Nephrotoxicity of Chemotherapy Agents



Fig. 4.1
Risk of chemotherapy-associated AKI depends on the interactions among patient, drug, and kidney specific risk factors. ADE adverse drug effects, AKI acute kidney injury, GFR glomerular filtration rate



In addition to kidney- and drug-specific risk factors, patient characteristics are critical in understanding how chemotherapy drugs affect the kidney. For example, the risk for excessive drug dosing is higher in older and female patients because they have reduced total body water and excessive serum drug concentrations may occur in this population. Additionally, decreased muscle mass in these patients results in lower creatinine, which may be misinterpreted as preserved or normal glomerular filtration rate (GFR) rather than reduction of muscle mass. One approach that may reduce excessive dosing in these patients is to base chemotherapy dosing on measured rather than estimated creatinine clearance. If that is not feasible, use of estimating equations for GFR other than Cockcroft–Gault may be more precise. While there is scant data examining the accuracy of various estimating equations, one retrospective study found that the Wright equation was superior to Cockcroft–Gault in accurately calculating GFR (using [51Cr]-ethylenediamine tetraacetic acid measured clearance as the standard) [3] .

Comorbidities in the cancer patients also influence the development of nephrotoxicity. These may include cirrhosis and congestive heart failure, which result in a functional prerenal state via decreased effective circulating volume. Adverse nonrenal side effects of chemotherapy including vomiting and diarrhea similarly predispose the cancer patient to kidney injury via creating a prerenal state. Furthermore, certain types of cancers are associated with higher proclivity for renal injury. For example, patients who have malignancies affecting the biliary system may also have obstructive jaundice, which may lead to renal hypoperfusion and bile salts-related tubular toxicity. Malignancies of the hematopoietic system such as leukemia and lymphoma may cause kidney injury directly via infiltration into renal parenchyma or indirectly via tumor lysis syndrome. Finally, paraproteinemic disorders such as multiple myeloma induce a diverse spectrum of kidney disease including amyloidosis, light and heavy chain deposition, and cast nephropathy. In patients with the aforementioned cancers, additional risk factors for nephrotoxicity compound the propensity for adverse renal drug effects .



Clinical and Pathological Classification of Nephrotoxicity from Chemotherapy Agents


Chemotherapy agents can cause kidney disease that fits the traditional grouping into prerenal, intrarenal, and postrenal states . However, most of these agents cause intrinsic renal injury at various parts of the nephron (Fig. 4.2), with a couple of notable exceptions. For example, interleukin-2 (IL-2) is associated with capillary leak syndrome, which can cause intravascular volume depletion and prerenal azotemia. Postrenal injury is rare with chemotherapy agents but case reports have linked cyclophosphamide with bladder outlet obstruction from vesicular thrombi in the setting of hemorrhagic cystitis [3]. In the following sections, we discuss intrarenal injury from chemotherapy agents common in current clinical practice, using a case-based approach to highlight clinical syndromes of acute tubular necrosis (ATN), tubulopathies, vascular injury, glomerular disease, acute interstitial nephritis (AIN), and crystal nephropathy (Fig. 4.2, Table 4.1) .

A314216_1_En_4_Fig2_HTML.gif


Fig. 4.2
Chemotherapy-associated nephrotoxicity acts on each part of the nephron to produce distinct clinical syndromes. ATN acute tubular necrosis, FSGS focal and segmental glomerulosclerosis



Table 4.1
Clinical syndromes of nephrotoxicity associated with chemotherapy agents

















































































































































ATN

Tubulopathies

Renal vasculature

AIN

Platinum agents

Fanconi syndrome

Hemodynamic AKI (capillary leak)

Ipilimumab, tremelimumab

Ifosfamide

Ifosfamide

IL-2

Sorafenib

Pemetrexed

Cisplatin

Denileukin diftitox

Sunitinib

Imatinib

Azacitidine

TMA
 

Mithramycin

Imatinib

Antiangiogenesis agents (VEGF and tyrosine kinase inhibitors)
 

