12,524
Views
144
CrossRef citations to date
0
Altmetric
Reviews

Asparaginase-associated toxicity in children with acute lymphoblastic leukemia

&
Pages 748-757 | Received 25 Jun 2015, Accepted 21 Sep 2015, Published online: 20 Nov 2015

Abstract

Asparaginase is an integral component of multiagent chemotherapy regimens for the treatment of children with acute lymphoblastic leukemia. Positive outcomes are seen in patients who are able to complete their entire prescribed course of asparaginase therapy. Toxicities associated with asparaginase use include hypersensitivity (clinical and subclinical), pancreatitis, thrombosis, encephalopathy, and liver dysfunction. Depending on the nature and severity of the toxicity, asparaginase therapy may be altered or discontinued in some patients. Clinical hypersensitivity is the most common asparaginase-associated toxicity requiring treatment discontinuation, occurring in up to 30% of patients receiving Escherichia coli–derived asparaginase. The ability to rapidly identify and manage asparaginase-associated toxicity will help ensure patients receive the maximal benefit from asparaginase therapy. This review will provide an overview of the common toxicities associated with asparaginase use and recommendations for treatment management.

Introduction

With current multiagent chemotherapy regimens, long-term outcomes for children with acute lymphoblastic leukemia (ALL) have greatly improved, with reported overall survival rates > 90% compared with < 30% in the 1960s [Citation1,Citation2]. These substantial gains in survival are due, at least in part, to the increased use of intense and prolonged asparaginase therapy [Citation3–5]. However, asparaginase use is associated with a number of toxicities. Failure to receive the full course of asparaginase therapy due to treatment-emergent toxicity has been associated with poor outcomes in children with ALL [Citation3,Citation6–8]. This review provides a concise overview of common toxicities associated with asparaginase therapy and recommendations for management.

Mechanism of action of asparaginase

The administration of asparaginase reduces plasma asparagine concentrations by catalyzing the deamination of asparagine into aspartic acid and ammonia [Citation8]. At sufficient enzyme activity levels, asparaginase therapy results in the complete depletion of serum asparagine concentrations, depriving leukemic blasts of this amino acid [Citation9], resulting in reduced protein synthesis and ultimately leukemic cell death.

Three different asparaginase preparations are currently used for the treatment of patients with ALL. Two preparations, native Escherichia coli (E. coli) asparaginase and polyethylene glycolated (PEG)-asparaginase, are derived from the bacterium E. coli [Citation10]. Native E. coli asparaginase has been widely used as a first-line treatment in ALL; however, the supply of this preparation has recently ceased in the United States and has largely been replaced with PEG-asparaginase [Citation11]. The third preparation, Erwinia asparaginase, is derived from the bacterium Erwinia chrysanthemi [Citation12]. The distinct bacterial origins of Erwinia asparaginase give it a unique immunogenic profile, showing no cross-reactivity with native E. coli asparaginase or PEG-asparaginase [Citation13]. Erwinia asparaginase is indicated as a component of a multiagent chemotherapy regimen for the treatment of patients with ALL who have developed hypersensitivity to E. coli–derived asparaginases [Citation14]. Each of the three asparaginase preparations displays markedly different pharmacokinetics that must be accounted for when determining dosing schedules [Citation15]. The half-life of PEG-asparaginase has been estimated at 4.8–7.0 days and is longer than that reported with native E. coli asparaginase and Erwinia asparaginase [Citation16–19]. In comparison, the half-life of native E. coli asparaginase and Erwinia asparaginase are 1.28 days and 15.6 hours, respectively [Citation14,Citation19]. Due to the shorter half-life of Erwinia asparaginase, patients who switch to this formulation should receive a higher dose at a greater frequency in order to maintain therapeutic levels of asparagine depletion [Citation20].

Asparaginase toxicity

Hypersensitivity

The asparaginases used in ALL treatment protocols are large molecules of bacterial origin, and thus have the ability to elicit an immune response in patients [Citation8]. Immune reactions to asparaginase are classified as either clinical or subclinical hypersensitivity (also referred to as “silent inactivation”). Clinical hypersensitivity is one of the most common reasons for the discontinuation of asparaginase therapy.

Rates of clinical hypersensitivity reactions in the literature vary. Clinical hypersensitivity to native E. coli asparaginase has been reported in up to 75% of patients with ALL [Citation21], although rates generally range from 10–30% [Citation7,Citation22–26]. Clinical hypersensitivity reactions appear to be less prevalent with PEG-asparaginase, with rates from 3–24% reported in clinical trials [Citation3,Citation7,Citation24,Citation26]. Hypersensitivity reactions to PEG-asparaginase are more common when patients have been previously exposed to native E. coli asparaginase, owing to their common bacterial source [Citation27].

Rates of clinical hypersensitivity in patients receiving Erwinia asparaginase, which is derived from an alternative bacterial source, have been reported in 3–37% of patients in clinical trials [Citation7,Citation14,Citation20,Citation26,Citation28–34]. Patients who develop clinical hypersensitivity to asparaginase have shown increased antibody formation and decreased asparaginase activity levels compared with patients who do not develop hypersensitivity [Citation32,Citation35,Citation36]. Tong and colleagues [Citation32] reported clinical hypersensitivity (grades 1–4) in 20 of 89 patients (22%) administered PEG-asparaginase during the intensification phase after receiving native E. coli asparaginase during the induction phase of the Dutch Childhood Oncology Group (DCOG) ALL-10 protocol. All 20 patients with hypersensitivity showed PEG-asparaginase activity levels of 0 IU/L, and E. coli antibodies were found in all patients with hypersensitivity during intensification.

The likelihood of asparaginase eliciting an immune response in patients may be influenced by a number of factors, including the asparaginase preparation, treatment intensity, and use of concomitant medications [Citation23,Citation24,Citation29,Citation36,Citation48]. The risk of antibody formation for patients increases with repeated exposure to asparaginase; consolidation and reinduction phases of treatment show the greatest incidence of hypersensitivity reactions and antibody formation [Citation30,Citation49]. However, prolonged exposure to asparaginase, without gaps in treatment, has been associated with decreased antibody levels [Citation36,Citation50]. Due to this, hypersensitivity reactions are most common within the first few doses of asparaginase after a break in treatment [Citation32]. Additionally, the concomitant administration of corticosteroids has been associated with reduced signs of clinical hypersensitivity [Citation24,Citation25]. However, as clinical hypersensitivity is often the only observable symptom of antibody formation, suppression of these symptoms may only mask signs of hypersensitivity and result in prolonged periods of suboptimal asparaginase activity levels in patients. Whenever possible, therapeutic dose monitoring of asparaginase activity levels should be utilized to identify patients with suboptimal activity levels to adjust treatment in these patients accordingly [Citation32].

