Adverse Renal Effects of Anticancer Immunotherapy: Comparison
Please note this is a comparison between Version 2 by Jason Zhu and Version 3 by Jason Zhu.

Modern oncological therapy utilizes various types of immunotherapy. Immune checkpoint inhibitors (ICIs), chimeric antigen receptor T cells (CAR-T) therapy, cancer vaccines, tumor-targeting monoclonal antibodies (TT-mAbs), bispecific antibodies and cytokine therapy improve patients’ outcomes. However, stimulation of the immune system, beneficial in terms of fighting against cancer, generates the risk of harm to other cells in a patient’s body. Kidney damage belongs to the relatively rare adverse events (AEs). Best described, but still, superficially, are renal AEs in patients treated with ICIs. International guidelines issued by the European Society for Medical Oncology (ESMO) and the American Society of Clinical Oncology (ASCO) cover the management of immune-related adverse events (irAEs) during ICI therapy. 

  • cancer
  • immunotherapy
  • adverse events
  • immune checkpoint inhibitors

1. Introduction

Epidemiologists predict approximately 3.4 million new cases of diagnosed cancers in the European Union (EU) and European Free Trade Association (EFTA) countries and 1.9 million in the USA, which may become the cause of about 1.7 million and 410 thousand deaths a year in 2040, respectively [1][2]. All of these make malignant neoplastic diseases a very important field of research, leading to the development of novel therapies, which have been continuously improving patients’ survival. Being very promising, this dynamic situation results in the necessity for physicians to learn how to deal with patients treated for cancer and how to manage long-term side effects caused by treatment [3].
Immunotherapy is believed to be one of the most popular and promising therapeutic approaches for cancer patients. This method is derived from the observation that cancer cells can escape from the control of the immune system and evade destruction by immunocompetent cells [4][5]. A few mechanisms, such as the production of immunosuppressive factors (e.g., TGF-β) by tumor cells [6][7] and the recruitment of cells that can mitigate immunological response [8][9][10], are considered to underlie this phenomenon. Reversing these effects and boosting the natural immune system is the key principle of immunotherapy [11]. There are various ways to achieve this, and they are evolving over time, starting from brave trials to induce erysipelas in patients with inoperable sarcomas to reduce tumor size by William B. Coley at the end of the 19th century [12][13]. Other invented strategies based on immunological response include the use of therapeutic cancer vaccines, oncolytic viruses and cytokines [14][15][16]. A milestone for present-day therapies was the research on cytotoxic T cell antigen 4 (CTLA-4) and programmed death receptor 1 (PD-1) proteins by James P. Allison and Tasuku Honjo, respectively, who were awarded the Nobel Prize in Physiology or Medicine in 2018 [17][18][19]. Since then, the era of immune checkpoint inhibitors (ICIs) has started. The advances in numerous different branches of medicine enabled the invention of chimeric antigen receptor T cells (CAR-T) therapy, which consists in collecting T cells from a patient’s peripheral blood, placing chimeric antigen receptors in collected cells via genetic engineering methods and then transferring them back to the appropriately prepared patient [20][21].
In common with other cancer treatments, such as chemotherapy, radiotherapy and surgery, methods based on mobilizing the immune system to destroy neoplastic cells are not free from adverse drug reactions. In the case of ICIs, they are specific to these therapeutic regimens and are called immune-related adverse events (irAEs). Damaging healthy cells by an agitated immune system lies at the root of them. Early recognition and adequate management of irAEs are crucial for patients’ safety and therapeutic success; therefore, being familiar with them is essential for physicians dealing with oncological patients [22]. Immune-related adverse events may affect every organ and system, especially the skin, the gastrointestinal tract, and the nervous, and endocrine systems [23][24]. Nephrotoxicity after ICIs is thought to be a relatively rare complication, but it may be underreported [25][26]. Since early identification of renal injury plays a meaningful role in a patient’s outcome, being aware of its possible manifestations and their management is a vital part of a physician’s knowledge.

2. Immune Checkpoint Inhibitors (ICIs)

2.1. Mechanism of Action

The human immune system is not helpless in its confrontation with cancer cells. Not only does it fight infections that may lead to tumorigenesis, but it also recognizes and gets rid of suspicious cells in a process called immunosurveillance [27]. Immune cells identify neoplastic cells via neoantigens, defined as the proteins absent in healthy cells, which were produced in a process of transcription and translation of changed DNA sequence in cancerous cells [28]. The most important role in the recognition and further activation of immunological response involves antigen presenting cells (APCs), such as dendritic cells (DCs), which collect neoantigens, process and transfer them to secondary lymphoid organs where the antigen presentation takes place [29][30][31]. It happens due to the displaying of properly prepared tumor antigens via the major histocompatibility complex (MHC) present on the surface of the APC, which is then recognized by the T cell receptor (TCR) present on the surface of the T cells [32]. Complete activation of T cells is only possible if co-stimulation takes place. It is done by the interaction of the following proteins: CD28 on T cells and CD80/CD86 on APCs [33]. The proliferation of T cells then takes place, stimulated by autocrine or paracrine production of cytokines, especially interleukin 2 (IL-2) secreted by T cells [34]. Finally, activated T cells infiltrate the cancer tissue and recognize previously presented neoantigens, which enables the destruction of cancer cells. Neoantigens released by dead neoplastic cells amplify the immunologic response and therefore fulfill the cancer-immunity cycle [35][36]. The mechanism of T cell activation, however advantageous in terms of eliminating cancer cells, requires precise control to avoid excessive stimulation, which may result in damaging healthy tissues. The precise balance between optimal and excessive immune stimulation is maintained as a consequence of the interaction of special surface proteins called immune checkpoints, which takes place during the crosstalk between APC and T cells [37][38]. One of these proteins expressed on the surface of T cells, namely CTLA-4, competes with CD28 for binding with CD80/CD86. When the binding takes place, the signal for the proliferation of lymphocytes is suppressed [39][40]. The other important interaction, which weakens immunosurveillance, is the interplay of PD-1 with PD-L1 and programmed death-ligand 2 (PD-L2). PD-1 is present on the surface of the active T cells, whereas PD-L1 is expressed either on APCs or tumor cells [41][42]. All of these make a promising target for cancer therapies because suppressing inhibitory signals may improve the immune system’s capability to eradicate neoplastic cells [43]. This group of drugs, which actually are immunomodulatory monoclonal antibodies (mAbs), has been intensively studied and is still developing.

2.2. Possible Manifestations and Pathophysiology of Renal irAEs

Blockade of immune checkpoints enhances patients’ immune cells’ capability to detect foreign cells, which simultaneously results in the possibility of classifying their own cells as foreign ones. This lies at the basis of irAEs that may occur during therapy with ICIs and that may affect almost every system of the human body [44]. The estimated total incidence of irAEs varies among different studies between 15% and 90% [45]. What is important is that irAEs may be recognized even several months after their administration [46]. These AEs may be mild to life-threatening or even result in death and are classified in five grades according to the Common Terminology Criteria for Adverse Events (CTCAE), where fifth grade means death [47]. Renal irAEs are less common than those involving the skin, lungs, bowels, liver, or endocrine glands [48]. Their frequency is estimated at up to 2% of cases [23], albeit some researchers anticipate that acute kidney injury (AKI) may occur even in 29% of cases [49][50]. Of note, AKI is less common during monotherapy than while combining two ICIs [51]. Clinically, renal toxicities may present as AKI, proteinuria, and dyselectrolytemia. There are also numerous possible types of renal injury after administration of ICIs, but acute tubulointerstitial nephritis (ATIN) is the most frequent one [52]. Other types include lupus-like immune complex glomerulonephritis [53], minimal change disease (MCD) [54][55], membranous nephritis (MN) [56], focal segmental glomerulosclerosis (FSGS) [57] and thrombotic microangiopathy (TMA) [58]. The exact mechanism leading to systemic or organ injury during (or after) therapy with ICIs is still unclear and necessitates further studies. Four possible mechanisms were proposed leading to renal irAEs [59]. The first one embraces the fact of expression of immune checkpoint molecules such as PD-L1 in kidneys, which may protect healthy tissue from T cell infiltration and cytotoxicity. Therefore, blockade of PD-L1 may result in tissue damage [59][60][61]. Another mechanism concerns activated T cells that can infiltrate either normal tissue or tumor and, in both cases, recognize antigens by TCRs. In the first case, TCR binds to antigens expressed on healthy cells that sequences are similar enough to neoantigens [59]. The next proposed mechanism involved in kidney injury is extensive production of pro-inflammatory cytokines such as IL-1Ra, CXCL10 and TNF-α [62], but it is still unclear whether increased levels of serum cytokines are a cause or an effect of tissue damage [59]. Last but not least, ICIs may contribute to the synthesis of different autoantibodies damaging normal organs [59]. In terms of the kidneys, there is a described case of anti-double-stranded DNA antibodies occurring in a patient’s blood after administering ICIs [63]. Some investigators concluded that tubulointerstitial nephritis caused by ICIs presents some differences from the classical ATIN caused by other drugs. Draibe et al. compared 13 patients with renal injury after taking ICIs with 34 patients with tubulonephritis related to other drugs and suggested that patients with ATIN related to ICIs had lower serum creatinine levels at the time of diagnosis (3.8  ±  1.0  vs. 6.0  ±  4.1 mg/dL, p < 0.01), and that the time from starting the treatment with the responsible drug to the diagnosis was longer in this group (197  ±  185 vs. 114  ±  352 days, p < 0.01) [64]. This suggests a milder course of kidney damage caused by ICIs.