Pentostatin

Pemetrexed

Gemcitabine
 

Zoledronate

Diaziquone

Cisplatin
 

Diaziquone

RSW

Mitomycin C
 
 
Cisplatin

IFN
 
 
Azacitidine
   
 
Magnesium wasting
   
 
Cetuximab
   
 
Panitumumab
   
 
Cisplatin
   
 
SIADH
   
 
Cyclophosphamide
   
 
Vincristine
   
 
NDI
   
 
Cisplatin
   
 
Ifosfamide
   
 
Pemetrexed
   

CKD

Nephrotic syndrome

Urinary tract and crystal nephropathy

Prerenal

Nitrosoureas, Ifosfamide

Minimal change disease

Methotrexate

Interleukin-2

Interferon α, β, γ

Cyclophosphamide

(Capillary Leak Syndrome)

FSGS

(hemorrhagic cystitis)
 

Interferon α, β, γ
   

Pamidronate, zoledronate (rare)
   


AKI acute kidney injury, RSW renal salt wasting, ADH antidiuretic hormone, SIADH syndrome of inappropriate antidiuretic hormone, NDI nephrogenic diabetes insipidus, AIN acute interstitial nephritis, CKD chronic kidney disease, FSGS focal segmental glomerulosclerosis, VEGF vascular endothelial growth factor TMA thrombotic microangiopathy, IL-2 interleukin-2, ATN acute tubular necrosis, IFN interferon


Acute Tubular Necrosis (ATN)



Case #1

A 66-year-old male has a history of hypertension (HTN) and chronic kidney disease (CKD), with baseline creatinine of 1.5 mg/dL. He is diagnosed with stage IV non-small cell lung cancer and is initiated on therapy with cisplatin, bevacizumab, and pemetrexed. Bevacizumab is discontinued after one cycle due to the development of a stomach ulcer. Cisplatin and pemetrexed are continued for seven more cycles. During the most recent infusion, the patient develops the following laboratory abnormalities: creatinine of 2.2 mg/dL, BUN of 37 mg/dL, bicarbonate of 20 meq/L, and potassium of 4.5 meq/L. Urinalysis shows no proteinuria but a urine microscopy shows granular casts and RTEC. Urinary sodium is 35 meq/L. A kidney sonogram reveals no hydronephrosis.

Which one of the following is true?



a.

Furosemide or mannitol given in addition to saline is proven to decrease the risk of AKI in cisplatin treatment

 

b.

The basolateral OCT-2 channel may mediate the tubular injury seen with both cisplatin and ifosfamide.

 

c.

This patient’s baseline renal insufficiency is not a risk factor for AKI due to cisplatin treatment

 

d.

Substituting oxaliplatin or carboplatin for cisplatin would not have reduced his risk for AKI

 

e.

Only pemetrexed is a likely cause for this patient’s AKI

 

As noted above, some drugs used for the treatment of malignancies cause direct injury to the cells of the renal tubules (Fig. 4.2), resulting in cellular death and ATN. Several chemotherapy drugs are implicated in causing ATN (Table 4.1). Depending on the particular drug or the severity and duration of injury, renal recovery may not be complete and result in CKD. Clinically, ATN presents with a rise in creatinine and decline in GFR. Tubular sodium reabsorption may be compromised, and a high urine sodium or FeNa > 2 % thus helps distinguish ATN from a prerenal insult. Microscopy may reveal RTEC and either granular or RTEC casts. Occasionally, severe injury can result in oliguric AKI, necessitating renal replacement therapy .


Cisplatin


Of the many drugs reported to cause ATN, cisplatin is the most well-known and best-studied nephrotoxic chemotherapy agent. It has been one of the oldest and the most frequently used platinum-based compounds in the treatment of cancer [4], and its effect on the kidney is a prototype for chemotherapy-induced nephrotoxicity. Cisplatin is known to cause dose-dependent ATN in up to one third of the patients receiving therapy [4], and appears to exert its deleterious effect mainly on proximal tubular cells. The kidney serves as the principal pathway for cisplatin excretion from the body, and the drug tends to accumulate in the kidney more so than other organs. Cells of the proximal tubule uptake cisplatin via OCT-2 [5], which leads to drug accumulation within proximal renal tubular cells that is five times greater than serum concentration [6]. This accumulation of cisplatin and its metabolites explains the drug’s preference for toxicity and damage to proximal tubule cell [7]. Most commonly, this injury results in tubular necrosis with AKI but as discussed later, an isolated proximal cell tubulopathy with acidosis and electrolyte abnormalities can also develop .

