Background: Tumor lysis syndrome (TLS) is an oncologic emergency due to a rapid break down of malignant cells usually induced by cytotoxic therapy, with hyperuricemia, hyperkalemia, hyperphosphatemia, hypocalcemia, and serious clinical consequences such as acute renal injury, cardiac arrhythmia, hypotension, and death. Rapidly expanding knowledge of cancer immune evasion mechanisms and host-tumor interactions has significantly changed our therapeutic strategies in hemato-oncology what resulted in the expanding spectrum of neoplasms with a risk of TLS. Summary: Since clinical TLS is a life-threatening condition, identifying patients with risk factors for TLS development and implementation of adequate preventive measures remains the most critical component of its medical management. In general, these consist of vigilant laboratory and clinical monitoring, vigorous IV hydration, urate-lowering therapy, avoidance of exogenous potassium, use of phosphate binders, and – in high-risk cases – considering cytoreduction before the start of the aggressive agent or a gradual escalation of its dose. Key Messages: In patients with a high risk of TLS, cytotoxic chemotherapy should be given in the facility with ready access to dialysis and a treatment plan discussed with the nephrology team. In the case of hyperkalemia, severe hyperphosphatemia or acidosis, and fluid overload unresponsive to diuretic therapy, the early renal replacement therapy (RRT) should be considered. One must remember that in TLS, the threshold for RRT initiation may be lower than in other clinical situations since the process of cell breakdown is ongoing, and rapid increases in serum electrolytes cannot be predicted.

Tumor lysis syndrome (TLS) is a hemato-oncologic emergency, characterized by hyperuricemia, hyperkalemia, hyperphosphatemia, hypocalcemia, and metabolic acidosis. It is due to a rapid break down of malignant cells, with a massive release of intracellular contents: potassium, phosphate, nucleic acids, and cytokines into the bloodstream. Catabolism of the nucleic acids leads to hyperuricemia, while hypocalcemia is a consequence of acute hyperphosphatemia with phosphate binding to calcium and calcium phosphate deposition in the body tissues. All this may be accompanied by a systemic inflammatory response triggered by cytokines released from tumor cells.

Usually, TLS is induced by cytotoxic therapy and appears in the first 48–72 h after its initiation, with first laboratory signs usually observed already 6–24 h after its initiation. However, it may be spontaneous, as in the case of rapidly proliferating high-grade hematologic malignancies, such as Burkitt’s lymphoma, acute myeloid leukemia (AML), and anaplastic large T-cell or diffuse large B-cell lymphoma [1]. TLS may have only laboratory form, or the metabolic disturbances may overwhelm the patient’s homeostatic capacity, leading to severe clinical consequences such as acute kidney injury (AKI), cardiac arrhythmia, hypotension, and/or neurologic complications, called then clinical TLS. The criteria for the diagnosis of both laboratory and clinical TLS are presented in Figure 1[2, 3].

Fig. 1.

Diagnosis of TLS in adults [2, 3]. TLS, tumor lysis syndrome; AKI, acute kidney injury; ULN, upper limit of normal.

Fig. 1.

Diagnosis of TLS in adults [2, 3]. TLS, tumor lysis syndrome; AKI, acute kidney injury; ULN, upper limit of normal.

Close modal

The incidence and prevalence of TLS are not well defined since they vary depending on several tumor-, anticancer therapy-, and patient-related risk factors, as well as prophylactic procedures undertaken. The most epidemiological data come mostly from the 90s, with laboratory TLS described in about 40% adults with hematologic malignancies [4] and even up to 70% of children with acute leukemia [5] and clinical form in <10% [6-8]. Then, in the first-year decade of the 21st century, we witnessed a rapid growth of highly effective novel anticancer therapies, and the risk of TLS – at least in certain diseases like chronic lymphocytic leukemia (CLL) – seemed to be even higher. This early experience resulted in the development of strict preventive measures and step-wise dosing and therapeutic sequencing strategies, which remarkably decreased the incidence of TLS [9]. It is now much less common, provided the adequate prophylaxis and monitoring; however, it must be kept in mind that the consequences may be fatal if they happen.

The condition is life threatening, and when not recognized early enough and aggressively treated, it is linked to a significantly increased risk for poor outcomes, with overall in-hospital mortality ranging in different series from 21 to 32%, with the highest reported rates of 79% in AML patients during induction therapy [6, 10, 11]. Among many predictors of short- and long-term mortality in patients with TLS, AKI appears to be the important one. In a single-center study in France, in-hospital and 6-month mortality rates were significantly higher in patients with TLS-related AKI (51 and 66%, respectively) than in patients with TLS but without AKI (7 and 21%) [11]. After adjustment for acute disease severity, the presence of AKI was associated with higher hospital mortality (OR: 10.41; 95% CI: 2.01–19.170; p = 0.005) and 6-month mortality (OR: 5.61; 95% CI: 1.64–54.66; p = 0.006), compared to patients without renal injury.

Long-term outcomes of TLS and TLS-related AKI are much more challenging to evaluate since there are too many confounding factors, with underlying malignancy being one of the most important. TLS-induced AKI may also decrease the probability of getting a long-term remission of the malignancy. No studies to date have evaluated the renal recovery from this specific type of AKI. It is known that AKI is a strong, independent risk factor for later CKD development and long-term mortality [12, 13]. Therefore, the role of early and adequate prophylaxis is crucial. Although there are no hard data, the studies performed in the last decade strongly suggest that the incorporation of strict preventive measures such as vigorous hydration, urate-lowering therapy, and close monitoring of the patient, together with more sophisticated cancer treatment strategies, may significantly reduce the risk, hasten recovery, and prevent complications.

The AKI is usually oliguric, and it is mostly a crystal-dependent injury due to precipitation of uric acid and calcium phosphate in renal tubules with obstruction of the tubular lumen. Calcium phosphate also precipitates in the interstitium and renal microvasculature, leading to nephrocalcinosis. Both types of crystals are toxic to the tubular epithelium, inducing local active inflammatory and pro-oxidative responses [14, 15]. Soluble uric acid may induce hemodynamic changes, with decreased renal blood flow due to vasoconstriction and impaired autoregulation (crystal-independent pathway) [15].

Additionally, uric acid may interfere with regenerative processes in proximal tubule cells, affecting their proliferation [16]. The aggravating factors include volume depletion, hypotension, nephrotoxins, radio-contrast exposure, sepsis, and pre-existing kidney dysfunction. In patients treated with high doses of allopurinol, the urinary concentration of xanthine may exceed its solubility leading to xanthine nephropathy or urolithiasis [17].

Since TLS is a life-threatening complication, prevention is the key component of its medical management. A simplified 3-step algorithm for the prevention of TLS is presented in Figure 2. The first step is to identify patients with risk factors for TLS development, the second is to implement adequate prophylactic measures that reduce the risk, and the third is a vigilant laboratory and clinical monitoring.

Fig. 2.

Key steps in TLS prevention. TLS, tumor lysis syndrome; CKD, chronic kidney disease; LDH, lactate dehydrogenase; RRT, renal replacement therapy.

Fig. 2.

Key steps in TLS prevention. TLS, tumor lysis syndrome; CKD, chronic kidney disease; LDH, lactate dehydrogenase; RRT, renal replacement therapy.

Close modal

Risk Stratification

Every patient who is going to receive chemotherapy for any hemato-oncological malignancy should be assessed for the risk of TLS. The risk depends on (1) the type of cancer, its mass, and cell-lysis potential; (2) the anticancer regimen, its effectiveness, and dosing; and (3) the clinical condition of the patient, particularly the presence of kidney dysfunction or involvement by the disease (Fig. 3).

Fig. 3.

TLS risk stratification for hemato-oncologic malignancies in adults [20, 80]. The dotted lines indicate the final TLS risk adjustment based on renal function and baseline uric acid, phosphate, and potassium serum concentrations. TLS, tumor lysis syndrome; CLL, chronic lymphocytic leukemia; WBC, white blood cells; AML, acute myeloid leukemia; LDH, lactate dehydrogenase; ULN, upper limit of normal.

Fig. 3.

TLS risk stratification for hemato-oncologic malignancies in adults [20, 80]. The dotted lines indicate the final TLS risk adjustment based on renal function and baseline uric acid, phosphate, and potassium serum concentrations. TLS, tumor lysis syndrome; CLL, chronic lymphocytic leukemia; WBC, white blood cells; AML, acute myeloid leukemia; LDH, lactate dehydrogenase; ULN, upper limit of normal.