Patients who display a hypersensitivity reaction to an E. coli–derived asparaginase should immediately discontinue their current therapy and be switched to Erwinia asparaginase () [Citation33, Citation37]. Patients with hypersensitivity switched to Erwinia asparaginase show therapeutic levels of asparaginase activity, and the majority of patients with hypersensitivity are able to complete their prescribed course of treatment [Citation33]. The FDA-approved dose substitution of Erwinia asparaginase for each planned dose of PEG-asparaginase is 25,000 IU/m2 administered intramuscularly (IM) or intravenously (IV) 3 times a week (Monday/Wednesday/Friday) for six doses [Citation14]. No established guidelines exist for when treatment with the new preparation should be initiated in patients who switch asparaginase preparations because of clinical hypersensitivity. In practice, many patients with hypersensitivity begin treatment with the new preparation at the time of their next scheduled dose. Given the strong association between clinical hypersensitivity and reduced asparaginase activity [Citation32], this practice may result in suboptimal asparagine depletion in the patients between doses. To avoid a prolonged gap in asparagine depletion, patients with hypersensitivity to asparaginase should begin treatment with an alternative asparaginase as early as possible, preferably within 48–72 hours after the hypersensitivity event if symptoms are fully resolved [Citation51,Citation52]. Patients who develop hypersensitivity to both E. coli– and Erwinia chrysanthemiderived formulations are forced to discontinue asparaginase treatment [Citation32].

Table I. Asparaginase-associated toxicities and recommendations for management of asparaginase therapy [Citation33,Citation37Citation47].

Subclinical hypersensitivity is characterized by the development of antiasparaginase antibodies and significantly reduced asparaginase activity levels [Citation19,Citation37]. Although difficult to identify because of the lack of clinical symptoms, subclinical hypersensitivity has been reported in 8–29% of patients receiving E. coli–derived asparaginases [Citation25,Citation32,Citation37]. Subclinical hypersensitivity is strongly associated with poor clinical outcomes if not readily identified and addressed [Citation25,Citation37]. The ability to prospectively identify patients with subclinical hypersensitivity and switch these patients to an alternate asparaginase formulation has been associated with improved outcomes in a clinical trial [Citation37].

Following the IV administration of asparaginase, localized non-antibody-mediated infusion reactions may occur in patients [Citation53]. Unlike true clinical hypersensitivity reactions, patients who display an infusion reaction to asparaginase do not show antiasparaginase antibodies and infusion reactions are not associated with a decrease in asparaginase activity levels. Patients with a non-antibody-mediated infusion reaction to asparaginase may be rechallenged with a longer infusion duration and appropriate premedication. Differentiating between clinical hypersensitivity, subclinical hypersensitivity, and non-antibody-mediated infusion reactions can be difficult in practice. But measurement of asparaginase activity levels might help to differentiate between these reactions.

Hyperglycemia

The use of asparaginase is associated with reduced insulin production and possibly a decrease in the expression of insulin receptors [Citation38,Citation54]. Corticosteroid use is associated with greater insulin resistance and an increase in hepatic gluconeogenesis [Citation55,Citation56]. Hyperglycemia is more common during phases of therapy when asparaginase and corticosteroids are administered together and in relatively higher doses [Citation38,Citation57].

Asparaginase-associated hyperglycemia has been reported in 4–20% of pediatric patients receiving E. coli asparaginase for ALL [Citation57–60] and in 4–17% of patients receiving Erwinia asparaginase [Citation14,Citation33,Citation34]. The asparaginase preparation and the type of corticosteroid (prednisone or dexamethasone) used in treatment do not seem to influence the risk of hyperglycemia [Citation24,Citation58,Citation60]. Insulin therapy may be required in severe cases; however, asparaginase therapy can continue if the patient shows normal glucose levels (< 200 mg/dL or 11 mmol/L) with insulin [] [Citation8,Citation38,Citation40]. The final decision regarding the continuation of asparagainse should be made by the treating physician based on the patient’s general status.

Pancreatitis

While the precise pathogenesis of pancreatitis is unknown, the reduction in protein synthesis resulting from asparaginase-induced depletion of asparagine has been implicated [Citation61]. In clinical trials, pancreatitis has been reported in 2–18% of patients undergoing asparaginase therapy for ALL, with grade 3/4 pancreatitis occurring in 5–10% of patients [Citation39,Citation61–65]. The formulation of asparaginase does not influence the incidence of pancreatitis in patients [Citation61].

Diagnosis of pancreatitis is based on a combination of clinical, biochemical (amylase, lipase), and radiological evidence. Asparaginase therapy can be continued with mildly elevated amylase or lipase levels if clinical signs are absent; however, asparaginase treatment is generally discontinued in the case of severe pancreatitis [] [Citation39,Citation40]. Patients with mild pancreatitis may be rechallenged with asparaginase if within 48 hours the patient displays no clinical symptoms, amylase and lipase levels are below 3 times the upper limit of normal (ULN), and there are no signs of pseudocysts or necrosis on imaging [Citation61]. Caution should be exercised, as recurrence of pancreatitis has been reported in up to 63% of patients following re-exposure to asparaginase [Citation62].

