JAK/STAT pathway inhibition overcomes IL7-induced glucocorticoid resistance in a subset of human T-cell acute lymphoblastic leukemias

C Delgado-Martin, L K Meyer, B J Huang, K A Shimano, M S Zinter, J V Nguyen, G A Smith, J Taunton, S S Winter, J R Roderick, M A Kelliher, T M Horton, B L Wood, D T Teachey, M L Hermiston, C Delgado-Martin, L K Meyer, B J Huang, K A Shimano, M S Zinter, J V Nguyen, G A Smith, J Taunton, S S Winter, J R Roderick, M A Kelliher, T M Horton, B L Wood, D T Teachey, M L Hermiston

Abstract

While outcomes for children with T-cell acute lymphoblastic leukemia (T-ALL) have improved dramatically, survival rates for patients with relapsed/refractory disease remain dismal. Prior studies indicate that glucocorticoid (GC) resistance is more common than resistance to other chemotherapies at relapse. In addition, failure to clear peripheral blasts during a prednisone prophase correlates with an elevated risk of relapse in newly diagnosed patients. Here we show that intrinsic GC resistance is present at diagnosis in early thymic precursor (ETP) T-ALLs as well as in a subset of non-ETP T-ALLs. GC-resistant non-ETP T-ALLs are characterized by strong induction of JAK/STAT signaling in response to interleukin-7 (IL7) stimulation. Removing IL7 or inhibiting JAK/STAT signaling sensitizes these T-ALLs, and a subset of ETP T-ALLs, to GCs. The combination of the GC dexamethasone and the JAK1/2 inhibitor ruxolitinib altered the balance between pro- and anti-apoptotic factors in samples with IL7-dependent GC resistance, but not in samples with IL7-independent GC resistance. Together, these data suggest that the addition of ruxolitinib or other inhibitors of IL7 receptor/JAK/STAT signaling may enhance the efficacy of GCs in a biologically defined subset of T-ALL.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
ETP T-ALL samples are intrinsically more resistant to GCs than are non-ETP T-ALLs. PDX samples were thawed, rested 1 h, and then exposed to vehicle or dexamethasone (Dex). Cells were stained with Hoechst dye at 48 h and subjected to flow cytometry. (a) Percentage of viable cells, relative to vehicle, of non-ETP T-ALL (circles, n=22) and ETP T-ALL (squares, n=10) PDX samples after 48 h treatment with 2.5 μm dexamethasone. Data were analyzed using the parametric unpaired t-test with Welch’s correction (unequal variances). ***P<0.001. (b) Percentage of viable cells, relative to vehicle, of representative samples of ETP T-ALLs (n=8) and non-ETP T-ALLs (n=10) exposed to increasing doses of dexamethasone (0, 100, 500 nm and 2.5 μm) for 48 h. Data are represented as mean±s.e.m. of the biological replicates. Error bars show s.e.m.
Figure 2
Figure 2
IL7 deprivation sensitizes a subset of T-ALL samples to dexamethasone-induced cell death. (a) Percentage of viable cells, relative to vehicle, after 48 h of treatment with 2.5 μm dexamethasone in the presence (filled shapes) or absence (empty shapes) of 25 ng/ml IL7. Dashed line indicates 75% relative viability. Non-ETP T-ALL (n=22), ETP T-ALL (n=10). (b) Non-ETP and (c) ETP PDX samples were subjected to 48 h treatment with 2.5 μm dexamethasone, 125 ng/ml AraC, 500 nM etoposide (etop), 250 nM vorinostat (vor), 2.5 μM methotrexate (MTX), or 5 μm vincristine (VCR) in the presence (gray) or absence (white) of 25 ng/ml IL7 and processed as above (n=4 to 22). Paired T-test (Wilcoxon—non normal distribution—in the case of ETPs) was used to evaluate statistical significance. *P<0.05; **P<0.01; ***P<0.001. Data are represented as mean±s.e.m. of the biological replicates. Error bars show s.e.m.