Close modal

Cancer-Related Factors

In general, due to a rapid rate of cell turnover and sensitivity to chemotherapy, TLS has been observed more often in patients’ hematologic malignancies [18]. The highest risk of TLS has been described in patients with Burkitt lymphoma, other rapidly growing high-grade non-Hodgkin lymphomas, B-cell acute lymphoblastic leukemia, AML with high white blood counts (>50K), and – more recently – in CLL and when treated with newer anticancer therapies. In patients with high-grade non-Hodgkin lymphomas, the predisposing cancer-related factors that must be assessed include the disease bulk, advanced stage, cancer and its proliferation potential (indicated by serum LDH ≥2 × upper limit of normal – ULN), and treatment sensitivity [19]. In acute leukemias, the most important seem to be the bone marrow involvement and high white blood counts (≥100 × 109/L or <100 × 109/L + LDH ≥2 × ULN), while in CLL, treatment with venetoclax and lymph node ≥10 or ≥5 cm and absolute lymphocyte count ≥25 × 109/L and elevated uric acid and spleno- and hepatomegaly [19]. Therefore, before TLS risk stratification, every patient with CLL or non-Hodgkin lymphoma should have a staging computed tomography of the chest, abdomen, and pelvis.

The intermediate-risk category for TLS includes the same (1) non-Hodgkin lymphomas, but in early stages, not bulky or with low proliferation potential (LDH <2 × ULN), (2) acute leukemias with lower WBC counts, and (3) CLL treated with fludarabine, rituximab or lenalidomide, or treated with venetoclax and with lymph node ≥5 cm, or with absolute lymphocyte count ≥25 × 109/L, or CLL with WBC ≥50 × 109/L. Additionally, there have been some classified highly chemotherapy-sensitive neoplasms such as neuroblastoma, germ cell tumors, and small-cell lung cancer, when bulky and in advanced stages [20].

All these factors must be interpreted together (Fig. 3). Keeping in mind the crucial role of preserved renal function, it is logical that renal dysfunction/involvement automatically upgrades the risk category.

Therapy-Related Factors

During the last twenty years, particularly the last decade, the expanding knowledge of cancer immune evasion mechanisms and host-tumor interactions has significantly changed our therapeutic strategies in hemato-oncology [21-25]. Several novel targeted molecular and immune cell-based agents are now available, usually to be used in combination with conventional cytotoxic agents [26]. Thereby, more neoplasms with a low proliferation rate, often refractory to the traditional cytotoxic chemotherapies, became responsive to much more effective new anticancer therapies. This concerns some solid tumors (pulmonary, gynecologic, gastrointestinal, neurologic, and sarcomas, especially advanced and metastatic); however, the most dramatic changes have been observed in CLL and small lymphocytic lymphoma (SLL) [3, 18, 27]. The introduction of small molecule inhibitors has dramatically transformed this otherwise rather indolent hematological malignancy into a disease with a clinically significant risk for TLS, concerning 10–18% of patients in early phase studies [19].

In general, the treatment of CLL/SLL targets 3 major cell pathways involved in the pathogenesis of B-cell proliferation: Bruton tyrosine kinase (BTK), phosphoinositide-3-kinase (PI3K), and the B-cell lymphoma-2 receptor (BCL-2). According to the current guidelines, targeted therapy with BTK inhibitors (ibrutinib) and BCL-2 inhibitor, venetoclax, is the preferred first-line treatment of all patients with CLL [28]. Fludarabine, cyclophosphamide, and rituximab are preferred for patients <65 years with untreated IGHV-mutated CLL. BTK inhibitors (ibrutinib and acalabrutinib), PI3K inhibitors (idelalisib, alt. with rituximab, and duvelisib), and venetoclax ± rituximab remain effective treatment options for relapsed/refractory CLL/SLL.

BCL-2 Inhibitors

BCL-2-type proteins are key molecules in cell regulatory pathways for apoptosis. The high efficacy of the first BCL-2 inhibitor, venetoclax, used in CLL, seems to be due to the pathophysiology of the disease, which is characterized by an overexpression of BCL-2 receptors [29]. In early clinical studies, the treatment with venetoclax was associated with the significant risk of TLS (8.3 and 8.9% of studied patients), with 2 TLS-associated fatalities, which led to a suspension of the trials [30, 31]. This resulted in the development of effective TLS mitigation strategies with step-wise dosing (dose ramp-up) of the drug, lowering WBC below 25 × 109/L with hydroxyurea before starting treatment with venetoclax or chemoimmunotherapy (BTK inhibitor and CD20 monoclonal antibody) [19, 32]. The debulking strategies, together with adequate hydration and more careful biochemical monitoring, resulted in a considerable decrease in TLS risk to approximately 1–3.8% and clinical TLS to <1% [19, 29, 33-36]. In other hematological diseases (multiple myeloma and AML), TLS is uncommon [37-43].

BTK Inhibitors

BTK inhibitors are the other agents currently used in CLL, targeting B-cell receptor signaling pathways. They may be given as a monotherapy or concurrently with venetoclax since there is a marked synergy between these drugs and their complementary activity. Since BTK inhibitors are highly active in treating and shrinking nodal disease and BCL-2 inhibitors are highly effective at clearing blood and bone marrow, their combination and therapeutic sequencing may permit treatment of shorter duration and lower intensity than chemotherapy, while still preserving the disease-control benefits [28, 29, 44]. The results of the phase II CAPTIVATE study, in which the BTK inhibitor, ibrutinib, was given with venetoclax as frontline therapy, are encouraging for both efficacy and safety [44]. There were 3 cases of laboratory TLS and no clinical manifestation. Moreover, the ibrutinib lead-in period resulted in a considerable tumor bulk reduction, with downgrading of the TLS risk category in 80% of the high-risk patients and 48% of the medium-risk patients [36]. Similarly, promising results were achieved in the CLARITY study, in which the ibrutinib-venetoclax combination was given to patients with relapsed/refractory CLL, with TLS 2% (one of 50 patients) [35].

PI3K Inhibitors

Similar to BTK inhibitors, PI3K inhibitors block B-cell receptor signaling, interfering with several pathways required for leukemia cell survival. Two of them, idelalisib and duvelisib, have been approved by the FDA to treat relapsed/refractory CLL and SLL based on the results of the phase III randomized DUO study and the DYNAMO study [45-47]. There were single 3 case reports of TLS in patients treated with idelalisib, but no laboratory or clinical TLS was observed in major clinical trials [46-48].

CDK Inhibitors

The regulatory role of cyclin-dependent kinases (CDKs) in the transition of cell steps that are necessary for its proliferation makes these enzymes a natural target for cancer therapy. The first CDK inhibitors to enter clinical trials were alvocidib and dinaciclib, given in high-risk CCL, acute leukemias, multiple myeloma, lymphomas, and some tumors. Both drugs exerted nonselective, inhibitory effects on a wide range of CKDs, including CKD9, which is implicated in CLL. In phase I and II trials, the major issue of these drugs was their toxicity profile to healthy cells, which led to severe side effects with a strikingly high incidence of TLS, reaching 40–50% patients for laboratory TLS and 15% for clinical TLS, with many of them requiring immediate hemodialysis [49-53]. Although a combination with other anticancer drugs was shown to mitigate their toxicity [54], both drugs were abandoned and replaced by selective CDK4/6 inhibitors palbociclib, ribociclib, and abemaciclib, with more favorable safety profile with no TLS syndrome reported. These drugs are FDA approved as a frontline treatment of breast cancer [55].

Proteasome Inhibitors

Proteasome inhibitors (bortezomib, oprozomib, carfilzomib, and ixazomib) block the function of the proteasome, the garbage disposal system of the cell, from the degradation of excess proteins, which in consequence build up in a cell leading to its death. They are an important class of drugs for the treatment of multiple myeloma and certain types of lymphoma, which generate a lot of additional proteins. Generally, the risk of TLS in multiple myeloma is low, given the disease’s low proliferation rate. However, after the introduction of proteasome inhibitors, an increasing frequency of TLS has been reported, with an incidence of 1.4–5% for bortezomib, 0.4–4.3% for carfilzomib, and 2.4% for oprozomib [9, 56-58].

Monoclonal Antibodies

The rate of TLS after treatment with first-generation anti-CD20 monoclonal antibody rituximab is low, in case reports or case series [56, 59]. Among the next-generations of these drugs, obinutuzumab, used in combination with chlorambucil in patients with relapsed/refractory CLL, was associated with 4.8% incidence of TLS in phase 1 and 2 studies [60] and with 4.3% in a recent phase 3 ILLUMINATE study [61].

