Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Clinical Practice: Review Article

Nephron 2023;147:65–77 Received: January 18, 2022


Accepted: April 24, 2022
DOI: 10.1159/000525029 Published online: June 17, 2022

Onconephrology: Update in Anticancer


Drug-Related Nephrotoxicity
Clara García-Carro a Juliana Draibe b María José Soler c
aNephrology
Department, San Carlos Clinical University Hospital, Madrid, Spain; bNephrology Department, Bellvitge
Hospital, Hospitalet Llobregat, Barcelona, Spain; cNephrology Department, Vall d’Hebron University Hospital,

Downloaded from http://karger.com/nef/article-pdf/147/2/65/3915619/000525029.pdf by guest on 22 June 2023


Vall d’Hebron Institute of Research, CSUR National unit of Expertise for Complex Glomerular diseases of Spain,
Barcelona, Spain

Keywords therapy. In addition, strategies for prevention and manage-


Onconephrology · Immunotherapy · Chemotherapy · ment recommendations in patients with malignancies and
Kidney injury · Cancer kidney disease will also be addressed.
© 2022 S. Karger AG, Basel

Abstract
The relation that connects cancer and renal damage is bidi- Introduction
rectional and this renal damage worsens quality of life and
increases morbidity in high-complexity patients such as pa- Cancer is one of the leading causes of death and in-
tients with cancer and kidney injury. Strikingly, in the last creased morbidity in the world [1]. In the last decade, new
decade, the treatment of advanced cancer has clearly ad- therapies have changed life expectancy in patients with
vanced in terms of new therapeutic strategies with the abil- malignancy, however, as well as classical chemotherapy
ity to transform the advanced metastatic cancer in a chronic agents, they are not free of adverse events. Renal function
condition. In this new era of cancer therapies, cancer treat- impairment and nephrotoxicity are one of the major lim-
ment including conventional chemotherapy, targeted can- itations of cancer treatment. The relation that connects
cer agents and immunotherapies among others are signifi- cancer and renal damage is bidirectional and this renal
cantly associated with kidney injury. Renal toxicity that is damage increases mortality and morbidity in patients
currently seen in onconephrology departments is in part re- with cancer. Patients with cancer who develop acute kid-
lated to the new therapies such as immunotherapy, and to ney injury (AKI) during oncologic treatments and pa-
the prolonged survival achieved at the expense of increasing tients with chronic kidney disease (CKD) who develop
therapy lines, and a combination of different drugs. In this cancer are frequently medicated with suboptimal thera-
review, we will discuss in a practical way, nephrotoxicity
caused by the main oncospecific treatments such as classical Clara García-Carro and Juliana Draibe contributed equally to this
chemotherapy agents, targeted therapies, and immuno- work.

Karger@karger.com © 2022 S. Karger AG, Basel Correspondence to:


www.karger.com/nef María José Soler, mjsoler01 @ gmail.com
Table 1. Conventional chemotherapy and nephrotoxicity

Medication Nephrotoxicity Mechanism of nephrotoxicity Prevention and management


recommendations

Platinum compounds ATI Direct tubular toxicity Hydration


(cisplatin, carboplatin, Hypomagnesemia Magnesium supplementation
oxaliplatin) Nephrogenic diabetes insipidus Forced diuresis with mannitol
Proximal tubulopathy
TMA
Ifosfamide ATI Mitochondrial tubular toxicity No current strategy for preventing
Proximal tubulopathy ifosfamide nephrotoxicity
CKD NAC: some beneficial effects
Pemetrexed ATI Antifolate effect that impairs DNA Leucovorin therapy
Proximal tubulopathy and RNA
Nephrogenic diabetes insipidus

Downloaded from http://karger.com/nef/article-pdf/147/2/65/3915619/000525029.pdf by guest on 22 June 2023


Methotrexate ATI Direct tubular toxicity Volume filling/urine alkalization (pH >7.5)
Crystalline nephropathy Induction of oxygen free radical Leucovorin therapy
formation Prolonged hemodialysis
Glucarpidase
Gemcitabine TMA Endothelial injury Eculizumab
Microvascular thrombosis
Mitomycin C TMA Endothelial injury Drug discontinuation
Microvascular thrombosis Supportive care

ATI, acute tubular Injury; CKD, chronic kidney injury; TMA, thrombotic microangiopathy; NAC, N-acetylcysteine.

peutic choices because of the lack of randomized clinical by nephrology specialists. With the aim to avoid CKD
trials in population with impaired renal function [2, 3], and/or renal dysfunction progression in patients with po-
this fact has a probably deleterious impact in survival out- tentially curable malignancies, nephrologists should be
comes. With the aim to identify, treat, and, if possible, able to manage renal toxicities of oncospecific drugs. In
prevent kidney damage in patients with cancer, the sub- this review, we will discuss in a practical way, nephrotox-
specialist of onconephrology has been created in the last icity caused by main oncospecific treatments: classical
years [4]. chemotherapy agents, targeted therapies, and immuno-
New oncospecific therapies have achieved dramatical- therapy. Furthermore, prevention and management rec-
ly better survival outcomes in different types of cancer; ommendations in patients with malignancies and kidney
however, renal toxicity seems not to be decreasing. Renal disease are also addressed.
toxicity that is nowadays seen in onconephrology depart-
ments is in part related to the new therapies such as im-
munotherapy, and to the prolonged survival achieved at Conventional Chemotherapy
the expense of increasing therapy lines, and a combina-
tion of different drugs. The admission of elderly patients As it is already known, conventional chemotherapy
with many comorbidities, such as hypertension (HTN) or has been classically associated with AKI; caused primar-
diabetes mellitus, and baseline renal dysfunction, to on- ily by acute tubular toxicity, acute tubulointerstitial ne-
cospecific and potentially nephrotoxic treatments, could phritis (ATIN), and different glomerulonephritis [5–7].
also in part play an important role in the development of It is frequently related with drug dosage and treatment
kidney toxicity. duration. Briefly, we will assess the more important che-
Currently, in this new onconephrology scenario, on- motherapeutic agents associated with renal toxicity (see
cologic patients with renal disease are commonly treated Table 1).

66 Nephron 2023;147:65–77 García-Carro/Draibe/Soler


DOI: 10.1159/000525029
Downloaded from http://karger.com/nef/article-pdf/147/2/65/3915619/000525029.pdf by guest on 22 June 2023
Fig. 1. Glomerular and tubular patterns of anticancer drug-related kidney injury.

Platinum Compounds (Cisplatin, Carboplatin, nephrotoxic than cisplatin but can still cause AKI in pa-
Oxaliplatin) tients with lower albumin (higher unbound fraction of
Cisplatin is a widely used and highly effective cancer platinum resulting in greater peak plasma concentra-
chemotherapeutic agent. It has been mainly related to re- tions), preexisting kidney damage, and concurrent use of
versible AKI approximately 3–5 days after drug exposure, nephrotoxic medications (like nonsteroidal anti-inflam-
although repeated doses (>100 mg/m2) may cause a per- matory drugs NSAIDS, iodinated contrast) [13].
manent kidney injury [5–7]. Besides AKI, cisplatin has
been also associated with hypomagnesemia [8], nephro- Ifosfamide
genic diabetes insipidus [9], proximal tubulopathy [10], Ifosfamide is an alkylating drug; it is often combined
and less frequently with thrombotic microangiopathy with cisplatin for the treatment of sarcomas, testicular tu-
[11] (see Fig. 1). Appropriate knowledge regarding the mors, and some refractory lymphomas. Its renal toxicity
pharmacokinetics and toxicological profile of cisplatin has been associated with proximal tubular dysfunction
may help physicians prevent renal toxicity. Accurate hy- (Fanconi Syndrome) and AKI, especially when combined
dration remains the main fundamental strategy for re- with platinum drugs, and can progress to CKD [14, 15].
ducing the risk of cisplatin-induced nephrotoxicity. In- When ATN occurs in the context of ifosfamide treatment,
terestingly, magnesium supplementation may have a role a special histological feature might be found by optical
as a nephroprotective agent, and forced diuresis with microscope the presence of karyomegalic cells. Karyome-
mannitol may be appropriate in some patients receiving galic changes in tubular epithelial cells result from aber-
cisplatin [12]. rant cell division related to exposure to the toxic ifos-
Carboplatin and oxaliplatin, are the second and third famide, secondarily inducing interstitial inflammation
generation of platinum agents, respectively. They are less [15]. In this setting, Akilesh et al. [16] described intersti-