Figure 3
Figure 3
IL7 responsiveness distinguishes non-ETP T-ALL samples and correlates with GC sensitivity. (a) PDX samples were thawed, rested 1 h, and then exposed to vehicle or 100 ng/ml IL7 for 15 min. STAT5 phosphorylation (pSTAT5) was analyzed by phosphoflow cytometry. Non-ETPs were placed into subsets based upon their IL7 responsiveness. Top depicts representative histograms of pSTAT5 in the basal state (shaded) and after 15 min of stimulation with 100 ng/ml IL7 for each subset (empty line). Bottom shows quantitation of pSTAT5 median fluorescence intensity (MFI) in the basal state and in response to IL7 exposure for each sample. (b) PDX samples were exposed to vehicle or 2.5 μm dexamethasone for 48 h in the presence (black circles) or absence (gray squares) of 25 ng/ml of IL7. Percentage of viable cells, relative to vehicle, for each of the subgroups (Responders, n=10, partial responders, n=7, non-responders, n=5, and ETPs, n=6) was measured by quantification of the Hoechst-negative fraction using flow cytometry. Paired T test (Wilcoxon) was used to evaluate statistical significance. *P<0.05, **P<0.01. Data are represented as mean±s.e.m. of the biological replicates. Error bars show s.e.m.
Figure 4
Figure 4
Ruxolitinib blocks IL7R signaling and abrogates the protective effect of IL7 on GC activity. (a) pSTAT5 of a representative ETP T-ALL PDX sample incubated with increasing doses (67.5, 125, 250, 500 and 1000 nm) of ruxolitinib (rux) and then stimulated with 100 ng/ml of IL7 for 15 min. (b) Representative histogram of pSTAT5 after 48 h of culture with vehicle (tinted line) or 500 nm ruxolitinib (empty line) in the presence of 25 ng/ml IL7. (c) PDX samples (Responder n=10, Partial n=5, non-responder n=4, ETP n=6) were subjected to 48 h treatment with vehicle, 2.5 μm dexamethasone, 500 nm ruxolitinib or the combination of dexamethasone and ruxolitinib in the presence of 25 ng/ml IL7. Percentage of viable (Hoechst negative) cells relative to vehicle was quantified for each subset. Data are represented as mean±s.e.m. of the biological replicates. Error bars show s.e.m.
Figure 5
Figure 5
Treatment with ruxolitinib and dexamethasone alters the balance between BCL2 and BIM. Representative PDX samples that either need IL7 to survive GC-induced death (‘IL7-dependent’, n=5) or that were GC-resistant (remained >75% viable) even in the absence of IL7 (‘IL7-independent’, n=4), were treated for 48 h with vehicle, 2.5 μm dexamethasone, 500 nm ruxolitinib or the combination of both drugs in IL7-containing media. Expression of BCL2 (a) and BIM (b) was determined for cells in the viable, active caspase-3 negative gate. One sample t-test was used to evaluate statistical significance. *P<0.05; **P<0.01. Data are represented as mean±s.e.m. of the biological replicates. Error bars show s.e.m.
Figure 6
Figure 6
IL7R signaling prevents a dexamethasone-induced increase in apoptotic priming. (a) Apoptotic priming of IL7-dependent (n=3) and -independent (n=3) samples treated for 16 h with vehicle, 1 μm dexamethasone, 500 nm ruxolitinib, or a combination of the two in IL7-containing media, as assessed by depletion of intracellular cytochrome c following a 1-h exposure to 100 nm BIM BH3 peptide. % priming values are normalized to the vehicle control condition. (b) Apoptotic priming of IL7-dependent and -independent samples following a 1-h exposure to 1 μm ABT-199. % priming values are normalized to the vehicle control condition. (c) Cell viability of IL7-dependent and -independent samples with the addition of 2.5 μm dexamethasone relative to vehicle, 500 nm ruxolitinib, or 200 nm ABT-199 alone. Data are represented as mean±s.e.m. of the biological replicates. Error bars show s.e.m.

