Conventional chemotherapy


Introduction

Several conventional chemotherapeutics have been associated with renal side effects including kidney function deterioration, electrolyte disorders, tubular injury, glomerular lesions, tubulointerstitial nephritis, and the development of thrombotic microangiopathy (TMA) ( Tables 16.1–16.3 ). Multiple risk factors for chemotherapy-induced nephrotoxicity have been identified ( Box 16.1 ). The quality of data available regarding these kidney side effects is low, because subtle kidney damage goes frequently unrecognized and therefore the true incidence of nephrotoxicity is difficult to determine. Most episodes of medication-induced acute kidney injury (AKI) are reversible, but chronic kidney disease (CKD) can develop because of glomerular scarring or tubulointerstitial inflammation.

Table 16.1
Conventional Chemotherapeutics, Mechanism of Action, Cancer Types Treated
Drug Class Drug Anti-Cancer MoA Types of Cancers Treated
Platinum-based chemotherapeutics Carboplatin Inhibition of DNA replication Head/neck, lung, ovarian, breast, and testicular
Cisplatin Testicular, germ cell, ovarian, cervical, breast, bladder, head/neck, esophageal, lung, mesothelioma
Oxaliplatin Colorectal, gastrointestinal
Alkylating agents Busulfan Cross-linking of DNA strands
Inhibition of RNA production
Bone marrow transplantation conditioning, leukemia, lymphoma, myeloproliferative disorders
Cyclophosphamide Lymphoma, multiple myeloma, leukemia, ovarian, breast, small cell lung, neuroblastoma, sarcoma
Diaziquone Primary brain
Ifosfamide Hodgkin and non-Hodgkin lymphoma, soft tissue sarcomas, osteosarcoma, testicular, breast, ovarian, cervical
Melphalan Multiple myeloma
Procarbazine Hodgkin lymphoma, brain
Temozolomide Brain, melanoma
Trabectedin Soft tissue sarcoma, ovarian
Antimetabolites Azacitidine Inhibition of DNA production by incorporation of chemically altered nucleotides or by depletion of nucleotides Myelodysplastic syndrome, acute myeloid leukemia
Capecitabine Metastatic breast and colorectal, esophageal, gastric, prostate, ovarian, pancreas
Cladribine Hairy cell leukemia, chronic lymphocytic leukemia
Clofarabine Relapsed or refractory acute myeloid leukemia, lymphoblastic leukemia
Cytarabine Acute myeloid leukemia, acute lymphocytic leukemia, chronic myelogenous leukemia, Hodgkin and non-Hodgkin lymphoma
Deoxycofomycin Hairy cell leukemia, chronic lymphocytic leukemia, adult T-cell leukemia/lymphoma
Fludarabine Acute myeloid/lymphocytic leukemia, chronic lymphocytic leukemia, non-Hodgkin lymphoma
5-Fluorouracil Colorectal, esophageal, gastric, pancreatic, breast, cervical
Gemcitabine Breast, ovarian, non-small cell lung, pancreatic, bladder, cholangiobiliary
Mercaptopurine Acute leukemia, chronic leukemia
Methotrexate Breast, lung, head/neck, bladder, leukemia, lymphoma, osteosarcoma
Pemetrexed Non-small lung cancer, mesothelioma
Thioguanine Acute leukemia, chronic myeloid leukemia
Antitumor antibiotics Anthracyclines DNA intercalation resulting in blocking of DNA/RNA synthesis Acute myeloid leukemia, acute lymphoid leukemia, chronic myelogenous leukemia, neuroblastoma, Kaposi sarcoma, breast, non-Hodgkin lymphoma
Bleomycin Hodgkin and non-Hodgkin lymphoma, testicular, ovarian, cervical
Mitomycin C Bladder, esophageal, anal, breast
Chloroethylnitrosourea Interstrand cross-linking of DNA Brain, multiple myeloma, Hodgkin and non-Hodgkin lymphoma, beta cell, chronic lymphocytic leukemia, prostate
Microtubule inhibitors Taxanes Inhibition of microtubules assembly/function Breast, head/neck, gastric, prostate, non-small cell lung, ovarian, Kaposi sarcoma, cervical, pancreatic, urothelial
Vinca alkaloids Acute leukemia, Hodgkin lymphoma, lung, bladder, breast, small cell lung, sarcoma
Topoisomerase inhibitors Etoposide Single-DNA strand breaks Lung, lymphoma, testicular
Irinotecan Colon, small cell lung
Topotecan Ovarian, small cell lung
Others Tamoxifen Degradation of PML-RARα Acute promyelocytic leukemia
Binding of estrogen-receptor Breast
DNA , Deoxyribonucleic acid; PML-RAR α, promyelocytic leukemia retinoic acid receptor; RNA , ribonucleic acid.

Table 16.2
Conventional Chemotherapeutics and Clinical Renal Syndromes
Drug Class Drug Clinical Renal Syndromes
Platinum-based chemotherapeutics Carboplatin AKI, CKD, hypomagnesemia, hypokalemia, hyponatremia, TMA
Cisplatin AKI, CKD, hypomagnesemia, Fanconi syndrome, distal RTA, polyuria, TMA, salt-wasting syndrome, SIADH
Oxaliplatin AKI, ATN, hypokalemia, TMA
Alkylating agents Busulfan AKI
Cyclophosphamide SIADH, hyponatremia, hemorrhagic cystitis
Diaziquone AKI
Ifosfamide AKI, CKD, ATN, Fanconi syndrome, distal and proximal RTA, nephrogenic diabetes insipidus, hypokalemia, interstitial nephritis, glomerular disease, hemorrhagic cystitis, SIADH
Melphalan AKI, nephrotic syndrome, SIADH
Procarbazine AKI
Temozolomide AKI
Trabectedin AKI
Antimetabolites Azacitidine AKI, Fanconi syndrome, RTA, polyuria, nephrogenic diabetes insipidus
Capecitabine AKI
Cladribine AKI
Clofarabine AKI, proteinuria, collapsing glomerulopathy
Cytarabine AKI, TMA
Deoxycoformycin AKI, hematuria, dysuria, TMA
Fludarabine AKI
5-Fluorouracil AKI, hyponatremia, hypokalemia
Gemcitabine AKI, proteinuria, hematuria, TMA
Mercaptopurine AKI, Fanconi syndrome
Methotrexate Crystalline nephropathy, ATN, AKI
Pemetrexed ATN, interstitial edema, distal RTA, nephrogenic diabetes insipidus, CKD
Thioguanine AKI
Antitumor antibiotics Anthracyclines Nephrotic syndrome, glomerular diseases (MCD, FSGS), TMA, CKD
Bleomycin TMA
Mitomycin C TMA
Chloroethylnitrosourea AKI, chronic interstitial nephritis, CKD, uric acid nephrolithiasis, nephrogenic diabetes insipidus, Fanconi syndrome, proximal RTA, TMA
Microtubule inhibitors Taxanes Hyponatremia, TMA, (ATN), hypokalemia
Vinca alkaloids (AKI), SIADH
Topoisomerase inhibitors Etoposide None
Irinotecan AKI
Topotecan AKI
Others Arsenic trioxide Acute tubulointerstitial injury
Tamoxifen Nephrotic syndrome
AKI , Acute kidney injury; ATN , acute tubular necrosis; CKD , chronic kidney disease; FSGS , focal segmental glomerulosclerosis, MCD , minimal change disease, RTA , renal tubular acidosis; SIADH , syndrome of inappropriate antidiuretic hormone secretion; TMA , thrombotic microangiopathy.