2.3. Risk Factors

In a cohort study including 309 patients who were given ICIs and where 51 of them (16.5%) developed AKI, Meraz-Muñoz et al. performed the identification of risk factors for ICI-induced nephrotoxicity. The presence of hypertension (OR 4.3; 95%CI: 1.8–6.1), and cerebrovascular disease (OR 9.2; 95%CI: 2.1–40), administration of angiotensin-converting enzyme inhibitors/angiotensin-receptor blockers (OR 2.9; 95%CI: 1.5–5.7), diuretics (OR 4.3; 95%CI: 1.9–9.8) and corticosteroids (OR 1.9; 95%CI: 1.1–3.6), and other irAEs (OR 3.2; 95%CI: 1.6–6.0) predicted development of AKI in an univariate analysis. However, the multivariable analysis revealed an association only with hypertension (OR 2.96; 95%CI: 1.33–6.59) and other irAEs (OR 2.82; 95%CI: 1.45–5.48) [65]. Cortazar et al. in a multicenter study with 138 patients receiving ICI therapy found a lower estimated glomerular filtration rate (eGFR) (OR 1.99; 95%CI: 1.43–2.76), usage of proton pump inhibitors (PPIs) (OR 2.38; 95%CI: 1.57–3.62) and combination of anti-CTLA-4 with anti-PD1/anti-PD-L1 drug (OR 2.71; 95%CI: 1.62–4.53) to be risk factors of AKI [66]. Similarly, another cohort study, which included 429 patients treated with ICIs and 429 control patients, confirmed that PPIs administration (OR 2.40; 95%CI: 1.79–3.23) and the presence of other irAEs (OR 2.07; 95%CI: 1.53–2.78) are risk factors of AKI in patients treated with the mentioned type of immunotherapy [67]. Of note, PPIs are known to be able to induce interstitial nephritis manifested by AKI. It is estimated that omeprazole may induce acute interstitial nephritis in 2–20/100,000 treated patients [68][69]. Their impact on AKI development in patients treated with ICIs was an object of interest in numerous studies. Apart from the research mentioned above, such an association was also documented in other studies [70][71].

2.4. Occurrence and Specific Nephrotoxicities

The first ICI to draw attention to possible renal adverse drug reactions was ipilimumab, an anti-CTLA-4 drug. In 2009 Fadel et al. noticed the possible harmful effects of ipilimumab on the kidneys. They reported a case of a 64-year-old man with metastatic melanoma who developed nephrotic syndrome after the treatment with this anti-CTLA-4 drug. The renal biopsy suggested lupus nephritis and anti-double-stranded DNA antibodies were detected. The treatment with ipilimumab was discontinued and prednisone was administered. After 3 months, anti-double-stranded DNA antibodies were undetectable and the nephrotic syndrome subsided [63]. In 2014, Izzedine et al. presented two case reports of patients with metastatic melanoma treated with ipilimumab with deteriorated kidney excretory function. In both cases a renal biopsy was performed and revealed interstitial inflammation. Both patients received prednisone administered orally and subsequently their kidney function improved [72]. In 2015 Thajudeen et al. described, as they claimed, the first case of biopsy-proven granulomatous interstitial nephritis after ipilimumab in a 74-year-old man with metastatic melanoma. The patient received treatment consisting of ipilimumab and dacarbazine. After the third cycle of therapy, the patient’s serum creatinine level doubled from 1.1–1.2 mg/dL to 2.2 mg/dL. Additionally, the patient complained of a rash. When the diagnosis was established based on biopsy, the treatment was interrupted and prednisone was applied. After 6 weeks kidney function improved. Finally, treatment with ipilimumab was resumed and the renal AE did not occur again [73]. Cortazar et al. in their work collected and summed up 13 cases of AKI after treatment with ICIs. Ten out of 13 patients were treated with ipilimumab alone or in combination. The period from starting the treatment to the development of AKI varied from 21 to 245 days with a median of 91 days. The median serum creatinine measured in these patients was 4.5 mg/dL. Seven patients had other irAEs recognized before the onset of AKI. All these patients had kidney biopsies performed. In 12 cases the histological diagnosis was ATIN and in one case it was TMA. Most of the patients (10) were treated with glucocorticoids and nine of them improved their renal function after the treatment. The remaining one, whose renal function did not recover after glucocorticoids, was the one with TMA. Patients who did not receive glucocorticoid therapy also did not improve their kidney function [51]. The ImmuNoTox study identified 14 ICI-induced AKI cases in 13 patients, retrospectively analyzing medical data from 352 patients treated with ICIs in one medical center in France. In most cases, the renal injury was classified as stage 1 (43%) and none of the patients needed hemodialysis therapy. Ten (77%) of these patients presented with irAEs affecting other systems. Six patients had renal biopsies which showed tubulointerstitial nephritis in all cases. The ICI therapy was withheld in all these patients and half of them received glucocorticoids. This had some limitations related to its retrospective character [25]. It is worth remembering the fact that ICI-induced AKI was described not only after the treatment with ipilimumab but after other ICIs as well. There were also reported cases of nephrotoxicities associated with pembrolizumab [51][74][75] and nivolumab [76][77][78]. As far as pembrolizumab, an anti-PD-1 monoclonal antibody, is concerned, Izzedine et al. described a series of renal AEs in patients treated with this drug in one medical center. It is observed a cohort consisting of 676 patients treated with pembrolizumab. In 12 participants (1.77%) renal side effects were observed, in 10 it was AKI, and in two proteinuria. In all mentioned cases of renal side effects, the kidney biopsy was performed, revealing acute tubular injury (ATI) in five patients, AIN in four patients, MCD alone in one patient, ATI and MCD in one patient, and finally nonspecific changes in one patient. In 10 patients pembrolizumab was withdrawn and seven of them received glucocorticoids. In one patient dialysis was started and this patient died in one month due to the progression of neoplastic disease. Others treated with glucocorticoids restored their renal function by about 50%. In one patient the treatment with pembrolizumab was restarted and resulted in an AIN relapse which was more severe. In patients who were not treated with glucocorticoids, the renal function remained stable. In two patients, in whom the treatment with pembrolizumab was maintained, their renal function improved [74]. In cases of biopsy-proven nephrotoxicity caused by nivolumab, the following findings occurred in the histological diagnosis: ATIN, IgA nephropathy, diffusive tubular injury, and complex-mediated glomerulonephritis. In the majority of the described cases, glucocorticoids were used in the treatment of these AEs resulting in renal function recovery [78]. Less frequent were renal AEs in patients treated with atezolizumab, durvalumab, avelumab and cemiplimab. Renal AEs were described for the first time for atezolizumab in a patient treated for renal cancer who developed AKI. The patient complained of an elevated body temperature and mild diarrhea. His blood tests revealed an elevation of serum creatinine to 5.6 mg/dL (in the previous tests the serum creatinine level was about 1.2 mg/dL). What is more, the urine tests showed proteinuria. This patient had a renal biopsy performed in which AIN was found. In the treatment methylprednisolone was used. The patient’s clinical improvement was observed after 8–10 weeks with partial normalization of serum creatinine level to 1.45 mg/dL [79]. In terms of durvalumab, there was a presented case of a patient who developed a nephrotic syndrome with MCD confirmed in a histological examination. The patient was treated with prednisolone and his symptoms withdrew [80]. In a phase II trial that included 88 patients treated with avelumab for chemotherapy-refractory metastatic Merkel cell carcinoma, four episodes of AKI occurred [81]. There is also reported a case of AKI with biopsy-proven AIN in a patient treated for squamous cell carcinoma of the skull with cemiplimab. The patient reported weakness and fatigue. His serum creatinine level was elevated in comparison to previous results (2.87 mg/dL and 1.3 mg/dL, respectively). The patient was treated with glucocorticoids and his renal function improved. The real incidence of AKI induced by ICIs is a subject of vivid debate. Cortazar et al. investigated the data from phase II and III clinical trials, with 3695 patients who were receiving ICIs. AKI occurred in 2.2% and severe AKI, defined as an increase of serum creatinine to a level higher than 4 mg/dL or tripling of initial creatinine level, emerged in 0.6% of patients. Furthermore, the incidence of AKI differed between the patients treated with various ICIs, ranging from 1.4% for pembrolizumab, 1.9% for nivolumab, and 2.0% for ipilimumab to 4.9% for combined therapy with ipilimumab and nivolumab [51]. However, some researchers suggest the real incidence of ICI-induced AKI may be much higher with a range of 9.9–29% [49].

2.5. Management and Outcomes

Due to more and more frequent use of ICIs in cancer treatment and still rising awareness of possible renal adverse effects of the mentioned therapy, both the ESMO and ASCO included recommendations on renal toxicities management in their guidelines concerning immunotherapy. ESMO guidelines divide patients with nephritis related to ICIs into four grades depending on serum creatinine elevation in relation to its baseline or upper limit of normal (ULN) (G1: 1.5 × baseline or >1.5 × ULN; G2: 1.5–3 × baseline or >1.5–3 ULN; G3: 3 × baseline or >3–6 ULN; G4: >6 × ULN) and recommend different strategies in each group. In general, according to these guidelines, every patient should have their serum sodium, potassium, creatinine, and urea level checked before each ICI application. In the case of abnormalities, other causes of renal function impairment such as dehydration, infection or obstruction in the urinary tract should be taken into account and then systematically excluded. What is more, potentially nephrotoxic drugs should be withdrawn. As for serious disturbance in renal parameters, ICI therapy should be suspended and administration of glucocorticoids should be considered (0.5–2 mg/kg/day methylprednisolone or equivalent). In dubious situations, renal biopsy may be taken into consideration as well as nephrology consultation [82]. Of note, urea level measurement is not recommended by nephrological guidelines for the diagnosis of AKI [83]. In the ASCO guidelines patients with worsened renal function caused by ICI therapy are also divided into four groups, but based on slightly different criteria which include direct creatinine elevation instead of elevation over ULN (G1: 1.5–2 × above baseline or >0.3 mg/dL; G2: 2–3 × above baseline; G3: >3 × above baseline or >4.0 mg/dL; G4: 6 × above baseline). Similarly to the ESMO guidelines this also highlights the importance of looking for other causes of kidney function deterioration. Diagnosis of ICI-induced AKI is empirical, and the biopsy is not indicated in most cases except for the ones not susceptible to standard treatment. In addition, the ASCO in the first line of treatment suggests glucocorticoids in doses of 0.5–2 mg/kg/day prednisone equivalents. Interestingly, they mention using other immunosuppressive drugs such as infliximab, azathioprine, cyclophosphamide, cyclosporine A, and mycophenolate in cases refractory to glucocorticoids [84]. The possible utility of infliximab [85] and mycophenolate [86][87] in the treatment of irAEs after ICI regimens was suggested in a few scientific paperesearchs. In the analysis performed by Cortazar et al. which included 138 patients with ICI-induced AKI, 40% had complete renal function recovery, while 45% had partial recovery and 15% did not improve their renal function after treatment. These patients were treated in 86% of cases with glucocorticoids and in 97% their ICI therapy was suspended. The therapy was resumed in 31 patients with previously diagnosed ICI-induced AKI and AKI relapsed in seven patients out of 31 (23%) [51]. An observational cohort study published by Baker et al. showed that AKI was associated with higher mortality in the group of patients treated with ICIs (HR 2.28; 95%CI: 1.90–2.72) but patients with AKI related to ICI therapy had significantly lower mortality (HR 0.43; 95%CI: 0.21–0.89) than patients with the other causes of AKI [88].