Anticancer Drug-Related Nephrotoxicity Nephron 2023;147:65–77 67


DOI: 10.1159/000525029
tial fibrosis and tubular atrophy in patients treated with Mitomycin C
ifosfamide, whereas several biopsies also contained inter- Mitomycin C is also related with dose-dependent
stitial inflammation. TMA. Drug discontinuation and supportive care are in-
dicated in that case, as plasmapheresis is ineffective [27].
Pemetrexed Gemcitabine, mitomycin C, and platinum compounds
Pemetrexed is a methotrexate analog that inhibits 3 were categorized by Izzedine and Perazella [28] as “Type
enzymes involved in folate metabolism and thus pu- I” cancer drug TMA and anti-vascular endothelial growth
rine/pyrimidine production needed for DNA synthesis. factor (VEGF) agents (discussed below) as “Type II” can-
Regarding its renal toxicity, it has been associated with cer drug TMA, on the basis of their different mechanisms
AKI, proximal tubulopathy, and nephrogenic diabetes of kidney injury and outcomes. In type I, endothelial in-
insipidus. Renal histology reveals ATN with mild-to- jury and/or microvascular thrombosis are involved in the
moderate interstitial fibrosis and, in a minority of cases, pathogenesis; it has been related to cumulative drug dose,
mild interstitial inflammation [17, 18]. Pemetrexed is may present many months after beginning or ending
currently contraindicated when the estimated creati- treatment, and is refractory to therapy. In relation to renal
nine clearance is <45 mL/min because of fatal toxicities histology, fibrin thrombi are often found in glomerular

Downloaded from http://karger.com/nef/article-pdf/147/2/65/3915619/000525029.pdf by guest on 22 June 2023


in patients with renal impairment [19]. In that line, in- and arteriolar compartments, whereas in “Type II” only
novative interventions as leucovorin therapy, used for glomerular thrombi are seen.
methotrexate toxicity, are needed to allow a safe treat-
ment [20].
Target Cancer Agents
Methotrexate
As permetrexed, methotrexate inhibits folate metabo- Several families of transmembrane receptors or intra-
lism, specifically dihydrofolate reductase. Methotrexate cellular tyrosine kinase inhibitors (TKIs) participate in
precipitation and metabolites within tubular lumen lead the tumor growth when aberrant activation is present.
to AKI and crystalline nephropathy [7, 21]. After AKI oc- The main strategies of inhibition include inhibitory anti-
curs, the excretion of the drug is reduced and the patient bodies (bevacizumab: anti-VEGF, cetuximab: anti-epi-
can develop systemic toxicity. Several strategies as high- dermal growth factor receptor [EGFR]) or small molecu-
dose of leucovorin therapy (a folate compound), pro- lar TKIs (gefitinib/erlotinib: anti-EGFR, sunitinib: anti-
longed hemodialysis [22] (daily for several days, to avoid ALK) [29] (see Table 2).
rebound), or glucarpidase (an enzyme that inactivates the
drug) can be used when AKI and severe systemic toxici- Anti-VEGF (Aflibercept, Bevacizumab)
ties developed [23]. The hypothesis that some kind of tumor growth is de-
pendent on angiogenesis has been the rationale to devel-
Gemcitabine op antiangiogenic drugs. Antiangiogenic drugs can target
Thrombotic microangioapthy (TMA) is a rare compli- the VEGF itself (bevacizumab, aflibercept), block the
cation related with gemcitabine with a high mortality VEGF receptor (ramucirumab), or be TKIs of the VEGF
rate. It is characterized by microangiopathic hemolytic receptor [30]. TKIs of VEGF receptor will be reviewed in
anemia, thrombocytopenia, and end-organ damage. In the next section.
the largest case series of TMA related to gemcitabine, au- Bevacizumab and aflibercept are increasingly applied
thors describe new-onset or exacerbated HTN and edema to treat malignancies like metastatic or recurrent colorec-
as early signs of TMA, suggesting that in front of these tal, non-small cell lung, breast, and renal cell cancers and
clinical symptoms close monitoring may be useful [24]. ramucirumab has been approved as a second line therapy
A kidney biopsy can reveal endothelial cell swelling, fi- for gastric, non-small-cell lung, and metastatic colorectal
brin thrombi within capillary loops and arterioles, me- cancers [31]. Despite their mechanisms of action are not
sangiolysis and thickened arterioles, and glomerular cap- the same, the three drugs inhibit endothelial cell prolif-
illaries [25]. Some case reports suggest that complement eration and vessel development and can cause HTN,
inhibition with eculizumab could be a reasonable therapy asymptomatic albuminuria, nephrotic syndrome, and
when plasmapheresis failed in patients with gemcitabine- TMA. VEGF is mainly expressed in podocytes and re-
associated TMA [26]. quired to maintain the integrity of glomerular endothe-
lial function and glomerular filtration barrier. The regula-

68 Nephron 2023;147:65–77 García-Carro/Draibe/Soler


DOI: 10.1159/000525029
Table 2. Target cancer agents and nephrotoxicity

Medication Mechanism of action Nephrotoxicity Prevention and management


recommendations

EGFR pathway EGFR-TKI inhibitors (erlotinib, gefitinib) MCD Drug discontinuation


inhibitors Membranous nephropathy
anti-EGFR receptor Hypomagnesemia Drug discontinuation
Antibodies (cetuximab, panitumumab) Hypokalemia
Nephrotic syndrome
IgA glomerulonephritis
Anaplastic Crizotinib AKI (NTA and ATIN) Drug discontinuation
lymphoma kinase ALK TKI inhibitor Formation or increase of renal cysts
inhibitors c-MET, ROS1 inhibitor Hypophosphatemia
BRAF inhibitors Inhibitors of mutant BRAF (vemurafenib, Fanconi syndrome Drug discontinuation
debrafenib) Hypophosphatemia

Downloaded from http://karger.com/nef/article-pdf/147/2/65/3915619/000525029.pdf by guest on 22 June 2023


Hyponatremia
Hypokalemia
AKI (NTA and ATIN)
VEGF pathway VEGF-TKI inhibitors (axitinib, pazopanib, FSGS/MCD (sunitinib/sorafenib) Drug discontinuation
inhibitors sorafenib, regorafenib, sunitinib) TMA (sunitinib) Angiotensin-converting
NTA (sunitinib/sorafenib) enzyme inhibitor
Hypophosphatemia, hypocalcemia,
hyponatremia, hypokalemia (regorafenib)
Anti- VEGF receptor antibodies TMA Drug discontinuation
Bevacizumab: mAb against VEGFA. IgA nephropathy Angiotensin-converting
Aflibercept: Recombinant fusion protein ANCA glomerulonephritis enzyme inhibitor
(VEGF trap) Diabetic nephropathy
Ramucirumab: mAb against VEGFR2 FSGS/MCD
Membranous nephropathy
Bcr-abl TKIs 1st generation: Nilotinib, imatinib Hypophosphatemia Monitoring of renal function
(Bcr-abl TKI) ATP-competitive TKI Reversible decrease in GFR Dose reduction when
Src family kinases inhibition AKI (TLS) decrease GFR
NTA (imatinib)
2nd generation: dasatinib Proteinuria, nephrotic syndrome: Dose reduction o drug
Platelet-derived growth factor receptor (dasatinib) discontinuation
Tyrosine kinase receptor KIT (CD117) NTA (ibrutinib) Change 1st generation Bcr-
VEGF inhibition abl tyrosine kinase inhibitors
Ibrutinib

