Abstract
The overall survival rate of patients with T-cell acute lymphoblastic leukemia (T-ALL) is now 90%, although patients with relapsed T-ALL face poor prognosis. The ubiquitin-proteasome system maintains normal protein homeostasis, and aberrations in this pathway are associated with T-ALL. Here we demonstrate the in vitro and in vivo activity of ixazomib, a second-generation orally available, reversible, and selective proteasome inhibitor against pediatric T-ALL cell lines and patient-derived xenografts (PDXs) grown orthotopically in immune-deficient NSG mice. Ixazomib was highly potent in vitro, with IC50 values in the low nanomolar range. As a monotherapy, ixazomib significantly extended mouse event-free survival of five out of eight T-ALL PDXs in vivo.
Keywords: Ixazomib, Patient-derived xenografts, Proteosome inhibitor, Pediatric ALL, Preclinical drug testing
INTRODUCTION
While overall survival rates for pediatric acute lymphoblastic leukemia (ALL) are approximately 90%, patients with relapsed T-cell ALL (T-ALL) have a particularly poor prognosis.1 Therefore, there is an urgent need to develop novel therapies that target relapsed/refractory ALL. The ubiquitin-proteasome system (UPS) maintains normal protein homeostasis and is responsible for a host of cellular processes including intracellular protein degradation, apoptosis, inflammation, and cell cycle control.2 Dysregulation of UPS-controlled processes has been linked to many cancers, rendering the UPS an attractive therapeutic target leading to the development of proteasome inhibitors.3
Clinical validation of proteasome inhibitors has been demonstrated for bortezomib, a first-in-class proteasome inhibitor that obtained US Food and Drug Administration (FDA) approval for the treatment of multiple myeloma (MM) and mantle cell lymphoma in 2003.4,5 Although bortezomib exhibited limited activity against pediatric leukemia as a single agent,6 phase I and II combination studies elicited complete remission (CR) rates of 67–73%.7,8 However, a phase III study of bortezomib added to induction therapy and to the delayed intensification treatment block failed to show benefit for children with newly diagnosed T-ALL.9
The next generation proteasome inhibitor carfilzomib obtained FDA approval for MM in 20125 and exhibited increased specificity, potency, and cellular apoptotic sensitivity compared to bortezomib in preclinical ALL models.10,11 A phase I study investigating the efficacy of carfilzomib in combination with standard chemotherapy in newly-diagnosed adults with ALL elicited a 100% CR rate and minimal residual disease negativity in 80% of patients.10
Ixazomib is a second-generation orally available, reversible and selective inhibitor of the β5 site of the constitutive proteasome, and the β1i and β5i sites of the immunoproteasome.12–14 Preclinical studies demonstrated improved pharmacokinetic, pharmacodynamic, and antitumor properties of ixazomib over bortezomib, including a shorter proteasome-dissociation half-life from red blood cells, which may lead to greater biodistribution.12,15 In 2015, ixazomib received FDA approval for use in combination with lenalidomide and dexamethasone in MM.16 Given that leukemia cells are more susceptible to apoptosis following proteasome inhibition compared to healthy cells,2,17 ixazomib is a promising candidate for preclinical ALL studies. Here we report the activity of ixazomib as a single agent against a panel of pediatric T-ALL patient-derived xenografts (PDXs).
METHODS
In vitro assessment of drug activity
Cytotoxicity assays were performed as described previously.18,19 Briefly, Jurkat cells (American Type Culture Collection, Manassas, VA) were suspended in RPMI media supplemented with 10% fetal calf serum (Biosera, Shanghai, China) while PDX cells were cultured in QBSF-60 medium (Quality Biological Inc., Gaithersberg, MD) supplemented with Flt-3 ligand (20 ng/mL, BioNovus Life Sciences, Cherrybrook, NSW, Australia). Cells were plated according to the optimal seeding density (0.2–4 × 105 cells/well) and PDX cells were incubated for 3 hours to equilibrate (37°C, 5% CO2). Jurkat cells and PDX cells were treated with ixazomib at 10-fold dilutions (1 pM - 10 μM) or vehicle control (at the equivalent DMSO concentration) for 48 h, with the final concentration of DMSO being <0.1%. Viability was determined using the AlamarBlue reduction assay and read on a Victor X3 Multilabel Plate Reader (560 nm excitation and 590 nm emission; PerkinElmer, Waltham, MA). All readings were calculated with 0 h background subtraction and normalized as a percentage of control wells, and the half-maximal inhibitory concentration (IC50) was calculated by interpolation of nonlinear regression curves calculated by GraphPad Prism 9 software. Full characterization of Jurkat cells and T-ALL PDX samples is provided in Table 1 and Supplementary Figure 1.