Table 16.3
Causes of the Most Common Clinical Renal Syndromes Associated With Conventional Chemotherapeutics
Clinical Renal Syndrome Conventional Chemotherapeutic
AKI/ATN Carboplatin, cisplatin, oxaliplatin, (busulfan), diaziquone, ifosfamide, melphalan, procarbazine, temozolomide, trabectedin, azacitidine, (capecitabine), cladarabine, clofarabine, cytarabine, deoxycofymycin, fludarabine, 5-fluorouracil, gemcitabine, mercaptopurine, methotrexate, pemetrexed, thioguanine, chloroethylnitrosourea, (taxanes, vinca alkaloids), irinotecan, (topotecan)
CKD Carboplatin, cisplatin, ifosfamide, pemetrexed, anthracyclines, chloroethylnitrosourea
Crystalline nephropathy Methotrexate
Distal RTA Cisplatin, ifosfamide, pemetrexed
Dysuria Deoxycofomycin
Fanconi syndrome Cisplatin, ifosfamide, mercaptopurine, chloroethylnitrosourea
Glomerular disease Ifosfamide, clofarbine, anthracyclines
Hematuria Deoxycoformycin, gemcitabine
Hemorrhagic cystitis Cyclophosphamide, ifosfamide
Hypokalemia Platinum derivatives, ifosfamide, 5-fluorouracil, taxanes
Hypomagnesemia Carboplatin, cisplatin
Hyponatremia Platinum derivatives, cyclophosphamide, 5-fluorouracil, taxanes
Interstitial nephritis/injury Ifosfamide, pemetrexed, chloroethylnitrosourea, arsenic trioxide
Nephrogenic diabetes insipidus Ifosfamide, azacitidine, pemetrexed, chloroethylnitrosourea
Nephrotic syndrome Melphalan, methotrexate, anthracyclines, tamoxifen
Proteinuria Clofarabine, gemcitabine
Proximal RTA Ifosfamide, azacitidine, chloroethylnitrosourea
SIADH Cyclophosphamide, ifosfamide, melphalan, vinca alkaloids, cisplatin
TMA Carboplatin, cisplatin, oxaliplatin, cytarabine, deoxycofomycin, gemcitabine, anthracyclines, bleomycin, mitomycin C, chloroethylnitrosourea, taxanes
Uric acid nephrolithiasis Chloroethylnitrosourea
AKI , Acute kidney injury; ATN , acute tubular necrosis; CKD , chronic kidney disease; RTA , renal tubular acidosis; SIADH , syndrome of inappropriate antidiuretic hormone secretion; TMA , thrombotic microangiopathy.

Box 16.1
NSAID, Nonsteroidal anti-inflammatory drug.
Risk Factors for Chemotherapy-Induced Nephrotoxicity

Patient factors

  • Older age

  • Acute or chronic renal dysfunction

  • Genetic factors: gene mutations in hepatic/renal CYP450 enzyme systems and transport proteins

Intrinsic chemotherapeutic nephrotoxicity

  • Cumulative drug dose

  • Crystal formation by drug or metabolites

  • Direct nephrotoxic effects

  • Combination with other nephrotoxins (e.g., NSAID, iodine-containing contrast agent, nephrotoxic antibiotics)

Tumor-related risk factors

  • Volume depletion (diarrhea, vomiting, malignant ascites or pleuritis, sepsis)

  • Tumor lysis syndrome

  • Hyperuricemia, hypercalcemia

  • Renal infiltration

  • Urinary obstruction caused by tumor or radiation therapy-induced fibrosis

  • Paraprotein-related kidney disease

  • Paraneoplastic glomerulonephritis

Renal drug handling

  • Accumulation and high metabolic rate in renal tubular cells

  • Hypoxic renal environment

  • Concentration of drug/toxin in the interstitium and medulla

Adapted from Perazella MA. Onco-nephrology: renal toxicities of chemotherapeutic agents. Clin J Am Soc Nephrol . 2012; 7:1713-1721.

AKI is a common condition in cancer patients and is associated with higher cost, longer length of hospital stay, increased morbidity, and mortality. A Danish population study reported an 18% incidence of AKI in the first year after cancer diagnosis. A study from the MD Anderson Cancer Center demonstrated that the rate of AKI in hospitalized cancer patients is significantly higher than in noncancer patients, and AKI occurrence correlated significantly with the administration of chemotherapy. AKI developed in many patients during admission and was associated with longer hospital stay, increased costs, and increased mortality. Even small increases in serum creatinine in cancer patients are associated with a prolonged stay at the intensive care unit (ICU) and increased mortality. In cancer patients admitted to an ICU, 32% required dialysis and hospital, and 6-month mortality rates were 64% and 73%, respectively. In survivors, kidney function recovered completely in 82% and partially in 12%, whereas 6% required chronic dialysis therapy.

Recently, CKD has been reported as a long-term complication in cancer survivors and has been associated with decreased survival. , Chronic nephrotoxicity in cancer patients may have multiple causes, including chemotherapy, radiotherapy exposure of the kidneys, kidney surgery, supportive care drugs, iodine-containing contrast agents, and tumor-related factors. A particularly vulnerable group of patients to develop chronic renal side effects are pediatric patients. Long-term childhood cancer survivors are at increased risk to develop elevated blood pressure and decreased kidney function. , Unilateral nephrectomy, abdominal radiotherapy, treatment with cisplatin and ifosfamide, higher cisplatin dose, and cumulative carboplatin dose have all been associated with an increased risk of persistent nephrotoxicity. Chronic glomerular and tubular nephrotoxicity developed in 20% to 50% and 60% to 80% of children treated with ifosfamide and cisplatin, respectively, and after nephrectomy, 20% of children displayed evidence of chronic glomerular damage. Overall, childhood cancer survivors have a ninefold higher risk of developing kidney failure compared with their siblings. Each drug has its own pattern of injury and in this chapter, we attempt to provide a detailed overview of specific chemotherapeutic drug-associated kidney side effects.

