15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      JAK/STAT Pathway Mutations in T-ALL, Including the STAT5B N642H Mutation, are Sensitive to JAK1/JAK3 Inhibitors

      letter
      1 , 2 , 1 , 2 , 1 , 2 , 1 , 2
      HemaSphere
      Wolters Kluwer Health

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy of the lymphocytes that is driven by the cooperation of various mutations. Constitutive activation of JAK-STAT signaling is observed in one-third of T-ALL patients and is caused by activating mutations in the interleukin 7 receptor alpha chain (IL7R), in the Janus kinases JAK1 or JAK3, or in the Signal transducer and activator of transcription 5B (STAT5B). 1–3 STAT5B mutations are most frequently the N642H variant and are associated with an unfavorable prognosis and higher risk of relapse. 1,2,4,5 We set out to test the efficacy of JAK1/JAK3 kinase inhibitors ruxolitinib, tofacitinib, upadacitinib, baricitinib, decernotinib, and peficitinib in the murine Ba/F3 cell model transformed by different mutated components of JAK/STAT signaling. We show that cells carrying mutations in any component of IL7 receptor signaling are sensitive to JAK-inhibition, including mutations in STAT5B. JAK2 inhibition by ruxolitinib is a well-established treatment mechanism for polycythemia vera and myelofibrosis. 6,7 The central role for the JAK1 and JAK3 kinases in IL7R/JAK/STAT driven T-ALL provides a rationale for JAK inhibition in this disease. The use of ruxolitinib, which has selectivity towards JAK1 and JAK2 and tofacitinib, targeting JAK1 and JAK3, in T-ALL is supported by preclinical evidence, showing sensitivity of IL7R, JAK1, and JAK3 mutated cases to these inhibitors in vitro 8 and in vivo in patient-derived xenograft models. 9 Indeed, mutant IL7R and mutant JAK3 typically require JAK1 activity and are sensitive to JAK1 inhibition. 10,11 PhaseI/II clinical trials for acute lymphoblastic leukemia with ruxolitinib are currently ongoing. IL7R signaling is important for normal development, maturation and homeostasis of the B- and T-cell compartment. The IL7 receptor is a heterodimer that consists of the IL7Ralpha chain and the common gamma receptor protein (IL2Rγ). Upon binding of interleukin 7, the ligand, to the IL7R, a conformational change occurs that brings both chains closer together, exposing tyrosine kinases JAK1 and JAK3 for phosphorylation and activation. They in turn phosphorylate downstream effectors including STAT5A and STAT5B. After phosphorylation of tyrosine 694, the STATs dimerize and translocate to the nucleus to activate transcription of STAT5 target genes, 3 including known oncogenes (eg, Myc) and anti-apoptotic factors such as BCL2. Mutations in IL7R or JAK1/3 increase phosphorylation activity of the JAKs, while the STAT5B(N642H) mutant displays stronger intra-dimer interaction, increased and prolonged phosphorylation of tyrosine Y694 as well as increased DNA binding capacities and transcriptional activity compared to wild type STAT5B. 4,5 We used BaF/3 cells that were transformed to cytokine-independent growth by expression of IL7R(insSRCL), JAK1(A634D), JAK3(M511I), JAK3(L857Q), JAK3(L875H), STAT5B(N642H), or NRAS(G12D). We treated these cells with ruxolitinib, tofacitinib, upadacitinib, baricitinib, decernotinib, and peficitinib in a concentration range from 1 nM to 10 μM for 24 hours. We used ATPlite luminescence as a read-out for proliferation and established dose-response curves and calculated IC50 values (Fig. 1A, Table 1). Cells expressing mutant JAK1 or JAK3 were sensitive to JAK-inhibition with IC50 values between 203 nM and 2395 nM. IC50 values for different mutants were determined by both specificity and potency of the inhibitor, as well as intrinsic sensitivity to inhibition of the given mutant. Importantly, all tested new inhibitors showed efficacy in inhibiting proliferation of JAK mutants comparable to well established JAK inhibitor ruxolitinib. The JAK3(L875H) mutant was the least sensitive mutant and showed high resistance towards upadacitinib and baricitinib. (Fig. 1A) Both inhibitors selectively inhibit JAK1, confirming the JAK1 independent character of mutations in the kinase domain of JAK3. The IL7R(insSRCL) transformed Ba/F3 cells showed the highest sensitivity to all inhibitors, except to decernotinib, which is a more JAK3 selective