patient-specific factors ronment is relatively hypoxic due to metabolic demands.
(Table 1). In fact, both factors often work in concert to en- These metabolic tasks include drug modification, for
hance the toxicity of any particular drug regimen. Drug- which the kidney contains multiple enzyme systems
specific factors refer to the inherent properties of the (including cytochrome p450) that oxidize parent com-
pharmacological compound. For example, the cytotoxic pounds into smaller and potentially toxic metabolites.4
effect of these drugs can affect malignant cells but also In addition, these enzymatic actions generate reactive ox-
healthy tissues, including kidney parenchymal cells. ygen species, leading to another plausible route of kidney
However, in several ways, the kidney’s role in drug han- injury.2
Chemical characteristics of the kidney microenviron-
From Section of Nephrology, Department of Internal Medicine, Yale Uni-
ment may also contribute to a specific drug’s toxic effects.
versity School of Medicine, New Haven, CT. For example, methotrexate (MTX) and its downstream
The authors report no financial conflicts of interest. metabolites require high pH for solubility. Normal hu-
Address correspondence to Anushree C. Shirali, MD, Department of Inter- man urine in a host with standard protein intake does
nal Medicine, Yale University School of Medicine, PO Box 208029, New Ha- not naturally attain high pH. As such, MTX and related
ven, CT 06520-8029. E-mail: anushree.shirali@yale.edu
Ó 2014 by the National Kidney Foundation, Inc. All rights reserved.
compounds are likely to remain precipitated within the
1548-5595/$36.00 renal tubules and cause direct cytotoxicity. Finally, it is
http://dx.doi.org/10.1053/j.ackd.2013.06.010 important to note that patients are often exposed to

56 Advances in Chronic Kidney Disease, Vol 21, No 1 (January), 2014: pp 56-63


Tubulointerstitial Nephrotoxicity of Chemotherapy 57

multiple potential nephrotoxic agents, which increases are associated with various types of kidney injury includ-
the susceptibility to kidney injury from any particular in- ing cast nephropathy, light- and heavy-chain deposition
sult. This is especially true of patients with malignancies, disease, and amyloidosis. Lastly, metabolic abnormalities
for whom use of these agents may be necessary for such as hypercalcemia, either in isolation or as part of the
treatment or diagnostic purposes (eg, nonsteroidal anti- tumor lysis syndrome, promote afferent arteriolar vaso-
inflammatory drugs prescribed for pain or osmolar con- constriction and a functionally prerenal state.
trast agents used for imaging).
Clinical and Pathologic Spectra of
Patient Factors Tubulointerstitial Disease
Along with kidney handling and drug-specific factors,
Overview
patient-specific characteristics also modify the risk for
nephrotoxicity. Some of these characteristics may be The spectrum of kidney disease from pharmaceutical
fixed, such as age or gender. The prevalence of cancer in- agents can be grouped into the customary prerenal, intra-
creases with aging, so older persons carry a higher bur- renal, and postrenal compartments. Although most drugs
den of malignant disease. Older persons also often have claim a singular mechanism of kidney injury, other drugs
more risk factors for AKI that make them more suscepti- may affect kidney function via several pathways. Chemo-
ble to chemotherapy-associated nephrotoxicity. These therapy drugs may induce prerenal injury (eg, with the
risk factors include (1) decreased muscle mass, which re- capillary leak syndrome tied to interleukin-2 administra-
sults in reduced serum creatinine and masks reduced tion). Postrenal disease is unusual, but it has been
glomerular filtration rate (GFR); (2) decreased total described with hemorrhagic cystitis from cyclophospha-
body water, which leads to mide leading to intravesicu-
increased serum drug con- lar thrombi and bladder
CLINICAL SUMMARY
centrations; and (3) de- outlet obstruction.6 How-
creased drug binding to ever, intrinsic kidney dis-
 Nephrotoxicity is commonly associated with some
protein in states of hypoal- chemotherapy drugs. ease is the most common
buminemia, which may be site of injury with chemo-
 Risk factors for nephrotoxicity are influenced by drug and
present in the patient with therapeutic drugs and is
patient characteristics.
cancer and contributes to in- further subdivided into
creased free drug concentra-  Nephrotoxicity is clinically manifested by acute kidney glomerular, tubular, and
injury, isolated tubulopathies, and CKD.
tions in the serum. Female interstitial pathology. Glo-
sex is also associated with  In general, nephrotoxicity can be avoided or ameliorated by merular disease from che-
reductions in total body wa- ensuring that patients are volume replete and not on motherapeutic agents is not
additional agents with nephrotoxic potential.