Cisplatin-induced nephrotoxicity is mediated through several pathways, including oxidative stress injury, upregulation of inflammatory mediators, and triggering of cell apoptosis [8]. The primary step occurs when a chloride ion on the parent drug is hydrolyzed and releases free hydroxyl radicals [4]. Through this and various other pathways, the production of reactive oxygen species is upregulated in cisplatin-exposed tubular cells. The resulting free radicals are directly toxic to various cell structures and promote apoptosis [6]. Inflammatory changes that occur include enhanced renal expression of tumor necrosis factor-alpha (TNF-apha), a potent pro-inflammatory molecule. TNF-alpha, along with mitogen-activated protein kinase and p53, leads to kidney injury via apoptosis [4] , and further production of cytokines, chemokines, and reactive oxygen species [6] . Apoptosis also occurs through other mechanisms, including activation of initiator caspases through mitochondrial dysfunction and oxidative stress. A direct effect of cisplatin on caspase 1 which activates caspase 3, a final pathway in the apoptosis cascade, may also contribute [8]. Finally, cisplatin therapy may also decrease renal blood flow via damage to the renal vasculature, resulting in direct ischemic or hypoxic effects to the proximal tubule. Other platinum-based drugs appear to carry less of a risk for nephrotoxicity. In in vitro studies, carboplatin and oxaliplatin display no affinity for OCT-2, and both lack chloride ions on their stem [5, 9, 10]. However, in high cumulative doses and in patients with the appropriate risk factors, there is still an appreciable risk of ATN from these newer derivative platinum drugs [11T, 12].

The management of cisplatin-induced renal injury is centered on prevention. Volume repletion, usually with isotonic saline, is a standard treatment. Once ATN has occurred, avoiding further dosing of the drug is key, and the concomitant use of other potential nephrotoxins should be strictly avoided. Though diuretics have been used to increase urine flow as a prophylactic strategy, a randomized control trial by Santoso et al. comparing saline alone, furosemide with saline, and mannitol with saline, was closed early due to a trend toward increased AKI in the mannitol group [13]. There was no difference at the time between the subjects receiving furosemide compared to saline alone [13] . Other strategies to reduce AKI include the glutathione analog amifostine and sodium thiosulfate, which may protect against free radical injury [14]. However, use of these drugs is currently limited in clinical practice due to the lack of rigorous clinical data regarding their efficacy, significant side effects and cost of these drugs, and persistent concern that their use may hinder the antitumor effect of cisplatin. Several other agents studied in animals have not yet made it into clinical practice (nucleophilic sulfur thiols, neurotrophins, phosphonic acid, melanocortins, and free oxygen radical scavengers) [15]. Chemical substrates such as cimetidine compete with cisplatin for uptake via OCT-2, and have been suggested as therapeutic interventions to prevent intracellular concentration in the proximal tubule cells, but thus far there is a lack of clinical studies targeting this mechanism .


Ifosfamide


Similar to cisplatin, ifosfamide and its metabolites are known to be directly toxic to the cells of the proximal tubule. It is an alkylating agent commonly used for the treatment of several different solid organ tumors, as well as certain lymphomas and sarcomas. Renal toxicity occurs in up to 30 % of those on treatment [16], but unlike cisplatin, AKI is less frequent, and the more common manifestation is Fanconi syndrome (FS) or an isolated proximal renal tubular acidosis (Type 2 RTA) [17].

Unlike its parent drug, cyclophosphamide, ifosfamide (Fig. 4.2) produces the nephrotoxic metabolite chlorocetaldehyde. The kidney may be more susceptible to injury from ifosfamide and its metabolite because the cytochrome p450 enzymes that are responsible for the metabolism of ifosfamide are highly expressed in the kidney [18] . Furthermore, similar to cisplatin, ifosfamide is actively transported into the tubular cells via the basolateral transporter OCT-2, whereas cyclophosphamide is not, again suggesting a possible therapeutic target for renal injury prevention [19]. Despite this, the overall risk of AKI is less when compared to cisplatin. A cumulative lifetime dose of greater than 60–80 g/m2 is associated with an increased risk for nephrotoxicity, but renal injury may happen at lower levels as well. Prior cisplatin use may also be an independent risk factor for injury [20]. Mesna, which is commonly used to prevent hemorrhagic cystitis, does not help to prevent AKI [16]. Not much is known about the mechanism of ifosfamide toxicity, but its effect on long-term renal function and the development of CKD will be discussed later.