EGFR, epidermal growth factor receptor, TKI: EGFR-tyrosine kinase inhibitors; 2. BRAF, encoding gene human protein B-Raf; VEGF,
vascular endothelial growth factor; MCD, minimal change disease; FSGF, focal segmental glomerulosclerosis; AKI, acute kidney injury; ATN,
acute tubular necrosis; NTIA, acute tubulointerstitial nephritis; TMA, thrombotic microangiopathy; CKD, chronic kidney injury; TLS, tumor
lysis syndrome.

tion of susceptibility to VEGF inhibition is not well un- angiotensin receptor blockers (ARB) are the first-line
derstood, and it is unknown why some patients tolerate therapy and calcium channel blockers are the second
long-term anti-VEGF therapies and others develop TMA, choice. Diuretic agents can also be added. Normally,
mainly limited to the kidney. blood pressure can be controlled and it is not necessary
The less severe renal adverse event associated with an- to stop the cancer treatment [30, 32]. All degree of pro-
ti-VEGF is HTN, which appears in 16%–43% of patients. teinuria is more common in patients on aflibercept than
Angiotensin-converting enzyme inhibitors (ACEIs) or in those on bevacizumab (33.9% vs. 13.3%) and it is as-

Anticancer Drug-Related Nephrotoxicity Nephron 2023;147:65–77 69


DOI: 10.1159/000525029
sociated to the disruption of the glomerular filtration bar- mitogen-activated protein kinase pathway, increasing is-
rier. Treatment can be continued in most cases involving chemic tubular damage. Although there is a paucity of
non-nephrotic-range proteinuria, and HTN and protein- renal biopsy, it seems that this drug causes mainly a tubu-
uria can be aggressively managed with ACEIs or ARBs. lar injury, as ATN and ATIN, reported in some case re-
Risk factors for the development of proteinuria are base- ports. In these cases, drug discontinuation seems to lead
line elevated systolic blood pressure, number of cycles of the recovery of kidney function [43].
anti-VEGF drug, and previous use of calcium channel
blockers [33]. If possible, kidney biopsy is recommended ALK Inhibitors (Crizotinib)
when proteinuria appears, but even more when it is over Crizotinib is approved for non-small cell lung cancer,
3 g per day or associated to AKI, since histological find- inhibiting the ALK, MET, and ROS1 kinases. Renal tox-
ings could influence prognosis and treatment options. icities include AKI (NTA and ATIN) and the formation
Anti-VEGF therapies have been related with minimal or increase of renal cysts during treatment for lung cancer
change disease and focal segmental glomerulosclerosis [44]. Schnell et al. [45] reviewed 272 patients treated with
(FSGS), but the most common histologic pattern is TMA. this drug in clinical trials and found new renal cysts (9%)
Anti-VEGF-associated TMA, as well as nephrotic range or increase in the size of renal cysts (2%) of treated pa-

Downloaded from http://karger.com/nef/article-pdf/147/2/65/3915619/000525029.pdf by guest on 22 June 2023


proteinuria, are considered a reason to discontinue the tients [45]. Furthermore, reductions in GFR have been
culprit drug. Renal response to discontinuation of the of- observed in patients who underwent this therapy.
fending agent is normally good with no need of other spe-
cific treatment [30, 32]. Recently, some case series of bev- EGFR Inhibitors (Cetuximab, Erlotinib, Gefitinib,
acizumab-associated TMA treated with eculizumab have Panitumumab)
been reported, suggesting that complement blockade Inhibition of EGFR, that results in phosphorylation of
should be an option for patients without response to the the receptor tyrosine kinase modulating cell differentia-
classical management [34, 35]. tion, proliferation, and survival; is crucial for the treat-
ment of some cancers such as non-small cell lung cancer.
TKIs (Axitinib, Pazopanib, Sorafenib, Regorafenib, EGFR inhibitors have been associated with electrolyte
Sunitinib) disorders; mainly hypomagnesemia; but also hypophos-
Decreased GFR has been reported during therapy with phatemia, and hypokalemia. Increased renal magnesium
axitinib, sunitinib, and sorafenib, although renal failure is has been found in 37% of patients, which resolves with
rare. TKIs are also related with TMA. In particular, suni- therapy discontinuation [46]. Cetuximab has also been
tinib in a case report of 5 patients has been associated with associated with a single case of IgA glomerulonephritis
AKI, eosinophilia, and/or nephrotic syndrome, and/or [47] and nephrotic syndrome [48].
TMA; although none were biopsied [36]. NTIA has been
also diagnosed in a biopsy-proven cases related with suni- Bcr-Abl TKIs
tinib and sorafenib [37, 38]. Besides, small series and case Bcr-Abl TKIs are the first-line therapy in chronic my-
reports related, these two drugs with minimal change, elogenous leukemia, caused by chromosomic abnormali-
FSGS, and immune complex deposition have been pub- ties resulting in the so-called Philadelphia chromosome
lished [39, 40]. Regorafenib has been associated with sev- in more than 90% of patients. The presence of the chime-
eral electrolyte abnormalities, including hypophosphate- ric oncogene BCR-Abl constitutes an active BRC-Abl ty-
mia, hypocalcemia, hyponatremia, and hypokalemia rosine kinase, a pathogenic factor of the disease. These
[41]. inhibitors are imatinib, dasatinib, nilotinib, and pona-
tinib [49]. Imatinib has been related to episodes of AKI in
BRAF Inhibitors (Dabrafenib, Vemurafenib) 7% and also to CKD. The decrease in the glomerular fil-
Many melanomas and other tumors cause point muta- tration rate has been associated with the treatment dura-
tions in the serine-threonine kinase BRAF. Dabrafenib tion [50]. AKI could appear as a part of a tumor lysis syn-
and vemurafenib are the two approved inhibitors of mu- drome (TLS) and drive to tubular damage. Interestingly,
tant BRAF for use in melanoma. They have been related acute tubular necrosis has been described as the histo-
with AKI and some electrolytes disorders as Fanconi syn- logic lesion in a patient on imatinib treatment who devel-
drome, hypophosphatemia, hyponatremia, and hypoka- oped AKI and required renal replacement therapy [51].
lemia [42]. The mechanism of kidney injury is unknown; Proteinuria could also appear in patients under imatinib
however, BRAF inhibitors interfere with the downstream treatment, often related de VEGF inhibition. Data about

70 Nephron 2023;147:65–77 García-Carro/Draibe/Soler


DOI: 10.1159/000525029
AKI related to nilotinib or ponatinib are scarce, but both inhibition of dopamine, norepinephrine, and serotonin
cause HTN in 10% and 50% of patients, respectively. Hy- reuptake. Patients on niraparib therapy should have their
pophosphatemia is also common in BCR-Abl TKIs treat- blood pressure monitored at least monthly for the first
ments, because they inhibit phosphorus tubular reab- year of treatment. If HTN is developed, it should be man-
sorption. aged with standard antihypertensive drugs and, if neces-
Ibrutinib inhibits the Bruton tyrosine kinase and is sary, adjusting PARP inhibitor dosing [59].
commonly used as treatment of chronic lymphocytic leu-
kemia. It has been related to HTN and also to TLS in a few BCL-2 Inhibitor (Venetoclax)
cases. Recently, two cases of biopsy-proven acute tubular The members of the B-cell leukemia/lymphoma-2
injury secondary to ibrutinib have been published, and (BCL-2) family of proteins are key regulators of the in-
the authors review adverse events related to this drug: a trinsic apoptotic pathway; dysregulation of this pathway
total of 25% from more than 600 adverse events were re- leads to the pathologic survival of cancer cells [60]. Vene-
ported as AKI [52, 53]. toclax is a BCL-2 inhibitor approved for chronic lympho-
cytic leukemia and acute myeloid leukemia [60, 61].
Cyclin-Dependent Kinases 4/6 Inhibitors and Venetoclax can lead to rapid TLS in CLL patients and, in