ter and muscle mass and common, but it may present
may be prone to drug over- as thrombotic microangio-
dosing for the same reasons. pathy, particularly with anti-
Finally, with decreased kid- angiogenesis agents and
ney reserve, any patient with CKD is at higher risk of gemcitabine.7,8 Other manifestations include focal seg-
AKI, regardless of age or gender. In summary, multiple mental glomerular sclerosis with all subclasses of inter-
factors may contribute to the kidney dysfunction asso- feron and pamidronate.9,10 Because further discussion
ciated with chemotherapy in patients with malignant of these glomerular pathologies is limited by space
diseases. considerations in the current article, we will focus on
The pathologic effects of cancer or side effects from injury to the tubular and interstitial compartments
the treatments of cancer also increase the risk of because it is the most likely to be associated with present-
chemotherapy-associated nephrotoxicity. These include day oncologic drugs. Five major pathologic states and their
vomiting and diarrhea, which increase propensity for kid- clinical presentations will be discussed: acute tubular ne-
ney injury through true volume depletion. Comorbid crosis (ATN), tubulopathies, crystal nephropathy, acute in-
conditions in the cancer patient such as congestive heart terstitial nephritis (AIN), and chronic interstitial nephritis
failure (from preexisting or drug-induced cardiomyopa- (Table 2). Representative examples of the most common
thy), malignant ascites, and sepsis reduce effective drugs in each category will be highlighted in the following
intravascular volume. Cholemic nephrosis, or jaundice- text; however, all drugs are listed in Table 2.
associated AKI, may result in the cancer patient with ob-
structive jaundice, putatively via kidney hypoperfusion ATN
5
and direct tubular toxicity from bile salts. Kidney disease Severe injury to the renal tubules results in cellular death
may also develop from malignancies such as leukemia and acute tubular injury and necrosis. Depending on the
and lymphoma via direct infiltration. Dysproteinemias particular drug, patients experience reduced GFR, usually
58 Shirali and Perazella

Table 1. Patient and Drug Characteristics That Increase Risk of Table 2. Chemotherapy Drugs Associated With Kidney Syndromes
Chemotherapy-Associated Nephrotoxicity of the Tubulointerstitium
Patient-Specific Factors Acute Tubular Necrosis
 Female gender  Cisplatin . carboplatin/oxalipatin
 Age . 65 y  Pemetrexed
 Nephrotic syndrome/hypoalbuminemia  Mithramycin
 Obstructive jaundice  Streptozocin
 Acute/chronic kidney disease  Pentostatin
B Cancer-related kidney disease  Zoledronate
B Other causes of kidney disease Tubulopathies, Fanconi syndrome
 True or effective volume depletion  Cisplatin
B Decreased GFR  Ifosfamide
B Decline in urine flow rates  Streptozocin
 Metabolic perturbations Salt wasting
B Urine pH  Cisplatin
 Immune response genes  Azacitidine
 Pharmacogenetics favoring drug toxicity Syndrome of inappropriate antidiuresis
B Polymorphisms in hepatic and kidney CYP450 system,  Cyclophosphamide
kidney transporters  Vincristine
Kidney-Specific Factors Nephrogenic diabetes insipidus
 High rate of blood delivery (20-25% of cardiac output)  Cisplatin
 Biotransformation of substances to reactive oxygen species  Ifosfamide
 High metabolic rate of tubular cells Magnesium wasting
 Proximal tubular uptake of toxins  Cetuximab, panitumumab
B Apical uptake via endocytosis/pinocytosis  Cisplatin
B Basolateral transport via OAT and OCT Crystal nephropathy
Drug-Specific Factors  Methotrexate
 Prolonged drug exposure Acute interstitial nephritis
 Direct drug or metabolite cytotoxicity of the drug or metab-  Anti-CTLA-4 antibody
olite Chronic interstitial nephritis
 Drug (or other nephrotoxin) combination that enhances  Nitrosureas
nephrotoxicity  Other drugs causing AKI
 Insoluble parent drug or metabolite with intratubular crystal
precipitation Abbreviations: AKI, acute kidney injury; CTLA-4, cytotoxic T-lym-
phocyte-associated antigen-4.
