Combinatorial efficacy of entospletinib and chemotherapy in patient-derived xenograft models of infant acute lymphoblastic leukemia

Joseph P Loftus, Anella Yahiaoui, Patrick A Brown, Lisa M Niswander, Asen Bagashev, Min Wang, Allyson Schauf, Stacey Tannheimer, Sarah K Tasian, Joseph P Loftus, Anella Yahiaoui, Patrick A Brown, Lisa M Niswander, Asen Bagashev, Min Wang, Allyson Schauf, Stacey Tannheimer, Sarah K Tasian

Abstract

Survival of infants with KMT2A-rearranged (R) acute lymphoblastic leukemia (ALL) remains dismal despite intensive chemotherapy. We observed constitutive phosphorylation of spleen tyrosine kinase (SYK) and associated signaling proteins in infant ALL patient-derived xenograft (PDX) model specimens and hypothesized that the SYK inhibitor entospletinib would inhibit signaling and cell growth in vitro and leukemia proliferation in vivo. We further predicted that combined entospletinib and chemotherapy could augment anti-leukemia effects. Basal kinase signaling activation and HOXA9/MEIS1 expression differed among KMT2A-R (KMT2A-AFF1 [n=4], KMT2A-MLLT3 [n=1], KMT2A-MLLT1 [n=4]) and non-KMT2A-R [n=3] ALL specimens and stratified by genetic subgroup. Incubation of KMT2A-R ALL cells in vitro with entospletinib inhibited methylcellulose colony formation and SYK pathway signaling in a dose-dependent manner. In vivo inhibition of leukemia proliferation with entospletinib monotherapy was observed in RAS-wild-type KMT2A-AFF1, KMT2A-MLLT3, and KMT2A-MLLT1 ALL PDX models with enhanced activity in combination with vincristine chemotherapy in several models. Surprisingly, entospletinib did not decrease leukemia burden in two KMT2A-AFF1 PDX models with NRAS/ or KRAS mutations, suggesting potential RAS-mediated resistance to SYK inhibition. As hypothesized, superior inhibition of ALL proliferation was observed in KMT2A-AFF1 PDX models treated with entospletinib and the MEK inhibitor selumetinib versus vehicle or inhibitor monotherapies (p.

Trial registration: ClinicalTrials.gov NCT00557193.