Downloaded from http://karger.com/nef/article-pdf/147/2/65/3915619/000525029.pdf by guest on 22 June 2023


Poly(ADP-Ribose) Polymerase Inhibitors order to minimize this adverse effect, a weekly dose ramp-
Cyclin-dependent kinases (CDK) inhibitors targeting up is recommended [62]. Furthermore, a recently pub-
CDK4 and CDK6 are currently commonly used in hor- lished case series suggested that TLS prophylaxis with al-
mone receptor-positive breast cancer, mainly in advanced lopurinol >72 h prior to venetoclax initiation is beneficial
stages [54]. Abemaciclib and other inhibitors of CDK 4/6 [63].
have been associated with reversible increases in serum
creatinine (SCr) levels of ∼15%–40% over baseline. Data Proteasome Inhibitors
of a recent in vitro study of abemaciclib renal effect Proteasome inhibitors (PIs) are a cornerstone for the
showed that this increase is caused by inhibition of renal treatment of multiple myeloma (MM), a plasma cell dis-
transporters and CDK4/6 inhibitors seem not to affect order characterized by abnormal proliferation of plasma
renal function when assessed by measured GFR, or mod- cells resulting in overproduction of paraprotein that rep-
ify the concentrations of urinary biomarkers of renal in- resents 10% of hematological malignancies. First ap-
jury such as NGAL or KIM-1. Patients under abemaciclib proved PI was bortezomib, and then carfilzomib and ixa-
will experience a mild (∼10%–40%) reversible increase in zomib [64]. There are increasing reports of TMA in as-
SCr due to renal transport inhibition. The increase in SCr sociation with PI exposure. It is interesting to mention
following may be higher in patients with CKD. When that prior to the use of PIs, the incidence of TMA in pa-
clinically indicated, alternative measurements of renal tients with MM was much lower, most frequently associ-
function different from creatinine-derived EGFR should ated with autologous stem-cell transplant and that there
be used in patients taking these drugs [55]. are no reported cases of PI-related TMA in patients with
Olaparib, niraparib, rucaparib, and talazoparib are lymphoma, where PI are also used. Thus, MM itself could
poly (ADP-ribose) polymerase (PARP) inhibitors that be a risk factor for PI-induced TMA. The morbidity and
are also used in gynecological cancer: both ovary and mortality of PI-related TMA are notable and for that rea-
breast cancer [56, 57]. Despite their mechanism of action son, clinical vigilance is required for early detection and
is not similar to CDK inhibitors, this group of drugs could intervention. TMA is not limited to the kidney and
also cause inhibition of renal transporters leading to a re- supportive treatment, including renal replacement thera-
versible and dose-dependent increase in SCr not affecting pies and blood transfusions, is needed, as well as a cessa-
GFR, since the median cystatin C-derived EGFR was tion of PI and any other offending agent. TMA seems to
comparable before/off treatment and during treatment be more frequent in patients on carfilzomib than in those
with olaparib in a study by Bruin et al. [58]. As with CKD on bortezomib according to the literature [65].
inhibitors, the use of the creatinine-derived EGFR can
give an underestimation of GFR in patients taking olapa-
rib. Therefore, an alternative renal marker should be used
to accurately calculate GFR in these patients. Interesting-
ly, PARP inhibitors, and especially niraparib, have been
related to any-grade HTN, any-grade HTN secondary to

Anticancer Drug-Related Nephrotoxicity Nephron 2023;147:65–77 71


DOI: 10.1159/000525029
Table 3. Cancer immunotherapies and nephrotoxicity

Medication Nephrotoxicity Mechanism of nephrotoxicity Prevention and


management
recommendations

IFN MCD/FSGS Direct effects of IFN on the podocyte Drug discontinuation


TMA Indirect effects via altered cytokine milieu Steroids (MCD/FSGS)
Damage microvascular endothelial cells by
inducing apoptosis in a dose-dependent manner
Interleukin-2 Hemodynamic AKI Capillary leak syndrome leading to AKI Volume filling
ATI
CAR-T Hemodynamic AKI CRS Previous chemotherapy/
ATI steroids
TLS Supportive treatment
Anti-IL-6: Tocilizumab

Downloaded from http://karger.com/nef/article-pdf/147/2/65/3915619/000525029.pdf by guest on 22 June 2023


Immune checkpoint ATIN Formation of new or reactivated T cells against CPI withdrawal
inhibitors (CTLA-4 ATI tumor antigens that cross-react with kidney Steroids
inhibitors, PD-1 Lupus-like glomerulonephritis antigens
inhibitors, PD-L1 Podocytopathies Loss of tolerance with reactivation of drug-specific
inhibitors) Necrotizing glomerulonephritis and T cell induced by ICI
vasculitis Increase in proinflammatory cytokines in kidney
C3 glomerulonephritis tissue
TMA Generation of autoantibodies against kidney
tissues

MCD, minimal change disease; FSGS, focal segmental glomerulosclerosis; TMA, thrombotic microangiopathy; ATI, acute tubular injury;
CTLA-4, cytotoxic T-lymphocyte-associated protein 4; PD-1, programmed death protein 1; PD-L1, programmed death-ligand 1.

Cancer Immunotherapies drug discontinuation is favorable in published series and


steroids can also be used [68]. Black race and some APOL1
Outcomes in advanced neoplasia’s have dramatically gene variants have been identified as risk factors for the
changed over the last decades. The use of drugs that mod- development of nephrotic syndrome under IFN treat-
ulate the immune response instead of direct tumor target- ment [67, 69].
ing plays an important role in this improvement in On-
cology [66]. However, the goal of modifying immune re- Interleukin-2
sponse has been linked to renal-related adverse effects Interleukin-2 (IL2) has been used for years in the treat-
that could limit the potential use of these treatments (see ment of renal-cell carcinoma and melanoma. High doses
Table 3). are necessary and IL2 was initially associated with an im-
portant treatment-related mortality, that has substantial-
Interferons ly decreased as long as experience has been gained [70].
Interferons (IFNs) are a group of pleiotropic cytokines The main toxicity of IL2 is a capillary leak syndrome in
that play important roles in intercellular communication relation to an increased level of circulating cytokines.
during innate and acquired immune responses as a host This could drive to a hypovolemic/pre-renal AKI within
defensive response against viral and bacterial infections. 24–48 h after the drug infusion. Generally, AKI improves
They have been proposed for different diseases, such as after IL2 discontinuation and supportive care. However,
hepatitis or multiple sclerosis, and they have also been an acute tubular injury could be present if ischemia was
identified as oncospecific therapies. IFNs have been re- important and renal function recovery could be partial or
lated to nephrotic syndrome and AKI secondary to mini- delayed [4].
mal change disease and FSGS [67]. Renal response to

72 Nephron 2023;147:65–77 García-Carro/Draibe/Soler


DOI: 10.1159/000525029
Chimeric Antigen Receptor T-Cell patient age, impaired baseline renal function, concomi-
Chimeric antigen receptor (CAR) T-cell (CAR-T) tant use of proton pomp inhibitors, a combination of
therapy has been a dramatic success in the treatment of more than one CPI and the presence of previous im-
previously refractory hematologic malignancies. CAR-T mune-related adverse events [78–81]. A special charac-
cells are host T cells submitted to engineering to express teristic of AKI-related to CPI is that it occurs from some
receptors that recognize tumor antigens [71]. These ex weeks to many months after treatment initiation [82].
vivo modified T cells are able to target and directly de- The most frequent renal histologic pattern in patients
stroy cancer cells. The main adverse effect of CAR-T cells with biopsy-proven CPI-related AKI is acute tubuloint-
therapy is cytokine release syndrome (CRS) in the onset erstitial nephritis (ATIN) [83] but glomerular lesions
of immune activation [72], that could drive to hemody- have also been described, such as renal vasculitis, podo-
namic AKI or acute tubular injury in approximately 20% cytopathies, and C3 glomerulonephritis [84]. When com-
of patients. The rapid onset of tumor destroy by CAR-T pared to classical ATIN, CPI-related ATIN presented
cells could also promote a TLS and secondary renal in- with lower creatinine at diagnosis, higher urinary leuco-
jury [71]. Acute tubular necrosis is related to a longer hos- cyte counts, and time from initiation of the culprit drug
pital stay and a higher 60 days mortality [71]. Previous to ATIN diagnosis was longer in patients with CPI-relat-