Proapoptotic Agents

The rate of TLS with a proapoptotic agent, lenalidomide, seems to be low (0–4%), particularly when used in patients with relapsed/refractory CLL (<5%) [3, 17, 62, 63].

CAR T-Cell Therapy

The therapy uses the patient’s cells, which are ex vivo genetically engineered to produce specific chimeric antigen receptors (CARs) on their surface, to direct them against the leukemic cells. Then, these modified cells are multiplied and infused back to the patient as therapy. TLS has been described in patients with acute and chronic hematologic malignancies after CAR T-cell therapy; however, the true incidence may be not yet known being overshadowed by coincident cytokine release syndrome [64-66]. In 2 large clinical studies conducted in adult patients with refractory large B-cell lymphoma, one with tisagenlecleucel and a second with axicabtagene ciloleucel CAR T-cell therapy, no cases of TLS were reported [67, 68]. However, in the recent phase 2, 25-center study, a global study of tisagenlecleucel in children and young adults with relapsed or refractory B-cell ALL, TLS occurred in 3 of 75 patients (4%) [65]. Therefore, regular – daily if possible – inpatient monitoring and TLS prophylaxis are recommended for patients with a high disease burden or elevated serum uric acid [69].

Immune Checkpoint Inhibitors

Immune checkpoint inhibitors are monoclonal antibodies that enhance tumor killing by T cells by releasing a natural brake (checkpoint proteins) on patient’s immune system so that the patient’s T cells could recognize and kill tumor cells. In patients treated with immune checkpoint inhibitors, TLS seems to be a rare complication, but it can be life threatening. Cordrey and Wang [70], in their systemic review of the literature, identified only 5 publications (4 case reports and 1 phase I clinical trial report) with a total of 6 cases of TLS after treatment with the immune checkpoint inhibitors. The median time from treatment to TLS was 14 days, with a range of 2 to 33 days. Four of these patients died; all of them had extensive liver metastases. The authors postulate the lack of awareness of TLS risk, together with its delayed occurrence in solid tumors, may contribute to the higher mortality as well as to the underdiagnosing and underreporting.

Prophylactic Measures

The prophylactic measures are vigorous IV hydration, urate-lowering therapy, avoidance of exogenous potassium and phosphate, and – in high-risk cases – considering cytoreduction before the start of the aggressive agent or a gradual escalation of its dose. Prophylactic use of phosphate binders or potassium binding resins is not recommended while avoiding additive potassium in intravenous fluids seems reasonable.

Intravenous Fluid Expansion

Patients should be instructed to drink 1.5–2.0 L of water daily starting 2 days before and throughout the dose-titration phase. Intravenous fluids should be administered as indicated based on overall risk of TLS or for those who cannot maintain an adequate level of oral hydration. It should be started at least 24 h before the anticancer drug dosing, provided that the patient is well hydrated, and continued for 24–48 h after completion of the therapy. The infusion rates should be high enough to keep urinary output >100 mL/h, with daily urine volumes of at least 3 L. In patients with evidence of fluid overload, or with insufficient diuresis despite well hydration, the loop diuretics may be considered. Thiazide diuretics are contradicted since they increase uric acid levels and interact with allopurinol. In the era of rasburicase, urinary alkalinization is no longer recommended since it may cause heavy calcium phosphate precipitation [17, 71].

Urate-Lowering Therapy

In the aforementioned guidelines, 2 uric acid-decreasing agents, the conventional xanthine oxidase inhibitor allopurinol and the recombinant uricase rasburicase, are recommended for the prophylaxis and management of TLS. Allopurinol is used to treat patients at low and intermediate risk of TLS, and rasburicase for patients with high risk, renal failure, and those with already existing TLS [17, 72].

Allopurinol

Allopurinol, a xanthine oxidase inhibitor, blocks the conversion of nucleic acids released from cancer cells to hypoxanthine to xanthine and xanthine to uric acid (Fig. 3), which are much more easily cleared by the kidney. Since it does not remove the existing uric acid, it usually takes a few days to reduce its concentration. Therefore, it is recommended that the treatment should be started 2–3 days before chemotherapy and continued at least for 10–14 days or until the signs of massive tumor lysis are absent [27, 73]. Typically, the drug is given orally at a dose of 600–800 mg daily; if necessary, it can also be given intravenously. Allopurinol is excreted by the kidney, which imposes the dose reduction in renal dysfunction and significantly limits the number of patients who may benefit from the treatment. The dose should also be reduced or the drug avoided in patients concomitantly treated with azathioprine, cyclophosphamide, or 6-mercaptopurine since it can potentiate their cytotoxic effects. The adverse effects are usually mild and include pruritic rash, diarrhea, leukopenia, and thrombocytopenia, occurring in 3–5% of the patients; however, more severe hypersensitivity skin reactions, acute interstitial nephritis, and xanthine nephropathy have been described.

Febuxostat

Recently, a novel potent nonpurine xanthine oxidase inhibitor, febuxostat, the medication approved for gout treatment, is being tested in preventing TLS. Since the drug is metabolized via glucuronidation and oxidation, with only 1–6% of the dose being excreted unchanged via the kidneys, no dose adjustment is necessary for patients with mild or moderate renal impairment [74]. The first studies demonstrated its efficacy in uric acid reduction and suggested that febuxostat may serve as an alternative to allopurinol in patients with renal dysfunction, allopurinol intolerance, or allopurinol resistance [75, 76]. The drug is usually started 24 h before chemotherapy and discontinued after the risk of TLS is minimal or absent. In a recent meta-analysis which included 6 studies with a total of 659 patients, febuxostat achieved a similar response rate, TLS incidence, and the rate of adverse events when compared to allopurinol [77]. However, the drug is not free from side effects, the most serious being Stevens-Johnson syndrome, anaphylaxis, and, as suggested by a recent safety trial, an increased risk of cardiac and all-cause mortality [78]. On this basis, in 2019, the FDA released a boxed warning limiting febuxostat use to the patients with hyperuricemia who cannot tolerate allopurinol in a setting in which rasburicase is not available or is contraindicated [79, 80].

Rasburicase

Rasburicase is a recombinant urate oxidase produced in genetically modified Saccharomyces cerevisiae. It decreases serum uric acid concentrations by converting it into an inactive metabolite, allantoin, easily soluble in water and excreted in the urine. Unlike allopurinol, its action is immediate, with a rapid decrease in serum acid concentration. In contrast to allopurinol, there is no need for rasburicase dose adjustment in patients with renal dysfunction. It is noteworthy that since rasburicase degrades uric acid in blood and plasma at room temperature, to avoid false results, the blood samples must be collected in pre-chilled tubes and immediately sent to the laboratory on ice, with the assay performed in 4 h of collection.

In several clinical trials, rasburicase has been shown to be effective and safe in both adults and children. The drug should be administered 4–24 h before starting chemotherapy. The labeled dose is 0.2 mg/kg, daily given as a 30-min IV infusion, for up to 5 days [72]; however, in patients with low-intermediate risk of TLS, the smaller doses (0.1–0.15 mg/kg) were demonstrated to be also efficient, allowing for substantial cost reduction [81]. Alternatively, for patients with the low to moderate risk, a single fixed (3, 4.5, 6, or 7.5 mg) or weight-based (0.15–0.20 mg/kg) dose regimen has been proposed, and its efficacy is demonstrated [82-88]. A systematic review and meta-analysis of 19 studies by Yu et al. [89] revealed that single doses of rasburicase: 6 mg for adults and 1.5 and 0.15 mg/kg for children, were sufficient to normalize and sustain lower uric acid and creatinine levels in adults with TLS. According to these authors, the 3- and 4.5-mg single doses can be considered if the baseline uric acid level is <12 mg/dL. In patients treated with the single-dose regimen, a reassessment of clinical and biochemical parameters is necessary, with repeating the dose if required [3, 89]. Cortes et al. [90] demonstrated that sequential therapy with rasburicase followed by allopurinol was similarly effective and may be a reasonable and cost-effective approach with similar efficacy in adults with hyperuricemia or at high risk for TLS.