inhibitor. Surprisingly, the STAT5B(N642H) expressing Ba/F3 cells that were grown in the absence of cytokines and do not contain JAK kinase mutations, were also sensitive to JAK-inhibition with IC50 values within range of the other cells and even higher sensitivity than JAK3(L875H). Figure 1 Ba/F3 cells transformed by any mutated member of IL7-JAK-STAT signaling are sensitive to JAK-inhibition. (A) Dose-response curves for ruxolitinib, tofacitinib, upadacitinib, baricitinib, decernotinib, and peficitinib treatment on Ba/F3 cells transformed by IL7R(insSRCL), JAK1(A634D), JAK3(M511I), JAK3(L857Q), JAK3(L875H), STAT5B(N642H), and NRAS(G12D) after 24 hours incubation. Data represent the mean of 3 experiments. (B) Mean fluorescence intensity values for p-STAT5B Y694 of Ba/F3 cells transformed by IL7R(insSRCL), JAK1(A634D), JAK3(M511I), JAK3(L857Q), JAK3(L875H), STAT5B(N642H), and NRAS(G12D) after treatment with upadacitinib, tofacitinib, ruxolitinib 2 μM or DMSO for 3 hours. Symbols represent biological replicates, mean and standard deviation (SD) are shown. Significance was calculated using one-way analysis of variance (ANOVA) with Bonferroni multiple comparisons correction. ns = not significant (p ≥ 0.05). Upa: upadacitinib, Tofa: tofacitinib, Ruxo: ruxolitinib. (C) Western blot analysis of phospho-STAT5 Y694 and STAT5 in STAT5B(N642H) transformed Ba/F3 cells after treatment with baricitinib, decernotinib, peficitinib, ruxolitinib, tofacitinib, and upadacitinib 2 μM or DMSO for 90 minutes. Table 1 IC50 values (nM) for all inhibitors in Figure 1 in Ba/F3 cells overexpressing IL7R(insSRCL), JAK1(A634D), JAK3(M511I), JAK3(L857Q), JAK3(L875H), STAT5B(N642H), or NRAS(G12D). To determine if the sensitivity to JAK-inhibition was an on-target effect, we performed flow cytometry analysis of phospho-STAT5B Y694 after treatment of the different Ba/F3 variants with 2 μM ruxolitinib, tofacitinib, upadacitinib, or DMSO for 3 hours (Fig. 1B). These data showed that growth inhibition correlated with loss of phospho-STAT5 signal for all sensitive JAK-STAT mutants. The JAK3(L875H) mutant was not very sensitive to these inhibitors and showed also no response in the phospho-STAT5 measurements. The STAT5B(N642H) cells were sensitive to ruxolitinib, tofacitinib, upadacitinib, and showed clear decreases in STAT5 phosphorylation upon treatment with these inhibitors. This was observed by phospho-flow measurement after 3 hours of treatment (Fig. 1B) or Western blot analysis of STAT5 Y694 phosphorylation after 90 minutes of treatment (Fig. 1C). Moreover, we observed reduced expression of STAT5 target genes in the STAT5B(N642H) expressing cells upon ruxolitinib treatment, as measured by quantitative RT-PCR (Fig. 2A). These results confirm a rapid loss of phosphorylation of STAT5 and loss of its transcriptional activity upon treatment of the cells with JAK inhibitors. Thus, despite the absence of activating mutations in the JAK kinases, the STAT5B(N642H) mutant showed high levels of phosphorylation that could be repressed with the use of JAK-inhibitors (Fig. 1). Figure 2 JAK-inhibition in Ba/F3 STAT5B(N642H) reduces expression of STAT5 target genes and causes a proliferation deficit that is synergistic with Bcl2-inhibition. (A) RT-qPCR of Ba/F3 STAT5B(N642H) for STAT5 target genes Bcl2, Cish, Pim1, Myc, and Osm after treatment with 1 μM ruxolitinib or DMSO for 3 hours. Data show the mean of 3 experiments +/− SD. ∗ p < 0.05. in unpaired two-tailed t test with equal variance. (B) RT-qPCR of Ba/F3 STAT5B(N642H) for JAK1 and JAK3 24 hours after electroporation with negative control siRNA (NC), siRNA targeting JAK3 (195, 196) or siRNA targeting JAK1 (198, 199). Data represent the mean of 3 experiments +/− SD. ∗∗ p < 0.0001, p values were calculated using one-way analysis of variance (ANOVA) with Bonferroni multiple comparisons correction. Data represent the mean of 3 experiments +/− SD. (C) Relative fold change of proliferation of Ba/F3 STAT5B(N642H) 72 hours after electroporation with negative control siRNA (NC), siRNA targeting JAK3 (195, 196) or siRNA targeting JAK1 (198, 199). ∗∗ p < 0.0001, significance was calculated using one-way analysis of variance (ANOVA) with the Bonferroni multiple comparisons correction. Data are presented as mean of 3 experiments +/− SD. (D-E) Ba/F3 STAT5B(N642H) were cultured