TABLE 1.
Patient demographics, key molecular lesions and in vivo ixazomib responses of the pediatric ALL PDXs used in the study
| Patient Demographics |
PDX Molecular Characteristics |
Ixazomib Response |
|||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|
| PDX | ALL subtype | Age at diagnosis (years) | Sex | Disease status at biopsy | Structural variations | Copy number variations | Single nucleotide variants, gene (amino acid change, variant allele frequency) | TALL Sorts classification | EFST-C | EFST/C | Median ORM |
|
|
|
|
|||||||||
| ALL-8 | T-ALL | 12.6 | M | Relapse | nd | CDKN 2A (HOM DEL), CDKN 2B (HOM DEL) | FAT2 (R2678*, 0.43); NT5C2 (R367Q, 0.56); FBXW7 (R465C, 0.42); ASXL1 (D863G, 0.47); SMARCA4 (R1189Q, 0.49); IL1B (R75K, 0.47); FBN2 (D1008H, 0.51) | TAL/LMO | 2.8 | 1.24 | PD1 |
| ALL-16 | T-ALL | 10.2 | F | Diag nosis | nd | nd | AURKB (V91M, 0.42); DYM (F573L, 0.44); BCL11B (C429fs, 0.26); ARID1A (1334_1335ins Q, 0.32); NOTCH1 (L1600P, 0.58); RPL10 (R98S, 0.65); PHLPP1 (S323P, 0.39) | NKX2 | 2.9 | 1.17 | PD1 |
| ALL-27 | T-ALL | 8.5 | M | Diag nosis | nd | CDKN 2A (HOM DEL), CDKN 2B (HOM DEL) | ACTB (357_364ISKQ EYDE>M, 0.29); ACTB (E364fs, 0.39); NOTCH1 (L1678P, 0.51); CREBBP (S1934P, 0.52); FBXW7 (R479Q, 0.53); PIK3CD (C416R, 0.23) | TAL/LMO | 1.1 | 1.21 | PD1 |
| ALL-30 | T-ALL | 7.4 | M | Diag nosis | STIL::TAL | CDKN 2A (HOM DEL), CDKN 2B (DEL) | SETD2 (DGRES1788d el, 0.31); FLG (A2307T, 0.27); GLI2 (R1543C, 0.54); EYS (TDG985del, 0.36) | TAL/LMO | −1.0 | 0.91 | PD1 |
| ALL-31 | T-ALL | 10.1 | M | Diag nosis | PTMA::NP M1; LMO1::TR BC1; LMO1::TR BC2 | CDKN 2A (HOM DEL), CDKN 2B (HOM DEL) | NRG2 (A792T, 0.49); MLLT4 (R915H, 0.47) | TAL/LMO | 1.9 | 1.17 | PD1 |
| ALL-32 | T-ALL | 11.1 | M | Relapse | NUP214::A BL1 | CDKN 2A (HOM DEL), CDKN 2B (HOM DEL) | ATF7IP (H115R, 0.48); ABL1 (E255K, 0.37); NOTCH2 (P2087T, 0.42); CTCF (, 0.45); NOTCH1 (L1593P, 0.47); WHSC1 (E1099K, 0.52); MTOR (A461V, 0.59); FGF3 (R145Q, 0.48) | TLX3 | 0.3 | 1.05 | PD1 |
| ETP-1 | ETP | 16 | M | Diag nosis | ATRX::MI R325HG; TP53::SNX 31 | nd | NOTCH1 (S2492*, 0.44); RUNX1 (A122fs, 0.47); NUP214 (A1376fs, 0.39); PHF6 (N147fs, 0.92); FGFR2 (E586Q, 0.43); ZFHX3 (G3511S, 0.51); EZH2 (S695L, 0.97); MED12 (R516H, 1); ZFP36L2 (A348T, 0.48); GNB1 (D76G, 0.51) | Diverse | 3.6 | 1.26 | PD1 |