One of the important factors limiting use of rasburicase is the considerable cost of the drug. However, in a retrospective study, Cairo et al. [91] compared reductions in uric acid, length of ICU, hospital stay, and hospitalization costs in 26 rasburicase-treated and 104 allopurinol-treated patients with TLS (matched in the ratio 1:4). Length of ICU and hospital stay was 2.5 and 5 days less for the rasburicase group (p < 0.0001 and p = 0.02, respectively). Total treatment costs per patient (Δ20,038$; p < 0.02) as well as cost per percentage of uric acid reduction ($3,899 vs. $16,894; p < 0.001) occurred to be significantly lower for patients treated with rasburicase than for those treated with allopurinol.

Although generally very well tolerated, rasburicase may cause serious and fatal adverse events, such as hypersensitivity reactions, including anaphylaxis, as well as severe oxidative hemolytic anemia and methemoglobinemia in patients with glucose-6-phosphate dehydrogenase deficiency [92-94]. For that reason, it carries the FDA boxed warning to alert the physician to these risks and take all appropriate precautions when introducing the drug [92]. Rasburicase is contradicted in patients with glucose-6-phosphate dehydrogenase deficiency. Therefore, the patients at risk (males of African, Asian, and Mediterranean descent) should be screened for this genetic abnormality before its administration [93]. The repeated exposure to rasburicase should be avoided since anaphylaxis after the second course of rasburicase appears to occur more frequently [94]. In any patient developing clinical symptoms of hypersensitivity, the drug should be immediately and permanently discontinued [92].

Taking into account the aforementioned risks, there is a need for clear recommendations for its use in the setting of TLS. According to the US Prescribing Information (USPI), rasburicase is recommended in patients with high risk of TLS, particularly those with elevated baseline uric acid or in already existing syndrome (Table 1) [95]. It seems also reasonable to use it in patients with high or even intermediate TLS risk and renal or heart failure who cannot tolerate aggressive hydration.

Table 1.

Recommended TLS prophylaxis based on tumor burden in patients with CLL and SLL [95]

Recommended TLS prophylaxis based on tumor burden in patients with CLL and SLL [95]
Recommended TLS prophylaxis based on tumor burden in patients with CLL and SLL [95]

Monitoring Approach

In all patients who are beginning anticancer therapy, several laboratory and clinical parameters should be carefully monitored. These include serum potassium, uric acid, phosphate, calcium, creatinine concentration, and LDH activity, as well as diuresis and fluid balance, which should be assessed on an ongoing basis. The frequency of laboratory parameter monitoring depends on the risk of TLS. While in low-risk patients, it can be done once daily, those at intermediate risk should undergo laboratory monitoring every 8–12 h, and those at high risk, every 4–6 h [3]. It is recommended to start the monitoring before initiation of chemotherapy and continue as long as the patient is at risk for TLS, which depends on the therapeutic regimen [3].

Hyperphosphatemia

In some patients, other factors besides hyperuricemia may play a key role in the development of TLS and AKI, and the urate-lowering therapy may not be sufficient as a preventive measure. The second most common metabolic disorder responsible for AKI, which cannot be underestimated, is hyperphosphatemia. Malignant cells contain up to 4 times more phosphate than normal cells and this increases further in hyperproliferative states such as blast crisis [96].

Phosphate-induced nephropathy may be aggravated when urinary alkalinization is used, as high urine pH favors precipitation of calcium phosphate in the renal tubules. In a multicenter prospective French study by Darmon et al. [97], which enrolled 153 high-risk patients, TLS developed in 30.7% of cases (in 11.1% laboratory and 19.6% clinical from), despite urate-lowering treatment. In a logistic regression model, the serum phosphate concentration occurred to be the main risk factor associated with clinical TLS and AKI, with OR 5.3; 95% CI: 1.5–18.3 (per mmol/L). The authors stressed the role of acute phosphate nephropathy in AKI in a setting of TLS, which in some cases may be overlooked, especially in the light of current guidelines focused on the antiuricemic agents [97].

Therefore, the treatment of hyperphosphatemia cannot be underestimated and should be started immediately, with phosphate intake restriction and elimination from IV solutions, avoidance of bicarbonates, and use of oral noncalcium phosphate binders. For patients with severe acute serum phosphate increase, the prophylactic intensive care unit admission and the consideration of early institution of renal replacement therapy (RRT) seem reasonable, to prevent disseminated metastatic calcium deposition.

Hyperkalemia

Hyperkalemia is the most life-threatening abnormality in TLS; therefore, it must be aggressively treated. It may be aggravated by metabolic acidosis. In every patient with hyperkalemia, continuous cardiac rhythm monitoring, as well as an immediate nephrology consultation, is recommended, and urgent hemodialysis should be considered. In case of emergency (serum potassium >6.5 mmol/L, cardiac conduction abnormalities, arrhythmia, lengthening of the PR interval and widening of QRS, muscle weakness, or paralysis), while waiting for hemodialysis, the rapidly acting therapies should be administered. These consist of IV infusion of 10% dextrose with rapid-acting insulin, to drive potassium into the cells, and IV calcium chlorate or gluconate to antagonize the membrane actions of hyperkalemia. If there is a risk of a longer delay before the dialysis is started, we administer oral gastrointestinal sodium-potassium exchange resins.

Hypocalcemia

Asymptomatic hypocalcemia often resolves as serum phosphate concentration is corrected, and the treatment, which may aggravate metastatic calcifications, is not recommended. Symptomatic patients should be treated with calcium at the lowest doses required to relieve symptoms [17, 80, 81].

Renal Replacement Therapy

In patients with a high risk of TLS, cytotoxic chemotherapy should be given in the facility with ready access to RRT and a treatment plan discussed with the nephrology team [81]. It must be remembered that in TLS, the threshold for RRT initiation may be lower than in other clinical situations, for 3 reasons. First is that the process of the cell break down is still ongoing and one cannot predict rapid increases in serum electrolytes in the individual patients, particularly in those with kidney dysfunction and oliguria. Secondly, early institution of RRT interrupts the pathological cascade with avoidance of life-threatening complications. Last but not least, it may prevent irreversible kidney injury. Early intervention is particularly favored in patients with congestive heart failure who cannot tolerate large fluid volumes. TLS in end-stage renal disease patients on dialysis seems to be extremely rare and we have found only 1 case report in the literature [98]. Therefore, no guidelines for optimal therapy in this population are available.

The choice of the RRT technique depends on the laboratory data and the malignant cell turnover rate as well as the clinical condition of the patient, volume status, hemodynamics, degree of impairment of other organs, and nutritional support. Intermittent hemodialysis is preferred in patients with severe hyperkalemia and severe hyperuricemia. In critically ill, continuous veno-venous hemofiltration, continuous veno-venous hemodialysis, or sustained low-efficiency daily dialysis seem to be a reasonable choice since they offer greater hemodynamic stability and better volume control, without the rebound hyperphosphatemia and hyperkalemia [99, 100]. For patients with severe hyperphosphatemia, continuous veno-venous hemodiafiltration has been proved as the most effective method of phosphate control [100, 101]. Peritoneal dialysis is not recommended for the treatment of TLS.

The authors have no conflicts of interest to declare.

The authors did not receive any funding.

J.M.R. and J.M.: design of the study, manuscript preparation, and final approval; J.M.R.: preparation of figures.

