Abstract
Asparagine is a non-essential amino acid since it can either be taken up via the diet or synthesized by asparagine synthetase. Acute lymphoblastic leukemia (ALL) cells do not express asparagine synthetase or express it only minimally, which makes them completely dependent on extracellular asparagine for their growth and survival. This dependency makes ALL cells vulnerable to treatment with L-asparaginase, an enzyme that hydrolyzes asparagine. To date, all clinically approved L-asparaginases have significant L-glutaminase co-activity, associated with non-immune related toxic side effects observed during therapy. Therefore, reduction of L-glutaminase co-activity with concomitant maintenance of its anticancer L-asparaginase effect may effectively improve the tolerability of this unique drug. Previously, we designed a new alternative variant of Erwinia chrysanthemi (ErA; Erwinaze) with decreased L-glutaminase co-activity, while maintaining its L-asparaginase activity, by the introduction of three key mutations around the active site (ErA-TM). However, Erwinaze and our ErA-TM variant have very short half-lives in vivo. Here, we show that the fusion of ErA-TM with an albumin binding domain (ABD)-tag significantly increases its in vivo persistence. In addition, we evaluated the in vivo therapeutic efficacy of ABD-ErA-TM in a B-ALL xenograft model of SUP-B15. Our results show a comparable long-lasting durable antileukemic effect between the standard-of-care pegylated-asparaginase and ABD-ErA-TM L-asparaginase, but with fewer co-glutaminase-related acute side effects. Since the toxic side effects of current L-asparaginases often result in treatment discontinuation in ALL patients, this novel ErA-TM variant with ultra-low L-glutaminase co-activity and long in vivo persistence may have great clinical potential.
Introduction
L- a sparaginases (L-ASNases) are cornerstone drugs in the treatment of acute lymphoblastic leukemia (ALL) and have greatly improved patients’ outcomes over the last 40 years.1,2 Since the introduction of L-ASNases, mortality rates in pediatric ALL patients have drastically decreased, currently reaching overall 5-year survival rates of almost 90%.3 While the depletion of blood asparagine (Asn) represents a critical component of the multiple-drug ALL treatment regimen,3-5 the use of L-ASNase is also associated with a variety of toxic side effects.6-9 A large part of these unwanted effects can be attributed to the secondary L-glutaminase co-activity displayed by all clinically approved bacterial L-ASNases.10 These L-ASNases not only convert Asn into L-aspartate (Asp) and ammonia, but also hydrolyse glutamine (Gln) into L-glutamate (Glu) and ammonia.11-13 For all clinically approved L-ASNases, this L-glutaminase co-activity ranges from 2-10% of their primary L-ASNase activity depending on their microbiological origin.10,14,15 Importantly, Gln is much more abundant than Asn in the blood, with levels that range between 500-800 mM compared to ~50 mM for Asn.16,17 Because Gln levels are much higher, extensive amounts of ammonia are produced which might contribute to L-ASNase-induced neurotoxicity.18-20 The L-glutaminase co-activity also has an impact on protein synthesis in the liver and pancreas resulting in hepatotoxicity, pancreatitis, hyperglycemia, leukopenia and thrombosis.6,14,21
Over the past years, several attempts have been made to develop less toxic, low-glutaminase variants from various biological sources with better enzymatic parameters.10,21,22 Important criteria to keep in mind while designing alternative L-ASNases are efficacy, toxicity, kinetic properties, pharmacokinetics (i.e. in vivo half-life) and immunogenicity.22-24 The active degradation of native L-ASNase enzymes and their short half-life represents one of the biggest problems observed in the L-ASNase research field.25 Currently, the first-line L-ASNase used in the clinic is polyethylene glycol (PEG)-asparaginase, a L-ASNase derived from Escherichia coli (EcA) which is conjugated to a PEG-tag that not only improves the half-life of the enzyme, but also the solubility and masks the bacterial enzyme from the immune system.25-31 However, studies have shown that PEG itself can be immunogenic and that this anti-PEG immune response facilitates clearance in vivo.10,32,33 Considering the bacterial origin of clinically used L-ASNases, immune responses against the enzyme itself are also commonly observed during therapy, including bronchospasms, anaphylactic shock, and hypersensitivity reactions.21,34,35
Currently, debate is ongoing whether the L-glutaminase co-activity of clinically approved L-ASNases is essential, contributory or fully dispensable for their anticancer effect based on contradictory results.10,36-39 Several studies have shown that the cancer cell killing potential of L-glutaminase co-activity has been linked to asparagine synthetase (ASNS) expression levels.10 Chan et al. found that L-glutaminase co-activity is not required for the cell killing potential of L-ASNase against ASNS-negative ALL cancer cells in vitro using a newly designed L-glutaminase-deficient EcA Q59L mutant (EcAQ59L),40 while other groups reported that Gln depletion contributed to the overall anti-leukemic effect.38,40,41 In addition, Panosyan et al. showed that deamination of Gln may enhance the anti-leukemic effect of L-ASNase.42 Recently, Nguyen et al. engineered a variant of Erwinia chrysanthemi (ErA) with three mutations, ErA-TM, that acquired ultra-low L-glutaminase activity while maintaining its L-ASNase activity.15 ErA has a very short in vivo half-life, but the authors reported that the addition of a His-SUMO tag N-terminally to ErA-TM enhanced the drug’s in vivo stability.43 Using short-term His-SUMO-ErA-TM treatment, in vivo antileukemic efficacy was successfully demonstrated in a B-ALL xenograft model of SUP-B15 cells and a patient-derived T-ALL xenograft model,43 confirming the previous in vitro results from Chan et al.40 However, it was recently shown that the EcAQ59L of Chan et al. did not induce a durable in vivo anticancer effect against xenografts of the ASNS-negative SUP-B15 leukemia cell line in NOD/SCID gamma mice (NSG) compared to the L-ASNase wild-type (L-ASNaseWT),44 therefore again contradicting previous reports.40
To clarify whether L-glutaminase co-activity is required for a durable long-term anti-leukemic response, we took advantage of the superior tolerability of our low-glutaminase ErA-TM variant. To further improve the in vivo persistence (even longer than that afforded by the His-SUMO tag43), we introduced an albumin-binding domain (ABD) N-terminal to ErA-TM, which we refer to as ABD-ErA-TM. We show that the introduction of an ABD-tag stabilizes ErA-TM and dramatically enhances its half-life. In addition, we evaluated the in vivo efficacy of this low-glutaminase variant in a SUP-B15 ALL xenograft model. Our results show a comparable long-lasting durable antileukemic effect between the mice treated with the standard-of-care PEG-asparaginase and ABD-ErA-TM, but with fewer co-glutaminase related nonimmunological toxicities. In the future, the implementation of this new L-ASNase variant in the clinic could improve ALL patients’ outcomes and quality of life.