Furthermore, the kidneys are a major excretion pathway for a number of antineoplastic drugs and their metabolites. Thus kidney impairment often results in delayed and/or decreased drug excretion and metabolism of cytotoxic chemotherapeutics, possibly leading to increased toxicity. Therefore many antineoplastic agents require dose adjustment when administered in the setting of acute kidney disease or CKD ( Table 16.4 ). Unfortunately, because severe renal impairment is usually an exclusion criterion in clinical trials, we do lack information on the excretion and metabolism of anticancer agents in this setting, though specific studies in these patients have been advocated; consequently, in patients with end-stage renal disease (ESRD) undergoing dialysis, the situation is even more complex, because antineoplastic agents’ excretion and metabolism are not fully known. Recommendations on dose adjustments of chemotherapeutics in patients with kidney dysfunction are based on our own experience and inspired by different published guidelines (see Table 16.4 ).

Table 16.4
Dose Adjustment of Chemotherapeutic Drugs in Patients With Renal Dysfunction
Chemotherapeutic agent > 90 90–80 80–70 70–60 60–50 50–40 40–30 30–20 20–10 10–0 HD PD
Carboplatin No DR (Calvert Formula) 250 mg/m ² max dose CI
Cisplatin no DR DR 25% DR 50% (consider carboplatin) DR 50–75% DR 50%
Oxaliplatin no DR consider DR ? ?
Busulfan no DR
Cyclophosphamide no DR DR25% DR50% DR 25%
Diaziquone no DR
Ifosfamide no DR DR 25% DR50% CI DR 50% DR 50%
Melphalan no DR DR25% DR 50% DR 50% ?
Temozolomide no DR LD LD
Trabectedin no DR CI CI CI
Cytarabine no DR DR 40% DR 50% CI for high dose LD LD
Deoxycofomycin no DR DR 25% DR 50% CI
Fludarabine no DR DR 20–50% CI LD DR 50%
5-Fluorouracil no DR consider DR LD LD
Gemcitabine no DR DR no DR LD
Mercaptopurine no DR increase dose interval 24–36h increase dose interval 48hr
Methotrexate no DR avoid high dose DR 50% CI DR 50–75% CI
Pemetrexed no DR CI
Thioguanine no DR consider DR
Bleomycin no DR DR 25% DR 50%
Mitomycin C no DR DR 25% DR 25%?
Streptozocin no DR DR 25% stop LD
Iomustine no DR DR 25% DR 50% DR 50%–75%
Bendamustine no DR CI
Carmustine no DR DR 20% DR25% no DR
Estramustine no DR
Lomustine no DR DR 25% DR 50% CI
Taxanes no DR no DR
Vincristine no DR
Vinblastine no Dr
Vinorelbine no DR
Eribulin no DR consider DR CI
Etoposide no DR DR 25% DR 50% DR 50% DR 50%
Topotecan no DR DR 50% DR 75% CI
CI , contraindicated; DR dose reduction; HD , hemodialysis; LD , limited data available; PD , peritoneal dialysis.

Estimation of kidney function

There are two main pathways for drug excretion by the kidney: glomerular filtration and tubular secretion. For those drugs that are excreted by the kidneys, dose adjustments are often required when kidney function is impaired, which is common in cancer patients. Because it is practically impossible to evaluate serum drug concentrations in every cancer patient, the most precise evaluation of kidney function possible is mandatory. Different methods can be used, although each method is in some way flawed and there is no consensus on an ideal tool to be used, especially in oncologic patients. , A creatinine clearance (CrCl) calculation, based upon a 24-hour collection of urine, is cumbersome and subject to error, mainly because of the need for urine collection, which is often troublesome for the patient, especially in the outpatient setting. Estimation equations, such as the Cockcroft-Gault (CG), the Modification of Diet in Renal Disease (MDRD), and the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) are presently the most common methods used in routine clinical practice. Most of the available data suggest that all these formulae provide similar levels of concordance in the measurement of glomerular filtration rate (GFR) in cancer patients, and are thus adequate enough for the purpose of dosing cancer drugs excreted by the kidneys. However, a recent report suggested that the CG equation underestimates GFR compared with the MDRD and CKD-EPI formulas, at least in Asian patients. More recently, the most accurate and least biased method to estimate GFR was described; this method applied a new multivariable linear model for GFR calculation using statistic regression analysis. Chromium-51 labeled ethylenediamine tetraacetic acid ( Cr-EDTA) GFR was compared with the estimated GFR (eGFR) from seven published models and the new model proposed; the new model improved the eGFR accuracy compared with all published models. Importantly, the new model reduced the fraction of patients with a carboplatin dose absolute percentage error greater than 20% to 14.17%, in contrast to 18.62% for the body surface area (BSA)-adjusted CKD-EPI, and 25.51% for the CG formula. Notably enough, this new model has been externally validated, and should now be considered a new “gold standard.”

Platinum-based chemotherapeutics

Carboplatin

Carboplatin is a second-generation platinum-based chemotherapeutic (including cisplatin, heptaplatin, lobaplatin, nedaplatin, oxaliplatin) and is used to treat a variety of cancers, such as head/neck cancer, lung cancer, ovarian cancer, brain cancer, and testicular cancer. Compared with cisplatin, carboplatin is better tolerated but less potent.

Mechanism of action

The mechanism of cytotoxicity of carboplatin is similar to that of cisplatin (discussed in detail in the cisplatin section), because it also interferes with deoxyribonucleic acid (DNA) duplication.