Thrombosis

Asparaginase is associated with a decrease in the production of a number of proteins involved in coagulation and fibrinolysis, and may increase the risk of thrombosis or bleeding [Citation66–70]. The majority of asparaginase-associated thrombotic events occur during induction and are likely attributable to multiple factors, including indwelling central venous catheter, treatment with corticosteroids, treatment with asparaginase, and leukemia itself [Citation41,Citation71]. The incidence of symptomatic thrombosis ranges from 2–7% in clinical trials and has been reported with both E. coli– and Erwinia-derived asparaginase [Citation14,Citation33,Citation34,Citation41,Citation71–73]. A meta-analysis of 17 studies focused on thrombotic complications in children with ALL found that the overall incidence of thrombosis is 5.2% [Citation71]. The authors report that the majority of events (53.8%) occurred in the central nervous system (CNS), and 28.6% of the total events were classified as central venous thrombosis. Of the non-CNS events, the greatest incidence was seen in the upper limbs and was categorized as deep venous thrombosis and central venous catheter–related [Citation71].

Patients with deep venous thrombosis should be managed with anticoagulation therapy, preferably with low-molecular-weight heparin (LMWH) [Citation42]. Asparaginase use should be temporarily discontinued in the case of clinically significant bleeding or thrombotic events; however, reports suggest that re-exposure to asparaginase with LMWH is safe and feasible in patients who develop thrombosis once clinical symptoms have resolved [] [Citation43,Citation73]. Screening patients for prothrombotic risk factors has been suggested in some reports [Citation71]; however, the evidence linking thrombophilia and other risk factors to the incidence of thrombotic events is mixed [Citation74–78].

Encephalopathy

Encephalopathy has been reported in patients receiving asparaginase treatment for ALL, although the precise relationship between asparaginase and neurotoxicity is unclear [Citation40,Citation44,Citation79,Citation80]. Posterior reversible encephalopathy is one of the encephalopathies sometimes seen in patients with ALL. The majority of reported cases of posterior reversible encephalopathy in patients with ALL occur during induction treatment, which could also be related to hypertension caused by glucocorticoids, and resolve without serious complications in most cases. [Citation80–83].

Elevated plasma ammonia levels, due to the asparaginase-driven breakdown of asparagine into aspartic acid and ammonia, are sometimes associated with encephalopathy in patients undergoing asparaginase therapy [Citation79,Citation80,Citation84]; however, hyperammonemia alone does not typically result in symptoms, and reduced expression of the glutamine transporter proteins may also play a role [Citation85]. Patients with existing liver disease may be at an increased risk for developing symptoms of hyperammonemia. There is no standard therapy for patients with asparaginase-induced hyperammonemia. Treatments with decreased protein intake, lactulose treatment, benzoic acid, and arginine and sodium phenylbutyrate have been reported; however, there is sparse evidence for these treatments and their efficacy is unclear [Citation84,Citation86,Citation87].

Myelosuppression

Although a number of early studies described an association between asparaginase use and myelosuppression [Citation88,Citation89], asparaginase itself is not typically considered a myelosuppressive agent. Asparaginase may cause myelosuppression directly or indirectly by altering the myelosuppressive effects of other agents, such as methotrexate (MTX) or 6-mercaptopurine (6-MP) [Citation45,Citation90,Citation91]. A recent report of pediatric patients (<10 years of age) with ALL treated on the Dana-Farber Cancer Institute ALL Consortium Protocol 05-01 found increased myelosuppression during prolonged asparaginase therapy in consolidation [Citation45]. Patients received 30 weeks of asparaginase treatment during consolidation, but no asparaginase was administered during continuation. A greater percentage of patients required dose reductions of MTX and/or 6-MP during consolidation compared with continuation (24% vs. 9%, respectively), suggesting a myelosuppressive role of asparaginase. Dose reduction of concurrently administered myelosuppressive agents may be used to manage asparaginase-associated myelosuppression during consolidation ()[Citation45].

Hypertriglyceridemia

Asparaginase use is associated with a number of abnormalities in lipid metabolism, including hypertriglyceridemia [Citation8,Citation92]. Corticosteroids are adipokinetic agents that alter lipid synthesis, clearance, and metabolism, and thus contribute to a transient elevation of triglyceride levels [Citation92,Citation93]. Transient elevations in triglyceride levels are most often seen when patients receive high doses of asparaginase and corticosteroids [Citation94]. Combined treatment with asparaginase and corticosteroids leads to hypertriglyceridemia in up to 67% of patients receiving asparaginase treatment for ALL [Citation95].

Data from patients treated on the DCOG ALL-10 protocol showed a significant positive relationship between asparaginase activity levels and triglyceride levels [Citation46]. This study prospectively evaluated the incidence and clinical course of hypertriglyceridemia and hypercholesterolemia in 89 pediatric patients undergoing prolonged asparaginase therapy. Hypertriglyceridemia (grade 3/4) was more prevalent in patients receiving PEG-asparaginase compared with patients administered Erwinia asparaginase (47% vs. 0%, respectively) [Citation46]. Additionally, hypercholesterolemia (grade 3/4) was reported in 25% of patients receiving PEG-asparaginase compared with no patients administered Erwinia asparaginase. In this study, asparaginase activity levels were consistently higher in patients receiving PEG-asparaginase compared with patients receiving Erwinia asparaginase, possibly contributing to the greater incidence of hypertriglyceridemia and hypercholesterolemia.

Hypertriglyceridemia is often transient and asymptomatic in patients. Adjustments in asparaginase therapy are generally not required; however, patients with elevated triglyceride levels should be closely monitored for signs of pancreatitis [] [Citation46,Citation93,Citation95,Citation96]. Several treatment approaches have been reported for patients with hypertriglyceridemia, including short-term fasting or low-fat diet, oral fibrates, omega-3, and plasmapheresis [Citation47,Citation97–101]. Strong evidence for any specific option is lacking, and there is currently no standard treatment for hypertriglyceridemia. Tong and colleagues [Citation102] reported the successful resolution of severe hypertriglyceridemia in a pediatric patient by temporarily omitting dexamethasone courses without any further changes to therapy.