References

    1. Graux C, Cools J, Michaux L, Vandenberghe P, Hagemeijer A. Cytogenetics and molecular genetics of T-cell acute lymphoblastic leukemia: from thymocyte to lymphoblast. Leukemia 2006; 20: 1496–1510.
    1. Hunger SP, Lu X, Devidas M, Camitta BM, Gaynon PS, Winick NJ et al. Improved survival for children and adolescents with acute lymphoblastic leukemia between 1990 and 2005: a report from the children’s oncology group. J Clin Oncol 2012; 30: 1663–1669.
    1. Bhojwani D, Pui C-H. Relapsed childhood acute lymphoblastic leukaemia. Lancet Oncol 2013; 14: e205–e217.
    1. Coustan-Smith E, Mullighan CG, Onciu M, Behm FG, Raimondi SC, Pei D et al. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia. Lancet Oncol 2009; 10: 147–156.
    1. Dördelmann M, Reiter A, Borkhardt A, Ludwig WD, Götz N, Viehmann S et al. Prednisone response is the strongest predictor of treatment outcome in infant acute lymphoblastic leukemia. Blood 1999; 94: 1209–1217.
    1. Riehm H, Reiter A, Schrappe M, Berthold F, Dopfer R, Gerein V et al. [Corticosteroid-dependent reduction of leukocyte count in blood as a prognostic factor in acute lymphoblastic leukemia in childhood (therapy study ALL-BFM 83)]. Klin Padiatr 1987; 199: 151–160.
    1. Kaspers GJL, Wijnands JJM, Hartmann R, Huismans L, Loonen AH, Stackelberg A et al. Immunophenotypic cell lineage and in vitro cellular drug resistance in childhood relapsed acute lymphoblastic leukaemia. Eur J Cancer 2005; 41: 1300–1303.
    1. Klumper E, Pieters R, Veerman AJ, Huismans DR, Loonen AH, Hählen K et al. In vitro cellular drug resistance in children with relapsed/refractory acute lymphoblastic leukemia. Blood 1995; 86: 3861–3868.
    1. Yamamoto KR. Steroid receptor regulated transcription of specific genes and gene networks. Annu Rev Genet 1985; 19: 209–252.
    1. Schmidt S, Rainer J, Ploner C, Presul E, Riml S, Kofler R. Glucocorticoid-induced apoptosis and glucocorticoid resistance: molecular mechanisms and clinical relevance. Cell Death Differ 2004; 11 (Suppl 1): S45–555.
    1. Tissing WJE, Meijerink JPP, den Boer ML, Brinkhof B, van Rossum EFC, van Wering ER et al. Genetic variations in the glucocorticoid receptor gene are not related to glucocorticoid resistance in childhood acute lymphoblastic leukemia. Clin Cancer Res 2005; 11: 6050–6056.
    1. Irving JAE, Minto L, Bailey S, Hall AG. Loss of heterozygosity and somatic mutations of the glucocorticoid receptor gene are rarely found at relapse in pediatric acute lymphoblastic leukemia but may occur in a subpopulation early in the disease course. Cancer Res 2005; 65: 9712–9718.
    1. Piovan E, Yu J, Tosello V, Herranz D, Ambesi-Impiombato A, Da Silva AC et al. Direct reversal of glucocorticoid resistance by AKT inhibition in acute lymphoblastic leukemia. Cancer Cell 2013; 24: 766–776.
    1. Marke R, Havinga J, Cloos J, Demkes M, Poelmans G, Yuniati L et al. Tumor suppressor IKZF1 mediates glucocorticoid resistance in B-cell precursor acute lymphoblastic leukemia. Leukemia 2016; 30: 1599–1603.
    1. Wuchter C, Ruppert V, Schrappe M, Dörken B, Ludwig W-D, Karawajew L. In vitro susceptibility to dexamethasone- and doxorubicin-induced apoptotic cell death in context of maturation stage, responsiveness to interleukin 7, and early cytoreduction in vivo in childhood T-cell acute lymphoblastic leukemia. Blood 2002; 99: 4109–4115.