Abbreviations: CYP450, cytochrome p450 system; GFR, glomerular
filtration rate; OAT, organic anion transporter; OCT, organic cation chief mechanism of cisplatin-associated kidney injury ap-
transporter. pears to be oxidative damage, which occurs when a chlo-
ride ion on the parent drug is hydrolyzed and releases
hydroxyl radicals.12 Cisplatin causes injury primarily to
with AKI, although prolonged injury or delayed recovery
may also result in CKD. Tubular sodium reabsorption may
be compromised, thus leading to urine Na1 mEq/L
greater than 20 and fractional excretion of sodium
(FeNa) . 2%. Urine sediment may reveal RTECs and
RTEC casts and/or granular casts. With prolonged injury,
oliguric AKI may result, requiring renal replacement ther-
apy. In consultation with the oncologist, the offending
agent should be discontinued and kidney function must
be closely monitored. Several chemotherapy drugs
(Table 1) are implicated in causing ATN. In the following
sections, we will concentrate on 3 chemotherapeutic
agents that the nephrologist is likely to encounter because
they often cause renal tubular injury.
Cisplatin. Cisplatin claims undisputed tumoricidal ef-
ficacy against various solid organ tumors. However, it is Figure 1. Cisplatin-associated nephrotoxicity. Cisplatin
also potent in causing dose-dependent ATN in up to 20% causes kidney injury via multiple mechanisms. Uptake into
to 30% of patients receiving platinum therapy.11 AKI is proximal tubular cells is associated with toxicity via cell sig-
usually nonoliguric, but it may be severe enough to re- naling pathways, inflammation, and increased oxidative
stress. Vascular injury also causes tubular ischemia and
quire dialysis. Cisplatin can also result in proximal tubul- acute kidney injury. OCT-2, organic cation transporter-2;
opathy, which will be discussed later. As seen in Figure 1, RTEC, renal tubular epithelial cell; TNF-a, tumor necrosis
cisplatin may cause AKI through several pathways. The factor-a.
Tubulointerstitial Nephrotoxicity of Chemotherapy 59

the proximal tubule, partly because these cells uptake


drug via organic cationic transporter-2 (OCT-2).13 The
proximal tubular cells accumulate cisplatin, and intracel-
lular conversion to metabolites favors toxic injury via
several mechanisms, including oxidative stress
and proinflammatory cytokine production.12,14 Tumor
necrosis factor-a is a particularly potent inflammatory
mediator, playing a role in mediating cisplatin-induced
kidney injury through apoptosis.12 Cisplatin also acti-
vates signaling pathways, such as mitogen-activated pro-
tein kinase, and p53, which promote cell death.12 Finally,
cisplatin may also induce injury in the kidney vascula-
ture, leading to ischemic tubular cell death and AKI.
Other platinum-based agents, such as carboplatin and
oxaliplatin, are less nephrotoxic than cisplatin because
they lack chloride ions on their stem and display de-
creased OCT-2 cellular uptake.15,16 Nonetheless, in high
doses and in patients with the aforementioned risk
factors for nephrotoxicity, there is also a measurable
risk of ATN with these derivative platinum drugs. Figure 2. Ifosfamide-associated nephrotoxicity. Unlike cy-
Prevention of cisplatin nephrotoxicity is focused on clophosphamide, methotrexate is transported into proximal
volume repletion with saline and avoidance of other tubular cells via the OCT-2 on the basolateral membrane and
is metabolized to the tubular toxic substance chloracetalde-
nephrotoxic agents. Other prophylactic strategies to hyde. AKI, acute kidney injury; ATN, acute tubular necrosis;
reduce kidney injury include the glutathione analog OCT, organic cation transporter-2.