Figures

Figure 1.
Figure 1.
Constitutive SYK signaling occurs in infant acute lymphoblastic leukemia (ALL). Simple Western analysis of splenic lysates from human ALL patient-derived xenograft (PDX) models demonstrated high basal phosphorylated SYK (pSYK) levels in the majority of infant non-KMT2A-rearranged (R) (light blue) and KMT2A-R (dark blue) ALL specimens. pSYK levels were lower in most childhood non-KMT2A-R ALL specimens (red) and absent in splenic tissue from non-leukemia-injected NSG mice (gray). Total SYK levels were similar across all models. ALL PDX model names are specified above corresponding Simple Western data.
Figure 2.
Figure 2.
Activity and dose optimization of entospletinib monotherapy in KMT2A-R acute lymphoblastic leukemia (ALL). (A) Viably cryopreserved harvested human KMT2A-R ALL cells from murine PDX spleens (model ALL3103 with KMT2A-MLLT3 fusion) demonstrated dose-dependent inhibition of colony formation in vitro in methylcellulose colony assays after ENTO treatment for 14 days. Samples were plated in triplicate in methylcellulose-based medium and grown in 10% leukocyteconditioned medium with 25% FBS and 2% BSA. Data are displayed as mean ± SEM. (B) ALL3103 PDX mice were treated with vehicle (control) or ENTO chow at the specified concentrations for 4 weeks. Human CD45+ CD19+ ALL flow cytometric analysis of murine blood at weekly time points and (C) spleens at study endpoint demonstrated significant inhibition of ALL proliferation with ENTO treatment (mean ± SEM). No difference in ALL burden was observed in 0.03% versus 0.07% ENTOtreated animals. (D) Terminal blood was collected from animals after 4 weeks of continuous ENTO chow consumption and evaluated for entospletinib levels. Data from individual animals are plotted as median interquartile range. ns: not significant by t-test. (E) Terminal spleens from individual mice were harvested, viably cryopreserved, lysed, and evaluated for levels of pSYK, SYK, cMYC, pERK and β-actin by Simple Western. *P<0.05, **P<0.01, ****P<0.0001 as compared to control chow-fed mice by ANOVA with Tukey’s post-test.
Figure 3.
Figure 3.
In vitro activity of entospletinib in KMT2A-R acute lymphoblastic leukemia (ALL). Viably cryopreserved KMT2A-R ALL PDX cells were exposed in vitro to 0.1% DMSO (vehicle control) or increasing concentrations of entospletinib (200 nM, 500 nM, 1 uM) for 2 hours, then lysed and analyzed by Simple Western. Additional untreated (baseline) cells were lysed immediately following sample thaw. (A) Dose-dependent inhibition of the specified phosphoproteins was observed with ENTO in the ALL135MR PDX model (KMT2A-MLLT1, RAS wild-type), while (B) no treatment effect was seen in the 142MR PDX model (KMT2A-AFF1, NRAS-mutant).
Figure 4.
Figure 4.
HOXA9 and MEIS1 expression signatures of KMT2A-R and non-KMT2A-R acute lymphoblastic leukemia (ALL) patient-derived xenograft (PDX) specimens. (A) Splenic PDX samples were analyzed for expression of mRNA for HOXA9 and MEIS1 by NanoString, with human bone marrow mononuclear cells (BMMC) and KG- 1 cell line as negative and positive controls, respectively. Increased MEIS1 and/or HOXA9 expression was seen in KMT2A-R ALL PDX models versus non-KMT2A-R (WT) models and generally clustered by genetic subtype. (B) Total and phosphorylated signal transduction proteins from murine splenic lysates were evaluated using Simple Western. Basal kinase signaling activation differed among KMT2A-R and non-KMT2A-R ALL samples and stratified by genetic subgroup (KMT2A-AFF1, KMT2A-MLLT1, KMT2A-MLLT3, and non-KMT2A-R). β-actin was used as a protein loading control.
Figure 5.
Figure 5.
Entospletinib potently inhibits in vivo acute lymphoblastic leukemia (ALL) proliferation with enhanced efficacy in combination with chemotherapy. Animals engrafted with KMT2A-R (ALL3103, ALL135MR, ALL142MR, ALL150MD, ALL3113) or non-KMT2A-R (ALL132GD, ALL185GD, ALL83GD) ALL were treated with control chow, 0.05% ENTO chow, 0.1 mg/kg vincristine (VCR) IP weekly, or both ENTO and VCR. Human CD45+CD19+ ALL cells were quantified by flow cytometry in end-of-study murine spleens and peripheral blood. (A) Combined ENTO+VCR significantly inhibited leukemia proliferation with enhanced activity compared to ENTO and/or VCR monotherapies in KMT2A-R PDX models without RAS mutations. (B) Conversely, potent VCR effects were observed in KMT2A-R ALL PDX models with NRAS or KRAS mutations without additional activity of combination treatment. (C) A KMT2A-R RAS wild-type ALL PDX model was sensitive to ENTO and not to VCR. (D) No treatment effects of ENTO or VCR were observed in a non-KMT2A-R KRAS-mutant ALL PDX model, while single-agent activity of VCR and/or ENTO and enhanced effects of combination treatment were detected in (E) non-KMT2A-R RAS wild-type PDX control models with other ALL-associated translocations. Data were analyzed by one-way ANOVA with Tukey’s post-test for multiple comparisons. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001.
Figure 6.
Figure 6.
Superior preclinical activity of entospletinib and selumetinib in KMT2A-R acute lymphoblastic leukemia (ALL) patient-derived xenograft (PDX) models. (A) PDX models 142MR (KMT2A-AFF1, NRAS-mutant) and (B) ALL3113 (KMT2A-AFF1, RAS wild-type) were treated with vehicle control chow, 0.05% ENTO chow, 100 mg/kg selumetinib (SEL) via oral gavage twice daily 5 days/week, or both ENTO and SEL for 2 or 4 weeks. Human CD45+ CD19+ ALL cells were quantified by flow cytometry in peripheral blood and end-study murine spleens. Enhanced anti-leukemia efficacy was observed in both models with combined ENTO and SEL treatment versus ENTO or SEL alone, as measured by one-way ANOVA with post Tukey’s post-test for multiple comparisons. *P<0.05, **P<0.01, ****P<0.0001. (C) Ex vivo phosphoflow cytometry analysis of gated human CD19+ CD45+ ALL cells in end-study murine spleens after 2 weeks (ALL142MR) or 4 weeks (ALL3113) of ENTO and/or SEL treatment demonstrate inhibition of pSYK, pERK, and/or pS6 versus vehicle control (gray). ns: not significant, *P<0.05, **P<0.01 by one-way ANOVA with post Tukey’s post-test for multiple comparisons.