3. Tumor-Targeting Monoclonal Antibodies (TT-mAbs)

3.1. Mechanism of Action

There are several mechanisms where neoplastic cells may be affected by TT-mAbs. Generally, most frequently they are able to inhibit signaling pathways essential for both the survival and progression of cancer cells, which takes place as a result of modifying the receptor proteins function [89][90] or binding to specific tumor-associated antigens (TAA) [89][91]. Another common feature for a significant part of TT-mAbs is their ability to promote antibody-dependent cell-mediated cytotoxicity (ADCC) [92][93] or complement-dependent cytotoxicity [94], which is strictly connected with the opsonization of malignant cells. The most popular and widely used anticancer drugs among TT-mAbs include: (1) anti-human epidermal growth factor receptor-2 (HER-2) mAbs such as trastuzumab, trasuzumab emtasine [T-DM1] and pertuzumab mainly in breast cancer [95][96] and gastric/gastroesophageal junction cancer [97]; (2) anti-epidermal growth factor receptor (EGFR) mAbs represented by panitumumab and cetuximab which are utilized in the treatment of colorectal [98] and head and neck cancers [99][100]; (3) anti-CD20 mAbs exemplified by rituximab, obinutuzumab and ofatumumab administered mostly in hematological malignancies such as chronic lymphocytic leukemia (CLL) [101] and non-Hodgkin lymphomas (NHLs) [102]; (4) anti-CD30 mAb brentuximab vedotin and (5) anti-CD52 mAb alemtuzumab, which are also used especially in different hematological malignancies [103][104].

3.2. Renal Adverse Effects

Trastuzumab, an anti-HER-2 mAb, is known for its possible cardiotoxicity [105]. In a study comparing chemotherapy alone versus chemotherapy with trastuzumab, no significant difference in the occurrence of AKI in both groups was detected [106]. However, there is described a case report of a patient treated with ado-trastuzumab emtansine (T-DM1), a drug that consists of trastuzumab and maytansinoid DM1 which has cytotoxic properties, in whom nephrotic syndrome developed after the beginning of the therapy. In this patient, a renal biopsy revealed FSGS and ATI [107]. Worth mentioning is the fact that decreased kidney filtration existing before trastuzumab therapy may increase the risk of cardiotoxicity of this therapy [108]. In the case of the other anti-HER-2 mAb, pertuzumab, no important renal AEs were reported [109][110]. As far as anti-EGFR mAbs (panitumumab, cetuximab) are concerned, the most important renal AE of these therapies is dyselectrolytemia, in particular hypomagnesemia and hypokalemia [111][112]. A meta-analysis performed by Petrelli et al. showed that treatment with these mAbs may induce hypomagnesemia. The estimated incidence of lowered levels of serum magnesium (Mg2+) was 17% and is thought to be higher in the group treated with panitumumab than in patients treated with cetuximab [113]. Hypomagnesemia is believed to be caused by decreased activation of the renal EGFR, which results in lowered activation of the TRPM6 (transient receptor potential cation channel), leading to reduced reabsorption of Mg2+ [114]. In terms of hypokalemia in patients treated with cetuximab, hypokalemia of any grade was observed in 8% of patients [115], while in patients undergoing therapy with panitumumab hypokalemia of all grades was detected in 34% of patients [116]. The exact mechanism of hypokalemia is still not fully explained. Management of these electrolyte imbalances is based on watchful monitoring of patients at risk and proper supplementation of potassium and magnesium when needed [111]. Boku et al., in a post-marketing surveillance study assessing the safety of panitumumab in 3085 patients, found 12 (0.4%) renal and urinary disorders without further specifying its exact character [117]. There was also a case of nephrotic syndrome, AKI and leukocytoclastic vasculitis in a patient treated with panitumumab [118]. Furthermore, there were reported cases of AKIs and nephrotic syndromes in patients treated with cetuximab. Histopathological findings in these cases included crescentic diffuse proliferative glomerulonephritis [119], diffuse proliferative glomerulonephritis [120] and TMA [121]. The next group of mAbs contains an anti-cluster of differentiation 20 (anti-CD20) antibodies. The most common AEs caused by this group of mAbs are infusion-related reactions, infections and cytopenias caused by reversible myelosuppression [122][123][124]. In terms of kidneys, AKI may be caused by tumor lysis syndrome (TLS) which may occur after treatment with rituximab, obinutuzumab and ofatumumab; therefore, physicians supervising therapy with anti-CD20 agents should be aware of this possible side effect [125][126]. In addition, the infections mentioned above may affect the urinary tract [127]. The most common AEs in the course of treatment with anti-CD30 antibody conjugate with auristatin E include fatigue, nausea, diarrhea, neutropenia and peripheral sensory neuropathy [128]. In a study comparing the efficacy and safety of treatment with brentuximab vedotin versus treatment with pembrolizumab, in one (0.7%) patient out of 152 receiving brentuximab vedotin ATIN occurred, while in this group AKI or nephritis was not spotted [129]. Side effects of alemtuzumab, anti-CD52 mAb in most cases manifest as an influenza-like syndrome, transient cytopenias and increased susceptibility to infections [130]. In a study designed to assess the safety of therapy for CLL with alemtuzumab in which 149 patients received alemtuzumab, no particular nephrotoxicity was spotted [131]. However, in patients treated with alemtuzumab because of non-oncological indications, for multiple sclerosis, there were several cases of anti-glomerular basement membrane (anti-GBM) disease and membranous glomerulonephropathy [132]. Interestingly, there was noted a case of prostate and kidney aspergillosis in a patient treated for CLL connected with the immunosuppressive properties of alemtuzumab [133].

4. Chimeric Antigen Receptor T Cell (CAR-T Cell) Therapy

4.1. Mechanism of Action

CAR-T cell therapy is a particular example of adoptive T-cell therapy (ACT) which generally relies on using a patient’s own T cells to destroy cancer cells. These T cells need to be previously collected and then properly modified to enable them to recognize abnormal cells [134]. As far as CAR-T cells are concerned, preparing them starts with collecting the patient’s peripheral blood and further isolation of T cells using leukapheresis. Then T cells proliferated, and CARs are placed in their cell membrane using molecular biology techniques [135]. CARs are transmembrane proteins that consist of an extracellular part that binds to the selected antigen, a spacer/hinge part, a transmembrane part and an intracellular one that is involved in signal processing and T cell activation [136][137][138]. Following such a preparation, CAR-T cells are re-infused into patients’ circulation after the administration of the lymphodepleting chemotherapy [139]. Binding of CAR to the targeted antigen activates effector functions of T cells independently from MHC [140]. Activation of T cells induces the production of cytokines or cytotoxic activity with expected anti-cancer effects [141]. This type of therapy is mainly used in patients with refractory or resistant hematological malignancies such as B cell acute lymphoblastic leukemia (B-ALL) and diffuse large B-cell lymphoma (DLBCL). Nowadays, there are more and more attempts to use CAR-T cells to fight solid tumors [142][143][144][145].

4.2. Renal Adverse Effects and Their Pathomechanisms

One of the main limitations of CAR-T cell therapy is its toxicity, often severe and life-threatening. Predominantly AEs of this therapy include cytokine release syndrome (CRS) and neurotoxicity, also called CAR T-cell related encephalopathy syndrome (CRES) [146]. CRS is associated with a massive production of cytokines in response to the binding of CAR to the targeted antigen and following activation of the immune response. Main cytokines involved in CRS include IL-6, IL-10 and interferon (IFN-γ) [146][147][148][149]. Symptoms and severity of CRS vary among patients, starting from influenza-like symptoms to the dysfunction of almost all organs and systems [150][151][152]. There are some grading systems used to assess the severity of CRS [153][154]. Renal AEs include AKI related to CRS [155], related to prerenal and renal mechanisms [156]. Prerenal AKI after CAR-T treatment is associated with impaired renal perfusion caused predominantly by CRS complications such as fever or vomiting, which may lead to dehydration resulting in a reduction in the intravascular volume [157]. In addition, severe CRS may lead to vasodilation, capillary leak syndrome and reduction of cardiac output. All of these affect kidney perfusion and result in a decrease in the glomerular filtration rate [158][159]. The renal mechanisms of AKI are also the consequences of prolonged hypovolemia leading to tubular ischaemic injury [160] and direct tubular toxicity of cytokines [161][162]. As a result of treatment and damage to neoplastic cells, tumor lysis syndrome (TLS) may develop. This syndrome is caused by the release of the contents from destroyed cells, inter alia intracellular ions, nucleic acids, proteins, and their metabolites. Substances such as uric acid and phosphate may contribute to the damaging of renal tubules when they precipitate, which in consequence leads to renal function impairment [163][164]. Other possible mechanisms of nephrotoxicity of CAR-T therapy are associated with the consequences of the development of hemophagocytic lymphohistiocytosis (HLH) in which AIN or TMA may be identified [165][166][167].