Methods
Expression and purification of ABD-ErA-TM and ABD-ErA-WT, crystal structure of ABD-ErA-TM, surface plasmon resonance and isothermal titration calorimetry
For detailed methodology, see the Online Supplementary Appendix.
Enzyme activity assay
The catalytic activities of the L-ASNase variants were determined using the aspartic acid b-hydroxamate (AHA) method as previously described.45-47 A detailed protocol can be found in the Online Supplementary Materials and Methods.
In vivo ABD-ErA-TM and PEG-asparaginase pharmacokinetics and pharmacodynamics study
The pilot pharmacodynamic study was performed at The University of Texas MD Anderson Cancer Center under protocol ACUF #00001658-RN01 following published methods.44,48 For the other pharmacodynamic and pharmacokinetic study, healthy wild-type C57BL/6J were randomly divided into vehicle phosphate-buffered saline (PBS) (n=5), PEG-asparaginase (n=15) and ABD-ErA-TM (n=15) treatment groups. One single administration/dose of 2,500 IU/kg PEG-asparaginase or ABD-ErA-TM was administered via intraperitoneal injection. For the activity determinations, 50 mL of peripheral blood was collected and samples were processed according to the protocol described by Nguyen et al.43 For the amino acid level determinations, 120 mL of peripheral blood was collected and amino acids were measured as previously described.43 For a more detailed description of the methods used, see the Online Supplementary Appendix.
Single-dose acute toxicity study
Healthy wild-type C57BL/6J (Charles River) mice (9 weeks old; n=18) received one single administration/dose (2,500 IU/kg) of PEG-asparaginase or ABD-ErA-TM via intraperitoneal injection. After 96 hours, blood was collected and hematologic (Vetscan® HM5 Hematology Analyzer; Abaxis) and biochemical analyses (VetScan® VS2 Chemistry Analyzer; Abaxis) were performed. For detailed methodology, see the Online Supplementary Appendix.
In vivo treatment of a SUP-B15 cell line xenograf model with L-ASNases
NSG (#005557; The Jackson Laboratory) female mice (n=15) were intravenously injected at 6 weeks of age with 150 mL PBS containing 5 × 106 luciferase-positive SUP-B15 cells. At regular time points, the bioluminescence was measured using the IVIS Lumina II imaging system (PerkinElmer). After evidence of leukemic cell engraftment in the spleen and/or bone marrow, the mice were randomly divided into vehicle PBS (n=5), PEG-asparaginase (n=5) and ABD-ErA-TM (n=5) treatment groups. PEG-asparaginase was administered every 10 days via intraperitoneal injection at a dose of 2,500 IU/kg (total of 5 administration/doses), while ABD-ErA-T M was given at the same dose every 7 days (total of 7 administration/doses). During the experiment, leukemic burden was evaluated via bioluminescent imaging at regular time points.
The ethical committee on animal welfare at Ghent University Hospital approved all the in vivo mice experiments.
Statistical analysis
Statistical power was calculated before every experiment (80% statistical power, a=0.05). The Shapiro-Wilk test was used to check for normality of the data. The amino acid level data were not normally distributed and therefore non-parametric Mann-Whitney U tests were performed. A two-sided P value <0.05 was considered statistically significant. For the toxicity data, Mann-Whitney U tests, followed by Bonferroni correction for multiple comparisons, were performed. Results are expressed as mean ± standard deviation where appropriate. GraphPad Prism 9 (GraphPad Software Inc., La Jolly, CA, USA) was used to analyze the data.
Results
Introduction of an ABD-tag extends the in vivo persistence of ErA-TM L-ASNase variants
Dennis et al.49 used phage display to select peptides that bind to serum albumins. The 18-residue peptide SA21 with the highest binding affinity to the human serum albumin (467 nM) contains two cysteines speculated to form an intramolecular disulfide bond that would stabilize the binding to albumin. We fused this SA21 sequence, from now on referred to as ABD, to our low-glutaminase ErA-TM variant (Figure 1A). The challenge in the purification was the undesired buildup of inter-ABD disulfide bonds instead of the desired intra-ABD disulfide bonds. The lack of cysteine residues in ErA/ErA-TM suggested that any undesired intermolecular disulfide bridges are between the cysteine of an ABD in a protomer and a cysteine of an ABD in another protomer. In fact, using gel filtration we could see that ~25% of the protein eluted as an octamer instead of the physiological tetramer, presumably due to this incorrect disulfide bond formation between ABD and cysteine residues located in different tetramers (Online Supplementary Figure S1). For the experiments described, only the physiological tetrameric fraction was used.
We first verified if the kcat and Km values of ABD-ErA-T M were comparable to the ones from untagged ErA-TM and our previously reported His-SUMO-ErA-TM variant43 (Table 1). The Km property is key for obtaining complete Asn depletion in vivo. The introduction of the triple mutations to ErA increased the Km from ~50 mM to ~100 mM, and the fusion of ErA-TM to ABD further increased the Km to ~150 mM.
