Persistent janus kinase-signaling in chronic lymphocytic leukemia patients on ibrutinib: Results of a phase I trial

David E Spaner, Lindsay McCaw, Guizhei Wang, Hubert Tsui, Yonghong Shi, David E Spaner, Lindsay McCaw, Guizhei Wang, Hubert Tsui, Yonghong Shi

Abstract

Methods to deepen clinical responses to ibrutinib are needed to improve outcomes for patients with chronic lymphocytic leukemia (CLL). This study aimed to determine the safety and efficacy of combining a janus kinase (JAK)-inhibitor with ibrutinib because JAK-mediated cytokine-signals support CLL cells and may not be inhibited by ibrutinib. The JAK1/2 inhibitor ruxolitinib was prescribed to 12 CLL patients with abnormal serum beta-2 microglobulin levels after 6 months or persistent lymphadenopathy or splenomegaly after 12 months on ibrutinib using a 3 + 3 phase 1 trial design (NCT02912754). Ibrutinib was continued at 420 mg daily and ruxolitinib was added at 5, 10, 15, or 20 mg BID for 3 weeks out of five for seven cycles. The break was mandated to avoid anemia and thrombocytopenia observed with ruxolitinib as a single agent in CLL. The combination was well-tolerated without dose-limiting toxicities. Cyclic changes in platelets, lymphocytes, and associated chemokines and thrombopoietic factors were observed and partial response criteria were met in 2 of 12 patients. The results suggest that JAK-signaling helps CLL cells persist in the presence of ibrutinib and ruxolitinib with ibrutinib is well-tolerated and may be a useful regiment to use in combination therapies for CLL.

Keywords: chemokines; chronic lymphocytic leukemia; ibrutinib; janus kinases; ruxolitinib; thrombopoiesis.

Conflict of interest statement

D. E. Spaner reports grants from Novartis to support the submitted work and personal fees from Janssen outside the submitted work. The other authors declare no conflicts of interest with respect to this work.

© 2019 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
Effect of ruxolitinib on hemoglobin (Hb), platelets, and β2M. (A) Hb was measured before starting ruxolitinib while on ibrutinib (C1D1) and after completing the seventh cycle of ruxolitinib (C7D21). The lines indicate results for individual patients and data for groups 1 and 2 (5 and 10 mg ruxolitinib) and groups 3 and 4 (15 and 20 mg ruxolitinib) are shown in different graphs. (B) Platelet counts were measured at C1D1 and at end of treatment (EOT). (C) β2M was measured at each time‐point and normalized to the initial value at C1D1. The average and standard error of these ratios for all patients are plotted as a function of time and show that β2M levels decreased during treatment with ruxolitinib (indicated by the solid bars labeled “C”) but recovered during the 2 week break from ruxolitinib in each cycle (indicated by the dashed bars labeled “B”). *P < 0.05; ns, not significant
Figure 2
Figure 2
Effect of ruxolitinib on blood lymphocytes. (A) Lymphocyte numbers at each time‐point were taken from the medical record. Differences between lymphocyte counts at each time‐point and the initial count at C1D1 were plotted as a function of time. Each line represents results for an individual patient. Groups 1 and 2 are distinguished from groups 3 and 4. The bars labeled C1, C2, C3, etc. represent the 3‐week period on ruxolitinib during each treatment cycle. Results for JAK2011 were considered uninterpretable as a result of confounding clinical events not related to the trial and were not included in the analysis. (B) Blood from JAK2014 was collected at C6D21, C7D1, and C7D21 and analyzed by 10‐color flow cytometry. Percentages of selected lymphocyte populations at each time‐point are shown below the respective dot‐plots and suggest CD5+ CD19+ CLL cells and CD38+ NK cells cycle in and out of the blood in the presence and absence of ruxolitinib
Figure 3
Figure 3
Effect of ruxolitinib on platelets and thrombopoietic cytokines. (A) Platelet counts were taken from the medical record. Differences between the number at each time‐point and initial platelet count at C1D1 were plotted as a function of time. Each line represents the calculations for a single patient. Results for groups 1 and 2 are shown separately from groups 3 and 4. (B‐D) TPO (B), PDGF‐AA (C, right panel), and CD40L (D, right panel) were measured in plasma from bone marrow aspirates obtained at C1D1 and CD3D21. PDGF‐AA (C, left panel) and CD40L (D, left panel) were also measured in plasma from blood at day 1 and day 21 of a treatment cycle. Each line represents results for an individual patient. *P < 0.05
Figure 4
Figure 4
Effect of ruxolitinib on IL10, CXCL10, and CXCL1. IL10, CXCL10, and CXCL1 were measured in plasma from blood at day 1 and day 21 of a treatment cycle (left panels) and also in plasma from marrow aspirates obtained at C1D1 and CD3D21 (right panels). Each line represents results for an individual patient. *P < 0.05