Pharmacokinetics

Carboplatin or cis-diammine (1,1-cyclobutane dicarboxylate) platinum(II) is administered intravenously. In general, the dosage of carboplatin is four times the dosage of cisplatin. More than 50% of the administered dosage is excreted unchanged in the urine within 24 hours. The importance of renal clearance to the metabolism and excretion of carboplatin is highlighted by its usual dosing schema, which is based upon an eGFR, along with the desired level of drug exposure, according to the area under the concentration-time curve (AUC, mg/mL × min), rather than the more common dosing calculation based upon the BSA (mg/m 2 ). Using the desired target AUC (which typically varies between 5 and 7 mg/mL/min) and the eGFR, the dose of carboplatin is then calculated using the Calvert formula: dose (mg) = AUC (mg/mL × min) × [GFR (mL/min) + 25 (mL/min)]. This dose calculation using the Calvert formula has to be repeated before each course of carboplatin to take into account possible changes in weight or renal function. A possible issue related to the use of this formula is the appropriate weight to use when calculating the eGFR. The original CG formula to estimate GFR used actual body weight, but none of the patients considered were obese. Most clinicians use actual body weight in the CG formula for nonobese patients. However, the use of actual body weight in the CG calculation can overestimate the GFR resulting in higher carboplatin dose in obese individuals. The Gynecological Oncology Group (GOG) recommends that actual weight be used to determine eGFR when using the CG equation as long as patients have a body mass index (BMI) of less than 25. For other patients, the use of an adjusted weight is suggested (adjusted weight [kg] = [{actual weight – ideal weight} × 0.40] + ideal weight). Another issue is that the Calvert formula was developed measuring GFR using Cr-EDTA, whereas in clinical practice CrCl or eGFR (using the MDRD or CKD-EPI formula) are used. It is not clear at this point how accurate it is to calculate the carboplatin dosage using these estimates. Carboplatin should not be administered to patients with CrCl lower than 20 mL/min and a maximum carboplatin dose of 250 mg/m 2 has been suggested in patients with CrCl=20 to 39 mL/min.

As far as carboplatin administration in patients undergoing dialysis concerns, polychemotherapy regimens incorporating carboplatin have been successfully used in patients undergoing hemodialysis (HD) and peritoneal dialysis (PD) without evidence of increased toxicity or reduced efficacy. Few cases of HD patients treated with carboplatin, either alone or in combination, are reported in the literature. The Calvert formula has been initially used in HD patients by assuming that GFR is zero. This is applicable to patients who receive HD within 12 to 18 hours after carboplatin infusion. After the first 24 hours, the majority of carboplatin, which is bound to serum proteins, is not easily dialyzable and remains in the blood stream despite repeated sessions of HD. Guddati et al. have proposed a correction factor to calculate the resultant AUC in HD patients; the AUC can increase by eightfold in patients who received the adjusted dose, but whose HD was delayed beyond 24 hours after infusion. The correction factor proposed can also be used to calculate the dose adjustment required, a priori, in patients who may receive delayed HD. The same authors have also developed a formula to adjust carboplatin dosage in PD patients; this formula takes into account the frequency of dialysis sessions and the time delay between carboplatin infusion and the initiation of dialysis, and predicts an approximately similar dosage of carboplatin as that of the Calvert formula in patients undergoing PD 4 times per day, if dialysis is initiated 12 hours after infusion. In another patient, HD was performed for 3.5 hours starting 90 minutes after completion of carboplatin, and pharmacokinetic assessments were performed at 1, 2, 4, and 12 hours after its infusion. Total carboplatin concentrations in plasma and platinum ultrafiltrate were measured. The plasma concentration of free platinum at the end of the infusion was 31,000 ng/mL and the AUC was 2.9 minutes times mg/mL. No significant carboplatin-related toxicities were reported. This case report clearly indicates that carboplatin can be safely administered in HD patients.

Toxicity

In general, platinum-based chemotherapeutics are associated with numerous and potentially severe side effects. Most common side effects include nausea, bone marrow suppression, and electrolyte disturbances.

Kidney toxicity

Although carboplatin is a nephrotoxic drug, its nephrotoxicity is much lower compared with cisplatin. Carboplatin is significantly less nephrotoxic than carboplatin because of its enhanced stability; this is mainly related to the fact that carboplatin has carboxylate and cyclobutane moieties in the cis position, rather than chloride. In a study involving patients treated with carboplatin in combination with vincristine, a 19% decrease in CrCl was observed as early as from the second course. In contrast, in another study comparing patients treated with carboplatin alone or in combination with etoposide and bleomycin, no significant change in kidney function before and after (1 month, < 3 months, and > 3 months) treatment with carboplatin was noted. AKI has been anecdotally reported in patients who have been treated with intraperitoneal carboplatin, in association with high dose carboplatin (1500–2000 mg/m 2 ) , or in combination with ifosfamide and etoposide. In this setting, AKI occurs within days after carboplatin administration and is often only partially reversible.

In a single center study including 50 children with Wilms tumors, treated between 2002 and 2012 and followed for 2 years, it was reported that patients treated with cyclophosphamide and carboplatin were at higher risk of kidney function deterioration. Skinner et al. evaluated the long-term nephrotoxicity of platinum-based chemotherapy in 63 childhood cancer survivors (27 cisplatin, 24 carboplatin, and 12 both cisplatin and carboplatin). There was no significant overall change in kidney function over time in any treatment group except for a slight reduced median GFR (84 mL/min/1.73 m 2 ) and serum magnesium level (0.68 mmol/L) in the cisplatin-treated group. At 10 years, reduced GFR (< 60 mL/min/1.73 m 2 ) and need for magnesium supplements were present in 11% and 7% of cisplatin-treated patients, respectively. In the patients treated with carboplatin, older age was associated with lower GFR during follow-up, higher cumulative carboplatin dose associated with lower serum magnesium levels at 1 year, and increased nephrotoxicity at 1 and 10 years. Hypomagnesemia is a frequent complication in childhood cancer survivors (7%–29%). Both cisplatin and carboplatin are associated with the development of Fanconi syndrome. The occurrence of hypomagnesemia after carboplatin administration has been widely reported. In one study, 6 months following carboplatin therapy, hypomagnesemia was observed in 15.6% of the children treated for solid tumors. In combination with the antiepidermal growth factor receptor monoclonal antibody, cetuximab (which also causes hypomagnesemia ), grade 3 to 4 hypomagnesemia occurred in 3% to 7.5% in lung cancer patients. , Also in combination with proton pump inhibitors, carboplatin is associated with the development of severe hypomagnesemia. Hypokalemia and hyponatremia may also rarely occur after carboplatin administration. , , Carboplatin has only rarely been associated with the development of TMA and most often in combination with other chemotherapeutics (gemcitabine, cyclophosphamide and thiotepa, docotaxel and trastumab). Finally, there are case reports concerning obstructive AKI caused by bladder obstruction by blood clots in two patients with ovarian cancer treated with carboplatin, in combination with paclitaxel. ,

Prevention of nephrotoxicity of platinum-based chemotherapeutics through sufficient hydration is essential. In patients with preexisting kidney impairment secondary to cisplatin, the effectiveness of hydration was shown in patients treated with carboplatin (800 mg/m 2 ) with or without hydration (250 mL/h of isotonic saline 3 hours before and after carboplatin infusion). Without hydration, a decrease in CrCl of 36% to 61% was noted versus no reduction in patients receiving hydration.