To verify that ABD-ErA-TM can efficiently deplete Asn levels in vivo, we conducted a pilot pharmacodynamic properties study that showed a significant decrease of Asn levels upon ABD-ErA-TM administration (single administration/dose 2,500 IU/kg) (Online Supplementary Figure S2). At the same time, we were encouraged to see a significant increase in half-life compared to that of the HisSUMO-ErA-TM protein43 (38.3 vs. 15.7 hours) (Table 2). Presumably, this increase in half-life is due to the interaction between the ABD moiety and the mouse serum albumin, and that this interaction slows down the degradation of the enzyme. To verify that our molecule binds serum albumins, we used surface plasmon resonance and isothermal titration calorimetry to determine the in vitro interaction with mouse and human serum albumins. Whereas ErA-TM shows no binding of either mouse or human serum albumin, ABD-ErA-TM binds with a low micromolar affinity to both (Table 3; Online Supplementary Figures S3 and S4). To test the prediction that a disulfide bridge between the two ABD cysteines is required for the interaction with albumins, we generated a variant in which these cysteines were mutated to serines (ABD(C→S)-ErA-TM). As expected, this variant exhibited no affinity to either mouse or human serum albumin. Indeed, the in vivo half-life of the ABD(C→S)-ErA-TM variant was essentially identical to that of ErA-TM (2.5 vs. 2.3 hours) (Table 2).
Finally, to obtain a molecular understanding of the ABD, we solved the crystal structure of ABD-ErA-TM at 1.75 Å resolution. Data collection and refinement statistics are shown in Online Supplementary Table S1. In the tetrameric molecule, one protomer had clear electron density for the ABD. Presumably, the ABD of the other three protomers were not seen due to being highly flexible. As shown in Figure 1B, the two cysteines of the ABD form a disulfide bridge that acts to rigidify the loop structure. Thus, for the first time, we have a view of the ABD-conformation that is required for the interaction with serum albumins.
In vivo pharmacokinetic and pharmacodynamic profiling of ABD-ErA-TM in comparison to PEG-asparaginase
To further document the pharmacokinetic profile and pharmacodynamic properties of our ABD-stabilized L-ASNase variant in comparison to those of the standard-of-care, healthy C57BL/6J mice were injected intraperitoneally with 2,500 IU/kg of PEG-asparaginase or ABD-ErA-TM. The L-ASNase activity was measured at days 1, 3, 5, 7, 10 and 14. Although a similar pharmacokinetic profile was seen with the PEG-asparaginase, the activity of ABD-ErA-TM decreased more rapidly compared to that of the PEG-asparaginase (Figure 2A). In addition, we determined the Asn, Asp, Gln, and Glu levels in blood plasma prior to the treatment and at days 1, 3, 5, 7, 10 and 14 after L-ASNase administration. Both enzymes efficiently depleted the blood Asn in a comparable manner (P=0.0079; Mann-Whitney U test), with levels slowly recovering after day 5 for ABD-ErA-TM and after day 7 for PEG-asparaginase (Figure 2B). In line with these results, plasma Asp levels increased significantly at days 1 and 3 (P=0.0079; Mann-Whitney U test) compared to baseline levels and then decreased to the pretreatment level by day 14 for both treatment groups (Online Supplementary Figure S5A). For Gln, only at 24 hours after administration was a minimal but significant reduction in Gln plasma levels seen for PEG-asparaginase, while no decrease of Gln levels was observed in ABD-ErA-TM mice (Online Supplementary Figure S5B). On the other hand, a consistent differential effect was demonstrated on the plasma Glu levels, with a significant increase in the PEG-asparaginase mice up to day 7 after administration, followed by a decrease towards baseline levels again. These increased Glu levels upon PEG-asparaginase treatment point out that long-term L-glutaminase co-activity is maintained. Notably, a minimal increase was observed in the ABD-ErA-TM group but not to the extent as in the PEG-asparaginase mice, as to be expected from this low-glutaminase variant (Online Supplementary Figure S5C).
Toxicity profiling of ABD-ErA-TM
Next, we evaluated the acute toxicity of ABD-ErA-TM versus PEG-asparaginase in healthy wild-type C57BL/6J female mice at a dose of 2,500 IU/kg. We compared the impact of both enzymes on the animals' body weight, one of the main parameters to evaluate L-ASNase toxicity. At the end of the experiment, PEG-asparaginase mice lost a greater percentage of their starting body weight (P=0.0022; Mann-Whitney U test), while the ABD-ErA-TM mice showed no significant decrease in body weight (Figure 2C). In addition, we also analyzed hematologic parameters, including white blood cell count, red blood cell count and blood platelet levels. While no effect was observed on red blood cell counts, significant differences were seen in white blood cell and blood platelet levels (Online Supplementary Figure S6A-C). As previously mentioned, side effects of L-ASNase treatment, in particular liver and pancreas toxicity, are often observed. Blood chemistry analysis showed a significant decrease in globulin and albumin levels, both markers of hepatic dysfunction, in the PEG-asparaginase group compared to the vehicle group (P=0.0366 and P=0.0084, respectively) (Online Supplementary Figure S6D-E). Other blood parameters, including alanine aminotransferase (Online Supplementary Figure S6F), total bilirubin (Online Supplementary Figure S6G), blood urea nitrogen (Online Supplementary Figure S6H) and amylase (Online Supplementary Figure S6I) did not show any significant changes between groups.