References

    1. Pui CH, Yang JJ, Hunger SP, et al. . Childhood acute lymphoblastic leukemia: progress through collaboration. J Clin Oncol. 2015;33(27):2938-2948.
    1. Geng H, Hurtz C, Lenz KB, et al. . Selfenforcing feedback activation between BCL6 and pre-B cell receptor signaling defines a distinct subtype of acute lymphoblastic leukemia. Cancer Cell. 2015;27(3):409-425.
    1. Nguyen K, Devidas M, Cheng SC, et al. . Factors influencing survival after relapse from acute lymphoblastic leukemia: a Children's Oncology Group study. Leukemia. 2008;22(12):2142-2150.
    1. Teachey DT, Hunger SP. Predicting relapse risk in childhood acute lymphoblastic leukaemia. Brit J Haematol. 2013;162(5): 606-620.
    1. Sun W, Malvar J, Sposto R, et al. . Outcome of children with multiply relapsed B-cell acute lymphoblastic leukemia: a therapeutic advances in childhood leukemia and lymphoma study. Leukemia. 2018; 32(11):2316-2325.
    1. Marks DI, Moorman AV, Chilton L, et al. . The clinical characteristics, therapy and outcome of 85 adults with acute lymphoblastic leukemia and t(4;11)(q21;q23)/MLL-AFF1 prospectively treated in the UKALLXII/ECOG2993 trial. Haematologica. 2013;98(6):945-952.
    1. Behm FG, Raimondi SC, Frestedt JL, et al. . Rearrangement of the MLL gene confers a poor prognosis in childhood acute lymphoblastic leukemia, regardless of presenting age. Blood. 1996;87(7):2870-2877.
    1. Winters AC, Bernt KM. MLL-rearranged leukemias-an update on science and clinical approaches. Front Pediatr. 2017;5:4.
    1. Brown P. Treatment of infant leukemias: challenge and promise. Hematology Am Soc Hematol Educ Program. 2013; 2013:596-600.
    1. Pieters R, Schrappe M, De Lorenzo P, et al. . A treatment protocol for infants younger than 1 year with acute lymphoblastic leukaemia (Interfant-99): an observational study and a multicentre randomised trial. Lancet. 2007;370(9583):240-250.
    1. Driessen EMC, de Lorenzo P, Campbell M, et al. . Outcome of relapsed infant acute lymphoblastic leukemia treated on the interfant-99 protocol. Leukemia. 2017; 31(12):2854.
    1. Brown P, Pieters R, Biondi A. How I treat infant leukemia. Blood. 2019;133(3):205-214.
    1. Vrooman LM, Blonquist TM, Harris MH, et al. . Refining risk classification in childhood B acute lymphoblastic leukemia: results of DFCI ALL Consortium Protocol 05-001. Blood Advances. 2018;2(12):1449-1458.
    1. Lafage-Pochitaloff M, Baranger L, Hunault M, et al. . Impact of cytogenetic abnormalities in adults with Ph-negative B-cell precursor acute lymphoblastic leukemia. Blood. 2017;130(16):1832-1844.
    1. Iacobucci I, Li Y, Roberts KG, et al. . Truncating erythropoietin receptor rearrangements in acute lymphoblastic leukemia. Cancer Cell. 2016;29(2):186-200.
    1. Tkachuk DC, Kohler S, Cleary ML. Involvement of a homolog of Drosophila trithorax by 11q23 chromosomal translocations in acute leukemias. Cell. 1992;71(4):691-700.
    1. Gu Y, Nakamura T, Alder H, et al. . The t(4;11) chromosome translocation of human acute leukemias fuses the ALL-1 gene, related to Drosophila trithorax, to the AF-4 gene. Cell. 1992;71(4):701-708.
    1. Zangrando A, Dell'Orto MC, te Kronnie G, Basso G. MLL rearrangements in pediatric acute lymphoblastic and myeloblastic leukemias: MLL specific and lineage specific signatures. BMC Med Genomics. 2009;2(1):36.
    1. de Boer J, Walf-Vorderwulbecke V, Williams O. In focus: MLL-rearranged leukemia. Leukemia. 2013;27(6):1224-1228.
    1. Slany RK. MLL fusion proteins and transcriptional control. Biochim Biophys Acta Gene Regul Mech. 2020;1863(3):194503.
    1. Lin S, Luo RT, Ptasinska A, et al. . Instructive role of MLL-fusion proteins revealed by a model of t(4;11) pro-B acute Lymphoblastic Leukemia. Cancer Cell. 2016;30(5):737-749.
    1. Krivtsov AV, Armstrong SA. MLL translocations, histone modifications and leukaemia stem-cell development. Nat Rev Cancer. 2007;7(11):823-833.