Downloaded from http://karger.com/nef/article-pdf/147/2/65/3915619/000525029.pdf by guest on 22 June 2023


chemotherapy and steroids are recommended with the ed ATIN than in those with classical ATIN [85]. Recom-
aim to reduce tumor size and to minimize CRS in patients mendations for kidney biopsy in patients with CPI-relat-
who will be under CAR-T cells therapy for preventing ed AKI are becoming more limited as long as experience
AKI [72]. In patients who develop severe CRS, admission in treating this entity has grown. Currently, a biopsy is
in critical care units for supportive treatment is justified. recommended for AKI stages 2 and 3 and when a clinical
Moreover, interleukin-6 receptor blockers, such as toci­ suspicion of glomerular injury exists (i.e., nephrotic range
lizumab, are useful to reduce systemic inflammation and, proteinuria, oliguria, dysmorphic hematuria) [4]. CPI
consequently, are useful in preventing the development withdrawal is mandatory in CPI-related AKI and cortico-
and in the management of AKI related to CAR-T cells steroids therapy should be initiated if ATIN is clinically
therapies [71]. suspected [78, 86].

Immune Checkpoint Inhibitors (CTLA-4 Inhibitors,


PD-1 Inhibitors, PD-L1 Inhibitors) Other Therapies
Immune checkpoint inhibitors (CPI) have revolution-
ized the treatment of some malignancies, and their use is Bisphosphonates
increasing day by day [73, 74] because of its striking effect Bisphosphonates have the ability to inhibit osteoclast-
on improving survival rates [75, 76]. CPI enhance tumor- mediated bone destruction, so they are used as the treat-
directed immune responses by binding inhibitory recep- ment of malignant bone diseases. Furthermore, they have
tors such as cytotoxic T-lymphocyte-associated protein 4, been shown antitumor activity by acting together with
and the programmed death protein 1 or its ligand on T other antineoplastic drugs [87]. In contrast to oral
cells and other immune cells causing a downregulation of bisphosphonates, that are used mainly to treat postmeno-
its activation. CPI binds to cytotoxic T-lymphocyte-asso- pausal osteoporosis, intravenous bisphosphonates such
ciated protein 4, programmed death protein 1 or its li- as zoledronate and pamidronate are related to potential
gand to activate immune cells from a quiescent state – nephrotoxicity that limits its use. Kidney damage is re-
triggering a vigorous response against tumor cells [77]. lated to dose and infusion time and both pamidronate
Since CPI mechanisms are not selective, these drugs cause and zoledronate could cause acute tubular necrosis and
diverse autoimmune phenomena known as immune-re- nephrotic syndrome with FSGS histologic pattern. For
lated adverse events. AKI incidence in patients under CPI that reason, renal function should be monitored prior to
treatment has been increasing to 13–29% as long as CPI treatment infusion and, if SCr rises, the drug should be
use has become more ordinary, however biopsy-proven discontinued. In addition, renal damage could be pre-
or clinically CPI-related AKI has been only found in 2%– vented by increasing the time interval between doses and
3% [78, 79]. In patients under CPI treatment, the develop- by strict adherence to guidelines dose recommendations
ment of AKI has been identified as a risk factor for mor- adjustment by baseline renal function. Ibandronate has
tality [80] as well as an absence of renal recovery after an not been related to renal damage even in patients with
AKI episode [78]. Risk factors for AKI related to CPI are CKD [4, 88].

Anticancer Drug-Related Nephrotoxicity Nephron 2023;147:65–77 73


DOI: 10.1159/000525029
mTOR Inhibitors combination with the classical ones have nicely demon-
Everolimus is approved for the treatment of breast strated to increase the life expectancy of the patients with
cancer, neuroendocrine tumors (pancreas, lung, or gas- advanced malignancy; however, they have been highly as-
trointestinal), advanced renal-cell carcinoma, angiomyo- sociated with renal toxicity that worsens the patient qual-
lipoma in tuberous sclerosis complex, astrocytoma, and ity of life and increases mortality. As there is a bidirec-
other types of malignancies, as well as sirolimus, a related tional axis between cancer and kidney injury, the con-
agent [89]. The main renal complication of everolimus solidation of dedicated onconephrology clinics is crucial
and sirolimus is proteinuria, reported in 3%–36% of pa- at least in referral hospitals.
tients. Sirolimus has also been associated with nephrotic
syndrome in 2% of cases, with an FSGS and membrano-
proliferative histologic pattern. mTOR inhibitor-related Conflict of Interest Statement
proteinuria mechanism is not well known but it could be
M.J. Soler reports honorarium for conferences, consulting fees,
in part related to a decreased production of VEGF in
and advisory boards from Astra Zeneca, Novo Nordsik, Esteve,
podocytes or to a decreased expression of cubilin and Vifor, Bayer, Mundipharma, Ingelheim Lilly, Jansen, ICU Medi-
megalin that leads to a decreased tubular albumin reup- cal, Travere Therapeutics, and Boehringer. C.G.-C. has received

Downloaded from http://karger.com/nef/article-pdf/147/2/65/3915619/000525029.pdf by guest on 22 June 2023


take. If proteinuria is mild to moderate, ACE inhibitors travel and congress fees support from AstraZeneca, Esteve, Novo-
or ARB and lifestyle modifications are recommended, Nordisk, Boehringer Ingelheim Lilly, Astellas, Otsuka, Novartis,
Astellas, and Baxter and has given scientific lectures and partici-
and mTOR inhibitor withdrawal is not necessary. In con-
pated in advisory boards organized by AstraZeneca, Boehringer
trast, in patients with severe proteinuria or nephrotic syn- Ingelheim Lilly, Mundipharma, and NovoNordisk.
drome, discontinuation of the culprit drug may be the
strategy to prevent AKI. Plasmapheresis could be benefi-
cial in patients with focal and segmental glomeruloscle- Funding Sources
rosis or membranoproliferative glomerulonephritis [4,
49]. This work was supported by grants from Fondo de Investig-
ación Sanitaria-FEDER, ISCIII, Río Hortega CM20/00111,
PI17/00257, EIN2020-1123381, RD16/0009/0030 (REDINREN),
Calcineurin Inhibitors and RD21/0005/0016 (RICORS 2040).
Calcineurin inhibitors (CNIs), both tacrolimus and
cyclosporine A, are drugs that are well known by nephrol-
ogists because they are part of the classical immunosup- Author Contributions
pressive regimen in renal transplantation, as well as in
other solid organ transplantation. However, CNI are also Conceptualization; writing – revision draft preparation, re-
used in allogenic hematopoietic stem-cell transplantation view, and editing; and supervision: Juliana Draibe, Clara García-
(HSCT) and, as happens in renal transplantation, their Carro, and María José Soler. All authors have read and agreed to
the published version of the manuscript.
use may be associated with AKI. Major renal effects of
CNI are renal vessel constriction, endothelial damage,
and tubular toxicity. However, cyclosporine A and tacro-
limus can also cause CNI-related TMA [4], that some- References 1 Bray F, Ferlay J, Soerjomataram I, Siegel RL,
times is limited to kidney but can also have peripheral Torre LA, Jemal A. Global cancer statistics
expression that sometimes requires supportive care in re- 2018: GLOBOCAN estimates of incidence
and mortality worldwide for 36 cancers in 185
cent HSCT recipients. CNI are also used as immunosup- countries. CA Cancer J Clin. 2018 Nov;68(6):
pressive treatment of aplastic anemia in ineligible pa- 394–424.
tients for first-line therapy, HSCT [90]. 2 Launay-Vacher V, Oudard S, Janus N, Gli-
gorov J, Pourrat X, Rixe O, et al. Prevalence of
renal insufficiency in cancer patients and im-
plications for anticancer drug management:
Conclusions the Renal Insufficiency and Anticancer Medi-
cations (IRMA) Study. Cancer. 2007 Sep;
110(6):1376–84.
Cancer treatment including conventional chemother- 3 Zoccali C, Blankestijn PJ, Bruchfeld A, Capas-
apy, targeted cancer agents, and immunotherapies among so G, Fliser D, Fouque D, et al. Children of a
lesser god: exclusion of chronic kidney dis-
others are significantly associated with kidney injury in ease patients from clinical trials. Nephrol Dial
terms of acute and CKD. The new therapies alone or in Transplant. 2019 Jul;34(7):1112–4.