In vivo therapeutic evaluation of ABD-ErA-TM in a B-cell acute lymphoblastic leukemia cell line xenograf model
We evaluated the in vivo efficacy of long-term Asn deprivation using ABD-ErA-TM in a luciferase positive SUP-B15 xenograft mouse model. To examine the need for co-glutaminase activity to kill ALL cells, we compared our stabilized low-glutaminase variant ABD-ErA-TM with PEG-asparaginase. Based on the results of our pharmacokinetic and pharmacodynamic studies, a dose of ABD-ErA-TM (2,500 IU/kg) was administered every 7 days, while PEG-asparaginase (2,500 IU/kg) was given every 10 days guaranteeing continuous Asn deprivation in the blood with minimal residual L-ASNase activity building up before each new dose. In total, mice received either five doses of PEG-asparaginase or seven doses of ABD-ErA-TM (Figure 3A). Results not only show that both ABD-ErA-TM and PEG-asparaginase were equally capable of rapidly lowering the leukemic burden, but also that a comparable long-term survival could be accomplished (Figure 3B). During the treatment time course, both PEG-asparaginase and ABD-ErA-TM treated mice displayed lower bioluminescent signals, below the background detection levels at the end of the treatment, while PBS control mice clearly showed an increase in leukemic burden (Figure 3C). At day 55, control mice had to be euthanized due to ethical humane end-points, while both PEG-asparaginase and ABD-ErA-TM mice showed no sign of ALL burden up to day 204, with the exception of one PEG-asparaginase treated mouse that relapsed at day 180 and clearly showed clinical signs of tumor formation.
The L-ASNase activity in blood plasma samples was measured 24 hours after administration of ABD-ErA-TM (at days 1, 8, 15, 22, 36 and 43) and PEG-asparaginase (at days 1, 11, 21 and 41). Results indicate that acceptable stable enzyme activity levels were maintained during the whole experiment (threshold for adequate activity >100 IU/L) without any excessive build-up of L-ASNase activity (Figure 3D). In addition, we also determined the amino acid levels in treated mice to confirm efficacy of Asn depletion during L-ASNase treatment. Both enzymes depleted the blood Asn, which was below detection at every time point (Figure 3E).
Besides the therapeutic effect, the impact and tolerability of both drugs was evaluated on the body weight of the animals. At the end of treatment, we observed a mean weight loss of 8.8% in the ABD-ErA-TM treatment group, while the PEG-asparaginase mice had a mean weight loss of 13.9% (Figure 3F). At the end of this survival experiment (day 204), mice were sacrificed and analyzed for signs of leukemia development. No significant difference between spleen/body weight in both treated groups was observed (Online Supplementary Figure S7A). In addition, analysis of human CD45+ cells in peripheral blood, spleen and bone marrow showed undetectable levels of leukemic cells in both PEG-asparaginase- and ABD-ErA-TM-treated mice, with exception of the PEG-asparaginase mouse that relapsed during survival monitoring (Online Supplementary Figure S7B).
Together, these results convincingly show that the ABD-tag stabilizes this ErA-TM variant in vivo and ABD-ErA-TM can lower ALL leukemic disease burden in a manner comparable to that of the high-glutaminase standard-of-care PEG-asparaginase, but with a better tolerability.
Discussion
Although L-ASNase is a key chemotherapeutic agent in childhood ALL, particularly during the induction phase, its administration is associated with multiple and severe toxicities. Unfortunately, these side effects prevent many patients from receiving their prescribed course of treatment, resulting in a detrimental impact on their outcome.50,51 Several studies showed that the L-glutaminase co-activity of clinically used L-ASNases contributes partially to these unwanted side effects.21 Therefore, lowering L-glutaminase co-activity while maintaining its anticancer effect might provide the required safety profile that reduces treatment interruptions.
Immune reactions during L-ASNase therapy occur frequently together with the development of anti-L-ASNase antibodies.52 Patients suffering from hypersensitivity reactions or silent inactivation to EcA are switched to native ErA.50,52-54 Although the latter is a useful second-line therapy in ALL, some disadvantages are associated with this drug, including a much shorter half-life compared to that of PEG-asparaginase.6,55 As patients can also develop antibodies against the PEG-tag itself, thereby limiting tolerance and efficacy,33,56 additional research on more immunotolerant tags such as XTEN57 and PASylation58,59 is currently ongoing. In our previous study,43 we presented an ErA-TM variant with significantly lower L-glutaminase but comparable L-ASNase activity relative to ErA-WT. We also achieved an extended circulation time by the fusion of a SUMO tag to these molecules. With the goal of further increasing blood circulation time, we analyzed several other tags, and discovered that the fusion of an ABD to ErA (WT or TM variant) resulted in respective molecules with much longer in vivo half-lives. In this study, we present ABD-ErA-TM, a novel ErA derived variant that is superior to our previous ones43 by combining both low L-glutaminase co-activity and an exceptionally extended in vivo persistence.
Acute toxicological evaluation of ABD-ErA-TM revealed reduced non-immunological toxicity compared to the current first-in-line EcA. Our toxicity data show a clear correlation between reduced L-glutaminase co-activity and reduced drug toxicity, especially on the average percentage of body weight loss. We note that other biochemical parameters of liver and pancreas toxicity including alanine aminotransferase, total bilirubin, blood urea nitrogen and amylase levels did not show any changes. This may not be surprising since L-ASNase toxicity in patients is often only observed after multiple L-ASNase administration/doses.
The clinical relevance of L-glutaminase co-activity, present in all clinically Food and Drug Administration (FDA)-approved bacterial L-ASNases, in the context of ALL remains largely unclear. First, it was shown that this L-glutaminase co-activity was not required to induce an in vivo L-ASNase effect in the leukemic SUP-B15 cell line.43 However, Chan et al.44 showed that a durable and long-lasting antileukemic effect in the same SUP-B15 xenograft could only be obtained using a L-ASNase variant that retained its L-glutaminase co-activity.44 Our results clearly contradict these findings and demonstrate that SUP-B15 leukemic cells can be durably killed with a L-ASNase variant with ultra-low glutaminase co-activity. We speculate that the lack of durable response noted by Chan et al.44 was due to insufficient long-term Asn depletion that resulted from using a non-stabilized L-ASNase variant, which means that it needs to be injected more frequently to observe a therapeutic effect. Our results clearly show a similar long-lasting durable anti-leukemic effect between PEG-asparaginase and ABD-ErA-TM.