    1. Maude SL, Dolai S, Delgado-Martin C, Vincent T, Robbins A, Selvanathan A et al. Efficacy of JAK/STAT pathway inhibition in murine xenograft models of early T-cell precursor (ETP) acute lymphoblastic leukemia. Blood 2015; 125: 1759–1767.
    1. Smith GA, Uchida K, Weiss A, Taunton J. Essential biphasic role for JAK3 catalytic activity in IL-2 receptor signaling. Nat Chem Biol 2016; 12: 373–379.
    1. Karawajew L, Ruppert V, Wuchter C, Kösser A, Schrappe M, Dörken B et al. Inhibition of in vitro spontaneous apoptosis by IL-7 correlates with bcl-2 up-regulation, cortical/mature immunophenotype, and better early cytoreduction of childhood T-cell acute lymphoblastic leukemia. Blood 2000; 96: 297–306.
    1. Ryan J, Letai A. BH3 profiling in whole cells by fluorimeter or FACS. Methods 2013; 61: 156–164.
    1. Pieters R, Huismans DR, Loonen AH, Hählen K, van der Does-van den Berg A, van Wering ER et al. Relation of cellular drug resistance to long-term clinical outcome in childhood acute lymphoblastic leukaemia. Lancet 1991; 338: 399–403.
    1. Kaspers GJ, Veerman AJ, Pieters R, Van Zantwijk CH, Smets LA, Van Wering ER et al. In vitro cellular drug resistance and prognosis in newly diagnosed childhood acute lymphoblastic leukemia. Blood 1997; 90: 2723–2729.
    1. Den Boer ML, Harms DO, Pieters R, Kazemier KM, Gobel U, Körholz D et al. Patient stratification based on prednisolone-vincristine-asparaginase resistance profiles in children with acute lymphoblastic leukemia. J Clin Oncol 2003; 21: 3262–3268.
    1. Schmitz M, Breithaupt P, Scheidegger N, Cario G, Bonapace L, Meissner B et al. Xenografts of highly resistant leukemia recapitulate the clonal composition of the leukemogenic compartment. Blood 2011; 118: 1854–1864.
    1. Liem NLM, Papa RA, Milross CG, Schmid MA, Tajbakhsh M, Choi S et al. Characterization of childhood acute lymphoblastic leukemia xenograft models for the preclinical evaluation of new therapies. Blood 2004; 103: 3905–3914.
    1. Jiang Q, Li WQ, Aiello FB, Mazzucchelli R, Asefa B, Khaled AR et al. Cell biology of IL-7, a key lymphotrophin. Cytokine Growth Factor Rev 2005; 16: 513–533.
    1. Silva A, Laranjeira ABA, Martins LR, Cardoso BA, Demengeot J, Yunes JA et al. IL-7 contributes to the progression of human T-cell acute lymphoblastic leukemias. Cancer Res 2011; 71: 4780–4789.
    1. Winter SS, Dunsmore KP, Devidas M, Eisenberg N, Asselin BL, Wood BL et al. Safe integration of nelarabine into intensive chemotherapy in newly diagnosed T-cell acute lymphoblastic leukemia: Children’s Oncology Group Study AALL0434. Pediatr Blood Cancer 2015; 62: 1176–1183.
    1. Foxwell BM, Beadling C, Guschin D, Kerr I, Cantrell D. Interleukin-7 can induce the activation of Jak 1, Jak 3 and STAT 5 proteins in murine T cells. Eur J Immunol 1995; 25: 3041–3046.
    1. Quintás-Cardama A, Vaddi K, Liu P, Manshouri T, Li J, Scherle PA et al. Preclinical characterization of the selective JAK1/2 inhibitor INCB018424: therapeutic implications for the treatment of myeloproliferative neoplasms. Blood 2010; 115: 3109–3117.