1.
Gangireddy
M
,
Shrimanker
I
,
Nookala
VK
,
Peroutka
KA
.
Spontaneous tumor lysis syndrome in diffuse large B-cell lymphoma: early diagnosis and management
.
Cureus
.
2019
May
;
11
(
5
):
e4679
.
[PubMed]
2168-8184
2.
Cairo
MS
,
Bishop
M
.
Tumour lysis syndrome: new therapeutic strategies and classification
.
Br J Haematol
.
2004
Oct
;
127
(
1
):
3
11
.
[PubMed]
0007-1048
3.
Howard
SC
,
Jones
DP
,
Pui
CH
.
The tumor lysis syndrome
.
N Engl J Med
.
2011
May
;
364
(
19
):
1844
54
.
[PubMed]
0028-4793
4.
Jeha
S
.
Tumor lysis syndrome
.
Semin Hematol
.
2001
Oct
;
38
(
4
Suppl 10
):
4
8
.
[PubMed]
0037-1963
5.
Wolf
G
,
Hegewisch-Becker
S
,
Hossfeld
DK
,
Stahl
RA
.
Hyperuricemia and renal insufficiency associated with malignant disease: urate oxidase as an efficient therapy?
Am J Kidney Dis
.
1999
Nov
;
34
(
5
):
E20
.
[PubMed]
0272-6386
6.
Montesinos
P
,
Lorenzo
I
,
Martín
G
,
Sanz
J
,
Pérez-Sirvent
ML
,
Martínez
D
, et al.
Tumor lysis syndrome in patients with acute myeloid leukemia: identification of risk factors and development of a predictive model
.
Haematologica
.
2008
Jan
;
93
(
1
):
67
74
.
[PubMed]
0390-6078
7.
Hande
KR
,
Garrow
GC
.
Acute tumor lysis syndrome in patients with high-grade non-Hodgkin’s lymphoma
.
Am J Med
.
1993
Feb
;
94
(
2
):
133
9
.
[PubMed]
0002-9343
8.
Annemans
L
,
Moeremans
K
,
Lamotte
M
,
Garcia Conde
J
,
van den Berg
H
,
Myint
H
, et al.
Incidence, medical resource utilisation and costs of hyperuricemia and tumour lysis syndrome in patients with acute leukaemia and non-Hodgkin’s lymphoma in four European countries
.
Leuk Lymphoma
.
2003
Jan
;
44
(
1
):
77
83
.
[PubMed]
1042-8194
9.
Howard
SC
,
Trifilio
S
,
Gregory
TK
,
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
Mar
;
95
(
4
):
563
73
.
[PubMed]
0939-5555
10.
Durani
U
,
Shah
ND
,
Go
RS
.
In-hospital outcomes of tumor lysis syndrome: A population-based study using the national inpatient sample
.
Oncologist
.
2017
Dec
;
22
(
12
):
1506
9
.
[PubMed]
1083-7159
11.
Darmon
M
,
Guichard
I
,
Vincent
F
,
Schlemmer
B
,
Azoulay
E
.
Prognostic significance of acute renal injury in acute tumor lysis syndrome
.
Leuk Lymphoma
.
2010
Feb
;
51
(
2
):
221
7
.
[PubMed]
1042-8194
12.
Coca
SG
,
Singanamala
S
,
Parikh
CR
.
Chronic kidney disease after acute kidney injury: a systematic review and meta-analysis
.
Kidney Int
.
2012
Mar
;
81
(
5
):
442
8
.
[PubMed]
0085-2538
13.
Kim
CS
,
Bae
EH
,
Ma
SK
,
Kweon
SS
,
Kim
SW
.
Impact of transient and persistent acute kidney injury on chronic kidney disease progression and mortality after gastric surgery for gastric cancer
.
PLoS One
.
2016
Dec
;
11
(
12
):
e0168119
.
[PubMed]
1932-6203
14.
Ejaz
AA
,
Mu
W
,
Kang
DH
,
Roncal
C
,
Sautin
YY
,
Henderson
G
, et al.
Could uric acid have a role in acute renal failure?
Clin J Am Soc Nephrol
.
2007
Jan
;
2
(
1
):
16
21
.
[PubMed]
1555-9041
15.
Shimada
M
,
Johnson
RJ
,
May
WS
 Jr
,
Lingegowda
V
,
Sood
P
,
Nakagawa
T
, et al.
A novel role for uric acid in acute kidney injury associated with tumour lysis syndrome
.
Nephrol Dial Transplant
.
2009
Oct
;
24
(
10
):
2960
4
.
[PubMed]
0931-0509
16.
Han
HJ
,
Lim
MJ
,
Lee
YJ
,
Lee
JH
,
Yang
IS
,
Taub
M
.
Uric acid inhibits renal proximal tubule cell proliferation via at least two signaling pathways involving PKC, MAPK, cPLA2, and NF-kappaB
.
Am J Physiol Renal Physiol
.
2007
Jan
;
292
(
1
):
F373
81
.
[PubMed]
1931-857X
17.
LaRosa
C
,
McMullen
L
,
Bakdash
S
,
Ellis
D
,
Krishnamurti
L
,
Wu
HY
, et al.
Acute renal failure from xanthine nephropathy during management of acute leukemia
.
Pediatr Nephrol
.
2007
Jan
;
22
(
1
):
132
5
.
[PubMed]
0931-041X
18.
Alakel
N
,
Middeke
JM
,
Schetelig
J
,
Bornhäuser
M
.
Prevention and treatment of tumor lysis syndrome, and the efficacy and role of rasburicase
.
OncoTargets Ther
.
2017
Feb
;
10
:
597
605
.
[PubMed]
1178-6930
19.
Tambaro
FP
,
Wierda
WG
.
Tumour lysis syndrome in patients with chronic lymphocytic leukaemia treated with BCL-2 inhibitors: risk factors, prophylaxis, and treatment recommendations
.
Lancet Haematol
.
2020
Feb
;
7
(
2
):
e168
76
.
[PubMed]
2352-3026
20.
Cairo
MS
,
Coiffier
B
,
Reiter
A
,
Younes
A
;
TLS Expert Panel
.
Recommendations for the evaluation of risk and prophylaxis of tumour lysis syndrome (TLS) in adults and children with malignant diseases: an expert TLS panel consensus
.
Br J Haematol
.
2010
May
;
149
(
4
):
578
86
.
[PubMed]
0007-1048
21.
Rosenberg
SA
,
Restifo
NP
.
Adoptive cell transfer as personalized immunotherapy for human cancer
.
Science
.
2015
Apr
;
348
(
6230
):
62
8
.
[PubMed]
0036-8075
22.
Park
JH
,
Rivière
I
,
Gonen
M
,
Wang
X
,
Sénéchal
B
,
Curran
KJ
, et al.
Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia
.
N Engl J Med
.
2018
Feb
;
378
(
5
):
449
59
.
[PubMed]
0028-4793
23.
Kochenderfer
JN
,
Somerville
RP
,
Lu
T
,
Yang
JC
,
Sherry
RM
,
Feldman
SA
, et al.
Long-duration complete remissions of diffuse large B cell lymphoma after anti-CD19 chimeric antigen receptor T cell therapy
.
Mol Ther
.
2017
Oct
;
25
(
10
):
2245
53
.
[PubMed]
1525-0016
24.
Podar
K
,
Jager
D
.
Targeting the immune niche within the bone marrow microenvironment: the rise of immunotherapy in multiple myeloma
.
Curr Cancer Drug Targets
.
2017
;
17
(
9
):
782
805
.
[PubMed]
1568-0096
25.
Comoli
P
,
Chabannon
C
,
Koehl
U
,
Lanza
F
,
Urbano-Ispizua
A
,
Hudecek
M
, et al.;
European Society for Blood and Marrow Transplantation, Cellular Therapy & Immunobiology Working Party – Solid Tumor Sub-committee
.
Development of adaptive immune effector therapies in solid tumors
.
Ann Oncol
.
2019
Nov
;
30
(
11
):
1740
50
.
[PubMed]
0923-7534
26.
Apetoh
L
,
Ladoire
S
,
Coukos
G
,
Ghiringhelli
F
.
Combining immunotherapy and anticancer agents: the right path to achieve cancer cure?
Ann Oncol
.
2015
Sep
;
26
(
9
):
1813
23
.
[PubMed]
0923-7534
27.
McBride
A
,
Trifilio
S
,
Baxter
N
,
Gregory
TK
,
Howard
SC
.
Managing tumor lysis syndrome in the era of novel cancer therapies
.
J Adv Pract Oncol
.
2017
Nov-Dec
;
8
(
7
):
705
20
.
[PubMed]
2150-0878
28.
Wierda
WG
,
Byrd
JC
,
Abramson
JS
,
Bilgrami
SF
,
Bociek
G
,
Brander
D
, et al.
Chronic lymphocytic leukemia/small lymphocytic lymphoma, Version 4.2020