Hepatic toxicity

Hepatic toxicity associated with asparaginase use is rarely associated with fatal complications; however, clinical outcomes may be negatively affected if significant delays in treatment are required due to asparaginase-associated dysfunction. Asparaginase use is more commonly associated with abnormalities in liver function and hepatic transaminases as well as elevations in bilirubin and alkaline phosphates [Citation7]. The mechanism of action by which asparaginase causes hepatic dysfunction is unknown; however, the reduction in protein synthesis associated with asparaginase therapy is believed to play a role [Citation40]. The degree to which hepatic dysfunction in patients undergoing treatment for ALL can be attributed to asparaginase use is unclear, as many regimens include the use of several potentially hepatotoxic drugs (e.g. corticosteroids, vinca alkaloids, anthracyclines, and antimetabolites). Elevated levels of hepatic transaminase, alkaline phosphatase, and bilirubin have been reported in 30–60% of patients receiving asparaginase as part of multiagent therapy for ALL [Citation40,Citation95]. In a study of 118 children receiving native E. coli asparaginase or PEG-asparaginase, abnormal liver function (grade 3/4), including elevated transaminases and hyperbilirubinemia, was found in 8% of patients receiving native E. coli asparaginase and in 5% of patients receiving PEG-asparaginase [Citation24,Citation103,Citation104]. In children and adults receiving Erwinia asparaginase, grade 3/4 liver toxicity has been reported in approximately 4% of patients [Citation14,Citation33,Citation34,Citation105]. There are no clear pediatric guidelines for the management of asparaginase in patients with hepatic toxicity, and treatment recommendations vary across protocols. In the DCOG ALL-11 pediatric protocol, patients are required to display aspartate aminotransferase/alanine aminotransferase < 10 × ULN and no signs of jaundice with bilirubin < 3 × ULN prior to starting asparaginase treatment [Citation106]. Recommendations for adolescent and young adults (AYA) call for witholding asparaginase in patients with grade 3/4 hepatotoxicity (alanine or glutamine aminotransferase elevation >5 × ULN) with the option to rechallenge patients with careful monitoring if complications resolve [] [Citation40].

Toxicity in AYA patients

The treatment of AYA patients (16–39 years of age) diagnosed with ALL represents a unique challenge. Older patients are believed to be at a greater risk for asparaginase-associated toxicities, and therefore, many adult protocols limit asparaginase use [Citation107]. However, a number of retrospective studies have reported significantly greater long-term survival when AYA patients are included in pediatric protocols, which use high-intensity asparaginase therapy [Citation108–112]. A large prospective study, C10403, evaluated the feasibility of using a pediatric regimen in 318 AYA patients (16–39 years of age) treated by adult hematologists and oncologists [Citation113]. Safety and toxicity results were compared with data from AYA patients (16–21 years of age) treated on the pediatric Children’s Oncology Group AALL0232 trial. The rates of adverse events were relatively similar in the two study populations, and overall treatment-related mortality was low (3%) in the C10403 study. Investigators concluded that treatment with a pediatric regimen was feasible in AYA patients up to 40 years of age [Citation113]. Differences in toxicities between age groups were not evaluated in C10403; however, results from another recent trial suggest a similar safety profile in pediatric and AYA patients. In this compassionate-use trial, 147 AYA patients (16 to <40 years of age) were switched to Erwinia asparaginase following hypersensitivity to an E. coli–derived asparaginase [Citation34]. Rates of asparaginase-related toxicity were reportedly similar in AYA patients and in patients <16 years of age. Hypersensitivity to Erwinia asparaginase was reported in 10.9% of patients ≥16 years of age compared with 15.1% in patients ≤10 years of age. Overall, toxicities were manageable, and the majority of AYA patients (73%) were able to complete their planned course of asparaginase therapy with Erwinia asparaginase [Citation34].

Conclusions

Asparaginase is a critical component of all pediatric ALL protocols and is increasingly used to treat AYA patients. With many protocols incorporating prolonged and high-intensity asparaginase treatment, it is important that practitioners be aware of all potential treatment-related toxicities. Effective management of asparaginase toxicity will help ensure patients receive their complete course of asparaginase therapy and obtain optimal treatment outcomes.

Supplemental material

ICMJE Form for Disclosure of Potential Conflicts of Interest

Download Zip (2.7 MB)

Acknowledgements

This study was supported by Jazz Pharmaceuticals plc or its subsidiaries.

The authors wish to thank Cory Hussar, PhD, of The Curry Rockefeller Group, LLC, Tarrytown, NY, for providing editorial assistance that was supported by Jazz Pharmaceuticals plc or its subsidiaries.

Potential conflict of interest

Disclosure forms provided by the authors are available with the full text of this article at http://dx.doi.org/10.3109/10428194.2015.1101098.