4.3. Occurrence and Outcomes

In a study performed by Gupta et al., researchers evaluated the incidence of AKI in 78 patients treated with axicabtagene ciloleucel (YESCARTA®) or tisagenlecleucel (KYMRIAH®) for refractory DLBCL. Among 15 patients (19%) with AKI, eight of them had lowered kidney perfusion, six developed ATN and one had an urinary tract obstruction related to the progression of the lymphoma. Grade 3 of AKI was confirmed in six patients and three of them required kidney replacement therapy. However, the average length of hospitalization and 60-day mortality was similar in patients with and without AKI [168]. Gutgarts et al. analyzed data from 46 adult patients treated for Non-Hodgkin lymphoma (NHL) with axicabtagene ciloleucel (YESCARTA®) or tisagenlecleucel (KYMRIAH®). They assessed kidney function up to 100 days after initiation of the treatment. They reported AKI of any grade in 14 (30% of patients) and grade 2 or 3 in 4 (8.7%) patients. None of the patients required kidney replacement therapy and most of them recovered kidney function within 30 days [169]. Another study included 38 patients treated with tisagenlecleucel (KYMRIAH®) for DLBCL. AKI was diagnosed in two (5%) patients; both of them had grade 3 AKI. One of them died 4 days after treatment and the second one 28 days after treatment [170]. Finally, a research performed by Kanduri et al., based on 22 cohort studies including 3376 patients treated with CAR-T cells, revealed an 18.6% (95%CI: 14.3–23.8) incidence of AKI, while 4.4% (95%CI: 2.1–8.9) of patients required renal replacement therapy [171].

4.4. Management

Patients who underwent CAR-T cells therapy and developed AKI should have been treated for the cause of renal function impairment. In the case of a prerenal mechanism, patients with hypovolemia should receive proper fluid resuscitation and vasopressors when needed. In such cases, norepinephrine is the first-choice drug for these patients [172]. Those with a clinically significant deterioration in cardiac output should be considered to be candidates for inotropic agents such as milrinone, dopamine, epinephrine, norepinephrine, or vasopressin [173]. Patients who developed AKI in the course of CRS or HLH should receive supportive care and proper treatment for these disease entities. ASCO guidelines cover these issues in detail [174]. The use of tocilizumab—an anti-human interleukin-6 receptor (anti-IL-6R) monoclonal antibody (mAb)— or in some cases of CRS, may be beneficial [175]. Treatment of HLH is based on immunosuppression with glucocorticoids, IL-6 antagonists or etoposide [174][176]. When it comes to TLS, patients at risk should be identified before therapy and proper precautions should be taken. Hydration and administration of hypouricemic drugs such as allopurinol or rasburicase should be considered [177].