Industrial production problems recently resulted in a shortage of ErA and reduced treatment options for patients who developed hypersensitivity or silent inactivation against PEG-asparaginase. To overcome these manufacturing issues, a recombinant version of ErA, JZP-458, was recently developed. While this JZP-458 has been approved by the FDA as part of a treatment regimen for children and adults with ALL or lymphoblastic lymphoma,60 our ABD-ErA-TM has multiple benefits over the JZP-458 preparation. One is avoiding the toxicities that arise due to the glutaminase co-activity. Secondly, the in vivo persistence of JZP-458 is short whereas the albumin binding, provided by the ABD domain, increases the half-life >10-fold. This will provide a dramatic clinical advantage, allowing patients to be dosed once a week instead of three times a week as currently required for Erwinaze or JZP-458. In addition, our new Erwinia L-ASNase preparation requires no additional post-translational modifications resulting in high protein yields. As our ABD-ErA-T M would only need to be administered once a week, smaller amounts will be necessary to treat the same number of patients compared to current standard-of-care L-ASNases.
Until now, all immune-related side-effects were ignored in our experimental setup, as the in vivo efficacy study was done with human ALL cells xenotransplanted in immuno-deficient NSG mice. Unfortunately, good preclinical models to evaluate the immunogenicity of L-ASNases are not available and this is one of the biggest challenges for future L-ASNase research.
In conclusion, this study convincingly shows that high L-glutaminase co-activity present in standard-of-care PEG-asparaginase is not essential for durable anti-leukemic properties in vivo, therefore contradicting previous reports. Furthermore, our findings also suggest a decline in in vivo toxicity when L-glutaminase co-activity is reduced. In addition, the fusion of an ABD-tag to our alternative ErA-TM variant increases its half-life and in vivo stability. Identifying a L-ASNase variant with fewer side effects will not only improve the quality of life of ALL patients directly, but will also allow treatment protocols to be maintained or prolonged, which will translate into improved outcomes, especially for adult ALL patients. In addition, a few aggressive solid cancer subtypes with poor prognosis and a clinical unmet need have been identified as sensitive to Asn deprivation, including ovarian and metastatic breast cancer. However, because of the adverse side effects in adults, clinical trials have been discontinued so far. By identifying a L-ASNase variant with fewer unwanted toxicities, we hope to revive the clinical trials and provide extra therapeutic opportunities for these patients.
Footnotes
- Received May 11, 2022
- Accepted August 4, 2022
Correspondence
Disclosures
AMS, AL and HAN declare competing financial interests by being founders with equity stake in Enzyme by Design Inc.: a start-up developing new L-asparaginases. AL and HAN are inventors of the Int. Patent. Appl. PCT/US2017/020090 (WO2017151707A1) that describes the ABD-ErA-TM variant. None of the other authors declare any competing financial interests.
Contributions
MVT, AMS, HAN, TL, SG, PVV and AL designed the study; MVT, AMS, HAN, HL and WC performed experiments; LR and KV provided excellent technical assistance; TL, SG, PVV and AL supervised this study; MVT and AL drafted the manuscript; MVT, AL, HUN and YDV edited the figures; MVT, AMS, YDV, HAN, LR, KV, EP, YS, HL, PLL, WC, VM, BDM, TL, SG, PVV and AL proofread the original draft and made valuable adjustments; AMS, PLL, TL, SG, PVV, and AL obtained funding for this study. All authors have read and agreed to the published version of the manuscript.
Data-sharing statement
The authors confirm that the data supporting the findings of this study are available within the article and its supplementary materials. More detailed data are available from the co-senior authors PVV and AL upon reasonable request.
Funding
This work was supported by The Belgian Foundation Against Cancer (project grants to PVV and SG), Belgian Stand Up to Cancer Foundation (project grants to PVV and SG) and vzw Kinderkankerfonds (grant to TL); and in part by the Department of Veterans Affairs, Veterans Health Administration, Office of Research and Development Bio-medical Laboratory Research and Development MERIT Award I01BX001919 and Chicago Biomedical Consortium Accelerator Award (AL), and NIH SBIR grant R43-CA232798 (AMS). WKC and PLL are supported in part by NIH grants S10OD012304-01 and P30CA016672. This research used resources of the Advanced Photon Source, a U.S. Department of Energy (DOE) Office of Science User Facility operated for the DOE Office of Science by Argonne National Laboratory under Contract No. DE-AC02-06CH11357. Use of the LS-CAT Sector 21 was supported by the Michigan Economic Development Corporation and the Michigan Technology Tri-Corridor (Grant 085P1000817). The views expressed in this article are those of the authors and do not necessarily reflect the position or policy of the Department of Veterans Affairs or the United States government.
Acknowledgments
The authors would like to thank Sri Ramya Donepudi for assistance with assessment of pharmacodynamics. We also thank the Biostatistics Unit of the Faculty of Medicine and Health Sciences of Ghent University for assistance with the statistics.