References

    1. Byrd JC, Furman RR, Coutre SE, et al. Three‐year follow‐up of treatment‐naive and previously treated patients with CLL and SLL receiving single‐agent ibrutinib. Blood. 2015;125:2497‐2506.
    1. Pinilla‐Ibarz J, Chavez JC. Life after ibrutinib? A new unmet need in CLL. Blood. 2015;125:2013‐2014.
    1. Mertens D, Stilgenbauer S. Ibrutinib‐resistant CLL: unwanted and unwonted!. Blood. 2017;129:1407‐1409.
    1. Li Y, Shi Y, McCaw L, et al. Microenvironmental interleukin‐6 suppresses toll‐like receptor signaling in human leukemia cells through miR‐17/19A. Blood. 2015;126:766‐778.
    1. Oppermann S, Lam AJ, Tung S, et al. Janus and PI3‐kinases mediate glucocorticoid resistance in activated chronic leukemia cells. Oncotarget. 2016;7:72608‐72621.
    1. Niemann CU, Herman SE, Maric I, et al. Disruption of in vivo CLL tumor‐microenvironment interactions by ibrutinib ‐ findings from an investigator initiated phase 2 study. Clin Cancer Res. 2015;22:1572‐1582.
    1. Aguilar‐Hernandez MM, Blunt MD, Dobson R, et al. IL‐4 enhances expression and function of surface IgM in CLL cells. Blood. 2016;127:3015‐3025.
    1. Verstovsek S, Mesa RA, Gotlib J, et al. A double‐blind, placebo‐controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366:799‐807.
    1. Passamonti F, Griesshammer M, Palandri F, et al. Ruxolitinib for the treatment of inadequately controlled polycythaemia vera without splenomegaly (RESPONSE‐2): a randomised, open‐label, phase 3b study. Lancet Oncol. 2017;18:88‐99.
    1. Spaner DE, Wang G, McCaw L, et al. Activity of the janus kinase inhibitor Ruxolitinib in CLL: results of a phase II trial. Haematologica. 2016;101:e192‐e195.
    1. Jain P, Keating M, Renner S, et al. Ruxolitinib for symptom control in patients with CLL: a single‐group, phase 2 trial. Lancet Haematol. 2017;4:e67‐e74.
    1. de Jong J, Skee D, Murphy J, et al. Effect of CYP3A perpetrators on ibrutinib exposure in healthy participants. Pharmacol Res Perspect. 2015;3:e00156.
    1. Thompson PA, O'Brien SM, Xiao L, et al. β2‐microglobulin normalization within 6 months of ibrutinib‐based treatment is associated with superior progression‐free survival in patients with chronic lymphocytic leukemia. Cancer. 2016;122:565‐573.
    1. Linguraru MG, Sandberg JK, Jones EC, Summers RM. Assessing splenomegaly: automated volumetric analysis of the spleen. Acad Radiol. 2013;20:675‐684.
    1. Le Tourneau C, Lee JJ, Siu LL. Dose escalation methods in phase I cancer clinical trials. JNCI. 2009;101:708‐720.
    1. Gehan EA. The determination of the number of patients required in a preliminary and a follow‐up trial of a new chemotherapeutic agent. J Chronic Dis. 1961;13:346‐353.
    1. Hallek M, Cheson BD, Catovsky D, et al. International workshop on chronic lymphocytic leukemia. Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute‐Working Group 1996 guidelines. Blood. 2008;111:5446‐5456.
    1. International CLL‐IPI Working Group . An international prognostic index for patients with chronic lymphocytic leukaemia (CLL‐IPI): a meta‐analysis of individual patient data. Lancet Oncol. 2016;17:779‐790.