References

  1. Dyba, T.; Randi, G.; Martos, C.; Giusti, F.; Calvalho, R.; Neamtiu, L.; Nicholson, N.; Flego, M.; Dimitrova, N.; Bettio, M. 1501O Long-term estimates of cancer incidence and mortality for the EU and EFTA countries according to different demographic scenarios. Ann. Oncol. 2021, 32, S1102.
  2. Rahib, L.; Wehner, M.R.; Matrisian, L.M.; Nead, K.T. Estimated Projection of US Cancer Incidence and Death to 2040. JAMA Netw. Open 2021, 4, e214708.
  3. Schmielau, J.; Rick, O.; Reuss-Borst, M.; Kalusche-Bontemps, E.-M.; Steimann, M. Rehabilitation of Cancer Survivors with Long-Term Toxicities. Oncol. Res. Treat. 2017, 40, 764–771.
  4. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674.
  5. Zindl, C.L.; Chaplin, D.D. Tumor Immune Evasion. Science 2010, 328, 697–698.
  6. Yang, L.; Pang, Y.; Moses, H.L. TGF-β and immune cells: An important regulatory axis in the tumor microenvironment and progression. Trends Immunol. 2010, 31, 220–227.
  7. Shields, J.D.; Kourtis, I.C.; Tomei, A.A.; Roberts, J.M.; Swartz, M.A. Induction of Lymphoidlike Stroma and Immune Escape by Tumors That Express the Chemokine CCL21. Science 2010, 328, 749–752.
  8. Mougiakakos, D.; Choudhury, A.; Lladser, A.; Kiessling, R.; Johansson, C.C. Regulatory T Cells in Cancer. Adv. Cancer Res. 2010, 107, 57–117.
  9. Ostrand-Rosenberg, S.; Sinha, P. Myeloid-Derived Suppressor Cells: Linking Inflammation and Cancer. J. Immunol. 2009, 182, 4499–4506.
  10. Ohue, Y.; Nishikawa, H. Regulatory T (Treg) cells in cancer: Can Treg cells be a new therapeutic target? Cancer Sci. 2019, 110, 2080–2089.
  11. Zhang, Y.; Zhang, Z. The history and advances in cancer immunotherapy: Understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell. Mol. Immunol. 2020, 17, 807–821.
  12. Coley, W.B. The Treatment of Malignat Tumors by Repeated Inoculations of Erysipelas. Am. J. Med Sci. 1893, 105, 487–510.
  13. Wiemann, B.; Starnes, C.O. Coley’s toxins, tumor necrosis factor and cancer research: A historical perspective. Pharmacol. Ther. 1994, 64, 529–564.
  14. Sathyanarayanan, V.; Neelapu, S.S. Cancer immunotherapy: Strategies for personalization and combinatorial approaches. Mol. Oncol. 2015, 9, 2043–2053.
  15. Karmakar, S.; Dhar, R.; Seethy, A.; Singh, S.; Pethusamy, K.; Srivastava, T.; Talukdar, J.; Rath, G.K. Cancer immunotherapy: Recent advances and challenges. J. Cancer Res. Ther. 2021, 17, 834.
  16. Velcheti, V.; Schalper, K. Basic Overview of Current Immunotherapy Approaches in Cancer. Am. Soc. Clin. Oncol. Educ. Book 2016, 35, 298–308.
  17. Leach, D.R.; Krummel, M.F.; Allison, J.P. Enhancement of Antitumor Immunity by CTLA-4 Blockade. Science 1996, 271, 1734–1736.
  18. Kwon, E.D.; Hurwitz, A.A.; Foster, B.A.; Madias, C.; Feldhaus, A.L.; Greenberg, N.M.; Burg, M.B.; Allison, J.P. Manipulation of T cell costimulatory and inhibitory signals for immunotherapy of prostate cancer. Proc. Natl. Acad. Sci. USA 1997, 94, 8099–8103.
  19. Ishida, Y.; Agata, Y.; Shibahara, K.; Honjo, T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992, 11, 3887–3895.
  20. Han, D.; Xu, Z.; Zhuang, Y.; Ye, Z.; Qian, Q. Current Progress in CAR-T Cell Therapy for Hematological Malignancies. J. Cancer 2021, 12, 326–334.
  21. Salter, A.; Pont, M.J.; Riddell, S.R. Chimeric antigen receptor–modified T cells: CD19 and the road beyond. Blood 2018, 131, 2621–2629.
  22. Darnell, E.P.; Mooradian, M.J.; Baruch, E.N.; Yilmaz, M.; Reynolds, K.L. Immune-Related Adverse Events (irAEs): Diagnosis, Management, and Clinical Pearls. Curr. Oncol. Rep. 2020, 22, 1–11.
  23. Martins, F.; Sofiya, L.; Sykiotis, G.P.; Lamine, F.; Maillard, M.; Fraga, M.; Shabafrouz, K.; Ribi, C.; Cairoli, A.; Guex-Crosier, Y.; et al. Adverse effects of immune-checkpoint inhibitors: Epidemiology, management and surveillance. Nat. Rev. Clin. Oncol. 2019, 16, 563–580.
  24. Barber, F.D. Adverse Events of Oncologic Immunotherapy and Their Management. Asia-Pacific J. Oncol. Nurs. 2019, 6, 212–226.
  25. Espi, M.; Teuma, C.; Novel-Catin, E.; Maillet, D.; Souquet, P.; Dalle, S.; Koppe, L.; Fouque, D. Renal adverse effects of immune checkpoints inhibitors in clinical practice: ImmuNoTox study. Eur. J. Cancer 2021, 147, 29–39.
  26. El Rassy, E.; Kourie, H.R.; Rizkallah, J.; El Karak, F.; Hanna, C.; Chelala, D.N.; Ghosn, M. Immune checkpoint inhibitors renal side effects and management. Immunotherapy 2016, 8, 1417–1425.
  27. Swann, J.B.; Smyth, M.J. Immune surveillance of tumors. J. Clin. Investig. 2007, 117, 1137–1146.
  28. Zhang, Z.; Lu, M.; Qin, Y.; Gao, W.; Tao, L.; Su, W.; Zhong, J. Neoantigen: A New Breakthrough in Tumor Immunotherapy. Front. Immunol. 2021, 12, 672356.
  29. Gaudino, S.J.; Kumar, P. Cross-Talk Between Antigen Presenting Cells and T Cells Impacts Intestinal Homeostasis, Bacterial Infections, and Tumorigenesis. Front. Immunol. 2019, 10, 360.
  30. Wang, S.; He, Z.; Wang, X.; Li, H.; Liu, X.-S. Antigen presentation and tumor immunogenicity in cancer immunotherapy response prediction. eLife 2019, 8, e49020.
  31. Zagorulya, M.; Duong, E.; Spranger, S. Impact of anatomic site on antigen-presenting cells in cancer. J. Immunother. Cancer 2020, 8, e001204.
  32. Diesendruck, Y.; Benhar, I. Novel immune check point inhibiting antibodies in cancer therapy—Opportunities and challenges. Drug Resist. Updat. 2017, 30, 39–47.
  33. Collins, M.; Ling, V.; Carreno, B.M. The B7 family of immune-regulatory ligands. Genome Biol. 2005, 6, 223.
  34. Lanzavecchia, A.; Sallusto, F. The instructive role of dendritic cells on T cell responses: Lineages, plasticity and kinetics. Curr. Opin. Immunol. 2001, 13, 291–298.
  35. Chen, D.S.; Mellman, I. Oncology Meets Immunology: The Cancer-Immunity Cycle. Immunity 2013, 39, 1–10.
  36. Horton, B.L.; Fessenden, T.B.; Spranger, S. Tissue Site and the Cancer Immunity Cycle. Trends Cancer 2019, 5, 593–603.
  37. Cai, X.; Zhan, H.; Ye, Y.; Yang, J.; Zhang, M.; Li, J.; Zhuang, Y. Current Progress and Future Perspectives of Immune Checkpoint in Cancer and Infectious Diseases. Front. Genet. 2021, 12, 785153.
  38. He, X.; Xu, C. Immune checkpoint signaling and cancer immunotherapy. Cell Res. 2020, 30, 660–669.
  39. Collins, A.V.; Brodie, D.W.; Gilbert, R.J.C.; Iaboni, A.; Manso-Sancho, R.; Walse, B.; Stuart, D.I.; van der Merwe, P.A.; Davis, S.J. The Interaction Properties of Costimulatory Molecules Revisited. Immunity 2002, 17, 201–210.
  40. Chambers, C.A.; Kuhns, M.S.; Egen, J.G.; Allison, J.P. CTLA-4-Mediated Inhibition in Regulation of T Cell Responses: Mechanisms and Manipulation in Tumor Immunotherapy. Annu. Rev. Immunol. 2001, 19, 565–594.
  41. Jiang, Y.; Chen, M.; Nie, H.; Yuan, Y. PD-1 and PD-L1 in cancer immunotherapy: Clinical implications and future considerations. Hum. Vaccines Immunother. 2019, 15, 1111–1122.
  42. Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am. J. Clin. Oncol. 2016, 39, 98–106.
  43. Bagchi, S.; Yuan, R.; Engleman, E.G. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu. Rev. Pathol. Mech. Dis. 2021, 16, 223–249.
  44. Esfahani, K.; Meti, N.; Miller, W.H.; Hudson, M. Adverse events associated with immune checkpoint inhibitor treatment for cancer. Can. Med Assoc. J. 2019, 191, E40–E46.
  45. Winer, A.; Bodor, J.N.; Borghaei, H. Identifying and managing the adverse effects of immune checkpoint blockade. J. Thorac. Dis. 2018, 10, S480–S489.
  46. Tang, S.-Q.; Tang, L.-L.; Mao, Y.-P.; Li, W.-F.; Chen, L.; Zhang, Y.; Guo, Y.; Liu, Q.; Sun, Y.; Xu, C.; et al. The Pattern of Time to Onset and Resolution of Immune-Related Adverse Events Caused by Immune Checkpoint Inhibitors in Cancer: A Pooled Analysis of 23 Clinical Trials and 8436 Patients. Cancer Res. Treat. 2021, 53, 339–354.
  47. Common Terminology Criteria for Adverse Events (CTCAE) Common Terminology Criteria for Adverse Events (CTCAE) V5.0; 2017. Available online: http://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/ctcae_v5_quick_reference_5x7.pdf (accessed on 25 May 2022).
  48. Xu, C.; Chen, Y.; Du, X.-J.; Liu, J.-Q.; Huang, C.-L.; Chen, L.; Zhou, G.-Q.; Li, W.-F.; Mao, Y.-P.; Hsu, C.; et al. Comparative safety of immune checkpoint inhibitors in cancer: Systematic review and network meta-analysis. BMJ 2018, 363, k4226.
  49. Wanchoo, R.; Karam, S.; Uppal, N.N.; Barta, V.S.; Deray, G.; Devoe, C.; Launay-Vacher, V.; Jhaveri, K.D.; on behalf of Cancer and Kidney International Network Workgroup on Immune Checkpoint Inhibitors. Adverse Renal Effects of Immune Checkpoint Inhibitors: A Narrative Review. Am. J. Nephrol. 2017, 45, 160–169.
  50. Mamlouk, O.; Abudayyeh, A. Cancer immunotherapy and its renal effects. J. Onco-Nephrol. 2019, 3, 151–159.
  51. Cortazar, F.B.; Marrone, K.A.; Troxell, M.L.; Ralto, K.M.; Hoenig, M.P.; Brahmer, J.R.; Le, D.T.; Lipson, E.J.; Glezerman, I.G.; Wolchok, J.; et al. Clinicopathological features of acute kidney injury associated with immune checkpoint inhibitors. Kidney Int. 2016, 90, 638–647.