Cisplatin

Cisplatin, the best known of the platinums, is widely used to treat testicular cancer, germ cell cancer, ovarian cancer, cervical cancer, breast cancer, bladder cancer, head/neck cancer, esophageal cancer, lung cancer, mesothelioma, brain tumors, and neuroblastoma. All platinum-based chemotherapeutics have been associated with early nephrotoxicity. Although carboplatinum and oxaliplatin have fewer side effects compared with cisplatin, cisplatin is still used extensively, because it is more potent in some cancers, resulting in better survival rates. ,

Pharmacokinetics

Cisplatin is nonenzymatically transformed into multiple metabolites after its administration. Cisplatin and its metabolites are removed from the body through the kidney (20%–80% within 24 hours) and the dose has to be reduced in patients with a decreased CrCl. In clinical trials, it is commonly required that patients receiving cisplatin have a serum creatinine of less than 2.0 mg/dL or a CrCl of 60 mL/min or higher. We suggest a 25% dose reduction for CrCl 46 to 60 mL/min and a 50% dose reduction for CrCl 30 to 45 mL/min. Cisplatin has been administered to patients undergoing HD , , and 50% to 75% and 50% dose reductions are suggested for patients undergoing HD and PD, respectively. , , HD should be performed within 3 hours after administration of cisplatin. Furthermore, it has been suggested to consider carboplatin instead of cisplatin in patients with CrCl less than 45 mL/min.

Mechanism of action

The main mechanism of action of cisplatin is the binding of DNA and inhibition of DNA replication ( Fig. 16.1 ). Inhibition of DNA replication mainly affects rapidly proliferating cells. After administration, cisplatin undergoes aquation intracellularly, in which one of the two chloride ligands is replaced by H 2 O. This aqua ligand is thereafter displaced by a DNA base, preferentially guanine. Cross-linking of DNA interferes with mitosis and results in apoptosis.

Fig. 16.1, Cisplatin nephrotoxicity. Cisplatin enters the proximal tubular cells through organic cation transporter-2 (OCT2) and accumulates in the cell cytoplasm. Cisplatin-mediated cellular injury involves several mechanisms. Cisplatin induces endoplasmic reticular stress (ER stress) and production of reactive-oxygen species (ROS), resulting in apoptosis. Moreover, cisplatin induces mitochondrial dysfunction, which is also associated with caspase-independent and caspase-dependent apoptosis. Moreover, cisplatin induces deoxyribonucleic acid (DNA) damage with phosphorylation and activation of p53. p53 subsequently induces transcription of apoptic genes such as PUMA- α and PIDD . PIDD forms the PIDDosome and activation of caspase 2 and caspase-independent apoptosis. PUMA-α neutralizes Bcl-XL, resulting in mitochondrial dysfunction and caspase-dependent apoptosis. Finally, cisplatin activates the death receptor pathway apoptosis pathway. Cisplatin also induces inflammation through the production of tumor necrosis factor (TNF)α and other chemokines. MRP , Multidrug-resistant protein; OAT , organic anion transporter; OCT , organic cation transporter; Pgp, P glycoprotein.

Toxicity

The most commonly reported side effects associated with cisplatin include nephrotoxicity, nausea/vomiting, bone marrow suppression and hemolytic anemia, ototoxicity, and neurotoxicity.

Kidney toxicity

As far as the clinical manifestations of cisplatin kidney injury, AKI has been reportedly observed in approximately 20% to 30% of patients, but the more common one is hypomagnesemia, observed between 40% and 100% of the cases. Other rarer, but not less challenging, manifestations include Fanconi-like syndrome, distal renal tubular acidosis (RTA), renal concentrating defect, and TMA. Cisplatin-induced nephrotoxicity mostly affects the corticomedullary S3 segment of the proximal tubule and to a lesser degree the loop of Henle and distal tubular segments. In proximal tubular cells in the S3 segment, cisplatin accumulates to a great degree, resulting in a cisplatin concentration 5 times higher than the serum concentration. After cisplatin enters the tubular cell, it appears to undergo a complex series of reactions that results in the formation of more potent (toxic) metabolites. However, it is unclear if all cisplatin-induced cytotoxic effects are mediated via its activated metabolites. Once within the tubular cell, cisplatin exerts many different biological effects, which culminate in cell apoptosis/necrosis (see Fig. 16.1 ). Although cisplatin targets nuclear DNA, it has been reported that only 1% of the cytosolic cisplatin is present within the nucleus. Multiple cellular targets have been identified for cisplatin toxicity, including: (1) DNA damage; (2) cytoplasmic organelle dysfunction, with endoplasmic reticulum stress and mitochondrial dysfunction; (3) apoptotic pathways both caspase-dependent and death receptor-mediated; (4) oxidative stress with formation of reactive oxygen species; and (5) inflammation mediated via tumor necrosis factor and other chemokines. In addition, there also appears to be an immune component to cisplatin-induced nephrotoxicity. , Several nonmodifiable factors have been identified to increase the risk for cisplatin-induced nephrotoxicity, such as genetic factors, race, gender, age, cardiac disease, malnutrition, dehydration, comorbidities such as diabetes or cirrhosis (hypoalbuminemia is associated with a higher unbound fraction of cisplatin, resulting in greater peak plasma concentrations), concomitant use of other nephrotoxic agents, or preexisting kidney diseases.

It is well known that cisplatin-induced kidney injury is dose-, duration-, and frequency-dependent. , Higher peak plasma concentrations result in greater injury. Furthermore, a higher cumulative dose has also been shown to increase the risk for future kidney injury. Early trials with cisplatin reported that greater than 70% of patients developed dose-related AKI. In more recent trials, at high cisplatin doses, 42% of treated patients had nephrotoxic injury. A recent study reported cisplatin-induced nephrotoxicity to occur in more than one-third of patients after the fourth cycle of chemotherapy and preventive strategies were ineffective. In a meta-analysis of randomized phase 2 and 3 clinical trials comparing first-line platinum-based chemotherapy with the same regimen without platinum, platinum was associated with a significant increase in nephrotoxicity (18 trials; 4384 patients; odds ratio, 3.09; 95% confidence interval, 1.88–5.06; p < .0001). With improved survival in cancer patients, it has become evident that cytotoxic chemotherapeutics can also result in CKD. In childhood cancer, loss of GFR occurs, especially in older children treated with ifosfamide and higher doses of cisplatin. , The long-term effect of cisplatin on kidney function in 859 adult patients treated with cisplatin who had survived 5 years or more after initial dose showed most patients experienced small but permanent declines in eGFR, although none progressed to ESRD requiring HD.