74 Nephron 2023;147:65–77 García-Carro/Draibe/Soler


DOI: 10.1159/000525029
4 Rosner MH, Jhaveri KD, McMahon BA, Pera- 19 Mita AC, Sweeney CJ, Baker SD, Goetz A, 32 Eremina V, Jefferson JA, Kowalewska J,
zella MA. Onconephrology: the intersections Hammond LA, Patnaik A, et al. Phase I and Hochster H, Haas M, Weisstuch J, et al. VEGF
between the kidney and cancer. CA Cancer J Pharmacokinetic Study of pemetrexed ad- inhibition and renal thrombotic microangi-
Clin. 2021 Jan;71(1):47–77. ministered every 3 weeks to advanced cancer opathy. N Engl J Med. 2008 Mar; 358(11):
5 Porta C, Cosmai L, Gallieni M, Pedrazzoli P, patients with normal and impaired renal 1129–36.
Malberti F. Renal effects of targeted antican- function. J Clin Oncol. 2006 Feb; 24(4): 552– 33 Kanbayashi Y, Ishikawa T, Tabuchi Y, Saka-
cer therapies. Nat Rev Nephrol. 2015 Jun; 62. guchi K, Ouchi Y, Otsuji E, et al. Predictive
11(6):354–70. 20 Bleyer WA. The clinical pharmacology of factors for the development of proteinuria in
6 Perazella MA. Onco-nephrology: renal tox- methotrexate: new applications of an old cancer patients treated with bevacizumab,
icities of chemotherapeutic agents. Clin J Am drug. Cancer. 1978 Jan;41(1):36–51. ramucirumab, and aflibercept: a single-insti-
Soc Nephrol. 2012 Oct;7(10):1713–21. 21 Celtikci B, Leclerc D, Lawrance AK, Deng L, tution retrospective analysis. Sci Rep. 2020
7 Perazella MA, Moeckel GW. Nephrotoxicity Friedman HC, Krupenko NI, et al. Altered ex- Dec;10(1):2011.
from chemotherapeutic agents: clinical mani- pression of methylenetetrahydrofolate reduc- 34 Vakiti A, Singh D, Pilla R, Alhaj-Moustafa M,
festations, pathobiology, and prevention/ tase modifies response to methotrexate in Fitzpatrick KW. Bevacizumab-induced atypi-
therapy. Semin Nephrol. 2010 Nov; 30(6): mice. Pharmacogenet Genomics. 2008 Jul; cal hemolytic uremic syndrome and treat-
570–81. 18(7):577–89. ment with eculizumab. J Oncol Pharm Pract.
8 Sutton RA, Walker VR, Halabe A, Swenerton 22 Wall SM, Johansen MJ, Molony DA, DuBose 2019 Jun;25(4):1011–5.
K, Coppin CM. Chronic hypomagnesemia TD, Jaffe N, Madden T. Effective clearance of 35 Hilburg R, Geara AS, Qiu MK, Palmer MB,
caused by cisplatin: effect of calcitriol. J Lab methotrexate using high-flux hemodialysis Chiang EY, Burger RA, et al. Bevacizumab-
Clin Med. 1991 Jan [cited 2022 Jan 9];117(1): membranes. Am J Kidney Dis. 1996 Dec; associated thrombotic microangiopathy

Downloaded from http://karger.com/nef/article-pdf/147/2/65/3915619/000525029.pdf by guest on 22 June 2023