  52. Eijgelsheim, M.; Sprangers, B. Kidney Biopsy Should Be Performed to Document the Cause of Immune Checkpoint Inhibitor–Associated Acute Kidney Injury: PRO. Kidney360 2020, 1, 158–161.
  53. Izzedine, H.; Mateus, C.; Boutros, C.; Robert, C.; Rouvier, P.; Amoura, Z.; Mathian, A. Renal effects of immune checkpoint inhibitors. Nephrol. Dial. Transplant. 2016, 32, 936–942.
  54. Gao, B.; Lin, N.; Wang, S.; Wang, Y. Minimal change disease associated with anti-PD1 immunotherapy: A case report. BMC Nephrol. 2018, 19, 156.
  55. Bickel, A.; Koneth, I.; Enzler-Tschudy, A.; Neuweiler, J.; Flatz, L.; Früh, M. Pembrolizumab-associated minimal change disease in a patient with malignant pleural mesothelioma. BMC Cancer 2016, 16, 656.
  56. Wakabayashi, K.; Yamamoto, S.; Hara, S.; Okawara, M.; Teramoto, K.; Ikeda, N.; Kusunoki, Y.; Takeji, M. Nivolumab-induced membranous nephropathy in a patient with stage IV lung adenocarcinoma. CEN Case Rep. 2022, 11, 171–176.
  57. Kim, D.W.; Jeon, H.; Kim, S.; Lee, W.; Kim, H.J.; Rhee, H.; Song, S.H.; Seong, E.Y. Pembrolizumab-induced focal segmental glomerulosclerosis. Medicine 2021, 100, e27546.
  58. Hayata, M.; Shimanuki, M.; Ko, T.; Date, R.; Hamaguchi, A.; Tominaga, A.; Miura, R.; Mizumoto, T.; Mukoyama, M. Pembrolizumab-associated thrombotic microangiopathy in a patient with urothelial cancer: A case report and literature review. Ren. Replace. Ther. 2020, 6, 1–6.
  59. Sury, K.; Perazella, M.A.; Shirali, A.C. Cardiorenal complications of immune checkpoint inhibitors. Nat. Rev. Nephrol. 2018, 14, 571–588.
  60. Ding, H.; Wu, X.; Gao, W. PD-L1 is expressed by human renal tubular epithelial cells and suppresses T cell cytokine synthesis. Clin. Immunol. 2005, 115, 184–191.
  61. Waeckerle-Men, Y.; Starke, A.; Wüthrich, R.P. PD-L1 partially protects renal tubular epithelial cells from the attack of CD8+cytotoxic T cells. Nephrol. Dial. Transplant. 2007, 22, 1527–1536.
  62. Murakami, N.; Borges, T.J.; Yamashita, M.; Riella, L.V. Severe acute interstitial nephritis after combination immune-checkpoint inhibitor therapy for metastatic melanoma. Clin. Kidney J. 2016, 9, 411–417.
  63. Fadel, F.; El Karoui, K.; Knebelmann, B. Anti-CTLA4 Antibody–Induced Lupus Nephritis. N. Engl. J. Med. 2009, 361, 211–212.
  64. Draibe, J.B.; García-Carro, C.; Martinez-Valenzuela, L.; Agraz, I.; Fulladosa, X.; Bolufer, M.; Tango, A.; Torras, J.; Soler, M.J. Acute tubulointerstitial nephritis induced by checkpoint inhibitors versus classical acute tubulointerstitial nephritis: Are they the same disease? Clin. Kidney J. 2020, 14, 884–890.
  65. Meraz-Muñoz, A.; Amir, E.; Ng, P.; Avila-Casado, C.; Ragobar, C.; Chan, C.; Kim, J.; Wald, R.; Kitchlu, A. Acute kidney injury associated with immune checkpoint inhibitor therapy: Incidence, risk factors and outcomes. J. Immunother. Cancer 2020, 8, e000467.
  66. Cortazar, F.B.; Kibbelaar, Z.A.; Glezerman, I.G.; Abudayyeh, A.; Mamlouk, O.; Motwani, S.S.; Murakami, N.; Herrmann, S.M.; Manohar, S.; Shirali, A.C.; et al. Clinical Features and Outcomes of Immune Checkpoint Inhibitor–Associated AKI: A Multicenter Study. J. Am. Soc. Nephrol. 2020, 31, 435–446.
  67. Gupta, S.; Short, S.A.P.; E Sise, M.; Prosek, J.M.; Madhavan, S.M.; Soler, M.J.; Ostermann, M.; Herrmann, S.M.; Abudayyeh, A.; Anand, S.; et al. Acute kidney injury in patients treated with immune checkpoint inhibitors. J. Immunother. Cancer 2021, 9, e003467.
  68. Nast, C.C. Medication-Induced Interstitial Nephritis in the 21st Century. Adv. Chronic Kidney Dis. 2017, 24, 72–79.
  69. Al-Aly, Z.; Maddukuri, G.; Xie, Y. Proton Pump Inhibitors and the Kidney: Implications of Current Evidence for Clinical Practice and When and How to Deprescribe. Am. J. Kidney Dis. 2020, 75, 497–507.
  70. Seethapathy, H.; Zhao, S.; Chute, D.F.; Zubiri, L.; Oppong, Y.; Strohbehn, I.; Cortazar, F.B.; Leaf, D.E.; Mooradian, M.J.; Villani, A.-C.; et al. The Incidence, Causes, and Risk Factors of Acute Kidney Injury in Patients Receiving Immune Checkpoint Inhibitors. Clin. J. Am. Soc. Nephrol. 2019, 14, 1692–1700.
  71. Kato, K.; Mizuno, T.; Koseki, T.; Ito, Y.; Hatano, M.; Takahashi, K.; Yamada, S.; Tsuboi, N. Concomitant Proton Pump Inhibitors and Immune Checkpoint Inhibitors Increase Nephritis Frequency. Vivo 2021, 35, 2831–2840.
  72. Izzedine, H.; Gueutin, V.; Gharbi, C.; Mateus, C.; Robert, C.; Routier, E.; Thomas, M.; Baumelou, A.; Rouvier, P. Kidney injuries related to ipilimumab. Investig. New Drugs 2014, 32, 769–773.
  73. Thajudeen, B.; Madhrira, M.; Bracamonte, E.; Cranmer, L.D. Ipilimumab Granulomatous Interstitial Nephritis. Am. J. Ther. 2015, 22, e84–e87.
  74. Izzedine, H.; Mathian, A.; Champiat, S.; Picard, C.; Mateus, C.; Routier, E.; Varga, A.; Malka, D.; Leary, A.; Michels, J.; et al. Renal toxicities associated with pembrolizumab. Clin. Kidney J. 2018, 12, 81–88.
  75. Ishibuchi, K.; Iwakura, T.; Kaneko, M.; Fukasawa, H.; Furuya, R. Pembrolizumab-associated nephrotic syndrome recovered from transient hemodialysis in a patient with lung cancer. CEN Case Rep. 2020, 9, 215–219.
  76. Stein, C.; Burtey, S.; Mancini, J.; Pelletier, M.; Sallée, M.; Brunet, P.; Berbis, P.; Grob, J.J.; Honoré, S.; Gaudy, C.; et al. Acute kidney injury in patients treated with anti-programmed death receptor-1 for advanced melanoma: A real-life study in a single-centre cohort. Nephrol. Dial. Transplant. 2020, 36, 1664–1674.
  77. Uchida, N.; Tsuji, S.; Fujita, K.; Koizumi, M.; Moriyoshi, K.; Mio, T. Nivolumab-induced severe acute kidney injury with a long latent phase in a patient with non-small-cell lung cancer: A case report. Clin. Case Rep. 2018, 6, 2185–2188.
  78. Georgianos, P.I.; Vaios, V.; Leontaridou, E.; Karayannopoulou, G.; Koletsa, T.; Sioulis, A.; Balaskas, E.V.; Zebekakis, P.E. Acute Interstitial Nephritis in a Patient with Non-Small Cell Lung Cancer under Immunotherapy with Nivolumab. Case Rep. Nephrol. 2019, 2019, 1–5.
  79. Xipell, M.; Victoria, I.; Hoffmann, V.; Villarreal, J.; Garcia-Herrera, A.; Reig, O.; Rodas, L.; Blasco, M.; Poch, E.; Mellado, B.; et al. Acute tubulointerstitial nephritis associated with atezolizumab, an anti-programmed death-ligand 1 (pd-l1) antibody therapy. OncoImmunology 2018, 7, e1445952.
  80. Toda, M.G.; Fujii, K.; Kato, A.; Yoshifuji, A.; Komatsu, M.; Amino, Y.; Kitazono, S.; Hashiguchi, A.; Ryuzaki, M. Minimal Change Disease Associated with Durvalumab. Kidney Int. Rep. 2021, 6, 2733–2734.
  81. Kaufman, H.L.; Russell, J.; Hamid, O.; Bhatia, S.; Terheyden, P.; D’Angelo, S.P.; Shih, K.C.; Lebbé, C.; Linette, G.P.; Milella, M.; et al. Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: A multicentre, single-group, open-label, phase 2 trial. Lancet Oncol. 2016, 17, 1374–1385.
  82. Haanen, J.; Carbonnel, F.; Robert, C.; Kerr, K.; Peters, S.; Larkin, J.; Jordan, K. Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2017, 28, iv119–iv142.
  83. Khwaja, A. KDIGO Clinical Practice Guidelines for Acute Kidney Injury. Nephron Exp. Nephrol. 2012, 120, c179–c184.
  84. Schneider, B.J.; Naidoo, J.; Santomasso, B.D.; Lacchetti, C.; Adkins, S.; Anadkat, M.; Atkins, M.B.; Brassil, K.J.; Caterino, J.M.; Chau, I.; et al. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: ASCO Guideline Update. J. Clin. Oncol. 2021, 39, 4073–4126.
  85. Lin, J.S.; Mamlouk, O.; Selamet, U.; Tchakarov, A.; Glass, W.F.; Sheth, R.A.; Layman, R.M.; Dadu, R.; Abdel-Wahab, N.; Abdelrahim, M.; et al. Infliximab for the treatment of patients with checkpoint inhibitor associated acute tubular interstitial nephritis. OncoImmunology 2021, 10, 1877415.
  86. Jessel, S.; Austin, M.; Kluger, H.M. Mycophenolate as Primary Treatment for Immune Checkpoint Inhibitor Induced Acute Kidney Injury in a Patient with Concurrent Immunotherapy-Associated Diabetes: A Case Report. Clin. Oncol. Case Rep. 2021, 4, 156.
  87. Omori, G.; Takada, K.; Murase, K.; Hayasaka, N.; Nakamura, H.; Iyama, S.; Ohnuma, H.; Miyanishi, K.; Fukuta, F.; Tanaka, T.; et al. Successful mycophenolate mofetil treatment of a patient with severe steroid-refractory hepatitis evoked by nivolumab plus ipilimumab treatment for relapsed bladder cancer. Clin. Case Rep. 2020, 9, 654–659.
  88. Baker, M.L.; Yamamoto, Y.; A Perazella, M.; Dizman, N.; Shirali, A.C.; Hafez, N.; Weinstein, J.; Simonov, M.; Testani, J.M.; Kluger, H.M.; et al. Mortality after acute kidney injury and acute interstitial nephritis in patients prescribed immune checkpoint inhibitor therapy. J. Immunother. Cancer 2022, 10, e004421.
  89. Galluzzi, L.; Vacchelli, E.; Bravo-San Pedro, J.M.; Buqué, A.; Senovilla, L.; Baracco, E.E.; Bloy, N.; Castoldi, F.; Abastado, J.-P.; Agostinis, P.; et al. Classification of current anticancer immunotherapies. Oncotarget 2014, 5, 12472–12508.