References

  • Pui CH, Robison LL, Look AT. Acute lymphoblastic leukaemia. Lancet 2008; 371: 1030–1043.
  • Hunger SP, Loh ML, Whitlock JA, et al. Children’s Oncology Group’s 2013 blueprint for research: acute lymphoblastic leukemia. Pediatr Blood Cancer 2013; 60: 957–963.
  • Silverman LB, Gelber RD, Dalton VK, et al. Improved outcome for children with acute lymphoblastic leukemia: results of Dana-Farber Consortium Protocol 91-01. Blood 2001; 97: 1211–1218.
  • Pession A, Valsecchi MG, Masera G, et al. Long-term results of a randomized trial on extended use of high dose L-asparaginase for standard risk childhood acute lymphoblastic leukemia. J Clin Oncol 2005; 23: 7161–7167.
  • Amylon MD, Shuster J, Pullen J, et al. Intensive high-dose asparaginase consolidation improves survival for pediatric patients with T cell acute lymphoblastic leukemia and advanced stage lymphoblastic lymphoma: a Pediatric Oncology Group study. Leukemia 1999; 13: 335–342.
  • Stock W. Adolescents and young adults with acute lymphoblastic leukemia. Hematology Am Soc Hematol Educ Program 2010; 2010: 21–29.
  • Raetz EA, Salzer WL. Tolerability and efficacy of L-asparaginase therapy in pediatric patients with acute lymphoblastic leukemia. J Pediatr Hematol Oncol 2010; 32: 554–563.
  • Muller HJ, Boos J. Use of L-asparaginase in childhood ALL. Crit Rev Oncol Hematol 1998; 28: 97–113.
  • Riccardi R, Holcenberg JS, Glaubiger DL, Wood JH, Poplack DG. L-asparaginase pharmacokinetics and asparagine levels in cerebrospinal fluid of rhesus monkeys and humans. Cancer Res 1981; 41: 4554–4558.
  • Panetta JC, Gajjar A, Hijiya N, et al. Comparison of native E. coli and PEG asparaginase pharmacokinetics and pharmacodynamics in pediatric acute lymphoblastic leukemia. Clin Pharmacol Ther 2009; 86: 651–658.
  • Keding R. RE: Discontinuation of Elspar®, (asparaginase for injection) 10,000 IU; August 12, 2012. Accessed July 30, 2014 from: http://www.fda.gov/downloads/Drugs/DrugSafety/DrugShortages/UCM321556.pdf.
  • Salzer W, Seibel N, Smith M. Erwinia asparaginase in pediatric acute lymphoblastic leukemia. Expert Opin Biol Ther 2012; 12: 1407–1414.
  • Zalewska-Szewczyk B, Gach A, Wyka K, Bodalski J, Mlynarski W. The cross-reactivity of anti-asparaginase antibodies against different L-asparaginase preparations. Clin Exp Med 2009; 9: 113–116.
  • ERWINAZE [package insert]. Palo Alto, CA: Jazz Pharmaceuticals, Inc.; 2014. Accessed January 30, 2015 from: http://erwinaze.com/ERWINAZEPI.pdf
  • Asselin BL, Whitin JC, Coppola DJ, Rupp IP, Sallan SE, Cohen HJ. Comparative pharmacokinetic studies of three asparaginase preparations. J Clin Oncol 1993; 11: 1780–1786.
  • Avramis VI, Panosyan EH. Pharmacokinetic/pharmacodynamic relationships of asparaginase formulations: the past, the present and recommendations for the future. Clin Pharmacokinet 2005; 44: 367–393.
  • Douer D, Yampolsky H, Cohen LJ, et al. Pharmacodynamics and safety of intravenous pegaspargase during remission induction in adults aged 55 years or younger with newly diagnosed acute lymphoblastic leukemia. Blood 2007; 109: 2744–2750.
  • Angiolillo AL, Schore RJ, Devidas M, et al. Pharmacokinetic and pharmacodynamic properties of calaspargase pegol Escherichia coli L-asparaginase in the treatment of patients with acute lymphoblastic leukemia: results from Children's Oncology Group Study AALL07P4. J Clin Oncol 2014; 32: 3874–3882.
  • Asselin BL. The three asparaginases. Comparative pharmacology and optimal use in childhood leukemia. Adv Exp Med Biol 1999; 457: 621–629.
  • Pieters R, Hunger SP, Boos J, et al. L-asparaginase treatment in acute lymphoblastic leukemia: a focus on Erwinia asparaginase. Cancer 2011; 117: 238–249.
  • Wacker P, Land VJ, Camitta BM, et al. Allergic reactions to E. coli L-asparaginase do not affect outcome in childhood B-precursor acute lymphoblastic leukemia: a Children’s Oncology Group Study. J Pediatr Hematol Oncol 2007; 29: 627–632.
  • Muller HJ, Beier R, Loning L, et al. Pharmacokinetics of native Escherichia coli asparaginase (Asparaginase medac) and hypersensitivity reactions in ALL-BFM 95 reinduction treatment. Br J Haematol 2001; 114: 794–799.
  • Woo MH, Hak LJ, Storm MC, et al. Hypersensitivity or development of antibodies to asparaginase does not impact treatment outcome of childhood acute lymphoblastic leukemia. J Clin Oncol 2000; 18: 1525–1532.
  • Avramis VI, Sencer S, Periclou AP, et al. A randomized comparison of native Escherichia coli asparaginase and polyethylene glycol conjugated asparaginase for treatment of children with newly diagnosed standard-risk acute lymphoblastic leukemia: a Children’s Cancer Group study. Blood 2002; 99: 1986–1994.
  • Panosyan EH, Seibel NL, Martin-Aragon S, et al. Asparaginase antibody and asparaginase activity in children with higher-risk acute lymphoblastic leukemia: Children’s Cancer Group Study CCG-1961. J Pediatr Hematol Oncol 2004; 26: 217–226.
  • Vrooman LM, Supko JG, Neuberg DS, et al. Erwinia asparaginase after allergy to E coli asparaginase in children with acute lymphoblastic leukemia. Pediatr Blood Cancer 2010; 54: 199–205.
  • Seibel NL, Steinherz PG, Sather HN, et al. Early postinduction intensification therapy improves survival for children and adolescents with high-risk acute lymphoblastic leukemia: a report from the Children’s Oncology Group. Blood 2008; 111: 2548–2555.
  • Billett AL, Carls A, Gelber RD, Sallan SE. Allergic reactions to Erwinia asparaginase in children with acute lymphoblastic leukemia who had previous allergic reactions to Escherichia coli asparaginase. Cancer 1992; 70: 201–206.
  • Moghrabi A, Levy DE, Asselin B, et al. Results of the Dana-Farber Cancer Institute ALL Consortium Protocol 95-01 for children with acute lymphoblastic leukemia. Blood 2007; 109: 896–904.