Cisplatin is associated with the development of TMA either as monotherapy , or in combination with other chemotherapeutics (with bleomycin and vincristine/vinblastine, , with bleomycin and epirubicin, , with bleomycin and methotrexate, with gemcitabine, with 5-fluorouracil [5-FU], with cyclophosphamide and adriamycin ). TMA can develop up to 4 months after the last administration of cisplatin. There is no established treatment for cisplatin-induced TMA as variable efficacies have been described for plasma exchange and infusion of fresh frozen plasma, aspirin, and dipyridamole.

Clinical practice guidelines have been published on the prevention of cisplatin-induced kidney injury. , , Different preventive strategies have been tested to reduce cisplatin-induced nephrotoxicity. A recent systematic review concluded that: (1) hydration is essential for all patients; (2) short-duration, low-volume, outpatient hydration regimens appear to be safe and feasible, even in patients receiving intermediate- to high-dose cisplatin; (3) magnesium supplementation (8–16 mEq) may limit cisplatin-induced nephrotoxicity; and (4) mannitol might be considered for high-dose cisplatin regimens and/or for patients with preexisting hypertension. These findings have broad implications for clinical practice and represent best practice principles for the prevention of cisplatin-induced nephrotoxicity. In animal models, amifostine (a free radical scavenger) ameliorates cisplatin-mediated nephrotoxicity. However, cisplatin-induced nephrotoxicity cannot be completely prevented with current available measures and several aspects regarding optimal management are still controversial, such as duration of hydration, use of magnesium, and use of diuretics. , Diuretics should probably be avoided in patients receiving platinum-based chemotherapy, because they are potentially harmful. Cisplatin is transported into the tubular cell by the basolateral organic cation transporter 2 (OCT2) channel; therefore blockers of the OCT2 channel have potential to prevent/reduce nephrotoxicity. A recent trial demonstrated that proton pump inhibitors (which are known blockers of the OCT2 channel) are associated with reduced cisplatin-mediated nephrotoxicity.

Oxaliplatin

Oxaliplatin is a member of platinum-based chemotherapeutics. Oxaliplatin is used to treat colorectal cancer (and other gastrointestinal cancers) and is often combined with 5-FU and leucovorin.

Mechanism of action

As other platinum-based chemotherapeutics, its action is through the blockade of DNA production. Oxaliplatin forms both inter- and intrastrand cross-links in DNA, causing cell death. However, recently it was reported that oxaliplatin does not only kill cells through DNA damage response, but also by inducing ribosome biogenesis stress. This might explain the distinct clinical implementation of oxaliplatin compared with other platinum-based chemotherapeutics.

Pharmacokinetics

Oxaliplatin undergoes extensive nonenzymatic biotransformation and platinum is mainly excreted by the kidneys. Approximately half of the total dose is recovered in the urine by day 5. Dose reduction is recommended in patients with a CrCl less than 20 mL/min. Doses up to 130 mg/m 2 every 3 weeks proved to be well tolerated in patients with CrCl greater than 20 mL/min and do not require dose reduction. Some experts recommend oxaliplatin dose reduction in patients with ESRD undergoing HD. Although others suggest this is not necessary if HD is performed shortly after drug administration, and the dosing interval is extended to 3 weeks.

Toxicity

Common side effects include neurotoxicity (peripheral neuropathy), fatigue, nausea and vomiting, neutropenia, nephrotoxicity, and ototoxicity (less than cisplatin and carboplatin). Other serious side effects include allergic reactions and rhabdomyolysis.

Kidney toxicity

Oxaliplatin is less nephrotoxic than cisplatin and carboplatin, but all platinum-based chemotherapeutics cause proximal tubular cell damage. There are few cases of AKI associated with oxaliplatin-induced immune-mediated intravascular hemolysis and the occurrence of acute tubular necrosis (ATN). , In these reported cases, the evolution was favorable with recovery of kidney function. Another well-known side effect of oxaliplatin is the development of hypokalemia during treatment. , In a prospective study of 772 patients treated with oxaliplatin, seven patients developed hypokalemia rapidly after oxaliplatin administration. It is suggested that oxaliplatin shifts potassium into cells. Oxaliplatin has also been associated with the development of TMA. Data from the Oklahoma thrombotic thrombocytopenic purpura-hemolytic uremic syndrome (TTP-HUS) registry and BloodCenter of Wisconsin concluded that there was a definite association between oxaliplatin and the occurrence of TMA. It has been suggested that TMA associated with oxaliplatin is immune mediated. Prevention of platinum-based chemotherapeutic-related nephrotoxicity is based on intravenous (IV) administration of isotonic saline fluids and treatment of nausea and vomiting.

Alkylating agents

Alkylating agents were among the earliest anticancer drugs, dating back to the 1940s. Alkylating agents react with electron-rich atoms to form covalent bonds. The common alkane transferred by classical alkylating agents is a single-carbon methyl group that also includes longer hydrocarbons. The most important reactions with regard to the agents’ antitumor activities are reactions with DNA bases; indeed, some alkylating agents are monofunctional and react with only one strand of DNA, whereas others are bifunctional and react with an atom on both strands of DNA, producing a cross-link that covalently links the two strands of the DNA double helix. Unless repaired, this lesion will prevent the cell from replicating effectively.

Busulfan

Busulfan is part of the conditioning regimen for bone marrow transplantation in patients with leukemia, lymphoma, and myeloproliferative disorders.

Mechanism of action

Busulfan is an alkylating chemotherapeutic.

Pharmacokinetics

Busulfan is extensively metabolized in the liver and these metabolites are predominantly excreted as inactive metabolites by renal excretion. No dose reductions are recommended in patients with renal dysfunction.

Toxicity

Busulfan-associated side effects include interstitial pulmonary fibrosis, seizures, hepatic veno-occlusive disease, and wasting syndrome.

Kidney toxicity

The nephrotoxicity of busulfan seems to be limited. In a phase I study, only one of 15 refractory acute leukemia patients received busulfan and clofarabine, as preparation for allogeneic hematopoietic stem cell transplantation, developed AKI (attributed to clofarabine).

Cyclophosphamide

Cyclophosphamide is used to treat lymphoma, multiple myeloma, leukemia, ovarian cancer, breast cancer, small cell lung cancer, neuroblastoma, and sarcoma.