Pemetrexed


An antifolate agent that is a structural analogue of MTX , pemetrexed acts to inhibit enzymes involved in purine and pyrimidine metabolism, thus interfering with the DNA and RNA synthesis which is necessary for cell replication of rapidly dividing tumor cells. It is used commonly for malignancies such as mesothelioma and non-small cell lung cancer among others. Uptake into proximal tubule cells occurs via both apical folate receptor-α transporters and basolateral-reduced folate carriers, and once in the cells, pemetrexed becomes trapped by polyglutamylation (Fig. 4.3) [15]. Increasing intracellular concentrations lead to further inhibition of folate metabolism, and likely contribute to renal tubular cell injury. Thus, both the toxicity and therapeutic effect of pemetrexed are tied to its antifolate action .

A314216_1_En_4_Fig3_HTML.gif


Fig. 4.3
Simplified schema illustrating effect of methotrexate (MTX) and pemetrexed (PTXD) on enzymes involved in de novo purine and pyrimidine metabolism. DHFR dihydrofolate reductase, dTMP deoxythymidine monophosphate, dUMP deoxyuridine monophosphate, EC extracellular, IC intracellular, TS thymidylate synthase

In addition to pemetrexed-induced AKI due to ATN [21], cases of nephrogenic diabetes insipidus (DI) and RTA have also been reported [22]. Patients who have received prior chemotherapy agents that are potential nephrotoxins, and those who have risk factors for CKD such as diabetes or HTN, have a higher risk of kidney injury. Biopsies of kidneys have shown loss of brush borders, tubular atrophy, with some interstitial inflammation [21].


Crizotinib


As the armamentarium of chemotherapeutic agents expands, new drugs are now rapidly entering clinical use. Some of these newer drugs have been reported to cause AKI in a similar pattern to ATN, though their exact mechanisms of toxicity are not often well defined. Crizotinib, a tyrosine kinase inhibitor of multiple pathways including anaplastic lymphoma kinase (ALK), is in increasing use as a therapy for metastatic ALK-positive non-small cell lung cancer. Due to its expedited FDA approval, some of its side effects were not clearly known, and since its release in 2011, several reports of AKI and renal insufficiency have been reported [2326]. A review of 38 patients who were given crizotinib for an average of 16 months reported a 23.9 % decline in estimated GFR during the initial 12 weeks of therapy. On withdrawal of the drug, the majority of patients recovered kidney function [25]. In one report of an episode of AKI associated with crizotinib, investigators were able to obtain a renal biopsy, which showed ATN as the primary lesion [24], though the mechanism of nephrotoxicity was not defined. In addition to ATN, non-nephrotic range and non-albuminuric proteinuria has also been reported [24] .


Carfilzomib


Another example of drug-induced injury is being reported in the treatment of multiple myeloma. Renal disease is not uncommon in multiple myeloma and therefore, a direct causal effect from a single drug is difficult to prove. Nonetheless, there are reports of AKI with carfilzomib, a next generation selective proteasome inhibitor approved for the treatment of relapsed and refractory multiple myeloma. Jhaveri and colleagues reported a case of a patient with IgG kappa multiple myeloma undergoing chemotherapy with carfilzomib and steroids who presented with fever and AKI 9 days after his last treatment, which resolved after conservative management, including drug discontinuation [27]. In a phase 2 trial of carfilzomib in the treatment of multiple myeloma, both acute and chronic renal failure were reported (5 and 3.8 %), and some of these subjects were managed with drug cessation, interruption, or dose reduction [28]. Though the mechanism of renal dysfunction is unknown, some have postulated that N-acetylcysteine may play a protective role in injury prevention, perhaps suggesting a partly prerenal or vasoconstrictive etiology to the AKI [29] .