amifostine and sodium thiosulfate.2 However, these ther-
apies are not well tolerated (nausea and vomiting) and
potentially reduce tumor killing. Several other agents famide is less compared with cisplatin and correlates with
studied in animals have not made it into clinical practice cumulative lifetime drug exposure. In addition to total
(nucleophilic sulfur thiols, neurotrophins, phosphonic drug exposure of more than 100 g/m2, previous cisplatin
acid, melanocortins, and free oxygen radical scaven- use may be an independent risk factor.18 More common
gers).2 Treatment of cisplatin-induced ATN begins with than AKI, ifosfamide is associated with proximal tubular
cessation of further dosing. If electrolyte and acid-base dysfunction that may present as Fanconi syndrome (FS)
abnormalities are severe (hyperkalemia, metabolic acido- or as isolated renal tubular acidosis.19
sis), then a period of renal replacement therapy may be Pemetrexed. Pemetrexed is a structural analog of the
necessary. Sodium thiosulfate and other antioxidant antifolate agent MTX, which causes tubular injury via
drugs have been proposed as therapy for ATN with plat- crystal deposition (discussed later). However, data from
inum drugs, but the side effects of these drugs and vari- a recent case report suggest that pemetrexed induces di-
able efficacy have limited their clinical use thus far. rect tubular injury, causing ATN.20 The risk is higher in
Another theoretical approach to prevent cisplatin neph- patients who received prior potentially nephrotoxic che-
rotoxicity involves the use of substrates that would com- motherapy and in those with risk factors for CKD such
pete for uptake via OCT-2 such as cimetidine, but thus far diabetes and hypertension. Apical folate receptors and/
this approach has not been studied clinically. or basolateral folate carriers may transport pemetrexed
Ifosfamide. In producing the nephrotoxic metabolite into tubular cells; thus, cytotoxicity is tied to an antifolate
chloracetaldyhyde (Fig 2), ifosfamide is unique from its effect that impairs DNA and RNA synthesis. Kidney bi-
parent drug, cyclophosphamide, in inducing tubular opsies in patients with pemetrexed-associated AKI dis-
injury. The kidney may be susceptible because the cyto- play features typical of ATN, including loss of brush
chrome p450 enzymes that are responsible for the metabo- border and tubular atrophy.20
lism of ifosfamide into toxic substrates may be highly
expressed in the kidney.17 In addition, basolateral uptake Tubulopathies
of ifosfamide via the OCT-2 transporter, which does not Some agents that cause ATN and AKI may also display
transport cyclophosphamide, enhances ifosfamide neph- metabolic and water derangements. These abnormalities
rotoxicity. Antagonism of the OCT-2 transporter with reflect isolated tubular injury at distinct segments and
a drug such as cimetidine is a potential strategy to reduce may occur with either reduced GFR or preserved kidney
tubular injury.2 Ifosfamide is an alkylating agent that is function.
used in treating several solid organ tumors as well as cer- Proximal Tubule. FS is the clinical description for gener-
tain lymphomas and sarcomas. The risk of AKI with ifos- alized dysfunction of the proximal tubule, which is
60 Shirali and Perazella

responsible for bicarbonate, amino acid, glucose, and able gastrointestinal side effects that limit patient
phosphate reabsorption from newly filtered fluid. Thus, adherence and are not always effective, intravenous
the major clinical features of proximal tubulopathy in- dosing is often required. Fortunately, kidney mag-
clude nondiabetic glucosuria, renal tubular acidosis, nesium wasting resolves 4 to 6 weeks after drug
and urinary phosphate wasting. Urine studies are reveal- discontinuation.
ing of these features, although incomplete FS will not dis- Water handling at the collecting tubule is under the
play all of these abnormalities. In these subclinical cases, control of the hormone vasopressin (or antidiuretic hor-
other urinary biomarkers have been suggested for early mone). In the kidney, vasopressin binds primarily to the
diagnosis, including retinol binding protein and b-2 mi- vasopressin receptor 2, initiating a G-protein-coupled
croglobulin.21 Among chemotherapy drugs, ifosfamide signaling cascade that results in the insertion of water-
and cisplatin are most commonly seen with complete permeable channels in the apical membrane of the collect-
FS, whereas imatinib use is reported to cause hypophos- ing duct. A luminal osmotic gradient allows water to
phatemia from a partial proximal tubulopathy.22 Despite move into the cells of the collecting duct, after which it
cessation of ifosfamide therapy, tubulopathy manifested moves across the freely permeable basolateral membrane
by impaired phosphate reabsorption may persist for into the systemic circulation. Chemotherapy drugs such
years in survivors of childhood malignancies.21 Although as cisplatin and ifosfamide interfere with vasopressin ac-
these persistent changes may not result in serum electro- tivation of the arginine vasopressin receptor 2, resulting
lyte abnormalities, the effect on more subtle parameters in nephrogenic diabetes insipidus.19 Clinically, patients
such as bone growth, etc, is unclear.21 experience polyuria and polydipsia, and urine studies
Loop of Henle. In addition to ATN and FS, cisplatin may demonstrate low specific gravity and osmolarity, indicat-
also cause a salt-wasting nephropathy. Although much ing an inability to appropriately concentrate urine. Unless
more rare than a diagnosis of ATN or FS, in a single- access to free water is limited or thirst is greatly dimin-
center report, 10% of patients treated with conventional ished, serum sodium levels are usually in the normal to
cisplatin doses developed profound volume depletion, high-normal range. Severe hypernatremia can occur