Mechanism of action

Cyclophosphamide is an alkylating agent and a member of the nitrogen mustard family. It acts through interfering with DNA replication and ribonucleic acid (RNA) production. The active metabolite of cyclophosphamide is phosphoramide mustard. This metabolite is only formed in cells that have low levels of aldehyde dehydrogenase activity. Phosphoramide mustard forms DNA inter- and intrastrand cross-links, which leads to cell apoptosis.

Pharmacokinetics

When administered orally, cyclophosphamide is rapidly absorbed and converted to its active metabolites in the liver. Several metabolites are produced: 4-hydroxycyclophosphamide, aldophosphamide, carboxycyclophosphamide ( Fig. 16.2 ). A fraction of aldophosphamide diffuses into cells and is there metabolized into phosphoramide mustard (the active metabolite of cyclophosphamide) and acrolein. Aldophosphamide is oxidized to carboxycyclophosphamide by the enzyme aldehyde dehydrogenase, and the formation of phosphoramide mustard only occurs in cells with low aldehyde dehydrogenase activity. Similarly, it has been demonstrated that high aldehyde dehydrogenase activity results in cyclophosphamide resistance in certain leukemia cell lines. Conversion of aldophosphamide to carboxycyclophosphamide by aldehyde dehydrogenase prevents toxicity as carboxycyclophosphamide does not result in nitrogen mustard formation and alkylation.

Fig. 16.2, Ifosfamide nephrotoxicity. Ifosfamide is transported in the proximal tubular cells through organic cation transporter-2 (OCT2). Ifosfamide undergoes substantial metabolization with the production of acrolein (responsible for bladder irritation and hemorrhagic cystitis) and chloroacetaldehyde (responsible for development of proximal tubulopathy). In this figure, the metabolism of ifosfamide is represented in the renal tubular cells, while this happens mainly in liver cells. FR-α , folate receptor-alpha; RFC, reduced folate carrier.

Some 30% of the cyclophosphamide dose is excreted in unchanged form in the urine, whereas highly protein-bound metabolites are primarily excreted through the kidney. Cyclophosphamide pharmacokinetics are clearly altered in patients with renal insufficiency. However, there is no consensus whether cyclophosphamide dose adjustments are needed in patients with reduced kidney function, because studies are unable to demonstrate an association between kidney function and cyclophosphamide clearance or hematologic toxicity. , We recommend no dose reduction, a 25% dose reduction, and a 50% dose reduction in patients with a CrCl greater than 20 mL/min, 10 to 20 mL/min, and less than 10 mL/min, respectively. Cyclophosphamide is moderately hemodialyzable and should always be administered after HD and HD held until 12 hours later. , In PD patients, a 25% dose reduction has been suggested. ,

Toxicity

Common adverse effects include low white blood cell counts, nausea/vomiting, hair loss, and hemorrhagic cystitis. Other severe adverse effects include increased future risk of cancer, infertility, allergic reactions, and pulmonary fibrosis. Cardiotoxicity occurs mainly with high cumulative doses of cyclophosphamide. Infertility has been reported in both females and males, and the risk for infertility increases with both cumulative dose of cyclophosphamide and patient age.

Kidney toxicity

Cyclophosphamide nephrotoxicity includes syndrome of inappropriate antidiuretic hormone secretion (SIADH) with hyponatremia, and hemorrhagic cystitis. Cyclophosphamide might either directly induce SIADH or stimulate ADH through cyclophosphamide-induced nausea. , Hyponatremia typically occurs with high dose IV cyclophosphamide but has also been reported after oral cyclophosphamide or low-dose IV cyclophosphamide. Hyponatremia often resolves after drug discontinuation, but high-dose IV cyclophosphamide and concomitant IV fluids to prevent hemorrhagic cystitis, the combination of SIADH, and enhanced water administration can lead to severe, occasionally fatal hyponatremia. Therefore it is recommended to administer isotonic saline rather than hypotonic solutions.

The cyclophosphamide metabolite acrolein is highly toxic to the bladder epithelium and may cause hemorrhagic cystitis. The risk of hemorrhagic cystitis can be minimized by administration of fluids and sodium 2-mercaptoethane sulfonate (mesna), which binds and inactivates acrolein. ,

Diaziquone

Diaziquone is mainly used in patients with primary brain tumors.

Mechanism of action

Diaziquone is an aziridinylbenzoquinone, which rapidly penetrates the central nervous system. The most probable mechanism of action is that of bioreductive alkylation.

Pharmacokinetics

After administration, diaziquone is rapidly absorbed and it has a volume of distribution exceeding that of total body water. Diaziquone is rapidly metabolized by the liver.

Toxicity

The main toxicity associated with diaziquone is bone marrow suppression resulting in leukopenia, granulocytopenia, and thrombocytopenia.

Kidney toxicity

Data regarding diaziquone-related nephrotoxicity are very limited, although acute renal failure has been reported subsequent to treatment with diaziquone.

Ifosfamide

Ifosfamide is used in the treatment of Hodgkin and non-Hodgkin lymphoma, soft tissue and osteosarcoma, lung cancer, testicular cancer, breast cancer, ovarian cancer, and cervical cancer.

Mechanism of action

Ifosfamide is an alkylating agent and a member of the nitrogen mustard family. It is believed to act through interfering with DNA replication and RNA production.

Pharmacokinetics

Ifosfamide is primarily excreted in the urine (80% to the total dose as unchanged ifosfamide). Guidelines vary substantially in regard to dosing in patients with kidney failure. We recommend a 25% dose reduction in patients with a CrCL 40 to 60 mL/min, a 50% dose reduction for patients with a CrCl 10 to 40 mL/min, and drug discontinuation in patients with CrCl lower than 10 mL/min. Ifosfamide is dialyzable, and it should be administered after HD with a dose reduction of 50%. The same dose reduction is recommended for PD patients. No dialysis should be performed for 12 hours after ifosfamide administration.

Toxicity

Common side effects include hair loss, vomiting, nephrotoxicity, neurotoxicity (encephalopathy and peripheral neuropathy), and bone marrow suppression.