References
- Radadiya A, Zhu W, Coricello A, Alcaro S, Richards NGJ. Improving the treatment of acute lymphoblastic leukemia. Biochemistry. 2020; 59(35):3193-3200. https://doi.org/10.1021/acs.biochem.0c00354PubMedPubMed CentralGoogle Scholar
- Malard F, Mohty M. Acute lymphoblastic leukaemia. Lancet. 2020; 395(10230):1146-1162. https://doi.org/10.1016/S0140-6736(19)33018-1PubMedGoogle Scholar
- Kumar K, Kaur J, Walia S, Pathak T, Aggarwal D. L-asparaginase: an effective agent in the treatment of acute lymphoblastic leukemia. Leuk Lymphoma. 2014; 55(2):256-262. https://doi.org/10.3109/10428194.2013.803224PubMedGoogle Scholar
- Micallef B, Nisticò R, Sarac SB. The changing landscape of treatment options in childhood acute lymphoblastic leukaemia. Drug Discov Today. 2022; 27(5):1483-1494. https://doi.org/10.1016/j.drudis.2022.01.002PubMedGoogle Scholar
- Endicott M, Jones M, Hull J. Amino acid metabolism as a therapeutic target in cancer: a review. Amino Acids. 2021; 53(8):1169-1179. https://doi.org/10.1007/s00726-021-03052-1PubMedPubMed CentralGoogle Scholar
- Cecconello DK, Magalhães MR, Werlang ICR, Lee MLM, Michalowski MB, Daudt LE. Asparaginase: an old drug with new questions. Hematol Transfus Cell Ther. 2020; 42(3):275-282. https://doi.org/10.1016/j.htct.2019.07.010PubMedPubMed CentralGoogle Scholar
- Hijiya N, van der Sluis IM. Asparaginase-associated toxicity in children with acute lymphoblastic leukemia. Leuk Lymphoma. 2016; 57(4):748-757. https://doi.org/10.3109/10428194.2015.1101098PubMedPubMed CentralGoogle Scholar
- Thu Huynh V, Bergeron S. Asparaginase toxicities: identification and management in patients with acute lymphoblastic leukemia. Clin J Oncol Nurs. 2017; 21(5):E248-E259. https://doi.org/10.1188/17.CJON.E248-E259PubMedGoogle Scholar
- Schmidt MP, Ivanov AV, Coriu D, Miron IC. L-asparaginase toxicity in the treatment of children and adolescents with acute lymphoblastic leukemia. J Clin Med. 2021; 10(19):4419. https://doi.org/10.3390/jcm10194419PubMedPubMed CentralGoogle Scholar
- Fonseca MHG, Fiúza TDS, Morais SB, Souza T, Trevizani R. Circumventing the side effects of L-asparaginase. Biomed Pharmacother. 2021; 139:111616. https://doi.org/10.1016/j.biopha.2021.111616PubMedGoogle Scholar
- Grigoryan RS, Panosyan EH, Seibel NL, Gaynon PS, Avramis IA, Avramis VI. Changes of amino acid serum levels in pediatric patients with higher-risk acute lymphoblastic leukemia (CCG-1961). In Vivo. 2004; 18(2):107-112. Google Scholar
- Ollenschläger G, Roth E, Linkesch W, Jansen S, Simmel A, Mödder B. Asparaginase-induced derangements of glutamine metabolism: the pathogenetic basis for some drug-related side-effects. Eur J Clin Invest. 1988; 18(5):512-516. https://doi.org/10.1111/j.1365-2362.1988.tb01049.xPubMedGoogle Scholar
- Emadi A, Law JY, Strovel ET. Asparaginase Erwinia chrysanthemi effectively depletes plasma glutamine in adult patients with relapsed/refractory acute myeloid leukemia. Cancer Chemother Pharmacol. 2018; 81(1):217-222. https://doi.org/10.1007/s00280-017-3459-6PubMedGoogle Scholar
- Narta UK, Kanwar SS, Azmi W. Pharmacological and clinical evaluation of L-asparaginase in the treatment of leukemia. Crit Rev Oncol Hematol. 2007; 61(3):208-221. https://doi.org/10.1016/j.critrevonc.2006.07.009PubMedGoogle Scholar
- Nguyen HA, Su Y, Lavie A. Design and characterization of Erwinia chrysanthemi l-asparaginase variants with diminished l-glutaminase activity. J Biol Chem. 2016; 291(34):17664-17676. https://doi.org/10.1074/jbc.M116.728485PubMedPubMed CentralGoogle Scholar
- Cruzat V, Macedo Rogero M, Noel Keane K, Curi R, Newsholme P. Glutamine: metabolism and immune function, supplementation and clinical translation. Nutrients. 2018; 10(11):1564. https://doi.org/10.3390/nu10111564PubMedPubMed CentralGoogle Scholar
- Watford M. Glutamine and glutamate: nonessential or essential amino acids?. Anim Nutr. 2015; 1(3):119-122. https://doi.org/10.1016/j.aninu.2015.08.008PubMedPubMed CentralGoogle Scholar
- Tong WH, Pieters R, de Groot-Kruseman HA. The toxicity of very prolonged courses of PEGasparaginase or Erwinia asparaginase in relation to asparaginase activity, with a special focus on dyslipidemia. Haematologica. 2014; 99(11):1716-21. https://doi.org/10.3324/haematol.2014.109413PubMedPubMed CentralGoogle Scholar
- Jaing TH, Lin JL, Lin YP, Yang SH, Lin JJ, Hsia SH. Hyperammonemic encephalopathy after induction chemotherapy for acute lymphoblastic leukemia. J Pediatr Hematol Oncol. 2009; 31(12):955-956. https://doi.org/10.1097/MPH.0b013e3181b8beb1PubMedGoogle Scholar
- Frantzeskaki F, Rizos M, Papathanassiou M. L-asparaginase fatal toxic encephalopathy during consolidation treatment in an adult with acute lymphoblastic leukemia. Am J Case Rep. 2013; 14:311-314. https://doi.org/10.12659/AJCR.889268PubMedPubMed CentralGoogle Scholar