which was characterized by orthostatic blood pressures when water intake is impaired. A recent systematic re-
changes, polyuria, and inappropriately high urine so- view suggested that remission of nephrogenic diabetes
dium concentrations.23 It is interesting to note that half insipidus usually occurs within several days to weeks
of the patients required continued management of kid- of drug cessation.26 On the opposite spectrum to im-
ney sodium wasting for months after discontinuation of paired kidney responsiveness to vasopressin, certain che-
cisplatin, indicating severe and irreversible injury in motherapeutic agents cause a heightened vasopressin
some cases.23 The mechanism is thought to involve injury response, which results in water intoxication and hypona-
at the loop of Henle, which impairs the kidney’s ability to tremia. Patients with various malignancies are already
concentrate urine and leads to polyuria. Proximal tubular predisposed to hyponatremia because they often suffer
injury is likely the primary mechanism behind the salt- from nausea/vomiting and pain, which are potent nonos-
wasting phenomenon. The loop of Henle also par- motic stimuli for vasopressin release. Cyclophosphamide
ticipates in paracellular magnesium transport; thus, and vincristine are among the chemotherapeutic agents
hypomagnesemia may accompany the salt-wasting syn- that potentiate the release and effect of vasopressin.19
drome with cisplatin. Crystal Nephropathy. Some chemotherapeutic drugs are
Collecting Duct and Distal Tubule. The distal nephron is directly cytotoxic to renal tubular cells. Other drugs medi-
the primary site for magnesium reabsorption in the ate tubular toxicity via formation of crystalline drug parti-
kidney. This is principally mediated by the epithelial cles. Crystal nephropathy refers to acute and chronic
channel transient receptor potential M6, which depends kidney injury resulting from precipitation and deposition
on epidermal growth factor receptor signaling for of crystals within the renal tubular lumen. Crystal precip-
activation. Hypomagnesemia due to urinary magne- itation related to drugs occurs either with the parent drug
sium wasting is increasingly being described in as- or its metabolites and is influenced by several factors.
sociation with novel anti-epidermal growth factor Among these factors, the principal one that increases the
receptor targeted antibody therapies including cetuxi- risk of crystal nephropathy is volume depletion. Whether
mab and panitumumab. Patients receiving these agents, true or effective volume depletion, clinical states that de-
which are gaining favor in the treatment of metastatic crease urinary flow rates predispose to crystal precipita-
colon cancer and other malignant diseases, have an tion. CKD (particularly with GFR , 60) also increases
incidence rate of hypomagnesemia that ranges between the risk of crystal nephropathy in 2 ways. First, with a re-
10% and 36% in clinical trials.24,25 Fractional excretion of duced number of functioning nephrons, the kidney is
magnesium in the urine exceeding 15% in the setting of more susceptible to any insult, including crystal nephrop-
hypomagnesemia is diagnostic of kidney magnesium athy. Second, with reduced GFR, excessive drug dosing is
wasting. Oral magnesium supplements are the first more likely to occur, especially when kidney impairment
line of treatment; however, because they have intoler- is masked and goes unrecognized. Increased serum drug
Tubulointerstitial Nephrotoxicity of Chemotherapy 61

levels lead to an increased filtered load and an intratubu- Preventive strategies such as leucovorin with or with-
lar drug burden that can cause kidney injury. out thymidine rescue to protect against high MTX levels
Malignancy-associated crystal nephropathy is gener- and administration of alkaline fluid to increase urine
ally associated with uric acid nephropathy, as seen in solubility (Fig 3) are paramount in avoiding toxic drug
the tumor lysis syndrome associated with high-grade effects.25 However, specific treatment strategies also ex-
leukemias and lymphomas. However, this type of crystal ist. Hemodialysis-based methods have also been used,
nephropathy is drug independent because any drug that with high-flux dialysis displaying the greatest potency
can cause rapid tumor killing and release large amounts in decreasing MTX levels.27 However, a limitation of
of uric acid can lead to deposition of uric acid crystals in this method is that MTX plasma levels rebound quickly
tubular lumens. However, drug-specific crystal nephrop- after cessation of dialysis. The U.S. Food and Drug
athy in malignant states is specifically seen with MTX. Administration-approved drug Carboxypeptidase G2
MTX is a drug with antimetabolite activity that stems (glucarpidase) is a recombinant enzyme that metabo-
from its antagonism of the folate pathway. It is used in lizes MTX into nontoxic derivatives. In a retrospective
treatment of several cancers, including several solid tu- trial that enrolled patients with MTX-induced nephro-
mors and leukemias. In addition, it is also included in toxicity and elevated plasma MTX levels, glucarpidase
the treatment regimen against autoimmune diseases lowered levels by 98% within 15 minutes.29 More impor-
such as rheumatoid arthritis, psoriasis, and to a lesser de- tantly, the levels remained low, and most patients did
gree systemic lupus erythematosis. The principal mecha- not require additional drug dosing. The metabolites
nism by which MTX exhibits anticancer activity is via generated by glucarpidase can cross-react with commer-
inhibition of dihydrofolate reductase, a key enzyme cial MTX assays; therefore, care must be taken in inter-
that is responsible for maintaining the active folate—tet- preting levels that are obtained with these assays.