.
J Natl Compr Canc Netw
.
2020
Feb
;
18
(
2
):
185
217
.
[PubMed]
1540-1413
29.
Seymour
J
.
Venetoclax, the first BCL-2 inhibitor for use in patients with chronic lymphocytic leukemia
.
Clin Adv Hematol Oncol
.
2019
Aug
;
17
(
8
):
440
3
.
[PubMed]
1543-0790
30.
Roberts
AW
,
Davids
MS
,
Pagel
JM
,
Kahl
BS
,
Puvvada
SD
,
Gerecitano
JF
, et al.
Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia
.
N Engl J Med
.
2016
Jan
;
374
(
4
):
311
22
.
[PubMed]
0028-4793
31.
Seymour
JF
,
Ma
S
,
Brander
DM
,
Choi
MY
,
Barrientos
J
,
Davids
MS
, et al.
Venetoclax plus rituximab in relapsed or refractory chronic lymphocytic leukaemia: a phase 1b study
.
Lancet Oncol
.
2017
Feb
;
18
(
2
):
230
40
.
[PubMed]
1470-2045
32.
Richard-Carpentier
G
,
DiNardo
CD
.
Venetoclax for the treatment of newly diagnosed acute myeloid leukemia in patients who are ineligible for intensive chemotherapy
.
Ther Adv Hematol
.
2019
Oct
;
10
:
2040620719882822
.
[PubMed]
2040-6207
33.
Seymour
JF
,
Kipps
TJ
,
Eichhorst
B
,
Hillmen
P
,
D’Rozario
J
,
Assouline
S
, et al.
Venetoclax-rituximab in relapsed or refractory chronic lymphocytic leukemia
.
N Engl J Med
.
2018
Mar
;
378
(
12
):
1107
20
.
[PubMed]
0028-4793
34.
Fischer
K
,
Al-Sawaf
O
,
Bahlo
J
,
Fink
AM
,
Tandon
M
,
Dixon
M
, et al.
Venetoclax and obinutuzumab in patients with CLL and coexisting conditions
.
N Engl J Med
.
2019
Jun
;
380
(
23
):
2225
36
.
[PubMed]
0028-4793
35.
Hillmen
P
,
Rawstron
AC
,
Brock
K
,
Muñoz-Vicente
S
,
Yates
FJ
,
Bishop
R
, et al.
Ibrutinib plus venetoclax in relapsed/refractory chronic lymphocytic leukemia: the CLARITY study
.
J Clin Oncol
.
2019
Oct
;
37
(
30
):
2722
9
.
[PubMed]
0732-183X
36.
Jain
N
,
Keating
M
,
Thompson
P
,
Ferrajoli
A
,
Burger
J
,
Borthakur
G
, et al.
Ibrutinib and venetoclax for first-line treatment of CLL
.
N Engl J Med
.
2019
May
;
380
(
22
):
2095
103
.
[PubMed]
0028-4793
37.
Kumar
S
,
Kaufman
JL
,
Gasparetto
C
,
Mikhael
J
,
Vij
R
,
Pegourie
B
, et al.
Efficacy of venetoclax as targeted therapy for relapsed/refractory t(11;14) multiple myeloma
.
Blood
.
2017
Nov
;
130
(
22
):
2401
9
.
[PubMed]
0006-4971
38.
Moreau
P
,
Chanan-Khan
A
,
Roberts
AW
,
Agarwal
AB
,
Facon
T
,
Kumar
S
, et al.
Promising efficacy and acceptable safety of venetoclax plus bortezomib and dexamethasone in relapsed/refractory MM
.
Blood
.
2017
Nov
;
130
(
22
):
2392
400
.
[PubMed]
0006-4971
39.
DiNardo
CD
,
Pratz
KW
,
Letai
A
,
Jonas
BA
,
Wei
AH
,
Thirman
M
, et al.
Safety and preliminary efficacy of venetoclax with decitabine or azacitidine in elderly patients with previously untreated acute myeloid leukaemia: a non-randomised, open-label, phase 1b study
.
Lancet Oncol
.
2018
Feb
;
19
(
2
):
216
28
.
[PubMed]
1470-2045
40.
Aldoss
I
,
Yang
D
,
Aribi
A
,
Ali
H
,
Sandhu
K
,
Al Malki
MM
, et al.
Efficacy of the combination of venetoclax and hypomethylating agents in relapsed/refractory acute myeloid leukemia
.
Haematologica
.
2018
Sep
;
103
(
9
):
e404
7
.
[PubMed]
0390-6078
41.
DiNardo
CD
,
Pratz
K
,
Pullarkat
V
,
Jonas
BA
,
Arellano
M
,
Becker
PS
, et al.
Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia
.
Blood
.
2019
Jan
;
133
(
1
):
7
17
.
[PubMed]
0006-4971
42.
Mei
M
,
Aldoss
I
,
Marcucci
G
,
Pullarkat
V
.
Hypomethylating agents in combination with venetoclax for acute myeloid leukemia: update on clinical trial data and practical considerations for use
.
Am J Hematol
.
2019
Mar
;
94
(
3
):
358
62
.
[PubMed]
1096-8652
43.
Ram
R
,
Amit
O
,
Zuckerman
T
,
Gurion
R
,
Raanani
P
,
Bar-On
Y
, et al.
Venetoclax in patients with acute myeloid leukemia refractory to hypomethylating agents-a multicenter historical prospective study
.
Ann Hematol
.
2019
Aug
;
98
(
8
):
1927
32
.
[PubMed]
0939-5555
44.
Wierda
WG
,
Siddiqi
T
,
Flinn
I
, et al.
Phase 2 CAPTIVATE results of ibrutinib (ibr) plus venetoclax (ven) in first-line chronic lymphocytic leukemia (CLL). J Clin Oncol.
2018
;36 (suppl; abstr 7502).
45.
Miller
BW
,
Przepiorka
D
,
de Claro
RA
,
Lee
K
,
Nie
L
,
Simpson
N
, et al.
FDA approval: idelalisib monotherapy for the treatment of patients with follicular lymphoma and small lymphocytic lymphoma
.
Clin Cancer Res
.
2015
Apr
;
21
(
7
):
1525
9
.
[PubMed]
1078-0432
46.
Flinn
IW
,
Hillmen
P
,
Montillo
M
,
Nagy
Z
,
Illés
Á
,
Etienne
G
, et al.
The phase 3 DUO trial: duvelisib vs ofatumumab in relapsed and refractory CLL/SLL
.
Blood
.
2018
Dec
;
132
(
23
):
2446
55
.
[PubMed]
0006-4971
47.
Flinn
IW
,
Miller
CB
,
Ardeshna
KM
,
Tetreault
S
,
Assouline
SE
,
Mayer
J
, et al.
DYNAMO: a phase II study of duvelisib (IPI-145) in patients with refractory indolent non-Hodgkin lymphoma
.
J Clin Oncol
.
2019
Apr
;
37
(
11
):
912
22
.
[PubMed]
0732-183X
48.
Crombie
JL
,
Teykucheva
S
,
Savell
A
, et al.
A phase I study of duvelisib and venetoclax in patients with relapsed or refractory CLL / SLL
.
Blood
.
2019
;
134
Suppl. 1
:
1763
. 0006-4971
49.
Karp
JE
,
Smith
BD
,
Levis
MJ
,
Gore
SD
,
Greer
J
,
Hattenburg
C
, et al.
Sequential flavopiridol, cytosine arabinoside, and mitoxantrone: a phase II trial in adults with poor-risk acute myelogenous leukemia
.
Clin Cancer Res
.
2007
Aug
;
13
(
15 Pt 1
):
4467
73
.
[PubMed]
1078-0432
50.
Blum
KA
,
Ruppert
AS
,
Woyach
JA
,
Jones
JA
,
Andritsos
L
,
Flynn
JM
, et al.
Risk factors for tumor lysis syndrome in patients with chronic lymphocytic leukemia treated with the cyclin-dependent kinase inhibitor, flavopiridol
.
Leukemia
.
2011
Sep
;
25
(
9
):
1444
51
.
[PubMed]
0887-6924
51.
Lin
TS
,
Ruppert
AS
,
Johnson
AJ
,
Fischer
B
,
Heerema
NA
,
Andritsos
LA
, et al.
Phase II study of flavopiridol in relapsed chronic lymphocytic leukemia demonstrating high response rates in genetically high-risk disease
.
J Clin Oncol
.
2009
Dec
;
27
(
35
):
6012
8
.
[PubMed]
0732-183X
52.
Lanasa
MC
,
Andritsos
L
,
Brown
JR
,
Gabrilove
J
,
Caligaris-Cappio
F
,
Ghia
P
, et al.
Final results of EFC6663: a multicenter, international, phase 2 study of alvocidib for patients with fludarabine-refractory chronic lymphocytic leukemia
.
Leuk Res
.
2015
May
;
39
(
5
):
495
500
.
[PubMed]
0145-2126
53.
Gojo
I
,
Sadowska
M
,
Walker
A
,
Feldman
EJ
,
Iyer
SP
,
Baer
MR
, et al.