- Van Trimpont M, Peeters E, De Visser Y. Novel insights on the use of L-asparaginase as an efficient and safe anti-cancer therapy. Cancers (Basel). 2022; 14(4):902. https://doi.org/10.3390/cancers14040902PubMedPubMed CentralGoogle Scholar
- Beckett A, Gervais D. What makes a good new therapeutic l-asparaginase?. World J Microbiol Biotechnol. 2019; 35(10):152. https://doi.org/10.1007/s11274-019-2731-9PubMedGoogle Scholar
- Avramis VI, Panosyan EH. Pharmacokinetic/pharmacodynamic relationships of asparaginase formulations: the past, the present and recommendations for the future. Clin Pharmacokinet. 2005; 44(4):367-393. https://doi.org/10.2165/00003088-200544040-00003PubMedGoogle Scholar
- Asselin B, Rizzari C. Asparaginase pharmacokinetics and implications of therapeutic drug monitoring. Leuk Lymphoma. 2015; 56(8):2273-2280. https://doi.org/10.3109/10428194.2014.1003056PubMedPubMed CentralGoogle Scholar
- Brumano LP, da Silva FVS, Costa-Silva TA. Development of L-asparaginase biobetters: current research status and review of the desirable quality profiles. Front Bioeng Biotechnol. 2019; 6:212-212. https://doi.org/10.3389/fbioe.2018.00212PubMedPubMed CentralGoogle Scholar
- Müller HJ, Boos J. Use of l-asparaginase in childhood ALL. Crit Rev Oncol Hematol. 1998; 28(2):97-113. https://doi.org/10.1016/S1040-8428(98)00015-8PubMedGoogle Scholar
- Kawedia JD, Rytting ME. Asparaginase in acute lymphoblastic leukemia. Clin Lymphoma Myeloma Leuk. 2014; 14:Suppl. https://doi.org/10.1016/j.clml.2014.06.017PubMedGoogle Scholar
- Soares AL, Guimarães GM, Polakiewicz B, de Moraes Pitombo RN, Abrahão-Neto J. Effects of polyethylene glycol attachment on physicochemical and biological stability of E. coli L-asparaginase. Int J Pharm. 2002; 237(1-2):163-170. https://doi.org/10.1016/S0378-5173(02)00046-7PubMedGoogle Scholar
- Meneguetti GP, Santos J, Obreque KMT. Novel site-specific PEGylated L-asparaginase. PLoS One. 2019; 14(2):e0211951. https://doi.org/10.1371/journal.pone.0211951PubMedPubMed CentralGoogle Scholar
- Verhoef JJF, Anchordoquy TJ. Questioning the use of PEGylation for drug delivery. Drug Deliv Transl Res. 2013; 3(6):499-503. https://doi.org/10.1007/s13346-013-0176-5PubMedPubMed CentralGoogle Scholar
- Heo YA, Syed YY, Keam SJ. Pegaspargase: a review in acute lymphoblastic leukaemia. Drugs. 2019; 79(7):767-777. https://doi.org/10.1007/s40265-019-01120-1PubMedPubMed CentralGoogle Scholar
- Zhang F, Liu M-R, Wan H-T. Discussion about several potential drawbacks of PEGylated therapeutic proteins. Biol Pharm Bull. 014; 37(3):335-339. https://doi.org/10.1248/bpb.b13-00661PubMedGoogle Scholar
- Kloos R, van der Sluis IM, Mastrobattista E, Hennink W, Pieters R, Verhoef JJ. Acute lymphoblastic leukaemia patients treated with PEGasparaginase develop antibodies to PEG and the succinate linker. Br J Haematol. 2020; 189(3):442-451. https://doi.org/10.1111/bjh.16254PubMedGoogle Scholar
- Krishna M, Nadler SG. Immunogenicity to biotherapeutics - the role of anti-drug immune complexes. Front Immunol. 2016; 7:21. https://doi.org/10.3389/fimmu.2016.00021PubMedPubMed CentralGoogle Scholar
- Mondelaers V, Ferster A, Uyttebroeck A. Prospective, realtime monitoring of pegylated Escherichia coli and Erwinia asparaginase therapy in childhood acute lymphoblastic leukaemia and non-Hodgkin lymphoma in Belgium. Br J Haematol. 2020; 190(1):105-114. https://doi.org/10.1111/bjh.16495PubMedGoogle Scholar
- Chiu M, Tardito S, Pillozzi S. Glutamine depletion by crisantaspase hinders the growth of human hepatocellular carcinoma xenografts. Br J Cancer. 2014; 111(6):1159-1167. https://doi.org/10.1038/bjc.2014.425PubMedPubMed CentralGoogle Scholar
- Long S, Zhang X, Rao Z. Amino acid residues adjacent to the catalytic cavity of tetramer l-asparaginase II contribute significantly to its catalytic efficiency and thermostability. Enzyme and Microbial Technology. 2016; 82:15-22. https://doi.org/10.1016/j.enzmictec.2015.08.009PubMedGoogle Scholar
- Offman MN, Krol M, Patel N. Rational engineering of L-asparaginase reveals importance of dual activity for cancer cell toxicity. Blood. 2011; 117(5):1614-1621. https://doi.org/10.1182/blood-2010-07-298422PubMedGoogle Scholar
- Parmentier JH, Maggi M, Tarasco E, Scotti C, Avramis VI, Mittelman SD. Glutaminase activity determines cytotoxicity of L-asparaginases on most leukemia cell lines. Leuk Res. 2015; 39(7):757-762. https://doi.org/10.1016/j.leukres.2015.04.008PubMedPubMed CentralGoogle Scholar
- Chan WK, Lorenzi PL, Anishkin A. The glutaminase activity of L-asparaginase is not required for anticancer activity against ASNS-negative cells. Blood. 2014; 123(23):3596-3606. https://doi.org/10.1182/blood-2013-10-535112PubMedPubMed CentralGoogle Scholar