Mammalian Target of Rapamycin (mTOR) inhibitors


Proteinuria is a well-known side effect of mammalian target of rapamycin (mTOR) inhibitors in some patients. However, a new association of mTOR inhibitors with ATN has recently surfaced. Four cases of biopsy-proven ATN occurred in patients undergoing therapy for lymphoma or metastatic malignancy [30]. Two of the cases rapidly recovered after drug discontinuation while the other two remained in renal failure, and one of the cases clearly showed signs of concomitant focal segmental glomerulosclerosis (FSGS). mTOR activity increases in the kidney after ischemic injury and may be involved in cell growth and repair; mTOR complex 1 (mTORC1), a protein complex formed in part by mTOR kinase, is specifically involved in the upstream inhibition of autophagy [30]. Therefore, induction of autophagy, particularly during times of renal tubular cell stress or injury, may be the mechanism by which these drugs cause renal damage [30]. MTOR inhibitors are used at much higher doses in cancer treatment compared to post-transplant immunosuppression, which may explain the lack of AKI associated with MTOR-inhibitor use in the solid organ transplant population [30] .


Clofarabine


Clofarabine is a purine nucleoside analog that exerts its anti-neoplastic effect by inhibiting DNA synthesis and the enzyme ribonucleotide reductase (RNR). It is used routinely for the treatment of relapsed acute lymphoblastic leukemia in children, and it is increasingly also being used for relapsed or refractory acute myeloid leukemia in adults. Two case reports outline patients treated with clofarabine who developed severe renal injury shortly after drug administration; one of the patients was found to have 4 g of proteinuria, and the other was anuric and required dialysis [31, 32]. No biopsy data exist to help propose a mechanism of injury, but RNR inhibition may be contributing to podocyte injury [31] .


Androgen Deprivation Therapy (ADT)


Recent epidemiological data has linked androgen deprivation therapy (ADT) to an increased risk of AKI in patients undergoing treatment for prostate cancer. In a group of over 10,000 patients with prostate cancer followed-up for 10 years and matched against selected controls, current ADT was associated with an adjusted odds ratio for hospitalization for AKI of 2.68; this was higher for those who received combined agent versus single agent regimens [33]. Given that ADT is still the mainstay of treatment for advanced prostate cancer, these results will require replication for clinical implications of these data to be confirmed. More population-wide epidemiological studies such as this one may alert us in the future to subtle associations of nephrotoxicity and commonly used chemotherapy agents .


Case #1 Discussion and Follow-Up:

In case #1, both cisplatin and pemetrexed are potential precipitants of AKI. The patient has a higher propensity for AKI due to his baseline CKD. Though volume expansion is important in preventing injury from cisplatin, there is no evidence to suggest that furosemide or mannitol may be helpful, and mannitol may be detrimental in certain situations. Many newer regimens substitute carboplatin or oxaliplatin for cisplatin, and these are on average less nephrotoxic than the latter. The right answer is b, as the OCT-2 channel transports both cisplatin and ifosfamide into proximal tubular cells, and is critical to their mechanism of injury.


Tubulopathies and Electrolyte Disorders



Case #2

A 47-year-old male and former chewing tobacco user who is diagnosed with surgically unresectable oral squamous cell carcinoma undergoes treatment with docetaxel, cisplatin, and 5-fluorouracil. Follow-up imaging shows progression of disease. Decision is made to start cetuximab monotherapy. After 2 months of therapy, the patient presents with fatigue, weakness, light-headedness, and complains of muscle “twitches.” Serum laboratory tests show the following levels: sodium of 129 meq/L, potassium of 3.1 meq/L, chloride of 101 meq/L, HCO3 of 18 meq/L, BUN of 34 mg/dL, and creatinine of 0.9 mg/dL. Glucose is 88 mg/dL, calcium is 7.7 mg/dL, and magnesium is 0.9 mg/dL. His urinary pH is 5.5.

Which of the following is false?



a.

His prior cisplatin use likely caused his hyponatremia by potentiation of antidiuretic hormone (ADH) and increased water reabsorption in the collecting duct

 

b.

Both cetuximab and prior cisplatin use could explain his hypomagnesemia

 

c.

His urine should be evaluated for glucose, phosphate, and magnesium

 

d.