Kidney toxicity

Ifosfamide is associated with numerous possible adverse kidney manifestations; AKI caused by ATN, Fanconi syndrome, , interstitial nephritis, glomerular disease, and hemorrhagic cystitis. Ifosfamide-induced tubular toxicity can be associated with metabolic acidosis with a normal anion gap (hyperchloremic acidosis) because of type 1 (distal) or type 2 (proximal) RTA; hypophosphatemia induced by decreased proximal phosphate reabsorption; renal glucosuria, aminoaciduria, and a marked increase in β 2 -microglobulin excretion, all from generalized proximal dysfunction; polyuria caused by nephrogenic diabetes insipidus; and hypokalemia, resulting from increased urinary potassium losses. Ifosfamide is more nephrotoxic than cyclophosphamide and this is caused by selective uptake of ifosfamide in proximal tubular cells through the OCT2 (see Fig. 16.2 ). Several risk factors for ifosfamide-induced nephrotoxicity have been identified including preexisting kidney disease, combination with platinum-based chemotherapy and/or other nephrotoxins, cumulative dose of ifosfamide (> 119 g/m 2 ), and renal irradiation. Although there are few data on long-term kidney function in ifosfamide-treated adults, a progressive decline in GFR has been described after as little as one course of ifosfamide in adults. This reduction in kidney function occurs in a minority of patients, is permanent and progressive, and can also occur long after exposure to ifosfamide. In a Dutch study, it was demonstrated that patients who received ifosfamide in childhood had a lower GFR than patients with the same pathologies who did not receive this treatment. The main risk factors for nephrotoxicity in children are a cumulative dose greater than 45 mg/m 2 , young age (< 3 years), previous or concurrent cisplatin treatment, Wilms tumor, and unilateral nephrectomy. Hemorrhagic cystitis can occur even when mesna is used.

Tubular involvement is generally very prolonged, potentially progressive and may lead to advanced CKD. , Ifosfamide-induced proximal tubular toxicity is characterized by aminoaciduria (28%), glucosuria (90%), low molecular weight proteinuria, Fanconi syndrome (1%–7%), hypophosphatemia, proximal RTA, hypokalemia, phosphaturia, and more rarely, calciuria, magnesiuria, and natriuria. Distal tubular toxicity has also been reported with type 1 RTA, a defect in urine concentration, and nephrogenic diabetes insipidus. Ifosfamide can induce SIADH characterized by hyponatremia, plasma hypoosmolality, and inadequate urinary osmolality. ,

Mesna and hydration are the main measures to minimize the urogenital and nephrologic involvement of ifosfamide, but N-acetylcysteine is recommended by some. , Patients who received ifosfamide should be followed long-term to detect CKD early.

Melphalan

Melphalan is an alkylating agent and is used in the treatment of multiple myeloma.

Mechanism of action

Melphalan is a member of the nitrogen mustard alkylating agent family. Melphalan alkylates guanine, which results in inter- and intrastrand links of DNA. This results in the inhibition of DNA and RNA synthesis and cytotoxicity.

Pharmacokinetics

Melphalan is metabolized primarily in the liver, and 10% to 30% is eliminated by the kidneys. Melphalan is both secreted and reabsorbed in the renal tubules. In patients with decreased kidney function, elimination of melphalan is reduced and systemic exposure is increased, which may result in bone marrow suppression. Recommendations regarding dose reduction for decreased kidney function differ substantially. We recommend a 25% dose reduction in patients with CrCl between 10 and 50 mL/min and a 50% dose reduction with CrCl less than 10 mL/min. Melphalan is removed during dialysis and should be administered at 50% of the dose after HD.

Toxicity

The most common side effects are nausea, vomiting, and bone marrow suppression.

Kidney toxicity

Nearly 19% of 80 consecutive patients with AL amyloidosis treated with high-dose melphalan followed by peripheral blood stem cell transplantation developed AKI. Patients who developed AKI had a worse 1-year survival ( p =.03). A few case reports note nephrotic syndrome after melphalan administration. , During 1-year follow-up, kidney function improved with disappearance of the nephrotic syndrome. Finally, melphalan has been associated with the development of SIADH. ,

Procarbazine

Procarbazine is used, in combination with other chemotherapeutics, to treat Hodgkin lymphoma and brain cancers, such as glioblastoma multiforme.

Mechanism of action

Procarbazine is an alkylating agent. Its mechanism of action is incompletely understood but involves inducing breaks in DNA strands.

Pharmacokinetics

Oral procarbazine is rapidly metabolized in the liver. After 24 hours, up to 70% of the dose is recovered in the urine. The use of procarbazine is not recommended in patients with severe kidney impairment, and a 50% dose reduction is recommended with a serum creatinine greater than 2.0 mg/dL.

Toxicity

Common side effects include: bone marrow suppression, nausea, vomiting, peripheral neuropathy, and fatigue. In combination with ethanol, procarbazine may cause a disulfiram-like reaction.

Kidney toxicity

Procarbazine monotherapy is not nephrotoxic. However, renal insufficiency after administration of procarbazine and high doses of methotrexate has been reported. The authors suggested procarbazine augmented the nephrotoxicity of methotrexate.

Temozolomide

Temozolomide is an alkylating agent used in the treatment of brain tumors (astrocytoma and glioblastoma multiforme) and melanomas, at least before the area of targeted agents and immunotherapy.

Mechanism of action

Temozolomide is an alkylating agent that alkylates/methylates guanine residues, resulting in DNA damage and cell death.

Pharmacokinetics

Temozolomide is administered orally and the kidney is the major route of elimination (5%–10% as unchanged temozolomide and 90–95% as metabolites). However, studies in patients with mild to moderate kidney dysfunction showed unaltered pharmacokinetics, suggesting no dose adjustment is required. Experience with temozolomide in HD patients is very limited.

Toxicity

The main toxicity associated with the use of temozolomide is bone marrow suppression.

Kidney toxicity

Temozolomide nephrotoxicity appears to be very limited. Few AKI cases were described with temozolomide treatment. Only one of 42 patients treated with temozolomide and irinotecan died after developing AKI.

Trabectedin

Trabectedin is an alkylating agent used to treat soft tissue sarcoma and ovarian cancer.

Mechanism of action

Trabectedin is an alkylating agent that binds DNA at the N2 position of guanine promoting degradation of RNA polymerase and generating DNA double-strand breaks. Furthermore, trabectedin blocks DNA binding of the transcription factor FUS-CHOP and reverses the oncogenic phenotype of liposarcoma cells.

Pharmacokinetics

Dose adjustments are not recommended in patients with CrCl greater than 30 mL/min and no data are available in patients with more severe kidney disease. Therefore the drug, which is not dialyzable, should not be used in patients with CrCl lower than 30 mL/min. ,

Toxicity

Adverse effects associated with trabectedin include bone marrow suppression, hepatotoxicity, nausea, vomiting, and fatigue.

Kidney toxicity

Cases of AKI (occasionally fatal) have been reported, some of which are attributable to rhabdomyolysis. Furthermore, elevations in serum creatinine grade 3 or 4 were observed in 4.2% of patients treated with trabectedin for liposarcoma or leiomyosarcoma. Nephrotoxicity complicated by hepatic failure and death was reported in a 79-year-old patient with sarcoma after a second course of trabectedin.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here