| ETP-3 | ETP | 14 | M | Diag nosis | ERG::IGJ | CDKN 2A (HOM DEL), CDKN 2B (HOM DEL) | GATA3 (TSTPLW270f s, 0.27); GATA3 (STPLW271fs, 0.32); NOTCH1 (PEQ1582del, 0.34); NOTCH1 (P1582fs, 0.52); NRXN1 (L607W, 0.51); FBN2 (I1248V, 0.49); NOTCH1 (Q1584fs, 0.5); TERT (A67P, 0.34); NOTCH1 (P1582T, 0.57); GATA3 (N286T, 0.69); JAK1 (S703I, 1); SOS1 (R982Q, 0.38); GIGYF2 (R407Q, 0.38); NOTCH4 (T684M, 0.45) | TAL/LMO | 0.4 | 1.03 | PD1 |
DEL, shallow deletion; del, deletion; EFS, event-free survival; ETP, early T-cell precursor; fs, frameshift mutation; HOMDEL, homozygous deletion; ins, insertional mutation; Median ORM, median objective response measure; nd, not detected; T-C, treated-control; T/C, treated/control
nonsense mutation.
Development of PDXs, drug treatments and assessment of in vivo drug activity
All animal experiments were approved by the Animal Care and Ethics Committee of UNSW Sydney (Sydney, Australia). Experiments used continuous PDXs previously established in 20–25 g female non-obese diabetic/severe combined immuno-deficient (NOD.CB17-Prkdcscid/SzJ, NOD/SCID) or NOD/SCID/interleukin-2 receptor γ–negative (NOD.Cg-PrkdcscidIL2rgtm1Wjl/SzJAusb, NSG) mice, as described previously.19–21 Briefly, leukemia cells were inoculated intravenously into 6–8 week-old NOD/SCID or NSG mice (Australian BioResources, Moss Vale, NSW, Australia) and leukemic burden was monitored by enumeration of human CD45+ (%huCD45+) cells versus total CD45+ leukocytes (human plus murine) in the peripheral blood (PB) and tissues, as described previously. 19–21 Details of the in vivo Ixazomib citrate treatment including objective response measures used to assess event-free survival (EFS) is provided in supplementary methods.
RESULTS & DISCUSSION
Previous research using T-ALL and acute myeloid leukemia (AML) cell lines showed that bortezomib and ixazomib preferentially and reversibly inhibited the β5 subunit of the constitutive proteasome, and the equivalent β5i subunit of the immunoproteasome, although the activity of bortezomib was more potent.13 Conversely, ixazomib elicited greater β1 and β1i inhibition, even in bortezomib-resistant cell lines, indicating that the mechanism of action of ixazomib, while similar to bortezomib, is not identical.13 Therefore, we first assessed the efficacy of ixazomib using in vitro cytotoxicity assays against a T-ALL cell line (Jurkat) and two T-ALL PDXs (ALL-30, ALL-31, Table 1). Ixazomib elicited IC50 values in the low nM range (Jurkat, 1.9 nM; ALL-30, 1.6 nM; ALL-31, 2.8 nM; Supplementary Figure 2), demonstrating excellent in vitro potency of ixazomib in cell lines and more clinically relevant T-ALL PDXs.