  90. Kaplan-Lefko, P.J.; Graves, J.D.; Zoog, S.J.; Pan, Y.; Wall, J.; Branstetter, D.G.; Moriguchi, J.; Coxon, A.; Huard, J.N.; Xu, R.; et al. Conatumumab, a fully human agonist antibody to death receptor 5, induces apoptosis via caspase activation in multiple tumor types. Cancer Biol. Ther. 2010, 9, 618–631.
  91. Coulie, P.G.; Van den Eynde, B.J.; Van Der Bruggen, P.; Boon, T. Tumour antigens recognized by T lymphocytes: At the core of cancer immunotherapy. Nat. Rev. Cancer 2014, 14, 135–146.
  92. Hubert, P.; Amigorena, S. Antibody-dependent cell cytotoxicity in monoclonal antibody-mediated tumor immunotherapy. OncoImmunology 2012, 1, 103–105.
  93. Kawaguchi, Y.; Kono, K.; Mimura, K.; Sugai, H.; Akaike, H.; Fujii, H. Cetuximab induce antibody-dependent cellular cytotoxicity against EGFR-expressing esophageal squamous cell carcinoma. Int. J. Cancer 2006, 120, 781–787.
  94. Zipfel, P.F.; Skerka, C. Complement regulators and inhibitory proteins. Nat. Rev. Immunol. 2009, 9, 729–740.
  95. Loibl, S.; Gianni, L. HER2-positive breast cancer. Lancet 2016, 389, 2415–2429.
  96. Verma, S.; Miles, D.; Gianni, L.; Krop, I.E.; Welslau, M.; Baselga, J.; Pegram, M.; Oh, D.-Y.; Diéras, V.; Guardino, E.; et al. Trastuzumab Emtansine for HER2-Positive Advanced Breast Cancer. N. Engl. J. Med. 2012, 367, 1783–1791.
  97. Boku, N. HER2-positive gastric cancer. Gastric Cancer 2013, 17, 1–12.
  98. Martinelli, E.; Ciardiello, D.; Martini, G.; Troiani, T.; Cardone, C.; Vitiello, P.; Normanno, N.; Rachiglio, A.; Maiello, E.; Latiano, T.; et al. Implementing anti-epidermal growth factor receptor (EGFR) therapy in metastatic colorectal cancer: Challenges and future perspectives. Ann. Oncol. 2020, 31, 30–40.
  99. Guidi, A.; Codecà, C.; Ferrari, D. Chemotherapy and immunotherapy for recurrent and metastatic head and neck cancer: A systematic review. Med. Oncol. 2018, 35, 37.
  100. Martínez-Trufero, J.; Borbalas, A.L.; Bernad, I.P.; Sanz, M.T.; Izquierdo, E.O.; Cirauqui, B.C.; Rubió-Casadevall, J.; Serrahima, M.P.; Ortega, J.P.; Toledano, I.P.; et al. Sequential chemotherapy regimen of induction with panitumumab and paclitaxel followed by radiotherapy and panitumumab in patients with locally advanced head and neck cancer unfit for platinum derivatives. The phase II, PANTERA/TTCC-2010-06 study. Clin. Transl. Oncol. 2021, 23, 1666–1677.
  101. Brown, J.R.; Cymbalista, F.; Sharman, J.; Jacobs, I.; Nava-Parada, P.; Mato, A. The Role of Rituximab in Chronic Lymphocytic Leukemia Treatment and the Potential Utility of Biosimilars. Oncologist 2017, 23, 288–296.
  102. Klein, C.; Jamois, C.; Nielsen, T. Anti-CD20 treatment for B-cell malignancies: Current status and future directions. Expert Opin. Biol. Ther. 2020, 21, 161–181.
  103. Van Der Weyden, C.; Dickinson, M.; Whisstock, J.; Prince, H.M. Brentuximab vedotin in T-cell lymphoma. Expert Rev. Hematol. 2018, 12, 5–19.
  104. Ravandi, F.; O’Brien, S. Alemtuzumab in CLL and Other Lymphoid Neoplasms. Cancer Investig. 2006, 24, 718–725.
  105. Nemeth, B.T.; Varga, Z.V.; Wu, W.J.; Pacher, P. Trastuzumab cardiotoxicity: From clinical trials to experimental studies. J. Cereb. Blood Flow Metab. 2016, 174, 3727–3748.
  106. Slamon, D.; Eiermann, W.; Robert, N.; Pienkowski, T.; Martin, M.; Press, M.; Mackey, J.; Glaspy, J.; Chan, A.; Pawlicki, M.; et al. Adjuvant Trastuzumab in HER2-Positive Breast Cancer. N. Engl. J. Med. 2011, 365, 1273–1283.
  107. Hakroush, S.; Wulf, S.; Gallwas, J.; Tampe, B. Case Report: Collapsing Focal Segmental Glomerulosclerosis After Initiation of Ado-Trastuzumab Emtansine Therapy. Front. Oncol. 2021, 11, 5007.
  108. Russo, G.; Cioffi, G.; Di Lenarda, A.; Tuccia, F.; Bovelli, D.; Di Tano, G.; Alunni, G.; Gori, S.; Faggiano, P.; Tarantini, L. Role of renal function on the development of cardiotoxicity associated with trastuzumab-based adjuvant chemotherapy for early breast cancer. Intern. Emerg. Med. 2012, 7, 439–446.
  109. Ishii, K.; Morii, N.; Yamashiro, H. Pertuzumab in the treatment of HER2-positive breast cancer: An evidence-based review of its safety, efficacy, and place in therapy. Core EÉvid. 2019, 14, 51–70.
  110. Zhu, C.; Ling, W.; Zhang, J.; Gao, H.; Shen, K.; Ma, X. Safety and efficacy evaluation of pertuzumab in patients with solid tumors. Medicine 2017, 96, e6870.
  111. Abbas, A.; Mirza, M.M.; Ganti, A.K.; Tendulkar, K. Renal Toxicities of Targeted Therapies. Target. Oncol. 2015, 10, 487–499.
  112. Cosmai, L.; Gallieni, M.; Porta, C. Renal toxicity of anticancer agents targeting HER2 and EGFR. J. Nephrol. 2015, 28, 647–657.
  113. Petrelli, F.; Borgonovo, K.; Cabiddu, M.; Ghilardi, M.; Barni, S. Risk of anti-EGFR monoclonal antibody-related hypomagnesemia: Systematic review and pooled analysis of randomized studies. Expert Opin. Drug Saf. 2011, 11, S9–S19.
  114. Groenestege, W.M.T.; Thébault, S.; van der Wijst, J.; Berg, D.V.D.; Janssen, R.; Tejpar, S.; Heuvel, L.P.V.D.; Van Cutsem, E.; Hoenderop, J.G.; Knoers, N.V.; et al. Impaired basolateral sorting of pro-EGF causes isolated recessive renal hypomagnesemia. J. Clin. Investig. 2007, 117, 2260–2267.
  115. Cao, Y.; Liu, L.; Liao, C.; Tan, A.; Gao, F. Meta-analysis of incidence and risk of hypokalemia with cetuximab-based therapy for advanced cancer. Cancer Chemother. Pharmacol. 2009, 66, 37–42.
  116. Giusti, R.M.; Cohen, M.H.; Keegan, P.; Pazdur, R. FDA Review of a Panitumumab (Vectibix™) Clinical Trial for First-Line Treatment of Metastatic Colorectal Cancer. Oncologist 2009, 14, 284–290.
  117. Boku, N.; Sugihara, K.; Kitagawa, Y.; Hatake, K.; Gemma, A.; Yamazaki, N.; Muro, K.; Hamaguchi, T.; Yoshino, T.; Yana, I.; et al. Panitumumab in Japanese patients with unresectable colorectal cancer: A post-marketing surveillance study of 3085 patients. Jpn. J. Clin. Oncol. 2014, 44, 214–223.
  118. Kamo, H.; Shinozaki, E.; Sugase, T.; Mizunuma, N.; Taniguchi, S.; Gotoh, T.; Chin, K.; Tanaka, T.; Koga, K.; Yamaguchi, K. Leukocytoclastic vasculitis with purpura and renal failure induced by the anti-epidermal growth factor receptor antibody panitumumab: A case report. J. Med. Case Rep. 2019, 13, 13.
  119. Manthri, S.; Bandaru, S.; Chang, A.; Hudali, T. Cetuximab-Associated Crescentic Diffuse Proliferative Glomerulonephritis. Case Rep. Nephrol. 2017, 2017, 1–4.
  120. Sasaki, K.; Anderson, E.; Shankland, S.J.; Nicosia, R.F. Diffuse Proliferative Glomerulonephritis Associated With Cetuximab, an Epidermal Growth Factor Receptor Inhibitor. Am. J. Kidney Dis. 2013, 61, 988–991.
  121. Koizumi, M.; Takahashi, M.; Murata, M.; Kikuchi, Y.; Seta, K.; Yahata, K. Thrombotic microangiopathy associated with cetuximab, an epidermal growth factor receptor inhibitor. Clin. Nephrol. 2017, 87, 51–54.
  122. Freeman, C.L.; Sehn, L.H. A tale of two antibodies: Obinutuzumabversusrituximab. Br. J. Haematol. 2018, 182, 29–45.
  123. Korycka-Wołowiec, A.; Wołowiec, D.; Robak, T. Ofatumumab for treating chronic lymphocytic leukemia: A safety profile. Expert Opin. Drug Saf. 2015, 14, 1945–1959.
  124. Wu, Y.; Wang, Y.; Gu, Y.; Xia, J.; Kong, X.; Qian, Q.; Hong, Y. Safety and efficacy of Ofatumumab in chronic lymphocytic leukemia: A systematic review and meta-analysis. Hematology 2017, 22, 578–584.
  125. Howard, S.C.; Trifilio, S.; Gregory, T.K.; Baxter, N.; McBride, A. Tumor lysis syndrome in the era of novel and targeted agents in patients with hematologic malignancies: A systematic review. Ann. Hematol. 2016, 95, 563–573.
  126. Kasi, P.M.; A Tawbi, H.; Oddis, C.V.; Kulkarni, H.S. Clinical review: Serious adverse events associated with the use of rituximab—A critical care perspective. Crit. Care 2012, 16, 231.
  127. Caldito, N.G.; Shirani, A.; Salter, A.; Stuve, O. Adverse event profile differences between rituximab and ocrelizumab: Findings from the FDA Adverse Event Reporting Database. Mult. Scler. J. 2020, 27, 1066–1076.
  128. Scott, L.J. Brentuximab Vedotin: A Review in CD30-Positive Hodgkin Lymphoma. Drugs 2017, 77, 435–445.
  129. Kuruvilla, J.; Ramchandren, R.; Santoro, A.; Paszkiewicz-Kozik, E.; Gasiorowski, R.; A Johnson, N.; Fogliatto, L.M.; Goncalves, I.; de Oliveira, J.S.R.; Buccheri, V.; et al. Pembrolizumab versus brentuximab vedotin in relapsed or refractory classical Hodgkin lymphoma (KEYNOTE-204): An interim analysis of a multicentre, randomised, open-label, phase 3 study. Lancet Oncol. 2021, 22, 512–524.
  130. Robak, T. Alemtuzumab for B-cell chronic lymphocytic leukemia. Expert Rev. Anticancer Ther. 2008, 8, 1033–1051.
  131. Hillmen, P.; Skotnicki, A.B.; Robak, T.; Jaksic, B.; Dmoszynska, A.; Wu, J.; Sirard, C.; Mayer, J. Alemtuzumab Compared With Chlorambucil As First-Line Therapy for Chronic Lymphocytic Leukemia. J. Clin. Oncol. 2007, 25, 5616–5623.