Clinical and laboratory studies of the novel cyclin-dependent kinase inhibitor dinaciclib (SCH 727965) in acute leukemias
.
Cancer Chemother Pharmacol
.
2013
Oct
;
72
(
4
):
897
908
.
[PubMed]
0344-5704
54.
Stephens
DM
,
Ruppert
AS
,
Maddocks
K
,
Andritsos
L
,
Baiocchi
R
,
Jones
J
, et al.
Cyclophosphamide, alvocidib (flavopiridol), and rituximab, a novel feasible chemoimmunotherapy regimen for patients with high-risk chronic lymphocytic leukemia
.
Leuk Res
.
2013
Oct
;
37
(
10
):
1195
9
.
[PubMed]
0145-2126
55.
Gao
JJ
,
Cheng
J
,
Bloomquist
E
,
Sanchez
J
,
Wedam
SB
,
Singh
H
, et al.
CDK4/6 inhibitor treatment for patients with hormone receptor-positive, HER2-negative, advanced or metastatic breast cancer: a US Food and Drug Administration pooled analysis
.
Lancet Oncol
.
2020
Feb
;
21
(
2
):
250
60
.
[PubMed]
1470-2045
56.
Williams
SM
,
Killeen
AA
.
Tumor lysis syndrome
.
Arch Pathol Lab Med
.
2019
Mar
;
143
(
3
):
386
93
.
[PubMed]
0003-9985
57.
Terzi Demirsoy
E
,
Birtas Atesoglu
E
,
Eren
N
,
Gedük
A
,
Mehtap
O
,
Tarkun
P
, et al.
Carfilzomib-induced tumor lysis syndrome in relapsed multiple myeloma: a report of two cases
.
Tumori
.
2019
Dec
;
105
(
6
):
NP24
7
.
[PubMed]
0300-8916
58.
Suzuki
K
,
Nishiwaki
K
,
Gunji
T
,
Katori
M
,
Hosoba
R
,
Hirano
K
, et al.
Tumor-lysis syndrome in relapsed or refractory multiple myeloma patients treated with proteasome inhibitors
.
Blood
.
2018
;
132
Supplement 1
:
5631
. 0006-4971
59.
Yang
H
,
Rosove
MH
,
Figlin
RA
.
Tumor lysis syndrome occurring after the administration of rituximab in lymphoproliferative disorders: high-grade non-Hodgkin’s lymphoma and chronic lymphocytic leukemia
.
Am J Hematol
.
1999
Dec
;
62
(
4
):
247
50
.
[PubMed]
0361-8609
60.
Cartron
G
,
de Guibert
S
,
Dilhuydy
MS
,
Morschhauser
F
,
Leblond
V
,
Dupuis
J
, et al.
Obinutuzumab (GA101) in relapsed/refractory chronic lymphocytic leukemia: final data from the phase 1/2 GAUGUIN study
.
Blood
.
2014
Oct
;
124
(
14
):
2196
202
.
[PubMed]
0006-4971
61.
Moreno
C
,
Greil
R
,
Demirkan
F
,
Tedeschi
A
,
Anz
B
,
Larratt
L
, et al.
Ibrutinib plus obinutuzumab versus chlorambucil plus obinutuzumab in first-line treatment of chronic lymphocytic leukaemia (iLLUMINATE): a multicentre, randomised, open-label, phase 3 trial
.
Lancet Oncol
.
2019
Jan
;
20
(
1
):
43
56
.
[PubMed]
1470-2045
62.
Cheson
BD
,
Heitner Enschede
S
,
Cerri
E
,
Desai
M
,
Potluri
J
,
Lamanna
N
, et al.
Tumor lysis syndrome in chronic lymphocytic leukemia with novel targeted agents
.
Oncologist
.
2017
Nov
;
22
(
11
):
1283
91
.
[PubMed]
1083-7159
63.
Badoux
XC
,
Keating
MJ
,
Wen
S
,
Wierda
WG
,
O’Brien
SM
,
Faderl
S
, et al.
Phase II study of lenalidomide and rituximab as salvage therapy for patients with relapsed or refractory chronic lymphocytic leukemia
.
J Clin Oncol
.
2013
Feb
;
31
(
5
):
584
91
.
[PubMed]
0732-183X
64.
Porter
DL
,
Levine
BL
,
Kalos
M
,
Bagg
A
,
June
CH
.
Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia
.
N Engl J Med
.
2011
Aug
;
365
(
8
):
725
33
.
[PubMed]
0028-4793
65.
Maude
SL
,
Laetsch
TW
,
Buechner
J
,
Rives
S
,
Boyer
M
,
Bittencourt
H
, et al.
Tisagenlecleucel in children and young adults with B- cell lymphoblastic leukemia
.
N Engl J Med
.
2018
Feb
;
378
(
5
):
439
48
.
[PubMed]
0028-4793
66.
Kochenderfer
JN
,
Dudley
ME
,
Carpenter
RO
,
Kassim
SH
,
Rose
JJ
,
Telford
WG
, et al.
Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation
.
Blood
.
2013
Dec
;
122
(
25
):
4129
39
.
[PubMed]
0006-4971
67.
Schuster
SJ
,
Svoboda
J
,
Chong
EA
,
Nasta
SD
,
Mato
AR
,
Anak
Ö
, et al.
Chimeric antigen receptor T cells in refractory B-Cell lymphomas
.
N Engl J Med
.
2017
Dec
;
377
(
26
):
2545
54
.
[PubMed]
0028-4793
68.
Neelapu
SS
,
Locke
FL
,
Bartlett
NL
,
Lekakis
LJ
,
Miklos
DB
,
Jacobson
CA
, et al.
Axicabtagene ciloleucel CAR T-Cell therapy in refractory large B-Cell lymphoma
.
N Engl J Med
.
2017
Dec
;
377
(
26
):
2531
44
.
[PubMed]
0028-4793
69.
Mahadeo
KM
,
Khazal
SJ
,
Abdel-Azim
H
,
Fitzgerald
JC
,
Taraseviciute
A
,
Bollard
CM
, et al.;
Pediatric Acute Lung Injury and Sepsis Investigators (PALISI) Network
.
Management guidelines for paediatric patients receiving chimeric antigen receptor T cell therapy
.
Nat Rev Clin Oncol
.
2019
Jan
;
16
(
1
):
45
63
.
[PubMed]
1759-4774
70.
Cordrey
EO
,
Wang
J
.
Tumor lysis syndrome associated with immune checkpoint blockade in solid tumors
.
Jpn J Cancer Oncol Res.
2018
;
1
(
1
):
1005
.
71.
Jones
GL
,
Will
A
,
Jackson
GH
,
Webb
NJ
,
Rule
S
;
British Committee for Standards in Haematology
.
Guidelines for the management of tumour lysis syndrome in adults and children with haematological malignancies on behalf of the British Committee for Standards in Haematology
.
Br J Haematol
.
2015
Jun
;
169
(
5
):
661
71
.
[PubMed]
0007-1048
72.
Sanofi-Aventis
. (
2011
). Elitek (rasburicase) package insert. Re¬trieved from http://products.sanofi.us/elitek/elitek.html
73.
Zelenetz
AD
,
Gordon
LI
,
Wierda
WG
,
Abramson
JS
,
Advani
RH
,
Andreadis
CB
, et al.
NCCN Clinical Practice Guidelines in Oncology: Non-Hodgkin’s lymphomas, Version 4.2014
.
Natl Compr Canc Netw.
2014
;
12
(
9
):
1282
303
.
74.
Khosravan
R
,
Grabowski
BA
,
Wu
JT
,
Joseph-Ridge
N
,
Vernillet
L
.
Pharmacokinetics, pharmacodynamics and safety of febuxostat, a non-purine selective inhibitor of xanthine oxidase, in a dose escalation study in healthy subjects
.
Clin Pharmacokinet
.
2006
;
45
(
8
):
821
41
.
[PubMed]
0312-5963
75.
Spina
M
,
Nagy
Z
,
Ribera
JM
,
Federico
M
,
Aurer
I
,
Jordan
K
, et al.;
FLORENCE Study Group
.
FLORENCE: a randomized, double-blind, phase III pivotal study of febuxostat versus allopurinol for the prevention of tumor lysis syndrome (TLS) in patients with hematologic malignancies at intermediate to high TLS risk
.
Ann Oncol
.
2015
Oct
;
26
(
10
):
2155
61
.
[PubMed]
0923-7534
76.
Tamura
K
,
Kawai
Y
,
Kiguchi
T
,
Okamoto
M
,
Kaneko
M
,
Maemondo
M
, et al.
Efficacy and safety of febuxostat for prevention of tumor lysis syndrome in patients with malignant tumors receiving chemotherapy: a phase III, randomized, multi-center trial comparing febuxostat and allopurinol
.
Int J Clin Oncol
.
2016
Oct
;
21
(
5
):
996
1003
.
[PubMed]
1341-9625
77.
Bellos