40–3. 28(6):846–54. treated with eculizumab: A case series and
9 Seguro AC, Massola Shimizu MH, Harami 23 Widemann BC, Schwartz S, Jayaprakash N, systematic review of the literature. Clin
Kudo L, Dos Santos Rocha A. Renal concen- Christensen R, Pui CH, Chauhan N, et al. Ef- Nephrol. 2021 Jul;96(1):51–9.
tration defect induced by cisplatinThe role of ficacy of glucarpidase (carboxypeptidase g2) 36 Jhaveri KD, Flombaum CD, Kroog G, Glezer-
thick ascending limb and papillary collecting in patients with acute kidney injury after man IG. Nephrotoxicities associated with the
duct. Am J Nephrol. 1989;9(1):59–65. high-dose methotrexate therapy. Pharmaco- use of tyrosine kinase inhibitors: a single-cen-
10 Wangila GW, Nagothu KK, Steward R, Bhatt therapy. 2014;34(5):427–39. ter experience and review of the literature.
R, Iyere PA, Willingham WM, et al. Preven- 24 Daviet F, Rouby F, Poullin P, Moussi-Francès Nephron Clin Pract. 2011 Apr;117(4):c312.
tion of cisplatin-induced kidney epithelial cell J, Sallée M, Burtey S, et al. Thrombotic micro- 37 Winn SK, Ellis S, Savage P, Sampson S, Marsh
apoptosis with a Cu superoxide dismutase- angiopathy associated with gemcitabine use: JE. Biopsy-proven acute interstitial nephritis
mimetic [copper2II(3,5-ditertiarybutylsalicyl Presentation and outcome in a national associated with the tyrosine kinase inhibitor
ate)4(ethanol)4]. Toxicol In Vitro. 2006 Dec; French retrospective cohort. Br J Clin Phar- sunitinib: a class effect? Nephrol Dial Trans-
20(8):1300–12. macol. 2019 Feb;85(2):403–12. plant. 2009 Feb;24(2):673–5.
11 Jackson AM, Rose BD, Graff LG, Jacobs JB, 25 Weitz IC. Thrombotic microangiopathy in 38 Izzedine H, Brocheriou I, Rixe O, Deray G.
Schwartz JH, Strauss GM, et al. Thrombotic cancer. Thromb Res. 2018 Apr; 164(Suppl 1): Interstitial nephritis in a patient taking
microangiopathy and renal failure associated S103–5. sorafenib. Nephrol Dial Transplant. 2007
with antineoplastic chemotherapy. Ann In- 26 Al Ustwani O, Lohr J, Dy G, LeVea C, Con- Aug;22(8):2411.
tern Med. 1984;101(1):41–4. nolly G, Arora P, et al. Eculizumab therapy for 39 Chen YS, Chen CL, Wang JS. Nephrotic syn-
12 Ruggiero A, Ariano A, Triarico S, Capozza gemcitabine induced hemolytic uremic syn- drome and acute renal failure apparently in-
MA, Romano A, Maurizi P, et al. Cisplatin- drome: case series and concise review. J Gas- duced by sunitinib. Case Rep Oncol. 2009
induced nephrotoxicity in children: what is trointest Oncol. 2014;5(1):E30. Oct;2(3):172–6.
the best protective strategy? J Oncol Pharm 27 Groff JA, Kozak M, Boehmer JP, Demko TM, 40 Turan N, Benekli M, Ozturk SC, Inal S, Me-
Pract. 2021 Jan;27(1):180–6. Diamond JR. Endotheliopathy: a continuum mis L, Guz G, et al. Sunitinib- and sorafenib-
13 Isnard-Bagnis C, Moulin B, Launay-Vacher of hemolytic uremic syndrome due to mito- induced nephrotic syndrome in a patient with
V, Izzedine H, Tostivint I, Deray G. Antican- mycin therapy. Am J Kidney Dis. 1997;29(2): gastrointestinal stromal tumor. Ann Pharma-
cer drug-induced nephrotoxicity. Nephrol 280–4. cother. 2012 Oct;46(10):e27.
Ther. 2005;1(2):101–14. 28 Izzedine H, Perazella MA. Thrombotic mi- 41 Grothey A, Van Cutsem E, Sobrero A, Siena
14 Ensergueix G, Pallet N, Joly D, Levi C, croangiopathy, cancer, and cancer drugs. Am S, Falcone A, Ychou M, et al. Regorafenib
Chauvet S, Trivin C, et al. Ifosfamide nephro- J Kidney Dis. 2015 Nov;66(5):857–68. monotherapy for previously treated metastat-
toxicity in adult patients. Clin Kidney J. 2020 29 Troxell ML, Higgins JP, Kambham N. Anti- ic colorectal cancer (CORRECT): an interna-
Aug;13(4):660–5. neoplastic treatment and renal injury: an up- tional, multicentre, randomised, placebo-
15 Higgs D, Nagy C, Einhorn LH. Ifosfamide: a date on renal pathology due to cytotoxic and controlled, phase 3 trial. Lancet. 2013;
clinical review. Semin Oncol Nurs. 1989; 5(2 targeted therapies. Adv Anat Pathol. 2016 381(9863):303–12.
Suppl 1):70–7. Sep;23(5):310–29. 42 Wanchoo R, Jhaveri KD, Deray G, Launay-
16 Akilesh S, Juaire N, Duffield JS, Smith KD. 30 Jhaveri KD, Wanchoo R, Sakhiya V, Ross Vacher V. Renal effects of BRAF inhibitors: a
Chronic ifosfamide toxicity: kidney patholo- DW, Fishbane S. Adverse renal effects of nov- systematic review by the Cancer and the Kid-
gy and pathophysiology. Am J Kidney Dis. el molecular oncologic targeted therapies: a ney International Network. Clin Kidney J.
2014 May;63(5):843–50. narrative review. Kidney Int Rep. 2016 Jan; 2016 Apr;9(2):245–51.
17 Stavroulopoulos A, Nakopoulou L, Xydakis 2(1):108–23. 43 Jhaveri KD, Sakhiya V, Fishbane S. Nephro-
AM, Aresti V, Nikolakopoulou A, Klouvas G. 31 Yamada R, Okawa T, Matsuo K, Suzuki M, toxicity of the BRAF inhibitors vemurafenib
Interstitial nephritis and nephrogenic diabe- Mori N, Mori K. Renal-limited thrombotic and dabrafenib. JAMA Oncol. 2015 Nov;1(8):
tes insipidus in a patient treated with peme- microangiopathy after switching from beva- 1133–4.
trexed. Ren Fail. 2010 Sep;32(8):1000–4. cizumab to ramucirumab: a case report. BMC 44 Martín Martorell P, Huerta Alvaro M, Solís
18 Perazella MA, Izzedine H. New drug toxicities nephrol. 2019 Jan;20(1):14. Salguero MA, Insa Molla A. Crizotinib and
in the onco-nephrology world. Kidney Int. renal insufficiency: a case report and review of
2015 May;87(5):909–17. the literature. Lung cancer. 2014;84(3):310–3.

Anticancer Drug-Related Nephrotoxicity Nephron 2023;147:65–77 75


DOI: 10.1159/000525029
45 Schnell P, Bartlett CH, Solomon BJ, Tassell V, 59 LaFargue CJ, Dal Molin GZ, Sood AK, Cole- 73 Larkin J, Chiarion-Sileni V, Gonzalez R, Grob
Shaw AT, de Pas T, et al. Complex renal cysts man RL. Exploring and comparing adverse JJ, Cowey CL, Lao CD, et al. Combined
associated with crizotinib treatment. Cancer events between PARP inhibitors. Lancet On- nivolumab and ipilimumab or monotherapy
Med. 2015 Jun;4(6):887. col. 2019 Jan;20(1):e15–28. in untreated melanoma. N Engl J Med. 2015
46 Schrag D, Chung KY, Flombaum C, Saltz L. 60 Ryan CE, Davids MS. BCL-2 inhibitors, pres- Jul;373(1):23–34.
Cetuximab therapy and symptomatic hypo- ent and future. Cancer J. 2019 Nov;25(6):401– 74 Gandhi L, Rodríguez-Abreu D, Gadgeel S, Es-
magnesemia. J Natl Cancer Inst. 2005 Aug; 9. teban E, Felip E, De Angelis F, et al. Pembro-
97(16):1221–4. 61 Cozad M, Stump SE, Buhlinger K, Collins J, lizumab plus chemotherapy in metastatic
47 Sasaki K, Anderson E, Shankland SJ, Nicosia Muir M, Coombs CC, et al. Evaluation of an non-small-cell lung cancer. N Engl J Med.
RF. Diffuse proliferative glomerulonephritis interdisciplinary venetoclax initiation pro- 2018 May;378(22):2078–92.
associated with cetuximab, an epidermal cess in minimizing risk of tumor lysis syn- 75 Hamid O, Robert C, Daud A, Hodi FS, Hwu
growth factor receptor inhibitor. Am J Kidney drome. Leuk Lymphoma. 2022:1–8. WJ, Kefford R, et al. Five-year survival out-
Dis. 2013 Jun;61(6):988–91. 62 Roberts AW, Davids MS, Pagel JM, Kahl BS, comes for patients with advanced melanoma
48 Harari PM. Epidermal growth factor receptor Puvvada SD, Gerecitano JF, et al. Targeting treated with pembrolizumab in KEY-
inhibition strategies in oncology. Endocr BCL2 with venetoclax in relapsed chronic NOTE-001. Ann Oncol. 2019 Apr [cited 2022
Relat Cancer. 2004 Dec;11(4):689–708. lymphocytic leukemia. N Engl J Med. 2016 Jan 9];30(4):582–8.
49 Abbas A, Mirza MM, Ganti AK, Tendulkar K. Jan;374(4):311–22. 76 Hodi FS, Chesney J, Pavlick AC, Robert C,
Renal toxicities of targeted therapies. Target 63 Esparza S, Muluneh B, Galeotti J, Matson M, Grossmann KF, McDermott DF, et al. Com-
Oncol. 2015 Dec;10(4):487–99. Richardson DR, Montgomery ND, et al. bined nivolumab and ipilimumab versus ipi-
50 Marcolino MS, Boersma E, Clementino NCD, Venetoclax-induced tumour lysis syndrome limumab alone in patients with advanced

Downloaded from http://karger.com/nef/article-pdf/147/2/65/3915619/000525029.pdf by guest on 22 June 2023