    1. Meyer C, Burmeister T, Groger D, et al. . The MLL recombinome of acute leukemias in 2017. Leukemia. 2018;32(2):273-284.
    1. Andersson AK, Ma J, Wang J, et al. . The landscape of somatic mutations in infant MLL-rearranged acute lymphoblastic leukemias. Nat Genet. 2015;47(4):330-337.
    1. Mohr S, Doebele C, Comoglio F, et al. . Hoxa9 and Meis1 cooperatively induce addiction to Syk signaling by suppressing miR-146a in acute myeloid leukemia. Cancer Cell. 2017;31(4):549-562.e11.
    1. Perova T, Grandal I, Nutter LM, et al. . Therapeutic potential of spleen tyrosine kinase inhibition for treating high-risk precursor B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6(236):236ra62.
    1. Mócsai A, Ruland J, Tybulewicz VLJ. The SYK tyrosine kinase: a crucial player in diverse biological functions. Nat Rev Immunol. 2010;10:387.
    1. Efremov DG, Laurenti L. The Syk kinase as a therapeutic target in leukemia and lymphoma. Expert Opin Investig Drugs. 2011;20(5):623-636.
    1. Sharman J, Di Paolo J. Targeting B-cell receptor signaling kinases in chronic lymphocytic leukemia: the promise of entospletinib. Ther Adv Hematol. 2016;7(3):157-170.
    1. Kohrer S, Havranek O, Seyfried F, et al. . Pre- BCR signaling in precursor B-cell acute lymphoblastic leukemia regulates PI3K/AKT, FOXO1 and MYC, and can be targeted by SYK inhibition. Leukemia. 2016;30(6):1246-1254.
    1. Loftus JP, Yahiaoui A, Shen F, et al. . Enhanced efficacy of the SYK inhibitor entospletinib and vincristine in KMT2Arearranged acute lymphoblastic leukemia. EHA Annual Congress. 2018:abstract PF164.
    1. Currie KS, Kropf JE, Lee T, et al. . Discovery of GS-9973, a selective and orally efficacious inhibitor of spleen tyrosine kinase. J Med Chem. 2014;57(9):3856-3873.
    1. Walker AR, Byrd JC, Bhatnagar B, et al. . Results of a phase 1b/2 study of entospletinib (GS-9973) monotherapy and in combination with induction chemotherapy in newly diagnosed patients with acute myeloid leukemia. EHA Annual Congress. 2018:abstract S118.
    1. Salzer WL, Jones TL, Devidas M, et al. . Decreased induction morbidity and mortality following modification to induction therapy in infants with acute lymphoblastic leukemia enrolled on AALL0631: a report from the Children's Oncology Group. Pediatr Blood Cancer. 2015; 62(3):414-418.
    1. Maude SL, Tasian SK, Vincent T, et al. . Targeting JAK1/2 and mTOR in murine xenograft models of Ph-like acute lymphoblastic leukemia. Blood. 2012; 120(17):3510-3518.
    1. Maude SL, Dolai S, Delgado-Martin C, et al. . Efficacy of JAK/STAT pathway inhibition in murine xenograft models of early Tcell precursor (ETP) acute lymphoblastic leukemia. Blood. 2015;125(11):1759-1767.
    1. Tasian SK, Teachey DT, Li Y, et al. . Potent efficacy of combined PI3K/mTOR and JAK or ABL inhibition in murine xenograft models of Ph-like acute lymphoblastic leukemia. Blood. 2017;129(2):177-187.
    1. Tasian SK, Hurtz C, Wertheim GB, et al. . High incidence of Philadelphia chromosome- like acute lymphoblastic leukemia in older adults with B-ALL. Leukemia. 2017;31(4):981-984.
    1. Ding YY, Stern JW, Jubelirer TF, et al. . Clinical efficacy of ruxolitinib and chemotherapy in a child with Philadelphia chromosome-like acute lymphoblastic leukemia with GOLGA5-JAK2 fusion and induction failure. Haematologica. 2018:103(9):e427-e431.
    1. Irving J, Matheson E, Minto L, et al. . Ras pathway mutations are prevalent in relapsed childhood acute lymphoblastic leukemia and confer sensitivity to MEK inhibition. Blood. 2014;124(23):3420-3430.
    1. Walker AR, Byrd JC, Blachly JS, et al. . Entospletinib in combination with induction chemotherapy in previously untreated acute myeloid leukemia: response and predictive significance of HOXA9 and MEIS1 expression. Clin Cancer Res. 2020; 26(22):5852-5859.