We previously reported that bortezomib induced regressions in two B- and two T-ALL PDXs and significantly extended mouse event-free survival (EFS).22 The bortezomib data, along with its demonstrated activity in relapsed/refractory pediatric ALL,7,8 provided strong rationale for the testing of additional proteasome inhibitors against T-lineage PDXs. Prior to in vivo efficacy testing, we first evaluated the tolerability of ixazomib in both naïve and leukemia engrafted NSG mice
In the naïve cohort (n=18), ixazomib was generally well tolerated with only one case of toxicity observed at each of the 3 mg/kg and 4.5 mg/kg doses after the second treatment. Although some weight loss was observed throughout the treatment, all remaining mice recovered (Supplementary Figure A.3). Biochemistry (Supplementary Table 1) and hematology (Supplementary Table 2) analyses fell mostly within the normal range and were not suggestive of acute toxicity. In mice engrafted with a T-ALL PDX, ALL-31 (Supplementary Figure B.3 and C.3; n=24), ixazomib was poorly tolerated at the two highest doses, with severe toxicity observed after the first dose (Supplementary Figure D.3). Ixazomib was better tolerated at the two lower doses (1.5 and 3 mg/kg), and no mice were euthanized due to toxicity. Biochemistry and hematology analyses revealed no drug-related perturbations (Supplementary Table 3 and Supplementary Table 4). These findings necessitated a dose reduction of ixazomib to 3 mg/kg for the efficacy study which was below the company recommended maximum tolerated dose (MTD).13
Using this revised dose, the efficacy of ixazomib as a monotherapy was evaluated in vivo against six T-ALL PDXs and two early T-cell precursor ALL PDXs (ETP-ALL; Table 1) which have previously been molecularly annotated.23 Ixazomib significantly delayed disease progression in five out of eight PDXs, with Treated-Control (T-C) values ranging from −1.0 to 3.6 days and T/C values from 0.91 to 1.26 (Figures 1 and 2, Table 1). Each PDX achieved a score of Progressive Disease 1 (PD1) according to stringent objective response measures.23 (Supplementary Table 5).
FIGURE 1.

In vivo activity of ixazomib against pediatric ALL PDXs. Individual mouse %huCD45+ cells in peripheral blood (left panels) and Kaplan–Meier curves for EFS (right panels) for T-ALL PDXs ALL-8, ALL-16, ALL-27, ALL-30. Vehicle controls: gray lines; drug treated: orange lines. In engraftment plots, individual mice are represented by pale colored lines, while the median value for the cohort is shown as a dark line. The dashed horizontal line at 25% indicates the threshold for event. (PD1 = progressive disease 1 as part of the objective response criteria; EFS = event free survival; T-C = the difference in median time-to-event (days) between Treatment and Control groups)
FIGURE 2.

In vivo activity of ixazomib against pediatric ALL PDXs. Individual mouse %huCD45+ cells in peripheral blood (left panels) and Kaplan–Meier curves for EFS (right panels) for T-ALL PDXs ALL-31 and ALL-32 and ETP-ALL PDXs ETP-1 and ETP-3. Vehicle controls: gray lines; drug treated: orange lines. In engraftment plots, individual mice are represented by pale colored lines, while the median value for the cohort is shown as a dark line. The dashed horizontal line at 25% indicates the threshold for event. (PD1 = progressive disease 1 as part of the objective response criteria; EFS = event free survival; T-C = the difference in median time-to-event (days) between Treatment and Control groups)
To date, ixazomib has been evaluated in combination with standard-of-care agents in several hematological malignancies. Ixazomib has been evaluated in multiple phase I/II/III trials for both newly-diagnosed and relapsed/refractory MM, and has FDA approval for use in combination with lenalidomide and dexamethasone for the treatment of patients with MM who have received at least one prior therapy. The combination of ixazomib, cyclophosphamide and dexamethasone elicited high overall response rates in newly-diagnosed MM.24–26 Ixazomib has also been evaluated in combination regimens for patients with AML and ALL, although the contribution of ixazomib to the activity observed for these regimens was not isolated in these clinical trials.27,28 Ixazomib is currently being evaluated in a phase I/II clinical trial in combination with chemotherapy (vincristine, dexamethasone, asparaginase, doxorubicin [VXLD]) in children with relapsed/refractory ALL or lymphoblastic lymphoma (ClinicalTrials.gov Identifier NCT03817320). In adult ALL, ixazomib was evaluated in a Phase I trial in newly-diagnosed patients aged 51–75 who were treated with ixazomib in combination with prednisone, vincristine, and doxorubicin, with Philadelphia chromosome positive patients also treated with dasatinib.28 Ixazomib was well tolerated and, despite the poor outcomes associated with adult-onset ALL, the combination therapy induced an overall remission rate of 79%.28 Combination of bortezomib with VXLD in relapsed/refractory pediatric ALL in a phase I/II clinical trial resulted in an overall response rate of 72.9% with no significant difference observed in 2-year overall survival in B- versus T-lineage patients.29 Additionally, bortezomib in combination with chemotherapy in relapse pediatric ALL exhibited a compete response rate of 68 ± 10% in T-ALL patients.30 However, when bortezomib was added to standard therapy for children with newly diagnosed T-ALL in the Children’s Oncology Group AALL1231 clinical trial, there was no added benefit.9 Therefore, further evaluation of ixazomib in combination with standard of care therapies for T-ALL will require new biological insights to guide patient selection and the selection of agents for combination testing.