rahydrofolate—levels that are necessary for pyrimidine Because leucovorin is also a substrate for glucarpidase,
synthesis. Thus, use of MTX blocks the DNA and RNA it should not be dosed within 2 hours of the enzyme be-
synthesis and rapid cell division that are characteristic ing dosed. At this time, there are no trials examining the
of tumor cells. efficacy of hemodialysis vs glucarpidase in the treat-
Up to 90% of MTX is cleared by the kidneys, and crys- ment of MTX toxicity complicated by AKI. Because
tallization within tubular fluid is enhanced by acidic pH. plasma levels rebound after high-flux hemodialysis,
MTX metabolizes into 2 primary derivatives, 7-hydroxy- glucarpidase appears to be the most efficient strategy
MTX and 2,4-diamino-N10-methylpteroic acid, which are in achieving sustained reduction in MTX levels.
6- and 10-fold less soluble in the urine than MTX, respec-
tively. Because MTX is used to effectively treat various
diseases, it is administered over a wide dose range
from 20 mg/m2 to 1000-33,000 mg/m2; within this range
it has a generally safe record when combined with leuco-
vorin rescue. For treatment of malignancies, MTX deliv-
ery is through prolonged intravenous infusions. This
regimen is well tolerated in patients with normal kidney
function when care is taken to ensure volume repletion
that favors high urine flow rate and urine alkalinization.
These steps are preventive measures to avoid crystalliza-
tion because an increase in urine pH from 6.0 to 7.0 re-
sults in a 5- to 8-fold increase in the solubility of MTX
and its metabolites.27 Crystallization of these substances
is the primary etiology of MTX nephrotoxicity, although
direct tubular toxicity may also play a role.
MTX-induced kidney dysfunction is tied to high se-
rum drug and metabolite levels and usually manifests
as nonoliguric AKI. With proper preventive strategies,
the risk of AKI is estimated at 1.8% on the basis of osteo-
sarcoma trials.28 Although most cases display peak Figure 3. Prevention and treatment of methotrexate nephro-
serum creatinine concentration at 1 week postdosing, toxicity. High-risk patients more commonly develop nephro-
with recovery 2 to 3 weeks later, some cases may develop toxicity from high-dose methotrexate. Preventive strategies
prolonged and severe AKI. In these cases, plasma MTX include intravenous fluids, urinary alkalinization, and leuco-
vorin rescue. When acute kidney injury develops and drug
concentrations may rise further and increase the risk for levels are excessive, glucarbidase and high-flux hemodialy-
systemic toxicities of MTX, which include bone marrow sis should be considered. HD, hemodialysis; IV, intravenous;
suppression and neurotoxicity. GFR, glomerular filtration rate.
62 Shirali and Perazella

However, at this time, the cost of this therapy remains Conclusion


somewhat prohibitive.
Chemotherapeutic agents lead to several types of kidney
tubulointerstitial disease, including AKI from ATN, tu-
AIN bulopathies, crystal nephropathy, and acute or chronic
Despite the many reports linking a wide variety of drugs tubulointerstitial nephritis. Clinically, patients may expe-
to AIN, these cases are rare with conventional chemo- rience AKI that recovers or progresses to CKD. Isolated
therapy drugs. A possible exception to this may be novel acid-base or electrolyte abnormalities are also common
biological chemotherapeutics that generally involve the kidney consequences of these drugs. It is important to
use of monoclonal antibodies to modulate anticancer note that patient- and drug-specific factors confer neph-
T cell immunity. For example, a single case report de- rotoxic risk and influence the development of these kid-
scribed AIN associated with use of an antibody directed ney abnormalities. Knowledge of these factors and the
against cytotoxic T-lymphocyte-associated antigen-4.30 prevention and treatment of chemotherapy-associated
This drug had been previously linked to drug-induced nephropathies is essential to the overall care of the pa-
lupus nephritis,31 but in the case of AIN, the authors tient with malignant disease.
describe kidney biopsy findings that include a dense
inflammatory infiltrate, predominantly consisting of
References
CD81 T cells, and no glomerular pathology.30 This drug
has been linked to inflammatory disease in other organs, 1. Lee W, Kim RB. Transporters and renal drug elimination. Annu Rev
including enterocolitis and thyroiditis, suggesting the Pharmacol Toxicol. 2004;44(1):137-166.