  • Albertsen BK, Schroder H, Jakobsen P, et al. Antibody formation during intravenous and intramuscular therapy with Erwinia asparaginase. Med Pediatr Oncol 2002; 38: 310–316.
  • Albertsen BK, Schroder H, Jakobsen P, et al. Monitoring of Erwinia asparaginase therapy in childhood ALL in the Nordic countries. Br J Clin Pharmacol 2001; 52: 433–437.
  • Tong WH, Pieters R, Kaspers GJ, et al. A prospective study on drug monitoring of PEGasparaginase and Erwinia asparaginase and asparaginase antibodies in pediatric acute lymphoblastic leukemia. Blood 2014; 123: 2026–2033.
  • Salzer WL, Asselin B, Supko JG, et al. Erwinia asparaginase achieves therapeutic activity after pegaspargase allergy: a report from the Children’s Oncology Group. Blood 2013; 122: 507–514.
  • Plourde PV, Jeha S, Hijiya N, et al. Safety profile of asparaginase Erwinia chrysanthemi in a large compassionate-use trial. Pediatr Blood Cancer 2014; 61: 1232–1238.
  • Hak LJ, Relling MV, Cheng C, et al. Asparaginase pharmacodynamics differ by formulation among children with newly diagnosed acute lymphoblastic leukemia. Leukemia 2004; 18: 1072–1077.
  • Liu C, Kawedia JD, Cheng C, et al. Clinical utility and implications of asparaginase antibodies in acute lymphoblastic leukemia. Leukemia 2012; 26: 2303–2309.
  • Vrooman LM, Stevenson KE, Supko JG, et al. Postinduction dexamethasone and individualized dosing of Escherichia coli L-asparaginase each improve outcome of children and adolescents with newly diagnosed acute lymphoblastic leukemia: results from a randomized study – Dana-Farber Cancer Institute ALL Consortium Protocol 00-01. J Clin Oncol 2013; 31: 1202–1210.
  • Howard SC, Pui CH. Endocrine complications in pediatric patients with acute lymphoblastic leukemia. Blood Rev 2002; 16: 225–243.
  • Raja RA, Schmiegelow K, Albertsen BK, et al. Asparaginase-associated pancreatitis in children with acute lymphoblastic leukaemia in the NOPHO ALL2008 protocol. Br J Haematol 2014; 165: 126–133.
  • Stock W, Douer D, DeAngelo DJ, et al. Prevention and management of asparaginase/pegasparaginase-associated toxicities in adults and older adolescents: recommendations of an expert panel. Leuk Lymphoma 2011; 52: 2237–2253.
  • Payne JH, Vora AJ. Thrombosis and acute lymphoblastic leukaemia. Br J Haematol 2007; 138: 430–445.
  • Truelove E, Fielding AK, Hunt BJ. The coagulopathy and thrombotic risk associated with L-asparaginase treatment in adults with acute lymphoblastic leukaemia. Leukemia 2013; 27: 553–559.
  • Grace RF, Dahlberg SE, Neuberg D, et al. The frequency and management of asparaginase-related thrombosis in paediatric and adult patients with acute lymphoblastic leukaemia treated on Dana-Farber Cancer Institute consortium protocols. Br J Haematol 2011; 152: 452–459.
  • Panis B, Vlaar AM, van Well GT, et al. Posterior reversible encephalopathy syndrome in paediatric leukaemia. Eur J Paediatr Neurol 2010; 14: 539–545.
  • Merryman R, Stevenson KE, Gostic WJ, 2nd, et al. Asparaginase-associated myelosuppression and effects on dosing of other chemotherapeutic agents in childhood acute lymphoblastic leukemia. Pediatr Blood Cancer 2012; 59: 925–927.
  • Tong WH, Pieters R, de Groot-Kruseman HA, et al. Toxicity of very prolonged PEGasparaginase and Erwinia asparaginase courses in relation to asparaginase activity levels with a special focus on dyslipidemia. Haematologica 2014; 99: 1716–1721.
  • Bhojwani D, Darbandi R, Pei D, et al. Severe hypertriglyceridaemia during therapy for childhood acute lymphoblastic leukaemia. Eur J Cancer 2014; 50: 2685–2694.
  • Panosyan EH, Grigoryan RS, Avramis IA, et al. Deamination of glutamine is a prerequisite for optimal asparagine deamination by asparaginases in vivo (CCG-1961). Anticancer Res 2004; 4(2C): 1121–1125.
  • Zalewska-Szewczyk B, Andrzejewski W, Mlynarski W, et al. The anti-asparagines antibodies correlate with L-asparagines activity and may affect clinical outcome of childhood acute lymphoblastic leukemia. Leuk Lymphoma 2007; 48: 931–936.
  • Kawahara Y, Morimoto A, Hayase T, et al. Monitoring of anti-L-asparaginase antibody and L-asparaginase activity levels in a pediatric patient with acute lymphoblastic leukemia and hypersensitivity to native Escherichia coli L-asparaginase during desensitization courses. J Pediatr Hematol Oncol 2014; 36: e91–e93.
  • Burke MJ. How to manage asparaginase hypersensitivity in acute lymphoblastic leukemia. Future Oncol 2014; 10: 2615–2627.
  • Wang B, Relling MV, Storm MC, et al. Evaluation of immunologic crossreaction of antiasparaginase antibodies in acute lymphoblastic leukemia (ALL) and lymphoma patients. Leukemia 2003; 17: 1583–1588.
  • Lenz HJ. Management and preparedness for infusion and hypersensitivity reactions. Oncologist 2007; 12: 601–609.
  • Carpentieri U, Balch MT. Hyperglycemia associated with the therapeutic use of L-asparaginase: possible role of insulin receptors. J Pediatr 1978; 93: 775–778.
  • Olefsky JM, Kimmerling G. Effects of glucocorticoids on carbohydrate metabolism. Am J Med Sci 1976; 271: 202–210.
  • Chan JC, Cockram CS, Critchley JA. Drug-induced disorders of glucose metabolism. Mechanisms and management. Drug Saf 1996; 15: 135–157.
  • Pui CH, Burghen GA, Bowman WP, et al. Risk factors for hyperglycemia in children with leukemia receiving L-asparaginase and prednisone. J Pediatr 1981; 99: 46–50.
  • Lowas SR, Marks D, Malempati S. Prevalence of transient hyperglycemia during induction chemotherapy for pediatric acute lymphoblastic leukemia. Pediatr Blood Cancer 2009; 52: 814–818.
  • Cetin M, Yetgin S, Kara A, et al. Hyperglycemia, ketoacidosis and other complications of L-asparaginase in children with acute lymphoblastic leukemia. J Med 1994; 25: 219–229.