for 10 days in the presence of DMSO, ruxolitinib, tofacitinib, upadacitinib, baricitinib, decernotinib, or peficitinib and analyzed by flowcytometry for concentration (D) and viablility (E). Data represent the mean of 3 experiments +/− SD. (F) Dose-response curves for venetoclax on Ba/F3 cells transformed by STAT5B(N642H) after 24 h incubation in the presence of 0 nM, 100 nM or 200 nM ruxolitinib. The mean of 3 experiments is shown. (G) Synergy matrix plot showing δ-scores for Ba/F3 STAT5B(N642H) treated with ruxolitinib and venetoclax. δ-score = the average d-score for the whole range of concentrations shown in the synergy matrix. These data indicate that the STAT5B(N642H) mutant itself induces activation of the JAK kinases and that this is required for its phosphorylation on Y694 and its transcriptional activity. To further confirm this, we electroporated Ba/F3 STAT5B(N642H) cells with siRNA targeting Jak1, Jak3 or negative control and confirmed knockdown by RT-PCR (Fig. 2B). Proliferation of the STAT5B(N642H) transformed cells was significantly reduced by Jak1 or Jak3 knock-down (Fig. 2C), supporting the hypothesis that JAK kinases are essential for proliferation of STAT5B(N642H) mutated cells. While this work was in progress, Pham and colleagues 12 came to similar conclusions in a mouse T-cell lymphoma/leukemia model driven by STAT5B(N642H). They documented strong phosphorylation of STAT5B(N642H) and sensitivity of the leukemia cells to JAK inhibition. To investigate whether JAK-inhibition could sustainably inhibit proliferation of STAT5B(N642H) overexpressing cells, we cultured the cells at a starting concentration of 100.000/mL for 10 days in culture medium supplemented with 2 μM concentration of inhibitor or equal amount of DMSO as control (Fig. 2D). When compared to DMSO, proliferation was clearly reduced for every inhibitor. However, in contrast to what has previously been described for JAK1 and JAK3 mutants, complete eradication of the cultured STAT5B(N642H) overexpressing cells could not be achieved. This corresponds with the inability to reduce relative proliferation to zero in the dose-response curves (Fig. 1A). Upon treatment, a fast decrease in viability occurs. For tofacitinib, upadacitinib, baricitinib, decernotinib, and peficitinib this is accompanied by a strong reduction in rate of proliferation. Treatment with ruxolitinib causes cell death during the first 4 days of treatment, after which viability starts to increase for all inhibitors (Fig. 2E). Despite incomplete response, proliferation rates remained strongly reduced for the entire duration of the growth experiment (Fig. 2D). It can be expected that Ba/F3 STAT5B(N642H) cells would rely on the expression of STAT5B target genes such as BCL2 for survival. Indeed, these cells were sensitive to BCL2-inhibition by venetoclax (IC50 1492 nM) and addition of 100 nM or 200 nM ruxolitinib showed a drastic decrease of the IC50 value of venetoclax to 107 nM and 28 nM respectively (Fig. 2F). This synergistic activity between ruxolitinib and venetoclax was indicated by high synergy scores calculated according to the zero interaction potential method. 13 We observed a clear overall synergistic effect (δ-score 30.09), with a maximum ZIP synergy score of 43.23, which means that there was >40% additional inhibition compared to the additive effect of the two compounds 14 (Fig. 2G). From this work, we conclude that the recently developed JAK-inhibitors with higher specificity towards JAK1 and JAK3 when compared to ruxolitinib, all show activity against hyperactivation of JAK-STAT signaling in T-ALL with similar or higher efficacy compared to ruxolitinib. Interestingly, despite its position downstream of the JAK kinases, we find that the STAT5B(N642H) mutant also depends on the JAK1/JAK3 kinase activity. Therefore, expression of STAT5 target genes and cell growth was reduced by JAK-inhibition in cells that express mutant STAT5B and this effect was synergistic with the BCL2-inhibitor venetoclax. In vivo, the sensitivity to JAK-inhibition is affected by the mutational landscape and clonal architecture of the leukemia for any given patient. 4,15 However, our data suggest that T-ALL patients who carry the STAT5B(N642H) mutation may still benefit from treatment with a JAK-inhibitor especially in combination with a BCL2 inhibitor.