    1. Kohrer S, Havranek O, Seyfried F, et al. . Pre- BCR signaling in precursor B-cell acute lymphoblastic leukemia regulates PI3K/AKT, FOXO1 and MYC, and can be targeted by SYK inhibition. Leukemia. 2016;30(6):1246-1254.
    1. van der Veer A, van der Velden VHJ, Willemse ME, et al. . Interference with pre- B-cell receptor signaling offers a therapeutic option for TCF3-rearranged childhood acute lymphoblastic leukemia. Blood Cancer J. 2014;4(2):e181.
    1. Hurtz C, Tasian SK, Wertheim GB, et al. . Redundant JAK, SRC and PI3 kinase signaling pathways regulate cell survival in human Ph-like ALL cell lines and primary cells. Blood. 2017;130(Suppl 1):717.
    1. Hurtz C, Wertheim GB, Loftus JP, et al. . Oncogene-independent adaptation of pre-B cell receptor signaling confers drug resistance and signaling plasticity in Ph-like ALL. Blood. 2019;134(S1):747.
    1. Matheson EC, Thomas H, Case M, et al. . Glucocorticoids and selumetinib are highly synergistic in RAS pathway mutated childhood acute lymphoblastic leukemia through upregulation of BIM. Haematologica. 2019;104(9):1804-1811.
    1. Agraz-Doblas A, Bueno C, Bashford- Rogers R, et al. . Unraveling the cellular origin and clinical prognostic markers of infant B-cell acute lymphoblastic leukemia using genome-wide analysis. Haematologica. 2019;104(6):1176-1188.
    1. Marschalek R. Another piece of the puzzle added to understand t(4;11) leukemia better. Haematologica. 2019;104(6):1098-1100.
    1. Bueno C, Calero-Nieto FJ, Wang X, et al. . Enhanced hemato-endothelial specification during human embryonic differentiation through developmental cooperation between AF4-MLL and MLL-AF4 fusions. Haematologica. 2019;104(6):1189-1201.
    1. Levis M, Brown P, Smith BD, et al. . Plasma inhibitory activity (PIA): a pharmacodynamic assay reveals insights into the basis for cytotoxic response to FLT3 inhibitors. Blood. 2006;108(10):3477-3483.
    1. Brown PA, Kairalla J, Hilden JM, et al. . FLT3 inhibitor correlative laboratory assays impact outcomes in KMT2A-rearranged infant acute lymphoblastic leukemia (ALL) patients treated with lestaurtinib: AALL0631, a Children's Oncology Group Study. Blood. 2019;134(S1):1293.
    1. Cooper TM, Cassar J, Eckroth E, et al. . A phase I study of quizartinib combined with chemotherapy in relapsed childhood leukemia: a therapeutic advances in Childhood Leukemia and Lymphoma (TACL) Study. Clin Cancer Res. 2016;22(16):4014-4022.
    1. Bernt KM, Zhu N, Sinha AU, et al. . MLLrearranged leukemia is dependent on aberrant H3K79 methylation by DOT1L. Cancer Cell. 2011;20(1):66-78.
    1. Daigle SR, Olhava EJ, Therkelsen CA, et al. . Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell. 2011;20(1):53-65.
    1. Shukla N, Wetmore C, O'Brien MM, et al. . Final report of phase 1 study of the DOT1L inhibitor, pinometostat (EPZ-5676), in children with relapsed or refractory MLL-r acute leukemia. Blood. 2016;128(22):2780.
    1. von Stackelberg A, Locatelli F, Zugmaier G, et al. . Phase I/phase II study of blinatumomab in pediatric patients with relapsed/refractory acute lymphoblasticl eukemia. J Clin Oncol. 2016;34(36):4381-4389.
    1. Driessen EM, van Roon EH, Spijkers-Hagelstein JA, et al. . Frequencies and prognostic impact of RAS mutations in MLLrearranged acute lymphoblastic leukemia in infants. Haematologica. 2013;98(6):937-944.
    1. Prelle C, Bursen A, Dingermann T, Marschalek R. Secondary mutations in t(4;11) leukemia patients. Leukemia. 2013;27(6):1425-1427.
    1. Kerstjens M, Driessen EM, Willekes M, et al. . MEK inhibition is a promising therapeutic strategy for MLL-rearranged infant acute lymphoblastic leukemia patients carrying RAS mutations. Oncotarget. 2017; 8(9):14835-14846.
    1. Cremer A, Ellegast JM, Alexe G, et al. . Resistance mechanisms to SYK inhibition in acute myeloid leukemia. Cancer Discov. 2020; 10(2):214-231.

Source: PubMed

3
Iratkozz fel