    1. Loh ML, Tasian SK, Rabin KR, Brown P, Magoon D, Reid JM et al. A phase 1 dosing study of ruxolitinib in children with relapsed or refractory solid tumors, leukemias, or myeloproliferative neoplasms: A Children’s Oncology Group phase 1 consortium study (ADVL1011). Pediatr Blood Cancer 2015; 62: 1717–1724.
    1. Jing D, Bhadri VA, Beck D, Thoms JAI, Yakob NA, Wong JWH et al. Opposing regulation of BIM and BCL2 controls glucocorticoid-induced apoptosis of pediatric acute lymphoblastic leukemia cells. Blood 2015; 125: 273–283.
    1. Akashi K, Kondo M, von Freeden-Jeffry U, Murray R, Weissman IL. Bcl-2 rescues T lymphopoiesis in interleukin-7 receptor-deficient mice. Cell 1997; 89: 1033–1041.
    1. Amos CL, Woetmann A, Nielsen M, Geisler C, Odum N, Brown BL et al. The role of caspase 3 and BclxL in the action of interleukin 7 (IL-7): a survival factor in activated human T cells. Cytokine 1998; 10: 662–668.
    1. Opferman JT, Letai A, Beard C, Sorcinelli MD, Ong CC, Korsmeyer SJ. Development and maintenance of B and T lymphocytes requires antiapoptotic MCL-1. Nature 2003; 426: 671–676.
    1. Letai A, Bassik MC, Walensky LD, Sorcinelli MD, Weiler S, Korsmeyer SJ. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell. 2002; 2: 183–192.
    1. Wood BL, Winter SS, Dunsmore KP, Devidas M, chen si, Asselin BL et al. T-lymphoblastic leukemia (T-ALL) shows esscellent outcome, lack of significance of the early thymic precursor (ETP) immunophenotype, and validation of the prognostic value of end-induction minimal residual disease (MRD) in Children’s Oncology Group (COG) study AALL0434. Blood 2014; 124: 1-1. (Abstract 1).
    1. Patrick K, Wade R, Goulden N, Mitchell C, Moorman AV, Rowntree C et al. Outcome for children and young people with Early T-cell precursor acute lymphoblastic leukaemia treated on a contemporary protocol, UKALL 2003. Br J Haematol 2014; 166: 421–424.
    1. Conter V, Valsecchi MG, Buldini B, Parasole R, Locatelli F, Colombini A et al. Early T-cell precursor acute lymphoblastic leukaemia in children treated in AIEOP centres with AIEOP-BFM protocols: a retrospective analysis. Lancet Haematol 2016; 3: e80–e86.
    1. Berki T, Pálinkás L, Boldizsár F, Németh P. Glucocorticoid (GC) sensitivity and GC receptor expression differ in thymocyte subpopulations. Int Immunol 2002; 14: 463–469.
    1. Veis DJ, Sentman CL, Bach EA, Korsmeyer SJ. Expression of the Bcl-2 protein in murine and human thymocytes and in peripheral T lymphocytes. J Immunol 1993; 151: 2546–2554.
    1. Gratiot-Deans J, Ding L, Turka LA, Nuñez G. bcl-2 proto-oncogene expression during human T cell development. Evidence for biphasic regulation. J Immunol 1993; 151: 83–91.
    1. Kim GY, Hong C, Park J-H. Seeing is believing: illuminating the source of in vivo interleukin-7. Immune Netw 2011; 11: 1–10.
    1. Barata JT, Cardoso AA, Nadler LM, Boussiotis VA. Interleukin-7 promotes survival and cell cycle progression of T-cell acute lymphoblastic leukemia cells by down-regulating the cyclin-dependent kinase inhibitor p27(kip1). Blood 2001; 98: 1524–1531.
    1. Barata JT, Cardoso AA, Boussiotis VA. Interleukin-7 in T-cell acute lymphoblastic leukemia: an extrinsic factor supporting leukemogenesis? Leuk Lymphoma 2005; 46: 483–495.