          Related collections

          Most cited references9

          • Record: found
          • Abstract: found
          • Article: not found

          THE GENOMIC LANDSCAPE OF PEDIATRIC AND YOUNG ADULT T-LINEAGE ACUTE LYMPHOBLASTIC LEUKEMIA

          Genetic alterations activating NOTCH1 signaling and T cell transcription factors, coupled with inactivation of the INK4/ARF tumor suppressors are hallmarks of T-ALL, but detailed genome-wide sequencing of large T-ALL cohorts has not been performed. Using integrated genomic analysis of 264 T-ALL cases, we identify 106 putative driver genes, half of which were not previously described in childhood T-ALL (e.g. CCND3, CTCF, MYB, SMARCA4, ZFP36L2 and MYCN). We described new mechanisms of coding and non-coding alteration, and identify 10 recurrently altered pathways, with associations between mutated genes and pathways, and stage or subtype of T-ALL. For example, NRAS/FLT3 mutations were associated with immature T-ALL, JAK3/STAT5B mutations in HOX1 deregulated ALL, PTPN2 mutations in TLX1 T-ALL, and PIK3R1/PTEN mutations in TAL1 ALL, suggesting that different signaling pathways have distinct roles according to maturational stage. This genomic landscape provides a logical framework for the development of faithful genetic models and new therapeutic approaches.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Efficacy of JAK/STAT pathway inhibition in murine xenograft models of early T-cell precursor (ETP) acute lymphoblastic leukemia.