- Maggi M, Chiarelli LR, Valentini G, Scotti C. Engineering of Helicobacter pylori L-asparaginase: characterization of two functionally distinct groups of mutants. PLoS One. 2015; 10(2):e0117025. https://doi.org/10.1371/journal.pone.0117025PubMedPubMed CentralGoogle Scholar
- Panosyan EH, Grigoryan RS, Avramis IA. Deamination of glutamine is a prerequisite for optimal asparagine deamination by asparaginases in vivo (CCG-1961). Anticancer Res. 2004; 24(2c):1121-1125. Google Scholar
- Nguyen HA, Su Y, Zhang JY. A novel l-asparaginase with low l-glutaminase coactivity Is highly efficacious against both Tand B-cell acute lymphoblastic leukemias in vivo. Cancer Res. 2018; 78(6):1549-1560. https://doi.org/10.1158/0008-5472.CAN-17-2106PubMedPubMed CentralGoogle Scholar
- Chan WK, Horvath TD, Tan L. Glutaminase activity of L-asparaginase contributes to durable preclinical activity against acute lymphoblastic leukemia. Mol Cancer Ther. 2019; 18(9):1587-1592. https://doi.org/10.1158/1535-7163.MCT-18-1329PubMedPubMed CentralGoogle Scholar
- Fernandez CA, Cai X, Elozory A. High-throughput asparaginase activity assay in serum of children with leukemia. Int J Clin Exp Med. 2013; 6(7):478-487. Google Scholar
- Hejazi M, Piotukh K, Mattow J. Isoaspartyl dipeptidase activity of plant-type asparaginases. Biochem J. 2002; 364(Pt 1):129-136. https://doi.org/10.1042/bj3640129PubMedPubMed CentralGoogle Scholar
- Lanvers-Kaminsky C, Rüffer A, Würthwein G. Therapeutic drug monitoring of asparaginase activity-method comparison of MAAT and AHA test used in the international AIEOP-BFM ALL 2009 trial. Ther Drug Monit. 2018; 40(1):93-102. https://doi.org/10.1097/FTD.0000000000000472PubMedGoogle Scholar
- Horvath TD, Chan WK, Pontikos MA. Assessment of l-asparaginase pharmacodynamics in mouse models of cancer. Metabolites. 2019; 9(1):10. https://doi.org/10.3390/metabo9010010PubMedPubMed CentralGoogle Scholar
- Dennis MS, Zhang M, Meng YG. Albumin binding as a general strategy for improving the pharmacokinetics of proteins. J Biol Chem. 2002; 277(38):35035-35043. https://doi.org/10.1074/jbc.M205854200PubMedGoogle Scholar
- Pieters R, Hunger SP, Boos J. L-asparaginase treatment in acute lymphoblastic leukemia: a focus on Erwinia asparaginase. Cancer. 2011; 117(2):238-249. https://doi.org/10.1002/cncr.25489PubMedPubMed CentralGoogle Scholar
- Gupta S, Wang C, Raetz EA. Impact of asparaginase discontinuation on outcome in childhood acute lymphoblastic Leukemia: a report From the Children's Oncology Group. J Clin Oncol. 2020; 38(17):1897-1905. https://doi.org/10.1200/JCO.19.03024PubMedPubMed CentralGoogle Scholar
- Burke MJ. How to manage asparaginase hypersensitivity in acute lymphoblastic leukemia. Future Oncol. 2014; 10(16):2615-2627. https://doi.org/10.2217/fon.14.138PubMedGoogle Scholar
- Salzer WL, Asselin BL, Plourde PV, Corn T, Hunger SP. Development of asparaginase Erwinia chrysanthemi for the treatment of acute lymphoblastic leukemia. Ann N Y Acad Sci. 2014; 1329:81-92. https://doi.org/10.1111/nyas.12496PubMedGoogle Scholar
- Tong WH, Pieters R, Kaspers GJ. A prospective study on drug monitoring of PEGasparaginase and Erwinia asparaginase and asparaginase antibodies in pediatric acute lymphoblastic leukemia. Blood. 2014; 123(13):2026-2033. https://doi.org/10.1182/blood-2013-10-534347PubMedPubMed CentralGoogle Scholar
- Salzer WL, Asselin B, Supko JG. Erwinia asparaginase achieves therapeutic activity after pegaspargase allergy: a report from the Children's Oncology Group. Blood. 2013; 122(4):507-514. https://doi.org/10.1182/blood-2013-01-480822PubMedPubMed CentralGoogle Scholar
- Armstrong JK, Hempel G, Koling S, Chan LS, Fisher T, Meiselman HJ, Garratty G. Antibody against poly(ethylene glycol) adversely affects PEG-asparaginase therapy in acute lymphoblastic leukemia patients. Cancer. 2007; 110(1):103-111. https://doi.org/10.1002/cncr.22739PubMedGoogle Scholar
- Ding S, Song M, Sim BC. Multivalent antiviral XTEN-peptide conjugates with long in vivo half-life and enhanced solubility. Bioconjug Chem. 2014; 25(7):1351-1359. https://doi.org/10.1021/bc500215mPubMedPubMed CentralGoogle Scholar
- Schlapschy M, Binder U, Börger C. PASylation: a biological alternative to PEGylation for extending the plasma half-life of pharmaceutically active proteins. Protein Eng Des Sel. 2013; 26(8):489-501. https://doi.org/10.1093/protein/gzt023PubMedPubMed CentralGoogle Scholar
- Gebauer M, Skerra A. Prospects of PASylation® for the design of protein and peptide therapeutics with extended half-life and enhanced action. Bioorg Med Chem. 2018; 26(10):2882-2887. https://doi.org/10.1016/j.bmc.2017.09.016PubMedGoogle Scholar
- Maese L, Rizzari C, Coleman R, Power A, van der Sluis I, Rau RE. Can recombinant technology address asparaginase Erwinia chrysanthemi shortages. Pediatr Blood Cancer. 2021; 68(10):e29169. https://doi.org/10.1002/pbc.29169PubMedGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.