Macedo AV, Marx-Neto AD, Silva MHCR, et in acute myeloid leukaemia. Br J Haematol. melanoma: 2-year overall survival outcomes
al. Imatinib treatment duration is related to 2020 Jan;188(1):173–7. in a multicentre, randomised, controlled,
decreased estimated glomerular filtration rate 64 Haddadin M, Al-Sadawi M, Madanat S, Tam phase 2 trial. Lancet Oncol. 2016 Nov;17(11):
in chronic myeloid leukemia patients. Ann E, Taiwo E, Luhrs C, et al. Late presentation 1558–68.
Oncol. 2011;22(9):2073–9. of carfilzomib associated thrombotic micro- 77 Champiat S, Lambotte O, Barreau E, Belkhir
51 Pou M, Saval N, Vera M, Saurina A, Solé M, angiopathy. Am J Med Case Rep. 2019 Jul; R, Berdelou A, Carbonnel F, et al. Manage-
Cervantes F, et al. Acute renal failure second- 7(10):240–3. ment of immune checkpoint blockade dysim-
ary to imatinib mesylate treatment in chronic 65 Monteith BE, Venner CP, Reece DE, Kew AK, mune toxicities: a collaborative position pa-
myeloid leukemia. Leuk Lymphoma. 2003 Jul; Lalancette M, Garland JS, et al. Drug-induced per. Annals of Oncol. 2016 Apr;27(4):559–74.
44(7):1239–41. thrombotic microangiopathy with concur- 78 Cortazar FBB, Kibbelaar ZAA, Glezerman
52 Manohar S, Bansal A, Wanchoo R, Sakhiya V, rent proteasome inhibitor use in the treat- IGG, Abudayyeh A, Mamlouk O, Motwani
Lucia S, Jhaveri KD. Ibrutinib induced acute ment of multiple myeloma: a case series and SSS, et al. Clinical features and outcomes of
tubular injury: a case series and review of the review of the literature. Clin Lymphoma My- immune checkpoint inhibitor-associated
literature. Am J Hematol. 2019 Sep; 94(9): eloma Leuk. 2020 Nov;20(11):e791–800. AKI: a Multicenter Study. J Am Soc Nephrol.
E223–5. 66 Yang Y. Cancer immunotherapy: harnessing 2020;31(2):435–46.
53 Kaur V, Mehta P, Johnsurd J, Govindarajan R. the immune system to battle cancer. J Clin In- 79 Seethapathy H, Zhao S, Chute DF, Zubiri L,
Ibrutinib-associated tumor lysis syndrome in vest. 2015 Sep;125(9):3335–7. Oppong Y, Strohbehn I, et al. The incidence,
a patient with chronic lymphocytic leukemia. 67 Markowitz GS, Bomback AS, Perazella MA. causes, and risk factors of acute kidney injury
Blood. 2014 Nov;124(23):3503–5. Drug-induced glomerular disease: direct cel- in patients receiving immune checkpoint in-
54 Chou J, Quigley DA, Robinson TM, Feng FY, lular injury. Clin J Am Soc Nephrol. 2015 Jul; hibitors. Clin J Am Soc Nephrol. 2019 Dec;
Ashworth A. Transcription-associated cy- 10(7):1291–9. 14(12):1692–700.
clin-dependent kinases as targets and bio- 68 Markowitz GS, Nasr SH, Stokes MB, D’Agati 80 García-Carro C, Bolufer M, Bury R, Castañe-
markers for cancer therapy. Cancer Discov. VD. Treatment with IFN-(alpha), -(beta), or da Z, Muñoz E, Felip E, et al. Acute kidney
2020 Mar;10(3):351–70. -(gamma) is associated with collapsing focal injury as a risk factor for mortality in onco-
55 Chappell JC, Turner PK, Pak YA, Bacon J, segmental glomerulosclerosis. Clin J Am Soc logical patients receiving check-point inhibi-
Chiang AY, Royalty J, et al. Abemaciclib in- Nephrol. 2010 Apr;5(4):607–15. tors. Nephrol Dial Transplant. 2021 Feb;
hibits renal tubular secretion without chang- 69 Nichols B, Jog P, Lee JH, Blackler D, Wilmot 37(5):887–894.
ing glomerular filtration rate. Clin Pharmacol M, D’agati V, et al. Innate immunity pathways 81 Meraz-Muñoz A, Amir E, Ng P, Avila-Casado
Ther. 2019 May;105(5):1187–95. regulate the nephropathy gene Apolipopro- C, Ragobar C, Chan C, et al. Acute kidney in-
56 Foo T, George A, Banerjee S. PARP inhibitors tein L1. Kidney Int. 2015 Feb;87(2):332–42. jury associated with immune checkpoint in-
in ovarian cancer: an overview of the practice- 70 Schwartz RN, Stover L, Dutcher JP. Managing hibitor therapy: incidence, risk factors and
changing trials. Genes Chromosomes Cancer. toxicities of high-dose interleukin-2. Oncol- outcomes. J Immunother Cancer. 2020 Jun;
2021 May;60(5):385–97. ogy (Williston Park). 2002 Nov [cited 2022 8(1):e000467.
57 Arun B, Akar U, Gutierrez-Barrera AM, Hor- Jan 9];16(11 Suppl 13):11–20. 82 Wanchoo R, Karam S, Uppal NN, Barta VS,
tobagyi GN, Ozpolat B. The PARP inhibitor 71 Gupta S, Seethapathy H, Strohbehn IA, Frig- Deray G, Devoe C, et al. Adverse renal effects
AZD2281 (Olaparib) induces autophagy/mi- ault MJ, O’Donnell EK, Jacobson CA, et al. of immune checkpoint inhibitors: a narrative
tophagy in BRCA1 and BRCA2 mutant breast Acute kidney injury and electrolyte abnor- review. Am J Nephrol. 2017 Feb;45(2):160–9.
cancer cells. Int J Oncol. 2015 Jul;47(1):262–8. malities after chimeric antigen receptor T-cell 83 Oleas D, Bolufer M, Agraz I, Felip E, Muñoz
58 Bruin MAC, Korse CM, van Wijnen B, de (CAR-T) therapy for diffuse large B-cell lym- E, Gabaldón A, et al. Acute interstitial nephri-
Jong VMT, Linn SC, van Triest B, et al. A real phoma. Am J Kidney Dis. 2020 Jul;76(1): 63– tis associated with immune checkpoint inhib-
or apparent decrease in glomerular filtration 71. itors: a single-centre experience. Clin Kidney
rate in patients using olaparib? Eur J Clin 72 Bonifant CL, Jackson HJ, Brentjens RJ, Cur- J. 2020 Apr;14(5):1364–70.
Pharmacol. 2021 Feb;77(2):179–88. ran KJ. Toxicity and management in CAR T-
cell therapy. Mol Ther Oncolytics. 2016 Apr;
3:16011.

76 Nephron 2023;147:65–77 García-Carro/Draibe/Soler


DOI: 10.1159/000525029
84 Kitchlu A, Jhaveri KD, Wadhwani S, Desh- 86 Perazella MA, Shirali AC. Immune check- 88 Perazella MA, Markowitz GS. Bisphospho-
pande P, Harel Z, Kishibe T, et al. A system- point inhibitor nephrotoxicity: what do we nate nephrotoxicity. Kidney Int. 2008 Dec;
atic review of immune checkpoint inhibitor- know and what should we do? Kidney Int. 74(11):1385–93.
associated glomerular disease. Kidney Int 2020 Jan;97(1):62–74. 89 Hasskarl J. Everolimus. Recent Results Can-
Rep. 2020 Jan;6(1):66–77. 87 Stresing V, Daubiné F, Benzaid I, Mönkkönen cer Res. 2018;211:101–23.
85 Draibe JB, Garcia-Carro C, Martinez-Valen- H, Clézardin P. Bisphosphonates in cancer 90 Huang C, Huang L, Wang Y, Li X, Ren L, Gu
zuela L, Agraz I, Fulladosa X, Bolufer M, et al. therapy. Cancer Lett. 2007 Nov;257(1):16–35. X, et al. 6-month consequences of COVID-19
Acute tubulointerstitial nephritis induced by in patients discharged from hospital: a Cohort
checkpoint inhibitors versus classical acute Study. Lancet. 2021 Jan;397(10270):220–32.
tubulointerstitial nephritis: are they the same
disease? Clin Kidney J. 2020 Mar; 14(3): 884–
90.

Downloaded from http://karger.com/nef/article-pdf/147/2/65/3915619/000525029.pdf by guest on 22 June 2023

Anticancer Drug-Related Nephrotoxicity Nephron 2023;147:65–77 77


DOI: 10.1159/000525029

You might also like