            Early T-cell precursor (ETP) acute lymphoblastic leukemia (ALL) is a recently described subtype of T-ALL characterized by a unique immunophenotype and genomic profile, as well as a high rate of induction failure. Frequent mutations in cytokine receptor and Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathways led us to hypothesize that ETP-ALL is dependent on JAK/STAT signaling. Here we demonstrate aberrant activation of the JAK/STAT pathway in ETP-ALL blasts relative to non-ETP T-ALL. Moreover, ETP-ALL showed hyperactivation of STAT5 in response to interleukin-7, an effect that was abrogated by the JAK1/2 inhibitor ruxolitinib. In vivo, ruxolitinib displayed activity in 6 of 6 patient-derived murine xenograft models of ETP-ALL, with profound single-agent efficacy in 5 models. Ruxolitinib treatment decreased peripheral blast counts relative to pretreatment levels and compared with control (P < .01) in 5 of 6 ETP-ALL xenografts, with marked reduction in mean splenic blast counts (P < .01) in 6 of 6 samples. Surprisingly, both JAK/STAT pathway activation and ruxolitinib efficacy were independent of the presence of JAK/STAT pathway mutations, raising the possibility that the therapeutic potential of ruxolitinib in ETP-ALL extends beyond those cases with JAK mutations. These findings establish the preclinical in vivo efficacy of ruxolitinib in ETP-ALL, a biologically distinct subtype for which novel therapies are needed.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A pooled analysis of overall survival in COMFORT-I and COMFORT-II, 2 randomized phase III trials of ruxolitinib for the treatment of myelofibrosis.

              Ruxolitinib, a potent Janus kinase 1/2 inhibitor, resulted in rapid and durable improvements in splenomegaly and disease-related symptoms in the 2 phase III COMFORT studies. In addition, ruxolitinib was associated with prolonged survival compared with placebo (COMFORT-I) and best available therapy (COMFORT-II). We present a pooled analysis of overall survival in the COMFORT studies using an intent-to-treat analysis and an analysis correcting for crossover in the control arms. Overall, 301 patients received ruxolitinib (COMFORT-I, n=155; COMFORT-II, n=146) and 227 patients received placebo (n=154) or best available therapy (n=73). After a median three years of follow up, intent-to-treat analysis showed that patients who received ruxolitinib had prolonged survival compared with patients who received placebo or best available therapy [hazard ratio=0.65; 95% confidence interval (95%CI): 0.46-0.90; P=0.01]; the crossover-corrected hazard ratio was 0.29 (95%CI: 0.13-0.63). Both patients with intermediate-2- or high-risk disease showed prolonged survival, and patients with high-risk disease in the ruxolitinib group had survival similar to that of patients with intermediate-2-risk disease in the control group. The Kaplan-Meier estimate of overall survival at week 144 was 78% in the ruxolitinib arm, 61% in the intent-to-treat control arm, and 31% in the crossover-adjusted control arm. While larger spleen size at baseline was prognostic for shortened survival, reductions in spleen size with ruxolitinib treatment correlated with longer survival. These findings are consistent with previous reports and support that ruxolitinib offers a survival benefit for patients with myelofibrosis compared with conventional therapies. (clinicaltrials.gov identifiers: COMFORT-I, NCT00952289; COMFORT-II, NCT00934544).
                Bookmark

                Author and article information

                Journal
                Hemasphere
                Hemasphere
                HS9
                HemaSphere
                Wolters Kluwer Health
                2572-9241
                December 2019
                22 October 2019
                : 3
                : 6
                : e313
                Affiliations
                [1 ]VIB Center for Cancer Biology, Leuven, Belgium.
                [2 ]KU Leuven Center for Human Genetics, Leuven, Belgium.
                Author notes
                Correspondence: Jan Cools (e-mail: jan.cools@ 123456kuleuven.be)
                Article
                HemaSphere-2019-0161 00021
                10.1097/HS9.0000000000000313
                6924561
                31976485
                0ea60d28-910a-4666-bef3-f7ae8f6a2492
                Copyright © 2019 the Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the European Hematology Association.

                This is an open access article distributed under the terms of the Creative Commons Attribution-Non Commercial-No Derivatives License 4.0 (CCBY-NC-ND), where it is permissible to download and share the work provided it is properly cited. The work cannot be changed in any way or used commercially without permission from the journal. http://creativecommons.org/licenses/by-nc-nd/4.0

                History
                : 6 August 2019
                : 14 October 2019
                : 16 October 2019
                Categories
                Letter
                002
                Custom metadata
                TRUE

                Comments

                Comment on this article