2. Perazella MA. Renal vulnerability to drug toxicity. Clin J Am Soc
possibility of autoimmunity rather than true drug sensi-
Nephrol. 2009;4(7):1275-1283.
tivity as the basis for kidney inflammation.32 However, 3. Khrunin AV, Moisseev A, Gorbunova V, Limborska S. Genetic poly-
despite this lone report implicating cytotoxic T-lympho- morphisms and the efficacy and toxicity of cisplatin-based chemo-
cyte-associated antigen-4 antagonism, there is not a col- therapy in ovarian cancer patients. Pharmacogenomics J. 2010;10(1):
lected body of evidence at this point to suggest a strong 54-61.
4. Anders MW. Metabolism of drugs by the kidney. Kidney Int.
association between AIN and chemotherapeutic agents.
1980;18(5):636-647.
5. Betjes MG, Bajema I. The pathology of jaundice-related renal in-
Chronic Interstitial Disease sufficiency: cholemic nephrosis revisited. J Nephrol. 2006;19(2):
229-233.
As mentioned earlier, chemotherapeutic drugs may cause 6. Wong TM, Yeo W, Chan LW, Mok TSK. Hemorrhagic pyelitis, ure-
prolonged AKI with delayed recovery, ultimately result- teritis, and cystitis secondary to cyclophosphamide: case report and
ing in irreversible kidney damage and CKD. This is review of the literature. Gynecol Oncol. 2000;76(2):223-225.
most often ascribed but not limited to those drugs that 7. Izzedine H, Isnard-Bagnis C, Launay-Vacher V, et al. Gemcitabine-
cause toxic tubular injury such as cisplatin, MTX, and ifos- induced thrombotic microangiopathy: a systematic review. Nephrol
Dial Transplant. 2006;21(11):3038-3045.
famide. Histologically, the kidney parenchyma displays 8. Eremina V, Jefferson JA, Kowalewska J, et al. VEGF inhibition and
chronic changes, including tubular atrophy and general- renal thrombotic microangiopathy. N Engl J Med. 2008;358(11):
ized interstitial fibrosis. Glomerular scarring is also noted 1129-1136.
in these histological sections. Such severe disease is more 9. Markowitz GS, Appel GB, Fine PL, et al. Collapsing focal segmental
likely to develop if certain drug- and patient-specific risk glomerulosclerosis following treatment with high-dose pamidro-
nate. J Am Soc Nephrol. 2001;12(6):1164-1172.
factors are present. These include excessive drug levels, 10. Markowitz GS, Nasr SH, Stokes MB, D’Agati VD. Treatment with
concomitant use of other nephrotoxic agents, and under- IFN-a, -b, or -g is associated with collapsing focal segmental glo-
lying diseases such as diabetes mellitus and hypertension merulosclerosis. Clin J Am Soc Nephrol. 2010;5(4):607-615.
that confer higher risk of CKD if present. Other agents 11. Miller RP, Tadagavadi RK, Ramesh G, Reeves WB. Mechanisms of
such as the nitrosureas are less likely to cause AKI, but cisplatin nephrotoxicity. Toxins (Basel). 2010;2(11):2490-2518.
12. Arany I, Safirstein RL. Cisplatin nephrotoxicity. Semin Nephrol.
they lead to progressive CKD over a period of months 2003;23(5):460-464.
to years because of chronic tubulointerstitial nephritis. 13. Ciarimboli G, Ludwig T, Lang D, et al. Cisplatin nephrotoxicity is
The nitrosureas are alkylating agents that are the fa- critically mediated via the human organic cation transporter 2.
vored therapy for certain malignancies because of their Am J Pathol. 2005;167(6):1477-1484.
ability to cross the blood-brain barrier. They are generally 14. Hartmann J, Lipp H. Toxicity of platinum compounds. Expert Opin
Pharmacother. 2003;4(6):889-901.
associated with dose-dependent nephrotoxicity that is clin- 15. Raymond E, Chaney SG, Taamma A, Cvitkovic E. Oxaliplatin: a
ically characterized by slowly progressive CKD; however, review of preclinical and clinical studies. Ann Oncol. 1998;9(10):
streptozocin has also been associated with AKI and FS. 1053-1071.