  132. Phelps, R.; A Winston, J.; Wynn, D.; Habek, M.; Hartung, H.-P.; Havrdová, E.K.; Markowitz, G.S.; Margolin, D.H.; E Rodriguez, C.; Baker, D.P.; et al. Incidence, management, and outcomes of autoimmune nephropathies following alemtuzumab treatment in patients with multiple sclerosis. Mult. Scler. J. 2019, 25, 1273–1288.
  133. Roux, C.; Thyss, A.; Gari-Toussaint, M. Prostatic and renal aspergillosis due to Aspergillus fumigatus in a patient receiving alemtuzumab for chronic lymphocytic leukaemia. J. Mycol. Médicale 2013, 23, 270–273.
  134. Perica, K.; Varela, J.C.; Oelke, M.; Schneck, J.P. Adoptive T Cell Immunotherapy for Cancer. Rambam Maimonides Med J. 2015, 6, e0004.
  135. Mohanty, R.; Chowdhury, C.R.; Arega, S.; Sen, P.; Ganguly, P.; Ganguly, N. CAR T cell therapy: A new era for cancer treatment (Review). Oncol. Rep. 2019, 42, 2183–2195.
  136. Wang, Z.; Wu, Z.; Liu, Y.; Han, W. New development in CAR-T cell therapy. J. Hematol. Oncol. 2017, 10, 1–11.
  137. Dai, H.; Wang, Y.; Lu, X.; Han, W. Chimeric Antigen Receptors Modified T-Cells for Cancer Therapy. JNCI J. Natl. Cancer Inst. 2016, 108, djv439.
  138. Eshhar, Z.; Waks, T.; Gross, G.; Schindler, D.G. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc. Natl. Acad. Sci. USA 1993, 90, 720–724.
  139. Wäsch, R.; Munder, M.; Marks, R. Teaming up for CAR-T cell therapy. Haematologica 2019, 104, 2335–2336.
  140. Sterner, R.C.; Sterner, R.M. CAR-T cell therapy: Current limitations and potential strategies. Blood Cancer J. 2021, 11, 1–11.
  141. Gross, G.; Waks, T.; Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl. Acad. Sci. USA 1989, 86, 10024–10028.
  142. Ma, S.; Li, X.; Wang, X.; Cheng, L.; Li, Z.; Zhang, C.; Ye, Z.; Qian, Q. Current Progress in CAR-T Cell Therapy for Solid Tumors. Int. J. Biol. Sci. 2019, 15, 2548–2560.
  143. Zhang, Q.; Ping, J.; Huang, Z.; Zhang, X.; Zhou, J.; Wang, G.; Liu, S.; Ma, J. CAR-T Cell Therapy in Cancer: Tribulations and Road Ahead. J. Immunol. Res. 2020, 2020, 1–11.
  144. Schepisi, G.; Cursano, M.C.; Casadei, C.; Menna, C.; Altavilla, A.; Lolli, C.; Cerchione, C.; Paganelli, G.; Santini, D.; Tonini, G.; et al. CAR-T cell therapy: A potential new strategy against prostate cancer. J. Immunother. Cancer 2019, 7, 258.
  145. Anwar, M.Y.; Williams, G.R.; Paluri, R.K. CAR T Cell Therapy in Pancreaticobiliary Cancers: A Focused Review of Clinical Data. J. Gastrointest. Cancer 2020, 52, 1–10.
  146. Brudno, J.N.; Kochenderfer, J.N. Recent advances in CAR T-cell toxicity: Mechanisms, manifestations and management. Blood Rev. 2019, 34, 45–55.
  147. Shimabukuro-Vornhagen, A.; Gödel, P.; Subklewe, M.; Stemmler, H.J.; Schlößer, H.A.; Schlaak, M.; Kochanek, M.; Böll, B.; Von Bergwelt-Baildon, M.S. Cytokine release syndrome. J. Immunother. Cancer 2018, 6, 56.
  148. Lee, D.W.; A Gardner, R.; Porter, D.L.; Louis, C.U.; Ahmed, N.; Jensen, M.C.; Grupp, S.A.; Mackall, C.L. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014, 124, 188–195.
  149. Norelli, M.; Camisa, B.; Barbiera, G.; Falcone, L.; Purevdorj, A.; Genua, M.; Sanvito, F.; Ponzoni, M.; Doglioni, C.; Cristofori, P.; et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat. Med. 2018, 24, 739–748.
  150. Chen, H.; Wang, F.; Zhang, P.; Zhang, Y.; Chen, Y.; Fan, X.; Cao, X.; Liu, J.; Yang, Y.; Wang, B.; et al. Management of cytokine release syndrome related to CAR-T cell therapy. Front. Med. 2019, 13, 610–617.
  151. Xu, X.-J.; Tang, Y.-M. Cytokine release syndrome in cancer immunotherapy with chimeric antigen receptor engineered T cells. Cancer Lett. 2014, 343, 172–178.
  152. Gauthier, J.; Turtle, C.J. Insights into cytokine release syndrome and neurotoxicity after CD19-specific CAR-T cell therapy. Curr. Res. Transl. Med. 2018, 66, 50–52.
  153. Liu, D.; Zhao, J. Cytokine release syndrome: Grading, modeling, and new therapy. J. Hematol. Oncol. 2018, 11, 1–7.
  154. Frey, N.V.; Porter, D.L. Cytokine release syndrome with novel therapeutics for acute lymphoblastic leukemia. Hematology 2016, 2016, 567–572.
  155. Jhaveri, K.D.; Rosner, M.H. Chimeric Antigen Receptor T Cell Therapy and the Kidney. Clin. J. Am. Soc. Nephrol. 2018, 13, 796–798.
  156. Zhou, H.; Yang, M.; Cui, L.; Jiang, J. Chimeric antigen receptor T cell therapy and nephrotoxicity: From diagnosis to treatment strategies. Int. Immunopharmacol. 2020, 89, 107072.
  157. Maude, S.L.; Barrett, D.; Teachey, D.; Grupp, S.A. Managing Cytokine Release Syndrome Associated with Novel T Cell-Engaging Therapies. Cancer J. 2014, 20, 119–122.
  158. Siddall, E.; Khatri, M.; Radhakrishnan, J. Capillary leak syndrome: Etiologies, pathophysiology, and management. Kidney Int. 2017, 92, 37–46.
  159. Shalabi, H.; Sachdev, V.; Kulshreshtha, A.; Cohen, J.W.; Yates, B.; Rosing, D.R.; Sidenko, S.; Delbrook, C.; Mackall, C.; Wiley, B.; et al. Impact of cytokine release syndrome on cardiac function following CD19 CAR-T cell therapy in children and young adults with hematological malignancies. J. Immunother. Cancer 2020, 8, e001159.
  160. Kellum, J.A. Persistent Acute Kidney Injury*. Crit. Care Med. 2015, 43, 1785–1786.
  161. Ramesh, G.; Reeves, W.B. Inflammatory cytokines in acute renal failure. Kidney Int. 2004, 66, S56–S61.
  162. Wu, W.; Wang, X.; Yu, X.; Lan, H.-Y. Smad3 Signatures in Renal Inflammation and Fibrosis. Int. J. Biol. Sci. 2022, 18, 2795–2806.
  163. Cairo, M.S.; Bishop, M. Tumour lysis syndrome: New therapeutic strategies and classification. Br. J. Haematol. 2004, 127, 3–11.
  164. Abu-Alfa, A.K.; Younes, A. Tumor Lysis Syndrome and Acute Kidney Injury: Evaluation, Prevention, and Management. Am. J. Kidney Dis. 2010, 55, S1–S13.
  165. Hines, M.R.; Keenan, C.; Alfaro, G.M.; Cheng, C.; Zhou, Y.; Sharma, A.; Hurley, C.; Nichols, K.E.; Gottschalk, S.; Triplett, B.M.; et al. Hemophagocytic lymphohistiocytosis-like toxicity (carHLH) after CD19-specific CAR T-cell therapy. Br. J. Haematol. 2021, 194, 701–707.
  166. Santoriello, D.; Hogan, J.; D’Agati, V.D. Hemophagocytic Syndrome With Histiocytic Glomerulopathy and Intraglomerular Hemophagocytosis. Am. J. Kidney Dis. 2016, 67, 978–983.
  167. Malaga-Dieguez, L.; Ming, W.; Trachtman, H. Direct Reversible Kidney Injury in Familial Hemophagocytic Lymphohistiocytosis Type 3. J. Am. Soc. Nephrol. 2015, 26, 1777–1780.
  168. Gupta, S.; Seethapathy, H.; Strohbehn, I.A.; Frigault, M.J.; O’Donnell, E.K.; Jacobson, C.A.; Motwani, S.S.; Parikh, S.M.; Curhan, G.C.; Reynolds, K.L.; et al. Acute Kidney Injury and Electrolyte Abnormalities After Chimeric Antigen Receptor T-Cell (CAR-T) Therapy for Diffuse Large B-Cell Lymphoma. Am. J. Kidney Dis. 2020, 76, 63–71.
  169. Gutgarts, V.; Jain, T.; Zheng, J.; Maloy, M.A.; Ruiz, J.D.; Pennisi, M.; Jaimes, E.A.; Perales, M.-A.; Sathick, J. Acute Kidney Injury after CAR-T Cell Therapy: Low Incidence and Rapid Recovery. Biol. Blood Marrow Transplant. 2020, 26, 1071–1076.
  170. Lee, M.D.; Strohbehn, I.A.; Seethapathy, H.S.; Rusibamayila, N.; Casey, K.S.; Gupta, S.; Leaf, D.E.; Frigault, M.J.; Sise, M.E. Acute Kidney Injury After the CAR-T Therapy Tisagenlecleucel. Am. J. Kidney Dis. 2021, 77, 990–992.
  171. Kanduri, S.R.; Cheungpasitporn, W.; Thongprayoon, C.; Petnak, T.; Lin, Y.; Kovvuru, K.; Manohar, S.; Kashani, K.; Herrmann, S.M. Systematic Review of Risk factors and Incidence of Acute Kidney Injury among Patients Treated with CAR-T Cell Therapies. Kidney Int. Rep. 2021, 6, 1416–1422.
  172. Joannidis, M.; Druml, W.; Forni, L.G.; Groeneveld, A.B.J.; Honore, P.M.; Hoste, E.; Ostermann, M.; Straaten, H.M.O.-V.; Schetz, M. Prevention of acute kidney injury and protection of renal function in the intensive care unit: Update 2017. Intensiv. Care Med. 2017, 43, 730–749.
  173. Burstein, D.S.; Maude, S.; Grupp, S.; Griffis, H.; Rossano, J.; Lin, K.; Burstein, D.S.; Maude, S.; Grupp, S.; Griffis, H.; et al. Cardiac Profile of Chimeric Antigen Receptor T Cell Therapy in Children: A Single-Institution Experience. Biol. Blood Marrow Transplant. 2018, 24, 1590–1595.
  174. Santomasso, B.D.; Nastoupil, L.J.; Adkins, S.; Lacchetti, C.; Schneider, B.J.; Anadkat, M.; Atkins, M.B.; Brassil, K.J.; Caterino, J.M.; Chau, I.; et al. Management of Immune-Related Adverse Events in Patients Treated With Chimeric Antigen Receptor T-Cell Therapy: ASCO Guideline. J. Clin. Oncol. 2021, 39, 3978–3992.
  175. Kotch, C.; Barrett, D.; Teachey, D.T. Tocilizumab for the treatment of chimeric antigen receptor T cell-induced cytokine release syndrome. Expert Rev. Clin. Immunol. 2019, 15, 813–822.
  176. Bergsten, E.; Horne, A.; Aricó, M.; Astigarraga, I.; Egeler, R.M.; Filipovich, A.H.; Ishii, E.; Janka, G.; Ladisch, S.; Lehmberg, K.; et al. Confirmed efficacy of etoposide and dexamethasone in HLH treatment: Long-term results of the cooperative HLH-2004 study. Blood 2017, 130, 2728–2738.
  177. Belay, Y.; Yirdaw, K.; Enawgaw, B. Tumor Lysis Syndrome in Patients with Hematological Malignancies. J. Oncol. 2017, 2017, 1–9.
More
ScholarVision Creations