  • Belgaumi AF, Al-Bakrah M, Al-Mahr M, et al. Dexamethasone-associated toxicity during induction chemotherapy for childhood acute lymphoblastic leukemia is augmented by concurrent use of daunomycin. Cancer 2003; 97: 2898–2903.
  • Raja RA, Schmiegelow K, Frandsen TL. Asparaginase-associated pancreatitis in children. Br J Haematol 2012; 159: 18–27.
  • Kearney SL, Dahlberg SE, Levy DE, et al. Clinical course and outcome in children with acute lymphoblastic leukemia and asparaginase-associated pancreatitis. Pediatr Blood Cancer 2009; 53: 162–167.
  • Knoderer HM, Robarge J, Flockhart DA. Predicting asparaginase-associated pancreatitis. Pediatr Blood Cancer 2007; 49: 634–639.
  • Samarasinghe S, Dhir S, Slack J, et al. Incidence and outcome of pancreatitis in children and young adults with acute lymphoblastic leukaemia treated on a contemporary protocol, UKALL 2003. Br J Haematol 2013; 162: 710–713.
  • Treepongkaruna S, Thongpak N, Pakakasama S, et al. Acute pancreatitis in children with acute lymphoblastic leukemia after chemotherapy. J Pediatr Hematol Oncol 2009; 31: 812–815.
  • Hernandez-Espinosa D, Minano A, Martinez C, et al. L-asparaginase-induced antithrombin type I deficiency: implications for conformational diseases. Am J Pathol 2006; 169: 142–153.
  • Mitchell LG, Halton JM, Vegh PA, et al. Effect of disease and chemotherapy on hemostasis in children with acute lymphoid leukemia. Am J Pediatr Hematol Oncol 1994; 16: 120–126.
  • Hongo T, Okada S, Ohzeki T, et al. Low plasma levels of hemostatic proteins during the induction phase in children with acute lymphoblastic leukemia: a retrospective study by the JACLS. Japan Association of Childhood Leukemia Study. Pediatr Int 2002; 44: 293–299.
  • Mitchell L, Hoogendoorn H, Giles AR, et al. Increased endogenous thrombin generation in children with acute lymphoblastic leukemia: risk of thrombotic complications in l’asparaginase-induced antithrombin III deficiency. Blood 1994; 83: 386–391.
  • Andrew M, Brooker L, Mitchell L. Acquired antithrombin III deficiency secondary to asparaginase therapy in childhood acute lymphoblastic leukaemia. Blood Coagul Fibrinolysis 1994; 5(Suppl. 1): S24–S36; discussion S59–S64.
  • Caruso V, Iacoviello L, Di Castelnuovo A, et al. Thrombotic complications in childhood acute lymphoblastic leukemia: a meta-analysis of 17 prospective studies comprising 1752 pediatric patients. Blood 2006; 108: 2216–2222.
  • Nowak-Gottl U, Kenet G, Mitchell LG. Thrombosis in childhood acute lymphoblastic leukaemia: epidemiology, aetiology, diagnosis, prevention and treatment. Best Pract Res Clin Haematol 2009; 22: 103–114.
  • Qureshi A, Mitchell C, Richards S, et al. Asparaginase-related venous thrombosis in UKALL 2003 – re-exposure to asparaginase is feasible and safe. Br J Haematol 2010; 149: 410–413.
  • Nowak-Gottl U, Wermes C, Junker R, et al. Prospective evaluation of the thrombotic risk in children with acute lymphoblastic leukemia carrying the MTHFR TT 677 genotype, the prothrombin G20210A variant, and further prothrombotic risk factors. Blood 1999; 93: 1595–1599.
  • Mauz-Korholz C, Junker R, Gobel U, et al. Prothrombotic risk factors in children with acute lymphoblastic leukemia treated with delayed E. coli asparaginase (COALL-92 and 97 protocols). Thromb Haemost 2000; 83: 840–843.
  • Mitchell LG, Andrew M, Hanna K, et al. A prospective cohort study determining the prevalence of thrombotic events in children with acute lymphoblastic leukemia and a central venous line who are treated with L-asparaginase: results of the Prophylactic Antithrombin Replacement in Kids with Acute Lymphoblastic Leukemia Treated with Asparaginase (PARKAA) Study. Cancer 2003; 97: 508–516.
  • Wermes C, von Depka Prondzinski M, et al. Clinical relevance of genetic risk factors for thrombosis in paediatric oncology patients with central venous catheters. Eur J Pediatr 1999; 158: S143–S146.
  • Elhasid R, Lanir N, Sharon R, et al. Prophylactic therapy with enoxaparin during L asparaginase treatment in children with acute lymphoblastic leukemia. Blood Coagul Fibrin 2001; 12: 367–370.
  • Jaing TH, Lin JL, Lin YP, et al. Hyperammonemic encephalopathy after induction chemotherapy for acute lymphoblastic leukemia. J Pediatr Hematol Oncol 2009; 31: 955–956.
  • Frantzeskaki F, Rizos M, Papathanassiou M, et al. L-asparaginase fatal toxic encephalopathy during consolidation treatment in an adult with acute lymphoblastic leukemia. Am J Case Rep 2013; 14: 311–314.
  • Morris EB, Laningham FH, Sandlund JT, et al. Posterior reversible encephalopathy syndrome in children with cancer. Pediatr Blood Cancer 2007; 48: 152–159.
  • Norman JK, Parke JT, Wilson DA, et al. Reversible posterior leukoencephalopathy syndrome in children undergoing induction therapy for acute lymphoblastic leukemia. Pediatr Blood Cancer 2007; 49: 198–203.
  • D’Angelo P, Farruggia P, Lo Bello A, et al. Reversible posterior leukoencephalopathy syndrome: report of 2 simultaneous cases in children. J Pediatr Hematol Oncol 2006; 28: 177–181.
  • Sudour H, Schmitt C, Contet A, et al. Acute metabolic encephalopathy in two patients treated with asparaginase and ondasetron. Am J Hematol 2011; 86: 323–325.
  • Butterworth RF. Pathophysiology of brain dysfunction in hyperammonemic syndromes: the many faces of glutamine. Mol Genet Metab 2014; 113: 113–117.
  • Jorck C, Kiess W, Weigel JF, et al. Transient hyperammonemia due to L-asparaginase therapy in children with acute lymphoblastic leukemia or non-Hodgkin lymphoma. Pediatr Hematol Oncol 2011; 28: 3–9.
  • D’Onofrio V, Poma F, Enea A, et al. Hyperammonemic coma in a patient with late-onset OTC deficiency. Pediatr Med Chir 2014; 36: 9.