Biopsy studies demonstrate chronic changes including 16. Ludwig T, Riethmuller C, Gekle M, Schwerdt G, Oberleithner H.
tubular atrophy, interstitial fibrosis, and glomerulosclero- Nephrotoxicity of platinum complexes is related to basolateral
organic cation transport. Kidney Int. 2004;66(1):196-202.
sis.33 Although all nitrosureas carry some nephrotoxic 17. Aleksa K. Cytochrome P450 3A and 2B6 in the developing kidney:
risk, streptozocin and semustine maintain higher risk implications for ifosfamide nephrotoxicity. Pediatr Nephrol. 2005;
when compared with carmustine and lomustine.34 20(7):872-885.
Tubulointerstitial Nephrotoxicity of Chemotherapy 63

18. Skinner R. Nephrotoxicity–what do we know and what don’t we insipidus: a systematic review. Am J Kidney Dis. 2005;45(4):
know? J Pediatr Hematol Oncol. 2011;33(2):128-134. 626-637.
19. Perazella MA, Moeckel GW. Nephrotoxicity from chemotherapeu- 27. Widemann BC, Adamson PC. Understanding and managing meth-
tic agents: clinical manifestations, pathobiology, and prevention/ otrexate nephrotoxicity. Oncologist. 2006;11(6):694-703.
therapy. Semin Nephrol. 2010;30(6):570-581. 28. Widemann BC, Balis FM, Kempf-Bielack B, et al. High-dose
20. Glezerman IG, Pietanza MC, Miller V, Seshan SV. Kidney tubular methotrexate-induced nephrotoxicity in patients with osteosar-
toxicity of maintenance pemetrexed therapy. Am J Kidney Dis. coma. Cancer. 2004;100(10):2222-2232.
2011;58(5):817-820. 29. Widemann BC, Balis FM, Kim A, et al. Glucarpidase, leucovorin,
21. Skinner R. Nephrotoxicity—what do we know and what don’t we and thymidine for high-dose methotrexate-induced renal dysfunc-
know? J Pediatr Hematol Oncol. 2011;33(2):128-134. tion: clinical and pharmacologic factors affecting outcome. J Clin
22. François H, Coppo P, Hayman J, et al. Partial Fanconi syndrome in- Oncol. 2010;28(25):3979-3986.
duced by imatinib therapy: A novel cause of urinary phosphate 30. Jolly EC, Clatworthy MR, Lawrence C, Nathan PD, Farrington K.
loss. Am J Kidney Dis. 2008;51(2):298-301. Anti-CTLA-4 (CD 152) monoclonal antibody-induced autoimmune
23. Hutchison FN, Perez EA, Gandara DR, Lawrence HJ, Kaysen GA. interstitial nephritis. NDT Plus. 2009;2(4):300-302.
Renal salt wasting in patients treated with cisplatin. Ann Intern 31. Fadel F, Karoui KE, Knebelmann B. Anti-CTLA4 Antibody–In-
Med. 1988;108(1):21-25. duced lupus nephritis. N Engl J Med. 2009;361(2):211-212.
24. Fakih MG, Wilding G, Lombardo J. Cetuximab-induced hypomag- 32. Beck KE, Blansfield JA, Tran KQ, et al. Enterocolitis in pa-
nesemia in patients with colorectal cancer. Clin Colorectal Cancer. tients with cancer after antibody blockade of cytotoxic T-
2006;6(2):152-156. lymphocyte-associated antigen 4. J Clin Oncol. 2006;24(15):
25. Tejpar S, Piessevaux H, Claes K, et al. Magnesium wasting associated 2283-2289.
with epidermal-growth-factor receptor-targeting antibodies in colo- 33. Schacht RG, Feiner HD, Gallo GR, Lieberman A, Baldwin DS.
rectal cancer: A prospective study. Lancet Oncol. 2007;8(5):387-394. Nephrotoxicity of nitrosoureas. Cancer. 1981;48(6):1328-1334.
26. Garofeanu CG, Weir M, Rosas-Arellano MP, Henson G, 34. Sahni V, Choudhury D, Ahmed Z. Chemotherapy-associated renal
Garg AX, Clark WF. Causes of reversible nephrogenic diabetes dysfunction. Nat Rev Nephrol. 2009;5(8):450-462.

You might also like