    1. Yang M, Chesterman CN, Chong BH. Recombinant PDGF enhances megakaryocytopoiesis in vitro. Br J Haematol. 1995;91:285‐289.
    1. Viallard JF, Solanilla A, Gauthier B, et al. Increased soluble and platelet‐associated CD40 ligand in essential thrombocythemia and reactive thrombocytosis. Blood. 2002;99:2612‐2624.
    1. Pattison MJ, Mackenzie KF, Arthur JS. Inhibition of JAKs in macrophages increases lipopolysaccharide‐induced cytokine production by blocking IL‐10‐mediated feedback. J Immunol. 2012;189:2784‐2792.
    1. Long M, Beckwith K, Do P, et al. Ibrutinib treatment improves T cell number and function in CLL patients. J Clin Invest. 2017;127:3052‐3064.
    1. Antonelli A, Ferrari SM, Giuggioli D, Ferrannini E, Ferri C, Fallahi P. Chemokine (C‐X‐C motif) ligand (CXCL)10 in autoimmune diseases. Autoimmun Rev. 2014;13:272‐280.
    1. Lv M, Xu Y, Tang R, et al. miR141‐CXCL1‐CXCR2 signaling‐induced Treg recruitment regulates metastases and survival of non‐small cell lung cancer. Mol Cancer Ther. 2014;13:3152‐3162.
    1. Burger JA, Montserrat E. Coming full circle: 70 years of CLL cell redistribution, from glucocorticoids to inhibitors of B‐cell receptor signaling. Blood. 2013;121:1501‐1509.
    1. Ahr B, Denizot M, Robert‐Hebmann V, Brelot A, Biard‐Piechaczyk M. Identification of the cytoplasmic domains of CXCR4 involved in Jak2 and STAT3 phosphorylation. J Biol Chem. 2005;280:6692‐6700.
    1. Johnson CS, Cook CA, Furmanski P. In vivo suppression of erythropoiesis by tumor necrosis factor‐alpha (TNF‐alpha): reversal with exogenous erythropoietin (EPO). Exp Hematol. 1990;18:109‐113.
    1. Mizutani H, Furubayashi T, Imai Y, et al. Mechanisms of corticosteroid action in immune thrombocytopenic purpura (ITP): experimental studies using ITP‐prone mice, (NZW x BXSB) F1. Blood. 1992;79:942‐947.
    1. Meyer SC, Keller MD, Woods BA, et al. Genetic studies reveal an unexpected negative regulatory role for Jak2 in thrombopoiesis. Blood. 2014;124:2280‐2294.
    1. Sosman JA, Verma A, Moss S, et al. Interleukin 10‐induced thrombocytopenia in normal healthy adult volunteers: evidence for decreased platelet production. Br J Haematol. 2000;111:104‐111.
    1. Shi Y, Wang G, Muhowski EM, et al. Ibrutinib reprograms the glucocorticoid receptor in CLL cells. Leukemia. 2019. [Epub ahead of print]. 10.1038/s41375-019-0381-4.
    1. Rogers KA, Huang Y, Ruppert AS, et al. Phase 1b study of obinutuzumab, ibrutinib, and venetoclax in relapsed and refractory chronic lymphocytic leukemia. Blood. 2018;132:1568‐1572.
    1. Oppermann S, Ylanko J, Shi Y, et al. High‐content screening identifies kinase inhibitors that overcome venetoclax resistance in activated CLL cells. Blood. 2016;128:934‐947.
    1. Guarini A, Peragine N, Messina M, et al. Unravelling the suboptimal response of TP53‐mutated chronic lymphocytic leukaemia to ibrutinib. Br J Haematol. 2019;184:392‐396.
    1. Tomic J, Lichty B, Spaner DE. Aberrant interferon‐signaling is associated with aggressive chronic lymphocytic leukemia. Blood. 2011;117:2668‐2680.

Source: PubMed

3
Subskrybuj