In summary, here we show that ixazomib has low nanomolar activity against a T-ALL cell line and T-ALL-PDX cells ex vivo and elicited modest in vivo activity as a monotherapy against pediatric T-ALL PDXs.
Supplementary Material
Highlights:
Ixazomib is an orally available, reversible selective proteasome inhibitor
Ixazomib elicited IC50 values in the low nM range in T-ALL cell models in vitro
Ixazomib treatment of PDX models in vivo resulted in a modest increase in survival
ACKNOWLEDGMENTS
This research was funded by grants from the National Cancer Institute (CA199000, CA199222 and CA263963) and the National Health and Medical Research Council of Australia (NHMRC Fellowship APP1157871 to R.B.L.). Children’s Cancer Institute Australia is affiliated with UNSW Sydney and The Sydney Children’s Hospitals Network.
Footnotes
CONFLICTS OF INTEREST STATEMENT
The authors declare no conflicts of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
REFERENCES
- 1.Teachey DT & O’Connor D How I treat newly diagnosed T-cell acute lymphoblastic leukemia and T-cell lymphoblastic lymphoma in children. Blood 135, 159–166 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Niewerth D, Dingjan I, Cloos J, Jansen G & Kaspers G Proteasome inhibitors in acute leukemia. Expert Rev. Anticancer Ther. 13, 327–337 (2013). [DOI] [PubMed] [Google Scholar]
- 3.Micel LN, Tentler JJ, Smith PG & Eckhardt GS Role of ubiquitin ligases and the proteasome in oncogenesis: novel targets for anticancer therapies. J. Clin. Oncol. 31, 1231–1238 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kane RC, Bross PF, Farrell AT & Pazdur R Velcade: U.S. FDA approval for the treatment of multiple myeloma progressing on prior therapy. Oncologist 8, 508–513 (2003). [DOI] [PubMed] [Google Scholar]
- 5.Fisher RI et al. Multicenter phase II study of bortezomib in patients with relapsed or refractory mantle cell lymphoma. J. Clin. Oncol. 24, 4867–4874 (2006). [DOI] [PubMed] [Google Scholar]
- 6.Horton TM et al. A phase 1 study of the proteasome inhibitor bortezomib in pediatric patients with refractory leukemia: a Children’s Oncology Group study. Clin. Cancer Res. 13, 1516–1522 (2007). [DOI] [PubMed] [Google Scholar]
- 7.Messinger YH et al. Bortezomib with chemotherapy is highly active in advanced B-precursor acute lymphoblastic leukemia: Therapeutic Advances in Childhood Leukemia & Lymphoma (TACL) Study. Blood 120, 285–290 (2012). [DOI] [PubMed] [Google Scholar]
- 8.Messinger Y et al. Phase I study of bortezomib combined with chemotherapy in children with relapsed childhood acute lymphoblastic leukemia (ALL): a report from the therapeutic advances in childhood leukemia (TACL) consortium. Pediatr. Blood Cancer 55, 254–259 (2010). [DOI] [PubMed] [Google Scholar]
- 9.Teachey DT et al. Children’s Oncology Group trial AALL1231: A phase III clinical trial testing bortezomib in newly diagnosed T-cell acute lymphoblastic leukemia and lymphoma. J. Clin. Oncol. 40, 2106–2118 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Jonas BA et al. Phase I study of escalating doses of carfilzomib with HyperCVAD in patients with newly diagnosed acute lymphoblastic leukemia. Am. J. Hematol. 96, E114–E117 (2021). [DOI] [PubMed] [Google Scholar]