His low calcium is likely related to his low magnesium levels

 

Certain chemotherapy agents impact renal handling of water and electrolytes either by direct cell injury (as described above), or by their effects on specific receptors or channels in distinct segments of the nephron. Due to this some patients develop electrolyte and acid–base derangements as their primary manifestation of renal toxicity. AKI may or may not be present, but regardless of the effect on GFR, the consequences can still be significant and important to recognize .


Proximal Tubule


Injury to the proximal tubule can impede reabsorption of several important electrolytes and compounds, including glucose, phosphate, bicarbonate, and amino acids. The clinical entity that ensues is named FS , and is thus characterized by glucosuria in the absence of hyperglycemia, phosphate wasting, and an RTA due to bicarbonate spilling. Incomplete or partial FS can present with some of these abnormalities but not all.

Ifosfamide is most commonly implicated in inducing FS, and though much of the literature is described in children, a few case reports in adults exist [34]. Even after cessation of therapy, tubular dysfunction from ifosfamide can persist for years, manifesting as partial FS with persistent phosphaturia, as described in childhood malignancy survivors [20]. In adults, this has been reported to lead to osteomalacia [35], and in children may possibly lead to issues with growth and bone development [20]. The doses of ifosfamide associated with FS are variable and not always at the levels associated with AKI or CKD, and the time to onset of symptoms can be immediate or delayed several months. Cisplatin, a proximal tubule cell toxin, is also associated with FS [36], though less commonly than its association with ATN. Imatinib use has also been reported to cause hypophosphatemia from hyperphosphaturia due to a partial proximal tubulopathy [37]. These defects can often go unrecognized and are important to monitor, if undergoing therapy with potential tubular toxins .


Loop of Henle


A principal site of sodium reabsorption, a defect in the loop of Henle can lead to salt wasting and volume depletion (Fig. 4.2, Table 4.1). Cisplatin, in addition to inducing ATN and FS, has been reported to induce a renal salt-wasting syndrome (RSWS) [38]. Affected patients can have profound volume depletion with orthostasis and polyuria, with laboratory tests indicating a hypoosmolar hyponatremia in the setting of a high rate of urinary sodium excretion. With these serum and urine indices, RSWS may be mistaken for syndrome of inappropriate antidiuretic hormone (SIADH) but the key difference between the two is that RSWS has a negative sodium balance despite hypovolemia. A single center report recorded an incidence of cisplatin-induced salt wasting as high as 10 % [39], though in another series a rate of < 1 % was noted [40]. Many of the patients in the former series had sodium wasting for months after discontinuation of cisplatin, and some cases were severe and irreversible [39]. As cases are uncommon, the mechanism is not well characterized at present. Proximal tubular damage likely leads to sodium delivery distally, but in other patients with FS the distal tubules assist in reabsorbing the increased sodium. Loop of Henle dysfunction is therefore postulated, as this is the site of sodium reabsorption that is critical to generating the medullary concentration gradient, which is noted to be impaired in cisplatin toxicity [38]. In support of the loop of Henle being a site of cisplatin injury, magnesium reabsorption occurs via paracellular pathways at the loop of Henle, and hypomagnesemia is reported with cisplatin use [41] (Fig. 4.2).


Collecting Duct


In addition to sodium, potassium, and water handling, the collecting duct is also responsible for magnesium homeostasis. Maintenance of magnesium stores occurs primarily via the epithelial channel transient receptor potential melastin subtype 6 (TRPM6) on the luminal surface of collecting duct cells. Magnesium reabsorption via TRPM6 has been found to be regulated by epidermal growth factor (EGF) receptor signaling. EGF binds to its receptor on the basolateral surface of the cell; this in turn sets off cellular signaling that facilitates insertion of TRPM6 channels into the apical membrane, thus allowing for magnesium reabsorption (Fig. 4.4) [42]. This interaction is directly antagonized in patients receiving anti-EGFR monoclonal antibodies such as cetuximab or panitumumab therapy, a novel class of chemotherapy agents increasingly associated with renal magnesium wasting.
Jul 17, 2017 | Posted by in NEPHROLOGY | Comments Off on Nephrotoxicity of Chemotherapy Agents

Full access? Get Clinical Tree

Get Clinical Tree app for offline access