  • Johnston PG, Hardisty RM, Kay HE, et al. Myelosuppressive effect of colaspase (L-asparaginase) in initial treatment of acute lymphoblastic leukaemia. Br Med J 1974; 3: 81–83.
  • Oehlers MJ, Fetawadjieff W, Woodliff HJ. Profound leukopenia following asparaginase treatment in a patient with acute lymphoblastic leukaemia. Med J Aust 1969; 2: 907–909.
  • Harris MB, Shuster JJ, Pullen J, et al. Treatment of children with early pre-B and pre-B acute lymphocytic leukemia with antimetabolite-based intensification regimens: a Pediatric Oncology Group Study. Leukemia 2000; 14: 1570–1576.
  • Salzer WL, Devidas M, Shuster JJ, et al. Intensified PEG-L-asparaginase and antimetabolite-based therapy for treatment of higher risk precursor-B acute lymphoblastic leukemia: a report from the Children’s Oncology Group. J Pediatr Hematol Oncol 2007; 29: 369–375.
  • Cremer P, Lakomek M, Beck W, et al. The effect of L-asparaginase on lipid metabolism during induction chemotherapy of childhood lymphoblastic leukaemia. Eur J Pediatr 1988; 147: 64–67.
  • Steinherz PG. Transient, severe hyperlipidemia in patients with acute lymphoblastic leukemia treated with prednisone and asparaginase. Cancer 1994; 74: 3234–3239.
  • Solano-Paez P, Villegas JA, Colomer I, et al. L-asparaginase and steroids-associated hypertriglyceridemia successfully treated with plasmapheresis in a child with acute lymphoblastic leukemia. J Pediatr Hematol Oncol 2011; 33: e122–e124.
  • Parsons SK, Skapek SX, Neufeld EJ, et al. Asparaginase-associated lipid abnormalities in children with acute lymphoblastic leukemia. Blood 1997; 89: 1886–1895.
  • Salvador C, Meister B, Crazzolara R, et al. Management of hypertriglyceridemia in children with acute lymphoblastic leukemia under persistent therapy with glucocorticoids and L-asparaginase during induction chemotherapy. Pediatr Blood Cancer 2012; 59: 771.
  • Ridola V, Buonuomo PS, Maurizi P, et al. Severe acute hypertriglyceridemia during acute lymphoblastic leukemia induction successfully treated with plasmapheresis. Pediatr Blood Cancer 2008; 50: 378–380.
  • Cohen H, Bielorai B, Harats D, et al. Conservative treatment of L-asparaginase-associated lipid abnormalities in children with acute lymphoblastic leukemia. Pediatr Blood Cancer 2010; 54: 703–706.
  • Berrueco R, Rives S, Lopez-Garcia VS, et al. Very high hypertriglyceridemia induced: is plasmapheresis needed? Pediatr Blood Cancer 2011; 57: 532.
  • Abourbih S, Filion KB, Joseph L, et al. Effect of fibrates on lipid profiles and cardiovascular outcomes: a systematic review. Am J Med 2009; 122: 962. e1–e8.
  • Bostrom B. Successful management of extreme hypertriglyceridemia from pegaspargase with omega-3. Pediatr Blood Cancer 2012; 59: 350.
  • Tong WH, Pieters R, van der Sluis IM. Successful management of extreme hypertriglyceridemia in a child with acute lymphoblastic leukemia by temporarily omitting dexamethasone while continuing asparaginase. Pediatr Blood Cancer 2012; 58: 317–318.
  • Dinndorf PA, Gootenberg J, Cohen MH, et al. FDA drug approval summary: pegaspargase (Oncaspar®) for the first-line treatment of children with acute lymphoblastic leukemia (ALL). Oncologist 2007; 12: 991–998.
  • Kurre HA, Ettinger AG, Veenstra DL, et al. A pharmacoeconomic analysis of pegaspargase versus native Escherichia coli L-asparaginase for the treatment of children with standard-risk, acute lymphoblastic leukemia: the Children’s Cancer Group study (CCG-1962). J Pediatr Hematol Oncol 2002; 24: 175–181.
  • Duval M, Suciu S, Ferster A, et al. Comparison of Escherichia coli-asparaginase with Erwinia-asparaginase in the treatment of childhood lymphoid malignancies: results of a randomized European Organisation for Research and Treatment of Cancer-Children’s Leukemia Group phase 3 trial. Blood 2002; 99: 2734–2739.
  • Dutch Childhood Oncology Group. Treatment study protocol of the Dutch Childhood Oncology Group for children and adolescents (1–19 year) with newly diagnosed acute lymphoblastic leukemia; April 10, 2013. Accessed December 1, 2014 from: https://www.skion.nl/workspace/uploads/Onderzoeksprotocol-ALL11-version-4-1-april-2013.pdf
  • Douer D. Is asparaginase a critical component in the treatment of acute lymphoblastic leukemia? Best Pract Res Clin Haematol 2008; 21: 647–658.
  • Boissel N, Auclerc MF, Lhéritier V, et al. Should adolescents with acute lymphoblastic leukemia be treated as old children or young adults? Comparison of the French FRALLE-93 and LALA-94 trials. J Clin Oncol 2003; 21: 774–780.
  • Stock W, La M, Sanford B, et al. What determines the outcomes for adolescents and young adults with acute lymphoblastic leukemia treated on cooperative group protocols? A comparison of Children’s Cancer Group and Cancer and Leukemia Group B studies. Blood 2008; 112: 1646–1654.
  • de Bont JM, Holt B, Dekker AW, et al. Significant difference in outcome for adolescents with acute lymphoblastic leukemia treated on pediatric vs adult protocols in the Netherlands. Leukemia 2004; 18: 2032–2035.
  • Hallbook H, Gustafsson G, Smedmyr B, et al. Treatment outcome in young adults and children >10 years of age with acute lymphoblastic leukemia in Sweden: a comparison between a pediatric protocol and an adult protocol. Cancer 2006; 107: 1551–1561.
  • Ramanujachar R, Richards S, Hann I, et al. Adolescents with acute lymphoblastic leukaemia: outcome on UK national paediatric (ALL97) and adult (UKALLXII/E2993) trials. Pediatr Blood Cancer 2007; 48: 254–261.
  • Advani AS, Sanford B, Luger S, et al. Frontline-treatment of acute lymphoblastic leukemia (ALL) in older adolescents and young adults (AYA) using a pediatric regimen is feasible: toxicity results of the prospective US intergroup trial C10403 (Alliance) [abstract]. Blood 2013; 122: Abstract 3903.