- 11.Niewerth D et al. Higher ratio immune versus constitutive proteasome level as novel indicator of sensitivity of pediatric acute leukemia cells to proteasome inhibitors. Haematologica 98, 1896–1904 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lee EC et al. Antitumor activity of the investigational proteasome inhibitor MLN9708 in mouse models of B-cell and plasma cell malignancies. Clin. Cancer Res. 17, 7313–7323 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Roeten MSF et al. Pre-clinical evaluation of the proteasome inhibitor ixazomib against Bortezomib-resistant leukemia cells and primary acute leukemia cells. Cells 10, 665 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Smith DC et al. Phase 1 study of ixazomib, an investigational proteasome inhibitor, in advanced non-hematologic malignancies. Invest. New Drugs 33, 652–663 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kupperman E et al. Evaluation of the proteasome inhibitor MLN9708 in preclinical models of human cancer. Cancer Res. 70, 1970–1980 (2010). [DOI] [PubMed] [Google Scholar]
- 16.Shirley M Ixazomib: First global approval. Drugs 76, 405–411 (2016). [DOI] [PubMed] [Google Scholar]
- 17.Orlowski RZ & Kuhn DJ Proteasome inhibitors in cancer therapy: lessons from the first decade. Clin. Cancer Res. 14, 1649–1657 (2008). [DOI] [PubMed] [Google Scholar]
- 18.Moradi Manesh D et al. AKR1C3 is a biomarker of sensitivity to PR-104 in preclinical models of T-cell acute lymphoblastic leukemia. Blood 126, 1193–1202 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Evans K et al. OBI-3424, a novel AKR1C3-activated prodrug, exhibits potent efficacy against preclinical models of T-ALL. Clin. Cancer Res. 25, 4493–4503 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Liem NLM et al. Characterization of childhood acute lymphoblastic leukemia xenograft models for the preclinical evaluation of new therapies. Blood 103, 3905–3914 (2004). [DOI] [PubMed] [Google Scholar]
- 21.Lock RB et al. The nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse model of childhood acute lymphoblastic leukemia reveals intrinsic differences in biologic characteristics at diagnosis and relapse. Blood 99, 4100–4108 (2002). [DOI] [PubMed] [Google Scholar]
- 22.Houghton PJ et al. Initial testing (stage 1) of the proteasome inhibitor bortezomib by the pediatric preclinical testing program. Pediatr. Blood Cancer 50, 37–45 (2008). [DOI] [PubMed] [Google Scholar]
- 23.Rokita JL et al. Genomic profiling of childhood tumor patient-derived xenograft models to enable rational clinical trial design. Cell Rep. 29, 1675–1689.e9 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Brayer J & Baz R The potential of ixazomib, a second-generation proteasome inhibitor, in the treatment of multiple myeloma. Ther. Adv. Hematol. 8, 209–220 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kumar SK et al. Phase 1/2 trial of ixazomib, cyclophosphamide and dexamethasone in patients with previously untreated symptomatic multiple myeloma. Blood Cancer J. 8, 70 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Dimopoulos MA et al. All-oral ixazomib, cyclophosphamide, and dexamethasone for transplant-ineligible patients with newly diagnosed multiple myeloma. Eur. J. Cancer 106, 89–98 (2019). [DOI] [PubMed] [Google Scholar]
- 27.Advani AS et al. A phase I/II trial of MEC (mitoxantrone, etoposide, cytarabine) in combination with ixazomib for relapsed refractory acute myeloid leukemia. Clin. Cancer Res. 25, 4231–4237 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Amrein P et al. Ixazomib in addition to chemotherapy for the treatment of acute lymphoblastic leukemia in older adults. Leuk. Lymphoma 63, 1428–1435 (2022). [DOI] [PubMed] [Google Scholar]
- 29.Bertaina A et al. The combination of bortezomib with chemotherapy to treat relapsed/refractory acute lymphoblastic leukaemia of childhood. Br. J. Haematol. 176, 629–636 (2017). [DOI] [PubMed] [Google Scholar]
- 30.Horton TM et al. Bortezomib reinduction chemotherapy in high-risk ALL in first relapse: a report from the Children’s Oncology Group. Br. J. Haematol. 186, 274–285 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