I
,
Kontzoglou
K
,
Psyrri
A
,
Pergialiotis
V
.
Febuxostat administration for the prevention of tumour lysis syndrome: A meta-analysis
.
J Clin Pharm Ther
.
2019
Aug
;
44
(
4
):
525
33
.
[PubMed]
0269-4727
78.
Choi
H
,
Neogi
T
,
Stamp
L
,
Dalbeth
N
,
Terkeltaub
R
.
Implications of the cardiovascular safety of febuxostat and allopurinol in patients with gout and cardiovascular morbidities (CARES) trial and associated FDA public safety alert
.
Arthritis Rheumatol
.
2018
;
70
(
11
):
1702
9
.
[PubMed]
2326-5191
80.
Larson
RA
,
Pui
CH
. Tumor lysis syndrome: Prevention and treatment. UpToDate,
2019
; https://www.uptodate.com
81.
Coiffier
B
,
Altman
A
,
Pui
CH
,
Younes
A
,
Cairo
MS
.
Guidelines for the management of pediatric and adult tumor lysis syndrome: an evidence-based review
.
J Clin Oncol
.
2008
Jun
;
26
(
16
):
2767
78
.
[PubMed]
0732-183X
82.
Herrington
JD
,
Dinh
BC
.
Fixed, low-dose rasburicase for the treatment or prevention of hyperuricemia in adult oncology patients
.
J Oncol Pharm Pract
.
2015
Apr
;
21
(
2
):
111
7
.
[PubMed]
1078-1552
83.
Vadhan-Raj
S
,
Fayad
LE
,
Fanale
MA
,
Pro
B
,
Rodriguez
A
,
Hagemeister
FB
, et al.
A randomized trial of a single-dose rasburicase versus five-daily doses in patients at risk for tumor lysis syndrome
.
Ann Oncol
.
2012
Jun
;
23
(
6
):
1640
5
.
[PubMed]
0923-7534
84.
Trifilio
SM
,
Pi
J
,
Zook
J
,
Golf
M
,
Coyle
K
,
Greenberg
D
, et al.
Effectiveness of a single 3-mg rasburicase dose for the management of hyperuricemia in patients with hematological malignancies
.
Bone Marrow Transplant
.
2011
Jun
;
46
(
6
):
800
5
.
[PubMed]
0268-3369
85.
Coutsouvelis
J
,
Wiseman
M
,
Hui
L
,
Poole
S
,
Dooley
M
,
Patil
S
, et al.
Effectiveness of a single fixed dose of rasburicase 3 mg in the management of tumour lysis syndrome
.
Br J Clin Pharmacol
.
2013
Feb
;
75
(
2
):
550
3
.
[PubMed]
0306-5251
86.
Patel
KS
,
Lau
JE
,
Zembillas
AS
,
Gallagher
EM
.
Single 4.5 mg fixed-dose of rasburicase for hyperuricemia associated with tumor lysis syndrome
.
J Oncol Pharm Pract
.
2017
Jul
;
23
(
5
):
333
7
.
[PubMed]
1078-1552
87.
Nauffal
M
,
Redd
R
,
Ni
J
,
Stone
RM
,
DeAngelo
DJ
,
McDonnell
AM
.
Single 6-mg dose of rasburicase: the experience in a large academic medical center
.
J Oncol Pharm Pract
.
2019
Sep
;
25
(
6
):
1349
56
.
[PubMed]
1078-1552
88.
Feng
X
,
Dong
K
,
Pham
D
,
Pence
S
,
Inciardi
J
,
Bhutada
NS
.
Efficacy and cost of single-dose rasburicase in prevention and treatment of adult tumour lysis syndrome: a meta-analysis
.
J Clin Pharm Ther
.
2013
Aug
;
38
(
4
):
301
8
.
[PubMed]
0269-4727
89.
Yu
X
,
Liu
L
,
Nie
X
,
Li
J
,
Zhang
J
,
Zhao
L
, et al.
The optimal single-dose regimen of rasburicase for management of tumour lysis syndrome in children and adults: a systematic review and meta-analysis
.
J Clin Pharm Ther
.
2017
Feb
;
42
(
1
):
18
26
.
[PubMed]
0269-4727
90.
Cortes
J
,
Moore
JO
,
Maziarz
RT
,
Wetzler
M
,
Craig
M
,
Matous
J
, et al.
Control of plasma uric acid in adults at risk for tumor Lysis syndrome: efficacy and safety of rasburicase alone and rasburicase followed by allopurinol compared with allopurinol alone—results of a multicenter phase III study
.
J Clin Oncol
.
2010
Sep
;
28
(
27
):
4207
13
.
[PubMed]
0732-183X
91.
Cairo
MS
,
Thompson
S
,
Tangirala
K
,
Eaddy
MT
.
A clinical and economic comparison of rasburicase and allopurinol in the treatment of patients with clinical or laboratory tumor lysis syndrome
.
Clin Lymphoma Myeloma Leuk
.
2017
Mar
;
17
(
3
):
173
8
.
[PubMed]
2152-2650
92.
Rasburicase [package insert]. Sanofi-Aventis US LLC; Bridgewater:
2009
.
93.
Relling
MV
,
McDonagh
EM
,
Chang
T
,
Caudle
KE
,
McLeod
HL
,
Haidar
CE
, et al.;
Clinical Pharmacogenetics Implementation Consortium
.
Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for rasburicase therapy in the context of G6PD deficiency genotype
.
Clin Pharmacol Ther
.
2014
Aug
;
96
(
2
):
169
74
.
[PubMed]
0009-9236
94.
Allen
KC
,
Champlain
AH
,
Cotliar
JA
,
Belknap
SM
,
West
DP
,
Mehta
J
, et al.
Risk of anaphylaxis with repeated courses of rasburicase: a Research on Adverse Drug Events and Reports (RADAR) project
.
Drug Saf
.
2015
Feb
;
38
(
2
):
183
7
.
[PubMed]
0114-5916
95.
AbbVie
.
Highlights of prescribing information.
2016
. http://www.rxabbvie.com/pdf/venclexta.pdf (accessed Jan 1, 2020).
96.
Flombaum
CD
.
Metabolic emergencies in the cancer patient
.
Semin Oncol
.
2000
Jun
;
27
(
3
):
322
34
.
[PubMed]
0093-7754
97.
Darmon
M
,
Vincent
F
,
Camous
L
,
Canet
E
,
Bonmati
C
,
Braun
T
, et al.;
Groupe de Recherche en Réanimation Respiratoire et Onco-Hématologique (GRRR-OH)
.
Tumour lysis syndrome and acute kidney injury in high-risk haematology patients in the rasburicase era. A prospective multicentre study from the Groupe de Recherche en Réanimation Respiratoire et Onco-Hématologique
.
Br J Haematol
.
2013
Aug
;
162
(
4
):
489
97
.
[PubMed]
0007-1048
98.
Lin
CJ
,
Chen
HH
,
Hsieh
RK
,
Chen
YC
,
Wu
CJ
.
Acute tumor lysis syndrome in a hemodialysis patient with diffuse large B cell lymphoma
.
Med Oncol
.
2009
;
26
(
1
):
93
5
.
[PubMed]
1357-0560
99.
Wang
Y
,
Lu
J
,
Tao
Y
.
Impact of daytime continuous veno-venous haemofiltration on treatment of paediatric tumour lysis syndrome
.
J Int Med Res
.
2018
Sep
;
46
(
9
):
3613
20
.
[PubMed]
0300-0605
100.
Choi
KA
,
Lee
JE
,
Kim
YG
,
Kim
DJ
,
Kim
K
,
Ko
YH
, et al.
Efficacy of continuous venovenous hemofiltration with chemotherapy in patients with Burkitt lymphoma and leukemia at high risk of tumor lysis syndrome
.
Ann Hematol
.
2009
Jul
;
88
(
7
):
639
45
.
[PubMed]
0939-5555
101.
Tan
HK
,
Bellomo
R
,
M’Pis
DA
,
Ronco
C
.
Phosphatemic control during acute renal failure: intermittent hemodialysis versus continuous hemodiafiltration
.
Int J Artif Organs
.
2001
Apr
;
24
(
4
):
186
91
.
[PubMed]
0391-3988
Open Access License / Drug Dosage / Disclaimer
This article is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License (CC BY-NC-ND). Usage and distribution for commercial purposes as well as any distribution of modified material requires written permission. Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug. Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.