    1. Hernández-Caselles T, Martínez-Esparza M, Sancho D, Rubio G, Aparicio P. Interleukin-7 rescues human activated T lymphocytes from apoptosis induced by glucocorticoesteroids and regulates bcl-2 and CD25 expression. Hum Immunol 1995; 43: 181–189.
    1. Bolotin E, Annett G, Parkman R, Weinberg K. Serum levels of IL-7 in bone marrow transplant recipients: relationship to clinical characteristics and lymphocyte count. Bone Marrow Transplant. 1999; 23: 783–788.
    1. Napolitano LA, Grant RM, Deeks SG, Schmidt D, De Rosa SC, Herzenberg LA et al. Increased production of IL-7 accompanies HIV-1-mediated T-cell depletion: implications for T-cell homeostasis. Nat Med 2001; 7: 73–79.
    1. Zubkova I, Mostowski H, Zaitseva M. Up-regulation of IL-7, stromal-derived factor-1 alpha, thymus-expressed chemokine, and secondary lymphoid tissue chemokine gene expression in the stromal cells in response to thymocyte depletion: implication for thymus reconstitution. J Immunol 2005; 175: 2321–2330.
    1. Miller CN, Hartigan-O’Connor DJ, Lee MS, Laidlaw G, Cornelissen IP, Matloubian M et al. IL-7 production in murine lymphatic endothelial cells and induction in the setting of peripheral lymphopenia. Int Immunol 2013; 25: 471–483.
    1. Cleaver AL, Beesley AH, Firth MJ, Sturges NC, O’Leary RA, Hunger SP et al. Gene-based outcome prediction in multiple cohorts of pediatric T-cell acute lymphoblastic leukemia: a Children’s Oncology Group study. Mol Cancer 2010; 9: 105.
    1. Li Y, Buijs-Gladdines JGCAM, Canté-Barrett K, Stubbs AP, Vroegindeweij EM, Smits WK et al. IL-7 receptor mutations and steroid resistance in pediatric t cell acute lymphoblastic leukemia: A Genome Sequencing Study. PLoS Med 2016; 13: e1002200.
    1. Hofmeister R, Khaled AR, Benbernou N, Rajnavolgyi E, Muegge K, Durum SK. Interleukin-7: physiological roles and mechanisms of action. Cytokine Growth Factor Rev 1999; 10: 41–60.
    1. Zenatti PP, Ribeiro D, Li W, Zuurbier L, Silva MC, Paganin M et al. Oncogenic IL7R gain-of-function mutations in childhood T-cell acute lymphoblastic leukemia. Nat Genet 2011; 43: 932–939.
    1. Canté-Barrett K, Spijkers-Hagelstein JaP, Buijs-Gladdines JGC a. M, Uitdehaag JCM, Smits WK, van der Zwet J et al. MEK and PI3K-AKT inhibitors synergistically block activated IL7 receptor signaling in T-cell acute lymphoblastic leukemia. Leukemia 2016; 30: 1832–1843.
    1. Treanor LM, Zhou S, Janke L, Churchman ML, Ma Z, Lu T et al. Interleukin-7 receptor mutants initiate early T cell precursor leukemia in murine thymocyte progenitors with multipotent potential. J Exp Med 2014; 211: 701–713.
    1. Schultz KR, Bowman WP, Aledo A, Slayton WB, Sather H, Devidas M et al. Improved early event-free survival with imatinib in Philadelphia chromosome-positive acute lymphoblastic leukemia: a children’s oncology group study. J Clin Oncol 2009; 27: 5175–5181.
    1. Rogatsky I, Hittelman AB, Pearce D, Garabedian MJ. Distinct glucocorticoid receptor transcriptional regulatory surfaces mediate the cytotoxic and cytostatic effects of glucocorticoids. Mol Cell Biol 1999; 19: 5036–5049.

Source: PubMed

3
Abonnieren